throbber
KJH
`THE KOREAN JOURNAL OF HEMATOLOGY
`~
`
`V O L U M E 4 6 (cid:564) N U M B E R 2 (cid:564) J u n e 2 0 1 1
`
`R E V I E W A R T I C L E
`
`The pathophysiology of chronic graft-versus-host disease: the
`unveiling of an enigma
`Chang-Ki Min
`Division of Hematology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
`
`p-ISSN 1738-7949 / e-ISSN 2092-9129
`DOI: 10.5045/kjh.2011.46.2.80
`Korean J Hematol 2011;46:80-7.
`
`Received on June 9, 2011
`Accepted on June 10, 2011
`
`*This study was supported by the Korea
`Healthcare Technology R&D Project,
`Ministry of Health, Welfare & Family
`Affairs, Republic of Korea (grant no.
`A092258).
`
`Correspondence to
`Chang-Ki Min, M.D.
`Department of Internal Medicine, Seoul St.
`Mary’s Hospital, The Catholic University
`of Korea, 505 Banpo-dong, Seocho-gu,
`Seoul 137-701, Korea
`Tel: (cid:1024)82-2-2258-6053
`Fax: (cid:1024)82-2-599-3589
`E-mail: ckmin@catholic.ac.kr
`(cid:59926) 2011 Korean Society of Hematology
`
`Chronic graft-versus-host disease (CGVHD) is one of the most significant complications
`of long-term survivors after allogeneic hematopoietic stem cell transplantation
`(allo-HSCT). CGVHD may have protean manifestations and can pose unique diagnostic
`and therapeutic challenges. New recommendations that emphasize the importance of
`qualitative differences, as opposed to time of onset after HSCT, are now being used to
`standardize the diagnosis and clinical assessment of CGVHD, but they require validation.
`During the past 3 decades, experimental studies and clinical observations have elucidated
`the mechanisms of acute GVHD, but its biology is much less well-understood.
`Experimental studies have generated at least 4 theories to explain the pathophysiology
`of CGVHD: (1) thymic damage and the defective negative selection of T cells, (2) regu-
`latory T cell deficiencies, (3) auto-antibody production by aberrant B cells, and (4) the
`formation of profibrotic lesions. Mouse models have provided important insights into
`the pathophysiology of CGVHD, and these have helped improve clinical outcomes follow-
`ing allo-HSCT, but no animal model fully replicates all of the features of CGVHD in humans.
`In this article, recent clinical changes, the pathogenesis of CGHVD, the cellular and cyto-
`kine networks implicated in its pathogenesis, and the animal models used to devise strat-
`egies to prevent and treat CGVHD are reviewed.
`
`Key Words Chronic graft-versus-host disease, Pathophysiology, Acute graft-versus-
`host disease, Fibrosis, Mouse model
`
`INTRODUCTION
`
` Chronic graft-versus-host disease (CGVHD) remains a ma-
`jor cause of late morbidity and mortality after allogeneic
`hematopoietic stem cell transplantation (allo-HSCT) [1]. The
`incidence of CGVHD following allo-HSCT ranges from 25%
`to 80%; the occurrence of CGVHD is associated with immune
`dysfunction and thus, a risk of infection and reduced quality
`of life [2], even though CGVHD is also associated with a
`lower relapse rate, presumably because of graft-versus-leuke-
`mia effects [3]. Over the past 10 years, CGVHD has emerged
`as the most troublesome complication of allo-HSCT. Impro-
`vements in human histocompatibility antigen (HLA) typing
`for unrelated transplantation, the adoption of new acute
`GVHD prophylaxis measures, reductions in conditioning reg-
`imen intensity, the introduction of new antimicrobial agents,
`and advances in supportive care have all helped to mitigate
`early morbidity and mortality in patients after allo-HSCT.
`However, because more and more patients survive the early
`post-transplant period, the number of individuals at risk
`• •o
`This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0)
`which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`for CGVHD continues to grow. This trend, in conjunction
`with the escalating use of mobilized peripheral blood cells
`as a preferred stem cell source, has led to a significant increase
`in the number of transplant survivors living with, and in
`some cases dying from, CGVHD [3-8]. Unfortunately, the
`treatment of established CGVHD remains unsatisfactory.
`Corticosteroids are the mainstay of therapy, but are often
`not fully effective, and their long-term use leads to multiple
`complications [1, 9]. Other agents such as calcineurin in-
`hibitors, sirolimus, mycophenolate mofetil, thalidomide,
`pentostatin, mesenchymal stem cells, and extracorporeal
`photopheresis have all produced responses in phase 2 studies,
`but no agent has yet demonstrated superiority to steroids
`alone in a randomized clinical trial [10-19].
`During the past 3 decades, experimental studies and clin-
`ical observations have elucidated the pathophysiology of
`acute GVHD, but the biology of CGVHD has not been deter-
`mined. In particular, defining the pathophysiology of CGVHD
`has been complicated by the absence of animal models that
`accurately recapitulate the disease or its clinical setting; this
`is in contrast to acute GVHD, in which murine models of
`
`

`

`Pathogenesis of chronic GVHD
`
`81
`
`major histocompatibility (MHC) mismatched HSCT provide
`a reasonably comprehensive picture of its pathophysiology
`as a clinical disease [20]. The purpose of this review is to
`briefly describe the pathophysiology of CGVHD, on the basis
`of phenotype and immunologic mechanisms that encompass
`the majority of murine CGVHD models described to date.
`Their relevance to clinical CGVHD is also discussed.
`
`DEFINITION
`
`CGVHD was initially defined as a GVHD syndrome that
`presents more than 100 days post-transplant, either as an
`extension of acute GVHD (progressive onset CGVHD), after
`a disease-free interval (quiescent CGVHD), or without pre-
`ceding acute GVHD (de novo CGVHD) [21]. In patients
`with CGVHD, the skin can exhibit erythema with macules
`and plaques, desquamation, dyspigmentation, lichen planus,
`atrophy, and in severe cases, chronic ulcers. Chronic choles-
`tatic liver disease can develop, as can involvement of the
`gastrointestinal tract, which may result in weight loss and
`malnutrition. CGVHD commonly produces sicca syndrome,
`which is caused by lymphocytic destruction of exocrine
`glands, most frequently affecting the eyes and mouth. The
`pathologic findings of CGVHD in the immune system include
`involution of thymic epithelium, lymphocyte depletion, and
`absence of secondary germinal centers in lymph nodes [22].
`The skin pathology shows epidermal atrophy, dermal fibrosis,
`and sclerosis. Gastrointestinal lesions include inflammation
`and rarely, stenosis and stricture formation, particularly in
`the esophagus. Positive histological findings in the liver are
`often intensified versions of acute GVHD and include chronic
`changes, such as fibrosis, the hyalinization of portal triads,
`and bile duct obliteration. The glands of the skin and digestive
`tract show destruction of centrally draining ducts and secon-
`dary involvement of alveolar components. Pulmonary tissue
`can also be involved, although histological distinctions from
`bacterial and viral infections are sometimes difficult. Never-
`theless, bronchiolitis obliterans, similar to that observed dur-
`ing lung transplant rejection, is now generally considered
`a manifestation of CGVHD.
`
`IMPORTANT CHANGES IN CLINICAL
`CONSIDERATIONS
`
`It is apparent that the clinical and histological changes
`considered characteristic of CGVHD can develop as early
`as 40 or 50 days post-transplant and thus, overlap with those
`of acute GVHD. Hence, the time of onset is increasingly
`becoming an arbitrary criterion, and it has become more
`meaningful to define the disease on the basis of clinical,
`histological, and immunologic findings.
`The National Institutes of Health (NIH) have proposed
`new consensus criteria for the diagnosis and clinical assess-
`ment of CGVHD, which emphasize the manifestations of
`GVHD and not time of onset after allo-HSCT (day 100)
`
`[23]. This proposal involves 2 categories for GVHD (acute
`or chronic), each with 2 subcategories (classic acute and
`late acute or classic chronic and overlap syndrome). In addi-
`tion, a new scoring system is proposed to describe the extent
`and severity of CGVHD at each organ or site at any given
`time and that takes functional impact into account. The
`global composite scores produced and the numbers of organs
`or sites involved have been proposed as a means of assessing
`CGVHD severity, and it is expected that this system will
`replace the old grading system (limited versus extensive
`types). The feasibility of the NIH consensus criteria has been
`examined by us and others, and all studies have demonstrated
`the applicability of the new NIH criteria and described possi-
`ble roles for the new global scoring system in the assessment
`of CGVHD severity [24-26].
`
`PATHOPHYSIOLOGY OF CGVHD
`
`Acute GVHD resembles a toxic, sepsis-like syndrome. Host
`antigen-presenting cells (APCs), especially dendritic cells
`(DCs), present alloantigens to incoming alloreactive cyto-
`toxic T cells, and the subsequent actions of these T cells
`result in tissue damage to the epidermis, hepatic bile ducts,
`and gut epithelium. This process is amplified by cytokine
`release from damaged tissues and the ingress of lipopoly-
`saccharide and other pathogen-associated molecular entities
`through damaged gut mucosa, which in turn up-regulate
`the innate immune system [27]. Not surprisingly, the immune
`mechanisms implicated in the induction and propagation
`of CGVHD differ from those of acute GVHD. However,
`the pathophysiology of CGVHD, unlike that of acute GVHD,
`remains obscure, as do its effective prevention and treatment.
`Fundamental research for the pathophysiology of CGVHD
`is required to develop more effective prophylactic and treat-
`ment regimens. So far, at least 4 theories have been generated
`to explain how CGVHD develops (Fig. 1).
`
`1. Breakage of immune tolerance to self-antigens (central tol-
`erance)
`It has been suggested that immune tolerance to self-anti-
`gens is disrupted in CGVHD, and that these give rise to
`the autoimmune manifestations of the disorder. One attrac-
`tive hypothesis is that thymic epithelial damage caused by
`conditioning regimens and/or acute GVHD leads to dysregu-
`lation of central tolerance mechanisms during the recon-
`stitution of the immune system post-transplantation [28].
`CD4+ T cells generated de novo from donor stem cells appear
`to mediate the evolution of CGVHD from acute GVHD [29].
`In fact, CGVHD occurs, even though it may not be preceded
`by acute GVHD. In healthy individuals, 95-99% of dou-
`ble-positive CD4+ CD8+ immature T cells in the cortex of
`the thymus die through apoptosis, having failed to receive
`survival signals through their T cell receptors, a process
`referred to as “death by neglect.” Those cells that do bind
`with low affinity to MHC class I or class II upregulate CD8
`or CD4 respectively (positive selection) and survive. Since
`
`Korean J Hematol 2011;46:80-7.
`
`

`

`82
`
`Chang-Ki Min
`
`Donor
`
`Recipient
`
`Acute GVHD
`
`Bcell
`
`a
`
`~ •
`
`a
`
`. ,.
`z--iS'I
`z
`r~--
`
`"
`
`Deficiency of regulatory T cells
`
`Thymic
`dysfunction
`
`B-cell
`activation
`
`Fibrosis
`
`Cytokines
`
`Organ faliure
`
`Fig. 1. The pathophysiology of chronic graft-versus-host disease.
`
`self-antigens are also presented in the context of the cortical
`epithelial MHC complex, such T cells will have low self
`affinity. Furthermore, in the thymic medulla, single-positive
`T cells will encounter marrow-derived APCs also bearing
`self-antigens (sequestered from the blood) and if strongly
`autoreactive, will die by apoptosis (negative selection).
`Through these processes, low-affinity, self-reacting naïve
`T cells will enter the periphery, and when they encounter
`the same self-antigen/MHC complex, will receive survival
`but not activation signals. This balance between negative
`and positive selections may be lost in a pro-inflammatory
`environment and high tissue-specific autoantigen load. In
`this inflammatory environment, peripheral tolerance mecha-
`nisms would be critical for regulating GVHD.
`In the setting of CGVHD, central tolerance failure could
`lead to an immune disease state resembling autoimmune
`disease. Although strategies based on the administration of
`keratinocyte growth factor at the time of transplant to pre-
`vent injury or repair thymic epithelium have been successful
`in experimental models [30], they failed in clinical trials
`[31]. Zhang found that host thymus is not required for the
`induction of CGVHD and that quiescent autoreactive T and
`
`B cells in transplants from non-autoimmune donors might
`be activated and expanded to cause CGVHD [32]. In addition,
`Imado found that transfection of the hepatocyte growth fac-
`tor (HGF) gene in vivo prevented the development of CGVHD
`in a murine model [33]. HGF also protected against thymic
`injury caused by acute GVHD and thus, prevented the gen-
`eration of host-reactive T cells and the development of
`CGVHD.
`
`2. CD4+CD25+ regulatory T cells (Tregs) and their relation-
`ship to CGVHD
`In several series, regulatory T cell (Treg) numbers (deter-
`mined by CD4+CD25+FoxP3+ staining) have been reported
`to be diminished in CGVHD [34-36], although data reported
`on Treg numbers and the occurrence of CGVHD are con-
`tradictory. Clark found that CGVHD is associated with ele-
`vated numbers of peripheral blood Tregs and that these num-
`bers returned to normal in patients with resolved CGVHD,
`thus indicating that CGVHD injury is not the result of Treg
`deficiency [37]. The mechanism by which Tregs suppress
`CGVHD remains uncertain, but there is evidence that sup-
`pression is mediated by cytokines, such as transforming
`
`Korean J Hematol 2011;46:80-7.
`
`

`

`Pathogenesis of chronic GVHD
`
`83
`
`growth factor (TGF)-(cid:69) and interleukin (IL)-10, or by contact
`with plasmacytoid DCs through indoleamine 2,3-dioxyge-
`nase [38]. Tregs may also exert an inhibitory influence di-
`rectly in target tissues [39]. For example, mucosal Treg num-
`bers have been documented to be lower in patients with
`GVHD than in normal controls or patients without GVHD
`[34]. Interestingly, extracorporeal photochemotherapy in-
`creases levels of circulating functional Tregs in CGVHD pa-
`tients [40], and recently, a novel photodepleting approach
`was found to both preserve and expand Treg numbers while
`selectively eliminating CD4+ effector T cells from patients
`with CGVHD [41].
`The adoptive transfer of Tregs in animal models of GVHD
`has demonstrated their efficacy, which suggests that Tregs
`can be exploited in the clinical setting [42]. Giorgini con-
`cluded that alloantigen-driven expansion, rather than ho-
`meostatic proliferation, is critical for the effectiveness of
`Tregs in CGVHD, and suggested that cellular therapy with
`alloantigen-induced Tregs in combination with glucocorti-
`coids could prevent CGVHD after immune reconstitution
`[43]. Zhang, using a murine study, suggested that peripheral
`tolerance may be more critical and abrogated by donor Tregs
`[32], and Chen associated the absence of Treg control of
`T helper (Th) 1 and Th17 cells with an autoimmune-mediated
`pathology in CGVHD [44].
`
`3. The roles of B cells and the antibodies they produce
`Historically, research into the prevention and treatment
`of GVHD centered on donor T lymphocytes and strategies
`designed to suppress or deplete these cells. The roles of
`B lymphocytes in the pathogenesis of GVHD were high-
`lighted by a case report of a patient with CGVHD who
`responded to B cell depletion therapy based on rituximab
`[45]. Considerable laboratory evidence has since revealed
`complex interactions between B and T cells that culminate
`in CGVHD. There are numerous examples of autoantibody
`formation in patients with CGVHD, but the role of autoanti-
`body formation in its pathogenesis has not been elucidated
`[46]. One study, in which antibodies to platelet-derived
`growth factor (PDGF) were observed in patients with
`CGVHD but not in those without CGVHD, was of particular
`note [47]. These antibodies were found to have the capacity
`to induce both tyrosine phosphorylation of the PDGF re-
`ceptor and type I collagen gene expression in fibroblasts.
`The role of B cell activity in CGVHD is underscored by
`the observation of high plasma levels of B cell activating
`factor (BAFF), a cytokine that appears to drive B cell auto-
`immunity, in patients with CGVHD [48]. In fact, high plasma
`levels of BAFF at 6 months post-transplantation were found
`to predict the subsequent development of CGVHD in asymp-
`tomatic patients. The development of antibodies to minor
`histocompatibility antigens (mHA) encoded on the Y chro-
`mosome in male patients receiving female grafts has been
`strongly associated with CGVHD incidence [49]. Since this
`was originally observed [45], several clinical trials and case
`series have been conducted on the use of B cell depletion
`using rituximab to treat CGVHD. The evidence obtained
`
`supports the roles of B cells and antibodies in CGVHD and
`prompted trials of rituximab. In phase 2 trials, responses
`were documented in over 50% of subjects [50]. Recently,
`Korean researchers performed a definitive trial in an attempt
`to establish the efficacy of rituximab and concluded that
`B cells represent a promising target for the prevention and
`treatment of CGVHD [51]. Although studies on B cell deple-
`tion in CGVHD have demonstrated the clinical effectiveness
`of this strategy, the mechanisms underlying the exact role
`of B cells on CGVHD are not entirely clear. In a murine
`study on the topic, it was suggested that donor B cell depletion
`protected mice from CGVHD [32], and therefore, it is con-
`ceivable that alloreactive donor CD4+ T cells could be acti-
`vated by host B cells, and that this, in turn, promotes the
`activation and expansion of quiescent autoreactive donor
`B cells in stem cell grafts. Furthermore, these autoreactive
`B cells could have a central role in amplification of auto-
`immune responses and in the epitope spreading of autor-
`eactive T and B cells [52]. Another model of lupus CGVHD
`showed that the cytotoxic T lymphocyte (CTL)- promoting
`properties of CD40 stimulation outweigh CD4+ T cell-driven
`B cell hyperactivity [53].
`
`4. Fibrotic changes
`In the skin, the initial phase of CGVHD is characterized
`by an intense mononuclear inflammatory infiltrate and de-
`structive changes at the dermal-epidermal junction, accom-
`panied by irregular acanthosis, hyperkeratosis, atrophy, pro-
`gressing to dermal fibrosis and sclerosis [54]. Other hallmarks
`of CGVHD include the destruction of tubuloalveolar glands,
`ducts in the skin, salivary and lacrimal glands, respiratory
`epithelium, and bile ducts. A large number of experimental
`models have indicated an association between type 2 polar-
`ized immune responses and the development of fibrosis [55].
`In particular, donor type 2 immune responses were found
`to be required for the induction of cutaneous GVHD in
`mice [56]. Furthermore, Hillebrandt found that complement
`factor 5 (C5) dose-dependently modified liver fibrosis in
`mice and humans [57]. C5b-9 complexes are deposited in
`the skin, liver, lung, and kidney in mice with GVHD [58].
`C3 is deposited at the dermal-epidermal junction in humans
`with CGVHD [59], but deposition of C5b-9 complexes has
`not been described in man. One study found that serum
`levels of TGF-(cid:69) were higher in patients with CGVHD than
`in patients without CGVHD [60]. The interpretation of this
`result is complicated because assays were carried out with
`serum and not plasma, and serum contains large amounts
`of TGF-(cid:69) released by platelets during clotting. Gene ex-
`pression studies have demonstrated that increased TGF-(cid:69)
`signaling in CD4 cells and CD8 cells is associated with a
`reduced risk of CGVHD in man [61]. The association between
`increased TGF-(cid:69) activity and a reduced risk of CGVHD might
`result from a lower risk of acute GVHD, since acute GVHD
`is a well-recognized risk factor of CGVHD. Furthermore,
`skin fibrosis and the upregulation of TGF-(cid:69)1 and collagen
`mRNAs commonly occur in human scleroderma and murine
`sclerodermatous GVHD following transplantation of B10.D2
`
`Korean J Hematol 2011;46:80-7.
`
`

`

`84
`
`Chang-Ki Min
`
`lymphoid cells into irradiated BALB/c recipients [62].
`There is now considerable evidence that the preferential
`expansion of Th2 cells after allo-HSCT is associated with
`the development of CGVHD in both murine models and
`humans [63-68]. As is shown by most experimental models
`of fibrosis, CD4+ T cells play an important role in the pro-
`gression of CGVHD, and the type of CD4+ T-cell response
`that develops is crucial. Studies using various cytokine-defi-
`cient mice have shown that fibrogenesis is strongly linked
`with the development of a Th2 CD4+ T-cell response and
`that this involves IL-4, IL-5, and IL-13 [69]. Although an
`equally potent inflammatory response develops when Th1
`CD4+ T cells, which produce interferon (IFN)-(cid:74), dominate
`[70], under these circumstances, the development of tissue
`fibrosis is almost completely attenuated. These studies show
`that chronic inflammation does not always induce the deposi-
`tion of connective-tissue elements and that the magnitude
`of fibrosis is tightly regulated by the phenotype of the devel-
`oping Th-cell response. Furthermore, IL-13 and IL-4 bind
`to the same signaling receptor (IL-4R(cid:68)-IL-13R(cid:68)1) on fibro-
`blasts [71]. Indeed, studies carried out using several fibroblast
`subtypes have demonstrated the potent collagen-inducing
`activities of IL-4 and IL-13 [72-74]. In addition, when the
`productions of IL-4 and IL-13 from fibroblasts are compared,
`the concentration of IL-13 often exceeds that of IL-4 by
`a factor of 10-100. This suggests that IL-13 uses a signaling
`pathway that is different in some way from that used by
`IL-4, which could provide a means of augmenting its fibro-
`genic potential. In contrast to IL-13, the extent to which
`IL-5 and eosinophils participate in fibrotic processes varies
`greatly, and no clear explanation has been proposed that
`adequately explains the widely divergent findings. However,
`Jacobsohn found that monitoring the peripheral eosinophil
`count post-transplantation might provide a means of detect-
`ing the development of CGVHD [75].
`Chemokines are potent leukocyte chemoattractants that
`cooperate with pro-fibrotic cytokines such as IL-13 and
`TGF-(cid:69) during the development of fibrosis by recruiting mac-
`rophages and other effector cells to sites of tissue damage.
`Chemokines and their receptors have been implicated in
`the pathogenesis of scleroderma by recruiting immune cells
`to target tissues and thus, contribute to tissue damage [76].
`Although numerous chemokine signaling pathways are prob-
`ably involved in fibrogenesis, the CC-chemokine family has
`been shown to play an important regulatory role. In partic-
`ular, CCL3 (macrophage inflammatory protein 1(cid:68), MIP-1(cid:68))
`and CCL2 (monocyte chemoattractant protein 1, MCP-1)
`are chemotactic for mononuclear phagocytes and have been
`identified to be essential pro-fibrotic mediators. In a murine
`CGVHD model, high levels of chemokine mRNAs, i.e.,
`MCP-1, CCL5 (RANTES), CCL17, and IFN-(cid:74)-inducible che-
`mokines (CXCL9/Mig, CXCL10/IP-10, and CXCL11/I-TAC),
`which are all monocyte/macrophage- and T cell-related,
`were observed from days 7 to 120 post-transplantation [77].
`In a previous study, we found that pravastatin attenuates
`murine CGVHD by blocking the influx of effector cells into
`target organs and by downregulating the protein expressions
`
`Korean J Hematol 2011;46:80-7.
`
`of MCP-1 and RANTES, thereby reducing collagen synthesis
`[78].
`
`CGVHD ANIMAL MODELS
`
`Several murine allo-HSCT models have been used to study
`the pathogenesis of CGVHD. The first type of model involves
`the transplantation of parental lymphocytes into non-irradi-
`ated MHC-mismatched F1 recipients [79, 80]. In this model
`type, F1 recipients develop high levels of serum anti-double-
`strand DNA (dsDNA) and glomerulonephritis, and autoanti-
`body production is the result of a cognate interaction between
`donor CD4+ T cells and host B cells [79, 81-83]. However,
`it is not clear whether mechanisms revealed by this model
`reflect the pathogenesis of CGVHD in human transplant
`recipients receiving conditioning.
`The second type of model involves the transplantation
`of donor lymphocytes into MHC-matched but mHA-mis-
`matched irradiated recipients. In this model, donor LP/J
`(H-2b) bone marrow and spleen cells were transplanted into
`lethally irradiated C57BL/6 (H-2b) recipients, which later
`developed acute and chronic forms of GVHD [84]. Clonal
`analysis of T cells from the C57BL/6 recipients indicated
`that acute GVHD development was due primarily to recipi-
`ent-specific donor CTL, whereas CGVHD development was
`caused by autoreactive CD4+ T lymphocytes [84].
`In another mHA-disparate model, B10D2 (H-2d) donor
`spleen cells were transplanted into lethally irradiated BALB/c
`recipients, which then developed sclerodermatous organ
`damage [32, 85]. Skin changes in this model include a mono-
`nuclear infiltrate deep in dermis, loss of dermal fat, increased
`collagen deposition, and “dropout” of dermal appendages,
`such as hair follicles; unlike that found in acute GVHD,
`the apoptosis of basal epithelial cells at the dermal-epidermal
`junction does not occur. Clinical manifestations begin as
`early as day 11 post-transplantation, and cutaneous fibrosis
`is apparent as early as day 21. Deposits of IgG, IgA, and
`IgM appear at the dermal epidermal junction in recipients
`[85]. Additional features of CGVHD in this model include
`inflammation and fibrosis in salivary and lacrimal glands,
`sclerosing cholangitis, progressive renal and gastrointestinal
`fibrosis, and the development of anti-Scl-70 antibody [86].
`Naïve donor CD4 cells initiate the disease in this strain
`combination [87], and the dermal infiltrate is comprised
`of T cells, monocytes, and macrophages [88]. T cells and
`macrophages in skin express TGF-(cid:69)1 but not TGF-(cid:69)2 or
`TGF-(cid:69)3 mRNA [89]. In a microarray analysis study, the
`expression of type 1 (IFN-(cid:74)) and type 2 (IL-6, IL-10, and
`IL-13) cytokines, chemokines, and a variety of growth factors
`and cell adhesion molecules were upregulated in recipients
`with CGVHD compared to recipients without it [77].
`Zhang developed a new type of CGVHD model based
`on the transplantation of DBA/2 (H-2d) spleen cells into
`MHC-matched but mHA-mismatched, sub-lethally irradi-
`ated BALB/c (H-2d) recipients; in this model, both donor
`CD25-CD4+ T cells and B cells were required for CGVHD
`
`

`

`Pathogenesis of chronic GVHD
`
`85
`
`development [32]. However, the relevance of this model
`for human CGVHD is questionable because even though
`dsDNA-specific autoantibodies, immune complex glomer-
`ulonephritis, and proteinuria are characteristic of systemic
`lupus, they rarely occur in patients with CGVHD [90].
`
`CONCLUSION
`
`Alloreactivity forms the basis of the pathogenesis of
`CGVHD, but the phenotypes and origins of the alloreactive
`cells involved remain somewhat ambiguous. Attempts to
`study CGVHD experimentally have been somewhat ham-
`pered by the absence of a reliable animal model that exactly
`represents variable manifestations in humans. Nevertheless,
`thymic dysfunction, Treg deficiency, autoantibody formation
`with B cell activation, and dysregulatory fibrotic processes
`have been shown to be associated with the occurrence of
`CGVHD. Fundamental research on the pathophysiology of
`CGVHD is required for the development of more effective
`prophylactic and treatment regimens. Finally, improved
`methods of diagnosis and staging based on an understanding
`of the pathogenesis of CGVHD should help to exploit novel
`therapeutic approaches in the future.
`
`REFERENCES
`
`1. Lee SJ, Vogelsang G, Flowers ME. Chronic graft-versus-host
`disease. Biol Blood Marrow Transplant 2003;9:215-33.
`2. Baird K, Pavletic SZ. Chronic graft versus host disease. Curr Opin
`Hematol 2006;13:426-35.
`3. Lee SJ, Klein JP, Barrett AJ, et al. Severity of chronic graft-ver-
`sus-host disease: association with treatment-related mortality
`and relapse. Blood 2002;100:406-14.
`4. Socié G, Stone JV, Wingard JR, et al. Long-term survival and late
`deaths after allogeneic bone marrow transplantation. Late Effects
`Working Committee of the International Bone Marrow Trans-
`plant Registry. N Engl J Med 1999;341:14-21.
`5. Lee SJ, Kim HT, Ho VT, et al. Quality of life associated with acute
`and chronic graft-versus-host disease. Bone Marrow Transplant
`2006;38:305-10.
`6. Fraser CJ, Bhatia S, Ness K, et al. Impact of chronic graft-ver-
`sus-host disease on the health status of hematopoietic cell trans-
`plantation survivors: a report from the Bone Marrow Transplant
`Survivor Study. Blood 2006;108:2867-73.
`7. Cutler C, Giri S, Jeyapalan S, Paniagua D, Viswanathan A, Antin
`JH. Acute and chronic graft-versus-host disease after allogeneic
`peripheral-blood stem-cell and bone marrow transplantation: a
`meta-analysis. J Clin Oncol 2001;19:3685-91.
`8. Schmitz N, Eapen M, Horowitz MM, et al. Long-term outcome
`of patients given transplants of mobilized blood or bone marrow:
`a report from the International Bone Marrow Transplant Regi-
`stry and the European Group for Blood and Marrow Transplanta-
`tion. Blood 2006;108:4288-90.
`9. Akpek G, Lee SM, Anders V, Vogelsang GB. A high-dose pulse ste-
`roid regimen for controlling active chronic graft-versus-host
`
`disease. Biol Blood Marrow Transplant 2001;7:495-502.
`10. Arora M, Wagner JE, Davies SM, et al. Randomized clinical trial
`of thalidomide, cyclosporine, and prednisone versus cyclosporine
`and prednisone as initial therapy for chronic graft-versus-host
`disease. Biol Blood Marrow Transplant 2001;7:265-73.
`11. Lopez F, Parker P, Nademanee A, et al. Efficacy of mycophenolate
`mofetil in the treatment of chronic graft-versus-host disease. Biol
`Blood Marrow Transplant 2005;11:307-13.
`12. Busca A, Locatelli F, Marmont F, Audisio E, Falda M. Response to
`mycophenolate mofetil therapy in refractory chronic graft-ver-
`sus-host disease. Haematologica 2003;88:837-9.
`13. Goldberg JD, Jacobsohn DA, Margolis J, et al. Pentostatin for the
`treatment of chronic graft-versus-host disease in children. J
`Pediatr Hematol Oncol 2003;25:584-8.
`14. Carnevale-Schianca F, Martin P, Sullivan K, et al. Changing from
`cyclosporine to tacrolimus as salvage therapy for chronic graft-
`versus-host disease. Biol Blood Marrow Transplant 2000;6:613-
`20.
`15. Couriel DR, Hosing C, Saliba R, et al. Extracorporeal photo-
`chemotherapy for the treatment of steroid-resistant chronic
`GVHD. Blood 2006;107:3074-80.
`16. Couriel DR, Saliba R, Escalón MP, et al. Sirolimus in combination
`with tacrolimus and corticosteroids for the treatment of resistant
`chronic graft-versus-host disease. Br J Haematol 2005;130:409-
`17.
`17. Foss FM, DiVenuti GM, Chin K, et al. Prospective study of ex-
`tracorporeal photopheresis in steroid-refractory or steroid-re-
`sistant extensive chronic graft-versus-host disease: analysis of re-
`sponse and survival incorporating prognostic factors. Bone
`Marrow Transplant 2005;35:1187-93.
`18. Greinix HT, Volc-Platzer B, Rabitsch W, et al. Successful use of
`extracorporeal photochemotherapy in the treatment of severe
`acute and chronic graft-versus-host disease. Blood 1998;92:3098-
`104.
`19. Johnston LJ, Brown J, Shizuru JA, et al. Rapamycin (sirolimus) for
`treatment of chronic graft-versus-host disease. Biol Blood
`Marrow Transplant 2005;11:47-55.
`20. Ferrara JL, Levy R, Chao NJ. Pathophysiologic mechanisms of
`acute graft-vs.-host disease. Biol Blood Marrow Transplant
`1999;5:347-56.
`21. Sullivan KM, Shulman HM, Storb R, et al. Chronic graft-ver-
`sus-host disease in 52 patients: adverse natural course and success-
`ful treatment with combination immunosuppression. Blood
`1981;57:267-76.
`22. Imanguli MM, Alevizos I, Brown R, Pavletic SZ, Atkinson JC. Oral
`graft-versus-host disease. Oral Dis 2008;14:396-412.
`23. Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes
`of Health consensus development project on criteria for clinical
`trials in chronic graft-versus-host disease: I. Diagnosis and staging
`working group report. Biol Blood Marrow Transplant 2005;11:
`945-56.
`24. Arora M, Nagaraj S, Witte J, et al. New classification of chronic
`GVHD: added clarity from the consensus diagnoses. Bone Marrow
`Transplant 2009;43:149-53.
`25. Cho BS, Min CK, Eom KS

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