throbber
Pathogenesis and
`Management of
`Graft-versus-Host
`Disease
`
`Sung W. Choi, MDa,*, John E. Levine, MDb,
`James L.M. Ferrara, MD, DScc
`
`KEYWORDS
` Acute graft-versus-host disease
` Chronic graft-versus-host disease
` Hematopoietic cell transplantation  GVHD
`
`Allogeneic hematopoietic cell transplantation (HCT) is an important therapeutic option
`for various malignant and nonmalignant conditions.1 The indication for its use has
`expanded, especially among older patients, in recent years through novel strategies
`using donor leukocyte infusions, nonmyeloablative conditioning and umbilical cord
`blood (UCB) transplantation.2 As allogeneic HCT continues to increase, with more
`than 20,000 allogeneic transplantations performed annually worldwide, greater
`attention is given to improvements in supportive care, infectious prophylaxis, immuno-
`suppressive medications, and DNA-based tissue typing. Despite advances, graft-
`versus-host disease (GVHD) remains the most frequent and serious complication
`following allogeneic HCT and limits the broader application of this important therapy.3
`GVHD can be considered an exaggerated manifestation of a normal inflammatory
`mechanism in which donor lymphocytes encounter foreign antigens in a milieu that
`fosters inflammation. In the context of hematological malignancies, a delicate balance
`exists between the harmful consequences of GVHD and the beneficial effects incurred
`
`Dr Choi is a St Baldrick’s Career Development Scholar.
`Dr Ferrara is an American Cancer Society Clinical Research Professor.
`a Department of Pediatrics, Blood and Marrow Transplant Program, University of Michigan
`Medical School, 1500 E. Medical Center Drive, 6303 Comprehensive Cancer Center, Ann Arbor,
`MI 48109-5942, USA
`b Department of Internal Medicine and Pediatrics, Blood and Marrow Transplant Program,
`University of Michigan Medical School, 1500 E. Medical Center Drive, 5303 Comprehensive
`Cancer Center, Ann Arbor, MI 48109-5941, USA
`c Department of Internal Medicine and Pediatrics, Blood and Marrow Transplant Program,
`University of Michigan Medical School, 1500 E. Medical Center Drive, 6303 Comprehensive
`Cancer Center, Ann Arbor, MI 48109-5942, USA
`* Corresponding author.
`E-mail address: sungchoi@med.umich.edu (S.W. Choi).
`
`Immunol Allergy Clin N Am 30 (2010) 75–101
`doi:10.1016/j.iac.2009.10.001
`immunology.theclinics.com
`0889-8561/10/$ – see front matter ª 2010 Elsevier Inc. All rights reserved.
`
`

`

`76
`
`Choi et al
`
`when donor lymphocytes attack recipient malignant cells, a process referred to as the
`graft-versus-leukemia/tumor (GVL) effect. Given the increasing number of transplant
`recipients, there will be an increasing population of patients with GVHD. Recent
`advances in the understanding of the pathogenesis of GVHD have led to new
`approaches to its management, including using it to preserve the GVL effect following
`allogeneic transplant. This article reviews the important elements in the complex
`immunologic interactions involving cytokine networks, chemokine gradients, and
`the direct mediators of cellular cytotoxicity that cause clinical GVHD, and discusses
`the risk factors and strategies for management of GVHD.
`
`ACUTE GVHD
`Epidemiology and Risk Factors
`
`In 1966, Billingham4 formulated three requirements for the development of GVHD:
`the graft must contain immunologically competent cells; the recipient must express
`tissue antigens that are not present in the transplant donor; and the recipient must be
`incapable of mounting an effective response to eliminate the transplanted cells. It is
`now known that T cells are the immunologically competent cells, and when tissues
`containing T cells (blood products, bone marrow [BM], and solid organs) are trans-
`ferred from one person to another who is unable to eliminate those cells, GVHD
`can develop.5,6
`Allogeneic HCT is the most common setting for the development of GVHD, in which
`recipients receive immunoablative chemotherapy or radiation before hematopoietic
`cell infusion containing donor T cells. GVHD ultimately develops when donor T cells
`respond to recipient tissue antigens secondary to mismatches between major or
`minor histocompatibility antigens between the donor and recipient. The major histo-
`compatibility complex (MHC) contains the genes that encode tissue antigens. In hu-
`mans, the MHC region lies on the short arm of chromosome 6 and is called the
`human leukocyte antigen (HLA) region.7 Class I HLA (A, B, and C) proteins are ex-
`pressed on almost all nucleated cells of the body at varying densities. Class II (DR,
`DQ, and DP) proteins are primarily expressed on hematopoietic cells (B cells, dendritic
`cells, monocytes, and activated T cells), but their expression can be induced on many
`other cell types following inflammation or injury. High-resolution DNA typing of HLA
`genes with polymerase chain reaction (PCR)-based techniques has now largely
`replaced earlier methods. The incidence of GVHD is directly related to HLA disparity8,9
`and with more HLA mismatches, the likelihood of developing GVHD increases.10,11
`Recent data from the National Marrow Donor Program (NMDP) suggest that high-
`resolution matching for HLA-A, -B, -C, and -DRBI (8/8 match) maximizes post trans-
`plant survival.12,13
`Despite HLA identity between a patient and donor, the incidence of acute GVHD
`ranges from 26% to 32% in recipients of sibling donor grafts, and 42% to 52% in
`recipients of unrelated donor grafts (Center for International Blood and Marrow Trans-
`plant Research [CIBMTR] Progress Report January–December 2008). The incidence is
`likely related to genetic differences that lie outside the HLA loci, or ‘‘minor’’ histocom-
`patibility antigens (HA), which are immunogenic peptides derived from polymorphic
`proteins presented on the cell surface by MHC molecules.14 Some minor HAs, such
`as HY and HA-3, are expressed on all tissues and are targets for GVHD and GVL,
`whereas other minor HAs, such as HA-1 and HA-2, are expressed abundantly on
`hematopoietic cells (including leukemic cells) and may induce a greater GVL effect
`with less GVHD.14,15 However, the precise elucidation of most human minor antigens
`remains to be accomplished.14,16
`
`

`

`Management of Graft-versus-Host Disease
`
`77
`
`The impact of donor and recipient polymorphisms in cytokine genes with critical
`roles in the classic ‘‘cytokine storm’’ of GVHD has been examined as a risk factor
`for GVHD.17 Various polymorphic genes, including tumor necrosis factor a (TNF-a),
`interleukin 10 (IL-10), and interferon-g (IFN-g) variants, have been associated with
`GVHD, although not always.18–20 There is no unequivocal evidence that polymorphic
`genes for cytokines or other proteins involved in innate immunity 21–24 sufficiently influ-
`ence GVHD and transplant outcome to change clinical practice. Nonetheless, future
`strategies to identify the best possible transplant donor will likely incorporate HLA
`and non-HLA genetic factors.
`In addition to genetic factors, other risk factors which have been associated with the
`development of GVHD include older donor and recipient age,25–28 multiparous female
`donor,28,29 advanced malignant condition at transplantation,9,29 donor type,28 and
`donor hematopoeitic cell source.30–32 In the last decade, there has been a shift in
`clinical practice from the use of intraoperative harvested BM to granulocyte colony-
`stimulating factor mobilized peripheral blood stem cells (PBSC) as the donor hemato-
`poietic cell source. However, definitive data demonstrating long-term advantages of
`PBSC rather than BM are lacking. One meta-analysis found that acute and chronic
`GVHD are more common following peripheral blood stem cell transplant (PBSCT)
`compared with bone marrow transplant (BMT) and indicated a trend toward decreased
`relapse rate following PBSCT.31 The relative risk (RR) for acute GVHD after PBSCT was
`1.16 (95% confidence interval [CI], 1.04–1.28) compared with BMT; the RR for chronic
`GVHD after PBSCT was 1.53 (95% CI, 1.25–2.05); and the RR for relapse after PBSCT
`was 0.81 (95% CI, 0.62–1.05). Thus, the survival benefit of PBSC versus BM remains in
`question. A large prospective, randomized, multicenter clinical trial of PBSC versus BM
`in unrelated donor transplantation conducted through the Blood and Marrow Trans-
`plant Clinical Trials Network (BMT CTN) has recently finished accrual.
`For individuals without a suitable HLA-matched donor, UCB has become an alter-
`native to BM or PBSC.33–36 The incidence and severity of acute GVHD seem to be
`lower following UCB transplant than after HLA-matched marrow unrelated donor
`transplant, despite HLA disparities between the donor and recipient.37,38 In an effort
`to meet the minimum cell dose required to ensure reliable engraftment, the simulta-
`neous transplantation of 2 partially HLA-matched UCB units has been studied.39 A
`recent report comparing transplantation with 2 partially HLA-matched UCB units
`versus a single unit demonstrated an increased incidence and earlier presentation
`of acute GVHD associated with the double UCB graft.40
`
`Prevention
`
`Prevention of acute GVHD is an integral component to the management of patients
`undergoing allogeneic HCT. The primary strategy employed is in the use of pharma-
`cologic GVHD prophylaxis. The most widely used GVHD prophylaxis following full
`intensity conditioning includes a combination of a calcineurin inhibitor (eg, cyclo-
`sporine, tacrolimus) with methotrexate (MTX). This standard regimen was initially
`described in 198641 and since then several clinical trials have shown the superiority
`of this combination in reducing the incidence of GVHD and improving survival
`compared with either agent alone.42–45 The calcineurin inhibitors cyclosporine and ta-
`crolimus impede the function of the cytoplasmic enzyme calcineurin, which is critical
`to the activation of T cells. The most common side effects include hypomagnesemia,
`hyperkalemia, hypertension, and nephrotoxicity.46 Large randomized studies
`comparing tacrolimus-MTX with cyclosporine-MTX have demonstrated a reduced
`incidence of grade II to IV acute GVHD with tacrolimus, but no overall survival
`advantage.43,46
`
`

`

`78
`
`Choi et al
`
`Recently, sirolimus, a widely used immunosuppressant in solid organ transplanta-
`tion,47 has become attractive as a GVHD prophylactic agent because of the nonover-
`lapping toxicities with calcineurin inhibitors and the different mechanism of action.
`Sirolimus binds uniquely to FK binding protein 12 (FKBP12) and then complexes
`with mammalian Target of Rapamycin (mTOR).48 Several studies have shown that
`the combination of sirolimus and tacrolimus has resulted in rapid engraftment, low
`incidence of acute GVHD,
`reduced transplant-related toxicity, and improved
`survival.49 A prospective, randomized, multicenter trial is being conducted through
`the BMT CTN (protocol 0402) comparing sirolimus-tacrolimus versus tacrolimus-
`MTX following HLA-matched, related donor PBSCT.
`A commonly used GVHD prophylaxis following reduced-intensity conditioning
`includes a combination of a calcineurin inhibitor (eg, cyclosporine, tacrolimus) with
`mycophenolate mofetil (MMF) instead of MTX. MMF, the prodrug of mycophenolic
`acid, selectively inhibits inosine monophosphate dehydrogenase, an enzyme critical
`to the de novo synthesis of guanosine nucleotide, which is needed for proliferation
`of T cells. In a prospective randomized trial, patients who received MMF as part of
`GVHD prophylaxis experienced significantly less severe mucositis and more rapid
`neutrophil engraftment than those who received MTX.50 Although the optimal prophy-
`laxis regimen following reduced-intensity HCT is not well established, MMF has been
`shown to be safe in this context.51–55 MMF is also often preferred to MTX in UCB trans-
`plants because of its advantageous toxicity profile with respect to neutropenia and
`mucositis.
`Many centers have previously attempted to decrease the risk of GVHD by ex vivo
`T cell depletion. Despite significant reductions in the incidence and severity of
`GVHD, T cell depletion has not achieved wide acceptance because of high rates of
`graft rejection, life-threatening infections, and leukemia relapse.56–58 In vivo T cell
`depletion has also been widely studied using alemtuzumab, a monoclonal antibody
`specific for CD52 antigen expressed abundantly on the surface of normal and malig-
`nant lymphocytes,59,60 or antithymocyte globulin (ATG), a polyclonal antibody mixture
`of either horse or rabbit origin directed against multiple epitopes of human T cells.61
`These approaches are associated with significant reduction in acute GVHD, but at
`the cost of impaired immune reconstitution and increased risk of leukemia relapse.
`Thus, the focus of most prevention strategies remains pharmacologic manipulation
`of T cells following transplant.
`
`Pathophysiology
`
`Acute GVHD is mediated by donor lymphocytes infused into the recipient, in whom
`they encounter profoundly damaged tissues from the effects of the underlying
`disease, prior infections, and the transplant conditioning regimen. The allogeneic
`donor cells encounter a foreign environment that has been altered to promote the acti-
`vation and proliferation of inflammatory cells. Thus, acute GVHD reflects an exagger-
`ated response of the normal inflammatory mechanisms that involves donor T cells and
`multiple innate and adaptive cells and mediators. Three sequential phases can be
`conceptualized to illustrate the complex cellular interactions and inflammatory
`cascades that ultimately evolve to acute GVHD: (1) activation of antigen-presenting
`cells (APCs); (2) donor T cell activation, proliferation, differentiation and migration;
`and (3) target tissue destruction.62
`
`Phase 1: activation of APCs
`In the first phase, APCs are activated by the underlying disease and the HCT
`conditioning regimen.63 Animal models 63,64 and clinical HCT 65 have supported the
`
`

`

`Management of Graft-versus-Host Disease
`
`79
`
`observation that increased risk of GVHD is associated with intensive conditioning
`regimens that contribute to extensive tissue injury and subsequent release of inflam-
`matory cytokines. Damage to host tissues leads to the secretion of proinflammatory
`cytokines, such as TNF-a and IL-1, and chemokines, such as CCL2-5 and CXCL9-
`11, thereby producing increased expression of adhesion molecules, MHC antigens
`and costimulatory molecules on host APCs. For example,
`increase of plasma
`TNF-a receptor 1 levels, a surrogate marker for TNF-a, at 1 week after HCT strongly
`correlates with the later development of GVHD.66 Systemic translocation of immunos-
`timulatory microbial products, such as lipopolysaccharide (LPS), as a result of damage
`to the gastrointestinal (GI) tract induced by the conditioning regimen, enhances the
`activation of host APCs.67,68 The initial site of interaction between activated APCs
`and donor T cells is likely the secondary lymphoid tissue in the GI tract.69 Different
`distinct subsets of APCs, including host and donor type APCs,68,70,71 dendritic
`cells,72,73 Langerhans cells,74 and monocytes/macrophages,75 have been implicated
`in this phase. However, the relative contributions of these various APCs remain to be
`elucidated. The intensity of the conditioning regimen and the degree of tissue injury
`seem to be associated with the risk of GVHD. Reduced intensity conditioning
`regimens have thus become more widely employed in an effort to reduce acute
`GVHD by decreasing the damage to host tissues.65,76
`
`Phase 2: donor T cell activation
`Donor T cell activation, proliferation, differentiation, and migration in response to
`primed APCs occur during the second phase of acute GVHD. The T cell receptors
`(TCR) of donor T cells recognize alloantigens on host and donor type APCs that are
`present in secondary lymphoid organs.77,78 During direct presentation, donor T cells
`recognize either the peptide bound to host MHC molecules, or the foreign MHC mole-
`cules themselves.79 During indirect presentation, donor T cells respond to the
`peptides generated by degradation of the host MHC molecules that are presented
`on donor-derived MHC.80
`Following antigen recognition, signaling through the TCR induces a conformational
`change in adhesion molecules, resulting in high affinity binding to the APC.81 The
`complex interaction between T cell costimulatory molecules and their ligands on
`APCs facilitates full T cell activation. Many T cell costimulatory molecules display unique
`and overlapping functions.82 Receptors of the B7 family and the TNF family play espe-
`cially critical roles in GVHD, and are known to deliver positive and negative signals to T
`cells.83 Blockade of costimulatory and inhibitory pathways can reduce acute GVHD in
`murine models, but this approach has not yet been tested in clinical trials.2
`Murine studies have shown that control of alloreactive responses responsible for
`GVHD depends at least in part on CD41 CD251 regulatory T cells. Studies in mice
`suggest that regulatory T cells added to donor grafts can prevent or delay GVHD.84
`However, the role of regulatory T cells in clinical allogeneic HCT has not been well
`established, in part because of the lack of clear identification of human regulatory T
`cell phenotype. In contrast to murine studies, more severe acute GVHD developed
`clinically when donor grafts contained larger numbers of donor CD41 CD251 T
`cells.85 One recent study found that HCT recipients with higher absolute numbers of
`FOXP31 CD41 T cells were associated with a reduced risk of developing GVHD.86
`However, FOXP3 expression in humans is not specific for regulatory T cell pheno-
`type,87 and improved techniques to identify and expand regulatory T cells are required
`for its wider application in clinical BMT.
`Several intracellular biochemical pathways are rapidly amplified following T cell acti-
`vation. Activated T cells secrete cytokines that are generally classified as Th1 (IFN-g,
`
`

`

`80
`
`Choi et al
`
`IL-2 and TNF-a) or Th2 (IL-4, IL-5, IL-10, and IL-13) and Th17. Although Th1 cytokines
`induce GVHD efficiently, the balance of Th1 and Th2 cytokines is important in the im-
`munopathogenesis of GVHD, but remains incompletely understood.88 TNF-a, already
`discussed as an inducer of APC activation in phase I,
`functions to amplify
`T cell activation and proliferation in this second phase of GVHD. IL-2 production is
`also integral in this early development of acute GVHD,89 and remains the principal
`target of many current clinical therapeutic and prophylactic approaches to GVHD,
`such as cyclosporine, tacrolimus, and monoclonal antibodies against IL-2 and its
`receptor.44 However, recent data suggest that regulatory T cell function is dependent
`on the presence of calcineurin-dependent IL-2, and that interference with IL-2 may
`possibly antagonize the induction of tolerance.90
`IFN-g is a cytokine with diverse effects in vivo that can amplify and suppress acute
`GVHD. With regards to amplification, IFN-g increases the expression of molecules
`such as chemokine receptors, MHC proteins, and adhesion molecules. IFN-g also
`sensitizes monocytes and macrophages to stimuli such as LPS, thereby accelerating
`intracellular cascades in response to these stimuli.88 IFN-g may also amplify GVHD by
`directly damaging target cells in the GI tract and in the skin while conversely inducing
`nitric oxide-mediated immunosuppression.91 IFN-g itself can prevent experimental
`GVHD by hastening the apoptosis of activated donor T cells.92 Thus, this complexity
`makes it a challenging target with respect to therapeutic intervention.
`IL-10 can suppress the expression of proinflammatory cytokines, chemokines, and
`adhesion molecules, and antigen-presenting and costimulatory molecules in mono-
`cytes/macrophages, neutrophils, and T cells.93 Recent clinical data suggest that
`genetic polymorphism in the IL-10 promoter region of the recipient has a significant
`impact on the risk of developing acute GVHD.94 Increased IL-10 production by recip-
`ient cells stimulated ex vivo has been associated with a reduced risk of acute GVHD.95
`Experimental data have also demonstrated that transforming growth factor b (TGF-b),
`another suppressive cytokine, attenuates acute GVHD, but may lead to chronic
`GVHD.96 Thus multiple cytokines are important in GVHD pathogenesis and regulation.
`Furthermore, the timing and duration of cytokine expression may be a critical factor
`determining the induction of the GVH reaction, and cytokine dysregulation could
`potentially contribute to the severity of acute GVHD.62
`
`Phase 3: cellular and inflammatory effector phase
`A complex cascade of cellular and inflammatory mediators occurs during the effector
`phase of acute GVHD. These mediators synergize to amplify local tissue injury and
`damage target tissues. As such, the effector phase of GVHD involves aspects of the
`innate and adaptive immune response and interactions with the proinflammatory cells
`and cytokines generated during phase 1 and 2.
`
`Cellular effectors Cytotoxic T cells are the major cellular effectors of acute GVHD and
`lyse target cells using principally the Fas/Fas ligand (FasL) and the perforin/granzyme
`pathways.97 The Fas-FasL pathway seems to predominate in hepatic GVHD whereas
`the perforin/granzyme pathways are more important in the GI tract and skin.2 The
`‘‘danger’’ signals generated in phase 1, augmented by the expression of costimulatory
`molecules in phase 2, induce the production of chemokines. The migration of donor
`T cells from lymphoid tissues to the target organs in which damage occurs is directed
`by these chemokine gradients and adhesion molecules such as L-selectin and aLb2
`integrin. The upregulation of chemokines, such as CCL2, CCL3, CCL4, CCL5,
`CXCL9, CXCL10, and CXCL11 in GVHD target organs (skin, liver, and gut), play
`primary roles in this homing process.98 Furthermore, integrin receptors, which are
`
`

`

`Management of Graft-versus-Host Disease
`
`81
`
`regulated by chemokines, mediate many adhesive interactions that are critical for
`successful T cell migration.99
`
`Inflammatory effectors The secretion of inflammatory cytokines, such as TNF-a or IL-1
`also results in GVHD target organ injury. The secretion signal may be provided through
`toll-like receptors (TLRs) by LPS and other microbial products that have leaked through
`damaged intestinal mucosa by the conditioning regimen in phase 1.100 TNF-a is
`produced by donor and host cells and is a critical component of GVHD pathophysi-
`ology. TNF-a can (1) activate APCs and enhance alloantigen presentation, (2) recruit
`effector cells to target organs through the induction of inflammatory chemokines, and
`(3) directly cause tissue damage via apoptosis and necrosis. IL-1 is also involved in
`the pathogenesis of acute GVHD.17 Its secretion occurs predominantly in the spleen
`and skin during the effector phase of experimental GVHD.101 Increased expression of
`mononuclear IL-1 mRNA has been associated with the development of GVHD.102
`However, the blockade of IL-1 using recombinant human IL-1 receptor antagonist in
`patients undergoing HCT did not reduce the risk of GVHD.103
`
`Clinical Features
`
`The three main organs involved in acute GVHD are the skin, liver, or GI tract. GVHD
`presents clinically in an acute or chronic form. Historically, the acute and chronic forms
`were arbitrarily defined based on the timeframe post transplant (less than or more than
`100 days, respectively).104 However, current consensus is that clinical manifestations
`guide whether the signs and symptoms of GVHD are acute, chronic, or an overlap
`syndrome (wherein diagnostic or distinctive features of acute and chronic GVHD
`appear together).105
`The extent (stage) of involvement of the three principal target organs determines the
`overall severity (grade) of acute GVHD (Table 1).106 The overall grades are classified as
`I (mild), II (moderate), III (severe), and IV (very severe). A poor prognosis is associated
`with severe grade III or IV GVHD, with 25% and 5% overall survival, respectively.107
`Skin is generally the first and most commonly affected organ.104 The presentation of
`skin involvement generally coincides with donor cell engraftment and is characterized
`by an erythematous, maculopapular rash that is often pruritic. In severe cases the skin
`may blister and ulcerate.108 The pathognomonic histopathologic finding is apoptosis
`at the base of epidermal rete ridges. Other features include dyskeratosis, exocytosis
`of lymphocytes, satellite lymphocytes adjacent to dyskeratotic epidermal keratino-
`cytes, and a perivascular lymphocytic infiltration in the dermis.1,109
`Liver GVHD can be a challenging diagnosis. The initial feature typically includes the
`development of jaundice or an increase in the alkaline phosphatase and bilirubin;
`hepatomegaly may be noted. However, it is often difficult to distinguish from other
`causes of liver dysfunction following allogeneic HCT, such as drug toxicity, venoocclu-
`sive disease, opportunistic (bacterial, viral, and fungal) infections, total parenteral
`nutrition, acalculous cholecystitis, and iron overload, because of the overlapping
`patterns of clinical history, physical examination, and laboratory and imaging data.
`Thus, a biopsy is often required to confirm the diagnosis of liver GVHD.110 The histo-
`logic features are endothelialitis, lymphocytic infiltration of the portal areas, pericho-
`langitis, and bile duct destruction.111 However, the increased risk of bleeding
`associated with thrombocytopenia in the immediate post transplant period can
`preclude obtaining a biopsy. As such, the diagnosis of liver GVHD is often a clinical
`diagnosis of exclusion.
`GI tract involvement of acute GVHD may present as nausea, vomiting, anorexia,
`diarrhea, or abdominal pain. It is a panintestinal process with focal lesions of varying
`
`

`

`82
`
`Choi et al
`
`Table 1
`Acute GVHD grading criteria
`
`Stage
`
`Skin
`
`Liver (Bilirubin)
`
`GI Tract (Stool Output/d)
`
`0
`
`1
`
`2
`
`3
`
`4
`
`No GVHD rash
`
`<2 mg/dL
`
`Maculopapular rash
`<25% BSA
`
`2–3 mg/dL
`
`Maculopapular rash
`25%–50% BSA
`
`Maculopapular rash
`>50% BSA
`
`Generalized erythroderma
`with bullous formation
`
`3.1–6 mg/dL
`
`6.1–15 mg/dL
`
`>15 mg/dL
`
`Adult: <500 mL/d
`Child: <10 mL kg/d
`
`Adult: 500–999 mL/d
`Child: 10–19.9 mL/kg/d or
`persistent nausea,
`vomiting, or anorexia,
`with a positive upper GI
`biopsy
`
`Adult: 1000–1500 mL/d
`Child: 20–30 mL/kg/d
`
`Adult: >1500 mL/d
`Child: >30 mL/kg/d
`
`Severe abdominal pain
`with or without ileus
`
`Overall clinical grade: grade 0, no stage 1–4 of any organ; 1, stage 1–2 skin rash and no liver or GI
`involvement; 2, stage 3 skin rash, or stage 1 liver involvement, or stage 1 GI involvement; 3, stage
`0–3 skin rash, with stage 2–3 liver involvement, or stage 2–3 GI involvement; 4, stage 4 skin rash,
`liver, or GI involvement.
`The standard GVHD grading criteria were developed by Glucksberg in 1974. (Data from
`Glucksberg H, Storb R, Fefer A, et al. Clinical manifestations of graft-versus-host disease in human
`recipients of marrow from HL-A-matched sibling donors. Transplantation 1974;18(4):295–304) and
`then revised at the 1994 Consensus Conference in Keystone. (Data from Pzrepiorka D, Weisdorf D,
`Martin P, et al. Consensus conference on acute GVHD grading. Bone Marrow Transpl 1995;15:825–8.)
`The grading system did not initially take into consideration the staging of the GI tract for pediatric
`patients. However, most pediatric centers have adopted the modified Glucksberg grading scale and
`adjusted the staging of the GI tract to be based on volume per kg of body weight.
`Abbreviation: BSA, body surface area.
`
`is not always
`intensity. Gastric involvement causes postprandial vomiting that
`preceded by nausea. The diarrhea of GVHD is secretory and may be accompanied
`by significant GI bleeding as a result of mucosal ulceration, which is a prognostic
`factor for poor outcome.112 In advanced disease, diffuse, severe abdominal pain
`and distension are accompanied by voluminous diarrhea. The histologic features
`include patchy ulcerations, apoptotic bodies in the base of crypts, crypt abscesses,
`and loss and flattening of the epithelium surface.113
`
`Diagnostic Approach
`
`The diagnosis of acute GVHD is based entirely on clinical criteria that can be
`confirmed by biopsy of one of the three target organs. Laboratory data or imaging
`studies are also useful tests in the diagnostic approach to GVHD. However, a major
`challenge with the diagnosis is the absence of laboratory tests that can reliably predict
`or screen for the condition before its onset: that can establish a diagnosis in real time;
`that can distinguish it from other conditions that present with similar symptoms, such
`as infection; or that can stratify patients according to response to therapy. Thus,
`experimental blood tests with predictive value for GVHD such as a 4-biomarker
`panel114,115 may ultimately be useful to further identify high-risk groups and their
`outcomes (Fig. 1). These biomarkers could result in immunosuppressive treatment
`schemas tailored to patients in several risk groups.
`
`

`

`Management of Graft-versus-Host Disease
`
`83
`
`GVHD+
`
`•
`
`-■
`•
`
`■
`
`■ -
`
`I I. I -I •
`- ■- ■I .. • ■
`■ -- • •
`L ••
`'I ■ -•
`
`LJ
`
`.J
`
`•
`
`■
`■
`Fold-change from the mean
`
`1/4
`
`1/2
`
`2
`
`4
`
`Composite
`
`- .,
`
`I I
`
`L
`
`•
`
`0.017
`0.018
`0.035
`0.07
`0.12
`0.13
`
`• 0.17
`•
`-r 0.37
`
`0.24
`0.27
`
`0.38
`0.80
`
`•
`• 0.0002
`• • 0.007
`
`0.0003
`0.0004
`
`Fold•
`change
`P-value
`from T-test GVHD +/.
`1.4E-07
`2.82
`5.9E-06
`5.44
`2.1 E-05
`4.45
`7.9E-05
`1.69
`2.33
`1.58
`2.53
`2.79
`1.42
`2.73
`1.34
`2.14
`1.30
`1.38
`0.76
`1.28
`1.38
`0.83
`1.20
`0.94
`
`A
`Protein
`IL-2Rn
`CRP
`IL-8
`ICAM-1
`TIMP-1
`TNFR1
`HGF
`CA19.9
`VEGF-O
`IL-12p70
`VCAM-1
`IL-10
`Anglostatin
`EotaXin
`IGF-1
`FGF-basic
`MMP-2
`Anglopoietin-2
`TGF-JI
`FasL
`
`B
`
`Q)
`
`..... cu
`
`0
`T"""
`
`<X)
`0
`
`(0
`0
`
`N
`0
`
`0::
`Q)
`>
`~ ·u5
`0 V
`a..
`0
`Q)
`=-i
`'-I-
`
`. : . . I
`~
`
`.
`
`r· -
`'
`
`I
`
`_.;
`
`HGF
`
`IL-2Ra_ . . <·?•~·
`.~·· · ""
`.·x'
`X
`
`_:_;
`,, ; ,
`
`IL-8
`
`TNFR1
`
`.: ,
`(
`r ···
`
`0
`0
`
`,·
`0.0
`
`0.2
`
`0.8
`
`1.0
`
`0.6
`0.4
`False Positive Rate
`Fig. 1. From discovery to validation of plasma biomarkers of acute GVHD. (A) The heatmap
`of proteins levels measured sequential enzyme-linked immunosorbent assay in the discovery
`set samples. Samples from 21 GVHD patients (left) and 21 GVHD1 patients (right) are rep-
`resented. Eleven proteins gave a P value for differences between GVHD1 and GVHD
`patient plasma <0.05. (B) The receiver-operating characteristic (ROC) curves of 4 individual
`discriminator proteins and the composite panel in the training set. Individual ROC curves
`for IL-2Ra, TNFR1, human growth factor, and IL-8 and the composite panel. (From Paczesny
`S, Krijanovski OI, Braun TM, et al. A biomarker panel for acute graft-versus-host disease.
`Blood 2009;113(2):273–8; with permission.)
`
`Treatment
`
`Despite routine treatment with calcineurin-based prophylaxis, GVHD remains a major
`complication of allogeneic HCT. The most important predictor of long-term survival in
`patients with acute GVHD is the primary response to the first line of treatment.116 Many
`centers treat mild skin GVHD (grade I) with topical corticosteroids alone, but for more
`severe skin GVHD or visceral GVHD involvement (grade II or greater), systemic
`
`

`

`84
`
`Choi et al
`
`corticosteroids are the mainstay of therapy, typically starting at 1 to 2 mg/kg/d.
`Durable responses occur in less than half of patients with grade II to IV GVHD,117
`and with increasing severity of the disease, the likelihood of response decreases.104
`Treatment with high-dose steroids often continues for 7 days or more, with a gradual
`dose reduction depending on the clinical response. In a recent retrospective analysis,
`low-dose (1 mg/kg/d) versus high-dose (2 mg/kg/d) prednisone was compared for
`initial treatment of acute GVHD. In patients with grades I to II GVHD, the nonrelapse
`mortality and overall survival were similar between regimens, with a reduced risk of
`invasive fungal
`infections and shorter hospitalizations in the low-dose prednisone
`group. The number of patients with grades III to IV at onset was too few to draw
`any significant conclusions.
`In addition to topical corticosteroid therapy for skin GVHD, nonabsorbed steroid
`therapy is commonly used in GI GVHD. Oral budesonide in combination with systemic
`corticosteroids in patients with grade II or higher acute GI GVHD has shown complete
`responses in 77% of patients compared with 32% of historical controls.118 In a more
`recent randomized, placebo-controlled trial of oral beclomethasone for GI GVHD, oral
`beclomethasone dipropionate (BDP) reduced GVHD flares following a prednisone
`taper and resulted in superior survival at 1 year post transplant.119 Intraarterial admin-
`istration of steroids for GI and hepatic GVHD has also been attempted to deliver
`steroids directly to the target organ.120
`Prolonged therapy with steroids invariab

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