throbber
.•
`
`Mouse models of graft-versus-host
`disease*
`Pavan Reddy1 and James L.M. Ferrara1,§, 1University of Michigan Cancer
`Center, Ann Arbor, Ml 48109-5942, USA
`
`Table of Contents
`1. Introduction ............................................................................... 1
`2. Mouse models ............................................................................. 2
`3. Immunobiology ............................................................................ 3
`3.1. Phase 1: Activation of Antigen Presenting Cells (APCs) ................................... 3
`3.2. Phase 2: Donor T cell activation, differentiation and migration ............................. 5
`3.2.1. Costimulation ................................................................. 5
`3.2.2. T cell subsets .................................................................. 5
`3.2.3. Cytokines and T cell differentiation ............................................... 7
`3.2.4. Leukocyte migration ............................................................ 8
`3.3. Phase 3: Effector phase .............................................................. 8
`3.3.1. Cellular effectors .............................................................. 9
`3.3.2. Inflammatory effectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
`4. Conclusion ............................................................................... 11
`5. References ............................................................................... 11
`
`1. Introduction
`
`Allogeneic hematopoietic cell transplantation (HCT) represents an important therapy for many hematological
`and some epithelial malignancies and for a spectrum of nonmalignant diseases (Appelbaum, 2001). The development
`of novel strategies such as donor leukocyte infusions (DLI), nonmyeloablative HCT and cord blood transplantation
`(CBT) have helped expand the indications for allogeneic HCT over the last several years, especially among older
`patients (Welniak et al., 2007). However, the major toxicity of allogeneic HCT, Graft-Versus-Host disease (GVHD),
`remains a lethal complication that limits its wider application (Ferrara and Reddy, 2006). Depending on when it occurs
`after HCT, GVHD can be either acute or chronic (Deeg, 2007; Weiden et al., 1979; Weiden et al., 1981; Lee, 2005).
`Acute GVHD is responsible for 15% to 40% of mortality and is the major cause of morbidity after allogeneic HCT,
`while chronic GVHD occurs in up to 50% of patients who survive three months after HCT. Mouse models have
`provided the majority of insights into the biology of this complex disease process.
`
`*Edited by Diane Mathis and Jerome Ritz. Last revised October 20, 2008. Published February 28, 2009. This chapter should be cited as: Reddy,
`P. and Ferrara, J.L.M., Mouse models of graft-versus-host disease (February 28, 2009), StemBook, ed. The Stem Cell Research Community,
`StemBook, doi/10.3824/stembook. l .36. l, http://www.stembook.org.
`Copyright: © 2009 Pavan Reddy and James L.M. Ferrara. This is an open-access article distributed under the terms of the Creative Commons
`Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
`§To whom correspondence should be addressed. E-mail: ferrara@umich.edu
`
`stembook.org
`
`Pharmacyclics Exhibit 2054
`Sandoz v. Pharmacyclics
`IPR2019-00865
`
`

`

`Mouse models of graft-versus-host disease
`
`The GVHD reaction was first noted when irradiated mice were infused with allogeneic marrow and spleen
`cells (van Bekkum and De Vries, 1967). Although mice recovered from radiation injury and marrow aplasia, they
`subsequently died with "secondary disease" (van Bekkum and De Vries, 1967), a syndrome that causes diarrhea,
`weight loss, skin changes, and liver abnormalities. This phenomenon was subsequently recognized as GVHD disease
`(GVHD). Three requirements for the developing of GVHD were formulated by Billingham (Billingham, 1966-1967).
`First, the graft must contain immunologically competent, now recognized as mature T cells. In both experimental and
`clinical allogeneic BMT, the severity of GVHD correlates with the number of transfused donor T cells (Kernan et
`al., 1986; Komgold et al., 1987). The precise nature of these cells and the mechanisms they use are now understood
`in greater detail (discussed below). Second, the recipient must be incapable of rejecting the transplanted cells (i.e.,
`immunocompromised). A patient with a normal immune system will usually reject cells from a foreign donor. In
`allogeneic BMT, the recipients are usually immunosuppressed with chemotherapy and/or radiation before stem cell
`infusion (Welniak et al., 2007). Third, the recipient must express tissue antigens that are not present in the transplant
`donor. This area has been the focus of intense research that has led to the discovery of the major histocompatibility
`complex (MHC; Petersdorf and Malkki, 2006). Human leukocyte antigens (HLA) are proteins that are the gene
`products of the MHC and that are expressed on the cell surfaces of all nucleated cells in the human body, HLA proteins
`are essential to the activation of allogeneic T cells (Petersdorf and Malkki, 2006; Krensky et al., 1990) discussed
`below. This chapter on mouse models of acute GVHD will place the immuno-biological mechanisms of Billingham 's
`postulates in perspective.
`
`In addition to these seminal postulates on GVH reaction, the critical requirement of immune cells from the
`donor graft for optimal leukemia/tumor elimination: a process called graft-versus-leukemia (GVL) effect, and its tight
`link with GVHD were initially made from mouse models (43). Other models such as the canine, nonhuman primate,
`and rat models also played important roles, particularly in the development of clinically used immuno-suppressants.
`Nonetheless, the presence of well-characterized in-bred strains, availability of knock-out and transgenic animals, easy
`availability of reagents, and the relative low cost have made mouse models the most utilized systems for investigating
`the mechanisms of GVH responses.
`
`2. Mouse models
`
`Mouse models of GVHD can be grouped into those in which GVHD is directed to MHC (class I, class II, or
`usually both) or to isolated multiple minor HA alone. Although multiple minor HA mismatches also exist in the former,
`their impact is usually limited relative to that induced by full MHC disparities (Reddy et al., 2008). The GVHD that
`develops in response to a full (class I and II) MHC disparity is dependent on CD4 T cells and CDS T cells provide
`additive pathology. These systems result in an inflammatory "cytokine storm," capable of inducing GVHD in target
`tissues without the requirement for cognate T cell interaction with MHC on tissue (Teshima et al., 2002). In contrast
`to CD4-dependent GVHD, CDS T cells induce GVHD primarily by their cytolytic machinery, which requires the
`TCR to engage MHC on target tissue (Reddy et al., 2008) The induction of GVHD to multiple minor HA results in
`a process where either CDS T cells, CD4 T cells, or both, depending on the strain combination (see Table 1) may
`play a role in disease. These different models have helped dissect and refine the various other complex aspects of
`GVHD (see below). It is critical from the outset to understand that although most clinical BMT recipients are MHC
`matched but minor HA disparate with the donor, there is no one single most appropriate mouse model of clinical BMT.
`Experimentally both the MHC disparate and minor HA disparate systems can also induce the full or certain specific
`aspects of the spectrum of clinically relevant GVHD while permitting the dissection of immunologic mechanisms.
`
`Most mouse models employ radiation for conditioning the recipient animals. Inbred mouse strains demonstrate
`variable sensitivity to radiation, so maximal tolerated total body irradiation (TBI) doses differ from strain to strain.
`For example, B6 are more resistant that BALB/C mice, and Fl hybrids are usually either more resistant than parental
`strain. Generally, the higher the TBI dose, the earlier and greater the intensity of the inflammatory arm of GVHD(see
`below) and BMT models utilizing low TBI doses and high donor T cell doses will result in GVHD dominated by
`later onset T cell-dependent pathology (Reddy et al., 2008). Chemotherapeutic conditioning with cyclophosphamide,
`fludarabine, and busulfan can also be delivered in mouse systems (Ferrara et al., 2005).
`
`Available mouse models (see Table 1) nicely mimic the spectrum of acute GVHD but the induction of clinically
`relevant chronic GVHD in mouse models using nonmutated inbred strains is challenging. Amongst the commonly
`utilized models, they either mimic only a few and not all of the manifestations or the kinetics of chronic GVHD. As
`such, this paucity of appropriate mouse models for chronic GVHD has resulted in a lack of significant understanding
`of the immunobiology of chronic GVHD when compared with acute GVHD. Below we briefly discuss the current
`understanding of immuno-biological mechanisms of acute GVHD derived from utilizing mouse models.
`
`2
`
`stembook.org
`
`

`

`Mouse models of graft-versus-host disease
`
`Donor
`
`Host
`
`GVHD targets
`
`T cell dependence
`
`Acute GVHD Models
`B6
`B6
`BALB/c
`B6
`B6
`C3H.SW
`B6
`B10.D2
`DBA/2
`BIO.BR
`Chronic GVHD Models
`B10.D2
`LP/J
`DBA/2
`B6
`BALB/c
`
`(B6 x DBA/2)Fl
`BALB/c
`B6
`bmI
`bm 12
`B6
`BALB/b
`DBA/2
`B10.D2
`
`CBA .
`
`BALB/c
`B6
`B6D2Fl
`(B6 x DBA/c)Fl
`BALB/c x A)Fl
`
`I, Il, mHAs
`I, Il, mHAs
`I, Il, mHAs
`I
`II
`mHAs
`mHAs
`mHAs
`mHAs
`mHAs
`
`mHAs
`mHAs
`I, Il, mHAs
`I, II, mHAs
`I, II, mHAs
`
`CD4 +/orCD8
`CD4 +/orCD8
`CD4 +/orCD8
`CD8
`CD4
`CD8
`CD4
`CD4
`CD8
`CD8
`
`CD4
`CD4
`CD4
`CD4
`CD4
`
`Table 1. Mouse models of BMT.
`Donor and host strains used in common BMT models, usual total body irradiation (TBI) doses (delivered)
`in two split doses on a single day at < 150 cGy/min), target GVHD antigens-MHC class I (I), or minor
`HA (mHA), T cell dependence of subsequent GVHD (CD4 and/or CD8). Source: Biol Blood Marrow
`Transplantation 14:129-135(2008) PMID SJ083-8791(07)00551-4.
`
`3. lmmunobiology
`
`It is helpful to remember two important principles when considering the pathophysiology of acute GVHD.
`First, acute GVHD reflects exaggerated, but normal inflammatory mechanisms that occur in a setting where they
`are undesirable. The donor lymphocytes that have been infused into the recipient function appropriately, given the
`foreign environment they encounter. Second, donor lymphocytes encounter tissues in the recipient that have often
`been profoundly damaged. The effects of the underlying disease, prior infections, and the intensity of conditioning
`regimen all result in substantial changes not only in the immune cells, but also in the endothelial and epithelial cells.
`Thus, the allogeneic donor cells rapidly encounter not only a foreign environment, but one that has been altered to
`promote the activation and proliferation of inflammatory cells. Thus, the pathophysiology of acute GVHD may be
`considered a distortion of the normal inflammatory cellular responses (Reddy and Ferrara 2003). The development
`and evolution of acute GVHD can be conceptualized in three sequential phases (see Figure 1) to provide a unified
`perspective on the complex cellular interactions and inflammatory cascades that lead to acute GVHD: (1) activation
`of the antigen-presenting cells (APCs; 2) donor T cell activation, differentiation and migration and (3) effector phase
`(Reddy and Ferrara 2003).
`
`3.1. Phase 1: Activation of Antigen Presenting Cells (APCs)
`
`The earliest phase of acute GVHD is set into motion by the profound damage caused by the underlying disease
`and its treatment or infections that might be further exacerbated by the BMT conditioning regimens of variable intensity
`which include total body irradiation (TBI and/or chemotherapy) that are administered even before the infusion of donor
`cells (Clift et al., 1990; Gale eta!., 1987; Hill and Ferrara, 2000; Paris et al., 2001; Xun et al., 1994). This first step results
`in activating the APCs. Specifically, damaged host tissues respond with multiple changes, including the secretion of
`proinflammatory cytokines, such as TNF-a and IL-1, described as the "cytokine storm" (Hill and Ferrara, 2000; Xun
`et al., 1994; Hill et al., 1997). Such changes increase expression of adhesion molecules, costimulatory molecules,
`MHC antigens and chemokines gradients that alert the residual host and the infused donor immune cells (Hill and
`Ferrara, 2000). These "danger signals" activate host APCs (Matzinger, 2002; Shlomchik et al., 1999). Damage to the
`gastrointestinal (GI) tract from the conditioning is particularly important in this process because it allows for systemic
`translocation of immuno-stimulatory microbial products such as lipopolysaccaride (LPS) that further enhance the
`activation of host APCs and the secondary lymphoid tissue in the GI tract is likely the initial site of interaction
`
`3
`
`stembook.org
`
`

`

`Mouse models of graft-versus-host disease
`
`GVHD
`pathology
`
`(I) Recipient c:ondltloning
`tissue damage
`
`Host
`
`IL-1
`IL-6
`
`Small
`intestine
`
`(II) Donor
`T cell activation
`
`Figure 1. T hree phases of GVHD immuno-biology.
`
`Target cell
`apoptosis
`
`{Ill) Effecto
`
`between activated APCs and donor T cells (Hill and Ferrara, 2000; Paris et al., 2001 ; Cooke et al. , 1998; Murai
`et al. , 2003). This scenario accords with the observation that an increased risk of GVHD is associated with intensive
`conditioning regimens that cause extensive injury to epithelial and endothelial surfaces with a subsequent release of
`inflammatory cytokines, and increases the expression of cell surface adhesion mol ecules (Hill and Ferrara, 2000; Paris
`et al. , 2001). The relationship among conditioning intensity, inflammatory cytokine, and GVHD severity has been
`supported by elegant murine studies (Paris et al. , 2001; Hill et al., 1997). Furthermore, the observations from these
`experimental studi es have led to two recent clinical innovations to reduce clinical acute GVHD: (a) reduced-intensity
`conditioning to decrease the damage to host ti ssues and, thus, limit activation of host APC and (b) KIR mismatches
`between donor and rec ipients to eliminate the host APCs by the alloreactive NK cells (Slavin, 2000; Velardi et al.,
`2002). However, reduced intensity conditioning also causes substantial GVHD. This suggests that in out-bred species
`that are exposed to infectious agents and in some parent into Fl mouse models , tissue stress and inflammation not
`caused by conditioning regimen are also sufficient to prime and induce a GVH response.
`
`Host type APCs that are present and have been primed by conditioning are critical for the induction of this
`phase; recent ev idence suggests that donor type APCs exacerbate GVHD, but in certain experimental models donor
`type APC chimeras also induce GVHD (Teshima et al. , 2002; Shlomchik et al., 1999; Jones et al. , 2003; Reddy et al.,
`2005). In clinical situations, if donor type APCs are present in sufficient quantity and have been appropriately primed,
`they too might play a role in the initiation and exacerbation of GVHD (Arpinati et al., 2000; Auffermann-Gretzinger
`et al. , 2002; MacDonald et al. , 2005 ). Amongst the cells with antigen-presenting capability, DCs are the most potent
`and play an important role in the induction of GVHD (Banchereau and Steinman, 1998). Experimental data suggest
`that GVHD can be regulated by qualitatively or quantitatively modulating distinct DC subsets (Chorny et al., 2006;
`Duffner et al. , 2004; Macdonald et al. , 2007; Paraiso et al. , 2007; Sato et al., 2003). In one clinical study persistence
`of host DC after day 100 correlated with the severity of acute GVHD while elimination of host DCs was associated
`with reduced severity of acute GVHD (Auffermann-Gretzinger et al. , 2002). The allo-stimulatory capacity of mature
`monocyte derived DCs (mDCs) after reduced-intensity transplants was lower for up to six months compared to the
`mDCs from myeloablative transplant recipients, thu s suggesting a role for host DCs and the reduction in "danger
`signals" secondary to less intense conditioning in acute GVHD (Nachbaur et al. , 2003). Nonetheless thi s concept of
`
`4
`
`stembook.org
`
`

`

`Mouse models of graft-versus-host disease
`
`enhanced host APC activation explains a number of clinical observations, such as increased risks of acute GVHD
`associated with advanced stage malignancy, conditioning intensity and histories of viral infections. This has been
`further suggested by recent NOD2, MBL and TLR4 polymorphism studies in humans (Holler, 2006; Rocha, 2002;
`Lorenz et al., 2001).
`
`Other professional APCs such as monocytes/macrophages or semi-professional APCs might also play a role
`in this phase. For example, recent data suggests that host type B cells might play a regulatory role under certain
`contexts (Rowe, 2006). Also host or donor type nonhematopoietic stem cells, such as mesenchymal stem cells or
`stromal cells when acting as APCs have been shown to reduce T cell allogeneic responses, although the mechanism
`for such inhibition remains unclear. The relative contributions of various APCs, professional or otherwise, remain to
`be elucidated.
`
`The other aspects of the innate immune system such as complement activation, PMNs, and defensins remain
`poorly understood and they too might play a role in enhancing or regulating the induction and propagation of GVHD.
`In this regard, a recent study suggests that target tissue inflammation might account for the unique organ specificity of
`acute GVHD (Chakraverty, 2006).
`
`3.2. Phase 2: Donor T cell activation, differentiation and migration
`
`The infused donor T cells interact with the primed APCs and initiate the second phase of acute GVHD. This
`phase includes antigen presentation by primed APCs, the subsequent activation, proliferation, differentiation and
`migration of alloreactive donor T cells.
`
`After allogeneic HSC transplants, both host- and donor-derived APCs are present in secondary lymphoid organs
`(Beilhack et al., 2005; Korngold and Sprent, 1980). The T cell receptor (TCR) of the donor T cells can recognize
`alloantigens either on host APCs (direct presentation) or donor APCs (indirect presentation; Lechler et al., 2001;
`Shlomchik, 2003). In direct presentation, donor T cells recognize either the peptide bound to allogeneic MHC
`molecules or allogeneic MHC molecules without peptide (Lechler et al., 2001; Sayegh and Carpenter, 1996). During
`indirect presentation, T cells respond to the peptide generated by degradation of the allogeneic MHC molecules
`presented on self-MHC (Sayegh and Carpenter, 1996). An experimental study demonstrated that APCs derived from
`the host, rather than from the donor, are critical in inducing GVHD across MiHA mismatch (Shlomchik, 2003). Recent
`data suggest that presenting distinct target antigens by the host and donor type APCs might play a differential role in
`mediating target organ damage (Shlomchik, 2003; Anderson et al., 2005; Kaplan et al., 2004). In humans, most cases
`of acute GVHD developed when both host DCs and donor dendritic cells (DCs) were present in peripheral blood after
`BMT (Auffermann-Gretzinger et al., 2002).
`
`3.2. 1. Costimulation
`
`The interaction of donor lymphocyte TCR with the host allo-peptide presented on the MHC of APCs alone is insufficient
`to induce T cell activation (Appleman and Boussiotis, 2003). Both TCR ligation and costimulation via a "second"
`signal through interaction between the T cell costimulatory molecules and their ligands on APCs are required to achieve
`T proliferation, differentiation and survival (Sharpe and Freeman, 2002). The danger signals generated in phase 1
`augment these interactions and significant progress has been made on the nature and impact of these "second" signals
`(Bromley et al., 2001; Dustin, 2001). Costimulatory pathways are now known to deliver both positive and negative
`signals and molecules from two major families; the B7 family and the TNF receptor (TNFR) family play pivotal
`roles in GVHD (Greenwald et al., 2005). Interrupting the second signal by blockade of various positive costimulatory
`molecules (CD28, ICOS, CD40, CD30, 4-lBB and OX40) reduces acute GVHD in several murine models while
`antagonism of the inhibitory signals (PD-1 and CTLA-4) exacerbates the severity of acute GVHD (Welniak et al.,
`2007; Blazar et al., 1994; Blazar et al., 1995; Blazar et al., 1997; Blazar et al., 2001; Blazar et al., 2003; Blazar et
`al., 2003). The various T cell and APC costimulatory molecules and the impact on acute GVHD are summarized in
`Table 2. The specific context and the hierarchy in which each of these signals play a dominant role in the modulation
`of GVHD remain to be determined.
`
`3.2.2. T cell subsets
`
`T cells consist of several subsets whose responses differ based on antigenic stimuli, activation thresholds and effector
`functions. The alloantigen composition of the host determines which donor T cell subsets proliferate and differentiate.
`
`5
`
`stembook.org
`
`

`

`Mouse models of graft-versus-host disease
`
`T cell costimulation
`
`Tcell
`
`APC
`
`Adhesion
`
`Recognition
`
`Costimulation
`
`ICAMs
`LEA-1
`CD2 (LEA-2)
`TCR/CD4
`TCR/CD8
`CD28
`CD152 (CTLA-4)
`ICOS
`PD-1
`Unknown
`CD 154 (CD4OL)
`CD134 (0X40)
`CD137 (4-IBB
`HVEM
`
`LEA-1
`ICAM~
`LFA-3
`NIIIC hi
`Mi-Ice 1
`CD80/86
`CD80/86
`B7H/B7RP-1
`PD-Ll, PD-L2
`B7-H3
`CD40
`CD13L(OX40L)
`CD137L (4-lIBBL
`LIGHT
`
`Table 2. T-cell-APC Interactions that Regulate GVHD.
`
`• CD4+ and CD8+ cells
`
`CD4 and CD8 proteins are coreceptors for constant portions of MHC class II and class I molecules, respectively
`(Csencsits and Bishop, 2003). Therefore, MHC class I (HLA-A, -B, -C) differences stimulate CD8+ T cells and MHC
`class II (HLA-DR -DP, -DQ) differences stimulate CD4+T cells (Csencsits and Bishop, 2003; Ferrara et al., 1993;
`Komgold and Sprent, 1982; Komgold and Sprent, 1985; Komgold and Sprent, 1987). But clinical trials of CD4+ or
`CDS+ depletion have been inconclusive (Wu and Ritz, 2006). This may not be surprising because GVHD is induced
`by MiHAs in the majority of HLA-identical BMT, which are peptides derived from polymorphic cellular proteins that
`are presented by MHC molecules (Goulmy, 2006). Because the manner of protein processing depends on genes of the
`MHC, two siblings will have many different peptides in the MHC groove (Goulmy, 2006). Thus, in the majority of
`HLA-identical BMT, acute GVHD may be induced by either or both CD4+ and CDS+ subsets in response to minor
`histocompatibility antigens (Wu and Ritz, 2006). The peptide repertoire for class I or class II MHC remains unknown
`and likely to be distinct between one individual to the next (Spierings et al., 2006). But it is plausible that only a few
`of these many peptides might behave as immunodominant "major minor" antigens that can potentially induce GVHD.
`In any event, such antigens remain to be identified and validated in large patient populations.
`
`Central deletion by establishment of stable mixed hematopoietic chimeric state is an effective way to eliminate
`continued thymic production of both CD4+ and CDS+ alloreactive T cells and thus reduce GVHD (Sykes, 2001;
`Wekerle et al., 1998; Wekerle et al., 2000). In contrast peripheral mechanisms to induce tolerance of CD4+ and CDS+
`Tcells appears to be distinct (Wells et al., 1999; Wells et al., 2001). The T cell apoptosis pathways by which peripheral
`deletion occurs can be broadly categorized into activation-induced cell death (AICD) and passive cell death (PCD;
`Lechler et al., 2003). Experimental data suggests that deletional tolerance by AICD is operative via the Fas (for CD4+)
`or TNFR (CDS+) pathways in Thl cells and when there is a higher frequency of alloreactive cells (Combadiere et al.,
`1998; Min et al., 2004; Siegel et al., 2000; Zhang et al., 1997; Zheng et al., 1995). PCD or "death by neglect" is due to
`rapid downregulation of Bcl-2 and appears to be critical in non-irradiated, but not after irradiated BMT (Drobyski et
`al., 2002). Thus, distinct mechanisms of tolerance induced by apoptosis have a dominant role depending on the T cell
`subsets, the conditioning regimens and the histocompatibility differences. Nonetheless strategies aimed at selective
`elimination of donor T cells in vivo after HCT, either by targeting a suicide gene to the allo-T cells or by photodynamic
`cell purging appear promising in reducing experimental acute GVHD (Bondanza et al., 2006; Bonini et al., 1997;
`Bordignon et al., 1995; Chen et al., 2002; Chen et al., 2002; Drobyski and Gendelman, 2002).
`
`• Na'ive and Memory Subsets
`
`Several independent groups have intriguingly found that, unlike memory (CD62L -) T cells, the na'ive (CD62L +)
`T cells were alloreactive and caused acute GVHD across different donor/recipient strain combinations (Anderson et
`al., 2003; Chen et al., 2004; Maeda et al., 2007; Zhang et al., 2004). Furthermore, expression of the na'ive T cell
`
`6
`
`stembook.org
`
`

`

`Mouse models of graft-versus-host disease
`
`marker CD62L was also found to be critical to regulate GVHD by donor natural regulatory T cells (Ermann et al.,
`2005; Taylor et al., 2004). By contrast, another recent study demonstrated that alloreactive memory T cells and their
`precursor cells (memory stem cells) sustain and also cause robust GVHD (Zhang et al., 2005; Zhang et al., 2005).
`
`• Regulatory T cells
`
`Recent advances indicate that distinct subsets of regulatory CD4+co25+, CD4+co25-1uo+ Tr cells, yoT
`cells, DN- T cells, NK T cells and regulatory DCs control immune responses by inducing anergy or active suppression
`of alloreactive T cells (Sato et al., 2003; Blazar and Taylor, 2005; Cohen et al., 2002; Cohen and Boyer, 2006; Hoffmann
`et al., 2002; Johnson et al., 2002; Lowsky et al., 2005; Maeda et al., 2005; Roncarolo, 1997; Young et al., 2003; Zeng
`et al., 1999; Zhang et al., 2004). Several studies have demonstrated a critical role for the natural donor co4+co25+
`Foxp3+ regulatory T (Treg) cells, obtained from naive animals or generated ex-vivo, in the outcome of acute GVHD.
`Donor CD4+co25+ T cells suppressed the early expansion of alloreactive donor T cells and their capacity to induce
`acute GVHD without abrogating GVL effector function against these tumors (Edinger et al., 2003; Nguyen et al.,
`2007). co4+co25+ T cells induced/generated by immature or regulatory host type DCs and by regulatory donor
`type myeloid APCs were also able to suppress acute GVHD (Sato et al., 2003). One of the clinical studies that
`evaluated the relationship between donor CD4+co25+ cells and acute GVHD in humans after matched sibling donor
`grafts and found that, in contrast to the murine studies, donor grafts containing larger numbers of CD4+ CD25+T
`cells developed more severe acute GVHD (Stanzani et al., 2003). These data suggest that coexpression of CD4+ and
`CD25+ is insufficient because an increase in CD25+ T cells in donor grafts is associated with greater risks of acute
`GVHD after clinical HCT. Another recent study found that Foxp3 mRNA expression (considered a specific marker
`for naturally occurring CD4+CD25+Tregs) was significantly decreased in peripheral blood mononuclear cells from
`patients with acute GVHD (Miura et al., 2004; Zorn et al., 2005). But Foxp3 expression in humans, unlike mice, may
`not be specific for T cells with a regulatory phenotype (Ziegler, 2006). It is likely that the precise role of regulatory T
`cells in clinical acute GVHD will, therefore, not only depend upon identifying specific molecular markers in addition
`to Foxp3, but also on the ability for ex vivo expansion of these cells in sufficient numbers. Several clinical trials are
`underway in the United States and Europe to substantially expand these cells ex vivo and use for prevention of GVHD.
`
`Host NKl .1 + T cells are another T cell subset with suppressive functions that have also been shown to suppress
`acute GVHD in an IL-4 dependent manner. (Lowsky et al., 2005; Zeng et al., 1999; Hashimoto et al., 2005) By contrast,
`donor NKT cells were found to reduce GVHD (Morris et al., 2005; Morris et al., 2006) and enhance perforin mediated
`GVL in an IFN-y dependent manner. Recent clinical data suggests that enhancing recipient NKT cells by repeated TLI
`conditioning promoted Th2 polarization and dramatically reduced GVHD (Lowsky et al., 2005). Experimental data
`also show that activated donor NK cells can reduce GVHD through the elimination of host APCs or by secretion of
`transforming growth factor-,B (TGF-,B; Morris et al., 2006). A murine BMT study using mice lacking SH2-containing
`inositol phosphatase (SHIP), in which the NK compartment is dominated by cells that express two inhibitory receptors
`capable of binding either self or allogeneic MHC ligands, suggests that host NK cells may play a role in the initiation
`of GVHD (Wang et al., 2002).
`
`3.2.3. Cytokines and T cell differentiation
`
`APC and T cell activation result in rapid intracellular biochemical cascades that induce transcription of many genes
`including cytokines and their receptors. The Th 1 cytokines (IFN-y, IL-2 and TNF-a) have been implicated in the patho(cid:173)
`physiology of acute GVHD (Antin and Ferrara, 1992; Ferrara and Krenger, 1998; Liu et al., 2007; Ratanatharathom et
`al., 1998; Reddy, 2003). IL-2 production by donor T cells remains the main target of many current clinical therapeutic
`and prophylactic approaches, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) against the IL-2 and
`its receptor to control acute GVHD (Ferrara and Krenger, 1998; Liu et al., 2007; Ratanatharathom et al., 1998; Reddy,
`2003; Ferrara, 1994). But emerging data indicate an important role for IL-2 in the generation and maintenance of
`co4+co25+ Foxp3+ Tregs, suggesting that prolonged interference with IL-2 may have an unintended consequence
`of preventing the development of long-term tolerance after allogeneic HCT (Gavin et al., 2007; Liston and Rudensky,
`2007; Zeiser et al., 2006; Zhang et al., 2005).
`
`Similarly the role of other Thi cytokines IFN-y or their inducers as regulators or inducers of GVHD severity
`depends on the degree of allo-mismatch, the intensity of conditioning and the T cell subsets that are involved after
`BMT (Reddy, 2004; Sykes et al., 1999; Yang et al., 1997). Thus, although the "cytokine storm" initiated in phase 1
`and amplified by the Thi cytokines correlates with the development of acute GVHD, early Thi polarization of donor
`T cells to HCT recipients can attenuate acute GVHD suggesting that physiological and adequate amounts of Thi
`
`7
`
`stembook.org
`
`

`

`Mouse models of graft-versus-host disease
`
`cytokines are critical for GVHD induction, while inadequate production (extremely low or high) could modulate acute
`GVHD through a breakdown of negative feedback mechanisms for activated donor T cells (Reddy, 2003; Reddy, 2004;
`Reddy et al., 2001; Sykes et al., 1995). Several different cytokines that polarize donor T cells to Th2 such as IL-4,
`G-CSF, IL-18, IL-11, rapamycin and the secretion of IL-4 by NKl .1 + T cells can reduce acute GVHD (Fowler et al.,
`1994; Fowler and Gress, 2000; Hill et al., 1998; Jung et al., 2006; Krenger et al., 1995; Pan et al., 1995; Reddy et al.,
`2003). But Thi and Th2 subsets cause injury of distinct acute GVHD target tissues, and some studies failed to show
`a beneficial effect of Th2 polarization on acute GVHD (Nikolic et al., 2000). Thus the Thl/Th2 paradigm of donor
`T cells in the immuno-pathogenesis of acute GVHD has evolved over the last few years and its causal role in acute
`GVHD is complex and incompletely understood.
`
`IL-10 plays a key role in suppressing immune responses and its role in regulating experimental acute GVHD
`is unclear (Blazar et al., 1998). Recent clinical data demonstrate an unequivocal association between IL-10 polymor(cid:173)
`phisms and the severity of acute GVHD (Lin et al., 2003). TGF-,B, another suppressive cytokine, was shown to suppress
`acute GVHD, but to exacerbate chronic GVHD (Banovic et al., 2005). The roles of some other cytokines, such as
`IL-7 (that promotes immune reconstitution) and IL-13, remain unclear (Alpdogan et al., 2001; Alpdogan et al., 2003;
`Gendelman et al., 2004; Sinha et al., 2002). The role for Th 17 cells, a recently described novel T cell differentiation
`in many immunological processes, is not yet known (Weaver and Murphy, 2007). In any

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