Conventional chromosome analysis (CCA) and interphase fluorescence in situ hybridization (FISH) was performed in 42 patients with mantle-cell lymphoma (MCL), with BCL1 rearrangement. The t(11;14)(q13;q32) or 11q abnormalities were detected by CCA in 34 cases, 20 of which had additional aberrations. A normal karyotype was observed in 8 cases. Probes detecting the chromosome aberrations that were observed in at least 3 cases by CCA, ie, +12, 13q14 deletion, and 17p deletion, were used for interphase FISH analysis. FISH detected total or partial +12, 13q14 deletion and 17p- in 28.5%, 52.4%, and 26% of the cases, respectively. The presence of these anomalies was not a function of karyotype complexity. Based on the results of CCA/FISH, three groups of increasing karyotype complexity were recognized: group 1, including 11 patients without detectable aberrations in addition to BCL1 rearrangement; group 2, including 14 patients with 1 to 2 additional anomalies; and group 3, including 17 patients with three or more additional anomalies. Clinical parameters associated with shorter survival were male sex (P= .006) and primary lymph-node involvement compared with primary bone marrow involvement (P = .015). Trisomy 12 was the only single cytogenetic parameter predictive of a poor prognosis (P = .006) and the best prognostic indicator was the derived measure of karyotype complexity (P < .0001), which maintained statistical significance in multivariate analysis (P< .0001). We arrived at the following conclusions: 13q14 deletion occurs at a high incidence in MCL; 17p deletion and total/partial +12 are relatively frequent events in MCL, the latter aberration being associated with a shorter survival; and the degree of karyotype complexity has a strong impact on prognosis in this neoplasia.

USING SENSITIVE MOLECULAR cytogenetic techniques, the majority of cases of lymphoma of follicle mantle lineage were shown to carry the 11;14 translocation1-4 and its molecular counterpart, consisting of the juxtaposition of theBCL1 locus and the immunoglobulin heavy-chain locus on the derivative 14q+ chromosome.5-7 This disease, which was recognized as a distinct clinicopathological entity as early as 1982,8 accounts for 3% to 9% of all non-Hodgkin’s lymphomas (NHL) in western countries.9 It more frequently affects middle-aged to elderly males, who usually have advanced disease involving lymph nodes and spleen as well as extranodal sites, especially the gastrointestinal (GI) tract and the bone marrow (BM). Frequently, peripheral blood (PB) involvement also occurs, either during disease evolution10 or at diagnosis, mimicking chronic lymphocytic leukemia (CLL).11 

Although a number of hematologic and biologic variables were shown to have correlation with survival in MCL,9,12,13 there is interstudy variability as to the prognostic significance of each of these variables,13-17 partially on account of the limited number of cases studied and of the heterogeneity of patient population.

There is evidence that genetic lesions may help identify prognostically different subgroups of NHL.18-20 Thus, p53 lesions were reproducibly associated with aggressive disease in MCL21,22and in B-cell CLL,23 as was the case with chromosome 17p deletions20,24,25 and aberrations of chromosomes 1, 6, and 1120,26,27 that were found to have prognostic significance in several subtypes of NHL.

In an attempt to better define the incidence and nature of chromosome lesions in MCL and to disclose cytogenetic patterns having prognostic significance, we identified 42 patients with BCL1 involvement and performed conventional cytogenetic analysis (CCA), that identified 13q14 deletion, 17p- and trisomy 12q as the most frequently occurring aberrations in addition to the t(11;14)(q13;q32). These chromosome lesions were subsequently investigated by the more-sensitive interphase fluorescence in situ hybridization (FISH) technique and an analysis was performed of the correlation between these chromosome lesions and salient hematologic parameters.

Patients and clinical parameters.

Fifty-nine patients with non-Hodgkin’s lymphoma (NHL) of follicle mantle lineage were diagnosed at the Institute of Hematology, University of Ferrara (Ferrara, Italy), over a 10-year period. Histologic diagnosis was performed according to recently summarized criteria9,28,29 on lymph-node specimens and/or bone-biopsy sections.

Of these 59 patients, 42 cases fulfilling the following criteria were included in the present study: (1) karyotype available for review. (2) Presence of the t(11;14) (q13;q32), or the corresponding BCL1involvement as detected by FISH technique on representative lymph-node or PB samples. FISH was shown in previous studies to be a very sensitive method for the detection of rearrangements occurring in the BCL1 locus.2,3 (3) Histologic picture of MCL on lymph-node specimens (28 cases with primary lymph-node involvement); or bone biopsy sections consistent with infiltration by MCL (14 patients with primary involvement of the BM and PB who had no superficial adenopathy available for biopsy). (4) Mantle-cell immunophenotype in cases with leukemic expression, ie, CD5/CD19+, CD22+, CD23, CD10, and bright expression of surface immunoglobulins (sIg). Other forms of NHL, CLL, and related disorders (ie, CLL/PL and prolymphocytic leukemia) as well as other chronic (mature) B-cell lymphoproliferative disorders30 were excluded from this analysis, irrespective of the presence of the t(11;14).

Staging procedures included physical examination, a routine laboratory profile, a chest radiograph, and abdomen ultrasonography. When indicated, barium contrast radiography was performed. Computed tomography (CT) scan was performed for staging purposes in 32 cases, bone biopsy was performed in 34 cases (including 14 cases with primary BM involvement). BM aspiration was performed in all cases. PB involvement was studied by light microscopy examination of smears stained by the May-Grunwald-Giemsa method and by immunophenotyping using the following panel of commercially available monoclonal antibodies: anti CD2, CD3, CD5, CD19, CD22, CD23, CD10, and FMC7. Double labeling with anti-CD5/CD19 was performed and the expression of sIg was studied using rabbit antihuman antibodies against the Ig heavy and light chains as previously reported.31 

Treatment was not homogeneous. Depending on age, stage, performance status, and on the clinical course the following first-line treatments were used: single-agent therapy (chlorambucil) in 10 cases and multiagent chemotherapy using cyclophosphamide, vincristine, and prednisone, with (26 cases) or without (6 cases) an anthracycline.

Clinical records were surveyed for all cases and the parameters outlined in Table 1 were collected.

Table 1.

Clinical and Biological Parameters at Presentation in 42 Patients With MCL and BCL1 Involvement

Median (Range)
Age yr  68 (40-88)  
Hb g/dL 12.5 (7.8-15.0)  
WBC × 109/L  8.5 (1.8-840) 
Platelet × 109/L  184 (10-677)  
LDH U/L  440 (250-1200)  
Albumin g/dL  3.8 (3.1-4.9) 
 No. of cases/total  
Splenomegaly  19/42 (45.2%) 
Male/female  29/13  
LDH abnormal  18/41 (43.9%) 
Advanced stage* 31/42 (73.8%)  
B-symptoms 14/42 (33.3%)  
BM involvement 27/42 (64.3%) 
Leukemic expression 31/42 (73.8%)  
Performance status (ECOG) 
 0-1  36/42 (85.7%)  
 2-4 6/42 (14.3%)  
HISTOLOGY 
 Diffuse/nodular1-153 26/2  
 Classical/blastoid  39/3 
Median (Range)
Age yr  68 (40-88)  
Hb g/dL 12.5 (7.8-15.0)  
WBC × 109/L  8.5 (1.8-840) 
Platelet × 109/L  184 (10-677)  
LDH U/L  440 (250-1200)  
Albumin g/dL  3.8 (3.1-4.9) 
 No. of cases/total  
Splenomegaly  19/42 (45.2%) 
Male/female  29/13  
LDH abnormal  18/41 (43.9%) 
Advanced stage* 31/42 (73.8%)  
B-symptoms 14/42 (33.3%)  
BM involvement 27/42 (64.3%) 
Leukemic expression 31/42 (73.8%)  
Performance status (ECOG) 
 0-1  36/42 (85.7%)  
 2-4 6/42 (14.3%)  
HISTOLOGY 
 Diffuse/nodular1-153 26/2  
 Classical/blastoid  39/3 
*

Stage III or IV.

Assessed by bone biopsy in 34 cases and by BM aspirate in 8 cases.

Positive cases defined by >10% CD5/CD19-positive cells in the lymphocyte gate.

F1-153

The histologic architecture of lymphoma growth was assessed on lymph node specimens and not on bone biopsy sections.

CCA.

Cytogenetic investigations were performed on lymph-node and/or PB samples obtained within 3 months of diagnosis in all patients. Single-cell suspensions were prepared, as previously described,32 after collection of a portion of surgically removed lymph node (28 cases, 6 of which were also studied on PB samples), and on PB mononuclear cells obtained by separation over a 1,077 mg/mL density gradient in 14 cases with prominent leukemic involvement. PB and lymph-node cell suspensions containing greater than 90% CD5/CD19+ lymphocytes were cultured for 24 to 72 hours, with and without the following mitogens: phorbol miristate acetate (50 ng/mL), lipopolysaccaride from Escherichia coli (100 mg/mL), and phytohemagglutinin M-form (100 mg/mL). Whenever possible, 20 to 30 metaphases were studied and karyotypes described according to the ISCN.33 

Interphase cytogenetics.

Interphase FISH studies were performed using probes detecting BCL1 rearrangements and those chromosome lesions that were found in at least three patients by CCA. These studies were performed on cells taken from the same samples that were used for cytogenetic analysis.

BCL1 involvement was documented as previously reported,34 using the yeast-artificial-chromosome (YAC) probe 214D11, spanning a 390 kb region encompassing the major translocation cluster and the minor translocation clusters of theBCL1 locus at 11q13.35 The presence of three signals in interphase cells (one deriving from the normal allele and two deriving from the split BCL1 allele) was considered indicative of BCL1 involvement. To prevent data misinterpretation due to the presence of trisomy or monosomy 11, a chromosome 11-specific centromeric probe (Oncor, Gaithersburg, MD), or the cosmid cCI11-395, located at 11p15.5, which was obtained from the Japanese Cancer Research Resources Bank,3 were used as control probes. To document the juxtaposition of BCL1/14q sequences, dual-color FISH was performed in those cases without cytogenetic evidence of the t(11;14), using the BCL1 probe and a 14q telomere probe, or the cos a2 IgH constant-region probe (prepared by H. Dohner, Ruprecht-Karls-Universitat, Medizinische Klinik und Poliklinik V, Heidelberg, Germany) because deletions of 13q, 17p, and total or partial trisomy 12 were the most frequent abnormalities in addition to the 11;14 translocation, having been found in at least 3 cases. Probes for the detection of these anomalies in interphase cells were used to increase the sensitivity of our analysis. The 13q14 C21 cosmid, recognizing DNA sequences between the Rb gene and the D13S25 marker,36 was isolated as previously described.34 Simultaneous hybridization with a chromosome 13-telomere probe (Oncor) was performed. The p53.3 cosmid recognizing p53 gene sequences at the 17p13 chromosome band was prepared and distributed by H. Döhner through F. Birg (Institut de Cancérologie e d’Immunologie de Marseille, INSERM 119, Marseille, France) in the context of the Biomed I programme, “E.U. concerted action for cytogenetic diagnosis of hematologic malignancies” (Project leader: A. Hagemeijer, Centre for Human Genetic, K.U.L., Leuven, Belgium). Simultaneous hybridization with a chromosome 17-centromeric probe was performed (Oncor). A commercially available chromosome 12-pericentromeric probe was used for the detection of total/partial trisomy 12 (Oncor).

Hybridization and signal screening.

The hybridization protocol was described in detail in previous studies.3,34 To prevent false-positive results due to inefficient hybridization, signal screening was performed on slides with a high hybridization efficiency, having greater than 80% interphase cells showing two signals with the control probe. The evaluation was performed on a fluorescence microscope (Nikon Italia, Florence, Italy); 200 cells with well-delineated signals were observed and images were captured with a charged-coupled camera device (Genevision, Nikon Italia).

Five control slides obtained from non-neoplastic tissue were tested with each probe. The cut off for the recognition of BCL1involvement and +12 was set at 5% interphase cells with 3 signals, whereas greater than 10% cells with 1 signal were required for the recognition of 13q14 deletion and 17p13 deletion.

Cytogenetic classification.

To analyze the correlation of cytogenetic data and clinicobiologic findings, three cytogenetic groups were identified based on the number of chromosome lesions as detected by CCA and interphase FISH. Group 1, including those BCL1-rearranged patients with a normal karyotype in at least 20 metaphases and those patients with the t(11;14) as the sole change. Group 2, including patients with 1 to 2 aberrations in addition to the t(11;14)/BCL1 rearrangement, and Group 3, defined by those patients with three or more aberrant events in addition to the t(11;14).

Statistical analysis.

Analysis of variance (ANOVA) with Bonferroni correction for multiple comparisons was used in the analysis of continuous variables whereas χ2 test was applied for categorical variables. Patient survival was estimated by the Kaplan-Meier method from the date of diagnosis until death due to any cause or until the last patient follow-up. The survival curves were statistically compared by the log-rank test. Because many statistical tests were undertaken in the evaluation of prognosis, a P value of .02 was used as the criterion for statistical significance. Proportional hazards regression analysis was used to identify the most significant independent prognostic variables on survival. P values less than .05 were considered statistically significant.

Chromosome lesions: CCA and FISH.

All cases had BCL1 involvement in the area covered by the 390kb YAC probe (Fig 1), resulting in the presence of three signals in 41% to 97% of the interphase cells (median value 73%). The t(11;14)(q13;q32) was documented by chromosome banding in a total of 29 cases, 15 of which had additional aberrations, rearrangements of 11q with complex karyotypes were observed in 5 patients and a normal karyotype was observed at diagnosis in 8 patients. Results are detailed in Table 2.

Fig. 1.

BCL1 rearranged leukemic MCL with primary BM involvement. Note the heterogeneity of cell size and morphology with irregular nuclear outline (left). A cell showing juxtaposition (arrowed) of BCL1 (green) to IgH (red) sequences is shown on the right.

Fig. 1.

BCL1 rearranged leukemic MCL with primary BM involvement. Note the heterogeneity of cell size and morphology with irregular nuclear outline (left). A cell showing juxtaposition (arrowed) of BCL1 (green) to IgH (red) sequences is shown on the right.

Close modal
Table 2.

Karyotypes and Interphase Cytogenetic Findings at Diagnosis in 42 Patients With BCL1 Translocation

Case No. Karyotype [No. of metaphases]Sample FISH*
13q− +12 17p−
Group 1  
 19.30  46,XX [20]  PB  —  —  — 
 8.29  46,XX, t(11;14)(q13;q32) [16]  PB  —  — —  
 23.31  46,XX [20]  PB  —  —  — 
 28.32  46,XY [20]  PB  —  —  —  
 31.8 46,XX,t(11;14)(q13;q32) [18] / 46, XX [2]  LN  —  — —  
 35.20  46,XY, t(11;14)(q13;q32) [15]  LN  — —  —  
 39.25  47,XX,t(11;14) (q13;q32) [4] / 46XX [18]  LN  —  —  —  
 33.16  46,XY, t(11;14)(q13;q32) [19]  LN  —  —  —  
 37.23 46,XX,t(11;14)(q13;q32), [20]  LN  —  —  — 
 41.37  46,XY, t(11;14)(q13;q32) [4] / 46,XX [17]  PB —  —  —  
 42.40  46,XX, t(11;14)(q13;q32) [19] PB  —  —  —  
Group 2  
 3.10 46,XY,t(11;14)(q13;q32) [16] / 46,XY[2]  LN  —  — 55%  
 7.19  47XY, +15, t(11;14)(q13;q32) [13]  LN 80%  —  —  
 9.34  46,XX, t(11;14)(q13;q32) [4] / 46, idem, del(13)(q14q21) [2] / 46,XX [12]  PB  80%  — —  
 12.42  46,XY [25]  PB  64%  —  — 
 15.9  46,XY,t(11;14)(q13;q32) [6] / 46,XY [18]  LN 90%  58%  —  
 17.13  46,XY,t(11;14)(q13;q32) [4] / 46,XY [16]  LN + PB  75%  38%  —  
 18.22 46, XY, t(11;14)(q13;q32) [3] / 46, XY, [19]  LN + PB 68%  44%  —  
 20.38  46,XY, t(11;14)(q13;q32) [6]/ 46,XY [18]  PB  78%  28%  —  
 21.26  46, XX, der(14)t(11;14)(q13;q32) [2], 46, XX [17]  LN + PB 70%  —  75%  
 22.27  46,XX [22]  LN  66% —  62%  
 24.41  46,XX[29]  PB  90%  — 45%  
 30.5 46-48,XY, +3,t(11;14)(q13;q32),add(12)(q?) [20]  LN  —  —  —  
 6.18  46,XY [21]  LN 74%  —  —  
 34.17  46-48XY, t(11;14)(q13;q32), +21 + mar [15]  LN  —  —  —  
Group 3  
 1.3 46-49,XY, del(2)(q?), der(3)(q?), +5,+der(11)(q?), +7,+12, del(17)(p1?2) inc [cp18]  LN  —  —  75%  
 2.7 38-45,X,−X,dup(3)(q21q24),t(6;12)(q23;q12) t(11;X?) (q11;p?), del(11)(q1?3q2?1) [cp8]  LN  —  —  65%  
 4.36 46-48,XY, add(1)(p?), del (10)(q?) t(11;14)(q13;q32), del(17)(p12) +2 mar [cp15]  PB  77%  68%  59%  
 5.15 45-46XY, −3,del(3)(p21;p23),t(3;11)(q14;q11), t(4;17)(q21;q21), t(8;17)(q12;p13), t(11;14)(q13;q32), −13 [cp19]  LN + PB 84%  —  —  
 10.35  46,XY, del(1)(p22p34), add(4)(p12), der(13)t(13;?)(q14;?), der(11)t(11;?) (q13;?) inc [10] PB  90%  —  —  
 11.39  47,XY, add(1)(p36), add(2)(p22), add(6)(q23), t(11;14)(q13;q32), del(13)(q12q14)+M[cp15] PB  74%  —  —  
 13.1 44-47,XY,der(4)t(4;12)(q?;q13), t(11;14) (q13;q32), +der(12)ins(12;13)(q13;q?13q?21), +mar [5] / 47, idem, add(17p) [6]  LN + PB  80%  42%  —  
 14.6 47,XY, +der(3)t(3;?)(q?;?),t(11;14)(q13;q32)[6]  LN  75%  55% —  
 16.11  46-75,XY, +3,t(11;14)(q13;q32),+12 [20]  LN 75%  38%  —  
 25.2  46,XY [25]  LN + PB 85%  40%  38%  
 26.4 44-46,XY, del(6)(q2?4), t(11;14)(q13;q32), +der(13)t(3;13) (q12;q34), +der(13)t(12;13)(q12;q34), del(17)(p11) [cp8]  LN  78% 45%  75%  
 27.12  43-46,XY, −3,−7, t(11;14)(q13;q32),+12 [12] /46,XY [4]  LN  64%  55% 88%  
 29.33  46,XX, add(10)(q22), t(11;14)(q13;q32) del(13)(q14q21) [9] / 46, idem, add(1)(p36) [6]  PB  72% 30%  66%  
 32.14  46,XY,t(11;14)(q13;q32) [8] / 45, idem, del(1) (p22;p32),inv(6)(p21;q2?3), −15 [4]  LN  — —  —  
 38.24  46, XY, idic (1)(q10), dup(3)(q25;q27), del(11)(q?) [3] / 47, idem, +14 [14] / 46XY [3]  LN  — —  —  
 36.21  46,XY, t(11;14)(q13;q32) [19] / 44, idem, −8,−9, del(11)(q13), add(13)(p13), add(17)(p13) [3]  LN —  —  —  
 40.28  47, XY,add(10)(p15), add(1)(q?),del(2)(p22), add(11)(q?) [15]  LN  —  — — 
Case No. Karyotype [No. of metaphases]Sample FISH*
13q− +12 17p−
Group 1  
 19.30  46,XX [20]  PB  —  —  — 
 8.29  46,XX, t(11;14)(q13;q32) [16]  PB  —  — —  
 23.31  46,XX [20]  PB  —  —  — 
 28.32  46,XY [20]  PB  —  —  —  
 31.8 46,XX,t(11;14)(q13;q32) [18] / 46, XX [2]  LN  —  — —  
 35.20  46,XY, t(11;14)(q13;q32) [15]  LN  — —  —  
 39.25  47,XX,t(11;14) (q13;q32) [4] / 46XX [18]  LN  —  —  —  
 33.16  46,XY, t(11;14)(q13;q32) [19]  LN  —  —  —  
 37.23 46,XX,t(11;14)(q13;q32), [20]  LN  —  —  — 
 41.37  46,XY, t(11;14)(q13;q32) [4] / 46,XX [17]  PB —  —  —  
 42.40  46,XX, t(11;14)(q13;q32) [19] PB  —  —  —  
Group 2  
 3.10 46,XY,t(11;14)(q13;q32) [16] / 46,XY[2]  LN  —  — 55%  
 7.19  47XY, +15, t(11;14)(q13;q32) [13]  LN 80%  —  —  
 9.34  46,XX, t(11;14)(q13;q32) [4] / 46, idem, del(13)(q14q21) [2] / 46,XX [12]  PB  80%  — —  
 12.42  46,XY [25]  PB  64%  —  — 
 15.9  46,XY,t(11;14)(q13;q32) [6] / 46,XY [18]  LN 90%  58%  —  
 17.13  46,XY,t(11;14)(q13;q32) [4] / 46,XY [16]  LN + PB  75%  38%  —  
 18.22 46, XY, t(11;14)(q13;q32) [3] / 46, XY, [19]  LN + PB 68%  44%  —  
 20.38  46,XY, t(11;14)(q13;q32) [6]/ 46,XY [18]  PB  78%  28%  —  
 21.26  46, XX, der(14)t(11;14)(q13;q32) [2], 46, XX [17]  LN + PB 70%  —  75%  
 22.27  46,XX [22]  LN  66% —  62%  
 24.41  46,XX[29]  PB  90%  — 45%  
 30.5 46-48,XY, +3,t(11;14)(q13;q32),add(12)(q?) [20]  LN  —  —  —  
 6.18  46,XY [21]  LN 74%  —  —  
 34.17  46-48XY, t(11;14)(q13;q32), +21 + mar [15]  LN  —  —  —  
Group 3  
 1.3 46-49,XY, del(2)(q?), der(3)(q?), +5,+der(11)(q?), +7,+12, del(17)(p1?2) inc [cp18]  LN  —  —  75%  
 2.7 38-45,X,−X,dup(3)(q21q24),t(6;12)(q23;q12) t(11;X?) (q11;p?), del(11)(q1?3q2?1) [cp8]  LN  —  —  65%  
 4.36 46-48,XY, add(1)(p?), del (10)(q?) t(11;14)(q13;q32), del(17)(p12) +2 mar [cp15]  PB  77%  68%  59%  
 5.15 45-46XY, −3,del(3)(p21;p23),t(3;11)(q14;q11), t(4;17)(q21;q21), t(8;17)(q12;p13), t(11;14)(q13;q32), −13 [cp19]  LN + PB 84%  —  —  
 10.35  46,XY, del(1)(p22p34), add(4)(p12), der(13)t(13;?)(q14;?), der(11)t(11;?) (q13;?) inc [10] PB  90%  —  —  
 11.39  47,XY, add(1)(p36), add(2)(p22), add(6)(q23), t(11;14)(q13;q32), del(13)(q12q14)+M[cp15] PB  74%  —  —  
 13.1 44-47,XY,der(4)t(4;12)(q?;q13), t(11;14) (q13;q32), +der(12)ins(12;13)(q13;q?13q?21), +mar [5] / 47, idem, add(17p) [6]  LN + PB  80%  42%  —  
 14.6 47,XY, +der(3)t(3;?)(q?;?),t(11;14)(q13;q32)[6]  LN  75%  55% —  
 16.11  46-75,XY, +3,t(11;14)(q13;q32),+12 [20]  LN 75%  38%  —  
 25.2  46,XY [25]  LN + PB 85%  40%  38%  
 26.4 44-46,XY, del(6)(q2?4), t(11;14)(q13;q32), +der(13)t(3;13) (q12;q34), +der(13)t(12;13)(q12;q34), del(17)(p11) [cp8]  LN  78% 45%  75%  
 27.12  43-46,XY, −3,−7, t(11;14)(q13;q32),+12 [12] /46,XY [4]  LN  64%  55% 88%  
 29.33  46,XX, add(10)(q22), t(11;14)(q13;q32) del(13)(q14q21) [9] / 46, idem, add(1)(p36) [6]  PB  72% 30%  66%  
 32.14  46,XY,t(11;14)(q13;q32) [8] / 45, idem, del(1) (p22;p32),inv(6)(p21;q2?3), −15 [4]  LN  — —  —  
 38.24  46, XY, idic (1)(q10), dup(3)(q25;q27), del(11)(q?) [3] / 47, idem, +14 [14] / 46XY [3]  LN  — —  —  
 36.21  46,XY, t(11;14)(q13;q32) [19] / 44, idem, −8,−9, del(11)(q13), add(13)(p13), add(17)(p13) [3]  LN —  —  —  
 40.28  47, XY,add(10)(p15), add(1)(q?),del(2)(p22), add(11)(q?) [15]  LN  —  — — 

Abbreviations: BM, bone marrow, PB, peripheral blood; LN, lymph node.

*

— indicates the absence of the chromosome lesion (% interphase with abnormal hybridization pattern below the cut-off point for positivity); the percentage of interphase nuclei with deletion 13q14 or 17p13 (1 signal) and with +12 (3 signals) is shown in parentheses.

Patients previously published.3 

Additional chromosome changes observed in three or more cases were loss of chromosome material at 13q and 17p- and total or partial trisomy 12. Monosomy 13 and structural 13q14 aberrations were detected by CCA in six cases. FISH documented the presence of 13q14 deletion in these cases and disclosed cytogenetically undetected 13q14 deletion in 16 additional cases (percentage of cells with one signal 64% to 90%). Three patients had total/partial trisomy 12 at banding analysis and nine additional patients were shown to carry extra chromosome 12 material in 28% to 68% of the cells by interphase FISH. Three patients had deletion 17p at metaphase banding analysis; cytogenetically undetected 17p13 deletions in 38% to 88% interphase nuclei were observed in eight additional cases. The frequency of these chromosome lesions in patients with complex karyotype (ie, patients with three or more aberrations in addition to the 11;14 translocation) compared with patients with noncomplex karyotype was as follows: 8 of 14 versus 14 of 28 for 13q14 deletion (P = 0.662); 5 of 14 versus 7 of 28 for +12 (P = .469) and 6 of 14 versus 5 of 28 for 17p- (P = .082).

The composition of cytogenetic groups 1, 2, 3 was the following (see Table 2): using data obtained by CCA, 22 patients had the t(11;14) as the sole change or a normal karyotype in more than 20 metaphases (group 1), 6 patients had one to two additional chromosome changes (group 2) and 14 patients had three or more additional aberrant events (group 3); when considering CCA plus interphase cytogenetic findings, 11 patients were found to have no detectable chromosome lesions in addition to the t(11;14)/BCL1 involvement (group 1), 14 patients had one to two additional aberrations (group 2) and 17 patients had three or more additional aberrations (group 3).

Hematologic and clinical features.

The salient hematologic features at presentation are summarized in Table 1. Primary sites of disease involvement at presentation were the lymph-node system in 28 cases, whereas the BM and PB (with or without splenomegaly) were primarily involved in 14 patients. Globally, extranodal involvement of the BM, spleen, gastrointestinal tract and Waldeyer’s ring occurred at presentation in 66% of the cases.

BM involvement usually consisted of interstitial or intertrabecular infiltrates of small- to medium-sized lymphocytes, with round-to-oval nuclei and nuclear indentations. Leukemic expression was found in 31 of 42 cases, having 10% to 98% CD5/C19+ cells in the lymphocyte gate (absolute lymphocyte count ranging between 1 and 800 × 109/L, median 22.4 × 109/L). The morphology of these cells showed heterogeneity of cell size and irregularities of nuclear outline (Fig 1); some small lymphocytes indistinguishable from CLL cells and prolymphocyte-like cells were also present. All cases with leukemic expression were CD5/CD19+, CD22+, FMC7+, CD23, and CD10 and sIg+ with a bright pattern of expression and surface κ/λ restriction.

Thirteen patients are alive at 12 to 79 months, with a median follow-up of 36 months, whereas 29 died at 2 to 86 months. Median overall survival in this series was 40 and 34 months in the less than 60 and greater than 60 year age groups, respectively.

Survival.

The correlation of clinical outcome and clinicobiologic parameters is summarized in Table 3, showing that male sex and primary lymph-node involvement were predictive of a shorter survival, whereas age, LDH level, advanced stage at presentation, serum albumin level, PB/BM involvement, PS, and splenomegaly were not. Trisomy 12 as detected by FISH was the only single cytogenetic parameter having prognostic significance (Fig 2), however the strongest prognostic indicator of shorter survival in univariate analysis was the degree of karyotype complexity (Table 3 and Fig 3). Except for male sex, which was associated with complex karyotype, the distribution of other salient clinical and hematologic parameters did not show any statistically significant correlation with cytogenetic features, as summarized in Table 4. Few patients presented blastic morphology or a nodular pattern of growth in lymph-node specimens (Table 1), precluding a meaningful analysis of the correlation of these histologic parameters with cytogenetic patterns.

Table 3.

Outcome According to Clinical and Biological Parameters in 42 Patients With t(11;14): Uniparameter Analysis

Variable No. Patients Median Survival (Months)95% Confidence Interval P
Age yr 
 ≤60  13  34  24-44  .92  
 >60  29  40 29-52  
LDH  
 Normal  23  43  33-54  .069 
 Increased  18  29  17-40  
Sex  
 f  13  61 49-73  .006  
 m  29  31  21-42  
Splenomegaly 
 yes  19  33  21-45  .184  
 no  23  47 33-61  
PB involvement  
 no  11  33  20-45  .71 
 yes  31  47  30-52  
BM involvement  
 yes  27 47  34-59  .026  
 no  15  25  17-32 
Primary site of involvement  
 lymph node  28 28  21-36  .015  
 BM ± spleen  14  55  40-71 
Stage  
 advanced  31  34  25-43  .107 
 initial  11  54  33-75  
PS (ECOG)  
 0-1  36 42  28-56  .121  
 2-4  6  19  7-31  
Serum albumin  
 <4.0  29  43  30-55  .45  
 ≥4.0  13 34  21-47  
+12 by FISH  
 yes  12  22  13-30 .006  
 no  30  47  36-59  
17p by FISH  
 yes 11  28  18-39  .073  
 no  31  45  33-57 
13q by FISH  
 yes  22  30  22-38  .053  
 no 20  48  33-64  
CCA  
 1  21  64  50-78  
 2 7  24  18-30  .0001  
 3  14  12  8-16 
CCA + FISH  
 1  11  86* Not reached at 86 months  
 2  14 48  36-60  <.0001  
 3  17  15  9-21 
Variable No. Patients Median Survival (Months)95% Confidence Interval P
Age yr 
 ≤60  13  34  24-44  .92  
 >60  29  40 29-52  
LDH  
 Normal  23  43  33-54  .069 
 Increased  18  29  17-40  
Sex  
 f  13  61 49-73  .006  
 m  29  31  21-42  
Splenomegaly 
 yes  19  33  21-45  .184  
 no  23  47 33-61  
PB involvement  
 no  11  33  20-45  .71 
 yes  31  47  30-52  
BM involvement  
 yes  27 47  34-59  .026  
 no  15  25  17-32 
Primary site of involvement  
 lymph node  28 28  21-36  .015  
 BM ± spleen  14  55  40-71 
Stage  
 advanced  31  34  25-43  .107 
 initial  11  54  33-75  
PS (ECOG)  
 0-1  36 42  28-56  .121  
 2-4  6  19  7-31  
Serum albumin  
 <4.0  29  43  30-55  .45  
 ≥4.0  13 34  21-47  
+12 by FISH  
 yes  12  22  13-30 .006  
 no  30  47  36-59  
17p by FISH  
 yes 11  28  18-39  .073  
 no  31  45  33-57 
13q by FISH  
 yes  22  30  22-38  .053  
 no 20  48  33-64  
CCA  
 1  21  64  50-78  
 2 7  24  18-30  .0001  
 3  14  12  8-16 
CCA + FISH  
 1  11  86* Not reached at 86 months  
 2  14 48  36-60  <.0001  
 3  17  15  9-21 

Abbreviations: CCA, patients subdivided in group 1, 2, 3 based solely on conventional cytogenetics; CCA + FISH, group 1, 2, and 3 compiled using metaphase banding analysis and interphase cytogenetic data.

Fig. 2.

Survival according to the presence (n = 12) or absence (n = 30) of total/partial trisomy 12 (P .006).

Fig. 2.

Survival according to the presence (n = 12) or absence (n = 30) of total/partial trisomy 12 (P .006).

Close modal
Fig. 3.

Survival according to the degree of karyotype complexity as defined by CCA + FISH analysis (group 1 = 11 patients; group 2 = 14 patients; group 3 = 17 patients) (P < .0001).

Fig. 3.

Survival according to the degree of karyotype complexity as defined by CCA + FISH analysis (group 1 = 11 patients; group 2 = 14 patients; group 3 = 17 patients) (P < .0001).

Close modal
Table 4.

Clinicopathologic Findings in Correlation With Cytogenetics in 42 MCL

+12 13q−17p− Cytogenetic Groups
Yes n = 12 No n = 30 Yes n = 22No n = 20 Yes n = 11 No n = 31 1 n = 11 2 n = 14 3 n = 17
Male  11  18 17  12  6  23  4  10  15  
Female  1  12 5  8  5  8  7  4  
 P = .045  P = .227  P = .226  P = .015  
Stage  
 Advanced  10 21  17  14  9  22  6  11  14  
 Initial 2  9  5  6  2  9  5  3  
 P = .375  P = .592  P = .482  P = .232  
PS 0-1  11  25  19 17  9  27  11  12  13  
PS 2-4  1  5  3  2  4  0  2  
 P = .486  P = .90  P = .667  P = .221  
Splenomegaly  
 Yes  13  13  6  6  13  3  8  8  
 No  17  9  14  5  18  8  6  
 P = .695  P = .059  P = .470  P = .324  
Albumin  
 <4.0 gr/dl  23  13  16  6  23  7  11  11  
 >4.0 gr/dl  5  7  9  4  5  8  4  3  
 P = .091  P = .143  P = .226  P = .639  
BM involved  6  21  16 11  5  22  8  11  8  
BM not involved  6  6  9  6  9  3  3  
 P = .222  P = .231  P = .129  P = .151  
PB involved  9  22  17 14  7  24  8  9  14  
PB not involved  3  5  6  4  7  3  5  
 P = .912  P = .592  P = .372  P = .521  
Age <60  3  10  5  4  9  2  4  7  
Age >60  9  20  14 15  7  22  9  10  10 
 P = .598  P = .426  P = .426  P = .426  
LDH normal  6  17  11 12  4  19  8  8  7  
LDH elevated  6  12 11  7  7  11  2  6  10 
 P = .613  P = .397  P = .123  P = .145  
Extranodal disease  
 No 3  11  8  6  3  11  6  4  4  
 Yes  19  14  14  8  20  5  10  13 
 P = .469  P = .662  P = .142  P = .212  
+12 13q−17p− Cytogenetic Groups
Yes n = 12 No n = 30 Yes n = 22No n = 20 Yes n = 11 No n = 31 1 n = 11 2 n = 14 3 n = 17
Male  11  18 17  12  6  23  4  10  15  
Female  1  12 5  8  5  8  7  4  
 P = .045  P = .227  P = .226  P = .015  
Stage  
 Advanced  10 21  17  14  9  22  6  11  14  
 Initial 2  9  5  6  2  9  5  3  
 P = .375  P = .592  P = .482  P = .232  
PS 0-1  11  25  19 17  9  27  11  12  13  
PS 2-4  1  5  3  2  4  0  2  
 P = .486  P = .90  P = .667  P = .221  
Splenomegaly  
 Yes  13  13  6  6  13  3  8  8  
 No  17  9  14  5  18  8  6  
 P = .695  P = .059  P = .470  P = .324  
Albumin  
 <4.0 gr/dl  23  13  16  6  23  7  11  11  
 >4.0 gr/dl  5  7  9  4  5  8  4  3  
 P = .091  P = .143  P = .226  P = .639  
BM involved  6  21  16 11  5  22  8  11  8  
BM not involved  6  6  9  6  9  3  3  
 P = .222  P = .231  P = .129  P = .151  
PB involved  9  22  17 14  7  24  8  9  14  
PB not involved  3  5  6  4  7  3  5  
 P = .912  P = .592  P = .372  P = .521  
Age <60  3  10  5  4  9  2  4  7  
Age >60  9  20  14 15  7  22  9  10  10 
 P = .598  P = .426  P = .426  P = .426  
LDH normal  6  17  11 12  4  19  8  8  7  
LDH elevated  6  12 11  7  7  11  2  6  10 
 P = .613  P = .397  P = .123  P = .145  
Extranodal disease  
 No 3  11  8  6  3  11  6  4  4  
 Yes  19  14  14  8  20  5  10  13 
 P = .469  P = .662  P = .142  P = .212  

The Cox regression model for survival showed that among the above-reported variables having prognostic significance in univariate analysis, only the measure of karyotype complexity as assessed by FISH and CCA maintained prognostic significance, with a P < .0001 (4,7805 hazard ratio; 2.2884 to 9.9874, 95% confidence limits).

A preliminary methodological problem in this study was represented by the definition of the inclusion criteria, given the heterogeneity of clinicopathological manifestations of MCL.9,12,21,37 Because lymphoma of follicle mantle lineage have in common a specific genetic marker and the immunophenotypic profile,9 we included patients with the t(11;14)/BCL1 rearrangement and with cytoimmunologic features characteristic of MCL. These biological markers of MCL were particularly important in distinguishing those cases presenting with primary BM and PB involvement from other leukemic NHL and chronic B-cell disorders, which may present similar morphologic features. Inclusion criteria in this series accounted on the one hand for the absence of cases with primary extranodal disease, all biopsy material from extranodal sites having been sent to the pathologist for histologic diagnosis and, on the other hand, for the relatively high number of cases with primary BM involvement and leukemic expression. Other hematologic features in our patients (Tables 1 and 3) did not differ significantly as compared with those reported in recent studies,12,13,15,16 with frequent presentation in advanced stage (74% of the cases, compared with 77% to 91%), male sex preponderance (2:1 compared with 1.6:1 up to 3:1), relatively old age (68 years, compared with 62 to 65 years) and short survival (34 and 40 months in the greater than 60 and less than 60 year age groups, compared with 43 to 56 months median overall survival). The histologic features, with a majority of cases presenting diffuse infiltration pattern and few cases displaying a predominantly blastic morphology, are in line with previous observations.12 

The overall cytogenetic picture and the chromosome lesions that were found in this series in addition to BCL1involvement/t(11;14)(q13;q32) improve our knowledge on the clinicobiologic significance of cytogenetics in MCL, showing that total/partial trisomy 12 and a derived measure of karyotype complexity may have a correlation with survival. 13q14 deletion, total or partial trisomy 12, and 17p13 deletion occurred at a relatively high frequency in this series and were not simply a function of karyotype complexity, suggesting that these anomalies are acquired early during the cytogenetic evolution of MCL.

We found a relatively high incidence of normal karyotype (19%) and isolated t(11;14) (33%) in newly diagnosed BCL1+MCL. These findings add to the data by Pittaluga et al29who described a 55% and 8% incidence of normal karyotype and t(11;14) as the sole change, respectively, in 38 cases and with the data collected by Johansson et al,38 who presented a 19.8% incidence of isolated t(11;14) in 91 cases published in the literature. FISH studies,39 however, showed that cytogenetic findings in MCL are partially influenced by suboptimal culture conditions and inadequate banding resolution, having three consequences: (1) detection of normal karyotypes due to divisions occurring in residual normal lymphocytes; (2) detection of the primary chromosome change defining the stemline with failure to show additional anomalies present in the sidelines; and (3) impossibility to recognize subtle rearrangements, especially small deletions, occurring in the context of abnormal karyotypes. Point 1 is illustrated in our study by eight BCL1rearranged cases with apparently normal karyotype. Though representing a laboratory artifact, the finding of a normal karyotype in low-grade lymphomas is clinically important, in that it reflects low mitotic activity of the neoplastic clone, a feature that was associated with a favorable prognosis in B-CLL40 and in follicle center cell lymphomas in which the percentage of normal metaphases in lymph-node samples was associated with a more favorable outcome.20,24Our finding that patients with normal cytogenetics or with t(11;14)/BCL1 rearrangement as the only detectable change (group 1) fared better than patients with additional chromosome changes represents, to the best of our knowledge, the first demonstration that MCL may benefit of cytogenetic investigations for a more accurate assessment of prognosis.

Point 2 is better illustrated in our series by those patients having a t(11;14) as the sole aberration by CCA, who were shown to carry extra chromosome 12 material in 26% to 58% of the interphase cells (cases 15.9; 17.13; 18.22; 20.38; 29.33). The possibility that in some patients with complex karyotype (cases 13.1; 14.6; 26.4), partial trisomy 12 was not recognized at karyotyping due to the presence of marker chromosomes, should be considered. It is noteworthy that in B-CLL, CCA was reported to underestimate the incidence of +12 compared with interphase cytogenetics, both in cases with normal karyotypes41 and with complex rearrangements.42,43 The acquisition of extra chromosome 12 material was detected much less frequently in follicle center cell lymphoma and in marginal zone B-cell lymphoma in three studies using comparative genomic hybridization,44-46 suggesting that this chromosome imbalance may be preferentially associated with CD5+ B-cell lymphoproliferative disorders. These considerations have practical implications, because total/partial trisomy 12 was the only single cytogenetic parameter that was predictive of an adverse outcome in this series, and point at the importance of FISH for the refinement of cytogenetic diagnosis in MCL.

Point 3 is illustrated by those cases having cytogenetically undetected 13q14 and 17p13 deletions. Our findings are reassuring with respect to the specificity of metaphase banding analysis and show that the sensitivity of CCA in detecting loss of chromosome material is unsatisfactory.

The high incidence of 13q14 deletions (52.3%) that was found in this series is a relatively new finding, this anomaly having been associated with 40% of B-CLL studied by molecular cytogenetics.36,43,47 Interestingly, 13q14 deletion was found to occur by FISH at a relatively high incidence in atypical CLL carrying the 11;14 translocation34 and in a recent study on MCL.48 Because a 8.8% incidence of 13q14 deletion was found at our Institution in 91 samples of B-NHL excluding MCL and small lymphocytic lymphoma (data not shown), we suggest that loss of genetic material involving the 13q14 region may represent an important step in the transformation of CD5+ B-cell neoplasias.

Whereas the 13q-anomaly carries a favorable prognostic significance in B-CLL, no statistically significant survival difference was noted in our patients with and without 13q14 deletion. A trend towards a shorter survival was observed instead in 13q-patients, partially accounted for by the presence of additional aberrations in half of the cases. It is noteworthy that 13q14 deletions are found, albeit at a lower frequency, in a spectrum of lymphoid neoplasias,49 including multiple myeloma, in which they have been associated with an inferior prognosis.50,51 

The 26.2% incidence of 17p13 deletion in this series is comparable with the 18% incidence that was reported by Clodi et al,52who analyzed by interphase FISH 79 unselected NHL using a 17p13/p53 probe. No correlation was found in this study and in the analysis by Clodi et al52 between deletion involving this region and survival; hence it is reasonable to assume that p53 gene deletion per se does not have a major impact on prognosis in NHL. As for the case with 13q14 deletions, the possibility should be considered that more cases need to be studied to allow a single cytogenetic parameter to reach statistical significance on survival analysis, even more so that a number of chromosome changes was frequently found in association with each of these anomalies.

This consideration prompted us to define a measure of karyotype complexity to compare survival among different patient groups. Derived measures of karyotype complexity, ie, modal chromosome number, number of translocation breakpoints, and number of marker chromosomes were shown to have prognostic significance in low-grade NHL and in follicle center cell lymphomas.20,26 The difference in survival according to the number of chromosome lesions that was observed in our study was evident using data obtained by CCA and the observed difference had an optimal statistical significance using CCA plus FISH data. This classification clearly depends on the probes that we chose to test in interphase cells, based on the identification of the most frequent additional changes by CCA. Other chromosome regions that may potentially have prognostic significance20,26 were shown to be deleted or rearranged in a fraction of MCL,38 the most frequent being 6q15-21, 6q25, 1p21-32, and 1q21-23. Attention was devoted in a recent study on a cycline-dependent kinase (CDK) inhibitor, referred to as p16INK4a and located on chromosome 9p21, the deletion of which was shown to be associated with a high proliferation index.53 Interestingly, the gene encoding for a closely related CDK inhibitor, p18INK4c, was mapped to 1p31-35,54 in a chromosome segment that was deleted in two of our cases. In general, it is reasonable to predict that, as more cases will be studied, new recurring chromosome breaks will be recognized that will allow for the definition of a wider panel of FISH probes, possibly resulting in the identification of other clinically important genomic imbalances.

Supported by BMH-1 EU CA: CT 94-1703, and by C.N.R., ACRO project and M.U.R.S.T, fondi 40% and 60%.

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

1
Vandenberghe
E
De Wolf-Peeters
C
Van Den Oord
J
Wlodar ska
I
Delabie
J
Stul
M
Thomas
J
Michaux
JL
Mecuc ci
C
Cassiman
JJ
Vandenberghe
H
Translocation (11;14): A cytogenetic anomaly associated with B cell lymphomas of non-follicle centre cell lineage.
J Pathol
163
1991
13
2
Vaandrager
JW
Schuuring
E
Zwikstra
E
de Boer
CJ
Kleiverda
KK
van Krieken
JHJM
Kluin-Nelemans
HC
van Ommen
GJB
Raap
AK
Kluin
PM
Direct visualization of dispersed 11q13 chromosomal traslocations in mantle cell lymphoma by multi-color DNA fiber FISH.
Blood
88
1996
1177
3
Bigoni
R
Negrini
M
Veronese
ML
Cuneo
A
Castoldi
G
Croce
CM
Characterization of t(11;14) translocation in mantle cell lymphoma by fluorescent in situ hybridization.
Oncogene
13
1996
797
4
Siebert
R
Matthiesen
P
Harder
S
Zhang
Y
Borowski
A
Zuhlke-Jenisch
R
Plendl
H
Metzke
S
Joos
S
Zucca
E
Weber-Matthiesen
K
Roggero
E
Grote
W
Schlegelberger
B
Application of interphase cytogenetics for the detection of t(11;14)(q13;q32) in mantle cell lymphomas.
Annals Oncol
9
1998
519
5
Tsujimoto
Y
Yunis
J
Onorato-Showe
L
Erikson
J
Nowell
PC
Croce
CM
Molecular cloning of the chromosomal breakpoint of B cell lymphomas and leukemias with the t(11;14) chromosome translocation.
Science
224
1984
1043
6
Rosenberg
CL
Wong
E
Petty
EM
Bale
ARE
Tsujimoto
Y
Harris
NL
Arnold
A
PRAD1, a candidate bcl1 oncogene: Mapping and expression in centrocytic lymphoma.
Proc Natl Acad Sci USA
88
1991
9638
7
Rimoch
R
Berger
F
Delsol
G
Charrin
C
Bertheas
MF
Ffrench
M
Garoscio
M
Felman
P
Coiffier
B
Bryon
PA
Rochet
M
Gentilhomme
O
Germain
D
Magaud
JP
Rearrangement and overexpression of the BCL-1/PRAD-1 gene in intermediate lymphocytic lymphomas and in t(11q13)-bearing leukemias.
Blood
81
1993
3063
8
Weisenburger
DD
Kim
H
Rappaport
H
Mantle-zone lymphoma. A follicular variant of intermediate lymphocytic lymphoma.
Cancer
49
1982
1429
9
Weisenburger
DD
Armitage
JO
Mantle Cell Lymphoma—An Entity Comes of Age.
Blood
87
1996
4483
10
Pombo De Oliveira
MS
Jaffe
ES
Catovsky
D
Leukaemic phase of mantle zone (intermediate) lymphoma. Its characterisation in 11 cases.
J Clin Pathol
42
1989
962
11
Raffeld
M
Jaffe
ES
Bcl-1, t(11;14), and mantle cell derived lymphomas.
Blood
78
1991
259
12
Argatoff
LH
Connors
JM
Klasa
RJ
Horsman
DE
Gascoyne
RD
Mantle cell lymphoma: A clinicopathologic study of 80 cases.
Blood
89
1996
2067
13
Vandenberghe
E
De Wolf-Peeters
C
Vaughan Hudson
G
Vaughan Hudson
B
Pittaluga
S
Anderson
L
Linch
DC
The clinical outcome of 65 cases of mantle cell lymphoma initially treated with non-intensive therapy by the British National Lymphoma Investigation Group.
Br J Haematol
99
1997
842
14
Zucca
E
Stein
H
Coiffier
B
European Lymphoma Task Force (ELTF) report of the Workshop on mantle cell lymphoma (MCL).
Ann Oncol
5
1994
507
15
Bertini
M
Rus
C
Freilone
R
Botto
B
Calvi
R
Novero
D
Orsucci
L
Vitolo
U
Palestro
G
Resegotti
L
Mantle cell lymphoma: a retrospective study on 27 patients. Clinical features and natural history.
Haematologica
83
1998
312
16
Bosch
F
Lòpez-Guillermo
A
Campo
E
Ribera
JM
Conde
E
Piris
MA
Vallespı̀
T
Woessner
S
Montserrat
E
Mantle cell lymphoma. Presenting features, response to therapy, and prognostic factors.
Cancer
82
1998
567
17
Norton
AJ
Matthews
J
Pappa
V
Shamash
J
Rohatiner
AZS
Lister
TA
Mantle cell lymphoma: Natural history defined in a serially biopsied population over 20-year period.
Ann Oncol
6
1995
249
18
Yunis
JJ
Frizzera
G
Oken
MM
McKenna
J
Theologides
A
Arnesen
M
Multiple recurrent genomic defects in follicular lymphoma: A possible model for cancer.
N Engl J Med
316
1987
79
19
Offitt
K
Lo Coco
F
Louie
DC
Parsa
NZ
Leung
D
Portlock
C
Ye
BH
Lista
F
Filippa
DA
Rosenbaum
A
Landanyi
M
Jhanwar
S
Dalla-Favera
R
Chaganti
RSK
Rearrangement of BCL-6 gene as a prognostic marker in diffuse large-cell lymphoma.
N Engl J Med
331
1994
74
20
Tilly
H
Rossi
A
Stamatoullas
A
Lenormand
B
Bigorgne
C
Kunlin
A
Monconduit
M
Bastard
C
Prognostic value of chromosomal abnormalities in follicular lymphoma.
Blood
84
1994
1043
21
Louie
DC
Offit
K
Jaslow
R
Parsa
NZ
Murty
VVVS
Schluger
A
Chaganti
RSK
p53 overexpression as a marker of poor prognosis in mantle cell lymphoma with t(11;14)(q13;q32).
Blood
86
1995
2892
22
Greiner
TC
Moynihan
MJ
Chan
WC
Lytle
DM
Pedersen
A
Anderson
JR
Weisenburger
DD
p53 mutations in mantle cell lymphoma are associated with variant cytology and predict a poor prognosis.
Blood
87
1996
4302
23
Döhner
H
Fisher
K
Bentz
M
Hansen
K
Benner
A
Cabot
G
Diehl
D
Schlenk
R
Coy
J
Stilgenbauer
S
Volkmann
M
Galle
PR
Poustka
A
Hustein
W
Lichter
P
p53 gene deletion predicts for poor survival and non-response to therapy with purine analogs in chronic B-cell leukemias.
Blood
85
1995
1580
24
Cabanillas
F
Pathak
S
Grant
G
Hagermeister
FB
McLaughlin
P
Swan
F
Rodriguez
MA
Trujillo
J
Cork
A
Butler
JJ
Katz
R
Bourne
S
Freireich
EJ
Refractoriness to chemotherapy and poor survival related to abnormalities of chromosomes 17 and 7 in lymphoma.
Am J Med
87
1989
167
25
Levine
EG
Arthur
DC
Frizzera
G
Peterson
BA
Hurd
DD
Bloomfield
CD
Cytogenetic abnormalities predict clinical outcome in non-Hodgkin’s lymphoma.
Ann Intern Med
108
1988
14
26
Offitt
K
Wong
G
Philippa
DA
Tao
Y
Chaganti
RSK
Cytogenetic analysis of 434 consecutively ascertained specimens of non-Hodgkin’s lymphoma: Clinical correlations.
Blood
77
1991
1508
27
Döhner
H
Stilgenbauer
S
James
MR
Benner
A
Weilguni
T
Bentz
M
Fischer
K
Hunstein
W
Lichter
P
11q deletions define a new subset of B-cell chronic lymphocytic leukemia characterized by extensive nodal involvement and inferior prognosis.
Blood
89
1997
2516
28
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
Muller-Hermelink
HK
Pileri
SA
Piris
MA
Ralfkiaer
E
Warnke
RA
A revised European-American classification of lymphoid neoplasms: A proposal from the international lymphoma study group.
Blood
84
1994
1361
29
Pittaluga
S
Wlodarska
I
Stul
MS
Thomas
J
Verhoef
G
Cassiman
JJ
Van Den Berghe
H
De Wolf-Peeters
C
Mantle cell lymphoma: A clinicopathologic study of 55 cases.
Histopathology
26
1995
17
30
Bennett
JM
Catovsky
D
Daniel
MT
Flandrin
G
Galton
DA
Gralnick
HR
Sultan
C
The French-American-British (FAB) Cooperative Group
Proposals for the classification of chronic (mature) B and T lymphoid leukemias.
J Clin Pathol
42
1989
567
31
Cuneo
A
Balboni
M
Piva
N
Rigolin
GM
Roberti
MG
Mejak
C
Moretti
S
Bigoni
R
Balsamo
R
Cavazzini
PL
Castoldi
GL
Atypical chronic lymphocytic leukaemia with the t(11;14)(q13;q32): Karyotype evolution and prolymphocy tic tranformation.
Br J Haematol
90
1995
409
32
Leroux
D
Le Marc’ hardour
F
Gressin
R
Jacob
MC
Keddari
E
Monteil
M
Caillot
P
Jalbert
P
Sotto
JJ
Non Hodgkin’s lymphomas with t(11;14)(q13;q32): A subset of mantle zone/intermediate lymphocytic limphoma.
Br J Haematol
77
1991
346
33
Mitelman
F
ISCN (1991). Guidelines for cancer cytogenetics. Supplement to an international system for human cytogenetic nomenclature.
1995
Karger
Basel, Switzerland
34
Cuneo
A
Bigoni
R
Negrini
M
Bullrich
F
Veronese
ML
Roberti
MG
Bardi
A
Rigolin
GM
Cavazzini
PL
Croce
CM
Castoldi
GL
Cytogenetic and interphase cytogenetic characterization of atypical chronic lymphocytic leukemia carryng BCL1 translocation.
Cancer Res
57
1997
1144
35
Szepetowsky
P
Perucca-Lostanlen
D
Grosgeorge
J
LePaslier
D
Brownstein
BH
Carle
GF
Gaudray
P
Description of a 700-kb yeast artificial chromosome contig containing the bcl-1 translocation breakpoint region at 11q13.
Cytogenet Cell Genet
69
1995
101
36
Bullrich
F
Veronese
ML
Kitada
S
Jurlander
J
Caligiuri
MA
Reed
J
Croce
CM
Minimal region of loss at 13q14 in B-CLL.
Blood
88
1996
3109
37
Hiddemann W: Mantle cell lymphoma. Educational session, Congress of the ISH/EHA, Amsterdam 4-9 July, 1998, pp 125-128.
38
Johansson
B
Mertens
F
Mitelman
F
Cytogenetic evolution patterns in non-Hodgkin’s lymphoma.
Blood
86
1995
3905
39
Coignet
LJA
Schuuring
E
Kibbelaar
RE
Raap
TK
Kleiverda
KK
Berthes
MF
Wiegant
J
Beverstock
G
Kluin
P
Detection of 11q13 rearrangements in hematologic neoplasias by double-color fluorescence in situ hybridization.
Blood
87
1996
1512
40
Juliusson
G
Oscier
DG
Fitchett
M
Ross
FM
Stockdill
G
Mackie
MJ
Parker
A
Castoldi
GL
Cuneo
A
Knuutila
S
Elonen
E
Gahrton
G
Prognostic subgroups in B-cell chronic lymphocytic leukemia defined by specific chromosomal abnormalities.
N Engl J Med
323
1990
720
41
Cuneo
A
Wlodarska
I
Sayed Aly
M
Piva
N
Carli
MG
Fagioli
F
Tallarico
A
Pazzi
I
Ferrari
L
Cassiman
JJ
Van Den Berghe
H
Castoldi
GL
Nonradioactive in situ hybridization for the detection and monitoring of trisomy 12 in B cell chronic lymphocytic leukaemia.
Br J Haematol
81
1992
192
42
Que
TH
Marco
JG
Ellis
J
Matutes
E
Babapulle
VB
Boyle
S
Catovsky
D
Trisomy 12 in chronic lymphocytic leukemia detected by fluorescence in situ hybridization: Analysis by stage, immunophenotype, and morphology.
Blood
82
1993
571
43
Bigoni
R
Cuneo
A
Roberti
MG
Bardi
A
Rigolin
GM
Piva
N
Scapoli
G
Spanedda
R
Negrini
M
Bullrich
F
Veronese
ML
Croce
CM
Castoldi
G
Chromosome aberrations in chronic lymphocytic leukemia mixed cell type. A cytogenetic and interphase cytogenetic study.
Leukemia
11
1997
1933
44
Bentz
M
Werner
CA
Döhner
H
Joos
S
Barth
TFE
Siebert
R
Schroder
M
Stilgenbauer
S
Fischer
K
Moller
P
Lichter
P
High incidence of chromosomal imbalances and gene amplifications in the classical follicular variant of follicle center lymphoma.
Blood
88
1996
1437
45
Avet-Loiseau
H
Vigier
M
Moreau
A
Mellerin
MP
Gaillard
F
Harousseau
JL
Bataille
R
Milpied
N
Comparative genomic hybridization detects genomic abnormalities in 80% of follicular lymphomas.
Br J Haematol
97
1997
119
46
Dierlamm
J
Rosenberg
C
Stul
M
Pittaluga
S
Wlodarska
I
Michaux
L
Dehaen
M
Verhoef
G
Thomas
J
de Kelver
W
Bakker-Shut
T
Cassiman
JJ
Raap
AK
De Wolf-Peeters
C
Van den Berghe
H
Hagemeijer
A
Characteristic pattern of chromosomal gains and losses in marginal zone B cell lymphoma detected by comparative genomic hybridization.
Leukemia
11
1997
747
47
Corcoran
MM
Rasool
O
Liu
Y
Iyengar
A
Grander
D
Ibbotson
RE
Merup
M
Wu
X
Brodyansky
V
Gardiner
AC
Juliusson
G
Chapman
RM
Ivanova
G
Tiller
M
Gahrton
G
Yankovsky
N
Zabarovsky
E
Oscier
DG
Eihorn
S
Detailed molecular delineation of 13q14.3 loss in B-cell chronic lymphocytic leukemia.
Blood
91
1998
1382
48
Stilgenbauer
S
Nickolenko
J
Wilhelm
J
Wolf
S
Weitz
S
Fisher
K
Boehm
T
Döhner
H
Lichter
P
Expressed sequences as candidates for a novel tumor suppressor gene at abnd 13q14 in B-cell chronic lymphocytic leukemia and mantle cell lymphoma.
Oncogene
16
1998
1891
49
Liu
Y
Hermanson
M
Grander
D
Merup
M
Wu
X
Heyman
M
Rasool
O
Juliusson
G
Gahrton
G
Detlofsson
R
Nikiforova
N
Buys
C
Soderhall
S
Yankovsky
N
Zabarovsky
E
Einhorn
S
13q deletions in lymphoid malignancies.
Blood
86
1995
1911
50
Tricot
G
Barlogie
B
Jagannath
S
Bracy
D
Mattox
S
Vesole
D
Naucke
S
Sawyer
J
Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving 11q and not with other karyotype abnormalities.
Blood
86
1995
4250
51
Seong
C
Delasalle
K
Hayes
K
Weber
D
Dimopoulos
M
Swantkowski
J
Huh
Y
Glassman
A
Champlin
R
Alexanian
R
Prognostic value of cytogenetics in multiple myeloma.
Br J Haematol
101
1998
189
52
Clodi
K
Younes
A
Goodacre
A
Roberts
M
Palmer
J
Younes
M
Cabanillas
F
Andeeff
M
Analysis of p53 gene deletions in patients with non-Hodgkin’s lymphoma by dual-colour fluorescence in-situ hybridation.
Br J Haematol
98
1997
913
53
Dreyling
MH
Bullinger
L
Ott
G
Stilgenbauer
S
Muller-Hermelink
Hk
Bentz
M
Hiddemann
W
Dohner
H
Alterations of the cyclin D1/p16-pRB pathway in mantle cell lymphoma.
Cancer Res
57
1997
4608
54
Tahara
H
Smith
AP
Gaz
AD
Zariwala
M
Xiong
Y
Arnold
A
Parathyroid tumor suppressor on 1p: analysis of the p18 cyclin-dependent kinase inhibitor gene as a candidate.
J Bone Miner Res
12
1997
1330
Sign in via your Institution