Next Article in Journal
Review on the Use of Heavy Metal Deposits from Water Treatment Waste towards Catalytic Chemical Syntheses
Next Article in Special Issue
The Role of Tumor Microenvironment in the Pathogenesis of Sézary Syndrome
Previous Article in Journal
GPCRs Are Optimal Regulators of Complex Biological Systems and Orchestrate the Interface between Health and Disease
Previous Article in Special Issue
Malignant and Benign T Cells Constituting Cutaneous T-Cell Lymphoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Safety and Danger Considerations of Novel Treatments for Atopic Dermatitis in Context of Primary Cutaneous Lymphomas

by
Karol Kołkowski
1,*,
Magdalena Trzeciak
2 and
Małgorzata Sokołowska-Wojdyło
2
1
Dermatological Students Scientific Association, Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland
2
Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(24), 13388; https://doi.org/10.3390/ijms222413388
Submission received: 3 November 2021 / Revised: 5 December 2021 / Accepted: 6 December 2021 / Published: 13 December 2021
(This article belongs to the Special Issue Pathogenesis and Treatment of Cutaneous Lymphoma)

Abstract

:
The impact of new and emerging therapies on the microenvironment of primary cutaneous lymphomas (PCLs) has been recently raised in the literature. Concomitantly, novel treatments are already used or registered (dupilumab, upadacitinib) and others seem to be added to the armamentarium against atopic dermatitis. Our aim was to review the literature on interleukins 4, 13, 22, and 31, and JAK/STAT pathways in PCLs to elucidate the safety of using biologics (dupilumab, tralokinumab, fezakinumab, nemolizumab) and small molecule inhibitors (upadacitinib, baricitinib, abrocitinib, ruxolitinib, tofacitinib) in the treatment of atopic dermatitis. We summarized the current state of knowledge on this topic based on the search of the PubMed database and related references published before 21 October 2021. Our analysis suggests that some of the mentioned agents (dupilumab, ruxolitinib) and others may have a direct impact on the progression of cutaneous lymphomas. This issue requires further study and meticulous monitoring of patients receiving these drugs to ensure their safety, especially in light of the FDA warning on tofacitinib. In conclusion, in the case of the rapid progression of atopic dermatitis/eczema, especially in patients older than 40 years old, there is a necessity to perform a biopsy followed by a very careful pathological examination.

1. Introduction

Primary cutaneous lymphomas (PCLs) are a rare entity of lymphoproliferative disorders that have no evidence of extracutaneous involvement at the time of diagnosis [1]. An important impact of the tumor microenvironment on the progression of the disease has been raised in literature [2]. Currently, a variety of drugs affecting the cytokines and pathways are essential in the pathogenesis of atopic dermatitis (AD) and are in the clinical trials phase, whereas dupilumab targeting interleukin-4 (IL-4) and interleukin-13 (IL-13), tralokinumab targeting IL-13 and two Janus kinase inhibitors (JAKi): upadacitinib (JAK1 inhibitor) and baricitinib (JAK1/JAK2 inhibitor), are already registered in the EU [3,4]. Agents blocking interleukin-22 (IL-22) and interleukin-31 (IL-31), fezakinumab, and nemolizumab, as well as lebrikizumab will be available for patients soon [3]. There is a controversy regarding a potential of increased risk of lymphoma in patients with atopic dermatitis (AD). Our aim is to elucidate the role of IL-4, IL-13, IL-22, IL-31, and the JAK/STAT pathway in PCLs in the context of novel treatment of AD.

2. Discussion

AD is a chronic, inflammatory skin disease characterized by strong pruritus that less commonly affects adults [5]. This condition is associated with a poorer quality of life in comparison with the general population and causes sleep disturbances and coexisting comorbidities [6]. As reported by the epidemiological studies, the prevalence of the childhood AD is between 12% and 20% in the United States, Europe, and Eastern Asia, whereas in the elderly population it ranges from 2% to 5% [7,8,9,10,11,12]. Moreover, the secular trends tend to show an increase in the number of AD patients in both children and adults [9,10]. Unfortunately, a significant number of these patients present moderate to severe AD. Despite the scale of the problem, the arsenal of drugs with a safe profile of action, characterized by a low risk of serious side effects, and appropriate for long-term use is scarce [13]. Therefore, doctors and patients hope for the end of “the draught”, which may happen thanks to biologic drugs, e.g., monoclonal antibodies (mAb) like dupilumab/tralokinumab or small molecule inhibitors, e.g., upadacitinib/baricitinib, which are proven to be effective and are registered in the EU [13]. In fact, a few of these medications are already approved for topical and systemic treatment of AD. However, despite the unquestionable potential these drugs hold for AD patients in relieving their burden, we believe that some important issues must be raised.
Among PCLs, heterogenous groups of B-, T- and NK-Cell lymphomas have been differentiated [1]. Mycosis fungoides (MF) belongs to cutaneous T-cell lymphoma (CTCL) and its classical variant is the most common PCL [1]. Our review focuses on the CTCLs; however, when PCLs are mentioned, we refer to the entire spectrum of primary cutaneous lymphomas. Major meta-analysis has shown a relative risk ratio (RR) of developing a lymphoma of 1.43 (95% CI, 1.12–1.81) in patients with AD [14]. The risk of lymphoma is higher in cases where highly potent TCSs are used and in a severe course of the disease [14]. In a recent study, the hazard ratios of developing Non-Hodgkin’s lymphoma (NHL) increase with the severity of the eczema [15]. This was the only epidemiological study in which we could find any biologic drug taken into consideration. Dupilumab has been analyzed in the Danish cohort together with the influence of other immunosuppressive drugs, including cyclosporine, azathioprine, mycophenolate and methotrexate [15]. According to some studies, the risk of developing NHL with cutaneous manifestation is especially high, but we have to bear in mind the possible misdiagnosis bias [14,15,16]. We were not able to find any other studies that describe the incidence of lymphomas in patients treated with biologics or small molecule inhibitors referring to AD except clinical trials and case reports. Incidence of lymphomas in the mentioned studies will now become a baseline for the further analysis of the effects of new immunosuppressives brought to the market.
It may be difficult to clinically differentiate AD and PCL, especially in the case of erythroderma. If a patient develops adult-onset AD, erythrodermic CTCL, and Sézary syndrome (SS), they should always be excluded, as these diseases require distinct treatment and have drastically varying prognoses [17]. Similarities concerning both diseases, which are crucial in their pathogenesis, are illustrated in the Table 1 [17,18,19,20,21,22,23,24,25,26,27,28].
Some authors indicate that, due to the significant quantity of similarities, both diseases may require the same treatment at certain stages [29]. However, in our opinion, the safety of emerging drugs used in AD treatment, in the context of a PCL coexistence/induction risk, should be raised. We decided to analyze theoretical and clinical data regarding interleukins and JAK-STAT pathways, which recently have been proven to be attractive targets in the treatment of AD. On that basis, we excluded IL-5, IL-17, and IL-33 from the analysis, as the trials of drugs targeting them are either terminated, are of unknown status, or they did not meet the primary endpoints [30,31,32,33,34,35]. In this review, we also omit the IL-12/IL-23 axis affected by ustekinumab for two reasons. Firstly, a recent review on the effectiveness of this agent concluded that the IL-12/IL-23 pathway is not an attractive target for the treatment of AD [36]. Furthermore, the largest cohort of patients receiving ustekinumab has shown that more novel and effective treatments are available for the therapy of this atopic disease [36]. Second, IL-12 has been shown to be one of the possible treatments, despite the fact that it is not currently clinically developed [37]. Therefore, blocking it should be a factor facilitating the lymphoma progression by down-regulating the Th-1 cytotoxicity against malignant clones.

2.1. New Medications in AD

AD is thought to be the hallmark of Th-2 microenvironment diseases. Th-2 profile cytokines, such as IL-4, IL-5 and IL-13, play a significant role in the pathogenesis of the disease by switching the immunoglobulin class to IgE and stimulating afferent neurons via IL-4Rα, thereby promoting pruritus [38]. Therefore, drugs blocking these pathways should be clinically effective in reducing the symptoms of this eczematous disease, as they act against the inflammation [39].
One of them is dupilumab—a fully human monoclonal antibody that blocks IL-4Rα, a shared receptor unit for IL-4 and IL-13, actively participating in the decrease of Th-2 mediated immunological response [3]. It is already used in America, Europe, and in several other countries on children, adolescents, and adults. The analysis of four phase-three trials has revealed that patients treated with this monoclonal antibody achieve a significantly higher percentage reduction from the baseline in the most important AD management scales—Eczema Area and Severity Index (EASI), SCORing Atopic Dermatitis (SCORAD), Dermatology Life Quality Index (DLQI), and Patient-Oriented Eczema Measure (POEM) versus control [40]. Notably, these superior effects have been achieved in monotherapy without topical corticosteroids, regardless of previous use of systemic non-steroidal immunosuppressants, e.g., methotrexate or cyclosporine [40].
Other drugs targeting the IL-13 are lebrikizumab and tralokinumab. IL-13 binds and neutralizes the activity of the mentioned cytokine with high affinity [41]. In phase IIb of several randomized clinical trials, it showed promising results [42,43]. Even though adverse effects of this drug were reported in the significant group of patients, they were mostly mild to moderate [42,43]. Phase III clinical trials on patients who suffer from moderate to severe AD are currently ongoing [44,45,46,47,48,49,50]. Another promising emerging drug is tralokinumab—a fully human, monoclonal anti-IL-13 IgG4 antibody that binds to two subunits of IL-13R (IL-13Rα1 and IL-13Rα2), thus neutralizing the cytokine from the interaction [3,51]. Recently, three phase III clinical trials (ECZTRA1, ECZTRA2, and ECZTRA3) were completed for this drug [52,53]. Tralokinumab, in combination with topical corticosteroids, is not only effective in reducing the pruritus and improving sleep quality, but it is also well tolerated for up to 52 weeks of treatment, which brings a promising perspective we mentioned earlier [52]. Moreover, this medicament is safe and well tolerated in combination with topical corticosteroids [53]. Interestingly, a long-term extension trial for patients who were participants in the previous studies is currently ongoing and the estimated completion date is in 2024 [54].
IL-22 and IL-31 are also the targets of new drugs, which have been or currently still are under investigation in phases IIa and III of clinical trials [55,56,57]. Fezakinumab, an anti-IL-22 antibody, has been shown in the IIa randomized, double-blind clinical trial on adults with moderate to severe AD to be well tolerated and to have sustainable improvements after the last dose [55]. Despite the small sample size and common adverse effects, which were upper respiratory tract infections, improvements in SCOring AD (SCORAD) were significant in patients with severe disease [3,55]. Thus, this drug is thought to be suitable for patients with severe AD, but no further clinical trials are currently ongoing [3]. Another interesting medication, especially for managing the pruritus in patients with AD is nemolizumab, a human monoclonal IL-31 receptor α (IL-31Rα) antagonist [3,57,58]. This drug targets small-diameter neurons and it is thought that the relieving effect of nemolizumab is due to action on cutaneous sensory neurons [3,58]. In the phase III trial, the patients who could not achieve proper control of pruritus by solely using topical treatment were recruited and enrolled [56]. Not only were the primary end points of the study achieved with a significant decrease in pruritus measured in the VAS scale, but also a series of secondary endpoints including EASI, DLQI or Insomnia Severity Index were met [56]. Other phase III trials are currently ongoing [59].
Various systemic and topical JAK inhibitors are about to be widely used in the treatment of AD [4]. The data on the double blind control trials evaluating the efficacy of these drugs in the treatment of AD are promising [60,61]. Baricitinib, abrocitinib, and upadacitinib belong to the group of oral drugs, while ruxolitinib is known as a topical agent considered in the therapy of AD [60].
Baricitinib is known as the first-generation JAK1/2 selective inhibitor [60,62,63]. The efficacy of the drug in monotherapy and combined with topical corticosteroids has been evaluated and the dose of 4 mg appears to significantly improve symptoms [64,65,66]. In the pooled safety analysis of baricitinib in adults, which contained previously mentioned studies, there were four major cardiovascular-adverse events and one death, however, no malignancies were reported [67].
Abrocitinib is an oral selective JAK1 inhibitor that achieved satisfying results in the phase III trial, proving that it is effective and well tolerated in monotherapy [64,68]. Patients from these studies have been enrolled in the extended trial (NCT03422822) and in the 48th week of this trial, it has been shown, that between 24 and 36 weeks, the proportion of patients meeting primary endpoints increased and was stable thereafter [60]. Comparing abrocitinib, dupilumab, and the placebo in clinical trial, both drugs significantly more reduced AD symptoms; however, a 200 mg dose of abrocitinib was superior to dupilumab in limiting itchiness [69].
The next oral selective JAK1 inhibitor is upadacitinib [60]. It safe and efficient in the monotherapy of moderate to severe AD in three phase III trials [70,71]. Moreover, in comparison with dupilumab it was superior, showing significantly higher proportion of patients who achieved the primary and secondary endpoints of the study [72]. Extension of the mentioned trials and also new ones with pediatric patients are ongoing [73,74].
Ruxolitinib (JAK1/2 inhibitor) and delgocitinib (pan-JAK inhibitor) have proved to be effective topical drugs in AD [60,61]. In the two phase III trials, ruxolitinib has shown anti-inflammatory and anti-pruritic effects superior to the vehicle cream [75]. Adverse effects were infrequent and clinically insignificant [75]. Clinical trials with atopic children are underway [76]. Delgocitinib also seems to be satisfactory, since in the phase III trial it was effective and well tolerated in Japanese patients for up to 28 weeks [77]. Currently, two phase III trials on moderate to severe chronic hand eczema are ongoing [78,79].

2.2. Role of Interleukin-4 and Interleukin-13 in PCL

Interleukin-4 (IL-4) and interleukin-13 (IL-13) are the characteristic molecules that induce, drive, and prolong the Th-2 answer both in AD and in advanced stages of CTCL [21,22,23,38]. Along with the progression of CTCL, Th-2 cytokines are most commonly overproduced, skewing the Th1/Th2 axis towards the latter side, a well-known phenomenon [80,81,82]. Certain genetic markers are associated with the predisposition to develop the progressive MF. One of them has an increased expression of IL-4 relative to CD3 expression levels, which are significantly associated with lymphoma progression [83]. Each of the discussed cytokines are elevated in the biopsies of MF and SS lesions [84,85,86,87]. IL-4 levels are raised in sera of patients; however, it does not always concern the cases of low-grade lymphoma [18,21,81,82,84,88]. The elevated concentrations of these cytokines are related to the ability of neoplastic cells to secrete IL-4 and IL-13 as well as in the skin and in the blood in vivo [81,82,85].
Also, IL-4 is a potent factor that polarizes tumor-associated macrophages into type 2 cells (M2 Macrophages) [89]. The ability to produce several Th-2 cytokines is characteristic for these phagocytes, thereby affecting the formation of CTCL by stromal factors [86,89]. Furthermore, IL-4 and IL-13 are important growth factors for PCLs and IL-13 acts in an autocrine manner on the neoplastic lymphocytes [81,87]. IL-4 and IL-33 are also found to induce the secretion of IL-31, which is one of the elements alongside with the discussed interleukins, causing pruritus in AD and in CTCL—but the data are ambivalent concerning lymphomas [3,90]. These properties contribute to driving the Th-2 type inflammatory answer propelled in a vicious circle, leading to the depletion of the Th-1 microenvironment. The arrest of the IL-4/IL-13 pathway by neutralizing IL-4 and IL-13 cytokines leads to inhibition of tumor-cell proliferation [81]. Interestingly, blocking certain types of IL-13 receptors (IL-13Rα2) revealed an even stronger inhibition effect. However, this receptor binds with IL-13 stronger than the first IL-13 receptor (IL-13Rα1) and is thought to be a decoy in the normal tissues [81,91]. Recent studies have shown that the tumoral microenvironment created by the malignant lymphocytes in leukemic CTCL is a global bias, which refers also to benign T cells [82]. Thus, in comparison to the normal lymphocytes in healthy individuals, non-tumorous cells are strongly Th-2 biased [82]. Such drastic reduction of the cytotoxic environment is thought to be one of major factors leading to infections which is the most common reason of death in this group of patients [1,92,93].
It is also proven that cytoplasmic IL-4 concentration is the predictor of the advanced stage of MF and SS [94]. Moreover, increased IL-4 concentration is observed frequently in advanced stages of CTCL; it correlates with T-cell immunophenotype differences found in advanced lymphoma stages and is associated with clonality of MF and SS cells [94]. Some available methods of treatment used in clinical practice can reduce the Th-2 polarization in advanced stages of CTCLs. One study revealed that extracorporeal photopheresis (ECP) effectively restores the imbalance in Th1/Th2 microenvironments of peripheral blood mononuclear cells (PBMC) [95]. After one year of such therapy, the concentrations of IL-4, interferon gamma (IFNγ), and IL-12 did not differ from the healthy controls [95]. Also, after administration of the T-cell depleting antibody directed against CD52, alemtuzumab (Campath), which is used to treat refractory leukemic CTCL (L-CTCL), skin T cells have been shown to secrete less IL-4 and more interferon gamma (IFNγ) than before the treatment [96]. Finally, Guenova et al. suggested that inhibiting the Th-2 microenvironment and restoration of Th-1 cytotoxicity should enhance both anti-tumorous and antibacterial responses [82].
Concluding this section, the listed Th-2 cytokines play an essential role in the pathogenesis of PCLs. It is especially prominent in advanced stages of the disease. Therefore, blocking these pathways may be beneficial and may result at least in the stabilization of the lymphoma.

2.3. Role of Interleukin-22 in PCL

Interleukin-22 (IL-22) is secreted mainly by the subpopulation of Th22 lymphocytes, but also by other immune cell subsets and is involved in the modification of tissue responses at the inflammation [97]. In two studies and one case report, levels of this cytokine were significantly elevated in both skin lesions and sera of CTCL patients [18,98,99]. Other researchers found that cultured CTCL cells overexpress IL-22 receptor subunit alpha1 (IL22Rα1) and overproduce IL-22 as well as chemokine ligand 20 (CCL20) [100]. Cytokine induces the expression of CCL20 and signal transducer and activator of transcription 3 (STAT3), the latter of plays a role in the pathogenesis of CTCL [98,101]. Moreover, the Sézary cells of one patient who developed sepsis stained positive for CD8 and produced IL-22 [98]. Genetic analysis of SS patients and SS cell lines (SaEx) showed the disruption in the IL-22 receptor subunit alpha2 (IL22Rα2) gene twice [102]. Subsequently, a fusion of several genes occurred and one of them was the CCDC28A-IL22Rα2, which was transcribed on the messenger RNA level [102]. CCL20 and IL-22 serum levels correlate with the LDH and sIL-2R and thus they correlate with CTCL severity [18]. Their activity seems significant in the pathogenesis of the disease.
Enhanced CCL20 activity may induce epidermal hyperplasia as well as the migration of chemokine receptor 6 (CCR6) positive Langerhans/Dendritic cells to the skin, which are crucial in the evolution of lymphoma cells as the activation of T-cell receptors is crucial for the malignant transformation of MF [18,103]. CCL20 is a ligand of CCR6 [100]. Activation of IL-22 has also been shown to lead to chronic CCR6-CCL20 interaction with CTCL cells [100]. Furthermore, the continuous upregulation of CCR6 was discovered, which results in the continuous activation of CCR6-CCL20, leading the lymphoma cells to metastasize to internal organs [100]. These important findings are supported by recent studies, showing that triggering a STAT3/CCL20/CCR6 cascade that blocks CCR6-CCL20 interaction, may be crucial in stopping lymphomagenesis [104]. Today, it is considered one of the promising strategies in the treatment of advanced CTCL [104].

2.4. Role of Interleukin-31 in PCL

The role of Interleukin-31 (IL-31) in PCL is difficult to establish despite numerous studies covering this topic. Importantly in other lymphomas, such as follicular lymphoma, IL-31 promotes the growth of tumors in an autocrine and paracrine manner [90]. Certainly, this cytokine seems to be involved in the pathogenesis of PCLs [105]. It is typically secreted by Th-2 cells [106]. Signal transducers and activators of transcription 6 (STAT6) and NF-κB induced by IL-4 are main players in mediating the production of IL-31 [106]. Both STAT6 and NF-κB play some role in the pathogenesis of CTCL [101,107]. IL-31 is elevated in both lesions and sera of patients in the majority of studies (five); however, one study found no differences in comparison to control groups [54,105,108,109,110,111]. Malignant T-cells may secrete IL-31 [109]. Researchers did not establish with certainty whether IL-31 concentration is correlated with CTCL progression or/and pruritus [54,105,108,109,110,111]. Despite the proven central role of this cytokine in mediating pruritus in AD patients and the assumptions to have the same role in CTCL, it is rather not the case here based on our results [58,105,110]. The postulated patomechanism seems to be specific to AD [110].
IL-31, a chemokine ligand expressed by monocytes and dendritic cells, is also correlated with CCL18 and may be associated with the development of CTCL [84]. Both of these cells are important in the pathogenesis of PCL [89,103]. Moreover, exposition of Staphylococcal enterotoxin B, a potent superantigen, to patients with AD rapidly elevates the IL-31 levels secreted by T cells [112]. In cultured patients’ tumor cell samples, IL-2 acted as the previously mentioned superantigen, resulting in the expression of IL-31 in 9 of 11 cases [113]. Both illustrated mechanisms could give a reasonable explanation to the observed elevation of IL-31 levels in patients with PCL. Moreover, it may explain why, in some studies, the concentrations were higher in advanced stages of the disease.

2.5. Role of JAK-STAT Pathways in PCL

As of today, four types of Janus kinases (JAKs) (JAK1, JAK2, JAK3 and TYK2) and seven different signal transducers and activators (STATs) (STAT1, STAT2, STAT3, STAT4, STAT5a and STAT5b, STAT6) have been identified [114,115]. These molecules have important roles in the transmission of the cytokine signal in various human cells in vivo [114]. They are abundantly expressed in the healthy human epidermis [115]. Numerous studies have shown the impact of different JAK/STAT pathways on the pathogenesis of PCLs [90,101,103,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181].
Strong evidence has been collected on genetic abnormalities of JAKs and STATs in cutaneous lymphoma cells, both on the cell lines and PBMCs from patients [103,125,129,137,139,143,144,149,153,157,162,170]. Studies highlight the activating mutations of JAK3, which occurred in 3.3–10.8% of patients with PCL according to different study groups [125,137,139]. Suppressors of cytokine signaling 1 (SOCS-1), a potent CTCL suppressor, regulates the JAK3/STAT5 signaling [119,143,153]. Moreover, mutations of SOCS-1 that abolished its binding to JAK3 reinforced the aggressive course of the lymphoma [143]. Other researchers found that SOCS-1 deletion was one of the most common events in the group they studied and happened especially in the early stages of MF [153]. Interestingly, JAK3 is activated by the IL-2 and is in its pathway, which may be clinically relevant especially in more aggressive types of the disease [139]. JAK3 is expressed in the nuclei of CTCL cells, both from cell lines and PBMCs, and may also play a novel role in malignant clones [167]. Interestingly, tofacitinib (JAK1 and JAK3 selective inhibitor) could not block the kinase activity inside the nucleus, in contrast to the normal blockage of the IL-2/JAK3 pathway [167,177].
Other genetic studies regarding PCL found the pathological variants in JAK1 and JAK2 genes only in individual cases, and concerning common JAK2 alterations detected in other lymphoid malignancies are worth noticing [129,137,162,170,176]. Interestingly, JAK2 may play a role in keeping the Th-1 cytotoxic answer against the tumor present by mediating the IL-12 signaling and thereby phosphorylating STAT4 [135]. STAT4 and STAT6 genes are inversely regulated in CTCL and the loss of expression of the former may play a role in switching to Th-2 answer in the advanced lymphoma stages [131]. In contrast, JAK2 inhibition resulted in a decreased viability of SaEx, suggesting that it may be important for tumorous survival [164]. Concomitantly, with these results acquired from cell lines, the PBMC isolates showed that JAK inhibition potentiates the cytotoxicity of other agents (e.g., histone deacetylase inhibitors (HDACi)) and allows us to achieve a more generalized lethal effect against the malignant clones [165]. Other studies seem to be consistent with these results by showing the results of combinative cytotoxic effect of ruxolitinib (selective JAK1 and JAK2 inhibitor) and reminostat (HDACi) on the CTCL cell lines [168]. Moreover, another study showed the synergistic role of JAK/STAT inhibition on the in vivo SS model, in which romidepsin (HDACi) and mechloretamine were successfully used in the treatment [173]. A blockage of the malignant cell growth mechanisms in CTCL after the administration of increasing doses of ruxolitinib has also been suggested [137]. Further use of JAK inhibitors in the treatment of SS may be due to their ability to stop the constitutive activation of the activated kinases [176]. In advanced stages of a lymphoma, STAT3 and STAT5 are completely dependent on the constitutively activated JAK1 and JAK3 [135]. However, one study showed that apoptosis in CTCL lines may be augmented via the JAK1 pathway and this activity was blocked by ruxolitinib [145].
The further parts of the JAK/STAT pathway, especially STAT3, STAT5, and STAT6 also play the established role of mediators in the PCLs oncogenesis [101,157,173,175]. These proteins act upon the regulation of several gene transcriptions after being phosphorylated by JAKs. One study demonstrated the constitutive activation of STAT5 in CTCL; however, in most cases, after blocking the specific Janus kinase, these molecules should not be able to maintain their function in regulating different gene transcriptions [134,135]. Currently, there are no ongoing and upcoming trials concerning AD and STATs inhibition. Therefore, the STATs role in PCLs is beyond the scope of this review.

2.6. Safety and Danger Concerns of Administering the New Drugs in the Context of PCLs

AD and psoriasis are distinct medical conditions. However, we have to think about the possible interactions between drugs modifying immune responses in AD and pathogenesis of PCL (followed by Dequidt et al.), especially because of the higher risk of lymphoma in severe cases of AD as well as the problem of differentiation between AD and PCL in some cases [182]. Also, in our opinion, it is crucial to first deduce whether the biologic and small molecule treatments in AD may induce lymphoma and second, in case of an overlap or misdiagnosis between those two diseases, a PCL may progress after administration of these agents.
We would like to shortly summarize the above theoretical assumptions. Decreasing the concentration and/or stopping the secretion of IL-4 and IL-13 could lead to the restoration of the Th-1 microenvironment, which may enhance tumorous toxicity. The reduction in the levels of these interleukins after receiving certain treatments discussed earlier is one of the supporting facts for this theory. Therefore, dupilumab, lebrikizumab, and tralokinumab may appear to be clinically efficient in the treatment of the PCL. Agents blocking IL-22, i.e., fezakinumab, could also stop the lymphomagenesis and additionally reduce the ability of the tumorous cells to metastasize in the advanced stages of the lymphoma. We also show the possible involvement of IL-31 in the pathogenesis of PCLs, which is still elusive. Theoretically, blocking the role in the establishment of the Th-2 microenvironment and in the growth of the tumor might be beneficial for the lymphoma patients after administration of nemolizumab, similar to other lymphomas. Lastly, we described the current state of knowledge on the influence of JAKs on PCLs. JAK1 and JAK3 seem to have the pathogenic role by activating the STAT3, STAT5, and STAT6, which contribute significantly to lymphomagenesis. Therefore, blocking them may reduce tumor development. In contrast, JAK2 may also play some role in preventing the growth of lymphomas. Despite the mentioned effects of ruxolitinib on the CTCL cell lines, obstructing this pathway may appear to be harmful for the patients by reducing the Th-1 cytotoxicity directed to the clones. Figure 1 summarizes the most important aspects of the above assumptions.
However, despite the theoretical expectations of stopping the progression of the disease, after administering the immunomodulating agents, PCL may progress or be induced for reasons currently unknown [183]. Dirk Elston, in his letter discussing the role of dupilumab in CTCL, agrees with our findings. He points out that if a cytokine is upregulated, it does not mean we must down regulate it, contrary to the saying “If it’s wet, dry it. If it’s dry, wet it’’ [184]. Concerning the theory we have previously described, we will now discuss what researchers and medical agencies have found when the aforementioned drugs have been used clinically. We were unable to find any reports on the impact of lebrikizumab, tralokinumab, fezakinumab, and nemolizumab on PCLs in PubMed. Also, we performed the search in the “Drug Safety-related Labeling Changes (SrLC)”—the Federal Drug Agency (FDA) database—and could not find any records on these biologic agents [185]. When searching the European Medicines Agency (EMA) website, found information on tralokinumab (Adtralza), which is authorized for use in the European Union in the treatment of moderate to severe AD [186]. We also found an agreement on the investigation plan for pediatric use of lebrikizumab [187].
Despite a multitude of evidence on potential benefits of using JAKi in the treatment of other lymphoid malignancies, the only JAK inhibitor with reported effects on patients with PCLs is ruxolitinib [188]. Baricitinib, upadacitinib, tofacitinib, and ruxolitinib are authorized for use in the European Union [189,190,191,192]. Abrocitinib was recently positively opinioned for marketing authorization [193]. Prior to 22 October 2021, the only JAK inhibitor registered for AD treatment in the EU is baricitinib (Olumiant) [192]. Interestingly, according to the U.S. Food and Drug Administration Database, baricitinib (Olumiant) and tofacitinib (Xeljanz) may increase risk of developing lymphomas, including those of the skin [194,195]. This warning especially raises the problem of potential increased risk of serious adverse events, including cardiovascular and malignant complication in the treatment of chronic inflammatory conditions [195]. Ruxolitinib and upadacitinib do not have such warnings or precautions in their records because they are not used for treatment of arthritis or other inflammatory conditions [195,196,197]. However, the issue of JAK inhibitor safety in the treatment of AD is raised, since they share the same mechanisms of action [198]. Moreover, currently the only JAK inhibitor that has been studied in a big, four-year surveillance study is tofacitinib [198].
Only dupilumab and ruxolitinib will be discussed in the subsequent paragraph, because to the best of our knowledge, only these drugs are reported to have been administered in the treatment of CTCL misdiagnosed as AD or eczema, CTCL itself, and AD followed by CTCL. These cases are recorded in Table 2.
An expert opinion on the safety of dupilumab shows that it is a safe, well-tolerated drug in AD [214]. Noticeably, CTCLs which occur during treatment with this drug may be unrelated, but a long-term follow-up performed with a large cohort of patients is needed to elucidate this subject [214]. Another opinion describes the cases of MF or SS identified in patients treated with dupilumab and concludes that in a limited subset of patients, this drug might appear to be beneficial [91]. However, generally it should be avoided and in some cases contraindicated for CTCL treatment [91,215]. Our research of the Pubmed database has led us to identify a total of 23 cases in which a PCL and use of dupilumab coexisted [199,200,201,202,203,204,205,206,207,208,209,210,211]. A total of 21 people in this group were above 40 years old [199,200,201,202,203,204,205,206,207,209,210,211]. What may be surprising in the context of our theoretical assumptions is that the most common event in the mentioned group was the progression of the lymphoma, which led to the death of two patients, who progressed to SS [199,200,201,202,203,204,205,206,207,209,210,211]. No clinical improvement of the CTCL was observed four times, whereas the disease course improved in three cases [199,203,206,207,211]. In 16 cases, the original diagnosis was AD or eczema while remaining patients were treated for PCL or mogalizumab-associated rash off-label [199,200,201,202,203,204,205,206,207,209,210,211]. Interestingly, dupilumab appeared to be effective for the treatment of lichenoid reaction associated with mogalizumab in a patient with CD8+ MF [211].
Ruxolitinib, which targets JAK1/JAK2 is used in the treatment of psoriatic arthritis, AD, and several lymphoid malignancies, e.g., myelofibrosis and polycythemia vera [215,216]. Moreover, trials on animal models of hemophagocytic lymphohistiocytosis (HLH) prove this JAK inhibitor to be efficient in the treatment of this condition [217,218]. The genetic abnormalities in the JAK/STAT pathway have been the rationale for therapeutic uses that we discussed earlier. Assuming that JAK inhibitors prove to be effective in the treatment of cutaneous lymphomas, clinicians may feel comfortable administering them if the final diagnosis is difficult to make [215]. These facts led the researchers to administer ruxolitinib to nine patients with PCLs (four MF, three non-specified CTCL, one primary cutaneous anaplastic large cell lymphoma (pcALCL), and one subcutaneous panniculitis-like-T-cell lymphoma (SPTCL)) [212,213]. Some was observed in three cases (one MF, one pcALCL and one STPCL), but the disease course remained stable or worsened in the others [212,213]. Interestingly, despite that five of the seven CTCLs showed the signs of JAK/STAT activation, only one patient whose tumor showed 20% overactivation of pSTAT3 responded to the treatment [213].
The conflicting data revealed in this article seems to be in line with our previous considerations on the safety and danger of the use of biologics in the treatment of psoriasis. Blockage of several mechanisms by which the interleukins act and occur in PCLs should be beneficial in the treatment of the disease. However, dupilumab, in most of patients with lymphoma misdiagnosed as AD or eczema, makes it fully apparent. This drug does not seem to be beneficial for CTCL patients in most cases. Accordingly, despite the JAK/STAT activation, most of the lymphomas did not respond to ruxolitinib. With this paper, we would like to raise awareness to the issue of a development or a misdiagnosis of a cutaneous lymphoma in patients with AD. Especially for patients that are 40 years old or above, the chronic and severe course of AD and the sudden worsening of the symptoms should be considered “red flags” to exclude the potential oncologic risk by taking and carefully verifying the biopsy.

3. Materials and Methods

A comprehensive search of the literature using the PubMed (https://pubmed.ncbi.nlm.nih.gov/) electronic database using the search queries “(IL-4 and cutaneous lymphoma) OR (IL-4 and mycosis fungoides)”, “IL-22 and cutaneous lymphoma”, and “IL-31 and cutaneous lymphoma” was performed in the second week of August 2021, from the database inception to the 14th of August 2021. Further research using the queries “(dupilumab and lymphoma)”, “(fezakinumab and cutaneous lymphoma) or (fezakinumab and mycosis fungoides)”, “(lebrikizumab and cutaneous lymphoma) or (lebrikizumab and mycosis fungoides)”, “(tralokinumab and cutaneous lymphoma) or (tralokinumab and mycosis fungoides)”, “(baricitinib and cutaneous lymphoma) or (baricitinib and mycosis fungoides)”, “(ruxolitinib and cutaneous lymphoma) or (ruxolitinib and mycosis fungoides)”, “(upadacitinib and cutaneous lymphoma) or (upadacitinib and mycosis fungoides)”, and “(jak inhibitor and cutaneous lymphoma) or (jak inhibitor and mycosis fungoides)” was performed in the third week of August 2021, from the database inception to the 25th of August 2021 and a “((jak) OR (stat)) AND (cutaneous lymphoma)” search was performed in the second week of September 2021, from the database inception to the 11th of September 2021. After the initial search, titles and abstracts were screened for the inclusion and exclusion criteria. Based on title and abstract analysis, we included articles concerning the role of IL-4, IL-13, IL-22, IL-31, JAK/STAT, and biologic drugs affecting cytokine profiles and JAK inhibitors on PCLs. At this step, we excluded records not related to the topic, non-English manuscripts, personal opinions, and duplicates. The remaining were qualified as eligible for full-text reading. After reading the full manuscripts, some were excluded (not relevant, not original, and not providing information concerning earlier mentioned cytokines, pathways, and new drugs’ impact on PCLs). Finally, additional relevant, eligible records identified through a references search were included, in which information on the effect of PCL microenvironmental influence on the specific lymphoma subtypes were included. Concentration of cytokines in the biopsies and in the blood of the patients, genetic alterations concerning genes linked to the featured subject, the possible effects of interleukins, pathways, and administration of the agents blocking them in the clone cells were analyzed and summarized.

4. Conclusions

IL-4, IL-13, IL-22, and IL-31 are detectable in the lesions and sera of patients suffering from PCL. The JAK-STAT pathway has an established role in the oncogenesis of this tumor. We summarized the effects of these cytokines in the course of cutaneous lymphomas. We have shown that IL-4, IL-13, and activation of certain JAKs and STATs to be crucial in the development of the tumoral microenvironment as well as in the progression of the disease. IL-22 and IL-31, however, are not as important in the pathogenesis of PCL as they are in AD. Based on the publications, we have also described the effect of dupilumab and ruxolitinib in the PCL patients misdiagnosed as AD, in PCL itself or AD itself and with PCL in follow up during treatment (coexistence of two diseases). The progression of the lymphoma was observed in most of cases. Thus, we would like to highlight, that in case of severe AD or eczema, especially in a case of rapid evolution of the symptoms in an older individual, it is necessary to perform a biopsy from the skin lesion. Subsequently, a close pathological examination to exclude the possibility of PCL misdiagnosis/evolution should be performed. However, as the authors, we would like to mention that we should not avoid dupilumab in severe AD, especially concerning the portfolio of other systemic drugs used in AD, such as cyclosporine A, methotrexate, and azathioprine, all with known immunosuppressive potential. We realize the necessity of further research concerning the role of IL-22 and IL-31 in PCL. Furthermore, the new biologic and small molecule drugs should be used carefully in the treatment of AD in order not to worsen the course of the cutaneous lymphomas.

Funding

The APC was funded by funds from the Polish Ministry of Science and Higher Education (02-0066/07/253).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All the data can be found in the PubMed database—https://pubmed.ncbi.nlm.nih.gov/ or under the links cited of cited websites.

Conflicts of Interest

The authors declare no conflict of interest.

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. Gonzalez, B.R.; Zain, J.; Rosen, S.T.; Querfeld, C. Tumor microenvironment in mycosis fungoides and Sézary syndrome. Curr. Opin. Oncol. 2016, 28, 88–96. [Google Scholar] [CrossRef]
  3. Yang, N.; Chen, Z.; Zhang, X.; Shi, Y. Novel Targeted Biological Agents for the Treatment of Atopic Dermatitis. BioDrugs 2021 2021, 35, 401–415. [Google Scholar] [CrossRef] [PubMed]
  4. Traidl, S.; Freimooser, S.; Werfel, T. Janus kinase inhibitors for the therapy of atopic dermatitis. Allergol. Sel. 2021, 5, 293–304. [Google Scholar] [CrossRef] [PubMed]
  5. Szalus, K.; Trzeciak, M.; Nowicki, R.J. Jak-stat inhibitors in atopic dermatitis from pathogenesis to clinical trials results. Microorganisms 2020, 8, 1743. [Google Scholar] [CrossRef]
  6. Hui, R.L.; Lide, W.; Chan, J.; Schottinger, J.; Yoshinaga, M.; Millares, M. Association between exposure to topical tacrolimus or pimecrolimus and cancers. Ann. Pharmacother. 2009, 43, 1956–1963. [Google Scholar] [CrossRef]
  7. Andersen, L.; Nyeland, M.E.; Nyberg, F. Higher self-reported severity of atopic dermatitis in adults is associated with poorer self-reported health-related quality of life in France, Germany, the U.K. and the U.S.A. Br. J. Dermatol. 2020, 182, 1176–1183. [Google Scholar] [CrossRef] [Green Version]
  8. Asher, M.I.; Montefort, S.; Björkstén, B.; Lai, C.K.; Strachan, D.P.; Weiland, S.K.; Williams, H. Worldwide time trends in the prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and eczema in childhood: ISAAC Phases One and Three repeat multicountry cross-sectional surveys. Lancet 2006, 368, 733–743. [Google Scholar] [CrossRef]
  9. Hou, A.; Silverberg, J.I. Secular trends of atopic dermatitis and its comorbidities in United States children between 1997 and 2018. Arch. Dermatol. Res. 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  10. Cheng, J.; Wu, J.J.; Han, G. Epidemiology and Characterization of Atopic Dermatitis in East Asian Populations: A Systematic Review. Dermatol. Ther. 2021, 11, 707–717. [Google Scholar] [CrossRef]
  11. Chello, C.; Carnicelli, G.; Sernicola, A.; Gagliostro, N.; Paolino, G.; Di Fraia, M.; Faina, V.; Muharremi, R.; Grieco, T. Atopic dermatitis in the elderly Caucasian population: Diagnostic clinical criteria and review of the literature. Int. J. Dermatol. 2020, 59, 716–721. [Google Scholar] [CrossRef]
  12. Barbarot, S.; Auziere, S.; Gadkari, A.; Girolomoni, G.; Puig, L.; Simpson, E.L.; Margolis, D.J.; de Bruin-Weller, M.; Eckert, L. Epidemiology of atopic dermatitis in adults: Results from an international survey. Allergy Eur. J. Allergy Clin. Immunol. 2018, 73, 1284–1293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Paller, A.S.; Kabashima, K.; Bieber, T. Therapeutic pipeline for atopic dermatitis: End of the drought? J. Allergy Clin. Immunol. 2017, 140, 633–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Legendre, L.; Barnetche, T.; Mazereeuw-Hautier, J.; Meyer, N.; Murrell, D.; Paul, C. Risk of lymphoma in patients with atopic dermatitis and the role of topical treatment: A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2015, 72, 992–1002. [Google Scholar] [CrossRef] [PubMed]
  15. Mansfield, K.E.; Schmidt, S.A.J.; Darvalics, B.; Mulick, A.; Abuabara, K.; Wong, A.Y.S.; Sørensen, H.T.; Smeeth, L.; Bhaskaran, K.; Dos Santos Silva, I.; et al. Association between Atopic Eczema and Cancer in England and Denmark. JAMA Dermatol. 2020, 156, 1086–1097. [Google Scholar] [CrossRef] [PubMed]
  16. Arellano, F.M.; Arana, A.; Wentworth, C.E.; Fernández-Vidaurre, C.; Schlienger, R.G.; Conde, E. Lymphoma among patients with atopic dermatitis and/or treated with topical immunosuppressants in the United Kingdom. J. Allergy Clin. Immunol. 2009, 123, 1111–1116.e13. [Google Scholar] [CrossRef]
  17. 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]
  18. Miyagaki, T.; Sugaya, M.; Suga, H.; Kamata, M.; Ohmatsu, H.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; Sato, S. IL-22, but not IL-17, Dominant environment in cutaneous T-cell lymphoma. Clin. Cancer Res. 2011, 17, 7529–7538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Jackow, C.M.; Cather, J.C.; Hearne, V.; Asano, A.T.; Musser, J.M.; Duvic, M. Association of erythrodermic cutaneous T-cell lymphoma, superantigen- positive Staphylococcus aureus, and oligoclonal T-cell receptor Vβ gene expansion. Blood 1997, 89, 32–40. [Google Scholar] [CrossRef] [PubMed]
  20. Gong, J.Q.; Lin, L.; Lin, T.; Hao, F.; Zeng, F.Q.; Bi, Z.G.; Yi, D.; Zhao, B. Skin colonization by Staphylococcus aureus in patients with eczema and atopic dermatitis and relevant combined topical therapy: A double-blind multicentre randomized controlled trial. Br. J. Dermatol. 2006, 155, 680–687. [Google Scholar] [CrossRef] [PubMed]
  21. 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] [Green Version]
  22. 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] [Green Version]
  23. Saed, G.; Fivenson, D.P.; Naidu, Y.; Nickoloff, B.J. Mycosis fungoides exhibits a Th1-type cell-mediated cytokine profile whereas sezary syndrome expresses a Th2-type profile. J. Investig. Dermatol. 1994, 103, 29–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Guttman-Yassky, E.; Krueger, J.G. Atopic dermatitis and psoriasis: Two different immune diseases or one spectrum? Curr. Opin. Immunol. 2017, 48, 68–73. [Google Scholar] [CrossRef]
  25. Bieber, T. Atopic dermatitis. N. Engl. J. Med. 2008, 358, 1483–1494. [Google Scholar] [CrossRef]
  26. Trzeciak, M.; Olszewska, B.; Sakowicz-Burkiewicz, M.; Sokołowska-Wojdyło, M.; Jankau, J.; Nowicki, R.J.; Pawełczyk, T. Expression Profiles of Genes Encoding Cornified Envelope Proteins in Atopic Dermatitis and Cutaneous T-Cell Lymphomas. Nutrients 2020, 12, 862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Suga, H.; Sugaya, M.; Miyagaki, T.; Ohmatsu, H.; Kawaguchi, M.; Takahashi, N.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; et al. Skin Barrier Dysfunction and Low Antimicrobial Peptide Expression in Cutaneous T-cell Lymphoma. Clin. Cancer Res. 2014, 20, 4339–4348. [Google Scholar] [CrossRef] [Green Version]
  28. Kopfnagel, V.; Harder, J.; Werfel, T. Expression of antimicrobial peptides in atopic dermatitis and possible immunoregulatory functions. Curr. Opin. Allergy Clin. Immunol. 2013, 13, 531–536. [Google Scholar] [CrossRef] [PubMed]
  29. Saulite, I.; Hoetzenecker, W.; Weidinger, S.; Cozzio, A.; Guenova, E.; Wehkamp, U. Sézary Syndrome and Atopic Dermatitis: Comparison of Immunological Aspects and Targets. BioMed Res. Int. 2016, 2016, 9717530. [Google Scholar] [CrossRef] [Green Version]
  30. A Study Investigating the Efficacy, Safety, and PK Profile of ANB020 Administered to Adult Subjects with Moderate-to-Severe AD (ATLAS). Available online: https://clinicaltrials.gov/ct2/show/NCT03533751?term=ANB020&cond=Atopic+Dermatitis&rank=1 (accessed on 16 October 2021).
  31. A Study to Evaluate the Safety and Efficacy of PF-06826647 in Participants with Moderate to Severe Ulcerative Colitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04209556?term=PF-06826647&cond=ulcerative+colitis&draw=2&rank=1 (accessed on 16 October 2021).
  32. NCT03568071—A Study to Assess Efficacy, Safety, Tolerability and Pharmacokinetics (PK)/Pharmacodynamics (PD) of MOR106 in Subjects with Moderate to Severe Atopic Dermatitis. Available online: https://www.cochranelibrary.com/central/doi/10.1002/central/CN-01660617/full (accessed on 16 October 2021).
  33. A Study to Test Safety, Tolerability, and the Way the Body Absorbs, Distributes, and Gets Rid of a Study Drug Called MOR106, in Healthy Subjects and in Patients with Moderate to Severe Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03689829?term=MOR106&cond=Atopic+Dermatitis&draw=2&rank=3 (accessed on 16 October 2021).
  34. Study of Single and Multiple Doses of ALS-008176 in Healthy Volunteers—Full Text View—ClinicalTrials.gov. Available online: http://www.clinicaltrials.gov/ct2/show/NCT01906164?term=ALS-008176&rank=1 (accessed on 16 October 2021).
  35. Kang, E.G.; Narayana, P.K.; Pouliquen, I.J.; Lopez, M.C.; Ferreira-Cornwell, M.C.; Getsy, J.A. Efficacy and safety of mepolizumab administered subcutaneously for moderate to severe atopic dermatitis. Allergy Eur. J. Allergy Clin. Immunol. 2020, 75, 950–953. [Google Scholar] [CrossRef]
  36. Husein-ElAhmed, H.; Steinhoff, M. Effectiveness of ustekinumab in patients with atopic dermatitis: Analysis of real-world evidence. J. Dermatol. Treat. 2021, in press. [Google Scholar] [CrossRef]
  37. 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. 2020, 84, 597–604. [Google Scholar] [CrossRef] [PubMed]
  38. Gandhi, N.A.; Bennett, B.L.; Graham, N.M.H.; Pirozzi, G.; Stahl, N.; Yancopoulos, G.D. Targeting key proximal drivers of type 2 inflammation in disease. Nat. Rev. Drug Discov. 2016, 15, 35–50. [Google Scholar] [CrossRef] [PubMed]
  39. Gandhi, N.A.; Pirozzi, G.; Graham, N.M.H. Commonality of the IL-4/IL-13 pathway in atopic diseases. Expert Rev. Clin. Immunol. 2017, 13, 425–437. [Google Scholar] [CrossRef] [PubMed]
  40. Griffiths, C.; de Bruin-Weller, M.; Deleuran, M.; Fargnoli, M.C.; Staumont-Sallé, D.; Hong, C.H.; Sánchez-Carazo, J.; Foley, P.; Seo, S.J.; Msihid, J.; et al. Dupilumab in Adults with Moderate-to-Severe Atopic Dermatitis and Prior Use of Systemic Non-Steroidal Immunosuppressants: Analysis of Four Phase 3 Trials. Dermatol. Ther. 2021, 11, 1357–1372. [Google Scholar] [CrossRef] [PubMed]
  41. Ultsch, M.; Bevers, J.; Nakamura, G.; Vandlen, R.; Kelley, R.F.; Wu, L.C.; Eigenbrot, C. Structural basis of signaling blockade by anti-IL-13 antibody Lebrikizumab. J. Mol. Biol. 2013, 425, 1330–1339. [Google Scholar] [CrossRef] [PubMed]
  42. Guttman-Yassky, E.; Blauvelt, A.; Eichenfield, L.F.; Paller, A.S.; Armstrong, A.W.; Drew, J.; Gopalan, R.; Simpson, E.L. Efficacy and Safety of Lebrikizumab, a High-Affinity Interleukin 13 Inhibitor, in Adults With Moderate to Severe Atopic Dermatitis: A Phase 2b Randomized Clinical Trial. JAMA Dermatol. 2020, 156, 411–420. [Google Scholar] [CrossRef] [Green Version]
  43. Simpson, E.L.; Flohr, C.; Eichenfield, L.F.; Simpson, E.L.; Bieber, T.; Sofen, H.; Taïeb, A.; Owen, R.; Putnam, W.; Castro, M.; et al. Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). J. Am. Acad. Dermatol. 2018, 78, 863–871.e11. [Google Scholar] [CrossRef] [Green Version]
  44. A Study of Lebrikizumab (LY3650150) on Vaccine Response in Adults with Atopic Dermatitis (ADopt-VA)—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04626297?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=1 (accessed on 17 August 2021).
  45. Study to Assess the Safety and Efficacy of Lebrikizumab (LY3650150) in Adolescent Participants with Moderate-to-Severe Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04250350?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=2 (accessed on 17 August 2021).
  46. Long-Term Safety and Efficacy Study of Lebrikizumab (LY3650150) in Participants with Moderate-to-Severe Atopic Dermatitis (ADjoin)—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04392154?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=3 (accessed on 17 August 2021).
  47. Evaluation of the Efficacy and Safety of Lebrikizumab (LY3650150) in Moderate to Severe Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04178967?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=4 (accessed on 17 August 2021).
  48. Evaluation of the Efficacy and Safety of Lebrikizumab (LY3650150) in Moderate to Severe Atopic Dermatitis (ADvocate1)—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04146363?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=5 (accessed on 17 August 2021).
  49. Safety and Efficacy of Lebrikizumab (LY3650150) in Combination with Topical Corticosteroid in Moderate-to-Severe Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04250337?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=6 (accessed on 17 August 2021).
  50. A Study of Lebrikizumab (LY3650150) in Combination with Topical Corticosteroids in Japanese Participants with Moderate-to-Severe Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04760314?term=lebrikizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=7 (accessed on 17 August 2021).
  51. Popovic, B.; Breed, J.; Rees, D.G.; Gardener, M.J.; Vinall, L.M.K.; Kemp, B.; Spooner, J.; Keen, J.; Minter, R.; Uddin, F.; et al. Structural Characterisation Reveals Mechanism of IL-13-Neutralising Monoclonal Antibody Tralokinumab as Inhibition of Binding to IL-13Rα1 and IL-13Rα2. J. Mol. Biol. 2017, 429, 208–219. [Google Scholar] [CrossRef] [PubMed]
  52. Wollenberg, A.; Blauvelt, A.; Guttman-Yassky, E.; Worm, M.; Lynde, C.; Lacour, J.P.; Spelman, L.; Katoh, N.; Saeki, H.; Poulin, Y.; et al. Tralokinumab for moderate-to-severe atopic dermatitis: Results from two 52-week, randomized, double-blind, multicentre, placebo-controlled phase III trials (ECZTRA 1 and ECZTRA 2). Br. J. Dermatol. 2021, 184, 437–449. [Google Scholar] [CrossRef]
  53. Silverberg, J.I.; Toth, D.; Bieber, T.; Alexis, A.F.; Elewski, B.E.; Pink, A.E.; Hijnen, D.; Jensen, T.N.; Bang, B.; Olsen, C.K.; et al. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: Results from the double-blind, randomized, multicentre, placebo-controlled phase III ECZTRA 3 trial. Br. J. Dermatol. 2021, 184, 450–463. [Google Scholar] [CrossRef] [PubMed]
  54. 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, 5582581. [Google Scholar] [CrossRef]
  55. Guttman-Yassky, E.; Brunner, P.M.; Neumann, A.U.; Khattri, S.; Pavel, A.B.; Malik, K.; Singer, G.K.; Baum, D.; Gilleaudeau, P.; Sullivan-Whalen, M.; et al. Efficacy and safety of fezakinumab (an IL-22 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by conventional treatments: A randomized, double-blind, phase 2a trial. J. Am. Acad. Dermatol. 2018, 78, 872–881.e6. [Google Scholar] [CrossRef] [Green Version]
  56. Kabashima, K.; Matsumura, T.; Komazaki, H.; Kawashima, M. Trial of Nemolizumab and Topical Agents for Atopic Dermatitis with Pruritus. N. Engl. J. Med. 2020, 383, 141–150. [Google Scholar] [CrossRef] [PubMed]
  57. Ruzicka, T.; Hanifin, J.M.; Furue, M.; Pulka, G.; Mlynarczyk, I.; Wollenberg, A.; Galus, R.; Etoh, T.; Mihara, R.; Yoshida, H.; et al. Anti–Interleukin-31 Receptor A Antibody for Atopic Dermatitis. N. Engl. J. Med. 2017, 376, 826–835. [Google Scholar] [CrossRef] [PubMed]
  58. Saleem, M.D.; Oussedik, E.; D’Amber, V.; Feldman, S.R. Interleukin-31 pathway and its role in atopic dermatitis: A systematic review. J. Dermatol. Treat. 2017, 28, 591–599. [Google Scholar] [CrossRef] [PubMed]
  59. Long-Term Safety and Efficacy of Nemolizumab with Moderate-to-Severe Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03989206?term=nemolizumab&cond=Atopic+Dermatitis&phase=2&draw=2&rank=3 (accessed on 17 August 2021).
  60. Chovatiya, R.; Paller, A.S. JAK inhibitors in the treatment of atopic dermatitis. J. Allergy Clin. Immunol. 2021, 148, 927–940. [Google Scholar] [CrossRef]
  61. Chapman, S.; Kwa, M.; Gold, L.S.; Lim, H.W. Janus kinase inhibitors in dermatology: Part, I. A comprehensive review. J. Am. Acad. Dermatol. 2021, in press. [Google Scholar] [CrossRef]
  62. Simpson, E.L.; Lacour, J.P.; Spelman, L.; Galimberti, R.; Eichenfield, L.F.; Bissonnette, R.; King, B.A.; Thyssen, J.P.; Silverberg, J.I.; Bieber, T.; et al. Baricitinib in patients with moderate-to-severe atopic dermatitis and inadequate response to topical corticosteroids: Results from two randomized monotherapy phase III trials. Br. J. Dermatol. 2020, 183, 242–255. [Google Scholar] [CrossRef] [PubMed]
  63. Simpson, E.L.; Forman, S.; Silverberg, J.I.; Zirwas, M.; Maverakis, E.; Han, G.; Guttman-Yassky, E.; Marnell, D.; Bissonnette, R.; Waibel, J.; et al. Baricitinib in patients with moderate-to-severe atopic dermatitis: Results from a randomized monotherapy phase 3 trial in the United States and Canada (BREEZE-AD5). J. Am. Acad. Dermatol. 2021, 85, 62–70. [Google Scholar] [CrossRef]
  64. Silverberg, J.I.; Simpson, E.L.; Thyssen, J.P.; Gooderham, M.; Chan, G.; Feeney, C.; Biswas, P.; Valdez, H.; Dibonaventura, M.; Nduaka, C.; et al. Efficacy and Safety of Abrocitinib in Patients with Moderate-to-Severe Atopic Dermatitis: A Randomized Clinical Trial. JAMA Dermatol. 2020, 156, 863–873. [Google Scholar] [CrossRef] [PubMed]
  65. Reich, K.; Kabashima, K.; Peris, K.; Silverberg, J.I.; Eichenfield, L.F.; Bieber, T.; Kaszuba, A.; Kolodsick, J.; Yang, F.E.; Gamalo, M.; et al. Efficacy and safety of baricitinib combined with topical corticosteroids for treatment of moderate to severe atopic dermatitis: A randomized clinical trial. JAMA Dermatol. 2020, 156, 1333–1343. [Google Scholar] [CrossRef]
  66. Lilly Lilly and Incyte Announce Top-Line Results from Phase 3 Study (BREEZE-AD4) of Oral Selective JAK Inhibitor Baricitinib in Combination with Topical Corticosteroids in Patients with Moderate to Severe Atopic Dermatitis Not Controlled with Cyclosporine. Available online: https://investor.lilly.com/news-releases/news-release-details/lilly-and-incyte-announce-top-line-results-phase-3-study-breeze (accessed on 20 October 2021).
  67. Bieber, T.; Thyssen, J.P.; Reich, K.; Simpson, E.L.; Katoh, N.; Torrelo, A.; De Bruin-Weller, M.; Thaci, D.; Bissonnette, R.; Gooderham, M.; et al. Pooled safety analysis of baricitinib in adult patients with atopic dermatitis from 8 randomized clinical trials. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 476–485. [Google Scholar] [CrossRef]
  68. Simpson, E.L.; Sinclair, R.; Forman, S.; Wollenberg, A.; Aschoff, R.; Cork, M.; Bieber, T.; Thyssen, J.P.; Yosipovitch, G.; Flohr, C.; et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): A multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet 2020, 396, 255–266. [Google Scholar] [CrossRef]
  69. Bieber, T.; Simpson, E.L.; Silverberg, J.I.; Thaçi, D.; Paul, C.; Pink, A.E.; Kataoka, Y.; Chu, C.-Y.; DiBonaventura, M.; Rojo, R.; et al. Abrocitinib versus Placebo or Dupilumab for Atopic Dermatitis. N. Engl. J. Med. 2021, 384, 1101–1112. [Google Scholar] [CrossRef] [PubMed]
  70. Silverberg, J.I.; de Bruin-Weller, M.; Bieber, T.; Soong, W.; Kabashima, K.; Costanzo, A.; Rosmarin, D.; Lynde, C.; Liu, J.; Gamelli, A.; et al. Upadacitinib plus topical corticosteroids in atopic dermatitis: Week 52 AD Up study results. J. Allergy Clin. Immunol. 2021. [Google Scholar] [CrossRef] [PubMed]
  71. Guttman-Yassky, E.; Teixeira, H.D.; Simpson, E.L.; Papp, K.A.; Pangan, A.L.; Blauvelt, A.; Thaçi, D.; Chu, C.Y.; Hong, C.H.; Katoh, N.; et al. Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (Measure Up 1 and Measure Up 2): Results from two replicate double-blind, randomised controlled phase 3 trials. Lancet 2021, 397, 2151–2168. [Google Scholar] [CrossRef]
  72. Blauvelt, A.; Teixeira, H.D.; Simpson, E.L.; Costanzo, A.; De Bruin-Weller, M.; Barbarot, S.; Prajapati, V.H.; Lio, P.; Hu, X.; Wu, T.; et al. Efficacy and Safety of Upadacitinib vs Dupilumab in Adults with Moderate-to-Severe Atopic Dermatitis: A Randomized Clinical Trial. JAMA Dermatol. 2021, 157, 1047–1055. [Google Scholar] [CrossRef]
  73. Open-Label Extension Study of Upadacitinib in Adult Participants with Moderate to Severe Atopic Dermatitis. Available online: https://clinicaltrials.gov/ct2/show/NCT04195698?term=NCT04195698&draw=2&rank=1 (accessed on 21 October 2021).
  74. A Study to Assess Real-World Use, Safety, and Effectiveness of Oral Upadacitinib in Adult and Adolescent (≥12 Years Old) Participants with Atopic Dermatitis—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT05081557?term=upadacitinib&cond=Atopic+Dermatitis&draw=2&rank=6 (accessed on 21 October 2021).
  75. Papp, K.; Szepietowski, J.C.; Kircik, L.; Toth, D.; Eichenfield, L.F.; Leung, D.Y.M.; Forman, S.B.; Venturanza, M.E.; Sun, K.; Kuligowski, M.E.; et al. Efficacy and safety of ruxolitinib cream for the treatment of atopic dermatitis: Results from 2 phase 3, randomized, double-blind studies. J. Am. Acad. Dermatol. 2021, 85, 863–872. [Google Scholar] [CrossRef]
  76. A Study to Assess the Efficacy and Safety of Ruxolitinib Cream in Children with Atopic Dermatitis (TRuE-AD3)—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04921969 (accessed on 21 October 2021).
  77. Nakagawa, H.; Nemoto, O.; Igarashi, A.; Saeki, H.; Kaino, H.; Nagata, T. Delgocitinib ointment, a topical Janus kinase inhibitor, in adult patients with moderate to severe atopic dermatitis: A phase 3, randomized, double-blind, vehicle-controlled study and an open-label, long-term extension study. J. Am. Acad. Dermatol. 2020, 82, 823–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Efficacy and Safety of Delgocitinib Cream in Adults with Moderate to Severe Chronic Hand Eczema—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04871711?term=delgocitinib&draw=2&rank=9 (accessed on 23 October 2021).
  79. Efficacy and Safety of Delgocitinib Cream in Adults with Moderate to Severe Chronic Hand Eczema (DELTA 2)—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04872101?term=delgocitinib&draw=2&rank=8 (accessed on 23 October 2021).
  80. Asadullah, K.; Döcke, W.D.; Haeuler, A.; Sterry, W.; Volk, H.D. Progression of mycosis fungoides is associated with increasing cutaneous expression of interleukin-10 mRNA. J. Investig. Dermatol. 1996, 107, 833–837. [Google Scholar] [CrossRef] [Green Version]
  81. 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] [Green Version]
  82. 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] [PubMed] [Green Version]
  83. 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] [Green Version]
  84. Miyagaki, T.; Sugaya, M.; Suga, H.; Ohmatsu, H.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; Sato, S. Increased CCL18 expression in patients with cutaneous T-cell lymphoma: Association with disease severity and prognosis. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 24–28. [Google Scholar] [CrossRef]
  85. Harwix, S.; Zachmann, K.; Neumann, C. T-cell clones from early-stage cutaneous T-cell lymphoma show no polarized Th-1 or Th-2 cytokine profile. Arch. Dermatol. Res. 2000, 292, 1–8. [Google Scholar] [CrossRef]
  86. 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]
  87. Bang, K.; Lund, M.; Mogensen, S.C.; Thestrup-Pedersen, K. In vitro culture of skin-homing T lymphocytes from inflammatory skin diseases. Exp. Dermatol. 2005, 14, 391–397. [Google Scholar] [CrossRef]
  88. Yamanaka, K.I.; Clark, R.; Rich, B.; Dowgiert, R.; Hirahara, K.; Hurwitz, D.; Shibata, M.; Mirchandani, N.; Jones, D.A.; Goddard, D.S.; et al. Skin-derived interleukin-7 contributes to the proliferation of lymphocytes in cutaneous T-cell lymphoma. Blood 2006, 107, 2440–2445. [Google Scholar] [CrossRef] [Green Version]
  89. 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]
  90. 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] [Green Version]
  91. Sugaya, M. Is blocking IL-4 receptor alpha beneficial for patients with mycosis fungoides or Sézary syndrome? J. Dermatol. 2021, 48, e225–e226. [Google Scholar] [CrossRef]
  92. Vidulich, K.A.; Talpur, R.; Bassett, R.L.; Duvic, M. Overall survival in erythrodermic cutaneous T-cell lymphoma: An analysis of prognostic factors in a cohort of patients with erythrodermic cutaneous T-cell lymphoma. Int. J. Dermatol. 2009, 48, 243–252. [Google Scholar] [CrossRef] [PubMed]
  93. Talpur, R.; Singh, L.; Daulat, S.; Liu, P.; Seyfer, S.; Trynosky, T.; Wei, W.; Duvic, M. Long term outcomes of 1263 patients with Mycosis fungoides and Sézary syndrome from 1982 to 2009. Clin. Cancer Res. 2012, 18, 5051–5060. [Google Scholar] [CrossRef] [Green Version]
  94. 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]
  95. Di Renzo, M.; Rubegni, P.; De Aloe, G.; Paulesu, L.; Pasqui, A.L.; Andreassi, L.; Auteri, A.; Fimiani, M. Extracorporeal photochemotherapy restores Th1/Th2 imbalance in patients with early stage cutaneous T-cell lymphoma. Immunology 1997, 92, 99–103. [Google Scholar] [CrossRef] [PubMed]
  96. Clark, R.A.; Watanabe, R.; Teague, J.E.; Schlapbach, C.; Tawa, M.C.; Adams, N.; Dorosario, A.A.; Chaney, K.S.; Cutler, C.S.; LeBoeuf, N.R.; et al. Skin effector memory T cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients. Sci. Transl. Med. 2012, 4, 117ra7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Fujita, H. The role of IL-22 and Th22 cells in human skin diseases. J. Dermatol. Sci. 2013, 72, 3–8. [Google Scholar] [CrossRef]
  98. Shimauchi, T.; Sasada, K.; Kito, Y.; Mori, T.; Hata, M.; Fujiyama, T.; Ito, T.; Hirakawa, S.; Tokura, Y. CD8+ Sézary syndrome with interleukin-22 production modulated by bacterial sepsis. Br. J. Dermatol. 2013, 168, 881–883. [Google Scholar] [CrossRef]
  99. 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 Dermatol. Venereol. 2020, 100, 1–6. [Google Scholar] [CrossRef]
  100. Ito, M.; Teshima, K.; Ikeda, S.; Kitadate, A.; Watanabe, A.; Nara, M.; Yamashita, J.; Ohshima, K.; Sawada, K.; Tagawa, H. MicroRNA-150 inhibits tumor invasion and metastasis by targeting the chemokine receptor CCR6, in advanced cutaneous T-cell lymphoma. Blood 2014, 123, 1499–1511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Olszewska, B.; Żawrocki, A.; Lakomy, J.; Karczewska, J.; Gleń, J.; Zabłotna, M.; Malek, M.; Jankau, J.; Lange, M.; Biernat, W.; et al. Mapping signal transducer and activator of transcription (STAT) activity in different stages of mycosis fungoides and Sezary syndrome. Int. J. Dermatol. 2020, 59, 1106–1112. [Google Scholar] [CrossRef] [PubMed]
  102. Izykowska, K.; Zawada, M.; Nowicka, K.; Grabarczyk, P.; Braun, F.C.M.; Delin, M.; Möbs, M.; Beyer, M.; Sterry, W.; Schmidt, C.A.; et al. Identification of multiple complex rearrangements associated with deletions in the 6q23-27 region in sézary syndrome. J. Investig. Dermatol. 2013, 133, 2617–2625. [Google Scholar] [CrossRef] [Green Version]
  103. Fanok, M.H.; Sun, A.; Fogli, L.K.; Narendran, V.; Eckstein, M.; Kannan, K.; Dolgalev, I.; Lazaris, C.; Heguy, A.; Laird, M.E.; et al. Role of Dysregulated Cytokine Signaling and Bacterial Triggers in the Pathogenesis of Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2018, 138, 1116–1125. [Google Scholar] [CrossRef] [Green Version]
  104. Ikeda, S.; Kitadate, A.; Ito, M.; Abe, F.; Nara, M.; Watanabe, A.; Takahashi, N.; Miyagaki, T.; Sugaya, M.; Tagawa, H. Disruption of CCL20-CCR6 interaction inhibits metastasis of advanced cutaneous T-cell lymphoma. Oncotarget 2016, 7, 13563–13574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. 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] [Green Version]
  106. 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] [Green Version]
  107. Tracey, L.; Villuendas, R.; Dotor, A.M.; Spiteri, I.; Ortiz, P.; García, J.F.; Rodríguez Peralto, J.L.; Lawler, M.; Piris, M.A. Mycosis fungoides shows concurrent deregulation of multiple genes involved in the TNF signaling pathway: An expression profile study. Blood 2003, 102, 1042–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. 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] [Green Version]
  109. 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] [Green Version]
  110. 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. Adv. Dermatol. Allergol. 2020, in press. [Google Scholar] [CrossRef]
  111. 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] [Green Version]
  112. Sonkoly, E.; Muller, A.; Lauerma, A.I.; Pivarcsi, A.; Soto, H.; Kemeny, L.; Alenius, H.; Dieu-Nosjean, M.C.; Meller, S.; Rieker, J.; et al. IL-31: A new link between T cells and pruritus in atopic skin inflammation. J. Allergy Clin. Immunol. 2006, 117, 411–417. [Google Scholar] [CrossRef]
  113. Möbs, M.; Gryzik, S.; Haidar, A.; Humme, D.; Beyer, M.; Vandersee, S. Analysis of the IL-31 pathway in mycosis fungoides and sézary syndrome. Arch. Dermatol. Res. 2015, 307, 479–485. [Google Scholar] [CrossRef]
  114. Leonard, W.J. Role of JAK kinases and stats in cytokine signal transduction. Int. J. Hematol. 2001, 73, 271–277. [Google Scholar] [CrossRef] [PubMed]
  115. Nishio, H.; Matsui, K.; Tsuji, H.; Tamura, A.; Suzuki, K. Immunolocalisation of the janus kinases (JAK)-signal transducers and activators of transcription (STAT) pathway in human epidermis. J. Anat. 2001, 198, 581–589. [Google Scholar] [CrossRef] [PubMed]
  116. Mitchell, T.J.; Whittaker, S.J.; John, S. Dysregulated Expression of COOH-Terminally Truncated Stat5 and Loss of IL2-Inducible Stat5-Dependent Gene Expression in Sezary Syndrome. Cancer Res. 2003, 63, 9048–9054. [Google Scholar] [PubMed]
  117. Bladon, J.; Taylor, P. Extracorporeal photopheresis differentially regulates the expression of phosphorylated STAT-1 and STAT-5 in treated monocytes and T cells, respectively. J. Cutan. Med. Surg. 2004, 8, 148–156. [Google Scholar] [CrossRef] [PubMed]
  118. Sommer, V.H.; Clemmensen, O.J.; Nielsen, O.; Wasik, M.; Lovato, P.; Brender, C.; Eriksen, K.W.; Woetmann, A.; Kaestel, C.G.; Nissen, M.H.; et al. In vivo activation of STAT3 in cutaneous T-cell lymphoma. Evidence for an antiapoptotic function of STAT3. Leukemia 2004, 18, 1288–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Brender, C.; Lovato, P.; Sommer, V.H.; Woetmann, A.; Mathiesen, A.M.; Geisler, C.; Wasik, M.; Ødum, N. Constitutive SOCS-3 expression protects T-cell lymphoma against growth inhibition by IFNα. Leukemia 2005, 19, 209–213. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, C.; Li, B.; Gaikwad, A.S.; Haridas, V.; Xu, Z.; Gutterman, J.U.; Duvic, M. Avicin D selectively induces apoptosis and downregulates p-STAT-3, bcl-2, and survivin in cutaneous T-cell lymphoma cells. J. Investig. Dermatol. 2008, 128, 2728–2735. [Google Scholar] [CrossRef] [Green Version]
  121. Fantin, V.R.; Loboda, A.; Paweletz, C.P.; Hendrickson, R.C.; Pierce, J.W.; Roth, J.A.; Li, L.; Gooden, F.; Korenchuk, S.; Hou, X.S.; et al. Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res. 2008, 68, 3785–3794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Van Kester, M.S.; Out-Luiting, J.J.; Von Dem Borne, P.A.; Willemze, R.; Tensen, C.P.; Vermeer, M.H. Cucurbitacin I inhibits Stat3 and induces apoptosis in Sézary cells. J. Investig. Dermatol. 2008, 128, 1691–1695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zhang, C.; Li, B.; Zhang, X.; Hazarika, P.; Aggarwal, B.B.; Duvic, M. Curcumin selectively induces apoptosis in cutaneous T-cell lymphoma cell lines and patients PBMCs: Potential role for STAT-3 and NF-B signaling. J. Investig. Dermatol. 2010, 130, 2110–2119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Verma, N.K.; Davies, A.M.; Long, A.; Kelleher, D.; Volkov, Y. STAT3 knockdown by siRNA induces apoptosis in human cutaneous T-cell lymphoma line Hut78 via downregulation of Bcl-xL. Cell. Mol. Biol. Lett. 2010, 15, 342–355. [Google Scholar] [CrossRef]
  125. Kameda, T.; Shide, K.; Shimoda, H.K.; Hidaka, T.; Kubuki, Y.; Katayose, K.; Taniguchi, Y.; Sekine, M.; Kamiunntenn, A.; Maeda, K.; et al. Absence of gain-of-function JAK1 and JAK3 mutations in adult T cell leukemia/lymphoma. Int. J. Hematol. 2010, 92, 320–325. [Google Scholar] [CrossRef]
  126. Wu, J.; Siddiqui, J.; Nihal, M.; Vonderheid, E.C.; Wood, G.S. Structural alterations of the FAS gene in cutaneous T-cell lymphoma (CTCL). Arch. Biochem. Biophys. 2011, 508, 185–191. [Google Scholar] [CrossRef] [Green Version]
  127. Persson, J.L. MiR-155 meets the JAK/STAT pathway. Cell Cycle 2013, 12, 2170. [Google Scholar] [CrossRef] [Green Version]
  128. Kopp, K.L.; Ralfkiaer, U.; Gjerdrum, L.M.R.; Helvad, R.; Pedersen, I.H.; Litman, T.; Jønson, L.; Hagedorn, P.H.; Krejsgaard, T.; Gniadecki, R.; et al. STAT5-mediated expression of oncogenic miR-155 in cutaneous T-cell lymphoma. Cell Cycle 2013, 12, 1939–1947. [Google Scholar] [CrossRef] [PubMed]
  129. Ehrentraut, S.; Nagel, S.; Scherr, M.E.; Schneider, B.; Quentmeier, H.; Geffers, R.; Kaufmann, M.; Meyer, C.; Prochorec-Sobieszek, M.; Ketterling, R.P.; et al. t(8;9)(p22;p24)/PCM1-JAK2 Activates SOCS2 and SOCS3 via STAT5. PLoS ONE 2013, 8, e53767. [Google Scholar] [CrossRef] [PubMed]
  130. Döbbeling, U.; Waeckerle-Men, Y.; Zabel, F.; Graf, N.; Kündig, T.M.; Johansen, P. The antihistamines clemastine and desloratadine inhibit STAT3 and c-Myc activities and induce apoptosis in cutaneous T-cell lymphoma cell lines. Exp. Dermatol. 2013, 22, 119–124. [Google Scholar] [CrossRef]
  131. 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] [Green Version]
  132. Van Der Fits, L.; Out-Luiting, J.J.; Tensen, C.P.; Zoutman, W.H.; Vermeer, M.H. Exploring the IL-21-STAT3 axis as therapeutic target for Sézary syndrome. J. Investig. Dermatol. 2014, 134, 2639–2647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. 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] [Green Version]
  134. Zhang, Q.; Wang, H.Y.; Wei, F.; Liu, X.; Paterson, J.C.; Roy, D.; Mihova, D.; Woetmann, A.; Ptasznik, A.; Odum, N.; et al. Cutaneous T Cell Lymphoma Expresses Immunosuppressive CD80 (B7-1) Cell Surface Protein in a STAT5-Dependent Manner. J. Immunol. 2014, 192, 2913–2919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. 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]
  136. Zhang, Q.; Nowak, I.; Vonderheid, E.C.; Rook, A.H.; Kadin, M.E.; Nowell, P.C.; Shaw, L.M.; Wasik, M.A. Activation of Jak/STAT proteins involved in signal transduction pathway mediated by receptor for interleukin 2 in malignant T lymphocytes derived from cutaneous anaplastic large T-cell lymphoma and Sezary syndrome. Proc. Natl. Acad. Sci. USA 1996, 93, 9148–9153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Pérez, C.; González-Rincón, J.; Onaindia, A.; Almaráz, C.; García-Díaz, N.; Pisonero, H.; Curiel-Olmo, S.; Gómez, S.; Cereceda, L.; Madureira, R.; et al. Mutated JAK kinases and deregulated STAT activity are potential therapeutic targets in cutaneous T-cell lymphoma. Haematologica 2015, 100, e450–e453. [Google Scholar] [CrossRef] [Green Version]
  138. 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] [Green Version]
  139. 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] [Green Version]
  140. Litvinov, I.V.; Shtreis, A.; Kobayashi, K.; Glassman, S.; Tsang, M.; Woetmann, A.; Sasseville, D.; Ødum, N.; Duvic, M. Investigating potential exogenous tumor initiating and promoting factors for Cutaneous T-Cell Lymphomas (CTCL), a rare skin malignancy. Oncoimmunology 2016, 5, 1175799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Chevret, E.; Merlio, J.P. Sézary Syndrome: Translating Genetic Diversity into Personalized Medicine. J. Investig. Dermatol. 2016, 136, 1319–1324. [Google Scholar] [CrossRef] [Green Version]
  142. Woollard, W.J.; Pullabhatla, V.; Lorenc, A.; Patel, V.M.; Butler, R.M.; Bayega, A.; Begum, N.; Bakr, F.; Dedhia, K.; Fisher, J.; et al. Candidate driver genes involved in genome maintenance and DNA repair in Sézary syndrome. Blood 2016, 127, 3387–3397. [Google Scholar] [CrossRef] [Green Version]
  143. Ehrentraut, S.; Schneider, B.; Nagel, S.; Pommerenke, C.; Quentmeier, H.; Geffers, R.; Feist, M.; Kaufmann, M.; Meyer, C.; Kadin, M.E.; et al. Th17 cytokine differentiation and loss of plasticity after SOCS1 inactivation in a cutaneous T-cell lymphoma. Oncotarget 2016, 7, 34201–34216. [Google Scholar] [CrossRef] [Green Version]
  144. Benoit, B.M.; Jariwala, N.; O’Connor, G.; Oetjen, L.K.; Whelan, T.M.; Werth, A.; Troxel, A.B.; Sicard, H.; Zhu, L.; Miller, C.; et al. CD164 identifies CD4+ T cells highly expressing genes associated with malignancy in Sézary syndrome: The Sézary signature genes, FCRL3, Tox, and miR-214. Arch. Dermatol. Res. 2017, 309, 11–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liszewski, W.; Naym, D.G.; Biskup, E.; Gniadecki, R. Psoralen with ultraviolet A-induced apoptosis of cutaneous lymphoma cell lines is augmented by type I interferons via the JAK1–STAT1 pathway. Photodermatol. Photoimmunol. Photomed. 2017, 33, 164–171. [Google Scholar] [CrossRef] [PubMed]
  146. Ni, X.; Zhang, X.; Hu, C.H.; Langridge, T.; Tarapore, R.S.; Allen, J.E.; Oster, W.; Duvic, M. ONC201 selectively induces apoptosis in cutaneous T-cell lymphoma cells via activating pro-apoptotic integrated stress response and inactivating JAK/STAT and NF-κB pathways. Oncotarget 2017, 8, 61761–61776. [Google Scholar] [CrossRef] [Green Version]
  147. Nielsen, M.; Kaltoft, K.; Nordahl, M.; Röpke, C.; Geisler, C.; Mustelin, T.; Dobson, P.; Svejgaard, A.; Ødum, N. Constitutive activation of a slowly migrating isoform of Stat3 in mycosis fungoides: Tyrphostin AG490 inhibits Stat3 activation and growth of mycosis fungoides tumor cell lines. Proc. Natl. Acad. Sci. USA 1997, 94, 6764–6769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Waldmann, T.A.; Chen, J. Disorders of the JAK/STAT pathway in T cell lymphoma pathogenesis: Implications for immunotherapy. Annu. Rev. Immunol. 2017, 35, 533–550. [Google Scholar] [CrossRef] [PubMed]
  149. Panagopoulos, I.; Gorunova, L.; Spetalen, S.; Bassarova, A.; Beiske, K.; Micci, F.; Heim, S. Fusion of the genes ataxin 2 like, ATXN2L, and Janus kinase 2, JAK2, in cutaneous CD4 positive T-cell lymphoma. Oncotarget 2017, 8, 103775–103784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Torres, A.N.B.; Najidh, S.; Tensen, C.P.; Vermeer, M.H. Molecular advances in cutaneous T-cell lymphoma. Semin. Cutan. Med. Surg. 2018, 37, 81–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Seto, A.G.; Beatty, X.; Lynch, J.M.; Hermreck, M.; Tetzlaff, M.; Duvic, M.; Jackson, A.L. Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. Br. J. Haematol. 2018, 183, 428–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Jacobsen, E.D.; Weinstock, D.M. Challenges and implications of genomics for T-cell lymphomas. Hematology 2018, 2018, 63–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Bastidas Torres, A.N.; Cats, D.; Mei, H.; Szuhai, K.; Willemze, R.; Vermeer, M.H.; Tensen, C.P. Genomic analysis reveals recurrent deletion of JAK-STAT signaling inhibitors HNRNPK and SOCS1 in mycosis fungoides. Genes Chromosom. Cancer 2018, 57, 653–664. [Google Scholar] [CrossRef] [Green Version]
  154. Lindahl, L.M.; Willerslev-Olsen, A.; Gjerdrum, L.M.R.; Nielsen, P.R.; Blümel, E.; Rittig, A.H.; Celis, P.; Herpers, B.; Becker, J.C.; Stausbøl-Grøn, B.; et al. Antibiotics inhibit tumor and disease activity in cutaneous T-cell lymphoma. Blood 2019, 134, 1072–1083. [Google Scholar] [CrossRef]
  155. Yang, H.; Tao, Y.; Zhang, M.; Ma, P.; Li, L.; Diao, Q. Effects of 9-cis-retinoic acid on the proliferation and apoptosis of cutaneous T-cell lymphoma cells. Anticancer Drugs 2019, 30, 56–64. [Google Scholar] [CrossRef]
  156. Moosic, K.B.; Paila, U.; Olson, K.C.; Dziewulska, K.; Wang, T.T.; Xing, J.C.; Ratan, A.; Feith, D.J.; Loughran, T.P.; Olson, T.L. Genomics of LGL leukemia and select other rare leukemia/lymphomas. Best Pract. Res. Clin. Haematol. 2019, 32, 196–206. [Google Scholar] [CrossRef]
  157. Seffens, A.; Herrera, A.; Tegla, C.; Buus, T.B.; Hymes, K.B.; Ødum, N.; Geskin, L.J.; Koralov, S.B. STAT3 dysregulation in mature T and NK cell lymphomas. Cancers 2019, 11, 1711. [Google Scholar] [CrossRef] [Green Version]
  158. Sun, W.H.; Pabon, C.; Alsayed, Y.; Huang, P.P.; Jandeska, S.; Uddin, S.; Platanias, L.C.; Rosen, S.T. Interferon-α resistance in a cutaneous T-cell lymphoma cell line is associated with lack of STAT1 expression. Blood 1998, 91, 570–576. [Google Scholar] [CrossRef] [PubMed]
  159. Trochopoulos, A.G.X.; Zaharieva, M.M.; Marinova, M.H.; Yoncheva, K.; Tibi, P.E.; Berger, M.R.; Konstantinov, S.M. Antineoplastic effect of a novel nanosized curcumin on cutaneous T cell lymphoma. Oncol. Lett. 2020, 20, 304. [Google Scholar] [CrossRef]
  160. Pérez, C.; Mondéjar, R.; García-Díaz, N.; Cereceda, L.; León, A.; Montes, S.; Durán Vian, C.; Pérez Paredes, M.G.; González-Morán, A.; Alegre de Miguel, V.; et al. Advanced-stage mycosis fungoides: Role of the signal transducer and activator of transcription 3, nuclear factor-κB and nuclear factor of activated T cells pathways. Br. J. Dermatol. 2020, 182, 147–155. [Google Scholar] [CrossRef]
  161. Zhu, M.; Yang, L.; Shi, X.; Gong, Z.; Yu, R.; Zhang, D.; Zhang, Y.; Ma, W. TPD7 inhibits the growth of cutaneous T cell lymphoma H9 cell through regulating IL-2R signalling pathway. J. Cell. Mol. Med. 2020, 24, 984–995. [Google Scholar] [CrossRef] [Green Version]
  162. Maurus, K.; Appenzeller, S.; Roth, S.; Brändlein, S.; Kneitz, H.; Goebeler, M.; Rosenwald, A.; Geissinger, E.; Wobser, M. Recurrent Oncogenic JAK and STAT Alterations in Cutaneous CD30-Positive Lymphoproliferative Disorders. J. Investig. Dermatol. 2020, 140, 2023–2031.e1. [Google Scholar] [CrossRef] [PubMed]
  163. García-Colmenero, L.; González, J.; Sandoval, J.; Guillén, Y.; Diaz-Lagares, A.; Andrades, E.; Iglesias, A.; Nonell, L.; Pujol, R.M.; Bigas, A.; et al. Epigenetic Silencing of Tumor Suppressor miR-124 Directly Supports STAT3 Activation in Cutaneous T-Cell Lymphoma. Cells 2020, 9, 2692. [Google Scholar] [CrossRef]
  164. Brouwer, I.J.; Out-Luiting, J.J.; Vermeer, M.H.; Tensen, C.P. Cucurbitacin E and I target the JAK/STAT pathway and induce apoptosis in Sézary cells. Biochem. Biophys. Rep. 2020, 24, 100832. [Google Scholar] [CrossRef] [PubMed]
  165. Yumeen, S.; Mirza, F.N.; Lewis, J.M.; King, A.L.O.; Kim, S.R.; Carlson, K.R.; Umlauf, S.R.; Surovtseva, Y.V.; Foss, F.M.; Girardi, M. JAK inhibition synergistically potentiates BCL2, BET, HDAC, and proteasome inhibition in advanced CTCL. Blood Adv. 2020, 4, 2213–2226. [Google Scholar] [CrossRef]
  166. Le, M.; Ghazawi, F.M.; Netchiporouk, E.; Litvinov, I.V. The Novel Role of Antibiotic Treatment in the Management of Cutaneous T-Cell Lymphoma (CTCL) Patients. J. Cutan. Med. Surg. 2020, 24, 410–411. [Google Scholar] [CrossRef]
  167. 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]
  168. Karagianni, F.; Piperi, C.; Mpakou, V.; Spathis, A.; Foukas, P.G.; Dalamaga, M.; Pappa, V.; Papadavid, E. Ruxolitinib with resminostat exert synergistic antitumor effects in Cutaneous T-cell Lymphoma. PLoS ONE 2021, 16, e0248298. [Google Scholar] [CrossRef] [PubMed]
  169. Woetmann, A.; Nielsen, M.; Christensen, S.T.; Brockdorff, J.; Kaltoft, K.; Engel, A.M.; Skov, S.; Brender, C.; Geisler, C.; Svejgaard, A.; et al. Inhibition of protein phosphatase 2A induces serine/threonine phosphorylation, subcellular redistribution, and functional inhibition of STAT3. Proc. Natl. Acad. Sci. USA 1999, 96, 10620–10625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Bastidas Torres, A.N.; Cats, D.; Out-Luiting, J.J.; Fanoni, D.; Mei, H.; Venegoni, L.; Willemze, R.; Vermeer, M.H.; Berti, E.; Tensen, C.P. Deregulation of JAK2 signaling underlies primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma. Haematologica 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  171. Willerslev-Olsen, A.; Gjerdrum, L.M.R.; Lindahl, L.M.; Buus, T.B.; Pallesen, E.M.H.; Gluud, M.; Bzorek, M.; Nielsen, B.S.; Kamstrup, M.R.; Rittig, A.H.; et al. Staphylococcus aureus Induces Signal Transducer and Activator of Transcription 5–Dependent miR-155 Expression in Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2021, 141, 2449–2458. [Google Scholar] [CrossRef]
  172. Pavlidis, A.; Piperi, C.; Papadavid, E. Novel therapeutic approaches for cutaneous T cell lymphomas. Expert Rev. Clin. Immunol. 2021, 17, 629–641. [Google Scholar] [CrossRef] [PubMed]
  173. Cortes, J.R.; Patrone, C.C.; Quinn, S.A.; Gu, Y.; Sanchez-Martin, M.; Mackey, A.; Cooke, A.J.; Shih, B.B.; Laurent, A.P.; Trager, M.H.; et al. Jak-STAT Inhibition Mediates Romidepsin and Mechlorethamine Synergism in Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2021, 141, 2908–2920.e7. [Google Scholar] [CrossRef] [PubMed]
  174. Nihal, M.; Wu, J.; Stonesifer, C.J.; Daniels, J.; Choi, J.; Geskin, L.; Rook, A.H.; Wood, G.S. Epigenetic Regulation of Apoptosis in Cutaneous T-Cell Lymphoma: Implications for Therapy with Methotrexate, Jak Inhibitors, and Resveratrol. J. Investig. Dermatol. 2021, in press. [Google Scholar] [CrossRef]
  175. García-Díaz, N.; Piris, M.Á.; Ortiz-Romero, P.L.; Vaqué, J.P. Mycosis fungoides and sézary syndrome: An integrative review of the pathophysiology, molecular drivers, and targeted therapy. Cancers 2021, 13, 1391. [Google Scholar] [CrossRef]
  176. McKenzie, R.C.T.; Jones, C.L.; Tosi, I.; Caesar, J.A.; Whittaker, S.J.; Mitchell, T.J. Constitutive activation of STAT3 in Sézary syndrome is independent of SHP-1. Leukemia 2012, 26, 323–331. [Google Scholar] [CrossRef]
  177. Willerslev-Olsen, A.; Krejsgaard, T.; Lindahl, L.M.; Litvinov, I.V.; Fredholm, S.; Petersen, D.L.; Nastasi, C.; Gniadecki, R.; Mongan, N.P.; Sasseville, D.; et al. Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma. Blood 2016, 127, 1287–1296. [Google Scholar] [CrossRef] [PubMed]
  178. Shreberk-Hassidim, R.; Ramot, Y.; Zlotogorski, A. Janus kinase inhibitors in dermatology: A systematic review. J. Am. Acad. Dermatol. 2017, 76, 745–753.e19. [Google Scholar] [CrossRef]
  179. 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] [Green Version]
  180. Eriksen, K.W.; Kaltoft, K.; Mikkelsen, G.; Nielsen, M.; Zhang, Q.; Geisler, C.; Nissen, M.H.; Röpke, C.; Wasik, M.A.; Ødum, N. Constitutive STAT3-activation in Sezary syndrome: Tyrphostin AG490 inhibits STAT3-activation, interleukin-2 receptor expression and growth of leukemic Sezary cells. Leukemia 2001, 15, 787–793. [Google Scholar] [CrossRef] [Green Version]
  181. 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]
  182. Dequidt, L.; Franck, N.; Sanchez-Pena, P.; Dalle, S.; Adamski, H.; Boulinguez, S.; Ingen-Housz-Oro, S.; Ram-Wolff, C.; Boccara, O.; Bonnet, N.; et al. Cutaneous lymphomas appearing during treatment with biologics: 44 cases from the French Study Group on Cutaneous Lymphomas and French Pharmacovigilance Database. Br. J. Dermatol. 2019, 181, 616–618. [Google Scholar] [CrossRef]
  183. Kołkowski, K.; Sokołowska-Wojdyło, M. Safety and danger of biologic treatments in psoriasis in context of cutaneous T-cell lymphoma (CTCL). Adv. Dermatol. Allergol. 2021, in press. [Google Scholar] [CrossRef]
  184. Elston, D.M. Dupilumab and cutaneous T-cell lymphoma. J. Am. Acad. Dermatol. 2020, 83, 33–34. [Google Scholar] [CrossRef] [PubMed]
  185. Drug Safety-Related Labeling Changes (SrLC). Available online: https://www.accessdata.fda.gov/scripts/cder/safetylabelingchanges/index.cfm?event=searchResult.page (accessed on 16 October 2021).
  186. European Medicines Agency. Adtralza. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/adtralza#authorisation-details-section (accessed on 21 October 2021).
  187. European Medicines Agency. EMEA-002536-PIP01-18. Available online: https://www.ema.europa.eu/en/medicines/human/paediatric-investigation-plans/emea-002536-pip01-18 (accessed on 21 October 2021).
  188. Klein, K.; Stoiber, D.; Sexl, V.; Witalisz-Siepracka, A. Untwining anti-tumor and immunosuppressive effects of JAK inhibitors—A strategy for hematological malignancies? Cancers 2021, 13, 2611. [Google Scholar] [CrossRef] [PubMed]
  189. European Medicines Agency. Octagam. Available online: http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/referrals/Octagam/human_referral_000265.jsp&mid=WC0b01ac0580024e99 (accessed on 20 October 2021).
  190. European Medicines Agency. Xeljanz. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/xeljanz#patient-safety-section (accessed on 20 October 2021).
  191. European Medicines Agency. Beromun. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/beromun (accessed on 20 October 2021).
  192. European Medicines Agency. Olumiant. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/olumiant#product-information-section (accessed on 20 October 2021).
  193. European Medicines Agency. EMA Tagrisso: Pending EC Decision. Available online: https://www.ema.europa.eu/en/medicines/human/summaries-opinion/tagrisso (accessed on 20 October 2021).
  194. Drug Safety-Related Labeling Changes (SrLC). Available online: https://www.accessdata.fda.gov/scripts/cder/safetylabelingchanges/index.cfm?event=searchdetail.page&DrugNameID=2084 (accessed on 16 October 2021).
  195. Food and Drug Administration. FDA Requires Warnings about Increased Risk of Serious Heart-Related Events, Cancer, Blood Clots, and Death for JAK Inhibitors That Treat Certain Chronic Inflammatory Conditions. Available online: https://www.fda.gov/drugs/drug-safety-and-availability/fda-requires-warnings-about-increased-risk-serious-heart-related-events-cancer-blood-clots-and-death (accessed on 22 October 2021).
  196. Drug Safety-Related Labeling Changes (SrLC). Available online: https://www.accessdata.fda.gov/scripts/cder/safetylabelingchanges/index.cfm?event=searchdetail.page&DrugNameID=2285 (accessed on 16 October 2021).
  197. Drug Safety-Related Labeling Changes (SrLC). Available online: https://www.accessdata.fda.gov/scripts/cder/safetylabelingchanges/index.cfm?event=searchdetail.page&DrugNameID=87 (accessed on 16 October 2021).
  198. Murphrey, M.; Waldman, R.A.; Druso, T.; Grant-Kels, J.M. Special Editorial: When Prescribing Janus Kinase Inhibitors for Dermatologic Conditions, Be Mindful of the FDA’s 9/1/2021 Data Safety Communication. J. Am. Acad. Dermatol. 2021, in press. [Google Scholar] [CrossRef]
  199. Chiba, T.; Nagai, T.; Osada, S.I.; Manabe, M. Diagnosis of mycosis fungoides following administration of dupilumab for misdiagnosed atopic dermatitis. Acta Derm.-Venereol. 2019, 99, 818–819. [Google Scholar] [CrossRef] [Green Version]
  200. Tran, J.; Morris, L.; Vu, A.; Duvic, M. Development of Sézary syndrome following the administration of dupilumab. Dermatol. Online J. 2020, 26, 17. [Google Scholar] [CrossRef]
  201. Miyashiro, D.; Vivarelli, A.G.; Gonçalves, F.; Cury-Martins, J.; Sanches, J.A. Progression of mycosis fungoides after treatment with dupilumab: A case report. Dermatol. Ther. 2020, 33, e13880. [Google Scholar] [CrossRef]
  202. Espinosa, M.L.; Nguyen, M.T.; Aguirre, A.S.; Martinez-Escala, M.E.; Kim, J.; Walker, C.J.; Pontes, D.S.; Silverberg, J.I.; Choi, J.; Pro, B.; et al. Progression of cutaneous T-cell lymphoma after dupilumab: Case review of 7 patients. J. Am. Acad. Dermatol. 2020, 83, 197–199. [Google Scholar] [CrossRef] [PubMed]
  203. Claire Hollins, L.; Wirth, P.; Fulchiero, G.J.; Foulke, G.T. Long-standing dermatitis treated with dupilumab with subsequent progression to cutaneous T-cell lymphoma. Cutis 2020, 106, E8–E11. [Google Scholar] [CrossRef]
  204. Mollanazar, N.K.; Savage, K.T.; Pousti, B.T.; Jariwala, N.; Del Guzzo, C.; Haun, P.; Vittorio, C.C.; Rook, A.H.; Kim, E.J. Cutaneous T-cell lymphoma and concomitant atopic dermatitis responding to dupilumab. Cutis 2020, 106, 131–132. [Google Scholar] [CrossRef]
  205. Lazaridou, I.; Ram-Wolff, C.; Bouaziz, J.D.; Begon, E.; Battistella, M.; Rivet, J.; Jachiet, M.; Bagot, M.; De Masson, A. Dupilumab treatment in two patients with cutaneous T-cell lymphomas. Acta Derm.-Venereol. 2020, 100, 1–2. [Google Scholar] [CrossRef] [PubMed]
  206. Steck, O.; Bertschi, N.L.; Luther, F.; van den Berg, J.; Winkel, D.J.; Holbro, A.; Schlapbach, C. Rapid and sustained control of itch and reduction in Th2 bias by dupilumab in a patient with Sézary syndrome. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 1331–1337. [Google Scholar] [CrossRef] [PubMed]
  207. Umemoto, N.; Demitsu, T.; Otaki, K.; Matsumoto, T.; Takazawa, M.; Yamada, A.; Kimura, S.-i.; Kakurai, M. Dupilumab therapy in Sézary syndrome misdiagnosed as atopic dermatitis: A case report. J. Dermatol. 2020, 47, e356–e357. [Google Scholar] [CrossRef] [PubMed]
  208. Ayasse, M.; Kamaria, N.; Glass, F.; Silverberg, J.I. Mycosis Fungoides Unmasked by Dupilumab Treatment in a Patient With a History of Atopic Dermatitis. Dermatitis 2020, 32, e88–e89. [Google Scholar] [CrossRef] [PubMed]
  209. Russomanno, K.; Carver DeKlotz, C.M. Acceleration of cutaneous T-cell lymphoma following dupilumab administration. JAAD Case Rep. 2021, 8, 83–85. [Google Scholar] [CrossRef]
  210. Newsom, M.; Hrin, M.L.; Hamid, R.N.; Strowd, L.C.; Ahn, C.; Jorizzo, J.L.; Feldman, S.R. Two cases of mycosis fungoides diagnosed after treatment non-response to dupilumab. Dermatol. Online J. 2021, 27, 18. [Google Scholar] [CrossRef]
  211. Trum, N.A.; Zain, J.; Abad, C.; Rosen, S.T.; Querfeld, C. Dupilumab as a therapy option for treatment refractory mogamulizumab-associated rash. JAAD Case Rep. 2021, 14, 37–42. [Google Scholar] [CrossRef]
  212. Lévy, R.; Fusaro, M.; Guerin, F.; Chetouani, A.; Moshous, D.; Fischer, A.; de Saint Basile, G.; Sepulveda, F.E.; Neven, B. Efficacy of ruxolitinib in subcutaneous panniculitis-like T-cell lymphoma and hemophagocytic lymphohistiocytosis. Blood Adv. 2020, 4, 1383–1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Moskowitz, A.J.; Ghione, P.; Jacobsen, E.D.; Ruan, J.; Schatz, J.H.; Noor, S.J.; Myskowski, P.L.; Vardhana, S.A.; Ganesan, N.; Hancock, H.; et al. Phase II biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas. Blood 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  214. Francuzik, W.; Alexiou, A.; Worm, M. Safety of dupilumab in patients with atopic dermatitis: Expert opinion. Expert Opin. Drug Saf. 2021, 9, 997–1004. [Google Scholar] [CrossRef]
  215. Sugaya, M. Clinical Guidelines and New Molecular Targets for Cutaneous Lymphomas. Int. J. Mol. Sci. 2021, 22, 11079. [Google Scholar] [CrossRef] [PubMed]
  216. Roskoski, R. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol. Res. 2016, 111, 784–803. [Google Scholar] [CrossRef] [PubMed]
  217. Das, R.; Guan, P.; Sprague, L.; Verbist, K.; Tedrick, P.; An, Q.A.; Cheng, C.; Kurachi, M.; Levine, R.; Wherry, E.J.; et al. Janus kinase inhibition lessens inflammation and ameliorates disease in murine models of hemophagocytic lymphohistiocytosis. Blood 2016, 127, 1666–1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Maschalidi, S.; Sepulveda, F.E.; Garrigue, A.; Fischer, A.; De Saint Basile, G. Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood 2016, 128, 60–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The influence of agents targeting interleukins (IL) 4, 13, 22, and 31 and JAK/STAT pathways on the primary cutaneous lymphomas (PCLs) cells and tumorous microenvironment. The up and down arrows stand for increase/decrease of the interleukins concentration, cell count or receptor’s upregulation. IL-12 promotes phosphorylation of STAT4, thereby stimulating the cytotoxic mediated CD8(+) answer. Concomitantly, IL-4, IL-13, and IL-31 contribute to forming the Th-2 cytokine profile, which results in decreased cytotoxic immunosurveillance and lymphomagenesis. IL-4, IL-13, and IL-22 activate different Janus kinases, which promote the STAT3, STAT5, and STAT6 activation contributing to the transcription of pro-tumorous factors. In the advanced stages of the disease, this phenomenon may be seen more prominently. By blocking several pathways or cytokines, biologic drugs and small molecule inhibitors may affect both the malignant microenvironment and pathways in the PCLs cells.
Figure 1. The influence of agents targeting interleukins (IL) 4, 13, 22, and 31 and JAK/STAT pathways on the primary cutaneous lymphomas (PCLs) cells and tumorous microenvironment. The up and down arrows stand for increase/decrease of the interleukins concentration, cell count or receptor’s upregulation. IL-12 promotes phosphorylation of STAT4, thereby stimulating the cytotoxic mediated CD8(+) answer. Concomitantly, IL-4, IL-13, and IL-31 contribute to forming the Th-2 cytokine profile, which results in decreased cytotoxic immunosurveillance and lymphomagenesis. IL-4, IL-13, and IL-22 activate different Janus kinases, which promote the STAT3, STAT5, and STAT6 activation contributing to the transcription of pro-tumorous factors. In the advanced stages of the disease, this phenomenon may be seen more prominently. By blocking several pathways or cytokines, biologic drugs and small molecule inhibitors may affect both the malignant microenvironment and pathways in the PCLs cells.
Ijms 22 13388 g001
Table 1. Clinical and immunological similarities between atopic dermatitis (AD) and cutaneous T-cell lymphoma (CTCL).
Table 1. Clinical and immunological similarities between atopic dermatitis (AD) and cutaneous T-cell lymphoma (CTCL).
SimilaritiesAtopic DermatitisCutaneous T-Cell Lymphoma
EosinophiliaOften presentMay be present in the advanced stage
Immunoglobulin E (IgE)Often elevatedMay be elevated in the advanced stage
Lactate dehydrogenase (LDH)May be elevatedSeverity marker of MF/SS
Soluble interleukin receptor 2 (sIL-2R)May be elevatedSeverity marker of MF/SS
Th-2 microenvironment activationAlways presentPresent in the advanced stage
Levels of filaggrinSignificantly loweredMay be significantly lowered
Transepidermal water loss (TEWL)Significantly loweredMay be significantly lowered
Levels of antimicrobial peptides (AMPs)Significantly loweredSignificantly lowered
Colonization of S. aureus80% of patients50–60% of patients
Table 2. Cutaneous T-cell lymphoma cases treated with dupilumab or ruxolitinib. We have updated the table continuing the results by doctor Sugaya [91].
Table 2. Cutaneous T-cell lymphoma cases treated with dupilumab or ruxolitinib. We have updated the table continuing the results by doctor Sugaya [91].
DrugAge (Years)SexPre-DiagnosisFinal DiagnosisResponse to TreatmentDeathReference
Dupilumab58MADMFProgression of MFNo[199]
Dupilumab64MADSSProgression of SSNo[200]
Dupilumab51FADMFProgression of MFNo[201]
Dupilumab64MADCTCL-NOSProgression of erythrodermaNo[202]
Dupilumab72MADMFProgression of MFNo[202]
Dupilumab59FADMF and ADProgression of MFNo[202]
Dupilumab40FADMFProgression of MFNo[202]
Dupilumab67MMFSSProgression of SSYes[202]
Dupilumab58MMFSSProgression of SSYes[202]
Dupilumab77FMFSSProgression of SSNo[202]
Dupilumab61MEczemaMFProgression of MFNo[203]
Dupilumab52MEczemaMFNo clinical improvementNo[203]
Dupilumab60FEczemaMFNo clinical improvementNo[203]
Dupilumab68MSS and ADSS and ADImprovement in SS and ADNo[204]
Dupilumab37FEczemaSSProgression of SSNo[205]
Dupilumab55MMF and ADMF and ADImprovement of MF and ADNo[205]
Dupilumab74FSSSSImprovement of SSNo[206]
Dupilumab48FADSS and ADNo clinical improvementNo[207]
Dupilumab40FADMFProgression of MFNo[208]
Dupilumab43MADMF and ADProgression of MFNo[209]
Dupilumab48FADMFProgression of MFNo[210]
Dupilumab55MADMFProgression of MFNo[210]
Dupilumab26MMFMFNo clinical improvementNo[211]
Ruxolitinib13MHLHHLH and SPTCLImprovement of SPTCL and HLHNo[212]
RuxolitinibNSNSMFMFProgression of MFNo[213]
RuxolitinibNSNSCTCLCTCLNo clinical improvement/Stable diseaseNo[213]
RuxolitinibNSNSCTCLCTCLProgression of CTCLNo[213]
RuxolitinibNSNSCTCLCTCLProgression of CTCLNo[213]
RuxolitinibNSNSMFMFProgression of MFNo[213]
RuxolitinibNSNSMFMFNo clinical improvement/Stable diseaseNo[213]
RuxolitinibNSNSMFMFImprovement of MF/Partial remissionNo[213]
RuxolitinibNSNSpcALCLpcALCLImprovement of MF/Complete responseNo[213]
Abbreviations: NS: not specified; M: male; F: female; MF: mycosis fungoides; AD: atopic dermatitis; CTCL: cutaneous t-cell lymphoma; pcALCL: primary cutaneous anaplastic large-cell lymphoma; SS: Sézary Syndrome; HLH: hemophagocytic lymphohistiocytosis; SPTCL: subcutaneous panniculitis-like T-cell lymphoma; CTCL-NOS: CTCL-not otherwise specified.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kołkowski, K.; Trzeciak, M.; Sokołowska-Wojdyło, M. Safety and Danger Considerations of Novel Treatments for Atopic Dermatitis in Context of Primary Cutaneous Lymphomas. Int. J. Mol. Sci. 2021, 22, 13388. https://doi.org/10.3390/ijms222413388

AMA Style

Kołkowski K, Trzeciak M, Sokołowska-Wojdyło M. Safety and Danger Considerations of Novel Treatments for Atopic Dermatitis in Context of Primary Cutaneous Lymphomas. International Journal of Molecular Sciences. 2021; 22(24):13388. https://doi.org/10.3390/ijms222413388

Chicago/Turabian Style

Kołkowski, Karol, Magdalena Trzeciak, and Małgorzata Sokołowska-Wojdyło. 2021. "Safety and Danger Considerations of Novel Treatments for Atopic Dermatitis in Context of Primary Cutaneous Lymphomas" International Journal of Molecular Sciences 22, no. 24: 13388. https://doi.org/10.3390/ijms222413388

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