ArticlePDF Available

Immunohistochemical and genomic profiles of diffuse large B-cell lymphomas: Implications for targeted EZH2 inhibitor therapy?

Authors:

Abstract and Figures

Enhancer of Zeste Homolog 2 (EZH2) plays an essential epigenetic role in Diffuse Large B Cell Lymphoma (DLBCL) development. Recurrent somatic heterozygous gain-of-function mutations of EZH2 have been identified in DLBCL, most notably affecting tyrosine 641 (Y641), inducing hyper-trimethylation of H3K27 (H3K27me3). Novel EZH2 inhibitors are being tested in phase 1 and 2 clinical trials but no study has examined which patients would most benefit from this treatment. We evaluated the immunohistochemical (IHC) methylation profiles of 82 patients with DLBCL, as well as the mutational profiles of 32 patients with DLBCL using NGS analysis of a panel of 34 genes involved in lymphomagenesis. A novel IHC score based on H3K27me2 and H3K27me3 expression was developed, capable of distinguishing patients with wild-type (WT) EZH2 and patients with EZH2 Y641 mutations (p = 10-5). NGS analysis revealed a subclonal EZH2 mutation pattern in EZH2 mutant patients with WT-like IHC methylation profiles, while associated mutations capable of upregulating EZH2 were detected in WT EZH2 patients with mutant-like IHC methylation profiles. IHC and mutational profiles highlight in vivo hyper-H3K27me3 and hypo-H3K27me2 status, pinpoint associated activating mutations and determine EZH2 mutation clonality, maximizing EZH2 inhibitor potential by identifying patients most likely to benefit from treatment.
Content may be subject to copyright.
Oncotarget16712
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No. 18
Immunohistochemical and genomic profiles of diffuse large
B-cell lymphomas: Implications for targeted EZH2 inhibitor
therapy?
Sydney Dubois1, Sylvain Mareschal1, Jean-Michel Picquenot1,2, Pierre-Julien
Viailly1, Elodie Bohers1, Marie Cornic2, Philippe Bertrand1, Elena Liana Veresezan1,2,
Philippe Ruminy1, Catherine Maingonnat1, Vinciane Marchand1, Hélène Lanic1,3,
Dominique Penther1, Christian Bastard1, Hervé Tilly1,3, Fabrice Jardin1,3
1INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
2Department of Pathology, Centre Henri Becquerel, Rouen, France
3Department of Clinical Hematology, Centre Henri Becquerel, Rouen, France
Correspondence to:
Fabrice Jardin, e-mail: fabrice.jardin@chb.unicancer.fr
Keywords: DLBCL, EZH2, Methylation, Immunohistochemistry, NGS
Received: October 20, 2014 Accepted: January 15, 2015 Published: February 05, 2015
ABSTRACT
Enhancer of Zeste Homolog 2 (EZH2) plays an essential epigenetic role in Diffuse
Large B Cell Lymphoma (DLBCL) development. Recurrent somatic heterozygous gain-
of-function mutations of EZH2 have been identified in DLBCL, most notably affecting
tyrosine 641 (Y641), inducing hyper-trimethylation of H3K27 (H3K27me3). Novel
EZH2 inhibitors are being tested in phase 1 and 2 clinical trials but no study has
examined which patients would most benefit from this treatment. We evaluated the
immunohistochemical (IHC) methylation profiles of 82 patients with DLBCL, as well
as the mutational profiles of 32 patients with DLBCL using NGS analysis of a panel of
34 genes involved in lymphomagenesis. A novel IHC score based on H3K27me2 and
H3K27me3 expression was developed, capable of distinguishing patients with wild-
type (WT) EZH2 and patients with EZH2 Y641 mutations (p = 10−5). NGS analysis
revealed a subclonal EZH2 mutation pattern in EZH2 mutant patients with WT-like
IHC methylation profiles, while associated mutations capable of upregulating EZH2
were detected in WT EZH2 patients with mutant-like IHC methylation profiles. IHC and
mutational profiles highlight in vivo hyper-H3K27me3 and hypo-H3K27me2 status,
pinpoint associated activating mutations and determine EZH2 mutation clonality,
maximizing EZH2 inhibitor potential by identifying patients most likely to benefit
from treatment.
Diffuse large B-cell lymphoma (DLBCL) is the
most common lymphoid malignancy, accounting for 30–
40% of all Non Hodgkin Lymphomas (NHL) [1]. Gene
expression proling has identied two main subtypes:
Germinal Center B-cell like (GCB) and Activated
B-Cell like (ABC), with the ABC subtype having the
most unfavorable prognosis [2, 3]. The development of
immuno-chemotherapy, and most notably rituximab, has
revolutionized the standard-of-care treatment of DLBCL
but a large part of patients still relapses or is refractory to
treatment.
Recently, epigenetic regulation has been shown to
be a crucial element in DLBCL development, and gene
repression mediated by Polycomb Repressive Complexes
1 and 2 (PRC1 and PRC2) has garnered attention.
Enhancer of Zeste Homolog 2 (EZH2), the catalytic
subunit of PRC2 [4], is a histone methyl-transferase
capable of specically mono-, di- and tri-methylating
histone H3 lysine 27 (H3K27me1, H3K27me2, and
H3K27me3) [5].
Recurrent somatic heterozygous gain-of-function
mutations of EZH2 have been identied in DLBCL, most
notably affecting tyrosine 641 (Y641), inducing increased
H3K27me3 [6, 7]. More recently, multiple studies have
shown cell lines with EZH2 mutations to be dependent
on the higher catalytic activity of mutant EZH2 Y641
Oncotarget16713
www.impactjournals.com/oncotarget
for proliferation, leading to the development of novel
EZH2 inhibitors for therapeutic use, capable of reversing
malignant phenotype [8–11].
Two EZH2 inhibitors are currently being tested
in phase 1 and 2 clinical trials both in patients with and
without EZH2 Y641 mutations (NCT01897571 and
NCT02082977), but no study has specically examined
which patients would be most susceptible to benet from
this treatment and how to screen for them. Patients with
EZH2 gain-of-function mutations have been pinpointed
as ideal EZH2 inhibitor recipients [8–11]; nevertheless, in
today’s targeted therapy era, it seems essential to establish
a method of detecting optimal candidates for EZH2
inhibitor treatment.
In the current study, we examined whether a simple
immunohistochemical (IHC) technique could be used to
distinguish wild-type (WT) -like and mutant-like EZH2
IHC methylation proles, and thus screen for patients
with conrmed overactive EZH2 at the protein level. We
also used Next Generation Sequencing (NGS) analysis to
further detail patients’ genomic proles and to determine
whether associated mutations could justify EZH2 inhibitor
treatment for patients otherwise not considered. We
propose that these methods, used in conjunction, could
serve to better determine candidates most likely to respond
to EZH2 inhibitor treatment.
MATERIALS & METHODS
Patients and biological samples
96 patients with de novo DLBCL at diagnosis
with available tumor DNA and Formalin-Fixed
Parafn-Embedded (FFPE) samples were included
for EZH2 Sanger sequencing analysis and subsequent
immunohistochemistry experiments. To provide a
comprehensive genomic description of DLBCL, targeted
NGS experiments were performed in 32 patients (20/96
and 12 additional cases not in our initial cohort). A
owchart summarizes the experimental methods used
on the entire cohort (Supplementary Figure 1). Table 1
summarizes the patients’ clinical characteristics.
Median follow-ups for overall survival and progression-
free survival were respectively 4.9 and 3.9 years. All
experiments were in accordance with the Helsinki
Declaration and the study was approved by the internal
review board.
Immunohistochemistry
Sections from FFPE tissue samples were used
to build Tissue Microarrays (TMAs). Information
on the primary antibodies used in this study (EZH2,
H3K27me1, H3K27me2 and H3K27me3) is summarized
in Supplementary Table 1. Deparafnization, rehydration,
and epitope retrieval was performed by PT Link
following the manufacturer’s instructions at pH 6
(DAKO, California, USA) and deparafnized sections
were stained using Vectastain kits (Vector Laboratories
Inc, California, USA) according to the manufacturer’s
instructions. The slides were then incubated with DAB+
chromogen for 5 minutes and counterstained with
hematoxyline for 2 minutes. Slides were scored in a
blinded fashion by an experienced anatomopathologist
(JMP). Slides were also scored in a blinded fashion by
a second independent anatomopathologist (ELV) in
order to assess correlation. Cases with lost TMA cores or
non-tumoral tissue were excluded. Tumors were scored
according to staining intensity (1–3, with 1 being weak
and 3 strong) and proportion of tumor cells stained (0–
10, with 0 representing negative staining, 1 representing
1–10% of positive tumor cells and 10 representing 91–
100% of positive tumor cells). For each antibody, a score
that ranged from 0 to 30 was calculated as the product
of staining intensity and proportion of tumor cells stained
[12]. Each tumor was represented 3 times on the TMAs
and the highest score was kept. For each patient, a me3/
me2 score was calculated:
me3
me2 score = log 2
a
me3 score +1
me2
score +1
GCB/ABC cell of origin (COO) subclassication
The GCB/ABC subtype was determined by cDNA-
mediated Annealing, Selection, extension, and Ligation
(DASL) technology based on the expression of 19 genes,
as previously described [13].
Ion torrent personal genome machine (PGM)
sequencing
Genomic DNA was submitted to Next Generat-
ion Sequencing (NGS) using a laboratory-developed
“Lymphopanel” set, designed to identify mutations in 34
genes important for lymphomagenesis (Supplementary
Table 2). This design covers 87 703 bases and generates
872 amplicons. Amplied libraries were submitted to
emulsion PCR with the Ion OneTouch™ 200 Template
Kit (Life Technologies, California, USA) using the Ion
OneTouch™ System (Life Technologies) according to the
manufacturer’s instructions. The generated Ion Sphere™
Particles (ISPs) were enriched with the Ion OneTouch™
Enrichment System and loaded and sequenced on Ion
316™ v2 Chips (Life Technologies).
PGM data analysis
Torrent Suite™ version 4.0 (Life Technologies)
software was used to perform primary analysis, including
signal processing, base calling, sequence alignment to
the reference genome (hg19) and generation of Binary
Alignment/Map (BAM) les. BAM les were used by
Torrent Suite™’s Variant Caller to detect point mutations
Oncotarget16714
www.impactjournals.com/oncotarget
as well as short insertions and deletions using the PGM
Somatic Low Stringency prole. VCF les generated by
Variant Caller were annotated by ANNOVAR [14].
Samples were considered of sufcient quality
when more than 90% of targeted bases were read at
least 20 times with sequencing and mapping precisions
of at least Q20. Only frameshift deletions and insertions,
nonframeshift deletions and substitutions, splicing,
nonsynonymous, stopgain or stoploss Single Nucleotide
Variations (SNVs) were kept. Variants present in dbSNP
(version 138) and absent in COSMIC (version 64)
were discarded, as were variants with a predictive SIFT
score > 0.05 [15]. A normal probability plot dened
thresholds separating true positives (conrmed by
Sanger sequencing, TVC score ≥ 22) from true negatives
(discredited by Sanger sequencing, TVC score < 9.5)
and highlighted a gray zone (9.5 < TVC score < 22) in
which variants must be conrmed by Sanger sequencing
or pyrosequencing.
Further verication by Sanger sequencing was
performed using a BigDye® Terminator v3.1 Cycle
Sequencing Kit (Life Technologies) and an ABI PRISM
3130 analyzer (Life Technologies). Primer sequences are
provided in Supplementary Table 3. Further verication by
pyrosequencing was performed using the PyroMark PCR
kit (Qiagen, France) with internal and sequencing primers
designed using PyroMark software (Qiagen). Bubble
charts to visualize validated variants per patient were
generated usingHighcharts.com (Highsoft AS, Norway).
Karyotyping and uorescent in situ
hybridization (FISH)
Cytogenetic analysis was performed according to
standard techniques. Slides were RHG-banded according
to Sehested [16] and karyotypes were described according
to the International System for Human Cytogenetic
Nomenclature. FISH using the LSI IGH/BCL2 Dual
Color, Dual Fusion Translocation Probe (Vysis, Downers
Grove, USA) was performed on metaphase preparations
according to the manufacturer’s instructions.
Statistical analysis
All statistical analyses except kappa scores were
performed using R software version 3.0.2 [17]. Kappa
scores were calculated using Medcalc software version
10.0.2.0. Overall Survival (OS) was calculated from
beginning of treatment to date of death or last patient
follow-up. Progression-Free Survival (PFS) was calculated
Table 1: Clinical characteristics of patients at diagnosis
Clinical parameter Patients at diagnosis (n = 96)
Gender M/F, n48/48
Age (years), median (range) 66 (17–87)
Adverse prognostic factors, n (%)
Age > 60 years 60 (63)
Ann Arbor stage III–IV 68 (71)
LDH > N 9 (9)
Extranodal sites ≥ 2 37 (39)
Bulky mass ≥ 10 cm 20 (21)
Performance status ≥ 2 26 (27)
IPI, n (%)
0–2 42 (44)
3–5 54 (56)
Treatment, n (%)
R-CHOP 38 (40)
R-ACVBP 17 (18)
R-mCHOP 13 (14)
R-IVA 1 (1)
Abbreviations: LDH, Lactate Dehydrogenase; IPI, International Prognostic Index; R, Rituximab; CHOP,
Cyclophosphamide, Hydroxydaunorubicin, Vincristine and Prednisone; ACVBP, Doxorubicin, Cyclophosphamide,
Vindesine, Bleomycin and Prednisone; mCHOP, miniCHOP; IVA, Ifosfamide, Etoposide and Cytarabine
Oncotarget16715
www.impactjournals.com/oncotarget
from beginning of treatment until disease progression,
relapse, death or last patient follow-up. Log-rank tests
(“survival” R package version 2.37.7) were used to assess
differences in OS and PFS rates calculated by Kaplan-
Meir estimates, as well as to perform univariate analysis.
Multivariate analysis was performed with a Cox regression
model. K-means cluster analysis was performed, with
cluster number set to k = 2. Statistical differences between
all other parameters were determined using χ2, Mann–
Whitney, or Fisher’s exact test when appropriate. p values
< 0.05 were considered statistically signicant.
RESULTS
Patient characteristics according to EZH2
somatic mutation status
Table 2 classies all patients with DASL and
Sanger sequencing data available based on their COO
subtype and EZH2 mutation status, and also highlights
the 82 patients usable for IHC. Of the 49 GCB subtype
patients, 12 were EZH2 Y641 mutant (24%), slightly
higher than the original report of 22% [18]. One EZH2
mutant patient in our 100-patient cohort was of the ABC
subtype, examples of which have been reported in the
literature [19]. IHC-usable WT EZH2 patients were
quite evenly split between ABC (n = 37/70) and GCB
(n = 30/70) subtype, while all IHC-usable EZH2 mutant
patients were of the GCB subtype (n = 12/12), as is
most frequent [18, 20]. EZH2 Y641 mutations showed
signicant association with t(14;18) translocation in
our cohort ( p < 10−4), corroborating previous studies
(Table 2) [20, 21].
Differential methylation levels of H3K27 are
distinguishable by IHC
FFPE samples of DLBCL placed on TMAs were
used for IHC with antibodies targeting EZH2, H3K27me1,
H3K27me2 and H3K27me3 (Supplementary Table 1).
We used breast cancer samples of different histological
subtypes, as well as DLBCL samples, as a guide to
determine primary antibody concentrations and incubation
times in order to observe gradients of EZH2 and H3K27
methylation IHC expression [12]. Figures 1A–1D show
representative images of differential IHC expression
of H3K27me2 and H3K27me3 from samples with WT
or Y641 mutant EZH2. Larger versions of the same
images are shown in Supplementary Figure 2. EZH2 IHC
expression was also able to showcase differential levels
of expression (not shown). H3K27me1 IHC expression
showed high expression levels for all patients, with no
differences observed (not shown).
Patients with EZH2 Y641 mutations present
distinct IHC methylation proles
There was no signicant difference in EZH2 or
H3K27me1 IHC expression between patients with mutant
and WT EZH2 (Table 2). Patients with EZH2 Y641 mutations
presented a signicantly lower H3K27me2 score ( p = 0.005)
and a signicantly higher H3K27me3 score ( p = 0.01) than
patients with WT EZH2 (Table 2). Hyper-trimethylation and
hypo-dimethylation in patients with EZH2 Y641 mutations
is therefore evident at the IHC level. There was no signicant
difference in either EZH2 or H3K27me1/2/3 IHC scores
between ABC and GCB subtypes (data not shown).
Table 2: Patients according to their EZH2 mutation status
Characteristics Total WT EZH2 EZH2 Y641 mutant p-value
Patients, n92 78 14
me3/me2 score usable, n82 70 12 0.65a
EZH2 IHC score, median (range) 18 (0–30) 18 (0–30) 21 (0–27) 0.8b
H3K27me1 IHC score, median (range) 30 30 30 1b
H3K27me2 IHC score, median (range) 27 (0–27) 27 (0–27) 18 (0–27) 0.005b
H3K27me3 IHC score, median (range) 18 (0–30) 18 (0–30) 27 (0–27) 0.01b
me3/me2 score, median (range) 0 (–4.8–4.8) –0.25 (–4.8–3.3) 0.56 (–0.56–4.8) 8.30E–05b
t(14;18), n17 89 3.50E–05a
Age (years), median (range) 66 (17–87) 66 (17–87) 63 (37–77) 0.23b
IPI: 0–2/3–5, n40/52 32/46 8/6 0.38a
GCB / ABC, n49/43 36/39 12/1 0.005a
aFisher’s Exact Test
bWilcoxon Rank Sum Test
Abbreviations: IPI, International Prognostic Index; GCB, Germinal Center B-Cell-like; ABC, Activated B-Cell-like
Oncotarget16716
www.impactjournals.com/oncotarget
We thus decided to implement a score based on the
ratio of me3 and me2 expression levels, in order to take
into account both criteria and gain statistical strength.
A logarithmic approach was used to obtain a wider
distribution (me3/me2 score detailed in methods).
Y641 EZH2 mutant patients had signicantly higher
me3/me2 scores than patients with WT EZH2 (p < 10−4)
(Figure 1E and Table 2) As me3/me2 scores for patients
with WT or mutant EZH2 overlapped at zero, three distinct
IHC methylation proles emerged, centered around zero: a
H3K27me3-high/H3K27me2-low prole (me3/me2 score
> 0, n = 12/82), a H3K27me3-low/H3K27me2-high prole
(me3/me2 score < 0, n = 38/82) and an intermediate
prole (me3/me2 score = 0, n = 32/82). Blinded analysis
by an independent pathologist without prior consultation
rendered a weighted kappa score of 0.55 (Kmax = 0.8,
k = 69% of Kmax).
The me3/me2 score is capable of distinguishing
patients based on their EZH2 mutation status. Indeed,
patients with EZH2 Y641 mutations mostly exhibit a
H3K27me3-high/H3K27me2-low prole (n = 7/12),
with 4/12 exhibiting an intermediate prole and 1/12
exhibiting a H3K27me3-low/H3K27me2-high prole.
On the other hand, patients with WT EZH2 status are
split between intermediate (n = 28/70) and H3K27me3-
low/H3K27me2-high proles (n = 37/70) (p = 10−5).
The maximum accuracy of the me3/me2 score was
88%, demonstrated for a threshold > 0 (Supplementary
Figure 3), leading us to merge patients with me3/me2
scores ≤ 0 into a single WT-like IHC methylation prole
group, compared to the me3/me2 score > 0 mutant-like
IHC methylation prole group.
NGS mutational proles allow more thorough
understanding of IHC methylation proles
In order to better understand the unexpected
IHC methylation proles observed for certain patients
of our cohort, we performed an NGS analysis of their
mutational proles using our Lymphopanel set of genes
Figure 1: Differential IHC H3K27me2/me3 expression can distinguish WT and mutant EZH2 DLBCL. (A–D) All images
are taken at 20× magnication. (A) and (B) are images from the same WT EZH2 tumor sample. (C) and (D) are images from the same Y641
EZH2 mutant sample. IHC scores for images A–D are respectively 27/30, 9/30, 9/30 and 27/30. (E) is a boxplot representation of me3/
me2 score according to EZH2 mutation status, showing signicantly higher score in EZH2 mutant tumor samples. The width of bars in E
is proportionate to sample size. p-values in E were calculated by a Mann–Whitney test.
Oncotarget16717
www.impactjournals.com/oncotarget
based on literature data obtained from whole exome
sequencing [22]. To this end, we sequenced all Y641
EZH2 mutant patients, as well as all WT EZH2 patients
with mutant-like IHC methylation proles. We also
included 12 additional patients to extend NGS analysis
to a total of 15 Y641 EZH2 mutant patients (13 GCB,
1 ABC, 1 other) and 17 WT EZH2 patients (13 ABC, 2
GCB, 2 other).
NGS results were sorted by quality scores and
Sanger or pyrosequencing when possible, as described
in the methods section (detailed in Supplementary
Table 4). The average overall depth was 215x and
the average depth for EZH2 Y646 codon was 414x.
A total of 127 variants were validated in this fashion
(Supplementary Table 5).
All EZH2 Y641 mutations found by Sanger
sequencing were conrmed by NGS, and their VAFs
as shown were calculated as the percentage of mutant
reads among total number of reads. No additional EZH2
Y641 mutations were found by NGS among our cohort,
and no A677 or A687 mutations were identied either.
The 15 EZH2 Y641 mutants were therefore exclusively
mutated at position Y641 and the 17 EZH2 WT patients
were conrmed to be WT. VAFs for EZH2 mutations
calculated by pyrosequencing were highly correlated with
VAFs calculated by NGS analysis (Pearson’s r = 0.93,
p < 10−5), legitimizing our NGS calculation method
of VAFs for the other genes of the Lymphopanel
(Supplementary Figure 4).
Variants and their VAFs were represented in a
bubble chart format according to their COO subtype
(Figure 2A, 2B).
EZH2 mutations are majoritarily clonal in
DLBCL
The clonal status of EZH2 mutations was
established by comparing VAFs of EZH2 Y641 mutations
with the average of those of the associated mutations
in each patient. Although the direct comparison was
complicated by taking into account all VAFs available
(including those > 50% potentially due to CNVs), we
were able to distinguish two different patterns for EZH2
mutations. The majority of EZH2 mutations (n = 12/15,
80%) represented true clonal events with similar VAFs
for other genes mutated in the same sample (Figure
2A, blue bubbles and Figure 2B, patient 445). Of note,
patient 1687 seems to present a clonal mutation of EZH2
but low tumor content. True subclonal EZH2 mutations,
with lower EZH2 mutation VAFs compared to other
mutations, were found in 3/15 (20%) samples (Figure
2A, orange bubbles). K-means clustering was performed
to separate clonal and subclonal mutations (k = 2) and
successfully segregated these three patients (Figure 3).
The percentages of clonal and subclonal EZH2 mutations
in our cohort are very similar to those found in a cohort
of 43 Follicular Lymphomas (FL) in a recent study by
Bödör et al [23]. A recent study in DLBCL also found
a similar distribution of clonal versus subclonal EZH2
mutations [24].
Interestingly, three EZH2 mutant GCB patients
(1528, 1639 and 1478) also harbor a mutation in MYD88.
While they remain anectodal, given the low sample size,
two of these (1639 and 1478) present similar VAFs in
both EZH2 and MYD88 mutations (38.9% and 36.4%
respectively for 13944 and 23.8% and 27.4% respectively
for 16995), indicating driver/clonal mutation status for
both EZH2 and MYD88. On the other hand, sample 1528
hosts an EZH2 mutation with a low VAF of 6% and a
MYD88 mutation with 38.1% VAF, suggesting a driver
MYD88 mutation with a subsequent EZH2 mutation,
indicative of a secondary EZH2 mutation acquisition in a
de novo case of DLBCL.
Higher number of Lymphopanel variants among
the GCB subtype
On average, patients of GCB subtype (n = 15)
presented 5.2 validated variants among the Lymphopanel
genes (Figure 2A) while patients of ABC subtype
(n = 13) presented only 2.9 validated variants (Figure 2B)
(p = 0.02). Only 1 GCB patient (6.7%) presented no
variants according to our criteria, compared to 3 ABC
patients (23.1%).
Furthermore, there were 11 cases of genes
displaying more than 1 variant in GCB patients (n =
9/15, 60%), and only 4 such cases in ABC patients (n =
3/13, 23.1%). Such genes in GCB patients in our cohort
included KMT2D, GNA13 and CREBBP (respectively 4,
4, and 2 cases each of patients with more than 1 variant).
This mutational prole was very similar to that described
in FL [23]. By contrast, such cases in ABC patients
were evenly distributed among 4 genes (PIM1, PRDM1,
TNFAIP3 and TNFRSF14), with 1 case in each, indicating
no particular variability hotspot.
Subclonal and low-VAF EZH2 mutations may
explain unexpected WT-like IHC proles
Potentially contributing to explain EZH2 mutant
patients with WT-like IHC methylation proles, we noted
that, despite small sample size, the me3/me2 score tended
to correlate with EZH2 mutation VAF (p = 0.09, Pearson’s
r = 0.51). Of the ve EZH2 mutant patients presenting
a me3/me2 score 0 (304, 494, 1524, 1528 and 1623),
two (1528 and 1623) present low VAFs of 6% and 8.4%
respectively. These two patients also exhibit a subclonal
EZH2 mutation, as determined by NGS and clustering
(Figure 2A and Figure 3), suggesting that EZH2 inhibitor
treatment might be less efcient.
Oncotarget16718
www.impactjournals.com/oncotarget
Figure 2: Genomic proles of patients according to DLBCL subtype. Validated variants for each patient are plotted in a bubble chart,
with bubble size reecting variant VAF, not corrected for CNVs. Patients are represented by Unique Personal Number (UPN). The value of
each sample’s me3/me2 score is shown, with NA corresponding to samples not present in our IHC study. Genes are ordered from most frequent
to least frequent, with EZH2 rst. (A) represents all GCB subtype patients with at least one mutation in our cohort, with clonal and subclonal
EZH2 mutations outlined in blue and orange respectively. (B) Represents all ABC subtype patients with at least one mutation in our cohort.
Oncotarget16719
www.impactjournals.com/oncotarget
Associated mutations may explain unexpected
IHC methylation proles
The idea behind establishing mutational proles for
patients was to identify associated mutations, which might
give reasonable cause to accept or deny treatment options,
including EZH2 inhibitors, for a patient.
In our cohort, ve WT EZH2 patients (1768, 1342,
1631, 773 and 478) presented a mutant-like IHC methylation
prole. Four of these patients are of the ABC subtype,
suggesting a potential EZH2 mutation bypass in ABC
patients. Furthermore, two of these patients (1768 and 1631)
showed remarkably similar mutational proles (Figure 2B),
both of them harboring mutations in TP53, MYD88, and
PRDM1, whereas no other ABC-subtype patient in our
cohort exhibited an association of either of these mutated
genes. An additional mutation in PIM1 (patient 1768)
proved interesting as well, as these were the only cases of
mutations in PRDM1 and/or PIM1 in our cohort.
Low EZH2 IHC expression is associated
with better prognosis in ABC-DLBCL
Survival analysis was performed on the 70 patients
treated with R-chemotherapy, as detailed in Table 1. The
median follow-up for OS and PFS was 5.1 and 4.5 years
respectively.
Following the thresholds dened by a previous
study [25], low EZH2 IHC expression (< 70% of tumoral
cells stained) was observed in 36% of patients (55% ABC
and 39% GCB), whereas high EZH2 IHC expression
(≥ 70% of tumoral cells stained) was observed in 64% of
patients (41% ABC and 53% GCB). In univariate analysis,
low EZH2 IHC expression was signicantly associated
with superior OS ( p = 0.035, OS = 77% at 3 years versus
35%) and PFS ( p = 0.02, PFS = 77% at 3 years versus
29%) in ABC patients treated with R-chemotherapy
(Figure 4A, 4B). However, in a multivariate analysis
including IPI and EZH2 IHC expression in this ABC-
DLBCL subgroup, neither low EZH2 IHC expression
nor IPI was a statistically signicant prognostic factor,
with low sample number potentially responsible for
this drawback. Of note, the prognostic impact of
EZH2 expression was not observed in GCB patients
(Figure 4C, 4D). Furthermore, no correlation was found
between prognosis and IHC methylation prole in our
cohort (data not shown) [23, 26].
DISCUSSION
We have analyzed EZH2, H3K27me1, H3K27me2
and H3K27me3 IHC expression in relation to EZH2 somatic
mutation status in a cohort of patients with DLBCL and
shown that a simple IHC experiment is able to distinguish
Figure 3: Clustering by EZH2 mutation VAF relative to associated mutation VAFs enables subclonal mutation
detection. The log ratio of EZH2 VAF and average of associated mutation VAFs was calculated for each patient. K means clustering (k = 2)
was performed and isolated patients 1528, 1251 and 1623 as a unique group with subclonal EZH2 mutations. Horizontal lines indicate
means for each cluster and vertical dotted lines represent each point’s distance to the cluster’s mean.
Oncotarget16720
www.impactjournals.com/oncotarget
patients with WT EZH2 and patients with EZH2 Y641
mutations according to their me3/me2 score in the majority
of cases. This result conrms the accumulation of steady
state levels of H3K27me2 in WT EZH2 patients and the
increase in H3K27me3 levels with lower H3K27me2 steady
state levels in patients with EZH2 Y641 mutations at the
IHC level. To our knowledge, this is the rst such study in
DLBCL. A previous study showed variable H3K27me3 and
EZH2 IHC expression regardless of EZH2 mutation status in
FL and H3K27me2 IHC expression was not analyzed [26].
We have also shown that no signicant difference
exists between patients with WT or mutant EZH2 in either
EZH2 or H3K27me1 IHC expression. Lower H3K27me1
expression could have been expected in EZH2 mutant
samples; however, decreased H3K27me1 in EZH2 mutant
cell lines is not always observed [6] and H3K27me1
formation can also be catalyzed by noncanonical PRC2
complexes containing WT EZH1 [27]. The lack of
difference in EZH2 IHC expression between patients
with WT or mutant EZH2, previously shown in FL [26],
conrms that the mutation mostly affects EZH2 activity,
although a recent study has identied a mechanism by
which it also affects EZH2 stability [28].
Most importantly, our me3/me2 score highli-
ghts patients with “mutant-like” and “WT-like” IHC
methylation proles. In patients with DLBCL, our IHC
assay should be carried out alongside Sanger sequencing
for EZH2. We propose that when both parameters are
concordant, no further testing would be necessary: EZH2
mutant patients with mutant-like IHC methylation proles
would be recommended for EZH2 inhibitor treatment,
whereas WT EZH2 patients with WT-like IHC methylation
Figure 4: Low IHC EZH2 expression is a positive prognostic indicator in ABC-DLBCL. Survival was calculated on ABC-
subtype and GCB-subtype patients with R-chemotherapy treatment (n = 30 and n = 31 respectively), divided into EZH2-low (< 70%) and
EZH2-high (≥ 70%) groups. (A) and (B) show OS and PFS respectively, calculated for ABC subtype patients. (C) and (D) show OS and
PFS respectively, calculated for GCB subtype patients. Low EZH2 expression is associated with signicantly higher OS and PFS in ABC-
DLBCL patients, whereas no difference is observed in GCB-DLBCL patients.
Oncotarget16721
www.impactjournals.com/oncotarget
proles would not. For patients with discordant IHC assay
and Sanger results, NGS sequencing should be performed
in order to detect EZH2 mutation VAF or associated
mutations which might justify accepting or denying
EZH2 inhibitor treatment (Figure 5). Thus, a fast and
readily accessible combination strategy including Sanger
sequencing and an IHC assay would serve an initial
ltering purpose, successfully singling out patients most
likely to benet from EZH2 inhibitor treatment, while
restricting the number of patients screened by NGS for
EZH2 inhibitor treatment approval.
Further comforting our hypothesis that immunohisto
chemistry is a valuable tool in the determina tion of
patients apt for EZH2 inhibitor treatment, Mccabe et al’s
study showed that among EZH2 mutant cell lines,
H3K27me3 Western Blot levels were signicantly higher
in transcriptionally responsive cell lines, indicating
that the association of EZH2 mutation status and
hypertrimethylation might be a more sensitive marker
for EZH2 inhibitor treatment than EZH2 mutation status
only [8]. Additionally, a study showed that cell lines
presenting low H3K27me2 levels in association with high
H3K27me3 levels in Western Blot were more respon-
sive to the anti-proliferative effects of EZH2 inhibitors,
highlighting the importance of a mutant-like methylation
prole in prospective patients [11]. IHC assays do present
drawbacks in terms of inter-laboratory reproducibility,
although differences could be reduced by using pixel
analysis software to score staining for instance [29].
We found a minority of patients with unexpected
WT-like or mutant-like IHC methylation proles,
given their mutation status, potentially predicting a
respectively impaired or improved response to EZH2
inhibitor treatment. One explanation comes in the form
of EZH2 mutation clonality analysis, and associated
mutations might point to explanations for the remaining
cases. Overall, our NGS study revealed similar mutation
frequencies in genes previously analyzed in large DLBCL
genomic studies [22, 30, 31] Interestingly, GCB-DLBCL
with EZH2 mutations in our cohort showed genomic
proles similar to those previously described for FL, with
frequent associated mutations in CREBBP, KMT2D and
TNFRSF14, potentially indicating a common genetic
history between GCB-EZH2 mutant-DLBCL and FL [23].
Similar data was obtained in a large DLBCL genomic
study, where 5 of 7 patients with EZH2 mutations
presented associated mutations of TNFRSF14 and 4
presented associated mutations of KMT2D [30].
NGS analysis has highlighted cases of interesting
associated mutations in either patients with WT or Y641
mutant EZH2. Although these patients represent anecdotal
evidence only at this time, they lay the groundwork for the
premise that associated mutations should also be taken into
account when deciding which patients to treat with EZH2
inhibitors. For instance, we detected unique mutations in
PIM1 and PRDM1 in patients 1768 and 1631 with WT
EZH2 but mutant-like IHC methylation proles. These
genes are part of the gene network heavily affected by
Figure 5: An IHC/Sanger combination approach as a decision aid for EZH2 inhibitor treatment. By using an initial
combination approach at time of diagnosis, three patient groups emerge, potentially simplifying EZH2 inhibitor treatment guidelines.
Further analysis by NGS would thus be restricted to patients with discordant Sanger sequencing and IHC methylation prole results.
Oncotarget16722
www.impactjournals.com/oncotarget
EZH2 binding and are involved in GC reaction [32, 33].
Interestingly, PIM1 was mutated in only one patient in
our cohort, whereas previous genomic studies showed
signicantly higher mutation frequencies [30, 31]. While a
previous study showed ABC-DLBCL cells to be refractory
to EZH2 inhibitor treatment, patient-specic associated
mutations such as these might modify their response and
should be evaluated [34].
Furthermore, associated mutations are essential
information when deciding on individual targeted
therapeutic cocktails. Patients with several targetable
mutations, such as patient 304 with mutations in both
EZH2 and MYC, might greatly benet from an inhibitor
combination approach [35, 36]. Indeed, in a recent
mouse model, it was shown that only the association of
an EZH2 Y641 mutation and MYC overexpression, and
not the EZH2 Y641 mutation alone, led to lymphoma
development [37].
Four of the ve WT EZH2 patients with mutant-
like IHC proles were of the ABC subtype. While this
may not be relevant for clinical trials which administer
EZH2 inhibitor treatment to GCB subtype patients
exclusively, it is indeed pertinent for clinical trials
where the main inclusion criterion is the presence of
EZH2 gain-of-function mutations. Although rare, EZH2
mutations in ABC subtype patients do exist, either linked
to misclassication or a change in subtype during disease
progression [19]. In any case, this result adds to the
still-open question of the extent to which EZH2 mutant
ABC subtype patients will benet from EZH2 inhibitor
treatment.
Our me3/me2 score was not correlated with prognosis,
although this was not unexpected, given previous studies
showing no correlation between EZH2 mutation status and
prognosis in FL [23, 26]. On the other hand, we showed
that low IHC EZH2 expression is correlated with superior
OS and PFS among ABC-DLBCL patients, identifying
a prognostic impact of our assay, although not present in
multivariate analysis, potentially due to low sample size. A
previous study in breast cancer also showed that low EZH2
expression is correlated with better Distant Disease Free
Survival (DDFS) [12], corroborating our ndings. On the
contrary, Lee et al recently analyzed EZH2 IHC expression
in a cohort of DLBCL patients of similar size and showed
that high EZH2 expression was associated with superior OS,
with EZH2-high ABC patients being the subgroup with the
highest OS, although this nding was not quite statistically
signicant in multivariate analysis [25]. Compared to
Lee et al, our cohort was marginally older, with a larger
percentage of patients over 60 years old or with Ann Arbor
stage III–IV at diagnosis. The molecular characteristics
of DLBCL have indeed been shown to be age-dependent
[38, 39]; however, although this might be a contributing
factor, the reasons for our discrepant ndings are still unclear.
EZH2 inhibitors are currently being tested in
clinical trials in DLBCL as novel and promising weapons
in clinicians’ therapeutic arsenal. This study has shown
that IHC and genomic proles can identify patients who
are most likely to benet from treatment with EZH2
inhibitors by highlighting a specic in vivo H3K27me3-
high/H3K27me2-low prole, determining EZH2 mutation
clonality and pinpointing associated activating mutations.
Immunohistochemistry could thus serve as a convenient,
fast, and easily accessible method to pre-screen patients
exhibiting high me3/me2 scores for sequencing for
associated mutations, thus reducing time and expenses
before determining optimal, patient-specic treatment. As
such, analyzing these parameters could maximize EZH2
inhibitor benet and potentially serve to grant access to
patients who would otherwise not have been considered.
ACKNOWLEDGMENTS
This study was funded by grants from the Ligue
Contre le Cancer (Comité de la Seine Maritime, France)
and from the Institut National du Cancer.
REFERENCES
1.
Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA,
Stein H, Thiele J, Vardiman JW. WHO classication of
tumours of haematopoietic and lymphoid tissues. Lyon:
IARC; 2008.
2.
Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS,
Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X,
Powell JI, Yang L, Marti GE, et al. Distinct types of diffuse
large B-cell lymphoma identied by gene expression prol-
ing. Nature. 2000; 403:503–11.
3.
Wright G, Tan B, Rosenwald A, Hurt EH, Wiestner A,
Staudt LM. A gene expression-based method to diagnose clini-
cally distinct subgroups of diffuse large B cell lymphoma. Proc
Natl Acad Sci U S A. 2003; 100:9991–6. [Internet] Available
from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?art
id=187912&tool=pmcentrez&rendertype=abstract
4.
Margueron R, Reinberg D. The Polycomb complex PRC2
and its mark in life. Nature. 2011; 469:343–9. [cited 2014
Jul 9] [Internet] Available from: http://www.pubmedcentral.
nih.gov/articlerender.fcgi?artid=3760771&tool=pmcentrez
&rendertype=abstract
5.
Viré E, Brenner C, Deplus R, Blanchon L, Fraga M,
Didelot C, Morey L, Van Eynde A, Bernard D,
Vanderwinden J-M, Bollen M, Esteller M, Di Croce L,
et al. The Polycomb group protein EZH2 directly controls
DNA methylation. Nature. 2006; 439:871–4. [cited 2014
Jul 10] [Internet] Available from: http://www.ncbi.nlm.nih.
gov/pubmed/16357870
6.
Sneeringer CJ, Scott MP, Kuntz KW, Knutson SK,
Pollock RM, Richon VM, Copeland RA. Coordinated activ-
ities of wild-type plus mutant EZH2 drive tumor-associated
hypertrimethylation of lysine 27 on histone H3 (H3K27) in
Oncotarget16723
www.impactjournals.com/oncotarget
human B-cell lymphomas. Proc Natl Acad Sci U S A. 2010;
107:20980–5. [cited 2014 Jul 11] [Internet] Available from:
http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=
3000297&tool=pmcentrez&rendertype=abstract
7.
Yap DB, Chu J, Berg T, Schapira M, Cheng S-WG,
Moradian A, Morin RD, Mungall AJ, Meissner B, Boyle M,
Marquez VE, Marra M a, Gascoyne RD, et al. Somatic
mutations at EZH2 Y641 act dominantly through a mech-
anism of selectively altered PRC2 catalytic activity, to
increase H3K27 trimethylation. Blood. 2011; 117:2451–9.
[Internet] Available from: http://www.pubmedcentral.nih.
gov/articlerender.fcgi?artid=3062411&tool=pmcentrez&re
ndertype=abstract
8.
McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C,
Van Aller GS, Liu Y, Graves AP, Della Pietra A, Diaz E,
LaFrance L V, Mellinger M, Duquenne C, et al. EZH2 inhi-
bition as a therapeutic strategy for lymphoma with EZH2-
activating mutations. Nature. Nature Publishing Group.
2012; 492:108–12. [Internet] [cited 2013 Jan 29] Available
from: http://www.ncbi.nlm.nih.gov/pubmed/23051747
9.
Knutson SK, Kawano S, Minoshima Y, Warholic NM,
Huang K-C, Xiao Y, Kadowaki T, Uesugi M, Kuznetsov G,
Kumar N, Wigle TJ, Klaus CR, Allain CJ, et al. Selective
inhibition of EZH2 by EPZ-6438 leads to potent antitumor
activity in EZH2-mutant non-hodgkin lymphoma. Mol
Cancer Ther. 2014; 13:842–54. [Internet] Available from:
http://www.ncbi.nlm.nih.gov/pubmed/24563539
10.
Qi W, Chan H, Teng L, Li L, Chuai S, Zhang R, Zeng J,
Li M, Fan H. Selective inhibition of Ezh2 by a small mol-
ecule inhibitor blocks tumor cells proliferation. Proc Natl
Acad Sci. 2012; 109:21360–5.
11.
Copeland RA, Richon VM, Scott MD, Sneeringer CJ,
Knutson SK, Pollock RM. US 8691507. 2014. [Internet]
Available from: http://www.google.com/patents/US8691507?
12.
Holm K, Grabau D, Lövgren K, Aradottir S, Gruvberger-
Saal S, Howlin J, Saal LH, Ethier SP, Bendahl P-O, Stål O,
Malmström P, Fernö M, Rydén L, et al. Global H3K27
trimethylation and EZH2 abundance in breast tumor sub-
types. Mol Oncol. 2012; [cited 2014 Jul 28]; 6:494–506.
[Internet] Available from: http://www.ncbi.nlm.nih.gov/
pubmed/22766277
13.
Bohers E, Mareschal S, Bouzelfen A, Marchand V,
Ruminy P, Maingonnat C, Ménard A-L, Etancelin P,
Bertrand P, Dubois S, Alcantara M, Bastard C,
Tilly H, et al. Targetable activating mutations are very fre-
quent in GCB and ABC diffuse large B-cell lymphoma.
Genes Chromosome Cancer. 2014; 014 [cited 2013 Dec
19]; 53:144–53. [Internet] Available from: http://www.ncbi.
nlm.nih.gov/pubmed/24327543
14.
Wang K, Li M, Hakonarson H. ANNOVAR: functional
annotation of genetic variants from high-throughput
sequencing data. Nucleic Acids Res. 2010; [cited 2014
Jul 15]; 38:e164. [Internet] Available from: http://www.
pubmedcentral.nih.gov/articlerender.fcgi?artid=2938201&
tool=pmcentrez&rendertype=abstract
15.
Kumar P, Henikoff S, Ng PC. Predicting the effects of cod-
ing non-synonymous variants on protein function using
the SIFT algorithm. Nat Protoc. 2009; [cited 2014 Jul 14];
4:1073–81. [Internet] Available from: http://www.ncbi.nlm.
nih.gov/pubmed/19561590
16.
Sehested J. A simple method for R banding of human chro-
mosomes, showing a pH-dependent connection between
R and G bands. Humangenetik. 1974; 21:55–8.
17.
Foundation for Statistical Computing: R: A language and
environment for statistical computing. Vienna, Austria.
[Internet] Available from: http://www.r-project.org/
18.
Morin RD, Johnson NA, Severson TM, Mungall AJ, An J,
Goya R, Paul JE, Boyle M, Woolcock BW, Kuchenbauer F,
Yap D, Humphries RK, Grifth OL, et al. Somatic muta-
tions altering EZH2 (Tyr641) in follicular and diffuse large
B-cell lymphomas of germinal-center origin. Nat Genet.
Nature Publishing Group. 2010; 42:181–5. [Internet] [cited
2013 Jan 29] Available from: http://www.pubmedcentral.
nih.gov/articlerender.fcgi?artid=2850970&tool=pmcentrez
&rendertype=abstract
19.
Pasqualucci L, Trifonov V, Fabbri G, Ma J, Rossi D,
Chiarenza A, Wells V a, Grunn A, Messina M, Elliot O,
Chan J, Bhagat G, Chadburn A, et al. Analysis of the cod-
ing genome of diffuse large B-cell lymphoma. Nat Genet.
Nature Publishing Group. 2011; 43:830–7. [cited 2013
Feb 5] [Internet] Available from: http://www.pubmedcen-
tral.nih.gov/articlerender.fcgi?artid=3297422&tool=pmcen
trez&rendertype=abstract
20.
Ryan RJH, Nitta M, Borger D, Zukerberg LR, Ferry JA,
Harris NL, Iafrate AJ, Bernstein BE, Sohani AR, Le LP.
EZH2 codon 641 mutations are common in BCL2-
rearranged germinal center B cell lymphomas. PLoS One.
2011; [cited 2014 May 6]; 6:e28585. [Internet] Available
from: http://www.pubmedcentral.nih.gov/articlerender.fcgi
?artid=3237460&tool=pmcentrez&rendertype=abstract
21.
Guo S, Chan JKC, Iqbal J, McKeithan T, Fu K, Meng B,
Pan Y, Cheuk W, Luo D, Wang R, Zhang W, Greiner TC,
Chan WC. EZH2 mutations in follicular lymphoma from
different ethnic groups and associated gene expression
alterations. Clin Cancer Res. 2014; [cited 2014 Jul 28];
20:3078–86. [Internet] Available from: http://www.ncbi.
nlm.nih.gov/pubmed/24634383
22.
Bohers E, Mareschal S, Bertrand P, Viailly PJ, Dubois S,
Maingonnat C, Ruminy P, Tilly H, Jardin F. Activating
somatic mutations in diffuse large B-cell lymphomas: les-
sons from next generation sequencing and key elements in
the precision medicine era. Leuk Lymphoma. 2014; [cited
2014 Dec 4]; (May):1–10. [Internet] Available from: http://
www.ncbi.nlm.nih.gov/pubmed/25130477
23.
Bödör C, Grossmann V. EZH2 mutations are frequent and
represent an early event in follicular lymphoma. Blood.
2013; [cited 2014 Jul 31]; [cited 2014 Jul 31]; 122:3165–8.
[Internet] Available from: http://bloodjournal.hematologyli-
brary.org/lookup/pmid?view=long&pmid=24052547
Oncotarget16724
www.impactjournals.com/oncotarget
24.
Jiang Y, Redmond D, Nie K, Eng KW, Clozel T, Martin
P, Tan LH, Melnick AM, Tam W, Elemento O. Deep
sequencing reveals clonal evolution patterns and muta-
tion events associated with relapse in B-cell lympho-
mas. Genome Biol. 2014; [cited 2014 Nov 18]; 15:432.
[Internet] Available from: http://www.pubmedcentral.nih.
gov/articlerender.fcgi?artid=4158101&tool=pmcentrez&r
endertype=abstract
25.
Lee HJ, Shin DH, Kim K Bin, Shin N, Park WY, Lee
JH, Choi KU, Kim JY, Lee CH, Sol MY. Polycomb pro-
tein EZH2 expression in diffuse large B-cell lymphoma
is associated with better prognosis in patients treated with
rituximab, cyclophosphamide, doxorubicin, vincristine and
prednisone. Leuk Lymphoma. 2014; [cited 2014 May 5];
55:2056–63. [Internet] Available from: http://www.ncbi.
nlm.nih.gov/pubmed/24304372
26.
Bodor C, O’Riain C, Wrench D, Matthews J, Iyengar S,
Tayyib H, Calaminici M, Clear A, Iqbal S, Quentmeier H,
Drexler H, Montoto S, Lister A, et al. EZH2 Y641 muta-
tions in follicular lymphoma. Leukemia. 2011; 25:726–9.
27.
Shen X, Liu Y, Hsu Y-J, Fujiwara Y, Kim J, Mao X,
Yuan G-C, Orkin SH. EZH1 mediates methylation on his-
tone H3 lysine 27 and complements EZH2 in maintaining
stem cell identity and executing pluripotency. Mol Cell.
2008; [cited 2014 Jul 15]; 32:491–502. [Internet] Available
from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?
artid=2630502&tool=pmcentrez&rendertype=abstract
28.
Sahasrabuddhe A, Chen X, Chung F, Velusamy T,
Lim MS, Elenitoba-Johnson KSJ. Oncogenic Y641 muta-
tions in EZH2 prevent Jak2/β-TrCP-mediated degradation.
Oncogene. Nature Publishing Group. 2014; [cited 2014 Oct
1];(August 2013) 1–10. [Internet] Available from: http://
www.ncbi.nlm.nih.gov/pubmed/24469040
29.
Rizzardi AE, Johnson AT, Vogel RI, Pambuccian SE,
Henriksen J, Skubitz AP, Metzger GJ, Schmechel SC.
Quantitative comparison of immunohistochemical stain-
ing measured by digital image analysis versus patholo-
gist visual scoring. Diagn Pathol. 2012; 7:42. [Internet]
Available from: http://www.pubmedcentral.nih.gov/arti-
clerender.fcgi?artid=3379953&tool=pmcentrez&renderty
pe=abstract
30. Lohr JG, Stojanov P, Lawrence MS, Auclair D, Chapuy B,
Sougnez C, Cruz-Gordillo P, Knoechel B, Asmann YW,
Slager SL, Novak AJ, Dogan A, Ansell SM, et al. Discovery
and prioritization of somatic mutations in diffuse large
B-cell lymphoma (DLBCL) by whole-exome sequenc-
ing. Proc Natl Acad Sci U S A. 2012; [cited 2014 Oct
4]; 109:3879–84. [Internet] Available from: http://www.
pubmedcentral.nih.gov/articlerender.fcgi?artid=3309757&
tool=pmcentrez&rendertype=abstract
31.
Morin RD, Mungall K, Pleasance E, Mungall AJ, Goya R,
Huff RD, Scott DW, Ding J, Roth A, Chiu R, Corbett RD,
Chan FC, Mendez-lago M, et al. Mutational and structural
analysis of diffuse large B-cell lymphoma using whole-
genome sequencing. Blood. 2013; 122:1256–66.
32. Turner CA, Mack DH, Davis MM. Pillars article: Blimp-1, a
novel zinc nger-containing protein that can drive the matura-
tion of B lymphocytes into immunoglobulin-secreting cells.
1994. J Immunol. 2010; 185:5–14. [Internet] Available from:
http://www.ncbi.nlm.nih.gov/pubmed/20562267
33.
Velichutina I, Shaknovich R, Geng H, Johnson NA,
Gascoyne RD, Melnick AM, Elemento O. EZH2-mediated
epigenetic silencing in germinal center B cells contrib-
utes to proliferation and lymphomagenesis. Blood. 2010;
[cited 2014 Oct 28]; 116:5247–55. [Internet] Available
from: http://bloodjournal.hematologylibrary.org/ content/
116/24/5247.short
34.
Béguelin W, Popovic R, Teater M, Jiang Y. EZH2 is
required for germinal center formation and somatic EZH2
mutations promote lymphoid transformation. Cancer Cell.
2013; 23:677–92. [Internet] Available from: http://www.
sciencedirect.com/science/article/pii/S1535610813001797
35.
Zhao X, Lwin T, Zhang X, Huang a, Wang J, Marquez VE,
Chen-Kiang S, Dalton WS, Sotomayor E, Tao J. Disruption
of the MYC-miRNA-EZH2 loop to suppress aggressive
B-cell lymphoma survival and clonogenicity. Leukemia.
2013; [cited 2014 Sep 4]; 27:2341–50. [Internet] Available
from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?
artid=4015113&tool=pmcentrez&rendertype=abstract
36.
Sander S, Bullinger L, Klapproth K, Fiedler K, Kestler HA,
Barth TFE, Möller P, Stilgenbauer S, Pollack JR, Wirth T.
MYC stimulates EZH2 expression by repression of its nega-
tive regulator miR-26a. Blood. 2008; [cited 2014 Sep 4];
112:4202–12. [Internet] Available from: http://blood-
journal.hematologylibrary.org/content/112/10/4202.short
37.
Berg T, Thoene S, Yap D, Wee T, Schoeler N, Roste P,
Lim E, Bilenky M, Mungall AJ, Oellerich T, Lee S, Lai CK,
Umlandt P, et al. A transgenic mouse model demonstrating the
oncogenic role of mutations in the polycomb-group gene EZH2
in lymphomagenesis. Blood. 2014; 123:3914–25. [Internet]
Available from: http://bloodjournal. hematologylibrary.org/
content/early/2014/05/06/blood-2012-12-473439.short
38.
Mareschal S, Lanic H, Ruminy P, Bastard C, Tilly H,
Jardin F. The proportion of activated B-cell like subtype
among de novo diffuse large B-cell lymphoma increases
with age. Haematologica. 2011; [cited 2014 Oct 15];
96:1888–90. [Internet] Available from: http://www.haema-
tologica.org/content/96/12/1888
39.
Klapper W, Kreuz M, Kohler C, Burkhardt B,
Szczepanowski M, Salaverria I, Hummel M, Loefer M,
Pillizzery S, Woessmann W, Schwänen C, Trümper L,
Wessendorf S, et al. Patient age at diagnosis is associated
with the molecular characteristics of diffuse large B-cell
lymphoma. Blood. 2012; [cited 2014 Sep 1]; 119:1882–7.
[Internet] Available from: http://bloodjournal.hematology-
library.org/content/119/8/1882.short
... One of the most important oncogenic mutations that occurs in the SET domain is found at residue Y641 [8,17]. A subset of hematologic malignancies, including diffuse large B-cell lymphoma (DLBCL), is characterized by expression of heterozygous EZH2(Y641) mutations and increased levels of histone methylation [5,8,10,17,18]. ...
... Chemoimmunotherapy, the combination of the monoclonal antibody, Rituximab, with standard chemotherapy, is the standard-of-care treatment for DLBCL; however, a high percentage of patients relapse or are refractory to this treatment [21,22]. Multiple studies have shown cells with EZH2 mutations to be dependent on the higher catalytic activity of mutant EZH2 Y641 for proliferation [8,18,[23][24][25]. In line with these findings, heterozygous gain-of-function mutations of the amino acid Y641 have been observed in 22~24% of GCB-DLBCL and 10% of follicular B-cell lymphoma (FL) cases [8,17,18], with a much higher percentage than is observed in other hematologic malignancies, such as Myelodysplastic syndromes (MDS) and AML [9,10,[26][27][28]. ...
... Multiple studies have shown cells with EZH2 mutations to be dependent on the higher catalytic activity of mutant EZH2 Y641 for proliferation [8,18,[23][24][25]. In line with these findings, heterozygous gain-of-function mutations of the amino acid Y641 have been observed in 22~24% of GCB-DLBCL and 10% of follicular B-cell lymphoma (FL) cases [8,17,18], with a much higher percentage than is observed in other hematologic malignancies, such as Myelodysplastic syndromes (MDS) and AML [9,10,[26][27][28]. Thus, lymphoma is a more clinically relevant disease target for inhibition of this mutation [25]. ...
Article
Full-text available
Activating mutations in EZH2, the catalytic component of PRC2, promote cell proliferation, tumorigenesis, and metastasis through enzymatic or non-enzymatic activity. The EZH2-Y641 gain-of-function mutation is one of the most significant in diffuse large B-cell lymphoma (DLBCL). Although EZH2 kinase inhibitors, such as EPZ-6438, provide clinical benefit, certain cancer cells are resistant to the enzymatic inhibition of EZH2 because of the inability to functionally target mutant EZH2, or because of cells’ dependence on the non-histone methyltransferase activity of EZH2. Consequently, destroying mutant EZH2 protein may be more effective in targeting EZH2 mutant cancers that are dependent on the non-catalytic activity of EZH2. Here, using extensive selectivity profiling, combined with genetic and animal model studies, we identified USP47 as a novel regulator of mutant EZH2. Inhibition of USP47 would be anticipated to block the function of mutated EZH2 through induction of EZH2 degradation by promoting its ubiquitination. Moreover, targeting of USP47 leads to death of mutant EZH2-positive cells in vitro and in vivo. Taken together, we propose targeting USP47 with a small molecule inhibitor as a novel potential therapy for DLBCL and other hematologic malignancies characterized by mutant EZH2 expression.
... Despite the well-established mutational status and driver function of this gene in DLBCL, the prognostic value of its mutational status remains controversial, with the few published data mostly pointing towards a lack thereof [7,11]. In spite of this, analysis of publicly available data from patients included in two different studies [5]-warranting cautious interpretation, given the potential use of different inclusion and outcome criteria-associates EZH2 mutated DLBCL with worse Overall and Progression Free Survival, albeit not significantly ( Figure A1) Furthermore, when considering EZH2 expression, some studies suggest that it may be a promising biomarker in DLBCL [12][13][14][15][16][17][18][19]. Nonetheless, published data are inconsistent, possibly due to the use of different methodologies (universally semiquantitative), different positivity threshold, inclusion of heterogeneously treated patients and small number of patients per study (median number of patients per study: 68, interquartile range: 37 to 92). ...
... Several reports [12,13,16,20] have described an association between EZH2 expression and patient outcome, mostly between high expression levels and worse prognosis [12,16,20]. As previously stated, we failed to find a statistically significant association between isolated EZH2 expression and patient outcome. ...
... Several reports [12,13,16,20] have described an association between EZH2 expression and patient outcome, mostly between high expression levels and worse prognosis [12,16,20]. As previously stated, we failed to find a statistically significant association between isolated EZH2 expression and patient outcome. ...
Article
Full-text available
Background: DLBCL represent a heterogeneous group of aggressive diseases. High grade B-cell lymphomas (HGBCL) were recently individualized from DLBCL as a discrete diagnostic entity due to their worse prognosis. Currently, although most patients are successfully treated with RCHOP regimens, 1/3 will either not respond or ultimately relapse. Alterations in histone modifying enzymes have emerged as the most common alterations in DLBCL, but their role as prognostic biomarkers is controversial. We aimed to ascertain the prognostic value of EZH2 immunoexpression in RCHOP-treated DLBCL and HGBCL. Results: We performed a retrospective cohort study including 125 patients with RCHOP-treated DLBCL or HGBCL. EZH2 expression levels did not differ between diagnostic groups or between DLBCL-NOS molecular groups. We found no associations between EZH2 expression levels and outcome, including in the subgroup analysis (GC versus non-GC). Nonetheless, EZH2/BCL2 co-expression was significantly associated with worse outcome (event free survival and overall survival). Conclusion: Although EZH2 mutations are almost exclusively found in GC-DLBCL, we found similar EZH2 expression levels in both DLBCL-NOS molecular groups, suggesting non-mutational mechanisms of EZH2 deregulation. These findings suggest that the use of EZH2 antagonists might be extended to non-GC DLBCL patients with clinical benefit. EZH2/BCL2 co-expression was associated with a worse outcome.
... Изучение филогенетической эволюции опухолевых клеток, а также множества опухолевых клонов при ФЛ показывает, что прогрессирование болезни происходит либо путем прямой клональной (линейной) эволюции -последовательного накопления соматических мутаций в опухолевой клетке, либо путем дивергентной эволюции от общей клетки-предшественницы -единовременного образования нескольких независимых опухолевых клонов из общей клетки-предшественницы [11,12]. Нарушения в опухолевых клетках ФЛ, включая реаранжировки гена BCL-2/гиперэкспрессию белка BCL-2 и мутации в гене EZH2/гиперэкспрессию белка EZH2, свидетельствуют о линейном пути эволюции опухолевой клетки [13,14], в то время как реаранжировки гена MYC/гиперэкспрессия белка MYC, мутации в генах TP53 и IRF8 свидетельствуют о дивергентном пути эволюции опухолевой клетки [15,16]. ...
... В связи с тем, что клинических и морфологических характеристик [21][22][23][24][25][26][27][28] на этапе диагностики ФЛ, как правило, недостаточно для надежного прогнозирования течения заболевания, определение молекулярно-генетических факторов приобретает особую актуальность. Данные литературы по изучению прогностической значимости мутаций в гене EZH2 при ФЛ представлены в незначительном объеме, а опубликованные сообщения крайне противоречивы [11][12][13][14][15][16]. Поскольку в настоящее время разработаны селективные ингибиторы EZH2 [29], являющиеся высокоэффективной терапевтической опцией для лечения ФЛ при наличии мутаций в гене EZH2, мы инициировали исследование по изучению мутационного статуса гена EZH2 в когорте больных с ФЛ 1-3А цитологического типа. ...
Article
Full-text available
Aim. To determine the incidence and prognostic value of mutations in exon 16 of EZH2 as well as those of polymorphism с.1582-21А>G (rs2072407) in EZH2 in patients with follicular lymphoma (FL) grades 1–3А in relation to morphologic and cytogenetic tumor characteristics. Materials & Methods. The prospective cohort study conducted by the National Research Center for Hematology from January 2017 to April 2021 enrolled 80 patients with newly diagnosed FL grades 1/2 and 3А. The median follow-up was 53 months. Molecular and cytogenetic analyses were based on biopsy samples of lymph nodes obtained before chemotherapy. The mutation status of exon 16 in EZH2 and the presence of intronic polymorphism rs2072407 in EZH2 were examined by Sanger sequencing method. Translocation t(14;18)(q32;q21) was detected by karyotyping or FISH. Results. Mutations in exon 16 of EZH2 (mutEZH2) were identified in 10/80 (13 %) patients. All patients showed missense mutation in codon 646 of EZH2. Translocation t(14;18) was detected in 45/80 (56 %) cases. Poor outcome in the cohort with no t(14;18) was observed 3 times more often than in the group of patients with t(14;18) (p = 0.0001). The presence of t(14;18) was associated with favorable prognosis irrespective of either the mutation status of exon 16 in EZH2 or the FL grade. The analysis of the polymorphism rs2072407 status yielded the following genotypes: AA in 24 % (n = 19), AG in 42 % (n = 34), and GG in 34 % (n = 27) of cases. The variants АА and AG were associated with higher risk of death (hazard ratio 2.9; 95% confidence interval 1.2–10.6; p = 0.01), whereas the genotype GG was associated with wtEZH2 (10 % vs. 37 %) and favorable prognosis (p = 0.065). Conclusion. Significant biological markers for favorable prognosis in FL appeared to be the presence of t(14;18)(q32;q21) and GG genotype of polymorphism rs2072407 in EZH2. The previously identified prognostic factors (grade 3А, bulky tumor lesions > 6 cm, Ki-67 > 35 %, and a short interval between symptom onset and chemotherapy start) were incorporated into a new unified personalized predictive (index PPI) FL model by supplementing it with two additional biological markers: the presence of t(14;18)(q32;q21) and GG genotype of polymorphism rs2072407. This approach may increase the prognostic value of the new personalized design which will provide the basis for risk-adapted algorithms for FL treatment.
... In our study, the Se of this approach was 95%, with 100% Sp, and, as has been described [38], no false-positive cases have been documented with this method. On the other hand, Dubois et al. [39] initially reported 22%, and thereafter up to 24%, frequency of EZH2 Y641 mutations in GC-DLBCL, which is slightly higher than initially reported by Morin et al. (n = 18/83, 21.7%) [40]. In our study, we found a similar frequency of EZH2 Y641 mutations (n = 20/98, 20.4%) in the same population. ...
Article
Full-text available
(1) Background: The epigenetic regulator EZH2 is a subunit of the polycomb repressive complex 2 (PRC2), and methylates H3K27, resulting in transcriptional silencing. It has a critical role in lymphocyte differentiation within the lymph node. Therefore, mutations at this level are implicated in lymphomagenesis. In fact, the mutation at the Y641 amino acid in the EZH2 gene is mutated in up to 40% of B-cell lymphomas. (2) Methods: We compared the presence of exon 16 EZH2 mutations in tumor samples and ctDNA in a prospective trial. These mutations were determined by Sanger sequencing and ddPCR. (3) Results: One hundred and thirty-eight cases were included. Ninety-eight were germinal center, and twenty had EZH2 mutations. Mean follow-up (IQR 25–75) was 23 (7–42) months. The tumor samples were considered the standard of reference. Considering the results of the mutation in ctDNA by Sanger sequencing, the sensibility (Se) and specificity (Sp) were 52% and 99%, respectively. After adding the droplet digital PCR (ddPCR) analysis, the Se and Sp increased to 95% and 100%, respectively. After bivariate analysis, only the presence of double-hit lymphoma (p = 0.04) or EZH2 mutations were associated with relapse. The median Progression free survival (PFS) (95% interval confidence) was 27.7 (95% IC: 14–40) vs. 44.1 (95% IC: 40–47.6) months for the mutated vs. wild-type (wt) patients. (4) Conclusions: The ctDNA is useful for analyzing EZH2 mutations, which have an impact on PFS.
... The human reference genome (GRCh38.p13/) alignment of obtained sequences was performed using Burrows-Wheeler Aligner (BWA) program [24,25]. Identification of somatic mutations was done using LoFreq (version 2) variant caller [26,27]. ...
Article
Full-text available
Introduction: Next-generation sequencing (NGS) elucidates the diffuse large B-cell lymphoma (DLBCL) genetic characteristics by finding recurrent and novel somatic mutations. This observational study attempted to create an NGS panel with a focus on identifying novel somatic mutations which could have potential clinical and therapeutic implications. This panel was created to look for mutations in 133 genes chosen on basis of a literature review and it was used to sequence the tumor DNA of 20 DLBCL patients after a centralized histopathologic review. Methods: The study included 20 patients having DLBCL. The quality and quantity of tumor cells were accessed by H&E staining and correlated with histopathology and Immunohistochemistry (IHC) status. Patients were grouped as ABC (activated B-cell), PMBL (primary mediastinal large B-cell lymphoma), and other or unclassified subtypes. The lymphoma panel of 133 was designed on targeted sequencing of multiple genes for the coding regions through NGS. The libraries were prepared and sequenced using the Illumina platform. The alignment of obtained sequences was performed using Burrows-Wheeler Aligner and identification of somatic mutations was done using LoFreq (version 2) variant caller. The mutations were annotated using an annotation pipeline (VariMAT). Previously published literature and databases were used for the annotation of clinically relevant mutations. The common variants were filtered for reporting based on the presence in various population databases (1000G, ExAC, EVS, 1000Japanese, dbSNP, UK10K, MedVarDb). A custom read-depth-based algorithm was used to determine CNV (Copy Number Variants) from targeted sequencing experiments. Rare CNVs were detected using a comparison of the test data read-depths with the matched reference dataset. Reportable mutations were prioritized and prepared based on AMPASCO-CAP (Association for Molecular Pathology-American Society of Clinical Oncology-College of American Pathologists), WHO guidelines, and also based on annotation metrics from OncoMD (a knowledge base of genomic alterations). Results: The informativity of the panel was 95 percent. NOTCH 1 was the most frequently mutated gene in 16.1% of patients followed by 12.9% who had ARID1A mutations. MYD88 and TP53 mutations were detected in 9.6% of the patient while 6.4% of patients had CSF3R mutations. NOTCH 1 and TP 53 are the most frequently reported gene in the middle age group (40-60). Mutation in MYD88 is reported in every age group. MYD88 (51%) is the most common mutation in ABC subtypes of DLBCL, followed by NOTCH 1 (44%) and SOCS 1 (33%) according to our findings. NOTCH 1 mutations are frequent in ABC and PMBL subtypes. Closer investigation reveals missense mutation is the most frequent mutation observed in the total cohort targeting 68.4% followed by frameshift deletion reported in 26.3%. Six novel variants have been discovered in this study. Conclusions: This study demonstrates the high yield of information in DLBCL using the NGS Lymphoma panel. Results also highlight the molecular heterogeneity of DLBCL subtypes which indicates the need for further studies to make the results of the NGS more clinically relevant.
... Hence, quantification of EZH2 protein expression can be a surrogate measure of EZH2 function. In this study, we evaluated the functional disruption of EZH2 by immunohistochemistry. Due to loss-of-function phenotype of EZH2 mutations in MDS, the expression levels correlated with mutation status in 73% of MDS patients, unlike diffuse large B cell lymphoma cases, where EZH2 expression was independent of mutation status 43,44 . The possible reasons for the incomplete correlation in the remaining (27%) MDS patients are elaborated below. ...
Article
EZH2 coding mutation (EZH2MUT), resulting in loss-of-function, is an independent predictor of overall survival in MDS. EZH2 function can be altered by other mechanisms including copy number changes, and mutations in other genes and non-coding regions of EZH2. Assessment of EZH2 protein can identify alterations of EZH2 function missed by mutation assessment alone. Precise evaluation of EZH2 function and gene-protein correlation in clinical MDS cohorts is important in the context of upcoming targeted therapies aimed to restore EZH2 function. In this study, we evaluated the clinicopathologic characteristics of newly diagnosed MDS patients with EZH2MUT and correlated the findings with protein expression using immunohistochemistry. There were 40 (~6%) EZH2MUT MDS [33 men, seven women; median age 74 years (range, 55–90)]. EZH2 mutations spanned the entire coding region. Majority had dominant EZH2 clone [median VAF, 30% (1–92)], frequently co-occurring with co-dominant TET2 (38%) and sub-clonal ASXL1 (55%) and RUNX1 (43%) mutations. EZH2MUT MDS showed frequent loss-of-expression compared to EZH2WT (69% vs. 27%, p = 0.001). Interestingly, NINE (23%) EZH2WT MDS also showed loss-of-expression. EZH2MUT and loss-of-expression significantly associated with male predominance and chr(7) loss. Further, only EZH2 loss-of-expression patients showed significantly lower platelet counts, a trend for higher BM blast% and R-IPSS scores. Over a 14-month median follow-up, both EZH2MUT (p = 0.027) and loss-of-expression (p = 0.0063) correlated with poor survival, independent of R-IPSS, age and gender. When analyzed together, loss-of-expression showed a stronger correlation than mutation (p = 0.061 vs. p = 0.43). In conclusion, immunohistochemical assessment of EZH2 protein, alongside mutation, is important for prognostic workup of MDS.
... In our study, the Se of this approach was 95 %, with a 100 % Sp, and as has been described [37], no falsepositive cases have been documented with this method. Dubois et al. [38] documented initially 22 %, and thereafter up to 24 % frequency of EZH2 Y641 mutations in GC-DLBCL, which is slightly higher than initially reported by Morin et al. (n=18/83, 21.7%) [39]. In our study, we found a similar frequency of EZH2 Y641 mutations (n=20/98, 20.4 %) in the same population. ...
Preprint
Full-text available
Background: The epigenetic regulator EZH2 is a subunit of the polycomb repressive complex 2 (PRC2), methylates H3K27, resulting in transcriptional silencing. The mutation at Y646 amino acid in the EZH2 gene is mutated in up to 40 % of B-cell lymphomas. We compared the presence of exon 16 EZH2 mutations in tumor samples and ctDNA in a prospective trial. The mutations were determined by sanger sequencing, and by ddPCR. We also evaluated the impact of these mutations on response, relapse, and survival. Results: One hundred and thirty-eight cases were included. Ninety-eight were germinal center, and twenty had EZH2 mutations. Mean follow-up (IQR 25-75) was 23 (7- 42) months. The tumor samples were considered the standard of reference. Considering the results of the mutation in ctDNA by Sanger sequencing, the sensibility (Se) and specificity (Sp) were 52 % and 99 %, respectively. After adding the droplet digital PCR (ddPCR) analysis, the Se and Sp increased to 95 and 100 %, respectively. After bivariate analysis, only the presence of double-hit lymphoma (p=0.04), or EZH2 mutations were associated with relapse. The median PFS (95 % Interval confidence) was 27.7 (95 % IC: 14-40) vs 44.1 (95 % IC: 40-47.6) months for the mutated vs wt patients. Conclusions: The ctDNA is usefull to analyse EZH2 mutations, which have an impact in PFS.
Article
Diffuse large B cell lymphoma, not otherwise specified (DLBCL, NOS) is the most common type of non-Hodgkin lymphoma (NHL). The 2016 World Health Organization (WHO) classification defined DLBCL, NOS and its subtypes based on clinical findings, morphology, immunophenotype, and genetics. However, even within the WHO subtypes, it is clear that additional clinical and genetic heterogeneity exists. Significant efforts have been focused on utilizing advanced genomic technologies to further subclassify DLBCL, NOS into clinically relevant subtypes. These efforts have led to the implementation of novel algorithms to support optimal risk-oriented therapy and improvement in the overall survival of DLBCL patients. We gathered an international group of experts to review the current literature on DLBCL, NOS, with respect to genomic aberrations and the role they may play in the diagnosis, prognosis and therapeutic decisions. We comprehensively surveyed clinical laboratory directors/professionals about their genetic testing practices for DLBCL, NOS. The survey results indicated that a variety of diagnostic approaches were being utilized and that there was an overwhelming interest in further standardization of routine genetic testing along with the incorporation of new genetic testing modalities to help guide a precision medicine approach. Additionally, we present a comprehensive literature summary on the most clinically relevant genomic aberrations in DLBCL, NOS. Based upon the survey results and literature review, we propose a standardized, tiered testing approach which will help laboratories optimize genomic testing in order to provide the maximum information to guide patient care.
Article
Full-text available
Approximately 15% of follicular lymphomas (FL) lack overexpression of BCL2 and the underlying translocation t(14;18). These cases can be diagnostically challenging, especially regarding follicular hyperplasia (FH). In a subset of FL, mutations in genes encoding for epigenetic modifiers, such as the histone-lysine N-methyltransferase EZH2 (enhancer of zeste homolog 2), were found, which might be used diagnostically. These molecular alterations can lead to an increased tri-methylation of histone H3 at position lysine 27 (H3K27m3) that, in turn, can be visualized immunohistochemically. The aim of this study was to analyze the expression of H3K27m3 in FL, primary cutaneous follicle center lymphomas (PCFCL), and pediatric-type FL (PTFL) in order to investigate its value in the differential diagnosis to FH and other B cell lymphomas and to correlate it to BCL2 expression and the presence of t(14;18). Additionally, the mutational profile of selected cases was considered to address H3K27m3’s potential use as a surrogate parameter for mutations in genes encoding for epigenetic modifiers. Eighty-nine percent of FL and 100% of PCFCL cases overexpressed H3K27m3, independently of BCL2, EZH2, and the presence of mutations. In contrast, 95% of FH and 100% of PTFL cases lacked H3K27m3 overexpression. Other B cell lymphomas considered for differential diagnosis also showed overexpression of H3K27m3 in the majority of cases. In summary, overexpression of H3K27m3 can serve as a new, BCL2 independent marker in the differential diagnosis of FL and PCFCL, but not PTFL, to FH, while being not of help in the differential diagnosis of FL to other B cell lymphomas.
Preprint
Full-text available
Background: The epigenetic regulator EZH2 is a subunit of the polycomb repressive complex 2 (PRC2), methylates H3K27, resulting in transcriptional silencing. The mutation at Y646 amino acid in the EZH2 gene is mutated in up to 40 % of B-cell lymphomas. Methods: We compared the presence of exon 16 EZH2 mutations in tumor samples and ctDNA in a prospective trial. The mutations were determined by sanger sequencing, and by ddPCR. We also evaluated the impact of these mutations on response, relapse, and survival. Results: One hundred and thirty-eight cases were included. Ninety-eight were germinal center, and twenty had EZH2 mutations. Mean follow-up (IQR 25-75) was 23 (7- 42) months. The tumor samples were considered the standard of reference. Considering the results of the mutation in ctDNA by Sanger sequencing, the sensibility (Se) and specificity (Sp) were 52 % and 99 %, respectively. After adding the ddPCR analysis, the Se and Sp increased to 95 and 100 %, respectively. After bivariate analysis, only the presence of double-hit lymphoma (p=0.04), or EZH2 mutations were associated with relapse. The median PFS (95 % Interval confidence) was 27.7 (95 % IC: 14-40) vs 44.1 (95 % IC: 40-47.6) months for the mutated vs wt patients. Conclusions: The ctDNA is usefull to analyse EZH2 mutations, which have an impact in PFS.
Article
Full-text available
Abstract Diffuse large B-cell lymphoma (DLBCL) is the most common form of lymphoma, accounting for 30-40% of newly diagnosed non-Hodgkin lymphomas. Historically, DLBCL has been thought to involve recurrent translocations of the IGH locus and the deregulation of rearranged oncogenes. Whole exome sequencing (WES) of more than two hundred DLBCL has completely redefined the genetic landscape of the disease by identifying recurrent single nucleotide variants and providing new therapeutic opportunities in DLBCL molecular subtypes. Some of these somatic mutations target genes that play a crucial role in B-cell function (BCR signaling, NFκ-B pathway, Toll-like receptor signaling, and the PI3K pathway), immunity, cell cycle/apoptosis, or chromatin modification. In this review, following an overview of the somatic mutations reported in DLBCL, we focus on activating and clustered mutations targeting genes including MYD88, CD79A/B, EZH2 and CARD11 and discuss their clinical and therapeutic relevance in the precision medicine era.
Article
Full-text available
Background Molecular mechanisms associated with frequent relapse of diffuse large B-cell lymphoma (DLBCL) are poorly defined. It¿s especially unclear how primary tumor clonal heterogeneity contributes to relapse. Here, we explore unique features of B-cell lymphomas - VDJ recombination and somatic hypermutation - to address this question.ResultsWe performed high-throughput sequencing of rearranged VDJ junctions in 14 pairs of matched diagnosis-relapse tumors, among which 7 pairs were further characterized by exome sequencing. We identify two distinctive modes of clonal evolution of DLBCL relapse: an early-divergent mode in which clonally related diagnosis and relapse tumors diverged early and developed in parallel; and a late-divergent mode in which relapse tumors developed directly from diagnosis tumors with minor divergence. By examining mutation patterns in the context of phylogenetic information provided by VDJ junctions, we identified mutations in epigenetic modifiers such as KMT2D as potential early driving events in lymphomagenesis and immune escape alterations as relapse-associated events.Conclusions Altogether, our study for the first time provides important evidence that DLBCL relapse may result from multiple, distinct tumor evolutionary mechanisms, providing rationale for therapies for each mechanism. Moreover, this study highlights the urgent need to understand the driving roles of epigenetic modifier mutations in lymphomagenesis, and immune surveillance factor genetic lesions in relapse.
Article
Full-text available
Gain-of-function mutations of EZH2 occur frequently in diffuse large B-cell lymphomas and in follicular lymphomas (FLs). However, the frequency of EZH2 mutation in Chinese FLs and the potential targets affected by this mutation are unknown. We determined EZH2 codon 641 mutations in Chinese FLs (n=124) and compared with Western FLs (n=70) using sensitive pyrosequencing assay. Gene expression profiling (GEP) was performed to determine differential gene-expression between the mutated vs unmutated subgroups, and selected genes were validated using immunohistochemistry (IHC). Our results showed similar frequencies of EZH2 codon 641 mutations in Chinese and Western FL cohorts (16.9% vs 18.6%,χ2-test, p=0.773), including all five reported mutation variants. We observed significant association of EZH2 mutation with low morphologic grade FLs (Grade 1-2, 23.6% vs Grade3, 7.7%,χ2-test, p=0.02). EZH2 mutations also showed significant association with BCL2 rearrangement in the Chinese cohort (26.8% vs 8.8%χ2-test, p=0.008) and combined cohorts (26.3% vs 9.1%,χ2-test, p=0.002). GEP analysis identified genes including TCF4, FOXP1, TCL1A, BIK and RASSF6P with significantly lower (p<0.01) in mutated cases and the potential target TCL1A showed consistency at the protein level. Similar prevalence of EZH2 mutation in two ethnic groups suggests shared pathogenetic mechanisms. The much lower frequency of EZH2 mutation in cases without BCL2 translocation suggests a different pattern of evolution of this subtype of FL. GEP studies showed a set of differentially expressed genes and suggested that EZH2 mutation may help to lock the tumor cells at the GC stage of differentiation.
Article
Full-text available
Mutations within the catalytic domain of the histone methyltransferase (HMT) EZH2 have been identified in subsets of non-Hodgkin Lymphoma (NHL) patients. These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. While both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties including good oral bioavailability in animals. Here we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2 mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 day after stopping compound treatment in two EZH2 mutant xenograft models. These data confirm the dependency of EZH2 mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers.
Article
Full-text available
EZH2 (enhancer of zeste homolog 2) is a critical enzymatic subunit of the polycomb repressive complex 2 (PRC2), which trimethylates histone H3 (H3K27) to mediate gene repression. Somatic mutations, overexpression and hyperactivation of EZH2 have been implicated in the pathogenesis of several forms of cancer. In particular, recurrent gain-of-function mutations targeting EZH2 Y641 occur most frequently in follicular lymphoma and aggressive diffuse large B-cell lymphoma and are associated with H3K27me3 hyperactivation, which contributes to lymphoma pathogenesis. However, the post-translational mechanisms of EZH2 regulation are not completely understood. Here we show that EZH2 is a novel interactor and substrate of the SCF E3 ubiquitin ligase β-TrCP (FBXW1). β-TrCP ubiquitinates EZH2 and Jak2-mediated phosphorylation on Y641 directs β-TrCP-mediated EZH2 degradation. RNA interference-mediated silencing of β-TrCP or inhibition of Jak2 results in EZH2 stabilization with attendant increase in H3K27 trimethylation activity. Importantly, the EZH2(Y641) mutants recurrently implicated in lymphoma pathogenesis are unable to bind β-TrCP. Further, endogenous EZH2(Y641) mutants in lymphoma cells exhibit increased EZH2 stability and H3K27me3 hyperactivity. Our studies demonstrate that β-TrCP has an important role in controlling H3K27 trimethylation activity and lymphoma pathogenesis by targeting EZH2 for degradation.Oncogene advance online publication, 27 January 2014; doi:10.1038/onc.2013.571.
Article
The histone methyltransferase EZH2 is frequently mutated in germinal center-derived diffuse large B-cell lymphoma and follicular lymphoma. To further characterize these EZH2 mutations in lymphomagenesis, we generated a mouse line where EZH2(Y641F) is expressed from a lymphocyte-specific promoter. Spleen cells isolated from the transgenic mice displayed a global increase in trimethylated H3K27, but the mice did not show an increased tendency to develop lymphoma. As EZH2 mutations often coincide with other mutations in lymphoma, we combined the expression of EZH2(Y641F) by crossing these transgenic mice with Eµ-Myc transgenic mice. We observed a dramatic acceleration of lymphoma development in this combination model of Myc and EZH2(Y641F). The lymphomas show histologic features of high-grade disease with a shift toward a more mature B-cell phenotype, increased cycling and gene expression, and epigenetic changes involving important pathways in B-cell regulation and function. Furthermore, they initiate disease in secondary recipients. In summary, EZH2(Y641F) can collaborate with Myc to accelerate lymphomagenesis demonstrating a cooperative role of EZH2 mutations in oncogenesis. This murine lymphoma model provides a new tool to study global changes in the epigenome caused by this frequent mutation and a promising model system for testing novel treatments.
Article
Diffuse large B cell lymphoma (DLBCL) is an aggressive and heterogeneous malignancy that can be divided in two major subgroups, germinal center B-cell-like (GCB) and activated B-cell-like (ABC). Activating mutations of genes involved in the BCR and NF-κB pathways (CD79A, CD79B, MYD88, and CARD11) or in epigenetic regulation (EZH2) have been recently reported, preferentially in one of the two DLBCL subtypes. We analyzed the mutational status of these five recurrently mutated genes in a cohort of 161 untreated de novo DLBCL. Overall, 93 mutations were detected, in 61 (38%) of the patients. The L265P MYD88 mutation was the most frequent MYD88 variant (n = 18), observed exclusively in the ABC subtype. CD79A/CD79B ITAM domains were targeted in ABC DLBCL (12/77; 16%), whereas CARD11 mutations were equally distributed in the two subtypes. The EZH2 Y641 substitution was found almost exclusively in the GCB subgroup (15/62; 24%). Twenty cases (12%) displayed two activating mutations, including the most frequent CD79/MYD88 variants combination (n = 8) which is observed exclusively in the ABC subtype. When considering only ABC DLBCL patients treated by rituximab plus chemotherapy, the presence of an activating NF-κB mutation was associated with an unfavorable outcome (3-years OS 26% for mutated cases versus 67% for the cases without mutations, P = 0.0337). Our study demonstrates that activating and targetable mutations are observed at a very high frequency in DLBCL at the time of diagnosis, indicating that sequencing of a limited number of genes could help tailor an optimal treatment strategy in DLBCL. © 2013 Wiley Periodicals, Inc.