Next Article in Journal
Current Status and Perspectives of Dual-Targeting Chimeric Antigen Receptor T-Cell Therapy for the Treatment of Hematological Malignancies
Next Article in Special Issue
Classifying Germinal Center Derived Lymphomas—Navigate a Complex Transcriptional Landscape
Previous Article in Journal
Tumor Microenvironment and Immunotherapy-Based Approaches in Mantle Cell Lymphoma
Previous Article in Special Issue
Biological and Clinical Implications of Gene-Expression Profiling in Diffuse Large B-Cell Lymphoma: A Proposal for a Targeted BLYM-777 Consortium Panel as Part of a Multilayered Analytical Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Approaches in Molecular Characterization of Classical Hodgkin Lymphoma

by
Diede A. G. van Bladel
1,2,
Wendy B. C. Stevens
3,
Michiel van den Brand
1,4,
Leonie I. Kroeze
1,
Patricia J. T. A. Groenen
1,
J. Han J. M. van Krieken
1,
Konnie M. Hebeda
1 and
Blanca Scheijen
1,2,*
1
Radboud University Medical Center, Department of Pathology, 6525 GA Nijmegen, The Netherlands
2
Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
3
Radboud University Medical Center, Department of Hematology, 6525 GA Nijmegen, The Netherlands
4
Pathology-DNA, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(13), 3222; https://doi.org/10.3390/cancers14133222
Submission received: 29 May 2022 / Revised: 24 June 2022 / Accepted: 27 June 2022 / Published: 30 June 2022
(This article belongs to the Special Issue Molecular Characterization of Hematological Tumors)

Abstract

:

Simple Summary

The unique tumor composition of classical Hodgkin lymphoma (cHL), with only a small fraction of malignant Hodgkin and Reed–Sternberg cells within the tumor tissue, has created many challenges to characterize the genetic alterations that drive this lymphoid malignancy. Major advances in sequencing technologies and detailed analysis of circulating tumor DNA in blood samples of patients have provided important contributions to enhance our understanding of the pathogenesis of cHL. In this review, we provide an overview of the recent advances in genotyping the clonal and mutational landscape of cHL. In addition, we discuss different next-generation sequencing applications to characterize tumor tissue and cell-free DNA, which are now available to improve the diagnosis of cHL, and to monitor therapeutic response or disease progression during treatment and follow up of cHL patients.

Abstract

Classical Hodgkin lymphoma (cHL) represents a B-cell lymphoproliferative disease characterized by clonal immunoglobulin gene rearrangements and recurrent genomic aberrations in the Hodgkin Reed–Sternberg cells in a reactive inflammatory background. Several methods are available for the molecular analysis of cHL on both tissue and cell-free DNA isolated from blood, which can provide detailed information regarding the clonal composition and genetic alterations that drive lymphoma pathogenesis. Clonality testing involving the detection of immunoglobulin and T cell receptor gene rearrangements, together with mutation analysis, represent valuable tools for cHL diagnostics, especially for patients with an atypical histological or clinical presentation reminiscent of a reactive lesion or another lymphoma subtype. In addition, clonality assessment may establish the clonal relationship of composite or subsequent lymphoma presentations within one patient. During the last few decades, more insight has been obtained on the molecular mechanisms that drive cHL development, including recurrently affected signaling pathways (e.g., NF-κB and JAK/STAT) and immune evasion. We provide an overview of the different approaches to characterize the molecular composition of cHL, and the implementation of these next-generation sequencing-based techniques in research and diagnostic settings.

Graphical Abstract

1. Introduction

Classical Hodgkin lymphoma (cHL) is one of the most common lymphoma types in the Western world, with an age-adjusted incidence rate of about 3 cases per 100,000 people [1]. Most cHL patients are diagnosed around young adulthood, followed by a second peak at the age of 60 years and older. There are four different histological subtypes defined for cHL, of which nodular sclerosis (NSCHL) and mixed cellularity (MCCHL) are the most common [2].
The cellular composition and morphology of cHL are unique among the different B-cell lymphomas. cHL consists of a minor neoplastic B-cell component (usually <1–5%) represented by enlarged mononucleated Hodgkin cells and multinucleated Reed–Sternberg cells, referred to as Hodgkin and Reed–Sternberg (HRS) cells, which are surrounded by a variable mixture of inflammatory cells, including reactive B- and T-cells, histiocytes and granulocytes [3,4,5]. The diagnosis for most patients is based on clinical presentation and histopathology, including the detection of CD30-positive HRS cells. Occasionally, cHL is difficult to distinguish from reactive lymphoproliferations or B- or T-cell lymphomas with HRS-like cells [6]. In such cases, molecular assays, including the detection of clonal immunoglobulin (IG) and T-cell receptor (TR) gene rearrangements or gene mutation analysis, can be essential for the correct diagnosis and treatment. Besides de novo cHL, this specific lymphoma subtype may occasionally develop as a transformation from low-grade B-cell lymphoma [7,8]. Although cHL is highly curable with multiagent chemotherapy and modern radiation techniques, up to 30% of patients with advanced stage cHL have progressive disease or develop a relapse [2,9,10]. Clonality testing and mutational profiling can be valuable techniques to assess the clonal relationship between the primary tumor and the clinical recurrence or a second lymphoid malignancy.
Because of the small fraction of neoplastic HRS cells in cHL, conventional clonality assays and mutation analysis have often failed to detect the molecular profiles of the HRS cells. With specialized techniques to isolate or enrich HRS cells, mutation and clonality analyses appeared technically more feasible, but these approaches are time- and labor-intensive and therefore not suitable for clinical practice. The major advances of next-generation sequencing (NGS) technologies and analysis of cell-free DNA (cfDNA) in the blood of lymphoma patients in the last decade have made important contributions to improve our understanding of the molecular pathogenesis of cHL [11,12,13]. Despite the relatively low number of HRS cells in cHL tumor tissues, there is a relatively high concentration of tumor-derived cfDNA, also known as circulating tumor DNA (ctDNA), present in the peripheral blood of most reported cHL patients, with potential for improved diagnostics and monitoring of cHL patients during treatment [14]. Here, we provide an overview of the different NGS-based molecular approaches that are available to characterize cHL in both tumor tissue and cfDNA.

2. Clonality Assessment in Classical Hodgkin Lymphoma Tissue

During early B- and T-lymphoid development, the different germline variable (V), diversity (D) and joining (J) genes of the IG and TR genes are recombined, a process called V(D)J recombination (Figure 1). This recombination process results in a unique antigen-receptor molecule on the surface of the mature lymphocytes. During further maturation of B cells within the germinal centers, the B lymphocytes extend their IG repertoire for antigen recognition by somatic hypermutation (SHM) [15,16]. The unique clonal IG/TR gene rearrangements, due to the shared cell of origin of the malignant lymphoid cells within one tumor, make the detection of these tumor-specific V(D)J rearrangements (i.e., clonotypes) a highly valuable molecular marker for clonality testing of lymphoproliferative diseases. Although cHL is characterized by the lack of a B-cell receptor on the cell surface, which is in contrast to most other B-cell lymphomas [17], genomic IG gene rearrangements have occurred during the premalignant B-cell development stage. These IG gene rearrangements can therefore be detected as a molecular fingerprint for an individual cHL tumor, and may involve both functional and non-functional IG gene rearrangements [17,18]. The low number of HRS cells in a background of many reactive immune cells, including B cells, and the high level of SHM potentially hampering efficient primer binding, makes detection of clonal IG rearrangements challenging in cHL whole tumor tissue specimens. Furthermore, the presence of reactive B cells within the tumor microenvironment (TME) may further complicate the analysis of clonality testing in cHL. These non-malignant B cells may undergo clonal expansion, which eventually results in minor clones.
Initially, clonality detection was performed using Southern blot analysis, with a large variety in the IG clone detection rate for cHL cases, ranging from 0% to 87% [19,20,21,22,23,24,25,26]. The introduction of more sensitive PCR-based techniques improved clonality detection for this type of lymphoma [27,28,29] (Table 1, upper panel). In the early 2000s, clonality detection was further optimized by the introduction of standardized multiplex PCR protocols as developed by the BIOMED-2 Concerted Action, nowadays called the EuroClonality consortium [30]. This BIOMED-2/EuroClonality assay in combination with GeneScan fragment size analysis yielded a mean clonality detection rate of 57% (range 26–79%) in cHL (Table 1, upper panel) [31,32,33,34,35,36,37,38,39]. The improvements in clonality testing based on the BIOMED-2/EuroClonality assay were achieved by the complementarity of the assays using multiple targets in both IGH and IGK loci [40,41]. Other groups introduced an additional semi-nested PCR protocol [35]. Other approaches that have been employed for the detection of IG gene rearrangements in cHL involved the enrichment or isolation of single HRS cells, which obviously increased the sensitivity and specificity of clonality detection (Table 1, upper panel) [17,18,26,42,43,44,45,46,47,48,49]. However, not every study was able to detect IG gene rearrangements in these cases, which could relate to the lack of HRS cell nuclei in thin tissue sections used for isolation, or mismatches of consensus primers due to SHM.
An important recent innovation for clonality assessment involved the introduction of NGS-based clonality testing on tissue biopsies by the EuroClonality-NGS Working Group [55,56]. This approach clearly improved clonality assessment in cHL as compared to the BIOMED-2/EuroClonality assay [51]. Other NGS-based assays, either amplicon-based (e.g., ClonoSEQ technology [50]) or hybrid capture-based [52], have also been used for clonality detection in cHL. These approaches have resulted in higher clone detection rates (range 38–89%) compared to conventional clonality testing, and included analysis in both cHL tissue DNA and cfDNA (Table 1, upper panel) [50,51,52]. A clear advantage of NGS-based clonality testing is the availability of the exact clonotype sequence. This allows for the specific detection of minor clones in a polyclonal background, as is the case in cHL. Several studies have also investigated the presence of clonal TR gene rearrangements in cHL (Table 1, lower panel). Although cHL is considered a B-cell lymphoma, the detection of clonal TR gene rearrangements in isolated HRS cells has suggested a T-cell origin in some cases of cHL [42,47,53,54,57,58]. TR clonality testing can also assist in the differential diagnosis of cHL with Hodgkin mimickers, such as angioimmunoblastic T-cell lymphoma (AITL) with a Hodgkin-like B-cell component or the follicular variant of peripheral T-cell lymphoma [36,59,60].
Finally, clonality assessment is highly informative to establish the clonal relationship of cHL recurrences, which has been investigated in only one study with a significant cohort size [61]. For 20 cHL patients with cHL recurrences, laser microdissection (LMD) was performed followed by Sanger sequencing of isolated HRS cells to compare tumor-specific clonotypes in the primary and subsequent cHL presentation. This study showed that in 8 out of 20 cHL patients (40%), the primary diagnosis and recurrence represented clonally unrelated lymphomas, suggesting the occurrence of second primary lymphoma in a significant number of cHL patients. Obviously, additional studies in larger cHL patient cohorts are required to confirm this observation.

3. New Developments in Molecular Testing of Classical Hodgkin Lymphoma with cfDNA

Currently, there is emerging interest in cfDNA analysis as a tool for non-invasive tumor detection and monitoring in clinical practice. Although cfDNA is commonly present in healthy subjects due to apoptosis of hematopoietic cells, studies show that cfDNA of cancer patients harbors tumor-derived fragmented DNA, known as circulating tumor DNA (ctDNA), which may provide molecular information related to the tumor [62]. The high sensitivity of sequencing techniques and droplet digital PCR (ddPCR) have enabled the use of ctDNA for early tumor detection, molecular profiling and monitoring of therapy response or disease progression [14,52,63,64,65,66,67]. Furthermore, the introduction of unique molecular identifiers (UMIs) for error suppression during next-generation sequencing [68,69] has resulted in even more sensitive tests for mutation detection in diagnostic settings. Since cfDNA has a short half-life of about 15 min to a few hours in the bloodstream, analysis of this type of DNA reflects the mutation status of the tumor at time of sampling, making it highly suitable for disease monitoring during treatment [70]. The analysis of cfDNA may also be of added value when tissue sampling is difficult due to localization of the tumor, or for better characterization of a heterogeneous tumor [70]. Of note, ctDNA isolation from plasma after blood sampling requires a special workflow of either EDTA tubes (processing time < 4 h) or nucleic acid preservation tubes (Roche or Streck cfDNA BCT; processing time < 24 h). A short processing time is necessary to avoid degradation of leukocytes and subsequent contamination of the ctDNA fraction.
A substantial part of cfDNA that is present in blood is derived from hematopoietic cells and may harbor non-tumor-derived mutations as a result of clonal hematopoiesis [71]. Clonal hematopoiesis is a normal process of aging and involves the accumulation of somatic mutations in hematopoietic stem cells, leading to clonal expansion of such mutations in blood cells [72]. The genes most frequently affected in clonal hematopoiesis are DNMT3A, TET2, ASXL1, TP53, JAK2, SF3B1, SRSF2 and PPM1D [73,74,75,76]. Although there is only limited overlap with cHL-associated genes, the presence of such variants in cfDNA of lymphoma patients may complicate ctDNA analysis, unless paired deep sequencing of plasma cfDNA and DNA from white blood cells has been performed [77].
One of the first studies providing evidence that genetic features of HRS cells can be detected in cfDNA involved non-invasive prenatal testing in an asymptomatic pregnant woman with an abnormal genomic profile who subsequently was diagnosed with cHL [78]. The possibility to detect genetic features in cfDNA was confirmed in nine additional patients with newly diagnosed cHL. Subsequent studies have confirmed the potential added value of analyzing cfDNA for cHL diagnostics and monitoring [52,63,64,65,66,67]. Using targeted sequencing, tumor-derived somatic mutations in cfDNA were identified with high detection rates, which were validated by the analysis of paired tumor tissue biopsies. In a recent study, gene-specific mutations were detected with ddPCR in cfDNA samples, demonstrating that this approach is also feasible in cHL diagnostics [79]. Taken together, these studies have shown that genotyping of cfDNA in cHL allows for at least equal performance of mutation detection as in whole tissue. This might be related to the high turnover rate of HRS cells, resulting in relatively larger amounts of ctDNA, despite the low HRS cell number in tumor tissues [14].
Potentially clinically relevant applications of cfDNA involve the detection of minimal residual disease (MRD) to monitor therapy response, as has been applied in different lymphoma types [80,81], including cHL [50,82], and for prognostic stratification [14,52,82,83,84,85]. This requires sensitive NGS-based assays, such as ClonoSEQ (formerly known as LymphoSIGHT) [81,86], CAPP-Seq [87], PhasED-Seq [88], or the EuroClonality-NGS DNA capture (EuroClonality-NDC) assay [89]. Despite the promising results obtained in different laboratories that have performed mutation and clonality analysis in cfDNA of cHL patients, there are still several challenges regarding the sensitivity and reproducibility of these assays. Large prospective clinical studies are required before cfDNA testing can be implemented in routine diagnostics and monitoring of cHL.

4. Molecular Pathogenesis of Classical Hodgkin Lymphoma

Accumulating genetic alterations within the HRS cells and infection by Epstein–Barr virus (EBV) play an important role in the pathogenesis of cHL [11,13,90]. Given the scarcity of the malignant HRS cells in cHL tissue, major advances to unravel the molecular pathogenesis of cHL have been achieved by whole exome sequencing (WES) on enriched HRS cells [91,92,93]. This confirmed the high genomic instability of HRS cells that was known from cytogenetic studies [12,94,95,96], which includes somatic mutations, translocations involving, among others, IG loci with various translocation partners [97], and recurrent copy number alterations (CNAs), including gains of chromosomal arms 2p, 9p, 16p, and 17q and losses of 13q, 6q, and 11q [95,98,99,100]. In fact, 9p24.1 gains are detected in almost all cHL cases and represent a hallmark of cHL. The genes encoding JAK2, as well as PD-1 ligands PD-L1 and PD-L2, are located on this locus, which all play an important role in cHL pathogenesis [101]. More recent studies were also able to detect mutations in genomic DNA of cHL whole tissue specimens, using targeted deep sequencing [79]. These approaches have been complemented by hybrid capture-based sequencing of ctDNA, which has allowed the detection of both tumor-specific gene mutations [52,63,64,65,66,67], and CNAs in cHL [102].

4.1. Mutational Landscape of Classical Hodgkin Lymphoma

Detailed molecular analyses of HRS cells and ctDNA in cHL patients have revealed the presence of recurrent gene mutations and genomic alterations that are involved in signaling pathways promoting cell proliferation and survival, as well as immune evasion (Figure 2 and Table 2) [14,52,64,66,67,79,91,92,93]. The most frequently affected pathway in cHL represents the JAK/STAT signaling pathway, which plays a central role in promoting cell proliferation and survival. JAK/STAT signaling is constitutively activated in HRS cells, as a result of inactivating mutations in its negative regulators, such as SOCS1 and PTPN1, activating mutations in STAT6 and overexpression of JAK2, due to focal gene amplifications or copy number gains of 9p24. HRS cell proliferation and survival are also regulated by the NF-κB pathway, which is constitutively activated by genetic alterations targeting TNFAIP3, NFKBIA, NFKBIE, IKBKB, and REL. In addition, HRS cells escape antitumor immune responses via inhibition of PD-1-expressing immune cells, such as cytotoxic T cells, due to overexpression of the PD-1 ligands PD-L1 and PD-L2. Other factors that induce immune evasion of HRS cells include inactivating alterations of B2M and CIITA, encoding components or regulators of MHC class I and II expression, respectively. Inactivating B2M mutations have been related to the NSCHL subtype, and a lack of B2M correlates with better clinical outcomes [91]. STAT6 and TNFAIP3 are more frequently mutated in NSCHL, and STAT6 mutations are more often observed in younger patients [67]. Others have also observed a correlation between ITPKB and B2M mutations and more disseminated disease [66]. However, additional studies are required to confirm the prognostic value of specific gene alterations and associations with certain cHL subtypes.
Other pathways and gene functions that are perturbed in cHL include the PI3K/AKT pathway, chromatin remodeling, nuclear–cytoplasmic transport and the p53 pathway. Activation of the PI3K/AKT pathway involves mutations in ITPKB and PIK3CA, which, together with inactivating GNA13 mutations [103,104,105], promote lymphoma cell survival. Gene mutations affecting proteins involved in chromatin remodeling are frequently observed in different B-cell malignancies. In cHL, the tumor suppressor gene ARID1A is among the most frequently mutated genes, and a component of the SWI/SNF chromatin remodeling complex. Recently, it was shown that ARID1A deficiency correlates with a higher mutation load and improved sensitivity to immune checkpoint blockade using anti-PD-L1 treatment in a preclinical ovarian cancer model [106]. It remains to be determined whether ARID1A mutations show predictive value in cHL immune checkpoint blockade. Furthermore, about 20% of analyzed cHL patients show mutations in XPO1, of which the majority shows the E571K hotspot mutation [52,64,65,67,92,93]. This gene is involved in nuclear–cytoplasmic transport and its alteration results in disturbed homeostasis of HRS cells. Interestingly, detectable XPO1 mutations at the end of treatment seem to result in shorter progression-free survival compared to patients with undetectable XPO1 mutations after treatment, and could potentially be used as a predictive marker [65].
The importance of the p53 pathway in cHL pathogenesis has remained controversial. In some studies, TP53 mutations were found to be rare (<10%) [52,92,107], while others reported higher frequencies of TP53 mutations (~25%) [79,108,109]. It is very plausible that EBV status may act as a confounding factor, since TP53 mutations are mainly detected in EBV-negative cases [108]. Mutations in TP53 can stabilize p53 protein due to conformational changes, which is visible by increased expression of p53 protein by immunohistochemistry, as is frequently observed in the nuclei of HRS cells [107,108,110]. Since mutated p53 protein can promote the survival of genomically instable cells, such as HRS cells, it is likely that reduced p53 function plays a role in the pathogenesis of cHL.

4.2. The Role of Epstein–Barr Virus Infection in Classical Hodgkin Lymphoma Pathogenesis

In 20–40% of the cHL cases in the Western world and more than 90% of pediatric cHL cases in developing countries, the lymphoma is EBV-positive, and there is compelling evidence that EBV serves as a tumor-initiating factor in cHL [5,111,112,113]. There are various EBV infection patterns known for lymphoid cells, each with a different range of EBV latent antigen expression. EBV-infected HRS cells in cHL display the latency type II program with expression of EBV latent genes LMP1, LMP2A, LMP2B and EBNA1 together with EBER non-coding RNAs and BART miRNAs, which all play a role in the pathogenesis of cHL (Figure 3) [113,114]. In most B cells, expression of the BCR serves as a survival signal. Since HRS cells lack expression of a functional BCR, LMP1 and LMP2A rescue HRS cells from apoptosis. Transmembrane protein LMP1 shows functional similarities with constitutive active CD40 signaling, while LMP2A functions as a BCR mimic with the ability to activate the RAS/MAPK pathway [90,113,115,116]. Together, these EBV proteins mediate activation of downstream signaling pathways (e.g., NF-κB, JAK/STAT and PI3K/AKT), and modulate the expression of cellular genes regulating B-cell proliferation, survival, lineage commitment/differentiation and immune escape [114,117]. The EBV latent protein EBNA1 is a key viral replication factor and important for maintaining the viral genome during cell division. In addition, it is capable of acting as a transcription factor, thereby regulating gene expression that promotes cell survival and contributing to immune evasion of EBV-infected HRS cells [90,118]. EBV-encoded small RNAs, or EBERs, inhibit apoptosis and induce the transcription of cytokines, resulting in HRS cell survival [114,119,120]. The exact roles of BART miRNAs in cHL pathogenesis are just starting to be explored. BART miRNAs contribute to HRS cell survival and reduction in tumor elimination by the immune system [121]. The pathogenic role of EBV infection in cHL development is further supported by the observation that EBV-negative cHL cases show a significantly higher mutational burden and more chromosomal abnormalities, compared to EBV-positive cases [92,93,122,123,124]. This underscores the finding that these viral proteins substitute the requirement for specific oncogene and tumor suppressor gene alterations in cHL pathogenesis. Indeed, TNFAIP3 and TP53 mutations and aberrant activation of receptor tyrosine kinases occur predominantly in EBV-negative cases [111,125,126].

4.3. T-Cell Immune Microenvironment in Classical Hodgkin Lymphoma

The tumor microenvironment (TME) of cHL is unique, as it contains a large admixture of inflammatory immune cells and stromal cells. The composition of the TME is actively orchestrated by the HRS cells via the production of different chemokines and interactions with the surrounding immune cells in the TME [111,127,128]. Many of the immune cells within the TME support the survival and proliferation of HRS cells, while others have the potential to eliminate the cHL tumor cells [129]. There is significant heterogeneity in the TME composition between individual cHL specimens, but T cells represent one of the more prevalent populations. These include mainly CD4+ T helper (Th) and regulatory T cells (Tregs), and to a lesser extent CD8+ cytotoxic T cells [127,129]. Single-cell expression profiling has revealed the expansion of Treg cell features in cHL as compared to reactive lymph nodes, displaying relatively high expression of LAG3+ and CTLA4+ Treg cell clusters in cHL samples [130]. Other non-Treg CD4+ T-cell clusters that are enriched in cHL samples include type 17 T helper (Th17) cells, which are more predominant in EBV-negative cases [130]. On the other hand, the proportion of type 1 T helper (Th1) cells is significantly enriched in lymphocyte-rich and mixed cellularity cHL [131]. Other quantitative approaches to investigate the cHL immune microenvironment involved mass cytometry, which showed concomitant expansion of Th1-polarized T effector cells and Tregs in cHL [132]. Due to the T cell-rich TME of cHL, minor T-cell clones can be detected with clonality testing in whole tumor tissue samples. These may represent clonal expansions of tumor- or EBV-reactive T cells.
A unique feature of cHL is the close proximity of rosetting CD4+ T cells. Such associations involve soluble factors and surface ligand–receptor interactions (e.g., CD40), and via pairs of adhesion molecules (i.e., CD54–CD11a and CD58–CD2). Furthermore, this CD40–CD40L interaction serves as an activation signal for the NF-κB pathway, which is important in cHL pathogenesis as described above. The rosetting T cells are also believed to protect the HRS cells from recognition and elimination by cytotoxic T cells and natural killer cells [127,133]. This is especially important in EBV-positive cHL, as such HRS cells express viral antigens that may be recognized by cytotoxic cells. However, HRS cells often have lost MHC class I and II molecules on their cell surface, which also contributes to immune escape via a reduced immunogenicity and impaired activation of T cells [129,134].

5. Conclusions and Future Perspectives

Although the molecular analysis of cHL has been hampered for many years by the low frequencies of malignant HRS cells, recent developments in sequencing technologies and analysis of cfDNA have made it possible to unravel the genetic features of cHL, which will further advance cHL diagnostics. This includes both improvements in clonality detection and the identification of recurrently mutated genes. In particular, the combination of IG and TR clonality testing may be of added value to correctly diagnose patients with cHL or T-non-Hodgkin lymphoma (T-NHL) with HRS-like cells. To further support the diagnosis of a possible T-NHL, targeted mutation analysis can be performed for recurrently mutated genes (e.g., RHOA, TET2, DNMT3A and IDH2 for AITL [135]). With the advances of single cell analyses (whole exome and/or genome sequencing), additional insights in cHL driver mutations and clonal dynamics can be obtained.
Until now, no universal molecular marker has been identified that could serve as a disease-specific genetic marker for diagnosis and longitudinal monitoring of therapy response in cHL patients. One interesting development in molecular testing is the molecular profiling of ctDNA within the fraction of plasma cfDNA in lymphoma patients. Given the high concordance of mutations between cfDNA and tumor DNA, and higher variant allele frequencies in cfDNA, this represents a promising approach for future molecular studies. Especially for risk-stratification, monitoring of therapy response and early detection of recurrences, screening of robust biomarkers in routine diagnostics with targeted NGS or ddPCR will be highly valuable. The recurrently mutated XPO1 gene might be a candidate biomarker, since the vast majority of XPO1 mutations involve the E571K hotspot mutation. Detectable XPO1 mutation by digital PCR at the end of treatment was shown to have potential predictive value for a shorter progression-free survival [65]. Additional patient-specific biomarkers can be retrieved from the identified IG gene rearrangements that could be monitored in cfDNA for the detection of MRD. Furthermore, cfDNA could be a highly interesting source to study the clonal evolution of relapsed disease, as previously described for different types of solid tumors [136,137,138,139] and B-cell lymphomas [140,141], including cHL [67]. Such longitudinal monitoring may provide information about the mechanisms of therapy resistance, and improve personalized treatment.
In conclusion, the major improvements in molecular testing of cHL have enhanced our understanding regarding cHL pathogenesis. These advances in NGS-based technologies will provide novel opportunities to improve diagnostics, disease monitoring and personalized therapies for cHL patients in daily clinical practice.

Author Contributions

Conceptualization, D.A.G.v.B., B.S. and K.M.H.; writing—original draft preparation, D.A.G.v.B.; writing—review and editing, B.S., W.B.C.S., M.v.d.B., L.I.K., P.J.T.A.G., J.H.J.M.v.K. and K.M.H.; visualization, D.A.G.v.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Dutch Cancer Society (KWF Kankerbestrijding), grant number 11137.

Acknowledgments

All figures were created with BioRender.com (accessed on 8 February 2022).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Glaser, S.L.; Clarke, C.A.; Keegan, T.H.; Chang, E.T.; Weisenburger, D.D. Time Trends in Rates of Hodgkin Lymphoma Histologic Subtypes: True Incidence Changes or Evolving Diagnostic Practice? Cancer Epidemiol. Biomark. Prev. 2015, 24, 1474–1488. [Google Scholar] [CrossRef] [Green Version]
  2. Swerdlow, S.H.; Campo, E.; Harris, N.L.; Pileri, S.A. Myeloproliferative neoplasms. In WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues; International Agency for Research on Cancer: Lyon, France, 2017. [Google Scholar]
  3. Aldinucci, D.; Gloghini, A.; Pinto, A.; De Filippi, R.; Carbone, A. The classical Hodgkin’s lymphoma microenvironment and its role in promoting tumour growth and immune escape. J. Pathol. 2010, 221, 248–263. [Google Scholar] [CrossRef]
  4. Campo, E.; Harris, N.L.; Pileri, S.A.; Jaffe, E.S.; Stein, H.; Thiele, J. Mature B-cell neoplasms. In WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues; International Agency for Research on Cancer: Lyon, France, 2017. [Google Scholar]
  5. Kuppers, R.; Engert, A.; Hansmann, M.L. Hodgkin lymphoma. J. Clin. Investig. 2012, 122, 3439–3447. [Google Scholar] [CrossRef]
  6. Parente, P.; Zanelli, M.; Sanguedolce, F.; Mastracci, L.; Graziano, P. Hodgkin Reed-Sternberg-Like Cells in Non-Hodgkin Lymphoma. Diagnostics 2020, 10, 1019. [Google Scholar] [CrossRef]
  7. Agbay, R.L.; Loghavi, S.; Medeiros, L.J.; Khoury, J.D. High-grade Transformation of Low-grade B-cell Lymphoma: Pathology and Molecular Pathogenesis. Am. J. Surg. Pathol. 2016, 40, e1–e16. [Google Scholar] [CrossRef]
  8. Montoto, S.; Fitzgibbon, J. Transformation of indolent B-cell lymphomas. J. Clin. Oncol. 2011, 29, 1827–1834. [Google Scholar] [CrossRef] [Green Version]
  9. Montanari, F.; Diefenbach, C. Relapsed Hodgkin lymphoma: Management strategies. Curr. Hematol. Malig. Rep. 2014, 9, 284–293. [Google Scholar] [CrossRef] [Green Version]
  10. Shanbhag, S.; Ambinder, R.F. Hodgkin lymphoma: A review and update on recent progress. CA Cancer J. Clin. 2018, 68, 116–132. [Google Scholar] [CrossRef]
  11. Brune, M.M.; Juskevicius, D.; Haslbauer, J.; Dirnhofer, S.; Tzankov, A. Genomic Landscape of Hodgkin Lymphoma. Cancers 2021, 13, 682. [Google Scholar] [CrossRef]
  12. Cuceu, C.; Hempel, W.M.; Sabatier, L.; Bosq, J.; Carde, P.; M’Kacher, R. Chromosomal Instability in Hodgkin Lymphoma: An In-Depth Review and Perspectives. Cancers 2018, 10, 91. [Google Scholar] [CrossRef] [Green Version]
  13. Weniger, M.A.; Küppers, R. Molecular biology of Hodgkin lymphoma. Leukemia 2021, 35, 968–981. [Google Scholar] [CrossRef]
  14. Camus, V.; Jardin, F. Cell-Free DNA for the Management of Classical Hodgkin Lymphoma. Pharmaceuticals 2021, 14, 207. [Google Scholar] [CrossRef]
  15. Kuppers, R.; Klein, U.; Hansmann, M.L.; Rajewsky, K. Cellular origin of human B-cell lymphomas. N. Engl. J. Med. 1999, 341, 1520–1529. [Google Scholar] [CrossRef]
  16. Neuberger, M.S.; Milstein, C. Somatic hypermutation. Curr. Opin. Immunol. 1995, 7, 248–254. [Google Scholar] [CrossRef]
  17. Marafioti, T.; Hummel, M.; Foss, H.D.; Laumen, H.; Korbjuhn, P.; Anagnostopoulos, I.; Lammert, H.; Demel, G.; Theil, J.; Wirth, T.; et al. Hodgkin and reed-sternberg cells represent an expansion of a single clone originating from a germinal center B-cell with functional immunoglobulin gene rearrangements but defective immunoglobulin transcription. Blood 2000, 95, 1443–1450. [Google Scholar] [CrossRef]
  18. Kuppers, 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. [Google Scholar] [CrossRef] [Green Version]
  19. Brinker, M.G.; Poppema, S.; Buys, C.H.; Timens, W.; Osinga, J.; Visser, L. Clonal immunoglobulin gene rearrangements in tissues involved by Hodgkin’s disease. Blood 1987, 70, 186–191. [Google Scholar] [CrossRef]
  20. Daus, H.; Schwarze, G.; Pees, H.; Radtke, H.; Kumel, G.; Scheurlen, P.G. Immunoglobulin and T cell receptor gene rearrangements in lymphoproliferative disorders. Exp. Cell Biol. 1989, 57, 177–184. [Google Scholar] [CrossRef]
  21. Griesser, H.; Feller, A.C.; Mak, T.W.; Lennert, K. Clonal rearrangements of T-cell receptor and immunoglobulin genes and immunophenotypic antigen expression in different subclasses of Hodgkin’s disease. Int. J. Cancer 1987, 40, 157–160. [Google Scholar] [CrossRef]
  22. Herbst, H.; Tippelmann, G.; Anagnostopoulos, I.; Gerdes, J.; Schwarting, R.; Boehm, T.; Pileri, S.; Jones, D.B.; Stein, H. Immunoglobulin and T-cell receptor gene rearrangements in Hodgkin’s disease and Ki-1-positive anaplastic large cell lymphoma: Dissociation between phenotype and genotype. Leuk. Res. 1989, 13, 103–116. [Google Scholar] [CrossRef]
  23. Knowles, D.M., 2nd; Neri, A.; Pelicci, P.G.; Burke, J.S.; Wu, A.; Winberg, C.D.; Sheibani, K.; Dalla-Favera, R. Immunoglobulin and T-cell receptor beta-chain gene rearrangement analysis of Hodgkin’s disease: Implications for lineage determination and differential diagnosis. Proc. Natl. Acad. Sci. USA 1986, 83, 7942–7946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Raghavachar, A.; Binder, T.; Bartram, C.R. Immunoglobulin and T-cell receptor gene rearrangements in Hodgkin’s disease. Cancer Res. 1988, 48, 3591–3594. [Google Scholar] [PubMed]
  25. Roth, M.S.; Schnitzer, B.; Bingham, E.L.; Harnden, C.E.; Hyder, D.M.; Ginsburg, D. Rearrangement of immunoglobulin and T-cell receptor genes in Hodgkin’s disease. Am. J. Pathol. 1988, 131, 331–338. [Google Scholar] [PubMed]
  26. Sundeen, J.; Lipford, E.; Uppenkamp, M.; Sussman, E.; Wahl, L.; Raffeld, M.; Cossman, J. Rearranged antigen receptor genes in Hodgkin’s disease. Blood 1987, 70, 96–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kamel, O.W.; Chang, P.P.; Hsu, F.J.; Dolezal, M.V.; Warnke, R.A.; van de Rijn, M. Clonal VDJ recombination of the immunoglobulin heavy chain gene by PCR in classical Hodgkin’s disease. Am. J. Clin. Pathol. 1995, 104, 419–423. [Google Scholar] [CrossRef]
  28. Manzanal, A.; Santon, A.; Oliva, H.; Bellas, C. Evaluation of clonal immunoglobulin heavy chain rearrangements in Hodgkin’s disease using the polymerase chain reaction (PCR). Histopathology 1995, 27, 21–25. [Google Scholar] [CrossRef] [PubMed]
  29. Manzanal, A.I.; Santon, A.; Acevedo, A.; Aguilera, B.; Oliva, H.; Bellas, C. Molecular analysis of the IgH gene in 212 cases of Hodgkin’s disease: Correlation of IgH clonality with the histologic and the immunocytochemical features. Mod. Pathol. 1997, 10, 679–685. [Google Scholar]
  30. van Dongen, J.J.; Langerak, A.W.; Bruggemann, M.; Evans, P.A.; Hummel, M.; Lavender, F.L.; Delabesse, E.; Davi, F.; Schuuring, E.; Garcia-Sanz, R.; et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: Report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia 2003, 17, 2257–2317. [Google Scholar] [CrossRef] [Green Version]
  31. Burack, W.R.; Laughlin, T.S.; Friedberg, J.W.; Spence, J.M.; Rothberg, P.G. PCR assays detect B-lymphocyte clonality in formalin-fixed, paraffin-embedded specimens of classical hodgkin lymphoma without microdissection. Am. J. Clin. Pathol. 2010, 134, 104–111. [Google Scholar] [CrossRef]
  32. Chute, D.J.; Cousar, J.B.; Mahadevan, M.S.; Siegrist, K.A.; Silverman, L.M.; Stoler, M.H. Detection of immunoglobulin heavy chain gene rearrangements in classic hodgkin lymphoma using commercially available BIOMED-2 primers. Diagn. Mol. Pathol. 2008, 17, 65–72. [Google Scholar] [CrossRef]
  33. Ghorbian, S.; Jahanzad, I.; Estiar, M.A.; Ziae, J.E.; Asvadi-Kermani, I.; Andalib, S.; Javadi, G.R.; Sakhinia, E. Molecular Analysis of IGH and Incomplete IGH D-J Clonality Gene Rearrangements in Hodgkin Lymphoma Malignancies. Clin. Lab. 2015, 61, 951–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ghorbian, S.; Jahanzad, I.; Javadi, G.R.; Sakhinia, E. Evaluation of IGK and IGL molecular gene rearrangements according to the BIOMED-2 protocols for clinical diagnosis of Hodgkin lymphoma. Hematology 2016, 21, 133–137. [Google Scholar] [CrossRef] [PubMed]
  35. Han, S.; Masaki, A.; Sakamoto, Y.; Takino, H.; Murase, T.; Iida, S.; Inagaki, H. Improved clonality detection in Hodgkin lymphoma using a semi-nested modification of the BIOMED-2 PCR assay for IGH and IGK rearrangements: A paraffin-embedded tissue study. Pathol. Int. 2018, 68, 287–293. [Google Scholar] [CrossRef] [PubMed]
  36. Hartmann, S.; Helling, A.; Doring, C.; Renne, C.; Hansmann, M.L. Clonality testing of malignant lymphomas with the BIOMED-2 primers in a large cohort of 1969 primary and consultant biopsies. Pathol. Res. Pract. 2013, 209, 495–502. [Google Scholar] [CrossRef]
  37. Hebeda, K.M.; Van Altena, M.C.; Rombout, P.; Van Krieken, J.H.; Groenen, P.J. PCR clonality detection in Hodgkin lymphoma. J. Hematop. 2009, 2, 34–41. [Google Scholar] [CrossRef] [Green Version]
  38. McClure, R.F.; Kaur, P.; Pagel, E.; Ouillette, P.D.; Holtegaard, C.E.; Treptow, C.L.; Kurtin, P.J. Validation of immunoglobulin gene rearrangement detection by PCR using commercially available BIOMED-2 primers. Leukemia 2006, 20, 176–179. [Google Scholar] [CrossRef]
  39. Tapia, G.; Sanz, C.; Mate, J.L.; Munoz-Marmol, A.M.; Ariza, A. Improved clonality detection in Hodgkin lymphoma using the BIOMED-2-based heavy and kappa chain assay: A paraffin-embedded tissue study. Histopathology 2012, 60, 768–773. [Google Scholar] [CrossRef]
  40. Evans, P.A.; Pott, C.; Groenen, P.J.; Salles, G.; Davi, F.; Berger, F.; Garcia, J.F.; van Krieken, J.H.; Pals, S.; Kluin, P.; et al. Significantly improved PCR-based clonality testing in B-cell malignancies by use of multiple immunoglobulin gene targets. Report of the BIOMED-2 Concerted Action BHM4-CT98-3936. Leukemia 2007, 21, 207–214. [Google Scholar] [CrossRef]
  41. Gameiro, P.; Sebastião, M.; Spetalen, S.; da Silva, M.G.; Cabeçadas, J. The added value of immunoglobulin Kappa light chain gene (IGK) rearrangement analysis in suspected B-cell lymphomas: Three illustrative cases. J. Hematop. 2012, 5, 45–56. [Google Scholar] [CrossRef]
  42. Aguilera, N.S.; Chen, J.; Bijwaard, K.E.; Director-Myska, A.E.; Barekman, C.L.; Millward, C.; Lichy, J.; Abbondanzo, S.L. Gene rearrangement and comparative genomic hybridization studies of classic Hodgkin lymphoma expressing T-cell antigens. Arch. Pathol. Lab. Med. 2006, 130, 1772–1779. [Google Scholar] [CrossRef]
  43. Deng, F.; Lü, G.; Li, G.; Yang, G. Hodgkin’s disease: Immunoglobulin heavy and light chain gene rearrangements revealed in single Hodgkin/Reed-Sternberg cells. Mol. Pathol. 1999, 52, 37–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Hummel, M.; Ziemann, K.; Lammert, H.; Pileri, S.; Sabattini, E.; Stein, H. Hodgkin’s disease with monoclonal and polyclonal populations of Reed-Sternberg cells. N. Engl. J. Med. 1995, 333, 901–906. [Google Scholar] [CrossRef] [PubMed]
  45. Izban, K.F.; Nawrocki, J.F.; Alkan, S.; Hsi, E.D. Monoclonal IgH gene rearrangement in microdissected nodules from nodular sclerosis Hodgkin disease. Am. J. Clin. Pathol. 1998, 110, 599–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Roth, J.; Daus, H.; Trumper, L.; Gause, A.; Salamon-Looijen, M.; Pfreundschuh, M. Detection of immunoglobulin heavy-chain gene rearrangement at the single-cell level in malignant lymphomas: No rearrangement is found in Hodgkin and Reed-Sternberg cells. Int. J. Cancer 1994, 57, 799–804. [Google Scholar] [CrossRef]
  47. Seitz, V.; Hummel, M.; Marafioti, T.; Anagnostopoulos, I.; Assaf, C.; Stein, H. Detection of clonal T-cell receptor gamma-chain gene rearrangements in Reed-Sternberg cells of classic Hodgkin disease. Blood 2000, 95, 3020–3024. [Google Scholar] [CrossRef]
  48. 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. [Google Scholar] [CrossRef]
  49. Brauninger, A.; Wacker, H.H.; Rajewsky, K.; Kuppers, R.; Hansmann, M.L. Typing the histogenetic origin of the tumor cells of lymphocyte-rich classical Hodgkin’s lymphoma in relation to tumor cells of classical and lymphocyte-predominance Hodgkin’s lymphoma. Cancer Res. 2003, 63, 1644–1651. [Google Scholar]
  50. Oki, Y.; Neelapu, S.S.; Fanale, M.; Kwak, L.W.; Fayad, L.; Rodriguez, M.A.; Wallace, M.; Klinger, M.; Carlton, V.; Kong, K.; et al. Detection of classical Hodgkin lymphoma specific sequence in peripheral blood using a next-generation sequencing approach. Br. J. Haematol. 2015, 169, 689–693. [Google Scholar] [CrossRef]
  51. van Bladel, D.A.G.; van den Brand, M.; Rijntjes, J.; Pamidimarri Naga, S.; Haacke, D.; Luijks, J.; Hebeda, K.M.; van Krieken, J.; Groenen, P.; Scheijen, B. Clonality assessment and detection of clonal diversity in classic Hodgkin lymphoma by next-generation sequencing of immunoglobulin gene rearrangements. Mod. Pathol. 2022, 35, 757–766. [Google Scholar] [CrossRef]
  52. Desch, A.K.; Hartung, K.; Botzen, A.; Brobeil, A.; Rummel, M.; Kurch, L.; Georgi, T.; Jox, T.; Bielack, S.; Burdach, S.; et al. Genotyping circulating tumor DNA of pediatric Hodgkin lymphoma. Leukemia 2020, 34, 151–166. [Google Scholar] [CrossRef]
  53. Muschen, M.; Rajewsky, K.; Brauninger, A.; Baur, A.S.; Oudejans, J.J.; Roers, A.; Hansmann, M.L.; Kuppers, R. Rare occurrence of classical Hodgkin’s disease as a T cell lymphoma. J. Exp. Med. 2000, 191, 387–394. [Google Scholar] [CrossRef] [PubMed]
  54. Venkataraman, G.; Song, J.Y.; Tzankov, A.; Dirnhofer, S.; Heinze, G.; Kohl, M.; Traverse-Glehen, A.; Eberle, F.C.; Hanson, J.C.; Raffeld, M.A.; et al. Aberrant T-cell antigen expression in classical Hodgkin lymphoma is associated with decreased event-free survival and overall survival. Blood 2013, 121, 1795–1804. [Google Scholar] [CrossRef] [PubMed]
  55. Bruggemann, M.; Kotrova, M.; Knecht, H.; Bartram, J.; Boudjogrha, M.; Bystry, V.; Fazio, G.; Fronkova, E.; Giraud, M.; Grioni, A.; et al. Standardized next-generation sequencing of immunoglobulin and T-cell receptor gene recombinations for MRD marker identification in acute lymphoblastic leukaemia; a EuroClonality-NGS validation study. Leukemia 2019, 33, 2241–2253. [Google Scholar] [CrossRef] [Green Version]
  56. Scheijen, B.; Meijers, R.W.J.; Rijntjes, J.; van der Klift, M.Y.; Möbs, M.; Steinhilber, J.; Reigl, T.; van den Brand, M.; Kotrová, M.; Ritter, J.M.; et al. Next-generation sequencing of immunoglobulin gene rearrangements for clonality assessment: A technical feasibility study by EuroClonality-NGS. Leukemia 2019, 33, 2227–2240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Nguyen, T.T.; Warnke, R.A.; Seo, K.; Rosenberg, S.A.; Arber, D.A. Rare presentation of classical Hodgkin lymphoma with a clonal T-cell receptor gene rearrangement in the tissue. Leuk. Lymphoma 2010, 51, 1356–1359. [Google Scholar] [CrossRef] [PubMed]
  58. Willenbrock, K.; Ichinohasama, R.; Kadin, M.E.; Miura, I.; Terui, T.; Meguro, K.; Fukuhara, O.; DeCoteau, J.F.; Hansmann, M.L. T-cell variant of classical Hodgkin’s lymphoma with nodal and cutaneous manifestations demonstrated by single-cell polymerase chain reaction. Lab. Investig. 2002, 82, 1103–1109. [Google Scholar] [CrossRef] [Green Version]
  59. Xie, Y.; Jaffe, E.S. How I Diagnose Angioimmunoblastic T-Cell Lymphoma. Am. J. Clin. Pathol. 2021, 156, 1–14. [Google Scholar] [CrossRef]
  60. Moroch, J.; Copie-Bergman, C.; de Leval, L.; Plonquet, A.; Martin-Garcia, N.; Delfau-Larue, M.H.; Molinier-Frenkel, V.; Belhadj, K.; Haioun, C.; Audouin, J.; et al. Follicular peripheral T-cell lymphoma expands the spectrum of classical Hodgkin lymphoma mimics. Am. J. Surg. Pathol. 2012, 36, 1636–1646. [Google Scholar] [CrossRef]
  61. Obermann, E.C.; Mueller, N.; Rufle, A.; Menter, T.; Mueller-Garamvoelgyi, E.; Cathomas, G.; Dirnhofer, S.; Tzankov, A. Clonal relationship of classical hodgkin lymphoma and its recurrences. Clin. Cancer Res. 2011, 17, 5268–5274. [Google Scholar] [CrossRef] [Green Version]
  62. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef]
  63. Alcoceba, M.; García-Álvarez, M.; Chillón, M.C.; Jiménez, C.; Medina, A.; Antón, A.; Blanco, O.; Díaz, L.G.; Tamayo, P.; González-Calle, V.; et al. Liquid biopsy: A non-invasive approach for Hodgkin lymphoma genotyping. Br. J. Haematol. 2021, 195, 542–551. [Google Scholar] [CrossRef] [PubMed]
  64. Bessi, L.; Viailly, P.J.; Bohers, E.; Ruminy, P.; Maingonnat, C.; Bertrand, P.; Vasseur, N.; Beaussire, L.; Cornic, M.; Etancelin, P.; et al. Somatic mutations of cell-free circulating DNA detected by targeted next-generation sequencing and digital droplet PCR in classical Hodgkin lymphoma. Leuk. Lymphoma 2019, 60, 498–502. [Google Scholar] [CrossRef] [PubMed]
  65. Camus, V.; Stamatoullas, A.; Mareschal, S.; Viailly, P.J.; Sarafan-Vasseur, N.; Bohers, E.; Dubois, S.; Picquenot, J.M.; Ruminy, P.; Maingonnat, C.; et al. Detection and prognostic value of recurrent exportin 1 mutations in tumor and cell-free circulating DNA of patients with classical Hodgkin lymphoma. Haematologica 2016, 101, 1094–1101. [Google Scholar] [CrossRef] [PubMed]
  66. 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] [Green Version]
  67. 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] [Green Version]
  68. Eijkelenboom, A.; Kamping, E.J.; Kastner-van Raaij, A.W.; Hendriks-Cornelissen, S.J.; Neveling, K.; Kuiper, R.P.; Hoischen, A.; Nelen, M.R.; Ligtenberg, M.J.; Tops, B.B. Reliable Next-Generation Sequencing of Formalin-Fixed, Paraffin-Embedded Tissue Using Single Molecule Tags. J. Mol. Diagn. 2016, 18, 851–863. [Google Scholar] [CrossRef] [Green Version]
  69. Kou, R.; Lam, H.; Duan, H.; Ye, L.; Jongkam, N.; Chen, W.; Zhang, S.; Li, S. Benefits and Challenges with Applying Unique Molecular Identifiers in Next Generation Sequencing to Detect Low Frequency Mutations. PLoS ONE 2016, 11, e0146638. [Google Scholar] [CrossRef] [Green Version]
  70. Lin, C.; Liu, X.; Zheng, B.; Ke, R.; Tzeng, C.M. Liquid Biopsy, ctDNA Diagnosis through NGS. Life 2021, 11, 890. [Google Scholar] [CrossRef]
  71. Lui, Y.Y.; Chik, K.W.; Chiu, R.W.; Ho, C.Y.; Lam, C.W.; Lo, Y.M. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin. Chem. 2002, 48, 421–427. [Google Scholar] [CrossRef] [Green Version]
  72. Chan, H.T.; Chin, Y.M.; Nakamura, Y.; Low, S.K. Clonal Hematopoiesis in Liquid Biopsy: From Biological Noise to Valuable Clinical Implications. Cancers 2020, 12, 2277. [Google Scholar] [CrossRef]
  73. Jaiswal, S.; Fontanillas, P.; Flannick, J.; Manning, A.; Grauman, P.V.; Mar, B.G.; Lindsley, R.C.; Mermel, C.H.; Burtt, N.; Chavez, A.; et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 2014, 371, 2488–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Buscarlet, M.; Provost, S.; Zada, Y.F.; Barhdadi, A.; Bourgoin, V.; Lépine, G.; Mollica, L.; Szuber, N.; Dubé, M.P.; Busque, L. DNMT3A and TET2 dominate clonal hematopoiesis and demonstrate benign phenotypes and different genetic predispositions. Blood 2017, 130, 753–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Genovese, G.; Kähler, A.K.; Handsaker, R.E.; Lindberg, J.; Rose, S.A.; Bakhoum, S.F.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Xie, M.; Lu, C.; Wang, J.; McLellan, M.D.; Johnson, K.J.; Wendl, M.C.; McMichael, J.F.; Schmidt, H.K.; Yellapantula, V.; Miller, C.A.; et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 2014, 20, 1472–1478. [Google Scholar] [CrossRef] [PubMed]
  77. Venanzi, A.; Marra, A.; Schiavoni, G.; Milner, S.G.; Limongello, R.; Santi, A.; Pettirossi, V.; Ultimo, S.; Tasselli, L.; Pucciarini, A.; et al. Dissecting Clonal Hematopoiesis in Tissues of Classical Hodgkin Lymphoma Patients. Blood Cancer Discov. 2021, 2, 216–225. [Google Scholar] [CrossRef]
  78. Vandenberghe, P.; Wlodarska, I.; Tousseyn, T.; Dehaspe, L.; Dierickx, D.; Verheecke, M.; Uyttebroeck, A.; Bechter, O.; Delforge, M.; Vandecaveye, V.; et al. Non-invasive detection of genomic imbalances in Hodgkin/Reed-Sternberg cells in early and advanced stage Hodgkin’s lymphoma by sequencing of circulating cell-free DNA: A technical proof-of-principle study. Lancet Haematol. 2015, 2, e55–e65. [Google Scholar] [CrossRef] [Green Version]
  79. Liang, W.S.; Vergilio, J.A.; Salhia, B.; Huang, H.J.; Oki, Y.; Garrido-Laguna, I.; Park, H.; Westin, J.R.; Meric-Bernstam, F.; Fabrizio, D.; et al. Comprehensive Genomic Profiling of Hodgkin Lymphoma Reveals Recurrently Mutated Genes and Increased Mutation Burden. Oncologist 2019, 24, 219–228. [Google Scholar] [CrossRef] [Green Version]
  80. Hussaini, M.O.; Srivastava, J.; Lee, L.W.; Nishihori, T.; Shah, B.D.; Alsina, M.; Pinilla-Ibarz, J.; Shain, K.H. Assessment of Clonotypic Rearrangements and Minimal Residual Disease in Lymphoid Malignancies: A Large Cancer Center Experience Using clonoSEQ. Arch. Pathol. Lab. Med. 2021, 146, 485–493. [Google Scholar] [CrossRef]
  81. Monter, A.; Nomdedeu, J.F. ClonoSEQ assay for the detection of lymphoid malignancies. Expert Rev. Mol. Diagn. 2019, 19, 571–578. [Google Scholar] [CrossRef]
  82. Sobesky, S.; Mammadova, L.; Cirillo, M.; Drees, E.E.E.; Mattlener, J.; Dörr, H.; Altmüller, J.; Shi, Z.; Bröckelmann, P.J.; Weiss, J.; et al. In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. Med (N. Y.) 2021, 2, 1171–1193. [Google Scholar] [CrossRef]
  83. Mussolin, L.; Burnelli, R.; Pillon, M.; Carraro, E.; Farruggia, P.; Todesco, A.; Mascarin, M.; Rosolen, A. Plasma cell-free DNA in paediatric lymphomas. J. Cancer 2013, 4, 323–329. [Google Scholar] [CrossRef] [PubMed]
  84. Primerano, S.; Burnelli, R.; Carraro, E.; Pillon, M.; Elia, C.; Farruggia, P.; Sala, A.; Vinti, L.; Buffardi, S.; Basso, G.; et al. Kinetics of Circulating Plasma Cell-Free DNA in Paediatric Classical Hodgkin Lymphoma. J. Cancer 2016, 7, 364–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Shi, Y.; Su, H.; Song, Y.; Jiang, W.; Sun, X.; Qian, W.; Zhang, W.; Gao, Y.; Jin, Z.; Zhou, J.; et al. Circulating tumor DNA predicts response in Chinese patients with relapsed or refractory classical hodgkin lymphoma treated with sintilimab. EBioMedicine 2020, 54, 102731. [Google Scholar] [CrossRef] [PubMed]
  86. Faham, M.; Zheng, J.; Moorhead, M.; Carlton, V.E.; Stow, P.; Coustan-Smith, E.; Pui, C.H.; Campana, D. Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia. Blood 2012, 120, 5173–5180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Newman, A.M.; Bratman, S.V.; To, J.; Wynne, J.F.; Eclov, N.C.; Modlin, L.A.; Liu, C.L.; Neal, J.W.; Wakelee, H.A.; Merritt, R.E.; et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 2014, 20, 548–554. [Google Scholar] [CrossRef]
  88. Kurtz, D.M. Bringing circulating tumor DNA to the clinic in Hodgkin lymphoma. Haematologica 2021, 106, 5–6. [Google Scholar] [CrossRef]
  89. Stewart, J.P.; Gazdova, J.; Darzentas, N.; Wren, D.; Proszek, P.; Fazio, G.; Songia, S.; Alcoceba, M.; Sarasquete, M.E.; Villarese, P.; et al. Validation of the EuroClonality-NGS DNA capture panel as an integrated genomic tool for lymphoproliferative disorders. Blood Adv. 2021, 5, 3188–3198. [Google Scholar] [CrossRef]
  90. Murray, P.G.; Young, L.S. An etiological role for the Epstein-Barr virus in the pathogenesis of classical Hodgkin lymphoma. Blood 2019, 134, 591–596. [Google Scholar] [CrossRef]
  91. Reichel, J.; Chadburn, A.; Rubinstein, P.G.; Giulino-Roth, L.; Tam, W.; Liu, Y.; Gaiolla, R.; Eng, K.; Brody, J.; Inghirami, G.; et al. Flow sorting and exome sequencing reveal the oncogenome of primary Hodgkin and Reed-Sternberg cells. Blood 2015, 125, 1061–1072. [Google Scholar] [CrossRef] [Green Version]
  92. Tiacci, E.; Ladewig, E.; Schiavoni, G.; Penson, A.; Fortini, E.; Pettirossi, V.; Wang, Y.; Rosseto, A.; Venanzi, A.; Vlasevska, S.; et al. Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma. Blood 2018, 131, 2454–2465. [Google Scholar] [CrossRef] [Green Version]
  93. Wienand, K.; Chapuy, B.; Stewart, C.; Dunford, A.J.; Wu, D.; Kim, J.; Kamburov, A.; Wood, T.R.; Cader, F.Z.; Ducar, M.D.; et al. Genomic analyses of flow-sorted Hodgkin Reed-Sternberg cells reveal complementary mechanisms of immune evasion. Blood Adv. 2019, 3, 4065–4080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tilly, H.; Bastard, C.; Delastre, T.; Duval, C.; Bizet, M.; Lenormand, B.; Daucé, J.P.; Monconduit, M.; Piguet, H. Cytogenetic studies in untreated Hodgkin’s disease. Blood 1991, 77, 1298–1304. [Google Scholar] [CrossRef] [PubMed]
  95. Hartmann, S.; Martin-Subero, J.I.; Gesk, S.; Hüsken, J.; Giefing, M.; Nagel, I.; Riemke, J.; Chott, A.; Klapper, W.; Parrens, M.; et al. Detection of genomic imbalances in microdissected Hodgkin and Reed-Sternberg cells of classical Hodgkin’s lymphoma by array-based comparative genomic hybridization. Haematologica 2008, 93, 1318–1326. [Google Scholar] [CrossRef]
  96. Re, D.; Zander, T.; Diehl, V.; Wolf, J. Genetic instability in Hodgkin’s lymphoma. Ann. Oncol. 2002, 13 (Suppl. 1), 19–22. [Google Scholar] [CrossRef]
  97. Martín-Subero, J.I.; Klapper, W.; Sotnikova, A.; Callet-Bauchu, E.; Harder, L.; Bastard, C.; Schmitz, R.; Grohmann, S.; Höppner, J.; Riemke, J.; et al. Chromosomal breakpoints affecting immunoglobulin loci are recurrent in Hodgkin and Reed-Sternberg cells of classical Hodgkin lymphoma. Cancer Res. 2006, 66, 10332–10338. [Google Scholar] [CrossRef] [Green Version]
  98. Chui, D.T.; Hammond, D.; Baird, M.; Shield, L.; Jackson, R.; Jarrett, R.F. Classical Hodgkin lymphoma is associated with frequent gains of 17q. Genes Chromosomes Cancer 2003, 38, 126–136. [Google Scholar] [CrossRef] [PubMed]
  99. Joos, S.; Menz, C.K.; Wrobel, G.; Siebert, R.; Gesk, S.; Ohl, S.; Mechtersheimer, G.; Trümper, L.; Möller, P.; Lichter, P.; et al. Classical Hodgkin lymphoma is characterized by recurrent copy number gains of the short arm of chromosome 2. Blood 2002, 99, 1381–1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Steidl, C.; Telenius, A.; Shah, S.P.; Farinha, P.; Barclay, L.; Boyle, M.; Connors, J.M.; Horsman, D.E.; Gascoyne, R.D. Genome-wide copy number analysis of Hodgkin Reed-Sternberg cells identifies recurrent imbalances with correlations to treatment outcome. Blood 2010, 116, 418–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Roemer, M.G.; Advani, R.H.; Ligon, A.H.; Natkunam, Y.; Redd, R.A.; Homer, H.; Connelly, C.F.; Sun, H.H.; Daadi, S.E.; Freeman, G.J.; et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J. Clin. Oncol. 2016, 34, 2690–2697. [Google Scholar] [CrossRef] [Green Version]
  102. Buedts, L.; Wlodarska, I.; Finalet-Ferreiro, J.; Gheysens, O.; Dehaspe, L.; Tousseyn, T.; Fornecker, L.M.; Lazarovici, J.; Casasnovas, R.O.; Gac, A.C.; et al. The landscape of copy number variations in classical Hodgkin lymphoma: A joint KU Leuven and LYSA study on cell-free DNA. Blood Adv. 2021, 5, 1991–2002. [Google Scholar] [CrossRef]
  103. Healy, J.A.; Nugent, A.; Rempel, R.E.; Moffitt, A.B.; Davis, N.S.; Jiang, X.; Shingleton, J.R.; Zhang, J.; Love, C.; Datta, J.; et al. GNA13 loss in germinal center B cells leads to impaired apoptosis and promotes lymphoma in vivo. Blood 2016, 127, 2723–2731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. O’Hayre, M.; Inoue, A.; Kufareva, I.; Wang, Z.; Mikelis, C.M.; Drummond, R.A.; Avino, S.; Finkel, K.; Kalim, K.W.; DiPasquale, G.; et al. Inactivating mutations in GNA13 and RHOA in Burkitt’s lymphoma and diffuse large B-cell lymphoma: A tumor suppressor function for the Gα13/RhoA axis in B cells. Oncogene 2016, 35, 3771–3780. [Google Scholar] [CrossRef] [PubMed]
  105. Xia, Z.; Zhang, X.; Liu, P.; Zhang, R.; Huang, Z.; Li, D.; Xiao, X.; Wu, M.; Ning, N.; Zhang, Q.; et al. GNA13 regulates BCL2 expression and the sensitivity of GCB-DLBCL cells to BCL2 inhibitors in a palmitoylation-dependent manner. Cell Death Dis. 2021, 12, 54. [Google Scholar] [CrossRef] [PubMed]
  106. Shen, J.; Ju, Z.; Zhao, W.; Wang, L.; Peng, Y.; Ge, Z.; Nagel, Z.D.; Zou, J.; Wang, C.; Kapoor, P.; et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat. Med. 2018, 24, 556–562. [Google Scholar] [CrossRef] [PubMed]
  107. Maggio, E.M.; Stekelenburg, E.; Van den Berg, A.; Poppema, S. TP53 gene mutations in Hodgkin lymphoma are infrequent and not associated with absence of Epstein-Barr virus. Int. J. Cancer 2001, 94, 60–66. [Google Scholar] [CrossRef]
  108. Chen, W.G.; Chen, Y.Y.; Kamel, O.W.; Koo, C.H.; Weiss, L.M. p53 mutations in Hodgkin’s disease. Lab. Investig. 1996, 75, 519–527. [Google Scholar]
  109. Mata, E.; Fernández, S.; Astudillo, A.; Fernández, R.; García-Cosío, M.; Sánchez-Beato, M.; Provencio, M.; Estévez, M.; Montalbán, C.; Piris, M.A.; et al. Genomic analyses of microdissected Hodgkin and Reed-Sternberg cells: Mutations in epigenetic regulators and p53 are frequent in refractory classic Hodgkin lymphoma. Blood Cancer J. 2019, 9, 34. [Google Scholar] [CrossRef]
  110. Elenitoba-Johnson, K.S.; Medeiros, L.J.; Khorsand, J.; King, T.C. P53 expression in Reed-Sternberg cells does not correlate with gene mutations in Hodgkin’s disease. Am. J. Clin. Pathol. 1996, 106, 728–738. [Google Scholar] [CrossRef] [Green Version]
  111. Liu, Y.; Sattarzadeh, A.; Diepstra, A.; Visser, L.; van den Berg, A. The microenvironment in classical Hodgkin lymphoma: An actively shaped and essential tumor component. Semin. Cancer Biol. 2014, 24, 15–22. [Google Scholar] [CrossRef]
  112. Shannon-Lowe, C.; Rickinson, A.B.; Bell, A.I. Epstein-Barr virus-associated lymphomas. Philos. Trans. R. Soc. B Biol. Sci. 2017, 372, 20160271. [Google Scholar] [CrossRef]
  113. Vrzalikova, K.; Pugh, M.; Mundo, L.; Murray, P. The contribution of ebv to the pathogenesis of classical hodgkin lymphoma. Ann. Lymphoma 2021, 5, 30. [Google Scholar] [CrossRef]
  114. Yin, H.; Qu, J.; Peng, Q.; Gan, R. Molecular mechanisms of EBV-driven cell cycle progression and oncogenesis. Med. Microbiol. Immunol. 2019, 208, 573–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Chen, S.Y.; Lu, J.; Shih, Y.C.; Tsai, C.H. Epstein-Barr virus latent membrane protein 2A regulates c-Jun protein through extracellular signal-regulated kinase. J. Virol. 2002, 76, 9556–9561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Portis, T.; Longnecker, R. Epstein-Barr virus (EBV) LMP2A alters normal transcriptional regulation following B-cell receptor activation. Virology 2004, 318, 524–533. [Google Scholar] [CrossRef] [Green Version]
  117. Luo, Y.; Liu, Y.; Wang, C.; Gan, R. Signaling pathways of EBV-induced oncogenesis. Cancer Cell Int. 2021, 21, 93. [Google Scholar] [CrossRef]
  118. Vrzalikova, K.; Sunmonu, T.; Reynolds, G.; Murray, P. Contribution of Epstein(-)Barr Virus Latent Proteins to the Pathogenesis of Classical Hodgkin Lymphoma. Pathogens 2018, 7, 59. [Google Scholar] [CrossRef] [Green Version]
  119. Liu, T.Y.; Wu, S.J.; Huang, M.H.; Lo, F.Y.; Tsai, M.H.; Tsai, C.H.; Hsu, S.M.; Lin, C.W. EBV-positive Hodgkin lymphoma is associated with suppression of p21cip1/waf1 and a worse prognosis. Mol. Cancer 2010, 9, 32. [Google Scholar] [CrossRef] [Green Version]
  120. Iwakiri, D.; Takada, K. Role of EBERs in the pathogenesis of EBV infection. Adv. Cancer Res. 2010, 107, 119–136. [Google Scholar] [CrossRef]
  121. De Re, V.; Caggiari, L.; De Zorzi, M.; Fanotto, V.; Miolo, G.; Puglisi, F.; Cannizzaro, R.; Canzonieri, V.; Steffan, A.; Farruggia, P.; et al. Epstein-Barr virus BART microRNAs in EBV- associated Hodgkin lymphoma and gastric cancer. Infect. Agent Cancer 2020, 15, 42. [Google Scholar] [CrossRef]
  122. Lake, A.; Shield, L.A.; Cordano, P.; Chui, D.T.; Osborne, J.; Crae, S.; Wilson, K.S.; Tosi, S.; Knight, S.J.; Gesk, S.; et al. Mutations of NFKBIA, encoding IkappaB alpha, are a recurrent finding in classical Hodgkin lymphoma but are not a unifying feature of non-EBV-associated cases. Int. J. Cancer 2009, 125, 1334–1342. [Google Scholar] [CrossRef] [Green Version]
  123. Montgomery, N.D.; Coward, W.B.t.; Johnson, S.; Yuan, J.; Gulley, M.L.; Mathews, S.P.; Kaiser-Rogers, K.; Rao, K.W.; Sanger, W.G.; Sanmann, J.N.; et al. Karyotypic abnormalities associated with Epstein-Barr virus status in classical Hodgkin lymphoma. Cancer Genet. 2016, 209, 408–416. [Google Scholar] [CrossRef] [PubMed]
  124. Schmitz, R.; Stanelle, J.; Hansmann, M.L.; Küppers, R. Pathogenesis of classical and lymphocyte-predominant Hodgkin lymphoma. Annu. Rev. Pathol. 2009, 4, 151–174. [Google Scholar] [CrossRef] [PubMed]
  125. Renné, C.; Hinsch, N.; Willenbrock, K.; Fuchs, M.; Klapper, W.; Engert, A.; Küppers, R.; Hansmann, M.L.; Bräuninger, A. The aberrant coexpression of several receptor tyrosine kinases is largely restricted to EBV-negative cases of classical Hodgkin’s lymphoma. Int. J. Cancer 2007, 120, 2504–2509. [Google Scholar] [CrossRef] [PubMed]
  126. Schmitz, R.; Hansmann, M.L.; Bohle, V.; Martin-Subero, J.I.; Hartmann, S.; Mechtersheimer, G.; Klapper, W.; Vater, I.; Giefing, M.; Gesk, S.; et al. TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. J. Exp. Med. 2009, 206, 981–989. [Google Scholar] [CrossRef] [Green Version]
  127. Aldinucci, D.; Celegato, M.; Casagrande, N. Microenvironmental interactions in classical Hodgkin lymphoma and their role in promoting tumor growth, immune escape and drug resistance. Cancer Lett. 2016, 380, 243–252. [Google Scholar] [CrossRef]
  128. Re, D.; Kuppers, R.; Diehl, V. Molecular pathogenesis of Hodgkin’s lymphoma. J. Clin. Oncol. 2005, 23, 6379–6386. [Google Scholar] [CrossRef] [Green Version]
  129. Wein, F.; Küppers, R. The role of T cells in the microenvironment of Hodgkin lymphoma. J. Leukoc. Biol. 2016, 99, 45–50. [Google Scholar] [CrossRef] [Green Version]
  130. Aoki, T.; Chong, L.C.; Takata, K.; Milne, K.; Hav, M.; Colombo, A.; Chavez, E.A.; Nissen, M.; Wang, X.; Miyata-Takata, T.; et al. Single-Cell Transcriptome Analysis Reveals Disease-Defining T-cell Subsets in the Tumor Microenvironment of Classic Hodgkin Lymphoma. Cancer Discov. 2020, 10, 406–421. [Google Scholar] [CrossRef] [Green Version]
  131. Aoki, T.; Chong, L.C.; Takata, K.; Milne, K.; Marshall, A.; Chavez, E.A.; Miyata-Takata, T.; Ben-Neriah, S.; Unrau, D.; Telenius, A.; et al. Single-cell profiling reveals the importance of CXCL13/CXCR5 axis biology in lymphocyte-rich classic Hodgkin lymphoma. Proc. Natl. Acad. Sci. USA 2021, 118, e2105822118. [Google Scholar] [CrossRef]
  132. Cader, F.Z.; Schackmann, R.C.J.; Hu, X.; Wienand, K.; Redd, R.; Chapuy, B.; Ouyang, J.; Paul, N.; Gjini, E.; Lipschitz, M.; et al. Mass cytometry of Hodgkin lymphoma reveals a CD4(+) regulatory T-cell-rich and exhausted T-effector microenvironment. Blood 2018, 132, 825–836. [Google Scholar] [CrossRef]
  133. Carbone, A.; Gloghini, A.; Gruss, H.J.; Pinto, A. CD40 ligand is constitutively expressed in a subset of T cell lymphomas and on the microenvironmental reactive T cells of follicular lymphomas and Hodgkin’s disease. Am. J. Pathol. 1995, 147, 912–922. [Google Scholar] [PubMed]
  134. Steidl, C.; Connors, J.M.; Gascoyne, R.D. Molecular pathogenesis of Hodgkin’s lymphoma: Increasing evidence of the importance of the microenvironment. J. Clin. Oncol. 2011, 29, 1812–1826. [Google Scholar] [CrossRef] [PubMed]
  135. Steinhilber, J.; Mederake, M.; Bonzheim, I.; Serinsöz-Linke, E.; Müller, I.; Fallier-Becker, P.; Lemonnier, F.; Gaulard, P.; Fend, F.; Quintanilla-Martinez, L. The pathological features of angioimmunoblastic T-cell lymphomas with IDH2(R172) mutations. Mod. Pathol. 2019, 32, 1123–1134. [Google Scholar] [CrossRef] [PubMed]
  136. Kujala, J.; Hartikainen, J.M.; Tengström, M.; Sironen, R.; Auvinen, P.; Kosma, V.M.; Mannermaa, A. Circulating Cell-Free DNA Reflects the Clonal Evolution of Breast Cancer Tumors. Cancers 2022, 14, 1332. [Google Scholar] [CrossRef]
  137. Imamura, F.; Uchida, J.; Kukita, Y.; Kumagai, T.; Nishino, K.; Inoue, T.; Kimura, M.; Oba, S.; Kato, K. Monitoring of treatment responses and clonal evolution of tumor cells by circulating tumor DNA of heterogeneous mutant EGFR genes in lung cancer. Lung Cancer 2016, 94, 68–73. [Google Scholar] [CrossRef] [PubMed]
  138. Siravegna, G.; Mussolin, B.; Buscarino, M.; Corti, G.; Cassingena, A.; Crisafulli, G.; Ponzetti, A.; Cremolini, C.; Amatu, A.; Lauricella, C.; et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med. 2015, 21, 795–801. [Google Scholar] [CrossRef] [Green Version]
  139. Perdigones, N.; Murtaza, M. Capturing tumor heterogeneity and clonal evolution in solid cancers using circulating tumor DNA analysis. Pharmacol. Ther. 2017, 174, 22–26. [Google Scholar] [CrossRef]
  140. Scherer, F.; Kurtz, D.M.; Newman, A.M.; Stehr, H.; Craig, A.F.; Esfahani, M.S.; Lovejoy, A.F.; Chabon, J.J.; Klass, D.M.; Liu, C.L.; et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci. Transl. Med. 2016, 8, 364ra155. [Google Scholar] [CrossRef] [Green Version]
  141. Rossi, D.; Diop, F.; Spaccarotella, E.; Monti, S.; Zanni, M.; Rasi, S.; Deambrogi, C.; Spina, V.; Bruscaggin, A.; Favini, C.; et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood 2017, 129, 1947–1957. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of immunoglobulin and T-cell receptor V(D)J gene rearrangements. (a) Immunoglobulin (IG) gene rearrangements generate a B-cell receptor, where IG heavy chain (IGH) genes are represented in blue, while IG kappa (IGK) and IG lambda (IGL) light chain genes are indicated in green. In this figure, only (potentially) productive IGH and IGK gene rearrangements are shown. In case of a non-productive IGK rearrangement, the kappa deleting element will rearrange with an upstream IGK-V gene or the Intron-RSS sequence (not shown in this figure). If both alleles result in a non-productive IGK light chain, the IGL light chain will rearrange, according to a similar process as IGK (not shown in this figure). Please note that the structure of the IGL chain is slightly different compared to the IGK chain shown here, since each IGL constant region is preceded by a single IGL-J gene. (b) V(D)J gene rearrangements of alpha/beta T-cell receptor. TR alpha (TRA) chain is represented in yellow, TR beta (TRB) chain in orange. Gamma/delta T-cell receptors are generated similarly, where TR gamma (TRG) chain undergoes VJ rearrangement similar to TRA, while TR delta (TRD) chain is generated by VDJ rearrangement, like TRB. Please note that the TRD gene cluster is located between the TRA-V and TRA-J gene clusters, as indicated with an asterisk. This TRD gene cluster will be deleted upon TRA gene rearrangements. For both IG and TCR, dark shades represent the constant regions, while the lighter shades correspond to variable regions, which are composed of rearranged V, (D) and J genes.
Figure 1. Schematic overview of immunoglobulin and T-cell receptor V(D)J gene rearrangements. (a) Immunoglobulin (IG) gene rearrangements generate a B-cell receptor, where IG heavy chain (IGH) genes are represented in blue, while IG kappa (IGK) and IG lambda (IGL) light chain genes are indicated in green. In this figure, only (potentially) productive IGH and IGK gene rearrangements are shown. In case of a non-productive IGK rearrangement, the kappa deleting element will rearrange with an upstream IGK-V gene or the Intron-RSS sequence (not shown in this figure). If both alleles result in a non-productive IGK light chain, the IGL light chain will rearrange, according to a similar process as IGK (not shown in this figure). Please note that the structure of the IGL chain is slightly different compared to the IGK chain shown here, since each IGL constant region is preceded by a single IGL-J gene. (b) V(D)J gene rearrangements of alpha/beta T-cell receptor. TR alpha (TRA) chain is represented in yellow, TR beta (TRB) chain in orange. Gamma/delta T-cell receptors are generated similarly, where TR gamma (TRG) chain undergoes VJ rearrangement similar to TRA, while TR delta (TRD) chain is generated by VDJ rearrangement, like TRB. Please note that the TRD gene cluster is located between the TRA-V and TRA-J gene clusters, as indicated with an asterisk. This TRD gene cluster will be deleted upon TRA gene rearrangements. For both IG and TCR, dark shades represent the constant regions, while the lighter shades correspond to variable regions, which are composed of rearranged V, (D) and J genes.
Cancers 14 03222 g001
Figure 2. Schematic overview of the pathways affected by genetic alterations in classical Hodgkin lymphoma. The main affected pathways in the pathogenesis of cHL involve JAK/STAT, NF-κB, PI3K/AKT and immune evasion, caused by inactivating mutations (in red), activating mutations (in green), translocations and overexpression due to gene amplifications. Nuclear–cytoplasmic (e.g., XPO1) transport and epigenetic regulation (e.g., ARID1A) may be disturbed in HRS cells, contributing to oncogenesis.
Figure 2. Schematic overview of the pathways affected by genetic alterations in classical Hodgkin lymphoma. The main affected pathways in the pathogenesis of cHL involve JAK/STAT, NF-κB, PI3K/AKT and immune evasion, caused by inactivating mutations (in red), activating mutations (in green), translocations and overexpression due to gene amplifications. Nuclear–cytoplasmic (e.g., XPO1) transport and epigenetic regulation (e.g., ARID1A) may be disturbed in HRS cells, contributing to oncogenesis.
Cancers 14 03222 g002
Figure 3. Schematic overview of EBV-associated gene products in the pathogenesis of classical Hodgkin lymphoma. EBV-associated transcripts and proteins are able to activate pathways and cellular processes that are recurrently affected by mutations in EBV-negative HRS cells. These alterations thereby play an important role in the pathogenesis and survival of EBV-positive HRS cells.
Figure 3. Schematic overview of EBV-associated gene products in the pathogenesis of classical Hodgkin lymphoma. EBV-associated transcripts and proteins are able to activate pathways and cellular processes that are recurrently affected by mutations in EBV-negative HRS cells. These alterations thereby play an important role in the pathogenesis and survival of EBV-positive HRS cells.
Cancers 14 03222 g003
Table 1. Overview of clonality assessment in classical Hodgkin lymphoma by the analysis of IG and TR gene.
Table 1. Overview of clonality assessment in classical Hodgkin lymphoma by the analysis of IG and TR gene.
IG Gene Rearrangements
Study CohortsClone Detection (IG)
Clonality AssayStudies (n)Samples (n)MeanRangeReferences
Southern blot *88–3945%0–87%[19,20,21,22,23,24,25,26]
PCR-based *332–21230%23–44%[27,28,29]
BIOMED-2 *812–5857%26–79%[31,32,33,34,35,36,37,38,39]
NGS-based
Tissue gDNA *216–1772%56–88%[50,51]
cfDNA29–7264%38–89%[50,52]
Enriched HRS cells #113–2562%0–100%[17,18,26,42,43,44,45,46,47,48,49]
TR Gene Rearrangements
Study cohortsClone detection (TR)
Clonality assayStudies (n)Samples (n)MeanRangeReferences
Southern blot *88–3915%0–68%[19,20,21,22,23,24,25,26]
BIOMED-2 *15817%NA[36]
Enriched HRS cells #43–1928%11–50%[42,47,53,54]
* Whole tumor genomic DNA (gDNA). # Multiple assay types used. cfDNA: cell-free DNA.
Table 2. Overview of recurrent genetic aberrations in classical Hodgkin lymphoma and their effects.
Table 2. Overview of recurrent genetic aberrations in classical Hodgkin lymphoma and their effects.
GeneGenetic Aberration(s) *Pathways and Biological Processes
PD-L1 and PD-L2CNAs (9p24 gain)Immune evasion
B2M, HLA-A/BInactivating mutations
CIITATranslocations
JAK2CNAs (9p24 gain)JAK/STAT signaling
STAT6Activating mutations
SOCS1Inactivating mutations
CSFR2BActivating mutations
PTPN1Inactivating mutations
TNFAIP3Inactivating mutations and deletionsNF-κB pathway
IKBKBActivating mutations
NFKBIA, NFKBIEInactivating mutations
BIRC3CNAs (11q loss)
RELCNAs (2p gain)
GNA13Inactivating mutationsPI3K/AKT pathway
ITPKBInactivating mutations
RBM38Inactivating mutations
PIK3CAActivating mutations
XPO1Activating mutationsNuclear–cytoplasmic transport
TP53Inactivating mutationsGenomic stability
ATMInactivating mutations
ARID1AInactivating mutationsEpigenetic regulation
KMT2CInactivating mutations
KMT2DInactivating mutations
* Activating mutations target oncogenes, while inactivating mutations target tumor suppressor genes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

van Bladel, D.A.G.; Stevens, W.B.C.; van den Brand, M.; Kroeze, L.I.; Groenen, P.J.T.A.; van Krieken, J.H.J.M.; Hebeda, K.M.; Scheijen, B. Novel Approaches in Molecular Characterization of Classical Hodgkin Lymphoma. Cancers 2022, 14, 3222. https://doi.org/10.3390/cancers14133222

AMA Style

van Bladel DAG, Stevens WBC, van den Brand M, Kroeze LI, Groenen PJTA, van Krieken JHJM, Hebeda KM, Scheijen B. Novel Approaches in Molecular Characterization of Classical Hodgkin Lymphoma. Cancers. 2022; 14(13):3222. https://doi.org/10.3390/cancers14133222

Chicago/Turabian Style

van Bladel, Diede A. G., Wendy B. C. Stevens, Michiel van den Brand, Leonie I. Kroeze, Patricia J. T. A. Groenen, J. Han J. M. van Krieken, Konnie M. Hebeda, and Blanca Scheijen. 2022. "Novel Approaches in Molecular Characterization of Classical Hodgkin Lymphoma" Cancers 14, no. 13: 3222. https://doi.org/10.3390/cancers14133222

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