Monoclonal B-cell lymphocytosis (MBL) is classified as chronic lymphocytic leukemia (CLL)–like, atypical CLL, and CD5 MBL. The number of B cells per microliter divides CLL-like MBL into MBL associated with lymphocytosis (usually detected in a clinical setting) and low-count MBL detected in the general population (usually identified during population screening). After a median follow-up of 34 months we reevaluated 76 low-count MBLs with 5-color flow cytometry: 90% of CLL-like MBL but only 44.4% atypical CLL and 66.7% CD5 MBL persisted over time. Population-screening CLL-like MBL had no relevant cell count change, and none developed an overt leukemia. In 50% of the cases FISH showed CLL-related chromosomal abnormalities, including monoallelic or biallelic 13q deletions (43.8%), trisomy 12 (1 case), and 17p deletions (2 cases). The analysis of the T-cell receptor β (TRBV) chains repertoire showed the presence of monoclonal T-cell clones, especially among CD4highCD8low, CD8highCD4low T cells. TRBV2 and TRBV8 were the most frequently expressed genes. This study indicates that (1) the risk of progression into CLL for low-count population-screening CLL-like MBL is exceedingly rare and definitely lower than that of clinical MBL and (2) chromosomal abnormalities occur early in the natural history and are possibly associated with the appearance of the typical phenotype.

The presence of monoclonal B-cell expansions in the peripheral blood (PB) of asymptomatic persons has long been known.1  Multiparameter flow cytometry2,3  recently allowed to recognize a preclinical hematologic condition characterized by small B-cell clones detectable in the PB of otherwise healthy persons and named monoclonal B-cell lymphocytosis (MBL).1,4  Most (75%) MBLs have the phenotype (CD5+, CD23+, CD20low, sIglow) of chronic lymphocytic leukemia (CLL-like MBL). The remaining MBLs are classified as atypical CLL (CD5+, CD20bright) and CD5-negative (CD5) MBL.1,4  The prevalence of CLL-like MBL strikingly increases with age, reaching a frequency > 20% in persons > 60 years5,6  and being also more frequent among relatives of patients affected by CLL.7-10 

Because among the elderly the MBL frequency is 100 times higher than that of CLL, MBL might represent a “normal” aspect of the immune system, especially of the immunosenescence process. Therefore, the central problem of CLL-like MBL is to define how it relates to CLL,11  specifically to understanding which are the molecular features12-14  that make them a potential preleukemic condition12  and which is their clinical effect.

The International Workshop on Chronic Lymphocytic Leukemia guidelines15  indicate that the diagnosis is CLL-like MBL and not yet stage 0 CLL when the number of circulating clonal B cells is below the threshold of 5 × 109 cells/L. This brings in the issue that, within MBL, the number of circulating clonal B cells can be heterogeneous, and CLL-like MBL may be further divided into 2 different general subgroups, clinical MBL and population-screening MBL.4,12  Clinical MBL is usually diagnosed in a clinical setting, is associated with lymphocytosis, and has a concentration of clonal B cells > ∼ 1500/μL.13  On the contrary, population-screening MBL, also defined “low-count MBL,”14  is only detected during screening studies of healthy persons in the general population with the use of highly sensitive technical procedures and is characterized by < ∼ 50 clonal B cells/μL.4  The median absolute count of clonal B cells in population-screening MBL is 1 cell/μL, and in 75% of the cases the count is < 3 cells/μL.12  These numbers strikingly differ from the median concentration of clonal B cells in clinical MBL, which is ∼ 3000/μL, thus representing the majority of lymphocytes. Although the potential risk of progression of clinical MBL (previously defined MBL with lymphocytosis) into clinically overt CLL is ∼ 1.1% per year,13  limited data are available on the outcome of low-count population-screening MBL even if it is the most common form in the general population and the prevalence is ≥ 100-fold than the prevalence of CLL. Conceivably, the progression of population-screening MBL into CLL is uncommon,16  and most might simply be an epiphenomenon of immunosenescence, that is, an intensification of the progressive restriction of Ig and TCR repertoire observed in the elderly.17-20  It becomes therefore important to investigate which molecular and biologic features of low-count population-screening MBL are associated with the risk of progression to focus medical attention onto the rare risky subjects and to refrain from useless and prolonged monitoring most of the otherwise healthy persons.

To this end we took advantage of our cohort of 137 MBL cases, detected during a population screening of 1779 healthy persons > 18 years of age.14  More than one-half of the originally cases of diagnosed MBL were reevaluated after a median follow-up of ∼ 3 years. We here report that 90% of CLL-like MBL clones persisted over time, whereas atypical CLL and CD5 CLL MBL tended to be transient. None of the low-count population-screening MBLs developed an overt leukemia even if they were found to carry 13q deletions with the same frequency as CLL.21  This suggests that 13q deletion occurs early in the natural history of MBL/CLL and is probably associated with the typical CLL phenotype rather than with the progression into an overt disease.

Study population

The baseline study was conducted between 2005 and 2008 and enrolled 1779 healthy persons from Val Borbera, a rural valley in Northern Italy that is isolated from the surrounding areas by geographic barriers.14  The whole population is currently involved in a study not aimed at elucidating any particular health problem but rather at dissecting potential genetic components of common diseases because it is probable to derive from a limited number of common ancestors (ie, “genetically isolated”).

Among the initial participants we identified 138 cases of MBL; 1 male was affected by a splenic marginal zone lymphoma whose leukemic phase was detected by our analysis and was subsequently excluded from the study (final prevalence, 137 of 1779; 7.7%). Of the 137 originally diagnosed MBL subjects, 76 persons participated to a second visit after a median follow-up of 34 months (range, 11-50 months). Forty-five persons refused to take part in the follow-up visit, and 16 subjects were deceased, for unrelated causes, at the time of the second evaluation. The persons studied were > 18 years of age with a median age of 66 years (range, 25-92 years); 29 were women and 47 men. The study population has been carefully investigated from the clinical and genealogic point of view during the previous study. Clinical records were collected on all 76 persons who underwent routine blood tests, including a complete blood count with differential. No person had a known history of CLL. The follow-up evaluation was performed over 1 year (2009).

The research protocol was approved by the Institutional Ethics Committee at San Raffaele Scientific Institute, Milano, Italy, and all participants gave written informed consent in accordance with the Declaration of Helsinki.

Blood samples, cell preparation, staining, and FACS analysis

EDTA PB samples obtained from all persons enrolled were processed within 24 hours after blood withdrawal as previously described.14  The following antibody mixes were used: FITC-conjugated F(ab)2–anti-κ, PE–anti-λ light chain (Dako), PE-cyanin7 (Cy7)–labeled anti-CD20, PE-cyanin5 (Cy5)–conjugated anti-CD5 and PE-Texas Red–conjugated anti-CD19 (Beckman Coulter). For each sample, up to 500 000 events were acquired on a FC500 equipped with 488 argon ion laser and 635 red HeNe laser and analyzed with the CXP software system (Beckman Coulter) according to the gating strategy previously described.14  MBL has been divided into CLL-like MBL, atypical CLL MBL, and CD5 MBL, based on the immunophenotypic profile.1,4  The latter categories are based on the occurrence of an unbalanced κ/λ ratio (> 3:1 or < 1:3) within CD5+ or CD5 B lymphocytes, respectively. In contrast CLL-like MBL is defined on the basis of the distinct CD5brightCD20dim expression pattern on CD19+ B cells.14  Of the 54 persistent CLL-like MBL clones, 43 cases (79.6%) were clearly monoclonal, expressing κ light chains in 37 cases (68.5%) and λ light chain in 6 cases (11.1%); in 5 cases (9.3%) surface light chain expression was not detectable, this being suggestive of monoclonality,14  whereas the 6 remaining cases (11.1%) were polyclonal.14 

T-cell study

To study lymphocytes subpopulations the following antibodies mix has been used: CD25 FITC-labeled, CD4 PE-conjugated, and CD16 PE-Cy5–conjugated mixed with CD56 PE-Cy5–conjugated (Beckman Coulter). For each sample, up to 100 000 events were acquired and analyzed on FC500.

To study the repertoire of the TCR variable genes of the β chain (TRBV) used by T lymphocytes circulating in the blood of persons with CLL-like MBL, PB mononuclear cells have been stained with the “Io test Beta Mark TCR Vβ Repertoire kit” (Immunotech, Beckman Coulter), and a mix of PE-Cy5–conjugated anti-CD4 and PE-Cy7–labeled anti-CD8 (Beckman Coulter). T-cell subsets have been analyzed according to the previously described gating strategy, and a restricted TRBV expression was considered when the expression of a single TRBV chain was higher than the mean value plus 3 SDs (generally > 20%).17 

For quality control purposes, we daily used 0.4 mL of Flow-Check Fluorospheres (Beckman Coulter) mixed with 0.2 mL of Flow-Check 770 (Beckman Coulter PC7 [770/488] Setup Kit) to assess flow cytometric optical alignment and fluidics system. In addition, we daily controlled light scatter intensity, fluorescence intensity, and hydrodynamics with the use of 0.4 mL of Flow-Set Fluorospheres (Beckman Coulter) mixed with 0.2 mL of Flow-Set 770 (Beckman Coulter PC7 [770/488] Setup Kit) to assess optimal conditions for quantitative analysis of human leukocytes.

PCR amplification of IGHV-D-J rearrangements and sequence analysis

IGHV-D-J rearrangements have been analyzed in selected samples after DNA extraction, PCR amplification, and nucleotide sequencing as previously described.14  Systematic resequencing of Ig genes has not been performed because of paucity of material, which was mainly used to perform FISH analysis. A monoclonal IGHV-D-J rearrangement was determined in 3 additional cases in addition to those previously published14 : case VB094, expressing IGHV4-59/61 (95.6% of identity to germ line), case VB266, expressing IGHV3-21 (89.1% of identity); and case VB1698 expressing IGHV3-15 (93.0% of identity) genes, respectively.

FACS and FISH

CLL-like MBL cells have been purified on High Speed Sorter MoFLo (Dako). The following antibody mixes were used: PE-Cy7–labeled anti-CD20, PE-Cy5–conjugated anti-CD5 and PE-Texas Red–conjugated anti-CD19 (Beckman Coulter).

Sorted CD5+CD20dim monoclonal B cells underwent interphase FISH analysis after fixation in methanol/acetic acid (3:1), using standard protocols.22,23  The FISH panel included probes for the detection of trisomy 12, deletions of 11q22.3 (ATM), 13q14.3 (D13S319), 13q34 (LSI13q34), and 17p13 (TP53) (Vysis LSI p53/LSI ATM and LSI D13S319/LSI 13q34/CEP 12 Multi-color Probe; Abbott Molecular). Nikon Eclipse 90i microscope (Nikon Instruments) and a Qicam Fast 1394 (Qimaging) camera were used. Acquisition was done with a Genikon (Nikon Instruments S.p.a. Italy) software with the use of a FISH 3D Acquisition Module. When possible, FISH signals for each probe were scored in ≥ 50 interphase nuclei. Cutoffs were set on mean values ± 2 SDs, derived from analysis on healthy donor sorted B cells: Del17p, 6.7% ± 2.2%; Del11q, 3.4% ± 4.6%; Del13q14.3, 4.9% ± 10.8%; Del13q34, 5.4% ± 15.5%; and trisomy 12, 0.7% ± 2.3%.

Statistical methods

Median, mean, SD, maximum, minimum, 25th and 75th percentiles (for continuous variables), and relative frequencies (for categorical variables) were calculated with SPSS software (SPSS 16.0; SPSS Inc). To define statistically significant differences between subgroups, Mann-Whitney test and Fisher exact test were used for continuous and categorical variables, respectively. All P values were 2-sided and regarded as statistically significant only if < .05.

Low-count population-screening CLL-like MBL persists over time

Between 2005 and 2008 we enrolled 1779 healthy persons, and flow cytometry detected the presence of MBL in 137 subjects (128 previously described14 ), 96 CLL-like MBL, 21 atypical-CLL MBL, and 20 CD5 MBL.1 

After a median follow-up of 34 months (range, 11-50 months), we reanalyzed the PB of 76 subjects (47 men, 29 women; median age, 66 years; range, 25-92 years). Sixteen subjects with MBL had died of unrelated reasons, and the remaining subjects were not available for the follow-up study. All subjects had a normal absolute white blood cell count (mean value, 7.0 × 109 cells/L; range 2.7-12.8 × 109 cells/L) and a normal absolute lymphocyte count (ALC; mean value, 2.4 × 109 cells/L; range, 0.8-3.9 × 109 cells/L), except for 4 cases whose lymphocyte counts were slightly above the value of 4.0 × 109 cells/L (range, 4.1-5.3 × 109 cells/L). The mean absolute B-cell count was 184.5 × 106 cells/L (range, 18.2-1508.8 × 106 cells/L). Among the 76 reevaluated subjects, 60 originally had a CLL-like MBL, 9 an atypical-CLL MBL, and 9 a CD5 MBL. Two subjects with MBL had a double clone.

At reevaluation 54 of 60 CLL-like MBL (90.0%), but only 4 of 9 atypical-CLL MBL (44.4%) and 6 of 9 CD5 MBL (66.7%) clones were still detectable in the same original carriers (Figure 1). The 6 transient CLL-like MBL clones were originally present in small numbers (median concentration of aberrant B cells, 0.40/μL) close to the detection limit of the acquisition protocol used. The 2 subjects with double clones were reconfirmed.

Figure 1

CLL-like MBL remains stable over time. Percentages of persistent or transient clones at follow-up analysis in CLL-like, atypical-CLL, and CD5 MBL. At the first evaluation the mean clonal B-cell count of persistent CLL-like MBL was 50.6 cells/μL, whereas transient CLL-like clones had a mean value of 0.40 cells/μL.

Figure 1

CLL-like MBL remains stable over time. Percentages of persistent or transient clones at follow-up analysis in CLL-like, atypical-CLL, and CD5 MBL. At the first evaluation the mean clonal B-cell count of persistent CLL-like MBL was 50.6 cells/μL, whereas transient CLL-like clones had a mean value of 0.40 cells/μL.

Close modal

Among the persistent MBL, 61 of 64 (95.3%) had a normal ALC (mean value, 2.4 × 109 cells/L; range, 0.8-3.9 × 109 cells/L). In the remaining 3 cases, ALCs were slightly above the 4.0 × 109 cells/L value (4.1, 4.3, and 5.3 × 109 cells/L). The mean absolute B-cell count of the persistent MBL was 190.8 × 106 cells/L (range, 21.6-1508.8 × 106 cells/L), and clonal B cells had a mean value of 62.3 × 106 cells/L (range, 0.2-1249.3 × 106 cells/L).

Population-screening CLL-like MBL remains stable over time

Among the 54 CLL-like persistent cases, 35 were men and 19 were women, with a median age of 66 years (range, 40-92 years). One case (VB094) was excluded from further analysis because of being treated with chemotherapy for a solid tumor.

The mean white blood cell count of the 53 CLL-like MBL cases was 7.1 × 109 cells/L (range, 2.7-12.8 × 109 cells/L), the mean ALC was 2.5 × 109 cells/L (range, 0.8-4.1 × 109 cells/L), and the mean absolute B-cell count was 197.0 × 109 cells/L (range, 20.8-1508.8 × 109 cells/L). The clonal CD19+CD5+ B cells were 51.4 × 109 cells/L (range, 0.2-1249.3 × 109 cells/L). All values were not significantly different from the previous assessment after a median follow-up of 34 months (Figure 2A-B). None of the CLL-like clones developed a clinically relevant lymphocytosis; one case only had 4.1 × 109 lymphocytes/L, decreasing from 5.3 × 109 cells/L at the original evaluation.

Figure 2

B-cell count and absolute clone values in persistent CLL-like MBL are not significantly different between first and follow-up evaluations. The box-plots show the distribution of B-cell count (A) and absolute clone (B) values in persistent CLL-like MBL at follow-up evaluation. Notched boxes represent 25th and 75th percentile values, and the line in the middle corresponds to median value. Vertical lines represent the highest and the lowest values that are not outliers or extreme values. The differences between the 2 time points are not statistically significant.

Figure 2

B-cell count and absolute clone values in persistent CLL-like MBL are not significantly different between first and follow-up evaluations. The box-plots show the distribution of B-cell count (A) and absolute clone (B) values in persistent CLL-like MBL at follow-up evaluation. Notched boxes represent 25th and 75th percentile values, and the line in the middle corresponds to median value. Vertical lines represent the highest and the lowest values that are not outliers or extreme values. The differences between the 2 time points are not statistically significant.

Close modal

When we considered the size of the clone, 48 of 53 clones had a clonal cell count of ≤ 50 aberrant B cells/μL (mean value, 3.6 cells/μL; range, 0.2-50.4 cells/μL) and represented only a small proportion of total B-cell count (mean, 2.8%; range, 0.1%-33.5%). Of the 5 of 53 remaining cases, 4 had < 500 cells/μL (247.1, 319.1, 336.7, 399.3 cells/μL) and 1 had < 1500 cells/μL (1249.3 cells/μL), all below the characteristic limit of clinical MBL.

After a median follow-up of 34 months, none of the CLL-like clones showed relevant changes in the cell count (Figure 2B). In particular, among the 48 cases with < 50 clonal B cells/μL, the mean difference between the first and the follow-up evaluation was < 1 cell/μL (0.93).

Low-count population-screening CLL-like MBL has recurrent CLL-related genomic aberrations

We sorted clonal B cells from 16 low-count population-screening CLL-like MBLs with FACS (median value, 5.3 cells/μL) and performed FISH analysis (Table 1) to evaluate the 4 most frequent aberrations detected in patients with CLL (del13q, trisomy 12, del11q, and del17p).21 

Table 1

CLL-like clones molecular parameters

IDSexAge, yFollow-up, moClone, cells/μL
Light chainFISHIGHV geneIGHV identity, %T-cell clones
First visitFollow-upCD4+CD8+CD4highCD8lowCD4lowCD8high
VB0010 65 50 5.35 6.10 κ ND IGHV3-30 97.7 No clones No clones TRBV3 = 22.4% TRBV7.1 = 22.4% TRBV8 = 33.9% TRBV13.6 = 32.1% No clones 
VB0094* 73 48 16.11 1.63 κ Normal IGHV4-59/-61 95.6 No clones No clones TRBV8 = 23.1% No clones 
VB0106 83 48 1.03 0.35 λ ND IGHV3-30 97.8 ND ND ND ND 
VB0266 74 47 6.56 8.16 κ ND IGHV3-21 89.1 No clones No clones TRBV2 = 25.3% TRBV8 = 27.4% No clones 
VB0274 67 47 7.26 11.26 κ ND IGHV4-59/-61 95.0 No clones TRBV9 = 55.0% No clones No clones 
VB0314 78 47 5.79 4.82 λ Del13q14.3 (19%), trisomy 12 (28%) IGHV4-59/-61 94.4 No clones No clones No clones No clones 
VB0323 75 47 0.51 0.61 κ ND IGHV4-34 97.7 ND ND ND ND 
VB0399 66 46 0.74 0.22 κ ND IGHV3-48 93.9 No clones No clones No clones TRBV17 = 21.4% 
VB0400 65 46 0.61 0.32 κ ND IGHV3-64 86.8 No clones No clones No clones TRBV2 = 22.2% 
VB0457 83 46 0.44 2.22 κ Normal IGHV3-23 100.0 No clones TRBV8 = 28.4% TRBV2 = 21.1% TRBV22 = 21.4% 
VB0537 74 47 1.79 0.75 κ ND IGHV3-30 97.5 ND ND ND ND 
VB0588 65 39 96.73 319.10 κ Del13q14.3 (31%) IGHV4-59 93.5 No clones No clones TRBV5.2 = 21.5% No clones 
VB0643 40 39 0.88 0.61 κ ND IGHV3-64 93.9 No clones No clones TRBV22 = 50.8% No clones 
VB0656 49 39 0.88 0.46 κ ND IGHV3-7 97.0 No clones No clones No clones No clones 
VB0667 65 38 0.24 0.68 κ Normal ND ND No clones No clones No clones TRBV22 = 26.1% 
VB0670 68 37 0.24 0.21 κ ND ND ND ND ND ND ND 
VB0679 67 38 6.34 9.25 λ ND IGHV4-59/-61 89.4 No clones No clones TRBV7.2 = 39.4% TRBV8 = 30.1% No clones 
VB0689 60 38 221.33 247.09 κ Normal IGHV3-73 92.6 No clones No clones TRBV2 = 25.7% No clones 
VB0777 68 37 0.66 0.79 κ ND ND ND ND ND ND ND 
VB0790 61 36 1.50 13.55 κ Normal IGHV4-30-4 94.4 No clones No clones No clones TRBV13.1 = 48.1% 
VB0819 85 37 0.19 0.46 κ ND IGHV3-48 88.3 ND ND ND ND 
VB0902 69 36 0.96 6.76 κ Normal IGHV4-39 100.0 No clones TRBV2 = 35.7% No clones No clones 
VB1002 63 35 1.67 1.60 κ Del17p (80%) IGHV4-59/-61 97.0 No clones No clones TRBV9 = 24.8% TRBV16 = 38.9% TRBV5.3 = 32.6% TRBV16 = 38.3% 
VB1091 46 35 1.12 1.61 Polyclonal ND IGHV3-15 93.5 No clones TRBV18 = 37.5% TRBV2 = 30.2% TRBV8 = 19.4% No clones 
VB1134 58 34 2.36 0.46 κ Del13q34 (25%) ND ND No clones No clones No clones TRBV13.6 = 22.2% 
VB1135 62 35 0.35 0.35 Negative ND ND ND ND ND ND ND 
VB1170 58 34 0.85 0.33 κ ND ND ND No clones No clones TRBV2 = 30.2% TRBV17 = 21.1% TRBV2 = 25.0% TRBV22 = 21.7% 
VB1189 80 34 189.76 399.29 κ Normal IGHV3-21 88.2 No clones TRBV8 = 23.1% TRVB13.6 = 42.8% No clones 
VB1202 61 34 4.72 18.56 κ Del13q14.3 (48%) ND ND No clones No clones No clones No clones 
VB1231 64 35 50.38 50.59 κ ND IGHV3-64 100.0 No clones No clones No clones TRBV1 = 22.7% 
VB1239 75 34 0.41 0.50 κ ND IGHV3-23 92.0 ND ND ND ND 
VB1240 84 31 1763.90 1249.29 λ Normal IGHV3-15 98.3 No clones No clones No clones No clones 
VB1309 59 24 0.50 0.56 Negative ND ND ND ND ND ND ND 
VB1320 61 24 0.41 3.82 λ Del13q14.3-q34 (25% and 30%), Del17p (90%) ND ND No clones No clones TRBV5.3 = 35.7% TRBV8 = 48.4% TRBV17 = 33.8% TRBV16 = 32.1% 
VB1349 72 24 0.75 1.11 κ ND ND ND No clones TRBV1 = 61.5% No clones TRBV17 = 21.4% 
VB1377 68 24 3.78 2.42 κ Del13q34 (28%) ND ND No clones No clones No clones No clones 
VB1378 77 25 0.54 0.42 κ ND ND ND ND ND ND ND 
VB1393 75 25 0.55 1.84 κ ND ND ND No clones TRBV13.2 = 25.8% No clones No clones 
VB1395 76 25 1.53 1.22 Polyclonal ND ND ND No clones No clones No clones No clones 
VB1451 66 23 0.46 2.36 κ ND ND ND No clones No clones TRBV2 = 45.7% No clones 
VB1470 73 23 0.94 4.02 κ Del13q14.3 (37%) ND ND No clones TRBV13.2 = 38.3% TRBV16 = 40.3% TRBV21.3 = 42.1% No clones 
VB1484 64 24 0.25 0.73 Polyclonal ND ND ND ND ND ND ND 
VB1487 84 23 0.06 0.53 κ ND IGHV4-59/-61 94.1 No clones No clones No clones TRBV7.1 = 46.4% TRBV8 = 29.4% TRBV17 = 26.9% TRBV22 = 28.6% 
VB1518 53 22 1.21 1.16 Negative ND ND ND ND ND ND ND 
VB1523 60 22 0.84 1.54 Polyclonal ND ND ND No clones No clones No clones No clones 
VB1531 61 23 0.50 0.75 Negative ND ND ND ND ND ND ND 
VB1592 92 21 0.29 0.20 Polyclonal ND IGHV3-64 98.1 ND ND ND ND 
VB1646 62 22 9.03 4.84 Negative ND ND ND No clones No clones No clones No clones 
VB1686 83 22 0.96 1.31 κ ND ND ND No clones No clones No clones No clones 
VB1688 55 21 1.22 1.07 Polyclonal ND IGHV3-21 95.1 ND ND ND ND 
VB1698 48 21 264.98 336.74 λ ND IGHV3-15 93.0 No clones No clones TRBV2 = 42.2% TRBV17 = 42.2% 
VB1711 56 11 0.41 0.32 κ ND ND ND ND ND ND ND 
VB1712 72 11 0.35 1.53 κ ND ND ND ND ND ND ND 
VB1736 48 11 0.68 0.26 Kappa ND ND ND ND ND ND ND 
IDSexAge, yFollow-up, moClone, cells/μL
Light chainFISHIGHV geneIGHV identity, %T-cell clones
First visitFollow-upCD4+CD8+CD4highCD8lowCD4lowCD8high
VB0010 65 50 5.35 6.10 κ ND IGHV3-30 97.7 No clones No clones TRBV3 = 22.4% TRBV7.1 = 22.4% TRBV8 = 33.9% TRBV13.6 = 32.1% No clones 
VB0094* 73 48 16.11 1.63 κ Normal IGHV4-59/-61 95.6 No clones No clones TRBV8 = 23.1% No clones 
VB0106 83 48 1.03 0.35 λ ND IGHV3-30 97.8 ND ND ND ND 
VB0266 74 47 6.56 8.16 κ ND IGHV3-21 89.1 No clones No clones TRBV2 = 25.3% TRBV8 = 27.4% No clones 
VB0274 67 47 7.26 11.26 κ ND IGHV4-59/-61 95.0 No clones TRBV9 = 55.0% No clones No clones 
VB0314 78 47 5.79 4.82 λ Del13q14.3 (19%), trisomy 12 (28%) IGHV4-59/-61 94.4 No clones No clones No clones No clones 
VB0323 75 47 0.51 0.61 κ ND IGHV4-34 97.7 ND ND ND ND 
VB0399 66 46 0.74 0.22 κ ND IGHV3-48 93.9 No clones No clones No clones TRBV17 = 21.4% 
VB0400 65 46 0.61 0.32 κ ND IGHV3-64 86.8 No clones No clones No clones TRBV2 = 22.2% 
VB0457 83 46 0.44 2.22 κ Normal IGHV3-23 100.0 No clones TRBV8 = 28.4% TRBV2 = 21.1% TRBV22 = 21.4% 
VB0537 74 47 1.79 0.75 κ ND IGHV3-30 97.5 ND ND ND ND 
VB0588 65 39 96.73 319.10 κ Del13q14.3 (31%) IGHV4-59 93.5 No clones No clones TRBV5.2 = 21.5% No clones 
VB0643 40 39 0.88 0.61 κ ND IGHV3-64 93.9 No clones No clones TRBV22 = 50.8% No clones 
VB0656 49 39 0.88 0.46 κ ND IGHV3-7 97.0 No clones No clones No clones No clones 
VB0667 65 38 0.24 0.68 κ Normal ND ND No clones No clones No clones TRBV22 = 26.1% 
VB0670 68 37 0.24 0.21 κ ND ND ND ND ND ND ND 
VB0679 67 38 6.34 9.25 λ ND IGHV4-59/-61 89.4 No clones No clones TRBV7.2 = 39.4% TRBV8 = 30.1% No clones 
VB0689 60 38 221.33 247.09 κ Normal IGHV3-73 92.6 No clones No clones TRBV2 = 25.7% No clones 
VB0777 68 37 0.66 0.79 κ ND ND ND ND ND ND ND 
VB0790 61 36 1.50 13.55 κ Normal IGHV4-30-4 94.4 No clones No clones No clones TRBV13.1 = 48.1% 
VB0819 85 37 0.19 0.46 κ ND IGHV3-48 88.3 ND ND ND ND 
VB0902 69 36 0.96 6.76 κ Normal IGHV4-39 100.0 No clones TRBV2 = 35.7% No clones No clones 
VB1002 63 35 1.67 1.60 κ Del17p (80%) IGHV4-59/-61 97.0 No clones No clones TRBV9 = 24.8% TRBV16 = 38.9% TRBV5.3 = 32.6% TRBV16 = 38.3% 
VB1091 46 35 1.12 1.61 Polyclonal ND IGHV3-15 93.5 No clones TRBV18 = 37.5% TRBV2 = 30.2% TRBV8 = 19.4% No clones 
VB1134 58 34 2.36 0.46 κ Del13q34 (25%) ND ND No clones No clones No clones TRBV13.6 = 22.2% 
VB1135 62 35 0.35 0.35 Negative ND ND ND ND ND ND ND 
VB1170 58 34 0.85 0.33 κ ND ND ND No clones No clones TRBV2 = 30.2% TRBV17 = 21.1% TRBV2 = 25.0% TRBV22 = 21.7% 
VB1189 80 34 189.76 399.29 κ Normal IGHV3-21 88.2 No clones TRBV8 = 23.1% TRVB13.6 = 42.8% No clones 
VB1202 61 34 4.72 18.56 κ Del13q14.3 (48%) ND ND No clones No clones No clones No clones 
VB1231 64 35 50.38 50.59 κ ND IGHV3-64 100.0 No clones No clones No clones TRBV1 = 22.7% 
VB1239 75 34 0.41 0.50 κ ND IGHV3-23 92.0 ND ND ND ND 
VB1240 84 31 1763.90 1249.29 λ Normal IGHV3-15 98.3 No clones No clones No clones No clones 
VB1309 59 24 0.50 0.56 Negative ND ND ND ND ND ND ND 
VB1320 61 24 0.41 3.82 λ Del13q14.3-q34 (25% and 30%), Del17p (90%) ND ND No clones No clones TRBV5.3 = 35.7% TRBV8 = 48.4% TRBV17 = 33.8% TRBV16 = 32.1% 
VB1349 72 24 0.75 1.11 κ ND ND ND No clones TRBV1 = 61.5% No clones TRBV17 = 21.4% 
VB1377 68 24 3.78 2.42 κ Del13q34 (28%) ND ND No clones No clones No clones No clones 
VB1378 77 25 0.54 0.42 κ ND ND ND ND ND ND ND 
VB1393 75 25 0.55 1.84 κ ND ND ND No clones TRBV13.2 = 25.8% No clones No clones 
VB1395 76 25 1.53 1.22 Polyclonal ND ND ND No clones No clones No clones No clones 
VB1451 66 23 0.46 2.36 κ ND ND ND No clones No clones TRBV2 = 45.7% No clones 
VB1470 73 23 0.94 4.02 κ Del13q14.3 (37%) ND ND No clones TRBV13.2 = 38.3% TRBV16 = 40.3% TRBV21.3 = 42.1% No clones 
VB1484 64 24 0.25 0.73 Polyclonal ND ND ND ND ND ND ND 
VB1487 84 23 0.06 0.53 κ ND IGHV4-59/-61 94.1 No clones No clones No clones TRBV7.1 = 46.4% TRBV8 = 29.4% TRBV17 = 26.9% TRBV22 = 28.6% 
VB1518 53 22 1.21 1.16 Negative ND ND ND ND ND ND ND 
VB1523 60 22 0.84 1.54 Polyclonal ND ND ND No clones No clones No clones No clones 
VB1531 61 23 0.50 0.75 Negative ND ND ND ND ND ND ND 
VB1592 92 21 0.29 0.20 Polyclonal ND IGHV3-64 98.1 ND ND ND ND 
VB1646 62 22 9.03 4.84 Negative ND ND ND No clones No clones No clones No clones 
VB1686 83 22 0.96 1.31 κ ND ND ND No clones No clones No clones No clones 
VB1688 55 21 1.22 1.07 Polyclonal ND IGHV3-21 95.1 ND ND ND ND 
VB1698 48 21 264.98 336.74 λ ND IGHV3-15 93.0 No clones No clones TRBV2 = 42.2% TRBV17 = 42.2% 
VB1711 56 11 0.41 0.32 κ ND ND ND ND ND ND ND 
VB1712 72 11 0.35 1.53 κ ND ND ND ND ND ND ND 
VB1736 48 11 0.68 0.26 Kappa ND ND ND ND ND ND ND 

ND indicates not done.

*

Case excluded from further analysis because treated with chemotherapy for a solid tumor.

Biallelic Del13q14.3.

In 8 of 16 subjects analyzed (50.0%) we found genomic abnormalities (Table 1). Aberrations involving the chromosome 13 long arm were the most frequent (7 of 16 cases; 43.8%), 5 cases (31.3%) had a deletion of 13q14.3 that in 2 subjects was biallelic; 2 subjects (12.5%) had only a deletion of 13q34 (Figure 3A), and one had both 13q34 and 13q14.3 regions deleted. The percentage of CLL-like cells carrying these abnormalities was 19%-48%. One case had trisomy 12 in 28% of isolated B cells (Figure 3B) and 2 subjects had a deletion of the 17p region in 80% and 90% of sorted CLL-like cells (Figure 3C), respectively. More than 1 cytogenetic abnormality was identified in 2 cases (12.5%); one presented concomitant trisomy 12 and del13q14.3, the other one showed del13q34/13q14.3 and del17p. Deletion of 11q was never detected. The distribution of the cytogenetic abnormalities was not different between κ and λ clones.

Figure 3

Genomic aberrations are frequently detected by FISH in CLL-like MBL clones. FISH analysis has been performed on CD5+CD20dim purified MBL cells and shows, (A) on the left, a normal nucleus with 2 signals for each probe (Spectrum Aqua, Orange, and Green) corresponding to LSI13q34 locus 13q34, LSI D13S319 locus 13q14.3, and CEP 12 locus 12p11.1-q11, respectively; on the right another nucleus with a heterozygous deletion of the LSI D13S319 locus 13q14.3 (1 Spectrum Orange signal and 2 normal Spectrum Aqua and 2 Spectrum Green signals). (B) Both nuclei show trisomy of CEP 12 locus 12p11.1-q11 (3 Spectrum Green signals) and 2 normal Spectrum Orange signals (LSI D13S319 locus 13q14.3). (C) Both nuclei show the presence of only one signal for LSI p53 locus 17p13.1 (Spectrum Orange) and a normal double signal for LSI ATM locus 11q22.3 (Spectrum Green). Microscope used for these images was Nikon Eclipse 90i with a magnification ×1000 (Objective PLAN APO VC 100X/1:40 oil; 10× ocular; room temperature), and camera used was Qicam Fast 1394 (Qimaging). The imaging medium used was DAPI (Vectashield Mounting Medium with DAPI, vector). Acquisition software was Genikon (Nikon Instruments S.P.A.) and a FISH 3D Acquisition Module was used.

Figure 3

Genomic aberrations are frequently detected by FISH in CLL-like MBL clones. FISH analysis has been performed on CD5+CD20dim purified MBL cells and shows, (A) on the left, a normal nucleus with 2 signals for each probe (Spectrum Aqua, Orange, and Green) corresponding to LSI13q34 locus 13q34, LSI D13S319 locus 13q14.3, and CEP 12 locus 12p11.1-q11, respectively; on the right another nucleus with a heterozygous deletion of the LSI D13S319 locus 13q14.3 (1 Spectrum Orange signal and 2 normal Spectrum Aqua and 2 Spectrum Green signals). (B) Both nuclei show trisomy of CEP 12 locus 12p11.1-q11 (3 Spectrum Green signals) and 2 normal Spectrum Orange signals (LSI D13S319 locus 13q14.3). (C) Both nuclei show the presence of only one signal for LSI p53 locus 17p13.1 (Spectrum Orange) and a normal double signal for LSI ATM locus 11q22.3 (Spectrum Green). Microscope used for these images was Nikon Eclipse 90i with a magnification ×1000 (Objective PLAN APO VC 100X/1:40 oil; 10× ocular; room temperature), and camera used was Qicam Fast 1394 (Qimaging). The imaging medium used was DAPI (Vectashield Mounting Medium with DAPI, vector). Acquisition software was Genikon (Nikon Instruments S.P.A.) and a FISH 3D Acquisition Module was used.

Close modal

Expansions of monoclonal T-cell populations are frequent in CLL-like MBL

In the hypothesis that CLL-like MBL may coexist with additional abnormalities of the immune system, we investigated also the TRBV with a panel of TRBV antibodies covering 70% of the expressed TCR repertoire17  in 36 cases of MBLs (20 men and 16 women, 14 below and 22 above the age of 65 years; Table 1).

We focused the analysis on CD4+, CD8+, and CD4+/CD8+ double-positive T lymphocytes, the latter being further divided into CD4highCD8low and CD4lowCD8high T cells (Figure 4A,C).17  On the basis of the previously published cutoff value of 20% of the cells expressing any TCR,17  we found ≥ 1 T-cell clone in 27 subjects (27 of 36; 75%) with a total of 53 clones detected.

Figure 4

T-cell clones in persons carrying CLL-like MBL. (A) PB T lymphocytes from a representative case (VB0902) are subdivided, based on CD4 and CD8 expression, in 4 subpopulations: CD8+ (gate A), CD4lowCD8high (gate B), CD4highCD8low (gate C), and CD4+ (gate D). (B) CD8+ T lymphocytes (gated through A) of subject VB0902 show a restricted expression of TRBV2 (35.7%, upper right quadrant). (C) T-cell subpopulations in case VB1002. (D) CD4highCD8low T lymphocytes in this second subject (gated through C) show restricted expression for distinct TRBV chain (TRBV9, 24.8%, upper left; and TRBV16, 38.9%, lower right), indicating the presence of 2 distinct clones. In the TCR-Vβ repertoire kit each tube includes a FITC-conjugated mAb, a PE-conjugated mAb, and a third mAb derived from a balanced mixture of a PE- and a FITC-conjugated form. Because of the mutually exclusive expression of each TRBV gene, the “virtual” double-positive population on the upper left quadrant represents a third different TRBV domain.

Figure 4

T-cell clones in persons carrying CLL-like MBL. (A) PB T lymphocytes from a representative case (VB0902) are subdivided, based on CD4 and CD8 expression, in 4 subpopulations: CD8+ (gate A), CD4lowCD8high (gate B), CD4highCD8low (gate C), and CD4+ (gate D). (B) CD8+ T lymphocytes (gated through A) of subject VB0902 show a restricted expression of TRBV2 (35.7%, upper right quadrant). (C) T-cell subpopulations in case VB1002. (D) CD4highCD8low T lymphocytes in this second subject (gated through C) show restricted expression for distinct TRBV chain (TRBV9, 24.8%, upper left; and TRBV16, 38.9%, lower right), indicating the presence of 2 distinct clones. In the TCR-Vβ repertoire kit each tube includes a FITC-conjugated mAb, a PE-conjugated mAb, and a third mAb derived from a balanced mixture of a PE- and a FITC-conjugated form. Because of the mutually exclusive expression of each TRBV gene, the “virtual” double-positive population on the upper left quadrant represents a third different TRBV domain.

Close modal

No CD4+ clones were found; 8 clones were observed among CD8+ cells (Figure 4B). The double-positive T-cell subsets were present in low percentages with CD4highCD8low cells representing 0.96% of lymphocytes and CD8highCD4low cells 0.24% of lymphocytes. As expected,17  these 2 subsets presented the highest amount of clones with 16 of 36 (44.4%) carrying ≥ 1 clonal population of the CD4highCD8low (Figure 4D) and 13 of 36 (36.1%) of the CD8highCD4low compartment.

Overall, 27 subjects presented ≥ 1 T-cell clonal expansions (range, 1-4 T-cell clonal expansions), of whom 17 of 22 subjects (77.3%) > 65 years of age and 10 of 14 (71.4%) < 65 years. Both frequencies were significantly higher (P = .002) than those observed in the general population.17 

TRBV2 (11 of 27; 40.7%) and TRBV8 (9 of 27; 33.3%) were the most frequent genes in the clonal populations. Both genes were more frequent in the MBL population than in the T-cell clones detected in the general population (TRBV2, 18.8%; TRBV8, 7.8%).17 

Not all MBLs are equal. We have previously found that MBLs identified through population-screening studies,14  with aberrant B cells representing a tiny minority of total B cells, have different biologic and laboratory features compared with clinical MBL associated with lymphocytosis.12  Clinical CLL-like MBL has a risk of progression into overt CLL of ∼ 1.1% per year,13  whereas the progression risk of low-count population-screening MBL is unknown. This is an important issue because the prevalence of population-screening MBL is much higher than that of clinical MBL with a peak of > 75% in persons > 90 years.5  To investigate which features are associated with the risk of progression we have reevaluated, after a median follow-up of 34 months, 76 subjects with MBL previously detected in the PB of a general adult population belonging to a rural community in Northern Italy. We here show that (1) 90.0% of population-screening CLL-like MBL persists over time in contrast to only 44.4% and 66.7% of atypical-CLL and CD5 MBL; (2) most of the population-screening MBLs remain stable, and no progression to clinically overt disease has been recorded; and (3) 50% of population-screening CLL-like MBL carries CLL-related genomic abnormalities, especially monoallelic or biallelic 13q deletions.

The fact that CLL-like MBL clones persist over time strongly implies that a stable modification has occurred within the B lineage and that these clones are not simply related to seasonal or occasional antigenic exposures. Because approximately one-half (44.4% and 66.7%) of atypical-CLL and CD5 MBL became undetectable after 3 years, MBL other than CLL-like may be transient. A plausible possibility is that they are “reactive” monoclonal expansions, conceivably triggered by self-limiting immune stimulations that occur randomly during one's lifetime. Our results differ from those recently published in a series of 12 atypical-CLL and CD5 MBL reevaluated 12 months after the first immunophenotypic analysis.24  All clones were confirmed and even showed a significant increase in the median concentration of clonal B cells. Although this discrepancy may be accounted for both by the original higher median value of the clonal populations and especially by the different length of the follow-up, further studies with larger cohorts are needed to settle the issue.

The persistence of CLL-like MBL closely resembles that of clonal double-positive CD4/CD8 T-cell populations in the elderly, linked to chronic and persistent viral infections (eg, CMV).25,26  Whether persistent stimulation by infectious agents (or self-antigens) may also trigger the development of MBL remains to be elucidated, although the increased frequency of CLL-like MBL in HCV-infected persons27  is in line with this possibility. Clonal T-cell populations, especially double-positive CD4/CD8, were found to be increased in population-screening MBL compared with the general population,17  with TRBV2 and TRBV8 as the most frequently involved genes, possibly suggesting a distinct restriction of T-cell specificity. Finally, more than one-half of the subjects analyzed presented multiple T-cell clones, suggesting a widespread deregulation of the immune system in MBL, another feature potentially related to a prolonged exposure to chronic antigenic stimulation.

After a median follow-up of ∼ 3 years, none of the 76 participants have developed CLL or any other lymphoproliferative disorder, and no one has required medical attention because of the development of clinically relevant lymphocytosis. Most subjects with population-screening MBL remained stable, some even decreased. These results confirm the hypothesis that the natural history of population-screening MBL differs from that of clinical MBL and are in line with the molecular and biologic differences we previously demonstrated.14  Low-count population-screening CLL-like MBL uses an IGHV gene repertoire different from clinical MBL and CLL, shows a distinct gene usage, a higher rate of mutated IGHV, and a rare occurrence of stereotyped receptors.14  On the basis of these evidences, molecular features of the clone could be potentially used to identify those rare subjects at risk of evolution into frank CLL and refrain from useless and prolonged monitoring in most of otherwise healthy persons. To our surprise, in the present work, when we studied by FISH analysis the presence of CLL-related genetic abnormalities, we found that ≤ 50% of cases with low-count population-screening MBL do carry the same CLL-related abnormalities with monoallelic or biallelic 13q deletions in 43.8% of the cases. We also observed trisomy 12 in 1 case and 17p deletion in 2 cases (in 1 case as a sole abnormality). The frequency of 13q deletions is similar to that observed in newly diagnosed CLL and in subjects with clinical MBL5,13,21  and strongly indicates that these lesions occur rather early during the development of MBL and appear to be associated with the acquisition of the typical immunophenotypic profile rather than with the fully fledged leukemic state.16  This finding is in keeping with the observation that the mouse model carrying deletions of the DLEU2/mir15-a/16-1 cluster located at 13q14 region develops a spectrum of lymphoproliferative disorders that range from MBL to CLL to diffuse large B-cell lymphoma.28 

Of interest we also detected 17p deletion in 2 cases of population-screening MBL with low numbers of clonal B cells (1.6 and 3.8 cells/μL, respectively); del17p was observed in 80% and 90% of clonal B cells, and in case VB1320 it was coexisting with an additional genetic alteration involving chromosome 13q, but the increase of aberrant B cells was minor at reevaluation (from 0.4 to 3.8 cells/μL, respectively). The detection of 17p deletion per se may not be detrimental because patients with CLL with this abnormality can have a stable disease especially if they also have mutated IGHV genes.29,30  Given the small size of the clone, we were able to sequence the IGHV gene in only 1 of the 2 MBL cases and in particular that one with the 17p deletion as sole abnormality (VB1002) that showed indeed a 97% identity with the germ line IGHV sequence.

In conclusion, our study shows that low-count population-screening CLL-like MBL, as opposed to atypical CLL and CD5 MBL, tends to persist over time. The potential risk of progression into overt CLL is exceedingly rare and definitely less than that of clinical MBL even if they carry CLL genomic aberrations. This suggests that these abnormalities occur early during the natural history of MBL/CLL and are probably associated with the acquisition of the typical phenotype rather than with the progression into a fully fledged leukemic disease.

An Inside Blood analysis of this article appears at the front of this issue.

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 was supported by Associazione Italiana per la Ricerca sul Cancro (AIRC; Investigator Grant and Special Program Molecular Clinical Oncology, 5 per mille no. 9965); “Fondazione Piera, Pietro e Giovanni Ferrero”; Fondazione CARIPLO; “CLLGRF–US/European Alliance for the Therapy of CLL,” FIRB and PRIN–Ministero Istruzione, Università e Ricerca (MIUR), Roma, Progetti Integrati Oncologia (PIO)–Ministero della Salute, Roma, Compagnia di San Paolo e Progetto Finalizzato Sanità 2007. C. Scielzo is supported by the EHA Fellowship Program (2009/18), and A.J. is supported by a EHA Partner fellowship program.

Contribution: C.F. and L.S. performed the experiments, analyzed the data, and wrote the manuscript; L.P. and A.T. performed FISH experiments; F.C. performed the experiments and analyzed the data; A.D and A.J. designed and performed immunoglobulin studies; C. Scielzo performed the experiments; C. Sala and D.T. updated clinical and genealogic data; F.C.-C. supervised analysis and wrote the paper; and P.G. designed the study, analyzed the data, and wrote the paper.

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

Correspondence: Federico Caligaris-Cappio, Department of Onco-Hematology, Via Olgettina 60, 20132, Milano, Italy; e-mail: caligaris.federico@hsr.it

1
Marti
 
GE
Rawstron
 
AC
Ghia
 
P
, et al. 
Diagnostic criteria for monoclonal B-cell lymphocytosis.
Br J Haematol
2005
, vol. 
130
 
3
(pg. 
325
-
332
)
2
Rawstron
 
AC
Green
 
MJ
Kuzmicki
 
A
, et al. 
Monoclonal B lymphocytes with the characteristics of “indolent” chronic lymphocytic leukemia are present in 3.5% of adults with normal blood counts.
Blood
2002
, vol. 
100
 
2
(pg. 
635
-
639
)
3
Ghia
 
P
Prato
 
G
Scielzo
 
C
, et al. 
Monoclonal CD5+ and CD5- B-lymphocyte expansions are frequent in the peripheral blood of the elderly.
Blood
2004
, vol. 
103
 
6
(pg. 
2337
-
2342
)
4
Shanafelt
 
TD
Ghia
 
P
Lanasa
 
MC
Landgren
 
O
Rawstron
 
AC
Monoclonal B-cell lymphocytosis (MBL): biology, natural history and clinical management.
Leukemia
2010
, vol. 
24
 
3
(pg. 
512
-
520
)
5
Nieto
 
WG
Almeida
 
J
Romero
 
A
, et al. 
Increased frequency (12%) of circulating chronic lymphocytic leukemia-like B-cell clones in healthy subjects using a highly sensitive multicolor flow cytometry approach.
Blood
2009
, vol. 
114
 
1
(pg. 
33
-
37
)
6
Rawstron
 
AC
Monoclonal B-cell lymphocytosis.
Hematology Am Soc Hematol Educ Program
2009
(pg. 
430
-
439
)
7
Rawstron
 
AC
Yuille
 
MR
Fuller
 
J
, et al. 
Inherited predisposition to CLL is detectable as subclinical monoclonal B-lymphocyte expansion.
Blood
2002
, vol. 
100
 
7
(pg. 
2289
-
2290
)
8
Marti
 
GE
Carter
 
P
Abbasi
 
F
, et al. 
B-cell monoclonal lymphocytosis and B-cell abnormalities in the setting of familial B-cell chronic lymphocytic leukemia.
Cytometry B Clin Cytom
2003
, vol. 
52
 
1
(pg. 
1
-
12
)
9
de Tute
 
R
Yuille
 
M
Catovsky
 
D
Houlston
 
RS
Hillmen
 
P
Rawstron
 
AC
Monoclonal B-cell lymphocytosis (MBL) in CLL families: substantial increase in relative risk for young adults.
Leukemia
2006
, vol. 
20
 
4
(pg. 
728
-
729
)
10
Goldin
 
LR
Lanasa
 
MC
Slager
 
SL
, et al. 
Common occurrence of monoclonal B-cell lymphocytosis among members of high-risk CLL families.
Br J Haematol
2010
, vol. 
151
 
2
(pg. 
152
-
158
)
11
Landgren
 
O
Albitar
 
M
Ma
 
W
, et al. 
B-cell clones as early markers for chronic lymphocytic leukemia.
N Engl J Med
2009
, vol. 
360
 
7
(pg. 
659
-
667
)
12
Rawstron
 
AC
Shanafelt
 
T
Lanasa
 
MC
, et al. 
Different biology and clinical outcome according to the absolute numbers of clonal B-cells in monoclonal B-cell lymphocytosis (MBL).
Cytometry B Clin Cytom
2010
, vol. 
78
 
Suppl 1
(pg. 
S19
-
S23
)
13
Rawstron
 
AC
Bennett
 
FL
O'Connor
 
SJ
, et al. 
Monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia.
N Engl J Med
2008
, vol. 
359
 
6
(pg. 
575
-
583
)
14
Dagklis
 
A
Fazi
 
C
Sala
 
C
, et al. 
The immunoglobulin gene repertoire of low-count chronic lymphocytic leukemia (CLL)-like monoclonal B lymphocytosis is different from CLL: diagnostic implications for clinical monitoring.
Blood
2009
, vol. 
114
 
1
(pg. 
26
-
32
)
15
Hallek
 
M
Cheson
 
BD
Catovsky
 
D
, et al. 
Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines.
Blood
2008
, vol. 
111
 
12
(pg. 
5446
-
5456
)
16
Scarfo
 
L
Dagklis
 
A
Scielzo
 
C
Fazi
 
C
Ghia
 
P
CLL-like monoclonal B-cell lymphocytosis: are we all bound to have it?
Semin Cancer Biol
2010
, vol. 
20
 
6
(pg. 
384
-
390
)
17
Ghia
 
P
Prato
 
G
Stella
 
S
Scielzo
 
C
Geuna
 
M
Caligaris-Cappio
 
F
Age-dependent accumulation of monoclonal CD4+CD8+ double positive T lymphocytes in the peripheral blood of the elderly.
Br J Haematol
2007
, vol. 
139
 
5
(pg. 
780
-
790
)
18
Ghia
 
P
Melchers
 
F
Rolink
 
AG
Age-dependent changes in B lymphocyte development in man and mouse.
Exp Gerontol
2000
, vol. 
35
 
2
(pg. 
159
-
165
)
19
Klinman
 
NR
Kline
 
GH
The B-cell biology of aging.
Immunol Rev
1997
, vol. 
160
 (pg. 
103
-
114
)
20
LeMaoult
 
J
Szabo
 
P
Weksler
 
ME
Effect of age on humoral immunity, selection of the B-cell repertoire and B-cell development.
Immunol Rev
1997
, vol. 
160
 (pg. 
115
-
126
)
21
Dohner
 
H
Stilgenbauer
 
S
Benner
 
A
, et al. 
Genomic aberrations and survival in chronic lymphocytic leukemia.
N Engl J Med
2000
, vol. 
343
 
26
(pg. 
1910
-
1916
)
22
Haferlach
 
C
Dicker
 
F
Schnittger
 
S
Kern
 
W
Haferlach
 
T
Comprehensive genetic characterization of CLL: a study on 506 cases analysed with chromosome banding analysis, interphase FISH, IgV(H) status and immunophenotyping.
Leukemia
2007
, vol. 
21
 
12
(pg. 
2442
-
2451
)
23
Dicker
 
F
Schnittger
 
S
Haferlach
 
T
Kern
 
W
Schoch
 
C
Immunostimulatory oligonucleotide-induced metaphase cytogenetics detect chromosomal aberrations in 80% of CLL patients: a study of 132 CLL cases with correlation to FISH, IgVH status, and CD38 expression.
Blood
2006
, vol. 
108
 
9
(pg. 
3152
-
3160
)
24
Nieto
 
WG
Teodosio
 
C
Lopez
 
A
, et al. 
Non-CLL-like monoclonal B-cell lymphocytosis in the general population: prevalence and phenotypic/genetic characteristics.
Cytometry B Clin Cytom
2010
, vol. 
78
 
Suppl 1
(pg. 
S24
-
S34
)
25
Khan
 
N
Shariff
 
N
Cobbold
 
M
, et al. 
Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals.
J Immunol
2002
, vol. 
169
 
4
(pg. 
1984
-
1992
)
26
Colombatti
 
A
Doliana
 
R
Schiappacassi
 
M
, et al. 
Age-related persistent clonal expansions of CD28(-) cells: phenotypic and molecular TCR analysis reveals both CD4(+) and CD4(+)CD8(+) cells with identical CDR3 sequences.
Clin Immunol Immunopathol
1998
, vol. 
89
 
1
(pg. 
61
-
70
)
27
Fazi
 
C
Dagklis
 
A
Cottini
 
F
, et al. 
Monoclonal B cell lymphocytosis in hepatitis C virus infected individuals.
Cytometry B Clin Cytom
2010
, vol. 
78
 
Suppl 1
(pg. 
S61
-
S68
)
28
Klein
 
U
Lia
 
M
Crespo
 
M
, et al. 
The DLEU2/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia.
Cancer Cell
2010
, vol. 
17
 
1
(pg. 
28
-
40
)
29
Best
 
OG
Gardiner
 
AC
Davis
 
ZA
, et al. 
A subset of Binet stage A CLL patients with TP53 abnormalities and mutated IGHV genes have stable disease.
Leukemia
2009
, vol. 
23
 
1
(pg. 
212
-
214
)
30
Tam
 
CS
Shanafelt
 
TD
Wierda
 
WG
, et al. 
De novo deletion 17p13.1 chronic lymphocytic leukemia shows significant clinical heterogeneity: the M.D. Anderson and Mayo Clinic experience.
Blood
2009
, vol. 
114
 
5
(pg. 
957
-
964
)

Author notes

*

C.F. and L.S. contributed equally to this study.

Sign in via your Institution