Next Article in Journal
Skeletal Morphogenesis and Anomalies in Gilthead Seabream: A Comprehensive Review
Next Article in Special Issue
An Overview of Recent Developments in the Management of Burn Injuries
Previous Article in Journal
A Weakly Supervised Learning Method for Cell Detection and Tracking Using Incomplete Initial Annotations
Previous Article in Special Issue
AML under the Scope: Current Strategies and Treatment Involving FLT3 Inhibitors and Venetoclax-Based Regimens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Pre-Emptive Use of Rituximab in Epstein–Barr Virus Reactivation: Incidence, Predictive Factors, Monitoring, and Outcomes

1
Hematology Department, BMT Unit, General Hospital “George Papanicolaou”, 57010 Thessaloniki, Greece
2
2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(22), 16029; https://doi.org/10.3390/ijms242216029
Submission received: 6 September 2023 / Revised: 30 October 2023 / Accepted: 3 November 2023 / Published: 7 November 2023

Abstract

:
Post-transplant lymphoproliferative disease (PTLD) is a fatal complication of hematopoietic cell transplantation (HCT) associated with the Epstein–Barr virus (EBV). Multiple factors such as transplant type, graft-versus-host disease (GVHD), human leukocyte antigens (HLA) mismatch, patient age, and T-lymphocyte-depleting treatments increase the risk of PTLD. EBV reactivation in hematopoietic cell transplant recipients is monitored through periodic quantitative polymerase chain reaction (Q-PCR) tests. However, substantial uncertainty persists regarding the clinically significant EBV levels for these patients. Guidelines recommend initiating EBV monitoring no later than four weeks post-HCT and conducting it weekly. Pre-emptive therapies, such as the reduction of immunosuppressive therapy and the administration of rituximab to treat EBV viral loads are also suggested. In this study, we investigated the occurrence of EBV-PTLD in 546 HCT recipients, focusing on the clinical manifestations and risk factors associated with the disease. We managed to identify 67,150 viral genomic copies/mL as the cutoff point for predicting PTLD, with 80% sensitivity and specificity. Among our cohort, only 1% of the patients presented PTLD. Anti-thymocyte globulin (ATG) and GVHD were independently associated with lower survival rates and higher treatment-related mortality. According to our findings, prophylactic measures including regular monitoring, pre-emptive therapy, and supportive treatment against infections can be effective in preventing EBV-related complications. This study also recommends conducting EBV monitoring at regular intervals, initiating pre-emptive therapy when viral load increases, and identifying factors that increase the risk of PTLD. Our study stresses the importance of frequent and careful follow-ups of post-transplant complications and early intervention in order to improve survival rates and reduce mortality.

1. Introduction

Post-transplant lymphoproliferative disease (PTLD) is one of the most fatal complications of hematopoietic cell transplantation (HCT) associated with the Epstein–Barr virus (EBV) in immunocompromised individuals [1,2]. EBV is a ubiquitous herpesvirus that infects over 90% of adults and 50–89% of children worldwide [1,3,4,5,6]. After infecting B-lymphocytes, EBV extends their lifespan, thereby increasing the likelihood of mutations. These mutations can involve alterations in BCL6 and MYC expression, the activation of signaling pathways such as NF-kB, BCL2, PI3K/AKT/mTOR, and changes in immunoglobulin switching. These mutations may lead to stepwise advancement from early lesions to polymorphic PTLD and then to clonally expanded monoclonal ones [7,8,9,10,11]. Regarding solid organ transplant recipients, more than 90% of EBV-PTLD cases are attributed to the host, whereas in HCT, the majority of EBV-PTLD cases are of donor origin [12]. PTLD cases ought to be categorized utilizing the tumor classification system for hematopoietic and lymphoid tissues of the World Health Organization 2022 [13]. Hyperplasia, polymorphic or monomorphic post-transplant lymphoproliferative disorders, and classic Hodgkin lymphoma are categorized as “Lymphoid proliferations and lymphomas associated with immune deficiency and dysregulation”. The incidence of PTLD after allogeneic hematopoietic stem cell transplantation is reported to vary between 0.2% and 17% [14,15]. The majority (60%) of cases develop within the first year post-transplantation [14].
Risk factors that have been identified include the transplant type, acute and chronic graft-versus-host disease (GVHD, reduced intensified conditioning (RIC), human leukocyte antigens (HLA)-mismatch, EBV seromismatch (R−/D+), patient age, ex vivo and in vivo T-lymphocyte-depleting treatments (anti-thymocyte globulin (ATG), primary immunodeficiency disorders, splenectomy, and cytomegalovirus reactivation [2,14,16,17,18,19,20,21,22,23,24]. The incidence of PTLD is on the rise, due to the expansion of the types of transplantation like haploidentical HCT [25].
PTLD manifests with persistent fever, lethargy, anorexia, signs of bowel perforation, tonsillar enlargement and/or inflammation, symptomatic hepatosplenomegaly, subcutaneous nodules, and gradual decline in peripheral blood cell counts [1,15,26]. Furthermore, patients with PTLD may exhibit headaches or focal neurological deficits [27].
The quantification of viral load is mostly performed via quantitative polymerase chain reaction (Q-PCR); however, there is no agreement on the EBV threshold that should trigger further investigations and pre-emptive therapy [28,29]. This lack of consensus can be attributed to varying sample materials and the absence of standardized PCR techniques [30,31]. In the absence of universally accepted laboratory assays [32,33,34], some researchers working with plasma samples suggest either a threshold of 1000 EBV copies/mL on two consecutive occasions, or 10,000 EBV copies/mL on one sample [22,35]. When whole blood samples are examined, a cutoff of 40,000 EBV copies/mL has been proposed [31,36,37,38], given that EBV load in plasma is 10- to 100-fold lower than in whole blood samples [39]. Guidelines from the European Conference on Infections in Leukemia (ECIL-6) advise that EBV monitoring should be initiated no later than four weeks post-HCT and occur at least once weekly, until cellular immunity has been reconstituted [31].
The types of pre-emptive therapies which can be regularly employed are the reduction of immunosuppressive therapy [40] and the administration of an anti-CD20 monoclonal antibody, rituximab. The use of rituximab resulted in better short-term outcomes in PTLD [41,42,43,44,45,46,47,48,49,50], although concerns exist regarding its long-term side effects. These side effects include B-cell suppression, hypogammaglobulinemia, progressive multifocal leukoencephalopathy, and increased susceptibility to infections [51,52,53,54,55]. The pre-emptive use of rituximab is recommended by American and European guidelines in order to treat rising EBV viral loads in peripheral blood without clinical symptoms, before the diagnosis of EBV-PTLD. However, pre-emptive therapy can lead to the administration of rituximab to patients who would not have developed PTLD. This exposes patients to the potential toxicities associated with the use of this treatment, such as cytopenias and infections.
Furthermore, noteworthy advancements have been achieved in the field of adoptive immunotherapies for treating PTLD, particularly regarding in vitro autologous donor T-cells, which is practiced as a clinical study in our center, or HLA-matched banked third-party donor polyclonal EBV-specific cytotoxic T-cells (EBVSTs) [56,57,58,59,60,61]. Additionally, chimeric antigen receptor T-cell (CAR-T) therapy has been proven effective in certain cases [62,63,64,65]. Although it has shown some promising outcomes in HCT-PTLD [66], its utilization is limited by the need for immunosuppressive regimens during the initial stages after HCT, which is when HCT-PTLD generally develops. This restriction may impair the effectiveness of CAR-T therapy for treating HCT-PTLD. Moreover, agents like lenalidomide [67,68] and bortezomib [69,70] have demonstrated remarkable outcomes. However, the availability of these treatment methods is often limited [31], despite recent advances [56].
Therefore, it is crucial to investigate approaches to prevent, pre-emptively treat, and eliminate the disease once it manifests. Nonetheless, due to the low incidence of PTLD, there have been limited clinical trials examining such approaches. This article seeks to investigate the occurrence of EBV-PTLD in HCT recipients, focusing on the clinical manifestations and risk factors associated with the disease.

2. Results

Out of 546 recipients of hematopoietic cell transplants, clinically relevant EBV reactivation occurred in 100 patients (18%). The majority of these cases had hematologic malignancy (n = 98) while 2 had aplastic anemia. Graft sources included matched sibling (n = 20), unrelated (n = 68), or haploidentical donors (n = 12) (Figure 1). Haploidentical donors were significantly higher in patients with clinically relevant EBV reactivation compared to our transplant population (12% versus 6%, p < 0.001).
The majority (88/100) of patients received myeloablative conditioning, while 12 received reduced-intensity conditioning. The median time to detection of clinically relevant EBV reactivation was 65 (range 20–2970) days post-transplant, with a median viral load of 24,500 viral genome copies (VGC)/mL (range 8690–2,670,000 VGC/mL) (Supplementary Figures S1 and S3 and Supplementary File entitled “Patients-Viral loads”). Pre-emptive rituximab was administered to all 74 patients at a median of 4 days post-EBV reactivation, as soon as the reactivation was detected. The majority, 63 out of 74, received one cycle, until the EBV load became undetectable. The number of rituximab cycles (median 1, range 1–3) were not associated with survival outcomes.
Relapse of clinically relevant EBV reactivation occurred in 13 out of 100 patients, with a higher incidence among those with delayed resolution of infection (27 versus 14 days in non-relapsed patients, p < 0.01). Late-onset neutropenia related to rituximab was noted in 16 out of 74 patients and significantly correlated with increased EBV loads. Multivariate analysis confirmed that haploidentical donors (p = 0.001), use of ATG (p < 0.001) and rituximab-related late-onset neutropenia (p < 0.001) were independently associated with increased viral loads. Concurrent cytomegalovirus (CMV) reactivation occurred in 47 patients, with significantly delayed resolution of EBV infections noted in patients receiving pre-emptive anti-CMV treatments, indicating greater immunosuppression.
Five patients (two with haploidentical and three with unrelated donors) developed PTLD at 41 days post-transplantation. The patients presented fever (5/5), anorexia (5/5), lymphadenopathy (4/5), hepatosplenomegaly (3/5), gradual decline in peripheral blood counts (4/5), and focal neurological deficits (1/5). Receiver operating characteristic (ROC) curve analysis identified a cutoff of 67,150 VGC/mL that predicted PTLD with 80% sensitivity and specificity (see green line in Figure 2).
Relapse-free survival (RFS), overall survival (OS), and treatment-related mortality (TRM) (Supplementary Figures S2–S5) in the entire cohort were similar regardless of EBV viral load (<50,000 VGC/mL) or PTLD [4-year RFS 32.2%; 4-year OS was 48.1% with a median follow-up of 29 months (4–216)]. Multivariate analysis revealed that ATG and chronic GVHD were independently associated with OS (p < 0.001, p < 0.05, respectively), while ATG, chronic GVHD and age at transplant were independently associated with higher TRM (HR: 0.1, 1.16, 1.03, 95%CI: 0.15–0.5, 0.008–1.16, 1.007–1.05, respectively, p < 0.05). A trend for higher TRM was also noted among patients with EBV loads higher than 50,000 VGC/mL.

3. Discussion

In the present study, we investigated the incidence, clinical significance, and relapse rate of EBV reactivation in a large cohort of HCT recipients. With the median time to detection being 65 days post-transplantation, 18% of patients had clinically relevant EBV reactivation. By multivariate analysis, we managed to associate multiple risk factors (late-onset neutropenia, ATG, and haploidentical donors) with increased viral loads. Additionally, CMV coincidental reactivation and subsequent antiviral treatment were correlated with the delayed resolution of EBV infections, most likely due to greater immunosuppression.
In our cohort, PTLD was observed in approximately 1% of patients, a percentage within range of the previously published literature [14,15,17,50]. Notably, this rate was particularly lower than in other studies, where pre-emptive treatment with rituximab was either not a standard of care [71] or utilized a higher viral load threshold for initiating therapy [72]. There was no significant difference in the studied outcomes between responders and non-responders. By ROC curve analysis, we identified a 67,150 viral genomic copies/mL threshold with high sensitivity and specificity for predicting PTLD. Considering the variability observed in the threshold ranges within the literature (ranging from 1000 to 40,000 EBV copies/mL) and depending on the center-specific cutoff values, the application of this threshold has the potential to reduce the unnecessary administration of rituximab and its associated adverse effects in a significant number of patients. In a similar setting, a recent study reported a decreased EBV-DNA clinically significant threshold, but with lower sensitivity and specificity [73].
Considering rituximab’s detrimental effects, we identified late-onset increased neutropenia in 16 patients receiving therapy, leading to increased immunosuppression. Correlation with increased viral loads was observed as expected. Cycles of treatment did not seem to be associated with survival outcomes. Our study aligns with current guidelines [31] that recommend a single infusion, as most patients required only one cycle of rituximab until an undetectable EBV load. Despite concerns about the effects of rituximab on immune reconstitution, a study of 319 consecutive allo-HCTs [52] found that short- and long-term survival were not inferior in patients who received rituximab. This underscores the effectiveness of pre-emptive rituximab, in agreement with our study’s results.
Relapse-free survival, overall survival and treatment-related mortality in the entire cohort were similar regardless of EBV viral load (<50,000 VGC/mL) or PTLD, in agreement with other studies reporting similar results [42,72,74,75]. For instance, Raberahona et al. reported that blood EBV viral load is frequently detectable after HCT but suggests no strong association with survival [42]. Solano et al. found no significant difference in the initial plasma EBV-DNA load among episodes of self-resolving EBV DNAemia, those requiring rituximab treatment or those leading to PTLDs [75]. Moreover, Duver et al. stated that the overall survival of patients with or without viral infections did not differ significantly [74]. However, in our study, ATG and chronic GVHD were independently associated with OS. Finally, ATG, chronic GVHD, age at transplant, and EBV loads above 50,000 VGC/mL were linked with higher TRM, confirming their role as risk factors.
Our study is limited by its single-center retrospective nature. Nevertheless, its strengths include standard operating procedures (SOPs) in a large patient population reflecting international experience. Another limitation, the use of VCG/mL instead of IU/mL as suggested by recent ECIL guidelines for the quantification of EBV-DNA viral loads, has been overcome by the routine use of standard commercial kit for all the patients [31].
As demonstrated in our study, regular monitoring and the use of pre-emptive therapy are effective strategies for preventing EBV-related complications, given its rapid and aggressive nature, especially in immunocompromised individuals. A useful cutoff point (67,150 VGC/mL) for the prevention of PTLD has been identified, with 80% specificity and sensitivity. Our study adds valuable data to the relevant literature, helping to address the significant uncertainty around the clinically important EBV levels in these patients. Our findings suggest that our diagnostic and management approach has led to acceptable survival rates in patients with PTLD.
However, our findings can be used by other laboratories as a protocol and not as absolute values. For PLTD, the diagnosis remains clinical. Therefore, our suggestion is that these levels should serve as alerts to clinicians for an extensive investigation.

4. Materials and Methods

4.1. Study Population

In this retrospective study, we investigated the molecular clinical features of EBV reactivation in patients who underwent hematopoietic cell transplantation at our JACIE (Joint Accreditation Committee-ISCT & EBMT) accredited unit between 2007 and 2019 (Table 1).

4.2. Conditioning Regimens—Monitoring

All patients with unrelated or haploidentical donors received rabbit ATG, as previously described [76,77]. To avoid reactions to ATG, methylprednisolone was used on the ATG infusion days. The pre-emptive administration of rituximab was the standard of care in our unit during this period. Treatment with rituximab was received at a scheme determined by the physician according to the dynamics of the infection. The assessment and grading of acute GVHD were performed according to the criteria of Glucksberg et al. [78], while chronic GVHD was assessed and graded according to the 2014 National Health Institute criteria [79,80]. In addition, prophylactic granulocyte-colony stimulating factor (GCSF) was used in all regimens after transplantation. Supportive care also comprised prophylactic platelets transfusion if platelet counts decreased to <20 × 109/L or prophylactic red blood cells transfusion if hemoglobin levels decreased to <8 g/dL. All patients received supportive treatment against bacterial, fungal, and viral infections. Trimethoprim-sulfamethoxazole was used as prophylaxis for Pneumocystis jirovecii infection. We measured EBV-DNA every 15 days over a minimum period of 12 months, or longer if immunosuppression was present, in all patients. DNA was extracted from whole blood using a QIAamp® DNA Mini Kit (Qiagen, Hilden, Germany) and EBV- DNA was measured by an artus® EBV RG PCR Kit (Qiagen, Hilden, Germany) on a Rotor-Gene Q MDx Instrument (Qiagen, Hilden, Germany). The analytical detection limit of the assay is 1.02 copies/µL. Clinically relevant EBV reactivation was characterized by the presence of >8500 viral genomic copies (VGC)/mL in whole blood, which was documented during regular molecular monitoring using quantitative real-time PCR. Recurrent infections have been treated with the same protocol. We considered undetectable levels of EBV as indicative of the resolution of infection. Patients with PTLD, confirmed by lymph node biopsy, were managed according to international standards.

4.3. Statistical Analysis

Data analysis was carried out using the statistical program SPSS 22.0 (IBM SPSS Statistics for Windows, Version 22.0., Armonk, NY, USA). Descriptive statistics were performed using the median and range for continuous variables and frequency for categorical variables. Continuous variables were assessed for normality and compared using a Mann–Whitney-test or t-test. The following factors were studied: age, type of disease/donor/graft, infections, graft-versus-host disease, rituximab cycles, late-onset neutropenia, ATG conditioning, treatment-related mortality, overall, and relapse-free survival. Categorical variables were compared using the chi-square test. The Kaplan–Meier method was used for performing survival analysis, and a log-rank test was utilized for comparison of survival curves. Cox regression analysis was conducted to identify univariate and multivariate predictors of survival, with time-dependent covariates computed through SPSS analysis. Statistical significance was assessed by the Gray test and Fine and Gray regression modeling. The level of statistical significance was defined at 0.05.

5. Conclusions

Our study investigated the incidence, clinical significance and risk factors associated with EBV reactivation in a substantial cohort of HCT recipients. 18% of the patients experienced clinically relevant EBV reactivation around 65 days post-transplantation. Multivariate analysis allowed us to identify crucial risk factors such as ATG administration, late-onset neutropenia, and haploidentical donor grafts, which all correlated with elevated viral loads. The observed PTLD incidence of (1%) aligns with the previous literature. Furthermore, we proposed a practical threshold of 67,150 VGC/mL with high sensitivity and specificity for PTLD prediction in an attempt to address the uncertainty regarding clinically significant EBV levels in these patients. Despite the inherent limitations, our findings underscore the significance of regular monitoring and pre-emptive therapy for managing EBV-related complications, particularly within the context of immunocompromised individuals.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms242216029/s1.

Author Contributions

Conceptualization, A.P. and E.G.; data curation, E.G. and N.K.; methodology, E.G., A.V., C.D., N.C., T.T., P.Z., F.K., M.I., I.B., D.M., E.Y., A.A. and I.S.; project administration, A.P. and E.G.; supervision, A.P.; validation, E.G.; visualization, E.G. and N.K.; writing—original draft, E.G. and N.K.; writing—review and editing, A.P., E.Y. and A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no funding.

Institutional Review Board Statement

The study was approved by the Institutional Review Boards of the referral centers and conducted in accordance with the Declaration of Helsinki.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data will be immediately available upon reasonable request.

Acknowledgments

We would like to thank Margarita Sakellari for proof-reading our manuscript.

Conflicts of Interest

E.G. is supported by the ASH Global Research Award and has consulted for Omeros Cooperation. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article, or the decision to submit it for publication.

References

  1. Heslop, H.E. How I treat EBV lymphoproliferation. Blood 2009, 114, 4002–4008. [Google Scholar] [CrossRef]
  2. Sundin, M.; Le Blanc, K.; Ringdén, 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] [PubMed]
  3. Cohen, J.I. Epstein–Barr Virus Infection. N. Engl. J. Med. 2000, 343, 481–492. [Google Scholar] [CrossRef] [PubMed]
  4. Balfour, H.H.; Dunmire, S.K.; Hogquist, K.A. Infectious mononucleosis. Clin. Transl. Immunol. 2015, 4, e33. [Google Scholar] [CrossRef] [PubMed]
  5. Young, L.S.; Rickinson, A.B. Epstein–Barr virus: 40 years on. Nat. Rev. Cancer 2004, 4, 757–768. [Google Scholar] [CrossRef]
  6. Kieff, E.; Rickinson, A.B. Epstein-Barr Virus and Its Replication. In Fields Virology; Knipe, D.M., Howley, P.M., Eds.; Lippinkott-Williams & Wilkins: Philadelphia, PA, USA, 2007; Volume 2, pp. 2603–2654. [Google Scholar]
  7. Vereide, D.T.; Sugden, B. Lymphomas differ in their dependence on Epstein-Barr virus. Blood 2011, 117, 1977–1985. [Google Scholar] [CrossRef] [PubMed]
  8. Burns, D.M.; Tierney, R.; Shannon-Lowe, C.; Croudace, J.; Inman, C.; Abbotts, B.; Nagra, S.; Fox, C.P.; Chaganti, S.; Craddock, C.F.; et al. Memory B-cell reconstitution following allogeneic hematopoietic stem cell transplantation is an EBV-associated transformation event. Blood 2015, 126, 2665–2675. [Google Scholar] [CrossRef]
  9. Krams, S.M.; Martinez, O.M. Epstein–Barr virus, rapamycin, and host immune responses. Curr. Opin. Organ Transplant. 2008, 13, 563–568. [Google Scholar] [CrossRef]
  10. Capello, D.; Rossi, D.; Gaidano, G. Post-transplant lymphoproliferative disorders: Molecular basis of disease histogenesis and pathogenesis. Hematol. Oncol. 2005, 23, 61–67. [Google Scholar] [CrossRef]
  11. Henderson, S.; Rowe, M.; Gregory, C.; Croom-Carter, D.; Wang, F.; Longnecker, R.; Kieff, E.; Rickinson, A. Induction of bcl-2 expression by epstein-barr virus latent membrane protein 1 protects infected B cells from programmed cell death. Cell 1991, 65, 1107–1115. [Google Scholar] [CrossRef]
  12. Swerdlow, S.; Campo, E.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.T.J. Post-transplant Lymphoproliferative Disorders. In WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues; Swerdlow, S.H., Webber, S.A., Ferry, J.A., Eds.; AARC: Lyon, France, 2017; pp. 453–464. [Google Scholar]
  13. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef] [PubMed]
  14. Landgren, O.; Gilbert, E.S.; Rizzo, J.D.; Socié, 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]
  15. Al Hamed, R.; Bazarbachi, A.H.; Mohty, M. Epstein-Barr virus-related post-transplant lymphoproliferative disease (EBV-PTLD) in the setting of allogeneic stem cell transplantation: A comprehensive review from pathogenesis to forthcoming treatment modalities. Bone Marrow Transplant. 2020, 55, 25–39. [Google Scholar] [CrossRef]
  16. Juvonen, E.; Aalto, S.M.; Tarkkanen, J.; Volin, L.; Mattila, P.S.; Knuutila, S.; Ruutu, T.; Hedman, K. High incidence of PTLD after non-T-cell-depleted allogeneic haematopoietic stem cell transplantation as a consequence of intensive immunosuppressive treatment. Bone Marrow Transplant. 2003, 32, 97–102. [Google Scholar] [CrossRef] [PubMed]
  17. 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 2013, 99, 346–352. [Google Scholar] [CrossRef] [PubMed]
  18. Cohen, J.M.; Cooper, N.; Chakrabarti, S.; Thomson, K.; Samarasinghe, S.; Cubitt, D.; Lloyd, C.; Woolfrey, A.; Veys, P.; Amrolia, P.J. EBV-related disease following haematopoietic stem cell transplantation with reduced intensity conditioning. Leuk. Lymphoma 2007, 48, 256–269. [Google Scholar] [CrossRef]
  19. Cockfield, S.M. Identifying the patient at risk for post-transplant lymphoproliferative disorder. Transpl. Infect. Dis. 2001, 3, 70–78. [Google Scholar] [CrossRef]
  20. San-Juan, R.; Manuel, O.; Hirsch, H.H.; Fernández-Ruiz, M.; López-Medrano, F.; Comoli, P.; Caillard, S.; Grossi, P.A.; Aguado, J.M.; Alamo-Martinez, J.M.; et al. Current preventive strategies and management of Epstein–Barr virus-related post-transplant lymphoproliferative disease in solid organ transplantation in Europe. Results of the ESGICH Questionnaire-based Cross-sectional Survey. Clin. Microbiol. Infect. 2015, 21, 604.e1–604.e9. [Google Scholar] [CrossRef]
  21. Gross, T.G.; Steinbuch, M.; DeFor, T.; Shapiro, R.S.; McGlave, P.; Ramsay, N.K.C.; Wagner, J.E.; Filipovich, A.H. B cell lymphoproliferative disorders following hematopoietic stem cell transplantation: Risk factors, treatment and outcome. Bone Marrow Transplant. 1999, 23, 251–258. [Google Scholar] [CrossRef]
  22. Kinch, A.; Öberg, 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]
  23. Brunstein, C.G.; Weisdorf, D.J.; DeFor, T.; Barker, J.N.; Tolar, J.; van Burik, J.-A.H.; 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] [PubMed]
  24. Sampaio, M.S.; Cho, Y.W.; Shah, T.; Bunnapradist, S.; Hutchinson, I.V. Impact of Epstein-Barr virus donor and recipient serostatus on the incidence of post-transplant lymphoproliferative disorder in kidney transplant recipients. Nephrol. Dial. Transplant. 2012, 27, 2971–2979. [Google Scholar] [CrossRef] [PubMed]
  25. Dierickx, D.; Habermann, T.M. Post-Transplantation Lymphoproliferative Disorders in Adults. N. Engl. J. Med. 2018, 378, 549–562. [Google Scholar] [CrossRef]
  26. Allen, U.D.; Preiksaitis, J.K. Post-transplant lymphoproliferative disorders, Epstein-Barr virus infection, and disease in solid organ transplantation: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin. Transplant. 2019, 33, e13652. [Google Scholar] [CrossRef] [PubMed]
  27. Lieberman, F.; Yazbeck, V.; Raptis, A.; Felgar, R.; Boyiadzis, M. Primary central nervous system post-transplant lymphoproliferative disorders following allogeneic hematopoietic stem cell transplantation. J. Neuro-Oncol. 2011, 107, 225–232. [Google Scholar] [CrossRef] [PubMed]
  28. Preiksaitis, J.K.; Pang, X.L.; Fox, J.D.; Fenton, J.M.; Caliendo, A.M.; Miller, G.G. Interlaboratory Comparison of Epstein-Barr Virus Viral Load Assays. Am. J. Transplant. 2009, 9, 269–279. [Google Scholar] [CrossRef] [PubMed]
  29. Preiksaitis, J.K. Epstein—Barr Viral Load Testing: Role in the Prevention, Diagnosis and Management of Posttransplant Lymphoproliferative Disorders. In Post-Transplant Lymphoproliferative Disorders; Dharnidharka, V.R., Green, M., Webber, S., Eds.; Springer: Berlin/Heidelberg, Germany, 2010; pp. 45–67. [Google Scholar] [CrossRef]
  30. Gärtner, B.; Preiksaitis, J.K. EBV viral load detection in clinical virology. J. Clin. Virol. 2010, 48, 82–90. [Google Scholar] [CrossRef]
  31. Styczynski, J.; Van Der Velden, W.; Fox, C.P.; Engelhard, D.; de la Camara, R.; Cordonnier, C.; Ljungman, P. Management of Epstein-Barr Virus infections and post-transplant lymphoproliferative disorders in patients after allogeneic hematopoietic stem cell transplantation: Sixth European Conference on Infections in Leukemia (ECIL-6) guidelines. Haematologica 2016, 101, 803–811. [Google Scholar] [CrossRef]
  32. Tang, L.; Su, Y.; Pounds, S.; Hayden, R.T. Quantitative Inference of Commutability for Clinical Viral Load Testing. J. Clin. Microbiol. 2018, 56, e00146-18. [Google Scholar] [CrossRef]
  33. Buelow, D.; Sun, Y.; Tang, L.; Gu, Z.; Pounds, S.; Hayden, R. Comparative Evaluation of Four Real-Time PCR Methods for the Quantitative Detection of Epstein-Barr Virus from Whole Blood Specimens. J. Mol. Diagn. 2016, 18, 527–534. [Google Scholar] [CrossRef]
  34. Rychert, J.; Danziger-Isakov, L.; Yen-Lieberman, B.; Storch, G.; Buller, R.; Sweet, S.C.; Mehta, A.K.; Cheeseman, J.A.; Heeger, P.; Rosenberg, E.S.; et al. Multicenter comparison of laboratory performance in cytomegalovirus and Epstein-Barr virus viral load testing using international standards. Clin. Transplant. 2014, 28, 1416–1423. [Google Scholar] [CrossRef] [PubMed]
  35. 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]
  36. Ahmad, I.; Cau, N.V.; Kwan, J.; Maaroufi, Y.; Meuleman, N.; Aoun, M.; Lewalle, P.; Martiat, P.; Crokaert, F.; Bron, D. Preemptive Management of Epstein-Barr Virus Reactivation After Hematopoietic Stem-Cell Transplantation. Transplantation 2009, 87, 1240–1245. [Google Scholar] [CrossRef] [PubMed]
  37. Worth, A.; Conyers, R.; Cohen, J.; Jagani, M.; Chiesa, R.; Rao, K.; Goulden, N.; Veys, P.; Amrolia, P.J. Pre-emptive rituximab based on viraemia and T cell reconstitution: A highly effective strategy for the prevention of Epstein–Barr virus-associated lymphoproliferative disease following stem cell transplantation. Br. J. Haematol. 2011, 155, 377–385. [Google Scholar] [CrossRef] [PubMed]
  38. DeStefano, C.B.; Desai, S.H.; Shenoy, A.G.; Catlett, J.P. Management of post-transplant lymphoproliferative disorders. Br. J. Haematol. 2018, 182, 330–343. [Google Scholar] [CrossRef]
  39. Kullberg-Lindh, C.; Olofsson, S.; Brune, M.; Lindh, M. Comparison of serum and whole blood levels of cytomegalovirus and Epstein-Barr virus DNA. Transpl. Infect. Dis. 2008, 10, 308–315. [Google Scholar] [CrossRef]
  40. Reshef, R.; Vardhanabhuti, S.; Luskin, M.R.; Heitjan, D.F.; Hadjiliadis, D.; Goral, S.; Krok, K.L.; Goldberg, L.R.; Porter, D.L.; Stadtmauer, E.A.; et al. Reduction of Immunosuppression as Initial Therapy for Posttransplantation Lymphoproliferative Disorder★. Am. J. Transplant. 2011, 11, 336–347. [Google Scholar] [CrossRef]
  41. 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]
  42. Raberahona, M.; Wackenheim, C.; Carré, M.; Thiébaut, A.; Lupo, J.; Semenova, T.; Morand, P.; Bulabois, C.-E.; Cahn, J.-Y.; Germi, R.; et al. Dynamics of Epstein-Barr viral load after hematopoietic stem cell transplantation and effect of preemptive rituximab therapy. Transpl. Infect. Dis. 2016, 18, 889–895. [Google Scholar] [CrossRef]
  43. Chang, Y.-J.; Xu, L.-P.; Wang, Y.; Zhang, X.-H.; Chen, H.; Chen, Y.-H.; Wang, F.-R.; Han, W.; Sun, Y.-Q.; Yan, C.-H.; et al. Rituximab for desensitization during HLA-mismatched stem cell transplantation in patients with a positive donor-specific anti-HLA antibody. Bone Marrow Transplant. 2020, 55, 1326–1336. [Google Scholar] [CrossRef]
  44. Coppoletta, S.; Tedone, E.; Galano, B.; Soracco, M.; Raiola, A.M.; Lamparelli, T.; Gualandi, F.; Bregante, S.; Ibatici, A.; di Grazia, C.; et al. Rituximab Treatment for Epstein-Barr Virus DNAemia after Alternative-Donor Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2011, 17, 901–907. [Google Scholar] [CrossRef] [PubMed]
  45. 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]
  46. Messahel, B.; Taj, M.M.; Hobson, R.; Hadzic, N.; Ramsay, A.; Hann, I.; Pinkerton, R. Single agent efficacy of rituximab in childhood immunosuppression related lymphoproliferative disease: A United Kingdom Children’s Cancer Study Group (UKCCSG) retrospective review. Leuk. Lymphoma 2006, 47, 2584–2589. [Google Scholar] [CrossRef] [PubMed]
  47. Rouce, R.H.; Louis, C.U.; Heslop, H.E. Epstein–Barr virus lymphoproliferative disease after hematopoietic stem cell transplant. Curr. Opin. Hematol. 2014, 21, 476–481. [Google Scholar] [CrossRef]
  48. 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]
  49. Chiereghin, A.; Bertuzzi, C.; Piccirilli, G.; Gabrielli, L.; Squarzoni, D.; Turello, G.; Ferioli, M.; Sessa, M.; Bonifazi, F.; Zanoni, L.; et al. Successful management of EBV-PTLD in allogeneic bone marrow transplant recipient by virological–immunological monitoring of EBV infection, prompt diagnosis and early treatment. Transpl. Immunol. 2016, 34, 60–64. [Google Scholar] [CrossRef] [PubMed]
  50. Stocker, N.; Labopin, M.; Boussen, I.; Paccoud, O.; Bonnin, A.; Malard, F.; Amiel, C.; Gozlan, J.; Battipaglia, G.; Duléry, R.; et al. Pre-emptive rituximab treatment for Epstein–Barr virus reactivation after allogeneic hematopoietic stem cell transplantation is a worthwhile strategy in high-risk recipients: A comparative study for immune recovery and clinical outcomes. Bone Marrow Transplant. 2019, 55, 586–594. [Google Scholar] [CrossRef]
  51. Avivi, I.; Stroopinsky, D.; Katz, T. Anti-CD20 monoclonal antibodies: Beyond B-cells. Blood Rev. 2013, 27, 217–223. [Google Scholar] [CrossRef]
  52. Kinch, A.; Hallböök, H.; Arvidson, J.; Sällström, K.; Bondeson, K.; Pauksens, K. Long-term outcome of Epstein–Barr virus DNAemia and PTLD with the use of preemptive rituximab following allogeneic HSCT. Leuk. Lymphoma 2017, 59, 1172–1179. [Google Scholar] [CrossRef]
  53. Luterbacher, F.; Bernard, F.; Baleydier, F.; Ranza, E.; Jandus, P.; Blanchard-Rohner, G. Case Report: Persistent Hypogammaglobulinemia More Than 10 Years After Rituximab Given Post-HSCT. Front. Immunol. 2021, 12, 773853. [Google Scholar] [CrossRef]
  54. Petropoulou, A.D.; Porcher, R.; de Latour, R.P.; Xhaard, A.; Weisdorf, D.; Ribaud, P.; Rodriguez-Otero, P.; Agbalika, F.; Talbot, A.; Toubert, A.; et al. Increased Infection Rate After Preemptive Rituximab Treatment for Epstein-Barr Virus Reactivation After Allogeneic Hematopoietic Stem-Cell Transplantation. Transplantation 2012, 94, 879–883. [Google Scholar] [CrossRef] [PubMed]
  55. 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]
  56. Heslop, H.E.; Sharma, S.; Rooney, C.M. Adoptive T-Cell Therapy for Epstein-Barr Virus–Related Lymphomas. J. Clin. Oncol. 2021, 39, 514–524. [Google Scholar] [CrossRef] [PubMed]
  57. Heslop, H.E.; Slobod, K.S.; Pule, M.A.; Hale, G.A.; Rousseau, A.; Smith, C.A.; Bollard, C.M.; Liu, H.; Wu, M.-F.; Rochester, R.J.; et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 2010, 115, 925–935. [Google Scholar] [CrossRef] [PubMed]
  58. Saglio, F.; Hanley, P.J.; Bollard, C.M. The time is now: Moving toward virus-specific T cells after allogeneic hematopoietic stem cell transplantation as the standard of care. Cytotherapy 2014, 16, 149–159. [Google Scholar] [CrossRef] [PubMed]
  59. Moosmann, A.; Bigalke, I.; Tischer, J.; Schirrmann, L.; Kasten, J.; Tippmer, S.; Leeping, M.; Prevalšek, D.; Jaeger, G.; Ledderose, G.; et al. Effective and long-term control of EBV PTLD after transfer of peptide-selected T cells. Blood 2010, 115, 2960–2970. [Google Scholar] [CrossRef]
  60. Keller, M.D.; Darko, S.; Lang, H.; Ransier, A.; Lazarski, C.A.; Wang, Y.; Hanley, P.J.; Davila, B.J.; Heimall, J.R.; Ambinder, R.F.; et al. T-cell receptor sequencing demonstrates persistence of virus-specific T cells after antiviral immunotherapy. Br. J. Haematol. 2019, 187, 206–218. [Google Scholar] [CrossRef]
  61. Papadopoulou, A.; Alvanou, M.; Karavalakis, G.; Tzannou, I.; Yannaki, E. Pathogen-specific T Cells: Targeting Old Enemies and New Invaders in Transplantation and Beyond. Hemasphere 2023, 7, e809. [Google Scholar] [CrossRef]
  62. Luttwak, E.; Hagin, D.; Perry, C.; Wolach, O.; Itchaki, G.; Amit, O.; Bar-On, Y.; Freund, T.; Kay, S.; Eshel, R.; et al. Anti-CD19 CAR-T therapy for EBV-negative posttransplantation lymphoproliferative disease—A single center case series. Bone Marrow Transplant. 2020, 56, 1031–1037. [Google Scholar] [CrossRef]
  63. Dang, B.N.; Ch’ng, J.; Russell, M.; Cheng, J.C.; Moore, T.B.; Alejos, J.C. Treatment of post-transplant lymphoproliferative disorder (PTLD) in a heart transplant recipient with chimeric antigen receptor T-cell therapy. Pediatr. Transplant. 2020, 25, e13861. [Google Scholar] [CrossRef]
  64. Krishnamoorthy, S.; Ghobadi, A.; Santos, R.D.; Schilling, J.D.; Malone, A.F.; Murad, H.; Bartlett, N.L.; Alhamad, T. CAR-T therapy in solid organ transplant recipients with treatment refractory posttransplant lymphoproliferative disorder. Am. J. Transplant. 2020, 21, 809–814. [Google Scholar] [CrossRef] [PubMed]
  65. Mamlouk, O.; Nair, R.; Iyer, S.P.; Edwards, A.; Neelapu, S.S.; Steiner, R.E.; Adkins, S.A.; Hawkins, M.; Saini, N.Y.; Devashish, K.; et al. Safety of CAR T-cell therapy in kidney transplant recipients. Blood 2021, 137, 2558–2562. [Google Scholar] [CrossRef] [PubMed]
  66. Yan, N.; Wang, N.; Zhang, P.; Wang, G.; Mao, X.; Peng, D.; Kuang, D.; Chen, L.; Zhu, L.; Zhou, J.; et al. Case Report: Successful Chimeric Antigen Receptor T Cell Therapy in Haploidentical-Allogeneic Stem Cell Transplant Patients with Post-Transplant Lymphoproliferative Disorder. Front. Oncol. 2021, 11, 709370. [Google Scholar] [CrossRef] [PubMed]
  67. Portell, C.; Nand, S. Single agent lenalidomide induces a response in refractory T-cell posttransplantation lymphoproliferative disorder. Blood 2008, 111, 4416–4417. [Google Scholar] [CrossRef] [PubMed]
  68. Läubli, H.; Tzankov, A.; Juskevicius, D.; Degen, L.; Rochlitz, C.; Stenner-Liewen, F. Lenalidomide monotherapy leads to a complete remission in refractory B-cell post-transplant lymphoproliferative disorder. Leuk. Lymphoma 2015, 57, 945–948. [Google Scholar] [CrossRef]
  69. Blaes, A.H.; Hammerschmidt, D.E.; Peterson, B.A. Bortezomib and Rituximab in the Treatment of Post-Transplant Lymphoproliferative Disorders. Blood 2011, 118, 4947. [Google Scholar] [CrossRef]
  70. Reid, E.G. Bortezomib-induced Epstein–Barr virus and Kaposi sarcoma herpesvirus lytic gene expression. Curr. Opin. Oncol. 2011, 23, 482–487. [Google Scholar] [CrossRef]
  71. 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. Epstein-barr virus DNAemia monitoring for the management of post-transplant lymphoproliferative disorder. Cytotherapy 2018, 20, 706–714. [Google Scholar] [CrossRef]
  72. García-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]
  73. Chen, J.; Sun, Y.Q.; Xu, L.P.; Zhang, X.H.; Liu, K.Y.; Mo, X.D.; Cheng, Y.F.; Huang, X.J.; Wang, Y. Dynamic monitoring of plasma Epstein-Barr Virus DNA load can predict the occurrence of lymphoproliferative disorders after haploidentical hematopoietic stem cell transplantation. Zhonghua Xue Ye Xue Za Zhi 2023, 44, 284–288. [Google Scholar] [CrossRef]
  74. Düver, F.; Weißbrich, B.; Eyrich, M.; Wölfl, 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]
  75. Solano, C.; Mateo, E.M.; Pérez, A.; Talaya, A.; Terol, M.J.; Albert, E.; Giménez, E.; Vinuesa, V.; Piñana, J.L.; Boluda, J.C.H.; et al. Epstein-Barr virus DNA load kinetics analysis in allogeneic hematopoietic stem cell transplant recipients: Is it of any clinical usefulness? J. Clin. Virol. 2017, 97, 26–32. [Google Scholar] [CrossRef] [PubMed]
  76. Sakellari, I.; Batsis, I.; Bousiou, Z.; Mallouri, D.; Constantinou, V.; Gavriilaki, E.; Smias, C.; Yannaki, E.; Kaloyannidis, P.; Papaioannou, G.; et al. The Role of Low-dose Anti-thymocyte Globulin as Standard Prophylaxis in Mismatched and Matched Unrelated Hematopoietic Peripheral Stem Cell Transplantation for Hematologic Malignancies. Clin. Lymphoma Myeloma Leuk. 2017, 17, 658–666. [Google Scholar] [CrossRef]
  77. Gavriilaki, E.; Sakellari, I.; Chatzikonstantinou, T.; Mallouri, D.; Batsis, I.; Vardi, A.; Bousiou, Z.; Koravou, E.-E.; Masmanidou, M.; Touloumenidou, T.; et al. Endothelial and Complement Activation as Predictors of Survival in Adult Allogeneic Hematopoietic Cell Transplantation. Hemasphere 2020, 5, e487. [Google Scholar] [CrossRef] [PubMed]
  78. Glucksberg, H.; Storb, R.; Fefer, A.; Buckner, C.D.; Neiman, P.E.; Clift, R.A.; Lerner, K.G.; Thomas, E.D. Clinical manifestations of graft-versus-host disease in human recipients of marrow from hl-a-matched sibling donors. Transplantation 1974, 18, 295–304. [Google Scholar] [CrossRef]
  79. Lee, S.J.; Wolff, D.; Kitko, C.; Koreth, J.; Inamoto, Y.; Jagasia, M.; Pidala, J.; Olivieri, A.; Martin, P.J.; Przepiorka, D.; et al. Measuring Therapeutic Response in Chronic Graft-versus-Host Disease. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: IV. The 2014 Response Criteria Working Group Report. Biol. Blood Marrow Transplant. 2015, 21, 984–999. [Google Scholar] [CrossRef]
  80. Przepiorka, D.; Weisdorf, D.; Martin, P.; Klingemann, H.G.; Beatty, P.; Hows, J.; Thomas, E.D. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995, 15, 825–828. [Google Scholar]
Figure 1. Patient sampling scheme employed in the study.
Figure 1. Patient sampling scheme employed in the study.
Ijms 24 16029 g001
Figure 2. A cutoff of 67,150 copies predicted post-transplant lymphoproliferative disease (PTLD) with 80% sensitivity and specificity.
Figure 2. A cutoff of 67,150 copies predicted post-transplant lymphoproliferative disease (PTLD) with 80% sensitivity and specificity.
Ijms 24 16029 g002
Table 1. Descriptive table of 100 patients with clinically relevant Epstein–Barr virus (EBV) reactivation. Mean and median viral genome copies (VGC)/mL were calculated from all positive pre-treatment samples.
Table 1. Descriptive table of 100 patients with clinically relevant Epstein–Barr virus (EBV) reactivation. Mean and median viral genome copies (VGC)/mL were calculated from all positive pre-treatment samples.
Biographical DataCategoryFrequencyPercentageMean
(Copies/mL)
Median
(Copies/mL)
Min
(Copies/mL)
Max
(Copies/mL)
GenderMale56/10056%112,94427,95091202,760,000
Female44/10044%102,34820,00086901,150,000
Age group0–186/1006%64,56717,70010,400180,000
18–4449/10049%118,48027,20087002,760,000
44+45/10045%103,00521,00086901,150,000
Type of transplantSibling20/10020%95,62220,50099301,150,000
VUD68/10068%74,10022,8508690951,000
Haplo12/10012%323,07548,90010,8002,760,000
Type of conditioningMyeloablative88/10088%118,79627,25086902,760,000
RIC12/10012%19,22517,250870047,100
aGVHDYes64/10064%95,70626,65086902,760,000
No36/10036%130,63722,75099301,150,000
cGVHDYes82/10082%76,34426,65086901,150,000
No18/10018%253,77217,55010,0002,760,000
Rituximab treatmentYes74/10074%135,78830,40086902,760,000
No26/10026%29,99616,35010,300148,000
Rituximab cyclesOne64/7486.5%138,66329,30086902,760,000
Two9/7412.1%83,24429,80010,400499,000
Three1/741.4%383,000383,000383,000383,000
PTLDYes5/1005%684,68078,40031,0002,760,000
No95/10095%77,49521,00086901,150,000
EBV relapseYes13/10013%281,32340,00011,0002,760,000
No87/10087%76,33920,50086901,150,000
CMV reactivationYes47/10047%82,76620,30086901,150,000
No53/10053%130,77824,60091202,760,000
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Papalexandri, A.; Gavriilaki, E.; Vardi, A.; Kotsiou, N.; Demosthenous, C.; Constantinou, N.; Touloumenidou, T.; Zerva, P.; Kika, F.; Iskas, M.; et al. Pre-Emptive Use of Rituximab in Epstein–Barr Virus Reactivation: Incidence, Predictive Factors, Monitoring, and Outcomes. Int. J. Mol. Sci. 2023, 24, 16029. https://doi.org/10.3390/ijms242216029

AMA Style

Papalexandri A, Gavriilaki E, Vardi A, Kotsiou N, Demosthenous C, Constantinou N, Touloumenidou T, Zerva P, Kika F, Iskas M, et al. Pre-Emptive Use of Rituximab in Epstein–Barr Virus Reactivation: Incidence, Predictive Factors, Monitoring, and Outcomes. International Journal of Molecular Sciences. 2023; 24(22):16029. https://doi.org/10.3390/ijms242216029

Chicago/Turabian Style

Papalexandri, Apostolia, Eleni Gavriilaki, Anna Vardi, Nikolaos Kotsiou, Christos Demosthenous, Natassa Constantinou, Tasoula Touloumenidou, Panagiota Zerva, Fotini Kika, Michalis Iskas, and et al. 2023. "Pre-Emptive Use of Rituximab in Epstein–Barr Virus Reactivation: Incidence, Predictive Factors, Monitoring, and Outcomes" International Journal of Molecular Sciences 24, no. 22: 16029. https://doi.org/10.3390/ijms242216029

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