You are on page 1of 31

BJD

GUIDELINES British Journal of Dermatology

British Association of Dermatologists and U.K. Cutaneous


Lymphoma Group guidelines for the management of primary
cutaneous lymphomas 2018
D. Gilson,1 S.J. Whittaker iD ,2 F.J. Child,2 J.J. Scarisbrick iD ,3 T.M. Illidge iD ,4 E.J. Parry,5 M.F. Mohd Mustapa,6
L.S.Exton,6 E. Kanfer,7 K. Rezvani,8 C.E. Dearden iD 9 and S.L. Morris10
1
Leeds Cancer Centre, St James’s University Hospital, Leeds LS9 7TF, U.K.
2
St John’s Institute of Dermatology, Guy’s and St Thomas NHS Foundation Trust, St Thomas’ Hospital, London SE1 7EH, U.K.
3
Queen Elizabeth Hospital, University Hospital Birmingham, Birmingham B15 2TH, U.K.
4
Institute of Cancer Sciences, University of Manchester, The Christie NHS Foundation Trust, Manchester M20 4BX, U.K.
5
Tameside Hospital Integrated Care NHS Foundation Trust, Ashton-under-Lyne OL6 9RW, U.K.
6
British Association of Dermatologists, Willan House, 4 Fitzroy Square, London, W1T 5HQ, U.K.
7
Haematology Department, Hammersmith Hospital, Du Cane Road, London W12 0HS, U.K.
8
The University of Texas MD Anderson Cancer Centre, Houston, TX, U.S.A.
9
Chronic Lymphocytic Leukaemia (CLL) Unit, The Royal Marsden NHS Foundation Trust, Sutton SW3 6JJ, U.K.
10
Guy’s and St Thomas’ NHS Foundation Trust, Guy’s Hospital, London SE1 9RT, U.K.

Linked Editorial: Wang and Bagot. Br J Dermatol 2019; 180:

Correspondence 1.0 Purpose and scope


Sean Whittaker. The overall objective of the guideline is to provide up-to-date,
E-mails: sean.whittaker@kcl.ac.uk; guidelines@bad.org.uk
evidence-based recommendations on the management of pri-
mary cutaneous lymphoma in the U.K. The document aims
Accepted for publication
14 September 2018
to: (i) offer an appraisal of all relevant literature up to Febru-
ary 2018 focusing on any key developments; (ii) address
Funding sources important, practical clinical questions relating to the primary
None. guideline objective; (iii) provide guideline recommendations
with, where appropriate, some health economic implications;
Conflicts of interest and (iv) discuss potential developments and future directions.
S.J.W. has been on advisory boards for Actelion, Takeda and Innate Pharma (specific) and has The guideline is presented as a detailed review with high-
a collaborative research agreement with Galderma. J.J.S. has been a consultant for Innate lighted recommendations for practical use in the clinic (see
Pharma, Actelion Pharmaceuticals, Therakos (Mallinckrodt), Takeda and 4SC (specific) and section 12). A patient information leaflet on mycosis fun-
has received honoraria and meeting sponsorship from Teva, Therakos, Johnson & Johnson and
goides is available on the British Association of Dermatologists
Takeda (specific). C.E.D. has been a consultant for Genzyme (specific). T.M.I. has been a con-
(BAD) website: www.bad.org.uk/for-the-public/patient-inf
sultant and invited speaker for Takeda (specific). E.J.P. has been on the advisory board for and
ormation-leaflets. Further information can also be found at
an invited speaker for Actelion. The other authors declare no conflicts of interest.
www.lymphomas.org.uk/about-lymphoma.
D.G., S.J.W., F.J.C., J.J.S., T.M.I., E.J.P., E.K., K.R., C.E.D. and S.L.M. are members of the
guideline development group, with technical support provided by L.S.E. and M.F.M.M.
1.1 Exclusions
These guidelines were first produced jointly by the British Association of Dermatologists and the
U.K. Cutaneous Lymphoma Group (2003); reviewed and updated 2018. These guidelines do not cover patients presenting with or found
This is an updated guideline prepared for the BAD Clinical Standards Unit, which includes the to have skin involvement as part of a systemic lymphoma.
Therapy & Guidelines Subcommittee. Members of the Clinical Standards Unit who have been
involved are P.M. McHenry (Chairman Therapy & Guidelines), T.A. Leslie, S. Wakelin, R.Y.P.
Hunasehally, M. Cork, G.A. Johnston, N. Chiang, F.S. Worsnop, A. Salim, D. Buckley, G. Petrof, 2.0 Methodology
N. Callachand (British National Formulary), T. Flavell (British Dermatological Nursing Group),
A.A. Salad (BAD Scientific Administrator), L.S. Exton (BAD Guideline Research Fellow) and M.F. This set of guidelines has been developed using the BAD’s recom-
Mohd Mustapa (BAD Clinical Standards Manager). mended methodology1 and with reference to the Appraisal of
DOI 10.1111/bjd.17240
Guidelines Research and Evaluation (AGREE II) instrument
(www.agreetrust.org).2 The recommendations were developed
NICE has renewed accreditation of the process used by the British for implementation in the National Health Service using a process
Association of Dermatologists to produce clinical guidelines. The
renewed accreditation is valid until 31 May 2021 and applies to of considered judgement based on the evidence (Appendix S1;
guidance produced using the processes described in the updated guid-
ance for writing a British Association of Dermatologists clinical see Supporting Information). Targeted literature searches were
guideline – the adoption of the GRADE methodology 2016. The
original accreditation term began on 12 May 2010. More infor- carried out in the PubMed, MEDLINE and Embase databases and
mation on accreditation can be viewed at www.nice.org.uk/accredi
tation. the Cochrane Library, for meta-analyses, randomized and

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 1


2 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

nonrandomized controlled clinical trials, case series, case reports, Table 1 World Health Organization/European Organisation for
open studies and cohort studies on primary cutaneous lymphoma Research and Treatment of Cancer classification of primary cutaneous
and specified treatments to February 2018. The search terms and lymphomas (italics indicate provisional entities)
strategies are detailed in Appendix S2 (see Supporting Informa-
Cutaneous B-cell lymphoma
tion). Additional relevant references were also retrieved from
• Indolent clinical behaviour
citations in the reviewed literature. ○ Primary cutaneous marginal zone B-cell lymphomaa
All identified English-language titles were screened and those ○ Primary cutaneous follicle centre lymphoma
relevant for first-round inclusion were selected for further scrutiny.
• Intermediate behaviour
The abstracts for the shortlisted references were then reviewed and
the full papers of relevant material were obtained; disagreements in
○ Primary cutaneous diffuse large B-cell
lymphoma, leg type
the final selections were resolved by discussion with the entire Cutaneous T-cell lymphoma
Guideline Development Group (GDG). The structure of the 2003 • Indolent clinical behaviour
guidelines was then discussed and re-evaluated, with headings and ○ Mycosis fungoides (and variants)
subheadings revised where appropriate; different coauthors were ○ Primary cutaneous CD30+ lymphoproliferative
disorder: anaplastic large cell lymphoma
allocated separate subsections. Each coauthor then performed a
detailed appraisal of the selected literature with discussions within ○ Primary cutaneous CD30+ lymphoproliferative
disorder: lymphomatoid papulosis
the GDG to resolve any issues, for example the quality of evidence
○ Subcutaneous panniculitis-like T-cell lymphoma
and making the appropriate recommendations. All subsections ○ Primary cutaneous CD4+ small/medium pleomorphic T-cell
were subsequently collated, circulated within the GDG and edited lymphoproliferative disorderb
to produce the final guideline. ○ Primary cutaneous acral CD8+ T-cell lymphomab
○ Hydroa vacciniforme-like lymphoproliferative
disorderb
3.0 Introduction • Aggressive clinical behaviour
○ Sezary syndrome
3.1. Incidence and epidemiology ○ Extranodal natural killer/T-cell lymphoma, nasal type
○ Primary cutaneous aggressive epidermotropic cytotoxic
Primary cutaneous lymphomas represent a heterogeneous CD8+ T-cell lymphoma
group of extranodal non-Hodgkin lymphomas, consisting of ○ Primary cutaneous c/d T-cell lymphoma
cutaneous B-cell lymphoma (CBCL) and cutaneous T-cell lym- ○ Primary cutaneous peripheral T-cell lymphoma,
phoma (CTCL). CTCL represents around 70% and CBCL about unspecified
30% of primary cutaneous lymphomas. Primary cutaneous a
Classified as extranodal marginal zone lymphoma of mucosa-
lymphomas have been defined by the European Organisation
associated lymphoid tissue (MALT lymphoma) in the 2016 ver-
for Research and Treatment of Cancer (EORTC)–World Health
sion. bChanges from the 2008 version.
Organization (WHO) classification3,4 and incorporated into
the most recent WHO classification (Table 1).5
These guidelines are broadly consistent with U.S. National
Comprehensive Cancer Network guidelines (www.nccn.org), Of these rarer CTCLs, the primary cutaneous CD30+ lymphoprolif-
and the EORTC and European Society for Medical Oncology erative disorders have an excellent prognosis, while some of the
clinical practice guidelines.4,6,7 other CTCL variants have poor prognosis and are more closely
Mycosis fungoides (MF) and Sezary syndrome (SS) are the related to peripheral T-cell lymphomas (PTCLs).9
most common clinicopathological subtypes of CTCL.3 A recent CBCL is classified into three main types: the more common,
U.K. National Cancer Information Network audit of newly low-grade primary cutaneous follicle centre lymphoma
diagnosed cases of CTCL from 2009 to 2013 suggests an (PCFCL) and primary cutaneous marginal zone lymphoma
annual incidence of 07 per 100 000 U.K. population, with a (PCMZL), which can occur at any age, and the rare, primary
significantly higher male incidence (16 : 10) and a peak age cutaneous, diffuse, large B-cell lymphoma (PCLBCL) leg type,
between 50 and 74 years. Completion of staging data was which occurs typically in elderly female patients.3
poor (7% in 2009, 20% in 2013).8 While the peak incidence A list of the abbreviations used in this guideline is available
of MF/SS occurs in the 50–74-year age group, approximately in Appendix S3 (see Supporting Information).
20% of patients are diagnosed with CTCL aged 25–49 years,
and MF may rarely present in childhood. Increasing age is a
4.0 Diagnosis, staging, prognosis and end
poor prognostic factor and the vast majority of younger
points
patients present with early-stage disease where the outlook is
excellent. However, management of younger adults with
4.1 Diagnosis
intermediate or advanced stages of CTCL may require a more
aggressive approach than usual. The diagnosis of different cutaneous lymphoma variants is
MF and SS are closely related both clinically and pathogenetically based on an assessment of the clinical and pathological fea-
but are distinct from other relatively rare types of primary CTCL. tures, which forms the basis for the National Cancer

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 3

Information Network minimum dataset for primary cutaneous In SS, an identical clonal T-cell-receptor gene rearrangement
lymphomas. MF is characterized by a distinct clinical morphol- is detected in skin and peripheral blood in combination with
ogy consisting of polymorphic patches or plaques, with the (i) a total Sezary cell count > 1000 cells lL1, (ii) an
more advanced cutaneous stage associated with tumours and expanded CD4+ T-cell population with a CD4 : CD8 ratio
erythroderma; peripheral adenopathy may or may not be pre- ≥ 10 or (iii) an expanded CD4+ T-cell population with abnor-
sent. SS is much less common (5% of CTCL) and is defined mal immunophenotype including CD4+ CD7 (> 30%) or
according to the WHO criteria by the presence of erythro- CD4+ CD26 (> 40%). Bone marrow aspirate and trephine
derma, peripheral lymphadenopathy and Sezary cells compris- biopsies are rarely indicated in MF, SS and LyP, unless there is
ing > 1000 cells lL1, or with a CD4 : CD8 ratio > 10 or an unexplained haematological abnormality, but they are more
loss of one or more T-cell antigens on flow cytometry and a commonly required for other CTCL variants and for high-
T-cell clone (stage B2). Human T-lymphotropic virus-1 serol- grade PCLBCL leg type. Bone marrow biopsy and aspirate is
ogy should be checked in all patients to exclude adult T-cell considered optional for low-grade PCMZL but should be con-
leukaemia/lymphoma, which can mimic CTCL. As for all new sidered for PCFCL, as systemic follicle centre lymphoma with
patients with lymphoma, HIV status should also be checked, secondary cutaneous involvement may occur.19,20 Serum lac-
in line with the Department of Health recommendations.10 tate dehydrogenase has prognostic significance and should be
Repeated biopsies may be required to establish the diagno- checked at diagnosis.21
sis, especially in MF. MF has a characteristic CD4+ epider- Existing staging systems for nodal lymphomas are not appli-
motropic infiltrate of T cells, although rare CD8+ phenotypic cable for primary cutaneous lymphomas. The original TNM
variants occur. T-cell receptor gene analysis is an important (primary tumour, regional nodes, metastasis) and clinical stag-
diagnostic and staging technique especially in patients with ing system for MF and SS22 has now been replaced by an
MF/SS, where analysis of peripheral blood provides critical International Society for Cutaneous Lymphomas (ISCL)–EORTC
prognostic information. It is essential that results are always revised staging system,23 which distinguishes patches and pla-
interpreted in the context of the clinicopathological features. ques, incorporates a molecular assessment of lymph node and
Patients with other CTCL variants do not have the character- peripheral blood, and provides a quantitative method for
istic polymorphic patches and plaques seen in MF and may assessing peripheral blood disease in SS (Tables S1–S3; see
present with cutaneous tumours, nodules or subcutaneous Supporting Information).24 These revised staging systems have
infiltration with distinctive pathology and immunophenotypic been adopted by the American Joint Committee on Cancer.25
findings.3 Lymphomatoid papulosis (LyP) represents part of a
spectrum of primary cutaneous CD30+ lymphoproliferative
4.3 Prognosis
disorders and is defined by a history of recurrent, self-healing
papules and nodules, and characteristic pathology. Recent studies have defined prognostic risks with CTCL and
CBCL presents with nodules, plaques and tumours with a CBCL, providing rates for overall survival (OS), disease-specific
characteristic pathology and immunophenotype, and site local- survival and progression-free survival (PFS) (Tables 2 and
ization can be an important distinguishing feature. A definitive 3).21,26–30
diagnosis can only be made on a representative biopsy, prefer- Unlike CTCL, prognosis for CBCL is determined by the type
ably an excision or elliptical biopsy. Borrelia burgdorferi infection of primary CBCL, not its stage (Table 3).3,11,31–33 Most
has been reported in association with PCMZL, and serology is patients with early stages of MF, primary cutaneous CD30+
indicated in all patients.11,12 Clonal IgH gene rearrangements lymphoproliferative disorders, and marginal zone and follicle
can aid diagnosis.13 centre lymphomas have a good prognosis, but disease progres-
sion and disease-specific mortality may occur. The 5- and 10-
year OS rates for MF are 80% and 57%, respectively, with dis-
4.2 Staging
ease-specific survival rates of 89% and 75% at 5 and 10 years,
Staging investigations should include a computed tomography respectively.27 Approximately 25% of early-stage patients
(CT) scan of the neck, chest, abdomen and pelvis in all patients develop disease progression.30 In contrast, patients with SS
with CTCL and CBCL, with the exception of those with early have an 11% 5-year survival, with a median survival of
stages of MF (IA/IB) and LyP unless there is palpable lym- 32 months from diagnosis. Patients with low-grade PCMZL
phadenopathy.14,15 Positron emission tomography (PET) CT and PCFCL have 5-year OS rates approaching 100%, but for
should be considered for patients with PCLBCL leg type in keep- PCLBCL leg type the prognosis is poor, with 5-year OS rates
ing with local policies for PET CT in aggressive B-cell lym- of between 36% and 45%.
phomas.16 The value of PET scans in MF/SS and other CTCL Several multivariate analyses of large cohorts of patients with
variants remains unclear at present, although this may indicate MF/SS have identified potential independent prognostic factors
which nodal basin to sample.17,18 In MF and SS, morphological at diagnosis. A study of 1502 U.K. patients identified separate
assessment of peripheral blood for Sezary cells is required (de- prognostic models for early and advanced MF.30,34 Specifically
fined as lymphocytes with hyperconvoluted nuclei). This should for early-stage disease, male sex, age (≥ 60 years), presence of
be carried out by flow cytometry to quantify the total number plaques (T1b/T2b), histological evidence of folliculotropic dis-
of Sezary cells and T-cell-receptor gene analysis. ease and palpable or histologically confirmed dermatopathic

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


4 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

Table 2 Prognosis in mycosis fungoides/Sezary syndrome21,26–30

Overall survival Disease-free survival Progression-free survival


Stage 5 year 10 year 5 year 10 year 5 year 10 year
IA T1a 97% 91% 100% 96% 95% 91%
T1b 91% 80% 96% 92% 88% 82%
IB T2a 85% 75% 90% 82% 85% 72%
T2b 81% 64% 86% 72% 75% 56%
IIA 78% 52% 89% 67% 83% 67%
IIB 40–65% 34% 50–80% 42% 52% 42%
IIIA 47% 37% 54% 45% 47% 38%
IIIB 40% 25% 48% 45% 18% 27%
IVA1 37% 18% 41% 20% 38% 17%
IVA2 18% 15% 23% 20% 23% 20%
IVB 18% – 18–20% – 18% –

Table 3 Prognosis in primary non-mycosis fungoides/Sezary standardized definitions of end points or response criteria. Con-
syndrome cutaneous lymphomas3,11,31–33 sequently, the detail and quality of response assessments have
been variable, and most studies have reported responses based
5-Year survival Overall Disease specific
on assessment of skin disease alone. This has led to the publica-
pcCD30+ anaplastic large 90% NA tion of ISCL–EORTC consensus criteria for disease end points,
cell lymphoma and assessment tools for MF/SS, which are based on utilization
pcCD30+ lymphomatoid papulosis 100% NA
of the modified Severity Weighted Assessment Tool technique
pc marginal zone lymphoma MZL > 95% 100%
(Table S4; see Supporting Information)35 and include a global
pc follicle centre lymphoma > 95% > 95%
pc diffuse large B-cell lymphoma 36–45% 61% scoring system based on responses of cutaneous and noncuta-
neous disease (Table S5; see Supporting Information).35
pc, primary cutaneous; NA, not available. In addition, a similar consensus proposal is in preparation for
other primary non-MF/non-SS cutaneous lymphomas. These stan-
peripheral nodes (N1/Nx) were adverse factors for progression dards for disease assessment should now be routinely used for
and survival. For late-stage disease, nodal involvement (N2/3) monitoring clinical outcomes in all patients with primary cutaneous
and blood and visceral disease are additional important prognos- lymphomas in conjunction with quality-of-life measurements.
tic factors. Patients with stage IIB disease have a poor prognosis,
but published data show marked variation in survival, perhaps 5.0 National Institute for Health and Care
reflecting the heterogeneous nature of tumour-stage disease in Excellence Improving Outcomes Guidance for
terms of both tumour burden and biology.26,27 The probability primary cutaneous lymphomas
of survival for patients with stage III erythrodermic MF, without
evidence of lymph node or peripheral blood involvement, is 5.1 Specialist multidisciplinary teams
broadly similar to that for stage IIB MF.
A Cutaneous Lymphoma International Consortium study of Primary cutaneous lymphomas are rare and heterogeneous
prognostic factors in 1275 patients with advanced MF/SS from malignancies that can present considerable diagnostic difficul-
29 international sites confirmed similar survival probability for ties. In addition, effective standard treatments have yet to be
patients with stage IIB and III disease, with significantly worse defined for many variants, particularly those with a poor prog-
survival in stage IV.21 Four independent prognostic factors nosis. As such, it is critical that patients have effective pathways
were identified, namely age > 60 years, large cell transforma- of care that allow rapid access to centres with broad experience
tion in skin, raised lactate dehydrogenase and stage IV disease. of primary cutaneous lymphomas. Such pathways have been
A prognostic index model combining these four factors identi- defined in the National Institute for Health and Care Excellence
fied three risk groups with significantly different 5-year sur- (NICE) Improving Outcomes Guidance (IOG) for skin cancers
vival rates: low risk (68%), intermediate risk (44%) and high including melanoma (Table 4).36,37 Patients outside the groups
risk (28%).21 This prognostic index is in the process of being where review by the supranetwork skin lymphoma multidisci-
prospectively validated. plinary team (MDT) is mandated may benefit from review by
this team, especially where there is difficulty in establishing a
diagnosis or the patient’s management is proving challenging,
4.4 Disease response and end points
depending on the expertise of the specialist skin MDT.
Historically, most trials in cutaneous lymphomas have been ret- All patients with suspected or proven primary cutaneous lym-
rospective reports of case series or phase II studies, with no phomas should be reviewed at regional specialist skin cancer

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 5

Table 4 Clinical pathway for cutaneous lymphomas Table 5 Supranetwork multidisciplinary teams (MDTs) in England, as
per the National Institute for Health and Care Excellence Improving
Supranetwork Outcomes Guidance for skin cancer36 and other network skin
Category Specialist MDT MDT lymphoma services in the U.K.
a
Stage IA–IIA MF Required
Stage IA–IIA MF Required Essential TSEB ECP
refractory to skin-directed Supranetwork MDTs
therapy Birmingham (Queen Yes (Coventry and Yes
Stage III–IVA1 erythrodermic Required Essential Elizabeth Hospital) Warwickshire
MF/SS University Hospital)
Stage IIB–IV MF/SS Required Essential Leeds (St James’s Yes No, referred to
pcCD30+ lymphoproliferative Required a
University Hospital) Rotherham
disorders Hospital
CTCL variants Required Essential Liverpool (Royal Yes (Clatterbridge Yes
a
CBCL Required Liverpool Hospital) Hospital, Wirral)
London (Guy’s and St Yes Yes
MF, mycosis fungoides; SS, Sezary syndrome; pc, primary cuta-
Thomas’ Hospital)
neous; CTCL, cutaneous T-cell lymphoma; CBCL, cutaneous B-
Manchester (The Yes Yes
cell lymphoma; MDT, multidisciplinary team. aThese patients Christie Hospital)
may benefit from review by the supranetwork skin lymphoma Newcastle Yes Yes
MDT. Table adapted from the National Institute for Health and (Freeman Hospital)
Care Excellence guidance.36 Nottingham Yes Yes
(City Hospital)
Other network skin lymphoma services
Belfast Yes No, would be
MDT meetings. There must be a close working relationship
referred
between the regional specialist skin cancer and haemato-oncol- Cardiff No, referred to Guy’s No, referred to
ogy MDTs, especially for patients who develop systemic disease. and St Thomas’ Bristol University
Hospitals
Glasgow No, referred to Yes
5.2 Supranetwork multidisciplinary teams Newcastle
The NICE IOG also defines which patients with primary cuta- TSEB, total skin electron beam; ECP, extracorporeal electrophore-
neous lymphomas should be referred to supranetwork MDTs sis.
across England (Table 5), at which dermatologists, clinical
oncologists, haemato-oncologists and both dermatopatholo-
gists and haematopathologists review the patient’s diagnosis biologics, chemotherapy and stem cell transplantation. Selection
and management plan. The IOG recommends that all patients of appropriate treatment is based on the stage of disease. Follow-
with stage IIB or higher MF, and all with SS and rare CTCL ing treatment, patients with advanced disease often develop
variants should be reviewed by the supranetwork MDT. These recurrent, low-grade disease that can be responsive to SDT. There
supranetwork MDTs also provide the appropriate pathway to have been very few randomized controlled studies. Recently
review cases where diagnostic or classification difficulties arise. Quaglino et al. published a retrospective analysis of 853 patients
Patients should be reviewed at supranetwork centres to pro- with advanced MF/SS from 21 centres.38 This large, multicentre
vide access to clinical trials, which are often the standard of retrospective study showed a substantial heterogeneity of treat-
care, and to agree on use of newly approved therapies. ment approaches in advanced MF/SS between U.S. and non-U.S.
The supranetwork MDT is also responsible for reviewing centres, with up to 24 different drugs, modalities or combina-
patients who require specialist treatment options, such as total tions used as first-line treatment, but these differences did not
skin electron beam therapy (TSEB) and extracorporeal photo- influence survival outcome. However, it was observed that
pheresis (ECP). Although not stipulated in the IOG, patients chemotherapy, when used as first-line treatment, is associated
with stage IB disease that is refractory to skin-directed therapy with a higher risk of death and/or change of therapy. For the pur-
(SDT) should be referred to the supranetwork MDT to ensure poses of this guideline, separate recommendations for MF/SS
that they have appropriate access to clinical trials and TSEB. All have been made according to stage.
patients who are being considered for stem cell transplantation
should be reviewed by a supranetwork MDT.
6.1 Skin-directed treatment

zary
6.0 Treatment of mycosis fungoides/Se 6.1.1 Topical therapies
syndrome
6.1.1.1 Stage IA–IIA mycosis fungoides
Therapeutic options range from SDT consisting of topical agents, There have been few randomized controlled trials (RCTs) of
phototherapy and radiotherapy, to systemic treatment with topical therapies for MF. The lack of well-controlled studies

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


6 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

Table 6 Topical therapy in mycosis fungoides

Level of
Therapy Design evidencea Stage Response rate
40
Mechlorethamine RCT 1+ IA–IIA 59% gel vs. 48% ointment
(noninferiority design by CAILS)
Retrospective case series41–46 3 IA–IB 51–80% (IA) 26–68% (IB)
Mechlorethamine + Prospective open-label phase II47 2+ IA–IB 58% complete response (duration 77 months)
betamethasone
Bexarotene gel Phase II50 2+ IA–IB 63%
Phase III51 IA–IIA 44%
Carmustine (BCNU) Case series48,49 3 IA–IIA 76%
Corticosteroids Case series39 3 IA–IB 94% (IA), 82% (IB)

RCT, randomized controlled trial; CAILS, Composite Assessment of Index Lesion Severity. aSee Appendix S1 in the Supporting Information.

limits the quality of evidence, although all of the topical thera- may be cured. Irritant contact dermatitis is the most com-
pies listed have some clinical efficacy for patches and thin pla- mon adverse effect, occurring in 10–40% of patients,
ques (Table 6). However, accurate data on end points such as depending on the preparation used. A recent 30-year popula-
duration of response and freedom from relapse are lacking. tion-based cohort study showed no increased risk of sec-
Many of the topical therapies reviewed remain unlicensed for ondary cancers (including nonmelanoma skin cancer or
use in MF. melanoma) in patients treated with mechlorethamine. Mortal-
ity and cause-specific mortality were not influenced by
6.1.1.1.1 Topical corticosteroids Topical corticosteroids, especially
mechlorethamine.46
class 1 (very potent) compounds, are effective for patches and
plaques in some patients with early-stage IA/IB MF, but 6.1.1.1.3 Topical carmustine (BCNU) Limited data suggest that
responses are rarely complete or durable.39 topical carmustine has similar efficacy to mechloretha-
mine.48,49 However, it is more extensively absorbed, increas-
6.1.1.1.2 Topical mechlorethamine (nitrogen mustard) The single lar-
ing the risk of bone marrow suppression, although the
gest RCT randomized 260 patients to a standard nitrogen
incidence of irritant contact dermatitis is lower (10%).
mustard 002% ointment or a novel nitrogen mustard 002%
gel preparation, used once daily for up to 12 months; no 6.1.1.1.4 Other topical therapies Topical bexarotene (1% gel) has
adjunctive therapy was allowed. The gel preparation was sta- been shown to be effective in prospective phase I/II open
tistically noninferior to the ointment, with response rates of studies50 and phase III trials for refractory or persistent, early-
585% (gel) vs. 477% (ointment) as measured by the Com- stage MF.51 Although approved by the U.S. Food and Drug
posite Assessment of Index Lesion Severity (CAILS).40 An Administration (FDA), topical bexarotene is not licensed in
extension study of 98 patients who failed to achieve com- Europe. Irritant contact dermatitis is common and was
plete response used a nitrogen mustard 004% gel, with a reported in 12% of patients.
CAILS response rate of 26% (6% complete response). Euro- Limited data from case reports and series suggest that imi-
pean Medicines Agency (EMA) approval of this new gel quimod 5% cream,52–59 5-fluorouracil cream,60 topical reti-
preparation (Ledagaâ) provides a more practical formulation noid preparations (tazarotene 01%, tretinoin 01%)61 and
than the historical, in-house compounded ointment and tacrolimus 01% ointment62 may have efficacy in early stages
aqueous solutions. of MF. The value of simple emollient therapy in MF was high-
These trials confirm that mechlorethamine is an effective lighted by a 24% placebo response rate to ointment base alone
topical therapy for early-stage MF. Previous studies have been in an RCT of topical peldesine (28% response rate, difference
predominantly retrospective and confounded by the use of not statistically significant).63
other therapeutic modalities.41–46 A prospective, nonrandom-
ized study of 64 patients with early-stage MF treated with a 6.1.1.2 Stage IIB mycosis fungoides (MF); stage III–IVA1 MF/Sezary syn-
002% aqueous solution of mechlorethamine followed by drome (SS); stage IVA2–B MF/SS
betamethasone cream twice weekly for 6 months reported There is no evidence to suggest that topical therapies have a
58% complete responses.47 significant impact on the course of the disease for advanced
The duration of response to mechlorethamine varies, and stages of MF/SS, although SDT can alleviate skin symptoms
the efficacy of maintenance therapy and whole-body applica- such as pain and pruritus, and most patients will require inter-
tion remains unclear, but rare patients with stage IA disease mittent topical treatments, especially topical steroids.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 7

Table 7 Phototherapy in mycosis fungoides

Therapy Design (all nonrandomized) Level of evidencea Stage Response rate Response duration (months)
66
UVB Retrospective 2 IA–IB 75% 51
UVB Retrospective65 2 IA–IB 71% 22
NB UVB Retrospective68 2 IA–IB 84% 125 weeks
NB UVB Retrospective69 2 IA–IB 84% (IA), 78% (IB)
NB UVB Retrospective91 2 IA–IB 100% > 60 (80% CR and 60% no recurrence)
PUVA Retrospective74 2 IA–IB 95% 43
PUVA Prospective79 2+ IA–IB 100% 20 (IA), 17 (IB)
PUVA Retrospective80 2 IA–II 63% (CR) 39

Regimens: psoralen–ultraviolet A (PUVA), twice weekly for 12–14 weeks; Narrowband ultraviolet B (NB UVB), two to three times weekly
for 12–14 weeks. CR, complete response. aSee Appendix S1 in the Supporting Information.

long-term outcomes following complete remission from


6.1.2 Phototherapy
PUVA monotherapy at a single institution reported that 30–
6.1.2.1 Stage IA–IIA mycosis fungoides 50% of patients had a durable remission (10-year disease-
Phototherapy (Table 7) is the standard of care for patients free survival), but maintenance PUVA was given to almost
with early stages of MF whose disease is not controlled by all responding patients.80 A total of 30% of patients showed
topical therapy, producing high complete remission rates.64–91 chronic photodamage and secondary skin cancers. This study
However, the duration of response is often limited. Repeated showed no significant difference in the OS rate for the non-
courses may be considered but the increased risk of skin can- relapsing and relapsing patient cohorts. Recent data from a
cer (including melanoma) limits the number of phototherapy prospective cohort study suggest that maintenance therapy
courses in a lifetime. does not prevent future relapse.81,82
There are few comparative studies or RCTs involving pso- For PUVA, an increased risk has been identified for
ralen–ultraviolet A (PUVA), and most case series lack a vali- patients receiving more than 250 treatments and/or
dated scoring system to assess tumour burden. The critical > 2000 J cm2.83,84 Based on the data available, the cumula-
questions as to whether PUVA affects time to progression and tive lifetime PUVA exposure should be limited (1200 J cm2
disease-specific survival remain unanswered. and/or 250 sessions).85 For maintenance PUVA the risks
may outweigh the benefits, and generally this should be
6.1.2.1.1 Ultraviolet B phototherapy Both narrowband ultraviolet
avoided for those patients who achieve excellent clinical
(UV)B (TL-01: 311–313 nm) and broadband UVB (290–
responses, although there are some exceptional refractory
320 nm) phototherapy can produce high complete response
patients for whom maintenance treatment may provide
rates.64–69 Responses are more likely in patients who have
important symptomatic relief.
only patches. There have been no prospective RCTs of nar-
rowband UVB, but a retrospective case series showed it to be 6.1.2.1.4 Photodynamic therapy Photodynamic therapy has been
as effective as PUVA for treatment of early-stage disease, with reported as a treatment for solitary plaques that are resistant
no difference in time to relapse.70 In a paediatric case series, to topical treatment. Small case series have shown durable
high response rates were seen (> 80%), including a number responses in 70–75% of patients.86,87 Photodynamic therapy
of complete responses in children with the hypopigmented is safe and well tolerated and may be appropriate for certain
variant of MF.71 sites, but a specific therapeutic role in MF has yet to be
6.1.2.1.2 Ultraviolet A phototherapy High-dose UVA1 photother- defined.
apy (340–400 nm), which penetrates more deeply than both
6.1.2.1.5 Excimer laser Several small case series have demon-
UVB and UVA, has shown clinical efficacy in a small case
strated the efficacy of the 308-nm excimer laser in early-stage
series.72
skin disease. Treatment with an excimer laser appears to be
6.1.2.1.3 Psoralen–ultraviolet A photochemotherapy Extensive, non- safe, effective and well tolerated for patches, but its precise
randomized and retrospective case studies have reported therapeutic role remains to be established.88–90
high complete response rates for PUVA in early stages.73–76
Data from an RCT showed that a minority (25–30% free- 6.1.2.2 Stage IIB mycosis fungoides
dom from relapse at 5 years) of patients achieving a com- Patients with tumours invariably have coexisting patches
plete response77 may have a prolonged duration of and plaques and may benefit from phototherapy if individ-
response. Many patients receive maintenance therapy but ual tumours respond to other options such as localized
the benefits are uncertain.78,79 A retrospective study of radiotherapy.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


8 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

Table 8 Combination psoralen–ultraviolet A (PUVA) in mycosis fungoides/Sezary syndrome

Duration
Therapy Design Level of evidencea Stage ORR (CR), % Number of patients (months) 5-year RFS, %
PUVA + interferon Case studies94
2 IA–IV 89 9
PUVA + interferon Case series97 2 IA–IV 68 (45) 22 20–75
PUVA + interferon Prospective phase II96 2+ IB–IIA 98 (84) 89 14
PUVA + interferon Case studies101 2 100 (100) 5 74
PUVA + interferon Case studies93 2 IB–IVB 90 (100) 39
PUVA + interferon Prospective phase II95 2+ IA–IVA 81 (75) 63 32 75
PUVA + bexarotene RCT EORTC-2101177 1+ IB–IIA 77 (31) 87 58 25
PUVA + bexarotene Case series100 2 I–III 67 (29) 14 2–10
PUVA + bexarotene Case series102 2 IA–IIB 100 (63) 8

ORR, overall response rate; RFS, relapse-free survival; RCT, randomized controlled trial; EORTC, European Organisation for Research and
Treatment of Cancer. aSee Appendix S1 in the Supporting Information.

6.1.2.3 Stage III/IVA1 erythrodermic mycosis fungoides/Sezary syndrome 6.1.3.2 Stage IIB mycosis fungoides (MF); stage III erythrodermic MF/
Patients with erythrodermic MF/SS are often intolerant of Sezary syndrome (SS); stage IVA2–B MF/SS
phototherapy due to aggravation of pruritus, but they can Although there is a lack of data, combination regimens
rarely respond. involving PUVA have been utilized as first-line treatment for
patients with erythrodermic MF, but patients with SS toler-
6.1.2.4 Stage IVA2–B mycosis fungoides/Sezary syndrome ate PUVA poorly. In contrast, combination PUVA regimens
PUVA can often be used as a salvage therapy, as patients with are rarely indicated as first-line therapy for tumour or nodal
advanced disease often relapse or have persistent low-grade disease, but are often used as an adjuvant or salvage ther-
skin disease characterized by patches and plaques following apy for those patients with persistent cutaneous disease fol-
systemic treatments. lowing debulking treatment for cutaneous tumours
or nodal/visceral disease, despite a lack of supportive
evidence.
6.1.3 Combination psoralen–ultraviolet A regimens
PUVA combined with interferon-alpha or bexarotene does not
improve overall response but may improve duration of 6.1.4 Radiotherapy (localized radiotherapy)
response and can the reduce cumulative UVA dose (Table 8).
6.1.4.1 Stage IA–IIA mycosis fungoides
MF is a highly radiosensitive malignancy, and localized radio-
6.1.3.1 Stage IA–IIA mycosis fungoides
therapy remains an effective treatment for patients with all
6.1.3.1.1 Psoralen–ultraviolet A and interferon-alpha RCTs have estab-
stages of disease.103–105 Palliative radiotherapy is very effec-
lished that combined PUVA and interferon-alpha are associated
tive for plaques and small or large tumours, which respond
with a reduced cumulative UVA dose to best response and an
to relatively low doses of radiotherapy, such as 8 Gy in two
improved duration of response compared with both PUVA
fractions.106 The dose-fractionation regimen should take into
alone and interferon-alpha combined with acitretin.92–94 How-
account the size of the treatment area, the treatment site and
ever, overall complete response rates for PUVA and interferon-
potential risk of acute and late damage to adjacent organs.
alpha appear to be similar to those with PUVA alone.93,95–102
MF is a multifocal disease, and while local control with
6.1.3.1.2 Psoralen–ultraviolet A and retinoids The combination of radiotherapy is readily achievable, this is usually a palliative
PUVA and retinoids (acitretin) is associated with a reduction approach. Therefore, it is appropriate to use the minimum
in cumulative UVA dose to best response, but with no dif- dose of radiotherapy to obtain local control. Using such a
ference in response rates compared with PUVA alone.99 low-dose approach also facilitates, if required, repeat treat-
PUVA and bexarotene have also been combined safely, with ment or treatment to an adjacent area where fields may over-
similar response rates to PUVA alone.100 Weaknesses of lap with a previous treatment field. This is especially
these studies include the lack of a validated scoring system important for sites with relatively poor skin tolerance to
to assess tumour burden, and data regarding the duration radiotherapy, such as the lower legs. Single fractions of 7–
of response and disease-free survival/OS. A recent RCT 8 Gy have been shown to be effective and convenient for
(EORTC-21011) comparing PUVA with combined PUVA the patient, but allow less re-treatment and may not be suit-
and bexarotene showed no significant difference in response able for areas of skin with poor tolerance.107 The use of
rates or duration of response, but a trend for lower cumu- higher doses of radiotherapy is seldom justified. Localized
lative UVA dose to achieve complete response in the combi- radiotherapy can be used at the same time as other SDT,
nation arm was noted.77 including phototherapy.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 9

For treatment of large areas of skin such as the trunk, limbs


6.1.5 Radiotherapy: total skin electron beam therapy
or whole scalp, smaller doses per fraction should be considered,
for example 20 Gy in 10 fractions. Regions such as the eyelids TSEB is a highly effective treatment for MF with excellent
may also benefit from smaller doses per fraction to limit acute complete response rates for all stages.115–134 EORTC consensus
and long-term toxicity; 4 Gy in two fractions may produce guidelines for the use of TSEB in CTCL have been pub-
symptomatic benefit in this situation lished.115 Critical issues to be addressed include maintenance
To define the radiotherapy clinical target volume (CTV) a of response to TSEB.
margin of ≥ 1 cm should be allowed around the disease pla-
que or tumour (gross tumour volume), and larger margins 6.1.5.1 Stage IA–IIA mycosis fungoides
may be required for some techniques. Further margins need A systematic review and meta-analysis of mostly nonrandom-
to be added according to the local protocol and radiotherapy ized studies has established that the rate of complete response
mode to determine the planning target volume and field size in patients with early-stage disease treated with TSEB is depen-
required to treat the area. To ensure that the full dermis (3– dent on the stage of disease, skin-surface dose and energy deliv-
5 mm thick) and a 5-mm-deep margin are being treated, the ered, with very high complete response rates observed.116
treatment dose should be delivered to a depth of 8–10 mm Greater skin-surface dose (32–36 Gy) and higher energy (4–6-
below the normal skin surface. MeV electrons) are associated with a higher rate of complete
Rarely, MF can present as a solitary patch or plaque, and in response and reasonable 5-year relapse-free survival
this setting local radiotherapy may be used with curative (Table 9).116–118 The conventional Stanford schedule of TSEB is
intent (doses of 20–30 Gy in 2-Gy fractions).108–111 36 Gy delivered with a six dual-field technique to the whole
Modern radiotherapy techniques such as complex matched skin, delivering 2-Gy fractions per 2-day cycle over 9 weeks,
electrons, intensity-modulated radiotherapy, helical Hi-ART with local boost treatments to tumours and thick plaques, and
therapy (TomoTherapyâ) and high-dose-rate brachytherapy supplemental radiotherapy to a median dose of 20 Gy to ‘shad-
mould techniques are increasingly being used to treat large owed’ or self-shielding areas, such as the perineum, soles of
areas of disease on curved surfaces of the skin such as the feet, scalp and inframammary folds.119 The standard U.K. mod-
scalp, trunk and limbs, and can provide excellent durable dis- ified Stanford protocol involved 30 Gy to the whole skin deliv-
ease control for many patients. ering 15-Gy fractions per 1-day cycle over 5 weeks.117 TSEB
delivering 30–36 Gy is associated with significant toxicity in
6.1.4.2 Stage IIB mycosis fungoides some patients. Fatigue has been reported in 38% of patients,
Low-dose palliative radiotherapy (e.g. 12 Gy in three fractions) erythema and desquamation in 47–76%, alopecia in 95%,
is very effective for MF tumours. Tumours involving large areas lower-leg oedema in 26%, blisters in 19–52% and skin infec-
require smaller doses per fraction, with schedules of 20–30 Gy tion in 31–32%.117,120
in 10–15 fractions. Radiotherapy field margins on the skin A recent review has shown similar response rates and dura-
should be determined as outlined above. The treatment depth tion of response with lower doses of TSEB (10–30 Gy),121 but
required is determined by the thickness of the tumour and its a lower median response duration was reported following a
depth of invasion, which in turn determines the radiotherapy total dose of 10 Gy.122 Very low doses of TSEB (4 Gy) have
mode required, for example orthovoltage X-rays, electrons or been tested, with reported inadequate responses.123 The results
photons. The depth of invasion may need to be assessed using of low-dose TSEB using 12 Gy have been reported from a
ultrasound, CT or magnetic resonance imaging. pooled analysis from three phase II trials. They showed an over-
all response rate (ORR) of 88%, with a median duration of clin-
6.1.4.3 Stage III–IVA1 erythrodermic mycosis fungoides/Sezary syndrome ical benefit of 707 weeks.124 A series of low-dose TSEB (12 Gy
Local radiotherapy may be used for patients who develop iso- in eight fractions over 2 weeks) in 103 patients has shown a
lated, symptomatic fissured lesions or tumours on a back- similar very high ORR, with a median duration of response of
ground of erythrodermic disease. It can also be useful for 18 months in patients with stage IB MF and 10 months in
treating extensive involvement of the hands and feet. patients with stage IIB MF, with significantly less toxicity than
standard-dose TSEB.125 Studies of low-dose TSEB in combina-
6.1.4.4 Stage IVA2–B mycosis fungoides/Sezary syndrome tion with newer agents are currently under way.
Localized, peripheral nodal disease can be treated with local One RCT comparing TSEB with topical mechlorethamine
external beam radiotherapy, as per standard protocols for non- showed similar rates of complete response and duration of
Hodgkin lymphoma. Visceral metastases can also be treated responses in early stages (IA–IB) of disease.126
with external beam radiotherapy using the same dosing sched- The use of adjuvant PUVA and topical mechlorethamine after
ules. Relatively low doses or single fractions can provide effec- TSEB has been reported, but the data are based on limited retro-
tive palliation and should be considered.112 Central nervous spective studies of small cohorts.127,128 It is not possible to rec-
system disease in MF has a very poor prognosis. Palliative ommend this as standard treatment, although PUVA can be used
low-dose, whole-brain radiotherapy can be used in patients as an effective early salvage therapy following TSEB. Further-
who are fit for treatment with a good performance sta- more, a recent report has shown no clinical advantage to the use
tus.113,114 of topical mechlorethamine as adjuvant treatment after TSEB.119

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


10 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

Table 9 Total skin electron beam (TSEB) therapy in mycosis fungoides/Sezary syndrome

Therapy No. of patients Dose (Gy) Level of evidencea Stage ORR, % CR, % Median RFS/PFS (months)
127
High dose 114 30–36 2+ IA 100 97 50
High dose117,119 103, 17 30–36 2+ IB 100 59–75 18–29
High dose117,119 77, 19 30–36 2+ IIB 95–100 47 9
High dose117,131 3, 45 30–36 2+ III 100 33–60 6–9
Low dose122,134 21 10 2+ IB–IV 95 57 VGPR 58
Low dose124 33 12 3 IB–III 88 27 MDOCB 16–17
Low dose121 33 5 to < 10 2+ IB–III 85–100 0–25 12
Low dose121 51 10 to < 20 2+ IB–III 96–100 7–52 257
Low dose121 32 20 to < 30 2+ IB–III 83–100 29–37 293
Low dose125 103 12 2+ IB–IV 87 18 132
54 IB 94 20 265
33 IIB 97 21 87
12 III 50 8 106
4 IV 25 0 –

ORR, overall response rate; CR, complete response; RFS, relapse-free survival; PFS, progression-free survival; VGPR, very good partial remis-
sion with modified Severity Weighted Assessment Tool score < 1; MDOCB, mean duration of clinical benefit (date of TSEB to date of next
whole-skin-equivalent treatment). aSee Appendix S1 in the Supporting Information.

TSEB should be considered as second-line treatment for improved disease-free and cause-specific survival when cor-
stage IB MF that has relapsed or is refractory to other SDT. It rected for peripheral blood and nodal involvement, but
can also be considered as first-line treatment in patients with these retrospective data may be open to selection bias.132
extensive cutaneous disease (stage T2b). There may be potential for TSEB to be used alone or in
combination for both skin and blood debulking in patients
6.1.5.2 Stage IIB-IVB mycosis fungoides with SS, but this needs further investigation in clinical
Patients with tumour stage IIB MF can achieve high rates of trials.133
durable complete responses to TSEB.121 One RCT included
103 patients with MF/SS (all stages) and compared TSEB and
6.2 Systemic biological therapies
multiagent chemotherapy (CAVE; cyclophosphamide, doxoru-
bicin, vincristine and etoposide) with sequential topical ther-
6.2.1 Interferon-alpha
apy including superficial radiotherapy and phototherapy. It
revealed a higher complete response rate in the TSEB– Interferon-alpha is licensed in the European Union for the
chemotherapy group (38% vs. 18%), but without a significant treatment of MF/SS (Table 10). There have been numerous,
difference in disease-free survival and OS.129 Nevertheless, a relatively small open nonrandomized studies of interferon-
combination of chemotherapy and TSEB can be invaluable for alpha in all stages of pretreated MF/SS, with variable dose
patients with severe skin disease and late stages of disease schedules (3–9 megaunits, three to seven times weekly). ORRs
(stage IVA2–B). In addition, TSEB can be utilized as debulking > 50% and complete response rates > 20% have been
treatment prior to allogeneic stem cell transplantation.130 reported. Response rates are higher in early stages of disease
and with higher doses of interferon.135–137 A small, random-
6.1.5.3 Stage III-IVA1 erythrodermic mycosis fungoides/Sezary syndrome ized study comparing PEGylated interferon-alpha-2b plus
A retrospective study of TSEB as monotherapy for erythroder- PUVA with standard interferon-alpha-2b plus PUVA showed a
mic CTCL noted a higher complete response rate and lower higher ORR with less constitutional side-effects for patients
rate of disease progression in patients without peripheral treated with PEGylated interferon-alpha-2b plus PUVA.94
blood involvement (stage III disease) and in those receiving a However, the patients in the PEGylated interferon arm had a
more intense regimen (32–40 Gy).131 These retrospective data higher rate of myelosuppression and liver toxicity than those
suggest that TSEB can be considered as second-line, and even treated with standard interferon.
first-line therapy for patients with erythrodermic MF, although
patients with erythrodermic disease can be very sensitive to 6.2.1.1 Stage IA–IIA mycosis fungoides
radiotherapy and have severe acute radiation reactions. A Although higher response rates have been reported for inter-
radiotherapy test dose to a limited area of skin should be used feron in patients with early-stage MF,135–137 interferon-alpha
before treatment, and careful patient selection is advised. should not be used in patients with early-stage MF who are
A retrospective study comparing TSEB alone with TSEB responsive to SDT, as these patients have a good prognosis
followed by ECP in erythrodermic MF/SS suggests that the and there is no evidence that interferon affects long-term
combination of TSEB and ECP is associated significantly with outcome.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 11

Table 10 Biological therapies in mycosis fungoides/Sezary syndrome

Therapy Design Level of evidenceb Stage ORR, % CR, % Duration


135
Interferon-a Case series 2 IA–IVB 45–74 10–27 NA
Interferon-a Case series136 2 IA–IIA 88 ? NA
Interferon-a Case series138 2 IIB–IVB 29–63 ? NA
Bexarotene Phase II145 2+ IA–IIA 54 0 516 days
Bexarotene Phase II148 2+ IIB–IVB 45–51 0 299 days
Brentuximab vedotin Phase II160 2+ IB–IVB 73 35 32 weeks (range 3–93)
Brentuximab vedotin Phase II161 2+ IB–IVB 70 3 54% of responders
progression free at 12 months
Brentuximab vedotin RCT Phase III162 1+ 1A–IVB 56 16 167 months
Denileukin diftitoxa Phase II163 2++ IB–IVA 30 10 69 months
Denileukin diftitoxa RCT phase III164 1+ IA–III 44 10 > 2 years (PFS)
Alemtuzumab high dose Case series151,152 2 IIB–IV 37–100 25–47 6–9 months
Alemtuzumab low dose Case series154,155 2 IIB–IV 85 21 TTF 12 months
Vorinostata Phase II165 2+ IB–IVA 24–297 0 106–185 days
Romidepsina Phase II166 2+ IB–IVA 34 6 137–149 months
Bortezomib Case series167 2 III–IV 70 10 7–14 months

ORR, overall response rate; CR, complete response; RCT, randomized controlled trial; NA, not available; PFS, progression-free survival; TTF,
time to treatment failure. aApproved by the U.S. Food and Drug Administration but not the European Medicines Agency. bSee Appendix S1
in the Supporting Information.

However, for patients with early-stage MF who are refractory time to response, and improve duration of response,143,144
to SDT, interferon and PUVA can be used in combination, and although the only RCT92 suggested no benefit.
evidence suggests that this may reduce the cumulative UV dose Prospective studies of bexarotene, a retinoid X receptor rex-
required to achieve a complete response.99 One RCT comparing inoid, have shown significant efficacy and good duration of
PUVA and interferon with interferon plus acitretin in stage I response with low rates of disease progression in early-stage
and II patients demonstrated that PUVA and interferon were disease.145 Bexarotene has been used in combination with
superior, with a 70% complete response rate compared with PUVA;102,146 however, the EORTC-21011 RCT in low-risk MF
38% for the drug combination.92 comparing PUVA alone with PUVA plus bexarotene showed
no difference in response rates.77
6.2.1.2 Stage IIB mycosis fungoides (MF); stage III–IVA1 erythrodermic
MF/Sezary syndrome (SS); stage IVA2–B MF/SS 6.2.2.2 Stage IIB mycosis fungoides (MF); stage III–IVA1 erythrodermic
Most studies of interferon have involved patients with refrac- MF/Sezary syndrome (SS); stage IVA2–B MF/SS
tory, advanced-stage disease but have not distinguished Bexarotene is licensed in the European Union for the treat-
between patients with erythroderma and either tumour-stage ment of late-stage MF/SS refractory to at least one systemic
disease or nodal involvement. Responses are dose dependent agent, and U.K. consensus guidelines for bexarotene prescrib-
but complete response is rare.135,136,138 Responses have been ing and management have been published.147 Maximum
reported with combined interferon-alpha and acitretin in responses may take 6 months, and bexarotene should be con-
patients with MF and SS resistant to interferon alone.139 Aviles tinued until loss of response.
et al.140 reported good response rates (80%) at 1 year with Prospective studies of bexarotene in refractory, late-stage
combinations of interferon (5 megaunits three times a week) disease have shown encouraging ORRs and response durations
and methotrexate or interferon and trans-retinoic acid. Data on at doses of 300 mg m2 daily,148 which have been confirmed
combined interferon-alpha and bexarotene are currently lim- in subsequent case series.146,149 These results suggest that bex-
ited to case series.141 Patients with erythrodermic MF treated arotene has a useful role, particularly for erythrodermic dis-
with immunosuppressive therapy such as ciclosporin for sus- ease, but monotherapy is unlikely to be effective for tumours
pected inflammatory disorders may have a more aggressive or nodal disease. Whether adjuvant bexarotene has a role in
disease course and worse outcome.142 maintaining remission or partial responses in those patients
with pretreated advanced disease is currently unknown.

6.2.2 Retinoids and rexinoids


6.2.3 Antibody therapies
6.2.2.1 Stage IA–IIA mycosis fungoides
Retinoic acid receptor retinoids (acitretin) have modest effects 6.2.3.1 Stage IA–IIA mycosis fungoides
in early stages of MF as monotherapy, but in combination At present, there is no evidence that antibody therapies should
with PUVA they may reduce the cumulative UVA dose and be used for patients with early-stage disease.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


12 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

expression appeared to correlate with tumour responses. Those


6.2.4 Alemtuzumab (CAMPATH-IH)
with < 5% CD30 expression had a lower likelihood of global
6.2.4.1 Stage IIB mycosis fungoides (MF); stage III–IVA1 erythrodermic response than did those with CD30 expression ≥ 5%.161
MF/Sezary syndrome (SS); stage IIVA2–B MF/SS Further to these phase II trials, a randomized, open-label
Phase II trials of alemtuzumab (a humanized anti-CD52 anti- phase III trial (ALCANZA) was designed to evaluate single-agent
body) in small cohorts of patients with advanced MF/SS have brentuximab vedotin vs. a control arm of the investigator’s
shown encouraging ORRs.150–156 Reported overall response choice of standard therapies, namely methotrexate or bexaro-
and complete remission rates are high, but typically responses tene, in patients with CD30-expressing CTCL, including those
are short lived, with only a minority of patients achieving with primary cutaneous anaplastic large cell lymphoma
responses for longer than 12 months. Clinical responses are (pcALCL) or MF. The primary end point was objective response
best in SS, where symptomatic benefit may be dramatic. In lasting at least 4 months (ORR4) as assessed by Global Response
contrast, recent evidence suggests that alemtuzumab is ineffec- Score. Brentuximab vedotin resulted in a statistically significant
tive in patients with MF with tumours and large cell transfor- improvement in the rate of ORR4 (563% vs. 125% in the con-
mation.157 trol arm). The median PFS was 167 months vs. 35 months, a
Reports of possible cardiac toxicity158 have not been con- significant difference favouring brentuximab (P < 0001).
firmed and the safety profile has been acceptable even in In November 2016, the FDA granted brentuximab vedotin
elderly patients, although infection and specifically cytomega- Breakthrough Therapy designation for the treatment of
lovirus reactivation remains an issue and patients need irradi- patients with CD30-expressing MF and pcALCL who require
ated blood products.155 In the majority of studies, systemic therapy and have received one prior systemic ther-
alemtuzumab has been administered intravenously at the stan- apy. The phase III data from this trial have also led to EMA
dard dose of 30 mg three times per week. However, one approval in patients with CD30+ CTCL.162 Peripheral neuropa-
study examined the use of a lower-dosage subcutaneous thy is a frequent side-effect, occurring in over 60% of
schedule at 10 mg three times per week, with similar efficacy patients, but resolution or improvement of symptoms on ces-
but reduction in toxicity.154 Alemtuzumab remains an impor- sation of treatment is reported.
tant second-line therapeutic option for patients with advanced Other biological therapies are listed in Table 10.163–167
disease, particularly erythrodermic stage III–IVA1 MF/SS.
6.3 Chemotherapy
6.2.5 Brentuximab vedotin
Systemic chemotherapy (Table 11) is usually reserved for
Brentuximab vedotin is an antibody–drug conjugate comprised of patients with advanced disease, or disease refractory to SDT or
an anti-CD30 monoclonal antibody attached by an enzyme-cleava- immunobiological therapy, and is palliative rather than cura-
ble linker to the antimicrotubule agent, monomethyl auristatin E, tive.168–189 Although good responses are reported with both
which is released upon internalization into CD30-expressing single-agent and combination regimens, overall the results are
tumour cells. It is administered intravenously at 18 mg kg1 over disappointing when compared with other lymphomas. A par-
30 min every 21 days, for a maximum of 16 cycles.159 ticular difficulty in interpreting data in this area is the variable
Phase II trials demonstrated encouragingly high response and largely suboptimal reporting of response assessment and
rates,160,161 where the proportion of CD30 tumour cell the heterogeneity of risk factors within the spectrum of the

Table 11 Chemotherapy in mycosis fungoides/Sezary syndrome

Therapy Design Level of evidencea ORR, % CR, % Median RFS or PFS (months)
174–177,181
Pentostatin Case series 2 35–71 6–32 9
Cladribine Case series178 2 28–41 14–19 45
Fludarabine Case series179,188 2 30–51 114 59
Liposomal doxorubicin Case series183 2 80–88 45–66 15
Liposomal doxorubicin EORTC-21012184 2+ 408 61 6
Gemcitabine Case series180–182 2 68–75 8–22 10–15
Methotrexate Case series170 2 58 41 31
Trimetrexate Case series171 2 45 Not recorded
Pralatrexate Phase II189 2+ 45 Not recorded
Combination chemotherapy Systematic review168 2+ 81 38 5–41
Chlorambucil Case series169 2 89 36 Not recorded
EPOCH Phase II187 2+ 80 27 8

ORR, overall response rate; CR, complete response; RFS, relapse-free survival; PFS, progression-free survival; EORTC, European Organisation
for Research and Treatment of Cancer; EPOCH, etoposide, prednisolone, vincristine, cyclophosphamide, doxorubicin. aSee Appendix S1 in
the Supporting Information.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 13

patients with MF and SS who are treated. In the absence of nonrandomized studies of other multiagent chemotherapy reg-
randomized, phase III trials, comparison of response rates imens – VICOP-B (etoposide, idarubicin, cyclophosphamide,
across phase II studies is fraught with difficulties of interpreta- vincristine, prednisolone, bleomycin) and EPOCH (etoposide,
tion, and therefore recommendations are subject to inherent prednisolone, vincristine, cyclophosphamide, doxorubicin) –
investigator and reporting bias. have revealed similar OR rates and durations of response to
those for CHOP.186,187 Overall survival rates remain
unchanged for these more intensive regimens and toxicity
6.3.1 Stage IA–IIA mycosis fungoides
may be considerable; therefore, these regimens have little clin-
Chemotherapy is contraindicated for early stages of disease, as ical benefit over less intensive regimens.
low-grade disease is relatively resistant to chemotherapy and The patient’s quality of life should always be considered
response duration is short. before embarking on chemotherapy regimens with limited
efficacy. Patients with CTCL are at high risk of neutropenic
sepsis, and therapy-related mortality with combination
6.3.2 Stage IIB mycosis fungoides (MF); stage III–IVA1
chemotherapy is a significant risk. Single-agent regimens
erythrodermic MF/Sezary syndrome (SS); stage IVA2–B
appear to have similar efficacy to combination regimens but
MF/SS
with lower toxicity, and therefore are preferable as palliative
Quaglino et al. found treatment heterogeneity between U.S. therapy in late stages of MF and SS. Durable responses with
and non-U.S. centres but no difference in survival.38 However, either single- or multiagent regimens are rare.
a constant geographical feature was the finding that first-line The limited data suggest that methotrexate and pentostatin
chemotherapy was associated with a higher risk of death. are most appropriate for erythrodermic SS, while gemcitabine
One RCT comparing combination chemotherapy and TSEB and liposomal doxorubicin are more appropriate for advanced
against conservative SDT showed a better overall response in stages of MF. Maintenance or adjuvant treatment with bioim-
the combination arm but greater morbidity and no improve- munotherapy, SDT or TSEB for patients achieving a response
ment in OS.129 A systematic review of uncontrolled open with chemotherapy may be considered. Patients frequently
studies of single-agent regimens in 526 patients with CTCL treated relapse, with early stages of disease manifested by patches and
between 1988 and 1994 revealed OR rates of 62%, with com- plaques, although there is no trial evidence suggesting that
plete response rates of 33% and median response durations of any such approach improves outcomes, and further well-
3–22 months.168 Single-agent therapies showing clinical effi- designed studies are required.
cacy in MF/SS include alkylating agents (chlorambucil and While outcomes are invariably poor for patients with
cyclophosphamide),169 antimetabolites (methotrexate and advanced disease, with median survival of < 12 months (stage
trimetrexate),170,171 purine analogues (pentostatin, 2-chloro- IV), for those stage IVA2 patients with a good performance sta-
deoxyadenosine and fludarabine),172–179 the pyrimidine tus, the aim is to achieve a durable complete or good partial
antimetabolite gemcitabine180–182 and liposomal doxoru- remission and to proceed to a reduced-intensity allogeneic
bicin.183,184 A recent U.K. prospective phase II study assessing transplant.
a combination regimen incorporating gemcitabine and bexaro-
tene in advanced MF/SS (GEMBEX) showed a low response
6.4 Extracorporeal photopheresis
rate following chemotherapy at 12 weeks (31%, all with par-
tial response), reducing to a 14% ORR at 24 weeks despite
6.4.1 Stage IA–IIA mycosis fungoides
bexarotene maintenance. In addition, control of hyperlipi-
daemia was challenging with this combination.185 Although responses to ECP have been reported in early-stage
A systematic review of different combination chemotherapy regi- MF, this is based mostly on a small number of patients in
mens in patients with MF and SS stages IIB–IVB treated between poorly controlled single-centre studies, including some in
1988 and 1994 showed an OR rate of 81% in 331 patients which patients were treated with adjuvant therapies
and a complete response rate of 38%, with response duration (Table 12).190–192 The only randomized study looking at
ranging from 5 to 41 months.168 The most commonly patients with early-stage MF treated with either PUVA or ECP
reported regimen used in MF/SS is CHOP (cyclophosphamide, showed that PUVA was more effective over a 6-month treat-
doxorubicin, vincristine, prednisone). Prospective, ment period.193 Therefore, there is no evidence at present to

Table 12 Electrophoresis (ECP) in mycosis fungoides/Sezary syndrome (adapted from Scarisbrick et al.)196

Therapy Design Level of evidencea Stage ORR, % CR, % OS (months)


ECP Systematic review 2+ IB–IV 63 20
ECP Systematic review 2+ III 35–71 14–26 39–100
ECP Multiple case series 2 III 31–86 0–33

ORR, overall response rate; CR, complete response; OS, overall survival. aSee Appendix S1 in the Supporting Information.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


14 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

suggest that ECP should be used for patients with early-stage 6.5.1.1 Stage IA–IIA mycosis fungoides
disease. Toxin therapies should not be used for early-stage disease in
view of potential adverse effects, but denileukin diftitox can
be effective for early-stage patients who are resistant to SDT.
6.4.2 Stage IIB mycosis fungoides
ECP is not effective for stage IIB MF.194,195 6.5.1.2 Stage IIB mycosis fungoides (MF); stage III–IVA1 erythrodermic
MF/Sezary syndrome (SS); stage IVA2–B) MF/SS
Denileukin diftitox (18 lg kg1 per day for 5 days, as a 21-
6.4.3 Stage III–IVA1 erythrodermic mycosis fungoides/
day cycle) is effective in heavily pretreated patients with late
zary syndrome
Se
stages of disease,163,164,214 and efficacy may be improved by
A systematic review of nonrandomized and mostly retrospective combining with bexarotene.215
studies of ECP in 650 patients from 30 published studies
showed an ORR of 63% and a complete response rate of
6.5.2 Histone deacetylase inhibitors
20%.196 Response rates are highest in those with erythrodermic
stage III–IVA1 MF/SS.190,197–200 Treatment is given on two Histone deacetylase (HDAC) inhibitors are an emerging class of
consecutive days and repeated every 4 weeks, but it may be drugs that increase acetylation of histones and nonhistone pro-
given more frequently (e.g. every 2 weeks) in those with a teins affecting gene transcription, which results in cell-cycle
high blood-tumour burden.194,196,201 Responses may take up arrest and apoptosis. Several phase II studies have established
to 6 months to develop and therapy should continue until loss that HDAC inhibitors have clinical efficacy in all stages of MF/
of response. The U.K. consensus guidelines for ECP in CTCL SS, with an acceptable safety profile.165,166,216,217 These studies
recommend ECP as first-line systemic treatment in patients with led to FDA approval for vorinostat (suberoylanilide hydroxamic
erythrodermic CTCL with evidence of blood involvement, treat- acid) and romidepsin for the treatment of refractory MF/SS.
ing with one to two cycles per month. The guidelines were Neither HDAC inhibitor is licensed for use in Europe.
recently updated in 2017 with the recommendation that ECP
treatment should be continued until loss of response.196,202 6.5.2.1 Stage IA–IIA mycosis fungoides
A recent U.K. audit report of 97 patients with CTCL treated HDAC inhibitors should not be used for early-stage MF but
since 2010 found that 45% had received ECP as the first-line can be effective for patients resistant or refractory to SDT.
systemic agent.203 The response rate at 6 months was 61%
and the median length of treatment was 9 months. There have 6.5.2.2 Stage IIB mycosis fungoides (MF); stage III–IVA1 erythrodermic
been numerous small case series of erythrodermic patients MF/Sezary syndrome (SS); stage IVA2–B MF/SS
treated with combinations of ECP, interferon-alpha and/or Phase II studies of vorinostat and romidepsin have shown sim-
retinoids, as well as bexarotene, which suggest that combina- ilar ORRs in advanced disease including stage III
tion therapy can be highly effective, as both clinical and patients.165,166,216,217 Evaluation of patients on vorinostat for
molecular remissions have been documented.196,204–213 over 2 years provides evidence of long-term safety and tolera-
Although these studies were uncontrolled, the evidence sug- bility.218 One study also showed a significant improvement in
gests that ECP can be an effective and well-tolerated therapy pruritus in 43% of patients, including those who had no
for patients with erythrodermic stage III–IVA1 MF/SS and that objective clinical response.166
such patients may benefit from combination regimens com-
bining ECP with other systemic therapies. ECP is an FDA-
6.5.3 Other systemic chemotherapies
approved treatment for MF/SS. In contrast, there is a lack of
evidence for use of ECP in stage IVA2–B MF/SS. Pralatrexate belongs to a class of folate analogues, known as
the 10-deazaaminopterins, and is emerging as a promising
new agent in the treatment of T-cell lymphomas, including
6.5 Systemic therapies (Food and Drug Administration
CTCL. Phase II trials have demonstrated good response rates,
but not European Medicines Agency approved)
although adverse effects, including mucositis, are a con-
cern.219 A further study, using a lower dose of pralatrexate,
6.5.1 Toxin therapies (denileukin diftitox: diphtheria
reported better tolerance but similar response rates.189
interleukin-2 fusion toxin; Ontak)

Denileukin diftitox is a fusion protein targeting the interleukin-


6.6 Systemic therapies (not Food and Drug
2 receptor (CD25). Large RCTs have shown significant efficacy
Administration or European Medicines Agency approved)
and improved PFS in all stages of disease (Table 9). Denileukin
diftitox is FDA but not EMA approved for MF/SS, but it is cur- Bortezomib, a proteasome inhibitor, has shown clinical efficacy
rently not available in the U.S.A. because of a manufacturing in a small series of patients with advanced stages of CTCL,167
change, and results of an ongoing trial are awaited. Toxicity but its clinical role remains to be established. Temozolomide
includes hypersensitivity reactions and vascular leak. has also been reported to be effective in a minority of patients

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 15

in a small multicentre study of patients with advanced MF/ infusion, but this in turn may lead to significant graft-versus-
SS220 and those with central nervous system progression.114 host disease.
Maximal benefit from RIC-HSCT is observed when it is per-
formed before patients develop highly refractory disease and
6.7 Autologous or allogeneic peripheral blood or bone
when there is low disease bulk at the time of transplanta-
marrow stem cell transplant
tion.225 Appropriate data registration is required to identify
Autologous haemopoietic stem cell transplant (HSCT) appears the optimal conditioning regimen for allo-HSCT and to define
to be associated with only short-term remission and is there- clinical outcomes. Considering the numerous biological agents
fore difficult to justify.221–223 In contrast, allogeneic HSCT is a that are becoming available and the potential efficacy of allo-
complex form of immunotherapy aimed at consolidating treat- HSCT, younger patients with stage IIB–IV disease and poor
ment of advanced MF/SS to produce a durable complete prognostic factors or aggressive disease should be reviewed at
remission.224 A retrospective study from the European Group supranetwork MDTs and, if appropriate, referred early to a
for Blood and Marrow Transplant (EBMT) included a hetero- specialist transplant centre.
geneous group of patients who received both myeloablative
and reduced-intensity conditioning (RIC) regimens. The study
6.8 Future perspectives
showed an estimated 3-year survival rate of 54%.225 However,
the treatment-related mortality of 25% at 1 year for CTCL was FDA-approved therapies include topical bexarotene gel,
a concern. The EBMT data suggested that alemtuzumab should mechlorethamine gel, oral bexarotene, denileukin diftitox,
not be used as part of the pretransplant conditioning regimen photopheresis and the HDAC inhibitors vorinostat and romi-
because of a higher rate of early relapse and increased treat- depsin.
ment-related mortality.225 In contrast, only topical mechlorethamine ointment for all
A number of other case reports and small retrospective stages of MF, and interferon-alpha, brentuximab and oral bex-
series have also described clinical responses and durable arotene for late-stage refractory MF/SS, are EMA approved.
remissions of more than 3 years following myeloablative Currently, there is an unmet need for improved treatments in
allo-HSCT226–229 and reduced-intensity conditioning allo- CTCL and an urgent need for well-designed RCTs with appro-
HSCT.229–234 A single-centre report described outcomes in 19 priate clinical end points.
recipients of allo-HSCT using an RIC regimen that included Until recently, denileukin diftitox was not available world-
TSEB irradiation prior to transplant. Eleven patients achieved wide because of manufacturing problems, although new clini-
sustained complete remissions, some of which were following cal trials have commenced.
donor lymphocyte infusion, immunosuppression withdrawal
and/or chemotherapy.130 An update of this study utilizing Recommendations
pretransplant TSEB in the majority of patients showed that
60% achieved complete skin remission as a result.235 • The diagnosis of MF and SS is based on a combination
A more recent analysis of the EBMT cohort (60 patients of clinical and pathological criteria and requires close
with MF/SS) in 2014 reported an OS of 46% and PFS of 32% MDT collaboration between different specialities.
at 5 years.236 Relapse or progression occurred in 45% at a (Strength of recommendation D (GPP))
median of 38 months following HSCT, but many patients • All patients with early-stage MF refractory to SDT and
were rescued with donor lymphocyte infusions. Nonrelapse late-stage MF/SS should be reviewed by supranetwork
mortality was 22%, with the last nonrelapse death occurring MDTs to agree a management plan and provide the
14 months following HSCT. Multivariate analysis confirmed opportunity for consideration in appropriate clinical
worse outcomes for myeloablative conditioning regimens and trials. (Strength of recommendation D (GPP))
transplants from unrelated donors.236 A meta-analysis of allo- • The aim of treatment is to control the patient’s disease
geneic or autologous SCT in MF and SS supports the view that and symptoms with the minimum of intervention.
allogeneic transplantation provides a better survival and dis- Management may range from an expectant policy
ease-free outcome than autologous transplantation.237 through to nonmyeloablative allogeneic stem cell trans-
There is increasing enthusiasm for RIC allograft approaches plantation. (Strength of recommendation D (GPP))
for a number of reasons. CTCL affects older patients, many of • SDTs, including phototherapy and local radiotherapy,
whom have chronically infected skin tumours, and the risk of are the standard of care for patients with early-stage
life-threatening sepsis is lower with reduced-intensity allo- IA–IIA MF. (Strength of recommendation C)
grafts. Allogeneic transplant is successful in part because of • Patients with stage IA–IIA MF who are refractory to
the graft-versus-lymphoma effect of the donor graft, indepen- SDT often require TSEB and systemic biological thera-
dent of the conditioning regimen, but this has to be balanced pies. There is no evidence to support the use of main-
against the morbidity associated with chronic graft-versus-host tenance phototherapy. (Strength of recommendation C)
disease.225 In addition, and relevant to the above, a major risk • Patients with stage IIB MF can have an unpredictable
following allogeneic transplant is disease relapse. Clearly, clinical course: some patients develop only small and
some patients can be salvaged with donor lymphocyte infrequent skin tumours and often obtain durable

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


16 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

responses to localized radiotherapy and other SDT 7.1.1 Primary cutaneous CD30+ anaplastic large cell
options for persistent patches and plaques; other lymphoma
patients develop extensive bulky skin tumours and pcALCL is characterized by solitary tumours, grouped tumours
rapidly progressive disease requiring TSEB and systemic or thick plaques of disease. Rarely, patients present with gener-
chemotherapy. (Strength of recommendation C) alized multifocal lesions. Partial regression may occur but it is
• Following treatment, patients with advanced MF may rarely complete, and this clinical feature helps to distinguish
develop recurrent, low-grade disease where SDT is a pcALCL from LyP. The tumour phenotype is CD30+ ALK, and
treatment option. (Strength of recommendation D (GPP)) this CTCL variant is biologically distinct from its nodal counter-
• Patients with erythrodermic MF (stage III) and SS part. The prognosis is excellent, with 5-year survival rates of
(stage IVA1) often require single or combination sys- 96%.31 Staging is based on the non-MF/SS staging recommen-
temic therapies such as methotrexate, photopheresis, dations,24 and CT scans and bone marrow trephine biopsies are
bexarotene and interferon-alpha as first-line treatment. required to confirm that the skin disease is not a manifestation
(Strength of recommendation C) of a systemic lymphoma. Primary cutaneous CD30+ ALK+ ALCL
• Options for SS (stage IVA1–2) also include clinical tri- has been rarely described but there is no evidence that such
als and alemtuzumab. (Strength of recommendation C) patients have a worse prognosis.
• For stage IVA2–B MF/SS, radiotherapy (including TSEB
for selected stage IV patients) and single-agent 7.1.1.1 Surgery
chemotherapy regimens are the preferred option, but Surgical excision can be an effective treatment for solitary
response duration is often short. (Strength of recommenda- tumours. If the lesion has been completely excised with clear
tion C) histological margins, it may be appropriate to manage the
• Brentuximab offers an effective option for refractory patient with adjuvant radiotherapy or active surveillance.
stage IB disease and advanced stages of MF/SS with
tumour CD30 expression. (Strength of recommendation B) 7.1.1.2 Radiotherapy
• All patients with early-stage MF refractory to SDT and Radiotherapy is an effective treatment modality for pcALCL,
stage IIB–IV MF/SS should be offered the opportunity with very high complete remission rates reported. Cutaneous
to participate in well-designed clinical trials. (Strength of recurrences have been reported in 41–64% of patients.15
recommendation D (GPP)) International consensus recommends a dose of 24–30 Gy.238
• Autologous HSCT should not be considered for
advanced stages of MF/SS. (Strength of recommendation A) 7.1.1.3 Chemotherapy
• Reduced-intensity allogeneic HSCT should be considered Doxorubicin-based, multiagent chemotherapy regimens are
for selected groups of patients with advanced MF/SS to effective with high complete response rates, but response
consolidate treatment responses. (Strength of recommendation B) duration is limited with relapses in > 60% of patients.15 Vin-
• The conditioning regimens and outcomes of RIC allo- blastine has been effective as a single agent in pcALCL.239
HSCT should be collected through data registries such There are only limited reports of single-agent chemotherapy
as the EBMT registry. (Strength of recommendation D (GPP)) agents such as methotrexate, etoposide and gemcitabine.

7.1.1.4 Antibodies
The fusion toxin brentuximab vedotin (SGN-35) in a large
7.0 Treatment of non-mycosis fungoides/
phase III RCT in pcALCL has shown significant benefit in
Sezary syndrome primary cutaneous T-cell
terms of durable response and improved PFS compared with
lymphomas
methotrexate or bexarotene.162
7.1 Primary cutaneous CD30+ lymphoproliferative
disorders 7.1.2 Lymphomatoid papulosis
Primary cutaneous CD30+ lymphoproliferative disorders rep- LyP is characterized by recurrent, papulonodular eruptions that
resent a spectrum of disorders, and the diagnosis is depen- resolve spontaneously to leave varioliform scars. The pathology
dent on careful assessment of both clinical and pathological shows variable proportions of CD30+ large cells (type A–D).
features. Specifically, MF must be excluded by a clinical
evaluation that fails to identify pre-existing or coexistent 7.1.2.1 Expectant policy
polymorphic patches and plaques in a characteristic limb- For patients with limited disease and infrequent eruptions, an
girdle distribution. There is only one RCT involving this active monitoring policy is acceptable.
condition, and the evidence is based on the EORTC, ISCL
and United States Cutaneous Lymphoma Consortium consen- 7.1.2.2 Phototherapy
sus statement,15 which is the appropriate source for refer- Phototherapy is effective in LyP but there is a lack of data on
ences supporting all the treatment recommendations below. dosage, and relapse is common when treatment is withdrawn.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 17

difficult to distinguish from reactive lymphoid proliferations.


7.1.2.3 Chemotherapy
Surgical excision and local radiotherapy are acceptable treat-
Weekly low-dose oral methotrexate is highly effective in LyP,
ment options.245 Rare CD8+ acral CTCL variants also have an
but only a minority of patients experience a durable remission
excellent prognosis and can also be treated with surgical exci-
following treatment withdrawal.15
sion or radiotherapy.246 However, the disease may follow a
more aggressive path in patients who present with large
7.1.2.4 Radiotherapy
rapidly progressive tumours or multifocal disease; this requires
Radiotherapy can be very effective especially for patients with
treatment as for PTCLs.247
regional LyP, and responses can be durable.15

7.1.2.5 Immunobiological therapy 7.2.3 CD8+ aggressive epidermotropic cytotoxic cutaneous


There are very limited data on retinoids, interferon-alpha and T-cell lymphoma and primary cutaneous cd T-cell
bexarotene, but responses have been reported with all of these lymphoma
agents in LyP. Brentuximab is effective.160
These rare CTCL variants are diagnosed on the basis of typical
clinicopathological features with usually widespread ulcerated
7.2 Rare cutaneous T-cell lymphoma variants plaques. Systemic dissemination (central nervous system) is
common. Multiagent chemotherapy options such as CHOP-like
There are various rare and aggressive CTCL variants including
regimens are appropriate, and TSEB can be an effective pallia-
cd CTCL and PTCL (not otherwise specified, NOS), which can
tive approach. Transplant options should be considered for eli-
primarily involve cutaneous tissues. The diagnosis in such
gible patients.
cases requires careful evaluation of the clinical features, stag-
ing investigations and pathology. Human T-lymphotropic
virus-1 virology should be considered to exclude a cutaneous
presentation of adult T-cell leukaemias/lymphoma; there are Recommendations
no controlled trial data. Treatment options are the same as • Primary cutaneous CD30+ lymphoproliferative disor-
those recommended in the British Committee for Standards in ders are diagnosed on the basis of careful clinicopatho-
Haematology 2013 guidelines.240 logical correlation. Staging investigations are not
Rare CTCL variants such as subcutaneous panniculitis-like T- appropriate for LyP but are essential for ALCL or bor-
cell lymphoma (SPTCL), CD4+ small/medium T-cell lympho- derline cases. (Strength of recommendation D (GPP))
proliferative disorder, acral CD8+ T-cell lymphoma and • Treatment for LyP ranges from an expectant policy for
aggressive epidermotropic cytotoxic CD8+ CTCL are character- patients with limited disease to phototherapy, radio-
ized by distinct clinical and/or pathological features and, with therapy, low-dose methotrexate or interferon-alpha for
the exception of the latter, have an excellent prognosis. patients with extensive disease. (Strength of recommendation
C)
7.2.1 Subcutaneous panniculitis-like T-cell lymphoma • Treatment for CD30+ pcALCL consists of surgical exci-
sion and/or radiotherapy for localized disease. (Strength
SPTCL is a rare CTCL variant characterized by subcutaneous of recommendation B)
plaques and tumours and cytologically atypical CD8+ T cells • Combination chemotherapy or brentuximab may be
rimming fat lobules.241 Localized disease may respond to appropriate for patients with CD30+ pcALCL with
radiotherapy. Patients may be steroid responsive but are extensive cutaneous disease or those with systemic pro-
likely to relapse, although immunosuppressive drug therapy gression. (Strength of recommendation B)
has been more effective than chemotherapy for some • Treatment of rare CTCL variants depends on the sub-
patients.242 Patients with haemophagocytic syndrome and type, as some (SPTCL, CD4+ small/medium pleomor-
steroid-resistant disease respond to doxorubicin-based phic, CD8+ acral T-cell lymphoma) are indolent with
chemotherapy regimens (CHOP), with durable remissions an excellent prognosis, while others are aggressive and
reported.241 Durable responses have also been reported fol- may require chemotherapy and consideration for stem
lowing high-dose chemotherapy and stem cell transplanta- cell transplant in line with recommendations for sys-
tion.243 Ciclosporin may be effective in patients failing temic PTCL (NOS). (Strength of recommendation C)
cytotoxic chemotherapy.244 • Primary cutaneous PTCLs (NOS) are rare and heteroge-
neous in behaviour. Some present with limited skin
disease that can be managed with skin-directed options
7.2.2 CD4+ small/medium pleomorphic T-cell
such as radiotherapy, while others present with exten-
lymphoproliferative disorder and primary cutaneous CD8+
sive or bulky skin disease and will require treatment
acral T-cell lymphoma
options as for systemic PTCL (NOS). (Strength of recom-
Primary cutaneous small/medium CD4+ lymphoproliferative mendation C)
disorder has an excellent prognosis, although it can be

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


18 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

that the radiotherapy CTV should be the tumour plus a 15-


8.0 Treatment of primary cutaneous B-cell cm margin of normal skin around the tumour.238
lymphomas In the U.K., the standard radiotherapy dose for indolent
For the purpose of this guideline, separate recommendations non-Hodgkin lymphoma is 24 Gy in 12 fractions, based on
have been made for low-grade lymphomas (PCMZL and the results of a multicentre, prospective RCT comparing 40 Gy
PCFCL), high-grade PCLBCL and the rare PCLBCL (NOS). vs. 24 Gy in 2-Gy fractions for indolent follicular and mar-
PCMZL and PCFCL have an excellent prognosis, and although ginal zone lymphoma.252 Lower doses or shorter fractionation
cutaneous relapses are frequent, systemic spread is rare. schedules (e.g. 8 Gy in two fractions for multiple lesions;
PCLBCLs are aggressive lymphomas with considerable mortal- 15 Gy in five fractions for solitary lesions) have been used in
ity and require systemic chemotherapy in most instances. many centres with good results.253 The Leiden group have
There are no RCTs but there are published EORTC consensus reported complete response rates of 72% and re-treatment
guidelines.11 rates of 29% with 4 Gy in two fractions, and complete
response achieved in all patients retreated with 20 Gy in eight
fractions.106 A U.K. RCT has shown that 4 Gy in two fractions
8.1 Primary cutaneous marginal zone and follicle centre
can be effective in the palliative setting, with an ORR of
lymphoma
741% (complete response rate 443%). However, this was
significantly inferior to 24 Gy in 12 fractions, with an ORR of
8.1.1 Excision
81% and complete response rate of 603%.254 The OS rate
Complete surgical excision may be considered for small was the same with both fractionations but there were fewer
lesions for both diagnosis and therapy, but there is no litera- patients with PCMZL or PCFCL in this study.
ture to guide margins of resection. In patients with PCMZL
and PCFCL treated with complete excision, over 40% are
8.1.4 Intralesional interferon
reported to relapse.11 Where the lesion has been excised com-
pletely, a policy of monitoring or radiotherapy to the scar There are limited data on the use of interferon-alpha. Vandersee et al.
with a 15-cm margin to CTV around the scar238 can be con- reported a 67% response rate in 15 patients, with 90% of responders
sidered to reduce the risk of local recurrence. In larger lesions relapsing. The median time to relapse was 155 months.255
where complete excision would result in a significant cosmetic
scar or functional defect, a diagnostic biopsy should be per-
8.1.5 Rituximab
formed and followed by radiotherapy.
The limited data on rituximab are based on case reports and
small case series. Treatment is either as systemic therapy
8.1.2 Antibiotics
375 mg m2 intravenously weekly for 4–8 weeks, or as
In patients with PCMZL and positive B. burgdorferi serology intralesional therapy 5–30 mg one to three times per week.
there are reports of complete response following antibiotic For intralesional therapy, the complete response rates for both
treatment.11 The U.K. Health Protection Agency guidance on PCMZL and PCFCL are reported to be 70–90%, with relapse
treatment for Borrelia with oral antibiotics should be followed rates of 40–60%.256 Systemic therapy is preferred for more
before considering other therapies. widespread disease. For systemic therapy, the complete
response rates for PCMZL and PCFCL are similar, ranging from
67% to 875%, but again the relapse rates are relatively high,
8.1.3 Radiotherapy
with a median time to relapse of 25 months.257,258
Radiotherapy is the standard treatment for both PCMZL and
PCFCL. The EORTC consensus recommendations11 reported data
8.1.6 Chemotherapy
on 132 patients with PCMZL and 460 with PCFCL treated with
radiotherapy. Of the patients with PCMZL, 99% achieved com- There are relatively little data published on the results of
plete remission and only 46% of patients showed one or more chemotherapy for PCMZL and PCFCL. For PCMZL, chlorambu-
relapses. Extracutaneous progression was reported in only three cil for multifocal skin lesions is associated with a complete
of 132 patients, one of whom died of lymphoma. The radio- response rate of 64% and relapse rate of 33%, based on
therapy doses reported varied from 10 Gy to 50 Gy, and most reports of 14 patients. CHOP is associated with a complete
studies used a treatment margin around the tumour of 1–5 cm. response rate of 85% and relapse rate of 57%.11
Of the patients with PCFCL, almost all achieved complete For PCFCL, cumulative data on 104 patients from eight
response with radiotherapy. Three major studies in PCFCL studies treated with CHOP or CHOP-like regimens for wide-
combining 147 patients reported a relapse rate of 30% when spread skin lesions or high skin tumour burden give a com-
margins > 2 cm were used.248–250 This differs from the plete response rate of 85% with a relapse rate of 48%.11 Only
relapse rate of 67% in a small study that used margins of 05– five patients received chemotherapy without an anthracycline
1 cm, where frequent in-field and marginal recurrences were (COP or CVP; see section 632). There is a lack of data on
reported.251 On the basis of this evidence, it is recommended the efficacy of rituximab–CVP and rituximab–bendamustine,

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 19

which are the most commonly used regimens in nodal or sys- review of 32 patients from eight separate studies treated
temic indolent lymphomas, in disease confined to the skin. with CHOP-like chemotherapy, 81% achieved complete
As for systemic indolent lymphomas, it is appropriate to response but 56% relapsed.11 Rituximab as a single treat-
manage patients with disease confined to the skin with watch- ment has an ORR of 75% in eight reported cases but all
ful waiting. Current evidence supports a role for single-agent patients relapsed, with a median disease-free survival of
chemotherapy or rituximab for those patients with extensive 525 months.259 Current international consensus and guideli-
cutaneous disease and no systemic involvement. Patients with nes recommend that PCLBCL leg type should be treated like
systemic disease or high disease burden failing to respond to a systemic diffuse large B-cell lymphoma (DLBCL), with
single-agent systemic therapy should be managed according to three to six cycles of R-CHOP and local radiotherapy of
local guidelines for the management of systemic disease. 30–36 Gy.6

Recommendations 8.2.2 Primary cutaneous diffuse large B-cell lymphoma


(not otherwise specified)
• Solitary lesions should be treated with the intention of
cure. (Strength of recommendation B) PCLBCL (NOS) includes rare morphological variants of DLBCL
• Small individual skin lesions can be completely excised such as anaplastic lymphoma, plasmablastic lymphoma, T-
and monitored, but treatment with radiotherapy (tu- cell/histiocyte rich large B-cell lymphoma and intravascular
mour or scar with a 2-cm margin of skin to define large B-cell lymphoma, which may rarely present primarily in
CTV) is preferred to reduce the risk of local relapse. the skin.3 These variants should be treated with systemic lym-
(Strength of recommendation B) phoma chemotherapy protocols.
• Following a diagnostic biopsy, the first-line treatment
for individual skin lesions should be radiotherapy. The
radiotherapy CTV should be the tumour plus a 15-cm Recommendations
margin of normal skin around the tumour. (Strength of
recommendation B)
• PCLBCL leg type should be treated with R-CHOP
chemotherapy followed by local radiotherapy as per
• Active monitoring is an option for patients with local guidelines for systemic DLBCL. Current protocols
asymptomatic disease. (Strength of recommendation D (GPP)) recommend three to six cycles of R-CHOP chemother-
• Systemic chemotherapy should be reserved for apy followed by 30–36 Gy in 2-Gy fractions of local
patients with widespread extensive lesions or high radiotherapy. (Strength of recommendation B)
tumour burden (stage T2c–T3), or patients with
nodal or visceral progression. Treatment is palliative,
• Palliative radiotherapy alone can be considered for
patients unfit for chemotherapy. (Strength of recommenda-
and for disease confined to the skin consideration tion D)
should be given to single-agent rituximab, or chlo-
rambucil, as this is well tolerated and effective. Fur-
• PCLBCL (NOS) should be treated as per local guidelines
for systemic DLBCL. (Strength of recommendation B)
ther choice of chemotherapy should be in line with
local guidelines for the management of indolent lym- Future research recommendation
phomas. (Strength of recommendation C)
• Patients who relapse with nodal or visceral lymphoma • Future trials should concentrate on improving clinical
should be treated according to local and national man- outcomes for patients with PCLBCL. (Strength of recommen-
agement guidelines for systemic lymphoma. (Strength of dation D (GPP))
recommendation B)

9.0 Economic and practical considerations


• Primary cutaneous lymphomas are rare and consequently
the economic costs of treatment are modest, especially for
8.2 Primary cutaneous diffuse large B-cell lymphoma
early stages of CTCL and primary cutaneous B-cell lym-
phomas. In contrast, advanced cutaneous T-cell lym-
8.2.1 Primary cutaneous diffuse large B-cell lymphoma leg
phomas require complex and expensive interventions,
type
often with only palliative intent.
The number of reported cases of PCLBCL is small. The • There has been a growing difference between the U.S.A.
response rate to radiotherapy alone is high, but relapse rates and European Union for approval of novel drugs. How-
are ≥ 58% with a significant risk of extracutaneous ever, the emergence of several pharmaceutically sponsored
recurrences.11 Data on CHOP-like chemotherapy, R-CHOP phase III RCTs in the last few years suggests that this issue
and R-CHOP plus local radiotherapy are limited to small is likely to be resolved, allowing U.K. patients to have
numbers of patients and short follow-up periods. In a access to these new treatments.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


20 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

Fig 1. Treatment guidelines for mycosis fungoides. EBRT, external beam radiotherapy with photons or electrons for lymph node, soft tissue or
visceral lymphoma; ECP, extracorporeal photopheresis; IFN, interferon; MTX, methotrexate; PD, progressive disease; RIC-allo-SCT, reduced-
intensity allogeneic stem cell transplantation; SDT, skin-directed therapy (topical steroids, ultraviolet B, psoralen–ultraviolet A, skin radiotherapy,
topical nitrogen mustard); TSEB, total skin electron beam radiotherapy. Skin radiotherapy indicates superficial radiotherapy or EBRT to skin
patches, plaques and tumours. #Supranetwork: refers to the supranetwork multidisciplinary team (MDT) meeting for treatment decision. *PD and
exhausted first- and second-line options. **Chemotherapy as recommended by the supranetwork MDT. ***Consider only if the patient has
durable complete response. ↔ indicates that after treatment, patients may respond to treatments included in earlier ‘line’ options. Patients can
move between first- and second-line options.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 21

Fig 2. Treatment guidelines for Sezary syndrome. Palliative skin-directed therapy can be used for symptom control if required. Chemotherapy is as directed
by the supranetwork multidisciplinary team (MDT). EBRT, external beam radiotherapy; ECP, extracorporeal photopheresis; IFN, interferon; MTX,
methotrexate; PD, progressive disease; RIC-allo-SCT, reduced-intensity allogeneic stem cell transplantation; WCC, white cell count, 9 109. #Supranetwork: refer
to the supranetwork MDT meeting for treatment decision. †PD and exhausted first- and second-line options. ††May be used in combination.

• Because of disease rarity, few large RCTs have been con- • There is an urgent need for the identification of novel tar-
cluded. This situation is now improving as both the FDA gets in CTCL based on improved understanding of disease
and EMA have adopted the same approach for orphan drug pathogenesis.
indications, but for some interventions such as stem cell
transplantation the evidence base will develop only 11.0 Recommended audit points
through consistent use of data registries.
In the last 20 consecutive patients seen with primary cuta-
neous T-cell lymphoma:
10.0 Future directions
1 Was the WHO 2016 classification used in recording the
• Low-risk, early-stage IA–IIA MF: future trials should
following?
address the need to improve duration of response without
increasing toxicity following PUVA and TSEB. a Cutaneous lymphoma subtype.
• Early-stage IA–IIA MF refractory to SDT: future trials b Tumour phenotype.
should address the need to improve specific clinical end c Folliculotropism in MF.
points such as time to relapse or progression, PFS and OS. d Large cell transformation in MF.
• High-risk advanced-stage IIB–IV MF/SS: future trials are
2 Were patients staged appropriately?
required to address an urgent need to improve PFS and OS.
3 Is there a record of performance status?
• The conditioning regimens and outcomes of RIC allo-
4 Is there a record of Sezary cell count, CD4 : CD8 ratio or
HSCT for selected groups of patients with advanced disease
abnormal T-cell phenotype on flow cytometry at diagno-
should be collected through data registries such as the
sis and clinical evolution?
EBMT registry, and large cohort studies should be pub-
5 Is there an annual log of a cumulative treatment
lished with adequate follow-up.
summary?

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


22 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

Fig 3. Treatment guidelines for other cutaneous T-cell lymphoma (CTCL). LyP, lymphomatoid papulosis; NK, natural killer; NOS, not otherwise
specified; pc, primary cutaneous; SDT, skin-directed therapy (topical steroids, ultraviolet B, psoralen–ultraviolet A, skin radiotherapy).

The audit recommendation of 20 cases per department is to


reduce variation in the results due to a single patient, and to
13.0 Stakeholder involvement and peer review
allow benchmarking between different units. However, The GDG consisted of consultant clinical dermatologists, haema-
departments unable to achieve this recommendation may tologists and oncologists. The draft document was circulated to
choose to audit all cases seen in the preceding 12 months. the BAD membership and the British Society for Haematology,
Royal College of Pathologists, British Dermatological Nursing
Group, Primary Care Dermatological Society, Royal College of
12.0 Summary
Radiologists, British Lymphoma Pathology Group, British Soci-
Figures 1–4 show treatment guidelines for MF, SS, other ety for Dermatopathology, U.K. Lymphoma Association and
CTCLs and CBCL, respectively. See the manuscript for details Guy’s & St Thomas’ NHS Foundation Trust Skin Lymphoma Dis-
of the evidence. cussion Group for comments, and was peer reviewed by the

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 23

Fig 4. Treatment guidelines for cutaneous B-cell lymphoma. PCFCL, primary cutaneous follicle centre lymphoma; PCLBCL, primary cutaneous
diffuse large B-cell lymphoma; PCMZL, primary cutaneous marginal zone lymphoma; R-bendamustine, rituximab with bendamustine; RCVP,
rituximab, cyclophosphamide, vincristine and prednisolone; R-CHOP, rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone.
§
Treatment is palliative; further therapy should be in line with local guidelines for the management of indolent lymphomas. §§In line with local
guidelines for systemic diffuse large B-cell lymphoma.

Clinical Standards Unit of the BAD (made up of the Therapy &


Guidelines Subcommittee) prior to publication.
15.0 Plans for guideline revision
The proposed revision date for this set of recommendations is
scheduled for 2023; where necessary, important interim
14.0 Limitations of the guidelines changes will be updated on the BAD website: http://
This document has been prepared on behalf of the BAD and is www.bad.org.uk/healthcare-professionals/clinical-standards/
based on the best data available when the document was pre- clinical-guidelines.
pared. Please check the BAD website for any updates (http://
www.bad.org.uk/healthcare-professionals/clinical-standards/ Acknowledgments
clinical-guidelines).260 It is recognized that under certain con-
We are very grateful to Dr Jennie Wimperis (British Society
ditions it may be necessary to deviate from the guidelines
for Haematology) for her input and our U.K. CLG colleagues
and that the results of future studies may require some of
Eve Gallop Evans, Eleanor James and Richard Cowan, as well
the recommendations herein to be changed. Failure to
as everyone including the patient organizations who com-
adhere to these guidelines should not necessarily be consid-
mented on the draft during the consultation period.
ered negligent, nor should adherence to these recommenda-
tions constitute a defence against a claim of negligence.
Limiting the review to English-language references was a References
pragmatic decision but the authors recognize this may 1 Bell HK, Ormerod AD. Writing a British Association of Dermatol-
exclude some important information published in other ogists clinical guideline: an update on the process and guidance
languages. for authors. Br J Dermatol 2009; 160:725–8.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


24 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

2 Brouwers M, Kho ME, Browman GP et al. AGREE II: advancing 21 Scarisbrick JJ, Prince HM, Vermeer MH et al. Cutaneous Lym-
guideline development, reporting and evaluation in healthcare. phoma International Consortium study of outcome in advanced
Can Med Assoc J 2010; 182:E839–42. stages of mycosis fungoides and Sezary syndrome: effect of speci-
3 Willemze R, Jaffe ES, Burg G et al. WHO-EORTC classification for fic prognostic markers on survival and development of a prog-
cutaneous lymphomas. Blood 2005; 105:3768–85. nostic model. J Clin Oncol 2015; 33:3766–73.
4 Trautinger F, Eder J, Assaf C et al. European Organisation for 22 Bunn PA, Lamberg SI. Report of the committee on staging and
Research and Treatment of Cancer consensus recommendations classification of cutaneous T-cell lymphomas. Cancer Treat Rep
for the treatment of mycosis fungoides/Sezary syndrome – 1979; 63:725–8.
update 2017. Eur J Cancer 2017; 77:57–74. 23 Olsen E, Vonderheid E, Pimpinelli N et al. Revisions to the staging
5 Swerdlow SH, Campo E, Harris NL. WHO Classification of Tumours of and classification of mycosis fungoides and Sezary syndrome: a
Haematopoietic and Lymphoid Tissues, 4th edn. Lyon: IARC, 2017. proposal of the International Society for Cutaneous Lymphomas
6 National Comprehensive Cancer Network. NCCN guidelines. (ISCL) and the cutaneous lymphoma task force of the European
Available at: https://www.nccn.org/professionals/physician_gls/ Organization of Research and Treatment of Cancer (EORTC). Blood
f_guidelines.asp (last accessed 8 October 2018). 2007; 110:1713–22.
7 Willemze R, Hodak E, Zinzani PL et al. Primary cutaneous 24 Kim YH, Willemze R, Pimpinelli N et al. TNM classification
lymphomas: ESMO Clinical Practice Guidelines for diagnosis, system for primary cutaneous lymphomas other than mycosis
treatment and follow-up. Ann Oncol 2018; 29 (Suppl. 4):iv30–40. fungoides and Sezary syndrome: a proposal of the International
8 National Cancer Registration and Analysis Service. NCIN data Society for Cutaneous Lymphomas (ISCL) and the Cutaneous
briefings. Available at: http://www.ncin.org.uk/publications/da Lymphoma Task Force of the European Organization of
ta_briefings (last accessed 8 October 2018). Research and Treatment of Cancer (EORTC). Blood 2007;
9 Vose J, Armitage J, Weisenburger D et al. International peripheral 110:479–84.
T-cell and natural killer/T-cell lymphoma study: pathology find- 25 American Joint Committee on Cancer. AJCC Cancer Staging Manual,
ings and clinical outcomes. J Clin Oncol 2008; 26:4124–30. 8th edn. Chicago: American Joint Committee on Cancer/Springer,
10 British HIV Association. U.K. National Guidelines for HIV testing 2016.
2008. Available at: https://www.bhiva.org/HIV-testing-guidelines 26 Zackheim HS, Amin S, Kashani-Sabet M et al. Prognosis in cuta-
(last accessed 8 October 2018). neous T-cell lymphoma by skin stage: long-term survival in 489
11 Senff NJ, Noordijk EM, Kim YH et al. European Organization patients. J Am Acad Dermatol 1999; 40:418–25.
for Research and Treatment of Cancer and International Society 27 van Doorn R, Van Haselen CW, van Voorst Vader PC et al. Myco-
for Cutaneous Lymphoma consensus recommendations for the sis fungoides: disease evolution and prognosis of 309 Dutch
management of cutaneous B-cell lymphomas. Blood 2008; patients. Arch Dermatol 2000; 136:504–10.
112:1600–9. 28 de Coninck EC, Kim YH, Varghese A et al. Clinical characteristics
12 Goodlad JR, Davidson MM, Hollowood K et al. Primary cutaneous and outcome of patients with extracutaneous mycosis fungoides.
B-cell lymphoma and Borrelia burgdorferi infection in patients from J Clin Oncol 2001; 19:779–84.
the Highlands of Scotland. Am J Surg Pathol 2000; 24:1279–85. 29 Kim YH, Liu HL, Mraz-Gernhard S et al. Long-term outcome of
13 Lukowsky A, Marchwat M, Sterry W et al. Evaluation of B-cell 525 patients with mycosis fungoides and Sezary syndrome: clini-
clonality in archival skin biopsy samples of cutaneous B-cell lym- cal prognostic factors and risk for disease progression. Arch Derma-
phoma by immunoglobulin heavy chain gene polymerase chain tol 2003; 139:857–66.
reaction. Leuk Lymphoma 2006; 47:487–93. 30 Agar NS, Wedgeworth E, Crichton S et al. Survival outcomes and
14 Bunn PA, Huberman MS, Whang-Peng J et al. Prospective staging prognostic factors in mycosis fungoides/Sezary syndrome: valida-
evaluation of patients with cutaneous T-cell lymphomas. Demon- tion of the revised International Society for Cutaneous Lym-
stration of a high frequency of extracutaneous dissemination. Ann phomas/European Organisation for Research and Treatment of
Intern Med 1980; 93:223–30. Cancer staging proposal. J Clin Oncol 2010; 28:4730–9.
15 Kempf W, Pfaltz K, Vermeer MH et al. EORTC, ISCL, and USCLC 31 Bekkenk MW, Geelen FA, van Voorst Vader PC et al. Primary and
consensus recommendations for the treatment of primary cuta- secondary cutaneous CD30+ lymphoproliferative disorders: a
neous CD30-positive lymphoproliferative disorders: lymphoma- report from the Dutch Cutaneous Lymphoma Group on the long-
toid papulosis and primary cutaneous anaplastic large-cell term follow-up data of 219 patients and guidelines for diagnosis
lymphoma. Blood 2011; 118:4024–35. and treatment. Blood 2000; 95:3653–61.
16 Tilly H, Dreyling M. Diffuse large B-cell non-Hodgkin’s lym- 32 Bessell EM, Humber CE, O’Connor S et al. Primary cutaneous B-
phoma: ESMO Clinical Practice Guidelines for diagnosis, treat- cell lymphoma in Nottinghamshire U.K.: prognosis of subtypes
ment and follow-up. Ann Oncol 2010; 21:v172–4. defined in the WHO-EORTC classification. Br J Dermatol 2012;
17 Kuo PH, McClennan BL, Carlson K et al. FDG-PET/CT in the evaluation 167:1118–23.
of cutaneous T-cell lymphoma. Mol Imaging Biol 2008; 10:74–81. 33 Chan SA, Shah F, Chiganti S et al. Primary cutaneous B-cell
18 Tsai EY, Taur A, Espinosa L et al. Staging accuracy in mycosis fun- lymphoma: systemic spread is rare while cutaneous relapses
goides and Sezary syndrome using integrated positron emission and secondary malignancies are frequent. Br J Dermatol 2017;
tomography and computed tomography. Arch Dermatol 2006; 177:287–9.
142:577–84. 34 Benton EC, Crichton S, Talpur R et al. A cutaneous lymphoma
19 Senff NJ, Kluin-Nelemans HC, Willemze R. Results of bone mar- international prognostic index (CLIPi) for mycosis fungoides and
row examination in 275 patients with histological features that Sezary syndrome. Eur J Cancer 2013; 49:2859–68.
suggest an indolent type of cutaneous B-cell lymphoma. Br J Hae- 35 Olsen EA, Whittaker S, Kim YH et al. Clinical end points and
matol 2008; 142:52–6. response criteria in mycosis fungoides and Sezary syndrome: a
20 Quereux G, Frot AS, Brocard A et al. Routine bone marrow biopsy consensus statement of the International Society for Cutaneous
in the initial evaluation of primary cutaneous B-cell lymphoma Lymphomas, the United States Cutaneous Lymphoma Consor-
does not appear justified. Eur J Dermatol 2009; 19:216–20. tium, and the Cutaneous Lymphoma Task Force of the European

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 25

Organisation for Research and Treatment of Cancer. J Clin Oncol 53 Chong A, Loo WJ, Banney L et al. Imiquimod 5% cream in the
2011; 29:2598–607. treatment of mycosis fungoides – a pilot study. J Dermatolog Treat
36 National Institute for Health and Care Excellence. Improving out- 2004; 15:118–19.
comes for people with skin tumours including melanoma. Cancer 54 Deeths MJ, Chapman JT, Dellavalle RP et al. Treatment of patch
service guideline CSG8. Available at: https://www.nice.org.uk/ and plaque stage mycosis fungoides with imiquimod 5% cream. J
guidance/csg8 (last accessed 8 October 2018). Am Acad Dermatol 2005; 52:275–80.
37 National Institute for Health and Care Excellence. Improving out- 55 Coors EA, Schuler G, Von Den Driesch P. Topical imiquimod as
comes for people with skin tumours including melanoma (up- treatment for different kinds of cutaneous lymphoma. Eur J Derma-
date). Available at: https://www.nice.org.uk/guidance/csg8/ tol 2006; 16:391–3.
resources/improving-outcomes-for-people-with-skin-tumours-inc 56 Ardigo M, Cota C, Berardesca E. Unilesional mycosis fun-
luding-melanoma-2010-partial-update-773380189 (last accessed goides successfully treated with imiquimod. Eur J Dermatol 2006;
8 October 2018). 16:446.
38 Quaglino P, Maule M, Prince HM et al. Global patterns of care in 57 Ariffin N, Khorshid M. Treatment of mycosis fungoides with imi-
advanced stage mycosis fungoides/Sezary syndrome: a multicen- quimod 5% cream. Clin Exp Dermatol 2006; 31:822–3.
ter retrospective follow-up study from the Cutaneous Lymphoma 58 Chiam LY, Chan YC. Solitary plaque mycosis fungoides on the
International Consortium. Ann Oncol 2017; 28:2517–25. penis responding to topical imiquimod therapy. Br J Dermatol
39 Zackheim HS, Kashani-Sabet M, Amin S. Topical corticosteroids 2007; 156:560–2.
for mycosis fungoides. Experience in 79 patients. Arch Dermatol 59 Martinez-Gonzalez MC, Verea-Hernando MM, Yebra-Pimentel MT et al.
1998; 134:949–54. Imiquimod in mycosis fungoides. Eur J Dermatol 2008; 18:148–52.
40 Lessin SR, Duvic M, Guitart J et al. Topical chemotherapy in cuta- 60 Kannangara AP, Levitan D, Fleischer AB. Six patients with early-
neous T-cell lymphoma: positive results of a randomized, con- stage cutaneous T-cell lymphoma successfully treated with topical
trolled, multicenter trial testing the efficacy and safety of a novel 5-fluorouracil. J Drugs Dermatol 2010; 9:1017–18.
mechlorethamine, 0.02%, gel in mycosis fungoides. JAMA Dermatol 61 Aires D, Shaath T, Fraga G et al. Safe and efficacious use of a topi-
2013; 149:25–32. cal retinoid under occlusion for the treatment of mycosis fun-
41 Zachariae H, Thestrup-Pedersen K, Sogaard H. Topical nitrogen goides. J Drugs Dermatol 2014; 13:1479–80.
mustard in early mycosis fungoides. A 12-year experience. Acta 62 Rallis E, Economidi A, Verros C et al. Successful treatment of
Derm Venereol 1985; 65:53–8. patch type mycosis fungoides with tacrolimus ointment 0.1%. J
42 Hoppe RT, Abel EA, Deneau DG et al. Mycosis fungoides: man- Drugs Dermatol 2006; 5:906–7.
agement with topical nitrogen mustard. J Clin Oncol 1987; 63 Duvic M, Olsen EA, Omura GA et al. A phase III, randomized,
5:1796–803. double-blind, placebo-controlled study of peldesine (BCX-34)
43 Ramsay DL, Halperin PS, Zeleniuch-Jacquotte A. Topical mechlor- cream as topical therapy for cutaneous T-cell lymphoma. J Am
ethamine therapy for early stage mycosis fungoides. J Am Acad Der- Acad Dermatol 2001; 44:940–7.
matol 1988; 19:684–91. 64 Milstein HJ, Vonderheid EC, Van Scott EJ et al. Home ultraviolet
44 Vonderheid EC, Tan ET, Kantor AF et al. Long-term efficacy, cura- phototherapy of early mycosis fungoides: preliminary observa-
tive potential, and carcinogenicity of topical mechlorethamine tions. J Am Acad Dermatol 1982; 6:355–62.
chemotherapy in cutaneous T cell lymphoma. J Am Acad Dermatol 65 Ramsay DL, Lish KM, Yalowitz CB et al. Ultraviolet-B photother-
1989; 20:416–28. apy for early-stage cutaneous T-cell lymphoma. Arch Dermatol
45 Kim YH, Martinez G, Varghese A et al. Topical nitrogen mustard 1992; 128:931–3.
in the management of mycosis fungoides: update of the Stanford 66 Resnik KS, Vonderheid EC. Home UV phototherapy of early
experience. Arch Dermatol 2003; 139:165–73. mycosis fungoides: long-term follow-up observations in thirty-
46 Lindahl LM, Fenger-Gron M, Iversen L. Topical nitrogen mustard one patients. J Am Acad Dermatol 1993; 29:73–7.
therapy in patients with mycosis fungoides or parapsoriasis. J Eur 67 Clark C, Dawe RS, Evans AT et al. Narrowband TL-01 photother-
Acad Dermatol Venereol 2013; 27:163–8. apy for patch-stage mycosis fungoides. Arch Dermatol 2000;
47 de Quatrebarbes J, Esteve E, Bagot M et al. Treatment of early- 136:748–52.
stage mycosis fungoides with twice-weekly applications of 68 Gathers RC, Scherschun L, Malick F et al. Narrowband UVB pho-
mechlorethamine and topical corticosteroids: a prospective study. totherapy for early-stage mycosis fungoides. J Am Acad Dermatol
Arch Dermatol 2005; 141:1117–20. 2002; 47:191–7.
48 Zackheim HS, Epstein EH, Crain WR. Topical carmustine (BCNU) 69 Pavlotsky F, Barzilai A, Kasem R et al. UVB in the management of
for cutaneous T cell lymphoma: a 15-year experience in 143 early stage mycosis fungoides. J Eur Acad Dermatol Venereol 2006;
patients. J Am Acad Dermatol 1990; 22:802–10. 20:565–72.
49 Apisarnthanarax N, Wood GS, Stevens SR et al. Phase I clinical 70 Ponte P, Serrao V, Apetato M. Efficacy of narrowband UVB vs.
trial of O6-benzylguanine and topical carmustine in the treatment PUVA in patients with early-stage mycosis fungoides. J Eur Acad
of cutaneous T-cell lymphoma, mycosis fungoides type. Arch Der- Dermatol Venereol 2010; 24:716–21.
matol 2012; 148:613–20. 71 Boulos S, Vaid R, Aladily TN et al. Clinical presentation,
50 Breneman D, Duvic M, Kuzel T et al. Phase 1 and 2 trial of bexar- immunopathology, and treatment of juvenile-onset mycosis fun-
otene gel for skin-directed treatment of patients with cutaneous goides: a case series of 34 patients. J Am Acad Dermatol 2014;
T-cell lymphoma. Arch Dermatol 2002; 138:325–32. 71:1117–26.
51 Heald P, Mehlmauer M, Martin AG et al. Topical bexarotene ther- 72 Zane C, Leali C, Airo P et al. ‘High-dose’ UVA1 therapy of wide-
apy for patients with refractory or persistent early-stage cutaneous spread plaque-type, nodular, and erythrodermic mycosis fun-
T-cell lymphoma: results of the phase III clinical trial. J Am Acad goides. J Am Acad Dermatol 2001; 44:629–33.
Dermatol 2003; 49:801–15. 73 Briffa DV, Warin AP, Harrington CI et al. Photochemotherapy
52 Shipman AR, Scarisbrick J. New treatment options for mycosis in mycosis fungoides. A study of 73 patients. Lancet 1980; 2:49–
fungoides. Indian J Dermatol 2016; 61:119. 53.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


26 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

74 Herrmann JJ, Roenigk HH, Hurria A et al. Treatment of mycosis versus interferon-2a plus PUVA in patients with cutaneous T-cell
fungoides with photochemotherapy (PUVA): long-term follow- lymphoma stages I and II. Blood 1998; 92:3578–81.
up. J Am Acad Dermatol 1995; 33:234–42. 93 Kuzel TM, Roenigk HH, Samuelson E et al. Effectiveness of inter-
75 Oguz O, Engin B, Aydemir EH. The influence of psoralen + feron alfa-2a combined with phototherapy for mycosis fungoides
ultraviolet A treatment on the duration of remission and prog- and the Sezary syndrome. J Clin Oncol 1995; 13:257–63.
nosis in mycosis fungoides. J Eur Acad Dermatol Venereol 2003; 94 H€usken AC, Tsianakas A, Hensen P et al. Comparison of pegylated
17:483–5. interferon a-2b plus psoralen PUVA versus standard interferon a-
76 Anadolu RY, Birol A, Sanli H et al. Mycosis fungoides and Sezary 2a plus PUVA in patients with cutaneous T-cell lymphoma. J Eur
syndrome: therapeutic approach and outcome in 113 patients. Int Acad Dermatol Venereol 2012; 26:71–8.
J Dermatol 2005; 44:559–65. 95 Chiarion-Sileni V, Bononi A, Fornasa CV et al. Phase II trial of
77 Whittaker S, Ortiz P, Dummer R et al. Efficacy and safety of bex- interferon-a-2a plus psolaren with ultraviolet light A in patients
arotene combined with psoralen-ultraviolet A (PUVA) compared with cutaneous T-cell lymphoma. Cancer 2002; 95:569–75.
with PUVA treatment alone in stage IB–IIA mycosis fungoides: 96 Rupoli S, Goteri G, Pulini S et al. Long-term experience with low-
final results from the EORTC Cutaneous Lymphoma Task Force dose interferon-a and PUVA in the management of early mycosis
phase III randomized clinical trial (NCT00056056). Br J Dermatol fungoides. Eur J Haematol 2005; 75:136–45.
2012; 167:678–87. 97 Nikolaou V, Siakantaris MP, Vassilakopoulos TP et al. PUVA plus
78 Roenigk HH, Kuzel TM, Skoutelis AP et al. Photochemotherapy interferon a2b in the treatment of advanced or refractory to
alone or combined with interferon alpha-2a in the treatment PUVA early stage mycosis fungoides: a case series. J Eur Acad Der-
of cutaneous T-cell lymphoma. J Invest Dermatol 1990; 95:198S– matol Venereol 2011; 25:354–7.
205S. 98 Wozniak MB, Tracey L, Ortiz-Romero PL et al. Psoralen plus ultravi-
79 Honigsmann H, Brenner W, Rauschmeier W et al. Pho- olet A  interferon-a treatment resistance in mycosis fungoides:
tochemotherapy for cutaneous T cell lymphoma. A follow-up the role of tumour microenvironment, nuclear transcription factor-
study. J Am Acad Dermatol 1984; 10:238–45. jB and T-cell receptor pathways. Br J Dermatol 2009; 160:92–102.
80 Querfeld C, Rosen ST, Kuzel TM et al. Long-term follow-up of 99 Thomsen K, Hammar H, Molin L et al. Retinoids plus PUVA
patients with early-stage cutaneous T-cell lymphoma who (RePUVA) and PUVA in mycosis fungoides, plaque stage. A
achieved complete remission with psoralen plus UV-A monother- report from the Scandinavian Mycosis Fungoides Group. Acta Derm
apy. Arch Dermatol 2005; 141:305–11. Venereol 1989; 69:536–8.
81 Sanchez MA, Gonzalez T, Gaitan MF et al. Is PUVA maintenance 100 Papadavid E, Antoniou C, Nikolaou V et al. Safety and efficacy of
therapy necessary in patients with early-stage mycosis fungoides? low-dose bexarotene and PUVA in the treatment of patients with
Evaluation of a treatment guideline over a 28-month follow-up. mycosis fungoides. Am J Clin Dermatol 2008; 9:169–73.
Int J Dermatol 2011; 50:1086–93. 101 Mostow EN, Neckel SL, Oberhelman L et al. Complete remissions
82 Dogra S, Mahajan R. Phototherapy for mycosis fungoides. Indian J in psoralen and UV-A (PUVA)-refractory mycosis fungoides-type
Dermatol Venereol Leprol 2015; 81:124–35. cutaneous T-cell lymphoma with combined interferon alfa and
83 Stern RS. The risk of melanoma in association with long-term PUVA. Arch Dermatol 1993; 129:747–52.
exposure to PUVA. J Am Acad Dermatol 2001; 44:755–61. 102 Singh F, Lebwohl MG. Cutaneous T-cell lymphoma treatment
84 Stern RS, Nichols KT, Vakeva LH. Malignant melanoma in patients using bexarotene and PUVA: a case series. J Am Acad Dermatol
treated for psoriasis with methoxsalen (psoralen) and ultraviolet 2004; 51:570–3.
A radiation (PUVA). The PUVA Follow-Up Study. N Engl J Med 103 Cotter GW, Baglan RJ, Wasserman TH et al. Palliative radiation
1997; 336:1041–5. treatment of cutaneous mycosis fungoides – a dose response. Int J
85 Ling TC, Clayton TH, Crawley J et al. British Association of Der- Radiat Oncol Biol Phys 1983; 9:1477–80.
matologists and British Photodermatology Group guidelines for 104 Kirova YM, Piedbois Y, Haddad E et al. Radiotherapy in the man-
the safe and effective use of psoralen–ultraviolet A therapy 2015. agement of mycosis fungoides: indications, results, prognosis.
Br J Dermatol 2016; 174:24–55. Twenty years experience. Radiother Oncol 1999; 51:147–51.
86 Kim ST, Kang DY, Kang JS et al. Photodynamic therapy with 105 Hoppe RT. Mycosis fungoides: radiation therapy. Dermatol Ther
methyl-aminolaevulinic acid for mycosis fungoides. Acta Derm 2003; 16:347–54.
Venereol 2012; 92:264–8. 106 Neelis KJ, Schimmel EC, Vermeer MH et al. Low-dose palliative
87 Quereux G, Brocard A, Saint-Jean M et al. Photodynamic therapy radiotherapy for cutaneous B- and T-cell lymphomas. Int J Radiat
with methyl-aminolevulinic acid for paucilesional mycosis fun- Oncol Biol Phys 2009; 74:154–8.
goides: a prospective open study and review of the literature. J 107 Thomas TO, Agrawal P, Guitart J et al. Outcome of patients trea-
Am Acad Dermatol 2013; 69:890–7. ted with a single-fraction dose of palliative radiation for cuta-
88 Meisenheimer JL. Treatment of mycosis fungoides using a 308- neous T-cell lymphoma. Int J Radiat Oncol Biol Phys 2013; 85:747–
nm excimer laser: two case studies. Dermatol Online J 2006; 12:11. 53.
89 Mori M, Campolmi P, Mavilia L et al. Monochromatic excimer 108 Wilson LD, Kacinski BM, Jones GW. Local superficial radiother-
light (308 nm) in patch-stage IA mycosis fungoides. J Am Acad apy in the management of minimal stage IA cutaneous T-cell
Dermatol 2004; 50:943–5. lymphoma (mycosis fungoides). Int J Radiat Oncol Biol Phys 1998;
90 Deaver D, Cauthen A, Cohen G et al. Excimer laser in the treat- 40:109–15.
ment of mycosis fungoides. J Am Acad Dermatol 2014; 70:1058–60. 109 Micaily B, Miyamoto C, Kantor G et al. Radiotherapy for unile-
91 Felix P, Marwan D, Aviv B. The potential of narrow band UVB to sional mycosis fungoides. Int J Radiat Oncol Biol Phys 1998; 42:361–
induce sustained durable complete remission off-therapy in stage 4.
I mycosis fungoides. J Am Acad Dermatol 2018; https://doi.org/10. 110 Ally MS, Pawade J, Tanaka M et al. Solitary mycosis fungoides: a
1016/j.jaad.2018.05.023 distinct clinicopathologic entity with a good prognosis: a series
92 Stadler R, Otte HG, Luger T et al. Prospective randomized multi- of 15 cases and literature review. J Am Acad Dermatol 2012;
center clinical trial on the use of interferon-2a plus acitretin 67:736–44.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 27

111 Piccinno R, Caccialanza M, Percivalle S. Minimal stage IA mycosis 129 Kaye FJ, Bunn PA, Steinberg SM et al. A randomized trial compar-
fungoides. Results of radiotherapy in 15 patients. J Dermatolog Treat ing combination electron-beam radiation and chemotherapy with
2009; 20:165–8. topical therapy in the initial treatment of mycosis fungoides. N
112 Sawyer EJ, Timothy AR. Low dose palliative radiotherapy in low Engl J Med 1989; 321:1784–90.
grade non-Hodgkin’s lymphoma. Radiother Oncol 1997; 42:49–51. 130 Duvic M, Donato M, Dabaja B et al. Total skin electron beam and
113 Stein M, Farrar N, Jones GW et al. Central neurologic involvement non-myeloablative allogeneic hematopoietic stem-cell transplanta-
in mycosis fungoides: ten cases, actuarial risk assessment, and tion in advanced mycosis fungoides and Sezary syndrome. J Clin
predictive factors. Cancer J 2006; 12:55–62. Oncol 2010; 28:2365–72.
114 Bird TG, Whittaker S, Wain EM et al. Temozolomide for central 131 Jones GW, Rosenthal D, Wilson LD. Total skin electron radiation
nervous system involvement in mycosis fungoides. Int J Dermatol for patients with erythrodermic cutaneous T-cell lymphoma (my-
2016; 55:751–6. cosis fungoides and the Sezary syndrome). Cancer 1999;
115 Jones GW, Kacinski BM, Wilson LD et al. Total skin electron radi- 85:1985–95.
ation in the management of mycosis fungoides: consensus of the 132 Wilson LD, Jones GW, Kim D et al. Experience with total skin
European Organization for Research and Treatment of Cancer electron beam therapy in combination with extracorporeal photo-
(EORTC) Cutaneous Lymphoma Project Group. J Am Acad Dermatol pheresis in the management of patients with erythrodermic (T4)
2002; 47:364–70. mycosis fungoides. J Am Acad Dermatol 2000; 43:54–60.
116 Jones GW, Hoppe RT, Glatstein E. Electron beam treatment for 133 Introcaso CE, Micaily B, Richardson SK et al. Total skin electron
cutaneous T-cell lymphoma. Hematol Oncol Clin North Am 1995; beam therapy may be associated with improvement of peripheral
9:1057–76. blood disease in Sezary syndrome. J Am Acad Dermatol 2008;
117 Morris SL, McGovern M, Bayne S et al. Results of a 5-week sched- 58:592–5.
ule of modern total skin electron beam radiation therapy. Int J 134 Kamstrup MR, Gniadecki R, Iversen L et al. Low-dose (10-Gy)
Radiat Oncol Biol Phys 2013; 86:936–41. total skin electron beam therapy for cutaneous T-cell lymphoma:
118 Heumann TR, Esiashvili N, Parker S et al. Total skin electron ther- an open clinical study and pooled data analysis. Int J Radiat Oncol
apy for cutaneous T-cell lymphoma using a modern dual-field Biol Phys 2015; 92:138–43.
rotational technique. Int J Radiat Oncol Biol Phys 2015; 92:183–91. 135 Bunn PA, Ihde DC, Foon KA. The role of recombinant interferon
119 Navi D, Riaz N, Levin YS et al. The Stanford University experience alfa-2a in the therapy of cutaneous T-cell lymphomas. Cancer
with conventional-dose, total skin electron-beam therapy in the 1986; 57:1689–95.
treatment of generalized patch or plaque (T2) and tumor (T3) 136 Olsen EA, Rosen ST, Vollmer RT et al. Interferon alfa-2a in the
mycosis fungoides. Arch Dermatol 2011; 147:561–7. treatment of cutaneous T cell lymphoma. J Am Acad Dermatol 1989;
120 Lloyd S, Chen Z, Foss FM et al. Acute toxicity and risk of infec- 20:395–407.
tion during total skin electron beam therapy for mycosis fun- 137 Papa G, Tura S, Mandelli F et al. Is interferon alpha in cutaneous
goides. J Am Acad Dermatol 2013; 69:537–43. T-cell lymphoma a treatment of choice? Br J Haematol 1991; 79
121 Harrison C, Young J, Navi D et al. Revisiting low-dose total skin (Suppl. 1):S48–51.
electron beam therapy in mycosis fungoides. Int J Radiat Oncol Biol 138 Kohn EC, Steis RG, Sausville EA et al. Phase II trial of intermittent
Phys 2011; 81:e651–7. high-dose recombinant interferon alfa-2a in mycosis fungoides
122 Kamstrup MR, Lindahl LM, Gniadecki R et al. Low-dose total skin and the Sezary syndrome. J Clin Oncol 1990; 8:155–60.
electron beam therapy as a debulking agent for cutaneous T-cell 139 Dreno B, Claudy A, Meynadier J et al. The treatment of 45
lymphoma: an open-label prospective phase II study. Br J Dermatol patients with cutaneous T-cell lymphoma with low doses of
2012; 166:399–404. interferon-a2a and etretinate. Br J Dermatol 1991; 125:456–9.
123 Lindahl LM, Kamstrup MR, Petersen PM et al. Total skin electron 140 Aviles A, Neri N, Fernandez-Diez J et al. Interferon and low doses
beam therapy for cutaneous T-cell lymphoma: a nationwide of methotrexate versus interferon and retinoids in the treatment
cohort study from Denmark. Acta Oncol 2011; 50:1199–205. of refractory/relapsed cutaneous T-cell lymphoma. Hematology
124 Hoppe RT, Harrison C, Tavallaee M et al. Low-dose total skin 2015; 20:538–42.
electron beam therapy as an effective modality to reduce disease 141 Straus DJ, Duvic M, Kuzel T et al. Results of a phase II trial of oral
burden in patients with mycosis fungoides: results of a pooled bexarotene (Targretin) combined with interferon alfa-2b (Intron-
analysis from 3 phase-II clinical trials. J Am Acad Dermatol 2015; A) for patients with cutaneous T-cell lymphoma. Cancer 2007;
72:286–92. 109:1799–803.
125 Morris SL, Scarisbrick J, Frew JJ et al. The results of low dose total 142 Foo SH, Shah F, Chaganti S et al. Unmasking mycosis fungoides/
skin electron beam radiotherapy (TSEB), in patients with mycosis Sezary syndrome from preceding or co-existing benign inflam-
fungoides from the U.K. Cutaneous Lymphoma Group. Int J Radiat matory dermatoses requiring systemic therapies: patients fre-
Oncol Biol Phys 2017; 99:627–33. quently present with advanced disease and have an aggressive
126 Hamminga B, Noordijk EM, van Vloten WA. Treatment of myco- clinical course. Br J Dermatol 2016; 174:901–4.
sis fungoides: total-skin electron-beam irradiation versus topical 143 Molin L, Thomsen K, Volden G et al. Oral retinoids in mycosis
mechlorethamine therapy. Arch Dermatol 1982; 118:150–3. fungoides and Sezary syndrome: a comparison of isotretinoin and
127 Quiros PA, Jones GW, Kacinski BM et al. Total skin electron beam etretinate. A study from the Scandinavian Mycosis Fungoides
therapy followed by adjuvant psoralen/ultraviolet-A light in the Group. Acta Derm Venereol 1987; 67:232–6.
management of patients with T1 and T2 cutaneous T-cell lym- 144 Kessler JF, Jones SE, Levine N et al. Isotretinoin and cutaneous
phoma (mycosis fungoides). Int J Radiat Oncol Biol Phys 1997; helper T-cell lymphoma (mycosis fungoides). Arch Dermatol 1987;
38:1027–35. 123:201–4.
128 Chinn DM, Chow S, Kim YH et al. Total skin electron beam ther- 145 Duvic M, Martin AG, Kim Y et al. Phase 2 and 3 clinical trial of
apy with or without adjuvant topical nitrogen mustard or nitro- oral bexarotene (Targretin capsules) for the treatment of refrac-
gen mustard alone as initial treatment of T2 and T3 mycosis tory or persistent early-stage cutaneous T-cell lymphoma. Arch
fungoides. Int J Radiat Oncol Biol Phys 1999; 43:951–8. Dermatol 2001; 137:581–93.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


28 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

146 Talpur R, Ward S, Apisarnthanarax N et al. Optimizing bexarotene 164 Prince HM, Duvic M, Martin A et al. Phase III placebo-controlled
therapy for cutaneous T-cell lymphoma. J Am Acad Dermatol 2002; trial of denileukin diftitox for patients with cutaneous T-cell lym-
47:672–84. phoma. J Clin Oncol 2010; 28:1870–7.
147 Scarisbrick JJ, Morris S, Azurdia R et al. U.K. consensus statement 165 Olsen EA, Kim YH, Kuzel TM et al. Phase IIb multicenter trial of
on safe clinical prescribing of bexarotene for patients with cuta- vorinostat in patients with persistent, progressive, or treatment
neous T-cell lymphoma. Br J Dermatol 2013; 168:192–200. refractory cutaneous T-cell lymphoma. J Clin Oncol 2007;
148 Duvic M, Hymes K, Heald P et al. Bexarotene is effective and safe 25:3109–15.
for treatment of refractory advanced-stage cutaneous T-cell lym- 166 Whittaker SJ, Demierre MF, Kim EJ et al. Final results from a mul-
phoma: multinational phase II-III trial results. J Clin Oncol 2001; ticenter, international, pivotal study of romidepsin in refractory
19:2456–71. cutaneous T-cell lymphoma. J Clin Oncol 2010; 28:4485–91.
149 Abbott RA, Whittaker SJ, Morris SL et al. Bexarotene therapy for 167 Zinzani PL, Musuraca G, Tani M et al. Phase II trial of proteasome
mycosis fungoides and Sezary syndrome. Br J Dermatol 2009; inhibitor bortezomib in patients with relapsed or refractory cuta-
160:1299–307. neous T-cell lymphoma. J Clin Oncol 2007; 25:4293–7.
150 Lundin J, Osterborg A, Brittinger G et al. CAMPATH-1H mono- 168 Bunn PA, Hoffman SJ, Norris D et al. Systemic therapy of cuta-
clonal antibody in therapy for previously treated low-grade non- neous T-cell lymphomas (mycosis fungoides and the Sezary syn-
Hodgkin’s lymphomas: a phase II multicenter study. European drome). Ann Intern Med 1994; 121:592–602.
Study Group of CAMPATH-1H Treatment in Low-Grade Non- 169 Winkelmann RK, Diaz-Perez JL, Buechner SA. The treatment of
Hodgkin’s Lymphoma. J Clin Oncol 1998; 16: 3257–63. Sezary syndrome. J Am Acad Dermatol 1984; 10:1000–4.
151 Kennedy GA, Seymour JF, Wolf M et al. Treatment of patients 170 Zackheim HS, Kashani-Sabet M, Hwang ST. Low-dose methotrex-
with advanced mycosis fungoides and Sezary syndrome with ate to treat erythrodermic cutaneous T-cell lymphoma: results in
alemtuzumab. Eur J Haematol 2003; 71:250–6. twenty-nine patients. J Am Acad Dermatol 1996; 34:626–31.
152 Lundin J, Hagberg H, Repp R et al. Phase 2 study of alem- 171 Sarris AH, Phan A, Duvic M et al. Trimetrexate in relapsed T-cell
tuzumab (anti-CD52 monoclonal antibody) in patients with lymphoma with skin involvement. J Clin Oncol 2002; 20:2876–80.
advanced mycosis fungoides/Sezary syndrome. Blood 2003; 172 Cummings FJ, Kim K, Neiman RS et al. Phase II trial of pento-
101:4267–72. statin in refractory lymphomas and cutaneous T-cell disease. J Clin
153 Zinzani PL, Alinari L, Tani M et al. Preliminary observations of a Oncol 1991; 9:565–71.
phase II study of reduced-dose alemtuzumab treatment in 173 Greiner D, Olsen EA, Petroni G. Pentostatin (20 -deoxycoformy-
patients with pretreated T-cell lymphoma. Haematologica 2005; cin) in the treatment of cutaneous T-cell lymphoma. J Am Acad
90:702–3. Dermatol 1997; 36:950–5.
154 Bernengo MG, Quaglino P, Comessatti A et al. Low-dose intermit- 174 Kurzrock R, Pilat S, Duvic M. Pentostatin therapy of T-cell lym-
tent alemtuzumab in the treatment of Sezary syndrome: clinical phomas with cutaneous manifestations. J Clin Oncol 1999;
and immunologic findings in 14 patients. Haematologica 2007; 17:3117–21.
92:784–94. 175 Dearden C, Matutes E, Catovsky D. Pentostatin treatment of cuta-
155 Alinari L, Geskin L, Grady T et al. Subcutaneous alemtuzumab for neous T-cell lymphoma. Oncology (Williston Park) 2000; 14(6 Suppl.
Sezary syndrome in the very elderly. Leuk Res 2008; 32:1299– 2):37–40.
303. 176 Ho AD, Suciu S, Stryckmans P et al. Pentostatin in T-cell malig-
156 Querfeld C, Mehta N, Rosen ST et al. Alemtuzumab for relapsed nancies – a phase II trial of the EORTC. Leukemia Cooperative Group.
and refractory erythrodermic cutaneous T-cell lymphoma: a sin- Ann Oncol 1999; 10:1493–8.
gle institution experience from the Robert H. Lurie Comprehen- 177 Foss FM, Ihde DC, Breneman DL et al. Phase II study of pento-
sive Cancer Center. Leuk Lymphoma 2009; 50:1969–76. statin and intermittent high-dose recombinant interferon alfa-2a
157 de Masson A, Guitera P, Brice P et al. Long-term efficacy and in advanced mycosis fungoides/Sezary syndrome. J Clin Oncol
safety of alemtuzumab in advanced primary cutaneous T-cell 1992; 10:1907–13.
lymphomas. Br J Dermatol 2014; 170:720–4. 178 Kuzel TM, Hurria A, Samuelson E et al. Phase II trial of 2-chloro-
158 Lenihan DJ, Alencar AJ, Yang D et al. Cardiac toxicity of alem- deoxyadenosine for the treatment of cutaneous T-cell lymphoma.
tuzumab in patients with mycosis fungoides/Sezary syndrome. Blood 1996; 87:906–11.
Blood 2004; 104:655–8. 179 Quaglino P, Fierro MT, Rossotto GL et al. Treatment of advanced
159 Scarisbrick JJ. Brentuximab vedotin therapy for CD30-positive mycosis fungoides/Sezary syndrome with fludarabine and poten-
cutaneous T-cell lymphoma: a targeted approach to management. tial adjunctive benefit to subsequent extracorporeal pho-
Future Oncol 2017; 13:2405–11. tochemotherapy. Br J Dermatol 2004; 150:327–36.
160 Duvic M, Tetzlaff MT, Gangar P et al. Results of a phase II trial of 180 Zinzani PL, Baliva G, Magagnoli M et al. Gemcitabine treatment in
brentuximab vedotin for CD30+ cutaneous T-cell lymphoma and pretreated cutaneous T-cell lymphoma: experience in 44 patients.
lymphomatoid papulosis. J Clin Oncol 2015; 33:3759–65. J Clin Oncol 2000; 18:2603–6.
161 Kim YH, Tavallaee M, Sundram U et al. Phase II investigator- 181 Marchi E, Alinari L, Tani M et al. Gemcitabine as frontline treat-
initiated study of brentuximab vedotin in mycosis fungoides and ment for cutaneous T-cell lymphoma: phase II study of 32
Sezary syndrome with variable CD30 expression level: a multi- patients. Cancer 2005; 104:2437–41.
institution collaborative project. J Clin Oncol 2015; 33:3750–8. 182 Duvic M, Talpur R, Wen S et al. Phase II evaluation of gemc-
162 Prince HM, Kim YH, Horwitz SM et al. Brentuximab vedotin or itabine monotherapy for cutaneous T-cell lymphoma. Clin Lym-
physician’s choice in CD30-positive cutaneous T-cell lymphoma phoma Myeloma 2006; 7:51–8.
(ALCANZA): an international, open-label, randomised, phase 3, 183 Wollina U, Dummer R, Brockmeyer NH et al. Multicenter study
multicentre trial. Lancet 2017; 390:555–66. of pegylated liposomal doxorubicin in patients with cutaneous T-
163 Olsen E, Duvic M, Frankel A et al. Pivotal phase III trial of two cell lymphoma. Cancer 2003; 98:993–1001.
dose levels of denileukin diftitox for the treatment of cutaneous 184 Dummer R, Quaglino P, Becker JC et al. Prospective international
T-cell lymphoma. J Clin Oncol 2001; 19:376–88. multicenter phase II trial of intravenous pegylated liposomal

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 29

doxorubicin monochemotherapy in patients with stage IIB, IVA, consensus statement update from the UK Photopheresis Society.
or IVB advanced mycosis fungoides: final results from EORTC Br J Haematol 2017; 177:287–310.
21012. J Clin Oncol 2012; 30:4091–7. 203 Sanyal S, Child F, Alfred A et al. U.K. national audit of extracor-
185 Illidge T, Chan C, Counsell N et al. Phase II study of gemcitabine poreal photopheresis in CTCL. Br J Dermatol 2018; 178:589–70.
and bexarotene (GEMBEX) in the treatment of cutaneous T-cell 204 Gottlieb SL, Wolfe JT, Fox FE et al. Treatment of cutaneous T-
lymphoma. Br J Cancer 2013; 109:2566–73. cell lymphoma with extracorporeal photopheresis monotherapy
186 Fierro MT, Doveil GC, Quaglino P et al. Combination of etopo- and in combination with recombinant interferon alfa: a 10-
side, idarubicin, cyclophosphamide, vincristine, prednisone and year experience at a single institution. J Am Acad Dermatol 1996;
bleomycin (VICOP-B) in the treatment of advanced cutaneous T- 35:946–57.
cell lymphoma. Dermatology 1997; 194:268–72. 205 Bisaccia E, Gonzalez J, Palangio M et al. Extracorporeal pho-
187 Akpek G, Koh HK, Bogen S et al. Chemotherapy with etoposide, tochemotherapy alone or with adjuvant therapy in the treatment
vincristine, doxorubicin, bolus cyclophosphamide, and oral pred- of cutaneous T-cell lymphoma: a 9-year retrospective study at a
nisone in patients with refractory cutaneous T-cell lymphoma. single institution. J Am Acad Dermatol 2000; 43:263–71.
Cancer 1999; 86:1368–76. 206 Dippel E, Schrag H, Goerdt S et al. Extracorporeal photopheresis
188 Foss FM, Ihde DC, Linnoila IR et al. Phase II trial of fludarabine and interferon-a in advanced cutaneous T-cell lymphoma. Lancet
phosphate and interferon alfa-2a in advanced mycosis fungoides/ 1997; 350:32–3.
Sezary syndrome. J Clin Oncol 1994; 12:2051–9. 207 Vonderheid EC, Bigler RD, Greenberg AS et al. Extracorporeal
189 Horwitz SM, Kim YH, Foss F et al. Identification of an active, photopheresis and recombinant interferon alfa 2b in Sezary syn-
well-tolerated dose of pralatrexate in patients with relapsed drome. Use of dual marker labeling to monitor therapeutic
or refractory cutaneous T-cell lymphoma. Blood 2012; 119: response. Am J Clin Oncol 1994; 17:255–63.
4115–22. 208 Haley HR, Davis DA, Sams WM. Durable loss of a malignant T-
190 Zic JA, Stricklin GP, Greer JP et al. Long-term follow-up of cell clone in a stage IV cutaneous T-cell lymphoma patient treated
patients with cutaneous T-cell lymphoma treated with extracor- with high-dose interferon and photopheresis. J Am Acad Dermatol
poreal photochemotherapy. J Am Acad Dermatol 1996; 35:935–45. 1999; 41:880–3.
191 Talpur R, Demierre MF, Geskin L et al. Multicenter photopheresis 209 Yoo EK, Cassin M, Lessin SR et al. Complete molecular remission
intervention trial in early-stage mycosis fungoides. Clin Lymphoma during biologic response modifier therapy for Sezary syndrome is
Myeloma Leuk 2011; 11:219–27. associated with enhanced helper T type 1 cytokine production and
192 Miller JD, Kirkland EB, Domingo DS et al. Review of extracorpo- natural killer cell activity. J Am Acad Dermatol 2001; 45:208–16.
real photopheresis in early-stage (IA, IB, and IIA) cutaneous T- 210 Suchin KR, Cucchiara AJ, Gottleib SL et al. Treatment of cutaneous T-
cell lymphoma. Photodermatol Photoimmunol Photomed 2007; 23:163– cell lymphoma with combined immunomodulatory therapy: a 14-year
71. experience at a single institution. Arch Dermatol 2002; 138:1054–60.
193 Child FJ, Mitchell TJ, Whittaker SJ et al. A randomized cross-over 211 Duvic M, Apisarnthanarax N, Cohen DS et al. Analysis of long-
study to compare PUVA and extracorporeal photopheresis in the term outcomes of combined modality therapy for cutaneous T-
treatment of plaque stage (T2) mycosis fungoides. Clin Exp Derma- cell lymphoma. J Am Acad Dermatol 2003; 49:35–49.
tol 2004; 29:231–6. 212 Tsirigotis P, Pappa V, Papageorgiou S et al. Extracorporeal photo-
194 Knobler R, Jantschitsch C. Extracorporeal photochemoim- pheresis in combination with bexarotene in the treatment of
munotherapy in cutaneous T-cell lymphoma. Transfus Apher Sci mycosis fungoides and Sezary syndrome. Br J Dermatol 2007;
2003; 28:81–9. 156:1379–81.
195 Zic JA. The treatment of cutaneous T-cell lymphoma with photo- 213 Wollina U, Looks A, Meyer J et al. Treatment of stage II cuta-
pheresis. Dermatol Ther 2003; 16:337–46. neous T-cell lymphoma with interferon alfa-2a and extracorpo-
196 Scarisbrick JJ, Taylor P, Holtick U et al. U.K. consensus statement real photochemotherapy: a prospective controlled trial. J Am Acad
on the use of extracorporeal photopheresis for treatment of cuta- Dermatol 2001; 44:253–60.
neous T-cell lymphoma and chronic graft-versus-host disease. Br J 214 LeMaistre CF, Saleh MN, Kuzel TM et al. Phase I trial of a ligand
Dermatol 2008; 158:659–78. fusion-protein (DAB389IL-2) in lymphomas expressing the
197 Russell-Jones R. Extracorporeal photopheresis in cutaneous T-cell receptor for interleukin-2. Blood 1998; 91:399–405.
lymphoma. Inconsistent data underline the need for randomized 215 Foss F, Demierre MF, DiVenuti G. A phase-1 trial of bexarotene
studies. Br J Dermatol 2000; 142:16–21. and denileukin diftitox in patients with relapsed or refractory
198 Edelson R, Berger C, Gasparro F et al. Treatment of cutaneous T- cutaneous T-cell lymphoma. Blood 2005; 106:454–7.
cell lymphoma by extracorporeal photochemotherapy. Prelimi- 216 Duvic M, Talpur R, Ni X et al. Phase 2 trial of oral vorinostat
nary results. N Engl J Med 1987; 316:297–303. (suberoylanilide hydroxamic acid, SAHA) for refractory cuta-
199 Duvic M, Hester JP, Lemak NA. Photopheresis therapy for cuta- neous T-cell lymphoma (CTCL). Blood 2007; 109:31–9.
neous T-cell lymphoma. J Am Acad Dermatol 1996; 35:573–9. 217 Piekarz RL, Frye R, Turner M et al. Phase II multi-institutional
200 Heald P, Rook A, Perez M et al. Treatment of erythrodermic cuta- trial of the histone deacetylase inhibitor romidepsin as monother-
neous T-cell lymphoma with extracorporeal photochemotherapy. apy for patients with cutaneous T-cell lymphoma. J Clin Oncol
J Am Acad Dermatol 1992; 27:427–33. 2009; 27:5410–17.
201 McKenna KE, Whittaker S, Rhodes LE et al. Evidence-based prac- 218 Duvic M, Olsen EA, Breneman D et al. Evaluation of the long-
tice of photopheresis 1987–2001: a report of a workshop of the term tolerability and clinical benefit of vorinostat in patients with
British Photodermatology Group and the U.K. Skin Lymphoma advanced cutaneous T-cell lymphoma. Clin Lymphoma Myeloma
Group. Br J Dermatol 2006; 154:7–20. 2009; 9:412–16.
202 Alfred A, Taylor PC, Dignan F et al. The role of extracorporeal 219 O’Connor OA, Pro B, Pinter-Brown L et al. Pralatrexate in patients
photopheresis in the management of cutaneous T-cell lymphoma, with relapsed or refractory peripheral T-cell lymphoma: results
graft-versus-host disease and organ transplant rejection: a from the pivotal PROPEL study. J Clin Oncol 2011; 29:1182–9.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)


30 BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al.

220 Querfeld C, Rosen ST, Guitart J et al. Multicenter phase II trial of 237 Wu PA, Kim YH, Lavori PW et al. A meta-analysis of patients
temozolomide in mycosis fungoides/Sezary syndrome: correla- receiving allogeneic or autologous hematopoietic stem cell trans-
tion with O(6)-methylguanine-DNA methyltransferase and mis- plant in mycosis fungoides and Sezary syndrome. Biol Blood Marrow
match repair proteins. Clin Cancer Res 2011; 17:5748–54. Transplant 2009; 15:982–90.
221 Olavarria E, Child F, Woolford A et al. T-cell depletion and autol- 238 Specht L, Dabaja B, Illidge T et al. Modern radiation therapy for
ogous stem cell transplantation in the management of tumour primary cutaneous lymphomas: field and dose guidelines from
stage mycosis fungoides with peripheral blood involvement. Br J the International Lymphoma Radiation Oncology Group. Int J
Haematol 2001; 114:624–31. Radiat Oncol Biol Phys 2015; 92:32–9.
222 Ingen-Housz-Oro S, Bachelez H, Verola O et al. High-dose ther- 239 Laly P, Ingen-Housz-Oro S, Beylot-Barry M et al. Efficacy of vin-
apy and autologous stem cell transplantation in relapsing cuta- blastine in primary cutaneous anaplastic large cell lymphoma.
neous lymphoma. Bone Marrow Transplant 2004; 33:629–34. JAMA Dermatol 2015; 151:1030–1.
223 Bigler RD, Crilley P, Micaily B et al. Autologous bone marrow 240 Dearden CE, Johnson R, Pettengell R et al. Guidelines for the
transplantation for advanced stage mycosis fungoides. Bone Marrow management of mature T-cell and NK-cell neoplasms (excluding
Transplant 1991; 7:133–7. cutaneous T-cell lymphoma). Br J Haematol 2011; 153:451–85.
224 Burt RK, Guitart J, Traynor A et al. Allogeneic hematopoietic stem 241 Willemze R, Jansen PM, Cerroni L et al. Subcutaneous panniculi-
cell transplantation for advanced mycosis fungoides: evidence of a tis-like T-cell lymphoma: definition, classification, and prognostic
graft-versus-tumor effect. Bone Marrow Transplant 2000; 25:111–13. factors: an EORTC Cutaneous Lymphoma Group Study of 83
225 Duarte RF, Canals C, Onida F et al. Allogeneic hematopoietic cell cases. Blood 2008; 111:838–45.
transplantation for patients with mycosis fungoides and Sezary 242 Michonneau D, Petrella T, Ortonne N et al. Subcutaneous panni-
syndrome: a retrospective analysis of the Lymphoma Working culitis-like T-cell lymphoma: immunosuppressive drugs induce
Party of the European Group for Blood and Marrow Transplanta- better response than polychemotherapy. Acta Derm Venereol 2017;
tion. J Clin Oncol 2010; 28:4492–9. 97:358–64.
226 Koeppel MC, Stoppa AM, Resbeut M et al. Mycosis fungoides and 243 Go RS, Wester SM. Immunophenotypic and molecular features,
allogenic bone marrow transplantation. Acta Derm Venereol 1994; clinical outcomes, treatments, and prognostic factors associated
74:331–2. with subcutaneous panniculitis-like T-cell lymphoma: a system-
227 Masood N, Russell KJ, Olerud JE et al. Induction of complete atic analysis of 156 patients reported in the literature. Cancer
remission of advanced stage mycosis fungoides by allogeneic 2004; 101:1404–13.
hematopoietic stem cell transplantation. J Am Acad Dermatol 2002; 244 Rojnuckarin P, Nakorn TN, Assanasen T et al. Cyclosporin in sub-
47:140–5. cutaneous panniculitis-like T-cell lymphoma. Leuk Lymphoma 2007;
228 Guitart J, Wickless SC, Oyama Y et al. Long-term remission after 48:560–3.
allogeneic hematopoietic stem cell transplantation for refractory 245 James E, Sokhn JG, Gibson JF et al. CD4+ primary cutaneous
cutaneous T-cell lymphoma. Arch Dermatol 2002; 138:1359–65. small/medium-sized pleomorphic T-cell lymphoma: a retrospec-
229 Molina A, Zain J, Arber DA et al. Durable clinical, cytogenetic, tive case series and review of literature. Leuk Lymphoma 2015;
and molecular remissions after allogeneic hematopoietic cell 56:951–7.
transplantation for refractory Sezary syndrome and mycosis fun- 246 Virmani P, Jawed S, Myskowski PL et al. Long-term follow-up
goides. J Clin Oncol 2005; 23:6163–71. and management of small and medium-sized CD4+ T cell lym-
230 Herbert KE, Spencer A, Grigg A et al. Graft-versus-lymphoma phoma and CD8+ lymphoid proliferations of acral sites: a multi-
effect in refractory cutaneous T-cell lymphoma after reduced- center experience. Int J Dermatol 2016; 55:1248–54.
intensity HLA-matched sibling allogeneic stem cell transplanta- 247 Garcia-Herrera A, Colomo L, Camos M et al. Primary cutaneous
tion. Bone Marrow Transplant 2004; 34:521–5. small/medium CD4+ T-cell lymphomas: a heterogeneous group
231 Soligo D, Ibatici A, Berti E et al. Treatment of advanced mycosis of tumors with different clinicopathologic features and outcome.
fungoides by allogeneic stem-cell transplantation with a non- J Clin Oncol 2008; 26:3364–71.
myeloablative regimen. Bone Marrow Transplant 2003; 31:663–6. 248 Eich HT, Eich D, Micke O et al. Long-term efficacy, curative
232 Fijnheer R, Sanders CJ, Canninga MR et al. Complete remission of potential, and prognostic factors of radiotherapy in primary cuta-
a radiochemotherapy-resistant cutaneous T-cell lymphoma with neous B-cell lymphoma. Int J Radiat Oncol Biol Phys 2003; 55:899–
allogeneic non-myeloablative stem cell transplantation. Bone Mar- 906.
row Transplant 2003; 32:345–7. 249 Smith BD, Glusac EJ, McNiff JM et al. Primary cutaneous B-cell
233 Delioukina M, Zain J, Palmer JM et al. Reduced-intensity allo- lymphoma treated with radiotherapy: a comparison of the Euro-
geneic hematopoietic cell transplantation using fludarabine-mel- pean Organization for Research and Treatment of Cancer and the
phalan conditioning for treatment of mature T-cell lymphomas. WHO classification systems. J Clin Oncol 2004; 22:634–9.
Bone Marrow Transplant 2012; 47:65–72. 250 Senff NJ, Hoefnagel JJ, Neelis KJ et al. Results of radiotherapy
234 Paralkar VR, Nasta SD, Morrissey K et al. Allogeneic hematopoietic in 153 primary cutaneous B-cell lymphomas classified accord-
SCT for primary cutaneous T cell lymphomas. Bone Marrow Trans- ing to the WHO-EORTC classification. Arch Dermatol 2007;
plant 2012; 47:940–5. 143:1520–6.
235 Hosing C, Bassett R, Dabaja B et al. Allogeneic stem-cell transplan- 251 Piccinno R, Caccialanza M, Berti E et al. Radiotherapy of cuta-
tation in patients with cutaneous lymphoma: updated results neous B cell lymphomas: our experience in 31 cases. Int J Radiat
from a single institution. Ann Oncol 2015; 26:2490–5. Oncol Biol Phys 1993; 27:385–9.
236 Duarte RF, Boumendil A, Onida F et al. Long-term outcome of 252 Lowry L, Smith P, Qian W et al. Reduced dose radiotherapy for
allogeneic hematopoietic cell transplantation for patients with local control in non-Hodgkin lymphoma: a randomised phase III
mycosis fungoides and Sezary syndrome: a European Society for trial. Radiother Oncol 2011; 100:86–92.
Blood and Marrow Transplantation Lymphoma working party 253 Morris SL. Skin lymphoma. Clin Oncol (R Coll Radiol) 2012;
extended analysis. J Clin Oncol 2014; 32:3347–8. 24:371–85.

British Journal of Dermatology (2018) © 2018 British Association of Dermatologists


BAD and U.K. CLG guidelines for primary cutaneous lymphomas 2018, D. Gilson et al. 31

254 Hoskin P, Kirkwood A, Popova B et al. FoRT: a phase 3 multi-


center prospective randomized trial of low dose radiation therapy Supporting Information
for follicular and marginal zone lymphoma. Int J Radiat Oncol Biol
Additional Supporting Information may be found in the online
Phys 2013; 85:22.
255 Vandersee S, Terhorst D, Humme D et al. Treatment of indolent version of this article at the publisher’s website:
primary cutaneous B-cell lymphomas with subcutaneous inter- Appendix S1 Levels of evidence and strength of recom-
feron-alfa. J Am Acad Dermatol 2014; 70:709–15. mendation.
256 Pe~nate Y, Hernandez-Machın B, Perez-Mendez LI et al. Intrale- Appendix S2 Search strategy.
sional rituximab in the treatment of indolent primary cutaneous Appendix S3 List of abbreviations.
B-cell lymphomas: an epidemiological observational multicentre Table S1 Staging system for mycosis fungoides/Sezary syn-
study. The Spanish Working Group on Cutaneous Lymphoma. Br
drome.
J Dermatol 2012; 167:174–9.
257 Valencak J, Weihsengruber F, Rappersberger K et al. Rituximab Table S2 Staging for mycosis fungoides/Sezary syndrome
monotherapy for primary cutaneous B-cell lymphoma: adapted from Olsen et al.23
response and follow-up in 16 patients. Ann Oncol 2009; Table S3 The International Society for Cutaneous Lym-
20:326–30. phomas–European Organisation for Research and Treatment of
258 Brandenburg A, Humme D, Terhorst D et al. Long-term outcome of Cancer staging system for non-mycosis fungoides/non-Sezary
intravenous therapy with rituximab in patients with primary cuta- syndrome primary cutaneous lymphomas.24
neous B-cell lymphomas. Br J Dermatol 2013; 169:1126–32.
Table S4 Modified Severity Weighted Assessment Tool
259 Fenot M, Quereux G, Brocard A et al. Rituximab for primary
cutaneous diffuse large B-cell lymphoma-leg type. Eur J Dermatol (mSWAT) in mycosis fungoides/Sezary syndrome.
2010; 20:753–7. Table S5 Skin and global response in mycosis fungoides/
260 Wang and Bagot. Br J Dermatol 2019; 180:000–000. Sezary syndrome.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018)

You might also like