Next Article in Journal
Prostate-Specific Membrane Antigen-Positron Emission Tomography-Guided Radiomics and Machine Learning in Prostate Carcinoma
Previous Article in Journal
Cytotoxic Autophagy: A Novel Treatment Paradigm against Breast Cancer Using Oleanolic Acid and Ursolic Acid
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Tumor Microenvironment as a Therapeutic Target in Cutaneous T Cell Lymphoma

by
Louis Boafo Kwantwi
1,2,3,
Steven T. Rosen
2,4 and
Christiane Querfeld
1,2,4,5,*
1
Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, USA
2
Beckman Research Institute, Duarte, CA 91010, USA
3
Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
4
Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA 91010, USA
5
Division of Dermatology, City of Hope Medical Center, Duarte, CA 91010, USA
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(19), 3368; https://doi.org/10.3390/cancers16193368
Submission received: 1 September 2024 / Revised: 27 September 2024 / Accepted: 28 September 2024 / Published: 1 October 2024
(This article belongs to the Special Issue Cutaneous T Cell Lymphomas: From Pathology to Treatment)

Abstract

:

Simple Summary

Cutaneous T cell lymphomas (CTCLs) are a group of rare lymphoproliferative malignancies manifesting in the skin. Cutaneous T cell lymphomas are an incurable, disfiguring, and life-threatening disease. Emerging studies have implicated the surrounding cells of malignant T cells (tumor microenvironment) in the disease evolution. This has revealed that targeting the tumor microenvironment has therapeutic potential in cutaneous T cell lymphomas. This review provides a detailed insight into the contribution of the tumor microenvironment in cutaneous T cell lymphomas and the targeting strategies.

Abstract

Cutaneous T cell lymphomas (CTCLs) are a heterogeneous group of non-Hodgkin lymphomas, with mycosis fungoides and Sézary syndrome being the two common subtypes. Despite the substantial improvement in early-stage diagnosis and treatments, some patients still progress to the advanced stage with an elusive underpinning mechanism. While this unsubstantiated disease mechanism coupled with diverse clinical outcomes poses challenges in disease management, emerging evidence has implicated the tumor microenvironment in the disease process, thus revealing a promising therapeutic potential of targeting the tumor microenvironment. Notably, malignant T cells can shape their microenvironment to dampen antitumor immunity, leading to Th2-dominated responses that promote tumor progression. This is largely orchestrated by alterations in cytokines expression patterns, genetic dysregulations, inhibitory effects of immune checkpoint molecules, and immunosuppressive cells. Herein, the recent insights into the determining factors in the CTCL tumor microenvironment that support their progression have been highlighted. Also, recent advances in strategies to target the CTCL tumor micromovement with the rationale of improving treatment efficacy have been discussed.

1. Introduction

Cutaneous T cell lymphomas are a rare form of non-Hodgkin lymphomas characterized by the accumulation of malignant CD4+ lymphocytes homing into the skin [1]. Even though the etiology is still an enigma, the highest incidence rates are found in African American and aged populations with a four-fold increase in individuals over 70 years [2,3]. Although it is a heterogenous disease, mycosis fungoides (MF) and Sézary syndrome (SS) account for 60% of all cases, making them the most studied and common subtypes [4]. MF is defined by patches, plaques, tumors, and/or erythroderma, while SS is a more aggressive and leukemic form of CTCLs characterized by erythroderma and the presence of clonally similar neoplastic T cells with cerebriform nuclei (Sézary cells) in the peripheral blood, skin, and/or lymph nodes [5,6]. The heterogeneous presentation not only makes a definitive diagnosis of CTCLs often difficult but also the selection of appropriate therapeutic options [7]. Although efforts made to understand the pathophysiology of CTCLs have led to the development of new treatment modalities for the early stage [8] and advanced stage of the disease [9,10,11], most patients develop progressive disease due to treatment failure, which makes our knowledge of the exact molecular mechanisms underpinning the disease incomplete. Given the reduced survival rates together with the increasing aggressiveness of CTCLs, studies to map the underlying mechanisms have pinpointed the critical role of the CTCL tumor microenvironment (TME) in the disease processes. Notably, the interaction between malignant T cells and their niche via cytokines dysregulations, genetic alterations, and immune cells infiltrating the microenvironment have become crucial determinants in tumor initiation, metastasis, therapeutic resistance, and other hallmarks of CTCLs [1,12,13]. Therefore, this review aims to elucidate the molecular interaction between CTCLs and their microenvironment and evaluate how such an interaction affects the fate of malignant T cells. In addition, insights into the recent advances in strategies to target the TME are highlighted.

2. Role of CTCL Tumor Microenvironment in CTCL Progression

The CTCL TME is composed of malignant T cells, endothelial cells, fibroblast, keratinocytes, and immune cells, including macrophages, monocytes, B cells, neutrophils, mast cells, eosinophils, natural killer cells, dendritic cells, T cells, myeloid-derived suppressor cells, and regulatory T cells [1,14,15]. The cellular communication mediated by tumor-derived factors alters the physiological role of antitumor immune cells, which shields CTCL cells from therapeutic agents, hence promoting their progression. Notably, the hostile nature of the TME, which is partly attributed to hypoxia [16], causes endothelial cells in the CTCL TME to proliferate and form new vessels, thereby promoting tumor progression [17,18]. Moreover, malignant T cells also activate stem cells and epithelial-to-mesenchymal transition to support their continuous renewal and differentiation [12]. Hence, the microenvironmental niche of CTCLs contributes to all facets of the disease processes, including immunosuppression [19,20,21], therapeutic resistance [22,23], apoptosis resistance [21,24,25,26,27], invasion and migration [28,29,30,31], angiogenesis [17,18,32], and tumor proliferation [33,34,35,36].

2.1. The Immune Tumor Microenvironment and CTCL Progression

2.1.1. Macrophages

Macrophages are one of the major leukocytes infiltrating the tumor microenvironment. Depending on the prevailing conditions in the TME, macrophages can be polarized into M1 and M2 phenotypes. Functionally, tumor-associated macrophages (TAMs) designated as M1 exhibit pro-inflammatory and antitumor functions, while M2 macrophages are anti-inflammatory with tumor-promoting functions [37]. Indeed, it has been found that while the early stage of MF is characterized by the high infiltration of M1 macrophages, M2 macrophages predominate in advanced tumors [38]. In the large cell transformation of MF, miR-708 downregulation and upregulation of miR-146a and miR-21 were associated with the infiltration of M2 macrophages, suggesting their immunosuppressive role in the TME [39]. Furthermore, both CD68+ and CD163+ macrophages are associated with the advanced stage of CTCLs [40,41].
In several mechanistic studies, macrophages polarized by CTCL TME tend to support CTCL progression through the expression of cytokines and growth factors [42,43,44,45]. For example, inflammatory cytokines, including CXCL5, CCL13, and IL10, produced by periostin-induced TAMs were found to create an immunosuppressive tumor microenvironment, leading to MF development [42]. Furthermore, M2 macrophage cocultured with CTCL cells upregulated S100A9/TLR4 via NF-kB to induce apoptosis resistance, leading to CTCL progression [19]. More recently, Han et al. showed that PD-1+ M2 macrophages induced by CTCL TME through the NF-kB/STAT/JAK pathway can impair the phagocytic activity of macrophages, promoting CTCL growth [46]. Macrophages are also important drivers of angiogenesis in CTCLs, as described by Wu et al. [47]. The depletion of M2-like TAMs delayed CTCL development in xenograft mouse models, supporting their role in CTCL tumorigenesis [47].
Regarding monocytes, evidence suggests that malignant T cells can recruit monocytes via a CCL5-dependent manner to promote the survival of CTCL cells [48]. Furthermore, monocytes can interact with malignant T cells to promote immunosuppression and CTCL progression [19], as shown in Figure 1.

2.1.2. Mast Cells

Mast cells infiltrating the CTCL TME have been established as key players in the disease processes. In CTCL lesions, increased mast cells not only show a positive relationship with tumor stage but also microvessel density, suggesting their role in inducing angiogenesis. In support of this, delayed tumor growth was found in a cutaneous lymphoma mouse model deficient in mast cells [49]. In MF, a high number of mast cells and tryptase are drivers of itch and MF disease severity [50]. In contrast, Eder et al. found higher mast cells in clinical stage IA and IB patients than in the IIA and IIB stages [51], suggesting that a higher number of mast cells may not necessarily reflect the stage of CTCLs.

2.1.3. Eosinophils and Neutrophils

Eosinophils and neutrophils are important components of innate cells with key roles in host defense mechanisms. However, signals within the TME can alter their physiology to support tumor progression [52,53,54,55]. Studies have shown that a high density of eosinophils either in blood [56] or skin lesions is associated with the aggressiveness of the disease [14,56]. Similar to other innate immune cells, the activation of eosinophils drives inflammation in CTCLs to accelerate disease progression [14,57]. In a study aimed at elucidating eosinophils-activating factors in CTCL TME, IL5 and high mobility BOX-1 protein (HMGB1) expressed by malignant T cells were identified as key activators of eosinophils in MF [58]. Moreover, a high infiltration of neutrophils mediated by IL17 and IL8 is linked with MF and SS disease progression [59,60].

2.1.4. Dendritic Cells and Natural Killer Cells

Dendritic cells (DCs) are the most efficient antigen-presenting cells noted for capturing and presenting antigens to naïve T cells [61]. Dendritic cells can exhibit both anti-tumor and protumor functions depending on the prevailing conditions within the TME [61]. Whereas mature dendritic cells play antitumor functions, immature dendritic cells foster immune tolerance, thus promoting tumor progression [62]. Berger et al. cocultured immature dendritic cells with CTCL cells from SS patients and found that immature dendritic cells can sustain the growth of CTCLs [63]. Furthermore, DCs can promote the migration of SS and MF cells [64]. OX40 is a costimulatory signal that promotes T cell expansion and survival. In a recent study, the activation of benign T cells by OX40L+CD40L+ dendritic cells stimulated inflammation and the release of tumorigenic signals to CTCLs [65]. Natural killer cells (NKs) are lymphoid members of the innate immune system playing cytotoxic functions similar to CD8+ T cells. In CTCLs, malignant T cells from SS patients can reduce CD16+CD56dim NK cells and downregulate NKG2D, the main activator of antitumor activity in NK cells, to promote their escape from NK-induced antitumor immunity [66]. Additionally, the increased expression of NK cell receptor KIR3DL2 has been found in MF [67].

2.1.5. Myeloid-Derived Suppressor Cells (MDSCs)

MDSCs represent a heterogeneous population of immune cells implicated in many pathologic conditions, including cancers [15,68]. In CTCLs, MDSC accumulation is linked with worse clinical outcomes in patients [69] and the advanced stage of SS and MF [70]. Furthermore, MDSCs have been shown to express high levels of arginase and nitric oxide (NO) to potentiate immunosuppression and CTCL progression [20]. Maliniemi et al. showed that CD33+ myeloid suppressor cells express indoleamine 2,3-deoxygenase 1, an immune checkpoint molecule, supporting their role in immunosuppression [71].

2.1.6. Tumor-Infiltrating Lymphocytes

Tumor-infiltrating lymphocytes are composed of heterogeneous immune cells with the primary function of clearing tumor cells. However, immunosuppressive factors in the TME hampers their function to facilitate tumor escape and progression [72]. Largely, markers associated with T cell exhaustion, including PD-1, CTLA-4, LAG-3, TIGIT, and TIM-3 [73,74], have been shown as crucial players in cancer-mediated immunosuppression in CTCLs. Furthermore, emerging evidence shows that microbiota present in CTCL TME can negatively regulate antitumor immunity, as demonstrated by Blümel et al. [75]. Here, authors established that staphylococcal alpha-toxin can promote the escape of CTCL cells from CD8+ T cell-mediated killing, hence facilitating SS progression [75]. Additionally, the cytotoxic functions of lymphocytes can be impaired through CTCL-mediated inhibition of cytokines involved in T cell priming. For example, SS cells can attenuate the cytotoxic functions of CD8+ T cells by suppressing their responsiveness to IL10 [76]. Furthermore, Zhen et al. have established that increased expression of miR-155, -130, and -21 in Hut78 and Myla cell lines can induce CD8+ T cell exhaustion, leading to CTCL progression [77].
B cells infiltrating the CTCL tumor microenvironment contribute to the pathophysiology of the disease. It has been shown that there is a high infiltration of B cells in MF patients compared to healthy controls [78]. Additionally, the high infiltration of B cells correlates positively with MF progression [78]. Functionally, B cells in the MF tumor microenvironment release immunosuppressive cytokines, contributing significantly to tumor cell growth, dissemination, angiogenesis, and immunosuppression [13].

2.1.7. Regulatory T Cells

In MF, Tregs have diverse functions with contrasting roles having been reported. While the early patch stage of MF is associated with high Treg numbers, a low number of FOXP3+ cells are found in the advanced stage [16,79,80,81]. Indeed, the reduced expression of FOXP3+ cells relative to CD3+ T cells in the early stages of MF correlates with the disease progression [82]. On the contrary, a high infiltration of Tregs correlates with good clinical outcomes in MF patients [82]. In SS, IL10 and TGF-β secreted by Tregs can suppress the secretion of IL2 and IFN-γ and maintain DC immaturity, leading to CTCL proliferation [83]. Similarly, high Tregs(CD4+ CD25+) in SS patients have been found to suppress the proliferation of autologous CD4+ CD25- responder T cells [20].
Evidence has shown that the microbiota in CTCL TME can augment the tumor-promoting functions of Tregs [84]. According to Willerslev-Olsen et al. Staphylococcus aureus enterotoxins (SEA) can induce FOXP3 expression in malignant SS cells via the STAT5 pathway [84].

2.2. Role of Cancer-Associated Fibroblasts in CTCL

Cancer-associated fibroblasts (CAF) are major components of the TME known to potentiate the immune escape of tumor cells [85,86]. In CTCLs, CAF can modulate the expression of biomarkers associated with CTCL pathogenesis, attenuate Th1-related cytokines, and promote the Th2-dominant microenvironment, leading to CTCL progression [87,88]. For example, MF cells were found to induce normal fibroblast to express high levels of TWIST1 and TOX and Th2 markers, such as GATA3, IL6, and IL4 but low levels of Th1 markers, IFNG and TBX2 [87] (Figure 2).
Data have shown that chemokines secreted by CAF can potentiate their tumor-promoting functions [22,29,89]. Specifically, CAF-induced CXCR4/SDF promoted SS cell migration by downregulating CD26/dipeptidyl peptidase IV [29]. Relatedly, eotaxins derived from dermal fibroblast can interact with CCR3+ lymphocytes to promote CTCL development [89]. Furthermore, CXCL12/CXCR4 secreted by MF-derived CAF was found to protect malignant T cells from doxorubicin-induced apoptosis, thereby enhancing the migration of MF cells [22]. Beksac et al. also cocultured fibroblast and malignant MF cells isolated from the skin of early-stage CTCLs and found that fibroblast can enhance the proliferation of MF cells [33].

2.3. Role of Vascular or Endothelial Cells in CTCLs

Angiogenesis, characterized by vascular or lymphatic vessel formation, is an important process for tumor dissemination [90,91,92]. Several molecular and correlative studies have detailed the indispensable role of endothelial cells and their related markers in CTCL pathogenesis. In MF and SS, the expression levels of VEGFR-3, VEGF-C, and other angiogenic markers, such as CD31, podoplanin, and LYVE-1, correlate significantly with disease progression [93,94,95,96]. Besides tissue expressions, serum levels of VEGF-A reflect the severity of itching in MF and SS patients [97]. Furthermore, a positive correlation between podoplanin expression and lymphatic vessel density in malignant T cells has been linked to tumor aggressiveness and advanced stage of MF [98]. Moreover, intertumoral SOX18, a marker of neovascularization, correlates with MF disease progression, cutaneous involvement, and metastasis [28].
Mechanistically, in situ expression of LTα driven by the aberrant activation of the JAK3/STAT5 pathway acts in an autocrine fashion via TNF-alpha receptor 2 to induce IL6 expression in malignant T cells, which together with VEGF induce tube formation and endothelial cell sprouting [18]. According to Lauenborg et al. IL17F derived from Myla supernatant can stimulate angiogenesis through tube formation and sprouting to facilitate CTCL progression [17]. Furthermore, placental growth factor (PlGF) and VEGF-A expressed in CTCL skin were found to promote tumor growth via tumor vasculature formation [32]. The same study found serum levels of PIG4 to correlate with MF/SS disease severity, suggesting a possible utility of PIG4 either as a biomarker or potential therapeutic target [32]. Additionally, VEGFR-3 expressed in CTCL cell lines and a xenograft mouse model of MF exhibited a protective effect towards the suberoylanilide hydroxamic acid (SAHA)-mediated inhibition of tumor cells, hence promoting tumor progression [23], as shown in Figure 3.

3. Molecular Mechanisms of CTCL Immune Evasion

Immune evasion of CTCLs involves several mechanisms, including the secretion of immunosuppressive factors, such as cytokines and exosomal cargos, genetic alterations, immune checkpoint-mediated T cell inhibition, and apoptosis resistance, as shown in Figure 4.

3.1. Apoptosis Resistance

Apoptosis evasion is a hallmark of cancer progression. This process is characterized by a decrease in the function of pro-apoptotic proteins and/or an increase in anti-apoptotic proteins. They can block cell death signals, hence promoting apoptosis resistance [99]. Fas ligand (FasL) expressed on cytotoxic T cells plays an important role in the Fas-mediated killing of tumor cells. However, FasL expressed on tumor cells can counterattack the tumor-killing abilities of tumor-infiltrating lymphocytes [100]. In support of this, Ni et al. have found that FasL expressed by malignant T cells and epidermal keratinocytes can induce the apoptosis of CD8+ T cells and MF progression [101]. Indeed, fewer CD8+ T cells appear to be distributed in the vicinity of FasL-positive tumor cells.
CTCL progression is dependent on their ability to escape activation-induced cell death (AICD) [24,25,26,27]. According to Klemke et al., malignant T cells from SS patients show reduced surface expression of CD95L, an apoptosis-inducing ligand, upon TCR stimulation, leading to AICD resistance [24]. Relatedly, the overexpression of E3 ubiquitin ligase c-CBL in CTCL cells inhibits AICD [25]. Genomic instability [102], mutations in genes [103], dysregulation of cytokines, and signaling pathways [104,105] are other mechanisms implicated in apoptosis resistance in CTCLs.

3.2. Cytokine Dysregulation in CTCLs

The pathophysiology of cancers is impacted considerably by the cytokine milieu of its environment [91,106,107,108]. Specifically, alterations in cytokines such as IL32, IL22, IL17F, IL17A, IL16, IL15, and HMGB1 [109,110] can create an immunosuppressive tumor microenvironment in CTCLs to facilitate immune evasion and tumor progression. According to Ito et al., C-C motif chemokine ligand 20 (CCL20) induced by IL22/IL22RA1 axis interacts with CCR6 receptor to promote migration and distant organ metastasis of CTCLs [31]. Intriguingly, increased CCL20 in clinical samples correlates positively with CTCL progression [111]. The elevated expression of 1L10 in malignant T cells facilitates tumor growth in vivo through IL10-mediated macrophage infiltration and M2 polarization [112]. Moreover, IL10 expressed by malignant T cells can impair the differentiation of monocytes to matured DCs, leading to antitumor suppression [113]. In a similar study, IKZF2-induced IL10 expression in malignant T cells was found to dampen the antitumor immunity of MHC II molecules, hence promoting apoptosis resistance and CTCL progression [21]. Indeed, increased 1L10 expression is associated with the clinical course of MF [21,112]. Ohmatsu et al. have indicated that the increased mRNA expression of IL32 not only predicts MF severity but can enhance the proliferation of CTCL cells [114]. Additionally, IL32 is known to upregulate survival genes [114,115], important for the initiation, maintenance, and progression of CTCLs [116]. IL16 and thymic stromal lymphopoietin (TSLP) expressed in the early stages of MF not only enhances the infiltration of malignant T cells into the skin but also contributes to CTCL proliferation [30]. The contribution of IL31 to CTCL pathogenesis appears to be diverse. While elevated serum levels of IL31 correlate positively with advanced disease stage [60] and pruritus [117], Santen et al. found low levels of IL31 in pruritic folliculotropic (FMF) but no expression in non-pruritic patients (MF) [118]. According to Mishra et al., the overexpression of IL15 by CD4+ T cells is associated with histone deacetylase histone (HDAC)1/6 upregulation and miRNA-21 activation, promoting CTCL progression [119]. Furthermore, the activation of mTORC1 by IL15 and IL2 in malignant CD4+ T cells promoted CTCL proliferation [120]. In addition to the above, Thode et al. have demonstrated that IL15 expressed by malignant T cells activates epidermal keratinocytes to promote CTCL proliferation [121]. It is interesting to note that while IL15 expression in skin-homing CD4+ T cells and peripheral blood CD4+ T cells correlated with CTCL disease progression [119], no correlation was found between IL15 miRNA expression in malignant T cells and CTCL advancement [122]. Additionally, IL17F induced by malignant T cells has been shown to promote the malignant transformation of MF cells and angiogenesis in CTCLs [17]. Senda and coworkers assessed the role of HMGB1 in CTCLs and found that high levels of HMGB1 in skin lesions and sera are associated with increased Th2 immune response and the induction of angiogenesis [123].

3.3. Genetic Alterations in CTCLs

Genetic alterations, including somatic mutation and mutagenic pathways, are important regulators of several cancer types, including CTCLs [124]. Evidence indicates that p53 mutation is linked with MF progression and predicts poor survival in patients [34,125]. Consequentially, p53 mutation status has been proposed as a possible biomarker to stratify patients at risk of advanced MF disease [125]. In a more mechanistic study, the dysregulation of p53 function has been shown to protect CTCL cells from apoptosis [126,127]. Several lines of evidence have shown that p21 dysregulation is associated with increased proliferation of CTCL cells [35,36,128,129]. Moreover, KRAS mutation promotes apoptosis resistance and predicts poor prognosis in MF patients [130,131]. In MF and SS, disease progression can be potentiated by alterations in CARD11 [103], TNFRSF1B [132], PLCG1 [133], and KIT [134]. Additionally, evidence from several whole-genomic and whole-exon sequencing studies suggests that mutation in NOTCH2 [34], TNFRSF1B, CTLA4-CD28 fusion [132], RB1, PTEN, DNMT3A, CDKN1B [103], CARD11, CDKN2A, and CCR4 [135] can promote CTCL progression. Although mutations in these genes are not frequently encountered in MF and SS patients, they can serve as potential therapeutic targets for CTCL patients. According to McGirt et al., JAK3 mutation in MF cells can induce apoptosis resistance and enhance CTCL proliferation [34].
Cancer-associated microbiota are important players in cancer progression [107,136,137]. In a study by Willerslev-Olsen et al., staphylococcal enterotoxin A (SEA) cocultured with non-malignant T cells was found to activate the STAT3/JAK3 pathway and induce IL17 expression [137]. Relatedly, IL17 and IL22 induced by STAT3 hyperactivation in a bacterial-dominated environment were found to enhance the proliferation of CTCL cells [136]. In a genomic analysis conducted in mice and humans, genetic instability mediated by a mutation in telomere-binding factor (TBF) was linked to CTCL development [138].

3.4. Immune Checkpoint-Mediated Suppression of T Cells

Increased expression of immune checkpoint molecules, including CTLA-4 [73], PD-1 [21,139,140], PD-L1 [73,139,141], and ICOS [73,141], on malignant T cells correlates positively with the advanced disease stage of CTCLs. Detailed insight has revealed that PD-L1 expression in CTCL cell lines can induce M2 macrophages to promote CTCL growth [142]. Furthermore, it has been found that increased PD-1 can impair antitumor immune response and promote Th2 responses, which facilitate CTCL tumor growth [143,144]. The available evidence supports the notion that reversing T cell exhaustion is key to restoring T cell function. Although this has largely been welcomed as a potential therapeutic strategy, an integrated genomic analysis in humans and a mice model of T cell lymphomas has found that, while the loss of PD-1 function promotes the reversal of T cell exhaustion, this is associated with FOXM1-mediated transcriptional signature, leading to poor prognostic outcomes in SS and MF patients [145].

3.5. Exosomes in CTCLs

Studies on exosomes have demonstrated their involvement in all aspects of tumorigenesis, including invasion and migration, angiogenesis induction, and tumor escape from immunosurveillance [146,147,148]. In the CTCL context, the available evidence indicates that miR- 155 derived from MF cell lines can enhance the migratory effect of MF cells. Interestingly, plasma exosomes from MF patients enhances the migration of normal peripheral blood mononuclear cells in a coculture system [149]. However, considering the large body of evidence on the diverse role of exosomes in tumorigenesis across several cancer types, further insights are required to fully understand the role of exosomes in CTCLs.

4. Advances in Strategies to Target CTCLs

The past few years have seen a great improvement in cancer treatment through a combination of agents or drugs targeting the CTCL tumor microenvironment. Specifically, targeting immune evasion mechanisms of malignant T cells and other populations within the TME contributing to CTCL tumorigenesis is promising. Immune checkpoint inhibitors, including pembrolizumab, durvalumab, and ontorpacept (TTI-621; SIRPα-IgG1 Fc), have shown significant antitumor activity with durable and long-lasting responses with manageable toxicity profiles in CTCL patients [150,151,152,153,154]. The intralesional application of ontorpacept (TTI-621) has led to activity in adjacent or distal non-injected lesions, suggesting systemic and locoregional abscopal effects [153]. Studies have shown that anti-PD-L1 (durvalumab), lenalidomide, and TTI-621 can re-program M2 macrophages to boost antitumor functions against CTCL cells [46,142]. Furthermore, the depletion of macrophages in a CTCL murine model using CCR2 inhibitors can synergize with anti-PD-1 to suppress tumor growth [155]. Even in some refractory SS patients, anti-PD-1 in combination with HDAC inhibitors can promote durable clinical response [156]. In a Phase1/2 trial of anti-PD-L1 (durvalumab) and lenalidomide in CTCL patients, Querfeld et al. showed that durvalumab and lenalidomide are associated with significant clinical activity in refractory and advanced patients [157].
Given the role of cytokines and chemokines in CTCL pathogenesis, studies exploring their therapeutic potential on the CTCL TME have also emerged [151,158]. EQ101 (formerly known as BNZ-1) is a synthetic peptide, designed to selectively inhibit IL-2, IL-9, and IL-15 binding to the common gamma chain (γc) signaling receptor, leading to the depletion of Tregs and tumor growth suppression [159]. Moreover, denileukin diftitox, a recombinant fusion protein of IL2 and diphtheria toxin, targets the IL2 receptor on malignant T cells and Tregs [20]. The reengineered drug denileukin difitox-cxdl (E7777) shows improved safety and tolerability and was FDA-approved in August 2024 for relapsed/refractory CTCLs. Mogamulizumab, which is a humanized anti-CCR4, exhibits potent clinical efficacy against CCR4-positive CTCLs and other T cell lymphomas and was shown to efficiently decrease Tregs, leading to CTCL growth inhibition [160]. KIR3DL2 expression is upregulated on all subtypes of CTCLs. Anti-KIR3DL2 monoclonal antibody (IPH4102) has shown promise in depleting the KIR3DL2 receptor in malignant T cells of CTCL patients [161]. IPH4102 has been shown to recruit human effector NK cells as well as macrophages to eliminate KIR3DL2+ T cells via antibody-dependent cell cytotoxicity and antibody-dependent cell phagocytosis, respectively. Wang et al. compared the efficacy of CCR4-IL2 bispecific immunotoxin with brentuximab. Using an immunodeficient NSG mouse model of CTCLs, the study found that CCR4-IL2 bispecific immunotoxin was more effective in prolonging survival than brentuximab [158].

5. Conclusions

Malignant T cells can turn their environment into a hospitable home to promote their survival, growth, and progression. Hence, in our quest to uncover the therapeutic potential of CTCL TME, agents or drugs should not only target malignant T cells but interfere with their key defensive mechanisms and abrogate their ability to evade the antitumor immune response. To this end, the reprogramming of protumorigenic immune cells to gain their antitumor functions and apoptosis induction of CTLC cells holds promise in CTCL treatment. Such a holistic approach will open new opportunities in the treatment of relapsing and refractory CTCL patients to yield durable clinical responses.

Author Contributions

Conceptualization, L.B.K. and C.Q.; writing—original draft preparation, L.B.K.; writing—review and editing, S.T.R. and C.Q. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported in part through the National Institutes of Health (NIH)/National Cancer Institute (NCI) Cancer Center Support Grant (P30CA 033572) to the City of Hope, NIH/NCI grant (R01 CA229510-01) and The Leukemia and Lymphoma Society Clinical Scholar Award to C. Querfeld, used in the capacity of mentoring coauthor.

Acknowledgments

C. Querfeld is a Scholar in Clinical Research of The Leukemia and Lymphoma Society.

Conflicts of Interest

L.B.K: None to declare. S.T.R: Is a consultant with Pepromene Bio, Inc; Abbvie; is a member of the Educational Advisory Board of Pepromene Bio, Inc; and has stock options with Pepromene Bio, Inc. C.Q.: Consultant to Helsinn, Kyowa Kirin, and Citius Pharmaceuticals Inc; contracted clinical investigator to Kyowa Kirin, Sirpant immunotherapeutics, Bristol Myers Squibb, and BioInvent; received research grants from Helsinn and Kyowa Kirin.

References

  1. Rubio Gonzalez, B.; 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] [PubMed]
  2. Agar, N.S.; Wedgeworth, E.; Crichton, S.; Mitchell, T.J.; Cox, M.; Ferreira, S.; Robson, A.; Calonje, E.; Stefanato, C.M.; Wain, E.M.; et al. Survival outcomes and prognostic factors in mycosis fungoides/Sézary syndrome: Validation of the revised International Society for Cutaneous Lymphomas/European Organisation for Research and Treatment of Cancer staging proposal. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2010, 28, 4730–4739. [Google Scholar] [CrossRef] [PubMed]
  3. Criscione, V.D.; Weinstock, M.A. Incidence of cutaneous T-cell lymphoma in the United States, 1973–2002. Arch. Dermatol. 2007, 143, 854–859. [Google Scholar] [CrossRef] [PubMed]
  4. Trautinger, F.; Eder, J.; Assaf, C.; Bagot, M.; Cozzio, A.; Dummer, R.; Gniadecki, R.; Klemke, C.D.; Ortiz-Romero, P.L.; Papadavid, E.; et al. European Organisation for Research and Treatment of Cancer consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome—Update 2017. Eur. J. Cancer 2017, 77, 57–74. [Google Scholar] [CrossRef]
  5. 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]
  6. Najidh, S.; Tensen, C.P.; van der Sluijs-Gelling, A.J.; Teodosio, C.; Cats, D.; Mei, H.; Kuipers, T.B.; Out-Luijting, J.J.; Zoutman, W.H.; van Hall, T.; et al. Improved Sézary cell detection and novel insights into immunophenotypic and molecular heterogeneity in Sézary syndrome. Blood 2021, 138, 2539–2554. [Google Scholar] [CrossRef]
  7. Wilcox, R.A. Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2016, 91, 151–165. [Google Scholar] [CrossRef]
  8. Lessin, S.R.; Duvic, M.; Guitart, J.; Pandya, A.G.; Strober, B.E.; Olsen, E.A.; Hull, C.M.; Knobler, E.H.; Rook, A.H.; Kim, E.J.; et al. Topical chemotherapy in cutaneous T-cell lymphoma: Positive results of a randomized, controlled, multicenter trial testing the efficacy and safety of a novel mechlorethamine, 0.02%, gel in mycosis fungoides. JAMA Dermatol. 2013, 149, 25–32. [Google Scholar] [CrossRef]
  9. Quaglino, P.; Maule, M.; Prince, H.M.; Porcu, P.; Horwitz, S.; Duvic, M.; Talpur, R.; Vermeer, M.; Bagot, M.; Guitart, J.; et al. Global patterns of care in advanced stage mycosis fungoides/Sezary syndrome: A multicenter retrospective follow-up study from the Cutaneous Lymphoma International Consortium. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 2517–2525. [Google Scholar] [CrossRef]
  10. Kim, Y.H.; Bagot, M.; Pinter-Brown, L.; Rook, A.H.; Porcu, P.; Horwitz, S.M.; Whittaker, S.; Tokura, Y.; Vermeer, M.; Zinzani, P.L.; et al. Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): An international, open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2018, 19, 1192–1204. [Google Scholar] [CrossRef]
  11. Prince, H.M.; Kim, Y.H.; Horwitz, S.M.; Dummer, R.; Scarisbrick, J.; Quaglino, P.; Zinzani, P.L.; Wolter, P.; Sanches, J.A.; Ortiz-Romero, P.L.; et al. Brentuximab vedotin or physician’s choice in CD30-positive cutaneous T-cell lymphoma (ALCANZA): An international, open-label, randomised, phase 3, multicentre trial. Lancet 2017, 390, 555–566. [Google Scholar] [CrossRef] [PubMed]
  12. Guo, W.; Liu, G.M.; Guan, J.Y.; Chen, Y.J.; Zhao, Y.Z.; Wang, K.; Bai, O. Epigenetic regulation of cutaneous T-cell lymphoma is mediated by dysregulated lncRNA MALAT1 through modulation of tumor microenvironment. Front. Oncol. 2022, 12, 977266. [Google Scholar] [CrossRef] [PubMed]
  13. Gaydosik, A.M.; Stonesifer, C.J.; Tabib, T.; Lafyatis, R.; Geskin, L.J.; Fuschiotti, P. The mycosis fungoides cutaneous microenvironment shapes dysfunctional cell trafficking, antitumor immunity, matrix interactions, and angiogenesis. JCI Insight 2023, 8, e170015. [Google Scholar] [CrossRef] [PubMed]
  14. Ionescu, M.A.; Rivet, J.; Daneshpouy, M.; Briere, J.; Morel, P.; Janin, A. In situ eosinophil activation in 26 primary cutaneous T-cell lymphomas with blood eosinophilia. J. Am. Acad. Dermatol. 2005, 52, 32–39. [Google Scholar] [CrossRef] [PubMed]
  15. Kwantwi, L.B.; Rosen, S.T.; Querfeld, C. The role of signaling lymphocyte activation molecule family receptors in hematologic malignancies. Curr. Opin. Oncol. 2024, 36, 449–455. [Google Scholar] [CrossRef] [PubMed]
  16. Alcántara-Hernández, M.; Torres-Zárate, C.; Pérez-Montesinos, G.; Jurado-Santacruz, F.; Domínguez-Gómez, M.A.; Peniche-Castellanos, A.; Ferat-Osorio, E.; Neri, N.; Nambo, M.J.; Alvarado-Cabrero, I.; et al. Overexpression of hypoxia-inducible factor 1 alpha impacts FoxP3 levels in mycosis fungoides--cutaneous T-cell lymphoma: Clinical implications. Int. J. Cancer 2014, 134, 2136–2145. [Google Scholar] [CrossRef]
  17. Lauenborg, B.; Litvinov, I.V.; Zhou, Y.; Willerslev-Olsen, A.; Bonefeld, C.M.; Nastasi, C.; Fredholm, S.; Lindahl, L.M.; Sasseville, D.; Geisler, C.; et al. Malignant T cells activate endothelial cells via IL-17 F. Blood Cancer J. 2017, 7, e586. [Google Scholar] [CrossRef]
  18. Lauenborg, B.; Christensen, L.; Ralfkiaer, U.; Kopp, K.L.; Jønson, L.; Dabelsteen, S.; Bonefeld, C.M.; Geisler, C.; Gjerdrum, L.M.; Zhang, Q.; et al. Malignant T cells express lymphotoxin α and drive endothelial activation in cutaneous T cell lymphoma. Oncotarget 2015, 6, 15235–15249. [Google Scholar] [CrossRef]
  19. Du, Y.; Cai, Y.; Lv, Y.; Zhang, L.; Yang, H.; Liu, Q.; Hong, M.; Teng, Y.; Tang, W.; Ma, R.; et al. Single-cell RNA sequencing unveils the communications between malignant T and myeloid cells contributing to tumor growth and immunosuppression in cutaneous T-cell lymphoma. Cancer Lett. 2022, 551, 215972. [Google Scholar] [CrossRef]
  20. Geskin, L.J.; Akilov, O.E.; Kwon, S.; Schowalter, M.; Watkins, S.; Whiteside, T.L.; Butterfield, L.H.; Falo, L.D. Therapeutic reduction of cell-mediated immunosuppression in mycosis fungoides and Sézary syndrome. Cancer Immunol. Immunother. 2018, 67, 423–434. [Google Scholar] [CrossRef]
  21. Xu, B.; Liu, F.; Gao, Y.; Sun, J.; Li, Y.; Lin, Y.; Liu, X.; Wen, Y.; Yi, S.; Dang, J.; et al. High Expression of IKZF2 in Malignant T Cells Promotes Disease Progression in Cutaneous T Cell Lymphoma. Acta Derm. Venereol. 2021, 101, adv00613. [Google Scholar] [CrossRef] [PubMed]
  22. Aronovich, A.; Moyal, L.; Gorovitz, B.; Amitay-Laish, I.; Naveh, H.P.; Forer, Y.; Maron, L.; Knaneh, J.; Ad-El, D.; Yaacobi, D.; et al. Cancer-Associated Fibroblasts in Mycosis Fungoides Promote Tumor Cell Migration and Drug Resistance through CXCL12/CXCR4. J. Investig. Dermatol. 2021, 141, 619–627.e612. [Google Scholar] [CrossRef] [PubMed]
  23. Pedersen, I.H.; Willerslev-Olsen, A.; Vetter-Kauczok, C.; Krejsgaard, T.; Lauenborg, B.; Kopp, K.L.; Geisler, C.; Bonefeld, C.M.; Zhang, Q.; Wasik, M.A.; et al. Vascular endothelial growth factor receptor-3 expression in mycosis fungoides. Leuk. Lymphoma 2013, 54, 819–826. [Google Scholar] [CrossRef] [PubMed]
  24. Klemke, C.-D.; Brenner, D.; Weiβ, E.-M.; Schmidt, M.; Leverkus, M.; Gülow, K.; Krammer, P.H. Lack of T-Cell Receptor–Induced Signaling Is Crucial for CD95 Ligand Up-regulation and Protects Cutaneous T-Cell Lymphoma Cells from Activation-Induced Cell Death. Cancer Res. 2009, 69, 4175–4183. [Google Scholar] [CrossRef]
  25. Wu, J.; Salva, K.A.; Wood, G.S. c-CBL E3 ubiquitin ligase is overexpressed in cutaneous T-cell lymphoma: Its inhibition promotes activation-induced cell death. J. Investig. Dermatol. 2015, 135, 861–868. [Google Scholar] [CrossRef]
  26. Ni, X.; Zhang, C.; Talpur, R.; Duvic, M. Resistance to activation-induced cell death and bystander cytotoxicity via the Fas/Fas ligand pathway are implicated in the pathogenesis of cutaneous T cell lymphomas. J. Investig. Dermatol. 2005, 124, 741–750. [Google Scholar] [CrossRef]
  27. Wang, Y.; Su, M.; Zhou, L.L.; Tu, P.; Zhang, X.; Jiang, X.; Zhou, Y. Deficiency of SATB1 expression in Sezary cells causes apoptosis resistance by regulating FasL/CD95L transcription. Blood 2011, 117, 3826–3835. [Google Scholar] [CrossRef]
  28. Jankowska-Konsur, A.; Kobierzycki, C.; Reich, A.; Piotrowska, A.; Gomulkiewicz, A.; Olbromski, M.; Podhorska-Okołów, M.; Dzięgiel, P.; Szepietowski, J.C. Expression of SOX18 in Mycosis Fungoides. Acta Derm. Venereol. 2017, 97, 17–23. [Google Scholar] [CrossRef]
  29. Narducci, M.G.; Scala, E.; Bresin, A.; Caprini, E.; Picchio, M.C.; Remotti, D.; Ragone, G.; Nasorri, F.; Frontani, M.; Arcelli, D.; et al. Skin homing of Sézary cells involves SDF-1-CXCR4 signaling and down-regulation of CD26/dipeptidylpeptidase IV. Blood 2006, 107, 1108–1115. [Google Scholar] [CrossRef]
  30. Tuzova, M.; Richmond, J.; Wolpowitz, D.; Curiel-Lewandrowski, C.; Chaney, K.; Kupper, T.; Cruikshank, W. CCR4+ T cell recruitment to the skin in mycosis fungoides: Potential contributions by thymic stromal lymphopoietin and interleukin-16. Leuk. Lymphoma 2015, 56, 440–449. [Google Scholar] [CrossRef]
  31. 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]
  32. Miyagaki, T.; Sugaya, M.; Oka, T.; Takahashi, N.; Kawaguchi, M.; Suga, H.; Fujita, H.; Yoshizaki, A.; Asano, Y.; Sato, S. Placental Growth Factor and Vascular Endothelial Growth Factor Together Regulate Tumour Progression via Increased Vasculature in Cutaneous T-cell Lymphoma. Acta Derm. Venereol. 2017, 97, 586–592. [Google Scholar] [CrossRef] [PubMed]
  33. Beksaç, B.; Gleason, L.; Baik, S.; Ringe, J.M.; Porcu, P.; Nikbakht, N. Dermal fibroblasts promote cancer cell proliferation and exhibit fibronectin overexpression in early mycosis fungoides. J. Dermatol. Sci. 2022, 106, 53–60. [Google Scholar] [CrossRef] [PubMed]
  34. 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. Whole-genome sequencing reveals oncogenic mutations in mycosis fungoides. Blood J. Am. Soc. Hematol. 2015, 126, 508–519. [Google Scholar] [CrossRef]
  35. Gluud, M.; Fredholm, S.; Blümel, E.; Willerslev-Olsen, A.; Buus, T.B.; Nastasi, C.; Krejsgaard, T.; Bonefeld, C.M.; Woetmann, A.; Iversen, L.; et al. MicroRNA-93 Targets p21 and Promotes Proliferation in Mycosis Fungoides T Cells. Dermatology 2021, 237, 277–282. [Google Scholar] [CrossRef]
  36. Wang, Y.; Gu, X.; Li, W.; Zhang, Q.; Zhang, C. PAK1 overexpression promotes cell proliferation in cutaneous T cell lymphoma via suppression of PUMA and p21. J. Dermatol. Sci. 2018, 90, 60–67. [Google Scholar] [CrossRef]
  37. Kumari, N.; Choi, S.H. Tumor-associated macrophages in cancer: Recent advancements in cancer nanoimmunotherapies. J. Exp. Clin. Cancer Res. 2022, 41, 68. [Google Scholar] [CrossRef]
  38. Johanny, L.D.; Sokumbi, O.; Hobbs, M.M.; Jiang, L. Polarization of Macrophages in Granulomatous Cutaneous T Cell Lymphoma Granulomatous Mycosis Fungoides Microenvironment. Dermatopathology 2022, 9, 54–59. [Google Scholar] [CrossRef]
  39. Di Raimondo, C.; Han, Z.; Su, C.; Wu, X.; Qin, H.; Sanchez, J.F.; Yuan, Y.-C.; Martinez, X.; Abdulla, F.; Zain, J.; et al. Identification of a Distinct miRNA Regulatory Network in the Tumor Microenvironment of Transformed Mycosis Fungoides. Cancers 2021, 13, 5854. [Google Scholar] [CrossRef]
  40. Huang, S.; Liao, M.; Chen, S.; Zhang, P.; Xu, F.; Zhang, H. Immune signatures of CD4 and CD68 predicts disease progression in cutaneous T cell lymphoma. Am. J. Transl. Res. 2022, 14, 3037–3051. [Google Scholar]
  41. El-Guindy, D.M.; Elgarhy, L.H.; Elkholy, R.A.; Ali, D.A.; Helal, D.S. Potential role of tumor-associated macrophages and CD163/CD68 ratio in mycosis fungoides and Sézary syndrome in correlation with serum sCD163 and CCL22. J. Cutan. Pathol. 2022, 49, 261–273. [Google Scholar] [CrossRef] [PubMed]
  42. 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]
  43. 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, e60–e67. [Google Scholar] [CrossRef]
  44. Günther, C.; Zimmermann, N.; Berndt, N.; Grosser, M.; Stein, A.; Koch, A.; Meurer, M. Up-regulation of the chemokine CCL18 by macrophages is a potential immunomodulatory pathway in cutaneous T-cell lymphoma. Am. J. Pathol. 2011, 179, 1434–1442. [Google Scholar] [CrossRef]
  45. Sadhukhan, P.; Seiwert, T.Y. The role of macrophages in the tumor microenvironment and tumor metabolism. Semin. Immunopathol. 2023, 45, 187–201. [Google Scholar] [CrossRef]
  46. Han, Z.; Wu, X.; Qin, H.; Yuan, Y.C.; Schmolze, D.; Su, C.; Zain, J.; Moyal, L.; Hodak, E.; Sanchez, J.F.; et al. Reprogramming of PD-1+ M2-like tumor-associated macrophages with anti-PD-L1 and lenalidomide in cutaneous T cell lymphoma. JCI Insight 2023, 8, e163518. [Google Scholar] [CrossRef]
  47. Wu, X.; Schulte, B.C.; Zhou, Y.; Haribhai, D.; Mackinnon, A.C.; Plaza, J.A.; Williams, C.B.; Hwang, S.T. Depletion of M2-like tumor-associated macrophages delays cutaneous T-cell lymphoma development in vivo. J. Investig. Dermatol. 2014, 134, 2814–2822. [Google Scholar] [CrossRef]
  48. Wilcox, R.A.; David, W.A.; Feldman, A.L.; Elsawa, S.F.; Grote, D.M.; Ziesmer, S.C.; Novak, A.J.; Pittelkow, M.R.; Witzig, T.E.; Ansell, S.M. Monocytes Promote Survival of Malignant T Cells in Cutaneous T-Cell Lymphoma and Are Recruited to the Tumor Microenvironment by CCL5 (RANTES). Blood 2008, 112, 378. [Google Scholar] [CrossRef]
  49. Rabenhorst, A.; Schlaak, M.; Heukamp, L.C.; Förster, A.; Theurich, S.; von Bergwelt-Baildon, M.; Büttner, R.; Kurschat, P.; Mauch, C.; Roers, A.; et al. Mast cells play a protumorigenic role in primary cutaneous lymphoma. Blood 2012, 120, 2042–2054. [Google Scholar] [CrossRef]
  50. Terhorst-Molawi, D.; Lohse, K.; Ginter, K.; Puhl, V.; Metz, M.; Hu, M.; Maurer, M.; Altrichter, S. Mast cells and tryptase are linked to itch and disease severity in mycosis fungoides: Results of a pilot study. Front. Immunol. 2022, 13, 930979. [Google Scholar] [CrossRef]
  51. Eder, J.; Rogojanu, R.; Jerney, W.; Erhart, F.; Dohnal, A.; Kitzwögerer, M.; Steiner, G.; Moser, J.; Trautinger, F. Mast Cells Are Abundant in Primary Cutaneous T-Cell Lymphomas: Results from a Computer-Aided Quantitative Immunohistological Study. PLoS ONE 2016, 11, e0163661. [Google Scholar] [CrossRef] [PubMed]
  52. Peng, W.; Sheng, Y.; Xiao, H.; Ye, Y.; Kwantwi, L.B.; Cheng, L.; Wang, Y.; Xu, J.; Wu, Q. Lung Adenocarcinoma Cells Promote Self-Migration and Self-Invasion by Activating Neutrophils to Upregulate Notch3 Expression of Cancer Cells. Front. Mol. Biosci. 2022, 8, 762729. [Google Scholar] [CrossRef] [PubMed]
  53. Cai, Z.; Zhang, M.; Boafo Kwantwi, L.; Bi, X.; Zhang, C.; Cheng, Z.; Ding, X.; Su, T.; Wang, H.; Wu, Q. Breast cancer cells promote self-migration by secreting interleukin 8 to induce NET formation. Gene 2020, 754, 144902. [Google Scholar] [CrossRef] [PubMed]
  54. Sheng, Y.; Peng, W.; Huang, Y.; Cheng, L.; Meng, Y.; Kwantwi, L.B.; Yang, J.; Xu, J.; Xiao, H.; Kzhyshkowska, J.; et al. Tumor-activated neutrophils promote metastasis in breast cancer via the G-CSF-RLN2-MMP-9 axis. J. Leukoc. Biol. 2023, 113, 383–399. [Google Scholar] [CrossRef]
  55. Lee, T.-L.; Chen, T.-H.; Kuo, Y.-J.; Lan, H.-Y.; Yang, M.-H.; Chu, P.-Y. Tumor-associated tissue eosinophilia promotes angiogenesis and metastasis in head and neck squamous cell carcinoma. Neoplasia 2023, 35, 100855. [Google Scholar] [CrossRef]
  56. Diwan, A.H.; Prieto, V.G.; Herling, M.; Duvic, M.; Jone, D. Primary Sézary syndrome commonly shows low-grade cytologic atypia and an absence of epidermotropism. Am. J. Clin. Pathol. 2005, 123, 510–515. [Google Scholar] [CrossRef]
  57. Suzuki, H.; Sugaya, M.; Nakajima, R.; Oka, T.; Takahashi, N.; Nakao, M.; Miyagaki, T.; Asano, Y.; Sato, S. Serum amyloid A levels in the blood of patients with atopic dermatitis and cutaneous T-cell lymphoma. J. Dermatol. 2018, 45, 1440–1443. [Google Scholar] [CrossRef]
  58. Fredholm, S.; Gjerdrum, L.M.; Willerslev-Olsen, A.; Petersen, D.L.; Nielsen, I.; Kauczok, C.S.; Wobser, M.; Ralfkiaer, U.; Bonefeld, C.M.; Wasik, M.A.; et al. STAT3 activation and infiltration of eosinophil granulocytes in mycosis fungoides. Anticancer Res. 2014, 34, 5277–5286. [Google Scholar]
  59. Cirée, A.; Michel, L.; Camilleri-Bröet, S.; Jean Louis, F.; Oster, M.; Flageul, B.; Senet, P.; Fossiez, F.; Fridman, W.H.; Bachelez, H.; et al. Expression and activity of IL-17 in cutaneous T-cell lymphomas (Mycosis fungoides and Sezary syndrome). Int. J. Cancer 2004, 112, 113–120. [Google Scholar] [CrossRef]
  60. 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]
  61. Wylie, B.; Macri, C.; Mintern, J.D.; Waithman, J. Dendritic Cells and Cancer: From Biology to Therapeutic Intervention. Cancers 2019, 11, 521. [Google Scholar] [CrossRef] [PubMed]
  62. Devi, K.S.P.; Anandasabapathy, N. The origin of DCs and capacity for immunologic tolerance in central and peripheral tissues. Semin. Immunopathol. 2017, 39, 137–152. [Google Scholar] [CrossRef] [PubMed]
  63. Berger, C.L.; Hanlon, D.; Kanada, D.; Dhodapkar, M.; Lombillo, V.; Wang, N.; Christensen, I.; Howe, G.; Crouch, J.; El-Fishawy, P.; et al. The growth of cutaneous T-cell lymphoma is stimulated by immature dendritic cells. Blood 2002, 99, 2929–2939. [Google Scholar] [CrossRef] [PubMed]
  64. Schwingshackl, P.; Obermoser, G.; Nguyen, V.A.; Fritsch, P.; Sepp, N.; Romani, N. Distribution and maturation of skin dendritic cell subsets in two forms of cutaneous T-cell lymphoma: Mycosis fungoides and Sézary syndrome. Acta Derm. Venereol. 2012, 92, 269–275. [Google Scholar] [CrossRef]
  65. Vieyra-Garcia, P.; Crouch, J.D.; O’Malley, J.T.; Seger, E.W.; Yang, C.H.; Teague, J.E.; Vromans, A.M.; Gehad, A.; Win, T.S.; Yu, Z.; et al. Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma. JCI Insight 2019, 4, e124233. [Google Scholar] [CrossRef]
  66. Manfrere, C.K.; Torrealba, M.P.; Miyashiro, D.R.; Pereira, N.Z.; Yoshikawa, F.S.Y.; Oliveira, L.d.M.; Cury-Martins,, J.; Duarte, A.J.S.; Sanches, J.A.; Sato, M.N. Profile of differentially expressed Toll-like receptor signaling genes in the natural killer cells of patients with Sézary syndrome. Oncotarget 2017, 8, 92183–92194. [Google Scholar] [CrossRef]
  67. Sako, N.; Schiavon, V.; Bounfour, T.; Dessirier, V.; Ortonne, N.; Olive, D.; Ram-Wolff, C.; Michel, L.; Sicard, H.; Marie-Cardine, A.; et al. Membrane expression of NK receptors CD160 and CD158k contributes to delineate a unique CD4+ T-lymphocyte subset in normal and mycosis fungoides skin. Cytom. Part A 2014, 85, 869–882. [Google Scholar] [CrossRef]
  68. Kwantwi, L.B. SLAM family-mediated crosstalk between tumor and immune cells in the tumor microenvironment: A promising biomarker and a potential therapeutic target for immune checkpoint therapies. Clin. Transl. Oncol. 2024, 1–8. [Google Scholar] [CrossRef]
  69. Argyropoulos, K.V.; Pulitzer, M.; Perez, S.; Korkolopoulou, P.; Angelopoulou, M.; Baxevanis, C.; Palomba, M.L.; Siakantaris, M. Tumor-infiltrating and circulating granulocytic myeloid-derived suppressor cells correlate with disease activity and adverse clinical outcomes in mycosis fungoides. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. Natl. Cancer Inst. Mex. 2020, 22, 1059–1066. [Google Scholar] [CrossRef]
  70. Hergott, C.B.; Dudley, G.; Dorfman, D.M. Circulating Myeloid-Derived Suppressor Cells Reflect Mycosis Fungoides/Sezary Syndrome Disease Stage and Response to Treatment. Blood 2018, 132, 4127. [Google Scholar] [CrossRef]
  71. Maliniemi, P.; Laukkanen, K.; Väkevä, L.; Dettmer, K.; Lipsanen, T.; Jeskanen, L.; Bessede, A.; Oefner, P.J.; Kadin, M.E.; Ranki, A. Biological and clinical significance of tryptophan-catabolizing enzymes in cutaneous T-cell lymphomas. Oncoimmunology 2017, 6, e1273310. [Google Scholar] [CrossRef] [PubMed]
  72. Kwantwi, L.B.; Wang, S.; Zhang, W.; Peng, W.; Cai, Z.; Sheng, Y.; Xiao, H.; Wang, X.; Wu, Q. Tumor-associated neutrophils activated by tumor-derived CCL20 (C-C motif chemokine ligand 20) promote T cell immunosuppression via programmed death-ligand 1 (PD-L1) in breast cancer. Bioengineered 2021, 12, 6996–7006. [Google Scholar] [CrossRef] [PubMed]
  73. Querfeld, C.; Leung, S.; Myskowski, P.L.; Curran, S.A.; Goldman, D.A.; Heller, G.; Wu, X.; Kil, S.H.; Sharma, S.; Finn, K.J.; et al. Primary T Cells from Cutaneous T-cell Lymphoma Skin Explants Display an Exhausted Immune Checkpoint Profile. Cancer Immunol. Res. 2018, 6, 900–909. [Google Scholar] [CrossRef] [PubMed]
  74. Murray, D.; McMurray, J.L.; Eldershaw, S.; Pearce, H.; Davies, N.; Scarisbrick, J.J.; Moss, P. Progression of mycosis fungoides occurs through divergence of tumor immunophenotype by differential expression of HLA-DR. Blood Adv. 2019, 3, 519–530. [Google Scholar] [CrossRef]
  75. Blümel, E.; Munir Ahmad, S.; Nastasi, C.; Willerslev-Olsen, A.; Gluud, M.; Fredholm, S.; Hu, T.; Surewaard, B.G.J.; Lindahl, L.M.; Fogh, H.; et al. Staphylococcus aureus alpha-toxin inhibits CD8(+) T cell-mediated killing of cancer cells in cutaneous T-cell lymphoma. Oncoimmunology 2020, 9, 1751561. [Google Scholar] [CrossRef]
  76. Torrealba, M.P.; Manfrere, K.C.; Miyashiro, D.R.; Lima, J.F.; Oliveira, L.d.M.; Pereira, N.Z.; Cury-Martins, J.; Pereira, J.; Duarte, A.J.S.; Sato, M.N.; et al. Chronic activation profile of circulating CD8+ T cells in Sézary syndrome. Oncotarget 2018, 9, 3497–3506. [Google Scholar] [CrossRef]
  77. Han, Z.; Estephan, R.J.; Wu, X.; Su, C.; Yuan, Y.C.; Qin, H.; Kil, S.H.; Morales, C.; Schmolze, D.; Sanchez, J.F.; et al. MicroRNA Regulation of T-Cell Exhaustion in Cutaneous T Cell Lymphoma. J. Investig. Dermatol. 2022, 142, 603–612.e607. [Google Scholar] [CrossRef]
  78. Nielsen, P.R.; Eriksen, J.O.; Sørensen, M.D.; Wehkamp, U.; Lindahl, L.M.; Bzorek, M.; Iversen, L.; Woetman, A.; Ødum, N.; Litman, T.; et al. Role of B-cells in Mycosis Fungoides. Acta Derm. Venereol. 2021, 101, adv00413. [Google Scholar] [CrossRef]
  79. Shareef, M.M.; Elgarhy, L.H.; Wasfy Rel, S. Expression of Granulysin and FOXP3 in Cutaneous T Cell Lymphoma and Sézary Syndrome. Asian Pac. J. Cancer Cancer Prev. 2015, 16, 5359–5364. [Google Scholar] [CrossRef]
  80. Fried, I.; Cerroni, L. FOXP3 in Sequential Biopsies of Progressive Mycosis Fungoides. Am. J. Dermatopathol. 2012, 34, 263–265. [Google Scholar] [CrossRef]
  81. Wada, D.A.; Wilcox, R.A.; Weenig, R.H.; Gibson, L.E. Paucity of intraepidermal FoxP3-positive T cells in cutaneous T-cell lymphoma in contrast with spongiotic and lichenoid dermatitis. J. Cutan. Pathol. 2010, 37, 535–541. [Google Scholar] [CrossRef] [PubMed]
  82. 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. JEADV 2014, 28, 1431–1435. [Google Scholar] [CrossRef] [PubMed]
  83. Berger, C.L.; Tigelaar, R.; Cohen, J.; Mariwalla, K.; Trinh, J.; Wang, N.; Edelson, R.L. Cutaneous T-cell lymphoma: Malignant proliferation of T-regulatory cells. Blood 2005, 105, 1640–1647. [Google Scholar] [CrossRef] [PubMed]
  84. Willerslev-Olsen, A.; Buus, T.B.; Nastasi, C.; Blümel, E.; Gluud, M.; Bonefeld, C.M.; Geisler, C.; Lindahl, L.M.; Vermeer, M.; Wasik, M.A.; et al. Staphylococcus aureus enterotoxins induce FOXP3 in neoplastic T cells in Sézary syndrome. Blood Cancer J. 2020, 10, 57. [Google Scholar] [CrossRef]
  85. Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct. Target. Ther. 2021, 6, 218. [Google Scholar] [CrossRef]
  86. Bhattacharjee, S.; Hamberger, F.; Ravichandra, A.; Miller, M.; Nair, A.; Affo, S.; Filliol, A.; Chin, L.; Savage, T.M.; Yin, D.; et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J. Clin. Investig. 2021, 131. [Google Scholar] [CrossRef]
  87. Mehdi, S.J.; Moerman-Herzog, A.; Wong, H.K. Normal and cancer fibroblasts differentially regulate TWIST1, TOX and cytokine gene expression in cutaneous T-cell lymphoma. BMC Cancer 2021, 21, 492. [Google Scholar] [CrossRef]
  88. Miyagaki, T.; Sugaya, M.; Suga, H.; Morimura, S.; Ohmatsu, H.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; Sato, S. Low Herpesvirus Entry Mediator (HVEM) Expression on Dermal Fibroblasts Contributes to a Th2-Dominant Microenvironment in Advanced Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2012, 132, 1280–1289. [Google Scholar] [CrossRef]
  89. Miyagaki, T.; Sugaya, M.; Fujita, H.; Ohmatsu, H.; Kakinuma, T.; Kadono, T.; Tamaki, K.; Sato, S. Eotaxins and CCR3 Interaction Regulates the Th2 Environment of Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2010, 130, 2304–2311. [Google Scholar] [CrossRef]
  90. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef]
  91. Li, M.; Fang, L.; Kwantwi, L.B.; He, G.; Luo, W.; Yang, L.; Huang, Y.; Yin, S.; Cai, Y.; Ma, W.; et al. N-Myc promotes angiogenesis and therapeutic resistance of prostate cancer by TEM8. Med. Oncol. 2021, 38, 127. [Google Scholar] [CrossRef] [PubMed]
  92. Kwantwi, L.B. The dual and multifaceted role of relaxin-2 in cancer. Clin. Transl. Oncol. 2023, 25, 2763–2771. [Google Scholar] [CrossRef] [PubMed]
  93. Karpova, M.B.; Fujii, K.; Jenni, D.; Dummer, R.; Urosevic-Maiwald, M. Evaluation of lymphangiogenic markers in Sézary syndrome. Leuk. Lymphoma 2011, 52, 491–501. [Google Scholar] [CrossRef] [PubMed]
  94. Zohdy, M.; Abd El Hafez, A.; Abd Allah, M.Y.Y.; Bessar, H.; Refat, S. Ki67 and CD31 Differential Expression in Cutaneous T-Cell Lymphoma and Its Mimickers: Association with Clinicopathological Criteria and Disease Advancement. Clin. Cosmet. Investig. Dermatol. 2020, 13, 431–442. [Google Scholar] [CrossRef]
  95. Jankowska-Konsur, A.; Kobierzycki, C.; Grzegrzolka, J.; Piotrowska, A.; Gomulkiewicz, A.; Glatzel-Plucinska, N.; Olbromski, M.; Podhorska-Okolow, M.; Szepietowski, J.C.; Dziegiel, P. Expression of CD31 in Mycosis Fungoides. Anticancer Res. 2016, 36, 4575–4582. [Google Scholar] [CrossRef]
  96. Jankowska-Konsur, A.; Kobierzycki, C.; Grzegrzółka, J.; Piotrowska, A.; Gomulkiewicz, A.; Glatzel-Plucinska, N.; Reich, A.; Podhorska-Okołów, M.; Dzięgiel, P.; Szepietowski, J.C. Podoplanin Expression Correlates with Disease Progression in Mycosis Fungoides. Acta Derm. Venereol. 2017, 97, 235–241. [Google Scholar] [CrossRef]
  97. Sakamoto, M.; Miyagaki, T.; Kamijo, H.; Oka, T.; Takahashi, N.; Suga, H.; Yoshizaki, A.; Asano, Y.; Sugaya, M.; Sato, S. Serum vascular endothelial growth factor A levels reflect itch severity in mycosis fungoides and Sézary syndrome. J. Dermatol. 2018, 45, 95–99. [Google Scholar] [CrossRef]
  98. El-Ashmawy, A.A.; Shamloula, M.M.; Elfar, N.N. Podoplanin as a Predictive Marker for Identification of High-Risk Mycosis Fungoides Patients: An Immunohistochemical Study. Indian J. Dermatol. 2020, 65, 500–505. [Google Scholar] [CrossRef]
  99. Mohammad, R.M.; Muqbil, I.; Lowe, L.; Yedjou, C.; Hsu, H.Y.; Lin, L.T.; Siegelin, M.D.; Fimognari, C.; Kumar, N.B.; Dou, Q.P.; et al. Broad targeting of resistance to apoptosis in cancer. Semin. Cancer Biol. 2015, 35, S78–S103. [Google Scholar] [CrossRef]
  100. Kim, R.; Emi, M.; Tanabe, K.; Uchida, Y.; Toge, T. The role of Fas ligand and transforming growth factor β in tumor progression. Cancer 2004, 100, 2281–2291. [Google Scholar] [CrossRef]
  101. Ni, X.; Hazarika, P.; Zhang, C.; Talpur, R.; Duvic, M. Fas ligand expression by neoplastic T lymphocytes mediates elimination of CD8+ cytotoxic T lymphocytes in mycosis fungoides: A potential mechanism of tumor immune escape? Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2001, 7, 2682–2692. [Google Scholar]
  102. Ferenczi, K.; Ohtola, J.; Aubert, P.; Kessler, M.; Sugiyama, H.; Somani, A.K.; Gilliam, A.C.; Chen, J.Z.; Yeh, I.; Matsuyama, S.; et al. Malignant T cells in cutaneous T-cell lymphoma lesions contain decreased levels of the antiapoptotic protein Ku70. Br. J. Dermatol. 2010, 163, 564–571. [Google Scholar] [CrossRef] [PubMed]
  103. da Silva Almeida, A.C.; Abate, F.; Khiabanian, H.; Martinez-Escala, E.; Guitart, J.; Tensen, C.P.; Vermeer, M.H.; Rabadan, R.; Ferrando, A.; Palomero, T. The mutational landscape of cutaneous T cell lymphoma and Sezary syndrome. Nat. Genet 2015, 47, 1465–1470. [Google Scholar] [CrossRef] [PubMed]
  104. Vieyra-Garcia, P.A.; Wei, T.; Naym, D.G.; Fredholm, S.; Fink-Puches, R.; Cerroni, L.; Odum, N.; O’Malley, J.T.; Gniadecki, R.; Wolf, P. STAT3/5-Dependent IL9 Overexpression Contributes to Neoplastic Cell Survival in Mycosis Fungoides. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 3328–3339. [Google Scholar] [CrossRef] [PubMed]
  105. Kumar, S.; Dhamija, B.; Marathe, S.; Ghosh, S.; Dwivedi, A.; Karulkar, A.; Sharma, N.; Sengar, M.; Sridhar, E.; Bonda, A.; et al. The Th9 Axis Reduces the Oxidative Stress and Promotes the Survival of Malignant T Cells in Cutaneous T-Cell Lymphoma Patients. Mol. Cancer Res. 2020, 18, 657–668. [Google Scholar] [CrossRef]
  106. Kwantwi, L.B.; Wang, S.; Sheng, Y.; Wu, Q. Multifaceted roles of CCL20 (C-C motif chemokine ligand 20): Mechanisms and communication networks in breast cancer progression. Bioengineered 2021, 12, 6923–6934. [Google Scholar] [CrossRef]
  107. Kwantwi, L.B. The dual role of autophagy in the regulation of cancer treatment. Amino Acids 2024, 56, 7. [Google Scholar] [CrossRef]
  108. Kwantwi, L.B.; Tandoh, T. Focal adhesion kinase-mediated interaction between tumor and immune cells in the tumor microenvironment: Implications for cancer-associated therapies and tumor progression. Clin. Transl. Oncol. 2024, 1–8. [Google Scholar] [CrossRef]
  109. Krejsgaard, T.; Lindahl, L.M.; Mongan, N.P.; Wasik, M.A.; Litvinov, I.V.; Iversen, L.; Langhoff, E.; Woetmann, A.; Odum, N. Malignant inflammation in cutaneous T-cell lymphoma—A hostile takeover. In Seminars in Immunopathology; Springer: Berlin/Heidelberg, Germany, 2017; pp. 269–282. [Google Scholar]
  110. Tseng, P.Y.; Hoon, M.A. Oncostatin M can sensitize sensory neurons in inflammatory pruritus. Sci. Transl. Med. 2021, 13, eabe3037. [Google Scholar] [CrossRef]
  111. 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. Off. J. Am. Assoc. Cancer Res. 2011, 17, 7529–7538. [Google Scholar] [CrossRef]
  112. Wu, X.; Hsu, D.K.; Wang, K.-H.; Huang, Y.; Mendoza, L.; Zhou, Y.; Hwang, S.T. IL-10 is overexpressed in human cutaneous T-cell lymphoma and is required for maximal tumor growth in a mouse model. Leuk. Lymphoma 2019, 60, 1244–1252. [Google Scholar] [CrossRef] [PubMed]
  113. Wilcox, R.A.; Wada, D.A.; Ziesmer, S.C.; Elsawa, S.F.; Comfere, N.I.; Dietz, A.B.; Novak, A.J.; Witzig, T.E.; Feldman, A.L.; Pittelkow, M.R. Monocytes promote tumor cell survival in T-cell lymphoproliferative disorders and are impaired in their ability to differentiate into mature dendritic cells. Blood J. Am. Soc. Hematol. 2009, 114, 2936–2944. [Google Scholar] [CrossRef] [PubMed]
  114. Ohmatsu, H.; Humme, D.; Gulati, N.; Gonzalez, J.; Möbs, M.; Suárez-Fariñas, M.; Cardinale, I.; Mitsui, H.; Guttman-Yassky, E.; Sterry, W. IL32 is progressively expressed in mycosis fungoides independent of helper T-cell 2 and helper T-cell 9 polarization. Cancer Immunol. Res. 2014, 2, 890–900. [Google Scholar] [CrossRef] [PubMed]
  115. Yu, K.K.; Smith, N.P.; Essien, S.V.; Teague, J.E.; Vieyra-Garcia, P.; Gehad, A.; Zhan, Q.; Crouch, J.D.; Gerard, N.; Larocca, C.; et al. IL-32 Supports the Survival of Malignant T Cells in Cutaneous T-cell Lymphoma. J. Investig. Dermatol. 2022, 142, 2285–2288.e2282. [Google Scholar] [CrossRef] [PubMed]
  116. Suga, H.; Sugaya, M.; Miyagaki, T.; Kawaguchi, M.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; Sato, S. The role of IL-32 in cutaneous T-cell lymphoma. J. Investig. Dermatol. 2014, 134, 1428–1435. [Google Scholar] [CrossRef]
  117. 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. 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]
  118. Santen, S.v.; Out, J.; Zoutman, W.; Quint, K.; Willemze, R.; Vermeer, M.; Tensen, C. Serum and cutaneous transcriptional expression levels of IL31 are minimal in cutaneous T cell lymphoma variants. Biochem. Biophys. Rep. 2021, 26, 101007. [Google Scholar] [CrossRef]
  119. Mishra, A.; La Perle, K.; Kwiatkowski, S.; Sullivan, L.A.; Sams, G.H.; Johns, J.; Curphey, D.P.; Wen, J.; McConnell, K.; Qi, J.; et al. Mechanism, Consequences, and Therapeutic Targeting of Abnormal IL15 Signaling in Cutaneous T-cell Lymphoma. Cancer Discov. 2016, 6, 986–1005. [Google Scholar] [CrossRef]
  120. Marzec, M.; Liu, X.; Kasprzycka, M.; Witkiewicz, A.; Raghunath, P.N.; El-Salem, M.; Robertson, E.; Odum, N.; Wasik, M.A. IL-2- and IL-15-induced activation of the rapamycin-sensitive mTORC1 pathway in malignant CD4+ T lymphocytes. Blood 2008, 111, 2181–2189. [Google Scholar] [CrossRef]
  121. Thode, C.; Woetmann, A.; Wandall, H.H.; Carlsson, M.C.; Qvortrup, K.; Kauczok, C.S.; Wobser, M.; Printzlau, A.; Ødum, N.; Dabelsteen, S. Malignant T cells secrete galectins and induce epidermal hyperproliferation and disorganized stratification in a skin model of cutaneous T-cell lymphoma. J. Investig. Dermatol. 2015, 135, 238–246. [Google Scholar] [CrossRef]
  122. 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. IL-15 and IL-17F are differentially regulated and expressed in mycosis fungoides (MF). Cell Cycle 2014, 13, 1306–1312. [Google Scholar] [CrossRef] [PubMed]
  123. Senda, N.; Miyagaki, T.; Kamijo, H.; Nakajima, R.; Oka, T.; Takahashi, N.; Suga, H.; Yoshizaki, A.; Asano, Y.; Sugaya, M.; et al. Increased HMGB1 levels in lesional skin and sera in patients with cutaneous T-cell lymphoma. Eur. J. Dermatol. 2018, 28, 621–627. [Google Scholar] [CrossRef] [PubMed]
  124. Kwantwi, L.B. Genetic alterations shape innate immune cells to foster immunosuppression and cancer immunotherapy resistance. Clin. Exp. Med. 2023, 23, 4289–4296. [Google Scholar] [CrossRef] [PubMed]
  125. Wooler, G.; Melchior, L.; Ralfkiaer, E.; Rahbek Gjerdrum, L.M.; Gniadecki, R. TP53 gene status affects survival in advanced mycosis fungoides. Front. Med. 2016, 3, 51. [Google Scholar] [CrossRef]
  126. Yu, X.; Li, H.; Zhu, M.; Hu, P.; Liu, X.; Qing, Y.; Wang, X.; Wang, H.; Wang, Z.; Xu, J.; et al. Involvement of p53 Acetylation in Growth Suppression of Cutaneous T-Cell Lymphomas Induced by HDAC Inhibition. J. Investig. Dermatol. 2020, 140, 2009–2022.e2004. [Google Scholar] [CrossRef]
  127. Wei, T.; Biskup, E.; Gjerdrum, L.M.; Niazi, O.; Ødum, N.; Gniadecki, R. Ubiquitin-specific protease 2 decreases p53-dependent apoptosis in cutaneous T-cell lymphoma. Oncotarget 2016, 7, 48391–48400. [Google Scholar] [CrossRef]
  128. Gu, X.; Wang, Y.; Zhang, C.; Liu, Y. GFI-1 overexpression promotes cell proliferation and apoptosis resistance in mycosis fungoides by repressing Bax and P21. Oncol. Lett. 2021, 22, 521. [Google Scholar] [CrossRef]
  129. Yang, H.; Ma, P.; Cao, Y.; Zhang, M.; Li, L.; Wei, J.; Tao, L.; Qian, K. ECPIRM, a Potential Therapeutic Agent for Cutaneous T-Cell Lymphoma, Inhibits Cell Proliferation and Promotes Apoptosis via a JAK/STAT Pathway. Anti-Cancer Agents Med. Chem. 2018, 18, 401–411. [Google Scholar] [CrossRef]
  130. Yanagi, T.; Nishihara, H.; Fujii, K.; Nishimura, M.; Narahira, A.; Takahashi, K.; Iwata, H.; Hata, H.; Kitamura, S.; Imafuku, K.; et al. Comprehensive cancer-related gene analysis reveals that active KRAS mutation is a prognostic mutation in mycosis fungoides. J. Dermatol. Sci. 2017, 88, 367–370. [Google Scholar] [CrossRef]
  131. Kiessling, M.K.; Oberholzer, P.A.; Mondal, C.; Karpova, M.B.; Zipser, M.C.; Lin, W.M.; Girardi, M.; Macconaill, L.E.; Kehoe, S.M.; Hatton, C.; et al. High-throughput mutation profiling of CTCL samples reveals KRAS and NRAS mutations sensitizing tumors toward inhibition of the RAS/RAF/MEK signaling cascade. Blood 2011, 117, 2433–2440. [Google Scholar] [CrossRef]
  132. Ungewickell, A.; Bhaduri, A.; Rios, E.; Reuter, J.; Lee, C.S.; Mah, A.; Zehnder, A.; Ohgami, R.; Kulkarni, S.; Armstrong, R.; et al. Genomic analysis of mycosis fungoides and Sézary syndrome identifies recurrent alterations in TNFR2. Nat. Genet. 2015, 47, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  133. Vaqué, J.P.; Gómez-López, G.; Monsálvez, V.; Varela, I.; Martínez, N.; Pérez, C.; Domínguez, O.; Graña, O.; Rodríguez-Peralto, J.L.; Rodríguez-Pinilla, S.M.; et al. PLCG1 mutations in cutaneous T-cell lymphomas. Blood 2014, 123, 2034–2043. [Google Scholar] [CrossRef] [PubMed]
  134. Chang, L.W.; Patrone, C.C.; Yang, W.; Rabionet, R.; Gallardo, F.; Espinet, B.; Sharma, M.K.; Girardi, M.; Tensen, C.P.; Vermeer, M.; et al. An Integrated Data Resource for Genomic Analysis of Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2018, 138, 2681–2683. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, L.; Ni, X.; Covington, K.R.; Yang, B.Y.; Shiu, J.; Zhang, X.; Xi, L.; Meng, Q.; Langridge, T.; Drummond, J.; et al. Genomic profiling of Sézary syndrome identifies alterations of key T cell signaling and differentiation genes. Nat. Genet. 2015, 47, 1426–1434. [Google Scholar] [CrossRef] [PubMed]
  136. 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]
  137. 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]
  138. Pinzaru, A.M.; Hom, R.A.; Beal, A.; Phillips, A.F.; Ni, E.; Cardozo, T.; Nair, N.; Choi, J.; Wuttke, D.S.; Sfeir, A.; et al. Telomere Replication Stress Induced by POT1 Inactivation Accelerates Tumorigenesis. Cell Rep. 2016, 15, 2170–2184. [Google Scholar] [CrossRef]
  139. Kantekure, K.; Yang, Y.; Raghunath, P.; Schaffer, A.; Woetmann, A.; Zhang, Q.; Odum, N.; Wasik, M. Expression patterns of the immunosuppressive proteins PD-1/CD279 and PD-L1/CD274 at different stages of cutaneous T-cell lymphoma/mycosis fungoides. Am. J. Dermatopathol. 2012, 34, 126–128. [Google Scholar] [CrossRef]
  140. Gao, Y.; Liu, F.; Sun, J.; Wen, Y.; Tu, P.; Kadin, M.E.; Wang, Y. Differential SATB1 Expression Reveals Heterogeneity of Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2021, 141, 607–618.e606. [Google Scholar] [CrossRef]
  141. Di Raimondo, C.; Rubio-Gonzalez, B.; Palmer, J.; Weisenburger, D.D.; Zain, J.; Wu, X.; Han, Z.; Rosen, S.T.; Song, J.Y.; Querfeld, C. Expression of immune checkpoint molecules programmed death protein 1, programmed death-ligand 1 and inducible T-cell co-stimulator in mycosis fungoides and Sézary syndrome: Association with disease stage and clinical outcome*. Br. J. Dermatol. 2022, 187, 234–243. [Google Scholar] [CrossRef]
  142. Han, Z.; Wu, X.; Qin, H.; Yuan, Y.C.; Zain, J.; Smith, D.L.; Akilov, O.E.; Rosen, S.T.; Feng, M.; Querfeld, C. Blockade of the Immune Checkpoint CD47 by TTI-621 Potentiates the Response to Anti-PD-L1 in Cutaneous T Cell Lymphoma. J. Investig. Dermatol. 2023, 143, 1569–1578.e5. [Google Scholar] [CrossRef] [PubMed]
  143. Saulite, I.; Ignatova, D.; Chang, Y.T.; Fassnacht, C.; Dimitriou, F.; Varypataki, E.; Anzengruber, F.; Nägeli, M.; Cozzio, A.; Dummer, R.; et al. Blockade of programmed cell death protein 1 (PD-1) in Sézary syndrome reduces Th2 phenotype of non-tumoral T lymphocytes but may enhance tumor proliferation. Oncoimmunology 2020, 9, 1738797. [Google Scholar] [CrossRef]
  144. Samimi, S.; Benoit, B.; Evans, K.; Wherry, E.J.; Showe, L.; Wysocka, M.; Rook, A.H. Increased Programmed Death-1 Expression on CD4+ T Cells in Cutaneous T-Cell Lymphoma: Implications for Immune Suppression. Arch. Dermatol. 2010, 146, 1382–1388. [Google Scholar] [CrossRef] [PubMed]
  145. Park, J.; Daniels, J.; Wartewig, T.; Ringbloom, K.G.; Martinez-Escala, M.E.; Choi, S.; Thomas, J.J.; Doukas, P.G.; Yang, J.; Snowden, C.; et al. Integrated genomic analyses of cutaneous T-cell lymphomas reveal the molecular bases for disease heterogeneity. Blood 2021, 138, 1225–1236. [Google Scholar] [CrossRef] [PubMed]
  146. Kwantwi, L.B. Exosome-mediated crosstalk between tumor cells and innate immune cells: Implications for cancer progression and therapeutic strategies. J. Cancer Res. Clin. Oncol. 2023, 149, 9487–9503. [Google Scholar] [CrossRef]
  147. Yang, E.; Wang, X.; Gong, Z.; Yu, M.; Wu, H.; Zhang, D. Exosome-mediated metabolic reprogramming: The emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct. Target. Ther. 2020, 5, 242. [Google Scholar] [CrossRef]
  148. Kwantwi, L.B. Interplay between tumor-derived factors and tumor-associated neutrophils: Opportunities for therapeutic interventions in cancer. Clin. Transl. Oncol. 2023, 25, 1963–1976. [Google Scholar] [CrossRef]
  149. Moyal, L.; Arkin, C.; Gorovitz-Haris, B.; Querfeld, C.; Rosen, S.; Knaneh, J.; Amitay-Laish, I.; Prag-Naveh, H.; Jacob-Hirsch, J.; Hodak, E. Mycosis fungoides-derived exosomes promote cell motility and are enriched with microRNA-155 and microRNA-1246, and their plasma-cell-free expression may serve as a potential biomarker for disease burden. Br. J. Dermatol. 2021, 185, 999–1012. [Google Scholar] [CrossRef]
  150. Kwantwi, L.B. Overcoming anti-PD-1/PD-L1 immune checkpoint blockade resistance: The role of macrophage, neutrophils and mast cells in the tumor microenvironment. Clin. Exp. Med. 2023, 23, 3077–3091. [Google Scholar] [CrossRef]
  151. Cheng, M.; Zain, J.; Rosen, S.T.; Querfeld, C. Emerging drugs for the treatment of cutaneous T-cell lymphoma. Expert Opin. Emerg. Drugs 2022, 27, 45–54. [Google Scholar] [CrossRef]
  152. Khodadoust, M.; Rook, A.H.; Porcu, P.; Foss, F.M.; Moskowitz, A.J.; Shustov, A.R.; Shanbhag, S.; Sokol, L.; Shine, R.; Fling, S.P. Pembrolizumab for treatment of relapsed/refractory mycosis fungoides and Sezary syndrome: Clinical efficacy in a CITN multicenter phase 2 study. Blood 2016, 128, 181. [Google Scholar] [CrossRef]
  153. Querfeld, C.; Thompson, J.A.; Taylor, M.H.; DeSimone, J.A.; Zain, J.M.; Shustov, A.R.; Johns, C.; McCann, S.; Lin, G.H.Y.; Petrova, P.S.; et al. Intralesional TTI-621, a novel biologic targeting the innate immune checkpoint CD47, in patients with relapsed or refractory mycosis fungoides or Sezary syndrome: A multicentre, phase 1 study. Lancet Haematol. 2021, 8, e808–e817. [Google Scholar] [CrossRef] [PubMed]
  154. Querfeld, C.; Chen, L.; Wu, X.; Han, Z.; Su, C.; Banez, M.; Quach, J.; Barnhizer, T.; Crisan, L.; Rosen, S.T.; et al. Preliminary Analysis Demonstrates Durvalumab (Anti-PD-L1) & Lenalidomide Is Superior to Single-Agent Durvalumab (anti-PD-L1) in Refractory/Advanced Cutaneous T Cell Lymphoma in a Randomized Phase 2 Trial. Blood 2023, 142, 3077. [Google Scholar]
  155. Wu, X.; Singh, R.; Hsu, D.K.; Zhou, Y.; Yu, S.; Han, D.; Shi, Z.; Huynh, M.; Campbell, J.J.; Hwang, S.T. A Small Molecule CCR2 Antagonist Depletes Tumor Macrophages and Synergizes with Anti-PD-1 in a Murine Model of Cutaneous T-Cell Lymphoma (CTCL). J. Investig. Dermatol. 2020, 140, 1390–1400.e1394. [Google Scholar] [CrossRef]
  156. Chen, C.; Liu, Z.; Liu, J.; Zhang, W.; Zhou, D.; Zhang, Y. Case Report: Outcome and Adverse Events of Anti-PD-1 Antibody Plus Chidamide for Relapsed/Refractory Sézary Syndrome: Case Series and A Literature Review. Front. Oncol. 2022, 12, 842123. [Google Scholar] [CrossRef]
  157. Querfeld, C.; Zain, J.M.; Wakefield, D.L.; Jovanovic-Talisman, T.; Kil, S.H.; Estephan, R.; Sanchez, J.; Palmer, J.; Rosen, S.T. Phase 1/2 Trial of Durvalumab and Lenalidomide in Patients with Cutaneous T Cell Lymphoma (CTCL): Preliminary Results of Phase I Results and Correlative Studies. Blood 2018, 132, 2931. [Google Scholar] [CrossRef]
  158. Wang, Z.; Ma, J.; Zhang, H.; Ramakrishna, R.; Mintzlaff, D.; Mathes, D.W.; Pomfret, E.A.; Lucia, M.S.; Gao, D.; Haverkos, B.M.; et al. CCR4-IL2 bispecific immunotoxin is more effective than brentuximab for targeted therapy of cutaneous T-cell lymphoma in a mouse CTCL model. FEBS Open Bio. 2023, 13, 1309–1319. [Google Scholar] [CrossRef]
  159. Querfeld, C.; William, B.M.; Sokol, L.; Akilov, O.; Poligone, B.; Zain, J.; Tagaya, Y.; Azimi, N. Co-Inhibition of il-2, il-9 and il-15 by the novel immunomodulator, bnz-1, provides clinical efficacy in patients with refractory cutaneous T cell lymphoma in a phase 1/2 clinical trial. Blood 2020, 136, 37. [Google Scholar] [CrossRef]
  160. Ni, X.; Jorgensen, J.L.; Goswami, M.; Challagundla, P.; Decker, W.K.; Kim, Y.H.; Duvic, M.A. Reduction of regulatory T cells by Mogamulizumab, a defucosylated anti-CC chemokine receptor 4 antibody, in patients with aggressive/refractory mycosis fungoides and Sézary syndrome. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 274–285. [Google Scholar] [CrossRef]
  161. Bagot, M.; Porcu, P.; Marie-Cardine, A.; Battistella, M.; William, B.M.; Vermeer, M.; Whittaker, S.; Rotolo, F.; Ram-Wolff, C.; Khodadoust, M.S.; et al. IPH4102, a first-in-class anti-KIR3DL2 monoclonal antibody, in patients with relapsed or refractory cutaneous T-cell lymphoma: An international, first-in-human, open-label, phase 1 trial. Lancet Oncol. 2019, 20, 1160–1170. [Google Scholar] [CrossRef]
Figure 1. CTCL TME negatively regulates the tumor immune microenvironment to support CTCL progression. Immune cells infiltrating the CTCL tumor microenvironment promote angiogenesis, tumor growth, migration, and immunosuppression.
Figure 1. CTCL TME negatively regulates the tumor immune microenvironment to support CTCL progression. Immune cells infiltrating the CTCL tumor microenvironment promote angiogenesis, tumor growth, migration, and immunosuppression.
Cancers 16 03368 g001
Figure 2. Role of cancer-associated fibroblasts in CTCLs. Cancer-associated fibroblasts promote CTCL migration, growth, apoptosis resistance, and immunosuppression.
Figure 2. Role of cancer-associated fibroblasts in CTCLs. Cancer-associated fibroblasts promote CTCL migration, growth, apoptosis resistance, and immunosuppression.
Cancers 16 03368 g002
Figure 3. Contribution of endothelial cells to CTCLs. Endothelial cells promote angiogenesis, tumor metastasis, growth, and apoptosis resistance.
Figure 3. Contribution of endothelial cells to CTCLs. Endothelial cells promote angiogenesis, tumor metastasis, growth, and apoptosis resistance.
Cancers 16 03368 g003
Figure 4. Molecular mechanisms of CTCL progression: CTCL TME influences the malignant transformation of CTCLs through genetic alterations, apoptosis resistance, immune checkpoint-mediated immunosuppression, cytokine dysregulations, and exosome secretions.
Figure 4. Molecular mechanisms of CTCL progression: CTCL TME influences the malignant transformation of CTCLs through genetic alterations, apoptosis resistance, immune checkpoint-mediated immunosuppression, cytokine dysregulations, and exosome secretions.
Cancers 16 03368 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kwantwi, L.B.; Rosen, S.T.; Querfeld, C. The Tumor Microenvironment as a Therapeutic Target in Cutaneous T Cell Lymphoma. Cancers 2024, 16, 3368. https://doi.org/10.3390/cancers16193368

AMA Style

Kwantwi LB, Rosen ST, Querfeld C. The Tumor Microenvironment as a Therapeutic Target in Cutaneous T Cell Lymphoma. Cancers. 2024; 16(19):3368. https://doi.org/10.3390/cancers16193368

Chicago/Turabian Style

Kwantwi, Louis Boafo, Steven T. Rosen, and Christiane Querfeld. 2024. "The Tumor Microenvironment as a Therapeutic Target in Cutaneous T Cell Lymphoma" Cancers 16, no. 19: 3368. https://doi.org/10.3390/cancers16193368

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