Vous êtes sur la page 1sur 11

Cancer Cell

Article

Direct Reversal of Glucocorticoid Resistance


by AKT Inhibition in Acute Lymphoblastic Leukemia
Erich Piovan,1,2,3,21 Jiyang Yu,4,5,17,21 Valeria Tosello,1,6 Daniel Herranz,1 Alberto Ambesi-Impiombato,1
Ana Carolina Da Silva,1 Marta Sanchez-Martin,1 Arianne Perez-Garcia,1 Isaura Rigo,1 Mireia Castillo,7
Stefano Indraccolo,2 Justin R. Cross,8 Elisa de Stanchina,9 Elisabeth Paietta,10,11 Janis Racevskis,10,11 Jacob M. Rowe,12
Martin S. Tallman,13 Giuseppe Basso,14 Jules P. Meijerink,15 Carlos Cordon-Cardo,7 Andrea Califano,1,4,5,16,17,*
and Adolfo A. Ferrando1,7,18,19,20,*
1Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
2UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto—IRCCS, Padova 35128, Italy
3Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Universita’ di Padova, Padova, Veneto 35128, Italy
4Department of Biomedical Informatics, Columbia University, New York, NY 10032, USA
5Department of Systems Biology, Columbia University, New York, NY 10032, USA
6Istituto Oncologico Veneto, IRCCS, Padova, Veneto 35128, Italy
7Department of Pathology, Mount Sinai School of Medicine, New York, NY 10029 USA
8Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 USA
9Antitumor Assessment Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
10Department of Medicine, Albert Einstein School of Medicine, Bronx, NY 10461, USA
11Department of Oncology, Montefiore Medical Center-North Division, Bronx, NY 10466, USA
12Hematology Department, Shaare Zedek Hospital, Jerusalem 91031, Israel
13Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
14Dipartimento di Salute della Donna e del Bambino, Università di Padova, via Giustiniani 3, Padova 35128, Italy
15Department of Pediatric Oncology/Hematology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, South Holland 010 7040704,

the Netherlands
16Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
17Cancer Regulatory Network Program, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
18Lymphoid Malignancy and Development Program, Herbert Irving Comprehensive Cancer Center, Columbia University, New York,

NY 10032, USA
19Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
20Department of Pathology, Columbia University Medical Center, New York, NY 10032, USA
21These authors contributed equally to this work

*Correspondence: ac2248@columbia.edu (A.C.), af2196@columbia.edu (A.A.F.)


http://dx.doi.org/10.1016/j.ccr.2013.10.022

SUMMARY

Glucocorticoid resistance is a major driver of therapeutic failure in T cell acute lymphoblastic leukemia
(T-ALL). Here, we identify the AKT1 kinase as a major negative regulator of the NR3C1 glucocorticoid receptor
protein activity driving glucocorticoid resistance in T-ALL. Mechanistically, AKT1 impairs glucocorticoid-
induced gene expression by direct phosphorylation of NR3C1 at position S134 and blocking gluco-
corticoid-induced NR3C1 translocation to the nucleus. Moreover, we demonstrate that loss of PTEN and
consequent AKT1 activation can effectively block glucocorticoid-induced apoptosis and induce resistance
to glucocorticoid therapy. Conversely, pharmacologic inhibition of AKT with MK2206 effectively restores
glucocorticoid-induced NR3C1 translocation to the nucleus, increases the response of T-ALL cells to gluco-
corticoid therapy, and effectively reverses glucocorticoid resistance in vitro and in vivo.

Significance

Improving the outcomes of patients with acute lymphoblastic leukemia (ALL) will require the identification of specific
molecular mechanisms driving resistance to therapy and developing new pharmacologic strategies to neutralize them.
Primary resistance to glucocorticoids, defined as failure to achieve robust cytoreduction after a week of glucocorticoid
therapy, is of particular importance as it is associated with very poor prognosis. Here, we show that AKT1 can drive
resistance to glucocorticoids via direct inhibition of the glucocorticoid receptor. Consistently, pharmacologic inhibition
of AKT effectively reverses glucocorticoid resistance. These results highlight the potential role of AKT1 as therapeutic
target in the treatment of glucocorticoid-resistant T cell ALL.

766 Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc.
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

INTRODUCTION HA-NR3C1 immunoprecipitates, suggesting that AKT1 can


interact with NR3C1 in vivo (Figure 1A). Reciprocal immuno-
Glucocorticoids are small lipophilic compounds derived from precipitation experiments confirmed the association between
cortisol, a natural adrenal hormone that signals via the glucocor- Flag-AKT1 and HA-NR3C1 (Figure 1B). Moreover, immuno-
ticoid receptor alpha, a nuclear receptor ligand-activated tran- precipitation of NR3C1 protein complexes from the T-ALL cell
scription factor encoded by the NR3C1 gene (Schlossmacher lines DND41 and CCRF-CEM demonstrated that endogenous
et al., 2011). Glucocorticoids play a fundamental role in the NR3C1 and AKT1 can interact in T-ALL lymphoblast cells (Fig-
treatment of all lymphoid tumors because of their capacity to ure 1C and Figure S1A available online). In addition, immunoflu-
induce apoptosis in lymphoid progenitor cells (Inaba and Pui, orescence analysis showed colocalization of NR3C1 and AKT1
2010; Pui et al., 2011, 2012). The importance of glucocorticoid in DND41 and CCRF-CEM cells (Figures 1D and S1B). Finally,
therapy in lymphoid malignancies is underscored by the strong glutathione S-transferase (GST)-pulldown assays showed that
association of primary glucocorticoid resistance with poor recombinant GST-NR3C1 fusion protein can directly interact
prognosis in childhood acute lymphoblastic leukemia (ALL). with His-tagged AKT1 in vitro (Figure 1E).
Thus, prednisone poor response, defined as failure to show
effective cytoreduction after 7 days of glucocorticoid therapy, AKT1 Phosphorylates S134 in the NR3C1 Protein
is strongly associated with increased risk of relapse and thera- AKT1 kinase substrates are typically phosphorylated by AKT at
peutic failure (Dördelmann et al., 1999; Inaba and Pui, 2010), RXRXXS/T motifs (Mok et al., 1999; Ozes et al., 1999; Zimmer-
and in vitro resistance to glucocorticoids is associated with mann and Moelling, 1999). Phospho-AKT motif scanning anal-
unfavorable prognosis in this disease (Hongo et al., 1997; Inaba ysis of NR3C1 revealed a potential AKT phosphorylation motif
and Pui, 2010; Klumper et al., 1995). 131RSTS134 (Figures 1F and S1C), suggesting that the gluco-
Primary glucocorticoid resistance is particularly frequent in corticoid receptor could be an AKT1 substrate phosphorylated
T cell ALL (T-ALL) (Hongo et al., 1997; Inaba and Pui, 2010; at serine 134. To test this possibility, we expressed HA-tagged
Klumper et al., 1995), leading us to hypothesize that activation wild-type NR3C1 (HA-NR3C1) or an HA-tagged form of the
of one or more oncogenic signaling pathways implicated in glucocorticoid receptor with a serine-to-alanine substitution
T cell transformation could be driving primary glucocorticoid at position 134 (HA-NR3C1 S134A) in cells infected with retro-
resistance in T-ALL directly by interfering with glucocorticoid viruses expressing MYR-AKT1. Protein immunoprecipitation of
receptor function or indirectly via inhibition of glucocorticoid- NR3C1 with an antibody against HA and subsequent western
induced apoptosis. In this context, AKT1 emerged as a plausible blot analysis with an antibody recognizing the phospho-RXXS/
candidate as PI3K-AKT activation plays a major role in the T AKT phosphorylation motif showed the presence of an HA-
pathogenesis of T-ALL, particularly in leukemias harboring NR3C1 phospho-AKT band in cells expressing the wild-type
mutations and deletions in the PTEN tumor suppressor gene glucocorticoid receptor, but not in cells expressing the HA-
(Palomero et al., 2007). Moreover, in silico analysis of signaling NR3C1 S134A mutant (Figure 1G). Consistently, in vitro kinase
factors modulating transcriptional signatures associated with assays in which we analyzed the capacity of the AKT1 kinase
glucocorticoid resistance in T-ALL pointed to a potential role of to phosphorylate the wild-type or S134A glucocorticoid
AKT1 as driver of glucocorticoid resistance in T-ALL. These receptor proteins demonstrated that AKT1 can effectively
results, together with the association of mutational loss of phosphorylate recombinant wild-type NR3C1 protein in vitro,
PTEN and increased AKT1 phosphorylation with primary but not the serine 134 to alanine NR3C1 mutant protein (Fig-
glucocorticoid resistance in the clinic (Bandapalli et al., 2013; ure 1H). Mass spectrometry analysis of HA-NR3C1 protein
Morishita et al., 2012) and the availability of PI3K-AKT specific isolated from MYR-AKT1-expressing cells demonstrated the
inhibitors in clinical trials for the treatment of human cancer, presence of serine phosphorylation at position 134 of the gluco-
prompted us to analyze the mechanistic role of AKT1 in the corticoid receptor (Figures 1I and 1J), which was effectively
control of glucocorticoid resistance in T-ALL. abrogated (control S134P:non-P = 0.47; MK2206 S134P:non-
P = 0.03; Table S1) upon treatment with MK2206, a highly potent
RESULTS and selective inhibitor of AKT (Hirai et al., 2010). Finally, and most
notably, western blot analysis with the AKT phosphorylation
AKT1 Binds to the NR3C1 Glucocorticoid motif antibody showed decreased AKT phosphorylated NR3C1
Receptor Protein in NR3C1 immunoprecipitates from CCRF-CEM T-ALL cells
Activation of gene expression by glucocorticoids is a multistep upon AKT inhibition with MK2206 (Figure 1K).
process that requires effective release of the glucocorticoid
receptor from heat shock protein complexes, translocation to AKT Signaling Inhibits NR3C1 Nuclear Translocation
the nucleus, and formation of a multiprotein transcriptional following Glucocorticoid Treatment
complex on the promoter of glucocorticoid target genes After establishing the interaction and phosphorylation of the
(Heitzer et al., 2007). To test if AKT1 can interact with and glucocorticoid receptor by AKT1, we sought to elucidate the
inhibit the glucocorticoid receptor protein, we transfected relevance of the NR3C1 S134 phosphorylation for glucocorticoid
293T cells with plasmid constructs driving the expression of receptor function. Glucocorticoid-induced cytoplasmic-nuclear
Flag-tagged AKT1 and hemagglutinin (HA)-tagged NR3C1 and shuttling is strictly required for glucocorticoid receptor activity,
isolated glucocorticoid receptor-containing protein complexes which functions as a ligand-activated transcription factor
via immunoprecipitation using an anti-HA antibody. Western (Heitzer et al., 2007). U2OS cells, which express undetectable
blot analysis demonstrated the presence of Flag-AKT1 in levels of endogenous NR3C1 (Figure S2A), showed cytoplasmic

Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc. 767
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

Input Figure 1. AKT1 Interacts with the Gluco-


A (10%) anti HA IP B C
corticoid Receptor Protein and Regulates
Flag-AKT1 + + + Input IP

)
%
(20%) anti Flag IP NR3C1 S134 Phosphorylation

t(
HA-NR3C1 + + +

pu

C1
an IgG
HA-NR3C1 + + (A) Western blot analysis of AKT1 and activated

In

R3
Flag

e
10 5

ti N
us
Flag-AKT1 + + + + AKT1 after NR3C1 immunoprecipitation in 293T

mo
AKT1 cells expressing Flag-tagged AKT1 and HA-tag-
NR3C1 AKT1
ged NR3C1.
pAKT(S473)
AKT1 NR3C1 (B) NR3C1 western blot analysis after AKT1
HA immunoprecipitation in 293T cells expressing
Flag-tagged AKT1 and HA-tagged NR3C1.
D E F (C) Western blot analysis of AKT1 after NR3C1 pro-
GST 129 137
pulldown tein immunoprecipitation in DND-41 T-ALL cells.

1
AKT NR3C1 Merged Human NR3C1 LNRSTSVPE

3C
(D) Immunofluorescence colocalization analysis

R
146 155

-N
Input Mouse NR3C1 LNRSTSRPE
ST
ST
(20%) G of AKT (green) and NR3C1 (red) proteins in
G
138 146
Rat NR3C1 LNRSTSVPE DND41 cells. Cell nuclei stained with TO-PRO3
His-AKT1 138
130 are shown in blue. Scale bar, 10 mm.
Pig NR3C1 LSRSTSVPE
(E) Analysis of AKT1-NR3C1 interaction via west-
ern blot analysis of protein complexes recovered
G Input (15%) anti HA IP H after NR3C1-GST pulldown of recombinant His-
GST-NR3C1 + tagged AKT1.
HA-NR3C1 + + + + GST-NR3C1 S134A + (F) Partial alignment of the glucocorticoid receptor
HA-NR3C1 S134A + + + +
His- activated AKT1 + + +
MYR-AKT1 + + + + protein sequences flanking S134.
GFP + + + + (G) Western blot analysis of NR3C1 phospho-
GST-NR3C1
Coomassie P

Anti AKT Anti AKT rylation using an antibody against the AKT phos-
32

phospho phospho His-AKT1


motif motif pho-motif in NR3C1 protein immunoprecipitates
NR3C1 NR3C1 GST-NR3C1 from U2OS cells expressing MYR-AKT1 together
GST-NR3C1 S134A
pAKT1 pAKT1 His-AKT1 with HA-NR3C1 or HA-NR3C1 S134A.
(T308) (T308)
(H) In vitro kinase analysis of AKT1 phospho-
rylation of recombinant NR3C1 (GST-NR3C1) and
I J b2 b3 b4 NR3C1 S134A mutant (GST-NR3C1 S134A) pro-
S T pS V P E N P K m/z 519.72 teins. The top shows P-32 autoradiography after
y7 H2PO3 752.39

mass 1037.42
y5 584.30

y7 y6 y5 y4 y3 y2 y1
100 SDS-PAGE, and corresponding protein loading is
100 519.72 1:TOF MS ES+246
b4 357.17
Intensity (%)

shown at the bottom.


b3 258.11

520.23
MH 1038.45
y2 224.16

(I) Electrospray ionization MS/MS spectrum of


y6 683.37

y7 850.37
b2 189.09

y3 358.22

y4 487.25
y1 117.11
%

517.24
520.73 monophosphorylated peptide STpS134VPENPK
519.57 521.20
515.24 518.22 522.25 524.26 (S132 to K140) obtained after trypsin digestion of
0 m/z 0 mass NR3C1 isolated from cells expressing constitu-
515 516 517 518 519 520 521 522 523 524 100 200 300 400 500 600 700 800 900 1000 1100
tively active AKT1. TOF, time-of-flight.
(J) Collision-induced dissociation of the molecular
K ion, [M+2H]2+ at a mass-to-charge ratio (m/z) of
anti HA IP 120 519.72 (mass = 1,037.42 Da) corresponding to S134.
Band Intensity

MK2206 + 100
(% control)

Characteristic b- and y-fragment ions—including


80
Anti AKT 60 y7, which contains pSer and features the loss of
phospho 40 98 Da (elimination of phosphoric acid)—are shown.
motif 20
(K) Western blot analysis of NR3C1 phosphoryla-
0
NR3C1 MK2206
tion using an anti AKT phospho-motif antibody in
+
NR3C1 immunoprecipitates from CCRF-CEM
cells expressing HA-NR3C1 treated with vehicle or
the MK2206 AKT inhibitor.
See also Figure S1 and Table S1.

localization of retrovirally expressed HA-tagged glucocorticoid levels of AKT activation, showed cytoplasmic localization of
receptor protein, which was completely relocalized to the NR3C1 in basal conditions, which was only partially relocalized
nucleus upon dexamethasone treatment (Figure 2A). Notably, to the nucleus upon dexamethasone treatment (Figures 2E and
expression of MYR-AKT1 in these cells resulted in impaired nu- S2B). Inhibition of AKT with MK2206 effectively enhanced
clear relocalization of NR3C1 after dexamethasone treatment glucocorticoid-induced translocation of the NR3C1 protein to
(Figure 2B). In addition, and in contrast with wild-type glucocor- the nucleus in these cells (Figures 2E and S2). Notably, similar
ticoid receptor, the NR3C1 S134A mutant protein showed results were obtained in primograft T-ALL lymphoblasts in which
increased nuclear localization in basal conditions and effective inhibition of AKT with MK2206 increased the nuclear transloca-
nuclear relocalization upon dexamethasone treatment (Fig- tion of the NR3C1 protein following glucocorticoid treatment
ure 2C), even upon expression of MYR-AKT1 (Figure 2D). Next, (Figure 2F).
we analyzed the capacity of the MK2206 AKT inhibitor to Transcriptionally, MYR-AKT1 impaired the capacity of
modulate glucocorticoid-induced translocation of NR3C1 to NR3C1 to activate a luciferase reporter construct under the
the nucleus in T-ALL cells. CCRF-CEM and MOLT3, two PTEN control of a synthetic glucocorticoid response element (Fig-
null, glucocorticoid-resistant T-ALL cell lines expressing high ure 3A). Consistently, AKT activation via MYR-AKT1 expression

768 Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc.
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

MK2206+
A F DMSO Dexa MK2206 Dexa
p<0.001
NR3C1 DAPI Merge C N C N C N C N

NR3C1 distribution
100
DMSO

T-ALL#9 NR3C1
80

(% total)
100

NR3C1 distribution
60 Cytoplasm
80
Nucleus

(% total)
40 60
Dexa

20 40
20
0
0
DMSO Dexa
B n.s
pAKT1 (S473)

NR3C1 distribution
NR3C1 Merge
DAPI 100
D MSO

AKT1
80
(% total) 60 Tubulin
40
Dex a

20 MAX

0
DMSO Dexa
MK2206+
C p<0.001 DMSO Dexa MK2206 Dexa
C N C N C N C N
NR3C1 distribution

NR3C1 DAPI Merge 100


DMSO

T-ALL#10 NR3C1
80
(% total)

60 100
NR3C1 distribution
Cytoplasm
80 Nucleus
40 (% total) 60
Dexa

20 40
0 20
DMSO Dexa 0
D
p<0.001 pAKT1 (S473)
100
NR3C1 distribution

NR3C1 DAPI Merge


DMSO

AKT1
80
(% total)

60 Tubulin
40
MAX
Dexa

20
0
DMSO Dexa
MK2206+
Cytoplasm DMSO Dexa MK2206 Dexa
Nucleus C N C N C N C N
T-ALL#12 NR3C1
100
E
NR3C1 distribution

MK2206+ Cytoplasm
DMSO Dexa MK2206 Dexa 80
Nucleus
(% total)

C N C N C N C N 60
40
NR3C1
20
pAKT1 0
(S473)
pAKT1 (S473)
AKT1
AKT1
Tubulin
Tubulin
MAX
MAX

Figure 2. AKT1-Mediated S134 Phosphorylation of the NRC3C1 Protein Impairs Dexamethasone-Induced Glucocorticoid Receptor
Nuclear Translocation
(A–D) Confocal microscopy analysis and quantitation of the distribution of NR3C1 cellular localization in U2OS cells expressing HA-NRC31 (A), HA-NRC31 and
MYR-AKT1 (B), HA-NRC31 S134A (C), or HA-NRC31 S134A and MYR-AKT1 (D) in basal conditions (DMSO) and after dexamethasone (Dexa; 1 mM) stimulation.
Scale bars, 20 mm.
(E) Cellular localization analysis of NR3C1 via nuclear and cytoplasmic cell fractionation and analysis of AKT1 signaling in cell lysates from CCRF-CEM T-ALL
cells treated with vehicle only (DMSO), dexamethasone (Dexa; 1 mM), the MK2206 AKT inhibitor (1 mM), or MK2206 plus dexamethasone (1 mM each).
(F) Cellular localization analysis of NR3C1 via western blot analysis of nuclear and cytoplasmic cell fractions in cell lysates from primograft T-ALL lymphoblasts.
Tubulin and MAX proteins are shown as controls for cytosolic and nuclear fractions. C, cytoplasmic fraction; N, nuclear fraction. Data in (A) through (D) are
represented as mean ± SD.
See also Figure S2.

Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc. 769
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

A p<0.001
or PTEN knockdown in DND41 and KOPTK1 T-ALL cells
Relative promoter activity

14 impaired key effector responses mediating glucocorticoid-


12 NR3C1 + + + +
GFP induced cell death, including glucocorticoid receptor auto-
10 MYR-AKT1 Dexa + +
8 NR3C1 upregulation (Geng et al., 2008) and induction of BCL2L11,
6 pAKT1 (S473)
4 AKT1 a proapototic factor essential for glucocorticoid-induced cell
Renilla
2 death (Wang et al., 2003) (Figures 3B and 3C; Figures S3A and
0 S3B). Consistently, glucorticoid-induced apoptosis was signifi-
NR3C1 + + + +
Dexa + + cantly blunted in DND41 and KOPTK1 MYR-AKT-expressing
cells compared with controls (Figure S3C). To better analyze
the potential impact of mutational loss of PTEN in T-ALL on the
B C p<0.001 global transcriptional program induced by glucocorticoids, we
EN

p< 0.001
C

8 6
LU

PT

relative expression
relative expression

7 performed microarray gene expression analysis of DND41


A

A
N

6 5
R

R
sh

sh

BCL2L11
5 4
NR3C1

PTEN shRNA control and PTEN shRNA knockdown cells treated


4 3
NR3C1 3
2
with dexamethasone. In agreement with a global inhibition of
2
pAKT(S473) 1 1 glucocorticoid receptor activity by AKT, microarray analysis of
0 0 gene expression changes induced by dexamethasone treatment
AKT1
D O
a

SO
a

D O
a

SO
a
ex

ex

ex

ex
S

Beta-actin showed complete abrogation of glucocorticoid-induced sig-


M

M
D

D
D

shRNA shRNA shRNA shRNA natures upon activation of AKT1 via PTEN shRNA knockdown
LUC PTEN LUC PTEN
(Figure 3D). Moreover, gene set enrichment analysis (GSEA)
shLUC shPTEN of genes regulated by glucocorticoids in primary leukemia
D Control Dexa Control Dexa E
samples (Schmidt et al., 2006) showed a marked enrichment in
TSC22D3
TP53INP1
ISG20
Geneset: genes upregulated DND41 control cells treated with dexamethasone compared
PIK3IP1
CXCR4
by glucocorticoids in ALL with dexamethasone-treated PTEN shRNA knockdown samples
CD69
p= 0.002
Enrichment Score

CD53 0.1
CD97
0
(Figure 3E).
GZMA
BIRC3 −0.1
RUNX2 −0.2
DDIT4
LY96
−0.3 Pharmacologic Inhibition of AKT Reverses
SMAP2 −0.4
−0.5
LPAR6
DFNA5 −0.6 Glucocorticoid Resistance In Vitro and In Vivo
SESN1 −0.7
P2RX5
RGPD2
To test the therapeutic role of AKT inhibition in the treatment of
RGPD1 Dexa/DMSO Dexa/DMSO glucocorticoid-resistant leukemias, we first treated CCRF-CEM
FAM65B
ADM in shPTEN in shLUC
LOC400027
YPEL5 cells, a PTEN null glucocorticoid-resistant T-ALL cell line, with
UGP2
PRDM1
HHIP−AS1
dexamethasone and MK2206. In these experiments, AKT1 inhi-
UBASH3B
C5orf62 Geneset: genes downregulated bition of CCRF-CEM lymphoblasts with MK2206 effectively
ARHGEF3
by glucocorticoids in ALL
TMEM217
FAM135B
restored glucocorticoid-induced apoptosis and reversed gluco-
FAM69A
Enrichment Score

AKAP7
PRKCB 0.8 p< 0.0001 corticoid resistance in vitro (Figure 4A). Similar results were
JPH1 0.7
CD300A 0.6 obtained in the glucocorticoid-resistant MOLT3 and PF382
RRM2 0.5
PRIM1
AK2
0.4
0.3
T-ALL cell lines (Figure S4A). Analysis of an extended panel of
EXO1 0.2
MBP
GAPDH 0.1 cell lines including B precursor ALL (Figure S4B), diffuse large
MCM6 0
FAM216A −0.1 B cell lymphoma (Figure S4C), and multiple myeloma (Fig-
PAICS
TAGLN2
MCM2
Dexa/DMSO Dexa/DMSO ure S4D) lines showed additive effects with this combination,
H19
GINS2
in shPTEN in shLUC suggesting that the synergistic interaction between glucocorti-
-5 0 5
coids and AKT inhibition may be most prominently relevant in
the context of T-ALL.
Next, we analyzed the effects of MK2206 and glucocorticoid
Figure 3. AKT Activation Inhibits Glucocorticoid-Induced Gene
Expression in vivo in a xenograft model of glucocorticoid-resistant T-ALL.
(A) Luciferase reporter analysis of dexamethasone-induced glucocorticoid CCRF-CEM cells expressing the luciferase gene were injected
receptor transactivation in U2OS cells expressing MYR-AKT1 compared with intravenously in immunodeficient NOG mice, and tumor engraft-
GFP-only-expressing controls using a synthetic glucocorticoid response ment was assessed by in vivo bioimaging at day 18. Animals
element reporter. harboring homogeneous tumor burdens were treated with
(B) Western blot analysis of PTEN expression and AKT activation in DND41 T-
vehicle only (DMSO), MK2206, dexamethasone, or MK2206
ALL cells infected with lentiviruses expressing a PTEN shRNA construct
(shRNA PTEN) or a control shRNA targeting the Renilla luciferase gene (shRNA
plus dexamethasone for 3 days. In this experiment, animals
LUC). treated with dexamethasone or MK2206 showed progressive
(C) Real-time PCR analysis of glucocorticoid-regulated transcripts in control tumor growth similar to that observed in vehicle-treated controls,
and PTEN knockdown DND41 cells treated with vehicle (DMSO) only or 1 mM while mice treated with MK2206 plus dexamethasone had
dexamethasone (Dexa).
(D) Heat map representation of the top differentially expressed genes between
control DND41 cells treated with vehicle only (Control) versus 1 mM dexa- (E) GSEA of genes regulated by glucocorticoids in ALL patients undergoing
methasone (Dexa) and corresponding transcript levels in control and dexa- glucocorticoid therapy in DND41 shRNA LUC dexamethasone-treated cells
methasone-treated PTEN knockdown cells. The scale bar shows color-coded compared with DND41 shRNA PTEN dexamethasone-treated cells. Data in
differential expression, with red indicating higher levels and blue indicating (A) and (C) are represented as mean ± SD.
lower levels of expression. See also Figure S3.

770 Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc.
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

Dexa+ Figure 4. Pharmacologic Inhibition of


A DMSO Dexa MK2206 MK2206 AKT with MK2206 Reverses Glucocorticoid
4.1 6.1 5.7 56 Resistance
56
(A) Representative plots of apoptosis and cell

7-AAD
7-AAD
7-AAD

viability quantification in CCRF-CEM T-ALL cells

7-AAD
C DMSO treated with vehicle only, MK2206 (100 nM),
93.7 1.90 89.41 4.1 92.1 2.1 18.5 24.9
Annexin V Annexin V dexamethasone (Dexa; 1 mM), or dexamethasone
Annexin V Annexin V
PI+ plus MK2206 (Dexa + MK2206; 1 mM and 100 nM,
p<0.001 7.8

PI
p<0.001 respectively) in combination in vitro.
100 p<0.001 p<0.001 p<0.001 PI-
Apoptosis (%)

(B) Tumor load quantification in vivo by biolumi-


80 DMSO 100 91.8

Viability (%)
nescence imaging and analysis of luciferase
60 p<0.001 80
MK2206 activity and human CD45-expressing cells in the
40 p=0.004 60 Dexa
n.s Dexa bone marrow of CCRF-CEM T-ALL xenografts
20 40
Dexa+ treated with vehicle only, MK2206 (10 mg kg!1
0 MK2206 20
PI+
TO-, PI+ via oral gavage twice a day), dexamethasone
0 25.0
25.02

PI
(5 mg kg!1 via intraperitoneal injection), or
Dexa
PI- MK2206 (10 mg kg!1 twice a day) plus dexa-
73.2
TO+, PI-

methasone (5 mg kg!1).
73.20
20 +
D 06

6
le

B 25
K2 a

p<0.001
2
ic

M ex
a

(C and D) Representative plots (C) and quantifi-


Luciferase activity
K2
ex
h
106

Ve

20 Vehicle
D

p<0.001 MK2206
(x106 cells)

0.2
cation (D) of cell viability in primograft T-ALL
Photon flux x

0.4 15 p<0.001 samples treated with vehicle only, MK2206


MK2206
0.6 10 PI+ (100 nM–10 mM), dexamethasone (10 nM–1 mM)
0.8 Dexa 21.4
5 alone, and dexamethasone (10 nM–1 mM) plus
PI
1.0 Dexa+
p<0.031 0 MK2206 PI- MK2206 (100 nM–10 mM) in combination.
77.1 Percentages of viable (PI!), and nonviable
p<0.011
p<0.001 (PI+) cells are indicated. Bar graphs represent
8 p<0.045 120
(relative to control)

Dexa+
x108

p=0.006 mean ± SD.


% leukemia cells

100 MK2206
6 p=0.018 See also Figures S1 and S4.
80
Photon flux

4 60
PI+
40 64.8
2
PI

20
0 0 PI-
29.0
Day 0 +3 0 +3 0 +3 0 +3
To further test the efficacy of this
D p<0.001 p<0.001 treatment combination in vivo, we estab-
p<0.001 p=0.02
p<0.001 p<0.001 p<0.002 lished leukemia xenografts in NOD rag
p<0.001 p<0.001 60
100 100 p<0.001 p<0.001 p<0.001 gamma immunodeficient mice using
cell viability(%)

p<0.001
cell viability(%)
cell viability(%)

80 80 50
p<0.001
T-ALL #19

40 p=0.001 p<0.001 three independent glucocorticoid-resis-


T-ALL #6
T-ALL #9

60 80
cell viability(%)

60 30
T-ALL #10

40 40 20 60 tant primograft T-ALL samples infected


20 20 10 40 with lentiviruses expressing the luciferase
0 0 0
20 gene. Animals harboring homogeneous
Dexa Dexa Dexa
0
p<0.001
tumor burdens by in vivo bioimaging
p<0.001 p<0.001 Dexa
p<0.001 p=0.002 were treated with vehicle only (DMSO),
100 100 p<0.001 100
cell viability(%)
cell viability(%)

cell viability(%)

p<0.001 80 p<0.001 80 p=0.01 DMSO MK2206 MK2206, dexamethasone, or MK2206


80
T-ALL #28
T-ALL #37

T-ALL #39

60 60 p<0.001 60 p=0.006
40 p<0.001 40 40 Dexa Dexa+ plus dexamethasone. In this experiment,
20 20 MK2206
20 mice treated with dexamethasone
0 0 0
or MK2206 showed progressive tumor
Dexa Dexa Dexa
growth similar to that observed in vehicle-
treated controls, while mice treated with
significant antitumor responses (Figure 4B). Following these MK2206 plus dexamethasone showed significant antitumor
results, we evaluated the response to the combination treatment responses (Figure 5). In these experiments, analysis of
MK2206 plus dexamethasone in primary T-ALL lymphoblasts. AKT1 phosphorylation showed effective suppression of AKT1
Toward this goal, we established viable in vitro cultures of signaling in mice treated with MK2206 and MK2206 plus
T-ALL leukemia primograft samples supported by bone marrow dexamethasone in vivo (Figures S5A and S5B). Moreover,
MS5 stroma cells expressing the Delta-like 1 NOTCH1 ligand expression analysis of the TSC22D3 (GILZ) glucocorticoid re-
(Armstrong et al., 2009). Treatment of T-ALL leukemia cultures ceptor target (Ayroldi et al., 2007; D’Adamio et al., 1997; Riccardi
with MK2206 plus dexamethasone in combination showed et al., 1999) and PARP-1 cleavage showed increased glucocor-
significantly increased antileukemic effects compared with ticoid response and enhanced apoptosis in leukemia-infiltrated
treatment with dexamethasone or MK2206 alone (Figures 4C, tissues of mice treated with MK2206 plus dexamethasone in
4D, and S4E). Notably, the antileukemic effects of MK2206 combination, respectively (Figure S5A). Pharmacokinetic ana-
plus dexamethasone were prominent in tumor samples with lysis of MK2206 and dexamethasone ruled out that this effect
PTEN loss (TALL#6, TALL#9, and T-ALL#28) but were also real- could be mediated by decreased clearance of dexamethasone,
ized in PTEN-expressing tumors with moderate levels of AKT1 as the pharmacokinetic profile of this glucocorticoid was iden-
activation (T-ALL #19, TALL#37, and T-ALL#39) (Figure S4F). tical in animals treated with dexamethasone alone and mice

Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc. 771
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

A day 0 day 5 day 0 day 5 B day 0 day 4 day 0 day 4 C day 0 day 5 day 0 day 5

vehicle

Dexa
Dexa

vehicle

Dex a
vehicle
Photon flux x108

Photon flux x107

Photon flux x107


0.5 0.5 0.5
1.0 1.0 1.0
1.5 1.5 1.5
2.0 2.0 2.0

M K 2 20 6 &

MK2206 &

MK2206 &
2.5 2.5 2.5
MK2206

M K2 2 0 6

MK2206
Dexa

Dexa

Dexa
D p=0.00017 E p=0.0003 F p=0.0005
5.9 2.42 3.92 0.38 Mean 2.04 1.74 2.28 0.77 Mean 1.26 1.37 1.61 0.52 Mean
burden (photon flux)

burden (photon flux)

burden (photon flux)


10 10 10
Fold change tumor

Fold change tumor

Fold change tumor


1 1 1

0.1 0.1 0.1


Vehicle MK-2206 Vehicle MK-2206 Vehicle MK-2206
Dexa Dexa + Dexa Dexa + Dexa Dexa +
MK2206 MK2206 MK2206

G H I

06
06
06

6
6
6

K2 +
K2 +

le

20
e

22
20
K2 +

22
e

20
22

M a
cl

ic

a
M xa
cl

a
M xa
a

ex
K-
ex
hi

K-
ex

h
hi

K-
ex

Ve
e

Ve

D
D
Ve

D
D
M

D
D

J K L
Mean 554 310 380 70 Mean 498 347 425 150 Mean 409 317 324 133
p <0.0001 p =0.0004 800 p =0.0038
Spleen weight (mg)

Spleen weight (mg)

800 800
Spleen weight (mg)

p =0.0016 p =0.0079
p =0.0095
600 p =0.0003 600
600 p =0.0057 p =0.0079
400 400
400
200 200
200
0 0
0
Vehicle Dexa MK2206 Dexa + Vehicle Dexa MK2206 Dexa + Vehicle Dexa MK2206 Dexa +
MK2206 MK2206 MK2206

M N p =0.0043
O p <0.001
p <0.001
p <0.001 p =0.0127 p <0.001
luciferase activity x 103

luciferase activity x 103

luciferase activity x 103

60 p <0.001 80 100 p <0.001


p =0.045
Bone marrow

Bone marrow

Bone marrow

60 Vehicle 80 Vehicle
Vehicle
40 MK2206 MK2206 MK2206
Dexa
60
Dexa 40 Dexa
20 Dexa + Dexa + 40 Dexa +
MK2206 20 MK2206 MK2206
20
0 0 0

Figure 5. Pharmacologic Inhibition of AKT Reverses Glucocorticoid Resistance in Primary Human T-ALL Xenografts In Vivo
(A–C) Representative examples of tumor load analysis via in vivo bioluminescence imaging in mice xenografted with three independent human T-ALLs treated
with vehicle only, MK2206 (10 mg kg!1 twice a day), dexamethasone (5 mg kg!1), or MK2206 (10 mg kg!1 twice a day) plus dexamethasone (5 mg kg!1).
(D–F) Quantitative analysis of tumor load and therapy response based on luciferase activity.
(G–I) Representative images of spleens in primary T-ALL xenografted mice at the end of treatment.
(J–L) Quantitative analysis of tumor burden estimated by spleen weight in T-ALL xenografted mice at the end of treatment. (M–O) Quantitative analysis of tumor
burden estimated by luciferase counts in bone marrow in T-ALL xenografted mice at the end of treatment. Bar graphs represent mean ± SD.
(A), (D), (G), (J), and (M) correspond to T-ALL #27. (B), (E), (H), (K), and (N) correspond to T-ALL #19. (C), (F), (I), (L), and (O) correspond to T-ALL #9. Scale bars,
2 cm. See also Figure S5.

772 Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc.
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

A B Figure 6. Pharmacologic Inhibition of AKT


100 Dexa 100 Reverses Glucocorticoid Resistance in
a Mouse Model of Glucorticoid-Resistant

Survival (%)
80 80
Survival (%)

p < 0.01
T-ALL
60 60
p= n.s (A) Kaplan-Meier curve in mice treated with
40 Control 40
dexamethasone (Dexa) or vehicle (Control)
20 20
Dexa Control
after allograft transplantation of Pten-nondeleted
0 0 (Ptenf/f) NOTCH1-induced T-ALL tumor cells.
0 5 10 15 20 25 30 35 0 5 10 15 20 25 30 35
Arrows indicate drug treatment.
Time (days) Time (days)
(B) Kaplan-Meier curve in mice treated with
dexamethasone (Dexa) or vehicle (Control) after
C day 0 day 3 day 0 day 3

Relative photon flux Relative photon flux


Relative photon flux Relative photon flux

allograft transplantation of Pten-deleted (Pten!/!)


4 4

(fold change)
(fold change)

NOTCH1-induced T-ALL tumor cells. Arrows


2 2
Vehicle

indicate drug treatment.

Dexa
1 1
(C and D) Representative images and changes
0.5 0.5
in bioluminescence by in vivo imaging (C), and
0.25 day day 0.25 day day statistical analysis of differences in treatment
0 3 0 3
response (D) in mice allografted with NOTCH1-
4 4 induced Pten!/! mouse leukemia cells treated

(fold change)
(fold change)

2
MK2206

2 with vehicle only, MK2206 (10 mg kg!1 via oral


MK2206
Dexa +

1 1 gavage twice a day), dexamethasone (Dexa;


0.5 0.5 5 mg kg!1), or MK2206 (10 mg kg!1 twice a day)
0.25 0.25 plus dexamethasone (5 mg kg!1) (Dexa +
day day day day
0 3 0 3
0.2
0.4

1.0
0.6
0.8
0.2
0.4

1.0
0.6
0.8

MK2206).
Luminescence Luminescence (E) Kaplan-Meier overall survival curve in mice
photon flux (x106) photon flux (x106)
allografted with NOTCH1-induced Pten!/! mouse
D E leukemia cells and treated with vehicle only
Mean 1.73 1.25 1.72 0.66 (control), MK2206 (10 mg kg!1 via oral gavage
p <0.0001 twice a day), dexamethasone (Dexa; 5 mg kg!1), or
10 0 Vehicle
Fold change tumor burden

10 p <0.01 MK2206 (10 mg kg!1 twice a day), plus dexa-


Dexa methasone (5 mg kg!1) (Dexa + MK2206).
p <0.001
p <0.002
Survival (%)

MK2206 (F and G) Quantification of glucocorticoid-induced


(photon flux)

50 Dexa + loss of viability in isogenic NOTCH1-induced


1 MK2206 Ptenf/f or Pten!/! mouse leukemia cells infected
p <0.0001 with retroviruses expressing the wild-type (F) or
the S134A mutant (G) glucocorticoid receptor
0
0 5 10 15 20 NR3C1. Data are represented as mean ± SD.
0.1 Vehicle MK2206
See also Figure S6.
Dexa Dexa + Days
MK2206

F G
120 120
100 this model, transplantation of tamox- Ptenf/f Ptenf/f
100
Viability (%)

Viability (%)

80 ifen-inducible conditional Pten knockout Pten-/- 80 Pten-/-


60 (ROSA26Cre-ERT2/+;Ptenf/f) hematopoietic 60
40 40 progenitors infected with retroviruses
20 20 expressing a mutant constitutively
0 0
0 10 10 10 0 10 10 10 -8 active form of the NOTCH1 receptor
-7 -6 -8 -7 -6

Dexamethasone (M) Dexamethasone (M) (NOTCH1 L1601P DPEST) resulted in


the development of NOTCH1-driven
T-ALL tumors as described elsewhere
treated with MK2206 plus dexamethasone in combination (Fig- (Chiang et al., 2008). Next, we infected NOTCH1 L1601P
ure S5C). In addition, even though we observed a modest DPEST, ROSA26Cre-ERT2/+, Ptenf/f T-ALL lymphoblasts with a
decrease in MK2206 half life in mice treated with dexametha- luciferase-expressing retrovirus and transplanted them into
sone plus MK2206 (Figure S5C), AKT inhibitor levels obtained secondary recipients that were treated with vehicle only or
in animals treated with MK2206 as single agent already result tamoxifen in order to generate Pten-nondeleted and Pten-
in complete suppression of AKT activation in vivo (Figures S5A deleted isogenic tumors, respectively. Treatment of Pten-non-
and S5B), supporting the conclusion that this pharmacokinetic deleted tumor-bearing mice with dexamethasone showed a
interaction does not account for the increased antileukemic significant improvement in survival compared with controls
responses observed in mice treated with dexamethasone plus treated with vehicle only (Figure 6A). In contrast, and consis-
MK2206. tent with a role of Pten loss and AKT1 activation in promoting
Finally, we generated a mouse leukemia model in which glucocorticoid resistance, all mice harboring Pten-deleted
glucocorticoid resistance is specifically driven by genetic loss tumors failed to respond to dexamethasone treatment and
of Pten using a well-established retroviral transduction and showed no survival differences compared to vehicle-treated
bone marrow transplantation protocol (Chiang et al., 2008). In controls (Figure 6B).

Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc. 773
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

To test the efficacy of MK2206 and glucocorticoids in combi- Mechanistically, we show that AKT1 can induce glucocorti-
nation, we treated mice transplanted with NOTCH1-induced coid resistance by phosphorylation of the glucocorticoid recep-
Pten-deleted murine tumors expressing luciferase in secondary tor in position S134, which impairs the nuclear relocalization of
recipients, with vehicle only (DMSO), MK2206, dexamethasone, the glucocorticoid receptor protein and blocks transcriptional
or MK2206 plus dexamethasone and monitored their response regulation of glucocorticoid target genes. However, it should
to therapy by in vivo bioimaging. Animals treated with dexa- be noted that additional mechanisms downstream of AKT1 in
methasone or MK2206 in this experiment showed progressive addition to direct inactivation of the glucocorticoid receptor
tumor growth similar to that observed in vehicle-treated con- can also promote resistance to glucocorticoid therapy by
trols, while mice treated with MK2206 plus dexamethasone promoting cell growth, metabolism, and survival in T-ALL. In
showed significant antitumor responses (Figures 6C and 6D), this regard, mTOR phosphorylation by AKT impairs glucocorti-
which translated in significantly improved survival in this coid-induced apoptosis by increasing the expression of MCL1
group (Figure 6E). As before, analysis of AKT1 phosphorylation (Wei et al., 2006). In addition, AKT phosphorylation inhibits
showed effective suppression of AKT1 signaling in mice BAD-mediated apoptosis (Bornhauser et al., 2007; Datta
treated with MK2206 and MK2206 plus dexamethasone et al., 1997; del Peso et al., 1997); AKT-mediated phosphoryla-
in vivo (Figure S6A), while GILZ expression analysis showed tion of XIAP prevents the autoubiquitination and subsequent
increased glucocorticoid response (Figure S6A) and PARP-1 degradation of this antiapoptotic factor (Bornhauser et al.,
cleavage demonstrated increased apoptosis in tumor cells 2007; Dan et al., 2004); and increased metabolism induced
from animals treated with MK2206 plus dexamethasone in by AKT activation can antagonize metabolic inhibition induced
combination (Figure S6A). Consistently, histological analysis by glucocorticoids (Beesley et al., 2009). The convergent ef-
of leukemia-infiltrated bone marrow showed increased antileu- fects of direct and indirect mechanisms downstream of AKT1
kemic effects (Figure S6B) and marked increased apoptosis antagonizing the antileukemic effects of glucocorticoids further
(Figure S6C) in mice xenografted with NOTCH1-induced support the role of the PI3K-AKT pathway as therapeutic target
Pten!/! T-ALL cells treated with MK2206 plus dexamethasone for the reversal of primary glucocorticoid resistance in T-ALL.
in combination.
Finally, we analyzed the role of NR3C1 S134 phosphorylation EXPERIMENTAL PROCEDURES
in the therapeutic response to glucocorticoids and the effects
of Pten loss in glucocorticoid therapy in this model. Retroviral Patient Samples
expression of the glucocorticoid receptor enhanced the T-ALL samples were provided by Columbia Presbyterian Hospital, the Eastern
Cooperative Oncology Group (ECOG), University of Padova, and Hospital
response of NOTCH1-induced Pten nondeleted leukemias
Central de Asturias with informed consent and analyzed under the supervision
to glucocorticoid treatment, an effect that was effectively
of the Columbia University Medical Center Institutional Review Board
abrogated upon Pten loss (Figures 6F and S6D). In contrast, committee.
expression of the AKT-resistant NR3C1 S134A mutant protein
was equally effective at increasing the antileukemic effects of Inhibitors and Drugs
glucocorticoids in Pten nondeleted and Pten null lymphoblasts MK2206 or 8-[4-(1-aminocyclobutyl)phenyl]-9-phenyl-1,2,4-triazolo[3,4-f]
(Figures 6G and S6D). Overall, these results support that [1,6]naphthyridin-3 (2H)-one hydrochloride [1:1] was from Selleck Chemicals
pharmacologic inhibition of AKT can effectively enhance LLC. Dexamethasone and 4-Hydroxytamoxifen were from Sigma-Aldrich.
glucocorticoid response and reverse glucocorticoid resistance
in T-ALL. In Vitro Kinase Assays
Flag-tagged recombinant GST-NR3C1 and GST-NR3C1 S134A mutant
proteins were incubated with recombinant active His-AKT1 protein (Millipore)
DISCUSSION
in kinase buffer (Cell Signaling) containing g-32P-ATP and analyzed by autora-
diography after SDS-PAGE.
Numerous mechanisms have been proposed to explain the lack
of effective glucocorticoid-induced apoptosis in prednisone
Mass Spectrometry
poor responders (Bhadri et al., 2012). However, and despite HA-NR3C1 protein was immunoprecipitated from U2OS cells stably express-
much research over the last 2 decades, the molecular basis ing HA-tagged human NR3C1 and Myr-AKT with anti-HA antibody-conjugated
of primary glucocorticoid resistance in ALL remains poorly beads (Sigma), electrophoresed on a 3%–8% Tris-acetate gel, stained with
understood. Given that primary glucocorticoid resistance is a Simply Blue Stain (Invitrogen), excised, reduced with dithiothreitol, alkylated
cell-intrinsic property of leukemia lymphoblasts present before with iodoacetamide, digested with trypsin, and analyzed for phosphorylated
peptides by nanoscale liquid chromatographic tandem mass spectrometry
exposure to glucocorticoid therapy, we proposed that primary
(LC-MS/MS). MS/MS spectra were processed using ProteinLynx from the
oncogenic signaling pathways involved in leukemia transforma- MassLynx 4.0 software and searched against the Swiss-Prot protein database
tion could also function as negative upstream regulators of the using Mascot (http://www.matrixscience.com) with differential modifications
glucocorticoid receptor driving this phenotype. The identification for Ser/Thr/Tyr phosphorylation (+79.97).
of AKT1 as a driver of glucocorticoid resistance in T-ALL is To test the effects of AKT1 inhibition on NR3C1 S134 phosphorylation, we
perfectly consistent with this model. Notably, direct activation treated U2OS cells stably expressing HA-tagged human NR3C1 and the
constitutively active Myr-AKT with DMSO or MK2206 (10 mM, 5 hr). We
of AKT1 can drive T cell transformation (Kharas et al., 2010),
immunoprecipitated HA-NR3C1 protein with anti-HA antibody-conjugated
activation of PI3K-AKT signaling as a result of deletions and
beads (Sigma). Immunopurified HA-NR3C1 was then electrophoresed on a
mutations in PTEN (Palomero et al., 2007) is highly prevalent 4%–12% Bis-Tris gel, stained with Simply Blue Stain (Invitrogen), digested
in T-ALL, and PTEN mutations are associated with primary with trypsin, and analyzed for the phosphorylation status at residue S134 by
glucocorticoid resistance in the clinic (Bandapalli et al., 2013). microcapillary LC-MS/MS.

774 Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc.
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

Immunofluorescence ACKNOWLEDGMENTS
We analyzed U2OS cells stably expressing wild-type NR3C1 or the S134A
NR3C1 by NR3C1 immunofluorescence (1:500; Santa Cruz Biotechnology), This work was supported by the National Institutes of Health (grants
followed by Alexa Fluor 594 (1:1,000; Invitrogen) staining and confocal micro- R01CA129382 to A.A.F., U24 CA114737 to E.P., RC2 CA148308 to A.C. and
scopy. In colocalization studies, T-ALL cell lines were immunostained with A.F., and U54CA121852 to A.C.), the Stand Up To Cancer Innovative Research
rabbit antibodies against NR3C1 (1:300; Santa Cruz Biotechnology) and a Award (to A.A.F.), the Chemotherapy Foundation (to A.A.F.), the Leukemia &
mouse antibody against AKT (1:100) (Cell Signaling, #2920) using anti-rabbit Lymphoma Society Scholar Award (to A.A.F.), a Leukemia & Lymphoma Soci-
Alexa Fluor 594 (1:500; Invitrogen) and anti-mouse Alexa Fluor 488 (1:500; ety SCOR Grant (to A.A.F.), and the ECOG Leukemia Tissue Bank. We are
Invitrogen) as secondary antibodies. We stained nuclei with TO-PRO3 reagent grateful to A. Kung for the FUW-luc vector, J. Aster for the MigR1-NOTCH1
(Invitrogen) and visualized them by confocal imaging on a Zeiss LSM510-NLO L1601PDP vector, P.P. Pandolfi for the Ptenf conditional knockout mouse,
microscope. T. Ludwig for the ROSA26Cre-ERT2/+ mouse, S. Minuzzo for generation of
T-ALL xenografts, L. Xu for help in animal procedures, M.A. Gawinowicz
Luciferase Reporter Assays from the Proteomics Share Resource at the Herbert Irving Comprehensive
We performed NR3C1 reporter assays using the Cignal GRE Reporter (luc) Cancer Center at Columbia University for assistance in mass spectrometry
Kit (SABiosciences). analysis, and R. Baer for helpful discussions and revision of the manuscript.

Received: July 26, 2011


Microarray Expression Analysis of Glucocorticoid Response
Revised: May 25, 2013
DND41 shRNA LUC and DND41 PTEN shRNA cells were treated with vehicle
Accepted: October 31, 2013
only (DMSO) or dexamethasone (1 mM) for 24 hr in triplicate. RNA was iso-
Published: November 27, 2013
lated, labeled, and hybridized to the HumanHT-12 v4 Expression BeadChip
(Illumina) using standard procedures. Raw gene expression data were log
REFERENCES
transformed and quantile normalized using MATLAB. Differentially regulated
genes in DND41 shRNA LUC versus DND41 PTEN shRNA upon dexameth-
Armstrong, F., Brunet de la Grange, P., Gerby, B., Rouyez, M.C., Calvo, J.,
asone treatment with a fold change >2 were selected by t test. Glucocorti-
Fontenay, M., Boissel, N., Dombret, H., Baruchel, A., Landman-Parker, J.,
coid-regulated transcripts (t test, p < 0.01, and fold change >1.25) in primary
et al. (2009). NOTCH is a key regulator of human T-cell acute leukemia initiating
ALL were identified from processed expression data obtained from
cell activity. Blood 113, 1730–1740.
peripheral blood lymphoblasts from 13 pediatric ALL patients after 24 hr
Ayroldi, E., Zollo, O., Bastianelli, A., Marchetti, C., Agostini, M., Di Virgilio, R.,
of treatment with glucocorticoids (Schmidt et al., 2006). Enrichment of
and Riccardi, C. (2007). GILZ mediates the antiproliferative activity of
these glucocorticoid signature genes in shRNA PTEN versus shRNA
glucocorticoids by negative regulation of Ras signaling. J. Clin. Invest. 117,
LUC dexamethasone-treated samples normalized to DMSO controls was
1605–1615.
analyzed by GSEA using the t test metric and 1,000 permutations of the
genes. Bandapalli, O.R., Zimmermann, M., Kox, C., Stanulla, M., Schrappe, M.,
Ludwig, W.D., Koehler, R., Muckenthaler, M.U., and Kulozik, A.E. (2013).
NOTCH1 activation clinically antagonizes the unfavorable effect of PTEN
Mice
inactivation in BFM-treated children with precursor T-cell acute lymphoblastic
Animal procedures were approved by the Columbia University Institutional
leukemia. Haematologica 98, 928–936.
Animal Care and Use Committee. ROSA26Cre-ERT2/+ mice expressing a tamox-
ifen-inducible form of the Cre recombinase from the ubiquitous Rosa26 Beesley, A.H., Firth, M.J., Ford, J., Weller, R.E., Freitas, J.R., Perera, K.U., and
locus (Guo et al., 2007) and Pten conditional knockout mice (Ptenf) have Kees, U.R. (2009). Glucocorticoid resistance in T-lineage acute lymphoblastic
been described elsewhere (Trotman et al., 2003). To generate NOTCH1- leukaemia is associated with a proliferative metabolism. Br. J. Cancer 100,
induced T-ALL tumors in mice, we performed retroviral transduction of 1926–1936.
bone marrow cells with an activated form of the NOTCH1 oncogene Bhadri, V.A., Trahair, T.N., and Lock, R.B. (2012). Glucocorticoid resistance in
(NOTCH1 L1601P DPEST) and transplanted them via intravenous injection paediatric acute lymphoblastic leukaemia. J. Paediatr. Child Health 48,
into lethally irradiated recipients as described elsewhere (Chiang et al., 634–640.
2008). Primary human T-ALL xenografts experiments were generated via Bornhauser, B.C., Bonapace, L., Lindholm, D., Martinez, R., Cario, G.,
intravenous injection of human T-ALL lymphoblasts into NOD rag gamma Schrappe, M., Niggli, F.K., Schäfer, B.W., and Bourquin, J.P. (2007). Low-
mice. We evaluated disease progression and therapy response by in vivo dose arsenic trioxide sensitizes glucocorticoid-resistant acute lymphoblastic
bioimaging with the In Vivo Imaging System (Xenogen), human CD45 analysis leukemia cells to dexamethasone via an Akt-dependent pathway. Blood
by flow cytometry, and analysis of luciferase activity in bone marrow and 110, 2084–2091.
spleen.
Chiang, M.Y., Xu, L., Shestova, O., Histen, G., L’heureux, S., Romany, C.,
Childs, M.E., Gimotty, P.A., Aster, J.C., and Pear, W.S. (2008). Leukemia-
Statistical Analyses associated NOTCH1 alleles are weak tumor initiators but accelerate K-ras-
We performed statistical analysis using Student’s t test and Wilcoxon signed- initiated leukemia. J. Clin. Invest. 118, 3181–3194.
rank test. We considered results with p < 0.05 as statistically significant. D’Adamio, F., Zollo, O., Moraca, R., Ayroldi, E., Bruscoli, S., Bartoli, A.,
Survival in animal experiments was represented with Kaplan-Meier curves, Cannarile, L., Migliorati, G., and Riccardi, C. (1997). A new dexamethasone-
and significance was estimated with the log-rank test (Prism GraphPad). induced gene of the leucine zipper family protects T lymphocytes from TCR/
CD3-activated cell death. Immunity 7, 803–812.
ACCESSION NUMBERS Dan, H.C., Sun, M., Kaneko, S., Feldman, R.I., Nicosia, S.V., Wang, H.G.,
Tsang, B.K., and Cheng, J.Q. (2004). Akt phosphorylation and stabilization
Microarray gene expression data are available in Gene Expression Omnibus of X-linked inhibitor of apoptosis protein (XIAP). J. Biol. Chem. 279, 5405–
under accession numbers GSE41062 and GSE32215. 5412.
Datta, S.R., Dudek, H., Tao, X., Masters, S., Fu, H., Gotoh, Y., and Greenberg,
SUPPLEMENTAL INFORMATION M.E. (1997). Akt phosphorylation of BAD couples survival signals to the cell-
intrinsic death machinery. Cell 91, 231–241.
Supplemental Information includes Supplemental Experimental Procedures, del Peso, L., González-Garcı́a, M., Page, C., Herrera, R., and Nuñez, G. (1997).
six figures, and one table and can be found with this article online at http:// Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt.
dx.doi.org/10.1016/j.ccr.2013.10.022. Science 278, 687–689.

Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc. 775
Cancer Cell
AKT Inhibition Reverses Glucocorticoid Resistance

Dördelmann, M., Reiter, A., Borkhardt, A., Ludwig, W.D., Götz, N., Viehmann, Ozes, O.N., Mayo, L.D., Gustin, J.A., Pfeffer, S.R., Pfeffer, L.M., and Donner,
S., Gadner, H., Riehm, H., and Schrappe, M. (1999). Prednisone response D.B. (1999). NF-kappaB activation by tumour necrosis factor requires the
is the strongest predictor of treatment outcome in infant acute lymphoblastic Akt serine-threonine kinase. Nature 401, 82–85.
leukemia. Blood 94, 1209–1217. Palomero, T., Sulis, M.L., Cortina, M., Real, P.J., Barnes, K., Ciofani, M.,
Geng, C.D., Schwartz, J.R., and Vedeckis, W.V. (2008). A conserved molecular Caparros, E., Buteau, J., Brown, K., Perkins, S.L., et al. (2007). Mutational
mechanism is responsible for the auto-up-regulation of glucocorticoid recep- loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia.
tor gene promoters. Mol. Endocrinol. 22, 2624–2642. Nat. Med. 13, 1203–1210.
Guo, K., McMinn, J.E., Ludwig, T., Yu, Y.H., Yang, G., Chen, L., Loh, D., Li, C., Pui, C.H., Carroll, W.L., Meshinchi, S., and Arceci, R.J. (2011). Biology, risk
Chua, S., Jr., and Zhang, Y. (2007). Disruption of peripheral leptin signaling in stratification, and therapy of pediatric acute leukemias: an update. Journal
mice results in hyperleptinemia without associated metabolic abnormalities. of clinical oncology: official journal of the American Society of Clinical
Endocrinology 148, 3987–3997. Oncology 29, 551–565.
Heitzer, M.D., Wolf, I.M., Sanchez, E.R., Witchel, S.F., and DeFranco, D.B. Pui, C.H., Mullighan, C.G., Evans, W.E., and Relling, M.V. (2012). Pediatric
(2007). Glucocorticoid receptor physiology. Rev. Endocr. Metab. Disord. 8, acute lymphoblastic leukemia: where are we going and how do we get there?
321–330. Blood 120, 1165–1174.
Hirai, H., Sootome, H., Nakatsuru, Y., Miyama, K., Taguchi, S., Tsujioka, K.,
Riccardi, C., Cifone, M.G., and Migliorati, G. (1999). Glucocorticoid hormone-
Ueno, Y., Hatch, H., Majumder, P.K., Pan, B.S., and Kotani, H. (2010).
induced modulation of gene expression and regulation of T-cell death: role of
MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard
GITR and GILZ, two dexamethasone-induced genes. Cell Death Differ. 6,
chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol.
1182–1189.
Cancer Ther. 9, 1956–1967.
Schlossmacher, G., Stevens, A., and White, A. (2011). Glucocorticoid
Hongo, T., Yajima, S., Sakurai, M., Horikoshi, Y., and Hanada, R. (1997). In vitro
receptor-mediated apoptosis: mechanisms of resistance in cancer cells.
drug sensitivity testing can predict induction failure and early relapse of
J. Endocrinol. 211, 17–25.
childhood acute lymphoblastic leukemia. Blood 89, 2959–2965.
Schmidt, S., Rainer, J., Riml, S., Ploner, C., Jesacher, S., Achmüller, C., Presul,
Inaba, H., and Pui, C.H. (2010). Glucocorticoid use in acute lymphoblastic
E., Skvortsov, S., Crazzolara, R., Fiegl, M., et al. (2006). Identification of
leukaemia. Lancet Oncol. 11, 1096–1106.
glucocorticoid-response genes in children with acute lymphoblastic leukemia.
Kharas, M.G., Okabe, R., Ganis, J.J., Gozo, M., Khandan, T., Paktinat, M.,
Blood 107, 2061–2069.
Gilliland, D.G., and Gritsman, K. (2010). Constitutively active AKT depletes
hematopoietic stem cells and induces leukemia in mice. Blood 115, 1406– Trotman, L.C., Niki, M., Dotan, Z.A., Koutcher, J.A., Di Cristofano, A., Xiao, A.,
1415. Khoo, A.S., Roy-Burman, P., Greenberg, N.M., Van Dyke, T., et al. (2003).
Pten dose dictates cancer progression in the prostate. PLoS Biol. 1, E59.
Klumper, E., Pieters, R., Veerman, A.J., Huismans, D.R., Loonen, A.H., Hählen,
K., Kaspers, G.J., van Wering, E.R., Hartmann, R., and Henze, G. (1995). Wang, Z., Malone, M.H., He, H., McColl, K.S., and Distelhorst, C.W. (2003).
In vitro cellular drug resistance in children with relapsed/refractory acute Microarray analysis uncovers the induction of the proapoptotic BH3-only
lymphoblastic leukemia. Blood 86, 3861–3868. protein Bim in multiple models of glucocorticoid-induced apoptosis. J. Biol.
Chem. 278, 23861–23867.
Mok, C.L., Gil-Gómez, G., Williams, O., Coles, M., Taga, S., Tolaini, M., Norton,
T., Kioussis, D., and Brady, H.J. (1999). Bad can act as a key regulator of Wei, G., Twomey, D., Lamb, J., Schlis, K., Agarwal, J., Stam, R.W., Opferman,
T cell apoptosis and T cell development. J. Exp. Med. 189, 575–586. J.T., Sallan, S.E., den Boer, M.L., Pieters, R., et al. (2006). Gene expression-
Morishita, N., Tsukahara, H., Chayama, K., Ishida, T., Washio, K., Miyamura, based chemical genomics identifies rapamycin as a modulator of MCL1 and
T., Yamashita, N., Oda, M., and Morishima, T. (2012). Activation of Akt is glucocorticoid resistance. Cancer Cell 10, 331–342.
associated with poor prognosis and chemotherapeutic resistance in pediatric Zimmermann, S., and Moelling, K. (1999). Phosphorylation and regulation of
B-precursor acute lymphoblastic leukemia. Pediatr. Blood Cancer 59, 83–89. Raf by Akt (protein kinase B). Science 286, 1741–1744.

776 Cancer Cell 24, 766–776, December 9, 2013 ª2013 Elsevier Inc.

Vous aimerez peut-être aussi