Ta strona używa pliki cookies, w celu polepszenia użyteczności i funkcjonalności oraz w celach statystycznych. Dowiedz się więcej w Polityce prywatności.
Korzystając ze strony wyrażasz zgodę na używanie plików cookies, zgodnie z aktualnymi ustawieniami przeglądarki.
Akceptuję wykorzystanie plików cookies
Advances in Dermatology and Allergology
eISSN: 2299-0046
ISSN: 1642-395X
Advances in Dermatology and Allergology/Postępy Dermatologii i Alergologii
Current Issue Archive Manuscripts accepted About the journal Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2025
vol. 42
 
Share:
Share:
Review paper

Classical and biological treatments in mycosis fungoides/Sézary syndrome. New horizons in oncodermatology

Hanna R. Cisoń
1
,
Rafał K. Białynicki-Birula
1

  1. Department and Clinic of Dermatology, Venereology and Allergology, Medical University of Wroclaw, Poland
Adv Dermatol Allergol 2025; XLII (2): 125-133
Online publish date: 2024/11/14
Article file
- Classical.pdf  [0.17 MB]
Get citation
 
 

Introduction

Cutaneous T-cell lymphoma (CTCL) is a heterogeneous group of malignancies primarily affecting skin-tropic lymphocytes nodal non-Hodgkin lymphoma (NHL), predominantly B-cell-derived, approximately 75% of primary cutaneous lymphomas have a T-cell origin. Of these T-cell lymphomas, two-thirds can be categorized as mycosis fungoides (MF) or Sézary syndrome (SS), therefore, our review focuses on this subgroup [1].

Methods

A database search of PubMed was conducted using the medical subject headings (MeSH) terms “lymphoma, T-cell, cutaneous/diagnosis”, “lymphoma, T-cell, cutaneous/diagnostic imaging”, “lymphoma, T-cell, cutaneous/drug therapy”, “lymphoma, T-cell, cutaneous/therapy”. To narrow down the search, filters were applied such as “full text” and “humans”. Additional keywords used to refine the search included the names of specific biological treatments and related terms such as “immunotherapy”. An advanced search was also carried out on ClinicalTrials.gov using the terms “MF/SS” and “biological treatment”.

Results

Outlined below are the therapeutic options, including both topical and systemic agents, with their use determined by the disease staging. The first section details approved monotherapies and combination therapies with the most frequent side effects which are collected in Table 1 [24] while the subsequent section discusses investigational treatments currently under research.

Table 1

Standard stage-based treatment regimens for MF/SS according to MF regarding staging WHO sorted alphabetically (only registered) with the most common side effects [24]

Disease stageTreatment (alphabetical order)The most common side effects
Patch-plaque MF (stage I–IIA)Bexarotene 1% gelDermatitis, rash, swelling
Bexarotene p.o.Leukopenia, hypothyroidism, hyperlipidaemia
Bexarotene p.o. and MTXLeukopenia, hypothyroidism, hyperlipidaemia and myelotoxicity, hepatotoxicity, nephrotoxicity
Brentuximab vedotinNeuropathy, allergic reactions, shortness of breath, redness or swelling of the face
HDAC inhibitorDiarrhea, infections, low blood cell count
IFN-αAnemia, neutropenia, hepatotoxicity
IFN-α and MTXAnemia, neutropenia, hepatotoxicity and myelotoxicity, hepatotoxicity, nephrotoxicity
Local RTSkin redness and irritation (erythema), fatigue, skin peeling or blistering
MechlorethamineSkin irritation: rash, itching, or redness
MogamulizumabInfusion reactions (fever, chills, rash), skin rash, infections (especially upper respiratory tract infections)
Phototherapy (NBUVB, PUVA)Skin redness (erythema),dry or itchy skin, increased risk of skin aging and skin cancer (especially with PUVA)
Steroids (high potency)Atrophy of the skin, stretch marks (striae), increased risk of infection (fungal or bacterial)
TLR agonistInjection site reactions (pain, redness, or swelling), flu-like symptoms (fever, chills, fatigue), rash or skin irritation
TSEBTSkin dryness and peeling, hair loss, fatigue
Tumour MF (stage IIB)AlloSCTGraft-versus-host disease, graft failure, infections
Bexarotene p.o.Leukopenia, hypothyroidism, hyperlipidaemia
Bexarotene p.o. and MTXLeukopenia, hypothyroidism, hyperlipidaemia and myelotoxicity, hepatotoxicity, nephrotoxicity
Brentuximab vedotinNeuropathy, allergic reaction, shortness of breath, redness or swelling of the face
CHOPDiarrhea, infections, low blood cell count
HDAC inhibitorDiarrhea, infections, low blood cell count
IFN-αAnemia, neutropenia, hepatotoxicity
IFN-α and bexarotene p.o.Anemia, neutropenia, hepatotoxicity and myelotoxicity, hepatotoxicity, nephrotoxicity
IFN-α and phototherapy (NBUVB, PUVA)Anemia, neutropenia, hepatotoxicity and skin redness (erythema),dry or itchy skin, increased risk of skin aging and skin cancer (especially with PUVA)
Local RT ± other SDTsSkin redness and irritation (erythema), fatigue, skin peeling or blistering
MogamulizumabInfusion reactions (fever, chills, rash), skin rash, infections (especially upper respiratory tract infections)
MonochemotherapyNausea and vomiting, alopecia, bone marrow suppression
MTXMyelotoxicity, hepatotoxicity, nephrotoxicity
TSEBTSkin dryness and peeling, hair loss, fatigue
Erythrodermic MF/SS (stage IIB–III)AlloSCTGraft-versus-host disease, graft failure, infections
Bexarotene p.o.Leukopenia, hypothyroidism, hyperlipidaemia
Brentuximab vedotinNeuropathy, allergic reactions, shortness of breath, redness or swelling of the face
CHOPDiarrhea, infections, low blood cell count
ECPHypotension, fatigue, infections or site reactions
HDAC inhibitorDiarrhea, infections, low blood cell count
IFN-αAnemia, neutropenia, hepatotoxicity
IFN-α and bexarotene p.o.Anemia, neutropenia, hepatotoxicity and myelotoxicity, hepatotoxicity, nephrotoxicity
IFN-α and phototherapy (NBUVB, PUVA)Anemia, neutropenia, hepatotoxicity and skin redness (erythema), dry or itchy skin, increased risk of skin aging and skin cancer (especially with PUVA)
MogamulizumabInfusion reactions (fever, chills, rash), skin rash, infections (especially upper respiratory tract infections)
MonochemotherapyNausea and vomiting, alopecia, bone marrow suppression
MTXMyelotoxicity, hepatotoxicity, nephrotoxicity
TSEBTSkin dryness and peeling, hair loss, fatigue
Lymph node and internal organ involvement MF (stage IV)AlemtuzumabInfections, infusion-related reactions (fever, chills, rash, nausea, and low blood pressure during or shortly after the infusion)
AlloSCTGraft-versus-host disease, graft failure, infections
Bexarotene p.o. and MTXLeukopenia, hypothyroidism, hyperlipidaemia and myelotoxicity, hepatotoxicity, nephrotoxicity
Brentuximab vedotinNeuropathy, allergic reactions, shortness of breath, redness or swelling of the face
CHOPDiarrhea, infections, low blood cell count
ECPHypotension, fatigue, infections or site reactions
HDAC inhibitorsDiarrhea, infections, low blood cell count
IFN-αAnemia, neutropenia, hepatotoxicity
MogamulizumabInfusion reactions (fever, chills, rash), skin rash, infections (especially upper respiratory tract infections)
Phototherapy (NBUVB, PUVA)Skin redness (erythema), dry or itchy skin, increased risk of skin aging and skin cancer (especially with PUVA)
TSEBTSkin dryness and peeling, hair loss, fatigue

[i] AlloSCT – allogeneic blood stem cell transplantation, CHOP – cyclophosphamide + hydroxydaunomycin (doxorubicin) + oncovin (vincristine) + prednisone, ECP – extracorporeal photopheresis, HDAC – histone deacetylase, IFN-α – interferon α, MTX – methotrexate, NBUVB – narrow band UVB, PUVA – psoralen + ultraviolet A, RT – local radiation therapy, TSEBT – total skin electron beam therapy.

The choice of treatment depends on the severity of the disease, patient preferences, and the occurrence of unacceptable adverse effects. In the case of early-stage (stage I–IIA) patch-plaque MF, the first line of treatment consists of skin-directed therapy (SDT) alone or in combination with other SDTs. In the case of tumour-stage MF (stage IIB), the first line of treatment is local radiation therapy (RT) alone and/or other SDTs (most commonly PUVA), systemic therapy and/or SDT, systemic therapy and/or RT or total skin electron beam therapy (TSEBT) for generalized lesions. When a patient is diagnosed with erythrodermic MF/SS (stage IIB–III), the first line of treatment is systemic therapy combined with SDT (most commonly PUVA). Finally, when a patient is diagnosed with MF involving lymph nodes and internal organs (stage IV), the first line of therapy is systemic therapy and/or RT to control local skin involvement (Table 1) [24].

Registered therapies

Topical treatment

For early-stage MF, the principal treatment modality mainly encompasses topical therapies, due to their efficacious outcomes and minimized systemic side effects [5].

Glucocorticosteroids

Topical corticosteroids are frequently employed as an initial treatment strategy. In a study conducted by Zackheim et al. [6], 79 patients with patch or plaque stage MF were treated with topical corticosteroids of class I to III b.i.s. for 3 months. The overall response rate (ORR) was 94% for patients with stage T1 and 82% with stage T2 disease.

Chlormethine gel

An alternative topical therapy is chlormethine gel. In a clinical trial conducted by Lessin et al. [7] evaluating patients with early-stage MF (IA–IIA) a daily monotherapy of 0.02% gel was prescribed for a duration of 12 months. The results indicated an ORR of 58.5% with 13.8% achieving complete remission rate (CRR). Additionally although FDA prescribing guidelines recommend daily application of chlormethine gel, off-label modifications to the dosing schedule and the concomitant use of topical steroids are practiced by experienced clinicians. These adjustments may enhance treatment tolerance by minimizing adverse reactions, particularly during the initial months of therapy [8, 9]. Furthermore, the PROVe (a PROspective, observational study assessing outcomes, adverse events (AEs), treatment patterns, and quality of life (QoL), in patients diagnosed with MF treated with Valchlor and the other therapies) study demonstrated that chlormethine gel is an effective therapeutic option which effectively reduces skin lesion severity and improves QoL in real-world clinical practice [10]. In the PROVe study, 298 patients with MF were monitored. At 12 months, 44.4% (chlormethine + corticosteroids + other) and 45.1% (chlormethine + other) of efficacy-evaluable patients were responders. Peak response was observed at 18 months in the chlormethine + other group.

Bexarotene gel

Another option is the application of bexarotene gel 1%, a derivative of retinoid. In Phase I and II Breneman’s [11] clinical trials evaluating bexarotene gel 1% b.i.s. for 10.5 months for dermal-directed therapy in patients with MF/SS, an ORR of 63% was observed.

Topical Toll-like receptor (TLR) agonists

Additionally, topical Toll-like receptor (TLR) agonists like imiquimod 5% and resiquimod 0.03/0.06% represent a novel class of topical treatments. Imiquimod functions through the activation of TLR7, culminating in the subsequent release of cytokines and elicitation/induction of an inflammatory cascade. In a cohort study, 20 patients at early MF stages were treated with imiquimod 5%, as a result the ORR was 80% and CRR was 45% [12]. Resiquimod exhibits the potential to stimulate systemic cells by activation in circulating dendritic cells (DCs) and natural killer (NK) cells. In a Phase I clinical trial involving 12 patients with stage IA–IIA, 0.03% and 0.06% topical resiquimod gel treatments q.d. for 24 weeks led to ORR of 75% and CRR of 30% [13].

Phototherapy

Phototherapy represents a crucial therapeutic approach that can be employed either as a standalone treatment or in conjunction with topical therapies for patients with limited-stage disease [14]. These methods encompass narrowband ultraviolet B (NBUVB, 311 nm) and the combination of 8-methoxypsoralen or 5-methoxypsoralen plus ultraviolet A (PUVA). NBUVB is administered for early-stage MF and in those cases the CRR was from 90% to 95%. Meanwhile, PUVA therapy is generally preferred for patients with darker skin tones, with thick plaques and those who relapse after initial NBUVB therapy [12]. This treatment involves the administration of a photosensitizing drug metoxalen which, when activated by UVA light, induces changes in the DNA of skin cells to slow their growth. PUVA CRRs were calculated to be 85% for stage IA, 65% for stage IB, and 85% for stage IIA [15].

Methotrexate

A retrospective survey of 48 patients with MF/SS treated with low-dose methotrexate (MTX) and followed for at least 1 year was conducted by Alenezi et al. [16]. The initial weekly dosage of MTX ranged from 10 to 25 mg for 3.5 months. CRR was achieved in 10 (21%) patients and partial response rate (PRR) in 25 (52%) patients. Among the 35 responders, 57% relapsed after a median of 11 months. Methotrexate was discontinued in 44 patients, mainly due to primary or secondary failure and toxicity. Despite these limitations, the benefit/risk ratio of low-dose methotrexate remains favourable even though the study by Abd-el-Baki et al. [17] revealed that the incidence of transformation of MF to large cell lymphoma (LCL) was significantly higher in patients who received MTX compared to those who did not (14.3% vs. 1.8%, p = 0.03).

Extracorporeal photopheresis (ECP)

During ECP, whole blood is collected from a cubital vein or implanted catheter, and leukocytes are separated. The leukocytes, or buffy coat, are then exposed to UVA irradiation in the presence of the photosensitizing agent 8-methoxypsoralen before being reinfused into the patient [18]. In the context of extended observation of MF/SS patients subjected to treated with ECP, CRR was 41% after a median of 19.6 months, meaning 40 ECP treatments/cycles. ECP was administered on 2 consecutive days every 4 weeks until a threshold skin response was observed or after 12 treatments. If needed, ECP could have been accelerated to two treatments every 14 days for up to four cycles, or until a successful response was achieved [19].

Radiotherapy (RT)

MF/SS are radiosensitive diseases, so radiation therapy, administered with a curative intent, is a viable treatment option for patients with localized MF both limited and advanced-stage MF or SS [3].

In a study by O’Malley et al. [20], low-dose radiotherapy (LDRT, 8 Gy) administered to MF patients resulted in clone eradication in 5 of 16 lesions and a > 90% reduction in 11 of 16 lesions, with no recurrences. A retrospective review further showed that early-stage, high-risk MF patients who received radiation had significantly prolonged overall survival compared to those who did not, highlighting the potential benefit of incorporating radiotherapy into treatment regimens for these patients.

Brachytherapy is another treatment option for patients with MF. In a study by Kamieniecka et al. [21], 3D superficial brachytherapy was used to preserve lens function while successfully treating malignant lesions on the left upper eyelid and forehead. The patient received 10 Gy in 5 fractions, with the lens receiving 5 Gy (45–55% isodose range). High-dose-rate brachytherapy using an iridium-192 source and Flexitron afterloading was employed. Regression of the lesions was observed 3 weeks post-treatment.

Total skin electron beam therapy (TSEBT)

In addition to local radiation therapy, low-dose TSEBT treats widespread disease by irradiating the superficial dermis while sparing deeper tissues. Targeted external beam radiation therapy (EBRT) is effective for large plaques or nodules [22]. The current therapeutic inclination favours low-dose TSEBT, typically 10–12 Gy administered across 6–8 fractions. In one observation for 9.6 months the CRR was 92% with mild toxicities [23].

Bexarotene

The oral rexinoid bexarotene, selective to retinoid X receptors (RXR), was approved by the FDA in 1999 for CTCL treatment, and subsequently as a topical gel. In Poland, as part of a drug program, bexarotene is made available to patients with MF and SS in monotherapy. Bexarotene treatment triggers apoptosis in MF/SS cells by downregulating both its receptors and the apoptosis inhibitor surviving. Patients receiving oral doses of 300 mg/m²/day achieved ORR of 45% in 48 weeks [24].

Interferons a and g

Interferons (INF), namely INF α-2b and INF g-1b, manifest pleiotropic and immunomodulatory actions in MF/SS. These multifaceted effects are correlated with ORR ranging from 50% to 70% and CRR falling within the 20% to 30% range for 14.9 months. Such therapeutic efficacy is predominantly observed in patients with limited-stage disease [25, 26]. IFN-α, generated by plasmacytoid dendritic cells (DC), is specifically targeted to counteract various immunological deficiencies inherent in MF/SS, thereby assisting in the enhancement of the anti-tumour response [27]. IFN-γ mechanisms of action are analogous to those of IFN-α, serving to activate CD8+ T lymphocytes and NK cells.

Brentuximab vedotin

The study of brentuximab vedotin vs physician’s choice in CD30-positive cutaneous T-cell lymphoma (ALCANZA) validates the therapeutic superiority associated with brentuximab vedotin (BV). Patients received either BV (1.8 mg/kg IV every 3 weeks, up to 16 cycles) or the physician’s choice of MTX (5–50 mg orally weekly) or bexarotene (300 mg/m² orally daily, up to 48 weeks). Within the MF patient population, final data showed that BV significantly outperformed physician’s choice, with an objective response rate at 4 months (ORR4) of 54.7% vs 12.5% and CRR of 17.2% vs 1.6% [28]. In Poland, as part of a drug program, BV is available to patients with MF in monotherapy.

Classical systemic chemotherapy

In MF/SS sustained therapeutic responses to conventional chemotherapeutic regimens remain elusive, with the median interval to subsequent treatment interventions typically demarcated in months [29]. In the pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma (PROPEL) study patients with confirmed CTCL who had progressed after at least one prior therapy received pralatrexate 30 mg/m² weekly for 6 weeks in 7-week cycles. Of the 115 patients enrolled, 111 were treated, and the RR among 109 evaluable patients was 29% (31 of 109), including CR 12 (11%) and 20 PR (18%), with a median duration of 10.1 months [30]. In the study by Fierro et al. [31], 35 patients with MF/SS were treated with 6 cycles of cyclophosphamide, vincristine, and prednisone (COP) or cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) at standard doses. The ORR was 40%, with the CRR of 23%.

Gemcitabine, a pyrimidine antimetabolite, has shown to be an effective monotherapy in patients with advanced-stage MF/SS who have undergone extensive prior therapy. In a study by Zinzani et al. [32], 70% of the patients with relapsed MF (21 out of 30) achieved CR or PR. Pegylated liposomal doxorubicin, a liposome-encapsulated form of doxorubicin, allows higher tumour concentrations with improved safety. A prospective multicentre study by Quereux et al. [33] reported 56% RR in MF/SS patients (14/25).

Allogeneic blood stem cell transplantation

Allogeneic blood stem cell transplantation (alloSCT) has shown a strong graft-versus-lymphoma (GvL) effect in MF/SS. In a trial with 80% of around 20 advanced MF/SS patients achieved durable remissions after 3 years following alloSCT [34]. AlloSCT should be considered for patients with advanced disease and poor prognosis, but without comorbidities. Optimal results are achieved with complete or near-complete remission before transplantation [4].

Mogamulizumab

Mogamulizumab (KW-0761) is a defucosylated, humanized monoclonal antibody that selectively targets the C-C chemokine receptor type 4 (CCR4), thereby enhancing antibody-dependent cell-mediated cytotoxicity (ADCC). CCR4, involved in lymphocyte skin trafficking is consistently expressed on tumour cells in T-cell malignancies like CTCL including MF and SS. In the clinical trial called ”Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma“ (MAVORIC), the patients treated with 0.32 mg/kg mogamulizumab for 24 weeks, observed ORR of 50% [35].

The ongoing MAVORIC retrospective study, which is actively enrolling patients, aims to assess real-world outcomes of mogamulizumab treatment in Europe. A post hoc subgroup analysis of ORRs by clinical stage showed higher response rates in advanced stages: 26% in stage III, 36% in stage IV, 16% in stage IIB, and 19% in stages IA/IIA. Compartment-specific ORRs were 42% for skin, 68% for blood, and 17% for lymph nodes [36]. In 2018, the FDA approved mogamulizumab for the treatment of CTCL in patients who have failed at least one prior systemic therapy. In Poland, as part of a drug program, mogamulizumab is also available as monotherapy for patients with MF and SS, when the stage of MF is classified as IB or higher, according to the TNMB staging system (ISCL and EORTC classification), and after the patient had previously undergone at least one line of systemic therapy.

HDAC inhibitors

Vorinostat (suberoylanilide hydroxamic acid, SAHA) and romidepsin (depsipeptide) serve as examples of histone deacetylase (HDAC) inhibitors, specifically targeting and impeding the enzymatic functions of class I and II HDACs. By modulating the deacetylation of histones and non-histone proteins, these inhibitors exert a regulatory influence over gene expression and chromatin structure [37, 38].

In a trial conducted by Olsen et al. [39], a dose of 400 mg q.d. of oral vorinostat was established for 74 MF/SS patients. The observed ORR was 30%, with a median duration of response of 185 days. In two clinical trials, romidepsin was administered as a 4-hour i.v. infusion at a dose of 14 mg/m2 on days 1, 8, and 15 of a 4-week cycle. The data revealed the ORR of 38% in patients with advanced-stage disease, with the median duration of response surpassing 12 months.

Ongoing emerging clinical studies highlight the need for novel therapeutic agents and updated treatment standards for patients with MF/SS. These advancements are driven by extensive research into the molecular characteristics, intracellular signalling pathways, and interactions with the tumour microenvironment in MF/SS, which continue to unveil new potential therapeutic targets.

Therapies in clinical trials studies

Mogamulizumab and magrolimab

During the development of the study, a phase Ib/II clinical trial (NCT04541017) is being conducted to identify the optimal dose, as well as any potential benefits and/or side effects of administering Hu5F9-G4 (magrolimab) in combination with mogamulizumab for treating patients with stage IB-IV MF or SS that have relapsed or are refractory to treatment. Magrolimab is a monoclonal antibody that blocks CD47, a “don’t-eat-me” signal often overexpressed on cancer cells. By inhibiting CD47, magrolimab promotes macrophage-mediated phagocytosis of tumour cells.

Patients receive either the combination therapy or mogamulizumab alone every 28 days for up to 12 cycles. Follow-up is necessary for up to 2 years after completion of study treatment [40].

Programmed cell death protein 1: pembrolizumab and nivolumab

Immune checkpoint antibodies targeting programmed cell death protein 1 (PD-1) function by engaging inhibitory PD-1 molecules expressed on exhausted T cells. Notably, PD-1 is also expressed on malignant T cells in MF and SS. In a phase II trial, 24 patients with refractory MF/SS were treated with pembrolizumab, an anti-PD-1 monoclonal antibody, at a dose of 2 mg/kg every 3 weeks for up to 24 months. The treatment resulted in the ORR of 38%, with 9 patients responding, including 2 who achieved CRR [41]. Further studies have been conducted with nivolumab, another agent targeting PD-1. A phase I trial consisting of 13 patients with MF revealed the ORR of 15%. There are also case reports signifying a favourable response in refractory SS patients [42]. Current research is exploring the combination of PD-1 inhibitors with other therapeutic agents, such as IFN-γ, in order to ascertain the potential synergistic effects.

Alemtuzumab

Alemtuzumab, denoted as a humanized IgG1 monoclonal antibody, specifically binds to the CD52 antigen, which is ubiquitously present on B-cells, T-cells, and monocytes. A phase II trial assessing 22 patients with advanced-stage MF/SS, indicated the ORR of 55% and CRR of 32%. Alemtuzumab was administered using a rapidly escalating initial dosing regimen, followed by a maintenance dose of 30 mg three times per week for up to 12 weeks [43].

Given the inherent proclivity for infectious adversities, an alternative protocol employing 10 mg maximum per administration of alemtuzumab s.c. was appraised in 14 SS patients. 85.7% (12/14) of patients showed ORR, including 21.4% (3) with CRR [44].

SIRPαFc

CD47, a prevalent glycoprotein, acts to inhibit macrophage phagocytosis through its interaction with the macrophage-bound SIRP-α. It is noteworthy that Sézary cells exhibit an enhanced expression of CD47. Experimental studies (NCT02890368) have introduced a decoy receptor, TTI-621 (secreting CD47 blocker signal regulatory protein a-SIRPαFc), via administration in SS patients, resulting in a significant reduction in circulating Sézary cells. TTI-621 was injected intralesionally in a dose-escalation study (cohorts 1–5: 1 mg, 3 mg, or 10 mg single dose, or three 10 mg doses weekly for 1–2 weeks) and in expansion cohorts (cohorts 6–9: 2-week induction at the maximum tolerated dose, with optional weekly continuation) [45].

Systemic TLR-agonists

In the realm of MF/SS therapeutic investigations, systemic deployment of TLR-agonists has been subjected to limited trials. The utilization of cytosine-guanine oligodeoxynucleotides (CpG-ODN), potent binders of TLR-9, facilitates the activation of plasmacytoid DCs. Within a phase I trial, clinical advanced refractory MF/SS patients were subcutaneously treated with escalating CpG-ODN doses yielded the ORR of 32%. Notably, as the study did not ascend to the maximal tolerable dose threshold, these initial findings suggest a potential therapeutic efficacy and warrant further exploration [46].

NM-IL-12

Interleukin-12 (IL-12), mainly produced by monocytes and dendritic cells (DCs), activates NK cells and CD8+ T cells, promoting IFN-γ secretion. In MF/SS advanced-stage patients, particularly with SS, show marked Th1 immunity deficits, with reduced DCs and lower IFN-γ and IL-12 levels. Given the shift towards Th2 immunity, marked by increased IL-4, IL-13, and IL-5, IL-12’s ability to suppress T2 cytokine secretion in SS patients holds important scientific relevance [47]. Initial findings suggest that NM-IL-12 can be safely co-administered with low-dose TSEBT (12 Gy, 1 Gy per fraction over 3 weeks) in MF/SS patients. The ORR was 88% (29/33), with 9 CRR. Median time to response was 7.6 weeks [48].

Cobomarsen (MRG-106)

Imbalance of miRNA propels MF/SS, with heightened oncogenic miR-155 driving abnormal proliferation. Unravelling its gene interactions could refine therapies, restoring miRNA balance for effective CTCL intervention [49]. Cobomarsen (MRG-106), an oligonucleotide miR-155 inhibitor, underwent phase I for MF/SS. Involving 24 patients, MRG-106 was administered via subcutaneous/IV injection (300–900 mg/dose, weekly/biweekly) and intralesional injection (75 mg/dose). Positive outcomes were seen in 95% of subjects, improving lesions and skin via mSWAT scores [50]. Post-treatment MRG-106 reduced T cell clonality, and 50% of patients showed a 50% mSWAT reduction, especially after multiple cycles [51].

Enhanced KIR3DL

IPH4102 represents a novel monoclonal antibody that selectively targets killer cell immunoglobulin-like receptor 3DL2 (KIR3DL2), a cell surface protein with notable expression in MF/SS, particularly in SS, effectively eliminating these cells through antibody-dependent cell cytotoxicity. In the context of a phase I trial targeting relapsed MF/SS, 44 patients were administered IPH4102, with no instances of dose-limiting toxicity observed. The agent’s favourable tolerance profile yielded a noteworthy ORR in 16 out of 44 patients [52]. Presently, ongoing multicohort phase II trials are underway to further show IPH4102’s therapeutic prospects.

PI3K, SYK/JAK inhibitors

Phosphatidylinositol 3-kinase (PI3K) orchestrates downstream oncogenic pathways. Its δ and γ isoforms, prominent in hematopoietic cells, offer targets for MF/SS. Tenalisib (RP6530), a pioneering dual PI3K δ/γ inhibitor at doses of 200–800 mg b.i.d. p.o. in 28-day cycles. Among 35 evaluable patients, the ORR was 45.7% (3 CRR, 13 PRR), with a median response duration of 4.9 months. Responding tumours showed reduced levels of CD30, IL-31, and IL-32a. A phase I/II combination study with romidepsin is currently ongoing [53]. Critical in MF/SS are the SYK and JAK pathways. A phase IIa study assessed cerdulatinib, a reversible ATP competitive inhibitor of SYK/JAK. Administered at 30 mg b.i.d., cerdulatinib yielded the ORR of 35%, diverging between MF (45%) and SS (17%) [54]. Cerdulatinib exhibited notable clinical efficacy and a favourable safety profile, warranting further exploration in ongoing trials.

BNZ-1

BNZ-1, a pegylated peptide antagonist, selectively engages the shared g-chain signalling receptor of interleukin-2 (IL-2), IL-9, and IL-15 cytokines. This interaction culminates in the disruption of downstream signalling cascades activated by these cytokines, yielding potential therapeutic implications for MF/SS patients. In a clinical trial, individuals with refractory MF/SS were subjected to weekly i.v. administrations of BNZ-1 at varied doses (0.5/1/2/4 mg/kg). Clinical responses were 1 CRR and 11 ORR (58%). Importantly, BNZ-1 exhibited favourable tolerability, underscoring its prospective utility as an innovative therapeutic approach for MF/SS management [55].

Discussion

Therapeutic decisions are highly individualized, depending on patient age, functional health, disease stage, progression, with the most objective assessment of skin lesions being and prior treatment responses.

This personalized approach tailors treatment to the unique clinical profile of each patient. Thanks to stem from extensive research into the molecular characteristics, intracellular signalling pathways, and tumour microenvironment interactions in MF/SS, revealing new potential treatment targets. However, toxicity remains a significant concern with these therapies and must be carefully assessed when evaluating novel agents and combination regimens. The high-frequency ultrasound (HFUS) seems to be a useful tool for monitoring infiltration of the skin [56]. In summary, the field of investigational therapies for MF/SS currently encompasses a diverse array of promising interventions at varying stages of clinical development. These therapies target a range of mechanisms and pathways, reflecting the complexity of MF/SS and the need for tailored treatment approaches. Ongoing clinical investigations are essential to validate and expand upon these promising preliminary results, advancing the field of MF/SS therapeutics.

Ethical approval

Not applicable.

Conflict of interest

The authors declare no conflict of interest.

References

1 

Campo E, Jaffe ES, Cook JR, et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood 2022; 140: 1229-53.

2 

Lee H. Mycosis fungoides and Sézary syndrome. Blood Res 2023; 58 (Suppl 1): 66-82.

3 

Sokołowska-Wojdyło M, Olszewska B, Chmielowska E, et al. Primary cutaneous lymphomas. Diagnostic and therapeutic guidelines of the Polish Dermatological Society (PTD) and Polish Lymphoma Research Group (PLRG). Dermatol Rev 2023; 110: 647-74.

4 

Latzka J, Assaf C, Bagot M, et al. EORTC consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome – update 2023. Eur J Cancer 2023; 195: 113343.

5 

Hristov AC, Tejasvi T, Wilcox RA. Cutaneous T-cell lymphomas: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol 2023; 98: 193-209.

6 

Zackheim HS, Kashani-Sabet M, Amin S. Topical corticosteroids for mycosis fungoides. Experience in 79 patients. Arch Dermatol 1998; 134: 949-54.

7 

Lessin SR, Duvic M, Guitart 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.

8 

Kim EJ, Geskin L, Guitart J, et al. Real-world experience with mechlorethamine gel in patients with mycosis fungoides-cutaneous lymphoma: preliminary findings from a prospective observational study. J Am Acad Dermatol 2020; 83: 928-30.

9 

Garcia-Saleem TJ, Stonesifer CJ, Khaleel AE, Geskin LJ. Management of mycosis fungoides with topical chlormethine/mechlorethamine gel: a Columbia University Cutaneous Lymphoma Center Experience. Acta Derm Venereol 2021; 101: adv00544.

10 

Kim EJ, Guitart J, Querfeld C, et al. The PROVe Study: US real-world experience with chlormethine/mechlorethamine gel in combination with other therapies for patients with mycosis fungoides cutaneous T-cell lymphoma. Am J Clin Dermatol 2021; 22: 407-14.

11 

Breneman D, Duvic M, Kuzel T, et al. Phase 1 and 2 trial of bexarotene gel for skin-directed treatment of patients with cutaneous T-cell lymphoma. Arch Dermatol 2002; 138: 325-32.

12 

Shipman AR, Scarisbrick J. New treatment options for mycosis fungoides. Indian J Dermatol 2016; 61: 119.

13 

Rook AH, Gelfand JM, Wysocka M, et al. Topical resiquimod can induce disease regression and enhance T-cell effector functions in cutaneous T-cell lymphoma. Blood 2015; 126: 2765.

14 

Dogra S, Mahajan R. Phototherapy for mycosis fungoides. Indian J Dermatol Venereol Leprol 2015; 81: 124-35.

15 

Olsen EA, Hodak E, Anderson T, et al. Guidelines for phototherapy of mycosis fungoides and Sézary syndrome: a consensus statement of the United States Cutaneous Lymphoma Consortium. J Am Acad Dermatol 2016; 74: 27-58.

16 

Alenezi F, Girard C, Bessis D, et al. Benefit/risk ratio of low-dose methotrexate in cutaneous lesions of mycosis fungoides and Sézary syndrome. Acta Derm Venereol 2021; 101: adv00384.

17 

Abd-el-Baki J, Demierre MF, Li N, Foss FM. Transformation in mycosis fungoides: the role of methotrexate. J Cutan Med Surg 2002; 6: 109-16.

18 

Cho A, Jantschitsch C, Knobler R. Extracorporeal photopheresis-an overview. Front Med (Lausanne) 2018; 5: 236.

19 

Knobler R, Duvic M, Querfeld C, et al. Long-term follow-up and survival of cutaneous T-cell lymphoma patients treated with extracorporeal photopheresis. Photodermatol Photoimmunol Photomed 2012; 28: 250-7.

20 

O’Malley JT, de Masson A, Lowry EL, et al. Radiotherapy eradicates malignant T cells and is associated with improved survival in early-stage mycosis fungoides. Clin Cancer Res 2020; 26: 408-18.

21 

Kamieniecka H, Rutkowski J. 3D technique in superficial brachytherapy: new attitude in old field of mycosis fungoides. J Contemp Brachyther 2024; 16: 310-3.

22 

Thomas TO, Agrawal P, Guitart J, et al. Outcome of patients treated with a single-fraction dose of palliative radiation for cutaneous T-cell lymphoma. Int J Radiat Oncol Biol Phys 2013; 85: 747-53.

23 

Neelis KJ, Schimmel EC, Vermeer MH, et al. Low-dose palliative radiotherapy for cutaneous B- and T-cell lymphomas. Int J Radiat Oncol Biol Phys 2009; 74: 154-8.

24 

Zhang C, Duvic M. Retinoids: therapeutic applications and mechanisms of action in cutaneous T-cell lymphoma. Dermatol Ther 2003; 16: 322-30.

25 

Jumbou O, N’Guyen JM, Tessier MH, et al. Long-term follow-up in 51 patients with mycosis fungoides and Sézary syndrome treated by interferon-alfa. Br J Dermatol 1999; 140: 427-31.

26 

Kaplan EH, Rosen ST, Norris DB, et al. Phase II study of recombinant human interferon gamma for treatment of cutaneous T-cell lymphoma. J Natl Cancer Inst 1990; 82: 208-12.

27 

Yoo EK, Cassin M, Lessin SR, Rook AH. Complete molecular remission during biologic response modifier therapy for Sézary syndrome is associated with enhanced helper T type 1 cytokine production and natural killer cell activity. J Am Acad Dermatol 2001; 45: 208-16.

28 

Horwitz SM, Scarisbrick JJ, Dummer R, et al. Randomized phase 3 ALCANZA study of brentuximab vedotin vs physician’s choice in cutaneous T-cell lymphoma: final data. Blood Adv 2021; 5: 5098-106.

29 

Hanel W, Briski R, Ross CW, et al. A retrospective comparative outcome analysis following systemic therapy in mycosis fungoides and Sezary syndrome. Am J Hematol 2016; 91: E491-5.

30 

O’Connor OA, Pro B, Pinter-Brown L, et al. Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol 2011; 29: 1182-9.

31 

Fierro MT, Quaglino P, Savoia P, et al. Systemic polychemotherapy in the treatment of primary cutaneous lymphomas: a clinical follow-up study of 81 patients treated with COP or CHOP. Leuk Lymphoma 1998; 31: 583-8.

32 

Zinzani PL, Baliva G, Magagnoli M, et al. Gemcitabine treatment in pretreated cutaneous T-cell lymphoma: experience in 44 patients. J Clin Oncol 2000; 18: 2603-6.

33 

Quereux G, Marques S, Nguyen JM, et al. Prospective multicenter study of pegylated liposomal doxorubicin treatment in patients with advanced or refractory mycosis fungoides or Sézary syndrome. Arch Dermatol 2008; 144: 727-33.

34 

Schlaak M, Theurich S, Pickenhain J, et al. Allogeneic stem cell transplantation for advanced primary cutaneous T-cell lymphoma: a systematic review. Crit Rev Oncol Hematol 2013; 85: 21-31.

35 

Kim YH, Bagot M, Pinter-Brown 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: 1192-204.

36 

Horwitz S, Zinzani PL, Bagot M, et al. Lack of impact of type and extent of prior therapy on outcomes of mogamulizumab therapy in patients with cutaneous T cell lymphoma in the MAVORIC trial. Leuk Lymphoma 2021; 62: 3109-18.

37 

Gloghini A, Buglio D, Khaskhely NM, et al. Expression of histone deacetylases in lymphoma: implication for the development of selective inhibitors. Br J Haematol 2009; 147: 515-25.

38 

Qu K, Zaba LC, Satpathy AT, et al. Chromatin accessibility landscape of cutaneous T cell lymphoma and dynamic response to HDAC inhibitors. Cancer Cell 2017; 32: 27-41.e4.

39 

Olsen EA, Kim YH, Kuzel TM, et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol 2007; 25: 3109-15.

40 

Whittaker SJ, Demierre MF, Kim EJ, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 2010; 28: 4485-91.

42 

Khodadoust MS, Rook AH, Porcu P, et al. Pembrolizumab in relapsed and refractory mycosis fungoides and Sézary syndrome: a Multicenter Phase II Study. J Clin Oncol 2020; 38: 20-8.

43 

Shen K, Liu Y, Cao X, Zhou D, Li J. Successful treatment of refractory Sezary syndrome by anti-PD-1 antibody (nivolumab). Ann Hematol 2017; 96: 687-8.

44 

Lundin J, Hagberg H, Repp R, et al. Phase 2 study of alemtuzumab (anti-CD52 monoclonal antibody) in patients with advanced mycosis fungoides/Sezary syndrome. Blood 2003; 101: 4267-72.

45 

Bernengo MG, Quaglino P, Comessatti A, et al. Low-dose intermittent alemtuzumab in the treatment of Sézary syndrome: clinical and immunologic findings in 14 patients. Haematologica 2007; 92: 784-94.

46 

Querfeld C, Thompson JA, Taylor MH, et al. Intralesional TTI-621, a novel biologic targeting the innate immune checkpoint CD47, in patients with relapsed or refractory mycosis fungoides or Sézary syndrome: a multicentre, phase 1 study. Lancet Haematol 2021; 8: e808-17.

47 

Kim YH, Girardi M, Duvic M, et al. Phase I trial of a Toll-like receptor 9 agonist, PF-3512676 (CPG 7909), in patients with treatment-refractory, cutaneous T-cell lymphoma. J Am Acad Dermatol 2010; 63: 975-83.

48 

Suchin KR, Cassin M, Gottleib SL, et al. Increased interleukin 5 production in eosinophilic Sézary syndrome: regulation by interferon alfa and interleukin 12. J Am Acad Dermatol 2001; 44: 28-32.

49 

Kim YH, Hoppe RT, Rook A, Maity A. A single-arm PHASE 2A study of NM-IL-12 (rHu-IL12) in patients with mycosis fungoides-type CTCL (MF) undergoing low-dose total skin electron beam therapy (LD-TSEBT). Blood 2016; 128: 4165.

50 

Lai P, Wang Y. Epigenetics of cutaneous T-cell lymphoma: biomarkers and therapeutic potentials. Cancer Biol Med 2021; 18: 34-51.

51 

Querfeld C, Foss FM, Pinter-Brown LC, et al. Phase 1 study of the safety and efficacy of MRG-106, a synthetic inhibitor of microRNA-155, in CTCL patients. Blood 2017; 130 (Suppl 1): 820.

52 

Bagot M, Porcu P, Marie-Cardine A, 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: 1160-70.

53 

Huen A, Haverkos BM, Zain J, et al. Phase I/Ib study of tenalisib (RP6530), a dual PI3K δ/γ inhibitor in patients with relapsed/refractory T-cell lymphoma. Cancers (Basel) 2020; 12: 2293.

54 

Horwitz SM, Feldman TA, Hess BT, et al. A phase 2 study of the dual SYK/JAK inhibitor cerdulatinib demonstrates good tolerability and clinical response in relapsed/refractory peripheral T-cell lymphoma and cutaneous T-cell lymphoma. Blood 2019; 134: 466.

55 

Querfeld C, William BM, Sokol L, et al. 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.

56 

Wohlmuth-Wieser I, Ramjist JM, Shear N, Alhusayen R. Morphologic features of cutaneous T-cell lymphomas using dermoscopy and high frequency ultrasound. J Clin Med 2020; 10: 17.

Copyright: © 2024 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2025 Termedia Sp. z o.o.
Developed by Bentus.