Next Article in Journal
Controlling Recombination to Evolve Bacteriophages
Previous Article in Journal
Safety of Anti-Reelin Therapeutic Approaches for Chronic Inflammatory Diseases
Previous Article in Special Issue
Differential Upregulation of Th1/Th17-Associated Proteins and PD-L1 in Granulomatous Mycosis Fungoides
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets

by
Alba Guglielmo
1,2,†,
Corrado Zengarini
2,3,*,†,
Claudio Agostinelli
2,4,
Giovanna Motta
2,4,
Elena Sabattini
2,4 and
Alessandro Pileri
2,3,*
1
Institute of Dermatology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
2
Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
3
Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
4
Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2024, 13(7), 584; https://doi.org/10.3390/cells13070584
Submission received: 6 December 2023 / Revised: 21 March 2024 / Accepted: 26 March 2024 / Published: 28 March 2024

Abstract

:
Cutaneous T cell lymphomas (CTCLs), encompassing mycosis fungoides (MF) and Sézary syndrome (SS), present a complex landscape influenced by cytokines and cellular responses. In this work, the intricate relationship between these inflammatory proteins and disease pathogenesis is examined, focusing on what is known at the clinical and therapeutic levels regarding the most well-known inflammatory mediators. An in-depth look is given to their possible alterations caused by novel immunomodulatory drugs and how they may alter disease progression. From this narrative review of the actual scientific landscape, Interferon-gamma (IFN-γ) emerges as a central player, demonstrating a dual role in both promoting and inhibiting cancer immunity, but the work navigates through all the major interleukins known in inflammatory environments. Immunotherapeutic perspectives are elucidated, highlighting the crucial role of the cutaneous microenvironment in shaping dysfunctional cell trafficking, antitumor immunity, and angiogenesis in MF, showcasing advancements in understanding and targeting the immune phenotype in CTCL. In summary, this manuscript aims to comprehensively explore the multifaceted aspects of CTCL, from the immunopathogenesis and cytokine dynamics centred around TNF-α and IFN-γ to evolving therapeutic modalities. Including all the major known and studied cytokines in this analysis broadens our understanding of the intricate interplay influencing CTCL, paving the way for improved management of this complex lymphoma.

1. Introduction

Cutaneous T cell lymphomas (CTCLs) are a group of non-Hodgkin lymphomas characterized by the appearance of the skin and the absence of extra-cutaneous dissemination at least six months after diagnosis [1,2]. Mycosis fungoides (MF) and Sézary syndrome (SS) are the two most common CTCLs, accounting for at least 50% of all CTCL diagnoses. From an epidemiological point of view, CTCL incidence has increased over recent decades, especially for MF and SS [3,4,5,6]. Hence, clinicians’ efforts are focused on understanding the mechanisms involved in CTCL pathogenesis to develop more effective and tailored therapies [7,8,9]. MF/SS pathogenesis and the mechanisms involved in their progression from early to advanced phases are far from being fully understood [10,11,12]. Different players are involved in MF/SS, such as gene alterations, changes in the microenvironment composition, cytokine balance changes, and transcriptional pathways [10,11,12,13,14,15,16]. All these players are intrinsically linked.
The present paper aims to analyze the potential role of cytokine changes in MF/SS pathogenesis and progression and to investigate possible therapeutic targets by narratively reviewing the literature.
To list cytokines in the most concise but relevant manner, given the significantly changing panorama regarding the nomenclature, the number, and the definitions, we based our work on the papers of Dinarello [17] and Wautier et al. [18]. We then included the family of interleukins going from 1 to 33, tumor necrosis factor, interferons, platelets, fibroblasts, and epidermal growth factors.
For better readability of the text, a summary of the described cytokines, their mechanisms, and known and potential related therapies are listed in Table 1.

2. IL-1

The Interleukin 1 (IL-1) family includes many known pro-inflammatory cytokine subtypes that play crucial roles in amplifying the immune reaction and controlling various innate-response processes (IL-1β, IL-1α, IL-18, IL-33, IL-36α, IL-36β, IL-36γ, IL-36RA, IL-37, IL-38, and IL-1RA) [17,102]. IL-1 is also a known mediator of fever and a leukocytic endogenous cascade.
Nevertheless, in the tumoral environment, it behaves counterintuitively: in breast cancer, IL-1β is able to induce the recruitment of immunosuppressive cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), gaining advantages for neoplastic spread [102,103].

IL-1 in MF/SS

IL-1′s role in MF/SS has been the subject of a few studies [104]. In these published works, it has been reported that low levels of immunostimulating cytokines (among them, IL-1 was included) are expressed in MF samples, speculating whether IL-1 may play a potential role in CTCL pathogenesis.
Another study showed how the major IL-1 subtypes alpha and beta are relatively less expressed in CTCLs when compared to healthy donors [19]. It has not been explained if this phenomenon has a pathogenetic role in halting disease progression, but it has been described as a potential biomarker in responsive patients to photopheresis [19,105].

3. IL-2

The physiological role of interleukin (IL)-2, also known as T cell growth factor (TCGF), is to upregulate the proliferation and cytolytic activity of activated T cells and to increase the cytotoxicity of monocytes and natural killer (NK) cells [21].
Thus, IL-2 is essential in activating the immune system against infective agents [21,106]. The IL-2 receptor (IL-2R) can be present in three subunits, IL-2Rα (also known as CD25), IL-2Rβ, and IL-2Rγ, and these are expressed on the surfaces of different cells important for activating memory T cells and prompting the immune response. IL-2Rγ (also known as common gamma chain or γc) dimerization is shared with the IL-4, IL-7, IL-9, IL-15, and IL-21 cytokines [44]. The interaction between IL-2 and IL-2R is a crucial stage of immune system activation. However, the IL-2Rα subunit plays a vital role in controlling and generating immune-suppressive T-regulatory cells (T-regs) [21]. IL-2 can act as an immunostimulating cytokine by expanding and increasing the proliferation of natural killer and cytotoxic CD8+ T cells. Moreover, IL-2 can induce the apoptosis of proliferating T cells through the Fas-FasL pathway and positively regulate T-reg cells, leading to immune tolerance and suppression [107].

IL-2 in MF/SS

IL-2 has been the subject of some MF studies. Owing to IL-2’s ability to expand the immune response, several groups [108,109,110,111] have investigated the possible therapeutic role of a recombinant IL-2 in MF patients. However, contrasting results have been provided in the literature, and IL-2’s role in MF is still open. In 2001, Brockdorff et al. [22] highlighted the tumorigenic role of IL-2.
Indeed, the authors found high levels of Gab2, an adaptor molecule involved in IL-2 receptor signaling in CTCLs, via IL-2 stimulation; Brockdorff speculated whether Gab2 could be regarded as a possible MF/SS progression marker. However, the role of IL-2 and its receptor remains controversial in the literature.
Shohat et al. [23] hypothesized that IL-2R may have an immunosuppressive role rather than IL-2. By investigating the effect of AS101, a tellurium-based compound with immunomodulating properties, on the pattern of lymphokine production by peripheral blood mononuclear cells (PBMCs) from patients with mycosis fungoides, Shohat et al. [23] found that AS101 [24] inhibited the production of IL-2R, IL-5, and IL-10 and induced a significant increase in IL-2 levels in the mycosis fungoides PBMCs, suggesting that IL-2R may have an immunosuppressive role. In contrast, IL-2 may have an antitumor action.
Another potential drug targeting IL-2R is a fusion toxin called denileukin diftitox [112,113], a drug that intracellularly delivers diphtheria toxin and consecutively inhibits protein synthesis in IL-2R-expressing cells. In a murine model of IL-2R-expressing malignancies, denileukin diftitox prolonged survival compared with controls. The most common adverse events reported in patients who received denileukin diftitox were hypoalbuminemia, fever/chills, acute hypersensitivity reactions, nausea/vomiting, and asthenia.
Kaminetsky and Himes et al. used it in patients with IL-2R-positive cutaneous T cell lymphoma (CTCL), reaching an overall response of 36% [113].
Duvic et al. used the same drug in relapsed CTCLs in an open-label phase III trial with an overall response of 40%. However, progression-free survival was 205 days, the median duration of response was 274 days, and three patients withdrew because of toxicity. Still, these studies show how denileukin diftitox may provide a clinically meaningful response in CTCLs
Another potential drug targeting IL-2 signaling under development is CCR4-IL2 IT. IL2-CCR4 bispecific IT is a receptor-specific immunotoxin targeting both CCR4 and IL2Rα and has been shown in preclinical studies to be significantly more effective than IL2 fusion toxin and brentuximab [26]. Still, clinical trials are needed to assess its potential role in the treatment of human CD25+ and/or CCR4+ CTCL.

4. IL-3

Interleukin 3 (IL-3, also defined as multi-CSF), often in conjunction with other cytokines such as IL-5, regulates immune system activation against pathogens. Activated T cells and basophils secrete IL-3, fostering T cell growth and differentiation and stimulating macrophage and granulocyte maturation [27].

IL-3 in MF/SS

To date, no studies have provided data on a role of IL-3 in MF/SS progression.

5. IL-4

Interleukin 4 (IL-4) is a cytokine involved in the skew to a Th2 phenotype of naïve T cells. Differentiation to a Th2 phenotype determines the consequence of a positive loop towards an increased production of IL-4 molecules. The receptor for interleukin-4 (IL-4Rα) has three different complexes throughout the body. Type 1, IL-4Rα, is effective when dimerized with the common gamma chain (γc), sharing the same mechanism as IL-2, IL-7, IL-9, IL-15, and IL-21 [27] and specifically binding IL-4. Type 2 receptors are bound to a different subunit, known as IL-13Rα1, and are shared with IL-12. One of the many physiological actions of IL-4 is to negatively modulate Th1 T cells, dendritic cells, and IFNγ production [114], facilitating tumor growth.
On the other hand, it promotes Th2 T cell differentiation, promoting anti-inflammatory and immunosuppressive responses, specifically by promoting the recruitment and activation of immunosuppressive cells such as T-regs and MDSCs. In the literature, it is known that, in CTCLs, an imbalance towards Th2 polarization driven by IL-4 may contribute to immune evasion and tumor progression by suppressing anti-tumor immune responses and enhancing pro-tumorigenic microenvironments [115].
In brief, IL-4 exerts an immunosuppressive action and, from an oncologic point of view, acts as a tumorigenic cytokine [31,114,116].

IL-4 in MF/SS

Since the early 1990s, increased production of IL-4 has been linked with CTCL progression [117,118,119,120]. IL-4 is thought to be one of the cytokines driving the shift from an antitumor to a tumorigenic phenotype. It decreases the production of Th1-associated cytokines, such as IL-12 and IFN-γ [121], so IL-4 can be regarded as an immunosuppressive molecule. IL-4, in association with IL-13, promotes tumor cell growth and proliferation in CTCL [122].
Furudate et al. hypothesized that IL-4 may act on the innate immune response by polarizing macrophages into immunosuppressive M2 macrophages. M2 macrophages produce Th-2 cytokines and stromal factors, promoting Th2 polarization of the microenvironment [123,124]
IL-4 can also synergically act with IL-33, inducing the secretion of IL-31, an interleukin responsible for the pathogenesis of itch [84,125].
From another perspective, CTLCs are related to high levels of tumoral Th2 T cells, so blocking Th2 differentiation is thought to reduce or alter CTLC diseases.
However, Saulite et al. asserted that a functional PD-1 block with nivolumab, in their pre-clinical work, resulted in a reduced Th2 phenotype of non-tumor T lymphocytes and enhanced the proliferation of tumor T cells in SS patients [32], shedding some light in the use of checkpoint inhibitors and CTCLs.
Still, blocking malignant T cells’ Th2 differentiation and proliferation by addressing the intracellular signaling pathway could be another feasible option. Ritlecitinib is a Janus kinase (JAK) inhibitor which has shown the capacity to reduce Th2 cells [126], and there is a phase IIA clinical trial evaluating this drug in CTCLs [33]. Guenova et al. [127] observed, in their analysis, that malignant T cells from patients with the disease typically express Th2 cytokines such as IL-4 and IL-13 but are negative for IFNγ, highlighting the MF cell’s ability to reduce immune system control by IFNγ low levels. High IL-4 levels also stimulate dermal fibroblasts from CTCL patients to secrete increased extracellular matrix protein periostin [128]. As a cascade of events, periostin will induce thymic stromal lymphopoietin expression, activating the STAT5 pathway and eventually promoting both neoplastic cell proliferation and the production of IL-4 [128,129,130,131]. A new monoclonal antibody against IL-4 (and IL-13), dupilumab, has recently become available for atopic dermatitis treatment [34].
Theoretically, by blocking IL-4 immune suppressive action, a reawakening of patients’ immune systems should be expected. However, in the literature, cases of misdiagnosed MF treated with dupilumab are available, featuring a dramatic disease progression [35,132,133]. It has been hypothesized that dupilumab action may paradoxically activate the JAK/STAT pathway, leading to results opposite to those expected.

6. IL-5

Interleukin 5 (IL-5) is a cytokine secreted by the activated Th-2 cell subset. IL-5 gene expression is linked with the IL-4 and IL-13 genes in a Th2 cytokine gene cluster on chromosome 5q [53].
The function of IL-5 has been best studied in the context of airway inflammation, particularly asthma. IL-5 promotes the differentiation, proliferation, recruitment, and activity of eosinophils, which are responsible for airway hyperreactivity and epithelial damage in allergic asthma [54,55].

IL-5 in MF/SS

Eosinophilia and elevated levels of immunoglobulin E (IgE) are often observed in advanced CTCLs due to the shift to a Th2 response, and IL-5 represents the principal regulator of eosinophilia [134,135,136]. It has been assessed that IL-5 plays a crucial role in the development of eosinophilia in Hodgkin’s lymphoma. In CTCLs, the production of IL-5 has also been related to erythroderma and elevated serum levels of IgE [38]. It has been hypothesized that IL-5 may be overexpressed in CTCL, owing to the constitutive activation of the STAT3 pathway in neoplastic cells, while no evidence that non-malignant T cell lines may secrete IL-5 has been observed. [38] The potential immunosuppressive action of IL-5 was observed by Shohat et al. [23] by comparing the IL-2, IL-2R, IL-5, IL-10, and IFN gamma levels in the peripheral blood mononuclear cells (PBMCs) of healthy volunteers and CTCL patients. The Israeli group found significantly higher levels of IL-2R, IL-5, and IL-10 in the affected patients and significantly lower levels of interferon-gamma. After the use of AS101, a tellurium-based compound with immunomodulating properties, the cytokine profile changed, with an increase in IFN gamma and a decrease in immunosuppressive cytokines, evidence observed previously by Yamamoto et al. [39] after the administration of IFN gamma.
Changes in IL-5 levels and other immunosuppressive or antitumor cytokines have been observed by other groups, highlighting a solid correlation between patient progression (and consequently, an increase in tumor burden both at the skin and blood levels) and an increase in immunosuppressive cytokine profile, including IL-5 [137]. In addition, other authors have proposed a correlation between IL-5 action and the miR-155/SATB1 axis, which is thought to play an essential role in CTCL progression and in other lymphoma pathogeneses. Low SATB1 expression levels, as observed in malignant SS cells, have been related to increased IL-5 production and higher malignant cell proliferation [51]. Aiming to add new possible response markers to scheduled therapies, Suchin at al. proposed that a decrease in IL-5 levels in treated patients may be regarded as a marker of clinical response to scheduled treatment, owing to the negative modulation of high antitumor cytokine levels such as IFN-α and IL-12 along with IL-5 [138].

7. IL-6

Interleukin 6 (IL-6) is a cytokine that synergistically acts in its role with IL-1. IL-6 induces acute-phase inflammation protein release, determines B lymphocyte differentiation into plasma cells, and increases adhesion molecules in inflammatory infiltrates [40,139].

IL-6 in MF/SS

Since the 1990s, Lawlor et al. and Watson et al. [40,104] have provided evidence that IL-6 may induce lymphocyte activation and migration in MF samples. Lawlor et al. [104] observed high IL-6 levels in lesional samples, hypothesizing that IL-6 may be related to MF/SS pathogenesis. In their experiments, Watson et al. [78] observed that high T-lymphocyte concentrations in MF infiltrates were related to high IL-1 and IL-6 levels. The authors hypothesized that promoting lymphocyte adherence with IL-6 may be related to a selective modification in vascular adherence molecules. Kadin et al. [41] proposed a possible prognostic role of IL-6 in early MF patients. By analysing 96 early MF cases, the Authors found that high serum CD30, CD25, and IL-6 levels were related to a higher risk of MF progression.
Olszewska et al. [43] recently proposed that the polymorphism of IL-6/STAT3 signaling may be related to the aggressive behavior of MF/SS. By comparing the serum IL-6 levels in 106 CTCL patients to 198 control cases, the authors found that their IL-6 serum levels were significantly higher than those in healthy controls. The same experiment also demonstrated that two genotypes, CC of IL-6 and GG of STAT3, were overexpressed in the CTCL patients, determining an increased risk of malignancy development. Due to the hypothesis that the GG genotype of the STAT3 polymorphism may be akin to the presence or absence of itch in CTCL patients, Olszewska et al. [43] highlighted that IL-6 may be involved in MF/SS, but not in the presence of itch in affected patients.
Recently, IL-6 was been studied as a possible marker of therapeutic response after PUVA/nbUVB phototherapy by Karamova et al. [140]. The authors analysed lesional skin cytokine changes (IL-1β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21, IL-22, IL-23, IL-25, IL-31, IL-33, IFN-γ, sCD40L, and TNF-α) before and after the treatments, observing a positive correlation between an mSWAT decrease and the levels of IL22, IL33, and TNF-α in the tumor tissue. The levels of IL10 and IFN-γ after PUVA treatment were increased in comparison to baseline. However, no changes in IL-6 levels were found.

8. IL-7

Interleukin-7 (IL-7) is a hematopoietic growth factor secreted by stromal cells in the bone marrow and thymus [141]. It is also produced by keratinocytes, dendritic cells, hepatocytes, neurons, and epithelial cells, but not by normal lymphocytes [141].
It stimulates the development of lymphoid lineage by binding to its specific receptor and activating signal transduction when dimerized with the common gamma chain (γc), with the same mechanism as IL-2, IL-4, IL-9, IL-15, and IL-21 [44], activating intracellular signaling pathways through JAK/STAT that promote cell proliferation, survival, and differentiation (principally B cells, T cells, and NK cells) [45].

IL-7 in MF/SS

In the medical literature, it has been written that an increased production of IL-7 could cause the chronic activation of CD8+ T cell clones in Sézary syndrome [142,143,144,145] and has been linked to the development of cutaneous lymphomas, given its relationship with lymphoid lineage progenitors’ stimulation.
However, there are very few works on the subject [142,144,146], and some have contrasting opinions, concluding that there is no in vivo confirmation of this correlation [46]. It is being studied as a potential immunotherapeutic drug [147], but there are no data regarding this use for CTCLs.

9. IL-8

See the paragraphs on Section 29.

10. IL-9

Interleukin 9 (IL-9) is a cytokine produced by various immune cells. It exerts its main activity when dimerized with the common gamma chain (γc), with the exact mechanism of IL-2, IL-4, IL-7, IL-15, and IL-21 on the IL-9 receptor [44], which is mainly expressed on T-helper cells subtype Th9, which are the major CD4+ T cells that produce IL-9. IL-33 can induce its production and also TGF-β [50].
Its primary function is to stimulate various hematopoietic cells’ proliferation and prevent the apoptosis of immune cells [50]. It activates the IL-9 receptor (IL9R) and intracellularly STAT pathway. It has also been linked to the development of asthma [148]. Due to its hematopoietic stimulations, it seems to give rise to multiple hematologic neoplasias and Hodgkin’s lymphoma, but it also has antitumoral properties in solid tumors, such as melanoma [149].

IL-9 in MF/SS

It seems that its hematopoietic function may be related to the appearance and maintenance of lymphomatous neoplasms [50].
This has been confirmed in some studies in which increased levels of Il-9 have been linked to patients suffering from Sézary syndrome [51] and mycosis fungoides [52].
As with Il-7, interest has been shown in stimulating its receptor as immunotherapy; however, there are currently no active studies on the uptake of CTCLs [150].

11. IL-10

Interleukin 10 (IL-10) plays a central role in preventing autoimmune diseases. IL-10 negatively modulates immune response during infection, allergy, and autoimmunity by increasing immunosuppressive cell levels [151]. On the other hand, due to its intrinsic immunosuppressive activity, it has been hypothesized that IL-10 may contribute to chronic infections [152]. An autocrine loop also regulates the production of IL-10.
Indeed, IL-10 is thought to be increased by T-regs lymphocytes recruited from blood vessels. IL-10 exerts its immunosuppressive action by inhibiting proinflammatory Th1 lymphocytes and the proliferation and differentiation of B and Th2 cells.

IL-10 in MF/SS

Higher levels of IL-10 have been detected in MF/SS biopsies compared with normal skin, corroborating the hypothesis that IL-10 may be involved in MF/SS pathogenesis and progression. Unsurprisingly, in their experiments, Wu et al. observed that higher levels of IL-10 can be seen in advanced CTCL phases rather than early ones [153]. Similar evidence was observed by Akatsuka et al. [55], who found that the development of IL-10-producing Bregs is impaired in patients with advanced MF, and a decrease in IL-10-producing Bregs may play an essential role in the progression of advanced MF. Different groups have focused on IL-10 as a possible therapeutic target or marker.
Tiffon et al. proposed that one of the histone deacetylase inhibitors, such as vorinostat and romidepsin, may exert their therapeutic action due to the downregulation of IL-10 RNA expression [56]. A similar action has been proposed for some proteasome inhibitors, like bortezomib, that modulate cytokine expression in CTCL, acting on TGFβ1 and IL-10 down-regulation [57]. IL-10′s role has also been studied from a genetic point of view, and upon reviewing the studies on the polymorphisms of the cytokine genes involved in CTCL pathogenesis, the paper by Nedoszytko B et al. is of some interest. Indeed, the authors observed that a polymorphic variant in the promoter region of IL-10 can reduce transcription factor recognition, leading to a decreased level of IL-10. The authors questioned whether such a gene variant may be related to a reduced risk of developing MF [154].

12. IL-11

Interleukin-11 (IL-11) plays a pivotal physiological role in multiple systems in the body. Primarily known for its involvement in hematopoiesis, it stimulates the production of platelets from megakaryocytes and hemostasis. Additionally, IL-11 showcases anti-inflammatory properties, modulating immune responses to maintain a balanced and regulated inflammatory environment [58].

IL-11 in MF/SS

While research continues, no direct established link exists between IL-11, MF, or SS.

13. IL-12

Interleukin 12 (IL-12) is a heterodimeric cytokine encoded by two separate genes, IL-12A and IL-12B (in common with IL-23), mainly transduced and transcribed by antigen-presenting cells, such as macrophages and dendritic cells [155,156].
Being a heterodimeric protein makes it included, from a structural perspective, in a family with IL-23, IL-27, and IL-35 [59]. Despite this, it has a different biological function [59], which consists of activating natural killer (NK) cells and promoting the differentiation of naive T cells into Th1 cells, thereby enhancing the immune system’s ability to fight intracellular pathogens and regulate inflammatory responses [155].
Finally, it stimulates IFNγ production by T cells, which primes additional APCs for IL-12 production, facilitating Th1 differentiation, and can also induce the production of IFNγ by NK cells [59].

Il-12 in MF/SS

Due to IL-12’s observed pro-inflammatory type 1 induction, it has long been studied as a potential immunotherapy for cancers, but it seems that its in vitro antitumor efficacy has not yet been well-established in humans [157].
Even if there are dated findings of a reduced or suppressed IL-12 pathway in CTCL development, especially in SS patients [60,101,158], studies regarding the impairment of the IL-12/23 axis in psoriatic patients have shown no evidence of an increased risk of altered clinical history in developing CTCLs [159]. Still, IL12/23 therapies are recent, and more long-term data are needed to confirm these findings.
Regarding the potential role of IL-12 as a treatment, there are some studies regarding the potential of the use of recombinant IL-12 in CTCLs: this is due to IL-12 being strongly able to induce Th-1 differentiation and conversion and suppressing Th-2 cytokines, making it a possible mechanism to treat MF and SS [60,138].
A clinical trial tried to assess IL-12′s role in IIA and the early stages of MF, with a 43% response in the reported patient series, but the study’s progression was interrupted due to company purchase [61].

14. IL-13

Different T cell subsets and dendritic cells produce interleukin 13 (IL-13), which is crucial in inhibiting pro-inflammatory cytokine production. IL-13 shares different biological activities with IL-4 by binding the IL-4Rα subunit [160,161]. However, recent studies have demonstrated a specific and nonredundant role for IL-13 in host immunity against parasites, inflammatory airway susceptibility, and tumor progression. IL-13 has a crucial role in the immune response against gastrointestinal helminth infections, such as Nippostrongylus brasiliensis [162], and a controversial role in leishmaniasis. Noben-Trauth et al. and Alexander et al. demonstrated that IL-13 promotes disease progression in the early phases of L. major and L. mexicana infections [29,163].
On the other hand, a protective role seems to be played in the chronic phases of infection [164]. Walter et al. studied the role of IL-13 in airway hyperresponsiveness and asthma in an IL-13(−/−) murine model [62], demonstrating that mice with the deletion of IL-13 failed to develop allergen-induced airway hyper-responsiveness. On the contrary, the blockade of IL-13 was ineffective in chronic disease with airway remodeling [165]. IL-13 has recently been studied in the context of cancer immunosurveillance. IL-13 is overexpressed in gastric, pancreatic, and oesophageal cancer patients [166,167]. Interestingly, the overexpression of the IL-13Rα2 chain in gastric cancer tissue is associated with a poor prognosis after gastrectomy [168]. IL-13 plays a crucial role in disease pathogenesis in different types of lymphomas, acting as an autocrine growth factor in Hodgkin‘s lymphoma and extranodal natural killer/T cell lymphoma [63,169].

IL-13 in MF/SS

IL-13 acts as an immunosuppressive cytokine in CTCLs. Geskin et al. speculated whether IL-13 may act as an autocrine factor in lymphoma cell proliferation through IL-13Rα1 and IL-13-Rα2 signaling. In their experiments, the American group found that the IL-13 protein was expressed in mononuclear cells in close contact with malignant cell aggregates in immunohistochemistry. In addition, a higher expression of IL-13 and its receptors within the infiltrate correlated with late stages, while in the early stages, this expression was low [122].
As mentioned above, numerous studies have highlighted that IL-13 and IL-4 have a synergistic effect, and their expression promotes tumor cell growth and proliferation in CTCLs. Some recent studies have shown a correlation between IL-13 and the JAK/STAT pathway [170]. Indeed, neoplastic cells can secrete mediators (including IL-13, IL-22, and oncostatin M) that activate STAT3 signaling and downregulate filaggrin and filaggrin-2 expression in human keratinocytes in vivo and in vitro. The authors hypothesized that skin barrier defects in CTCLs may be related to the production of cytokines via a JAK1/STAT3-dependent mechanism, proposing a possible JAK inhibitor therapeutic candidate, such as tofacitinib.
Despite this theoretically promising approach, Dupilumab, an IL-4RA monoclonal antibody [36], has been demonstrated to be catastrophic in CTCL patients, as stated before [171]. Hashimoto et al. [171] analyzed the expression of IL-13 receptor α2 (IL-13RA2) in the lesional skin of an SS patient who progressed after Dupilumab administration. As a possible explanation for the disease progression, the authors proposed that, despite the action on IL-4 and IL-13RA1, IL-13 may bind to IL-13RA2, leading to the exacerbation and progression of the disease.

15. IL-14

Interleukin-14 (IL-14), also called Alpha-taxilinis or high-molecular-weight B-cell growth, controls B cells’ growth and proliferation. It is produced mainly by T cells and specific malignant B cells [64,172].

IL-14 and MF/SS

IL-14 seems to be higher in effusion fluids from patients with aggressive B-cell lymphomas [173] and in murine models related to the development of solid cancers [174]. A study in mice of the directed expression of IL-14 in CD19+ B cells led to marked splenomegaly and altered spleen morphology at baseline due to the expansion of marginal zone B cells [175].
Yet, its specific role has to be determined, and there are no data related to CTCLs.

16. IL-15 and IL-17

Interleukin 15 (IL-15) is produced by different types of cells, including fibroblasts, keratinocytes, monocytes, macrophages, and dendritic cells, and exerts its main activity when dimerized with the common gamma chain (γc), with the same mechanism as IL-2, IL-4, IL-7, IL-9, and IL-21 on the IL-15 receptor expressed on T, B, and NK cells. IL-15 enhances CD8 T cell cytotoxic activity, B cell differentiation and immunoglobulin synthesis, and dendritic cell maturation. IL-15 shares the same receptor as IL-2 (IL-2R/IL-15Rβ; CD122), leading to JAK1/JAK3/STAT5 pathway activation. Unlike IL-2, IL-15 is not involved in the differentiation of immunosuppressive T-regs [176].
The Interleukin 17 Family (IL-17) encompasses six cytokines, which are IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F (also known as IL-25; please refer to that paragraph for more information), produced by the Th17 subset of CD4+ T cells. Among them, Il-17A is the most described in the literature. IL-17A enhances the immune response against infectious agents by inducing pro-inflammatory cytokine expression (such as TNF, IL-1, and IL-6) and the production of chemokines, metalloproteinases, and antimicrobial peptides from different types of cells, including keratinocytes, fibroblasts, and epithelial cells. Moreover, IL-17 induces the expression of ICAM-1 in keratinocytes [71]. On the other hand, chronic activity of IL-17 is associated with tumorigenesis by cancer cell proliferation, MDSC recruitment, angiogenesis, and autoimmunity [177].

IL-15 and IL-17 in MF/SS

The first studies on IL-15 supposed that such a cytokine may be considered as a survival factor [142,178,179]. IL-15 can stimulate MF cells [67,180], and in MF samples, IL-15 is highly expressed, in contrast to what is observed in normal skin. In MF/SS, IL-15 has been implicated in the recruitment of CD4+ memory T cells to the skin, the induction of T cell proliferation, and the inhibition of apoptotic cell death [65].
Due to a higher expression in advanced CTCL stages, IL-15 was assumed to promote disease progression. However, later studies observed that IL-15 is constitutively expressed in CTCL, with no stage-related expression [181,182]. In a large cohort of patients, Willerslev-Olsen et al. [145] corroborated what had been previously reported by Leroy et al. [181] on IL-15’s heterogeneous expression. They suggested that IL-15 is constitutively expressed in MF/SS and how different IL-15 levels may be related to differences in treatment regimens and microbial infections.
Furthermore, Willerslev-Olsen et al. [66] proposed that the net effect of IL-15 expression may depend on the specific cytokine environment and cellular composition of the skin lesions, thus playing a far more complex role in CTCL pathogenesis than initially thought.
Indeed, IL-15 can exert an antitumor action as a growth and activation factor for non-malignant T cells. Combined with IFNa, IL-15 potentiates CD8 T cells’ and NK cells’ antitumor action and promotes the growth inhibition of malignant T cells [183,184]. On the other hand, in the literature, there is evidence that IL-15, produced by malignant T cells and/or stromal cells, promotes tumor progression via the autocrine and paracrine stimulation of malignant T cells [179]. A possible link between IL-15 and IL-17A expression via the STAT3 pathway was proposed by Dummer et al. [185]. However, Willerslev-Olsen et al. [145], who observed the presence of IL-17A after using IL-15 inhibitors, denied such a connection. In the same analysis, the Danish researchers proposed that IL1–7 may be critical in the pathogenesis of the advanced stages of CTCL [66].
Mishra et al. [69] speculated whether IL-15 might play a pivotal role in MF/SS pathogenesis, in part via the epigenetic inhibition of the transcriptional repressor, Zeb1. Zeb1 inhibition may lead to the overexpression of IL-15 and the activation of specific histone deacetylases (HDAC), opening the door to whether or not a selective inhibition of HDAC1/2 may determine a halt in disease progression. As a consequence, HDAC inhibitors may play a role in MF/SS treatment.
Several investigations have found that IL-17 stimulates angiogenesis (including lymphangiogenesis) and modulates stromal cells’ function, eventually leading to tumor progression [185,186]. The tumorigenic ability of IL-17 was the subject of a recent investigation provided by Papathemeli et al. [187]. In their research, the Greek group observed changes in IL-17 (low expression) in the mRNA levels in MF/SS samples compared to those in healthy donors. The authors speculated whether low levels of IL-17A and IL-17F in mycosis fungoides may be connected to impaired immune surveillance, thereby facilitating tumorigenesis. A specific T-helper cell subset, called TH22 cells, is responsible for IL-22, IL-10, and TNF-α production without IL-17A.

17. IL-16

Interleukin 16 (IL-16) is a chemokine and a modulator of T cell activation, also implied in viral replication inhibition [151]. It is produced by several APC cells, including epidermal cells, and has shown the ability to recruit and activate many other cells expressing CD4, including monocytes, eosinophils, and DCs.
It has been extensively studied for its role in immune response; IL-16 bioactivity has also been associated with the progression of some cancers [64,70].

IL-16 and MF/SS

IL-16′s role in most cancers is still debated, but in CTCLs, it has been proven to promote cell growth by decreasing p27(KIP1) levels, while the overexpression of the secreted IL-16 molecule induces proliferation in CTCL T cells [70]. It seems that intracellular IL-16 levels are linked to a loss of surface CD26, which occurs in the early onset of CTCL stages (with a peak to IB stage). The phenomenon seems to not be irreversible, with IL-16 levels recovered in the late stages of Sézary syndromes [188].
More data about its role in MF/SS are needed.

18. IL-18

Interleukin 18 (IL-18) plays a key role in IFN-γ production and in Th1 and NK cell response. IL-18 is primarily produced by dendritic cells, neutrophils, macrophages, and the epithelium, including keratinocytes. IL-18, together with IL-12 and IL-15, has a role in adaptive immunity due to its ability to induce IFN-γ production via the activation of the NF-kB and STAT-4 pathways. Without IL-12 or IL-15, IL-18 does not induce IFN-γ production, but produces IL-13 and IL-4, promoting Th2 cell differentiation [73,189,190].

IL-18 in MF/SS

IL-18’s role in MF/SS has been the subject of a few studies. Bostan et al. [191] performed an immunohistochemical study on skin biopsies, including MF, pityriasis lichenoides chronica (PLC), and control cases. They demonstrated that, in all stages of MF and PLC, the levels of IL-18 expression were elevated compared to those of control cases, suggesting that the activation of the inflammasome complex and subsequent IL-18 production might play a role in MF pathogenesis [191].
Manfrere et al. [74] studied the levels of inflammasome components, including IL-18, in SS patients. The IL-18 levels were lower in the epidermis and elevated in the dermis of SS patients compared to the control group (composed of healthy donors and patients with idiopathic erythroderma). In SS lymph nodes, IL-18 expression was elevated compared to controls. Similarly, in SS serum, IL-18 and the endogenous inhibitor IL-18-binding protein were elevated in the serum of SS patients. Interestingly, higher IL-18 levels were correlated with a reduced expression of IL-1B. The authors hypothesized that this imbalanced IL-1B and IL-18 expression in SS patients might represent a distinct inflammasome activation pathway leading to tumoral escape to apoptosis [74].

19. IL-19, IL-20, IL-22, IL-24, and IL-26

Interleukins 19, 20, 22 24, and 26 (IL-19, IL-20, IL-22, IL-24, and IL-26) are members of the IL-20 subfamily, which is part of the IL-10 family. These cytokines mainly exert anti-inflammatory and immunosuppressive activities [53,75].
IL-19 regulates T cells’ activity and the production of IL-4, IL-5, IL-10, and IL-13. It is involved in autoimmune and inflammatory diseases such as psoriasis and IBD. IL-19 has a controversial role in monocyte activity, inducing IL-10 production in human monocytes with the M2 phenotype, whereas it induced IL-6 and TNF-α expression in mouse M1 monocytes [192].
IL-20 is involved in the pathogenesis of inflammatory diseases, such as psoriasis, rheumatoid arthritis, and atherosclerosis [193]. Moreover, it is associated with breast cancer cell survival and bladder cancer cell migration [194,195].
IL-22 is a member of the IL-10 family and is produced by NK and CD4 T cells. It is considered as a cancer-promoting cytokine. Indeed, IL-22, after binding IL-22R, leads to STAT3 pathway activation and is related to the progression of different neoplasms, including pancreatic, colon, breast, and lung cancers [115].
IL-24 was originally identified as a tumor-suppressing protein named melanoma differentiation-associated 7 (MDA-7) [196]. Interestingly, IL-24 has unique antitumor activity in lung, breast, and colorectal cancer models [197].
IL-26 was initially named AK155. It is produced by epithelial cells, macrophages, NK cells, Th1, and Th17 cells, and it exerts a pro-inflammatory activity in infections and autoimmune responses [198].

IL-19, IL-20, IL-22, IL-24, and IL-26 in MF/SS

IL-19 expression has been investigated in the advanced stages of CTCL. Senda et al. demonstrated that IL-19 levels were correlated positively with HMGB1, a protein associated with angiogenesis, Th2 polarization, and CTCL progression [76].
Regarding IL-22, it was studied as being related to IL-17A by a Greek group [124]. IL-22 had a relatively high mRNA expression compared to IL-17 MF/SS samples. For the group, the upregulation of IL-22 could play a role in establishing the tumor microenvironment in mycosis fungoides. IL-22 also seems to play an essential role in keratinocyte proliferation, leading to epidermal hyperplasia. A specific T-helper cell subset, called TH22 cells, is responsible for IL-22, IL-10, and TNF-α production without IL-17A. Serum IL-22 levels are positively correlated with serum sIL-2R, LDH, and CCL27 levels, and all the molecules mentioned above are known to be related to disease activity in CTCLs [125,126]. Several reports present in the literature have reported that high levels of IL-22, IL-10, and CCL20 in serum patients are correlated with the advanced stages of CTCLs [127]. Furthermore, it has been proposed that high levels of IL-22, IL-10, and CCL20 may determine, as a cascade event, the overexpression of IL-22–STAT3–CCL20–CCR6 thought to be associated with a tumor’s ability to spread to the lymph nodes and internal organs [128,129].
No data are currently available about the roles of IL-20, IL-24, and IL-26 in MF/SS.

20. IL-21

Interleukin-21 (IL-21) exerts its main activity when dimerized with the common gamma chain (γc), with the exact mechanism of IL-2, IL-4, IL-7, IL-9, and IL-15 on the IL-21 receptor expressed on T, B, and NK cells. Its function is to regulate cell proliferation and activity. It is primarily produced by a subset of T cells known as T follicular helper (Tfh) cells [199].

IL-21 in MF/SS

Regarding its role in tumor genesis, IL-21 has been proven to induce cytotoxic reactions against tumors such as melanoma [199,200] and has been proven to be produced by Hodgkin’s lymphoma (HL) cancer cells (in contrast to what is known in the unaffected healthy human body), making it considered as a possible biomarker [201].

21. IL-22

See the paragraphs on Section 19.

22. IL-23

Interleukin 23 (IL-23) is an inflammatory heterodimeric cytokine composed of subunits IL-12B (in common with IL-12) and IL-23A [79]. Its structure makes it related to IL-12 and included in the heterodimeric cytokines family with Il-12, IL-27, and IL-35 [202].
It is primarily produced by activated antigen-presenting cells (APCs), such as dendritic cells, macrophages, or monocytes, but other immune cells, such as innate lymphoid cells and γδ T cells, can secrete it.
It is vital for T helper type 17 cell (Th17 cell) maintenance and expansion. Polarization to a Th17 phenotype is triggered by IL-6 and TGF-β, which activate the Th17 transcription factor RORγt [79].

IL-23 in MF/SS

Before IL-23 was discovered, IL-12 was thought to be one of the principal causes of inflammatory disease and autoimmunity [203], but after the work of Oppmann et al. [204], a new biological model showed how IL-23 was mainly driven in psoriasis, arthritis, and immuno-bowel diseases.
This led to multiple studies on targeting it and developing drugs such as ustekinumab, guselkumab, risankizumab, and tildrakizumab [205,206].
However, as for IL-12, due to the recent discovery and lack of data, IL-23 in CTCLs is poorly understood. The work of Sugaya et al. [207] showed how CD163+ receptors are related to M2-activated macrophages, which, in contrast to classically activated ones, are not driven by IL-23 and are increased in CTCL lesional skin.
Moreover, as for IL-12 cytokines, a study indicated no evidence of CTCL relation with IL-23 biological therapies [159], despite some case reports [208] and small patient series [209].
Actually, there are no data about the use of IL-23 biological drugs specifically for CTCLs.

23. IL-24

See the paragraphs on Section 19.

24. IL-25

Interleukin 25 (IL-25) belongs to the IL-17 cytokine family and is also known as IL-17E. IL-25 is mainly produced by dendritic cells and exerts its activity on various types of cells, including Th cells. IL-25 leads to Th2 phenotype polarization and IL-4, IL-5, and IL-13 production. Moreover, IL-25 inhibits TH1 and TH17 responses by inhibiting IL-12 and IL-23, respectively. IL-25 is mainly involved in autoimmune diseases, allergic disorders, and parasitic infections, but its role in different types of cancers has recently come under investigation [80,210].

IL-25 in MF/SS

IL-25’s role in MF/SS has been investigated. Nakajima et al. demonstrated that IL-25 expression was higher in MF and SS epidermal keratinocytes than in normal controls. IL-25 production is stimulated by IL-4 and IL-13 cytokines—produced by tumor cells and Th2 cells—and periostin produced by dermal fibroblasts. Moreover, the IL-25 levels in skin lesions and sera are correlated with disease progression, and IL-25 serum levels are correlated with LDH levels. IL-25 enhanced IL-13 production from tumor cells via STAT6 signaling pathways, resulting in the augmentation of a Th2-dominant microenvironment [211].

25. IL-26

See the paragraphs on Section 19.

26. IL-27

Interleukin 27 (IL-27) is a member of the IL-6/IL-12 family and regulates the immune response. IL-27 promotes Th1 immunity and IFN-γ production by NK and T cells [212] and inhibits Th2 response [82,190]. IL-27 seems to play a controversial role in cancer immunity. IL-27 promotes antitumor immunity by NK, NKT, and CD8+ T cells [213,214]. On the other hand, IL-27Rα expression in different tumor cell lines is associated with the inhibition of effector responses and the promotion of tumor growth [215].

IL-27 in MF/SS

Miyagaki et al. analyzed the serum levels of IL-27 in MF and SS patients. They observed that the IL-27 levels were higher in advanced stages compared to early stages or controls. However, the IL-27 serum level of patients with advanced-stage CTCL was inversely correlated with the number of eosinophils in peripheral blood. The authors hypothesized that IL-27 might be produced by microenvironment cells as a response to neoplastic cells to reduce Th2 immunity, typically associated with CTCL progression [129].

27. IL-28 and IL-29

Interleukin-28 (IL-28) presents two isoforms, IL-28A and IL-28B, belonging to the amino acid sequence and functions of the type III interferon family of cytokines, which are highly similar (in amino acid sequence) to IL-29 [54,97].
For their descriptions, refer to the paragraph on interferons.

28. IL-30

Interleukin-30 (IL-30) is a component of the IL-27 cytokine family, often associated with modulating immune responses. IL-30 is known for its potential role in regulating inflammation and immune reactions. However, its specific physiological functions are still being investigated [81].

IL-30 in MF/SS

To date, IL-30 has not been extensively studied or linked explicitly to MF. However, IL-30’s physiological role suggests a potential involvement in modulating immune responses and inflammation, which could have implications for various skin conditions.

29. IL-31 and IL-8

Interleukin 31 (IL-31) is mainly produced after IL-4 stimulation by CD4+ Th2 helper cells, mast cells, and dendritic cells and exerts its activity on fibroblasts and eosinophils. IL-31 binds the IL-31 receptor (IL-31R), leading to JAK1/JAK2 and STAT3 signaling. IL-31 has a prominent role in pruritus pathogenesis in atopic dermatitis, prurigo nodularis, and CTCL [30,216,217].
Interleukin 8 (IL-8), also called CXCL8, is a chemokine produced by macrophages, epithelial cells, and airway smooth muscle cells. IL-8 is a chemotactic factor for neutrophils and other granulocytes. IL-8 exerts its oncogenic role by binding the IL-8 R localized on cancer cells and microenvironment cells (including TAM and neutrophils), leading to neo-angiogenesis and an enhancement in metastatic potential in pancreatic and bladder cancer [218,219].

IL-31 and IL-8 in MF/SS

Several studies have investigated the pathogenetic role of IL-31 in CTCL [85,125,220]. IL-31 is secreted by Th-2 cells after IL-4 stimulation [221] and by malignant T cells [83]. In CTCL patients, increased IL-31 levels have been detected in both sera and skin lesions, and seem to be correlated with increased pruritus [216]. However, no univocal correlation has been established between IL-31 levels and disease progression or pruritus severity [83,220,222].
IL-8 is a chemotactic cytokine that is not directly involved in the pathogenesis of pruritus in CTCL [47]. However, several studies have described high IL-8 levels in pruritic dermatitis, such as atopic dermatitis [48]. In a study by Abreu et al. [49], IL-8 serum levels were similar in MF patients and healthy controls, suggesting that IL-8, per se, is not a key mediator of itch in MF/SS patients. However, owing to IL-8′s chemotactic action for neutrophils, one can speculate whether IL-8 may take part in exacerbating itch by recruiting and activating neutrophil cells. Kadin et al. [42] also proposed that high IL-8 levels may be a marker of worsened clinical outcomes in MF. Indeed, the American group found that IL-8 serum levels were correlated with CD30 serum levels. As a consequence, IL-8 may be secreted by CD30+ neoplastic cells, causing a general state of immune activation predisposing to MF progression [223]. Hence, IL-8 should be regarded as a marker of MF progression.

30. IL-32

Interleukin 32 (IL-32) is encoded by one gene, but has nine different isoforms because of the alternative mRNA splicing [224,225].
The most studied isoform is IL-32γ, and it does not belong to any cytokine family because it has almost no similar structure nor function to other cytokines.
Some isoforms seem to be related and are found in cancer cells [226,227], fostering tumor progression stimulating (NF-κB)-mediated cytokines and metalloproteinase production, as well as the stimulation of differentiation into immunosuppressive cell types in some cancer types [228]. For example, IL-32γ and IL-32β expression are associated with increased cancer cell death in colon cancer and melanoma, whereas the expression of these isoforms is associated with increased invasion and migration in breast cancer cells [224,229].
Also, the IL-32 isoforms α, β, and γ also play essential roles in regulating the anti-tumor immune response [230].

IL-32 in MF/SS

Van Kester et al.’s study showed how MF may be induced by IL-32-producing cells [231], while other groups showed IL-32 to have a progressive role as an independent factor unrelated to Th2 differentiation and autocrine stimulating factor in MF and SS cells in lesional skin [232,233]. IL-32 mRNA levels have been found to increase in tumoral samples, and, when cultured, tumor viability has been impaired by adding anti-IL-32 antibodies, confirming the potential effect of IL-32 on MF and SS viability and progression [86,232].
IL-32 was recently discovered, and there are no data regarding a potential targeted therapy such as humanized immunoglobulins for cancer or other diseases.

31. IL-33

Interleukin-33 (IL-33) is a member of the IL-1 family and acts like an alarmin because it induces an inflammatory response after skin damage. By binding to its receptor ST2, IL-33 can polarize macrophages into M2 macrophages, induce the maturation of dendritic cells, and promote Th1-mediated responses, including cell-mediated cytotoxicity [87,234] [Kurowska-Stolarska M J Immunol 2009; Liew FY Nat Rev Immunol 2016]. IL-33/ST2 signaling in sensory neurons mediates pruritus, a symptom shared by many pathological conditions of the skin. IL-33 is highly expressed in the skin and increases in inflammatory skin lesions in atopic dermatitis, psoriasis, and scleroderma [235]. In atopic dermatitis patients, high IL-33 serum levels are positively correlated with the severity of atopic dermatitis [235]. IL-33 is also implicated in allergy and asthma development [88]. IL-33 is also thought to be related to the promotion of local progression and metastasizing of myeloproliferative neoplasms and gastric, colon, and breast cancer [236].

IL-33 in MF/SS

Rustowska-Rogowska et al. [237] proposed that IL-33/ST2 signaling may be implicated in the pathogenesis of MF. Although MF patients had similar serum IL-33 concentrations as controls and IL-33 concentrations were unrelated to the MF stage, the authors hypothesised that high IL-33 levels may be confined to the skin and rapidly degraded by proteasomes. Hence, IL-33 levels may appear normal at the blood level, but not reveal increased IL-33 production in the skin of MF patients [89]. Therefore, IL-33 might accelerate MF progression via a paracrine action in the tumor microenvironment, like in patients with myeloproliferative syndromes [90]. Rustowska-Rogowska et al.’s [237] conclusion was that further studies should investigate IL-33’s expression in lesional skin.

32. TNF-α

TNF-α is a naturally occurring cytokine involved in normal inflammatory and immune responses. It plays an essential role in the pathogenesis of various hematologic malignancies, such as multiple myeloma, myelodysplastic syndrome, acute myelogenous leukaemia, lymphoproliferative disorders, and conditions such as graft-versus-host disease [238,239].

TNF-α in MF/SS

Tumor necrosis factor alpha (TNF–α) has been implicated in the development of CTCL by the promotion of epidermotropism via the induction of interferon-inducible protein (IP-10) [238,240]. In addition, TNF-a acts as an autocrine growth factor, enhancing its tumorigenic action and empowering the NF-κB pathway [241]. In cell lines, anti-TNF-α antibodies downregulate CTCL cell growth, as well as NF-κB overactivation. As a result, TNF-a plays a complex role in CTCL pathogenesis along with other cytokines, such as IL-7, IL-12, and 15, by allowing neoplastic cells to gain advantages against immune system antitumor action. TNF-a is also overexpressed in psoriasis, creating a link in etiologic mechanisms between psoriasis and CTCL, particularly a defect in the mechanisms involved in inducing cell death. The availability of anti-TNF-a drugs in psoriasis has given some hope in CTCL treatment. To date, no conclusions can be drawn in patients treated with anti-TNF- α with a misdiagnosed or concomitant MF/SS, owing to the presence in the literature of MF/SS cases unmasked by the use of anti-TNF-α, cases that progressed after biological treatment administration, and cases without clinical progression after anti-TNF-α treatment [242,243,244,245,246,247].
However, extreme caution before starting anti-TNF-α treatment, as well as supplementary biopsies in case of worsening after the beginning of the treatment, has been suggested by all the authors.

33. EGF

Epidermal Growth Factor (EGF) is a polypeptide produced in different tissues and was first discovered in the mouse salivary gland, but in the human body, it can be found in platelets. It is also in the urine, saliva, milk, tears, and blood plasma [248,249]. It can also be found in the submandibular glands and the parotid gland. Its mediator and receptors are modulated by cellular stress and injury mediators (such as TNF-α) [91], hormones such as estrogens [92], other growth factors (e.g., TGF-β) [250]), and environmental factors.
It is studied for causing epidermal proliferation, modulating cellular activities, cell survival, and general tissue repair [248]. EGF’s biological activities depend upon its binding to a specific cell membrane receptor, which leads to a cell cycle progression effect on most epithelial tissues, fibroblasts, and endothelial cells [251].

EGF in MF/SS

Only one article in the literature described a hypothetical but unproven therapeutic relationship between EGF and CTCLs, which claimed that MF develops in tissues with IFN type 1 deficiency. Since EGF has been proven to impair IFN production, it is speculated that reducing EFG functions may alter disease progression [95].

34. FGF

Fibroblast growth factors (FGF) regulate a broad spectrum of biological functions, including cellular proliferation, survival, migration, and differentiation. Alterations in FGF–FGF receptor expression, localization, and signaling have been implicated in several pathological processes, including malignant transformation, tumor spread, and metastasis [93,252].

FGF in MF/SS

Little is known about the role of FGF in MF and SS. Queen et al. hypothesized the roles of FGF and TGF-β in paraneoplastic scleroderma pathogenesis in a patient affected by MF with CD30+ large cell transformation. FGF and TGF-β stimulate the differentiation of mesenchymal stem cells into myofibroblasts, leading to fibrotic pathway activation in MF patients [253,254].

35. PDGR

Platelet-derived growth factors (PDGFs) and their receptor (platelet-derived growth factor alpha or PDGFRα) activate the intracellular signaling pathway through tyrosine kinases (RTKs). They play critical roles in various cellular processes, particularly in cell growth, proliferation, and differentiation. Upon activation, they trigger signaling cascades that regulate cell survival, migration, and tissue repair [94,255]. Dysregulation or mutations in PDGFRα signaling have been implicated in several diseases, including certain cancers, where aberrant activation of this receptor can contribute to uncontrolled cell growth and tumor progression. PDGFRα remains a significant target for therapeutic interventions, and drugs that inhibit its activity are utilized to treat certain cancers and other conditions driven by abnormal PDGFRα signaling [256,257].

PDGRα in MF/SS

PDGFRα is primarily known for its involvement in various cancers, especially gastrointestinal stromal tumors (GISTs), where mutations or dysregulation of PDGFRα play a significant role [139]. However, in MF/SS, the specific contribution or involvement of PDGFRα remains unexplored and far from beinb understood.

36. Interferon Type I, Type II, and Type III

Interferon alpha (IFN-α), interferon beta (IFN-β), which are included in the interferons type I family, interferon-gamma (IFN-γ), representing interferon type II, and the recent family of interferon type III (IFN-λ) [97] consist of cytokines produced as part of the innate immune response, and their immunomodulatory actions have been used in the treatment of several malignancies, including CTCL [258].
IFN-α is produced mainly by plasmacytoid dendritic cells (pDCs), enhancing the immune system response. Specifically, IFN- α has been found to stimulate CD8+ T cells and NK cells, thereby activating antitumor cytotoxicity [20,138,259]. Moreover, IFN- α may upregulate MHC class I expression and blunt the excess TH2 production of IL-4 and IL-5, restoring the host TH1/TH2 balance.
IFN-β, notably regulated and synthesized by fibroblasts, is known for its antiviral activity. In the human body, IFN-β is primarily produced by fibroblasts and other immune cells, including (pDCs), in response to viral infections or other immune triggers [260]. For therapeutic purposes, recombinant IFN-β produced using biotechnological methods is mainly used to treat multiple sclerosis [261,262].
The physiological role of IFN-γ is to orchestrate cellular immunity against infections and tumor surveillance. IFN-γ enhances the antigen recognition of antigen-presenting cells (APCs) and increases the production of ROS and NOS in macrophages [96]. Moreover, IFN-γ regulates humoral immunity by B-cell proliferation and antibody class switching.
IFN-γ receptor (IFNγR) can present in two subunits, IFNγR1 and IFNγR2, that interact with JAK1 and JAK2, respectively, leading to STAT1 activation [98,263]. IFN-γ plays a central role in all the phases of “cancer immunoediting” [264], showing antitumor and tumorigenic effects. In the elimination phase, IFN-γ promotes antigen presentation, CD4+ T cell polarization to Th1, and the maturation of CD8+ T cells. In the escape phase, INF-gamma induces PD-L1 up-regulation on cancerous cells, inhibiting antitumor CD8+ T cell cytotoxicity and enhancing tumor cell survival [265,266].
IFN- λ is a relatively recent family of anti-viral cytokines consisting of four molecules, which include IFN-λ1, IFN-λ2, IFN-λ3 (also known as IL29, IL28A, and IL28B, respectively), and IFN-λ4. Its function seems to be similar to the IFN type I family, although less intense [267].

Interferon Type I, Type II, and Type III in MF/SS

IFN-α presents an antitumor action by empowering CD8+ and NK cell action against MF cells. In addition, high IFN-alpha levels decrease the Th2 cytokinetic milieu and IL-4/IL-13 overexpression. IFN-α in different formulations has been used in MF/SS treatment, but, owing to these different formulations and the absence of prospective clinical trials, no definitive conclusions on IFN-α’s efficacy can be drawn [258,268].
A brief mention is made of IFN-β, which little is known about in the field of cutaneous lymphomas. In medical databases, it has only been the subject of small studies, mainly with small and retrospective series [260,269].
IFN-γ’s role has thoroughly been investigated in MF. Sarris et al. studied the underlying mechanisms of the epidermotropism pathogenesis in CTCL and demonstrated the cytokine loop between INF-gamma, produced by CD4+ lymphocytes, and IFN -gamma inducible protein 10 (IP-10), produced by keratinocytes. The study demonstrated the pivotal role of INF-γ in CTCL pathogenesis and Pautrier’s microabcess formations. Moreover, INF-γ and IP-10 were associated with the early stages of MF with marked epidermotropism and exclusive skin involvement. On the other hand, in SS, the level of INF-γ was lower and the Th2 response was prominent [270]. Asadullah et al. demonstrated that the overexpression of EL-10 mRNA leads to MF progression by the suppression of IFN-γ production. IFN-γ a was higher in the patch stage and decreased in a stage-dependent manner in advanced stages when epidermotropism was lost [271]. Further studies corroborated such a finding and revealed that monokine induced by IFN -γ contributes to epidermotropism, providing further evidence on the role of IFN-γ in early-stage pathogenesis [99]. Sigurdsson et al. studied the expressions of IFN γ and IL-4 in dermal infiltrate in patients with inflammatory skin disease, MF, and SS, revealing a higher expression of INF-γ in inflammatory skin disease and MF than SS [28].
Several groups have investigated the therapeutic role of recombinant IFN-γ a in monotherapy [245,272] or in association with other therapies, such as phototherapy, bexarotene, vorinostat, and IFN-α, providing no clear-cut results due to the small sample sizes of the studies [100,273,274,275].
The literature lacks information regarding IFN- λ and its relation to CTCLs, probably due to its recent discovery and limited knowledge.

37. Discussion

MF/SS are fascinating diseases owing to their intrinsic nature. Indeed, each clinical presentation has a peculiar pathogenesis [276], histology [2,277,278], and stage-related treatment [279,280,281,282]. Indeed, from a histological point of view, the diseases are characterized by neoplastic T cells resembling their regular counterparts surrounded by reactive T cells along with dendritic cells, histiocytes, and macrophages. Furthermore, in a hematologic setting, it is exceptional that lymphomas warrant skin-directed therapy as a treatment, not a systemic one (concerning the early stages). However, the mechanisms involved in the progression from early indolent stages to aggressive ones are still far from being understood. Several players are involved and connected, such as gene alterations, cytokines, and microenvironment cells [11,12,72,78,283,284,285,286,287,288,289].
Cytokines are a potential target of treatment and response to administered therapies. Indeed, Tsai YC et al. highlighted that a switch from a Th2 to a Th1 cytokine pattern is related to a response to extracorporeal photopheresis, stressing how the interaction between neoplastic cells and the microenvironment realized by the production of different cytokine patterns is meaningful [5,19]. The administration of IFN-α, a Th1 cytokine, is a well-known efficient treatment in advanced-stage CTCL [100,272,273,274], and its therapeutic action, at least in part, is related to restoring the host’s immune system action against the disease. Other cytokines that may be potential targets have been revealed to be harmful to patients, such as the IL-4/IL-13 inhibitor Dupilumab [34,132,133,171]. As mentioned above, the administration of Dupilumanb in misdiagnosed MF/SS had the opposite action on patients, with the development of a more aggressive disease. Caution on biologic treatment should be taken, especially for cases of non-conventional atopic dermatitis or psoriasis (another disease that can be treated with biologic therapies).
Anti-IL-17 is potentially tumorigenic in misdiagnosed MF/SS treated as “recalcitrant” psoriasis [66,185,186]. Although cases without progression have been described, anti-TNF-α treatment [231,232,233,234,235,236] should also be managed with extreme caution for the risk of developing or unmasking a CTCL. Such evidence in the literature has left the door open to the need for a clear-cut algorithm for patients eligible for biologic therapies. The restoration from a Th2 to a Th1 pattern is also one of the significant goals of the newly available drugs such as Mogamulizumab, Brentuximab Vedotin, and Chlormetine and molecules currently under clinical trial such as Pi3K inhibitors [5,127,290,291,292,293]. From the present paper reviewing the literature, it has emerged that cytokines can be a reliable marker of some tumorigenic pathway activity, such as JAK/STAT MAPK, and the suppression of those tumorigenic pathways may be monitored by analyzing cytokine composition changes in patients. The reawakening of the immune system and the empowerment of its antitumor action, along with the apoptosis of neoplastic T cells, are major targets of the currently available drugs and the candidate ones currently under investigation. In this light, interferon drugs should be regarded as the immunotherapy pioneer in MF/SS.
Other drugs are currently being investigated, such as Bruton’s tyrosine kinase inhibitor (Ibrutinib), which was hypothesized to decrease signaling through the T cell receptor pathway and promoting the antitumor immune response by driving selective cytotoxic Th1 CD4 effector T cell differentiation. However, a recent phase II trial [294] demonstrated limited activity on selected patients. Trials on IL-2 fusion toxin have demonstrated anti-tumor activity both as a single agent and in combination with other drugs [25,268,295]. However, the drug was not well-tolerated and its use has been limited. Due to the overexpression of the JAK3/STAT5 pathway and its cytokine expression in MF/SS [170,296,297,298], one can speculate whether JAK inhibitors may be considered as a possible MF/SS treatment. To date, limited data are available, including a case report [299] and a phase II trial [300], and no conclusions can be drawn (Figure 1).
Hypothetically, a blockade of the JAK3/STAT5 pathway should lead to the inhibition of immunosuppressive cytokines downstream. However, the lack of data demands extreme caution on JAK inhibitor administration. From the present review, it emerges that MF/SS treatment needs to be delivered at different levels, and one of them is to control the cytokine balance, with a reduction in immunosuppressive cytokines and an increase in antitumor ones.

Author Contributions

Conceptualization: A.G. and A.P.; data curation: A.G. and A.P.; formal analysis: C.Z., C.A., E.S., G.M. and A.P.; investigation: A.P.; methodology: C.Z. and A.P.; resources: A.P.; software: A.P.; supervision: C.A.; validation: A.G. and A.P.; visualization: A.G., C.A., E.S. and A.P.; writing—original draft: A.G. and A.P.; writing—review and editing: A.G., C.Z. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available upon request from the authors.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

APCAntigen-Presenting Cell
CDCluster of Differentiation
CCLC-C Motif Chemokine Ligand
CCRC-C Motif Chemokine Receptor
CTCLCutaneous Cutaneous T cell lymphoma
CXCLC-X-C Motif Chemokine Ligand
EGFEpdermal Growth Gactors
FGFFibroblast Growth Factors
JAKJAnus Kinase
GABGRB2-associated-binding protein
HDACHistone Deacetylases
HMGBHigh Mobility Group Box protein
ICAM-1Intercellular Adhesion Molecule 1
IFNInterferon
ILInterLeukin
IPInterferon-Inducible Protein
ITImmunoToxin
LDHLactate DeHydrogenase
MAPKMitogen-Activated Protein Kinase
MDA7Melanoma Differentiation-Associated protein 7
MFMycosis fungoides
nbUVBnarrow band Ultra Violet type B
NOSNitrogen Oxygen Species
PBMCPeripheral Blood Mononuclear Cell
PI3KsPhosphoinositide 3-kinase inhibitors
PDGFPlatelet-Derived Growth Factors
PUVAPsoralen and Ultra Violet type A
ROSReactive Oxygen Species
SATB1Special AT-rich sequence-Binding protein-1
STATSignal Transducer and Activator of Transcription
SSSézary syndrome
TAMTumor-Associated Macrophage
TCGFT cell growth factor
TfhT follicular helper cell
TGFTransforming Growth Factor
TNFTumor Necrosis Factor
Th(Lymphocyte) T helper (Cell)
T-Regs(lymphocyte) T-regulatory cells
NKNatural Killer (cell)
γcCommon gamma chain

References

  1. Willemze, R.; Cerroni, L.; Kempf, W.; Berti, E.; Facchetti, F.; Swerdlow, S.H.; Jaffe, E.S. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood 2019, 133, 1703–1714. [Google Scholar] [CrossRef]
  2. Pileri, A.; Patrizi, A.; Agostinelli, C.; Neri, I.; Sabattini, E.; Bacci, F.; Piccaluga, P.P.; Pimpinelli, N.; Pileri, S.A. Primary cutaneous lymphomas: A reprisal. Semin. Diagn. Pathol. 2011, 28, 214–233. [Google Scholar] [CrossRef] [PubMed]
  3. Criscione, V.D.; Weinstock, M.A. Incidence of cutaneous T-cell lymphoma in the United States, 1973–2002. Arch. Dermatol. 2007, 143, 854–859. [Google Scholar] [CrossRef] [PubMed]
  4. Dobos, G.; de Masson, A.; Ram-Wolff, C.; Beylot-Barry, M.; Pham-Ledard, A.; Ortonne, N.; Ingen-Housz-Oro, S.; Battistella, M.; d’Incan, M.; Rouanet, J.; et al. Epidemiological changes in cutaneous lymphomas: An analysis of 8593 patients from the French Cutaneous Lymphoma Registry. Br. J. Dermatol. 2021, 184, 1059–1067. [Google Scholar] [CrossRef]
  5. Pileri, A.; Morsia, E.; Zengarini, C.; Torre, E.; Goteri, G.; Quaglino, P.; Pimpinelli, N.; Paulli, M.; A Pileri, S.; Zinzani, P.L.; et al. Epidemiology of cutaneous T-cell lymphomas: State of the art and a focus on the Italian Marche region. Eur. J. Dermatol. 2023, 33, 360–367. [Google Scholar] [CrossRef] [PubMed]
  6. Kayishunge, D.; Ly, S.; Su, J.; Wong, H.K. Epidemiologic Trends of Cutaneous T-Cell Lymphoma in Arkansas Reveals Demographic Disparities. Cancers 2022, 14, 4329. [Google Scholar] [CrossRef]
  7. Miyashiro, D.; Sanches, J.A. Mycosis fungoides and Sézary syndrome: Clinical presentation, diagnosis, staging, and therapeutic management. Front. Oncol. 2023, 13, 1141108. [Google Scholar] [CrossRef] [PubMed]
  8. Dai, J.; Duvic, M. Cutaneous T-Cell Lymphoma: Current and Emerging Therapies. Oncology 2023, 37, 55–62. [Google Scholar] [CrossRef] [PubMed]
  9. Wojewoda, K.; Gillstedt, M.; Englund, H.; Ali, S.; Lewerin, C.; Osmancevic, A. Diagnostic Outcomes and Treatment Modalities in Patients with Mycosis Fungoides in West Sweden—A Retrospective Register-Based Study. Cancers 2022, 14, 4661. [Google Scholar] [CrossRef]
  10. Pileri, A.; Guglielmo, A.; Grandi, V.; Violetti, S.A.; Fanoni, D.; Fava, P.; Agostinelli, C.; Berti, E.; Quaglino, P.; Pimpinelli, N. The Microenvironment’s Role in Mycosis Fungoides and Sézary Syndrome: From Progression to Therapeutic Implications. Cells 2021, 10, 2780. [Google Scholar] [CrossRef] [PubMed]
  11. Alberti-Violetti, S.; Sapienza, M.R.; Del Corvo, M.; Melle, F.; Motta, G.; Venegoni, L.; Cerroni, L.; Cota, C.; Pileri, A.; Berti, E.; et al. A Microenvironment-Related Nine-Gene Signature May Predict Survival in Mycosis Fungoides Patients at Diagnosis. Cells 2023, 12, 1944. [Google Scholar] [CrossRef] [PubMed]
  12. Dobos, G.; Lazaridou, I.; de Masson, A. Mycosis Fungoides and Sézary Syndrome: Microenvironment and Cancer Progression. Cancers 2023, 15, 746. [Google Scholar] [CrossRef]
  13. Johnson, V.E.; Vonderheid, E.C.; Hess, A.D.; Eischen, C.M.; McGirt, L.Y. Genetic Markers Associated with Progression in Early Mycosis Fungoides. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 1431–1435. [Google Scholar] [CrossRef] [PubMed]
  14. Wobser, M.; Roth, S.; Appenzeller, S.; Houben, R.; Schrama, D.; Goebeler, M.; Geissinger, E.; Rosenwald, A.; Maurus, K. Targeted Deep Sequencing of Mycosis Fungoides Reveals Intracellular Signaling Pathways Associated with Aggressiveness and Large Cell Transformation. Cancers 2021, 13, 5512. [Google Scholar] [CrossRef]
  15. McGirt, L.Y.; Jia, P.; Baerenwald, D.A.; Duszynski, R.J.; Dahlman, K.B.; Zic, J.A.; Zwerner, J.P.; Hucks, D.; Dave, U.; Zhao, Z.; et al. Whole-genome sequencing reveals oncogenic mutations in mycosis fungoides. Blood 2015, 126, 508–519. [Google Scholar] [CrossRef] [PubMed]
  16. Gan, L.; Shi, H.; Zhang, Y.; Sun, J.; Chen, H. Proteomic Screening and Verification of Biomarkers in Different Stages of Mycosis Fungoides: A pilot Study. Front. Cell Dev. Biol. 2021, 9, 747017. [Google Scholar] [CrossRef] [PubMed]
  17. Dinarello, C.A. Immunological and inflammatory functions of the interleukin-1 family. Annu. Rev. Immunol. 2009, 27, 519–550. [Google Scholar] [CrossRef]
  18. Wautier, J.-L.; Wautier, M.-P. Pro- and Anti-Inflammatory Prostaglandins and Cytokines in Humans: A Mini Review. Int. J. Mol. Sci. 2023, 24, 9647. [Google Scholar] [CrossRef]
  19. Tsai, Y.-C.; Schlaepfer, T.; Ignatova, D.; Chang, Y.-T.; Valaperti, A.; Amarov, B.; Blanchard, G.; Pehr, K.; Vonow-Eisenring, M.; Urosevic-Maiwald, M.; et al. Boost of innate immunity cytokines as biomarkers of response to extracorporeal photopheresis in patients with leukaemic cutaneous T-cell lymphoma. Br. J. Dermatol. 2023, 189, 603–611. [Google Scholar] [CrossRef] [PubMed]
  20. Gottlieb, S.L.; Wolfe, J.T.; Fox, F.E.; DeNardo, B.J.; Macey, W.H.; Bromley, P.G.; Lessin, S.R.; Rook, A.H. 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. [Google Scholar] [CrossRef] [PubMed]
  21. Malek, T.R.; Bayer, A.L. Tolerance, not immunity, crucially depends on IL-2. Nat. Rev. Immunol. 2004, 4, 665–674. [Google Scholar] [CrossRef] [PubMed]
  22. Brockdorff, J.L.; Gu, H.; Mustelin, T.; Kaltoft, K.; Geisler, C.; Röpke, C.; Ødum, N. Gab2 is phosphorylated on tyrosine upon interleukin-2/interleukin-15 stimulation in mycosis-fungoides-derived tumor T cells and associates inducibly with SHP-2 and Stat5a. Exp. Clin. Immunogenet. 2001, 18, 86–95. [Google Scholar] [CrossRef] [PubMed]
  23. Shohat, M.; Hodak, E.; Sredni, B.; Shohat, B.; Sredni, D.; David, M. Cytokine profile of patients with mycosis fungoides and the immunomodulatory effect of AS101. Acta Derm. Venereol. 2001, 81, 255–257. [Google Scholar] [CrossRef] [PubMed]
  24. Kozenitzky, L.; David, M.; Sredai, B.; Albeck, M.; Shohat, B. Immunomodulatory effects of AS101 on interleukin-2 production and T-lymphocyte function of lymphocytes treated with psoralens and ultraviolet A. Photodermatol. Photoimmunol. Photomed. 1992, 9, 24–28. [Google Scholar] [PubMed]
  25. Foss, F.M. Interleukin-2 Fusion Toxin: Targeted Therapy for Cutaneous T Cell Lymphoma. Ann. N. Y. Acad. Sci. 2001, 941, 166–176. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, Z.; Ma, J.; Zhang, H.; Ramakrishna, R.; Mintzlaff, D.; Mathes, D.W.; Pomfret, E.A.; Lucia, M.S.; Gao, D.; Haverkos, B.M.; et al. CCR4-IL2 bispecific immunotoxin is more effective than brentuximab for targeted therapy of cutaneous T-cell lymphoma in a mouse CTCL model. FEBS Open Bio 2023, 13, 1309–1319. [Google Scholar] [CrossRef] [PubMed]
  27. Dougan, M.; Dranoff, G.; Dougan, S.K. GM-CSF, IL-3, and IL-5 Family of Cytokines: Regulators of Inflammation. Immunity 2019, 50, 796–811. [Google Scholar] [CrossRef] [PubMed]
  28. Sigurdsson, V.; Toonstra, J.; Bihari, I.C.; Bruijnzeel-Koomen, C.A.; van Vloten, W.A.; Thepen, T. Interleukin 4 and interferon-gamma expression of the dermal infiltrate in patients with erythroderma and mycosis fungoides. An immuno-histochemical study. J. Cutan. Pathol. 2000, 27, 429–435. [Google Scholar] [CrossRef] [PubMed]
  29. Noben-Trauth, N.; Paul, W.E.; Sacks, D.L. IL-4- and IL-4 receptor-deficient BALB/c mice reveal differences in susceptibility to Leishmania major parasite substrains. J. Immunol. 1999, 162, 6132–6140. [Google Scholar] [CrossRef] [PubMed]
  30. Murdaca, G.; Greco, M.; Tonacci, A.; Negrini, S.; Borro, M.; Puppo, F.; Gangemi, S. IL-33/IL-31 Axis in Immune-Mediated and Allergic Diseases. Int. J. Mol. Sci. 2019, 20, 5856. [Google Scholar] [CrossRef] [PubMed]
  31. Junttila, I.S. Tuning the Cytokine Responses: An Update on Interleukin (IL)-4 and IL-13 Receptor Complexes. Front. Immunol. 2018, 9, 888. [Google Scholar] [CrossRef] [PubMed]
  32. Saulite, I.; Ignatova, D.; Chang, Y.-T.; Fassnacht, C.; Dimitriou, F.; Varypataki, E.; Anzengruber, F.; Nägeli, M.; Cozzio, A.; Dummer, R.; 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, 1738797. [Google Scholar] [CrossRef]
  33. Ritlecitinib in CTCL. Available online: https://ctv.veeva.com/study/ritlecitinib-in-ctcl (accessed on 10 January 2024).
  34. Beck, L.A.; Thaçi, D.; Hamilton, J.D.; Graham, N.M.; Bieber, T.; Rocklin, R.; Ming, J.E.; Ren, H.; Kao, R.; Simpson, E.; et al. Dupilumab Treatment in Adults with Moderate-to-Severe Atopic Dermatitis. N. Engl. J. Med. 2014, 371, 130–139. [Google Scholar] [CrossRef] [PubMed]
  35. Buffon, S.; Alberti Violetti, S.; Avallone, G.; Venegoni, L.; Marzano, A.V.; Mastorino, L.; Fava, P.; Ribero, S.; Quaglino, P.; Ortoncelli, M.; et al. Mycosis fungoides and Sézary syndrome following dupilumab treatment: Experience of two Italian tertiary care centres. Clin. Exp. Dermatol. 2023, 48, 1376–1378. [Google Scholar] [CrossRef] [PubMed]
  36. Harb, H.; Chatila, T.A. Mechanisms of Dupilumab. Clin. Exp. Allergy 2020, 50, 5–14. [Google Scholar] [CrossRef] [PubMed]
  37. Pelaia, C.; Paoletti, G.; Puggioni, F.; Racca, F.; Pelaia, G.; Canonica, G.W.; Heffler, E. Interleukin-5 in the Pathophysiology of Severe Asthma. Front. Physiol. 2019, 10, 1514. [Google Scholar] [CrossRef] [PubMed]
  38. Nielsen, M.; Nissen, M.H.; Gerwien, J.; Zocca, M.-B.; Rasmussen, H.M.; Nakajima, K.; Röpke, C.; Geisler, C.; Kaltoft, K.; Ødum, N. Spontaneous interleukin-5 production in cutaneous T-cell lymphoma lines is mediated by constitutively activated Stat3. Blood 2002, 99, 973–977. [Google Scholar] [CrossRef] [PubMed]
  39. Yamamoto, T.; Katayama, I.; Nishioka, K. Expression of stem cell factor in the lesional skin of systemic sclerosis. Dermatology 1998, 197, 109–114. [Google Scholar] [CrossRef] [PubMed]
  40. Watson, C.; Whittaker, S.; Smith, N.; Vora, A.J.; Dumonde, D.C.; Brown, K.A. IL-6 acts on endothelial cells to preferentially increase their adherence for lymphocytes. Clin. Exp. Immunol. 1996, 105, 112–119. [Google Scholar] [CrossRef] [PubMed]
  41. Kadin, M.E.; Pavlov, I.Y.; Delgado, J.C.; Vonderheid, E.C. High soluble CD30, CD25, and IL-6 may identify patients with worse survival in CD30+ cutaneous lymphomas and early mycosis fungoides. J. Investig. Dermatol. 2012, 132, 703–710. [Google Scholar] [CrossRef] [PubMed]
  42. Kadin, M.E. What Cytokines Can Tell Us About the Pathogenesis of Breast Implant-Associated Anaplastic Large Cell Lymphoma (BIA-ALCL). Aesthetic Surg. J. 2019, 39, S28–S35. [Google Scholar] [CrossRef] [PubMed]
  43. Olszewska, B.; Gleń, J.; Zabłotna, M.; Nowicki, R.J.; Sokołowska-Wojdyło, M. The polymorphisms of IL-6/STAT3 signaling pathway may contribute to cutaneous T-cell lymphomas susceptibility. Arch. Dermatol. Res. 2021, 313, 25–31. [Google Scholar] [CrossRef] [PubMed]
  44. Asao, H.; Okuyama, C.; Kumaki, S.; Ishii, N.; Tsuchiya, S.; Foster, D.; Sugamura, K. Cutting Edge: The Common γ-Chain Is an Indispensable Subunit of the IL-21 Receptor Complex1. J. Immunol. 2001, 167, 1–5. [Google Scholar] [CrossRef] [PubMed]
  45. Winer, H.; Rodrigues, G.O.L.; Hixon, J.A.; Aiello, F.B.; Hsu, T.C.; Wachter, B.T.; Li, W.; Durum, S.K. IL-7: Comprehensive review. Cytokine 2022, 160, 156049. [Google Scholar] [CrossRef] [PubMed]
  46. Asadullah, K.; Haeussler, A.; Friedrich, M.; Siegling, A.; Olaizola-Horn, S.; Trefzer, U.; Volk, H.D.; Sterry, W. IL-7 mRNA is not overexpressed in mycosis fungoides and pleomorphic T-cell lymphoma and is likely to be an autocrine growth factor in vivo. Arch. Dermatol. Res. 1996, 289, 9–13. [Google Scholar] [CrossRef] [PubMed]
  47. Harada, A.; Sekido, N.; Akahoshi, T.; Wada, T.; Mukaida, N.; Matsushima, K. Essential involvement of interleukin-8 (IL-8) in acute inflammation. J. Leukoc. Biol. 1994, 56, 559–564. [Google Scholar] [CrossRef] [PubMed]
  48. Kimata, H.; Lindley, I. Detection of plasma interleukin-8 in atopic dermatitis. Arch. Dis. Child. 1994, 70, 119–122. [Google Scholar] [CrossRef]
  49. Abreu, M.; Miranda, M.; Castro, M.; Fernandes, I.; Cabral, R.; Santos, A.H.; Fonseca, S.; Rodrigues, J.; Leander, M.; Lau, C.; et al. IL-31 and IL-8 in Cutaneous T-Cell Lymphoma: Looking for Their Role in Itch. Adv. Hematol. 2021, 2021, e5582581. [Google Scholar] [CrossRef] [PubMed]
  50. Rojas-Zuleta, W.G.; Sanchez, E. IL-9: Function, Sources, and Detection. Methods Mol. Biol. 2017, 1585, 21–35. [Google Scholar] [CrossRef] [PubMed]
  51. Herrera, A.; Fredholm, S.; Cheng, A.; Mimitou, E.P.; Seffens, A.; Bar-Natan, M.; Sun, A.; Latkowski, J.-A.; Willerslew-Olsen, A.; Buus, T.B.; et al. Low SATB1 Expression Promotes IL-5 and IL-9 Expression in Sézary Syndrome. J. Investig. Dermatol. 2020, 140, 713–716. [Google Scholar] [CrossRef] [PubMed]
  52. Vieyra-Garcia, P.A.; Wei, T.; Naym, D.G.; Fredholm, S.; Fink-Puches, R.; Cerroni, L.; Odum, N.; O’Malley, J.T.; Gniadecki, R.; Wolf, P. STAT3/5-Dependent IL9 Overexpression Contributes to Neoplastic Cell Survival in Mycosis Fungoides. Clin. Cancer Res. 2016, 22, 3328–3339. [Google Scholar] [CrossRef] [PubMed]
  53. Wei, H.; Li, B.; Sun, A.; Guo, F. Interleukin-10 Family Cytokines Immunobiology and Structure. Adv. Exp. Med. Biol. 2019, 1172, 79–96. [Google Scholar] [CrossRef] [PubMed]
  54. Commins, S.; Steinke, J.W.; Borish, L. The extended IL-10 superfamily: IL-10, IL-19, IL-20, IL-22, IL-24, IL-26, IL-28, and IL-29. J. Allergy Clin. Immunol. 2008, 121, 1108–1111. [Google Scholar] [CrossRef]
  55. Akatsuka, T.; Miyagaki, T.; Nakajima, R.; Kamijo, H.; Oka, T.; Takahashi, N.; Suga, H.; Yoshizaki, A.; Asano, Y.; Sugaya, M.; et al. Decreased IL-10-producing regulatory B cells in patients with advanced mycosis fungoides. Eur. J. Dermatol. EJD 2018, 28, 314–319. [Google Scholar] [CrossRef] [PubMed]
  56. 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. [Google Scholar] [CrossRef] [PubMed]
  57. Chang, T.-P.; Poltoratsky, V.; Vancurova, I. Bortezomib Inhibits Expression of TGFβ1, IL-10, and CXCR4, Resulting in Decreased Survival and Migration of Cutaneous T Cell Lymphoma Cells. J. Immunol. 2015, 194, 2942–2953. [Google Scholar] [CrossRef]
  58. Fung, K.Y.; Louis, C.; Metcalfe, R.D.; Kosasih, C.C.; Wicks, I.P.; Griffin, M.D.W.; Putoczki, T.L. Emerging roles for IL-11 in inflammatory diseases. Cytokine 2022, 149, 155750. [Google Scholar] [CrossRef] [PubMed]
  59. Vignali, D.A.A.; Kuchroo, V.K. IL-12 family cytokines: Immunological playmakers. Nat. Immunol. 2012, 13, 722–728. [Google Scholar] [CrossRef] [PubMed]
  60. Rook, A.H.; Kubin, M.; Cassin, M.; Vonderheid, E.C.; Vowels, B.R.; Wolfe, J.T.; Wolf, S.F.; Singh, A.; Trinchieri, G.; Lessin, S.R. IL-12 reverses cytokine and immune abnormalities in Sezary syndrome. J. Immunol. 1995, 154, 1491–1498. [Google Scholar] [CrossRef] [PubMed]
  61. Duvic, M.; Sherman, M.L.; Wood, G.S.; Kuzel, T.M.; Olsen, E.; Foss, F.; Laliberté, R.J.; Ryan, J.L.; Zonno, K.; Rook, A.H. A phase II open-label study of recombinant human interleukin-12 in patients with stage IA, IB, or IIA mycosis fungoides. J. Am. Acad. Dermatol. 2006, 55, 807–813. [Google Scholar] [CrossRef] [PubMed]
  62. Walter, D.M.; McIntire, J.J.; Berry, G.; McKenzie, A.N.; Donaldson, D.D.; DeKruyff, R.H.; Umetsu, D.T. Critical role for IL-13 in the development of allergen-induced airway hyperreactivity. J. Immunol. 2001, 167, 4668–4675. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, Y.; Li, C.; Zhang, M.; Li, Z. IL-13 and IL-13Rα1 are overexpressed in extranodal natural killer/T cell lymphoma and mediate tumor cell proliferation. Biochem. Biophys. Res. Commun. 2018, 503, 2715–2720. [Google Scholar] [CrossRef] [PubMed]
  64. Kemp, E.H.; Ajjan, R.A.; Metcalfe, R.A.; Watson, P.F.; Weetman, A.P. IL-14 and IL-16 are expressed in the thyroid of patients with either Graves’ disease or Hashimoto’s thyroiditis. Clin. Endocrinol. 2015, 83, 726–732. [Google Scholar] [CrossRef] [PubMed]
  65. Marzec, M.; Liu, X.; Kasprzycka, M.; Witkiewicz, A.; Raghunath, P.N.; El-Salem, M.; Robertson, E.; Odum, N.; Wasik, M.A. IL-2- and IL-15-induced activation of the rapamycin-sensitive mTORC1 pathway in malignant CD4+ T lymphocytes. Blood 2008, 111, 2181–2189. [Google Scholar] [CrossRef] [PubMed]
  66. Willerslev-Olsen, A.; Litvinov, I.V.; Fredholm, S.M.; Petersen, D.L.; Sibbesen, N.A.; Gniadecki, R.; Zhang, Q.; Bonefeld, C.M.; Wasik, M.A.; Geisler, C.; et al. IL-15 and IL-17F are differentially regulated and expressed in mycosis fungoides (MF). Cell Cycle 2014, 13, 1306–1312. [Google Scholar] [CrossRef] [PubMed]
  67. Asadullah, K.; Haeussler-Quade, A.; Gellrich, S.; Hanneken, S.; Hansen-Hagge, T.E.; Döcke, W.D.; Volk, H.D.; Sterry, W. IL-15 and IL-16 overexpression in cutaneous T-cell lymphomas: Stage-dependent increase in mycosis fungoides progression. Exp. Dermatol. 2000, 9, 248–251. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, H.-J.; Bae, S.-C. Histone deacetylase inhibitors: Molecular mechanisms of action and clinical trials as anti-cancer drugs. Am. J. Transl. Res. 2011, 3, 166–179. [Google Scholar] [PubMed]
  69. Mishra, A.; La Perle, K.; Kwiatkowski, S.; Sullivan, L.A.; Sams, G.H.; Johns, J.; Curphey, D.P.; Wen, J.; McConnell, K.; Qi, J.; et al. Mechanism, Consequences, and Therapeutic Targeting of Abnormal IL15 Signaling in Cutaneous T-cell Lymphoma. Cancer Discov. 2016, 6, 986–1005. [Google Scholar] [CrossRef] [PubMed]
  70. Richmond, J.; Tuzova, M.; Cruikshank, W.; Center, D. Regulation of cellular processes by interleukin-16 in homeostasis and cancer. J. Cell Physiol. 2014, 229, 139–147. [Google Scholar] [CrossRef] [PubMed]
  71. Iwakura, Y.; Ishigame, H.; Saijo, S.; Nakae, S. Functional specialization of interleukin-17 family members. Immunity 2011, 34, 149–162. [Google Scholar] [CrossRef] [PubMed]
  72. Kołkowski, K.; Gleń, J.; Olszewska, B.; Zabłotna, M.; Nowicki, R.J.; Sokołowska-Wojdyło, M. Interleukin-17 Genes Polymorphisms are Significantly Associated with Cutaneous T-cell Lymphoma Susceptibility. Acta Derm. Venereol. 2022, 102, 2416. [Google Scholar] [CrossRef] [PubMed]
  73. Kaplanski, G. Interleukin-18: Biological properties and role in disease pathogenesis. Immunol. Rev. 2018, 281, 138–153. [Google Scholar] [CrossRef]
  74. Manfrere, K.C.G.; Torrealba, M.P.; Ferreira, F.M.; de Sousa, E.S.A.; Miyashiro, D.; Teixeira, F.M.E.; Custódio, R.W.A.; Nakaya, H.I.; Ramos, Y.A.L.; Sotto, M.N.; et al. Imbalanced IL-1B and IL-18 Expression in Sézary Syndrome. Int. J. Mol. Sci. 2023, 24, 4674. [Google Scholar] [CrossRef] [PubMed]
  75. Kempuraj, D.; Frydas, S.; Kandere, K.; Madhappan, B.; Letourneau, R.; Christodoulou, S.; Boucher, W.; Riccioni, G.; Conti, P.; Theoharides, T.C. Interleukin-19 (IL-19) network revisited. Int. J. Immunopathol. Pharmacol. 2003, 16, 95–97. [Google Scholar] [CrossRef]
  76. Senda, N.; Miyagaki, T.; Kamijo, H.; Nakajima, R.; Oka, T.; Takahashi, N.; Suga, H.; Yoshizaki, A.; Asano, Y.; Sugaya, M.; et al. Increased HMGB1 levels in lesional skin and sera in patients with cutaneous T-cell lymphoma. Eur. J. Dermatol. 2018, 28, 621–627. [Google Scholar] [CrossRef] [PubMed]
  77. Leonard, W.J.; Wan, C.-K. IL-21 Signaling in Immunity. F1000Research 2016, 5, 224. [Google Scholar] [CrossRef]
  78. Aulasevich, N.; Haist, M.; Försch, S.; Weidenthaler-Barth, B.; Mailänder, V. The Role of the Immune Phenotype in Tumor Progression and Prognosis of Patients with Mycosis Fungoides: A Quantitative Immunohistology Whole Slide Approach. Cells 2022, 11, 3570. [Google Scholar] [CrossRef] [PubMed]
  79. Duvallet, E.; Semerano, L.; Assier, E.; Falgarone, G.; Boissier, M.-C. Interleukin-23: A key cytokine in inflammatory diseases. Ann. Med. 2011, 43, 503–511. [Google Scholar] [CrossRef]
  80. Liu, Y.; Shao, Z.; Shangguan, G.; Bie, Q.; Zhang, B. Biological Properties and the Role of IL-25 in Disease Pathogenesis. J. Immunol. Res. 2018, 2018, 6519465. [Google Scholar] [CrossRef] [PubMed]
  81. Kourko, O.; Seaver, K.; Odoardi, N.; Basta, S.; Gee, K. IL-27, IL-30, and IL-35: A Cytokine Triumvirate in Cancer. Front. Oncol. 2019, 9, 969. [Google Scholar] [CrossRef] [PubMed]
  82. Yoshimoto, T.; Yoshimoto, T.; Yasuda, K.; Mizuguchi, J.; Nakanishi, K. IL-27 suppresses Th2 cell development and Th2 cytokines production from polarized Th2 cells: A novel therapeutic way for Th2-mediated allergic inflammation. J. Immunol. 2007, 179, 4415–4423. [Google Scholar] [CrossRef]
  83. Singer, E.M.; Shin, D.B.; Nattkemper, L.A.; Benoit, B.M.; Klein, R.S.; Didigu, C.A.; Loren, A.W.; Dentchev, T.; Wysocka, M.; Yosipovitch, G.; et al. IL-31 is produced by the malignant T-cell population in cutaneous T-Cell lymphoma and correlates with CTCL pruritus. J. Investig. Dermatol. 2013, 133, 2783–2785. [Google Scholar] [CrossRef] [PubMed]
  84. Ferretti, E.; Corcione, A.; Pistoia, V. The IL-31/IL-31 receptor axis: General features and role in tumor microenvironment. J. Leukoc. Biol. 2017, 102, 711–717. [Google Scholar] [CrossRef] [PubMed]
  85. Malek, M.; Gleń, J.; Rębała, K.; Kowalczyk, A.; Sobjanek, M.; Nowicki, R.; Ruckemann-Dziurdzińska, K.; Sokołowska-Wojdyło, M. Il-31 does not correlate to pruritus related to early stage cutaneous T-cell lymphomas but is involved in pathogenesis of the disease. Acta Derm. Venereol. 2015, 95, 283–288. [Google Scholar] [CrossRef] [PubMed]
  86. Yu, K.K.; Smith, N.P.; Essien, S.V.; Teague, J.E.; Vieyra-Garcia, P.; Gehad, A.; Zhan, Q.; Crouch, J.D.; Gerard, N.; Larocca, C.; et al. IL-32 Supports the Survival of Malignant T Cells in Cutaneous T-cell Lymphoma. J. Investig. Dermatol. 2022, 142, 2285–2288.e2. [Google Scholar] [CrossRef] [PubMed]
  87. Kurowska-Stolarska, M.; Stolarski, B.; Kewin, P.; Murphy, G.; Corrigan, C.; Ying, S.; Pitman, N.; Mirchandani, A.; Rana, B.; Van Rooijen, N.; et al. IL-33 Amplifies the Polarization of Alternatively Activated Macrophages That Contribute to Airway Inflammation1. J. Immunol. 2009, 183, 6469–6477. [Google Scholar] [CrossRef] [PubMed]
  88. Miller, A.M. Role of IL-33 in inflammation and disease. J. Inflamm. 2011, 8, 22. [Google Scholar] [CrossRef] [PubMed]
  89. Meephansan, J.; Tsuda, H.; Komine, M.; Tominaga, S.-I.; Ohtsuki, M. Regulation of IL-33 expression by IFN-γ and tumor necrosis factor-α in normal human epidermal keratinocytes. J. Investig. Dermatol. 2012, 132, 2593–2600. [Google Scholar] [CrossRef] [PubMed]
  90. Mager, L.F.; Riether, C.; Schürch, C.M.; Banz, Y.; Wasmer, M.-H.; Stuber, R.; Theocharides, A.P.; Li, X.; Xia, Y.; Saito, H.; et al. IL-33 signaling contributes to the pathogenesis of myeloproliferative neoplasms. J. Clin. Investig. 2015, 125, 2579–2591. [Google Scholar] [CrossRef] [PubMed]
  91. Yoo, J.; Rodriguez Perez, C.E.; Nie, W.; Edwards, R.A.; Sinnett-Smith, J.; Rozengurt, E. TNF-α induces upregulation of EGFR expression and signaling in human colonic myofibroblasts. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, G805–G814. [Google Scholar] [CrossRef] [PubMed]
  92. Filardo, E.J. Epidermal growth factor receptor (EGFR) transactivation by estrogen via the G-protein-coupled receptor, GPR30: A novel signaling pathway with potential significance for breast cancer. J. Steroid Biochem. Mol. Biol. 2002, 80, 231–238. [Google Scholar] [CrossRef] [PubMed]
  93. Ferguson, H.R.; Smith, M.P.; Francavilla, C. Fibroblast Growth Factor Receptors (FGFRs) and Noncanonical Partners in Cancer Signaling. Cells 2021, 10, 1201. [Google Scholar] [CrossRef] [PubMed]
  94. Horikawa, S.; Ishii, Y.; Hamashima, T.; Yamamoto, S.; Mori, H.; Fujimori, T.; Shen, J.; Inoue, R.; Nishizono, H.; Itoh, H.; et al. PDGFRα plays a crucial role in connective tissue remodeling. Sci. Rep. 2015, 5, 17948. [Google Scholar] [CrossRef] [PubMed]
  95. Yu, Z.; Vieyra-Garcia, P.; Benezeder, T.; Crouch, J.D.; Kim, I.R.; O’Malley, J.T.; Devlin, P.M.; Gehad, A.; Zhan, Q.; Gudjonsson, J.E.; et al. Phototherapy Restores Deficient Type I IFN Production and Enhances Antitumor Responses in Mycosis Fungoides. J. Investig. Dermatol. 2023, 144, 621–632.E1. [Google Scholar] [CrossRef]
  96. Kak, G.; Raza, M.; Tiwari, B.K. Interferon-gamma (IFN-γ): Exploring its implications in infectious diseases. Biomol. Concepts 2018, 9, 64–79. [Google Scholar] [CrossRef] [PubMed]
  97. Uzé, G.; Monneron, D. IL-28 and IL-29: Newcomers to the interferon family. Biochimie 2007, 89, 729–734. [Google Scholar] [CrossRef] [PubMed]
  98. Boehm, U.; Klamp, T.; Groot, M.; Howard, J.C. Cellular responses to interferon-gamma. Annu. Rev. Immunol. 1997, 15, 749–795. [Google Scholar] [CrossRef]
  99. Tensen, C.P.; Vermeer, M.H.; van der Stoop, P.M.; van Beek, P.; Scheper, R.J.; Boorsma, D.M.; Willemze, R. Epidermal Interferon-γ Inducible Protein-10 (IP-10) and Monokine Induced by γ-Interferon (Mig) but not IL-8 mRNA Expression is Associated with Epidermotropism in Cutaneous T Cell Lymphomas. J. Investig. Dermatol. 1998, 111, 222–226. [Google Scholar] [CrossRef] [PubMed]
  100. García-Vega, Y.; García-García, I.; Collazo-Caballero, S.E.; Santely-Pravia, E.E.; Cruz-Ramírez, A.; Tuero-Iglesias, A.D.; Alfonso-Alvarado, C.; Cabrera-Placeres, M.; Castro-Basart, N.; Duncan-Roberts, Y.; et al. Pharmacokinetic and pharmacodynamic characterization of a new formulation containing synergistic proportions of interferons alpha-2b and gamma (HeberPAG) in patients with mycosis fungoides: An open-label trial. BMC Pharmacol. Toxicol. 2012, 13, 20. [Google Scholar] [CrossRef] [PubMed]
  101. Seo, N.; Tokura, Y.; Matsumoto, K.; Furukawa, F.; Takigawa, M. Tumour-specific cytotoxic T lymphocyte activity in Th2-type Sézary syndrome: Its enhancement by interferon-gamma (IFN-gamma) and IL-12 and fluctuations in association with disease activity. Clin. Exp. Immunol. 1998, 112, 403–409. [Google Scholar] [CrossRef] [PubMed]
  102. Kaneko, N.; Kurata, M.; Yamamoto, T.; Morikawa, S.; Masumoto, J. The role of interleukin-1 in general pathology. Inflamm. Regen. 2019, 39, 12. [Google Scholar] [CrossRef] [PubMed]
  103. Guo, B.; Fu, S.; Zhang, J.; Liu, B.; Li, Z. Targeting inflammasome/IL-1 pathways for cancer immunotherapy. Sci. Rep. 2016, 6, 36107. [Google Scholar] [CrossRef] [PubMed]
  104. Lawlor, F.; Smith, N.P.; Camp, R.D.; Bacon, K.B.; Black, A.K.; Greaves, M.W.; Gearing, A.J. Skin exudate levels of interleukin 6, interleukin i and other cytokines in mycosis fungoides. Br. J. Dermatol. 1990, 123, 297–304. [Google Scholar] [CrossRef] [PubMed]
  105. Ying, Z.; Shiue, L.; Park, K.; Kollet, J.; Bijani, P.; Goswami, M.; Duvic, M.; Ni, X. Blood transcriptional profiling reveals IL-1 and integrin signaling pathways associated with clinical response to extracorporeal photopheresis in patients with leukemic cutaneous T-cell lymphoma. Oncotarget 2019, 10, 3183–3197. [Google Scholar] [CrossRef] [PubMed]
  106. Hodge, S.; Hodge, G.; Flower, R.; Han, P. Surface and intracellular interleukin-2 receptor expression on various resting and activated populations involved in cell-mediated immunity in human peripheral blood. Scand. J. Immunol. 2000, 51, 67–72. [Google Scholar] [CrossRef] [PubMed]
  107. Refaeli, Y.; Van Parijs, L.; London, C.A.; Tschopp, J.; Abbas, A.K. Biochemical mechanisms of IL-2-regulated Fas-mediated T cell apoptosis. Immunity 1998, 8, 615–623. [Google Scholar] [CrossRef] [PubMed]
  108. Rybojad, M.; Marolleau, J.P.; Flageul, B.; Baccard, M.; Brandely, M.; Morel, P.; Gisselbrecht, C. Successful interleukin-2 therapy of advanced cutaneous T-cell lymphoma. Br. J. Dermatol. 1992, 127, 63–64. [Google Scholar] [CrossRef] [PubMed]
  109. Baccard, M.; Marolleau, J.P.; Rybojad, M. Middle-Term Evolution of Patients with Advanced Cutaneous T-Cell Lymphoma Treated with High-Dose Recombinant Interleukin-2. Arch. Dermatol. 1997, 133, 656. [Google Scholar] [CrossRef] [PubMed]
  110. Nagatani, T.; Kin, S.T.; Baba, N.; Miyamoto, H.; Nakajima, H.; Katoh, Y. A case of cutaneous T cell lymphoma treated with recombinant interleukin 2 (rIL-2). Acta Derm. Venereol. 1988, 68, 504–508. [Google Scholar]
  111. Querfeld, C.; Rosen, S.T.; Guitart, J.; Rademaker, A.; Foss, F.; Gupta, R.; Kuzel, T.M. Phase II trial of subcutaneous injections of human recombinant interleukin-2 for the treatment of mycosis fungoides and Sézary syndrome. J. Am. Acad. Dermatol. 2007, 56, 580–583. [Google Scholar] [CrossRef] [PubMed]
  112. Figgitt, D.P.; Lamb, H.M.; Goa, K.L. Denileukin diftitox. Am. J. Clin. Dermatol. 2000, 1, 67–72; discussion 73. [Google Scholar] [CrossRef]
  113. Kaminetzky, D.; Hymes, K.B. Denileukin diftitox for the treatment of cutaneous T-cell lymphoma. Biologics 2008, 2, 717–724. [Google Scholar] [PubMed]
  114. Gadani, S.P.; Cronk, J.C.; Norris, G.T.; Kipnis, J. IL-4 in the brain: A cytokine to remember. J. Immunol. 2012, 189, 4213–4219. [Google Scholar] [CrossRef] [PubMed]
  115. Huanosta-Murillo, E.; Alcántara-Hernández, M.; Hernández-Rico, B.; Victoria-Acosta, G.; Miranda-Cruz, P.; Domínguez-Gómez, M.A.; Jurado-Santacruz, F.; Patiño-López, G.; Pérez-Koldenkova, V.; Palma-Guzmán, A.; et al. NLRP3 Regulates IL-4 Expression in TOX+ CD4+ T Cells of Cutaneous T Cell Lymphoma to Potentially Promote Disease Progression. Front. Immunol. 2021, 12, 668369. [Google Scholar] [CrossRef] [PubMed]
  116. Huang, X.; Li, Y.; Fu, M.; Xin, H.-B. Polarizing Macrophages In Vitro. Methods Mol. Biol. 2018, 1784, 119–126. [Google Scholar] [CrossRef] [PubMed]
  117. Vowels, B.R.; Lessin, S.R.; Cassin, M.; Jaworsky, C.; Benoit, B.; Wolfe, J.T.; Rook, A.H. Th2 cytokine mRNA expression in skin in cutaneous T-cell lymphoma. J. Investig. Dermatol. 1994, 103, 669–673. [Google Scholar] [CrossRef] [PubMed]
  118. Hoppe, R.T.; Medeiros, L.J.; Warnke, R.A.; Wood, G.S. CD8-positive tumor-infiltrating lymphocytes influence the long-term survival of patients with mycosis fungoides. J. Am. Acad. Dermatol. 1995, 32, 448–453. [Google Scholar] [CrossRef] [PubMed]
  119. Papadavid, E.; Economidou, J.; Psarra, A.; Kapsimali, V.; Mantzana, V.; Antoniou, C.; Limas, K.; Stratigos, A.; Stavrianeas, N.; Avgerinou, G.; et al. The relevance of peripheral blood T-helper 1 and 2 cytokine pattern in the evaluation of patients with mycosis fungoides and Sézary syndrome. Br. J. Dermatol. 2003, 148, 709–718. [Google Scholar] [CrossRef] [PubMed]
  120. Litvinov, I.V.; Cordeiro, B.; Fredholm, S.; Ødum, N.; Zargham, H.; Huang, Y.; Zhou, Y.; Pehr, K.; Kupper, T.S.; Woetmann, A.; et al. Analysis of STAT4 expression in cutaneous T-cell lymphoma (CTCL) patients and patient-derived cell lines. Cell Cycle 2014, 13, 2975–2982. [Google Scholar] [CrossRef] [PubMed]
  121. Vowels, B.R.; Cassin, M.; Vonderheid, E.C.; Rook, A.H. Aberrant cytokine production by Sezary syndrome patients: Cytokine secretion pattern resembles murine Th2 cells. J. Investig. Dermatol. 1992, 99, 90–94. [Google Scholar] [CrossRef] [PubMed]
  122. Geskin, L.J.; Viragova, S.; Stolz, D.B.; Fuschiotti, P. Interleukin-13 is overexpressed in cutaneous T-cell lymphoma cells and regulates their proliferation. Blood 2015, 125, 2798–2805. [Google Scholar] [CrossRef] [PubMed]
  123. Furudate, S.; Fujimura, T.; Kakizaki, A.; Kambayashi, Y.; Asano, M.; Watabe, A.; Aiba, S. The possible interaction between periostin expressed by cancer stroma and tumor-associated macrophages in developing mycosis fungoides. Exp. Dermatol. 2016, 25, 107–112. [Google Scholar] [CrossRef] [PubMed]
  124. Furudate, S.; Fujimura, T.; Kakizaki, A.; Hidaka, T.; Asano, M.; Aiba, S. Tumor-associated M2 macrophages in mycosis fungoides acquire immunomodulatory function by interferon alpha and interferon gamma. J. Dermatol. Sci. 2016, 83, 182–189. [Google Scholar] [CrossRef] [PubMed]
  125. Ferretti, E.; Hohaus, S.; Di Napoli, A.; Belmonte, B.; Cuccaro, A.; Cupelli, E.; Galli, E.; Rufini, V.; Tripodi, G.; Fraternali-Orcioni, G.; et al. Interleukin-31 and thymic stromal lymphopoietin expression in plasma and lymph node from Hodgkin lymphoma patients. Oncotarget 2017, 8, 85263–85275. [Google Scholar] [CrossRef] [PubMed]
  126. Guttman-Yassky, E.; Pavel, A.B.; Diaz, A.; Zhang, N.; Del Duca, E.; Estrada, Y.; King, B.; Banerjee, A.; Banfield, C.; Cox, L.A.; et al. Ritlecitinib and brepocitinib demonstrate significant improvement in scalp alopecia areata biomarkers. J. Allergy Clin. Immunol. 2022, 149, 1318–1328. [Google Scholar] [CrossRef] [PubMed]
  127. Guenova, E.; Watanabe, R.; Teague, J.E.; Desimone, J.A.; Jiang, Y.; Dowlatshahi, M.; Schlapbach, C.; Schaekel, K.; Rook, A.H.; Tawa, 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, 19, 3755–3763. [Google Scholar] [CrossRef]
  128. Takahashi, N.; Sugaya, M.; Suga, H.; Oka, T.; Kawaguchi, M.; Miyagaki, T.; Fujita, H.; Sato, S. Thymic Stromal Chemokine TSLP Acts through Th2 Cytokine Production to Induce Cutaneous T-cell Lymphoma. Cancer Res. 2016, 76, 6241–6252. [Google Scholar] [CrossRef] [PubMed]
  129. Miyagaki, T.; Sugaya, M.; Shibata, S.; Ohmatsu, H.; Fujita, H.; Tamaki, K. Serum interleukin-27 levels in patients with cutaneous T-cell lymphoma. Clin. Exp. Dermatol. 2010, 35, e143–e144. [Google Scholar] [CrossRef] [PubMed]
  130. Tuzova, M.; Richmond, J.; Wolpowitz, D.; Curiel-Lewandrowski, C.; Chaney, K.; Kupper, T.; Cruikshank, W. CCR4+T cell recruitment to the skin in mycosis fungoides: Potential contributions by thymic stromal lymphopoietin and interleukin-16. Leuk. Lymphoma 2015, 56, 440–449. [Google Scholar] [CrossRef]
  131. Stolearenco, V.; Namini, M.R.J.; Hasselager, S.S.; Gluud, M.; Buus, T.B.; Willerslev-Olsen, A.; Ødum, N.; Krejsgaard, T. Cellular Interactions and Inflammation in the Pathogenesis of Cutaneous T-Cell Lymphoma. Front. Cell Dev. Biol. 2020, 8, 851. [Google Scholar] [CrossRef]
  132. Jfri, A.; Smith, J.S.; Larocca, C. Diagnosis of mycosis fungoides or Sézary syndrome after dupilumab use: A systematic review. J. Am. Acad. Dermatol. 2023, 88, 1164–1166. [Google Scholar] [CrossRef] [PubMed]
  133. Yan, D.; Ramachandran, V.; Weston, G.; Kim, R.H.; Latkowski, J.-A. Diagnosing mycosis fungoides after initiation of therapy with dupilumab: A case report and literature review. Int. J. Dermatol. 2023, 62, e500–e503. [Google Scholar] [CrossRef] [PubMed]
  134. Miyagaki, T.; Sugaya, M. Erythrodermic cutaneous T-cell lymphoma: How to differentiate this rare disease from atopic dermatitis. J. Dermatol. Sci. 2011, 64, 1–6. [Google Scholar] [CrossRef] [PubMed]
  135. Schlaeger, M. Immunoglobulin E in cutaneous lymphomas and in parapsoriasis en plaques (Brocq) (author’s transl). Dermatologica 1980, 161, 1–7. [Google Scholar] [CrossRef] [PubMed]
  136. Guilhou, J.J.; Clot, J.; Robinet-Lévy, M.; Meynadier, J. IgE and cellular immunity in cutaneous lymphomas (author’s transl). Ann. Dermatol. Venereol. 1977, 104, 533–537. [Google Scholar] [PubMed]
  137. Guan, W.; Ni, Z.; Hu, Y.; Liang, W.; Ou, C.; He, J.; Liu, L.; Shan, H.; Lei, C.; Hui, D.S.C.; et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef]
  138. Suchin, K.R.; Cassin, M.; Gottleib, S.L.; Sood, S.; Cucchiara, A.J.; Vonderheid, E.C.; Rook, A.H. Increased interleukin 5 production in eosinophilic Sézary syndrome: Regulation by interferon alfa and interleukin 12. J. Am. Acad. Dermatol. 2001, 44, 28–32. [Google Scholar] [CrossRef] [PubMed]
  139. Heinrich, M.C.; Corless, C.L.; Duensing, A.; McGreevey, L.; Chen, C.-J.; Joseph, N.; Singer, S.; Griffith, D.J.; Haley, A.; Town, A.; et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003, 299, 708–710. [Google Scholar] [CrossRef]
  140. Karamova, A.E.; Verbenko, D.A.; Vorontsova, A.A.; Zhilova, M.B.; Nikonorov, A.A.; Gatiatulina, E.R.; Znamenskaya, L.F.; Kubanov, A.A. Effect of PUVA and NB-UVB Therapy on the Skin Cytokine Profile in Patients with Mycosis Fungoides. J. Oncol. 2022, 2022, 3149293. [Google Scholar] [CrossRef] [PubMed]
  141. Chen, D.; Tang, T.-X.; Deng, H.; Yang, X.-P.; Tang, Z.-H. Interleukin-7 Biology and Its Effects on Immune Cells: Mediator of Generation, Differentiation, Survival, and Homeostasis. Front. Immunol. 2021, 12, 747324. [Google Scholar] [CrossRef] [PubMed]
  142. Qin, J.Z.; Kamarashev, J.; Zhang, C.L.; 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. Investig. Dermatol. 2001, 117, 583–589. [Google Scholar] [CrossRef] [PubMed]
  143. Foss, F.M.; Koc, Y.; Stetler-Stevenson, M.A.; Nguyen, D.T.; O’Brien, M.C.; Turner, R.; Sausville, E.A. Costimulation of cutaneous T-cell lymphoma cells by interleukin-7 and interleukin-2: Potential autocrine or paracrine effectors in the Sézary syndrome. J. Clin. Oncol. 1994, 12, 326–335. [Google Scholar] [CrossRef] [PubMed]
  144. Netchiporouk, E.; Litvinov, I.V.; Moreau, L.; Gilbert, M.; Sasseville, D.; Duvic, M. Deregulation in STAT signaling is important for cutaneous T-cell lymphoma (CTCL) pathogenesis and cancer progression. Cell Cycle 2014, 13, 3331–3335. [Google Scholar] [CrossRef] [PubMed]
  145. Merle-Béral, H.; Schmitt, C.; Mossalayi, D.; Bismuth, G.; Dalloul, A.; Mentz, F. Interleukin-7 and malignant T cells. Nouv. Rev. Fr. Hematol. 1993, 35, 231–232. [Google Scholar] [PubMed]
  146. Bagot, M.; Charue, D.; Boulland, M.L.; Gaulard, P.; Revuz, J.; Schmitt, C.; Wechsler, J. Interleukin-7 receptor expression in cutaneous T-cell lymphomas. Br. J. Dermatol. 1996, 135, 572–575. [Google Scholar] [CrossRef] [PubMed]
  147. Sportès, C.; Gress, R.E. Interleukin-7 immunotherapy. Adv. Exp. Med. Biol. 2007, 601, 321–333. [Google Scholar] [CrossRef] [PubMed]
  148. Doherty, T.A.; Broide, D.H. Insights into the biology of IL-9 in asthma. J. Allergy Clin. Immunol. 2022, 150, 585–586. [Google Scholar] [CrossRef] [PubMed]
  149. Fang, Y.; Chen, X.; Bai, Q.; Qin, C.; Mohamud, A.O.; Zhu, Z.; Ball, T.W.; Ruth, C.M.; Newcomer, D.R.; Herrick, E.J.; et al. IL-9 inhibits HTB-72 melanoma cell growth through upregulation of p21 and TRAIL. J. Surg. Oncol. 2015, 111, 969–974. [Google Scholar] [CrossRef] [PubMed]
  150. Zheng, N.; Lu, Y. Targeting the IL-9 pathway in cancer immunotherapy. Hum. Vaccin. Immunother. 2020, 16, 2333–2340. [Google Scholar] [CrossRef] [PubMed]
  151. Ng, T.H.S.; Britton, G.; Hill, E.; Verhagen, J.; Burton, B.; Wraith, D. Regulation of Adaptive Immunity; The Role of Interleukin-10. Front. Immunol. 2013, 4, 129. [Google Scholar] [CrossRef]
  152. Howes, A.; Gabryšová, L.; O’Garra, A. Role of IL-10 and the IL-10 Receptor in Immune Responses. In Reference Module in Biomedical Sciences; Elsevier: Amsterdam, The Netherlands, 2014; ISBN 978-0-12-801238-3. [Google Scholar]
  153. Wu, X.; Hsu, D.K.; Wang, K.-H.; Huang, Y.; Mendoza, L.; Zhou, Y.; Hwang, S.T. IL-10 is overexpressed in human cutaneous T-cell lymphoma and is required for maximal tumor growth in a mouse model. Leuk. Lymphoma 2019, 60, 1244–1252. [Google Scholar] [CrossRef] [PubMed]
  154. Nedoszytko, B.; Olszewska, B.; Roszkiewicz, J.; Glen, J.; Zabłotna, M.; Ługowska-Umer, H.; Nowicki, R.; Sokołowska-Wojdyło, M. The role of polymorphism of interleukin-2, -10, -13 and TNF-α genes in cutaneous T-cell lymphoma pathogenesis. Postep. Dermatol. Alergol. 2016, 33, 429–434. [Google Scholar] [CrossRef] [PubMed]
  155. Dittrich, A.; Hessenkemper, W.; Schaper, F. Systems biology of IL-6, IL-12 family cytokines. Cytokine Growth Factor. Rev. 2015, 26, 595–602. [Google Scholar] [CrossRef] [PubMed]
  156. Glassman, C.R.; Mathiharan, Y.K.; Jude, K.M.; Su, L.; Panova, O.; Lupardus, P.J.; Spangler, J.B.; Ely, L.K.; Thomas, C.; Skiniotis, G.; et al. Structural basis for IL-12 and IL-23 receptor sharing reveals a gateway for shaping actions on T versus NK cells. Cell 2021, 184, 983–999.e24. [Google Scholar] [CrossRef] [PubMed]
  157. Nguyen, K.G.; Vrabel, M.R.; Mantooth, S.M.; Hopkins, J.J.; Wagner, E.S.; Gabaldon, T.A.; Zaharoff, D.A. Localized Interleukin-12 for Cancer Immunotherapy. Front. Immunol. 2020, 11, 575597. [Google Scholar] [CrossRef] [PubMed]
  158. Showe, L.C.; Fox, F.E.; Williams, D.; Au, K.; Niu, Z.; Rook, A.H. Depressed IL-12-mediated signal transduction in T cells from patients with Sézary syndrome is associated with the absence of IL-12 receptor beta 2 mRNA and highly reduced levels of STAT4. J. Immunol. 1999, 163, 4073–4079. [Google Scholar] [CrossRef] [PubMed]
  159. Davis, M.S.; Spencer, R.K.; Johnson, C.E.; Elhage, K.G.; Jin, J.Q.; Hakimi, M.; Bhutani, T.; Liao, W. Risk of Cutaneous T Cell Lymphoma with Psoriasis Biologic Therapies. Dermatol. Ther. 2023, 14, 15–30. [Google Scholar] [CrossRef] [PubMed]
  160. Wynn, T.A. IL-13 effector functions. Annu. Rev. Immunol. 2003, 21, 425–456. [Google Scholar] [CrossRef] [PubMed]
  161. de Vries, J.E. The role of IL-13 and its receptor in allergy and inflammatory responses. J. Allergy Clin. Immunol. 1998, 102, 165–169. [Google Scholar] [CrossRef] [PubMed]
  162. Urban, J.F.; Noben-Trauth, N.; Donaldson, D.D.; Madden, K.B.; Morris, S.C.; Collins, M.; Finkelman, F.D. IL-13, IL-4Ralpha, and Stat6 are required for the expulsion of the gastrointestinal nematode parasite Nippostrongylus brasiliensis. Immunity 1998, 8, 255–264. [Google Scholar] [CrossRef] [PubMed]
  163. Alexander, J.; Brombacher, F.; McGachy, H.A.; McKenzie, A.N.J.; Walker, W.; Carter, K.C. An essential role for IL-13 in maintaining a non-healing response following Leishmania mexicana infection. Eur. J. Immunol. 2002, 32, 2923–2933. [Google Scholar] [CrossRef] [PubMed]
  164. Mohrs, M.; Ledermann, B.; Köhler, G.; Dorfmüller, A.; Gessner, A.; Brombacher, F. Differences between IL-4- and IL-4 receptor alpha-deficient mice in chronic leishmaniasis reveal a protective role for IL-13 receptor signaling. J. Immunol. 1999, 162, 7302–7308. [Google Scholar] [CrossRef] [PubMed]
  165. Leigh, R.; Ellis, R.; Wattie, J.; Donaldson, D.D.; Inman, M.D. Is interleukin-13 critical in maintaining airway hyperresponsiveness in allergen-challenged mice? Am. J. Respir. Crit. Care Med. 2004, 170, 851–856. [Google Scholar] [CrossRef] [PubMed]
  166. Gabitass, R.F.; Annels, N.E.; Stocken, D.D.; Pandha, H.A.; Middleton, G.W. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol. Immunother. 2011, 60, 1419–1430. [Google Scholar] [CrossRef] [PubMed]
  167. Shi, J.; Song, X.; Traub, B.; Luxenhofer, M.; Kornmann, M. Involvement of IL-4, IL-13 and Their Receptors in Pancreatic Cancer. Int. J. Mol. Sci. 2021, 22, 2998. [Google Scholar] [CrossRef] [PubMed]
  168. Lin, C.; Liu, H.; Zhang, H.; He, H.; Li, H.; Shen, Z.; Qin, J.; Qin, X.; Xu, J.; Sun, Y. Interleukin-13 receptor α2 is associated with poor prognosis in patients with gastric cancer after gastrectomy. Oncotarget 2016, 7, 49281–49288. [Google Scholar] [CrossRef] [PubMed]
  169. Skinnider, B.F.; Kapp, U.; Mak, T.W. The role of interleukin 13 in classical Hodgkin lymphoma. Leuk. Lymphoma 2002, 43, 1203–1210. [Google Scholar] [CrossRef] [PubMed]
  170. Gluud, M.; Pallesen, E.M.H.; Buus, T.B.; Gjerdrum, L.M.R.; Lindahl, L.M.; Kamstrup, M.R.; Bzorek, M.; Danielsen, M.; Bech, R.; Monteiro, M.N.; et al. Malignant T cells induce skin barrier defects through cytokine-mediated JAK/STAT signaling in cutaneous T-cell lymphoma. Blood 2023, 141, 180–193. [Google Scholar] [CrossRef]
  171. Hashimoto, M.; Miyagaki, T.; Komaki, R.; Takeuchi, S.; Kadono, T. Development of Nodular Lesions after Dupilumab Therapy in Erythrodermic Mycosis Fungoides with Interleukin-13 Receptor alpha2 Expression. Acta Derm. Venereol. 2022, 102, adv00766. [Google Scholar] [CrossRef] [PubMed]
  172. Nogami, S.; Satoh, S.; Tanaka-Nakadate, S.; Yoshida, K.; Nakano, M.; Terano, A.; Shirataki, H. Identification and characterization of taxilin isoforms. Biochem. Biophys. Res. Commun. 2004, 319, 936–943. [Google Scholar] [CrossRef] [PubMed]
  173. Ford, R.; Tamayo, A.; Martin, B.; Niu, K.; Claypool, K.; Cabanillas, F.; Ambrus, J. Identification of B-cell growth factors (interleukin-14; high molecular weight-B-cell growth factors) in effusion fluids from patients with aggressive B-cell lymphomas. Blood 1995, 86, 283–293. [Google Scholar] [CrossRef] [PubMed]
  174. Bodine, B.G.; Bennion, B.G.; Leatham, E.; Jimenez, F.R.; Wright, A.J.; Jergensen, Z.R.; Erickson, C.J.; Jones, C.M.; Johnson, J.P.; Knapp, S.M.; et al. Conditionally induced RAGE expression by proximal airway epithelial cells in transgenic mice causes lung inflammation. Respir. Res. 2014, 15, 133. [Google Scholar] [CrossRef]
  175. Singh, A.R.; Peirce, S.K.; Joshi, S.; Durden, D.L. PTEN and PI-3 kinase inhibitors control LPS signaling and the lymphoproliferative response in the CD19+ B cell compartment. Exp. Cell Res. 2014, 327, 78–90. [Google Scholar] [CrossRef]
  176. Steel, J.C.; Waldmann, T.A.; Morris, J.C. Interleukin-15 biology and its therapeutic implications in cancer. Trends Pharmacol. Sci. 2012, 33, 35–41. [Google Scholar] [CrossRef]
  177. Li, X.; Bechara, R.; Zhao, J.; McGeachy, M.J.; Gaffen, S.L. IL-17 receptor-based signaling and implications for disease. Nat. Immunol. 2019, 20, 1594–1602. [Google Scholar] [CrossRef]
  178. Kopp, K.; Ralfkiaer, U.; Mette Gjerdrum, L.; Helvad, R.; Pedersen, I.; Litman, T.; Jønson, L.; Hagedorn, P.; Krejsgaard, T.; Gniadecki, R.; et al. STAT5-mediated expression of oncogenic miR-155 in cutaneous T-cell lymphoma. Cell Cycle 2013, 12, 1939–1947. [Google Scholar] [CrossRef] [PubMed]
  179. Döbbeling, U.; Dummer, R.; Laine, E.; Potoczna, N.; Qin, J.Z.; Burg, G. Interleukin-15 is an autocrine/paracrine viability factor for cutaneous T-cell lymphoma cells. Blood 1998, 92, 252–258. [Google Scholar] [CrossRef] [PubMed]
  180. Qin, J.Z.; Zhang, C.L.; Kamarashev, J.; Dummer, R.; Burg, G.; Döbbeling, U. Interleukin-7 and interleukin-15 regulate the expression of the bcl-2 and c-myb genes in cutaneous T-cell lymphoma cells. Blood 2001, 98, 2778–2783. [Google Scholar] [CrossRef]
  181. Leroy, S.; Dubois, S.; Tenaud, I.; Chebassier, N.; Godard, A.; Jacques, Y.; Dréno, B. Interleukin-15 expression in cutaneous T-cell lymphoma (mycosis fungoides and Sézary syndrome). Br. J. Dermatol. 2001, 144, 1016–1023. [Google Scholar] [CrossRef] [PubMed]
  182. Sugaya, M. The Role of Th17-Related Cytokines in Atopic Dermatitis. Int. J. Mol. Sci. 2020, 21, 1314. [Google Scholar] [CrossRef]
  183. Wysocka, M.; Benoit, B.M.; Newton, S.; Azzoni, L.; Montaner, L.J.; Rook, A.H. Enhancement of the host immune responses in cutaneous T-cell lymphoma by CpG oligodeoxynucleotides and IL-15. Blood 2004, 104, 4142–4149. [Google Scholar] [CrossRef] [PubMed]
  184. Wysocka, M.; Dawany, N.; Benoit, B.; Kossenkov, A.V.; Troxel, A.B.; Gelfand, J.M.; Sell, M.K.; Showe, L.C.; Rook, A.H. Synergistic enhancement of cellular immune responses by the novel Toll receptor 7/8 agonist 3M-007 and interferon-γ: Implications for therapy of cutaneous T-cell lymphoma. Leuk. Lymphoma 2011, 52, 1970–1979. [Google Scholar] [CrossRef] [PubMed]
  185. Krejsgaard, T.; Litvinov, I.V.; Wang, Y.; Xia, L.; Willerslev-Olsen, A.; Koralov, S.B.; Kopp, K.L.; Bonefeld, C.M.; Wasik, M.A.; Geisler, C.; et al. Elucidating the role of interleukin-17F in cutaneous T-cell lymphoma. Blood 2013, 122, 943–950. [Google Scholar] [CrossRef] [PubMed]
  186. Krejsgaard, T.; Vetter-Kauczok, C.S.; Woetmann, A.; Lovato, P.; Labuda, T.; Eriksen, K.W.; Zhang, Q.; Becker, J.C.; Ødum, N. Jak3- and JNK-dependent vascular endothelial growth factor expression in cutaneous T-cell lymphoma. Leukemia 2006, 20, 1759–1766. [Google Scholar] [CrossRef] [PubMed]
  187. Papathemeli, D.; Patsatsi, A.; Papanastassiou, D.; Koletsa, T.; Papathemelis, T.; Avgeros, C.; Pikou, O.; Lazaridou, E.; Georgiou, E. Protein and mRNA Expression Levels of Interleukin-17A, -17F and -22 in Blood and Skin Samples of Patients with Mycosis Fungoides. Acta Derm. -Venereol. 2020, 100, 1–6. [Google Scholar] [CrossRef] [PubMed]
  188. Richmond, J.; Tuzova, M.; Parks, A.; Adams, N.; Martin, E.; Tawa, M.; Morrison, L.; Chaney, K.; Kupper, T.S.; Curiel-Lewandrowski, C.; et al. Interleukin-16 as a marker of Sézary syndrome onset and stage. J. Clin. Immunol. 2011, 31, 39–50. [Google Scholar] [CrossRef] [PubMed]
  189. Hoshino, T.; Wiltrout, R.H.; Young, H.A. IL-18 is a potent coinducer of IL-13 in NK and T cells: A new potential role for IL-18 in modulating the immune response. J. Immunol. 1999, 162, 5070–5077. [Google Scholar] [CrossRef]
  190. Nakanishi, K.; Yoshimoto, T.; Tsutsui, H.; Okamura, H. Interleukin-18 regulates both Th1 and Th2 responses. Annu. Rev. Immunol. 2001, 19, 423–474. [Google Scholar] [CrossRef]
  191. Bostan, E.; Gokoz, O.; Atakan, N. The role of NLRP1 and NLRP3 inflammasomes in the etiopathogeneses of pityriasis lichenoides chronica and mycosis fungoides: An immunohistochemical study. Arch. Dermatol. Res. 2023, 315, 231–239. [Google Scholar] [CrossRef] [PubMed]
  192. Azuma, Y.-T.; Matsuo, Y.; Kuwamura, M.; Yancopoulos, G.D.; Valenzuela, D.M.; Murphy, A.J.; Nakajima, H.; Karow, M.; Takeuchi, T. Interleukin-19 protects mice from innate-mediated colonic inflammation. Inflamm. Bowel Dis. 2010, 16, 1017–1028. [Google Scholar] [CrossRef] [PubMed]
  193. Wei, C.-C.; Hsu, Y.-H.; Li, H.-H.; Wang, Y.-C.; Hsieh, M.-Y.; Chen, W.-Y.; Hsing, C.-H.; Chang, M.-S. IL-20: Biological functions and clinical implications. J. Biomed. Sci. 2006, 13, 601–612. [Google Scholar] [CrossRef] [PubMed]
  194. Hsu, Y.-H.; Hsing, C.-H.; Li, C.-F.; Chan, C.-H.; Chang, M.-C.; Yan, J.-J.; Chang, M.-S. Anti-IL-20 monoclonal antibody suppresses breast cancer progression and bone osteolysis in murine models. J. Immunol. 2012, 188, 1981–1991. [Google Scholar] [CrossRef] [PubMed]
  195. Lee, S.-J.; Cho, S.-C.; Lee, E.-J.; Kim, S.; Lee, S.-B.; Lim, J.-H.; Choi, Y.H.; Kim, W.-J.; Moon, S.-K. Interleukin-20 promotes migration of bladder cancer cells through extracellular signal-regulated kinase (ERK)-mediated MMP-9 protein expression leading to nuclear factor (NF-κB) activation by inducing the up-regulation of p21(WAF1) protein expression. J. Biol. Chem. 2013, 288, 5539–5552. [Google Scholar] [CrossRef] [PubMed]
  196. Subtraction Hybridization Identifies a Novel Melanoma Differentiation Associated Gene, mda-7, Modulated during Human Melanoma Differentiation, Growth and Progression—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/8545104/ (accessed on 9 January 2024).
  197. Whitaker, E.L.; Filippov, V.A.; Duerksen-Hughes, P.J. Interleukin 24: Mechanisms and therapeutic potential of an anti-cancer gene. Cytokine Growth Factor Rev. 2012, 23, 323–331. [Google Scholar] [CrossRef] [PubMed]
  198. Stephen-Victor, E.; Fickenscher, H.; Bayry, J. IL-26: An Emerging Proinflammatory Member of the IL-10 Cytokine Family with Multifaceted Actions in Antiviral, Antimicrobial, and Autoimmune Responses. PLoS Pathog. 2016, 12, e1005624. [Google Scholar] [CrossRef] [PubMed]
  199. Spolski, R.; Leonard, W.J. Interleukin-21: Basic biology and implications for cancer and autoimmunity. Annu. Rev. Immunol. 2008, 26, 57–79. [Google Scholar] [CrossRef] [PubMed]
  200. Li, Y.; Li, Y.-F.; Si, C.-Z.; Zhu, Y.-H.; Jin, Y.; Zhu, T.-T.; Liu, M.-Y.; Liu, G.-Y. CCL21/IL21-armed oncolytic adenovirus enhances antitumor activity against TERT-positive tumor cells. Virus Res. 2016, 220, 172–178. [Google Scholar] [CrossRef] [PubMed]
  201. Lamprecht, B.; Kreher, S.; Anagnostopoulos, I.; Jöhrens, K.; Monteleone, G.; Jundt, F.; Stein, H.; Janz, M.; Dörken, B.; Mathas, S. Aberrant expression of the Th2 cytokine IL-21 in Hodgkin lymphoma cells regulates STAT3 signaling and attracts Treg cells via regulation of MIP-3α. Blood 2008, 112, 3339–3347. [Google Scholar] [CrossRef] [PubMed]
  202. Tang, C.; Chen, S.; Qian, H.; Huang, W. Interleukin-23: As a drug target for autoimmune inflammatory diseases. Immunology 2012, 135, 112–124. [Google Scholar] [CrossRef] [PubMed]
  203. Prevention of experimental autoimmune encephalomyelitis by antibodies against interleukin 12. J. Exp. Med. 1995, 181, 381–386. [CrossRef] [PubMed]
  204. Oppmann, B.; Lesley, R.; Blom, B.; Timans, J.C.; Xu, Y.; Hunte, B.; Vega, F.; Yu, N.; Wang, J.; Singh, K.; et al. Novel p19 Protein Engages IL-12p40 to Form a Cytokine, IL-23, with Biological Activities Similar as Well as Distinct from IL-12. Immunity 2000, 13, 715–725. [Google Scholar] [CrossRef] [PubMed]
  205. Bardazzi, F.; Viviani, F.; Piraccini, B.M.; Lasagni, C.; Bigi, L.; Manfredini, M.; Pongetti, L.; Di Lernia, V.; Corazza, M.; Pepe, F. Tildrakizumab in Complex Psoriatic Patients: An Experience in Emilia-Romagna (Italy). J. Cutan. Med. Surg. 2023, 27, 126–132. [Google Scholar] [CrossRef] [PubMed]
  206. Näslund-Koch, C.; Zachariae, C.; Skov, L. Tildrakizumab: An Evidence-Based Review of Its Use in the Treatment of Moderate-to-Severe Chronic Plaque Psoriasis. Ther. Clin. Risk Manag. 2020, 16, 903–916. [Google Scholar] [CrossRef] [PubMed]
  207. Sugaya, M.; Miyagaki, T.; Ohmatsu, H.; Suga, H.; Kai, H.; Kamata, M.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; et al. Association of the numbers of CD163(+) cells in lesional skin and serum levels of soluble CD163 with disease progression of cutaneous T cell lymphoma. J. Dermatol. Sci. 2012, 68, 45–51. [Google Scholar] [CrossRef]
  208. Fahmy, L.M.; Schreidah, C.M.; Lapolla, B.A.; Magro, C.M.; Geskin, L.J. Mycosis fungoides diagnosed after exposure to risankizumab for psoriasis. JAAD Case Rep. 2023, 41, 85–89. [Google Scholar] [CrossRef] [PubMed]
  209. Amitay-Laish, I.; Guenova, E.; Ortiz-Romero, P.L.; Vico-Alonso, C.; Rozati, S.; Geskin, L.J.; Nikolaou, V.; Papadavid, E.; Barzilai, A.; Pavlovsky, L.; et al. The Course of Mycosis Fungoides under Cytokine Pathway Blockers: A Multicentre Analysis of Real-life Clinical Data. Acta Derm. Venereol. 2020, 100, adv00277. [Google Scholar] [CrossRef] [PubMed]
  210. Gowhari Shabgah, A.; Amir, A.; Gardanova, Z.R.; Olegovna Zekiy, A.; Thangavelu, L.; Ebrahimi Nik, M.; Ahmadi, M.; Gholizadeh Navashenaq, J. Interleukin-25: New perspective and state-of-the-art in cancer prognosis and treatment approaches. Cancer Med. 2021, 10, 5191–5202. [Google Scholar] [CrossRef] [PubMed]
  211. Nakajima, R.; Miyagaki, T.; Hirakawa, M.; Oka, T.; Takahashi, N.; Suga, H.; Yoshizaki, A.; Fujita, H.; Asano, Y.; Sugaya, M.; et al. Interleukin-25 is involved in cutaneous T-cell lymphoma progression by establishing a T helper 2-dominant microenvironment. Br. J. Dermatol. 2018, 178, 1373–1382. [Google Scholar] [CrossRef]
  212. Pflanz, S.; Timans, J.C.; Cheung, J.; Rosales, R.; Kanzler, H.; Gilbert, J.; Hibbert, L.; Churakova, T.; Travis, M.; Vaisberg, E.; et al. IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4+ T cells. Immunity 2002, 16, 779–790. [Google Scholar] [CrossRef] [PubMed]
  213. Hisada, M.; Kamiya, S.; Fujita, K.; Belladonna, M.L.; Aoki, T.; Koyanagi, Y.; Mizuguchi, J.; Yoshimoto, T. Potent antitumor activity of interleukin-27. Cancer Res. 2004, 64, 1152–1156. [Google Scholar] [CrossRef] [PubMed]
  214. Salcedo, R.; Hixon, J.A.; Stauffer, J.K.; Jalah, R.; Brooks, A.D.; Khan, T.; Dai, R.-M.; Scheetz, L.; Lincoln, E.; Back, T.C.; et al. Immunologic and Therapeutic Synergy of IL-27 and IL-2: Enhancement of T Cell Sensitization, Tumor-Specific CTL Reactivity and Complete Regression of Disseminated Neuroblastoma Metastases in the Liver and Bone Marrow. J. Immunol. 2009, 182, 4328–4338. [Google Scholar] [CrossRef] [PubMed]
  215. Dibra, D.; Cutrera, J.; Xia, X.; Li, S. WSX1 Expression in Tumors Induces Immune Tolerance via Suppression of Effector Immune Cells. PLoS ONE 2011, 6, e19072. [Google Scholar] [CrossRef] [PubMed]
  216. Nattkemper, L.A.; Martinez-Escala, M.-E.; Gelman, A.B.; Singer, E.M.; Rook, A.H.; Guitart, J.; Yosipovitch, G. Cutaneous T-cell Lymphoma and Pruritus: The Expression of IL-31 and its Receptors in the Skin. Acta Derm. Venereol. 2016, 96, 894–898. [Google Scholar] [CrossRef] [PubMed]
  217. Furue, M.; Yamamura, K.; Kido-Nakahara, M.; Nakahara, T.; Fukui, Y. Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy 2018, 73, 29–36. [Google Scholar] [CrossRef]
  218. Shi, Q.; Abbruzzese, J.L.; Huang, S.; Fidler, I.J.; Xiong, Q.; Xie, K. Constitutive and inducible interleukin 8 expression by hypoxia and acidosis renders human pancreatic cancer cells more tumorigenic and metastatic. Clin. Cancer Res. 1999, 5, 3711–3721. [Google Scholar] [PubMed]
  219. Inoue, K.; Slaton, J.W.; Kim, S.J.; Perrotte, P.; Eve, B.Y.; Bar-Eli, M.; Radinsky, R.; Dinney, C.P. Interleukin 8 expression regulates tumorigenicity and metastasis in human bladder cancer. Cancer Res. 2000, 60, 2290–2299. [Google Scholar] [CrossRef] [PubMed]
  220. Ohmatsu, H.; Sugaya, M.; Suga, H.; Morimura, S.; Miyagaki, T.; Kai, H.; Kagami, S.; Fujita, H.; Asano, Y.; Tada, Y.; et al. Serum IL-31 levels are increased in patients with cutaneous T-cell lymphoma. Acta Derm. Venereol. 2012, 92, 282–283. [Google Scholar] [CrossRef] [PubMed]
  221. Maier, E.; Werner, D.; Duschl, A.; Bohle, B.; Horejs-Hoeck, J. Human Th2 but not Th9 cells release IL-31 in a STAT6/NF-κB-dependent way. J. Immunol. 2014, 193, 645–654. [Google Scholar] [CrossRef] [PubMed]
  222. Olszewska, B.; Żawrocki, A.; Gleń, J.; Lakomy, J.; Karczewska, J.; Zabłotna, M.; Malek, M.; Jankau, J.; Lange, M.; Biernat, W.; et al. Interleukin-31 is overexpressed in skin and serum in cutaneous T-cell lymphomas but does not correlate to pruritus. Postep. Dermatol. Alergol. 2022, 39, 81–87. [Google Scholar] [CrossRef]
  223. Chechlinska, M.; Kowalewska, M.; Nowak, R. Systemic inflammation as a confounding factor in cancer biomarker discovery and validation. Nat. Rev. Cancer 2010, 10, 2–3. [Google Scholar] [CrossRef] [PubMed]
  224. Sloot, Y.J.E.; Smit, J.W.; Joosten, L.A.B.; Netea-Maier, R.T. Insights into the role of IL-32 in cancer. Semin. Immunol. 2018, 38, 24–32. [Google Scholar] [CrossRef] [PubMed]
  225. Ribeiro-Dias, F.; Saar Gomes, R.; de Lima Silva, L.L.; dos Santos, J.C.; Joosten, L.A.B. Interleukin 32: A novel player in the control of infectious diseases. J. Leukoc. Biol. 2017, 101, 39–52. [Google Scholar] [CrossRef] [PubMed]
  226. Lei, J.; Xu, F.; Deng, C.; Nie, X.; Zhong, L.; Wu, Z.; Li, J.; Wu, X.; He, S.; Chen, Y. Fusobacterium nucleatum promotes the early occurrence of esophageal cancer through upregulation of IL-32/PRTN3 expression. Cancer Sci. 2023, 114, 2414–2428. [Google Scholar] [CrossRef] [PubMed]
  227. Zhai, J.-M.; An, Y.-H.; Wang, W.; Fan, Y.-G.; Yao, G.-L. IL-32 expression indicates unfavorable prognosis in patients with colon cancer. Oncol. Lett. 2019, 17, 4655–4660. [Google Scholar] [CrossRef] [PubMed]
  228. Nold-Petry, C.A.; Rudloff, I.; Baumer, Y.; Ruvo, M.; Marasco, D.; Botti, P.; Farkas, L.; Cho, S.X.; Zepp, J.A.; Azam, T.; et al. IL-32 promotes angiogenesis. J. Immunol. 2014, 192, 589–602. [Google Scholar] [CrossRef] [PubMed]
  229. Han, L.; Chen, S.; Chen, Z.; Zhou, B.; Zheng, Y.; Shen, L. Interleukin 32 Promotes Foxp3+ Treg Cell Development and CD8+ T Cell Function in Human Esophageal Squamous Cell Carcinoma Microenvironment. Front. Cell Dev. Biol. 2021, 9, 704853. [Google Scholar] [CrossRef] [PubMed]
  230. Han, S.; Yang, Y. Interleukin-32: Frenemy in cancer? BMB Rep. 2019, 52, 165–174. [Google Scholar] [CrossRef] [PubMed]
  231. van Kester, M.S.; Borg, M.K.; Zoutman, W.H.; Out-Luiting, J.J.; Jansen, P.M.; Dreef, E.J.; Vermeer, M.H.; van Doorn, R.; Willemze, R.; Tensen, C.P. A meta-analysis of gene expression data identifies a molecular signature characteristic for tumor-stage mycosis fungoides. J. Investig. Dermatol. 2012, 132, 2050–2059. [Google Scholar] [CrossRef] [PubMed]
  232. Suga, H.; Sugaya, M.; Miyagaki, T.; Kawaguchi, M.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; Sato, S. The role of IL-32 in cutaneous T-cell lymphoma. J. Investig. Dermatol. 2014, 134, 1428–1435. [Google Scholar] [CrossRef] [PubMed]
  233. Ohmatsu, H.; Humme, D.; Gulati, N.; Gonzalez, J.; Möbs, M.; Suárez-Fariñas, M.; Cardinale, I.; Mitsui, H.; Guttman-Yassky, E.; Sterry, W.; et al. IL32 is progressively expressed in mycosis fungoides independent of helper T-cell 2 and helper T-cell 9 polarization. Cancer Immunol. Res. 2014, 2, 890–900. [Google Scholar] [CrossRef] [PubMed]
  234. Liew, F.Y.; Girard, J.-P.; Turnquist, H.R. Interleukin-33 in health and disease. Nat. Rev. Immunol. 2016, 16, 676–689. [Google Scholar] [CrossRef] [PubMed]
  235. Tamagawa-Mineoka, R.; Katoh, N.; Kishimoto, S. Platelet activation in patients with psoriasis: Increased plasma levels of platelet-derived microparticles and soluble P-selectin. J. Am. Acad. Dermatol. 2010, 62, 621–626. [Google Scholar] [CrossRef] [PubMed]
  236. Wasmer, M.-H.; Krebs, P. The Role of IL-33-Dependent Inflammation in the Tumor Microenvironment. Front. Immunol. 2016, 7, 682. [Google Scholar] [CrossRef] [PubMed]
  237. Rustowska-Rogowska, A.; Gleń, J.; Jarząbek, T.; Rogowski, W.; Rębała, K.; Zabłotna, M.; Czajkowska, K.; Nowicki, R.; Kowalczyk, A.; Sokołowska-Wojdyło, M. Interleukin-33 polymorphisms and serum concentrations in mycosis fungoides. Int. J. Dermatol. 2020, 59, 345–351. [Google Scholar] [CrossRef] [PubMed]
  238. Tsimberidou, A.-M.; Giles, F.J.; Duvic, M.; Kurzrock, R. Pilot study of etanercept in patients with relapsed cutaneous T-cell lymphomas. J. Am. Acad. Dermatol. 2004, 51, 200–204. [Google Scholar] [CrossRef]
  239. Tsimberidou, A.-M.; Alvarado, Y.; Giles, F.J. Evolving role of ribonucleoside reductase inhibitors in hematologic malignancies. Expert. Rev. Anticancer. Ther. 2002, 2, 437–448. [Google Scholar] [CrossRef] [PubMed]
  240. Daliani, D.; Ulmer, R.A.; Jackow, C.; Pugh, W.; Gansbacher, B.; Cabanillas, F.; Duvic, M.; Sarris, A.H. Tumor necrosis factor-alpha and interferon-gamma, but not HTLV-I tax, are likely factors in the epidermotropism of cutaneous T-cell lymphoma via induction of interferon-inducible protein-10. Leuk. Lymphoma 1998, 29, 315–328. [Google Scholar] [CrossRef] [PubMed]
  241. Giri, D.K.; Aggarwal, B.B. Constitutive Activation of NF-κB Causes Resistance to Apoptosis in Human Cutaneous T Cell Lymphoma HuT-78 Cells: Autocrine role of tumor necrosis factor and reactive oxygen intermediates. J. Biol. Chem. 1998, 273, 14008–14014. [Google Scholar] [CrossRef] [PubMed]
  242. Martinez-Escala, M.E.; Posligua, A.L.; Wickless, H.; Rutherford, A.; Sable, K.A.; Rubio-Gonzalez, B.; Zhou, X.A.; Kaplan, J.B.; Pro, B.; Choi, J.; et al. Progression of undiagnosed cutaneous lymphoma after anti-tumor necrosis factor-alpha therapy. J. Am. Acad. Dermatol. 2018, 78, 1068–1076. [Google Scholar] [CrossRef] [PubMed]
  243. Partarrieu-Mejías, F.; Díaz-Corpas, T.; Pérez-Ferriols, A.; Alegre-de Miquel, V. Mycosis fungoides after treatment with tumor necrosis factor-alpha inhibitors for psoriasis: Progression or onset? Int. J. Dermatol. 2019, 58, e103–e105. [Google Scholar] [CrossRef] [PubMed]
  244. Amitay-Laish, I.; Davidovici, B.; Moyal, L.; Gurevich, M.; Akerman, L.; Hodak, E. Lack of detectable effect of narrow-band ultraviolet B on peripheral blood mononuclear cell cytokine expression in early-stage mycosis fungoides. Photodermatol. Photoimmunol. Photomed. 2018, 34, 350–353. [Google Scholar] [CrossRef] [PubMed]
  245. Suga, H.; Sugaya, M.; Toyama, T.; Sumida, H.; Fujita, H.; Kogure, A.; Igarashi, A.; Sato, S. A Case of Mycosis Fungoides with Large Cell Transformation Associated with Infliximab Treatment. Acta Derm. -Venereol. 2014, 94, 233–234. [Google Scholar] [CrossRef] [PubMed]
  246. Adams, A.E.; Zwicker, J.; Curiel, C.; Kadin, M.E.; Falchuk, K.R.; Drews, R.; Kupper, T.S. Aggressive cutaneous T-cell lymphomas after TNFα blockade. J. Am. Acad. Dermatol. 2004, 51, 660–662. [Google Scholar] [CrossRef] [PubMed]
  247. Lourari, S.; Prey, S.; Livideanu, C.; Jamard, B.; Lamant, L.; Cantagrel, A.; Paul, C. Cutaneous T-cell lymphoma following treatment of rheumatoid arthritis with tumour necrosis factor-α blocking agents: Two cases. J. Eur. Acad. Dermatol. Venereol. 2009, 23, 967–968. [Google Scholar] [CrossRef]
  248. Boonstra, J.; Rijken, P.; Humbel, B.; Cremers, F.; Verkleij, A.; van Bergen en Henegouwen, P. The epidermal growth factor. Cell Biol. Int. 1995, 19, 413–430. [Google Scholar] [CrossRef] [PubMed]
  249. Wong, R.W.C.; Guillaud, L. The role of epidermal growth factor and its receptors in mammalian CNS. Cytokine Growth Factor. Rev. 2004, 15, 147–156. [Google Scholar] [CrossRef] [PubMed]
  250. Feng, P.; Catt, K.J.; Knecht, M. Transforming growth factor beta regulates the inhibitory actions of epidermal growth factor during granulosa cell differentiation. J. Biol. Chem. 1986, 261, 14167–14170. [Google Scholar] [CrossRef]
  251. Chen, J.; Zeng, F.; Forrester, S.J.; Eguchi, S.; Zhang, M.-Z.; Harris, R.C. Expression and Function of the Epidermal Growth Factor Receptor in Physiology and Disease. Physiol. Rev. 2016, 96, 1025–1069. [Google Scholar] [CrossRef] [PubMed]
  252. Yun, Y.-R.; Won, J.E.; Jeon, E.; Lee, S.; Kang, W.; Jo, H.; Jang, J.-H.; Shin, U.S.; Kim, H.-W. Fibroblast growth factors: Biology, function, and application for tissue regeneration. J. Tissue Eng. 2010, 2010, 218142. [Google Scholar] [CrossRef] [PubMed]
  253. Paraneoplastic Scleroderma in the Setting of CD30+ Large Cell Transformation of Mycosis Fungoides—PMC. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6357550/ (accessed on 9 January 2024).
  254. Krejsgaard, T.; Lindahl, L.M.; Mongan, N.P.; Wasik, M.A.; Litvinov, I.V.; Iversen, L.; Langhoff, E.; Woetmann, A.; Odum, N. Malignant inflammation in cutaneous T-cell lymphoma—A hostile takeover. Semin. Immunopathol. 2017, 39, 269–282. [Google Scholar] [CrossRef] [PubMed]
  255. Heldin, C.-H.; Lennartsson, J. Structural and functional properties of platelet-derived growth factor and stem cell factor receptors. Cold Spring Harb. Perspect. Biol. 2013, 5, a009100. [Google Scholar] [CrossRef] [PubMed]
  256. Demoulin, J.-B.; Montano-Almendras, C.P. Platelet-derived growth factors and their receptors in normal and malignant hematopoiesis. Am. J. Blood Res. 2012, 2, 44–56. [Google Scholar] [PubMed]
  257. Belinsky, M.G.; Cai, K.Q.; Zhou, Y.; Luo, B.; Pei, J.; Rink, L.; von Mehren, M. Succinate dehydrogenase deficiency in a PDGFRA mutated GIST. BMC Cancer 2017, 17, 512. [Google Scholar] [CrossRef] [PubMed]
  258. Weiner, D.M.; Durgin, J.S.; Wysocka, M.; Rook, A.H. The Immunopathogenesis and Immunotherapy of Cutaneous T Cell Lymphoma: Part II, Current and Future Approaches. J. Am. Acad. Dermatol. 2021, 84, 597–604. [Google Scholar] [CrossRef] [PubMed]
  259. Yoo, E.K.; Cassin, M.; Lessin, S.R.; Rook, A.H. Complete molecular remission during biologic response modifier therapy for Sézary syndrome is associated with enhanced helper T type 1 cytokine production and natural killer cell activity. J. Am. Acad. Dermatol. 2001, 45, 208–216. [Google Scholar] [CrossRef]
  260. Spaccarelli, N.; Rook, A.H. The Use of Interferons in the Treatment of Cutaneous T-Cell Lymphoma. Dermatol. Clin. 2015, 33, 731–745. [Google Scholar] [CrossRef] [PubMed]
  261. Filipi, M.; Jack, S. Interferons in the Treatment of Multiple Sclerosis: A Clinical Efficacy, Safety, and Tolerability Update. Int. J. MS Care 2020, 22, 165–172. [Google Scholar] [CrossRef] [PubMed]
  262. Killestein, J.; Polman, C.H. Determinants of interferon β efficacy in patients with multiple sclerosis. Nat. Rev. Neurol. 2011, 7, 221–228. [Google Scholar] [CrossRef] [PubMed]
  263. Zaidi, M.R.; Merlino, G. The two faces of interferon-γ in cancer. Clin. Cancer Res. 2011, 17, 6118–6124. [Google Scholar] [CrossRef] [PubMed]
  264. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
  265. Dong, H.; Strome, S.E.; Salomao, D.R.; Tamura, H.; Hirano, F.; Flies, D.B.; Roche, P.C.; Lu, J.; Zhu, G.; Tamada, K.; et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 2002, 8, 793–800. [Google Scholar] [CrossRef] [PubMed]
  266. Alspach, E.; Lussier, D.M.; Schreiber, R.D. Interferon γ and Its Important Roles in Promoting and Inhibiting Spontaneous and Therapeutic Cancer Immunity. Cold Spring Harb. Perspect. Biol. 2019, 11, a028480. [Google Scholar] [CrossRef] [PubMed]
  267. Kotenko, S.V.; Durbin, J.E. Contribution of type III interferons to antiviral immunity: Location, location, location. J. Biol. Chem. 2017, 292, 7295–7303. [Google Scholar] [CrossRef]
  268. Kuzel, T.M.; Roenigk, H.H.; Samuelson, E.; Herrmann, J.J.; Hurria, A.; Rademaker, A.W.; Rosen, S.T. Effectiveness of interferon alfa-2a combined with phototherapy for mycosis fungoides and the Sézary syndrome. J. Clin. Oncol. 1995, 13, 257–263. [Google Scholar] [CrossRef] [PubMed]
  269. Zinzani, P.L.; Mazza, P.; Gherlinzoni, F. Beta interferon in the treatment of mycosis fungoides. Haematologica 1988, 73, 547–548. [Google Scholar] [PubMed]
  270. Sarris, A.H.; Esgleyes-Ribot, T.; Crow, M.; Broxmeyer, H.E.; Karasawas, N.; Pugh, W.; Grossman, D.; Deisseroth, A.; Duvic, M. Cytokine Loops Involving Interferon-γ and IP-10, a Cytokine Chemotactic for CD4+ Lymphocytes: An Explanation for the Epidermotropism of Cutaneous T-Cell Lymphoma? Blood 1995, 86, 651–658. [Google Scholar] [CrossRef] [PubMed]
  271. Asadullah, K.; Haeussler, A.; Sterry, W.; Docke, W.; Volk, H. Interferon gamma and tumor necrosis factor alpha mRNA expression in mycosis fungoides progression [letter; comment]. Blood 1996, 88, 757–758. [Google Scholar] [CrossRef] [PubMed]
  272. Sugaya, M.; Tokura, Y.; Hamada, T.; Tsuboi, R.; Moroi, Y.; Nakahara, T.; Amano, M.; Ishida, S.; Watanabe, D.; Tani, M.; et al. Phase II study of i.v. interferon-gamma in Japanese patients with mycosis fungoides. J. Dermatol. 2014, 41, 50–56. [Google Scholar] [CrossRef] [PubMed]
  273. McGinnis, K.S.; Ubriani, R.; Newton, S.; Junkins-Hopkins, J.M.; Vittorio, C.C.; Kim, E.J.; Wysocka, M.; Rook, A.H. The addition of interferon gamma to oral bexarotene therapy with photopheresis for Sézary syndrome. Arch. Dermatol. 2005, 141, 1176–1178. [Google Scholar] [CrossRef]
  274. Gardner, J.M.; Introcaso, C.E.; Nasta, S.D.; Kim, E.J.; Vittorio, C.C.; Rook, A.H. A novel regimen of vorinostat with interferon gamma for refractory Sézary syndrome. J. Am. Acad. Dermatol. 2009, 61, 112–116. [Google Scholar] [CrossRef] [PubMed]
  275. Shimauchi, T.; Sugita, K.; Nishio, D.; Isoda, H.; Abe, S.; Yamada, Y.; Hino, R.; Ogata, M.; Kabashima, K.; Tokura, Y. Alterations of serum Th1 and Th2 chemokines by combination therapy of interferon-gamma and narrowband UVB in patients with mycosis fungoides. J. Dermatol. Sci. 2008, 50, 217–225. [Google Scholar] [CrossRef] [PubMed]
  276. Jonak, C.; Tittes, J.; Brunner, P.M.; Guenova, E. Mycosis fungoides and Sézary syndrome. J. Dtsch. Dermatol. Ges. 2021, 19, 1307–1334. [Google Scholar] [CrossRef] [PubMed]
  277. Willemze, R.; Jaffe, E.S.; Burg, G.; Cerroni, L.; Berti, E.; Swerdlow, S.H.; Ralfkiaer, E.; Chimenti, S.; Diaz-Perez, J.L.; Duncan, L.M.; et al. WHO-EORTC classification for cutaneous lymphomas. Blood 2005, 105, 3768–3785. [Google Scholar] [CrossRef]
  278. Pimpinelli, N.; Olsen, E.A.; Santucci, M.; Vonderheid, E.; Haeffner, A.C.; Stevens, S.; Burg, G.; Cerroni, L.; Dreno, B.; Glusac, E.; et al. Defining early mycosis fungoides. J. Am. Acad. Dermatol. 2005, 53, 1053–1063. [Google Scholar] [CrossRef] [PubMed]
  279. Latzka, J. EORTC consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome—Update 2023. Eur. J. Cancer 2023, 195, 113343. [Google Scholar] [CrossRef] [PubMed]
  280. Trautinger, F.; Eder, J.; Assaf, C.; Bagot, M.; Cozzio, A.; Dummer, R.; Gniadecki, R.; Klemke, C.-D.; Ortiz-Romero, P.L.; Papadavid, E.; 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, 77, 57–74. [Google Scholar] [CrossRef] [PubMed]
  281. Quaglino, P.; Prince, H.M.; Cowan, R.; Vermeer, M.; Papadavid, E.; Bagot, M.; Servitjie, O.; Berti, E.; Guenova, E.; Stadler, R.; et al. Treatment of early-stage mycosis fungoides: Results from the PROspective Cutaneous Lymphoma International Prognostic Index (PROCLIPI) study. Br. J. Dermatol. 2021, 184, 722–730. [Google Scholar] [CrossRef] [PubMed]
  282. Quaglino, P.; Maule, M.; Prince, H.M.; Porcu, P.; Horwitz, S.; Duvic, M.; Talpur, R.; Vermeer, M.; Bagot, M.; Guitart, J.; et al. Global patterns of care in advanced stage mycosis fungoides/Sezary syndrome: A multicenter retrospective follow-up study from the Cutaneous Lymphoma International Consortium. Ann. Oncol. 2017, 28, 2517–2525. [Google Scholar] [CrossRef] [PubMed]
  283. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [PubMed]
  284. Gaydosik, A.M.; Stonesifer, C.J.; Tabib, T.; Lafyatis, R.; Geskin, L.J.; Fuschiotti, P. The mycosis fungoides cutaneous microenvironment shapes dysfunctional cell trafficking, antitumor immunity, matrix interactions, and angiogenesis. JCI Insight 2023, 8, 170015. [Google Scholar] [CrossRef] [PubMed]
  285. Kalliara, E.; Belfrage, E.; Gullberg, U.; Drott, K.; Ek, S. Spatially Guided and Single Cell Tools to Map the Microenvironment in Cutaneous T-Cell Lymphoma. Cancers 2023, 15, 2362. [Google Scholar] [CrossRef] [PubMed]
  286. Xiao, A.; Akilov, O.E. Targeting the CD47-SIRPα Axis: Present Therapies and the Future for Cutaneous T-cell Lymphoma. Cells 2022, 11, 3591. [Google Scholar] [CrossRef] [PubMed]
  287. Johanny, L.D.; Sokumbi, O.; Hobbs, M.M.; Jiang, L. Polarization of Macrophages in Granulomatous Cutaneous T Cell Lymphoma Granulomatous Mycosis Fungoides Microenvironment. Dermatopathology 2022, 9, 54–59. [Google Scholar] [CrossRef] [PubMed]
  288. Patil, K.; Kuttikrishnan, S.; Khan, A.Q.; Ahmad, F.; Alam, M.; Buddenkotte, J.; Ahmad, A.; Steinhoff, M.; Uddin, S. Molecular pathogenesis of Cutaneous T cell Lymphoma: Role of chemokines, cytokines, and dysregulated signaling pathways. Semin. Cancer Biol. 2022, 86, 382–399. [Google Scholar] [CrossRef] [PubMed]
  289. Liu, J.; Zhu, S.; Tang, W.; Huang, Q.; Mei, Y.; Yang, H. Exosomes from tamoxifen-resistant breast cancer cells transmit drug resistance partly by delivering miR-9-5p. Cancer Cell Int. 2021, 21, 55. [Google Scholar] [CrossRef] [PubMed]
  290. Bresin, A.; Caprini, E.; Russo, G.; Narducci, M.G. Challenging Cutaneous T-Cell Lymphoma: What Animal Models Tell us So Far. J. Investig. Dermatol. 2022, 142, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  291. de Masson, A.; Darbord, D.; Dobos, G.; Boisson, M.; Roelens, M.; Ram-Wolff, C.; Cassius, C.; Le Buanec, H.; de la Grange, P.; Jouenne, F.; et al. Macrophage-derived CXCL9 and CXCL11, T-cell skin homing, and disease control in mogamulizumab-treated CTCL patients. Blood 2022, 139, 1820–1832. [Google Scholar] [CrossRef] [PubMed]
  292. Barta, S.K.; Liu, N.; DerSarkissian, M.; Chang, R.; Ye, M.; Duh, M.S.; Surinach, A.; Fanale, M.; Yu, K.S. Real-World Treatment Patterns and Clinical Outcomes with Brentuximab Vedotin or Other Standard Therapies in Patients with Previously Treated Cutaneous T-Cell Lymphoma in the United States. Clin. Lymphoma Myeloma Leuk. 2023, 24, e21–e32.e4. [Google Scholar] [CrossRef] [PubMed]
  293. Stuver, R.; Geller, S. Advances in the treatment of mycoses fungoides and Sézary syndrome: A narrative update in skin-directed therapies and immune-based treatments. Front. Immunol. 2023, 14, 1284045. [Google Scholar] [CrossRef] [PubMed]
  294. Kumar, A.; Vardhana, S.; Moskowitz, A.J.; Porcu, P.; Dogan, A.; Dubovsky, J.A.; Matasar, M.J.; Zhang, Z.; Younes, A.; Horwitz, S.M. Pilot trial of ibrutinib in patients with relapsed or refractory T-cell lymphoma. Blood Adv. 2018, 2, 871–876. [Google Scholar] [CrossRef] [PubMed]
  295. Duvic, M. Treatment of Cutaneous T-Cell Lymphoma from a Dermatologist’s Perspective. Clin. Lymphoma 2000, 1, S15–S20. [Google Scholar] [CrossRef] [PubMed]
  296. Sibbesen, N.A.; Kopp, K.L.; Litvinov, I.V.; Jønson, L.; Willerslev-Olsen, A.; Fredholm, S.; Petersen, D.L.; Nastasi, C.; Krejsgaard, T.; Lindahl, L.M.; et al. Jak3, STAT3, and STAT5 inhibit expression of miR-22, a novel tumor suppressor microRNA, in cutaneous T-Cell lymphoma. Oncotarget 2015, 6, 20555–20569. [Google Scholar] [CrossRef] [PubMed]
  297. Vadivel, C.K.; Gluud, M.; Torres-Rusillo, S.; Boding, L.; Willerslev-Olsen, A.; Buus, T.B.; Nielsen, T.K.; Persson, J.L.; Bonefeld, C.M.; Geisler, C.; et al. JAK3 Is Expressed in the Nucleus of Malignant T Cells in Cutaneous T Cell Lymphoma (CTCL). Cancers 2021, 13, 280. [Google Scholar] [CrossRef] [PubMed]
  298. Nielsen, M.; Kæstel, C.G.; Eriksen, K.W.; Woetmann, A.; Stokkedal, T.; Kaltoft, K.; Geisler, C.; Röpke, C.; Ødum, 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. [Google Scholar] [CrossRef] [PubMed]
  299. Castillo, D.E.; Romanelli, P.; Lev-Tov, H.; Kerdel, F. A case of erythrodermic mycosis fungoides responding to upadacitinib. JAAD Case Rep. 2022, 30, 91–93. [Google Scholar] [CrossRef] [PubMed]
  300. Moskowitz, A.J.; Ghione, P.; Jacobsen, E.; Ruan, J.; Schatz, J.H.; Noor, S.; Myskowski, P.; Vardhana, S.; Ganesan, N.; Hancock, H.; et al. A phase 2 biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas. Blood 2021, 138, 2828–2837. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Potential and actual therapies to address CTCLs and their relative targets.
Figure 1. Potential and actual therapies to address CTCLs and their relative targets.
Cells 13 00584 g001
Table 1. List of described cytokines, their main mechanisms and implications in MF/SS, and their potential therapeutic roles.
Table 1. List of described cytokines, their main mechanisms and implications in MF/SS, and their potential therapeutic roles.
CytokineFunctionRole in MF/SSPotential Targeted Therapies Evaluated
IL-1Pro-inflammatory [17].Elevated in treated patient; potential biomarker of photopheresis response [19,20].No studies available
IL-2Pro-inflammatory.
Upregulate T cells and to increase the cytotoxicity of monocytes and natural killer (NK) [21].
May have an CTCL suppressive action, but still controversial [22,23].AS101 inihbits IL2R and increases IL-2, suggesting an immunosuppressive role [24].
Denileukin Diftitox, which seems to reach a response of 36–40% of CTCLs in some studies [25].
CCR4-IL2 IT, in pre-clinical models seems to be promising in inducing CTCLs remission [26].
IL-3Pluripotent and hematopoietic factor required for survival and proliferation of hematopoietic progenitor cells [27].Unknown.No studies available.
IL-4Negatively modulates Th1 T cells; skews to a Th2 phenotype of naïve T cells [28].Contributes to immune evasion and tumor progression microenvironments (still debated) [29].
Can (with IL-33) induce IL-31 secretions, involved in itch pathogenesis [28,30].
Anti PD-1 (Nivolumab) could reduce malignant Th2 cells, but it is controversial [31,32].
JAK inhibitor (Ritlecitinib) showed promising effect in reducing Th2 neoplastic cells (IIA trial ongoing) [33].
Dupilumab, by blocking IL-4 and Il-13, could induce an immune system against the tumor and blocking of Th2 cells’ proliferation, but is debated due to reported misdiagnosed MFs treated with dupilumab with a dramatic progression [34,35,36].
IL-5Stimulates eosinophilic cascade [37].Related to erythroderma and elevated serum levels of IgE.
Seems to be overexpressed by CTCL T cells [38].
AS101 increases IFN-γ and a decreases Il-5, so to affect CTCL progression [24,39].
IL-6Pro-inflammatory.
Differentiates plasma cells and increases adhesion molecule production [40].
Hyperxpressed in CTCL samples. Seems to be related to a higher risk of MF progression [41,42]. Il-6 polymorphism may be related to worse disease prognosis [43].No studies available.
IL-7Hemapoietic factor stimulates the development of lymphoid lineage [44].Linked to activation of CD8 + SS clone cells, but studies are not concordant [45,46].No studies available.
IL-8IL-8 is a chemotactic factor for neutrophils and other granulocytes. The oncogenic role is achieved by binding the IL-8 R localized on cancer cells and on microenvironment cells [47].Involved in pruritus and CTCL progression [48,49].No studies available.
IL-9Stimulates various hematopoietic cells’ proliferation and prevents immune cells’ apoptosis.
Seems to be related to hematologic neoplasias [50].
High levels have been linked in patients with SS [51] and MF [52].No studies available.
IL-10Anti-inflammatory.
Prevention of autoimmune diseases.
Can contribute to infection and tumor progression [53,54].
Higher levels of IL-10 have been detected in MF/SS biopsies compared with normal skin [55,56].Vorinostat and romidepsin may exert their therapeutic action due to the downregulation of IL-10 RNA expression [56].

Bortezomib modulates cytokine expression in CTCL, acting on TGFβ1 and IL-10 down-regulation [57].
IL-11Anti-inflammatory properties.
Hematopoiesis, production of platelets from megakaryocytes and hemostasis [58].
Unknown.No studies available.
IL-12Activates NK cells and promotes the differentiation of Th1 cells [59].Reduces or suppresses IL-12 pathway in CTCL developement, especially in SS patients [60].Recombinant IL-12 may restore NK cells functions in CTCLs [61].
IL-13Anti-inflammatory.
Related to Th2 axis [62].
IL-13 may act as an autocrine factor in lymphoma cell proliferation through IL-13Rα1 and IL-13-Rα2 signaling [63].

A higher expression of IL-13 and its receptors correlates with late stages, while in the early stages, the expression is low [63].
Dupilumab, by blocking IL-4 and IL-13, could induce an immune system against the tumor and halt Th2 cell proliferation, but this is debated due to reported misdiagnosed MFs treated with dupilumab with a dramatic progression [34,35,36].
IL-14Growth of B cells. Produced by T cells and certain malignant B cells [64].Unknown.No studies available.
IL-15Enhances CD8 T cell cytotoxic activity, B cell differentiation, Ig synthesis, and DC maturation [65].This is implied in the recruitment of CD4+ memory T cells to the skin, induction of T cell proliferation, and inhibition of apoptotic cell death [65,66].
Linked to later stages of CTCL and assumed to promote disease progression [67].
HDAC inhibitors [68] could halt Zeb1, which leads to an overexpression of IL-15 [69].
IL-16Pro-inflammatory [70].Related to early MF stages [70].No studies available.
IL-17Enhances immune response against infectious agents. Upregulates pro-inflammatory cytokines, chemokines, metalloproteinases, and antimicrobial peptides [71].Low expression of IL-17 mRNA levels in MF/SS samples compared to healthy donors [66,72]. No studies available.
IL-18Has a role in adaptive immunity. Induces IFN-γ production [73].Higher expression in all MF stages compared to control cases. Role in tumor escape in SS [74].No studies available.
IL-19Pro-inflammatory [75].IL-19 levels correlated positively with HMGB1, a protein associated with angiogenesis, Th2 polarization, and CTCL progression [76]No studies available.
IL-20Immunosuppression [75].Unknown.No studies available.
IL-21IL-21 enhances cytotoxicity and induces the production of IFN-γ and perforin by NK cells [77].Unknown.No studies available.
IL-22Immunosuppression [54].IL-22 could play a role in establishing the tumor microenvironment in MF [78]. No studies available.
IL-23Pro-inflammatory [79].Unknown.No studies available.
IL-24Pro-inflammatory [54].Unknown.No studies available.
IL-25Causes Th2 phenotype polarization and IL-4, IL-5, and IL-13 production. It inhibits TH1 and TH17 responses through inhibition of IL-12 and IL-23 [80].Higher expression in MF and SS epidermal keratinocytes, compared to controls. IL-25 levels in skin lesions related to disease progression and serum levels correlated with LDH levels [41].
IL-25 enhances IL-13 production by tumor shifting to a Th2 dominant microenvironment [80].
No studies available.
IL-26Pro-inflammatory [54].Unknown.No studies available.
IL-27IL-27 promotes Th1 immunity, IFN-γ production by NK and T cells, and inhibits Th2 response [81].IL-27 levels were higher in advanced stages compared to early stages or controls in MF [81,82].No studies available.
IL-28See IFN-γ.Unknown.No studies available.
IL-29See IFN-γ.Unknown.No studies available.
IL-30Pro-inflammatory [81].Unknown.No studies available.
IL-31Produced by CD4+ Th2 cells, mast cells, and dendritic cells. It modulates fibroblasts and eosinophils.Increased levels have been detected in both serum and skin lesions [83].
It seems to be related to pruritus, but is still debated [49,84,85].
No studies available.
IL-32Immunosuppression and cancer progression [30].IL-32 is associated with MF development and progression [86].No studies available.
IL-33Causes M2 macrophages’ differentiation and induces maturation of dendritic cells [87].
Promotes Th1-mediated responses, including cell-mediated cytotoxicity [88].
IL-33 might accelerate MF progression via a paracrine action in the tumor microenvironment [89,90].No studies available.
TNF-APro-inflammatory [91].TNF–α has been implicated in the development of CTCL by the promotion of epidermotropism via the induction of interferon-inducible protein. TNF-a acts as an autocrine growth factor, enhancing its tumorigenic action and empowering the NF/KB pathway.No studies available.
EGFEpidermal stem cells’ proliferation and differentiation [92].Possible role due to its known effect in inhibiting IFN type 1, low levels of which are related to MF development [91].No studies available.
FGFMesenchimal stem cells’ proliferation, survival, migration, and differentiation [93].FGF’s role has been hypothesized in paraneoplastic scleroderma in MF [93].No studies available.
PDGFRVarous cellular lines’ growth, proliferation, and differentiation [94].Unknown.No studies available.
IFN [95,96]Pro-inflammatory [97,98,99,100].
-
IFN-α presents an antitumor action by empowering CD8+ and NK cells’ action against MF cells. IFN-α reduces the Th2 cytokinetic milieu and IL-4/IL-13 overexpression [95].
-
IFN-β’s role is under investigation [95].
-
IFN-γ is involved in epidermotropism pathogenesis [89].
-
IFN-λ’s role is unknown.
The administration of IFN -α is a well-known efficient treatment in advanced-stage CTCLS [96,97,98,99,100,101].

Recombinant IFN-γ a in monotherapy or in association with other therapies, such as phototherapy, bexarotene, vorinostat, and ifn-α, did not provide clear-cut results due to the small sample size of the studies [96,97,98,99,100,101].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guglielmo, A.; Zengarini, C.; Agostinelli, C.; Motta, G.; Sabattini, E.; Pileri, A. The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets. Cells 2024, 13, 584. https://doi.org/10.3390/cells13070584

AMA Style

Guglielmo A, Zengarini C, Agostinelli C, Motta G, Sabattini E, Pileri A. The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets. Cells. 2024; 13(7):584. https://doi.org/10.3390/cells13070584

Chicago/Turabian Style

Guglielmo, Alba, Corrado Zengarini, Claudio Agostinelli, Giovanna Motta, Elena Sabattini, and Alessandro Pileri. 2024. "The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets" Cells 13, no. 7: 584. https://doi.org/10.3390/cells13070584

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop