Graft-versus-host disease.
Journal: 2009/May - The Lancet
ISSN: 1474-547X
Abstract:
Haemopoietic-cell transplantation (HCT) is an intensive therapy used to treat high-risk haematological malignant disorders and other life-threatening haematological and genetic diseases. The main complication of HCT is graft-versus-host disease (GVHD), an immunological disorder that affects many organ systems, including the gastrointestinal tract, liver, skin, and lungs. The number of patients with this complication continues to grow, and many return home from transplant centres after HCT requiring continued treatment with immunosuppressive drugs that increases their risks for serious infections and other complications. In this Seminar, we review our understanding of the risk factors and causes of GHVD, the cellular and cytokine networks implicated in its pathophysiology, and current strategies to prevent and treat the disease. We also summarise supportive-care measures that are essential for management of this medically fragile population.
Relations:
Content
Citations
(527)
References
(144)
Clinical trials
(2)
Diseases
(1)
Conditions
(2)
Chemicals
(3)
Organisms
(1)
Processes
(2)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Lancet 373(9674): 1550-1561

Graft-versus-Host Disease

Introduction

The number of allogeneic hematopoietic cell transplantations (HCT) continues to increase with more than 25,000 allogeneic transplantations performed annually. The graft-versus-leukemia / tumor (GVL) effect during allogeneic HCT effectively eradicates many hematological malignancies.1 The development of novel strategies that use donor leukocyte infusions, non-myeloablative conditioning and umbilical cord blood (UCB) transplantation have helped expand the indications for allogeneic HCT over the last several years, especially among older patients.2 Improvements in infectious prophylaxis, immunosuppressive medications, supportive care and DNA-based tissue typing have also contributed to improved outcomes after allogeneic HCT.1 Yet the major complication of allogeneic HCT, graft-versus-host disease (GVHD), remains lethal and limits the use of this important therapy.2 Given current trends, the number of transplants from unrelated donors is expected to double within the next five years, significantly increasing the population of patients with GVHD. In this seminar we review advances made in identifying the genetic risk factors and pathophysiology of this major HCT complication, as well as its prevention, diagnosis and treatment.

Etiology and Clinical Features

Fifty years ago Billingham 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.3 We know now that the immunologically competent cells are T cells, and that GVHD can develop in various clinical settings when tissues containing T cells (blood products, bone marrow, and solid organs) are transferred from one person to another who is not able to eliminate those cells.45 Patients, whose immune systems are suppressed, and who receive white blood cells from another individual, are at particularly high risk for GVHD.

GVHD occurs when donor T cells respond to genetically defined proteins on host cells. The most important proteins are Human Leukocyte Antigens (HLA)267, which are highly polymorphic and are encoded by the major histocompatibility complex (MHC). Class I HLA (A, B, and C) proteins are expressed on almost all nucleated cells of the body at varying densities. Class II proteins (DR, DQ, and DP) are primarily expressed on hematopoietic cells (B cells, dendritic cells, monocytes), 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 have now largely replaced earlier methods. The incidence of acute GVHD is directly related to the degree of mismatch between HLA proteins89 and thus ideally, donors and recipients are matched at HLA-A, -B, -C, and -DRB1, (“8/8 matches”), but mismatches may be tolerated for UCB grafts (see below).1012

Non-HLA Genetics

Despite HLA identity between a patient and donor, approximately 40% of patients receiving HLA-identical grafts develop acute GVHD due to genetic differences that lie outside the HLA loci, or “minor” histocompatibility antigens (HA). Some minor HAs, such as HY and HA-3, are expressed on all tissues and are targets for both GVHD and GVL.13 Other minor HAs, such as HA-1 and HA-2, are expressed most abundantly on hematopoietic cells (including leukemic cells) and may therefore induce a greater GVL effect with less GVHD.1314

Polymorphisms in both donors and recipients for cytokines that are involved in the classical `cytokine storm' of GVHD (discussed below) have been implicated as risk factors for GVHD.15 Tumor Necrosis Factor (TNF)-α, Interleukin 10 (IL-10), Interferon-γ (IFNγ) variants have correlated with GVHD in some, but not all, studies.1618 Genetic polymorphisms of proteins involved in innate immunity, such as nucleotide oligomerization domain 2 and Keratin 18 receptors, have also been associated with GVHD.1922 Future strategies to identify the best possible transplant donor will probably incorporate both HLA and non-HLA genetic factors.

Clinical Features of Acute GVHD

Based on an early Seattle experience, acute GVHD was defined to occur prior to day 100, whereas chronic GVHD occurred after that time.2325 This definition is far from satisfactory, and a recent National Institutes of Health classification includes late-onset acute GVHD (after day 100) and an overlap syndrome with features of both acute and chronic GVHD.26 Late-onset acute GVHD and the overlap syndrome occur with greater frequency after reduced-intensity conditioning (RIC), an increasingly widespread technique (see below). As shown in Table 1, the clinical manifestations of acute GVHD occur in the skin, gastrointestinal tract and liver.27 In a comprehensive review, Martin et al found that at the onset of acute GVHD, 81% of patients had skin involvement, 54% had GI involvement, and 50% had liver involvement.23 Recent data suggest that lungs might also be targets of experimental GVHD.28

Table 1

Acute GVHD Symptoms

Skinmaculopapular skin rash
upper GI tractnausea and/or anorexia PLUS positive histology
lower GI tractwatery diarrhea ≥ 500ml +/- severe abdominal pain +/- bloody diarrhea or ileus (after exclusion of infectious etiology)
Livercholestatic hyperbilirubinemia

Skin is most commonly affected and is usually the first organ involved, often coinciding with engraftment of donor cells. The characteristic maculopapular rash is pruritic and can spread throughout the body, sparing the scalp (Figure 1). In severe cases the skin may blister and ulcerate.27 Apoptosis at the base of epidermal rete pegs is a characteristic pathologic finding. Other features include dyskeratosis, exocytosis of lymphocytes, satellite lymphocytes adjacent to dyskeratotic epidermal keratinocytes, and a perivascular lymphocytic infiltration in the dermis.2930

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0001.jpg
Acute GVHD of the skin (Grade I). Photograph courtesy of J. Levine, M.D.

Gastrointestinal tract involvement of acute GVHD usually presents as diarrhea but may also include vomiting, anorexia, and/or abdominal pain when severe.29 The diarrhea of GVHD is secretory and often voluminous (greater than two liters per day). Bleeding, which carries a poor prognosis, occurs as a result of mucosal ulceration31 but patchy involvement of the mucosa often leads to a normal appearance on endoscopy.32

Radiologic findings of the gastrointestinal (GI) tract include luminal dilatation with thickening of the wall of the small bowel (“ribbon sign” on CT scan) and air/fluid levels suggestive of an ileus.27 Histologic features include patchy ulcerations, apoptotic bodies in the base of crypts, crypt abscesses, and loss as well as flattening of the surface epithelium.33

Liver disease caused by GVHD may be difficult to distinguish from other causes of liver dysfunction following BMT such as veno-occlusive disease, drug toxicity, viral infection, sepsis, or iron overload. The histologic features of hepatic GVHD are endothelialitis, lymphocytic infiltration of the portal areas, pericholangitis and bile duct destruction.3435 The liver is rarely biopsied because thrombocytopenia early after transplant greatly increases the risks of the procedure, usually making the diagnosis one of exclusion.

The incidence of the severity of acute GVHD is determined by the extent of involvement of these three principal target organs (see Table 1). The overall grades are classified as I (mild), II (moderate), III (severe) and IV (very severe). Severe GVHD carries a poor prognosis, with 25% long term survival for grade III and 5% for grade IV.36

The incidence of acute GVHD is directly related to the degree of mismatch between HLA proteins and ranges from 35-45% in recipients of full matched sibling donor grafts89 to 60-80% in recipients of one-antigen HLA mismatched unrelated donor grafts.63739 The same degree of mismatch causes less GVHD using UCB grafts and incidence of acute GVHD is lower following the transplant of partially matched UCB units and ranges from 35-65%.12

Clinical Features of Chronic GVHD

Chronic GVHD is the major cause of late non-relapse death following HCT.40 Its presentation may be progressive (active or acute GVHD merging into chronic), quiescent (acute GVHD that resolves completely but is later followed by chronic GVHD) or it may occur de novo. Older recipient age and a history of acute GVHD are the greatest risk factors for chronic GVHD,41 and strategies to prevent acute GVHD may therefore help to prevent chronic GVHD. As shown in Table 2, the manifestations of chronic GVHD are somewhat protean, and are often of an autoimmune nature. Clinical signs often first appear in the buccal mucosa (see Figure 2). New consensus criteria for the diagnosis and staging of chronic GVHD have recently been developed.26

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0002.jpg
Chronic GVHD: Lichenoid changes of the buccal mucosa. Photograph courtesy of J. P. Guadagrini, D.D.S.

Table 2

Chronic GVHD Symptoms

SkinDyspigmentation, new onset alopecia, poikiloderma, lichen planus-like eruptions or sclerotic features
NailsNail dystrophy or loss
MouthXerostomia, ulcers, lichen-type features, restrictions of mouth opening from sclerosis
EyesDry eyes, sicca syndrome, cicatricial conjunctivitis
Muscles, fascia, jointsFasciitis, myositis, or joint stiffness from contractures
Female GenitaliaVaginal sclerosis, ulcerations
GI tractAnorexia, weight loss, esophageal web or strictures
LiverJaundice, transaminitis
LungsRestrictive or obstructive defects on pulmonary function tests, bronchiolitis obliterans, pleural effusions
KidneysNephrotic syndrome (rare)
HeartPericarditis
MarrowThrombocytopenia, anemia, neutropenia

Pathophysiology of Acute GVHD

Two important principles are important to consider regarding the pathophysiology of acute GVHD. First, acute GVHD reflects exaggerated but normal inflammatory mechanisms mediated by donor lymphocytes infused into the recipient where they function appropriately, given the foreign environment they encounter. Second, the recipient tissues that stimulate donor lymphocytes have usually been damaged by underlying disease, prior infections, and the transplant conditioning regimen.29 As a result, these tissues produce molecules (sometimes referred to as “danger” signals) that promote the activation and proliferation of donor immune cells.4245 Mouse models havebeen central to our identification and understanding of the pathophysiologic mechanisms of GVHD, and canine models have been critical to the development of clinically useful strategies for GVHD prophylaxis and treatment and to the development of donor leukocyte infusions.364647 Based largely on these experimental models, the development of acute GVHD can be conceptualized in three sequential steps or phases: (1) activation of the APCs; (2) donor T cell activation, proliferation, differentiation and migration; and (3) target tissue destruction (Figure 3).

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0003.jpg
GVHD Pathophysiology

In Phase I, the recipient conditioning regimen damages host tissues and causes release of inflammatory cytokines such as TNFα, IL-1 and IL-6. Increased levels of these cytokines leads to activation of host antigen presenting cells (APCs). In Phase II, host APCs activate mature donor cells. The subsequent proliferation and differentiation of these activated T cells produces additional effectors that mediate the tissue damage, including Cytotoxic T Lymphocytes, Natural Killer (NK) cells, TNFα and IL-1. Lipopolysaccharide (LPS) that has leaked through the damaged intestinal mucosa triggers additional TNFα production. TNFα can damage tissue directly by inducing necrosis and apoptosis in the skin and GI tract through either TNF receptors or the Fas pathway. TNFα plays a direct role in intestinal GVHD damage which further amplifies damage in the skin, liver and lung in a “cytokine storm.”

Phase I: Activation of Antigen Presenting Cells (APCs)

The first step involves the activation of APCs by the underlying disease and the HCT conditioning regimen. Damaged host tissues respond by producing “danger” signals, including proinflammatory cytokines (e.g., TNF-α), chemokines, and increased expression of adhesion molecules, MHC antigens and costimulatory molecules on host APCs.424850 A recent report demonstrated that at one week after HCT, increased levels of TNF-α receptor I, a surrogate marker for TNF-α, strongly correlated with the later development of GVHD.51 Damage to the GI tract from the conditioning is particularly important because it allows for systemic translocation of additional inflammatory stimuli such as microbial products including lipopolysaccaride (LPS) or other pathogen-associated molecular patterns that further enhance the activation of host APCs.49 The secondary lymphoid tissue in the GI tract is likely the initial site of interaction between activated APCs and donor T cells.52 These observations have led an important clinical strategy to reduce acute GVHD by reducing the intensity of the conditioning regimen. Experimental GVHD can also be reduced by manipulating distinct subsets of APCs.5354 In addition, non-hematopoietic stem cells, such as mesenchymal stem cells or stromal cells, can reduce allogeneic T cell responses, although the mechanism for such inhibition remains unclear.2

The concept that enhanced activation of host APCs increases the risk for acute GVHD unifies a number of seemingly disparate clinical associations with that risk, such as advanced stages of malignancy, more intense transplant conditioning regimens and histories of viral infections. APCs detect infections by recognizing conserved molecular patterns that are unique to microbes, called pathogen-associated molecular patterns (PAMPs). Among the classes of receptors that recognize such patterns, the Toll-like receptors (TLR) are the best characterized.55 For example, TLR4 recognizes LPS55 and mice with mutant TLR4 receptors that do not respond to LPS cause less GVHD when used as donors.56 Other TLRs that recognize viral DNA or RNA also activate APCs and may enhance GVHD, providing a potential mechanistic basis for increased GVHD associated with viral infections such as cytomegalovirus (CMV).57

Phase II: Donor T Cell Activation

The core of the GVH reaction is Step 2, where donor T cells proliferate and differentiate in response to host APCs. The “danger” signals generated in Phase I augment this activation at least in part by increasing the expression of costimulatory molecules.58 Blockade of co-stimulatory pathways to prevent GVHD is successful in animal models, but this approach has not yet been tested in large clinical trials.2

In mouse models, where genetic differences between donor and recipient strains can be tightly controlled, CD4 cells induce acute GVHD to MHC class II differences, and CD8 cells induce acute GVHD to MHC class I differences.5961 In the majority of HLA-identical HCTs, both CD4 and CD8 subsets respond to minor histocompatibility antigens and can cause GVHD in HLA-identical HCT.

Regulatory T cells can suppress the proliferation of conventional T cells and prevent GVHD in animal models when added to donor grafts containing conventional T cells.62 In mice, the Foxp3 protein functions as a master switch in the development of regulatory T cells, which normally constitute 5% of the CD4+ T cell population.62 Regulatory T cells secrete anti-inflammatory cytokines IL-10 and Transforming Growth Factor(TGF)-β and can also act through contact-dependent inhibition of APCs.62 It is likely that the use of regulatory T cells in clinical acute GVHD will require improved techniques to identify and expand them.

Natural Killer T cell (NKT) 1.1 subsets of both the host and donors that have been shown to modulate acute GVHD.63 Host NKT cells have been shown to suppress acute GVHD in an IL-4 dependent manner.64 A recent clinical trial of total lymphoid irradiation used as conditioning significantly reduced GVHD and enhanced host NKT cell function.65 By contrast, donor NKT cells can reduce GVHD and enhance perforin mediated GVL in an experimental model.66

Activation of immune cells results in rapid intracellular biochemical cascades that induce transcription of genes for many proteins including cytokines and their receptors. Th1 cytokines (IFN-γ, IL-2 and TNF-α) are produced in large amounts during acute GVHD. IL-2 production by donor T cells remains the principal target of many current clinical therapeutic and prophylactic approaches to GVHD, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) directed against IL-2 and its receptor.9 But emerging data indicate an important role for IL-2 in the generation and maintenance of CD4 CD25 T regs, suggesting that prolonged interference with IL-2 may have an unintended consequence of preventing the development of long term tolerance after allogeneic HCT.67 IFN-γ has multiple functions and can either amplify or reduce GVHD.6869 IFN-γ may amplify GVHD by increasing the expression of molecules such as chemokines receptors, MHC proteins, and adhesion molecules; it also increases the sensitivity of monocytes and macrophages to stimuli such as LPS and accelerates intracellular cascades in response to these stimuli.70 Early polarization of donor T cells so that they secrete less IFN-γ and more IL-4 can also attenuate experimental acute GVHD.71 IFN-γ may amplify GVHD by directly damaging epithelium in the GI tract and skin and inducing immnosuppression through the induction of nitric oxide.72 By contrast, IFN-γ may suppress GVHD by hastening the apoptosis of activated donor T cells.6973. This complexity means the manipulation of IFN-γ may have diverse effects in vivo, making it a challenging target with respect to therapeutic intervention. IL-10 plays a key role in suppression of immune responses, and clinical data suggest it may regulate acute GVHD.17 TGF-β, another suppressive cytokine can suppress acute GVHD but exacerbate chronic GVHD.74 Thus the timing and duration of the secretion of any given cytokine may determine the specific effects of that cytokine on GVHD severity.

Phase III: Cellular and Inflammatory Effector Phase

The effector phase of this process is a complex cascade of both cellular mediators such as cytotoxic T lymphocytes(CTLs) and NK cells and soluble inflammatory mediators such as TNF-α, IFN-γ, IL-1 and nitric oxide.229 These soluble and cellular mediators synergize to amplify local tissue injury and further promote inflammation and target tissue destruction.

Cellular Effectors

The cellular effectors of acute GVHD are primarily CTLs and NK cells.49 CTLs that preferentially use the Fas/FasL pathway of target lysis and appear to predominate in GVHD liver damage (hepatocytes express large amounts of Fas) whereas GVHD CTLs that use the perforin /granzyme pathways are more important in the GI tract and skin.275 Chemokines direct the migration of donor T cells from lymphoid tissues to the target organs where they cause damage. Macrophage inflammatory protein-1alpha (MIP-1α) and other chemokines such as CCL2-5, CXCL2, CXCL9-11, CCL17 and CCL27 are over-expressed and enhance the homing of cellular effectors to target organs during experimental GVHD.76 Expression of integrins, such as α4β7 and its ligand MadCAM-1, are also important for homing of donor T cells to Peyer's patches during intestinal GVHD.527778

Inflammatory Effectors

Microbial products such as LPS that leak through a damaged intestinal mucosa or skin may stimulate secretion of inflammatory cytokines through Toll-like receptors (TLRs).4979 The GI tract is particularly susceptible to damage from TNF-α, and plays a major role in the amplification and propagation of the “cytokine storm” characteristic of acute GVHD.49 TNF-α can be produced by both donor and host cells, and it acts in three different ways: first, it activates APCs and enhances alloantigen presentation; second, it recruits effector cells to target organs via the induction of inflammatory chemokines; and third, it directly causes tissue necrosis, as its name suggests.8082

Prevention of GVHD

Based on the evidence from animal models regarding the central role of T cells in initiating GVHD, numerous clinical studies evaluating T cell depletion (TCD) as prophylaxis for GVHD were performed in the 1980's and 1990's. There were three principal TCD strategies: (1) negative selection of T cells ex vivo, (2) positive selection of CD34 stem cells ex vivo; and (3) anti-T cell antibodies in vivo.83 Most strategies showed a significant limitation in both acute and chronic GVHD.8488 Unfortunately, the lower incidence of severe GVHD was offset by high rates of graft failure, relapse of malignancy, infections, and Epstein-Barr virus-associated lymphoproliferative disorders. Negative selection purging strategies using various anti-T cell antibodies achieved similar long-term results regardless of the breadth of antibody specificity.8993 One large registry study demonstrated that purging strategies using antibodies with broad specificities produced inferior leukemia-free survival than standard immunosuppression in patients receiving unrelated donor transplants.94 Several studies have investigated partial T cell depletion, either by eliminating specific T cell subsets (e.g., CD8) or by titrating the dose of T cells present in the inoculum.9597 None of these approaches, however, has convincingly demonstrated an optimal strategy that improves long-term survival.

Alemtuzumab is a monoclonal antibody that binds CD52, a protein expressed on a broad spectrum of leukocytes including lymphocytes, monocytes, and dendritic cells. Its use in GVHD prophylaxis in a Phase II trial decreased the incidence of acute and chronic GVHD following reduced intensity transplant.98 In two prospective studies, patients who received alemtuzumab rather than methotrexate showed significantly lower rates of acute and chronic GVHD,99 but experienced more infectious complications and higher rates of relapse, so that there was no overall survival benefit. Alemtuzumab may also contribute to graft failure when used with minimal intensity conditioning regimens.100

An alternative strategy to TCD attempted to induce anergy in donor T cells by ex vivo antibody blockade of co-stimulatory pathways prior to transplantation. A small study using this approach in haploidentical HCT recipients was quite encouraging, but has not yet been replicated.101 Thus the focus of most prevention strategies remains pharmacological manipulation of T cells after transplant.

Administration of anti-T cell antibodies in vivo as GVHD prophylaxis has also been extensively tested. The best studied drugs are anti-thymocyte globulin (ATG) or antilymphocyte globulin (ALG) preparations. These sera, which have high titers of polyclonal antibodies, are made by immunizing animals (horses or rabbits) to thymocytes or lymphocytes, respectively. A complicating factor in determining the role of these polyclonal sera in transplantation is the observation that even different brands of the same class of sera exert different biologic effects.102 However, the side effects of ATG/ALG infusions are common across different preparations and include fever, chills, headache, thrombocytopenia (from cross-reactivity to platelets), and, infrequently, anaphylaxis. In retrospective studies, rabbit ATG reduced the incidence of GVHD in related donor HSCT recipients without appearing to improve survival.103104 In recipients of unrelated donor HSCT, addition of ALG to standard GVHD prophylaxis effectively prevented severe GVHD, but did not result in improved survival because of increased infections.105 In a long term follow-up study, however, pretransplant ATG provided significant protection against extensive chronic GVHD and chronic lung dysfunction.106

The primary pharmacologic strategy to prevent GVHD is the inhibition of the cytoplasmic enzyme, calcineurin, that is critical for in the activation of T cells. The calcineurin inhibitors, cyclosporine and tacrolimus, have similar mechanisms of action, clinical effectiveness and toxicity profiles, including hypomagnesemia, hyperkalemia, hypertension, and nephrotoxicity.9107 Serious side effects include transplant-associated thrombotic microangiopathy (TAM) and neurotoxicity that can lead to premature discontinuation. Although clinically similar to thrombotic thrombocytopenic purpura, TAM does not reliably respond to therapeutic plasmapheresis, carries a high mortality rate, and removal of the offending agent does not always result in improvement.108 Posterior reversible encephalopathy syndrome includes mental status changes, seizures, neurological deficits and characteristic magnetic resonance imaging findings; this syndrome has been seen in 1-2% of HCT recipients receiving and calcineurin inhibitors.109 Side effects of these drugs decrease as the dose is tapered, usually two to four months after HCT.

Calcineurin inhibitors are often administered in combination with other immunosuppressants, such as methotrexate, which is given at low doses in the early post-transplant period.9107 The toxicities of methotrexate (neutropenia and mucositis) have led some investigators to replace it with mycophenolate mofetil (MMF). In one 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 methotrexate.110 The incidence and severity of acute GVHD was similar between the two groups, but the study closed early due to superiority of the MMF arm with respect to reduced mucositis and the speed of hematopoietic engraftment. A desire for faster neutrophil engraftment has led to the use of MMF in UCB blood transplants where graft failure is a major concern.111 MMF is also often used after RIC regimens for similar reasons.112113

Sirolimus is an immunosuppressant that is structurally similar to tacrolimus but does not inhibit calcineurin. In a small Phase II trial, it showed excellent efficacy in combination with tacrolimus;114 the drug damages endothelial cells, however, and it may enhance TAM that is associated with calcineurin inhibitors.115 The combination of tacrolimus and sirolimus is currently being compared in a large randomized multi-center trial.

RIC regimens attempt to suppress the host immune system sufficiently so that donor T cells can engraft and then ablate the lympho-hematopoietic compartment of the recipient. The term “non-myeloablative” is therefore somewhat misleading. RIC regimens produce less tissue damage and lower levels of the inflammatory cytokines that are important in the initiation of GVHD pathophysiology; this effect may explain the reduced incidence of severe GVHD following RIC compared to the full intensity conditioning used in historical controls.98116 The onset of acute GVHD may be delayed after RIC until after day 100, however, and it may present simultaneously with elements of chronic GVHD (“overlap syndrome”).116120

Treatment of Acute GVHD

GVHD generally first develops in the second month after HCT, during continued treatment with calcineurin-based prophylaxis.23121 Steroids, with their potent antilymphocyte and anti-inflammatory activity, are the gold standard for treatment of GVHD. Many centers treat mild GVHD of the skin (Grade I) with topical steroids alone, but for more severe skin GVHD and any degree of visceral GVHD involvement, high-dose systemic steroids are usually initiated. Steroid therapy results in complete remission in less than half of the patients,122 and more severe GVHD is less likely to respond to treatment.123124 In a prospective randomized study, the addition of ATG to steroids as primary therapy did not increase the response rate.124 In a retrospective study, the use of ATG in patients who showed early signs of steroid-resistance was beneficial,122 but not all studies show such benefit and ATG is not standardly used because of increased infection risks.106125126.

An increasingly common treatment for GVHD is extracorporeal photopheresis (ECP). During ECP, the patient's white blood cells are collected by apheresis, incubated with the DNA-intercalating agent, 8-methoxypsoralen, exposed to ultraviolet light (UVA), and returned to the patient. ECP is known to induce cellular apoptosis, which has strong anti-inflammatory effects in a number of systems, including prevention of rejection of solid organ grafts.127 Animal studies show that ECP reverses acute GVHD by increasing the number of regulatory T cells.128 A Phase II clinical study of steroid-dependent or steroid refractory GVHD showed resolution of GVHD in a large majority of patients, with 50% long-term survival in this very high risk group.129 Randomized multi-center studies of this approach are needed to determine its place in the management of acute GVHD.

Another interesting strategy to treat GVHD is the blockade of the inflammatory cytokine TNF-α. TNF-α can activate APCs, recruit effector cells and cause direct tissue damage.130 In animal models, TNF-α plays a central role in GVHD of the GI tract, which is central to the “cytokine storm” and plasma levels of TNFR I (a surrogate marker for TNF-α) rise in patients before the clinical manifestations of GVHD appear. 51 A recent Phase II trial of etanercept, a solubilized TNFR II, showed significant efficacy when added to systemic steroids as primary therapy for acute GVHD. Seventy percent of patients had complete resolution of all GVHD symptoms within one month, with 80% complete responses in the GI tract and the skin. The authors also showed that plasma levels of TNFR I were a significant biomarker for clinical GVHD.131

Treatment of Chronic GVHD

In contrast to acute GVHD, the pathophysiology of chronic GVHD remains poorly understood, and it is treated with a variety of immunosuppressive agents. The response of chronic GVHD to treatment is unpredictable, and mixed responses in different organs can occur in the same patient. Confounding variables such as infection and co-morbidities also make responses hard to measure. The use of corticosteroids (with or without a calcineurin inhibitor) is the standard of care, but a randomized trial of more than 300 patients with chronic GVHD found no difference between cyclosporine plus prednisone versus prednisone alone.132 Chronic immunosuppressants, especially those containing steroids, are highly toxic and result in infectious deaths. Many second line therapies have been studied, but none has achieved widespread acceptance. As mentioned above, ECP shows some promise, with significant response rates in high-risk patients. The best responses were observed in skin, liver, oral mucosa, eye, and lung.133 This observation is particularly relevant because lung GVHD has the potential to be a particularly devastating complication necessitating lung transplant as the only therapeutic option.134135

Essential Supportive Care in GVHD Patients

Meticulous supportive care is critical for patients with both acute and chronic GVHD because of the extended duration of immunosuppressive treatments and because the multiple medications required may have synergistic toxicities. Such care includes extensive infectious prophylaxis, early interventions in cases of suspected infections, and prophylaxis against non-infectious side effects of medications (See Table 3). These complications often require rapid responses to prevent serious or irreversible damage, and are best handled in close collaboration between the primary physician and the transplant specialist.

Table 3

Recommendations for Supportive Care

ComplicationClinical symptomsRoutine
Monitoring
ProphylaxisRecommended
Intervention

Bacterial infections:
Catheter-relatedFever, chills Pain and/or erythemaAssessment of catheter sitesSterile dressing, Regular line maintenanceObtain cultures Immediate IV antibiotic treatment, remove line

OtherFever, chills Sepsis symptomsClinical signs, Chest X ray or CT scan for possible pneumoniaAntibiotic prophylaxis in high risk patients (high dose corticosteroids or asplenism) IVIG if IgG level < 400 mg/dlObtain cultures Immediate IV broad spectrum antibiotics as risk of over-whelming sepsis within hours

Viral infections:
CMVGastroenteritis Interstitial pneumoniaBlood CMV PCR OR pp65 antigen levelsPreemptive treatment in patients with reactivationAntiviral therapy: Ganciclovir, Valganciclovir, or Foscarnet

Respiratory VirusesSymptoms of upper/lower respiratory tract infectionsClinical monitoringAnnual influenza vaccination against influenza (starting at 6 months post HCT) Vaccination of caregiversEarly treatment with neuraminidase inhibitors (Influenza), other antivirals

Varicella-zoster virusVesicular skin lesionsClinical monitoringAcyclovir prophylaxisTreatment doses of antivirals

Fungal and other infections:
Aspergillosis; other emerging fungal infectionsPulmonary lesions Sinusitis Skin nodulesGalactomannan assays in high risk patients CT scan if signs of infectionVoriconazole OR Posaconazole prophylaxis in high risk patients (eg, high dose steroids)Antifungal therapy

CandidaThrush Pulmonary lesionsClinical exam CT scan if signs of infectionFluconazole (Aspergillus prophylaxis protects against Candida as well)Antifungal therapy

PneumocystisFever Hypoxia Respiratory distressClinical assessmentCotrimoxazole OR Pentamadine until one month off ISTreatment doses of anti-PCP medications

Other Toxicities of immunosuppressive agents:
Calcineurin inhibitors (CNI)Tremor
Clinical assessment Drug levels
Adjust dose to desired trough levels

Neurotoxicity
Assess mental status

Stop CNI
Renal impairmentCreatinine levels &amp; GFRAdequate fluid uptake (~3L/day)IV fluids
HypertensionBlood pressure monitoringAntihypertensive treatment (ACE inhibitors, ß blocking agents)
Transplant associated microangiopathyAssess blood smear for hemolysis, schistocytesStop CNI Plasmapheresis

CorticosteroidsCushing's symptomsClinical assessmentTaper as recommended
DiabetesBlood glucose levelsNutritional guidanceInsulin treatment
Osteoporosis
Assessment of bone density
Calcium /Vit D supplementation
Bisphosphonate treatment if osteoporosis
MyopathyPhysiotherapy
Late graft failureBleeding symptoms AnemiaBlood countsStop or switch responsible drugsGrowth factors: G-CSF Erythropoetin Transfusions

All patients should receive at least fluconazole as prophylaxis against fungal infections. Invasive molds, especially aspergillus, are common in patients with prolonged steroid use.136 Prophylaxis with voriconazole or posaconazole should be considered for these patients. Usual sites of infection are the lungs, sinuses, brain, skin,137 and serial galactomannan assays may aid in the early detection.138 Candida can cause lesions in the lung and spleen, which may need screening with ultrasonography. Pneumocystis is another opportunistic infection that should receive cotrimoxazol (bactrim) prophylaxis.139

Viral infections are frequent in these patients with GVHD. Cytomegalovirus causes interstitial pneumonia and gastritis. Patients who are at risk should have their blood monitored several times monthly. Techniques that directly detect virus should be performed, such as CMV PCR or pp65 antigen, and evidence of increased viral load should prompt preemptive treatment with ganciclovir or foscarnet prior to clinical manifestations of disease. Shingles is not uncommon and acyclovir prophylaxis may be beneficial.140 Patients and caregivers should receive vaccinations against influenza, and treatment with neuraminidase inhibitors is recommended in the event of influenza infection.141142

Patients with GVHD often have IgG2 and IgG4 subclass deficiencies despite normal lgG levels, making them susceptible to infections with encapsulated organisms. Treatment of severe hypogammaglobulinemia with intravenous immunoglobulin is standard in many centers,143 but the level that triggers replacement varies considerably among transplant specialists. There is little supporting evidence for the routine use of intravenous immunoglobulin as prophylaxis144 but patients should receive routine prophylaxis (penicillin or its equivalent) due to the increased risk of streptococcal sepsis.145 Pneumococcal conjugate and hemophilus influenza vaccine may provide additional protection and are also recommended for all patients, including those with chronic GVHD.139146147 The sites of any indwelling catheters should be assessed regularly and early treatment of a suspected infection initiated. Early signs or symptoms of septic shock such as shaking chills or low blood pressure requires prompt evaluation with chest X-ray and/or CT scan, blood culture and broad spectrum antibiotics because shock may progress rapidly in these patients.

Chronic immunosuppressant therapy has multiple toxicities. Diabetes (which further increases the risks of infection), muscle weakness, osteoporosis, avascular necrosis (often requiring joint replacement) and other Cushingoid features are common with chronic steroid use. Frequent monitoring of blood glucose and screening bone density is recommended, and treatment includes insulin, calcium, Vitamin D and bisphosphonates.148149 Calcineurin inhibitors frequently cause renal impairment, hypertension, and neurologic paraesthesias. Standard supportive care includes blood pressure monitoring, assessment of renal function, and monitoring of drug blood levels, which should be maintained within therapeutic parameters. To prevent renal dysfunction, most centers recommend vigorous oral outpatient hydration. Some patients will be unable to tolerate calcineurin inhibitors and will require different immunosuppressive medications altogether. Cytopenias sometimes require treatment with growth factors such as G-CSF or cessation of the offending agent. These maneuvers should always be performed in close consultation with a transplant specialist.

Future Directions

As allogeneic transplantation becomes an increasingly attractive therapeutic option, the need for novel approaches to GVHD has accelerated. The number of patients receiving transplants from unrelated donors is expected to double in the next five years, significantly increasing the population of patients with GVHD. The advent of RIC regimens has reduced transplant-related mortality and lengthened the period during which acute GVHD may develop (many new cases present up to day 200) and the need for close monitoring of patients in this period has increased. Patients have often returned to the care of their primary hematologists by this time, increasing the need for these physicians to collaborate with transplant specialists in the management of GVHD and its complications. Identification of biomarkers for GVHD with diagnostic (and possibly prognostic) significance may eventually make the treatment of GVHD preemptive rather than prophylactic. The use of cellular component therapy, such as regulatory T cells that have been expanded ex vivo. will also enter clinical trials in the near future, but the extensive infrastructure required for such cellular approaches will likely limit their use initially to large academic centers, intensifying the need for close communication between transplant specialists and referring hematologists.

Search Strategy and Selection Criteria

Pubmed and Medline databases were searched using the search term GVHD that was thencross-referenced with each of the following words: clinical, cytokines, MHC and HLA antigens, biology and immunology. The criteria used to include mostly the peer-reviewed original and review journal articles published within the last decade except for those seminal articles that initially described the clinical features. All non-peer reviewed manuscripts, supplements and textbooks were excluded.

Etiology and Clinical Features

Fifty years ago Billingham 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.3 We know now that the immunologically competent cells are T cells, and that GVHD can develop in various clinical settings when tissues containing T cells (blood products, bone marrow, and solid organs) are transferred from one person to another who is not able to eliminate those cells.45 Patients, whose immune systems are suppressed, and who receive white blood cells from another individual, are at particularly high risk for GVHD.

GVHD occurs when donor T cells respond to genetically defined proteins on host cells. The most important proteins are Human Leukocyte Antigens (HLA)267, which are highly polymorphic and are encoded by the major histocompatibility complex (MHC). Class I HLA (A, B, and C) proteins are expressed on almost all nucleated cells of the body at varying densities. Class II proteins (DR, DQ, and DP) are primarily expressed on hematopoietic cells (B cells, dendritic cells, monocytes), 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 have now largely replaced earlier methods. The incidence of acute GVHD is directly related to the degree of mismatch between HLA proteins89 and thus ideally, donors and recipients are matched at HLA-A, -B, -C, and -DRB1, (“8/8 matches”), but mismatches may be tolerated for UCB grafts (see below).1012

Non-HLA Genetics

Despite HLA identity between a patient and donor, approximately 40% of patients receiving HLA-identical grafts develop acute GVHD due to genetic differences that lie outside the HLA loci, or “minor” histocompatibility antigens (HA). Some minor HAs, such as HY and HA-3, are expressed on all tissues and are targets for both GVHD and GVL.13 Other minor HAs, such as HA-1 and HA-2, are expressed most abundantly on hematopoietic cells (including leukemic cells) and may therefore induce a greater GVL effect with less GVHD.1314

Polymorphisms in both donors and recipients for cytokines that are involved in the classical `cytokine storm' of GVHD (discussed below) have been implicated as risk factors for GVHD.15 Tumor Necrosis Factor (TNF)-α, Interleukin 10 (IL-10), Interferon-γ (IFNγ) variants have correlated with GVHD in some, but not all, studies.1618 Genetic polymorphisms of proteins involved in innate immunity, such as nucleotide oligomerization domain 2 and Keratin 18 receptors, have also been associated with GVHD.1922 Future strategies to identify the best possible transplant donor will probably incorporate both HLA and non-HLA genetic factors.

Clinical Features of Acute GVHD

Based on an early Seattle experience, acute GVHD was defined to occur prior to day 100, whereas chronic GVHD occurred after that time.2325 This definition is far from satisfactory, and a recent National Institutes of Health classification includes late-onset acute GVHD (after day 100) and an overlap syndrome with features of both acute and chronic GVHD.26 Late-onset acute GVHD and the overlap syndrome occur with greater frequency after reduced-intensity conditioning (RIC), an increasingly widespread technique (see below). As shown in Table 1, the clinical manifestations of acute GVHD occur in the skin, gastrointestinal tract and liver.27 In a comprehensive review, Martin et al found that at the onset of acute GVHD, 81% of patients had skin involvement, 54% had GI involvement, and 50% had liver involvement.23 Recent data suggest that lungs might also be targets of experimental GVHD.28

Table 1

Acute GVHD Symptoms

Skinmaculopapular skin rash
upper GI tractnausea and/or anorexia PLUS positive histology
lower GI tractwatery diarrhea ≥ 500ml +/- severe abdominal pain +/- bloody diarrhea or ileus (after exclusion of infectious etiology)
Livercholestatic hyperbilirubinemia

Skin is most commonly affected and is usually the first organ involved, often coinciding with engraftment of donor cells. The characteristic maculopapular rash is pruritic and can spread throughout the body, sparing the scalp (Figure 1). In severe cases the skin may blister and ulcerate.27 Apoptosis at the base of epidermal rete pegs is a characteristic pathologic finding. Other features include dyskeratosis, exocytosis of lymphocytes, satellite lymphocytes adjacent to dyskeratotic epidermal keratinocytes, and a perivascular lymphocytic infiltration in the dermis.2930

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0001.jpg
Acute GVHD of the skin (Grade I). Photograph courtesy of J. Levine, M.D.

Gastrointestinal tract involvement of acute GVHD usually presents as diarrhea but may also include vomiting, anorexia, and/or abdominal pain when severe.29 The diarrhea of GVHD is secretory and often voluminous (greater than two liters per day). Bleeding, which carries a poor prognosis, occurs as a result of mucosal ulceration31 but patchy involvement of the mucosa often leads to a normal appearance on endoscopy.32

Radiologic findings of the gastrointestinal (GI) tract include luminal dilatation with thickening of the wall of the small bowel (“ribbon sign” on CT scan) and air/fluid levels suggestive of an ileus.27 Histologic features include patchy ulcerations, apoptotic bodies in the base of crypts, crypt abscesses, and loss as well as flattening of the surface epithelium.33

Liver disease caused by GVHD may be difficult to distinguish from other causes of liver dysfunction following BMT such as veno-occlusive disease, drug toxicity, viral infection, sepsis, or iron overload. The histologic features of hepatic GVHD are endothelialitis, lymphocytic infiltration of the portal areas, pericholangitis and bile duct destruction.3435 The liver is rarely biopsied because thrombocytopenia early after transplant greatly increases the risks of the procedure, usually making the diagnosis one of exclusion.

The incidence of the severity of acute GVHD is determined by the extent of involvement of these three principal target organs (see Table 1). The overall grades are classified as I (mild), II (moderate), III (severe) and IV (very severe). Severe GVHD carries a poor prognosis, with 25% long term survival for grade III and 5% for grade IV.36

The incidence of acute GVHD is directly related to the degree of mismatch between HLA proteins and ranges from 35-45% in recipients of full matched sibling donor grafts89 to 60-80% in recipients of one-antigen HLA mismatched unrelated donor grafts.63739 The same degree of mismatch causes less GVHD using UCB grafts and incidence of acute GVHD is lower following the transplant of partially matched UCB units and ranges from 35-65%.12

Clinical Features of Chronic GVHD

Chronic GVHD is the major cause of late non-relapse death following HCT.40 Its presentation may be progressive (active or acute GVHD merging into chronic), quiescent (acute GVHD that resolves completely but is later followed by chronic GVHD) or it may occur de novo. Older recipient age and a history of acute GVHD are the greatest risk factors for chronic GVHD,41 and strategies to prevent acute GVHD may therefore help to prevent chronic GVHD. As shown in Table 2, the manifestations of chronic GVHD are somewhat protean, and are often of an autoimmune nature. Clinical signs often first appear in the buccal mucosa (see Figure 2). New consensus criteria for the diagnosis and staging of chronic GVHD have recently been developed.26

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0002.jpg
Chronic GVHD: Lichenoid changes of the buccal mucosa. Photograph courtesy of J. P. Guadagrini, D.D.S.

Table 2

Chronic GVHD Symptoms

SkinDyspigmentation, new onset alopecia, poikiloderma, lichen planus-like eruptions or sclerotic features
NailsNail dystrophy or loss
MouthXerostomia, ulcers, lichen-type features, restrictions of mouth opening from sclerosis
EyesDry eyes, sicca syndrome, cicatricial conjunctivitis
Muscles, fascia, jointsFasciitis, myositis, or joint stiffness from contractures
Female GenitaliaVaginal sclerosis, ulcerations
GI tractAnorexia, weight loss, esophageal web or strictures
LiverJaundice, transaminitis
LungsRestrictive or obstructive defects on pulmonary function tests, bronchiolitis obliterans, pleural effusions
KidneysNephrotic syndrome (rare)
HeartPericarditis
MarrowThrombocytopenia, anemia, neutropenia

Pathophysiology of Acute GVHD

Two important principles are important to consider regarding the pathophysiology of acute GVHD. First, acute GVHD reflects exaggerated but normal inflammatory mechanisms mediated by donor lymphocytes infused into the recipient where they function appropriately, given the foreign environment they encounter. Second, the recipient tissues that stimulate donor lymphocytes have usually been damaged by underlying disease, prior infections, and the transplant conditioning regimen.29 As a result, these tissues produce molecules (sometimes referred to as “danger” signals) that promote the activation and proliferation of donor immune cells.4245 Mouse models havebeen central to our identification and understanding of the pathophysiologic mechanisms of GVHD, and canine models have been critical to the development of clinically useful strategies for GVHD prophylaxis and treatment and to the development of donor leukocyte infusions.364647 Based largely on these experimental models, the development of acute GVHD can be conceptualized in three sequential steps or phases: (1) activation of the APCs; (2) donor T cell activation, proliferation, differentiation and migration; and (3) target tissue destruction (Figure 3).

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0003.jpg
GVHD Pathophysiology

In Phase I, the recipient conditioning regimen damages host tissues and causes release of inflammatory cytokines such as TNFα, IL-1 and IL-6. Increased levels of these cytokines leads to activation of host antigen presenting cells (APCs). In Phase II, host APCs activate mature donor cells. The subsequent proliferation and differentiation of these activated T cells produces additional effectors that mediate the tissue damage, including Cytotoxic T Lymphocytes, Natural Killer (NK) cells, TNFα and IL-1. Lipopolysaccharide (LPS) that has leaked through the damaged intestinal mucosa triggers additional TNFα production. TNFα can damage tissue directly by inducing necrosis and apoptosis in the skin and GI tract through either TNF receptors or the Fas pathway. TNFα plays a direct role in intestinal GVHD damage which further amplifies damage in the skin, liver and lung in a “cytokine storm.”

Phase I: Activation of Antigen Presenting Cells (APCs)

The first step involves the activation of APCs by the underlying disease and the HCT conditioning regimen. Damaged host tissues respond by producing “danger” signals, including proinflammatory cytokines (e.g., TNF-α), chemokines, and increased expression of adhesion molecules, MHC antigens and costimulatory molecules on host APCs.424850 A recent report demonstrated that at one week after HCT, increased levels of TNF-α receptor I, a surrogate marker for TNF-α, strongly correlated with the later development of GVHD.51 Damage to the GI tract from the conditioning is particularly important because it allows for systemic translocation of additional inflammatory stimuli such as microbial products including lipopolysaccaride (LPS) or other pathogen-associated molecular patterns that further enhance the activation of host APCs.49 The secondary lymphoid tissue in the GI tract is likely the initial site of interaction between activated APCs and donor T cells.52 These observations have led an important clinical strategy to reduce acute GVHD by reducing the intensity of the conditioning regimen. Experimental GVHD can also be reduced by manipulating distinct subsets of APCs.5354 In addition, non-hematopoietic stem cells, such as mesenchymal stem cells or stromal cells, can reduce allogeneic T cell responses, although the mechanism for such inhibition remains unclear.2

The concept that enhanced activation of host APCs increases the risk for acute GVHD unifies a number of seemingly disparate clinical associations with that risk, such as advanced stages of malignancy, more intense transplant conditioning regimens and histories of viral infections. APCs detect infections by recognizing conserved molecular patterns that are unique to microbes, called pathogen-associated molecular patterns (PAMPs). Among the classes of receptors that recognize such patterns, the Toll-like receptors (TLR) are the best characterized.55 For example, TLR4 recognizes LPS55 and mice with mutant TLR4 receptors that do not respond to LPS cause less GVHD when used as donors.56 Other TLRs that recognize viral DNA or RNA also activate APCs and may enhance GVHD, providing a potential mechanistic basis for increased GVHD associated with viral infections such as cytomegalovirus (CMV).57

Phase II: Donor T Cell Activation

The core of the GVH reaction is Step 2, where donor T cells proliferate and differentiate in response to host APCs. The “danger” signals generated in Phase I augment this activation at least in part by increasing the expression of costimulatory molecules.58 Blockade of co-stimulatory pathways to prevent GVHD is successful in animal models, but this approach has not yet been tested in large clinical trials.2

In mouse models, where genetic differences between donor and recipient strains can be tightly controlled, CD4 cells induce acute GVHD to MHC class II differences, and CD8 cells induce acute GVHD to MHC class I differences.5961 In the majority of HLA-identical HCTs, both CD4 and CD8 subsets respond to minor histocompatibility antigens and can cause GVHD in HLA-identical HCT.

Regulatory T cells can suppress the proliferation of conventional T cells and prevent GVHD in animal models when added to donor grafts containing conventional T cells.62 In mice, the Foxp3 protein functions as a master switch in the development of regulatory T cells, which normally constitute 5% of the CD4+ T cell population.62 Regulatory T cells secrete anti-inflammatory cytokines IL-10 and Transforming Growth Factor(TGF)-β and can also act through contact-dependent inhibition of APCs.62 It is likely that the use of regulatory T cells in clinical acute GVHD will require improved techniques to identify and expand them.

Natural Killer T cell (NKT) 1.1 subsets of both the host and donors that have been shown to modulate acute GVHD.63 Host NKT cells have been shown to suppress acute GVHD in an IL-4 dependent manner.64 A recent clinical trial of total lymphoid irradiation used as conditioning significantly reduced GVHD and enhanced host NKT cell function.65 By contrast, donor NKT cells can reduce GVHD and enhance perforin mediated GVL in an experimental model.66

Activation of immune cells results in rapid intracellular biochemical cascades that induce transcription of genes for many proteins including cytokines and their receptors. Th1 cytokines (IFN-γ, IL-2 and TNF-α) are produced in large amounts during acute GVHD. IL-2 production by donor T cells remains the principal target of many current clinical therapeutic and prophylactic approaches to GVHD, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) directed against IL-2 and its receptor.9 But emerging data indicate an important role for IL-2 in the generation and maintenance of CD4 CD25 T regs, suggesting that prolonged interference with IL-2 may have an unintended consequence of preventing the development of long term tolerance after allogeneic HCT.67 IFN-γ has multiple functions and can either amplify or reduce GVHD.6869 IFN-γ may amplify GVHD by increasing the expression of molecules such as chemokines receptors, MHC proteins, and adhesion molecules; it also increases the sensitivity of monocytes and macrophages to stimuli such as LPS and accelerates intracellular cascades in response to these stimuli.70 Early polarization of donor T cells so that they secrete less IFN-γ and more IL-4 can also attenuate experimental acute GVHD.71 IFN-γ may amplify GVHD by directly damaging epithelium in the GI tract and skin and inducing immnosuppression through the induction of nitric oxide.72 By contrast, IFN-γ may suppress GVHD by hastening the apoptosis of activated donor T cells.6973. This complexity means the manipulation of IFN-γ may have diverse effects in vivo, making it a challenging target with respect to therapeutic intervention. IL-10 plays a key role in suppression of immune responses, and clinical data suggest it may regulate acute GVHD.17 TGF-β, another suppressive cytokine can suppress acute GVHD but exacerbate chronic GVHD.74 Thus the timing and duration of the secretion of any given cytokine may determine the specific effects of that cytokine on GVHD severity.

Phase III: Cellular and Inflammatory Effector Phase

The effector phase of this process is a complex cascade of both cellular mediators such as cytotoxic T lymphocytes(CTLs) and NK cells and soluble inflammatory mediators such as TNF-α, IFN-γ, IL-1 and nitric oxide.229 These soluble and cellular mediators synergize to amplify local tissue injury and further promote inflammation and target tissue destruction.

Cellular Effectors

The cellular effectors of acute GVHD are primarily CTLs and NK cells.49 CTLs that preferentially use the Fas/FasL pathway of target lysis and appear to predominate in GVHD liver damage (hepatocytes express large amounts of Fas) whereas GVHD CTLs that use the perforin /granzyme pathways are more important in the GI tract and skin.275 Chemokines direct the migration of donor T cells from lymphoid tissues to the target organs where they cause damage. Macrophage inflammatory protein-1alpha (MIP-1α) and other chemokines such as CCL2-5, CXCL2, CXCL9-11, CCL17 and CCL27 are over-expressed and enhance the homing of cellular effectors to target organs during experimental GVHD.76 Expression of integrins, such as α4β7 and its ligand MadCAM-1, are also important for homing of donor T cells to Peyer's patches during intestinal GVHD.527778

Inflammatory Effectors

Microbial products such as LPS that leak through a damaged intestinal mucosa or skin may stimulate secretion of inflammatory cytokines through Toll-like receptors (TLRs).4979 The GI tract is particularly susceptible to damage from TNF-α, and plays a major role in the amplification and propagation of the “cytokine storm” characteristic of acute GVHD.49 TNF-α can be produced by both donor and host cells, and it acts in three different ways: first, it activates APCs and enhances alloantigen presentation; second, it recruits effector cells to target organs via the induction of inflammatory chemokines; and third, it directly causes tissue necrosis, as its name suggests.8082

Prevention of GVHD

Based on the evidence from animal models regarding the central role of T cells in initiating GVHD, numerous clinical studies evaluating T cell depletion (TCD) as prophylaxis for GVHD were performed in the 1980's and 1990's. There were three principal TCD strategies: (1) negative selection of T cells ex vivo, (2) positive selection of CD34 stem cells ex vivo; and (3) anti-T cell antibodies in vivo.83 Most strategies showed a significant limitation in both acute and chronic GVHD.8488 Unfortunately, the lower incidence of severe GVHD was offset by high rates of graft failure, relapse of malignancy, infections, and Epstein-Barr virus-associated lymphoproliferative disorders. Negative selection purging strategies using various anti-T cell antibodies achieved similar long-term results regardless of the breadth of antibody specificity.8993 One large registry study demonstrated that purging strategies using antibodies with broad specificities produced inferior leukemia-free survival than standard immunosuppression in patients receiving unrelated donor transplants.94 Several studies have investigated partial T cell depletion, either by eliminating specific T cell subsets (e.g., CD8) or by titrating the dose of T cells present in the inoculum.9597 None of these approaches, however, has convincingly demonstrated an optimal strategy that improves long-term survival.

Alemtuzumab is a monoclonal antibody that binds CD52, a protein expressed on a broad spectrum of leukocytes including lymphocytes, monocytes, and dendritic cells. Its use in GVHD prophylaxis in a Phase II trial decreased the incidence of acute and chronic GVHD following reduced intensity transplant.98 In two prospective studies, patients who received alemtuzumab rather than methotrexate showed significantly lower rates of acute and chronic GVHD,99 but experienced more infectious complications and higher rates of relapse, so that there was no overall survival benefit. Alemtuzumab may also contribute to graft failure when used with minimal intensity conditioning regimens.100

An alternative strategy to TCD attempted to induce anergy in donor T cells by ex vivo antibody blockade of co-stimulatory pathways prior to transplantation. A small study using this approach in haploidentical HCT recipients was quite encouraging, but has not yet been replicated.101 Thus the focus of most prevention strategies remains pharmacological manipulation of T cells after transplant.

Administration of anti-T cell antibodies in vivo as GVHD prophylaxis has also been extensively tested. The best studied drugs are anti-thymocyte globulin (ATG) or antilymphocyte globulin (ALG) preparations. These sera, which have high titers of polyclonal antibodies, are made by immunizing animals (horses or rabbits) to thymocytes or lymphocytes, respectively. A complicating factor in determining the role of these polyclonal sera in transplantation is the observation that even different brands of the same class of sera exert different biologic effects.102 However, the side effects of ATG/ALG infusions are common across different preparations and include fever, chills, headache, thrombocytopenia (from cross-reactivity to platelets), and, infrequently, anaphylaxis. In retrospective studies, rabbit ATG reduced the incidence of GVHD in related donor HSCT recipients without appearing to improve survival.103104 In recipients of unrelated donor HSCT, addition of ALG to standard GVHD prophylaxis effectively prevented severe GVHD, but did not result in improved survival because of increased infections.105 In a long term follow-up study, however, pretransplant ATG provided significant protection against extensive chronic GVHD and chronic lung dysfunction.106

The primary pharmacologic strategy to prevent GVHD is the inhibition of the cytoplasmic enzyme, calcineurin, that is critical for in the activation of T cells. The calcineurin inhibitors, cyclosporine and tacrolimus, have similar mechanisms of action, clinical effectiveness and toxicity profiles, including hypomagnesemia, hyperkalemia, hypertension, and nephrotoxicity.9107 Serious side effects include transplant-associated thrombotic microangiopathy (TAM) and neurotoxicity that can lead to premature discontinuation. Although clinically similar to thrombotic thrombocytopenic purpura, TAM does not reliably respond to therapeutic plasmapheresis, carries a high mortality rate, and removal of the offending agent does not always result in improvement.108 Posterior reversible encephalopathy syndrome includes mental status changes, seizures, neurological deficits and characteristic magnetic resonance imaging findings; this syndrome has been seen in 1-2% of HCT recipients receiving and calcineurin inhibitors.109 Side effects of these drugs decrease as the dose is tapered, usually two to four months after HCT.

Calcineurin inhibitors are often administered in combination with other immunosuppressants, such as methotrexate, which is given at low doses in the early post-transplant period.9107 The toxicities of methotrexate (neutropenia and mucositis) have led some investigators to replace it with mycophenolate mofetil (MMF). In one 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 methotrexate.110 The incidence and severity of acute GVHD was similar between the two groups, but the study closed early due to superiority of the MMF arm with respect to reduced mucositis and the speed of hematopoietic engraftment. A desire for faster neutrophil engraftment has led to the use of MMF in UCB blood transplants where graft failure is a major concern.111 MMF is also often used after RIC regimens for similar reasons.112113

Sirolimus is an immunosuppressant that is structurally similar to tacrolimus but does not inhibit calcineurin. In a small Phase II trial, it showed excellent efficacy in combination with tacrolimus;114 the drug damages endothelial cells, however, and it may enhance TAM that is associated with calcineurin inhibitors.115 The combination of tacrolimus and sirolimus is currently being compared in a large randomized multi-center trial.

RIC regimens attempt to suppress the host immune system sufficiently so that donor T cells can engraft and then ablate the lympho-hematopoietic compartment of the recipient. The term “non-myeloablative” is therefore somewhat misleading. RIC regimens produce less tissue damage and lower levels of the inflammatory cytokines that are important in the initiation of GVHD pathophysiology; this effect may explain the reduced incidence of severe GVHD following RIC compared to the full intensity conditioning used in historical controls.98116 The onset of acute GVHD may be delayed after RIC until after day 100, however, and it may present simultaneously with elements of chronic GVHD (“overlap syndrome”).116120

Treatment of Acute GVHD

GVHD generally first develops in the second month after HCT, during continued treatment with calcineurin-based prophylaxis.23121 Steroids, with their potent antilymphocyte and anti-inflammatory activity, are the gold standard for treatment of GVHD. Many centers treat mild GVHD of the skin (Grade I) with topical steroids alone, but for more severe skin GVHD and any degree of visceral GVHD involvement, high-dose systemic steroids are usually initiated. Steroid therapy results in complete remission in less than half of the patients,122 and more severe GVHD is less likely to respond to treatment.123124 In a prospective randomized study, the addition of ATG to steroids as primary therapy did not increase the response rate.124 In a retrospective study, the use of ATG in patients who showed early signs of steroid-resistance was beneficial,122 but not all studies show such benefit and ATG is not standardly used because of increased infection risks.106125126.

An increasingly common treatment for GVHD is extracorporeal photopheresis (ECP). During ECP, the patient's white blood cells are collected by apheresis, incubated with the DNA-intercalating agent, 8-methoxypsoralen, exposed to ultraviolet light (UVA), and returned to the patient. ECP is known to induce cellular apoptosis, which has strong anti-inflammatory effects in a number of systems, including prevention of rejection of solid organ grafts.127 Animal studies show that ECP reverses acute GVHD by increasing the number of regulatory T cells.128 A Phase II clinical study of steroid-dependent or steroid refractory GVHD showed resolution of GVHD in a large majority of patients, with 50% long-term survival in this very high risk group.129 Randomized multi-center studies of this approach are needed to determine its place in the management of acute GVHD.

Another interesting strategy to treat GVHD is the blockade of the inflammatory cytokine TNF-α. TNF-α can activate APCs, recruit effector cells and cause direct tissue damage.130 In animal models, TNF-α plays a central role in GVHD of the GI tract, which is central to the “cytokine storm” and plasma levels of TNFR I (a surrogate marker for TNF-α) rise in patients before the clinical manifestations of GVHD appear. 51 A recent Phase II trial of etanercept, a solubilized TNFR II, showed significant efficacy when added to systemic steroids as primary therapy for acute GVHD. Seventy percent of patients had complete resolution of all GVHD symptoms within one month, with 80% complete responses in the GI tract and the skin. The authors also showed that plasma levels of TNFR I were a significant biomarker for clinical GVHD.131

Treatment of Chronic GVHD

In contrast to acute GVHD, the pathophysiology of chronic GVHD remains poorly understood, and it is treated with a variety of immunosuppressive agents. The response of chronic GVHD to treatment is unpredictable, and mixed responses in different organs can occur in the same patient. Confounding variables such as infection and co-morbidities also make responses hard to measure. The use of corticosteroids (with or without a calcineurin inhibitor) is the standard of care, but a randomized trial of more than 300 patients with chronic GVHD found no difference between cyclosporine plus prednisone versus prednisone alone.132 Chronic immunosuppressants, especially those containing steroids, are highly toxic and result in infectious deaths. Many second line therapies have been studied, but none has achieved widespread acceptance. As mentioned above, ECP shows some promise, with significant response rates in high-risk patients. The best responses were observed in skin, liver, oral mucosa, eye, and lung.133 This observation is particularly relevant because lung GVHD has the potential to be a particularly devastating complication necessitating lung transplant as the only therapeutic option.134135

Essential Supportive Care in GVHD Patients

Meticulous supportive care is critical for patients with both acute and chronic GVHD because of the extended duration of immunosuppressive treatments and because the multiple medications required may have synergistic toxicities. Such care includes extensive infectious prophylaxis, early interventions in cases of suspected infections, and prophylaxis against non-infectious side effects of medications (See Table 3). These complications often require rapid responses to prevent serious or irreversible damage, and are best handled in close collaboration between the primary physician and the transplant specialist.

Table 3

Recommendations for Supportive Care

ComplicationClinical symptomsRoutine
Monitoring
ProphylaxisRecommended
Intervention

Bacterial infections:
Catheter-relatedFever, chills Pain and/or erythemaAssessment of catheter sitesSterile dressing, Regular line maintenanceObtain cultures Immediate IV antibiotic treatment, remove line

OtherFever, chills Sepsis symptomsClinical signs, Chest X ray or CT scan for possible pneumoniaAntibiotic prophylaxis in high risk patients (high dose corticosteroids or asplenism) IVIG if IgG level < 400 mg/dlObtain cultures Immediate IV broad spectrum antibiotics as risk of over-whelming sepsis within hours

Viral infections:
CMVGastroenteritis Interstitial pneumoniaBlood CMV PCR OR pp65 antigen levelsPreemptive treatment in patients with reactivationAntiviral therapy: Ganciclovir, Valganciclovir, or Foscarnet

Respiratory VirusesSymptoms of upper/lower respiratory tract infectionsClinical monitoringAnnual influenza vaccination against influenza (starting at 6 months post HCT) Vaccination of caregiversEarly treatment with neuraminidase inhibitors (Influenza), other antivirals

Varicella-zoster virusVesicular skin lesionsClinical monitoringAcyclovir prophylaxisTreatment doses of antivirals

Fungal and other infections:
Aspergillosis; other emerging fungal infectionsPulmonary lesions Sinusitis Skin nodulesGalactomannan assays in high risk patients CT scan if signs of infectionVoriconazole OR Posaconazole prophylaxis in high risk patients (eg, high dose steroids)Antifungal therapy

CandidaThrush Pulmonary lesionsClinical exam CT scan if signs of infectionFluconazole (Aspergillus prophylaxis protects against Candida as well)Antifungal therapy

PneumocystisFever Hypoxia Respiratory distressClinical assessmentCotrimoxazole OR Pentamadine until one month off ISTreatment doses of anti-PCP medications

Other Toxicities of immunosuppressive agents:
Calcineurin inhibitors (CNI)Tremor
Clinical assessment Drug levels
Adjust dose to desired trough levels

Neurotoxicity
Assess mental status

Stop CNI
Renal impairmentCreatinine levels &amp; GFRAdequate fluid uptake (~3L/day)IV fluids
HypertensionBlood pressure monitoringAntihypertensive treatment (ACE inhibitors, ß blocking agents)
Transplant associated microangiopathyAssess blood smear for hemolysis, schistocytesStop CNI Plasmapheresis

CorticosteroidsCushing's symptomsClinical assessmentTaper as recommended
DiabetesBlood glucose levelsNutritional guidanceInsulin treatment
Osteoporosis
Assessment of bone density
Calcium /Vit D supplementation
Bisphosphonate treatment if osteoporosis
MyopathyPhysiotherapy
Late graft failureBleeding symptoms AnemiaBlood countsStop or switch responsible drugsGrowth factors: G-CSF Erythropoetin Transfusions

All patients should receive at least fluconazole as prophylaxis against fungal infections. Invasive molds, especially aspergillus, are common in patients with prolonged steroid use.136 Prophylaxis with voriconazole or posaconazole should be considered for these patients. Usual sites of infection are the lungs, sinuses, brain, skin,137 and serial galactomannan assays may aid in the early detection.138 Candida can cause lesions in the lung and spleen, which may need screening with ultrasonography. Pneumocystis is another opportunistic infection that should receive cotrimoxazol (bactrim) prophylaxis.139

Viral infections are frequent in these patients with GVHD. Cytomegalovirus causes interstitial pneumonia and gastritis. Patients who are at risk should have their blood monitored several times monthly. Techniques that directly detect virus should be performed, such as CMV PCR or pp65 antigen, and evidence of increased viral load should prompt preemptive treatment with ganciclovir or foscarnet prior to clinical manifestations of disease. Shingles is not uncommon and acyclovir prophylaxis may be beneficial.140 Patients and caregivers should receive vaccinations against influenza, and treatment with neuraminidase inhibitors is recommended in the event of influenza infection.141142

Patients with GVHD often have IgG2 and IgG4 subclass deficiencies despite normal lgG levels, making them susceptible to infections with encapsulated organisms. Treatment of severe hypogammaglobulinemia with intravenous immunoglobulin is standard in many centers,143 but the level that triggers replacement varies considerably among transplant specialists. There is little supporting evidence for the routine use of intravenous immunoglobulin as prophylaxis144 but patients should receive routine prophylaxis (penicillin or its equivalent) due to the increased risk of streptococcal sepsis.145 Pneumococcal conjugate and hemophilus influenza vaccine may provide additional protection and are also recommended for all patients, including those with chronic GVHD.139146147 The sites of any indwelling catheters should be assessed regularly and early treatment of a suspected infection initiated. Early signs or symptoms of septic shock such as shaking chills or low blood pressure requires prompt evaluation with chest X-ray and/or CT scan, blood culture and broad spectrum antibiotics because shock may progress rapidly in these patients.

Chronic immunosuppressant therapy has multiple toxicities. Diabetes (which further increases the risks of infection), muscle weakness, osteoporosis, avascular necrosis (often requiring joint replacement) and other Cushingoid features are common with chronic steroid use. Frequent monitoring of blood glucose and screening bone density is recommended, and treatment includes insulin, calcium, Vitamin D and bisphosphonates.148149 Calcineurin inhibitors frequently cause renal impairment, hypertension, and neurologic paraesthesias. Standard supportive care includes blood pressure monitoring, assessment of renal function, and monitoring of drug blood levels, which should be maintained within therapeutic parameters. To prevent renal dysfunction, most centers recommend vigorous oral outpatient hydration. Some patients will be unable to tolerate calcineurin inhibitors and will require different immunosuppressive medications altogether. Cytopenias sometimes require treatment with growth factors such as G-CSF or cessation of the offending agent. These maneuvers should always be performed in close consultation with a transplant specialist.

Future Directions

As allogeneic transplantation becomes an increasingly attractive therapeutic option, the need for novel approaches to GVHD has accelerated. The number of patients receiving transplants from unrelated donors is expected to double in the next five years, significantly increasing the population of patients with GVHD. The advent of RIC regimens has reduced transplant-related mortality and lengthened the period during which acute GVHD may develop (many new cases present up to day 200) and the need for close monitoring of patients in this period has increased. Patients have often returned to the care of their primary hematologists by this time, increasing the need for these physicians to collaborate with transplant specialists in the management of GVHD and its complications. Identification of biomarkers for GVHD with diagnostic (and possibly prognostic) significance may eventually make the treatment of GVHD preemptive rather than prophylactic. The use of cellular component therapy, such as regulatory T cells that have been expanded ex vivo. will also enter clinical trials in the near future, but the extensive infrastructure required for such cellular approaches will likely limit their use initially to large academic centers, intensifying the need for close communication between transplant specialists and referring hematologists.

Search Strategy and Selection Criteria

Pubmed and Medline databases were searched using the search term GVHD that was thencross-referenced with each of the following words: clinical, cytokines, MHC and HLA antigens, biology and immunology. The criteria used to include mostly the peer-reviewed original and review journal articles published within the last decade except for those seminal articles that initially described the clinical features. All non-peer reviewed manuscripts, supplements and textbooks were excluded.

Non-HLA Genetics

Despite HLA identity between a patient and donor, approximately 40% of patients receiving HLA-identical grafts develop acute GVHD due to genetic differences that lie outside the HLA loci, or “minor” histocompatibility antigens (HA). Some minor HAs, such as HY and HA-3, are expressed on all tissues and are targets for both GVHD and GVL.13 Other minor HAs, such as HA-1 and HA-2, are expressed most abundantly on hematopoietic cells (including leukemic cells) and may therefore induce a greater GVL effect with less GVHD.1314

Polymorphisms in both donors and recipients for cytokines that are involved in the classical `cytokine storm' of GVHD (discussed below) have been implicated as risk factors for GVHD.15 Tumor Necrosis Factor (TNF)-α, Interleukin 10 (IL-10), Interferon-γ (IFNγ) variants have correlated with GVHD in some, but not all, studies.1618 Genetic polymorphisms of proteins involved in innate immunity, such as nucleotide oligomerization domain 2 and Keratin 18 receptors, have also been associated with GVHD.1922 Future strategies to identify the best possible transplant donor will probably incorporate both HLA and non-HLA genetic factors.

Clinical Features of Acute GVHD

Based on an early Seattle experience, acute GVHD was defined to occur prior to day 100, whereas chronic GVHD occurred after that time.2325 This definition is far from satisfactory, and a recent National Institutes of Health classification includes late-onset acute GVHD (after day 100) and an overlap syndrome with features of both acute and chronic GVHD.26 Late-onset acute GVHD and the overlap syndrome occur with greater frequency after reduced-intensity conditioning (RIC), an increasingly widespread technique (see below). As shown in Table 1, the clinical manifestations of acute GVHD occur in the skin, gastrointestinal tract and liver.27 In a comprehensive review, Martin et al found that at the onset of acute GVHD, 81% of patients had skin involvement, 54% had GI involvement, and 50% had liver involvement.23 Recent data suggest that lungs might also be targets of experimental GVHD.28

Table 1

Acute GVHD Symptoms

Skinmaculopapular skin rash
upper GI tractnausea and/or anorexia PLUS positive histology
lower GI tractwatery diarrhea ≥ 500ml +/- severe abdominal pain +/- bloody diarrhea or ileus (after exclusion of infectious etiology)
Livercholestatic hyperbilirubinemia

Skin is most commonly affected and is usually the first organ involved, often coinciding with engraftment of donor cells. The characteristic maculopapular rash is pruritic and can spread throughout the body, sparing the scalp (Figure 1). In severe cases the skin may blister and ulcerate.27 Apoptosis at the base of epidermal rete pegs is a characteristic pathologic finding. Other features include dyskeratosis, exocytosis of lymphocytes, satellite lymphocytes adjacent to dyskeratotic epidermal keratinocytes, and a perivascular lymphocytic infiltration in the dermis.2930

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0001.jpg
Acute GVHD of the skin (Grade I). Photograph courtesy of J. Levine, M.D.

Gastrointestinal tract involvement of acute GVHD usually presents as diarrhea but may also include vomiting, anorexia, and/or abdominal pain when severe.29 The diarrhea of GVHD is secretory and often voluminous (greater than two liters per day). Bleeding, which carries a poor prognosis, occurs as a result of mucosal ulceration31 but patchy involvement of the mucosa often leads to a normal appearance on endoscopy.32

Radiologic findings of the gastrointestinal (GI) tract include luminal dilatation with thickening of the wall of the small bowel (“ribbon sign” on CT scan) and air/fluid levels suggestive of an ileus.27 Histologic features include patchy ulcerations, apoptotic bodies in the base of crypts, crypt abscesses, and loss as well as flattening of the surface epithelium.33

Liver disease caused by GVHD may be difficult to distinguish from other causes of liver dysfunction following BMT such as veno-occlusive disease, drug toxicity, viral infection, sepsis, or iron overload. The histologic features of hepatic GVHD are endothelialitis, lymphocytic infiltration of the portal areas, pericholangitis and bile duct destruction.3435 The liver is rarely biopsied because thrombocytopenia early after transplant greatly increases the risks of the procedure, usually making the diagnosis one of exclusion.

The incidence of the severity of acute GVHD is determined by the extent of involvement of these three principal target organs (see Table 1). The overall grades are classified as I (mild), II (moderate), III (severe) and IV (very severe). Severe GVHD carries a poor prognosis, with 25% long term survival for grade III and 5% for grade IV.36

The incidence of acute GVHD is directly related to the degree of mismatch between HLA proteins and ranges from 35-45% in recipients of full matched sibling donor grafts89 to 60-80% in recipients of one-antigen HLA mismatched unrelated donor grafts.63739 The same degree of mismatch causes less GVHD using UCB grafts and incidence of acute GVHD is lower following the transplant of partially matched UCB units and ranges from 35-65%.12

Clinical Features of Chronic GVHD

Chronic GVHD is the major cause of late non-relapse death following HCT.40 Its presentation may be progressive (active or acute GVHD merging into chronic), quiescent (acute GVHD that resolves completely but is later followed by chronic GVHD) or it may occur de novo. Older recipient age and a history of acute GVHD are the greatest risk factors for chronic GVHD,41 and strategies to prevent acute GVHD may therefore help to prevent chronic GVHD. As shown in Table 2, the manifestations of chronic GVHD are somewhat protean, and are often of an autoimmune nature. Clinical signs often first appear in the buccal mucosa (see Figure 2). New consensus criteria for the diagnosis and staging of chronic GVHD have recently been developed.26

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0002.jpg
Chronic GVHD: Lichenoid changes of the buccal mucosa. Photograph courtesy of J. P. Guadagrini, D.D.S.

Table 2

Chronic GVHD Symptoms

SkinDyspigmentation, new onset alopecia, poikiloderma, lichen planus-like eruptions or sclerotic features
NailsNail dystrophy or loss
MouthXerostomia, ulcers, lichen-type features, restrictions of mouth opening from sclerosis
EyesDry eyes, sicca syndrome, cicatricial conjunctivitis
Muscles, fascia, jointsFasciitis, myositis, or joint stiffness from contractures
Female GenitaliaVaginal sclerosis, ulcerations
GI tractAnorexia, weight loss, esophageal web or strictures
LiverJaundice, transaminitis
LungsRestrictive or obstructive defects on pulmonary function tests, bronchiolitis obliterans, pleural effusions
KidneysNephrotic syndrome (rare)
HeartPericarditis
MarrowThrombocytopenia, anemia, neutropenia

Pathophysiology of Acute GVHD

Two important principles are important to consider regarding the pathophysiology of acute GVHD. First, acute GVHD reflects exaggerated but normal inflammatory mechanisms mediated by donor lymphocytes infused into the recipient where they function appropriately, given the foreign environment they encounter. Second, the recipient tissues that stimulate donor lymphocytes have usually been damaged by underlying disease, prior infections, and the transplant conditioning regimen.29 As a result, these tissues produce molecules (sometimes referred to as “danger” signals) that promote the activation and proliferation of donor immune cells.4245 Mouse models havebeen central to our identification and understanding of the pathophysiologic mechanisms of GVHD, and canine models have been critical to the development of clinically useful strategies for GVHD prophylaxis and treatment and to the development of donor leukocyte infusions.364647 Based largely on these experimental models, the development of acute GVHD can be conceptualized in three sequential steps or phases: (1) activation of the APCs; (2) donor T cell activation, proliferation, differentiation and migration; and (3) target tissue destruction (Figure 3).

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0003.jpg
GVHD Pathophysiology

In Phase I, the recipient conditioning regimen damages host tissues and causes release of inflammatory cytokines such as TNFα, IL-1 and IL-6. Increased levels of these cytokines leads to activation of host antigen presenting cells (APCs). In Phase II, host APCs activate mature donor cells. The subsequent proliferation and differentiation of these activated T cells produces additional effectors that mediate the tissue damage, including Cytotoxic T Lymphocytes, Natural Killer (NK) cells, TNFα and IL-1. Lipopolysaccharide (LPS) that has leaked through the damaged intestinal mucosa triggers additional TNFα production. TNFα can damage tissue directly by inducing necrosis and apoptosis in the skin and GI tract through either TNF receptors or the Fas pathway. TNFα plays a direct role in intestinal GVHD damage which further amplifies damage in the skin, liver and lung in a “cytokine storm.”

Phase I: Activation of Antigen Presenting Cells (APCs)

The first step involves the activation of APCs by the underlying disease and the HCT conditioning regimen. Damaged host tissues respond by producing “danger” signals, including proinflammatory cytokines (e.g., TNF-α), chemokines, and increased expression of adhesion molecules, MHC antigens and costimulatory molecules on host APCs.424850 A recent report demonstrated that at one week after HCT, increased levels of TNF-α receptor I, a surrogate marker for TNF-α, strongly correlated with the later development of GVHD.51 Damage to the GI tract from the conditioning is particularly important because it allows for systemic translocation of additional inflammatory stimuli such as microbial products including lipopolysaccaride (LPS) or other pathogen-associated molecular patterns that further enhance the activation of host APCs.49 The secondary lymphoid tissue in the GI tract is likely the initial site of interaction between activated APCs and donor T cells.52 These observations have led an important clinical strategy to reduce acute GVHD by reducing the intensity of the conditioning regimen. Experimental GVHD can also be reduced by manipulating distinct subsets of APCs.5354 In addition, non-hematopoietic stem cells, such as mesenchymal stem cells or stromal cells, can reduce allogeneic T cell responses, although the mechanism for such inhibition remains unclear.2

The concept that enhanced activation of host APCs increases the risk for acute GVHD unifies a number of seemingly disparate clinical associations with that risk, such as advanced stages of malignancy, more intense transplant conditioning regimens and histories of viral infections. APCs detect infections by recognizing conserved molecular patterns that are unique to microbes, called pathogen-associated molecular patterns (PAMPs). Among the classes of receptors that recognize such patterns, the Toll-like receptors (TLR) are the best characterized.55 For example, TLR4 recognizes LPS55 and mice with mutant TLR4 receptors that do not respond to LPS cause less GVHD when used as donors.56 Other TLRs that recognize viral DNA or RNA also activate APCs and may enhance GVHD, providing a potential mechanistic basis for increased GVHD associated with viral infections such as cytomegalovirus (CMV).57

Phase II: Donor T Cell Activation

The core of the GVH reaction is Step 2, where donor T cells proliferate and differentiate in response to host APCs. The “danger” signals generated in Phase I augment this activation at least in part by increasing the expression of costimulatory molecules.58 Blockade of co-stimulatory pathways to prevent GVHD is successful in animal models, but this approach has not yet been tested in large clinical trials.2

In mouse models, where genetic differences between donor and recipient strains can be tightly controlled, CD4 cells induce acute GVHD to MHC class II differences, and CD8 cells induce acute GVHD to MHC class I differences.5961 In the majority of HLA-identical HCTs, both CD4 and CD8 subsets respond to minor histocompatibility antigens and can cause GVHD in HLA-identical HCT.

Regulatory T cells can suppress the proliferation of conventional T cells and prevent GVHD in animal models when added to donor grafts containing conventional T cells.62 In mice, the Foxp3 protein functions as a master switch in the development of regulatory T cells, which normally constitute 5% of the CD4+ T cell population.62 Regulatory T cells secrete anti-inflammatory cytokines IL-10 and Transforming Growth Factor(TGF)-β and can also act through contact-dependent inhibition of APCs.62 It is likely that the use of regulatory T cells in clinical acute GVHD will require improved techniques to identify and expand them.

Natural Killer T cell (NKT) 1.1 subsets of both the host and donors that have been shown to modulate acute GVHD.63 Host NKT cells have been shown to suppress acute GVHD in an IL-4 dependent manner.64 A recent clinical trial of total lymphoid irradiation used as conditioning significantly reduced GVHD and enhanced host NKT cell function.65 By contrast, donor NKT cells can reduce GVHD and enhance perforin mediated GVL in an experimental model.66

Activation of immune cells results in rapid intracellular biochemical cascades that induce transcription of genes for many proteins including cytokines and their receptors. Th1 cytokines (IFN-γ, IL-2 and TNF-α) are produced in large amounts during acute GVHD. IL-2 production by donor T cells remains the principal target of many current clinical therapeutic and prophylactic approaches to GVHD, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) directed against IL-2 and its receptor.9 But emerging data indicate an important role for IL-2 in the generation and maintenance of CD4 CD25 T regs, suggesting that prolonged interference with IL-2 may have an unintended consequence of preventing the development of long term tolerance after allogeneic HCT.67 IFN-γ has multiple functions and can either amplify or reduce GVHD.6869 IFN-γ may amplify GVHD by increasing the expression of molecules such as chemokines receptors, MHC proteins, and adhesion molecules; it also increases the sensitivity of monocytes and macrophages to stimuli such as LPS and accelerates intracellular cascades in response to these stimuli.70 Early polarization of donor T cells so that they secrete less IFN-γ and more IL-4 can also attenuate experimental acute GVHD.71 IFN-γ may amplify GVHD by directly damaging epithelium in the GI tract and skin and inducing immnosuppression through the induction of nitric oxide.72 By contrast, IFN-γ may suppress GVHD by hastening the apoptosis of activated donor T cells.6973. This complexity means the manipulation of IFN-γ may have diverse effects in vivo, making it a challenging target with respect to therapeutic intervention. IL-10 plays a key role in suppression of immune responses, and clinical data suggest it may regulate acute GVHD.17 TGF-β, another suppressive cytokine can suppress acute GVHD but exacerbate chronic GVHD.74 Thus the timing and duration of the secretion of any given cytokine may determine the specific effects of that cytokine on GVHD severity.

Phase III: Cellular and Inflammatory Effector Phase

The effector phase of this process is a complex cascade of both cellular mediators such as cytotoxic T lymphocytes(CTLs) and NK cells and soluble inflammatory mediators such as TNF-α, IFN-γ, IL-1 and nitric oxide.229 These soluble and cellular mediators synergize to amplify local tissue injury and further promote inflammation and target tissue destruction.

Clinical Features of Acute GVHD

Based on an early Seattle experience, acute GVHD was defined to occur prior to day 100, whereas chronic GVHD occurred after that time.2325 This definition is far from satisfactory, and a recent National Institutes of Health classification includes late-onset acute GVHD (after day 100) and an overlap syndrome with features of both acute and chronic GVHD.26 Late-onset acute GVHD and the overlap syndrome occur with greater frequency after reduced-intensity conditioning (RIC), an increasingly widespread technique (see below). As shown in Table 1, the clinical manifestations of acute GVHD occur in the skin, gastrointestinal tract and liver.27 In a comprehensive review, Martin et al found that at the onset of acute GVHD, 81% of patients had skin involvement, 54% had GI involvement, and 50% had liver involvement.23 Recent data suggest that lungs might also be targets of experimental GVHD.28

Table 1

Acute GVHD Symptoms

Skinmaculopapular skin rash
upper GI tractnausea and/or anorexia PLUS positive histology
lower GI tractwatery diarrhea ≥ 500ml +/- severe abdominal pain +/- bloody diarrhea or ileus (after exclusion of infectious etiology)
Livercholestatic hyperbilirubinemia

Skin is most commonly affected and is usually the first organ involved, often coinciding with engraftment of donor cells. The characteristic maculopapular rash is pruritic and can spread throughout the body, sparing the scalp (Figure 1). In severe cases the skin may blister and ulcerate.27 Apoptosis at the base of epidermal rete pegs is a characteristic pathologic finding. Other features include dyskeratosis, exocytosis of lymphocytes, satellite lymphocytes adjacent to dyskeratotic epidermal keratinocytes, and a perivascular lymphocytic infiltration in the dermis.2930

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0001.jpg
Acute GVHD of the skin (Grade I). Photograph courtesy of J. Levine, M.D.

Gastrointestinal tract involvement of acute GVHD usually presents as diarrhea but may also include vomiting, anorexia, and/or abdominal pain when severe.29 The diarrhea of GVHD is secretory and often voluminous (greater than two liters per day). Bleeding, which carries a poor prognosis, occurs as a result of mucosal ulceration31 but patchy involvement of the mucosa often leads to a normal appearance on endoscopy.32

Radiologic findings of the gastrointestinal (GI) tract include luminal dilatation with thickening of the wall of the small bowel (“ribbon sign” on CT scan) and air/fluid levels suggestive of an ileus.27 Histologic features include patchy ulcerations, apoptotic bodies in the base of crypts, crypt abscesses, and loss as well as flattening of the surface epithelium.33

Liver disease caused by GVHD may be difficult to distinguish from other causes of liver dysfunction following BMT such as veno-occlusive disease, drug toxicity, viral infection, sepsis, or iron overload. The histologic features of hepatic GVHD are endothelialitis, lymphocytic infiltration of the portal areas, pericholangitis and bile duct destruction.3435 The liver is rarely biopsied because thrombocytopenia early after transplant greatly increases the risks of the procedure, usually making the diagnosis one of exclusion.

The incidence of the severity of acute GVHD is determined by the extent of involvement of these three principal target organs (see Table 1). The overall grades are classified as I (mild), II (moderate), III (severe) and IV (very severe). Severe GVHD carries a poor prognosis, with 25% long term survival for grade III and 5% for grade IV.36

The incidence of acute GVHD is directly related to the degree of mismatch between HLA proteins and ranges from 35-45% in recipients of full matched sibling donor grafts89 to 60-80% in recipients of one-antigen HLA mismatched unrelated donor grafts.63739 The same degree of mismatch causes less GVHD using UCB grafts and incidence of acute GVHD is lower following the transplant of partially matched UCB units and ranges from 35-65%.12

Clinical Features of Chronic GVHD

Chronic GVHD is the major cause of late non-relapse death following HCT.40 Its presentation may be progressive (active or acute GVHD merging into chronic), quiescent (acute GVHD that resolves completely but is later followed by chronic GVHD) or it may occur de novo. Older recipient age and a history of acute GVHD are the greatest risk factors for chronic GVHD,41 and strategies to prevent acute GVHD may therefore help to prevent chronic GVHD. As shown in Table 2, the manifestations of chronic GVHD are somewhat protean, and are often of an autoimmune nature. Clinical signs often first appear in the buccal mucosa (see Figure 2). New consensus criteria for the diagnosis and staging of chronic GVHD have recently been developed.26

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0002.jpg
Chronic GVHD: Lichenoid changes of the buccal mucosa. Photograph courtesy of J. P. Guadagrini, D.D.S.

Table 2

Chronic GVHD Symptoms

SkinDyspigmentation, new onset alopecia, poikiloderma, lichen planus-like eruptions or sclerotic features
NailsNail dystrophy or loss
MouthXerostomia, ulcers, lichen-type features, restrictions of mouth opening from sclerosis
EyesDry eyes, sicca syndrome, cicatricial conjunctivitis
Muscles, fascia, jointsFasciitis, myositis, or joint stiffness from contractures
Female GenitaliaVaginal sclerosis, ulcerations
GI tractAnorexia, weight loss, esophageal web or strictures
LiverJaundice, transaminitis
LungsRestrictive or obstructive defects on pulmonary function tests, bronchiolitis obliterans, pleural effusions
KidneysNephrotic syndrome (rare)
HeartPericarditis
MarrowThrombocytopenia, anemia, neutropenia

Pathophysiology of Acute GVHD

Two important principles are important to consider regarding the pathophysiology of acute GVHD. First, acute GVHD reflects exaggerated but normal inflammatory mechanisms mediated by donor lymphocytes infused into the recipient where they function appropriately, given the foreign environment they encounter. Second, the recipient tissues that stimulate donor lymphocytes have usually been damaged by underlying disease, prior infections, and the transplant conditioning regimen.29 As a result, these tissues produce molecules (sometimes referred to as “danger” signals) that promote the activation and proliferation of donor immune cells.4245 Mouse models havebeen central to our identification and understanding of the pathophysiologic mechanisms of GVHD, and canine models have been critical to the development of clinically useful strategies for GVHD prophylaxis and treatment and to the development of donor leukocyte infusions.364647 Based largely on these experimental models, the development of acute GVHD can be conceptualized in three sequential steps or phases: (1) activation of the APCs; (2) donor T cell activation, proliferation, differentiation and migration; and (3) target tissue destruction (Figure 3).

An external file that holds a picture, illustration, etc.
Object name is nihms-115970-f0003.jpg
GVHD Pathophysiology

In Phase I, the recipient conditioning regimen damages host tissues and causes release of inflammatory cytokines such as TNFα, IL-1 and IL-6. Increased levels of these cytokines leads to activation of host antigen presenting cells (APCs). In Phase II, host APCs activate mature donor cells. The subsequent proliferation and differentiation of these activated T cells produces additional effectors that mediate the tissue damage, including Cytotoxic T Lymphocytes, Natural Killer (NK) cells, TNFα and IL-1. Lipopolysaccharide (LPS) that has leaked through the damaged intestinal mucosa triggers additional TNFα production. TNFα can damage tissue directly by inducing necrosis and apoptosis in the skin and GI tract through either TNF receptors or the Fas pathway. TNFα plays a direct role in intestinal GVHD damage which further amplifies damage in the skin, liver and lung in a “cytokine storm.”

Phase I: Activation of Antigen Presenting Cells (APCs)

The first step involves the activation of APCs by the underlying disease and the HCT conditioning regimen. Damaged host tissues respond by producing “danger” signals, including proinflammatory cytokines (e.g., TNF-α), chemokines, and increased expression of adhesion molecules, MHC antigens and costimulatory molecules on host APCs.424850 A recent report demonstrated that at one week after HCT, increased levels of TNF-α receptor I, a surrogate marker for TNF-α, strongly correlated with the later development of GVHD.51 Damage to the GI tract from the conditioning is particularly important because it allows for systemic translocation of additional inflammatory stimuli such as microbial products including lipopolysaccaride (LPS) or other pathogen-associated molecular patterns that further enhance the activation of host APCs.49 The secondary lymphoid tissue in the GI tract is likely the initial site of interaction between activated APCs and donor T cells.52 These observations have led an important clinical strategy to reduce acute GVHD by reducing the intensity of the conditioning regimen. Experimental GVHD can also be reduced by manipulating distinct subsets of APCs.5354 In addition, non-hematopoietic stem cells, such as mesenchymal stem cells or stromal cells, can reduce allogeneic T cell responses, although the mechanism for such inhibition remains unclear.2

The concept that enhanced activation of host APCs increases the risk for acute GVHD unifies a number of seemingly disparate clinical associations with that risk, such as advanced stages of malignancy, more intense transplant conditioning regimens and histories of viral infections. APCs detect infections by recognizing conserved molecular patterns that are unique to microbes, called pathogen-associated molecular patterns (PAMPs). Among the classes of receptors that recognize such patterns, the Toll-like receptors (TLR) are the best characterized.55 For example, TLR4 recognizes LPS55 and mice with mutant TLR4 receptors that do not respond to LPS cause less GVHD when used as donors.56 Other TLRs that recognize viral DNA or RNA also activate APCs and may enhance GVHD, providing a potential mechanistic basis for increased GVHD associated with viral infections such as cytomegalovirus (CMV).57

Phase II: Donor T Cell Activation

The core of the GVH reaction is Step 2, where donor T cells proliferate and differentiate in response to host APCs. The “danger” signals generated in Phase I augment this activation at least in part by increasing the expression of costimulatory molecules.58 Blockade of co-stimulatory pathways to prevent GVHD is successful in animal models, but this approach has not yet been tested in large clinical trials.2

In mouse models, where genetic differences between donor and recipient strains can be tightly controlled, CD4 cells induce acute GVHD to MHC class II differences, and CD8 cells induce acute GVHD to MHC class I differences.5961 In the majority of HLA-identical HCTs, both CD4 and CD8 subsets respond to minor histocompatibility antigens and can cause GVHD in HLA-identical HCT.

Regulatory T cells can suppress the proliferation of conventional T cells and prevent GVHD in animal models when added to donor grafts containing conventional T cells.62 In mice, the Foxp3 protein functions as a master switch in the development of regulatory T cells, which normally constitute 5% of the CD4+ T cell population.62 Regulatory T cells secrete anti-inflammatory cytokines IL-10 and Transforming Growth Factor(TGF)-β and can also act through contact-dependent inhibition of APCs.62 It is likely that the use of regulatory T cells in clinical acute GVHD will require improved techniques to identify and expand them.

Natural Killer T cell (NKT) 1.1 subsets of both the host and donors that have been shown to modulate acute GVHD.63 Host NKT cells have been shown to suppress acute GVHD in an IL-4 dependent manner.64 A recent clinical trial of total lymphoid irradiation used as conditioning significantly reduced GVHD and enhanced host NKT cell function.65 By contrast, donor NKT cells can reduce GVHD and enhance perforin mediated GVL in an experimental model.66

Activation of immune cells results in rapid intracellular biochemical cascades that induce transcription of genes for many proteins including cytokines and their receptors. Th1 cytokines (IFN-γ, IL-2 and TNF-α) are produced in large amounts during acute GVHD. IL-2 production by donor T cells remains the principal target of many current clinical therapeutic and prophylactic approaches to GVHD, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) directed against IL-2 and its receptor.9 But emerging data indicate an important role for IL-2 in the generation and maintenance of CD4 CD25 T regs, suggesting that prolonged interference with IL-2 may have an unintended consequence of preventing the development of long term tolerance after allogeneic HCT.67 IFN-γ has multiple functions and can either amplify or reduce GVHD.6869 IFN-γ may amplify GVHD by increasing the expression of molecules such as chemokines receptors, MHC proteins, and adhesion molecules; it also increases the sensitivity of monocytes and macrophages to stimuli such as LPS and accelerates intracellular cascades in response to these stimuli.70 Early polarization of donor T cells so that they secrete less IFN-γ and more IL-4 can also attenuate experimental acute GVHD.71 IFN-γ may amplify GVHD by directly damaging epithelium in the GI tract and skin and inducing immnosuppression through the induction of nitric oxide.72 By contrast, IFN-γ may suppress GVHD by hastening the apoptosis of activated donor T cells.6973. This complexity means the manipulation of IFN-γ may have diverse effects in vivo, making it a challenging target with respect to therapeutic intervention. IL-10 plays a key role in suppression of immune responses, and clinical data suggest it may regulate acute GVHD.17 TGF-β, another suppressive cytokine can suppress acute GVHD but exacerbate chronic GVHD.74 Thus the timing and duration of the secretion of any given cytokine may determine the specific effects of that cytokine on GVHD severity.

Phase III: Cellular and Inflammatory Effector Phase

The effector phase of this process is a complex cascade of both cellular mediators such as cytotoxic T lymphocytes(CTLs) and NK cells and soluble inflammatory mediators such as TNF-α, IFN-γ, IL-1 and nitric oxide.229 These soluble and cellular mediators synergize to amplify local tissue injury and further promote inflammation and target tissue destruction.

Cellular Effectors

The cellular effectors of acute GVHD are primarily CTLs and NK cells.49 CTLs that preferentially use the Fas/FasL pathway of target lysis and appear to predominate in GVHD liver damage (hepatocytes express large amounts of Fas) whereas GVHD CTLs that use the perforin /granzyme pathways are more important in the GI tract and skin.275 Chemokines direct the migration of donor T cells from lymphoid tissues to the target organs where they cause damage. Macrophage inflammatory protein-1alpha (MIP-1α) and other chemokines such as CCL2-5, CXCL2, CXCL9-11, CCL17 and CCL27 are over-expressed and enhance the homing of cellular effectors to target organs during experimental GVHD.76 Expression of integrins, such as α4β7 and its ligand MadCAM-1, are also important for homing of donor T cells to Peyer's patches during intestinal GVHD.527778

Inflammatory Effectors

Microbial products such as LPS that leak through a damaged intestinal mucosa or skin may stimulate secretion of inflammatory cytokines through Toll-like receptors (TLRs).4979 The GI tract is particularly susceptible to damage from TNF-α, and plays a major role in the amplification and propagation of the “cytokine storm” characteristic of acute GVHD.49 TNF-α can be produced by both donor and host cells, and it acts in three different ways: first, it activates APCs and enhances alloantigen presentation; second, it recruits effector cells to target organs via the induction of inflammatory chemokines; and third, it directly causes tissue necrosis, as its name suggests.8082

Prevention of GVHD

Based on the evidence from animal models regarding the central role of T cells in initiating GVHD, numerous clinical studies evaluating T cell depletion (TCD) as prophylaxis for GVHD were performed in the 1980's and 1990's. There were three principal TCD strategies: (1) negative selection of T cells ex vivo, (2) positive selection of CD34 stem cells ex vivo; and (3) anti-T cell antibodies in vivo.83 Most strategies showed a significant limitation in both acute and chronic GVHD.8488 Unfortunately, the lower incidence of severe GVHD was offset by high rates of graft failure, relapse of malignancy, infections, and Epstein-Barr virus-associated lymphoproliferative disorders. Negative selection purging strategies using various anti-T cell antibodies achieved similar long-term results regardless of the breadth of antibody specificity.8993 One large registry study demonstrated that purging strategies using antibodies with broad specificities produced inferior leukemia-free survival than standard immunosuppression in patients receiving unrelated donor transplants.94 Several studies have investigated partial T cell depletion, either by eliminating specific T cell subsets (e.g., CD8) or by titrating the dose of T cells present in the inoculum.9597 None of these approaches, however, has convincingly demonstrated an optimal strategy that improves long-term survival.

Alemtuzumab is a monoclonal antibody that binds CD52, a protein expressed on a broad spectrum of leukocytes including lymphocytes, monocytes, and dendritic cells. Its use in GVHD prophylaxis in a Phase II trial decreased the incidence of acute and chronic GVHD following reduced intensity transplant.98 In two prospective studies, patients who received alemtuzumab rather than methotrexate showed significantly lower rates of acute and chronic GVHD,99 but experienced more infectious complications and higher rates of relapse, so that there was no overall survival benefit. Alemtuzumab may also contribute to graft failure when used with minimal intensity conditioning regimens.100

An alternative strategy to TCD attempted to induce anergy in donor T cells by ex vivo antibody blockade of co-stimulatory pathways prior to transplantation. A small study using this approach in haploidentical HCT recipients was quite encouraging, but has not yet been replicated.101 Thus the focus of most prevention strategies remains pharmacological manipulation of T cells after transplant.

Administration of anti-T cell antibodies in vivo as GVHD prophylaxis has also been extensively tested. The best studied drugs are anti-thymocyte globulin (ATG) or antilymphocyte globulin (ALG) preparations. These sera, which have high titers of polyclonal antibodies, are made by immunizing animals (horses or rabbits) to thymocytes or lymphocytes, respectively. A complicating factor in determining the role of these polyclonal sera in transplantation is the observation that even different brands of the same class of sera exert different biologic effects.102 However, the side effects of ATG/ALG infusions are common across different preparations and include fever, chills, headache, thrombocytopenia (from cross-reactivity to platelets), and, infrequently, anaphylaxis. In retrospective studies, rabbit ATG reduced the incidence of GVHD in related donor HSCT recipients without appearing to improve survival.103104 In recipients of unrelated donor HSCT, addition of ALG to standard GVHD prophylaxis effectively prevented severe GVHD, but did not result in improved survival because of increased infections.105 In a long term follow-up study, however, pretransplant ATG provided significant protection against extensive chronic GVHD and chronic lung dysfunction.106

The primary pharmacologic strategy to prevent GVHD is the inhibition of the cytoplasmic enzyme, calcineurin, that is critical for in the activation of T cells. The calcineurin inhibitors, cyclosporine and tacrolimus, have similar mechanisms of action, clinical effectiveness and toxicity profiles, including hypomagnesemia, hyperkalemia, hypertension, and nephrotoxicity.9107 Serious side effects include transplant-associated thrombotic microangiopathy (TAM) and neurotoxicity that can lead to premature discontinuation. Although clinically similar to thrombotic thrombocytopenic purpura, TAM does not reliably respond to therapeutic plasmapheresis, carries a high mortality rate, and removal of the offending agent does not always result in improvement.108 Posterior reversible encephalopathy syndrome includes mental status changes, seizures, neurological deficits and characteristic magnetic resonance imaging findings; this syndrome has been seen in 1-2% of HCT recipients receiving and calcineurin inhibitors.109 Side effects of these drugs decrease as the dose is tapered, usually two to four months after HCT.

Calcineurin inhibitors are often administered in combination with other immunosuppressants, such as methotrexate, which is given at low doses in the early post-transplant period.9107 The toxicities of methotrexate (neutropenia and mucositis) have led some investigators to replace it with mycophenolate mofetil (MMF). In one 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 methotrexate.110 The incidence and severity of acute GVHD was similar between the two groups, but the study closed early due to superiority of the MMF arm with respect to reduced mucositis and the speed of hematopoietic engraftment. A desire for faster neutrophil engraftment has led to the use of MMF in UCB blood transplants where graft failure is a major concern.111 MMF is also often used after RIC regimens for similar reasons.112113

Sirolimus is an immunosuppressant that is structurally similar to tacrolimus but does not inhibit calcineurin. In a small Phase II trial, it showed excellent efficacy in combination with tacrolimus;114 the drug damages endothelial cells, however, and it may enhance TAM that is associated with calcineurin inhibitors.115 The combination of tacrolimus and sirolimus is currently being compared in a large randomized multi-center trial.

RIC regimens attempt to suppress the host immune system sufficiently so that donor T cells can engraft and then ablate the lympho-hematopoietic compartment of the recipient. The term “non-myeloablative” is therefore somewhat misleading. RIC regimens produce less tissue damage and lower levels of the inflammatory cytokines that are important in the initiation of GVHD pathophysiology; this effect may explain the reduced incidence of severe GVHD following RIC compared to the full intensity conditioning used in historical controls.98116 The onset of acute GVHD may be delayed after RIC until after day 100, however, and it may present simultaneously with elements of chronic GVHD (“overlap syndrome”).116120

Treatment of Acute GVHD

GVHD generally first develops in the second month after HCT, during continued treatment with calcineurin-based prophylaxis.23121 Steroids, with their potent antilymphocyte and anti-inflammatory activity, are the gold standard for treatment of GVHD. Many centers treat mild GVHD of the skin (Grade I) with topical steroids alone, but for more severe skin GVHD and any degree of visceral GVHD involvement, high-dose systemic steroids are usually initiated. Steroid therapy results in complete remission in less than half of the patients,122 and more severe GVHD is less likely to respond to treatment.123124 In a prospective randomized study, the addition of ATG to steroids as primary therapy did not increase the response rate.124 In a retrospective study, the use of ATG in patients who showed early signs of steroid-resistance was beneficial,122 but not all studies show such benefit and ATG is not standardly used because of increased infection risks.106125126.

An increasingly common treatment for GVHD is extracorporeal photopheresis (ECP). During ECP, the patient's white blood cells are collected by apheresis, incubated with the DNA-intercalating agent, 8-methoxypsoralen, exposed to ultraviolet light (UVA), and returned to the patient. ECP is known to induce cellular apoptosis, which has strong anti-inflammatory effects in a number of systems, including prevention of rejection of solid organ grafts.127 Animal studies show that ECP reverses acute GVHD by increasing the number of regulatory T cells.128 A Phase II clinical study of steroid-dependent or steroid refractory GVHD showed resolution of GVHD in a large majority of patients, with 50% long-term survival in this very high risk group.129 Randomized multi-center studies of this approach are needed to determine its place in the management of acute GVHD.

Another interesting strategy to treat GVHD is the blockade of the inflammatory cytokine TNF-α. TNF-α can activate APCs, recruit effector cells and cause direct tissue damage.130 In animal models, TNF-α plays a central role in GVHD of the GI tract, which is central to the “cytokine storm” and plasma levels of TNFR I (a surrogate marker for TNF-α) rise in patients before the clinical manifestations of GVHD appear. 51 A recent Phase II trial of etanercept, a solubilized TNFR II, showed significant efficacy when added to systemic steroids as primary therapy for acute GVHD. Seventy percent of patients had complete resolution of all GVHD symptoms within one month, with 80% complete responses in the GI tract and the skin. The authors also showed that plasma levels of TNFR I were a significant biomarker for clinical GVHD.131

Treatment of Chronic GVHD

In contrast to acute GVHD, the pathophysiology of chronic GVHD remains poorly understood, and it is treated with a variety of immunosuppressive agents. The response of chronic GVHD to treatment is unpredictable, and mixed responses in different organs can occur in the same patient. Confounding variables such as infection and co-morbidities also make responses hard to measure. The use of corticosteroids (with or without a calcineurin inhibitor) is the standard of care, but a randomized trial of more than 300 patients with chronic GVHD found no difference between cyclosporine plus prednisone versus prednisone alone.132 Chronic immunosuppressants, especially those containing steroids, are highly toxic and result in infectious deaths. Many second line therapies have been studied, but none has achieved widespread acceptance. As mentioned above, ECP shows some promise, with significant response rates in high-risk patients. The best responses were observed in skin, liver, oral mucosa, eye, and lung.133 This observation is particularly relevant because lung GVHD has the potential to be a particularly devastating complication necessitating lung transplant as the only therapeutic option.134135

Essential Supportive Care in GVHD Patients

Meticulous supportive care is critical for patients with both acute and chronic GVHD because of the extended duration of immunosuppressive treatments and because the multiple medications required may have synergistic toxicities. Such care includes extensive infectious prophylaxis, early interventions in cases of suspected infections, and prophylaxis against non-infectious side effects of medications (See Table 3). These complications often require rapid responses to prevent serious or irreversible damage, and are best handled in close collaboration between the primary physician and the transplant specialist.

Table 3

Recommendations for Supportive Care

ComplicationClinical symptomsRoutine
Monitoring
ProphylaxisRecommended
Intervention

Bacterial infections:
Catheter-relatedFever, chills Pain and/or erythemaAssessment of catheter sitesSterile dressing, Regular line maintenanceObtain cultures Immediate IV antibiotic treatment, remove line

OtherFever, chills Sepsis symptomsClinical signs, Chest X ray or CT scan for possible pneumoniaAntibiotic prophylaxis in high risk patients (high dose corticosteroids or asplenism) IVIG if IgG level < 400 mg/dlObtain cultures Immediate IV broad spectrum antibiotics as risk of over-whelming sepsis within hours

Viral infections:
CMVGastroenteritis Interstitial pneumoniaBlood CMV PCR OR pp65 antigen levelsPreemptive treatment in patients with reactivationAntiviral therapy: Ganciclovir, Valganciclovir, or Foscarnet

Respiratory VirusesSymptoms of upper/lower respiratory tract infectionsClinical monitoringAnnual influenza vaccination against influenza (starting at 6 months post HCT) Vaccination of caregiversEarly treatment with neuraminidase inhibitors (Influenza), other antivirals

Varicella-zoster virusVesicular skin lesionsClinical monitoringAcyclovir prophylaxisTreatment doses of antivirals

Fungal and other infections:
Aspergillosis; other emerging fungal infectionsPulmonary lesions Sinusitis Skin nodulesGalactomannan assays in high risk patients CT scan if signs of infectionVoriconazole OR Posaconazole prophylaxis in high risk patients (eg, high dose steroids)Antifungal therapy

CandidaThrush Pulmonary lesionsClinical exam CT scan if signs of infectionFluconazole (Aspergillus prophylaxis protects against Candida as well)Antifungal therapy

PneumocystisFever Hypoxia Respiratory distressClinical assessmentCotrimoxazole OR Pentamadine until one month off ISTreatment doses of anti-PCP medications

Other Toxicities of immunosuppressive agents:
Calcineurin inhibitors (CNI)Tremor
Clinical assessment Drug levels
Adjust dose to desired trough levels

Neurotoxicity
Assess mental status

Stop CNI
Renal impairmentCreatinine levels &amp; GFRAdequate fluid uptake (~3L/day)IV fluids
HypertensionBlood pressure monitoringAntihypertensive treatment (ACE inhibitors, ß blocking agents)
Transplant associated microangiopathyAssess blood smear for hemolysis, schistocytesStop CNI Plasmapheresis

CorticosteroidsCushing's symptomsClinical assessmentTaper as recommended
DiabetesBlood glucose levelsNutritional guidanceInsulin treatment
Osteoporosis
Assessment of bone density
Calcium /Vit D supplementation
Bisphosphonate treatment if osteoporosis
MyopathyPhysiotherapy
Late graft failureBleeding symptoms AnemiaBlood countsStop or switch responsible drugsGrowth factors: G-CSF Erythropoetin Transfusions

All patients should receive at least fluconazole as prophylaxis against fungal infections. Invasive molds, especially aspergillus, are common in patients with prolonged steroid use.136 Prophylaxis with voriconazole or posaconazole should be considered for these patients. Usual sites of infection are the lungs, sinuses, brain, skin,137 and serial galactomannan assays may aid in the early detection.138 Candida can cause lesions in the lung and spleen, which may need screening with ultrasonography. Pneumocystis is another opportunistic infection that should receive cotrimoxazol (bactrim) prophylaxis.139

Viral infections are frequent in these patients with GVHD. Cytomegalovirus causes interstitial pneumonia and gastritis. Patients who are at risk should have their blood monitored several times monthly. Techniques that directly detect virus should be performed, such as CMV PCR or pp65 antigen, and evidence of increased viral load should prompt preemptive treatment with ganciclovir or foscarnet prior to clinical manifestations of disease. Shingles is not uncommon and acyclovir prophylaxis may be beneficial.140 Patients and caregivers should receive vaccinations against influenza, and treatment with neuraminidase inhibitors is recommended in the event of influenza infection.141142

Patients with GVHD often have IgG2 and IgG4 subclass deficiencies despite normal lgG levels, making them susceptible to infections with encapsulated organisms. Treatment of severe hypogammaglobulinemia with intravenous immunoglobulin is standard in many centers,143 but the level that triggers replacement varies considerably among transplant specialists. There is little supporting evidence for the routine use of intravenous immunoglobulin as prophylaxis144 but patients should receive routine prophylaxis (penicillin or its equivalent) due to the increased risk of streptococcal sepsis.145 Pneumococcal conjugate and hemophilus influenza vaccine may provide additional protection and are also recommended for all patients, including those with chronic GVHD.139146147 The sites of any indwelling catheters should be assessed regularly and early treatment of a suspected infection initiated. Early signs or symptoms of septic shock such as shaking chills or low blood pressure requires prompt evaluation with chest X-ray and/or CT scan, blood culture and broad spectrum antibiotics because shock may progress rapidly in these patients.

Chronic immunosuppressant therapy has multiple toxicities. Diabetes (which further increases the risks of infection), muscle weakness, osteoporosis, avascular necrosis (often requiring joint replacement) and other Cushingoid features are common with chronic steroid use. Frequent monitoring of blood glucose and screening bone density is recommended, and treatment includes insulin, calcium, Vitamin D and bisphosphonates.148149 Calcineurin inhibitors frequently cause renal impairment, hypertension, and neurologic paraesthesias. Standard supportive care includes blood pressure monitoring, assessment of renal function, and monitoring of drug blood levels, which should be maintained within therapeutic parameters. To prevent renal dysfunction, most centers recommend vigorous oral outpatient hydration. Some patients will be unable to tolerate calcineurin inhibitors and will require different immunosuppressive medications altogether. Cytopenias sometimes require treatment with growth factors such as G-CSF or cessation of the offending agent. These maneuvers should always be performed in close consultation with a transplant specialist.

Future Directions

As allogeneic transplantation becomes an increasingly attractive therapeutic option, the need for novel approaches to GVHD has accelerated. The number of patients receiving transplants from unrelated donors is expected to double in the next five years, significantly increasing the population of patients with GVHD. The advent of RIC regimens has reduced transplant-related mortality and lengthened the period during which acute GVHD may develop (many new cases present up to day 200) and the need for close monitoring of patients in this period has increased. Patients have often returned to the care of their primary hematologists by this time, increasing the need for these physicians to collaborate with transplant specialists in the management of GVHD and its complications. Identification of biomarkers for GVHD with diagnostic (and possibly prognostic) significance may eventually make the treatment of GVHD preemptive rather than prophylactic. The use of cellular component therapy, such as regulatory T cells that have been expanded ex vivo. will also enter clinical trials in the near future, but the extensive infrastructure required for such cellular approaches will likely limit their use initially to large academic centers, intensifying the need for close communication between transplant specialists and referring hematologists.

Search Strategy and Selection Criteria

Pubmed and Medline databases were searched using the search term GVHD that was thencross-referenced with each of the following words: clinical, cytokines, MHC and HLA antigens, biology and immunology. The criteria used to include mostly the peer-reviewed original and review journal articles published within the last decade except for those seminal articles that initially described the clinical features. All non-peer reviewed manuscripts, supplements and textbooks were excluded.

Prevention of GVHD

Based on the evidence from animal models regarding the central role of T cells in initiating GVHD, numerous clinical studies evaluating T cell depletion (TCD) as prophylaxis for GVHD were performed in the 1980's and 1990's. There were three principal TCD strategies: (1) negative selection of T cells ex vivo, (2) positive selection of CD34 stem cells ex vivo; and (3) anti-T cell antibodies in vivo.83 Most strategies showed a significant limitation in both acute and chronic GVHD.8488 Unfortunately, the lower incidence of severe GVHD was offset by high rates of graft failure, relapse of malignancy, infections, and Epstein-Barr virus-associated lymphoproliferative disorders. Negative selection purging strategies using various anti-T cell antibodies achieved similar long-term results regardless of the breadth of antibody specificity.8993 One large registry study demonstrated that purging strategies using antibodies with broad specificities produced inferior leukemia-free survival than standard immunosuppression in patients receiving unrelated donor transplants.94 Several studies have investigated partial T cell depletion, either by eliminating specific T cell subsets (e.g., CD8) or by titrating the dose of T cells present in the inoculum.9597 None of these approaches, however, has convincingly demonstrated an optimal strategy that improves long-term survival.

Alemtuzumab is a monoclonal antibody that binds CD52, a protein expressed on a broad spectrum of leukocytes including lymphocytes, monocytes, and dendritic cells. Its use in GVHD prophylaxis in a Phase II trial decreased the incidence of acute and chronic GVHD following reduced intensity transplant.98 In two prospective studies, patients who received alemtuzumab rather than methotrexate showed significantly lower rates of acute and chronic GVHD,99 but experienced more infectious complications and higher rates of relapse, so that there was no overall survival benefit. Alemtuzumab may also contribute to graft failure when used with minimal intensity conditioning regimens.100

An alternative strategy to TCD attempted to induce anergy in donor T cells by ex vivo antibody blockade of co-stimulatory pathways prior to transplantation. A small study using this approach in haploidentical HCT recipients was quite encouraging, but has not yet been replicated.101 Thus the focus of most prevention strategies remains pharmacological manipulation of T cells after transplant.

Administration of anti-T cell antibodies in vivo as GVHD prophylaxis has also been extensively tested. The best studied drugs are anti-thymocyte globulin (ATG) or antilymphocyte globulin (ALG) preparations. These sera, which have high titers of polyclonal antibodies, are made by immunizing animals (horses or rabbits) to thymocytes or lymphocytes, respectively. A complicating factor in determining the role of these polyclonal sera in transplantation is the observation that even different brands of the same class of sera exert different biologic effects.102 However, the side effects of ATG/ALG infusions are common across different preparations and include fever, chills, headache, thrombocytopenia (from cross-reactivity to platelets), and, infrequently, anaphylaxis. In retrospective studies, rabbit ATG reduced the incidence of GVHD in related donor HSCT recipients without appearing to improve survival.103104 In recipients of unrelated donor HSCT, addition of ALG to standard GVHD prophylaxis effectively prevented severe GVHD, but did not result in improved survival because of increased infections.105 In a long term follow-up study, however, pretransplant ATG provided significant protection against extensive chronic GVHD and chronic lung dysfunction.106

The primary pharmacologic strategy to prevent GVHD is the inhibition of the cytoplasmic enzyme, calcineurin, that is critical for in the activation of T cells. The calcineurin inhibitors, cyclosporine and tacrolimus, have similar mechanisms of action, clinical effectiveness and toxicity profiles, including hypomagnesemia, hyperkalemia, hypertension, and nephrotoxicity.9107 Serious side effects include transplant-associated thrombotic microangiopathy (TAM) and neurotoxicity that can lead to premature discontinuation. Although clinically similar to thrombotic thrombocytopenic purpura, TAM does not reliably respond to therapeutic plasmapheresis, carries a high mortality rate, and removal of the offending agent does not always result in improvement.108 Posterior reversible encephalopathy syndrome includes mental status changes, seizures, neurological deficits and characteristic magnetic resonance imaging findings; this syndrome has been seen in 1-2% of HCT recipients receiving and calcineurin inhibitors.109 Side effects of these drugs decrease as the dose is tapered, usually two to four months after HCT.

Calcineurin inhibitors are often administered in combination with other immunosuppressants, such as methotrexate, which is given at low doses in the early post-transplant period.9107 The toxicities of methotrexate (neutropenia and mucositis) have led some investigators to replace it with mycophenolate mofetil (MMF). In one 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 methotrexate.110 The incidence and severity of acute GVHD was similar between the two groups, but the study closed early due to superiority of the MMF arm with respect to reduced mucositis and the speed of hematopoietic engraftment. A desire for faster neutrophil engraftment has led to the use of MMF in UCB blood transplants where graft failure is a major concern.111 MMF is also often used after RIC regimens for similar reasons.112113

Sirolimus is an immunosuppressant that is structurally similar to tacrolimus but does not inhibit calcineurin. In a small Phase II trial, it showed excellent efficacy in combination with tacrolimus;114 the drug damages endothelial cells, however, and it may enhance TAM that is associated with calcineurin inhibitors.115 The combination of tacrolimus and sirolimus is currently being compared in a large randomized multi-center trial.

RIC regimens attempt to suppress the host immune system sufficiently so that donor T cells can engraft and then ablate the lympho-hematopoietic compartment of the recipient. The term “non-myeloablative” is therefore somewhat misleading. RIC regimens produce less tissue damage and lower levels of the inflammatory cytokines that are important in the initiation of GVHD pathophysiology; this effect may explain the reduced incidence of severe GVHD following RIC compared to the full intensity conditioning used in historical controls.98116 The onset of acute GVHD may be delayed after RIC until after day 100, however, and it may present simultaneously with elements of chronic GVHD (“overlap syndrome”).116120

Treatment of Acute GVHD

GVHD generally first develops in the second month after HCT, during continued treatment with calcineurin-based prophylaxis.23121 Steroids, with their potent antilymphocyte and anti-inflammatory activity, are the gold standard for treatment of GVHD. Many centers treat mild GVHD of the skin (Grade I) with topical steroids alone, but for more severe skin GVHD and any degree of visceral GVHD involvement, high-dose systemic steroids are usually initiated. Steroid therapy results in complete remission in less than half of the patients,122 and more severe GVHD is less likely to respond to treatment.123124 In a prospective randomized study, the addition of ATG to steroids as primary therapy did not increase the response rate.124 In a retrospective study, the use of ATG in patients who showed early signs of steroid-resistance was beneficial,122 but not all studies show such benefit and ATG is not standardly used because of increased infection risks.106125126.

An increasingly common treatment for GVHD is extracorporeal photopheresis (ECP). During ECP, the patient's white blood cells are collected by apheresis, incubated with the DNA-intercalating agent, 8-methoxypsoralen, exposed to ultraviolet light (UVA), and returned to the patient. ECP is known to induce cellular apoptosis, which has strong anti-inflammatory effects in a number of systems, including prevention of rejection of solid organ grafts.127 Animal studies show that ECP reverses acute GVHD by increasing the number of regulatory T cells.128 A Phase II clinical study of steroid-dependent or steroid refractory GVHD showed resolution of GVHD in a large majority of patients, with 50% long-term survival in this very high risk group.129 Randomized multi-center studies of this approach are needed to determine its place in the management of acute GVHD.

Another interesting strategy to treat GVHD is the blockade of the inflammatory cytokine TNF-α. TNF-α can activate APCs, recruit effector cells and cause direct tissue damage.130 In animal models, TNF-α plays a central role in GVHD of the GI tract, which is central to the “cytokine storm” and plasma levels of TNFR I (a surrogate marker for TNF-α) rise in patients before the clinical manifestations of GVHD appear. 51 A recent Phase II trial of etanercept, a solubilized TNFR II, showed significant efficacy when added to systemic steroids as primary therapy for acute GVHD. Seventy percent of patients had complete resolution of all GVHD symptoms within one month, with 80% complete responses in the GI tract and the skin. The authors also showed that plasma levels of TNFR I were a significant biomarker for clinical GVHD.131

Treatment of Chronic GVHD

In contrast to acute GVHD, the pathophysiology of chronic GVHD remains poorly understood, and it is treated with a variety of immunosuppressive agents. The response of chronic GVHD to treatment is unpredictable, and mixed responses in different organs can occur in the same patient. Confounding variables such as infection and co-morbidities also make responses hard to measure. The use of corticosteroids (with or without a calcineurin inhibitor) is the standard of care, but a randomized trial of more than 300 patients with chronic GVHD found no difference between cyclosporine plus prednisone versus prednisone alone.132 Chronic immunosuppressants, especially those containing steroids, are highly toxic and result in infectious deaths. Many second line therapies have been studied, but none has achieved widespread acceptance. As mentioned above, ECP shows some promise, with significant response rates in high-risk patients. The best responses were observed in skin, liver, oral mucosa, eye, and lung.133 This observation is particularly relevant because lung GVHD has the potential to be a particularly devastating complication necessitating lung transplant as the only therapeutic option.134135

Essential Supportive Care in GVHD Patients

Meticulous supportive care is critical for patients with both acute and chronic GVHD because of the extended duration of immunosuppressive treatments and because the multiple medications required may have synergistic toxicities. Such care includes extensive infectious prophylaxis, early interventions in cases of suspected infections, and prophylaxis against non-infectious side effects of medications (See Table 3). These complications often require rapid responses to prevent serious or irreversible damage, and are best handled in close collaboration between the primary physician and the transplant specialist.

Table 3

Recommendations for Supportive Care

ComplicationClinical symptomsRoutine
Monitoring
ProphylaxisRecommended
Intervention

Bacterial infections:
Catheter-relatedFever, chills Pain and/or erythemaAssessment of catheter sitesSterile dressing, Regular line maintenanceObtain cultures Immediate IV antibiotic treatment, remove line

OtherFever, chills Sepsis symptomsClinical signs, Chest X ray or CT scan for possible pneumoniaAntibiotic prophylaxis in high risk patients (high dose corticosteroids or asplenism) IVIG if IgG level < 400 mg/dlObtain cultures Immediate IV broad spectrum antibiotics as risk of over-whelming sepsis within hours

Viral infections:
CMVGastroenteritis Interstitial pneumoniaBlood CMV PCR OR pp65 antigen levelsPreemptive treatment in patients with reactivationAntiviral therapy: Ganciclovir, Valganciclovir, or Foscarnet

Respiratory VirusesSymptoms of upper/lower respiratory tract infectionsClinical monitoringAnnual influenza vaccination against influenza (starting at 6 months post HCT) Vaccination of caregiversEarly treatment with neuraminidase inhibitors (Influenza), other antivirals

Varicella-zoster virusVesicular skin lesionsClinical monitoringAcyclovir prophylaxisTreatment doses of antivirals

Fungal and other infections:
Aspergillosis; other emerging fungal infectionsPulmonary lesions Sinusitis Skin nodulesGalactomannan assays in high risk patients CT scan if signs of infectionVoriconazole OR Posaconazole prophylaxis in high risk patients (eg, high dose steroids)Antifungal therapy

CandidaThrush Pulmonary lesionsClinical exam CT scan if signs of infectionFluconazole (Aspergillus prophylaxis protects against Candida as well)Antifungal therapy

PneumocystisFever Hypoxia Respiratory distressClinical assessmentCotrimoxazole OR Pentamadine until one month off ISTreatment doses of anti-PCP medications

Other Toxicities of immunosuppressive agents:
Calcineurin inhibitors (CNI)Tremor
Clinical assessment Drug levels
Adjust dose to desired trough levels

Neurotoxicity
Assess mental status

Stop CNI
Renal impairmentCreatinine levels &amp; GFRAdequate fluid uptake (~3L/day)IV fluids
HypertensionBlood pressure monitoringAntihypertensive treatment (ACE inhibitors, ß blocking agents)
Transplant associated microangiopathyAssess blood smear for hemolysis, schistocytesStop CNI Plasmapheresis

CorticosteroidsCushing's symptomsClinical assessmentTaper as recommended
DiabetesBlood glucose levelsNutritional guidanceInsulin treatment
Osteoporosis
Assessment of bone density
Calcium /Vit D supplementation
Bisphosphonate treatment if osteoporosis
MyopathyPhysiotherapy
Late graft failureBleeding symptoms AnemiaBlood countsStop or switch responsible drugsGrowth factors: G-CSF Erythropoetin Transfusions

All patients should receive at least fluconazole as prophylaxis against fungal infections. Invasive molds, especially aspergillus, are common in patients with prolonged steroid use.136 Prophylaxis with voriconazole or posaconazole should be considered for these patients. Usual sites of infection are the lungs, sinuses, brain, skin,137 and serial galactomannan assays may aid in the early detection.138 Candida can cause lesions in the lung and spleen, which may need screening with ultrasonography. Pneumocystis is another opportunistic infection that should receive cotrimoxazol (bactrim) prophylaxis.139

Viral infections are frequent in these patients with GVHD. Cytomegalovirus causes interstitial pneumonia and gastritis. Patients who are at risk should have their blood monitored several times monthly. Techniques that directly detect virus should be performed, such as CMV PCR or pp65 antigen, and evidence of increased viral load should prompt preemptive treatment with ganciclovir or foscarnet prior to clinical manifestations of disease. Shingles is not uncommon and acyclovir prophylaxis may be beneficial.140 Patients and caregivers should receive vaccinations against influenza, and treatment with neuraminidase inhibitors is recommended in the event of influenza infection.141142

Patients with GVHD often have IgG2 and IgG4 subclass deficiencies despite normal lgG levels, making them susceptible to infections with encapsulated organisms. Treatment of severe hypogammaglobulinemia with intravenous immunoglobulin is standard in many centers,143 but the level that triggers replacement varies considerably among transplant specialists. There is little supporting evidence for the routine use of intravenous immunoglobulin as prophylaxis144 but patients should receive routine prophylaxis (penicillin or its equivalent) due to the increased risk of streptococcal sepsis.145 Pneumococcal conjugate and hemophilus influenza vaccine may provide additional protection and are also recommended for all patients, including those with chronic GVHD.139146147 The sites of any indwelling catheters should be assessed regularly and early treatment of a suspected infection initiated. Early signs or symptoms of septic shock such as shaking chills or low blood pressure requires prompt evaluation with chest X-ray and/or CT scan, blood culture and broad spectrum antibiotics because shock may progress rapidly in these patients.

Chronic immunosuppressant therapy has multiple toxicities. Diabetes (which further increases the risks of infection), muscle weakness, osteoporosis, avascular necrosis (often requiring joint replacement) and other Cushingoid features are common with chronic steroid use. Frequent monitoring of blood glucose and screening bone density is recommended, and treatment includes insulin, calcium, Vitamin D and bisphosphonates.148149 Calcineurin inhibitors frequently cause renal impairment, hypertension, and neurologic paraesthesias. Standard supportive care includes blood pressure monitoring, assessment of renal function, and monitoring of drug blood levels, which should be maintained within therapeutic parameters. To prevent renal dysfunction, most centers recommend vigorous oral outpatient hydration. Some patients will be unable to tolerate calcineurin inhibitors and will require different immunosuppressive medications altogether. Cytopenias sometimes require treatment with growth factors such as G-CSF or cessation of the offending agent. These maneuvers should always be performed in close consultation with a transplant specialist.

Future Directions

As allogeneic transplantation becomes an increasingly attractive therapeutic option, the need for novel approaches to GVHD has accelerated. The number of patients receiving transplants from unrelated donors is expected to double in the next five years, significantly increasing the population of patients with GVHD. The advent of RIC regimens has reduced transplant-related mortality and lengthened the period during which acute GVHD may develop (many new cases present up to day 200) and the need for close monitoring of patients in this period has increased. Patients have often returned to the care of their primary hematologists by this time, increasing the need for these physicians to collaborate with transplant specialists in the management of GVHD and its complications. Identification of biomarkers for GVHD with diagnostic (and possibly prognostic) significance may eventually make the treatment of GVHD preemptive rather than prophylactic. The use of cellular component therapy, such as regulatory T cells that have been expanded ex vivo. will also enter clinical trials in the near future, but the extensive infrastructure required for such cellular approaches will likely limit their use initially to large academic centers, intensifying the need for close communication between transplant specialists and referring hematologists.

Search Strategy and Selection Criteria

Pubmed and Medline databases were searched using the search term GVHD that was thencross-referenced with each of the following words: clinical, cytokines, MHC and HLA antigens, biology and immunology. The criteria used to include mostly the peer-reviewed original and review journal articles published within the last decade except for those seminal articles that initially described the clinical features. All non-peer reviewed manuscripts, supplements and textbooks were excluded.

Acknowledgements

This work was supported in part by grants from the National Institutes of Health (Bethesda, MD), P01 CA 39542, and the Jose Carreras Foundation. James L.M. Ferrara, M. D. is a Doris Duke Distinguished Clinical Scientist and an American Cancer Society Professor.

University of Michigan, Pediatrics and Internal Medicine, Blood and Marrow Transplantation Program
University of Michigan, Pediatrics and Internal Medicine, Blood and Marrow Transplantation Program
University of Michigan, Internal Medicine, Blood and Marrow Transplantation Program
Internal Medicine and Department of Haematology/Oncology, University Hospital, Regensburg, Germany
Corresponding Author: James L.M. Ferrara, M.D., The University of Michigan, 1500 East Medical Center Drive, 6303 CCC, Ann Arbor, MI 48109-5942, Telephone: (734)615-1340, FAX (734)615-4947, email: ude.hcimu@ararref.

References

  • 1. Appelbaum FRHaematopoietic cell transplantation as immunotherapy. Nature. 2001;411:385–389.[PubMed][Google Scholar]
  • 2. Welniak LA, Blazar BR, Murphy WJImmunobiology of allogeneic hematopoietic stem cell transplantation. Annu Rev Immunol. 2007;25:139–170.[PubMed][Google Scholar]
  • 3. Billingham REThe biology of graft-versus-host reactions. Harvey Lect. 1966-67;62:21–78.[PubMed][Google Scholar]
  • 4. Korngold R, Sprent J. Purified T cell subsets and lethal graft-versus-host disease in mice. In: Gale RP, Champlin R, editors. Progress in Bone Marrow Transplantation. Alan R. Liss, Inc.; New York: 1987. pp. 213–218. [PubMed]
  • 5. Kernan NA, Collins NH, Juliano LL, Cartagena TT, Dupont BB, OReilly RJClonable T lymphocytes in T cell-depleted bone marrow transplants correlate with development of graft-v-host disease. Blood. 1986;68:770–773.[PubMed][Google Scholar]
  • 6. Petersdorf EW, Longton GM, Anasetti C, et al The significance of HLA-DRB1 matching on clinical outcome after HLA-A, B, DR identical unrelated donor marrow transplantation. Blood. 1995;1995;86:1606–1613.[PubMed][Google Scholar]
  • 7. Krensky AM, Weiss A, Crabtree G, Davis MM, Parham PT-lymphocyte-antigen interactions in transplant rejection. N Engl J Med. 1990;322:510–517.[PubMed][Google Scholar]
  • 8. Loiseau P, Busson M, Balere ML, et al HLA Association with hematopoietic stem cell transplantation outcome: the number of mismatches at HLA-A, -B, -C, -DRB1, or -DQB1 is strongly associated with overall survival. Biol Blood Marrow Transplant. 2007;13:965–974.[PubMed][Google Scholar]
  • 9. Ratanatharathorn V, Nash RA, Przepiorka D, et al Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation. Blood. 1998;92:2303–2314.[PubMed][Google Scholar]
  • 10. Cornetta K, Laughlin M, Carter S, et al Umbilical cord blood transplantation in adults: results of the prospective Cord Blood Transplantation (COBLT) Biol Blood Marrow Transplant. 2005;11:149–160.[PubMed][Google Scholar]
  • 11. Wagner JE, Barker JN, DeFor TE, et al Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood. 2002;100:1611–1618.[PubMed][Google Scholar]
  • 12. Barker JN, Wagner JEUmbilical-cord blood transplantation for the treatment of cancer. Nat Rev Cancer. 2003;3:526–532.[PubMed][Google Scholar]
  • 13. Bleakley MR, S. R. Molecules and mechanisms of the graft-versus-leukaemia effect. Nature Rev Cancer. 2004;4:371–380.[PubMed]
  • 14. Goulmy E, Schipper R, Pool J, et al Mismatches of minor histocompatibility antigens between HLA-identical donors and recipients and the development of graft-versus-host disease after bone marrow transplantation. N Engl J Med. 1996;334:281–285.[PubMed][Google Scholar]
  • 15. Antin JH, Ferrara JLCytokine dysregulation and acute graft-versus-host disease. Blood. 1992;80:2964.[PubMed][Google Scholar]
  • 16. Cavet J, Middleton PG, Segall M, Noreen H, Davies SM, Dickinson AMRecipient tumor necrosis factor-alpha and interleukin-10 gene polymorphisms associate with early mortality and acute graft-versus-host disease severity in HLA-matched sibling bone marrow transplants. Blood. 1999;94:3941–32946.[PubMed][Google Scholar]
  • 17. Lin MT, Storer B, Martin PJ, et al Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation. N Engl J Med. 2003;349:2201–2210.[PubMed][Google Scholar]
  • 18. Dickinson AM, Charron DNon-HLA immunogenetics in hematopoietic stem cell transplantation. Curr Opin Immunol. 2005;17:517–525.[PubMed][Google Scholar]
  • 19. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–3057.[PubMed][Google Scholar]
  • 20. Miller JS, Cooley S, Parham P, et al Missing KIR ligands are associated with less relapse and increased graft-versus-host disease (GVHD) following unrelated donor allogeneic HCT. Blood. 2007;109:5058–5061.[Google Scholar]
  • 21. Velardi A, Ruggeri L, Alessandro L, Moretta, Moretta NK cells: a lesson from mismatched hematopoietic transplantation. Trends Immunology. 2002;23:438–444.[PubMed]
  • 22. Holler E, Rogler G, Brenmoehl J, et al Prognostic significance of NOD2/CARD15 variants in HLA-identical sibling hematopoietic stem cell transplantation: effect on long-term outcome is confirmed in 2 independent cohorts and may be modulated by the type of gastrointestinal decontamination. Blood. 2006;107:4189–4193.[PubMed][Google Scholar]
  • 23. Martin PJ, Schoch G, Fisher L, et al A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment. Blood. 1990;76:1464–1472.[PubMed][Google Scholar]
  • 24. Sullivan KM, Agura E, Anasetti C, et al Chronic graft-versus-host disease and other late complications of bone marrow transplantation. Semin Hemat. 1991;28:250–259.[PubMed][Google Scholar]
  • 25. Sullivan KM, Mori M, Sanders J, et al Late complications of allogeneic and autologous marrow transplantation. Bone Marrow Transplant. 1992;10:127–134.[PubMed][Google Scholar]
  • 26. 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]
  • 27. Vogelsang GB, Lee L, Bensen-Kennedy DMPathogenesis and treatment of graft-versus-host disease after bone marrow transplant. Annu Rev Med. 2003;54:29–52.[PubMed][Google Scholar]
  • 28. Cooke KR, Kobzik LL, Martin TR, et al. An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood. 1996;88:3230–9.[PubMed]
  • 29. Ferrara JL, Deeg HJGraft-versus-host disease. N Engl J Med. 1991;324:667–674.[PubMed][Google Scholar]
  • 30. Goker H, Haznedaroglu IC, Chao NJAcute graft-vs-host disease: pathobiology and management. Exp Hematol. 2001;29:259–277.[PubMed][Google Scholar]
  • 31. Nevo S, Enger C, Swan V, et al Acute bleeding after allogeneic bone marrow transplantation: association with graft versus host disease and effect on survival. Transplant. 1999;67:681–689.[PubMed][Google Scholar]
  • 32. Ponec RJ, Hackman RC, McDonald GBEndoscopic and histologic diagnosis of intestinal graft-versus-host disease after marrow transplantation. Gastrointest Endosc. 1999;49:612–621.[PubMed][Google Scholar]
  • 33. Snover DC, Weisdorf SA, Vercellotti GM, Rank B, Hutton S, McGlave PA histopathologic study of gastric and small intestinal graft-versus-host disease following allogeneic bone marrow transplantation. Hum Pathol. 1985;16:387–392.[PubMed][Google Scholar]
  • 34. Snover DC, Weisdorf SA, Ramsay NK, McGlave P, Kersey JHHepatic graft versus host disease: a study of the predictive value of liver biopsy in diagnosis. Hepatology. 1984;4:123–130.[PubMed][Google Scholar]
  • 35. Fujii N, Takenaka K, Shinagawa K, et al Hepatic graft-versus-host disease presenting as an acute hepatitis after allogeneic peripheral blood stem cell transplantation. Bone Marrow Transplant. 2001;27:1007–1010.[PubMed][Google Scholar]
  • 36. Cahn JY, Klein JP, Lee SJ, et al Prospective evaluation of 2 acute graft-versus-host (GVHD) grading systems: a joint Societe Francaise de Greffe de Moelle et Therapie Cellulaire (SFGM-TC), Dana Farber Cancer Institute (DFCI), and International Bone Marrow Transplant Registry (IBMTR) prospective study. Blood. 2005;106:1495–1500.[Google Scholar]
  • 37. Anasetti C, Rybka W, Sullivan KM, Banaji M, Slichter SJGraft-v-host disease is associated with autoimmune-like thrombocytopenia. Blood. 1989;73:1054–1058.[PubMed][Google Scholar]
  • 38. Petersdorf EW, Longton G, Anasetti C, et al Donor-recipient disparities for HLAC genes is a risk factor for graft failure following marrow transplantation from unrelated donors. Blood. 1995;86:1606–1613.[PubMed][Google Scholar]
  • 39. Flomenberg N, Baxter-Lowe LA, Confer D, Fernandez-Vina M, et al Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome. Blood. 2004;104:1923–1930.[PubMed][Google Scholar]
  • 40. 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][Google Scholar]
  • 41. Carlens S, Ringden O, Remberger M, Lonnqvist B, Hagglund H, Klaesson S, et al Risk factors for chronic graft-versus-host disease after bone marrow transplantation: a retrospective single centre analysis. Bone Marrow Transplant. 1998;22:755–761.[PubMed][Google Scholar]
  • 42. Matzinger PThe danger model: a renewed sense of self. Science. 2002;296:301–305.[PubMed][Google Scholar]
  • 43. Shlomchik WD, Couzens MS, Tang CB, et al Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 1999;285:412–415.[PubMed][Google Scholar]
  • 44. Teshima T, Ordemann R, Reddy P, et al Acute graft-versus-host disease does not require alloantigen expression on host epithelium. Nat Med. 2002;8:575–581.[PubMed][Google Scholar]
  • 45. Reddy P, Maeda Y, Liu C, Krijanovski OI, Korngold R, Ferrara JLA crucial role for antigen-presenting cells and alloantigen expression in graft-versus-leukemia responses. Nat Med. 2005;11:1244–1249.[PubMed][Google Scholar]
  • 46. Kolb HJ, Holler EAdoptive immunotherapy with donor lymphocyte transfusions. Curr Opin Oncol. 1997;9:139–145.[PubMed][Google Scholar]
  • 47. Storb R, Rudolph RH, Thomas EDMarrow grafts between canine siblings matched by serotyping and mixed leukocyte culture. J Clin Invest. 1971;50:1272–1275.[Google Scholar]
  • 48. Xun CQ, Thompson JS, Jennings CD, Brown SA, Widmer MBEffect of total body irradiation, busulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-versus-host disease in H-2-incompatible transplanted SCID mice. Blood. 1994;83:2360–2367.[PubMed][Google Scholar]
  • 49. Hill GR, Ferrara JLThe primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 2000;95:2754–2759.[PubMed][Google Scholar]
  • 50. Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JLTotal body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood. 1997;90:3204–3213.[PubMed][Google Scholar]
  • 51. Choi SW, Kitko CL, Braun T, et al Change plasma tumor necrosis factor recept-1 levels in the first week post-myeloablative allogeneic transplant correlated with severity and incidence of GVHD and survival. Blood. (In Press)
  • 52. Murai M, Yoneyama H, Ezaki T, et al Peyer's patch is the essential site in initiating murine acute and lethal graft-versus-host reaction. Nat Immunol. 2003;4:154–160.[PubMed][Google Scholar]
  • 53. Duffner UA, Maeda Y, Cooke KR, et al Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease. J Immunol. 2004;172:7393–7398.[PubMed][Google Scholar]
  • 54. Sato K, Yamashita N, Baba M, Matsuyama TRegulatory dendritic cells protect mice from murine acute graft-versus-host disease and leukemia relapse. Immunity. 2003;18:367–379.[PubMed][Google Scholar]
  • 55. Medzhitov R, Janeway CA., Jr Decoding the patterns of self and nonself by the innate immune system. Science. 2002;296:298–300.[PubMed]
  • 56. Cooke KR, Gerbitz A, Crawford JM, et al LPS antagonism reduces graft-versus-host disease and preserves graft-versus-leukemia activity after experimental bone marrow transplantation. J Clin Invest. 2001;107:1581–1589.[Google Scholar]
  • 57. Akira S, Uematsu S, Takeuchi OPathogen recognition and innate immunity. Cell. 2006;124:783–801.[PubMed][Google Scholar]
  • 58. Dustin MLRole of adhesion molecules in activation signaling in T lymphocytes. J Clin Immunol. 2001;21:258–263.[PubMed][Google Scholar]
  • 59. Csencsits KL, Bishop DKContrasting alloreactive CD4+ and CD8+ T cells: there's more to it than MHC restriction. Am J Transplant. 2003;3:107–115.[PubMed][Google Scholar]
  • 60. Korngold R, Sprent JFeatures of T cells causing H-2-restricted lethal graft-vs.-host disease across minor histocompatibility barriers. J Exp Med. 1982;155:872–883.[Google Scholar]
  • 61. Korngold R, Sprent JSurface markers of T cells causing lethal graft-vs-host disease to class I vs class II H-2 differences. J Immunol. 1985;135:3004–3010.[PubMed][Google Scholar]
  • 62. Cohen JL, Boyer OThe role of CD4+CD25hi regulatory T cells in the physiopathogeny of graft-versus-host disease. Curr Opin Immunol. 2006;18:580–585.[PubMed][Google Scholar]
  • 63. Zeng D, Lewis D, Dejbakhsh-Jones S, et al Bone marrow NK1.1(-) and NK1.1(+) T cells reciprocally regulate acute graft versus host disease. J Exp Med. 1999;189:1073–1081.[Google Scholar]
  • 64. Zeng D, Dejbakhsh-Jones S, Strober SGranulocyte Colony-Stimulating Factor Reduces the Capacity of Blood Mononuclear Cells to Induce Graft-Versus-Host Disease: Impact on Blood Progenitor Cell Transplantation. Blood. 1997;90:453–463.[PubMed][Google Scholar]
  • 65. Lowsky R, Takahashi T, Liu YP, et al Protective conditioning for acute graft-versus-host disease. N Engl J Med. 2005;353:1321–1331.[PubMed][Google Scholar]
  • 66. Morris ES, MacDonald KP, Rowe V, et al NKT cell-dependent leukemia eradication following stem cell mobilization with potent G-CSF analogs. J Clin Invest. 2005;115:3093–3103.[Google Scholar]
  • 67. Zeiser R, Nguyen VH, Beilhack A, et al Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood. 2006;108:390–399.[Google Scholar]
  • 68. Fowler DH, Kurasawa K, Smith R, Eckhaus MA, Gress REDonor CD4-enriched cells of Th2 cytokine phenotype regulate graft-versus-host disease without impairing allogeneic engraftment in sublethally irradiated mice. Blood. 1994;84:3540–3549.[PubMed][Google Scholar]
  • 69. Yang YG, Dey BR, Sergio JJ, Pearson DA, Sykes MDonor-derived interferon gamma is required for inhibition of acute graft-versus-host disease by interleukin 12. J Clin Invest. 1998;102:2126–2135.[Google Scholar]
  • 70. Ferrara JLM Graft-versus-host disease. W.B. Saunders Company; Philadelphia: 2006. [PubMed][Google Scholar]
  • 71. Ferrara JL, Reddy PPathophysiology of graft-versus-host disease. Semin Hematol. 2006;43:3–10.[PubMed][Google Scholar]
  • 72. Krenger W, Falzarano G, Delmonte J, Jr., Snyder KM, Byon JC, Ferrara JLInterferon-gamma suppresses T-cell proliferation to mitogen via the nitric oxide pathway during experimental acute graft-versus-host disease. Blood. 1996;88:1113–1121.[PubMed][Google Scholar]
  • 73. Reddy P, Teshima T, Kukuruga M, et al Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis. J Exp Med. 2001;194:1433–1440.[Google Scholar]
  • 74. Banovic T, MacDonald KP, Morris ES, et al TGF-beta in allogeneic stem cell transplantation: friend or foe? Blood. 2005;106:2206–2214.[PubMed][Google Scholar]
  • 75. van den Brink MR, Burakoff SJCytolytic pathways in haematopoietic stem-cell transplantation. Nat Rev Immunol. 2002;2:273–281.[PubMed][Google Scholar]
  • 76. Wysocki CA, Panoskaltsis-Mortari A, Blazar BR, Serody JSLeukocyte migration and graft-versus-host disease. Blood. 2005;105:4191–4199.[Google Scholar]
  • 77. Waldman E, Lu SX, Hubbard VM, et al Absence of beta7 integrin results in less graft-versus-host disease because of decreased homing of alloreactive T cells to intestine. Blood. 2006;107:1703–1711.[Google Scholar]
  • 78. Welniak LA, Kuprash DV, Tumanov AV, et al Peyer's patches are not required for acute graft-versus-host disease after myeloablative conditioning and murine allogeneic bone marrow transplantation. Blood. 2005;107:410–412.[Google Scholar]
  • 79. Iwasaki A, Medzhitov RToll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5:987–995.[PubMed][Google Scholar]
  • 80. Hill GR, Teshima T, Rebel VI, et al The p55 TNF-alpha receptor plays a critical role in T cell alloreactivity. J Immunol. 2000;164:656–663.[PubMed][Google Scholar]
  • 81. Piguet PF, Grau GE, Allet B, Vassalli PTumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft-vs.-host disease. J Exp Med. 1987;166:1280–1289.[Google Scholar]
  • 82. Brown GR, Lee E, Thiele DLTNF-TNFR2 interactions are critical for the development of intestinal graft-versus-host disease in MHC class II-disparate (C57BL/6J-->C57BL/6J x bm12)F1 mice. J Immunol. 2002;168:3065–3071.[PubMed][Google Scholar]
  • 83. Soiffer R T-Cell Depletion to Prevent Graft-vs-host Disease. 3 ed. Blackwell; Carlton: 2008. [PubMed][Google Scholar]
  • 84. Marmont AM, Horowitz M, Gale RP, et al T-cell depletion of HLA-identical transplants in leukemia. Blood. 1991;78:2120–2130.[PubMed][Google Scholar]
  • 85. O'Reilly RJ, Brochstein JA, Collins NH, et al Evaluation of HLA-haplotype disparate parental marrow grafts depleted of T lymphocytes by differential agglutination with a soybean lectin and E-rosette depletion for the treatment of severe combined immunodeficiency. Vox Sang. 1986;51:81–86.[PubMed][Google Scholar]
  • 86. Platzbecker U, Ehninger G, Bornhäuser MAllogeneic transplantation of CD34+ selected hematopoietic cells--clinical problems and current challenges. Leukemia &amp; Lymphoma. 2004;45:447–453.[PubMed][Google Scholar]
  • 87. Urbano-Ispizua A, Rozman C, Martínez C, et al Rapid engraftment without significant graft-versus-host disease after allogeneic transplantation of CD34+ selected cells from peripheral blood. Blood. 1997;89:3967–3973.[PubMed][Google Scholar]
  • 88. Wagner JE, Thompson JS, Carter SL, Kernan NAEffect of graft-versus-host disease prophylaxis on 3-year disease-free survival in recipients of unrelated donor bone marrow (T-cell Depletion Trial): a multi-centre, randomised phase IIIII trial. Lancet. 2005;366:733–741.[PubMed][Google Scholar]
  • 89. Ash RC, Casper JT, Chitambar CR, et al Successful allogeneic transplantation of T-cell-depleted bone marrow from closely HLA-matched unrelated donors. N Engl J Med. 1990;322:485–494.[PubMed][Google Scholar]
  • 90. Bacigalupo AA, Lamparelli TT, Bruzzi PP, et al Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO) Blood. 2001;98:2942–2947.[PubMed][Google Scholar]
  • 91. Barge RMY, Starrenburg CWJ, Falkenburg JHF, Fibbe WE, Marijt EW, Willemze RRLong-term follow-up of myeloablative allogeneic stem cell transplantation using Campath "in the bag" as T-cell depletion: the Leiden experience. Bone Marrow Transplant. 2006;37:1129–1134.[PubMed][Google Scholar]
  • 92. Martin PJ, Hansen JA, Torok-Storb B, et al Effects of treating marrow with a CD3-specific immunotoxin for prevention of acute graft-versus-host disease. Bone Marrow Transplant. 1988;3:437–44.[PubMed][Google Scholar]
  • 93. Ringdén O, Remberger M, Aschan J, Lungman P, Lönnqvist B, Markling LLong-term follow-up of a randomized trial comparing T cell depletion with a combination of methotrexate and cyclosporine in adult leukemic marrow transplant recipients. Transplantation. 1994;58:887–891.[PubMed][Google Scholar]
  • 94. Champlin RE, Passweg JR, Zhang MJ, et al T-cell depletion of bone marrow transplants for leukemia from donors other than HLA-identical siblings: advantage of T-cell antibodies with narrow specificities. Blood. 2000;95:3996–4003.[PubMed][Google Scholar]
  • 95. Chalandon Y, Roosnek E, Mermillod B, Waelchli L, Helg C, Chapuis BCan only partial T-cell depletion of the graft before hematopoietic stem cell transplantation mitigate graft-versus-host disease while preserving a graft-versus-leukemia reaction? A prospective phase II study. Biol Blood Marrow Transplant. 2006;12:102–110.[PubMed][Google Scholar]
  • 96. Goerner M, Weber-Nordt R, Hoepfner S, Benner A, Luft T, Ho ADAddition of a low fixed number of CD3+ cells to CD34-enriched allografts: effects on engraftment, graft-versus-host disease, and survival after related and unrelated peripheral stem cell transplantation. J Hematotherapy &amp; stem cell research. 2003;12:309–320.[PubMed][Google Scholar]
  • 97. Ho VT, Kim HT, Li S, et al Partial CD8+ T-cell depletion of allogeneic peripheral blood stem cell transplantation is insufficient to prevent graft-versus-host disease. Bone Marrow Transplant. 2004;34:987–994.[PubMed][Google Scholar]
  • 98. Perez-Simon JA, Diez-Campelo M, Martino R, et al Influence of the intensity of the conditioning regimen on the characteristics of acute and chronic graft-versus-host disease after allogeneic transplantation. Br J Haematol. 2005;130:394–403.[PubMed][Google Scholar]
  • 99. Perez-Simon JA, Kottaridis PD, Martino R, et al Nonmyeloablative transplantation with or without alemtuzumab: comparison between 2 prospective studies in patients with lymphoproliferative disorders. Blood. 2002;100:3121–3127.[PubMed][Google Scholar]
  • 100. Delgado J, Thomson K, Russell N, et al Results of alemtuzumab-based reduced-intensity allogeneic transplantation for chronic lymphocytic leukemia: a British Society of Blood and Marrow Transplantation Study. Blood. 2006;107:1724–1730.[PubMed][Google Scholar]
  • 101. Guinan EC, Boussiotis VA, Neuberg D, et al Transplantation of anergic histoincompatible bone marrow allografts. N Engl J Med. 1999;340:1704–1714.[PubMed][Google Scholar]
  • 102. Bacigalupo AAntilymphocyte/thymocyte globulin for graft versus host disease prophylaxis: efficacy and side effects. Bone Marrow Transplant. 2005;35:225–231.[PubMed][Google Scholar]
  • 103. Kroger N, Zabelina T, Kruger W, et al In vivo T cell depletion with pretransplant anti-thymocyte globulin reduces graft-versus-host disease without increasing relapse in good risk myeloid leukemia patients after stem cell transplantation from matched related donors. Bone Marrow Transplant. 2002;29:683–689.[PubMed][Google Scholar]
  • 104. Remberger M, Mattsson J, Ringden OPolyclonal anti-T-cell globulin as part of the preparative regimen for pediatric allogeneic stem-cell transplantation. Pediatric Transplantation. 2001;5:285–92.[PubMed][Google Scholar]
  • 105. Bacigalupo A, Lamparelli T, Bruzzi P, et al Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO) Blood. 2001;98:2942–2947.[PubMed][Google Scholar]
  • 106. Bacigalupo A, Lamparelli T, Barisione G, et al Thymoglobulin prevents chronic graft-versus-host disease, chronic lung dysfunction, and late transplant-related mortality: long-term follow-up of a randomized trial in patients undergoing unrelated donor transplantation. Biol Blood Marrow Transplant. 2006;12:560–5.[PubMed][Google Scholar]
  • 107. Nash RA, Antin JH, Karanes C, et al Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors. Blood. 2000;96:2062–8.[PubMed][Google Scholar]
  • 108. Qu L, Kiss JEThrombotic microangiopathy in transplantation and malignancy. Semin Thromb Hemost. 2005;31:691–699.[PubMed][Google Scholar]
  • 109. Wong R, Beguelin GZ, de Lima M, et al Tacrolimus-associated posterior reversible encephalopathy syndrome after allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2003;122:128–34.[PubMed][Google Scholar]
  • 110. Bolwell B, Sobecks R, Pohlman B, et al A prospective randomized trial comparing cyclosporine and short course methotrexate with cyclosporine and mycophenolate mofetil for GVHD prophylaxis in myeloablative allogeneic bone marrow transplantation. Bone Marrow Transplant. 2004;34:621–5.[PubMed][Google Scholar]
  • 111. Barker JN, Weisdorf DJ, DeFor TE, Blazar BR, Miller JS, Wagner JERapid and complete donor chimerism in adult recipients of unrelated donor umbilical cord blood transplantation after reduced-intensity conditioning. Blood. 2003;102:1915–9.[PubMed][Google Scholar]
  • 112. Baron F, Baker JE, Storb R, et al Kinetics of engraftment in patients with hematologic malignancies given allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. Blood. 2004;104:2254–62.[PubMed][Google Scholar]
  • 113. Burroughs L, Mielcarek M, Leisenring W, et al Extending postgrafting cyclosporine decreases the risk of severe graft-versus-host disease after nonmyeloablative hematopoietic cell transplantation. Transplantation. 2006;81:818–25.[PubMed][Google Scholar]
  • 114. Cutler C, Kim HT, Hochberg E, et al Sirolimus and tacrolimus without methotrexate as graft-versus-host disease prophylaxis after matched related donor peripheral blood stem cell transplantation. Biol Blood Marrow Transplant. 2004;10:328–36.[PubMed][Google Scholar]
  • 115. Cutler C, Henry NL, Magee C, et al Sirolimus and thrombotic microangiopathy after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2005;11:551–557.[PubMed][Google Scholar]
  • 116. Mielcarek M, Martin PJ, Leisenring W, et al Graft-versus-host disease after nonmyeloablative versus conventional hematopoietic stem cell transplantation. Blood. 2003;102:756–62.[PubMed][Google Scholar]
  • 117. Levine JE, Uberti JP, Ayash L, et al Lowered-intensity preparative regimen for allogeneic stem cell transplantation delays acute graft-versus-host disease but does not improve outcome for advanced hematologic malignancy. Biol Blood Marrow Transplant. 2003;9:189–97.[PubMed][Google Scholar]
  • 118. Diaconescu R, Flowers CR, Storer B, et al Morbidity and mortality with nonmyeloablative compared with myeloablative conditioning before hematopoietic cell transplantation from HLA-matched related donors. Blood. 2004;104:1550–8.[PubMed][Google Scholar]
  • 119. Martino R, Iacobelli S, Brand R, et al Retrospective comparison of reduced-intensity conditioning and conventional high-dose conditioning for allogeneic hematopoietic stem cell transplantation using HLA-identical sibling donors in myelodysplastic syndromes. Blood. 2006;108:836–46.[PubMed][Google Scholar]
  • 120. Maris M, Storb RThe transplantation of hematopoietic stem cells after nonmyeloablative conditioning: a cellular therapeutic approach to hematologic and genetic diseases. Immunol Res. 2003;28:13–24.[PubMed][Google Scholar]
  • 121. MacMillan ML, Weisdorf DJ, Davies SM, et al Early antithymocyte globulin therapy improves survival in patients with steroid-resistant acute graft-versus-host disease. Biol Blood Marrow Transplant. 2002;8:40–6.[PubMed][Google Scholar]
  • 122. MacMillan ML, Weisdorf DJ, Wagner JE, et al Response of 443 patients to steroids as primary therapy for acute graft-versus-host disease: comparison of grading systems. Biol Blood Marrow Transplant. 2002;8:387–94.[PubMed][Google Scholar]
  • 123. Martin PJ, Schoch G, Fisher L, et al A retrospective analysis of therapy for acute graft-versus-host disease: secondary treatment. Blood. 1991;77:1821–8.[PubMed][Google Scholar]
  • 124. Cragg L, Blazar BR, Defor T, et al A randomized trial comparing prednisone with antithymocyte globulin/prednisone as an initial systemic therapy for moderately severe acute graft-versus-host disease. Biol Blood Marrow Transplant. 2000;6:441–7.[PubMed][Google Scholar]
  • 125. McCaul KG, Nevill TJ, Barnett MJ, et al Treatment of steroid-resistant acute graft-versus-host disease with rabbit antithymocyte globulin. J Hematother Stem Cell Res. 2000;9:367–74.[PubMed][Google Scholar]
  • 126. Arai S, Margolis J, Zahurak M, Anders V, Vogelsang GBPoor outcome in steroid-refractory graft-versus-host disease with antithymocyte globulin treatment. Biol Blood Marrow Transplant. 2002;8:155–60.[PubMed][Google Scholar]
  • 127. Barr ML, Meiser BM, Eisen HJ, et al. Photopheresis for the prevention of rejection in cardiac transplantation. Photopheresis Transplantation Study Group. N Engl J Med. 1998;339:1744–1751.[PubMed]
  • 128. Gatza E, Rogers CE, Clouthier SG, et al Extracorporeal photopheresis reverses experimental graft-versus-host disease through regulatory T cells. Blood. 2008 prepublished online April 14. DOI:10.1182/blood-2007-11-125542. [Google Scholar]
  • 129. Greinix HT, Knobler RM, Worel N, et al The effect of intensified extracorporeal photochemotherapy on long-term survival in patients with severe acute graft-versus-host disease. Haematologica. 2006;91:405–8.[PubMed][Google Scholar]
  • 130. Reddy P, Ferrara JLImmunobiology of acute graft-versus-host disease. Blood Rev. 2003;17:187–94.[PubMed][Google Scholar]
  • 131. Levine JE, Paczesny S, Mineishi S, et al Etanercept plus methylprednisolone as initial therapy for acute graft-versus-host disease. Blood. 2008;111:2470–5.[Google Scholar]
  • 132. Koc S, Leisenring W, Flowers ME, et al Therapy for chronic graft-versus-host disease: a randomized trial comparing cyclosporine plus prednisone versus prednisone alone. Blood. 2002;100:48–51.[PubMed][Google Scholar]
  • 133. Couriel D, Hosing C, Saliba R, et al Extracorporeal photopheresis for acute and chronic graft-versus-host disease: does it work? Biol Blood Marrow Transplant. 2006;12:37–40.[PubMed][Google Scholar]
  • 134. Sano Y, Date H, Nagahiro I, Aoe M, Shimizu NLiving-Donor Lobar Lung Transplantation for Bronchiolitis Obliterans After Bone Marrow Transplantation. Annals Thoracic Surgery. 2005;79:1051–1052.[PubMed][Google Scholar]
  • 135. Rabitsch W, Deviatko E, Keil F, et al Successful lung transplantation for bronchiolitis obliterans after allogeneic marrow transplantation. Transplant. 2001;71:1341–1343.[PubMed][Google Scholar]
  • 136. Marr KA, Carter RA, Boeckh M, Martin P, Corey LInvasive aspergillosis in allogeneic stem cell transplant recipients: changes in epidemiology and risk factors. Blood. 2002;100:4358–4366.[PubMed][Google Scholar]
  • 137. Baddley J, Stroud T, Salzman D, Pappas PInvasive Mold Infections in Allogeneic Bone Marrow Transplant Recipients. Clin Infect Diseases. 2001;32:1319–1324.[PubMed][Google Scholar]
  • 138. Rovira M, Jimenez M, De La Bellacasa J, et al Detection of Aspergillus galactomannan by enzyme immunoabsorbent assay in recipients of allogeneic hematopoietic stem cell transplantation: a prospective study. Transplant. 2004;77:1260–4.[PubMed][Google Scholar]
  • 139. Guidelines for preventing opportunistic infections among hematopoietic stem cell transplant recipients. Biol Blood Marrow Transplant. 2000;6:659–713.[PubMed]
  • 140. Boeckh M, Kim HW, Flowers ME, Meyers JD, Bowden RALong-term acyclovir for prevention of varicella zoster virus disease after allogeneic hematopoietic cell transplantation--a randomized double-blind placebo-controlled study. Blood. 2006;107:1800–1805.[Google Scholar]
  • 141. Machado CM, Cardoso MRA, da Rocha IF, Boas LSV, Dulley FL, Pannuti CSThe benefit of influenza vaccination after bone marrow transplantation. Bone Marrow Transplant. 2005;36:897–900.[PubMed][Google Scholar]
  • 142. Machado CM, Boas LSV, Mendes AVA, et al Use of Oseltamivir to control influenza complications after bone marrow transplantation. Bone Marrow Transplant. 2004;34:111–114.[PubMed][Google Scholar]
  • 143. Darabi K, Abdel-Wahab O, Dzik WHCurrent usage of intravenous immune globulin and the rationale behind it: the Massachusetts General Hospital data and a review of the literature. Transfusion. 2006;46:741–53.[PubMed][Google Scholar]
  • 144. Kumar A, Teuber SS, Gershwin MEIntravenous immunoglobulin: striving for appropriate use. Int Arch Allergy Immunol. 2006;140:185–98.[PubMed][Google Scholar]
  • 145. Kulkarni S, Powles R, Treleaven J, et al Chronic graft versus host disease is associated with long-term risk for pneumococcal infections in recipients of bone marrow transplants. Blood. 2000;95:3683–6.[PubMed][Google Scholar]
  • 146. Meisel R, Kuypers L, Dirksen U, et al Pneumococcal conjugate vaccine provides early protective antibody responses in children after related and unrelated allogeneic hematopoietic stem cell transplantation. Blood. 2007;109:2322–2326.[PubMed][Google Scholar]
  • 147. Ljungman P, Engelhard D, de la Cámara R, et al Vaccination of stem cell transplant recipients: recommendations of the Infectious Diseases Working Party of the EBMT. Bone Marrow Transplantation. 2005;35:737–746.[PubMed][Google Scholar]
  • 148. Carpenter PA, Hoffmeister P, Chesnut CH, III, et al Bisphosphonate Therapy for Reduced Bone Mineral Density in Children with Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2007;13:683–690.[PubMed][Google Scholar]
  • 149. Rizzo JD, Wingard JR, Tichelli A, et al Recommended screening and preventive practices for long-term survivors after hematopoietic cell transplantation: joint recommendations of the European Group for Blood and Marrow Transplantation, Center for International Blood and Marrow Transplant Research, and the American Society for Blood and Marrow Transplantation (EBMT/CIBMTR/ASBMT) Bone Marrow Transplant. 2006;37:249–261.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.