Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A mathematical model for dynamics of soluble form of DNAM-1 as a biomarker for graft-versus-host disease

  • Yuki Goshima,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Software, Validation, Visualization, Writing – original draft

    Affiliations Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan, School of Medicine, University of Tsukuba, Tsukuba, Japan

  • Shinji Nakaoka,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Software, Supervision

    Affiliations Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido, Japan, PRESTO, Japan Science and Technology Agency, Saitama, Japan

  • Kazuteru Ohashi,

    Roles Data curation

    Affiliation Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan

  • Hisashi Sakamaki,

    Roles Data curation

    Affiliation Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan

  • Kazuko Shibuya,

    Roles Conceptualization, Investigation, Supervision

    Affiliations Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan

  • Akira Shibuya

    Roles Conceptualization, Funding acquisition, Investigation, Project administration, Supervision, Writing – original draft, Writing – review & editing

    ashibuya@md.tsukuba.ac.jp

    Affiliations Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan, Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan

Abstract

DNAM-1 (CD226) is an activating immunoreceptor expressed on T cells and NK cells and involved in the pathogenesis of acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We previously reported that a soluble form of DNAM-1 (sDNAM-1) is generated by shedding from activated T cells. Moreover, higher serum levels of sDNAM-1 in patients before allo-HSCT is a predictive biomarker for the development of aGVHD based on the retrospective univariate and multivariate analyses in allo-HSCT patients. However, it remains unclear how the serum levels of sDNAM-1 are regulated after allo-HSCT and whether they are associated with the development of aGVHD. Here, we constructed a mathematical model to assess the dynamics of sDNAM-1 after allo-HSCT by assuming that there are three types of sDNAM-1 (the first and the second were from alloreactive and non-alloreactive donor lymphocytes, respectively, and the third from recipient lymphocytes). Our mathematical model fitted well to the data set of sDNAM-1 in patients (n = 67) who had undergone allo-HSCT and suggest that the high proportion of the first type of sDNAM-1 to the total of the first and second types is associated with high risk of the development of severe aGVHD. Thus, sDNAM-1 after allo-HSCT can be a biomarker for the development of aGVHD.

Introduction

Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Although the mechanism underlying the development of aGVHD has been extensively studied in vitro and in vivo [13], the diagnosis of and treatment for aGVHD are still problematic.

DNAM-1, also known as CD226, is an activating immunoreceptor expressed on CD4+ T cells, CD8+ T cells, natural killer (NK) cells, and monocytes [4]. We and others demonstrated that DNAM-1 plays an important role in the development of aGVHD in mouse models [5,6]. Moreover, we have recently identified a soluble form of DNAM-1 (sDNAM-1), which is shed from the membrane type of DNAM-1 expressed on the cell surface of activated T lymphocytes, in human sera [7]. We performed retrospective univariate and multivariate analyses of serum levels of sDNAM-1 in patients before and after allo-HSCT at a single center (n = 71) [7]. We demonstrated that cumulative incidences of all grade (grade I–IV) and grade II–IV aGVHD in patients with high maximal serum levels of sDNAM-1 (≥30 pM) in the 7 days before allo-HSCT were significantly higher than those in patients with low maximal serum levels of sDNAM-1 (<30 pM) in the same period, and concluded that the serum levels of sDNAM-1 can be a predictive biomarker for the development of aGVHD [7]. However, it remains unclear how the dynamics of serum levels of sDNAM-1 after allo-HSCT is regulated and whether it is associated with the development of aGVHD.

In this study, we constructed a mathematical model to assess the dynamics of serum levels of sDNAM-1, and revisited the data set of sDNAM-1, which had been analyzed previously [7], particularly after, rather than before, allo-HSCT to be applied by the mathematical model. We show that sDNAM-1 after allo-HSCT can be a biomarker for the development of aGVHD.

Materials and methods

Patients, samples, and inclusion criteria

Serum samples were obtained from 156 patients at the Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Japan, between March 2009 and November 2011. Data from some of the patients had already been analyzed [7]; we basically followed their methods in terms of such features as informed consent and sample handling but here we included other patients according to our new criteria which is shown in Fig 1. Written informed consent was obtained from each patient in accordance with the Declaration of Helsinki. This study was approved by the ethics committee of the University of Tsukuba (approval No., 505) and Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital (approval No., 571). Soluble DNAM-1 in sera was measured by sandwich enzyme-linked immunosorbent assay (ELISA), as described [7]. The data were collected from about 7 days before allo-HSCT up to a maximum of 249 days after allo-HSCT. Since our mathematical model required eight parameters and at least one parameter within 100 days and 7 days after allo-HSCT, respectively, we excluded patients who did not meet these criteria. As a result, 67 patients (GVHD (+) = 48 patients, GVHD (-) = 19 patients) satisfied our criteria (Fig 1); their characteristics are listed in Table 1.

thumbnail
Fig 1. Patient disposition.

From 156 patients analyzed for sDNAM-1 in the sera, we finally included 67 in our study. Details of the inclusion criteria are given in the Materials and Methods.

https://doi.org/10.1371/journal.pone.0228508.g001

thumbnail
Table 1. Patient characteristics and clinical information.

https://doi.org/10.1371/journal.pone.0228508.t001

Definition of grading system of acute GVHD and transplantation procedures

The GVHD grade of each patient was determined as previously described [8,9]. The transplantation procedures are described previously [7].

Mathematical model and statistical analysis

sDNAM-1 was detected in the sera of healthy people as well as patients before and after allo-HSCT. sDNAM-1 might be produced by either donor cells, recipient cells or both. Therefore, we modeled the dynamics of those cells using ordinary differential equations. Differential equations have been used for describing the virus and cell growth [10,11]. In our research, we firstly formulate the mathematical model to explain the dynamics of sDNAM-1. Secondly, we fitted this model to the patients’ data and got each patient’s parameter using Package FME [12] in R (version 3.4.3) (R Core Team (2018)). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. URL https://www.R-project.org/). As for statistical analysis, a t-test, Fisher’s exact test and Binomial test were used to analyze the value of Rday_n, patient demographic and clinical information, and comparison of Akaike’s Information Criterion (AIC) between two different mathematical models, respectively. For all tests, a P-value of less than 0.05 was considered as statistical significance.

Assessing goodness of fit to data and contribution of first type of sDNAM-1 to aGVHD.

We compared 2 mathematical models to assess their goodness of fit to the data. The model 1 is consisted of two secretors of sDNAM-1 (i.e., donor lymphocytes that persistently produce sDNAM-1 and recipient lymphocytes that had produced it before all-HSCT). On the other hand, the model 2 is consisted of three secretors of sDNAM-1 (i.e., donor lymphocytes that transiently release sDNAM-1 in addition to those in the model 1). Here, we used AIC, which can be used to compare two mathematical models [13].

Results

Mathematical model of sDNAM-1 dynamics

sDNAM-1 in sera of patients that received allo-HSCT can be derived from lymphocytes of either a donor, a recipient or both. Therefore, we assumed that sDNAM-1 in sera of patients consists of three types of sDNAM-1. The first and second types are transiently and persistently released from donor lymphocytes, respectively. The third one is a residual sDNAM-1 derived from recipient’s lymphocytes before allo-HSCT. Since it is difficult to discriminate the origins of serum sDNAM-1 detected by ELISA, we performed a computational simulation to estimate the concentration of each type of sDNAM-1. In our mathematical model, we denoted x1(t) as the concentration of the first type of sDNAM-1. We adopted a gamma distribution for x1(t), because the gamma function is a unimodal distribution and has a flexible shape, unlike the normal distribution. It can be used to express the feature of first type of sDNAM-1, namely transient growth. The second and third types of sDNAM-1 follows a logistic equation and an exponential decay pattern, which we denoted them as x2(t) and x3(t), respectively. The model is shown as follows: (1)

Definition of each parameter are given in S1 Table in the Supplementary Methods. x1(t)+x2(t)+x3(t) expresses the total amount of sDNAM-1. We fitted this model to the observed data and obtained the estimated parameters.

Definition of Rday_n and comparison of Rday_n between patients’ group with or without aGVHD

We defined Rday_n using a mathematical expression as follows: (2)

Integration of x1(t) gives an area under the curve (AUC). The AUC of x1(t) from 0 to day_n is the amount of sDNAM-1 that is secreted by the donor lymphocytes during n days after allo-HSCT. In order to assess the effect of sDNAM-1 from donor lymphocytes on acute GVHD, we excluded the third type of sDNAM-1, which is derived from recipient lymphocytes. Since donor-derived allo-reactive T cells attack the recipient’s tissues during acute GVHD [14], it is reasonable to focus on the first and second type of sDNAM-1 derived from donor lymphocytes. Therefore, Rday_n is the proportion of sDNAM-1, which is released transiently from donor lymphocytes. We plotted each distribution of Rday_n. The Rday_n distribution of patients with aGVHD (GVHD (+)) tended to shift to the right, whereas the distribution of patients without aGVHD (GVHD (-)) tended to shift to the left (Fig 2). We also compiled boxplots of Rday_n for GVHD (+) and GVHD (–) (n = 20, 30, 40, and 50). Rday_n of GVHD (+) was higher than that of GVHD (–) for each day (n = 20, 30, 40, and 50) (Fig 3). We performed t-test and Wilcoxon test and calculated the 95% CIs of mean & median of differences between GVHD (+) and GVHD (-) groups. The GVHD (+) group showed higher Rday_20, Rday_30, Rday_40, and Rday_50 than did the GVHD (–) group (Table 2). The differences in each estimated mean between the two groups exceeded 30% (Table 2). In addition, we analyzed the relationship between Rday_n (n = 20, 30, 40, and 50) with the grades of aGVHD. We found that the higher the grade, the more the value of Rday_n (n = 20, 30, 40, and 50) increased (Fig 4), suggesting that Rday_n reflected the grade of aGVHD. In addition, we also found the association of Rday_n with aGVHD in the skin and gastrointestinal tract, but not in the liver (S2S4 Figs, S2S4 Tables). In contrast, we did not observe the association of Rday_n with the factors of patient background (e.g., with or without CMV infection, total body irradiation, and HLA matching, and treatment for GVHD prophylaxis) (S5S8 Tables).

thumbnail
Fig 2. Distributions of Rday_n values for day n = 20, 30, 40, and 50.

Rday_n does not follow a normal distribution. The vertical axis is the number of patients, and the horizontal axis is the value of Rday_n.

https://doi.org/10.1371/journal.pone.0228508.g002

thumbnail
Fig 3. Box plots comparing Rday_n values between GVHD (+) and GVHD (–).

GVHD (+) and GVHD (-) indicate patients with and without aGVHD. A thick line in each box indicates the median value of Rday_n (n = 20,30,40 and 50).

https://doi.org/10.1371/journal.pone.0228508.g003

thumbnail
Fig 4. Box plots of Rday_n (n = 20, 30, 40, and 50) values per grade of aGVHD).

Rday_n (%) values (n = 20, 30, 40, and 50) in patients with aGVHD without (grade 0) and with (grade I-IV) aGVHD. There is no box plot for GradeⅣ, as it contained only one patient.

https://doi.org/10.1371/journal.pone.0228508.g004

Half-life of sDNAM-1

Based on our mathematical model, the half-life of sDNAM-1 was calculated as . The mean of half-life of sDNAM-1 was 12.5 +/- 9.2 days (n = 67). This calculation formula did not take the continuous production of sDNAM-1, which is variable in each patient, into account, resulting in the relatively high deviation of the half-life.

Days elapsed from HSCT to the development of aGVHD

We plotted a histogram of the days elapsed from allo-HSCT to the onset of aGVHD. The mean number of days was 23 (S1 Fig). Twenty-four patients (50.0%) developed aGVHD within 20 days after allo-HSCT, 39 (81.3%) within 30 days, 41 (85.4%) within 40 days, and 48 (100%) within 50 days.

Comparison between the models 1 and 2 using AIC

We compared 2 mathematical models to assess their goodness of fit to the data. The model 1 is consisted of two secretors of DNAM-1 (i.e., donor and recipient lymphocytes). On the other hand, the model 2 is consisted of three secretors of DNAM-1 (i.e., two types of donor lymphocytes that transiently and persistently release sDNAM-1 and recipient lymphocytes). The percentages of patients suitable for the models 1 and 2 is 29.9% and 70.1%, respectively (p-value = 0.0013) (Table 3). These results suggest that the model 2 consisting of three types of sDNAM-1 explained the dynamics of the observed sDNAM-1 better than did the model 1 consisting of two types of sDNAM-1.

thumbnail
Table 3. Comparison of data fitting between two models using Akaike’s information criterion.

https://doi.org/10.1371/journal.pone.0228508.t003

Simulation of dynamics of sDNAM-1 based on Rday_n

We simulated the dynamics of sDNAM-1 using the patients’ data (Fig 5). For example, patient ID3 who developed aGVHD (GVHD (+)) had high percentages of Rday_n (Rday_30 = 99.7%, Rday_40 = 98.8% and Rday_50 = 94.8%). Patient ID4 (GVHD (+)) also had high percentages of Rday_n (Rday_30 = 98.7%, Rday_40 = 78.3%, and Rday_50 = 61.0%). In contrast, patient ID2 who did not develop aGVHD (GVHD (–)) had a low concentration of total sDNAM-1 (maximum = 21.3 pM) and low percentages of Rday_n (Rday_30 = 44.3%, Rday_40 = 28.3%, and Rday_50 = 20.0%). Although the patient ID1 had a high concentration of total sDNAM-1 (maximum = 88.3 pM; see a highest purple dot), the patient did not develop aGVHD. Rather, the patient had low percentages of Rday_n (Rday_30 = 0.211%, Rday_40 = 7.91%, and Rday_50 = 19.4%).

thumbnail
Fig 5. Observed data and simulation of the dynamics of the soluble form of DNAM-1 (sDNAM-1) from different sources.

Representative patients Data of sDNAM-1 dynamics after transplantation are shown. The purple dots indicate the observed data. The blue line models the total amount of sDNAM-1; it represents the full model (= x1(t)+x2(t)+x3(t)). The green line models the first type of sDNAM-1, namely the transiently type (= x1(t)). The yellow-green line models the second type of sDNAM-1, namely the persistent type (= x2(t)). The red line models the third type of sDNAM-1, namely the residual type produced by recipient (= x3(t)). Thus, the blue line (total concentration of sDNAM-1) consists of the green, yellow-green, and red lines combined. Patients ID1 and ID2 did not develop aGVHD, whereas patients ID3 and ID4 did develop it.

https://doi.org/10.1371/journal.pone.0228508.g005

The AUC values of Rday_20, Rday_30, Rday_40, and Rday_50 were 0.77, 0.86, 0.85, and 0.82, respectively (Table 4). We set the cutoffs of Rday_n (n = 20, 30, 40, and 50) of 70%, 66%, 51%, and 37%, respectively based on the method of “The closest-to-(0,1) criterion” [15]. The respective sensitivities and specificities of Rday_20, Rday_30, Rday_40, and Rday_50 were 63%, 69%, 79%, and 85%, and 84%, 89%, 74%, and 68%, respectively. The accuracies were 69%, 75%, 78%, and 81%, respectively.

thumbnail
Table 4. Assessment of Rday_n by using receiver operating characteristics (ROCs).

https://doi.org/10.1371/journal.pone.0228508.t004

Discussion

The biomarkers for aGVHD have been identified and validated as promising tools for diagnosis, assessment, prediction of response to therapy, and prognostic risk [7,1820]. Several groups have also identified organ-specific biomarkers for aGVHD, such as elafin for skin aGVHD, regenerating islet-derived 3-alpha (Reg3α) for gastrointestinal tract aGVHD, and hepatocyte growth factor (HGF) and cytokeratin fragment 18 (KRT18) for liver aGVHD [7,2022]. In addition, soluble suppression of tumorigenicity 2 (ST2) and the plasma microRNA signature were identified as predictive biomarkers for resistance to systemic steroid therapy to aGVHD and survival of allo-HSCT patients with aGVHD [20,23].

In the current study, we constructed a mathematical model to assess the dynamics of sDNAM-1 in the sera of patients after allo-HSCT and applied it to the data set of sDNAM-1, which had been previously analyzed [7]. For a mathematical model, we assumed that there were three types of sDNAM-1. The first and second types of sDNAM-1 are released from alloreactive and non-alloreactive donor lymphocytes, respectively. The third type is residual one released from recipient lymphocytes before allo-HSCT. Based on these three types of sDNAM-1, we constructed a mathematical model for the dynamics of sDNAM-1 using either these three or two (the second and the third) types of sDNAM-1 and found that the model using the three types efficiently fitted into the data better than the model using two types of sDNAM-1, demonstrating that the first type of sDNAM-1 is required for the construction of a better mathematical model. By using this model, we showed that the greater the proportion of the first type of sDNAM-1 to the total of the first and second types of sDNAM-1 during n days (Rday_n) (n = 20, 30, 40, and 50) after allo-HSCT was, the more likely the patient developed aGVHD. Rday_n was a reliable index for high sensitivity of and specificity to the development of aGVHD. We also showed that Rday_n correlated well with the GVHD severity. Thus, Rday_n is a key biomarker for the development of aGVHD when sDNAM-1 was analyzed repeatedly over time after allo-HSCT.

Considering that allogeneic effector T cells derived from donors proliferate rapidly after transplantation and play a central role in the pathogenesis of aGVHD [24,25][26], donor lymphocytes producing the first type of sDNAM-1 are likely to be allogeneic effector T cells. On the other hand, donor lymphocytes that persistently release sDNAM-1 might be non-effector T cells. This scenario is in agreement with the previous report that certain T cells showed clonal expansions after allo-HSCT particularly in patients with aGVHD as demonstrated by the T cell antigen receptor repertory analyses [27].

Our study showed that our mathematical model for sDNAM-1 dynamics provides a useful biomarker for the development of aGVHD. However, a larger prospective study is required to generalize the significance of this mathematical model as an aGVHD biomarker. Nevertheless, our concept provides an important framework of a sensitive and specific biomarker for aGVHD.

Supporting information

S1 Fig. Histogram of onset day of aGVHD.

https://doi.org/10.1371/journal.pone.0228508.s001

(DOCX)

S2 Fig. Relation between Skin GVHD and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s002

(DOCX)

S3 Fig. Relation between gastrointestinal GVHD and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s003

(DOCX)

S4 Fig. Relation between liver GVHD and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s004

(DOCX)

S1 Table. Definitions of parameters used in the mathematical model.

https://doi.org/10.1371/journal.pone.0228508.s005

(DOCX)

S2 Table. Relation between skin GVHD and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s006

(DOCX)

S3 Table. Relation between gastrointestinal GVHD and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s007

(DOCX)

S4 Table. Relation between liver GVHD and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s008

(DOCX)

S6 Table. Relation between CMV infection and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s010

(DOCX)

S7 Table. Relation between GVHD prophylaxis and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s011

(DOCX)

S8 Table. Relation between HLA matching and Rday_n.

https://doi.org/10.1371/journal.pone.0228508.s012

(DOCX)

Acknowledgments

We thank S. Tochihara and W. Saito for secretarial assistance.

References

  1. 1. Korngold R, Sprent J. Variable capacity of L3T4+ T cells to cause lethal graft-versus-host disease across minor histocompatibility barriers in mice. J Exp Med [Internet]. 1987 Jun 1 [cited 2018 Nov 4];165(6):1552–64. Available from: http://www.ncbi.nlm.nih.gov/pubmed/3108446 pmid:3108446
  2. 2. Ferrara JLM, Levy R, Chao NJ. Pathophysiologic mechanisms of acute graft-vs.-host disease. Biol Blood Marrow Transplant [Internet]. 1999 Dec 1 [cited 2018 Nov 4];5(6):347–56. Available from: https://www.sciencedirect.com/science/article/pii/S108387919970011X?via%3Dihub pmid:10595812
  3. 3. Zhang L, Chu J, Yu J, Wei W. Cellular and molecular mechanisms in graft-versus-host disease. J Leukoc Biol [Internet]. 2016 Feb 1 [cited 2018 Nov 4];99(2):279–87. Available from: pmid:26643713
  4. 4. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, et al. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity [Internet]. 1996 Jun 1 [cited 2018 Jan 3];4(6):573–81. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8673704 pmid:8673704
  5. 5. Nabekura T, Shibuya K, Takenaka E, Kai H, Shibata K, Yamashita Y, et al. Critical role of DNAX accessory molecule-1 (DNAM-1) in the development of acute graft-versus-host disease in mice. Proc Natl Acad Sci U S A [Internet]. 2010 Oct 26 [cited 2017 Dec 18];107(43):18593–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20937876 pmid:20937876
  6. 6. Koyama M, Kuns RD, Olver SD, Lineburg KE, Lor M, Teal BE, et al. Promoting regulation via the inhibition of DNAM-1 after transplantation. Blood [Internet]. 2013 Apr 25 [cited 2018 Jan 3];121(17):3511–20. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9376604 pmid:23430112
  7. 7. Kanaya M, Shibuya K, Hirochika R, Kanemoto M, Ohashi K, Okada M, et al. Soluble DNAM-1, as a predictive biomarker for acute Graft-Versus-Host disease. PLoS One. 2016;11(6):1–12.
  8. 8. Glucksberg H, Storb R, Fefer A, Buckner CD, Neiman PE, Clift RA, et al. Clinical manifestations of graft-versus-host disease in human recipients of marrow from hl-a-matched sibling donors1. Transplantation. 1974;18(4):295–304. pmid:4153799
  9. 9. Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J, et al. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant [Internet]. 1995 Jun [cited 2018 Dec 4];15(6):825–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7581076 pmid:7581076
  10. 10. Nakaoka S, Aihara K. Mathematical study on kinetics of hematopoietic stem cells–theoretical conditions for successful transplantation. J Biol Dyn [Internet]. 2012 Mar [cited 2019 Feb 17];6(2):836–54. Available from: http://www.tandfonline.com/doi/abs/10.1080/17513758.2011.588343
  11. 11. Iwami S, Takeuchi JS, Nakaoka S, Mammano F, Clavel F, Inaba H, et al. Cell-to-cell infection by HIV contributes over half of virus infection. Elife [Internet]. 2015 Oct 6;4. Available from: https://elifesciences.org/articles/08150
  12. 12. Petzoldt T. Molecular epidemeology presentations 2018. 2010;33(3).
  13. 13. Akaike H. Information theory and an extension of the maximum likelihood principle. Csaki BNPF, editor. Budapest, Hungary CL - 2nd International Symposium on Information Theory, Tsahkadsor, Armenia, USSR, September 2–8, 1971: Akadémiai Kiadó; 1973. p. 267–81.
  14. 14. Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet [Internet]. 2009;373(9674):1550–61. Available from: pmid:19282026
  15. 15. Perkins NJ, Schisterman EF. The inconsistency of &quot;optimal&quot; cutpoints obtained using two criteria based on the receiver operating characteristic curve. Am J Epidemiol [Internet]. 2006 Apr 1 [cited 2018 Nov 25];163(7):670–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16410346 pmid:16410346
  16. 16. Paczesny S, Braun T, Vander Lugt M, Harris A, Fiema B, Hernandez J, et al. Three Biomarker Panel at Day 7 and 14 Can Predict Development of Grade II-IV Acute Graft-Versus-Host Disease. Blood [Internet]. 2010 [cited 2018 Nov 25];116(21). Available from: http://www.bloodjournal.org/content/116/21/675?sso-checked=true
  17. 17. Lee C, Haneuse S, Wang H-L, Rose S, Spellman SR, Verneris M, et al. Prediction of absolute risk of acute graft-versus-host disease following hematopoietic cell transplantation. PLoS One [Internet]. 2018 [cited 2018 Nov 1];13(1):e0190610. Available from: http://www.ncbi.nlm.nih.gov/pubmed/29346409 pmid:29346409
  18. 18. Harris AC, Ferrara JLM, Levine JE. Advances in predicting acute GVHD. Br J Haematol [Internet]. 2013 Feb [cited 2019 May 6];160(3):288–302. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23205489 pmid:23205489
  19. 19. Paczesny S, Krijanovski OI, Braun TM, Choi SW, Clouthier SG, Kuick R, et al. A biomarker panel for acute graft-versus-host disease. Blood [Internet]. 2009 Jan 8 [cited 2019 May 6];113(2):273–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8481507 pmid:18832652
  20. 20. Vander Lugt MT, Braun TM, Hanash S, Ritz J, Ho VT, Antin JH, et al. ST2 as a Marker for Risk of Therapy-Resistant Graft-versus-Host Disease and Death. N Engl J Med [Internet]. 2013 Aug 8 [cited 2018 Jan 7];369(6):529–39. Available from: http://www.nejm.org/doi/10.1056/NEJMoa1213299 pmid:23924003
  21. 21. Ferrara JLM, Harris AC, Greenson JK, Braun TM, Holler E, Teshima T, et al. Regenerating islet-derived 3-alpha is a biomarker of gastrointestinal graft-versus-host disease. Blood [Internet]. 2011 Dec 15 [cited 2019 May 6];118(25):6702–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21979939 pmid:21979939
  22. 22. Harris AC, Ferrara JLM, Braun TM, Holler E, Teshima T, Levine JE, et al. Plasma biomarkers of lower gastrointestinal and liver acute GVHD. Blood [Internet]. 2012 Mar 22 [cited 2019 May 6];119(12):2960–3. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22286196 pmid:22286196
  23. 23. Xiao B, Wang Y, Li W, Baker M, Guo J, Corbet K, et al. Plasma microRNA signature as a noninvasive biomarker for acute graft-versus-host disease. Blood [Internet]. 2013 Nov 7 [cited 2019 May 6];122(19):3365–75. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24041574 pmid:24041574
  24. 24. Matthews K, Lim Z, Afzali B, Pearce L, Abdallah A, Kordasti S, et al. Imbalance of effector and regulatory CD4 T cells is associated with graft-versus-host disease after hematopoietic stem cell transplantation using a reduced intensity conditioning regimen and alemtuzumab. Haematologica [Internet]. 2009 Jul [cited 2019 Apr 1];94(7):956–66. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19491336 pmid:19491336
  25. 25. Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Alloreactive memory T cells are responsible for the persistence of graft-versus-host disease. J Immunol [Internet]. 2005 Mar 1 [cited 2018 Jan 14];174(5):3051–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15728519 pmid:15728519
  26. 26. Alho AC, Kim HT, Chammas MJ, Reynolds CG, Matos TR, Forcade E, et al. Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD. 2016 [cited 2019 Apr 1]; Available from: http://www.broadinstitute.org/
  27. 27. Yew PY, Alachkar H, Yamaguchi R, Kiyotani K, Fang H, Yap KL, et al. Quantitative characterization of T-cell repertoire in allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant [Internet]. 2015 Sep 8 [cited 2019 Apr 18];50(9):1227–34. Available from: http://www.nature.com/articles/bmt2015133 pmid:26052909