throbber
Brief Reviews
`
`Th e
`
`Journal of
`Immunology
`
`Cytokines in Graft-versus-Host Disease
`
`Andrea S. Henden and Geoffrey R. Hill
`
`Graft-versus-host disease (GVHD) is a complication of
`allogeneic bone marrow transplantation whereby trans-
`planted naive and marrow-derived T cells damage recip-
`ient tissue through similar mechanisms to those that allow
`destruction of malignant cells, the therapeutic intent of
`bone marrow transplantation. The manifestations and
`severity of GVHD are highly variable and are influenced
`by the proportions of naive cells maturing along regula-
`tory T cell, Th1, Th2, or Th17 phenotypes. This matu-
`ration is largely influenced by local cytokines, which, in
`turn, activate transcription factors and drive development
`toward a dominant phenotype. In addition, proinflamma-
`tory cytokines exert direct effects on GVHD target tissues.
`Our knowledge of the role that cytokines play in orches-
`trating GVHD is expanding rapidly and parallels other
`infective and inflammatory conditions in which a predom-
`inant T cell signature is causative of pathology. Because
`a broad spectrum of cytokine therapies is now routinely
`used in clinical practice, they are increasingly relevant to
`transplant medicine. The Journal of Immunology, 2015,
`194: 4604–4612.
`
`G raft-versus-host disease (GVHD) is a phenomenon
`
`almost unique to allogeneic bone marrow trans-
`plantation (BMT) whereby lymphocytes are intro-
`duced and permitted to engraft and proliferate within an
`immunocompromised host. In this setting, naive (i.e., those that
`have not previously encountered Ag) donor T cells are able to
`recognize host or recipient Ags as foreign, an effect that constitutes
`the therapeutic intent of BMT, allowing destruction of leukemic
`or other malignant cells through activation of pathways of the
`adaptive immune response. This beneficial effect is termed “graft-
`versus-leukemia” (GVL). The relative contributions of memory
`T cells to GVHD and GVL were discussed elsewhere (1).
`However, the effect is not specific to malignant cells, and si-
`multaneous damage and destruction of healthy cells and tissues
`via the same or similar mechanisms give rise to GVHD. The
`morbidity and mortality of GVHD limit the clinical scenarios in
`which allogeneic hematopoietic stem cell transplantation may
`otherwise offer therapeutic benefit. Therefore, much research has
`focused on the separation of GVL and GVHD, although success
`has been limited because of the use of the same immune effector
`
`mechanisms. An example of this is T cell depletion of trans-
`plants: a reduction in GVHD is offset by attendant increases in
`the rates of relapse of primary malignancy (2, 3), in addition to
`more delayed immune reconstitution with increased morbidity
`and mortality due to opportunistic infection. An alternate focus
`has been to examine the influences on emerging innate and
`adaptive immune responses in an attempt to preserve beneficial
`GVL effects while eliminating the harmful “off-target” GVHD
`effects. In this setting, understanding the cytokine orchestration
`of the maturing immune response within allogeneic transplan-
`tation offers the opportunity to improve outcomes of this
`treatment through identification of rapidly translatable clinical
`therapeutic targets. Our understanding of events within the al-
`logeneic transplantation landscape also informs our under-
`standing of emerging innate and adaptive immune responses in
`scenarios other than BMT.
`
`Cytokines and acute GVHD
`
`The initiation of GVHD is necessarily influenced by the cy-
`tokine milieu in which it arises, and three distinct phases have
`been described (4, 5). The initial phase is triggered by tissue
`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
`a minimal residual level suitable for subsequent immune control
`and to ablate existing immune function, allowing engraftment of
`the naive donor inoculum. Myeloablative stem cell transplan-
`tation typically uses total body irradiation or busulphan-based
`chemotherapy to achieve these dual aims; however, they also
`result in damage to the GI tract mucosa and other cells con-
`tributing to the “cytokine storm,” which is characterized by the
`release of proinflammatory cytokines: classically TNF, IL-1, and
`IL-6 (4, 6). Although less well defined, there is an appreciation
`that a similar process occurs with reduced intensity–condition-
`ing transplantation, although the dominant cytokines and
`temporal relationships may differ (7).
`In addition to chemotherapy and radiation-induced tissue
`damage and inflammation, recognition of pathogen-associated
`molecular patterns, such as LPS, and danger-associated mo-
`lecular patterns arising from GI microbiota have significant
`bearing on GVHD pathophysiology. The inflammatory sig-
`nals generated in the emerging adaptive immune response are
`added to by recognition of molecular motifs from both
`pathogenic and commensal organisms and subsequent acti-
`
`Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute,
`Brisbane 4006, Queensland, Australia; and The Royal Brisbane and Women’s Hospital,
`Brisbane 4029, Queensland, Australia
`
`Abbreviations used in this article: aGVHD, acute GVHD; BMT, bone marrow trans-
`plantation; cGVHD, chronic GVHD; GI, gastrointestinal; GVHD, graft-versus-host
`disease; GVL, graft-versus-leukemia; TFH, T follicular helper; Treg, regulatory T cell.
`
`Received for publication January 20, 2015. Accepted for publication March 19, 2015.
`
`Address correspondence and reprint requests to Prof. Geoffrey R. Hill, QIMR Berghofer
`Medical Research Institute, 300 Herston Road, Herston, Brisbane 4006, QLD,
`Australia. E-mail address: geoff.hill@qimrberghofer.edu.au
`
`www.jimmunol.org/cgi/doi/10.4049/jimmunol.1500117
`
`Copyright Ó 2015 by The American Association of Immunologists, Inc. 0022-1767/15/$25.00
`
`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`

`

`The Journal of Immunology
`
`4605
`
`vation of innate lymphoid pathways. The diversity of the
`resident organisms can be affected by conditioning-associated
`inflammation and by GVHD itself; conversely, the microbiota
`present can influence the severity of GVHD (8). The quan-
`titative and qualitative contributions of this microbiota-driven
`inflammatory signal are influenced by the variety and path-
`ogenicity of organisms present, and has been demonstrated to
`affect the severity of GVHD (8–10). A reduction in the
`bacterial burden by use of antimicrobial decontamination in
`the posttransplant period also can reduce GVHD severity
`(10). Although our mechanistic understanding of this effect is
`not complete, it is apparent that GVHD mediates a loss of
`Paneth cell–derived antimicrobial peptides that play an im-
`portant role in shaping the diversity of microbiota (11), in
`addition to the use of pharmaceutical antimicrobials (9).
`Understanding these mechanisms may offer manipulable
`targets to alter this primary, inflammation-mediated, initia-
`tion phase of GVHD.
`Recognition of the range of triggers to the cytokine storm
`complements our knowledge of subsequent T cell and APC
`interactions that define the second phase of acute GVHD
`(aGVHD) pathophysiology and during which cytokines play
`a key role in driving naive T cell differentiation and expansion
`toward one maturation program or another. Type 1 or Tc1/
`Th1 maturation is recognized as the dominant pattern in
`aGVHD (12, 13), and is linked to severe GI tract pathology
`(14). Indeed, in animal models of aGVHD, Th2 and regu-
`latory T cells (Tregs) are rare (15). T cells expressing IL-17 are
`also rare, although this may reflect the plasticity of this lineage
`(16). Increased quantities of Th1-associated cytokines, TNF
`and IFN-g, in aGVHD are associated with earlier onset and
`more severe disease in preclinical models and clinical BMT (4,
`14, 17–19). Although the dominance of Th1 subsets is well
`established, Th2 and Th17 subsets are also involved in pa-
`thology, and the balance between subsets determines aGVHD
`severity (20), in addition to organ specificity (15, 20), and the
`pathogenic or protective effects of any subset cannot be
`viewed in isolation. Implicit in the known reciprocal regula-
`tion of T cell differentiation by these cytokines is the concept
`that inhibition of any one lineage may provoke unwanted and
`exaggerated differentiation down alternative differentiation
`pathways.
`Th2 differentiation is often seen as opposing Th1 differ-
`entiation; however, this subset is also recognized to cause
`aGVHD but with predominant pathology in pulmonary,
`hepatic, and cutaneous tissues (21), in contrast to the strong
`GI association with Th1. Cutaneous pathology also may be
`generated by Th17 cells; although they are more commonly
`associated with chronic GVHD (cGVHD), they also have
`been associated with acute pathology (22–24). Th17 differ-
`entiation is initiated by IL-6 (25), and RORgt is the defining
`transcription factor (26), whereas maintenance and amplifi-
`cation relies on IL-23 and IL-21, respectively (27). The use of
`RORC-deficient donor T cells results in attenuated aGVHD
`severity and lethality (26). Further studies are needed to better
`define the role of this subset in late aGVHD versus early
`cGVHD, as well as the relative contribution of IL-17 from
`CD4 and CD8 T cells to end-organ pathology.
`The third and final effector phase of aGVHD is charac-
`terized by target tissue damage, with the hallmark histological
`finding being apoptosis, most commonly in the GI tract, liver,
`
`and skin. This tissue damage is mediated by more than one
`immunological mechanism. First, cognate T cell–MHC
`interactions are required for effector, usually CD8, T cells that
`are able to evoke cytolytic machinery, including perforins and
`granzymes that induce target cell death via apoptosis (5, 6,
`28). Interestingly, granzyme B–deficient donor T cells me-
`diate less severe GVHD but may still generate GVL (29), via
`reduced activation-induced death of CD8 T cells (30). In
`a complementary pathway in which cognate T cell–MHC
`interactions are not required, myeloid cells, in addition to
`lymphoid cells, are primed during aGVHD to release cyto-
`pathic quantities of inflammatory cytokines (e.g., TNF, IL-6)
`that directly invoke apoptosis (31). Importantly, TNF is also
`involved in GVL effects, and inhibition can compromise
`antitumor immunity (32). Damage to the primary target
`organs of GVHD is driven by chemokine expression that
`results in tissue homing of lymphocyte populations. LPAM-1
`(a4b7 integrin) and L-selectin (CD62L) are associated with
`homing to GI and GALTs and cutaneous lymphoid Ag to
`skin (33) and are necessary for induction of GVHD tissue
`damage at these sites (34). Cytokines, including IFN-g, are
`known to induce upregulation of chemokines and recep-
`tors (35), and these mechanisms were shown to be important
`in determining the severity of GVHD within an inflam-
`matory environment (36–38). The expression of mole-
`cules associated with lymphocyte exhaustion (e.g., PD1) and
`their ligands (e.g., PD-L1) on nonlymphoid tissue also was
`shown to be cytokine (IFN) dependent and contribute to the
`constraint of
`lymphocyte-mediated tissue damage late in
`the aGVHD setting (39, 40). Therefore, the inflamma-
`tory cytokines present in this setting participate in positive-
`and negative-feedback loops in both lymphocyte and non-
`lymphocyte populations.
`
`Cytokines and cGVHD
`
`cGVHD represents a distinct pathophysiological entity from
`aGVHD that traditionally is separated by time of onset; how-
`ever, it is now recognized by its distinct end-organ pathology.
`Although the cardinal feature of aGVHD is apoptosis, fibrosis
`is the predominant mechanism of tissue damage in cGVHD.
`Additionally, primary target organs also differ, with lung and
`skin being the primary target organs in cGVHD, manifesting
`as bronchiolitis obliterans and scleroderma (41, 42). Sicca
`symptoms secondary to salivary and lacrimal gland destruc-
`tion and oral lichenoid GVHD are also prominent. Despite
`these disparate pathophysiological manifestations, clear roles
`for cytokine control of this phase of disease also were dem-
`onstrated, unaccompanied by large-scale conditioning-related
`tissue damage and the “cytokine storm” that initiates aGVHD.
`IL-17 and subsequent T cell differentiation along the Th17
`pathway are becoming more strongly associated with cGVHD.
`Initially identified with the use of G-CSF in stem cell mo-
`bilization of donors and prominent Th17 differentiation (43),
`IL-17 was shown more recently to result in CSF1-dependent
`macrophage accumulation in skin and lung, which drives tis-
`sue fibrosis (44). We demonstrated recently that, consistent
`with this, systemic IL-17 levels increase late after clinical
`BMT, at a time when cGVHD develops (45). It is also clear
`that T follicular helper (TFH) cells and IL-21 play important
`roles in the development of cGVHD via the stimulation of
`germinal center B cells and alloantibody generation (46).
`
`

`

`4606
`
`BRIEF REVIEWS: CYTOKINES IN GVHD
`
`This is particularly relevant to bronchiolitis obliterans, be-
`cause preliminary evidence suggests that Th17 differentia-
`tion and CSF1 dysregulation are also involved in this
`aberrant immunological pathway (44). Thus, inhibition of
`Th17 differentiation and CSF1 appear highly relevant to
`the prevention and treatment of cGVHD. Inhibition of ter-
`minal cytokines involved in fibrosis, such as TGF-b and
`IL-13, represent additional targets; however, TGF-b inhi-
`bition is problematic given its important role in Treg ho-
`meostasis (47).
`
`Cytokines and T cell–differentiation programs
`
`The ability of cytokines to drive cellular differentiation is
`recognized in situations other than GVHD, with the accep-
`tance that derivation of phenotypically distinct erythroid,
`myeloid, and lymphoid populations from a common long-
`term hematopoietic stem cell is dependent upon binding of
`cytokines to their cognate receptors (48, 49). Similarly, the
`maturation of the naive T cell population in the context of
`BMT and GVHD is also driven by cytokines and subsequent
`transcriptional pathways elicited thereafter (50). A summary
`of these effects is shown in Fig. 1 and outlined further below.
`Cytokines and Th1 differentiation. IFN-g, IL-2, and TNF are
`the key cytokines generated during Th1 differentiation (14),
`and phenotypic differentiation is initiated by IL-12 and
`controlled by the transcription factor T-bet (25, 48). IFN-g
`in this setting participates in positive feedback to reinforce Th1
`responses, in addition to exerting effects on nonlymphocytes,
`as well as on nonhematopoietic cells (18). Initial attempts
`to define the role of IFN-g in determining aGVHD severity
`were hampered by conflicting data supporting the exacerba-
`tion and amelioration of pathology; however, subsequent
`work demonstrated this to be related to differing effects on
`donor and host tissues, in addition to tissue-specific effects
`
`on nonhematopoietic tissues. Donor lymphocyte IFN-g
`signaling enhanced GVHD via the promotion of Th1 dif-
`ferentiation, and it also is directly cytotoxic to gut mucosa
`(18). Tissue-specific effects are also seen in pulmonary pa-
`renchyma in which a protective role for IFN-g was dem-
`onstrated and these effects have also been described by
`other groups (15). IFN-g provides evidence for a paradigm
`where cytokines may exert effects in nonhematopoietic tissue,
`in addition to specific effects on lymphocytes and other he-
`matopoietic cells. Evidence of similar patterns for other cy-
`tokines is continually being defined and allows selection of
`appropriate targets for inhibition in the clinic.
`With regard to other Th1-associated cytokines, a similarly
`complex effect is seen for IL-2, both mechanistically and in
`therapeutic outcomes (51). Initially used at a high dose in an
`attempt to augment proliferation of lymphocytes as “immu-
`notherapy” for solid malignancies (52, 53),
`it was subse-
`quently found, paradoxically, to have a critical role in sup-
`porting Treg populations and in controlling GVHD (51,
`54). The promotion of regulatory pathways in GVHD was
`demonstrated in small numbers of patients when used in
`a “low dose” (55). These apparently dose-dependent effects
`are likely explained by competition for consumption of this
`cytokine by maturing Treg and T effector cell populations
`(56); in the clinical transplantation scenario, they are further
`complicated by the use of calcineurin inhibitors, which also
`target this pathway, when used as GVHD prophylaxis post-
`transplant.
`Cytokines and Th2 differentiation. The presence of IL-25 and,
`subsequently, IL-4 supports the development of T cells of
`the Th2 lineage that traditionally have been described as
`being involved in allergy and host defense against parasites
`and helminths. Th2 cells produce IL-4, IL-5, IL-10, and
`IL-13, with transcriptional control exerted by GATA3 (57).
`
`.. Tissue
`
`damage
`
`Acute
`GVHD
`
`Chronic
`GVHD
`
`Cytokine driven
`cellular differentiation
`
`Naive
`cells
`
`chemotherapy -,,
`conditioning
`damage
`
`+ Radiation and _.,.
`(bO
`
`IL-4 -IL-12 -IL-1
`
`IL-6 -
`
`IFNy
`TNF
`
`Tcells
`
`Oo O
`
`NK/NKTI
`Innate
`Lymphoid
`Cells(ILC)
`
`Monocytes
`and
`macrophages
`
`Microbiota
`derived DAMPs
`and PAMPs
`
`Tc1/Th1; IFNy, TNF, IL-2
`
`NK/NKT/ILC activation
`
`-
`
`FIBROSIS
`
`APOPTOSIS
`
`y
`
`granzyme
`mediated (cid:173)
`cytolysis
`
`TNF,
`IL-1
`and IL-6
`
`Activated monocytes
`and macrophages
`
`......_ TGF-~
`
`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 gut microbiota 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 monocyte 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
`lung pathology (15, 18), as opposed to the strong association
`of Th1 cells with gut and liver damage. Recent work dem-
`onstrated a role for an IL-25–dependent immature non-B
`non-T cell innate immune effector cell population that is re-
`sponsible for propagation of Th2 responses and production
`of IL-4, IL-5, and IL-13; where lacking, this results in an
`impaired ability to expel helminths (58–60). This cytokine
`may be seen as having protective effects on GI tissues, an
`outcome that is clearly attractive in the context of aGVHD
`pathology.
`Other Th2 cytokines were shown to have protective roles in
`GVHD, including IL-10. Despite mixed results when initially
`given as therapy to patients, its production by B lymphocytes
`in animal models of transplantation reduces the severity of
`GVHD (61). These outcomes were mediated by effects on
`donor T cell expansion; similarly, reductions in IL-4 and IL-
`10 were demonstrated in patients with cGVHD (62, 63).
`Cytokines and Th17 differentiation. Th17 cells are a more recent
`addition to the Th1/Th2 paradigm (25), and roles for IL-17–
`producing cells of both Th17 and Tc17 varieties are still being
`defined in the GVHD setting and in other immune pathologies
`(64). Initiation of Th17 development is triggered by IL-6 and
`TGF-b and is associated with transcriptional activation of
`RORgt after phosphorylation of STAT3, as well as with the
`secretion of IL-17, IL-21, and IL-22. There is an increasing
`appreciation for the role that Th17 cells play in determining
`the severity of GVHD (65), with a particular role for IL-6
`becoming apparent (66, 67). Recently, our group demonstrated
`the importance of this effect in cohorts in which IL-6 inhibition
`represents a potentially effective therapeutic strategy to reduce the
`severity of aGVHD in clinical stem cell transplantation (45). IL-
`22 may be secreted by Th17 cells and, in this setting, it appears to
`be pathogenic (68); conversely, it may be secreted by innate
`lymphoid cells where, in the GI tract at least, it appears to be
`an important protective cytokine (69). IL-21 can be produced by
`both Th17 and TFH cells, and it promotes aGVHD by impairing
`Treg homeostasis (70). Given that it also has an important role in
`inducing aberrant, allospecific germinal center B cell responses
`and cGVHD, inhibition of this Th17-associated cytokine rep-
`resents another attractive therapeutic target for GVHD control
`after BMT.
`
`Heterogeneity of effect conferred by cellular target
`
`Increasingly, the effects mediated by a particular cytokine are
`being defined as dependent on the cells in which it transduces
`a signal and may be considered to regulate effector cell pop-
`ulations, as well as to confer susceptibility or protection to other
`inflammatory signals in target organs and tissues. Modes of
`signaling also influence these responses, with a recent appreci-
`ation for the differential pathology induced by the binding of
`cytokines by membrane-bound receptors as opposed to soluble
`receptors.
`Hematopoietic and nonhematopoietic cells. Cytokines may exert
`effects on cells of hematopoietic origin, in addition to non-
`hematopoietic target tissues directly. IFN-g (18) and IL-22
`(68, 69) are clear examples that were already discussed.
`Further examples are seen in cGVHD: IL-2, IL-10, and
`TGF-b may act directly on tissue fibroblasts in affected
`organs to mediate pathology (71),
`in addition to known
`effects of IL-2 in supporting Treg populations (51, 55) and
`
`B cell–derived IL-10 being protective in the initiation of
`aGVHD (61). A role for innate lymphoid cells as cytokine-
`responsive mediators of protection from GVHD is emerging
`(72), with ILC1, ILC2, and ILC3 subtypes demonstrating
`similar transcriptional control and cytokine profiles to Th1,
`Th2, and Th17 cells (73).
`Donor and host cells. The effect of a single cytokine can
`be dependent upon its roles in hematopoietic and non-
`hematopoietic tissue; however, in BMT, donor or host origin of
`the cell transducing the signal is as an additional factor influ-
`encing outcomes. Type I IFN is an example of a cytokine for
`which signaling through recipient APCs results in less severe class
`II–dependent GVHD in the colon, whereas signaling through
`donor APCs may amplify GVHD responses. The former is
`mediated through decreased donor CD4 proliferation, and the
`latter is mediated through more effective cross-presentation of
`alloantigens to CD8 T cells (19). IL-4 is another example. A
`subpopulation of recipient NKT cells secretes high levels of IL-4
`and indirectly expands donor Treg populations to promote
`tolerance after BMT (74). In contrast, IL-4 may drive donor
`Th2 differentiation directly and enhance GVHD that likely
`usually represents chronic disease. Appreciation of these
`mechanisms is important, because treatment of a recipient or
`graft can be temporally separated and offers the opportunity to
`select desirable effects while avoiding potentially deleterious
`outcomes.
`Receptor disposition. Additional complicating factors exist when
`considering cytokine-mediated effects in immune-mediated and
`inflammatory conditions. IL-6 is an example of a cytokine for
`which signaling via “classical” or membrane-bound receptor–
`ligand interactions produces differing pathology than does
`signaling mediated through soluble or trans receptor binding.
`These effects were described originally in mouse models of
`rheumatoid arthritis, in which trans signaling (by the IL-6–
`soluble IL-6R complex) recapitulated inflammatory joint
`disease in IL-6–deficient mice, whereas injection of the
`native IL-6 cytokine itself did not (75). Subsequently, IL-6
`signaling through the trans pathway has been thought to be
`more inflammatory in nature than classical signaling, in part
`relating to the ability of IL-6 to signal through cells that ex-
`press the gp130 receptor complex but do not basally express
`IL-6R (76). A similar paradigm was demonstrated in al-
`lergic asthma: Th2 expansion appears to be driven by trans
`signaling whereby expansion of Tregs was limited by classical
`IL-6 signaling, and inhibition with anti–IL-6R mAb resulted
`in increased numbers of Tregs (77), and an increase in asthma
`risk was associated with a single nucleotide polymorphism
`that results in an increase in soluble IL-6R and trans signaling
`(78). Appreciation of the mechanisms by which a cytokine can
`mediate differential effects is critical to understanding both
`disease pathophysiology and effective clinical translation of
`therapeutics. The availability of mAbs to cytokine receptors,
`such as tocilizumab for IL-6R, which inhibits all IL-6 signaling,
`in addition to more specific inhibitors of signalling pathway
`components, such as soluble gp130:Fc, which inhibits IL-6
`trans signaling only, is a clear example.
`
`Translational application
`
`Accepted murine models of transplantation and rapid and
`reproducible multiplexed techniques to measure cytokines in
`serum or cell culture supernatants or intracellular cytokine
`
`

`

`4608
`
`BRIEF REVIEWS: CYTOKINES IN GVHD
`
`production by flow cytometry have allowed identification of,
`and will continue to define, the key cytokines in aGVHD
`(45, 79), as well as facilitate clinical translation of findings.
`However, a number of factors must be considered when ex-
`trapolating laboratory observations
`into clinical cohorts.
`Variation exists in transplantation protocols, patient pop-
`ulations, modes of conditioning, and posttransplant immune-
`suppression strategies. The last factor is of particular im-
`portance when considering the translation of observations
`made in animal models to the clinical setting, where immune
`suppression with cyclosporin or tacrolimus, combined with
`methotrexate or mycophenolate, is considered standard of
`care to avoid life-threatening acute and severe GVHD. How-
`ever, most animal models of transplantation rely solely on
`radiation-based conditioning. Therefore, effective transla-
`
`tion will require validation of observations made in animal
`models with clinical cohorts, because standard immune-
`suppressing agents were shown to affect cytokine levels pro-
`duced by T cells and NK cells, and profiles vary with stem cell
`source (80, 81). Importantly, IFN-g, TNF, and IL-1 are not
`systemically dysregulated in clinical subjects after BMT who
`receive immune suppression in the same way as seen in rodent
`models (45). With this in mind, it should be noted that no
`cytokine-inhibition strategy or cytokine administration has
`proved efficacious in randomized studies. In general, en-
`couraging results seen in preclinical studies and early-phase
`clinical trials have either not progressed into phase III studies,
`or effects have not been robust within this context [e.g., IL-1
`(82) and TNF inhibition (83, 84)]. Table I provides a sum-
`mary of cytokines and inhibitors that have been explored for
`
`Table I.
`
`Summary of relevant cytokine-targeted therapeutic studies
`
`Cytokine Inhibitors
`
`Phase I and/or II Clinical Trial Data
`
`Randomized, Double-Blind
`Controlled and/or Phase III
`Clinical Trial Data
`
`TNF-aR2 (etanercept)
`Prophylaxis
`Treatment
`TNF-a binding mAb (infliximab)
`Prophylaxis
`Treatment
`IL-1Ra (anakinra)
`Prophylaxis
`Treatment
`IL-2Ra/anti-CD25 (basiliximab/daclizumaba)
`Prophylaxis
`Treatment
`IL-6R (tocilizumab)
`Prophylaxis
`Treatment
`Keratinocyte growth factor (palifermin)
`Prophylaxis
`
`Cytokines
`
`IL-2 (aldesleukin)
`Prophylaxis
`Treatment
`IL-11 (oprelvekin)
`Prophylaxis
`
`Cytokines with Non-GVHD Benefits
`
`IFN-a (INTRON A, Roferon-A)—promotion of GVL with concomitant
`promotion of GVHD
`Prophylaxis
`Treatment
`Keratinocyte growth factor (palifermin)—for reduction of oral mucositis
`Prophylaxis
`
`Potential GVHD Therapies
`
`IL-17
`IL-17A mAb (secukinumab, ixekizumab, perakizumab)
`IL-17RA mAb (brodalumab)
`IL-17A/TNF (ABT122)
`IL-22 (fezakinumab)
`IL-12p40/23 mAb (ustekinumab)
`IL-23p19 (guselkumab, tildrakizumab)
`
`IL-13 (lebrikizumab, tralokinumab)
`
`+ (83)
`+ (102–105)
`
`2 (84)
`+ (99, 100)
`
`+ (106)
`
`+ (107)
`+ (108–110)
`
`+ (45)
`+ (113, 114)
`
`2 (115–119)
`
`+ (55)
`+ (51)
`
`2 (121)
`
`+ (122–126)
`+ (127, 128)
`
`+ (116–118)
`
`•
`•
`
`•
`2 (101)
`
`2 (82)
`•
`
`•
`2 (111, 112)
`
`•
`•
`
`2 (120)
`
`•
`•
`
`•
`
`•
`•
`
`+ (120)
`
`Phase I and/or II Clinical
`Data Outside GVHD
`
`Phase III Clinical Trial
`Data Outside GVHD
`
`+ Ixekizumab (129)
`+ (130)
`Ongoing
`Ongoing
`+ (97, 132)
`+ Guselkumab (133)
`+ Tildrakizumab (134)
`+ Tralokinumab (135)
`
`+ Secukinumab (131)
`•
`•
`•
`•
`
`Tildrakizumab (ongoing)
`+ 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
`to agents with potential efficacy in GVHD that are being
`explored in other disease settings. Importantly, most studies
`in GVHD examine the usefulness of cytokine antagonists as
`an adjunct to standard modalities of immune suppression
`rather than their efficacy in isolation, as is usually the case in
`preclinical testing. Thus, it will be important to follow some
`recently defined general principles, taking into consideration
`concurrent immune suppression, when planning to translate
`findings from mice to patients (28).
`The effect of a particular cytokine in any one individual is
`also affected by human genetic heterogeneity, and data already
`demonstrated a clear impact of single nucleotide polymorphisms
`in cytokine loci on GVHD outcomes (85, 86). Despite these
`difficulties in directly translating laboratory observations to the
`clinic, cytokine therapy for GVHD remains fertile ground for
`new and effective therapeutics, because a number of agents that
`augment or antagonize cytokine pathways are already available,
`having been explored and validated in other autoimmune and
`inflammatory disease settings. Cytokine inhibition,
`initially
`with TNF and subsequently with IL-6, is considered routine
`care for rheumatoid arthritis patients whose disease is not
`controlled by more nonspecific immune suppression with
`corticosteroids, methotrexate, and calcineurin inhibitors (87–
`90). Imperfect disease control, when used as monotherapy, has
`paved the way for the use of combination cytokine inhibition,
`and the rational combination of TNF and IL-17 showed efficacy
`in preclinical models of disease (91). Novel targets, such as IL-
`32, IL-34, and IL-35, are also being explored (92). The success
`of cytokine inhibition in rheumatoid arthritis is also paralleled in
`other inflammatory diseases, including psoriatic dermatitis and
`arthritis (93), with TNF inhibition having a demonstrated role,
`in addition to promising newer targets, such as IL-22 and IL-23
`(94, 95). Evidence for the value of cytokine inhibition exists in
`diseases other than inflammatory arthropathies, with efficacy
`demonstrated for TNF (96), as well as IL-12/23 (97, 98), in
`inflammatory bowel disease. In this setting of proven efficacy for
`cytokine inhibition in other diseases of dysregulated immunity,
`further definition of the role of cytokines in the determination of
`GVHD severity is likely to translate rapidly into efficacious
`therapies for the transplant patient population.
`
`Conclusions
`Cytokines are a defining influence on evolving immune
`responses in BMT and in the generation of GVHD, the major
`pathology limiting the wider application of transplantation.
`The classical appreciation of a naive T cell being influenced by
`a cytokine to mature into a more differentiated phenotype is
`made more complex in the GVHD setting as the impact of
`cytokines acting in different tissue compartments (e.g., lym-
`phoid and nonlymphoid or donor and host) and the use of
`classical and trans signaling pathways become better appreci-
`ated. Although these factors require further work to better
`define these complex interactions,
`they go some way in
`explaining the previously mixed and conflicting results asso-
`ciated with some cytokine therapies (99–101). Appreciation
`of this complexity will allow for the development of more
`log

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket