Anaplastic, CD30+, large-cell lymphoma is now a well-recognized pathologic entity that accounts for 2% to 8% of all lymphomas. Recent progress has been made in the understanding of certain biologic features found in anaplastic large-cell lymphoma, but information about its clinical behavior, in comparison to other large-cell lymphomas, is limited. The pathologic review of a large multicenter study of the treatment of aggressive lymphoma identified 146 cases of anaplastic large-cell lymphoma (ALCL) on the basis of morphology and CD30 expression. We compared initial presentation, immunophenotype, and clinical outcome of these cases with those of the 1,695 nonanaplastic diffuse large-cell lymphomas (non-ALCL) included in the same trial. Patients with ALCL were more likely to be male (P = .018) and were younger (P < .0001) than those with non-ALCL. B symptoms were more frequent in ALCL (P = .006). Skin (P < .0001) and lung (P < .05) involvement was also more frequent in ALCL, but frequency of bone marrow involvement was identical (P = .5). Tumor cell phenotype was B in 56 cases (38%), T in 49 cases (34%), and null in 33 cases (22%). Response to chemotherapy (P = .001), event-free survival (P = .006), and overall survival (P = .0004) were better for ALCL than for non-ALCL. Multivariate analyses identified anaplastic character as an independent factor that predicted a longer survival. Tumor cell phenotype did not influence event-free survival (P = .72) or overall survival (P = .83). ALCL in adults is a clinicopathologic entity which, independent of its phenotypic characteristics, has a better outcome than other diffuse large-cell lymphomas.

ANAPLASTIC large cell lymphoma (ALCL), also called Ki-1 lymphoma, is a morphologically and immunologically distinct subset of non-Hodgkin's lymphoma (NHL) originally described by Stein et al,1 which accounts for 2% to 8% of all lymphomas. This disease is characterized by the proliferation of pleomorphic large neoplastic lymphoid cells, which strongly express the CD30 antigen (Ki-1 antigen), usually growing in a cohesive pattern and preferentially spreading in the lymph node sinuses.1-4 

Recent studies have shown that ALCL is an heterogeneous group of diseases, differing in their histology, phenotype, cytogenetics, and clinical course.5 Morphologic heterogeneity of ALCL explains why many cases were formerly diagnosed as malignant histiocytosis, Hodgkin's disease, or nonhematopoietic tumors.4-6 Although a majority of ALCL express antigens of T- or B-cell lineage, some cases may lack lymphoid antigens and very rare cases express both T- and B-cell markers.6 The recently proposed Revised European-American Lymphoma (REAL) classification of lymphoid neoplasms4 included the B-cell type of ALCL among the morphologic variants of diffuse large B-cell lymphoma, limiting the term of anaplastic large cell lymphoma to T- and null-cell types. The translocation t(2; 5)(p23; q35), initially described in T-cell ALCL,7 has now been cloned.8 The specificity of this translocation in the spectrum of ALCL and Hodgkin's disease is currently under investigation.9 10 

Although rare cases of ALCL occur in patients with a history of previous lymphoma, most cases are qualified as primary. Two distinct clinical forms of primary ALCL are now recognized: limited to the skin and systemic.4 The purely cutaneous ALCL may be indistinguishable from lymphomatoid papulomatosis and regressing atypical histiocytosis and, like these two entities, may undergo spontaneous regression.11 Conversely, systemic ALCL has an aggressive clinical course and patients frequently present with systemic symptoms, advanced-stage disease, and extranodal localizations.5,12 Response to treatment and overall survival of systemic ALCL in children is good. In adults, however, whether or not prognosis of ALCL is different from other diffuse large cell lymphomas is still controversial.12-27 

The purpose of this study was to compare clinical, immunopathologic, and evolutive features of 146 adult patients with primary ALCL to 1,695 cases of nonanaplastic diffuse large-cell lymphoma (non-ALCL). All of these patients were enrolled in a prospective multicenter study and were uniformly staged and treated.

Patients. In October 1987, the Groupe d'Etudes des Lymphomes de l'Adulte (GELA) initiated a prospective multicenter study, which was designated as LNH-87 study, for patients with aggressive NHL, in which patients were stratified in four treatment groups according to age and prognostic factors. Newly diagnosed patients older than 16 years with intermediate- or high-grade lymphoma were considered eligible for the study. Patients were not included if they had a positive serology to human immunodeficiency virus, a concomitant or previous cancer (except in situ cervix carcinoma or skin epithelioma), congestive heart failure, recent myocardial infarction or conduction abnormalities, uncontrolled diabetes mellitus, and liver or kidney failure.

A review of histologic documents by three independent pathologists from the GELA was planned in the protocol and performed in 88% of the 3,232 patients included. Lymphomas were classified according to the Working Formulation,28 but ALCL, as defined by the updated Kiel classification,29 was added to the categories of this classification. Only patients with a confirmed diagnosis of ALCL were selected for the present study.

Histopathologic and phenotypic analysis. Slides from routinely paraffin-embedded tissues were stained with hematoxylin-eosin, Giemsa, and Gordon-Sweet. For all cases with anaplastic morphology, slides were available for phenotypic study on paraffin sections using immunoperoxidase or alkaline-phosphatase antialkaline phosphatase (APAAP) labeling and a panel of paraffin-resistant antibodies were applied, which included CD45/LC (leukocyte common antigen, MO701; Dako, Glostrup, Denmark), CD30/Ber-H2 (MO751; Dako), CD20/L26 (MO786), CD45R0/UCHL1 (MO742; Dako), CD3 (AO452; Dako), CD68/KP1 (MO814; Dako), CD15 (MO733; Dako), EMA/E29 (epithelial membrane antigen, MO613; Dako), and BNH9 (MO743; Immunotech, Lumigny, France). Additional phenotyping could be obtained on frozen sections in 49 cases.

The histologic and phenotypic criteria used for the definition of ALCL were those defined by Stein et al.1 Briefly, the diagnosis of ALCL was based both on the histopathologic presence of large cells with anaplastic morphology (pleomorphic nuclei, proeminent nucleoli, and abundant cytoplasm) frequently involving preferentially lymph node sinuses and on the expression of CD30 antigen by the tumor cells. Hodgkin's related30 or Hodgkin's-like variants4 of ALCL were excluded from this study.

Lymphomas were considered of B-cell lineage when tumor cells expressed CD20. They were considered of T-cell origin when tumor cells expressed CD3 or, when CD3 negative, expressed CD45RO, but not CD20 and CD68. On frozen sections, cases were considered as T-cell lymphoma when they expressed CD3 or at least two other T-cell antigens (CD2, CD4, CD5, or CD7). Histiocytic type was suspected when only CD45RO and CD68 were expressed on paraffin sections and CD4 was the only T-cell marker expressed on frozen sections.

The group of patients used for comparison consisted of the nonanaplastic diffuse large-cell lymphomas (diffuse large-cell and immunoblastic types of the Working Formulation).27 These cases were only studied on paraffin sections.

Staging. The extent of the disease was studied by physical examination, computerized tomographic scan of chest and abdomen, cerebrospinal fluid examination, and bone marrow biopsy. The number of extranodal sites and the diameter of the largest tumor mass was determined. Patients were staged according to the Ann Arbor classification.31 Performance status was assessed according to the Eastern Cooperative Oncology Group (ECOG) scale (0 to 4).32 Serum lactate dehydrogenase (LDH) level was expressed as the ratio over the maximum normal value.

Treatment and assessment of response. Patients in the LNH-87 study were enrolled in four different randomized trials according to age and to the presence of any of the following adverse prognostic factors: ECOG performance status of 2 to 4, two or more extranodal sites, tumor burden ≥10 cm in largest dimension, bone marrow or central nervous system involvement, and Burkitt's or lymphoblastic subtypes.

Patients in group 1 were younger than 70 years and had no adverse prognostic factor. Treatment in this group randomly compared the LNH-84 regimen33 consisting of three cycles of ACVBP (doxorubicine, cyclophosphamide, vindesine, bleomycin, prednisone) chemotherapy followed by a consolidation phase with high-dose methotrexate, ifosfamide plus etoposide, asparaginase and cytosine-arabinoside, versus eight cycles of m-BACOD (methotrexate, bleomycin, cyclophosphamide, vincristine, dexamethasone) as described by Shipp et al.34 

Patients in group 2 were younger than 55 years and had at least one of the adverse prognostic factors.35 The induction treatment was randomized between ACVBP and NCVBP (mitoxantrone, cyclophosphamide, vindesine, bleomycin, prednisone) where mitoxantrone replaced doxorubicin. Patients in complete remission were then randomly assigned to receive either the LNH-84 consolidation chemotherapy or intensive consolidation and autologous bone marrow support.35 

Patients in group 3 were aged between 55 and 70 years and had at least one adverse prognostic factor. Patients in this group were randomly assigned to receive either the LNH-84 regimen or an alternating chemotherapy VIMMM (VM26, ifosfamide, mitoxantrone, methyl-gag, methotrexate)/ACVBP.36 

Patients in group 4 were patients over 70 years of age. This study prospectively investigated the effect of the addition of tetrahydropyranyl (THP)-doxorubicin to the CVP regimen (cyclophosphamide, vincristine, prednisone.37 

Criteria for assessment of response to treatment have been previously described.35 At the time of this analysis, the median follow-up period was 55 months, and the maximal follow-up period was 8 years.

Statistical methods. Patient characteristics and complete remission rates were compared by χ2 tests. The event-free survival was measured from the date of randomization to disease progression, relapse, or death. The overall survival was measured from the date of randomization to death from any cause or date of last follow-up evaluation. The rates of survival were estimated by the method of Kaplan and Meier38 and compared by log-rank tests. Stepwise Cox proportional-hazards regression analysis with the overall survival as the dependent variable was used to adjust the effect of anaplastic character for potential independent prognostic factors.39 Tests for comparison were regarded as significant if the two-sided P value was less than .05.

Among the 1,841 patients with diffuse large-cell lymphoma included in the LNH-87 trial who had a histologic review, 146 (8%) were classified as having ALCL.

Clinical presentation. Main clinical characteristics of patients with ALCL as compared with those with non-ALCL are listed in Table 1. The male/female ratio was 1.81:1 in ALCL, the male predominance being significantly more important than in non-ALCL. Patients with ALCL were significantly younger than those with non-ALCL (P < .0001). B symptoms were more frequent in ALCL. As in non-ALCL, a majority of patients with ALCL had a disseminated disease (Ann Arbor stage III or IV) and a limited number of extranodal sites. However, skin and lung lesions were more frequent in ALCL, occurring respectively in 15% (P < .0001) and 16% (P < .05) of these patients. Conversely, lesions of the digestive tract were less frequently found in ALCL (P < .002). Bone marrow involvement was detected in 15% of the patients with ALCL as compared with 17% of the patients with non-ALCL (P = .5). ALCL and non-ALCL had a similar distribution in the different risk groups of the lymphoma international prognostic index.32 

Table 1.

Characteristics of Patients With Either ALCL or Non-ALCL

CharacteristicAnaplasticNonanaplasticP
 
No. 146 1,695 
 n (%) n (%) 
Sex 
94 (64) 918 (54) 
52 (36) 777 (46) .018 
Age (yr) 
≤60 107 (73) 988 (58) 
>60 39 (27) 707 (42) <.0001 
Performance status* 
0-1 103 (72) 1,221 (74) 
>1 40 (28) 420 (26) .5 
B symptoms* 
Absent 67 (47) 939 (58) 
Present 77 (53) 673 (42) .006 
Mediastinal involvement 55 (38) 539 (32) .2 
Extranodal sites 
Bone marrow 21 (15) 268 (17) .5 
Liver 17 (12) 188 (11) .8 
Spleen 26 (18) 333 (20) .6 
Central nervous system 11 (8) 129 (8) 
Skin 22 (15) 93 (5) <.0001 
Lung 23 (16) 210 (12) .05 
Gut 10 (7) 277 (16) .002 
Bone 12 (8) 138 (8) 
Waldeyer's ring 11 (8) 187 (11) .2 
No. of extranodal sites 
0-1 site 112 (77) 1,290 (76) 
>1 sites 34 (23) 405 (24) .9 
Ann Arbor stage 
I-II 59 (40) 683 (41) 
III-IV 87 (60) 969 (59) .8 
LDH level* 
≤1 × normal 69 (50) 659 (43) 
>1 × normal 68 (50) 877 (57) .09 
Albumin level* 
≤30 g/L 34 (24) 271 (17) 
>30 g/L 106 (76) 1,294 (83) .04 
International prognostic index31* 
Low 63 (45) 564 (36) 
Low-intermediate 28 (20) 405 (26) 
High-intermediate 23 (17) 349 (22) 
High 25 (18) 253 (16) .08 
Treatment received 
ACVBP 69 (47) 714 (42) 
m-BACOD 22 (15) 187 (11) 
NCVBP 22 (15) 251 (15) 
VIMMM 18 (12) 241 (14) 
CVP/CTVP 15 (10) 302 (18) .2 
CharacteristicAnaplasticNonanaplasticP
 
No. 146 1,695 
 n (%) n (%) 
Sex 
94 (64) 918 (54) 
52 (36) 777 (46) .018 
Age (yr) 
≤60 107 (73) 988 (58) 
>60 39 (27) 707 (42) <.0001 
Performance status* 
0-1 103 (72) 1,221 (74) 
>1 40 (28) 420 (26) .5 
B symptoms* 
Absent 67 (47) 939 (58) 
Present 77 (53) 673 (42) .006 
Mediastinal involvement 55 (38) 539 (32) .2 
Extranodal sites 
Bone marrow 21 (15) 268 (17) .5 
Liver 17 (12) 188 (11) .8 
Spleen 26 (18) 333 (20) .6 
Central nervous system 11 (8) 129 (8) 
Skin 22 (15) 93 (5) <.0001 
Lung 23 (16) 210 (12) .05 
Gut 10 (7) 277 (16) .002 
Bone 12 (8) 138 (8) 
Waldeyer's ring 11 (8) 187 (11) .2 
No. of extranodal sites 
0-1 site 112 (77) 1,290 (76) 
>1 sites 34 (23) 405 (24) .9 
Ann Arbor stage 
I-II 59 (40) 683 (41) 
III-IV 87 (60) 969 (59) .8 
LDH level* 
≤1 × normal 69 (50) 659 (43) 
>1 × normal 68 (50) 877 (57) .09 
Albumin level* 
≤30 g/L 34 (24) 271 (17) 
>30 g/L 106 (76) 1,294 (83) .04 
International prognostic index31* 
Low 63 (45) 564 (36) 
Low-intermediate 28 (20) 405 (26) 
High-intermediate 23 (17) 349 (22) 
High 25 (18) 253 (16) .08 
Treatment received 
ACVBP 69 (47) 714 (42) 
m-BACOD 22 (15) 187 (11) 
NCVBP 22 (15) 251 (15) 
VIMMM 18 (12) 241 (14) 
CVP/CTVP 15 (10) 302 (18) .2 
*

Data were unavailable for some patients.

Histopathology and immunophenotype. Most cases (129 of 146) were consistent with the common type of ALCL.6 Nine cases were considered as small-cell variant,40 five cases as giant cell rich variant, and the remaining three as lymphohistiocytic type.6 We were unable to delineate the pleomorphic and the monomorphic types as described by Chott et al15 or the pale cell and basophilic cell types as described by Chan et al.13 Partial involvement with pseudo-metastatic pattern was found in 33 patients. Additional morphologic features were: important necrosis in 6 cases, erythrophagocytosis in 3, numerous eosinophils in 3, and angiocentrism in 1 case.

By definition, all cases of ALCL had evidence of expression of CD30 by tumor cells. In each specimen, 75% to 100% of the tumor cells reacted with the anti-CD30 antibodies as a dot in the Golgi zone and/or a membrane staining. Seventy-eight percent of the cases expressed CD45, 42% EMA, 23.5% BNH9, and 16.5% CD15. In the eight CD45 negative cases with a null phenotype, the lack of cytokeratin and melanoma markers was assessed.

As shown in Table 2, the phenotype was considered as B in 56 cases (38%). T-lineage antigens were detected in 49 cases (34%), confirmed on frozen material in 19 cases: 31 cases reacted with CD3 antibodies (with or without CD45RO coexpression), 12 cases with CD45RO only (in the absence of B-cell and CD68 markers) and six cases disclosed T-cell antigens on frozen sections only. B- and T-cell markers were coexpressed in four cases (3%) studied on both paraffin and frozen sections. Four other cases (3%), studied both on paraffin and frozen sections, were considered to have a histiocytic origin. The remaining 33 cases (23%) lacked B- and T-cell antigens expression using our limited panel. In the non-ALCL group, 1,368 cases (81%) expressed B-cell antigens, 144 T-cell antigens (8.5%), and 183 cases (11%) did not show detectable lineage antigens on paraffin-embedded sections.

Table 2.

Characteristics of Patients With Anaplastic Large-Cell Lymphoma According to the Phenotype of Tumor Cells

CharacteristicBTNullP
No.* 56 49 33 
Sex 
30 38 19 
26 11 14 .03 
Age (yr) 
≤60 36 41 23 
>60 20 10 .08 
Performance status 
0-1 41 36 21 
>1 14 11 12 .4 
B symptoms 
Absent 28 24 13 
Present 27 24 20 .5 
Mediastinal involvement 26 12 13 .07 
Extranodal sites 
Bone marrow .7 
Liver .3 
Spleen 14 .2 
Central nervous system .8 
Skin 13 .01 
Lung 10 .5 
Gut .8 
Bone .6 
Waldeyer's ring .8 
No. of extranodal sites 
0-1 site 41 38 27 
>1 sites 15 11 .6 
Ann Arbor stage 
I-II 20 27 10 
III-IV 36 22 23 .05 
LDH level 
≤1 × normal 19 32 13 
>1 × normal 31 16 18 .01 
Albumin level* 
≤30 g/L 10 15 
>30 g/L 43 33 25 .3 
International prognostic index31 
Low 16 31 12 
Low-intermediate 16 
High-intermediate 
High 10 .01 
Treatment received 
ACVBP 28 20 17 
m-BACOD 13 
NCVBP 
VIM3/ACVBP 
CVP/CTVP .3 
CharacteristicBTNullP
No.* 56 49 33 
Sex 
30 38 19 
26 11 14 .03 
Age (yr) 
≤60 36 41 23 
>60 20 10 .08 
Performance status 
0-1 41 36 21 
>1 14 11 12 .4 
B symptoms 
Absent 28 24 13 
Present 27 24 20 .5 
Mediastinal involvement 26 12 13 .07 
Extranodal sites 
Bone marrow .7 
Liver .3 
Spleen 14 .2 
Central nervous system .8 
Skin 13 .01 
Lung 10 .5 
Gut .8 
Bone .6 
Waldeyer's ring .8 
No. of extranodal sites 
0-1 site 41 38 27 
>1 sites 15 11 .6 
Ann Arbor stage 
I-II 20 27 10 
III-IV 36 22 23 .05 
LDH level 
≤1 × normal 19 32 13 
>1 × normal 31 16 18 .01 
Albumin level* 
≤30 g/L 10 15 
>30 g/L 43 33 25 .3 
International prognostic index31 
Low 16 31 12 
Low-intermediate 16 
High-intermediate 
High 10 .01 
Treatment received 
ACVBP 28 20 17 
m-BACOD 13 
NCVBP 
VIM3/ACVBP 
CVP/CTVP .3 
*

Four other patients had both B- and T-cell markers and four patients had histiocytic type.

Data were unavailable for some patients.

ALCL with skin involvement had preferentially a T-cell phenotype (P < .01). T-cell ALCL, as compared with B-cell and null ALCL, had a less disseminated disease (P < .05) and a lower LDH level (P < .01) at diagnosis. Consequently, ALCL of T-cell phenotype was more frequently classified in the low risk category of the international lymphoma prognostic index (P < .01).

Response to treatment and survival. The rate of patients who obtained a complete remission was 75% in the ALCL group and 61% in the non-ALCL group (P < .001). As shown in Fig 1, survival of patients with ALCL (estimated survival at 5 years, 66%; 95% confidence interval, 58 to 74) was significantly longer than those with non-ALCL (estimated survival at 5 years, 48.3%; 95% confidence interval, 45.8 to 50.8) (P = .0004). A significant difference was also observed in event-free survival (P = .006). Other factors were found to be related to survival in univariate analysis, namely: age, performance status, presence of B symptoms, LDH level, serum albumin level, Ann Arbor stage, bone marrow involvement, and number of extranodal sites of the disease. A multivariate analysis using these factors and histology (anaplastic v nonanaplastic) identified the anaplastic histology as an independent factor related to survival (P = .0028) (Table 3).

Fig. 1.

Overall survival from the time of inclusion in the LNH-87 trial. Comparison of patients with ALCL (n = 146) and patients with nonanaplastic diffuse large-cell lymphoma (n = 1,695); P = .0004.

Fig. 1.

Overall survival from the time of inclusion in the LNH-87 trial. Comparison of patients with ALCL (n = 146) and patients with nonanaplastic diffuse large-cell lymphoma (n = 1,695); P = .0004.

Close modal
Table 3.

Factors Independently Related to Overall Survival in 1,710 Cases of Diffuse Large-Cell Lymphoma

FactorRelative RiskP
Age (≤60 v >60) 1.72 <.0001 
Ann Arbor stage (I-II v III-IV) 1.60 <.0001 
LDH level (≤1 × normal v >1 × normal) 1.59 <.0001 
Performance status (0-1 v 2-4) 1.59 <.0001 
Histology (anaplastic v nonanaplastic) 1.60 .0028 
B symptoms (no v yes) 1.07 .0097 
Serum albumin level (≤30 g/L v <30 g/L) 1.21 .01 
Bone marrow involvement (no v yes) 1.04 .036 
Extranodal involvement (≤1 site v >1 site) 1.18 .05 
FactorRelative RiskP
Age (≤60 v >60) 1.72 <.0001 
Ann Arbor stage (I-II v III-IV) 1.60 <.0001 
LDH level (≤1 × normal v >1 × normal) 1.59 <.0001 
Performance status (0-1 v 2-4) 1.59 <.0001 
Histology (anaplastic v nonanaplastic) 1.60 .0028 
B symptoms (no v yes) 1.07 .0097 
Serum albumin level (≤30 g/L v <30 g/L) 1.21 .01 
Bone marrow involvement (no v yes) 1.04 .036 
Extranodal involvement (≤1 site v >1 site) 1.18 .05 

The International Lymphoma Prognostic Index,32 developed for aggressive lymphoma in general, predicted survival in patients with ALCL. Overall 5-year survival was, respectively, 82% for the low risk group (95% confidence interval, 72.7 to 92.5), 78.3% for the low-intermediate risk group (95% confidence interval, 62.9 to 93.7), 50.2% for the high-intermediate risk group (95% confidence interval, 28.8 to 71.6), and 25.7% for the high risk group (95% confidence interval, 6.5 to 44.9) (P < .0001). In the non-ALCL patients, overall 5-year survival was, respectively, 69.3% for the low risk group (95% confidence interval, 65.3 to 73.3), 47.1% for the low-intermediate risk group (95% confidence interval, 41.8 to 52.4), 34.5% for the high-intermediate risk group (95% confidence interval, 28.7 to 40.2), and 22% for the high risk group (95% confidence interval, 16.6 to 27.4). A second multivariate analysis was made to evaluate the additional prognostic value of histology on the International Prognostic Index. In this analysis, putting only the Index risk groups into the model along with histology (Table 4), the anaplastic character was again independently prognostic (P < .0011).

Table 4.

Multivariate Analysis of Overall Survival in Large-Cell Lymphoma Using the International Prognostic Index32 Risk Groups and Histology as Variables

FactorRelative RiskP
International Prognostic Index (low v low intermediate v high intermediate v high risk group) 1.70 <.0001 
Histology (anaplastic v nonanaplastic) 1.62 <.0011 
FactorRelative RiskP
International Prognostic Index (low v low intermediate v high intermediate v high risk group) 1.70 <.0001 
Histology (anaplastic v nonanaplastic) 1.62 <.0011 

n = 1,710.

In the ALCL group, the complete remission rate was 80% for B-cell subtype, 69% for T-cell subtype, and 64% for null subtype (P = .09). The differences between subtypes in overall survival (Fig 2) and event-free survival were not significant (respectively, P = .83 and P = .72).

Fig. 2.

Overall survival from the time of inclusion in the LNH-87 trial of patients with ALCL according to immunophenotype: B-cell (n = 56), T-cell (n = 49), and null type (n = 33). P = .83.

Fig. 2.

Overall survival from the time of inclusion in the LNH-87 trial of patients with ALCL according to immunophenotype: B-cell (n = 56), T-cell (n = 49), and null type (n = 33). P = .83.

Close modal

As shown in Fig 3 and 4, B-cell ALCL (estimated survival at 5 years, 71%; 95% confidence interval, 58.2 to 83.8) had a longer survival than non-ALCL of the same phenotype (estimated survival at 5 years, 51%; 95% confidence interval, 48.2 to 53.8) (P = .037) and T-cell ALCL (estimated survival at 5 years, 63.2%; 95% confidence interval, 47.8 to 78.6), a longer survival than T-cell non-ALCL (estimated survival at 5 years, 36%; 95% confidence interval, 27.5 to 44.5) (P = .0061).

Fig. 3.

Overall survival from the time of inclusion in the LNH-87 trial. Comparison of patients with ALCL of B-cell type (n = 56) and patients with nonanaplastic diffuse large-cell lymphoma of B-cell type (n = 1,368); P = .037.

Fig. 3.

Overall survival from the time of inclusion in the LNH-87 trial. Comparison of patients with ALCL of B-cell type (n = 56) and patients with nonanaplastic diffuse large-cell lymphoma of B-cell type (n = 1,368); P = .037.

Close modal
Fig. 4.

Overall survival from the time of inclusion in the LNH-87 trial. Comparison of patients with ALCL of T-cell type (n = 49) and patients with non-ALCL of T-cell type (n = 144); P = .0061.

Fig. 4.

Overall survival from the time of inclusion in the LNH-87 trial. Comparison of patients with ALCL of T-cell type (n = 49) and patients with non-ALCL of T-cell type (n = 144); P = .0061.

Close modal

Of the 22 patients with cutaneous involvement, only five had a disease limited to the skin. Three of these patients had a bulky mass (>10 cm) and two had multifocal disease. As shown in Fig 5, the overall survival of the 22 patients with cutaneous ALCL (estimated survival at 5 years, 90.9%; 95% confidence interval, 78.9 to 100) was significantly longer than those of the 124 patients without this localization (estimated survival at 5 years, 61.6%; 95% confidence interval, 52.6 to 70.6) (P = .0258).

Fig. 5.

Overall survival from the time of inclusion in the LNH-87 trial of patients with ALCL according to the presence (n = 22) or absence (n = 124) of cutaneous involvement at the time of diagnosis. P = .0258.

Fig. 5.

Overall survival from the time of inclusion in the LNH-87 trial of patients with ALCL according to the presence (n = 22) or absence (n = 124) of cutaneous involvement at the time of diagnosis. P = .0258.

Close modal

Since ALCL has been described as a morphologic entity, several clinicopathologic studies have been reported.13-27 However, the small number of patients in the series, the heterogeneity of the treatment received, and the lack of comparison with non-ALCL do not allow drawing firm conclusions in terms of clinical presentation and outcome. Moreover, the varying proportion of ALCL in reported series, ranging from 2% of all lymphomas18 to 22% of the large cell lymphomas22 reflects this difficulty.

The aim of this study was to individualize ALCL in a cohort of patients prospectively treated in a large multicenter trial and to compare their clinical characteristics and treatment outcome with those of patients with non-ALCL included in the same trial. An additional goal of this report was to study the influence of immunophenotype on these parameters.

Our study confirms that ALCL mostly affects young men,15,17 but male predominance appeared only for T-cell type. It is admitted that ALCL patients frequently present with advanced disease,5,17 but initial Ann Arbor stage and number of extranodal sites of the diseases were not different from those of non-ALCL. The higher frequency of B symptoms in ALCL is a common finding,15,22,24,25 which might be related to the elevated levels of the soluble form of CD30 molecule in this disease.41 In Hodgkin's disease, sCD30 levels have been found to be closely correlated with the presence of B symptoms.42,43 Circulating sCD30 in this disease might reflect the effects of cytokines produced by tumor cells.43 

The high frequency of skin involvement is recognized as a characteristic of ALCL.5,21 The clinical distinction between primary cutaneous lymphoma and cutaneous involvement of a systemic disease form is not always easy to establish. Some phenotypic20 and genotypic44 features might help in this distinction. In this series, we confirm the predominance of T-cell phenotype among the cutaneous forms of ALCL. The presence of skin involvement was associated with a longer survival, which is in accordance with the rather favorable outcome reported for adults with primary cutaneous ALCL.11,20,45 However, skin involvement has also been described to be associated with an increased risk of treatment failure in children46 and adults23 with ALCL. As only 15% of the patients with ALCL had skin involvement, their better survival cannot explain the favorable outcome of the whole population observed in our series.

Bone marrow involvement has been initially considered as a rare event in ALCL.6,12 Our study, using conventional methods, did not show any difference in this involvement between ALCL and non-ALCL. When immunohistochemistry with anti-CD30 and anti-EMA is used to study bone marrow samples, malignant cells could be detected in as much as 43% of the cases.47 We confirm the previously reported low incidence of digestive tract involvement, which could be a typical feature of ALCL.19 27 

As CD30 positivity appears more widespread in non-Hodgkin's lymphoma than originally thought, identification of ALCL requires the combination of an histologic and immunophenotypic approach.6,48 Immunohistochemical detection of CD30 expression is particularly useful for the diagnosis of the lymphohistiocytic variant,6 which was only observed in three patients. The development of antibodies directed against chimeric anaplastic lymphoma kinase might be useful in the diagnosis of ALCL, at least in those bearing the t(2; 5) translocation.9 49 

Although no large comparative studies have been published, most investigators reported that response of ALCL to chemotherapy was good, ranging from 60% to 90%.13-27 The overall survival of localized disease is known to be good, especially in children.17,21,27,46 More advanced stages have a high relapse rate and their prognosis in comparison to that of other large-cell lymphomas is controversial. Although few studies have suggested that advanced-stage ALCL may have a short disease-free survival and may require more intensive therapy,21,25 most investigators consider that ALCL generally behaves as a usual intermediate to high grade lymphoma.17,22,24 However, comparative studies in diffuse large-cell lymphomas have shown an association between CD30 expression and a favorable outcome.16,23 A recent study of a large cohort of patients with lymphoma identified ALCL of T/null-cell type as a subgroup with a particularly good survival.50 In our study, ALCL had a longer event-free and overall survival than non-ALCL. In multivariate analyses, anaplastic histology was an independent prognostic factor outweighing some variables, as bone marrow involvement or number of extranodal sites involved, and remaining independent from the risk groups defined by the International Prognostic Index.32 These findings question the introduction of histology as a prognostic factor in future predictive models used for aggressive non-Hodgkin's lymphoma. Finally, the better outcome of ALCL does not support the use of intensive therapy with bone marrow transplant as a front-line treatment in this disease.25 

The predominance of B-cell phenotype in our patients with ALCL is noteworthy. The same finding has only been reported in two recent series of ALCL.24,25 Although we cannot exclude that some ALCL classified as having a null phenotype could, in fact, bear T-cell antigens, only detectable on frozen sections, the proportion of 22% of null type lymphomas found in our study is in accordance with most published series.13-15,18,20-22,24,25,46 The explanation of the predominance of B-cell phenotype could be found in the population studied which excluded children, secondary forms of ALCL, and comprised only a few patients with purely cutaneous involvement, all conditions known to be predominantly of T-cell type.6,11,46 Phenotype was not found to influence the outcome of ALCL, B-, and T-cell ALCL lymphomas, each having a better outcome than their respective non-ALCL counterparts. The larger difference observed between the T-cell lymphomas could be attributed to the poor prognosis of T-cell non-ALCL.51 In contrast to the recent proposition of the REAL classification4 to consider B-cell ALCL as a variant of diffuse large B-cell lymphoma, these results indicate the clinical importance to recognize ALCL, whatever its phenotype could be.

In conclusion, ALCL, which is recognized as a pathologic entity with heterogeneous phenotypic and genotypic features, represents a homogeneous group of patients with an independent prognostic expression. Within the ALCL, phenotype does not influence prognosis, whereas the clinical variables of the International Prognostic Index predict survival. Whether ALCL requires a specific therapeutic approach remains to be determined.

Dr Marie-Françoise d'Agay initiated this work. This report is dedicated to her memory. We thank Catherine Balmale who did the computer programming for this analysis, Catherine Bellorgey for excellent techical assistance, Richard Medeiros for help in the preparation of the manuscript, and Drs Bertrand Coiffier, Christian Gisselbrecht, Félix Reyes, and Philippe Solal-Céligny for their comments on the manuscript.

APPENDIX

The following clinicians and pathologists participated in the LNH-87 study:

A. Abdalsamad, C. Allard, R. Angonin, J. d'Anjou, B. Audhuy, J. Audouin, G. Auzanneau, A.C. Baglin, C. Bailly, Y. Bastion, E. Baumelou, P. Bensimon, F. Berger, P. Biron, A.M. Blaise, M. Blanc, F. Boman-Ferrand, A. Boehn, J. Boniver, M. Bordes, D. Bordessoule, A. Bosly, R. Bouabdallah, S. Boucheron, J. Bouvier, P. Brice, N. Brousse, P. Brousset, P.A. Bryon, D. Caillot, J.P. Carbillet, R.O. Casasnovas, T. Caulet, D. Cazals, F. Charlotte, L. Charvillat, A.M. Chesneau, B. Christian, B. Coiffier, T. Conroy, J.F. Cordier, C. Cordonnier, J.P. Clauvel, E. Deconinck, M. Delage, A. Delannoy, G. Delsol, M. Delos, A. Devidas, M. Diviné, H. Dombret, C. Doyen, H. Duplay, B. Dupriez, C. Duval, J.C. Eisenmann, J.M. Emberger, B. Epardeau, B. Fabiani, P. Felman, J.P. Fermand, A. Ferrant, C. Fermé, A. Ferrand, M. French, M. Fievez, Y. Fonck, N. Froment, J. Gabarre, P. Galian, O. Gasser, C. Gisselbrecht, B. Gosselin, H. Guy, D. Guyotat, C. Haioun, J. Hamels, R. Herbrecht, C. Hopfner, N. Horschowski, F. Huguet, P. Jacomy, J. Jaubert, R. Jeandel, Y. Kerneis, J.P. Knopf, M. Kuentz, E. Labouyrie, B. Lancien, G. Laurent, A. Lavergne, C. Lavignac, V. Leblond, M. Lecomte-Houke, F. Lejeune, M.B. Leger-Ravet, R. Loire, R. Marcellin, J.P. Marolleau, G. Marit, C. Martin, C. Marty-Double, A. de Mascarel, S. Méhaut, J.P. Merlio, C. Merignargues, J.M. Micléa, J.L. Michaux, V. de Montpreville, M. Monconduit, P. Morel, F. Morvan, J.F. Mosnier, G. Nédellec, G. Netter-Pinon, H. Noel, C. Nouvel, M. Patey, P.Y. Peaud, G. Perie, M. Peuchmaur, B. Pignon, C. Platini, M. Pluot, J.P. Pollet, E. Pujade-Lauraine, M. Raphael, M.C. Raymond-Gelle, J. Reiffers, F. Reyes, M. Rochet, J.F. Rossi, A.M. Roucayrol, A. Rozenbaum, G. Salles, H. Schill, C. Sebban, M. Simon, Ph. Solal-Céligny, P. Straub, E. Suc, L. Sutton, G. Tertian, S. Thiebaut, A. Thyss, Ph. Travade, V. Trillet, J.P. Vernant, L. Xerri.

Supported in part by grants from the Délégation à la Recherche Clinique de l'Assistance Publique-Hôpitaux de Paris, Paris, France; the Fondation contre la Leucémie, Paris, France; and the Caisse Nationale d'Assurance Maladie des Travailleurs Salariés, Paris, France.

Address reprint requests to Hervé Tilly, MD, Centre Henri Becquerel, Rue d'Amiens, 76038 Rouen, France.

1
Stein
H
Mason
DY
Gerdes
J
O'Connor
N
Wainscoat
J
Pallesen
G
Gatter
K
Falini
B
Delsol
G
Lemke
H
Schwarting
R
Lemmert
K
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
2
Agnarsson
BA
Kadin
ME
Ki-1 positive large cell lymphoma. A morphologic and immunologic study of 19 cases.
Am J Surg Pathol
12
1988
264
3
Delsol
G
Al Saati
T
Gatter
K
Gerdes
J
Scwaring
R
Caverivière
P
Rigal-Huguet
F
Robert
A
Stein
H
Mason
DY
Coexpression of epithelial membrane antigen (EMA), Ki-1, and interleukin-2 receptor by anaplastic large cell lymphomas: Diagnostic value in so-called malignant histiocytosis.
Am J Pathol
130
1988
59
4
Harris
NL
Jaffe
ES
Stein
H
Banks
PM
Chan
JKC
Cleary
ML
Delsol
G
De Wolf-Peeters
C
Falini
B
Gatter
KC
Grogan
TM
Isaacson
PG
Knowles
DM
Mason
DY
HK
Muller-Hermelink
Pileri
S
Piris
MA
Ralfkaier
E
Warnke
RA
A revised European-American classification of lymphoid neoplasms: A proposal from the international lymphoma study group.
Blood
84
1994
1361
5
Kadin ME: Primary Ki-1-positive anaplastic large-cell lymphoma: A distinct clinicopathologic entity. Ann Oncol 5:25, 1994 (suppl 1)
6
Stein HD, Dallenbach F: Diffuse large cell lymphomas of B and T cell type, in Knowles DM (ed): Neoplastic Hematopathology. Baltimore, MD, Williams & Wilkins, 1992 p 675
7
Mason
DY
Bastard
C
Rimokh
R
Dastugue
N
Huret
JL
Kristofferson
U
Magaud
JP
Nezelof
C
Tilly
H
Vannier
JP
Hemet
J
Warnke
R
CD30 positive large cell lymphomas (Ki1 lymphomas) are associated with a chromosomal translocation involving 5q35.
Br J Haematol
74
1990
161
8
Morris
SW
Kirstein
MN
Valentine
MB
Dittmer
KG
Shapiro
DN
Saltman
DL
Look
AT
Fusion of a kinase gene ALK, to a nucleolar protein gene, NPM, in non Hodgkin's lymphoma.
Science
263
1994
1281
9
Lamant
L
Meggetto
F
Al Saati
T
Brugnières
L
Bressac de Paillerets
B
Dastugue
N
Bernheim
A
Rubie
H
Terrier-Lacombe
MJ
Robert
A
Brousset
P
Rigal
F
Schlaifer
D
Shiota
M
Mori
S
Delsol
G
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
10
Sarris
AH
Luthra
R
Papadimitracopoulou
V
Waasdorp
M
Meletios
A
Dimopoulos
JA
McBride
JA
Cabanillas
F
Duvic
M
Deisseroth
A
Morris
SW
Pugh
WC
Amplification of genomic DNA demonstrates the presence of the t(2; 5)(p23; q35) in anaplastic large cell lymphoma, but not other non-Hodgkin's lymphomas, Hodgkin's disease, or lymphomatoid papulosis.
Blood
88
1996
1771
11
Kadin
ME
The spectrum of Ki-1+ cutaneous lymphoma.
Curr Probl Dermatol
19
1990
132
12
Filippa
DA
Ladanyi
M
Wollner
N
Straus
DJ
O'Brien
JP
Portlock
C
Gangi
M
Sun
M
CD30 (Ki-1)-positive malignant lymphomas: Clinical, immunophenotypic, histologic, and genetic characteristics and differences with Hodgkin's disease.
Blood
87
1996
2905
13
Chan
JCK
Ng
CS
Hui
PK
Leung
TW
Lo
ESF
Lau
WH
McGuire
LJ
Anaplastic large-cell Ki-1 lymphoma: Delineation of two morphological types.
Histopathology
15
1989
11
14
Tashiro
K
Kikuchi
M
Takeshita
M
Yoshida
T
Oshima
K
Clinicopathological study of Ki-1-positive lymphomas.
Pathol Res Pract
185
1989
461
15
Chott
A
Kaserer
K
Augustin
I
Vesely
M
Heintz
R
Oehlinger
W
Hanak
H
Radszkiewcz
T
Ki-1 positive large-cell lymphoma. A clinico pathologic study of 41 cases.
Am J Surg Pathol
14
1990
439
16
Offit
K
Ladanyi
M
Gangi
MD
Ebrahim
SAD
Filippa
D
Chaganti
RSK
Ki-1 expression defines a favorable clinical subset of non-B cell non-Hodgkin's lymphoma.
Leukemia
4
1990
625
17
Greer
JP
Kinney
MC
Collins
RD
Salhany
KE
Wolff
SN
Hainsworth
JD
Flexner
JM
Stein
RS
Clinical features of 31 patients with Ki-1 anaplastic large-cell lymphoma.
J Clin Oncol
9
1991
539
18
Nakamura
S
Takagi
N
Kojima
M
Motoori
T
Kitoh
K
Osada
H
Suzuki
H
Ogura
M
Kurita
S
Oyama
A
Ueda
R
Takahashi
T
Suchi
T
Clinicopathologic study of anaplastic large cell lymphoma (Ki-1 positive large cell lymphoma) among the Japanese.
Cancer
68
1991
118
19
Penny
RJ
Blaustein
JC
Longtine
JA
Pinkus
GS
Ki-1 positive large-cell lymphoma, a heterogeneous group of neoplasms.
Cancer
68
1991
362
20
De Bruin
PC
Beljaards
RC
Van Heerde
P
Van Der Walk
P
Noorduyns
LA
Van Krieken
JHJM
Kluin-Nelemans
JC
Willemze
R
Meijer
CJLM
Differences in clinical behaviour and immunophenotype between primary cutaneous and primary nodal anaplastic large cell lymphoma of T-cell or null cell phenotype.
Histopathology
23
1993
127
21
Schulman
LN
Frisard
B
Antin
JH
Wheeler
C
Pinkus
G
Magauran
N
Mauch
P
Nobles
E
Mashal
R
Canellos
G
Tung
N
Kadin
M
Primary Ki-1 anaplastic large-cell lymphoma in adults: Clinical characteristics and therapeutic outcome.
J Clin Oncol
11
1993
937
22
Pileri
S
Bocchia
M
Baroni
CD
Martelli
M
Falini
B
Sabattini
E
Gherlinzoni
F
Amadori
S
Poggi
S
Mazza
P
Burgio
V
Zinzani
PL
Melilli
G
Benni
M
Saragoni
L
Martelle
MF
Stein
H
Mandelli
F
Tura
S
Anaplastic large cell lymphoma (CD30+/Ki-1+): Results of a prospective clinicopathological study of 69 cases.
Br J Haematol
86
1994
513
23
Romaguera
JE
Manning
JT Jr
Tornos
CS
Rodriguez
J
Brooks
TE
Pugh
WB
Ordonez
NG
Goodacre
AM
Cabanillas
F
Long-term prognostic importance of primary Ki1 (CD30) antigen expression and anaplastic morphology in adults patients with diffuse large cell lymphoma.
Ann Oncol
5
1994
317
24
Tirelli
U
Vaccher
E
Zagonel
V
Talamini
R
Bernardi
D
Tavia
M
Gloghini
A
Merola
MC
Monfardini
S
Carbone
A
CD30 (Ki-1)-positive anaplastic large-cell lymphomas in 13 patients with and 27 patients without human immunodeficiency virus infection: The first comparative clinicopathologic study from a single institution that also includes 80 patients with other human immunodeficiency virus-related lymphomas.
J Clin Oncol
13
1995
373
25
Fanin
R
Silvestri
F
Geromin
A
Cerno
M
Infanti
L
Zaja
F
Barillari
G
Savignano
C
Rinaldi
C
Damiani
D
Buffoli
A
Biffoni
F
Baccarani
M
Primary systemic CD30 (Ki-1)-positive anaplastic large cell lymphoma of the adult: Sequential intensive treatment with F-MACHOP regimen (± radiotherapy) and autologous bone marrow transplantation.
Blood
87
1996
1243
26
Zinzani
PL
Bendandi
M
Martelli
M
Falini
B
Sabattini
E
Amadori
S
Gherlinzoni
F
Martelli
MF
Mandelli
F
Tura
S
Pileri
SA
Anaplastic large-cell lymphoma: Clinical and prognostic evaluation of 90 adult patients.
J Clin Oncol
14
1996
955
27
Clavio
M
Rossi
E
Truini
M
Carrara
P
Ravetti
JL
Spriano
M
Vimercati
AR
Santini
G
Canepa
L
Pierri
I
Celesti
L
Miglino
M
Castellaneta
A
Damasio
E
Gobbi
M
Anaplastic large cell lymphoma: A clinicopathologic study of 53 cases.
Leuk Lymphoma
22
1996
319
28
Non-Hodgkin's lymphoma classification project
National Cancer Institute study of classification of non-Hodgkin's lymphomas: Summary and description of a working formulation for clinical usage.
Cancer
49
1982
2112
29
Stansfeld
AG
Diebold
J
Kapanci
Y
Kelényi
G
Lennert
K
Mioduszewska
O
Noel
H
Rilke
F
Sunstrom
C
Van Unnik
JAM
Wright
DH
Updated Kiel classification for lymphomas.
Lancet
1
1988
292
30
Stein
H
Herbst
H
Anagnostopoulos
I
Niedobitek
G
Dallenbach
F
Kratzsch
HC
The nature of Hodgkin and Reed-Sternberg cells, their association with EBV, and their relationship to anaplastic large cell lymphoma.
Ann Oncol
2
1991
33
31
Carbone
PP
Kaplan
HS
Musshoff
K
Smithers
DW
Tubiana
M
Report of the committee on Hodgkin's disease staging classification.
Cancer Res
31
1991
1860
32
The international non-Hodgkin's lymphoma prognostic factors project. A predictive model for aggressive non-Hodgkin's lymphoma. N Engl J Med 329:987, 1993
33
Coiffier
B
Gisselbrecht
C
Herbrecht
R
Tilly
H
Bosly
A
Brousse
N
LNH-84 regimen: A multicenter study of intensive chemotherapy in 737 patients with aggressive malignant lymphoma.
J Clin Oncol
7
1989
1018
34
Shipp
MA
Yeap
BY
Harrington
DP
Klatt
MM
Pinkus
GS
Jochelson
MS
Rosenthal
DS
Skarin
AT
Canellos
GP
The m-BACOD combination chemotherapy regimen in large-cell lymphoma: Analysis of the completed trial and comparison with the M-BACOD regimen.
J Clin Oncol
8
1990
84
35
Haioun
C
Lepage
E
Gisselbrecht
C
Coiffier
B
Bosly
A
Tilly
H
Morel
P
Nouvel
C
Herbrecht
R
d'Agay
MF
Gaulard
P
Reyes
F
Comparison of autologous bone marrow transplantation with sequential chemotherapy for intermediate-grade and high-grade non-Hodgkin's lymphoma in first complete remission: A study of 464 patients.
J Clin Oncol
12
1994
2543
36
Bosly A, Lepage E, Coiffier B, Dupriez B, Herbrecht R, Fillet G, Diviné M, Nouvel C, Tilly H, Bordessoule D, Gaulard P, Gisselbrecht C: In elderly patients with non-Hodgkin's lymphoma, alternance of chemotherapy is not superior to ACVBP + consolidation: A prospective randomized study on 884 patients LNH87 protocol group 3. Ann Oncol 7:60 1996 (suppl 3, abstr)
37
Bastion Y, Blay JY, Diviné M, Brice P, Bordessoule D, Sebban C, Blanc M, Tilly H, Lederlin P, Deconinck E, Salles B, Brière J, Coiffier B: Elderly patients with aggressive lymphoma survive longer if treated with a curative intent. A GELA study on 453 patients older than 69 years. J Clin Oncol (in press)
38
Kaplan
EL
Meier
P
Nonparametric estimation for incomplete observations.
J Am Stat Assoc
53
1958
457
39
Cox DR: Regression models and life-tables: J R Stat Soc [B] 34:187, 1972
40
Kinney
MC
Collins
RD
Greer
JP
Whitlock
JA
Sioutos
N
Kadin
ME
A small-cell variant of primary Ki-1 (CD30+) T-cell lymphoma.
Am J Surg Pathol
17
1993
859
41
Nadali
G
Vinante
F
Stein
H
Todeschini
G
Tecchio
C
Morosato
L
Chilosi
M
Menestrina
F
Kinney
MC
Greer
JP
Latza
U
Peronna
G
Pizzolo
G
Serum levels of the soluble form of CD30 molecule as a tumor marker in CD30+ anaplastic large-cell lymphoma.
J Clin Oncol
13
1995
1355
42
Gause
A
Pohl
C
Tschiersch
A
Da Costa
L
Jung
W
Diehl
V
Hasenclever
D
Pfreundschuh
M
Clinical significance of soluble CD30 antigen in the sera of patients with untreated Hodgkin's disease.
Blood
77
1991
1983
43
Nadali
G
Vinante
F
Ambrosetti
A
Todeschini
G
Veneri
D
Zanotti
R
Meneghini
V
Ricetti
MM
Benedetti
F
Vassanelli
A
Perona
G
Chilosi
M
Menestrina
F
Fiacchini
M
Stein
H
Pizzolo
G
Serum levels of soluble CD30 are elevated in the majority of untreated patients with Hodgkin's disease and correlate with clinical features and prognosis.
J Clin Oncol
12
1994
793
44
Wood
GS
Hardman
DL
Boni
R
Dummer
R
Kim
YH
Smoller
BR
Takeshita
M
Kikuchi
M
Burg
G
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
45
Paulli
M
Berti
E
Rosso
R
Boveri
E
Kindl
S
Klersy
C
Azzarino
M
Borroni
G
Menestrina
F
Santucci
M
Gambini
C
Vassalo
G
Magrini
U
Sterry
W
Burg
G
Geerts
ML
Meijer
CJLM
Willemze
R
Feller
AC
Müller-Hermelink
HK
Kadin
ME
CD30/Ki-1 positive lymphoproliferative disorders of the skin. Clinicopathologic correlation and statistical analysis of 86 cases: A multicentric study from the European Organization for Research and Treatment of Cancer cutaneous lymphoma project group.
J Clin Oncol
13
1995
1343
46
Reiter
A
Schrappe
M
Tiemann
M
Parwaresch
Zimmermann
Yakisan
E
Dopfer
R
Bucksy
P
Mann
G
Gadner
H
Riehm
H
Successful treatment strategy for Ki-1 anaplastic large-cell lymphoma of childhood: A prospective analysis of 62 patients enrolled in three consecutive Berlin-Frankfurt-Munster group studies.
J Clin Oncol
12
1994
899
47
Fraga
M
Brousset
P
Schlaifer
D
Payen
C
Robert
A
Rubie
H
Huguet-Rigal
F
Delsol
G
Bone marrow involvement in anaplastic large cell lymphoma. Immunohistochemical detection of minimal disease and its prognostic significance.
Am J Clin Pathol
103
1995
82
48
Kadin
ME
Ki1/CD30+ (Anaplastic) large-cell lymphoma: Maturation of a clinicopathologic entity with prospects of effective therapy.
J Clin Oncol
12
1994
884
49
Shiota
M
Fujimoto
J
Takenaga
M
Satoh
H
Ichinohasama
R
Abe
M
Nakano
M
Yamamoto
T
Mori
S
Diagnosis of t(2; 5)(p23; q35)-associated Ki-1 lymphoma with immunohistochemistry.
Blood
84
1994
3648
50
The Non-Hodgkin's Lymphoma Classification Project. A clinical evaluation of the International Lymphoma Study Groupe Classification of non-Hodgkin's lymphoma. Blood 89:3909, 1997
51
Coiffier
B
Brousse
N
Peuchmaur
M
Berger
F
Gisselbrecht
C
Bryon
PA
Diebold
J
Peripheral T-cell lymphomas have a worse prognosis than B-cell lymphomas: A prospective study of 361 immunophenotyped patients treated with the LNH-84 regimen.
Ann Oncol
1
1990
45
Sign in via your Institution