Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Transplant. Dec 24, 2016; 6(4): 608-619
Published online Dec 24, 2016. doi: 10.5500/wjt.v6.i4.608
Biology of chronic graft-vs-host disease: Immune mechanisms and progress in biomarker discovery
Richard B Presland, Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA 98195, United States
Richard B Presland, Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98195, United States
Author contributions: The author solely contributes to the manuscript.
Conflict-of-interest statement: The author declares no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Richard B Presland, PhD, Department of Oral Health Sciences, School of Dentistry, University of Washington, 1959 NE Pacific St, Box 357475, Seattle, WA 98195, United States. rp@uw.edu
Telephone: +1-206-6166706
Received: April 16, 2016
Peer-review started: April 19, 2016
First decision: June 7, 2016
Revised: August 15, 2016
Accepted: September 13, 2016
Article in press: September 18, 2016
Published online: December 24, 2016

Abstract

Chronic graft-vs-host disease (cGVHD) is the leading cause of long-term morbidity and mortality following allogeneic hematopoietic stem cell transplantation. It presents as a chronic inflammatory and sclerotic autoimmune-like condition that most frequently affects the skin, oral mucosa, liver, eyes and gastrointestinal tract. Both clinical and animal studies have shown that multiple T cell subsets including Th1, Th2, Th17, T follicular helper cells and regulatory T-cells play some role in cGVHD development and progression; B cells also play an important role in the disease including the production of antibodies to HY and nuclear antigens that can cause serious tissue damage. An array of cytokines and chemokines produced by different types of immune cells also mediate tissue inflammation and damage of cGVHD target tissues such as the skin and oral cavity. Many of these same immune regulators have been studied as candidate cGVHD biomarkers. Recent studies suggest that some of these biomarkers may be useful for determining disease prognosis and planning long-term clinical follow-up of cGVHD patients.

Key Words: Chronic graft-vs-host disease, Biomarker, Allogeneic hematopoietic stem cell transplantation, Cytokine

Core tip: Chronic graft-vs-host disease (cGVHD) is a frequent long-term medical complication of allogeneic hematopoietic stem cell transplantation which can have a devastating impact on overall health and quality of life. This immune-mediated disorder manifests as an inflammatory and autoimmune-like disorder that can affect multiple tissues in an individual patient. Both clinical and animal studies demonstrate that multiple T cell subsets, as well as B cells, and their secreted cytokines play important roles in cGVHD initiation and progression. In the last decade many molecular biomarkers have been identified that correlate with cGVHD onset and/or progression, and some might have applications clinically in the near future.



INTRODUCTION

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is utilized primarily as a curative treatment for both hematological and non-hematological malignancies[1], although it has been used successfully in small-scale clinical trials as a stem cell therapy for some inherited diseases such as Recessive Dystrophic Epidermolysis Bullosa[2]. In the case of hematologic malignancies, the graft-vs-leukemia or graft-vs-tumor (GVL or GVT) effect mediated by donor-derived T cells helps to eliminate malignant cells in the transplant recipient[3]. However, a major long-term complication of allo-HSCT is chronic graft-vs-host disease (cGVHD), which occurs in 30%-70% of patients, with adults more frequently affected than pediatric patients[4]. Chronic GVHD manifests as an autoimmune-like inflammatory disease that can affect a single organ, but more typically it presents as a multi-organ disease affecting the skin (75% of patients), oral mucosa (51%-63% of patients), liver, eyes and gastrointestinal tract (22%-51% of patients)[4]. Oral mucosal disease can include salivary gland pathology or sclerosis of the lamina propria or submucosa. Other tissues including the lung, esophagus, joints, muscles and genitalia can also be involved (Table 1). cGVHD is often preceded by acute GVHD, which typically occurs within 100 d after transplantation, although the acute form can persist longer.

Table 1 Signs, symptoms and prevalence of chronic graft-vs-host disease in selected organs and tissues.
Organ or tissuePrevalence, %1Diagnostic features2Distinctive features3
Skin75%Poikiloderma Lichen planus-like features Sclerosis Morphea-like featuresDepigmentation Papulosquamous lesions
Mouth51%-63%Lichen planus-like featuresXerostomia Mucoceles Mucosal atrophy Ulcers Pseudomembranes
Liver29%-51%None4None4
Eye22%-33%Dry, gritty or painful eyes Cicatricial conjunctivitis Keratoconjunctivitis-Sicca syndrome
GI tract and esophagus7%-45%Esophageal web Strictures or stenosis in upper esophagus
Lung4%-19%Bronchiolitis obliteransAir trapping and bronchiectasis on chest CT scan
Muscles, fascia and joints6%Fascitis Sclerosis Joint stiffness or contracturesMyositis or polymyositis
Genitalia1%Lichen planus-like features Lichen sclerosus-like featuresErosions, Fissures, Ulcers

In allo-HSCT patients, cGVHD is the most common cause of non-relapse mortality (NRM, which refers to mortality not related to the primary malignancy or disease) among patients surviving more than two years[5]. Other important contributing factors to patient mortality are viral or bacterial infection and secondary malignancies (Figure 1)[4,6]. A recent analysis by the Center for International Blood and Marrow Transplant Research (CIBMTR) of more than 26000 allo-HSCT patients demonstrated that the incidence of cGVHD is increasing worldwide, making it imperative that we fully understand the etiology of this disease[7].

Figure 1
Figure 1 Causes of death among allogeneic hematopoietic stem cell transplantation patients. Pie charts show causes of death among patients who received a cell graft from (A) an HLA-matched sibling or (B) an unrelated donor. Data is from the Center for International Blood and Marrow Transplant Research, for allogeneic hematopoietic stem cell transplants performed in 2012-13[88] (Available from: URL: http//http://www.cibmtr.org/data/resources/pages/index.aspx). GVHD: Graft-vs-host disease.

This review will focus on the pathobiology of cGVHD, which has features of both alloimmune and autoimmune disease and involves altered activities and function of various T cell populations [T helper (Th) 1, Th2, Th17, T follicular helper cells and regulatory T-cells] as well as of B cells. Equally important are the various cytokines and chemokines produced by immune cells and their target tissues, which cause inflammation and tissue damage. A second productive area of cGVHD research is biomarker discovery; high-throughput approaches including mass spectrometry have led to the identification of a number of molecular markers from blood and saliva that correlate with active disease. Not surprisingly, many of these markers are associated with altered host immunity and/or tissue inflammation. This review will not discuss current primary and secondary therapeutic strategies for cGVHD; for an in-depth discussion of this topic, the reader is referred elsewhere[8-10].

CLINICAL FEATURES

According to the NIH consensus criteria published in 2005, cGVHD can be subclassified into: (1) Classic cGVHD presenting with manifestations that can be ascribed only to cGVHD; and (2) Overlap syndrome that has diagnostic or distinctive cGVHD manifestations together with features typical of acute GVHD[11]. Acute GVHD occurs in 40%-60% of patients receiving allo-HSCT and is one of the major risk factors for subsequent cGVHD. To improve cGVHD classification, the NIH severity score was developed which documents the number of organs involved and numerically scores the degree of functional impairment. Generally, patients are assessed as having mild, moderate or severe disease on a scale of 1 to 4 for each tissue[11-13]. However, clinical symptoms of cGVHD often overlap with other autoimmune diseases such as lichen planus and scleroderma and the degree of organ involvement is highly variable, which can make diagnosis challenging[8,14]. Table 1 lists signs and symptoms that are considered to be diagnostic of cGVHD as well as some of the commonly observed clinical features that are considered to be insufficient for disease diagnosis. As many as three or more tissues can be affected in a single patient, as reflected in the NIH global severity classification of cGVHD[13]. Skin manifestations that are considered diagnostic include poikiloderma (altered pigmentation with erythema), lichen planus-like lesions, sclerosis and morphea-like features (Table 1). Distinctive features (often observed in skin cGVHD but not sufficient for diagnosis) include depigmentation, papulosquamous lesions, ichthyosis and pruritis. Skin appendages are often targeted as well but these signs are not considered diagnostic: Symptoms can include scalp hair thinning or alopecia, sweat impairment and nail dystrophy or onycholysis (nail loss)[11,13].

Cutaneous cGVHD can occur in two forms termed lichenoid and sclerodermatous[15]. Lichenoid lesions usually occur early in the course of the disease, presenting as erythematous papules or plaques, with a squamous surface. Typical affected sites include the face, ears, palms and soles. Sclerodermatous cGVHD, which generally develops as a later complication, appear as sclerotic, shiny, white or yellow plaques with patchy hyperpigmentation or a poikilodermal appearance[15]. Sclerodermatous cGVHD can be localized or generalized and affect underlying tissues including the fascia, ligaments and peripheral nerves, causing pain and morbidity for the affected patient.

Oral symptoms vary but commonly involve lichenoid changes, xerostomia as a result of salivary gland damage, mucositis, erythema, mucoceles and restricted mouth opening (trismus) due primarily to sclerosis[16-18]. However, under current guidelines only lichen planus-like features are considered to be diagnostic (Table 1)[13]. Oral sensitivity and pain are often observed, which in more severe cases manifests as dysphagia (difficulty with swallowing) and weight loss. In one recent study of 210 cGVHD patients, 29% of cases were classified as malnourished by measurement using the Patient-Generated Subjective Global Assessment tool. Malnutrition was correlated with a lower body-mass index and poorer overall survival[19]. Gingivitis and tooth decay also occur because of xerostomia and altered oral immunity related to immunosuppression and reduced salivary IgG production[20].

Clinical symptoms seen in other involved tissues such as the liver, eyes, gastrointestinal tract, lungs, muscles/fascia and genitalia are summarized in Table 1, and have been reviewed extensively elsewhere[9,13]. Neurological manifestations of cGVHD are rare, but when present can include Myositis and Myasthenia gravis that affect the peripheral nervous system, and less commonly, various complications that affect the central nervous system[21]. Clinical features of cGVHD do not seem to vary with patient age, graft source (typically either bone marrow or PBSCs) and type of pre-transplant conditioning[4,9]. Most cases of cGVHD occur 4-6 mo after allo-HSCT, but 5%-10% of patients are diagnosed more than one year following allogeneic transplantation.

RISK FACTORS

The best documented risk factors for cGVHD are a history of acute GVHD (seen in 40%-60% of cGVHD patients), the use of PBSCs for grafting, a female donor-male recipient combination, older patient age and the use of HLA-mismatched or unrelated donors[13,22,23]. The increasing use of PBSCs (which contain more T cells compared to aspirated bone marrow) is one factor that influences the incidence and severity of cGVHD, since alloreactive T cells are a major player in cGVHD pathobiology[7]. These risk factors appear to largely explain the increasing incidence of cGVHD worldwide in allo-HSCT; however, additionally, a significant decline in early NRM appears to be contributing to the increased incidence of cGVHD in long-term survivors[7]. Notably, the frequency of GVHD-associated mortality is similar in HLA-matched sibling transplants compared to transplants performed using an unrelated donor (Figure 1).

A number of studies have also implicated certain genetic polymorphisms in addition to HLA antigen disparity between donor and recipient in the risk of GVHD risk (reviewed in Pidala et al[24]). For example, polymorphisms in a considerable number of genes that encode cytokines, chemokines or their receptors are associated with increased risk of cGVHD. These include genetic variants in the donor and/or recipient IL-10 genes[25-27], donor IL-1α gene[28], recipient IL-6 gene[29], recipient MHC class I-related chain A (MICA) gene (Val allele)[30], and donor and recipient IL-1 receptor antagonist (IL-1ra) genes[25,28]. For MICA, which acts as an activating ligand for the NKG2D receptor on certain types of T-cells, cGVHD incidence was positively correlated with serum MICA levels in patients post-HSCT; on the other hand, the presence of MICA antibodies prior to transplantation conferred protection against cGVHD[30]. A smaller number of genetic polymorphisms have been associated with decreased risk of cGVHD[24].

Baron et al[31] utilized gene expression profiling of donor CD4+ and CD8+ T cells to develop a “GVHD-predictive signature”, demonstrating the central importance of the TGF-β signaling pathway in regulating donor T cell function[31]. Remarkably, the so-called “dangerous donor” trait derived from T cell gene expression profiling not only predicted early (acute) GVHD, but also cGVHD occurrence in the recipient at one year post-transplantation. These observations reinforces other studies in humans and mice showing that the growth factor TGF-β has pleiotropic effects on T cells, including inhibition of Th1 cell differentiation and promoting expansion of regulatory T cells that are protective against cGVHD[9,32,33]. It also suggests that the grafted stem cells can have a long-term, dominant influence on the transplant recipient’s T cell profile and consequently the overall health of the patient.

PATHOBIOLOGY OF CGVHD: ROLE OF T CELLS, B CELLS AND THEIR CYTOKINES
T cells

While the mechanisms that cause the inflammation and tissue damage of acute GVHD are now quite well understood, the pathobiology of cGVHD is more complex and less well understood. Many investigators believe that the destructive immunological and autoimmune mechanisms that cause cGVHD are distinct from acute GVHD, irrespective of whether or not the cGVHD evolves from acute GVHD[6,34]. Activated donor T cells are the most important cell population in cGVHD, since T cell depletion from the graft prevents cGVHD in both human and animal studies[35]. The use of rabbit anti-thymocyte globulin (ATG) in conditioning regimens prior to transplant reduces the risk of subsequent acute and cGVHD, either by depleting donor T cells or by interfering with their activation by recipient alloantigens[22,36]. The major T cell subsets proposed to be involved in cGVHD include CD4+ T cells, CD4+ regulatory T cells (Tregs) and CD8+ T cells (Table 2).

Table 2 Immune cell types and their function in chronic graft-vs-host disease.
Cell typeSubtypesKey cytokines or markersBrief summary of disease involvement
CD4+ T cellsTh1IFN-γ, TNF-αPro-inflammatory. Important in acute GVHD, but role in cGVHD unclear
Th2IL-4, IL-13Stimulate antibody production. Role in clinical cGVHD poorly defined
Th17IL-17; also IL-21, IL-22, TNF-αPro-inflammatory. IL-17 levels correlate with disease severity; IL-17 induces scleroderma of skin and lung
TregsTGF-β, required for Treg proliferation and (differentiation)Produced mostly in thymus. Suppress autoreactive T cells. Lower levels of Tregs present in cGVHD patients, associated with thymic damage and loss of self-tolerance in cGVHD
T follicular helper cells1Express CCR5, PD-1 and ICOSPromote abnormal B cell maturation into long-lived active plasma cells, and IgG secretion
CD8+ T cellsCXCL9, CXCL10Mediate graft-vs-tumor effect of transplant. Serum CXCL9 levels elevated in cGVHD patients
B cells (total)Increased BAFF/B-cell ratio, elevated serum BAFF levelsDecreased in active cGVHD. Remaining B cells are resistant to apoptosis
Naïve and transitional B cellsCD19Decreased in active cGVHD
Memory B cells (total)CD19, CD27Decreased in active cGVHD. Cells essential for a normal immune response to bacterial pathogens or opportunistic infections
Regulatory B cellsIL-10Decreased in active cGVHD. Function to maintain tolerance and help prevent autoimmune disease
Plasma cellsCD27, CD38Increased in active cGVHD. Cells secrete immunoglobulins including IgGs and are resistant to apoptosis

Th1, Th2 and Th17 cells: Alloreactive CD4+ T cells that react to foreign (donor-derived) antigens include several Th cell subsets, primarily Th1, Th2 and Th17 cells. A central role for Th1 cells in acute GVHD is well established[6]; however, the importance of Th1 (and Th2) cells in cGVHD is still a matter of debate, even though Th1 cytokines such as interferon-γ (IFN-γ) can be found in skin and other tissues of affected patients[37]. Infusion of murine IFN-γ-null donor T cells reduced cGVHD symptoms in skin and salivary glands, indicating a role for Th1 cells in certain tissues[38]. A role for Th2 cells has been suggested because of the role of Th2 cytokines such as IL-4 and IL-13 (Table 2) in the production of antibodies to both self and non-self-antigens in patients; murine studies support the involvement of the Th2 cytokines IL-4 and IL-10 in stimulating B cell expansion in cGVHD[39].

Th17 cells produce several cytokines including IL-17, IL-21 and IL-22, which have potent pro-inflammatory functions in cGVHD[40]. IL-17A, produced mainly by CD8+ T cells, stimulates scleroderma which is an important feature of cutaneous cGVHD; however, current data suggests that co-expressed Th1 cytokines such as tumor necrosis factor-α (TNF-α) contribute to the observed pathology[41]. Improvement in cGVHD symptoms correlates with a reduction in Th-17 cell numbers in peripheral blood[42]. In liver cGVHD, there are increased numbers of Th17 cells and an increased Th17/Treg ratio observed in liver biopsies, suggesting that Th17 cells are an important driver of clinical liver disease[43].

T follicular helper cells: T follicular helper (TFH) cells promote differentiation of naïve B cells into memory B cells and class switching of IgG genes in the germinal center, within secondary lymphoid organs. A recent study showed that TFH cells were unusually active with prolonged survival in cGVHD patients, which correlates with the aberrant survival of B cells and hypersecretion of immunoglobulins[44]. The increased survival of Th2- and Th17-type TFH cells was correlated with increased cellular expression of the pro-survival marker Bcl-2. Overall, the study by Forcade et al[44] suggests that aberrant B cell activity including production of antibodies is driven, at least in part, by abnormal TFH cell activity. Studies using a murine model have confirmed the importance of TFH cells in cGVHD pathogenesis, particularly for the development of bronchiolitis obliterans syndrome which is a signature feature of lung cGVHD[45] (Table 1).

Tregs: Tregs, which are CD4+ CD25+ and also express the transcription factor FOXP3, suppress autoreactive T cells and are important for immune system homeostasis. Specifically, Tregs are essential for the establishment and maintenance of tolerance after allo-HSCT[46]. Tregs are depleted in both acute and cGVHD, demonstrating their importance as suppressors of inflammation and disease development[23]. Impaired Treg production and function has been linked to thymic damage as a result of CD4+ lymphopenia following allo-HSCT, at least in myeloablative patients[47]. The presence of Tregs in the skin and oral mucosa of cGVHD patients in a functional (e.g., CXCR3+) state suggests they may play a role in limiting tissue damage by alloreactive T cells[48]. Pharmacological approaches used to treat steroid-refractory cGVHD that increased Treg cell numbers have shown promise clinically in treating cutaneous cGVHD[49]. Further, in a study of allo-HSCT patients with acute leukemia, direct infusion of Tregs together with conventional T cells protected against GVHD in almost 90% of engrafted patients, while still maintaining the GVT anti-tumor effect conferred by conventional T cells[50]. These studies suggest that manipulation of Tregs might be a feasible approach to reducing or preventing GVHD without compromising the anti-tumor surveillance capacity of the patient’s immune system.

CD8+ T cells: CD8+ T cells are another immune cell population present in tissues affected by cGVHD, including the skin and oral mucosa[17]. Donor CD8+ cells mediate the GVT effect of allo-HSCT that typically results in the eradication of malignant cells from the patient. Among the cytokines produced by CD8+ cells are CXCL9 and CXCL10; CXCL9 is elevated in the serum of early-stage cGVHD patients, with CXCL9 levels being correlated with disease severity[51] (Tables 2 and 3).

Table 3 Candidate biomarkers of chronic graft-vs-host disease1.
Gene/proteinFunctionBiofluid3Ref.
BAFF, soluble; BAFF/B cell ratioGrowth factor, promotes B cell expansion and activationBlood[59,60,77]
CXCL92Chemokine produced by activated T cellsBlood[51,79]
CD-13, solubleAntigen presentationBlood[59]
C-reactive protein3Acute phase proteinBlood[12]
Cystatin BInhibitor of cathepsin proteasesSaliva[80]
IL-1raInhibitor of IL-1 receptor signalingSaliva[80]
IL-2R, solubleIL-2 receptor, marker of activated T cellsBlood[59,76]
IL-6Pro-inflammatory Th2 cytokineBlood[42,75]
IL-10Th2 cytokineBlood[73]
IL-15Enhances anti-tumor function of CD8+ T cellsBlood[78]
LactoperoxidaseAnti-microbial enzymeSaliva[81]
LactoferrinIron-binding glycoproteinSaliva[81]
MICA, solubleStimulates T cell activity via NKG2D receptorBlood[30]
TGF-βAnti-inflammatory cytokine; stimulates activity of TregsBlood[33]
TNF-αPro-inflammatory Th1 cytokineBlood[73-75]
B cells

In addition to T cells, there is increasing evidence that B cells play a number of important roles in cGVHD pathogenesis[52]. Patients with active cGVHD consistently have lower numbers of naïve and transitional B cells as well as total B cells[53,54] (Table 3). Regulatory B cells that secrete the anti-inflammatory cytokine IL-10 (and form a subpopulation within the transitional and memory B cell compartments) were also less frequent in cGVHD patients and displayed a deficiency in IL-10 production[55]. Together with Tregs, regulatory B cells play a central role in graft tolerance and the prevention of autoimmune disease and hence represent a topic worthy of further investigation in relation to cGVHD[56]. CGVHD patients are susceptible to pneumococcal infection which can cause severe or fatal infections in long term transplant survivors[57]. This susceptibility to infections is associated with the abnormal B cell profile, including decreased numbers of memory B cells that are critical for a normal immune response including IgG production[53,58]. Like many other autoimmune conditions, cGVHD patients frequently produce allo- and auto-antibodies to DNA and/or other antigens such as male HY antigen, which can correlate with disease onset and severity (see below). Activated B cells secrete an array of Th1 and Th2 cytokines that can regulate the function of T cell populations including Tregs. Levels of B cell activation factor (BAFF), a cytokine that promotes the survival and differentiation of activated B cells, are consistently increased relative to B cell numbers in patients with cGVHD[59,60]. As discussed above, the increased activity of TFH cells appears to play a significant role in producing the abnormal B cell profile characteristic of cGVHD[44].

Perhaps the best evidence that B cells are functionally important in human cGVHD are the numerous clinical observations with Rituximab, a humanized monoclonal antibody that targets the membrane protein CD20 of B cells, causing their cell death. Rituximab (and other anti-CD20 drugs) are effective in the treatment of steroid-refractory cGVHD, resulting in rapid and selective depletion of B cells and diminished activation of cytotoxic T cells; concurrently, the number and activity of Tregs are elevated[52,61,62]. In one study, the prophylactic use of Rituximab after allo-HSCT significantly reduced the incidence of both acute and cGVHD as well as NRM[63]. Hence, inhibiting B cell function has profound effects on both B and T cell homeostasis, with significant benefits to cGVHD patients especially in cases where other primary and/or secondary treatments have been unsatisfactory.

ANIMAL MODELS OF CGVHD

Several types of murine models have been utilized for studies of GVHD pathobiology including: (1) a bone marrow transplantation (BMT) model involving lethal radiation (total body irradiation, TBI) and transplantation of syngeneic marrow into treated mice[64]; (2) a parent-into-F1 model where donor spleen cells are infused into non-irradiated mice[39]; and (3) a transgenic model utilizing a self-antigen, membrane-associated chick ovalbumin, expressed under the control of the K14 promoter (K14-mOVA), where autoreactive skin disease is promoted by adoptive transfer of CD8 T cells from a second mouse strain, OT-1, that has an engineered T cell receptor specific for an ovalbumin peptide[65,66]. These animal models each demonstrate one or more manifestations of clinical cGVHD including the presence of anti-DNA antibodies, sclerosis, weight loss and chronic inflammation of skin and mucosal tissues associated with elevated Th1, Th2 and/or Th17 cytokines. The K14-mOVA adoptive transfer model has been used to test the efficacy of novel anti-inflammatory biologics that target the Janus kinase (JAK)[67] and Histone Deacetylase 6[68] enzymes, which were shown to be effective at suppressing and/or reversing cutaneous disease. Tofacitinib, the JAK inhibitor, blocked the expansion and activation of CD8+ cells thereby reducing IFN-γ secretion by CD8+ cells and keratinocytes as well as preventing the downstream consequences of interferon signaling such as chemokine production and keratinocyte apoptosis[67]. Another JAK inhibitor, Ruxolitinib (INCB018424), reduced murine GVHD (acute GVHD) symptoms and the levels of pro-inflammatory cytokines by both impairing differentiation of CD4+ T cells into IFN-γ and IL-17-producing cells, and by promoting the production of protective Tregs[69]. Notably this JAK 1/2 inhibitor reduced GVHD symptoms and improved overall animal survival while still maintaining the anti-tumor (GVT) effect[70].

While these pre-clinical models have been valuable in defining the immune mediators of cGVHD, the animal models do not typically parallel the evolution of the human disease, especially the common clinical presentation of classic cGVHD[8]. Additionally, in mice receiving intensive conditioning regimens (especially radiation), there is a well characterized scenario of inflammatory cytokine release, T cell activation and homing to target organs where tissue destruction occurs through the action of PBMCs and their associated cytokines. However, in humans the preparative regimen is only one factor involved in GVHD initiation, and its influence may be diminished in patients who now receive reduced-intensity conditioning prior to allo-HSCT[8]. Some recently described animal models exhibit systemic disease with multi-organ involvement including the lung, which appears to more closely resemble human cGVHD[71]. Despite some weaknesses, animal models will undoubtedly continue to provide insight into specific aspects of cGVHD pathobiology and will be essential for preclinical testing of new therapies for acute and cGVHD.

CGVHD BIOMARKERS

An emerging area of cGVHD research involves the discovery and validation of biomarkers that might eventually be used in clinical diagnosis or treatment planning. To date, most studies have focused on protein and immune cell biomarkers, even though RNAs (including mRNAs and micro RNAs) might also have utility as disease biomarkers[54,60]. As defined at the first meeting of the NIH Biomarker Working Group in 2006, cGVHD biomarkers could be used in disease management or clinical trials to: (1) predict response to therapy; (2) measure disease activity; (3) predict the risk of developing cGVHD; (4) diagnose cGVHD or predict prognosis; and (5) serve as a surrogate end point for therapeutic response[37].

To date, researchers have utilized mass spectrometry-based discovery approaches as well as Luminex and antibody arrays to screen clinical samples for potential serum and saliva protein biomarkers. Biomarkers identified to date can be broadly divided into proteins that function as cytokines and chemokines, immune (e.g., cytokine) receptors and other types of immune or non-immune proteins (Paczesny et al[72] for a recent review). Identified serum biomarkers that might indicate overall disease (and/or altered immune cell) activity include B cell activation factor (BAFF), MICA and anti-MICA antibodies, TNF-α, IL-15 and Chemokine (C-X-C motif) ligand 9 (CXCL9). Salivary biomarkers, associated mainly with oral cGVHD, include IL-1ra, cystatin B, lactotransferrin and lactoperoxidase (Table 3). Cellular markers primarily comprise immune cell populations that are altered in cGVHD (Table 2).

Serum biomarkers

Chronic cGVHD onset and/or persistence is associated with increased levels of TNF-α, BAFF, IL-6, sIL-2R (soluble IL-2 receptor alpha), and IL-10, and decreased levels of TGF-β and IL-15 (Table 3). Several studies have reported elevated levels of the pro-inflammatory cytokine TNF-α in acute and cGVHD, with measured levels correlating with cGVHD severity[73-75]. IL-6 shows a similar trend and correlation with cGVHD severity[42,75]. Soluble IL-2 receptor alpha (sIL-2Rα) is another example of a serum marker that is increased in pediatric and adult patients with cGVHD[59,76]. BAFF, a growth factor that promotes B cell differentiation and immunoglobulin production, is increased in both pediatric and adult patients with cGVHD; levels of this growth factor are often reported relative to the number of B cells in blood samples[51,59,60,77]. High levels of BAFF protein were present in allo-HSCT patients who subsequently developed cGVHD, confirming its role in alloimmunity[77]. CXCL9 levels are also elevated in newly diagnosed cGVHD patients and were correlated with disease severity in three different cohorts studied at two transplant centers[51].

Other markers besides BAFF and CXCL9 have been shown to have potential predictive value in allo-HSCT patients for determining future disease. For example, elevated levels of soluble MICA protein post-allo-HSCT were associated with an increased risk of cGVHD (by contrast, as stated above, the presence of MICA antibodies before transplantation conferred some protection from cGVHD)[30]. Similarly, Pratt et al[78] have shown that patients with low serum levels of IL-15 at day 7 post-transplant had 3-fold higher risk of developing cGVHD subsequently. IL-15 levels were observed to be inversely correlated with CD8 T cell levels, which are important for the GVT effect but also influence the development of cGVHD (see above).

In addition to intrinsic, host-dependent (e.g., immune) factors, the levels of biomarkers such as BAFF and CXCL9 can be modified by extrinsic factors including immunosuppressive drugs such as corticosteroids[51,54]. Hence, as recognized by many investigators, independent validation of promising biomarker candidates is essential. CXCL9 was recently validated as a cGVHD biomarker in a multicenter United States study of allo-HSCT patients[79].

Salivary biomarkers

Two recent studies utilizing mass spectrometry approaches identified a total of 82 and 102 salivary proteins, respectively, that showed altered expression in oral cGVHD[80,81]. IL-1 receptor antagonist (IL-1ra) exhibited reduced expression in patients with oral cGVHD[80]. The changes in IL-1ra expression coupled with higher levels of IL-1 family cytokines[16] in saliva likely enhance oral inflammation and subsequent tissue damage. In particular, IL-6 levels have been shown to correlate with oral cGVHD severity[16]. Changes in expression of salivary lactoperoxidase and lactotransferrin have also been reported, indicative of impaired innate immunity in oral cGVHD[81] (Table 3). The alterations in the salivary proteome among proteins involved in innate and acquired immunity are consistent with the clinical features of oral cGVHD, in particular patient susceptibility to bacterial and viral infections[4,18]. Changes in inorganic salivary components, especially Na+ and Cl- ions and inorganic phosphate, also occur in concert with cGVHD onset, correlating with hyposalivation and damage to the salivary glands[82,83].

Other biofluids

Certain Th2 and Th17 cytokines, in particular IL-6, IL-10, IL-17A and TNF-α are elevated in the tear fluid of cGVHD patients and correlated with systemic cGVHD regardless of ocular symptoms; levels of three of these cytokines (IL-6, IL-10 and TNF-α) also were significantly correlated with ocular cGVHD parameters[84].

Cellular biomarkers

In addition to protein biomarkers, a large number of immune cell populations have been studied as potential cGVHD biomarkers. Some of the best studied are listed in Table 2. As discussed in the Pathobiology section above, CD4+ IL-17+ Th17 cells are elevated in active cGVHD while Tregs that express the markers CD4, CD25 and FoxP3 are typically decreased[42,47]. There are also complex changes in the B cell population of cGVHD patients. Overall, total B cell counts are decreased in cGVHD patients as are the levels of naïve, transitional and regulatory B cells. In contrast, differentiated CD38+ CD27+ IgG-secreting plasma cells are increased in patients with active cGVHD (Table 2)[53,54].

Antibodies

Antibodies including autoantibodies are another group of well-studied potential biomarkers that are produced in cGVHD patients by an aberrant B cell population. Up to 80% of allogeneic transplants involving a female donor-male recipient combination produce antibodies against Y-chromosome-encoded HY proteins, and these antibodies appear to predict the development of cGVHD[8,37]. Antibodies to Platelet-derived Growth Factor (PDGF) Receptor, double stranded DNA and anti-nuclear antibody (ANA) are also common in cGVHD patients[59]. Anti-PDGF receptor antibodies cause accumulation of reactive oxygen species and stimulate type I collagen expression, suggesting a role for these antibodies in skin and lung fibrosis[85,86]. Patients with classic cGVHD were found to have higher levels of ANA and anti-DNA antibodies compared to patients who had a prior history of acute GVHD where B cells have limited involvement[59].

CONCLUSION

CGVHD is a chronic inflammatory and autoimmune-like condition that involves a complex interplay between the immune systems of the transplant donor and recipient. Despite significant progress in understanding the risk factors, and the development of effective second-line treatments for steroid-refractory cGVHD, the incidence of cGVHD is increasing worldwide[7]. While donor-derived T cells are still considered to be the preeminent mediators of cGVHD, aberrant B cells clearly play a significant role in promoting autoimmunity and inflammation, and conferring susceptibility to serious, often life-threatening infections. The enhanced activity of T follicular helper cells in cGVHD also appears to play a key role in the aberrant B cell activity and the resulting autoimmune-like features of cGVHD, including the presence of antibodies that target HY and nuclear proteins[44].

In addition to the significant progress in our understanding of cGVHD immunobiology and pathobiology, guidelines for biomarker development and validation were recently updated. The updated guidelines include recommendations for biomarker identification, verification, qualification, and application with terminology based on Food and Drug Administration and European Medicines Agency guidelines[72]. Suggested areas of focus for validation include biomarkers that are prognostic, stratify cGVHD risk or are predictive of future disease. Biobank repositories that can serially collect peripheral blood and cell samples from allo-HSCT patients in a standardized format will also be an important tool for pre-clinical biomarker validation[72]. The French National Cryostem Project is one example of such a national effort[87,88] which, together with multicenter collaborations[51,79], should enable protein biomarkers to be added to the clinician’s toolkit for cGVHD patient care in the not-too-distant future.

ACKNOWLEDGMENTS

I thank Dr. Thomas Morton for critically reading this manuscript. I also thank the anonymous reviewers for their many helpful comments and suggestions.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Transplantation

Country of origin: United States

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B, B, B, B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Budde H, Crocchiolo R, Greinix HT, Inamoto Y, Kelly DL, Teofili L, Zhang LL S- Editor: Qiu S L- Editor: A E- Editor: Lu YJ

References
1.  Copelan EA. Hematopoietic stem-cell transplantation. N Engl J Med. 2006;354:1813-1826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1657]  [Cited by in F6Publishing: 1630]  [Article Influence: 90.6]  [Reference Citation Analysis (0)]
2.  Tolar J, Wagner JE. Allogeneic blood and bone marrow cells for the treatment of severe epidermolysis bullosa: repair of the extracellular matrix. Lancet. 2013;382:1214-1223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
3.  Appelbaum FR. Haematopoietic cell transplantation as immunotherapy. Nature. 2001;411:385-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 328]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
4.  Lee SJ, Flowers ME. Recognizing and managing chronic graft-versus-host disease. Hematology Am Soc Hematol Educ Program. 2008;134-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 149]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
5.  Socié G, Stone JV, Wingard JR, Weisdorf D, Henslee-Downey PJ, Bredeson C, Cahn JY, Passweg JR, Rowlings PA, Schouten HC. Long-term survival and late deaths after allogeneic bone marrow transplantation. Late Effects Working Committee of the International Bone Marrow Transplant Registry. N Engl J Med. 1999;341:14-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 544]  [Cited by in F6Publishing: 509]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
6.  Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet. 2009;373:1550-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1733]  [Cited by in F6Publishing: 1720]  [Article Influence: 114.7]  [Reference Citation Analysis (0)]
7.  Arai S, Arora M, Wang T, Spellman SR, He W, Couriel DR, Urbano-Ispizua A, Cutler CS, Bacigalupo AA, Battiwalla M. Increasing incidence of chronic graft-versus-host disease in allogeneic transplantation: a report from the Center for International Blood and Marrow Transplant Research. Biol Blood Marrow Transplant. 2015;21:266-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 280]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
8.  Socié G, Ritz J. Current issues in chronic graft-versus-host disease. Blood. 2014;124:374-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
9.  Jacobsohn DA. Optimal management of chronic graft-versus-host disease in children. Br J Haematol. 2010;150:278-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
10.  Wolff D, Schleuning M, von Harsdorf S, Bacher U, Gerbitz A, Stadler M, Ayuk F, Kiani A, Schwerdtfeger R, Vogelsang GB. Consensus Conference on Clinical Practice in Chronic GVHD: Second-Line Treatment of Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2011;17:1-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 237]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
11.  Filipovich AH, Weisdorf D, Pavletic S, Socie G, Wingard JR, Lee SJ, Martin P, Chien J, Przepiorka D, Couriel D. 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]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2645]  [Cited by in F6Publishing: 2726]  [Article Influence: 151.4]  [Reference Citation Analysis (0)]
12.  Grkovic L, Baird K, Steinberg SM, Williams KM, Pulanic D, Cowen EW, Mitchell SA, Hakim FT, Martires KJ, Avila DN. Clinical laboratory markers of inflammation as determinants of chronic graft-versus-host disease activity and NIH global severity. Leukemia. 2012;26:633-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
13.  Jagasia MH, Greinix HT, Arora M, Williams KM, Wolff D, Cowen EW, Palmer J, Weisdorf D, Treister NS, Cheng GS. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21:389-401.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1768]  [Cited by in F6Publishing: 1681]  [Article Influence: 186.8]  [Reference Citation Analysis (0)]
14.  Lee SJ, Vogelsang G, Flowers ME. Chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2003;9:215-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 645]  [Cited by in F6Publishing: 646]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
15.  Aractingi S, Chosidow O. Cutaneous graft-versus-host disease. Arch Dermatol. 1998;134:602-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 115]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
16.  Fall-Dickson JM, Mitchell SA, Marden S, Ramsay ES, Guadagnini JP, Wu T, St John L, Pavletic SZ. Oral symptom intensity, health-related quality of life, and correlative salivary cytokines in adult survivors of hematopoietic stem cell transplantation with oral chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2010;16:948-956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
17.  Mays JW, Fassil H, Edwards DA, Pavletic SZ, Bassim CW. Oral chronic graft-versus-host disease: current pathogenesis, therapy, and research. Oral Dis. 2013;19:327-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
18.  Schubert MM, Sullivan KM, Morton TH, Izutsu KT, Peterson DE, Flournoy N, Truelove EL, Sale GE, Buckner CD, Storb R. Oral manifestations of chronic graft-v-host disease. Arch Intern Med. 1984;144:1591-1595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 95]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
19.  Bassim CW, Fassil H, Dobbin M, Steinberg SM, Baird K, Cole K, Joe G, Comis LE, Mitchell SA, Grkovic L. Malnutrition in patients with chronic GVHD. Bone Marrow Transplant. 2014;49:1300-1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
20.  Nagler RM, Nagler A. The molecular basis of salivary gland involvement in graft--vs.--host disease. J Dent Res. 2004;83:98-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
21.  Grauer O, Wolff D, Bertz H, Greinix H, Kühl JS, Lawitschka A, Lee SJ, Pavletic SZ, Holler E, Kleiter I. Neurological manifestations of chronic graft-versus-host disease after allogeneic haematopoietic stem cell transplantation: report from the Consensus Conference on Clinical Practice in chronic graft-versus-host disease. Brain. 2010;133:2852-2865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 155]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
22.  Flowers ME, Inamoto Y, Carpenter PA, Lee SJ, Kiem HP, Petersdorf EW, Pereira SE, Nash RA, Mielcarek M, Fero ML. Comparative analysis of risk factors for acute graft-versus-host disease and for chronic graft-versus-host disease according to National Institutes of Health consensus criteria. Blood. 2011;117:3214-3219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 447]  [Article Influence: 34.4]  [Reference Citation Analysis (0)]
23.  Subramaniam DS, Fowler DH, Pavletic SZ. Chronic graft-versus-host disease in the era of reduced-intensity conditioning. Leukemia. 2007;21:853-859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
24.  Pidala J, Sarwal M, Roedder S, Lee SJ. Biologic markers of chronic GVHD. Bone Marrow Transplant. 2014;49:324-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
25.  Rocha V, Franco RF, Porcher R, Bittencourt H, Silva WA, Latouche A, Devergie A, Esperou H, Ribaud P, Socie G. Host defense and inflammatory gene polymorphisms are associated with outcomes after HLA-identical sibling bone marrow transplantation. Blood. 2002;100:3908-3918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 160]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
26.  Kim DH, Lee NY, Sohn SK, Baek JH, Kim JG, Suh JS, Lee KB, Shin IH. IL-10 promoter gene polymorphism associated with the occurrence of chronic GVHD and its clinical course during systemic immunosuppressive treatment for chronic GVHD after allogeneic peripheral blood stem cell transplantation. Transplantation. 2005;79:1615-1622.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Mullighan C, Heatley S, Doherty K, Szabo F, Grigg A, Hughes T, Schwarer A, Szer J, Tait B, To B. Non-HLA immunogenetic polymorphisms and the risk of complications after allogeneic hemopoietic stem-cell transplantation. Transplantation. 2004;77:587-596.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Cullup H, Dickinson AM, Cavet J, Jackson GH, Middleton PG. Polymorphisms of interleukin-1alpha constitute independent risk factors for chronic graft-versus-host disease after allogeneic bone marrow transplantation. Br J Haematol. 2003;122:778-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
29.  Cavet J, Dickinson AM, Norden J, Taylor PR, Jackson GH, Middleton PG. Interferon-gamma and interleukin-6 gene polymorphisms associate with graft-versus-host disease in HLA-matched sibling bone marrow transplantation. Blood. 2001;98:1594-1600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 189]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
30.  Boukouaci W, Busson M, Peffault de Latour R, Rocha V, Suberbielle C, Bengoufa D, Dulphy N, Haas P, Scieux C, Amroun H. MICA-129 genotype, soluble MICA, and anti-MICA antibodies as biomarkers of chronic graft-versus-host disease. Blood. 2009;114:5216-5224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 76]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
31.  Baron C, Somogyi R, Greller LD, Rineau V, Wilkinson P, Cho CR, Cameron MJ, Kelvin DJ, Chagnon P, Roy DC. Prediction of graft-versus-host disease in humans by donor gene-expression profiling. PLoS Med. 2007;4:e23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
32.  Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S, Negrin RS. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9:1144-1150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1005]  [Cited by in F6Publishing: 987]  [Article Influence: 47.0]  [Reference Citation Analysis (0)]
33.  Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 2006;24:99-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1535]  [Cited by in F6Publishing: 1636]  [Article Influence: 90.9]  [Reference Citation Analysis (0)]
34.  Shlomchik WD. Graft-versus-host disease. Nat Rev Immunol. 2007;7:340-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 550]  [Cited by in F6Publishing: 554]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
35.  Ho VT, Soiffer RJ. The history and future of T-cell depletion as graft-versus-host disease prophylaxis for allogeneic hematopoietic stem cell transplantation. Blood. 2001;98:3192-3204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 309]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
36.  Finke J, Bethge WA, Schmoor C, Ottinger HD, Stelljes M, Zander AR, Volin L, Ruutu T, Heim DA, Schwerdtfeger R. Standard graft-versus-host disease prophylaxis with or without anti-T-cell globulin in haematopoietic cell transplantation from matched unrelated donors: a randomised, open-label, multicentre phase 3 trial. Lancet Oncol. 2009;10:855-864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 499]  [Cited by in F6Publishing: 524]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
37.  Schultz KR, Miklos DB, Fowler D, Cooke K, Shizuru J, Zorn E, Holler E, Ferrara J, Shulman H, Lee SJ. Toward biomarkers for chronic graft-versus-host disease: National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: III. Biomarker Working Group Report. Biol Blood Marrow Transplant. 2006;12:126-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 112]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
38.  Nishimori H, Maeda Y, Teshima T, Sugiyama H, Kobayashi K, Yamasuji Y, Kadohisa S, Uryu H, Takeuchi K, Tanaka T. Synthetic retinoid Am80 ameliorates chronic graft-versus-host disease by down-regulating Th1 and Th17. Blood. 2012;119:285-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
39.  Kataoka Y, Iwasaki T, Kuroiwa T, Seto Y, Iwata N, Hashimoto N, Ogata A, Hamano T, Kakishita E. The role of donor T cells for target organ injuries in acute and chronic graft-versus-host disease. Immunology. 2001;103:310-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
40.  Serody JS, Hill GR. The IL-17 differentiation pathway and its role in transplant outcome. Biol Blood Marrow Transplant. 2012;18:S56-S61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
41.  Hill GR, Olver SD, Kuns RD, Varelias A, Raffelt NC, Don AL, Markey KA, Wilson YA, Smyth MJ, Iwakura Y. Stem cell mobilization with G-CSF induces type 17 differentiation and promotes scleroderma. Blood. 2010;116:819-828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 124]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
42.  Dander E, Balduzzi A, Zappa G, Lucchini G, Perseghin P, Andrè V, Todisco E, Rahal D, Migliavacca M, Longoni D. Interleukin-17-producing T-helper cells as new potential player mediating graft-versus-host disease in patients undergoing allogeneic stem-cell transplantation. Transplantation. 2009;88:1261-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 89]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
43.  Malard F, Bossard C, Brissot E, Chevallier P, Guillaume T, Delaunay J, Mosnier JF, Moreau P, Grégoire M, Gaugler B. Increased Th17/Treg ratio in chronic liver GVHD. Bone Marrow Transplant. 2014;49:539-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
44.  Forcade E, Kim HT, Cutler C, Wang K, Alho AC, Nikiforow S, Ho VT, Koreth J, Armand P, Alyea EP. Circulating T follicular helper cells with increased function during chronic graft-versus-host disease. Blood. 2016;127:2489-2497.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Flynn R, Du J, Veenstra RG, Reichenbach DK, Panoskaltsis-Mortari A, Taylor PA, Freeman GJ, Serody JS, Murphy WJ, Munn DH. Increased T follicular helper cells and germinal center B cells are required for cGVHD and bronchiolitis obliterans. Blood. 2014;123:3988-3998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 155]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
46.  Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, Shimizu J, Takahashi T, Nomura T. Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol Rev. 2006;212:8-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1190]  [Cited by in F6Publishing: 1191]  [Article Influence: 66.2]  [Reference Citation Analysis (0)]
47.  Matsuoka K, Kim HT, McDonough S, Bascug G, Warshauer B, Koreth J, Cutler C, Ho VT, Alyea EP, Antin JH. Altered regulatory T cell homeostasis in patients with CD4+ lymphopenia following allogeneic hematopoietic stem cell transplantation. J Clin Invest. 2010;120:1479-1493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 176]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
48.  Imanguli MM, Cowen EW, Rose J, Dhamala S, Swaim W, Lafond S, Yagi B, Gress RE, Pavletic SZ, Hakim FT. Comparative analysis of FoxP3(+) regulatory T cells in the target tissues and blood in chronic graft versus host disease. Leukemia. 2014;28:2016-2027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
49.  Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, Armand P, Cutler C, Ho VT, Treister NS. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med. 2011;365:2055-2066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 851]  [Cited by in F6Publishing: 825]  [Article Influence: 63.5]  [Reference Citation Analysis (0)]
50.  Martelli MF, Di Ianni M, Ruggeri L, Falzetti F, Carotti A, Terenzi A, Pierini A, Massei MS, Amico L, Urbani E. HLA-haploidentical transplantation with regulatory and conventional T-cell adoptive immunotherapy prevents acute leukemia relapse. Blood. 2014;124:638-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 288]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
51.  Kitko CL, Levine JE, Storer BE, Chai X, Fox DA, Braun TM, Couriel DR, Martin PJ, Flowers ME, Hansen JA. Plasma CXCL9 elevations correlate with chronic GVHD diagnosis. Blood. 2014;123:786-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
52.  Shimabukuro-Vornhagen A, Hallek MJ, Storb RF, von Bergwelt-Baildon MS. The role of B cells in the pathogenesis of graft-versus-host disease. Blood. 2009;114:4919-4927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 205]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
53.  Kuzmina Z, Greinix HT, Knobler R, Worel N, Kouba M, Weigl R, Körmöczi U, Rottal A, Pohlreich D, Zielinski C. Proportions of immature CD19+CD21- B lymphocytes predict the response to extracorporeal photopheresis in patients with chronic graft-versus-host disease. Blood. 2009;114:744-746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
54.  Sarantopoulos S, Stevenson KE, Kim HT, Cutler CS, Bhuiya NS, Schowalter M, Ho VT, Alyea EP, Koreth J, Blazar BR. Altered B-cell homeostasis and excess BAFF in human chronic graft-versus-host disease. Blood. 2009;113:3865-3874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 253]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
55.  Khoder A, Sarvaria A, Alsuliman A, Chew C, Sekine T, Cooper N, Mielke S, de Lavallade H, Muftuoglu M, Fernandez Curbelo I. Regulatory B cells are enriched within the IgM memory and transitional subsets in healthy donors but are deficient in chronic GVHD. Blood. 2014;124:2034-2045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 151]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
56.  Durand J, Chiffoleau E. B cells with regulatory properties in transplantation tolerance. World J Transplant. 2015;5:196-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Kulkarni S, Powles R, Treleaven J, Riley U, Singhal S, Horton C, Sirohi B, Bhagwati N, Meller S, Saso R. Chronic graft versus host disease is associated with long-term risk for pneumococcal infections in recipients of bone marrow transplants. Blood. 2000;95:3683-3686.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  D’Orsogna LJ, Wright MP, Krueger RG, McKinnon EJ, Buffery SI, Witt CS, Staples N, Loh R, Cannell PK, Christiansen FT. Allogeneic hematopoietic stem cell transplantation recipients have defects of both switched and igm memory B cells. Biol Blood Marrow Transplant. 2009;15:795-803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 51]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
59.  Fujii H, Cuvelier G, She K, Aslanian S, Shimizu H, Kariminia A, Krailo M, Chen Z, McMaster R, Bergman A. Biomarkers in newly diagnosed pediatric-extensive chronic graft-versus-host disease: a report from the Children’s Oncology Group. Blood. 2008;111:3276-3285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 112]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
60.  Levine JE, Paczesny S, Sarantopoulos S. Clinical applications for biomarkers of acute and chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2012;18:S116-S124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
61.  Cutler C, Miklos D, Kim HT, Treister N, Woo SB, Bienfang D, Klickstein LB, Levin J, Miller K, Reynolds C. Rituximab for steroid-refractory chronic graft-versus-host disease. Blood. 2006;108:756-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 330]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
62.  Mohty M, Marchetti N, El-Cheikh J, Faucher C, Fürst S, Blaise D. Rituximab as salvage therapy for refractory chronic GVHD. Bone Marrow Transplant. 2008;41:909-911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
63.  Arai S, Sahaf B, Narasimhan B, Chen GL, Jones CD, Lowsky R, Shizuru JA, Johnston LJ, Laport GG, Weng WK. Prophylactic rituximab after allogeneic transplantation decreases B-cell alloimmunity with low chronic GVHD incidence. Blood. 2012;119:6145-6154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
64.  Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JL. Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood. 1997;90:3204-3213.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Miyagawa F, Gutermuth J, Zhang H, Katz SI. The use of mouse models to better understand mechanisms of autoimmunity and tolerance. J Autoimmun. 2010;35:192-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
66.  Miyagawa F, Tagaya Y, Kim BS, Patel HJ, Ishida K, Ohteki T, Waldmann TA, Katz SI. IL-15 serves as a costimulator in determining the activity of autoreactive CD8 T cells in an experimental mouse model of graft-versus-host-like disease. J Immunol. 2008;181:1109-1119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
67.  Okiyama N, Furumoto Y, Villarroel VA, Linton JT, Tsai WL, Gutermuth J, Ghoreschi K, Gadina M, O’Shea JJ, Katz SI. Reversal of CD8 T-cell-mediated mucocutaneous graft-versus-host-like disease by the JAK inhibitor tofacitinib. J Invest Dermatol. 2014;134:992-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
68.  Tsuji G, Okiyama N, Villarroel VA, Katz SI. Histone deacetylase 6 inhibition impairs effector CD8 T-cell functions during skin inflammation. J Allergy Clin Immunol. 2015;135:1228-1239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
69.  Spoerl S, Mathew NR, Bscheider M, Schmitt-Graeff A, Chen S, Mueller T, Verbeek M, Fischer J, Otten V, Schmickl M. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood. 2014;123:3832-3842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 299]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
70.  Choi J, Cooper ML, Alahmari B, Ritchey J, Collins L, Holt M, DiPersio JF. Pharmacologic blockade of JAK1/JAK2 reduces GvHD and preserves the graft-versus-leukemia effect. PLoS One. 2014;9:e109799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
71.  Srinivasan M, Flynn R, Price A, Ranger A, Browning JL, Taylor PA, Ritz J, Antin JH, Murphy WJ, Luznik L. Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans. Blood. 2012;119:1570-1580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 184]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
72.  Paczesny S, Hakim FT, Pidala J, Cooke KR, Lathrop J, Griffith LM, Hansen J, Jagasia M, Miklos D, Pavletic S. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: III. The 2014 Biomarker Working Group Report. Biol Blood Marrow Transplant. 2015;21:780-792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 104]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
73.  Skert C, Damiani D, Michelutti A, Patriarca F, Arpinati M, Filì C, Lucchi P, Malagola M, Bergonzi C, Roccaro A. Kinetics of Th1/Th2 cytokines and lymphocyte subsets to predict chronic GVHD after allo-SCT: results of a prospective study. Bone Marrow Transplant. 2009;44:729-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
74.  Ritchie D, Seconi J, Wood C, Walton J, Watt V. Prospective monitoring of tumor necrosis factor alpha and interferon gamma to predict the onset of acute and chronic graft-versus-host disease after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2005;11:706-712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
75.  Barak V, Levi-Schaffer F, Nisman B, Nagler A. Cytokine dysregulation in chronic graft versus host disease. Leuk Lymphoma. 1995;17:169-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 57]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
76.  Kobayashi S, Imamura M, Hashino S, Tanaka J, Asaka M. Clinical relevance of serum soluble interleukin-2 receptor levels in acute and chronic graft-versus-host disease. Leuk Lymphoma. 1997;28:159-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
77.  Sarantopoulos S, Stevenson KE, Kim HT, Bhuiya NS, Cutler CS, Soiffer RJ, Antin JH, Ritz J. High levels of B-cell activating factor in patients with active chronic graft-versus-host disease. Clin Cancer Res. 2007;13:6107-6114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 185]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
78.  Pratt LM, Liu Y, Ugarte-Torres A, Hoegh-Petersen M, Podgorny PJ, Lyon AW, Williamson TS, Khan FM, Chaudhry MA, Daly A. IL15 levels on day 7 after hematopoietic cell transplantation predict chronic GVHD. Bone Marrow Transplant. 2013;48:722-728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
79.  Yu J, Storer BE, Kushekhar K, Abu Zaid M, Zhang Q, Gafken PR, Ogata Y, Martin PJ, Flowers ME, Hansen JA. Biomarker Panel for Chronic Graft-Versus-Host Disease. J Clin Oncol. 2016;34:2583-2590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 97]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
80.  Devic I, Shi M, Schubert MM, Lloid M, Izutsu KT, Pan C, Missaghi M, Morton TH, Mancl LA, Zhang J. Proteomic analysis of saliva from patients with oral chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2014;20:1048-1055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
81.  Bassim CW, Ambatipudi KS, Mays JW, Edwards DA, Swatkoski S, Fassil H, Baird K, Gucek M, Melvin JE, Pavletic SZ. Quantitative salivary proteomic differences in oral chronic graft-versus-host disease. J Clin Immunol. 2012;32:1390-1399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
82.  Izutsu KT, Sullivan KM, Schubert MM, Truelove EL, Shulman HM, Sale GE, Morton TH, Rice JC, Witherspoon RP, Storb R. Disordered salivary immunoglobulin secretion and sodium transport in human chronic graft-versus-host disease. Transplantation. 1983;35:441-446.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Boer CC, Correa ME, Tenuta LM, Souza CA, Vigorito AC. Post-allogeneic Hematopoietic Stem Cell Transplantation (HSCT) changes in inorganic salivary components. Support Care Cancer. 2015;23:2561-2567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
84.  Jung JW, Han SJ, Song MK, Kim TI, Kim EK, Min YH, Cheong JW, Seo KY. Tear Cytokines as Biomarkers for Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2015;21:2079-2085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
85.  Svegliati S, Olivieri A, Campelli N, Luchetti M, Poloni A, Trappolini S, Moroncini G, Bacigalupo A, Leoni P, Avvedimento EV. Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease. Blood. 2007;110:237-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 180]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
86.  Baroni SS, Santillo M, Bevilacqua F, Luchetti M, Spadoni T, Mancini M, Fraticelli P, Sambo P, Funaro A, Kazlauskas A. Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis. N Engl J Med. 2006;354:2667-2676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 454]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
87.  The French National Cryostem Effort 2015. [updated 2015 Jan 20].  Available from: http//www.cibmtr.org/NewsEvents/CryoStem_call_for_research_project.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Pasquini MC, Zhu X. Current uses and outcomes of hematopoietic stem cell transplantation: CIBMTR Summary Slides, 2015.  Available from: http//www.cibmtr.org.  [PubMed]  [DOI]  [Cited in This Article: ]