Abstract
Disease overview
Cutaneous T-cell lymphomas are a heterogenous group of T-cell lymphoproliferative disorders involving the skin, the majority of which may be classified as Mycosis Fungoides (MF) or Sézary Syndrome (SS).
Diagnosis
The diagnosis of MF or SS requires the integration of clinical and histopathologic data.
Risk-adapted therapy
TNMB (tumor, node, metastasis, blood) staging remains the most important prognostic factor in MF/SS and forms the basis for a “risk-adapted,” multi-disciplinary approach to treatment. For patients with disease limited to the skin, expectant management or skin-directed therapies is preferred, as both disease-specific and overall survival for these patients is favorable. In contrast, patients with advanced-stage disease with significant nodal, visceral or blood involvement are generally approached with biologic-response modifiers or histone deacetylase inhibitors prior to escalating therapy to include systemic, single-agent chemotherapy. In highly-selected patients, allogeneic stem-cell transplantation may be considered, as this may be curative in some patients.
Disease Overview
Primary cutaneous lymphomas are a heterogenous group of extranodal non-Hodgkin lymphomas which, by definition, are largely confined to the skin at diagnosis. The European Organization for Research and Treatment of Cancer (EORTC) and World Health Organization (WHO) published a consensus classification for cutaneous lymphomas in 2005 (1). In contrast to nodal non-Hodgkin lymphoma, most of which are B-cell derived, approximately 75% of primary cutaneous lymphomas are T-cell derived, two-thirds of which may be classified as Mycosis fungoides (MF) or Sézary Syndrome (SS) (1–3). The incidence of cutaneous T-cell lymphomas (CTCL) has been increasing and is currently 6.4 per million persons, based on Surveillance, Epidemiology, and End Results (SEER) registry data, with the highest incidence rates being reported among males and African-Americans (2). While CTCL may occur in children and young adults, this is very uncommon and often associated with histopathologic variants of MF (4–6). The incidence of CTCL increases significantly with age, with a median age at diagnosis in the mid-50’s and a four-fold increase in incidence appreciated in patients over 70 (2, 6).
Epidemiological studies have failed to consistently identify environmental or virally associated risk factors for most CTCL subtypes, with the notable exception of HTLV-1 infection in adult T-cell leukemia/lymphoma (7). Recent studies, however, have suggested that medications may induce an antigen-driven T-cell lymphoproliferation or dyscrasia (8, 9). A recent case series examined a subset of hypertensive MF patients using hydrochlorothiazide. When compared to hypertensive MF patients not using hydrochlorothiazide, these patients were more likely to have stage I disease, and were less likely to have a clonal TCR gene rearrangement (9). More importantly, in a subset of these patients, a complete or partial response was observed upon discontinuation of hydrochlorothiazide. In three patients, CTCL recurred upon reinitiating hydrochlorothiazide, and subsequently receded with its discontinuation. While these findings could be interpreted as a drug reaction, more specifically a drug-induced pseudolymphoma, the authors of this single center study speculate that hydrochlorothiazide may be associated with antigen-driven T-cell lymphoproliferation and could serve as a trigger for MF. Consequently, a therapeutic trial off hydrochlorothiazide may be warranted in selected patients. Moreover, as a variety of other medications may initiate a reaction mimicking MF, a careful medication history should be performed in these patients with a trial off any suspected offending drug. Individual genetic features have also been implicated in the development of CTCL. Rare reports of familial MF and the detection of specific HLA class II alleles in association with both sporadic and familial MF suggest that host genetic factors may contribute to MF development (10–12). While the role of environmental and host genetic factors in CTCL pathogenesis remains unclear, significant insights into disease ontogeny, molecular pathogenesis and disease-associated immune dysregulation have been realized (13–16).
Cell of origin
Naïve T cells, upon encountering antigen in skin-draining lymph nodes, inducibly express the E-selectin ligand cutaneous lymphocyte antigen (CLA) and chemokine receptors (e.g. CCR4, CCR8, CCR10) that are required for their subsequent trafficking to the skin (17–19). Clonal expansion of activated T cells is followed by their differentiation into multiple subsets of effector and memory cells. Central memory cells (TCM) retain the ability to access the peripheral blood and lymph nodes. Effector memory cells (TEM), in contrast, migrate into extranodal sites, including the skin, where a subset will remain, as tissue-resident memory cells (TRM). The majority of T cells in the skin are TRM (17, 20), express a high-affinity antigen receptor (21), and have a distinct gene-expression profile (22). Clonal T cells in MF are commonly TRM derived, thus explaining their tendency to remain confined to the skin (23). Immunophenotyping studies demonstrate that malignant T cells in patients with leukemic CTCL variants (Sézary Syndrome and MF with secondary leukemic involvement) express CCR7 and L-selectin, resembling TCM (24). This fundamental difference in the putative cell of origin between SS (TCM derived) and MF (TRM derived) is consistent with their distinct clinical behavior, as TCM may be found in both the peripheral blood, lymph node and skin and are long-lived cells resistant to apoptosis, while skin-resident TRM cells fail to circulate in peripheral blood, remaining fixed within the skin (24). In addition, a population of recirculating CCR7+L-selectin− migratory memory T cells (TMM) has been described in the skin (20). Therefore, a subset of MF patients with secondary leukemic involvement, poorly demarcated patches/plaques, more significant dermal involvement, and dermatopathic lymphadenopathy may harbor a TMM-derived clone (20). The contention that MF subtypes and SS originate from different T-cell subsets is consistent with comparative genomic hybridization (CGH) and gene-expression profiling data demonstrating that these CTCL subtypes are genetically distinct (25, 26).
Regulatory T cells (Treg) expressing the transcription factor FoxP3 are important in the maintenance of self-tolerance and form a minor subset of skin-resident T cells. Heid et. al. demonstrated that the malignant T cells in a subset of Sézary patients may be derived from Treg cells, as the malignant clone in these patients not only expressed FoxP3 and suppressed conventional T cells, but possessed a demethylated FoxP3 promoter (27). Uncertainties remain as to whether or not a subset of Sézary patients harbor a clone that is derived from bona fide skin resident Treg cells, or whether these cells aberrantly acquire a Treg phenotype during disease evolution (28). For example, immature dendritic cells, which are prevalent in CTCL (29), may upregulate FoxP3 expression in malignant T cells (30). Therefore, a subset of SS patients appears to harbor a Treg-derived (or “Treg-like”) clone, although the prognostic and therapeutic implications of this observation remain to be defined.
In contrast to regulatory T cells, which represent a minority of skin-resident T cells, the majority of T cells in the skin produce cytokines characteristic of distinct effector T-cell subsets, including Th1, Th2 and Th17 cells. This effector T-cell heterogeneity raises the possibility that future studies may subclassify CTCL based on these T-cell subsets (31, 32). Of note, MF/SS is associated with the expression of Th2-associated genes (e.g. GATA-3) and the production of Th2-associated cytokines (e.g. IL-4, IL-5, IL-13), raising the possibility that a significant subset of patients may harbor Th2-derived clones (33–37). Alternatively, recurrent mutations activating specific signaling pathways (e.g. NFAT, NFκB, JAK/STAT) may promote the acquisition of a particular phenotype independent of the cell of origin (38). T-cell differentiation is associated with considerable plasticity. Therefore, the phenotype of malignant T cells may be both heterogeneous and highly dependent upon cues within the microenvironment (30, 39, 40). As the genetic landscape and the putative cell of origin are further defined in subsets of CTCL, including MF/SS, one may anticipate that this data may have a significant impact on the classification, risk-stratification and treatment of these diseases.
Immunopathogenesis
The establishment of long-term CTCL cell lines is challenging, as these cells frequently undergo spontaneous cell death during in vitro culture (41, 42)(and personal observation). Therefore, the resistance to apoptosis observed in vivo is unlikely due to an intrinsic resistance to apoptosis alone. Rather, extrinsic factors present within the tumor microenvironment likely contribute to the growth and survival of malignant T cells, a contention supported by the observation that cytokine supplementation or the provision of T-cell costimulatory signals supports the growth of malignant T cells in vitro (41, 43, 44). Both gene-expression profiling and immunohistochemistry-based studies have recently highlighted the important contribution of non-malignant cells, including monocyte-derived lymphoma-associated macrophages, in the pathogenesis of both Hodgkin and non-Hodgkin lymphomas (45–47). Similarly, malignant T cells in the skin are frequently associated with dendritic cells and immunohistochemistry-based studies have clearly demonstrated an abundance of both lymphoma-associated macrophages and dendritic cells, many of which may be actively recruited into the tumor microenvironment by tumor-derived chemokines (29, 48). These monocyte-derived cells promote tumorigenesis both directly, by the production of factors which promote tumor cell growth and survival, and indirectly, by supporting tumor angiogenesis and suppressing host anti-tumor immunity (49). For example, monocyte-derived dendritic cells supported the long-term survival of malignant T cells during in vitro culture (42). More recently, peripheral blood monocytes (and their progeny) were shown to support the growth of malignant T cells in vitro, confer resistant to chemotherapy, and promote tumor engraftment in immunodeficient mice (29). Lymphoma-derived IL-10, which is upregulated in patients with advanced-stage, refractory disease (50), impairs the maturation of lymphoma-associated dendritic cells, rendering them immunologically incompetent, thus promoting escape from host anti-tumor immune surveillance. In addition, lymphoma-associated dendritic cells were observed to express the T-cell co-inhibitory ligand B7-H1 (PD-L1, CD274), which directly inhibits the proliferation of tumor-specific T cells, and indirectly impairs anti-tumor immunity by promoting the induction of suppressive regulatory T cells (51). Therefore, lymphoma-associated macrophages and dendritic cells appear to play an important role in cutaneous T-cell lymphomagenesis while contributing to the evasion and suppression of host anti-tumor immunity.
In addition to the tumor microenvironment’s role, widespread impairment of cellular immunity – the tumor “macroenvironment” – has long been appreciated in CTCL and contributes to the significant morbidity and mortality associated with infectious complications observed in CTCL. Approximately 50% of patients with CTCL, particularly those with advanced-stage disease, will ultimately succumb to infectious complications (52–54). Both quantitative and qualitative defects in natural killer (NK) cell (55, 56), dendritic cell (57) and T cell-mediated (58–60) immunity are observed in CTCL. In addition, CTCL is associated with a significant loss of the T-cell repertoire, analogous to that observed in HIV infection. T-cell receptor (TCR) diversity within multiple TCR beta-variable (Vβ) families was analyzed using complementarity-determining region 3 (CDR3) spectratyping and combined with a quantitative analysis of TCR-Vβ usage by flow cytometry (61). In patients with advanced-stage disease, and half of patients with limited-stage disease, a dramatic loss of TCR diversity was observed. Whether this observation may be explained by tumor-mediated suppression of non-malignant T cells, diminished thymic output of naïve T cells and compensatory homeostatic expansion of oligoclonal peripheral T cells, or some other mechanism, is unknown (50). As lymphopenia is an adverse prognostic factor in many hematologic malignancies (62–67), and undoubtedly contributes to the infectious complications observed in CTCL, improved understanding of the causative mechanism(s) leading to this dramatic loss of T-cell diversity may have significant therapeutic implications.
Molecular pathogenesis
Recurrent chromosomal translocations involving the IgH gene on chromosome 14 lead to the aberrant expression of anti-apoptotic (e.g. Bcl-2) and oncogenic (e.g. cyclinD1, Myc) proteins in B-cell lymphomas. These recurrent translocations arise in peripheral B cells undergoing class-switch recombination and somatic hypermutation. In contrast, the TCR gene loci, while involved in recurrent chromosomal translocations in precursor T-cell lymphoblastic leukemias/lymphomas, are rarely involved in recurrent translocations in mature T-cell lymphoproliferative disorders (68, 69). With the exception of translocations involving the interferon regulatory factor 4 (IRF4) gene (also known as MUM1) in a subset of cutaneous anaplastic large cell lymphomas, recurrent chromosomal translocations are infrequently observed in CTCL (70–74). Despite this, a number of signaling pathways regulating cell-cycle progression and survival have been implicated in CTCL pathogenesis.
The NF-kB family of transcription factors (i.e. c-rel, p65/RelA, RelB, p50/p105, p52/p100) plays an important role in normal lymphocyte development, activation and differentiation via the regulation of target genes involved in cell growth, survival and cytokine production. Multiple mechanisms, well described in B-cell lymphomas, lead to constitutive NF-kB activation, promoting lymphomagenesis (75). In a similar fashion, NF-kB is constitutively activated in CTCL (76–78). Immunohistochemical analysis of MF cases demonstrated nuclear localization of p65/RelA in over 90% of the cases examined (76). Furthermore, pharmacologic NF-kB inhibition in CTCL cell lines decreases NF-kB DNA binding activity, thus promoting cell death (76–79). While the molecular mechanisms leading to constitutive NF-kB activation in CTCL are poorly understood, the observation that IKK inhibition downregulates NF-kB activity implicates upstream IKK-activating elements (77, 78).
The signal transducers and activators of transcription (STATs) are a family of six transcription factors which become phosphorylated by one of four upstream receptor-associated Janus kinases (JAKs) following cytokine stimulation. Nuclear localization and DNA-binding of phosphorylated STAT3 has been convincingly demonstrated in CTCL (80, 81). Following nuclear translocation, STAT3 directly regulates a number of target genes in CTCL, including regulators of apoptosis (e.g. Bcl-2/Bax), cytokines (e.g. IL-5, IL-13) and suppressors of cytokine signaling (e.g. SOCS). In addition, STAT3 indirectly regulates gene expression by inducing the expression of DNA methyltransferase 1 (DNMT1), which promotes the epigenetic silencing of tumor suppressor genes (82). Not surprisingly then, pharmacologic inhibition of STAT3 promotes apoptosis in CTCL (80, 83–85). Cytogenetic gains involving STAT5A and STAT5B or their activation in response to cytokines present within the tumor microenvironment suggests a pathogenic role for other STATs (40, 86–88).
Normal T cells undergo a controlled process of activation-induced cell death following antigen-dependent activation and proliferation, thus maintaining lymphocyte homeostasis. Extrinsic death receptors, including Fas (CD95), play an important role in regulating this process. A number of mechanisms, including promoter methylation (89–91), gene mutations (92) and loss of the long arm of chromosome 10 (93) result in diminished Fas expression in CTCL and reduced sensitivity to apoptosis. In addition, promoter methylation and epigenetic instability leading to the inactivation of many tumor suppressor genes, including those involved in the induction of apoptosis, appear to be commonly employed mechanisms of lymphomagenesis in CTCL (94).
In addition to multiple defects in apoptosis, aberrant cell-cycle regulation, including inactivation of the CDKN2A-CDKN2B locus, is frequently observed in CTCL (95, 96). Cyclin upregulation, including cyclinD1, and loss of RB1 have also been described (97). As gene-expression profiling and next-generation sequencing technologies are employed, additional pathogenic pathways, including those involving transcription factors regulating T-cell differentiation (36, 37), c-MYC (98, 99), RAS/RAF/MEK signaling (100), among others (93, 101), may be identified in subsets of CTCL. For example, a gain of function mutation (S345F) in the phospholipase C, gamma 1 (PLCG1) gene was recently observed in 19% of CTCL cases (38). This mutation was associated with NFAT activation, and suggests that calcineurin inhibitors may be a rationale therapeutic approach in these patients.
Diagnosis
Mycosis fungoides
The definitive diagnosis of MF, particularly patch/plaque stage disease, is challenging, as many of its clinical and pathologic features are non-specific. Many patients will have had symptoms attributed to eczema or parapsoriasis for years prior to obtaining a definitive diagnosis. The median time from symptom onset to diagnosis in retrospective series is 3–4 years, but may exceed four decades (102–104). Given the importance of clinicopathological correlation in the diagnosis of MF and the variable association of specific histologic findings with the diagnosis, biopsy reports are not infrequently “suggestive of” the diagnosis. This occasional uncertainty implied in biopsy reports and apparent lack of a more definitive histopathologic diagnosis may be a source of frustration for clinicians unfamiliar with the challenges associated with rendering a pathologic diagnosis of MF. While a definitive diagnosis of MF may be made on the basis of clinical and histopathologic features alone, determination of T-cell clonality and assessment for the aberrant loss of T-cell antigen expression by immunohistochemical staining for CD2, CD3, CD5 and CD7 are useful ancillary studies in the diagnosis of MF (and SS). PCR-based methods are able to detect clonal rearrangements of the T-cell receptor (TCR) in formalin-fixed, paraffin-embedded biopsy specimens (105, 106). PCR-based methods, while sensitive, should be interpreted with caution, as clonal TCR gene rearrangements may be detected in normal elderly individuals and in patients with benign dermatoses or other disease states (107–111). However, detection of identical clones from two different sites is quite specific for MF (112). The extent to which MF/SS may be preceded by a pre-malignant state, analogous to monoclonal B-cell lymphocytosis (MBL) or monoclonal gammopathy of undetermined significance (MGUS), is debatable and poorly defined (113). The malignant lymphocytes in MF/SS are usually CD3+CD4+ and CD8−, but frequently lose the expression of other pan-T-cell antigens. Therefore, demonstration of a significant population of cells lacking CD2, CD5 and/or CD7 expression, either within the entire lesion or the epidermis alone, is highly specific (specificity >90%) for MF in most reported series (114, 115). Clinically, patch/plaque stage MF is frequently characterized by persistent and progressive lesions that develop in a “bathing suit” distribution and vary in size, shape and color. These lesions are frequently large (>5 cm), pruritic and multifocal in “classical” MF. However, a broad range of MF variants have been described with differences in tropism (e.g. follicular MF), distribution (e.g. palmoplantar MF), pigmentation (e.g. hypo- and hyperpigmented variants) and focality (e.g. unilesional MF), some of which are formally recognized in the WHO-EORTC classification (1, 116). Given the need for uniform diagnostic criteria in MF, the International Society for Cutaneous Lymphoma (ISCL) recently proposed a point-based diagnostic algorithm which integrates clinical, histopathologic and immunophenotyping data with an assessment of T-cell clonality (117).
Sézary Syndrome
Traditionally, SS is defined as a leukemic form of CTCL associated with erythroderma. A series of studies in the early to mid-20th century, beginning with Sezary’s initial landmark observation in 1938, identified a population of large lymphocytes in the peripheral blood with grooved, lobulated (that is, “cerebriform”) nuclei in patients with MF or SS (118–123). As in other chronic lymphoproliferative disorders, the Sezary cell count is preferably expressed in absolute terms, with ≥1000 cells/μl classified as B2 disease in the current ISCL/EORTC TNMB staging classification. The morphologic detection of Sezary cells in the peripheral blood is not specific for CTCL, as Sezary cells may be found in peripheral blood from normal donors and in benign conditions (124–126). The histologic findings in the skin often resemble those observed in MF, with less prominent epidermotropism, while lymph node involvement is characterized by complete effacement of the nodal architecture by infiltrating Sezary cells (127).
In SS, clonal T cells are generally CD3+CD4+ and CD8− by multi-color flow cytometry (128–131). As in MF, the aberrant loss of pan-T-cell antigens, including CD2, CD3, CD4, CD5 and CD7 is frequently observed (130, 132–134). Of these, the aberrant loss of CD7 expression is most common, being observed in approximately two-thirds of cases (132, 135, 136). Loss of CD26 expression is also useful in the identification of Sezary cells, being observed in the majority of cases (131, 137–139). More recently, the aberrant expression of the MHC class I-binding, killer immunoglobulin-like receptor (KIR) CD158κ, normally expressed by natural killer cells, was described in the majority of patients examined with SS (140, 141). Molecular studies, including detection of a clonal TCR gene rearrangement by PCR and the presence of a clonal cytogenetic abnormality, provide evidence of T-cell clonality. An alternative approach to demonstrate T-cell clonality incorporates multi-color flow cytometry using a panel of antibodies specific for various TCR beta-chain variable region family members (TCR-Vβ) (142–144). This approach is successful in identifying a clonal population of T cells if this population is significantly higher than the background frequency of polyclonal T cells harboring the same Vβ chain (142, 143). Clark et. al. recently observed that lymphocytes isolated from either peripheral blood or skin lesions of CTCL patients contained a population of cells with high forward and side scatter characteristics on flow cytometric analysis (145). A similar population of so-called high-scatter T cells (THS) was not observed in samples obtained from patients with benign conditions. More importantly, these high-scatter T cells, upon careful immunophenotyping and analysis of clonal TCR-Vβ chain expression, were convincingly shown to represent the malignant T cell clone. While additional confirmatory studies are warranted, detection of high-scatter T cells may be an easily performed method to detect a clonal T-cell population in patients with limited-stage MF and to monitor the response to therapy.
The currently proposed ISCL criteria for SS integrate clinical, histologic, immunophenotyping and molecular studies. In patients with erythroderma, criteria recommended for the diagnosis of SS by the ISCL include the following: absolute sezary count ≥1000/μl, a CD4/CD8 ratio ≥10 (due to the clonal expansion of CD4+ cells), aberrant expression of pan-T-cell antigens, demonstration of T-cell clonality by Southern blot or PCR-based methods, or cytogenetic demonstration of an abnormal clone (130). At a minimum, the WHO-EORTC recommends the demonstration of T-cell clonality in combination with the above-mentioned criteria for the diagnosis of SS (1). In addition to the ISCL criteria, the most recent WHO classification requires erythroderma, generalized lymphadenopathy, and clonally related T-cells (Sézary cells) in the skin, peripheral blood, and lymph nodes. On rare occasions, SS may be preceded by a prior history of classic MF. The ISCL recommends that such cases be designated as “SS preceded by MF.” Conversely, patients with MF, but without erythroderma, may meet hematologic criteria for SS. In these cases, the designation “MF with leukemic involvement” is recommended.
Non-MF/SS subtypes of CTCL
An important goal during a patient’s initial diagnostic evaluation is to distinguish non-MF/SS CTCL subtypes from MF/SS, as the natural history, prognosis, and treatment approach for each of the non-MF/SS lymphomas is highly variable. A detailed description of these CTCL subtypes is beyond the scope of this update, but the salient features of each have been recently summarized (1, 146).
Risk-stratification
Staging
In contrast to many other lymphoproliferative disorders in which cytogenetic and laboratory findings play a prominent role in risk stratification, TNMB (tumor, node, metastasis, blood) staging remains an important prognostic factor in MF/SS and forms the basis for a “risk-adapted” approach to treatment. In 2007, the ISCL and EORTC revised the TNMB staging of MF/SS (147). Patients with only patches and plaques have stage I disease, but may be further divided into stage IA (<10% body surface area involved or T1) or stage IB (>10% body surface area involved or T2) based on the extent of skin involvement. For practical purposes, the area of one hand (including both palm and digits) represents approximately 1% of body surface area. Current staging and diagnostic recommendations do not require a biopsy of clinically normal lymph nodes; however, an excisional biopsy of any abnormal lymph nodes (≥1.5 cm in diameter or firm/fixed) is recommended, with preference being given either to the largest lymph node draining an area of skin involvement or to the node with the greatest standardized uptake value (SUV) on FDG-PET imaging. In current practice, two pathologic staging systems are used to classify the extent of nodal involvement. In the Dutch system, lymph nodes are pathologically graded based on the presence of large cerebriform nuclei (>7.5 μm) and the degree of architectural effacement (148). In contrast, the NCI-VA classification uses the relative number of atypical lymphocytes (not size), along with nodal architecture to determine the extent of nodal involvement (149, 150). Patients with patch/plaque stage disease (T1/T2) and architectural preservation of any clinically abnormal lymph nodes are classified as stage IIA. Collectively, patients with stage I-IIA disease have “limited-stage” disease, as the overall survival in these patients is measured in decades, with survival in patients with stage IA disease resembling that of normal age-matched controls (6, 102, 103). At diagnosis, the majority of MF patients will have limited-stage disease (6). In contrast, patients with tumor stage disease (T3), erythroderma (T4), nodal involvement characterized by partial or complete architectural effacement (N3), visceral metastases (M1), or significant leukemic involvement (B2) have “advanced-stage” disease. Detection of a clonal TCR gene rearrangement by PCR, which has been incorporated into the revised ISCL/EORTC node(N) and blood(B) staging classification, is an adverse prognostic factor (6, 151–154). Unfortunately, median survivals from approximately 1–5 years are observed in these patients with more extensive disease (6). The revised ISCL/EORTC staging for MF/SS is summarized in Table 1.
TABLE 1.
Stage | TNMB Classification | Median OS (years) | 10-year(6) | |||||
---|---|---|---|---|---|---|---|---|
T | N | M | B | OS (%) | DSS (%) | RDP (%) | ||
IA | 1 | 0 | 0 | 0,1 | 35.5 | 88 | 95 | 12 |
| ||||||||
IB | 2 | 0 | 0 | 0,1 | 21.5 | 70 | 77 | 38 |
| ||||||||
IIA | 1, 2 | 1 | 0 | 0,1 | 15.8 | 52 | 67 | 33 |
| ||||||||
IIB | 3 | 0–2 | 0 | 0,1 | 4.7 | 34 | 42 | 58 |
| ||||||||
IIIA | 4 | 0–2 | 0 | 0 | 4.7 | 37 | 45 | 62 |
| ||||||||
IIIB | 4 | 0–2 | 0 | 1 | 3.4 | 25 | 45 | 73 |
| ||||||||
IVA1 | 1–4 | 0–2 | 0 | 2 | 3.8 | 18 | 20 | 83 |
| ||||||||
IVA2 | 1–4 | 3 | 0 | 0–2 | 2.1 | 15 | 20 | 80 |
| ||||||||
IVB | 1–4 | 0–3 | 1 | 0–2 | 1.4 | 18 (5 year) | 18 (5 year) | 82 (5 year) |
OS, overall survival; DSS, disease-specific survival; RDP, risk of disease progression
A recently reported retrospective study which included 1,398 MF patients, 71% with patch/plaque stage disease, and 104 SS patients has validated the revised ISCL/EORTC staging classification (6). On univariate and multivariate analyses, the revised T, N, M and B classification were significantly associated with overall and disease-specific survival. The median survival, disease-specific survival and risk of disease progression, by clinical stage, are summarized in Table 1. In addition to staging, male gender, increasing age, an elevated LDH and the folliculotropic variant of MF were also independently associated with poorer overall and disease-specific survival. In contrast to previous reports highlighting the aggressive clinical course associated with large cell transformation (155–159), defined as the presence of large, atypical lymphocytes comprising at least 25% of the total lymphoid infiltrate, large cell transformation was not an independent predictor of overall or disease-specific survival, but was associated with a higher risk (hazard ratio 3.32) of disease progression (6). Given the importance of the TNMB classification in risk stratification and defining disease burden, the ISCL/EORTC recommends its use in defining the initial, maximum and current burden of disease, which will ultimately play an important role in the selection of either skin-directed or systemic therapies (147).
Recognizing that the staging system used for MF/SS is less helpful for non-MF/SS cutaneous lymphomas, a new TNM classification was also proposed for these CTCL variants (160). Due to the significant heterogeneity of these lymphomas, this staging system does not provide prognostic information, but is intended to provide a uniform description of the disease burden.
Cytogenetics
In contrast to some B-cell lymphoproliferative disorders, like chronic lymphocytic leukemia and multiple myeloma, for which gene-expression profiling and cytogenetic findings have important prognostic implications, risk-stratification in CTCL based on cytogenetic findings has only recently been described, is poorly understood, and consequently is not routinely performed in clinical practice.
Shin et. al. performed a gene expression profiling analysis on lesional skin biopsy specimens obtained from 62 CTCL patients and identified 3 distinct gene expression clusters that were prognostically important (50), that were later confirmed by RT-PCR analysis (161). The first cluster was associated with the upregulation of genes involved in T-cell activation, homing and tumor necrosis factor (TNF) signaling. This cluster conferred an inferior event-free survival when compared with the other two clusters. The second cluster, associated with the upregulation of genes involved in keratinocyte and epidermal proliferation and differentiation, was comprised largely of patients with limited-stage disease and was, not surprisingly, associated with superior event-free survival. Cluster 3, associated with an event-free survival intermediate between the first two clusters, was associated with the upregulation of genes involved in keratinocyte function and WNT signaling.
Array-comparative genomic hybridization techniques have revealed chromosomal copy number alterations that are prognostically relevant. First, an inverse association between survival and the absolute number of copy number alterations, reflecting genomic instability, has been observed in both tumor-stage MF and SS (162, 163). For example, in a cohort of 28 SS patients, the presence of fewer than 3 copy number alterations was associated with a median overall-survival of 93 months, compared with a median overall-survival of 67 months for those with 3 or more copy number alterations (162). In addition to genomic complexity, specific chromosomal gains/losses have also been associated with inferior survival. Unfortunately, many of these studies are small and hindered by the inclusion of multiple histologies. For example, in a cohort of 58 patients with transformed MF, SS or cutaneous anaplastic large cell lymphoma (cALCL), loss of the CDKN2A-CDKN2B locus (at 9p21) was associated with inferior overall survival that was highly significant. However, 9p21 loss was only found in a single patient with cALCL. Therefore, when these patients were omitted from analysis, the loss of 9p21 was associated with decreased overall survival that approached, but did not reach, statistical significance (96). Despite this, the adverse prognostic significance of 9p21 loss is supported by multiple patient cohorts including both MF and SS (25, 26, 163). Additional cytogenetic abnormalities, involving gains of chromosomes 1q and 8q and losses of chromosome 10q, have been associated with inferior survival (146).
Treatment of limited-stage MF
As the majority of CTCL patients present with patch/plaque stage MF and have an excellent prognosis, the initial goal of therapy is to improve symptoms and quality of life while avoiding treatment-related toxicity. For many patients, this may involve either expectant management (i.e. “watch and wait”) or skin-directed therapies. A randomized trial comparing early combined modality therapy, including both radiation and multiagent chemotherapy (cyclophosphamide, doxorubicin, etoposide, and vincristine), with sequential topical therapies demonstrated that combined-modality therapy, while associated with a superior complete response rate, did not translate into improvements in disease-free or overall survival and was associated with significant toxicity (164). The limited efficacy associated with chemotherapy was recently highlighted in a large retrospective study in which the median time to next treatment following single or multiagent chemotherapy was <4 months(165). Therefore, patients with limited-stage disease who require therapy are best approached with skin-directed therapies, usually under the direction of a dermatologist and/or radiation oncologist. Excellent reviews and treatment guidelines are available (146, 166–171).
Treatment of advanced-stage MF/SS
Overview
Patients with advanced-stage MF/SS require a multidisciplinary approach, as various combinations of skin-directed therapies, biologic-response modifiers and ultimately the sequential use of systemic chemotherapeutic agents are frequently employed in the management of these patients. As for limited-stage disease, multiagent chemotherapy, with only few exceptions, is generally not appropriate (164). A “risk-adapted” stage-based approach is adopted, with biologic-response modifiers (e.g. bexarotene and interferon-alpha) and histone deacetylase inhibitors (e.g. vorinostat) generally preferred prior to escalating therapy to include systemic chemotherapy (172). Therapeutic decisions are individualized and based on a patient’s age, performance status, extent of disease burden, the rate of disease progression, and previous therapies (166–171).
Bexarotene
The endogenous retinoids all-trans retinoic acid and 9-cis retinoic acid (i.e. vitamin-A-derived compounds) regulate a diverse array of biologic processes, ranging from embryonic development to cell growth, differentiation and survival, upon binding two families of steroid hormone receptors, the retinoic acid receptors (RAR) and retinoid X receptors (RXR). Upon forming homo- or heterodimers, these receptors recruit various nuclear co-repressor or co-activator proteins depending whether or not they are bound by ligand. Multiple RAR retinoids have been used in MF/SS, either topically or systemically (reviewed in (173, 174)), with response rates exceeding 50%. However, in 1999 the oral RXR-selective “rexinoid” bexarotene was FDA approved for CTCL and was later approved as a topical gel formulation. Laboratory studies demonstrate that bexarotene promotes cell cycle arrest and apoptosis in CTCL cell lines (175, 176). In a multicenter phase II-III study, 94 patients with advanced-stage CTCL who had been previously treated with a median of five prior therapies, the vast majority of whom had disease refractory to at least one prior systemic therapy, received at least 300 mg/m2 of oral bexarotene daily (177). Among patients treated at the 300 mg/m2 dose, an overall response rate of 45% was observed, only 2% of which were complete. While an improved overall response rate was noted with the use of higher doses, this difference was not statistically significant, and dose-limiting toxicity was far more common (50% vs. 89%) in these patients. While a dose-response relationship is likely, the 300 mg/m2 dose appears to provide the optimal risk-benefit ratio. The most common toxicities associated with therapy were hypertriglyceridemia (in 82%) and central hypothyroidism (29%). Myelosuppression is infrequent and usually uncomplicated. Pancreatitis secondary to hypertriglyceridemia may be rarely observed, but is reversible upon discontinuation of treatment. Therefore, a baseline lipid panel and TSH should be obtained prior to the initiation of therapy. In one retrospective study, all patients treated with bexarotene developed hyperlipidemia and hypothyroidism, frequently within weeks of initiating treatment (178). Consequently, use of lipid-lowering agents (e.g. fenofibrate) and low-dose levothyroxine (e.g. 50 micrograms) prior to initiating bexarotene is generally recommended (179–181). In clinical practice, bexarotene is frequently initiated at a lower dose of 150 mg/m2 and subsequently titrated to full doses after 4 weeks of therapy, depending upon patient tolerability. Most responses occur within 2–3 months of treatment initiation, but may be delayed. Therefore, in the absence of disease progression or toxicity, treatment should be continued for up to 6 months. For responding patients, treatment should be continued until disease progression and, depending upon the quality of the response, adjunctive skin-directed therapies (e.g. PUVA, interferon) should be considered (182). Guidelines describing appropriate laboratory monitoring, supportive care, and safe clinical prescribing of bexarotene have been recently published (181). Future studies clarifying the optimal use of bexarotene, either in combination or sequentially with other agents, are needed.
HDAC inhibitors
Histone deacetylases (HDACs) catalyze the removal of acetyl groups from both histone and non-histone proteins. As histone acetylation is associated with an open chromatin configuration associated with active gene transcription, HDACs contribute to histone deacetylation and the epigenetic repression of gene transcription. As HDACs regulate a wide variety of processes involved in carcinogenesis, multiple mechanisms may explain the clinical activity of HDAC inhibitors (183, 184), including altered gene expression of cell-cycle and apoptotic regulatory proteins (185–189), acetylation of non-histone proteins regulating cell growth and survival (190–193), angiogenesis (194, 195), aggresome formation (196) and DNA repair (197). In addition, HDAC inhibitors may have important effects on the tumor microenvironment via reactive oxygen species (198, 199), enhanced antigen presentation (200) and downregulation of immunomodulatory cytokines, like IL-10 (201).
Vorinostat (suberoylanilide hydroxamic acid, SAHA) and romidepsin (depsipeptide) inhibit class I and II HDACs (i.e. pan-HDAC inhibitors), the former being widely expressed in various lymphoma subtypes (202). Early phase I studies of both vorinostat and romidepsin established their safety and potential efficacy in lymphoproliferative disorders, including CTCL (203), thus paving the way for larger phase II studies. An earlier phase II study established 400 mg of oral vorinostat once daily as the optimal dose that was investigated further in 74 previously treated patients with CTCL, most of whom (>80%) had advanced-stage disease (204, 205). The overall response rate was approximately 30% for patients with advanced-stage disease and was associated with a median duration of response estimated to exceed 185 days. Most responses were rapid (i.e. <2 months) and were also noted in patients with tumor-stage disease and Sézary syndrome (206). Patients who failed to achieve an objective response appeared to derive some clinical benefit, including stable disease, decreased lymphadenopathy and pruritis relief, with treatment. The most common non-hematologic adverse events, observed in almost 50% of patients, were gastrointestinal toxicities (nausea, vomiting, diarrhea). Hematologic toxicities, including anemia or thrombocytopenia, were observed in up to 20% of patients. Among responding patients, long-term therapy with vorinostat appears to be well tolerated (207). Prolongation of the QT interval was rarely observed, but monitoring and appropriate electrolyte replacement is recommended for those patients at risk for QT prolongation (208).
Romidepsin, administered as a 4-hour intravenous infusion (14 mg/m2) days 1, 8 and 15 every 4 weeks, was evaluated in two phase II studies, the largest of which included 96 patients, most with advanced-stage disease(209, 210). The overall response rate was 38% for patients with advanced-stage disease, with a median duration of response that exceeded one year. A toxicity profile similar to that described for vorinostat was observed. Intensive cardiac monitoring in a subset of these patients failed to demonstrate any clinically significant cardiotoxicity (211).
Additional HDAC inhibitors, including potent pan-HDAC inhibitors, appear to have activity in CTCL (189, 212, 213). Further studies are needed to fully define the mechanisms of resistance to HDAC inhibition in CTCL (189, 214–218), enabling the development of rational therapeutic combinations incorporating HDAC inhibitors in CTCL (219, 220).
Interferon-alpha
Interferon-alpha (i.e. interferon-alpha 2b), a type I interferon with immunomodulatory properties, has pleiotropic effects in CTCL and is associated with an overall response rate of 50–70% and a complete response rate of 20–30%, particularly in patients with limited-stage disease (221–224). While often considered as second-line therapy for limited-stage CTCL, interferon-alpha, frequently at doses ranging from 3–10 million units daily to three times weekly, is a treatment to be considered in the first-line setting in patients with advanced-stage disease. Responses, which may be achieved within a few months, are observed in patients with tumor-stage MF and SS, and are occasionally durable(165, 225). Furthermore, interferon-alpha may be successfully combined with a number of other therapeutic modalities frequently utilized in the management of these patients, including PUVA, bexarotene, chemotherapy and ECP (226–239). For example, in a cohort of 51, mostly advanced-stage patients treated with single-agent, low-dose, interferon-alpha, responses were observed in 34 (67%), including 21 (41%) with a complete response and 9 with a long-term remission (224). Similarly, in a cohort of 47 patients with stage III/IV disease, 89% of whom had peripheral blood involvement, a response rate exceeding 80% was observed in those treated with a combination of ECP and interferon-alpha (239). Interferon-alpha is associated with myelosuppression, transaminitis and dose-limiting flu-like side effects, particularly at higher doses.
Extracorporeal photophoresis
During extracorporeal photophoresis (ECP) pooled leukapheresis and plasmapheresis products are exposed to 8-methoxypsoralen (8-MOP) prior to extracorporeal circulation through a 1 mm thick disposable cassette exposed to UVA radiation. The irradiated leukocytes, representing approximately 5% of peripheral blood leukocytes, are subsequently reinfused. Psoralen covalently binds and crosslinks DNA following UVA exposure, leading to the induction of apoptosis in the majority of treated lymphocytes by multiple mechanisms involving bcl-2 family members, disruption of the mitochondrial membrane potential and extrinsic cell death pathways (240–242). In contrast, ECP leads to monocyte activation, including significant changes in gene expression (243), and dendritic cell differentiation, which is thought to culminate in enhanced antigen presentation and the initiation of a host immune response (244). In hopes of prolonging the exposure time between monocyte-derived dendritic cells and malignant lymphocytes undergoing apoptosis, investigators have developed a modified ECP protocol (i.e. “transimmunization”) whereby blood products are incubated overnight following UVA irradiation and prior to patient infusion (245). This novel adaptation is investigational and has not been widely employed given concerns about infectious risks and lack of a proven increase in efficacy.
Following the landmark study by Edelson and colleagues describing responses in 27 out of 37 patients with erythrodermic CTCL treated with ECP, ECP was approved by the Food and Drug Administration of the USA for the treatment of CTCL and is now considered the treatment of choice in the first-line management of patients with Sézary syndrome in many centers (246). While responses vary between case series, overall response rates hover around 60%, with a complete response rate of approximately 20% (247–250). As current treatment protocols no longer require the oral administration of 8-MOP, eliminating nausea, ECP is safe and generally very well tolerated. While alternative schedules have been investigated, ECP is generally performed for 2 consecutive days every 2–4 weeks. While the precise mechanism of action is incompletely understood, evidence suggests that ECP has immunomodulatory effects which may augment host anti-tumor immunity. It is not surprising then that the median time to response following the initiation of ECP is approximately 6 months. Median survival exceeding 8 years has been observed in ECP treated patients and among complete responders, many experience durable responses which may permit, for some, weaning from CTCL-directed therapies (247, 251–253). While patient- or disease-specific factors which may predict a response to therapy are imperfect, patients for whom treatment is initiated promptly after diagnosis who have circulating Sézary cells, but without significant nodal or visceral disease, may be more likely to respond. In addition, patients without profound immune deficiencies, reflected by normal or near-normal cytotoxic T-cell and CD4/CD8 values and the absence of prior exposure to systemic chemotherapy, may be more likely to respond to therapy (247, 249, 252). While effective as monotherapy, ECP has also been combined with other therapeutic strategies, including interferon, bexarotene and TSEBT (229, 239, 251, 254–256).
Monoclonal antibodies
In contrast to many B-cell lymphoproliferative disorders, where the incorporation of CD20-targeting monoclonal antibodies has become the standard of care, additional studies are needed to identify the optimal approach targeting T-cell specific antigens in advanced-stage MF/SS. Alemtuzumab is a humanized IgG1 monoclonal antibody directed against CD52, an antigen widely expressed by B-cells, T-cells and monocytes (257). In a phase II study in 22 patients with advanced-stage MF/SS, overall and complete response rates of 55% and 32%, respectively, were observed, with a median time to treatment failure of 1 year (258). Given the significant risk of infectious complications, low-dose subcutaneous alemtuzumab was investigated in 14 patients with SS, most of whom had relapsed/refractory disease (259). Most patients in this study received 3 mg of subcutaneous alemtuzumab on day 1 followed by a 10 mg dose on alternating days until the Sézary count was <1000/mm3. With the exception of a single patient whose best response was stable disease, 9 out of 10 patients treated in this manner achieved a response, 3 of which were complete. For most patients, the time to treatment failure exceeded 12 months. What is notable, however, is that infectious complications were not observed in patients treated with the lowest dose (i.e. 10 mg) of alemtuzumab. Similar results, with no infectious complications, were recently reported in a small cohort of patients treated with modified, low-dose, subcutaneous alemtuzumab for six weeks (260). In addition to hematologic toxicity, conventionally dosed alemtuzumab in advanced-stage MF/SS is associated with a high incidence of infectious complications (258, 259, 261–264). Overall, infectious complications have been observed in two-thirds of treated patients, most of which are bacterial, including sepsis. Cytomegalovirus (CMV) reactivation is the most common viral infection. In addition, Pneumocystis jirovecii pneumonia and invasive fungal infections have also been observed. Therefore, trimethoprim-sulphamethoxazole and acyclovir should be routinely administered for PJP and HSV/VZV prophylaxis, respectively, in patients receiving alemtuzumab. In addition, CMV surveillance should be performed every 1–2 weeks by quantitative PCR and suppressive therapy with ganciclovir or oral valganciclovir initiated in response to viral reactivation. Low-dose, subcutaneous alemtuzumab appears to be safe and efficacious in selected patients with advanced-stage MF/SS provided with appropriate supportive care. Monoclonal antibodies targeting additional T-cell specific antigens, including CD2 (265), CD4 (266), CD25 (267) and CCR4 (268–270) are being explored and appear promising. Mogamulizumab (KW-0761) is a humanized monoclonal antibody specific for the chemokine receptor CCR4 that has been defucosylated and is consequently associated with enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). In a phase I/2 study, mogamulizumab was well tolerated and was associated with an overall response rate of 37%. A similar response rate of 29% (2/7), all partial, was observed in a phase II Japanese study (270, 271). In addition to ADCC-mediated clearance of malignant T cells, mogamulizumab may inhibit Treg-mediate immune suppression (272, 273), and may warrant further investigation with immunomodulatory therapies, including immune checkpoint blockade (274). A randomized, phase III clinical trial comparing mogamulizumab and vorinostat in relapsed/refractory CTCL is ongoing in the US (NCT01728805). While capable of binding skin-resident T cells, monoclonal antibodies like mogamulizumab and alemtuzumab may be most efficacious in MF/SS patients with recirculating (and TMM or TCM-derived) clones (20). Brentuximab vedotin is an antibody-drug conjugate in which an anti-CD30 monoclonal antibody is linked with an anti-tubulin agent (monomethyl auristatin E). In a phase II study, 19 patients with relapsed/refractory MF received brentuximab vedotin. Among the 13 patients with stage IB or IIB disease, a response rate of 92% (all partial) was observed (275). As a single partial response was observed among the 6 patients with Stage IV disease, an overall response rate of 68% for the entire cohort was observed. Interestingly, quantitative image analysis for CD30 expression demonstrated CD30 positivity in all cases available for review, including those that were deemed CD30 negative by conventional immunohistochemistry. The response to brentuximab vedotin was not associated with CD30 expression in this cohort. As anticipated, neuropathy was the most common toxicity observed. A randomized, phase III clinical trial comparing brentuximab vedotin with an investigator’s choice (methotrexate or bexarotene) is ongoing (NCT01578499). Resimmune, a second-generation immunotoxin in which the catalytic and translocation domains of diphtheria toxin (DT390) have been fused to CD3-specific single chain antibody fragments [bisFv(UCHT1)], is associated with a response rate of 36% (16% complete), and is particularly active in patients with limited-stage disease (276). Much like its predecessor, resimmune is associated with a vascular leak syndrome (146).
Systemic Chemotherapy
Systemic chemotherapy is generally reserved for patients with advanced-stage MF/SS who have either relapsed following therapy with skin-directed therapies and the biologic-response modifiers described above or have extensive disease with visceral organ involvement. Multiple chemotherapeutic agents, including single-agent and combination chemotherapy regimens, while associated with high response rates in MF/SS (167, 169, 277), are infrequently durable (165), and frequently associated with significant myelosuppression and infectious complications (165, 278–280). Therefore, with the exceptions of refractory disease or in the setting of extensive or rapidly progressive disease where a rapid treatment response may be necessary, the administration of sequential, single-agent chemotherapy is preferred. Many oral and intravenous chemotherapeutic agents have been utilized in MF/SS (281–301). Unfortunately, the duration of response with these agents is frequently measured in months. Therefore, novel therapeutic agents, either alone or in combination, are needed.
Pralatrexate, a novel antifolate with a high affinity for the reduced folate carrier (RFC-1) and novel mechanism of resistance when compared with methotrexate (302–304), was associated with an overall response rate of 29% in the PROPEL study. This study was comprised largely of peripheral T-cell lymphoma patients, most of whom had refractory disease (305). Notably, twelve patients with transformed MF were included in the study (306). Many of these patients had received more than 5 prior systemic therapies, including CHOP or CHOP-like regimens. With only a single exception, these patients were refractory to their most recent therapy. Responses, as assessed by the study investigators, were observed in 58% of patients with a median duration of response and progression-free survival of 4–5 months. Results of a dose-finding study were reported in a larger cohort of CTCL patients (307). In this study, the optimal dose was identified as 15 mg/m2, given weekly 3 weeks out of 4, and was associated with an overall response rate of 43%. In an effort to reduce the incidence of mucositis, folic acid and vitamin B12 supplementation is routinely provided in these patients (308). Additional therapeutic approaches, including proteasome inhibition (309), immunomodulatory strategies (310), and more targeted approaches warrant further investigation (311). As there is no standard of care for patients with MF/SS requiring systemic chemotherapy and the decision to initiate therapy is individualized, including consideration of responses and complications related to prior therapies, participation in a well-designed clinical trial is always worth consideration.
High-dose chemotherapy and hematopoietic stem cell transplantation
The available experience with high-dose chemotherapy and autologous stem cell transplantation, largely confined to case series, suggests that responses following treatment are frequently transient. In contrast, the durable remissions observed following allogeneic transplantation may be explained by the graft versus lymphoma immune response (312, 313). A retrospective analysis of 60 patients with advanced-stage MF/SS who underwent allogeneic stem cell transplantation was recently reported (314). In this series, patients had received a median of 4 prior therapies prior to undergoing either reduced-conditioning (73%) or myeloablative (27%) conditioning prior to related (75%) or matched-unrelated donor (25%) transplantation. Non-relapse mortality at 1 year was 14% for patients receiving reduced-intensity conditioning or HLA identical/related donor stem cells and 38–40% for those undergoing myeloablative conditioning or receiving match-unrelated donor grafts. Transplantation during an early phase of disease (defined as first or second remission or relapse following 3 or fewer systemic therapies) was associated with lower relapse rates (25% vs. 44% at 1 year) and a statistically insignificant increase in 3-year overall survival (68% vs. 46%). Given the differences in non-relapse mortality, both reduced-intensity conditioning and use of matched-related donors were associated with superior overall survival (63% at 3 years). Seventeen out of 26 patients who relapsed received donor-lymphocyte infusions. Of these, 47% achieved a complete remission, thus providing evidence for a graft-versus-lymphoma effect in MF/SS. In contrast to the experience with B-cell non-Hodgkin lymphomas, chemotherapy sensitivity prior to transplantation or the extent of disease burden did not influence overall survival. The estimated 3-year progression-free and overall survival were 34% and 53%, respectively. Given the possibility of complete and durable remissions, allogeneic stem-cell transplantation in conjunction with total skin electron beam therapy may be considered in selected patients (225, 315).
Summary
Establishing a definitive diagnosis of CTCL, accurate disease staging and risk-stratification, and the selection of appropriate therapy requires a multidisciplinary approach. While high response rates may be achieved with systemic chemotherapy, these responses are frequently short-lived and associated with significant toxicities. As treatment of advanced-stage MF/SS is largely palliative, a stage-based approach utilizing sequential therapies in an escalated fashion is preferred. Participation in a well-designed clinical trial is encouraged, as the introduction of novel agents will continue to expand the therapeutic options available in the management of CTCL.
Acknowledgments
This work was supported in part by the National Institutes of Health (K08CA172215) and the Leukemia and Lymphoma Society Translational Research Program.
Footnotes
Conflict of interest: Nothing to report
References
- 1.Willemze R, Jaffe ES, Burg G, Cerroni L, Berti E, Swerdlow SH, Ralfkiaer E, Chimenti S, Diaz-Perez JL, Duncan LM, Grange F, Harris NL, Kempf W, Kerl H, Kurrer M, Knobler R, Pimpinelli N, Sander C, Santucci M, Sterry W, Vermeer MH, Wechsler J, Whittaker S, Meijer CJ. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:3768–3785. doi: 10.1182/blood-2004-09-3502. [DOI] [PubMed] [Google Scholar]
- 2.Criscione VD, Weinstock MA. Incidence of cutaneous T-cell lymphoma in the United States, 1973–2002. Arch Dermatol. 2007;143:854–859. doi: 10.1001/archderm.143.7.854. [DOI] [PubMed] [Google Scholar]
- 3.Bradford PT, Devesa SS, Anderson WF, Toro JR. Cutaneous lymphoma incidence patterns in the United States: a population-based study of 3884 cases. Blood. 2009;113:5064–5073. doi: 10.1182/blood-2008-10-184168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Burns MK, Ellis CN, Cooper KD. Mycosis fungoides–type cutaneous T-cell lymphoma arising before 30 years of age. Immunophenotypic, immunogenotypic and clinicopathologic analysis of nine cases. J Am Acad Dermatol. 1992;27:974–978. doi: 10.1016/0190-9622(92)70297-s. [DOI] [PubMed] [Google Scholar]
- 5.Pope E, Weitzman S, Ngan B, Walsh S, Morel K, Williams J, Stein S, Garzon M, Knobler E, Lieber C, Turchan K, Wargon O, Tsuchiya A. Mycosis fungoides in the pediatric population: report from an international Childhood Registry of Cutaneous Lymphoma. J Cutan Med Surg. 2010;14:1–6. doi: 10.2310/7750.2009.08091. [DOI] [PubMed] [Google Scholar]
- 6.Agar NS, Wedgeworth E, Crichton S, Mitchell TJ, Cox M, Ferreira S, Robson A, Calonje E, Stefanato CM, Wain EM, Wilkins B, Fields PA, Dean A, Webb K, Scarisbrick J, Morris S, Whittaker SJ. 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. Journal of Clinical Oncology. 28:4730–4739. doi: 10.1200/JCO.2009.27.7665. [DOI] [PubMed] [Google Scholar]
- 7.Whittemore AS, Holly EA, Lee IM, Abel EA, Adams RM, Nickoloff BJ, Bley L, Peters JM, Gibney C. Mycosis fungoides in relation to environmental exposures and immune response: a case-control study. J Natl Cancer Inst. 1989;81:1560–1567. doi: 10.1093/jnci/81.20.1560. [DOI] [PubMed] [Google Scholar]
- 8.Magro CM, Crowson AN, Kovatich AJ, Burns F. Drug-induced reversible lymphoid dyscrasia: a clonal lymphomatoid dermatitis of memory and activated T cells. Hum Pathol. 2003;34:119–129. doi: 10.1053/hupa.2003.4. [DOI] [PubMed] [Google Scholar]
- 9.Jahan-Tigh RR, Huen AO, Lee GL, Pozadzides JV, Liu P, Duvic M. Hydrochlorothiazide and cutaneous T cell lymphoma: prospective analysis and case series. Cancer. 2013;119:825–831. doi: 10.1002/cncr.27740. [DOI] [PubMed] [Google Scholar]
- 10.Hodak E, Klein T, Gabay B, Ben-Amitai D, Bergman R, Gdalevich M, Feinmesser M, Maron L, David M. Familial mycosis fungoides: report of 6 kindreds and a study of the HLA system. J Am Acad Dermatol. 2005;52:393–402. doi: 10.1016/j.jaad.2003.12.052. [DOI] [PubMed] [Google Scholar]
- 11.Hodak E, Lapidoth M, Kohn K, David D, Brautbar B, Kfir K, Narinski N, Safirman S, Maron M, Klein K. Mycosis fungoides: HLA class II associations among Ashkenazi and non-Ashkenazi Jewish patients. Br J Dermatol. 2001;145:974–980. doi: 10.1046/j.1365-2133.2001.04496.x. [DOI] [PubMed] [Google Scholar]
- 12.Jackow CM, McHam JB, Friss A, Alvear J, Reveille JR, Duvic M. HLA-DR5 and DQB1*03 class II alleles are associated with cutaneous T-cell lymphoma. J Invest Dermatol. 1996;107:373–376. doi: 10.1111/1523-1747.ep12363352. [DOI] [PubMed] [Google Scholar]
- 13.Tuyp E, Burgoyne A, Aitchison T, MacKie R. A case-control study of possible causative factors in mycosis fungoides. Arch Dermatol. 1987;123:196–200. [PubMed] [Google Scholar]
- 14.Wohl Y, Tur E. Environmental risk factors for mycosis fungoides. Curr Probl Dermatol. 2007;35:52–64. doi: 10.1159/000106410. [DOI] [PubMed] [Google Scholar]
- 15.Morales Suarez-Varela MM, Olsen J, Kaerlev L, Guenel P, Arveux P, Wingren G, Hardell L, Ahrens W, Stang A, Llopis-Gonzalez A, Merletti F, Guillen-Grima F, Johansen P. Are alcohol intake and smoking associated with mycosis fungoides? A European multicentre case-control study Eur J Cancer. 2001;37:392–397. doi: 10.1016/s0959-8049(00)00383-x. [DOI] [PubMed] [Google Scholar]
- 16.Morales-Suarez-Varela MM, Olsen J, Johansen P, Kaerlev L, Guenel P, Arveux P, Wingren G, Hardell L, Ahrens W, Stang A, Llopis A, Merletti F, Guillen-Grima F, Masala G. Occupational sun exposure and mycosis fungoides: a European multicenter case-control study. J Occup Environ Med. 2006;48:390–393. doi: 10.1097/01.jom.0000194160.95468.20. [DOI] [PubMed] [Google Scholar]
- 17.Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K, Dowgiert RK, Kupper TS. The vast majority of CLA+ T cells are resident in normal skin. J Immunol. 2006;176:4431–4439. doi: 10.4049/jimmunol.176.7.4431. [DOI] [PubMed] [Google Scholar]
- 18.Reiss Y, Proudfoot AE, Power CA, Campbell JJ, Butcher EC. CC chemokine receptor (CCR)4 and the CCR10 ligand cutaneous T cell-attracting chemokine (CTACK) in lymphocyte trafficking to inflamed skin. J Exp Med. 2001;194:1541–1547. doi: 10.1084/jem.194.10.1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Homey B, Alenius H, Muller A, Soto H, Bowman EP, Yuan W, McEvoy L, Lauerma AI, Assmann T, Bunemann E, Lehto M, Wolff H, Yen D, Marxhausen H, To W, Sedgwick J, Ruzicka T, Lehmann P, Zlotnik A. CCL27-CCR10 interactions regulate T cell-mediated skin inflammation. Nat Med. 2002;8:157–165. doi: 10.1038/nm0202-157. [DOI] [PubMed] [Google Scholar]
- 20.Watanabe R, Gehad A, Yang C, Scott LL, Teague JE, Schlapbach C, Elco CP, Huang V, Matos TR, Kupper TS, Clark RA. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Science translational medicine. 2015;7:279ra239. doi: 10.1126/scitranslmed.3010302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Frost EL, Kersh AE, Evavold BD, Lukacher AE. Cutting Edge: Resident Memory CD8 T Cells Express High-Affinity TCRs. J Immunol. 2015;195:3520–3524. doi: 10.4049/jimmunol.1501521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, Vega-Ramos J, Lauzurica P, Mueller SN, Stefanovic T, Tscharke DC, Heath WR, Inouye M, Carbone FR, Gebhardt T. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol. 2013;14:1294–1301. doi: 10.1038/ni.2744. [DOI] [PubMed] [Google Scholar]
- 23.Clark RA, Watanabe R, Teague JE, Schlapbach C, Tawa MC, Adams N, Dorosario AA, Chaney KS, Cutler CS, Leboeuf NR, Carter JB, Fisher DC, Kupper TS. Skin effector memory T cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients. Science translational medicine. 2012;4:117ra117. doi: 10.1126/scitranslmed.3003008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.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: 10.1182/blood-2009-11-251926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Laharanne E, Oumouhou N, Bonnet F, Carlotti M, Gentil C, Chevret E, Jouary T, Longy M, Vergier B, Beylot-Barry M, Merlio JP. Genome-wide analysis of cutaneous T-cell lymphomas identifies three clinically relevant classes. J Invest Dermatol. 2010;130:1707–1718. doi: 10.1038/jid.2010.8. [DOI] [PubMed] [Google Scholar]
- 26.van Doorn R, van Kester MS, Dijkman R, Vermeer MH, Mulder AA, Szuhai K, Knijnenburg J, Boer JM, Willemze R, Tensen CP. Oncogenomic analysis of mycosis fungoides reveals major differences with Sezary syndrome. Blood. 2009;113:127–136. doi: 10.1182/blood-2008-04-153031. [DOI] [PubMed] [Google Scholar]
- 27.Heid JB, Schmidt A, Oberle N, Goerdt S, Krammer PH, Suri-Payer E, Klemke CD. FOXP3+CD25− tumor cells with regulatory function in Sezary syndrome. J Invest Dermatol. 2009;129:2875–2885. doi: 10.1038/jid.2009.175. [DOI] [PubMed] [Google Scholar]
- 28.Krejsgaard T, Odum N, Geisler C, Wasik MA, Woetmann A. Regulatory T cells and immunodeficiency in mycosis fungoides and Sezary syndrome. Leukemia. 2012;26:424–432. doi: 10.1038/leu.2011.237. [DOI] [PubMed] [Google Scholar]
- 29.Wilcox RA, Wada DA, Ziesmer SC, Elsawa SF, Comfere NI, Dietz AB, Novak AJ, Witzig TE, Feldman AL, Pittelkow MR, Ansell SM. Monocytes promote tumor cell survival in T-cell lymphoproliferative disorders and are impaired in their ability to differentiate into mature dendritic cells. Blood. 2009;114:2936–2944. doi: 10.1182/blood-2009-05-220111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Berger CL, Tigelaar R, Cohen J, Mariwalla K, Trinh J, Wang N, Edelson RL. Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells. Blood. 2005;105:1640–1647. doi: 10.1182/blood-2004-06-2181. [DOI] [PubMed] [Google Scholar]
- 31.Clark RA. Skin-resident T cells: the ups and downs of on site immunity. J Invest Dermatol. 2010;130:362–370. doi: 10.1038/jid.2009.247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wang T, Feldman AL, Wada DA, Lu Y, Polk A, Briski R, Ristow K, Habermann TM, Thomas D, Ziesmer SC, Wellik LE, Lanigan TM, Witzig TE, Pittelkow MR, Bailey NG, Hristov AC, Lim MS, Ansell SM, Wilcox RA. GATA-3 expression identifies a high-risk subset of PTCL, NOS with distinct molecular and clinical features. Blood. 2014;123:3007–3015. doi: 10.1182/blood-2013-12-544809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Vowels BR, Lessin SR, Cassin M, Jaworsky C, Benoit B, Wolfe JT, Rook AH. Th2 cytokine mRNA expression in skin in cutaneous T-cell lymphoma. J Invest Dermatol. 1994;103:669–673. doi: 10.1111/1523-1747.ep12398454. [DOI] [PubMed] [Google Scholar]
- 34.Vowels BR, Cassin M, Vonderheid EC, Rook AH. Aberrant cytokine production by Sezary syndrome patients: cytokine secretion pattern resembles murine Th2 cells. J Invest Dermatol. 1992;99:90–94. doi: 10.1111/1523-1747.ep12611877. [DOI] [PubMed] [Google Scholar]
- 35.Suchin KR, Cassin M, Gottleib SL, Sood S, Cucchiara AJ, Vonderheid EC, Rook AH. Increased interleukin 5 production in eosinophilic Sezary syndrome: regulation by interferon alfa and interleukin 12. J Am Acad Dermatol. 2001;44:28–32. doi: 10.1067/mjd.2001.109853. [DOI] [PubMed] [Google Scholar]
- 36.Kari L, Loboda A, Nebozhyn M, Rook AH, Vonderheid EC, Nichols C, Virok D, Chang C, Horng WH, Johnston J, Wysocka M, Showe MK, Showe LC. Classification and prediction of survival in patients with the leukemic phase of cutaneous T cell lymphoma. J Exp Med. 2003;197:1477–1488. doi: 10.1084/jem.20021726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Nebozhyn M, Loboda A, Kari L, Rook AH, Vonderheid EC, Lessin S, Berger C, Edelson R, Nichols C, Yousef M, Gudipati L, Shang M, Showe MK, Showe LC. Quantitative PCR on 5 genes reliably identifies CTCL patients with 5% to 99% circulating tumor cells with 90% accuracy. Blood. 2006;107:3189–3196. doi: 10.1182/blood-2005-07-2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Vaque JP, Gomez-Lopez G, Monsalvez V, Varela I, Martinez N, Perez C, Dominguez O, Grana O, Rodriguez-Peralto JL, Rodriguez-Pinilla SM, Gonzalez-Vela C, Rubio-Camarillo M, Martin-Sanchez E, Pisano DG, Papadavid E, Papadaki T, Requena L, Garcia-Marco JA, Mendez M, Provencio M, Hospital M, Suarez-Massa D, Postigo C, San Segundo D, Lopez-Hoyos M, Ortiz-Romero PL, Piris MA, Sanchez-Beato M. PLCG1 mutations in cutaneous T-cell lymphomas. Blood. 2014;123:2034–2043. doi: 10.1182/blood-2013-05-504308. [DOI] [PubMed] [Google Scholar]
- 39.Kasprzycka M, Zhang Q, Witkiewicz A, Marzec M, Potoczek M, Liu X, Wang HY, Milone M, Basu S, Mauger J, Choi JK, Abrams JT, Hou JS, Rook AH, Vonderheid E, Woetmann A, Odum N, Wasik MA. Gamma c-signaling cytokines induce a regulatory T cell phenotype in malignant CD4+ T lymphocytes. J Immunol. 2008;181:2506–2512. doi: 10.4049/jimmunol.181.4.2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Adachi T, Kobayashi T, Sugihara E, Yamada T, Ikuta K, Pittaluga S, Saya H, Amagai M, Nagao K. Hair follicle-derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat Med. 2015 doi: 10.1038/nm.3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dalloul A, Laroche L, Bagot M, Mossalayi MD, Fourcade C, Thacker DJ, Hogge DE, Merle-Beral H, Debre P, Schmitt C. Interleukin-7 is a growth factor for Sezary lymphoma cells. J Clin Invest. 1992;90:1054–1060. doi: 10.1172/JCI115920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Berger CL, Hanlon D, Kanada D, Dhodapkar M, Lombillo V, Wang N, Christensen I, Howe G, Crouch J, El-Fishawy P, Edelson R. The growth of cutaneous T-cell lymphoma is stimulated by immature dendritic cells. Blood. 2002;99:2929–2939. [PubMed] [Google Scholar]
- 43.Yamanaka K, Clark R, Rich B, Dowgiert R, Hirahara K, Hurwitz D, Shibata M, Mirchandani N, Jones DA, Goddard DS, Eapen S, Mizutani H, Kupper TS. Skin-derived interleukin-7 contributes to the proliferation of lymphocytes in cutaneous T-cell lymphoma. Blood. 2006;107:2440–2445. doi: 10.1182/blood-2005-03-1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.McCusker ME, Garifallou M, Bogen SA. Sezary lineage cells can be induced to proliferate via CD28-mediated costimulation. J Immunol. 1997;158:4984–4991. [PubMed] [Google Scholar]
- 45.Dave SS, Wright G, Tan B, Rosenwald A, Gascoyne RD, Chan WC, Fisher RI, Braziel RM, Rimsza LM, Grogan TM, Miller TP, LeBlanc M, Greiner TC, Weisenburger DD, Lynch JC, Vose J, Armitage JO, Smeland EB, Kvaloy S, Holte H, Delabie J, Connors JM, Lansdorp PM, Ouyang Q, Lister TA, Davies AJ, Norton AJ, Muller-Hermelink HK, Ott G, Campo E, Montserrat E, Wilson WH, Jaffe ES, Simon R, Yang L, Powell J, Zhao H, Goldschmidt N, Chiorazzi M, Staudt LM. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med. 2004;351:2159–2169. doi: 10.1056/NEJMoa041869. [DOI] [PubMed] [Google Scholar]
- 46.Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermelink HK, Smeland EB, Giltnane JM, Hurt EM, Zhao H, Averett L, Yang L, Wilson WH, Jaffe ES, Simon R, Klausner RD, Powell J, Duffey PL, Longo DL, Greiner TC, Weisenburger DD, Sanger WG, Dave BJ, Lynch JC, Vose J, Armitage JO, Montserrat E, Lopez-Guillermo A, Grogan TM, Miller TP, LeBlanc M, Ott G, Kvaloy S, Delabie J, Holte H, Krajci P, Stokke T, Staudt LM. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med. 2002;346:1937–1947. doi: 10.1056/NEJMoa012914. [DOI] [PubMed] [Google Scholar]
- 47.Steidl C, Lee T, Shah SP, Farinha P, Han G, Nayar T, Delaney A, Jones SJ, Iqbal J, Weisenburger DD, Bast MA, Rosenwald A, Muller-Hermelink HK, Rimsza LM, Campo E, Delabie J, Braziel RM, Cook JR, Tubbs RR, Jaffe ES, Lenz G, Connors JM, Staudt LM, Chan WC, Gascoyne RD. Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med. 2010;362:875–885. doi: 10.1056/NEJMoa0905680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Schlapbach C, Ochsenbein A, Kaelin U, Hassan AS, Hunger RE, Yawalkar N. High numbers of DC-SIGN+ dendritic cells in lesional skin of cutaneous T-cell lymphoma. J Am Acad Dermatol. 2010;62:995–1004. doi: 10.1016/j.jaad.2009.06.082. [DOI] [PubMed] [Google Scholar]
- 49.Wilcox RA. Cancer-associated myeloproliferation: old association, new therapeutic target. Mayo Clin Proc. 2010;85:656–663. doi: 10.4065/mcp.2010.0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Shin J, Monti S, Aires DJ, Duvic M, Golub T, Jones DA, Kupper TS. Lesional gene expression profiling in cutaneous T-cell lymphoma reveals natural clusters associated with disease outcome. Blood. 2007;110:3015–3027. doi: 10.1182/blood-2006-12-061507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wilcox RA, Feldman AL, Wada DA, Yang ZZ, Comfere NI, Dong H, Kwon ED, Novak AJ, Markovic SN, Pittelkow MR, Witzig TE, Ansell SM. B7-H1 (PD-L1, CD274) suppresses host immunity in T-cell lymphoproliferative disorders. Blood. 2009;114:2149–2158. doi: 10.1182/blood-2009-04-216671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Epstein EH, Jr, Levin DL, Croft JD, Jr, Lutzner MA. Mycosis fungoides. Survival, prognostic features, response to therapy, and autopsy findings. Medicine (Baltimore) 1972;51:61–72. [PubMed] [Google Scholar]
- 53.Posner LE, Fossieck BE, Jr, Eddy JL, Bunn PA., Jr Septicemic complications of the cutaneous T-cell lymphomas. Am J Med. 1981;71:210–216. doi: 10.1016/0002-9343(81)90107-8. [DOI] [PubMed] [Google Scholar]
- 54.Axelrod PI, Lorber B, Vonderheid EC. Infections complicating mycosis fungoides and Sezary syndrome. JAMA. 1992;267:1354–1358. [PubMed] [Google Scholar]
- 55.Wysocka M, Benoit BM, Newton S, Azzoni L, Montaner LJ, Rook AH. Enhancement of the host immune responses in cutaneous T-cell lymphoma by CpG oligodeoxynucleotides and IL-15. Blood. 2004;104:4142–4149. doi: 10.1182/blood-2004-03-1190. [DOI] [PubMed] [Google Scholar]
- 56.Bouaziz JD, Ortonne N, Giustiniani J, Schiavon V, Huet D, Bagot M, Bensussan A. Circulating natural killer lymphocytes are potential cytotoxic effectors against autologous malignant cells in sezary syndrome patients. J Invest Dermatol. 2005;125:1273–1278. doi: 10.1111/j.0022-202X.2005.23914.x. [DOI] [PubMed] [Google Scholar]
- 57.Wysocka M, Zaki MH, French LE, Chehimi J, Shapiro M, Everetts SE, McGinnis KS, Montaner L, Rook AH. Sezary syndrome patients demonstrate a defect in dendritic cell populations: effects of CD40 ligand and treatment with GM-CSF on dendritic cell numbers and the production of cytokines. Blood. 2002;100:3287–3294. doi: 10.1182/blood-2002-01-0231. [DOI] [PubMed] [Google Scholar]
- 58.French LE, Huard B, Wysocka M, Shane R, Contassot E, Arrighi JF, Piguet V, Calderara S, Rook AH. Impaired CD40L signaling is a cause of defective IL-12 and TNF-alpha production in Sezary syndrome: circumvention by hexameric soluble CD40L. Blood. 2005;105:219–225. doi: 10.1182/blood-2004-03-1055. [DOI] [PubMed] [Google Scholar]
- 59.Samimi S, Benoit B, Evans K, Wherry EJ, Showe L, Wysocka M, Rook AH. Increased programmed death-1 expression on CD4+ T cells in cutaneous T-cell lymphoma: implications for immune suppression. Arch Dermatol. 2010;146:1382–1388. doi: 10.1001/archdermatol.2010.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lee BN, Duvic M, Tang CK, Bueso-Ramos C, Estrov Z, Reuben JM. Dysregulated synthesis of intracellular type 1 and type 2 cytokines by T cells of patients with cutaneous T-cell lymphoma. Clin Diagn Lab Immunol. 1999;6:79–84. doi: 10.1128/cdli.6.1.79-84.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yawalkar N, Ferenczi K, Jones DA, Yamanaka K, Suh KY, Sadat S, Kupper TS. Profound loss of T-cell receptor repertoire complexity in cutaneous T-cell lymphoma. Blood. 2003;102:4059–4066. doi: 10.1182/blood-2003-04-1044. [DOI] [PubMed] [Google Scholar]
- 62.Behl D, Ristow K, Markovic SN, Witzig TE, Habermann TM, Colgan JP, Inwards DJ, White WL, Ansell SM, Micallef IN, Johnston PB, Porrata LF. Absolute lymphocyte count predicts therapeutic efficacy of rituximab therapy in follicular lymphomas. Br J Haematol. 2007;137:409–415. doi: 10.1111/j.1365-2141.2007.06596.x. [DOI] [PubMed] [Google Scholar]
- 63.Porrata LF, Gertz MA, Inwards DJ, Litzow MR, Lacy MQ, Tefferi A, Gastineau DA, Dispenzieri A, Ansell SM, Micallef IN, Geyer SM, Markovic SN. Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood. 2001;98:579–585. doi: 10.1182/blood.v98.3.579. [DOI] [PubMed] [Google Scholar]
- 64.Porrata LF, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Gastineau DA, Litzow MR, Winters JL, Markovic SN. Early lymphocyte recovery predicts superior survival after autologous stem cell transplantation in non-Hodgkin lymphoma: a prospective study. Biol Blood Marrow Transplant. 2008;14:807–816. doi: 10.1016/j.bbmt.2008.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Porrata LF, Ristow K, Habermann TM, Witzig TE, Inwards DJ, Markovic SN. Absolute lymphocyte count at the time of first relapse predicts survival in patients with diffuse large B-cell lymphoma. Am J Hematol. 2009;84:93–97. doi: 10.1002/ajh.21337. [DOI] [PubMed] [Google Scholar]
- 66.Porrata LF, Rsitow K, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Habermann TM, Witzig TE, Colgan JP, Nowakowski GS, Thompson CA, Markovic SN. Lymphopenia assessed during routine follow-up after immunochemotherapy (R-CHOP) is a risk factor for predicting relapse in patients with diffuse large B-cell lymphoma. Leukemia. 2010;24:1343–1349. doi: 10.1038/leu.2010.108. [DOI] [PubMed] [Google Scholar]
- 67.Siddiqui M, Ristow K, Markovic SN, Witzig TE, Habermann TM, Colgan JP, Inwards DJ, White WL, Ansell SM, Micallef IN, Johnston PB, Call TG, Porrata LF. Absolute lymphocyte count predicts overall survival in follicular lymphomas. Br J Haematol. 2006;134:596–601. doi: 10.1111/j.1365-2141.2006.06232.x. [DOI] [PubMed] [Google Scholar]
- 68.Leich E, Haralambieva E, Zettl A, Chott A, Rudiger T, Holler S, Muller-Hermelink HK, Ott G, Rosenwald A. Tissue microarray-based screening for chromosomal breakpoints affecting the T-cell receptor gene loci in mature T-cell lymphomas. J Pathol. 2007;213:99–105. doi: 10.1002/path.2196. [DOI] [PubMed] [Google Scholar]
- 69.Feldman AL, Law M, Grogg KL, Thorland EC, Fink S, Kurtin PJ, Macon WR, Remstein ED, Dogan A. Incidence of TCR and TCL1 gene translocations and isochromosome 7q in peripheral T-cell lymphomas using fluorescence in situ hybridization. Am J Clin Pathol. 2008;130:178–185. doi: 10.1309/PNXUKA1CFJMVGCN1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Pham-Ledard A, Prochazkova-Carlotti M, Laharanne E, Vergier B, Jouary T, Beylot-Barry M, Merlio JP. IRF4 gene rearrangements define a subgroup of CD30-positive cutaneous T-cell lymphoma: a study of 54 cases. J Invest Dermatol. 2010;130:816–825. doi: 10.1038/jid.2009.314. [DOI] [PubMed] [Google Scholar]
- 71.Feldman AL, Law M, Remstein ED, Macon WR, Erickson LA, Grogg KL, Kurtin PJ, Dogan A. Recurrent translocations involving the IRF4 oncogene locus in peripheral T-cell lymphomas. Leukemia. 2009;23:574–580. doi: 10.1038/leu.2008.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wada DA, Law ME, Hsi ED, Dicaudo DJ, Ma L, Lim MS, Souza AD, Comfere NI, Weenig RH, Macon WR, Erickson LA, Ozsan N, Ansell SM, Dogan A, Feldman AL. Specificity of IRF4 translocations for primary cutaneous anaplastic large cell lymphoma: a multicenter study of 204 skin biopsies. Mod Pathol. 2010 doi: 10.1038/modpathol.2010.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Batista DA, Vonderheid EC, Hawkins A, Morsberger L, Long P, Murphy KM, Griffin CA. Multicolor fluorescence in situ hybridization (SKY) in mycosis fungoides and Sezary syndrome: search for recurrent chromosome abnormalities. Genes Chromosomes Cancer. 2006;45:383–391. doi: 10.1002/gcc.20302. [DOI] [PubMed] [Google Scholar]
- 74.Thangavelu M, Finn WG, Yelavarthi KK, Roenigk HH, Jr, Samuelson E, Peterson L, Kuzel TM, Rosen ST. Recurring structural chromosome abnormalities in peripheral blood lymphocytes of patients with mycosis fungoides/Sezary syndrome. Blood. 1997;89:3371–3377. [PubMed] [Google Scholar]
- 75.Staudt LM. Oncogenic activation of NF-kappaB. Cold Spring Harb Perspect Biol. 2010;2:a000109. doi: 10.1101/cshperspect.a000109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Izban KF, Ergin M, Qin JZ, Martinez RL, Pooley RJ, Saeed S, Alkan S. Constitutive expression of NF-kappa B is a characteristic feature of mycosis fungoides: implications for apoptosis resistance and pathogenesis. Hum Pathol. 2000;31:1482–1490. doi: 10.1053/hupa.2000.20370. [DOI] [PubMed] [Google Scholar]
- 77.Sors A, Jean-Louis F, Pellet C, Laroche L, Dubertret L, Courtois G, Bachelez H, Michel L. Down-regulating constitutive activation of the NF-kappaB canonical pathway overcomes the resistance of cutaneous T-cell lymphoma to apoptosis. Blood. 2006;107:2354–2363. doi: 10.1182/blood-2005-06-2536. [DOI] [PubMed] [Google Scholar]
- 78.Sors A, Jean-Louis F, Begue E, Parmentier L, Dubertret L, Dreano M, Courtois G, Bachelez H, Michel L. Inhibition of IkappaB kinase subunit 2 in cutaneous T-cell lymphoma down-regulates nuclear factor-kappaB constitutive activation, induces cell death, and potentiates the apoptotic response to antineoplastic chemotherapeutic agents. Clin Cancer Res. 2008;14:901–911. doi: 10.1158/1078-0432.CCR-07-1419. [DOI] [PubMed] [Google Scholar]
- 79.Juvekar A, Manna S, Ramaswami S, Chang TP, Vu HY, Ghosh CC, Celiker MY, Vancurova I. Bortezomib induces nuclear translocation of IkappaBalpha resulting in gene-specific suppression of NF-kappaB–dependent transcription and induction of apoptosis in CTCL. Mol Cancer Res. 2011;9:183–194. doi: 10.1158/1541-7786.MCR-10-0368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Nielsen M, Kaltoft K, Nordahl M, Ropke C, Geisler C, Mustelin T, Dobson P, Svejgaard A, Odum N. Constitutive activation of a slowly migrating isoform of Stat3 in mycosis fungoides: tyrphostin AG490 inhibits Stat3 activation and growth of mycosis fungoides tumor cell lines. Proc Natl Acad Sci U S A. 1997;94:6764–6769. doi: 10.1073/pnas.94.13.6764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sommer VH, Clemmensen OJ, Nielsen O, Wasik M, Lovato P, Brender C, Eriksen KW, Woetmann A, Kaestel CG, Nissen MH, Ropke C, Skov S, Odum N. In vivo activation of STAT3 in cutaneous T-cell lymphoma. Evidence for an antiapoptotic function of STAT3 Leukemia. 2004;18:1288–1295. doi: 10.1038/sj.leu.2403385. [DOI] [PubMed] [Google Scholar]
- 82.Zhang Q, Wang HY, Woetmann A, Raghunath PN, Odum N, Wasik MA. STAT3 induces transcription of the DNA methyltransferase 1 gene (DNMT1) in malignant T lymphocytes. Blood. 2006;108:1058–1064. doi: 10.1182/blood-2005-08-007377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Verma NK, Davies AM, Long A, Kelleher D, Volkov Y. STAT3 knockdown by siRNA induces apoptosis in human cutaneous T-cell lymphoma line Hut78 via downregulation of Bcl-xL. Cell Mol Biol Lett. 2010;15:342–355. doi: 10.2478/s11658-010-0008-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhang C, Li B, Zhang X, Hazarika P, Aggarwal BB, Duvic M. Curcumin selectively induces apoptosis in cutaneous T-cell lymphoma cell lines and patients’ PBMCs: potential role for STAT-3 and NF-kappaB signaling. J Invest Dermatol. 2010;130:2110–2119. doi: 10.1038/jid.2010.86. [DOI] [PubMed] [Google Scholar]
- 85.Nielsen M, Kaestel CG, Eriksen KW, Woetmann A, Stokkedal T, Kaltoft K, Geisler C, Ropke C, Odum N. Inhibition of constitutively activated Stat3 correlates with altered Bcl-2/Bax expression and induction of apoptosis in mycosis fungoides tumor cells. Leukemia. 1999;13:735–738. doi: 10.1038/sj.leu.2401415. [DOI] [PubMed] [Google Scholar]
- 86.Marzec M, Halasa K, Kasprzycka M, Wysocka M, Liu X, Tobias JW, Baldwin D, Zhang Q, Odum N, Rook AH, Wasik MA. Differential effects of interleukin-2 and interleukin-15 versus interleukin-21 on CD4+ cutaneous T-cell lymphoma cells. Cancer Res. 2008;68:1083–1091. doi: 10.1158/0008-5472.CAN-07-2403. [DOI] [PubMed] [Google Scholar]
- 87.Mao X, Lillington DM, Czepulkowski B, Russell-Jones R, Young BD, Whittaker S. Molecular cytogenetic characterization of Sezary syndrome. Genes Chromosomes Cancer. 2003;36:250–260. doi: 10.1002/gcc.10152. [DOI] [PubMed] [Google Scholar]
- 88.Barba G, Matteucci C, Girolomoni G, Brandimarte L, Varasano E, Martelli MF, Mecucci C. Comparative genomic hybridization identifies 17q11.2 approximately q12 duplication as an early event in cutaneous T-cell lymphomas. Cancer Genet Cytogenet. 2008;184:48–51. doi: 10.1016/j.cancergencyto.2008.03.007. [DOI] [PubMed] [Google Scholar]
- 89.Wu J, Nihal M, Siddiqui J, Vonderheid EC, Wood GS. Low FAS/CD95 expression by CTCL correlates with reduced sensitivity to apoptosis that can be restored by FAS upregulation. J Invest Dermatol. 2009;129:1165–1173. doi: 10.1038/jid.2008.309. [DOI] [PubMed] [Google Scholar]
- 90.Wu J, Wood GS. Reduction of Fas/CD95 Promoter Methylation, Upregulation of Fas Protein, and Enhancement of Sensitivity to Apoptosis in Cutaneous T-Cell Lymphoma. Arch Dermatol. 2011 doi: 10.1001/archdermatol.2010.376. [DOI] [PubMed] [Google Scholar]
- 91.Jones CL, Wain EM, Chu CC, Tosi I, Foster R, McKenzie RC, Whittaker SJ, Mitchell TJ. Downregulation of Fas gene expression in Sezary syndrome is associated with promoter hypermethylation. J Invest Dermatol. 2010;130:1116–1125. doi: 10.1038/jid.2009.301. [DOI] [PubMed] [Google Scholar]
- 92.Dereure O, Levi E, Vonderheid EC, Kadin ME. Infrequent Fas mutations but no Bax or p53 mutations in early mycosis fungoides: a possible mechanism for the accumulation of malignant T lymphocytes in the skin. J Invest Dermatol. 2002;118:949–956. doi: 10.1046/j.1523-1747.2002.01794.x. [DOI] [PubMed] [Google Scholar]
- 93.Scarisbrick JJ, Woolford AJ, Russell-Jones R, Whittaker SJ. Loss of heterozygosity on 10q and microsatellite instability in advanced stages of primary cutaneous T-cell lymphoma and possible association with homozygous deletion of PTEN. Blood. 2000;95:2937–2942. [PubMed] [Google Scholar]
- 94.van Doorn R, Zoutman WH, Dijkman R, de Menezes RX, Commandeur S, Mulder AA, van der Velden PA, Vermeer MH, Willemze R, Yan PS, Huang TH, Tensen CP. Epigenetic profiling of cutaneous T-cell lymphoma: promoter hypermethylation of multiple tumor suppressor genes including BCL7a, PTPRG, and p73. J Clin Oncol. 2005;23:3886–3896. doi: 10.1200/JCO.2005.11.353. [DOI] [PubMed] [Google Scholar]
- 95.Scarisbrick JJ, Woolford AJ, Calonje E, Photiou A, Ferreira S, Orchard G, Russell-Jones R, Whittaker SJ. Frequent abnormalities of the p15 and p16 genes in mycosis fungoides and sezary syndrome. J Invest Dermatol. 2002;118:493–499. doi: 10.1046/j.0022-202x.2001.01682.x. [DOI] [PubMed] [Google Scholar]
- 96.Laharanne E, Chevret E, Idrissi Y, Gentil C, Longy M, Ferrer J, Dubus P, Jouary T, Vergier B, Beylot-Barry M, Merlio JP. CDKN2A-CDKN2B deletion defines an aggressive subset of cutaneous T-cell lymphoma. Mod Pathol. 2010;23:547–558. doi: 10.1038/modpathol.2009.196. [DOI] [PubMed] [Google Scholar]
- 97.Mao X, Orchard G, Vonderheid EC, Nowell PC, Bagot M, Bensussan A, Russell-Jones R, Young BD, Whittaker SJ. Heterogeneous abnormalities of CCND1 and RB1 in primary cutaneous T-Cell lymphomas suggesting impaired cell cycle control in disease pathogenesis. J Invest Dermatol. 2006;126:1388–1395. doi: 10.1038/sj.jid.5700224. [DOI] [PubMed] [Google Scholar]
- 98.Kennah E, Ringrose A, Zhou LL, Esmailzadeh S, Qian H, Su MW, Zhou Y, Jiang X. Identification of tyrosine kinase, HCK, and tumor suppressor, BIN1, as potential mediators of AHI-1 oncogene in primary and transformed CTCL cells. Blood. 2009;113:4646–4655. doi: 10.1182/blood-2008-08-174037. [DOI] [PubMed] [Google Scholar]
- 99.Qin JZ, Dummer R, Burg G, Dobbeling U. Constitutive and interleukin-7/interleukin-15 stimulated DNA binding of Myc, Jun, and novel Myc-like proteins in cutaneous T-cell lymphoma cells. Blood. 1999;93:260–267. [PubMed] [Google Scholar]
- 100.Kiessling MK, Oberholzer PA, Mondal C, Karpova MB, Zipser MC, Lin WM, Girardi M, Macconaill LE, Kehoe SM, Hatton C, French LE, Garraway LA, Polier G, Suss D, Klemke CD, Krammer PH, Gulow K, Dummer R. High-throughput mutation profiling of CTCL samples reveals KRAS and NRAS mutations sensitizing tumors toward inhibition of the RAS/RAF/MEK signaling cascade. Blood. 2011;117:2433–2440. doi: 10.1182/blood-2010-09-305128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Krejsgaard T, Vetter-Kauczok CS, Woetmann A, Kneitz H, Eriksen KW, Lovato P, Zhang Q, Wasik MA, Geisler C, Ralfkiaer E, Becker JC, Odum N. Ectopic expression of B-lymphoid kinase in cutaneous T-cell lymphoma. Blood. 2009;113:5896–5904. doi: 10.1182/blood-2008-09-181024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Kim YH, Liu HL, Mraz-Gernhard S, Varghese A, Hoppe RT. Long-term outcome of 525 patients with mycosis fungoides and Sezary syndrome: clinical prognostic factors and risk for disease progression. Arch Dermatol. 2003;139:857–866. doi: 10.1001/archderm.139.7.857. [DOI] [PubMed] [Google Scholar]
- 103.van Doorn R, Van Haselen CW, van Voorst Vader PC, Geerts ML, Heule F, de Rie M, Steijlen PM, Dekker SK, van Vloten WA, Willemze R. Mycosis fungoides: disease evolution and prognosis of 309 Dutch patients. Arch Dermatol. 2000;136:504–510. doi: 10.1001/archderm.136.4.504. [DOI] [PubMed] [Google Scholar]
- 104.Arulogun SO, Prince HM, Ng J, Lade S, Ryan GF, Blewitt O, McCormack C. Long-term outcomes of patients with advanced-stage cutaneous T-cell lymphoma and large cell transformation. Blood. 2008;112:3082–3087. doi: 10.1182/blood-2008-05-154609. [DOI] [PubMed] [Google Scholar]
- 105.Morgan SM, Hodges E, Mitchell TJ, Harris S, Whittaker SJ, Smith JL. Molecular analysis of T-cell receptor beta genes in cutaneous T-cell lymphoma reveals Jbeta1 bias. J Invest Dermatol. 2006;126:1893–1899. doi: 10.1038/sj.jid.5700304. [DOI] [PubMed] [Google Scholar]
- 106.Ponti R, Quaglino P, Novelli M, Fierro MT, Comessatti A, Peroni A, Bonello L, Bernengo MG. T-cell receptor gamma gene rearrangement by multiplex polymerase chain reaction/heteroduplex analysis in patients with cutaneous T-cell lymphoma (mycosis fungoides/Sezary syndrome) and benign inflammatory disease: correlation with clinical, histological and immunophenotypical findings. Br J Dermatol. 2005;153:565–573. doi: 10.1111/j.1365-2133.2005.06649.x. [DOI] [PubMed] [Google Scholar]
- 107.Guitart J, Magro C. Cutaneous T-cell lymphoid dyscrasia: a unifying term for idiopathic chronic dermatoses with persistent T-cell clones. Arch Dermatol. 2007;143:921–932. doi: 10.1001/archderm.143.7.921. [DOI] [PubMed] [Google Scholar]
- 108.Posnett DN, Sinha R, Kabak S, Russo C. Clonal populations of T cells in normal elderly humans: the T cell equivalent to “benign monoclonal gammapathy”. J Exp Med. 1994;179:609–618. doi: 10.1084/jem.179.2.609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Epling-Burnette PK, Painter JS, Rollison DE, Ku E, Vendron D, Widen R, Boulware D, Zou JX, Bai F, List AF. Prevalence and clinical association of clonal T-cell expansions in Myelodysplastic Syndrome. Leukemia. 2007;21:659–667. doi: 10.1038/sj.leu.2404590. [DOI] [PubMed] [Google Scholar]
- 110.Martinez A, Pittaluga S, Villamor N, Colomer D, Rozman M, Raffeld M, Montserrat E, Campo E, Jaffe ES. Clonal T-cell populations and increased risk for cytotoxic T-cell lymphomas in B-CLL patients: clinicopathologic observations and molecular analysis. Am J Surg Pathol. 2004;28:849–858. doi: 10.1097/00000478-200407000-00002. [DOI] [PubMed] [Google Scholar]
- 111.Kohler S, Jones CD, Warnke RA, Zehnder JL. PCR-heteroduplex analysis of T-cell receptor gamma gene rearrangement in paraffin-embedded skin biopsies. Am J Dermatopathol. 2000;22:321–327. doi: 10.1097/00000372-200008000-00005. [DOI] [PubMed] [Google Scholar]
- 112.Thurber SE, Zhang B, Kim YH, Schrijver I, Zehnder J, Kohler S. T-cell clonality analysis in biopsy specimens from two different skin sites shows high specificity in the diagnosis of patients with suggested mycosis fungoides. J Am Acad Dermatol. 2007;57:782–790. doi: 10.1016/j.jaad.2007.06.004. [DOI] [PubMed] [Google Scholar]
- 113.Gniadecki R, Lukowsky A. Monoclonal T-cell dyscrasia of undetermined significance associated with recalcitrant erythroderma. Arch Dermatol. 2005;141:361–367. doi: 10.1001/archderm.141.3.361. [DOI] [PubMed] [Google Scholar]
- 114.Ormsby A, Bergfeld WF, Tubbs RR, Hsi ED. Evaluation of a new paraffin-reactive CD7 T-cell deletion marker and a polymerase chain reaction-based T-cell receptor gene rearrangement assay: implications for diagnosis of mycosis fungoides in community clinical practice. J Am Acad Dermatol. 2001;45:405–413. doi: 10.1067/mjd.2001.116342. [DOI] [PubMed] [Google Scholar]
- 115.Michie SA, Abel EA, Hoppe RT, Warnke RA, Wood GS. Discordant expression of antigens between intraepidermal and intradermal T cells in mycosis fungoides. Am J Pathol. 1990;137:1447–1451. [PMC free article] [PubMed] [Google Scholar]
- 116.Kazakov DV, Burg G, Kempf W. Clinicopathological spectrum of mycosis fungoides. J Eur Acad Dermatol Venereol. 2004;18:397–415. doi: 10.1111/j.1468-3083.2004.00937.x. [DOI] [PubMed] [Google Scholar]
- 117.Pimpinelli N, Olsen EA, Santucci M, Vonderheid E, Haeffner AC, Stevens S, Burg G, Cerroni L, Dreno B, Glusac E, Guitart J, Heald PW, Kempf W, Knobler R, Lessin S, Sander C, Smoller BS, Telang G, Whittaker S, Iwatsuki K, Obitz E, Takigawa M, Turner ML, Wood GS. Defining early mycosis fungoides. J Am Acad Dermatol. 2005;53:1053–1063. doi: 10.1016/j.jaad.2005.08.057. [DOI] [PubMed] [Google Scholar]
- 118.Sezary A, Bouvrain Y. Erythrodermie avec presence de cellules monstrueses dans le derme et le sang circulant. Bull Soc Fr Derm Syph. 1938;45:254–260. [Google Scholar]
- 119.Main RA, Goodall HB, Swanson WC. Sezary’s syndrome. Br J Dermatol. 1959;71:335–343. doi: 10.1111/j.1365-2133.1959.tb13359.x. [DOI] [PubMed] [Google Scholar]
- 120.Taswell HF, Winkelmann RK. Sezary syndrome–a malignant reticulemic erythroderma. Jama. 1961;177:465–472. doi: 10.1001/jama.1961.03040330001001. [DOI] [PubMed] [Google Scholar]
- 121.Lutzner MA, Emerit I, Durepaire R, Flandrin G, Grupper C, Prunieras M. Cytogenetic, cytophotometric, and ultrastructural study of large cerebriform cells of the Sezary syndrome and description of a small-cell variant. J Natl Cancer Inst. 1973;50:1145–1162. doi: 10.1093/jnci/50.5.1145. [DOI] [PubMed] [Google Scholar]
- 122.Lutzner MA, Jordan HW. The ultrastructure of an abnormal cell in Sezary’s syndrome. Blood. 1968;31:719–726. [PubMed] [Google Scholar]
- 123.Edelson RL, Lutzner MA, Kirkpatrick CH, Shevach EM, Green I. Morphologic and functional properties of the atypical T lymphocytes of the Sezary syndrome. Mayo Clin Proc. 1974;49:558–566. [PubMed] [Google Scholar]
- 124.Lutzner MA, Hobbs JW, Horvath P. Ultrastructure of abnormal cells in Sezary syndrome, mycosis fungoides, and parapsoriasis en plaque. Arch Dermatol. 1971;103:375–386. doi: 10.1001/archderm.103.4.375. [DOI] [PubMed] [Google Scholar]
- 125.Matutes E, Robinson D, O’Brien M, Haynes BF, Zola H, Catovsky D. Candidate counterparts of Sezary cells and adult T-cell lymphoma-leukaemia cells in normal peripheral blood: an ultrastructural study with the immunogold method and monoclonal antibodies. Leuk Res. 1983;7:787–801. doi: 10.1016/0145-2126(83)90073-5. [DOI] [PubMed] [Google Scholar]
- 126.Reinhold U, Herpertz M, Kukel S, Oltermann I, Uerlich M, Kreysel HW. Induction of nuclear contour irregularity during T-cell activation via the T-cell receptor/CD3 complex and CD2 antigens in the presence of phorbol esters. Blood. 1994;83:703–706. [PubMed] [Google Scholar]
- 127.Scheffer E, Meijer CJ, van Vloten WA, Willemze R. A histologic study of lymph nodes from patients with the Sezary syndrome. Cancer. 1986;57:2375–2380. doi: 10.1002/1097-0142(19860615)57:12<2375::aid-cncr2820571223>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- 128.Willemze R, van Vloten WA, Hermans J, Damsteeg MJ, Meijer CJ. Diagnostic criteria in Sezary’s syndrome: a multiparameter study of peripheral blood lymphocytes in 32 patients with erythroderma. J Invest Dermatol. 1983;81:392–397. doi: 10.1111/1523-1747.ep12521991. [DOI] [PubMed] [Google Scholar]
- 129.Boumsell L, Bernard A, Reinherz EL, Nadler LM, Ritz J, Coppin H, Richard Y, Dubertret L, Valensi F, Degos L, Lemerle J, Flandrin G, Dausset J, Schlossman SF. Surface antigens on malignant Sezary and T-CLL cells correspond to those of mature T cells. Blood. 1981;57:526–530. [PubMed] [Google Scholar]
- 130.Vonderheid EC, Bernengo MG, Burg G, Duvic M, Heald P, Laroche L, Olsen E, Pittelkow M, Russell-Jones R, Takigawa M, Willemze R. Update on erythrodermic cutaneous T-cell lymphoma: report of the International Society for Cutaneous Lymphomas. J Am Acad Dermatol. 2002;46:95–106. doi: 10.1067/mjd.2002.118538. [DOI] [PubMed] [Google Scholar]
- 131.Hristov AC, Vonderheid EC, Borowitz MJ. Simplified flow cytometric assessment in mycosis fungoides and Sezary syndrome. Am J Clin Pathol. 2011;136:944–953. doi: 10.1309/AJCP09OTJOYAVZZK. [DOI] [PubMed] [Google Scholar]
- 132.Bernengo MG, Quaglino P, Novelli M, Cappello N, Doveil GC, Lisa F, De Matteis A, Fierro MT, Appino A. Prognostic factors in Sezary syndrome: a multivariate analysis of clinical, haematological and immunological features. Ann Oncol. 1998;9:857–863. doi: 10.1023/a:1008397323199. [DOI] [PubMed] [Google Scholar]
- 133.Harmon CB, Witzig TE, Katzmann JA, Pittelkow MR. Detection of circulating T cells with CD4+CD7− immunophenotype in patients with benign and malignant lymphoproliferative dermatoses. J Am Acad Dermatol. 1996;35:404–410. doi: 10.1016/s0190-9622(96)90605-2. [DOI] [PubMed] [Google Scholar]
- 134.Klemke CD, Booken N, Weiss C, Nicolay JP, Goerdt S, Felcht M, Geraud C, Kempf W, Assaf C, Ortonne N, Battistella M, Bagot M, Knobler R, Quaglino P, Arheiliger B, Santucci M, Jansen P, Vermeer MH, Willemze R. 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. doi: 10.1111/bjd.13832. [DOI] [PubMed] [Google Scholar]
- 135.Bogen SA, Pelley D, Charif M, McCusker M, Koh H, Foss F, Garifallou M, Arkin C, Zucker-Franklin D. Immunophenotypic identification of Sezary cells in peripheral blood. Am J Clin Pathol. 1996;106:739–748. doi: 10.1093/ajcp/106.6.739. [DOI] [PubMed] [Google Scholar]
- 136.Ginaldi L, Matutes E, Farahat N, De Martinis M, Morilla R, Catovsky D. Differential expression of CD3 and CD7 in T-cell malignancies: a quantitative study by flow cytometry. Br J Haematol. 1996;93:921–927. doi: 10.1046/j.1365-2141.1996.d01-1720.x. [DOI] [PubMed] [Google Scholar]
- 137.Jones D, Dang NH, Duvic M, Washington LT, Huh YO. Absence of CD26 expression is a useful marker for diagnosis of T-cell lymphoma in peripheral blood. Am J Clin Pathol. 2001;115:885–892. doi: 10.1309/U1Y6-J4AG-5M4M-7AYV. [DOI] [PubMed] [Google Scholar]
- 138.Pierson DM, Jones D, Muzzafar T, Kersh MJ, Challagundla P, Medeiros LJ, Jorgensen JL. Utility of CD26 in flow cytometric immunophenotyping of T-cell lymphomas in tissue and body fluid specimens. Cytometry B Clin Cytom. 2008;74:341–348. doi: 10.1002/cyto.b.20431. [DOI] [PubMed] [Google Scholar]
- 139.Sokolowska-Wojdylo M, Wenzel J, Gaffal E, Steitz J, Roszkiewicz J, Bieber T, Tuting T. Absence of CD26 expression on skin-homing CLA+ CD4+ T lymphocytes in peripheral blood is a highly sensitive marker for early diagnosis and therapeutic monitoring of patients with Sezary syndrome. Clin Exp Dermatol. 2005;30:702–706. doi: 10.1111/j.1365-2230.2005.01904.x. [DOI] [PubMed] [Google Scholar]
- 140.Bahler DW, Hartung L, Hill S, Bowen GM, Vonderheid EC. CD158k/KIR3DL2 is a useful marker for identifying neoplastic T-cells in Sezary syndrome by flow cytometry. Cytometry B Clin Cytom. 2008;74:156–162. doi: 10.1002/cyto.b.20395. [DOI] [PubMed] [Google Scholar]
- 141.Poszepczynska-Guigne E, Schiavon V, D’Incan M, Echchakir H, Musette P, Ortonne N, Boumsell L, Moretta A, Bensussan A, Bagot M. CD158k/KIR3DL2 is a new phenotypic marker of Sezary cells: relevance for the diagnosis and follow-up of Sezary syndrome. J Invest Dermatol. 2004;122:820–823. doi: 10.1111/j.0022-202X.2004.22326.x. [DOI] [PubMed] [Google Scholar]
- 142.Klemke CD, Brade J, Weckesser S, Sachse MM, Booken N, Neumaier M, Goerdt S, Nebe TC. The diagnosis of Sezary syndrome on peripheral blood by flow cytometry requires the use of multiple markers. Br J Dermatol. 2008;159:871–880. doi: 10.1111/j.1365-2133.2008.08739.x. [DOI] [PubMed] [Google Scholar]
- 143.Morice WG, Kimlinger T, Katzmann JA, Lust JA, Heimgartner PJ, Halling KC, Hanson CA. Flow cytometric assessment of TCR-Vbeta expression in the evaluation of peripheral blood involvement by T-cell lymphoproliferative disorders: a comparison with conventional T-cell immunophenotyping and molecular genetic techniques. Am J Clin Pathol. 2004;121:373–383. doi: 10.1309/3A32-DTVM-H640-M2QA. [DOI] [PubMed] [Google Scholar]
- 144.Schwab C, Willers J, Niederer E, Ludwig E, Kundig T, Grob P, Burg G, Dummer R. The use of anti-T-cell receptor-Vbeta antibodies for the estimation of treatment success and phenotypic characterization of clonal T-cell populations in cutaneous T-cell lymphomas. Br J Haematol. 2002;118:1019–1026. doi: 10.1046/j.1365-2141.2002.03726.x. [DOI] [PubMed] [Google Scholar]
- 145.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: 10.1182/blood-2010-05-287664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Wilcox RA. Cutaneous T-cell lymphoma: 2011 update on diagnosis, risk-stratification, and management. Am J Hematol. 2011;86:928–948. doi: 10.1002/ajh.22139. [DOI] [PubMed] [Google Scholar]
- 147.Olsen E, Vonderheid E, Pimpinelli N, Willemze R, Kim Y, Knobler R, Zackheim H, Duvic M, Estrach T, Lamberg S, Wood G, Dummer R, Ranki A, Burg G, Heald P, Pittelkow M, Bernengo MG, Sterry W, Laroche L, Trautinger F, Whittaker S. 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: 10.1182/blood-2007-03-055749. [DOI] [PubMed] [Google Scholar]
- 148.Scheffer E, Meijer CJ, Van Vloten WA. Dermatopathic lymphadenopathy and lymph node involvement in mycosis fungoides. Cancer. 1980;45:137–148. doi: 10.1002/1097-0142(19800101)45:1<137::aid-cncr2820450124>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
- 149.Sausville EA, Worsham GF, Matthews MJ, Makuch RW, Fischmann AB, Schechter GP, Gazdar AF, Bunn PA., Jr Histologic assessment of lymph nodes in mycosis fungoides/Sezary syndrome (cutaneous T-cell lymphoma): clinical correlations and prognostic import of a new classification system. Hum Pathol. 1985;16:1098–1109. doi: 10.1016/s0046-8177(85)80177-5. [DOI] [PubMed] [Google Scholar]
- 150.Clendenning WE, Rappaport HW. Report of the Committee on Pathology of Cutaneous T Cell Lymphomas. Cancer Treat Rep. 1979;63:719–724. [PubMed] [Google Scholar]
- 151.Fraser-Andrews EA, Mitchell T, Ferreira S, Seed PT, Russell-Jones R, Calonje E, Whittaker SJ. Molecular staging of lymph nodes from 60 patients with mycosis fungoides and Sezary syndrome: correlation with histopathology and outcome suggests prognostic relevance in mycosis fungoides. Br J Dermatol. 2006;155:756–762. doi: 10.1111/j.1365-2133.2006.07428.x. [DOI] [PubMed] [Google Scholar]
- 152.Assaf C, Hummel M, Steinhoff M, Geilen CC, Orawa H, Stein H, Orfanos CE. Early TCR-beta and TCR-gamma PCR detection of T-cell clonality indicates minimal tumor disease in lymph nodes of cutaneous T-cell lymphoma: diagnostic and prognostic implications. Blood. 2005;105:503–510. doi: 10.1182/blood-2004-06-2220. [DOI] [PubMed] [Google Scholar]
- 153.Scarisbrick JJ, Whittaker S, Evans AV, Fraser-Andrews EA, Child FJ, Dean A, Russell-Jones R. Prognostic significance of tumor burden in the blood of patients with erythrodermic primary cutaneous T-cell lymphoma. Blood. 2001;97:624–630. doi: 10.1182/blood.v97.3.624. [DOI] [PubMed] [Google Scholar]
- 154.Fraser-Andrews EA, Woolford AJ, Russell-Jones R, Seed PT, Whittaker SJ. Detection of a peripheral blood T cell clone is an independent prognostic marker in mycosis fungoides. The Journal of investigative dermatology. 2000;114:117–121. doi: 10.1046/j.1523-1747.2000.00830.x. [DOI] [PubMed] [Google Scholar]
- 155.Vergier B, de Muret A, Beylot-Barry M, Vaillant L, Ekouevi D, Chene G, Carlotti A, Franck N, Dechelotte P, Souteyrand P, Courville P, Joly P, Delaunay M, Bagot M, Grange F, Fraitag S, Bosq J, Petrella T, Durlach A, De Mascarel A, Merlio JP, Wechsler J. Transformation of mycosis fungoides: clinicopathological and prognostic features of 45 cases. French Study Group of Cutaneious Lymphomas. Blood. 2000;95:2212–2218. [PubMed] [Google Scholar]
- 156.Greer JP, Salhany KE, Cousar JB, Fields JP, King LE, Graber SE, Flexner JM, Stein RS, Collins RD. Clinical features associated with transformation of cerebriform T-cell lymphoma to a large cell process. Hematol Oncol. 1990;8:215–227. doi: 10.1002/hon.2900080406. [DOI] [PubMed] [Google Scholar]
- 157.Salhany KE, Cousar JB, Greer JP, Casey TT, Fields JP, Collins RD. Transformation of cutaneous T cell lymphoma to large cell lymphoma. A clinicopathologic and immunologic study The American journal of pathology. 1988;132:265–277. [PMC free article] [PubMed] [Google Scholar]
- 158.Diamandidou E, Colome M, Fayad L, Duvic M, Kurzrock R. Prognostic factor analysis in mycosis fungoides/Sezary syndrome. J Am Acad Dermatol. 1999;40:914–924. doi: 10.1016/s0190-9622(99)70079-4. [DOI] [PubMed] [Google Scholar]
- 159.Diamandidou E, Colome-Grimmer M, Fayad L, Duvic M, Kurzrock R. Transformation of mycosis fungoides/Sezary syndrome: clinical characteristics and prognosis. Blood. 1998;92:1150–1159. [PubMed] [Google Scholar]
- 160.Kim YH, Willemze R, Pimpinelli N, Whittaker S, Olsen EA, Ranki A, Dummer R, Hoppe RT, EORTC Iat TNM classification system for primary cutaneous lymphomas other than 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;110:479–484. doi: 10.1182/blood-2006-10-054601. [DOI] [PubMed] [Google Scholar]
- 161.Litvinov IV, Jones DA, Sasseville D, Kupper TS. Transcriptional profiles predict disease outcome in patients with cutaneous T-cell lymphoma. Clin Cancer Res. 2010;16:2106–2114. doi: 10.1158/1078-0432.CCR-09-2879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Caprini E, Cristofoletti C, Arcelli D, Fadda P, Citterich MH, Sampogna F, Magrelli A, Censi F, Torreri P, Frontani M, Scala E, Picchio MC, Temperani P, Monopoli A, Lombardo GA, Taruscio D, Narducci MG, Russo G. 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. doi: 10.1158/0008-5472.CAN-09-2367. [DOI] [PubMed] [Google Scholar]
- 163.Salgado R, Servitje O, Gallardo F, Vermeer MH, Ortiz-Romero PL, Karpova MB, Zipser MC, Muniesa C, Garcia-Muret MP, Estrach T, Salido M, Sanchez-Schmidt J, Herrera M, Romagosa V, Suela J, Ferreira BI, Cigudosa JC, Barranco C, Serrano S, Dummer R, Tensen CP, Sole F, Pujol RM, Espinet B. Oligonucleotide array-CGH identifies genomic subgroups and prognostic markers for tumor stage mycosis fungoides. J Invest Dermatol. 2010;130:1126–1135. doi: 10.1038/jid.2009.306. [DOI] [PubMed] [Google Scholar]
- 164.Kaye FJ, Bunn PA, Jr, Steinberg SM, Stocker JL, Ihde DC, Fischmann AB, Glatstein EJ, Schechter GP, Phelps RM, Foss FM, et al. A randomized trial comparing combination electron-beam radiation and chemotherapy with topical therapy in the initial treatment of mycosis fungoides. N Engl J Med. 1989;321:1784–1790. doi: 10.1056/NEJM198912283212603. [DOI] [PubMed] [Google Scholar]
- 165.Hughes CF, Khot A, McCormack C, Lade S, Westerman DA, Twigger R, Buelens O, Newland K, Tam C, Dickinson M, Ryan G, Ritchie D, Wood C, Prince HM. Lack of durable disease control with chemotherapy for mycosis fungoides and Sezary syndrome: a comparative study of systemic therapy. Blood. 2015;125:71–81. doi: 10.1182/blood-2014-07-588236. [DOI] [PubMed] [Google Scholar]
- 166.Trautinger F, Knobler R, Willemze R, Peris K, Stadler R, Laroche L, D’Incan M, Ranki A, Pimpinelli N, Ortiz-Romero P, Dummer R, Estrach T, Whittaker S. EORTC consensus recommendations for the treatment of mycosis fungoides/Sezary syndrome. European Journal of Cancer. 2006;42:1014–1030. doi: 10.1016/j.ejca.2006.01.025. [DOI] [PubMed] [Google Scholar]
- 167.Lansigan F, Foss FM. Current and emerging treatment strategies for cutaneous T-cell lymphoma. Drugs. 2010;70:273–286. doi: 10.2165/11532190-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 168.Horwitz SM, Olsen EA, Duvic M, Porcu P, Kim YH. Review of the treatment of mycosis fungoides and sezary syndrome: a stage-based approach. J Natl Compr Canc Netw. 2008;6:436–442. doi: 10.6004/jnccn.2008.0033. [DOI] [PubMed] [Google Scholar]
- 169.Prince HM, Whittaker S, Hoppe RT. How I treat mycosis fungoides and Sezary syndrome. Blood. 2009;114:4337–4353. doi: 10.1182/blood-2009-07-202895. [DOI] [PubMed] [Google Scholar]
- 170.Whittaker SJ, Marsden JR, Spittle M, Russell Jones R. Joint British Association of Dermatologists and U.K. Cutaneous Lymphoma Group guidelines for the management of primary cutaneous T-cell lymphomas. Br J Dermatol. 2003;149:1095–1107. doi: 10.1111/j.1365-2133.2003.05698.x. [DOI] [PubMed] [Google Scholar]
- 171.Jones GW, Kacinski BM, Wilson LD, Willemze R, Spittle M, Hohenberg G, Handl-Zeller L, Trautinger F, Knobler R. Total skin electron radiation in the management of mycosis fungoides: Consensus of the European Organization for Research and Treatment of Cancer (EORTC) Cutaneous Lymphoma Project Group. J Am Acad Dermatol. 2002;47:364–370. doi: 10.1067/mjd.2002.123482. [DOI] [PubMed] [Google Scholar]
- 172.Wilcox RA. Cutaneous T-cell lymphoma: 2014 update on diagnosis, risk-stratification, and management. Am J Hematol. 2014;89:837–851. doi: 10.1002/ajh.23756. [DOI] [PubMed] [Google Scholar]
- 173.Kempf W, Kettelhack N, Duvic M, Burg G. Topical and systemic retinoid therapy for cutaneous T-cell lymphoma. Hematol Oncol Clin North Am. 2003;17:1405–1419. doi: 10.1016/s0889-8588(03)00107-2. [DOI] [PubMed] [Google Scholar]
- 174.Zhang C, Duvic M. Retinoids: therapeutic applications and mechanisms of action in cutaneous T-cell lymphoma. Dermatol Ther. 2003;16:322–330. doi: 10.1111/j.1396-0296.2003.01644.x. [DOI] [PubMed] [Google Scholar]
- 175.Nieto-Rementeria N, Perez-Yarza G, Boyano MD, Apraiz A, Izu R, Diaz-Perez JL, Asumendi A. Bexarotene activates the p53/p73 pathway in human cutaneous T-cell lymphoma. Br J Dermatol. 2009;160:519–526. doi: 10.1111/j.1365-2133.2008.08931.x. [DOI] [PubMed] [Google Scholar]
- 176.Zhang C, Hazarika P, Ni X, Weidner DA, Duvic M. Induction of apoptosis by bexarotene in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. Clin Cancer Res. 2002;8:1234–1240. [PubMed] [Google Scholar]
- 177.Duvic M, Hymes K, Heald P, Breneman D, Martin AG, Myskowski P, Crowley C, Yocum RC. Bexarotene is effective and safe for treatment of refractory advanced-stage cutaneous T-cell lymphoma: multinational phase II–III trial results. J Clin Oncol. 2001;19:2456–2471. doi: 10.1200/JCO.2001.19.9.2456. [DOI] [PubMed] [Google Scholar]
- 178.Abbott RA, Whittaker SJ, Morris SL, Russell-Jones R, Hung T, Bashir SJ, Scarisbrick JJ. Bexarotene therapy for mycosis fungoides and Sezary syndrome. Br J Dermatol. 2009;160:1299–1307. doi: 10.1111/j.1365-2133.2009.09037.x. [DOI] [PubMed] [Google Scholar]
- 179.Assaf C, Bagot M, Dummer R, Duvic M, Gniadecki R, Knobler R, Ranki A, Schwandt P, Whittaker S. Minimizing adverse side-effects of oral bexarotene in cutaneous T-cell lymphoma: an expert opinion. Br J Dermatol. 2006;155:261–266. doi: 10.1111/j.1365-2133.2006.07329.x. [DOI] [PubMed] [Google Scholar]
- 180.Gniadecki R, Assaf C, Bagot M, Dummer R, Duvic M, Knobler R, Ranki A, Schwandt P, Whittaker S. The optimal use of bexarotene in cutaneous T-cell lymphoma. Br J Dermatol. 2007;157:433–440. doi: 10.1111/j.1365-2133.2007.07975.x. [DOI] [PubMed] [Google Scholar]
- 181.Scarisbrick JJ, Morris S, Azurdia R, Illidge T, Parry E, Graham-Brown R, Cowan R, Gallop-Evans E, Wachsmuth R, Eagle M, Wierzbicki AS, Soran H, Whittaker S, Wain EM. U.K. consensus statement on safe clinical prescribing of bexarotene for patients with cutaneous T-cell lymphoma. Br J Dermatol. 2013;168:192–200. doi: 10.1111/bjd.12042. [DOI] [PubMed] [Google Scholar]
- 182.Huber MA, Kunzi-Rapp K, Staib G, Scharffetter-Kochanek K. Management of refractory early-stage cutaneous T-cell lymphoma (mycosis fungoides) with a combination of oral bexarotene and psoralen plus ultraviolet bath therapy. J Am Acad Dermatol. 2004;50:475–476. doi: 10.1016/s0190-9622(03)02488-5. [DOI] [PubMed] [Google Scholar]
- 183.Schrump DS. Cytotoxicity mediated by histone deacetylase inhibitors in cancer cells: mechanisms and potential clinical implications. Clin Cancer Res. 2009;15:3947–3957. doi: 10.1158/1078-0432.CCR-08-2787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Lemoine M, Younes A. Histone deacetylase inhibitors in the treatment of lymphoma. Discov Med. 2010;10:462–470. [PubMed] [Google Scholar]
- 185.Gui CY, Ngo L, Xu WS, Richon VM, Marks PA. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci U S A. 2004;101:1241–1246. doi: 10.1073/pnas.0307708100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Richon VM, Sandhoff TW, Rifkind RA, Marks PA. Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci U S A. 2000;97:10014–10019. doi: 10.1073/pnas.180316197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sandor V, Senderowicz A, Mertins S, Sackett D, Sausville E, Blagosklonny MV, Bates SE. P21-dependent g(1)arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br J Cancer. 2000;83:817–825. doi: 10.1054/bjoc.2000.1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zhang C, Richon V, Ni X, Talpur R, Duvic M. Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. J Invest Dermatol. 2005;125:1045–1052. doi: 10.1111/j.0022-202X.2005.23925.x. [DOI] [PubMed] [Google Scholar]
- 189.Shao W, Growney JD, Feng Y, O’Connor G, Pu M, Zhu W, Yao YM, Kwon P, Fawell S, Atadja P. Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: Defining molecular mechanisms of resistance. Int J Cancer. 2010;127:2199–2208. doi: 10.1002/ijc.25218. [DOI] [PubMed] [Google Scholar]
- 190.Tang Y, Zhao W, Chen Y, Zhao Y, Gu W. Acetylation is indispensable for p53 activation. Cell. 2008;133:612–626. doi: 10.1016/j.cell.2008.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Zhao Y, Lu S, Wu L, Chai G, Wang H, Chen Y, Sun J, Yu Y, Zhou W, Zheng Q, Wu M, Otterson GA, Zhu WG. Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21(Waf1/Cip1) Mol Cell Biol. 2006;26:2782–2790. doi: 10.1128/MCB.26.7.2782-2790.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Dai Y, Rahmani M, Dent P, Grant S. Blockade of histone deacetylase inhibitor-induced RelA/p65 acetylation and NF-kappaB activation potentiates apoptosis in leukemia cells through a process mediated by oxidative damage, XIAP downregulation, and c-Jun N-terminal kinase 1 activation. Mol Cell Biol. 2005;25:5429–5444. doi: 10.1128/MCB.25.13.5429-5444.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Zhang XD, Gillespie SK, Borrow JM, Hersey P. The histone deacetylase inhibitor suberic bishydroxamate regulates the expression of multiple apoptotic mediators and induces mitochondria-dependent apoptosis of melanoma cells. Mol Cancer Ther. 2004;3:425–435. [PubMed] [Google Scholar]
- 194.Kim SH, Jeong JW, Park JA, Lee JW, Seo JH, Jung BK, Bae MK, Kim KW. Regulation of the HIF-1alpha stability by histone deacetylases. Oncol Rep. 2007;17:647–651. [PubMed] [Google Scholar]
- 195.Heider U, Kaiser M, Sterz J, Zavrski I, Jakob C, Fleissner C, Eucker J, Possinger K, Sezer O. Histone deacetylase inhibitors reduce VEGF production and induce growth suppression and apoptosis in human mantle cell lymphoma. Eur J Haematol. 2006;76:42–50. doi: 10.1111/j.1600-0609.2005.00546.x. [DOI] [PubMed] [Google Scholar]
- 196.Catley L, Weisberg E, Kiziltepe T, Tai YT, Hideshima T, Neri P, Tassone P, Atadja P, Chauhan D, Munshi NC, Anderson KC. Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood. 2006;108:3441–3449. doi: 10.1182/blood-2006-04-016055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Munshi A, Kurland JF, Nishikawa T, Tanaka T, Hobbs ML, Tucker SL, Ismail S, Stevens C, Meyn RE. Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity. Clin Cancer Res. 2005;11:4912–4922. doi: 10.1158/1078-0432.CCR-04-2088. [DOI] [PubMed] [Google Scholar]
- 198.Rosato RR, Almenara JA, Grant S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res. 2003;63:3637–3645. [PubMed] [Google Scholar]
- 199.Martirosyan A, Leonard S, Shi X, Griffith B, Gannett P, Strobl J. Actions of a histone deacetylase inhibitor NSC3852 (5-nitroso-8-quinolinol) link reactive oxygen species to cell differentiation and apoptosis in MCF-7 human mammary tumor cells. J Pharmacol Exp Ther. 2006;317:546–552. doi: 10.1124/jpet.105.096891. [DOI] [PubMed] [Google Scholar]
- 200.Weiser TS, Ohnmacht GA, Guo ZS, Fischette MR, Chen GA, Hong JA, Nguyen DM, Schrump DS. Induction of MAGE-3 expression in lung and esophageal cancer cells. Ann Thorac Surg. 2001;71:295–301. doi: 10.1016/s0003-4975(00)02421-8. discussion 301–292. [DOI] [PubMed] [Google Scholar]
- 201.Tiffon C, Adams J, van der Fits L, Wen S, Townsend P, Ganesan A, Hodges E, Vermeer M, Packham G. The histone deacetylase inhibitors vorinostat and romidepsin downmodulate IL-10 expression in cutaneous T-cell lymphoma cells. Br J Pharmacol. 2011;162:1590–1602. doi: 10.1111/j.1476-5381.2010.01188.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Gloghini A, Buglio D, Khaskhely NM, Georgakis G, Orlowski RZ, Neelapu SS, Carbone A, Younes A. Expression of histone deacetylases in lymphoma: implication for the development of selective inhibitors. Br J Haematol. 2009;147:515–525. doi: 10.1111/j.1365-2141.2009.07887.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Prince HM, Bishton MJ, Harrison SJ. Clinical studies of histone deacetylase inhibitors. Clin Cancer Res. 2009;15:3958–3969. doi: 10.1158/1078-0432.CCR-08-2785. [DOI] [PubMed] [Google Scholar]
- 204.Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, Chiao JH, Reilly JF, Ricker JL, Richon VM, Frankel SR. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL) Blood. 2007;109:31–39. doi: 10.1182/blood-2006-06-025999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Olsen EA, Kim YH, Kuzel TM, Pacheco TR, Foss FM, Parker S, Frankel SR, Chen C, Ricker JL, Arduino JM, Duvic M. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol. 2007;25:3109–3115. doi: 10.1200/JCO.2006.10.2434. [DOI] [PubMed] [Google Scholar]
- 206.Kim E, Rook A, Kim Y, Demierre MF, Lerner A, Duvic M, Robak T, Becker JC, McCulloch W, Whittaker S. Romidepsin activity in all three disease compartments (skin, blood, lymph nodes) in patients with cutaneous T-cell lymphoma (CTCL) J Clin Oncol. 2010;28 abstract 8047. [Google Scholar]
- 207.Duvic M, Olsen EA, Breneman D, Pacheco TR, Parker S, Vonderheid EC, Abuav R, Ricker JL, Rizvi S, Chen C, Boileau K, Gunchenko A, Sanz-Rodriguez C, Geskin LJ. Evaluation of the long-term tolerability and clinical benefit of vorinostat in patients with advanced cutaneous T-cell lymphoma. Clin Lymphoma Myeloma. 2009;9:412–416. doi: 10.3816/CLM.2009.n.082. [DOI] [PubMed] [Google Scholar]
- 208.Sager PT, Balser B, Wolfson J, Nichols J, Pilot R, Jones S, Burris HA. Electrocardiographic effects of class 1 selective histone deacetylase inhibitor romidepsin. Cancer medicine. 2015;4:1178–1185. doi: 10.1002/cam4.467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Whittaker SJ, Demierre MF, Kim EJ, Rook AH, Lerner A, Duvic M, Scarisbrick J, Reddy S, Robak T, Becker JC, Samtsov A, McCulloch W, Kim YH. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol. 2010;28:4485–4491. doi: 10.1200/JCO.2010.28.9066. [DOI] [PubMed] [Google Scholar]
- 210.Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, Zain J, Prince HM, Leonard JP, Geskin LJ, Reeder C, Joske D, Figg WD, Gardner ER, Steinberg SM, Jaffe ES, Stetler-Stevenson M, Lade S, Fojo AT, Bates SE. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol. 2009;27:5410–5417. doi: 10.1200/JCO.2008.21.6150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Piekarz RL, Frye AR, Wright JJ, Steinberg SM, Liewehr DJ, Rosing DR, Sachdev V, Fojo T, Bates SE. Cardiac studies in patients treated with depsipeptide, FK228, in a phase II trial for T-cell lymphoma. Clin Cancer Res. 2006;12:3762–3773. doi: 10.1158/1078-0432.CCR-05-2095. [DOI] [PubMed] [Google Scholar]
- 212.Ellis L, Pan Y, Smyth GK, George DJ, McCormack C, Williams-Truax R, Mita M, Beck J, Burris H, Ryan G, Atadja P, Butterfoss D, Dugan M, Culver K, Johnstone RW, Prince HM. Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res. 2008;14:4500–4510. doi: 10.1158/1078-0432.CCR-07-4262. [DOI] [PubMed] [Google Scholar]
- 213.Pohlman B, Advani RH, Duvic M, Hymes K, Intragumtornchai T, Lekhakula A, Shpilberg O, Lerner A, Ben-Yehuda D, Beylot-Barry M, Hillen U, Fagerberg J, Foss F. Final Results of a Phase II Trial of Belinostat (PXD101) in Patients with Recurrent or Refractory Peripheral or Cutaneous T-Cell Lymphoma. Blood. 2009;114 abstract 920. [Google Scholar]
- 214.Fantin VR, Loboda A, Paweletz CP, Hendrickson RC, Pierce JW, Roth JA, Li L, Gooden F, Korenchuk S, Hou XS, Harrington EA, Randolph S, Reilly JF, Ware CM, Kadin ME, Frankel SR, Richon VM. Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res. 2008;68:3785–3794. doi: 10.1158/0008-5472.CAN-07-6091. [DOI] [PubMed] [Google Scholar]
- 215.Robey RW, Zhan Z, Piekarz RL, Kayastha GL, Fojo T, Bates SE. Increased MDR1 expression in normal and malignant peripheral blood mononuclear cells obtained from patients receiving depsipeptide (FR901228, FK228, NSC630176) Clin Cancer Res. 2006;12:1547–1555. doi: 10.1158/1078-0432.CCR-05-1423. [DOI] [PubMed] [Google Scholar]
- 216.Karpova MB, Gunz D, Okoniewski MJ, Cozzio A, Schad K, Conzett Baumann K, Dummer R. Transcriptome adaptation caused by vorinostat/bexarotene combination therapy in advanced cutaneous T-cell lymphoma. J Clin Oncol. 2010;28 abstract 8050. [Google Scholar]
- 217.Khan O, Fotheringham S, Wood V, Stimson L, Zhang C, Pezzella F, Duvic M, Kerr DJ, La Thangue NB. HR23B is a biomarker for tumor sensitivity to HDAC inhibitor-based therapy. Proc Natl Acad Sci U S A. 2010;107:6532–6537. doi: 10.1073/pnas.0913912107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Chakraborty AR, Robey RW, Luchenko VL, Zhan Z, Piekarz RL, Gillet JP, Kossenkov AV, Wilkerson J, Showe LC, Gottesman MM, Collie NL, Bates SE. MAPK pathway activation leads to Bim loss and histone deacetylase inhibitor resistance: rationale to combine romidepsin with an MEK inhibitor. Blood. 2013;121:4115–4125. doi: 10.1182/blood-2012-08-449140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Heider U, Rademacher J, Lamottke B, Mieth M, Moebs M, von Metzler I, Assaf C, Sezer O. Synergistic interaction of the histone deacetylase inhibitor SAHA with the proteasome inhibitor bortezomib in cutaneous T cell lymphoma. Eur J Haematol. 2009;82:440–449. doi: 10.1111/j.1600-0609.2009.01239.x. [DOI] [PubMed] [Google Scholar]
- 220.Dummer R, Hymes K, Sterry W, Steinhoff M, Assaf C, Kerl H, Ahern J, Rizvi S, Ricker JL, Whittaker S. Vorinostat in combination with bexarotene in advanced cutaneous T-cell lymphoma: A phase I study. J Clin Oncol. 2009;27 abstract 8572. [Google Scholar]
- 221.Olsen EA, Rosen ST, Vollmer RT, Variakojis D, Roenigk HH, Jr, Diab N, Zeffren J. Interferon alfa-2a in the treatment of cutaneous T cell lymphoma. J Am Acad Dermatol. 1989;20:395–407. doi: 10.1016/s0190-9622(89)70049-9. [DOI] [PubMed] [Google Scholar]
- 222.Sun WH, Pabon C, Alsayed Y, Huang PP, Jandeska S, Uddin S, Platanias LC, Rosen ST. Interferon-alpha resistance in a cutaneous T-cell lymphoma cell line is associated with lack of STAT1 expression. Blood. 1998;91:570–576. [PubMed] [Google Scholar]
- 223.Bunn PA, Jr, Foon KA, Ihde DC, Longo DL, Eddy J, Winkler CF, Veach SR, Zeffren J, Sherwin S, Oldham R. Recombinant leukocyte A interferon: an active agent in advanced cutaneous T-cell lymphomas. Ann Intern Med. 1984;101:484–487. doi: 10.7326/0003-4819-101-4-484. [DOI] [PubMed] [Google Scholar]
- 224.Jumbou O, N’Guyen JM, Tessier MH, Legoux B, Dreno B. Long-term follow-up in 51 patients with mycosis fungoides and Sezary syndrome treated by interferon-alfa. Br J Dermatol. 1999;140:427–431. doi: 10.1046/j.1365-2133.1999.02704.x. [DOI] [PubMed] [Google Scholar]
- 225.Polansky M, Talpur R, Daulat S, Hosing C, Dabaja B, Duvic M. Long-Term Complete Responses to Combination Therapies and Allogeneic Stem Cell Transplants in Patients With Sezary Syndrome. Clinical lymphoma, myeloma & leukemia. 2015;15:e83–93. doi: 10.1016/j.clml.2014.09.013. [DOI] [PubMed] [Google Scholar]
- 226.Olsen EA, Bunn PA. Interferon in the treatment of cutaneous T-cell lymphoma. Hematol Oncol Clin North Am. 1995;9:1089–1107. [PubMed] [Google Scholar]
- 227.Kuzel TM, Gilyon K, Springer E, Variakojis D, Kaul K, Bunn PA, Jr, Evans L, Roenigk HH, Jr, Rosen ST. Interferon alfa-2a combined with phototherapy in the treatment of cutaneous T-cell lymphoma. J Natl Cancer Inst. 1990;82:203–207. doi: 10.1093/jnci/82.3.203. [DOI] [PubMed] [Google Scholar]
- 228.Straus DJ, Duvic M, Kuzel T, Horwitz S, Demierre MF, Myskowski P, Steckel S. Results of a phase II trial of oral bexarotene (Targretin) combined with interferon alfa-2b (Intron-A) for patients with cutaneous T-cell lymphoma. Cancer. 2007;109:1799–1803. doi: 10.1002/cncr.22596. [DOI] [PubMed] [Google Scholar]
- 229.Dippel E, Schrag H, Goerdt S, Orfanos CE. Extracorporeal photopheresis and interferon-alpha in advanced cutaneous T-cell lymphoma. Lancet. 1997;350:32–33. doi: 10.1016/s0140-6736(05)66242-3. [DOI] [PubMed] [Google Scholar]
- 230.Foss FM, Ihde DC, Breneman DL, Phelps RM, Fischmann AB, Schechter GP, Linnoila I, Breneman JC, Cotelingam JD, Ghosh BC, et al. Phase II study of pentostatin and intermittent high-dose recombinant interferon alfa-2a in advanced mycosis fungoides/Sezary syndrome. J Clin Oncol. 1992;10:1907–1913. doi: 10.1200/JCO.1992.10.12.1907. [DOI] [PubMed] [Google Scholar]
- 231.Fritz TM, Kleinhans M, Nestle FO, Burg G, Dummer R. Combination treatment with extracorporeal photopheresis, interferon alfa and interleukin-2 in a patient with the Sezary syndrome. Br J Dermatol. 1999;140:1144–1147. doi: 10.1046/j.1365-2133.1999.02889.x. [DOI] [PubMed] [Google Scholar]
- 232.Zachariae H, Thestrup-Pedersen K. Interferon alpha and etretinate combination treatment of cutaneous T-cell lymphoma. J Invest Dermatol. 1990;95:206S–208S. doi: 10.1111/1523-1747.ep12875616. [DOI] [PubMed] [Google Scholar]
- 233.Papa G, Tura S, Mandelli F, Vegna ML, Defazio D, Mazza P, Zinzani PL, Simoni R, DePita O, Ferranti G, et al. Is interferon alpha in cutaneous T-cell lymphoma a treatment of choice? Br J Haematol. 1991;79(Suppl 1):48–51. doi: 10.1111/j.1365-2141.1991.tb08119.x. [DOI] [PubMed] [Google Scholar]
- 234.Rupoli S, Barulli S, Guiducci B, Offidani M, Mozzicafreddo G, Simonacci M, Filosa G, Giacchetti A, Ricotti G, Brandozzi G, Cataldi I, Serresi S, Ceschini R, Bugatti L, Offidani A, Giangiacomi M, Brancorsini D, Leoni P. Low dose interferon-alpha2b combined with PUVA is an effective treatment of early stage mycosis fungoides: results of a multicenter study. Cutaneous-T Cell Lymphoma Multicenter Study Group. Haematologica. 1999;84:809–813. [PubMed] [Google Scholar]
- 235.Kuzel TM, Roenigk HH, Jr, Samuelson E, Herrmann JJ, Hurria A, Rademaker AW, Rosen ST. Effectiveness of interferon alfa-2a combined with phototherapy for mycosis fungoides and the Sezary syndrome. J Clin Oncol. 1995;13:257–263. doi: 10.1200/JCO.1995.13.1.257. [DOI] [PubMed] [Google Scholar]
- 236.Roenigk HH, Jr, Kuzel TM, Skoutelis AP, Springer E, Yu G, Caro W, Gilyon K, Variakojis D, Kaul K, Bunn PA, Jr, et al. Photochemotherapy alone or combined with interferon alpha-2a in the treatment of cutaneous T-cell lymphoma. J Invest Dermatol. 1990;95:198S–205S. doi: 10.1111/1523-1747.ep12875523. [DOI] [PubMed] [Google Scholar]
- 237.Chiarion-Sileni V, Bononi A, Fornasa CV, Soraru M, Alaibac M, Ferrazzi E, Redelotti R, Peserico A, Monfardini S, Salvagno L. Phase II trial of interferon-alpha-2a plus psolaren with ultraviolet light A in patients with cutaneous T-cell lymphoma. Cancer. 2002;95:569–575. doi: 10.1002/cncr.10706. [DOI] [PubMed] [Google Scholar]
- 238.Foss FM, Ihde DC, Linnoila IR, Fischmann AB, Schechter GP, Cotelingam JD, Steinberg SM, Ghosh BC, Stocker JL, Bastian A, et al. Phase II trial of fludarabine phosphate and interferon alfa-2a in advanced mycosis fungoides/Sezary syndrome. J Clin Oncol. 1994;12:2051–2059. doi: 10.1200/JCO.1994.12.10.2051. [DOI] [PubMed] [Google Scholar]
- 239.Suchin KR, Cucchiara AJ, Gottleib SL, Wolfe JT, DeNardo BJ, Macey WH, Bromley PG, Vittorio CC, Rook AH. Treatment of cutaneous T-cell lymphoma with combined immunomodulatory therapy: a 14-year experience at a single institution. Arch Dermatol. 2002;138:1054–1060. doi: 10.1001/archderm.138.8.1054. [DOI] [PubMed] [Google Scholar]
- 240.Bladon J, Taylor PC. Lymphocytes treated by extracorporeal photopheresis demonstrate a drop in the Bcl-2/Bax ratio: a possible mechanism involved in extracorporeal-photopheresis-induced apoptosis. Dermatology. 2002;204:104–107. doi: 10.1159/000051826. [DOI] [PubMed] [Google Scholar]
- 241.Bladon J, Taylor PC. Extracorporeal photopheresis: a focus on apoptosis and cytokines. J Dermatol Sci. 2006;43:85–94. doi: 10.1016/j.jdermsci.2006.05.004. [DOI] [PubMed] [Google Scholar]
- 242.Osella-Abate S, Zaccagna A, Savoia P, Quaglino P, Salomone B, Bernengo MG. Expression of apoptosis markers on peripheral blood lymphocytes from patients with cutaneous T-cell lymphoma during extracorporeal photochemotherapy. J Am Acad Dermatol. 2001;44:40–47. doi: 10.1067/mjd.2001.108376. [DOI] [PubMed] [Google Scholar]
- 243.Berger C, Hoffmann K, Vasquez JG, Mane S, Lewis J, Filler R, Lin A, Zhao H, Durazzo T, Baird A, Lin W, Foss F, Christensen I, Girardi M, Tigelaar R, Edelson R. Rapid generation of maturationally synchronized human dendritic cells: contribution to the clinical efficacy of extracorporeal photochemotherapy. Blood. 2010;116:4838–4847. doi: 10.1182/blood-2009-11-256040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Berger CL, Xu AL, Hanlon D, Lee C, Schechner J, Glusac E, Christensen I, Snyder E, Holloway V, Tigelaar R, Edelson RL. Induction of human tumor-loaded dendritic cells. Int J Cancer. 2001;91:438–447. doi: 10.1002/1097-0215(200002)9999:9999<::aid-ijc1073>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
- 245.Girardi M, Berger CL, Wilson LD, Christensen IR, Thompson KR, Glusac EJ, Edelson RL. Transimmunization for cutaneous T cell lymphoma: a Phase I study. Leuk Lymphoma. 2006;47:1495–1503. doi: 10.1080/10428190600581419. [DOI] [PubMed] [Google Scholar]
- 246.Edelson R, Berger C, Gasparro F, Jegasothy B, Heald P, Wintroub B, Vonderheid E, Knobler R, Wolff K, Plewig G, et al. Treatment of cutaneous T-cell lymphoma by extracorporeal photochemotherapy. Preliminary results. N Engl J Med. 1987;316:297–303. doi: 10.1056/NEJM198702053160603. [DOI] [PubMed] [Google Scholar]
- 247.Knobler R, Jantschitsch C. Extracorporeal photochemoimmunotherapy in cutaneous T-cell lymphoma. Transfus Apher Sci. 2003;28:81–89. doi: 10.1016/S1473-0502(02)00103-9. [DOI] [PubMed] [Google Scholar]
- 248.Zic JA. The treatment of cutaneous T-cell lymphoma with photopheresis. Dermatol Ther. 2003;16:337–346. doi: 10.1111/j.1396-0296.2003.01646.x. [DOI] [PubMed] [Google Scholar]
- 249.Quaglino P, Knobler R, Fierro MT, Savoia P, Marra E, Fava P, Bernengo MG. Extracorporeal photopheresis for the treatment of erythrodermic cutaneous T-cell lymphoma: a single center clinical experience with long-term follow-up data and a brief overview of the literature. Int J Dermatol. 2013;52:1308–1318. doi: 10.1111/ijd.12121. [DOI] [PubMed] [Google Scholar]
- 250.Knobler R, Berlin G, Calzavara-Pinton P, Greinix H, Jaksch P, Laroche L, Ludvigsson J, Quaglino P, Reinisch W, Scarisbrick J, Schwarz T, Wolf P, Arenberger P, Assaf C, Bagot M, Barr M, Bohbot A, Bruckner-Tuderman L, Dreno B, Enk A, French L, Gniadecki R, Gollnick H, Hertl M, Jantschitsch C, Jung A, Just U, Klemke CD, Lippert U, Luger T, Papadavid E, Pehamberger H, Ranki A, Stadler R, Sterry W, Wolf IH, Worm M, Zic J, Zouboulis CC, Hillen U. Guidelines on the use of extracorporeal photopheresis. J Eur Acad Dermatol Venereol. 2014;28(Suppl 1):1–37. doi: 10.1111/jdv.12311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Gottlieb SL, Wolfe JT, Fox FE, DeNardo BJ, Macey WH, Bromley PG, Lessin SR, Rook AH. Treatment of cutaneous T-cell lymphoma with extracorporeal photopheresis monotherapy and in combination with recombinant interferon alfa: a 10-year experience at a single institution. J Am Acad Dermatol. 1996;35:946–957. doi: 10.1016/s0190-9622(96)90119-x. [DOI] [PubMed] [Google Scholar]
- 252.Heald P, Rook A, Perez M, Wintroub B, Knobler R, Jegasothy B, Gasparro F, Berger C, Edelson R. Treatment of erythrodermic cutaneous T-cell lymphoma with extracorporeal photochemotherapy. J Am Acad Dermatol. 1992;27:427–433. doi: 10.1016/0190-9622(92)70212-x. [DOI] [PubMed] [Google Scholar]
- 253.Zic JA, Stricklin GP, Greer JP, Kinney MC, Shyr Y, Wilson DC, King LE., Jr Long-term follow-up of patients with cutaneous T-cell lymphoma treated with extracorporeal photochemotherapy. J Am Acad Dermatol. 1996;35:935–945. doi: 10.1016/s0190-9622(96)90118-8. [DOI] [PubMed] [Google Scholar]
- 254.Wilson LD, Jones GW, Kim D, Rosenthal D, Christensen IR, Edelson RL, Heald PW, Kacinski BM. Experience with total skin electron beam therapy in combination with extracorporeal photopheresis in the management of patients with erythrodermic (T4) mycosis fungoides. J Am Acad Dermatol. 2000;43:54–60. doi: 10.1067/mjd.2000.105510. [DOI] [PubMed] [Google Scholar]
- 255.Wilson LD, Licata AL, Braverman IM, Edelson RL, Heald PW, Feldman AM, Kacinski BM. Systemic chemotherapy and extracorporeal photochemotherapy for T3 and T4 cutaneous T-cell lymphoma patients who have achieved a complete response to total skin electron beam therapy. Int J Radiat Oncol Biol Phys. 1995;32:987–995. doi: 10.1016/0360-3016(95)00073-8. [DOI] [PubMed] [Google Scholar]
- 256.Tsirigotis P, Pappa V, Papageorgiou S, Kapsimali V, Giannopoulou V, Kaitsa I, Girkas K, Papageorgiou E, Stavrianeas N, Economopoulos T, Dervenoulas J. Extracorporeal photopheresis in combination with bexarotene in the treatment of mycosis fungoides and Sezary syndrome. Br J Dermatol. 2007;156:1379–1381. doi: 10.1111/j.1365-2133.2007.07901.x. [DOI] [PubMed] [Google Scholar]
- 257.Ginaldi L, De Martinis M, Matutes E, Farahat N, Morilla R, Dyer MJ, Catovsky D. Levels of expression of CD52 in normal and leukemic B and T cells: correlation with in vivo therapeutic responses to Campath-1H. Leuk Res. 1998;22:185–191. doi: 10.1016/s0145-2126(97)00158-6. [DOI] [PubMed] [Google Scholar]
- 258.Lundin J, Hagberg H, Repp R, Cavallin-Stahl E, Freden S, Juliusson G, Rosenblad E, Tjonnfjord G, Wiklund T, Osterborg A. Phase 2 study of alemtuzumab (anti-CD52 monoclonal antibody) in patients with advanced mycosis fungoides/Sezary syndrome. Blood. 2003;101:4267–4272. doi: 10.1182/blood-2002-09-2802. [DOI] [PubMed] [Google Scholar]
- 259.Bernengo MG, Quaglino P, Comessatti A, Ortoncelli M, Novelli M, Lisa F, Fierro MT. Low-dose intermittent alemtuzumab in the treatment of Sezary syndrome: clinical and immunologic findings in 14 patients. Haematologica. 2007;92:784–794. doi: 10.3324/haematol.11127. [DOI] [PubMed] [Google Scholar]
- 260.Fisher DC, Tawa M, Walsh M, Clark RA, Kupper TS. Low-dose alemtuzumab is uniquely effective in refractory leukemic cutaneous T-cell lymphoma (L-CTCL) Blood. 2009;114 abstract 3748. [Google Scholar]
- 261.Thursky KA, Worth LJ, Seymour JF, Miles Prince H, Slavin MA. Spectrum of infection, risk and recommendations for prophylaxis and screening among patients with lymphoproliferative disorders treated with alemtuzumab*. Br J Haematol. 2006;132:3–12. doi: 10.1111/j.1365-2141.2005.05789.x. [DOI] [PubMed] [Google Scholar]
- 262.Enblad G, Hagberg H, Erlanson M, Lundin J, MacDonald AP, Repp R, Schetelig J, Seipelt G, Osterborg A. A pilot study of alemtuzumab (anti-CD52 monoclonal antibody) therapy for patients with relapsed or chemotherapy-refractory peripheral T-cell lymphomas. Blood. 2004;103:2920–2924. doi: 10.1182/blood-2003-10-3389. [DOI] [PubMed] [Google Scholar]
- 263.Gautschi O, Blumenthal N, Streit M, Solenthaler M, Hunziker T, Zenhausern R. Successful treatment of chemotherapy-refractory Sezary syndrome with alemtuzumab (Campath-1H) Eur J Haematol. 2004;72:61–63. doi: 10.1046/j.0902-4441.2004.00169.x. [DOI] [PubMed] [Google Scholar]
- 264.Kennedy GA, Seymour JF, Wolf M, Januszewicz H, Davison J, McCormack C, Ryan G, Prince HM. Treatment of patients with advanced mycosis fungoides and Sezary syndrome with alemtuzumab. Eur J Haematol. 2003;71:250–256. doi: 10.1034/j.1600-0609.2003.00143.x. [DOI] [PubMed] [Google Scholar]
- 265.O’Mahony D, Morris JC, Moses L, O’Hagan D, Gao W, Stetler-Stevenson M, Taylor M, Hammershaimb L, Waldman TA, Janik JE. Phase I Trial of Siplizumab in CD2-Positive Lymphoproliferative Disease. Blood. 2005;106 abstract 3353. [Google Scholar]
- 266.Kim YH, Duvic M, Obitz E, Gniadecki R, Iversen L, Osterborg A, Whittaker S, Illidge TM, Schwarz T, Kaufmann R, Cooper K, Knudsen KM, Lisby S, Baadsgaard O, Knox SJ. Clinical efficacy of zanolimumab (HuMax-CD4): two phase 2 studies in refractory cutaneous T-cell lymphoma. Blood. 2007;109:4655–4662. doi: 10.1182/blood-2006-12-062877. [DOI] [PubMed] [Google Scholar]
- 267.Kreitman RJ, Wilson WH, White JD, Stetler-Stevenson M, Jaffe ES, Giardina S, Waldmann TA, Pastan I. Phase I trial of recombinant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) in patients with hematologic malignancies. J Clin Oncol. 2000;18:1622–1636. doi: 10.1200/JCO.2000.18.8.1622. [DOI] [PubMed] [Google Scholar]
- 268.Suzuki R. Dosing of a phase I study of KW-0761, an anti-CCR4 antibody, for adult T-cell leukemia-lymphoma and peripheral T-cell lymphoma. J Clin Oncol. 2010;28:e404–405. doi: 10.1200/JCO.2010.29.7242. author reply e406. [DOI] [PubMed] [Google Scholar]
- 269.Yamamoto K, Utsunomiya A, Tobinai K, Tsukasaki K, Uike N, Uozumi K, Yamaguchi K, Yamada Y, Hanada S, Tamura K, Nakamura S, Inagaki H, Ohshima K, Kiyoi H, Ishida T, Matsushima K, Akinaga S, Ogura M, Tomonaga M, Ueda R. Phase I study of KW-0761, a defucosylated humanized anti-CCR4 antibody, in relapsed patients with adult T-cell leukemia-lymphoma and peripheral T-cell lymphoma. J Clin Oncol. 2010;28:1591–1598. doi: 10.1200/JCO.2009.25.3575. [DOI] [PubMed] [Google Scholar]
- 270.Duvic M, Pinter-Brown L, Foss F, Sokol L, Jorgensen J, Spitalny GL, Kim YH. Results of a phase 1/2 Study for KW-0761, a Monoclonal Antibody Directed Against CC Chemokine Receptor Type 4 (CCR4), In CTCL Patients. Blood. 2010;116 Abstract 285. [Google Scholar]
- 271.Ogura M, Ishida T, Hatake K, Taniwaki M, Ando K, Tobinai K, Fujimoto K, Yamamoto K, Miyamoto T, Uike N, Tanimoto M, Tsukasaki K, Ishizawa K, Suzumiya J, Inagaki H, Tamura K, Akinaga S, Tomonaga M, Ueda R. Multicenter Phase II Study of Mogamulizumab (KW-0761), a Defucosylated Anti-CC Chemokine Receptor 4 Antibody, in Patients With Relapsed Peripheral T-Cell Lymphoma and Cutaneous T-Cell Lymphoma. J Clin Oncol. 2014;32:1157–1163. doi: 10.1200/JCO.2013.52.0924. [DOI] [PubMed] [Google Scholar]
- 272.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: 10.1158/1078-0432.CCR-14-0830. [DOI] [PubMed] [Google Scholar]
- 273.Ni X, Langridge T, Duvic M. Depletion of regulatory T cells by targeting CC chemokine receptor type 4 with mogamulizumab. Oncoimmunology. 2015;4:e1011524. doi: 10.1080/2162402X.2015.1011524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Wilcox RA. Mogamulizumab: 2 birds, 1 stone. Blood. 2015;125:1847–1848. doi: 10.1182/blood-2015-02-625251. [DOI] [PubMed] [Google Scholar]
- 275.Krathen M, Sundram M, Bashey S, Sutherland K, Salva K, Wood GS, Advani R, Hoppe R, Reddy S, Armstrong R, Nagpal S, Pulitzer M, Horwitz S, Kim YH. Brentuximab Vedotin Demonstrates Significant Clinical Activity in Relapsed or Refractory Mycosis Fungoides with Variable CD30 Expression. Blood (ASH Annual Meeting Abstracts) 2012:120. [Google Scholar]
- 276.Frankel AE, Woo JH, Ahn C, Foss FM, Duvic M, Neville PH, Neville DM. Resimmune, an anti-CD3epsilon recombinant immunotoxin, induces durable remissions in patients with cutaneous T-cell lymphoma. Haematologica. 2015;100:794–800. doi: 10.3324/haematol.2015.123711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Whittaker SJ, Foss FM. Efficacy and tolerability of currently available therapies for the mycosis fungoides and Sezary syndrome variants of cutaneous T-cell lymphoma. Cancer Treat Rev. 2007;33:146–160. doi: 10.1016/j.ctrv.2006.08.006. [DOI] [PubMed] [Google Scholar]
- 278.Akpek G, Koh HK, Bogen S, O’Hara C, Foss FM. Chemotherapy with etoposide, vincristine, doxorubicin, bolus cyclophosphamide, and oral prednisone in patients with refractory cutaneous T-cell lymphoma. Cancer. 1999;86:1368–1376. [PubMed] [Google Scholar]
- 279.Molin L, Thomsen K, Volden G, Groth O, Hellbe L, Holst R, Knudsen EA, Roupe G, Schmidt H. Combination chemotherapy in the tumour stage of mycosis fungoides with cyclophosphamide, vincristine, vp-16, adriamycin and prednisolone (cop, chop, cavop): a report from the Scandinavian mycosis fungoides study group. Acta Derm Venereol. 1980;60:542–544. [PubMed] [Google Scholar]
- 280.Duvic M, Lemak NA, Redman JR, Eifel PJ, Tucker SL, Cabanillas FF, Kurzrock R. Combined modality therapy for cutaneous T-cell lymphoma. J Am Acad Dermatol. 1996;34:1022–1029. doi: 10.1016/s0190-9622(96)90282-0. [DOI] [PubMed] [Google Scholar]
- 281.Zackheim HS, Epstein EH., Jr Low-dose methotrexate for the Sezary syndrome. J Am Acad Dermatol. 1989;21:757–762. doi: 10.1016/s0190-9622(89)70251-6. [DOI] [PubMed] [Google Scholar]
- 282.Zackheim HS, Kashani-Sabet M, Hwang ST. Low-dose methotrexate to treat erythrodermic cutaneous T-cell lymphoma: results in twenty-nine patients. J Am Acad Dermatol. 1996;34:626–631. doi: 10.1016/s0190-9622(96)80062-4. [DOI] [PubMed] [Google Scholar]
- 283.Zackheim HS, Kashani-Sabet M, McMillan A. Low-dose methotrexate to treat mycosis fungoides: a retrospective study in 69 patients. J Am Acad Dermatol. 2003;49:873–878. doi: 10.1016/s0190-9622(03)01591-3. [DOI] [PubMed] [Google Scholar]
- 284.Vonderheid EC, Sajjadian A, Kadin ME. Methotrexate is effective therapy for lymphomatoid papulosis and other primary cutaneous CD30-positive lymphoproliferative disorders. J Am Acad Dermatol. 1996;34:470–481. doi: 10.1016/s0190-9622(96)90442-9. [DOI] [PubMed] [Google Scholar]
- 285.Zinzani PL, Magagnoli M, Bendandi M, Orcioni GF, Gherlinzoni F, Albertini P, Pileri SA, Tura S. Therapy with gemcitabine in pretreated peripheral T-cell lymphoma patients. Ann Oncol. 1998;9:1351–1353. doi: 10.1023/a:1008409601731. [DOI] [PubMed] [Google Scholar]
- 286.Zinzani PL, Baliva G, Magagnoli M, Bendandi M, Modugno G, Gherlinzoni F, Orcioni GF, Ascani S, Simoni R, Pileri SA, Tura S. Gemcitabine treatment in pretreated cutaneous T-cell lymphoma: experience in 44 patients. J Clin Oncol. 2000;18:2603–2606. doi: 10.1200/JCO.2000.18.13.2603. [DOI] [PubMed] [Google Scholar]
- 287.Marchi E, Alinari L, Tani M, Stefoni V, Pimpinelli N, Berti E, Pagano L, Bernengo MG, Zaja F, Rupoli S, Pileri S, Baccarani M, Zinzani PL. Gemcitabine as frontline treatment for cutaneous T-cell lymphoma: phase II study of 32 patients. Cancer. 2005;104:2437–2441. doi: 10.1002/cncr.21449. [DOI] [PubMed] [Google Scholar]
- 288.Duvic M, Talpur R, Wen S, Kurzrock R, David CL, Apisarnthanarax N. Phase II evaluation of gemcitabine monotherapy for cutaneous T-cell lymphoma. Clin Lymphoma Myeloma. 2006;7:51–58. doi: 10.3816/CLM.2006.n.039. [DOI] [PubMed] [Google Scholar]
- 289.Zinzani PL, Venturini F, Stefoni V, Fina M, Pellegrini C, Derenzini E, Gandolfi L, Broccoli A, Argnani L, Quirini F, Pileri S, Baccarani M. Gemcitabine as single agent in pretreated T-cell lymphoma patients: evaluation of the long-term outcome. Ann Oncol. 2010;21:860–863. doi: 10.1093/annonc/mdp508. [DOI] [PubMed] [Google Scholar]
- 290.Wollina U, Graefe T, Karte K. Treatment of relapsing or recalcitrant cutaneous T-cell lymphoma with pegylated liposomal doxorubicin. J Am Acad Dermatol. 2000;42:40–46. doi: 10.1016/s0190-9622(00)90007-0. [DOI] [PubMed] [Google Scholar]
- 291.Wollina U, Dummer R, Brockmeyer NH, Konrad H, Busch JO, Kaatz M, Knopf B, Koch HJ, Hauschild A. Multicenter study of pegylated liposomal doxorubicin in patients with cutaneous T-cell lymphoma. Cancer. 2003;98:993–1001. doi: 10.1002/cncr.11593. [DOI] [PubMed] [Google Scholar]
- 292.Pulini S, Rupoli S, Goteri G, Pimpinelli N, Alterini R, Tassetti A, Scortechini AR, Offidani M, Mulattieri S, Stronati A, Brandozzi G, Giacchetti A, Mozzicafreddo G, Ricotti G, Filosa G, Bettacchi A, Simonacci M, Novelli N, Leoni P. Pegylated liposomal doxorubicin in the treatment of primary cutaneous T-cell lymphomas. Haematologica. 2007;92:686–689. doi: 10.3324/haematol.10879. [DOI] [PubMed] [Google Scholar]
- 293.Quereux G, Marques S, Nguyen JM, Bedane C, D’Incan M, Dereure O, Puzenat E, Claudy A, Martin L, Joly P, Delaunay M, Beylot-Barry M, Vabres P, Celerier P, Sasolas B, Grange F, Khammari A, Dreno B. Prospective multicenter study of pegylated liposomal doxorubicin treatment in patients with advanced or refractory mycosis fungoides or Sezary syndrome. Arch Dermatol. 2008;144:727–733. doi: 10.1001/archderm.144.6.727. [DOI] [PubMed] [Google Scholar]
- 294.Cummings FJ, Kim K, Neiman RS, Comis RL, Oken MM, Weitzman SA, Mann RB, O’Connell MJ. Phase II trial of pentostatin in refractory lymphomas and cutaneous T-cell disease. J Clin Oncol. 1991;9:565–571. doi: 10.1200/JCO.1991.9.4.565. [DOI] [PubMed] [Google Scholar]
- 295.Dearden C, Matutes E, Catovsky D. Deoxycoformycin in the treatment of mature T-cell leukaemias. Br J Cancer. 1991;64:903–906. doi: 10.1038/bjc.1991.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Mercieca J, Matutes E, Dearden C, MacLennan K, Catovsky D. The role of pentostatin in the treatment of T-cell malignancies: analysis of response rate in 145 patients according to disease subtype. J Clin Oncol. 1994;12:2588–2593. doi: 10.1200/JCO.1994.12.12.2588. [DOI] [PubMed] [Google Scholar]
- 297.Greiner D, Olsen EA, Petroni G. Pentostatin (2′-deoxycoformycin) in the treatment of cutaneous T-cell lymphoma. J Am Acad Dermatol. 1997;36:950–955. doi: 10.1016/s0190-9622(97)80279-4. [DOI] [PubMed] [Google Scholar]
- 298.Ho AD, Suciu S, Stryckmans P, De Cataldo F, Willemze R, Thaler J, Peetermans M, Dohner H, Solbu G, Dardenne M, Zittoun R. Pentostatin in T-cell malignancies–a phase II trial of the EORTC. Leukemia Cooperative Group. Ann Oncol. 1999;10:1493–1498. doi: 10.1023/a:1008377724139. [DOI] [PubMed] [Google Scholar]
- 299.Kurzrock R, Pilat S, Duvic M. Pentostatin therapy of T-cell lymphomas with cutaneous manifestations. J Clin Oncol. 1999;17:3117–3121. doi: 10.1200/JCO.1999.17.10.3117. [DOI] [PubMed] [Google Scholar]
- 300.Tsimberidou AM, Giles F, Duvic M, Fayad L, Kurzrock R. Phase II study of pentostatin in advanced T-cell lymphoid malignancies: update of an M.D. Anderson Cancer Center series Cancer. 2004;100:342–349. doi: 10.1002/cncr.11899. [DOI] [PubMed] [Google Scholar]
- 301.Jidar K, Ingen-Housz-Oro S, Beylot-Barry M, Paul C, Chaoui D, Sigal-Grinberg M, Morel P, Dubertret L, Bachelez H. Gemcitabine treatment in cutaneous T-cell lymphoma: a multicentre study of 23 cases. Br J Dermatol. 2009;161:660–663. doi: 10.1111/j.1365-2133.2009.09230.x. [DOI] [PubMed] [Google Scholar]
- 302.O’Connor OA, Hamlin PA, Portlock C, Moskowitz CH, Noy A, Straus DJ, Macgregor-Cortelli B, Neylon E, Sarasohn D, Dumetrescu O, Mould DR, Fleischer M, Zelenetz AD, Sirotnak F, Horwitz S. Pralatrexate, a novel class of antifol with high affinity for the reduced folate carrier-type 1, produces marked complete and durable remissions in a diversity of chemotherapy refractory cases of T-cell lymphoma. Br J Haematol. 2007;139:425–428. doi: 10.1111/j.1365-2141.2007.06658.x. [DOI] [PubMed] [Google Scholar]
- 303.Serova M, Bieche I, Sablin MP, Pronk GJ, Vidaud M, Cvitkovic E, Faivre S, Raymond E. Single agent and combination studies of pralatrexate and molecular correlates of sensitivity. Br J Cancer. 2011;104:272–280. doi: 10.1038/sj.bjc.6606063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Zain J, O’Connor O. Pralatrexate: basic understanding and clinical development. Expert Opin Pharmacother. 2010;11:1705–1714. doi: 10.1517/14656566.2010.489552. [DOI] [PubMed] [Google Scholar]
- 305.O’Connor OA, Pro B, Pinter-Brown L, Bartlett N, Popplewell L, Coiffier B, Jo Lechowicz M, Savage KJ, Shustov AR, Gisselbrecht C, Jacobsen E, Zinzani PL, Furman R, Goy A, Haioun C, Crump M, Zain JM, Hsi E, Boyd A, Horwitz S. Pralatrexate in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results From the Pivotal PROPEL Study. J Clin Oncol. 2011;29:1182–1189. doi: 10.1200/JCO.2010.29.9024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Foss F, Horwitz S, Pinter-Brown L, Goy A, Pro B, Coiffier B, Popplewell L, Savage KJ, Shustov AR, Zain J, Koutsoukos T, Fruchtman SM, O’Connor OA. Pralatrexate Is An Effective Treatment for Heavily Pretreated Patients with Relapsed/Refractory Transformed Mycosis Fungoides (tMF) Blood. 2010;116 Abstract 1762. [Google Scholar]
- 307.Horwitz S, Kim YH, Foss F, Zain JM, Myskowski P, Lechowicz MJ, Fisher DC, Shustov AR, Bartlett N, Delioukina M, Koutsoukos T, Fruchtman SM, O’Connor OA, Duvic M. Identification of An Active, Well-Tolerated Dose of Pralatrexate In Patients with Relapsed or Refractory Cutaneous T-cell Lymphoma (CTCL): Final Results of a Multicenter Dose-Finding Study. Blood. 2010;116 doi: 10.1182/blood-2011-11-390211. Abstract 2800. [DOI] [PubMed] [Google Scholar]
- 308.Rueda A, Casanova M, Quero C, Medina-Perez A. Pralatrexate, a new hope for aggressive T-cell lymphomas? Clin Transl Oncol. 2009;11:215–220. doi: 10.1007/s12094-009-0343-9. [DOI] [PubMed] [Google Scholar]
- 309.Zinzani PL, Musuraca G, Tani M, Stefoni V, Marchi E, Fina M, Pellegrini C, Alinari L, Derenzini E, de Vivo A, Sabattini E, Pileri S, Baccarani M. Phase II trial of proteasome inhibitor bortezomib in patients with relapsed or refractory cutaneous T-cell lymphoma. J Clin Oncol. 2007;25:4293–4297. doi: 10.1200/JCO.2007.11.4207. [DOI] [PubMed] [Google Scholar]
- 310.Querfeld C, Rosen ST, Guitart J, Duvic M, Kim YH, Dusza SW, Kuzel TM. Results of an open-label multicenter phase 2 trial of lenalidomide monotherapy in refractory mycosis fungoides and Sezary syndrome. Blood. 2014;123:1159–1166. doi: 10.1182/blood-2013-09-525915. [DOI] [PubMed] [Google Scholar]
- 311.Wilcox RA. A three signal model of T-cell lymphoma pathogenesis. Am J Hematol. 2015 doi: 10.1002/ajh.24203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Wu PA, Kim YH, Lavori PW, Hoppe RT, Stockerl-Goldstein KE. A meta-analysis of patients receiving allogeneic or autologous hematopoietic stem cell transplant in mycosis fungoides and Sezary syndrome. Biology of Blood & Marrow Transplantation. 2009;15:982–990. doi: 10.1016/j.bbmt.2009.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313.Duarte RF, Schmitz N, Servitje O, Sureda A. Haematopoietic stem cell transplantation for patients with primary cutaneous T-cell lymphoma. Bone Marrow Transplantation. 2008;41:597–604. doi: 10.1038/sj.bmt.1705968. [DOI] [PubMed] [Google Scholar]
- 314.Duarte RF, Canals C, Onida F, Gabriel IH, Arranz R, Arcese W, Ferrant A, Kobbe G, Narni F, Deliliers GL, Olavarria E, Schmitz N, Sureda A. Allogeneic hematopoietic cell transplantation for patients with mycosis fungoides and Sezary syndrome: a retrospective analysis of the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. Journal of Clinical Oncology. 28:4492–4499. doi: 10.1200/JCO.2010.29.3241. [DOI] [PubMed] [Google Scholar]
- 315.Schlaak M, Theurich S, Pickenhain J, Skoetz N, Kurschat P, von Bergwelt-Baildon M. Allogeneic stem cell transplantation for advanced primary cutaneous T-cell lymphoma: a systematic review. Crit Rev Oncol Hematol. 2013;85:21–31. doi: 10.1016/j.critrevonc.2012.06.002. [DOI] [PubMed] [Google Scholar]