[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (99)

Search Parameters:
Keywords = CTCL

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 3902 KiB  
Article
Combination of JAKi and HDACi Exerts Antiangiogenic Potential in Cutaneous T-Cell Lymphoma
by Fani Karagianni, Christina Piperi, Sara Valero-Diaz, Camilla Amato, Jose Pedro Vaque, Berta Casar and Evangelia Papadavid
Cancers 2024, 16(18), 3176; https://doi.org/10.3390/cancers16183176 (registering DOI) - 17 Sep 2024
Viewed by 75
Abstract
Angiogenesis plays a pivotal role in the growth and metastasis of tumors, including the development and progression of cutaneous lymphomas. The chick embryo CAM model has been utilized in various studies to assess the growth rate, angiogenic potential, and metastatic capability of different [...] Read more.
Angiogenesis plays a pivotal role in the growth and metastasis of tumors, including the development and progression of cutaneous lymphomas. The chick embryo CAM model has been utilized in various studies to assess the growth rate, angiogenic potential, and metastatic capability of different tumor types and malignant cell lines. However, the precise mechanisms of angiogenesis in CTCL and the influence of Ruxolitinib or Resminostat on angiogenesis in hematological malignancies and solid tumors are not well understood. Recent in vitro and in vivo data have demonstrated the synergistic inhibition of tumorigenesis and metastasis in experimental models of CTCL when using the combination of Resminostat (HDACi) with Ruxolitinib (JAKi). The present work aims to elucidate the effects of this combination on the tumor microenvironment’s vascular components. We investigated the effects of Ruxolitinib (JAKi) in combination with Resminostat (HDACi) treatment in transendothelial migration of CTCL cells (106 MyLa and SeAx) by using a transwell-based transendothelial migration assay and tumor angiogenesis in vivo. We used the CTCL chick embryo CAM model with xenografted tumors derived from implanted MyLa and SeAx cells and administered topically 15 μM ruxolitinib and 5 μM Resminostat every two days during a 5-day period. JAKi and HDACi inhibited CTCL cell transendothelial migration by 75% and 82% (p < 0.05) in both CTCL engrafted cells (MyLa and SeAx, respectively) compared to the untreated group. Moreover, the combination of ruxolitinib with resminostat blocked angiogenesis by significantly reducing the number of blood vessel formation by 49% and 34% in both MyLa and SeAx, respectively (p < 0.05), indicating that the proposed combination exerted significant anti-angiogenic effects in the CAM CTCL model. Overall, these data provide valuable insights into potential therapeutic strategies targeting angiogenesis in CTCL, paving the way for more effective treatment approaches in the future. Full article
(This article belongs to the Special Issue Skin Cancers as a Paradigm Shift: From Pathobiology to Treatment)
Show Figures

Figure 1

Figure 1
<p>Ruxolitinib/Resminostat inhibited transendothelial migration in MyLa and SeAx engrafted cells in chick embryo model. Values represent mean ± standard error of the mean (SEM) for three independent experiments. Welchs’ <span class="html-italic">t</span>-test was implemented for comparison of two independent groups. One-way ANOVA analysis with Welch correction was performed for multiple comparison tests. Significance was defined as <span class="html-italic">p</span> ≤ 0.05 and denoted as: ** <span class="html-italic">p</span> &lt; 0.01 **** <span class="html-italic">p</span> &lt; 0.001, ns: not significant. Analysis was performed using GraphPad Prism 8 software (San Diego, CA, USA).</p>
Full article ">Figure 2
<p>Ruxolitinib/Resminostat blocked angiogenesis in MyLa (<b>A</b>) and SeAx (<b>B</b>) engrafted cells in chick embryo model. Representative images of (<b>A</b>) patterns of vascular branching and IKOSA blood vessels map (<b>left panel</b>) and its quantification (<b>right panel</b>) in grafted CTCL microtumors into the CAM treated as indicated. Arrows in the image indicate the effect of drugs on blood vessel density and integrity. Combination of ruxolitinib and resminostat resulted in the development of thin and collapsed blood vessels with reduced size lumens compared to control. Values represent mean ± SEM for four independent experiments, each employing 10–12 embryos per treatment variant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, ns: not significant by one-way ANOVA test.</p>
Full article ">Figure 3
<p>Expression levels of angiogenesis-related genes in MyLa/SeAx engrafted cells in chick embryo model after treatment with resminostat and/or ruxolitinib. Combination treatment decreased VEGFA (<b>A</b>) and increased VEGFB (<b>B</b>) in CTCL cells engrafted in chich embryos compared to vehicle. The bars are the means determined in three (<span class="html-italic">n</span> = 3) independent experiments using 10–12 embryos per variant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, ns: not significant by one-way ANOVA test.</p>
Full article ">Figure 4
<p>Ruxolitinib/Resminostat decreased hemoglobin levels in MyLa (<b>A</b>) and SeAx (<b>B</b>) engrafted cells in chick embryo model compared to vehicle. The bars are the means determined in three (<span class="html-italic">n</span> = 3) independent experiments using 10–12 embryos per variant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA test.</p>
Full article ">Figure 5
<p>Ruxolitinib/Resminostat decreased tumor mass in MyLa (<b>A</b>) and SeAx (<b>B</b>) engrafted cells in chick embryo model compared to vehicle. Data show mean ± SEM from three (<span class="html-italic">n</span> = 3) independent experiments, each employing 12–14 embryos per treatment variant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, **** <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA test.</p>
Full article ">Figure 6
<p><span class="html-italic">HDAC</span> gene expression and HDAC activity in MyLa (<b>A</b>) and SeAx (<b>B</b>) engrafted cells in chick embryo model. Combination of Ruxolitinib/Resminostat and resminostat alone decreased HDAC activity levels in MyLa and SeAx engrafted cells in chick embryo model. Data show mean ± SEM from three (<span class="html-italic">n</span> = 3) independent experiments, each employing 10–12 embryos per treatment variant. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005 , ns: not significant by one-way ANOVA test.</p>
Full article ">Figure 7
<p>Western blot analyses in CTCL CAM onplant microtumors for key implicated pathways. Portions of microtumors were lysed and analyzed in MyLa (A) and SeAx (B) CAM onplant microtumors for the activation levels p-AKT (Ser473), p-ERK (Tyr 204), p-STAT5 (Tyr694/699), and GADPH in . Data show mean ± SEM from three (<span class="html-italic">n</span> = 3) independent experiments. <span class="html-italic">p</span> values: * &lt; 0.05, ** &lt; 0.01, *** &lt; 0.005, **** &lt; 0.001 by one-way ANOVA test.</p>
Full article ">
10 pages, 1381 KiB  
Case Report
Mogamulizumab and Concomitant Hypofractionated Low-Dose Total Skin Electron Beam Therapy (2 × 4 Gy) in Cutaneous T-Cell Lymphoma: Proof of Principle, Report of Two Cases
by Mathias Oymanns, Michael Daum-Marzian and Chalid Assaf
Curr. Oncol. 2024, 31(9), 5412-5421; https://doi.org/10.3390/curroncol31090400 - 13 Sep 2024
Viewed by 234
Abstract
Patients with advanced-stage mycosis fungoides (MF IIB–IVB) and Sézary syndrome (SS) have poor prognoses, with survival ranging from 4.7 to 1.4 years depending on the disease stage. There is a need for therapeutic approaches that lead to long-lasting responses and improved quality of [...] Read more.
Patients with advanced-stage mycosis fungoides (MF IIB–IVB) and Sézary syndrome (SS) have poor prognoses, with survival ranging from 4.7 to 1.4 years depending on the disease stage. There is a need for therapeutic approaches that lead to long-lasting responses and improved quality of life and survival. Mogamulizumab, a humanized antibody against the CCR4 molecule, and low-dose total skin electron beam therapy (TSEBT) are two known established treatments for MF and SS as a monotherapy. However, little is known about the potential additive effect on the combination of both treatments. We report here for the first time the concurrent use of low-dose hypofractionated TSEBT (2 × 4 Gy) with mogamulizumab. Based on two relapsed/refractory and advanced-stage CTCL patients, we show that this combination may be well tolerated in advanced-stage MF or SS and may potentially lead to an additive treatment effect on response times, particularly in the skin and blood within two weeks. We propose that this combination may be a treatment option for patients with SS. Further research is needed to understand the efficacy and tolerability profile of this therapeutic combination and to determine if there is an additive effect of the combination on the response rates when compared with the monotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>Case 1: Concurrent mogamulizumab and TSEBT regimen. IV, intravenous; TSEBT, total skin electron beam therapy.</p>
Full article ">Figure 2
<p>Patient with Sézary syndrome—case 1—(<b>a</b>) before and (<b>b</b>) after 2 administrations of mogamulizumab and concurrent TSEBT at 2 Gy. TSEBT, total skin electron beam therapy.</p>
Full article ">Figure 3
<p>Case 2: Concurrent mogamulizumab and TSEBT regimen. IV, intravenous; TSEBT, total skin electron beam therapy.</p>
Full article ">Figure 4
<p>Patient 2 with Sézary syndrome (<b>a</b>) before and (<b>b</b>) after 11 administrations of mogamulizumab and 2 concurrent administrations of TSEBT at 4 Gy weekly.</p>
Full article ">
10 pages, 1172 KiB  
Review
Clinical and Histologic Variants of CD8+ Cutaneous T-Cell Lymphomas
by Madisen A. Swallow, Goran Micevic, Amanda Zhou, Kacie R. Carlson, Francine M. Foss and Michael Girardi
Cancers 2024, 16(17), 3087; https://doi.org/10.3390/cancers16173087 - 5 Sep 2024
Viewed by 353
Abstract
Although the vast majority of CTCL subtypes are of the CD4+ T-helper cell differentiation phenotype, there is a spectrum of CD8+ variants that manifest wide-ranging clinical, histologic, and phenotypic features that inform the classification of the disease. CD8, like CD4, and cytotoxic molecules [...] Read more.
Although the vast majority of CTCL subtypes are of the CD4+ T-helper cell differentiation phenotype, there is a spectrum of CD8+ variants that manifest wide-ranging clinical, histologic, and phenotypic features that inform the classification of the disease. CD8, like CD4, and cytotoxic molecules (including TIA and granzyme) are readily detectable via IHC staining of tissue and, when expressed on the phenotypically abnormal T-cell population, can help distinguish specific CTCL subtypes. Nonetheless, given that the histopathologic differential for CD8+ lymphoproliferative disorders and lymphomas may range from very indolent lymphomatoid papulosis (LyP) to aggressive entities like CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma (AECTCL), CD8 and/or cytotoxic molecule expression alone is insufficient for diagnosis and is not in itself an indicator of prognosis. We present a review of CTCL subtypes that can demonstrate CD8 positivity: CD8+ mycosis fungoides (MF), LyP type D, subcutaneous panniculitis-like T-cell lymphoma (SPTCL), primary cutaneous gamma/delta T-cell lymphoma (PCGDTL), CD8+ AECTCL, and acral CD8+ T-cell lymphoproliferative disorder (acral CD8+ TCLPD). These diseases may have different clinical manifestations and distinctive treatment algorithms. Due to the rare nature of these diseases, it is imperative to integrate clinical, histologic, and immunohistochemical findings to determine an accurate diagnosis and an appropriate treatment plan. Full article
(This article belongs to the Special Issue Cutaneous Lymphoma)
Show Figures

Figure 1

Figure 1
<p>The cutaneous T-cell lymphoma entities divided into typically CD4-positive and CD8-positive lymphomas.</p>
Full article ">Figure 2
<p>When constructing a diagnostic decision tree for CD8+ T-cell lymphoma, it is crucial to consider the key distinguishing features, particularly immunohistochemistry (IHC) differences and clinical decision points. The IHC differences, shown in the diamonds, include cytotoxic markers such as granzyme and TIA, as well as CD30 positivity. Clinical decision points, shown in rectangles, are also essential components of the decision-making process. Together, these elements help in accurately differentiating and diagnosing CD8+ T-cell lymphoma.</p>
Full article ">
13 pages, 639 KiB  
Review
Kinases Inhibitors as New Therapeutic Opportunities in Cutaneous T-Cell Lymphoma
by Sara Valero-Diaz, Camilla Amato and Berta Casar
Kinases Phosphatases 2024, 2(3), 255-267; https://doi.org/10.3390/kinasesphosphatases2030016 - 28 Aug 2024
Viewed by 368
Abstract
Cutaneous T-cell lymphomas (CTCLs) are a heterogeneous group of T-cell lymphomas characterised by high relapse rates and no curative treatments unless the allogeneic stem cell transplantation. The main complication in the management of this kind of malignancy is the variability that characterises the [...] Read more.
Cutaneous T-cell lymphomas (CTCLs) are a heterogeneous group of T-cell lymphomas characterised by high relapse rates and no curative treatments unless the allogeneic stem cell transplantation. The main complication in the management of this kind of malignancy is the variability that characterises the genetic and clinical features among the CTCL subtypes. JAK/STAT, MAPK/ERK, PI3K/Akt, and NF-kB are those signalling pathways that are found altered in CTCL and that are responsible for promoting both T-cell malignancy and the pro-tumorigenic microenvironment. Thus, targeting key players of these pathways can be an advantageous therapeutic option for CTCL. In this review, we aim to summarise the different approaches that precisely inhibit the kinases of each cited signalling. JAK inhibitors seem to be the most promising kinase inhibitors for CTCL. However, adverse events have been reported especially in patients with immunosuppression or an underlying autoimmune disease. More studies are needed, especially clinical trials, to investigate the benefits of these drugs for the treatment of cutaneous T-cell lymphomas. Full article
(This article belongs to the Special Issue Human Protein Kinases: Development of Small-Molecule Therapies)
Show Figures

Figure 1

Figure 1
<p>Kinase signalling pathways in CTCL.</p>
Full article ">
22 pages, 12037 KiB  
Article
H-1 Parvovirus-Induced Oncolysis and Tumor Microenvironment Immune Modulation in a Novel Heterotypic Spheroid Model of Cutaneous T-Cell Lymphoma
by Assia Angelova, Milena Barf, Alexandra Just, Barbara Leuchs, Jean Rommelaere and Guy Ungerechts
Cancers 2024, 16(15), 2711; https://doi.org/10.3390/cancers16152711 - 30 Jul 2024
Viewed by 991
Abstract
The rat protoparvovirus H-1 (H-1PV) is an oncolytic virus known for its anticancer properties in laboratory models of various human tumors, including non-Hodgkin lymphomas (NHL) of B-cell origin. However, H-1PV therapeutic potential against hematological malignancies of T-cell origin remains underexplored. The aim of [...] Read more.
The rat protoparvovirus H-1 (H-1PV) is an oncolytic virus known for its anticancer properties in laboratory models of various human tumors, including non-Hodgkin lymphomas (NHL) of B-cell origin. However, H-1PV therapeutic potential against hematological malignancies of T-cell origin remains underexplored. The aim of the present study was to conduct a pilot preclinical investigation of H-1PV-mediated oncolytic effects in cutaneous T-cell lymphoma (CTCL), a type of NHL that is urgently calling for innovative therapies. We demonstrated H-1PV productive infection and induction of oncolysis in both classically grown CTCL suspension cultures and in a novel, in vivo-relevant, heterotypic spheroid model, but not in healthy donor controls, including peripheral blood mononuclear cells (PBMCs). H-1PV-mediated oncolysis of CTCL cells was not prevented by Bcl-2 overexpression and was accompanied by increased extracellular ATP release. In CTCL spheroid co-cultures with PBMCs, increased spheroid infiltration with immune cells was detected upon co-culture treatment with the virus. In conclusion, our preclinical data show that H-1PV may hold significant potential as an ingenious viroimmunotherapeutic drug candidate against CTCL. Full article
(This article belongs to the Special Issue Oncolytic Viruses as an Emerging Aspect of Immune Oncology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>H-1PV entry and genomic DNA expression in CTCL cells. Positive fluorescence signals, indicative of virus P4 promoter activity, NS1 protein synthesis, and NS1-mediated virus P38 promoter activation, were detected on day three after infection with the recombinant H-1PV/EGFP virus (<b>left</b> panels, green) in all CTCL cell lines. Accordingly, H-1PV NS1 protein, the major inductor of virus-mediated oncotoxicity, was detected (<b>right</b> panels, red) in all cell lines after infection with the wt virus.</p>
Full article ">Figure 2
<p>H-1PV-mediated induction of cytotoxicity in CTCL cells. On day three after infection, significant viability suppression was observed in HH, HuT 78, and MyLa cells, while in SeAx cells, in contrast, the 50% TCID could not be reached. On day seven after infection, progression of virus-induced oncolysis was seen in HH and, of note, in SeAx cultures, in contrast to the overgrowth of HuT 78 and MyLa cell fractions, which survived infection on day three.</p>
Full article ">Figure 3
<p>H-1PV innocuousness for human primary healthy donor pan T-lymphocytes (panT) and peripheral blood mononuclear cells (PBMC). H-1PV infection of activated panT and PBMC cells failed to induce any significant toxicity, in contrast to the substantially impaired viability observed on day three in H-1PV-infected CTCL cultures (see above).</p>
Full article ">Figure 4
<p>Induction of eATP release in H-1PV-infected CTCL cultures. In HH cells, which are prone to efficient quick oncolysis, increased eATP accumulation was detected twenty-four hours after infection with high (&gt;10 PFU/cell) virus doses. In HuT 78, SeAx, and MyLa cells, which were found to display lower sensitivity to the virus, in comparison with HH, eATP secretion was induced up to seventy-two hours, and peaked forty-eight hours post infection. RLU, relative luminescence unit.</p>
Full article ">Figure 5
<p>Spheroid formation in hanging-drop CTCL cultures. (<b>a</b>) In contrast to HH and MyLa cells, which displayed intrinsic spheroid-building ability, HuT 78 and SeAx cells were only capable of two-dimensional clustering under monoculture conditions; (<b>b</b>) under co-culture conditions, keratinocytes (<b>left</b> panels) or fibroblasts (<b>right</b> panels) provided the CTCL cells with substantial 3D growth support resulting in the reproducible formation of compact, regular-shape spheroids.</p>
Full article ">Figure 6
<p>Immunofluorescence detection of cell type-specific markers in heterotypic CTCL spheroids. Immunofluorescence staining against CD4, vimentin (Vim), pancytokeratin (PCK), and CD31 (red) verified the incorporation into the heterotypic spheroid of lymphoma, fibroblast, keratinocyte, and endothelial cells, respectively.</p>
Full article ">Figure 7
<p>Heterotypic CTCL spheroid structure. (<b>a</b>) The characteristic “core-periphery” structure displayed by all types of heterotypic CTCL spheroids is illustrated by the immunofluorescence labeling of keratinocyte-containing HuT 78 spheroid sections. The typical heterotypic CTCL spheroid consisted of: (i) CTCL core (*, CD4, green), (ii) TME zone (**, pancytokeratin, red), and (iii) an invasive CTCL periphery (***, CD4, green). (<b>b</b>) In fibroblast-containing HH and HuT 78 spheroids (<b>left</b> panels), the endothelial cells (CD31, red) populated the entire TME zone. In contrast, in the keratinocyte-containing counterparts (<b>right</b> panels), the CD31+ cells were shown to cluster around the CTCL core.</p>
Full article ">Figure 8
<p>Reporter and viral gene expression in H-1PV-infected heterotypic CTCL spheroids. Reporter EGFP (green, <b>left</b> panels) and NS1 (red, <b>right</b> panels) protein expression generated positive immunofluorescence signals detected three days after spheroid infection with recombinant H-1PV/EGFP or wtH-1PV, respectively.</p>
Full article ">Figure 9
<p>H-1PV-induced suppression of heterotypic CTCL spheroid viability. Virus treatment of HH, SeAx, and MyLa spheroids led to progressive reduction in spheroid viability, in comparison with mock-treated controls, which ensued from day three to day seven. In HuT 78 spheroids, the moderate H-1PV-induced cytotoxicity detected on day three was not detectable on day seven. RLU, relative luminescence unit.</p>
Full article ">Figure 10
<p>H-1PV-induced shrinkage of heterotypic CTCL spheroids. The spheroids were treated, or not, with H-1PV and the reduction in brightfield object (BO) average area was evaluated using live-cell imaging analysis. Data obtained at 0 h (spheroid treatment and start of imaging) and 72 h are displayed and show significant shrinkage of all spheroids on day three after infection.</p>
Full article ">Figure 11
<p>H-1PV-induced cytotoxicity in heterotypic CTCL spheroids. On day three after spheroid treatment with H-1PV (n = 6 per treatment condition), virus dose-dependent increase in the uptake of the dead cell tracker IncuCyte<sup>®</sup> Cytotox Red Dye, in association with significant spheroid shrinkage, was observed.</p>
Full article ">Figure 12
<p>Spheroid infiltration with CD8+ cells in co-cultures of keratinocyte-containing MyLa spheroids and PBMCs. H-1PV treatment (<b>right</b> panel) of the co-culture enhanced the spontaneous (<b>left</b> panel) spheroid infiltration with cytotoxic T-lymphocytes (CD8, red). Mean CD8+ cell counts per 500 cells were compared between H-1PV-infected and mock-treated co-cultures.</p>
Full article ">Figure 13
<p>The multifaceted role of extracellular ATP in CTCL biology. CTCL cells release cytosolic ATP (cytATP) into the extracellular space (1). Extracellular ATP (eATP) may activate cellular purinergic P2X7 receptors (2), leading to Ca<sup>2+</sup> influx, NFAT activation, IL-2 transcription (3), and, ultimately, CTCL cell growth and proliferation (4). On the other hand, H-1PV infection of CTCL cells (5) may additionally trigger eATP secretion, resulting in the induction of danger signaling in the CTCL TME (7).</p>
Full article ">Figure A1
<p>Improved SeAx cell killing by combined H-1PV infection and Bcl-2 inhibition. Venetoclax (VCL, 100 nM)-mediated Bcl-2 inhibition combined with H-1PV infection (10 PFU/cell) of Bcl-2-overexpressing SeAx cells led to increased suppression of cell viability on both day three and day seven, in comparison with either virus or VCL treatment alone.</p>
Full article ">Figure A2
<p>Surface P2X7 purinergic receptor and CD73 ectonucleotidase expression in H-1PV-infected (<b>right</b> panels) versus mock-infected (<b>left</b> panels) CTCL cells. In HH and MyLa cells, both P2X7 upregulation and CD73 downregulation was induced by H-1PV infection. In contrast, marked CD73 upregulation was observed in infected HuT 78 cells.</p>
Full article ">
12 pages, 1837 KiB  
Article
Application of Photodynamic Therapy with 5-Aminolevulinic Acid to Extracorporeal Photopheresis in the Treatment of Cutaneous T-Cell Lymphoma: A First-in-Human Phase I/II Study
by Eidi Christensen, Olav Andreas Foss, Toril Holien, Petras Juzenas and Qian Peng
Pharmaceutics 2024, 16(6), 815; https://doi.org/10.3390/pharmaceutics16060815 - 16 Jun 2024
Viewed by 880
Abstract
Extracorporeal photopheresis (ECP) is a therapeutic modality used for T-cell-mediated disorders. This approach involves exposing isolated white blood cells to photoactivatable 8-methoxypsoralen (8-MOP) and UVA light, aiming to induce apoptosis in T-cells and thereby modulate immune responses. However, conventional 8-MOP-ECP lacks cell selectivity, [...] Read more.
Extracorporeal photopheresis (ECP) is a therapeutic modality used for T-cell-mediated disorders. This approach involves exposing isolated white blood cells to photoactivatable 8-methoxypsoralen (8-MOP) and UVA light, aiming to induce apoptosis in T-cells and thereby modulate immune responses. However, conventional 8-MOP-ECP lacks cell selectivity, killing both healthy and diseased cells, and has shown limited treatment efficacy. An alternative approach under investigation involves the use of 5-aminolevulinic acid (ALA) in conjunction with light, referred to as ALA-based photodynamic therapy. Our previous ex vivo studies suggest that ALA-ECP exhibits greater selectivity and efficiency in killing T-cells derived from patients with T-cell-mediated disorders compared to those treated with 8-MOP-ECP. We have conducted a clinical phase I–(II) study evaluating ALA-ECP safety and tolerability in cutaneous T-cell lymphoma (CTCL). Here, 20 ALA-ECP treatments were administered to one CTCL patient, revealing no significant changes in vital signs. Two adverse events were reported; both evaluated by the Internal Safety Review Committee as non-serious. In addition, five conceivable events with mainly mild symptoms took place. During the study period, a 53% reduction in skin involvement and a 50% reduction in pruritus was observed. In conclusion, the results indicate that ALA-ECP treatment is safe and well tolerated. Full article
(This article belongs to the Special Issue Photodynamic Therapy: Rising Star in Pharmaceutical Applications)
Show Figures

Figure 1

Figure 1
<p>Images of the anterior trunk taken (<b>A</b>) before and (<b>B</b>) after 20 ALA-ECP treatments, depicting multiple areas with erythema, patches, and plaques before treatment, and paler skin indicative of clinical remission on the upper chest and upper arms, along with predominantly erythematous patches on the abdomen.</p>
Full article ">Figure 2
<p>Images with extracts from the patient’s back taken (<b>A</b>) before and (<b>B</b>) after 20 ALA-ECP treatments, depicting patches and plaques before treatment and predominantly patches after treatment.</p>
Full article ">Figure 3
<p>Images displaying the front of the lower extremities taken (<b>A</b>) before and (<b>B</b>) after 20 ALA-ECP treatments, with predominantly plaque localized to the thighs before treatment and with mainly macular erythema after treatment.</p>
Full article ">Figure 4
<p>Images of the back of the lower extremities taken (<b>A</b>) before and (<b>B</b>) after 20 ALA-ECP treatments, with areas of erythema and plaque localized proximally and mid-extremity before and predominantly erythema proximally and with mild pigmentation mid-extremity, particularly on the left extremity after treatment.</p>
Full article ">
13 pages, 8790 KiB  
Review
Delving into the Metabolism of Sézary Cells: A Brief Review
by Carel Cherfan, Alain Chebly, Hamid Reza Rezvani, Marie Beylot-Barry and Edith Chevret
Genes 2024, 15(5), 635; https://doi.org/10.3390/genes15050635 - 17 May 2024
Viewed by 1037
Abstract
Primary cutaneous lymphomas (PCLs) are a heterogeneous group of lymphoproliferative disorders caused by the accumulation of neoplastic T or B lymphocytes in the skin. Sézary syndrome (SS) is an aggressive and rare form of cutaneous T cell lymphoma (CTCL) characterized by an erythroderma [...] Read more.
Primary cutaneous lymphomas (PCLs) are a heterogeneous group of lymphoproliferative disorders caused by the accumulation of neoplastic T or B lymphocytes in the skin. Sézary syndrome (SS) is an aggressive and rare form of cutaneous T cell lymphoma (CTCL) characterized by an erythroderma and the presence of atypical cerebriform T cells named Sézary cells in skin and blood. Most of the available treatments for SS are not curative, which means there is an urgent need for the development of novel efficient therapies. Recently, targeting cancer metabolism has emerged as a promising strategy for cancer therapy. This is due to the accumulating evidence that metabolic reprogramming highly contributes to tumor progression. Genes play a pivotal role in regulating metabolic processes, and alterations in these genes can disrupt the delicate balance of metabolic pathways, potentially contributing to cancer development. In this review, we discuss the importance of targeting energy metabolism in tumors and the currently available data on the metabolism of Sézary cells, paving the way for potential new therapeutic approaches aiming to improve clinical outcomes for patients suffering from SS. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Figure 1
<p>Glycolysis and its inhibitors. Glucose transporter (GLUT); Hexokinase (HK); Phosphoglucose isomerase (PGI); 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase3 (PFKFB3); Phosphofructokinase 1 (PFK1); Tripsephosphate isomerase (TP1); Glyceraldehyde phosphate dehydrogenase (GAPDH); Phosphoglycerate Kinase (PGK); Phosphoglycerate mutase (PGAM); Pyruvate Kinase M2 (PKM2); Lactate dehydrogenase A (LDHA); Benitrobenrazide (BNBZ).</p>
Full article ">Figure 2
<p>Mitochondrial targets for cancer therapy. Pyruvate dehydrogenase complex (PDC); Pyruvate dehydrogenase kinases (PDKs); Dichloroacetate (DCA); isocitrate dehydrogenase (IDH); Glutaminase (GLS); Glutamate dehydrogenase (GDH); Complexes I, II, III, IV; Coenzyme Q (CoQ); Cytochrome c (Cyt c).</p>
Full article ">
19 pages, 771 KiB  
Review
A Narrative Review of the State of the Art of CCR4-Based Therapies in Cutaneous T-Cell Lymphomas: Focus on Mogamulizumab and Future Treatments
by Corrado Zengarini, Alba Guglielmo, Martina Mussi, Giovanna Motta, Claudio Agostinelli, Elena Sabattini, Bianca Maria Piraccini and Alessandro Pileri
Antibodies 2024, 13(2), 32; https://doi.org/10.3390/antib13020032 - 22 Apr 2024
Viewed by 1768
Abstract
The CCR4 receptor is a pivotal target in cutaneous T-cell lymphoma (CTCL) therapy due to its role in impairing immune responses against malignant T-cells and expression profiles. Monoclonal antibodies like mogamulizumab effectively bind to CCR4, reducing tumour burden and enhancing patient outcomes by [...] Read more.
The CCR4 receptor is a pivotal target in cutaneous T-cell lymphoma (CTCL) therapy due to its role in impairing immune responses against malignant T-cells and expression profiles. Monoclonal antibodies like mogamulizumab effectively bind to CCR4, reducing tumour burden and enhancing patient outcomes by inhibiting the receptor’s interaction with ligands, thereby hindering malignant T-cell migration and survival. Combining CCR4 antibodies with chemotherapy, radiation, and other drugs is being explored for synergistic effects. Additionally, small-molecular inhibitors, old pharmacological agents interacting with CCR4, and CAR-T therapies are under investigation. Challenges include drug resistance, off-target effects, and patient selection, addressed through ongoing trials refining protocols and identifying biomarkers. Despite advancements, real-life data for most of the emerging treatments are needed to temper expectations. In conclusion, CCR4-targeted therapies show promise for CTCL management, but challenges persist. Continued research aims to optimise treatments, enhance outcomes, and transform CTCL management. This review aims to elucidate the biological rationale and the several agents under various stages of development and clinical evaluation with the actual known data. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic representation of the molecular pathways activated by CCR4.</p>
Full article ">
35 pages, 836 KiB  
Review
The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets
by Alba Guglielmo, Corrado Zengarini, Claudio Agostinelli, Giovanna Motta, Elena Sabattini and Alessandro Pileri
Cells 2024, 13(7), 584; https://doi.org/10.3390/cells13070584 - 28 Mar 2024
Cited by 1 | Viewed by 1633
Abstract
Cutaneous T cell lymphomas (CTCLs), encompassing mycosis fungoides (MF) and Sézary syndrome (SS), present a complex landscape influenced by cytokines and cellular responses. In this work, the intricate relationship between these inflammatory proteins and disease pathogenesis is examined, focusing on what is known [...] Read more.
Cutaneous T cell lymphomas (CTCLs), encompassing mycosis fungoides (MF) and Sézary syndrome (SS), present a complex landscape influenced by cytokines and cellular responses. In this work, the intricate relationship between these inflammatory proteins and disease pathogenesis is examined, focusing on what is known at the clinical and therapeutic levels regarding the most well-known inflammatory mediators. An in-depth look is given to their possible alterations caused by novel immunomodulatory drugs and how they may alter disease progression. From this narrative review of the actual scientific landscape, Interferon-gamma (IFN-γ) emerges as a central player, demonstrating a dual role in both promoting and inhibiting cancer immunity, but the work navigates through all the major interleukins known in inflammatory environments. Immunotherapeutic perspectives are elucidated, highlighting the crucial role of the cutaneous microenvironment in shaping dysfunctional cell trafficking, antitumor immunity, and angiogenesis in MF, showcasing advancements in understanding and targeting the immune phenotype in CTCL. In summary, this manuscript aims to comprehensively explore the multifaceted aspects of CTCL, from the immunopathogenesis and cytokine dynamics centred around TNF-α and IFN-γ to evolving therapeutic modalities. Including all the major known and studied cytokines in this analysis broadens our understanding of the intricate interplay influencing CTCL, paving the way for improved management of this complex lymphoma. Full article
Show Figures

Figure 1

Figure 1
<p>Potential and actual therapies to address CTCLs and their relative targets.</p>
Full article ">
16 pages, 1041 KiB  
Review
Vitamin D in Cutaneous T-Cell Lymphoma
by August-Witte Feentved Ødum and Carsten Geisler
Cells 2024, 13(6), 503; https://doi.org/10.3390/cells13060503 - 13 Mar 2024
Cited by 2 | Viewed by 1866
Abstract
Cutaneous T-cell lymphoma (CTCL) is characterized by the proliferation of malignant T cells in inflamed skin lesions. Mycosis fungoides (MF)—the most common variant of CTCL—often presents with skin lesions around the abdomen and buttocks (“bathing suit” distribution), i.e., in skin areas devoid of [...] Read more.
Cutaneous T-cell lymphoma (CTCL) is characterized by the proliferation of malignant T cells in inflamed skin lesions. Mycosis fungoides (MF)—the most common variant of CTCL—often presents with skin lesions around the abdomen and buttocks (“bathing suit” distribution), i.e., in skin areas devoid of sun-induced vitamin D. For decades, sunlight and vitamin D have been connected to CTCL. Thus, vitamin D induces apoptosis and inhibits the expression of cytokines in malignant T cells. Furthermore, CTCL patients often display vitamin D deficiency, whereas phototherapy induces vitamin D and has beneficial effects in CTCL, suggesting that light and vitamin D have beneficial/protective effects in CTCL. Inversely, vitamin D promotes T helper 2 (Th2) cell specific cytokine production, regulatory T cells, tolerogenic dendritic cells, as well as the expression of immune checkpoint molecules, all of which may have disease-promoting effects by stimulating malignant T-cell proliferation and inhibiting anticancer immunity. Studies on vitamin D treatment in CTCL patients showed conflicting results. Some studies found positive effects, others negative effects, while the largest study showed no apparent clinical effect. Taken together, vitamin D may have both pro- and anticancer effects in CTCL. The balance between the opposing effects of vitamin D in CTCL is likely influenced by treatment and may change during the disease course. Therefore, it remains to be discovered whether and how the effect of vitamin D can be tilted toward an anticancer response in CTCL. Full article
Show Figures

Figure 1

Figure 1
<p>Simplified overview of the vitamin D/VDR signaling pathway. PM, plasma membrane; VDR, vitamin D receptor; RXR, retinoid X receptor; VDRE, vitamin D response element.</p>
Full article ">Figure 2
<p>The many anti- and pro-cancer roles of vitamin D in CTCL. IL, interleukin; Th2, T helper 2 cell; DC, dendritic cell; PD-L1, programmed death ligand-1; Treg, regulatory T cell.</p>
Full article ">
9 pages, 241 KiB  
Article
New Genetic Markers of Skin T-Cell Lymphoma Treatment
by Vladimír Vašků, Petra Fialová and Anna Vašků
Genes 2024, 15(3), 358; https://doi.org/10.3390/genes15030358 - 13 Mar 2024
Viewed by 1319
Abstract
Aim: Cutaneous T-cell lymphomas (CTCL) can be described as chronic skin inflammation lesions with the content of malignant T cells and they are considered to be T-cell-mediated skin diseases. CD147 is recognized as a 58-kDa cell surface glycoprotein of the immunoglobulin superfamily; it [...] Read more.
Aim: Cutaneous T-cell lymphomas (CTCL) can be described as chronic skin inflammation lesions with the content of malignant T cells and they are considered to be T-cell-mediated skin diseases. CD147 is recognized as a 58-kDa cell surface glycoprotein of the immunoglobulin superfamily; it can induce the synthesis of MMPs (matrix metalloproteinases) on the surface of tumor cells where it was originally identified. It can also function in adjacent tumor fibroblasts using CD147–CD147 interactions. The polymorphism rs8259 T/A is situated in the untranslated region (3′UTR) of the CD147 gene. HLA DRB1*1501 takes part in the process of presentation and recognition of different antigens to T cells. It can be expressed by antigen-presenting cells—macrophages, dendritic cells, and B cells. The aim of the study is to test genotype–phenotype associations of both polymorphisms including therapy in a large cohort of CTCL patients. Materials and Methods: A final total of 104 CTCL patients were enrolled in the study. For the first remission at the clinic department, they were treated by means of local skin-directed therapy, phototherapy, and systemic therapy. Genomic DNA was isolated from peripheral blood leukocytes. A standard technique using proteinase K was applied. The polymorphisms rs8259 T/A (CD147 gene) and rs3135388 (HLA DRB1*1501) were detected through standard PCR-restriction fragment length polymorphism methods. Results: The severity of the disease (patients with parapsoriasis, stages IA and IB, vs patients with stages IIB, IIIA, and IIIB) was associated with the CD147 genotype: the AA variant was 3.38 times more frequent in more severe cases, which reflects the decision on systemic therapy (p = 0.02, specificity 0.965). The AA genotype in the CD147 polymorphism was 12 times more frequent in patients who underwent systemic therapy of CTCL compared to those not treated with this therapy (p = 0.009, specificity 0.976). The same genotype was also associated with radiotherapy—it was observed 14 times more frequently in patients treated with radiotherapy (p = 0.009, specificity 0.959). In patients treated with interferon α therapy, the AA genotype was observed to be 5.85 times more frequent compared to the patients not treated with interferon therapy (p = 0.03, specificity 0.963). The HLA DRB1*1501 polymorphism was associated with local skin-directed therapy of CTCL. The CC genotype of the polymorphism was observed to be 3.57 times more frequent in patients treated with local therapy (p = 0.008, specificity 0.948). When both polymorphisms had been calculated together, even better results were obtained: the AACC double genotype was 11 times more frequent in patients with severe CTCL (p = 0.009, specificity 0.977). The TACT double genotype was associated with local skin-directed therapy (0.09 times lower frequency, p = 0.007, sensitivity 0.982). The AACC genotype was 8.9 times more frequent in patients treated by means of systemic therapy (p = 0.02, specificity 0.976) and as many as 18.8 times more frequent in patients treated with radiotherapy (p = 0.005, specificity 0.969). Thus, the AACC double genotype of CD147 and DRB1*1501 polymorphisms seems to be a clinically highly specific marker of severity, systemic therapy and radiotherapy of patients with T-cell lymphoma. Conclusion: Although genotyping results were not known during the treatment decision and could not modify it, the clinical decision on severity and therapy reflected some aspects of the genetic background of this complicated T-cell-associated disease very well. Full article
15 pages, 1368 KiB  
Systematic Review
JAK Inhibitors in Cutaneous T-Cell Lymphoma: Friend or Foe? A Systematic Review of the Published Literature
by Seyed Mohammad Vahabi, Saeed Bahramian, Farzad Esmaeili, Bardia Danaei, Yasamin Kalantari, Patrick Fazeli, Sara Sadeghi, Nima Hajizadeh, Chalid Assaf and Ifa Etesami
Cancers 2024, 16(5), 861; https://doi.org/10.3390/cancers16050861 - 21 Feb 2024
Cited by 1 | Viewed by 2262
Abstract
Cutaneous T-cell lymphomas (CTCLs) are a group of lymphoid neoplasms with high relapse rates and no curative treatment other than allogeneic stem cell transplantation (allo-SCT). CTCL is significantly influenced by disruption of JAK/STAT signaling. Therefore, Janus kinase (JAK) inhibitors may be promising for [...] Read more.
Cutaneous T-cell lymphomas (CTCLs) are a group of lymphoid neoplasms with high relapse rates and no curative treatment other than allogeneic stem cell transplantation (allo-SCT). CTCL is significantly influenced by disruption of JAK/STAT signaling. Therefore, Janus kinase (JAK) inhibitors may be promising for CTCL treatment. This study is a systematic review aiming to investigate the role of JAK inhibitors in the treatment of CTCL, including their efficacy and safety. Out of 438 initially searched articles, we present 13 eligible ones. The overall response rate (ORR) in the treatment with JAK inhibitors in clinical trials was 11–35%, although different subtypes of CTCL showed different ORRs. Mycosis fungoides showed an ORR of 14–45%, while subcutaneous-panniculitis-like T-cell lymphoma (SPTCL) displayed an ORR ranging from 75% to 100%. Five cases were reported having a relapse/incident of CTCL after using JAK inhibitors; of these, three cases were de novo CTCLs in patients under treatment with a JAK inhibitor due to refractory arthritis, and two cases were relapsed disease after graft-versus-host disease treatment following allo-SCT. In conclusion, using JAK inhibitors for CTCL treatment seems promising with acceptable side effects, especially in patients with SPTCL. Some biomarkers, like pS6, showed an association with better responses. Caution should be taken when treating patients with an underlying autoimmune disease and prior immunosuppression. Full article
(This article belongs to the Special Issue Targets and Biomarkers in Cutaneous Lymphoma)
Show Figures

Figure 1

Figure 1
<p>Disruptions in the JAK/STAT pathway are linked to various subtypes of CTCL, each mostly impacted by distinct genetic mutations. The pathway becomes active when cytokines engage their respective cell surface receptors, triggering the activation of JAKs. In CTCL, the mutation spectrum varies across subtypes: for MF and SS, JAK1 and JAK3 exhibit single-nucleotide variants (SNVs), while STAT3 and STAT5B are similarly affected by SNVs. In SPTCL, JAK2 is particularly notable for SNVs. Primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma (pcAECyTCL) does not show JAK mutations but is characterized by SNVs in JAK2, JAK3, STAT3, and STAT5B. Additionally, increased expression of these STAT proteins is frequently correlated with copy number gains, particularly in JAK2 for MF/SS and in both STAT3 and STAT5B across the subtypes mentioned. Copy number variations (CNVs) and structural alterations such as fusions in these genes can lead to enhanced or constitutive activation of the pathway, driving the progression of CTCL. Moreover, the negative regulatory roles of SOCS1 and SH2B3 (LNK) are highlighted, where CNVs or loss-of-function mutations can lead to unregulated JAK/STAT pathway activity, emphasizing their potential as therapeutic targets. For instance, loss-of-function mutations and deletions in SOCS1 could contribute to the aberrant activation of the JAK/STAT pathway in CTCL subtypes. The detailed mutation profile for each subtype underscores the complexity and the necessity for subtype-specific therapeutic approaches, including the use of JAK inhibitors to correct the dysregulated signaling in these lymphomas. “P” stands for phosphate; triangles and squares stand for cytokines, chemokines and other proteins. Abbreviations: IFN-γ, interferon gamma; IL, interleukin; SOCS1, suppressor of cytokine signaling-1; LNK, lymphocyte adapter protein; JAK, Janus kinase; TYK2, tyrosine kinase-2; STAT, signal transducer and activator of transcription.</p>
Full article ">Figure 2
<p>Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Flow chart of the number of studies identified and selected into the systematic review and meta-analysis.</p>
Full article ">
11 pages, 273 KiB  
Review
The Complex Role of Infectious Agents in Human Cutaneous T-Cell Lymphoma Pathogenesis: From Candidate Etiological Factors to Potential Therapeutics
by Assia Angelova, Jean Rommelaere and Guy Ungerechts
Pathogens 2024, 13(3), 184; https://doi.org/10.3390/pathogens13030184 - 20 Feb 2024
Cited by 1 | Viewed by 1620
Abstract
Cutaneous T-cell lymphoma (CTCL) is a devastating, potentially fatal T-lymphocyte malignancy affecting the skin. Despite all efforts, the etiology of this disease remains unknown. Infectious agents have long been suspected as factors or co-factors in CTCL pathogenesis. This review deals with the panel [...] Read more.
Cutaneous T-cell lymphoma (CTCL) is a devastating, potentially fatal T-lymphocyte malignancy affecting the skin. Despite all efforts, the etiology of this disease remains unknown. Infectious agents have long been suspected as factors or co-factors in CTCL pathogenesis. This review deals with the panel of bacterial and viral pathogens that have been investigated so far in an attempt to establish a potential link between infection/carriage and CTCL development. A special focus is given to a recently discovered human protoparvovirus, namely the cutavirus (CutaV), which has emerged as a plausible CTCL etiological agent. Available evidence in support of this hypothesis as well as alternative interpretations and uncertainties raised by some conflicting data are discussed. The complexity and multifacetedness of the Parvoviridae family of viruses are illustrated by presenting another protoparvovirus, the rat H-1 parvovirus (H-1PV). H-1PV belongs to the same genus as the CutaV but carries considerable potential for therapeutic applications in cutaneous lymphoma. Full article
(This article belongs to the Section Vaccines and Therapeutic Developments)
21 pages, 3425 KiB  
Review
Clinical and Real-World Effectiveness of Mogamulizumab: A Narrative Review
by Montserrat Fernández-Guarino, Pablo Ortiz, Fernando Gallardo and Mar Llamas-Velasco
Int. J. Mol. Sci. 2024, 25(4), 2203; https://doi.org/10.3390/ijms25042203 - 12 Feb 2024
Cited by 1 | Viewed by 2415
Abstract
Mogamulizumab (MOG) is an antibody targeting the CCR4 receptor, authorized for relapsed or refractory peripheral T-cell (PTCL) and cutaneous T-cell lymphomas (CTCL). Its adoption in guidelines and endorsement by FDA and EMA established it as a systemic treatment, especially for advanced disease stages [...] Read more.
Mogamulizumab (MOG) is an antibody targeting the CCR4 receptor, authorized for relapsed or refractory peripheral T-cell (PTCL) and cutaneous T-cell lymphomas (CTCL). Its adoption in guidelines and endorsement by FDA and EMA established it as a systemic treatment, especially for advanced disease stages due to its comparatively lower toxicity. Clinical trials and real-world evidence have underscored its efficacy in advanced CTCLs, including mycosis fungoides and Sézary syndrome; PTCLs; and adult T-cell leukemia/lymphoma (ATLL), showcasing positive outcomes. Notably, the drug has demonstrated significant response rates, disease stability, and extended periods of progression-free survival, suggesting its applicability in cases with multiple treatment lines. Its safety profile is generally manageable, with adverse events (AEs) primarily related to the skin, infusion-related reactions, drug eruptions, autoimmune diseases, and skin disorders. The latter seem to appear as CCR4 can promote the skin-specific homing of lymphocytes, and MOG is directed against this receptor. While combination with immunostimulatory agents like interferon alpha and interleukin 12 has shown promising results, caution is urged when combining with PD1 inhibitors due to the heightened risk of immune-mediated AEs. The introduction of MOG as a systemic treatment implies a significant advancement in managing these diseases, supported by its favorable safety profile and complementary mechanisms. Full article
(This article belongs to the Special Issue Dermatology: Advances in Pathophysiology and Therapies (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Representative figure of mogamulizumab’s main mechanism of action by antibody-dependent cell-mediated cytotoxicity.</p>
Full article ">Figure 2
<p>Schematic summary of the guidelines for treatment of mycosis fungoides and Sezary syndrome. * meclorethamine is not available in every country.</p>
Full article ">Figure 2 Cont.
<p>Schematic summary of the guidelines for treatment of mycosis fungoides and Sezary syndrome. * meclorethamine is not available in every country.</p>
Full article ">Figure 3
<p>Morbiliform rash in the back of a patient during MOG treatment.</p>
Full article ">Figure 4
<p>Patient treated with MOG who developed lichenoid reactions that recurred several times during the follow-up.</p>
Full article ">Figure 5
<p>Patient treated with MOG who developed facial vitiligo months after the start of the treatment 2 years ago.</p>
Full article ">
12 pages, 471 KiB  
Review
Role of IL-4 and IL-13 in Cutaneous T Cell Lymphoma
by Roberto Mazzetto, Paola Miceli, Jacopo Tartaglia, Christian Ciolfi, Alvise Sernicola and Mauro Alaibac
Life 2024, 14(2), 245; https://doi.org/10.3390/life14020245 - 9 Feb 2024
Cited by 2 | Viewed by 2051
Abstract
The interleukins IL-4 and IL-13 are increasingly recognized contributors to the pathogenesis of cutaneous T cell lymphomas (CTCLs), and their role in disease-associated pruritus is accepted. The prevailing Th2 profile in advanced CTCL underscores the significance of understanding IL-4/IL-13 expression dynamics from the [...] Read more.
The interleukins IL-4 and IL-13 are increasingly recognized contributors to the pathogenesis of cutaneous T cell lymphomas (CTCLs), and their role in disease-associated pruritus is accepted. The prevailing Th2 profile in advanced CTCL underscores the significance of understanding IL-4/IL-13 expression dynamics from the early stages of disease, as a shift from Th1 to Th2 may explain CTCL progression. Targeted agents blocking key cytokines of type 2 immunity are established therapeutics in atopic disorders and have a promising therapeutic potential in CTCL, given their involvement in cutaneous symptoms and their contribution to the pathogenesis of disease. IL-4, IL-13, and IL-31 are implicated in pruritus, offering therapeutic targets with dupilumab, tralokinumab, lebrikizumab, and nemolizumab. This review analyzes current knowledge on the IL-4/IL-13 axis in mycosis fungoides and Sezary syndrome, the most common types of CTCL, examining existing literature on the pathogenetic implications with a focus on investigational treatments. Clinical trials and case reports are required to shed light on novel uses of medications in various diseases, and ongoing research into the role of IL-4/IL-13 axis blockers in CTCL therapy might not only improve the management of disease-related pruritus but also provide in-depth insights on the pathophysiologic mechanisms of CTCL. Full article
(This article belongs to the Special Issue Skin Cancer: From Molecular Basis to Therapy)
Show Figures

Figure 1

Figure 1
<p>Surface receptors related to type 2 inflammatory pathways expressed on malignant lymphocytes of T cell cutaneous lymphoma. Abbreviations: CCL17, C-C motif chemokine ligand 17; CCR4, C-C chemokine receptor type 4; CLA, cutaneous lymphocyte antigen; IL-4Ra, IL-4 receptor alpha; IL-13Ra1, IL-13 receptor alpha 1; IL-31RA, IL-31 receptor A; OSMRB, oncostatin M receptor beta; OX40, T cell co-stimulatory receptor CD134; TSLP and TSLPR, thymic stromal lymphopoietin and its receptor.</p>
Full article ">
Back to TopTop