Next Article in Journal
A Comprehensive Analysis of Tn and STn Antigen Expression in Esophageal Adenocarcinoma
Previous Article in Journal
Metabolomics, Transcriptome and Single-Cell RNA Sequencing Analysis of the Metabolic Heterogeneity between Oral Cancer Stem Cells and Differentiated Cancer Cells
Previous Article in Special Issue
Optimizing Treatment for Relapsed/Refractory Classic Hodgkin Lymphoma in the Era of Immunotherapy
 
 
cancers-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Impact of Serum β2-Microglobulin Levels in Hodgkin Lymphoma Treated with ABVD or Equivalent Regimens: A Comprehensive Analysis of 915 Patients

by
Theodoros P. Vassilakopoulos
1,*,†,
Maria Arapaki
1,†,
Panagiotis T. Diamantopoulos
2,
Athanasios Liaskas
1,
Fotios Panitsas
1,
Marina P. Siakantaris
1,
Maria Dimou
1,
Styliani I. Kokoris
1,
Sotirios Sachanas
1,
Marina Belia
1,
Chrysovalantou Chatzidimitriou
1,
Elianna A. Konstantinou
1,
John V. Asimakopoulos
1,
Kyriaki Petevi
1,
George Boutsikas
1,
Alexandros Kanellopoulos
1,
Alexia Piperidou
1,
Maria-Ekaterini Lefaki
1,
Angeliki Georgopoulou
1,
Anastasia Kopsaftopoulou
1,
Kalliopi Zerzi
1,
Ioannis Drandakis
1,
Maria N. Dimopoulou
1,
Marie-Christine Kyrtsonis
3,
Panayiotis Tsaftaridis
1,
Eleni Plata
1,
Eleni Variamis
2,
Gerassimos Tsourouflis
4,
Flora N. Kontopidou
5,
Kostas Konstantopoulos
1,
Gerassimos A. Pangalis
1,
Panayiotis Panayiotidis
1,‡ and
Maria K. Angelopoulou
1,‡
add Show full author list remove Hide full author list
1
Department of Haematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, School of Medicine, Laikon General Hospital, 17 Ag. Thoma Str., 11527 Athens, Greece
2
First Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Laikon General Hospital, 17 Ag. Thoma Str., 11527 Athens, Greece
3
First Department of Internal Medicine Propedeutic, National and Kapodistrian University of Athens, School of Medicine, Laikon General Hospital, 17 Ag. Thoma Str., 11527 Athens, Greece
4
Second Department of Surgery Propedeutic, National and Kapodistrian University of Athens, Laikon General Hospital, 11527Athens, Greece
5
Second Department of Internal Medicine, National and Kapodistrian University of Athens, Ippokration General Hospital, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
These authors contributed equally to this study.
These authors contributed equally to this study.
Cancers 2024, 16(2), 238; https://doi.org/10.3390/cancers16020238
Submission received: 4 December 2023 / Revised: 26 December 2023 / Accepted: 29 December 2023 / Published: 5 January 2024
(This article belongs to the Special Issue Hodgkin Lymphoma: Present Status and Future Strategies)

Abstract

:

Simple Summary

The significance of serum beta-2 microglobulin (sβ2m) in Hodgkin lymphoma (HL) is controversial. In an effort to investigate the prognostic significance of sβ2m levels in a large series of patients with HL, we analyzed 915 patients, who were treated with ABVD or equivalent regimens with or without radiotherapy. Sβ2m levels were measured by a radioimmunoassay (upper normal limit 2.4 mg/L). The median sβ2m levels were 2.20 mg/L. Freedom from progression (FFP) was significantly inferior in patients with a higher sβ2m at all tested cutoffs. The best cutoff was 2.0 mg/L (10-year FFP 83% vs. 70%, p = 0.001), which performed better than the 2.4 mg/L cutoff (“normal versus high”). Our data suggest that higher sβ2m is a significant independent predictor of FFP, OS and HLSS in HL but the optimal cutoff appears to lie within the normal limits in this predominantly young patient population.

Abstract

The significance of serum beta-2 microglobulin (sβ2m) in Hodgkin lymphoma (HL) is controversial. We analyzed 915 patients with HL, who were treated with ABVD or equivalent regimens with or without radiotherapy. Sβ2m levels were measured by a radioimmunoassay (upper normal limit 2.4 mg/L). Sequential cutoffs (1.8–3.0 by 0.1 mg/L increments, 3.5 and 4.0 mg/L) were tested along with ROC analysis. The median sβ2m levels were 2.20 mg/L and were elevated (>2.4 mg/L) in 383/915 patients (41.9%). Higher sβ2m was associated with inferior freedom from progression (FFP) at all tested cutoffs. The best cutoff was 2.0 mg/L (10-year FFP 83% vs. 70%, p = 0.001), which performed better than the 2.4 mg/L cutoff (“normal versus high”). In multivariate analysis, sβ2m > 2.0 mg/L was an independent adverse prognostic factor in the whole patient population. In multivariate overall survival analysis, sβ2m levels were predictive at 2.0 mg/L cutoff in the whole patient population and in advanced stages. Similarly, sβ2m > 2.0 mg/L independently predicted inferior HL-specific survival in the whole patient population. Our data suggest that higher sβ2m is an independent predictor of outcome in HL but the optimal cutoff lies within the normal limits (i.e., at 2.0 mg/L) in this predominantly young patient population, performing much better than a “normal versus high” cutoff set at 2.4 mg/L.

1. Introduction

The prognosis of Hodgkin lymphoma (HL) has dramatically changed over the last few decades, with the 5-year survival rate below 10% in the 1960s increasing to a 10-year survival rate exceeding 80% in the 2010s [1,2]. A further increase is expected with the use of novel immunotherapies for relapsed/refractory disease [3,4,5,6] or even their incorporation in earlier treatment lines [7,8]. The prognosis primarily depends on clinical stage as defined by the anatomic extent of the disease and the presence of B-symptoms according to the Ann Arbor staging system [9] and the Cotswolds [10] and Lugano modifications [11]. In early-stage disease, the presence of bulky mediastinal disease, the number of involved nodal sites, elevated erythrocyte sedimentation rate (ESR), extranodal involvement and age provide additional prognostic information [12,13,14,15,16,17,18,19,20,21,22,23,24], while the 7-factor international prognostic score (IPS) has become the standard prognostic tool for advanced stages [25] followed by simplified versions [26,27]. Another 7-factor advanced-stage Hodgkin lymphoma International Prognostic Index was recently published by the HoLISTIC consortium including significantly overlapping factors compared to IPS, albeit handled in a totally different logistic way [28].
Unfortunately, the above and other conventional prognostic systems cannot accurately classify patients with highly divergent levels of risk of relapse/progression and are unable to define either a very low-risk subgroup or any sizeable subgroup of patients with a>40-50% failure rate under modern ABVD-like fixed or Positron Emission Tomography driven therapy [25,26,27,28,29,30]. Many biological prognostic factors have been evaluated in this context, but none has been adopted in everyday prognostication for several reasons [30]. Thus, research is still focusing to the identification of novel, powerful, conventional and biological prognostic factors which might permit the reduction in chemotherapy and omission radiotherapy (RT) in an effort to minimize the long-term toxic effects in low-risk patients and guide the intensification/modification of treatment in high-risk groups.
Serum beta2-microglobulin (sβ2m) is a well-established prognostic factor in multiple myeloma, and has been incorporated in the international staging system (ISS) [31]. It is also an extensively evaluated prognostic factor in diffuse large B-cell lymphoma and other non-Hodgkin lymphoma subtypes [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46] and may work in acute myeloid leukemia as well [47], but has not been incorporated in current prognostic models for these diseases. Although tested as a prognostic factor in HL 30 years ago [48], its role has not yet been fully established; thus, sβ2m has not been used in any of the current prognostic systems for HL. However, there are several small- to medium-sized studies evaluating the potential prognostic role of sβ2m levels in patients with HL using heterogenous therapy, different endpoints, and various cutoffs with conflicting results [48,49,50,51,52,53,54,55,56]. The data from our group, published in 2002 and 2005 based on patients treated with ABVD or equivalent regimens, were also conflicting [52,53]. The updated analysis of 379 patients in 2005 demonstrated a role of sβ2m in predicting overall survival and also in predicting failure-free survival in the early stages only [53].
At this point, we extended our series to include 915 patients treated optimally with ABVD or equivalent regimens with or without RT with a much longer median follow-up of approximately 9 years extending up to almost 30 years. The size of this population is by far the largest ever recruited and permits the extraction of much more reliable conclusions. It also enables the reliable evaluation of multiple cutoffs, since the prognostic significance of sβ2m levels is not necessarily evident in a “normal versus elevated” analysis and optimal cutoffs for clinical use may also be different in different disease stages.

2. Patients and Methods

2.1. Patients, Staging, Treatment Strategies and Laboratory Assays

We analyzed 915 patients who received a diagnosis and first-line treatment for HL between 1990 and 2018, and had available sβ2m levels at diagnosis. The study period was extended from the beginning of sβ2m-level determination in clinical practice until the change in the method of sβ2m measurement implying a different cutoff in 2018. All patients were older than 14 years, were HIV-negative, and had received treatment with anthracycline-based CT with or without RT. In this retrospective study, patients were selected solely based on the availability of pretreatment sβ2m levels and their characteristics were comparable with those of patients who had also received anthracycline-based chemotherapy with or without RT during the same period, but did not have available serum β2-microglobulin levels, as previously reported [52,53].
All patients were clinically staged according to the Ann Arbor system [9], using standard staging procedures. Clinical Ann Arbor stages (AAS) IA and IIA were considered early, while clinical stages IB, IIB, III and IV were considered advanced for the purposes of this analysis. The number of involved anatomic sites was determined as described in previous publication of our group. Hemoglobin, white blood cell counts, and the differential erythrocyte sedimentation rate (ESR), serum albumin and serum LDH levels were measured by standard assays. Anemia was defined as the presence of hemoglobin levels <13 g/dL for males and <11.5 g/dL for females. Serum albumin was analyzed at a cutoff of 4 g/dL, as proposed by the IPS [25]. Severe lymphopenia was also defined according to the cutoff provided by the IPS (<0.6 × 109/L or <8%) [25].
Treatment strategies for early (IA, IIA) and advanced AAS (IB, IIB, III, IV) patients have been described previously [52,57]. PET-driven strategies have been adopted during the last 15 years, initially for advanced- and later for early-stage disease. The evolution of treatment strategies during the study period has been described in recent publications of our group [58,59].
Sβ2m was measured using a radioimmunoassay (Pharmacia). The range of normal values was 1.0–2.4 mg/L.
The study was approved by the appropriate Institutional Review Board. As a non-interventional retrospective study, informed consent was waived.

2.2. Statistical Analysis

The frequency of elevated sβ2m levels among various subgroups of patients were compared by the chi-square test. The Mann–Whitney and Kruskal–Wallis tests were used for non-parametric comparisons, as appropriate. The correlation between sβ2m levels and other variables evaluated as continuous was estimated by the Spearman’s rho coefficient. The optimal cutoff for sβ2m levels was determined by direct testing of sequential cutoffs and by the use of Receiver Operator Curves (ROCs). The results obtained by both approaches were very similar.
Freedom from progression (FFP) was defined as the time interval between treatment initiation and treatment failure or last follow-up. Treatment failure was defined as the inability to achieve complete or partial remission (CR, PR) during initial therapy, requiring a switch to alternative chemotherapy, or relapse/progression after an initial CR/PR or toxic death. Patients with deaths of unrelated causes were censored. Overall survival (OS) and Hodgkin lymphoma-specific survival (HLSS) were measured from treatment initiation to death from any cause or HL-related causes (progressive HL, death of treatment toxicity), respectively, or last follow-up. Deaths due to secondary malignancies or cardiovascular causes during CR were censored. Survival after failure (SAF) was defined as the time interval between the documentation of treatment failure (primary failure or relapse) and death from any cause or last follow-up. The estimation of actuarial FFP or survival was performed using the Kaplan–Meier method [60]. The identification of prognostic factors in univariate analysis was based on the log-rank test [61]. The identification of independent prognostic factors was performed using Cox’s proportional hazards model [62].

3. Results

3.1. Patients’ Characteristics

The median age of the patients was 32 years (14–86) and 513 (56.1%) were males. Among 915 patients, 515 (56.3%) had early- and 400 (33.7%) had advanced-stage disease, while 304 (33.2%) had B-symptoms. The histologic subtype of 891 patients with recorded information was nodular sclerosis in 610 (68.5%), mixed cellularity in 173 (19.4%), nodular lymphocyte predominance in 44 (4.9%), lymphocyte rich classical in 39 (4.4%), lymphocyte depletion in 3 (0.3%), and classical HL unclassified, overlapping or interfollicular in 22 (2.4%). In general, patients’ characteristics were compatible with other reported unselected series of patients with non-pediatric HL. As patients had been diagnosed between 1990 and 2018, the median follow-up of those who were alive at the time of the analysis, was 105.1 months (1.6–353.7).

3.2. Serum β2-Microglobulin Levels and Clinicopathologic Correlations

The median observed sβ2m levels were 2.20 mg/L, with an interquartile range (IQR) of 1.80-3.00 mg/L and a range of 0.50-14.40 mg/L. Elevated sβ2m levels (>2.40 mg/L) were found in 383/915 patients (41.9%).
The correlation between sβ2m levels and other potential prognostic factors is shown in Table 1. Sβ2m levels correlated strongly with all baseline features, including demographics (older age, male gender), non-nodular sclerosing classical HL, clinical and laboratory markers of disease extent and aggressiveness and the IPS (all p-values <0.001), with only correlations with leukocytosis, iliac/inguinal and lung involvement being looser but still statistically significant.
With respect to potential biological prognostic factors, highly significant correlations of moderate magnitude were observed between sβ2m and serum soluble CD30, serum interleukin-10 and serum ferritin (p < 0.001 but Spearman’s rho 0.333–0.455), as summarized in Table 2. However, there were no significant associations with bcl-2, activated caspase-3 or Epstein–Barr virus Latent Protein-1 (LMP-1) immunohistochemical expression.

3.3. Freedom from Progression

The 10-year FFP rate for the whole series was 76%. Among 208 events, only 3 were toxic deaths, while 205 were related to progressive or relapsing disease. As expected, most of the potential prognostic factors listed in Table 1 were statistically significant in the univariate analysis of FFP at the level of ≤0.001, with the exception of iliac/inguinal involvement (p = 0.047), age, gender, histology, leukocytosis, lung involvement and the number of nodal sites (in advanced disease only), which were not significant.

3.3.1. All Patients

When sβ2m levels were classified as quartiles, a consistent drop of 5–6% was observed for each one from Q1 to Q4, with 10-year rates of 84%, 78%, 73% and 68% (p = 0.001, Figure 1A), indicating a “dose–response” effect.
Patients with elevated sβ2m levels had inferior FFP (70% versus 80%, p = 0.001, Figure 1B). As the “normal versus elevated” comparison is arbitrary and may not be optimal, several cutoff points for sβ2m levels were evaluated to identify the optimal cutoff to predict FFP, starting from 1.8 mg/L and advancing in 0.1 mg/L steps up to 3.0 mg/L and then at 0.5 mg/L steps up to 4.0 mg/L. In the univariate analysis, FFP was significantly inferior in patients with higher sβ2m at all tested cutoffs, as shown in Table 3. Interestingly, the 2.4 mg/L cutoff (“normal versus elevated”) was not the best one, as the widest difference was observed at the cutoff of 2.0 mg/L(10-year FFP 83% versus 70%, p < 0.001; Figure 1C). ROC curve analysis confirmed this finding and provided a best cutoff at 2.02 mg/L. The area under the curve (AUC) was 0.573 (95% CI 0.53–0.62; p = 0.01).
2m levels >2.0 mg/L were an independent adverse prognostic factor in the large-scale multivariate analysis (see Table 4 footnote) of all 915 patients, along with stage and lymphocytopenia (hazard ratio (HR) 1.55, 95% confidence intervals (CI) 1.11–2.17, p = 0.01; Table 4). The “normal versus elevated” comparison was not significant in the multivariate analysis (Table 4).

3.3.2. Early Stages

Among 515 patients with early-stage HL (IA/IIA), the best cutoff was found at 1.9 mg/L, with 10-year FFP rates of 88% versus 78% (p = 0.003, Figure 1D). Significant results were also obtained at the cutoff of 2.0 mg/L, with 10-year FFP rates of 86% versus 78% (p = 0.007, Figure 1E). As shown in Table 3, cutoffs set at 2.2 mg/L or higher, including the “normal versus elevated” comparison were not predictive of FFP. Sβ2m levels > 2.0 mg/L were an independent adverse prognostic factor in a large-scale multivariate analysis of patients with early stages along with ≥3 nodal sites and ESR ≥ 50 mm/h (hazard ratio (HR) 1.65, 95% confidence intervals (CI) 1.04–2.62, and p = 0.034; Table 4). Unexpectedly, the “normal versus elevated” comparison was also significant in multivariate analysis with a similar HR (Table 4).

3.3.3. Advanced Stages

Among 400 patients with advanced-stage HL (IB/IIB/III/IV), none of the tested cutoffs, including the “normal versus elevated” comparison, were predictive of FFP in univariate analysis (Table 3). The best cutoff was set at 2.0 mg/Land resulted in a marginally significant prediction, with 10-year FFP rates of 74% versus 64% (p = 0.09, Figure 1F). Similarly to the univariate results, sβ2m levels > 2.0 mg/L were an independent adverse prognostic factor of borderline significance in the multivariate analysis of patients with advanced stages, including all the IPS factors (Table 4; see also footnote) along with stage IV, lymphopenia and leukocytosis (protective!!) (hazard ratio (HR) 1.44, 95% CI 0.94–2.21, and p = 0.098; Table 4). The “normal versus elevated” comparison was not significant in multivariate analysis (Table 4).

3.4. Overall Survival

The 10-year OS rate for the whole series was 85%. Among 131 deaths, 74 were disease-related and 57 unrelated.

3.4.1. All Patients

When sβ2m levels were classified as quartiles, a gradual drop was observed for each one from Q1 to Q4, with 10-year rates of 95%, 87%, 85% and 71% (p < 0.001; Figure 2A).
Patients with elevated sβ2m levels had inferior OS (90% versus 77%, p < 0.001; Figure 2B). At the cutoff of 2.0 mg/L, the difference was similar (92% versus 79%, p < 0.001; Figure 2C). Sβ2m levels > 2.0 mg/L were an independent adverse prognostic factor in a large-scale multivariate analysis of all 915 patients (see Table 2 footnote), along with older age, B-symptoms and lymphocytopenia (hazard ratio (HR) 1.96, 95% CI 1.21–3.19, and p = 0.006; Table 4). The “normal versus elevated” comparison yielded borderline results in the multivariate analysis (Table 4).

3.4.2. Early and Advanced Stages

In the multivariate analysis of OS in early-stage patients, sβ2m levels were neither an independent prognostic factor at the cutoff of 2.0 mg/L nor at a “normal versus elevated” basis (Table 4). In contrast, in advanced stages, sβ2m > 2.0 mg/L was an independent adverse prognostic factor along with older age, lymphopenia, anemia and a lack of leukocytosis (hazard ratio (HR) 2.07, 95% CI1.04-4.15, and p = 0.039; Table 4). The “normal versus elevated” comparison was not predictive in advanced-stage disease (Table 4).

3.5. Causes of Death, Hodgkin Lymphoma-Specific Survival and Survival after Failure

Up to publication of this study, 131 deaths were recorded. Among them, 74 (56%) were due to HL, with 58 being directly related to progressive HL, 5 toxic deaths (3 during first-line and 2 during salvage therapy), 9 secondary neoplasias plus active HL and 1 congestive heart failure directly after treatment. We also recorded 27 unrelated deaths and 30 deaths of secondary neoplasia during first complete remission.
The 10-year HLSS rate for the whole series was 91%. When sβ2m levels were classified as quartiles, a gradual drop was observed for each one from Q1 to Q4, with 10-year rates of 98%, 90%, 89% and 84% (p < 0.001; Figure 2D).
Patients with elevated sβ2m levels had inferior HLSS (93% versus 86%, p = 0.002; Figure 2E). At the cutoff of 2.0 mg/L, the difference was even more marked (96% versus 86%, p < 0.001; Figure 2F). Sβ2m levels > 2.0 mg/L were an independent adverse prognostic factor in the large-scale multivariate analysis (see Table 4 footnote) of all 915 patients, along with B-symptoms, lymphopenia and a lack of leukocytosis (hazard ratio (HR) 2.21, 95% CI 1.19–4.11, and p = 0.012; Table 4). The “normal versus elevated” comparison did not lead to a statistically significant result in the multivariate analysis (Table 4).
In the multivariate analysis of HLSS in early-stage patients, sβ2m levels were the only variable with a borderline-independent effect, only when evaluated at the cutoff of 2.0 mg/L (hazard ratio (HR) 2.30, 95% CI 0.89–5.94, and p = 0.085; Table 4), but not in a “normal versus elevated” basis (Table 4). In contrast, in the multivariate analysis of HLSS in advanced stages, sβ2m levels were neither an independent prognostic factor at the cutoff of 2.0 mg/L nor at a “normal versus elevated” basis (Table 4).
The 10-year SAF rate for the whole series was 61%. A statistically significant impact on SAF was observed when levels were classified as quartiles, with 10-year rates of 74%, 46%, 55% and 39% for Q1 through Q4 (p = 0.001; Figure 2G). Patients with elevated sβ2m levels had a similar SAF to those with normal levels (49% versus 55%, respectively, p = 0.177; Figure 2H). At the cutoff of 2.0 mg/L, the difference became borderline (62% versus 47%, p = 0.071; Figure 2I).

4. Discussion

B2m is synthesized in all nucleated cells, binds to major histocompatibility complex class I molecules, and is not directly attached to the cell membrane. Thus, free soluble β2m is detected in body fluids due to its release from the cell surface and cytoplasm. Since its identification 50 years ago, in 1972 [63], β2m has been widely investigated as a prognostic factor in hematologic malignancies. Further to the correlation with tumor burden [52], the mechanisms underlying the prognostic significance of β2mstill remain unclear. Indeed, several studies have shown that the prognostic significance of sβ2mmay be independent from factors reflecting disease burden [33,35], suggesting that it could either be related to other specific biologic features of lymphomas or simply overcome other markers of tumor burden, obscuring their significance.
The role of sβ2m as a prognostic factor in HL has been evaluated in several small- or medium-sized studies in the past, with partially conflicting results, probably owing to the sample sizes and the variable treatment approaches (Table 5) ([48,49,50,51,53,55,56,64,65,66,67,68,69], present study). Briefly, considering both the MD Anderson studies together [48,51], sβ2m appears to correlate with FFS in advanced stages (overall survival not reported), while it was associated with inferior overall survival in early stages, with only a borderline effect on FFS. It should be noted that treatment was inferior to ABVD and equivalents in the majority of these patients. In a study of the International Hodgkin Study Group, sβ2m was an independent predictor of FFS in early stage patients with favorable characteristics treated with RT alone [66].
In the present study, we evaluated the prognostic role of sβ2m in HL in the—by far—largest series published to date, consisting of a large cohort of homogeneously treated patients. Importantly, all patients had been treated with ABVD or equivalent regimens with or without RT, which are considered standard therapy for HL. Since it is known that more effective treatment may eliminate the significance of previously established prognostic factors, our study rules out a potential bias due to inferior treatment. Our results extend our previous observations and establish sβ2m as a potential independent prognostic factor in HL.
An important novel observation was made possible thanks to the very large size of this patient population: analyzing several potential cutoffs, we concluded that sβ2m may not work well as a prognostic factor, neither when analyzed on a “normal versus elevated” basis at the cutoff of 2.4 mg/L(as performed in our previous studies) [52,53] nor when analyzed at higher—clearly abnormal—cutoffs, as 2.5 mg/L [48,51,55,56] or 3.0 mg/L [65]. Instead, sβ2m worked better when the cutoff was set within the normal range at 2.0 mg/L. It is reasonable to wonder whether this observation is biologically relevant. In our opinion it is reasonable, because sβ2m levels are strongly and positively correlated with age in normal subjects. As the normal range is established from unselected normal individuals from the general population, the true upper normal limit for younger people might probably be lower. Along these lines, as patients with HL are much younger than the general population, the expected upper normal limit of sβ2m for the majority of them might be lower than the conventional 2.4 mg/L and might approach 2.0 mg/L.
The use of a sβ2m cutoff within the normal range of is supported by a recent Chinese study, in which 353 patients were evaluated, among whom 230 had received ABVD and 123 ABVD-like regimens, the latter with inferior progression-free survival (PFS). The levels of sβ2m were evaluated by ROC curves and the best cutoff was set at 1.85 mg/L, very similarly to our results. Although sβ2m levels above that cutoff were associated with inferior PFS and OS in the univariate analysis, the prognostic significance was independent of other factors only for OS. This is not unexpected, as sβ2m levels are more potent predictors of OS, as shown in the present and our previous studies, because of their strong association with age and renal function. However, the moderate size of the study by Wen et al. might have obscured an independent effect of sβ2m levels on PFS. In another small study of 67 patients, ROC analysis suggested a cutoff of 2.5 mg/L, which produced significant results in multivariate analysis for PFS, OS and DSS [55].
Reporting here our experience in 915 patients with HL, with 208 treatment failure events recorded so far (84 in early and 124 in advanced stages), this study was powered to detect moderate but clinically significant differences and to perform subgroup analyses according to clinical stage. Serum β2mlevels >2.0 mg/L independently predicted a lower FFP rate in the whole-patient population of this study, when evaluated in multivariate analysis including 11 additional and potentially strong prognostic covariates. The same was true for OS and HLSS. Notably, on a “normal versus elevated” basis, sβ2m had no independent effect on FFP and HLSS, presenting only a borderline association with OS. Among 515 patients with early-stage HL (IA/IIA), the best cutoff was found at 1.9 mg/L, but 2.0 mg/L was also highly significant and was used for further evaluation for reasons of consistency. Again, sβ2m was an independent predictor of FFP, when evaluated in multivariate analysis including eight additional covariates with established or strongly suspected prognostic significance in early-stage disease. Serum β2m levels were the only independent predictor of HLSS, albeit of marginal significance, but had no effect on OS. Among 400 patients with advanced-stage HL (IB/IIB/III/IV), sβ2m levels >2.0 mg/L were an independent predictor of FFP, when evaluated in multivariate analysis including all seven IPS covariates. This effect was more marked regarding OS but sβ2m had no independent effect on HLSS. On a “normal versus elevated” basis, sβ2m had no independent effect in any of the three endpoints.
Finally, this is the first study to show that sβ2m levels are significantly correlated with some established biological prognostic factors, such as serum sCD30 and serum IL-10 levels [66], but not with others, such as the tissue expression of bcl-2 and activated caspase-3 [30].

5. Conclusions

The present study has established the prognostic impact of sβ2m at a lower-than-expected cutoff, but also raises several new questions. It is not clear if sβ2mcan add to the prediction achieved by the IPS or other prognostics systems or just replace variables within the existing systems. Unfortunately, sβ2m was not evaluated during the development of the new holistic IPS [28], while its additive impact when biological prognostic factors are taken into account remains unknown. Some kind of “correction” according to the renal function should also be investigated in the effort to increase the prognostic significance of sβ2m levels. Another problem is that interim PET-guided therapy has predominated the field of treatment of Hodgkin lymphoma during the last decade. It is not clear whether sβ2m simply predicts a higher probability of interim PET positivity or may further help to discriminate which patients with a negative interim PET will relapse or who will be cured with intensified treatment following a positive interim PET. Similar considerations apply regarding the potential association of sβ2m levels with the results of end-of-treatment PET [70]. In addition, as novel prognostic factors appear, their correlation with sβ2m should be accurately determined. Unfortunately, there are no data regarding the correlation of sβ2m, neither with the circulating tumor DNA [71,72,73] and its changes during treatment nor with PET metrics, including baseline total metabolic tumor volume (TMTV) [74,75,76,77,78], total lesion glucolysis (TLG) [79,80] or lesion dissemination [81,82,83]. As sβ2m levels correlated strongly with almost all baseline features reflecting disease extent and aggressiveness in this study, it is reasonable to hypothesize a strong correlation with PET metrics, which, however, does not exclude the persistence of the independent prognostic significance of sβ2m.Finally, probably the main question to be asked in the near future is how sβ2m will affect the outcome of patients treated in the first line with chemotherapy plus novel agents such as BV-AVD or BreCADD, or—more importantly—how sβ2m will work as a prognostic factor under treatment with nivolumab-AVD [8].

Author Contributions

Conceptualization, T.P.V. and G.A.P.; methodology, T.P.V., M.A. and F.P.; software, T.P.V. and F.P.; investigation, T.P.V., M.A., P.T.D., A.L., F.P., M.P.S., M.D., S.I.K., S.S., M.B., C.C., E.A.K., J.V.A., K.P., G.B., A.K. (Alexandros Kanellopoulo), A.P., M.-E.L., A.G., A.K. (Anastasia Kopsaftopoulou), K.Z., I.D., M.N.D., M.-C.K., P.T., E.P., E.V., G.T., F.N.K., K.K., G.A.P., P.P. and M.K.A.; data curation, T.P.V., M.A., A.L. and M.K.A.; writing—original draft preparation, T.P.V., M.A. and P.T.D.; writing—review and editing, T.P.V., M.A., P.T.D., A.L., F.P., M.P.S., M.D., S.I.K., S.S., M.B., C.C., E.A.K., J.V.A., K.P., G.B., A.K. (Alexandros Kanellopoulo), A.P., M.-E.L., A.G., A.K. (Anastasia Kopsaftopoulou), K.Z., I.D., M.N.D., M.-C.K., P.T., E.P., E.V., G.T., F.N.K., K.K., G.A.P., P.P. and M.K.A.; supervision, T.P.V., K.K., G.A.P.,M.K.A. and P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of LAIKON General Hospital: 1000/07-12-2006.

Informed Consent Statement

As a retrospective, non interventional study, informed consent was waived.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Canellos, G.P.; Rosenberg, S.A.; Friedberg, J.W.; Lister, T.A.; Devita, V.T. Treatment of Hodgkin lymphoma: A 50-year perspective. J. Clin. Oncol. 2014, 32, 163–168. [Google Scholar] [CrossRef] [PubMed]
  2. Vassilakopoulos, T.P.; Angelopoulou, M.K. Advanced and relapsed/refractory Hodgkin lymphoma: What has been achieved during the last 50 years. Semin. Hematol. 2013, 50, 4–14. [Google Scholar] [CrossRef] [PubMed]
  3. Armand, P.; Zinzani, P.L.; Lee, H.J.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; Tomita, A.; et al. Five-year follow-up of KEYNOTE-087: Pembrolizumab monotherapy in relapsed/refractory classical Hodgkin lymphoma. Blood 2023, 142, 878–886. [Google Scholar] [CrossRef] [PubMed]
  4. Armand, P.; Engert, A.; Younes, A.; Fanale, M.; Santoro, A.; Zinzani, P.L.; Timmerman, J.M.; Collins, G.P.; Ramchandren, R.; Cohen, J.B.; et al. Nivolumab for Relapsed/Refractory Classic Hodgkin Lymphoma After Failure of Autologous Hematopoietic Cell Transplantation: Extended Follow-Up of the Multicohort Single-Arm Phase II CheckMate 205 Trial. J. Clin. Oncol. 2018, 36, 1428–1439. [Google Scholar] [CrossRef] [PubMed]
  5. Vassilakopoulos, T.P.; Chatzidimitriou, C.; Asimakopoulos, J.V.; Arapaki, M.; Tzoras, E.; Angelopoulou, M.K.; Konstantopoulos, K. Immunotherapy in Hodgkin Lymphoma: Present Status and Future Strategies. Cancers 2019, 11, 1071. [Google Scholar] [CrossRef] [PubMed]
  6. Vassilakopoulos, T.P.; Asimakopoulos, J.V.; Konstantopoulos, K.; Angelopoulou, M.K. Optimizing outcomes in relapsed/refractory Hodgkin lymphoma: A review of current and forthcoming therapeutic strategies. Ther. Adv. Hematol. 2020, 11, 2040620720902911. [Google Scholar] [CrossRef] [PubMed]
  7. Kuruvilla, J.; Ramchandren, R.; Santoro, A.; Paszkiewicz-Kozik, E.; Gasiorowski, R.; Johnson, N.A.; Fogliatto, L.M.; Goncalves, I.; de Oliveira, J.S.R.; Buccheri, V.; et al. Pembrolizumab versus brentuximab vedotin in relapsed or refractory classical Hodgkin lymphoma (KEYNOTE-204): An interim analysis of a multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2021, 22, 512–524. [Google Scholar] [CrossRef] [PubMed]
  8. Vassilakopoulos, T.P.; Liaskas, A.; Pereyra, P.; Panayiotidis, P.; Angelopoulou, M.K.; Gallamini, A. Incorporating Monoclonal Antibodies into the First-Line Treatment of Classical Hodgkin Lymphoma. Int. J. Mol. Sci. 2023, 24, 13187. [Google Scholar] [CrossRef]
  9. Carbone, P.P.; Kaplan, H.S.; Musshoff, K.; Smithers, D.W.; Tubiana, M. Report of the Committee on Hodgkin’s Disease Staging Classification. Cancer Res. 1971, 31, 1860–1861. [Google Scholar]
  10. Lister, T.A.; Crowther, D.; Sutcliffe, S.B.; Glatstein, E.; Canellos, G.P.; Young, R.C.; Rosenberg, S.A.; Coltman, C.A.; Tubiana, M. Report of a committee convened to discuss the evaluation and staging of patients with Hodgkin’s disease: Cotswolds meeting. J. Clin. Oncol. 1989, 7, 1630–1636. [Google Scholar] [CrossRef]
  11. Cheson, B.D.; Fisher, R.I.; Barrington, S.F.; Cavalli, F.; Schwartz, L.H.; Zucca, E.; Lister, T.A. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: The Lugano classification. J. Clin. Oncol. 2014, 32, 3059–3068. [Google Scholar] [CrossRef] [PubMed]
  12. Carde, P.; Hagenbeek, A.; Hayat, M.; Monconduit, M.; Thomas, J.; Burgers, M.J.; Noordijk, E.M.; Tanguy, A.; Meerwaldt, J.H.; Le Fur, R.; et al. Clinical staging versus laparotomy and combined modality with MOPP versus ABVD in early-stage Hodgkin’s disease: The H6 twin randomized trials from the European Organization for Research and Treatment of Cancer Lymphoma Cooperative Group. J. Clin. Oncol. 1993, 11, 2258–2272. [Google Scholar] [CrossRef] [PubMed]
  13. Noordijk, E.M.; Carde, P.; Dupouy, N.; Hagenbeek, A.; Krol, A.D.; Kluin-Nelemans, J.C.; Tirelli, U.; Monconduit, M.; Thomas, J.; Eghbali, H.; et al. Combined-modality therapy for clinical stage I or II Hodgkin’s lymphoma: Long-term results of the European Organisation for Research and Treatment of Cancer H7 randomized controlled trials. J. Clin. Oncol. 2006, 24, 3128–3135. [Google Scholar] [CrossRef]
  14. Fermé, C.; Eghbali, H.; Meerwaldt, J.H.; Rieux, C.; Bosq, J.; Berger, F.; Girinsky, T.; Brice, P.; van’t Veer, M.B.; Walewski, J.A.; et al. Chemotherapy plus involved-field radiation in early-stage Hodgkin’s disease. N. Engl. J. Med. 2007, 357, 1916–1927. [Google Scholar] [CrossRef] [PubMed]
  15. Raemaekers, J.M.; André, M.P.; Federico, M.; Girinsky, T.; Oumedaly, R.; Brusamolino, E.; Brice, P.; Fermé, C.; van der Maazen, R.; Gotti, M.; et al. Omitting radiotherapy in early positron emission tomography-negative stage I/II Hodgkin lymphoma is associated with an increased risk of early relapse: Clinical results of the preplanned interim analysis of the randomized EORTC/LYSA/FIL H10 trial. J. Clin. Oncol. 2014, 32, 1188–1194. [Google Scholar] [CrossRef]
  16. Fermé, C.; Thomas, J.; Brice, P.; Casasnovas, O.; Vranovsky, A.; Bologna, S.; Lugtenburg, P.J.; Bouabdallah, R.; Carde, P.; Sebban, C.; et al. ABVD or BEACOPP(baseline) along with involved-field radiotherapy in early-stage Hodgkin Lymphoma with risk factors: Results of the European Organisation for Research and Treatment of Cancer (EORTC)-Groupe d’Étude des Lymphomes de l’Adulte (GELA) H9-U intergroup randomised trial. Eur. J. Cancer 2017, 81, 45–55. [Google Scholar] [CrossRef]
  17. Engert, A.; Franklin, J.; Eich, H.T.; Brillant, C.; Sehlen, S.; Cartoni, C.; Herrmann, R.; Pfreundschuh, M.; Sieber, M.; Tesch, H.; et al. Two cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine plus extended-field radiotherapy is superior to radiotherapy alone in early favorable Hodgkin’s lymphoma: Final results of the GHSG HD7 trial. J. Clin. Oncol. 2007, 25, 3495–3502. [Google Scholar] [CrossRef]
  18. Engert, A.; Schiller, P.; Josting, A.; Herrmann, R.; Koch, P.; Sieber, M.; Boissevain, F.; De Wit, M.; Mezger, J.; Duhmke, E.; et al. Involved-field radiotherapy is equally effective and less toxic compared with extended-field radiotherapy after four cycles of chemotherapy in patients with early-stage unfavorable Hodgkin’s lymphoma: Results of the HD8 trial of the German Hodgkin’s Lymphoma Study Group. J. Clin. Oncol. 2003, 21, 3601–3608. [Google Scholar] [CrossRef]
  19. Engert, A.; Plütschow, A.; Eich, H.T.; Lohri, A.; Dörken, B.; Borchmann, P.; Berger, B.; Greil, R.; Willborn, K.C.; Wilhelm, M.; et al. Reduced treatment intensity in patients with early-stage Hodgkin’s lymphoma. N. Engl. J. Med. 2010, 363, 640–652. [Google Scholar] [CrossRef]
  20. Eich, H.T.; Diehl, V.; Görgen, H.; Pabst, T.; Markova, J.; Debus, J.; Ho, A.; Dörken, B.; Rank, A.; Grosu, A.L.; et al. Intensified chemotherapy and dose-reduced involved-field radiotherapy in patients with early unfavorable Hodgkin’s lymphoma: Final analysis of the German Hodgkin Study Group HD11 trial. J. Clin. Oncol. 2010, 28, 4199–4206. [Google Scholar] [CrossRef]
  21. Behringer, K.; Goergen, H.; Hitz, F.; Zijlstra, J.M.; Greil, R.; Markova, J.; Sasse, S.; Fuchs, M.; Topp, M.S.; Soekler, M.; et al. Omission of dacarbazine or bleomycin, or both, from the ABVD regimen in treatment of early-stage favourable Hodgkin’s lymphoma (GHSG HD13): An open-label, randomised, non-inferiority trial. Lancet 2015, 385, 1418–1427. [Google Scholar] [CrossRef] [PubMed]
  22. von Tresckow, B.; Plütschow, A.; Fuchs, M.; Klimm, B.; Markova, J.; Lohri, A.; Kral, Z.; Greil, R.; Topp, M.S.; Meissner, J.; et al. Dose-intensification in early unfavorable Hodgkin’s lymphoma: Final analysis of the German Hodgkin Study Group HD14 trial. J. Clin. Oncol. 2012, 30, 907–913. [Google Scholar] [CrossRef] [PubMed]
  23. Fuchs, M.; Goergen, H.; Kobe, C.; Kuhnert, G.; Lohri, A.; Greil, R.; Sasse, S.; Topp, M.S.; Schafer, E.; Hertenstein, B.; et al. Positron Emission Tomography-Guided Treatment in Early-Stage Favorable Hodgkin Lymphoma: Final Results of the International, Randomized Phase III HD16 Trial by the German Hodgkin Study Group. J. Clin. Oncol. 2019, 37, 2835–2845. [Google Scholar] [CrossRef]
  24. Borchmann, P.; Plutschow, A.; Kobe, C.; Greil, R.; Meissner, J.; Topp, M.S.; Ostermann, H.; Dierlamm, J.; Mohm, J.; Thiemer, J.; et al. PET-guided omission of radiotherapy in early-stage unfavourable Hodgkin lymphoma (GHSG HD17): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2021, 22, 223–234. [Google Scholar] [CrossRef] [PubMed]
  25. Hasenclever, D.; Diehl, V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N. Engl. J. Med. 1998, 339, 1506–1514. [Google Scholar] [CrossRef] [PubMed]
  26. Hayden, A.R.; Lee, D.G.; Villa, D.; Gerrie, A.S.; Scott, D.W.; Slack, G.W.; Sehn, L.H.; Connors, J.M.; Savage, K.J. Validation of a simplified international prognostic score (IPS-3) in patients with advanced-stage classic Hodgkin lymphoma. Br. J. Haematol. 2020, 189, 122–127. [Google Scholar] [CrossRef] [PubMed]
  27. Diefenbach, C.S.; Li, H.; Hong, F.; Gordon, L.I.; Fisher, R.I.; Bartlett, N.L.; Crump, M.; Gascoyne, R.D.; Wagner, H., Jr.; Stiff, P.J.; et al. Evaluation of the International Prognostic Score (IPS-7) and a Simpler Prognostic Score (IPS-3) for advanced Hodgkin lymphoma in the modern era. Br. J. Haematol. 2015, 171, 530–538. [Google Scholar] [CrossRef]
  28. Rodday, A.M.; Parsons, S.K.; Upshaw, J.N.; Friedberg, J.W.; Gallamini, A.; Hawkes, E.; Hodgson, D.; Johnson, P.; Link, B.K.; Mou, E.; et al. The Advanced-Stage Hodgkin Lymphoma International Prognostic Index: Development and Validation of a Clinical Prediction Model From the HoLISTIC Consortium. J. Clin. Oncol. 2023, 41, 2076–2086. [Google Scholar] [CrossRef]
  29. Moccia, A.A.; Donaldson, J.; Chhanabhai, M.; Hoskins, P.J.; Klasa, R.J.; Savage, K.J.; Shenkier, T.N.; Slack, G.W.; Skinnider, B.; Gascoyne, R.D.; et al. International Prognostic Score in advanced-stage Hodgkin’s lymphoma: Altered utility in the modern era. J. Clin. Oncol. 2012, 30, 3383–3388. [Google Scholar] [CrossRef]
  30. Brockelmann, P.J.; Angelopoulou, M.K.; Vassilakopoulos, T.P. Prognostic factors in Hodgkin lymphoma. Semin. Hematol. 2016, 53, 155–164. [Google Scholar] [CrossRef] [PubMed]
  31. Greipp, P.R.; San Miguel, J.; Durie, B.G.; Crowley, J.J.; Barlogie, B.; Bladé, J.; Boccadoro, M.; Child, J.A.; Avet-Loiseau, H.; Kyle, R.A.; et al. International staging system for multiple myeloma. J. Clin. Oncol. 2005, 23, 3412–3420. [Google Scholar] [CrossRef] [PubMed]
  32. Federico, M.; Guglielmi, C.; Luminari, S.; Mammi, C.; Marcheselli, L.; Gianelli, U.; Maiorana, A.; Merli, F.; Bellei, M.; Pozzi, S.; et al. Prognostic relevance of serum beta2 microglobulin in patients with follicular lymphoma treated with anthracycline-containing regimens. A GISL study. Haematologica 2007, 92, 1482–1488. [Google Scholar] [CrossRef] [PubMed]
  33. Federico, M.; Bellei, M.; Marcheselli, L.; Luminari, S.; Lopez-Guillermo, A.; Vitolo, U.; Pro, B.; Pileri, S.; Pulsoni, A.; Soubeyran, P.; et al. Follicular lymphoma international prognostic index 2: A new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. J. Clin. Oncol. 2009, 27, 4555–4562. [Google Scholar] [CrossRef] [PubMed]
  34. Shi, C.; Zhu, Y.; Su, Y.; Chung, L.W.; Cheng, T. Beta2-microglobulin: Emerging as a promising cancer therapeutic target. Drug Discov. Today 2009, 14, 25–30. [Google Scholar] [CrossRef] [PubMed]
  35. Yoo, C.; Yoon, D.H.; Suh, C. Serum beta-2 microglobulin in malignant lymphomas: An old but powerful prognostic factor. Blood Res. 2014, 49, 148–153. [Google Scholar] [CrossRef]
  36. Miyashita, K.; Tomita, N.; Taguri, M.; Suzuki, T.; Ishiyama, Y.; Ishii, Y.; Nakajima, Y.; Numata, A.; Hattori, Y.; Yamamoto, W.; et al. Beta-2 microglobulin is a strong prognostic factor in patients with DLBCL receiving R-CHOP therapy. Leuk. Res. 2015, 39, 1187–1191. [Google Scholar] [CrossRef] [PubMed]
  37. Melchardt, T.; Troppan, K.; Weiss, L.; Hufnagl, C.; Neureiter, D.; Tränkenschuh, W.; Hopfinger, G.; Magnes, T.; Deutsch, A.; Neumeister, P.; et al. A modified scoring of the NCCN-IPI is more accurate in the elderly and is improved by albumin and β2 -microglobulin. Br. J. Haematol. 2015, 168, 239–245. [Google Scholar] [CrossRef] [PubMed]
  38. Seo, S.; Hong, J.Y.; Yoon, S.; Yoo, C.; Park, J.H.; Lee, J.B.; Park, C.S.; Huh, J.; Lee, Y.; Kim, K.W.; et al. Prognostic significance of serum beta-2 microglobulin in patients with diffuse large B-cell lymphoma in the rituximab era. Oncotarget 2016, 7, 76934–76943. [Google Scholar] [CrossRef]
  39. Yoo, C.; Yoon, D.H.; Kim, S.; Huh, J.; Park, C.S.; Park, C.J.; Lee, S.W.; Suh, C. Serum beta-2 microglobulin as a prognostic biomarker in patients with mantle cell lymphoma. Hematol. Oncol. 2016, 34, 22–27. [Google Scholar] [CrossRef]
  40. Kanemasa, Y.; Shimoyama, T.; Sasaki, Y.; Tamura, M.; Sawada, T.; Omuro, Y.; Hishima, T.; Maeda, Y. Beta-2 microglobulin as a significant prognostic factor and a new risk model for patients with diffuse large B-cell lymphoma. Hematol. Oncol. 2017, 35, 440–446. [Google Scholar] [CrossRef]
  41. Montalbán, C.; Díaz-López, A.; Dlouhy, I.; Rovira, J.; Lopez-Guillermo, A.; Alonso, S.; Martín, A.; Sancho, J.M.; García, O.; Sánchez, J.M.; et al. Validation of the NCCN-IPI for diffuse large B-cell lymphoma (DLBCL): The addition of β(2) -microglobulin yields a more accurate GELTAMO-IPI. Br. J. Haematol. 2017, 176, 918–928. [Google Scholar] [CrossRef]
  42. Sorigue, M.; Bishton, M.; Domingo-Domenech, E.; McMillan, A.; Prusila, R.; García, O.; Kuusisto, M.; Condom, M.; Tapia, G.; Ribera, J.M.; et al. Refractoriness to rituximab-based therapy and elevated serum B2-microglobulin predict for inferior survival in marginal zone lymphoma. Leuk. Lymphoma 2019, 60, 2524–2531. [Google Scholar] [CrossRef]
  43. Khashab, T.; Hagemeister, F.; Romaguera, J.E.; Fanale, M.A.; Pro, B.; McLaughlin, P.; Rodriguez, M.A.; Neelapu, S.S.; Fayad, L.; Younes, A.; et al. Long-term overall- and progression-free survival after pentostatin, cyclophosphamide and rituximab therapy for indolent non-Hodgkin lymphoma. Br. J. Haematol. 2019, 185, 670–678. [Google Scholar] [CrossRef] [PubMed]
  44. Bento, L.; Díaz-López, A.; Barranco, G.; Martín-Moreno, A.M.; Baile, M.; Martín, A.; Sancho, J.M.; García, O.; Rodríguez, M.; Sánchez-Pina, J.M.; et al. New prognosis score including absolute lymphocyte/monocyte ratio, red blood cell distribution width and beta-2 microglobulin in patients with diffuse large B-cell lymphoma treated with R-CHOP: Spanish Lymphoma Group Experience (GELTAMO). Br. J. Haematol. 2020, 188, 888–897. [Google Scholar] [CrossRef] [PubMed]
  45. Advani, R.H.; Skrypets, T.; Civallero, M.; Spinner, M.A.; Manni, M.; Kim, W.S.; Shustov, A.R.; Horwitz, S.M.; Hitz, F.; Cabrera, M.E.; et al. Outcomes and prognostic factors in angioimmunoblastic T-cell lymphoma: Final report from the international T-cell Project. Blood 2021, 138, 213–220. [Google Scholar] [CrossRef] [PubMed]
  46. Kang, S.; Cho, H.; Kim, S.; Lee, K.; Kang, E.H.; Park, J.S.; Lee, Y.S.; Park, C.S.; Go, H.; Huh, J.; et al. A New Prognostic Index for Extranodal Natural Killer/T-Cell Lymphoma: Incorporation of Serum β-2 Microglobulin to PINK. Cancer Res. Treat. 2023, 55, 314–324. [Google Scholar] [CrossRef] [PubMed]
  47. Tsimberidou, A.M.; Kantarjian, H.M.; Wen, S.; O’Brien, S.; Cortes, J.; Wierda, W.G.; Koller, C.; Pierce, S.; Brandt, M.; Freireich, E.J.; et al. The prognostic significance of serum beta2 microglobulin levels in acute myeloid leukemia and prognostic scores predicting survival: Analysis of 1,180 patients. Clin. Cancer Res. 2008, 14, 721–730. [Google Scholar] [CrossRef]
  48. Dimopoulos, M.A.; Cabanillas, F.; Lee, J.J.; Swan, F.; Fuller, L.; Allen, P.K.; Hagemeister, F.B. Prognostic role of serum beta 2-microglobulin in Hodgkin’s disease. J. Clin. Oncol. 1993, 11, 1108–1111. [Google Scholar] [CrossRef]
  49. Fleury, J.; Tortochaux, J.; Legros, M.; Cure, H.; Kwiatkowski, F.; Ferrière, J.P.; Travade, P.; Dionet, C.; Gaillard, G.; Chassagne, J.; et al. Prognostic value of beta-2-microglobulin in Hodgkin disease in young adults. Bull. Cancer 1994, 81, 625–631. [Google Scholar]
  50. Raida, L.; Papajík, T.; Hlusí, A.; Faber, E.; Urbanová, R.; Heczko, M.; Jancíková, M.; Zapletalová, J.; Komenda, S.; Indrák, K. Importance of determination of serum beta-2-microglobulin levels in patients with Hodgkin’s lymphoma. Vnitr. Lek. 2002, 48, 91–95. [Google Scholar]
  51. Chronowski, G.M.; Wilder, R.B.; Tucker, S.L.; Ha, C.S.; Sarris, A.H.; Hagemeister, F.B.; Barista, I.; Hess, M.A.; Cabanillas, F.; Cox, J.D. An elevated serum beta-2-microglobulin level is an adverse prognostic factor for overall survival in patients with early-stage Hodgkin disease. Cancer 2002, 95, 2534–2538. [Google Scholar] [CrossRef] [PubMed]
  52. Vassilakopoulos, T.P.; Nadali, G.; Angelopoulou, M.K.; Siakantaris, M.P.; Dimopoulou, M.N.; Kontopidou, F.N.; Karkantaris, C.; Kokoris, S.I.; Kyrtsonis, M.C.; Tsaftaridis, P.; et al. The prognostic significance of beta(2)-microglobulin in patients with Hodgkin’s lymphoma. Haematologica 2002, 87, 701–708. [Google Scholar] [PubMed]
  53. Vassilakopoulos, T.P.; Nadali, G.; Angelopoulou, M.K.; Dimopoulou, M.N.; Siakantaris, M.P.; Kontopidou, F.N.; Karkantaris, C.; Kokoris, S.I.; Dimitriadou, E.M.; Calpadaki, C.; et al. Beta(2)-microglobulin in Hodgkin’s lymphoma: Prognostic significance in patients treated with ABVD or equivalent regimens. J. Buon 2005, 10, 59–69. [Google Scholar] [PubMed]
  54. Bien, E.; Balcerska, A. Serum soluble interleukin-2 receptor, beta2-microglobulin, lactate dehydrogenase and erythrocyte sedimentation rate in children with Hodgkin’s lymphoma. Scand. J. Immunol. 2009, 70, 490–500. [Google Scholar] [CrossRef] [PubMed]
  55. Nakajima, Y.; Tomita, N.; Watanabe, R.; Ishiyama, Y.; Yamamoto, E.; Ishibashi, D.; Itabashi, M.; Koyama, S.; Takahashi, H.; Numata, A.; et al. Prognostic significance of serum beta-2 microglobulin level in Hodgkin lymphoma treated with ABVD-based therapy. Med. Oncol. 2014, 31, 185. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, Q.; Qin, Y.; Zhou, S.; He, X.; Yang, J.; Kang, S.; Liu, P.; Yang, S.; Zhang, C.; Gui, L.; et al. Prognostic value of pretreatment serum beta-2 microglobulin level in advanced classical Hodgkin lymphoma treated in the modern era. Oncotarget 2016, 7, 72219–72228. [Google Scholar] [CrossRef] [PubMed]
  57. Vassilakopoulos, T.P.; Angelopoulou, M.K.; Siakantaris, M.P.; Kontopidou, F.N.; Dimopoulou, M.N.; Kokoris, S.I.; Kyrtsonis, M.C.; Tsaftaridis, P.; Karkantaris, C.; Anargyrou, K.; et al. Combination chemotherapy plus low-dose involved-field radiotherapy for early clinical stage Hodgkin’s lymphoma. Int. J. Radiat. Oncol. Biol. Phys. 2004, 59, 765–781. [Google Scholar] [CrossRef]
  58. Vassilakopoulos, T.P.; Dimopoulou, M.N.; Angelopoulou, M.K.; Petevi, K.; Pangalis, G.A.; Moschogiannis, M.; Dimou, M.; Boutsikas, G.; Kanellopoulos, A.; Gainaru, G.; et al. Prognostic Implication of the Absolute Lymphocyte to Absolute Monocyte Count Ratio in Patients With Classical Hodgkin Lymphoma Treated With Doxorubicin, Bleomycin, Vinblastine, and Dacarbazine or Equivalent Regimens. Oncologist 2016, 21, 343–353. [Google Scholar] [CrossRef]
  59. Karakatsanis, S.; Panitsas, F.; Arapaki, M.; Galopoulos, D.; Asimakopoulos, J.V.; Liaskas, A.; Chatzidimitriou, C.; Belia, M.; Konstantinou, E.; Vassilopoulos, I.; et al. Serum ferritin levels in previously untreated classical Hodgkin lymphoma: Correlations and prognostic significance. Leuk. Lymphoma 2022, 63, 799–812. [Google Scholar] [CrossRef]
  60. Kaplan, E.; Meier, P. Nonparametric estimation from incomplete observations. J. Am. Stat. Assoc. 1958, 53, 457–481. [Google Scholar] [CrossRef]
  61. Mantel, N. Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother. Rep. 1966, 50, 163–170. [Google Scholar] [PubMed]
  62. Cox, D. Regression models and life tables (with Discussion). J. R. Stat. Soc. B 1972, 34. [Google Scholar]
  63. Peterson, P.A.; Cunningham, B.A.; Berggård, I.; Edelman, G.M. β2-Microglobulin--A free immunoglobulin domain. Proc. Natl. Acad. Sci. USA 1972, 69, 1697–1701. [Google Scholar] [CrossRef] [PubMed]
  64. Axdorph, U.; Sjöberg, J.; Grimfors, G.; Landgren, O.; Porwit-MacDonald, A.; Björkholm, M. Biological markers may add to prediction of outcome achieved by the International Prognostic Score in Hodgkin’s disease. Ann. Oncol. 2000, 11, 1405–1411. [Google Scholar] [CrossRef] [PubMed]
  65. Oza, A.M.; Ganesan, T.S.; Dorreen, M.; Johnson, P.W.; Waxman, J.; Gregory, W.; Lim, J.; Wright, J.; Dadiotis, L.; Barbounis, V.; et al. Patterns of survival in patients with advanced Hodgkin’s disease (HD) treated in a single centre over 20 years. Br. J. Cancer 1992, 65, 429–437. [Google Scholar] [CrossRef] [PubMed]
  66. Visco, C.; Vassilakopoulos, T.P.; Kliche, K.O.; Nadali, G.; Viviani, S.; Bonfante, V.; Medeiros, L.J.; Notti, P.; Rassidakis, G.Z.; Peethambaram, P.; et al. Elevated serum levels of IL-10 are associated with inferior progression-free survival in patients with Hodgkin’s disease treated with radiotherapy. Leuk. Lymphoma 2004, 45, 2085–2092. [Google Scholar] [CrossRef] [PubMed]
  67. Itoh, K.; Kinoshita, T.; Watanabe, T.; Yoshimura, K.; Okamoto, R.; Chou, T.; Ogura, M.; Hirano, M.; Asaoku, H.; Kurosawa, M.; et al. Prognostic analysis and a new risk model for Hodgkin lymphoma in Japan. Int. J. Hematol. 2010, 91, 446–455. [Google Scholar] [CrossRef] [PubMed]
  68. Mirili, C.; Paydas, S.; Kapukaya, T.K.; Yılmaz, A. Systemic immune-inflammation index predicting survival outcome in patients with classical Hodgkin lymphoma. Biomark. Med. 2019, 13, 1565–1575. [Google Scholar] [CrossRef]
  69. Wen, Q.; Ge, J.; Lei, Y.; Zhang, Y.; Kong, X.; Wang, W.; Hou, H.; Wang, Z.; Qian, S.; Ding, M.; et al. Real-world evidence of ABVD-like regimens compared with ABVD in classical Hodgkin lymphoma: A 10-year study from China. J. Cancer Res. Clin. Oncol. 2022, 149, 3989–4003. [Google Scholar] [CrossRef]
  70. Picardi, M.; Fonti, R.; Della Pepa, R.; Giordano, C.; Pugliese, N.; Nicolai, E.; Salvatore, M.; Mainolfi, C.; Venetucci, P.; Rascato, M.G.; et al. 2-deoxy-2[F-18] fluoro-D-glucose positron emission tomography Deauville scale and core-needle biopsy to determine successful management after six doxorubicin, bleomycin, vinblastine and dacarbazine cycles in advanced-stage Hodgkin lymphoma. Eur. J. Cancer 2020, 132, 85–97. [Google Scholar] [CrossRef]
  71. Spina, V.; Bruscaggin, A.; Cuccaro, A.; Martini, M.; Di Trani, M.; Forestieri, G.; Manzoni, M.; Condoluci, A.; Arribas, A.; Terzi-Di-Bergamo, L.; et al. Circulating tumor DNA reveals genetics, clonal evolution, and residual disease in classical Hodgkin lymphoma. Blood 2018, 131, 2413–2425. [Google Scholar] [CrossRef]
  72. Camus, V.; Viennot, M.; Lequesne, J.; Viailly, P.J.; Bohers, E.; Bessi, L.; Marcq, B.; Etancelin, P.; Dubois, S.; Picquenot, J.M.; et al. Targeted genotyping of circulating tumor DNA for classical Hodgkin lymphoma monitoring: A prospective study. Haematologica 2021, 106, 154–162. [Google Scholar] [CrossRef]
  73. Pepe, F.; Giordano, C.; Russo, G.; Palumbo, L.; Vincenzi, A.; Acanfora, G.; Lisi, D.; Picardi, M.; Pane, F.; Troncone, G.; et al. Liquid biopsy: A promising tool for driving strategies and predicting failures in patients with classic Hodgkin lymphoma. Cytopathology 2023. [Google Scholar] [CrossRef]
  74. Mettler, J.; Müller, H.; Voltin, C.A.; Baues, C.; Klaeser, B.; Moccia, A.; Borchmann, P.; Engert, A.; Kuhnert, G.; Drzezga, A.E.; et al. Metabolic Tumour Volume for Response Prediction in Advanced-Stage Hodgkin Lymphoma. J. Nucl. Med. 2018, 60, 207–211. [Google Scholar] [CrossRef]
  75. Cottereau, A.S.; Versari, A.; Loft, A.; Casasnovas, O.; Bellei, M.; Ricci, R.; Bardet, S.; Castagnoli, A.; Brice, P.; Raemaekers, J.; et al. Prognostic value of baseline metabolic tumor volume in early-stage Hodgkin lymphoma in the standard arm of the H10 trial. Blood 2018, 131, 1456–1463. [Google Scholar] [CrossRef]
  76. Gallamini, A.; Sudria, A.; Kurlapski, M.; Gastaud, L. Revisiting the predictive role of 18F-fluorodeoxyglucose positron emission tomography/computerized tomography on treatment outcome in early-stage favorable Hodgkin lymphoma. Hematol. Oncol. 2023, 41, 608–611. [Google Scholar] [CrossRef]
  77. van Heek, L.; Stuka, C.; Kaul, H.; Müller, H.; Mettler, J.; Hitz, F.; Baues, C.; Fuchs, M.; Borchmann, P.; Engert, A.; et al. Predictive value of baseline metabolic tumor volume in early-stage favorable Hodgkin Lymphoma - Data from the prospective, multicenter phase III HD16 trial. BMC Cancer 2022, 22, 672. [Google Scholar] [CrossRef]
  78. Rossi, C.; André, M.; Dupuis, J.; Morschhauser, F.; Joly, B.; Lazarovici, J.; Ghesquières, H.; Stamatoullas, A.; Nicolas-Virelizier, E.; Feugier, P.; et al. High-risk stage IIB Hodgkin lymphoma treated in the H10 and AHL2011 trials: Total metabolic tumor volume is a useful risk factor to stratify patients at baseline. Haematologica 2022, 107, 2897–2904. [Google Scholar] [CrossRef]
  79. Moskowitz, A.J.; Schöder, H.; Gavane, S.; Thoren, K.L.; Fleisher, M.; Yahalom, J.; McCall, S.J.; Cadzin, B.R.; Fox, S.Y.; Gerecitano, J.; et al. Prognostic significance of baseline metabolic tumor volume in relapsed and refractory Hodgkin lymphoma. Blood 2017, 130, 2196–2203. [Google Scholar] [CrossRef]
  80. Guo, B.; Tan, X.; Ke, Q.; Cen, H. Prognostic value of baseline metabolic tumor volume and total lesion glycolysis in patients with lymphoma: A meta-analysis. PLoS ONE 2019, 14, e0210224. [Google Scholar] [CrossRef]
  81. Driessen, J.; Zwezerijnen, G.J.C.; Schöder, H.; Drees, E.E.E.; Kersten, M.J.; Moskowitz, A.J.; Moskowitz, C.H.; Eertink, J.J.; Vet, H.C.W.; Hoekstra, O.S.; et al. The Impact of Semiautomatic Segmentation Methods on Metabolic Tumor Volume, Intensity, and Dissemination Radiomics in (18)F-FDG PET Scans of Patients with Classical Hodgkin Lymphoma. J. Nucl. Med. 2022, 63, 1424–1430. [Google Scholar] [CrossRef] [PubMed]
  82. Durmo, R.; Donati, B.; Rebaud, L.; Cottereau, A.S.; Ruffini, A.; Nizzoli, M.E.; Ciavarella, S.; Vegliante, M.C.; Nioche, C.; Meignan, M.; et al. Prognostic value of lesion dissemination in doxorubicin, bleomycin, vinblastine, and dacarbazine-treated, interimPET-negative classical Hodgkin Lymphoma patients: A radio-genomic study. Hematol. Oncol. 2022, 40, 645–657. [Google Scholar] [CrossRef] [PubMed]
  83. Gallamini, A.; Rambaldi, A.; Patti, C.; Romano, A.; Viviani, S.; Silvia, B.; Silvia, O.; Trentin, L.; Cantonetti, M.; Roberto, S.; et al. Lesion Dissemination in Baseline PET/CT (D-MAX) and IPS Score Predict ABVD Treatment Outcome in PET-2 Negative Advanced-Stage Hodgkin Lymphoma Patients Enrolled in the Prospective GITIL/FIL HD0607 Trial. Blood 2021, 138, 2443. [Google Scholar] [CrossRef]
Figure 1. (A) Freedom from progression (FFP) according to sβ2m levels classified as quartiles in the whole patient population; (B) FFP according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); (C) FFP according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in the whole patient population; (D) FFP according to sβ2m levels (≤1.9 mg/L vs. >1.9 mg/L) in early-stage patients; (E) FFP according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in early-stage patients; and (F) FFP according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L)in advanced-stage patients.
Figure 1. (A) Freedom from progression (FFP) according to sβ2m levels classified as quartiles in the whole patient population; (B) FFP according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); (C) FFP according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in the whole patient population; (D) FFP according to sβ2m levels (≤1.9 mg/L vs. >1.9 mg/L) in early-stage patients; (E) FFP according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in early-stage patients; and (F) FFP according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L)in advanced-stage patients.
Cancers 16 00238 g001
Figure 2. (A) Overall survival (OS) according to sβ2m levels classified as quartiles in the whole patient population; (B) OS according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); (C) OS according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in the whole patient population; (D) Hodgkin lymphoma-specific survival (HLSS) according to sβ2m levels classified as quartiles in the whole patient population; (E) HLSS according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); (F) HLSS according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in the whole patient population; (G) survival after failure (SAF) according to sβ2m levels classified as quartiles in the whole patient population; (H) SAF according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); and (I) SAF according to sβ2m levels (≤2.0 mg/L vs. 2.0 mg/L) in the whole patient population.
Figure 2. (A) Overall survival (OS) according to sβ2m levels classified as quartiles in the whole patient population; (B) OS according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); (C) OS according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in the whole patient population; (D) Hodgkin lymphoma-specific survival (HLSS) according to sβ2m levels classified as quartiles in the whole patient population; (E) HLSS according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); (F) HLSS according to sβ2m levels (≤2.0 mg/L vs. >2.0 mg/L) in the whole patient population; (G) survival after failure (SAF) according to sβ2m levels classified as quartiles in the whole patient population; (H) SAF according to sβ2m levels as “normal vs. elevated” in the whole patient population (≤2.4 mg/L vs. >2.4 mg/L); and (I) SAF according to sβ2m levels (≤2.0 mg/L vs. 2.0 mg/L) in the whole patient population.
Cancers 16 00238 g002
Table 1. Patient characteristics and correlations with serum β2-microglobulin levels.
Table 1. Patient characteristics and correlations with serum β2-microglobulin levels.
Patient CharacteristicsValuePatientsSerum β2-Microglobulinp-Value
#%MedianIQR
Age (years)<45
≥45
659
256
72.0
28.0
2.00
3.00
1.70–2.63
2.20–4.20
<0.001
Genderfemale
male
402
513
43.9
56.1
2.00
2.37
1.70–2.80
1.89–3.20
<0.001
AnnArbor StageI/IIA
IB/IIB/III/IV
515
400
56.3
33.7
1.98
2.70
1.70–2.55
2.00–3.70
<0.001
AnnArbor StageIA/B
IIA/B
IIIA/B
IVA/B
182/13
320/111
75/87
34/93
19.9/1.4
35.0/12.1
8.2/9.5
3.7/10.2
2.00/2.90
1.95/2.30
2.40/3.20
2.49/2.93
0.85/2.15
0.82/1.54
1.40/1.96
1.59/2.03
<0.001
B-SymptomsA
B
611
304
66.8
33.2
2.00
2.80
1.70–2.61
2.03–3.93
<0.001
HistologyNLP
NS
MC
LD
LR
UCL
IF-NS/MC
44
610
173
3
39
16
4+2
4.9
68.5
19.4
0.3
4.4
1.8
0.6
1.90
2.13
2.61
2.48
2.30
3.39
1.90
1.51–2.58
1.76–2.80
1.90–3.70
1.90–5.30
1.90–2.95
2.38–4.33
1.87–3.04
<0.001
Bone marrow involvementno
yes
819
41
95.2
4.8
2.17
3.50
1.78–3.00
2.68–5.25
<0.001
Liver involvementno
yes
887
21
97.7
2.3
2.20
4.00
1.80–3.00
2.74–4.76
<0.001
Lung involvementno
yes
819
84
90.7
9.3
2.18
2.44
1.78–3.00
2.00–3.48
0.012
Iliac/inguinal involvementno
yes
818
82
90.9
9.1
2.10
3.00
1.77–2.80
2.03–3.80
0.022
Anemiano
yes
525
389
57.4
42.6
2.00
2.52
1.70–2.63
1.95–3.70
<0.001
Leukocytosis (×109/L)<10
≥10
533
377
58.6
41.4
2.20
2.22
1.79–3.24
1.82–2.90
0.83
Marked Leukocytosis(×109/L)<15
≥15
787
123
86.5
13.5
2.18
2.40
1.78–3.00
1.97–3.14
0.019
Severe Lymphocytopeniano
yes
771
97
88.8
11.2
2.20
2.54
1.80–3.00
2.02–3.84
<0.001
ESR (mm/h)<50
≥50
408
411
49.8
50.2
2.00
2.41
1.70–2.60
1.90–3.38
<0.001
LDHnormal
elevated
599
251
70.5
29.5
2.10
2.61
1.75–2.80
2.00–3.62
<0.001
Albumin (g/dL)≥4
<4
469
413
53.2
46.8
2.00
2.58
1.70–2.60
1.94–3.68
<0.001
IPS0–2
3–7
633
244
72.2
27.8
2.00
3.05
1.70–2.60
2.26–4.08
<0.001
Nodal sites (#; AAS I/IIA)1–2
≥3
371
143
72.2
27.8
2.00
1.93
1.70–2.60
1.70–2.47
0.69
Involved sites (#; AAS IIB-IV)≤4
≥5
205
183
52.8
47.2
2.40
3.00
1.87–3.45
2.21–4.00
<0.001
IQR = interquartile range, NLP = nodular lymphocyte predominant, NS = nodular sclerosis, MC = mixed cellularity, LD = lymphocyte depleted, LR = lymphocyte rich classical, UCL = unclassified classical Hodgkin lymphoma, IF = interfollicular classicl hodgkin lymphoma, NS/MC = classical Hodgkin lymphoma with overlapping features between nodular sclerosis and mixed cellularity, ESR = erythrocyte sedimentation rate, LDH = serum lactate dehydrogenase, IPS = international prognostic score, # = number, AAS = Ann-Arbor stage.
Table 2. Correlation between serum β2-microglobulin levels and other—rarely reported—biological prognostic factors.
Table 2. Correlation between serum β2-microglobulin levels and other—rarely reported—biological prognostic factors.
Biological Prognostic FactorPatients with Available Data (#)Statistical Methodp-ValueComments
Serum Ferritin(ng/mL)399Spearman’s rho = 0.455<0.001Positive correlation
Serum soluble CD30204Spearman’s rho = 0.333<0.001Positive correlation
Serum interleukin10(pg/mL)204Spearman’s rho = 0.336<0.001Positive correlation
Bcl-2 expression102Mann–Whitney0.64-
Activated caspase-373Mann–Whitney0.79-
LMP-1 expression189Mann–Whitney0.10↑β2m in positive cases
Table 3. Univariate analysis of the prognostic significance of serum β2-microglobulin levels on various cutoff points in terms of freedom from progression.
Table 3. Univariate analysis of the prognostic significance of serum β2-microglobulin levels on various cutoff points in terms of freedom from progression.
CutoffAll PatientsStages IA/IIAStages IB/IIB/III/IV
(mg/L)Pts/Failed10y-FFPpPts/Failed10y-FFPpPts/Failed10y-FFPp
≤1.8236/36840.002181/22880.01255/17700.569
>1.8679/16973334/6279345/10766
≤1.9311/5284<0.001231/28880.00380/24700.431
>1.9604/15672284/5678320/10066
≤2.0396/6783<0.001290/39860.007106/28710.090
>2.0519/14170225/4578294/9664
≤2.1424/7582<0.001304/41860.007120/34710.211
>2.1491/13370211/4377280/9065
≤2.2464/8781<0.001326/48850.07138/39710.182
>2.2451/12170189/3677262/8564
≤2.3496/97800.001345/51850.063151/46700.477
>2.3419/11170170/3378249/7865
≤2.4532/105800.001365/54850.06167/51700.504
>2.4393/10570150/3078233/7365
≤2.5566/113800.001384/58840.115182/55700.428
>2.5349/9569131/2678218/6964
≤2.6596/11780<0.001401/60850.08195/57710.243
>2.6319/9168114/2976205/6763
≤2.7623/12380<0.001418/65840.286205/58720.107
>2.7292/856797/1978195/6662
≤2.8646/138790.002427/69830.743219/61700.188
>2.8269/756988/1581181/6063
≤2.9664/137790.002437/70830.600227/67700.256
>2.9251/716878/1480173/5760
≤3.0695/144780.002452/73830.714243/71700.282
>3.0220/646863/1180157/5363
≤3.5759/161780.002477/77830.538282/84690.291
>3.5156/476738/781118/4063
≤4.0810/178770.044490/80830.887320/98680.604
>4.0105/306825/48280/2664
FFP = freedom from progression.
Table 4. Multivariate analysis of the prognostic significance of serum β2-microbulin levels of freedom from progression, and overall and Hodgkin lymphoma-specific survival. Analysis performed at the cutoff of 2.0 mg/L or a “normal vs. elevated” basis (cutoff 2.4 mg/L) in the whole potent population and in early and advanced stages separately.
Table 4. Multivariate analysis of the prognostic significance of serum β2-microbulin levels of freedom from progression, and overall and Hodgkin lymphoma-specific survival. Analysis performed at the cutoff of 2.0 mg/L or a “normal vs. elevated” basis (cutoff 2.4 mg/L) in the whole potent population and in early and advanced stages separately.
Covariates Entering the Multivariate ModelSerum β2-Microglobulin
at the 2.0 mg/L Cutoff
Covariates Entering the Multivariate ModelSerum β2-Microglobulin on a “Normal vs. Elevated” Basis
Hazard Ratio95% Clp-Value Hazard Ratio95% Clp-Value
All patients—Freedom From Progression *
Clinical Stage Clinical Stage
 Stage IIB/III vs. I/IIA1.651.16–2.360.005 Stage IIB/III vs. I/IIA1.841.30–2.600.001
 Stage IV vs. I/IIA2.291.53–3.42<0.001 Stage IV vs. I/IIA2.591.75–3.85<0.001
Lymphopenia (yes vs. no)1.761.19–2.590.004Lymphopenia (yes vs. no)1.841.24–2.720.002
2m (>2.0 vs. ≤2 mg/L)1.551.11–2.170.012m (>2.4 vs. ≤2.4 mg/L)Notselected-
Early stages—Freedom From Progression (I/IIA) **
Nodal Sites # (≥3 vs.<3)1.971.24–3.160.005Nodal Sites # (≥3 vs. <3)2.001.24–3.210.004
ESR (≥50 vs.<50 mm/h)1.520.94–2.450.085ESR (≥50 vs.<50 mm/h)1.580.98–2.530.059
2m(>2.0 vs. ≤2 mg/L)1.651.04–2.620.0342m (>2.4 vs. ≤2.4 mg/L)1.671.03–2.720.038
Advanced Stages—Freedom From Progression (IIB/III/IV) ***
Lymphopenia (yes vs. no)2.311.51–3.54<0.001Lymphopenia (yes vs. no)2.361.54–3.61<0.001
WBC (≥15 vs. <15 × 109/L)0.610.38–0.990.047WBC (≥15 vs. <15 × 109/L)0.620.38–1.020.058
Stage (IV vs. IB/IIB/III)1.420.98–2.060.067Stage (IV vs. IB/IIB/III)1.440.99–2.08 0.057
2m (>2.0 vs. ≤2 mg/L)1.440.94–2.210.0982m (>2.4 vs. ≤2.4 mg/L)notselected-
All Patients—Overall Survival *
Age (≥45 vs. <45 years)2.631.73–3.99<0.001Age (≥45 vs.<45)2.641.70–4.07<0.001
B-symptoms (yes vs. no)2.011.31–3.070.001B–symptoms (yes vs. no)2.071.34–3.180.001
Lymphopenia (yes vs. no)1.831.07–3.120.027Lymphopenia (yes vs. no)1.841.08–3.15 0.021
2m (>2.0 vs. ≤2 mg/L)1.961.21–3.190.0062m (>2.4 vs. ≤2.4 mg/L)1.530.97-2.410.067
Early Stages (I/IIA)—Overall Survival **
Age (≥45 vs. <45 years)2.341.23–4.460.01Age (≥45 vs.<45)2.341.23–4.460.010
Gender (male vs. female)2.251.14–4.420.019Gender (male vs. female)2.251.14–4.420.019
2m (>2.0 vs. ≤2 mg/L)Notselected 2m (>2.4 vs. ≤2.4 mg/L)Notselected
Advanced Stages (IB/IIB/III/IV)—Overall Survival ***
Age (≥45 vs. <45 years)4.022.44–6.62<0.001Age (≥45 vs.<45 years)4.923.03–8.00<0.001
Lymphopenia (yes vs. no)2.571.46–4.520.001Lymphopenia (yes vs. no)2.351.35–4.080.003
Anemia (yes vs. no)1.740.99–3.060.054Anemia (yes vs. no)1.791.03–3.130.04
WBC (≥15 vs. <15 × 109/L)0.580.31–1.130.10WBC (≥15 vs. <15 × 109/L)notselected-
2m (>2.0 vs. ≤2 mg/L)2.071.04–4.150.0392m (>2.4 vs. ≤2.4 mg/L)notselected-
All Patients—Hodgkin Lymphoma Specific Survival *
B-symptoms (yes vs. no)3.111.75–5.53<0.001Clinical Stage
Lymphopenia (yes vs. no)2.171.15–4.090.017 Stage IIB/III vs. I/IIA2.641.35–5.180.005
WBC (≥10 vs. <10 × 109/L)0.520.30–0.900.019 Stage IV vs. I/IIA3.101.43–6.720.004
2m (>2.0 vs. ≤2 mg/L)2.211.19–4.110.012Lymphopenia (yes vs. no)2.761.48–5.150.001
WBC (≥10 vs. <10 × 109/L)0.510.29–0.890.019
Anemia (yes vs. no)1.790.97–3.290.061
2m (>2.4 vs. ≤2.4 mg/L)notselected
Early Stages (I/IIA)—Hodgkin Lymphoma Specific Survival **
2m (>2.0 vs. ≤2 mg/L)2.300.89–5.940.085No model fitted
Advanced Stages (IB/IIB/III/IV)—Hodgkin Lymphoma Specific Survival ***
Age (≥45 vs. <45 years)2.451.38–4.360.002Age (≥45 vs.<45 years)2.451.38–4.360.002
Lymphopenia (yes vs. no)2.991.57–5.680.001Lymphopenia (yes vs. no)2.991.57–5.680.001
Anemia (yes vs. no)2.121.05–4.250.035Anemia (yes vs. no)2.121.05–4.250.035
WBC (≥15 vs. <15 × 109/L)0.470.21–1.050.067WBC (≥15 vs. <15 × 109/L)0.470.21–1.050.067
2m (>2.0 vs. ≤2 mg/L)notselected-2m (>2.4 vs. ≤2.4 mg/L)notselected-
WBC = White Blood Cell count * Variables examined in the multivariate model for all 915 patients: age (≥45 vs. <45 years), gender (male vs. female), stage (IV vs. IIB/III vs. I/IIA), B-symptoms (yes vs. no), infradiaphragmatic disease (yes vs. no), albumin (≥4 vs. <4 g/dL), leukocytosis (≥10 vs. <10 × 109/L), anemia (yes vs. no), involved nodal sites (≥3 vs. <3), lymphopenia (yes vs. no), ESR (≥50 vs. <50 mm/h), and Sβ2m levels (>2.0 vs. ≤2 mg/L) or Sβ2m levels (>2.4 vs. ≤2.4 mg/L). ** Variables examined in the multivariate model for early-stage patients: age (≥45 vs. <45 years), gender (male vs. female), stage (II vs. I), leukocytosis (≥10 vs. <10 × 109/L), anemia (yes vs. no), involved nodal sites (≥3 vs. <3), ESR (≥50 vs. <50 mm/h), and Sβ2m levels (>2.0 vs. ≤2 mg/L) or Sβ2m levels (>2.4 vs. ≤2.4 mg/L). *** Variables examined in the multivariate model for all advanced-stage patients: age (≥45 vs. <45 years), gender (male vs. female), stage (IV vs. IB/IIB/III), albumin (≥4 vs. <4 g/dL), marked leukocytosis (≥15 vs. <15 × 109/L), anemia (yes vs. no), lymphopenia (yes vs. no), and Sβ2m levels (>2.0 vs. ≤2 mg/L) or Sβ2m levels (>2.4 vs. ≤2.4 mg/L).
Table 5. Summary of published studies on the prognostic significance of serum β2-microglobulin levels in patients with Hodgkin lymphoma.
Table 5. Summary of published studies on the prognostic significance of serum β2-microglobulin levels in patients with Hodgkin lymphoma.
StudyNo. of PatientsTreatment Prognostic Significance of β2-Microglobulin in Multivariate Analysis
Early StagesAdvanced StagesOverall
CutoffPFS/TTFOSPFS/TTFOSPFS/TTFOS
Oza et al., 1992 [65]60
(IIIB, IV)
MVPP ± RT
ChlvPP ± RT
3 mg/LNANA+ -NANA
Dimopoulos et al., 1993 [48]160RT only
NOVP ± RT, MOPP ± RT
Anthracycline-based (minority)
2.5 mg/L± *NT+ *,¶¶NT+ *NT
Fleury et al., 1994 [49]64
(age < 50 y)
MOPP ± RT
MOPP/ABVD ± RT
2.4 mg/LNTNTNTNT+NT
Axdorphet al., 2000 [64]99RT only
MOPP or CCNU-OPP
MOPP/ABVD ± RT
NRNTNTNTNT- ***- ***
Raida et al., 2002 [50]69NRNRNRNRNRNR- *NR
Chronowski et al., 2002 [51]191
(ES)
NOVP + RT, MOPP + RT
ABVD + RT
CVPP/ABDIC + RT
2.5 mg/L± **+NANANANA
Visco et al., 2004 [66]61
(ES, non-X)
RT only“elevated”+ *NTNANANANA
Vassilakopoulos et al., 2005 [53]379ABVD or equivalents ± RT2.4 mg/L++--- *+
Itoh et al., 2010 [67]167 (111) §ABVd § ± RT2.0 mg/LNTNRNTNRNT-
Nakajima et al., 2014 [55]67ABVD ± RT2.5 mg/L §§NRNRNRNR+-
Wang et al., 2016 [56]202 (IIX, III/IV)ABVD ± RT2.5 mg/L §§NANA+ *+NANA
Miriliet al., 2019 [68]122RT only
ABVD ±RT
2.2 mg/L §NTNTNTNT-+
Wen et al., 2022 [69]365ABVD or equivalents ± RT1.85 mg/L §NTNTNTNT-+
Present Study, 2023915ABVD or equivalents ± RT2.0 mg/L+ *-± *++ *+
NA = Not Applicable, NT = Not Tested, NR = Not Reported, ES = early stage, non-X = non bulky. * Tumor control was the endpoint either as TTF or FFP (with progression/relapse counted as events along with treatment- or disease-related deaths or not; deaths of any other cause were censored). ** RFS was the endpoint (only recurrence counted as event). *** DFS and cause-specific survival were the endpoints. § 111/167 had sβ2m levels available; ABVd = ABVD with reduced dacarbazine doses. §§ Cutoff determined by ROC curve analysis. Independent prognostic factor for achievement of CR but not for disease-free survival. ¶¶ The effect of sβ2m within early and advanced stages was tested only in univariate analysis.
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

Vassilakopoulos, T.P.; Arapaki, M.; Diamantopoulos, P.T.; Liaskas, A.; Panitsas, F.; Siakantaris, M.P.; Dimou, M.; Kokoris, S.I.; Sachanas, S.; Belia, M.; et al. Prognostic Impact of Serum β2-Microglobulin Levels in Hodgkin Lymphoma Treated with ABVD or Equivalent Regimens: A Comprehensive Analysis of 915 Patients. Cancers 2024, 16, 238. https://doi.org/10.3390/cancers16020238

AMA Style

Vassilakopoulos TP, Arapaki M, Diamantopoulos PT, Liaskas A, Panitsas F, Siakantaris MP, Dimou M, Kokoris SI, Sachanas S, Belia M, et al. Prognostic Impact of Serum β2-Microglobulin Levels in Hodgkin Lymphoma Treated with ABVD or Equivalent Regimens: A Comprehensive Analysis of 915 Patients. Cancers. 2024; 16(2):238. https://doi.org/10.3390/cancers16020238

Chicago/Turabian Style

Vassilakopoulos, Theodoros P., Maria Arapaki, Panagiotis T. Diamantopoulos, Athanasios Liaskas, Fotios Panitsas, Marina P. Siakantaris, Maria Dimou, Styliani I. Kokoris, Sotirios Sachanas, Marina Belia, and et al. 2024. "Prognostic Impact of Serum β2-Microglobulin Levels in Hodgkin Lymphoma Treated with ABVD or Equivalent Regimens: A Comprehensive Analysis of 915 Patients" Cancers 16, no. 2: 238. https://doi.org/10.3390/cancers16020238

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