• Telomere length in MCL is variable but does not correlate with disease characteristics and survival.

Telomere shortening is of pathogenic and prognostic importance in cancers. In the present study, we analyzed telomere length in 73 mantle cell lymphoma (MCL), 55 chronic lymphocytic leukemia (CLL), and 20 normal B-cell samples using quantitative PCR (Q-PCR) to study its association with disease characteristics and outcome. Telomere length was found to be highly variable in MCL (range, 2.2-13.8 kb; median, 4.3 kb). Telomere dysfunction in MCL was evident from comparison with normal B cells (median, 7.5 kb), but had no significant association with any biologic or clinical feature. This was in contrast to CLL, in which a significant correlation of short telomeres with poor prognostic subgroups was confirmed. There was a trend toward an increased number of genomic aberrations with shortening of telomeres in MCL. No difference in survival was observed between the groups with short and long telomeres, indicating that, as opposed to CLL, telomere length is not of prognostic relevance in MCL.

Mantle cell lymphoma (MCL) is a specific subtype of non-Hodgkin lymphoma with a median overall survival of approximately 4-5 years and significant heterogeneity.1  Several studies have identified various pathogenic and prognostic factors in MCL ranging from genomic aberrations2,3 ; IGHV mutation status4,5 ; gene mutations in ATM,6 TP53,7 CCND1,8  and NOTCH19 ; proliferation index (Ki67)10 ; and expression levels of genes11,12  to the establishment of MCL international prognostic index (MIPI) based on age, performance status, lactate dehydrogenase, and leukocyte count at diagnosis.13  MCL shows a relatively high number of genomic alterations per case2,3  and one of the many causes for genomic instability is telomere dysfunction.

Short, dysfunctional telomeres have been associated with chromosomal end-to-end fusions that predispose the cells to chromosomal aberrations, genomic complexity, and malignant transformation.14  In cancers, there may be a relation between telomere dysfunction and an increase in genomic complexity, clonal evolution, and, consequently, to chemoresistance and poor survival.

In CLL, telomere length is highly variable and short telomeres have been associated with unmutated IGHV status, high-risk genomic aberrations, and poor outcome in univariate and multivariate analyses.15-17  In contrast, study of pathogenic and prognostic role of telomere dysfunction in MCL using a well-characterized, representative cohort has not been done. Earlier studies on telomere length in MCL were carried out with a limited number of samples18,19  and analyses of telomere length associations with other biologic disease characteristics and prognostic factors were not performed.19,20  In the present study, we analyzed telomere length in 73 well-characterized MCL samples from lymph nodes (n = 51) and peripheral blood (n = 22) to evaluate associations with biologic and clinical disease characteristics and outcome.

Tumor samples of 73 MCL and 55 CLL patients treated at the Universities of Ulm, Würzburg, and Heidelberg were obtained after informed consent in accordance with the Declaration of Helsinki and approval from the institutional review boards of each institution. DNA from lymph node samples was isolated from fresh/frozen tissue blocks and that from peripheral blood samples was isolated from PBMCs. Peripheral blood samples from older healthy donors (median age, 56 years; range, 46-68 years) were obtained for comparison of telomere length of MCL with normal B cells and DNA was isolated from CD19-sorted PBMCs. Clinical and molecular characteristics of the MCL and CLL cohorts used for the study are summarized in Table 1. Analyses of genomic aberrations by FISH, proliferation indices by Ki67 stain, array CGH, and sequencing of IGHV and TP53 were done as described previously.10,21,22  Measurement of telomere length was carried out using a Q-PCR–based technique.23  The primers used to amplify telomere and single-copy genes (SCG) were tel1b and tel2b and HBG3 and HBG4, respectively.16,24  Synthetic oligonucleotide standards for telomere (84 bp) and SCG (81 bp) PCR were used to quantify the amount of telomere sequence per sample. Twenty-five nanograms of DNA was used per reaction in triplicates for the telomere and SCG PCRs. Three telomere length controls were included in every plate to detect plate-to-plate variations. The Q-PCR technique was validated by terminal restriction fragment length (TRF) analysis25  and a correlation of R2 = 0.8516 was obtained (supplemental Figure 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article). The TRF value of telomere length for each sample was calculated from the linear regression of Q-PCR versus TRF. Tumor cell content of the samples was estimated by the percentage of cells harboring the most prevalent genomic aberrations as analyzed by FISH (n = 57). Samples with no (n = 10) or partial (7 of 57) FISH data were analyzed for relative expression of CD3 using Q-PCR to control for sample purity (median: 0.15) as described previously.4  Because the MCL samples used in the study were in general associated with high tumor load, 6 samples with no FISH or CD3 expression were also included for analysis. The lymph node and peripheral blood MCL samples were analyzed separately because of potential differences in telomere lengths between tissues. We included the CLL cohort and B cells from healthy donors as control groups for comparison with MCL.

Table 1

Characteristics of the MCL and CLL cohorts

Cohort characteristicsMCL
CLL
n%n%
Median age, y 65  65  
    Range 37-90  35-81  
Ki67 high (> 0.4) 13/39 33.3   
Blastoid morphology 7/59 11.9   
Unmutated IGHV 43/58 74.1 32/54 59.3 
TP53 mutation 16/68 23.5 20/54 37.0 
Deletion 9p21 25/62 40.3   
Deletion 11q22-q23 25/62 40.3 11/55 20 
Deletion 13q14 35/62 56.5 21/55 38.2 
Deletion 17p13 12/58 20.7 12/55 21.8 
No aberration 1/62 1.6 5/55 9.1 
Cohort characteristicsMCL
CLL
n%n%
Median age, y 65  65  
    Range 37-90  35-81  
Ki67 high (> 0.4) 13/39 33.3   
Blastoid morphology 7/59 11.9   
Unmutated IGHV 43/58 74.1 32/54 59.3 
TP53 mutation 16/68 23.5 20/54 37.0 
Deletion 9p21 25/62 40.3   
Deletion 11q22-q23 25/62 40.3 11/55 20 
Deletion 13q14 35/62 56.5 21/55 38.2 
Deletion 17p13 12/58 20.7 12/55 21.8 
No aberration 1/62 1.6 5/55 9.1 

Telomere lengths of MCL and CLL samples were significantly shorter than those of normal B cells, indicating the occurrence of telomere dysfunction in MCL and CLL (Figure 1A). Telomere length has been associated with IGHV mutation status in CLL in several studies.15-17  In the current series, the association of short telomeres with an unmutated IGHV status was confirmed in CLL samples (P < .0001), whereas in the MCL cohort, no significant correlation was found (Figure 1A). In addition, there was no association of telomere length with the presence or absence of TP53 mutations in MCL cases, whereas CLL cases with a TP53 mutation showed a significant association with short telomeres (Figure 1A). There were no significant differences in telomere length comparing MCL samples from the lymph nodes and peripheral blood (supplemental Figure 2).

Figure 1

Correlation of telomere length with biologic and clinical factors. (A) Telomere length distribution in MCL and CLL samples with mutated/unmutated IGHV and mutant/wild-type TP53 and in normal B cells from healthy donors. (B) Correlation of telomere length with proliferation index (Ki67) levels. (C) Telomere length distribution among MCL lymph node samples with and without different genomic aberrations such as del(9p21), del(11q22-q23), del(13q14), del(13qTerminal), and del(17p13). (D) Telomere length distribution among MCL peripheral blood samples with and without different genomic aberrations such as del(9p21), del(11q22-q23), del(13q14), del(13qTerminal), and del(17p13). (E) Telomere length variation with the number of genomic aberrations detected using FISH and/or array-CGH (2 outliers with 0 and 1 genomic aberration detected were removed). (F) Relation of telomere lengths to clinical features in MCL samples. (G) Overall survival in the short (< median) and long telomere (> median) subgroups (median, 48 months; P = .8174). (H) Progression-free survival in the short (< median) and long telomere (> median) subgroups (median 25 vs 22 months; P = .8644). The lines in panels A, C, D, and F represent the median.

Figure 1

Correlation of telomere length with biologic and clinical factors. (A) Telomere length distribution in MCL and CLL samples with mutated/unmutated IGHV and mutant/wild-type TP53 and in normal B cells from healthy donors. (B) Correlation of telomere length with proliferation index (Ki67) levels. (C) Telomere length distribution among MCL lymph node samples with and without different genomic aberrations such as del(9p21), del(11q22-q23), del(13q14), del(13qTerminal), and del(17p13). (D) Telomere length distribution among MCL peripheral blood samples with and without different genomic aberrations such as del(9p21), del(11q22-q23), del(13q14), del(13qTerminal), and del(17p13). (E) Telomere length variation with the number of genomic aberrations detected using FISH and/or array-CGH (2 outliers with 0 and 1 genomic aberration detected were removed). (F) Relation of telomere lengths to clinical features in MCL samples. (G) Overall survival in the short (< median) and long telomere (> median) subgroups (median, 48 months; P = .8174). (H) Progression-free survival in the short (< median) and long telomere (> median) subgroups (median 25 vs 22 months; P = .8644). The lines in panels A, C, D, and F represent the median.

Close modal

In somatic cells, the main mechanism of telomere shortening is attributed to the end replication problem during DNA replication, which therefore should be associated with the cellular proliferation index. However, we observed no correlation between telomere length and the proliferation index in the MCL samples (Figure 1B). This might be because of variable telomerase activity among the different MCL subsets. MCL cases of the blastoid subtype showed a tendency to have shorter telomeres, but the difference was not significant (Figure 1F).

Telomere lengths of the MCL samples did not show significant association with specific genomic aberrations. Only slight variations in median telomere length were observed among the MCL subgroups with different genomic aberrations in the lymph node (Figure 1C) and peripheral blood samples (Figure 1D). In contrast, in the CLL samples, a significant association of short telomeres with the high-risk aberrations del(17p13) and del(11q22-q23) was observed (supplemental Figure 3).16,17 

Because telomere shortening could contribute to genomic instability and therefore to an increased incidence of genomic alterations, we analyzed the association of telomere length with the number of genomic aberrations. A weak but significant correlation (R2 = 0.1534, P = .022) was observed comparing telomere length with the number of genomic aberrations (Figure 1E). However, analysis of the relation of telomere length with clinical features showed no correlation with lactate dehydrogenase levels, WBC, or with MIPI groups (Figure 1F and supplemental Figure 4).

In the present study, we analyzed the impact of telomere length on survival in MCL cases by categorizing the patients into short (< median) and long (> median) telomere subgroups. No significant difference in overall survival (median, 48 months; P = .8174; Figure 1G) or progression-free survival (median, 25 vs 22 months; P = .8644; Figure 1H) was identified between the 2 subgroups. In contrast, when evaluating the impact of established prognostic factors on outcome to verify that our cohort was representative, we observed the expected correlations of Ki67 (P = .0001), MIPI index (P = .007), and B symptoms (P = .0119) with disease outcome (data not shown).

In summary, the comprehensive characterization of MCL cases using a large cohort with mature follow-up showed that telomeres in MCL samples were possibly dysfunctional by comparison with normal B cells but were not associated with any of the established clinical or biologic factors relevant for pathogenesis or prognostication and disease outcome. This is in marked contrast to CLL, in which telomere length has been associated with biologic disease characteristics, pathogenic mechanisms, and survival in multivariate analysis.15-17 

The online version of this article contains a data supplement.

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.

The authors thank Sabrina Kless for excellent technical assistance.

This work was supported by funding from Deutsche Forschungsgemeinschaft (SFB1074, subproject B1) and the European Mantle Cell Lymphoma Network. B.M.C.J. was supported by a fellowship from the International Graduate School in Molecular Medicine, Ulm, Germany.

Contribution: B.M.C.J. performed the experiments, interpreted the data, and wrote the manuscript; D.K. interpreted the data, edited the manuscript, and provided vital material; A.L. and D.M. interpreted the data and edited the manuscript; M.H. performed the experiments; G.O., A.R., T.F.E.B., and P.M. interpreted the data, edited the manuscript, and provided vital material; T.Z. designed the research and conceived the study; T.Z. and H.D. interpreted the data and edited the manuscript; and S.S. conceived the study, designed the research, interpreted the data, and wrote the manuscript.

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

Correspondence: Stephan Stilgenbauer, MD, Department of Internal Medicine III, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; e-mail: stephan.stilgenbauer@uniklinik-ulm.de.

1
Herrmann
 
A
Hoster
 
E
Zwingers
 
T
, et al. 
Improvement of overall survival in advanced stage mantle cell lymphoma.
J Clin Oncol
2009
, vol. 
27
 
4
(pg. 
511
-
518
)
2
Sander
 
S
Bullinger
 
L
Leupolt
 
E
, et al. 
Genomic aberrations in mantle cell lymphoma detected by interphase fluorescence in situ hybridization. Incidence and clinicopathological correlations.
Haematologica
2008
, vol. 
93
 
5
(pg. 
680
-
687
)
3
Hartmann
 
EM
Campo
 
E
Wright
 
G
, et al. 
Pathway discovery in mantle cell lymphoma by integrated analysis of high-resolution gene expression and copy number profiling.
Blood
2010
, vol. 
116
 
6
(pg. 
953
-
961
)
4
Kienle
 
D
Kröber
 
A
Katzenberger
 
T
, et al. 
VH mutation status and VDJ rearrangement structure in mantle cell lymphoma: correlation with genomic aberrations, clinical characteristics, and outcome.
Blood
2003
, vol. 
102
 
8
(pg. 
3003
-
3009
)
5
Hadzidimitriou
 
A
Agathangelidis
 
A
Darzentas
 
N
, et al. 
Is there a role for antigen selection in mantle cell lymphoma? Immunogenetic support from a series of 807 cases.
Blood
2011
, vol. 
118
 
11
(pg. 
3088
-
3095
)
6
Schaffner
 
C
Idler
 
I
Stilgenbauer
 
S
Döhner
 
H
Lichter
 
P
Mantle cell lymphoma is characterized by inactivation of the ATM gene.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 
6
(pg. 
2773
-
2778
)
7
Hernandez
 
L
Fest
 
T
Cazorla
 
M
, et al. 
p53 gene mutations and protein overexpression are associated with aggressive variants of mantle cell lymphomas.
Blood
1996
, vol. 
87
 
8
(pg. 
3351
-
3359
)
8
Wiestner
 
A
Tehrani
 
M
Chiorazzi
 
M
, et al. 
Point mutations and genomic deletions in CCND1 create stable truncated cyclin D1 mRNAs that are associated with increased proliferation rate and shorter survival.
Blood
2007
, vol. 
109
 
11
(pg. 
4599
-
4606
)
9
Kridel
 
R
Meissner
 
B
Rogic
 
S
, et al. 
Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma.
Blood
2012
, vol. 
119
 
9
(pg. 
1963
-
1971
)
10
Katzenberger
 
T
Petzoldt
 
C
Höller
 
S
, et al. 
The Ki67 proliferation index is a quantitative indicator of clinical risk in mantle cell lymphoma.
Blood
2006
, vol. 
107
 
8
pg. 
3407
 
11
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
 
2
(pg. 
185
-
197
)
12
Fernàndez
 
V
Salamero
 
O
Espinet
 
B
, et al. 
Genomic and gene expression profiling defines indolent forms of mantle cell lymphoma.
Cancer Res
2010
, vol. 
70
 
4
(pg. 
1408
-
1418
)
13
Hoster
 
E
Dreyling
 
M
Klapper
 
W
, et al. 
A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma.
Blood
2008
, vol. 
111
 
2
(pg. 
558
-
565
)
14
Begus-Nahrmann
 
Y
Hartmann
 
D
Kraus
 
J
, et al. 
Transient telomere dysfunction induces chromosomal instability and promotes carcinogenesis.
J Clin Invest
2012
, vol. 
122
 
6
(pg. 
2283
-
2288
)
15
Damle
 
RN
Batliwalla
 
FM
Ghiotto
 
F
, et al. 
Telomere length and telomerase activity delineate distinctive replicative features of the B-CLL subgroups defined by immunoglobulin V gene mutations.
Blood
2004
, vol. 
103
 
2
(pg. 
375
-
382
)
16
Roos
 
G
Kröber
 
A
Grabowski
 
P
, et al. 
Short telomeres are associated with genetic complexity, high-risk genomic aberrations, and short survival in chronic lymphocytic leukemia.
Blood
2008
, vol. 
111
 
4
(pg. 
2246
-
2252
)
17
Rampazzo
 
E
Bonaldi
 
L
Trentin
 
L
, et al. 
Telomere length and telomerase levels delineate subgroups of B-cell chronic lymphocytic leukemia with different biological characteristics and clinical outcomes.
Haematologica
2012
, vol. 
97
 
1
(pg. 
56
-
63
)
18
Cottliar
 
AS
Panero
 
J
Pedrazzini
 
E
, et al. 
Analysis of telomere length in mantle cell lymphoma.
Eur J Haematol
2009
, vol. 
83
 
5
(pg. 
433
-
438
)
19
Ladetto
 
M
Compagno
 
M
Ricca
 
I
, et al. 
Telomere length correlates with histo- pathogenesis according to the germinal center in mature B-cell lymphoproliferative disorders.
Blood
2004
, vol. 
103
 
12
(pg. 
4644
-
4649
)
20
Walsh
 
SH
Grabowski
 
P
Berglund
 
M
, et al. 
Telomere length and correlation with histopathogenesis in B-cell leukemias/lymphomas.
Eur J Haematol
2007
, vol. 
78
 
4
(pg. 
283
-
289
)
21
Zenz
 
T
Häbe
 
S
Denzel
 
T
, et al. 
Detailed analysis of p53 pathway defects in fludarabine-refractory chronic lymphocytic leukemia (CLL): dissecting the contribution of 17p deletion, TP53 mutation, p53-p21 dysfunction, and miR34a in a prospective clinical trial.
Blood
2009
, vol. 
114
 
13
(pg. 
2589
-
2597
)
22
Kohlhammer
 
H
Schwaenen
 
C
Wessendorf
 
S
, et al. 
Genomic DNA-chip hybridization in t(11;14)-positive mantle cell lymphomas shows a high frequency of aberrations and allows a refined characterization of consensus regions.
Blood
2004
, vol. 
104
 
3
(pg. 
795
-
801
)
23
O'Callaghan
 
N
Dhillon
 
V
Thomas
 
P
Fenech
 
M
A quantitative real-time PCR method for absolute telomere length.
Biotechniques
2008
, vol. 
44
 
6
(pg. 
807
-
809
)
24
Cawthon
 
RM
Telomere measurement by quantitative PCR.
Nucleic Acids Res
2002
, vol. 
30
 
10
pg. 
e47
 
25
Kruk
 
PA
Rampino
 
NJ
Bohr
 
VA
DNA damage and repair in telomeres: Relation to aging.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 
1
(pg. 
258
-
262
)
Sign in via your Institution