Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 30.
Published in final edited form as: Cytometry B Clin Cytom. 2018 Oct 16;96(3):234–241. doi: 10.1002/cyto.b.21738

Naïve/Memory T-Cell Phenotypes in Leukemic Cutaneous T-Cell Lymphoma: Putative Cell of Origin Overlaps Disease Classification

Pedro Horna 1, Lynn C Moscinski 2, Lubomir Sokol 3, Haipeng Shao 2,*
PMCID: PMC7703846  NIHMSID: NIHMS1647344  PMID: 30328260

Abstract

Background:

Mycosis fungoides (MF) and Sézary Syndrome (SS) are clinically distinct cutaneous T-cell lymphomas with strikingly similar morphologic and phenotypic features. Prior studies have suggested phenotypic differences based on markers of antigen experience, suggesting a different cell of origin.

Methods:

Seventy-nine involved peripheral blood or bone marrow samples from 33 patients with SS and 19 patients with MF were studied by 10-color flow cytometry, including CD62L, CD45RA, CCR4, and PD-1. Gated tumor events were classified as naïve (TN), central memory (TCM), effector memory (TEM), or effector memory with reacquired CD45RA (TEMRA); based on CD62L+/CD45RA+, CD62L+/CD45RA, CD62L/CD45RA, or CD62L/CD45RA+ phenotype, respectively. Sequential specimens were compared to assess for phenotypic stability.

Results:

The naïve/memory phenotype of the neoplastic T-cells was markedly heterogeneous, with a dominant TN, TCM, TEM, or TEMRA subset on 11 (14%), 32 (41%), 30 (38%), and 6 (8%) cases, respectively. There was no correlation between the diagnosis of MF or SS and putative cell of origin (P = 0.4). Overexpression of CCR4 and PD1 was observed in most cases, with higher intensity in SS compared to MF. The naïve/memory phenotype remained the same for 10 patients up to 273 days after the initial analysis; while on six patients, the naïve/memory phenotype was different from the original phenotype.

Conclusions:

Both SS and MF can have phenotypic features of any of the major naïve/memory T-cell subsets, which questions the current principle of “cell-of-origin” distinction between SS and MF. Phenotypic shifts within these subsets are common, suggesting a functional state rather than a cell-of-origin surrogate.

Key terms: memory T-cells, naïve T-cells, Sézary syndrome, mycosis fungoides, flow cytometry

INTRODUCTION

Mycosis fungoides (MF) and Sézary syndrome (SS) are two clinically distinct cutaneous T-cell lymphomas (CTCLs) that share marked morphologic and phenotypic similarities, despite distinctively different clinical presentations. MF is the most common CTCL, with an incidence of 0.36–0.55 per 100,000 individuals in the United States (1,2), and characterized clinically by slowly progressive patch, plaque and tumor lesions (3). A subset of patients with MF progresses to more aggressive phase of the disease with large cell transformation or leukemic MF. In contrast, SS is a rare CTCL with an incidence of 0.01 per 100,000 individuals in the United States (2), and presents de-novo with diffuse erythroderma, lymphadenopathy, and overt leukemic disease. Both SS and MF cells are Sézary cells with cerebriform nuclei and are CD4+/CD8 αβ T-cells in most cases. Both exhibit similar T-cell antigen expression profiles when studied on peripheral blood, with frequent loss of CD26 and CD7 (4). Patients with MF may also develop diffuse erythroderma during the course of the disease (5). These cases of erythrodermic MF can have lymphadenopathy and circulating Sézary cells in the peripheral blood, and thus are indistinguishable clinically from SS. Nevertheless, patients with erythrodermic MF usually have a history of MF with patch/plaque lesions for years and should not be diagnosed as SS, which presents with erythroderma and peripheral blood involvement at initial presentation.

Advances in our understanding of T-cell immunology have stimulated an interest in determining the putative cell of origin of various T cell lymphomas, including CTCLs, based on their immunophenotypic resemblance to known benign T-cell subsets. Various discrete subpopulations of normal CD4+ T-cells have been described, corresponding to different stages of antigen experience, functional properties, migration capabilities and their ability to survive and/or self-renew (69). T-cells emigrating from the thymus that have not yet encountered their cognate antigen constitute the naïve T-cell subset (TN), which has the capacity to self-renew and home to secondary lymphoid tissues. When T-cells encounter their cognate antigen, they progress through central memory T-cells (TCM), effector memory T-cells (TEM), and finally terminally differentiated effector memory T-cells (TEMRA). TCM cells retain the capacity to home to secondary lymphoid tissues and self-renew, providing the basis for long term immunity. TEM cells are capable of migrating to sites of inflammation and best suited to exert their function as orchestrators of the immune response. TEMRA cells develop cytotoxic properties and express IFN-γ after activation through the T-cell receptor (TCR). The CD8+ TEMRA cells have been well characterized, but the CD4+ TEMRA cells are ill-defined and usually ignored in the literature, although both CD4+ and CD8+ TEMRA cells have similar functions. The progression of T-cells from TN to TEMRA cells is accompanied by characteristic changes in antigen expression of surface markers including CD45RA, CD45RO, CCR7, CD62L, CD27, and CD28, which can be used to define the T-cell subsets. TN cells are CD45RA+CD45ROCCR7+CD62L+CD27+CD28+. After antigen stimulation, TN cells lose CD45RA and gain expression of CD45RO and become TCM cells. As TCM cells progress to TEM cells, they further lose expression of CCR7, CD62L, CD27, and CD28. At the last stage, TEMRA cells regain expression of CD45RA with loss of CD45RO. TEMRA cells are otherwise differentiated from TN cells by the absence of expression of CCR7, CD62L, CD27, and CD28.

Immunophenotypic analyses on a limited number of CTCL cases have suggested that MF originates from TEM, while SS arises from TCM, indicating that MF and SS are biologically distinct diseases (1012). This concept has been widely embraced, as it helps explain the dramatically different clinical behavior of the two diseases with otherwise indistinguishable cytologic and immunophenotypic features (13,14). Nevertheless, recent studies have found a more heterogeneous expression (1517), challenging the above cell-of-origin hypothesis.

Using a 10-color flow cytometry T-cell panel, we prospectively analyzed the immunophenotypes of neoplastic T-cells in clinically defined MF and SS, detected on fresh peripheral blood or bone marrow samples. Our findings reveal a broad heterogeneity and variability in the expression of antigens associated with naïve or memory T-cell subsets, with no significant correlation with disease classification based on clinical features. Moreover, significant phenotypic shifts on the expression of these antigens were detected on follow-up specimens, suggesting that the naïve/memory phenotype of the neoplastic cells is likely representative of a functional state, rather than an indication of cell of origin.

MATERIALS AND METHODS

Patient Selection

Patients with MF or SS were evaluated at Moffitt Cancer Center (MCC) between February 2015 and October 2017. Staging studies performed upon initial evaluation included complete physical examination, computer tomography, peripheral blood cell counts, white blood cell differentials, and peripheral blood flow cytometry, following the 2007 criteria from the International Society for Cutaneous Lymphoma (ISCL) and the European Organization of Research and Treatment of Cancer (4). Histological slides, flow cytometry results, TCR gene rearrangement studies, imaging studies, photographs of skin lesions, and clinical history were evaluated at the MCC interdisciplinary tumor board to render a clinicopathologic diagnosis based on the World Health Organization/European Organization for Research and Treatment of Cancer (WHO/EORTC) cutaneous lymphoma guidelines (18), and recommendations from the ISCL (19). Control peripheral blood samples were retrospectively identified by searching the medical records for patients with a complete clinical history, comprehensive laboratory work up and no demonstrable hematological malignancy, on which a 10-color T-cell panel was performed. This study was approved by the University of South Florida Institutional Review Board.

Flow Cytometry

All samples were analyzed fresh and stained with antibodies within 24 hours after collection. Aliquots of 50 μL of whole blood or bone marrow aspirate in sodium heparin, were incubated in the dark for 15 min at room temperature with combinations of 10 monoclonal antibodies conjugated to Pacific Blue (PB), Krome Orange (KO), fluorescein isothiocyanate (FITC), phycoerythrin (PE), R-phycoerythin-Texas Red-X (ECD), phycoerythrin-cyanine 5.5 (PC5.5), phycoerythrin-cyanin 7 (PC7), allophycocyanin (APC), allophycocyanin-alexa fluor 700 (APC-A700), and allophycocyanin-alexa fluor 750 (APC-A750). Red blood cells were lysed with BD FACS lysing solution (BD Biosciences, San Jose, CA) and nucleated cell were resuspended in phosphate buffered saline containing 2% paraformaldehyde. A standardized 10-color T-cell panel (PB/KO/FITC/PE/ECD/PC5.5/PC7/APC/APC-A700/APC-A750) was utilized in all specimens, consisting of three stained tubes (CD8/CD45/CD2/CD26/CD3/CD5/CD7/CD30/CD19/CD4, CD8/CD45/TCRγδ/TCRαβ/CD3/PD-1/CD10/CCR4/CD25/CD4, and CD57/CD45/CD45RA/CD52/CD62L/CD56/CD7/CD16/CD4/CD3). All antibodies were obtained from Beckman Coulter (Brea, CA). Up to 1,000,000 events were acquired until 5,000 lymphocytes were collected on a Gallios flow cytometer (Beckman Coulter).

Listmode files were analyzed as previously described (20), based on ad-hoc templates created on Kaluza version 1.5a (Beckman Coulter, Brea, CA). In brief, each lymphoid population was defined as the intersection of three interdependent polygonal gates drawn on three separate two-dimensional dot-plots, representing the six most informative and biologically relevant parameters for each particular lymphoid subset. Real time color-coding for each population was used to visually narrow the polygonal gates in order to define single clusters of cells with homogeneous fluorescence properties in most parameters studied. All lymphoid populations were identified and categorized.

Delta median fluorescence values were calculated by comparing the fluorescence of gated neoplastic events to that of monocytes (for CD2, CD5, CD7, CCR4, and PD-1), neutrophils (for CD26 and CD52) or NK cells (for CD3 and CD4) within the same analysis tube.

Statistical Analysis

All statistic calculations were performed using GraphPad Prism, version 7.01 for Windows (GraphPad Software, San Diego, CA). Differences in antigen expression between groups were studied using a two-tailed Mann Whitney test on delta median fluorescence values. Differences in frequencies of antigen loss or naïve-memory T-cell phenotype were studied using the Chi-square test. A statistically significant P value was considered as <0.05.

RESULTS

We identified neoplastic T-cells by 10-color flow cytometry on 74 peripheral blood specimens and 5 bone marrow specimens from 52 patients with CTCL. Based on a comprehensive review of the clinical history, laboratory studies, and pathology material, 33 patients met criteria for de-novo SS while 19 patients were diagnosed as MF. The median age of the patients with SS was 71 years old (range: 28–90), and the male to female ratio was 1.4. At the time of diagnosis, prior and current staging studies were consistent with stage IVA1 (27 patients, 82%), IVA2 (2 patients, 6%), or IVB (4 patients, 12%) disease in patients with SS. The median age of the patients with MF was 69.5 years old (range: 33–87), and the male to female ratio was 5.7. At the time of initial diagnosis of MF, patients presented with stage IA (3 patients, 16%), IB (9 patients, 47%), IIB (3 patients, 16%), IIIA (1 patient, 5%), IIIB (1 patient, 5%), and IVA1 (2 patients, 11%) disease.

To standardize our immunophenotypic analysis, we studied 27 control individuals (patients with no demonstrable hematologic malignancy) and showed the presence of discrete CD4+ T-cell subsets corresponding to TN (CD62L+/CD45RA+, median: 21.8%, range: 2.2–74.3%), TCM (CD62L+/CD45RA, median: 51%, range: 19.9–70%), TEM (CD62L/CD45RA, median: 21.3%, range: 4.17–59.4%), and TEMRA (CD62L/CD45RA+, median: 1.2%, range: 0–11%, Figure 1A,B). In contrast, CD8+ T-cells showed a more prominent population of TEMRA cells (range: 1–72.8%, median: 29.6%) (Figure 1C), while most NK cells were positive for CD45RA and showed a spectrum or biphasic expression of CD62L (Figure 1D). Based on these findings, appropriate thresholds were set up on CD62L versus CD45RA 2-dimensional plots for the evaluation of tumor samples, with particular attention to background benign lymphoid subsets as internal controls (Figure 2).

Fig. 1.

Fig. 1.

A, Representative flow cytometry plots from a control individual with no demonstrable hematologic malignancy, showing the typical distribution of benign CD4+ T-cells (cyan), CD8+ T-cells (orange), and NK cells (gold), on CD62L versus CD45RA dot plots. Also shown are the quadrant gates utilized to quantify naïve (TN), central memory (TCM), effector memory (TEM), and effector memory with CD45RA expression (TEMRA) CD4+ T-cells. B, Percentages of TN, TCM, TEM, and TEMRA CD4+ T-cells, and C, similar subsets of CD8+ T-cells across 27 control individuals. D, Percentage of NK cells falling within the quadrant gates defining naïve/memory T-cell subsets.

Fig. 2.

Fig. 2.

Representative peripheral blood flow cytometry plots from patients with MF (A and C) or SS (B and D), demonstrating the presence of neoplastic T-cells with immunophenotypic features of TN (A), TCM (B), TEM (C), or TEMRA cells (D). Neoplastic T-cells (red) were identified based on immunophenotypic aberrancies, such as CD3dim/CD4dim (A), CD3dim/CD26 (B), CD2dim/−/CD26 (C), and CD3+/CD2 (D). Background benign CD4+ T-cells (cyan), CD8+ T-cells (orange), and NK cells (gold) are also shown. Expression of PD-1 and CCR4 is shown in the middle plots.

Immunophenotypic results for all cases are summarized in Figure 3. With the exception of 1 case of MF double negative for CD4/CD8, all neoplastic cells were positive for CD4, and negative for CD8 and TCRγδ. Except for one patient (four cases) with neoplastic cells lacking surface TCR expression (as determined by absent surface CD3 and TCR), all cases were positive for TCRαβ. As previously described (20), the most common immunophenotypic aberrancies in SS and MF were decreased expression or loss of CD26 (81%) and CD7 (64.6%), followed by variably diminished expression of CD2 (41.8%), CD3 (35.4%), CD4 (30.4%), and CD5 (15.2%). There was no statistically significant correlation between the incidence of these immunophenotypic abnormalities and a clinical diagnosis of MF or SS (data not shown). When gated based on these immunophenotypic aberrancies, neoplastic T-cells comprised a median of 58.8% of lymphoid events (range: 10–97.4%).

Fig. 3.

Fig. 3.

Heat maps summarizing the flow cytometric findings in leukemic CTCL, including the presence of neoplastic T-cells with predominantly TN, TCM, TEM, or TEMRA phenotype. Each row represents gated benign CD4+ T-cells from individuals with no hematopoietic malignancy (top), or gated aberrant neoplastic T-cells from patients with MF or SS. The color intensity on the left heat maps depicts the percentage of benign CD4+ T-cells (top) or neoplastic T-cells within the corresponding naïve/memory T-cell subset-defining quadrants, on a CD62L versus CD45RA dot plot. The color intensity on the right heat maps depicts the delta median fluorescence intensity (ΔMFI) compared to an internal control population, and normalized to the highest recorded value for the corresponding antigen (% maximum value).

Gated neoplastic T-cells from patients with CTCL exhibited a variety of staining patterns for CD62L and CD45RA (Figure 2). When comparing cases of MF and SS, the predominant naïve/memory phenotype was TN in 12% and 15% of cases, TCM in 33% and 46% of cases, TEM in 49% and 30% of cases, and TEMRA in 6 and 9% of cases, respectively (Figures 3). There was no correlation between naïve/memory phenotype and a clinical diagnosis of MF versus SS (P = 0.4). When sequential samples from the same patient were compared, the naïve/memory phenotype remained the same for 10 patients, up to 273 days after the initial evaluation. In six other patients, the naïve/memory phenotype was different on one or more follow up analyses (Figure 4), due to changes in CD62L expression (switching between TCM and TEM phenotype) in five cases, and loss of CD45RA expression in one case.

Fig. 4.

Fig. 4.

Representative peripheral blood flow cytometry plots from two patients with Sezary syndrome exhibiting naïve/memory phenotype shifts on follow up studies. A, CD3dim/CD26 neoplastic T-cells showed a similar TCM phenotype at diagnosis (not shown) and 27 days later (top). On day 234, a follow up study showed loss of CD62L expression on the neoplastic cells, corresponding to a TEM phenotype (bottom). B, CD3+/CD2dim/− neoplastic T-cells at first evaluation showed a TEM phenotype (top). On a follow up study 277 days later, the neoplastic cells showed gain of CD62L expression and a TCM phenotype (bottom).

Expression of the targetable surface antigens CD52, CCR4, and PD-1 was evaluated on CTCL patients and controls, excluding samples obtained after therapy with the corresponding therapeutic antibody (Alemtuzumab, Mogamulizumab, and anti-PD1 antibodies, respectively). As a group, neoplastic T-cells from patients with CTCL exhibited higher expression of PD-1 and CCR4 (P < 0.0001 for both), compared to benign CD4 T-cells from control individuals; although expression levels comparable with benign T-cells were observed in many cases (Figure 5). With the exception of 3 CTCL cases from different patients negative for CD52 (4 SS and 1 MF), neoplastic cells were positive for CD52 at levels comparable to benign T-cells (P = 0.36). SS cases exhibited higher levels of CCR4 compared to MF cases (P = 0.001), but with significant overlap of fluorescence values. In addition, negative PD-1 expression was not seen in SS but encountered in a few cases of MF (Figure 5B).

Fig. 5.

Fig. 5.

Scatter plots comparing the level of expression of CD52 (A), PD1 (B), and CCR4 (C) between benign CD4+ T-cells from control individuals, and malignant T-cells identified on samples from patients with MF and SS. Each dot represents one sample studied. For illustration purposes, values ≤0.1 is displayed against the axis. ΔMFI: Delta median fluorescence intensity, compared to an internal control population. Error lines show median and interquartile range. The P values for statistically significant differences are shown. NS: Not statistically significant.

DISCUSSION

Prior immunophenotypic studies on limited numbers of CTCL cases have reported that neoplastic T-cells in SS are usually positive for CD27, CCR7, and CD62L; while neoplastic T-cells from skin lesions in MF are usually negative for these surface antigens (10,11). In the normal CD4+ T-cell repertoire, expression of these antigens is strongly associated with the TN and TCM subsets, while lack of expression is characteristic of the TEM and TEMRA subsets. CD62L and CCR7 play an essential role in T-cell migration to lymph nodes while CD27 mediates costimulatory signals necessary for the establishment of T-cell immunity, consistent with the physiologic role of TN and TCM as circulating immune initiators and surveyors. In addition, CTCL has been shown to express surface molecules involved in T-cell homing to skin (namely CLA, CCR4, and CCR10), which can also be demonstrated in a subset of antigen-experienced benign T-cells. This evidence has fueled the concept that SS originates from CD4+ TCM cells, while MF arises from TEM cells.

The above “cell-of-origin” hypothesis has gained widespread support, as it helps explain the dramatic differences in clinical behavior between MF and SS, despite otherwise almost identical cytological and immunophenotypic features. Nevertheless, other lines of evidence have posed a challenge to this model. For example, at least two reports have demonstrated a high incidence of CCR7 expression in MF, particularly in advance-stage skin lesions (16,17), arguing against a TEM phenotype. When studied, CD45RA expression in MF and SS has been variable and not supportive of a TCM or TEM phenotype, as this marker is expected to be negative in these T-cell subsets (21). More recently, two immunophenotypic studies have revealed an unexpected heterogeneity of the naïve/memory maturation phenotype in SS, with remarkable disparities between blood- and skin-derived neoplastic T-cells (22,23). In addition, mass parallel gene sequencing has demonstrated a high number of ultraviolet light-associated dinucleotide mutations (CC> > TT) in both MF and SS (24,25), arguing against a TCM origin as this T-cell subset is largely circulatory and not skin-bound. We hereby report the largest published series of CTCL evaluated for naïve/memory phenotypic properties on neoplastic T-cells by flow cytometry. Importantly and for the first time, we compared the naïve/memory phenotype of circulating neoplastic T-cells in SS and MF on a large clinical series, to evaluate the veracity of the “cell of origin” hypothesis and the diagnostic utility of naïve/memory phenotyping to distinguish SS from leukemic MF. As suggested by recent lines of evidence, we demonstrated a highly variable spectrum of naïve/memory T-cell phenotype in CTCL, with no statistically significant correlation with a clinical diagnosis of MF versus SS.

Our naïve/memory T-cell phenotyping is based on two surface antigens widely utilized for this purpose: CD62L and CD45RA. It should be noted that the correlation between these and other markers utilized to define T-cell subsets, such as CD27, CD44, CD45RO, CD127, and CCR7, is not perfect when studying the normal CD4+ T-cell repertoire (8,21) and also on limited data on CTCL (10). Thus, attempts to identify naïve or memory-like phenotypes in CTCL using a different set of antigens might not give identical results. In the design of our panel, we favored using CD62L over the similarly expressed antigen CCR7, based on the essential role that CD62L plays in the rolling of lymphocytes on high endothelial venules, and the fact that CXCR4 (which is commonly expressed in CTCL) can compensate for a lack of CCR7 in facilitating T-cell homing to lymph nodes (26). Similarly, CD45RA expression was arbitrarily selected (over lack of CD44 and CD45RO expression) as the marker of choice to define TN cells, based on our desire to also identify the TEMRA subset.

CD62L (also known as l-selectin) is a cell adhesion molecule, which recognizes sialylated carbohydrate groups on CD34, GlyCAM, and MadCAM expressed on high endothelial venules. It is an indispensable surface molecule for the rolling and tethering of leukocytes entering lymphoid tissues, and it is believed to mediate the circulation of the TN and TCM subsets through lymph nodes. Interestingly, CD62L-positive neoplastic cells have been shown to be selectively depleted upon anti-CD52 (Alemtuzumab) therapy in CTCL (12), suggesting a mechanism for the resistance of CD62L-negative skin infiltrates in MF. In our series, we demonstrate that the expression of CD62L in circulating Sézary cells is highly variable, both in MF and SS, and is not necessarily stable. These findings might have important implications for therapy with Alemtuzumab and should be further studied in the setting of clinical trials.

The phenotypic shift observed in 37.5% of the patients (6 of 16 patients) was mainly due to changes in CD62L expression. These patients were not treated with Alemtuzumab. While CD62L expression is often lost after density gradient centrifugation and cryopreservation of samples, the change of CD62L was not believed to be due to these technical issues as all our samples were analyzed fresh and comparably between initial and subsequent samples. MF has been postulated to result from persistent antigen stimulation, and the change in the naïve/memory phenotype is an antigen driven event (27). Therefore, the phenotypic shift in these patients was most likely driven by an unknown antigenic challenge. This further supports the idea that the naïve/memory phenotype of CTCL reflects the functional status of the Sézary cells and is not a good surrogate for the classification of SS and MF.

CCR4 is the chemokine receptor for CCL17 and CCL22, and has been involved in the migration of activated T-cells into inflamed tissues, including skin. Indeed, CCR4 has been shown to mediate the migration of tumor cells from blood to skin in SS (28). Approximately 20–25% of peripheral blood CD4+ T-cells from healthy individuals exhibit some level of expression of CCR4 (29,30), while CCR4 expression in MF and SS has been reported to be consistently positive (1012) and higher than benign T-cells (30). In one study, higher levels of CCR4 expression were observed in SS compared to MF, which correlated with a better response to therapy with the anti-CCR4 antibody Mogamulizumab (31). We have independently confirmed a statistically significant higher expression of CCR4 in SS compared to MF, in agreement with the above reported finding. Although assessment of CCR4 expression by flow cytometry provides useful information when considering therapy with Mogamulizumab, significant overlaps of CCR4 expression between MF, SS, and reactive T-cells might pose challenges in defining and harmonizing positivity thresholds and expression intensity measures.

In conclusion, we report the immunophenotype of circulating MF and SS, defined based on expert multidisciplinary review of the clinical history and laboratory studies. In particular, we defined leukemic/advanced-stage MF based on a reliable history depicting clinical features typical for MF and eventual progression to leukemic disease. While MF and SS are recognized as two distinct CTCLs in the current WHO Classification of Diseases (32), ours, and other recent findings indicate that circulating MF and SS show similar naïve/memory maturation phenotypic heterogeneity and may represent a spectrum of same disease process. Thus other than the clinical history, the cases of “MF transformation to SS” or “secondary SS” cannot be reliably distinguished from “De novo” SS based on phenotypic analysis.

Footnotes

CONFLICT OF INTEREST

The authors declare no conflict of interest.

LITERATURE CITED

  • 1.Weinstock MA, Gardstein B. Twenty-year trends in the reported incidence of mycosis fungoides and associated mortality. Am J Public Health 1999;89:1240–1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Imam MH, Shenoy PJ, Flowers CR, Phillips A, Lechowicz MJ. Incidence and survival patterns of cutaneous T-cell lymphomas in the United States. Leuk Lymphoma 2013;54:752–759. [DOI] [PubMed] [Google Scholar]
  • 3.Yamashita T, Abbade LP, Marques ME, Marques SA. Mycosis fungoides and Sezary syndrome: Clinical, histopathological and immunohistochemical review and update. An Bras Dermatol 2012; 87:817–828. quiz 829–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.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. [DOI] [PubMed] [Google Scholar]
  • 5.Vonderheid EC, Bernengo MG, Burg G, Duvic M, Heald P, Laroche L, Olsen E, Pittelkow M, Russell-Jones R, Takigawa M. et al. Update on erythrodermic cutaneous T-cell lymphoma: Report of the International Society for Cutaneous Lymphomas. J Am Acad Dermatol 2002;46:95–106. [DOI] [PubMed] [Google Scholar]
  • 6.Koch S, Larbi A, Derhovanessian E, Ozcelik D, Naumova E, Pawelec G. Multiparameter flow cytometric analysis of CD4 and CD8 T cell subsets in young and old people. Immun Ageing 2008;5:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Appay V, van Lier RA, Sallusto F, Roederer M. Phenotype and function of human T lymphocyte subsets: Consensus and issues. Cytometry A 2008;73A:975–983. [DOI] [PubMed] [Google Scholar]
  • 8.Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999;401:708–712. [DOI] [PubMed] [Google Scholar]
  • 9.Roetynck S, Olotu A, Simam J, Marsh K, Stockinger B, Urban B, Langhorne J. Phenotypic and functional profiling of CD4 T cell compartment in distinct populations of healthy adults with different antigenic exposure. PLoS One 2013;8:e55195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Campbell JJ, Clark RA, Watanabe R, Kupper TS. Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: A biologic rationale for their distinct clinical behaviors. Blood 2010;116:767–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Clark RA, Shackelton JB, Watanabe R, Calarese A, Yamanaka K, Campbell JJ, Teague JE, Kuo HP, Hijnen D, Kupper TS. High-scatter T cells: A reliable biomarker for malignant T cells in cutaneous T-cell lymphoma. Blood 2011;117:1966–1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Clark RA, Watanabe R, Teague JE, Schlapbach C, Tawa MC, Adams N, Dorosario AA, Chaney KS, Cutler CS, Leboeuf NR. et al. Skin effector memory T cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients. Sci Transl Med 2012;4:117ra117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jawed SI, Myskowski PL, Horwitz S, Moskowitz A, Querfeld C. Primary cutaneous T-cell lymphoma (mycosis fungoides and Sezary syndrome). Part I. Diagnosis: Clinical and histopathologic features and new molecular and biologic markers. J Am Acad Dermatol 2014;70:205. e201–205 e216. [DOI] [PubMed] [Google Scholar]
  • 14.Song SX, Willemze R, Swerdlow SH, Kinney MC, Said JW. Mycosis fungoides: Report of the 2011 Society for Hematopathology/European Association for Haematopathology workshop. Am J Clin Pathol 2013;139:466–490. [DOI] [PubMed] [Google Scholar]
  • 15.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. [DOI] [PubMed] [Google Scholar]
  • 16.Hu SC, Lin CL, Hong CH, Yu HS, Chen GS, Lee CH. CCR7 expression correlates with subcutaneous involvement in mycosis fungoides skin lesions and promotes migration of mycosis fungoides cells (MyLa) through mTOR activation. J Dermatol Sci 2014;74: 31–38. [DOI] [PubMed] [Google Scholar]
  • 17.Kallinich T, Muche JM, Qin S, Sterry W, Audring H, Kroczek RA. Chemokine receptor expression on neoplastic and reactive T cells in the skin at different stages of mycosis fungoides. J Invest Dermatol 2003;121:1045–1052. [DOI] [PubMed] [Google Scholar]
  • 18.Willemze R, Jaffe ES, Burg G, Cerroni L, Berti E, Swerdlow SH, Ralfkiaer E, Chimenti S, Diaz-Perez JL, Duncan LM. et al. WHO-EORTC classification for cutaneous lymphomas. Blood 2005;105:3768–3785. [DOI] [PubMed] [Google Scholar]
  • 19.Pimpinelli N, Olsen EA, Santucci M, Vonderheid E, Haeffner AC, Stevens S, Burg G, Cerroni L, Dreno B, Glusac E. et al. Defining early mycosis fungoides. J Am Acad Dermatol 2005;53:1053–1063. [DOI] [PubMed] [Google Scholar]
  • 20.Horna P, Deaver DM, Qin D, Moscinski LC, Sotomayor EM, Glass LF, Sokol L. Quantitative flow cytometric identification of aberrant T cell clusters in erythrodermic cutaneous T cell lymphoma. Implications for staging and prognosis. J Clin Pathol 2014; 67:431–436. [DOI] [PubMed] [Google Scholar]
  • 21.Campbell JJ, Murphy KE, Kunkel EJ, Brightling CE, Soler D, Shen Z, Boisvert J, Greenberg HB, Vierra MA, Goodman SB. et al. CCR7 expression and memory T cell diversity in humans. J Immunol 2001;166:877–884. [DOI] [PubMed] [Google Scholar]
  • 22.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. [DOI] [PubMed] [Google Scholar]
  • 23.Moins-Teisserenc H, Daubord M, Clave E, Douay C, Felix J, Marie-Cardine A, Ram-Wolff C, Maki G, Beldjord K, Homyrda L. et al. CD158k is a reliable marker for diagnosis of Sezary syndrome and reveals an unprecedented heterogeneity of circulating malignant cells. J Invest Dermatol 2015;135:247–257. [DOI] [PubMed] [Google Scholar]
  • 24.McGirt LY, Jia P, Baerenwald DA, Duszynski RJ, Dahlman KB, Zic JA, Zwerner JP, Hucks D, Dave U, Zhao Z. et al. Whole-genome sequencing reveals oncogenic mutations in mycosis fungoides. Blood 2015;126:508–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang L, Ni X, Covington KR, Yang BY, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Scimone ML, Felbinger TW, Mazo IB, Stein JV, Von Andrian UH, Weninger W. CXCL12 mediates CCR7-independent homing of central memory cells, but not naive T cells, in peripheral lymph nodes. J Exp Med 2004;199:1113–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Boursalian TE, Bottomly K. Stability of naive and memory phenotypes on resting CD4 T cells in vivo. J Immunol 1999; 162:9–16. [PubMed] [Google Scholar]
  • 28.Hoeller C, Richardson SK, Ng LG, Valero T, Wysocka M, Rook AH, Weninger W. In vivo imaging of cutaneous T-cell lymphoma migration to the skin. Cancer Res 2009;69:2704–2708. [DOI] [PubMed] [Google Scholar]
  • 29.Wu XS, Lonsdorf AS, Hwang ST. Cutaneous T-cell lymphoma: Roles for chemokines and chemokine receptors. J Invest Dermatol 2009;129:1115–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ferenczi K, Fuhlbrigge RC, Pinkus J, Pinkus GS, Kupper TS. Increased CCR4 expression in cutaneous T cell lymphoma. J Invest Dermatol 2002;119:1405–1410. [DOI] [PubMed] [Google Scholar]
  • 31.Ni X, Jorgensen JL, Goswami M, Challagundla P, Decker WK, Kim YH, Duvic MA. Reduction of regulatory T cells by Mogamulizumab, a defucosylated anti-CC chemokine receptor 4 antibody, in patients with aggressive/refractory mycosis fungoides and Sezary syndrome. Clin Cancer Res 2015;21:274–285. [DOI] [PubMed] [Google Scholar]
  • 32.Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, Advani R, Ghielmini M, Salles GA, Zelenetz AD. et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127:2375–2390. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES