throbber
bjh
`
`review
`
`Advances in understanding the pathogenesis of graft-versus-
`host disease
`
`John Magenau, Lyndsey Runaas and Pavan Reddy
`
`Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan,
`Ann Arbor, MI, USA
`
`Summary
`
`Allogeneic haematopoietic stem cell transplantation (HCT) is
`a potent immunotherapy with curative potential for several
`haematological disorders. Overcoming the immunological
`barrier of acute graft-versus-host disease (GVHD) remains a
`fundamental impediment to expanding the efficacy of HCT.
`GVHD reflects a complex pathological interaction between
`the innate and adaptive immune systems of the host and
`donor. Over the past decade there has been a tremendous
`advancement in our understanding of the cellular and molec-
`ular underpinnings of this devastating disease. In this review,
`we cover several recently appreciated facets of GVHD patho-
`genesis including novel extracellular mediators of inflamma-
`tion, immune subsets, intracellular signal transduction, post-
`translation modifications and epigenetic regulation. We begin
`to develop general themes regarding the immunological path-
`ways in GVHD pathogenesis, discuss critical outstanding
`questions, and explore new avenues for GVHD treatment
`and prevention.
`
`Keywords: GVHD, allogeneic, transplantation.
`
`receive allogeneic
`While increasing numbers of patients
`haematopoietic stem cell transplantation (HCT) for aggressive
`haematological disorders, graft-versus-host disease (GVHD)
`remains a formidable barrier to maximizing its therapeutic
`efficacy. Despite the routine administration of immunosup-
`pressive prophylaxis, GVHD is the principle cause of trans-
`plant-related mortality (TRM). Clinically significant acute
`GVHD will occur in approximately 40% of patients undergo-
`ing human leucocyte antigen (HLA)-matched related HCT
`and upwards of 50–70% of patients receiving HLA-matched
`or -mismatched unrelated donor HCT (Jagasia et al, 2012).
`
`Correspondence: Pavan Reddy, M.D., Professor of Medicine Blood
`
`and Marrow Transplant Program, Division of Hematology/Oncology,
`
`Department of Internal Medicine, University of Michigan, 3312
`
`Cancer Center, 1500 E. Medical Center Drive, SPC 5932, Ann Arbor,
`
`MI 48109-5932, USA.
`
`E-mail: reddypr@umich.edu
`
`First published online 27 March 2016
`doi: 10.1111/bjh.13959
`
`b1 h BRITISH JOURNAL
`
`OF HAEMATOLOGY
`
`Unfortunately, less than half of the patients who develop acute
`GVHD experience a demonstrable response, partly due to the
`rapid pace of the disease as well as the marginal efficacy of pri-
`mary treatment with high dose corticosteroids (MacMillan
`et al, 2010; Choi & Reddy, 2014; Magenau & Reddy, 2014). In
`cases where organ damage is advanced or where high dose
`corticosteroid therapy is unsuccessful, mortality can exceed
`90% (Pasquini, 2008). Many patients who do ultimately
`respond to intensified immunosuppression must endure sig-
`nificant morbidity from infection, functional impairment and
`subsequent chronic GVHD. Finally, given that the efficacy of
`HCT relies heavily upon graft-versus-leukaemia
`(GVL)
`responses that are tightly linked to GVHD, intensive immune
`suppression may increase the risk of relapse and subsequent
`mortality (Rubio et al, 2015).
`
`Overview of GVHD biology
`
`The clinical features of acute GVHD manifest as an intense
`inflammatory injury primarily involving the skin,
`intestine
`and liver. Underlying
`this
`clinical presentation is
`an
`immunologically-mediated process of allogeneic donor cells
`responding to host tissues expressing polymorphic human
`leucocyte antigens (Blazar et al, 2012). The essential function
`of allogeneic donor T cells was theorized following an almost
`complete absence of GVHD in syngeneic and T cell-depleted
`HCT. However, it is now widely recognized that allogenic
`GVHD and GVL responses are fundamentally driven by ini-
`tial interactions between host and donor antigen presenting
`cells (APCs) that encounter mature T lymphocytes from the
`donor inoculum (Shlomchik et al, 1999; Matte et al, 2004;
`Reddy et al, 2005; Koyama et al, 2012; Toubai et al, 2012).
`The
`ensuing inflammatory
`cascade
`is
`self-perpetuating:
`release of pro-inflammatory cytokines results in expansion of
`alloreactive T cells with specificity to host tissues that,
`in
`turn, secrete additional inflammatory mediators.
`Over the past decade our knowledge of the intricacies that
`govern T cell and APC interactions have grown tremen-
`dously. A great number of mechanistic insights are derived
`from murine models of acute GVHD that sufficiently recapit-
`ulate acute GVHD in humans. As a consequence, a bewilder-
`ing array of cellular immune subsets, extracellular receptors,
`
`ª 2016 John Wiley & Sons Ltd
`British Journal of Haematology, 2016, 173, 190–205
`
`

`

`Immunobiology of GVHD
`
`mediators of inflammation and molecular signalling path-
`ways are now implicated in GVHD pathogenesis. In this
`review, we focus on critical advances in the understanding of
`acute GVHD pathogenesis. Emerging mechanistic themes are
`highlighted as well as opportunities for clinical translation,
`i.e. those that shape the allogeneic immune response to sup-
`press GVHD without compromising beneficial anti-tumour
`and anti-viral immunity (Table I).
`
`Initiating events
`
`According to Billingham’s postulates made almost 50 years
`ago, immunocompetent donor cells must experience histo-
`
`compatibility differences in the host for the development of
`GVHD (Billingham, 1966). It is now well established that
`disparities in major histocompatibility complex (MHC) loci
`on chromosome 6, as occurs in HLA-mismatched HCT, are
`directly proportional
`to the
`severity of
`acute GVHD
`(Flomenberg et al, 2004; Petersdorf et al, 2015). Today, the
`majority of HCT occurs with HLA-matched unrelated
`donors,
`thus differences in other polymorphic genes, so
`called minor histocompatibility antigens (mHA), provide the
`necessary tissue disparity to produce GVHD. Furthermore,
`given that adaptive immune responses require additional sig-
`nalling events to initiate GVHD, the importance of MHC
`independent events is becoming increasingly appreciated.
`
`Table I. Select pathways of GVHD pathogenesis with potential therapeutic approaches
`
`Aberrant pathway
`
`Approach
`
`Proposed mechanism(s)
`
`Clinical trial
`(phase I-II)
`
`Reference(s)
`
`CTLA4-Ig (Abatacept)
`
`Inhibiting costimulation
`CTLA4-Ig
`(Abatacept)
`Modulate cytokine(s)/chemokine(s)
`IL6
`anti-IL6 receptor mAb
`(Tocilizumab)
`anti-IL21 mAb
`Inhibit CCR5 coreceptor
`(Maraviroc)
`
`IL21
`CCR5
`
`Decrease DAMPs/PAMPs
`↑ protease
`Exogenous Alpha-1-antitrypsin
`inhibition
`(AAT/SERPINA1)
`
`Activate Siglecs
`
`CD24-Fc
`
`Alter microbiome
`
`Antibioltics, probiotics, other
`
`Improve peripheral tolerance
`Expand/induce
`Treg graft engineering, Graft
`Tregs
`Selection, Pharmacological
`(several)
`ex vivo iNKT expansion,
`adoptive transfer
`Graft Engineering (select
`memory T cells)
`
`iNKT
`
`Decreased activation of T cells
`
`↓Th1, Th17, Tissue Damage; ↑Treg;
`
`↓ Th1, Th17; ↑ iTreg
`↓ T cell homing to Gut
`
`Suppress release of DAMPs; Limit
`cytokine production
`
`Sequester DAMPs, ↓DAMP-
`mediated DC activation
`Gut protection through modifying
`microbiome and byproducts
`
`Inactivate/delete alloreactive T cells
`
`Promotes Treg expansion
`
`Reduce alloreactive T cells
`
`Naive T cell
`depletion
`Intracellular signalling
`Tyrosine kinases
`NF-jB
`
`STATs
`Epigenetic
`DNA methylation
`
`JAK-1/2 inhibition
`c-Rel inhibition, proteasome
`inhibition (Velcade),
`neddylation inhibition
`(MLN4924)
`STAT3 inhibition
`
`Increase Treg:Teff ratios, ↓STAT
`↓transcription of multiple
`inflammatory cytokines
`
`↑ FOX P3 Expression (iTREG)
`
`Azacitidine
`
`Promotes iTreg expansion
`
`Histone acetylation Vorinostat
`
`Bromodomain
`(BET) proteins
`
`I-BET-151, JQ1
`
`Increase IDO, decrease
`inflammatory cytokines
`↓DC activation; ↓T-cell
`proliferation; NF-kB signalling
`
`DAMPs, damage-associated molecular patterns; PAMPs, pathogen-associated molecular patterns.
`
`ª 2016 John Wiley & Sons Ltd
`British Journal of Haematology, 2016, 173, 190–205
`
`+
`
`+
`
`
`+
`
`+
`
`
`
`
`
`+
`
`
`
`+
`
`+
`+
`
`
`
`+
`
`+
`
`
`
`Miller et al (2010)
`
`Drobyski et al (2011);Kennedy
`et al (2014)
`Hippen et al (2012)
`Reshef et al (2012)
`
`Marcondes et al (2011); Tawara
`et al (2012); Brennan et al
`(2012)
`Chen et al (2009a); Toubai et al
`(2014)
`Atarashi et al (2013); Jenq et al
`(2015)
`
`Di Ianni et al (2011); Brunstein
`et al (2011)
`
`Schneidawind et al (2015)
`
`Bleakley et al (2015)
`
`Spoerl et al (2014)
`Shono et al (2014); Mathewson
`et al (2013)
`
`Laurence et al (2012)
`
`Choi et al (2010); Goodyear
`et al (2012)
`Reddy et al (2008); Choi et al
`(2014)
`Sun et al (2015a)
`
`191
`
`

`

`J. Magenau et al
`
`Costimulation
`
`While the primary interaction occurs between the MHC/al-
`lopeptide on APCs and the T cell receptor (TCR) of donor
`T cells, this signal alone is insufficient to induce T cell acti-
`vation. Several studies in HCT and related haematological
`fields have focused on the effect of the required second or
`co-stimulatory signal on T cell activation and GVHD
`(McDonald-Hyman et al, 2015). Second signals between
`APCs and T cells may positively or negatively influence the
`immune response (Fig 1A). CTLA4 is one of several co-sti-
`mulatory molecules
`expressed on differentiated T cells
`which functions as a key negative regulator of T cell activa-
`tion. CD28, a structural homolog of CTLA4, provides a
`positive co-stimulatory signal to T cells. By competing with
`greater affinity than CD28, CTLA4 preferentially binds
`ligands on APCs (B7-1/2 or CD80/86) thereby constraining
`T cell proliferation (Mueller, 2010; Fig 1B). CTLA4-immu-
`noglobulin or Abatacept is a US Food and Drug Adminis-
`tration (FDA) approved therapy for rheumatoid arthritis
`that fuses a modified Fc portion of human immunoglobu-
`lin-G1with CTLA4. Abatabcept increases the availability of
`CTLA4 which probably minimizes activating interactions
`with CD28. Based on encouraging pre-clinical evidence of
`GVHD suppression and pilot studies in humans (Miller
`et al, 2010), abatacept is currently being tested in a phase
`II multi-centre randomized study. This mechanism is nota-
`bly distinct from the alternate strategy of directly inhibiting
`CTLA4, which activates T cells (e.g.
`ipilimumab; Fig 1C).
`
`Interestingly, toxicity related to ipilimumab (without allo-
`geneic HCT) may resemble GVHD.
`Programmed cell death-1 (PD-1, also termed PCDC1) is
`another distinct inhibitory signal that has been implicated in
`maintaining peripheral
`tolerance (Mueller, 2010). Because
`PD-1 and PD-1 ligand (also termed CD274) interactions
`promote T cell exhaustion, this pathway may be physiologi-
`cally important
`for eliminating chronically self-reactive T
`cells. While pre-clinical studies after HCT suggest interrupt-
`ing these interactions may aggravate acute GVHD, delaying
`PD-1 blockade or selective interruption of PD ligands with
`differing
`tissue distribution may promote
`graft-versus-
`tumour (GVT) responses without aggravating GVHD (Koest-
`ner et al, 2011; Saha et al, 2013).
`Therapeutic
`interventions
`surrounding the TCR:MHC
`immune synapse must account for orchestrating co-stimula-
`tory and co-inhibitory signals, but also time-dependent effects
`specific to immune cells. As an example, the B7-H3 (also ter-
`med CD276) membrane protein, widely expressed on T cells,
`natural killer cells, dendritic cells (DCs) and macrophages, is
`an activating co-stimulatory signal that mediates transplant
`rejection (Wang et al, 2005). However, B7-H3 can also effec-
`tively curtail alloreactive T cells and GVHD in the early HCT
`period (Veenstra et al, 2015) and donor lymphocyte infusion
`with T cells lacking B7-H3 promotes GVL responses without
`GVHD. These studies illustrate the time dependence of co-sti-
`mulatory pathways, thus agonism or antagonism both may
`present viable therapeutic strategies after HCT.
`
`Fig 1. Co-stimulatory interactions between
`APCs & T cells. Co-stimulatory ligands and
`receptors (A) provide positive or negative sec-
`ond signals together with the TCR that deter-
`mine the activation status of T cells. Clinically
`available pharmacological agents modulate co-
`stimulatory interactions on APCs and T cells.
`Interrupting B7:CD28 interactions (B) can sup-
`press T cells through loss of activating second
`signals, potentially impeding graft-versus-host
`disease. Conversely, targeting CTLA4 or
`PDCD1 (C) can activate T cells through blunt-
`ing inhibitory second signals. MHC, major his-
`tocompatibility complex; TCR, T-cell receptor;
`APC, antigen presenting cell.
`
`ª 2016 John Wiley & Sons Ltd
`British Journal of Haematology, 2016, 173, 190–205
`
`192
`
`

`

`Mediators of inflammation: cytokines and chemokines. Once
`activated, T cells initiate transcriptional programmes that
`result in the massive release of pro-inflammatory mediators
`[tumour necrosis factor a (TNF-a, TNF), interleukin 1 (IL1),
`c interferon (IFN-c,
`IFNG)]
`that amplify the immune
`response and result in tissue damage (Paczesny et al, 2010).
`In addition, cytokines and chemokines influence prolifera-
`tion, differentiation and homing of effector cells to GVHD
`target
`tissues. This knowledge laid the foundation for
`attempts at blocking single cyto/chemokines to ameliorate
`GVHD. This approach has been met with modest clinical
`success, but
`is gaining renewed interest
`from pre-clinical
`studies suggesting that early interruption of novel cytokines
`can impede multiple cellular pathways in GVHD.
`Interleukin 6 (IL6): There is compelling evidence that IL6
`promotes GVHD by divergent mechanisms including increas-
`ing inflammatory T-helper cells types 1 and 17 (Th1 and
`Th17, respectively) subsets, decreasing regulatory T cells
`(Tregs) and by direct cytotoxicity (Chen et al, 2009b; Tawara
`et al, 2011). In models of HCT, levels of IL6 are elevated,
`following cytotoxicity from conditioning or GVHD.
`IL6
`blockade suppresses experimental GVHD without impairing
`the GVL response. Given the clinical availability of
`the
`humanized anti-IL6 receptor antibody,
`tocilizumab,
`for
`rheumatoid arthritis, IL6 blockade was piloted in steroid-
`refractory acute GVHD without untoward toxicity (Drobyski
`et al, 2011). Because IL6 is elevated early after HCT, a single
`centre phase I/II trial evaluated whether early intervention
`with tocilizumab might reduce GVHD in HLA-matched
`related and unrelated donors (Kennedy et al, 2014). Correla-
`tive studies revealed that downstream STAT3 was reduced in
`monocytes and T cells from patients receiving tocilizumab,
`implying successful IL6 blockade. The rate of grade II-IV
`GVHD was 12% and severe grade III-IV GVHD was 4%,
`supporting additional studies in GVHD prevention.
`Interleukin-21 (IL21):
`is capable of promoting Th1 and
`Th17 differentiation, NK cell expansion and formation of
`inducible Tregs (iTregs) in addition to impacting a wide
`range of
`lymphoid and myeloid cells
`including APCs.
`Administration of IL21 inhibitors or IL21-deficient T cells
`mitigates gastrointestinal
`(GI) GVHD without
`impairing
`GVL responses, perhaps due to its tissue-specific effects
`(Bucher
`et al, 2009).
`In humans,
`IL21 expression was
`increased in the GI tracts of patients with GVHD. In a xeno-
`graft model, prophylactic administration of anti-human IL21
`reduced GVHD lethality (Hippen et al, 2012). Exogenous
`IL21 is currently being evaluated in immunotherapy trials for
`cancer, while IL21 blocking strategies are being developed for
`autoimmune conditions.
`Interleukin-2 (IL2): Interrupting IL2 secretion by donor T
`cells, via the calcineurin inhibitors (CNIs), ciclosporin and
`tacrolimus, is an established standard of care in GVHD pre-
`vention. However, subsequent randomized trials directly tar-
`geting
`the
`IL2 receptor
`(i.e. daclizumab) have been
`unsuccessful in acute GVHD treatment, partly due to high
`
`ª 2016 John Wiley & Sons Ltd
`British Journal of Haematology, 2016, 173, 190–205
`
`Immunobiology of GVHD
`
`rates of relapse (Lee et al, 2004). While CNIs reduce effector
`T cell function, an unintended consequence of prolonged IL2
`inhibition may be that Treg generation is constrained. In
`support of
`this hypothesis, murine data and a recent
`prospective trial demonstrated that administration of
`low
`dose IL2 in chronic GVHD results in regression of disease
`(Koreth et al, 2011). The mechanism of IL2 may relate to
`differential effects on STAT5 proteins in dividing lympho-
`cytes that restore a more favourable balance between Tregs
`and conventional T cells (Matsuoka et al, 2013).
`Chemokine (C-C motif) receptor-5 (CCR5): CCR5 is upreg-
`ulated in T lymphocytes upon allogeneic stimulation and
`directs homing to target tissues such as the GI tract. HCT
`recipients with a missense mutation of CCR5 (Delta32)
`appear less susceptible to developing clinical GVHD (Bogu-
`nia-Kubik et al, 2006). A phase I/II clinical trial using a small
`molecule antagonist of CCR5 (maraviroc) demonstrated
`promising results for GVHD prevention (Reshef et al, 2012)
`and is now being prospectively evaluated in a larger clinical
`trials network (CTN) study (NCT02208037).
`Interleukin-22 (IL22): Our understanding of cytokines has
`expanded to include identification of molecules capable of
`tissue repair. IL22 acts upon intestinal stem cells (ISCs) that
`are critically involved in epithelial repair. Because the GI
`tract is exquisitely sensitive to the cytotoxic effects of condi-
`tioning, its damage responses are instrumental for propagat-
`ing GVHD. Deficiency of IL22 promotes loss of ISCs, greater
`intestinal damage and more severe experimental GVHD
`(Hanash et al, 2012).
`Suppression of Tumourigenicity 2 (ST2): ST2 functions as
`both a soluble (sST2) and membrane bound receptor (ST2L)
`for IL33, a cytokine in the IL1 receptor family with diverse
`immunological
`roles depending on disease, cell
`type or
`model system. Using proteomic methods to compare plasma
`levels prior to and during GVHD, elevations in sST2 have
`been identified as an independent biomarker of treatment
`resistance and mortality (Vander Lugt et al, 2013). While
`high ST2 levels predict GVHD mortality, non-specific tissue
`damage or the genetic background of the host could also
`influence plasma concentrations (Ito & Barrett, 2015). High
`levels of ST2 and IL33 are produced by non-haematopoietic
`cells in the GI tracts of animals with GVHD (Reichenbach
`et al, 2015; Zhang et al, 2015). IL33 binding to ST2 aggra-
`vates GVHD, which can be reversed in IL33(/) deficient
`hosts or in T cells lacking ST2 expression. In pre-clinical
`models, minimizing IL33/ST2 interactions by administering
`an ST2-Fc fusion protein reduced GVHD.
`FTY720 (FTY): FTY is an immunomodulator derived from
`a metabolite of the fungus Isaria sinclairii. It binds with high
`affinity to sphingosine 1-phosphate receptors found on all
`cell types and is thought to exert its immunomodulatory
`effects through sequestering of lymphocytes within secondary
`lymphoid organs. Given this, there was interest in evaluating
`FTY to prevent or treat GVHD as an oral FTY (fingolimod)
`was approved by the FDA in 2010 to treat relapsing multiple
`
`193
`
`

`

`J. Magenau et al
`
`sclerosis. Taylor et al (2007) were able to demonstrate that
`FTY significantly inhibited but did not completely prevent
`GVHD in a model of GVHD. Similarly, administration of
`FTY slightly mitigated but did not eliminate GVL effect
`(Taylor et al, 2007).
`Retinoic acids: Retinoic acid (RA) is produced by intestinal
`cells and is known to have a role in intestinal
`immune
`homeostasis; however, its exact role in the pathophysiology
`of GVHD remains uncertain. Nishimori et al (2012) demon-
`strated that administration of a RA analogue ameliorated
`cGVHD. The effects on acute GVHD appear more nuanced
`with administration of a vitamin-A deficient diet reducing
`gut acute GVHD but worsening liver GVHD (Koenecke et al,
`2012). Similarly, Aoyama et al (2013) found that inhibiting
`donor T-cell RA receptor signalling reduced GVHD while
`preserving graft-versus-lymphoma effects.
`
`Modulating innate immune responses
`
`As DCs are primary sensors of initial inflammatory signals,
`their regulation can significantly shape GVHD. Currently,
`several efforts are underway to understand if modulating DC
`function can be utilized for treating GVHD.
`
`Damage-associated molecular patterns (DAMPs)
`
`Immune and myeloablative conditioning therapy preceding
`HCT provides important anti-tumour effects, but also causes
`direct immune activation by tissue injury and release of pro-
`inflammatory mediators. Large scale observational studies
`
`have confirmed a modest yet significant increase in acute
`GVHD incidence with higher conditioning intensity, espe-
`cially in regimens containing total body irradiation (TBI)
`(Nakasone et al, 2015). A growing body of evidence suggests
`that highly conserved Toll-like receptors (TLRs) and other
`pattern recognition receptors on innate immune cells acutely
`sense endogenous ‘danger’ signals from DAMPs, such as
`ATP, HMGB1, uric acid (UA) and heat-shock proteins, that
`provide a crucial
`initiating step in alloreactive T cell
`responses (Wilhelm et al, 2010; Jankovic et al, 2013; Brennan
`et al, 2015; Fig 2).
`The purine nucleoside ATP, when present in the extracel-
`lular space following tissue injury, is one of several early dan-
`ger signals or DAMPs. ATP released into peritoneal fluids of
`humans and mice following radiation interacts with P2X7
`causing expression of co-stimulatory molecules, phosphoryla-
`tion of
`signal
`transducer and activator of
`transcription
`(STAT) proteins and production of inflammatory cytokines.
`Antagonizing ATP:P2X7 interactions
`limits mortality in
`models of GVHD (Wilhelm et al, 2010). Other DAMPs are
`increasingly being recognized as initiators of the alloimmune
`response. For example, tissue injury releases the extracellular
`matrix component UA, which, in turn, activates the NLRP3
`inflammasome (Jankovic et al, 2013). Mice under conditions
`of less UA or deficient in components of the NLRP3 complex
`demonstrate less severe GVHD. Heparin sulfate (HS), also a
`DAMP, is similarly capable of activating alloreactive T cell
`responses through binding TLR4 receptors on DCs and ele-
`vated levels correlate with GVHD (Brennan et al, 2012).
`While inhibiting single DAMPs is effective in suppressing
`
`Cytokines
`
`•• • A..._ Tocilizumab (anti-lL6)
`
`Maraviroc
`
`Activation of Adaptive
`Immunity
`
`AAT
`CD24Fc
`
`Activation of Innate Immunity
`
`Fig 2. Role of DAMPs & Novel Cytokines in GVHD Pathogenesis. Tissue damage following conditioning (chemotherapy/radiation) results in the
`release of numerous sterile inflammatory mediators, termed damage-associated molecular patterns (DAMPs), that together with cytokines con-
`tribute to the initiation of acute GVHD. Inflammatory mediators activate innate immunity through interactions with toll-like receptors (TLR)
`and cytokine receptors (not depicted) on APCs. These interactions promote activation of the adaptive immune response characterized by T cell
`differentiation, proliferation, and migration that perpetuates GVHD and worsens tissue damage. Certain investigational agents (e.g. AAT,
`CD24Fc) may mitigate GVHD by either sequestering DAMPs or regulating APC-mediated responses to DAMPs. GVHD, graft-versus-host disease;
`APC, antigen presenting cell; AAT, alpha-1-antitrypsin; CD24Fc, CD24 fusion protein; HS, heparin sulfate; UA, uric acid; HS, heat shock protein.
`
`194
`
`ª 2016 John Wiley & Sons Ltd
`British Journal of Haematology, 2016, 173, 190–205
`
`

`

`translation may
`clinical
`successful
`experimental GVHD;
`require inhibiting many DAMPs and their recognition by
`immune cells. Furthermore, it remains to be determined if
`DAMPs play as pivotal a role in settings of reduced intensity
`conditioning where tissue damage is less pronounced.
`
`Alpha-1-antitrypsin (AAT, also termed SERPINA1). AAT is
`an abundant serine protease inhibitor produced as an acute
`phase reactant by the liver which is classically described for
`its role in inhibiting neutrophil elastase. Congenital AAT
`deficiency results in emphysema, particularly under condi-
`tions of chronic noxious stimuli, such as cigarette smoke
`(Silverman & Sandhaus, 2009). Over the past several years,
`numerous additional roles of AAT have been identified, such
`as promoting tolerance in models of islet cell transplantation
`(Koulmanda et al, 2008). In allogeneic HCT models, human
`AAT administration reduces GVHD, proportionally increases
`Tregs without
`impairing T cell responses and suppresses
`multiple pro-inflammatory cytokines (Tawara et al, 2012).
`Similar results have been observed by other groups (Marcon-
`des et al, 2011; Brennan et al, 2012). The mechanisms by
`which AAT attenuates DC responses remain uncertain, but
`may involve less tissue release of DAMPs, such as HS, (Bren-
`nan et al, 2012) or by limiting the glycolytic capacity of
`metabolically active T cells (Marcondes et al, 2014).
`In
`humans, stool levels of AAT are increased in patients with
`severe GI GVHD (Rodriguez-Otero et al, 2012), suggesting
`patients may have a relative deficiency or be less able main-
`tain tissue concentrations that exert a protective effect. Given
`these observations, proof of concept clinical trials of exoge-
`nous AAT supplementation in steroid-refractory GVHD are
`now underway.
`
`Sialic-acid–binding
`lectin-G (Siglec-
`immunoglobulin-like
`G). Siglec-G residues, broadly expressed on B-cells, DCs and
`macrophages, serve as key negative regulators of DAMP dri-
`ven immune activation (Crocker et al, 2007). Recent pre-
`clinical data suggest Siglec-G expression is important
`for
`maintaining protective effects against GVHD (Toubai et al,
`2014). Following HLA-matched and -mismatched HCT,
`/
`Siglec-G
`deficient animals have heightened release of mul-
`tiple pro-inflammatory cytokines, stimulate greater numbers
`of alloreactive T cells and ultimately experience inferior sur-
`vival due to increased GVHD severity. Host tissue injuries
`caused by myeloablative HCT regimens, including high-dose
`chemotherapy and/or TBI not only promote release of
`DAMPs, but also reduce Siglec-G expression (Toubai et al,
`2014). Thus, conditioning therapy,
`through its effect on
`siglecs, may limit the capacity of immune cells to attenuate
`the damage response. The membrane glycoprotein CD24,
`expressed on haematopoietic cells including T lymphocytes,
`directly binds DAMPs and Siglec-G down-regulating immune
`stimulation (Liu et al, 2009).
`It is postulated that the CD24-Siglec-G pathway plays a
`critical role in preventing exaggerated responses to pathologi-
`
`ª 2016 John Wiley & Sons Ltd
`British Journal of Haematology, 2016, 173, 190–205
`
`Immunobiology of GVHD
`
`cal cell death, and most importantly, discriminating between
`exaggerated damage responses (i.e. DAMPs) while retaining
`immunity against pathogens (i.e. pathogen-associated molec-
`ular patterns, PAMPs) (Chen et al, 2009a). As HCT is associ-
`ated with tissue injury that additionally places hosts at risk
`for infection, the CD24-Siglec-G pathway is a compelling tar-
`get for GVHD prevention after myeloablative conditioning.
`For example, CD24Fc (CD24Fc immunoglobulin) is a fully
`humanized fusion protein consisting of
`the extracellular
`domain of mature CD24 linked to the human immunoglob-
`ulin G1 (IgG1) Fc domain. Similar to native CD24, in vitro
`studies demonstrate that CD24Fc binds murine Siglec-G
`(and its human orthologue, Siglec 10) (Chen et al, 2009a).
`In pre-clinical models, administration of exogenous CD24Fc
`promoted Siglec G activation and prevented GVHD (Toubai
`et al, 2014). Clinical studies of CD24Fc for GVHD preven-
`tion are currently underway in the settings of heightened tis-
`sue injury, such as myeloablative HCT. Nonetheless, whether
`enhanced signalling through other negative immune regula-
`tory pathways besides Siglecs can also mitigate GVHD
`remains to be explored.
`
`Role of microbiome in GVHD
`
`The interplay of the intestinal microbiome and GVHD has
`been a topic of investigation since the 1970s when van Bek-
`kum (1977) observed delayed GVHD after gut decontamina-
`tion. Rigorous examination of
`the relationship between
`GVHD and microbiota has only been possible with recent
`advances
`in technology allowing for culture-independent
`rRNA gene sequencing. With this new tool,
`investigators
`have noted a profound loss of bacterial diversity in murine
`models of GVHD (Eriguchi et al, 2012; Jenq et al, 2012;
`Tawara et al, 2013). Holler et al (2014) demonstrated that
`eliminating certain bacterial species prior to HCT, such as
`Lactobacillus, correlates with increased GVHD pathology.
`This process can be reversed by re-introduction of these
`organisms, suggesting that loss of certain commensal bacteria
`may promote GVHD severity. Similarly, the human micro-
`biome shifts towards potentially pathogenic organisms, such
`as Enterococci, after HCT. This shift is pronounced after
`antimicrobial usage and most notable after developing
`intestinal GVHD (Holler et al, 2014). Recently,
`increased
`bacterial diversity was associated with reduced mortality
`(Jenq et al, 2015). Specifically, higher proportions of bacteria
`belonging to the genus Blautia was linked to decreased
`GVHD-specific mortality and improved survival. Of particu-
`lar clinical interest, loss of Blautia is associated with use of
`antibiotics targeting anaerobes as well as parenteral nutrition;
`suggesting two potential
`interventions
`to mitigate acute
`GVHD. However, murine experiments suggest that the donor
`microbiome does not seem to affect GVHD severity in the
`hosts (Tawara et al, 2013).
`Given these findings, work is underway to actively manip-
`ulate the intestinal microbiome in a favourable manner.
`
`195
`
`

`

`J. Magenau et al
`
`Atarashi et al (2013) identified a cocktail of 17 species of
`Clostridia that potently enhance host Treg cell abundance
`and can reverse autoimmune colitis. In a murine model of
`GVHD, administration of this same cocktail was shown to
`reduce GVHD and improve survival
`(Mathewson et al,
`2014). Despite advances in technology that have allowed a
`better understanding into the microbiome, many questions
`remain, including the effect of prophylactic antimicrobials,
`optimal strategies for protection of regenerative cell popula-
`tions and advisability of the administration of exogenous
`commensal flora. These remain areas of active research.
`
`Cellular mediators of peripheral tolerance
`
`The actions of inflammatory signals play a large role in shap-
`ing the cellular milieu that follows HCT. As adults have
`undergone thymic involution, and mature lymphocytes from
`the donor inoculum drive GVHD, post-HCT immunity lacks
`normative central tolerance mechanisms, such as negative
`selection, that delete reactive T cells and B cells. These condi-
`tions imply a greater role for peripheral immune tolerance
`mechanisms in regulating the allogeneic responses. Induction
`of peripheral tolerance to reduce GVHD are being actively
`explored.
`
`Expansion of Tregs
`
`Tregs serve a crucial role in maintaining peripheral self-toler-
`ance. Natural Tregs are characterized by intracellular expres-
`sion of the transcription factor forkhead box P3 (FOXP3). In
`HCT, murine
`studies
`show that
`adoptive
`transfer of
`CD4+CD25+ Tregs protects against lethal GVHD (Edinger
`et al, 2003). A notable observation is that the suppressive
`effects of Tregs appear to have a nominal impact on immune
`recovery and GVL (Blazar et al, 2012). However, evaluating
`this in patients will require methods for generating high
`ratios of Tregs to T effectors that are sustainable in vivo. One
`approach is ex vivo expansion with adoptive transfer. In the
`setting of HLA-mismatched haploidentical HCT, infusion of
`donor Tregs prior to CD34+ selected stem cells resulted in
`very low rates of acute GVHD, without standard prophylactic
`immunosuppression (Di Ianni et al, 2011). In double cord
`unit HCT, escalating doses of third party expanded Tregs
`limited GVHD compared to historical controls (Brunstein
`et al, 2011). As further refinements occur, production of
`more stable populations of Tregs is anticipated, which will
`enable more definitive clinical studies of GVHD prevention.
`Nevertheless, these bench to clinic translations provide strik-
`ing examples of the potential of cellular therapy to mitigate
`human disease.
`
`Inducible Tregs
`
`Another approach to improving the availability of Tregs is
`the conversion of conventional T cells into iTregs. In fact,
`
`196
`
`some pharmacological agents described in this review exert
`their suppressive actions through iTreg expansion. The pro-
`cess of conferring antigen specificity to iTregs, as a means of
`improving the efficiency and selectively of their suppressive
`function is also being explored. For example, iTregs specific
`for the naturally occurring male Y chromosome minor histo-
`compatibility antigen (miHAg), termed HY, can be generated
`to prevent experimental GVHD (Li et al, 2015). As antici-
`pated, HY-specific iTregs exhibit enhanced expansion and
`stability in male hosts, making it conceivable that these tech-
`niques could expand Tregs against other relevant mHA.
`Whether iTregs and conventional T cells require simil

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