Next Article in Journal
Single-Cell Analysis
Next Article in Special Issue
Leukocytes, Systemic Inflammation and Immunopathology in Acute-on-Chronic Liver Failure
Previous Article in Journal
Investigating the Role of Methylation in Silencing of VDR Gene Expression in Normal Cells during Hematopoiesis and in Their Leukemic Counterparts
Previous Article in Special Issue
Infection and Activation of B Cells by Theiler’s Murine Encephalomyelitis Virus (TMEV) Leads to Autoantibody Production in an Infectious Model of Multiple Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gene Expression Comparison between Sézary Syndrome and Lymphocytic-Variant Hypereosinophilic Syndrome Refines Biomarkers for Sézary Syndrome

Department of Dermatology, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
*
Author to whom correspondence should be addressed.
Cells 2020, 9(9), 1992; https://doi.org/10.3390/cells9091992
Submission received: 27 July 2020 / Revised: 26 August 2020 / Accepted: 27 August 2020 / Published: 29 August 2020

Abstract

:
Sézary syndrome (SS), an aggressive cutaneous T-cell lymphoma (CTCL) with poor prognosis, is characterized by the clinical hallmarks of circulating malignant T cells, erythroderma and lymphadenopathy. However, highly variable clinical skin manifestations and similarities with benign mimickers can lead to significant diagnostic delay and inappropriate therapy that can lead to disease progression and mortality. SS has been the focus of numerous transcriptomic-profiling studies to identify sensitive and specific diagnostic and prognostic biomarkers. Benign inflammatory disease controls (e.g., psoriasis, atopic dermatitis) have served to identify chronic inflammatory phenotypes in gene expression profiles, but provide limited insight into the lymphoproliferative and oncogenic roles of abnormal gene expression in SS. This perspective was recently clarified by a transcriptome meta-analysis comparing SS and lymphocytic-variant hypereosinophilic syndrome, a benign yet often clonal T-cell lymphoproliferation, with clinical features similar to SS. Here we review the rationale for selecting lymphocytic-variant hypereosinophilic syndrome (L-HES) as a disease control for SS, and discuss differentially expressed genes that may distinguish benign from malignant lymphoproliferative phenotypes, including additional context from prior gene expression studies to improve understanding of genes important in SS.

1. Introduction

Cutaneous T-cell lymphomas (CTCL) represent a heterogeneous group of skin homing T-cell malignancies, with mycosis fungoides (MF) and its leukemic variant Sézary syndrome (SS) accounting for the majority of cases. These two variants present with highly variable clinical skin inflammation that can be mistaken for benign mimickers, such as psoriasis, atopic dermatitis and other benign inflammatory dermatoses (BID) [1,2,3]. A correct diagnosis of MF/SS can be delayed for many years, potentially leading to inappropriate therapy, disease progression, and death [4,5]. MF and SS are classified by the clinical staging system jointly established by the World Health Organization and the European Organization for Research and Treatment of Cancer (WHO-EORTC) [6,7]. Diagnosis can be challenging, and currently relies on a combination of several nonspecific clinical, histopathologic and diagnostic criteria [2,5,6,8]. Presently, there remains a need to identify reliable diagnostic, stage-associated and prognostic biomarkers of MF/SS to improve initial diagnosis and identify patients at risk for progression.
Gene expression biomarkers have enormous potential to improve clinical practice for MF/SS, and innovative efforts have been taken to identify sensitive and specific diagnostic and prognostic biomarkers [9,10,11,12]. Studies have employed “disease controls” like psoriasis and atopic dermatitis to filter out gene expression signals contributed by inflammatory processes [11,13,14,15,16,17]. Many BIDs display some clinical, histologic and immunologic features that resemble MF/SS [1,2,18]. While MF and SS are caused by neoplastic T cells, benign/reactive T cells play a prominent role in inflammatory symptoms and contribute inflammatory gene expression signatures that may confound early diagnosis of MF/SS [19]. The inflammatory milieu of late MF and SS acquires a T helper type-2 (Th2)-biased phenotype, with additional regulatory and Th17-like features [20]. SS shares many similarities with atopic dermatitis, which is also characterized by Th2 skin inflammation, and can acquire additional Th-phenotypes in chronic disease (reviewed by Saulite et al. [18]). While Th2 skin inflammation is common, Th1 and Th17 infiltrates are typical of psoriasis, and the inflammatory phenotype of contact dermatitis varies with exposure [21] (Table 1). Nevertheless, comparing the T cell phenotypes of BIDs to the dysregulated T cell phenotypes in CTCL has contributed important information to gene expression studies. Boonk et al. [8] effectively demonstrated that cases of benign erythroderma express some SS-biomarker genes at levels that exceed healthy donors, while the most specific SS-biomarker genes showed the least overlap in mRNA expression between SS and benign erythroderma cases. Inclusion of disease controls for benign/reactive inflammation can therefore improve the specificity of gene expression data to identify highly relevant genes unique to MF and/or SS, and exclude genes that could misclassify patients.
A remaining challenge to the understanding of SS is the lack of a well-defined, pre/early-neoplastic T-cell population suitable for studies of stage progression, and the evolution of neoplastic phenotypes. However, this role was recently filled by a benign T-cell lymphoproliferative disorder known as lymphocytic-variant hypereosinophilic syndrome (L-HES) [22], which has skin inflammation and hematologic abnormalities that resemble SS [23,24,25,26,27,28,29]. A dominant T-cell clone in the blood is detected in a large majority of L-HES cases [23,25], and atypical T cells often exceed 70% of circulating lymphocytes [30]. While clonal T-cell populations can occasionally be detected in BIDs, they are not typical, nor a defining feature [12]. BIDs are characterized by polyclonal infiltrates of reactive T cells, and are not considered cutaneous lymphoproliferative disorders [31,32]. The combination of benign inflammatory and clonal lymphoproliferative phenotypes makes L-HES a unique disease control (Table 1) that can provide meaningful biological context that was previously lacking in transcriptomic studies of SS [22].
In this review, we will present the clinical and molecular similarities between SS and L-HES, discuss the rationale for using L-HES as a disease control for SS, and review the genes highlighted in a recent meta-analysis of SS and L-HES transcriptomes. We will also incorporate into perspective prior gene expression and functional studies of SS that together may distinguish characteristics of malignant and benign lymphoproliferative phenotypes, and their significance.

2. Clinical Features of SS and L-HES

Early MF presents with limited skin involvement and an indolent clinical course that may last for decades. Disease progression can include an increase in affected skin area, thickening of skin lesions from flat patches and plaques to nodular tumors, and the spread of malignant T cells to lymph nodes and visceral tissues (Figure 1) [33]. A subset of MF patients develops limited blood involvement (stage IIIB), and some may progress to frank SS satisfying B2 blood criteria (stage IVA1, Figure 1). SS is an advanced, leukemic variant of MF, distinguished from MF by generalized erythroderma and a high burden of tumor cells circulating in peripheral blood (Figure 1). However, the vast majority of SS cases arise de novo, without prior definable skin lesions characteristic of MF. This observation is consistent with the notion that MF and SS arise from distinct memory T cell subsets [34]. Most cases of SS share a classic triad of symptoms: pruritic erythroderma, lymphadenopathy, and a high burden of circulating clonal T cells. The B2 threshold for blood involvement in SS is met by Sézary cells in excess of 1000 cells/μL, CD4/CD8 ≥ 10, CD4+CD7 cells ≥ 30%, or CD4+CD26 cells ≥ 40% [6,33].
While SS is a moderately aggressive peripheral T-cell lymphoma with a 5-year overall survival rate of 36% [7], L-HES is typically indolent [25,35]. Chronic L-HES is infrequently associated with cytogenetic changes [23,25], whereas SS T cells harbor frequent chromosomal abnormalities and widespread changes in epigenetic status that can alter gene expression and prognosis (Table 2) [36,37,38,39,40,41]. Despite these important differences, SS and L-HES have similarities in a number of clinical and molecular findings (Table 2). In both SS and L-HES, eosinophils and abnormal T cells can be detected in both blood and skin, and over two thirds of L-HES patients experience skin inflammation and pruritus [25,27]. While a clonal T cell population is detected in the majority of L-HES cases, there is no consensus threshold for circulating, abnormal T cells, which show variable immunophenotypes, most often CD3CD4+, with frequent CD7 loss similar to SS (Table 2) [24,35,42]. In contrast, CD26 loss is common to SS but has not been reported in L-HES. Importantly, prior studies have shown that L-HES CD3CD4+ T cells have a CD45RO+ memory phenotype [29,30,42], which is shared by the majority of abnormal T cells in SS [43].
Causes of eosinophilia can be broadly classified as primary/neoplastic or secondary/reactive. Primary eosinophilias are defined by the presence of clonal, neoplastic eosinophils, such as in chronic eosinophilic leukemia or myeloproliferative HES [44]. Neoplastic eosinophil/myeloid clones may bear cytogenetic abnormalities such as the FIP1-like-1-platelet-derived growth factor receptor-alpha (FIP1L1-PDGFRA) fusion, which is effectively targeted by imatinib therapy [45]. Secondary eosinophilias are polyclonal and reactive, and are thought to depend on cytokines and growth factors that support eosinophil maturation and/or proliferation. They may be the result of parasitic or other infectious, allergic diseases, or lymphoid neoplasms and lymphoproliferations, which include L-HES [29]. L-HES is defined by early and severe eosinophilia secondary to an over proliferation of abnormal, and often clonal, Th2 T-cells secreting the eosinophilopoietic cytokine interleukin-5 (IL-5). An important diagnostic criterion for L-HES is eosinophilia exceeding 1500/µL in blood. Similarly, blood eosinophils are elevated in a subset of SS patients, and eosinophilia >700/μL is an indicator for disease progression in SS (Table 2) [46]. Increased eosinophils, particularly in the skin, have been associated with activation of signal transducer and activator of transcription 3 (STAT3) in CTCL T cells [47,48], and additional Janus kinase (JAK)-STAT dysfunction is frequent in CTCL, other T-cell leukemia/lymphomas, and many BIDs [49,50]. STAT3 is required for Th2 T-cell differentiation [51], and mediates IL-5 production in CTCL cell lines [52]. The discovery of an activating mutation of STAT3 in L-HES suggests a role for STAT3 dysfunction in the Th2 phenotype typical of L-HES [53]. These findings underscore the similarities in SS and L-HES at the molecular level. In addition, certain therapies are effective for both SS and L-HES, such as interferon (IFN)-α (Table 2). IFN-α has been shown to suppress IL-5 production in peripheral blood mononuclear cells (PBMCs) obtained from patients with eosinophilic SS [54]. Thus, while SS, L-HES, and BIDs are all T-cell disorders with clinical skin inflammation (Table 1), L-HES shares lymphoproliferative and eosinophilic features with SS that are absent in classical BIDs like atopic dermatitis (Table 2).

3. Gene Expression in SS and L-HES

SS gene expression profiles have been explored in a number of transcriptomic studies [14,16,17,68,71,72,73,74,75,76], and several multi-gene panels have been proposed to distinguish SS from BID with high accuracy [8,15,16,77], detect SS patients with as few as 5% circulating tumor cells [71], or offer prognostic insight [11]. However, these comparisons lacked the ability to exclude Th2 and lymphoproliferation genes, which may be expressed in both SS and L-HES, as L-HES is a Th2 lymphoproliferation.
There are two published transcriptomic studies of atypical T cells from L-HES patients. Ravoet et al. [30] observed abnormal expression of 850 genes in L-HES CD3CD4+ T cells, with notable changes for growth control genes, including abnormally high expression of IL17RB (IL-25 receptor) and altered expression of transforming growth factor-β superfamily genes. Walker et al. [53] described significant upregulation of a STAT3-target gene signature, which may contribute to the Th2-like phenotype of L-HES T cells.
The public L-HES data set from Ravoet et al. [30] was recently compared to gene expression data from SS memory T cells [22] (Figure 2). Importantly, both data sets were obtained on the same microarray platform. The outcome of this meta-analysis approach was greater confidence in the identification of biomarker genes specific to the malignant phenotype of SS T cells, which eliminated Th2- and lymphoproliferation-associated genes inherent to L-HES. A common analysis workflow was used for both data sets to identify genes of interest, and changes in SS or L-HES gene expression compared to normal donors was based on a threshold of 2-fold with q ≤ 0.05 [22]. The outcome showed a highly significant degree of overlap between the abnormal gene expression profiles of SS and L-HES T cells compared to normal T cells (Figure 2), suggesting that gene expression shared by SS and L-HES reflects benign lymphoproliferative and Th2 phenotypes rather than malignant processes. Interestingly, shared genes included DNM3, CCR4 and GATA3, which have appeared in diagnostic and prognostic gene-expression panels previously proposed for SS [8,11,15,16]. Nevertheless, L-HES and SS are distinct diseases, and gene expression abnormalities unique to either SS or L-HES were also identified by the meta-analysis (Figure 2). Many frequently published SS-biomarker genes were found in the “SS-unique” group, including increased expression of PLS3, TOX and TWIST1, and reduced expression of STAT4, confirming their association with malignancy. By identifying groups of shared and SS-unique genes associated with benign and malignant phenotypes, respectively, this novel comparison offered a new perspective on abnormal gene expression in SS.
A limitation of the meta-analysis is the small number of cases included for both SS and L-HES. However, the SS and L-HES subjects have well-annotated clinical and immune characteristics that increase confidence in the results. In addition, we conducted a literature review of ten prior SS transcriptome studies (Table 3) to determine to what extent the SS-unique genes have been identified in other SS cohorts. Of the studies used for comparison, seven used microarrays, three used bulk RNA sequencing, and one used single-cell RNA sequencing. Eight studies compared SS patients to healthy donors, and one study included BIDs. Two studies compared malignant SS cells to non-malignant cells from the same patients.
After comparing results of the meta-analysis to the ten other studies in Table 3, we determined that thirty-seven upregulated and five downregulated SS-unique genes have also been reported in at least two additional transcriptomic studies of SS (Figure 3). The ten SS-unique genes most frequently reported as upregulated in other SS cohorts are TWIST1, PGN2L1, ANK1, IKZF2, KLHL42, NEDD4L, PLS3, ST8SIA1, TOX, and TPR (Figure 3A). Each of these genes has been reported in at least four other publications. SS-unique genes frequently reported as downregulated in other SS cohorts include STAT4, GSTP1, CTSW, SYTL3, and TBX21 (Figure 3B). The small number of downregulated SS-unique genes supported by multiple other studies may reflect under-reporting of downregulated genes in the literature, as no supplemental data were available for downregulated genes from three studies [16,68,73].
We also compared genes abnormally expressed in L-HES [22,30] with other SS studies from Table 3 to identify gene expression shared by multiple SS cohorts. For genes identified as shared between SS and L-HES by the meta-analysis, eleven upregulated and eleven downregulated genes were reported in at least two other transcriptomic studies of SS (Figure 3C,D). Upregulated shared genes include DNM3, TNFSF11, CCR4, TSPAN, CDCA7, CDH1, CMIP, CPNE2, GATA3, MLF1, and SGCE (Figure 3C), and downregulated shared genes include SATB1, APBA2, GZMK, KLRB1, CCL5, TGFBR2, BCL2L11, HOPX, IFI44, and PCSK5 (Figure 3D). We also identified seven genes upregulated in L-HES that were not shared with the SS cohort from the meta-analysis, but were concordantly differentially expressed with at least two prior transcriptomic studies for SS. These genes include CCDC167 [68,74], DUSP4 [16,73], LMNA [73,74], NINJ2 [71,74], PTTG1 [17,74], TNFSF10 [68,71], and GPR171 [16,17]. Thus, many of the shared and SS-unique genes identified by the meta-analysis of SS and L-HES gene expression are supported by prior studies in SS. How well the L-HES transcriptome data of Ravoet et al. represent other L-HES cohorts will remain an open question until additional studies are performed or added to public data repositories. The remainder of this review will consider the potential functional roles of shared and unique gene expression in SS.

3.1. Gene Expression Shared by SS and L-HES

While genes with expression changes common to SS and L-HES are not ideal diagnostic biomarkers, they can provide additional insight into molecular mechanisms that support similarities in disease phenotype, and may have prognostic value. Increased expression of GATA3 and decreased expression of SATB1 are important examples. GATA3 is a Zn-finger transcription factor and master regulator of Th2 differentiation, and both GATA3 and Th2 cytokine genes are frequently overexpressed in both SS and L-HES [30,53,71,74,81,82]. The Th2 cytokine IL-5 promotes eosinophilia in SS and L-HES, and activation of the IL5 promoter by GATA3 is directly opposed by the transcription factor SATB1 [83]. Decreased expression of SATB1 observed in both SS and L-HES reduces this check on IL5 expression.
Skin homing is another phenotype common to both SS and L-HES T cells, which is mediated by CCR4, a chemokine receptor essential for cutaneous homing of T cells [84]. Skin-resident SS T cells have a higher proliferative index than circulating SS cells, which may be dependent on cytokines and other factors present in the skin microenvironment [85]. The active role of CCR4 in SS is shown by the efficacy of anti-CCR4 immunotherapy approved for use in CTCL [86]. CCR4 expression is also suppressed in SS T cells and/or cell lines by other therapies with efficacy in CTCL, including the rexinoid bexarotene and histone deacetylase inhibitors (HDACi) romidepsin and suberoylanilide hydroxamic acid (SAHA/vorinostat) [87,88].
Genes with potential roles in lymphoproliferation were also concordantly regulated in SS and L-HES, including increased expression of CDCA7, DNM3 and TNFSF11, and decreased expression of SATB1. SATB1 can sensitize SS cells to activation induced cell death [72], and low expression of SATB1 is an independent prognostic factor in CTCL [89]. TNFSF11 encodes receptor activator nuclear factor-κB ligand (RANKL), which is frequently expressed by cancer cells. The many roles for RANKL-RANK signaling in cancer-niche development, metastatic processes, neoangiogenesis, and immune escape have been reviewed recently [90]. The shared expression of TNFSF11 in SS and L-HES is consistent with the enhanced proliferation of T cells. Increased expression of DNM3 was associated with a better overall survival in a large cohort of SS patients [91], which is supported by its ability to inhibit colony formation and increase the expression of p53 protein in hepatocellular carcinoma cell lines [92]. The shared expression of DNM3 in SS and L-HES is surprising because DNM3 is a component of two multi-gene panels that differentiated SS from MF and BID cases including psoriasis, atopic dermatitis, and benign erythroderma with sensitivity and specificity over 95% [8,16].
CDCA7 is a shared gene with a previously unrecognized role in SS, despite its detection in several prior transcriptomic studies of SS [16,72,74]. CDCA7 is frequently overexpressed in human cancers [93], including a large variety of B- and T-cell leukemias and lymphomas, and its loss has been shown to reduce T- and B-lymphomagenesis in vivo [94,95]. Thus, CDCA7 expression may be important to the proliferative nature of SS and L-HES. CDCA7 has weak transforming activity of its own, and is a direct target of C-Myc oncoprotein [96]. CDCA7 also contributes to anchorage-independent growth [94], and silencing CDCA7 impaired lymphoma cell migration and invasion in in vitro and in vivo models [95]. A single cell RNA sequencing study of malignant and nonmalignant T cells from one SS patient revealed that CDCA7 expression was an early event in the evolution of heterogeneous transcriptional states in SS clonal T cells [74], suggesting that high CDCA7 expression may provide a receptive environment for additional changes. In summary, many gene expression abnormalities shared by SS and L-HES T cells appear to fulfill roles that promote Th2-like and proliferative phenotypes. While SS biomarkers DNM3 and SATB1 may have promise as prognostic biomarkers, they do not reflect processes unique to malignant T cells.

3.2. Gene Expression Unique to SS

The meta-analysis strategy comparing SS and L-HES is supported by the inclusion in the SS-unique category of many genes with rich publication histories in SS, such as PLS3, TWIST1 and STAT4. In addition, the potential importance of some less well-recognized SS biomarker genes has been elevated by their new SS-unique status. SS-unique genes are associated with several cancer-promoting mechanisms including enhanced survival, oncogenic mi-RNAs, T-cell exhaustion and immunoregulation.

3.2.1. Well-Established SS-Unique Biomarker Genes

PLS3 and TWIST1 are two frequently reported and highly expressed SS-biomarker genes with established roles in the epithelial to mesenchymal transition, and frequent association with disease progression in solid malignancies. PLS3 is one of the earliest SS biomarker genes identified and is consistently upregulated in the majority of SS cases [15,97,98], including SS without erythroderma [99], but is absent in the blood of normal donors, BID, and MF patients without blood involvement [10,97,98,100]. Because PLS3 expression is limited to the CD26 negative T-cell population [98], and malignant T-cell clones [97], PLS3 expression correlates with blood disease burden in SS [10]. In solid cancers, PLS3 expression is a marker of circulating tumor cells with a metastatic phenotype, and this is associated with a poor prognosis [101,102]. In SS, PLS3 expression has been associated with enhanced migration and apoptosis resistance [97]. However, in contrast to solid tumors, PLS3 expression in SS is associated with a better prognosis [91]. Single cell RNA sequencing of SS T cells revealed that PLS3 was expressed in only one of two divergent evolutionary transcriptional states that developed in a malignant T-cell clone [74].
Numerous studies have reported overexpression of TWIST1 in SS [8,14,16,22,72]. TWIST1 is a highly conserved developmental transcription factor with diverse pathological functions in solid and hematological tumors [103,104]. In CTCL, the frequency and intensity of lesional TWIST1 expression was shown to increase with clinical stage, and was highest in SS; all late stage lesions that stained positive for TWIST1 were also positive for C-Myc [105]. This suggests that increased expression of TWIST1 plays a role in CTCL progression, and is consistent with the recent discovery that TWIST1 is involved in skin cancer initiation, maintenance and progression in a dose dependent manner [106]. TWIST1 is also an important suppressor of chronic Th1 inflammation [107,108], and ectopic expression of Twist1 reduced effector cytokine expression in both Th1- and Th2-polarized cells [108]. This feature of TWIST1 may contribute to poorly inducible cytokine gene expression observed in SS cells [22,109]. In addition, the mechanism of TWIST1 overexpression may vary between patients, resulting from promoter hypomethylation [110], or gain of chromosomal region 7p21.1 harboring the TWIST1 gene [111].
Reduced STAT4 expression is one of the most consistent findings in transcriptional studies of SS [8]. However, genes with reduced expression have limited usefulness as stand-alone biomarkers, and STAT4 has appeared in several multi-gene biomarker panels. Showe et al. [112] first described the loss of STAT4 expression in SS PBMCs, and Litvinov et al. [113] confirmed that low STAT4 expression in CTCL lesional skin is associated with progressive disease. However, in a separate study, higher STAT4 expression was associated with HDACi resistance in MF/SS patients [114], suggesting that alternative pathways to malignancy may be active in these patients.
STAT4 expression loss in SS appears to be related to STAT5-mediated expression of oncogenic microRNA-155 (miR-155) [113,115], as knockdown of miR-155 in MF cell lines increased STAT4 expression and improved the apoptotic response to SAHA [113,116]. Ralfkiaer et al. [117] demonstrated that a microRNA panel including miRNA-155, miRNA-203 and miRNA-205 could accurately classify 95% of CTCL from BID cases. In addition to targeting STAT4 mRNA, miR-155 increases mutations by simultaneously interfering with DNA repair and cell cycle checkpoints [118], and has been associated with several solid and hematological malignancies [119]. Phase 1 and phase 2 clinical trials are underway for Cobomarsen/MRG-106, a novel oligonucleotide inhibitor of miR-155 (NCT02580552, NCT03713320, NCT03837457), with promising interim results that emphasize the importance of miR-155 to CTCL pathogenesis [119].
In summary, TWIST1, PLS3 and STAT4 remain some of the most reliable diagnostic biomarkers of SS due to their selective dysregulation in malignant T cells, and absent expression in L-HES. Indeed, the combined altered expression of these three biomarker genes was demonstrated in a multicenter study to distinguish cases of SS from erythrodermic inflammatory dermatoses with 98% sensitivity and 100% specificity [8].

3.2.2. SS-Unique Genes Associated with Regulatory and Exhaustion Phenotypes

It has been suggested that the phenotype of SS T cells is plastic and may respond to microenvironmental factors [43,120,121]. While it is well known that SS malignant T cells express Th2 cytokines, they also variably express characteristics of regulatory T cells (Tregs), including expression of forkhead box P3 (FoxP3), inhibitory receptors, and immunoregulatory cytokines [81,122]. Moreover, these FoxP3+ malignant T cells have suppressive activity [123], and are associated with a worse prognosis [124].
CTCL T cells from lesional skin also exhibit phenotypes of exhausted T cells, potentially due to persistent antigen stimulation in the skin microenvironment. Increased surface expression of compensatory inhibitory receptors has been detected in early stage skin lesions, and a positive correlation was found between increased gene expression for checkpoint receptors and higher disease stage [125]. The SS-unique genes IKZF2, FCRL3, TIGIT and TOX have each been associated with Tregs and/or T-cell exhaustion. FCRL3 is an orphan Fc-like receptor with immunoregulatory properties [126], and the inhibitory receptor TIGIT is expressed by chronically activated, exhausted T cells and activated Tregs [127,128]. Coexpression of TIGIT and FCRL3 identifies a population of highly suppressive, Helios+FoxP3+ conventional Tregs [129], and several studies have described elevated surface expression of TIGIT and FCRL3 on circulating CD4+ T cells in SS [78,122,130]. In addition, high expression of TIGIT or FCRL3 has been shown to correlate with CD26 loss and a high tumor burden [78,122], and clinical remission coincided with the disappearance of CD4+CD26 T cells expressing FCRL3 [78], suggesting that the immunoregulatory phenotype of SS T cells plays an active role in disease.
The transcription factor TOX is required for the thymic development of all CD4 T-cell lineages [131], while in mature T cells, TOX is upregulated in exhausted T cells in the context of chronic antigen stimulation [132]. Strong TOX staining is observed in lesional skin from a large majority of SS and MF cases, but is much less common in BID skin. High TOX expression correlates with increased disease-specific mortality in SS [133], and predicts disease progression and poor survival in early MF [134]. Suppression of TOX expression increases cell cycle regulators and apoptosis in CTCL cell lines, and greatly reduces the growth of CTCL tumor xenografts in mice [133]. In addition, increased expression of both TIGIT and TOX was observed during the malignant progression of chronic L-HES to T-cell lymphoma [22]. These findings suggest that SS-unique gene expression is associated with regulatory and exhaustion phenotypes of malignant T cells in SS.

3.2.3. New and Promising SS-Unique Biomarker Genes

Gene expression studies for SS have identified many differentially expressed genes, but there are additional SS-unique genes that merit further discussion. ANK1, GATA6 and HDAC9 have not been well characterized as SS biomarkers, but recent developments provide insight into their potential roles in SS. Overexpression of ANK1 was detected by several gene expression profiling studies [16,75], and was independently validated by Moerman-Herzog et al. [22]. Importantly, the last intron of ANK1 harbors MIR486, which is also overexpressed in SS, and is involved in cell survival [135]. Co-transcription of ANK1 mRNA and miR-486 is activated in hematopoietic progenitor cells by MYB oncoprotein, in erythroid cells by GATA1 transcription factor, and in muscle cells by myocardin related transcription factor A (reviewed in [136]). In the meta-analysis of SS and L-HES [22], MYB was significantly upregulated in SS and down-regulated in L-HES, and the miRNA-486 target MAF [137] was significantly downregulated in SS and unchanged in L-HES. ANK1 and miR-486 can also be upregulated in a p53 dependent manner following DNA damage. In this context, miRNA-486 promoted G2/M arrest, and ANK1 enhanced cell motility [138]. In the same study, high ANK1 expression correlated with reduced survival in several cancers including chronic lymphocytic leukemia, and increased survival was demonstrated in patients with a high positive correlation between TP53 and ANK1 expression.
The transcription factor GATA6 is widely associated with tumorigenesis [139], including an oncogenic role in CTCL [140]. In SS T cells, GATA6 is overexpressed and directly induces expression of CD137L, which promotes proliferation, survival and migration of SS T cells and CTCL cell lines [22,110,140]. Higher numbers of GATA6+, CD137L+ and CD137+ cells have been detected in all stages of MF/SS lesional skin compared to normal skin [140]. Furthermore, expression of GATA6 mRNA in SS T cells and CTCL cell lines is enhanced by DNA hypomethylation and histone acetylation [110,140], suggesting that ectopic GATA6 expression is activated by epigenetic dysregulation common to CTCL T cells.
HDACi therapy differentially modifies the distinct chromatin accessibility signatures observed in leukemic and host T cells from CTCL patients. The only HDAC gene differentially expressed between host and SS cells is HDAC9, and the HDAC9 locus is strongly accessible only in leukemic T cells [40]. Several studies have shown that HDAC9 limits the proliferation and suppressive potential of Tregs (reviewed in [141]), but promotes B cell lymphomagenesis [142], suggesting that the function of HDAC9 is context dependent. Interestingly, in vitro acquired resistance to ricolinostat, a selective HDAC6 inhibitor, was associated with higher HDAC9 expression in a B-cell lymphoma cell line [143], and HDAC9 expression has been associated with drug resistance and poor prognosis in a variety of solid malignancies [144,145]. Thus, HDAC9, ANK1, and GATA6 are biomarkers with SS-unique expression discovered by the comparison of SS and L-HES that may illuminate important roles in SS pathogenesis.

4. Concluding Remarks

Identifying highly specific biomarkers for CTCL remains a challenge, but selecting informative controls can improve the outcome. SS and L-HES are skin-tropic diseases that share many clinical and immunological features that are mirrored in their gene expression profiles, which exhibit prominent Th2-like and lymphoproliferative signatures, more so than other inflammatory skin diseases. It is now clear that several previously described SS biomarkers are shared by L-HES, indicating that a reevaluation of their functional significance in oncogenesis unique to SS is needed. The remaining biomarker genes unique to SS are now more focused on malignant gene expression phenotypes, including immunomodulation, EMT, cell survival, and co-expression of oncogenic miRNAs. The refocused SS-unique gene set includes genes that have received little prior attention in SS, presenting an opportunity to gain insight from these less-studied genes [146] which may have potentially important roles in SS. One of the earliest gene expression profiling studies of SS, conducted by Kari et al. [71], noted that while as few as eight genes with high predictive power could be used to accurately classify SS patients with low tumor burden, the best 85 genes could be removed before classification dipped below 100%, and the remaining group of 300 genes with lower predictive power was still highly accurate. This suggests that expanded gene panels could mitigate the confounding effects of small sample size and disease heterogeneity, which have significantly impeded efforts to identify diagnostic gene expression panels that are both sensitive and specific in multiple cohorts, and to illuminate the underlying mechanisms of SS pathogenesis.
In closing, comparative analysis of SS and L-HES gene expression identified subsets of genes that are unique to each disease, and will serve to improve diagnostic accuracy. A new focus on gene expression associated only with the malignant T-cell phenotype in SS may also illuminate potential therapeutic targets for these T-cell diseases. Understanding the roles of these genes in SS, and the processes by which these genes become dysregulated will yield insight into the mechanisms driving these rare and diagnostically challenging diseases.

Author Contributions

Conceptualization, supervision, editing and critical review, H.K.W.; Conceptualization, original draft preparation, bibliography, A.M.-H.; Review and editing, S.J.M.; All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Martin and Dorothy Spatz Charitable Foundation (H.K.W.).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Hodak, E.; Amitay-Laish, I. Mycosis fungoides: A great imitator. Clin. Dermatol. 2019, 37, 255–267. [Google Scholar] [CrossRef] [PubMed]
  2. Klemke, C.D.; Booken, N.; Weiss, C.; Nicolay, J.P.; Goerdt, S.; Felcht, M.; Geraud, C.; Kempf, W.; Assaf, C.; Ortonne, N.; et al. Histopathological and immunophenotypical criteria for the diagnosis of Sezary syndrome in differentiation from other erythrodermic skin diseases: A European Organisation for Research and Treatment of Cancer (EORTC) Cutaneous Lymphoma Task Force Study of 97 cases. Br. J. Dermatol. 2015, 173, 93–105. [Google Scholar] [CrossRef] [PubMed]
  3. Mangold, A.R.; Thompson, A.K.; Davis, M.D.; Saulite, I.; Cozzio, A.; Guenova, E.; Hodak, E.; Amitay-Laish, I.; Pujol, R.M.; Pittelkow, M.R.; et al. Early clinical manifestations of Sezary syndrome: A multicenter retrospective cohort study. J. Am. Acad. Dermatol. 2017, 77, 719–727. [Google Scholar] [CrossRef] [PubMed]
  4. Foo, S.H.; Shah, F.; Chaganti, S.; Stevens, A.; Scarisbrick, J.J. Unmasking mycosis fungoides/Sezary syndrome from preceding or co-existing benign inflammatory dermatoses requiring systemic therapies: Patients frequently present with advanced disease and have an aggressive clinical course. Br. J. Dermatol. 2016, 174, 901–904. [Google Scholar] [CrossRef] [PubMed]
  5. Scarisbrick, J.J.; Quaglino, P.; Prince, H.M.; Papadavid, E.; Hodak, E.; Bagot, M.; Servitje, O.; Berti, E.; Ortiz-Romero, P.; Stadler, R.; et al. The PROCLIPI international registry of early-stage mycosis fungoides identifies substantial diagnostic delay in most patients. Br. J. Dermatol. 2019, 181, 350–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Scarisbrick, J.J.; Hodak, E.; Bagot, M.; Stranzenbach, R.; Stadler, R.; Ortiz-Romero, P.L.; Papadavid, E.; Evison, F.; Knobler, R.; Quaglino, P.; et al. Blood classification and blood response criteria in mycosis fungoides and Sezary syndrome using flow cytometry: Recommendations from the EORTC cutaneous lymphoma task force. Eur. J. Cancer 2018, 93, 47–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Willemze, R.; Cerroni, L.; Kempf, W.; Berti, E.; Facchetti, F.; Swerdlow, S.H.; Jaffe, E.S. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood 2019, 133, 1703–1714. [Google Scholar] [CrossRef]
  8. Boonk, S.E.; Zoutman, W.H.; Marie-Cardine, A.; van der Fits, L.; Out-Luiting, J.J.; Mitchell, T.J.; Tosi, I.; Morris, S.L.; Moriarty, B.; Booken, N.; et al. Evaluation of Immunophenotypic and Molecular Biomarkers for Sezary Syndrome Using Standard Operating Procedures: A Multicenter Study of 59 Patients. J. Investig. Dermatol. 2016, 136, 1364–1372. [Google Scholar] [CrossRef] [Green Version]
  9. Dulmage, B.; Geskin, L.; Guitart, J.; Akilov, O.E. The biomarker landscape in mycosis fungoides and Sezary syndrome. Exp. Dermatol. 2017, 26, 668–676. [Google Scholar] [CrossRef] [Green Version]
  10. Tang, N.; Gibson, H.; Germeroth, T.; Porcu, P.; Lim, H.W.; Wong, H.K. T-plastin (PLS3) gene expression differentiates Sezary syndrome from mycosis fungoides and inflammatory skin diseases and can serve as a biomarker to monitor disease progression. Br. J. Dermatol. 2010, 162, 463–466. [Google Scholar] [CrossRef]
  11. Litvinov, I.V.; Netchiporouk, E.; Cordeiro, B.; Dore, M.A.; Moreau, L.; Pehr, K.; Gilbert, M.; Zhou, Y.; Sasseville, D.; Kupper, T.S. The Use of Transcriptional Profiling to Improve Personalized Diagnosis and Management of Cutaneous T-cell Lymphoma (CTCL). Clin. Cancer Res. 2015, 21, 2820–2829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Walia, R.; Yeung, C.C.S. An Update on Molecular Biology of Cutaneous T Cell Lymphoma. Front. Oncol. 2019, 9, 1558. [Google Scholar] [CrossRef] [PubMed]
  13. Van Kester, M.S.; Borg, M.K.; Zoutman, W.H.; Out-Luiting, J.J.; Jansen, P.M.; Dreef, E.J.; Vermeer, M.H.; van Doorn, R.; Willemze, R.; Tensen, C.P. A meta-analysis of gene expression data identifies a molecular signature characteristic for tumor-stage mycosis fungoides. J. Investig. Dermatol. 2012, 132, 2050–2059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Van Doorn, R.; Dijkman, R.; Vermeer, M.H.; Out-Luiting, J.J.; van der Raaij-Helmer, E.M.; Willemze, R.; Tensen, C.P. Aberrant expression of the tyrosine kinase receptor EphA4 and the transcription factor twist in Sezary syndrome identified by gene expression analysis. Cancer Res. 2004, 64, 5578–5586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Nebozhyn, M.; Loboda, A.; Kari, L.; Rook, A.H.; Vonderheid, E.C.; Lessin, S.; Berger, C.; Edelson, R.; Nichols, C.; Yousef, M.; et al. Quantitative PCR on 5 genes reliably identifies CTCL patients with 5% to 99% circulating tumor cells with 90% accuracy. Blood 2006, 107, 3189–3196. [Google Scholar] [CrossRef] [Green Version]
  16. Booken, N.; Gratchev, A.; Utikal, J.; Weiss, C.; Yu, X.; Qadoumi, M.; Schmuth, M.; Sepp, N.; Nashan, D.; Rass, K.; et al. Sezary syndrome is a unique cutaneous T-cell lymphoma as identified by an expanded gene signature including diagnostic marker molecules CDO1 and DNM3. Leukemia 2008, 22, 393–399. [Google Scholar] [CrossRef] [Green Version]
  17. Hahtola, S.; Tuomela, S.; Elo, L.; Hakkinen, T.; Karenko, L.; Nedoszytko, B.; Heikkila, H.; Saarialho-Kere, U.; Roszkiewicz, J.; Aittokallio, T.; et al. Th1 response and cytotoxicity genes are down-regulated in cutaneous T-cell lymphoma. Clin. Cancer Res. 2006, 12, 4812–4821. [Google Scholar] [CrossRef] [Green Version]
  18. Saulite, I.; Hoetzenecker, W.; Weidinger, S.; Cozzio, A.; Guenova, E.; Wehkamp, U. Sezary Syndrome and Atopic Dermatitis: Comparison of Immunological Aspects and Targets. BioMed Res. Int. 2016, 2016, 9717530. [Google Scholar] [CrossRef] [Green Version]
  19. Vieyra-Garcia, P.; Crouch, J.D.; O’Malley, J.T.; Seger, E.W.; Yang, C.H.; Teague, J.E.; Vromans, A.M.; Gehad, A.; Win, T.S.; Yu, Z.; et al. Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma. JCI Insight 2019, 4. [Google Scholar] [CrossRef]
  20. Krejsgaard, T.; Lindahl, L.M.; Mongan, N.P.; Wasik, M.A.; Litvinov, I.V.; Iversen, L.; Langhoff, E.; Woetmann, A.; Odum, N. Malignant inflammation in cutaneous T-cell lymphoma-a hostile takeover. Semin. Immunopathol. 2017, 39, 269–282. [Google Scholar] [CrossRef]
  21. Dhingra, N.; Shemer, A.; da Rosa, J.C.; Rozenblit, M.; Fuentes-Duculan, J.; Gittler, J.K.; Finney, R.; Czarnowicki, T.; Zheng, X.; Xu, H.; et al. Molecular profiling of contact dermatitis skin identifies allergen-dependent differences in immune response. J. Allergy Clin. Immunol. 2014, 134, 362–372. [Google Scholar] [CrossRef] [PubMed]
  22. Moerman-Herzog, A.M.; Acheampong, D.A.; Brooks, A.G.; Blair, S.M.; Hsu, P.C.; Wong, H.K. Transcriptome analysis of Sezary syndrome and lymphocytic-variant hypereosinophilic syndrome T cells reveals common and divergent genes. Oncotarget 2019, 10, 5052–5069. [Google Scholar] [CrossRef] [PubMed]
  23. Roufosse, F.; Cogan, E.; Goldman, M. Lymphocytic Variant Hypereosinophilic Syndromes. Immunol. Allergy Clin. N. Am. 2007, 27, 389–413. [Google Scholar] [CrossRef]
  24. Roufosse, F.; Schandene, L.; Sibille, C.; Willard-Gallo, K.; Kennes, B.; Efira, A.; Goldman, M.; Cogan, E. Clonal Th2 lymphocytes in patients with the idiopathic hypereosinophilic syndrome. Br. J. Haematol. 2000, 109, 540–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lefevre, G.; Copin, M.C.; Roumier, C.; Aubert, H.; Avenel-Audran, M.; Grardel, N.; Poulain, S.; Staumont-Salle, D.; Seneschal, J.; Salles, G.; et al. CD3-CD4+ lymphoid variant of hypereosinophilic syndrome: Nodal and extranodal histopathological and immunophenotypic features of a peripheral indolent clonal T-cell lymphoproliferative disorder. Haematologica 2015, 100, 1086–1095. [Google Scholar] [CrossRef]
  26. Ogbogu, P.U.; Bochner, B.S.; Butterfield, J.H.; Gleich, G.J.; Huss-Marp, J.; Kahn, J.E.; Leiferman, K.M.; Nutman, T.B.; Pfab, F.; Ring, J.; et al. Hypereosinophilic syndrome: A multicenter, retrospective analysis of clinical characteristics and response to therapy. J. Allergy Clin. Immunol. 2009, 124, 1319–1325. [Google Scholar] [CrossRef] [Green Version]
  27. Leiferman, K.M.; Gleich, G.J.; Peters, M.S. Dermatologic manifestations of the hypereosinophilic syndromes. Immunol. Allergy Clin. N. Am. 2007, 27, 415–441. [Google Scholar] [CrossRef]
  28. Simon, H.U.; Plotz, S.G.; Dummer, R.; Blaser, K. Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N. Engl. J. Med. 1999, 341, 1112–1120. [Google Scholar] [CrossRef]
  29. Roufosse, F.; Garaud, S.; de Leval, L. Lymphoproliferative disorders associated with hypereosinophilia. Semin. Hematol. 2012, 49, 138–148. [Google Scholar] [CrossRef]
  30. Ravoet, M.; Sibille, C.; Gu, C.; Libin, M.; Haibe-Kains, B.; Sotiriou, C.; Goldman, M.; Roufosse, F.; Willard-Gallo, K. Molecular profiling of CD3-CD4+ T cells from patients with the lymphocytic variant of hypereosinophilic syndrome reveals targeting of growth control pathways. Blood 2009, 114, 2969–2983. [Google Scholar] [CrossRef] [Green Version]
  31. Piccolo, V.; Russo, T.; Agozzino, M.; Vitiello, P.; Caccavale, S.; Alfano, R.; Argenziano, G. Dermoscopy of Cutaneous Lymphoproliferative Disorders: Where Are We Now? Dermatology 2018, 234, 131–136. [Google Scholar] [CrossRef] [PubMed]
  32. Cerroni, L. Lymphoproliferative lesions of the skin. J. Clin. Pathol. 2006, 59, 813–826. [Google Scholar] [CrossRef] [PubMed]
  33. Olsen, E.A.; Whittaker, S.; Kim, Y.H.; Duvic, M.; Prince, H.M.; Lessin, S.R.; Wood, G.S.; Willemze, R.; Demierre, M.F.; Pimpinelli, N.; et al. Clinical end points and response criteria in mycosis fungoides and Sezary syndrome: A consensus statement of the International Society for Cutaneous Lymphomas, the United States Cutaneous Lymphoma Consortium, and the Cutaneous Lymphoma Task Force of the European Organisation for Research and Treatment of Cancer. J. Clin. Oncol. 2011, 29, 2598–2607. [Google Scholar] [CrossRef] [PubMed]
  34. Campbell, J.J.; Clark, R.A.; Watanabe, R.; Kupper, T.S. Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: A biologic rationale for their distinct clinical behaviors. Blood 2010, 116, 767–771. [Google Scholar] [CrossRef] [PubMed]
  35. Gotlib, J. World Health Organization-defined eosinophilic disorders: 2015 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2015, 90, 1077–1089. [Google Scholar] [CrossRef]
  36. Izykowska, K.; Przybylski, G.K.; Gand, C.; Braun, F.C.; Grabarczyk, P.; Kuss, A.W.; Olek-Hrab, K.; Bastidas Torres, A.N.; Vermeer, M.H.; Zoutman, W.H.; et al. Genetic rearrangements result in altered gene expression and novel fusion transcripts in Sezary syndrome. Oncotarget 2017, 8, 39627–39639. [Google Scholar] [CrossRef] [Green Version]
  37. Choi, J.; Goh, G.; Walradt, T.; Hong, B.S.; Bunick, C.G.; Chen, K.; Bjornson, R.D.; Maman, Y.; Wang, T.; Tordoff, J.; et al. Genomic landscape of cutaneous T cell lymphoma. Nat. Genet. 2015, 47, 1011–1019. [Google Scholar] [CrossRef]
  38. da Silva Almeida, A.C.; Abate, F.; Khiabanian, H.; Martinez-Escala, E.; Guitart, J.; Tensen, C.P.; Vermeer, M.H.; Rabadan, R.; Ferrando, A.; Palomero, T. The mutational landscape of cutaneous T cell lymphoma and Sezary syndrome. Nat. Genet. 2015, 47, 1465–1470. [Google Scholar] [CrossRef] [Green Version]
  39. Van Doorn, R.; Slieker, R.C.; Boonk, S.E.; Zoutman, W.H.; Goeman, J.J.; Bagot, M.; Michel, L.; Tensen, C.P.; Willemze, R.; Heijmans, B.T.; et al. Epigenomic Analysis of Sezary Syndrome Defines Patterns of Aberrant DNA Methylation and Identifies Diagnostic Markers. J. Investig. Dermatol. 2016, 136, 1876–1884. [Google Scholar] [CrossRef] [Green Version]
  40. Qu, K.; Zaba, L.C.; Satpathy, A.T.; Giresi, P.G.; Li, R.; Jin, Y.; Armstrong, R.; Jin, C.; Schmitt, N.; Rahbar, Z.; et al. Chromatin Accessibility Landscape of Cutaneous T Cell Lymphoma and Dynamic Response to HDAC Inhibitors. Cancer Cell 2017, 32, 27–41. [Google Scholar] [CrossRef] [Green Version]
  41. Caprini, E.; Cristofoletti, C.; Arcelli, D.; Fadda, P.; Citterich, M.H.; Sampogna, F.; Magrelli, A.; Censi, F.; Torreri, P.; Frontani, M.; et al. Identification of key regions and genes important in the pathogenesis of sezary syndrome by combining genomic and expression microarrays. Cancer Res. 2009, 69, 8438–8446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Roufosse, F.; Schandene, L.; Sibille, C.; Kennes, B.; Efira, A.; Cogan, E.; Goldman, M. T-cell receptor-independent activation of clonal Th2 cells associated with chronic hypereosinophilia. Blood 1999, 94, 994–1002. [Google Scholar] [CrossRef] [PubMed]
  43. Roelens, M.; Delord, M.; Ram-Wolff, C.; Marie-Cardine, A.; Alberdi, A.; Maki, G.; Homyrda, L.; Bensussan, A.; Bagot, M.; Toubert, A.; et al. Circulating and skin-derived Sezary cells: Clonal but with phenotypic plasticity. Blood 2017, 130, 1468–1471. [Google Scholar] [CrossRef] [PubMed]
  44. Kahn, J.E.; Groh, M.; Lefevre, G. (A Critical Appraisal of) Classification of Hypereosinophilic Disorders. Front. Med. 2017, 4, 216. [Google Scholar] [CrossRef] [Green Version]
  45. Pardanani, A.; Brockman, S.R.; Paternoster, S.F.; Flynn, H.C.; Ketterling, R.P.; Lasho, T.L.; Ho, C.L.; Li, C.Y.; Dewald, G.W.; Tefferi, A. FIP1L1-PDGFRA fusion: Prevalence and clinicopathologic correlates in 89 consecutive patients with moderate to severe eosinophilia. Blood 2004, 104, 3038–3045. [Google Scholar] [CrossRef]
  46. Tancrede-Bohin, E.; Ionescu, M.A.; de La Salmoniere, P.; Dupuy, A.; Rivet, J.; Rybojad, M.; Dubertret, L.; Bachelez, H.; Lebbe, C.; Morel, P. Prognostic value of blood eosinophilia in primary cutaneous T-cell lymphomas. Arch. Dermatol. 2004, 140, 1057–1061. [Google Scholar] [CrossRef] [Green Version]
  47. Netchiporouk, E.; Litvinov, I.V.; Moreau, L.; Gilbert, M.; Sasseville, D.; Duvic, M. Deregulation in STAT signaling is important for cutaneous T-cell lymphoma (CTCL) pathogenesis and cancer progression. Cell Cycle 2014, 13, 3331–3335. [Google Scholar] [CrossRef]
  48. Fredholm, S.; Gjerdrum, L.M.; Willerslev-Olsen, A.; Petersen, D.L.; Nielsen, I.O.; Kauczok, C.S.; Wobser, M.; Ralfkiaer, U.; Bonefeld, C.M.; Wasik, M.A.; et al. STAT3 activation and infiltration of eosinophil granulocytes in mycosis fungoides. Anticancer Res. 2014, 34, 5277–5286. [Google Scholar]
  49. Waldmann, T.A.; Chen, J. Disorders of the JAK/STAT Pathway in T Cell Lymphoma Pathogenesis: Implications for Immunotherapy. Annu. Rev. Immunol. 2017, 35, 533–550. [Google Scholar] [CrossRef]
  50. Damsky, W.; King, B.A. JAK inhibitors in dermatology: The promise of a new drug class. J. Am. Acad. Dermatol. 2017, 76, 736–744. [Google Scholar] [CrossRef]
  51. Stritesky, G.L.; Muthukrishnan, R.; Sehra, S.; Goswami, R.; Pham, D.; Travers, J.; Nguyen, E.T.; Levy, D.E.; Kaplan, M.H. The transcription factor STAT3 is required for T helper 2 cell development. Immunity 2011, 34, 39–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Nielsen, M.; Nissen, M.H.; Gerwien, J.; Zocca, M.B.; Rasmussen, H.M.; Nakajima, K.; Ropke, C.; Geisler, C.; Kaltoft, K.; Odum, N. Spontaneous interleukin-5 production in cutaneous T-cell lymphoma lines is mediated by constitutively activated Stat3. Blood 2002, 99, 973–977. [Google Scholar] [CrossRef] [PubMed]
  53. Walker, S.; Wang, C.; Walradt, T.; Hong, B.S.; Tanner, J.R.; Levinsohn, J.L.; Goh, G.; Subtil, A.; Lessin, S.R.; Heymann, W.R.; et al. Identification of a gain-of-function STAT3 mutation (p.Y640F) in lymphocytic variant hypereosinophilic syndrome. Blood 2016, 127, 948–951. [Google Scholar] [CrossRef] [PubMed]
  54. Suchin, K.R.; Cassin, M.; Gottleib, S.L.; Sood, S.; Cucchiara, A.J.; Vonderheid, E.C.; Rook, A.H. Increased interleukin 5 production in eosinophilic Sezary syndrome: Regulation by interferon alfa and interleukin 12. J. Am. Acad. Dermatol. 2001, 44, 28–32. [Google Scholar] [CrossRef]
  55. Olsen, E.; Vonderheid, E.; Pimpinelli, N.; Willemze, R.; Kim, Y.; Knobler, R.; Zackheim, H.; Duvic, M.; Estrach, T.; Lamberg, S.; et al. Revisions to the staging and classification of mycosis fungoides and Sezary syndrome: A proposal of the International Society for Cutaneous Lymphomas (ISCL) and the cutaneous lymphoma task force of the European Organization of Research and Treatment of Cancer (EORTC). Blood 2007, 110, 1713–1722. [Google Scholar] [CrossRef] [Green Version]
  56. Roufosse, F. Peripheral T-cell lymphoma developing after diagnosis of lymphocytic variant hypereosinophilic syndrome: Misdiagnosed lymphoma or natural disease progression? Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2014, 118, 506–510. [Google Scholar] [CrossRef]
  57. Whittaker, S.; Hoppe, R.; Prince, H.M. How I treat mycosis fungoides and Sezary syndrome. Blood 2016, 127, 3142–3153. [Google Scholar] [CrossRef] [Green Version]
  58. Olsen, E.A.; Rook, A.H.; Zic, J.; Kim, Y.; Porcu, P.; Querfeld, C.; Wood, G.; Demierre, M.F.; Pittelkow, M.; Wilson, L.D.; et al. Sezary syndrome: Immunopathogenesis, literature review of therapeutic options, and recommendations for therapy by the United States Cutaneous Lymphoma Consortium (USCLC). J. Am. Acad. Dermatol. 2011, 64, 352–404. [Google Scholar] [CrossRef]
  59. Klion, A.D. How I treat hypereosinophilic syndromes. Blood 2015, 126, 1069–1077. [Google Scholar] [CrossRef] [Green Version]
  60. Khoury, P.; Bochner, B.S. Consultation for Elevated Blood Eosinophils: Clinical Presentations, High Value Diagnostic Tests, and Treatment Options. J. Allergy Clin. Immunol. Pract. 2018, 6, 1446–1453. [Google Scholar] [CrossRef]
  61. Butt, N.M.; Lambert, J.; Ali, S.; Beer, P.A.; Cross, N.C.; Duncombe, A.; Ewing, J.; Harrison, C.N.; Knapper, S.; McLornan, D.; et al. Guideline for the investigation and management of eosinophilia. Br. J. Haematol. 2017, 176, 553–572. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, E.J.; Rook, A.H. Treatment of Sézary syndrome. In UpToDate; Kuzel, T.W., Zic, J.A., Eds.; Wolters Kluwer: Waltham, MA, USA, 2020; Available online: https://www.uptodate.com/contents/treatment-of-sezary-syndrome#H9585818 (accessed on 27 August 2020).
  63. Agar, N.S.; Wedgeworth, E.; Crichton, S.; Mitchell, T.J.; Cox, M.; Ferreira, S.; Robson, A.; Calonje, E.; Stefanato, C.M.; Wain, E.M.; et al. Survival outcomes and prognostic factors in mycosis fungoides/Sezary syndrome: Validation of the revised International Society for Cutaneous Lymphomas/European Organisation for Research and Treatment of Cancer staging proposal. J. Clin. Oncol. 2010, 28, 4730–4739. [Google Scholar] [CrossRef] [PubMed]
  64. Buus, T.B.; Willerslev-Olsen, A.; Fredholm, S.; Blumel, E.; Nastasi, C.; Gluud, M.; Hu, T.; Lindahl, L.M.; Iversen, L.; Fogh, H.; et al. Single-cell heterogeneity in Sezary syndrome. Blood Adv. 2018, 2, 2115–2126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sokolowska-Wojdylo, M.; Wenzel, J.; Gaffal, E.; Lenz, J.; Speuser, P.; Erdmann, S.; Abuzahra, F.; Bowman, E.; Roszkiewicz, J.; Bieber, T.; et al. Circulating clonal CLA(+) and CD4(+) T cells in Sezary syndrome express the skin-homing chemokine receptors CCR4 and CCR10 as well as the lymph node-homing chemokine receptor CCR7. Br. J. Dermatol. 2005, 152, 258–264. [Google Scholar] [CrossRef]
  66. Bernengo, M.G.; Novelli, M.; Quaglino, P.; Lisa, F.; De Matteis, A.; Savoia, P.; Cappello, N.; Fierro, M.T. The relevance of the CD4+ CD26- subset in the identification of circulating Sezary cells. Br. J. Dermatol. 2001, 144, 125–135. [Google Scholar] [CrossRef] [PubMed]
  67. Mishra, A.; La Perle, K.; Kwiatkowski, S.; Sullivan, L.A.; Sams, G.H.; Johns, J.; Curphey, D.P.; Wen, J.; McConnell, K.; Qi, J.; et al. Mechanism, Consequences, and Therapeutic Targeting of Abnormal IL15 Signaling in Cutaneous T-cell Lymphoma. Cancer Discov. 2016, 6, 986–1005. [Google Scholar] [CrossRef] [Green Version]
  68. Wang, L.; Ni, X.; Covington, K.R.; Yang, B.Y.; Shiu, J.; Zhang, X.; Xi, L.; Meng, Q.; Langridge, T.; Drummond, J.; et al. Genomic profiling of Sezary syndrome identifies alterations of key T cell signaling and differentiation genes. Nat. Genet. 2015, 47, 1426–1434. [Google Scholar] [CrossRef] [Green Version]
  69. Woollard, W.J.; Pullabhatla, V.; Lorenc, A.; Patel, V.M.; Butler, R.M.; Bayega, A.; Begum, N.; Bakr, F.; Dedhia, K.; Fisher, J.; et al. Candidate driver genes involved in genome maintenance and DNA repair in Sezary syndrome. Blood 2016, 127, 3387–3397. [Google Scholar] [CrossRef] [Green Version]
  70. Wong, H.K.; Mishra, A.; Hake, T.; Porcu, P. Evolving insights in the pathogenesis and therapy of cutaneous T-cell lymphoma (mycosis fungoides and Sezary syndrome). Br. J. Haematol. 2011, 155, 150–166. [Google Scholar] [CrossRef] [Green Version]
  71. Kari, L.; Loboda, A.; Nebozhyn, M.; Rook, A.H.; Vonderheid, E.C.; Nichols, C.; Virok, D.; Chang, C.; Horng, W.H.; Johnston, J.; et al. Classification and prediction of survival in patients with the leukemic phase of cutaneous T cell lymphoma. J. Exp. Med. 2003, 197, 1477–1488. [Google Scholar] [CrossRef]
  72. Wang, Y.; Su, M.; Zhou, L.L.; Tu, P.; Zhang, X.; Jiang, X.; Zhou, Y. Deficiency of SATB1 expression in Sezary cells causes apoptosis resistance by regulating FasL/CD95L transcription. Blood 2011, 117, 3826–3835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Fanok, M.H.; Sun, A.; Fogli, L.K.; Narendran, V.; Eckstein, M.; Kannan, K.; Dolgalev, I.; Lazaris, C.; Heguy, A.; Laird, M.E.; et al. Role of Dysregulated Cytokine Signaling and Bacterial Triggers in the Pathogenesis of Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2018, 138, 1116–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Borcherding, N.; Voigt, A.P.; Liu, V.; Link, B.K.; Zhang, W.; Jabbari, A. Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression. Clin. Cancer Res. 2019, 25, 2996–3005. [Google Scholar] [CrossRef] [Green Version]
  75. Lee, C.S.; Ungewickell, A.; Bhaduri, A.; Qu, K.; Webster, D.E.; Armstrong, R.; Weng, W.K.; Aros, C.J.; Mah, A.; Chen, R.O.; et al. Transcriptome sequencing in Sezary syndrome identifies Sezary cell and mycosis fungoides-associated lncRNAs and novel transcripts. Blood 2012, 120, 3288–3297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Pomerantz, R.G.; Mirvish, E.D.; Erdos, G.; Falo, L.D., Jr.; Geskin, L.J. Novel approach to gene expression profiling in Sezary syndrome. Br. J. Dermatol. 2010, 163, 1090–1094. [Google Scholar] [CrossRef] [Green Version]
  77. Michel, L.; Jean-Louis, F.; Begue, E.; Bensussan, A.; Bagot, M. Use of PLS3, Twist, CD158k/KIR3DL2, and NKp46 gene expression combination for reliable Sezary syndrome diagnosis. Blood 2013, 121, 1477–1478. [Google Scholar] [CrossRef] [Green Version]
  78. Wysocka, M.; Kossenkov, A.V.; Benoit, B.M.; Troxel, A.B.; Singer, E.; Schaffer, A.; Kim, B.; Dentchev, T.; Nagata, S.; Ise, T.; et al. CD164 and FCRL3 are highly expressed on CD4+CD26- T cells in Sezary syndrome patients. J. Investig. Dermatol. 2014, 134, 229–236. [Google Scholar] [CrossRef] [Green Version]
  79. Liberzon, A.; Subramanian, A.; Pinchback, R.; Thorvaldsdottir, H.; Tamayo, P.; Mesirov, J.P. Molecular signatures database (MSigDB) 3.0. Bioinformatics 2011, 27, 1739–1740. [Google Scholar] [CrossRef]
  80. Stelzer, G.; Rosen, N.; Plaschkes, I.; Zimmerman, S.; Twik, M.; Fishilevich, S.; Stein, T.I.; Nudel, R.; Lieder, I.; Mazor, Y.; et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr. Protoc. Bioinform. 2016, 54, 1.30.1–1.30.33. [Google Scholar] [CrossRef]
  81. Gibson, H.M.; Mishra, A.; Chan, D.V.; Hake, T.S.; Porcu, P.; Wong, H.K. Impaired proteasome function activates GATA3 in T cells and upregulates CTLA-4: Relevance for Sezary syndrome. J. Investig. Dermatol. 2013, 133, 249–257. [Google Scholar] [CrossRef] [Green Version]
  82. Geskin, L.J.; Viragova, S.; Stolz, D.B.; Fuschiotti, P. Interleukin-13 is overexpressed in cutaneous T-cell lymphoma cells and regulates their proliferation. Blood 2015, 125, 2798–2805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Fredholm, S.; Willerslev-Olsen, A.; Met, O.; Kubat, L.; Gluud, M.; Mathiasen, S.L.; Friese, C.; Blumel, E.; Petersen, D.L.; Hu, T.; et al. SATB1 in Malignant T Cells. J. Investig. Dermatol. 2018, 138, 1805–1815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Yoshie, O.; Matsushima, K. CCR4 and its ligands: From bench to bedside. Int. Immunol. 2015, 27, 11–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Cristofoletti, C.; Bresin, A.; Picozza, M.; Picchio, M.C.; Monzo, F.; Helmer Citterich, M.; Passarelli, F.; Frezzolini, A.; Scala, E.; Monopoli, A.; et al. Blood and skin-derived Sezary cells: Differences in proliferation-index, activation of PI3K/AKT/mTORC1 pathway and its prognostic relevance. Leukemia 2019, 33, 1231–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Duvic, M.; Pinter-Brown, L.C.; Foss, F.M.; Sokol, L.; Jorgensen, J.L.; Challagundla, P.; Dwyer, K.M.; Zhang, X.; Kurman, M.R.; Ballerini, R.; et al. Phase 1/2 study of mogamulizumab, a defucosylated anti-CCR4 antibody, in previously treated patients with cutaneous T-cell lymphoma. Blood 2015, 125, 1883–1889. [Google Scholar] [CrossRef]
  87. Kitadate, A.; Ikeda, S.; Abe, F.; Takahashi, N.; Shimizu, N.; Matsue, K.; Tagawa, H. Histone deacetylase inhibitors downregulate CCR4 expression and decrease mogamulizumab efficacy in CCR4-positive mature T-cell lymphomas. Haematologica 2018, 103, 126–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Richardson, S.K.; Newton, S.B.; Bach, T.L.; Budgin, J.B.; Benoit, B.M.; Lin, J.H.; Yoon, J.S.; Wysocka, M.; Abrams, C.S.; Rook, A.H. Bexarotene blunts malignant T-cell chemotaxis in Sezary syndrome: Reduction of chemokine receptor 4-positive lymphocytes and decreased chemotaxis to thymus and activation-regulated chemokine. Am. J. Hematol. 2007, 82, 792–797. [Google Scholar] [CrossRef]
  89. Grzanka, D.; Gagat, M.; Izdebska, M.; Marszalek, A. Expression of special AT-rich sequence-binding protein 1 is an independent prognostic factor in cutaneous T-cell lymphoma. Oncol. Rep. 2015, 33, 250–266. [Google Scholar] [CrossRef] [Green Version]
  90. Renema, N.; Navet, B.; Heymann, M.F.; Lezot, F.; Heymann, D. RANK-RANKL signalling in cancer. Biosci. Rep. 2016, 36. [Google Scholar] [CrossRef] [Green Version]
  91. Boonk, S.E.; Zoutman, W.H.; Putter, H.; Ram-Wolff, C.; Felcht, M.; Klemke, C.D.; Ranki, A.; Quaglino, P.; Whittaker, S.; Bagot, M.; et al. Increased Expression of PLS3 Correlates with Better Outcome in Sezary Syndrome. J. Investig. Dermatol. 2017, 137, 754–757. [Google Scholar] [CrossRef] [Green Version]
  92. Zhang, Z.; Chen, C.; Guo, W.; Zheng, S.; Sun, Z.; Geng, X. DNM3 Attenuates Hepatocellular Carcinoma Growth by Activating P53. Med. Sci. Monit. 2016, 22, 197–205. [Google Scholar] [CrossRef] [PubMed]
  93. Osthus, R.C.; Karim, B.; Prescott, J.E.; Smith, B.D.; McDevitt, M.; Huso, D.L.; Dang, C.V. The Myc target gene JPO1/CDCA7 is frequently overexpressed in human tumors and has limited transforming activity in vivo. Cancer Res. 2005, 65, 5620–5627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Jimenez, P.R.; Martin-Cortazar, C.; Kourani, O.; Chiodo, Y.; Cordoba, R.; Dominguez-Franjo, M.P.; Redondo, J.M.; Iglesias, T.; Campanero, M.R. CDCA7 is a critical mediator of lymphomagenesis that selectively regulates anchorage-independent growth. Haematologica 2018, 103, 1669–1678. [Google Scholar] [CrossRef] [PubMed]
  95. Martin-Cortazar, C.; Chiodo, Y.; Jimenez, P.R.; Bernabe, M.; Cayuela, M.L.; Iglesias, T.; Campanero, M.R. CDCA7 finely tunes cytoskeleton dynamics to promote lymphoma migration and invasion. Haematologica 2019. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, A.; Ho, C.S.; Ponzielli, R.; Barsyte-Lovejoy, D.; Bouffet, E.; Picard, D.; Hawkins, C.E.; Penn, L.Z. Identification of a novel c-Myc protein interactor, JPO2, with transforming activity in medulloblastoma cells. Cancer Res. 2005, 65, 5607–5619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Begue, E.; Jean-Louis, F.; Bagot, M.; Jauliac, S.; Cayuela, J.M.; Laroche, L.; Parquet, N.; Bachelez, H.; Bensussan, A.; Courtois, G.; et al. Inducible expression and pathophysiologic functions of T-plastin in cutaneous T-cell lymphoma. Blood 2012, 120, 143–154. [Google Scholar] [CrossRef] [Green Version]
  98. Jones, C.L.; Ferreira, S.; McKenzie, R.C.; Tosi, I.; Caesar, J.A.; Bagot, M.; Whittaker, S.J.; Mitchell, T.J. Regulation of T-plastin expression by promoter hypomethylation in primary cutaneous T-cell lymphoma. J. Investig. Dermatol. 2012, 132, 2042–2049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Henn, A.; Michel, L.; Fite, C.; Deschamps, L.; Ortonne, N.; Ingen-Housz-Oro, S.; Marinho, E.; Beylot-Barry, M.; Bagot, M.; Laroche, L.; et al. Sezary syndrome without erythroderma. J. Am. Acad. Dermatol. 2015, 72, 1003–1009 e1001. [Google Scholar] [CrossRef]
  100. Su, M.W.; Dorocicz, I.; Dragowska, W.H.; Ho, V.; Li, G.; Voss, N.; Gascoyne, R.; Zhou, Y. Aberrant expression of T-plastin in Sezary cells. Cancer Res. 2003, 63, 7122–7127. [Google Scholar]
  101. Yokobori, T.; Iinuma, H.; Shimamura, T.; Imoto, S.; Sugimachi, K.; Ishii, H.; Iwatsuki, M.; Ota, D.; Ohkuma, M.; Iwaya, T.; et al. Plastin3 is a novel marker for circulating tumor cells undergoing the epithelial-mesenchymal transition and is associated with colorectal cancer prognosis. Cancer Res. 2013, 73, 2059–2069. [Google Scholar] [CrossRef] [Green Version]
  102. Ueo, H.; Sugimachi, K.; Gorges, T.M.; Bartkowiak, K.; Yokobori, T.; Muller, V.; Shinden, Y.; Ueda, M.; Ueo, H.; Mori, M.; et al. Circulating tumour cell-derived plastin3 is a novel marker for predicting long-term prognosis in patients with breast cancer. Br. J. Cancer 2015, 112, 1519–1526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Zhao, Z.; Rahman, M.A.; Chen, Z.G.; Shin, D.M. Multiple biological functions of Twist1 in various cancers. Oncotarget 2017, 8, 20380–20393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Norozi, F.; Ahmadzadeh, A.; Shahjahani, M.; Shahrabi, S.; Saki, N. Twist as a new prognostic marker in hematological malignancies. Clin. Transl. Oncol. 2016, 18, 113–124. [Google Scholar] [CrossRef] [PubMed]
  105. Goswami, M.; Duvic, M.; Dougherty, A.; Ni, X. Increased Twist expression in advanced stage of mycosis fungoides and Sezary syndrome. J. Cutan. Pathol. 2012, 39, 500–507. [Google Scholar] [CrossRef] [PubMed]
  106. Beck, B.; Lapouge, G.; Rorive, S.; Drogat, B.; Desaedelaere, K.; Delafaille, S.; Dubois, C.; Salmon, I.; Willekens, K.; Marine, J.C.; et al. Different levels of Twist1 regulate skin tumor initiation, stemness, and progression. Cell Stem Cell 2015, 16, 67–79. [Google Scholar] [CrossRef] [Green Version]
  107. Pham, D.; Vincentz, J.W.; Firulli, A.B.; Kaplan, M.H. Twist1 regulates Ifng expression in Th1 cells by interfering with Runx3 function. J. Immunol. 2012, 189, 832–840. [Google Scholar] [CrossRef] [Green Version]
  108. Niesner, U.; Albrecht, I.; Janke, M.; Doebis, C.; Loddenkemper, C.; Lexberg, M.H.; Eulenburg, K.; Kreher, S.; Koeck, J.; Baumgrass, R.; et al. Autoregulation of Th1-mediated inflammation by twist1. J. Exp. Med. 2008, 205, 1889–1901. [Google Scholar] [CrossRef]
  109. Chong, B.F.; Dantzer, P.; Germeroth, T.; Hafner, M.; Wilson, A.J.; Xiao, G.; Wong, H.K. Induced Sezary syndrome PBMCs poorly express immune response genes up-regulated in stimulated memory T cells. J. Dermatol. Sci. 2010, 60, 8–20. [Google Scholar] [CrossRef] [Green Version]
  110. Wong, H.K.; Gibson, H.; Hake, T.; Geyer, S.; Frederickson, J.; Marcucci, G.; Caligiuri, M.A.; Porcu, P.; Mishra, A. Promoter-Specific Hypomethylation Is Associated with Overexpression of PLS3, GATA6, and TWIST1 in the Sezary Syndrome. J. Investig. Dermatol. 2015, 135, 2084–2092. [Google Scholar] [CrossRef] [Green Version]
  111. Vermeer, M.H.; van Doorn, R.; Dijkman, R.; Mao, X.; Whittaker, S.; van Voorst Vader, P.C.; Gerritsen, M.J.; Geerts, M.L.; Gellrich, S.; Soderberg, O.; et al. Novel and highly recurrent chromosomal alterations in Sezary syndrome. Cancer Res. 2008, 68, 2689–2698. [Google Scholar] [CrossRef] [Green Version]
  112. Showe, L.C.; Fox, F.E.; Williams, D.; Au, K.; Niu, Z.; Rook, A.H. Depressed IL-12-mediated signal transduction in T cells from patients with Sezary syndrome is associated with the absence of IL-12 receptor beta 2 mRNA and highly reduced levels of STAT4. J. Immunol. 1999, 163, 4073–4079. [Google Scholar] [PubMed]
  113. Litvinov, I.V.; Cordeiro, B.; Fredholm, S.; Odum, N.; Zargham, H.; Huang, Y.; Zhou, Y.; Pehr, K.; Kupper, T.S.; Woetmann, A.; et al. Analysis of STAT4 expression in cutaneous T-cell lymphoma (CTCL) patients and patient-derived cell lines. Cell Cycle 2014, 13, 2975–2982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Andrews, J.M.; Schmidt, J.A.; Carson, K.R.; Musiek, A.C.; Mehta-Shah, N.; Payton, J.E. Novel cell adhesion/migration pathways are predictive markers of HDAC inhibitor resistance in cutaneous T cell lymphoma. EBioMedicine 2019, 46, 170–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kopp, K.L.; Ralfkiaer, U.; Gjerdrum, L.M.; Helvad, R.; Pedersen, I.H.; Litman, T.; Jonson, L.; Hagedorn, P.H.; Krejsgaard, T.; Gniadecki, R.; et al. STAT5-mediated expression of oncogenic miR-155 in cutaneous T-cell lymphoma. Cell Cycle 2013, 12, 1939–1947. [Google Scholar] [CrossRef]
  116. Moyal, L.; Yehezkel, S.; Gorovitz, B.; Keren, A.; Gilhar, A.; Lubin, I.; Sherman, S.; Hodak, E. Oncogenic role of microRNA-155 in mycosis fungoides: An In Vitro and xenograft mouse model study. Br. J. Dermatol. 2017, 177, 791–800. [Google Scholar] [CrossRef]
  117. Ralfkiaer, U.; Hagedorn, P.H.; Bangsgaard, N.; Lovendorf, M.B.; Ahler, C.B.; Svensson, L.; Kopp, K.L.; Vennegaard, M.T.; Lauenborg, B.; Zibert, J.R.; et al. Diagnostic microRNA profiling in cutaneous T-cell lymphoma (CTCL). Blood 2011, 118, 5891–5900. [Google Scholar] [CrossRef]
  118. Tili, E.; Michaille, J.J.; Wernicke, D.; Alder, H.; Costinean, S.; Volinia, S.; Croce, C.M. Mutator activity induced by microRNA-155 (miR-155) links inflammation and cancer. Proc. Natl. Acad. Sci. USA 2011, 108, 4908–4913. [Google Scholar] [CrossRef] [Green Version]
  119. Witten, L.; Slack, F.J. miR-155 as a novel clinical target for hematological malignancies. Carcinogenesis 2020, 41, 2–7. [Google Scholar] [CrossRef]
  120. Kasprzycka, M.; Zhang, Q.; Witkiewicz, A.; Marzec, M.; Potoczek, M.; Liu, X.; Wang, H.Y.; Milone, M.; Basu, S.; Mauger, J.; et al. Gamma c-signaling cytokines induce a regulatory T cell phenotype in malignant CD4+ T lymphocytes. J. Immunol. 2008, 181, 2506–2512. [Google Scholar] [CrossRef] [Green Version]
  121. Krejsgaard, T.; Odum, N.; Geisler, C.; Wasik, M.A.; Woetmann, A. Regulatory T cells and immunodeficiency in mycosis fungoides and Sezary syndrome. Leukemia 2012, 26, 424–432. [Google Scholar] [CrossRef] [Green Version]
  122. Jariwala, N.; Benoit, B.; Kossenkov, A.V.; Oetjen, L.K.; Whelan, T.M.; Cornejo, C.M.; Takeshita, J.; Kim, B.S.; Showe, L.C.; Wysocka, M.; et al. TIGIT and Helios Are Highly Expressed on CD4(+) T Cells in Sezary Syndrome Patients. J. Investig. Dermatol. 2017, 137, 257–260. [Google Scholar] [CrossRef] [Green Version]
  123. Heid, J.B.; Schmidt, A.; Oberle, N.; Goerdt, S.; Krammer, P.H.; Suri-Payer, E.; Klemke, C.D. FOXP3+CD25- tumor cells with regulatory function in Sezary syndrome. J. Investig. Dermatol. 2009, 129, 2875–2885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Capriotti, E.; Vonderheid, E.C.; Thoburn, C.J.; Wasik, M.A.; Bahler, D.W.; Hess, A.D. Expression of T-plastin, FoxP3 and other tumor-associated markers by leukemic T-cells of cutaneous T-cell lymphoma. Leuk. Lymphoma 2008, 49, 1190–1201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Querfeld, C.; Leung, S.; Myskowski, P.L.; Curran, S.A.; Goldman, D.A.; Heller, G.; Wu, X.; Kil, S.H.; Sharma, S.; Finn, K.J.; et al. Primary T Cells from Cutaneous T-cell Lymphoma Skin Explants Display an Exhausted Immune Checkpoint Profile. Cancer Immunol. Res. 2018, 6, 900–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Swainson, L.A.; Mold, J.E.; Bajpai, U.D.; McCune, J.M. Expression of the autoimmune susceptibility gene FcRL3 on human regulatory T cells is associated with dysfunction and high levels of programmed cell death-1. J. Immunol. 2010, 184, 3639–3647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Joller, N.; Lozano, E.; Burkett, P.R.; Patel, B.; Xiao, S.; Zhu, C.; Xia, J.; Tan, T.G.; Sefik, E.; Yajnik, V.; et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 2014, 40, 569–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Harjunpaa, H.; Guillerey, C. TIGIT as an emerging immune checkpoint. Clin. Exp. Immunol. 2019. [Google Scholar] [CrossRef] [Green Version]
  129. Bin Dhuban, K.; d’Hennezel, E.; Nashi, E.; Bar-Or, A.; Rieder, S.; Shevach, E.M.; Nagata, S.; Piccirillo, C.A. Coexpression of TIGIT and FCRL3 identifies Helios+ human memory regulatory T cells. J. Immunol. 2015, 194, 3687–3696. [Google Scholar] [CrossRef] [Green Version]
  130. Anzengruber, F.; Ignatova, D.; Schlaepfer, T.; Chang, Y.T.; French, L.E.; Pascolo, S.; Contassot, E.; Bobrowicz, M.; Hoetzenecker, W.; Guenova, E. Divergent LAG-3 versus BTLA, TIGIT, and FCRL3 expression in Sezary syndrome. Leuk. Lymphoma 2019, 60, 1899–1907. [Google Scholar] [CrossRef] [Green Version]
  131. Aliahmad, P.; Kaye, J. Development of all CD4 T lineages requires nuclear factor TOX. J. Exp. Med. 2008, 205, 245–256. [Google Scholar] [CrossRef]
  132. Ando, M.; Ito, M.; Srirat, T.; Kondo, T.; Yoshimura, A. Memory T cell, exhaustion, and tumor immunity. Immunol. Med. 2019, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Huang, Y.; Su, M.W.; Jiang, X.; Zhou, Y. Evidence of an oncogenic role of aberrant TOX activation in cutaneous T-cell lymphoma. Blood 2015, 125, 1435–1443. [Google Scholar] [CrossRef]
  134. Lefrancois, P.; Xie, P.; Wang, L.; Tetzlaff, M.T.; Moreau, L.; Watters, A.K.; Netchiporouk, E.; Provost, N.; Gilbert, M.; Ni, X.; et al. Gene expression profiling and immune cell-type deconvolution highlight robust disease progression and survival markers in multiple cohorts of CTCL patients. Oncoimmunology 2018, 7, e1467856. [Google Scholar] [CrossRef] [Green Version]
  135. Narducci, M.G.; Arcelli, D.; Picchio, M.C.; Lazzeri, C.; Pagani, E.; Sampogna, F.; Scala, E.; Fadda, P.; Cristofoletti, C.; Facchiano, A.; et al. MicroRNA profiling reveals that miR-21, miR486 and miR-214 are upregulated and involved in cell survival in Sezary syndrome. Cell Death Dis. 2011, 2, e151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Tessema, M.; Yingling, C.M.; Picchi, M.A.; Wu, G.; Ryba, T.; Lin, Y.; Bungum, A.O.; Edell, E.S.; Spira, A.; Belinsky, S.A. ANK1 Methylation regulates expression of MicroRNA-486-5p and discriminates lung tumors by histology and smoking status. Cancer Lett. 2017, 410, 191–200. [Google Scholar] [CrossRef]
  137. Bianchi, E.; Bulgarelli, J.; Ruberti, S.; Rontauroli, S.; Sacchi, G.; Norfo, R.; Pennucci, V.; Zini, R.; Salati, S.; Prudente, Z.; et al. MYB controls erythroid versus megakaryocyte lineage fate decision through the miR-486-3p-mediated downregulation of MAF. Cell Death Differ. 2015, 22, 1906–1921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Hall, A.E.; Lu, W.T.; Godfrey, J.D.; Antonov, A.V.; Paicu, C.; Moxon, S.; Dalmay, T.; Wilczynska, A.; Muller, P.A.; Bushell, M. The cytoskeleton adaptor protein ankyrin-1 is upregulated by p53 following DNA damage and alters cell migration. Cell Death Dis. 2016, 7, e2184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Sun, Z.; Yan, B. Multiple roles and regulatory mechanisms of the transcription factor GATA6 in human cancers. Clin. Genet. 2020, 97, 64–72. [Google Scholar] [CrossRef] [Green Version]
  140. Kamijo, H.; Miyagaki, T.; Shishido-Takahashi, N.; Nakajima, R.; Oka, T.; Suga, H.; Sugaya, M.; Sato, S. Aberrant CD137 ligand expression induced by GATA6 overexpression promotes tumor progression in cutaneous T-cell lymphoma. Blood 2018, 132, 1922–1935. [Google Scholar] [CrossRef] [Green Version]
  141. Parra, M. Class IIa HDACs—New insights into their functions in physiology and pathology. FEBS J. 2015, 282, 1736–1744. [Google Scholar] [CrossRef]
  142. Gil, V.S.; Bhagat, G.; Howell, L.; Zhang, J.; Kim, C.H.; Stengel, S.; Vega, F.; Zelent, A.; Petrie, K. Deregulated expression of HDAC9 in B cells promotes development of lymphoproliferative disease and lymphoma in mice. Dis. Model Mech. 2016, 9, 1483–1495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Hutt, D.M.; Roth, D.M.; Vignaud, H.; Cullin, C.; Bouchecareilh, M. The histone deacetylase inhibitor, Vorinostat, represses hypoxia inducible factor 1 alpha expression through translational inhibition. PLoS ONE 2014, 9, e106224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Linares, A.; Assou, S.; Lapierre, M.; Thouennon, E.; Duraffourd, C.; Fromaget, C.; Boulahtouf, A.; Tian, G.; Ji, J.; Sahin, O.; et al. Increased expression of the HDAC9 gene is associated with antiestrogen resistance of breast cancers. Mol. Oncol. 2019, 13, 1534–1547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Milde, T.; Oehme, I.; Korshunov, A.; Kopp-Schneider, A.; Remke, M.; Northcott, P.; Deubzer, H.E.; Lodrini, M.; Taylor, M.D.; von Deimling, A.; et al. HDAC5 and HDAC9 in medulloblastoma: Novel markers for risk stratification and role in tumor cell growth. Clin. Cancer Res. 2010, 16, 3240–3252. [Google Scholar] [CrossRef] [Green Version]
  146. Stoeger, T.; Gerlach, M.; Morimoto, R.I.; Nunes Amaral, L.A. Large-scale investigation of the reasons why potentially important genes are ignored. PLoS Biol. 2018, 16, e2006643. [Google Scholar] [CrossRef]
Figure 1. Clinical criteria for staging of mycosis fungoides (MF) and Sézary syndrome (SS). SS is classified as stage IVA1 disease and is distinguished from MF by a high blood tumor burden. Diagnostic “B2” blood criteria required for SS include Sézary cells ≥ 1000 cells/μL, CD4/CD8 ≥ 10, CD4+CD7 cells > 30% or CD4+CD7 cells ≥ 40%, with an identical T-cell clone detected in blood and skin. Stage IIIB requires B1 blood involvement not meeting the B2 threshold for SS. The most distinguishing qualifier for each stage is noted. The table is adapted from the ISCL/European Organization for Research and Treatment of Cancer (EORTC) classification criteria in Olsen et al. [33]. BSA, body surface area affected. Erythroderma, BSA > 80%.
Figure 1. Clinical criteria for staging of mycosis fungoides (MF) and Sézary syndrome (SS). SS is classified as stage IVA1 disease and is distinguished from MF by a high blood tumor burden. Diagnostic “B2” blood criteria required for SS include Sézary cells ≥ 1000 cells/μL, CD4/CD8 ≥ 10, CD4+CD7 cells > 30% or CD4+CD7 cells ≥ 40%, with an identical T-cell clone detected in blood and skin. Stage IIIB requires B1 blood involvement not meeting the B2 threshold for SS. The most distinguishing qualifier for each stage is noted. The table is adapted from the ISCL/European Organization for Research and Treatment of Cancer (EORTC) classification criteria in Olsen et al. [33]. BSA, body surface area affected. Erythroderma, BSA > 80%.
Cells 09 01992 g001
Figure 2. Overlap of differentially expressed genes for L-HES and SS. Shared and unique gene expression in SS and L-HES are shown with relationships to their respective roles in disease, which may lead to diagnostic improvements. Red circle: Differential gene expression in SS CD3+CD4+CD45RO+ T cells compared to normal CD3+CD4+CD45RO+ T cells [22]. Blue circle: Differential gene expression in L-HES CD3CD4+ T cells compared to normal CD3+CD4+ T cells [30]. Center overlap: Gene expression abnormalities shared by SS and L-HES may reflect benign or pre-neoplastic proliferative and inflammatory phenotypes. Excluded areas to the right and left represent abnormal gene expression unique to SS or L-HES, respectively. SS-unique genes have been associated with a number of cancer-promoting phenotypes.
Figure 2. Overlap of differentially expressed genes for L-HES and SS. Shared and unique gene expression in SS and L-HES are shown with relationships to their respective roles in disease, which may lead to diagnostic improvements. Red circle: Differential gene expression in SS CD3+CD4+CD45RO+ T cells compared to normal CD3+CD4+CD45RO+ T cells [22]. Blue circle: Differential gene expression in L-HES CD3CD4+ T cells compared to normal CD3+CD4+ T cells [30]. Center overlap: Gene expression abnormalities shared by SS and L-HES may reflect benign or pre-neoplastic proliferative and inflammatory phenotypes. Excluded areas to the right and left represent abnormal gene expression unique to SS or L-HES, respectively. SS-unique genes have been associated with a number of cancer-promoting phenotypes.
Cells 09 01992 g002
Figure 3. Differentially identified genes from the meta-analysis of SS and L-HES are supported by prior SS studies. Gene expression results from Moerman-Herzog et al. were compared to prior transcriptomic profiling studies of SS (Table 3). Genes differentially expressed from SS of prior studies were identified from the manuscript and supplementary data, using the significance threshold defined by each study. Gene symbols were updated using the Molecular Signatures database [79] and/or the GeneCards database [80]. Gene groups are defined by expression pattern, (A) upregulated SS-unique genes, (B) downregulated SS-unique genes, (C) upregulated shared genes, (D) downregulated shared genes. Only genes reported in at least three studies are shown. For each gene, studies that reported significant differential expression for that gene are represented by color-coded boxes next to the gene symbol.
Figure 3. Differentially identified genes from the meta-analysis of SS and L-HES are supported by prior SS studies. Gene expression results from Moerman-Herzog et al. were compared to prior transcriptomic profiling studies of SS (Table 3). Genes differentially expressed from SS of prior studies were identified from the manuscript and supplementary data, using the significance threshold defined by each study. Gene symbols were updated using the Molecular Signatures database [79] and/or the GeneCards database [80]. Gene groups are defined by expression pattern, (A) upregulated SS-unique genes, (B) downregulated SS-unique genes, (C) upregulated shared genes, (D) downregulated shared genes. Only genes reported in at least three studies are shown. For each gene, studies that reported significant differential expression for that gene are represented by color-coded boxes next to the gene symbol.
Cells 09 01992 g003
Table 1. Shared and distinct features of Sézary syndrome and disease controls.
Table 1. Shared and distinct features of Sézary syndrome and disease controls.
Disease TypeSkin InflammationLympho-ProliferationMalignancy
Sézary syndromeTh2clonal+
L-HESTh2frequently clonal
Atopic DermatitisTh2reactive
PsoriasisTh1, Th17reactive
Contact DermatitisTh1, Th2 or Th17 reactive
L-HES, lymphocytic-variant hypereosinophilic syndrome; Th1, T-helper type 1; Th2, T-helper type 2; Th17, T-helper type 17.
Table 2. Clinical and molecular features of Sézary syndrome and lymphocytic-variant hypereosinophilic syndrome (HES).
Table 2. Clinical and molecular features of Sézary syndrome and lymphocytic-variant hypereosinophilic syndrome (HES).
Clinical FeaturesSézary SyndromeLymphocytic-Variant HES
Classificationlymphoma, stage IVbenign lymphoproliferation
Diagnostic criteriaSézary cells > 1000/µL (or, CD4/CD8 ratio ≥ 10, CD4+CD7 cells ≥ 40%, CD4+CD26 cells ≥ 30%), with identical T-cell clone in blood + skin [6,55]Rule out other causes of HES. Blood eosinophilia >1500/µL, abnormal T cells with no standardized threshold, frequent T-cell clonality, T cells secrete IL-5 [25,35,56].
Clinical coursemoderately aggressiveindolent
Skin and physical symptomswidespread erythroderma, pruritus, lymphadenopathylimited erythroderma, urticaria, pruritus [24,27]
Residence of T cellsblood, skin, lymph nodeblood, skin, lymph node, soft tissue [25]
Skin pathologyepidermotropic Sézary cells with cerebriform nuclei, eosinophils in some patientsabundant eosinophils, and perivascular, dermal infiltrate of small-medium size T cells with irregular nucleus and scarce cytoplasm [25,35]
Eosinophiliasome patients, late, moderate complications uncommon [29] >700/µL poor prognostic indicator [46]all patients, early, severe, can cause organ damage
First line therapySystemic immunomodulation: ECP combined with interferons or other systemic (bexarotene, romidepsin, low dose methotrexate) and/or skin-directed (topicals, TSEBT) therapy [57,58]systemic corticosteroids [35,59,60]
Second line therapyTargeted and immune enhancing/sparing therapies preferred: mogamulizumab. romidepsin, alemtuzumab, intermediate dose methotrexate. Refractory disease: clinical trials, allogeneic HCT, chemotherapy [57,58]IFN-α + glucocorticoids
Steroid-sparing: mepolizumab, alemtuzumab, mycophenolate mofetil, cyclosporin, methotrexate, JAK kinase inhibitors (ruxolitinib, tofacitinib) [35,59,60] and imatinib, despite lack of FIP1L1-PDFGRA fusion, for patients who fail other tharapies [61]
Follow upMonitor complete blood count with differential, liver function, LDH, flow cytometry for Sezary cells [33] in blood, physical examination for nodes, viscera and skin infections [62].monitor T-cell lymphoma risk with lymphocyte counts, PB flow cytometry, BM cytogenetics [59]
Progression symptomsIncreases in pruritus, erythroderma, or skin tumor burden, enlarging lymph nodes, visceral organ involvement, immune suppression [63]10–25% progress to T-cell lymphoma, cytogenetic changes
Nonspecific symptoms: rapid increase in lymphocytosis, lymph node involvement, infiltrative nodules [25]
Molecular FeaturesSézary SyndromeLymphocytic-VariantHES
T-cell phenotypememory T cell with heterogeneous molecular phenotype [43,64]memory T cell [30,42]
T-cell surface antigensCD3+/−CD4+, CD7 and/or CD26 loss
CLA+, CCR7+, CCR4+, CCR10+ [65,66]
CD3CD4+CD7CD5++, CD3+CD4+CD7, or, CD3+CD4CD8 [23,35]
CytokinesTh2 (IL-4, IL-5, IL-13), suppressive (IL-10), autocrine or paracrine growth stimulation (IL-15, IL-16, IL-32) [67,68]Th2 (IL-4, IL-5, IL-13) [28,42]
Molecular driversMutations in pathways related to DNA damage repair (TP53), apoptosis, (FAS), cell cycle (MYC, RB1), epigenetic modulators (DNMT3A, TET2), JAK/STAT (JAK3, STAT3, STAT5B), ARID1A, NF-κB (NFKB2, CARD11), TCR-signaling (CD28, PLCG1) [37,38,69,70]IL-5, GATA3, JAK/STAT, IL17RB, TGFβ signaling [30,53]
Genetic abnormalitiesFrequent SNV and CNV, C > T transitions consistent with UV damage, recurrent 10q and 17p deletions, recurrent 8q and 17q amplifications [37,38]; gene fusions [68]Seldom reported, partial 6q deletion and other karyotype abnormalities [23]
BM, bone marrow; CNV, copy number variation; ECP, extracorporeal photopheresis; HCT, hematopoietic stem cell transplant; LDH, lactate dehydrogenase; ND, normal donor; PB, peripheral blood; PUVA, psoralen plus ultraviolet A; SNV, single nucleotide variation; TCR, T-cell receptor; TSEBT, total skin electron beam therapy.
Table 3. SS transcriptomic profiling studies included in Figure 3.
Table 3. SS transcriptomic profiling studies included in Figure 3.
Meta-Analysis StudySézary PatientsHealthy DonorsBIDTechnology
Moerman-Herzog et al. [22]n = 3
CD3+CD4+CD45RO+
n = 3
CD3+CD4+CD45RO+
n.a.microarray
Prior StudySézary PatientsHealthy DonorsBIDTechnology
Fanok et al. [73]n = 8 CD3+CD4+CD7
and/or CD3+CD4+CD26
n =4 CD3+CD4+CD45RO+n.a.RNAseq
Wang et al. [68]n = 22 CD3+CD4+n = 5 CD3+CD4+n.a.RNAseq
Wysocka et al. [78]n = 6 CD3+CD4+n = 3 CD3+CD4+n.a.microarray
Wang et al. [72]n = 6 CD3+CD4+CD7-n = 9 CD3+CD4+n.a.microarray
Booken et al. [16]n = 10 PBMCn =10 PBMCn.a.microarray
Hahtola et al. [17]n = 4 PBMCn = 5 PBMCn.a.microarray
van Doorn et al. [14]n = 10 CD3+CD4+n = 3 CD3+CD4+n = 5 CD3+CD4+microarray
Kari et al. [71]n = 18
>60% CD4+
n = 12
Th2-skewed PBMC
n.a.microarray
Prior StudySézary Malignant CellsPatient-Matched
Non-Malignant Cells
BIDTechnology
Borcherding et al. [74]n = 1
CD3+CD4+CD5brightSSChi
n = 1
CD3+CD4+CD5intSSCint
n.a.scRNAseq
Lee et al. [75]n = 3
CD3+CD4++
n = 3
CD3+CD4+
n.a.RNAseq
n.a., not applicable; RNAseq, RNA sequencing; scRNAseq, single cell RNAseq.

Share and Cite

MDPI and ACS Style

Moerman-Herzog, A.; Mehdi, S.J.; Wong, H.K. Gene Expression Comparison between Sézary Syndrome and Lymphocytic-Variant Hypereosinophilic Syndrome Refines Biomarkers for Sézary Syndrome. Cells 2020, 9, 1992. https://doi.org/10.3390/cells9091992

AMA Style

Moerman-Herzog A, Mehdi SJ, Wong HK. Gene Expression Comparison between Sézary Syndrome and Lymphocytic-Variant Hypereosinophilic Syndrome Refines Biomarkers for Sézary Syndrome. Cells. 2020; 9(9):1992. https://doi.org/10.3390/cells9091992

Chicago/Turabian Style

Moerman-Herzog, Andrea, Syed J. Mehdi, and Henry K. Wong. 2020. "Gene Expression Comparison between Sézary Syndrome and Lymphocytic-Variant Hypereosinophilic Syndrome Refines Biomarkers for Sézary Syndrome" Cells 9, no. 9: 1992. https://doi.org/10.3390/cells9091992

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