Limited knowledge exists about the impact of specific genetic abnormalities on the proliferation of neoplastic B cells from chronic lymphoproliferative disorders (B-CLPDs). Here we analyze the impact of cytogenetic abnormalities on the proliferation of neoplastic B cells in 432 B-CLPD patients, grouped according to diagnosis and site of sampling, versus their normal counterparts. Overall, proliferation of neoplastic B cells highly varied among the different B-CLPD subtypes, the greatest numbers of proliferating cells being identified in diffuse large B-cell lymphoma (DLBCL) and Burkitt lymphoma (BL). Compared with normal B cells, neoplastic B-CLPD cells showed significantly increased S + G2/M-phase values in mantle cell lymphoma (MCL), B-chronic lymphocytic leukemia (B-CLL), BL, and some DLBCL cases. Conversely, decreased proliferation was observed in follicular lymphoma, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia (LPL/WM), and some DLBCL patients; hairy cell leukemia, splenic marginal zone, and MALT-lymphoma patients showed S + G2/M phase values similar to normal mature B lymphocytes from LN. Interestingly, in B-CLL and MCL significantly higher percentages of S + G2/M cells were detected in BM versus PB and in LN versus BM and PB samples, respectively. In turn, presence of 14q32.3 gene rearrangements and DNA aneuploidy, was associated with a higher percentage of S + G2/M-phase cells among LPL/WM and B-CLL cases, respectively.

Malignant transformation of neoplastic cells, including B lymphocytes, has long been associated with genetic abnormalities involving cell proliferation and death signaling pathways.1-5  Accordingly, high grade B-cell non-Hodgkin lymphomas (NHLs; eg, Burkitt lymphoma) have been considered to have an abnormally increased cell proliferation rate, linked to constitutive activation of proto-oncogenes (eg, C-MYC).6-8  In contrast, an increased cell survival because of a prominent antiapoptotic phenotype (eg, high bcl2 or cyclin D1 expression)9-13  has been associated with low-grade B-NHL and most chronic lymphoid leukemias.1  However, a careful analysis of the literature shows very limited information about the real impact of specific genetic abnormalities associated with different subtypes of both high- and low-grade B-cell chronic lymphoproliferative disorder (B-CLPD) on the proliferation rate of primary tumor cells, in comparison with their normal counterparts

Such comparison could contribute to a better understanding of the real impact of specific genetic abnormalities in the proliferation rate of tumor cells arrested at specific stages of maturation; in turn, this information is also of potential clinical relevance, since measurement of the proliferative activity of neoplastic cells by semiquantitative immunohistochemical techniques (eg, ki67 or PCNA immunostaining),14-19 3H thymidine incorporation,20  and single-parameter flow cytometric analysis of paraffin-embedded tumor samples21-27  is considered to be of great help for the prognostic stratification of B-cell chronic lymphoproliferative disorders (B-CLPDs),21-23,26-33  even within specific histologic and WHO subtypes (eg, mantle cell lymphoma).34 

Despite this, evaluation of tumor cell proliferation has only partially translated into routine clinical practice. This is due to a great variability with respect to numbers of cells analyzed, the use of different sampling (eg, fresh tissue vs paraffin-embedded material), and sample preparation and staining techniques. In addition, in the case of flow cytometry, the coefficients of variation of the G0/G1 peaks and the mathematical models for cell-cycle phase calculations are highly variable21,23,32 ; moreover, assessment of the overall percentage of proliferating cells in the whole sample, and not exclusively among neoplastic B cells, leads to an additional increasing degree of variability.35 

Here, we analyze the potential impact of the most frequent genetic abnormalities on the proliferative rate of neoplastic B cells from a series of 432 B-CLPD patients grouped according to diagnosis and site of sampling in comparison with their normal counterparts. For that purpose, a detailed analysis of the proliferative rate of different compartments of maturation-associated subsets of bone marrow (BM), peripheral blood (PB), and reactive lymph node (RLN) B cells is provided as a frame of reference for the identification of an altered cell-cycle distribution in B-CLPD.

This study was approved by the Ethical Committee of the University Hospital of Salamanca. Informed consent was obtained in accordance with the Declaration of Helsinki.

Patients and samples

A total of 432 untreated patients (260 males and 172 females with a mean age of 66 ± 13 years; range: 19 to 95 years), newly diagnosed with B-CLPD between September 2000 and February 2007, were included in this study. In all cases, diagnosis was established according to the WHO criteria36,37  with the following distribution: B-cell chronic lymphocytic leukemia (B-CLL), 210 patients; hairy cell leukemia (HCL), 7; mantle-cell lymphoma (MCL), 39; splenic marginal zone B-cell lymphoma (SMZL), 16; MALT lymphoma (MALT-NHL), 20; follicular lymphoma (FL), 71; diffuse large B-cell lymphoma (DLCL), 19; Burkitt lymphoma (BL), 14; and lymphoplasmacytic lymphoma/Waldenström macroglobulinemia (LPL/WM), 36. Immunophenotypic analyses, DNA cell content measurements, and interphase fluorescence in situ hybridization (iFISH) studies were performed on EDTA-anticoagulated PB (39%), BM (43%), and fine needle–aspirated (FNA) lymph node (LN; 18%) samples.

Immunophenotypical studies

Immunophenotypic analysis of neoplastic B cells was performed in all 432 B-CLPD cases after staining of erythrocyte-lysed BM and PB as well as LN samples, according to procedures that have been previously described in detail.38  The following fluorocrome-conjugated—fluorescein isothiocyanate (FITC)/phycoerythrin (PE)/peridinin chlorophyll protein-cyanin 5.5 (PerCPCy5.5)/allophycocyanin (APC)—combinations of monoclonal antibodies (MAbs) were used: CD8-sIgλ/CD56-sIgκ/CD4-CD19/CD3, CD22/CD23/CD19/CD20, CD103/CD25/CD19/CD11c, FMC7/CD24/CD19/CD34, cytoplasmatic (Cy) bcl2/CD10/CD19/CD38, surface immunoglobulin (sIg)λ/sIgκ/CD19/CD5, sIgμ/CD27/CD19, and CD43/CD79b/CD19. All MAb reagents were purchased from BD Biosciences (San Jose, CA) except anti-CD79b, CD24, CD10, and CD43, which were obtained from Immunotech (Marseille, France) and antihuman sIgμ, antihuman sIgλ, and antihuman sIgκ, which were purchased from DAKO (Glostrup, Denmark). For the staining of Cybcl2, the Fix & Perm reagent kit (Invitrogen, Burlingame, CA) was used, strictly following the recommendations of the manufacturer. In all cases, one tube stained with CD19-PerCPCy5.5 was used as a control for the specific evaluation of baseline green, orange, and deep red autofluorescence of B cells.

Immediately after sample preparation was completed, data acquisition was performed on a FACSCalibur flow cytometer using the CellQUEST software program (BD Biosciences). For each combination of MAbs, information about 5 × 104 events corresponding to the whole sample cellularity was acquired and stored. In a second step, information on events acquired through an electronic live gate containing only CD19+ cells was specifically stored (median of 201 643 and 95% confidence interval of 169 641 to 233 645 events). For data analysis, the PAINT-A-GATE-PRO software program (BD Biosciences) was used. For each sample, all major subpopulations of lymphocytes were measured including B cells, T cells, and their major CD3+/CD4+ and CD3+/CD8+ subsets and CD56+/CD3 natural killer (NK) cells. CD19 was used for the specific identification of B cells present in all sample aliquots, and their percentage was calculated after excluding cell debris and platelets according to conventional procedures.38  Identification of B-cell precursors versus mature B lymphocytes was performed on the basis of a low to intermediate CD45 expression on CD19+ cells. In 57 B-CLPD cases showing dim CD19 expression, CD20 was used to calculate the percentage of B cells in the sample.

Interphase fluorescence in situ hybridization (iFISH) studies

Analysis of the genetic alterations associated with B-CLPD was systematically performed by multicolor iFISH. For the detection of trisomy 12, del(11q22.3), del(11q23), del(17p13.1), and del(13q14), the following DNA probes purchased from Vysis (Downers Grove, IL) were used in multicolor stainings (Table 1): CEP12, LSI ATM (11q22.3) and LSI MLL (11q23.3) dual-color probe, LSI p53 (17p13.1) and both the LSI 13/RB1 gene (13q14) and LSI D13S25 (13q14.3) DNA probes. In turn, presence of t(14q32), t(18q21), t(14;18), t(11;14), and c-MYC gene rearrangements was assessed using the LSI IgH dual-color, LSI MALT1 dual-color, LSI IgH/Bcl2 dual-color, LSI IgH/CCND1 dual-color and both the LSI MYC dual-color breakapart and LSI IgH/MYC/CEP8 3-color probes, respectively. DNA probes for the detection of BCL6 gene rearrangements (LSI BCL6 dual-color breakapart probe; Vysis) and del(6q21) (6q21 cocktail probe; Q-BIOgene, Amsterdam, The Netherlands) were also used (Table 1).

Table 1

Chromosomal abnormalities detected by iFISH in B-CLPD subtypes

B-CLPD/geneChromosomal abnormalityProbe
B-CLL   
    MDM2?39  trisomy 12 CEP12* 
    RB1/D13S2540  del(13q) LSI 13/RB1 (13q14)/LSI D13S25 (13q14.3)* 
    ATM/MLL40,41  del(11q) LSI ATM (11q22.3)/LSI MLL (11q23.3)* 
    P5340  del(17p) LSI P53 (17p13.1)* 
MCL   
    CYCLIN D142,43  t(11;14) LSI IGH/CCND1* 
MALT   
    MLT144,45  t(18q21) LSI MALT1* 
    IGH t(14q32) LSI IGH* 
FL, BCL243,46  t(14;18) LSI IGH/BCL2* 
DLBCL, BCL647  t(3q27) LSI BCL6* 
BL, C-MYC48  t(8;14) LSI IGH/MYC/CEP8* 
LPL/WM   
    IGH49  t(14q32) LSI IGH* 
    BLIMP150,51  del(6q21) 6q21 cocktail probe 
B-CLPD/geneChromosomal abnormalityProbe
B-CLL   
    MDM2?39  trisomy 12 CEP12* 
    RB1/D13S2540  del(13q) LSI 13/RB1 (13q14)/LSI D13S25 (13q14.3)* 
    ATM/MLL40,41  del(11q) LSI ATM (11q22.3)/LSI MLL (11q23.3)* 
    P5340  del(17p) LSI P53 (17p13.1)* 
MCL   
    CYCLIN D142,43  t(11;14) LSI IGH/CCND1* 
MALT   
    MLT144,45  t(18q21) LSI MALT1* 
    IGH t(14q32) LSI IGH* 
FL, BCL243,46  t(14;18) LSI IGH/BCL2* 
DLBCL, BCL647  t(3q27) LSI BCL6* 
BL, C-MYC48  t(8;14) LSI IGH/MYC/CEP8* 
LPL/WM   
    IGH49  t(14q32) LSI IGH* 
    BLIMP150,51  del(6q21) 6q21 cocktail probe 

B-CLPD indicates B-cell chronic lymphoproliferative disorders; B-CLL, B-cell chronic lymphocytic leukemia; MCL, mantle-cell lymphoma; MALT, mucosa-associated lymphoid tissue lymphoma; FL, follicular lymphoma; DLBCL, diffuse large B-cell lymphoma; BL, Burkitt lymphoma; and LPL/WM, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia.

*

Vysis.

Q-BIOgene.

FISH studies were performed on freshly obtained, fluorescence-activated cell sorting (FACS)–purified neoplastic B cells (purity of 98% ± 0.82%) fixed in 3:1 (vol/vol) methanol–acetic acid, prepared, and hybridized as previously described in detail.52  The number of hybridization spots was evaluated using a BX60 fluorescence microscope (Olympus, Hamburg, Germany) equipped with a 100× oil objective. For each slide, the number of hybridization spots with a similar size, intensity, and shape was counted for at least 200 nuclei, the number of unhybridization cells in the areas assessed being less than 1%. Cutoff values used for the definition of each chromosomal abnormality were established according to the iFISH patterns observed in a group of 10 age- and sex-matched normal control BM samples studied with the same probes (mean percentage + 2 standard deviations [SD]): chromosome 12, more than 1%; del(13q14), more than 4%; del(17p13.1), more than 7%; del(11q22.3), more than 4%; del(11q23.3), more than 1%; t(14;18), more than 1%; c-MYC gene rearrangements, more than 1%; t(11;14), more than 1%; and; del(6q21), more than 6%.

Flow cytometric analysis of B-cell DNA contents

The DNA content of neoplastic B cells was measured in a FACSCalibur flow cytometer (BD Biosciences) using the Cycloscope NHL-B reagent kit (Cytognos, Salamanca, Spain), strictly following the recommendations of the manufacturer. DNA aneuploidy was defined by the observation of a different DNA content for G0/G1 tumor B cells compared with normal residual (T/NK) lymphocytes; DNA index of neoplastic B cells was calculated as the ratio between the modal fluorescence channel of G0/G1 tumor B cells and that of the normal residual G0/G1 diploid T/NK lymphocytes present in the same sample. Analysis of the distribution of B-CLPD cells in the G0/G1 and S + G2/M cell-cycle phases was performed according to well-established methods, after specifically selecting CD19/CD20/CD22/CD23 FITC-stained B cells and excluding debris and cell doublets.

In parallel, analysis of the cell-cycle distribution of normal B cells was performed on a total of 20 samples—10 BM, 5 PB, and 5 reactive LNs—obtained from an identical number of adult volunteers without any known hematologic disorder and normal blood cell counts. For this purpose, normal BM, PB, and RLN samples were stained with DRAQ5 (Cytognos), together with CD45-FITC/CD19-PE, CD38-FITC/CD19-PE for B17, CD20-FITC/CD23-PE for PB, and CD38-FITC/CD20-PE (all purchased from BD Biosciences), for LN sacrophage as previously described in detail.53  Briefly, after being stained with saturating amounts of the above-listed monoclonal antibodies (MAbs) and lysed (Quicklysis; Cytognos), cells were resuspended in 0.5 mL PBS to which 3 μL DRAQ5 (Vitro SA, Madrid, Spain) was added; afterward, an incubation was performed for 10 minutes in the dark at room temperature (RT). Immediately after this incubation period, sample aliquots were measured in a FACSCalibur flow cytometer using the CellQUEST software program. For each sample aliquot, information about 5 × 105 cells was stored. For data analysis, the PAINT-A-GATE software program (BD Biosciences) was used. For each sample, the following cell populations were identified after excluding dead cells and cell doublets in a sideward light scatter (SSC) versus DRAQ5 fluorescence area and a DRAQ5 fluorescence area versus DRAQ5 fluorescence width bivariate dot plot (Figure 1): early CD45lo/CD19lo B-cell progenitors, late CD45+/CD19hi B-cell precursors, mature CD45hi/CD19+ B lymphocytes and CD38hi/CD19−/+ plasma cells in BM samples (Figure 1A-D); mature CD20+/CD23+ and CD20+CD23 B lymphocytes in PB (Figure 1E-F); and mature CD38lo/CD20+, CD38+CD20hi B lymphocytes and CD38hi/CD20−/lo plasma cells in RLN samples (Figure 1G-I).

Figure 1

Representative bivariate dot plots illustrating the gating strategy used for the inmunophenotypic identification of each B-cell subpopulation identified and the analysis of their cell-cycle distribution. The strategy used for the identification of B-cell subsets is shown for normal bone marrow (BM; panels A-D), peripheral blood (PB; panels E,F) and reactive lymph node (LN; panels G-I) samples (left columns), together with the strategy used for the analysis of their cell-cycle distribution according to their DNA cell contents (DRAQ5 fluorescence area: panels J-R in the right column). In these dot plots for each B-cell population represented by the corresponding colored label, G0/G1 cells are depicted as grey events while, whenever present (panels J-M and P-R) S+G2/M cells are displayed as black dots.

Figure 1

Representative bivariate dot plots illustrating the gating strategy used for the inmunophenotypic identification of each B-cell subpopulation identified and the analysis of their cell-cycle distribution. The strategy used for the identification of B-cell subsets is shown for normal bone marrow (BM; panels A-D), peripheral blood (PB; panels E,F) and reactive lymph node (LN; panels G-I) samples (left columns), together with the strategy used for the analysis of their cell-cycle distribution according to their DNA cell contents (DRAQ5 fluorescence area: panels J-R in the right column). In these dot plots for each B-cell population represented by the corresponding colored label, G0/G1 cells are depicted as grey events while, whenever present (panels J-M and P-R) S+G2/M cells are displayed as black dots.

Close modal

Statistical methods

For all continuous variables studied, their means and standard deviations (SDs), medians, and ranges were calculated using the SPSS software program (SPSS 12; Chicago, IL). To establish the statistical significance of the differences observed between groups, either the ANOVA or the Student t test and either the Kruskal-Wallis or Mann-Whitney U tests were used for continuous variables showing a parametric and nonparametric distribution, respectively. For categoric variables, frequencies were calculated and the chi-square test was used to establish the statistical significance of differences observed between groups. Survival curves were plotted according to the method of Kaplan and Meier, and the log-rank test was used to assess the statistical significance of the differences observed between curves. P values less than .05 were considered to be associated with statistical significance.

Cell-cycle distribution of different normal B-cell compartments

In normal BM, the proliferative rate of the B-cell populations significantly (P < .001) decreased with B-cell maturation from early CD45lo/CD19lo B-cell progenitors, to the late CD45+/CD19hi B-cell precursors, and the more mature CD45hi/CD19+ B lymphocytes (S + G2/M 7% ± 2.4%, 3% ± 1.2%, and 0.5% ± 1.1%, respectively; Table 2). In PB, even lower percentages of S + G2/M cells were found among mature CD20+/CD23+ (0.1% ± 0.8%) and CD20+CD23 (0.1% ± 0.05%) B lymphocytes (Table 2).

Table 2

Proliferative rate of different maturation-associated B-cell compartments from normal adult bone marrow (BM), peripheral blood (PB), and reactive lymph node (RLN) samples

Type of sample/ B-cell compartmentProliferative rate (% of S+G2/M cells)P
BM  <.001* 
    CD45lo/CD19lo 7 ± 2.4  
    CD45+/CD19hi 3 ± 1.2  
    CD45hi/CD19+ 0.5 ± 1.1  
    CD38hi/CD19−/+ 3 ± 1.1  
PB  NS 
    CD20+/CD23+ 0.1 ± 0.8  
    CD20+/CD23 0.1 ± 0.05  
RLN  <.05 
    CD38lo/CD20+ 2 ± 3  
    CD38+/CD20hi 13.5 ± 8  
    CD38hi/CD20−/lo 5.4 ± 2  
Type of sample/ B-cell compartmentProliferative rate (% of S+G2/M cells)P
BM  <.001* 
    CD45lo/CD19lo 7 ± 2.4  
    CD45+/CD19hi 3 ± 1.2  
    CD45hi/CD19+ 0.5 ± 1.1  
    CD38hi/CD19−/+ 3 ± 1.1  
PB  NS 
    CD20+/CD23+ 0.1 ± 0.8  
    CD20+/CD23 0.1 ± 0.05  
RLN  <.05 
    CD38lo/CD20+ 2 ± 3  
    CD38+/CD20hi 13.5 ± 8  
    CD38hi/CD20−/lo 5.4 ± 2  

Results are expressed as means plus or minus 1 standard deviation (range).

NS indicates no statistically significant differences: P > .05.

*

P < .001 for all BM B-cell compartments versus CD45loCD19lo B-cell precursors.

P < .05 for all LN B-cell compartments versus CD38+CD20hi B lymphocytes.

Mature CD38lo/CD20+ B lymphocytes from reactive lymph nodes (RLNs) showed a higher proliferative rate (% of S + G2/M cells of 2% ± 3%) than mature BM and PB B lymphocytes (P < .005; Table 2). In turn, RLN B lymphocytes showing a germinal center–associated phenotype (CD38+CD20hi) represented the compartment of mature B cells showing the highest proliferation (P < .005 vs other mature B lymphocytes and plasma cells) with a mean percentage of S + G2/M-phase cells of 13.5% plus or minus 8% (Table 2). The proliferative rate of more mature B cells decreased thereafter with maturation into CD38hi/CD20−/lo LN and CD38hi/CD19−/+ BM plasma cells—percentage of S + G2/M plasma cells of 5.4% plus or minus 2% (P < .001) and of 3% plus or minus 1.1%, (P < .005), respectively.

Cell-cycle distribution of neoplastic B cells according to the type of sample analyzed

Comparative analysis of the proliferative rate of neoplastic B cells from B-CLPD patients in different tissues (BM, PB, and LN) showed significant differences in B-CLL and MCL cases (Table 3). Accordingly, in B-CLL, a significantly (P = .003) higher percentage of cells in the S + G2/M cell-cycle phases was detected in BM versus PB (0.7% ± 1% vs 0.4% ± 0.5%, respectively); in turn, significantly higher percentages of S + G2/M cells were detected in LN (7.7% ± 8%) versus BM (2% ± 1.3%; P = .02) and PB (2.4% ± 4.3; P = .004) samples from MCL patients. By contrast, no significant differences were observed for the other B-CLPD subgroups once the proliferative rate of neoplastic B cells from different types of samples was compared. However, both BL and LPL/WM showed a tendency toward a higher proliferation in LN versus PB and LN versus PB and BM, respectively (Table 3).

Table 3

Proliferative rate of neoplastic B-cells in B-CLPD patients according to the type of sample analyzed: bone marrow (BM), peripheral blood (PB) and lymph node (LN)

Subtype of B-CLPD/ type of sample (no. of cases)Proliferative rate, % of S+G2/M cellsP
B-CLL  .003* 
    BM (n=78) 0.7 ± 1  
    PB (n=123) 0.4 ± 0.4  
    LN (n=9) 0.5 ± 0.5  
SMZL  NS 
    BM (n=7) 1.7 ± 2.4  
    PB (n=9) 0.5 ± 0.2  
HCL   
    BM (n=2) 1.7 ± 2.4  
    LN (n=5) 0.5 ± 0.2  
MALT  NS 
    BM (n=15) 1.4 ± 1.4  
    PB (n=4) 0.7 ± 0.5  
    LN (n=1) 4.6  
MCL  .04 
    BM (n=15) 2 ± 1.3  
    PB (n=16) 2.4 ± 4.3  
    LN (n=8) 7.7 ± 8  
FL  NS 
    BM (n=37) 3 ± 2.5  
    PB (n=9) 2.1 ± 2  
    LN (n=25) 3 ± 2  
BL  NS 
    PB (n=8) 15 ± 3.4  
    LN (n=11) 26 ± 13  
DLBCL  NS 
    BM (n=4) 12.6 ± 19  
    LN (n=15) 14 ± 7  
LPL/WM  NS 
    BM (n=31) 2 ± 2  
    PB (n=2) 1.1 ± 1.3  
    LN (n=3) 10.6 ± 16.7  
Subtype of B-CLPD/ type of sample (no. of cases)Proliferative rate, % of S+G2/M cellsP
B-CLL  .003* 
    BM (n=78) 0.7 ± 1  
    PB (n=123) 0.4 ± 0.4  
    LN (n=9) 0.5 ± 0.5  
SMZL  NS 
    BM (n=7) 1.7 ± 2.4  
    PB (n=9) 0.5 ± 0.2  
HCL   
    BM (n=2) 1.7 ± 2.4  
    LN (n=5) 0.5 ± 0.2  
MALT  NS 
    BM (n=15) 1.4 ± 1.4  
    PB (n=4) 0.7 ± 0.5  
    LN (n=1) 4.6  
MCL  .04 
    BM (n=15) 2 ± 1.3  
    PB (n=16) 2.4 ± 4.3  
    LN (n=8) 7.7 ± 8  
FL  NS 
    BM (n=37) 3 ± 2.5  
    PB (n=9) 2.1 ± 2  
    LN (n=25) 3 ± 2  
BL  NS 
    PB (n=8) 15 ± 3.4  
    LN (n=11) 26 ± 13  
DLBCL  NS 
    BM (n=4) 12.6 ± 19  
    LN (n=15) 14 ± 7  
LPL/WM  NS 
    BM (n=31) 2 ± 2  
    PB (n=2) 1.1 ± 1.3  
    LN (n=3) 10.6 ± 16.7  

Results expressed as means plus or minus 1 standard deviation. P values provided correspond to comparisons indicated.

B-CLPD indicates B-cell chronic lymphoproliferative disorders; B-CLL, B-cell chronic lymphocytic leukemia; SMZL, splenic marginal zone B-cell lymphoma; NS, statistically not significantly different (P > .05); HCL, hairy cell leukemia; MCL, mantle-cell lymphoma; MALT, mucosa-associated lymphoid tissue lymphoma; FL, follicular lymphoma; BL, Burkitt lymphoma; DLBCL, diffuse large B-cell lymphoma; and; LPL/WM, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia.

*

BM versus PB samples.

LN versus BM and PB samples.

Cell-cycle distribution of neoplastic B cells from patients with different diagnostic subtypes of B-CLPD

Overall, large B-cell lymphomas (BL and DLBCL) were those subgroups of B-CLPD showing the highest percentage of S + G2/M-phase neoplastic B cells, with no significant differences being observed in the proliferative rate between these 2 subgroups of B-NHL. However, in comparison with normal CD38+/CD20hi germinal center B cells, half of all BL cases—7/14—while only 3 (16%) of 19 DLBCL cases showed an abnormally increased proliferative rate (P = .001 and P = .03, respectively); in addition 4 (21%) of 19 (P = .01) DLBCL cases, but none of the BL patients, displayed decreased S + G2/M cell percentages. In contrast, despite showing a germinal center immunophenotype, FL cases displayed an overall intermediate proliferative rate that was significantly (P < .001) lower than the proliferative rate of normal CD38+/CD20hi RLN B cells (% of S + G2/M cells of 13.5% ± 8% vs 2.4% ± 2%, respectively), the percentage of S + G2/M B cells being decreased in 66 (93%) of 71 cases (Figure 2).

Figure 2

Proliferative rate of neoplastic B cells in individual B-CLPD patients in comparison with their normal counterparts. The range of S + G2/M-phase cells for the corresponding normal B-cell counterpart is shown as a gray box, and the dotted line corresponds to normal median values. *P < .05 for B-CLL versus normal mature bone marrow (BM) CD45hiCD19+ B cells and CD20+CD23+ peripheral blood (PB) B lymphocytes; **P = .01 for MCL LN samples versus CD38loCD20+ reactive LN B lymphocytes; ***P < .001 for FL versus CD38+CD20hi reactive LN B lymphocytes; and ****P < .05 for LPL/WM versus normal BM and lymph node (LN) plasma cells. B-CLL indicates B-cell chronic lymphocytic leukemia; HCL, hairy cell leukemia; MCL, mantle-cell lymphoma; SMZL, splenic marginal zone B-cell lymphoma; MALT, mucosa-associated lymphoid tissue lymphoma; FL, follicular lymphoma; DLBCL, diffuse large B-cell lymphoma; BL, Burkitt lymphoma; and LPL/WM, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia.

Figure 2

Proliferative rate of neoplastic B cells in individual B-CLPD patients in comparison with their normal counterparts. The range of S + G2/M-phase cells for the corresponding normal B-cell counterpart is shown as a gray box, and the dotted line corresponds to normal median values. *P < .05 for B-CLL versus normal mature bone marrow (BM) CD45hiCD19+ B cells and CD20+CD23+ peripheral blood (PB) B lymphocytes; **P = .01 for MCL LN samples versus CD38loCD20+ reactive LN B lymphocytes; ***P < .001 for FL versus CD38+CD20hi reactive LN B lymphocytes; and ****P < .05 for LPL/WM versus normal BM and lymph node (LN) plasma cells. B-CLL indicates B-cell chronic lymphocytic leukemia; HCL, hairy cell leukemia; MCL, mantle-cell lymphoma; SMZL, splenic marginal zone B-cell lymphoma; MALT, mucosa-associated lymphoid tissue lymphoma; FL, follicular lymphoma; DLBCL, diffuse large B-cell lymphoma; BL, Burkitt lymphoma; and LPL/WM, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia.

Close modal

Other B-CLPDs showing intermediate numbers of S + G2/M-phase cells were HCL (2.4% ± 3%), MCL (3.3% ± 5%), MALT-NHL (1.4% ± 1.5%), and LPL/WM (2.5% ± 5%). Once compared with normal mature CD38lo/CD20+ LN B cells (% of S + G2/M cells of 2% ± 3%) no significant differences were found for HCL and MALT-NHL; in contrast, 6 (16%) of 39 MCL cases (P = .001) and 3 (37%) of 8 LN samples from MCL (P = .01) showed abnormally increased percentages of S + G2/M cells (13.8% ± 5% and 17% ± 5%, respectively). Among LPL/WM cases, 30 (83%) of 36 patients showed a significantly (P < .001) lower percentage of S + G2/M-phase cells (1.2% ± 1%) than normal reactive CD38hi/CD20lo LN and BM plasma cells (5.4% ± 2% and 3% ± 1.1%, respectively; Figure 2). In contrast, S + G2/M cell percentages detected in LPL/WM cases were similar to those of CD38lo/CD20+ mature RLN B lymphocytes (Figure 2).

B-CLL together with SMZL were those B-CLPDs showing the lowest proportion of S + G2/M-phase cells (0.6% ± 0.7% and 0.9% ± 1.6%, respectively). Despite this, 73 (60%) of 123 PB B-CLL and 58 (74%) of 78 BM B-CLL cases showed a significantly higher proportion of S + G2/M-phase cells (0.6% ± 0.5% and 2.7% ± 2.1%; respectively) compared with mature CD20+/CD23+ PB B cells and with mature CD45hi/CD19+ BM B cells (P < .001), respectively; in contrast, no significant differences were observed between neoplastic B cells from SMZL patients and normal/reactive CD38loCD20+ LN B cells (Figure 2).

Overall, there was an association between the proliferative rate of neoplastic B cells as evaluated by the percentage of S + G2/M phase cells and the Ki67 score by immunohistochemistry assessed in a subset of 76 lymph node biopsies from an identical number of patients, particularly when comparing cases with high Ki67 expression (> 90% of neoplastic B cells; % of S + G2/M of 17% ± 15%) with those showing low (< 10%; % of S + G2/M of 2% ± 2%; P < .001), intermediate (10%-60%; % of S + G2/M of 2% ± 1.5%; P = .002) and intermediate-high (60%-90%; % of S + G2/M of 3% ± 1.5%; P = .01) Ki67 expression.

Impact of common recurrent cytogenetic abnormalities in the proliferative rate of neoplastic B cells

Most (385/432; 89%) B-CLPD patients included in the present study showed a diploid DNA content, while the remaining 47 cases (11%) were DNA hyperdiploid. The highest frequency of DNA aneuploid cases was observed among DLBCL (42%), BL (29%), MCL (21%), and FL (18%) cases, while all HCL and SMZL analyzed had a diploid DNA cell content. In turn, the frequency of DNA aneuploid cases detected by flow cytometry among MALT-NHL (10%), B-CLL (5%), and LPL/WM cases (6%) was low. Interestingly, upon comparing the DNA index (DI) of aneuploid cases, significant differences were observed between B-CLL cases (DI of 1.05 ± 0.04) in comparison with MCL (DI of 1.6 ± 0.4; P < .001), FL (DI of 1.3 ± 0.4; P < .001), DLBCL (DI of 1.4 ± 0.4; P < .001), and BL (DI of 1.4 ± 0.2; P = .01) patients, in line with previous observations.21,24,28,29,54  In addition, FL cases also showed a significantly lower DNA index than MCL cases (P = .02). Among B-CLL patients, DNA aneuploidy was associated with a higher percentage of S + G2/M-phase cells (1% ± 0.7% vs 0.5% ± 0.7%; P = .002) as previously reported.55  In contrast, no statistically significant differences were found as regards the proliferative rate of neoplastic B cells in the other subgroups of B-CLPD with respect to the presence or absence of DNA aneuploidy.

Among B-CLL cases, de1(13q) and trisomy 12 were those chromosomal abnormalities more frequently observed (37% and 22% of the cases, respectively) followed by del(11q) (9%) and del(17p) (7%). DNA aneuploid B-CLL patients showed a higher frequency of trisomy 12 versus diploid B-CLL cases (63% vs 20%; P = .003). In turn, in MALT-NHL cases, t(18q21) was negative in 16 of 16 cases studied, while gene rearrangements involving the 14q32.3 chromosome region were detected in 4 (25%) of 16 patients. In addition, t(14;18), BCL6 gene rearrangements, and t(8;14) were found in 86% (n = 61/71), 17%, and 6% of all FL patients, respectively; t(8;14) was identified in 13 (93%) of 14 BL cases, t(11;14) was found in 30 (77%) of 39 MCL, and BCL6 gene rearrangements were present in 5 (38%) of 13 DLBCL patients, none of the later cases showing t(14;18). Finally, del(6q21) was identified in 9 (30%) of 30 LPL/WM patients, and translocations associated with the 14q32.3 region were detected in 2 (8%) of 24 cases analyzed. Interestingly, once we analyzed the potential impact of the presence versus absence of these different cytogenetic abnormalities within the specific subgroups of B-CLPD patients on the proliferative rate of neoplastic B cells, presence of translocations associated with the 14q32.3 region in LPL/WM cases was associated with a higher percentage of S + G2/M-phase cells in comparison with negative cases (17% ± 18% vs 1.3% ± 0.7%; P = .006). In contrast, no statistically significant differences were found between cases showing a distinct cytogenetic pattern for any of the other diagnostic subgroups of B-CLPD analyzed except for a higher proliferative rate among FL cases with t(8;14) (P = .05).

Association between the proliferative rate of neoplastic B cells and other clinical and biologic disease features

Among B-CLPD patients as a whole, a significant association was observed between a worse performance status (ECOG ≥ 2; (P < .001), presence of infiltration of 2 or more extranodal sites (P = .001), thrombocytopenia (< 100 × 109/L; P = .01), increased LDH serum levels (P < .001), and a higher tumor cell proliferation (Table 4). In addition, FL patients with histologic grade 1 (P = .003) and grade II (P = .01) had lower percentage of S + G2/M phase cells compared with grade 3 cases; in line with this, absence of superficial adenopathies (P = .03) and infiltration of 2 or more extranodal sites (P < .001) in FL was also associated with a higher percentage of S + G2/M neoplastic B cells (Table 4). Similarly, a significantly higher proliferation (P = .03) was observed among stage B and C versus stage A B-CLL cases (Table 4). In contrast, presence of leukocytosis (> 10 × 109/L; P < .001) and lymphocytosis (> 5 × 109/L; P < .001) was associated with lower percentages of S + G2/M cells, particularly among B-CLL (P ≤ .06) and low-grade B-CLPD other than B-CLL, MCL, and FL (P ≤ .09). Regarding patient outcome, a clear association between higher percentages of S + G2/M cells and both progressive disease and shorter survival was observed after a median follow-up of 25 months (P = .005) for the whole B-CLPD group as well as for MCL patients (P = .03; Table 4).

Table 4

B-CLPD: relationship between the proliferative rate of neoplastic B cells and the clinical and biologic characteristics of the disease

B-CLL, n=151
MCL, n=27
FL, n=54
Other low-grade B-CLPD*, n=51
High-grade B-CLPD,† n=21
All B-CLPD patients, n=304
% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP
Patient characteristic                   
    Performance status                   
        ECOG 2 or less 97 0.5 ± 0.3 NS 84 3 ±4 NS 86 2.7 ±2.1 NS 87 1.5 ±2 .03 44 20 ±13 NS 11 2 ±4 <.001 
        ECOG more than 2 0.5 ±0.5  16 7 ±6  14 2.6 ±2.3  13 6.5 ±11  56 15 ±8  89 6 ±8  
        Adenopathies                   
        No 45 0.5 ±0.4 NS 11 4 ±6 NS 5.7 ±1.2 .03 71 1.5 ±1.6 NS 25 19 ±7 NS 42 1.5 ±3 NS 
        Yes 55 0.5 ±0.3  89 3 ±5  96 2.5 ±2  29 4 ±7  75 17 ±12  58 3.1 ±6  
    Infiltration of 2 or more extranodal sites                   
        No 95 0.4 ±0.4 <.01 83 2.4 ±3 NS 73 2.4 ±1.6 <.001 83 4 ±6 NS 45 12 ±7 NS 86 2 ±3.5 .001 
        Yes 3.5 ±6  17 1.5 ±1  26 3.3 ±3  17 10 ±13        
    B-CLL Binet stage                   
        A 75 0.4 ±0.3                 
        B 15 0.6 ±0.7 .03                
        C 10 0.7 ±0.7                 
    FL histologic grade                   
        Grade I       57 2.3 ±1.6           
        Grade II       32 2.1 ±1.5 <.05§          
        Grade III       11 5 ±2           
    Standard IPI                   
        Low (0;1) 84 0.4 ±0.3  21 3.8 ±5  45 2.2 ±2  41 1.3 ±1.3  40 21 ±13  39 5 ±8  
        Low to intermediate (2) 11 0.5 ±0.4 <.001 47 1.5 ±1.1 NS 24 2.3 ±1.4 NS 37 2 ±2.4 NS 40 11 ±8 NS 35 3.5 ±5 NS 
        High to intermediate (3) 2.4 ±1.2  32 6.5 ±8  24 3.3 ±2.6  18 7 ±13  13 24 ±8  22 5.7 ±8  
        High (4;5)     3.8 ±2  2.2  31  3.4 ±2  
Laboratory parameters                   
    WBC count less than 10×109/L 14 1.1 ±2 .03 57 5 ±6 .06 83 2.5 ±1.6 .06 24 3 ±5 .09 74 15 ±12 NS 45 4.7 ±7 <.001 
    Leukocytosis more than 10×109/L 86 0.4 ±0.4  43 2.4 ±4  17 2 ±2.5  76 1 ±1.4  26 14 ±13  55 1 ±2  
    Lymphocyte count less than 5×109/L 10 1.4 ±2.4 .06 55 4 ±4 .09 83 2.7 ±2 NS 24 3 ±6 .01 96  NS 45 5.1 ±7 <.001 
    Lymphocytosis more than 5×109/L 90 0.5 ±0.6  45 4 ±7  17 1.5 ±2  76 0.6 ±0.4  4   55 1 ±2.1  
    Hemoglobin                   
        Anemia less than 100 g/dL 0.5 ±0.4 NS 4 ±5 NS 0.4 ±0.1 .03 13 7 ±13 NS 11 30 ±1.4 .07 4 ±9 NS 
        More than 100 g/dL 92 0.5 ±0.9  95 0.2  96 2.5 ±2  87 2 ±2  89 16 ±10  92 2.5 ±5  
    Platelet count                   
        Thrombocytopenia less than 100×109/L 1.4 ±3 .01 10 7 ±8 NS 2.5 ±3 NS 13 8 ±12 NS 11 7 ±5 NS 4.3 ±7 .01 
     More than 100×109/L 93 0.5 ±0.5  90 3 ±5  92 2.5 ±2  87 2 ±2  89 18 ±11  91 2.4 ±5  
    Serum LDH                   
    Normal 94 0.5 ±0.8 .06 72 3 ±4 NS 23 2.3 ±2 NS 87 2 ±2 .01 63 17 ±11 NS 86 2 ±4.6 <.001 
    Increased more than 450 U/L 0.7 ±0.8  28 4 ±7  77 3.6 ±2.6  13 7 ±11  37 18 ±1o  14 5 ±7  
Patient follow-up                   
    Outcome                   
        Stable disease 64 0.4 ±0.3 NS 19 1.3 ±1 NS 14 2 ±1.6 NS 50 1.3 ±1.4 NS NS 46 0.7 ±0.9 <.001 
        Progressive disease 0.4 ±0.3  33 6 ±7  25 2.7 ±1.7  1.2 ±1  28 20 ±10  14 3.4 ±5  
    Total deaths                   
        No 90 0.5 ±0.8 NS 50 1.3 ±1.1 .007 80 2.4 ±2 NS 84 2 ±5 NS 57 15 ±12 NS 82 2 ±5 0.009 
        Yes 10 0.5 ±0.8  50 6.5 ±6.2  20 3 ±1.6  16 2 ±3  43 19 ±8  18 5 ±6  
Median survival Cutoff value Median survival, mo P Cutoff value Median survival, mo P Cutoff value Median survival, mo P Cutoff value Median survival, mo P Cutoff valueMedian survival, mo P Cutoff value Median survival, mo P 
    Low proliferative rate <0.34% NR NS <1.4% NR .03 <1.8% NR NS <1% NR NS <13.7% NR NS <0.7% NR .005 
    High proliferative rate ≥0.34% NR  ≥1.4% 12  ≥1.8% NR  ≥1% 34  ≥13.7% NR  ≥0.7% 68  
B-CLL, n=151
MCL, n=27
FL, n=54
Other low-grade B-CLPD*, n=51
High-grade B-CLPD,† n=21
All B-CLPD patients, n=304
% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP% of cases% of S+G2/M cellsP
Patient characteristic                   
    Performance status                   
        ECOG 2 or less 97 0.5 ± 0.3 NS 84 3 ±4 NS 86 2.7 ±2.1 NS 87 1.5 ±2 .03 44 20 ±13 NS 11 2 ±4 <.001 
        ECOG more than 2 0.5 ±0.5  16 7 ±6  14 2.6 ±2.3  13 6.5 ±11  56 15 ±8  89 6 ±8  
        Adenopathies                   
        No 45 0.5 ±0.4 NS 11 4 ±6 NS 5.7 ±1.2 .03 71 1.5 ±1.6 NS 25 19 ±7 NS 42 1.5 ±3 NS 
        Yes 55 0.5 ±0.3  89 3 ±5  96 2.5 ±2  29 4 ±7  75 17 ±12  58 3.1 ±6  
    Infiltration of 2 or more extranodal sites                   
        No 95 0.4 ±0.4 <.01 83 2.4 ±3 NS 73 2.4 ±1.6 <.001 83 4 ±6 NS 45 12 ±7 NS 86 2 ±3.5 .001 
        Yes 3.5 ±6  17 1.5 ±1  26 3.3 ±3  17 10 ±13        
    B-CLL Binet stage                   
        A 75 0.4 ±0.3                 
        B 15 0.6 ±0.7 .03                
        C 10 0.7 ±0.7                 
    FL histologic grade                   
        Grade I       57 2.3 ±1.6           
        Grade II       32 2.1 ±1.5 <.05§          
        Grade III       11 5 ±2           
    Standard IPI                   
        Low (0;1) 84 0.4 ±0.3  21 3.8 ±5  45 2.2 ±2  41 1.3 ±1.3  40 21 ±13  39 5 ±8  
        Low to intermediate (2) 11 0.5 ±0.4 <.001 47 1.5 ±1.1 NS 24 2.3 ±1.4 NS 37 2 ±2.4 NS 40 11 ±8 NS 35 3.5 ±5 NS 
        High to intermediate (3) 2.4 ±1.2  32 6.5 ±8  24 3.3 ±2.6  18 7 ±13  13 24 ±8  22 5.7 ±8  
        High (4;5)     3.8 ±2  2.2  31  3.4 ±2  
Laboratory parameters                   
    WBC count less than 10×109/L 14 1.1 ±2 .03 57 5 ±6 .06 83 2.5 ±1.6 .06 24 3 ±5 .09 74 15 ±12 NS 45 4.7 ±7 <.001 
    Leukocytosis more than 10×109/L 86 0.4 ±0.4  43 2.4 ±4  17 2 ±2.5  76 1 ±1.4  26 14 ±13  55 1 ±2  
    Lymphocyte count less than 5×109/L 10 1.4 ±2.4 .06 55 4 ±4 .09 83 2.7 ±2 NS 24 3 ±6 .01 96  NS 45 5.1 ±7 <.001 
    Lymphocytosis more than 5×109/L 90 0.5 ±0.6  45 4 ±7  17 1.5 ±2  76 0.6 ±0.4  4   55 1 ±2.1  
    Hemoglobin                   
        Anemia less than 100 g/dL 0.5 ±0.4 NS 4 ±5 NS 0.4 ±0.1 .03 13 7 ±13 NS 11 30 ±1.4 .07 4 ±9 NS 
        More than 100 g/dL 92 0.5 ±0.9  95 0.2  96 2.5 ±2  87 2 ±2  89 16 ±10  92 2.5 ±5  
    Platelet count                   
        Thrombocytopenia less than 100×109/L 1.4 ±3 .01 10 7 ±8 NS 2.5 ±3 NS 13 8 ±12 NS 11 7 ±5 NS 4.3 ±7 .01 
     More than 100×109/L 93 0.5 ±0.5  90 3 ±5  92 2.5 ±2  87 2 ±2  89 18 ±11  91 2.4 ±5  
    Serum LDH                   
    Normal 94 0.5 ±0.8 .06 72 3 ±4 NS 23 2.3 ±2 NS 87 2 ±2 .01 63 17 ±11 NS 86 2 ±4.6 <.001 
    Increased more than 450 U/L 0.7 ±0.8  28 4 ±7  77 3.6 ±2.6  13 7 ±11  37 18 ±1o  14 5 ±7  
Patient follow-up                   
    Outcome                   
        Stable disease 64 0.4 ±0.3 NS 19 1.3 ±1 NS 14 2 ±1.6 NS 50 1.3 ±1.4 NS NS 46 0.7 ±0.9 <.001 
        Progressive disease 0.4 ±0.3  33 6 ±7  25 2.7 ±1.7  1.2 ±1  28 20 ±10  14 3.4 ±5  
    Total deaths                   
        No 90 0.5 ±0.8 NS 50 1.3 ±1.1 .007 80 2.4 ±2 NS 84 2 ±5 NS 57 15 ±12 NS 82 2 ±5 0.009 
        Yes 10 0.5 ±0.8  50 6.5 ±6.2  20 3 ±1.6  16 2 ±3  43 19 ±8  18 5 ±6  
Median survival Cutoff value Median survival, mo P Cutoff value Median survival, mo P Cutoff value Median survival, mo P Cutoff value Median survival, mo P Cutoff valueMedian survival, mo P Cutoff value Median survival, mo P 
    Low proliferative rate <0.34% NR NS <1.4% NR .03 <1.8% NR NS <1% NR NS <13.7% NR NS <0.7% NR .005 
    High proliferative rate ≥0.34% NR  ≥1.4% 12  ≥1.8% NR  ≥1% 34  ≥13.7% NR  ≥0.7% 68  

Significant differences were not found for sex, Ann Arbor stage, B symptoms, hepatomegaly, splenomegaly, bulky disease, and b-2 microglobulin serum levels.

B-CLPD indicates B-cell chronic lymphoproliferative disorders; B-CLL, B-cell chronic lymphocytic leukemia; MCL, mantle-cell lymphoma; FL, follicular lymphoma; LDH, lactic dehydrogenase; NS, no statistically significant differences found (P > .05); and NR, not reached.

Results expressed as mean plus or minus 1 standard deviation.

§

P value provided corresponds to comparisons between histologic grade I and II versus grade III FL, respectively.

*

Other low-grade B-NHL: hairy cell leukemia; splenic marginal zone B-cell lymphoma; mucosa-associated lymphoid tissue lymphoma; and lymphoplasmacytic lymphoma/Waldenström macroglobulinemia.

High-grade B-NHL: diffuse large B-cell lymphoma; Burkitt lymphoma; and other unclassifiable high-grade B-NHLs.

Only 1 case showing 5 x 109/L or more lymphocytes.

The cutoff value used for grouping into the low and high proliferative rate categories in each diagnostic subtype corresponds to the median value of the percentage of S+G2M cells.

The proliferative activity of neoplastic B cells has long been suggested to be a relevant prognostic factor in B-CLPD associated with both response to chemotherapy and patient survival,17,27,30-32,34,35,56  as also found here. However, evaluation of cell proliferation has only partially translated into routine clinical practice through the use of semiquantitative immunohistochemical stains for individual cell cycle–associated proteins such as Ki67 and PCNA.14-19,34,57-59  Although, Ki67 immunohistochemistry alone is a powerful tool to measure the distribution of proliferating cells and has been found to correlate with worse clinical outcome,59,60  to the best of our knowledge, no study has been reported so far in which the proliferative rate of neoplastic B-CLPD cells is analyzed in a reproducible and quantitative way in comparison with that of their normal counterpart. Such comparison would provide further insight on the impact of specific genetic abnormalities on the proliferation of neoplastic B cells in specific subtypes of both chronic lymphoid leukemias and B-NHL.

Early BM B-cell development concludes when a B-cell precursor successfully rearranges and expresses Ig heavy and light chain genes coupled with CD79 at the cell surface membrane, to form a functional antigen receptor (BCR) capable of recognizing non–self-antigens.3,61  During this period, B-cell maturation is associated with a significant expansion of the B-cell compartment until large numbers of mature, naive, resting B cells are generated.3  Afterward, mature resting B cells leave the BM62  and recirculate through PB and the LN.63  Under appropriate T cell–dependent stimulatory conditions, antigen recognition through the BCR induces the activation, (mono/oligo) clonal expansion, and further maturation of B cells; in the germinal center (GC) B-cell responses are associated with an increase affinity of the BCR for the antigen recognized and Ig class switch through the occurrence of both hypermutation and rearrangements of the Ig genes.62,64-66  In line with this, mature PB and marginal zone B lymphocytes are characterized by signals that help to maintain a quiescent state (eg, low expression of cyclins such as CCNA, CDK1, and CCNB1), while GC cells have a high proliferation gene expression signature (eg, high expression of CCNB1, PCNA, and Ki67).67  Further maturation of GC B cells into plasma cells (PCs) capable of producing high-affinity antibodies is followed by a progressive decreased proliferation3,63,66,68  also supported by the observation that CD45hi/bcl2lo PCs from secondary lymphoid organs and PB are highly proliferating, while CD45dim/bcl2+ PCs from BM have a lower proliferative rate decreasing in parallel to CD45 expression.69 

In this study, we accurately establish the magnitude of the variations in the proliferative fraction of B cells at different maturation stages providing a frame of reference for the understanding of the impact of neoplastic transformation of mature B cells in their proliferative rate. Accordingly, our results show the existence of a progressively lower proportion of S + G2/M cells from the more immature CD45lo/CD19lo and CD45+/CD19hi BM B-cell precursors to the more mature BM CD45hi/CD19+ and PB CD20+CD23+ and CD20+CD23 B lymphocytes. In turn, RLN B cells displaying a germinal center–associated immunophenotype (CD38+CD20hi B cells) showed a higher proliferative rate than that of marginal zone resting B lymphocytes, and the more mature plasma cells (PCs) from both RLN (CD38hiCD20−/lo) and BM (CD38+++CD19+). Overall, these results confirm the occurrence of variations on the proliferative rate of BM, PB, and RLN B cells in association with both the maturation stage and activation status of B cells.3,69,70 

Previous reports indicate that B-CLPDs displaying a low proliferative rate correspond to relatively indolent, low-grade tumors, while B-CLPDs with a higher proliferative rate typically (> 10%) show an aggressive clinical course.23,25,28  In line with these observations, we found a clear association between higher percentages of S + G2/M cells and both adverse prognostic features (eg, advanced patient performance status, disease stage, high LDH serum levels, and presence of thrombocytopenia), progressive disease, and a shorter overall survival for the whole B-CLPD group as well as for a specific diagnostic subgroups of the disease such as MCL.

Overall, a relatively high degree of heterogeneity between the different subgroups of B-CLPDs analyzed was also observed. Large cell lymphomas (BL and DLBCL) were those showing the highest percentage of S + G2/M-phase neoplastic B cells; intermediate proliferative rates were observed for HCL, MCL, MALT-LNH, FL, and LPL/WM, while B-CLL and SMZL patients were those subtypes of B-CLPD showing the lowest proportion of S + G2/M-phase cells. Upon comparing the proliferative rate of normal versus neoplastic B cells, similar median percentages of S + G2/M cells were found for HCL, SMZL, and MALT-NHL patients. However at present, the normal counterpart for these specific subgroups of B-CLPD—which are presumably derived from either marginal zone B cells or post-CG memory B cells—still remains to be fully identified.3,71  In contrast, a significantly increased proportion of S + G2/M-phase cells was observed among a relevant fraction of all B-CLL, BL, DLBCL, and MCL cases compared with their normal counterpart (CD20+CD23+ PB B lymphocytes, RLN CD38+CD20hi, CD38+CD20hi, and CD38loCD20+ B cells, respectively). In contrast, most FLs and LPL/WMs displayed a lower proliferative rate than their normal counterpart. Despite this, a more careful analysis of the results shows the existence of an important degree of variability even among individual cases within specific diagnostic subgroups of B-CLPD. Interestingly, significantly higher percentages of S + G2/M-phase cells were found in B-CLL cases in BM versus PB, and in RLN from MCL patients versus both BM and PB. The higher proliferative rate observed in specific tissues supports the existence of a nonrandom distribution of the proliferative compartment of neoplastic B cells in B-CLPD, with specific tissues providing a more favorable microenvironment for the expansion of the neoplastic clones.3,63,66,68,69,72  In line with this hypothesis, previous studies have shown that the patterns of tissue compartmentalization, intranodal growth, and hematologic spread of malignant cells in B-CLPD are influenced by variations in the adhesion properties of tumor B cells to BM and/or LN stromal cells.73-78  As an example, low-grade B-NHL typically show stronger expression of CD44 and CD54 in LN than in PB and BM.79,80  Similarly, among B-CLL cases, a relationship between the expression of CD44 and a diffuse pattern of BM infiltration has been reported,81  consistent with the finding that CD44 mediates adhesion of maturing B cells to BM stromal cells.82  In addition, it has also been shown that in individual patients, “pseudofollicular” proliferation centers in tissues involved by B-CLL, including BM on average, display a greater proportion of CD38+ cells and higher proliferation, versus neoplastic PB B cells.83-86  Of note, comparative evaluation of the Ki67 score by immunohistochemistry and the percentage of S + G2/M cells in lymph node biopsies from patients with B-CLPD showed a significant association between both parameters, in line with previous observations.14,22 

Despite this, variations in cell proliferation of neoplastic B cells within specific subtypes of B-CLPDs could not be fully explained by differences in the tissues analyzed. Accordingly, half of all BL patients displayed aberrantly increased percentages of S + G2/M cells in tumoral LN, compared with GC B lymphocytes. Similarly, one-fourth of all DLBCLs displayed altered (either increased or decreased) cell proliferation in lymphoid tissue samples. These results could suggest that, apart from reflecting the proliferative rate of their normal counterpart, specific genetic abnormalities associated with the different diagnostic subgroups of B-CLPDs analyzed could contribute to modulate the cell-cycling rate of neoplastic B cells.1  However, upon comparing within individual diagnostic subgroups of patients the proliferative rate of neoplastic B cells from cases carrying different genetic abnormalities, no major differences were observed between MALT, MCL, FL, BL, and DLBCL for the presence versus absence of t(14q32), t(11;14), t(14;18), t(8;14), and BCL6 rearrangements, respectively. In contrast, in line with previous observations,55  DNA aneuploid B-CLL cases as well as LPL/WM patients carrying 14q32 gene rearrangements and FL with t(8;14) were associated with an increased cell proliferation once compared with DNA diploid B-CLL, 14q32-negative LPL/WM, and t(8;14)-negative FL cases, respectively.

Overall, these findings confirm the notion that among the different genetic abnormalities analyzed, some are more likely related to an inhibitory effect on apoptosis and a prolonged B-cell survival in the absence of an increase in cell cycling (eg, t(14;18) with overexpression of bcl2 in FL87,88 ), while others more frequently lead to an augmented cell proliferation (eg, C-MYC gene rearrangements) in BL and FL.1  Similarly, overexpression of cyclin D1 among MCL patients does not appear to confer a proliferative advantage, in line with previous observations showing a similar gene expression signature and survival rates for typical MCL patients showing overexpression of cyclin D1 and MCL cases that are negative for this cell cycle–associated protein and that have acquired alternative mechanisms to accelerate entry into S-phase, independent of cyclin D1 expression.89 

In general, low- and high-grade B-NHL are both able to accumulate other alterations in cell-cycle regulators, most frequently involving tumor suppressor genes such as BCL6, p16, p53, and p27 that confer an increased aggressiveness and proliferative advantage to tumoral cells.1,90-92  However, in the present study, neither p53 deletion in B-CLL nor BCL6 gene rearrangements in DLBCL and FL were associated with an increased cell proliferation. In line with these observations, previous reports have shown that overexpression of BCL6 is often independent of chromosomal translocations involving the BCL6 gene, indicating that BCL6 expression might be associated with a high proliferation in both DLBCL and FL cases independently of the occurrence of 3q27 gene rearrangements.93,94  Similarly, no direct correlation has been observed between the percentage of proliferating Ki67+ B-CLL cells and p53-positive cells.95 

In summary, our results confirm that the proliferative rate of neoplastic cells from patients with B-CLPD is highly heterogeneous, such variability reflecting their maturation stage and to a certain extent also unique specific underlying genetic abnormalities; at the same time, it is associated with the clinical and biologic behavior of the disease. Further variations in B-CLL and MCL could also be related to the tissue compartment analyzed and the microenvironment of the neoplastic B cells.

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

This work has been partially supported by the following grants: FIS 06/0824, from the Ministerio de Sanidad y Consumo (Madrid, Spain) and RETICC RD06/0020/0035 from the Instituto de Salud Carlos III, Ministerio de Sanidad y Consumo (Madrid, Spain). S.Q. is supported by a grant from COLCIENCIAS (Bogotá, Colombia), J.M.S. is supported by a grant from the Ministerio de Sanidad y Consumo (Madrid, Spain; CP05/00321), A.R. is supported by a grant from the Ministerio de Ciencia y Tecnología (Madrid, Spain) y Fondo Social Europeo, and C.F. is supported by a grant from the Instituto de Salud Carlos III (Madrid, Spain; CM05/00250).

Contribution: S.Q. performed research, collected and analyzed data, and wrote the paper; A.L., A.R., S.B., M.L.S., J.F., C.F., and M.C.G. performed research and collected data; J.M.S., C.S.O., N.F., M. González, and P.G. performed research and analyzed data; M. Giralt performed research; M.C.P., J.M.M.-A., O.G., L.P., J.D.M., M.G.S., A.A.R., C.C., J.L.G., and R.B., collected data; J.A. collected and analyzed data, and wrote the paper; A.O. designed research, collected and analyzed data, and wrote the paper.

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

Correspondence: Alberto Orfao, Servicio General de Citometría, Laboratorio de Hematología, Hospital Universitario Salamanca, Paseo de San Vicente, 58-182, 37007 Salamanca, Spain; e-mail: orfao@usal.es.

1
Sanchez-Beato
 
M
Sanchez-Aguilera
 
A
Piris
 
MA
Cell cycle deregulation in B-cell lymphomas.
Blood
2003
, vol. 
101
 (pg. 
1220
-
1235
)
2
Sanchez-Beato
 
M
Saez
 
AI
Martinez-Montero
 
JC
, et al. 
Cyclin-dependent kinase inhibitor p27KIP1 in lymphoid tissue: p27KIP1 expression is inversely proportional to the proliferative index.
Am J Pathol
1997
, vol. 
151
 (pg. 
151
-
160
)
3
Kuppers
 
R
Mechanisms of B-cell lymphoma pathogenesis.
Nat Rev Cancer
2005
, vol. 
5
 (pg. 
251
-
262
)
4
Karin
 
M
Lin
 
A
NF-kappaB at the crossroads of life and death.
Nat Immunol
2002
, vol. 
3
 (pg. 
221
-
227
)
5
Dang
 
CV
c-Myc target genes involved in cell growth, apoptosis, and metabollism.
Mol Cell Biol
1999
, vol. 
19
 (pg. 
1
-
11
)
6
Boxer
 
LM
Dang
 
CV
Translocations involving c-myc and c-myc function.
Oncogene
2001
, vol. 
20
 (pg. 
5595
-
5610
)
7
Kramer
 
MH
Hermans
 
J
Wijburg
 
E
, et al. 
Clinical relevance of BCL2, BCL6, and MYC rearrangements in diffuse large B-cell lymphoma.
Blood
1998
, vol. 
92
 (pg. 
3152
-
3162
)
8
Hecht
 
JL
Aster
 
JC
Molecular biology of Burkitt's lymphoma.
J Clin Oncol
2000
, vol. 
18
 (pg. 
3707
-
3721
)
9
Wolowiec
 
D
Ciszak
 
L
Kosmaczewska
 
A
, et al. 
Cell cycle regulatory proteins and apoptosis in B-cell chronic lymphocytic leukemia.
Haematologica
2001
, vol. 
86
 (pg. 
1296
-
1304
)
10
Gottardi
 
D
Alfarano
 
A
De Leo
 
AM
, et al. 
Defective apoptosis due to Bcl-2 overexpression may explain why B-CLL cells accumulate in G0.
Curr Top Microbiol Immunol
1995
, vol. 
194
 (pg. 
307
-
312
)
11
Kitada
 
S
Andersen
 
J
Akar
 
S
, et al. 
Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with In vitro and In vivo chemoresponses.
Blood
1998
, vol. 
91
 (pg. 
3379
-
3389
)
12
Menendez
 
P
Vargas
 
A
Bueno
 
C
, et al. 
Quantitative analysis of bcl-2 expression in normal and leukemic human B-cell differentiation.
Leukemia
2004
, vol. 
18
 (pg. 
491
-
498
)
13
Ghia
 
P
Boussiotis
 
VA
Schultze
 
JL
, et al. 
Unbalanced expression of bcl-2 family proteins in follicular lymphoma: contribution of CD40 signaling in promoting survival.
Blood
1998
, vol. 
91
 (pg. 
244
-
251
)
14
Pich
 
A
Ponti
 
R
Valente
 
G
, et al. 
MIB-1, Ki67, and PCNA scores and DNA flow cytometry in intermediate grade malignant lymphomas.
J Clin Pathol
1994
, vol. 
47
 (pg. 
18
-
22
)
15
Rabenhorst
 
SH
Burini
 
RC
Schmitt
 
FC
Proliferating cell nuclear antigen (PCNA) in non-Hodgkin's lymphomas: correlation with working formulation and Kiel classification in formalin-fixed paraffin-embedded material.
Pathology
1996
, vol. 
28
 (pg. 
12
-
16
)
16
Sebo
 
TJ
Roche
 
PC
Witzig
 
TE
Kurtin
 
PJ
Proliferative activity in non-Hodgkin's lymphomas: a comparison of the bromodeoxyuridine labeling index with PCNA immunostaining and quantitative image analysis.
Am J Clin Pathol
1993
, vol. 
99
 (pg. 
668
-
672
)
17
Koster
 
A
Tromp
 
HA
Raemaekers
 
JM
, et al. 
The prognostic significance of the intra-follicular tumor cell proliferative rate in follicular lymphoma.
Haematologica
2007
, vol. 
92
 (pg. 
184
-
190
)
18
Tominaga
 
K
Yamaguchi
 
Y
Nozawa
 
Y
Abe
 
M
Wakasa
 
H
Proliferation in non-Hodgkin's lymphomas as determined by immunohistochemical double staining for Ki-67.
Hematol Oncol
1992
, vol. 
10
 (pg. 
163
-
169
)
19
Weiss
 
LM
Strickler
 
JG
Medeiros
 
LJ
, et al. 
Proliferative rates of non-Hodgkin's lymphomas as assessed by Ki-67 antibody.
Hum Pathol
1987
, vol. 
18
 (pg. 
1155
-
1159
)
20
Kvaloy
 
S
Marton
 
PF
Kaalhus
 
O
, et al. 
3H-thymidine uptake in B cell lymphomas: relationship to treatment response and survival.
Scand J Haematol
1985
, vol. 
34
 (pg. 
429
-
435
)
21
Saikia
 
UN
Dey
 
P
Vohra
 
H
Gupta
 
SK
DNA flow cytometry of non-Hodgkin's lymphomas: correlation with cytologic grade and clinical relapse.
Diagn Cytopathol
2000
, vol. 
22
 (pg. 
152
-
156
)
22
Horii
 
A
Yoshida
 
J
Hattori
 
K
, et al. 
DNA ploidy, proliferative activities, and immunophenotype of malignant lymphoma: application of flow cytometry.
Head Neck
1998
, vol. 
20
 (pg. 
392
-
398
)
23
Winter
 
JN
Andersen
 
J
Variakojis
 
D
, et al. 
Prognostic implications of ploidy and proliferative activity in the diffuse, aggressive non-Hodgkin's lymphomas.
Blood
1996
, vol. 
88
 (pg. 
3919
-
3925
)
24
Wooldridge
 
TN
Grierson
 
HL
Weisenburger
 
DD
, et al. 
Association of DNA content and proliferative activity with clinical outcome in patients with diffuse mixed cell and large cell non-Hodgkin's lymphoma.
Cancer Res
1988
, vol. 
48
 (pg. 
6608
-
6613
)
25
Holte
 
H
Suo
 
Z
Smeland
 
EB
, et al. 
Prognostic value of lymphoma-specific S-phase fraction compared with that of other cell proliferation markers.
Acta Oncol
1999
, vol. 
38
 (pg. 
495
-
503
)
26
Macartney
 
JC
Camplejohn
 
RS
DNA flow cytometry of non-Hodgkin's lymphomas.
Eur J Cancer
1990
, vol. 
26
 (pg. 
635
-
637
)
27
Macartney
 
JC
Proliferative activity in lymphomas.
J Pathol
1988
, vol. 
156
 (pg. 
7
-
8
)
28
Pinto
 
A
Cabecadas
 
J
Nobrega
 
SD
Mendonca
 
E
Flow cytometric S-phase fraction as a complementary biological parameter for the cytological grading of non-Hodgkin's lymphoma.
Diagn Cytopathol
2003
, vol. 
29
 (pg. 
194
-
199
)
29
Macartney
 
JC
Camplejohn
 
RS
Morris
 
R
, et al. 
DNA flow cytometry of follicular non-Hodgkin's lymphoma.
J Clin Pathol
1991
, vol. 
44
 (pg. 
215
-
218
)
30
Braylan
 
RC
Flow-cytometric DNA analysis in the diagnosis and prognosis of lymphoma.
Am J Clin Pathol
1993
, vol. 
99
 (pg. 
374
-
380
)
31
Holte
 
H
Zuo
 
H
Smeland
 
EB
, et al. 
Prognostic value of lymphoma-specific S-phase fraction compared with that of other cell proliferation markers.
Acta Oncol
1999
, vol. 
38
 (pg. 
495
-
503
)
32
Duque
 
RE
Andreeff
 
M
Braylan
 
RC
Diamond
 
LW
Peiper
 
SC
Consensus review of the clinical utility of DNA flow cytometry in neoplastic hematopathology.
Cytometry
1993
, vol. 
14
 (pg. 
492
-
496
)
33
Joensuu
 
H
Klemi
 
PJ
Soderstrom
 
KO
Jalkanen
 
S
Comparison of S-phase fraction, working formulation, and Kiel classification in non-Hodgkin's lymphoma.
Cancer
1991
, vol. 
68
 (pg. 
1564
-
1571
)
34
Katzenberger
 
T
Petzoldt
 
C
Holler
 
S
, et al. 
The Ki67 proliferation index is a quantitative indicator of clinical risk in mantle cell lymphoma [Letter].
Blood
2006
, vol. 
107
 pg. 
3407
 
35
Macartney
 
JC
Camplejohn
 
RS
Alder
 
J
Stone
 
MG
Powell
 
G
Prognostic importance of DNA flow cytometry in non-Hodgkin's lymphomas.
J Clin Pathol
1986
, vol. 
39
 (pg. 
542
-
546
)
36
Harris
 
NL
Jaffe
 
E
Diebold
 
J
, et al. 
The World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: Report of the Clinical Advisory Committee meeting, Airlie House, Virginia, November, 1997.
Ann Oncol
1999
, vol. 
10
 (pg. 
1419
-
1432
)
37
Matutes
 
E
Owusu-Ankomah
 
K
Morilla
 
R
, et al. 
The immunological profile of B-cell disorders and proposal of a scoring system for the diagnosis of CLL.
Leukemia
1994
, vol. 
8
 (pg. 
1640
-
1645
)
38
Sanchez
 
ML
Almeida
 
J
Vidriales
 
B
, et al. 
Incidence of phenotypic aberrations in a series of 467 patients with B chronic lymphoproliferative disorders: basis for the design of specific four-color stainings to be used for minimal residual disease investigation.
Leukemia
2002
, vol. 
16
 (pg. 
1460
-
1469
)
39
Watanabe
 
T
Hotta
 
T
Ichikawa
 
A
, et al. 
The MDM2 oncogene overexpression in chronic lymphocytic leukemia and low-grade lymphoma of B-cell origin.
Blood
1994
, vol. 
84
 (pg. 
3158
-
3165
)
40
Dohner
 
H
Stilgenbauer
 
S
Benner
 
A
, et al. 
Genomic aberrations and survival in chronic lymphocytic leukemia.
N Engl J Med
2000
, vol. 
343
 (pg. 
1910
-
1916
)
41
Sindelarova
 
L
Michalova
 
K
Zemanova
 
Z
, et al. 
Incidence of chromosomal anomalies detected with FISH and their clinical correlations in B-chronic lymphocytic leukemia.
Cancer Genet Cytogenet
2005
, vol. 
160
 (pg. 
27
-
34
)
42
Bosch
 
F
Jares
 
P
Campo
 
E
, et al. 
PRAD-1/cyclin D1 gene overexpression in chronic lymphoproliferative disorders: a highly specific marker of mantle cell lymphoma.
Blood
1994
, vol. 
84
 (pg. 
2726
-
2732
)
43
Bench
 
AJ
Erber
 
WN
Follows
 
GA
Scott
 
MA
Molecular genetic analysis of haematological malignancies, II: mature lymphoid neoplasms.
Int J Lab Hematol
2007
, vol. 
29
 (pg. 
229
-
260
)
44
Uren
 
AG
O'Rourke
 
K
Aravind
 
LA
, et al. 
Identification of paracaspases and metacaspases: two ancient families of caspase-like proteins, one of which plays a key role in MALT lymphoma.
Mol Cell
2000
, vol. 
6
 (pg. 
961
-
967
)
45
Dierlamm
 
J
Baens
 
M
Wlodarska
 
I
, et al. 
The apoptosis inhibitor gene API2 and a novel 18q gene, MLT, are recurrently rearranged in the t(11;18)(q21;q21) associated with mucosa-associated lymphoid tissue lymphomas.
Blood
1999
, vol. 
93
 (pg. 
3601
-
3609
)
46
Willis
 
TG
Dyer
 
MJ
The role of immunoglobulin translocations in the pathogenesis of B-cell malignancies.
Blood
2000
, vol. 
96
 (pg. 
808
-
822
)
47
Ye
 
BH
BCL-6 in the pathogenesis of non-Hodgkin's lymphoma.
Cancer Invest
2000
, vol. 
18
 (pg. 
356
-
365
)
48
Blum
 
KA
Lozanski
 
G
Byrd
 
JC
Adult Burkitt leukemia and lymphoma.
Blood
2004
, vol. 
104
 (pg. 
3009
-
3020
)
49
Iida
 
S
Rao
 
PH
Nallasivam
 
P
, et al. 
The t(9;14)(p13;q32) chromosomal translocation associated with lymphoplasmacytoid lymphoma involves the PAX-5 gene.
Blood
1996
, vol. 
88
 (pg. 
4110
-
4117
)
50
Schop
 
RF
Kuehl
 
WM
Van Wier
 
SA
, et al. 
Waldenstrom macroglobulinemia neoplastic cells lack immunoglobulin heavy chain locus translocations but have frequent 6q deletions.
Blood
2002
, vol. 
100
 (pg. 
2996
-
3001
)
51
Wong
 
KF
So
 
CC
Waldenstrom macroglobulinemia with karyotypic aberrations involving both homologous 6q.
Cancer Genet Cytogenet
2001
, vol. 
124
 (pg. 
137
-
139
)
52
Tabernero
 
MD
San Miguel
 
J
Garcia-Sanz
 
M
, et al. 
Incidence of chromosome numerical changes in multiple myeloma: fluorescence in situ hybridization analysis using 15 chromosome-specific probes.
Am J Clin Pathol
1996
, vol. 
149
 (pg. 
153
-
161
)
53
Primo
 
D
Flores
 
J
Quijano
 
S
, et al. 
Impact of BCR/ABL gene expression on the proliferative rate of different subpopulations of haematopoietic cells in chronic myeloid leukaemia.
Br J Haematol
2006
, vol. 
135
 (pg. 
43
-
51
)
54
Orfao
 
A
Ciudad
 
J
Gonzalez
 
M
, et al. 
Prognostic value of S-phase white blood cell count in B-cell chronic lymphocytic leukemia.
Leukemia
1992
, vol. 
6
 (pg. 
47
-
51
)
55
Quijano
 
S
Lopez
 
A
Rasillo
 
AI
, et al. 
Impact of trisomy 12, del(13q), del(17p) and del(11q) on the immunophenotype, DNA ploidy status and proliferative rate of leukemic B-cells in chronic lymphocytic leukemia.
Cytometry B Clin Cytom
2007
, vol. 
74B
 (pg. 
139
-
149
)
56
Saito
 
B
Shiozawa
 
E
Yamochi-Onizuka
 
T
, et al. 
Efficacy of rituximab plus chemotherapy in follicular lymphoma depends on Ki-67 expression.
Pathol Int
2004
, vol. 
54
 (pg. 
667
-
674
)
57
Kalogeraki
 
A
Tzardi
 
M
Panagiotides
 
I
, et al. 
MIB1 (Ki-67) expression in non-Hodgkin's lymphomas.
Anticancer Res
1997
, vol. 
17
 (pg. 
487
-
491
)
58
Romaguera
 
JE
Fayad
 
L
Rodriguez
 
MA
, et al. 
High rate of durable remissions after treatment of newly diagnosed aggressive mantle-cell lymphoma with rituximab plus hyper-CVAD alternating with rituximab plus high-dose methotrexate and cytarabine.
J Clin Oncol
2005
, vol. 
23
 (pg. 
7013
-
7023
)
59
Raty
 
R
Franssila
 
K
Joensuu
 
H
Teerenhovi
 
L
Elonen
 
E
Ki-67 expression level, histological subtype, and the International Prognostic Index as outcome predictors in mantle cell lymphoma.
Eur J Haematol
2002
, vol. 
69
 (pg. 
11
-
20
)
60
Bryant
 
RJ
Banks
 
PM
O'Malley
 
DP
Ki67 staining pattern as a diagnostic tool in the evaluation of lymphoproliferative disorders.
Histopathology
2006
, vol. 
48
 (pg. 
505
-
515
)
61
van Zelm
 
MC
van der Burg
 
M
de Ridder
 
D
, et al. 
Ig gene rearrangement steps are initiated in early human precursor B cell subsets and correlate with specific transcription factor expression.
J Immunol
2005
, vol. 
175
 (pg. 
5912
-
5922
)
62
Kuppers
 
R
Klein
 
U
Hansmann
 
ML
Rajewsky
 
K
Cellular origin of human B-cell lymphomas.
N Engl J Med
1999
, vol. 
341
 (pg. 
1520
-
1529
)
63
Dono
 
M
Cerruti
 
G
Zupo
 
S
The CD5+ B-cell.
Int J Biochem Cell Biol
2004
, vol. 
36
 (pg. 
2105
-
2111
)
64
Kuppers
 
R
la-Favera
 
R
Mechanisms of chromosomal translocations in B cell lymphomas.
Oncogene
2001
, vol. 
20
 (pg. 
5580
-
5594
)
65
Stevenson
 
FK
Sahota
 
SS
Ottensmeier
 
CH
, et al. 
The occurrence and significance of V gene mutations in B cell-derived human malignancy.
Adv Cancer Res
2001
, vol. 
83
 (pg. 
81
-
116
)
66
Liu
 
YJ
Arpin
 
C
de
 
BO
, et al. 
Sequential triggering of apoptosis, somatic mutation and isotype switch during germinal center development.
Semin Immunol
1996
, vol. 
8
 (pg. 
169
-
177
)
67
Shen
 
Y
Iqbal
 
J
Xiao
 
L
, et al. 
Distinct gene expression profiles in different B-cell compartments in human peripheral lymphoid organs.
BMC Immunol
2004
, vol. 
5
 pg. 
20
 
68
Good
 
KL
Tangye
 
SG
Decreased expression of Kruppel-like factors in memory B cells induces the rapid response typical of secondary antibody responses.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 (pg. 
13420
-
13425
)
69
Pellat-Deceunynck
 
C
Bataille
 
R
Normal and malignant human plasma cells: proliferation, differentiation, and expansions in relation to CD45 expression.
Blood Cells Mol Dis
2004
, vol. 
32
 (pg. 
293
-
301
)
70
Rajewsky
 
K
Clonal selection and learning in the antibody system.
Nature
1996
, vol. 
381
 (pg. 
751
-
758
)
71
Dogan
 
A
Isaacson
 
PG
Splenic marginal zone lymphoma.
Semin Diagn Pathol
2003
, vol. 
20
 (pg. 
121
-
127
)
72
Klein
 
U
Goossens
 
T
Fischer
 
M
, et al. 
Somatic hypermutation in normal and transformed human B cells.
Immunol Rev
1998
, vol. 
162
 (pg. 
261
-
280
)
73
De Rossi
 
G
Zarcone
 
D
Mauro
 
F
, et al. 
Adhesion molecule expression on B-cell chronic lymphocytic leukemia cells: malignant cell phenotypes define distinct disease subsets.
Blood
1993
, vol. 
81
 (pg. 
2679
-
2687
)
74
Callea
 
V
Morabito
 
F
Luise
 
F
, et al. 
Clinical significance of sIL2R, sCD23, sICAM-1, IL6 and sCD 14 serum levels in B-cell chronic lymphocytic leukemia.
Haematologica
1996
, vol. 
81
 (pg. 
310
-
315
)
75
Eistere
 
W
Hilbe
 
W
Stauder
 
R
, et al. 
An aggressive subtype of B-CLL is characterized by strong CD44 expression and lack of CD11c.
Br J Haematol
1996
, vol. 
93
 (pg. 
661
-
669
)
76
Beksac
 
M
Arat
 
M
Akan
 
H
, et al. 
Circulating CD44 and intercellular adhesion molecule-1 levels in low grade non-Hodgkin lymphoma and B-cell chronic lymphocytic leukemia patients during interferon-alpha-2a treatment.
Cancer
2000
, vol. 
89
 (pg. 
1474
-
1481
)
77
Barrena
 
S
Almeida
 
J
Yunta
 
M
, et al. 
Aberrant expression of tetraspanin molecules in B-cell chronic lymphoproliferative disorders and its correlation with normal B-cell maturation.
Leukemia
2005
, vol. 
19
 (pg. 
1376
-
1383
)
78
Angelopoulou
 
MK
Kontopidou
 
FN
Pangalis
 
GA
Adhesion molecules in B-chronic lymphoproliferative disorders.
Semin Hematol
1999
, vol. 
36
 (pg. 
178
-
197
)
79
Nadkarni
 
JJ
Perambakam
 
SM
Rathore
 
VB
, et al. 
Expression of adhesion molecules in B-cell chronic lymphocytic leukaemia: an analysis in lymphoid compartments: peripheral blood, bone marrow and lymph node.
Cancer Biother Radiopharm
1998
, vol. 
13
 (pg. 
269
-
274
)
80
Kimby
 
E
Rincon
 
J
Patarroyo
 
M
Mellstedt
 
H
Expression of adhesion molecules CD11/CD18 (Leu-CAMs, beta 2-integrins), CD54 (ICAM-1) and CD58 (LFA-3) in B-chronic lymphocytic leukemia.
Leuk Lymphoma
1994
, vol. 
13
 (pg. 
297
-
306
)
81
De Rossi
 
G
Zarcone
 
D
Mauro
 
F
, et al. 
Adhesion molecule expression on B-cell chronic lymphocytic leukemia cells: malignant cell phenotypes define distinct disease subsets.
Blood
1993
, vol. 
81
 (pg. 
2679
-
2687
)
82
Miyake
 
K
Underhill
 
CB
Lesley
 
J
Kincade
 
PW
Hyaluronate can function as a cell adhesion molecule and CD44 participates in hyaluronate recognition.
J Exp Med
1990
, vol. 
172
 (pg. 
69
-
75
)
83
Soma
 
LA
Craig
 
FE
Swerdlow
 
SH
The proliferation center microenvironment and prognostic markers in chronic lymphocytic leukemia/small lymphocytic lymphoma.
Hum Pathol
2006
, vol. 
37
 (pg. 
152
-
159
)
84
Swerdlow
 
SH
Murray
 
LJ
Habeshaw
 
JA
Stansfeld
 
AG
Lymphocytic lymphoma/B-chronic lymphocytic leukaemia–an immunohistopathological study of peripheral B lymphocyte neoplasia.
Br J Cancer
1984
, vol. 
50
 (pg. 
587
-
599
)
85
Ghia
 
P
Guida
 
G
Stella
 
S
, et al. 
The pattern of CD38 expression defines a distinct subset of chronic lymphocytic leukemia (CLL) patients at risk of disease progression.
Blood
2003
, vol. 
101
 (pg. 
1262
-
1269
)
86
Jaksic
 
O
Paro
 
MM
Kardum
 
SI
, et al. 
CD38 on B-cell chronic lymphocytic leukemia cells has higher expression in lymph nodes than in peripheral blood or bone marrow.
Blood
2004
, vol. 
103
 (pg. 
1968
-
1969
)
87
McDonnell
 
TJ
Deane
 
N
Platt
 
FM
, et al. 
bcl-2-immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation.
Cell
1989
, vol. 
57
 (pg. 
79
-
88
)
88
McDonnell
 
TJ
Korsmeyer
 
SJ
Progression from lymphoid hyperplasia to high-grade malignant lymphoma in mice transgenic for the t(14; 18).
Nature
1991
, vol. 
349
 (pg. 
254
-
256
)
89
Rosenwald
 
A
Wright
 
G
Wiestner
 
A
, et al. 
The proliferation gene expression signature is a quantitative integrator of oncogenic events that predicts survival in mantle cell lymphoma.
Cancer Cell
2003
, vol. 
3
 (pg. 
185
-
197
)
90
Sanchez-Beato
 
M
Saez
 
AI
Navas
 
IC
, et al. 
Overall survival in aggressive B-cell lymphomas is dependent on the accumulation of alterations in p53, p16, and p27.
Am J Pathol
2001
, vol. 
159
 (pg. 
205
-
213
)
91
Dogan
 
A
Bagdi
 
E
Munson
 
P
Isaacson
 
PG
CD10 and BCL-6 expression in paraffin sections of normal lymphoid tissue and B-cell lymphomas.
Am J Surg Pathol
2000
, vol. 
24
 (pg. 
846
-
852
)
92
Onizuka
 
T
Moriyama
 
M
Yamochi
 
T
, et al. 
BCL-6 gene product, a 92- to 98-kD nuclear phosphoprotein, is highly expressed in germinal center B cells and their neoplastic counterparts.
Blood
1995
, vol. 
86
 (pg. 
28
-
37
)
93
Ueda
 
C
Uchiyama
 
T
Ohno
 
H
Immunoglobulin (Ig)/BCL6 versus non-Ig/BCL6 gene fusion in diffuse large B-cell lymphoma corresponds to a high- versus low-level expression of BCL6 mRNA.
Blood
2002
, vol. 
99
 (pg. 
2624
-
2625
)
94
Artiga
 
MJ
Saez
 
AI
Romero
 
C
, et al. 
A short mutational hot spot in the first intron of BCL-6 is associated with increased BCL-6 expression and with longer overall survival in large B-cell lymphomas.
Am J Pathol
2002
, vol. 
160
 (pg. 
1371
-
1380
)
95
Cordone
 
I
Masi
 
S
Mauro
 
FR
, et al. 
p53 expression in B-cell chronic lymphocytic leukemia: a marker of disease progression and poor prognosis.
Blood
1998
, vol. 
91
 (pg. 
4342
-
4349
)
Sign in via your Institution