Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Sep 14.
Published in final edited form as: Biol Blood Marrow Transplant. 2016 May 5;22(9):1552–1564. doi: 10.1016/j.bbmt.2016.04.022

The role of biomarkers in the diagnosis and risk stratification of Acute Graft vs. Host Disease (aGvHD): A systematic review

Alaa M Ali 1, John F DiPersio 1, Mark A Schroeder 1,*
PMCID: PMC5599102  NIHMSID: NIHMS899267  PMID: 27158050

Abstract

Allogeneic hematopoietic cell transplantation (HCT) is an increasingly used curative modality for hematological malignancies and other benign conditions. Attempts to reduce the morbidity/mortality and improve survival in patients undergoing HCT are crucial. The ability to diagnose acute graft versus host disease (aGvHD) in a timely manner, or even predict aGvHD prior to clinical manifestations, along with the accurate stratification of these patients are critical steps to improve the treatment and outcomes of these patients. Many novel biomarkers that may help in achieving these goals have been studied recently. This overview is intended to assist clinicians and investigators by providing a comprehensive review and analytical interpretation of the current knowledge concerning GvHD and biomarkers likely to prove useful in diagnosis and risk-stratification of this condition, along with the difficulties that hamper this approach.

Keywords: Graft-versus-Host Disease (GvHD), hematopoietic stem cell transplantation (HSCT), biomarkers, proteomics

Introduction

Acute graft-versus-host disease (aGvHD) is a major complication of allogeneic hematopoietic-cell transplantation (allo-HCT), occurring in up to 60% of the transplant recipients, with well described risk factors including human leukocyte antigen (HLA) mismatch, age, stem cell source, gender disparity, and conditioning regimen[1]. Acute GvHD is a systemic disorder driven by donor T-cells with a pleomorphic clinical presentation involving multiple target organs, including the skin, liver and gastrointestinal (GI) tract [2]. The diagnosis of aGvHD is clinical and pathologic confirmation is helpful but lacks positive and negative predictive value[35]. Immunosuppressive therapy with steroids is the first line therapy for clinically significant GvHD. Mortality and morbidity from high dose systemic immunosuppression is significant and no other treatment to date added to upfront steroids has proven beneficial[6]. Although the clinical diagnosis can be readily made basis in patients with a classical presentation, some cases prove challenging[7]. Currently, the diagnosis of aGvHD can be made by combining the clinical impression, high pre-test likelihood of aGvHD, exclusion of other competing disorders, along with histological examination of the target tissue. Biopsy alone is not the gold standard for diagnosis due to the false negative (patchiness of the disease, absence of the typical changes at early stages) and false positive results (residual conditioning regimen toxicity, infections)[3]–[5]. Furthermore, other causes for a patients’ symptoms may be found in many cases, confounding the diagnosis of aGvHD [8]–[12]. Once diagnosed, the severity of aGvHD has historically been graded using the Keystone Consensus criteria[13] or CIBMTR criteria[14]. Grading is an important step to determine the appropriate management of aGvHD and to determine prognosis and response to treatment. It has been well recognized that current grading systems overgrade some patients with a high likelihood of responding to immunosuppressive therapy and can not predict who will respond to steroids with certainty[15]. There are several limitations to current prognostic grading systems: despite the general relationship to outcomes, inter-observer errors can occur due to subjective biases, initial grade may not reflect peak grade and the time to response after therapy initiation is not accounted for. Therefore, many patients who are classified as standard-risk have their treatment fail while others classified as high- risk are over-treated. A recent update to the acute GvHD grading system may help to better stratify patients’ non-relapse mortality risk but could still be plagued by inter-oberserver biases[15] with significant variability across BMT centers[16]. The use of biomarkers can add to prediction accuracy and eliminate some of these problems.

High-dose systemic steroids are the standard first-line therapy for patients with grade II or higher aGvHD[6], [17]. However, practices differ among institutions with respect to initial steroid dose, the use of additional immunosuppressive agents, and the approach to steroid tapering after initial response. In general, therapy of clinically suspected aGvHD is started before diagnosis is confirmed and potentially before peak grade. Such an approach may expose the already immunosuppressed patients to unnecessary systemic steroids with significant infectious and non-infectious complications. In fact, a study conducted to reduce the dose of initial steroid therapy for grades I – II acute GvHD observed no difference in outcome between standard dose 2mg/kg and reduced dose 1mg/kg arms[18]. Approximately half of patients have complete responses to steroid by day 28 after therapy initiation[19]. Steroid refractory aGvHD is associated with high transplant-related mortality and low overall survival, even without relapse of the underlying disease that required their transplant[20]. There are no standard second line therapies for steroid-refractory aGvHD, and responses vary, but based on small retrospective and phase I/II studies responses are around 30 – 50% with six month NRM approximately 50%[6]. The hope of a biomarker based grading system has the potential to more accurately stratify patients based on risk of failing to respond to steroids, to alternative treatment approaches, or in those that fail upfront therapy, may be used to help choose additional lines of therapy.

Although mortality related to GvHD has been reduced in recent years [21], [22], aGvHD remains a major cause of transplant-related mortality (TRM). Acute GvHD represents the primary limitation to more widespread use of allogeneic HCT as a potentially curative modality for patients with malignant and non-malignant diseases. The field of biomarker research may provide for more accurate grading/risk stratification and identification of patients at higher risk for refractoriness to therapy or GvHD progression. Furthermore, the treatment of aGvHD has recently evolved from a one-size-fits-all approach to a more refined strategy based on predicted outcomes. Patients who are predicted to have low-risk aGvHD may benefit from lower doses and shorter courses of immune suppression. In addition, since not all cases of aGvHD progress in the same way or have the same outcome, the therapy should be tailored not only to the severity of the disease but also to the predicted rate of progression. As a result, many researchers have examined whether adding novel plasma biomarkers levels at different time points before and after transplantation can add to the prediction accuracy when compared to other prognostic tools. Timely recognition of patients at high risk for aGvHD or who would likely demonstrate resistance to steroids early in the course of their transplantation may lead to a more stringent monitoring, preventive care and early introduction of alternative and more effective immunosuppressive treatments earlier in the course of treatment.

It is reasonable to assume that plasma proteins involved in the complex pathophysiology of aGvHD might be altered in these patients. For the past 20 years, various groups have been investigating potential biomarkers to enhance the early and more accurate diagnosis and risk stratification of patients with aGvHD. Recent research has applied proteomics technologies to identify aGvHD biomarkers. This has led, in a short period of time, to the identification of novel biological pathways and biomarkers predictive of and associated with aGvHD [23]. Nevertheless, no single biomarker nor a panel of biomarkers, has been validated for clinical use via large multicenter trials. In this article, we summarize the current knowledge of promising diagnostic and prognostic aGvHD biomarkers and analyze the supporting data available in the literature.

Review Design

We searched PubMed and Medline up to December 31, 2015 to identify studies evaluating biomarkers in the setting of aGvHD. Each biomarker (microRNA, ST2, TNF receptor 1, IL-7, sBAFF, REG3α, S100, TIM-3, CK-18, HGF, Elafin) was searched separately as well. Only full-text articles published in English were considered. The primary search was conducted using the terms “Graft-versus-host disease” and “biomarker” excluding “Reviews”. Relevant references in the publications identified were also reviewed. Eligible studies included clinical studies with more than 5 patients. Studies investigating the diagnostic and prognostic value of transcriptomic and proteomic biomarkers were reviewed. Biomarkers that were evaluated in at least 2 independent studies will be discussed. The primary statistical outcomes were sensitivity/specificity, positive/negative predictive values, area under the curve, and hazard ratios. The main outcomes of the remaining preclinical and clinical studies were reported in the table but not discussed in the text.

Diagnostic and Prognostic Biomarkers

The biomarkers of aGvHD that have been discovered so far can be classified in a variety of ways: target (systemic or particular organ/tissue) or via their biophysical properties. Systemic biomarkers lack organ specificity for skin- or GI-type aGvHD. These biomarkers rise in response to systemic injury rather than specific tissue damage. On the other hand, organ-specific biomarkers are expressed by target organs rather than the effector cells that are damaging all tissues. Identifying markers that are target-tissue specific has been technically challenging due to the cellular heterogeneity of tissues and the difficulty of amplifying the amount of protein required. Another way of classifying these novel biomarkers is based on their biophysical properties. Transcriptomic biomarkers are discovered by RNA expression profiling (mRNA, rRNA, tRNA and other non-coding RNA), while proteomic biomarker are discovered by the methodical studying of the protein profile of a biologic specimen. Finally, cellular biomarkers are discovered by the studying of the altered numbers and functions of several different immune cell subsets [24]–[26].

Below we review the the following systemic biomarkers (miRNA, ST2, and markers of immune activation) and organ-specific biomarkers (Reg3α, S100, TIM-3, CK-18, HGF, and Elafin) and summarize these results in Table 1.

Table 1.

A summery of the biomarkers of aGVHD classified according to their organ specificity.

Biomarker Studies Type/N Time Assay Conditioning
regimen
GVHD
prophylaxis
Outcomes
Systemic MicroRNA (e.g. MiR-155, MiR-586) Ranganathan et al32 Murine/Human (5) D 21 post HCT RT-PCR N/A N/A MiR-155 is up-regulated in T-cell from aGVHD. Blocking MiR-155 may prevent aGVHD.
Xie et al33 Human (64) 2–5w post GVHD RT-PCR MAC CSA, MMF, MTX MiR-155 is up-regulated in aGVHD following allo-PBSCT with correlation with severity.
Wang et al34 Human (98) D: 7, 14, 21, 30, 60, 90 & at onset of GVHD RT-PCR MAC Not specified MiR-586>2200 copies/uL at D7 may predict impending aGVHD (Sen: 87.5%. Spec: 55.0%. AUC: 0.739)
Xiao et al35 Human (196) 16 days prior to GVHD diagnosis qRT-PCR MAC&RIC Not specified 4-miRNA panel predicted the probability of aGVHD (Sen: 92%. Spec: 62%. AUC: 0.8)
The panel was an independent predictor for developing aGVHD (HR: 1.478) at median of 16 d before diagnosis.
The MiRNAs levels were significantly associated with severity
The panel was an independent unfavorable prognostic factor for aGVHD OS (HR: 2.110)
AUC of miRNA signature was higher than the AUC for sIL2Ra 2 weeks after HCT (0.86 vs 0.76)

ST2 Vander Lugt et al43 Human (673)
Bi-center
D 14 after HCT
D 16, 28 after therapy of GVHD
Mass spectrometry/ELISA MAC&RIC CNI/MMF
CNI/MTX
CNI/MTX/Enbrel
High ST2 levels on D14 is associated with an increased NRM within 6 months of HCT (58% overall NRI)
Patients with high ST2 levels at initiation of therapy for aGVHD were 2.3 times as likely to have resistant aGVHD and 3.7 as likely to die within 6 months compared to patients with low levels.
Ponce et al44 Human (113) D28 after CBT ELISA MAC&RIC CNI/MMF High D28 ST2 levels (≥ 33.9 ng/mL) were associated with increased risk of III–IV aGVHD (HR: 2.62. AUC: 0.59) and increased TRM (HR:4.2. AUC:0.75)

Ceruloplasmin Lv et al95 Human (98) d-9, -1, 7, 14, 21, 28, 55–60,90–100 ELISA MAC CSA+MMF+MTX Ceruloplasmin levels> 670 μg/ml on D 7, 14, 21 predicted aGVHD occurrence (Sen: 79.7%, Spec: 78.3%, AUC: 0.861). The levels failed to predict resistance to first-line therapy.

Organ-specific Immune activation TNFR1 Choi et al47 Human (438) Before and at d7 after HCT ELISA MAC FK506+mini-MTX Day7 TNFR1 ratio correlated with eventual development of II–IV GVHD (HR=2.44) and TRM (HR=2.40)
Kitko et al48 Human (82)
Pediatrics
Before and at d7 after HCT ELISA MAC FK506+MMF
FK506+mini MTX
Day7 TNFR1 ratio correlated with severity and 1-year OS in pediatric population
Willems et al49 Human (106) Before and at d7 after HCT ELISA RIC CNI+MMF Day7 TNFR1 ratio correlated with grade II–IV (HR=2.2) and grade III–IV (HR=2.9) aGVHD but no impact on OS
August et al50 Human (62)
Peds & adults
Before, d0, 5, 10, 15 after HCT ELISA RIC&MAC Not specified D15 levels of TNFR1 (among other biomarkers: sCD8, sIL-2R, sCD40, sCD28) correlated with severe aGVHD (AUC=0.77, PPV=0.45, NPV=0.87)

IL-7 Dean et al53 Human (31) D7, 14. Month 1, 2, 3, 6, 9,12 ELISA RIC CSA+MTX IL-7 level at D+14 (cutoff value of 13 pg/mL) predicted the subsequent development of aGVHD (Sens: 85.7%, Spec: 88.2%, PPV: 85.7%, NPV: 88.2%). Higher levels were strongly associated with more severe grades of aGVHD.
Thiant et al55 Human (45) D0,7, 14, 18, 25, 30, 60, 90 ELISA RIC CSA+MTX IL-7 and IL-15 have similar kinetic, peaking on day +14 at four- to fivefold over pre-conditioning values (medians of 15.8 and 38.7 pg/ml). The occurrence grade II–IV aGVHD is associated with peak IL-7 level (HR=5.38)
Thiant et al54 Human (40) D0, 7,14,18,25,30,60,90 ELISA MAC CSA+MTX

sBAFF Cho et al57 Human (45) D0, 7, 14 ELISA MAC CSA+MTX
FK506+ MTX
sBAFF levels > 43 pg/mL at each time point had a significantly lower cumulative incidence of aGVHD (Sen: 75%, Spec: 73–82%, AUC: 0.7–0.8)

TIRC7 Zhu et al96 Human (39) Before and after GVHD therapy q-PCR &ELISA MAC&RIC CSA+MTX Higher levels of TIRC7 in aGVHD. Levels correlated with severity and declined markedly after therapy.

Lower GI-specific REG3a Harris et al62 Human (954)
Multi-center
At onset of GVHD ELISA High&moderate intensity CNI/MMF
CNI/MTX
REG3a (>151 ng/mL) distinguished LGI GVHD from non-GVHD diarrhea (AUC: 0.79, PPV: 95%, NPV: 34%).
REG3a at onset of LGI GVHD predicts nonresponse to therapy at D28 (PPV: 51%, NPV: 76%).
Higher REG3a concentrations at onset of LGI GVHD (above the median >135 ng/mL) correlated significantly with higher 1-year NRM (52% vs 33%)
Ferrara et al63 Human (1014)
Multi-center
At onset of LGI GVHD ELISA High&moderate intensity At least 2 agents including CNI REG3a levels distinguished LGI GVHD from non-GVHD diarrhea (AUC: 0.80, PPV: 95%, NPV: 32%).
REG3a levels (>151 ng/mL) at GVHD onset predicted non response to therapy at 4 weeks (3-fold higher), 1-year NRM (59% vs 34%), and 1-year OS (27% vs 48%)
REG3a levels further risk-stratified patients who had either advanced clinical stage or histologic severity (34% vs 66% for 1 or 2 risk factors).

S100 Reinhardt et al66 Human (52) At onset or progression of GVHD Calprotectin assay&sandwi ch-ELISA MAC&RIC CNI, MMF, MTX, ATG S100A8/S100A9 and S100A12 levels were significantly increased in stool and serum of patients with GI aGVHD and extensive cGVHD compared with patients without GVHD. No correlation with the grade could be detected.
Rodriguez-Otero et al67 Human (72) At onset of GVHD Sandwich-ELISA MAC&RIC CSA+ MMF
CSA+ MTX
Fecal S100 concentrations were increased in stage II–III GI aGVHD but not stage I causing low sensitivity of the marker 31% (90% specificity)
A high fecal S100 levels (≥ 100 μg/g) was strongly associated with SR-GVHD (CI of SR-GVHD: 93% vs 33%). S100 ≥ 100 μg/g independently correlated with a lower probability of CR (HR: 0.47)
Chiusolo et al68 Human (59) At onset of GVHD ELISA Not specified Not specified Fecal S100 was higher in patients with aGVHD than in non-GI-aGVHD. Levels increase with disease severity (with arbitrary cut-off of 160 mg/kg: Sen: 100%, Spec: 81.8%, PPV: 86%, NPV: 100%, AUC: 0.942)

Liver-specific TIM-3 Oikawa et al74 Murine N/A Flow cytometry N/A N/A TIM-3 is up-regulated in aGVHD. Anti-TIM-3 antibody increases the severity of aGVHD
Veenstra et al75 Murine N/A Same N/A N/A TIM-3/gal-9 pathway acts as a suppressor of aGVHD
Hansen et al76 Human (127) At onset of aGVHD Sandwich ELISA/Flow cytometry MAC&RIC CNI+MTX
CNI+MMF
TIM-3 levels were significantly higher in patients with more severe mid-gut GVHD, compared with those with upper-gut GVHD, without GVHD, and normal control (medians: 11,550 vs 4670 vs 2710 vs 2285 pg/mL respectively). The levels were higher in samples collected closest to GVHD onset compared with earlier samples. The levels correlated with severity.
Samples from patients with common diseases didn’t yield increased levels of TIM-3

CK-18 Luft et al81 Human (55) At the time of maximum GVHD ELISA MAC&RIC Not specified Both intestinal and hepatic GVHD were consistently associated with significant elevations of CK18F levels over baseline (5.6-fold change) CK18F levels decreased in responsive GVHD to immunosuppressive therapy and persisted in resistant GVHD
Conditions that might represent relevant differential diagnoses (toxic mucositis, noncomplicated, infection-related diarrhea, and VOD) were not associated with CK18F elevation.
Luft et al82 Human (48) Longitudinally: before HCT, at escalation of immunosuppression, late ELISA MAC&RIC CNI/MMF
CNI/MTX
Significantly higher CK18F levels were detected in steroid-refractory GVHD indicating the ongoing epithelial death of target organs.

HGF Okamoto et al97 Human (38) Serially from D0–120 post HCT ELISA Not specified Not specified HGF levels were increased in patients with than without aGVHD. The levels correlated with the grade of the disease.
Paczesny et al88 Human (424) D0–7–14–21–28–56–100 and at onset of GVHD ELISA MAC&RIC FK506+MTX
M CNI/MMF
HGF as part of a panel (along with IL-2Ra, TNFR1, IL-8) discriminated patients with and without aGVHD (AUC: 0.86).
The panel predicted survival independently of GVHD severity.
HGF was higher in visceral GVHD compared to skin-only GVHD.
Harris et al62 Human (954)
Multi-center
At onset of GVHD ELISA High&moderate intensity CNI/MTX
MF+MTX
HGF concentrations were elevated in LGI GVHD compared with isolated skin GVHD
HGF levels perform poorly as a diagnostic biomarker distinguishing LGI GVHD from non-GVHD diarrhea (AUC: 0.60) and liver GVHD from other causes of hyperbilirubinemia (AUC: 0.59)
High HGF concentrations correlated with significantly higher 1-year NRM

Skin-specific Elafin Paczesny91 Human (492) At onset of skin GVHD ELISA MAC&RIC CNI+ another agent Plasma elafin levels were twice as high in patients with skin GVHD compared to other groups (No GVHD, GI GVHD, Non-GVHD rash).
Elafin levels at the time of diagnosis of skin GVHD correlated with maximum overall GVHD grade.
Elafin is the best single discriminator for the diagnosis of GVHD in BMT patients with a rash compared to other biomarkers (AUC: 0.77)
The 1-year NRM was more than double in the high elafin group (≥ 6000 pg/ml) compared with low elafin group (28% vs 11%)
High elafin level at the time of GVHD diagnosis was significantly associated with a greater risk of death (hazard ratio of 1.78)
Chacon98 N/A N/A N/A N/A N/A There is immunohistochemical overexpression of elafin in both skin aGVHD and engraftment syndrome (ES). However, serum elafin hasn’t been reported to be elevated in ES

Abbreviations:

CSA: cyclosporine, MMF: myecophenolate mofetil, MTX: methotrexate, CNI: calcineurin inhibitors, FK506: tacrolimus.

MAC: myeloablative conditioning, RIC: reduced-intensity conditioning.

RT-PCR: reverse transcription polymerase chain reaction. ELISA: enzyme-linked immunosorbent assay.

Sen: sensitivity. Spec: specificity. PPV: positive predictive value, NPV: negative predictive value. HR: hazard ratio. NRI: net reclassification index. CI: cumulative incidence. AUC: area under the curve

CBT: cord blood transplantation.

VOD: veno-occlusive disease

SR-GVHD: steroid resistant GVHD. CR: complete response

Systemic biomarkers

MicroRNAs

MicroRNAs (miRNAs) are a class of small noncoding RNA (21- to 25-nucleotides) that negatively and positively regulate gene expression by translational repression or via induction in alterations in messenger RNA stability. miRNAs regulate gene function in a variety of ways and at multiple levels, particularly transcription, translation, and protein degradation[27]. Circulating miRNAs have been studied as novel, noninvasive biomarkers for many diseases such as cancer, sepsis, cardiovascular disease, liver injury, organ transplant rejection, and diabetes[28]–[31].

Researchers have conducted animal and human studies to investigate the role of miRNAs in the pathogenesis of aGvHD, and to screen for promising biomarkers and therapeutic targets [32]–[35]. Studies have found some lymphocyte-related serum miRNAs such as MiR-155 to be significantly up-regulated in aGvHD with a correlation between the level of serum MiR-155 and the severity of the disease[33]. However, MiR-155 is known to play a key role in inflammation regulation and immune response[36] and is unlikely to be specific for aGvHD. Furthermore, the expression of MiR-155 was the lowest among other miRNAs measured in the serum of patients with aGvHD[35]. Allo-HCT patients with high levels of another MiRNA, MiR-586, at day 7 were found to be at high risk of developing aGvHD[34]. However, levels of MiR-586 were easily affected by infections that are common at day 7 after allo-HCT. The lack of specificity of these studied MiRNAs has led to the simultaneous use of several MiRNAs to increase the specificity and diagnostic performance of these biomarkers.

Plasma miRNA signature has been studied as a biomarker for aGvHD. Xiao et al showed that evaluation of a panel of four microRNAs (miR-423, miR-199a-3p, miR-93, miR-377) was able to distinguish patients who developed aGvHD from those who did not (AUC: 0.80), and to predict the severity of the disease [35]. High expression of the miRNA panel was associated with poor overall survival. More importantly, elevated miRNAs can be detected at a median of 16 days before the diagnosis of aGvHD and were found to predict for the development of aGvHD when compared with another biomarker, sIL2Rα. Furthermore, this miRNA signature for aGvHD, unlike other studies of other MiRNAs or polypeptides, was not seen in the plasma of lung transplant recipients undergoing rejection, non-transplanted sepsis patients or patients with veno-occlusive disease[35]. This suggests a higher specificity of this miRNA signature for aGvHD. Serum or plasma miRNAs are highly stable in human peripheral blood[37] and are tissue-specific[38], making these biomarkers excellent targets for studying. There are other advantages of a miRNA signature including its ease of measurement, simplicity and lack of expense. Organ-specific association studies of miRNAs in the setting of aGvHD are lacking and could add to our improved stratification of patients to organ specific treatment strategies.

ST2

ST2 (suppression of tumorigenicity 2) is a member of the interleukin (IL)-1 receptor family and specifically binds to IL-33. ST2 is present in 2 isoforms: a membrane form expressed on hematopoietic cells, specifically T helper 2 (Th2) cells, and a soluble form secreted by endothelial and epithelial cells in response to inflammatory injury [39], [40]. Soluble ST2 appears to act as a receptor for IL-33 limiting its access to Th2 cells and promoting the Th1 phenotype, which has been associated in aGvHD pathophysiology[41], [42].

In a large two center study of 673 recipients of myeloablative or nonmyeloablative HCT, patients with high ST2 measured as early as 14 days after transplant had an increased risk of non-relapse mortality (NRM)[43]. Moreover, ST2 level predicted the response of aGvHD to treatment. Patients with high ST2 levels (defined as levels greater than 50% above the responder’s median value) were less likely to respond to treatment, while those with low levels were more likely to respond to therapy even if they had high-grade aGvHD using clinical criteria. The addition of ST2 to the clinical risk factors has reclassified almost 60% of patients receiving all types of conditioning regimens. Furthermore, a strong additive effect was noted when including the concentrations of both ST2 and REG3a (see below) to the clinical characteristics in order to improve the accuracy of the risk stratification. Similar results were obtained when testing the efficacy of ST2 in risk-stratifying Cord Blood Transplantation (CBT) patients[44].

Despite the promising results of these studies in demonstrating a particular significance of ST2 measurements in post-transplant mortality risk stratification, many questions remain. ST2 concentrations seem to vary significantly across conditioning intensities, necessitating separate thresholds for each conditioning intensity to be identified. Furthermore, the timing for measuring ST2 was arbitrarily chosen in these studies, and prospective evaluation of serial ST2 measurements is needed to refine the predictive value of this biomarker.

Biomarkers of Immune Activation

The persistent activation of the immune system that occurs early after donor graft infusion leads to excessive cytokine production (cytokine storm). Cytokines are important to the pathogenesis of aGvHD[45]. Many studies have examined cytokines and their receptors as potential aGvHD biomarkers. Herein we review studies that examined the diagnostic and prognostic utility of serum levels of TNF receptor 1, IL-7, and sBAFF.

TNF receptor 1

TNF-α is an inflammatory cytokine frequently implicated in the pathogenesis of aGvHD. It is the most frequently reported immune activation marker to be elevated prior to GvHD onset[46]. Researchers have measured TNF receptor 1 (TNFR1) as a surrogate marker for TNF-α. Increased levels of TNFR1 at day 7 equal or greater than 2.5 x baseline prior to transplant significantly correlated with the eventual development of severe aGvHD and with treatment related mortality in several studies[4749]. Interestingly, day 7 serum solubleTNFR1 levels were not significantly associated with grade II–IV aGvHD in these studies, suggesting that TNFR1 day7/baseline ratio has better predictive value. Another important observation of these studies is the low sensitivity of TNFR1 ratio (equal to or greater than 2.5 x baseline) in predicting severe aGvHD (sensitivity <40%) since the majority of patients who develop grades II–IV aGvHD have a TNFR1 day7/baseline ratio less than 2.5. Nevertheless, in patients with a TNFR1 ratio more than or equal to 2.5 x baseline, the likelihood of developing significant aGvHD is adequately high (~60%) to justify the future use of TNFR1 ratio-based preemptive treatment strategies. The screening performance of TNFR1 ratio may be strengthened by combining it to other biomarkers as a composite panel (See Table 2) (AUC=0.77 and NPV= 0.87)[50]

Table 2.

Examples of biomarker panels that combine several markers of aGVHD to increase the specificity or predictive/diagnostic power.

Panel biomarkers Study Type/N Time Assay Conditioning regimen GVHD Prophylaxis Outcome
TIM3, IL6, sTNFR1, ST2 McDonald et al77 Single center (317) D 7 through d 70 after HCT in cohort 1&2 (with GI GVHD)

D 14±3 after HCT in patients without GVHD
ELISA MAC CNI+MTX
CNI+MMF
The panel predicted development of grade 3–4 GVHD at median of 4 days before initiation of therapy (AUC: 0.88).
TIM3 predicted subsequent grade 3–4 GVHD (AUC: 0.76).
Plasma ST2 and sTNFR1 predicted 1-year NRM (AUC: 0.90).
IL-2Ra, TNFR1, HGF, IL-8, elafin, REG3a Levine et al92 Multicenter (112) Onset of GVHD D 0, 14, 28 after therapy initiation ELISA MAC/RIC Not specified D 0 and 14 biomarker panel independently predicted nonresponse at d 28 (OR: 2.98, OR: 6.32).
D 0 biomarker panel independently predicted mortality by day 180 (OR: 4.61, AUC: 0.7210).
D 28 biomarker panel predicted d 180 NRM (OR: 7.43).
Several soluble and cellular biomarkers Te Boome et al94 Single center (48) before and at predetermined time points after first MSC infusion ELISA MAC/RIC Not specified The panel was predictive for mortality (HR 2.924) when measured before MSC-administration.
ST2 was only predictive for mortality 2 weeks after but not before MSC-administration (HR 2.389).
TNFR1, ST2, REG3a Levine et al93 Multicenter (492) 48 h before or after therapy initiation ELISA MAC/RIC CNI+MTX
CNI+MMF
CNI+Sirolimus
Post-HCT
cyclophosphamide
The CI of 12-month NRM significantly increased as GVHD score increased (8% vs 27% vs 46% for score 1, 2, 3).
The response rates to primary GVHD therapy decreased as the GVHD score increased (86% vs 67% vs 46% for score 1, 2, 3).
The development of GI GVHD in patients who presented with skin GVHD only increased as the the score increased (19% vs 29% vs 34% for score 1, 2, 3).

First biomarker-based score that can be used to guide risk-adapted therapy.

Abbreviations:

MMF: myecophenolate mofetil, MTX: methotrexate, CNI: calcineurin inhibitors.

MAC: myeloablative conditioning, RIC: reduced-intensity conditioning.

RT-PCR: reverse transcription polymerase chain reaction. ELISA: enzyme-linked immunosorbent assay.

MSC: mesenchymal stromal cells

CI: cumulative incidence. HR: hazard ratio. AUC: area under the curve.

IL-7

Interleukin-7 (IL-7), is a growth factor that represents the principal homeostatic cytokine for T cells and is important for B and T-cell maturation. IL-7 promotes immune reconstitution after allogeneic HSCT and is needed for the development of acute GvHD in murine models[51], [52]. High levels of IL-7 along with other cytokines leads to homeostatic peripheral expansion of donor T cells within 30 days post-transplant. Given this key role in initial T-cell recovery, many groups have measured the levels of IL-7 over sequential time points before and after allo-HCT. In small studies, IL-7 levels at D+14 predicted the subsequent development of aGvHD (PPV: 85%, NPV: 88%) and higher levels were strongly associated with more severe grades of aGvHD[53]. By applying a multivariate model, high serum levels of IL-7 by day 14 after transplantation was the factor most strongly associated with the probability of developing grades II–IV aGvHD both in myeolablative[54] and reduced intensity conditioning[55]. It is important to note, however, that some other studies have reported less TRM in patients who recover T cells more rapidly[56] suggesting that the ratio among the subsets of the lymphocytes rather than the size determines the aGvHD developments and the outcomes. The predictive value of IL-7 levels still requires support from larger, prospective trials.

sBAFF

B cell activating factor, also known as B cell survival and activation factor, acts as a potent B-cell activator and it has been shown to play a crucial role in the proliferation and differentiation of these cells in mice and humans. It has been linked to a variety of autoimmune diseases. sBAFF is required for the reconstitution of B cells after myeloablation in animal models. Cho et al found that elevated sBAFF levels at any time point in the early days post-HCT were associated with decreased risk of developing aGvHD[57]. These findings suggest not only a predictive role of sBAFF during the peri-transplantation period, but also suggest that elevated levels may even confer protection against the development of aGvHD. This is to be contrasted with the notable recent reports that demonstrated an association between high sBAFF levels and the occurrence of chronic GvHD (cGvHD)[58]. This highlights the difference in pathogenesis between aGvHD, which is mediated mainly by donor T cells, and cGvHD where donor B cells play an important role in the pathophysiology of cGvHD. Despite the recent reports that have linked B cells to aGvHD[59], the role of B cells in the pathogenesis of aGvHD remains uncertain. Furthermore, T cells have been shown to express BAFF receptors[60]. Therefore, there could be a role for sBAFF in the regulation of T-cell dependent immunity. Further experimental and clinical studies are needed to gain more insight into the roles of BAFF and B cells in the pathogenesis of aGvHD

The use of sBAFF as a biomarker for aGVDH prediction may be complicated by a number of factors. First, BAFF levels are increased in the setting of B lymphopenia. Furthermore, the levels are also affected by medications, particularly high-dose steroids that are commonly used in aGvHD. Finally, current ELISA assays underestimate BAFF levels and precise quantification of the biomarker is lacking.

Organ-specific biomarkers

Lower Gastrointestinal Tract (LGI)- specific Biomarkers

LGI GvHD occurs in up to 60% of patients following allo-HCT. The involvement of the lower GI tract with aGvHD is often severe, and is characterized by voluminous secretory diarrhea, with or without hematochezia, and abdominal cramps. The etiology of diarrhea following allo-HCT is a common diagnostic dilemma often indistinguishable from other causes of diarrhea (conditioning chemotherapy related, administration of antibiotics, clostridium difficile-associated diarrhea, or cytomegalovirus infection). The gastrointestinal involvement of aGvHD remains a major cause of morbidity; and the use of biomarkers to predict the occurrence or estimate the maximal severity would be helpful. The most studied LGI-specific biomarkers are: REG3α, CK-18, S100, and TIM-3.

REG3a

REG3α (regenerating islet-derived 3-α) is an antimicrobial protein expressed in Paneth cells and secreted into the crypt microenvironment. REG3a may play a protective effect for intestinal stem cells (ISCs), which are important cellular targets of GvHD in the GI tract[61].

REG3α has been shown to have a very good diagnostic and prognostic performance in multicenter studies with large number of patients. REG3a distinguishes LGI GvHD from non-GvHD diarrhea with excellent positive predictive value and AUC (95% and 0.8 respectively). REG3α was the best diagnostic biomarker for LGI GvHD and additional biomarkers (HGF and CK18) as a composite panel provided minimal increased sensitivity or specificity. Furthermore, the levels of REG3a at onset of LGI GvHD predicts the response to first-line therapy and 1-year NRM[62], [63].

REG3α, along with clinical severity and histologic severity, provides important prognostic information before the initiation of therapy rather than at the time of maximum grade of GvHD. The three factors (REG3α levels, clinical and histologic severity) independently predict the lack of response to therapy and likely integrated into single grading system that will permit better risk stratification of patients with severe LGI aGvHD.

S100

S100 are pro-inflammatory proteins that play an important role in many inflammatory disorders, such as inflammatory bowel diseases (IBD) and rheumatoid arthritis RA[64], [65]. S100 proteins are released by activated or damaged phagocytes under conditions of cell stress during infections and autoimmune disease and they represent promising novel therapeutic targets. Studies have attempted to evaluate the diagnostic and prognostic value of serum and fecal levels of some of S100 proteins (S100A12 and calprotectin: S100A8/S100A9) in the setting of LGI aGvHD[66]–[68]

The major advantage of these markers was not the capability to diagnose GI-GvHD but rather their ability to predict the response to steroids, and therefore risk-stratifying these patients. The sensitivity of fecal S100 proteins for the diagnosis of stage 1 GI aGvHD was quite low (with no sensitivity improvement with cutoff decrease) and was unable to discriminate aGvHD from other causes of diarrhea. However, the S100 levels were predictive for responses to treatment. Furthermore, fecal S100 (along with other fecal markers: α1-antitrypsin and elastase) were particularly useful re-stratifying patients with stage 2 GI GvHD, allowing patients with the worst prognosis to be identified. Interestingly, the probability of steroid-resistant GI aGvHD was as high as 100% if two fecal markers were high and as low as 0% if the 2 markers were low.

Fecal proteins represent attractive noninvasive biomarkers to evaluate LGI aGvHD. The biomarkers are immediately available and can be easily collected. Moreover, if validated they they may represent a better tool to grade, risk-stratify and monitor the response to treatment in patients with LGI aGvHD compared to histologically variable GI biopsies and measurement of stool volumes.

TIM-3

The TIM (T-cell immunoglobulin domain and mucin domain) family of genes was first described in 2001[69]. The critical role of these genes products (3 proteins: TIM-1, -3, and -4) in regulating immunity is beginning to emerge[70]. TIM-3 was the first described and has been the most studied member of the family[71]. To date, the in vivo functions of TIM-3 and its role in immunoregulation remained largely unknown with discrepancy between studies. Interactions between TIM-3 and its ligand galectin-9 (gal-9) play a role in autoimmune disorders, chronic infection, tumor immunity, and transplantation[72], [73]. In murine models of aGvHD, TIM-3 has been shown to be up-regulated with a possible inhibitory role to the interaction between TIM-3 and its ligand in the pathogenesis of aGvHD[74], [75]

The predictive value of serum TIM-3 levels in the setting of GvHD has been studied by Hansen et al[76]. Although TIM-3 levels correlated with the occurrence and severity of the disease, the area under the receiver operating characteristic (ROC) curve was significant only for severe mid-gut GvHD compared to all other aGvHDs including upper GI aGVHD (0.79 vs 0.59). These findings are inconsistent with the recent paper from McDonald et al[77], where the measurement of TIM-3 was the most useful analyte among others for predicting grade 3–4 vs grade 0–2 aGvHD (AUC: 0.76) (See Table 2).

CK-18

Cytokeratin-18 (CK18) is an intermediate filament that is typically expressed in the epithelia of the digestive, respiratory and urogenital tracts[78]. The initiation of the apoptotic process results in activation of certain caspases which cleave a variety of cellular substrate including CK18. Cleavage at a particular site (DALD-S) gives rise to a neo-epitope termed cytokeratin-18 fragments (CK18Fs) which are released into the serum [79]. Intra-cryptal apoptosis is the histopathologic hallmark in GvHD and is used in the grading of GvHD[80]. Based on the hypothesis that apoptosis at the level of target organs in aGvHD leads to increase in CK18F and that CK18F mirrors the pathogenetic end point of GvHD, researchers have investigated whether CK18F provides a tool for sensitive assessment of GvHD-associated apoptotic activity and grading of the disease[81], [82]. Although CK18F levels correlated with GI and hepatic GvHD as well as response to immunosuppressive therapy, these studies were not designed to systematically determine the diagnostic features of this biomarker (sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV)). Apoptosis is not GvHD-specific and more studies are needed to validate the diagnostic performance of its associated biomarkers in the setting of aGvHD.

Liver-specific Biomarkers

HGF

Hepatocyte growth factor (HGF) is a cellular growth factor that targets epithelial and endothelial cells including hepatocytes, by activating a tyrosine kinase signaling cascade after binding to proto-oncogene c-Met receptors[83], [84]. Serum HGF concentrations have been reported to be elevated in liver disease, chronic renal failure, systemic inflammatory response syndrome (SIRS), and malignant diseases such as leukemia, breast cancer, and gastric cancer[85]–[87].

Although the levels of HGF were found to be elevated in patients with visceral GvHD, HGF performs poorly as a diagnostic biomarker. HGF failed to distinguish LGI aGvHD from non-GvHD diarrhea (AUC: 0.6)[62]. Likewise, HGF concentrations are elevated in liver GvHD compared with asymptomatic patients but are comparable to concentrations in patients with non-GvHD hyperbilirubinemia (AUC:0.59)[62]. A small number of patients develop hyperbilirubinemia caused by non-GvHD transplantation-related complications such as veno-occlusive disease (VOD) and these complications can be discriminated clinically from GvHD. In spite of this, a biomarker(s) that differentiates liver aGvHD from these complications would be clinically meaningful. HGF seems to perform significantly better as a diagnostic biomarker when integrated in a composite biomarker panel[88]. (See Table 1)

In spite of the poor diagnostic capabilities of HGF, this biomarker has a significantly better prognostic performance. The biomarker panels that involved HGF were shown to predict 1-year NRM and long-term survival independently of GvHD severity, and HGF was the primary contributor to the prognostic significance of these panels[62], [88]

Skin-specific Biomarkers

Elafin

Skin-derived antileukoproteinase (SKALP), otherwise known as Elafin, is an elastase-specific protease inhibitor expressed mainly in epithelial cells. Elafin is not present in normal keratinocytes but overexpressed in a variety of inflammatory skin disorders such as psoriasis[89]. In vivo studies have shown that inflammatory cytokines that are secreted in patients with aGvHD can stimulate the expression of elafin in epidermal cells [90]. Immunohistochemistry studies revealed an overexpression of elafin in GvHD skin biopsies. Elafin is expressed by the target epidermal cells rather than the effector cells that injure all three-target organs (GI, Liver, skin), which explains the higher specificity of elafin as a biomarker for skin aGvHD. Although produced locally as an antiprotease secreted in response to cytokines, elafin is readily detected in the systemic circulation as well.

When the diagnostic features of serum elafin as a biomarker of skin aGvHD were compared to other biomarkers of aGvHD (TNFR1, IL2Rα, IL8, HGF) (See Table 1), elafin was found to be the best single discriminator for the diagnosis of skin GvHD from other etiologies of rash (drug rash, engraftment syndrome, leukemia cutis, and viral or fungal rash). The area under the curve (AUC) of elafin was the highest (0.77) compared to the above biomarkers. Furthermore, elafin correlated with the maximum overall GvHD grade and 1-year NRM, making its prognostic value appealing as well[91].

Biomarker Panel

When several clinically-useful biomarkers are present, typically no particular one is satisfactory alone in terms of sensitivity or specificity for the diagnosis or prediction of a specific disease. Researchers have identified potential biomarker panels for aGVHD either by combining individual biomarkers that have been studied previously or by performing discovery studies that compare samples from patients with severe aGVHD with samples from patients without aGVHD. By analyzing these samples, biomarkers that provide the best discrimination between the two groups are identified. Afterwards, the panel will be developed, validated and tested for operating characteristics. Summary of biomarker panels is presented in Table 2.

Recently, many groups have used biomarker panels to identify patients at the extremes of outcomes. These panels have been shown to provide prognostic and predictive information of aGvHD outcomes, including maximal severity, response to therapy, and mortality[77], [92]. The predictive values of these panels were independent and additive of the clinical status of the patient (onset grade, unrelated vs related donor and peripheral vs bone marrow vs cord as a source of stem cells) with very good area under the curve(0.7–0.9) (Table 2). However, the positive predictive values of the panels remained in the range of 40–50%, indicating that the false positives would still equal or outnumber the true positives. Recent reiteration of the biomarker panel by the University of Michigan group (Ann Arbor GvHD score) examined the utility of measuring 3 biomarkers (TNFR1, REG3α, and ST2) at the time of aGvHD diagnosis to create a model algorithm that predicts NRM 6 months later in a well-powered study[93]. The algorithm defined three distinct scores whose risk of NRM increased with each increasing grade (independently of other risk factors, such as donor type, age, conditioning regimen, and HLA match) and may prove useful in the guidance of aGvHD therapy. The value of biomarkers in predicting the risk of complications and mortality following HCT will be studied in BMT-CTN Protocol 1202.

Although the integration of a biomarker panel in clinical practice still needs further investigation, these panels may also provide a valuable alternative or addition to composite end points in clinical trials evaluating novel therapies for aGvHD and improve the success rate of these trials. Biomarker panels may provide an opportunity for a better selection of patients who are likely to benefit from expensive or potentially risky therapies tested in clinical trials as well as a close monitoring of response. To this end, Te Boome et al evaluated the utility of biomarkers in the prediction of either resolution of aGvHD or survival in patients with steroid-refractory aGvHD treated with mesenchymal stromal cells (MSCs) in a prospective phase II trial[94]. Surprisingly, biomarkers that were previously reported to be predictive of outcomes (ST2[43] and Levine panel[92]) failed to correlate with clinical severity or predict resolution of aGvHD, likely due to the relatively low number of patients.

These studies represent the first demonstrations of how GVHD biomarkers panels and biomarker-based scores may be potentially incorporated into clinical care.

Summary

Many flaws exist in the current clinically-based grading systems of aGvHD. Maximal clinical severity, which grading tools rely on, can only be assigned retrospectively after the response to treatment is known. Withholding treatment for GvHD or waiting for 14–28 days to evaluate for refractoriness to therapy may lead to extensive and irreversible damage, especially in the GI tract, resulting in unacceptable high rates of morbidity and mortality. If it were possible to predict the ultimate severity of aGvHD before or at the onset of symptoms, preemptive therapy (e.g. anti-T cell therapy) could be given to blunt the intensity of tissue damage. Whether this approach, even when implemented early will work to change the natural history of GvHD remains to be seen and proven in prospective clinical trials. Even patients who respond to low doses of steroids can experience unnecessary infectious complications as well as other morbidities such as diabetes mellitus and avascular necrosis.

Previous studies have clearly established correlation between individual as well as combined biomarkers and outcomes of aGvHD. However, inconsistent results among different clinical centers and studies are noted and mentioned in this review. This inconsistency can be attributed to the high heterogeneity of patient groups receiving various treatments as well as the variation in study protocols and treatments. Despite the statistically significant operating characteristics of many of the biomarkers discussed and the composite panels, the use of these data in clinical practice should currently be reserved for use in well designed prospective clinical trials. Although the data derived from biomarker-based scoring systems predicts outcomes better than clinical scoring systems, the positive predictive value of these biomarkers is still low enough and is most useful for patients who score at either end to justify preemptive therapy. Moreover, reconciling the results of the various studies done at different centers to examine the value of different biomarkers in the prediction of aGvHD outcomes will be difficult due to the differences in timing, intensity of conditioning therapy, choice of GvHD prophylaxis, source of hematopoietic cells, and statistical methods. This suggests that predictive biomarkers might need a coordinated multicenter approach with coordinated times of sampling and centralized analysis of biomarker levels. Another attractive approach would be to incorporate the clinical characteristics and risk factors with the biomarker scores (such as the Ann Arbor score) to improve the predictive power of these scores and their treatment algorithms. Biomarkers represent a compelling yet still unproven tool for reproducible prediction of aGvHD, the severity of aGvHD and the outcome of therapy.

Highlights.

  • Biomarkers predict the severity and outcomes of aGVHD better than clinical grading systems.

  • Integrating clinical characteristics into biomarker scores may improve the predictive power of both.

  • Multicenter studies are needed to validate these biomarkers in different settings.

Acknowledgments

Conflict of interest statement: The authors declare that there are no conflicts of interest.

Financial disclosure: The authors have nothing to disclose.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Lee S-E, Cho B-S, Kim J-H, Yoon J-H, Shin S-H, Yahng S-A, Eom K-S, Kim Y-J, Kim H-J, Lee S, Min C-K, Cho S-G, Kim D-W, Lee J-W, Min W-S, Park iC-W. Risk and prognostic factors for acute GVHD based on NIH consensus criteria. Bone Marrow Transplant. 2013 Kwi;48(4):587–92. doi: 10.1038/bmt.2012.187. [DOI] [PubMed] [Google Scholar]
  • 2.Ferrara JLM, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet (London, England) 2009 Maj;373(9674):1550–61. doi: 10.1016/S0140-6736(09)60237-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Iqbal N, Salzman D, Lazenby AJ, Wilcox CM. Diagnosis of gastrointestinal graft-versus-host disease. Am J Gastroenterol. 2000 Lis;95(11):3034–8. doi: 10.1111/j.1572-0241.2000.03250.x. [DOI] [PubMed] [Google Scholar]
  • 4.Roy J, Snover D, Weisdorf S, Mulvahill A, Filipovich A, Weisdorf D. Simultaneous upper and lower endoscopic biopsy in the diagnosis of intestinal graft-versus-host disease. Transplantation. 1991 Mar;51(3):642–6. doi: 10.1097/00007890-199103000-00019. [DOI] [PubMed] [Google Scholar]
  • 5.McDonald GB. How I treat acute graft-versus-host disease of the gastrointestinal tract and the liver. Blood. 2016 Sty; doi: 10.1182/blood-2015-10-612747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Martin PJ, Rizzo JD, Wingard JR, Ballen K, Curtin PT, Cutler C, Litzow MR, Nieto Y, Savani BN, Schriber JR, Shaughnessy PJ, Wall DA, Carpenter PA. First- and second-line systemic treatment of acute graft-versus-host disease: recommendations of the American Society of Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2012 Sie;18(8):1150–63. doi: 10.1016/j.bbmt.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dignan FL, Clark A, Amrolia P, Cornish J, Jackson G, Mahendra P, Scarisbrick JJ, Taylor PC, Hadzic N, Shaw BE, Potter MN. Diagnosis and management of acute graft-versus-host disease. Br J Haematol. 2012 Lip;158(1):30–45. doi: 10.1111/j.1365-2141.2012.09129.x. [DOI] [PubMed] [Google Scholar]
  • 8.Bombí JA, Nadal A, Carreras E, Ramírez J, Muñoz J, Rozman C, Cardesa A. Assessment of histopathologic changes in the colonic biopsy in acute graft-versus-host disease. Am J Clin Pathol. 1995 Cze;103(6):690–5. doi: 10.1093/ajcp/103.6.690. [DOI] [PubMed] [Google Scholar]
  • 9.Ponec RJ, Hackman RC, McDonald GB. Endoscopic and histologic diagnosis of intestinal graft-versus-host disease after marrow transplantation. Gastrointest Endosc. 1999 Maj;49(5):612–21. doi: 10.1016/s0016-5107(99)70390-1. [DOI] [PubMed] [Google Scholar]
  • 10.Liakopoulou E, Mutton K, Carrington D, Robinson S, Steward CG, Goulden NJ, Cornish JM, Marks DI. Rotavirus as a significant cause of prolonged diarrhoeal illness and morbidity following allogeneic bone marrow transplantation. Bone Marrow Transplant. 2005 Paÿ;36(8):691–4. doi: 10.1038/sj.bmt.1705127. [DOI] [PubMed] [Google Scholar]
  • 11.Selbst MK, Ahrens WA, Robert ME, Friedman A, Proctor DD, Jain D. Spectrum of histologic changes in colonic biopsies in patients treated with mycophenolate mofetil. Mod Pathol. 2009 Cze;22(6):737–43. doi: 10.1038/modpathol.2009.44. [DOI] [PubMed] [Google Scholar]
  • 12.Alonso CD, Treadway SB, Hanna DB, Huff CA, Neofytos D, Carroll KC, Marr KA. Epidemiology and outcomes of Clostridium difficile infections in hematopoietic stem cell transplant recipients. Clin Infect Dis. 2012 Kwi;54(8):1053–63. doi: 10.1093/cid/cir1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J, Thomas ED. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995 Cze;15(6):825–8. [PubMed] [Google Scholar]
  • 14.Rowlings PA, Przepiorka D, Klein JP, Gale RP, Passweg JR, Henslee-Downey PJ, Cahn JY, Calderwood S, Gratwohl A, Socié G, Abecasis MM, Sobocinski KA, Zhang MJ, Horowitz MM. IBMTR Severity Index for grading acute graft-versus-host disease: retrospective comparison with Glucksberg grade. Br J Haematol. 1997 Cze;97(4):855–64. doi: 10.1046/j.1365-2141.1997.1112925.x. [DOI] [PubMed] [Google Scholar]
  • 15.MacMillan ML, Robin M, Harris AC, DeFor TE, Martin PJ, Alousi A, Ho VT, Bolaños-Meade J, Ferrara JLM, Jones R, Arora M, Blazar BR, Holtan SG, Jacobsohn D, Pasquini M, Socie G, Antin JH, Levine JE, Weisdorf DJ. A refined risk score for acute graft-versus-host disease that predicts response to initial therapy, survival, and transplant-related mortality. Biol Blood Marrow Transplant. 2015 Kwi;21(4):761–7. doi: 10.1016/j.bbmt.2015.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Harris AC, Young R, Devine S, Hogan WJ, Ayuk F, Bunworasate U, Chanswangphuwana C, Efebera YA, Holler E, Litzow M, Ordemann R, Qayed M, Renteria AS, Reshef R, Wölfl M, Chen Y-B, Goldstein S, Jagasia M, Locatelli F, Mielke S, Porter D, Schechter T, Shekhovtsova Z, Ferrara JLM, Levine JE. International, Multicenter Standardization of Acute Graft-versus-Host Disease Clinical Data Collection: A Report from the Mount Sinai Acute GVHD International Consortium. Biol Blood Marrow Transplant. 2016 Sty;22(1):4–10. doi: 10.1016/j.bbmt.2015.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ruutu T, Gratwohl A, de Witte T, Afanasyev B, Apperley J, Bacigalupo A, Dazzi F, Dreger P, Duarte R, Finke J, Garderet L, Greinix H, Holler E, Kröger N, Lawitschka A, Mohty M, Nagler A, Passweg J, Ringdén O, Socié G, Sierra J, Sureda A, Wiktor-Jedrzejczak W, Madrigal A, Niederwieser D. Prophylaxis and treatment of GVHD: EBMT-ELN working group recommendations for a standardized practice. Bone Marrow Transplant. 2014 Luty;49(2):168–73. doi: 10.1038/bmt.2013.107. [DOI] [PubMed] [Google Scholar]
  • 18.Mielcarek M, Furlong T, Storer BE, Green ML, McDonald GB, Carpenter PA, Flowers MED, Storb R, Boeckh M, Martin PJ. Effectiveness and safety of lower dose prednisone for initial treatment of acute graft-versus-host disease: a randomized controlled trial. Haematologica. 2015 Luty;100(6):842–848. doi: 10.3324/haematol.2014.118471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.MacMillan ML, Weisdorf DJ, Wagner JE, DeFor TE, Burns LJ, Ramsay NKC, Davies SM, Blazar BR. Response of 443 patients to steroids as primary therapy for acute graft-versus-host disease: comparison of grading systems. Biol Blood Marrow Transplant. 2002 Sty;8(7):387–94. doi: 10.1053/bbmt.2002.v8.pm12171485. [DOI] [PubMed] [Google Scholar]
  • 20.Levine JE, Logan B, Wu J, Alousi AM, Ho V, Bolaños-Meade J, Weisdorf D. Graft-versus-host disease treatment: predictors of survival. Biol Blood Marrow Transplant. 2010 Grudz;16(12):1693–9. doi: 10.1016/j.bbmt.2010.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gooley TA, Chien JW, Pergam SA, Hingorani S, Sorror ML, Boeckh M, Martin PJ, Sandmaier BM, Marr KA, Appelbaum FR, Storb R, McDonald GB. Reduced mortality after allogeneic hematopoietic-cell transplantation. N Engl J Med. 2010 Lis;363(22):2091–101. doi: 10.1056/NEJMoa1004383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jenq RR, van den Brink MRM. Allogeneic haematopoietic stem cell transplantation: individualized stem cell and immune therapy of cancer. Nat Rev Cancer. 2010 Mar;10(3):213–21. doi: 10.1038/nrc2804. [DOI] [PubMed] [Google Scholar]
  • 23.Lochmanová G, Jedličková L, Potěšil D, Tomancová A, Verner J, Pospíšilová S, Doubek M, Mayer J, Zdráhal Z. Potential biomarkers for early detection of acute graft-versus-host disease. Proteomics Clin Appl. 2012 Sie;6(7–8):351–63. doi: 10.1002/prca.201100104. [DOI] [PubMed] [Google Scholar]
  • 24.Lau J, Sartor M, Bradstock KF, Vuckovic S, Munster DJ, Hart DNJ. Activated circulating dendritic cells after hematopoietic stem cell transplantation predict acute graft-versus-host disease. Transplantation. 2007 Kwi;83(7):839–46. doi: 10.1097/01.tp.0000258731.38149.61. [DOI] [PubMed] [Google Scholar]
  • 25.Magenau JM, Qin X, Tawara I, Rogers CE, Kitko C, Schlough M, Bickley D, Braun TM, Jang P-S, Lowler KP, Jones DM, Choi SW, Reddy P, Mineishi S, Levine JE, Ferrara JLM, Paczesny S. Frequency of CD4(+)CD25(hi)FOXP3(+) regulatory T cells has diagnostic and prognostic value as a biomarker for acute graft-versus-host-disease. Biol Blood Marrow Transplant. 2010 Lip;16(7):907–14. doi: 10.1016/j.bbmt.2010.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Teshima T, Maeda Y, Ozaki K. Regulatory T cells and IL-17-producing cells in graft-versus-host disease. Immunotherapy. 2011 Lip;3(7):833–52. doi: 10.2217/imt.11.51. [DOI] [PubMed] [Google Scholar]
  • 27.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009 Sty;136(2):215–33. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ng EKO, Chong WWS, Jin H, Lam EKY, Shin VY, Yu J, Poon TCW, Ng SSM, Sung JJY. Differential expression of microRNAs in plasma of patients with colorectal cancer: a potential marker for colorectal cancer screening. Gut. 2009 Paÿ;58(10):1375–81. doi: 10.1136/gut.2008.167817. [DOI] [PubMed] [Google Scholar]
  • 29.Li L-M, Hu Z-B, Zhou Z-X, Chen X, Liu F-Y, Zhang J-F, Shen H-B, Zhang C-Y, Zen K. Serum microRNA profiles serve as novel biomarkers for HBV infection and diagnosis of HBV-positive hepatocarcinoma. Cancer Res. 2010 Grudz;70(23):9798–807. doi: 10.1158/0008-5472.CAN-10-1001. [DOI] [PubMed] [Google Scholar]
  • 30.Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, Mayr A, Weger S, Oberhollenzer F, Bonora E, Shah A, Willeit J, Mayr M. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010 Wrz;107(6):810–7. doi: 10.1161/CIRCRESAHA.110.226357. [DOI] [PubMed] [Google Scholar]
  • 31.Mas VR, Dumur CI, Scian MJ, Gehrau RC, Maluf DG. MicroRNAs as biomarkers in solid organ transplantation. Am J Transplant. 2013 Sty;13(1):11–9. doi: 10.1111/j.1600-6143.2012.04313.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ranganathan P, Heaphy CEA, Costinean S, Stauffer N, Na C, Hamadani M, Santhanam R, Mao C, Taylor PA, Sandhu S, He G, Shana’ah A, Nuovo GJ, Lagana A, Cascione L, Obad S, Broom O, Kauppinen S, Byrd JC, Caligiuri M, Perrotti D, Hadley GA, Marcucci G, Devine SM, Blazar BR, Croce CM, Garzon R. Regulation of acute graft-versus-host disease by microRNA-155. Blood. 2012 Maj;119(20):4786–97. doi: 10.1182/blood-2011-10-387522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Xie L-N, Zhou F, Liu X-M, Fang Y, Yu Z, Song N-X, Kong F-S. Serum microRNA155 is increased in patients with acute graft-versus-host disease. Clin Transplant. 2014 Mar;28(3):314–23. doi: 10.1111/ctr.12314. [DOI] [PubMed] [Google Scholar]
  • 34.Wang Y, Zhao X, Ye X, Luo H, Zhao T, Diao Y, Zhang H, Lv M, Zhang W, Huang X, Wan J. Plasma microRNA-586 is a new biomarker for acute graft-versus-host disease. Ann Hematol. 2015 Wrz;94(9):1505–14. doi: 10.1007/s00277-015-2414-z. [DOI] [PubMed] [Google Scholar]
  • 35.Xiao B, Wang Y, Li W, Baker M, Guo J, Corbet K, Tsalik EL, Li Q-J, Palmer SM, Woods CW, Li Z, Chao NJ, He Y-W. Plasma microRNA signature as a noninvasive biomarker for acute graft-versus-host disease. Blood. 2013 Lis;122(19):3365–75. doi: 10.1182/blood-2013-06-510586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tili E, Croce CM, Michaille J-J. miR-155: on the crosstalk between inflammation and cancer. Int Rev Immunol. 2009 Sty;28(5):264–84. doi: 10.1080/08830180903093796. [DOI] [PubMed] [Google Scholar]
  • 37.Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008 Lip;105(30):10513–8. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chang J, Guo J-T, Jiang D, Guo H, Taylor JM, Block TM. Liver-specific microRNA miR-122 enhances the replication of hepatitis C virus in nonhepatic cells. J Virol. 2008 Sie;82(16):8215–23. doi: 10.1128/JVI.02575-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011 Kwi;117(14):3720–32. doi: 10.1182/blood-2010-07-273417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ito S, Barrett AJ. ST2: the biomarker at the heart of GVHD severity. Blood. 2015 Sty;125(1):10–1. doi: 10.1182/blood-2014-11-611780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Reichenbach DK, Schwarze V, Matta BM, Tkachev V, Lieberknecht E, Liu Q, Koehn BH, Pfeifer D, Taylor PA, Prinz G, Dierbach H, Stickel N, Beck Y, Warncke M, Junt T, Schmitt-Graeff A, Nakae S, Follo M, Wertheimer T, Schwab L, Devlin J, Watkins SC, Duyster J, Ferrara JLM, Turnquist HR, Zeiser R, Blazar BR. The IL-33/ST2 axis augments effector T-cell responses during acute GVHD. Blood. 2015 Maj;125(20):3183–92. doi: 10.1182/blood-2014-10-606830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, Zurawski G, Moshrefi M, Qin J, Li X, Gorman DM, Bazan JF, Kastelein RA. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005 Lis;23(5):479–90. doi: 10.1016/j.immuni.2005.09.015. [DOI] [PubMed] [Google Scholar]
  • 43.Vander Lugt MT, Braun TM, Hanash S, Ritz J, Ho VT, Antin JH, Zhang Q, Wong C-H, Wang H, Chin A, Gomez A, Harris AC, Levine JE, Choi SW, Couriel D, Reddy P, Ferrara JLM, Paczesny S. ST2 as a marker for risk of therapy-resistant graft-versus-host disease and death. N Engl J Med. 2013 Sie;369(6):529–39. doi: 10.1056/NEJMoa1213299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ponce DM, Hilden P, Mumaw C, Devlin SM, Lubin M, Giralt S, Goldberg JD, Hanash A, Hsu K, Jenq R, Perales M-A, Sauter C, van den Brink MRM, Young JW, Brentjens R, Kernan NA, Prockop SE, O’Reilly RJ, Scaradavou A, Paczesny S, Barker JN. High day 28 ST2 levels predict for acute graft-versus-host disease and transplant-related mortality after cord blood transplantation. Blood. 2015 Sty;125(1):199–205. doi: 10.1182/blood-2014-06-584789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Henden AS, Hill GR. Cytokines in Graft-versus-Host Disease. J Immunol. 2015 Maj;194(10):4604–12. doi: 10.4049/jimmunol.1500117. [DOI] [PubMed] [Google Scholar]
  • 46.Remberger M, Ringden O, Markling L. TNF alpha levels are increased during bone marrow transplantation conditioning in patients who develop acute GVHD. Bone Marrow Transplant. 1995 Sty;15(1):99–104. [PubMed] [Google Scholar]
  • 47.Choi SW, Kitko CL, Braun T, Paczesny S, Yanik G, Mineishi S, Krijanovski O, Jones D, Whitfield J, Cooke K, Hutchinson RJ, Ferrara JLM, Levine JE. Change in plasma tumor necrosis factor receptor 1 levels in the first week after myeloablative allogeneic transplantation correlates with severity and incidence of GVHD and survival. Blood. 2008 Sie;112(4):1539–42. doi: 10.1182/blood-2008-02-138867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kitko CL, Paczesny S, Yanik G, Braun T, Jones D, Whitfield J, Choi SW, Hutchinson RJ, Ferrara JLM, Levine JE. Plasma elevations of tumor necrosis factor-receptor-1 at day 7 postallogeneic transplant correlate with graft-versus-host disease severity and overall survival in pediatric patients. Biol Blood Marrow Transplant. 2008 Lip;14(7):759–65. doi: 10.1016/j.bbmt.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Willems E, Humblet-Baron S, Dengis O, Seidel L, Beguin Y, Baron F. Elevations of tumor necrosis factor receptor 1 at day 7 and acute graft-versus-host disease after allogeneic hematopoietic cell transplantation with nonmyeloablative conditioning. Bone Marrow Transplant. 2010 Wrz;45(9):1442–8. doi: 10.1038/bmt.2009.360. [DOI] [PubMed] [Google Scholar]
  • 50.August KJ, Chiang K-Y, Bostick RM, Flanders WD, Waller EK, Langston A, Worthington-White D, Rowland P, Moore KF, Khoury HJ, Horan JT. Biomarkers of immune activation to screen for severe, acute GVHD. Bone Marrow Transplant. 2011 Kwi;46(4):601–4. doi: 10.1038/bmt.2010.165. [DOI] [PubMed] [Google Scholar]
  • 51.Chung B, Dudl E, Toyama A, Barsky L, Weinberg KI. Importance of Interleukin-7 in the Development of Experimental Graft-Versus-Host Disease. Biol Blood Marrow Transplant. 2008 Sty;14(1):16–27. doi: 10.1016/j.bbmt.2007.07.015. [DOI] [PubMed] [Google Scholar]
  • 52.Chung B, Dudl EP, Min D, Barsky L, Smiley N, Weinberg KI. Prevention of graft-versus-host disease by anti IL-7Ralpha antibody. Blood. 2007 Paÿ;110(8):2803–10. doi: 10.1182/blood-2006-11-055673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Dean RM, Fry T, Mackall C, Steinberg SM, Hakim F, Fowler D, Odom J, Foley J, Gress R, Bishop MR. Association of serum interleukin-7 levels with the development of acute graft-versus-host disease. J Clin Oncol. 2008 Grudz;26(35):5735–41. doi: 10.1200/JCO.2008.17.1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Thiant S, Yakoub-Agha I, Magro L, Trauet J, Coiteux V, Jouet J-P, Dessaint J-P, Labalette M. Plasma levels of IL-7 and IL-15 in the first month after myeloablative BMT are predictive biomarkers of both acute GVHD and relapse. Bone Marrow Transplant. 2010 Paÿ;45(10):1546–52. doi: 10.1038/bmt.2010.13. [DOI] [PubMed] [Google Scholar]
  • 55.Thiant S, Labalette M, Trauet J, Coiteux V, de Berranger E, Dessaint J-P, Yakoub-Agha I. Plasma levels of IL-7 and IL-15 after reduced intensity conditioned allo-SCT and relationship to acute GVHD. Bone Marrow Transplant. 2011 Paÿ;46(10):1374–81. doi: 10.1038/bmt.2010.300. [DOI] [PubMed] [Google Scholar]
  • 56.Savani BN, Mielke S, Rezvani K, Montero A, Yong AS, Wish L, Superata J, Kurlander R, Singh A, Childs R, Barrett AJ. Absolute lymphocyte count on day 30 is a surrogate for robust hematopoietic recovery and strongly predicts outcome after T cell-depleted allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2007 Paÿ;13(10):1216–23. doi: 10.1016/j.bbmt.2007.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cho B-S, Min C-K, Kim H-J, Lee S, Kim Y-J, Lim J-Y, Jeong D-C, Cho B, Kim H-K, Eom K-S, Cho S-G, Kim D-W, Lee J-W, Min W-S, Kim C-C, Chung N-G. High levels of B cell activating factor during the peritransplantation period are associated with a reduced incidence of acute graft-versus-host disease following myeloablative allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2010 Maj;16(5):629–38. doi: 10.1016/j.bbmt.2009.11.023. [DOI] [PubMed] [Google Scholar]
  • 58.Sarantopoulos S, Stevenson KE, Kim HT, Cutler CS, Bhuiya NS, Schowalter M, Ho VT, Alyea EP, Koreth J, Blazar BR, Soiffer RJ, Antin JH, Ritz J. Altered B-cell homeostasis and excess BAFF in human chronic graft-versus-host disease. Blood. 2009 Kwi;113(16):3865–74. doi: 10.1182/blood-2008-09-177840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ratanatharathorn V, Logan B, Wang D, Horowitz M, Uberti JP, Ringden O, Gale RP, Khoury H, Arora M, Spellman S, Cutler C, Antin J, Bornhaüser M, Hale G, Verdonck L, Cairo M, Gupta V, Pavletic S. Prior rituximab correlates with less acute graft-versus-host disease and better survival in B-cell lymphoma patients who received allogeneic peripheral blood stem cell transplantation. Br J Haematol. 2009 Cze;145(6):816–24. doi: 10.1111/j.1365-2141.2009.07674.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ye Q, Wang L, Wells AD, Tao R, Han R, Davidson A, Scott ML, Hancock WW. BAFF binding to T cell-expressed BAFF-R costimulates T cell proliferation and alloresponses. Eur J Immunol. 2004 Paÿ;34(10):2750–9. doi: 10.1002/eji.200425198. [DOI] [PubMed] [Google Scholar]
  • 61.Takashima S, Kadowaki M, Aoyama K, Koyama M, Oshima T, Tomizuka K, Akashi K, Teshima T. The Wnt agonist R-spondin1 regulates systemic graft-versus-host disease by protecting intestinal stem cells. J Exp Med. 2011 Luty;208(2):285–94. doi: 10.1084/jem.20101559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Harris AC, Ferrara JLM, Braun TM, Holler E, Teshima T, Levine JE, Choi SW, Landfried K, Akashi K, Vander Lugt M, Couriel DR, Reddy P, Paczesny S. Plasma biomarkers of lower gastrointestinal and liver acute GVHD. Blood. 2012 Mar;119(12):2960–3. doi: 10.1182/blood-2011-10-387357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ferrara JLM, Harris AC, Greenson JK, Braun TM, Holler E, Teshima T, Levine JE, Choi SWJ, Huber E, Landfried K, Akashi K, Vander Lugt M, Reddy P, Chin A, Zhang Q, Hanash S, Paczesny S. Regenerating islet-derived 3-alpha is a biomarker of gastrointestinal graft-versus-host disease. Blood. 2011 Grudz;118(25):6702–8. doi: 10.1182/blood-2011-08-375006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Foell D, Roth J. Proinflammatory S100 proteins in arthritis and autoimmune disease. Arthritis Rheum. 2004 Grudz;50(12):3762–71. doi: 10.1002/art.20631. [DOI] [PubMed] [Google Scholar]
  • 65.Foell D, Wittkowski H, Ren Z, Turton J, Pang G, Daebritz J, Ehrchen J, Heidemann J, Borody T, Roth J, Clancy R. Phagocyte-specific S100 proteins are released from affected mucosa and promote immune responses during inflammatory bowel disease. J Pathol. 2008 Paÿ;216(2):183–92. doi: 10.1002/path.2394. [DOI] [PubMed] [Google Scholar]
  • 66.Reinhardt K, Foell D, Vogl T, Mezger M, Wittkowski H, Fend F, Federmann B, Gille C, Feuchtinger T, Lang P, Handgretinger R, Andreas Bethge W, Holzer U. Monocyte-induced development of Th17 cells and the release of S100 proteins are involved in the pathogenesis of graft-versus-host disease. J Immunol. 2014 Paÿ;193(7):3355–65. doi: 10.4049/jimmunol.1400983. [DOI] [PubMed] [Google Scholar]
  • 67.Rodriguez-Otero P, Porcher R, Peffault de Latour R, Contreras M, Bouhnik Y, Xhaard A, Andreoli A, Ribaud P, Kapel N, Janin A, Socié G, Robin M. Fecal calprotectin and alpha-1 antitrypsin predict severity and response to corticosteroids in gastrointestinal graft-versus-host disease. Blood. 2012 Cze;119(24):5909–17. doi: 10.1182/blood-2011-12-397968. [DOI] [PubMed] [Google Scholar]
  • 68.Chiusolo P, Metafuni E, Giammarco S, Bellesi S, Piccirillo N, Fanali C, Castagnola M, Zuppi C, De Michele T, Leone G, Sica S. Role of fecal calprotectin as biomarker of gastrointestinal GVHD after allogeneic stem cell transplantation. Blood. 2012 Lis;120(22):4443–4. doi: 10.1182/blood-2012-08-447326. [DOI] [PubMed] [Google Scholar]
  • 69.Kuchroo VK, Umetsu DT, DeKruyff RH, Freeman GJ. The TIM gene family: emerging roles in immunity and disease. Nat Rev Immunol. 2003 Cze;3(6):454–62. doi: 10.1038/nri1111. [DOI] [PubMed] [Google Scholar]
  • 70.Geng H, Zhang G-M, Li D, Zhang H, Yuan Y, Zhu H-G, Xiao H, Han L-F, Feng Z-H. Soluble Form of T Cell Ig Mucin 3 Is an Inhibitory Molecule in T Cell-Mediated Immune Response. J Immunol. 2006 Sty;176(3):1411–1420. doi: 10.4049/jimmunol.176.3.1411. [DOI] [PubMed] [Google Scholar]
  • 71.Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, Manning S, Greenfield EA, Coyle AJ, Sobel RA, Freeman GJ, Kuchroo VK. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002 Sty;415(6871):536–41. doi: 10.1038/415536a. [DOI] [PubMed] [Google Scholar]
  • 72.Sehrawat S, Reddy PBJ, Rajasagi N, Suryawanshi A, Hirashima M, Rouse BT. Galectin-9/TIM-3 interaction regulates virus-specific primary and memory CD8 T cell response. PLoS Pathog. 2010 Maj;6(5):e1000882. doi: 10.1371/journal.ppat.1000882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hastings WD, Anderson DE, Kassam N, Koguchi K, Greenfield EA, Kent SC, Zheng XX, Strom TB, Hafler DA, Kuchroo VK. TIM-3 is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. Eur J Immunol. 2009 Wrz;39(9):2492–501. doi: 10.1002/eji.200939274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Oikawa T, Kamimura Y, Akiba H, Yagita H, Okumura K, Takahashi H, Zeniya M, Tajiri H, Azuma M. Preferential involvement of Tim-3 in the regulation of hepatic CD8+ T cells in murine acute graft-versus-host disease. J Immunol. 2006 Paÿ;177(7):4281–7. doi: 10.4049/jimmunol.177.7.4281. [DOI] [PubMed] [Google Scholar]
  • 75.Veenstra RG, Taylor PA, Zhou Q, Panoskaltsis-Mortari A, Hirashima M, Flynn R, Liu D, Anderson AC, Strom TB, Kuchroo VK, Blazar BR. Contrasting acute graft-versus-host disease effects of Tim-3/galectin-9 pathway blockade dependent upon the presence of donor regulatory T cells. Blood. 2012 Lip;120(3):682–90. doi: 10.1182/blood-2011-10-387977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hansen JA, Hanash SM, Tabellini L, Baik C, Lawler RL, Grogan BM, Storer B, Chin A, Johnson M, Wong C-H, Zhang Q, Martin PJ, McDonald GB. A novel soluble form of Tim-3 associated with severe graft-versus-host disease. Biol Blood Marrow Transplant. 2013 Wrz;19(9):1323–30. doi: 10.1016/j.bbmt.2013.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.McDonald GB, Tabellini L, Storer BE, Lawler RL, Martin PJ, Hansen JA. Plasma biomarkers of acute GVHD and nonrelapse mortality: predictive value of measurements before GVHD onset and treatment. Blood. 2015 Lip;126(1):113–20. doi: 10.1182/blood-2015-03-636753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chu PG, Weiss LM. Keratin expression in human tissues and neoplasms. Histopathology. 2002 Maj;40(5):403–39. doi: 10.1046/j.1365-2559.2002.01387.x. [DOI] [PubMed] [Google Scholar]
  • 79.Bivén K, Erdal H, Hägg M, Ueno T, Zhou R, Lynch M, Rowley B, Wood J, Zhang C, Toi M, Shoshan MC, Linder S. A novel assay for discovery and characterization of pro-apoptotic drugs and for monitoring apoptosis in patient sera. Apoptosis. 2003 Cze;8(3):263–8. doi: 10.1023/a:1023672805949. [DOI] [PubMed] [Google Scholar]
  • 80.Shulman HM, Kleiner D, Lee SJ, Morton T, Pavletic SZ, Farmer E, Moresi JM, Greenson J, Janin A, Martin PJ, McDonald G, Flowers MED, Turner M, Atkinson J, Lefkowitch J, Washington MK, Prieto VG, Kim SK, Argenyi Z, Diwan AH, Rashid A, Hiatt K, Couriel D, Schultz K, Hymes S, Vogelsang GB. Histopathologic diagnosis of chronic graft-versus-host disease: National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: II. Pathology Working Group Report. Biol Blood Marrow Transplant. 2006 Sty;12(1):31–47. doi: 10.1016/j.bbmt.2005.10.023. [DOI] [PubMed] [Google Scholar]
  • 81.Luft T, Conzelmann M, Benner A, Rieger M, Hess M, Strohhaecker U, Görner M, Hegenbart U, Ho AD, Dreger P. Serum cytokeratin-18 fragments as quantitative markers of epithelial apoptosis in liver and intestinal graft-versus-host disease. Blood. 2007 Grudz;110(13):4535–42. doi: 10.1182/blood-2006-10-049817. [DOI] [PubMed] [Google Scholar]
  • 82.Luft T, Dietrich S, Falk C, Conzelmann M, Hess M, Benner A, Neumann F, Isermann B, Hegenbart U, Ho AD, Dreger P. Steroid-refractory GVHD: T-cell attack within a vulnerable endothelial system. Blood. 2011 Sie;118(6):1685–92. doi: 10.1182/blood-2011-02-334821. [DOI] [PubMed] [Google Scholar]
  • 83.Matsumoto K, Nakamura T. Emerging multipotent aspects of hepatocyte growth factor. J Biochem. 1996 Kwi;119(4):591–600. doi: 10.1093/oxfordjournals.jbchem.a021283. [DOI] [PubMed] [Google Scholar]
  • 84.Nakamura T, Nishizawa T, Hagiya M, Seki T, Shimonishi M, Sugimura A, Tashiro K, Shimizu S. Molecular cloning and expression of human hepatocyte growth factor. Nature. 1989 Lis;342(6248):440–3. doi: 10.1038/342440a0. [DOI] [PubMed] [Google Scholar]
  • 85.Shiota G, Okano J-I, Kawasaki H, Kawamoto T, Nakamura T. Serum hepatocyte growth factor levels in liver diseases: Clinical implications. Hepatology. 1995 Sty;21(1):106–112. [PubMed] [Google Scholar]
  • 86.Hino M, Inaba M, Goto H, Nishizawa Y, Tatsumi N, Nishino T, Morii H. Hepatocyte growth factor levels in bone marrow plasma of patients with leukaemia and its gene expression in leukaemic blast cells. Br J Cancer. 1996 Sty;73(1):119–23. doi: 10.1038/bjc.1996.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Taniguchi T, Toi M, Inada K, Imazawa T, Yamamoto Y, Tominaga T. Serum concentrations of hepatocyte growth factor in breast cancer patients. Clin Cancer Res. 1995 Wrz;1(9):1031–4. [PubMed] [Google Scholar]
  • 88.Paczesny S, Krijanovski OI, Braun TM, Choi SW, Clouthier SG, Kuick R, Misek DE, Cooke KR, Kitko CL, Weyand A, Bickley D, Jones D, Whitfield J, Reddy P, Levine JE, Hanash SM, Ferrara JLM. A biomarker panel for acute graft-versus-host disease. Blood. 2009 Sty;113(2):273–8. doi: 10.1182/blood-2008-07-167098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nonomura K, Yamanishi K, Yasuno H, Nara K, Hirose S. Up-regulation of elafin/SKALP gene expression in psoriatic epidermis. J Invest Dermatol. 1994 Lip;103(1):88–91. doi: 10.1111/1523-1747.ep12391802. [DOI] [PubMed] [Google Scholar]
  • 90.Pfundt R, Wingens M, Bergers M, Zweers M, Frenken M, Schalkwijk J. TNF-alpha and serum induce SKALP/elafin gene expression in human keratinocytes by a p38 MAP kinase-dependent pathway. Arch Dermatol Res. 2000 Kwi;292(4):180–7. doi: 10.1007/s004030050475. [DOI] [PubMed] [Google Scholar]
  • 91.Paczesny S, Braun TM, Levine JE, Hogan J, Crawford J, Coffing B, Olsen S, Choi SW, Wang H, Faca V, Pitteri S, Zhang Q, Chin A, Kitko C, Mineishi S, Yanik G, Peres E, Hanauer D, Wang Y, Reddy P, Hanash S, Ferrara JLM. Elafin is a biomarker of graft-versus-host disease of the skin. Sci Transl Med. 2010 Sty;2(13):13ra2. doi: 10.1126/scitranslmed.3000406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Levine JE, Logan BR, Wu J, Alousi AM, Bolaños-Meade J, Ferrara JLM, Ho VT, Weisdorf DJ, Paczesny S. Acute graft-versus-host disease biomarkers measured during therapy can predict treatment outcomes: a Blood and Marrow Transplant Clinical Trials Network study. Blood. 2012 Kwi;119(16):3854–60. doi: 10.1182/blood-2012-01-403063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Levine JE, Braun TM, Harris AC, Holler E, Taylor A, Miller H, Magenau J, Weisdorf DJ, Ho VT, Bolaños-Meade J, Alousi AM, Ferrara LM. A PROGNOSTIC SCORE FOR ACUTE GRAFT-VERSUS-HOST DISEASE BASED ON BIOMARKERS: A MULTICENTER STUDY. Lancet Haematol. 2015 Sty;2(1):e21–e29. doi: 10.1016/S2352-3026(14)00035-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Te Boome LCJ, Mansilla C, van der Wagen LE, Lindemans CA, Petersen EJ, Spierings E, Thus KA, Westinga K, Plantinga M, Bierings M, Broers AEC, Cuijpers MLH, van Imhoff GW, Janssen JJ, Huisman C, Zeerleder S, Huls G, Boelens JJ, Wulffraat NM, Slaper-Cortenbach ICM, Kuball J. Biomarker profiling of steroid-resistant acute GVHD in patients after infusion of mesenchymal stromal cells. Leukemia. 2015 Wrz;29(9):1839–46. doi: 10.1038/leu.2015.89. [DOI] [PubMed] [Google Scholar]
  • 95.Lv M, Ye H, Zhao X, Zhao X, Chang Y, Liu D. Ceruloplasmin is a potential biomarker for aGvHD following allogeneic hematopoietic stem cell transplantation. PLoS One. 2013 Styczeń;8(3):e58735. doi: 10.1371/journal.pone.0058735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhu F, Qiao J, Chen W, Pan B, Wu Q, Cao J. Increased expression of T cell immune response cDNA 7 in patients with acute graft-versus-host disease. Ann Hematol. 2015 Czerwiec;94(6):1025–32. doi: 10.1007/s00277-015-2300-8. [DOI] [PubMed] [Google Scholar]
  • 97.Okamoto T, Takatsuka H, Fujimori Y, Wada H, Iwasaki T, Kakishita E. Increased hepatocyte growth factor in serum in acute graft-versus-host disease. Bone Marrow Transplant. 2001 Lipiec;28(2):197–200. doi: 10.1038/sj.bmt.1703095. [DOI] [PubMed] [Google Scholar]
  • 98.Chacon AH, Farooq U, Shiman MI, Elgart GW. Elafin: a possible new biomarker and immunohistochemical stain for pre-engraftment syndrome. J Am Acad Dermatol sierpień. 2013;69(2):e102–3. doi: 10.1016/j.jaad.2012.11.024. [DOI] [PubMed] [Google Scholar]

RESOURCES