<<

in Graft-versus- Andrea S. Henden and Geoffrey R. Hill J Immunol 2015; 194:4604-4612; ; This information is current as doi: 10.4049/jimmunol.1500117 of September 25, 2021. http://www.jimmunol.org/content/194/10/4604 Downloaded from References This article cites 134 articles, 49 of which you can access for free at: http://www.jimmunol.org/content/194/10/4604.full#ref-list-1

Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication

*average by guest on September 25, 2021 Subscription Information about subscribing to The Journal of is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2015 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Th eJournal of Brief Reviews Immunology

Cytokines in Graft-versus-Host Disease Andrea S. Henden and Geoffrey R. Hill Graft-versus-host disease (GVHD) is a complication of mechanisms. An example of this is depletion of trans- allogeneic bone marrow transplantation whereby trans- plants: a reduction in GVHD is offset by attendant increases in planted naive and marrow-derived T cells damage recip- the rates of relapse of primary malignancy (2, 3), in addition to ient tissue through similar mechanisms to those that allow more delayed immune reconstitution with increased morbidity destruction of malignant cells, the therapeutic intent of and mortality due to opportunistic . An alternate focus bone marrow transplantation. The manifestations and has been to examine the influences on emerging innate and severity of GVHD are highly variable and are influenced adaptive immune responses in an attempt to preserve beneficial by the proportions of naive cells maturing along regula- GVL effects while eliminating the harmful “off-target” GVHD effects. In this setting, understanding the orchestration

tory T cell, Th1, Th2, or Th17 phenotypes. This matu- Downloaded from ration is largely influenced by local cytokines, which, in of the maturing within allogeneic transplan- turn, activate transcription factors and drive development tation offers the opportunity to improve outcomes of this treatment through identification of rapidly translatable clinical toward a dominant phenotype. In addition, proinflamma- therapeutic targets. Our understanding of events within the al- tory cytokines exert direct effects on GVHD target tissues. logeneic transplantation landscape also informs our under- Our knowledge of the role that cytokines play in orches- standing of emerging innate and adaptive immune responses in trating GVHD is expanding rapidly and parallels other scenarios other than BMT. http://www.jimmunol.org/ infective and inflammatory conditions in which a predom- inant T cell signature is causative of . Because Cytokines and acute GVHD a broad spectrum of cytokine is now routinely The initiation of GVHD is necessarily influenced by the cy- used in clinical practice, they are increasingly relevant to tokine milieu in which it arises, and three distinct phases have transplant . The Journal of Immunology, 2015, been described (4, 5). The initial phase is triggered by tissue 194: 4604–4612. damage and associated loss of mucosal barrier function, pri- marily in the gastrointestinal (GI) tract, which is caused by the

conditioning regimens needed to bring malignant disease to by guest on September 25, 2021 raft-versus-host disease (GVHD) is a phenomenon a minimal residual level suitable for subsequent immune control almost unique to allogeneic bone marrow trans- and to ablate existing immune function, allowing engraftment of G plantation (BMT) whereby are intro- the naive donor inoculum. Myeloablative stem cell transplan- duced and permitted to engraft and proliferate within an tation typically uses total body irradiation or busulphan-based immunocompromised host. In this setting, naive (i.e., those that to achieve these dual aims; however, they also have not previously encountered Ag) donor T cells are able to result in damage to the GI tract mucosa and other cells con- recognize host or recipient Ags as foreign, an effect that constitutes tributing to the “cytokine storm,” which is characterized by the the therapeutic intent of BMT, allowing destruction of leukemic release of proinflammatory cytokines: classically TNF, IL-1, and or other malignant cells through activation of pathways of the IL-6 (4, 6). Although less well defined, there is an appreciation adaptive immune response. This beneficial effect is termed “graft- that a similar process occurs with reduced intensity–condition- versus-leukemia” (GVL). The relative contributions of memory ing transplantation, although the dominant cytokines and T cells to GVHD and GVL were discussed elsewhere (1). temporal relationships may differ (7). However, the effect is not specific to malignant cells, and si- In addition to chemotherapy and radiation-induced tissue multaneous damage and destruction of healthy cells and tissues damage and inflammation, recognition of pathogen-associated via the same or similar mechanisms give rise to GVHD. The molecular patterns, such as LPS, and danger-associated mo- morbidity and mortality of GVHD limit the clinical scenarios in lecular patterns arising from GI microbiota have significant which allogeneic hematopoietic stem cell transplantation may bearing on GVHD pathophysiology. The inflammatory sig- otherwise offer therapeutic benefit. Therefore, much research has nals generated in the emerging adaptive immune response are focused on the separation of GVL and GVHD, although success added to by recognition of molecular motifs from both has been limited because of the use of the same immune effector pathogenic and commensal organisms and subsequent acti-

Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Abbreviations used in this article: aGVHD, acute GVHD; BMT, bone marrow trans- Brisbane 4006, Queensland, Australia; and The Royal Brisbane and Women’s , plantation; cGVHD, chronic GVHD; GI, gastrointestinal; GVHD, graft-versus-host Brisbane 4029, Queensland, Australia disease; GVL, graft-versus-leukemia; TFH, T follicular helper; Treg, . Received for publication January 20, 2015. Accepted for publication March 19, 2015. Copyright 2015 by The American Association of Immunologists, Inc. 0022-1767/15/$25.00 Address correspondence and reprint requests to Prof. Geoffrey R. Hill, QIMR Berghofer Medical Research Institute, 300 Herston , Herston, Brisbane 4006, QLD, Australia. E-mail address: [email protected]

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1500117 The Journal of Immunology 4605 vation of innate lymphoid pathways. The diversity of the and . This tissue damage is mediated by more than one resident organisms can be affected by conditioning-associated immunological mechanism. First, cognate T cell–MHC inflammation and by GVHD itself; conversely, the microbiota interactions are required for effector, usually CD8, T cells that present can influence the severity of GVHD (8). The quan- are able to evoke cytolytic machinery, including perforins and titative and qualitative contributions of this microbiota-driven granzymes that induce target cell via apoptosis (5, 6, inflammatory signal are influenced by the variety and path- 28). Interestingly, granzyme B–deficient donor T cells me- ogenicity of organisms present, and has been demonstrated to diate less severe GVHD but may still generate GVL (29), via affect the severity of GVHD (8–10). A reduction in the reduced activation-induced death of CD8 T cells (30). In bacterial burden by use of antimicrobial decontamination in a complementary pathway in which cognate T cell–MHC the posttransplant period also can reduce GVHD severity interactions are not required, myeloid cells, in addition to (10). Although our mechanistic understanding of this effect is lymphoid cells, are primed during aGVHD to release cyto- not complete, it is apparent that GVHD mediates a loss of pathic quantities of inflammatory cytokines (e.g., TNF, IL-6) Paneth cell–derived antimicrobial peptides that play an im- that directly invoke apoptosis (31). Importantly, TNF is also portant role in shaping the diversity of microbiota (11), in involved in GVL effects, and inhibition can compromise addition to the use of pharmaceutical antimicrobials (9). antitumor (32). Damage to the primary target Understanding these mechanisms may offer manipulable organs of GVHD is driven by chemokine expression that targets to alter this primary, inflammation-mediated, initia- results in tissue homing of populations. LPAM-1 tion phase of GVHD. (a4b7 ) and L-selectin (CD62L) are associated with Downloaded from Recognition of the range of triggers to the cytokine storm homing to GI and GALTs and cutaneous lymphoid Ag to complements our knowledge of subsequent T cell and APC skin (33) and are necessary for induction of GVHD tissue interactions that define the second phase of acute GVHD damage at these sites (34). Cytokines, including IFN-g, are (aGVHD) pathophysiology and during which cytokines play known to induce upregulation of chemokines and recep- a key role in driving naive T cell differentiation and expansion tors (35), and these mechanisms were shown to be important toward one maturation program or another. Type 1 or Tc1/ in determining the severity of GVHD within an inflam- http://www.jimmunol.org/ Th1 maturation is recognized as the dominant pattern in matory environment (36–38). The expression of mole- aGVHD (12, 13), and is linked to severe GI tract pathology cules associated with lymphocyte exhaustion (e.g., PD1) and (14). Indeed, in animal models of aGVHD, Th2 and regu- their ligands (e.g., PD-L1) on nonlymphoid tissue also was latory T cells (Tregs) are rare (15). T cells expressing IL-17 are shown to be cytokine (IFN) dependent and contribute to the also rare, although this may reflect the plasticity of this lineage constraint of lymphocyte-mediated tissue damage late in (16). Increased quantities of Th1-associated cytokines, TNF the aGVHD setting (39, 40). Therefore, the inflamma- and IFN-g, in aGVHD are associated with earlier onset and tory cytokines present in this setting participate in positive-

more severe disease in preclinical models and clinical BMT (4, and negative-feedback loops in both lymphocyte and non- by guest on September 25, 2021 14, 17–19). Although the dominance of Th1 subsets is well lymphocyte populations. established, Th2 and Th17 subsets are also involved in pa- thology, and the balance between subsets determines aGVHD Cytokines and cGVHD severity (20), in addition to organ specificity (15, 20), and the cGVHD represents a distinct pathophysiological entity from pathogenic or protective effects of any subset cannot be aGVHD that traditionally is separated by time of onset; how- viewed in isolation. Implicit in the known reciprocal regula- ever, it is now recognized by its distinct end-organ pathology. tion of T cell differentiation by these cytokines is the concept Although the cardinal feature of aGVHD is apoptosis, fibrosis that inhibition of any one lineage may provoke unwanted and is the predominant mechanism of tissue damage in cGVHD. exaggerated differentiation down alternative differentiation Additionally, primary target organs also differ, with and pathways. skin being the primary target organs in cGVHD, manifesting Th2 differentiation is often seen as opposing Th1 differ- as obliterans and (41, 42). Sicca entiation; however, this subset is also recognized to cause symptoms secondary to salivary and lacrimal gland destruc- aGVHD but with predominant pathology in pulmonary, tion and oral lichenoid GVHD are also prominent. Despite hepatic, and cutaneous tissues (21), in contrast to the strong these disparate pathophysiological manifestations, clear roles GI association with Th1. Cutaneous pathology also may be for cytokine control of this phase of disease also were dem- generated by Th17 cells; although they are more commonly onstrated, unaccompanied by large-scale conditioning-related associated with chronic GVHD (cGVHD), they also have tissue damage and the “cytokine storm” that initiates aGVHD. been associated with acute pathology (22–24). Th17 differ- IL-17 and subsequent T cell differentiation along the Th17 entiation is initiated by IL-6 (25), and RORgt is the defining pathway are becoming more strongly associated with cGVHD. transcription factor (26), whereas maintenance and amplifi- Initially identified with the use of G-CSF in stem cell mo- cation relies on IL-23 and IL-21, respectively (27). The use of bilization of donors and prominent Th17 differentiation (43), RORC-deficient donor T cells results in attenuated aGVHD IL-17 was shown more recently to result in CSF1-dependent severity and lethality (26). Further studies are needed to better accumulation in skin and lung, which drives tis- define the role of this subset in late aGVHD versus early sue fibrosis (44). We demonstrated recently that, consistent cGVHD, as well as the relative contribution of IL-17 from with this, systemic IL-17 levels increase late after clinical CD4 and CD8 T cells to end-organ pathology. BMT, at a time when cGVHD develops (45). It is also clear The third and final effector phase of aGVHD is charac- that T follicular helper (TFH) cells and IL-21 play important terized by target tissue damage, with the hallmark histological roles in the development of cGVHD via the stimulation of finding being apoptosis, most commonly in the GI tract, liver, germinal center B cells and alloantibody generation (46). 4606 BRIEF REVIEWS: CYTOKINES IN GVHD

This is particularly relevant to bronchiolitis obliterans, be- on nonhematopoietic tissues. Donor lymphocyte IFN-g cause preliminary evidence suggests that Th17 differentia- signaling enhanced GVHD via the promotion of Th1 dif- tion and CSF1 dysregulation are also involved in this ferentiation, and it also is directly cytotoxic to gut mucosa aberrant immunological pathway (44). Thus, inhibition of (18). Tissue-specific effects are also seen in pulmonary pa- Th17 differentiation and CSF1 appear highly relevant to renchyma in which a protective role for IFN-g was dem- the prevention and treatment of cGVHD. Inhibition of ter- onstrated and these effects have also been described by minal cytokines involved in fibrosis, such as TGF-b and other groups (15). IFN-g provides evidence for a paradigm IL-13, represent additional targets; however, TGF-b inhi- where cytokines may exert effects in nonhematopoietic tissue, bition is problematic given its important role in Treg ho- in addition to specific effects on lymphocytes and other he- meostasis (47). matopoietic cells. Evidence of similar patterns for other cy- tokines is continually being defined and allows selection of Cytokines and T cell–differentiation programs appropriate targets for inhibition in the . With regard to other Th1-associated cytokines, a similarly The ability of cytokines to drive cellular differentiation is complex effect is seen for IL-2, both mechanistically and in recognized in situations other than GVHD, with the accep- therapeutic outcomes (51). Initially used at a high dose in an tance that derivation of phenotypically distinct erythroid, attempt to augment proliferation of lymphocytes as “immu- myeloid, and lymphoid populations from a common long- term hematopoietic stem cell is dependent upon binding of notherapy” for solid malignancies (52, 53), it was subse- cytokines to their cognate receptors (48, 49). Similarly, the quently found, paradoxically, to have a critical role in sup- Downloaded from maturation of the naive T cell population in the context of porting Treg populations and in controlling GVHD (51, BMT and GVHD is also driven by cytokines and subsequent 54). The promotion of regulatory pathways in GVHD was transcriptional pathways elicited thereafter (50). A summary demonstrated in small numbers of patients when used in of these effects is shown in Fig. 1 and outlined further below. a “low dose” (55). These apparently dose-dependent effects g are likely explained by competition for consumption of this Cytokines and Th1 differentiation. IFN- , IL-2, and TNF are http://www.jimmunol.org/ the key cytokines generated during Th1 differentiation (14), cytokine by maturing Treg and T effector cell populations and phenotypic differentiation is initiated by IL-12 and (56); in the clinical transplantation scenario, they are further controlled by the transcription factor T-bet (25, 48). IFN-g complicated by the use of inhibitors, which also in this setting participates in positive feedback to reinforce Th1 target this pathway, when used as GVHD prophylaxis post- responses, in addition to exerting effects on nonlymphocytes, transplant. as well as on nonhematopoietic cells (18). Initial attempts Cytokines and Th2 differentiation. The presence of IL-25 and, to define the role of IFN-g in determining aGVHD severity subsequently, IL-4 supports the development of T cells of were hampered by conflicting data supporting the exacerba- the Th2 lineage that traditionally have been described as tion and amelioration of pathology; however, subsequent being involved in allergy and host defense against parasites by guest on September 25, 2021 work demonstrated this to be related to differing effects on and helminths. Th2 cells produce IL-4, IL-5, IL-10, and donor and host tissues, in addition to tissue-specific effects IL-13, with transcriptional control exerted by GATA3 (57).

FIGURE 1. Cytokine drivers in the three phases of aGVHD initiation and end-organ pathology. Initial inflammatory signals are elicited by cellular damage from chemo- and radiotherapy, in addition to those derived from following GI tract damage and loss of integrity. Cytokines act on naive T cell, ILC, and myeloid cell populations, resulting in differentiation to Th1, Th2, and Th17 cell subsets, activated ILC subsets, and activated myeloid cells. End-organ tissue damage in aGVHD is caused by apoptosis elicited by Th1/Tc1 cytokines and cytolytic machinery, including perforin and granzyme, following cognate TCR–MHC interactions. Additional inflammatory pathways that are not dependent on cognate T cell pathways, including IL-6– and TNF–mediated apoptosis, following release of these cytokines from activated and macrophage populations. End-organ damage in cGVHD classically follows aGVHD and is mediated by Th2/Th17 cells and monocyte/macrophage populations secreting TGF-b that result in tissue fibrosis. The influence of ILCs on GVHD requires further de- lineation but they may be regulatory, at least early after BMT. The Journal of Immunology 4607

In aGVHD, the Th2 program appears to mediate skin and –derived IL-10 being protective in the initiation of lung pathology (15, 18), as opposed to the strong association aGVHD (61). A role for innate lymphoid cells as cytokine- of Th1 cells with gut and liver damage. Recent work dem- responsive mediators of protection from GVHD is emerging onstrated a role for an IL-25–dependent immature non-B (72), with ILC1, ILC2, and ILC3 subtypes demonstrating non-T cell innate immune effector cell population that is re- similar transcriptional control and cytokine profiles to Th1, sponsible for propagation of Th2 responses and production Th2, and Th17 cells (73). of IL-4, IL-5, and IL-13; where lacking, this results in an Donor and host cells. The effect of a single cytokine can impaired ability to expel helminths (58–60). This cytokine be dependent upon its roles in hematopoietic and non- may be seen as having protective effects on GI tissues, an hematopoietic tissue; however, in BMT, donor or host origin of outcome that is clearly attractive in the context of aGVHD the cell transducing the signal is as an additional factor influ- pathology. encing outcomes. Type I IFN is an example of a cytokine for Other Th2 cytokines were shown to have protective roles in which signaling through recipient APCs results in less severe class GVHD, including IL-10. Despite mixed results when initially II–dependent GVHD in the colon, whereas signaling through given as to patients, its production by B lymphocytes donor APCs may amplify GVHD responses. The former is in animal models of transplantation reduces the severity of mediated through decreased donor CD4 proliferation, and the GVHD (61). These outcomes were mediated by effects on latter is mediated through more effective cross-presentation of donor T cell expansion; similarly, reductions in IL-4 and IL- alloantigens to CD8 T cells (19). IL-4 is another example. A

10 were demonstrated in patients with cGVHD (62, 63). subpopulation of recipient NKT cells secretes high levels of IL-4 Downloaded from Cytokines and Th17 differentiation. Th17 cells are a more recent and indirectly expands donor Treg populations to promote addition to the Th1/Th2 paradigm (25), and roles for IL-17– tolerance after BMT (74). In contrast, IL-4 may drive donor producing cells of both Th17 and Tc17 varieties are still being Th2 differentiation directly and enhance GVHD that likely defined in the GVHD setting and in other immune usually represents chronic disease. Appreciation of these (64). Initiation of Th17 development is triggered by IL-6 and mechanisms is important, because treatment of a recipient or TGF-b and is associated with transcriptional activation of graft can be temporally separated and offers the opportunity to http://www.jimmunol.org/ RORgt after phosphorylation of STAT3, as well as with the select desirable effects while avoiding potentially deleterious secretion of IL-17, IL-21, and IL-22. There is an increasing outcomes. appreciation for the role that Th17 cells play in determining disposition. Additional complicating factors exist when the severity of GVHD (65), with a particular role for IL-6 considering cytokine-mediated effects in immune-mediated and becoming apparent (66, 67). Recently, our group demonstrated inflammatory conditions. IL-6 is an example of a cytokine for the importance of this effect in cohorts in which IL-6 inhibition which signaling via “classical” or membrane-bound receptor– represents a potentially effective therapeutic strategy to reduce the ligand interactions produces differing pathology than does severity of aGVHD in clinical stem cell transplantation (45). IL- signaling mediated through soluble or trans receptor binding. by guest on September 25, 2021 22maybesecretedbyTh17cellsand, in this setting, it appears to These effects were described originally in mouse models of be pathogenic (68); conversely, it may be secreted by innate , in which trans signaling (by the IL-6– lymphoid cells where, in the GI tract at least, it appears to be soluble IL-6R complex) recapitulated inflammatory joint an important protective cytokine (69). IL-21 can be produced by disease in IL-6–deficient mice, whereas injection of the both Th17 and TFH cells, and it promotes aGVHD by impairing native IL-6 cytokine itself did not (75). Subsequently, IL-6 Treg homeostasis (70). Given that it also has an important role in signaling through the trans pathway has been thought to be inducing aberrant, allospecific germinal center B cell responses more inflammatory in nature than classical signaling, in part and cGVHD, inhibition of this Th17-associated cytokine rep- relating to the ability of IL-6 to signal through cells that ex- resents another attractive therapeutic target for GVHD control press the gp130 receptor complex but do not basally express after BMT. IL-6R (76). A similar paradigm was demonstrated in al- lergic : Th2 expansion appears to be driven by trans Heterogeneity of effect conferred by cellular target signaling whereby expansion of Tregs was limited by classical Increasingly, the effects mediated by a particular cytokine are IL-6 signaling, and inhibition with anti–IL-6R mAb resulted being defined as dependent on the cells in which it transduces in increased numbers of Tregs (77), and an increase in asthma a signal and may be considered to regulate effector cell pop- risk was associated with a single nucleotide polymorphism ulations, as well as to confer susceptibility or protection to other that results in an increase in soluble IL-6R and trans signaling inflammatory signals in target organs and tissues. Modes of (78). Appreciation of the mechanisms by which a cytokine can signaling also influence these responses, with a recent appreci- mediate differential effects is critical to understanding both ation for the differential pathology induced by the binding of disease pathophysiology and effective clinical translation of cytokines by membrane-bound receptors as opposed to soluble therapeutics. The availability of mAbs to cytokine receptors, receptors. such as for IL-6R, which inhibits all IL-6 signaling, Hematopoietic and nonhematopoietic cells. Cytokines may exert in addition to more specific inhibitors of signalling pathway effects on cells of hematopoietic origin, in addition to non- components, such as soluble gp130:Fc, which inhibits IL-6 hematopoietic target tissues directly. IFN-g (18) and IL-22 trans signaling only, is a clear example. (68, 69) are clear examples that were already discussed. Further examples are seen in cGVHD: IL-2, IL-10, and Translational application TGF-b may act directly on tissue fibroblasts in affected Accepted murine models of transplantation and rapid and organs to mediate pathology (71), in addition to known reproducible multiplexed techniques to measure cytokines in effects of IL-2 in supporting Treg populations (51, 55) and serum or cell culture supernatants or intracellular cytokine 4608 BRIEF REVIEWS: CYTOKINES IN GVHD production by flow cytometry have allowed identification of, tion will require validation ofobservationsmadeinanimal and will continue to define, the key cytokines in aGVHD models with clinical cohorts, because standard immune- (45, 79), as well as facilitate clinical translation of findings. suppressing agents were shown to affect cytokine levels pro- However, a number of factors must be considered when ex- duced by T cells and NK cells, and profiles vary with stem cell trapolating laboratory observations into clinical cohorts. source (80, 81). Importantly, IFN-g, TNF, and IL-1 are not Variation exists in transplantation protocols, patient pop- systemically dysregulated in clinical subjects after BMT who ulations, modes of conditioning, and posttransplant immune- receive immune suppression in the same way as seen in rodent suppression strategies. The last factor is of particular im- models (45). With this in mind, it should be noted that no portance when considering the translation of observations cytokine-inhibition strategy or cytokine administration has made in animal models to the clinical setting, where immune proved efficacious in randomized studies. In general, en- suppression with cyclosporin or , combined with couraging results seen in preclinical studies and early-phase or mycophenolate,isconsideredstandardof clinical trials have either not progressed into phase III studies, care to avoid life-threatening acute and severe GVHD. How- or effects have not been robust within this context [e.g., IL-1 ever, most animal models of transplantation rely solely on (82) and TNF inhibition (83, 84)]. Table I provides a sum- radiation-based conditioning. Therefore, effective transla- mary of cytokines and inhibitors that have been explored for

Table I. Summary of relevant cytokine-targeted therapeutic studies Downloaded from

Randomized, Double-Blind Controlled and/or Phase III Cytokine Inhibitors Phase I and/or II Clinical Trial Data Clinical Trial Data TNF-aR2 () Prophylaxis + (83) •

Treatment + (102–105) • http://www.jimmunol.org/ TNF-a binding mAb (infliximab) Prophylaxis 2 (84) • Treatment + (99, 100) 2 (101) IL-1Ra () Prophylaxis 2 (82) Treatment + (106) • IL-2Ra/anti-CD25 (/daclizumaba) Prophylaxis + (107) • Treatment + (108–110) 2 (111, 112) IL-6R (tocilizumab) Prophylaxis + (45) • by guest on September 25, 2021 Treatment + (113, 114) • Keratinocyte growth factor (palifermin) Prophylaxis 2 (115–119) 2 (120)

Cytokines IL-2 (aldesleukin) Prophylaxis + (55) • Treatment + (51) • IL-11 () Prophylaxis 2 (121) •

Cytokines with Non-GVHD Benefits IFN-a (INTRON A, Roferon-A)—promotion of GVL with concomitant promotion of GVHD Prophylaxis + (122–126) • Treatment + (127, 128) • Keratinocyte growth factor (palifermin)—for reduction of oral mucositis Prophylaxis + (116–118) + (120)

Phase I and/or II Clinical Phase III Clinical Trial Potential GVHD Therapies Data Outside GVHD Data Outside GVHD IL-17 IL-17A mAb (, , perakizumab) + Ixekizumab (129) + Secukinumab (131) IL-17RA mAb () + (130) • IL-17A/TNF (ABT122) Ongoing • IL-22 () Ongoing • IL-12p40/23 mAb () + (97, 132) • IL-23p19 (, ) + Guselkumab (133) + Tildrakizumab (134) Tildrakizumab (ongoing) IL-13 (, ) + Tralokinumab (135) + Lebrikizumab (136) Tralokinumab (ongoing) Cytokines and their antagonists are included that have been tested within a trial setting to prevent or treat GVHD or other complications of allogeneic BMT. Also included is a list of newer therapeutics with potential application to GVHD that are undergoing testing in other disease settings. +, positive data; •, lack of data in this setting; 2, negative data. The Journal of Immunology 4609 efficacy in the treatment or prevention of GVHD, in addition effective prevention and treatment for GVHD. In the future, to agents with potential efficacy in GVHD that are being cytokine therapies may become a standard component of explored in other disease settings. Importantly, most studies transplantation protocols as we have seen in other inflam- in GVHD examine the usefulness of cytokine antagonists as matory and autoimmune pathologies. an adjunct to standard modalities of immune suppression rather than their efficacy in isolation, as is usually the case in Disclosures preclinical testing. Thus, it will be important to follow some recently defined general principles, taking into consideration The authors have no financial conflicts of interest. concurrent immune suppression, when planning to translate findings from mice to patients (28). References The effect of a particular cytokine in any one individual is 1. Anderson, B. E., H. Zheng, P. A. Taylor, C. Matte-Martone, J. M. McNiff, also affected by human genetic heterogeneity, and data already D. Jain, A. J. Demetris, A. Panoskaltsis-Mortari, A. Ager, B. R. Blazar, et al. 2008. Memory T cells in GVHD and GVL. Biol. Marrow Transplant. 14(1, Suppl. demonstrated a clear impact of single nucleotide polymorphisms 1)19–20. in cytokine loci on GVHD outcomes (85, 86). Despite these 2. Wagner, J. E., J. S. Thompson, S. L. Carter, and N. A. Kernan, Unrelated Donor Marrow Transplantation Trial. 2005. Effect of graft-versus-host disease prophy- difficulties in directly translating laboratory observations to the laxis on 3-year disease-free survival in recipients of unrelated donor bone marrow clinic, cytokine therapy for GVHD remains fertile ground for (T-cell Depletion Trial): a multi-centre, randomised phase II-III trial. Lancet 366: new and effective therapeutics, because a number of agents that 733–741. 3. Horowitz, M. M., R. P. Gale, P. M. Sondel, J. M. Goldman, J. Kersey, H. J. Kolb, augment or antagonize cytokine pathways are already available, A. A. Rimm, O. Ringde´n, C. Rozman, B. Speck, et al. 1990. Graft-versus- Downloaded from having been explored and validated in other autoimmune and leukemia reactions after bone marrow transplantation. Blood 75: 555–562. 4. Hill, G. R., J. M. Crawford, K. R. Cooke, Y. S. Brinson, L. Pan, and J. L. Ferrara. inflammatory disease settings. Cytokine inhibition, initially 1997. Total body irradiation and acute graft-versus-host disease: the role of gas- with TNF and subsequently with IL-6, is considered routine trointestinal damage and inflammatory cytokines. Blood 90: 3204–3213. 5. Ferrara, J. L., and H. J. Deeg. 1991. Graft-versus-host disease. N. Engl. J. Med. care for rheumatoid arthritis patients whose disease is not 324: 667–674. controlled by more nonspecific immune suppression with 6. Hill, G. R. 2009. Inflammation and bone marrow transplantation. Biol. Blood Marrow Transplant. 15(1, Suppl.)139–141. http://www.jimmunol.org/ , methotrexate, and calcineurin inhibitors (87– 7. Mohty, M., D. Blaise, C. Faucher, N. Vey, R. Bouabdallah, A. M. Stoppa, 90). Imperfect disease control, when used as monotherapy, has F. Viret, G. Gravis, D. Olive, and B. Gaugler. 2005. Inflammatory cytokines and paved the way for the use of combination cytokine inhibition, acute graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation. Blood 106: 4407–4411. and the rational combination of TNF and IL-17 showed efficacy 8. Jenq, R. R., C. Ubeda, Y. Taur, C. C. Menezes, R. Khanin, J. A. Dudakov, C. Liu, in preclinical models of disease (91). Novel targets, such as IL- M. L. West, N. V. Singer, M. J. Equinda, et al. 2012. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J. 32,IL-34,andIL-35,arealsobeing explored (92). The success Exp. Med. 209: 903–911. of cytokine inhibition in rheumatoid arthritis is also paralleled in 9. Holler, E., P. Butzhammer, K. Schmid, C. Hundsrucker, J. Koestler, K. Peter, W. Zhu, D. Sporrer, T. Hehlgans, M. Kreutz, et al. 2014. Metagenomic analysis other inflammatory , including psoriatic and of the stool microbiome in patients receiving allogeneic stem cell transplantation:

arthritis (93), with TNF inhibition having a demonstrated role, loss of diversity is associated with use of systemic and more pronounced by guest on September 25, 2021 in addition to promising newer targets, such as IL-22 and IL-23 in gastrointestinal graft-versus-host disease. Biol. Blood Marrow Transplant. 20: 640–645. (94, 95). Evidence for the value of cytokine inhibition exists in 10. Beelen, D. W., A. Elmaagacli, K. D. Muller,€ H. Hirche, and U. W. Schaefer. diseases other than inflammatory arthropathies, with efficacy 1999. Influence of intestinal bacterial decontamination using metronidazole and ciprofloxacin or ciprofloxacin alone on the development of acute graft-versus-host demonstratedforTNF(96),aswellasIL-12/23(97,98),in disease after marrow transplantation in patients with hematologic malignancies: inflammatory bowel disease. In this setting of proven efficacy for final results and long-term follow-up of an open-label prospective randomized trial. Blood 93: 3267–3275. cytokine inhibition in other diseases of dysregulated immunity, 11. Eriguchi, Y., S. Takashima, H. Oka, S. Shimoji, K. Nakamura, H. Uryu, further definition of the role of cytokines in the determination of S. Shimoda, H. Iwasaki, N. Shimono, T. Ayabe, et al. 2012. Graft-versus-host disease disrupts intestinal microbial ecology by inhibiting Paneth cell production of GVHD severity is likely to translate rapidly into efficacious a-defensins. Blood 120: 223–231. therapies for the transplant patient population. 12. Teshima, T., Y. Maeda, and K. Ozaki. 2011. Regulatory T cells and IL-17- producing cells in graft-versus-host disease. 3: 833–852. 13. Pan, B., Y. Zhang, Y. Sun, H. Cheng, Y. Wu, G. Song, W. Chen, L. Zeng, and Conclusions K. Xu. 2014. Deviated balance between Th1 and Th17 cells exacerbates acute graft-versus-host disease in mice. Cytokine 68: 69–75. Cytokines are a defining influence on evolving immune 14. Hill, G. R., and J. L. Ferrara. 2000. The primacy of the as responses in BMT and in the generation of GVHD, the major a target organ of acute graft-versus-host disease: rationale for the use of cytokine pathology limiting the wider application of transplantation. shields in allogeneic bone marrow transplantation. Blood 95: 2754–2759. 15. Yi, T., Y. Chen, L. Wang, G. Du, D. Huang, D. Zhao, H. Johnston, J. Young, The classical appreciation of a naive T cell being influenced by I. Todorov, D. T. Umetsu, et al. 2009. Reciprocal differentiation and tissue- a cytokine to mature into a more differentiated phenotype is specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease. Blood 114: 3101–3112. made more complex in the GVHD setting as the impact of 16. Hirota, K., J. H. Duarte, M. Veldhoen, E. Hornsby, Y. Li, D. J. Cua, H. Ahlfors, cytokines acting in different tissue compartments (e.g., lym- C. Wilhelm, M. Tolaini, U. Menzel, et al. 2011. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12: 255–263. phoid and nonlymphoid or donor and host) and the use of 17. Xun, C. Q., J. S. Thompson, C. D. Jennings, S. A. Brown, and M. B. Widmer. classical and trans signaling pathways become better appreci- 1994. Effect of total body irradiation, busulfan-, or cyclophos- phamide conditioning on inflammatory cytokine release and development of acute ated. Although these factors require further work to better and chronic graft-versus-host disease in H-2-incompatible transplanted SCID define these complex interactions, they go some way in mice. Blood 83: 2360–2367. explaining the previously mixed and conflicting results asso- 18. Burman, A. C., T. Banovic, R. D. Kuns, A. D. Clouston, A. C. Stanley, E. S. Morris, V. Rowe, H. Bofinger, R. Skoczylas, N. Raffelt, et al. 2007. IFN- ciated with some cytokine therapies (99–101). Appreciation gamma differentially controls the development of idiopathic pneumonia syndrome of this complexity will allow for the development of more and GVHD of the gastrointestinal tract. Blood 110: 1064–1072. 19. Robb, R. J., E. Kreijveld, R. D. Kuns, Y. A. Wilson, S. D. Olver, A. L. Don, logical trials in this field, and early positive results are already N. C. Raffelt, N. A. De Weerd, K. E. Lineburg, A. Varelias, et al. 2011. Type I- being realized (45). The use of murine models of disease, in IFNs control GVHD and GVL responses after transplantation. Blood 118: 3399– 3409. parallel with observations in clinical cohorts, will allow further 20. Yi, T., D. Zhao, C.-L. Lin, C. Zhang, Y. Chen, I. Todorov, T. LeBon, F. Kandeel, refinement of our current understanding and result in more S. Forman, and D. Zeng. 2008. Absence of donor Th17 leads to augmented Th1 4610 BRIEF REVIEWS: CYTOKINES IN GVHD

differentiation and exacerbated acute graft-versus-host disease. Blood 112: 2101– 45. Kennedy, G. A., A. Varelias, S. Vuckovic, L. Le Texier, K. H. Gartlan, P. Zhang, 2110. G. Thomas, L. Anderson, G. Boyle, N. Cloonan, et al. 2014. Addition of 21. Nikolic, B., S. Lee, R. T. Bronson, M. J. Grusby, and M. Sykes. 2000. Th1 and -6 inhibition with tocilizumab to standard graft-versus-host disease Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets. J. prophylaxis after allogeneic stem-cell transplantation: a phase 1/2 trial. Lancet Clin. Invest. 105: 1289–1298. Oncol. 15: 1451–1459. 22. Carlson, M. J., M. L. West, J. M. Coghill, A. Panoskaltsis-Mortari, B. R. Blazar, 46. Flynn, R., J. Du, R. G. Veenstra, D. K. Reichenbach, A. Panoskaltsis-Mortari, and J. S. Serody. 2009. In vitro-differentiated TH17 cells mediate lethal acute P. A. Taylor, G. J. Freeman, J. S. Serody, W. J. Murphy, D. H. Munn, et al. 2014. graft-versus-host disease with severe cutaneous and pulmonary pathologic mani- Increased T follicular helper cells and germinal center B cells are required for festations. Blood 113: 1365–1374. cGVHD and bronchiolitis obliterans. Blood 123: 3988–3998. 23.Varelias,A.,K.H.Gartlan,E.Kreijveld,S.D.Olver,M.Lor,R.D.Kuns, 47. Banovic, T., K. P. MacDonald, E. S. Morris, V. Rowe, R. Kuns, A. Don, J. Kelly, K. E. Lineburg, B. E. Teal, N. C. Raffelt, M. Cheong, et al. 2015. Lung parenchyma- S. Ledbetter, A. D. Clouston, and G. R. Hill. 2005. TGF-beta in allogeneic stem derived IL-6 promotes IL-17A-dependent acute lung injury after allogeneic stem cell transplantation: friend or foe? Blood 106: 2206–2214. cell transplantation. Blood. DOI: 10.1182/blood-2014-07-590232. 48. Lotem, J., and L. Sachs. 2002. Cytokine control of developmental programs in 24. Kappel, L. W., G. L. Goldberg, C. G. King, D. Y. Suh, O. M. Smith, C. Ligh, normal hematopoiesis and leukemia. Oncogene 21: 3284–3294. A. M. Holland, J. Grubin, N. M. Mark, C. Liu, et al. 2009. IL-17 contributes to 49. Metcalf, D. 2008. Hematopoietic cytokines. Blood 111: 485–491. CD4-mediated graft-versus-host disease. Blood 113: 945–952. 50. Weaver, C. T., R. D. Hatton, P. R. Mangan, and L. E. Harrington. 2007. IL-17 25. Steinman, L. 2007. A brief history of TH17, the first major revision in the TH1/ cytokines and the expanding diversity of effector T cell lineages. Annu. Rev. TH2 hypothesis of T cell–mediated tissue damage. Nat. Med. 13: 139–145. Immunol. 25: 821–852. 26. Fulton, L. M., M. J. Carlson, J. M. Coghill, L. E. Ott, M. L. West, 51. Koreth, J., K. Matsuoka, H. T. Kim, S. M. McDonough, B. Bindra, E. P. Alyea, A. Panoskaltsis-Mortari, D. R. Littman, B. R. Blazar, and J. S. Serody. 2012. III, P. Armand, C. Cutler, V. T. Ho, N. S. Treister, et al. 2011. Interleukin-2 and Attenuation of acute graft-versus-host disease in the absence of the transcription regulatory T cells in graft-versus-host disease. N. Engl. J. Med. 365: 2055–2066. factor RORgt. J. Immunol. 189: 1765–1772. 52. Dutcher, J. 2002. Current status of interleukin-2 therapy for metastatic renal cell 27. Serody, J. S., and G. R. Hill. 2012. The IL-17 differentiation pathway and its role carcinoma and metastatic melanoma. (Huntington) 16: 4–10. in transplant outcome. Biol. Blood Marrow Transplant. 18(1, Suppl.)S56–S61. 53. Murphy, W. J., J. L. M. Ferrara, and T. Malek. 2012. A delicate balance: tweaking 28. Markey, K. A., K. P. MacDonald, and G. R. Hill. 2014. The biology of graft-versus- IL-2 immunotherapy. Nat. Med. 18: 208–209. host disease: experimental systems instructing clinical practice. Blood 124: 354–362. 54. Pe´rol, L., G. H. Martin, S. Maury, J. L. Cohen, and E. Piaggio. 2014. Potential Downloaded from 29. Graubert, T. A., J. F. DiPersio, J. H. Russell, and T. J. Ley. 1997. Perforin/ limitations of IL-2 administration for the treatment of experimental acute graft- granzyme-dependent and independent mechanisms are both important for the versus-host disease. Immunol. Lett. 162(2 Pt B): 173–184. development of graft-versus-host disease after murine bone marrow transplanta- 55. Kennedy-Nasser, A. A., S. Ku, P. Castillo-Caro, Y. Hazrat, M.-F. Wu, H. Liu, tion. J. Clin. Invest. 100: 904–911. J. Melenhorst, A. J. Barrett, S. Ito, A. Foster, et al. 2014. Ultra low-dose IL-2 for 30. Bian, G., X. Ding, N. D. Leigh, Y. Tang, M. L. Capitano, J. Qiu, P. L. McCarthy, GVHD prophylaxis after allogeneic hematopoietic stem cell transplantation H. Liu, and X. Cao. 2013. Granzyme B-mediated damage of CD8+ T cells impairs mediates expansion of regulatory T cells without diminishing antiviral and anti- graft-versus-tumor effect. J. Immunol. 190: 1341–1350. leukemic activity. Clin. Cancer Res. 20: 2215–2225.

31. Mantovani, A., A. Sica, S. Sozzani, P. Allavena, A. Vecchi, and M. Locati. 2004. 56. Ho¨fer, T., O. Krichevsky, and G. Altan-Bonnet. 2012. Competition for IL-2 between http://www.jimmunol.org/ The chemokine system in diverse forms of macrophage activation and polarization. regulatory and effector T cells to chisel immune responses. Front. Immunol. 3: 268. Trends Immunol. 25: 677–686. 57. Nurieva, R. I., and Y. Chung. 2010. Understanding the development and function 32. Hill, G. R., T. Teshima, A. Gerbitz, L. Pan, K. R. Cooke, Y. S. Brinson, of T follicular helper cells. Cell. Mol. Immunol. 7: 190–197. J. M. Crawford, and J. L. Ferrara. 1999. Differential roles of IL-1 and TNF-alpha 58. Fallon, P. G., S. J. Ballantyne, N. E. Mangan, J. L. Barlow, A. Dasvarma, on graft-versus-host disease and graft versus leukemia. J. Clin. Invest. 104: 459– D. R. Hewett, A. McIlgorm, H. E. Jolin, and A. N. McKenzie. 2006. Identifi- 467. cation of an interleukin (IL)-25-dependent cell population that provides IL-4, IL- 33. Sackstein, R. 2006. A revision of Billingham’s tenets: the central role 5, and IL-13 at the onset of helminth expulsion. J. Exp. Med. 203: 1105–1116. of lymphocyte migration in acute graft-versus-host disease. Biol. Blood Marrow 59. Saenz, S. A., M. C. Siracusa, J. G. Perrigoue, S. P. Spencer, J. F. Urban, Jr., Transplant. 12(1, Suppl. 1)2–8. J. E. Tocker, A. L. Budelsky, M. A. Kleinschek, R. A. Kastelein, T. Kambayashi, 34. Dutt, S., J. Ermann, D. Tseng, Y. P. Liu, T. I. George, C. G. Fathman, and et al. 2010. IL25 elicits a multipotent progenitor cell population that promotes S. Strober. 2005. L-selectin and beta7 integrin on donor CD4 T cells are required T(H)2 cytokine responses. Nature 464: 1362–1366.

for the early migration to host mesenteric lymph nodes and acute colitis of graft- 60. Neill, D. R., S. H. Wong, A. Bellosi, R. J. Flynn, M. Daly, T. K. Langford, by guest on September 25, 2021 versus-host disease. Blood 106: 4009–4015. C. Bucks, C. M. Kane, P. G. Fallon, R. Pannell, et al. 2010. Nuocytes represent a 35. He, T., C. Tang, S. Xu, T. Moyana, and J. Xiang. 2007. gamma new innate effector leukocyte that mediates type-2 immunity. Nature 464: 1367– stimulates cellular maturation of line DC2.4 leading to induction of 1370. efficient responses and antitumor immunity. Cell. Mol. Immunol. 61. Rowe, V., T. Banovic, K. P. MacDonald, R. Kuns, A. L. Don, E. S. Morris, 4: 105–111. A. C. Burman, H. M. Bofinger, A. D. Clouston, and G. R. Hill. 2006. Host B cells 36. Cooke, K. R., A. Gerbitz, J. M. Crawford, T. Teshima, G. R. Hill, A. Tesolin, produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone D. P. Rossignol, and J. L. Ferrara. 2001. LPS antagonism reduces graft-versus-host marrow transplantation. Blood 108: 2485–2492. disease and preserves graft-versus-leukemia activity after experimental bone marrow 62. Tanaka, J., M. Imamura, M. Kasai, S. Hashino, S. Kobayashi, S. Noto, T. Higa, transplantation. J. Clin. Invest. 107: 1581–1589. K. Sakurada, and M. Asaka. 1996. Th2 cytokines (IL-4, IL-10 and IL-13) and IL- 37. He, W., J. J. Racine, H. F. Johnston, X. Li, N. Li, K. Cassady, C. Liu, R. Deng, 12 mRNA expression by concanavalin A-stimulated peripheral blood mononuclear P. Martin, S. Forman, and D. Zeng. 2014. Depletion of host CCR7(+) dendritic cells during chronic graft-versus-host disease. Eur. J. Haematol. 57: 111–113. cells prevented donor T cell tissue tropism in anti-CD3-conditioned recipients. 63. Rozmus, J., K. R. Schultz, K. Wynne, A. Kariminia, P. Satyanarayana, M. Krailo, Biol. Blood Marrow Transplant. 20: 920–928. S. A. Grupp, A. L. Gilman, and F. D. Goldman. 2011. Early and late extensive 38. Li, N., Y. Chen, W. He, T. Yi, D. Zhao, C. Zhang, C.-L. Lin, I. Todorov, chronic graft-versus-host disease in children is characterized by different Th1/Th2 F. Kandeel, S. Forman, and D. Zeng. 2009. Anti-CD3 preconditioning separates cytokine profiles: findings of the Children’s Oncology Group Study ASCT0031. GVL from GVHD via modulating host dendritic cell and donor T-cell migration Biol. Blood Marrow Transplant. 17: 1804–1813. in recipients conditioned with TBI. Blood 113: 953–962. 64. Gaffen, S. L., R. Jain, A. V. Garg, and D. J. Cua. 2014. The IL-23-IL-17 immune 39. Blazar, B. R., B. M. Carreno, A. Panoskaltsis-Mortari, L. Carter, Y. Iwai, axis: from mechanisms to therapeutic testing. Nat. Rev. Immunol. 14: 585–600. H. Yagita, H. Nishimura, and P. A. Taylor. 2003. Blockade of programmed death- 65. van der Waart, A. B., W. J. van der Velden, N. M. Blijlevens, and H. Dolstra. 1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma- 2014. Targeting the IL17 pathway for the prevention of graft-versus-host disease. dependent mechanism. J. Immunol. 171: 1272–1277. Biol. Blood Marrow Transplant. 20: 752–759. 40. Li, X., R. Deng, W. He, C. Liu, M. Wang, J. Young, Z. Meng, C. Du, W. Huang, 66. Tawara, I., M. Koyama, C. Liu, T. Toubai, D. Thomas, R. Evers, P. Chockley, L. Chen, et al. 2012. Loss of B7-H1 expression by recipient parenchymal cells leads E. Nieves, Y. Sun, K. P. Lowler, et al. 2011. Interleukin-6 modulates graft-versus- to expansion of infiltrating donor CD8+ T cells and persistence of graft-versus-host host responses after experimental allogeneic bone marrow transplantation. Clin. disease. J. Immunol. 188: 724–734. Cancer Res. 17: 77–88. 41. Filipovich, A. H., D. Weisdorf, S. Pavletic, G. Socie, J. R. Wingard, S. J. Lee, 67. Chen, X., R. Das, R. Komorowski, A. Beres, M. J. Hessner, M. Mihara, and P. Martin, J. Chien, D. Przepiorka, D. Couriel, et al. 2005. National Institutes of W. R. Drobyski. 2009. Blockade of interleukin-6 signaling augments regulatory Health consensus development project on criteria for clinical trials in chronic graft- T-cell reconstitution and attenuates the severity of graft-versus-host disease. Blood versus-host disease: I. Diagnosis and staging working group report. Biol. Blood 114: 891–900. Marrow Transplant. 11: 945–956. 68. Couturier, M., B. Lamarthe´e, J. Arbez, J.-C. Renauld, C. Bossard, F. Malard, 42. Afessa, B., M. R. Litzow, and A. Tefferi. 2001. Bronchiolitis obliterans and other F. Bonnefoy, M. Mohty, S. Perruche, P. Tiberghien, et al. 2013. IL-22 deficiency late onset non-infectious pulmonary complications in hematopoietic stem cell in donor T cells attenuates murine acute graft-versus-host disease mortality while transplantation. Bone Marrow Transplant. 28: 425–434. sparing the graft-versus-leukemia effect. Leukemia 27: 1527–1537. 43. Hill, G. R., S. D. Olver, R. D. Kuns, A. Varelias, N. C. Raffelt, A. L. Don, 69. Hanash, A. M., J. A. Dudakov, G. Hua, M. H. O’Connor, L. F. Young, K. A. Markey, Y. A. Wilson, M. J. Smyth, Y. Iwakura, et al. 2010. Stem cell N. V. Singer, M. L. West, R. R. Jenq, A. M. Holland, L. W. Kappel, et al. 2012. mobilization with G-CSF induces type 17 differentiation and promotes sclero- Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage derma. Blood 116: 819–828. and regulates sensitivity to graft versus host disease. Immunity 37: 339–350. 44. Alexander, K. A., R. Flynn, K. E. Lineburg, R. D. Kuns, B. E. Teal, S. D. Olver, 70. Bucher, C., L. Koch, C. Vogtenhuber, E. Goren, M. Munger, A. Panoskaltsis- M. Lor, N. C. Raffelt, M. Koyama, L. Leveque, et al. 2014. CSF-1–dependant Mortari, P. Sivakumar, and B.R. Blazar. 2009. IL-21 blockade reduces graft- donor-derived mediate chronic graft-versus-host disease. J. Clin. versus-host disease mortality by supporting inducible T regulatory cell generation. Invest. 124: 4266–4280. Blood 114: 5375–5384. The Journal of Immunology 4611

71. Blazar, B. R., W. J. Murphy, and M. Abedi. 2012. Advances in graft-versus-host et al. 2010. Infliximab, , or combination therapy for Crohn’s disease. disease biology and therapy. Nat. Rev. Immunol. 12: 443–458. N. Engl. J. Med. 362: 1383–1395. 72. Munneke, J. M., A. T. Bjo¨rklund, J. M. Mjo¨sberg, K. Garming-Legert, 97. Burakoff, R., C. F. Barish, D. Riff, R. Pruitt, W. Y. Chey, F. A. Farraye, I. Shafran, J. H. Bernink, B. Blom, C. Huisman, M. H. van Oers, H. Spits, K.-J. Malmberg, S. Katz, C. L. Krone, M. Vander Vliet, et al. 2006. A phase 1/2A trial of STA and M. D. Hazenberg. 2014. Activated innate lymphoid cells are associated with 5326, an oral interleukin-12/23 inhibitor, in patients with active moderate to a reduced susceptibility to graft-versus-host disease. Blood 124: 812–821. severe Crohn’s disease. Inflamm. Bowel Dis. 12: 558–565. 73. Spits, H., and T. Cupedo. 2012. Innate lymphoid cells: emerging insights in de- 98. Sandborn, W. J., B. G. Feagan, R. N. Fedorak, E. Scherl, M. R. Fleisher, S. Katz, velopment, lineage relationships, and function. Annu. Rev. Immunol. 30: 647–675. J. Johanns, M. Blank, and P. Rutgeerts, Ustekinumab Crohn’s Disease Study 74. Pillai, A. B., T. I. George, S. Dutt, and S. Strober. 2009. Host natural killer T cells Group. 2008. A randomized trial of Ustekinumab, a human interleukin-12/23 induce an interleukin-4-dependent expansion of donor CD4+CD25+Foxp3+ T monoclonal , in patients with moderate-to-severe Crohn’s disease. Gas- regulatory cells that protects against graft-versus-host disease. Blood 113: 4458–4467. troenterology 135: 1130–1141. 75. Nowell, M. A., P. J. Richards, S. Horiuchi, N. Yamamoto, S. Rose-John, 99. Patriarca, F., A. Sperotto, D. Damiani, G. Morreale, F. Bonifazi, A. Olivieri, N. Topley, A. S. Williams, and S. A. Jones. 2003. Soluble IL-6 receptor governs F. Ciceri, G. Milone, S. Cesaro, G. Bandini, et al. 2004. Infliximab treatment for IL-6 activity in experimental arthritis: blockade of arthritis severity by soluble steroid-refractory acute graft-versus-host disease. Haematologica 89: 1352–1359. . J. Immunol. 171: 3202–3209. 100. Couriel, D., R. Saliba, K. Hicks, C. Ippoliti, M. de Lima, C. Hosing, I. Khouri, 76. Scheller, J., N. Ohnesorge, and S. Rose-John. 2006. Interleukin-6 trans-signalling B. Andersson, J. Gajewski, M. Donato, et al. 2004. -alpha in chronic inflammation and cancer. Scand. J. Immunol. 63: 321–329. blockade for the treatment of acute GVHD. Blood 104: 649–654. 77. Doganci, A., T. Eigenbrod, N. Krug, G. T. De Sanctis, M. Hausding, 101. Couriel, D. R., R. Saliba, M. de Lima, S. Giralt, B. Andersson, I. Khouri, V. J. Erpenbeck, B. Haddad, H. A. Lehr, E. Schmitt, T. Bopp, et al. 2005. The IL- C. Hosing, C. Ippoliti, E. J. Shpall, R. Champlin, and A. Alousi. 2009. A phase III 6R a chain controls lung CD4+CD25+ Treg development and function during study of infliximab and corticosteroids for the initial treatment of acute graft- allergic airway inflammation in vivo. J. Clin. Invest. 115: 313–325. versus-host disease. Biol. Blood Marrow Transplant. 15: 1555–1562. 78. Revez, J. A., L. Bain, B. Chapman, J. E. Powell, R. Jansen, D. L. Duffy, 102. Levine, J. E., S. Paczesny, S. Mineishi, T. Braun, S. W. Choi, R. J. Hutchinson, J. Y. Tung, AAGC Collaborators, P. M. Penninx, P. M. Visscher, E. J. De Geus, D. Jones, Y. Khaled, C. L. Kitko, D. Bickley, et al. 2008. Etanercept plus methyl- et al. 2013. A new regulatory variant in the interleukin-6 receptor gene associates as initial therapy for acute graft-versus-host disease. Blood 111: 2470–2475. with asthma risk. Genes Immun. 14: 441–446. 103. Alousi, A. M., D. J. Weisdorf, B. R. Logan, J. Bolan˜os-Meade, S. Carter, 79. DiCarlo, J., R. Agarwal-Hashmi, A. Shah, P. Kim, L. Craveiro, R. Killen, N. Difronzo, M. Pasquini, S. C. Goldstein, V. T. Ho, B. Hayes-Lattin, et al. 2009. Downloaded from Y. Rosenberg-Hasson, and H. Maecker. 2014. Cytokine and chemokine patterns Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for across 100 days after hematopoietic stem cell transplantation in children. Biol. acute graft-versus-host disease: a randomized phase 2 trial from the Blood and Blood Marrow Transplant. 20: 361–369. Marrow Transplant Clinical Trials Network. Blood 114: 511–517. 80. Derniame, S., J. Perazzo, F. Lee, A. Domogala, M. Escobedo-Cousin, 104. Busca, A., F. Locatelli, F. Marmont, C. Ceretto, and M. Falda. 2007. Recombi- R. Alnabhan, M. Luevano, I. Pedroza-Pacheco, N. Cooper, A. Madrigal, and nant human soluble tumor necrosis factor receptor fusion as treatment for A. Saudemont. 2014. Differential effects of mycophenolate mofetil and cyclo- steroid refractory graft-versus-host disease following allogeneic hematopoietic stem sporine A on peripheral blood and cord blood natural killer cells activated with cell transplantation. Am. J. Hematol. 82: 45–52.

interleukin-2. Cytotherapy 16: 1409–1418. 105. Wolff, D., V. Roessler, B. Steiner, S. Wilhelm, V. Weirich, J. Brenmoehl, http://www.jimmunol.org/ 81. Derniame, S., F. Lee, A. Domogala, A. Madrigal, and A. Saudemont. 2014. M. Leithaeuser, N. Hofmeister, C. Junghanss, J. Casper, et al. 2005. Treatment of Unique effects of mycophenolate mofetil on cord blood T cells: implications for steroid-resistant acute graft-versus-host disease with and etanercept. GVHD prophylaxis. Transplantation 97: 870–878. Bone Marrow Transplant. 35: 1003–1010. 82. Antin, J. H., D. Weisdorf, D. Neuberg, R. Nicklow, S. Clouthier, S. J. Lee, 106. Antin, J. H., H. J. Weinstein, E. C. Guinan, P. McCarthy, B. E. Bierer, E. Alyea, C. McGarigle, B. R. Blazar, S. Sonis, et al. 2002. Interleukin-1 blockade D. G. Gilliland, S. K. Parsons, K. K. Ballen, I. J. Rimm, G. Falzarano, et al. 1994. does not prevent acute graft-versus-host disease: results of a randomized, double- Recombinant human interleukin-1 receptor antagonist in the treatment of steroid- blind, -controlled trial of interleukin-1 receptor antagonist in allogeneic resistant graft-versus-host disease. Blood 84: 1342–1348. bone marrow transplantation. Blood 100: 3479–3482. 107. Fang, J., C. Hu, M. Hong, Q. Wu, Y. You, Z. Zhong, W. Li, P. Zou, Y. Hu, and 83. Choi, S. W., P. Stiff, K. Cooke, J. L. Ferrara, T. Braun, C. Kitko, P. Reddy, L. Xia. 2012. Prophylactic effects of interleukin-2 receptor antagonists against G. Yanik, S. Mineishi, S. Paczesny, et al. 2012. TNF-inhibition with etanercept for graft-versus-host disease following unrelated donor peripheral blood stem cell graft-versus-host disease prevention in high-risk HCT: lower TNFR1 levels cor- transplantation. Biol. Blood Marrow Transplant. 18: 754–762. € relate with better outcomes. Biol. Blood Marrow Transplant. 18: 1525–1532. 108. Schmidt-Hieber, M., T. Fietz, W. Knauf, L. Uharek, W. Hopfenmuller, E. Thiel, by guest on September 25, 2021 84. Hamadani, M., C. C. Hofmeister, B. Jansak, G. Phillips, P. Elder, W. Blum, and I. W. Blau. 2005. Efficacy of the interleukin-2 receptor antagonist basiliximab S. Penza, T. S. Lin, R. Klisovic, G. Marcucci, et al. 2008. Addition of infliximab to in steroid-refractory acute graft-versus-host disease. Br. J. Haematol. 130: 568–574. standard acute graft-versus-host disease prophylaxis following allogeneic peripheral 109. Przepiorka, D., N. A. Kernan, C. Ippoliti, E. B. Papadopoulos, S. Giralt, blood cell transplantation. Biol. Blood Marrow Transplant. 14: 783–789. I. Khouri, J. G. Lu, J. Gajewski, A. Durett, K. Cleary, et al. 2000. Daclizumab, 85. Karimi, M. H., S. Salek, R. Yaghobi, M. Ramzi, B. Geramizadeh, and S. Hejr. a humanized anti-interleukin-2 receptor alpha chain antibody, for treatment of 2014. Association of IL-17 gene polymorphisms and serum level with graft versus acute graft-versus-host disease. Blood 95: 83–89. host disease after allogeneic hematopoietic stem cell transplantation. Cytokine 69: 110. Willenbacher, W., N. Basara, I. W. Blau, A. A. Fauser, and M. G. Kiehl. 2001. 120–124. Treatment of steroid refractory acute and chronic graft-versus-host disease with 86. Resende, R. G., J. d. F. Correia-Silva, T. A. Silva, U. E. Saloma˜o, L. Marques- daclizumab. Br. J. Haematol. 112: 820–823. Silva, E´. L. M. Vieira, W. O. Dutra, and R. S. Gomez. 2014. IL-17 genetic and 111. Cahn, J. Y., P. Bordigoni, P. Tiberghien, N. Milpied, A. Brion, J. Widjenes, immunophenotypic evaluation in chronic graft-versus-host disease. Mediators B. Lioure, G. Michel, S. Burdach, H. J. Kolb, et al. 1995. Treatment of acute Inflamm. DOI: http://dx.doi.org/10.1155/2014/571231 graft-versus-host disease with methylprednisolone and cyclosporine with or with- 87. Buch, M. H., and P. Emery. 2011. New therapies in the management of rheu- out an anti-interleukin-2 receptor . A multicenter phase III matoid arthritis. Curr. Opin. Rheumatol. 23: 245–251. study. Transplantation 60: 939–942. 88. Monaco, C., J. Nanchahal, P. Taylor, and M. Feldmann. 2015. Anti-TNF ther- 112. Lee, S. J., D. Zahrieh, E. Agura, M. L. MacMillan, R. T. Maziarz, P. L. McCarthy, apy: past, present and future. Int. Immunol. 27: 55–62. Jr., V. T. Ho, C. Cutler, E. P. Alyea, J. H. Antin, and R. J. Soiffer. 2004. Effect of 89. Singh, J. A., S. Beg, and M. A. Lopez-Olivo. 2011. Tocilizumab for rheumatoid up-front daclizumab when combined with steroids for the treatment of acute graft- arthritis: a Cochrane systematic review. J. Rheumatol. 38: 10–20. versus-host disease: results of a randomized trial. Blood 104: 1559–1564. 90. Gabay, C., P. Emery, R. van Vollenhoven, A. Dikranian, R. Alten, K. Pavelka, 113. Gergis, U., J. Arnason, R. Yantiss, T. Shore, U. Wissa, E. Feldman, and M. Klearman, D. Musselman, S. Agarwal, J. Green, and A. Kavanaugh, ADACTA T. Woodworth. 2010. Effectiveness and safety of tocilizumab, an anti-interleukin- Study Investigators. 2013. Tocilizumab monotherapy versus mono- 6 receptor monoclonal antibody, in a patient with refractory GI graft-versus-host therapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double- disease. J. Clin. Oncol. 28: e602–e604. blind, controlled phase 4 trial. Lancet 381: 1541–1550. 114. Drobyski, W. R., M. Pasquini, K. Kovatovic, J. Palmer, J. Douglas Rizzo, A. Saad, 91. Fischer, J. A., A. J. Hueber, S. Wilson, M. Galm, W. Baum, C. Kitson, J. Auer, W. Saber, and P. Hari. 2011. Tocilizumab for the treatment of steroid refractory S. H. Lorenz, J. Moelleken, M. Bader, et al. 2015. Combined inhibition of tumor graft-versus-host disease. Biol. Blood Marrow Transplant. 17: 1862–1868. necrosis factor a and interleukin-17 as a therapeutic opportunity in rheumatoid 115. Blazar, B. R., D. J. Weisdorf, T. Defor, A. Goldman, T. Braun, S. Silver, and arthritis: development and characterization of a novel bispecific antibody. Arthritis J. L. Ferrara. 2006. Phase 1/2 randomized, placebo-control trial of palifermin to Rheumatol. 67: 51–62. prevent graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell 92. Venkatesha, S. H., S. Dudics, B. Acharya, and K. D. Moudgil. 2015. Cytokine- transplantation (HSCT). Blood 108: 3216–3222. modulating strategies and newer cytokine targets for arthritis therapy. Int. J. Mol. 116. Nasilowska-Adamska, B., P. Rzepecki, J. Manko, A. Czyz, M. Markiewicz, Sci. 16: 887–906. I. Federowicz, A. Tomaszewska, B. Piatkowska-Jakubas, A. Wrzesien-Kus, 93. Mandell, B. F., and J. M. Sobell. 2014. The role of TNF inhibitors in psoriatic M. Bieniaszewska, et al. 2007. The influence of palifermin (Kepivance) on oral disease. Semin. Cutan. Med. Surg. 33(4, Suppl.)S64–S68. mucositis and acute graft versus host disease in patients with hematological diseases 94. Mitra, A., S. K. Raychaudhuri, and S. P. Raychaudhuri. 2012. Functional role of undergoing hematopoietic stem cell transplant. Bone Marrow Transplant. 40: 983– IL-22 in psoriatic arthritis. Arthritis Res. Ther. 14: R65. 988. 95.Tonel,G.,C.Conrad,U.Laggner,P.DiMeglio,K.Grys, 117. Langner, S., P. Staber, N. Schub, M. Gramatzki, W. Grothe, G. Behre, T.K.McClanahan,W.M.Blumenschein,J.-Z.Qin,H.Xin,E.Oldham, W. Rabitsch, C. Urban, W. Linkesch, and P. Neumeister. 2008. Palifermin et al. 2010. Cutting edge: A critical functional role for IL-23 in . J. reduces incidence and severity of oral mucositis in allogeneic stem-cell transplant Immunol. 185: 5688–5691. recipients. Bone Marrow Transplant. 42: 275–279. 96. Colombel, J. F., W. J. Sandborn, W. Reinisch, G. J. Mantzaris, A. Kornbluth, 118. Levine, J. E., B. R. Blazar, T. DeFor, J. L. Ferrara, and D. J. Weisdorf. 2008. D. Rachmilewitz, S. Lichtiger, G. D’Haens, R. H. Diamond, D. L. Broussard, Long-term follow-up of a phase I/II randomized, placebo-controlled trial of pal- 4612 BRIEF REVIEWS: CYTOKINES IN GVHD

ifermin to prevent graft-versus-host disease (GVHD) after related donor allogeneic lowing peripheral blood stem cell transplant for chronic myeloid leukaemia: hematopoietic cell transplantation (HCT). Biol. Blood Marrow Transplant. 14: a clinico-pathological study. Bone Marrow Transplant. 20: 83–85. 1017–1021. 128. Klingemann, H. G., A. P. Grigg, K. Wilkie-Boyd, M. J. Barnett, A. C. Eaves, 119. Nasilowska-Adamska, B., R. Szydlo, P. Rzepecki, A. Czyz, A. Tomaszewska, D. E. Reece, J. D. Shepherd, and G. L. Phillips. 1991. Treatment with M. Markiewicz, T. Torosian, M. Bieniaszewska, A. Hellman, W. W. Jedrzejczak, recombinant interferon (alpha-2b) early after bone marrow transplantation in et al. 2011. Palifermin does not influence the incidence and severity of GvHD nor patients at high risk for relapse [corrected]. Blood 78: 3306–3311. long-term survival of patients with hematological diseases undergoing HSCT. Ann. 129. Gordon, K. B., C. L. Leonardi, M. Lebwohl, A. Blauvelt, G. S. Cameron, Transplant. 16: 47–54. D. Braun, J. Erickson, and M. Heffernan. 2014. A 52-week, open-label study of 120. Jagasia, M. H., R. Abonour, G. D. Long, B. J. Bolwell, G. G. Laport, T. B. Shore, the efficacy and safety of ixekizumab, an anti-interleukin-17A monoclonal anti- S. Durrant, J. Szer, M.-G. Chen, R. Lizambri, and E. K. Waller. 2012. Palifermin body, in patients with chronic plaque psoriasis. J. Am. Acad. Dermatol. 71: 1176–1182. for the reduction of acute GVHD: a randomized, double-blind, placebo-controlled 130. Papp, K., C. Leonardi, A. Menter, E. H. Z. Thompson, C. E. Milmont, trial. Bone Marrow Transplant. 47: 1350–1355. G. Kricorian, A. Nirula, and P. Klekotka. 2014. Safety and efficacy of brodalumab 121. Antin, J. H., S. J. Lee, D. Neuberg, E. Alyea, R. J. Soiffer, S. Sonis, and for psoriasis after 120 weeks of treatment. J. Am. Acad. Dermatol. 71: 1183–1190.e3. J. L. Ferrara. 2002. A phase I/II double-blind, placebo-controlled study of 131. Langley, R. G., B. E. Elewski, M. Lebwohl, K. Reich, C. E. Griffiths, K. Papp, recombinant human interleukin-11 for mucositis and acute GVHD prevention in L. Puig, H. Nakagawa, L. Spelman, B. Sigurgeirsson, et al. 2014. Secukinumab in allogeneic stem cell transplantation. Bone Marrow Transplant. 29: 373–377. plaque psoriasis—results of two phase 3 trials. N. Engl. J. Med. 371: 326–338. 122. Porter, D. L., M. S. Roth, C. McGarigle, J. L. Ferrara, and J. H. Antin. 1994. 132. Krueger, G. G., R. G. Langley, C. Leonardi, N. Yeilding, C. Guzzo, Y. Wang, Induction of graft-versus-host disease as immunotherapy for relapsed chronic L. T. Dooley, and M. Lebwohl, CNTO 1275 Psoriasis Study Group. 2007. A myeloid leukemia. N. Engl. J. Med. 330: 100–106. human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N. 123. Samson, D., L. Volin, U. Schanz, A. Bosi, and G. Gahrtron. 1996. Feasibility and Engl. J. Med. 356: 580–592. toxicity of interferon maintenance therapy after allogeneic BMT for multiple 133. Sofen, H., S. Smith, R. T. Matheson, C. L. Leonardi, C. Calderon, myeloma: a pilot study of the EBMT. Bone Marrow Transplant. 17: 759–762. C. Brodmerkel, K. Li, K. Campbell, S. J. Marciniak, Jr., Y. Wasfi, et al. 2014. 124. Ratanatharathorn, V., J. Uberti, C. Karanes, L. G. Lum, E. Abella, M. E. Dan, Guselkumab (an IL-23-specific mAb) demonstrates clinical and molecular response M. Hussein, and L. L. Sensenbrenner. 1994. Phase I study of alpha-interferon in patients with moderate-to-severe psoriasis. J. Allergy Clin. Immunol. 133: 1032– augmentation of cyclosporine-induced graft versus host disease in recipients of 1040. autologous bone marrow transplantation. Bone Marrow Transplant. 13: 625–630. 134. Zandvliet, A., S. Glasgow, A. Horowitz, D. Montgomery, J. Marjason, A. Mehta, Downloaded from 125. Streetly, M., M. Kazmi, D. Radia, C. Hoyle, and S. A. Schey. 2004. Second C. Xu, M. van Vugt, and S. Khalilieh. 2015. Tildrakizumab, a novel anti-IL-23 autologous transplant with cyclosporin/interferon alpha-induced graft versus host monoclonal antibody, is unaffected by ethnic variability in Caucasian, Chinese, disease for patients who have failed first-line consolidation. Bone Marrow Trans- and Japanese subjects. Int. J. Clin. Pharmacol. Ther. 53: 139–146. plant. 33: 1131–1135. 135. Piper, E., C. Brightling, R. Niven, C. Oh, R. Faggioni, K. Poon, D. She, 126. Kolb, H. J., J. Mittermuller,€ C. Clemm, E. Holler, G. Ledderose, G. Brehm, C.Kell,R.D.May,G.P.Geba,andN.A.Molfino.2013.AphaseIIplacebo- M. Heim, and W. Wilmanns. 1990. Donor leukocyte transfusions for treatment of controlled study of tralokinumab in moderate-to-severe asthma. Eur. Respir. J. recurrent chronic myelogenous leukemia in marrow transplant patients. Blood 76: 41: 330–338.

2462–2465. 136. Corren, J., R. F. Lemanske, N. A. Hanania, P. E. Korenblat, M. V. Parsey, http://www.jimmunol.org/ 127. Lim, S. H., S. Coleman, A. Bull, U. O’Callaghan, R. Evely, and M. Booth. 1997. J. R. Arron, J. M. Harris, H. Scheerens, L. C. Wu, Z. Su, et al. 2011. Leb- Cyclosporin A/alpha interferon-induced autologous graft-versus-host disease fol- rikizumab treatment in adults with asthma. N. Engl. J. Med. 365: 1088–1098. by guest on September 25, 2021