Cutaneous T-cell lymphoma (CTCL) is a heterogeneous disease group of unknown etiology with a complex immunological background. As CTCL arises from T cells that have a vital role in the antitumor response, their therapy is largely aimed at reversing the immunological mechanisms leading to or manifesting during this malignancy. Early disease stages can be controlled with skin-directed therapy in most CTCL cases. Still, advanced CTCL has a dismal prognosis and warrants systemic therapy. Despite considerable progress in understanding the pathophysiology of the disease and the numerous systemic treatment options available, long-term remission rates with conventional treatments alone are still low. Allogeneic hematopoietic stem cell transplantation is currently the only curative option for advanced CTCL, including mycosis fungoides and Sézary syndrome. The aims of this review is to summarize the recent findings on the immunology of this heterogeneous disease and to present the advances in its clinical management.

Cutaneous T-cell lymphomas (CTCL) are a heterogeneous group of non-Hodgkin lymphomas arising from the malignant proliferation of skin-homing or skin-resident T cells [1, 2]. Although CTCL manifestation in children exists [3], most CTCL typically affect the elderly, with a median age at diagnosis of 55–60 years and an average number of 6.4 new cases per year and per million people [4].

The 2 main subtypes of CTCL include the most frequent, i.e., mycosis fungoides (MF), accounting for approximately 60% of CTCL cases and 50% of all primary cutaneous lymphomas, and the rare leukemic variant Sézary syndrome (SS), representing around 5% of CTCL cases. The second most common group, representing approximately 25% of CTCL, is the group of primary cutaneous CD30+ lymphoproliferative disorders including primary cutaneous anaplastic large lymphoma and lymphomatoid papulosis [5]. Other CTCL subtypes are very rare [5, 6].

MF and SS originate from distinct CD4+ T-cell populations [7] and vary in terms of their prognosis as well as their clinical manifestations [8], with some overlapping symptoms such as chronic cutaneous lesions, associated with scaly rash, pruritus, burning, and sometimes pain [9] with recurrent infections, thus having a significant impact on the quality of life due to subsequent psychological problems and sleep disorders [10, 11]. Most patients with MF present a prolonged, indolent clinical course with initial skin involvement and clinical presentation depending on the stage at diagnosis [12]. Of all MF patients, 71.5% are in an early stage and 28.5% are in an advanced stage of the disease, and progression to a higher stage of the disease occurs in 9.7–11.6% of patients [13]. MF restricted to the skin progresses from a persistent patch stage with finely scaling lesions to a plaque and tumor stage, typically in sun-protected areas over years [8]. Extracutaneous disease initially involves regional lymph nodes and is mostly present with extensive skin involvement with tumors or erythroderma.

In CTCL leukemic variants, including SS and a portion of advanced-stage MF, malignant CD4+ T cells accumulate in the peripheral blood and visceral organs, with a subsequently dismal prognosis. SS is defined as an aggressive leukemic CTCL clinically defined by the triad of erythroderma, generalized lymphadenopathy, and the presence of neoplastic T cells in the skin, lymph nodes, and peripheral blood (Sézary cells) [8]. Cutaneous lesions include erythroderma, plantar and palmar keratoderma, onychodystrophy, ectropion, and diffuse alopecia [14].

The etiology of CTCL remains largely unknown. Viruses, such as human T-cell leukemia virus type 1, have been previously suggested as drivers of the disease [15], but recent studies do not present enough evidence to support the viral hypothesis in the pathogenesis of MF and SS [16]. Antigen-driven T-cell lymphoproliferation or dyscrasia following medication use [17], as well as genetic factors, i.e., HLA class II alleles predisposing to the disease [18], have been reported. Recent attempts to profile the genomic landscape of CTCL have demonstrated its high heterogeneity. Although the pathogenesis of the disease cannot be attributed to a small subset of well-defined somatic mutations, copy number variations, fusion proteins, and somatic mutations in diverse cellular and signaling pathways might contribute to the pathogenesis of the disease [19-22]. Those include alterations in factors functioning in epigenetic regulation, DNA damage response, cell cycle control, programmed cell death, and T-cell receptor (TCR) signaling, as well as nuclear factor (NF)-κB and Janus kinase (Jak)/signal transducer and activator of transcription (STAT) pathways [19, 20, 23-26]. Next to those intrinsic drivers, also extrinsic drivers, most commonly Staphylococcus aureus (SA) and its toxins, are under debate [27]. However, epidemiological studies could so far not reliably identify environmental exposure as a trigger for the disease [28].

As the name indicates, CTCL is a disease of T cells, although MF and SS appear to have their origin in different T-cell subtypes [7]. Based on flow cytometry analysis of lymph node or skin-homing molecules and differentiation markers, MF has been characterized as a malignancy of skin-resident effector memory T (TEM) cells (CCR4+ and CLA+), while SS cells present a phenotype of central memory T (TCM) cells (CCR7+, CD27+, and L-selectin+) [7]. These findings explain some of the differences in the clinical manifestation of the disease. TEM cells are skin-resident stationary polarized effector cells producing high amounts of inflammatory cytokines [29] that result in a skin-limited presentation in the form of patches, plaques, or tumors that remain stable for years. A progressive disease with the involvement of blood, lymph nodes, and viscera develops only in a small subset of patients [2]. In contrast, TCM are highly proliferative and actively recirculate between the blood, lymph nodes, and skin [2].

Differences in the genetic background of T-cell precursors in these CTCL subtypes have been further proven by comparative genomic hybridization and gene expression analyses [22, 30, 31]. However, malignant T cells differ not only between CTCL subtypes but also between patients with the same disease. In a cohort of SS patients it was observed that not all malignant T-cells were characterized by the TCMphenotype, but malignant cells from a subgroup of SS patients (18 out of 47 patients) expressed markers of a naive, more stem cell-like phenotype, such as high levels of CD45RA [32].

Finally, it has become evident that malignant T-cells show a high intrapatient variability, and follow-up data of SS patients suggests also an evolution in the phenotype of SS cells, demonstrating their plasticity [32]. Single-cell RNA sequencing combined with flow cytometry showed a high heterogeneity of classical T-cell markers within individual patients, with a common cluster of only 5 proteins (S100A4, S100A10, IL7R, CCR7, and CXCR4) [33].

Single-cell RNA sequencing of SS cells suggests also the existence of cases with a shift from a T-regulatory-like phenotype (characterized by FOXP3 expression) to a more central memory phenotype (characterized by the expression of a major Th2 driver – GATA3 or IKZF2) in malignant CD4+ cells [34]. In line with this finding, FOXP3 could be an important factor to predict early disease in CTCL, along with another 19 genes suggested to correlate with different CTCL stages [34]. Nevertheless, the role of Treg cells in the pathogenesis of SS is still controversial [35-37]. Some clinical cases in which CTCL appeared with the phenotype of malignant proliferation of Tregs have been observed [38]. Also, a recent study by Borcherding et al. [34] confirmed the presence of CD25- FOXP3+ tumor cells in a subset of SS patients. Of interest, in murine modes, an inadequate CD25 expression guarantees Treg plasticity and their differentiation into a Th phenotype depending on the cytokine milieu [39]. Hence, the CD25– FOXP3+ population may be an intermediate state of SS cells as well. Recent analyses suggest a marked decrease in the number of TCM following HDAC inhibition [33] which may be attributed to an open chromatin state leading to FOXP3 activation [40].

Early-Stage CTCL: Equilibrium Phase

The patient’s immune system has an important influence on the tumor fate. Many MF patients can have indolent disease over years, most likely presenting an equilibrium phase where the adaptive immune response can still control tumor outgrowth [11]. Early-stage skin lesions (stages IA to IIA) are infiltrated by a small number of malignant T cells surrounded by reactive immune cells, including large numbers of activated CD8+ T cells and T helper 1 (Th1) cells, resulting in the establishment of cell-mediated antitumor responses and secretion of cytotoxic molecules, including the proinflammatory cytokines interferon (IFN)-α and IFN-γ (Fig. 1a) [41-46]. In early-stage CTCL, the Th1 phenotype is maintained by the expression of signal transducer and activator of transcription 4 (STAT-4) and interleukin (IL)-12 signaling via JAK2/TYK2 [47].

Fig. 1.

Immune responses in early- and advanced-stage CTCL. a Early-stage CTCL. Upper panel: schematic illustration of the cellular composition in patch-stage disease. Small populations of tumor T cells are present mostly in the epidermis and are held in check by surrounding healthy T cells and regulatory T cells. Lower panel: immunosurveillance mechanisms are partially intact, the antitumor Th1 immune response can control tumor progression, leading to a tumor equilibrium. b Advanced-stage CTCL. Upper panel: schematic illustration of the cellular composition in plaque/tumor stage disease. The number of infiltrating tumor T cells is largely increased and present now also in the dermis, while the number of healthy T cells and T regulatory cells is decreased. Lower panel: immunosurveillance of the tumor is largely defective. A skewed Th1 immune response and an increased Th2 response, among other mechanisms, lead to immune escape of tumor cells and allow tumor progression.

Fig. 1.

Immune responses in early- and advanced-stage CTCL. a Early-stage CTCL. Upper panel: schematic illustration of the cellular composition in patch-stage disease. Small populations of tumor T cells are present mostly in the epidermis and are held in check by surrounding healthy T cells and regulatory T cells. Lower panel: immunosurveillance mechanisms are partially intact, the antitumor Th1 immune response can control tumor progression, leading to a tumor equilibrium. b Advanced-stage CTCL. Upper panel: schematic illustration of the cellular composition in plaque/tumor stage disease. The number of infiltrating tumor T cells is largely increased and present now also in the dermis, while the number of healthy T cells and T regulatory cells is decreased. Lower panel: immunosurveillance of the tumor is largely defective. A skewed Th1 immune response and an increased Th2 response, among other mechanisms, lead to immune escape of tumor cells and allow tumor progression.

Close modal

Advanced-Stage CTCL: Tumor Progression Phase

The escape from immune recognition can lead to tumor progression as observed in advanced-stage CTCL (stages IIB to IVB) with a large increase in infiltrating tumor T cells presenting on the skin with plaques and tumors (Fig. 1b). During disease progression, the expression of Th2 markers (e.g., GATA-3) and cytokines (e.g., IL-4, IL-5, and IL-10) increases, whereas the expression of Th1 transcription factors, such as T-cell-specific T-box transcription factor (T-bet), IFN-γ, STAT4, and IL-12 decreases [48, 49]. What shifts the balance in favor of tumor progression remains to be largely unknown in CTCL. One contributor might be mutations in the JAK/STAT pathway, making it persistently active in cancer T cells [49]. In early disease, there is constitutive activation of STAT5; in later disease there is activation of STAT3. STAT-5 via miR-155 reduces STAT4 expression, which is critical for the Th1 phenotype and thereby contributes to a Th1 to Th2 switch [49]. Moreover, STAT5 is known to be involved in the transcription of antiapoptotic proteins (bcl-2 and bcl-xl), cell cycle genes (cyclin D and c-myc), and IL-4 cytokines and its activation could be therefore an important driver of tumor cell proliferation [50, 51].

In addition to the dominance of the Th2 phenotype in advanced stage CTCL, several types of immune cells have been shown to contribute to a state of immune evasion of the tumor cells. Among them are subpopulations of dendritic cells (DC; immature CD209/DC-DIGN+ DC) [52] and subpopulations of CD4+ and CD8+ T cells with a high expression of immune checkpoint inhibitors [53]. Moreover, as already mentioned, also an impaired IL-12 production by DC contributes to the Th2 switch [54].

A clinical characteristic of advanced stage CTCL encompasses the susceptibility to infections as a consequence of impaired antigen-specific T-cell responses and decreased CD8 cytotoxic response [55]. The susceptibility to infections together with peripheral eosinophilia and high IgE and IgA levels further confirms a Th2-driven immunological process [55]. Interestingly, high IgE to environmental and food allergens in Sézary patients has been associated with a lower survival rate [56]. Therapies targeting the Th2 phenotype that would reinvert it into a Th1 immunological response hold potential for improving both the anticancer and the antipathogen response [41].

The presence and role of natural killer (NK) cells in CTCL skin lesions is still under debate. CTCL cells have been demonstrated to be susceptible to NK-induced killing in vitro [57]. Recently, an analysis of NK cells in peripheral blood showed no significant difference in the number of NK cells in CTCL versus healthy individuals [58]. This is in contrast with previous reports of decreased NK cell numbers in SS patients that suggested that their function may be increased by Toll-like receptor (TLR) stimulation [59]. On the other hand, higher numbers of circulating NK cells have been correlated with a poorer prognosis both in MF and in SS [58] and related to findings in another Th2-mediated disease, i.e., atopic dermatitis (AD), where NK dysregulation contributed to AD pathogenesis [60]. Although the reason for these findings is not entirely clear, they suggest that the existence of the inhibitory mechanism of the CTCL microenvironment has a significant role in suppressing the anti-tumor activity of NK cells in vivo. One such factors may be chronic IL-15 stimulation leading to an exhaustion state of NK cells, defined as their hyperactivation together with impaired recognition of malignant cells [58]. However, the involvement of NK cells in providing an antitumor response in the skin remains controversial, as NK cells have been described to be present in the skin in scarce amounts [61, 62]. The effectiveness of rituximab (anti-CD20 mAb) in the therapy of primary cutaneous B-cell lymphoma indicates that NK cells in the skin have valid antibody-dependent cell-mediated cytotoxicity (ADCC). However, in a phase 3 clinical trial, the reported response rate (RR) of mogamulizumab (anti-CCR4 mAb) was only 28% despite the high CCR4 expression in the study cohort [63]. Also, the RR in the skin was much lower compared blood. Impaired ADCC may also contribute to the inefficacy of anti-CD52 mAb (alemtuzumab) in MF [64, 65].

Neutrophils may also contribute to CTCL pathophysiology, as hyperactivated neutrophils are found in peripheral blood of CTCL patients (even in early disease) and the secreted IL-8 and LTB4 contribute to skin inflammation [66]. Moreover, an increased number of myeloid-derived suppressor cells (MDSCs) in comparison to healthy subjects has been observed in both MF and SS and a decrease in their numbers has been noted after successful therapy [67-69].

Treatment of CTCL depends on the stage of the disease and the general condition of the patient. Early disease stages of MF (IA-IIA) can be controlled with skin – directed therapy, such as topical steroids, light treatment and radiation [4, 70]. Systemic treatments, such as retinoids and IFN-α have been recommended as second-line treatment of MF stages IA, IB and IIA [71]. The 5-year survival for these patients is around 90%, significantly better compared with 30%–50% for advanced disease (IIB–IVB) [72]. Chemotherapy agents, such as monochemotherapy (gemcitabine, pegylated liposomal doxorubicine) and polychemotherapy, as well as newer systemic agents (brentuximab-vedotin and mogamulizumab) are recommended in advanced disease stage. Large cell transformation in histology, expression of CD30 and folliculotropic subtype of MF have been reported to have a more aggressive course and poorer prognosis. Although rare cases of CD4- CD8+ , CD4- CD8- , or CD4+CD8+ immunophenotypes have been described in MF, these differences in phenotype do not affected the prognosis of the disease [73]. However, a phenotypic switch from CD4+ to CD4- in the lesion has been suggested a portend of poor outcome [74].

SS is associated with a poor prognosis and overall survival rates varying from 7.5 to 22.4 months [75]. Apart from conventional treatments (e.g. extracorporeal photopheresis (ECP), light treatment (PUVA), retinoids, IFN-α, low dose methotrexate and polychemotherapy), small-molecule inhibitors e.g. histone deacetylase inhibitors (HDACi) and monoclonal antibodies (mAbs) are currently being explored as therapeutic options in SS [76, 77]. A comprehensive review on investigative drugs in CTCL has been published recently [78]. Despite their initial moderate to good response to most treatments, SS patients often develop resistance to treatment in the course of the disease [79]. Selection of more robust and aggressive tumor clones has been hypothesized as one possible driver for resistance; e.g. in a small cohort of SS patients, tumor cell subpopulations were heterogeneous and showed different sensitivity to HDACi [33]. Also, although no single common driver mutation has been identified, novel findings demonstrate aberrant functioning of cell division mechanisms in CTCL that lead to genetic instability and possible escape to treatment [80]. Further, the role of STAT3 activation in conferring resistance in both SS and advanced-stage MF has become clearer [81, 82]. And finally, SS cells have been shown to display resistance to apoptosis when chromatin remodeling is defect, such as in SNF5- and SATB1-deficient cells, or when Fas-mediated apoptosis is defect [83, 84]. If and how exactly these mechanisms contribute to treatment resistance in SS cell subpopulations has yet to be addressed in detail. Taken together, despite the numerous systemic treatment options available, long – term remission rates with conventional treatments alone are still low and allogeneic hematopoietic stem cell transplantation (alloHSCT) is currently the only curative option in advanced MF/SS [85, 86].

Thorough reviews analyzing the therapeutic options available for CTCL have been lastly identified in the literature [70, 85, 87, 88]. Herein, we summarize the recent advances in the immunotherapies of CTCL.

ECP and PUVA

Both extracorporeal photopheresis (ECP) and psoralen plus ultraviolet A (PUVA) consist of the systemic or local application of 8-metoxypsolaren and subsequent photoactivation resulting in apoptosis of malignant lymphocytes [89-92]. However, despite the established use in the management of CTCL, their mechanism of action is still not sufficiently investigated. While PUVA is applied to patients with earlier stages of CTCL and skin involvement only, ECP is used mainly in erythrodermic patients with blood involvement [93]. PUVA induces remission lasting up to 10 years in 72% of MF patients [2, 94] and is described to have an immunomodulatory effect i.e. a shift from Th2 to Th1 phenotype [95]. A recent study by Vieyra-Garcia et al. [96]shed new light on the mechanisms beyond PUVA efficacy in CTCL. The authors have demonstrated a switch from a Th2 with CCL18 overexpression, to a Th1 phenotype characterized by CXCL9, CXCL10, and CXCL11 expression. Moreover, they have shown that while in low-burden disease the efficacy of PUVA results in the reduction of the number of malignant T-cells, in high-burden disease it relies mostly on the recruitment of clonal cytotoxic T cells. The study identified the c-Kit+OX40L+CD40L+ DC as the major drivers of inflammation and clonal expansion of malignant T cells due to their pro-survival impact and suggested targeting c-Kit, OX40, and CD40 signaling may be a novel therapeutic option in the treatment of MF.

ECP induces response rates of approximately 60%, with complete responses of 14–26% [89, 97] that can be further improved when combined with other immunomodulatory agents, such as IFN-α or systemic retinoids [89, 98]. The immunomodulatory effect of ECP is thought to rely on the induction of monocyte differentiation to DC that are capable of phagocytosing and efficiently presenting tumor antigens [99, 100]. Moreover, a shift from Th2 bias to a Th1 proinflammatory phenotype has been reported [90]. Recently, there has been considerable interest in defining the role of NK cells in the success of ECP. Recent studies have reported a significant increase in the percentage of CD56+dim NK cells characterized by a high cytotoxic potential 3 months after the start of therapy [20]. Interestingly, Mundy-Bosse et al.[58] reported that increased NK cell cytotoxicity and higher NK-cell numbers before ECP were associated with decreased short-term survival.

Monoclonal Antibodies

Mogamulizumab (Anti-CCR4)

Mogamulizumab, a glycol-engineered mAb targeting chemokine receptor type 4 (CCR4) with an increased affinity to FcγRIIIa (CD16) and enhanced ADCC [101], was approved in August 2018 for patients with relapsed or refractory, advanced CTCL with at least 1 prior systemic therapy [63]. Apart from the clinical efficacy, patients treated with mogamulizumab reported an improvement in their quality of life, including skin pain and fatigue [63]. Mogamulizumab is generally well-tolerated; however some skin-related toxicities due to the induction of autoantibodies recognizing human keratinocytes or melanocytes that induce complement-dependent cytotoxicity have been reported [102]. Of note, targeting CCR4 may also result in the depletion of nonmalignant Tregs, thus leading to or aggravating autoimmune disorders [103]. In this way, it also increases the risk of graft-versus-host disease following allogeneic bone marrow transplantation [104], which should be therefore delayed by at least 50 days from the administration of the last dose [105].

Immune Checkpoint Inhibitors

Based on the increased response rates associated with targeting of negative immune regulators through mAbs, also known as immune checkpoints inhibitors (ICI), in a wide spectrum of malignancies, including melanoma, there is considerable interest in applying these targets in the management of CTCL [87]. The particularity of CTCL in the context of implementation of ICI relies on the fact that the tumor itself arises from CD4+ T cells, a population of lymphocytes responsible for priming of the cytotoxic response. Increasing evidence suggests that in CTCL both CD4+ and CD8+ cells have characteristics of immune exhaustion [53, 106, 107], and therefore targeting immune checkpoints would have implications on the functionality of both helper and cytotoxic T cells. One immune-inhibitory axis is the programmed death (PD-)1 axis, binding to its ligands PD-L1/L2. PD-1 expression has been shown to be high in the blood and skin of SS patients [108, 109] and has already been proposed as a factor responsible for drug resistance in SS [110]. A phase 2 clinical trial of the anti-PD1 mAb pembrolizumab in heavily pretreated advanced-stage MF and SS patients reported an ORR of 38%, with a 1-year progression-free survival of 69% and a duration of response of 64 months [111, 112]. Moreover, there are also ongoing trials targeting PD-L1, using anti-PD-L1 mAbs, i.e., atezolizumab (NCT03357224) and durvalumab (NCT03011814).

A recent analysis investigating PD-1 expression in SS showed a high PD-1 expression on tumor T cells compared to nontumor CD4+ T cells from SS patients or to normal CD4+ cells from healthy individuals [113]. In contrast, PD-L1 showed a decreased expression on tumor T cells, while PD-L2 expression is low and did not show any significant differences between groups. This is in line with other studies [114], where PD-L1 showed a high expression in the tumor environment, particularly in monocyte-derived compartments, where it was expressed by 73% of cells. Also a recent study by Querfeld et al. [53] showed high PD-L1 levels in DC émigrés from the skin but a low expression by T cells themselves.

While PD-1/PD-L1 ICI have gained much interest in the therapy of CTCL, much less is known about the expression of other immune receptors. In an analysis of CTCL skin samples, Querfeld et al. [53] observed a higher expression of CTLA-4 on both CD4+ and CD8+ T cells. In another study no significant differences were found in CTLA-4 expression in CD4+ malignant versus bystander T cells in SS patients and healthy controls [115]. However, as the combination of anti-PD-1 (nivolumab) with anti-CTLA-4 showed no benefit compared to nivolumab alone [116], there are no active or recruiting clinical studies testing the efficacy of CTLA-4 targeting in CTCL. A recent analysis [115] of a panel of checkpoint inhibitors in a small cohort of SS patients revealed a significant upregulation of FRCL3 and TIGIT expression, which is in line with the previous reports [117, 118], together with a reduced expression of LAG-3 on CD4+ tumor cells. As several advanced clinical studies address TIGIT as a target molecule, it may also be of interest in CTCL. Interestingly Querfeld et al. [53] observed increased LAG-3 expression in lesional MF skin samples, which may be explained by the fact that MF and SS arise from distinct T-cell subsets [7]. Further, a hitherto nondemonstrated overexpression of BTLA on CD4+ cells in SS has been reported [115]. This is of possible interest, as BTLA blockade would arrest T-cell proliferation and can be therapeutically targeted by a specific fragment (HVEM 26–28) [119]. However, no in vivo studies have been conducted so far. As immune checkpoints have nonredundant roles in cancer progression, other emerging regulatory molecules (TIM-3, CD96 [TACTILE], and PD-1H [VISTA]) may be of interest in CTCL, too.

CD47

CD47 is highly expressed on Sézary cells in the peripheral blood and skin and correlates with a worse overall survival [120]. Inhibiting the binding of CD47 to its ligand, SIRPa activates both innate and adaptive antitumor responses by promoting phagocytosis and subsequent activation of CD8+ T cells [reviewed in 121]. Targeting CD47 with TTI-621 not only blocks the “do-not-eat-me signal” of CD47 but it also enhances phagocytosis of tumor cells by monocytes. The data gained in 2 phase I trials suggest its satisfactory activity combined with a good safety profile [120, 122].

Alemtuzumab (Anti-CD52)

Alemtuzumab is a monoclonal Ab directed against the surface glycoprotein CD52. CD52 expression on malignant T cells has been shown in 14 out of 16 CTCL cases by flow cytometry [123]. In several clinical studies, alemtuzumab appears to reach better responses in Sézary patients, with an ORR of 81%, as compared to MF patients, with an ORR of 29%[124]. A retrospective study of 39 advanced CTCL patients also showed long-term remission in Sézary patients but not in MF patients [65]. The treatment is associated with a high toxicity, but this can be lowered by subcutaneous administration (as compared to intravenous) and lower doses of the antibody. Despite a good response rate in Sézary patients, it is not approved anymore for CTCL treatment. Currently, there is an ongoing phase I study using a combination treatment of IL-15 and alemtuzumab in different T-cell leukemia and lymphoma conditions (NCT02689453).

KIR3DL2 Targeting

KIR3DL2, also called CD158k, is overexpressed by transformed advanced MF and SS cells correlating with the disease stage and large cell transformation [125] as well as a shorter survival [126]. IPH4102, an anti-KIR2DL2 humanized IgG1 mAb, effectively induces ADCC and immunophagocytosis [127], delays tumor growth, and improved the overall survival in a xenograft mouse model. The results of a phase I study in MF and SS patients (NCT02593045) demonstrated a confirmed global overall response in 16 of 44 patients (36.4%; 95% CI 23.8–51.1), and, of those, 15 responses were observed in 35 patients with SS (43%; 95% CI 28.0–59.1) [128]. Moreover, a phase II trial of IPH4102 alone or in combination with chemotherapy in patients with advanced T-cell lymphoma (TELLOMAK) (NCT03902184) is currently recruiting.

Chimeric Antigen Receptor-Based Therapies

Given their success in the treatment of B-cell malignancies, chimeric antigen receptor (CAR)-modified lymphocytes raise interest as a therapeutic option in CTCL. However, their use poses some considerable issues. First of all, there is a lack of specific targets expressed uniquely on malignant T cells. This results in 2 different concerns. One is that, when using a specific target for a tumor (sub)population, not all malignant cells are affected by the treatment and relapse of the disease occurs. The other is that, when using a more broadly expressed marker, targeting also of healthy T cells can occur, which can lead to life-threatening T-cell aplasia. While the first concern is difficult to solve, unless a unique marker expressed by all malignant cells is discovered, the second concern could be overcome by a transient CAR expression or expression of suicide genes allowing for reconstitution of T cells. By now CD4, CD5, CD7, CD30, CD37, CCR4, and TCR β chains (TRBC1/TRBC2) have been tested as possible targets of CAR-based therapies [reviewed in 129]. An alternative to targeting T cells is the application of CAR-transduced NK cells. This may offer an alternative as it eliminates the phenomenon of fratricide, i.e., mutual killing of CAR-expressing cells due to shared T-cell antigens [129].

Cytokines

Interferons

Next to IFN-α, which is an approved and recommended treatment for MF and SS [130], IFN-γ has emerged as a novel option for the patients who have failed IFN-α. IFN-γ abrogates the Th1/Th2 bias inducing a Th1-dominated tumor microenvironment and stimulates macrophages, DC, and cytotoxicity, mediated by CD8 T cells and NK cells [131]. As demonstrated by a small-scale study by Kaplan et al. [132], who treated 16 MF and SS patients with IFN-γ, 31% of patients had an objective partial response. None of the patients showed a complete response. A subsequent small-dose-escalating study by Dummer et al.[133] demonstrated the therapeutic effect of intratumoral injections of TG1042 (a third-generation, nonreplicating human adenovirus vector containing a human IFN-γ cDNA insert). The observations included 9 patients (CTCL, n = 7; CBCL, n = 2) injected with the following doses of TG1042: 3 × 109, 3 × 1010, and 3 × 1011 total particles. A local clinical response was observed in 5 of the 9 treated patients (3 patients with a complete response and 2 patients with a partial response). Three patients reported a complete systemic response, represented by clearance of noninjected skin lesions. The duration of the clinical response was on average 3 months (range 1–6 months) and only grade 1 and 2 adverse events were reported [133].

Interleukin-12

IL-12 is a Th1-promoting cytokine produced by the antigen-presenting cells. It is a potent inducer of IFN-γ. IL-12 boosts NK cell activity and cytotoxic T-cell responses in CTCL [134]. As discussed before, a Th2-dominant cytokine milieu and a reduced production of Th1-inducing cytokines such as IFN-γ and IL-12 are a hallmark of immune evasion in advanced MF and SS [135]. In vitro data have shown increased lysis of malignant cells derived from SS patients when cultured with IL-12, providing additional evidence of the role of cytokines in antitumor immune function [134]. The therapeutic effect of IL-12 administration was tested in a small-scale (n = 10) phase I trial using 50–300 ng/kg IL-12. The study cohort included patients with MF and SS, stages T1 to T4. Unfortunately, the observations in SS patients were scarce because 2 of the 3 SS patients withdrew from the treatment. Overall, 20% of the patients showed a complete response, 20% showed a partial response, and the remaining patients had no response or a local response.

TLR Agonists

TLR are key players in innate immunity. TLR are predominantly expressed by DC and macrophages and recognize pathogen antigens in the microenvironment [136]. Once bound to their ligand, they induce antigen presentation and cell proliferation and cause surface upregulation of costimulatory molecules on the antigen-presenting cells, leading to T cell activation and increased cytotoxicity [137]. The therapeutic targets addressed in CTCL are TLR7 and TLR8, which bind viral RNA particles, and TLR9, which binds bacterial and viral DNA.

Imiquimod (TLR 7 Agonist)

TLR7 is expressed on plasmacytoid DC (pDC) [138]. Targeting TLR7 with a synthetic ligand, i.e., imiquimod, induces the production of proinflammatory cytokines, i.e. IFN-α, which induces a Th1 type immune response and shift towards cell-mediated immunity. There are several case reports and case series describing the efficacy of imiquimod treatment in CTCL patients [139-145]. Summarized results of the studies demonstrate that 71% of the patients have a complete response and 17% stable disease. Despite the promising results, imiquimod is not approved for use in CTCL because larger trials are necessary to evaluate its efficacy and safety in MF and SS.

Resiquimod (TLR7 and TLR8 Agonist)

Resiquimod binds to both TLR7 and TLR8. Despite their structural similarity, the 2 receptors differ in their signaling, and their activation leads to secretion of different cytokines. The TLR7 agonist activates predominantly pDC, inducing the production of IFN-α and IFN-regulated chemokines such as IFN-inducible protein and IFN-inducible T-cell α chemoattractant. The TLR8 agonist activates mDC, monocytes, and MDC to secrete TNF-α, IL-12, and MIP-1α [146]. In summary, TLR7 activation results in a 5–10 times greater production of IFN-α, whereas TLR7/8 stimulation increases TNF-α and IL-12 levels approximately 10 times. A phase I clinical trial with 12 early-stage patients (from IA to IIA MF) illustrated a 17% complete response rate and a 75% partial response rate in patients. Moreover, the dose escalation correlates with a better response [147]. The elevated after-treatment levels of TNF-α, IL-12, and IFN-γ induce the cytotoxic function of CD8 and NK cells and partial or complete reduction of the clonal malignant T cells in skin biopsies and correlate positively with treatment response.

TLR9 Agonist

TLR9 is an intracellular receptor expressed on numerous immune cells and it is activated by unmethylated CpG sequences found in bacterial or viral DNA. Its binding initiates the release of proinflammatory cytokines such as type I IFN and IL-12 [148]. In a phase 1/2 study by Kim et al. [149] 15 MF patients received intratumoral injections with TLR9 agonist CpG oligodeoxynucleotides combined with localized radiation. The combination of TLR9 agonist treatment and radiation was chosen because local radiation increases the number of available tumor antigens for pDC, which in parallel are primed by unmethylated CpG sequences. The overall response rate is approximately 36% with a median response duration of 7 weeks.

Antibiotics

A diminished diversity of skin microbiome with a tendency toward increased colonization with SA has been reportedin both MF and SS [150, 151]. It has been suggested that SA enterotoxins stimulate a reciprocal cross-talk between nonmalignant and malignant T cells resulting in IL-2-dependent proliferation of the malignant clone [152] and that interaction of T cells with the bacterial microenvironment may accelerate disease progression by promoting STAT signaling [153]. Thus, microbiome targeting may be a therapeutic strategy. Indeed, a recent small clinical study by Lindahl et al. [154] demonstrated that a short-term aggressive treatment with a SA-targeting antibiotic regimen resulted in a marked long-lasting clinical improvement in advanced-stage CTCL patients [154], leading to a decrease in the proliferation of malignant cells, STAT3 signaling, and the expression of CD25. These observations encourage the conduction of studies combining the targeting of SA together with STAT signaling [155].

CTCL is a rare skin lymphoma arising from malignant T-cell homing to or sessile in the skin. Both intrinsic and extrinsic factors may play a critical role in the pathophysiology of CTCL and cutting-edge research is currently focused on how increased levels and/or overactivation of key molecules, such as STAT3, GATA3, CCR4, and KIR3DL2, contribute to the initiation and progression of the disease. The extensive research performed in recent years has greatly advanced our understanding of CTCL and its impact on patient care and quality of life.

Naturally occurring molecules and genes, involved directly in or acting as surrogate parameters for active pathophysiological disease processes, if easily detectable, are convenient for use as biomarkers, disease-classifying molecules, or treatment targets. However, the heterogeneity of CTCL renders the detection of common markers and/or therapeutic targets difficult. Novel technologies and large-scale data generation combined with analytical approaches supported by artificial intelligence are expected to deliver the necessary knowledge and allow personalized medicine for the domain of CTCL.

The rarity of the diseases of the CTCL group necessitates an international collaborative effort to successfully accomplish high-evidence clinical research. In the last few years, the set-up has been established and successfully applied for at least 2 large prospective randomized clinical trials.

F.D. has received intermittent travel support from Pierre Fabre outside of this work. E.G. has intermittent, project-focused consulting and/or advisory relationships with Mallinckrodt, Takeda, Helsinn, Scaylite, and Novartis outside of this work. M.B., C.F., D.I., and Y.-T.C. declare no conflict of interests.

This project was supported by the Jubiläumsstiftung von SwissLife, the Promedica Stiftung (1406/M and 1412/M), a further anonymous foundation, the Swiss Cancer Research Foundation (KFS-4243-08-2017), the Clinical Research Priority Program (CRPP) of the University of Zurich, the Swiss National Science Foundation (PMPDP3_151326), and the European Academy of Dermatology and Venereology (PPRC-2019–20). The funders had no role in the study design, data collection and analysis, the decision to publish, or the preparation of this paper.

All of the authors searched for and collected literature data and participated in the writing and approved the final version of this paper.

1.
Kempf
W
,
Zimmermann
AK
,
Mitteldorf
C
.
Cutaneous lymphomas-An update 2019
.
Hematol Oncol
.
2019
Jun
;
37
(
S1
Suppl 1
):
43
7
.
[PubMed]
0278-0232
2.
Willemze
R
,
Cerroni
L
,
Kempf
W
,
Berti
E
,
Facchetti
F
,
Swerdlow
SH
, et al.
The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas
.
Blood
.
2019
Apr
;
133
(
16
):
1703
14
.
[PubMed]
0006-4971
3.
Ferenczi
K
,
Makkar
HS
.
Cutaneous lymphoma: kids are not just little people
.
Clin Dermatol
.
2016
Nov - Dec
;
34
(
6
):
749
59
.
[PubMed]
0738-081X
4.
Wilcox
RA
.
Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management
.
Am J Hematol
.
2017
Oct
;
92
(
10
):
1085
102
.
[PubMed]
0361-8609
5.
Willemze
R
,
Jaffe
ES
,
Burg
G
,
Cerroni
L
,
Berti
E
,
Swerdlow
SH
, et al.
WHO-EORTC classification for cutaneous lymphomas
.
Blood
.
2005
May
;
105
(
10
):
3768
85
.
[PubMed]
0006-4971
6.
Geller
S
,
Myskowski
PL
,
Pulitzer
M
,
Horwitz
SM
,
Moskowitz
AJ
.
Cutaneous T-cell lymphoma (CTCL), rare subtypes: five case presentations and review of the literature
.
Linchuang Zhongliuxue Zazhi
.
2019
Feb
;
8
(
1
):
5
.
[PubMed]
1009-0460
7.
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
Aug
;
116
(
5
):
767
71
.
[PubMed]
0006-4971
8.
Larocca
C
,
Kupper
T
.
Mycosis Fungoides and Sézary Syndrome: an Update
.
Hematol Oncol Clin North Am
.
2019
Feb
;
33
(
1
):
103
20
.
[PubMed]
0889-8588
9.
Serrano
L
,
Martinez-Escala
ME
,
Zhou
XA
,
Guitart
J
.
Pruritus in Cutaneous T-Cell Lymphoma and Its Management
.
Dermatol Clin
.
2018
Jul
;
36
(
3
):
245
58
.
[PubMed]
0733-8635
10.
Demierre
MF
,
Gan
S
,
Jones
J
,
Miller
DR
.
Significant impact of cutaneous T-cell lymphoma on patients’ quality of life: results of a 2005 National Cutaneous Lymphoma Foundation Survey
.
Cancer
.
2006
Nov
;
107
(
10
):
2504
11
.
[PubMed]
0008-543X
11.
Scarisbrick
JJ
,
Quaglino
P
,
Prince
HM
,
Papadavid
E
,
Hodak
E
,
Bagot
M
, et al.
The PROCLIPI international registry of early-stage mycosis fungoides identifies substantial diagnostic delay in most patients
.
Br J Dermatol
.
2019
Aug
;
181
(
2
):
350
7
.
[PubMed]
0007-0963
12.
Mourad
A
,
Gniadecki
R
.
Overall Survival in Mycosis Fungoides: A Systematic Review and Meta-Analysis
.
J Invest Dermatol
.
2020
Feb
;
140
(
2
):
495
497.e5
.
[PubMed]
0022-202X
13.
Talpur
R
,
Singh
L
,
Daulat
S
,
Liu
P
,
Seyfer
S
,
Trynosky
T
, et al.
Long-term outcomes of 1,263 patients with mycosis fungoides and Sézary syndrome from 1982 to 2009
.
Clin Cancer Res
.
2012
Sep
;
18
(
18
):
5051
60
.
[PubMed]
1078-0432
14.
Mangold
AR
,
Thompson
AK
,
Davis
MD
,
Saulite
I
,
Cozzio
A
,
Guenova
E
, et al.
Early clinical manifestations of Sézary syndrome: A multicenter retrospective cohort study
.
J Am Acad Dermatol
.
2017
Oct
;
77
(
4
):
719
27
.
[PubMed]
0190-9622
15.
Whittemore
AS
,
Holly
EA
,
Lee
IM
,
Abel
EA
,
Adams
RM
,
Nickoloff
BJ
, et al.
Mycosis fungoides in relation to environmental exposures and immune response: a case-control study
.
J Natl Cancer Inst
.
1989
Oct
;
81
(
20
):
1560
7
.
[PubMed]
0027-8874
16.
Netchiporouk
E
,
Gantchev
J
,
Tsang
M
,
Thibault
P
,
Watters
AK
,
Hughes
JM
, et al.
Analysis of CTCL cell lines reveals important differences between mycosis fungoides/Sézary syndrome vs. HTLV-1+ leukemic cell lines
.
Oncotarget
.
2017
Oct
;
8
(
56
):
95981
98
.
[PubMed]
1949-2553
17.
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
Feb
;
119
(
4
):
825
31
.
[PubMed]
0008-543X
18.
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
Sep
;
107
(
3
):
373
6
.
[PubMed]
0022-202X
19.
Choi
J
,
Goh
G
,
Walradt
T
,
Hong
BS
,
Bunick
CG
,
Chen
K
, et al.
Genomic landscape of cutaneous T cell lymphoma
.
Nat Genet
.
2015
Sep
;
47
(
9
):
1011
9
.
[PubMed]
1061-4036
20.
da Silva Almeida
AC
,
Abate
F
,
Khiabanian
H
,
Martinez-Escala
E
,
Guitart
J
,
Tensen
CP
, et al.
The mutational landscape of cutaneous T cell lymphoma and Sézary syndrome
.
Nat Genet
.
2015
Dec
;
47
(
12
):
1465
70
.
[PubMed]
1061-4036
21.
Park
J
,
Yang
J
,
Wenzel
AT
,
Ramachandran
A
,
Lee
WJ
,
Daniels
JC
, et al.
Genomic analysis of 220 CTCLs identifies a novel recurrent gain-of-function alteration in RLTPR (p.Q575E)
.
Blood
.
2017
Sep
;
130
(
12
):
1430
40
.
[PubMed]
0006-4971
22.
Wang
L
,
Ni
X
,
Covington
KR
,
Yang
BY
,
Shiu
J
,
Zhang
X
, et al.
Genomic profiling of Sézary syndrome identifies alterations of key T cell signaling and differentiation genes
.
Nat Genet
.
2015
Dec
;
47
(
12
):
1426
34
.
[PubMed]
1061-4036
23.
McGirt
LY
,
Jia
P
,
Baerenwald
DA
,
Duszynski
RJ
,
Dahlman
KB
,
Zic
JA
, et al.
Whole-genome sequencing reveals oncogenic mutations in mycosis fungoides
.
Blood
.
2015
Jul
;
126
(
4
):
508
19
.
[PubMed]
0006-4971
24.
Ungewickell
A
,
Bhaduri
A
,
Rios
E
,
Reuter
J
,
Lee
CS
,
Mah
A
, et al.
Genomic analysis of mycosis fungoides and Sézary syndrome identifies recurrent alterations in TNFR2
.
Nat Genet
.
2015
Sep
;
47
(
9
):
1056
60
.
[PubMed]
1061-4036
25.
Woollard
WJ
,
Pullabhatla
V
,
Lorenc
A
,
Patel
VM
,
Butler
RM
,
Bayega
A
, et al.
Candidate driver genes involved in genome maintenance and DNA repair in Sézary syndrome
.
Blood
.
2016
Jun
;
127
(
26
):
3387
97
.
[PubMed]
0006-4971
26.
Guenova
E
,
Metzler
G
,
Hoetzenecker
W
,
Berneburg
M
,
Rocken
M
.
Classic Mediterranean Kaposi’s sarcoma regression with sirolimus treatment
.
Arch Dermatol
.
2008
May
;
144
(
5
):
692
3
.
[PubMed]
0003-987X
27.
Blümel
E
,
Munir Ahmad
S
,
Nastasi
C
,
Willerslev-Olsen
A
,
Gluud
M
,
Fredholm
S
, et al.
Staphylococcus aureus alpha-toxin inhibits CD8+ T cell-mediated killing of cancer cells in cutaneous T-cell lymphoma
.
OncoImmunology
.
2020
Apr
;
9
(
1
):
1751561
.
[PubMed]
2162-4011
28.
Ghazawi
FM
,
Alghazawi
N
,
Le
M
,
Netchiporouk
E
,
Glassman
SJ
,
Sasseville
D
, et al.
Environmental and Other Extrinsic Risk Factors Contributing to the Pathogenesis of Cutaneous T Cell Lymphoma (CTCL)
.
Front Oncol
.
2019
Apr
;
9
:
300
.
[PubMed]
2234-943X
29.
Clark
RA
.
Skin-resident T cells: the ups and downs of on site immunity
.
J Invest Dermatol
.
2010
Feb
;
130
(
2
):
362
70
.
[PubMed]
0022-202X
30.
van Doorn
R
,
van Kester
MS
,
Dijkman
R
,
Vermeer
MH
,
Mulder
AA
,
Szuhai
K
, et al.
Oncogenomic analysis of mycosis fungoides reveals major differences with Sezary syndrome
.
Blood
.
2009
Jan
;
113
(
1
):
127
36
.
[PubMed]
0006-4971
31.
Laharanne
E
,
Oumouhou
N
,
Bonnet
F
,
Carlotti
M
,
Gentil
C
,
Chevret
E
, et al.
Genome-wide analysis of cutaneous T-cell lymphomas identifies three clinically relevant classes
.
J Invest Dermatol
.
2010
Jun
;
130
(
6
):
1707
18
.
[PubMed]
0022-202X
32.
Roelens
M
,
Delord
M
,
Ram-Wolff
C
,
Marie-Cardine
A
,
Alberdi
A
,
Maki
G
, et al.
Circulating and skin-derived Sézary cells: clonal but with phenotypic plasticity
.
Blood
.
2017
Sep
;
130
(
12
):
1468
71
.
[PubMed]
0006-4971
33.
Buus
TB
,
Willerslev-Olsen
A
,
Fredholm
S
,
Blümel
E
,
Nastasi
C
,
Gluud
M
, et al.
Single-cell heterogeneity in Sézary syndrome
.
Blood Adv
.
2018
Aug
;
2
(
16
):
2115
26
.
[PubMed]
2473-9529
34.
Borcherding
N
,
Voigt
AP
,
Liu
V
,
Link
BK
,
Zhang
W
,
Jabbari
A
.
Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression
.
Clin Cancer Res
.
2019
May
;
25
(
10
):
2996
3005
.
[PubMed]
1078-0432
35.
Heid
JB
,
Schmidt
A
,
Oberle
N
,
Goerdt
S
,
Krammer
PH
,
Suri-Payer
E
, et al.
FOXP3+CD25- tumor cells with regulatory function in Sézary syndrome
.
J Invest Dermatol
.
2009
Dec
;
129
(
12
):
2875
85
.
[PubMed]
0022-202X
36.
Tiemessen
MM
,
Mitchell
TJ
,
Hendry
L
,
Whittaker
SJ
,
Taams
LS
,
John
S
.
Lack of suppressive CD4+CD25+FOXP3+ T cells in advanced stages of primary cutaneous T-cell lymphoma
.
J Invest Dermatol
.
2006
Oct
;
126
(
10
):
2217
23
.
[PubMed]
0022-202X
37.
Gjerdrum
LM
,
Woetmann
A
,
Odum
N
,
Burton
CM
,
Rossen
K
,
Skovgaard
GL
, et al.
FOXP3+ regulatory T cells in cutaneous T-cell lymphomas: association with disease stage and survival
.
Leukemia
.
2007
Dec
;
21
(
12
):
2512
8
.
[PubMed]
0887-6924
38.
Berger
CL
,
Tigelaar
R
,
Cohen
J
,
Mariwalla
K
,
Trinh
J
,
Wang
N
, et al.
Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells
.
Blood
.
2005
Feb
;
105
(
4
):
1640
7
.
[PubMed]
0006-4971
39.
Komatsu
N
,
Mariotti-Ferrandiz
ME
,
Wang
Y
,
Malissen
B
,
Waldmann
H
,
Hori
S
.
Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity
.
Proc Natl Acad Sci USA
.
2009
Feb
;
106
(
6
):
1903
8
.
[PubMed]
0027-8424
40.
Qu
K
,
Zaba
LC
,
Satpathy
AT
,
Giresi
PG
,
Li
R
,
Jin
Y
, et al.
Chromatin Accessibility Landscape of Cutaneous T Cell Lymphoma and Dynamic Response to HDAC Inhibitors. Cancer Cell.
2017
;32(1):27-41 e4.
41.
Guenova
E
,
Watanabe
R
,
Teague
JE
,
Desimone
JA
,
Jiang
Y
,
Dowlatshahi
M
, et al.
TH2 cytokines from malignant cells suppress TH1 responses and enforce a global TH2 bias in leukemic cutaneous T-cell lymphoma
.
Clin Cancer Res
.
2013
Jul
;
19
(
14
):
3755
63
.
[PubMed]
1078-0432
42.
Wood
GS
,
Edinger
A
,
Hoppe
RT
,
Warnke
RA
.
Mycosis fungoides skin lesions contain CD8+ tumor-infiltrating lymphocytes expressing an activated, MHC-restricted cytotoxic T-lymphocyte phenotype
.
J Cutan Pathol
.
1994
Apr
;
21
(
2
):
151
6
.
[PubMed]
0303-6987
43.
Asadullah
K
,
Friedrich
M
,
Döcke
WD
,
Jahn
S
,
Volk
HD
,
Sterry
W
.
Enhanced expression of T-cell activation and natural killer cell antigens indicates systemic anti-tumor response in early primary cutaneous T-cell lymphoma
.
J Invest Dermatol
.
1997
May
;
108
(
5
):
743
7
.
[PubMed]
0022-202X
44.
Bagot
M
,
Echchakir
H
,
Mami-Chouaib
F
,
Delfau-Larue
MH
,
Charue
D
,
Bernheim
A
, et al.
Isolation of tumor-specific cytotoxic CD4+ and CD4+CD8dim+ T-cell clones infiltrating a cutaneous T-cell lymphoma
.
Blood
.
1998
Jun
;
91
(
11
):
4331
41
.
[PubMed]
0006-4971
45.
Echchakir
H
,
Bagot
M
,
Dorothée
G
,
Martinvalet
D
,
Le Gouvello
S
,
Boumsell
L
, et al.
Cutaneous T cell lymphoma reactive CD4+ cytotoxic T lymphocyte clones display a Th1 cytokine profile and use a fas-independent pathway for specific tumor cell lysis
.
J Invest Dermatol
.
2000
Jul
;
115
(
1
):
74
80
.
[PubMed]
0022-202X
46.
Hsi
AC
,
Lee
SJ
,
Rosman
IS
,
Carson
KR
,
Kelley
A
,
Viele
V
, et al.
Expression of helper T cell master regulators in inflammatory dermatoses and primary cutaneous T-cell lymphomas: diagnostic implications
.
J Am Acad Dermatol
.
2015
Jan
;
72
(
1
):
159
67
.
[PubMed]
0190-9622
47.
Showe
LC
,
Fox
FE
,
Williams
D
,
Au
K
,
Niu
Z
,
Rook
AH
. Depressed IL-12-mediated signal transduction in T cells from patients with Sezary syndrome is associated with the absence of IL-12 receptor beta 2 mRNA and highly reduced levels of STAT4. Journal of immunology (Baltimore, Md : 1950).
1999
;163(7):4073-9.
48.
Vowels
BR
,
Lessin
SR
,
Cassin
M
,
Jaworsky
C
,
Benoit
B
,
Wolfe
JT
, et al.
Th2 cytokine mRNA expression in skin in cutaneous T-cell lymphoma
.
J Invest Dermatol
.
1994
Nov
;
103
(
5
):
669
73
.
[PubMed]
0022-202X
49.
Netchiporouk
E
,
Litvinov
IV
,
Moreau
L
,
Gilbert
M
,
Sasseville
D
,
Duvic
M
.
Deregulation in STAT signaling is important for cutaneous T-cell lymphoma (CTCL) pathogenesis and cancer progression
.
Cell Cycle
.
2014
;
13
(
21
):
3331
5
.
[PubMed]
1538-4101
50.
Qin
JZ
,
Kamarashev
J
,
Zhang
CL
,
Dummer
R
,
Burg
G
,
Döbbeling
U
.
Constitutive and interleukin-7- and interleukin-15-stimulated DNA binding of STAT and novel factors in cutaneous T cell lymphoma cells
.
J Invest Dermatol
.
2001
Sep
;
117
(
3
):
583
9
.
[PubMed]
0022-202X
51.
Litvinov
IV
,
Pehr
K
,
Sasseville
D
.
Connecting the dots in cutaneous T cell lymphoma (CTCL): STAT5 regulates malignant T cell proliferation via miR-155
.
Cell Cycle
.
2013
Jul
;
12
(
14
):
2172
3
.
[PubMed]
1538-4101
52.
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
Jun
;
62
(
6
):
995
1004
.
[PubMed]
0190-9622
53.
Querfeld
C
,
Leung
S
,
Myskowski
PL
,
Curran
SA
,
Goldman
DA
,
Heller
G
, et al.
Primary T Cells from Cutaneous T-cell Lymphoma Skin Explants Display an Exhausted Immune Checkpoint Profile
.
Cancer Immunol Res
.
2018
Aug
;
6
(
8
):
900
9
.
[PubMed]
2326-6066
54.
Wysocka
M
,
Zaki
MH
,
French
LE
,
Chehimi
J
,
Shapiro
M
,
Everetts
SE
, et al.
Sézary 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
Nov
;
100
(
9
):
3287
94
.
[PubMed]
0006-4971
55.
Rook
AH
,
Heald
P
.
The immunopathogenesis of cutaneous T-cell lymphoma
.
Hematol Oncol Clin North Am
.
1995
Oct
;
9
(
5
):
997
1010
.
[PubMed]
0889-8588
56.
Scala
E
,
Abeni
D
,
Palazzo
P
,
Liso
M
,
Pomponi
D
,
Lombardo
G
, et al.
Specific IgE toward allergenic molecules is a new prognostic marker in patients with Sézary syndrome
.
Int Arch Allergy Immunol
.
2012
;
157
(
2
):
159
67
.
[PubMed]
1018-2438
57.
Bouaziz
JD
,
Ortonne
N
,
Giustiniani
J
,
Schiavon
V
,
Huet
D
,
Bagot
M
, et al.
Circulating natural killer lymphocytes are potential cytotoxic effectors against autologous malignant cells in sezary syndrome patients
.
J Invest Dermatol
.
2005
Dec
;
125
(
6
):
1273
8
.
[PubMed]
0022-202X
58.
Mundy-Bosse
B
,
Denlinger
N
,
McLaughlin
E
,
Chakravarti
N
,
Hwang
S
,
Chen
L
, et al.
Highly cytotoxic natural killer cells are associated with poor prognosis in patients with cutaneous T-cell lymphoma
.
Blood Adv
.
2018
Aug
;
2
(
15
):
1818
27
.
[PubMed]
2473-9529
59.
ManfrereC
KC
,
Torrealba
MP
,
Miyashiro
DR
,
Pereira
NZ
,
Yoshikawa
FS
,
de M Oliveira
L
, et al.
Profile of differentially expressed Toll-like receptor signaling genes in the natural killer cells of patients with Sézary syndrome
.
Oncotarget
.
2017
Sep
;
8
(
54
):
92183
94
.
[PubMed]
1949-2553
60.
Mack
MR
,
Brestoff
JR
,
Niu
H
,
Whelan
TM
,
Oetjen
LK
,
Bodet
ND
, et al.
Natural killer cell dysregulation underlies atopic dermatitis.
The Journal of Immunology.
2018
;200(1 Supplement):45.37-45.37.
61.
Batista
MD
,
Ho
EL
,
Kuebler
PJ
,
Milush
JM
,
Lanier
LL
,
Kallas
EG
, et al.
Skewed distribution of natural killer cells in psoriasis skin lesions
.
Exp Dermatol
.
2013
Jan
;
22
(
1
):
64
6
.
[PubMed]
0906-6705
62.
von Bubnoff
D
,
Andrès
E
,
Hentges
F
,
Bieber
T
,
Michel
T
,
Zimmer
J
.
Natural killer cells in atopic and autoimmune diseases of the skin
.
J Allergy Clin Immunol
.
2010
Jan
;
125
(
1
):
60
8
.
[PubMed]
0091-6749
63.
Kim
YH
,
Bagot
M
,
Pinter-Brown
L
,
Rook
AH
,
Porcu
P
,
Horwitz
SM
, et al.;
MAVORIC Investigators
.
Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): an international, open-label, randomised, controlled phase 3 trial
.
Lancet Oncol
.
2018
Sep
;
19
(
9
):
1192
204
.
[PubMed]
1470-2045
64.
Clark
RA
,
Watanabe
R
,
Teague
JE
,
Schlapbach
C
,
Tawa
MC
,
Adams
N
, et al.
Skin effector memory T cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients
.
Sci Transl Med
.
2012
Jan
;
4
(
117
):
117ra7
.
[PubMed]
1946-6234
65.
de Masson
A
,
Guitera
P
,
Brice
P
,
Moulonguet
I
,
Mouly
F
,
Bouaziz
JD
, et al.
Long-term efficacy and safety of alemtuzumab in advanced primary cutaneous T-cell lymphomas
.
Br J Dermatol
.
2014
Mar
;
170
(
3
):
720
4
.
[PubMed]
0007-0963
66.
Goddard
DS
,
Yamanaka
K
,
Kupper
TS
,
Jones
DA
.
Activation of neutrophils in cutaneous T-cell lymphoma
.
Clin Cancer Res
.
2005
Dec
;
11
(
23
):
8243
9
.
[PubMed]
1078-0432
67.
Hergott
CB
,
Dudley
G
,
Dorfman
DM
.
Circulating Myeloid-Derived Suppressor Cells Reflect Mycosis Fungoides/Sezary Syndrome Disease Stage and Response to Treatment
.
Blood
.
2018
;
132
Suppl 1
:
4127
. 0006-4971
68.
Pileri
A
,
Agostinelli
C
,
Quaglino
P
,
Patrizi
A
,
Pimpinelli
N
.
The role of myeloid derived suppressor cells in mycosis fungoides
.
Cancer Immunol Immunother
.
2018
Jul
;
67
(
7
):
1175
6
.
[PubMed]
0340-7004
69.
Geskin
LJ
,
Akilov
OE
,
Kwon
S
,
Schowalter
M
,
Watkins
S
,
Whiteside
TL
, et al.
Therapeutic reduction of cell-mediated immunosuppression in mycosis fungoides and Sézary syndrome
.
Cancer Immunol Immunother
.
2018
Mar
;
67
(
3
):
423
34
.
[PubMed]
0340-7004
70.
Lovgren
ML
,
Scarisbrick
JJ
.
Update on skin directed therapies in mycosis fungoides
.
Linchuang Zhongliuxue Zazhi
.
2019
Feb
;
8
(
1
):
7
.
[PubMed]
1009-0460
71.
Trautinger
F
,
Eder
J
,
Assaf
C
,
Bagot
M
,
Cozzio
A
,
Dummer
R
, et al.
European Organisation for Research and Treatment of Cancer consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome - Update 2017
.
Eur J Cancer
.
2017
May
;
77
:
57
74
.
[PubMed]
0959-8049
72.
Scarisbrick
JJ
,
Prince
HM
,
Vermeer
MH
,
Quaglino
P
,
Horwitz
S
,
Porcu
P
, et al.
Cutaneous Lymphoma International Consortium Study of Outcome in Advanced Stages of Mycosis Fungoides and Sézary Syndrome: Effect of Specific Prognostic Markers on Survival and Development of a Prognostic Model
.
J Clin Oncol
.
2015
Nov
;
33
(
32
):
3766
73
.
[PubMed]
0732-183X
73.
Jaque
A
,
Mereniuk
A
,
Walsh
S
,
Shear
NH
,
Sade
S
,
Zagorski
B
, et al.
Influence of the phenotype on mycosis fungoides prognosis, a retrospective cohort study of 160 patients
.
Int J Dermatol
.
2019
Aug
;
58
(
8
):
933
9
.
[PubMed]
0011-9059
74.
Marks
E
,
Shi
Y
,
Wang
Y
.
Two cases of phenotypic switch of primary cutaneous T cell lymphoma after treatment with an aggressive course and review of the literature
.
Virchows Arch
.
2019
Nov
;
475
(
5
):
637
48
.
[PubMed]
0945-6317
75.
Janiga
J
,
Kentley
J
,
Nabhan
C
,
Abdulla
F
.
Current systemic therapeutic options for advanced mycosis fungoides and Sézary syndrome
.
Leuk Lymphoma
.
2018
Mar
;
59
(
3
):
562
77
.
[PubMed]
1042-8194
76.
Guenova
E
,
Hoetzenecker
W
,
Rozati
S
,
Levesque
MP
,
Dummer
R
,
Cozzio
A
.
Novel therapies for cutaneous T-cell lymphoma: what does the future hold?
Expert Opin Investig Drugs
.
2014
Apr
;
23
(
4
):
457
67
.
[PubMed]
1354-3784
77.
Saulite
I
,
Hoetzenecker
W
,
Weidinger
S
,
Cozzio
A
,
Guenova
E
,
Wehkamp
U
.
Sézary Syndrome and Atopic Dermatitis: Comparison of Immunological Aspects and Targets
.
BioMed Res Int
.
2016
;
2016
:
9717530
.
[PubMed]
2314-6133
78.
Ramelyte
E
,
Dummer
R
,
Guenova
E
.
Investigative drugs for the treatment of cutaneous T-cell lymphomas (CTCL): an update
.
Expert Opin Investig Drugs
.
2019
Sep
;
28
(
9
):
799
809
.
[PubMed]
1354-3784
79.
Alpdogan
O
,
Kartan
S
,
Johnson
W
,
Sokol
K
,
Porcu
P
.
Systemic therapy of cutaneous T-cell lymphoma (CTCL)
.
Linchuang Zhongliuxue Zazhi
.
2019
Feb
;
8
(
1
):
10
.
[PubMed]
1009-0460
80.
Tsang
M
,
Gantchev
J
,
Netchiporouk
E
,
Moreau
L
,
Ghazawi
FM
,
Glassman
S
, et al.
A study of meiomitosis and novel pathways of genomic instability in cutaneous T-cell lymphomas (CTCL)
.
Oncotarget
.
2018
Dec
;
9
(
102
):
37647
61
.
[PubMed]
1949-2553
81.
Perez
C
,
Mondejar
R
,
Garcia-Diaz
N
,
Cereceda
L
,
Leon
A
,
Montes
S
, et al.
Advanced-stage Mycosis Fungoides. Role of STAT3, NFKB and NFAT pathways
.
Br J Dermatol
.
2019
.0007-0963
82.
Butler
RM
,
McKenzie
RC
,
Jones
CL
,
Flanagan
CE
,
Woollard
WJ
,
Demontis
M
, et al.
Contribution of STAT3 and RAD23B in Primary Sézary Cells to Histone Deacetylase Inhibitor FK228 Resistance
.
J Invest Dermatol
.
2019
Sep
;
139
(
9
):
1975
1984.e2
.
[PubMed]
0022-202X
83.
Li
Y
,
Wang
J
,
Yu
M
,
Wang
Y
,
Zhang
H
,
Yin
J
, et al.
SNF5 deficiency induces apoptosis resistance by repressing SATB1 expression in Sézary syndrome
.
Leuk Lymphoma
.
2018
Oct
;
59
(
10
):
2405
13
.
[PubMed]
1042-8194
84.
Contassot
E
,
French
LE
.
Epigenetic causes of apoptosis resistance in cutaneous T-cell lymphomas
.
J Invest Dermatol
.
2010
Apr
;
130
(
4
):
922
4
.
[PubMed]
0022-202X
85.
Johnson
WT
,
Mukherji
R
,
Kartan
S
,
Nikbakht
N
,
Porcu
P
,
Alpdogan
O
.
Allogeneic hematopoietic stem cell transplantation in advanced stage mycosis fungoides and Sézary syndrome: a concise review
.
Linchuang Zhongliuxue Zazhi
.
2019
Feb
;
8
(
1
):
12
.
[PubMed]
1009-0460
86.
Doerschner
M
,
Pekar-Lukacs
A
,
Messerli-Odermatt
O
,
Dommann-Scherrer
C
,
Rütti
M
,
Müller
AM
, et al.
Interferon alfa-2a maintenance after salvage autologous stem cell transplantation in atypical mycosis fungoides with central nervous system involvement
.
Br J Dermatol
.
2019
Dec
;
181
(
6
):
1296
302
.
[PubMed]
0007-0963
87.
Shalabi
D
,
Bistline
A
,
Alpdogan
O
,
Kartan
S
,
Mishra
A
,
Porcu
P
, et al.
Immune evasion and current immunotherapy strategies in mycosis fungoides (MF) and Sézary syndrome (SS)
.
Linchuang Zhongliuxue Zazhi
.
2019
Feb
;
8
(
1
):
11
.
[PubMed]
1009-0460
88.
Cristofoletti
C
,
Narducci
MG
,
Russo
G
.
Sézary Syndrome, recent biomarkers and new drugs
.
Linchuang Zhongliuxue Zazhi
.
2019
Feb
;
8
(
1
):
2
.
[PubMed]
1009-0460
89.
Alfred
A
,
Taylor
PC
,
Dignan
F
,
El-Ghariani
K
,
Griffin
J
,
Gennery
AR
, et al.
The role of extracorporeal photopheresis in the management of cutaneous T-cell lymphoma, graft-versus-host disease and organ transplant rejection: a consensus statement update from the UK Photopheresis Society
.
Br J Haematol
.
2017
Apr
;
177
(
2
):
287
310
.
[PubMed]
0007-1048
90.
Cho
A
,
Jantschitsch
C
,
Knobler
R
.
Extracorporeal Photopheresis-An Overview
.
Front Med (Lausanne)
.
2018
Aug
;
5
:
236
.
[PubMed]
2296-858X
91.
Trautinger
F
.
Phototherapy of mycosis fungoides
.
G Ital Dermatol Venereol
.
2017
Dec
;
152
(
6
):
597
606
.
[PubMed]
1827-1820
92.
Berneburg
M
,
Herzinger
T
,
Rampf
J
,
Hoetzenecker
W
,
Guenova
E
,
Meisner
C
, et al.
Efficacy of bath psoralen plus ultraviolet A (PUVA) vs. system PUVA in psoriasis: a prospective, open, randomized, multicentre study
.
Br J Dermatol
.
2013
Sep
;
169
(
3
):
704
8
.
[PubMed]
0007-0963
93.
Geskin
L
.
ECP versus PUVA for the treatment of cutaneous T-cell lymphoma
.
Skin Therapy Lett
.
2007
Jun
;
12
(
5
):
1
4
.
[PubMed]
1201-5989
94.
Querfeld
C
,
Rosen
ST
,
Kuzel
TM
,
Kirby
KA
,
Roenigk
HH
 Jr
,
Prinz
BM
, et al.
Long-term follow-up of patients with early-stage cutaneous T-cell lymphoma who achieved complete remission with psoralen plus UV-A monotherapy
.
Arch Dermatol
.
2005
Mar
;
141
(
3
):
305
11
.
[PubMed]
0003-987X
95.
Holtick
U
,
Marshall
SR
,
Wang
XN
,
Hilkens
CM
,
Dickinson
AM
.
Impact of psoralen/UVA-treatment on survival, activation, and immunostimulatory capacity of monocyte-derived dendritic cells
.
Transplantation
.
2008
Mar
;
85
(
5
):
757
66
.
[PubMed]
0041-1337
96.
Vieyra-Garcia
P
,
Crouch
JD
,
O’Malley
JT
,
Seger
EW
,
Yang
CH
,
Teague
JE
, et al.
Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma
.
JCI Insight
.
2019
Jan
;
4
(
1
):
124233
.
[PubMed]
2379-3708
97.
Atta
M
,
Papanicolaou
N
,
Tsirigotis
P
.
The role of extracorporeal photopheresis in the treatment of cutaneous T-cell lymphomas
.
Transfus Apheresis Sci
.
2012
Apr
;
46
(
2
):
195
202
.
[PubMed]
1473-0502
98.
Raphael
BA
,
Shin
DB
,
Suchin
KR
,
Morrissey
KA
,
Vittorio
CC
,
Kim
EJ
, et al.
High clinical response rate of Sezary syndrome to immunomodulatory therapies: prognostic markers of response
.
Arch Dermatol
.
2011
Dec
;
147
(
12
):
1410
5
.
[PubMed]
0003-987X
99.
Edelson
RL
.
Mechanistic insights into extracorporeal photochemotherapy: efficient induction of monocyte-to-dendritic cell maturation
.
Transfus Apheresis Sci
.
2014
Jun
;
50
(
3
):
322
9
.
[PubMed]
1473-0502
100.
Ventura
A
,
Vassall
A
,
Robinson
E
,
Filler
R
,
Hanlon
D
,
Meeth
K
, et al.
Extracorporeal Photochemotherapy Drives Monocyte-to-Dendritic Cell Maturation to Induce Anticancer Immunity
.
Cancer Res
.
2018
Jul
;
78
(
14
):
4045
58
.
[PubMed]
0008-5472
101.
Shinkawa
T
,
Nakamura
K
,
Yamane
N
,
Shoji-Hosaka
E
,
Kanda
Y
,
Sakurada
M
, et al.
The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity
.
J Biol Chem
.
2003
Jan
;
278
(
5
):
3466
73
.
[PubMed]
0021-9258
102.
Suzuki
Y
,
Saito
M
,
Ishii
T
,
Urakawa
I
,
Matsumoto
A
,
Masaki
A
, et al.
Mogamulizumab treatment elicits autoantibodies attacking the skin in patients with adult T cell leukemia-lymphoma
.
Clin Cancer Res
.
2019
Jul
;
25
(
14
):
4388
99
.
[PubMed]
1078-0432
103.
Bonnet
P
,
Battistella
M
,
Roelens
M
,
Ram-Wolff
C
,
Herms
F
,
Frumholtz
L
, et al.
Association of autoimmunity and long-term complete remission in patients with Sézary syndrome treated with mogamulizumab
.
Br J Dermatol
.
2019
Feb
;
180
(
2
):
419
20
.
[PubMed]
0007-0963
104.
Ishida
T
,
Jo
T
,
Takemoto
S
,
Suzushima
H
,
Suehiro
Y
,
Choi
I
, et al.
Follow-up of a randomised phase II study of chemotherapy alone or in combination with mogamulizumab in newly diagnosed aggressive adult T-cell leukaemia-lymphoma: impact on allogeneic haematopoietic stem cell transplantation
.
Br J Haematol
.
2019
Feb
;
184
(
3
):
479
83
.
[PubMed]
0007-1048
105.
Fuji
S
,
Shindo
T
.
Friend or foe? Mogamulizumab in allogeneic hematopoietic stem cell transplantation for adult T-cell leukemia/lymphoma
.
Stem Cell Investig
.
2016
Oct
;
3
:
70
.
[PubMed]
2313-0792
106.
Rubio Gonzalez
B
,
Zain
J
,
Rosen
ST
,
Querfeld
C
.
Tumor microenvironment in mycosis fungoides and Sézary syndrome
.
Curr Opin Oncol
.
2016
Jan
;
28
(
1
):
88
96
.
[PubMed]
1040-8746
107.
Torrealba
MP
,
Manfrere
KC
,
Miyashiro
DR
,
Lima
JF
,
de M Oliveira
L
,
Pereira
NZ
, et al.
Chronic activation profile of circulating CD8+ T cells in Sézary syndrome
.
Oncotarget
.
2017
Dec
;
9
(
3
):
3497
506
.
[PubMed]
1949-2553
108.
Samimi
S
,
Benoit
B
,
Evans
K
,
Wherry
EJ
,
Showe
L
,
Wysocka
M
, et al.
Increased programmed death-1 expression on CD4+ T cells in cutaneous T-cell lymphoma: implications for immune suppression
.
Arch Dermatol
.
2010
Dec
;
146
(
12
):
1382
8
.
[PubMed]
0003-987X
109.
Cetinözman
F
,
Jansen
PM
,
Vermeer
MH
,
Willemze
R
.
Differential expression of programmed death-1 (PD-1) in Sézary syndrome and mycosis fungoides
.
Arch Dermatol
.
2012
Dec
;
148
(
12
):
1379
85
.
[PubMed]
0003-987X
110.
Wada
DA
,
Wilcox
RA
,
Harrington
SM
,
Kwon
ED
,
Ansell
SM
,
Comfere
NI
.
Programmed death 1 is expressed in cutaneous infiltrates of mycosis fungoides and Sézary syndrome
.
Am J Hematol
.
2011
Mar
;
86
(
3
):
325
7
.
[PubMed]
0361-8609
111.
Khodadoust
M
,
Rook
AH
,
Porcu
P
,
Foss
FM
,
Moskowitz
AJ
,
Shustov
AR
, et al.
Pembrolizumab for Treatment of Relapsed/Refractory Mycosis Fungoides and Sezary Syndrome: Clinical Efficacy in a Citn Multicenter Phase 2 Study
.
Blood
.
2016
;
128
(
22
):
181
. 0006-4971
112.
Khodadoust
MS
,
Rook
A
,
Porcu
P
,
Foss
FM
,
Moskowitz
AJ
,
Shustov
AR
, et al.
Durable Responses with Pembrolizumab in Relapsed/Refractory Mycosis Fungoides and Sézary Syndrome: Final Results from a Phase 2 Multicenter Study
.
Blood
.
2018
;
132
Suppl 1
:
2896
. 0006-4971
113.
Saulite
I
,
Ignatova
D
,
Chang
YT
,
Fassnacht
C
,
Dimitriou
F
,
Varypataki
E
, et al.
Blockade of programmed cell death protein 1 (PD-1) in Sézary syndrome reduces Th2 phenotype of non-tumoral T lymphocytes but may enhance tumor proliferation
.
OncoImmunology
.
2020
;
9
(
1
):
1738797
. 2162-4011
114.
Wilcox
RA
,
Feldman
AL
,
Wada
DA
,
Yang
ZZ
,
Comfere
NI
,
Dong
H
, et al.
B7-H1 (PD-L1, CD274) suppresses host immunity in T-cell lymphoproliferative disorders
.
Blood
.
2009
Sep
;
114
(
10
):
2149
58
.
[PubMed]
0006-4971
115.
Anzengruber
F
,
Ignatova
D
,
Schlaepfer
T
,
Chang
YT
,
French
LE
,
Pascolo
S
, et al.
Divergent LAG-3 versus BTLA, TIGIT, and FCRL3 expression in Sézary syndrome
.
Leuk Lymphoma
.
2019
Aug
;
60
(
8
):
1899
907
.
[PubMed]
1042-8194
116.
Ansell
S
,
Gutierrez
ME
,
Shipp
MA
,
Gladstone
D
,
Moskowitz
A
,
Borello
I
, et al.
A Phase 1 Study of Nivolumab in Combination with Ipilimumab for Relapsed or Refractory Hematologic Malignancies (CheckMate 039)
.
Blood
.
2016
;
128
(
22
):
183
. 0006-4971
117.
Jariwala
N
,
Benoit
B
,
Kossenkov
AV
,
Oetjen
LK
,
Whelan
TM
,
Cornejo
CM
, et al.
TIGIT and Helios Are Highly Expressed on CD4+ T Cells in Sézary Syndrome Patients
.
J Invest Dermatol
.
2017
Jan
;
137
(
1
):
257
60
.
[PubMed]
0022-202X
118.
Wysocka
M
,
Kossenkov
AV
,
Benoit
BM
,
Troxel
AB
,
Singer
E
,
Schaffer
A
, et al.
CD164 and FCRL3 are highly expressed on CD4+CD26- T cells in Sézary syndrome patients
.
J Invest Dermatol
.
2014
Jan
;
134
(
1
):
229
36
.
[PubMed]
0022-202X
119.
Spodzieja
M
,
Lach
S
,
Iwaszkiewicz
J
,
Cesson
V
,
Kalejta
K
,
Olive
D
, et al.
Design of short peptides to block BTLA/HVEM interactions for promoting anticancer T-cell responses
.
PLoS One
.
2017
Jun
;
12
(
6
):
e0179201
.
[PubMed]
1932-6203
120.
Johnson
LD
,
Banerjee
S
,
Kruglov
O
,
Viller
NN
,
Horwitz
SM
,
Lesokhin
A
, et al.
Targeting CD47 in Sézary syndrome with SIRPαFc
.
Blood Adv
.
2019
Apr
;
3
(
7
):
1145
53
.
[PubMed]
2473-9529
121.
Folkes
AS
,
Feng
M
,
Zain
JM
,
Abdulla
F
,
Rosen
ST
,
Querfeld
C
.
Targeting CD47 as a cancer therapeutic strategy: the cutaneous T-cell lymphoma experience
.
Curr Opin Oncol
.
2018
Sep
;
30
(
5
):
332
7
.
[PubMed]
1040-8746
122.
Ansell
S
,
Chen
RW
,
Flinn
IW
,
Maris
MB
,
O’Connor
OA
,
Johnson
LD
, et al.
A Phase 1 Study of TTI-621, a Novel Immune Checkpoint Inhibitor Targeting CD47, in Patients with Relapsed or Refractory Hematologic Malignancies
.
Blood
.
2016
;
128
(
22
):
1812
. 0006-4971
123.
Jiang
L
,
Yuan
CM
,
Hubacheck
J
,
Janik
JE
,
Wilson
W
,
Morris
JC
, et al.
Variable CD52 expression in mature T cell and NK cell malignancies: implications for alemtuzumab therapy
.
Br J Haematol
.
2009
Apr
;
145
(
2
):
173
9
.
[PubMed]
0007-1048
124.
Lundin
J
,
Hagberg
H
,
Repp
R
,
Cavallin-Ståhl
E
,
Fredén
S
,
Juliusson
G
, et al.
Phase 2 study of alemtuzumab (anti-CD52 monoclonal antibody) in patients with advanced mycosis fungoides/Sezary syndrome
.
Blood
.
2003
Jun
;
101
(
11
):
4267
72
.
[PubMed]
0006-4971
125.
Battistella
M
,
Leboeuf
C
,
Ram-Wolff
C
,
Hurabielle
C
,
Bonnafous
C
,
Sicard
H
, et al.
KIR3DL2 expression in cutaneous T-cell lymphomas: expanding the spectrum for KIR3DL2 targeting
.
Blood
.
2017
Dec
;
130
(
26
):
2900
2
.
[PubMed]
0006-4971
126.
Hurabielle
C
,
Thonnart
N
,
Ram-Wolff
C
,
Sicard
H
,
Bensussan
A
,
Bagot
M
, et al.
Usefulness of KIR3DL2 to Diagnose, Follow-Up, and Manage the Treatment of Patients with Sézary Syndrome
.
Clin Cancer Res
.
2017
Jul
;
23
(
14
):
3619
27
.
[PubMed]
1078-0432
127.
Marie-Cardine
A
,
Viaud
N
,
Thonnart
N
,
Joly
R
,
Chanteux
S
,
Gauthier
L
, et al.
IPH4102, a humanized KIR3DL2 antibody with potent activity against cutaneous T-cell lymphoma
.
Cancer Res
.
2014
Nov
;
74
(
21
):
6060
70
.
[PubMed]
0008-5472
128.
Bagot
M
,
Porcu
P
,
Marie-Cardine
A
,
Battistella
M
,
William
BM
,
Vermeer
M
, et al.
IPH4102, a first-in-class anti-KIR3DL2 monoclonal antibody, in patients with relapsed or refractory cutaneous T-cell lymphoma: an international, first-in-human, open-label, phase 1 trial
.
Lancet Oncol
.
2019
Aug
;
20
(
8
):
1160
70
.
[PubMed]
1470-2045
129.
Scarfò
I
,
Frigault
MJ
,
Maus
MV
.
CAR-Based Approaches to Cutaneous T-Cell Lymphoma
.
Front Oncol
.
2019
Apr
;
9
:
259
.
[PubMed]
2234-943X
130.
Spaccarelli
N
,
Rook
AH
.
The Use of Interferons in the Treatment of Cutaneous T-Cell Lymphoma
.
Dermatol Clin
.
2015
Oct
;
33
(
4
):
731
45
.
[PubMed]
0733-8635
131.
Olsen
EA
,
Rook
AH
,
Zic
J
,
Kim
Y
,
Porcu
P
,
Querfeld
C
, et al.
Sézary syndrome: immunopathogenesis, literature review of therapeutic options, and recommendations for therapy by the United States Cutaneous Lymphoma Consortium (USCLC)
.
J Am Acad Dermatol
.
2011
Feb
;
64
(
2
):
352
404
.
[PubMed]
0190-9622
132.
Kaplan
EH
,
Rosen
ST
,
Norris
DB
,
Roenigk
HH
 Jr
,
Saks
SR
,
Bunn
PA
 Jr
.
Phase II study of recombinant human interferon gamma for treatment of cutaneous T-cell lymphoma
.
J Natl Cancer Inst
.
1990
Feb
;
82
(
3
):
208
12
.
[PubMed]
0027-8874
133.
Dummer
R
,
Hassel
JC
,
Fellenberg
F
,
Eichmüller
S
,
Maier
T
,
Slos
P
, et al.
Adenovirus-mediated intralesional interferon-gamma gene transfer induces tumor regressions in cutaneous lymphomas
.
Blood
.
2004
Sep
;
104
(
6
):
1631
8
.
[PubMed]
0006-4971
134.
Rook
AH
,
Kubin
M
,
Fox
FE
,
Niu
Z
,
Cassin
M
,
Vowels
BR
, et al.
The potential therapeutic role of interleukin-12 in cutaneous T-cell lymphoma
.
Ann N Y Acad Sci
.
1996
Oct
;
795
1 Interleukin 1
:
310
8
.
[PubMed]
0077-8923
135.
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
Jul
;
99
(
1
):
90
4
.
[PubMed]
0022-202X
136.
Morrison
C
,
Baer
MR
,
Zandberg
DP
,
Kimball
A
,
Davila
E
.
Effects of Toll-like receptor signals in T-cell neoplasms
.
Future Oncol
.
2011
Feb
;
7
(
2
):
309
20
.
[PubMed]
1479-6694
137.
Terhorst
D
,
Kalali
BN
,
Ollert
M
,
Ring
J
,
Mempel
M
.
The role of toll-like receptors in host defenses and their relevance to dermatologic diseases
.
Am J Clin Dermatol
.
2010
;
11
(
1
):
1
10
.
[PubMed]
1175-0561
138.
Gibson
SJ
,
Lindh
JM
,
Riter
TR
,
Gleason
RM
,
Rogers
LM
,
Fuller
AE
, et al.
Plasmacytoid dendritic cells produce cytokines and mature in response to the TLR7 agonists, imiquimod and resiquimod
.
Cell Immunol
.
2002
Jul-Aug
;
218
(
1-2
):
74
86
.
[PubMed]
0008-8749
139.
Suchin
KR
,
Junkins-Hopkins
JM
,
Rook
AH
.
Treatment of stage IA cutaneous T-Cell lymphoma with topical application of the immune response modifier imiquimod
.
Arch Dermatol
.
2002
Sep
;
138
(
9
):
1137
9
.
[PubMed]
0003-987X
140.
Dummer
R
,
Urosevic
M
,
Kempf
W
,
Kazakov
D
,
Burg
G
.
Imiquimod induces complete clearance of a PUVA-resistant plaque in mycosis fungoides
.
Dermatology
.
2003
;
207
(
1
):
116
8
.
[PubMed]
1018-8665
141.
Chong
A
,
Loo
WJ
,
Banney
L
,
Grant
JW
,
Norris
PG
.
Imiquimod 5% cream in the treatment of mycosis fungoides—a pilot study
.
J Dermatolog Treat
.
2004
Apr
;
15
(
2
):
118
9
.
[PubMed]
0954-6634
142.
Deeths
MJ
,
Chapman
JT
,
Dellavalle
RP
,
Zeng
C
,
Aeling
JL
.
Treatment of patch and plaque stage mycosis fungoides with imiquimod 5% cream
.
J Am Acad Dermatol
.
2005
Feb
;
52
(
2
):
275
80
.
[PubMed]
0190-9622
143.
Chiam
LY
,
Chan
YC
.
Solitary plaque mycosis fungoides on the penis responding to topical imiquimod therapy
.
Br J Dermatol
.
2007
Mar
;
156
(
3
):
560
2
.
[PubMed]
0007-0963
144.
Martínez-González
MC
,
Verea-Hernando
MM
,
Yebra-Pimentel
MT
,
Del Pozo
J
,
Mazaira
M
,
Fonseca
E
.
Imiquimod in mycosis fungoides
.
Eur J Dermatol
.
2008
Mar-Apr
;
18
(
2
):
148
52
.
[PubMed]
1167-1122
145.
Lewis
DJ
,
Byekova
YA
,
Emge
DA
,
Duvic
M
.
Complete resolution of mycosis fungoides tumors with imiquimod 5% cream: a case series
.
J Dermatolog Treat
.
2017
Sep
;
28
(
6
):
567
9
.
[PubMed]
0954-6634
146.
Gorden
KB
,
Gorski
KS
,
Gibson
SJ
,
Kedl
RM
,
Kieper
WC
,
Qiu
X
, et al.
Synthetic TLR agonists reveal functional differences between human TLR7 and TLR8.
Journal of immunology (Baltimore, Md : 1950).
2005
;174(3):1259-68.
147.
Rook
AH
,
Gelfand
JM
,
Wysocka
M
,
Troxel
AB
,
Benoit
B
,
Surber
C
, et al.
Topical resiquimod can induce disease regression and enhance T-cell effector functions in cutaneous T-cell lymphoma
.
Blood
.
2015
Sep
;
126
(
12
):
1452
61
.
[PubMed]
0006-4971
148.
Iwasaki
A
,
Medzhitov
R
.
Toll-like receptor control of the adaptive immune responses
.
Nat Immunol
.
2004
Oct
;
5
(
10
):
987
95
.
[PubMed]
1529-2908
149.
Kim
YH
,
Gratzinger
D
,
Harrison
C
,
Brody
JD
,
Czerwinski
DK
,
Ai
WZ
, et al.
In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: a phase 1/2 study
.
Blood
.
2012
Jan
;
119
(
2
):
355
63
.
[PubMed]
0006-4971
150.
Talpur
R
,
Bassett
R
,
Duvic
M
.
Prevalence and treatment of Staphylococcus aureus colonization in patients with mycosis fungoides and Sézary syndrome
.
Br J Dermatol
.
2008
Jul
;
159
(
1
):
105
12
.
[PubMed]
0007-0963
151.
Blümel
E
,
Willerslev-Olsen
A
,
Gluud
M
,
Lindahl
LM
,
Fredholm
S
,
Nastasi
C
, et al.
Staphylococcal alpha-toxin tilts the balance between malignant and non-malignant CD4+ T cells in cutaneous T-cell lymphoma
.
OncoImmunology
.
2019
Jul
;
8
(
11
):
e1641387
.
[PubMed]
2162-4011
152.
Tokura
Y
,
Heald
PW
,
Yan
SL
,
Edelson
RL
.
Stimulation of cutaneous T-cell lymphoma cells with superantigenic staphylococcal toxins
.
J Invest Dermatol
.
1992
Jan
;
98
(
1
):
33
7
.
[PubMed]
0022-202X
153.
Fanok
MH
,
Sun
A
,
Fogli
LK
,
Narendran
V
,
Eckstein
M
,
Kannan
K
, et al.
Role of Dysregulated Cytokine Signaling and Bacterial Triggers in the Pathogenesis of Cutaneous T-Cell Lymphoma
.
J Invest Dermatol
.
2018
May
;
138
(
5
):
1116
25
.
[PubMed]
0022-202X
154.
Lindahl
LM
,
Willerslev-Olsen
A
,
Gjerdrum
LM
,
Nielsen
PR
,
Blümel
E
,
Rittig
AH
, et al.
Antibiotics inhibit tumor and disease activity in cutaneous T-cell lymphoma
.
Blood
.
2019
Sep
;
134
(
13
):
1072
83
.
[PubMed]
0006-4971
155.
Vermeer
MH
.
Antibiotics can improve CTCL
.
Blood
.
2019
Sep
;
134
(
13
):
1000
1
.
[PubMed]
0006-4971

Edited by: H.-U. Simon, Bern.Malgorzata Bobrowicz and Christina Fassnacht contributed equally to this work.

Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC). Usage and distribution for commercial purposes requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.