You are on page 1of 11

Articles

Cancer mediates effector T cell dysfunction by


targeting microRNAs and EZH2 via glycolysis
restriction
Ende Zhao1,2,13, Tomasz Maj1,13, Ilona Kryczek1, Wei Li1,2, Ke Wu1,2, Lili Zhao3, Shuang Wei1,
Joel Crespo1, Shanshan Wan1, Linda Vatan1, Wojciech Szeliga1, Irene Shao1, Yin Wang1, Yan Liu1,
Sooryanarayana Varambally4,5, Arul M Chinnaiyan5, Theodore H Welling1, Victor Marquez6,
© 2015 Nature America, Inc. All rights reserved.

Jan Kotarski7, Hongbo Wang8, Zehua Wang8, Yi Zhang9, Rebecca Liu10, Guobin Wang2 & Weiping Zou1,11,12

Aerobic glycolysis regulates T cell function. However, whether and how primary cancer alters T cell glycolytic metabolism and
affects tumor immunity in cancer patients remains a question. Here we found that ovarian cancers imposed glucose restriction
on T cells and dampened their function via maintaining high expression of microRNAs miR-101 and miR-26a, which constrained
expression of the methyltransferase EZH2. EZH2 activated the Notch pathway by suppressing Notch repressors Numb and
Fbxw7 via trimethylation of histone H3 at Lys27 and, consequently, stimulated T cell polyfunctional cytokine expression and
promoted their survival via Bcl-2 signaling. Moreover, small hairpin RNA–mediated knockdown of human EZH2 in T cells elicited
poor antitumor immunity. EZH2+CD8+ T cells were associated with improved survival in patients. Together, these data unveil a
metabolic target and mechanism of cancer immune evasion.

Effector T cells, particularly polyfunctional effector T cells, produce we found that EZH2 in human T cells controls effector T cell poly-
multiple effector cytokines and cytotoxic proteases after activation, functionality and survival. Notably, EZH2 is a central target and
mediate potent protective immunity and are associated with beneficial sensor of glycolytic metabolism in the tumor microenvironment.
immune responses against viral infection1–5. Inadequate T cell Furthermore, we found that EZH2 expression in T cells is regulated
activation might result in dysfunctional states of T cells, including by glycolytic metabolism via microRNAs (miRNAs) and is function-
anergy, exhaustion and senescence6–8. It has been reported that a ally and clinically relevant in patients with ovarian cancer.
switch from oxidative phosphorylation to glycolysis is important
for memory T cell activation and effector function9,10. Given that RESULTS
nutrients (including glucose) are poorly replenished in the tumor, EZH2+ T cells are polyfunctional and apoptosis resistant
T cell glycolytic metabolism is assumed, reasonably, to be altered in Immunohistochemical analysis has demonstrated that memory T cell
the tumor microenvironment10. However, whether and how human tumor infiltration is associated with improved cancer patient sur-
primary tumor-associated glycolytic metabolism affects effector vival15–17. However, it is unknown which specific and functional T cell
T cell phenotype, function and in vivo antitumor immunity and clinical subsets mediate antitumor immunity in vivo and are associated with
outcome remains to be defined in patients with cancer. long-term patient survival. EZH2 has been reported to control both
The methyltransferase EZH2 is the catalytic subunit of the TH1 and TH2 cell differentiation from naïve T cells in mice13,14. We
polycomb-group family that trimethylates histone H3 on Lys27 hypothesized that EZH2 might regulate the effector cytokine pro-
(H3K27me3)11,12. H3K27me3 is a repressive epigenetic mark and file of memory T cells in humans and, particularly, in patients with
mediates transcriptional repression in cancer cells12. Published studies cancer. To explore the link between EZH2 and T cell function, we
suggest that EZH2 is involved in differentiation of type 1 and type examined EZH2+ T cells in different human tissues and analyzed
2 helper T cells (TH1 and TH2 cells) in mice13,14. In the current study, their phenotype. Immunofluorescence staining revealed the presence

1Department of Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA. 2Department of Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China. 3Department of Biostatistics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA.
4Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA. 5Department of Pathology, University of Michigan School of Medicine,

Ann Arbor, Michigan, USA. 6Chemical Biology Laboratory, Center for Cancer Research, NCI-Frederick, Frederick, Maryland, USA. 7The First Department of Gynecologic
Oncology and Gynecology, Medical University of Lublin, Lublin, Poland. 8Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China. 9Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA.
10Department of Obstetrics and Gynecology, University of Michigan School of Medicine, Arbor, Michigan, USA. 11Graduate Programs in Immunology and Cancer Biology,

University of Michigan, Ann Arbor, Michigan, USA. 12Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA. 13These authors contributed
equally to this work. Correspondence should be addressed to W.Z. (wzou@med.umich.edu) or G.W. (wangguobin1954@126.com).

Received 11 November 2014; accepted 30 September 2015; published online 2 November 2015; doi:10.1038/ni.3313

nature immunology  aDVANCE ONLINE PUBLICATION 


Articles

of EZH2+CD3+ T cells in tonsil, spleen and ulcerative colitic colon tis- DZNep20 resulted in fewer CD8+ T cells with double- or triple-positive
sues (Supplementary Fig. 1a). Polychromatic flow cytometry analysis expression of IFN-γ, TNF and granzyme B, as compared to untreated
demonstrated that peripheral blood EZH2+ T cells were confined cells (Fig. 2b,c). To confirm these results, we knocked down EZH2
to CD45RA−CD62L−CD45RO+ memory cells (Fig. 1a). Neither expression with EZH2-specific short hairpin RNA (shEZH2). As
EZH2+CD4+ nor EZH2+CD8+ T cells expressed KLRG1, Tim-3 expected, shEZH2 caused reduced expression of EZH2 and H3K27me3
or CD57, markers associated with T cell anergy and senescence6,8 and decreased percentages of polyfunctional CD8+ T cells, as com-
(Fig. 1b). Thus, EZH2+ T cells are different from anergic and pared to a scramble vector (Fig. 2d–f).
senescent memory T cells. To test the effect of EZH2-associated H3K27me3 on T cell polyfunc-
We further examined the effector cytokine profile and cytotoxic tionality, we used GSK126, an EZH2-specific S-adenosylmethionine-
protease of EZH2+ T cells (Supplementary Fig. 1b). EZH2+CD4+ competitive inhibitor compound21. T cells treated with GSK126
T cells were enriched with cells expressing two or three of the showed complete loss of EZH2-associated H3K27me3 without
effector cytokines interleukin 2 (IL-2), interferon-γ (IFN-γ) and any change in EZH2 protein abundance (Fig. 2g). GSK126 treat-
tumor necrosis factor (TNF) (Fig. 1c,d). EZH2+CD8+ T cells were ment inhibited polyfunctionality, as shown by reduced percentages
enriched with cells expressing two (double positive) or three (tri- of double- and triple-positive CD8+ T cells (Fig. 2h,i). These data
ple positive) of the effector molecules IFN-γ, TNF and granzyme indicate that EZH2 regulates T cell polyfunctionality via EZH2-
B (Fig. 1c,d). These data indicate that a high proportion of EZH2+ associated H3K27me3.
T cells express multiple effector cytokines (i.e., are polyfunctional) As EZH2+ T cells are more resistant to apoptosis, we wondered
(Supplementary Fig. 1b). whether EZH2 is also involved in the control of effector T cell sur-
Polychromatic flow cytometry analysis also detected EZH2+CD8+ vival. Flow cytometry analysis showed that treatment with DZNep or
T cells in human ovarian cancer tissue. Ovarian cancer–infiltrating GSK126 increased the percentage of apoptotic (annexin V+) T cells
© 2015 Nature America, Inc. All rights reserved.

EZH2+CD8+ T cells were phenotypically distinct from KLRG1+CD8+ under T cell antigen receptor (TCR) engagement (Fig. 2j). Similarly,
T cells, Tim-3+CD8+ T cells and CD57+CD8+ T cells (Fig. 1e). shEZH2 treatment increased T cell apoptosis (Fig. 2j). In further sup-
Ovarian cancer–infiltrating EZH2+CD8+ T cells were also enriched port of the nature of EZH2-mediated resistance to apoptosis, DZNep
with polyfunctional T cells (Fig. 1f). TUNEL assay showed that there treatment lowered Bcl-2 mRNA and protein expression in T cells, as
were fewer spontaneously apoptotic T cells in EZH2+ T cells than in compared to untreated cells (Supplementary Fig. 2b and Fig. 2k).
EZH2− T cells in ovarian cancer tissues (Fig. 1g). Consistent with the Inhibition of EZH2 promoted the expression of genes encoding the
antiapoptotic role of Bcl-2 in human effector T cells18,19, we observed proapoptotic proteins BAK, BAX and BIM in T cells (Supplementary
that polyfunctional T cells and EZH2+ T cells expressed high amounts Fig. 2c). T cells transduced with shEZH2 expressed less Bcl-2 than
of Bcl-2 (Fig. 1h,i). These data suggest that EZH2+ T cells may have a did cells treated with scrambled vector (Fig. 2l). Furthermore, knock-
survival advantage. To further explore this possibility, we examined down of EZH2 reduced BCL2 promoter activity (Fig. 2m). These data
T cell survival in the presence of cisplatin, a first-line chemother- indicate that EZH2 may regulate Bcl-2 expression and consequently
apeutic agent for ovarian cancer. Cisplatin induced CD8+ T cell control effector T cell survival. Thus, EZH2 is a key regulatory gene
apoptosis (Supplementary Fig. 1c) and increased the percentage of controlling effector T cell polyfunctionality and survival.
polyfunctional CD8+ T cells, as compared to cells cultured without
cisplatin. Cisplatin treatment did not affect the absolute numbers of EZH2 inhibits T cell Notch repressors
polyfunctional CD8+ T cells (Fig. 1j). These data indicate that poly- We next explored how EZH2 regulates effector T cell polyfunctional-
functional CD8+ T cells are relatively resistant to cisplatin-induced ity and survival. The Notch signaling pathway regulates effector T cell
apoptosis. Consistent with this observation, EZH2 expression was survival19,22 and effector function, including cytokine expression23. We
higher in CD8+ T cells treated with cisplatin than in untreated cells hypothesized that EZH2 alters the Notch signaling pathway and in turn
(Supplementary Fig. 1d). EZH2+CD8+ T cells were enriched with affects T cell cytokine profile and survival. As expected, when human
polyfunctional T cells (Fig. 1f) and were less susceptible to sponta- T cells were activated, EZH2 and intracellular domain of Notch (NICD)
neous (Fig. 1g) and chemotherapy-induced (Fig. 1j) apoptosis than were synchronically induced (Fig. 3a). Knockdown of EZH2 with
were EZH2− T cells in the human ovarian cancer microenvironment. shEZH2 decreased the amount of NICD (Fig. 3b). DZNep treatment
Thus, EZH2+ T cells may be functional effector T cells in the tumor suppressed Notch signaling activation as shown by reduced expres-
microenvironment and may mediate potent antitumor immunity. sion of HES1, HEY1 and HEY2 , which are direct targets of NOTCH
However, the ratios between the percentage of EZH2+CD8+ effector (Fig. 3c). Those data indicate that EZH2 may promote acivation of
T cells and that of dysfunctional T cells (KLRG1+, Tim-3+ or CD57+) Notch signaling in T cells. EZH2 is one of the Polycomb group family
were lower in ovarian cancer than in peripheral blood (Fig. 1k–m) members that functions in transcriptional repression12. We hypoth-
and noncancerous ovary cysts (Fig. 1n–p). The imbalance between esized that EZH2 targets and represses Notch repressors and conse-
functional effector T cells and dysfunctional T cells suggests a loss of quently activates Notch. To test this hypothesis, we analyzed expression
polyfunctional effector T cells in the cancer microenvironment. of NUMB and FBXW7, two important Notch suppressors24,25, after
blockade of EZH2 expression. We found that blockade with DZNep
EZH2 regulates effector T cell polyfunctionality and survival led to elevated expression of NUMB and FBXW7 in comparison to
Given that EZH2 expression coincided in polyfunctional and apoptosis- vehicle treatment (Fig. 3d). ChIP assays revealed high occupancies
resistant effector T cells, we hypothesized that EZH2 may functionally of H3K27me3 in the proximal promoter areas of NUMB and FBXW7,
regulate T cell polyfunctional cytokine profile and survival. To test this whereas genetic knockdown of EZH2 abrogated these occupancies
hypothesis, we activated T cells with antibodies to CD3 (anti-CD3) and (Fig. 3e). These data suggest that EZH2 targets Notch repressors and
anti-CD28. We observed an increase (as compared to freshly isolated promotes Notch activation and may, consequently, regulate effector T
T cells) in expression of EZH2 and H3K27me3 upon T cell activation cell polyfunctionality and survival. In line with this, we observed that
(Fig. 2a and Supplementary Fig. 2a). Notably, biochemical inhibi- Notch inhibitors DAPT and GSI-I suppressed CD8+ T cells double or
tion of EZH2 expression with the histone methyltransferase inhibitor triple- positive for IFN-γ, TNF and granzyme B, and increased effector

 aDVANCE ONLINE PUBLICATION  nature immunology


Articles

a b c d 70 CD4
*
CD8

CD4 CD4 60
CD4 CD8 *

EZH2

EZH2
50
Triple-

SSC
66% 46% positive

EZH2 (%)
40
Double- 30
CD45RA CD62L CD45RO KLRG1 Tim-3 CD57 16% 31% positive
CD8 CD8 20
Single-
11% 36% positive

EZH2
EZH2

10

Negative 0
7% 4%

b os e
ip os e
os e
ve

b o e
ip os e
os e
ve
ou -p iv
Tr le-p itiv
-p iv

ou -p tiv
Tr le-p sitiv
-p iv
iti

iti
D gle gat

le it

le it
D gle ga
CD45RA CD62L CD45RO KLRG1 Tim-3 CD57 EZH2

n e

n e
Si N

Si N
e f g CD3 DAPI CD3 TUNEL h
Ovarian cancer
80
*
35 * 1,200
EZH2

8 5 15 3 11 3

Triple-positive CD8 T cells (%)


35 28 23 30 60

CD3+ TUNEL+ (%)


1,000

Bcl-2 (MFI)
25 800
40
+

20 600
KLRG1 Tim-3 CD57 *
15 400
Ovarian cyst 20
CD3 EZH2 Merged
© 2015 Nature America, Inc. All rights reserved.

10 200
5 0.1 6 0.4 10 0.2
EZH2

5 0 0
4 0.7 3

+

ve
os l
tro
2
2

iti
0

H
H

on
EZ
EZ

C
– +

-p
EZH2 EZH2

le
ip
Tr
KLRG1 Tim-3 CD57

i j k l m n 3
12 * 30 1.2 1.5
Triple-positive cell number (×104)

1,600 8 2.5
Triple-positive CD8+ T cells (%)

5
Total CD8 cell number (×10 )
5

EZH2/KLRG1 ratio
7
10 25 1.0
* * 2 *
4
EZH2/KLRG1 ratio

EZH2/Tim-3 ratio
6

EZH2/CD57 ratio
1,200
8 20 0.8 1.0
Bcl-2 (MFI)

5 1.5
3
800 6 15 0.6 4
1.0
* * 2 3
+

4 10 0.4 0.5
400 2 0.5
2 5 0.2 1
1 0

st

er
0 0 0 0 0 0 0

cy

nc

ca
n
d

er

er

ca d
er
in

in

in
l

l
tro

tro

tro

ria
oo

oo

oo
2

at

at

at

nc

nc

nc

n
H

on

on

on

va
Bl

Bl

Bl
pl

pl

pl

ria
ca

ca
EZ

EZ

is

is

is
C

O
va
C

n
ria

ria

ria

O
va

va

va
O

O
Figure 1  EZH2+ T cells endow polyfunctional and apoptosis-resistant features. (a,b) Phenotype of EZH2+ 4 0.6 o p
T cells. Peripheral blood mononuclear cells from healthy donors were stained with antibodies to EZH2 and
3.5 *
CD45RA, CD62L and CD45RO (a) or KLRG1, Tim-3 and CD57 (b) and T cell markers and analyzed *
3
with LSR II. One representative of 8 donors is shown. (c,d) EZH2 and polyfunctional T cells in blood.
EZH2/CD57 ratio
EZH2/Tim-3 ratio

2.5 0.4
Intracellular staining was performed in peripheral blood mononuclear cells for IFN-γ, TNF and granzyme B.
2
EZH2 expression was analyzed in CD4+ T cells that were negative or single- , double- or triple-positive
+ 1.5
for IL-2, IFN-γ and TNF or CD8 T cells that were negative or single- , double- or triple-positive for IFN-γ, 0.2
1.0
TNF and granzyme B. Shown are results from 1 of 6 flow cytometry dot plots (c) and N = 6 donors,
0.5
mean and s.e.m. Numbers on dot plots represents the percentage of EZH2-positive T cells. (d). *P < 0.05
0 0
(Wilcoxon rank-sum test (d)). (e) EZH2+ T cells in ovarian cancer. Single cells were made from ovarian cancer
st

er

st

er

tissues and were stained for T cell markers, KLRG1, Tim-3, CD57 and EZH2. CD8+ T cells were analyzed
cy

cy
nc

nc
ca

ca
n

n
ria

ria

by flow cytometry. Numbers in quadrants indicate the percentages of the single- and double-positive cells in the
n

n
va

va
ria

ria
O

O
va

va

CD45+CD3+CD8+gate. Data are from 1 representative donor of 20. (f) EZH2 and polyfunctional T cells in ovarian
O

cancer. Intracellular staining was performed in single cells made from ovarian cancer tissues for T cell markers, IFN-γ, TNF, granzyme B, and EZH2. IFN-γ,
TNF, and granzyme B triple positive (polyfunctional) CD8+ T cells were analyzed on the basis of EZH2 expression. N = 5 patients. *P < 0.01 (Wilcoxon
rank-sum test). (g) Relationship between EZH2 expression and T cell apoptosis in ovarian cancer. Frozen ovarian cancer tissues were stained with anti-
CD3 (green), anti-EZH2 (white), TUNEL (red) and DAPI (blue). TUNEL+CD3+EZH2+ T cells (white) are marked with white arrows. The percentage of
TUNEL+CD3+ T cells was determined in EZH2+ and EZH2− T cells. N = 10 patients (mean and s.e.m.). *P < 0.05 (Wilcoxon rank-sum test). (h) Expression
of Bcl-2 in polyfunctional T cells. CD8+ T cells were stimulated with anti-CD3 and anti-CD28 for 2 d. Expression of IFN-γ, TNF, granzyme B and Bcl-2 was
analyzed by flow cytometry. Results are shown as the mean fluorescent intensity (MFI) of Bcl-2 expression in polyfunctional (triple-positive) CD8 + T cells.
N = 6 donors (mean and s.d.). *P < 0.05 (Wilcoxon rank-sum test). (i) Expression of Bcl-2 in EZH2+CD8+ T cells. CD8+ T cells were stimulated with
anti-CD3 and anti-CD28 for 2 d. Expression of EZH2 and Bcl-2 was analyzed by flow cytometry. Results are shown as the MFI of Bcl-2 expression in
EZH2+CD8+ T cells. N = 6 (mean and s.d.). *P < 0.05 (Wilcoxon rank-sum test). (j) Effects of cisplatin on polyfunctional T cells. CD8+ T cells were
stimulated with anti-CD3 and anti-CD28 for 5 d. Cisplatin was added in culture on day 4. Cells were stained for polyfunctional phenotype (IFN-γ, TNF and
granzyme B). Total T cell number, the percentage and absolute numbers of triple-positive CD8 + T cells is shown. N = 4 donors. *P < 0.05 (Wilcoxon rank-
sum test). (k–m) Ratios of EZH2/KLRG1 (k), EZH2/Tim-3 (l), and EZH2/CD57 (m) in ovarian cancer infiltrating T cells and peripheral blood T cells. The
levels of KLRG1, Tim-3, and EZH2 expressing CD8+ T cells were analyzed by flow cytometry. N = 30 patients (mean and s.e.m.). *P < 0.05 (Mann-Whitney
U -test). (n–p) Ratios of EZH2/KLRG1 (n), EZH2/Tim-3 (o) and EZH2/CD57 (p) in ovarian cancer infiltrating T cells and T cells in normal ovary cysts. CD8 +
T cells were analyzed by flow cytometry. Ovary cyst tissues, N = 11; cancer samples, N = 7 (mean and s.e.m.). *P < 0.05 (Mann-Whitney U -test).

nature immunology  aDVANCE ONLINE PUBLICATION 


Articles

a b c d e f g h i

Double-positive T cells (%)

Double-positive T cells (%)


Triple-positive T cells (%)

Triple-positive T cells (%)


40 20 40 20

Double-positive T cells (%)

Triple-positive T cells (%)


35 10
CD3 and CD28 – + + 30 15 30 8 CD3 and CD28 + + 30 15
DZNep – – + EZH2 25 * GSK126 – +
EZH2 20 * 10 H3K27me3 20 * 6 EZH2 20 10
H3
β-actin
β-actin 15 4 H3K27me3 *
H3K27me3 10 5 * 10 H3 10 5
H3 2 *

sh ble
5 β-actin

2
H
m
EZ
0 0 0 0 0 0

ra
Sc
ep

ep

6
EZ e
D trol

EZ e

SK l
6
D trol

SK l
2

G ntro
2

G ntro
12
sh mbl

sh mbl

12
H

H
ZN

ZN
on

on

o
ra

ra
C

C
C

C
Sc

Sc
j k l

7-AAD
Figure 2  EZH2 regulates effector T cell polyfunctionality and survival. (a) Effects of DZNep

shEZH2 GSK126 DZNep Control


5
Bcl-2 Bcl-2
on EZH2 and H3K27me3 expression in T cells. T cells were stimulated with anti-CD3 and
EZH2 EZH2
anti-CD28 antibodies for 3 d in the absence or presence of 5 µM DZNep. The expression of β-actin β-actin
20
EZH2, H3K27me3, H3 and β-actin was measured with immunoblotting. Shown are data from 1

ep

EZ e
2
D trol

sh bl
H
+
representative experiment of 4. (b,c) Effects of DZNep on polyfunctionality of CD8 T cells. CD8 +

ZN

m
on

ra
C
T cells were activated with anti-CD3 and anti-CD28 antibodies for 3 d in the absence or presence

Sc
18
of 5 µM DZNep. Expression of IFN-γ, TNF, and granzyme B was analyzed by flow cytometry. The
percentage of double- (b) and triple-positive (c) cells is shown. N = 5 donors (mean and s.e.m.).
Bcl-2 promoter activity (%) m
0 25 50 75 100
*P < 0.05 (Wilcoxon rank-sum test (b,c)). (d–f) Effects of shEZH2 on EZH2 and H3K27me3 19
+
expression and polyfunctionality in CD8 T cells. T cells transfected with shEZH2 or scramble Scramble
shEZH2 *
vectors were activated for 3 d. The expression of EZH2 and H3K27me3 was measured with Annexin V
© 2015 Nature America, Inc. All rights reserved.

immunoblotting (d). Expression of IFN-γ, TNF and granzyme B was analyzed by flow cytometry.
The percentage of double- (e) and triple-positive (f) cells is shown. N = 3 donors analyzed in duplicate. *P < 0.05 (Student’s t-test (e,f)). (g–i) Effects of
GSK126 on H3K27me3 expression and polyfunctionality in CD8 + T cells. T cells were stimulated with anti-CD3 and anti-CD28 for 4 d. 10 µM GSK126
was added on days 0 and 2. The expression of EZH2, H3K27me3, H3 and β-actin was detected with immunoblotting in 3 donors analyzed in duplicate (g).
Expression of IFN-γ, TNF and granzyme B was analyzed by flow cytometry. The percentage of double- (h) and triple-positive (i) cells is shown. N = 5
donors (mean and s.e.m.). *P < 0.05 (Wilcoxon rank-sum test (h,i)). (j) Effects of EZH2 on T cell apoptosis. T cells were treated with 5 µM DZNep, 10 µM
GSK126 or shEZH2 transfection and activated with anti-CD3 and anti-CD28 for 2 d. T cell apoptosis was evaluated with annexin V and 7-AAD staining.
Representative dot plots for 1 of 5 donors tested are shown. Numbers on dot plots represent fraction of annexin V–binding apoptotic cells. (k,l) Effects of
EZH2 blockade on Bcl-2 expression. T cells were treated with 5 µM DZNep (k) or transfected with shEZH2 (l) and activated for 3 d. Expression of Bcl-2,
EZH2 and β-actin was detected with immunoblotting. One of 5 experiments per condition is shown. (m) Effect of EZH2 blockade on Bcl-2 promoter activity.
293T cells were co-transfected with vectors encoding shEZH2 or scramble and a vector carrying GFP-tagged Bcl-2 for 2 d. The promoter activity was analyzed
by flow cytometry and expressed as the percentage of GFP expression. N = 4 independent experiments (mean and s.d.). *P < 0.05 (Mann-Whitney U -test).

T cell apoptosis, in comparison to vehicle treatment (Fig. 3f–h and Glucose restriction impairs T cell function in tumors
Supplementary Fig. 3a). Furthermore, ectopic expression of the Polyfunctional T cells mediate antiviral immunity1–5. Many studies
Notch active domain (Notch-IC) increased BCL2 promoter activity have focused on T cell exhaustion and senescence. However, the reason
(Supplementary Fig. 3b). These data suggest that EZH2 targets Notch for the relative loss of EZH2+ polyfunctional T cells in the cancer
repressors and promotes Notch activation, affects Bcl-2 activation and microenvironment is poorly understood. To address this question,
controls effector T cell polyfunctionality and survival. we initially examined how polyfunctional T cells were regulated in

a c d e IgG f g h *
Stimulation 0.06 Scramble
1 Control Control shEZH2 50 20 15
0 h 24 h 72 h *
DZNep *
Double-positive T cells (%)

DZNep 0.05
Triple-positive T cells (%)

NICD
Annexin V+ T cells (%)

0.8 40
6 * 15
Gene expression

EZH2
* 5
* 0.04 10
Input (%)

*
Gene expression

β-actin 0.6 30 *
* 4 * 0.03 10
0.4 3 20 *
0.02 5
b NICD
0.2
2
0.01 10
5
*
EZH2 1 *
β-actin 0 0 0 0 0 0
NUMB FBXW7
N rol
FB B

G l
D -I
T
7

l
-I

T
l
1

H 1

G ol
D -I
T
2

tro

tro
tro

SI
AP
XW
ES
EY

SI
AP
EY

SI
AP
tr
sh ble

t
on

on

on
on

on
H

G
H
H

D
EZ
m

C
C

C
ra
Sc

Figure 3  EZH2 inhibits Notch repressors, T cell polyfunctionality and survival. (a) Kinetic activation of EZH2 and Notch in T cells. T cells were
stimulated with anti-CD3 and anti-CD28 for the indicated time. Expression of NICD, EZH2 and β-actin was detected with immunoblotting. One
representative experiment of 3 is shown. (b) Effect of EZH2 blockade on Notch activation. ShEZH2 lentivirus transduced T cells were activated for 3 d.
Expression of NICD, EZH2 and β-actin was detected with immunoblotting. One representative experiment of 3 experiments is shown. (c,d) Effect of
DZNep on Notch signaling gene expression. Human T cells were activated in the presence of 5 µM DZNep for 2 d. Expression of active Notch signaling
genes (HES1, HEY1 and HEY2) donors (c), and Notch repressor genes (NUMB and FBXW7) (d) was quantified by real-time PCR. Results are shown
as expression relative to vehicle-treated cells. N = 5 donors (mean and s.d.). *P < 0.05 (Wilcoxon test (c,d)). (e) H3K27me3 binding to the proximal
promoter areas of NUMB and FBXW7. Jurkat cells were transfected with shEZH2 or scramble. ChIP assay was performed with anti-H3K27me3 antibody.
The occupancy of H3K27me3 in the proximal promoter areas of NUMB and FBXW7 was examined and expressed as the percentages of input. N = 4
transfections, each done in triplicate (mean and s.e.m.). *P < 0.05 (Wilcoxon test). (f–h) Effects of Notch signaling inhibition on T cell polyfunctionality and
apoptosis. CD8+ T cells were activated with anti-CD3 and anti-CD28 in the presence of two gamma-secretase inhibitors: DAPT and GSI-I. Polyfunctionality
(f,g) and apoptosis (h) were analyzed by flow cytometry. Data from 3 donors are shown (mean and s.e.m.). *P < 0.05 (Mann-Whitney U -test).

 aDVANCE ONLINE PUBLICATION  nature immunology


Articles

a b Normal medium
Tumor medium
c
Normal medium + glucose
6 Tumor medium + glucose
10 0.16
Activation
9 0.14 Activation
8
* 0.12

ECAR (mpH/min)
Glucose (mg/ml)

4 7

ECAR/OCR
6 0.10
5 0.08
4 0.06
2 3
0.04
2
* 1 0.02
0 0
0 0 17 35 52 69 86 104 121 138 155 0 17 35 52 69 86 104 121 138 155
or me m
um

m co m

uc um
iu

Time (min)
e

Time (min)
di

Tu gl edi

gl di
m ed

or s

os
+ me
Tu l m

+ m
u
or

al
a
m

m
or

d e f g h
N

Control
Figure 4  Tumor impairs T cell Control Glucose
80 30 15 8
Control 25
polyfunctionality and survival via glucose Glucose
*
70 Glucose

Annexin V–positive cells (%)


7

Annexin V–positive cells (%)


restriction. (a) The concentrations of 25
Double-positive cells (%)

Triple-positive cells (%)

Triple-positive cells (%)


60 6 20
glucose in different culture media.
20 10 *
Glucose was detected in normal medium, 50
* 5
15
*
© 2015 Nature America, Inc. All rights reserved.

primary ovarian cancer cell medium, 40 15 * 4


and these two media supplemented with 30 10
10 3
additional glucose. Tumor medium was 5
20 2
supplemented with an additional 2 mg/ml 5 5
glucose after culture with tumor cells. 10 1
N = 4 donors (mean and s.e.m.). *P < 0.05 0 0 0 0 0
Control Tumor Control Tumor Control Tumor Control 2-DG Control 2-DG
(Wilcoxon rank-sum test). (b,c) Metabolic
profile of normal and tumor-conditioned memory CD8 + T cells. Memory CD8+ T cells were pre-incubated overnight in medium or tumor-conditioned
supernatants with or without glucose supplementation, and activated with anti-CD3 and anti-CD28. ECAR (b) and ECAR/OCR ratio (c) were assayed in
different time points in a Seahorse XF24 analyzer. Representative data from one of 4 donors, each condition was repeated 4 times (mean and s.e.m.).
(d–f) Effects of tumor-associated glucose restriction on CD8 + T cell polyfunctionality and apoptosis. Memory CD8 + T cells were pre-incubated overnight
in medium or tumor-conditioned supernatants with or without glucose supplementation and washed and stimulated for 48 h with anti-CD3 and anti-
CD28 antibodies. Polyfunctional profile was analyzed by flow cytometry. Results are shown for double-positive (d) and triple-positive (e) cells for IFN-γ,
TNF and granzyme B. T cell apoptosis (f) was determined by annexin V staining and analyzed by flow cytometry. Results are from 4 donors (mean and
s.e.m.). *P < 0.05 (Mann-Whitney U -test). (g,h) Effects of 2-DG on CD8+ T cell polyfunctionality and apoptosis. CD8+ T cells were activated with anti-
CD3 and anti-CD28 for 3 d in the presence or absence of 2-DG. Polyfunctionality (g) and annexin V binding (h) were assessed by flow cytometry. Results
are from 4 donors (mean and s.d.). *P < 0.05 (Wilcoxon rank-sum test).

the tumor microenvironment. A switch from oxidative phosphoryla- Again, glucose supplementation reversed these effects (Fig. 4d–f).
tion to aerobic glycolysis is crucial for T cell acquisition of effector Similar results were obtained in polyfunctional CD4+ T cells
functions9,10. We hypothesized that glucose restriction in tumor tis- (Supplementary Fig. 4a,b). We also cultured CD8+ T cells in nor-
sue reduces numbers of polyfunctional T cells. To test this hypoth- mal medium containing glucose at concentrations similar to that
esis, we cultured CD8+ T cells with RPMI 1640 with 10% FCS and of tumor medium (0–1 mg/ml). We found that low concentrations
2 mg/ml glucose (normal medium), tumor-conditioned medium from of glucose resulted in reduced numbers of polyfunctional CD8+
primary ovarian cancer cells26, normal medium supplemented with T cells (Supplementary Fig. 4c) and a higher frequency of apoptosis
2 mg/ml glucose and tumor medium supplemented with 2 mg/ml (Supplementary Fig. 4d). We also examined the influence of glu-
glucose (Fig. 4a). We subsequently analyzed the ability of these cells cose on the expression of ligand for PD-1 (PD-L1 (B7-H1)) and B7
to undergo a metabolic switch and its association with polyfunctional homolog 4 (B7-H4, B7x, B7s) in ovarian cancer cells. Glucose restric-
T cells. Tumor medium contained low concentrations of glucose (Fig. 4a). tion or supplementation did not alter their expression (Supplementary
As expected, T cells cultured in normal medium responded efficiently Fig. 4e). Furthermore, the hexokinase inhibitor 2-deoxy-d-glucose
to TCR-engaged activation and increased extracellular acidification (2-DG) treatment reduced the number of polyfunctional T cells
rate (ECAR). In contrast, CD8+ T cells cultured in tumor medium and increased the percentage of apoptotic T cells, as compared to
were poorly responsive to activation, as demonstrated by reduced untreated cells (Fig. 4g,h and Supplementary Fig. 4f). Taken together,
ECAR (Fig. 4b). Glucose supplementation partially rescued ECAR these data suggest that tumor impairs T cell glycolysis, inhibits their
in T cells cultured with tumor medium but had no effect on ECAR in polyfunctionality and promotes their apoptosis.
T cells cultured with normal medium (Fig. 4a,b). We further evalu-
ated the ratio between ECAR and oxygen consumption rate (OCR) in Tumor controls T cell EZH2 via specific miRNAs
CD8+ T cells conditioned with ovarian cancer medium. We observed Given our observations that EZH2 regulates T cell polyfunctionality
a lower ECAR/OCR ratio in tumor-conditioned T cells than in and survival, tumor microenvironments impairs T cell polyfunction-
T cells conditioned in normal medium (Fig. 4c). Accompanied with ality and survival via glucose restriction and T cell activation induces
a reduced ECAR and ECAR/OCR ratio, culture with tumor medium EZH2 expression, we hypothesized that the tumor environment
resulted in lower numbers of polyfunctional CD8+ T cells, as shown controls T cell EZH2 induction via glucose restriction and, in turn,
by reduced frequencies of double-positive and triple-positive CD8+ regulates T cell polyfunctionality and apoptosis. To test this hypoth-
T cells and high frequencies of apoptotic CD8+ T cells (Fig. 4d–f). esis, we activated and cultured T cells in the presence of medium

nature immunology  aDVANCE ONLINE PUBLICATION 


Articles

Figure 5  Tumor controls T cell EZH2 by a Control Control b Control c d e


miR-101 and miR-26a via glucose restriction. Glucose Glucose 2-DG
(a,b) Effect of tumor-associated glucose 600 1,000 1 1 1

EZH2 expression (MFI)


EZH2 expression (MFI)

miR-26a expression
miR-101 expression
restriction on T cell EZH2 expression. CD4+ (a) 500

expression (MFI)
800 0.8 0.8 0.8

Relative EZH2
and CD8+ (b) T cells were activated with anti- 400 *
* 600 * 0.6 0.6 0.6
CD3 and anti-CD28 for 3 d in medium or tumor 300 *
400 0.4 0.4 0.4
conditioned medium with or without glucose 200
Control Control
supplementation. EZH2 protein expression was 100 200 0.2 0.2 0.2
2-DG 2-DG
determined by flow cytometry. Results are from 0 0 0 0 0
0 2 4 6 8 1012 0 2 4 6 8 1012

+
3 donors (mean fluorescence intensity (MFI) of

l
or

Tu l
or
tro

tro

8
m

D
n

n
Tu
Time (h) Time (h)

Co

Co

C
EZH2 and s.e.m.). *P < 0.05 (Mann-Whitney
U -test). (c) Effect of 2-DG on T cell EZH2
expression. T cells were activated with anti-
f 2 2 1 g 25 25 h i j
*
* *

miR-26a expression
miR-101 expression

Annexin V–positive
CD3 and anti-CD28 for 3 d in the presence or 0.8 20 20
*

expression (MFI)

CD8+ T cells (%)


CD8+ T cells (%)
1.5 1.5 *

Relative EZH2

Triple-positive
absence (control) of 2-DG. EZH2 expression was * 0.6 15 15
determined in viable T cells by flow cytometry. *
1 1 *
Results are normalized to expression in the 0.4 * 10 * 10
* *
control group (N = 3 donors, mean and s.e.m.). 0.5 0.5 0.2 5 5
*P < 0.05 (Mann-Whitney U -test).
0 0 0 0 0
(d,e) Dynamic changes of miR-101 and
T um
+ uc or
uc e
2- se
G

T um
+ luc or
uc e
2- se
G

-2 m ol
m ic
ic

6a im l
m ic
ic

-2 m rol
m ic
ic
-2 m ro
miR-26a in activated CD8+ T cells. CD8+ T cells gl os

gl os

6a im
im

im

6a im
im
iR 1 tr
D

D
or G um

or G um
o

iR 01 nt

iR 1 nt
m -10 Con
i

i
ed

ed

m -1 Co

m -10 Co
were activated with anti-CD3 and anti-CD28 in
l
M

iR

iR

iR
the presence or absence of 2-DG. MiR-101
© 2015 Nature America, Inc. All rights reserved.

m
Tu

Tu
(d) and miR-26a (e) were measured by qPCR
in different time points. Results are shown as the expression of miRNA relative to that at time point 0. One of 2 per group is shown. (f,g) Effect of
tumor-associated glucose restriction on miRNA expression in CD8 + T cells. Memory CD8+ T cells were pre-incubated overnight in medium or tumor-
conditioned supernatant with or without glucose supplementation, then washed and stimulated for 12 h with anti-CD3 and anti-CD28. The abundance
of miR-101 (f) and miR-26a (g) was measured by qPCR. N = 3 donors in triplicate (mean and s.e.m.). *P < 0.05 (Mann-Whitney U -test). (h–j) Effects
of miRNA mimics on T cell EZH2 expression, polyfunctional profile and apoptosis. Memory CD8 + T cells were transfected with miR-101 mimic,
miR-26a mimic or control oligonucleotide, and activated with anti-CD3 and anti-CD28 for 48 h. T cell EZH2 expression (h), polyfunctionality (i) and
apoptosis (j) were determined by flow cytometry. N = 4 donors (mean and s.e.m.). *P < 0.05 (Mann-Whitney U -test).

from cultured primary ovarian cell cultures and examined their EZH2 and enhanced T cell apoptosis (Fig. 5g–j). Accordingly, the absolute
expression. We observed that tumor medium impaired EZH2 expres- numbers of viable T cells were reduced in cells transfected with miR-
sion in both CD4+ and CD8+ T cells (Fig. 5a,b and Supplementary 101 and miR-26a mimics (Supplementary Fig. 5e).
Fig. 5a). Notably, glucose supplementation of growth medium rescued miR-101 (ref. 27) and miR-26a28 target the gene encoding EZH2
EZH2 expression (Fig. 5a,b and Supplementary Fig. 5a). Low con- through its 3′ untranslated region (UTR). To determine whether
centrations of glucose in fresh culture medium also caused reduced miR-101 and miR-26a may target other genes and regulate T cell
EZH2 expression (Supplementary Fig. 5b). These data suggest that survival and polyfunctionality, we constructed plasmid encoding
T cell EZH2 expression was impaired via glucose restriction. In line EZH2 without a 3′ UTR27 and overexpressed this construct in T cells.
with this possibility, we observed that 2-DG strongly inhibited EZH2 As expected, miR-101 and miR-26a mimics had no effect on EZH2
expression in CD4+ and CD8+ T cells (Fig. 5c). Thus, tumor micro- expression in T cells with overexpression of EZH2 without 3′ UTR
environments reduce EZH2 expression via impairment of glucose (Supplementary Fig. 6a). miR-101 and miR-26a mimics inhibited
metabolism. Bcl2 expression, Hey1 expression and polyfunctional CD8+ T cell for-
Next, we examined how tumor cells impair EZH2 expression in mation in control T cells transfected with empty plasmid, but not in
T cells. The miRNAs miR-101 (ref. 27) and miR-26a (ref. 28) regulate EZH2-overexpressing T cells (Supplementary Fig. 6b–d). Thus, we
EZH2 expression in tumor cells. In parallel to the increase in EZH2 propose that cancer mediates effector T cell dysfunction by target-
expression, we observed a rapid reduction in abundance of miR-101 ing miRNAs, EZH2 and the Notch repressor signaling pathway via
and miR-26a in the course of T cell activation, whereas many miR- restriction of aerobic glycolysis.
NAs, including miR-223, miR-106b and miR-181a were not affected
(Fig. 5d,e and Supplementary Fig. 5c). In the presence of tumor cells, EZH2 affects tumor immunity and patient survival
however, the expression of miR-101 and miR-26a remained high and EZH2 marks polyfunctional T cells and controls their survival. We
was significantly reduced by the addition of glucose, whereas supple- wondered whether EZH2 might affect T cell–mediated antitumor
mentation of normal medium with additional glucose did not affect immunity in vivo. To investigate this question, we employed a mouse
miRNA expression (Fig. 5f,g). In line with this observation, miR- model of melanoma with lung metastasis. Adoptively transferred
101 and miR-26a were more abundant in T cells cultured in normal tumor-specific T cells protected mice from lung metastasis (Fig. 6a).
medium supplemented with limited glucose than in T cells grown Pre-treatment of T cells with DZNep inhibited the expression of EZH2
in medium containing normal glucose concentrations (2 mg/ml) and H3K27me3 and impaired the protective role of T cells, as indi-
(Supplementary Fig. 5d). This observation suggests that the tumor cated by an increased number of tumor foci in the lung (Fig. 6a).
microenvironment may promote and sustain miR-101 and miR-26a Similarly, EZH2 knockdown in T cells increased lung metastasis as
expression in T cells via glucose restriction. In further support of this, compared to a control shRNA with a scrambled sequence (Fig. 6b).
2-DG treatment prevented the loss of miR-101 and miR-26a in T cells We further examined the effects of EZH2 on T cell–mediated anti-
activated with TCR engagement (Fig. 5d–g). As expected, transfec- tumor immunity in a humanized ovarian cancer model19,29. Human
tion of T cells with miR-101 and miR-26a mimics led to the reduction ovarian cancer–specific T cells were treated with DZNep and intrave-
of EZH2 expression, decreased the frequency of polyfunctional T cells nously transfused into ovarian cancer–bearing mice19. We observed

 aDVANCE ONLINE PUBLICATION  nature immunology


Articles

a T cell DZNep Lung metastasis foci b 160 c 2,500


d
* Control

Lung metastasis foci

Tumor volume (mm3)


1
– –
120 2,000 T cells * High CD8+
T cell + DZNep
0.8 Low CD8+

Overall survival
1,500
+ – 80
1,000
* 0.6
0.4
40 500
0.2
+ + 0
0 0

sh ble
ra S

2
H

12

16

20

24

0
20
40
60
80

0
0
0
0
0
Sc PB

10
12
14
16
18
m
EZ
Time after inoculation (d) Time (months)

e 1 +
High CD8 f 1 High CD8+EZH2+ g 1 +
High CD8 EZH2 +
h 1 High CD8+EZH2+(%)
0.8 Low CD8+ 0.8 Low CD8+EZH2+ 0.8 Low CD8+EZH2+ 0.8 Low CD8+EZH2+(%)

Overall survival
Overall survival
Disease-free

Disease-free
survival

survival
0.6 0.6 0.6 0.6
0.4 0.4 0.4 0.4
0.2 0.2 0.2 0.2
0 0 0 0
0
20
40
60
80

0
0
0
0
0

0
20
40
60
80

0
0
0
0
0

0
20
40
60
80

0
0
0
0
0

0
20
40
60
80

0
0
0
0
0
10
12
14
16
18

10
12
14
16
18

10
12
14
16
18

10
12
14
16
18
Time (months) Time (months) Time (months) Time (months)

Figure 6  EZH2 affects T cell–mediated i 120 months (DFS) j ROC curve at


120 months (OS) k ROC curve at

antitumor immunity and patient survival.


© 2015 Nature America, Inc. All rights reserved.

+
AUC P + AUC P
(a) Effects of biochemical blockade CD8 74.76 CD8 62.11
+ + + + 0.01 + + 0.004
1 High CD8 EZH2 (%) CD8 EZH2 82.79 0.03 CD8 EZH2 73.45 0.007
of T cell EZH2 on mouse melanoma
Disease-free survival

Low CD8+EZH2+(%)
CD8+EZH2+ 92.44 0.09 CD8+EZH2+ 90.13 0.04
metastasis. Melanoma cells were 0.8 1 1
intravenously injected into C57BL/6 0.6
0.8 0.8
mice to induce lung metastasis. Tumor- 0.4

Sensitivity
Sensitivity
0.6 0.6
specific T cells were activated in the 0.2
presence or absence of 1 µM DZNep for 0.4 0.4
0
2 d and expanded in IL-2 for 3 d. These 0.2 0.2
0
20
40
60
80

0
0
0
0
0
10
12
14
16
18

T cells were intravenously transfused into


Time (months) 0 0
tumor-bearing mice. Lungs from all mice are shown;
N = 5 mice per group. (b) Effects of genetic blockade of T cell EZH2 on mouse 0 0.2 0.4 0.6 0.8 1 0 0.2 0.4 0.6 0.8 1
melanoma metastasis. Tumor-specific T cells transfected with lentiviral vector expressing Specificity Specificity
shEZH2 or scramble were activated for 2 d and expanded in IL-2 for 3 d. These T cells were intravenously transfused into tumor-bearing mice. Lung
metastatic loci were counted on day 16. N = 5–7 per group; each dot represents an individual mouse. *P < 0.05 (Mann-Whitney U -test, shEZH2 vs.
scramble). (c) Effects of T cell EZH2 blockade on human ovarian cancer growth. Human primary ovarian cancer cells were subcutaneously injected into
female NSG mice. Tumor-specific T cells were initially treated with or without 1 µM DZNep in vitro and intravenously transferred into ovarian cancer–
bearing NSG mice. Tumor volume was measured. N = 5 mice per group (mean ± s.e.m.). *P < 0.05 (Mann-Whitney U -test). (d–i) Tumor-infiltrating T cells
and patient outcome. Ovarian cancer tissue microarray was subject to immunohistochemistry staining with anti-CD8 and anti-EZH2 antibodies. CD8 +
and EZH2+CD8+ T cells were quantified. Kaplan-Meier estimates (Supplementary Tables 1–3) of overall survival and progression-free interval were done
according to the median values of CD8+ T cell numbers (d,e), EZH2+CD8+ T cell numbers (f,g) and the percentage of EZH2+CD8+ T cells (h,i).
(j,k) Relative impact of CD8+ T cell numbers, EZH2+CD8+ T cell numbers and the percentage of EZH2+CD8+ T cells on long-term survival. Time-
dependent receiver-operating characteristic (ROC) curve analysis was applied to evaluate the predictive accuracy of each marker for a 10-year overall
survival (OS) (j) and disease-free survival (DFS) (k). AUC, area under the curve.

greater tumor growth in mice that received tumor-specific T cells (Fig. 6e and Supplementary Table 1). However, EZH2+CD8+ T cell
pretreated with DZNep in vitro than in mice that received T cells numbers and the percentage of EZH2+CD8+ T cells highly correlated
not treated with DZNep (Fig. 6c). These results, together with our with improved overall survival and disease-free interval (Fig. 6f–i
observations that EZH2 inhibition reduces T cell polyfunctional- and Supplementary Table 1). Age, tumor stage and debulking and
ity, indicate that EZH2 controls polyfunctional T cell–mediated grade were important prognostic factors for overall and disease-free
antitumor immunity in vivo. survival (Supplementary Table 1). After adjusting for these clini-
The surface phenotype of tumor-infiltrating T cell subsets has been cal prognostic factors and histology, the number and percentage of
studied in human cancers29,30. Given the polyfunctional profile of EZH2+CD8+ T cells remained significant (P < 0.05) predicators of
EZH2+ T cells, we hypothesized that tumor-infiltrating EZH2+CD8+ overall and disease-free survival (Supplementary Tables 2–4).
T cells positively affect cancer survival. To test this hypothesis, we To estimate the accuracy of CD8+ T cell numbers, EZH2+CD8+
quantified the frequencies of EZH2+CD8+ T cells via immuno- T cell numbers and the percentages of EZH2+CD8+ T cells for predict-
histochemistry in tissue samples from 135 patients with ovarian ing survival, we used time-dependent receiver-operating-characteristic
cancer for whom clinical and pathological information was avail- (ROC) curve analysis31. We calculated the area under the ROC
able (Supplementary Table 1). On the basis of the median num- curve (AUC) to evaluate the predictive accuracy of each marker for
bers of CD8+ T cells and EZH2+CD8+ T cells and the percentage of estimating long-term (10-year) survival. The AUC for CD8+ T cells
EZH2+CD8+ T cells in total CD8+ T cells, we divided patients into was significantly lower than that for EZH2+CD8+ cells, expressed in
two groups, designated ‘low’ and ‘high’. Univariate analysis dem- both number and percentage (Fig. 6j,k). This finding indicates that
onstrated no significant correlations between the number of CD8+ expression of EZH2 in CD8+ T cells is a stronger predictor for long-
T cells and overall patient survival (Fig. 6d and Supplementary Table 1) term overall survival and disease-free interval than is CD8 expression
or between the number of CD8+ T cells and disease-free interval alone (Fig. 6j,k). Moreover, the percentage of EZH2+CD8+ T cells was

nature immunology  aDVANCE ONLINE PUBLICATION 


Articles

associated with a significantly higher AUC were the numbers of a metabolic target and mechanism of cancer immune evasion: cancer
CD8+ T cells or EZH2+CD8+ T cells, for both overall survival and restricts T cell EZH2 expression by limiting their aerobic glycolysis
disease-free interval (Fig. 6j,k). Similar results were obtained for and consequently weakens T cell–mediated antitumor immunity.
survival from 6–8 years. These data suggest that the percentage This study has notable clinical application. The ability of persistence
of EZH2+CD8+ T cells precisely predicts outcomes and long-term and survival is critical for tumor-associated antigen (TAA)-specific
survival for patients with ovarian cancer. Taken together, our results T cells to exert their antitumor effects 34. TH17 cells have survival
demonstrate how EZH2 marks polyfunctional T cells and controls advantages with stem cell properties19,36,38,40. In line with this, we
tumor immunity and affects patient outcome. have found that polyfunctional T cells express EZH2 and have sur-
vival advantages in the cancer microenvironment. It is logical that the
DISCUSSION fraction of EZH2+CD8+ T cells significantly and precisely predicts
Effector memory T cells, including polyfunctional T cells, mediate patient outcome. Our work indicates that polyfunctional CD8 +
potent protective immunity in viral infection1–5. Although T cell infil- T cells are a precise prognostic factor for long-term cancer survival.
tration of tumors is associated with improved patient survival15–17, Furthermore, EZH2+CD8+ T cells may be a valuable marker for test-
the nature of T cell effector functions, including survival and poly- ing and screening clinical efficacy of immune therapy and vaccination.
functionality, remains poorly understood. Furthermore, memory It is reasonable to assume that enhancing and/or maintaining EZH2
T cell subsets are under active investigation in the context of immune expression in T cells may be therapeutically meaningful in treating
vaccination, T cell therapy and inhibitory checkpoint blockade patients with cancer; however, EZH2 is highly expressed in various
therapy6,19,32–36. It has been proposed that memory T cell differentia- human cancer cells27,41. Systemic epigenetic therapy, including EZH2
tion status, survival and ‘stemness’ properties19,34–37 and the percent- blockade, has been considered a powerful regimen to treating patients
age of CD27+CD28+ T cells37 may be crucial factors in determining with cancer42,43. Given the current overwhelming interest in epige-
© 2015 Nature America, Inc. All rights reserved.

their antitumor potency19,34–36,38. However, it is not clear which T cell netic therapy, our data suggest that the potential effect of systemic
subset truly mediates antitumor immunity in patients with cancer. epigenetic manipulation on T cell effector function should be taken
Understanding the features of the protective T cells in cancer patients into consideration. T cells and tumor cells may be differentially sensi-
will provide significant guidance in designing T cell therapies and tive to epigenetic manipulation. A tumor-specific targeting approach
evaluating therapeutic efficacy of immune vaccination and therapy. or the use of minimal effective dosage would be a wise option in
Here we have phenotypically, functionally and clinically demonstrated clinical exploration of epigenetic therapy. Furthermore, the combina-
that EZH2+ T cells mediate potent antitumor immunity in human tion of epigenetic therapy and immunotherapy may be sequentially
cancer and are associated with long-term survival. used to enhance the therapeutic efficacy and minimize the potential
Epigenetic and chromatin remodeling may occur rapidly in acti- side effect of certain epigenetic modifier(s). In the context of autoim-
vated memory T cells9. We found that 48 h after TCR engagement, munity, T cells express high amounts of EZH2 in the microenvi-
substantial EZH2 expression was induced in T cells. EZH2 expres- ronment of ulcerative colitis, and it is assumed that inhibition of
sion highly coincides in polyfunctional memory T cells. Major tran- EZH2 expression or EZH2-associated signaling in T cells might be
scriptional factors, including T-bet, GATA-3 and RORγt control the therapeutically beneficial.
differentiation and function of TH1, TH2 and TH17 effector T cell In summary, we have demonstrated that EZH2+CD8+ T cells
subsets. It is unknown whether polyfunctional T cells are control- constitute a functional effector T cell subset in human cancer; that
led by a major transcription factor. Although EZH2 is not known EZH2 marks and controls effector T cell polyfunctionality and
as a transcription factor, we have shown that EZH2 is involved in survival through an miRNA-EZH2-Notch signaling pathway; and
defining the survival and polyfunctional cytokine profile of effector that this pathway is regulated by glucose metabolism in the tumor
T cells. To further study the mechanism by which EZH2 controls microenvironment. Manipulation of this pathway may be useful in
effector T cell survival and polyfunctionality, we have shown that treating patients with cancer.
EZH2 directly binds to the promoter areas of genes encoding Notch
repressors, NUMB and FBXW7, and represses their transcription Methods
via H3K27me3 and subsequently causes Notch activation, which Methods and any associated references are available in the online
results in antiapoptotic gene activation and effector cytokine expres- version of the paper.
sion. Thus, EZH2 targets the Notch signaling pathway to regulate
Note: Any Supplementary Information and Source Data files are available in the
effector T cell polyfunctionality and survival. online version of the paper.
Given the functional and mechanistic importance of EZH2 in poly-
functional T cells in cancer, it is essential to dissect how T cell EZH2 Acknowledgments
We thank D. Postiff, M. Vinco, R. Craig and J. Barikdar for tissue procurement
is controlled in the tumor microenvironment, a hypoglycemic envi- core at the University of Michigan; G. Lv, W. Dong and L. Li for assistance;
ronment owing to the Warburg effect. Notably, we have found that R. Zhang (Wistar Institute) for shEZH2 plasmids; P. King for discussions; and
primary ovarian cancer cells reduce the EZH2 expression and limit B. Leclair and D. Leclair for support. Supported by the US National Institutes of
polyfunctional cytokine expression and survival of effector T cells, Health (the Intramural Research Program; and CA123088, CA099985, CA156685,
CA171306 and 5P30CA46592), the Chinese Ministry of Science and Technology
which can subsequently be rescued by glucose supplementation and
(973 program, 2015CB554000), the Wuhan Union Hospital Research Fund,
mimicked by 2-DG treatment. This observation suggests that T cell the Ovarian Cancer Research Fund, and Marsha Rivkin Center for Ovarian
EZH2 is controlled by glycolytic metabolism in the cancer environ- Cancer Research.
ment. In line with our observations, studies have demonstrated that
glycolytic switch regulates memory T cell activation, proliferation and AUTHOR CONTRIBUTIONS
IFN-γ expression in mouse models9,10,39. Further mechanistic studies E.Z., T.M. and I.K. designed and performed most of the experiments, interpreted
the data and drafted the paper; W.L., K.W., L.Z., S. Wei, J.C., S. Wan, L.V., W.S.
have shown that tumor cells sustain high expression of miR-26a and and I.S. performed experiments; Y.W., Y.L., S.V., A.M.C., T.H.W., V.E.M., J.K.,
miR-101 in effector T cells. Consequently, these miRNAs lessen EZH2 H.W., Y.Z., Z.W., R.L. and G.W. provided reagents or clinical specimens and
expression and cause effector T cell dysfunction. Thus, our data reveal clinopathological information and interpreted the data. W.Z., I.K. and G.W.

 aDVANCE ONLINE PUBLICATION  nature immunology


Articles

supported, conceived of and supervised the research, designed experiments 21. McCabe, M.T. et al. EZH2 inhibition as a therapeutic strategy for lymphoma with
and wrote the paper. EZH2-activating mutations. Nature 492, 108–112 (2012).
22. Ciofani, M. & Zuniga–Pflucker, J.C. Notch promotes survival of pre–T cells at the
COMPETING FINANCIAL INTERESTS beta–selection checkpoint by regulating cellular metabolism. Nat. Immunol. 6,
The authors declare no competing financial interests. 881–888 (2005).
23. Amsen, D. et al. Instruction of distinct CD4 T helper cell fates by different notch
Reprints and permissions information is available online at http://www.nature.com/ ligands on antigen-presenting cells. Cell 117, 515–526 (2004).
24. Wirtz–Peitz, F., Nishimura, T. & Knoblich, J.A. Linking cell cycle to asymmetric
reprints/index.html.
division: Aurora–A phosphorylates the Par complex to regulate Numb localization.
1. Ladell, K. et al. A molecular basis for the control of preimmune escape variants Cell 135, 161–173 (2008).
by HIV–specific CD8+ T cells. Immunity 38, 425–436 (2013). 25. Oberg, C. et al. The Notch intracellular domain is ubiquitinated and negatively
2. Almeida, J.R. et al. Superior control of HIV–1 replication by CD8+ T cells is reflected regulated by the mammalian Sel-10 homolog. J. Biol. Chem. 276, 35847–35853
by their avidity, polyfunctionality, and clonal turnover. J. Exp. Med. 204, (2001).
2473–2485 (2007). 26. Cui, T.X. et al. Myeloid-derived suppressor cells enhance stemness of cancer cells
3. Harari, A. et al. An HIV–1 clade C DNA prime, NYVAC boost vaccine regimen by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity 39,
induces reliable, polyfunctional, and long-lasting T cell responses. J. Exp. Med. 611–621 (2013).
205, 63–77 (2008). 27. Varambally, S. et al. Genomic loss of microRNA-101 leads to overexpression of
4. Gaucher, D. et al. Yellow fever vaccine induces integrated multilineage and histone methyltransferase EZH2 in cancer. Science 322, 1695–1699 (2008).
polyfunctional immune responses. J. Exp. Med. 205, 3119–3131 (2008). 28. Sander, S. et al. MYC stimulates EZH2 expression by repression of its negative
5. Precopio, M.L. et al. Immunization with vaccinia virus induces polyfunctional and regulator miR-26a. Blood 112, 4202–4212 (2008).
phenotypically distinctive CD8+ T cell responses. J. Exp. Med. 204, 1405–1416 29. Curiel, T.J. et al. Specific recruitment of regulatory T cells in ovarian carcinoma
(2007). fosters immune privilege and predicts reduced survival. Nat. Med. 10, 942–949
6. Wherry, E.J. T cell exhaustion. Nat. Immunol. 12, 492–499 (2011). (2004).
7. Zheng, Y., Zha, Y., Driessens, G., Locke, F. & Gajewski, T.F. Transcriptional regulator 30. Galon, J. et al. Type, density, and location of immune cells within human colorectal
early growth response gene 2 (Egr2) is required for T cell anergy in vitro and tumors predict clinical outcome. Science 313, 1960–1964 (2006).
in vivo. J. Exp. Med. 209, 2157–2163 (2012). 31. Heagerty, P.J., Lumley, T. & Pepe, M.S. Time-dependent ROC curves for censored
8. Crespo, J., Sun, H., Welling, T.H., Tian, Z. & Zou, W. T cell anergy, exhaustion, survival data and a diagnostic marker. Biometrics 56, 337–344 (2000).
© 2015 Nature America, Inc. All rights reserved.

senescence, and stemness in the tumor microenvironment. Curr. Opin. Immunol. 32. Scholler, J. et al. Decade-long safety and function of retroviral-modified chimeric
25, 214–221 (2013). antigen receptor T cells. Sci. Transl. Med. 4, 132ra153 (2012).
9. Gubser, P.M. et al. Rapid effector function of memory CD8+ T cells requires an 33. Ahmed, R., Bevan, M.J., Reiner, S.L. & Fearon, D.T. The precursors of memory:
immediate-early glycolytic switch. Nat. Immunol. 14, 1064–1072 (2013). models and controversies. Nat. Rev. Immunol. 9, 662–668 (2009).
10. Chang, C.H. et al. Posttranscriptional control of T cell effector function by aerobic 34. Rosenberg, S.A., Restifo, N.P., Yang, J.C., Morgan, R.A. & Dudley, M.E. Adoptive
glycolysis. Cell 153, 1239–1251 (2013). cell transfer: a clinical path to effective cancer immunotherapy. Nat. Rev. Cancer
11. Viré, E. et al. The Polycomb group protein EZH2 directly controls DNA methylation. 8, 299–308 (2008).
Nature 439, 871–874 (2006). 35. Gattinoni, L. et al. A human memory T cell subset with stem cell–like properties.
12. Cao, R. et al. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Nat. Med. 17, 1290–1297 (2011).
Science 298, 1039–1043 (2002). 36. Zou, W. & Restifo, N.P. TH17 cells in tumour immunity and immunotherapy.
13. Tumes, D.J. et al. The polycomb protein Ezh2 regulates differentiation and plasticity Nat. Rev. Immunol. 10, 248–256 (2010).
of CD4+ T helper type 1 and type 2 cells. Immunity 39, 819–832 (2013). 37. Powell, D.J. Jr., Dudley, M.E., Robbins, P.F. & Rosenberg, S.A. Transition of late-
14. Tong, Q. et al. Ezh2 regulates transcriptional and posttranslational expression of stage effector T cells to CD27+CD28+ tumor-reactive effector memory T cells in
T-bet and promotes Th1 cell responses mediating aplastic anemia in mice. humans after adoptive cell transfer therapy. Blood 105, 241–250 (2005).
J. Immunol. 192, 5012–5022 (2014). 38. Wei, S., Zhao, E., Kryczek, I. & Zou, W. Th17 cells have stem cell-like features
15. Pagès, F. et al. Effector memory T cells, early metastasis, and survival in colorectal and promote long-term tumor immunity. OncoImmunology 1, 516–519 (2012).
cancer. N. Engl. J. Med. 353, 2654–2666 (2005). 39. Yang, K. et al. T cell exit from quiescence and differentiation into Th2 cells depend
16. Zhang, L. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian on Raptor-mTORC1-mediated metabolic reprogramming. Immunity 39, 1043–1056
cancer. N. Engl. J. Med. 348, 203–213 (2003). (2013).
17. Kryczek, I. et al. Phenotype, distribution, generation, and functional and clinical 40. Muranski, P. et al. Th17 cells are long lived and retain a stem cell–like molecular
relevance of Th17 cells in the human tumor environments. Blood 114, 1141–1149 signature. Immunity 35, 972–985 (2011).
(2009). 41. Varambally, S. et al. The Polycomb group protein EZH2 is involved in progression
18. Marrack, P. & Kappler, J. Control of T cell viability. Annu. Rev. Immunol. 22, of prostate cancer. Nature 419, 624–629 (2002).
765–787 (2004). 42. Tan, J. et al. Pharmacologic disruption of Polycomb-repressive complex 2–mediated
19. Kryczek, I. et al. Human TH17 cells are long–lived effector memory cells. gene repression selectively induces apoptosis in cancer cells. Genes Dev. 21,
Sci. Transl. Med. 3, 104ra100 (2011). 1050–1063 (2007).
20. Miranda, T.B. et al. DZNep is a global histone methylation inhibitor that reactivates 43. Fiskus, W. et al. Combined epigenetic therapy with the histone methyltransferase
developmental genes not silenced by DNA methylation. Mol. Cancer Ther. 8, EZH2 inhibitor 3–deazaneplanocin A and the histone deacetylase inhibitor
1579–1588 (2009). panobinostat against human AML cells. Blood 114, 2733–2743 (2009).

nature immunology  aDVANCE ONLINE PUBLICATION 


ONLINE METHODS Cell apoptosis detection. Cell apoptosis was measured with an annexin V
Ovarian cancer patients and human tissues. Patients diagnosed with ovarian apoptosis detection kit (BD Bioscience). 2–5 µM DZNep and 10 µM GSK126
cancer were recruited for this study. We collected 135 formalin-fixed, paraffin- (Glixx Laboratories) were added into the cell culture as indicated.
embedded ovarian tumor tissue blocks, 20 snap-frozen ovarian tumor
tissues, and 37 fresh ovarian cancer tissues and 11 ovary cysts for this study, Metabolic assay. T cell metabolism was measured in a Seahorse XFe24 analyzer
as described29,44–46. After pathological review, a tissue microarray (TMA) was on V7-PET XF24 cell culture microplates (Seahorse Bioscience). 1 d before
constructed from the most representative area of paraffin-embedded ovar- the assay, the plate was coated with Cell-Tak reagent (BD Biosciences) accord-
ian tumor tissue. For each tumor, a minimum of two representative tumor ingly to the manufacturer’s instruction. Next, 5 × 105 T cells were seeded into
areas were selected from a hematoxylin- and eosin-stained section of a donor the wells and gently centrifuged twice for uniform distribution in the well.
block. Core cylinders (1 mm) were punched from each of these areas and After 30 min of incubation, the assay medium was added to the cells and
deposited into a recipient paraffin block. Consecutive 6 µm-thick TMA the experiment was performed. Normal medium was RPMI 1640 contain-
sections were cut and placed on charged poly-l-lysine-coated slides for immu- ing 2 mg/ml glucose without sodium bicarbonate and FCS adjusted to pH
nohistochemistry analyses. All donors provided written, informed consent. In 7.4. Normal medium and ovarian cancer culture medium were used in the
addition to ovarian cancer tissues, human spleen, tonsil, and ulcerative colitic assay. Different metabolic modifiers were added. Before seeding on the test
colon tissues were obtained from the tissue procurement core at the University plate, the cells were washed three times in assay medium to remove buffering
of Michigan Hospital. The study was approved by the Institutional Review compounds. T cells were stimulated with 2.5 µg/ml anti-CD3 (clone UCHT1)
Boards of the University of Michigan School of Medicine, Tongji Medical and 1.25 µg/ml anti-CD28 (clone CD28.2) with or without 2-deoxy-d-glucose
College and Medical University of Lublin. (2-DG, 50 mg/ml) (Sigma-Aldrich).

Animal studies. For the B16 melanoma lung metastasis model, we intra- T cell culture. Human peripheral blood T cell subsets (106 per milliliter) were
venously injected B16 melanoma cells into C57BL/6 mice and established activated with anti-CD3 (clone UCHT1, 2.5 µg/ml) and anti-CD28 (clone
lung metastasis. Melanoma-specific T cells were generated as described 44. CD28.2, 1.25 µg/ml) (BD Bioscience) for 3–5 d. To obtain tumor medium,
© 2015 Nature America, Inc. All rights reserved.

Melanoma-specific T cells were activated in the presence or absence of 1 µM primary ovarian cancer cells were cultured for 3 d, and the cells and culture
DZNep for 2 d or transfected with lentivirus vectors encoding shEZH2 or medium were subsequently frozen and thawed 5 times. The cancer cell cul-
scramble and activated for 2 d, then washed and expanded in 80 IU/ml IL-2 for ture supernatant was obtained through centrifugation (20,000 × g, 1 h, 4 °C),
3 d and intravenously transfused into mice bearing melanoma lung metastasis. and used to culture T cells with or without glucose (2 mg/ml, Sigma-Aldrich)
After 7 d, the mice were killed. Lung metastatic foci were recorded. supplementation for 16–24 h. To obtain normal medium containing differ-
ent concentrations of glucose, normal medium without glucose (Invitrogen)
Human ovarian cancer chimeric model. Primary ovarian cancer cells were iso- was supplemented with 1 or 5 mg/ml glucose. Chemical inhibitors of
lated from ovarian cancer ascites fluid and ovarian cancer tissues44,47. Ovarian metabolic pathways, Notch signaling or EZH2 were added as indicated in
cancers were subcutaneously established in female NOD/scid/IL-2Rγnull figure legends. Total numbers of viable cells at the endpoint of experiments
(NSG) mice (~6–8 weeks old; Jackson Laboratory)44,47. Autologous tumor- were 1–2 × 106 cells.
specific T cells were generated as described19. Tumor-specific T cells were
treated with 1 µM DZNep for 3 d, washed and intravenously transfused into Immunoblot analysis. Immunoblotting was performed using primary anti-
mice bearing autologous ovarian cancer. Tumor volume was measured every bodies to the following: EZH2 (1:1,000, 612667, BD Bioscience), H3K27me3
4 d. All animal studies were conducted under the approval of the University (1:1,000, 07-449, Millipore), H3 (1:2,000, 4499, Cell Signaling), NICD (1:1,000,
of Michigan Committee on Use and Care of Animals. ab52301, Abcam), Bcl-2 (1:1,000, 551107, BD Bioscience), and β-actin (1:5,000,
A5441, Sigma). Signals were detected by ECL reagent (GE Healthcare).
Manipulation of EZH2 and Notch signaling pathways. Short hairpin RNA
with an EZH2-specific (shEZH2) or scramble sequence was packed into a len- Quantitative real-time PCR. Total RNA was isolated with TRIzol Reagent
tivirus packaging construct and transfected into HEK293T cells (ATCC) with (Invitrogen). Quantitative real-time PCR was carried out to quantify
Lipofectamine 2000 (Invitrogen). EZH2 expression was genetically blocked genes associated with cell cycling, Notch signaling, and apoptosis 19
in human primary memory T cells with lentivirus encoding shEZH2 or (Supplementary Table 5).
scramble. 2–5 µM 3-deazaneplanocin A HCl (DZNep) and 10 µM GSK126
(Glixx Laboratories) were used to biochemically block EZH2 and EZH2 meth- MicroRNA detection and manipulation. MiRNAs were analyzed with miRNA
yltransferase activity, respectively. The vectors encoding an active domain of TaqMan assays (Life Technologies) accordingly to the manufacturer’s instruc-
Notch (Notch-IC) or a negative domain were used to genetically activate or tion. To examine the roles of miRNAs, we transfected T cells with specific
block Notch signaling, respectively19,48. The Notch signaling pathway was miRNA mimics or irrelevant oligonucleotide controls on day 2 using a human
biochemically manipulated with two Notch inhibitors: DAPT (Santa Cruz Nucleofector Kit (Lonza). The cells were analyzed on day 3.
Biotech) and GSI-I (Calbiochem). HEK293T cells were routinely tested for
mycoplasma contamination. EZH2 overexpression. T cells were transfected with EZH2-pCMV plasmid27,41
before activation by human Nucleofector Kit (Lonza).
Fluorescence-activated cell sorting and flow cytometry analysis. Cells were
first stained extracellularly with specific antibodies to human CD3 (clone BCL2 promoter assay. The human BCL2 promoter sequence (−850 to +134,
HIT3a), CD4 (clone RPA-T4), CD8 (clone HIT8a), CD45RA (clone HI100), transcription start site as +1) was initially linked to GFP. HEK293T cells were
CD45RO (clone UCHL1), CD57 (clone NK-1), CD62L (clone DREG56) transfected with viral vector encoding the human BCL2 promoter in con-
(BD Bioscience), KLRG-1 (clone 13F12F2, eBioscience) or Tim-3 (clone junction with vector controls or vectors containing cDNA encoding shEZH2,
#344823) (R&D), fixed and permeabilized with Fixation/Permeabilization Notch-IC, and Notch-DN48. The promoter activity was measured by the
solution (eBioscience), and stained intracellularly with antibodies to green fluorescence intensity of transfected cells. The intensity of control-GFP
tumor necrosis factor (TNF, clone Mab11), IFN-γ (clone B27), granzyme reporter is defined as 100.
B (clone GB11), Bcl-2 (clone Bcl-2/100), or EZH2 (clone 11/EZH2) (BD
Biosciences). Samples were acquired on a flow cytometry analyzer (LSR II; BD Chromatin immunoprecipitation (ChIP). ChIP was performed as
Biosciences) and data were analyzed with DIVA software (BD Biosciences). described26,49. Briefly, nuclear extracts were prepared from primary human
For lentivirus-infected cells, GFP+ cells were sorted with a FACSAria cell cells. Mouse anti-human H3K27me3 (ab6002, Abcam) and normal IgG
sorter (BD Biosciences). (sc-2027, Santa Cruz) antibodies were used for immunoprecipitation. ChIP

nature immunology doi:10.1038/ni.3313


primers (Supplementary Table 5) were designed to detect promoter fragments Pearson correlation (r) for continuous data) and P values (null hypothesis is
near transcription start sites on NUMB and FBXW7 genes. that r is in fact 0) were computed to measure the degree of association between
biomarkers. Overall patient survival was defined as time from date of diagnosis
Immunofluorescence and immunohistochemistry staining. We performed to disease-related death. Disease-free interval was defined as time from date
immunofluorescence staining in frozen tissue sections. The primary anti- of diagnosis to disease progression or disease-related death. Data were cen-
bodies were mouse anti-human EZH2 (1:100, 612667, IgG1, BD Bioscience), sored at the last follow up for patients who were disease free or alive at the
rabbit anti-human CD3 (1:100, A0452, IgG, Dako), and TUNEL (S7165, time of analysis. Survival functions were estimated by Kaplan-Meier methods.
Millipore). The secondary antibodies were goat anti-mouse IgG conjugated Cox’s proportional hazards regression was performed to model survival as a
with Alexa Fluor 488 (1:2,000, A11001, Life Technologies) and goat anti-rabbit function of the levels of CD8+ T cells, EZH2+CD8+ T cells or the percentage
IgG conjugated with Alexa Fluor 568 (1:2,000, A11036, Life Technologies). of EZH2+CD8+ T cells in CD8+ T cells (all classified as low and high on the
The stained tissue sections were analyzed under a fluorescent microscope. basis of the median value), after adjusting for age, stage, histology, and debulk-
Immunohistochemistry staining was performed in paraffin-fixed ovarian can- ing. We assessed the adequacy of the Cox regression model by graphical and
cer tissues (Dako EnVision Double stain System, Dako). Paraffin-embedded numerical methods as described26,49,50. To estimate the performance of the
TMA sections were deparaffinized and stained with rabbit anti-human EZH2 levels of CD8+ T cells, EZH2+CD8+ T cells and the percentage of EZH2+CD8+
(1:200, 187395, Invitrogen) followed by Polymer-HRP and DAB Chromogen T cells for predicting patient survival, we used the time-dependent receiver-
treatment. TMA sections were subsequently stained with mouse anti-human operating-characteristic (ROC) curve analysis 31. The area under the ROC
CD8 (1:50, M7103, Dako), followed by treatment with rabbit-mouse LINK, curve (AUC) was calculated to evaluate each marker’s predictive accuracy.
polymer-AP, permanent red chromogen and hematoxylin counterstaining. We used this analysis to evaluate and compare the predictive accuracy of the
The TMA cores were quantified and analyzed for expression of EZH2 + and levels of CD8+ T cells, EZH2+CD8+ T cells, and the percentage of EZH2+CD8+
CD8+ T cells with an Aperio imaging system (Genetix). The specimens were T cells for long-term survivals. All analyses were done using SAS 9.3 software.
digitalized with an automated platform (Aperio Technologies) and ScanScope P < 0.05 was considered as significant.
XT and Spectrum Plus using TMA software version 9.1 scanning system. Cores
© 2015 Nature America, Inc. All rights reserved.

were scored in high resolution at 40× magnification. Tissue sections were


scored manually on a computer screen, and a mean score for duplicate cores 44. Kryczek, I. et al. B7–H4 expression identifies a novel suppressive macrophage
from each individual was calculated. Any discrepancies were resolved by sub- population in human ovarian carcinoma. J. Exp. Med. 203, 871–881 (2006).
45. Curiel, T.J. et al. Blockade of B7–H1 improves myeloid dendritic cell–mediated
sequent consultation with diagnostic pathologists. CD8 was localized in the antitumor immunity. Nat. Med. 9, 562–567 (2003).
cell membrane and cytoplasm and EZH2 was localized in the nuclei. CD8 and 46. Zou, W. et al. Stromal–derived factor–1 in human tumors recruits and alters the
EZH2 were scored quantitatively (the numbers of positive cells). The tissues function of plasmacytoid precursor dendritic cells. Nat. Med. 7, 1339–1346
were divided into high and low CD8+ T cells (>7, <7) and EZH2+CD8+ T cells (2001).
47. Kryczek, I. et al. Expression of aldehyde dehydrogenase and CD133 defines ovarian
(>2, <2), or by the percentage (>22%, <22%) of EZH2+CD8+ T cells in cancer stem cells. Int. J. Cancer 130, 29–39 (2012).
CD8+ T cells on the basis of their median values per core (Supplementary 48. Yamamoto, N. et al. Role of Deltex-1 as a transcriptional regulator downstream of
Tables 1–4). Staining with isotype antibody was used as negative control. the Notch receptor. J. Biol. Chem. 276, 45031–45040 (2001).
49. Kryczek, I. et al. IL–22CD4 T cells promote colorectal cancer stemness via STAT3
transcription factor activation and induction of the methyltransferase DOT1L.
Statistical analysis. Wilcoxon rank-sum tests were used to compare two Immunity 40, 772–784 (2014).
independent groups, and for paired groups, Wilcoxon signed rank tests were 50. Lin, D.Y., Wei, L.J. & Ying, Z. Checking the Cox model with cumulative sums of
used. Correlation coefficients (Spearman correlation (r) for ordinal data and martingale-based residuals. Biometrika 80, 557–572 (1993).

doi:10.1038/ni.3313
 nature immunology

You might also like