Anaplastic large cell lymphoma (ALCL) is a distinct entity of non-Hodgkin lymphoma, characterized by a proliferation of pleomorphic large lymphoid cells that express CD30. Recent studies have found that a subset of ALCL aberrantly expresses a chimeric anaplastic lymphoma kinase (ALK) protein as a result of t(2;5)(p23;q35) or variant translocations. ALK-positive ALCLs feature good prognosis, but some of them lead to poor outcomes. Since CD56 is expressed in some ALCLs, its clinical significance was examined in a series of T/null cell type ALCLs. Of 143 patients, 83 (58%) showed ALK-positive staining, and of 140 patients, 25 (18%) expressed CD56. The ALK-positive subgroup was characterized by a younger age of onset (P < .0001), lower serum lactate dehydrogenase level (P = .01), better performance status (P = .03), less frequent extranodal involvement (P = .01), lower international prognostic index (IPI) categories (P = .002), and superior survival (P = .0009) in comparison with the ALK-negative group, suggesting that ALK is a specific marker defining a distinct subtype. CD56+ cases showed a significantly poor prognosis overall (P = .002) as well as in both ALK-positive and ALK-negative subgroups (P = .02 andP = .04, respectively). Multivariate analysis confirmed that CD56 is independent of other prognostic factors, including IPI. Although CD56+ cases showed a higher incidence of bone involvement, no other differences in clinicopathologic parameters were found between the CD56+ and CD56 groups. These findings suggest that CD56 is not a marker to identify a distinct subtype of ALCL, but a strong clinical prognostic factor. Effective therapeutic approaches should be explored for high-risk ALCL patients, who can be identified by means of a prognostic model, including CD56.

Anaplastic large cell lymphoma (ALCL) was first described by Stein et al1 as a large-cell non-Hodgkin lymphoma (NHL) characterized by a bizarre morphology that often shows intrasinusoidal and paracortical infiltration of lymph nodes. The tumor cells of ALCL express CD30 antigen, which is also expressed on Reed-Sternberg cells in Hodgkin disease (HD) and on a subset of various T-cell neoplasms.1-3 Both the B- and T/null cell type ALCLs were initially recognized in the updated Kiel Classification,4,5 but only T/null cell type ALCL has been included in the Revised European American Lymphoma (REAL) classification6 and the World Health Organization (WHO) classification.7 Although some morphological variants were proposed afterward, ALCL has been recognized as a distinct disease entity.

A nonrandom chromosomal translocation t(2;5)(p23;q35) has been reported in ALCL.8-10 This translocation has been cloned and shown to result in the fusion of the NPM gene on chromosome 5 and the ALK gene on chromosome 2, resulting in the expression of an aberrant fusion protein, p80NPM/ALK.11,12The polyclonal antibody against p80NPM/ALK, which recognizes anaplastic lymphoma kinase (ALK),13 and the subsequently established monoclonal antibodies ALK114 and ALKc15 have made it possible to further categorize ALCL as an entity separate from HD, lymphomatoid papulosis, and primary cutaneous ALCL.16-23 Accumulated evidence, such as immunohistochemical, cytogenetic, and reverse genetic detection, also supports the recognition of ALK-positive ALCL as a distinct subtype with a much younger age distribution, nodal predilection, and good prognosis.14,15,18-21,24-32 However, these issues are as yet only marginally dealt with within the REAL/WHO classifications because they have not been sufficiently confirmed by data from large series of ALCL cases.

A further issue is the expression of CD56, a neural cell-adhesion molecule, which is expressed on natural killer (NK) cells and a subset of T cells and monocytes.33,34 Its expression is well recognized in hematolymphoid malignancies of NK-cell lineage,35,36 but also in some cases of acute myeloid leukemia (AML). CD56 expression has been found to be a risk factor for AMLs with t(8;21) and t(15;17),37,38 but its significance in malignant lymphomas other than those of NK-cell lineage awaits further clarification. For this study, we investigated 143 cases of T/null cell type ALCL to determine the biologic and prognostic significance of p80/ALK and CD56 for the category of ALCL.

Patient selection

From the patient records of Aichi Cancer Center and collaborating institutions, 143 patients with ALCL of T/null cell phenotype were identified. These included 42 patients of cytotoxic-molecule–positive ALCL previously reported by us.39 All specimens were obtained at the initial presentation of the patients and were reviewed by 2 independent pathologists (T.S. and S.N.). Patients meeting the original criteria of Stein et al,1 supplemented by the description of Suchi et al,5 were enrolled in this study. Excluded were those occasional patients whose primary diagnostic material was not optimal for the identification of features relevant to this series, including minute biopsy specimens, tissues with extensive necrosis, and tissue materials used for rapid frozen-section diagnosis. Patients with ALCL of B-cell phenotype, primary cutaneous ALCL, and secondary ALCL were also excluded from this study, as were patients with retrovirus (human T-cell leukemia/lymphoma virus type 1 and human immunodeficiency virus) infection. The patients' records and clinical data were investigated retrospectively.

Histopathology

Tissue was fixed in 10% formalin and embedded in paraffin. Sections (5 μm thick) were stained with hematoxylin and eosin, periodic acid–Schiff, Giemsa, and Gomori silver impregnation.

Immunophenotypic study

Immunoperoxidase studies for the following antigens were performed on the formalin-fixed, paraffin-embedded sections by means of the avidin-biotin peroxidase complex method.40 The antibodies comprised Ber-H2/CD30 (Dako, Santa Fe, CA), LeuM1/CD15 (Becton Dickinson, Sunnyvale, CA), L26/CD20 (Dako), CD79a (Dako), UCHL1/CD45RO (Dako), MT1/CD43 (Bio-Science Products, Emmenbrucke, Switzerland), CD3 (Dako), CD4 (Novocastra Laboratories, Newcastle, UK), CD8 (Dako), E29/EMA (Coulter, Hialeah, FL), CD56 (Novocastra), Leu7/CD57 (Becton Dickinson), LMP-1 (Dako), DO-7/p53 (Dako), BCL-2 (Dako), TIA-1 (Coulter), granzyme B (Monosan, Uden, The Netherlands), βF1 (T Cell Science, Cambridge, MA), ALK1 (Dako), and p80 (courtesy of S. Mori, University of Tokyo, Japan). Detection of Epstein-Barr virus (EBV) small RNAs by means of in situ hybridization using EBV-encoded small RNA (EBER) oligonucleotides was also performed on formalin-fixed paraffin-embedded sections by means of the Dako hybridization kit with a cocktail of fluorescein-isothiocyanate–labeled EBER oligonucleotides (one oligonucleotide corresponding to EBER-1 and one to EBER-2, both 30 bases long).41 The cell lineage of each case was identified as previously described.28 Briefly, cases were classified as T lineage if they reacted with one or more antibodies against the T-cell antigens CD45RO, CD43, and CD3 and lacked reactivity for the B-cell–associated antigens CD20 and CD79a. They were classified as B-lineage if the opposite pattern of reactivity was observed. A null phenotype was assigned to cases that did not express either T- or B-cell–associated markers.

Statistical analysis

Correlation between the 2 groups was examined with the χ2 test, the Fisher exact test, the Student ttest, and the Mann-Whitney U test. Patient survival data were analyzed with the Kaplan-Meier method and were compared by means of the log-rank test. Univariate and multivariate analyses were performed with the Cox proportional hazard regression model, and variables were selected with the stepwise method. Data were analyzed with the SAS system (SAS Institute Inc, Cary, NC).

ALK immunohistochemistry and histopathological features

With the use of p80 and/or ALK1 antibodies, 83 of the 143 patients (58%) were shown to have ALK-positive ALCL cases, 1 of which showed nuclear-restricted ALK staining and 49 of which showed nuclear-positive cytoplasmic staining (ALK-N/NC), suggesting that the tumor cells of these cases harbored the NPM-ALK chimeric protein.42Another 25 cases displayed cytoplasmic-restricted staining (ALK-C), which indicated that the ALK gene may remain intact or may fuse with genes other than NPM. The staining pattern (ALK-N/NC vs ALK-C) could not be determined for the remaining 8 cases, mainly owing to the unsuitable condition of the paraffin blocks.

Histologically, 128 patients were categorized as classical type ALCL, 11 as HD-like ALCL, and 4 as lymphohistiocytic (LH)/small-cell (SC) variants. All of the 4 LH/SC variants showed ALK expression, but only 1 of the 11 HD-like ALCL types did. The diagnosis of HD-like ALCL was based on the histological appearance. All of these 11 cases showed sinusoidal involvements and a cohesive growth pattern of neoplastic cells, which led to a diagnosis of NHL rather than HD. These cases also had occasional Hodgkin/Reed-Sternberg–like cells, the absence of which would result in a diagnosis of classical or common type ALCL.

Clinical features

There were 97 males and 44 females with an age range from 1 to 85 years (median age, 32 years). Patients' clinical characteristics and subgroups according to ALK expression are summarized in Table1. The ALK-positive group showed a dramatically younger age distribution (mean: 25.0 ± 17.6 vs 50.6 ± 20.6 years). ALK-negative cases were male predominant, although the difference was not statistically significant. No differences in stage or B symptoms between ALK-positive and ALK-negative subgroups were observed. In ALK-positive cases, the performance status (PS) showed significantly better distribution (P = .03), and the serum lactate dehydrogenase (LDH) level was lower (P = .01). Most of the patients in both groups showed nodal presentation of the lymphoma, but the incidence of extranodal involvement was significantly higher in the ALK-negative group (P = .01). The incidence of BM or skin involvement tended to be higher in the ALK-negative group, and that of bone disease higher in the ALK-positive group, although the difference was not significant. The incidence of extranodal involvement at 2 or more sites did not show any difference. The international prognostic index (IPI) categories of the ALK-positive group showed lower distribution than those of the ALK-negative group (P = .002).

Table 1.

Patient characteristics according to ALK expression

CharacteristicsALK+ ALCLALKALCLP value
Total number 83 60  
Age (y), median (range) 21 (1-73) 57 (8-85) < .0001 
 ≦ 10 19 (23%) 1 (2%)  
 ≦ 30 54 (65%) 12 (20%)  
 > 60 4 (5%) 23 (38%)  
Sex (male/female) 52/31 45/15 .12 
Stage   .33 
 I 10 (12%) 5 (8%)  
 II 17 (21%) 14 (23%)  
 III 25 (30%) 12 (20%)  
 IV 31 (37%) 29 (48%)  
PS   .03 
 0 43 (53%) 22 (37%)  
 1 22 (27%) 17 (29%)  
 2 7 (9%) 6 (10%)  
 3 5 (6%) 7 (12%)  
 4 4 (5%) 7 (12%)  
LDH > normal 34 (42%) 36 (63%) .01  
B symptoms 45 (56%) 33 (57%) .88  
Extranodal involvement    
 Bone marrow 9 (11%) 12 (20%) .13 
 Skin 17 (21%) 19 (32%) .13 
 Liver 7 (8%) 5 (8%) .98 
 Spleen 9 (11%) 9 (15%) .46 
 Bone 10 (12%) 3 (5%) .15 
 Lung 10 (12%) 5 (8%) .70 
 Mediastinum 5 (6%) 7 (12%) .23  
 ≧ 1 site 44 (53%) 44 (73%) .01  
 ≧ 2 sites 22 (27%) 14 (24%) .68  
IPI   .002 
 Low 40 (50%) 16 (28%)  
 Low-intermediate 22 (28%) 18 (31%)  
 High-intermediate 13 (16%) 12 (21%)  
 High 5 (6%) 12 (21%)  
CharacteristicsALK+ ALCLALKALCLP value
Total number 83 60  
Age (y), median (range) 21 (1-73) 57 (8-85) < .0001 
 ≦ 10 19 (23%) 1 (2%)  
 ≦ 30 54 (65%) 12 (20%)  
 > 60 4 (5%) 23 (38%)  
Sex (male/female) 52/31 45/15 .12 
Stage   .33 
 I 10 (12%) 5 (8%)  
 II 17 (21%) 14 (23%)  
 III 25 (30%) 12 (20%)  
 IV 31 (37%) 29 (48%)  
PS   .03 
 0 43 (53%) 22 (37%)  
 1 22 (27%) 17 (29%)  
 2 7 (9%) 6 (10%)  
 3 5 (6%) 7 (12%)  
 4 4 (5%) 7 (12%)  
LDH > normal 34 (42%) 36 (63%) .01  
B symptoms 45 (56%) 33 (57%) .88  
Extranodal involvement    
 Bone marrow 9 (11%) 12 (20%) .13 
 Skin 17 (21%) 19 (32%) .13 
 Liver 7 (8%) 5 (8%) .98 
 Spleen 9 (11%) 9 (15%) .46 
 Bone 10 (12%) 3 (5%) .15 
 Lung 10 (12%) 5 (8%) .70 
 Mediastinum 5 (6%) 7 (12%) .23  
 ≧ 1 site 44 (53%) 44 (73%) .01  
 ≧ 2 sites 22 (27%) 14 (24%) .68  
IPI   .002 
 Low 40 (50%) 16 (28%)  
 Low-intermediate 22 (28%) 18 (31%)  
 High-intermediate 13 (16%) 12 (21%)  
 High 5 (6%) 12 (21%)  

PS indicates performance status; IPI, international prognostic index.

Expression of phenotypic markers and cytotoxic molecules

The results are summarized in Table2 and categorized according to ALK-positive and ALK-negative subgroups. Immunohistochemical profile of a CD56-positive case is shown in Figure1. All cases but 1 were positive for CD30, and CD56 was positive in 13 of 81 cases (18%) of the ALK-positive group and in 12 of 59 cases (20%) of the ALK-negative group, so that the incidence of expression was almost the same. None of the ALK-positive group showed any expression of CD15, BCL-2, or EBV, but most of them were positive for epithelial membrane antigen (EMA). On the other hand, the expression of these markers was somewhat heterogeneous for the ALK-negative group, resulting in a statistically significant difference between these 2 groups. In HD-like ALCL, the expression of CD15 was found in 4 out of 10, the expression of EMA in 7 out of 9, and the presence of EBV in 2 out of 10 cases. Of the 4 CD15+ cases, 3 were also positive for EMA; the remaining case was not examined for EMA. Only one case showed simultaneous expression of CD15 and EBV, but the neoplastic cells were also positive for EMA and CD45RO. The expression of cytotoxic molecules (TIA-1 and granzyme B) was significantly higher in the ALK-positive group (P < .0001 and P = .007, respectively).

Table 2.

Expression of phenotypic markers and cytotoxic molecules

ALK+ALCL (n = 83)ALK ALCL (n = 60)P value
CD56 13/81 (16%) 12/59 (20%) .51 
CD30 81/83 (98%) 60/60 (100%) .34 
CD15 0/78 11/55 (20%) < .0001 
CD20 0/83 0/60 1.00 
CD45RO 29/76 (38%) 21/48 (44%) .54 
CD43 34/68 (50%) 24/46 (52%) .82 
CD3 31/74 (42%) 26/50 (52%) .27 
CD4 34/74 (46%) 18/46 (39%) .46 
CD8 10/73 (14%) 6/47 (13%) .88 
EMA 80/83 (96%) 26/55 (47%) < .0001 
BCL2 0/45 14/27 (52%) < .0001 
TIA-1 59/75 (79%) 23/54 (43%) < .0001  
Granzyme B 53/81 (65%) 24/57 (42%) .007 
EBERs 0/80 12/58 (21%) < .0001 
ALK+ALCL (n = 83)ALK ALCL (n = 60)P value
CD56 13/81 (16%) 12/59 (20%) .51 
CD30 81/83 (98%) 60/60 (100%) .34 
CD15 0/78 11/55 (20%) < .0001 
CD20 0/83 0/60 1.00 
CD45RO 29/76 (38%) 21/48 (44%) .54 
CD43 34/68 (50%) 24/46 (52%) .82 
CD3 31/74 (42%) 26/50 (52%) .27 
CD4 34/74 (46%) 18/46 (39%) .46 
CD8 10/73 (14%) 6/47 (13%) .88 
EMA 80/83 (96%) 26/55 (47%) < .0001 
BCL2 0/45 14/27 (52%) < .0001 
TIA-1 59/75 (79%) 23/54 (43%) < .0001  
Granzyme B 53/81 (65%) 24/57 (42%) .007 
EBERs 0/80 12/58 (21%) < .0001 

EBER indicates Epstein-Barr–encoded small RNA; EMA, epithelial membrane antigen.

Fig. 1.

Immunohistochemistry of CD56 in ALCL.

CD56 is expressed on the cell surface membrane of the lymphoma cells, and its expression is more intense on the adjacent membrane of neighboring cells.

Fig. 1.

Immunohistochemistry of CD56 in ALCL.

CD56 is expressed on the cell surface membrane of the lymphoma cells, and its expression is more intense on the adjacent membrane of neighboring cells.

Close modal

Therapeutic response and prognosis

The treatment consisted of chemotherapeutic regimens containing doxorubicin for 125 patients and without doxorubicin for 9. Five patients with stage I disease did not receive chemotherapy and were treated with radiation or operative resection alone; the 3 patients who had not received any therapy because of their poor PS died of the disease; and 1 patient was lost to follow-up before receiving any therapy. In total, 100 of the 139 patients (71.9%) attained complete remission, and 18 (12.9%) partial remission. Therapeutic response was significantly better for the ALK-positive group (P = .009, Mann-Whitney U test).

The overall survival curves of ALK-positive and ALK-negative ALCLs, shown in Figure 2A, demonstrate a significantly better survival for ALK-positive ALCLs (P = .0009). The ALK-negative group showed no differences in survival between HD-like and common ALCL. The ALK-N/NC and ALK-C groups showed almost identical survival (Figure 2B).

Fig. 2.

Overall survival of ALK-positive and ALK-negative ALCL.

(A) ALK-positive ALCL shows significantly better prognosis (P = .0009). (B) No difference is seen in the pattern of ALK positivity (nuclear/nuclear + cytoplasmic vs cytoplasmic) (P = .61).

Fig. 2.

Overall survival of ALK-positive and ALK-negative ALCL.

(A) ALK-positive ALCL shows significantly better prognosis (P = .0009). (B) No difference is seen in the pattern of ALK positivity (nuclear/nuclear + cytoplasmic vs cytoplasmic) (P = .61).

Close modal

Comparison of CD56+ and CD56 cases

A comparison of the clinical characteristics of CD56+and CD56 cases is summarized in Table3. Although CD56+ cases showed a significant preponderance of bone disease (P = .005), none of the other clinical factors or the expression of phenotypic markers and cytotoxic molecules registered any significant difference between the CD56+ and CD56 groups (Table 4). However, the overall survival was significantly different, with the CD56+ cases showing a much poorer prognosis (Figure3, P = .002). In both the ALK-positive and ALK-negative subgroups, CD56+ cases showed a poorer prognosis than CD56 cases (Figure4A-B).

Table 3.

Clinical characteristics according to CD56 expression

CharacteristicsCD56+ ALCLCD56ALCLP value
Total 25 115  
Age (y), median (range) 39 (4-85) 29 (1-85) .14  
Sex (male/female) 17/8 78/37 .99 
Stage   .31 
 I 6 (24%) 9 (8%)  
 II 4 (16%) 25 (22%)  
 III 5 (20%) 31 (27%)  
 IV 10 (40%) 50 (44%)  
PS   .99 
 0 12 (48%) 51 (46%)  
 1 5 (20%) 33 (30%)  
 2 4 (16%) 9 (8%)  
 3 3 (12%) 9 (8%)  
 4 1 (4%) 10 (9%)  
LDH > normal 11 (44%) 56 (51%) .56  
B symptoms 14 (58%) 63 (56%) .82  
Extranodal involvement    
 BM 2 (8%) 19 (17%) .28 
 Skin 3 (12%) 33 (29%) .19 
 Liver 2 (8%) 10 (9%) .91 
 Spleen 2 (8%) 16 (14%) .43 
 Bone 6 (24%) 7 (6%) .005 
 Lung 5 (20%) 11 (10%) .13 
 Mediastinum 2 (8%) 9 (8%) .97  
 ≧ 1 site 16 (64%) 72 (62%) .90  
 ≧ 2 sites 8 (32%) 28 (25%) .46  
IPI   .63 
 Low 11 (44%) 44 (40%)  
 Low-intermediate 3 (12%) 36 (32%)  
 High-intermediate 7 (28%) 18 (16%)  
 High 4 (16%) 13 (12%)  
CharacteristicsCD56+ ALCLCD56ALCLP value
Total 25 115  
Age (y), median (range) 39 (4-85) 29 (1-85) .14  
Sex (male/female) 17/8 78/37 .99 
Stage   .31 
 I 6 (24%) 9 (8%)  
 II 4 (16%) 25 (22%)  
 III 5 (20%) 31 (27%)  
 IV 10 (40%) 50 (44%)  
PS   .99 
 0 12 (48%) 51 (46%)  
 1 5 (20%) 33 (30%)  
 2 4 (16%) 9 (8%)  
 3 3 (12%) 9 (8%)  
 4 1 (4%) 10 (9%)  
LDH > normal 11 (44%) 56 (51%) .56  
B symptoms 14 (58%) 63 (56%) .82  
Extranodal involvement    
 BM 2 (8%) 19 (17%) .28 
 Skin 3 (12%) 33 (29%) .19 
 Liver 2 (8%) 10 (9%) .91 
 Spleen 2 (8%) 16 (14%) .43 
 Bone 6 (24%) 7 (6%) .005 
 Lung 5 (20%) 11 (10%) .13 
 Mediastinum 2 (8%) 9 (8%) .97  
 ≧ 1 site 16 (64%) 72 (62%) .90  
 ≧ 2 sites 8 (32%) 28 (25%) .46  
IPI   .63 
 Low 11 (44%) 44 (40%)  
 Low-intermediate 3 (12%) 36 (32%)  
 High-intermediate 7 (28%) 18 (16%)  
 High 4 (16%) 13 (12%)  

See Table 1 footnote for explanation of abbreviations.

Table 4.

Phenotypic characteristics according to CD56 expression

CD56+ ALCL (n = 25)CD56 ALCL (n = 115)P value
ALK 13/25 (52%) 68/115 (59%) .51 
CD30 25/25 (100%) 113/115 (98%) .67 
CD15 1/22 (5%) 8/108 (8%) .52 
CD20 0/25 0/115 1.00 
CD45RO 9/18 (50%) 40/103 (39%) .37 
CD43 8/17 (47%) 49/94 (52%) .70 
CD3 11/23 (48%) 46/101 (46%) .84 
CD4 12/22 (55%) 40/98 (41%) .24 
CD8 3/22 (14%) 13/98 (13%) .67 
EMA 19/23 (83%) 85/112 (76%) .49 
BCL2 1/8 (13%) 13/64 (20%) .51 
TIA-1 11/22 (50%) 71/107 (66%) .15  
Granzyme B 15/25 (60%) 62/113 (55%) .64 
EBERs 4/24 (17%) 8/111 (7%) .14 
CD56+ ALCL (n = 25)CD56 ALCL (n = 115)P value
ALK 13/25 (52%) 68/115 (59%) .51 
CD30 25/25 (100%) 113/115 (98%) .67 
CD15 1/22 (5%) 8/108 (8%) .52 
CD20 0/25 0/115 1.00 
CD45RO 9/18 (50%) 40/103 (39%) .37 
CD43 8/17 (47%) 49/94 (52%) .70 
CD3 11/23 (48%) 46/101 (46%) .84 
CD4 12/22 (55%) 40/98 (41%) .24 
CD8 3/22 (14%) 13/98 (13%) .67 
EMA 19/23 (83%) 85/112 (76%) .49 
BCL2 1/8 (13%) 13/64 (20%) .51 
TIA-1 11/22 (50%) 71/107 (66%) .15  
Granzyme B 15/25 (60%) 62/113 (55%) .64 
EBERs 4/24 (17%) 8/111 (7%) .14 

See Table 2 for explanation of abbreviations.

Fig. 3.

Overall survival of CD56+ and CD56 ALCL cases.

The CD56+ group has a significantly worse prognosis (P = .002).

Fig. 3.

Overall survival of CD56+ and CD56 ALCL cases.

The CD56+ group has a significantly worse prognosis (P = .002).

Close modal
Fig. 4.

Prognostic difference between CD56+ and CD56 ALCL according to ALK expression.

The CD56+ group shows a significantly lower survival for both ALK-positive (A, P = .02) and ALK-negative (B,P = .04) subtypes.

Fig. 4.

Prognostic difference between CD56+ and CD56 ALCL according to ALK expression.

The CD56+ group shows a significantly lower survival for both ALK-positive (A, P = .02) and ALK-negative (B,P = .04) subtypes.

Close modal

Prognostic factors for ALCL

Univariate Cox analysis identified the following prognostic factors: age, clinical stage, PS, ALK expression, CD56 expression, EBV positivity, serum LDH level, presence of B symptoms, extranodal involvement of more than one site, and IPI (Table5). Multivariate analysis excluding IPI categories showed age older than 60, advanced stage (III or IV), CD56 positivity, and PS greater than one to be significant and independent prognostic factors (Table 5).

Table 5.

Prognostic factors affecting overall survival

VariablesUnfavorable factorsUnivariateMultivariate5-150
Hazard ratio (CI)P valueHazard ratio (CI)P value
Comparison with risk factors      
 Age > 60 years 3.1  (1.7-5.7) .0003 4.1  (2.1-7.8) .00003  
 CD56 Positive 2.7  (1.4-5.1) .003 3.1  (1.5-6.1) .001 
 Stage III/IV 2.4  (1.2-4.8) .01 2.8  (1.3-6.1) .008  
 PS 2-4 3.4  (1.9-6.1) .00003 2.5  (1.4-4.6) .003  
 EBV Positive 3.2  (1.6-6.7) .002 —  
 ALK Negative 2.5  (1.4-4.9) .001 —  
 B symptom Present 2.1  (1.1-3.7) .01 —  
 Extranodal disease ≧ 2 sites 1.9  (1.1-3.5) .03 —  
 LDH > normal 1.8  (1.0-3.1) .05 —  
Comparison with IPI category      
 IPI category H-I/H 4.1  (2.2-7.3) .000004 4.0  (2.2-7.2) .00001 
 CD56 Positive 2.7  (1.4-5.1) .003 2.6  (1.3-5.0) .004 
VariablesUnfavorable factorsUnivariateMultivariate5-150
Hazard ratio (CI)P valueHazard ratio (CI)P value
Comparison with risk factors      
 Age > 60 years 3.1  (1.7-5.7) .0003 4.1  (2.1-7.8) .00003  
 CD56 Positive 2.7  (1.4-5.1) .003 3.1  (1.5-6.1) .001 
 Stage III/IV 2.4  (1.2-4.8) .01 2.8  (1.3-6.1) .008  
 PS 2-4 3.4  (1.9-6.1) .00003 2.5  (1.4-4.6) .003  
 EBV Positive 3.2  (1.6-6.7) .002 —  
 ALK Negative 2.5  (1.4-4.9) .001 —  
 B symptom Present 2.1  (1.1-3.7) .01 —  
 Extranodal disease ≧ 2 sites 1.9  (1.1-3.5) .03 —  
 LDH > normal 1.8  (1.0-3.1) .05 —  
Comparison with IPI category      
 IPI category H-I/H 4.1  (2.2-7.3) .000004 4.0  (2.2-7.2) .00001 
 CD56 Positive 2.7  (1.4-5.1) .003 2.6  (1.3-5.0) .004 

CI indicates confidence interval; H, high; H-I, high-intermediate.

F5-150

Final model.

When the IPI was included instead of its constitutive factors, only IPI (relative risk [RR] = 4.0; confidence interval [CI], 2.2-7.2;P = .00001) and CD56 (RR = 2.6 CI; 1.3-5.0;P = .004) were identified as independent and significant prognostic factors. According to these findings, all patients were divided into 4 groups with different prognoses on the basis of IPI and CD56 (Figure 5).

Fig. 5.

Overall survival of all ALCL patients stratified according to CD56 expression and IPI category.

The CD56+ and IPI high-intermediate/high subgroups have an extremely poor prognosis (P < .0001).

Fig. 5.

Overall survival of all ALCL patients stratified according to CD56 expression and IPI category.

The CD56+ and IPI high-intermediate/high subgroups have an extremely poor prognosis (P < .0001).

Close modal

In our series of ALCL patients, clear clinicopathologic differences were found between ALK-positive and ALK-negative subtypes, which is consistent with most of the studies in the literature.24,30-32 ALK-positive ALCLs are characterized by a younger age distribution, lower serum LDH level, better PS, less frequent extranodal involvement, lower IPI categories, and better prognosis. Although we could demonstrate that the expression of CD56 on the lymphoma cells is an independent prognostic factor for T/null cell type ALCL, the clinical manifestations of CD56+ and CD56 ALCLs were quite similar. This suggests that CD56 expression is not a relevant factor for the identification of a novel subtype of ALCL but a purely clinical risk factor.

Initial investigation did not identify the IPI risk category as prognostic for ALCL,43 but recent studies with large populations have shown that the IPI is highly prognostic for ALCL.30-32 Our study also confirmed the prognostic significance of IPI for T/null cell type ALCL. In our study, however, multivariate analyses did not identify ALK expression as an independent prognostic factor, but this does not contradict the fact that ALK-positive ALCL is a distinct subtype, because the expression of ALK is closely correlated with age and IPI. It is therefore not fruitful to discuss whether age or ALK expression is more prognostically significant for ALCL, since these 2 factors are interrelated and have the same impact on the prognosis for ALCL. When age or the IPI was not included in the multivariate analysis models, ALK instead of age or the IPI was identified as prognostic in both models. These results confirm that ALK-positive ALCL constitutes a distinct entity.

CD56 has been well documented as being expressed in a variety of NK-cell neoplasms,44-47 but it is also expressed in hematolymphoid malignancies other than those of NK-cell lineage, ie, AML,37,38,48,49 acute lymphoblastic leukemia of both T- and B-cell lineage,49 and some types of T- and B-cell lymphoma.50-53 Its expression is rare in diffuse large B-cell lymphoma,54,55 but common and well investigated in multiple myeloma.56,57 CD56 is expressed mostly on myeloma cells in bone marrow but less frequently on those in peripheral blood (plasma cell leukemia) or extramedullary sites (plasmacytoma), suggesting its adhesive function to bone marrow stroma cells.58-60 In ALCL, Felgar et al61 detected CD56 expression in 8 of 17 cases (47%) of T/null cell type ALCL, but Krenacs et al62 reported much lower frequency of CD56+ cases (1 of 32), and Foss et al63 no CD56 expression in 13 cases. In our larger series, CD56 was expressed in 25 of 140 cases (17%) and was shown to be a prognostic factor. The expression of CD56 has also been shown to be a risk factor in AMLs with t(8;21) and t(15;17),37,38 but is controversial in multiple myeloma. In an early report by Van Camp et al,56CD56 patients were shown to have aggressive clinical courses, but this might simply represent the tumor cell localization of myeloma cells. Mathew et al64 reported similar survival curves for CD56+ and CD56 myeloma, whereas Garcia-Sanz et al65 showed poorer prognosis of myeloma cases with high CD56+ CD3-plasma cells in peripheral blood. For none of the specific entities of malignant lymphoma have any prognostic implications of CD56 expression been argued. The expression of CD56 might also be a prognostic factor for other types of hematolymphoid malignancy. The clinical and prognostic significance of CD56 expression in various types of leukemias and lymphomas therefore deserves to be examined.

The reason CD56 functions as a prognostic factor in ALCL remains unclear. Although CD56 is a neural cell adhesion molecule, the frequency of extranodal involvements for the CD56+ and CD56 groups was not different in our ALCL cases. The expression of CD56 is associated with disease localization in multiple myeloma,58-60 but did not correlate with extramedullary involvement in AML studies with large numbers.48,49 The role of CD56 might be different in different subtypes of hematolymphoid malignancy. Integrin β1, an adhesion molecule that interacts with the extracellular matrix, was recently shown to mediate the anti-apoptotic signal resulting in drug resistance of small-cell lung cancer cells.66 CD56 is a homophilic-binding adhesion molecule, and its expression often appears to be more intense on the adjacent membrane of neighboring cells. It is therefore possible that CD56 also mediates certain intercellular signals and functions as an adverse prognostic factor in various hematolymphoid malignancies, as well as in those with NK-cell lineage.

From the viewpoint of lymphoma classification, HD-like ALCL is not a well-defined entity, although it was included in the REAL classification as a provisional entity.6 A workshop report on HD and related diseases67 and the recently published WHO classification7 emphasized that HD-like ALCL should be separated into T-lineage ALCL and B-lineage HD. Expression of T-cell–related antigens (CD3, CD43, CD45RO) or EMA is strongly in favor of a diagnosis of ALCL; the presence of B-cell markers (CD20) EBV or CD15 favors a diagnosis of HD. For our 11 cases of HD-like ALCL, 4 were positive for CD15. However, 3 of the 4 CD15+ cases were also positive for EMA, and 2 coexpressed T-lineage antigens (CD45RO and CD4, respectively). Phenotypical results do not conflict with the inclusion of these HD-like cases in a category of ALCL. CD15 and EBV were also found, respectively, in 7 and 8 cases without HD-like appearance, indicating that the presence of CD15+ or EBV-positive cases in the ALK-negative group were not because of the inclusion of HD-like ALCLs. The proportions of CD15+ or EBV-positive cases in common and HD-like ALCLs are consistent with those reported in a previous study by Zinzani et al,68although B-cell type ALCLs were also included in their study. In addition, no prognostic differences were found for the HD-like cases in the ALK-negative ALCL, so that we have included the HD-like cases in this study. However, the possibility that these HD-like ALCLs may represent tumor-cell–rich cases of HD deserves further investigation before a strict border is drawn between ALCL and HD.

In our study, 65 of 81 (85.0%) ALK-positive ALCL cases and 30 of 57 (52.6%) ALK-negative ALCL cases expressed cytotoxic molecules, granzyme B, and/or TIA-1. This is consistent with the observations by others of high-frequency cytotoxic molecule expression in ALCL and suggests a possible derivation of ALCL from cytotoxic T cells.30,61-63 In our cases, no differences between cytotoxic-molecule–positive and cytotoxic-molecule–negative cases, including differences in clinicopathologic features and prognosis, could be identified (data not shown). The negative cases, however, especially those in the ALK-positive group, may express cytotoxic molecules other than TIA-1 or granzyme B. Further investigations are needed to determine the origin of these cytotoxic-molecule–negative ALCLs.

Recently, Falini et al42 established that the staining pattern of ALK is defined by the chimeric partner of ALKgene as a result of its oncogenic translocations. They showed that 44 of 59 ALK-positive ALCL cases (75%) possess NPM-ALK and 15 to have variant ALK chimera. ALK-N/NC staining means that lymphoma cells have an NPM-ALK fusion protein as a result of t(2;5)(p23;q35), whereas ALK-C staining is derived from other variant ALK fusion and 2p23 abnormalities.14,69-73 Some of the fusion partners of ALK in these variant translocations have recently been cloned and identified as TPM3, TFG, AITC, andCLTCL genes.74-79 Although these 4 genes have no homologous region or function, they were fused to the ALKgene at a similar break point, at just 3′ of the transmembrane region. As a result, this transmembrane portion was lost in the x-ALK chimeric products, whereas the tyrosine kinase domain was preserved. These findings suggest that the oncogenic event accounting for these 2p23 translocations is the deregulation of the aberrant ALK gene expression by the promoter/enhancer of the fusion partners. We determined that 50 of 75 cases (67%) showed ALK-N/NC staining, but could not identify any clinicopathologic, immunophenotypic, or prognostic differences between ALK-N/NC and ALK-C groups. Our result suggests that the staining pattern of ALK does not define a distinct subtype. This is consistent with the speculation that the oncogenicity of the aberrant ALK expression is not affected by the fusion partners.

Several recent studies have been performed on the basis of age categorization as pediatric80,81 or adult.32,67,82 In our ALK-positive ALCL cases, however, both pediatric and adult patients showed identical clinicopathologic characteristics. The entity of ALK-positive ALCL therefore transcends the arbitrary boundaries of 15 or 20 years of age, so that there seems to be no good reason to divide this disease into 2 age categories, pediatric and adult, for a more accurate understanding of the disease. Pediatric and adult cases tended to be treated with different therapeutic protocols, however, mainly owing to the physician's specialization, either pediatrics or internal medicine. The appropriate therapeutic approach for ALCL should be investigated from the viewpoint of a continuous spectrum of ALCL, at least for children and adolescents/young adults. A prospective clinical trial is needed to explore an effective therapeutic approach for ALCL. For this, we recommend that both pediatric and adult patients be treated with a consistent strategy.

In conclusion, we propose that for clinical studies of ALCL, CD56 expression as well as the IPI should be included in the prognostic factors used for patient stratification.

We thank H. Ishida and Y. Tokoro for technical assistance, and the collaborators from the following institutions for providing the patients' data and specimens: National Sapporo Hospital; Sapporo Municipal Hospital; Akita University School of Medicine; Japanese Red Cross Ashikaga Hospital; Gunma University School of Medicine; Kitazato University School of Medicine; Tsukuba University School of Medicine; Saitama Cancer Center; Chiba University School of Medicine; Hamamatsu Medical School; Seirei Hamamatsu Hospital; Iida Municipal Hospital; Takaoka Hospital; Toyama Central Hospital; Toyohashi Municipal Hospital; Japanese Red Cross Nagoya First Hospital; Aichi Prefectural Hospital; Okazaki Municipal Hospital; Kariya General Hospital; Kousei Hospital; Tokoname Municipal Hospital; Ichinomiya Municipal Hospital; Nagoya University School of Medicine; Nagoya City University School of Medicine; Higashi Municipal Hospital; Nagoya Ekisaikai Hospital; Nagoya Memorial Hospital; National Nagoya Hospital; National Higashi Nagoya Hospital; Aichi Medical School; Showa Hospital; Gifu Municipal Hospital; Yokkaichi Municipal Hospital; Mie University School of Medicine; Suzuka Central General Hospital; Fukui Saiseikai Hospital; Youka Hospital; National Kyoto Hospital; National Osaka Hospital; Chugoku-chuou Hospital; Okayama Saiseikai Hospital; Okayama Rousai Hospital; Japanese Red Cross Okayama Hospital; Mitoyo General Hospital; Fukuyama National Hospital; Kawasaki Medical School; Japanese Red Cross Takamatsu Hospital; Fukuoka University School of Medicine.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Stein
H
Mason
DY
Gerdes
J
et al
The expression of the Hodgkin's disease associated antigen Ki-1 in reactive and neoplastic lymphoid tissue: evidence that Reed-Sternberg cells and histiocytic malignancies are derived from activated lymphoid cells.
Blood.
66
1985
848
858
2
Piris
M
Brown
DC
Gatter
KC
Mason
DY
CD30 expression in non-Hodgkin's lymphoma.
Histopathology.
17
1990
211
218
3
Nakamura
S
Suchi
T
Koshikawa
T
et al
Clinicopathologic study of 212 cases of peripheral T-cell lymphoma among the Japanese.
Cancer.
72
1993
1762
1772
4
Stansfeld
AG
Diebold
J
Kapanci
Y
et al
Updated Kiel classification for lymphomas.
Lancet.
1
1988
292
293
5
Suchi
T
Lennert
K
Tu
L-Y
et al
Histopathology and immunohistochemistry of peripheral T cell lymphomas: a proposal for their classification.
J Clin Pathol.
40
1987
995
1015
6
Harris
NL
Jaffe
ES
Stein
H
et al
A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group.
Blood.
84
1994
1361
1392
7
Harris
NL
Jaffe
ES
Diebold
J
et al
World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee Meeting–Airlie House, Virginia, November 1997.
J Clin Oncol.
17
1999
3835
3849
8
Benz-Lemoine
E
Brizard
A
Huret
JL
et al
Malignant histiocytosis: a specific t(2;5)(p23;q35) translocation? Review of the literature.
Blood.
72
1988
1045
1047
9
Fischer
P
Nacheva
E
Mason
DY
et al
A Ki-1 (CD30)-positive human cell line (Karpas 299) established from a high grade non-Hodgkin's lymphoma, showing a 2;5 translocation and rearrangement of the T-cell receptor beta-chain gene.
Blood.
72
1988
234
250
10
Kaneko
Y
Frizzera
G
Edamura
S
et al
A novel translocation t(2;5)(p23;q35), in childhood phagocytic large T-cell lymphoma mimicking malignant histiocytosis.
Blood.
73
1989
806
813
11
Morris
SW
Kirstein
MN
Valentine
MB
et al
Fusion of a kinase gene, ALK, to a molecular protein gene, NPM, in non-Hodgkin's lymphoma.
Science.
263
1994
1281
1284
12
Shiota
M
Fujimoto
J
Semba
T
Satoh
H
Yamamoto
T
Mori
S
Hyperphosphorylation of a novel 80 kDa protein-tyrosine kinase similar to Ltk in a human Ki-1 lymphoma cell line, AMS3.
Oncogene.
9
1994
1567
1574
13
Shiota
M
Fujimoto
J
Takenaga
M
et al
Diagnosis of t(2;5)(p23;q35)-associated Ki-1 lymphoma with immunohistochemistry.
Blood.
84
1994
3648
3652
14
Pulford
K
Lamant
L
Morris
SW
et al
Detection of anaplastic lymphoma kinase (ALK) and nucleolar protein nucleophosmin (NPM)-ALK proteins in normal and neoplastic cells with the monoclonal antibody ALK1.
Blood.
89
1997
1394
1404
15
Falini
B
Bigerna
B
Fizzotti
M
et al
ALK expression defines a distinct group of T/null lymphomas (“ALK lymphomas”) with a wide morphological spectrum.
Am J Pathol.
153
1998
875
886
16
Ladanyi
M
Cavalchire
G
Morris
SW
Downing
J
Filippa
DA
Reverse transcriptase polymerase chain reaction for the Ki-1 anaplastic large cell lymphoma-associated t(2;5) translocation in Hodgkin's disease.
Am J Pathol.
145
1994
1296
1300
17
Weiss
LM
Lopategui
JR
Sun
LH
Kamel
OW
Koo
CH
Glackin
C
Absence of the t(2;5) in Hodgkin's disease.
Blood.
85
1995
2845
2847
18
Herbst
H
Anagnostopoulos
J
Heinze
B
Durkop
H
Hummel
M
Stein
H
ALK gene products in anaplastic large cell lymphomas and Hodgkin's disease.
Blood.
86
1995
1694
1700
19
Wellmann
A
Otsuki
T
Vogelbrich
M
Clark
HM
Jaffe
ES
Raffeld
M
Analysis of the t(2;5)(p23;q35) translocation by reverse transcription polymerase chain reaction in CD30+ anaplastic large cell lymphomas and in other non-Hodgkin's lymphomas of T-cell phenotype.
Blood.
86
1995
2321
2328
20
Elmberger
G
Lozano
MD
Weisenburger
DD
Sanger
W
Chan
WC
Transcripts of the npm-alk fusion gene in anaplastic large cell lymphoma, Hodgkin's disease, and reactive lymphoid lesions.
Blood.
86
1995
3517
3521
21
Lamant
L
Meggetto
F
Al Saati
T
et al
High incidence of the t(2;5)(p23;q35) translocation in anaplastic large cell lymphoma and its lack of detection in Hodgkin's disease: comparison of cytogenetic analysis, reverse transcriptase-polymerase chain reaction, and P-80 immunostaining.
Blood.
87
1996
284
291
22
Wood
GS
Hardman
DL
Boni
R
et al
Lack of the t(2;5) or other mutations resulting in expression of anaplastic lymphoma kinase catalytic domain in CD30+ primary cutaneous lymphoproliferative disorders and Hodgkin's disease.
Blood.
88
1996
1765
1770
23
Sarris
AH
Luthra
R
Papadimitracopoulou
V
et al
Amplification of genomic DNA demonstrates the presence of the t(2;5) (p23;q35) in anaplastic large cell lymphoma, but not in other non-Hodgkin's lymphomas, Hodgkin's disease, or lymphomatoid papulosis.
Blood.
88
1996
1771
1779
24
Shiota
M
Nakamura
S
Ichinohasama
R
et al
Anaplastic large cell lymphomas expressing the novel chimeric protein p80NPM/ALK: a distinct clinicopathologic entity.
Blood.
86
1995
1954
1960
25
DeCoteau
JF
Butmarc
JR
Kinney
MC
Kadin
ME
The t(2;5) chromosomal translocation is not a common feature of primary cutaneous CD30+ lymphoproliferative disorders: comparison with anaplastic large-cell lymphoma of nodal origin.
Blood.
87
1996
3437
3441
26
Weisenburger
DD
Gordon
BG
Vose
JM
et al
Occurrence of the t(2;5)(p23;q35) in non-Hodgkin's lymphoma.
Blood.
87
1996
3860
3868
27
Pittaluga
S
Wiodarska
I
Pulford
K
et al
The monoclonal antibody ALK1 identifies a distinct morphological subtype of anaplastic large cell lymphoma associated with 2p23/ALK rearrangements.
Am J Pathol.
151
1997
343
351
28
Nakamura
S
Shiota
M
Nakagawa
A
et al
Anaplastic large cell lymphoma: a distinct molecular pathologic entity: a reappraisal with special reference to p80NPM/ALK expression.
Am J Surg Pathol.
21
1997
1420
1432
29
Benharroch
D
Meguerian-Bedoyan
Z
Lamant
L
et al
ALK-positive lymphoma: a single disease with a broad spectrum of morphology.
Blood.
91
1998
2076
2084
30
ten Berge
RL
Dukers
DF
Oudejans
JJ
et al
Adverse effects of activated cytotoxic T lymphocytes on the clinical outcome of nodal anaplastic large cell lymphoma.
Blood.
93
1999
2688
2696
31
Falini
B
Pileri
S
Zinzani
PL
et al
ALK+ lymphoma: clinico-pathological findings and outcome.
Blood.
93
1999
2697
2706
32
Gascoyne
RD
Aoun
P
Wu
D
et al
Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma.
Blood.
93
1999
3913
3921
33
Griffin
JD
Hercend
T
Beveridge
R
Schlossman
SF
Characterization of an antigen expressed by human natural killer cells.
J Immunol.
130
1983
2947
2951
34
Lanier
LL
Le
AM
Civin
CI
Loken
MR
Phillips
JH
The relationship of CD16(Leu-11) and Leu-19(NKH-1) antigen expression of human peripheral blood NK cells and cytotoxic T lymphocytes.
J Immunol.
136
1986
4480
4486
35
Jaffe
ES
Classification of natural killer (NK) cell and NK-like T-cell malignancies.
Blood.
87
1996
1207
1210
36
Suzuki
R
Nakamura
S
Malignancies of natural killer (NK) cell precursor: myeloid/NK cell precursor acute leukemia and blastic NK cell lymphoma/leukemia.
Leuk Res.
23
1999
615
624
37
Baer
MR
Stewart
CC
Lawrence
D
et al
Expression of the neural cell adhesion molecule CD56 is associated with short remission duration and survival in acute myeloid leukemia with t(8;21)(q22;q22).
Blood.
90
1997
1643
1648
38
Murray
CK
Estey
E
Paietta
E
et al
CD56 expression in acute promyelocytic leukemia: a possible indicator of poor treatment outcome?
J Clin Oncol.
17
1999
293
297
39
Kagami
Y
Suzuki
R
Taji
H
et al
Nodal cytotoxic lymphoma spectrum: a clinicopathologic study of 66 patients.
Am J Surg Pathol.
23
1999
1184
1200
40
Hsu
SM
Raine
L
Fanger
H
Use of avidin-biotin-peroxidase complex (ABC) in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody (PAP) procedures.
J Histochem Cytochem.
29
1981
577
580
41
Kobashi
Y
Nakamura
S
Sasajima
Y
et al
Inconsistent association of Epstein-Barr virus with CD56 (NCAM)-positive angiocentric lymphoma occurring in sites other than the upper and lower respiratory tract.
Histopathology.
28
1996
111
120
42
Falini
B
Pulford
K
Pucciarini
A
et al
Lymphomas expressing ALK fusion protein(s) other than NPM-ALK.
Blood.
94
1999
3509
3515
43
Armitage
JO
Weisenburger
DD
for the Non-Hodgkin's Lymphoma Classification Project
New approach to classifying non-Hodgkin's lymphomas: clinical features of the major histologic subtypes.
J Clin Oncol.
16
1998
2780
2795
44
Suzuki
R
Yamamoto
K
Seto
M
et al
CD7+ and CD56+ myeloid/natural killer cell precursor acute leukemia: a distinct hematolymphoid disease entity.
Blood.
90
1997
2417
2428
45
Nakamura
S
Suchi
T
Koshikawa
T
et al
Clinicopathologic study of CD56 (NCAM)-positive angiocentric lymphoma occurring in sites other than the upper and lower respiratory tract.
Am J Surg Pathol.
19
1995
284
296
46
Fernandez
LA
Pope
B
Lee
C
Zayed
E
Aggressive natural killer cell leukemia in an adult and establishment of an NK cell line.
Blood.
67
1986
925
930
47
Suzumiya
J
Takeshita
M
Kimura
M
et al
Expression of adult and fetal natural killer cell markers in sinonasal lymphomas.
Blood.
83
1994
2255
2260
48
Seymour
JF
Pierce
SA
Kantarjian
HM
Keating
MI
Estey
EH
Investigation of karyotypic, morphologic and clinical features in patients with acute myeloid leukemia blast cells expressing the neural cell adhesion molecule (CD56).
Leukemia.
8
1994
823
826
49
Thomas
X
Vila
L
Campos
L
Sabido
O
Archimbaud
E
Expression N-CAM (CD56) on acute leukemia cells: relationship with disease characteristics and outcome.
Leuk Lymphoma.
19
1995
295
300
50
Hanson
CA
Bockenstedt
PL
Schnitzer
B
Fox
DA
Kueck
B
Braun
DK
S100-positive, T-cell chronic lymphoproliferative disease: an aggressive disorder of an uncommon T-cell subset.
Blood.
78
1991
1803
1813
51
Macon
WR
Williams
ME
Greer
JP
et al
Natural killer-like T-cell lymphomas: aggressive lymphomas of T-large granular lymphocytes.
Blood.
87
1996
1474
1483
52
Cooke
CB
Krenacs
L
Stetler-Stevenson
M
et al
Hepatosplenic T-cell lymphoma: a distinct clinicopathologic entity of cytotoxic γδ T-cell origin.
Blood.
88
1996
4265
4274
53
Hammer
RD
Vnencak-Jones
CL
Manning
SS
Glick
AD
Kinney
MC
Microvillous lymphomas are B-cell neoplasms that frequently express CD56.
Mod Pathol.
11
1998
239
246
54
Kern
WF
Spier
CM
Hanneman
EH
Miller
TP
Matzner
M
Grogan
TM
Neural cell adhesion molecule-positive peripheral T-cell lymphoma: a rare variant with a propensity for unusual sites of involvement.
Blood.
79
1992
2432
2437
55
Sekita
T
Tamaru
J-I
Isobe
K
et al
Diffuse large B cell lymphoma expressing the natural killer cell marker CD56.
Pathol Int.
49
1999
752
758
56
Van Camp
B
Durie
BGM
Spier
C
et al
Plasma cells in multiple myeloma express a natural killer cell-associated antigen: CD56 (NKH-1; Leu-19).
Blood.
76
1990
377
382
57
Harada
H
Kawano
MM
Huang
N
et al
Phenotypic difference of normal plasma cells from mature myeloma cells.
Blood.
81
1993
2658
2663
58
Pellat-Deceunynck
C
Barille
S
Jego
G
et al
The absence of CD56 (NCAM) on malignant plasma cells is a hallmark of plasma cell leukemia and of a special subset of multiple myeloma.
Leukemia.
12
1998
1977
1982
59
Rawstron
A
Barrans
S
Blythe
D
et al
Distribution of myeloma plasma cells in peripheral blood and bone marrow correlates with CD56 expression.
Br J Haematol.
104
1999
138
143
60
Garcia-Sanz
R
Orfao
A
Gonzalez
M
et al
Primary plasma cell leukemia: clinical, immunophenotypic, DNA ploidy, and cytogenetic characteristics.
Blood.
93
1999
1032
1037
61
Felgar
RE
Salhany
KE
Macon
WR
Pietra
GG
Kinney
MC
The expression of TIA-1+ cytolytic-type granules and other cytolytic lymphocyte-associated markers in CD30+ anaplastic large cell lymphomas (ALCL): correlation with morphology, immunophenotype, ultrastructure, and clinical features.
Hum Pathol.
30
1999
228
236
62
Krenacs
L
Wellmann
A
Sorbara
L
et al
Cytotoxic cell antigen expression in anaplastic large cell lymphomas of T- and null-cell type and Hodgkin's disease: evidence for distinct cellular origin.
Blood.
89
1997
980
989
63
Foss
H-D
Anagnostopoulos
I
Araujo
I
et al
Anaplastic large-cell lymphomas of T-cell and null-cell phenotype express cytotoxic molecules.
Blood.
88
1996
4005
4011
64
Mathew
P
Ahmann
GJ
Witzig
TE
Roche
PC
Kyle
RA
Greipp
PR
Clinicopathological correlates of CD56 expression in multiple myeloma: a unique entity?
Br J Haematol.
90
1995
459
461
65
Garcia-Sanz
R
Gonzalez
M
Orfao
A
et al
Analysis of natural killer-associated antigens in peripheral blood and bone marrow of multiple myeloma patients and prognostic implications.
Br J Haematol.
93
1996
81
88
66
Sethi
T
Rintoul
RC
Moore
SM
et al
Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo.
Nat Med.
5
1999
662
668
67
Rudiger
T
Jaffe
ES
Delsol
G
et al
Workshop report on Hodgkin's disease and related diseases (‘grey zone’ lymphoma).
Ann Oncol.
9 (suppl 5)
1998
S31
S38
68
Zinzani
PL
Bendandi
M
Marteli
M
et al
Anaplastic large-call lymphoma: clinical and prognostic evaluation of 90 adult patients.
J Clin Oncol.
14
1996
955
962
69
Sainati
L
Montaldi
A
Stella
M
Putti
MC
Zanesco
L
Basso
G
A novel variant translocation t(2;13) (p23;q34) in Ki-1 large cell anaplastic lymphoma.
Br J Haematol.
75
1990
621
622
70
Pulford
K
Lamant
L
Morris
SW
et al
Detection of anaplastic lymphoma kinase (ALK) and nucleolar protein nucleophosmin (NPM)-ALK proteins in normal and neoplastic cells with the monoclonal antibody ALK1.
Blood.
89
1997
1394
1404
71
Wlodarska
I
De Wolf-Peeters
C
Falini
B
et al
The cryptic inv(2) (p23q35) defines a new molecular genetic subtype of ALK-positive anaplastic large-cell lymphoma.
Blood.
92
1998
2688
2695
72
Mitev
L
Christova
S
Hadjiev
E
et al
A new variant chromosomal translocation t(2;2)(p23;q23) in CD30+/Ki-1+ anaplastic large cell lymphoma.
Leuk Lymphoma.
28
1998
613
616
73
Rosenwald
A
Ott
G
Pulford
K
et al
t(1;2)(q21;p23) and t(2;3)(p23;q21): two novel variant translocations of the t(2;5)(p23;q35) in anaplastic large cell lymphoma.
Blood.
94
1999
362
364
74
Lamant
L
Dastugue
N
Pulford
K
Delsol
G
Mariame
B
A new fusion gene, TPM3-ALK, in anaplastic large-cell lymphoma created by a (1;2) (q25;p23) translocation.
Blood.
93
1999
3088
3095
75
Hernandez
L
Pinyol
M
Hernandez
S
et al
TRK-fused gene (TFG) is a new partner of ALK in anaplastic large cell lymphoma producing two structurally different TFG-ALK translocations.
Blood.
94
1999
3265
3268
76
Ma
Z
Cools
J
Marynen
P
et al
Inv(2)(p23q35) in anaplastic large-cell lymphoma induces constitutive anaplastic lymphoma kinase (ALK) tyrosine kinase activation by fusion to ATIC, an enzyme involved in purine nucleotide biosynthesis.
Blood.
95
2000
2144
2149
77
Colleoni
GW
Bridge
JA
Garicochea
B
Liu
J
Filippa
DA
Ladanyi
M
ATIC-ALK: a novel variant ALK gene fusion in anaplastic large cell lymphoma resulting from the recurrent cryptic chromosomal inversion, inv(2)(p23q35).
Am J Pathol.
156
2000
781
789
78
Trinei
M
Lanfrancone
L
Campo
E
et al
A new variant anaplastic lymphoma kinase (ALK)-fusion protein (ATIC-ALK) in a case of ALK-positive anaplastic large cell lymphoma.
Cancer Res.
60
2000
793
798
79
Touriol
C
Greenland
C
Lamant
L
et al
Further demonstration of the diversity of chromosomal changes involving 2p23 in ALK-positive lymphoma: 2 cases expressing ALK kinase fused to CLTCL (clathrin chain polypeptide-like).
Blood.
95
2000
3204
3207
80
Hutchison
RE
Banki
K
Shuster
D
et al
Use of an anti-ALK antibody in the characterization of anaplastic large-cell lymphoma of childhood.
Ann Oncol.
8(suppl 1)
1997
S37
S42
81
Brugihres
L
LeDeley
MC
Pacquement
H
et al
CD30+ anaplastic large cell lymphoma in children: analysis of 82 patients enrolled in two consecutive studies of the French Society of Pediatric Oncology.
Blood.
92
1998
3591
3598
82
Tilly
H
Gaulard
P
Lepage
E
et al
Primary anaplastic large-cell lymphoma in adults: clinical presentation, immunophenotype, and outcome.
Blood.
90
1997
3727
3734

Author notes

Ritsuro Suzuki, Division of Molecular Medicine, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan; e-mail: rsuzuki@aichi-cc.pref.aichi.jp.

Sign in via your Institution