Next Article in Journal
Short or Long Interval between Priming and Boosting: Does It Impact on the Vaccine Immunogenicity?
Next Article in Special Issue
Humoral Immunity to Varicella Zoster Virus in Patients with Systemic Lupus Erythematosus and Rheumatoid Arthritis Compared to Healthy Controls
Previous Article in Journal
Evaluation of Typhoid Conjugate Vaccine Effectiveness in Ghana (TyVEGHA) Using a Cluster-Randomized Controlled Phase IV Trial: Trial Design and Population Baseline Characteristics
Previous Article in Special Issue
Ageing of Immune System and Response to a Live-Attenuated Herpes Zoster Vaccine in Lung Transplant Candidates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Factors Associated with Post-Transplant Active Epstein-Barr Virus Infection and Lymphoproliferative Disease in Hematopoietic Stem Cell Transplant Recipients: A Systematic Review and Meta-Analysis

1
Department of Social and Preventive Medicine, Université de Montréal, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
2
Department of Pediatrics, Division of Infectious Diseases, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
3
Department of Pediatrics, Division of Hematology-Oncology, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
4
Research Center of CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
5
Department of Pediatrics, Division of Pediatric Intensive Care Medicine, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
6
Departement of Microbiology, Infectiology and Immunology, Université de Montréal, CHU Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
*
Author to whom correspondence should be addressed.
Vaccines 2021, 9(3), 288; https://doi.org/10.3390/vaccines9030288
Submission received: 25 January 2021 / Revised: 15 March 2021 / Accepted: 15 March 2021 / Published: 19 March 2021

Abstract

:
This systematic review was undertaken to identify risk factors associated with post-transplant Epstein–Barr virus (EBV) active infection and post-transplant lymphoproliferative disease (PTLD) in pediatric and adult recipients of hematopoietic stem cell transplants (HSCT). A literature search was conducted in PubMed and EMBASE to identify studies published until 30 June 2020. Descriptive information was extracted for each individual study, and data were compiled for individual risk factors, including, when possible, relative risks with 95% confidence intervals and/or p-values. Meta-analyses were planned when possible. The methodological quality and potential for bias of included studies were also evaluated. Of the 3362 titles retrieved, 77 were included (62 for EBV infection and 22 for PTLD). The overall quality of the studies was strong. Several risk factors were explored in these studies, but few statistically significant associations were identified. The use of anti-thymocyte globulin (ATG) was identified as the most important risk factor positively associated with post-transplant active EBV infection and with PTLD. The pooled relative risks obtained using the random-effect model were 5.26 (95% CI: 2.92–9.45) and 4.17 (95% CI: 2.61–6.68) for the association between ATG and post-transplant EBV infection and PTLD, respectively. Other risk factors for EBV and PTLD were found in the included studies, such as graft-versus-host disease, type of conditioning regimen or type of donor, but results are conflicting. In conclusion, the results of this systematic review indicate that ATG increases the risk of EBV infection and PTLD, but the link with all other factors is either nonexistent or much less convincing.

1. Introduction

Hematopoietic stem cell transplant (HSCT) recipients are at risk of developing post-transplant lymphoproliferative disease (PTLD) following primary or reactivated infection by the Epstein–Barr virus (EBV) [1,2,3,4,5,6,7]. EBV is a ubiquitous human herpesvirus with a seroprevalence approximating 50–55% of the pediatric population living in countries with high hygienic standards and reaching 90–99% by mid-adulthood [8,9,10]. EBV is the etiologic agent of infectious mononucleosis and is also associated with the development of some cancers, most notably Hodgkin’s lymphoma, African Burkitt’s lymphoma and nasopharyngeal carcinoma [11,12,13], as well as lymphoproliferative disease in immunocompromised individuals [14]. After primary infection, EBV establishes latent infection in B cells [15]. In immunocompetent individuals, primary infection is often subclinical and latent infection is usually well controlled by the immune system throughout life. However, when the cytolytic T-lymphocyte arm of the immune system is suppressed, primary infection can be more consequential, and latent EBV can reactivate to cause a spectrum of EBV-associated diseases ranging from fever, EBV end-organ diseases, such as pneumonia, hepatitis and encephalitis, to PTLD [16]. PTLD is a complex disorder whereby an interplay of factors is involved in facilitating tumorigenesis [17].
The occurrence of PTLD in patients receiving an allogeneic HSCT can reach 24%, depending on the presence of risk factors [16,18,19]. The highest incidence of PTLD is seen in the first six months post-transplant, with most cases occurring during the first year post-transplant [4,6,20]. Infants are generally at higher risk because they are most often EBV-naïve before transplant [21]. Several clinical risk factors have been associated with EBV infection and PTLD in HSCT, including T-cell depletion of the graft, use of unrelated donors or of two or more HLA-mismatches in related donors, use of anti-lymphocyte serum for prevention or treatment of acute graft-versus-host disease (GvHD) and use of anti-CD3 monoclonal antibodies for acute GvHD [19].
The rapid increase of EBV viral load (EBV-VL) in the blood is a well-documented predictive biomarker of EBV-associated diseases. Following transplantation, regular monitoring of EBV-VL is usually performed for better management of patients who show large spikes in VL [22,23]. A reduction in the intensity of immunosuppression or treatment with the anti-CD20 monoclonal (rituximab) is effective in decreasing EBV-VL to prevent PTLD [24]. In patients receiving HSCT, rituximab use is the more common option [7]. However, both methods for lowering EBV-VL have important disadvantages. Reduction in the intensity of immunosuppression can increase the risk of GvHD [6], while rituximab use in patients who are already immunosuppressed can incur the development of other fatal infections [6]. Rituximab targets CD20-expressing malignant B cells as well as all mature B cells, thus impeding the production of antimicrobial immunoglobulins [6]. There is clearly an important clinical advantage in preventing EBV disease rather than attempting to cure it. Numerous studies have sought to better understand the determinants of EBV infection following allogeneic HSCT. The literature contains numerous important studies that consider one or more risk factors in small to large sample sizes of patients with different characteristics; however, no systematic review is available summarizing the determinants of EBV infection in HSCT. Therefore, the aim of this work was to synthesize, through a systematic review and meta-analysis, the risk factors associated with active EBV infection and with PTLD in HSCT recipients.

2. Methods

We conducted, using Medline and EMBASE, a systematic search of all articles on risk factors for active EBV infection (including EBV primary infection as well as EBV reactivation) and PTLD in pediatric and adult recipients of HSCT published in peer-reviewed journals between 1946 and 30 June 2020. A non-exhaustive list of concepts and keywords was obtained by referring to articles related to active EBV infection and PTLD; the list was broadened using medical subject heading (MeSH) descriptors in Medline and Emtree in EMBASE. The ovidSP interface was used to search in both databases. The search equations are presented in Table S1. The selection of the articles was done in four steps: (1) title exploration, (2) review of abstracts, (3) review of the articles’ contents, and (4) review of the references of selected articles. All selection steps were performed independently by two authors (PE, MZ); in cases of disagreement, a third author (HT) was solicited for a consensual decision.
For the systematic review, three inclusion conditions were applied: (1) the study population had to be composed of pediatric and/or adult HSCT recipients; (2) risk factors for EBV infection or for PTLD had to be analyzed using univariate and/or multivariate statistical methods; and (3) the paper had to be in English or French. Abstracts, conference papers, congress papers, editorials, guidelines, reviews and case reports were excluded from the systematic review.
Two independent authors (PE and MZ) extracted the following information from the selected articles: authors, publication year, location, study type, post-transplant follow-up duration, transplant type, sample size, population (child or adult, and median or mean age, range or interquartile range), the definition of PTLD or definition of EBV infection, frequency of EBV-VL testing, blood compartment used to measure EBV-VL, and statistical methods used. In addition, for all potential risk factors explored in the studies, point estimates, such as odds ratio (OR), risk ratio (RR), the hazard ratio (HR) and subhazard ratio (SHR), confidence intervals (CI) and p-values were extracted when reported. In some cases, the corresponding author was contacted to clarify ambiguities.
The quality of each individual study was independently evaluated by two authors (PE & MZ) using a modified version of the Effective Public Health Practice Project (EPHPP) quality assessment tool for quantitative studies [25,26]. The quality assessment was based on the following components: selection bias, study design, confounders and data collection methods; it was rated as strong, moderate or weak (from high-quality to low-quality) according to the definition presented in Table S2a.
Finally, risk factors explored in these studies were described by providing the total number of studies showing a statistically significant association contrasted to the total number of studies investigating the risk factor.
The data reported made it possible to perform a meta-analysis solely to measure the association between the use of anti-thymocyte globulin (ATG) and two outcomes: post-transplant EBV infection and PTLD. Studies using multivariate analysis were considered for the meta-analysis except for the study by Liu et al. [27] because only the p-value was reported (not the measure of association). Since post-transplant EBV infection is not a rare event in this population, to obtain pooled estimates, results from studies that reported adjusted HR or SHR were combined separately from those that reported adjusted OR. This distinction was not made for PTLD, which is a relatively less frequent event. Adjusted estimates were combined using the inverse variance method with the fixed-effect model or random-effect model. The choice between these two models was guided by the value of statistic I, which revealed the proportion of the total variance observed due to a real difference in the measures of effects between studies. The fixed-effect model was used when I2 < 25% and the random-effect model when I2 ≥ 25% [28,29]. We also performed a sensitivity analysis to assess the contribution of each study to the pooled estimate. To this end, the pooled estimate was recalculated, each time excluding only one of the studies considered [30]. The analysis was performed with software R version 3.6.1.

3. Results

In total, 3362 titles were identified in the research bases, 1883 in EMBASE and 1479 in Medline. Once duplicates and papers with exclusion criteria were removed, 77 articles [4,16,23,24,27,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102] fulfilled the inclusion criteria for our systematic review (62 for EBV and 22 for PTLD). Among the 22 articles selected for PTLD, seven were also retained for EBV. Detailed information on the selection procedure is provided in the flow diagram (Figure 1). Among the 62 articles included to analyze risk factors for post-transplant EBV infection, two relate exactly to the same patient cohort (Bogunia-Kubik et al. [36] and Bogunia-Kubik et al. [35]) and 11 relate to non-disjoint samples (include some of the same patients) (Cesaro et al. [41] and Cesaro et al. [42]; Liu et al. [74] and Liu et al. [27]; Xuan et al. [99] and Liu et al. [74]; Liu et al. [75] and Liu et al. [73]; Wang et al. [97] and Ru et al. [86]; Zhou et al. [102] and Zhou et al. [101]). However, none of these studies but one [101] were excluded from the qualitative synthesis for duplication because the risk factors explored were different. The study by Zhou et al. [102] was excluded because all variables in this paper were explored using univariate analysis and were considered in the study by Zhou et al. [101] using multivariate analysis. With respect to the analysis of PTLD risk factors, the studies by Sundin et al. [103] and Omar et al. [81] were discarded because Uhlin et al. [94] explored the same factors and sample population as these two studies. Hoegh-Petersen et al. [60] and Kalra et al. [67] used non-disjoint samples. However, these two studies were retained in the review because the risk factors explored were not completely identical. For the same risk factors explored in both studies, those from Kalra et al. [67] were retained as the analyses were done on a larger sample. In addition, the Ali et al. [31] and Althubaiti et al. [32] studies were carried out with non-disjoint samples, but both were retained because different variables were explored.
Characteristics of the selected studies are described in Table 1; more details are provided in Table S3. Briefly, among the 77 studies, seven were conducted in France, seven in Italy, six in Poland, one in Belgium, three in Spain, six in the United Kingdom, one in Finland, five in the United States, three in Japan, 16 in China, two in Korea, one in Russia, five in Canada, three in Sweden, one in Turkey, three in Germany, two in the Netherlands, one in Greece, one in Portugal and three were multi-national. Twenty-three studies were prospective, 51 were retrospective, two were case–control studies and one was a randomized control trial. The sample size ranged from 26 to 64,539 HSCT recipients (Figure 2). It is noteworthy that most studies were performed with pooled pediatric and adult populations (n = 41), while 19 included only children and 17 only adults.
The definition of post-transplant EBV infection and the diagnostic criteria for PTLD differed among studies. Active post-transplant EBV infection was diagnosed when the EBV-VL in blood, determined using a PCR test, was above a given threshold. In some cases, thresholds were not readily comparable because there was no direct conversion between the units of measurement used. In two studies [45,91], active EBV infection was defined as a reactivation event because all patients showed positive EBV serology when the follow-up period started. In other studies, no distinction was made between primary infection and reactivation: both were considered active EBV infection. There was also some variability between studies with respect to the frequency of PCR testing, but it was performed weekly in most studies during the early post-transplant period. The type of specimen tested by PCR varied, with peripheral blood in 19 studies, plasma in 11 studies, serum in five studies, serum or plasma in one study, whole blood in 13 studies, peripheral blood or whole blood in two studies, and whole blood and plasma in one study; 10 studies provided no information on specimen type. The method used to diagnose PTLD was not detailed in one study. The length of follow-up was an important source of variation between studies; in some cases the follow-up period was not reported [31,32,34,40,46,48,52,55,59,61,63,73,81,83,86,90,94,98,100].
Various statistical methods were used. Logistic regression was used in 13 studies, Cox model in 22, survival analysis using the log-rank test in one, multiple linear regression in two, Fine and Gray competitive risk model in 10 and Poisson regression for grouped survival data in one. Among the 28 studies employing univariate analysis, the statistical method was not explicitly reported in one study; one study used univariate logistic regression, another used the univariate Cox model, another used time-dependent landmark, while other studies used at least one of the following tests: Log-rank test, Gray’s test, Chi 2 test, Wilcoxon nonparametric test, Kruskal–Wallis test, Fisher’s exact test, Mann–Whitney test or Wald test. Among the 49 studies in which a multivariate analysis was performed, the criteria for selecting variables for the multivariate model were explicitly indicated in 20. Variables with a p-value ˂ 0.1 in univariate analysis were retained for multivariate analysis in eight studies, a p-value ˂ 0.2 in three, a p-value ˂ 0.3 in one, a p-value ˂ 0.05 in one; a p-value < 0.05 was used for the multivariate analysis in three studies. In four studies, the investigators used a p-value < 0.1 in the univariate analysis combined with a p-value < 0.05 in multivariate analysis. Altogether, 52 studies were considered as properly adjusted for confounding bias.
Table S2b reports the quality assessment of the 74 articles included in the review according to the outcome. Regarding post-transplant EBV infection, 27 (42.9%) were rated “strong”, 15 (23.8%) “moderate” and 21 (33.3%) “weak”. For PTLD, 12 (57.1%) were rated “strong”, three (14.3%) “moderate” and six (28.6%) “weak”. The lack of information on retention and potential for selection bias, as well as the absence of control for potential confounding bias, were the main contributors to the lower overall rating of articles. It is important to note that the absence of control for confusion in several articles could be justified by the fact that their main focus did not involve analysis of factors associated with either post-transplant EBV infection or PTLD.
Table S4 provides a detailed list of the risk factors for post-transplant EBV infection and for PTLD explored in the 77 included studies with a description, when possible, of the measures of association and CI and/or p-value. Figure 3 presents, for every individual risk factor, the total number of studies that investigated the risk factor contrasted to the number that showed a statistically significant association. Table 2 presents a summary of the risk factors (with measures of association) for post-transplant EBV infection and for PTLD that were analyzed in studies using multivariate analysis. The presence of GvHD, use of ATG and type of conditioning regimen were the three risk factors most frequently associated with EBV infection and PTLD.

3.1. Graft-versus-Host Disease

The association between acute (a)GvHD and post-transplant EBV infection was examined in 21 studies [4,39,41,46,51,52,56,57,59,65,66,68,74,76,81,85,86,88,89,91,101]. Six statistically significant associations were highlighted using different grade categorization of the outcome. In two studies, aGvHD was dichotomized in grade ≥3 versus <3; one of them (Juvonen et al. [66]) showed that patients with aGvHD grade ≥3 had a higher risk of active EBV infection (HR = 1.70 (95% CI: 1.11–2.62)). However, this result was not corroborated by other studies [51,91,101]. Among eight studies [4,39,41,57,59,74,86,89] that compared grade ≥2 versus <2, two [59,89] showed that an aGvHD grade ≥2 significantly increased the risk of active EBV infection, one reported a positive association without reporting the relative risk (Hiwarkar et al. [58]), and the other (Sirvent-Von Bueltzingsloewen et al. [89]) reported an OR = 3.4 (95% CI: 1.2–9.7). The potential effect of aGvHD, however, was not confirmed by the other studies [4,39,41,57,74]. One study (Peric et al. [85]) categorized aGvHD according to grade 0–1, grade 2 and grades 3–4 and did not show a statistically significant association with active EBV infection. Seven studies considered the presence versus absence of aGvHD; three highlighted a statistically significant association. Elmahdi et al. [52] showed that the presence of aGvHD, whatever the grade, increased the risk of EBV infection (HR = 3.29 (95% CI: 1.26–8.58)). Similar results were obtained by Cohen et al. [46] (OR = 2.2 (95% CI: 2.12–15.08)) and by Omar et al. [81] who showed that patients with aGvHD had on average a higher EBV-VL than patients without aGvHD (p = 0.009). However, these results were not corroborated in four other studies [56,65,68,76]. Seven studies examined if chronic (c) GvHD was a risk factor of post-transplant EBV infection [4,41,46,68,74,86,88], two of which showed a statistically significant relationship [68,86]. Two studies [24,69] did not differentiate aGvHD and cGvHD; one [69] showed a statistically significant association.
In regards to PTLD, its occurrence was associated with aGvHD grade ≥2 in Landgren et al. (RR = 1.7 [1.2–2.5)) [70], Uhlin et al. (SHR = 2.65 (1.32–5.35)) [94] and Fujimoto et al. (HR = 1.93 (1.48–2.52)) [55]. No statistically significant association was identified in eight other studies [16,27,46,56,57,88,95,98] that explored the association between aGvHD and PTLD. Four studies [16,27,70,88] analyzed the association between cGvHD and PTLD; only Landgren et al. [70] found a statistically significant association (RR = 2.0 [1.1–3.2)). In contrast, the study by Kalra et al. [67]. showed a lower risk of PTLD in patients with aGvHD grade ≥2 or cGvHD that required systemic therapy (SHR = 0.47; p = 0.04).

3.2. Graft-versus-Host Disease Prophylaxis/Treatment

ATG use appears to be an important risk factor for the development of active post-transplant EBV infection or PTLD. Among 15 multivariate studies [4,41,45,46,51,53,56,66,68,69,74,79,85,86,96] that examined the association between ATG and active post-transplant EBV infection, 10 found a statistically significant association: Cesaro et al. [41] (HR = 13.0 (95% CI: 2–96)), Fan et al. [53] (OR = 7.69 (95% CI: 1.17–50.49)), Juvonen et al. [66] (HR = 5.78 (95% CI: 2.47–13.5)), Liu et al. [74] (HR = 14.081 (95% CI: 6.02–32.92)), Peric et al. [85] (SHR = 4.9 (95% CI: 1.1–21.0)), Van Esser et al. [96] (HR = 3.4 (95% CI: 1.6–1)), Gao et al. [56] (HR = 6.3 (95% CI: 1.6–24.0)), Düver et al. [51] (OR = 10.68 (95% CI: 1.15–98.86)), Ru et al. [86] (HR = 4.29 (95% CI: 2.64–6.97)) and Kullberg-Lindh et al. [68] (slope = 1.34; p = 0.004). All studies compared patients who received ATG versus those who did not, but one: Van Esser et al. [96] reported the risk of EBV infection in patients receiving T-cell depleted (TCD) grafts with ATG versus patients receiving non-TCD grafts. A statistically significant association between active post-transplant EBV infection and TCD grafts was shown by Bordon et al. [37] (p = 0.04) as well as for CD4+ depleted grafts by Torre-Cisneros et al. [91] (OR = 11.5 (95% CI: 5.8–22.8)). Corticosteroid use for GvHD prophylaxis (OR = 23.68 (95% CI: 1.92–291.45)) was associated with EBV infection in the study by Fan et al. [53].
An association between ATG and PTLD was reported by Landgren et al. [70] (RR = 3.8 [2.5–5.8)), Van der Velden et al. [95] (OR = 2.4 (1.3–4.2)), Liu et al. [27] (p = 0.038), Xuan et al. [16] (HR = 13.03 (1.67–101.58)) as well by Fujimoto et al. [55] (HR = 6.13 (95% CI: 4.33–8.68] for GvHD prophylaxis and HR = 2.09 (95% CI: 1.17–3.72] for GvHD treatment). The association was not statistically significant in the study by Gao et al. [56] (HR = 2.9 (95% CI: 0.3–27.5)). Brunstein et al. [38] found a higher risk for the composite outcome ‘post-transplant EBV infection or PTLD’ in patients with non-myeloablative conditioning regimen (NMAC) + ATG (HR = 15.4 (2.0–116.1)), but a similar risk for those receiving NMAC without ATG (HR = 0.7 (0.1–6.5)) compared to those who received myeloablative conditioning (MAC). This highlights the role of ATG as a significant risk factor. Buyck et al. [40] reported a dose-response relationship: the risk of PTLD increased with the number of prior courses of ATG (HR = 7.23 (1.67–31.32)). Lin et al. [72] found a higher risk of post-transplant EBV infection in patients, who received a higher dose of ATG (10.0 mg/kg versus 7.5 mg/kg: HR = 2.02 (95% CI: 1.37–2.97)). The study by Cohen et al. [46] compared patients who received Campath versus ATG; no statistically significant association was found (unadjusted OR = 0.56 (0.15–2.05)).
The meta-analyses that we performed are presented in Figure 4 and Figure 5. The pooled HR for the association between ATG use and post-transplant EBV infection obtained using the random-effect model was 5.26 (95% CI: 2.92–9.45) with an I2 = 63.2% (Figure 4). We performed sensitivity analyses by recalculating the pooled estimate after excluding only one study at a time: the results vary between 4.13 and 6.49, and the I2 heterogeneity statistic varies between 22% and 69%. The studies by Laberko et al. [69] and Liu et al. [74] had the greatest influence on the pooled estimate and on the level of heterogeneity. However, regardless of the study excluded, the overall result remains statistically significant. With respect to studies that estimated an adjusted OR to report the association between ATG and post-transplant EBV infection, the pooled estimate was 2.74 [1.03–7.31] and I2 = 40.3% (Figure 5). The sensitivity analyses highlighted a variation of the pooled estimate from 2.07 to 4.00 and of I2 from 28% to 58%. The studies by Christopeit et al. [45] and Cohen et al. [46] had the greatest influence on the pooled estimate and heterogeneity. The pooled estimate was no longer significant if a single study was removed from the analysis, except for the study by Christopeit et al. [45], which was carried out with the smallest sample. The pooled RR for the association between ATG and PTLD obtained using the random-effect model was 4.17 (95% CI: 2.61–6.68) with an I2 = 56.7%. The sensitivity analysis revealed that the pooled estimate ranged from 3.34 to 5.02 and the I2 from 9% to 67%. The studies by Fujimoto et al. [55] and Van der Velden et al. [95] had the biggest influence on the pooled estimate and the I2. The sensitivity analysis did not question the statistically significant association between ATG and PTLD.
The results of these meta-analyses should be understood cautiously given the high-level of heterogeneity observed between studies. Due to the small number of articles, we did not explore the sources of heterogeneity further by performing a subgroup analysis or a meta-regression.

3.3. Other Risk Factors

Other possible risk factors were analyzed in the retained studies; these factors were not associated with EBV infection or PTLD, or their relationship was more ambiguous (Table 2 and Table S4). The association between the primary diagnosis and post-transplant active EBV infection was explored in several reports [4,23,35,39,46,56,59,69,85,86,88,101]; three showed a strong positive and statistically significant association [23,39,88]. According to Carpenter et al. [23], the risk of active EBV infection was greater in patients with Hodgkin’s lymphoma (HR = 3.53 (95% CI: 1.51–8.25)) or chronic lymphocytic leukemia (HR = 3.77 (95% CI: 1.38–10.32)) compared to patients with acute myeloid leukemia. Sanz et al. [88] reported that the risk was greater in patients with Hodgkin disease compared to other patients (SHR = 11.6 (95% CI: 3.4–40.0)). However, Burns et al. [39] found that the risk of active post-transplant EBV infection was lower in patients with non-Hodgkin’s lymphoma compared to patients with acute myeloid leukemia/myelodysplastic syndrome (AML/MDS) (HR = 0.18 (95% CI: 0.05–0.57)). Furthermore, Fujimoto et al. [55] showed a higher risk of PTLD in patients with aplastic anemia compared to those with AML/MDS (HR = 5.19 (95% CI: 3.32–8.11)). No statistically significant association between the PTLD outcome and the patient’s primary diagnosis was found in the other studies [46,56,88,94,95,98].
The use of reduced-intensity conditioning regimen was determined as a risk factor for post-transplant EBV infection by Sanz et al. [88] (SHR = 6.0 (2.0–17.6)) and as a risk factor for PTLD by Sanz et al. [88] (SHR = 5.5 (1.8–17.1)) and Uhlin et al. [94] (SHR = 3.25 (1.53–6.89)). Two studies reported that intensified myeloablative conditioning regimen (MAC) increased the risk of post-transplant EBV infection: Liu et al. [74] (HR = 1.72 (1.03–2.88)) and Lin et al. [72] (HR = 1.73 (95% CI: 1.18–2.54)). Liu et al. [74] also found an association between intensified MAC and PTLD (p = 0.018), but Xuan et al. [16] found an association in the opposite direction (standard versus intensified regimen: HR = 4.46 (1.20–16.61)). Gao et al. [56] found that the use of fludarabine would increase the risk of PTLD (HR = 3.8 (1.4–10.6)). EBV viral load was higher in the study by Kullberg-Lindh et al. [68] when total body irradiation (TBI) was performed.
Recipient age did not seem to be an important risk factor for active EBV infection post-transplant. Of the 20 studies [4,23,35,39,50,51,52,56,65,68,69,74,76,85,86,88,89,93,96,101] to consider this factor, only Bogunia-Kubik et al. [35] highlighted a statistically significant association, showing that the propensity for active post-transplant EBV infection was higher in people over 25 years compared to others (OR = 1.54 (95% CI: 1.14–2.70)). Concerning the association between age and PTLD, only two [56,70] of the 10 studies [16,27,40,56,67,70,88,94,95,98] that explored this factor indicated a statistically significant association. A higher risk of PTLD has been observed in patients aged 50 years or more (RR = 5.1 (2.8–8.7)) [70]. Conversely, a lower risk of PTLD was observed in patients 40 years or older in another study (HR = 0.4 (0.2–0.9)) [56].
Several studies used multivariate analysis to examine the relationship between recipient sex and post-transplant active EBV infection [4,35,39,50,52,56,65,68,69,72,74,76,85,86,88,89,96,101] or PTLD [16,27,40,56,88,94,95,98]; none found a significant association. Three out of six studies [35,53,56,65,79,101] that analyzed the association between donor sex and post-HSCT active EBV infection showed a statistically significant association but in the opposite direction. In two studies, the risk for active EBV infection post-HSCT was higher in patients receiving a male donor transplant [53,56] while, in the other, patients receiving a female donor transplant appeared to be at greater risk [65]. The only study [56] that explored the association between donor sex and PTLD did not find a statistically significant association. Moreover, no statistically significant association was found between the donor/recipient sex combination and post-transplant EBV infection [35,41,53,57,65]. Among all studies that examined the sex of the dyad donor/recipient and PTLD [57,82,94,98], only one [82] found a statistically significant association suggesting a higher risk of PTLD in patients who received a transplant from a different sex donor.
With regard to viral infections, a study by Zallio et al. [24] suggests that the risk of active post-transplant EBV infection is higher in patients with CMV reactivation compared to those who are CMV negative (p < 0.05). Similar results were found by Gao et al. (HR = 5.9 (2.5–13.9)) [56] and by Zhou et al. (HR = 97.75 (9.48–1008.30)) [101]. Xu et al. [98] and Gao et al. [56] found that the risk of PTLD was higher in patients with CMV DNAemia: HR = 5.68 (1.17–7.57) and HR = 11.6 (1.2–114.4), respectively. According to Lakerko et al. [69], the risk of post-transplant EBV active infection was higher among EBV seronegative patients (compared to seropositive patients) in HSCT recipients receiving a graft from an EBV-seropositive donor (HR = 2.85 (95% CI: 1.12–7.28)). Lin et al. [72] found a higher risk of post-transplant EBV infection in EBV-seropositive patients who received an EBV-negative graft (HR = 1.58 (1.01–2.46)). Other risk factors associated with active EBV infection post-transplant include: (1) two human genotypes, namely the interferon-ɣ (IFNɣ) gene 3/3 (OR = 7.28; p = 0.005) [36] and the CC-chemokine receptor-5 (CCR5) (OR = 0.17 (95% CI: 0.03–0.80)) [35], (2) the volume of platelets transfused (>2530 vs. <1260 mL) (HR = 2.19 (95% CI: 1.21–3.97)) [92], (3) unrelated or mismatched related donor (p = 0.04) [81], (4) unrelated donor ((HR = 8.8, p = 0.030) [76], (OR = 5.05 (1.24–20.63)) [51], (HR = 2.63 (1.02–6.67)) [93]), (5) HLA incompatibility ((OR = 5 [1.5–16.4)) [89], (HR = 1.83 (1.27–2.63)) [86]), (6) CD3+ count in the graft ≥ median (OR = 0.11 (0.02–0.78)) [4], (7) CD3+CD8+ count in the graft ≥ median (OR = 0.05 (0.01–0.43)) [45], (8) CD34+ count in the graft >1.35 × 106/kg (HR = 2.6 (1.5–4.6)) [96], (9) CD4+ lymphocyte/µl at one month after HSCT ≥ 50 (OR = 0.1 (0.02–0.481)) [4], (10) Vδ2+ T cell count 30 days post-transplant (HR = 0.347 (0.161–0.747)) [73], (11) IgM level ≥ median 30 days after HSCT (HR = 0.27 (0.10–0.75)) [98], (12) proportion (%) of NKp30/total NK cells one month after HSCT (HR = 0.96 (0.918–0.998)) [100], and (13) prior HSCT (HR = 2.6 (1.1–6.4)) [57]. Also, a higher risk of post-transplant EBV infection was observed by Tsoumakas et al. [93] in patients receiving a peripheral blood transplant compared to those receiving a bone marrow transplant (HR = 2.51 (1.04–6.05)), but an opposite result was found by Wang et al. [97] (HR = 18.69; p < 0.001).
Other factors associated with PTLD include: (1) CD8+ count (≥median vs. <median) 30 days after HSCT (HR = 0.34 (0.13–0.92)), (2) prior HSCT ((HR = 2.6 (1.1–6.4)) [57], (RR = 3.5 [1.7–6.3)) [70]), (3) splenectomy (SHR = 4.81 (1.51–15.4)) [94], (4) infusion of mesenchymal stromal cells (SHR = 3.05 (1.25–7.48)) [94], (5) a stepwise increase of EBV-DNA by 1 log (HR = 2.9 (1.7–4.8)) [96], (6) HLA DRB1*11:01 (SHR = 4.85 (1.57–14.97)) [82], and (7) HLA mismatch (SHR = 5.89 (2.43–14.3)) [94]. Fujimoto et al. [55] found that, compared to matched related donor grafts, the risk of PTLD is higher when using mismatched related donor grafts (HR = 4.39 (2.39–8.07)), matched unrelated donor grafts (HR = 4.08 (2.39–6.99)), mismatched unrelated donor grafts (HR = 3.20 (1.58–6.47)) or cord blood grafts (HR = 8.03 (4.72–13.7)).

4. Discussion

This systematic review includes 77 papers. It aims to characterize risk factors associated with active post-transplant EBV infection and PTLD in HSCT recipients. Active EBV infection can result in rapidly increasing EBV-VL, which is a high-risk marker for PTLD development. Proper identification of the risk factors associated with active EBV infection and PTLD is needed for effective patient management.
In this systematic review, we focused on risk factors explored in published studies; very few statistically significant associations were found. The use of ATG was identified as one of the most important risk factors for the development of active post-transplant EBV infection and PTLD. The pooled relative risks estimated from the meta-analysis that was carried out confirmed a positive and statistically significant association between ATG and EBV infection (RR = 3.98 (95% CI: 2.20–7.18) and PTLD (RR = 3.69 (95% CI: 2.24–6.08)). ATG is a potent immunosuppressive agent that obliterates the T-cell pool [104,105,106], thereby enabling reactivation of latent EBV contained in mature B cells along with the malignant expansion of infected cells [104]. In the HSCT setting, ATG is used for the prevention of aGvHD, given its ability to target and deplete T lymphocytes [107,108].
Some studies included in this review also found an association with the presence of GvHD, which is an immune-mediated complication of HSCT whereby donor T cells present in the graft initiate an alloreactive process that ultimately causes destruction of host tissues [109]. aGvHD usually occurs within the first three months post-transplant and is categorized into four grades ranging from 1 (light disease) to 4 (severe disease) [110]. cGvHD usually occurs beyond the initial three months post-transplant. The pathophysiology of GvHD, especially that of cGvHD, is complex [111]. T and B lymphocytes are probably involved in the pathophysiology of GvHD, although the mechanism linking these cells to GvHD is not well-known [108]. In short, the etiology of GvHD is complex, and it is difficult to conclude whether GvHD is an independent risk factor for EBV and PTLD or whether the relationship found in some studies is the result of confounding by indication related to the use of ATG. The analyses that we are currently running among pediatric HSCT recipients recruited in our TREASuRE cohort study [112] confirm that EBV is strongly associated with ATG but not with GvHD, following adequate control for confounding bias.
Many other variables were analyzed in the 77 included studies, but results were either inconsistent, failed to find an association, or limited in terms of the number of studies that investigated the risk factor. Some studies showed that primary diagnosis was associated with post-transplant active EBV infection [23,39,88], more specifically in the case of Hodgkin disease [23,88]. Some forms of Hodgkin’s lymphoma are etiologically linked to EBV [113,114] and may occur in individuals who are not able to properly control EBV infection. These individuals may be thought to be more susceptible to other EBV diseases (such as post-transplant active EBV infection) along the continuum of care, but HSCT should have corrected any immune cell problem. Although interesting, further studies are needed to confirm the potential association between Hodgkin’s disease and post-transplant active EBV infection in HSCT patients.
Discordant results were found for other variables, and, in other cases, the number of studies investigating risk factors was limited. These variables are recipient age, recipient gender, donor type, conditioning regimen, graft source, graft history, graft content (CD34+, CD3+, CD8+, CD3+/CD8+), genotype (IFNɣ gene 3/3, CCR5), splenectomy, mesenchymal stromal cells, donor gender and transfusion (red blood cells, platelets, plasma) (Table S4). In our recent study, although no relationship was statistically found between EBV and blood product transfusion, we linked a case of EBV infection in an EBV-seronegative pediatric HSCT recipient to a blood donor through viral genotype analysis [112]. One cause of discordant results is the heterogeneity observed among the various studies, most notably with regard to the different specimen types used to perform PCR tests (Table S3). The sensitivity of PCR tests is greater when whole blood is used as opposed to plasma [115]. Other sources of discordance include variations in the statistical approach and experimental design. We also noted the absence of controls for confounding and failure to report results when associations lacked statistical significance. In addition, only 42.9% of studies included in the systematic review of factors associated with post-transplant EBV infection were classified as being of strong quality, and 23.8% were classified as moderate quality; with respect to PTLD as an outcome, the proportions were, respectively 57.1% and 14.3%. An important risk for bias includes uncontrolled confounding bias and the lack of information on retention, a potential source of selection bias in cohort studies.
This review was not able to discern whether differences exist between children and adults. While statistical power was higher in studies combining both groups, differences in terms of risk factors may exist. Immune restoration through T-cell reconstitution after transplantation is different in children and adults [116], and risk factors may differ. It should be noted that 25 of the 77 studies selected in this systematic review have a sample size of less than 100; therefore, it is possible that type II error may explain why positive associations were not statistically significant in many studies. Moreover, the included studies were limited to the identification of factors associated with the first occurrence of active EBV infection post-transplant, although during follow-up a patient may experience several episodes of active EBV infection [23,116]. This latter aspect should be considered in order to better understand the dynamics of the evolution of active EBV infection post-transplant in HSCT recipients. Risk factors for the occurrence of active EBV infection may be different from those that explain the dynamics of infection. Finally, there was insufficient information on attrition, which may be the primary source of selection bias in this type of study. While we initially intended to perform a meta-analysis of all risk factors associated with active EBV infection and PTLD, this was not possible because of the diversity of outcome definitions, the variability in the definition of risk factors and the non-systematic reporting of point estimates, confidence intervals and p-values. However, as indicated above, a meta-analysis was carried out to measure the association between ATG use and post-transplant EBV infection and PTLD, respectively. The results, however, must be considered with caution, as the definition of outcome was quite variable from one study to another. Based on all the above arguments, further studies using large cohorts of children and adults are needed to better elucidate the determinants of active EBV infection and PTLD among HSCT recipients.
In conclusion, we found ATG as the most important risk factor for the development of active post-transplant EBV infection and PTLD in HSCT patients. ATG considerably increases the risk of EBV and PTLD. Other risk factors have been linked with EBV and PTLD in studies, such as GvHD or type of donor, but the association for these other factors is less clear due to conflicting results, the potential for bias, particularly confounding, or because of the low number of studies that considered these risk factors. Further studies using large cohorts of children and adults with appropriate control for confounding are needed to better characterize other determinants of active EBV infection and PTLD among HSCT recipients.

Supplementary Materials

The following are available online at https://www.mdpi.com/2076-393X/9/3/288/s1, Table S1: Search equation used in Medline and EMBASE, Table S2a: Component ratings of study (a modified version of the Effective Public Health Practice Project (EPHPP) Quality Assessment Tool for Quantitative Studies, Table S2b: Results of the quality evaluation of the 77 articles included in this systematic review, Table S3: Characteristics of the 77 studies included in the systematic review and Table S4: Risk factors for post-transplant EBV infection and for PTLD explored in the 77 retained studies.

Author Contributions

P.R.E.B.: developed the study protocol, performed the research and the selection of articles, carried out the data extraction and synthesis, elaborated the draft of the evaluation grid of selected articles, evaluated the quality of the articles, wrote the first draft of the manuscript and performed updates of the manuscript. M.Z.: conducted the research and selection of articles, performed the data extraction, revised the evaluation grid of the articles, assessed the quality of the articles and revised all versions of the manuscript. C.B., M.D., J.L.: provided clinical expertise for analysis and interpretation and revised the versions of the manuscript. L.L.: checked all the data extracted and proofread all versions of the manuscript. C.A.: provided expertise on EBV, revised all versions of the manuscript and co-supervised all steps. H.T.: validated the results of article selection, revised and validated the evaluation grid of articles, revised all versions of the manuscript and supervised all steps. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant from the Canadian Blood Services (grant CBS #201209–293922) to HT, CB, MD, JL, CA, as well as by an operating grant from the Cancer Research Society and the C17 Council (OG #23276) to CA for review of the pediatric studies. PREB was supported by doctoral awards from the CHU Sainte-Justine and Université de Montréal (tuition fee waiver scholarships for international students and graduate scholarship for the final year of PhD studies). HT holds a salary award (Chercheur–Boursier, Junior-2) from the Fonds de la recherche du Québec en santé (FRQ-S) and from Canadian Institutes of Health Research (CIHR) (New investigator salary award).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable (we did a systematic review and meta-analysis of published studies).

Data Availability

We confirm that our data are available.

Acknowledgments

We thank Sylvie Fontaine, librarian at the paramedical library of the Université de Montréal, for her support in the identification of descriptors and keywords in the Medline and EMBASE databases.

Conflicts of Interest

The authors of this manuscript have no conflicts of interest to disclose.

Abbreviations

aGvHDAcute graft-versus-host disease
ATGanti-thymocyte globulin
cGvHDchronic graft-versus-host disease
CCR5CC chemokine receptor-5
CIconfidence interval
CMVcytomegalovirus
EPHPPEffective Public Health Practice Project
EBVEpstein–Barr Virus
HRthe hazard ratio
HSCThemamiddleoietic stem cell transplant
IFNɣinterferon-ɣ
ORodds ratio
PCRpolymerase chain reaction
PTLDpost-transplant lymphoproliferative disease
SHRsubhazard ratio
TCDT-cell depleted
TBItotal body irradiation
VLviral load

References

  1. Aalto, S.M.; Juvonen, E.; Tarkkanen, J.; Volin, L.; Haario, H.; Ruutu, T.; Hedman, K. Epstein-Barr viral load and disease prediction in a large cohort of allogeneic stem cell transplant recipients. Clin. Infect. Dis. 2007, 45, 1305–1309. [Google Scholar] [CrossRef] [Green Version]
  2. Faraci, M.; Caviglia, I.; Morreale, G.; Lanino, E.; Cuzzubbo, D.; Giardino, S.; Di Marco, E.; Cirillo, C.; Scuderi, F.; Dallorso, S.; et al. Viral-load and B-lymphocyte monitoring of EBV reactivation after allogeneic hemopoietic SCT in children. Bone Marrow Transplant. 2010, 45, 1052–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Herreman, A.; Dierickx, D.; Morscio, J.; Camps, J.; Bittoun, E.; Verhoef, G.; De Wolf-Peeters, C.; Sagaert, X.; Tousseyn, T. Clinicopathological characteristics of posttransplant lymphoproliferative disorders of T-cell origin: Single-center series of nine cases and meta-analysis of 147 reported cases. Leuk. Lymphoma 2013, 54, 2190–2199. [Google Scholar] [CrossRef] [PubMed]
  4. Patriarca, F.; Medeot, M.; Isola, M.; Battista, M.L.; Sperotto, A.; Pipan, C.; Toffoletti, E.; Dozzo, M.; Michelutti, A.; Gregoraci, G.; et al. Prognostic factors and outcome of Epstein-Barr virus DNAemia in high-risk recipients of allogeneic stem cell transplantation treated with preemptive rituximab. Transpl. Infect. Dis. 2013, 15, 259–267. [Google Scholar] [CrossRef]
  5. Rasche, L.; Kapp, M.; Einsele, H.; Mielke, S. EBV-induced post transplant lymphoproliferative disorders: A persisting challenge in allogeneic hematopoetic SCT. Bone Marrow Transplant. 2014, 49, 163–167. [Google Scholar] [CrossRef] [Green Version]
  6. Reddy, N.; Rezvani, K.; Barrett, A.J.; Savani, B.N. Strategies to prevent EBV reactivation and posttransplant lymphoproliferative disorders (PTLD) after allogeneic stem cell transplantation in high-risk patients. Biol. Blood Marrow Transplant. 2011, 17, 591–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Styczynski, J.; Einsele, H.; Gil, L.; Ljungman, P. Outcome of treatment of Epstein-Barr virus-related post-transplant lymphoproliferative disorder in hematopoietic stem cell recipients: A comprehensive review of reported cases. Transpl. Infect. Dis. 2009, 11, 383–392. [Google Scholar] [CrossRef] [PubMed]
  8. International Agency for Research on Cancer (IARC). Epstein-Barr Virus and Kaposi’s Sarcoma Herpesvirus/Human Herpesvirus 8. In Proceedings of the IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, Lyon, France, 17–24 June 1997; Volume 70, pp. 1–492. [Google Scholar]
  9. Allen, U.; Alfieri, C.; Preiksaitis, J.; Humar, A.; Moore, D.; Tapiero, B.; Tellier, R.; Green, M.; Davies, D.; Hebert, D.; et al. Epstein-Barr virus infection in transplant recipients: Summary of a workshop on surveillance, prevention and treatment. Can. J. Infect. Dis. 2002, 13, 89–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Pariente, M.; Bartolome, J.; Lorente, S.; Crespo, M.D. Age distribution of serological profiles of Epstein-Barr virus infection: Review of results from a diagnostic laboratory. Enferm. Infecc. Microbiol. Clin. 2007, 25, 108–110. [Google Scholar] [CrossRef]
  11. Corssmit, E.P.; Hall, M.A.L.-V.; Portegies, P.; Bakker, P. Severe neurological complications in association with Epstein-Barr virus infection. J. Neurovirol. 1997, 3, 460–464. [Google Scholar] [CrossRef]
  12. Jenson, H.B. Acute complications of Epstein-Barr virus infectious mononucleosis. Curr. Opin. Pediatr. 2000, 12, 263–268. [Google Scholar] [CrossRef] [PubMed]
  13. Parkin, D.M. The global health burden of infection-associated cancers in the year 2002. Int. J. Cancer 2006, 118, 3030–3044. [Google Scholar] [CrossRef] [Green Version]
  14. Cohen, J.I. Epstein-Barr virus lymphoproliferative disease associated with acquired immunodeficiency. Medicine 1991, 70, 137–160. [Google Scholar] [CrossRef]
  15. Cohen, J.I. Epstein–Barr virus infection. N. Engl. J. Med. 2000, 343, 481–492. [Google Scholar] [CrossRef] [PubMed]
  16. Xuan, L.; Jiang, X.; Sun, J.; Zhang, Y.; Huang, F.; Fan, Z.; Guo, X.; Dai, M.; Liu, C.; Yu, G.; et al. Spectrum of Epstein-Barr virus-associated diseases in recipients of allogeneic hematopoietic stem cell transplantation. Transplantation 2013, 96, 560–566. [Google Scholar] [CrossRef]
  17. Tanner, J.; Alfieri, C. The Epstein-Barr virus and post-transplant lymphoproliferative disease: Interplay of immunosuppression, EBV, and the immune system in disease pathogenesis. Transpl. Infect. Dis. 2001, 3, 60–69. [Google Scholar] [CrossRef] [PubMed]
  18. Curtis, R.E.; Travis, L.B.; Rowlings, P.A.; Socie, G.; Kingma, D.W.; Banks, P.M.; Jaffe, E.S.; Sale, G.E.; Horowitz, M.M.; Witherspoon, R.P.; et al. Risk of lymphoproliferative disorders after bone marrow transplantation: A multi-institutional study. Blood 1999, 94, 2208–2216. [Google Scholar] [PubMed]
  19. Faye, A.; Vilmer, E. Post-Transplant Lymphoproliferative Disorder in Children. Paediatr. Drugs 2005, 7, 55–65. [Google Scholar] [CrossRef]
  20. Kinch, A.; Oberg, G.; Arvidson, J.; Falk, K.I.; Linde, A.; Pauksens, K. Post-transplant lymphoproliferative disease and other Epstein-Barr virus diseases in allogeneic haematopoietic stem cell transplantation after introduction of monitoring of viral load by polymerase chain reaction. Scand. J. Infect. Dis. 2007, 39, 235–244. [Google Scholar] [CrossRef]
  21. Hussein, K.; Tiede, C.; Maecker-Kolhoff, B.; Kreipe, H. Posttransplant lymphoproliferative disorder in pediatric patients. Pathobiology 2013, 80, 289–296. [Google Scholar] [CrossRef]
  22. Blaes, A.H.; Cao, Q.; Wagner, J.E.; Young, J.A.; Weisdorf, D.J.; Brunstein, C.G. Monitoring and preemptive rituximab therapy for Epstein-Barr virus reactivation after antithymocyte globulin containing nonmyeloablative conditioning for umbilical cord blood transplantation. Biol. Blood Marrow Transplant. 2010, 16, 287–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Carpenter, B.; Haque, T.; Dimopoulou, M.; Atkinson, C.; Roughton, M.; Grace, S.; Denovan, S.; Fielding, A.; Kottaridis, P.D.; Griffiths, P.; et al. Incidence and dynamics of Epstein-Barr virus reactivation after alemtuzumab-based conditioning for allogeneic hematopoietic stem-cell transplantation. Transplantation 2010, 90, 564–570. [Google Scholar] [CrossRef] [PubMed]
  24. Zallio, F.; Primon, V.; Tamiazzo, S.; Pini, M.; Baraldi, A.; Corsetti, M.T.; Gotta, F.; Bertassello, C.; Salvi, F.; Rocchetti, A.; et al. Epstein-Barr virus reactivation in allogeneic stem cell transplantation is highly related to cytomegalovirus reactivation. Clin. Transplant. 2013, 27, E491–E497. [Google Scholar] [CrossRef] [PubMed]
  25. Effective Public Health Practice Project. Quality Assessment Tool for Quantitative Studies. Hamilton, on: Effective Public Health Practice Project. Available online: https://merst.ca/wp-content/uploads/2018/02/quality-assessment-tool_2010.pdf (accessed on 30 September 2019).
  26. Effective Public Health Practice Project. Dictionary for the Effective Public Health Practice Project Quality Assessment Tool For Quantitative Studies. Available online: https://merst.ca/wp-content/uploads/2018/02/qualilty-assessment-dictionary_2017.pdf (accessed on 30 September 2019).
  27. Liu, Q.F.; Ling, Y.W.; Fan, Z.P.; Jiang, Q.L.; Sun, J.; Wu, X.L.; Zhao, J.; Wei, Q.; Zhang, Y.; Yu, G.P.; et al. Epstein-Barr virus (EBV) load in cerebrospinal fluid and peripheral blood of patients with EBV-associated central nervous system diseases after allogeneic hematopoietic stem cell transplantation. Transpl. Infect. Dis. 2013, 15, 379–392. [Google Scholar] [CrossRef]
  28. Borenstein, M. Introduction to Meta-Analysis; John Wiley & Sons: Chichester, West Sussex, UK; Hoboken, NJ, USA, 2009; p. xxviii, 421p. [Google Scholar]
  29. Higgins, J.P.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef] [Green Version]
  30. Harrer, M.; Cuijpers, P.; Furukawa, T.; Ebert, D. Doing Meta-Analysis in R: A Hands-on Guide; PROTECT Lab Erlangen: Erlangen, Germany, 2019. [Google Scholar]
  31. Ali, S.; Al Thubaiti, S.; Renzi, S.; Krueger, J.; Chiang, K.Y.; Naqvi, A.; Schechter, T.; Punnett, A.; Ali, M. Hemophagocytic lymphohistiocytosis is a sign of poor outcome in pediatric Epstein-Barr virus-associated post-transplant lymphoproliferative disease after allogeneic hematopoietic stem cell transplantation. Pediatr. Transplant. 2019, 23, e13319. [Google Scholar] [CrossRef] [Green Version]
  32. Althubaiti, S.; Ali, S.; Renzi, S.; Krueger, J.; Chiang, K.Y.; Schechter, T.; Punnett, A.; Ali, M. Lymphocyte subset at time of Epstein-Barr viremia post-allogeneic hematopoietic stem cell transplantation in children may predict development of post-transplant lymphoproliferative disease: CD8:CD20 ratio as a sensitive predictor. Pediatr. Transplant. 2019, 23. [Google Scholar] [CrossRef]
  33. Atay, D.; Akcay, A.; Erbey, F.; Ozturk, G. The impact of alternative donor types on viral infections in pediatric hematopoietic stem cell transplantation. Pediatr. Transplant. 2018, 22. [Google Scholar] [CrossRef]
  34. Auger, S.; Orsini, M.; Ceballos, P.; Fegueux, N.; Kanouni, T.; Caumes, B.; Klein, B.; Villalba, M.; Rossi, J.F. Controlled Epstein-Barr virus reactivation after allogeneic transplantation is associated with improved survival. Eur. J. Haematol. 2014, 92, 421–428. [Google Scholar] [CrossRef]
  35. Bogunia-Kubik, K.; Jaskula, E.; Lange, A. The presence of functional CCR5 and EBV reactivation after allogeneic haematopoietic stem cell transplantation. Bone Marrow Transplant. 2007, 40, 145–150. [Google Scholar] [CrossRef] [Green Version]
  36. Bogunia-Kubik, K.; Mlynarczewska, A.; Jaskula, E.; Lange, A. The presence of IFNG 3/3 genotype in the recipient associates with increased risk for Epstein-Barr virus reactivation after allogeneic haematopoietic stem cell transplantation. Br. J. Haematol. 2005, 132, 326–332. [Google Scholar] [CrossRef] [PubMed]
  37. Bordon, V.; Padalko, E.; Benoit, Y.; Dhooge, C.; Laureys, G. Incidence, kinetics, and risk factors of Epstein-Barr virus viremia in pediatric patients after allogeneic stem cell transplantation. Pediatr. Transplant. 2012, 16, 144–150. [Google Scholar] [CrossRef]
  38. Brunstein, C.G.; Weisdorf, D.J.; DeFor, T.; Barker, J.N.; Tolar, J.; van Burik, J.A.; Wagner, J.E. Marked increased risk of Epstein-Barr virus-related complications with the addition of antithymocyte globulin to a nonmyeloablative conditioning prior to unrelated umbilical cord blood transplantation. Blood 2006, 108, 2874–2880. [Google Scholar] [CrossRef]
  39. Burns, D.M.; Rana, S.; Martin, E.; Nagra, S.; Ward, J.; Osman, H.; Bell, A.I.; Moss, P.; Russell, N.H.; Craddock, C.F.; et al. Greatly reduced risk of EBV reactivation in rituximab-experienced recipients of alemtuzumab-conditioned allogeneic HSCT. Bone Marrow Transplant. 2016, 51, 825–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Buyck, H.C.E.; Ball, S.; Junagade, P.; Marsh, J.; Chakrabarti, S. Prior immunosuppressive therapy with antithymocyte globulin increases the risk of EBV-related lymphoproliferative disorder following allo-SCT for acquired aplastic anaemia. Bone Marrow Transplant. 2009, 43, 813–816. [Google Scholar] [CrossRef]
  41. Cesaro, S.; Murrone, A.; Mengoli, C.; Pillon, M.; Biasolo, M.A.; Calore, E.; Tridello, G.; Varotto, S.; Alaggio, R.; Zanesco, L.; et al. The real-time polymerase chain reaction-guided modulation of immunosuppression enables the pre-emptive management of Epstein-Barr virus reactivation after allogeneic haematopoietic stem cell transplantation. Br. J. Haematol. 2004, 128, 224–233. [Google Scholar] [CrossRef]
  42. Cesaro, S.; Pegoraro, A.; Tridello, G.; Calore, E.; Pillon, M.; Varotto, S.; Abate, D.; Barzon, L.; Mengoli, C.; Carli, M.; et al. A prospective study on modulation of immunosuppression for Epstein-Barr virus reactivation in pediatric patients who underwent unrelated hematopoietic stem-cell transplantation. Transplantation 2010, 89, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  43. Chiereghin, A.; Prete, A.; Belotti, T.; Gibertoni, D.; Piccirilli, G.; Gabrielli, L.; Pession, A.; Lazzarotto, T. Prospective Epstein-Barr virus-related post-transplant lymphoproliferative disorder prevention program in pediatric allogeneic hematopoietic stem cell transplant: Virological monitoring and first-line treatment. Transpl. Infect. Dis. 2016, 18, 44–54. [Google Scholar] [CrossRef]
  44. Chiereghin, A.; Piccirilli, G.; Belotti, T.; Prete, A.; Bertuzzi, C.; Gibertoni, D.; Gabrielli, L.; Turello, G.; Borgatti, E.C.; Barbato, F.; et al. Clinical utility of measuring Epstein-Barr virus-specific cell-mediated immunity after HSCT in addition to virological monitoring: Results from a prospective study. Med. Microbiol. Immunol. 2019, 208, 825–834. [Google Scholar] [CrossRef] [PubMed]
  45. Christopeit, M.; Janssen, N.; Weber, T.; Bacher, U.; Lautenschlager, C.; Oehme, A.; Kekule, A.S.; Schmoll, H.J. Cyclosporine area under the curve after allogeneic hematopoietic stem cell transplantation is an indicator of Epstein-Barr virus DNAemia. Leuk. Lymphoma 2013, 54, 133–137. [Google Scholar] [CrossRef]
  46. Cohen, J.; Gandhi, M.; Naik, P.; Cubitt, D.; Rao, K.; Thaker, U.; Davies, E.G.; Gaspar, H.B.; Amrolia, P.J.; Veys, P. Increased incidence of EBV-related disease following paediatric stem cell transplantation with reduced-intensity conditioning. Br. J. Haematol. 2005, 129, 229–239. [Google Scholar] [CrossRef]
  47. Comoli, P.; Basso, S.; Zecca, M.; Pagliara, D.; Baldanti, F.; Bernardo, M.E.; Barberi, W.; Moretta, A.; Labirio, M.; Paulli, M.; et al. Preemptive therapy of EBV-related lymphoproliferative disease after pediatric haploidentical stem cell transplantation. Am. J. Transplant. 2007, 7, 1648–1655. [Google Scholar] [CrossRef] [PubMed]
  48. Czyzewski, K.; Styczynski, J.; Giebel, S.; Fraczkiewicz, J.; Salamonowicz, M.; Zajac-Spychala, O.; Zaucha-Prazmo, A.; Drozd-Sokolowska, J.; Waszczuk-Gajda, A.; Dybko, J.; et al. Age-dependent determinants of infectious complications profile in children and adults after hematopoietic cell transplantation: Lesson from the nationwide study. Ann. Hematol. 2019, 98, 2197–2211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. D’Aveni, M.; Aissi-Rothe, L.; Venard, V.; Salmon, A.; Falenga, A.; Decot, V.; Virion, J.M.; Wang, Y.; Clement, L.; Latger-Cannard, V.; et al. The clinical value of concomitant Epstein Barr virus (EBV)-DNA load and specific immune reconstitution monitoring after allogeneic hematopoietic stem cell transplantation. Transpl. Immunol. 2011, 24, 224–232. [Google Scholar] [CrossRef]
  50. Dumas, P.Y.; Ruggeri, A.; Robin, M.; Crotta, A.; Abraham, J.; Forcade, E.; Bay, J.O.; Michallet, M.; Bertrand, Y.; Socie, G.; et al. Incidence and risk factors of EBV reactivation after unrelated cord blood transplantation: A Eurocord and Societe Francaise de Greffe de Moelle-Therapie Cellulaire collaborative study. Bone Marrow Transplant. 2013, 48, 253–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Duver, F.; Weisbrich, B.; Eyrich, M.; Wolfl, M.; Schlegel, P.G.; Wiegering, V. Viral reactivations following hematopoietic stem cell transplantation in pediatric patients—A single center 11-year analysis. PLoS ONE 2020, 15, e0228451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Elmahdi, S.; Muramatsu, H.; Narita, A.; Torii, Y.; Ismael, O.; Kawashima, N.; Okuno, Y.; Sekiya, Y.; Xu, Y.; Wang, X.; et al. Correlation of rabbit antithymocyte globulin serum levels and clinical outcomes in children who received hematopoietic stem cell transplantation from an alternative donor. Pediatr. Transplant. 2016, 20, 105–113. [Google Scholar] [CrossRef] [PubMed]
  53. Fan, J.; Jing, M.; Yang, M.; Xu, L.; Liang, H.; Huang, Y.; Yang, R.; Gui, G.; Wang, H.; Gong, S.; et al. Herpesvirus infections in hematopoietic stem cell transplant recipients seropositive for human cytomegalovirus before transplantation. Int. J. Infect. Dis. 2016, 46, 89–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Figgins, B.; Hammerstrom, A.; Ariza-Heredia, E.; Oran, B.; Milton, D.R.; Yeh, J. Characterization of Viral Infections after Antithymocyte Globulin-Based Conditioning in Adults Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 1837–1843. [Google Scholar] [CrossRef]
  55. Fujimoto, A.; Hiramoto, N.; Yamasaki, S.; Inamoto, Y.; Uchida, N.; Maeda, T.; Mori, T.; Kanda, Y.; Kondo, T.; Shiratori, S.; et al. Risk Factors and Predictive Scoring System For Post-Transplant Lymphoproliferative Disorder after Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 1441–1449. [Google Scholar] [CrossRef]
  56. Gao, X.N.; Lin, J.; Wang, L.J.; Li, F.; Li, H.H.; Wang, S.H.; Huang, W.R.; Gao, C.J.; Yu, L.; Liu, D.H. Risk factors and clinical outcomes of Epstein-Barr virus DNAemia and post-transplant lymphoproliferative disorders after haploidentical and matched-sibling PBSCT in patients with hematologic malignancies. Ann. Hematol. 2019, 98, 2163–2177. [Google Scholar] [CrossRef]
  57. Garcia-Cadenas, I.; Castillo, N.; Martino, R.; Barba, P.; Esquirol, A.; Novelli, S.; Orti, G.; Garrido, A.; Saavedra, S.; Moreno, C.; et al. Impact of Epstein Barr virus-related complications after high-risk allo-SCT in the era of pre-emptive rituximab. Bone Marrow Transplant. 2015, 50, 579–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Han, S.B.; Bae, E.Y.; Lee, J.W.; Jang, P.S.; Lee, D.G.; Chung, N.G.; Jeong, D.C.; Cho, B.; Lee, S.J.; Kang, J.H.; et al. Features of Epstein-Barr virus reactivation after allogeneic hematopoietic cell transplantation in Korean children living in an area of high seroprevalence against Epstein-Barr virus. Int. J. Hematol. 2014, 100, 188–199. [Google Scholar] [CrossRef] [PubMed]
  59. Hiwarkar, P.; Gaspar, H.B.; Gilmour, K.; Jagani, M.; Chiesa, R.; Bennett-Rees, N.; Breuer, J.; Rao, K.; Cale, C.; Goulden, N.; et al. Impact of viral reactivations in the era of pre-emptive antiviral drug therapy following allogeneic haematopoietic SCT in paediatric recipients. Bone Marrow Transplant. 2013, 48, 803–808. [Google Scholar] [CrossRef]
  60. Hoegh-Petersen, M.; Goodyear, D.; Geddes, M.N.; Liu, S.; Ugarte-Torres, A.; Liu, Y.; Walker, J.T.; Fonseca, K.; Daly, A.; Duggan, P.; et al. High incidence of post transplant lymphoproliferative disorder after antithymocyte globulin-based conditioning and ineffective prediction by day 28 EBV-specific T lymphocyte counts. Bone Marrow Transplant. 2011, 46, 1104–1112. [Google Scholar] [CrossRef]
  61. Hoshino, Y.; Kimura, H.; Tanaka, N.; Tsuge, I.; Kudo, K.; Horibe, K.; Kato, K.; Matsuyama, T.; Kikuta, A.; Kojima, S.; et al. Prospective monitoring of the Epstein-Barr virus DNA by a real-time quantitative polymerase chain reaction after allogenic stem cell transplantation. Br. J. Haematol. 2001, 115, 105–111. [Google Scholar] [CrossRef]
  62. Islam, M.S.; Anoop, P.; Gordon-Smith, E.C.; Rice, P.; Datta-Nemdharry, P.; Marsh, J.C. Epstein-Barr virus infections after allogeneic stem cell transplantation: A comparison between non-malignant and malignant hematological disorders. Hematology 2010, 15, 344–350. [Google Scholar] [CrossRef] [PubMed]
  63. Issa, H.; Sharma, N.; Zhao, Q.; Ruppert, A.S.; Elder, P.; Benson, D.M.; Penza, S.; Vasu, S.; William, B.; Jaglowski, S.; et al. Comparison of Two Doses of Antithymocyte Globulin in Reduced-Intensity Conditioning Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 1993–2001. [Google Scholar] [CrossRef]
  64. Kutnik, P.; Kwiatkowska, A.; Krawczyk, D.; Polak, O.; Jawoszek, P.; Puchala, D.; Zaucha-Prazmo, A.; Kowalczyk, J. The impact of donor-recipient sex matching on transplantrelated complications in children after allogeneic haematopoietic stem cell transplantation—A single-centre, retrospective study. Pediatr. Pol. 2019, 94, 158–161. [Google Scholar] [CrossRef]
  65. Jaskula, E.; Dlubek, D.; Sedzimirska, M.; Duda, D.; Tarnowska, A.; Lange, A. Reactivations of cytomegalovirus, human herpes virus 6, and Epstein-Barr virus differ with respect to risk factors and clinical outcome after hematopoietic stem cell transplantation. Transplant. Proc. 2010, 42, 3273–3276. [Google Scholar] [CrossRef]
  66. Juvonen, E.; Aalto, S.; Tarkkanen, J.; Volin, L.; Hedman, K.; Ruutu, T. Retrospective evaluation of serum Epstein Barr virus DNA levels in 406 allogeneic stem cell transplant patients. Haematologica 2007, 92, 819–825. [Google Scholar] [CrossRef]
  67. Kalra, A.; Roessner, C.; Jupp, J.; Williamson, T.; Tellier, R.; Chaudhry, A.; Khan, F.; Taparia, M.; Jimenez-Zepeda, V.H.; Stewart, D.A.; et al. Risk factors for post-transplant lymphoproliferative disorder after Thymoglobulin-conditioned hematopoietic cell transplantation. Clin. Transplant. 2018, 32, e13150. [Google Scholar] [CrossRef]
  68. Kullberg-Lindh, C.; Mellgren, K.; Friman, V.; Fasth, A.; Ascher, H.; Nilsson, S.; Lindh, M. Opportunistic virus DNA levels after pediatric stem cell transplantation: Serostatus matching, anti-thymocyte globulin, and total body irradiation are additive risk factors. Transpl. Infect. Dis. 2011, 13, 122–130. [Google Scholar] [CrossRef] [PubMed]
  69. Laberko, A.; Bogoyavlenskaya, A.; Shelikhova, L.; Shekhovtsova, Z.; Balashov, D.; Voronin, K.; Kurnikova, E.; Boyakova, E.; Raykina, E.; Brilliantova, V.; et al. Risk Factors for and the Clinical Impact of Cytomegalovirus and Epstein-Barr Virus Infections in Pediatric Recipients of TCR-alpha/beta- and CD19-Depleted Grafts. Biol. Blood Marrow Transplant. 2017, 23, 483–490. [Google Scholar] [CrossRef] [Green Version]
  70. Landgren, O.; Gilbert, E.S.; Rizzo, J.D.; Socie, G.; Banks, P.M.; Sobocinski, K.A.; Horowitz, M.M.; Jaffe, E.S.; Kingma, D.W.; Travis, L.B.; et al. Risk factors for lymphoproliferative disorders after allogeneic hematopoietic cell transplantation. Blood 2009, 113, 4992–5001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Li, S.; Wang, B.; Fu, L.; Pang, Y.; Zhu, G.; Zhou, X.; Ma, J.; Su, Y.; Qin, M.; Wu, R. Hematopoietic stem cell transplantation without in vivo T-cell depletion for pediatric aplastic anemia: A single-center experience. Pediatr. Transplant. 2018, 22, e13204. [Google Scholar] [CrossRef] [PubMed]
  72. Lin, R.; Wang, Y.; Huang, F.; Fan, Z.; Zhang, S.; Yang, T.; Xu, Y.; Xu, N.; Xuan, L.; Ye, J.; et al. Two dose levels of rabbit antithymocyte globulin as graft-versus-host disease prophylaxis in haploidentical stem cell transplantation: A multicenter randomized study. BMC Med. 2019, 17, 156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Liu, J.; Gao, H.; Xu, L.P.; Mo, X.D.; Liu, R.; Liang, S.; Wu, N.; Wang, M.; Wang, Z.; Chang, Y.J.; et al. Immunosuppressant indulges EBV reactivation and related lymphoproliferative disease by inhibiting Vdelta2+ T cells activities after hematopoietic transplantation for blood malignancies. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Liu, Q.; Xuan, L.; Liu, H.; Huang, F.; Zhou, H.; Fan, Z.; Zhao, K.; Wu, M.; Xu, L.; Zhai, X.; et al. Molecular monitoring and stepwise preemptive therapy for Epstein-Barr virus viremia after allogeneic stem cell transplantation. Am. J. Hematol. 2013, 88, 550–555. [Google Scholar] [CrossRef]
  75. Liu, J.; Bian, Z.; Wang, X.; Xu, L.P.; Fu, Q.; Wang, C.; Chang, Y.J.; Wang, Y.; Zhang, X.H.; Jiang, Z.; et al. Inverse correlation of Vdelta2(+) T-cell recovery with EBV reactivation after haematopoietic stem cell transplantation. Br. J. Haematol. 2018, 180, 276–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Marinho-Dias, J.; Baldaque, I.; Pinho-Vaz, C.; Leite, L.; Branca, R.; Campilho, F.; Campos, A.; Medeiros, R.; Sousa, H. Association of Epstein-Barr virus infection with allogeneic hematopoietic stem cell transplantation in patients in Portugal. Mol. Med. Rep. 2019, 19, 1435–1442. [Google Scholar] [CrossRef] [Green Version]
  77. Meijer, E.; Dekker, A.W.; Lokhorst, H.M.; Petersen, E.J.; Nieuwenhuis, H.K.; Verdonck, L.F. Low incidence of infectious complications after nonmyeloablative compared with myeloablative allogeneic stem cell transplantation. Transpl. Infect. Dis. 2004, 6, 171–178. [Google Scholar] [CrossRef] [PubMed]
  78. Mountjoy, L.; Jain, T.; Kunze, K.L.; Khera, N.; Sproat, L.Z.; Jennifer, W.; McCallen, M.; Leis, J.F.; Noel, P.; Slack, J.L.; et al. Clinical outcomes with low dose anti-thymocyte globulin in patients undergoing matched unrelated donor allogeneic hematopoietic cell transplantation. Leuk. Lymphoma 2020, 61, 1996–2002. [Google Scholar] [CrossRef] [PubMed]
  79. Neumann, T.; Schneidewind, L.; Thiele, T.; Pink, D.; Schulze, M.; Schmidt, C.; Krüger, W. No indication of increased infection rates using low-dose alemtuzumab instead of anti-thymocyte globulin as graft-versus-host disease prophylaxis before allogeneic stem cell transplantation. Transpl. Infect. Dis. 2018, 20, e12822. [Google Scholar] [CrossRef] [PubMed]
  80. Nowak, J.; Gwozdowicz, S.; Graczyk-Pol, E.; Mika-Witkowska, R.; Rogatko-Koros, M.; Nestorowicz, K.; Szlendak, U.; Malinowska, A.; Kaczmarek, B.; Nasilowska-Adamska, B.; et al. Epstein-Barr virus infections are strongly dependent on activating and inhibitory KIR-HLA pairs after T-cell replate unrelated hematopoietic stem cell transplantation, the principles, and method of pairing analysis. HLA 2019, 94, 40–48. [Google Scholar] [CrossRef] [PubMed]
  81. Omar, H.; Hagglund, H.; Gustafsson-Jernberg, A.; LeBlanc, K.; Mattsson, J.; Remberger, M.; Ringden, O.; Sparrelid, E.; Sundin, M.; Winiarski, J.; et al. Targeted monitoring of patients at high risk of post-transplant lymphoproliferative disease by quantitative Epstein-Barr virus polymerase chain reaction. Transpl. Infect. Dis. 2009, 11, 393–399. [Google Scholar] [CrossRef] [Green Version]
  82. Pagliuca, S.; Bommier, C.; Michonneau, D.; Meignin, V.; Salmona, M.; Robin, M.; Prata, P.H.; Xhaard, A.; de Fontbrune, F.S.; Feghoul, L.; et al. Epstein-Barr Virus-Associated Post-Transplantation Lymphoproliferative Disease in Patients Who Received Anti-CD20 after Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 2490–2500. [Google Scholar] [CrossRef]
  83. Park, J.; Lim, S.H.; Kim, S.H.; Yun, J.; Kim, C.K.; Lee, S.C.; Won, J.H.; Hong, D.S.; Park, S.K. Is immunological recovery clinically relevant at 100 days after allogeneic transplantation? Korean J. Intern. Med. 2020, 21. [Google Scholar] [CrossRef] [Green Version]
  84. Peric, Z.; Cahu, X.; Chevallier, P.; Brissot, E.; Malard, F.; Guillaume, T.; Delaunay, J.; Ayari, S.; Dubruille, V.; Le Gouill, S.; et al. Features of EBV reactivation after reduced intensity conditioning unrelated umbilical cord blood transplantation. Bone Marrow Transplant. 2012, 47, 251–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Peric, Z.; Cahu, X.; Chevallier, P.; Brissot, E.; Malard, F.; Guillaume, T.; Delaunay, J.; Ayari, S.; Dubruille, V.; Le Gouill, S.; et al. Features of Epstein-Barr Virus (EBV) reactivation after reduced intensity conditioning allogeneic hematopoietic stem cell transplantation. Leukemia 2011, 25, 932–938. [Google Scholar] [CrossRef] [Green Version]
  86. Ru, Y.; Zhang, X.; Song, T.; Ding, Y.; Zhu, Z.; Fan, Y.; Xu, Y.; Sun, A.; Qiu, H.; Jin, Z.; et al. Epstein-Barr virus reactivation after allogeneic hematopoietic stem cell transplantation: Multifactorial impact on transplant outcomes. Bone Marrow Transplant. 2020, 55, 1754–1762. [Google Scholar] [CrossRef]
  87. Rustia, E.; Violago, L.; Jin, Z.; Foca, M.D.; Kahn, J.M.; Arnold, S.; Sosna, J.; Bhatia, M.; Kung, A.L.; George, D.; et al. Risk Factors and Utility of a Risk-Based Algorithm for Monitoring Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections in Pediatric Recipients after Allogeneic Hematopoietic Cell Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 1646–1653. [Google Scholar] [CrossRef] [Green Version]
  88. Sanz, J.; Arango, M.; Senent, L.; Jarque, I.; Montesinos, P.; Sempere, A.; Lorenzo, I.; Martin, G.; Moscardo, F.; Mayordomo, E.; et al. EBV-associated post-transplant lymphoproliferative disorder after umbilical cord blood transplantation in adults with hematological diseases. Bone Marrow Transplant. 2014, 49, 397–402. [Google Scholar] [CrossRef]
  89. Bueltzingsloewen, A.S.-V.; Morand, P.; Buisson, M.; Souillet, G.; Chambost, H.; Bosson, J.L.; Bordigoni, P. A prospective study of Epstein-Barr virus load in 85 hematopoietic stem cell transplants. Bone Marrow Transplant. 2002, 29, 21–28. [Google Scholar] [CrossRef] [Green Version]
  90. Styczynski, J.; Gil, L.; Tridello, G.; Ljungman, P.; Donnelly, J.P.; Van Der Velden, W.; Omar, H.; Martino, R.; Halkes, C.; Faraci, M.; et al. Response to rituximab-based therapy and risk factor analysis in epstein barr virus-related lymphoproliferative disorder after hematopoietic stem cell transplant in children and adults: A study from the infectious diseases working party of the european group for blood and marrow transplantation. Clin. Infect. Dis. 2013, 57, 794–802. [Google Scholar] [PubMed] [Green Version]
  91. Torre-Cisneros, J.; Roman, J.; Torres, A.; Herrera, C.; Caston, J.J.; Rivero, A.; Mingot, E.; Rojas, R.; Martin, C.; Martinez, F.; et al. Control of Epstein-Barr virus load and lymphoproliferative disease by maintenance of CD8+ T lymphocytes in the T lymphocyte-depleted graft after bone marrow transplantation. J. Infect. Dis. 2004, 190, 1596–1599. [Google Scholar] [CrossRef] [Green Version]
  92. Trottier, H.; Buteau, C.; Robitaille, N.; Duval, M.; Tucci, M.; Lacroix, J.; Alfieri, C. Transfusion-related Epstein-Barr virus infection among stem cell transplant recipients: A retrospective cohort study in children. Transfusion 2012, 52, 2653–2663. [Google Scholar] [CrossRef]
  93. Tsoumakas, K.; Giamaiou, K.; Goussetis, E.; Graphakos, S.; Kossyvakis, A.; Horefti, E.; Mentis, A.; Elefsiniotis, I.; Pavlopoulou, I.D. Epidemiology of viral infections among children undergoing hematopoietic stem cell transplant: Alpha prospective single-center study. Transpl. Infect. Dis. 2019, 21, e13095. [Google Scholar] [CrossRef]
  94. Uhlin, M.; Wikell, H.; Sundin, M.; Blennow, O.; Maeurer, M.; Ringden, O.; Winiarski, J.; Ljungman, P.; Remberger, M.; Mattsson, J. Risk factors for Epstein-Barr virus-related post-transplant lymphoproliferative disease after allogeneic hematopoietic stem cell transplantation. Haematologica 2014, 99, 346–352. [Google Scholar] [CrossRef] [PubMed]
  95. Van Der Velden, W.J.F.M.; Mori, T.; Stevens, W.B.C.; De Haan, A.F.J.; Stelma, F.F.; Blijlevens, N.M.A.; Donnelly, J.P. Reduced PTLD-related mortality in patients experiencing EBV infection following allo-SCT after the introduction of a protocol incorporating pre-emptive rituximab. Bone Marrow Transplant. 2013, 48, 1465–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Van Esser, J.W.J.; Van Der Holt, B.; Meijer, E.; Niesters, H.G.M.; Trenschel, R.; Thijsen, S.F.T.; Van Loon, A.M.; Frassoni, F.; Bacigalupo, A.; Schaefer, U.W.; et al. Epstein-Barr virus (EBV) reactivation is a frequent event after allogeneic stem cell transplantation (SCT) and quantitatively predicts EBV-lymphoproliferative disease following T-cell-depleted SCT. Blood 2001, 98, 972–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Wang, H.; Zhang, T.T.; Qi, J.Q.; Chu, T.T.; Miao, M.; Qiu, H.Y.; Fu, C.C.; Tang, X.W.; Ruan, C.G.; Wu, D.P.; et al. Incidence, risk factors, and clinical significance of Epstein-Barr virus reactivation in myelodysplastic syndrome after allogeneic haematopoietic stem cell transplantation. Ann. Hematol. 2019, 98, 987–996. [Google Scholar] [CrossRef] [PubMed]
  98. Xu, L.P.; Zhang, C.L.; Mo, X.D.; Zhang, X.H.; Chen, H.; Han, W.; Chen, Y.H.; Wang, Y.; Yan, C.H.; Wang, J.Z.; et al. Epstein-Barr Virus-Related Post-Transplantation Lymphoproliferative Disorder after Unmanipulated Human Leukocyte Antigen Haploidentical Hematopoietic Stem Cell Transplantation: Incidence, Risk Factors, Treatment, and Clinical Outcomes. Biol. Blood Marrow Transplant. 2015, 21, 2185–2191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Xuan, L.; Huang, F.; Fan, Z.; Zhou, H.; Zhang, X.; Yu, G.; Zhang, Y.; Liu, C.; Sun, J.; Liu, Q. Effects of intensified conditioning on Epstein-Barr virus and cytomegalovirus infections in allogeneic hematopoietic stem cell transplantation for hematological malignancies. J. Hematol. Oncol. 2012, 5, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Yu, X.X.; Cao, X.H.; Yan, H.; Luo, X.Y.; Zhao, X.S.; Sun, Y.Q.; Wang, Y.; Xu, L.P.; Zhang, X.H.; Chang, Y.J.; et al. Delay expression of NKp30 on NK cells correlates with long-term mycophenolate mofetil treatment and higher EBV viremia post allogenic hematological stem cells transplantation. Clin. Immunol. 2019, 205, 49–56. [Google Scholar] [CrossRef] [PubMed]
  101. Zhou, L.; Gao, Z.Y.; Lu, D.P. Incidence, risk factors, and clinical outcomes associated with Epstein-Barr virus-DNAemia and Epstein-Barr virus-associated disease in patients after haploidentical allogeneic stem cell transplantation: A single-center study. Clin. Transplant. 2020, 34. [Google Scholar] [CrossRef]
  102. Zhou, L.; Gao, Z.Y.; Lu, D.P. Comparison of ATG-thymoglobulin with ATG-Fresenius for Epstein-Barr virus infections and graft-versus-host-disease in patients with hematological malignances after haploidentical hematopoietic stem cell transplantation: A single-center experience. Ann. Hematol. 2020, 99, 1389–1400. [Google Scholar] [CrossRef]
  103. Sundin, M.; Le Blanc, K.; Ringden, O.; Barkholt, L.; Omazic, B.; Lergin, C.; Levitsky, V.; Remberger, M. The role of HLA mismatch, splenectomy and recipient Epstein-Barr virus seronegativity as risk factors in post-transplant lymphoproliferative disorder following allogeneic hematopoietic stem cell transplantation. Haematologica 2006, 91, 1059–1067. [Google Scholar]
  104. Hoegh-Petersen, M.; Amin, M.; Liu, Y.; Ugarte-Torres, A.; Williamson, T.; Podgorny, P.; Russell, J.; Grigg, A.; Ritchie, D.; Storek, J. Anti-thymocyte globulins capable of binding to T and B cells reduce graft-vs-host disease without increasing relapse. Bone Marrow Transplant. 2013, 48, 105–114. [Google Scholar] [CrossRef] [Green Version]
  105. Mohty, M. Mechanisms of action of antithymocyte globulin: T-cell depletion and beyond. Leukemia 2007, 21, 1387–1394. [Google Scholar] [CrossRef] [Green Version]
  106. Kekre, N.; Antin, J.H. ATG in allogeneic stem cell transplantation: Standard of care in 2017? Counterpoint. Blood Adv. 2017, 1, 573–576. [Google Scholar] [CrossRef] [Green Version]
  107. Ram, R.; Storb, R. Pharmacologic Prophylaxis Regimens for Acute Graft-Versus-Host Disease: Past, Present and Future; Taylor & Francis: Oxfordshire, UK, 2013. [Google Scholar]
  108. Shimabukuro-Vornhagen, A.; Hallek, M.J.; Storb, R.F.; von Bergwelt-Baildon, M.S. The role of B cells in the pathogenesis of graft-versus-host disease. Blood 2009, 114, 4919–4927. [Google Scholar] [CrossRef] [Green Version]
  109. Zeiser, R.; Blazar, B.R. Acute graft-versus-host disease—Biologic process, prevention, and therapy. N. Engl. J. Med. 2017, 377, 2167–2179. [Google Scholar] [CrossRef] [PubMed]
  110. Société Française de Greffe de Moelle et Thérapie Cellulaire. Greffe de Cellules Souches Hématopoïétiques: Livret D’information et D’aide à la Décision à L’usage des Patients; Société Française de Greffe de Moelle et Thérapie Cellulaire: Toulouse, France, 2013. [Google Scholar]
  111. Andréoli, A.L.; de Latour, R.P.; Thépot, S.; Socié, G. La maladie chronique du greffon contre l’hôte. Hématologie 2010, 16, 420–429. [Google Scholar]
  112. Enok Bonong, P.R.; Buteau, C.; Delage, G.; Tanner, J.E.; Lacroix, J.; Duval, M.; Laporte, L.; Tucci, M.; Robitaille, N.; Spinella, P.C.; et al. Transfusion-related Epstein-Barr virus (EBV) infection: A multicenter prospective cohort study among pediatric recipients of hematopoietic stem cell transplants (TREASuRE study). Transfusion 2021, 61, 144–158. [Google Scholar] [CrossRef]
  113. Čičkušić, E.; Mustedanagić-Mujanović, J.; Iljazović, E.; Karasalihović, Z.; Škaljić, I. Association of Hodgkin’s lymphoma with Epstein Barr virus infection. Bosn. J. Basic Med. Sci. 2007, 7, 58. [Google Scholar] [CrossRef] [Green Version]
  114. Glaser, S.L.; Lin, R.J.; Stewart, S.L.; Ambinder, R.F.; Jarrett, R.F.; Brousset, P.; Pallesen, G.; Gulley, M.L.; Khan, G.; O’Grady, J. Epstein-Barr virus-associated Hodgkin’s disease: Epidemiologic characteristics in international data. Int. J. Cancer 1997, 70, 375–382. [Google Scholar] [CrossRef]
  115. Hakim, H.; Gibson, C.; Pan, J.; Srivastava, K.; Gu, Z.; Bankowski, M.J.; Hayden, R.T. Comparison of various blood compartments and reporting units for the detection and quantification of Epstein-Barr virus in peripheral blood. J. Clin. Microbiol. 2007, 45, 2151–2155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Greenfield, H.M.; Gharib, M.I.; Turner, A.J.; Guiver, M.; Carr, T.; Will, A.M.; Wynn, R.F. The impact of monitoring Epstein-Barr virus PCR in paediatric bone marrow transplant patients: Can it successfully predict outcome and guide intervention? Pediatr. Blood Cancer 2006, 47, 200–205. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Search strategy flowchart. * The reasons for exclusion of these articles were as follows: Two articles were excluded because their sample is a subset of the sample from two other articles. There was no univariate or multivariate statistical analysis for the identification of risk factors for post-transplant active EBV infection or PTLD in 26 articles and in three articles. EBV post-transplant infection was combined with other viral infections in a single variable.
Figure 1. Search strategy flowchart. * The reasons for exclusion of these articles were as follows: Two articles were excluded because their sample is a subset of the sample from two other articles. There was no univariate or multivariate statistical analysis for the identification of risk factors for post-transplant active EBV infection or PTLD in 26 articles and in three articles. EBV post-transplant infection was combined with other viral infections in a single variable.
Vaccines 09 00288 g001
Figure 2. Summary of some characteristics of the studies included in the systematic review. (A) Number of studies by year of publication; (B) Proportion (number) of studies by type of population; (C) Proportion (number) of studies according to the type of outcome; (D) Descriptive statistics on sample size by type of population; (E) Number of studies according to the type of statistical analysis carried out, the type of population and the outcome; (F) Number of studies by type of outcome and by quality level. EBV and PTLD: The two outcomes were studied separately in the same article; EBV/PTLD: The two outcomes were combined into one. * Studies with the outcome PTLD/EBV and the studies with outcome PTLD were considered together.
Figure 2. Summary of some characteristics of the studies included in the systematic review. (A) Number of studies by year of publication; (B) Proportion (number) of studies by type of population; (C) Proportion (number) of studies according to the type of outcome; (D) Descriptive statistics on sample size by type of population; (E) Number of studies according to the type of statistical analysis carried out, the type of population and the outcome; (F) Number of studies by type of outcome and by quality level. EBV and PTLD: The two outcomes were studied separately in the same article; EBV/PTLD: The two outcomes were combined into one. * Studies with the outcome PTLD/EBV and the studies with outcome PTLD were considered together.
Vaccines 09 00288 g002
Figure 3. Summary of risk factors for post-transplant EBV infection (A) and for PTLD (B) explored in the studies that controlled for confounding. Abbreviations: ADV: adenovirus; aGvHD: acute graft-versus-host disease; ATG: anti-thymocyte globulin; BM: bone marrow; CB: cord blood; CCR5: C–C chemokine receptor 5; cGvHD: chronic graft-versus-host disease; CMV: cytomegalovirus; CsA: cyclosporine A; D/R: donor/recipient; GvHD: graft-versus-host disease; HLA: human leukocyte antigen; HSCT: hematopoietic stem cell transplantation; IFNG: interferon-γ gene; MAC: myeloablative conditioning; MMF: mycophenolate mofetil; MSC: mesenchymal stromal cells; MTX: methotrexate; NK: natural killer cells; NMAC: nonmyeloablative conditioning; PBSC: peripheral blood stem cells; PLT: platelets; RIC: reduced-intensity conditioning; TBI: total body irradiation; TCD: T-cell depletion; URD: unrelated donor.
Figure 3. Summary of risk factors for post-transplant EBV infection (A) and for PTLD (B) explored in the studies that controlled for confounding. Abbreviations: ADV: adenovirus; aGvHD: acute graft-versus-host disease; ATG: anti-thymocyte globulin; BM: bone marrow; CB: cord blood; CCR5: C–C chemokine receptor 5; cGvHD: chronic graft-versus-host disease; CMV: cytomegalovirus; CsA: cyclosporine A; D/R: donor/recipient; GvHD: graft-versus-host disease; HLA: human leukocyte antigen; HSCT: hematopoietic stem cell transplantation; IFNG: interferon-γ gene; MAC: myeloablative conditioning; MMF: mycophenolate mofetil; MSC: mesenchymal stromal cells; MTX: methotrexate; NK: natural killer cells; NMAC: nonmyeloablative conditioning; PBSC: peripheral blood stem cells; PLT: platelets; RIC: reduced-intensity conditioning; TBI: total body irradiation; TCD: T-cell depletion; URD: unrelated donor.
Vaccines 09 00288 g003
Figure 4. Forest plots for the association between ATG use and post-transplant EBV infection according to studies estimating adjusted HR/SHR and adjusted OR. (*) In the study by Laberko et al., two estimates of the hazard ratio (HR) of the association between the use of ATG and post-transplant EBV infection were reported, corresponding to the use of horse ATG on one hand and rabbit ATG on the other. These two HRs were combined using a meta-analysis with inverse variance as a method. The results obtained were used to carry out the meta-analysis, including the other studies. Abbreviations: OR: odds ratio; HR: hazard ratio; SHR: subhazard ratio; CI: confidence intervals; ATG: anti-thymocyte globulin.
Figure 4. Forest plots for the association between ATG use and post-transplant EBV infection according to studies estimating adjusted HR/SHR and adjusted OR. (*) In the study by Laberko et al., two estimates of the hazard ratio (HR) of the association between the use of ATG and post-transplant EBV infection were reported, corresponding to the use of horse ATG on one hand and rabbit ATG on the other. These two HRs were combined using a meta-analysis with inverse variance as a method. The results obtained were used to carry out the meta-analysis, including the other studies. Abbreviations: OR: odds ratio; HR: hazard ratio; SHR: subhazard ratio; CI: confidence intervals; ATG: anti-thymocyte globulin.
Vaccines 09 00288 g004
Figure 5. Forest plots for the association between ATG use and post-transplant lymphoproliferative disease (PTLD). Abbreviations: OR: odds ratio; HR: hazard ratio; SHR: subhazard ratio; CI: confidence intervals; ATG: anti-thymocyte globulin.
Figure 5. Forest plots for the association between ATG use and post-transplant lymphoproliferative disease (PTLD). Abbreviations: OR: odds ratio; HR: hazard ratio; SHR: subhazard ratio; CI: confidence intervals; ATG: anti-thymocyte globulin.
Vaccines 09 00288 g005
Table 1. Characteristics of the 77 studies included in the systematic review.
Table 1. Characteristics of the 77 studies included in the systematic review.
First Author, YearCountryStudy TypeStudy PopulationSample SizeOutcomeMedian (Range) of Follow-UpStatistical AnalysisOverall Rating (Table S2)
Ali, 2019 [31]CanadaRetrospectiveP408PTLDNRUnivariateWeak
Althubaiti, 2019 [32]CanadaRetrospectiveP26PTLDNRUnivariateWeak
Atay, 2018 [33]TurkeyRetrospectiveP171EBV 14 monthsUnivariateWeak
Auger, 2014 [34]FranceRetrospectiveA190EBV36.6 months (95% IC 31.5–45.7)MultivariateWeak
Bogunia-Kubik, 2007 [35]PolandRetrospectiveP and A92EBVNRMultivariateStrong
Bogunia-Kubik, 2005 [36]PolandRetrospectiveP and A83EBVNRMultivariateStrong
Bordon, 2012 [37]BelgiumRetrospectiveP80EBVNRMultivariateModerate
Brunstein, 2006 [38]USAMulticenter retrospectiveP and A335EBV/PTLD1.2 (77 days–9.2 years)MultivariateModerate
Burns, 2016 [39]United KingdomRetrospectiveP and A186EBV28 monthsMultivariateStrong
Buyck, 2009 [40]United KingdomRetrospectiveP and A87PTLDNRMultivariateModerate
Carpenter, 2010 [23]United KingdomRetrospectiveP and A111 aEBV2.4 yearsMultivariateStrong
Cesaro, 2004 [41]ItalyRetrospectiveP79 bEBVNRMultivariateModerate
Cesaro, 2010 [42]ItalyRetrospectiveP89EBVNRUnivariateWeak
Chiereghin, 2016 [43]ItalyProspectiveP28EBV7.1 monthsUnivariateWeak
Chiereghin, 2019 [44]ItalyProspectiveP and A51EBVNRUnivariateWeak
Christopeit, 2013 [45]USARetrospectiveA28 cEBVNRMultivariateModerate
Cohen, 2005 [46]United KingdomProspectiveP128EBVNRMultivariateModerate
Cohen, 2005 [46]United KingdomProspectiveP128PTLDNRMultivariateModerate
Comoli, 2007 [47]ItalyProspectiveP and A27EBV23 monthsUnivariateWeak
Czyżewski, 2019 [48]PolandRetrospective multicenter studyP and A1569EBVNRUnivariateWeak
D’Aveni, 2011 [49]FranceRetrospectiveP and A40 dEBVNRUnivariateWeak
Dumas, 2013 [50]FranceMulticenter retrospectiveP and A175EBVNRMultivariateModerate
Düver, 2020 [51]GermanyRetrospectiveP107EBV365 (range: 22–365) daysMultivariateStrong
Elmahdi, 2016 [52]JapanRetrospectiveP37EBVNRMultivariateModerate
Fan, 2016 [53]ChinaRetrospectiveP and A44 eEBV NRMultivariateModerate
Figgins, 2019 [54]USARetrospectiveA123EBV12.8 (range: 1.0–23.1) monthsUnivariateWeak
Fujimoto, 2019 [55]JapanMulticenter retrospectiveP and A64,539PTLDNRMultivariateStrong
Gao, 2019 [56]ChinaRetrospectiveP and A200EBVNRMultivariateStrong
Gao, 2019 [56]ChinaRetrospectiveP and A200PTLDNRMultivariateStrong
Garcia-Cadenas, 2015 [57]SpainProspectiveA93EBVNRMultivariateStrong
Garcia-Cadenas, 2015 [57]SpainProspectiveA93PTLDNRMultivariateStrong
Han, 2014 [58]KoreaRetrospectiveP248EBVNRUnivariateWeak
Hiwarkar, 2013 [59]United KingdomRetrospectiveP278EBVNRMultivariateModerate
Hoegh-Petersen, 2011 [60]CanadaRetrospectiveA307PTLD375 (28–1727) daysUnivariateWeak
Hoshino, 2001 [61]JapanProspectiveP and A38EBVNRUnivariateWeak
Islam, 2010 [62]United KingdomRetrospectiveP and A83EBV4.2 (0.9–8.1) yearsUnivariateWeak
Issa, 2019 [63]USARetrospectiveA357EBVNRUnivariateWeak
Kutnik, 2019 [64]PolandRetrospectiveP198EBV12 monthsUnivariateWeak
Jaskula, 2010 [65]PolandProspectiveP and A102EBVNRMultivariateModerate
Juvonen, 2007 [66]FinlandRetrospectiveA406EBVNRMultivariateStrong
Kalra, 2018 [67]CanadaRetrospectiveP and A554PTLD509 daysMultivariateStrong
Kullberg-Lindh, 2015 [68]SwedenRetrospectiveP47EBVNRMultivariateStrong
Laberko, 2017 [69]RussiaRetrospectiveP182EBV27 monthsMultivariateStrong
Landgren, 2009 [70]CIBMTRMulticenter retrospectiveP and A26,901PTLD>12 monthsMultivariateStrong
Li, 2018 [71]ChinaRetrospectiveP62EBV 32.5 (0.5–132) monthsUnivariateWeak
Lin, 2019 [72]ChinaMulticenter randomized studyP and A408EBVNRMultivariateStrong
Liu, 2020 [73]ChinaProspectiveA170EBVNRMultivariateStrong
Liu, 2020 [73]ChinaProspectiveA170PTLDNRUnivariateWeak
Liu, 2013 [74]ChinaProspectiveP and A251 fEBV327 (27–1408) daysMultivariateStrong
Liu, 2013 [27]ChinaProspectiveP and A172EBV495 (45–1158) daysMultivariateStrong
Liu, 2013 [27]ChinaProspectiveP and A172PTLD495 (45–1158) daysMultivariateStrong
Liu, 2018 [75]ChinaProspectiveA132EBVNRUnivariate Strong
Marinho-Dias, 2019 [76]PortugalProspectiveP and A40EBV>120 daysMultivariateStrong
Meijer, 2004 [77]NetherlandsProspectiveA78 gEBV(6–32) monthsUnivariateWeak
Mountjoy, 2020 [78]USARetrospectiveA209EBVNon-ATG group
677 (7–3147) days
ATG group
504 (33–2156) days
UnivariateWeak
Neumann, 2018 [79]GermanyCase–controlA44EBV NRUnivariate §Strong
Nowak, 2019 [80]PolandRetrospectiveP and A239EBV2.1 (0.2–67.8) monthsUnivariateWeak
Omar, 2009 [81]SwedenProspectiveP and A131EBVNRMultivariateModerate
Pagliuca, 2019 [82]FranceRetrospectiveP and A208PTLD47.33 (3.18–126.20) monthsMultivariateStrong
Park, 2020 [83]KoreaRetrospectiveP and A114EBVNRUnivariateWeak
Patriarca, 2013 [4]ItalyProspectiveA100 hEBV7 (2–36) monthsMultivariateStrong
Peric, 2012 [84]FranceRetrospectiveA33EBV468 (92–1277) daysUnivariateWeak
Peric, 2011 [85]FranceRetrospectiveA175EBV655 (92–1542) daysMultivariateStrong
Ru, 2020 [86]ChinaRetrospectiveP and A890EBVNRMultivariateStrong
Rustia, 2016 [87]USARetrospectiveP140EBVNRUnivariateWeak
Sanz, 2014 [88]SpainRetrospectiveP and A288EBV>6 monthsMultivariateStrong
Sanz, 2014 [88]SpainRetrospectiveP and A288PTLD>6 monthsMultivariateStrong
Sirvent-von Bueltzingsloewen, 2002 [89]FranceMulticenter prospectiveP and A85 iEBV306 (26–867) daysMultivariateStrong
Styczynski, 2013 [90]EBMTMulticenter retrospectiveP and A4466PTLDNRUnivariateWeak
Torre-Cisneros, 2004 [91]SpainProspectiveP and A100 jEBVNRMultivariateModerate
Trottier, 2012 [92]CanadaRetrospectiveP238EBVNRMultivariateModerate
Tsoumakas, 2019 [93]GreeceProspectiveP110EBV≥1 yearMultivariateStrong
Uhlin, 2014 [94]SwedenRetrospectiveP and A1021PTLDNRMultivariateStrong
Van der Velden, 2013 [95]NetherlandsRetrospectiveA273EBV/PTLD≥6 monthsMultivariateModerate
Van Esser, 2001 [96]Italy, Germany, NetherlandsMulticenter prospectiveP and A152EBVNRMultivariateStrong
Van Esser, 2001 [96]Italy, Germany, NetherlandsMulticenter prospectiveP and A152PTLDNRMultivariateStrong
Wang, 2019 [97]ChinaRetrospectiveP and A186EBVNRMultivariateStrong
Xu, 2015 [98]ChinaCase–controlP and A180PTLDNRMultivariateStrong
Xuan, 2012 [99]ChinaProspectiveP and A185EBV319 (27–1194) daysMultivariateStrong
Xuan, 2013 [16]ChinaProspectiveP and A263PTLD374 (27–1554) daysMultivariateStrong
Yu, 2019 [100]ChinaProspectiveP and A90EBVNRMultivariateModerate
Zallio, 2013 [24]ItalyProspectiveA100EBVNRMultivariateModerate
Zhou, 2020 [101]ChinaRetrospectiveP and A131EBV59.2 (range: 2.03–113.8) monthsMultivariateStrong
Zhou, 2020 [102]ChinaRetrospectiveP and A160PTLD64.7 (range: 2.03–113.8) monthsUnivariateWeak
a Alemtuzumab was considered in the conditioning protocol of all patients, and only patients with at least 6 months of follow-up were considered. b Almost all patients received the standard conditioning regimen. c All of these patients had positive EBV serology, survived beyond 40 days and received cyclosporine beyond 30 days post-transplant. d Of the 40 patients, five were excluded: three because of related early transplant mortality and two dues to relapse before 60 days of follow-up. e All patients in the study had positive CMV serology and negative PCR tests for herpesviruses (EBV, CMV, and HHV-6) one week after transplantation. f All patients had a negative EBV PCR test at the start of follow-up. g All except 1 (receiving bone marrow) received a peripheral blood stem cell graft. h All patients had a follow-up duration > 30 days post-transplant. i Five patients with post-transplant lymphoproliferative syndrome were excluded. Analysis of risk factors for EBV reactivation involved 80 patients. j All patients had positive EBV serology before transplantation. § The individuals were matched according to the variables age, diagnosis, and conditioning regimen. Chi 2 test and Mann–Whitney U test were used to verify that the distributions of potential confounding factors were not significantly different. The outcome has not been explicitly defined. Abbreviation: ATG: anti-thymocyte globulin; CIBMTR: Center for International Blood and Marrow Transplant Research; EBV: Epstein–Barr virus; EMBT: European Group for Blood and Marrow Transplantation; NR: not reported; P: pediatrics; P and A: pediatrics and adults; PTLD: post-transplant lymphoproliferative disease.
Table 2. Summary of risk factors for post-transplant EBV infection and for PTLD in the studies using multivariate analysis.
Table 2. Summary of risk factors for post-transplant EBV infection and for PTLD in the studies using multivariate analysis.
First Author, YearOutcomeStudy PopulationRisk FactorsEstimate (95% CI); p-Value *
Recipient age
Bogunia-Kubik, 2007 [35]EBVP and A> vs. ≤25 yearsOR = 1.54 (1.136–2.703); p = 0.034
Ru, 2020 [86]EBVP and A<30 vs. ≥30 yearsHR = 1.041 (0.763–1.420); p = 0.799
Düver, 2020 [51]EBVPAge (continuous)OR = 1.08 (1.00–1.17); p = 0.057
Kullberg-Lindh, 2011 [68]EBVPContinuousSlope = −0.06; p = 0.09
Gao, 2019 [56]PTLDP and A≥40 vs. <40 yearsHR = 0.4 (0.2–0.9); p = 0.032
Landgren, 2009 [70]PTLDP and A≥50 yearsRR = 5.1 (2.8–8.7)
Diagnosis
Burns, 2016 [39]EBVP and ANHL vs. AML/MDSHR = 0.18 (0.05–0.57); p = 0.004
ALL vs. AML/MDSHR = 0.89 (0.45–1.75); p = 0.734
HL vs. AML/MDSHR = 1.63 (0.64–4.16); p = 0.308
CLL vs. AML/MDSHR = 0.87 (0.41–1.85); p = 0.724
MPD vs. AML/MDSHR = 0.95 (0.43–2.11); p = 0.907
Other vs. AML/MDSHR = 3.01 (0.94–9.65); P = 0.063
Carpenter, 2010 [23]EBVP and AHL vs. AMLHR = 3.53 (1.51–8.25); p = 0.004
NHL vs. AMLHR = 0.678 (0.249–1.848); p = 0.448
MPD vs. AMLHR = 2.01 (0.828–4.858); p = 0.123
CLL vs. AMLHR = 3.767 (1.375–10.322); p = 0.01
Other disease vs. AMLHR = 1.449 (0.486–4.319); p = 0.506
Sanz, 2014 [88]EBVP and AHodgkin’s disease vs. other diagnosisSHR = 11.6 (3.4–40.0); p < 0.0001
Zhou, 2020 [101]EBVP and AUnderlying disease (AA vs. AL)HR = 4.369 (0.484–39.451); p = 0.189
Fujimoto, 2019 [55]PTLDP and AALL vs. AML/MDSHR = 1.08 (0.75–1.57); p = 0.68
CML/MPD vs. AML/MDSHR = 1.55 (0.89–2.69); p = 0.12
Lymphoid malignancies vs. AML/MDSHR = 1.33 (0.92–1.92); p = 0.13
AA vs. AML/MDSHR = 5.19 (3.32–8.11); p < 0.001
Others vs. AML/MDSHR = 1.94 (0.97–3.89); p = 0.06
Genotype
Bogunia-Kubik, 2005 [36]EBVP and ARecipient having IFNG 3/3 genotype vs. other IFNGOR = 7.28; p = 0.005
Bogunia-Kubik, 2007 [35]EBVP and APresence of CCR5 deletion mutation (yes vs. no)OR = 0.17 (0.034–0.803); p = 0.026
Pagliuca, 2019 [82]PTLDP and APresence of HLA DRB1*11:01 (yes vs. no)SHR = 4.85 (1.57–14.97); p = 0.006
Recipient, donor EBV, CMV serostatus
Hiwarkar, 2013 [59]EBVPD+ and R+ (CMV or EBV) or host adenoviral infectionSignificant, but NR
Laberko, 2017 [69]EBVP and AEBV D+/R− vs. D+/R+HR = 2.85 (1.12–7.28); p = 0.028
EBV D−/R+ vs. D+/R+HR = 0.32 (0.05–2.0); p = 0.22
EBV D−/R− vs. D+/R+No events
EBV Unknown vs. D+/R+HR = 1.23 (0.53–2.9); p = 0.63
Lin, 2019 [72]EBVP and AD/R EBV serostatus (D−/R+ vs. Other)HR = 1.58 (1.01–2.46); p = 0.046
Uhlin, 2014 [94]PTLDP and AEBV D+ R− vs. OtherSHR = 4.97 (2.30–10.7); p < 0.001
Brunstein, 2006 [38]EBV/PTLDP and ACMV (R− vs. R+)HR = 3.0 (0.9–9.7) p = 0.07
Donor sex
Fan, 2016 [53]EBVP and AMale donorOR = 13.24 (2.006–87.387); p = 0.007
Jaskula, 2010 [65]EBVP and AFemale donorOR = 2.816; p = 0.044
Donor type
Düver, 2020 [51]EBVPUnrelated donor vs. Related donorOR = 5.05 (1.24–20.63); p = 0.024
Marinho-Dias, 2019 [76]EBVP and AUnrelated donor (yes vs. no)HR = 8.8, p = 0.030 at D + 150
Tsoumakas, 2019 [93]EBVPRelated donor vs. unrelated donorHR = 0.38 (0.15–0.98); p = 0.045
Omar, 2009 [81]EBVP and AURD + MMRD vs. HLA-matched donorp = 0.04
Pagliuca, 2019 [82]PTLDP and AUnrelated (yes vs. no)SHR = 2.11 (1.00–4.45); p = 0.051
Fujimoto, 2019 [55]PTLDP and AMMRD vs. MRDHR = 4.39 (2.39–8.07); p < 0.001
MURD vs. MRDHR = 4.08 (2.39–6.99); p < 0.001
MMURD vs. MRDHR = 3.20 (1.58–6.47); p = 0.001
CB vs. MRDHR = 8.03 (4.72–13.7); p < 0.001
Sirvent-von Bueltzingsloewen, 2002 [89]EBVP and AHLA incompatibility (yes vs. no)OR = 5 (1.5–16.4)
Torre-Cisneros, 2004 [91]EBVP and ANo HLA-matched sibling donorHR = 2.1 (0.8–6.2); p = 0.069
Gao, 2019 [56]EBVP and AHaploidentical donors vs. matched sibling donorsHR = 2.0 (0.8–5.1); p = 0.130
Ru, 2020 [86]EBVP and AHLA-haploidentical vs. HLA-identicalHR = 1.830 (1.275–2.627); p = 0.001
Gao, 2019 [56]PTLDP and AHaploidentical donors vs. matched sibling donorsHR = 2.0 (0.5–8.3); p = 0.350
Uhlin, 2014 [94]PTLDP and AHLA mismatch vs. matchSHR = 5.89 (2.43–14.3) p < 0.001
Graft source
Tsoumakas, 2019 [93]EBVPPBSC vs. BMHR = 2.51 (1.04–6.05); p = 0.041
Wang, 2019 [97]EBVP and APB + BM vs. PBHR = 7.89; p = 0.003
BM vs. PBHR = 18.69; p < 0.001
Graft content
Christopeit, 2013 [45]EBVACD3+ (≥ vs. < median)OR = 0.11 (0.02–0.78); p = 0.027
CD3+CD8+ (≥ vs. < median)OR = 0.05 (0.006–0.431); p = 0.007
Van Esser, 2001 [96]EBVP and ACD34+ (>1.35 × 106/kg)HR = 2.6 (1.5–4.6); p = 0.001
Conditioning regimens and GvHD prophylaxis/treatment
Kullberg-Lindh, 2011 [68]EBVPTBI (yes vs. no)Slope = 1.60; p = 0.001
Liu, 2013 [74]EBVP and AIntensified MAC vs. standard MACHR = 1.72 (1.03–2.88); p = 0.038
Lin, 2019 [72]EBVP and AIntensified conditioning vs. standard MACHR = 1.73 (1.18–2.54); p = 0.005
Sanz, 2014 [88]EBVP and ARIC vs. MACSHR = 6.0 (2.0–17.6); p = 0.001
PTLDRIC vs. MACSHR = 5.5 (1.8–17.1); p = 0.003
Fujimoto, 2019 [55]PTLDP and ARIC vs. MACHR = 0.82 (0.60–1.12); p = 0.22
Uhlin, 2014 [94]PTLDP and ARIC vs. no RICSHR = 3.25 (1.53–6.89) p = 0.002
Xuan, 2013 [17]PTLDP and AStandard vs. intensifiedHR = 4.46 (1.20–16.61); p = 0.026
Liu, 2013 [27]PTLDP and AIntensified MAC vs. standard MACp = 0.018
Brunstein, 2006 [38]EBV/PTLDP and ANMAC without ATG vs. MACHR = 0.7 (0.1–6.5); p = 0.51
NMAC with ATG vs. MACHR = 15.4 (2.0–116.1); p < 0.01
Van der Velden, 2013 [95]PTLDAMAC without ATGOR = 2.6 (1.05–7.15); p = 0.01
NMAC with ATGOR = 2.1 (0.92–4.8); p = 0.08
Gao, 2019 [56]PTLDP and AUse of fludarabine (yes vs. no)HR = 3.8 (1.4–10.6); p = 0.010
Cohen, 2005 [46]EBVPATG vs. CampathOR = 2.09 (0.83–5.29)
Cesaro, 2004 [41]EBVPUse of ATG (yes vs. no)HR = 13.0 (2–96); p = 0.01
Düver, 2020 [51]EBVPUse of ATG (yes vs. no)OR = 10.68 (1.15–98.86); p = 0.037
Gao, 2019 [56]EBVP and AUse of ATG (yes vs. no)HR = 6.3 (1.6–24.0); p = 0.008
Kullberg-Lindh, 2011 [68]EBVPUse of ATG (yes vs. no)Slope = 1.34; p = 0.004
Juvonen, 2007 [66]EBVAUse of ATG (yes vs. no) HR = 5.78 (2.47–13.5); p < 0.001
Peric, 2011 [85]EBVAUse of ATG (yes vs. no)SHR = 4.9 (1.1–21.0); p = 0.03
Fan, 2016 [53]EBVP and AUse of ATG (yes vs. no)OR = 7.69 (1.17–50.49); p = 0.034
Laberko, 2017 [69]EBVP and AHorse ATG vs. no serotherapyHR = 2.47 (0.95–6.38); p = 0.063
Rabbit ATG vs. no serotherapyHR = 1.22 (0.467–3.18); p = 0.69
Christopeit, 2013 [45]EBVAUse of ATG (yes vs. no)OR = 0.83 (0.17–4.01); p = 0.820
Liu, 2013 [74]EBVP and AUse of ATG (yes vs. no)HR = 14.08 (6.02–32.92); p < 0.001
Ru, 2020 [86]EBVP and AUse of ATG (yes vs. no)HR = 4.288(2.638–6.97); p < 0.001
Liu, 2013 [27]PTLDP and AUse of ATG (yes vs. no)p = 0.038
Van der Velden, 2013 [95]PTLDAUse of ATG (yes vs. no)OR = 2.4 (1.3–4.2) p = 0.001
Landgren, 2009 [70]PTLDP and AUse of ATG (yes vs. no) RR = 3.8 (2.5–5.8)
Xuan, 2013 [16]PTLDP and AUse of ATG (yes vs. no)HR = 13.03 (1.67–101.58) p = 0.014
Fujimoto, 2019 [55]PTLDP and AUse of ATG in conditioning regimen (yes vs. no)HR = 6.13 (4.33–8.68); p < 0.001
Fujimoto, 2019 [55]PTLDP and AUse of ATG for GvHD treatment (yes vs. no) HR = 2.09 (1.17–3.72); p = 0.01
Gao, 2019 [56]PTLDP and AUse of ATG (yes vs. no)HR = 2.9 (0.3–27.5); p = 0.350
Lin, 2019 [72]EBVP and AATG dose (10.0 mg/kg vs. 7.5 mg/kg)HR = 2.02 (1.37–2.97); p < 0.001
Buyck, 2009 [40]PTLDP and ANumber of prior courses of ATGHR = 7.23 (1.67–31.32); p = 0.008;
Fan, 2016 [53]EBVP and AMMF + CsA + prednisone vs. MMF + CsAOR = 23.68 (1.924–291.449); p = 0.013
Christopeit, 2013 [45]EBVACsA AUC (≥ vs. <6000 ng/mL x days)OR = 6.067 (1.107–33.238); p = 0.038
T-cell depletion
Bordon, 2012 [37]EBVPIn vivo TCD (yes vs. no)p = 0.04
Torre-Cisneros, 2004 [91]EBVP and AUse of CD4+ lymphocyte-depleted graft (yes vs. no)HR = 11.5 (5.8–22.8); p < 0.0001
Van Esser, 2001 [96]EBVP and ATCD without ATG vs. non-TCDHR = 1.5 (0.8–2.9); p = 0.3
TCD with ATG vs. non-TCDHR = 3.4 (1.6–7.1); p = 0.001
Landgren, 2009 [70]PTLDP and ABroad lymphocyte depletion vs. no TCDRR = 3.1 (1.2–6.7)
Selective TCD vs. no TCDRR = 9.4 (6.0–14.7)
Method of T-cell depletion
Landgren, 2009 [70]PTLDP and AAlemtuzumab MoAb vs. no TCDRR = 3.1 (0.7–8.4)
Elutriation/density gradient centrifugation vs. no TCDRR = 3.2 (0.8–8.8)
Anti-T or anti-T + NK MoAb vs. no TCDRR = 8.4 (5.1–13)
SRBC rosetting vs. no TCDRR = 14.6 (5.9–31)
Lectins with/without SRBC or anti-T MoAb vs. no TCDRR = 15.8 (7.2–32)
Unclassified/unknown method vs. no TCDRR = 6.0 (0.96–20)
Graft-versus-host disease
Cohen, 2005 [46]EBVPaGvHD (yes vs. no)OR = 2.20 (2.12–15.08)
Elmahdi, 2016 [52]EBVPaGvHD (yes vs. no)HR = 3.29 (1.26–8.58); p = 0.015
Hiwarkar, 2013 [59]EBVPaGvHD ≥ grade IISignificant, but NR
Kullberg-Lindh, 2011 [68]EBVPcGvHD (yes vs. no)Slope = −1.12; p = 0.023
Juvonen, 2007 [66]EBVAaGvHD ≥ grade IIIHR = 1.70 (1.11–2.62); p = 0.015
Sirvent-von Bueltzingsloewen, 2002 [89]EBVP and AaGvHD ≥ grade IIOR = 3.4 (1.2–9.7)
Omar, 2009 [81]EBVP and AaGvHD (yes vs. no)p = 0.009
Gao, 2019 [56]EBVP and AaGvHD (yes vs. no)HR = 1.0 (0.7–1.6); p = 0.960
Gao, 2019 [56]PTLDP and AaGvHD (yes vs. no)HR = 1.4 (0.5–3.8); p = 0.480
Laberko, 2017 [69]EBVP and AGvHD (yes vs. no)HR = 1.97 (1.04–3.72); p = 0.037
Landgren, 2009 [70]PTLDP and AaGvHD ≥ grade II RR = 1.7 (1.2–2.5)
Ru, 2020 [86]EBVP and AaGvHD (grade II-IV vs. none or grade I)HR = 1.26 (0.89–1.78); p = 0.193
Fujimoto, 2019 [55]PTLDP and AaGvHD grade II-IV (yes vs. no) HR = 1.93 (1.48–2.52); p < 0.001
Uhlin, 2014 [94]PTLDP and AaGvHD ≥ grade IISHR = 2.65 (1.32–5.35) p = 0.006
Landgren, 2009 [70]PTLDP and AcGvHD moderate/severe or clinical extensive RR = 2.0 (1.1–3.2)
Ru, 2020 [86]EBVP and AcGvHD (yes vs. no)HR = 1.413 (1.013–1.971); p = 0.042
Kalra, 2018 [67]PTLDP and AaGvHD grade II-IV or chronic NST (yes vs. no)SHR = 0.47, p = 0.04
Immunological reconstitution
Patriarca, 2013 [4]EBVAPeripheral blood CD4+ lymphocyte/µL at +1 month after HSCT (≥50 vs. <50)OR = 0.1 (0.02–0.48); p = 0.004
Yu, 2019 [100]EBVP and ANKp30 in 1-month post-transplant (1 M) (% of total NK cells)HR = 0.957 (0.918–0.998); p = 0.04
Liu, 2020 [73]EBVAVδ2+ cell recovery at day 30 post-transplantationHR = 0.347 (0.161–0.747); p = 0.007
Liu, 2020 [73]EBVACD8+ cell recovery at day 30 post-transplantationHR = 0.499 (0.207–1.201); p = 0.121
Xu, 2015 [98]PTLDP and ACD8+ cell count at day 30 after HSCT (≥median vs. < median)HR = 0.34 (0.13–0.92) p = 0.033
PTLDP and AIgM count at day 30 after HSCT (≥median vs. <median)HR = 0.27 (0.10–0.75) p = 0.012
CMV reactivation
Gao, 2019 [56]EBVP and ACMV DNAemia (yes vs. no)HR = 5.9 (2.5–13.9); p < 0.001
Torre-Cisneros, 2004 [91]EBVP and ACMV load > 2500 copies/mLHR = 2.1 (0.9–7); p = 0.061
Zallio, 2013 [24]EBVAyes vs. noSignificant, but NR
Zhou, 2020 [101]EBVP and ACMV DNAemia (yes vs. no)HR = 97.754 (9.477–1008.304)
Gao, 2019 [56]PTLDP and ACMV DNAemia (yes vs. no)HR = 11.6 (1.2–114.4); p = 0.036
Xu, 2015 [98]PTLDP and ACMV DNAemia (yes vs. no)HR = 5.68 (1.17–27.57) p = 0.031
Transfusion
Trottier, 2012 [92]EBVPRBC transfusion volume (mL)<850 vs. 0HR = 1.99 (0.47–8.44)p-value trend = 0.047
850–1890 vs. 0HR = 2.40 (0.56–10.24)
>1890 vs. 0HR = 2.86 (0.68–12.11)
PFFP transfusion volume (mL)≤200 vs. 0HR = 0.70 (0.22–2.25)p-value trend = 0.079
>200 vs. 0HR = 3.16 (1.00–11.17)
PPLT transfusion volume (mL)1260–2530 vs. <1260HR = 1.65 (0.86–3.18)p-value trend = 0.012
>2530 vs. <1260HR = 2.19 (1.21–3.97)
Other factors
Garcia-Cadenas, 2015 [57]EBVAPrior SCT (yes vs. no)HR: 2.6 (1.1–6.4); p = 0.04
PTLDAPrior SCT (yes vs. no)HR: 6.4 (1.3–31.9); p = 0.02
Fujimoto, 2019 [55]PTLDP and ANumber of allogeneic HSCT (two or more vs. one)HR = 1.50 (1.05–2.15); p = 0.03
Landgren, 2009 [70]PTLDP and ASecond transplant (yes vs. no) RR = 3.5 (1.7–6.3)
Uhlin, 2014 [94]PTLDP and ASplenectomy (yes vs. no)SHR = 4.81 (1.51–15.4) p = 0.008
PTLDP and AMSC treatment (yes vs. no)SHR = 3.05 (1.25–7.48) p = 0.015
Landgren, 2009 [70]PTLDP and A2+ HLA MMRD or URD, no ATG, no selective TCD vs. matched sibling or 1 HLA-Ag mismatched relativeRR = 0.9 (0.3–2.2)
2+ HLA MMRD or URD, ATG and/or selective TCD vs. matched sibling or 1 HLA-Ag mismatched relativeRR = 3.8 (2.4–6.1)
Van Esser, 2001 [96]PTLDP and AA stepwise increase of EBV-DNA by 1 logHR = 2.9 (1.7–4.8); p < 0.001
Pagliuca, 2019 [82]PTLDP and AFever at onset of EBV infection (yes vs. no)SHR = 6.12 (1.74–21.58); p = 0.005
Fujimoto, 2019 [55]PTLDP and AYear of HSCT (2010–2015 vs. 1990–2009)HR = 1.87 (1.38–2.52); p < 0.001
Abbreviations: A: adults; Ag: antigen; aGvHD: acute graft-versus-host disease; ALL: acute lymphocytic leukemia; AML: acute myeloid leukemia; ATG: anti-thymocyte globulin; AUC: area under curve; BM: bone marrow; CB: cord blood; CCR5: C–C chemokine receptor 5; cGvHD: chronic graft-versus-host disease; CI: confidence interval; CLL: chronic lymphocytic leukemia; CMV: cytomegalovirus; CsA: cyclosporine A; D+: donor positive; D−: donor negative; D/R: donor/recipient; EBV: Epstein–Barr virus; FFP: fresh-frozen plasma; GvHD: graft-versus-host disease; HL: Hodgkin’s lymphoma; HLA: human leukocyte antigen; HR: hazard ratio; HSCT: hematopoietic stem cell transplantation; IFNG: interferon-γ gene; MAC: myeloablative conditioning; MDS: myelodysplastic syndrome; MMF: mycophenolate mofetil; MMRD: mismatched related donor; MMUD: mismatched unrelated donor; MoAb: monoclonal antibody; MPD: myeloproliferative disease; MRD: matched related donor; MSC: mesenchymal stromal cells; MURD: matched unrelated donor; NHL: non-Hodgkin’s lymphoproliferative disease; NK: natural killer cells; NMAC: non-myeloablative conditioning; NR: not reported; NST: needing systemic therapy; OR: odds ratio; P: pediatric; P and A: pediatric and adult; PB: peripheral blood; PBSC: peripheral blood stem cells; PTLD: post-transplant lymphoproliferative disease; PLT: platelets; R+: recipient positive; R−: recipient negative; RBC: red blood cell; RIC: reduced-intensity conditioning; RR: relative risk; SCT: stem cell transplant; SHR: subhazard ratio; SRBC: sheep red blood cell; TBI: total body irradiation; TCD: T-cell depletion; URD: unrelated donor; vs.: versus. Time-dependent covariate. * Statistically significant associations are shown in bold.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Enok Bonong, P.R.; Zahreddine, M.; Buteau, C.; Duval, M.; Laporte, L.; Lacroix, J.; Alfieri, C.; Trottier, H. Factors Associated with Post-Transplant Active Epstein-Barr Virus Infection and Lymphoproliferative Disease in Hematopoietic Stem Cell Transplant Recipients: A Systematic Review and Meta-Analysis. Vaccines 2021, 9, 288. https://doi.org/10.3390/vaccines9030288

AMA Style

Enok Bonong PR, Zahreddine M, Buteau C, Duval M, Laporte L, Lacroix J, Alfieri C, Trottier H. Factors Associated with Post-Transplant Active Epstein-Barr Virus Infection and Lymphoproliferative Disease in Hematopoietic Stem Cell Transplant Recipients: A Systematic Review and Meta-Analysis. Vaccines. 2021; 9(3):288. https://doi.org/10.3390/vaccines9030288

Chicago/Turabian Style

Enok Bonong, Pascal Roland, Monica Zahreddine, Chantal Buteau, Michel Duval, Louise Laporte, Jacques Lacroix, Caroline Alfieri, and Helen Trottier. 2021. "Factors Associated with Post-Transplant Active Epstein-Barr Virus Infection and Lymphoproliferative Disease in Hematopoietic Stem Cell Transplant Recipients: A Systematic Review and Meta-Analysis" Vaccines 9, no. 3: 288. https://doi.org/10.3390/vaccines9030288

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop