Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2020 Oct 23;192(4):683–696. doi: 10.1111/bjh.17117

The changing therapeutic landscape, burden of disease, and unmet needs in patients with cutaneous T‐cell lymphoma

Julia J Scarisbrick 1,, Martine Bagot 2, Pablo L Ortiz‐Romero 3
PMCID: PMC7894136  PMID: 33095448

Summary

Cutaneous T‐cell lymphomas (CTCLs) have a chronic, relapsing course, and the most common subtypes are mycosis fungoides and Sézary syndrome. The disease causes visible skin alterations and can also cause alopecia, pruritus and pain, all of which can impact patients’ health‐related quality of life (HRQoL). The goal of treatment is to reduce symptoms and prevent disease progression. However, treatment recommendations are often based on low levels of evidence due to the lack of well‐designed randomised clinical trials and treatment guidelines, and approved drugs vary considerably across different countries and regions. Currently, available treatments rarely lead to durable remissions and eventually become less effective, meaning patients often require multiple therapy changes. Skin‐directed therapies (SDTs) are first‐line treatments for early‐stage CTCL, whereas systemic therapies may be needed for early‐stage disease that does not respond to SDT or for advanced‐stage disease. However, patients can experience significant side‐effects with these treatments or may be unable to tolerate them. Hence, there is an unmet need for effective therapies with good safety profiles for the treatment of early‐ and late‐stage CTCL. Here, we review current treatment guidelines, investigational and approved treatments, the impact of CTCL on patients’ HRQoL, and the treatment of pruritus.

Keywords: CTCL, mycosis fungoides, quality of life, Sézary syndrome, treatment strategies

Current Management of CTCL

Cutaneous T‐cell lymphomas (CTCLs) represent <4% of non‐Hodgkin lymphomas in the USA 1 and present as erythema, papules, patches, plaques, nodules or tumours. 2 Several CTCL subtypes exist, but the most prevalent form is mycosis fungoides (MF), 1 , 3 followed by Sézary syndrome (SS), 4 which have estimated incidences of 0·3–0·9/100 000 and 0·1/1 000 000 respectively in Europe. 5 , 6 Patients with early‐stage disease have a good prognosis, with 10‐year overall survival (OS) rates of 50–90% depending on various prognostic factors. However, the prognosis for patients with advanced‐stage disease is poorer, with 10‐year OS rates of 15–53% 7 , 8 and a median OS of 2–5 years. 9 Diagnosis of MF and SS is based on multicompartmental assessment of disease in the skin (T), lymph nodes (N), visceral organs (M), and peripheral blood (B), referred to collectively as TNMB staging. Previously, blood involvement was not included in clinical staging of the disease, 10 but this was updated in 2007 11 following evidence that showed increased blood involvement adversely affects disease prognosis. 12 , 13

Durable remission is uncommon in MF and SS, and patients tend to be treated with consecutive treatments until loss of response or intolerability. Periods of no therapy are common, particularly in the early stages, and are termed ‘expectant’ therapy or ‘watch and wait’. In addition, symptom relief via methods such as anti‐pruritic medication and emollients may be used. Allogeneic haematopoietic stem cell transplantation (allo‐HSCT) may be indicated in a small number of eligible patients and is the only curative option. 8 , 14 , 15 , 16 Therefore, the goals of therapy are to improve or clear lesions and to control symptoms across all affected disease compartments (skin, blood, lymph nodes, and viscera), to minimise the risk of progression 17 and to avoid treatment‐related toxicity. 18

Earlier lines of therapy generally consist of skin‐directed therapies (SDTs), radiotherapy, and biological response modifiers [National Comprehensive Cancer Network (NCCN) 2020]. 8 , 19 Most treatments result in only partial responses 8 and most patients receive therapies until loss of response, meaning they move on to receive consecutive or combination treatments and often recycle their previous therapies. When systemic chemotherapy is required, single agents are preferred over combination chemotherapy such as the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) because of the short duration of responses and the higher toxicity profiles associated with combination chemotherapy (NCCN 2020). 19 Therapies with fewer side‐effects and an absence of cumulative toxicity are often given in an ongoing or maintenance fashion 20 to improve and maintain disease control and quality of life (QoL) (NCCN 2020). 19 Systemic therapies are frequently combined with SDT.

Treatment recommendations are generally based on low levels of evidence due to the scarcity of well‐designed randomised clinical trials 8 , 20 , 21 , 22 (NCCN 2020). 19 Few CTCL therapies are approved by the European Medicines Agency (EMA), with the majority having orphan status (Table I) 23 , 24 , 25 , 26 , 27 , 28 , 29 . In the latest treatment guidelines from the British Association of Dermatologists (BAD) and UK Cutaneous Lymphoma Group (UKCLG), clinical trials are included as second‐ and third‐line treatment options for all disease stages except MF Stage IA/IIA, confirming a need for new therapies. 8

Table I.

Current systemic therapies approved by the EMA for treatment of CTCL.

Drug Year approved Reference
Roferon‐A 1996 Roferon‐A Patient Information Leaflet[ 28 ]
Oral bexarotene 2001 Targretin EPAR Medicine Overview[ 29 ]
Topical mechlorethamine gel 2012 (Orphan Designation) Ledaga EPAR Summary[ 25 ]
2017 (Marketing Authorisation) Ledaga Product Information[ 25 ]
Brentuximab vedotin 2012 (Orphan Designation) Brentuximab vedotin Orphan Designation[ 24 ]
2012 (Marketing Authorisation) Adcetris Product Information 23
Mogamulizumab 2018 (Orphan Designation) Mogamulizumab Orphan Designation[ 26 ]
2018 (Marketing Authorisation) Poteligeo Product Information 27

CTCL, cutaneous T‐cell lymphoma; EMA, European Medicines Agency; EPAR, European public assessment report.

The identification of specific CTCL biomarkers has allowed for the development of targeted therapies such as monoclonal antibodies, histone deacetylase inhibitors (HDACi), and proteasome inhibitors, 30 but there is still a lack of curative therapies.

Cutaneous T‐cell lymphomas can also greatly impact patients’ health‐related QoL (HRQoL) through physical symptoms, such as pruritus and pain, 31 , 32 and non‐physical factors, such as insomnia, anxiety and depression. 33 As CTCL is visible on the skin, patients experience relatively poor HRQoL, 34 , 35 , 36 even in the very early stages of disease. 34 Further, women with newly diagnosed MF or SS and those with alopecia have significantly impaired HRQoL. 37 Hence, it is important to consider the patient’s HRQoL when selecting treatment, particularly in patients with late‐stage MF or SS. 38 The effect of CTCL on patient QoL should however be considered during treatment option selection, particularly the effect of leaving early‐stage disease untreated. Patients themselves may wish for early intervention even with a low chance of success.

In addition, there is a profound effect upon those family members who become caregivers to a person diagnosed with CTCL. Caregivers for those with advanced CTCL in particular can face extreme distress as the disease progresses, given the poor disease prognosis and lack of curative therapies. 39 This burden and its increase with time, is reflected in their own loss in QoL. 40

The present review will describe the unmet needs, novel therapies (Table II) 23 , 27 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , and disease burden experienced by patients with CTCL.

Table II.

Novel therapies in ongoing clinical trials for early‐ and late‐stage CTCL.

Disease stage Name Treatment type Mechanism Reference
Early FLASH Phototherapy Synthetic hypericin in the form of a topical gel is used as a photosensitiser with fluorescent light therapy

NCT02448381

NIHR Innovation Observatory Evidence Briefing (2017) 53

E7777 Recombinant cytotoxic fusion protein Diphtheria toxin fragments A and B are combined with human IL‐2 to target the IL‐2 receptor, which is overexpressed in CD25+ CTCL NCT01871727
Low‐dose PUVA Phototherapy Low‐dose PUVA used as maintenance therapy Vieyra‐Garcia et al. (2019) 58
Resiquimod Skin‐directed therapy A toll‐like agonist against TLR7 and TLR8, leading to an increase in IFN‐γ and TNF‐α Rook et al. (2015) 56
Lenalidomide Immunomodulatory drug An analogue of thalidomide that induces Th1 cytokine production and NK cell and T‐cell activation Querfeld et al. (2014) 55
MNA‐D Microneedle array Doxorubicin is delivered directly into patches and plaques via a dissolvable MNA NCT02192021
Pimecrolimus Immunomodulatory topical cream Inhibits calcineurin, which inhibits cytokine expressions and blocks T‐cell activation

EudraCT Number: 2014‐001377‐14

Eichenfield and Beck (2003) 48

Late Brentuximab vedotin Monoclonal antibody Directed against CD30 and conjugated with auristatin E, a tubulin inhibitor, which causes apoptosis by disrupting microtubules within malignant cells

Adcetris Prescribing Information 41

Adcetris Product Information 23

Cobomarsen (MRG‐106) Oligonucleotide inhibitor Inhibits microRNA‐155, which is involved in the proliferation of malignant cells, and helps to induce apoptosis

T‐cell Lymphoma Forum Industry Innovations miRagen MicroRNA Oral Presentation (2019) 57

NCT03713320

Mogamulizumab Monoclonal antibody Directed against CCR4, which is overexpressed on malignant T cells, and defucosylated to improve ADCC

Poteligeo Prescribing Information 54

Poteligeo Product Information 27

Panobinostat Pan‐histone deacetylase inhibitor Acts against all class I, II, and IV histone deacetylase enzymes

Atadja (2009) 43

Duvic et al. (2013) 47

Pembrolizumab Monoclonal antibody Targets programmed death‐1 receptor Khodadoust et al. (2020) 49
Pegylated liposomal doxorubicin Chemotherapy Pegylation of doxorubicin in liposomal form leads to a longer half‐life with reduced toxicity

Alpdogan et al. (2019) 42

Dummer et al. (2012) 46

Alemtuzumab Monoclonal antibody Directed against CD52 and kills malignant cells through ADCC or non‐complement‐mediated killing

Lowenstein et al. (2006) 51

Bernengo et al. (2007) 44

Bortezomib Proteasome inhibitor Inhibits transcription factor NF‐κB, leading to the suppression of TGF‐β1 and IL‐10, which induces apoptosis and may inhibit CTCL cell migration

Zinzani et al. (2007) 59

Chang et al. (2015) 45

Dimethyl fumarate Immunomodulatory drug Inhibits transcription factor NF‐κB, which is used by cancer cells to block apoptosis

Kiessling et al. (2009) 50

Nicolay et al. (2016) 52

IPH4102 (alone or in combination with chemotherapy) Monoclonal antibody Directed against KIR3DL2 (immunoglobulin‐like receptor), which is overexpressed in SS NCT03902184
TTI‐621 Recombinant fusion protein Binds to CD47 and prevents an inhibitory antiphagocytic (do not eat) signal to macrophages NCT02890368

ADCC, antibody‐dependent cellular cytotoxicity; CCR4, C‐C chemokine receptor 4; CTCL, cutaneous T‐cell lymphoma; IFN, interferon; IL, interleukin; KIR3DL2, killer cell immunoglobulin‐like receptor, three Ig domains and long cytoplasmic tail 2; NF‐κB, nuclear factor κ‐light‐chain‐enhancer of activated B cells; NK cell, natural killer cell; PUVA, psoralen and ultraviolet A; SS, Sézary syndrome; TGF, transforming growth factor; TLR, toll‐like receptor; TNF, tumour necrosis factor.

Treatment guidelines

Various groups have published guidelines for MF and SS treatment, such as the International Society for Cutaneous Lymphoma (ISCL); the European Organisation for Research and Treatment of Cancer (EORTC); BAD and UKCLG; and the National Comprehensive Cancer Network (NCCN, USA‐based; NCCN 2020). 8 , 19 , 20 In general, CTCL treatments are listed as first‐, second‐ or third‐line, and no preference is given for treatments within each line of therapy. SDTs tend to be recommended for early‐stage disease, with biological response modifiers reserved for late‐stage disease. Patients may be given several first‐line treatments before they move to second‐line therapies.

There is no contemporary, global consensus on treatment guidelines. In the NCCN guidelines, systemic therapies, such as brentuximab vedotin, are recommended as second‐line therapy even for Stage IA patients with persistent skin disease, while the BAD recommends the treatment as second‐line therapy from Stage IIB only. Regional guidelines mean that treatment can vary greatly between countries. The available treatments also differ (e.g. vorinostat is approved by the United States Food and Drug administration [FDA] but not by the EMA) and even the structure of guidelines; e.g. the EORTC divides therapies into first‐ and second‐line treatments for each disease stage, while the BAD and UKCLG designate first‐, second‐ and third‐line treatments. 8 , 20 However, several first‐line therapies are usually tried before moving on to second‐line options. Although more drugs are licensed by the FDA than by the EMA, a large‐scale, global study of patients with advanced MF or SS found no difference in survival rates between patients in the USA and in Europe 9 , 60 .

Recommendations in updated treatment guidelines have also changed. For example, total skin electron beam therapy (TSEB) was previously recommended by the EORTC as a first‐line treatment for early‐stage disease, but is now recommended as a second‐line treatment. Additionally, monochemotherapy has changed from being a second‐line treatment to a first‐line treatment for Stage IIB MF. 20 , 61 For TSEB, these changes are most likely due to an increase in the assessed quality of evidence available for TSEB in the most recent guidelines. In the case of monochemotherapy, it is most likely due to considering evidence for poly‐ and monochemotherapy separately in the most recent guidelines; polychemotherapy is still considered as a second‐line treatment. Allo‐HSCT is recommended for advanced‐stage disease in the updated 2017 EORTC guidelines, 20 whereas it was not recommended in the previous version. 61 While allo‐HSCT is potentially curative, 15 , 16 there is still much work to be done in understanding the optimal conditioning regimens, the best moment to recommend HSCT, and how prior systemic therapies will affect the transplant outcome. 15 , 62 Furthermore, allo‐HSCT is only available in limited, specialist centres, making it not easily accessible.

Early disease

Unmet needs

Patients with early‐stage disease (Stage IA/IIA) have the best long‐term survival prognosis, 7 , 8 with Stage IA patients having a prognosis similar to that of the age‐matched general population. 63 However, 20–25% of patients with early‐stage disease progress to more advanced disease. 7 , 64 Additionally, patients with early‐stage disease can exhibit significant blood involvement, 65 which is generally associated with a poorer prognosis compared with patients without blood involvement. 66 Currently, the Cutaneous Lymphoma International Prognostic Index (CLIPi) identifies patients as low‐, intermediate‐ and high‐risk of progression, 7 but its predictive ability may be limited. 64

Cutaneous T‐cell lymphoma has a chronic, relapsing course and patients generally undergo multiple consecutive therapies, with the intent of clearing the skin disease, minimising recurrence, preserving patients’ HRQoL and preventing disease progression. 36 , 67 First‐line treatments for early‐stage CTCL are skin‐directed, including topical corticosteroids, nitrogen mustards, and topical retinoids, as well as phototherapy, photochemotherapy and radiotherapy. 8 , 20 Response rates to topical therapies in patients with early‐stage disease range from 54% to 94% (topical corticosteroids: 82–94%, nitrogen mustards: 58·5–94%, topical retinoids: 54–80%), with complete response rates of 10–86% (topical corticosteroids: 25–63%, nitrogen mustards: 13·8–86%, topical retinoids: 10–60%); 68 however, side‐effects may limit treatment tolerability. 68 Phototherapy can produce relatively high response rates (63–100%), but is associated with side‐effects such as secondary cancers, and patients often relapse soon after therapy ends. 8 , 20 In addition, access to treatments can also be an issue. For example, nitrogen mustards are not currently available in the UK and Spain.

Therefore, there is still a clear need for more effective and tolerable therapies for early‐stage patients, including those who do not respond adequately to SDTs.

Novel therapies for early‐stage disease

Phase I and II trials

Phase I and II trials are summarised in Table III. 55 , 56 , 69

Table III.

Summary of Phase I and II trials related to early‐stage disease.

Drug Details
Immunomodulatory drug
Resiquimod

Immune response modifier with anti‐viral and anti‐tumour activity.

Phase I results: 11 out 12 patients (CTCL, Stage IA/IIA) experienced significant improvement. 56

Phase II study (NCT03292406): expected completion March 2021.

Lenalidomide

Approved for use in patients with multiple myeloma, myelodysplastic syndromes, and various B‐cell lymphomas. 69

Phase II results: 28% ORR, all PR, seen across all disease stages, including refractory patients; patients had received a median of six prior therapies. 55

Phase I and I/II studies: evaluating lenalidomide in combination with durvalumab (NCT03011814) and brentuximab vedotin (NCT03302728).

Pimecrolimus Phase II study (EudraCT no. 2014‐001377‐14): for patients with MF Stage IA/IIA currently ongoing.
Chemotherapy
MNA‐D

Small adhesive‐like patches (aMNA), which have dozens of very small microneedles loaded with extremely low doses of doxorubicin.

Phase I study (NCT02192021): the safety and dose‐finding phase is complete; the efficacy and safety phase expected completion date is December 2022.

CTCL, cutaneous T‐cell lymphoma; MF, mycosis fungoides; MNA, microneedle applicator; MNA‐D, microneedle array‐doxorubicin; ORR, overall response rate; PR, partial response.

Phase III trials

Fluorescent Light Activated Synthetic Hypericin (FLASH)

The Fluorescent Light Activated Synthetic Hypericin (FLASH) study is an ongoing, multicentre, randomised, double‐blind, placebo‐controlled Phase III trial in patients with Stage IA/IIA MF (NCT02448381). Patients receive photodynamic therapy using a topical gel comprising synthetic hypericin or placebo ointment in combination with fluorescent light therapy.

Hypericin is a natural compound found in St John’s wort (Hypericum perforatum). It contains a photosensitizer that can be excited at wavelengths within the visible spectrum, giving it an advantage over other photosensitizers that require ultraviolet light. 70 In the FLASH trial, the synthetic hypericin SGX301 (Soligenix, Inc., Princeton, NJ, USA) is used as an ointment that can be applied topically to skin lesions and that is more actively taken up by cancer cells than normal cells. After exposure to fluorescent light, reactive oxygen species develop, which kill the cancer cells. In early studies, 58% of patients with CTCL responded to SGX301 versus 8% of patients treated with placebo, with an acceptable safety profile. 71 SGX301 has received orphan drug designation in the European Union and in the USA.

SGX301 may provide a safer, more convenient alternative to photochemotherapy with psoralen plus ultraviolet A (PUVA), which has been associated with an increased risk of secondary skin cancers in patients exposed to high numbers of PUVA treatments. 8 , 72

Denileukin diftitox

Denileukin diftitox is a recombinant cytotoxic fusion protein, combining human interleukin (IL)‐2 with diphtheria toxin fragments A and B. 73 The drug targets cells expressing the IL‐2 receptor (IL‐2R), which is highly expressed in CD25‐positive (+) CTCL. 74 In a placebo‐controlled pivotal trial, 75 response rates in patients treated with two different doses of denileukin diftitox were 38–49%, including complete responses in 9–11% of cases, which was statistically significant compared with placebo. 76 Progression‐free survival (PFS) was also significantly longer compared with the placebo group (794–971 days). In another open‐label Phase III study, patients with MF and SS Stage IA–III achieved an overall response rate (ORR) of 30·6%. 77

Denileukin diftitox was approved by the FDA under the trade name Ontak® (Seragen, Inc., San Diego, CA, USA ) 75 in 1999 for patients with persistent or recurrent CD25+ CTCL, and was the first drug in its class approved to treat a human disease. 74 However, the drug was discontinued due to production issues. 78

E7777 is a new, improved formulation that was evaluated in a Phase I dose‐finding study of patients with persistent and recurrent CTCL in Japan, in whom it showed anti‐tumour activity. 73 Additionally, a Phase II study with the ORR as the primary objective was completed in April 2019 (NCT02676778), and a Phase III, multicentre, open‐label, single‐arm study assessing dose, efficacy and safety (NCT01871727) was completed in March 2020.

Low‐dose PUVA

Psoralen plus ultraviolet A was used as maintenance therapy in patients with early‐stage MF by 88% of respondents to an online survey of members of the ISCL conducted in 2004, 79 despite the recognised increase in incidence of squamous cell cancer and melanoma in patients receiving multiple courses of phototherapy. Additionally, respondents indicated a lack of consistency in the frequencies and durations of maintenance phototherapy. 79

A clinical trial was conducted to investigate the benefits of using low‐dose PUVA as a maintenance treatment in patients with MF (NCT01686594). 58 At the time of study initiation, the EORTC recommended against the use of PUVA for maintenance therapy due to a lack of evidence for efficacy and safety. 61 In the updated EORTC recommendations, published in 2017, the use of PUVA as maintenance therapy is acknowledged, although the guidelines state that there is still a lack of supportive evidence. 20

In the low‐dose PUVA maintenance trial, patients received PUVA during a 12–24‐week induction phase. 58 Patients with complete responses were then randomised to either PUVA maintenance twice weekly for 9 months or to no maintenance; however, the trial closed early due to low accrual. The median disease‐free remission period was 15 months in patients who received maintenance PUVA compared with 4 months in patients without maintenance. In biomarker assessments, high baseline levels of C‐X‐C motif chemokine ligand 9 (CXCL9), CXCL11, CXCL12, tumour necrosis factor ligand superfamily member 13 (TNFSF13), and CXCL13 were negatively associated with response to PUVA, while high baseline levels of TNF‐related weak inducer of apoptosis (TWEAK, also known as TNFSF12) were positively associated and patients who achieved a complete response had high levels of TWEAK. Owing to the low accrual, this study is limited by the small sample size in the randomised group (n = 19), but it is the first prospective randomised trial evaluating the use of PUVA as maintenance therapy.

Advanced disease

Unmet needs

Patients with Stage IIB–IV disease have a very poor median OS, ranging from 3–5 years. 9 Although allo‐HSCT is a curative treatment option for patients with CTCL, at least half of patients who receive this treatment relapse within 1 year, 15 , 62 , 80 and graft‐versus‐host disease (GvHD) can lead to severe morbidity and death. 81 Additionally, further investigation is needed into factors affecting outcomes after allo‐HSCT, such as the optimal conditioning regimens as well as patient selection and the best time for transplantation during the disease course. 81

There is also a great unmet need for more effective therapies to improve outcomes in this advanced‐stage patient population. Immunotherapies are the preferred first‐line therapy over chemotherapy, 8 , 20 (NCCN 2020) 19 a systematic review of 13 chemotherapy regimens in CTCL found a short median time to next therapy (TTNT) of only 3·9 months for mono‐ or polychemotherapy. 82 Additionally, a second review found the relative risk of death for both mono‐ and polychemotherapy to be elevated compared with other lines of treatment. 60 Biomarker and genetic analyses have led to the development of several new targeted therapies. 30

Novel therapies for late‐stage disease

Phase I and II trials

Phase I and II trials are summarised in Table IV. 45 , 46 , 47 , 49 , 50 , 59 , 83 , 84 , 85 , 86 , 87 , 88 , 89 , 90 , 91 , 92 , 93 , 94 , 95 , 96 , 97

Table IV.

Summary of phase I and II trials related to late‐stage disease.

Drug Details
HDACi
Panobinostat

Approved in 2015 by the FDA for multiple myeloma.

Phase II study results (NCT00425555): patients were stratified by previous bexarotene treatment. ORR was 17·3% for all patients, 15·2% for patients previously treated with bexarotene, and 20·0% for bexarotene‐naïve patients. Median DOR was 5·6 months in patients previously treated with bexarotene, and had not been reached at the time of publication in the bexarotene‐naïve group. 47 Median PFS was 4·2 and 3·7 months for patients with and without previous bexarotene exposure respectively. 47

Preclinical data suggested that panobinostat may be useful in treating CTCL patients whose disease is resistant to vorinostat through downregulation of STAT protein activity. 90

Resminostat

Previously tested in Phase I or II studies in patients with hepatocellular carcinoma, Hodgkin lymphoma, biliary tract and pancreatic cancer, and non‐small cell lung cancer. 86 , 92 , 93 , 95

Phase II study (NCT02953301): assessing the ability of resminostat as a maintenance treatment for patients with advanced‐stage MF or SS who achieved disease control with another systemic therapy. Estimated completion date December 2021.

Humanised monoclonal antibody
Pembrolizumab (anti‐PD1)

Targets PD‐1 receptor.

Phase II study results: patients had received a median of ≥4 prior systemic therapies. ORR was 38%, with two patients achieving CR and seven achieving PR. Of the nine responders, all had ongoing responses at last follow‐up. Median DOR had not been reached at the time of publication, with a median response follow‐up time of 58 weeks. 49

Ongoing interventional trials include a Phase II study evaluating pembrolizumab the radiation therapy in MF and SS (NCT03385226); a Phase I study evaluating pembrolizumab with total skin electron beam radiation therapy in MF and SS (NCT03617224); a Phase I dose‐finding study of pembrolizumab in combination with pralatrexate, decitabine, or both drugs (NCT03240211); and a Phase II study of pembrolizumab combined with romidepsin in patients with relapsed/refractory MF (NCT03278782).

Tislelizumab (anti‐PD1) Phase II study (NCT03493451): estimated completion date June 2020.
Atezolizumab (anti‐PD1) Phase II study (NCT03357224): estimated completion date June 2025.
Durvalumab (anti‐PD1) Phase I/II study (NCT03011814): estimated completion date June 2023.
Alemtuzumab (anti‐CD52) Phase II study results: safety and efficacy of alemtuzumab was evaluated in 22 patients with advanced, pretreated MF and SS in a single‐arm Phase II trial. 87 Objective responses were seen in 55% of patients, with a CR rate of 32% and PR rate of 23%. The treatment appeared to be particularly effective for patients with SS, with six of seven experiencing complete clearance of Sézary cells. In patients with lymph node involvement, six of 11 achieved CR. Patients with erythroderma also responded better than those with plaques or tumours (ORR: 69% and 40%; CR: 38% and 30% respectively).
AFM13 (anti‐CD30, anti‐CD16A)

Phase Ia/IIb study (NCT03192202): estimated completion date August 2020.

Phase II study (NCT04101331): estimated completion date February 2023.

IPH4102 (anti‐KIR3DL2)

KIR3DL2 is expressed predominantly in SS. 84

Phase II study (NCT03902184): evaluate IPH4102 alone or in combination with chemotherapy in patients with relapsed/refractory MF/SS.

Chemotherapy
PLD

Showed efficacy as a monotherapy in patients with relapsed/refractory CTCL. 88 , 89 , 97

Phase II study results: 20 patients (40·8%) achieved an objective response and three achieved CR. Patients with Stage IVA/B disease had lower response rates than those with Stage IIB (22% vs. 52%). Median DOR was 6 months and 80% of patients had a substantial improvement in cutaneous lymphoma manifestations. 46

Romidepsin

Approved by the FDA for the treatment of patients with CTCL who have received ≥1 systemic therapy.

Phase I study (NCT02512497): investigating the use of romidepsin in combination with fludarabine and busulfan, both before and after allogeneic HSCT, as a maintenance therapy to help to control lymphoma (including CTCL) or leukaemia, as well as to determine the optimal dose and safety of the treatment (NCT02512497). Estimated completion date December 2020.

Proteasome inhibitor
Bortezomib

Approved by the FDA for treatment of multiple myeloma and mantle cell lymphoma. It leads to apoptosis and may also inhibit CTCL migration by suppressing TGF‐β1 and IL‐10 through NF‐κB inhibition. 45

Phase II study results: 15 patients with relapsed/refractory CTCL or peripheral T‐cell lymphoma. 59 ORR was 70% in patients with MF and one patient achieved CR.

Phase II study (NCT03487133): investigation in combination with dexamethasone in patients with relapsed/refractory CTCL. Estimated completion date December 2023.

Immunomodulatory drug
DMF

Currently approved for multiple sclerosis (Tecfidera Product Information) 94 and plaque psoriasis (Skilarence Product Information). 91

Inhibitor of transcription factor NF‐κB, which appears to block apoptosis in cancer cells. 83 , 85 , 96 Inhibition of NF‐κB‐induced apoptosis in malignant cells from patients with SS, while leaving healthy T cells unaffected. 50 Inhibition of NF‐κB led to downregulation of FHC in malignant cells, which in turn lead to a rise in free cytosolic iron and ultimately cell death through reactive oxygen species. Downregulation of FHC was not seen in healthy T cells.

Phase IIa study (NCT02546440): estimated completion date September 2021.

TTI‐621 Phase I study (NCT02890368): dose‐escalation trial involving patients with relapsed/refractory solid tumours or MF. Estimated completion date December 2019.

CR, complete response; DMF, dimethyl fumarate; DOR, duration of response; FDA, USA Food and Drug Administration; FHC, ferritin heavy chain; HSCT, haematopoietic stem cell transplantation; KIR3DL2, killer cell immunoglobulin‐like receptor, three Ig domains and long cytoplasmic tail 2; MF, mycosis fungoides; NF‐κB, nuclear factor κ‐light‐chain‐enhancer of activated B cells; ORR, overall response rate; PD‐1, programmed death‐1; PFS, progression‐free survival; PLD, pegylated liposomal doxorubicin; PR, partial response; SS, Sézary syndrome; STAT, signal transducer and activator of transcription.

Phase III trials

The current E7777 trial (NCT01871727), as mentioned previously, is open to patients with any stage of CTCL. A previous Phase III study using the old formulation, denileukin diftitox, included 15 (42%) patients with late‐stage CTCL, of whom four (27%) achieved a response. 77

For late‐stage disease, systemic therapies are recommended.

Brentuximab vedotin

Brentuximab vedotin (Adcetris®, Takeda Pharmaceutical Co. Ltd., Tokyo, Japan) is an anti‐CD30 monoclonal antibody conjugated with monomethyl auristatin E (MMAE), a cytotoxic drug that inhibits tubulin, leading to apoptosis through microtubule disruption (Adcetris Product Information). 23 It specifically targets CD30+ CTCL, which is present in ~30% of patients with CTCL. 98 In a multicentre, international, open‐label, randomised, Phase III trial, patients were randomised to brentuximab vedotin or physician’s choice (methotrexate or bexarotene, 64 patients in each treatment arm). 99 Patients must have had CD30+ MF or primary cutaneous anaplastic large‐cell lymphoma (pcALCL) and have had received at least one prior therapy. Patients with additional diagnoses of SS or B2 blood involvement were excluded from the study. In total, 128 patients were in the intent‐to‐treat population: 97 patients with CD30+ MFand 31 with pcALCL. The primary end‐point was an ORR of ≥4 months.

At a median follow‐up of 22·9 months, the 4‐month ORR was 56·3% in the brentuximab vedotin arm and 12·5% in the physician’s choice arm. The PFS was significantly improved and symptom reduction was also seen. 99 The median TTNT was significantly greater for brentuximab vedotin‐treated patients (14·2 months) compared with those receiving the physician’s choice (5·6 months). 100 Peripheral neuropathy was highly prevalent in the brentuximab vedotin‐treated patients compared with physician’s choice‐treated patients (44/66 and four of 62 respectively). 99 Based on these data, brentuximab vedotin was approved by the FDA in 2017 for the treatment of CD30+ MF and pcALCL following more than one prior systemic therapy (Adcetris Prescribing Information) 41 and by the EMA in 2018 for CD30+ CTCL (not including patients with SS) following more than one prior systemic therapy (Adcetris Product Information), 23 but it can only be administered for a maximum of 16 infusions. Early reports of real‐world data of brentuximab vedotin have shown similar results in CD30+ CTCL. 101

Brentuximab vedotin is also being evaluated in combination with lenalidomide in patients with relapsed/refractory CTCL regardless of CD30 expression status (NCT03302728) and in patients with Stage IB–IVB CTCL (NCT03409432).

Mogamulizumab

Mogamulizumab (Poteligeo®, Kyowa Kirin, Inc., Tokyo, Japan) is a defucosylated, humanised, monoclonal antibody targeting C‐C chemokine receptor 4 (CCR4), which is thought to play a critical role in the migration of malignant T cells to the skin and is overexpressed in CTCL at all disease stages. 102 Defucosylation improves the antibody‐dependent cellular cytotoxicity (ADCC) of the drug. 102 , 103 Mogamulizumab has demonstrated efficacy in patients with MF Stage IB–IVB and SS across disease compartments of the skin, blood, and lymph nodes.

Mogamulizumab was investigated in an international, open‐label, randomised controlled Phase III trial for relapsed/refractory MF or SS. This study was the largest randomised study of systemic therapy in CTCL and the first to use PFS as a primary end‐point, in line with international consensus guidelines. 11 , 104 In total, 372 patients with relapsed/refractory Stage IB–IVB MF or SS (204 and 168 patients respectively) who had failed at least one prior systemic therapy were randomised 1:1 to receive vorinostat or mogamulizumab. There were similar numbers of patients with MF and SS in each treatment arm: 105 patients with MF and 81 with SS for mogamulizumab; and 99 patients with MF and 87 SS for vorinostat. Vorinostat‐treated patients who experienced intolerable toxicity or who had confirmed disease progression following at least two treatment cycles could crossover to mogamulizumab treatment.

The PFS, based on global composite scores of response in each disease compartment (i.e. skin, blood, lymph nodes, and viscera), was significantly longer with mogamulizumab (7·7 months) compared with vorinostat (3·1 months). 104 The ORR in skin, blood, and lymph node compartments was improved for 42%, 68%, and 17% of mogamulizumab‐treated patients and 16%, 19%, and 4% of vorinostat‐trested patients respectively. The ORR was improved overall for 28% of patients treated with mogamulizumab compared with 5% of the patients treated with vorinostat. 104 The TTNT was significantly prolonged for the mogamulizumab‐treated patients compared with the vorinostat‐treated patients (11 and 3·5 months respectively). 105 Between the mogamulizumab and vorinostat treatment discontinuation due to adverse events was similar (21·7% and 23·7% respectively), and there were also similar rates of drug‐related serious treatment‐emergent adverse events (19·6% and 16·7% respectively). 106

Mogamulizumab was approved by the Japanese Pharmaceuticals and Medical Devices Agency (PDMA) in 2018 for relapsed/refractory CTCL, 107 by the FDA in 2018 for adult patients with relapsed/refractory MF or SS after at least one prior systemic therapy (Poteligeo Prescribing Information), 54 and by the EMA in 2018 for adult patients with MF or SS who had received at least one prior systemic therapy (Poteligeo Product Information). 102 , 27

Lacutamab

Another monoclonal antibody currently under investigation is IPH4102 (Lacutamab, Innate Pharma, Marseille, France), 108 which targets the cell surface protein killer cell immunoglobulin (Ig)‐like receptor, three Ig domains and long cytoplasmic tail 2 (KIR3DL2) and, upon binding induces apoptosis of the cell via ADCC and antibody‐dependent cellular phagocytosis. 109 This protein is overexpressed in T‐cell lymphomas, including in >85% of patients with SS. In a Phase I trial, 43% (15/35) of patients with SS had a confirmed global overall response. 84 A Phase II trial (TELLOMAK) is ongoing (NCT03902184).

Vorinostat

The HDACi vorinostat (Zolinza®, Merck & Co., Inc., Whitehouse Station, NJ, USA) is also approved by the FDA for the treatment of cutaneous manifestations in patients with CTCL who have progressive, persistent, or recurrent disease or following two systemic therapies. In a Phase II trial in patients with MF or SS refractory to or intolerant of prior therapies, the response rate was 24% by modified severity weighted assessment tool (mSWAT), with a median duration of response of 15 weeks. 110 In a Phase IIB trial in patients with MF and SS Stage ≥IB with progressive, persistent, or recurrent disease after two prior systemic therapies, the response rate was 29·7% and the median duration of response was ≥185 days. 11

Romidepsin

Romidepsin (Istodax®, Celgene Corp., Summit, NJ, USA) is another HDACi that is FDA‐approved for patients with CTCL and peripheral T‐cell lymphoma (PTCL) following at least one prior systemic therapy. In a pivotal Phase II trial of patients with Stage IB–IVA CTCL who had received more than one prior systemic therapy, the ORR was 34% by mSWAT, with six patients achieving a complete response. 111 There are no approved HDACis in Europe for CTCL.

Effect of CTCL on HRQoL

Along with minimising symptoms and preventing progression, improving patients’ HRQoL is an important therapy goal in CTCL. CTCL is a generally incurable disease that is characterised by visible skin alterations, such as patches, plaques and tumours, as well as by alopecia, pruritus and pain. The disease requires chronic treatment with a series of drugs, which can have significant side‐effects and often lose effectiveness, meaning patients may require multiple therapy changes.

There are a variety of tools to assess patients’ HRQoL that include domains for symptoms, function and emotions. Instruments commonly used in epidemiological and clinical studies of patients with CTCL are Skindex‐29, a tool developed specifically for patients with skin disease, and generic tools such as the Functional Assessment of Cancer Therapy‐General (FACT‐G), 36‐item Short Form Health Survey, EORTC Quality of Life Core Questionnaire (QLQ‐C30), General Health Questionnaire and the Spitzer Quality of Life Index. 112

Using the FACT‐G questionnaire and the skin disease‐specific Skindex‐29 instrument, Demierre et al. 34 assessed HRQoL in 22 patients with CTCL. Their findings showed that patients with CTCL had low scores in physical, social/family, emotional, and functional well‐being and symptoms. 34 In addition, patients with more advanced‐stage disease had worse scores across all scales of both HRQoL instruments, possibly reflecting more severe symptoms in advanced‐stage CTCL and a greater risk of death. 34

Wright et al. 36 also found lower HRQoL scores using Skindex‐29 in patients with advanced‐stage CTCL compared with patients with earlier‐stage disease, with 88% of the participants reporting pruritus in the 4 weeks prior to completing the questionnaire; 46% reported experiencing pruritus ‘often’ or ‘all the time’. However, advanced‐stage CTCL is more than just a skin disease, and as such HRQoL may be affected in other ways that are not addressed by Skindex‐29. In a survey conducted in 630 patients with CTCL, 88% of respondents indicated that they were bothered by itching, 83% by scaling, and 94% by skin redness. Some pain was reported by 41% of respondents and 13% reported ‘quite a bit’ to ‘very much’ pain. 35 Pruritus incidence and severity were associated with both early‐ and advanced‐stage CTCL, measured using the visual analogue scale (VAS) for itch. 113 Further, CTCL impacted patients’ abilities to meet the needs of their family, interfered with their job and led to missed work days, and significantly impacted their social interactions and sleep. 35 Fatigue was also reported by 66% of respondents and the disease led to patients feeling frustrated, angry and embarrassed, and worrying about the seriousness of their disease and possible death.

Jonak et al. (2019) 112 reviewed the Skindex‐29 HRQoL assessments from three different epidemiological studies 34 , 36 , 114 and found reproducible results, with all studies showing that a majority of patients with CTCL worry about the seriousness of their illness and progression of the disease, and experience sleeping disorders and difficulties during their work, leisure time and social life.

Molloy et al. (2020) 37 used the Skindex‐29 tool to examine the HRQoL of patients enrolled in the international, observational, MF/SS Prospective Cutaneous Lymphoma International Prognostic Index (PROCLIPI) study. They found that newly diagnosed women and those with alopecia experienced the worst HRQoL. 37 They suggested that a questionnaire specific to CTCL is necessary in order to better understand the HRQoL of these patients.

The need for a CTCL‐specific tool

Due to the lack of effective curative therapies for CTCL, patients may live for many years experiencing the effects of the disease, with unpleasant symptoms such as pain, itching and functional deficit if the disease affects their hands or feet, and the emotional effect of living with long‐term cancer.

In a 2018 two‐arm clinical study comparing mogamulizumab with vorinostat, Skindex‐29 and FACT‐G were used to measure patient HRQoL as a secondary end‐point. 104 Overall, both Skindex‐29 and FACT‐G showed that mogamulizumab was favoured over vorinostat. 115 However, when looking at the functioning domain, the differences between the results at treatment cycles seven and nine were significantly different for Skindex‐29, but not for FACT‐G. In the emotional domain, Skindex‐29 showed vorinostat to improve on the baseline results, while FACT‐G showed it to worsen. The discrepancies in results between these different tools emphasise the importance of using more disease‐specific tools to measure HRQoL in patients with CTCL.

Molloy et al. (2020) 37 reported on 237 patients [60·3% male; median age 60 years (interquartile range 49–70)], of whom 179 had early‐stage MF and 58 had advanced‐stage MF/SS. HRQoL, as measured by Skindex‐29, was worse in women, patients with SS, patients with late‐stage MF and in those with raised lactate dehydrogenase, alopecia, higher mSWAT score and confluent erythema. Female sex and alopecia were also independent predictors of worse global HRQoL.

McCaffrey et al. (2019) 116 recently developed a CTCL‐specific HRQoL tool (named MF/SS‐CTCL QoL) in collaboration with the Cutaneous Lymphoma Foundation in order to fill this gap. Validation of the tool was performed through the PatientsLikeMe Open Research Exchange, by having patients (n = 126) complete the MF/SS‐CTCL QoL questionnaire and also Skindex‐29 as a comparator. A proportion of the patients (n = 66) then repeated MF/SS‐CTL QoL after 5 days in order to assess test–retest reliability. However, the patient group that participated in the development of the tool predominantly had Stage I disease; therefore, data from advanced‐stage patients are also required to improve the robustness of the tool.

The importance of pruritus control in improving patients’ HRQoL

Pruritus affects a high proportion of patients and can affect other HRQoL factors by leading to difficulty falling asleep, depression, 34 , 35 and suicidal ideation (personal communication from P Ortiz‐Romero, February 2020). Clinical studies of CTCL treatment showed that responders to the therapy had improved HRQoL, as well as improved skin and pruritus severity, suggesting that effective management and relief from pruritus impacts HRQoL. However, CTCL treatment guidelines focus on treating the disease itself and do not provide specific guidance on how to manage pruritus. 8 , 22

Pathophysiology of pruritus in CTCL

The exact mechanism that causes pruritus in patients with CTCL is not fully understood, so pruritus is often managed by treating the disease rather than by using specific anti‐itch treatments. The neuronal itch pathway can be stimulated by histamine, but itch in patients with CTCL is often unresponsive to treatment with anti‐histamine agents or topical corticosteroids and may be due to ‘histamine‐independent’ stimuli, 117 so other mediators must be considered.

There is evidence to suggest that itch in CTCL is related to inflammatory processes, with particular focus on IL‐31. 118 Nattkemper et al. (2016) 118 found that IL‐31 was significantly increased in the epidermis and lymphocytic infiltrate of patients with CTCL, and that patients with moderate‐to‐severe itch had the highest levels of IL‐31 in the skin where itch was evaluated using an itch‐specific tool, the VAS‐itch. IL‐31 levels did not correlate with disease stage, implying that IL‐31 is not directly involved in the pathogenesis of CTCL. However, a study in mice showed that repeated exposure to IL‐31 led to long‐lasting scratching, 119 implying that ongoing IL‐31 exposure is needed for pruritus to develop. Altogether, this suggests that the ongoing, increased levels of IL‐31 are a possible mechanism for pruritus in CTCL.

Pruritus therapies

Some therapies other than those that target the lymphoma itself have been effective in patients with CTCL. 117 For example, corticosteroids, both topical and oral formulations, can provide relief in some patients with CTCL. 120 Gabapentin, an anti‐convulsant, and mirtazapine, an anti‐depressant, have shown effectiveness in treating pruritus in patients with advanced‐stage MF/SS. 121 In addition, aprepitant blocks the binding of substance P to the neurokinin‐1 receptor and is approved for chemotherapy‐induced nausea and vomiting. Substance P localises to mast cells, but has also been shown to induce itch in mast cell‐deficient mice. 122 In a small case series of four patients with CTCL, 123 aprepitant improved pruritus, and a retrospective review involving 17 patients showed an ORR of 84% 124 ; however, large‐scale studies are needed to confirm these results. Naltrexone 125 and naloxone, both mu‐opioid receptor antagonists, have also been used. Currently, a Phase III clinical trial is evaluating the effectiveness of naloxone lotion in patients with CTCL (NCT02811783), with results expected in 2021.

Conclusions

CTCL is characterised by visible skin alterations, such as patches, plaques and tumours, and also causes alopecia, pruritus and pain. Patient assessment requires a multicompartmental approach, including assessment of the level of blood involvement, and the same approach should be used when examining patient response to help determine the effect of treatment on disease stage.

The disease also requires chronic treatment with a series of therapies, which can have significant side‐effects and often lose effectiveness, meaning patients may require multiple therapy changes. Patients with early‐stage disease have a good prognosis; however, a subpopulation with refractory disease has limited treatment options when SDTs are no longer effective. Patients with advanced‐stage disease have a poorer prognosis and therefore it is important to halt or slow disease progression as much as possible with improved duration of response and novel therapies. There is an unmet need for safe and effective therapies with good safety profiles for patients with both early‐ and late‐stage CTCL. However, the recent development of targeted immunotherapies with good PFS, such as mogamulizumab and brentuximab vedotin, is changing the way that CTCL is now being managed, particularly for refractory patients. Another avenue to explore is the efficacy of maintenance therapy in order to improve the duration of response to existing therapies.

Allo‐HSCT can offer a cure in some patients but comes at a price, including the likelihood of relapse, GvHD, or infection 15 , 62 , 80 and risk of death. 126 Further research is required into the optimal conditioning regimens and the best timing of HSCT following systemic therapy.

Cutaneous T‐cell lymphoma also significantly impacts patients’ HRQoL, which worsens as the disease advances. Patients experience itching and pain, as well as poor sleep, depression and anxiety. There are a variety of HRQoL measurement tools available, but the development of a CTCL‐specific tool would more accurately reflect the effect of the disease on patients’ lives. It is important that efforts be made to evaluate the patient perspective on their disease qualitatively and used to inform the development of any CTCL‐specific HRQoL tool.

Author contributions

All authors contributed equally to the planning, draft revisions and final approval of this manuscript.

Conflict of interest

Julia J. Scarisbrick receives honoraria from 4SC, Kyowa Kirin, Mallinckrodt, and Takeda; serves as a consultant/advisor for 4SC, Innate Pharma, Kyowa Kirin, Mallinckrodt, and Takeda; and receives research funding from Kyowa Kirin and Takeda. Martine Bagot has stock ownership in and patents/royalties with Innate Pharma, and serves as a consultant/advisor for Innate Pharma, Kyowa Kirin, and miRagen. Pablo L. Ortiz‐Romero serves as a consultant/advisor for 4SC, Actelion, Innate, Kyowa Kirin, miRagen, Ricordati Rare Diseases, and Takeda; has patents/royalties with PLCG1 mutation; and receives travel funding from Almirall, Janssen, Leo Pharma, and Novartis.

Acknowledgements

This work is supported by Kyowa Kirin, Inc. The authors would like to thank Emma Butterworth, PhD, of Excerpta Medica, for medical writing assistance, funded by Kyowa Kirin, Inc.

References

  • 1. Criscione VD, Weinstock MA. Incidence of cutaneous T‐cell lymphoma in the United States, 1973–2002. Arch Dermatol. 2007;143:854–9. [DOI] [PubMed] [Google Scholar]
  • 2. Pulitzer M. Cutaneous T‐cell lymphoma. Clin Lab Med. 2017;37:527–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. NCRAS CTCL Report . [cited 2020 Sep 19]. Available from: http://www.ncin.org.uk/view?rid=3275
  • 4. Krejsgaard T, Lindahl LM, Mongan NP, Waski MA, Litvinov IV, Iversen L, et al. Malignant inflammation in cutaneous T‐cell lymphoma – a hostile takeover. Semin Immunopathol. 2017;39:269–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Orphanet: Classic mycosis fungoides . [cited 2020 Sep 21]. Available from: https://www.orpha.net/consor/cgi‐bin/OC_Exp.php?Lng=GB&Expert=2584
  • 6. Orphanet: Sézary syndrome . [cited 2020 Sep 21]. Available from: https://www.orpha.net/consor/cgi‐bin/OC_Exp.php?Expert=3162
  • 7. Benton EC, Crichton S, Talpur R, Agar NS, Fields PA, Wedgeworth E, et al. A cutaneous lymphoma international prognostic index (CLIPi) for mycosis fungoides and Sezary syndrome. Eur J Cancer. 2013;49:2859–68. [DOI] [PubMed] [Google Scholar]
  • 8. Gilson D, Whittaker S, Child F, Scarisbrick JJ, Illidge TM, Parry EJ, et al. British Association of Dermatologists and U.K. Cutaneous Lymphoma Group guidelines for the management of primary cutaneous lymphomas 2018. Br J Dermatol. 2019;180:496–526. [DOI] [PubMed] [Google Scholar]
  • 9. Scarisbrick JJ, Prince H, Vermeer MH, Quaglino P, Horwitz S, Porcu P, et al. Cutaneous Lymphoma International Consortium study of outcome in advanced stages of mycosis fungoides and Sézary syndrome: effect of specific prognostic markers on survival and development of a prognostic model. J Clin Oncol. 2015;33:3766–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Kim YH, Willemze R, Pimpinelli N, Whittaker S, Olsen EA, Ranki A, et al. TNM classification system for primary cutaneous lymphomas other than mycosis fungoides and Sézary syndrome: a proposal of the International Society for Cutaneous Lymphomas (ISCL) and the Cutaneous Lymphoma Task Force of the European Organization of Research and Treatment of Cancer (EORTC). Blood. 2007;110:479–84. [DOI] [PubMed] [Google Scholar]
  • 11. Olsen EA, Kim YH, Kuzel TM, Pacheco TR, Foss FM, Parker S, 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. [DOI] [PubMed] [Google Scholar]
  • 12. Fraser‐Andrews EA, Woolford AJ, Russell‐Jones R, Seed PT, Whittaker SJ. Detection of a peripheral blood T cell clone is an independent prognostic marker in mycosis fungoides. J Investig Dermatol. 2000;114:117–21. [DOI] [PubMed] [Google Scholar]
  • 13. Kim YH, Liu HL, Mraz‐Gernhard S, Verghese A, Hoppe RT. Long‐term outcome of 525 patients with mycosis fungoides and Sezary syndrome: clinical prognostic factors and risk for disease progression. Arch Dermatol. 2003;139:857–66. [DOI] [PubMed] [Google Scholar]
  • 14. NCCN T‐Cell Lymphomas Guidelines . [cited 2020 Sep 19]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/t‐cell.pdf
  • 15. Ritchie S, Qureshi I, Molloy K, Yoo J, Shah F, Stevens A, et al. Evaluation of haematopoietic stem cell transplantation in patients diagnosed with cutaneous T‐cell lymphoma at a tertiary care centre: should we avoid chemotherapy in conditioning regimes? Br J Dermatol. 2020;182:807–9. [DOI] [PubMed] [Google Scholar]
  • 16. Schmitz N, Lenz G, Stelljes M. Allogeneic hematopoietic stem cell transplantation for T‐cell lymphomas. Blood. 2018;132:245–53. [DOI] [PubMed] [Google Scholar]
  • 17. Shalabi D, Bistline A, Alpdogan O, Kartan S, Mishra A, Porcu P, et al. Immune evasion and current immunotherapy strategies in mycosis fungoides (MF) and Sézary syndrome (SS). Chin Clin Oncol. 2019;8:11. [DOI] [PubMed] [Google Scholar]
  • 18. Hristov AC, Trilokraj T, Wilcox RA. Mycosis fungoides and Sézary syndrome: 2019 update on diagnosis, risk‐stratification, and management. Am J Hematol. 2019;94:1027–41. [DOI] [PubMed] [Google Scholar]
  • 19. Nccn guidelines. [cited 2020 Sep 22]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/primary_cutaneous.pdf
  • 20. Trautinger F, Eder J, Assaf C, Bagot M, Cozzio A, Dummer R, et al. European Organisation for Research and Treatment of Cancer consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome – update 2017. Eur J Cancer. 2017;77:57–74. [DOI] [PubMed] [Google Scholar]
  • 21. Kempf W, Pfaltz K, Vermeer MH, Cozzio A, Ortiz‐Romero PL, Bagot M, et al. EORTC, ISCL, and USCLC consensus recommendations for the treatment of primary cutaneous CD30‐positive lymphoproliferative disorders: lymphomatoid papulosis and primary cutaneous anaplastic large‐cell lymphoma. Blood. 2011;118:4024–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Willemze R, Hodak E, Zinzani PL, Specht L, Ladetto M, ESMO Guidelines Committee . Primary cutaneous lymphomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up. Ann Oncol. 2018;29:iv30–iv40. [DOI] [PubMed] [Google Scholar]
  • 23. Adcetris Product Information . [cited 2020 Sep 19]. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/adcetris
  • 24. Brentuximab vedotin Orphan Designation . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/medicines/human/orphan‐designations/eu311939
  • 25. Ledaga EPAR Summary . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/medicines/human/EPAR/ledaga
  • 26. Mogamulizumab Orphan Designation . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/medicines/human/orphan‐designations/eu3161756
  • 27. Poteligeo Product Information . [cited 2020 Sep 19]. Available from: https://www.ema.europa.eu/en/documents/product‐information/poteligeo‐epar‐product‐information_en.pdf
  • 28. Roferon‐A Patient Information Leaflet. [cited 2020 Sep 19]. Available from: www.medicines.org.uk/emc/product/1117
  • 29. Targretin EPAR Medicine Overview . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/documents/overview/targretin‐epar‐medicine‐overview_en.pdf
  • 30. Cristofoletti C, Narducci MG, Russo G. Sézary syndrome, recent biomarkers and new drugs. Chin Clin Oncol. 2019;8:2. [DOI] [PubMed] [Google Scholar]
  • 31. Green SB, Byar DP, Lamberg SI. Prognostic variables in mycosis fungoides. Cancer. 1981;47:2671–7. [DOI] [PubMed] [Google Scholar]
  • 32. Lebon B, Beynon TA, Whittaker SJ. Palliative care in patients with primary cutaneous lymphoma: symptom burden and characteristics of hospital palliative care team input. Arch Dermatol. 2007;143:423–4. [DOI] [PubMed] [Google Scholar]
  • 33. Sampogna F, Frontani M, Baliva G, Lombardo GA, Alvetreti G, Di Pietro C, et al. Quality of life and psychological distress in patients with cutaneous lymphoma. Br J Dermatol. 2009;160:815–22. [DOI] [PubMed] [Google Scholar]
  • 34. Demierre MF, Tien A, Miller D. Health‐related quality‐of‐life assessment in patients with cutaneous T‐cell lymphoma. Arch Dermatol. 2005;141:325–30. [DOI] [PubMed] [Google Scholar]
  • 35. Demierre MF, Gan S, Jones J, Miller DR. Significant impact of cutaneous T‐cell lymphoma on patients’ quality of life: results of a 2005 national cutaneous lymphoma foundation survey. Cancer. 2006;107:2504–11. [DOI] [PubMed] [Google Scholar]
  • 36. Wright A, Wijeratne A, Hung T, Gao W, Whittaker S, Morris S, et al. Prevalence and severity of pruritus and quality of life in patients with cutaneous T‐cell lymphoma. J Pain Symptom Manag. 2013;45:114–9. [DOI] [PubMed] [Google Scholar]
  • 37. Molloy K, Jonak C, Woei‐A‐Ji FJ, Guenova E, Busschots AM, Bervoets A, et al. Characteristics associated with significantly worse quality of life in mycosis fungoides/Sézary syndrome from the Prospective Cutaneous Lymphoma International Prognostic Index (PROCLIPI) study. Br J Dermatol. 2020;182:770–9. [DOI] [PubMed] [Google Scholar]
  • 38. Herbosa CM, Semenov YR, Rosenberg AR, Mehta‐Shah N, Musiek AC. Clinical severity measures and quality of life burden in patients with mycosis fungoides and Sézary syndrome: comparison of generic and dermatology‐specific instruments. J Eur Acad Dermatol Venereol. 2020;34:995–1003. [DOI] [PubMed] [Google Scholar]
  • 39. Orlowska D, Selman LE, Beynon T, Radcliffe E, Whittaker S, Child F, et al. ‘It’s a traumatic illness, traumatic to witness’: a qualitative study of the experiences of bereaved family caregivers of patients with cutaneous T‐cell lymphoma. Br J Dermatol. 2018;179:882–8. [DOI] [PubMed] [Google Scholar]
  • 40. Williams K, Gibson A, McNamara L, Jones T, Llyod AJ. Health state utilities associated with caring for an individual with cutaneous T‐cell lymphoma (CTCL). J Med Econ. 2020. [Online ahead of print]. 10.1080/13696998.2020.1793764 [DOI] [PubMed] [Google Scholar]
  • 41. Adcetris Prescribing Information . [cited 2020 Sep 19]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125388s100lbl.pdf
  • 42. Alpdogan O, Kartan S, Johnson W, Sokol K, Porcu P. Systemic therapy of cutaneous T‐cell lymphoma (CTCL). Chin Clin Oncol. 2019;8:20. [DOI] [PubMed] [Google Scholar]
  • 43. Atadja P. Development of the pan‐DAC inhibitor panobinostat (LBH589): successes and challenges. Cancer Lett. 2009;280:233–41. [DOI] [PubMed] [Google Scholar]
  • 44. Bernengo MG, Quaglino P, Comessatti A, Ortoncelli M, Novelli M, Lisa F, et al. Low‐dose intermittent alemtuzumab in the treatment of Sézary Syndrome: clinical and immunologic findings in 14 patients. Haematologica. 2007;92:784–94. [DOI] [PubMed] [Google Scholar]
  • 45. Chang TP, Poltoratsky V, Vancurova I. Bortezomib inhibits expression of TGF‐β1, IL‐10, and CXCR4, resulting in decreased survival and migration of cutaneous T cell lymphoma cells. J Immunol. 2015;194:2942–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Dummer R, Quaglino P, Becker JC, Hasan B, Karrasch M, Whittaker S, et al. Prospective international multicenter phase II trial of intravenous pegylated liposomal doxorubicin monochemotherapy in patients with stage IIB, IVA, or IVB advanced mycosis fungoides: final results from EORTC 21012. J Clin Oncol. 2012;30:4091–7. [DOI] [PubMed] [Google Scholar]
  • 47. Duvic M, Dummer R, Becker JC, Poulalhon N, Romero NO, Bernengo MG, et al. Panobinostat activity in both bexarotene‐exposed and ‐naïve patients with refractory cutaneous T‐cell lymphoma: results of a phase II trial. Eur J Cancer. 2013;49:386–94. [DOI] [PubMed] [Google Scholar]
  • 48. Eichenfield LF, Beck L. Elidel (Pimecrolimus) cream 1%: a nonsteroidal topical agent for the treatment of atopic dermatitis. J Allergy Clin Immunol. 2003;111:1153–68. [DOI] [PubMed] [Google Scholar]
  • 49. Khodadoust M, Rook A, Procu P, Foss F, Moskowitz AJ, Shustov A, et al. Pembrolizumab in relapsed and refractory Mycosis Fungoides and Sézary Syndrome: a multicenter phase II study. J Clin Oncol. 2020;38:20–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Kiessling MK, Klemke CD, Kaminski MM, Galani IE, Krammer PH, Gülow K. inhibition of constitutively activated nuclear factor‐κB induces reactive oxygen species‐ and iron‐dependent cell death in cutaneous T‐cell lymphoma. Cancer Res. 2009;69:2365–74. [DOI] [PubMed] [Google Scholar]
  • 51. Lowenstein H, Shah A, Chant A, Khan A. Different mechanisms of Campath‐1H‐mediated depletion for CD4+ and CD8+ T cells in peripheral blood. Transplant Int. 2006;19:927–36. [DOI] [PubMed] [Google Scholar]
  • 52. Nicolay JP, Muller‐Decker K, Schroeder A, Brechmann M, Möbs M, Géraud C, et al. Dimethyl fumarate restores apoptosis sensitivity and inhibits tumor growth and metastasis in CTCL by targeting NF‐kB. Blood. 2016;128:805–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. NIHR Innovation Observatory Evidence Briefing . Synthetic hypericin (SGX301; VIMRxyn) for cutaneous T‐cell lymphoma, unspecified; 2017. [cited 2020 Sep 19]. Available from: www.io.nihr.ac.uk/wp‐content/uploads/migrated_new/10761‐Synthetic‐Hypericin.pdf
  • 54. Poteligeo Prescribing Information . [cited 2020 Sep 19]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761051s000lbl.pdf
  • 55. Querfeld C, Rosen ST, Guitart J, Duvic M, Kim YH, Dusza SW, et al. Results of an open‐label multicenter phase 2 trial of lenalidomide monotherapy in refractory mycosis fungoides and Sézary syndrome. Blood. 2014;123:1159–66. [DOI] [PubMed] [Google Scholar]
  • 56. Rook AH, Gelfand JM, Wysocka M, Troxel AB, Benoit B, Surber C, et al. Topical resiquimod can induce disease regression and enhance T‐cell effector functions in cutaneous T‐cell lymphoma. Blood. 2015;126:1452–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. T‐cell Lymphoma Forum Industry Innovations miRagen MicroRNA Oral Presentation. Cobomarsen, an emerging potential treatment for patients with miR‐155 elevated cancers. [cited 2020 Sep 19]. Available from: www.miragen.com/publications/2019/02/T‐cell‐Lymphoma‐Forum‐Industry‐Innovations‐miRagen‐MicroRNA‐Oral‐Presentation.pdf
  • 58. Vieyra‐Garcia P, Fink‐Puches R, Porkert S, Lang R, Pöchlauer S, Ratzinger G, et al. Evaluation of low‐dose, low‐frequency oral psoralen–UV‐a treatment with or without maintenance on early‐stage mycosis fungoides: a randomized clinical trial. JAMA Dermatol. 2019;155:538–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Zinzani PL, Musuraca G, Tani M, Stefoni V, Marchi E, Fina M, et al. Phase II trial of proteasome inhibitor bortezomib in patients with relapsed or refractory cutaneous T‐cell lymphoma. J Clin Oncol. 2007;25:4293–7. [DOI] [PubMed] [Google Scholar]
  • 60. Quaglino P, Maule M, Prince HM, Porcu P, Horwitz S, Duvic M, et al. Global patterns of care in advanced stage mycosis fungoides/sezary syndrome a multicenter retrospective follow‐up study from the Cutaneous Lymphoma International Consortium. Ann Oncol. 2017;28:2517–25. [DOI] [PubMed] [Google Scholar]
  • 61. Trautinger F, Knobler R, Willemze R, Peris K, Stadler R, Laroche L, et al. EORTC consensus recommendations for the treatment of mycosis fungoides/sézary syndrome. Eur J Cancer. 2006;42:1014–30. [DOI] [PubMed] [Google Scholar]
  • 62. Johnson WT, Mukherji R, Kartan S, Nikbakht N, Porcu P, Alpdogan O. Allogeneic hematopoietic stem cell transplantation in advanced stage mycosis fungoides and Sézary syndrome: a concise review. Chin Clin Oncol. 2019;8:12. [DOI] [PubMed] [Google Scholar]
  • 63. Litvinov IV, Jones DA, Sasseville D, Kupper TS. Transcriptional profiles predict disease outcome in patients with cutaneous t‐cell lymphoma. Clin Cancer Res. 2010;16:2106–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Amorim GM, Corbellini JPN, Quintella DC, Cuzzi T, Ramos‐E‐Silva M. Evaluation of the Cutaneous Lymphoma International Prognostic Index in patients with early stage mycosis fungoides. An Bras Dermatol. 2018;93:680–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Talpur R, Singh L, Daulat S, Liu P, Seyfer S, Trynosky T, et al. Long‐term outcomes of 1,263 patients with mycosis fungoides and sezary syndrome from 1982 to 2009. Clin Cancer Res. 2012;18:5051–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Scarisbrick JJ, Kim YH, Whittaker SJ, Wood GS, Vermeer MH, Prince HM, et al. Prognostic factors, prognostic indices and staging in mycosis fungoides and Sezary syndrome: where are we now? Br J Dermatol. 2014;170:1226–36. [DOI] [PubMed] [Google Scholar]
  • 67. Jawed SI, Myskowski PL, Horwitz S, Moskowitz A, Querfeld C. Primary cutaneous T‐cell lymphoma (mycosis fungoides and Sézary syndrome): part II. Prognosis, management, and future directions. J Am Acad Dermatol. 2014;70:223.e1–17. [DOI] [PubMed] [Google Scholar]
  • 68. Tarabadkar ES, Shinohara MM. Skin directed therapy in cutaneous T‐cell lymphoma. Front Oncol. 2019;9:260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Revlimid Prescribing Information . [cited 2020 Sep 19]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021880s049lbl.pdf
  • 70. Mühleisen L, Alev M, Unterweger H, Subatzus D, Pöttler M, Friedrich RP, et al. Analysis of hypericin‐mediated effects and implications for targeted photodynamic therapy. Int J Mol Sci. 2017;18:1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Rook AH, Wood GS, Duvic M, Vonderheid EC, Tobia A, Cabana B. A phase II placebo‐controlled study of photodynamic therapy with topical hypericin and visible light irradiation in the treatment of cutaneous T‐cell lymphoma and psoriasis. J Am Acad Dermatol. 2010;63:984–90. [DOI] [PubMed] [Google Scholar]
  • 72. Stern RS. The risk of melanoma in association with long‐term exposure to PUVA. J Am Acad Dermatol. 2001;44:755–61. [DOI] [PubMed] [Google Scholar]
  • 73. Ohmachi K, Ando K, Ogura M, Uchida T, Tobinai K, Maruyama D, et al. E7777 in Japanese patients with relapsed/refractory peripheral and cutaneous T‐cell lymphoma: a phase I study. Cancer Sci. 2018;109:794–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Lansigan F, Stearn DM, Foss F. Role of denileukin diftitox in the treatment of persistent or recurrent cutaneous T‐cell lymphoma. Cancer Manag Res. 2010;2:53–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Olsen E, Duvic M, Frankel A, Kim Y, Martin A, Vonderheid E, et al. Pivotal phase III trial of two dose levels of denileukin diftitox for the treatment of cutaneous T‐cell lymphoma. J Clin Oncol. 2001;19:376–88. [DOI] [PubMed] [Google Scholar]
  • 76. Prince HM, Duvic M, Martin A, Sterry W, Assaf C, Sun Y, et al. Phase III placebo‐controlled trial of denileukin diftitox for patients with cutaneous T‐cell lymphoma. J Clin Oncol. 2010;28:1870–7. [DOI] [PubMed] [Google Scholar]
  • 77. Prince HM, Martin AG, Olsen EA, Fivenson DP, Duvic M. Denileukin diftitox for the treatment of CD25 low‐expression mycosis fungoides and Sézary syndrome. Leuk Lymphoma. 2013;54:69–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Wang Z, Zheng Q, Zhang H, Bronson RT, Madsen JC, Sachs DH, et al. Ontak‐like human IL‐2 fusion toxin. J Immunol Methods. 2017;448:51–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Carter J, Zug KA. Phototherapy for cutaneous T‐cell lymphoma: online survey and literature review. J Am Acad Dermatol. 2009;60:39–50. [DOI] [PubMed] [Google Scholar]
  • 80. Lechowicz MJ, Lazarus HM, Carreras J, Laport GG, Cutler CS, Wiernik PH, et al. Allogeneic hematopoietic cell transplantation for mycosis fungoides and Sezary syndrome. Bone Marrow Transplant. 2014;49:1360–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Virmani P, Zain J, Rosen ST, Myskowski PL, Querfeld C. Hematopoietic stem cell transplant for mycosis fungoides and sézary syndrome. Dermatol Clin. 2015;33:807–18. [DOI] [PubMed] [Google Scholar]
  • 82. Hughes CF, Khot A, McCormack C, Lade S, Westerman DA, Twigger R, et al. Lack of durable disease control with chemotherapy for mycosis fungoides and Sézary syndrome: a comparative study of systemic therapy. Blood. 2015;125:71–81. [DOI] [PubMed] [Google Scholar]
  • 83. Annunziata CM, Davis RE, Demchanko Y, Bellamy W, Gabrea A, Zhan F, et al. Frequent engagement of the classical and alternative NF‐κB pathways by diverse genetic abnormalities in multiple myeloma. Cancer Cell. 2007;12:115–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bagot M, Porcu P, Cardine AM, Battistella M, William BM, Vermeer M, 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. [DOI] [PubMed] [Google Scholar]
  • 85. Bassères DS, Baldwin AS. Nuclear factor‐κB and inhibitor of κB kinase pathways in oncogenic initiation and progression. Oncogene. 2006;25:6817–30. [DOI] [PubMed] [Google Scholar]
  • 86. Ikeda M, Ohno I, Ueno H, Mitsunaga S, Hashimoto Y, Okusaka T, et al. Phase I study of resminostat, an HDAC inhibitor, combined with S‐1 in patients with pre‐treated biliary tract or pancreatic cancer. Investig New Drugs. 2019;37:109–17. [DOI] [PubMed] [Google Scholar]
  • 87. Lundin J, Hagberg H, Repp R, Cavallin‐Ståhl E, Fredén S, Juliusson G, et al. Phase 2 study of alemtuzumab (anti‐CD52 monoclonal antibody) in patients with advanced mycosis fungoides/Sézary syndrome. Blood. 2003;101:4267–72. [DOI] [PubMed] [Google Scholar]
  • 88. Pulini S, Rupoli S, Goteri G, Pimpinelli N, Alterini R, Tassetti A, et al. Pegylated liposomal doxorubicin in the treatment of primary cutaneous T‐cell lymphomas. Haematologica. 2007;92:686–9. [DOI] [PubMed] [Google Scholar]
  • 89. Quereux G, Marques S, Nguyen JM, Bedane C, D'incan M, Dereure O, 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. [DOI] [PubMed] [Google Scholar]
  • 90. Shao W, Growney JD, Feng Y, O’Connor G, Pu M, Zhu W, et al. Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T‐cell lymphoma models: defining molecular mechanisms of resistance. Int J Cancer. 2010;127:2199–208. [DOI] [PubMed] [Google Scholar]
  • 91. Skilarence Product Information . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/documents/product‐information/skilarence‐epar‐product‐information_en.pdf
  • 92. Tak WY, Ryoo BY, Lim HY, Kim DY, Okusaka T, Ikeda M, et al. Phase I/II study of first‐line combination therapy with sorafenib plus resminostat, an oral HDAC inhibitor, versus sorafenib monotherapy for advanced hepatocellular carcinoma in east Asian patients. Investig New Drugs. 2018;36:1072–84. [DOI] [PubMed] [Google Scholar]
  • 93. Tambo Y, Hosomi Y, Sakai H, Nogami N, Atagi S, Sasaki Y, et al. Phase I/II study of docetaxel combined with resminostat, an oral hydroxamic acid HDAC inhibitor, for advanced non‐small cell lung cancer in patients previously treated with platinum‐based chemotherapy. Investig New Drugs. 2017;35:217–26. [DOI] [PubMed] [Google Scholar]
  • 94. Tecfidera Product Information . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/documents/product‐information/tecfidera‐epar‐product‐information_en.pdf
  • 95. Walewski J, Paszkiewicz‐Kozik E, Borsaru G, Hellmann A, Janikova A, Warszewska A, et al. Resminostat in patients with relapsed or refractory Hodgkin lymphoma: results of the phase II SAPHIRE study. Leuk Lymphoma. 2019;60:675–84. [DOI] [PubMed] [Google Scholar]
  • 96. Weston VJ, Austen B, Wei W, Marston E, Alvi A, Lawson S, et al. Apoptotic resistance to ionizing radiation in pediatric B‐precursor acute lymphoblastic leukemia frequently involves increased NF‐κB survival pathway signaling. Blood. 2004;104:1465–73. [DOI] [PubMed] [Google Scholar]
  • 97. Wollina U, Dummer R, Brockmeyer NH, Konrad H, Busch JO, Kaatz M, et al. Multicenter study of pegylated liposomal doxorubicin in patients with cutaneous T‐cell lymphoma. Cancer. 2003;98:993–1001. [DOI] [PubMed] [Google Scholar]
  • 98. Willemze R, Jaffe ES, Burg G, Cerroni L, Berti E, Swerdlow SH, et al. WHO‐EORTC classification for cutaneous lymphomas. Blood. 2005;105:3768–85. [DOI] [PubMed] [Google Scholar]
  • 99. Prince HM, Kim Y, Horwitz S, Dummer R, Scarisbrick J, Quaglino P, et al. Brentuximab vedotin or physician's choice in CD30‐positive cutaneous T‐cell lymphoma (ALCANZA): an international, open‐label, randomised, phase 3, multicentre trial. Lancet. 2017;390:555–66. [DOI] [PubMed] [Google Scholar]
  • 100. Scarisbrick JJ, Horwitz SM, Prince HM, Whittaker S, Duvic M, Kim YH, et al. Brentuximab vedotin (BV) versus physician’s choice (PC) of methotrexate or bexarotene in adult patients with previously treated CD30‐positive cutaneous T‐cell lymphoma (CTCL; mycosis fungoides [MF] or primary cutaneous anaplastic large cell lymphoma [pcALCL]): final time to next therapy (TTNT) results from the phase 3 ALCANZA study. Eur J Cancer. 2019;119:S31. [Google Scholar]
  • 101. Engelina S, Saggu M, Yoo J, Shah F, Stevens A, Irwin C, et al. Brentuximab a novel antibody therapy: real‐world use confirms efficacy and tolerability for CD30‐positive cutaneous lymphoma. Br J Dermatol. 2020;182:799–800. [DOI] [PubMed] [Google Scholar]
  • 102. Ollila TA, Sahin I, Olszewski AJ. Mogamulizumab: a new tool for management of cutaneous T‐cell lymphoma. OncoTargets Ther. 2019;12:1085–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Ishida T, Ueda R. CCR4 as a novel molecular target for immunotherapy of cancer. Cancer Sci. 2006;97:1139–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Kim YH, Bagot M, Pinter‐Brown L, Rook AH, Porcu P, Horwitz SM, 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. [DOI] [PubMed] [Google Scholar]
  • 105. Pro B, Kim YH, Ortiz‐Romero PL, Sokol L, Scarisbrick J, Musiek A, et al. Time to next treatment in patients with previously treated cutaneous T‐cell lymphoma (CTCL) receiving mogamulizumab or vorinostat: a MAVORIC post‐hoc analysis. J Clin Oncol. 2019;37:7539. [Google Scholar]
  • 106. Kim YH, Bagot M, Zinzani PL, Duvic M, Morris S, Kim E, et al. Safety of mogamulizumab in mycosis fungoides and Sézary Syndrome: final results from the phase 3 MAVORIC study. Blood. 2019;134:5300. [Google Scholar]
  • 107. Kyowa Kirin Press Release . Approval for Additional Indication for PTCL and CTCL of Mogamulizumab; 2018. [cited 2020 Sep 19]. Available from: https://www.kyowakirin.com/media_center/news_releases/2018/e20180821_01.html
  • 108. Humanised IgG1 monoclonal antibody against human KIR3DL2 for the treatment of cutaneous T‐cell lymphoma Orphan Designation . [cited 2020 Sep 19]. Available from: www.ema.europa.eu/en/documents/orphan‐designation/eu/3/14/1322‐public‐summary‐opinion‐orphan‐designation‐humanised‐igg1‐monoclonal‐antibody‐against‐human_en.pdf
  • 109. Marie‐Cardine A, Viaud N, Thonnart N, Joly R, Chanteux S, Gauthier L, et al. IPH4102, a humanized KIR3DL2 antibody with potent activity against cutaneous T‐cell lymphoma. Cancer Res. 2014;74:6060–70. [DOI] [PubMed] [Google Scholar]
  • 110. Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, et al. Phase 2 trial of oral vorinostat (Suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T‐cell lymphoma (CTCL). Blood. 2007;109:31–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Whittaker SJ, Demierre MF, Kim EJ, Rook AH, Lerner A, Duvic M, 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. [DOI] [PubMed] [Google Scholar]
  • 112. Jonak C, Porkert S, Oerlemans S, Papadavid E, Molloy K, Lehner‐Baumgartner E, et al. Health‐related quality of life in cutaneous lymphomas: past, present and future. Acta Derm Venereol. 2019;99:640–6. [DOI] [PubMed] [Google Scholar]
  • 113. Vij A, Duvic M. Prevalence and severity of pruritus in cutaneous T cell lymphoma. Int J Dermatol. 2012;51:930–4. [DOI] [PubMed] [Google Scholar]
  • 114. Porkert S, Lehner‐Baumgartner E, Valencak J, Knobler R, Riedl E, Jonak C. Patients’ illness perception as a tool to improve individual disease management in primary cutaneous lymphomas. Acta Derm Venereol. 2018;98:240–5. [DOI] [PubMed] [Google Scholar]
  • 115. Quaglino P, Hudgens S, Porcu P, Cowan R, Floden L, Tsianakas A, et al. Quality of life in cutaneous T‐cell lymphoma subjects treated with the anti‐CCR4 monoclonal antobody mogamulizumab versus vorinostat: results from the Phase 3 MAVORIC Trial. HemaSphere. 2018;2:85 10.1097/HS9.0000000000000060 [DOI] [Google Scholar]
  • 116. McCaffrey S, Black RA, Nagao M, Sepassi M, Sharma G, Thronton S, et al. Measurement of quality of life in patients with mycosis fungoides/Sézary syndrome cutaneous T‐cell lymphoma: development of an electronic instrument. J Med Internet Res. 2019;21:e11302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Ahern K, Gilmore ES, Poligone B. Pruritus in cutaneous T‐cell lymphoma: a review. J Am Acad Dermatol. 2012;67:760–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Nattkemper LA, Martinez‐Escala M, Gelman LB, Singer EM, Rook AH, Guitart J, et al. Cutaneous T‐cell lymphoma and pruritus: the expression of IL‐31 and its receptors in the skin. Acta Derm Venereol. 2016;96:894–8. [DOI] [PubMed] [Google Scholar]
  • 119. Arai I, Tsuji M, Miyagawa K, Takeda H, Akiyama N, Saito S. Repeated administration of IL‐31 upregulates IL‐31 receptor A (IL‐31RA) in dorsal root ganglia and causes severe itch‐associated scratching behaviour in mice. Exp Dermatol. 2015;24:75–8. [DOI] [PubMed] [Google Scholar]
  • 120. Meyer N, Paul C, Misery L. Pruritus in cutaneous T‐cell lymphomas: frequent, often severe and difficult to treat. Acta Derm Venereol. 2010;90:12–7. [DOI] [PubMed] [Google Scholar]
  • 121. Demierre MF, Taverna J. Mirtazapine and gabapentin for reducing pruritus in cutaneous T‐cell lymphoma. J Am Acad Dermatol. 2006;55:543–4. [DOI] [PubMed] [Google Scholar]
  • 122. Andoh T, Nagasawa T, Satoh M, Kuraishi Y. Substance P induction of itch‐associated response mediated by cutaneous NK1 tachykinin receptors in mice. J Pharmacol Exp Therap. 1998;286:1140–5. [PubMed] [Google Scholar]
  • 123. Song JS, Tawa M, Chau NG, Kupper TS, LeBoeuf NR. Aprepitant for refractory cutaneous T‐cell lymphoma‐associated pruritus: 4 cases and a review of the literature. BMC Cancer. 2017;17:200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Maroñas‐Jiménez L, Estrach T, Gallardo F, Pérez A, Andrés Borja H, Servitje O, et al. Aprepitant improves refractory pruritus in primary cutaneous T‐cell lymphomas: experience of the Spanish Working Group on Cutaneous Lymphomas. Br J Dermatol. 2018;178:e273–e274. [DOI] [PubMed] [Google Scholar]
  • 125. Brune A, Metze D, Luger TA, Ständer S. Antipruritic therapy with the oral opioid receptor antagonist naltrexone. Open, non‐placebo controlled administration in 133 patients. Der Hautarzt. 2004;55:1130–6. [DOI] [PubMed] [Google Scholar]
  • 126. Duarte RF, Boumendil A, Onida F, Gabriel I, Arranza R, Arcese W, et al. Long‐term outcome of allogeneic hematopoietic cell transplantation for patients with mycosis fungoides and Sézary syndrome: a European society for Blood and Marrow Transplantation Lymphoma Working Party extended analysis. J Clin Oncol. 2014;32:3347–8. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Haematology are provided here courtesy of Wiley

RESOURCES