Review Article

Split Viewer

Korean J Hematol 2011; 46(2):

Published online June 21, 2011

https://doi.org/10.5045/kjh.2011.46.2.80

© The Korean Society of Hematology

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.

Correspondence to : 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: +82-2-2258-6053, Fax: +82-2-599-3589, ckmin@catholic.ac.kr

Received: June 9, 2011; Accepted: June 10, 2011

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.

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) regulatory 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 following 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 cytokine networks implicated in its pathogenesis, and the animal models used to devise strategies to prevent and treat CGVHD are reviewed.

Keywords Chronic graft-versus-host disease, Pathophysiology, Acute graft-versus-host disease, Fibrosis, Mouse model

Chronic graft-versus-host disease (CGVHD) remains a major 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-leukemia effects [3]. Over the past 10 years, CGVHD has emerged as the most troublesome complication of allo-HSCT. Improvements in human histocompatibility antigen (HLA) typing for unrelated transplantation, the adoption of new acute GVHD prophylaxis measures, reductions in conditioning regimen 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 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 inhibitors, 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 clinical observations have elucidated the pathophysiology of acute GVHD, but the biology of CGVHD has not been determined. 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 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.

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 preceding 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 cholestatic 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 secondary involvement of alveolar components. Pulmonary tissue can also be involved, although histological distinctions from bacterial and viral infections are sometimes difficult. Nevertheless, bronchiolitis obliterans, similar to that observed during lung transplant rejection, is now generally considered a manifestation of CGVHD.

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 assessment 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 addition, 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 possible roles for the new global scoring system in the assessment of CGVHD severity [24-26].

Acute GVHD resembles a toxic, sepsis-like syndrome. Host antigen-presenting cells (APCs), especially dendritic cells (DCs), present alloantigens to incoming alloreactive cytotoxic 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 lipopolysaccharide 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 treatment 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 tolerance)

It has been suggested that immune tolerance to self-antigens is disrupted in CGVHD, and that these give rise to the autoimmune manifestations of the disorder. One attractive hypothesis is that thymic epithelial damage caused by conditioning regimens and/or acute GVHD leads to dysregulation of central tolerance mechanisms during the reconstitution 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 double-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 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 mechanisms 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 prevent 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 factor (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 generation of host-reactive T cells and the development of CGVHD.

2. CD4<sup>+</sup>CD25<sup>+</sup> regulatory T cells (Tregs) and their relationship to CGVHD

In several series, regulatory T cell (Treg) numbers (determined 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 contradictory. Clark found that CGVHD is associated with elevated numbers of peripheral blood Tregs and that these numbers 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 suppression is mediated by cytokines, such as transforming growth factor (TGF)-β and interleukin (IL)-10, or by contact with plasmacytoid DCs through indoleamine 2,3-dioxygenase [38]. Tregs may also exert an inhibitory influence directly in target tissues [39]. For example, mucosal Treg numbers have been documented to be lower in patients with GVHD than in normal controls or patients without GVHD [34]. Interestingly, extracorporeal photochemotherapy increases levels of circulating functional Tregs in CGVHD patients [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 concluded that alloantigen-driven expansion, rather than homeostatic proliferation, is critical for the effectiveness of Tregs in CGVHD, and suggested that cellular therapy with alloantigen-induced Tregs in combination with glucocorticoids 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 highlighted 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 autoantibody 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 receptor 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 autoimmunity, 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 asymptomatic patients. The development of antibodies to minor histocompatibility antigens (mHA) encoded on the Y chromosome 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 depletion 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 conceivable that alloreactive donor CD4+ T cells could be activated 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 autoimmune responses and in the epitope spreading of autoreactive 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 destructive changes at the dermal-epidermal junction, accompanied by irregular acanthosis, hyperkeratosis, atrophy, progressing 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 polarized 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-β 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-β released by platelets during clotting. Gene expression studies have demonstrated that increased TGF-β signaling in CD4 cells and CD8 cells is associated with a reduced risk of CGVHD in man [61]. The association between increased TGF-β 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-β1 and collagen mRNAs commonly occur in human scleroderma and murine sclerodermatous GVHD following transplantation of B10.D2 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 progression of CGVHD, and the type of CD4+ T-cell response that develops is crucial. Studies using various cytokine-deficient 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)-γ, 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 deposition of connective-tissue elements and that the magnitude of fibrosis is tightly regulated by the phenotype of the developing Th-cell response. Furthermore, IL-13 and IL-4 bind to the same signaling receptor (IL-4Rα-IL-13Rα1) on fibroblasts [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 fibrogenic 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 detecting the development of CGVHD [75].

Chemokines are potent leukocyte chemoattractants that cooperate with pro-fibrotic cytokines such as IL-13 and TGF-β during the development of fibrosis by recruiting macrophages 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 probably involved in fibrogenesis, the CC-chemokine family has been shown to play an important regulatory role. In particular, CCL3 (macrophage inflammatory protein 1α, MIP-1α) 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-γ-inducible chemokines (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 of MCP-1 and RANTES, thereby reducing collagen synthesis [78].

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-irradiated 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 autoantibody 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-mismatched 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 recipient-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 mononuclear 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-β1 but not TGF-β2 or TGF-β3 mRNA [89]. In a microarray analysis study, the expression of type 1 (IFN-γ) 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 irradiated BALB/c (H-2d) recipients; in this model, both donor CD25-CD4+ T cells and B cells were required for CGVHD development [32]. However, the relevance of this model for human CGVHD is questionable because even though dsDNA-specific autoantibodies, immune complex glomerulonephritis, and proteinuria are characteristic of systemic lupus, they rarely occur in patients with CGVHD [90].

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 hampered 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.

  1. Lee, SJ, Vogelsang, G, Flowers, ME. Chronic graft-versus-host disease. Biol Blood Marrow Transplant, 2003;9;215-233.
    Pubmed
  2. Baird, K, Pavletic, SZ. Chronic graft versus host disease. Curr Opin Hematol, 2006;13;426-435.
    Pubmed
  3. Lee, SJ, Klein, JP, Barrett, AJ, et al. Severity of chronic graft-versus-host disease: association with treatment-related mortality and relapse. Blood, 2002;100;406-414.
    Pubmed
  4. Socié, G, Stone, JV, Wingard, JR, et al, Late Effects Working Committee of the International Bone Marrow Transplant Registry. Long-term survival and late deaths after allogeneic bone marrow transplantation. N Engl J Med, 1999;341;14-21.
    Pubmed
  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;305-310.
    Pubmed
  6. Fraser, CJ, Bhatia, S, Ness, K, et al. Impact of chronic graft-versus-host disease on the health status of hematopoietic cell transplantation survivors: a report from the Bone Marrow Transplant Survivor Study. Blood, 2006;108;2867-2873.
    Pubmed
  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;3685-3691.
    Pubmed
  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 Registry and the European Group for Blood and Marrow Transplantation. Blood, 2006;108;4288-4290.
    Pubmed
  9. Akpek, G, Lee, SM, Anders, V, Vogelsang, GB. A high-dose pulse steroid regimen for controlling active chronic graft-versus-host disease. Biol Blood Marrow Transplant, 2001;7;495-502.
    Pubmed
  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;265-273.
    Pubmed
  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;307-313.
    Pubmed
  12. Busca, A, Locatelli, F, Marmont, F, Audisio, E, Falda, M. Response to mycophenolate mofetil therapy in refractory chronic graft-versus-host disease. Haematologica, 2003;88;837-839.
    Pubmed
  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;584-588.
    Pubmed
  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;613-620.
    Pubmed
  15. Couriel, DR, Hosing, C, Saliba, R, et al. Extracorporeal photochemotherapy for the treatment of steroid-resistant chronic GVHD. Blood, 2006;107;3074-3080.
    Pubmed
  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;409-417.
    Pubmed
  17. Foss, FM, DiVenuti, GM, Chin, K, et al. Prospective study of extracorporeal photopheresis in steroid-refractory or steroid-resistant extensive chronic graft-versus-host disease: analysis of response and survival incorporating prognostic factors. Bone Marrow Transplant, 2005;35;1187-1193.
    Pubmed
  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;3098-3104.
    Pubmed
  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.
    Pubmed
  20. Ferrara, JL, Levy, R, Chao, NJ. Pathophysiologic mechanisms of acute graft-vs.-host disease. Biol Blood Marrow Transplant, 1999;5;347-356.
    Pubmed
  21. Sullivan, KM, Shulman, HM, Storb, R, et al. Chronic graft-versus-host disease in 52 patients: adverse natural course and successful treatment with combination immunosuppression. Blood, 1981;57;267-276.
    Pubmed
  22. Imanguli, MM, Alevizos, I, Brown, R, Pavletic, SZ, Atkinson, JC. Oral graft-versus-host disease. Oral Dis, 2008;14;396-412.
    Pubmed
  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;945-956.
    Pubmed
  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;149-153.
    Pubmed
  25. Cho, BS, Min, CK, Eom, KS, et al. Feasibility of NIH consensus criteria for chronic graft-versus-host disease. Leukemia, 2009;23;78-84.
    Pubmed
  26. Vigorito, AC, Campregher, PV, Storer, BE, et al. Evaluation of NIH consensus criteria for classification of late acute and chronic GVHD. Blood, 2009;114;702-708.
    Pubmed
  27. Choi, SW, Levine, JE, Ferrara, JL. Pathogenesis and management of graft-versus-host disease. Immunol Allergy Clin North Am, 2010;30;75-101.
    Pubmed
  28. Sprent, J, Kishimoto, H. The thymus and central tolerance. Transplantation, 2001;72;S25-S28.
    Pubmed
  29. Dutt, S, Tseng, D, Ermann, J, et al. Naive and memory T cells induce different types of graft-versus-host disease. J Immunol, 2007;179;6547-6554.
    Pubmed
  30. Panoskaltsis-Mortari, A, Lacey, DL, Vallera, DA, Blazar, BR. Keratinocyte growth factor administered before conditioning ameliorates graft-versus-host disease after allogeneic bone marrow transplantation in mice. Blood, 1998;92;3960-3967.
    Pubmed
  31. Blazar, BR, Weisdorf, DJ, Defor, T, et al. Phase 1/2 randomized, placebo-control trial of palifermin to prevent graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Blood, 2006;108;3216-3222.
    Pubmed
  32. Zhang, C, Todorov, I, Zhang, Z, et al. Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations. Blood, 2006;107;2993-3001.
    Pubmed
  33. Imado, T, Iwasaki, T, Kataoka, Y, et al. Hepatocyte growth factor preserves graft-versus-leukemia effect and T-cell reconstitution after marrow transplantation. Blood, 2004;104;1542-1549.
    Pubmed
  34. Rieger, K, Loddenkemper, C, Maul, J, et al. Mucosal FOXP3+ regulatory T cells are numerically deficient in acute and chronic GvHD. Blood, 2006;107;1717-1723.
    Pubmed
  35. Anderson, BE, McNiff, JM, Matte, C, Athanasiadis, I, Shlomchik, WD, Shlomchik, MJ. Recipient CD4+ T cells that survive irradiation regulate chronic graft-versus-host disease. Blood, 2004;104;1565-1573.
    Pubmed
  36. Zorn, E, Kim, HT, Lee, SJ, et al. Reduced frequency of FOXP3+CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease. Blood, 2005;106;2903-2911.
    Pubmed
  37. Clark, FJ, Gregg, R, Piper, K, et al. Chronic graft-versus-host disease is associated with increased numbers of peripheral blood CD4+CD25high regulatory T cells. Blood, 2004;103;2410-2416.
    Pubmed
  38. Sharma, MD, Baban, B, Chandler, P, et al. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest, 2007;117;2570-2582.
    Pubmed
  39. Nguyen, VH, Zeiser, R, Dasilva, DL, et al. In vivo dynamics of regulatory T-cell trafficking and survival predict effective strategies to control graft-versus-host disease following allogeneic transplantation. Blood, 2007;109;2649-2656.
    Pubmed
  40. Di Biaso, I, Di Maio, L, Bugarin, C, et al. Regulatory T cells and extracorporeal photochemotherapy: correlation with clinical response and decreased frequency of proinflammatory T cells. Transplantation, 2009;87;1422-1425.
    Pubmed
  41. Bastien, JP, Krosl, G, Therien, C, et al. Photodepletion differentially affects CD4+ Tregs versus CD4+ effector T cells from patients with chronic graft-versus-host disease. Blood, 2010;116;4859-4869.
    Pubmed
  42. Hoffmann, P, Ermann, J, Edinger, M, Fathman, CG, Strober, S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med, 2002;196;389-399.
    Pubmed
  43. Giorgini, A, Noble, A. Blockade of chronic graft-versus-host disease by alloantigen-induced CD4+CD25+Foxp3+ regulatory T cells in nonlymphopenic hosts. J Leukoc Biol, 2007;82;1053-1061.
    Pubmed
  44. Chen, X, Vodanovic-Jankovic, S, Johnson, B, Keller, M, Komorowski, R, Drobyski, WR. Absence of regulatory T-cell control of TH1 and TH17 cells is responsible for the autoimmune-mediated pathology in chronic graft-versus-host disease. Blood, 2007;110;3804-3813.
    Pubmed
  45. Ratanatharathorn, V, Carson, E, Reynolds, C, et al. Anti-CD20 chimeric monoclonal antibody treatment of refractory immune-mediated thrombocytopenia in a patient with chronic graft-versus-host disease. Ann Intern Med, 2000;133;275-279.
    Pubmed
  46. Patriarca, F, Skert, C, Sperotto, A, et al. The development of autoantibodies after allogeneic stem cell transplantation is related with chronic graft-vs-host disease and immune recovery. Exp Hematol, 2006;34;389-396.
    Pubmed
  47. Svegliati, S, Olivieri, A, Campelli, N, et al. Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease. Blood, 2007;110;237-241.
    Pubmed
  48. Sarantopoulos, S, Stevenson, KE, Kim, HT, et al. High levels of B-cell activating factor in patients with active chronic graft-versus-host disease. Clin Cancer Res, 2007;13;6107-6114.
    Pubmed
  49. Miklos, DB, Kim, HT, Miller, KH, et al. Antibody responses to H-Y minor histocompatibility antigens correlate with chronic graft-versus-host disease and disease remission. Blood, 2005;105;2973-2978.
    Pubmed
  50. Cutler, C, Miklos, D, Kim, HT, et al. Rituximab for steroid-refractory chronic graft-versus-host disease. Blood, 2006;108;756-762.
    Pubmed
  51. Kim, SJ, Lee, JW, Jung, CW, et al. Weekly rituximab followed by monthly rituximab treatment for steroid-refractory chronic graft-versus-host disease: results from a prospective, multicenter, phase II study. Haematologica, 2010;95;1935-1942.
    Pubmed
  52. Alousi, AM, Uberti, J, Ratanatharathorn, V. The role of B cell depleting therapy in graft versus host disease after allogeneic hematopoietic cell transplant. Leuk Lymphoma, 2010;51;376-389.
    Pubmed
  53. Puliaev, R, Puliaeva, I, Welniak, LA, et al. CTL-promoting effects of CD40 stimulation outweigh B cell-stimulatory effects resulting in B cell elimination and disease improvement in a murine model of lupus. J Immunol, 2008;181;47-61.
    Pubmed
  54. Shulman, HM, Kleiner, D, Lee, SJ, et al. Histopathologic diagnosis of chronic graft-versus-host disease: National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: II. Pathology Working Group Report. Biol Blood Marrow Transplant, 2006;12;31-47.
    Pubmed
  55. Wynn, TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol, 2004;4;583-594.
    Pubmed
  56. Nikolic, B, Lee, S, Bronson, RT, Grusby, MJ, Sykes, M. Th1 and Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets. J Clin Invest, 2000;105;1289-1298.
    Pubmed
  57. Hillebrandt, S, Wasmuth, HE, Weiskirchen, R, et al. Complement factor 5 is a quantitative trait gene that modifies liver fibrogenesis in mice and humans. Nat Genet, 2005;37;835-843.
    Pubmed
  58. Niculescu, F, Niculescu, T, Nguyen, P, et al. Both apoptosis and complement membrane attack complex deposition are major features of murine acute graft-vs.-host disease. Exp Mol Pathol, 2005;79;136-145.
    Pubmed
  59. Tsoi, MS, Storb, R, Jones, E, et al. Deposition of IgM and complement at the dermoepidermal junction in acute and chronic cutaneous graft-vs-host disease in man. J Immunol, 1978;120;1485-1492.
    Pubmed
  60. Liem, LM, Fibbe, WE, van Houwelingen, HC, Goulmy, E. Serum transforming growth factor-beta1 levels in bone marrow transplant recipients correlate with blood cell counts and chronic graft-versus-host disease. Transplantation, 1999;67;59-65.
    Pubmed
  61. Baron, C, Somogyi, R, Greller, LD, et al. Prediction of graft-versus-host disease in humans by donor gene-expression profiling. PLoS Med, 2007;4;e23.
    Pubmed
  62. Jaffee, BD, Claman, HN. Chronic graft-versus-host disease (GVHD) as a model for scleroderma. I. Description of model systems. Cell Immunol, 1983;77;1-12.
    Pubmed
  63. Allen, RD, Staley, TA, Sidman, CL. Differential cytokine expression in acute and chronic murine graft-versus-host-disease. Eur J Immunol, 1993;23;333-337.
    Pubmed
  64. De Wit, D, Van Mechelen, M, Zanin, C, et al. Preferential activation of Th2 cells in chronic graft-versus-host reaction. J Immunol, 1993;150;361-366.
    Pubmed
  65. Doutrelepont, JM, Moser, M, Leo, O, et al. Hyper IgE in stimulatory graft-versus-host disease: role of interleukin-4. Clin Exp Immunol, 1991;83;133-136.
    Pubmed
  66. Garlisi, CG, Pennline, KJ, Smith, SR, Siegel, MI, Umland, SP. Cytokine gene expression in mice undergoing chronic graft-versus-host disease. Mol Immunol, 1993;30;669-677.
    Pubmed
  67. Tanaka, J, Imamura, M, Kasai, M, et al. The important balance between cytokines derived from type 1 and type 2 helper T cells in the control of graft-versus-host disease. Bone Marrow Transplant, 1997;19;571-576.
    Pubmed
  68. Umland, SP, Razac, S, Nahrebne, DK, Seymour, BW. Effects of in vivo administration of interferon (IFN)-gamma, anti-IFN-gamma, or anti-interleukin-4 monoclonal antibodies in chronic autoimmune graft-versus-host disease. Clin Immunol Immunopathol, 1992;63;66-73.
    Pubmed
  69. Wynn, TA, Cheever, AW, Jankovic, D, et al. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature, 1995;376;594-596.
    Pubmed
  70. Hoffmann, KF, Cheever, AW, Wynn, TA. IL-10 and the dangers of immune polarization: excessive type 1 and type 2 cytokine responses induce distinct forms of lethal immunopathology in murine schistosomiasis. J Immunol, 2000;164;6406-6416.
    Pubmed
  71. Murata, T, Husain, SR, Mohri, H, Puri, RK. Two different IL-13 receptor chains are expressed in normal human skin fibroblasts, and IL-4 and IL-13 mediate signal transduction through a common pathway. Int Immunol, 1998;10;1103-1110.
    Pubmed
  72. Chiaramonte, MG, Donaldson, DD, Cheever, AW, Wynn, TA. An IL-13 inhibitor blocks the development of hepatic fibrosis during a T-helper type 2-dominated inflammatory response. J Clin Invest, 1999;104;777-785.
    Pubmed
  73. Oriente, A, Fedarko, NS, Pacocha, SE, Huang, SK, Lichtenstein, LM, Essayan, DM. Interleukin-13 modulates collagen homeostasis in human skin and keloid fibroblasts. J Pharmacol Exp Ther, 2000;292;988-994.
    Pubmed
  74. Saito, A, Okazaki, H, Sugawara, I, Yamamoto, K, Takizawa, H. Potential action of IL-4 and IL-13 as fibrogenic factors on lung fibroblasts in vitro. Int Arch Allergy Immunol, 2003;132;168-176.
    Pubmed
  75. Jacobsohn, DA, Schechter, T, Seshadri, R, Thormann, K, Duerst, R, Kletzel, M. Eosinophilia correlates with the presence or development of chronic graft-versus-host disease in children. Transplantation, 2004;77;1096-1100.
    Pubmed
  76. Atamas, SP, White, B. The role of chemokines in the pathogenesis of scleroderma. Curr Opin Rheumatol, 2003;15;772-777.
    Pubmed
  77. Zhou, L, Askew, D, Wu, C, Gilliam, AC. Cutaneous gene expression by DNA microarray in murine sclerodermatous graft-versus-host disease, a model for human scleroderma. J Invest Dermatol, 2007;127;281-292.
    Pubmed
  78. Yoon, HK, Lim, JY, Kim, TJ, Cho, CS, Min, CK. Effects of pravastatin on murine chronic graft-versus-host disease. Transplantation, 2010;90;853-860.
    Pubmed
  79. Morris, SC, Cheek, RL, Cohen, PL, Eisenberg, RA. Allotype-specific immunoregulation of autoantibody production by host B cells in chronic graft-versus host disease. J Immunol, 1990;144;916-922.
    Pubmed
  80. van der Veen, FM, Rolink, AG, Gleichmann, E. Autoimmune disease strongly resembling systemic lupus erythematosus (SLE) in F1 mice undergoing graft-versus-host reaction (GVHR). Adv Exp Med Biol, 1982;149;669-677.
    Pubmed
  81. Sekiguchi, DR, Eisenberg, RA, Weigert, M. Secondary heavy chain rearrangement: a mechanism for generating anti-double-stranded DNA B cells. J Exp Med, 2003;197;27-39.
    Pubmed
  82. Sekiguchi, DR, Jainandunsing, SM, Fields, ML, et al. Chronic graft-versus-host in Ig knockin transgenic mice abrogates B cell tolerance in anti-double-stranded DNA B cells. J Immunol, 2002;168;4142-4153.
    Pubmed
  83. Via, CS, Shearer, GM. T-cell interactions in autoimmunity: insights from a murine model of graft-versus-host disease. Immunol Today, 1988;9;207-213.
    Pubmed
  84. Parkman, R. Clonal analysis of murine graft-vs-host disease. I. Phenotypic and functional analysis of T lymphocyte clones. J Immunol, 1986;136;3543-3548.
    Pubmed
  85. Claman, HN, Jaffee, BD, Huff, JC, Clark, RA. Chronic graft-versus-host disease as a model for scleroderma. II. Mast cell depletion with deposition of immunoglobulins in the skin and fibrosis. Cell Immunol, 1985;94;73-84.
    Pubmed
  86. Ruzek, MC, Jha, S, Ledbetter, S, Richards, SM, Garman, RD. A modified model of graft-versus-host-induced systemic sclerosis (scleroderma) exhibits all major aspects of the human disease. Arthritis Rheum, 2004;50;1319-1331.
    Pubmed
  87. Anderson, BE, McNiff, J, Yan, J, et al. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest, 2003;112;101-108.
    Pubmed
  88. McCormick, LL, Zhang, Y, Tootell, E, Gilliam, AC. Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J Immunol, 1999;163;5693-5699.
    Pubmed
  89. Zhang, Y, McCormick, LL, Desai, SR, Wu, C, Gilliam, AC. Murine sclerodermatous graft-versus-host disease, a model for human scleroderma: cutaneous cytokines, chemokines, and immune cell activation. J Immunol, 2002;168;3088-3098.
    Pubmed
  90. Rouquette-Gally, AM, Boyeldieu, D, Prost, AC, Gluckman, E. Autoimmunity after allogeneic bone marrow transplantation. A study of 53 long-term-surviving patients. Transplantation, 1988;46;238-240.
    Pubmed

Article

Review Article

Korean J Hematol 2011; 46(2): 80-87

Published online June 21, 2011 https://doi.org/10.5045/kjh.2011.46.2.80

Copyright © The Korean Society of Hematology.

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.

Correspondence to: 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: +82-2-2258-6053, Fax: +82-2-599-3589, ckmin@catholic.ac.kr

Received: June 9, 2011; Accepted: June 10, 2011

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.

Abstract

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) regulatory 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 following 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 cytokine networks implicated in its pathogenesis, and the animal models used to devise strategies to prevent and treat CGVHD are reviewed.

Keywords: Chronic graft-versus-host disease, Pathophysiology, Acute graft-versus-host disease, Fibrosis, Mouse model

INTRODUCTION

Chronic graft-versus-host disease (CGVHD) remains a major 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-leukemia effects [3]. Over the past 10 years, CGVHD has emerged as the most troublesome complication of allo-HSCT. Improvements in human histocompatibility antigen (HLA) typing for unrelated transplantation, the adoption of new acute GVHD prophylaxis measures, reductions in conditioning regimen 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 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 inhibitors, 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 clinical observations have elucidated the pathophysiology of acute GVHD, but the biology of CGVHD has not been determined. 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 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 preceding 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 cholestatic 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 secondary involvement of alveolar components. Pulmonary tissue can also be involved, although histological distinctions from bacterial and viral infections are sometimes difficult. Nevertheless, bronchiolitis obliterans, similar to that observed during 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 assessment 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 addition, 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 possible 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 cytotoxic 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 lipopolysaccharide 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 treatment 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 tolerance)

It has been suggested that immune tolerance to self-antigens is disrupted in CGVHD, and that these give rise to the autoimmune manifestations of the disorder. One attractive hypothesis is that thymic epithelial damage caused by conditioning regimens and/or acute GVHD leads to dysregulation of central tolerance mechanisms during the reconstitution 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 double-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 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 mechanisms 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 prevent 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 factor (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 generation of host-reactive T cells and the development of CGVHD.

2. CD4<sup>+</sup>CD25<sup>+</sup> regulatory T cells (Tregs) and their relationship to CGVHD

In several series, regulatory T cell (Treg) numbers (determined 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 contradictory. Clark found that CGVHD is associated with elevated numbers of peripheral blood Tregs and that these numbers 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 suppression is mediated by cytokines, such as transforming growth factor (TGF)-β and interleukin (IL)-10, or by contact with plasmacytoid DCs through indoleamine 2,3-dioxygenase [38]. Tregs may also exert an inhibitory influence directly in target tissues [39]. For example, mucosal Treg numbers have been documented to be lower in patients with GVHD than in normal controls or patients without GVHD [34]. Interestingly, extracorporeal photochemotherapy increases levels of circulating functional Tregs in CGVHD patients [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 concluded that alloantigen-driven expansion, rather than homeostatic proliferation, is critical for the effectiveness of Tregs in CGVHD, and suggested that cellular therapy with alloantigen-induced Tregs in combination with glucocorticoids 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 highlighted 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 autoantibody 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 receptor 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 autoimmunity, 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 asymptomatic patients. The development of antibodies to minor histocompatibility antigens (mHA) encoded on the Y chromosome 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 depletion 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 conceivable that alloreactive donor CD4+ T cells could be activated 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 autoimmune responses and in the epitope spreading of autoreactive 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 destructive changes at the dermal-epidermal junction, accompanied by irregular acanthosis, hyperkeratosis, atrophy, progressing 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 polarized 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-β 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-β released by platelets during clotting. Gene expression studies have demonstrated that increased TGF-β signaling in CD4 cells and CD8 cells is associated with a reduced risk of CGVHD in man [61]. The association between increased TGF-β 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-β1 and collagen mRNAs commonly occur in human scleroderma and murine sclerodermatous GVHD following transplantation of B10.D2 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 progression of CGVHD, and the type of CD4+ T-cell response that develops is crucial. Studies using various cytokine-deficient 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)-γ, 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 deposition of connective-tissue elements and that the magnitude of fibrosis is tightly regulated by the phenotype of the developing Th-cell response. Furthermore, IL-13 and IL-4 bind to the same signaling receptor (IL-4Rα-IL-13Rα1) on fibroblasts [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 fibrogenic 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 detecting the development of CGVHD [75].

Chemokines are potent leukocyte chemoattractants that cooperate with pro-fibrotic cytokines such as IL-13 and TGF-β during the development of fibrosis by recruiting macrophages 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 probably involved in fibrogenesis, the CC-chemokine family has been shown to play an important regulatory role. In particular, CCL3 (macrophage inflammatory protein 1α, MIP-1α) 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-γ-inducible chemokines (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 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-irradiated 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 autoantibody 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-mismatched 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 recipient-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 mononuclear 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-β1 but not TGF-β2 or TGF-β3 mRNA [89]. In a microarray analysis study, the expression of type 1 (IFN-γ) 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 irradiated BALB/c (H-2d) recipients; in this model, both donor CD25-CD4+ T cells and B cells were required for CGVHD development [32]. However, the relevance of this model for human CGVHD is questionable because even though dsDNA-specific autoantibodies, immune complex glomerulonephritis, 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 hampered 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.

Fig 1.

Figure 1.

The pathophysiology of chronic graft-versus-host disease.

Blood Research 2011; 46: 80-87https://doi.org/10.5045/kjh.2011.46.2.80

References

  1. Lee, SJ, Vogelsang, G, Flowers, ME. Chronic graft-versus-host disease. Biol Blood Marrow Transplant, 2003;9;215-233.
    Pubmed
  2. Baird, K, Pavletic, SZ. Chronic graft versus host disease. Curr Opin Hematol, 2006;13;426-435.
    Pubmed
  3. Lee, SJ, Klein, JP, Barrett, AJ, et al. Severity of chronic graft-versus-host disease: association with treatment-related mortality and relapse. Blood, 2002;100;406-414.
    Pubmed
  4. Socié, G, Stone, JV, Wingard, JR, et al, Late Effects Working Committee of the International Bone Marrow Transplant Registry. Long-term survival and late deaths after allogeneic bone marrow transplantation. N Engl J Med, 1999;341;14-21.
    Pubmed
  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;305-310.
    Pubmed
  6. Fraser, CJ, Bhatia, S, Ness, K, et al. Impact of chronic graft-versus-host disease on the health status of hematopoietic cell transplantation survivors: a report from the Bone Marrow Transplant Survivor Study. Blood, 2006;108;2867-2873.
    Pubmed
  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;3685-3691.
    Pubmed
  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 Registry and the European Group for Blood and Marrow Transplantation. Blood, 2006;108;4288-4290.
    Pubmed
  9. Akpek, G, Lee, SM, Anders, V, Vogelsang, GB. A high-dose pulse steroid regimen for controlling active chronic graft-versus-host disease. Biol Blood Marrow Transplant, 2001;7;495-502.
    Pubmed
  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;265-273.
    Pubmed
  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;307-313.
    Pubmed
  12. Busca, A, Locatelli, F, Marmont, F, Audisio, E, Falda, M. Response to mycophenolate mofetil therapy in refractory chronic graft-versus-host disease. Haematologica, 2003;88;837-839.
    Pubmed
  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;584-588.
    Pubmed
  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;613-620.
    Pubmed
  15. Couriel, DR, Hosing, C, Saliba, R, et al. Extracorporeal photochemotherapy for the treatment of steroid-resistant chronic GVHD. Blood, 2006;107;3074-3080.
    Pubmed
  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;409-417.
    Pubmed
  17. Foss, FM, DiVenuti, GM, Chin, K, et al. Prospective study of extracorporeal photopheresis in steroid-refractory or steroid-resistant extensive chronic graft-versus-host disease: analysis of response and survival incorporating prognostic factors. Bone Marrow Transplant, 2005;35;1187-1193.
    Pubmed
  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;3098-3104.
    Pubmed
  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.
    Pubmed
  20. Ferrara, JL, Levy, R, Chao, NJ. Pathophysiologic mechanisms of acute graft-vs.-host disease. Biol Blood Marrow Transplant, 1999;5;347-356.
    Pubmed
  21. Sullivan, KM, Shulman, HM, Storb, R, et al. Chronic graft-versus-host disease in 52 patients: adverse natural course and successful treatment with combination immunosuppression. Blood, 1981;57;267-276.
    Pubmed
  22. Imanguli, MM, Alevizos, I, Brown, R, Pavletic, SZ, Atkinson, JC. Oral graft-versus-host disease. Oral Dis, 2008;14;396-412.
    Pubmed
  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;945-956.
    Pubmed
  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;149-153.
    Pubmed
  25. Cho, BS, Min, CK, Eom, KS, et al. Feasibility of NIH consensus criteria for chronic graft-versus-host disease. Leukemia, 2009;23;78-84.
    Pubmed
  26. Vigorito, AC, Campregher, PV, Storer, BE, et al. Evaluation of NIH consensus criteria for classification of late acute and chronic GVHD. Blood, 2009;114;702-708.
    Pubmed
  27. Choi, SW, Levine, JE, Ferrara, JL. Pathogenesis and management of graft-versus-host disease. Immunol Allergy Clin North Am, 2010;30;75-101.
    Pubmed
  28. Sprent, J, Kishimoto, H. The thymus and central tolerance. Transplantation, 2001;72;S25-S28.
    Pubmed
  29. Dutt, S, Tseng, D, Ermann, J, et al. Naive and memory T cells induce different types of graft-versus-host disease. J Immunol, 2007;179;6547-6554.
    Pubmed
  30. Panoskaltsis-Mortari, A, Lacey, DL, Vallera, DA, Blazar, BR. Keratinocyte growth factor administered before conditioning ameliorates graft-versus-host disease after allogeneic bone marrow transplantation in mice. Blood, 1998;92;3960-3967.
    Pubmed
  31. Blazar, BR, Weisdorf, DJ, Defor, T, et al. Phase 1/2 randomized, placebo-control trial of palifermin to prevent graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Blood, 2006;108;3216-3222.
    Pubmed
  32. Zhang, C, Todorov, I, Zhang, Z, et al. Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations. Blood, 2006;107;2993-3001.
    Pubmed
  33. Imado, T, Iwasaki, T, Kataoka, Y, et al. Hepatocyte growth factor preserves graft-versus-leukemia effect and T-cell reconstitution after marrow transplantation. Blood, 2004;104;1542-1549.
    Pubmed
  34. Rieger, K, Loddenkemper, C, Maul, J, et al. Mucosal FOXP3+ regulatory T cells are numerically deficient in acute and chronic GvHD. Blood, 2006;107;1717-1723.
    Pubmed
  35. Anderson, BE, McNiff, JM, Matte, C, Athanasiadis, I, Shlomchik, WD, Shlomchik, MJ. Recipient CD4+ T cells that survive irradiation regulate chronic graft-versus-host disease. Blood, 2004;104;1565-1573.
    Pubmed
  36. Zorn, E, Kim, HT, Lee, SJ, et al. Reduced frequency of FOXP3+CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease. Blood, 2005;106;2903-2911.
    Pubmed
  37. Clark, FJ, Gregg, R, Piper, K, et al. Chronic graft-versus-host disease is associated with increased numbers of peripheral blood CD4+CD25high regulatory T cells. Blood, 2004;103;2410-2416.
    Pubmed
  38. Sharma, MD, Baban, B, Chandler, P, et al. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest, 2007;117;2570-2582.
    Pubmed
  39. Nguyen, VH, Zeiser, R, Dasilva, DL, et al. In vivo dynamics of regulatory T-cell trafficking and survival predict effective strategies to control graft-versus-host disease following allogeneic transplantation. Blood, 2007;109;2649-2656.
    Pubmed
  40. Di Biaso, I, Di Maio, L, Bugarin, C, et al. Regulatory T cells and extracorporeal photochemotherapy: correlation with clinical response and decreased frequency of proinflammatory T cells. Transplantation, 2009;87;1422-1425.
    Pubmed
  41. Bastien, JP, Krosl, G, Therien, C, et al. Photodepletion differentially affects CD4+ Tregs versus CD4+ effector T cells from patients with chronic graft-versus-host disease. Blood, 2010;116;4859-4869.
    Pubmed
  42. Hoffmann, P, Ermann, J, Edinger, M, Fathman, CG, Strober, S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med, 2002;196;389-399.
    Pubmed
  43. Giorgini, A, Noble, A. Blockade of chronic graft-versus-host disease by alloantigen-induced CD4+CD25+Foxp3+ regulatory T cells in nonlymphopenic hosts. J Leukoc Biol, 2007;82;1053-1061.
    Pubmed
  44. Chen, X, Vodanovic-Jankovic, S, Johnson, B, Keller, M, Komorowski, R, Drobyski, WR. Absence of regulatory T-cell control of TH1 and TH17 cells is responsible for the autoimmune-mediated pathology in chronic graft-versus-host disease. Blood, 2007;110;3804-3813.
    Pubmed
  45. Ratanatharathorn, V, Carson, E, Reynolds, C, et al. Anti-CD20 chimeric monoclonal antibody treatment of refractory immune-mediated thrombocytopenia in a patient with chronic graft-versus-host disease. Ann Intern Med, 2000;133;275-279.
    Pubmed
  46. Patriarca, F, Skert, C, Sperotto, A, et al. The development of autoantibodies after allogeneic stem cell transplantation is related with chronic graft-vs-host disease and immune recovery. Exp Hematol, 2006;34;389-396.
    Pubmed
  47. Svegliati, S, Olivieri, A, Campelli, N, et al. Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease. Blood, 2007;110;237-241.
    Pubmed
  48. Sarantopoulos, S, Stevenson, KE, Kim, HT, et al. High levels of B-cell activating factor in patients with active chronic graft-versus-host disease. Clin Cancer Res, 2007;13;6107-6114.
    Pubmed
  49. Miklos, DB, Kim, HT, Miller, KH, et al. Antibody responses to H-Y minor histocompatibility antigens correlate with chronic graft-versus-host disease and disease remission. Blood, 2005;105;2973-2978.
    Pubmed
  50. Cutler, C, Miklos, D, Kim, HT, et al. Rituximab for steroid-refractory chronic graft-versus-host disease. Blood, 2006;108;756-762.
    Pubmed
  51. Kim, SJ, Lee, JW, Jung, CW, et al. Weekly rituximab followed by monthly rituximab treatment for steroid-refractory chronic graft-versus-host disease: results from a prospective, multicenter, phase II study. Haematologica, 2010;95;1935-1942.
    Pubmed
  52. Alousi, AM, Uberti, J, Ratanatharathorn, V. The role of B cell depleting therapy in graft versus host disease after allogeneic hematopoietic cell transplant. Leuk Lymphoma, 2010;51;376-389.
    Pubmed
  53. Puliaev, R, Puliaeva, I, Welniak, LA, et al. CTL-promoting effects of CD40 stimulation outweigh B cell-stimulatory effects resulting in B cell elimination and disease improvement in a murine model of lupus. J Immunol, 2008;181;47-61.
    Pubmed
  54. Shulman, HM, Kleiner, D, Lee, SJ, et al. Histopathologic diagnosis of chronic graft-versus-host disease: National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: II. Pathology Working Group Report. Biol Blood Marrow Transplant, 2006;12;31-47.
    Pubmed
  55. Wynn, TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol, 2004;4;583-594.
    Pubmed
  56. Nikolic, B, Lee, S, Bronson, RT, Grusby, MJ, Sykes, M. Th1 and Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets. J Clin Invest, 2000;105;1289-1298.
    Pubmed
  57. Hillebrandt, S, Wasmuth, HE, Weiskirchen, R, et al. Complement factor 5 is a quantitative trait gene that modifies liver fibrogenesis in mice and humans. Nat Genet, 2005;37;835-843.
    Pubmed
  58. Niculescu, F, Niculescu, T, Nguyen, P, et al. Both apoptosis and complement membrane attack complex deposition are major features of murine acute graft-vs.-host disease. Exp Mol Pathol, 2005;79;136-145.
    Pubmed
  59. Tsoi, MS, Storb, R, Jones, E, et al. Deposition of IgM and complement at the dermoepidermal junction in acute and chronic cutaneous graft-vs-host disease in man. J Immunol, 1978;120;1485-1492.
    Pubmed
  60. Liem, LM, Fibbe, WE, van Houwelingen, HC, Goulmy, E. Serum transforming growth factor-beta1 levels in bone marrow transplant recipients correlate with blood cell counts and chronic graft-versus-host disease. Transplantation, 1999;67;59-65.
    Pubmed
  61. Baron, C, Somogyi, R, Greller, LD, et al. Prediction of graft-versus-host disease in humans by donor gene-expression profiling. PLoS Med, 2007;4;e23.
    Pubmed
  62. Jaffee, BD, Claman, HN. Chronic graft-versus-host disease (GVHD) as a model for scleroderma. I. Description of model systems. Cell Immunol, 1983;77;1-12.
    Pubmed
  63. Allen, RD, Staley, TA, Sidman, CL. Differential cytokine expression in acute and chronic murine graft-versus-host-disease. Eur J Immunol, 1993;23;333-337.
    Pubmed
  64. De Wit, D, Van Mechelen, M, Zanin, C, et al. Preferential activation of Th2 cells in chronic graft-versus-host reaction. J Immunol, 1993;150;361-366.
    Pubmed
  65. Doutrelepont, JM, Moser, M, Leo, O, et al. Hyper IgE in stimulatory graft-versus-host disease: role of interleukin-4. Clin Exp Immunol, 1991;83;133-136.
    Pubmed
  66. Garlisi, CG, Pennline, KJ, Smith, SR, Siegel, MI, Umland, SP. Cytokine gene expression in mice undergoing chronic graft-versus-host disease. Mol Immunol, 1993;30;669-677.
    Pubmed
  67. Tanaka, J, Imamura, M, Kasai, M, et al. The important balance between cytokines derived from type 1 and type 2 helper T cells in the control of graft-versus-host disease. Bone Marrow Transplant, 1997;19;571-576.
    Pubmed
  68. Umland, SP, Razac, S, Nahrebne, DK, Seymour, BW. Effects of in vivo administration of interferon (IFN)-gamma, anti-IFN-gamma, or anti-interleukin-4 monoclonal antibodies in chronic autoimmune graft-versus-host disease. Clin Immunol Immunopathol, 1992;63;66-73.
    Pubmed
  69. Wynn, TA, Cheever, AW, Jankovic, D, et al. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature, 1995;376;594-596.
    Pubmed
  70. Hoffmann, KF, Cheever, AW, Wynn, TA. IL-10 and the dangers of immune polarization: excessive type 1 and type 2 cytokine responses induce distinct forms of lethal immunopathology in murine schistosomiasis. J Immunol, 2000;164;6406-6416.
    Pubmed
  71. Murata, T, Husain, SR, Mohri, H, Puri, RK. Two different IL-13 receptor chains are expressed in normal human skin fibroblasts, and IL-4 and IL-13 mediate signal transduction through a common pathway. Int Immunol, 1998;10;1103-1110.
    Pubmed
  72. Chiaramonte, MG, Donaldson, DD, Cheever, AW, Wynn, TA. An IL-13 inhibitor blocks the development of hepatic fibrosis during a T-helper type 2-dominated inflammatory response. J Clin Invest, 1999;104;777-785.
    Pubmed
  73. Oriente, A, Fedarko, NS, Pacocha, SE, Huang, SK, Lichtenstein, LM, Essayan, DM. Interleukin-13 modulates collagen homeostasis in human skin and keloid fibroblasts. J Pharmacol Exp Ther, 2000;292;988-994.
    Pubmed
  74. Saito, A, Okazaki, H, Sugawara, I, Yamamoto, K, Takizawa, H. Potential action of IL-4 and IL-13 as fibrogenic factors on lung fibroblasts in vitro. Int Arch Allergy Immunol, 2003;132;168-176.
    Pubmed
  75. Jacobsohn, DA, Schechter, T, Seshadri, R, Thormann, K, Duerst, R, Kletzel, M. Eosinophilia correlates with the presence or development of chronic graft-versus-host disease in children. Transplantation, 2004;77;1096-1100.
    Pubmed
  76. Atamas, SP, White, B. The role of chemokines in the pathogenesis of scleroderma. Curr Opin Rheumatol, 2003;15;772-777.
    Pubmed
  77. Zhou, L, Askew, D, Wu, C, Gilliam, AC. Cutaneous gene expression by DNA microarray in murine sclerodermatous graft-versus-host disease, a model for human scleroderma. J Invest Dermatol, 2007;127;281-292.
    Pubmed
  78. Yoon, HK, Lim, JY, Kim, TJ, Cho, CS, Min, CK. Effects of pravastatin on murine chronic graft-versus-host disease. Transplantation, 2010;90;853-860.
    Pubmed
  79. Morris, SC, Cheek, RL, Cohen, PL, Eisenberg, RA. Allotype-specific immunoregulation of autoantibody production by host B cells in chronic graft-versus host disease. J Immunol, 1990;144;916-922.
    Pubmed
  80. van der Veen, FM, Rolink, AG, Gleichmann, E. Autoimmune disease strongly resembling systemic lupus erythematosus (SLE) in F1 mice undergoing graft-versus-host reaction (GVHR). Adv Exp Med Biol, 1982;149;669-677.
    Pubmed
  81. Sekiguchi, DR, Eisenberg, RA, Weigert, M. Secondary heavy chain rearrangement: a mechanism for generating anti-double-stranded DNA B cells. J Exp Med, 2003;197;27-39.
    Pubmed
  82. Sekiguchi, DR, Jainandunsing, SM, Fields, ML, et al. Chronic graft-versus-host in Ig knockin transgenic mice abrogates B cell tolerance in anti-double-stranded DNA B cells. J Immunol, 2002;168;4142-4153.
    Pubmed
  83. Via, CS, Shearer, GM. T-cell interactions in autoimmunity: insights from a murine model of graft-versus-host disease. Immunol Today, 1988;9;207-213.
    Pubmed
  84. Parkman, R. Clonal analysis of murine graft-vs-host disease. I. Phenotypic and functional analysis of T lymphocyte clones. J Immunol, 1986;136;3543-3548.
    Pubmed
  85. Claman, HN, Jaffee, BD, Huff, JC, Clark, RA. Chronic graft-versus-host disease as a model for scleroderma. II. Mast cell depletion with deposition of immunoglobulins in the skin and fibrosis. Cell Immunol, 1985;94;73-84.
    Pubmed
  86. Ruzek, MC, Jha, S, Ledbetter, S, Richards, SM, Garman, RD. A modified model of graft-versus-host-induced systemic sclerosis (scleroderma) exhibits all major aspects of the human disease. Arthritis Rheum, 2004;50;1319-1331.
    Pubmed
  87. Anderson, BE, McNiff, J, Yan, J, et al. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest, 2003;112;101-108.
    Pubmed
  88. McCormick, LL, Zhang, Y, Tootell, E, Gilliam, AC. Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J Immunol, 1999;163;5693-5699.
    Pubmed
  89. Zhang, Y, McCormick, LL, Desai, SR, Wu, C, Gilliam, AC. Murine sclerodermatous graft-versus-host disease, a model for human scleroderma: cutaneous cytokines, chemokines, and immune cell activation. J Immunol, 2002;168;3088-3098.
    Pubmed
  90. Rouquette-Gally, AM, Boyeldieu, D, Prost, AC, Gluckman, E. Autoimmunity after allogeneic bone marrow transplantation. A study of 53 long-term-surviving patients. Transplantation, 1988;46;238-240.
    Pubmed
Blood Res
Volume 59 2024

Stats or Metrics

Share this article on

  • line

Related articles in BR

Blood Research

pISSN 2287-979X
eISSN 2288-0011
qr-code Download