Zum Hauptinhalt springen

Impact of Tumour Epstein-Barr Virus Status on Clinical Outcome in Patients with Classical Hodgkin Lymphoma (cHL): A Review of the Literature and Analysis of a Clinical Trial Cohort of Children with cHL.

Nohtani, M ; Vrzalikova, K ; et al.
In: Cancers, Jg. 14 (2022-09-01), Heft 17
Online academicJournal

Impact of Tumour Epstein–Barr Virus Status on Clinical Outcome in Patients with Classical Hodgkin Lymphoma (cHL): A Review of the Literature and Analysis of a Clinical Trial Cohort of Children with cHL 

Simple Summary: The Epstein–Barr virus (EBV) contributes to different forms of human cancer, including a subset of classical Hodgkin lymphoma (cHL), a B-cell lymphoma with unusual histological features. Although the pathogenesis of EBV-associated cHL remains to be elucidated, biological investigations point to an important aetiological role for the virus in the development of this tumour. This is even more relevant now considering the potential opportunities that exist to treat EBV-associated disorders, for example, with immunotherapeutics or small molecule inhibitors targeting viral proteins. For this reason, we believe it is now timely to review the association between EBV and cHL and in particular to re-evaluate the impact of EBV status on clinical outcomes in cHL patients. Herein, we also report the impact of EBV on clinical outcomes in a cohort of children and adolescents with cHL. In this study, we have re-evaluated how EBV status influences clinical outcome. To accomplish this, we performed a literature review of all studies that have reported the effect of EBV status on patient outcome and also explored the effect of EBV positivity on outcome in a clinical trial of children with cHL from the UK. Our literature review revealed that almost all studies of older adults/elderly patients have reported an adverse effect of an EBV-positive status on outcome. In younger adults with cHL, EBV-positive status was either associated with a moderate beneficial effect or no effect, and the results in children and adolescents were conflicting. Our own analysis of a series of 166 children with cHL revealed no difference in overall survival between EBV-positive and EBV-negative groups (p = 0.942, log rank test). However, EBV-positive subjects had significantly longer event-free survival (p = 0.0026). Positive latent membrane protein 1 (LMP1) status was associated with a significantly lower risk of treatment failure in a Cox regression model (HR = 0.21, p = 0.005). In models that controlled for age, gender, and stage, EBV status had a similar effect size and statistical significance. This study highlights the age-related impact of EBV status on outcome in cHL patients and suggests different pathogenic effects of EBV at different stages of life.

Keywords: classic Hodgkin lymphoma (cHL); Epstein–Barr virus (EBV); clinical trial

1. Introduction

Classical Hodgkin lymphoma (cHL) is characterised by rare malignant Hodgkin/Reed Sternberg (HRS) cells surrounded by a florid tumour microenvironment (TME) comprising different inflammatory cells. HRS cells are germinal centre (GC)-experienced B cells [[1]] but do not express a functional B-cell receptor (BCR); survival of these cells is mediated by activation of anti-apoptosis pathways crucial for cHL pathogenesis [[2]].

Following B-cell infection with EBV, extrachromosomal copies of circular genomes, known as episomes, are created by fusion of the viral terminal repeats (TRs) creating termini of a unique length [[3]]. The detection of TRs of different lengths indicates more than one infection event [[3]]. In contrast, TRs with an identical number of repeats are found in cHL and other EBV-associated cancers, indicating expansion from a single infected cell [[4]]. EBV is crucial also for cHL progression, as it is found during the course of cHL [[5]].

EBV protein expression in HRS cells is limited to (i) Epstein–Barr virus nuclear antigen-1 (EBNA1) [[6]], an essential replication and virus maintenance factor, as well as a transcription factor that regulates the expression of both viral and cellular genes [[7], [9]]; EBNA1 has been shown to promote the growth and survival of cHL-derived cell lines [[11]] and (ii) two latent membrane proteins (LMPs) [[13], [15]]; LMP1 is a constitutively active CD40 receptor [[16]] that activates oncogenic pathways including NF-κB, JAK/STAT, AP-1, and phospatidylinositol-3 kinase (PI3K)/AKT [[17], [19]], whereas LMP2A is a BCR homologue [[21]] that can promote the survival of BCR-negative B cells [[23], [25]].

Serological studies showing that antibody levels to EBV antigens were raised in HL patients provided early evidence for an aetiological role [[26]]. Later, people who develop EBV-positive cHL were shown to be more likely to have elevated antibody levels to EBV antigens compared to those who develop EBV-negative cHL [[28]]. Furthermore, infectious mononucleosis (IM), representing symptomatic primary EBV infection, was shown to increase the risk of EBV-positive, but not EBV-negative, cHL [[29], [31]], and a seasonal peak in children diagnosed with cHL was shown for EBV-positive, but not for EBV-negative, cHL [[33]]; these data point to primary EBV infection as a trigger for the development of EBV-positive cHL. While only a small fraction of people infected with EBV will develop EBV-positive cHL [[34]], susceptibility is associated with variation in the human leukocyte antigen (HLA) region [[35], [37]]; those with HLA-A*01 have an increased risk, and those with HLA-A*02 have a decreased risk [[36], [38]].

EBV is present in HRS cells more frequently in male patients, in those with mixed cellularity disease, and in patients from resource-poor countries. Geographical differences might be explained by differing susceptibilities between ethnic groups [[40]]. For example, EBV-positive cHL is more common in Asians and Hispanics compared with whites or blacks and in South Asian children compared with non-South Asian children in the UK [[40], [42]]. In resource-rich nations, the proportion of cHL with EBV is higher in older people and in children, with lower rates in young adults [[43]]. Jarrett et al. have proposed a four-disease model: (1) childhood cHL, which is frequently EBV-positive; (2) cHL of older adults, which is also often EBV-related; it is well known that aging is associated with decreased immune function, so this entity may be related to EBV reactivation; (3) EBV-negative disease in young adults, usually, but not always, of nodular sclerosis type; (4) EBV-associated cHL occurring after late EBV exposure [[44]].

While first-line combination chemotherapy and radiotherapy is effective for the majority of cHL patients, prognosis for patients with refractory or relapsed disease remains dismal, despite the use of newer targeted therapies such as brentuximab vedotin. The current treatments also cause significant long-term toxicities that include secondary malignancies, cardiopulmonary toxicity, hypothyroidism, and infertility. Outcomes are particularly poor for older patients (5-year survival 30–50% in patients older than 70 years). Despite overwhelming evidence supporting an aetiological role for EBV in cHL, EBV status does not currently influence patient management. Moreover, therapies designed to specifically target EBV are yet to be adopted.

In this study, we have performed a comprehensive literature review encompassing all relevant previous studies reporting the effect of EBV status on patient outcome. We have also separately explored the effect of EBV positivity in a clinical trial of children and adolescents with cHL from the United Kingdom in an attempt to clarify the apparently conflicting effects of EBV status on clinical outcomes in the paediatric setting.

2. Materials and Methods

2.1. Literature Review

Using the keywords "EBV" and "Hodgkin lymphoma," we conducted a search on PubMed (http://www.ncbi.nlm.nih.gov/pubmed;accessedon2February2022). Additionally, we looked through the reference lists of the papers and manually included the publications missed by the original search. Using the title, names and affiliations of the authors, duplicate information or overlapping articles were eliminated. We confined our analysis to studies that used either in situ hybridisation to detect the Epstein–Barr-encoded RNAs (EBER1 and EBER2) and/or immunohistochemistry to identify expression of the EBV oncogene, LMP1. We excluded studies in which other virus proteins (LMP2A and EBNA1, which are less abundant in EBV-positive HRS cells) were used as targets. We also excluded studies that exclusively used PCR to detect EBV. NLPHL is regarded as a distinct disease entity, and it differs from cHL primarily in terms of morphology, phenotype, genetics, clinical behaviour and EBV positivity. Therefore, inclusion of these cases in studies was taken into account.

2.2. Analysis of a Clinical Trial Cohort of Children and Adolescents with cHL

Patients

Eligible patients included all children and adolescents (<18 years old) with newly diagnosed, untreated, biopsy-proven classical (i.e., excluding lymphocyte predominant subtype) HL who had been enrolled onto the HD 2000 02 (HD 3 trial) and for whom archival pathological material was available as FFPE sections (n = 189). Ethical approval and written informed consent were obtained from all patients and/or their parents/guardians in accordance with the then-current institutional and ethical committee guidelines. The results of this clinical trial and details of patient recruitment and treatment regimens have been reported previously [[45]].

2.3. Event-Free Survival and Overall Survival

Event-free survival (EFS) was calculated as the time from the date of diagnosis to the date of relapse, progression, or death from any cause. Patients who did not experience any events were censored at their last follow-up visit. Overall survival (OS) was measured from the date of diagnosis to the date of last follow-up visit or to the date of death.

2.4. EBV Detection

Immunohistochemistry for LMP1 was performed on paraffin sections from each case to detect the presence of EBV infection as previously described [[14]]. Specimens were recorded as either EBV-positive (LMP1 present within HRS cells) or EBV-negative (LMP1 not detectable in HRS cells).

2.5. Statistical Methods

Mann–Whitney U- and chi-squared tests were used to detect differences between EBV-positive and EBV-negative groups in terms of patient and disease characteristics. Differences in survival and event-free survival between EBV-positive and EBV-negative patients were investigated using Kaplan–Meier curves and log-rank tests. A Cox proportional hazards univariate analysis was also performed to ascertain the hazard ratio (HR) for event-free survival of each variable. The variables considered were EBV, age (≤12 and >12), sex, and disease stage early stages I/IIA and advanced stages IIB/III/IV). Multivariate Cox analysis was performed to determine which factors were independently predictive of event-free survival. Life-table methods were used to derive treatment failure rates (hazard) and the hazard ratio by EBV status at various time intervals after diagnosis. In order to determine that the 189patients that were eventually used in our analysis were representative of the whole cohort, chi-squared and t-tests were used to compare age, gender, subtype, stage, and symptoms between the two groups. All analyses used either SPSS Version 16 (SPSS Inc., Chicago, IL, USA) or R-4.1.2, and differences were deemed significant if the p-value was less than 0.05.

3. Results

3.1. Literature Review

A total of 40 studies met the inclusion criteria set out above (Supplementary Table S1). We focused our analysis on those studies that had categorized patients by age group. We summarize the results of our analysis below.

Older adults/elderly patients with cHL: our literature review revealed that an EBV-positive status was associated with poor prognosis in older adults/elderly patients in six of seven studies [[46], [48], [50], [52]].

Young adults with cHL: A modest beneficial effect of an EBV-positive status for young adults with cHL was demonstrated in four of eight studies. Three studies showed no significant effect of EBV, and one showed a negative impact of EBV on prognosis [[46], [48], [53], [55], [57]].

Paediatric and adolescent cHL: Eight studies explored the effect of EBV status on outcome in children or adolescents with cHL (Table 1). Engel et al. studied 47 patients (with follow up available on 36) and found significantly fewer deaths and longer median survival in EBV-positive cases [[58]]. Barros et al. studied 104 patients and showed that EBV was significantly associated with lower-risk nodular sclerosis disease [[59]]. Keegan et al. showed that in children <15 years old, EBV positivity was associated with longer survival, but this was only of borderline significance [[46]]. Three studies reported no significant effect of EBV status on outcome [[60], [62]]. Two studies reported a negative effect of EBV on outcome in children. In the largest study to date, Claviez [[63]] reported the impact of EBV status on outcome in 842 children and adolescents. They found that LMP1 positivity was associated with significantly poorer overall survival (OS) but found no effect on failure-free survival (FFS); in fact, in their study, FFS was higher in EBV-positive (89.1%) compared with EBV-negative (84.1%) patients. In the second study, Koh et al. reported that in 135 children with HL, EBER positivity was associated with a significantly higher international prognostic score (IPS) and significantly lower OS, although positivity was not an independent risk factor for OS [[64]]. The authors did not report the influence of EBV status on FFS.

3.2. Analysis of a Clinical Trial Cohort of Children and Adolescents with cHL

Given the observed uncertainty of EBV's effects on outcome in childhood and adolescent HL, we next examined the effects of EBV status on EFS and OS in a cohort of children and adolescents with cHL recruited to a clinical trial in the United Kingdom, where full clinical annotation of the sample collection, including reliable treatment, follow up, and outcome data were available. This trial has been reported previously, but EBV status has not been examined in this cohort before [[45]].

EBV status was tested in tissue from 189 trial subjects. Of these samples, 23 were excluded (six—identity uncertain; five—not lymphoid tissue; seven—test failed; one—repeated tests contradictory; four—not HL at Pathology Review). Sixty-two of the remaining 166 cases were EBV positive (34.4%, 95% CU 27.6–41.2%). The 189 patients included in this study were representative of the whole clinical trial cohort of 387 patients (Table 2).

Characteristics of the subjects, according to EBV status, are shown in Table 3. EBV-positive patients were significantly younger than EBV-negative patients, with a median age of 10.0 years, compared to 14.2 for EBV-negative subjects. EBV-positive cases were also less likely to have stage IV disease. The subtypes also differed significantly, EBV-positive tumours being less likely to be of nodular sclerosis subtype and more likely to be of mixed cellularity subtype.

In total, 14 deaths and 47 events were recorded (relapses, disease progression, persistent disease). Figure 1 shows Kaplan–Meier survival curves for overall survival (Figure 1a) and event-free survival (Figure 1b), by EBV status. There was no evidence of a difference between groups for overall survival (p = 0.942, log rank test). Event-free survival, however, was significantly poorer in EBV-negative subjects (p = 0.0026). As described above, fewer EBV-positive patients presented with stage IV disease than patients with EBV-negative cHL. The treatment for patients in this trial varied according to stage: patients with stage II and stage III disease received three cycles of ChlVbPP/ABVcD chemotherapy, whilst those with stage IV disease received four cycles. Therefore, the improved EFS observed for EBV-positive patients is not likely to be a consequence of more intensive chemotherapy.

In a Cox regression model, positive LMP1 status was associated with a significantly reduced risk of treatment failure (HR = 0.21, p = 0.005, Figure 2). EBV status retained a similar effect size and statistical significance in models adjusting for age, sex, and stage. We defined stage in two groups, early (stages I/IIA) and advanced (IIB/III/IV), based on risk stratification and treatment management guidelines [[86]]. Patients within the early group had reduced risk of failure compared to advanced stages, but this difference was not statistically significant (p = 0.45), and EBV status remained the most predictive factor. Age also has a negative effect on patients' outcome, with older patients having poorer outcomes, but this was not statistically significant.

4. Discussion

Notwithstanding some of the difficulties in drawing comparisons across different studies included in our literature review (different methodologies, patient recruitment, inclusion of NLPHL in some studies, different age cut-offs, etc.), we can draw several general conclusions. Thus, almost all studies of older adults/elderly patients reported an adverse effect of an EBV-positive status on outcome. In contrast, in younger adults with cHL, EBV-positive status was either associated with a moderate beneficial effect or no effect. It is possible that within the older adult group, EBV is not itself a driver of poorer outcomes but might be associated with other co-morbidities including reduced immunity and/or generally poorer health. Nevertheless, it should be noted that many older patients with cHL are unfit to receive standard-of-care combination chemotherapy; therefore, older people with EBV-positive cHL might benefit from therapies that specifically target EBV if they prove to be better tolerated than conventional chemotherapy. For a more detailed discussion of targeting EBV therapeutically in EBV-positive cHL and other cancers, the reader is referred to recent reviews [[88], [90]].

The situation at first sight would appear to be less clear in children and adolescents. Thus, we found that of the eight studies that reported an effect of EBV on outcome, three showed a beneficial effect, three no effect, and two an adverse effect. However, it must be pointed out that of the three studies showing no effect, two of these, one from India [[60]] and the other from Turkey [[61]], showed very high rates of EBV detection (96.6% and 82.5%, respectively), meaning they were likely insufficiently powered to detect differences even if present. Moreover, of the two studies showing an adverse effect of EBV, one showed poorer overall survival (OS) only in some subgroups of the disease but no effect on failure-free survival (FFS) [[63]]. The other study found a significantly shorter OS for EBV-positive patients but did not report FFS [[64]]. It has been suggested previously that OS may not be the best end-point since factors other than those related to the primary treatment may influence outcome [[66]].

Given the uncertainty of EBV's effects in the paediatric setting, we decided to study a separate cohort of children and adolescents with cHL recruited to a clinical trial [[45]]. We found that among 166 patients, EBV had no effect on OS but was associated with a significantly longer FFS. In a Cox regression model, EBV positivity was found to be associated with a significantly reduced risk of treatment failure. Moreover, EBV status retained a similar effect size and statistical significance in models adjusting for age, sex, and stage. Thus, our new data suggest that EBV might be associated with a better prognosis in children and adolescents, at least in the UK, and that it should be considered as a factor that could help stratify patients, for example, for de-escalation of therapy. Given the overall success of conventional therapies in children, OS may not be the best measure of outcome when assessing biological factors such as EBV status and which may be confounded by disease-unrelated deaths.

The cHL TME has been shown to vary with age and with EBV status (Figure 1). Thus, cytotoxic markers on T cells and numbers of CD16+ natural killer cells are increased in EBV-positive vs. EBV-negative cHL (cases included age-matched paediatric, adult, and elderly patients) [[91]]. EBV-positive paediatric cHL also exhibits a more cytotoxic TME with predominant Th1 polarisation, overexpression of CD8, TIA1, TBET, and granzyme B, and reduced FoxP3+ regulatory T cells (Tregs) compared with EBV-negative disease (Th2 and Th17) [[92]]. However, increased PDL1+ cells in EBV-positive paediatric cHL might blunt T-cell-mediated cytotoxicity [[94]]. In adult/elderly EBV-positive cHL, reduced granzyme B-positive T cells, increased Tregs, and limited interferon beta production indicate a more immunosuppressive TME, potentially contributing to the unfavourable outcome found in elderly cHL patients [[95]]. Furthermore, PDL1 is expressed more frequently on EBV-positive HRS cells rather than in the TME, where PD1 is predominantly found [[97]]. Thus, we speculate here that at least some of the age-related differences we have observed in our analysis might be because of differences in the TME between different patient groups (Figure 3). In this regard, it will be important to determine if EBV regulates the TME differently in these groups. We already know that EBV is a major regulator of the TME; effects that are mediated by LMP1 [[98]], LMP2A, and EBNA1 [[99]]. Conversely, the TME can also regulate virus gene expression in tumour cells [[101]].

In summary, a re-evaluation of the published literature presented here shows that in older adults and the elderly, an EBV-positive status is associated with poorer outcomes and that EBV-targeted therapies could be particularly valuable in this group of patients. Our literature review combined with a new analysis of a clinical trial cohort of children and adolescents with cHL suggest that while EBV-targeted therapies could also be useful in children, measuring EBV status might also be helpful in stratifying patients in future clinical trials.

5. Conclusions

Our literature review of the effects of EBV on outcomes for cHL patients combined with our own analysis of a cohort of paediatric and adolescent patients has shown important age-related effects. Currently, treatment for cHL patients is not stratified by EBV status, but this should be considered in future studies. For example, EBV status could be evaluated with the aim of reducing the harmful effects of harsh chemotherapeutic regimens used to treat cHL patients, particularly in childhood where there is a high risk of secondary malignancy and other complications. There is also a need to explore new opportunities to target EBV specifically, for example, with EBNA1 inhibitors or immunotherapies that target the virus. This could be particularly relevant for older adult/elderly patients where outcomes for EBV-positive patients are especially poor.

Figures and Tables

Graph: Figure 1 Overall survival (a) and event-free survival (b) for 166 subjects with cHL, by EBV status.

Graph: Figure 2 Multivariate analysis of the effect of EBV positivity on event-free survival in 166 cHL patients, adjusting for sex, age, and stage. ** marks p-values less than 0.01.

Graph: Figure 3 Schematic representation of differences in the cHL TME in different age groups. EBV-positive paediatric cHL has a more cytotoxic TME with predominant Th1 polarisation, overexpression of CD8, TIA1, TBET, and granzyme B, and fewer FoxP3+ regulatory T cells (Tregs) compared with EBV-negative disease. Fewer granzyme B-positive T cells, increased Tregs, and limited interferon beta production in older adult/elderly EBV-positive cHL indicate a more immunosuppressive TME.

Table 1 Summary of the impact of EBV on clinical outcome from published literature. Nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) cases were included in some studies. POS: positive effect of EBV on clinical outcome; NEG: negative effect of EBV on clinical outcome. "no" means no significant effect of EBV on clinical outcome. "No entry" means that the effect on clinical outcome was not studied in that group. * This was a meta-analysis that did not take age into account.

StudyPopulationNo. of PatientsNLPHL IncludedAge (Years)Effect (No Age Split)Children/AdolescentsYoung AdultsOlder Adults
Claviez (2005) [63] Multinational842yes2–20NEG
Koh (2018) [64]South Korea135no<15NEG
Dinand (2007) [60]India118yes<15no
Aktas (2007) [61]Turkey63noPaediatric patientsno
Chabay (2008) [62]Brazil, Argentina176yes0–18no
Engel (2000) [58]South Africa36no≤14POS
Keegan (2005) [46]USA922noup to 96POSnoNEG
Barros (2010) [59]Brazil104noup to 18POS
Koh (2012) [53]S Korea159yes4–77NEGNEG
Jarrett (2005) [47]UK437no16–74NEGnoNEG
Clarke (2001) [48]USA311yes19–79noNEG
Kwon (2006) [54]Korea56yes6–77NEGPOS
Glavina-Durdov (2001) [55]Croatia100yes13–84noPOS
Murray (1999) [56]UK190yes22–49POS
Flavell (2003) [57]UK273yes≥15noPOS
Stark (2002) [49]UK102yes≥60NEG
Diepstra (2009) [50]Netherlands412no7–91noNEG
Wang (2021) [51]China134yes5–74noNEG
Enblad (1999) [65]Sweden117yes11–87NEG
Proctor (2002) [52]UK94no>60no
Herling(2003) [66]USA, Italy, Greece303noadultsno
Axdorph (1999) [67]Sweden95no14–77no
Enblad (1997) [68]Sweden107yes6–87no
Keresztes (2006) [69]Hungary109no>61no
Krugmann (2003) [70]Austria119no14–83POS
Naresh (2000) [71] India110no4–61POS
Morente (1997) [72]Spain140yes5–83POS
Montalban (2000) [73]Spain110yesNKPOS
Trimeche (2007) [74]Belgium111no8–88NEG
Quijano (2004) [75]Columbia67noNKPOS
Myriam (2017) [76]Tunisia131no4–83NEG
Santisteban-Espejo (2021) [77]Spain88no19–82NEG
Elsayed (2014) [78]Japan389no4–89NEG
Souza (2010) [79]Brazil97no>18no
Cheriyalinkal Parambil (2020) [80]India189no≥15POS
Vestlev (1992) [81]Denmark66no12.8–60.5no
Armstrong (1994) [82]UK59yesNKno
Levy (2000) [83]Israel134yes4–50+NEG
Vassalo (2003) [84]Brazil78no>15POS
Lee (2014) [85]various*NANAno

Table 2 Comparison of clinical trial patients included or not included in the EBV study. Interquartile range (IQR).

In EBV Study (n = 189)Not in EBV Study (n = 198)p-Value
Age at diagnosis0.9
Median, IQR13.32 (10.25–14.86)13.20 (10.00–14.69)
Gender0.9
Male119 (65.0%)116 (58.5%)
Female64 (35.0%)82 (41.5%)
Subtype0.8
Nodular sclerosing126 (75.9%)129 (65.1%)
Mixed cellularity26 (15.6%)39 (19.6%)
Other/unknown14 (8.4%)30 (15.1%)
Stage0.2
I23 (12.5%)31 (15.6%)
II77 (44.8%)103 (52.0%)
III38 (22.4%)32 (16.1%)
IV37 (20.2%)32 (16.1%)
Symptoms0.9
A98 (59.0%)114 (57.5%)
B68 (41.0%)84 (42.4%)

Table 3 Clinicopathological variables according to EBV status. Stage, subtype and symptoms were defined following review. Interquartile range (IQR).

EBV+ (n = 62)EBV− (n = 104)p-Value
Age at diagnosis<0.001
Median, IQR10.0 (7.1–13.8)14.2 (12.2–15.3)
Gender0.067
Male44 (71.0%)59 (56.7%)
Female18 (29.0%)45 (43.3%)
Subtype0.005
Nodular sclerosing41 (66.1%)85 (81.7%)
Mixed cellularity17 (27.4%)9 (8.7%)
Other/unknown4 (6.5%)10 (9.6%)
Stage0.027
I8 (12.9%)5 (4.8%)
II29 (46.8%)48 (46.2%)
III18 (29.0%)22 (21.2%)
IV7 (11.3%)29 (27.9%)
Symptoms0.897
A37 (59.7%)61 (58.7%)
B25 (40.3%)43 (41.3%)

Author Contributions

Conceptualisation, P.G.M., P.K., M.N., K.B., J.B., S.M. and K.V.; methodology, J.E.P., M.N., É.F., P.K., S.M. and M.I.; resources, R.G. and K.M.; data curation, J.E.P., M.N., É.F., M.I. and S.D.; writing—original draft preparation and manuscript writing, P.G.M., M.N., K.V. and P.K.; visualisation, M.N., É.F. and J.E.P. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the UK National Research Ethics Committee (REC reference 16/WM/0037; IRAS Project no. 181189).

Informed Consent Statement

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

Data Availability Statement

The data presented in this study are available in this article and supplementary material.

Conflicts of Interest

The authors declare no conflict of interest.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14174297/s1, Table S1: Supplementary Table S1, rawData.xls.

Footnotes 1 Publisher's Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. References Küppers R., Rajewsky K., Zhao M., Simons G., Laumann R., Fischer R., Hansmann M.L. Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proc. Natl. Acad. Sci. USA. 1994; 91: 10962-10966. 10.1073/pnas.91.23.10962. 7971992 2 Kanzler H., Kuppers R., Hansmann M.L., Rajewsky K. Hodgkin and Reed-Sternberg cells in Hodgkin's disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. J. Exp. Med. 1996; 184: 1495-1505. 10.1084/jem.184.4.1495. 8879220 3 Raab-Traub N., Flynn K. The structure of the termini of the Epstein-Barr virus as a marker of clonal cellular proliferation. Cell. 1986; 47: 883-889. 10.1016/0092-8674(86)90803-2 4 Lardelli P., Garcia del Moral R. Clonal Epstein-Barr virus virus genome in Hodgkin's and non-Hodgkin's lymphoma. Blood. 1990; 75: 1589-1590. 10.1182/blood.V75.7.1589.1589 5 Coates P.J., Slavin G., D'Ardenne A.J. Persistence of Epstein-Barr virus in Reed-Sternberg cells throughout the course of Hodgkin's disease. J. Pathol. 1991; 164: 291-297. 10.1002/path.1711640404. 1656004 6 Grasser F.A., Murray P.G., Kremmer E., Klein K., Remberger K., Feiden W., Reynolds G., Niedobitek G., Young L.S., Mueller-Lantzsch N. Monoclonal antibodies directed against the Epstein-Barr virus-encoded nuclear antigen 1 (EBNA1): Immunohistologic detection of EBNA1 in the malignant cells of Hodgkin's disease. Blood. 1994; 84: 3792-3798. 10.1182/blood.V84.11.3792.bloodjournal84113792 7 Frappier L. Contributions of Epstein–Barr Nuclear Antigen 1 (EBNA1) to Cell Immortalization and Survival. Viruses. 2012; 4: 1537-1547. 10.3390/v4091537. 23170171 8 Frappier L. EBNA1 and host factors in Epstein–Barr virus latent DNA replication. Curr. Opin. Virol. 2012; 2: 733-739. 10.1016/j.coviro.2012.09.005 9 Frappier L. The Epstein-Barr Virus EBNA1 Protein. Scientifica. 2012; 2012: 438204. 10.6064/2012/438204 Tempera I., De Leo A., Kossenkov A.V., Cesaroni M., Song H., Dawany N., Showe L., Lu F., Wikramasinghe P., Lieberman P.M. Identification of MEF2B, EBF1, and IL6R as Direct Gene Targets of Epstein-Barr Virus (EBV) Nuclear Antigen 1 Critical for EBV-Infected B-Lymphocyte Survival. J. Virol. 2015; 90: 345-355. 10.1128/JVI.02318-15 Flavell J.R., Baumforth K.R., Wood V.H., Davies G.L., Wei W., Reynolds G.M., Morgan S., Boyce A., Kelly G.L., Young L.S. Down-regulation of the TGF-beta target gene, PTPRK, by the Epstein-Barr virus encoded EBNA1 contributes to the growth and survival of Hodgkin lymphoma cells. Blood. 2008; 111: 292-301. 10.1182/blood-2006-11-059881 Wood V.H., O'Neil J.D., Wei W., Stewart S.E., Dawson C.W., Young L.S. Epstein-Barr virus-encoded EBNA1 regulates cellular gene transcription and modulates the STAT1 and TGFbeta signaling pathways. Oncogene. 2007; 26: 4135-4147. 10.1038/sj.onc.1210496. 17486072 Deacon E.M., Pallesen G., Niedobitek G., Crocker J., Brooks L., Rickinson A.B., Young L.S. Epstein-Barr virus and Hodgkin's disease: Transcriptional analysis of virus latency in the malignant cells. J. Exp. Med. 1993; 177: 339-349. 10.1084/jem.177.2.339. 8381153 Murray P.G., Young L.S., Rowe M., Crocker J. Immunohistochemical demonstration of the Epstein-Barr virus-encoded latent membrane protein in paraffin sections of Hodgkin's disease. J. Pathol. 1992; 166: 1-5. 10.1002/path.1711660102. 1311374 Niedobitek G., Kremmer E., Herbst H., Whitehead L., Dawson C.W., Niedobitek E., Von Ostau C., Rooney N., Grasser F.A., Young L.S. Immunohistochemical detection of the Epstein-Barr virus-encoded latent membrane protein 2A in Hodgkin's disease and infectious mononucleosis. Blood. 1997; 90: 1664-1672. 10.1182/blood.V90.4.1664 Lam N., Sugden B. CD40 and its viral mimic, LMP1: Similar means to different ends. Cell Signal. 2003; 15: 9-16. 10.1016/S0898-6568(02)00083-9 Bargou R.C., Emmerich F., Krappmann D., Bommert K., Mapara M.Y., Arnold W., Royer H.D., Grinstein E., Greiner A., Scheidereit C. Constitutive nuclear factor-kappaB-relA activation is required for proliferation and survival of Hodgkin's disease tumor cells. J. Clin. Investig. 1997; 100: 2961-2969. 10.1172/JCI119849 Dutton A., Reynolds G.M., Dawson C.W., Young L.S., Murray P.G. Constitutive activation of phosphatidyl-inositide 3 kinase contributes to the survival of Hodgkin's lymphoma cells through a mechanism involving Akt kinase and mTOR. J. Pathol. 2005; 205: 498-506. 10.1002/path.1725 Heath E., Begue-Pastor N., Chaganti S., Croom-Carter D., Shannon-Lowe C., Kube D., Feederle R., Delecluse H.-J., Rickinson A.B., Bell A.I. Epstein-Barr Virus Infection of Naïve B Cells In Vitro Frequently Selects Clones with Mutated Immunoglobulin Genotypes: Implications for Virus Biology. PLoS Pathog. 2012; 8e1002697. 10.1371/journal.ppat.1002697 Holtick U., Vockerodt M., Pinkert D., Schoof N., Stürzenhofecker B., Kussebi N., Lauber K., Wesselborg S., Löffler D., Horn F. STAT3 is essential for Hodgkin lymphoma cell proliferation and is a target of tyrphostin AG17 which confers sensitization for apoptosis. Leukemia. 2005; 19: 936-944. 10.1038/sj.leu.2403750 Caldwell R.G., Wilson J.B., Anderson S.J., Longnecker R. Epstein-Barr Virus LMP2A Drives B Cell Development and Survival in the Absence of Normal B Cell Receptor Signals. Immunity. 1998; 9: 405-411. 10.1016/S1074-7613(00)80623-8 Merchant M., Swart R., Katzman R.B., Ikeda M., Ikeda A., Longnecker R., Dykstra M.L., Pierce S.K. The Effects of the Epstein-Barr Virus Latent Membrane Protein 2a on B Cell Function. Int. Rev. Immunol. 2001; 20: 805-835. 10.3109/08830180109045591 Chaganti S., Bell A., Pastor N.B., Milner A.E., Drayson M., Gordon J., Rickinson A.B. Epstein-Barr virus infection in vitro can rescue germinal center B cells with inactivated immunoglobulin genes. Blood. 2005; 106: 4249-4252. 10.1182/blood-2005-06-2327. 16123211 Bechtel D., Kurth J., Unkel C., Küppers R. Transformation of BCR-deficient germinal-center B cells by EBV supports a major role of the virus in the pathogenesis of Hodgkin and posttransplantation lymphomas. Blood. 2005; 106: 4345-4350. 10.1182/blood-2005-06-2342. 16131568 Mancao C., Altmann M., Jungnickel B., Hammerschmidt W. Rescue of "crippled" germinal center B cells from apoptosis by Epstein-Barr virus. Blood. 2005; 106: 4339-4344. 10.1182/blood-2005-06-2341 Levine P.H., Berard C.W., Carbone P.P., Waggoner D.E., Malan L. Elevated antibody titers to Epstein-Barr virus in Hodgkin's disease. Cancer. 1971; 7: 416-421. 10.1002/1097-0142(197102)27:2<416::AID-CNCR2820270227>3.0.CO;2-W Mueller N., Evans A., Harris N.L., Comstock G.W., Jellum E., Magnus K., Orentreich N., Polk B.F., Vogelman J. Hodgkin's disease and Epstein-Barr virus. Altered antibody pattern before diagnosis. N. Engl. J. Med. 1989; 320: 689-695. 10.1056/NEJM198903163201103 Levin L.I., Chang E.T., Ambinder R.F., Lennette E.T., Rubertone M.V., Mann R.B., Borowitz M., Weir E.G., Abbondanzo S.L., Mueller N.E. Atypical prediagnosis Epstein-Barr virus serology restricted to EBV-positive Hodgkin lymphoma. Blood. 2012; 120: 3750-3755. 10.1182/blood-2011-12-390823 Connelly R.R., Christine B.W. A cohort study of cancer following infectious mononucleosis. Cancer Res. 1974; 34: 1172-1178 Rosdahl N., Larsen S.O., Clemmesen J. Hodgkin's Disease in Patients with Previous Infectious Mononucleosis: 30 Years' Experience. BMJ. 1974; 2: 253-256. 10.1136/bmj.2.5913.253 Hjalgrim H., Ekström Smedby K., Rostgaard K., Molin D., Hamilton-Dutoit S., Chang E.T., Ralfkiaer E., Sundstrom C., Adami H.O., Glimelius B. Infectious Mononucleosis, Childhood Social Environment, and Risk of Hodgkin Lymphoma. Cancer Res. 2007; 67: 2382-2388. 10.1158/0008-5472.CAN-06-3566. 17332371 Hjalgrim H., Munksgaard L., Melbye M. Epstein-Barr virus and Hodgkin's lymphoma. Ugeskr. Laeger. 2002; 164: 5924-5927. 12553111 Reiman A., Powell J.E., Flavell K.J., Grundy R.G., Mann J.R., Parkes S., Redfern D., Young L.S., Murray P.G. Seasonal differences in the onset of the EBV-positive and -negative forms of paediatric Hodgkin's lymphoma. Br. J. Cancer. 2003; 89: 1200-1201. 10.1038/sj.bjc.6601277. 14520445 Young L., Yap L.-F., Murray P.G. Epstein–Barr virus: More than 50 years old and still providing surprises. Nat. Cancer. 2016; 16: 789-802. 10.1038/nrc.2016.92. 27687982 Kushekhar K., Berg A.V.D., Nolte I., Hepkema B., Visser L., Diepstra A. Genetic Associations in Classical Hodgkin Lymphoma: A Systematic Review and Insights into Susceptibility Mechanisms. Cancer Epidemiol. Biomarkers Prev. 2014; 23: 2737-2747. 10.1158/1055-9965.EPI-14-0683 Hjalgrim H., Rostgaard K., Johnson P.C.D., Lake A., Shield L., Little A.-M., Ekstrom-Smedby K., Adami H.-O., Glimelius B., Hamilton-Dutoit S. HLA-A alleles and infectious mononucleosis suggest a critical role for cytotoxic T-cell response in EBV-related Hodgkin lymphoma. Proc. Natl. Acad. Sci. USA. 2010; 107: 6400-6405. 10.1073/pnas.0915054107 Cozen W., Timofeeva M.N., Li D., Diepstra A., Hazelett D., Delahaye-Sourdeix M., Edlund C.K., Franke L., Rostgaard K., Berg D.J.V.D. A meta-analysis of Hodgkin lymphoma reveals 19p13.3 TCF3 as a novel susceptibility locus. Nat. Commun. 2014; 5: 1-10. 10.1038/ncomms4856 Niens M., Jarrett R., Hepkema B., Nolte I.M., Diepstra A., Platteel M., Kouprie N., Delury C.P., Gallagher A., Visser L. HLA-A*02 is associated with a reduced risk and HLA-A*01 with an increased risk of developing EBV+ Hodgkin lymphoma. Blood. 2007; 110: 3310-3315. 10.1182/blood-2007-05-086934 Niens M., van den Berg A., Diepstra A., Nolte I.M., van der Steege G., Gallagher A., Taylor G.M., Jarrett R.F., Poppema S., te Meerman G.J. The human leukocyte antigen class I region is associated with EBV-positive Hodgkin's lymphoma: HLA-A and HLA complex group 9 are putative candidate genes. Cancer Epidemiol. Biomark. Prev. 2006; 15: 2280-2284. 10.1158/1055-9965.EPI-06-0476 Glaser S.L., Clarke C.A., Chang E.T., Yang J., Gomez S.L., Keegan T.H. Hodgkin lymphoma incidence in California Hispanics: Influence of nativity and tumor Epstein-Barr virus. Cancer Causes Control. 2014; 25: 709-725. 10.1007/s10552-014-0374-6 Glaser S.L. Hodgkin's disease in black populations: A review of the epidemiologic literature. Semin. Oncol. 1990; 17: 643-659. 2251512 Flavell K.J., Biddulph J.P., Powell J.E., Parkes S.E., Redfern D., Weinreb M., Nelson P., Mann J.R., Young L.S., Murray P.G. South Asian ethnicity and material deprivation increase the risk of Epstein-Barr virus infection in childhood Hodgkin's disease. Br. J. Cancer. 2001; 85: 350-356. 10.1054/bjoc.2001.1872. 11487264 Glaser S.L., Jarrett R.F. The epidemiology of Hodgkin's disease. Baillieres Clin. Haematol. 1996; 9: 401-416. 10.1016/S0950-3536(96)80018-7 Jarrett R.F. Viruses and Hodgkin's lymphoma. Ann. Oncol. 2002; 13(Suppl. S1): 23-29. 10.1093/annonc/13.S1.23 Shankar A., Visaduraki M., Hayward J., Morland B., McCarthy K., Hewitt M. Clinical outcome in children and adolescents with Hodgkin lymphoma after treatment with chemotherapy alone—The results of the United Kingdom HD3 national cohort trial. Eur. J. Cancer. 2012; 48: 108-113. 10.1016/j.ejca.2011.05.029 Keegan T.H., Glaser S.L., Clarke C.A., Gulley M.L., Craig F.E., DiGiuseppe J.A., Dorfman R.F., Mann R.B., Ambinder R.F. Epstein-Barr virus as a marker of survival after Hodgkin's lymphoma: A population-based study. J. Clin. Oncol. 2005; 23: 7604-7613. 10.1200/JCO.2005.02.6310 Jarrett R., Stark G.L., White J., Angus B., Alexander F.E., Krajewski A.S., Freeland J., Taylor G.M., Taylor P.R.A. The Scotland and Newcastle Epidemiology of Hodgkin Disease Study Group Impact of tumor Epstein-Barr virus status on presenting features and outcome in age-defined subgroups of patients with classic Hodgkin lymphoma: A population-based study. Blood. 2005; 106: 2444-2451. 10.1182/blood-2004-09-3759 Clarke C.A., Glaser S.L., Dorfman R.F., Mann R., DiGiuseppe J.A., Prehn A.W., Ambinder R.F. Epstein-Barr virus and survival after Hodgkin disease in a population-based series of women. Cancer. 2001; 91: 1579-1587. 10.1002/1097-0142(20010415)91:8<1579::AID-CNCR1169>3.0.CO;2-L Stark G.L., Wood K.M., Jack F., Angus B., Proctor S.J., Taylor P.R. Northern Region Lymphoma Group Hodgkin's disease in the elderly: A population-based study. Br. J. Haematol. 2002; 119: 432-440. 10.1046/j.1365-2141.2002.03815.x Diepstra A., van Imhoff G.W., Schaapveld M., Karim-Kos H., Berg A.V.D., Vellenga E., Poppema S. Latent Epstein-Barr Virus Infection of Tumor Cells in Classical Hodgkin's Lymphoma Predicts Adverse Outcome in Older Adult Patients. J. Clin. Oncol. 2009; 27: 3815-3821. 10.1200/JCO.2008.20.5138 Wang C., Zou S.-P., Chen D.-G., Wang J.-S., Zheng Y.-B., Chen X.-R., Yang Y. Latent Epstein–Barr virus infection status and prognosis in patients with newly diagnosed Hodgkin lymphoma in Southeast China: A single-center retrospective study. Hematology. 2021; 26: 675-683. 10.1080/16078454.2021.1971864. 34493172 Proctor S.J., Rueffer J.U., Angus B., Breuer K., Flechtner H., Jarrett R., Levis A., Taylor P., Tirelli U. Hodgkin's disease in the elderly: Current status and future directions. Ann. Oncol. 2002; 13: 133-137. 10.1093/annonc/13.S1.133. 12078895 Koh Y.W., Yoon D.H., Suh C., Huh J. Impact of the Epstein–Barr virus positivity on Hodgkin's lymphoma in a large cohort from a single institute in Korea. Ann. Hematol. 2012; 91: 1403-1412. 10.1007/s00277-012-1464-8. 22526365 Kwon J.M., Park Y.H., Kang J.H., Kim K., Ko Y.H., Ryoo B.Y., Lee S.S., Lee S.I., Koo H.H., Kim W.S. The effect of Epstein–Barr virus status on clinical outcome in Hodgkin's lymphoma. Ann. Hematol. 2006; 85: 463-468. 10.1007/s00277-006-0081-9 Glavina-Durdov M., Jakic-Razumovic J., Capkun V., Murray P. Assessment of the prognostic impact of the Epstein–Barr virus-encoded latent membrane protein-1 expression in Hodgkin's disease. Br. J. Cancer. 2001; 84: 1227-1234. 10.1054/bjoc.2001.1774 Murray P.G., Billingham L.J., Hassan H.T., Flavell J.R., Nelson P.N., Scott K., Reynolds G., Constandinou C.M., Kerr D.J., Devey E.C. Effect of Epstein-Barr virus infection on response to chemotherapy and survival in Hodgkin's disease. Blood. 1999; 94: 442-447. 10.1182/blood.V94.2.442.414a46_442_447 Flavell K.J., Billingham L.J., Biddulph J.P., Gray L., Flavell J.R., Constandinou C.M., Young L.S., Murray P.G. The effect of Epstein–Barr virus status on outcome in age- andsex-defined subgroups of patients with advanced Hodgkin's disease. Ann. Oncol. 2003; 14: 282-290. 10.1093/annonc/mdg065 Engel M., Essop M.F., Close P., Hartley P., Pallesen G., Sinclair-Smith C. Improved prognosis of Epstein-Barr virus associated childhood Hodgkin's lymphoma: Study of 47 South African cases. J. Clin. Pathol. 2000; 53: 182-186. 10.1136/jcp.53.3.182 Barros M.H.M., Scheliga A., De Matteo E., Minnicelli C., Soares F.A., Zalcberg I.R., Hassan R. Cell cycle characteristics and Epstein–Barr virus are differentially associated with aggressive and non-aggressive subsets of Hodgkin lymphoma in pediatric patients. Leuk. Lymphoma. 2010; 51: 1513-1526. 10.3109/10428194.2010.489243 Dinand V., Dawar R., Arya L.S., Unni R., Mohanty B., Singh R. Hodgkin's lymphoma in Indian children: Prevalence and significance of Epstein-Barr virus detection in Hodgkin's and Reed-Sternberg cells. Eur. J. Cancer. 2007; 43: 161-168. 10.1016/j.ejca.2006.08.036 Aktas S., Kargı A., Olgun N., Diniz G., Erbay A., Vergin C., Kargi A. Prognostic Significance of Cell Proliferation and Apoptosis-Regulating Proteins in Epstein-Barr Virus Positive and Negative Pediatric Hodgkin Lymphoma. Lymphat. Res. Biol. 2007; 5: 175-182. 10.1089/lrb.2007.5305. 18035936 Chabay P., Lara J., Lorenzetti M., Cambra P., Haab G.A., Aversa L., De Matteo E., Preciado M. Epstein Barr virus in relation to apoptosis markers and patients' outcome in pediatric B-cell Non-Hodgkin lymphoma. Cancer Lett. 2011; 307: 221-226. 10.1016/j.canlet.2011.04.006. 21546156 Claviez A., Tiemann M., Luüders H., Krams M., Parwaresch R., Schellong G., Dorffel W. Impact of latent Epstein-Barr virus infection on outcome in children and adolescents with Hodgkin's lymphoma. J. Clin. Oncol. 2005; 23: 4048-4056. 10.1200/JCO.2005.01.701 Koh Y.W., Han J.H., Yoon D.H., Suh C., Huh J. Epstein-Barr virus positivity is associated with angiogenesis in, and poorer survival of, patients receiving standard treatment for classical Hodgkin's lymphoma. Hematol. Oncol. 2018; 36: 182-188. 10.1002/hon.2468 Enblad G., Sandvej K., Sundstrom C., Pallesen G., Glimelius B. Epstein-Barr virus distribution in Hodgkin's disease in an unselected Swedish population. Acta Oncol. 1999; 38: 425-429. 10.1080/028418699431942. 10418708 Herling M., Rassidakis G.Z., Medeiros L.J., Vassilakopoulos T.P., Kliche K.O., Nadali G., Viviani S., Bonfante V., Giardini R., Chilosi M. Expression of Epstein-Barr virus latent membrane protein-1 in Hodgkin and Reed-Sternberg cells of classical Hodgkin's lymphoma: Associations with presenting features, serum interleukin 10 levels, and clinical outcome. Clin. Cancer Res. 2003; 9: 2114-2120 Axdorph U., Porwit-MacDonald A., Sjöberg J., Grimfors G., Ekman M., Wang W., Biberfeld P., Björkholm M. Epstein–Barr virus expression in Hodgkin's disease in relation to patient characteristics, serum factors and blood lymphocyte function. Br. J. Cancer. 1999; 81: 1182-1187. 10.1038/sj.bjc.6690827 Enblad G., Sandvej K., Lennette E., Sundstrom C., Klein G., Glimelius B., Pallesen G. Lack of correlation between EBV serology and presence of EBV in the Hodgkin and Reed-Sternberg cells of patients with Hodgkin's disease. Int. J. Cancer. 1997; 72: 394-397. 10.1002/(SICI)1097-0215(19970729)72:3<394::AID-IJC3>3.0.CO;2-K Keresztes K., Miltenyi Z., Bessenyei B., Beck Z., Szollosi Z., Nemes Z., Olah E., Illes A. Association between the Epstein-Barr Virus and Hodgkin's Lymphoma in the North-Eastern Part of Hungary: Effects on Therapy and Survival. Acta Haematol. 2006; 116: 101-107. 10.1159/000093639 Krugmann J., Tzankov A., Gschwendtner A., Fischhofer M., Greil R., Fend F., Dirnhofer S. Longer Failure-Free Survival Interval of Epstein-Barr Virus–Associated Classical Hodgkin's Lymphoma: A Single-Institution Study. Mod. Pathol. 2003; 16: 566-573. 10.1097/01.MP.0000071843.09960.BF Naresh K.N., Johnson J., Srinivas V., Soman C.S., Saikia T., Advani S.H., Badwe R.A., Dinshaw K.A., Muckaden M., Magrath I. Epstein—Barr virus association in classical Hodgkin's disease provides survival advantage to patients and correlates with higher expression of proliferation markers in Reed—Sternberg cells. Ann. Oncol. 2000; 11: 91-96. 10.1023/A:1008337100424. 10690394 Morente M.M., Piris M.A., Abraira V., Acevedo A., Aguilera B., Bellas C., Fraga M., Garcia-Del-Moral R., Gomez-Marcos F., Menarguez J. Adverse clinical outcome in Hodgkin's disease is associated with loss of retinoblastoma protein expression, high Ki67 proliferation index, and absence of Epstein-Barr virus-latent membrane protein 1 expression. Blood. 1997; 90: 2429-2436. 9310494 Montalban C., Abraira V., Morente M., Acevedo A., Aguilera B., Bellas C., Fraga M., Del Moral R.G., Menarguez J., Oliva H. Epstein-Barr virus-latent membrane protein 1 expression has a favorable influence in the outcome of patients with Hodgkin's Disease treated with chemotherapy. Leuk. Lymphoma. 2000; 39: 563-572. 10.3109/10428190009113386. 11342339 Trimèche M., Bonnet C., Korbi S., Boniver J., Leval L.D. Association between Epstein-Barr virus and Hodgkin's lymphoma in Belgium: A pathological and virological study. Leuk. Lymphoma. 2007; 48: 1323-1331. 10.1080/10428190701411177. 17613761 Quijano S., Saavedra C., Fiorentino S., Orozco O., Bravo M.M. Epstein-Barr virus presence in Colombian Hodgkin lymphoma cases and its relation to treatment response. Biomedica. 2004; 24: 163-173. 10.7705/biomedica.v24i2.1262 Myriam B.D., Sonia Z., Hanene S., Teheni L., Mounir T. Prognostic significance of Epstein–Barr virus (EBV) infection in Hodgkin lymphoma patients. J. Infect. Chemother. 2017; 23: 121-130. 10.1016/j.jiac.2016.09.004 Santisteban-Espejo A., Perez-Requena J., Atienza-Cuevas L., Moran-Sanchez J., Fernandez-Valle M.D.C., Bernal-Florindo I., Romero-Garcia R., Garcia-Rojo M. Prognostic Role of the Expression of Latent-Membrane Protein 1 of Epstein–Barr Virus in Classical Hodgkin Lymphoma. Viruses. 2021; 132523. 10.3390/v13122523 Elsayed A.A., Asano N., Ohshima K., Izutsu K., Kinoshita T., Nakamura S. Prognostic significance of CD 20 expression and E pstein-B arr virus (EBV) association in classical H odgkin lymphoma in J apan: A clinicopathologic study. Pathol. Int. 2014; 64: 336-345. 10.1111/pin.12175 Souza E.M., Baiocchi O.C., Zanichelli M.A., Alves A.C., Assis M.G., Eiras D.P., Dobo C., Oliveira J.S. Impact of Epstein–Barr virus in the clinical evolution of patients with classical Hodgkin's lymphoma in Brazil. Hematol. Oncol. 2010; 28: 137-141. 10.1002/hon.933 Cheriyalinkal Parambil B., Narula G., Dhamne C., Roy Moulik N., Shet T., Shridhar E., Gujral S., Shah S., Laskar S., Khanna N. Assessment of tumor Epstein-Barr Virus status and its impact on outcomes in intermediate and high-risk childhood classic Hodgkin Lymphoma treated at a tertiary cancer center in India. Leuk. Lymphoma. 2020; 61: 3217-3225. 10.1080/10428194.2020.1800005 Vestlev P.M., Pallesen G., Sandvej K., Hamilton-Duroit S.J., Bendtzen S.M. Prognosis of Hodgkin's disease in not influenced by epstein-barry virus latent membrane protein. Int. J. Cancer. 1992; 50: 670-671. 10.1002/ijc.2910500432. 1311289 Armstrong A., Lennard A., Alexander F., Angus B., Proctor S., Onions D., Jarrett R. Prognostic significance of Epstein-Barr virus association in Hodgkin's disease. Eur. J. Cancer. 1994; 30: 1045-1046. 10.1016/0959-8049(94)90157-0 Levy A., Diomin V., Gopas J., Ariad S., Sacks M., Benharroch D. Hodgkin's lymphoma in the Bedouin of southern Israel: Epidemiological and clinical features. Isr. Med. Assoc. J. 2000; 2: 501-503. 10979320 Vassallo J., Metze K., Traina F.A., de Souza C., Lorand-Metze I. The prognostic relevance of apoptosis-related proteins in classical Hodgkin's lymphomas. Leuk. Lymphoma. 2003; 44: 483-488. 10.1080/1042819021000037958 Lee E.K., Kim S.Y., Noh K.-W., Joo E.H., Zhao B., Kieff E., Kang M.-S. Small molecule inhibition of Epstein–Barr virus nuclear antigen-1 DNA binding activity interferes with replication and persistence of the viral genome. Antivir. Res. 2014; 104: 73-83. 10.1016/j.antiviral.2014.01.018 Eichenauer D.A., Aleman B.M.P., André M., Federico M., Hutchings M., Illidge T., Engert A., Ladetto M. ESMO Guidelines Committee Hodgkin lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018; 29: iv19-iv29. 10.1093/annonc/mdy080 Kaseb H., Babiker H.M. Hodgkin Lymphoma. StatPearls; StatPearls Publishing: Treasure Island, FL, USA. 2022 Cui X., Snapper C.M. Epstein Barr Virus: Development of Vaccines and Immune Cell Therapy for EBV-Associated Diseases. Front. Immunol. 2021; 12: 734471. 10.3389/fimmu.2021.734471 Toner K., Bollard C.M. EBV+ lymphoproliferative diseases: Opportunities for leveraging EBV as a therapeutic target. Blood. 2022; 139: 983-994. 10.1182/blood.2020005466. 34437680 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. 10.1200/JCO.20.01709 Wu R., Sattarzadeh A., Rutgers B., Diepstra A., Berg A.V.D., Visser L. The microenvironment of classical Hodgkin lymphoma: Heterogeneity by Epstein–Barr virus presence and location within the tumor. Blood Cancer, J. 2016; 6: e417. 10.1038/bcj.2016.26 Barros M.H.M., Vera-Lozada G., Soares F.A., Niedobitek G., Hassan R. Tumor microenvironment composition in pediatric classical Hodgkin lymphoma is modulated by age and Epstein-Barr virus infection. Int. J. Cancer. 2011; 131: 1142-1152. 10.1002/ijc.27314. 22025264 Jimenez O., Barros M.H., De Matteo E., Lombardi M.G., Preciado M.V., Niedobitek G., Chabay P. M1-like macrophage polarization prevails in young children with classic Hodgkin Lymphoma from Argentina. Sci. Rep. 2019; 9: 1-6. 10.1038/s41598-019-49015-1 Jimenez O., Colli S., Lombardi M.G., Preciado M.V., De Matteo E., Chabay P. Epstein–Barr virus recruits PDL1-positive cells at the microenvironment in pediatric Hodgkin lymphoma. Cancer Immunol. Immunother. 2020; 70: 1519-1526. 10.1007/s00262-020-02787-2 Satoh T., Wada R., Yajima N., Imaizumi T., Yagihashi S. Tumor Microenvironment and RIG-I Signaling Molecules in Epstein Barr Virus-Positive and -Negative Classical Hodgkin Lymphoma of the Elderly. J. Clin. Exp. Hematop. 2014; 54: 75-84. 10.3960/jslrt.54.75. 24942949 Assis M.C.G., Campos A.H.F.M., de Oliveira J.S.R., Soares F.A., Silva J.M.K., Silva P.B., Penna A.D., Souza E.M., Baiocchi O.C.G. Increased expression of CD4+CD25+FOXP3+ regulatory T cells correlates with Epstein–Barr virus and has no impact on survival in patients with classical Hodgkin lymphoma in Brazil. Med. Oncol. 2012; 29: 3614-3619. 10.1007/s12032-012-0299-4. 22791223 Ozturk V., Yikilmaz A.S., Kilicarslan A., Bakanay S.M., Akinci S., Dilek I. The Triple Positivity for EBV, PD-1, and PD-L1 Identifies a Very High Risk Classical Hodgkin Lymphoma. Clin. Lymphoma Myeloma Leuk. 2020; 20: e375-e381. 10.1016/j.clml.2019.11.021 Sueur C., Lupo J., Mas P., Morand P., Boyer V. Difference in cytokine production and cell cycle progression induced by Epstein-Barr virus Lmp1 deletion variants in Kmh2, a Hodgkin lymphoma cell line. Virol. J. 2014; 11: 94. 10.1186/1743-422X-11-94 IIncrocci R., McAloon J., Montesano M., Bardahl J., Vagvala S., Stone A., Swanson-Mungerson M. Epstein-Barr virus LMP2A utilizes Syk and PI3K to activate NF-kappaB in B-cell lymphomas to increase MIP-1alpha production. J. Med. Virol. 2019; 91: 845-855. 10.1002/jmv.25381 Baumforth K.R., Birgersdotter A., Reynolds G.M., Wei W., Kapatai G., Flavell J.R., Kalk E., Piper K., Lee S., Machado L. Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin's lymphoma cells mediates Up-regulation of CCL20 and the migration of regulatory T cells. Am. J. Pathol. 2008; 173: 195-204. 10.2353/ajpath.2008.070845 Kis L.L., Salamon D., Persson E.K., Nagy N., Scheeren F.A., Spits H., Klein G., Klein E. IL-21 imposes a type II EBV gene expression on type III and type I B cells by the repression of C- and activation of LMP-1-promoter. Proc. Natl. Acad. Sci. USA. 2009; 107: 872-877. 10.1073/pnas.0912920107 Kis L.L., Takahara M., Nagy N., Klein G., Klein E. Cytokine mediated induction of the major Epstein–Barr virus (EBV)-encoded transforming protein, LMP-1. Immunol. Lett. 2006; 104: 83-88. 10.1016/j.imlet.2005.11.003. 16386314

By Mahdi Nohtani; Katerina Vrzalikova; Maha Ibrahim; Judith E. Powell; Éanna Fennell; Susan Morgan; Richard Grundy; Keith McCarthy; Sarah Dewberry; Jan Bouchal; Katerina Bouchalova; Pamela Kearns and Paul G. Murray

Reported by Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author

Titel:
Impact of Tumour Epstein-Barr Virus Status on Clinical Outcome in Patients with Classical Hodgkin Lymphoma (cHL): A Review of the Literature and Analysis of a Clinical Trial Cohort of Children with cHL.
Autor/in / Beteiligte Person: Nohtani, M ; Vrzalikova, K ; Ibrahim, M ; Powell, JE ; Fennell, É ; Morgan, S ; Grundy, R ; McCarthy, K ; Dewberry, S ; Bouchal, J ; Bouchalova, K ; Kearns, P ; Murray, PG
Link:
Zeitschrift: Cancers, Jg. 14 (2022-09-01), Heft 17
Veröffentlichung: Basel, Switzerland : MDPI, 2022
Medientyp: academicJournal
ISSN: 2072-6694 (print)
DOI: 10.3390/cancers14174297
Sonstiges:
  • Nachgewiesen in: MEDLINE
  • Sprachen: English
  • Publication Type: Journal Article
  • Language: English
  • [Cancers (Basel)] 2022 Sep 01; Vol. 14 (17). <i>Date of Electronic Publication: </i>2022 Sep 01.
  • References: N Engl J Med. 1989 Mar 16;320(11):689-95. (PMID: 2537928) ; Blood. 2008 Jan 1;111(1):292-301. (PMID: 17720884) ; Cancer Res. 1974 May;34(5):1172-8. (PMID: 4842360) ; Eur J Cancer. 2007 Jan;43(1):161-8. (PMID: 17113770) ; Br J Cancer. 1999 Dec;81(7):1182-7. (PMID: 10584880) ; Blood Cancer J. 2016 May 06;6:e417. (PMID: 27152844) ; Blood. 2005 Dec 15;106(13):4249-52. (PMID: 16123211) ; Ann Oncol. 2018 Oct 1;29(Suppl 4):iv19-iv29. (PMID: 29796651) ; Leuk Lymphoma. 2020 Dec;61(13):3217-3225. (PMID: 32729791) ; Cancer. 2001 Apr 15;91(8):1579-87. (PMID: 11301409) ; Ann Oncol. 2003 Feb;14(2):282-90. (PMID: 12562657) ; J Clin Oncol. 2021 Feb 10;39(5):514-524. (PMID: 33434061) ; Hematology. 2021 Dec;26(1):675-683. (PMID: 34493172) ; Isr Med Assoc J. 2000 Jul;2(7):501-3. (PMID: 10979320) ; Ann Hematol. 2006 Jul;85(7):463-8. (PMID: 16534596) ; J Clin Oncol. 2009 Aug 10;27(23):3815-21. (PMID: 19470931) ; Antiviral Res. 2014 Apr;104:73-83. (PMID: 24486954) ; Blood. 1994 Dec 1;84(11):3792-8. (PMID: 7949135) ; Cancer Epidemiol Biomarkers Prev. 2014 Dec;23(12):2737-47. (PMID: 25205514) ; Blood. 2005 Dec 15;106(13):4339-44. (PMID: 16076866) ; Immunity. 1998 Sep;9(3):405-11. (PMID: 9768760) ; Cancer. 1971 Feb;27(2):416-21. (PMID: 4322464) ; Hematol Oncol. 2010 Sep;28(3):137-41. (PMID: 20128016) ; J Exp Med. 1993 Feb 1;177(2):339-49. (PMID: 8381153) ; Cancer Epidemiol Biomarkers Prev. 2006 Nov;15(11):2280-4. (PMID: 17119058) ; J Med Virol. 2019 May;91(5):845-855. (PMID: 30609049) ; Hematol Oncol. 2018 Feb;36(1):182-188. (PMID: 28744882) ; Viruses. 2021 Dec 15;13(12):. (PMID: 34960792) ; Biomedica. 2004 Jun;24(2):163-73. (PMID: 15495596) ; J Clin Pathol. 2000 Mar;53(3):182-6. (PMID: 10823135) ; Front Immunol. 2021 Oct 08;12:734471. (PMID: 34691042) ; Leuk Lymphoma. 2007 Jul;48(7):1323-31. (PMID: 17613761) ; Cell. 1986 Dec 26;47(6):883-9. (PMID: 3022942) ; Blood. 2007 Nov 1;110(9):3310-5. (PMID: 17630352) ; Leuk Lymphoma. 2000 Nov;39(5-6):563-72. (PMID: 11342339) ; J Infect Chemother. 2017 Mar;23(3):121-130. (PMID: 28034523) ; Leuk Lymphoma. 2010 Aug;51(8):1513-22. (PMID: 20687799) ; Baillieres Clin Haematol. 1996 Sep;9(3):401-16. (PMID: 8922237) ; Med Oncol. 2012 Dec;29(5):3614-9. (PMID: 22791223) ; Proc Natl Acad Sci U S A. 2010 Jan 12;107(2):872-7. (PMID: 20080768) ; J Clin Exp Hematop. 2014;54(1):75-84. (PMID: 24942949) ; Mod Pathol. 2003 Jun;16(6):566-73. (PMID: 12808062) ; Ann Hematol. 2012 Sep;91(9):1403-12. (PMID: 22526365) ; Br J Cancer. 2003 Oct 6;89(7):1200-1. (PMID: 14520445) ; Leukemia. 2005 Jun;19(6):936-44. (PMID: 15912144) ; Acta Oncol. 1999;38(4):425-9. (PMID: 10418708) ; J Clin Oncol. 2005 Jun 20;23(18):4048-56. (PMID: 15961758) ; Blood. 1990 Apr 1;75(7):1589-90. (PMID: 2156583) ; Virol J. 2014 May 19;11:94. (PMID: 24886620) ; Cancer Res. 2007 Mar 1;67(5):2382-8. (PMID: 17332371) ; Semin Oncol. 1990 Dec;17(6):643-59. (PMID: 2251512) ; Ann Oncol. 2000 Jan;11(1):91-6. (PMID: 10690394) ; Clin Cancer Res. 2003 Jun;9(6):2114-20. (PMID: 12796376) ; Ann Oncol. 2002;13 Suppl 1:133-7. (PMID: 12078895) ; Blood. 1999 Jul 15;94(2):442-7. (PMID: 10397711) ; Cancer Causes Control. 2014 Jun;25(6):709-25. (PMID: 24722952) ; Ugeskr Laeger. 2002 Dec 9;164(50):5924-7. (PMID: 12553111) ; Leuk Lymphoma. 2003 Mar;44(3):483-8. (PMID: 12688319) ; PLoS Pathog. 2012;8(5):e1002697. (PMID: 22589726) ; J Pathol. 2005 Mar;205(4):498-506. (PMID: 15714459) ; Ann Oncol. 2002;13 Suppl 1:23-9. (PMID: 12078898) ; Proc Natl Acad Sci U S A. 1994 Nov 8;91(23):10962-6. (PMID: 7971992) ; Cell Signal. 2003 Jan;15(1):9-16. (PMID: 12401515) ; Clin Lymphoma Myeloma Leuk. 2020 Jul;20(7):e375-e381. (PMID: 32295735) ; Am J Pathol. 2008 Jul;173(1):195-204. (PMID: 18502823) ; Immunol Lett. 2006 Apr 15;104(1-2):83-8. (PMID: 16386314) ; J Clin Invest. 1997 Dec 15;100(12):2961-9. (PMID: 9399941) ; Sci Rep. 2019 Sep 3;9(1):12687. (PMID: 31481738) ; Nat Commun. 2014 Jun 12;5:3856. (PMID: 24920014) ; Blood. 2005 Oct 1;106(7):2444-51. (PMID: 15941916) ; Int Rev Immunol. 2001;20(6):805-35. (PMID: 11913951) ; Int J Cancer. 2012 Sep 1;131(5):1142-52. (PMID: 22025264) ; J Virol. 2015 Oct 14;90(1):345-55. (PMID: 26468528) ; Eur J Cancer. 1994;30A(7):1045-6. (PMID: 7946575) ; Nat Rev Cancer. 2016 Dec;16(12):789-802. (PMID: 27687982) ; Curr Opin Virol. 2012 Dec;2(6):733-9. (PMID: 23031715) ; Scientifica (Cairo). 2012;2012:438204. (PMID: 24278697) ; Lymphat Res Biol. 2007;5(3):175-82. (PMID: 18035936) ; Br Med J. 1974 May 4;2(5913):253-6. (PMID: 4406463) ; Cancer Lett. 2011 Aug 28;307(2):221-6. (PMID: 21546156) ; Viruses. 2012 Sep;4(9):1537-47. (PMID: 23170171) ; J Exp Med. 1996 Oct 1;184(4):1495-505. (PMID: 8879220) ; Blood. 2022 Feb 17;139(7):983-994. (PMID: 34437680) ; Int J Cancer. 1997 Jul 29;72(3):394-7. (PMID: 9247279) ; Eur J Cancer. 2012 Jan;48(1):108-13. (PMID: 21703849) ; J Pathol. 1991 Aug;164(4):291-7. (PMID: 1656004) ; Pathol Int. 2014 Jul;64(7):336-45. (PMID: 25047504) ; Int J Cancer. 1992 Feb 20;50(4):670-1. (PMID: 1311289) ; Br J Haematol. 2002 Nov;119(2):432-40. (PMID: 12406082) ; J Clin Oncol. 2005 Oct 20;23(30):7604-13. (PMID: 16186595) ; Blood. 1997 Aug 15;90(4):1664-72. (PMID: 9269787) ; Acta Haematol. 2006;116(2):101-7. (PMID: 16914904) ; J Pathol. 1992 Jan;166(1):1-5. (PMID: 1311374) ; Br J Cancer. 2001 Aug 3;85(3):350-6. (PMID: 11487264) ; Oncogene. 2007 Jun 14;26(28):4135-47. (PMID: 17486072) ; Blood. 2005 Dec 15;106(13):4345-50. (PMID: 16131568) ; Br J Cancer. 2001 May 4;84(9):1227-34. (PMID: 11336475) ; Proc Natl Acad Sci U S A. 2010 Apr 6;107(14):6400-5. (PMID: 20308568) ; Blood. 2012 Nov 1;120(18):3750-5. (PMID: 22972983) ; Blood. 1997 Sep 15;90(6):2429-36. (PMID: 9310494) ; Cancer Immunol Immunother. 2021 Jun;70(6):1519-1526. (PMID: 33184699)
  • Contributed Indexing: Keywords: Epstein–Barr virus (EBV); classic Hodgkin lymphoma (cHL); clinical trial
  • Entry Date(s): Date Created: 20220909 Latest Revision: 20220913
  • Update Code: 20231215
  • PubMed Central ID: PMC9454639

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -