To the editor:

Primary cutaneous CD30+ lymphoproliferative disorders (CD30+ LPDs) are a disease spectrum including primary cutaneous anaplastic large cell lymphoma (C-ALCL) and lymphomatoid papulosis (LyP).1  C-ALCL presents as solitary, grouped, or, rarely, multifocal nodules and tumors that often ulcerate. Cutaneous relapses are common, but extracutaneous dissemination occurs in only 10% to 15% of patients and mainly involves regional lymph nodes.2-4  LyP is characterized by a chronic course of recurrent, self-healing papulonecrotic or nodular skin lesions. Extracutaneous disease rarely develops.3  The prognosis of both conditions is usually excellent with a 5-year disease-specific survival of approximately 90% for C-ALCL and almost 100% for LyP.2-4  However, about 10% of patients with primary cutaneous CD30+ LPD run a more aggressive clinical course. Apart from presentation with extensive skin lesions on legs or arms, risk factors for this unusually aggressive clinical course are currently unknown.2,3,5 

Primary systemic ALCL is divided into ALK+ ALCL and ALK ALCL, based on the presence or absence, respectively, of a rearrangement in the ALK gene that can reliably be detected by ALK immunohistochemistry.6  Systemic ALK+ ALCL has a better overall survival than systemic ALK ALCL, with 5-year survival rates of approximately 80% for ALK+ ALCL and approximately 50% for ALK ALCL.7,8 

In systemic ALK ALCL, recent studies detected 2 additional genes (DUSP22 and TP63) with recurrent chromosomal rearrangements that were shown to be mutually exclusive and absent in ALK+ ALCL. DUSP22 is rearranged in approximately 30% of the ALK ALCL patients and is associated with a more favorable prognosis with a 5-year survival rate of 90%.8,9  Chromosomal rearrangements involving the TP63 gene, found in 8% of patients with ALK ALCL, showed a poorer overall survival than ALK ALCL without a DUSP22 or TP63 rearrangement with 5-year survival rates of 17% and 42%, respectively.8-10 

In C-ALCL, ALK rearrangements are generally absent.11  However, unusual cases of ALK+ C-ALCL have been reported, many with an excellent prognosis.12-15  Rearrangements of DUSP22 have been detected in approximately 30% of C-ALCL patients and in a small subset of LyP.11,16,17  Clinical behavior and prognosis were similar to that of patients without DUSP22 rearrangements. TP63 rearrangements have only been detected in 2 of 41 patients with C-ALCL (5%), and both patients had an unusually aggressive disease course.10,18 TP63 rearrangements also have been detected in a single patient with transformed mycosis fungoides (MF) with an aggressive clinical course, but were not detected in 32 patients with LyP.18 

The association of TP63 rearrangements with poor outcome, both in patients with systemic ALK ALCL and 2 patients with C-ALCL, prompted us to study the presence of this rearrangement in a selected group of patients with C-ALCL and LyP with an aggressive clinical course.

From the 118 patients with C-ALCL included in the Cutaneous Lymphoma Registry of the Leiden University Medical Center (LUMC) between 1985 and 2015, those patients were particularly selected who had developed extracutaneous disease and/or died of ALCL (Table 1; patients 1-8 and 12) and patients who had presented with extensive skin lesions on the legs (patients 1-4, 8, 13, and 14). From the 185 patients with LyP registered in the same period, we selected the only 2 patients who died of systemic ALCL during follow-up (patients 15 and 16) and 1 patient who developed a large persistent tumor during follow-up (patient 17). The final study group contained 17 patients with CD30+ LPDs, including 14 patients with C-ALCL and 3 with LyP, type C. In all patients with C-ALCL, routine staging procedures, including physical examination, blood tests, computed tomography scans of neck, chest, and abdomen, and in most cases bone marrow biopsies, showed no signs of extracutaneous disease at the time of diagnosis.

Table 1

Clinical follow-up data of 17 patients with primary cutaneous CD30+ LPD and 1 patient with MF

No.SexAge (y)DiagnosisSite of relapse (mo after diagnosis)Current statusDuration of follow-up (mo)p63 expressionTP63 FISH
63 C-ALCL S, LN (29), muscle (29) DOL 31 * Normal 
59 C-ALCL S, LN (6) DOL 19 − Normal 
45 C-ALCL S, LN (18), lung (18), blood (19) DOL 20 ++ Normal 
37 C-ALCL S, LN (161), bone (167), lung (167), muscle (167) DOL 171 + Abnormal (43% polyploidy) 
62 C-ALCL S, LN (28), lung (32) DOL 36 ++ Normal 
88 C-ALCL DOL 10 − Normal 
47 C-ALCL LN (4) DOL ±§ Normal 
67 C-ALCL S, LN (12) DUD 34 Abnormal (12% polyploidy) 
71 C-ALCL — DUD 13 − Normal 
10 78 C-ALCL — DUD 34 ± Normal 
11 54 C-ALCL AWD 170 Abnormal (6% polyploidy) 
12 63 C-ALCL Bone (2) ACR 90 ++ Normal 
13 80 C-ALCL ACR 80 ± Normal 
14 44 C-ALCL ACR 141 − Normal 
15 43 LyP S, LN (48) DOL 51 ± Normal 
16 71 LyP S, LN (48), bone marrow (50) DOL 53 − Normal 
17 62 LyP DUD 158 − Normal 
PC 80 MF DUD 87 Abnormal (unbalanced translocation with gain of green signal) 
No.SexAge (y)DiagnosisSite of relapse (mo after diagnosis)Current statusDuration of follow-up (mo)p63 expressionTP63 FISH
63 C-ALCL S, LN (29), muscle (29) DOL 31 * Normal 
59 C-ALCL S, LN (6) DOL 19 − Normal 
45 C-ALCL S, LN (18), lung (18), blood (19) DOL 20 ++ Normal 
37 C-ALCL S, LN (161), bone (167), lung (167), muscle (167) DOL 171 + Abnormal (43% polyploidy) 
62 C-ALCL S, LN (28), lung (32) DOL 36 ++ Normal 
88 C-ALCL DOL 10 − Normal 
47 C-ALCL LN (4) DOL ±§ Normal 
67 C-ALCL S, LN (12) DUD 34 Abnormal (12% polyploidy) 
71 C-ALCL — DUD 13 − Normal 
10 78 C-ALCL — DUD 34 ± Normal 
11 54 C-ALCL AWD 170 Abnormal (6% polyploidy) 
12 63 C-ALCL Bone (2) ACR 90 ++ Normal 
13 80 C-ALCL ACR 80 ± Normal 
14 44 C-ALCL ACR 141 − Normal 
15 43 LyP S, LN (48) DOL 51 ± Normal 
16 71 LyP S, LN (48), bone marrow (50) DOL 53 − Normal 
17 62 LyP DUD 158 − Normal 
PC 80 MF DUD 87 Abnormal (unbalanced translocation with gain of green signal) 

ACR, alive and in complete remission; AWL, alive with disease; DOL, died of lymphoma; DUD, died of unrelated disease; F, female; M, male; LN, lymph node; PC, positive control; S, skin.

*

Tumor cells completely negative.

>75% of tumor cells positive.

30% to 75% of tumor cells positive.

§

<30% of tumor cells positive.

For the purpose of this study, p63 immunohistochemistry and fluorescence in situ hybridization (FISH) with break-apart probes for TP63 were performed on the formalin-fixed and paraffin-embedded (FFPE) skin biopsies of 16 patients and an involved lymph node of 1 patient (patient 15); these were collected from the archives of the LUMC Department of Pathology in accordance with the Dutch Code for Proper Secondary Use of Human Tissue and approved by the medical ethics committee of the LUMC. In all FFPE skin biopsies, CD30+ tumor cells made up at least 70% of the total infiltrate. As a positive control, the FFPE skin biopsy of 1 patient with tumor stage MF with a TP63 rearrangement (t(3,6)(q28p22.3)) detected by whole genome sequencing (unpublished data) was included. Immunohistochemistry was automatically performed using Dako Autostainer Link 48 with monoclonal mouse anti-human p63 antibodies from Dako (DAK-P63, M7317) in a dilution of 1:50. For positive staining, a cutoff value of 30% was used.9  For the FISH probes, DNA was isolated from bacterial artificial chromosome clones: RP11-718B1, RP11-179O12, and RP11-24F1 proximal and RP11-791J2, RP11-10K11, and RP11-204624 distal of the TP63 gene, purchased from the BACPAC resources center, Children’s Hospital Oakland Research Institute, California, and based on the probe design as used by Vasmatzis et al.10  The isolated DNA was labeled using nick translation either with digoxigenin- or biotin-coupled dideoxynucleotides as haptens. FFPE whole-tissue sections were deparaffinized in xylene, pretreated in 10 mM citric acid buffer, digested in 0.4% pepsin in 0.02M HCL, and codenatured and hybridized with the designed probes. The hapten-labeled probes were subsequently incubated with the mouse anti-digoxigenin antibody conjugated with fluorescein isothiocyanate (FITC) and streptavidin conjugated with Cy3 followed by a second detection using RabbitαMouse anti-biotin antibody conjugated with FITC, followed by counterstaining with 4,6 diamidino-2-phenylindole. In some cases, a probe for the centromere of chromosome 3 was added. For the analysis, 100 to 250 tumor cells per section were manually scored.

In all CD30+ LPD patients and the MF-positive control, both p63 immunohistochemistry and FISH for TP63 were successfully performed (Table 1; Figure 1). Expression of p63 in more than 30% of the tumor cells was present in 6/17 patients (35%), of which 3 showed expression in more than 75% of the tumor cells. In 7 patients, staining was completely negative. The MF-positive control expressed p63 in more than 30% of the neoplastic T cells. There was no relationship between p63 expression and the clinical course of the patients. A TP63 rearrangement was detected by FISH analysis in none of the 17 CD30+ LPD patients, suggesting that expression of p63 protein is transcriptionally regulated by the p63 promotor and/or influenced by epigenetic factors (eg, chromatin modifications, mitochondrial RNAs). In 3 patients, 1 to 3 extra fusion signals were seen in more than 5% of the scored cells: 6%, 12%, and 43%, respectively. In these cases, the probe for chromosome 3 also showed extra signals, indicating gain of chromosome 3 or polyploid tumor cells. It is unknown whether this has any implications on the regulation of the TP63 gene, though no clear association with p63 expression or with the clinical course of these patients was noted. The MF-positive control showed an unbalanced translocation with gain of the green signal (distal to TP63) (Figure 1).

Figure 1

Histologic features of 2 representative patients with primary C-ALCL and a patient with tumor stage MF included as a positive control. The dermis of the first patient with C-ALCL (patient 10) shows a dense dermal infiltrate of CD30+ tumor cells with infiltration of the hair shaft (A). The tumor cells are predominantly negative for p63 (B). FISH with break-apart probes for TP63 (TP63 FISH) shows 2 normal fusion signals (C). The second patient with C-ALCL (patient 4) also shows a dense dermal infiltrate of CD30+ tumor cells (D). The tumor cells variably express p63 (E). TP63 FISH shows 3 extra copies of the fusion signal, indicating gain of chromosome 3 or polyploid tumor cells (F). The hematoxylin and eosin staining of the patient with MF shows some epidermotropism and diffuse infiltration of the dermis by blastic tumor cells (G). The tumor cells variably express p63 (H). TP63 FISH shows 2 fusion signals and 2 separate green signals (I; arrows), suggesting gain of chromosome 3 with an unbalanced translocation of TP63. Note that the epidermis shows expression of p63 in the basal and suprabasal layers (B,E,H). Original magnification ×200 for panels A-B,G-H, ×100 for panels D-E, and ×400 for insets.

Figure 1

Histologic features of 2 representative patients with primary C-ALCL and a patient with tumor stage MF included as a positive control. The dermis of the first patient with C-ALCL (patient 10) shows a dense dermal infiltrate of CD30+ tumor cells with infiltration of the hair shaft (A). The tumor cells are predominantly negative for p63 (B). FISH with break-apart probes for TP63 (TP63 FISH) shows 2 normal fusion signals (C). The second patient with C-ALCL (patient 4) also shows a dense dermal infiltrate of CD30+ tumor cells (D). The tumor cells variably express p63 (E). TP63 FISH shows 3 extra copies of the fusion signal, indicating gain of chromosome 3 or polyploid tumor cells (F). The hematoxylin and eosin staining of the patient with MF shows some epidermotropism and diffuse infiltration of the dermis by blastic tumor cells (G). The tumor cells variably express p63 (H). TP63 FISH shows 2 fusion signals and 2 separate green signals (I; arrows), suggesting gain of chromosome 3 with an unbalanced translocation of TP63. Note that the epidermis shows expression of p63 in the basal and suprabasal layers (B,E,H). Original magnification ×200 for panels A-B,G-H, ×100 for panels D-E, and ×400 for insets.

Close modal

In conclusion, this study suggests that in patients with CD30+ LPD, an aggressive clinical course cannot be defined by the presence of TP63 rearrangements, as was recently shown in systemic ALK ALCL. In addition, immunohistochemical expression of p63 is variable in CD30+ LPD and is not correlated with overall survival.

Acknowledgments: The authors thank D. de Jong, Department of Molecular Cell Biology, Leiden University Medical Center, The Netherlands, for providing valuable assistance in performing FISH.

This work was supported by funding from the Dutch Cancer Society (grant UL2013-6104).

Contribution: A.M.R.S. performed the research, analyzed the data, and wrote the paper; Y.-Y.C. performed the research, contributed critical reagents, and analyzed the data; P.M.J. analyzed the data; A.N.B.T. contributed critical reagents; K.S. contributed analytical tools and analyzed the data; C.P.T. designed the research and contributed analytical tools; and R.W. designed the research, analyzed the data, and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interest.

Correspondence: Anne M. R. Schrader, Leiden University Medical Center, Department of Pathology, P.O. Box 9600, 2300 RC Leiden, The Netherlands; e-mail: a.m.r.schrader@gmail.com.

1
Willemze
 
R
Jaffe
 
ES
Burg
 
G
, et al. 
WHO-EORTC classification for cutaneous lymphomas.
Blood
2005
, vol. 
105
 
10
(pg. 
3768
-
3785
)
2
Benner
 
MF
Willemze
 
R
Applicability and prognostic value of the new TNM classification system in 135 patients with primary cutaneous anaplastic large cell lymphoma.
Arch Dermatol
2009
, vol. 
145
 
12
(pg. 
1399
-
1404
)
3
Bekkenk
 
MW
Geelen
 
FA
van Voorst Vader
 
PC
, et al. 
Primary and secondary cutaneous CD30(+) lymphoproliferative disorders: a report from the Dutch Cutaneous Lymphoma Group on the long-term follow-up data of 219 patients and guidelines for diagnosis and treatment.
Blood
2000
, vol. 
95
 
12
(pg. 
3653
-
3661
)
4
Liu
 
HL
Hoppe
 
RT
Kohler
 
S
Harvell
 
JD
Reddy
 
S
Kim
 
YH
CD30+ cutaneous lymphoproliferative disorders: the Stanford experience in lymphomatoid papulosis and primary cutaneous anaplastic large cell lymphoma.
J Am Acad Dermatol
2003
, vol. 
49
 
6
(pg. 
1049
-
1058
)
5
Woo
 
DK
Jones
 
CR
Vanoli-Storz
 
MN
, et al. 
Prognostic factors in primary cutaneous anaplastic large cell lymphoma: characterization of clinical subset with worse outcome.
Arch Dermatol
2009
, vol. 
145
 
6
(pg. 
667
-
674
)
6
Campo
 
E
Swerdlow
 
SH
Harris
 
NL
Pileri
 
S
Stein
 
H
Jaffe
 
ES
The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications.
Blood
2011
, vol. 
117
 
19
(pg. 
5019
-
5032
)
7
Savage
 
KJ
Harris
 
NL
Vose
 
JM
, et al. 
International Peripheral T-Cell Lymphoma Project
ALK− anaplastic large-cell lymphoma is clinically and immunophenotypically different from both ALK+ ALCL and peripheral T-cell lymphoma, not otherwise specified: report from the International Peripheral T-Cell Lymphoma Project.
Blood
2008
, vol. 
111
 
12
(pg. 
5496
-
5504
)
8
Parrilla Castellar
 
ER
Jaffe
 
ES
Said
 
JW
, et al. 
ALK-negative anaplastic large cell lymphoma is a genetically heterogeneous disease with widely disparate clinical outcomes.
Blood
2014
, vol. 
124
 
9
(pg. 
1473
-
1480
)
9
King
 
RL
Dao
 
LN
McPhail
 
ED
, et al. 
Morphologic features of ALK-negative anaplastic large cell lymphomas with DUSP22 rearrangements.
Am J Surg Pathol
2016
, vol. 
40
 
1
(pg. 
36
-
43
)
10
Vasmatzis
 
G
Johnson
 
SH
Knudson
 
RA
, et al. 
Genome-wide analysis reveals recurrent structural abnormalities of TP63 and other p53-related genes in peripheral T-cell lymphomas.
Blood
2012
, vol. 
120
 
11
(pg. 
2280
-
2289
)
11
Xing
 
X
Feldman
 
AL
Anaplastic large cell lymphomas: ALK positive, ALK negative, and primary cutaneous.
Adv Anat Pathol
2015
, vol. 
22
 
1
(pg. 
29
-
49
)
12
Kadin
 
ME
Pinkus
 
JL
Pinkus
 
GS
, et al. 
Primary cutaneous ALCL with phosphorylated/activated cytoplasmic ALK and novel phenotype: EMA/MUC1+, cutaneous lymphocyte antigen negative.
Am J Surg Pathol
2008
, vol. 
32
 
9
(pg. 
1421
-
1426
)
13
Oschlies
 
I
Lisfeld
 
J
Lamant
 
L
, et al. 
ALK-positive anaplastic large cell lymphoma limited to the skin: clinical, histopathological and molecular analysis of 6 pediatric cases. A report from the ALCL99 study.
Haematologica
2013
, vol. 
98
 
1
(pg. 
50
-
56
)
14
Quintanilla-Martinez
 
L
Jansen
 
PM
Kinney
 
MC
Swerdlow
 
SH
Willemze
 
R
Non-mycosis fungoides cutaneous T-cell lymphomas: report of the 2011 Society for Hematopathology/European Association for Haematopathology workshop.
Am J Clin Pathol
2013
, vol. 
139
 
4
(pg. 
491
-
514
)
15
Attygalle
 
AD
Cabeçadas
 
J
Gaulard
 
P
, et al. 
Peripheral T-cell and NK-cell lymphomas and their mimics; taking a step forward - report on the lymphoma workshop of the XVIth meeting of the European Association for Haematopathology and the Society for Hematopathology.
Histopathology
2014
, vol. 
64
 
2
(pg. 
171
-
199
)
16
Wada
 
DA
Law
 
ME
Hsi
 
ED
, et al. 
Specificity of IRF4 translocations for primary cutaneous anaplastic large cell lymphoma: a multicenter study of 204 skin biopsies.
Mod Pathol
2011
, vol. 
24
 
4
(pg. 
596
-
605
)
17
Pham-Ledard
 
A
Prochazkova-Carlotti
 
M
Laharanne
 
E
, et al. 
IRF4 gene rearrangements define a subgroup of CD30-positive cutaneous T-cell lymphoma: a study of 54 cases.
J Invest Dermatol
2010
, vol. 
130
 
3
(pg. 
816
-
825
)
18
Chavan
 
RN
Bridges
 
AG
Knudson
 
RA
, et al. 
Somatic rearrangement of the TP63 gene preceding development of mycosis fungoides with aggressive clinical course.
Blood Cancer J
2014
, vol. 
4
 pg. 
e253
 
Sign in via your Institution