• CDKN2A and TP53 deletions remain of bad prognostic value in younger MCL patients treated according to the current standard of care.

  • CDKN2A and TP53 deletions have independent deleterious effects and should be considered for treatment decisions in addition to MIPI and Ki-67 index.

We revisited the prognostic value of frequently detected somatic gene copy number alterations (CNAs) in mantle cell lymphoma (MCL) patients treated first line with immunochemotherapy and autologous stem cell transplantation (ASCT), with or without high-dose cytarabine, in the randomized European MCL Younger trial. DNA extracted from tumor material of 135 patients (median age, 56 years) was analyzed by multiplex ligation-dependent probe amplification and/or quantitative multiplex polymerase chain reaction of short fluorescent fragments. As expected, MYC (18%) was the more frequently gained, whereas RB1 (26%), ATM (25%), CDKN2A (p16) (25%), and TP53 (22%) were the more frequently deleted. Whether adjusted for MCL International Prognostic Index (MIPI) or not, deletions of RB1, CDKN2A, TP53, and CDKN1B were associated with shorter overall survival (OS), similarly in both treatment arms, whereas CNAs in MYC, ATM, CDK2, CDK4, and MDM2 had no prognostic value. Additive effects were seen for CDKN2A (hazard ratio, 2.3; P = .007, MIPI-adjusted) and TP53 deletions (hazard ratio, 2.4; P = .007), reflected in a dismal outcome with simultaneous deletions (median OS, 1.8 years) compared with single deletions (median OS, 4.3 and 5.1 years) or without these deletions (median OS, 7 years), again similarly in both treatment arms. The additive prognostic effects of CDKN2A and TP53 deletions were independent of the Ki-67 index. Despite immunochemotherapy, high-dose cytarabine, and ASCT, younger MCL patients with deletions of CDKN2A (p16) and TP53 show an unfavorable prognosis and are candidates for alternative therapeutic strategies. This trial was registered at www.clinicaltrials.gov as #NCT00209222.

Mantle cell lymphoma (MCL) is a well-defined subtype of B-cell lymphoma characterized by t(11;14)(q13;q32)-driven overexpression of cyclin D1.1,2  It is rare, representing only 5% to 9% of all lymphomas.3  The median age at diagnosis is ∼65 years. Other than occasional indolent forms,4  MCL is considered to be an aggressive disease, and its treatment is a challenge.5,6  Despite considerable improvement in progression-free survival7  (∼2 years after first-line conventional immunochemotherapy and >5 years after dose-intensified therapy including high-dose cytarabine8 ), individual heterogeneity in clinical behavior is still encountered, ranging from primary refractory disease to a progression-free survival of >7 years.9 

Although numerous genetic abnormalities are involved in the pathophysiology of MCL,10,11  not all are of prognostic value. The CDKN2A locus (9p21), which encodes for both the CDK4/6 inhibitor INK4a (p16) and the positive TP53 regulator ARF (p14), is one of the most frequently deleted and is consistently associated with a poor prognosis in MCL.12-19  Additional genes frequently targeted by secondary genetic alterations,20  eg, CDKN1B/CDK2/CDK4/RB1, involved in cell cycle regulation,16,17,21,22  and ATM/MDM2/TP53, involved in DNA damage response,14,18,21 are less consistently associated with prognosis. Alterations of the 8q24 (MYC) locus are uncommon in MCL but have been identified in cases with blastoid MCL.23,24  The association of the overexpression of the primary cell cycle regulator cyclin D1 and secondary alterations in cell cycle regulation translates into high level cell proliferation and shorter survival.13  Accordingly, the tumor cell proliferation marker Ki-67 index25,26  adds to the prognostic value of the MCL International Prognostic Index (MIPI),27  the first clinical prognostic index specific for MCL.28 

Previous reports on the prognostic value of gene copy number alterations (CNAs) in MCL are mainly based on smaller retrospective patient cohorts and/or with heterogeneous treatment, including only 0% to 40% of patients with rituximab-containing chemotherapy. Using the clinical data of the randomized European MCL Younger trial,29  the aim of our study was to re-evaluate the prognostic value of somatic CNAs in the context of the current standard of care, including immunochemotherapy, high-dose cytarabine, and autologous stem cell transplantation (ASCT).8 

Patients

Patients up to 65 years old with previously untreated MCL of Ann Arbor stages II to IV were enrolled in the randomized MCL Younger trial of the European MCL Network (#NCT00209222). Patients received either 6 cycles of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), followed by stem cell mobilization and myeloablative radio-chemotherapy with ASCT, or 6 cycles of alternating R-CHOP or rituximab, dexamethasone, high-dose cytarabine, and cisplatin (R-DHAP), followed by high-dose cytarabine containing myeloablative radio-chemotherapy and ASCT29  (supplemental Figure 1 available on the Blood Web site). The trial was performed in accordance with the Declaration of Helsinki, and all patients gave written informed consent to participate and to provide blood, bone marrow, and tissue for biologic studies.

The inclusion criteria for patients analyzed in the present biological study were (1) a confirmed histologic diagnosis of MCL with overexpression of cyclin D1 assessed by immunohistochemistry and/or molecular biology; (2) the availability of diagnostic tumor DNA; and (3) complete clinical data. When no frozen tissue DNA was available, peripheral samples with >50% tumor cells were considered eligible for CNA analysis. Diagnostic peripheral blood and bone marrow aspirations had been prospectively collected during the minimal residual disease program linked to the MCL Younger trial,30  with tumor load prospectively assessed by 4-color flow cytometry and/or clonospecific quantitative polymerase chain reaction (PCR). Finally, when no other diagnostic material was available, formalin-fixed, paraffin-embedded (FFPE) tumor sections were collected from the LYSA-Pathology platform.

Histopathologic and clinical variables

The histologic diagnosis was confirmed by the central pathology review at one of the designated pathology reference centers (European MCL Pathology Panel). MCL cytology was classified as blastoid (pleomorphic or blastic) or nonblastoid (classical or small cell).31  The Ki-67 index was defined as the percentage of Ki-67–positive tumor cells in representative areas of the lymphoma and quantified according to published guidelines.32  We used the established 30% threshold to distinguish patients with a low or high Ki-67 index.33  The individual clinical prognostic information was summarized in the quantitative MIPI score using age, performance status, lactate dehydrogenase (LDH), and white blood cell count (WBC) at trial entry.27  Outcome variables were overall survival (OS) and time to treatment failure (TTF). OS was measured from trial registration to death from any cause. Patients who had not died during the observation period were censored for OS at the latest contact date. TTF was measured from start of treatment to stable disease at end of induction, progression, or death from any cause. Patients with no failure, progression, or death during observation time were censored for TTF at the latest tumor assessment date. The clinical data cutoff date was April 7, 2013.

DNA extraction and quality control

DNA was extracted from peripheral blood and/or bone marrow samples using standard proteinase K digestion and phenol-chloroform extraction or the QIAGEN Blood Mini Kit (QIAGEN).30  DNAs extracted from tumor tissues were sent to the Creteil biological center by French locoregional “Centres de Ressources Biologiques.” The quality of tumor DNA samples was assessed using the BIOMED-2 specimen control “barcode“ multiplex PCR reaction.34 

CNA analysis

Quantitative multiplex PCR of short fluorescent fragments

An MCL-dedicated quantitative multiplex PCR of short fluorescent fragments (QMPSF) assay was used as described (supplemental Figure 2A).18  The following target genes were analyzed: MYC (8q24), RB1 (13q14), ATM (11q22-23), CDKN2A (p16) (9p21), TP53 (17p13), CDK2 (12q13), CDKN1B (p27) (12p13), and MDM2 (12q14). The reference gene was CECR1, located at 22q11. Two control DNAs were used to calculate the mean template/control peak height ratio (commercial DNA; Roche Diagnostics, Mylan, France). PCR fragments ranged from 150 to 250 bp. QMPSF is able to detect CNAs when tumor cells represent >30% of total cells analyzed.18 

Multiplex ligation-dependent probe amplification

In contrast to QMPSF, multiplex ligation-dependent probe amplification (MLPA) (supplemental Figure 2A) probe-targeted sequences are small (50-70 nucleotides), an attractive property for FFPE-derived analysis. Hybridization and ligation steps were performed on 100 ng genomic DNA, and probes were subjected to PCR reactions using SALSA P038-A2 CLL probemix-2 according to the manufacturer's instructions (MRC-Holland, Amsterdam, The Netherlands). Targeted loci included 8q24 (MYC), several targets on 13q14, including one specific for RB1, 9p21 (CDKN2A [p16]), 11q23 (ATM), 17p13 (TP53), and 12q14 (CDK4). Products were run on an automated sequencer (ABI 3130X1 genetic analyzer). The calculation of probe ratios consisted of a mathematical comparison between relative quantities of target DNA amplified from a test patient sample and those generated in 6 normal control samples. It was performed using Coffalyser software (MRC-Holland BV) A decrease or increase in peak height values resulting in ratios <0.80 or >1.20, respectively, indicated copy loss or gain in tumor cells. MLPA has been reported to detect CNAs when tumor cells represent >40% of total cells analyzed.35,36  In a validation step using the GRANTA cell line, we were able to detect the homozygous CDKN2A (p16) deletion when only 25% of tumor cells were present in studied samples (sample/reference allele ratio = 0.5; supplemental Figure 2B).

CNA result encoding

The first technique used was QMPSF. However, this technique was not suited for paraffin-embedded tissues, and several patients had an alteration of the gene used for normalization. We moved to the MLPA technique and reanalyzed all patients for whom enough tumor material remained available. From 560 comparisons for shared loci, 13 discrepancies between the 2 techniques were observed (supplemental Figure 2C). In discrepant cases, MLPA was repeated. If another repeat could not be performed (no more material), results were considered inconclusive. When repeated, MLPA confirmed precedent MLPA results in all performed cases, and the obtained MLPA values were considered for analyses. For samples with an estimated 50% tumor cell infiltration and a sample/reference ratio between 0.8 and 0.9, results were reported as inconclusive to avoid false negativities.

Statistical methods

Clinical characteristics, histopathologic features, and clinical outcome of CNA evaluable MCL trial patients were compared with nonevaluable patients. Univariable analyses of the prognostic value of CNAs were done using Kaplan-Meier estimates and log-rank tests. For all genes with CNAs detected in >10 patients, we performed univariable and multivariable Cox regression analyses. We adjusted for clinical prognostic factors summarized in the quantitative MIPI score and for the Ki-67 index. In multivariable analyses, we identified independently prognostic CNAs by backward elimination using the Wald statistic, keeping MIPI score constantly included. Initially, all CNAs with prognostic impact in univariable analyses, indicated by P < .05, were included in multiple Cox regression models, and subsequently backward elimination was performed using an exclusion level of P < .01. As sensitivity analyses, we added all CNAs not included in multivariable analyses for backward elimination separately to the final Cox model.

We performed subgroup analyses according to treatment group and investigated the potential interaction of prognostic effects with treatment using interaction terms in multivariable Cox regression. For statistical comparisons, we used Fisher’s exact, Mann-Whitney U, log-rank, and Wald tests. All reported P values are 2 sided and descriptive.

Patient characteristics

One hundred thirty-five patients from the MCL Younger trial had suitable tumor DNA available for the CNA study (supplemental Figure 3); 72 patients were in the experimental R-CHOP/R-DHAP group. The median age at trial registration was 56 years (Table 1). Eighty-six percent of patients had stage IV disease, and according to MIPI, 49%, 26%, and 25% of patients were of low, intermediate, and high risk, respectively. Eight (9%) of 89 patients with assessed cytology had blastoid MCL. The median Ki-67 index was 19%, and the Ki-67 index was ≥30% in 32% of the 72 patients with an available Ki-67 index. After a median follow-up of 4.6 years, the estimated median OS and TTF for the 135 patients was 6.8 (50 events) and 4.6 years (72 events), respectively. Compared with the 440 MCL patients not included in the biologic study, selected patients had similar age, more frequent bone marrow involvement, increased LDH, and higher WBC and thus had a less favorable risk profile (Table 1). Accordingly, OS and TTF were slightly shorter in the studied patients compared with the nonstudied patients (5-year OS rates, 66% vs 73%; P = .0083; 5-year TTF rates, 46% vs 55%; P = .0122).

Table 1

Patient baseline characteristics

VariableValueIncluded (n = 135)Not included (n = 440)P value
Age (years) Median (min-max) 56 (35-65) 55 (30-66) .33 
Male No. (%) 109 (81) 350 (80) .81 
Stage    .55 
 I No. (%) 0 (0) 2 (0)  
 II No. (%) 3 (2) 20 (5)  
 III No. (%) 16 (12) 61 (14)  
 IV No. (%) 116 (86) 357 (81)  
B-symptoms No. (%) 51 (38) 163 (37) .92 
ECOG PS    .061 
 0-1 No. (%) 126 (93) 428 (97)  
 2 No. (%) 9 (7) 12 (3)  
Bone marrow involvement No. (%) 113 (84) 323 (73) .016 
Elevated LDH No. (%) 63 (47) 147 (33) .006 
LDH/ULN Median (min-max) 0.97 (0.41-4.8) 0.89 (0.29-12.2) .038 
WBC (G/L) Median (min-max) 8.8 (0.052-388) 7.2 (1.1-1110) .001 
MIPI score Median (min-max) 5.73 (3.63-8.03) 5.51 (4.07-8.68) <.001 
MIPI risk    <.001 
 Low No. (%) 66 (49) 294 (67)  
 Intermediate No. (%) 35 (26) 96 (22)  
 High No. (%) 34 (25) 50 (11)  
Ki-67 index (%)* Median (min-max) 19 (2-87) 21 (0-97) .52 
Ki-67 index ≥ 30% No. (%) 23 (32) 63 (26) .30 
Blastoid cytology No. (%) 8 (9) 22 (8) .32 
Treatment    .83 
 R-CHOP No. (%) 63 (47) 174 (44)  
 R-CHOP/R-DHAP No. (%) 72 (53) 217 (55)  
VariableValueIncluded (n = 135)Not included (n = 440)P value
Age (years) Median (min-max) 56 (35-65) 55 (30-66) .33 
Male No. (%) 109 (81) 350 (80) .81 
Stage    .55 
 I No. (%) 0 (0) 2 (0)  
 II No. (%) 3 (2) 20 (5)  
 III No. (%) 16 (12) 61 (14)  
 IV No. (%) 116 (86) 357 (81)  
B-symptoms No. (%) 51 (38) 163 (37) .92 
ECOG PS    .061 
 0-1 No. (%) 126 (93) 428 (97)  
 2 No. (%) 9 (7) 12 (3)  
Bone marrow involvement No. (%) 113 (84) 323 (73) .016 
Elevated LDH No. (%) 63 (47) 147 (33) .006 
LDH/ULN Median (min-max) 0.97 (0.41-4.8) 0.89 (0.29-12.2) .038 
WBC (G/L) Median (min-max) 8.8 (0.052-388) 7.2 (1.1-1110) .001 
MIPI score Median (min-max) 5.73 (3.63-8.03) 5.51 (4.07-8.68) <.001 
MIPI risk    <.001 
 Low No. (%) 66 (49) 294 (67)  
 Intermediate No. (%) 35 (26) 96 (22)  
 High No. (%) 34 (25) 50 (11)  
Ki-67 index (%)* Median (min-max) 19 (2-87) 21 (0-97) .52 
Ki-67 index ≥ 30% No. (%) 23 (32) 63 (26) .30 
Blastoid cytology No. (%) 8 (9) 22 (8) .32 
Treatment    .83 
 R-CHOP No. (%) 63 (47) 174 (44)  
 R-CHOP/R-DHAP No. (%) 72 (53) 217 (55)  

ECOG PS, Eastern Cooperative Oncology Group performance status; ULN, upper limit of normal range.

*

Ki-67 assessed in 72 of 135 included and in 245 of 440 not included patients.

Cytology assessed in 68 of 135 included and 270 of 440 not included patients.

In 48 patients treatment not documented, 1 R-bendamustin.

CNA detection

DNA with >50% tumor cells was obtained from lymph node biopsies for 79 patients (including 61 frozen and 18 FFPE samples), peripheral blood for 54 patients, and bone marrow aspirates for 2 patients. The most frequent copy gain involved MYC (18%; Table 2), whereas the most frequent deletion involved the 13q14 locus (36%), including RB1 in 26%. CNAs in ATM were mostly deletions (25%), but copy gains were found in 3 of 129 patients. As expected, CDKN2A (p16) and TP53 deletions were frequent (25% and 22%, respectively). Of 114 patients analyzed with both QMPSF and MLPA, only 24 (21%) displayed no CNA in any analyzed loci. Details of the CNA distribution for each MCL case are shown in supplemental Figure 4. The distribution of CNAs did not differ substantially according to the type of sample analyzed (supplemental Table 1).

Table 2

Distribution of gene copy number alterations in the studied cohort

Allelic statusMYC (8q24)Locus (13q14)RB1 (13q14)ATM (11q23)CDKN2A (9p21)TP53 (17p13)CDK2 (12q13)CDK4 (12q14)CDKN1B (12p13)MDM2 (12q14)
Total interpretable (n) 130 130 131 129 134 134 116 129 109 114 
Diploid locus (n) 107 82 96 94 100 104 106 119 93 108 
Diploid locus (%) 82% 63% 73% 73% 75% 78% 91% 92% 85% 95% 
Monoallelic deletion (n) 44 33 31 19 30 13 
Monoallelic deletion (%) 0% 34% 25% 24% 14% 22% 1% 2% 12% 2% 
Biallelic deletion (n) 15 
Biallelic deletion (%) 0% 2% 1% 1% 11% 0% 0% 0% 0% 0% 
Gain (n) 23 
Gain (%) 18% 1% 1% 2% 0% 0% 8% 6% 3% 4% 
Total deletions (%) 0% 36% 26% 25% 25% 22% 1% 2% 12% 2% 
Total abnormalities (%) 18% 37% 27% 27% 25% 22% 9% 8% 15% 5% 
Allelic statusMYC (8q24)Locus (13q14)RB1 (13q14)ATM (11q23)CDKN2A (9p21)TP53 (17p13)CDK2 (12q13)CDK4 (12q14)CDKN1B (12p13)MDM2 (12q14)
Total interpretable (n) 130 130 131 129 134 134 116 129 109 114 
Diploid locus (n) 107 82 96 94 100 104 106 119 93 108 
Diploid locus (%) 82% 63% 73% 73% 75% 78% 91% 92% 85% 95% 
Monoallelic deletion (n) 44 33 31 19 30 13 
Monoallelic deletion (%) 0% 34% 25% 24% 14% 22% 1% 2% 12% 2% 
Biallelic deletion (n) 15 
Biallelic deletion (%) 0% 2% 1% 1% 11% 0% 0% 0% 0% 0% 
Gain (n) 23 
Gain (%) 18% 1% 1% 2% 0% 0% 8% 6% 3% 4% 
Total deletions (%) 0% 36% 26% 25% 25% 22% 1% 2% 12% 2% 
Total abnormalities (%) 18% 37% 27% 27% 25% 22% 9% 8% 15% 5% 

CNAs, Ki-67 index, and cytology

Among the patients with high Ki-67 index (≥30%), deletions in 13q14, RB1, CDKN2A (p16), and CDKN1B (p27) were substantially more frequent compared with patients with low Ki-67 index (65% vs 25%, 48% vs 19%, 43% vs 10%, and 24% vs 8%, respectively), whereas the frequencies of the remaining CNAs, including TP53 deletion, were independent of the Ki-67 index (22% vs 18%; supplemental Table 2). Frequencies of CNAs were similar in blastoid vs nonblastoid MCL, except for TP53, which was more frequently deleted in blastoid MCL (4 of 8) compared with nonblastoid MCL (11 of 81, 14%; supplemental Table 2).

Prognostic relevance of CNAs

In analyses including single genes without and with adjustment for MIPI, deletions of the 13q14 locus, RB1, CDKN2A (p16), TP53, and CDKN1B (p27) were associated with shorter OS and TTF (Table 3; Figure 1; supplemental Figure 5), whereas CNAs in MYC, ATM, CDK2, CDK4, and MDM2 were not prognostic (Table 3). The MIPI-adjusted OS hazard ratio (HR) for deletions in the 13q14 locus was 2.1 (P = .010), whereas it was 2.2 (P = .014) for RB1 deletions and 2.2 (P = .010) for CDKN2A (p16) deletions. Patients with heterozygous or homozygous deletions of CDKN2A (p16) showed a similar outcome. For TP53 deletions, the adjusted OS HR was 2.4 (P = .008) and for CDKN1B (p27) deletions was 1.9 (P = .083). Similar results were seen for TTF (Table 3). There was no indication for an interaction of the prognostic effect for any CNA with the treatment group. In particular, deletions of CDKN2A (p16) or TP53 had similar prognostic value in the experimental (Figure 1) and control treatment groups (supplemental Figures 5 and 6).

Table 3

Prognostic impact of CNAs by Cox regression analyses without or with adjustment for MIPI score

VariableAdjustmentOSTTF
HR95% CI, lower bound95% CI, upper boundPHR95% CI, lower bound95% CI, upper boundP
MYC gain None 0.89 0.43 1.84 .74 1.01 0.56 1.82 .98 
 MIPI score 1.03 0.49 2.16 .93 1.10 0.61 2.00 .75 
13q14 deletion None 2.03 1.15 3.60 .015 2.40 1.47 3.92 <.001 
 MIPI score 2.13 1.19 3.80 .010 2.49 1.52 4.06 <.001 
RB1 deletion None 1.75 0.96 3.22 .070 1.91 1.14 3.19 .013 
 MIPI score 2.20 1.18 4.11 .014 2.25 1.33 3.79 .002 
ATM deletion None 0.65 0.33 1.29 .22 0.81 0.47 1.38 .44 
 MIPI score 0.77 0.38 1.55 .47 0.91 0.52 1.57 .72 
CDKN2A deletion None 2.97 1.67 5.29 <.001 2.62 1.59 4.33 <.001 
 MIPI score 2.22 1.21 4.07 .010 2.15 1.28 3.62 .004 
TP53 deletion None 2.95 1.58 5.48 .001 2.39 1.41 4.06 .001 
 MIPI score 2.36 1.26 4.42 .008 1.96 1.14 3.36 .015 
CDKN1B deletion None 2.31 1.13 4.70 .021 2.42 1.28 4.57 .007 
 MIPI score 1.89 0.92 3.89 .083 2.03 1.07 3.85 .031 
VariableAdjustmentOSTTF
HR95% CI, lower bound95% CI, upper boundPHR95% CI, lower bound95% CI, upper boundP
MYC gain None 0.89 0.43 1.84 .74 1.01 0.56 1.82 .98 
 MIPI score 1.03 0.49 2.16 .93 1.10 0.61 2.00 .75 
13q14 deletion None 2.03 1.15 3.60 .015 2.40 1.47 3.92 <.001 
 MIPI score 2.13 1.19 3.80 .010 2.49 1.52 4.06 <.001 
RB1 deletion None 1.75 0.96 3.22 .070 1.91 1.14 3.19 .013 
 MIPI score 2.20 1.18 4.11 .014 2.25 1.33 3.79 .002 
ATM deletion None 0.65 0.33 1.29 .22 0.81 0.47 1.38 .44 
 MIPI score 0.77 0.38 1.55 .47 0.91 0.52 1.57 .72 
CDKN2A deletion None 2.97 1.67 5.29 <.001 2.62 1.59 4.33 <.001 
 MIPI score 2.22 1.21 4.07 .010 2.15 1.28 3.62 .004 
TP53 deletion None 2.95 1.58 5.48 .001 2.39 1.41 4.06 .001 
 MIPI score 2.36 1.26 4.42 .008 1.96 1.14 3.36 .015 
CDKN1B deletion None 2.31 1.13 4.70 .021 2.42 1.28 4.57 .007 
 MIPI score 1.89 0.92 3.89 .083 2.03 1.07 3.85 .031 

By log-rank tests, no prognostic effects were seen for CNAs in CDK2 (P = .87 for TTF and P = .45 for OS), CDK4 (P = .34 for TTF and P = .14 for OS), and MDM2 (P = .43 for TTF and P = .69 for OS). Due to the low number of CNAs, these genes were not included in Cox regression analyses. CI, confidence interval.

Figure 1

Prognostic value of CDKN2A (p16) and TP53 deletions on OS in pooled treatment arms or in the experimental treatment arm (R-CHOP/R-DHAP). (A) OS according to the presence or absence of CDKN2A (p16) deletion in pooled treatment arms. (B) OS according to the presence or absence of CDKN2A (p16) deletion in the experimental treatment arm. (C) OS according to the presence or absence of TP53 deletion in pooled treatment arms. (D) OS according to the presence or absence of TP53 deletion in the experimental treatment arm. (E) OS according to the presence or absence of CDKN2A (p16) and TP53 deletions in pooled treatment arms. (F) OS according to the presence or absence of CDKN2A (p16) and TP53 deletions in the experimental treatment arm.

Figure 1

Prognostic value of CDKN2A (p16) and TP53 deletions on OS in pooled treatment arms or in the experimental treatment arm (R-CHOP/R-DHAP). (A) OS according to the presence or absence of CDKN2A (p16) deletion in pooled treatment arms. (B) OS according to the presence or absence of CDKN2A (p16) deletion in the experimental treatment arm. (C) OS according to the presence or absence of TP53 deletion in pooled treatment arms. (D) OS according to the presence or absence of TP53 deletion in the experimental treatment arm. (E) OS according to the presence or absence of CDKN2A (p16) and TP53 deletions in pooled treatment arms. (F) OS according to the presence or absence of CDKN2A (p16) and TP53 deletions in the experimental treatment arm.

Close modal

In multivariable analyses including deletions of RB1, CDKN2A (p16), TP53, and CDKN1B (p27) and adjusting for MIPI, only deletions of CDKN2A (p16) (OS HR, 2.3; P = .007) and TP53 (OS HR, 2.4; P = .007) showed independent prognostic impact. The group with both CDKN2A (p16) and TP53 deletions showed the worst prognosis, with median OS and TTF of 1.8 and 0.5 years, respectively (Figure 1; supplemental Figure 5), and again, as there was no interaction with the treatment group, the independent prognostic impact of the CDKN2A (p16) deletion and TP53 deletion on OS and TTF was clearly seen in both the experimental (R-CHOP/R-DHAP) arm (Figure 1; supplemental Figure 5) and the control (R-CHOP) arm (supplemental Figure 6). No other CNA investigated showed an additional prognostic impact to CDKN2A (p16) and TP53 deletions.

When adjusted for the Ki-67 index, the prognostic impact of CDKN2A (p16) deletion as single variable on OS still persisted but was reduced (HR unadjusted, 5.3; adjusted for Ki-67 index, 3.8; adjusted for Ki-67 index and MIPI, 3.6), whereas the prognostic impact of TP53 deletion as single variable was only slightly modified by the inclusion of the Ki-67 index (HR unadjusted, 2.7; adjusted for Ki-67 index without or with MIPI, 2.4). In multivariable models including CDKN2A (p16), TP53, and the Ki-67 index on 72 patients with available Ki-67 index (26 events), without or with adjustment for MIPI, the independent prognostic effect on OS of deletions in CDKN2A (p16) (HR, 4.2 without or with adjustment for MIPI) and TP53 (HR, 2.5 and 2.8 without and with adjustment for MIPI) was shown to be independent of the Ki-67 index (Table 4).

Table 4

Multivariable Cox regression analyses for overall survival including CDKN2A deletion and TP53 deletion status, excluding or including Ki-67 index (in %), without or with adjustment for MIPI score

VariableAdjustmentOS
HR95% CI (lower bound)95% CI, upper boundP
CDKN2A deletion None 5.51 2.42 12.54 <.001 
TP53 deletion  2.92 1.23 6.93 .015 
CDKN2A deletion MIPI score 4.46 1.85 10.78 .001 
TP53 deletion  2.86 1.20 6.80 .017 
CDKN2A deletion None 4.15 1.55 11.14 .005 
TP53 deletion  2.54 1.04 6.24 .041 
Ki-67 index (10% increase)  1.11 0.92 1.33 .27 
CDKN2A deletion MIPI score 4.19 1.50 11.66 .006 
TP53 deletion  2.79 1.15 6.78 .024 
Ki-67 index (10% increase)  1.03 0.83 1.28 .81 
VariableAdjustmentOS
HR95% CI (lower bound)95% CI, upper boundP
CDKN2A deletion None 5.51 2.42 12.54 <.001 
TP53 deletion  2.92 1.23 6.93 .015 
CDKN2A deletion MIPI score 4.46 1.85 10.78 .001 
TP53 deletion  2.86 1.20 6.80 .017 
CDKN2A deletion None 4.15 1.55 11.14 .005 
TP53 deletion  2.54 1.04 6.24 .041 
Ki-67 index (10% increase)  1.11 0.92 1.33 .27 
CDKN2A deletion MIPI score 4.19 1.50 11.66 .006 
TP53 deletion  2.79 1.15 6.78 .024 
Ki-67 index (10% increase)  1.03 0.83 1.28 .81 

We report here the prognostic value of CNAs detected at diagnosis in MCL patients treated in a randomized trial according to current standard of care, including a high-dose cytarabine regimen in the experimental arm.9  We show that deletions of CDKN2A (p16) and TP53 are of poor prognostic value for both OS and TTF, independently of MIPI and that CDKN2A (p16) and TP53 deletions are independent additive poor prognostic factors in both treatment arms. Moreover, we observed that the prognostic effects of CDKN2A (p16) and TP53 deletions were independent of the proliferation marker Ki-67 index.

Our CNA frequencies are in line with previously reported abnormalities detected by accurate techniques such as array or matrix comparative genomic hybridization (CGH), fluorescence in situ hybridization, real-time quantitative polymerase chain reaction (RQ-PCR), or QMPSF (supplemental Table 3). Deletions in 13q14 and ATM (11q23) are consistently the most frequently detected and in the same order of magnitude, followed by CDKN2A (9p21) and TP53 deletions.

We found that 22% of the studied patients had a TP53 deletion. The prognostic value of this abnormality is controversial in the MCL literature, with reports either showing no or a deleterious prognostic value (supplemental Table 3). However, these mostly smaller studies mixed patients with no need for treatment and patients with very aggressive disease. Moreover, treatment was heterogeneous, with <50% of patients receiving rituximab in combination with chemotherapy at induction. To our knowledge, we show here for the first time that the addition of rituximab to chemotherapy does not erase the poor prognostic value of TP53 deletion in MCL, despite a clinical improvement of disease outcome.37  The same has recently been observed in B-cell chronic lymphocytic leukemia.38  Our data from a randomized trial enable us to further demonstrate that induction treatment including high-dose cytarabine followed by ASCT, the new standard of care for younger patients, does not solve the problem of TP53 deletions for MCL patients. This is in accordance with Nordström et al, who recently demonstrated a poor prognostic value of p53 overexpression, as assessed by immunohistochemistry, in MCL patients treated with intensive immunochemotherapy including high-dose cytarabine.39 

Most of our studied genes can be grouped into 2 major pathways, related to either cell cycle regulation (CDKN1B [p27], CDK4, and RB1) or apoptosis and DNA damage response pathways (ATM/TP53/MDM2). The CDKN2A (p16) deletion is thought to alter both cell cycle regulation (p16) and DNA damage response (p14) pathways in MCL. Previous reports in aggressive lymphomas of various histology40,41  or focusing on DLBCL42  have shown that reinforcing the p14-induced deficit of apoptosis and DNA damage response pathways through TP53 deletion, in addition to CDKN2A (p16) deletion, has an additional deleterious effect on OS. We now confirm these observations in the era of new treatments and in a larger cohort of MCL patients, highlighting the biologic power of these oncogenic alterations that are not overrun yet by the new therapeutic strategies.

Those 2 major alterations that accelerate cell cycle and inhibit apoptosis can be targeted by new therapies. Recent phase 2 studies have shown high response rates of small molecules targeting the B-cell receptor pathway, including Bruton tyrosine kinase inhibitors (such as ibrutinib)43  and phosphatidylinositol-4,5-bisphosphate 3-kinase inhibitors (like idelalisib),44  as well as proteasome inhibitors (bortezomib)45  and the immunomodulatory agents (lenalidomide).46  Based on the involved pathways, it would be logical to evaluate combined approaches targeting the different pathways. In fact, preclinical data suggest that the combination of ibrutinib and ABT-19947  or ibrutinib and bortezomib48,49  has synergistic efficacy. In addition, refractoriness to one or another compound may be overcome by such a combined approach.48 

The population we studied for CNAs did not fully reflect the total population of the clinical trial, as bone marrow involvement, elevated LDH, and high-risk MIPI were more frequently observed. Because our patients were selected by the availability of a high content tumor cell sample, 42% had >50% circulating tumor cells. This selection could have affected the frequency of CNAs reported here. However, apart from the CDK2 and CDK4 gains, which tended to be more frequently detected in lymph nodes than in peripheral blood, no difference was detected between the 2 sample types analyzed. In particular, the deletion of CDKN2A (p16), which is associated with a proliferative signature,13  is not higher in frankly leukemic forms. Accordingly, the Ki-67 index was similar in studied and unstudied patients. We did not detect an additive prognostic impact of any other CNA investigated to CDKN2A (p16) and TP53 deletions. However, although our cohort of homogenously treated MCL patients analyzed for CNAs is relatively large compared with other reports, an even larger statistical power might be necessary to detect a prognostic impact especially of the more uncommon CNAs.

In conclusion, despite being associated with substantial improvement in clinical outcome, high-dose cytarabine as part of immunochemotherapy followed by ASCT does not seem to erase the bad prognostic value of CDKN2A (p16) and TP53 deletions. Furthermore, patients with simultaneous deletions of CDKN2A (p16) and TP53 show a very dismal outcome and are therefore candidates for specific therapeutic strategies. We therefore recommend that all patients benefit from CNA evaluation of CDKN2A and TP53 at diagnosis, in addition to Ki-67 index and MIPI.

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 Plateformes de Ressources Biologiques: PRB Créteil (PRB BB-0033-00021; Dijon, Lyon, Nancy, and IGR) and Dr N. Grardel and Prof de Leval who provided DNA from frozen tumor material. The authors also thank Isabelle Texier and Tony Noel from the Creteil laboratory for technical performance and Catherine Bely for secretarial assistance.

This work was supported by Institut National du Cancer (INCA), grant Programmes d'actions intégrées de recherche (PAIR Lymphome) 2008-1-LYM-01 (O.H.), Association pour la recherche sur le cancer (ARC) (subvention 3730) (M.-H.D.-L.), Fondation de France grant 2004004029 (M.-H.D.-L.), Association pour la recherche thérapeutique génétique et immunologique dans les lymphomes (ARTGIL) (C.H. and M.-H.D.-L.), Lymphoma Research Foundation (LRF) (M.D.), Framework Programmes 6 European Commission (contract LSHC-CT-2004-503351) (M.D.), and Roche (support for the MCL Younger trial) (M.D.). This work was supported in part by MRC Holland, which provided free kits for MLPA technology development.

Contributions: M.-H.D.-L. designed the research, performed genetic experiments, and drafted the manuscript; W.K., F.B., and J.B. performed pathologic review and Ki-67 counts; E.M., C.P., and M.-H.D.-L. collected and characterized tumor infiltration of diagnostic samples during the clinical trial; G.S., O.C., P.F., C.H., V.R., C.T., and M.D. included studied patients in the clinical trial, helped in tumor tissue recovery, and, with F.J. and E.M., edited the manuscript; E.H. planned and performed the statistical analysis; O.H., M.U., and M.D. coordinated the MCL Younger trial; and all authors reviewed and accepted the final version of the manuscript. A list of the European mantle cell network members is available on http://www.european-mcl.net/en/members.php.

Conflict-of-interest disclosure: M.-H.D.-L., W.K., F.B., F.J., J.B., O.C., V.R., M.U., E.M., C.P., and O.H. declare no competing financial interests. E.H. received travel support by Roche. P.F. and C.H. received honoraria for advisory board participations from Roche. G.S. received research support and honoraria for conferences or advisory board participations from Roche/Genentech and honoraria for conferences or advisory board participations from Celgene and Janssen. M.D. received speaker’s honoraria and support of investigator-initiated trials (institution) from Roche.

Correspondence: Marie-Helene Delfau-Larue, Département d’Hématologie et Immunologie Biologiques, GH Mondor, AP-HP, INSERM U955 eq9, 94000 Créteil, France; e-mail: marie-helene.delfau@hmn.aphp.fr.

1
Harris
 
NL
Jaffe
 
ES
Stein
 
H
, et al. 
A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group.
Blood
1994
, vol. 
84
 
5
(pg. 
1361
-
1392
)
2
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
 
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon, France: IARC Press; 2008
3
A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin’s lymphoma
The Non-Hodgkin’s Lymphoma Classification Project.
Blood
1997
, vol. 
89
 
11
(pg. 
3909
-
3918
)
4
Martin
 
P
Chadburn
 
A
Christos
 
P
, et al. 
Outcome of deferred initial therapy in mantle-cell lymphoma.
J Clin Oncol
2009
, vol. 
27
 
8
(pg. 
1209
-
1213
)
5
Vose
 
JM
Mantle cell lymphoma: 2012 update on diagnosis, risk-stratification, and clinical management.
Am J Hematol
2012
, vol. 
87
 
6
(pg. 
604
-
609
)
6
Harel
 
S
Delarue
 
R
Ribrag
 
V
Dreyling
 
M
Hermine
 
O
Treatment of younger patients with mantle cell lymphoma.
Semin Hematol
2011
, vol. 
48
 
3
(pg. 
194
-
207
)
7
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
)
8
Dreyling
 
M
Geisler
 
C
Hermine
 
O
, et al. 
ESMO Guidelines Working Group
Newly diagnosed and relapsed mantle cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.
Ann Oncol
2014
, vol. 
25
 
Suppl 3
(pg. 
iii83
-
iii92
)
9
Delarue
 
R
Haioun
 
C
Ribrag
 
V
, et al. 
Groupe d’Etude des Lymphomes de l’Adulte (GELA)
CHOP and DHAP plus rituximab followed by autologous stem cell transplantation in mantle cell lymphoma: a phase 2 study from the Groupe d’Etude des Lymphomes de l’Adulte.
Blood
2013
, vol. 
121
 
1
(pg. 
48
-
53
)
10
Fernàndez
 
V
Hartmann
 
E
Ott
 
G
Campo
 
E
Rosenwald
 
A
Pathogenesis of mantle-cell lymphoma: all oncogenic roads lead to dysregulation of cell cycle and DNA damage response pathways.
J Clin Oncol
2005
, vol. 
23
 
26
(pg. 
6364
-
6369
)
11
Jares
 
P
Colomer
 
D
Campo
 
E
Molecular pathogenesis of mantle cell lymphoma.
J Clin Invest
2012
, vol. 
122
 
10
(pg. 
3416
-
3423
)
12
Pinyol
 
M
Hernandez
 
L
Cazorla
 
M
, et al. 
Deletions and loss of expression of p16INK4a and p21Waf1 genes are associated with aggressive variants of mantle cell lymphomas.
Blood
1997
, vol. 
89
 
1
(pg. 
272
-
280
)
13
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
)
14
Rubio-Moscardo
 
F
Climent
 
J
Siebert
 
R
, et al. 
Mantle-cell lymphoma genotypes identified with CGH to BAC microarrays define a leukemic subgroup of disease and predict patient outcome.
Blood
2005
, vol. 
105
 
11
(pg. 
4445
-
4454
)
15
Salaverria
 
I
Zettl
 
A
Beà
 
S
, et al. 
Specific secondary genetic alterations in mantle cell lymphoma provide prognostic information independent of the gene expression-based proliferation signature.
J Clin Oncol
2007
, vol. 
25
 
10
(pg. 
1216
-
1222
)
16
Katzenberger
 
T
Kienle
 
D
Stilgenbauer
 
S
, et al. 
Delineation of distinct tumour profiles in mantle cell lymphoma by detailed cytogenetic, interphase genetic and morphological analysis.
Br J Haematol
2008
, vol. 
142
 
4
(pg. 
538
-
550
)
17
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
)
18
Jardin
 
F
Picquenot
 
JM
Parmentier
 
F
, et al. 
Detection of gene copy number aberrations in mantle cell lymphoma by a single quantitative multiplex PCR assay: clinicopathological relevance and prognosis value.
Br J Haematol
2009
, vol. 
146
 
6
(pg. 
607
-
618
)
19
Dreyling
 
MH
Bullinger
 
L
Ott
 
G
, et al. 
Alterations of the cyclin D1/p16-pRB pathway in mantle cell lymphoma.
Cancer Res
1997
, vol. 
57
 
20
(pg. 
4608
-
4614
)
20
Royo
 
C
Salaverria
 
I
Hartmann
 
EM
Rosenwald
 
A
Campo
 
E
Beà
 
S
The complex landscape of genetic alterations in mantle cell lymphoma.
Semin Cancer Biol
2011
, vol. 
21
 
5
(pg. 
322
-
334
)
21
Espinet
 
B
Salaverria
 
I
Beà
 
S
, et al. 
Incidence and prognostic impact of secondary cytogenetic aberrations in a series of 145 patients with mantle cell lymphoma.
Genes Chromosomes Cancer
2010
, vol. 
49
 
5
(pg. 
439
-
451
)
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
Hao
 
S
Sanger
 
W
Onciu
 
M
Lai
 
R
Schlette
 
EJ
Medeiros
 
LJ
Mantle cell lymphoma with 8q24 chromosomal abnormalities: a report of 5 cases with blastoid features.
Mod Pathol
2002
, vol. 
15
 
12
(pg. 
1266
-
1272
)
24
Michaux
 
L
Wlodarska
 
I
Theate
 
I
, et al. 
Coexistence of BCL1/CCND1 and CMYC aberrations in blastoid mantle cell lymphoma: a rare finding associated with very poor outcome.
Ann Hematol
2004
, vol. 
83
 
9
(pg. 
578
-
583
)
25
Gerdes
 
J
Dallenbach
 
F
Lennert
 
K
Lemke
 
H
Stein
 
H
Growth fractions in malignant non-Hodgkin’s lymphomas (NHL) as determined in situ with the monoclonal antibody Ki-67.
Hematol Oncol
1984
, vol. 
2
 
4
(pg. 
365
-
371
)
26
Tiemann
 
M
Schrader
 
C
Klapper
 
W
, et al. 
European MCL Network
Histopathology, cell proliferation indices and clinical outcome in 304 patients with mantle cell lymphoma (MCL): a clinicopathological study from the European MCL Network.
Br J Haematol
2005
, vol. 
131
 
1
(pg. 
29
-
38
)
27
Hoster
 
E
Klapper
 
W
Hermine
 
O
, et al. 
Confirmation of the mantle-cell lymphoma International Prognostic Index in randomized trials of the European Mantle-Cell Lymphoma Network.
J Clin Oncol
2014
, vol. 
32
 
13
(pg. 
1338
-
1346
)
28
Hoster
 
E
Dreyling
 
M
Klapper
 
W
, et al. 
German Low Grade Lymphoma Study Group (GLSG); European Mantle Cell Lymphoma Network
A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma.
Blood
2008
, vol. 
111
 
2
(pg. 
558
-
565
)
29
Hermine
 
O
Hoster
 
E
Walewski
 
J
, et al. 
Alternating courses of 3x CHOP and 3x DHAP plus rituximab followed by a high dose ARA-C containing myeloablative regimen and autologous stem cell transplantation (ASCT) increases overall survival when compared to 6 courses of CHOP plus rituximab followed by myeloablative radiochemotherapy and ASCT in mantle cell lymphoma: final analysis of the MCL Younger Trial of the European Mantle Cell Lymphoma Network (MCL net).
ASH Annu Mtg Abstr
2012
, vol. 
120
 
21
pg. 
151
 
30
Pott
 
C
Hoster
 
E
Delfau-Larue
 
M-H
, et al. 
Molecular remission is an independent predictor of clinical outcome in patients with mantle cell lymphoma after combined immunochemotherapy: a European MCL intergroup study.
Blood
2010
, vol. 
115
 
16
(pg. 
3215
-
3223
)
31
Klapper
 
W
Histopathology of mantle cell lymphoma.
Semin Hematol
2011
, vol. 
48
 
3
(pg. 
148
-
154
)
32
Klapper
 
W
Hoster
 
E
Determann
 
O
, et al. 
European MCL Network
Ki-67 as a prognostic marker in mantle cell lymphoma-consensus guidelines of the pathology panel of the European MCL Network.
J Hematop
2009
, vol. 
2
 
2
(pg. 
103
-
111
)
33
Determann
 
O
Hoster
 
E
Ott
 
G
, et al. 
European Mantle Cell Lymphoma Network and the German Low Grade Lymphoma Study Group
Ki-67 predicts outcome in advanced-stage mantle cell lymphoma patients treated with anti-CD20 immunochemotherapy: results from randomized trials of the European MCL Network and the German Low Grade Lymphoma Study Group.
Blood
2008
, vol. 
111
 
4
(pg. 
2385
-
2387
)
34
van Dongen
 
JJM
Langerak
 
AW
Brüggemann
 
M
, et al. 
Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2257
-
2317
)
35
van Veghel-Plandsoen
 
MM
Wouters
 
CH
Kromosoeto
 
JNR
den Ridder-Klünnen
 
MC
Halley
 
DJ
van den Ouweland
 
AM
Multiplex ligation-depending probe amplification is not suitable for detection of low-grade mosaicism.
Eur J Hum Genet
2011
, vol. 
19
 
9
(pg. 
1009
-
1012
)
36
Coll-Mulet
 
L
Santidrián
 
AF
Cosialls
 
AM
, et al. 
Multiplex ligation-dependent probe amplification for detection of genomic alterations in chronic lymphocytic leukaemia.
Br J Haematol
2008
, vol. 
142
 
5
(pg. 
793
-
801
)
37
Lenz
 
G
Dreyling
 
M
Hoster
 
E
, et al. 
Immunochemotherapy with rituximab and cyclophosphamide, doxorubicin, vincristine, and prednisone significantly improves response and time to treatment failure, but not long-term outcome in patients with previously untreated mantle cell lymphoma: results of a prospective randomized trial of the German Low Grade Lymphoma Study Group (GLSG).
J Clin Oncol
2005
, vol. 
23
 
9
(pg. 
1984
-
1992
)
38
Stilgenbauer
 
S
Schnaiter
 
A
Paschka
 
P
, et al. 
Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial.
Blood
2014
, vol. 
123
 
21
(pg. 
3247
-
3254
)
39
Nordström
 
L
Sernbo
 
S
Eden
 
P
, et al. 
SOX11 and TP53 add prognostic information to MIPI in a homogenously treated cohort of mantle cell lymphoma—a Nordic Lymphoma Group study.
Br J Haematol
2014
, vol. 
166
 
1
(pg. 
98
-
108
)
40
Grønbaek
 
K
de Nully Brown
 
P
Møller
 
MB
, et al. 
Concurrent disruption of p16INK4a and the ARF-p53 pathway predicts poor prognosis in aggressive non-Hodgkin’s lymphoma.
Leukemia
2000
, vol. 
14
 
10
(pg. 
1727
-
1735
)
41
Pinyol
 
M
Hernández
 
L
Martínez
 
A
, et al. 
INK4a/ARF locus alterations in human non-Hodgkin’s lymphomas mainly occur in tumors with wild-type p53 gene.
Am J Pathol
2000
, vol. 
156
 
6
(pg. 
1987
-
1996
)
42
Jardin
 
F
Ruminy
 
P
Kerckaert
 
J-P
, et al. 
Detection of somatic quantitative genetic alterations by multiplex polymerase chain reaction for the prediction of outcome in diffuse large B-cell lymphomas.
Haematologica
2008
, vol. 
93
 
4
(pg. 
543
-
550
)
43
Wang
 
ML
Rule
 
S
Martin
 
P
, et al. 
Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma.
N Engl J Med
2013
, vol. 
369
 
6
(pg. 
507
-
516
)
44
Kahl
 
BS
Spurgeon
 
SE
Furman
 
RR
, et al. 
A phase 1 study of the PI3Kδ inhibitor idelalisib in patients with relapsed/refractory mantle cell lymphoma (MCL).
Blood
2014
, vol. 
123
 
22
(pg. 
3398
-
3405
)
45
Goy
 
A
Bernstein
 
SH
Kahl
 
BS
, et al. 
Bortezomib in patients with relapsed or refractory mantle cell lymphoma: updated time-to-event analyses of the multicenter phase 2 PINNACLE study.
Ann Oncol
2009
, vol. 
20
 
3
(pg. 
520
-
525
)
46
Goy
 
A
Sinha
 
R
Williams
 
ME
, et al. 
Single-agent lenalidomide in patients with mantle-cell lymphoma who relapsed or progressed after or were refractory to bortezomib: phase II MCL-001 (EMERGE) study.
J Clin Oncol
2013
, vol. 
31
 
29
(pg. 
3688
-
3695
)
47
Zhao
 
X
Bodo
 
J
Sun
 
D
, et al. 
Combination of ibrutinib with ABT-199: synergistic effects on proliferation inhibition and apoptosis in mantle cell lymphoma cells through perturbation of BTK, AKT and BCL2 pathways.
Br J Haemato
2015
, vol. 
168
 
5
(pg. 
765
-
768
)
48
Axelrod
 
M
Ou
 
Z
Brett
 
LK
, et al. 
Combinatorial drug screening identifies synergistic co-targeting of Bruton’s tyrosine kinase and the proteasome in mantle cell lymphoma.
Leukemia
2014
, vol. 
28
 
2
(pg. 
407
-
410
)
49
Portell
 
CA
Axelrod
 
M
Brett
 
LK
, et al. 
Synergistic cytotoxicity of ibrutinib and the BCL2 antagonist, ABT-199(GDC-0199), in mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL): molecular analysis reveals mechanisms of target interactions.
Blood
2014
, vol. 
124
 
21
pg. 
509
 
Sign in via your Institution