vulnerabilities of pten tp53 -defi cient prostate cancers ......896 | cancer discoveryaugust 2014...

10
896 | CANCER DISCOVERYAUGUST 2014 www.aacrjournals.org Vulnerabilities of PTEN TP53 -Deficient Prostate Cancers to Compound PARP–PI3K Inhibition Enrique González-Billalabeitia 1,2,11 , Nina Seitzer 1,2 , Su Jung Song 1,2 , Min Sup Song 1,2 , Akash Patnaik 3,4,7 , Xue-Song Liu 1,2 , Mirjam T. Epping 1,2 , Antonella Papa 1,2 , Robin M. Hobbs 1,2 , Ming Chen 1,2 , Andrea Lunardi 1,2 , Christopher Ng 1,2 , Kaitlyn A. Webster 1,2 , Sabina Signoretti 5,9 , Massimo Loda 6,8,9,10 , John M. Asara 7 , Caterina Nardella 1,2,5 , John G. Clohessy 1,2,5 , Lewis C. Cantley 4,7 , and Pier Paolo Pandolfi 1,2 RESEARCH BRIEF ABSTRACT Prostate cancer is the most prevalent cancer in males, and treatment options are limited for advanced forms of the disease. Loss of the PTEN and TP53 tumor suppressor genes is commonly observed in prostate cancer, whereas their compound loss is often observed in advanced prostate cancer. Here, we show that PARP inhibition triggers a p53-dependent cellular senescence in a PTEN-deficient setting in the prostate. Surprisingly, we also find that PARP- induced cellular senescence is morphed into an apoptotic response upon compound loss of PTEN and p53. We further show that superactivation of the prosurvival PI3K–AKT signaling pathway limits the efficacy of a PARP single-agent treatment, and that PARP and PI3K inhibitors effectively synergize to suppress tumorigenesis in human prostate cancer cell lines and in a Pten/ Trp53–deficient mouse model of advanced prostate cancer. Our findings, therefore, identify a combinatorial treatment with PARP and PI3K inhibitors as an effective option for PTEN-deficient prostate cancer. SIGNIFICANCE: The paucity of therapeutic options in advanced prostate cancer displays an urgent need for the preclinical assessment of novel therapeutic strategies. We identified differential therapeutic vulnerabilities that emerge upon the loss of both PTEN and p53, and observed that combined inhibition of PARP and PI3K provides increased efficacy in hormone-insensitive advanced prostate cancer. Cancer Discov; 4(8); 896–904. ©2014 AACR. Authors’ Affiliations: 1 Cancer Research Institute, Beth Israel Deaconess Can- cer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; 2 Department of Medicine; Divisions of 3 Hematology/Oncology and 4 Signal Transduction, Department of Medicine; 5 Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center; 6 Department of Pathology, Brigham and Women’s Hospital; 7 Depart- ment of Systems Biology, Harvard Medical School; 8 Center for Molecular Oncologic Pathology; 9 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; 10 Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts; and 11 On leave of absence: Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseg- uer, Murcia, Spain Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/). E. González-Billalabeitia and N. Seitzer contributed equally to this work. Corresponding Author: Pier Paolo Pandolfi, Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Dea- coness Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215. Phone: 617-735-2121/2141; Fax: 617-735-2120; E-mail: [email protected] doi: 10.1158/2159-8290.CD-13-0230 ©2014 American Association for Cancer Research. INTRODUCTION There is an urgent need for the development of novel suc- cessful strategies for advanced prostate cancer to improve patient outcomes (1, 2). The loss of at least one allele of phosphatase and tensin homolog deleted on chromosome 10 ( PTEN) is observed in more than 40% of prostate cancers (3, 4). As a lipid phos- phatase, PTEN suppresses the activation of the PI3K–AKT signaling cascade that is a central proto-oncogenic signaling on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Upload: others

Post on 17-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

896 | CANCER DISCOVERY�AUGUST 2014 www.aacrjournals.org

Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K Inhibition Enrique González-Billalabeitia 1 , 2 , 11 , Nina Seitzer 1 , 2 , Su Jung Song 1 , 2 , Min Sup Song 1 , 2 , Akash Patnaik 3 , 4 , 7 , Xue-Song Liu 1 , 2 , Mirjam T. Epping 1 , 2 , Antonella Papa 1 , 2 , Robin M. Hobbs 1 , 2 , Ming Chen 1 , 2 , Andrea Lunardi 1 , 2 , Christopher Ng 1 , 2 , Kaitlyn A. Webster 1 , 2 , Sabina Signoretti 5 , 9 , Massimo Loda 6 , 8 , 9 , 10 , John M. Asara 7 , Caterina Nardella 1 , 2 , 5 , John G. Clohessy 1 , 2 , 5 , Lewis C. Cantley 4 , 7 , and Pier Paolo Pandolfi 1 , 2

RESEARCH BRIEF

ABSTRACT Prostate cancer is the most prevalent cancer in males, and treatment options

are limited for advanced forms of the disease. Loss of the PTEN and TP53 tumor

suppressor genes is commonly observed in prostate cancer, whereas their compound loss is often

observed in advanced prostate cancer. Here, we show that PARP inhibition triggers a p53-dependent

cellular senescence in a PTEN -defi cient setting in the prostate. Surprisingly, we also fi nd that PARP-

induced cellular senescence is morphed into an apoptotic response upon compound loss of PTEN and

p53. We further show that superactivation of the prosurvival PI3K–AKT signaling pathway limits the

effi cacy of a PARP single-agent treatment, and that PARP and PI3K inhibitors effectively synergize to

suppress tumorigenesis in human prostate cancer cell lines and in a Pten / Trp53 –defi cient mouse model

of advanced prostate cancer. Our fi ndings, therefore, identify a combinatorial treatment with PARP and

PI3K inhibitors as an effective option for PTEN -defi cient prostate cancer.

SIGNIFICANCE: The paucity of therapeutic options in advanced prostate cancer displays an urgent need

for the preclinical assessment of novel therapeutic strategies. We identifi ed differential therapeutic

vulnerabilities that emerge upon the loss of both PTEN and p53, and observed that combined inhibition

of PARP and PI3K provides increased effi cacy in hormone-insensitive advanced prostate cancer. Cancer

Discov; 4(8); 896–904. ©2014 AACR.

Authors’ Affi liations: 1 Cancer Research Institute, Beth Israel Deaconess Can-cer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; 2 Department of Medicine; Divisions of 3 Hematology/Oncology and 4 Signal Transduction, Department of Medicine; 5 Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center; 6 Department of Pathology, Brigham and Women’s Hospital; 7 Depart-ment of Systems Biology, Harvard Medical School; 8 Center for Molecular Oncologic Pathology; 9 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; 10 Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts; and 11 On leave of absence: Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseg-uer, Murcia, Spain

Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).

E. González-Billalabeitia and N. Seitzer contributed equally to this work.

Corresponding Author: Pier Paolo Pandolfi , Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Dea-coness Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215. Phone: 617-735-2121/2141; Fax: 617-735-2120; E-mail: [email protected]

doi: 10.1158/2159-8290.CD-13-0230

©2014 American Association for Cancer Research.

INTRODUCTION

There is an urgent need for the development of novel suc-

cessful strategies for advanced prostate cancer to improve

patient outcomes ( 1, 2 ).

The loss of at least one allele of phosphatase and tensin

homolog deleted on chromosome 10 ( PTEN ) is observed in

more than 40% of prostate cancers ( 3, 4 ). As a lipid phos-

phatase, PTEN suppresses the activation of the PI3K–AKT

signaling cascade that is a central proto-oncogenic signaling

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 2: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

AUGUST 2014�CANCER DISCOVERY | 897

PI3K and PARP Inhibition in PTEN-Defi cient Prostate Cancer RESEARCH BRIEF

hub in prostate cancer development ( 5–9 ). Notably, PTEN

also exerts tumor-suppressive functions in the nucleus by reg-

ulating genomic instability, and DNA-repair defects driven

by PTEN loss have been proposed as a rationale for the sen-

sitivity to PARP inhibitors comparable with the enhanced

sensitivity of BRCA -defi cient cancers ( 5 , 10–12 ). On this basis,

we therefore aimed to test whether PARP inhibition, alone

or combined with PI3K inhibition, would represent effective

therapeutic modalities in prostate cancer.

RESULTS Common Genetic Alterations in Prostate Cancer Morph Senescence into Apoptosis in Response to PARP Inhibition

We fi rst aimed to determine whether common genetic altera-

tions in prostate cancer respond toward olaparib. To mimic

different stages of prostate cancer progression, we selected

Pten +/− (early stage), Pten Lx/Lx (advanced hormone sensitive),

and Pten Lx/Lx ; Trp53 Lx/Lx mouse embryonic fi broblasts (MEF;

advanced hormone insensitive; Supplementary Fig. S1A) to per-

form a cell proliferation assay upon treatment with 10 μmol/L

olaparib. Interestingly, MEFs of all genotypes ( Fig.  1A–C )

showed a signifi cant growth inhibition upon exposure to ola-

parib. Surprisingly, in contrast with Pten Lx/Lx ; Trp53 Lx/Lx MEFs,

Pten +/− and Pten Lx/Lx MEFs showed a robust senescence response

(Supplementary Fig.  S1B and S1C) that was signifi cant and

dose dependent in Pten +/− ( Fig. 1D ) as well as Pten Lx/Lx ( Fig. 1E )

compared with wild-type MEFs. Strikingly and surprisingly,

further analysis revealed that in contrast to Pten Lx/Lx MEFs,

Pten Lx/Lx ; Trp53 Lx/Lx MEFs showed a signifi cant increase in apop-

tosis rather than senescence (Supplementary Fig. S1C and 1F )

as analyzed by Annexin V staining (Supplementary Fig.  S1D

and S1E) and detection of cleaved caspase-3/7 ( Fig. 1G ).

Western blot analysis of Pten Lx/Lx and Pten Lx/Lx ; Trp53 Lx/Lx

MEFs treated with increasing concentrations of olaparib

revealed that whereas Pten Lx/Lx MEFs showed a further increase

in p53 protein levels, both Pten Lx/Lx and Pten Lx/Lx ;Trp53 Lx/Lx

MEFs showed increased DNA damage as visualized by γH2AX

staining ( Fig. 1H ). This analysis demonstrates that the senes-

cence response in Pten Lx/Lx is likely driven by the induction of

p53, as previously described ( 13 ). However, the concomitant

loss of p53 induces increased DNA damage that in turn

morphs this phenotype into an apoptotic response.

PARP Inhibition Induces a Differential Response In Vivo with a Modest Effect on Overall Tumor Response

To validate our fi ndings in vivo , we next studied the sen-

sitivity of genetically engineered mouse models (GEMM) of

Figure 1.   Common genetic alterations in prostate cancer morph senescence into apoptosis in response to PARP inhibition. Growth of Pten +/− (A), Pten Lx/Lx (B), and Pten Lx/Lx ;Trp53 Lx/Lx (C) MEFs in the presence of 10 μmol/L olaparib (**, P < 0.0032; ***, P < 0.0001). D, quantifi cation of SA-β-gal positiv-ity in Pten +/− and Pten Lx/Lx MEFs. (continued on following page)

A

0 2 4 6

1

2

3

4

5

6

**

MEF Pten+/–

Olaparib 10 μmol/L

MEF Pten+/– DMSO

MEF PtenLx/Lx;Trp53Lx/Lx

Olaparib 10 μmol/L

MEF Pten+/–

MEF PtenLx/Lx;Trp53Lx/Lx DMSOMEF WT

MEF PtenLx/Lx

Olaparib 10 μmol/L

MEF PtenLx/Lx DMSO

Days

0 7 9

15

10

5

0

***

DC

Days

970

2.5

2

1.5

0

3

**N

orm

aliz

ed g

row

th (

AU

)N

orm

aliz

ed g

row

th (

AU

)

SA

-β-G

ala

cto

sid

ase (

%)

Norm

aliz

ed g

row

th (

AU

)

B

Olaparib (μmol/L)

0 1 3 10

60

40

20

0

*

*

Days

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 3: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

898 | CANCER DISCOVERY�AUGUST 2014 www.aacrjournals.org

González-Billalabeitia et al.RESEARCH BRIEF

prostate cancer toward PARP inhibition. Similar to our in vitro

analysis, we enrolled Pten +/− , Pten Lx/Lx ; Probasin-Cre (referred to

as Pten pc−/− ) and Pten Lx/Lx ; Trp53 Lx/Lx Probasin-Cre (referred to

as Pten pc−/− ; Trp53 pc−/− ) mice. In line with the data observed in

MEFs, pharmacologic inhibition of PARP induced a strong

and signifi cant induction of senescence in Pten +/− (Supplemen-

tary Fig. S2A) and Pten pc−/− ( Fig. 2A and B ) models compared

with vehicle-treated controls. In Pten pc−/− mice, the senescence

response was accompanied by increased DNA damage as

analyzed by γH2AX staining of treated prostate tumors (Sup-

plementary Fig.  S2B). Histologic analysis of Pten pc−/− tumors

treated with olaparib revealed a modest decrease in high-grade

prostatic intraepithelial neoplasia (HGPIN; Fig. 2C ). However,

this trend did not reach statistical signifi cance.

Next, we tested whether Pten pc−/− ; Trp53 pc−/− mice showed

a similar apoptotic response upon treatment with olaparib

as observed in vitro . In line with the in vitro data, olaparib

treatment increased γH2AX in dorsolateral prostate (DLP)

tumors of Pten pc−/− ; Trp53 pc−/− mice (Supplementary Fig. S2C).

Surprisingly, macroscopic analysis and cytokeratin 8 stain-

ing (luminal cells) of olaparib-treated prostates revealed that

more glands were lined by a single layer compared with the

vehicle control ( Fig.  2D and Supplementary S3A). In addi-

tion, analysis of cytokeratin 14 showed a reduction of the

intermediate basal cell population in single-layered glands,

suggesting a certain degree of normalization after treat-

ment (Supplementary Fig.  S3B). Terminal deoxynucleotidyl

transferase–mediated dUTP nick end labeling (TUNEL) and

caspase-3 staining further revealed a signifi cant increase in

apoptotic cells upon olaparib treatment ( Fig.  2E and F and

Supplementary S2D). However, similar to Pten pc−/− mice,

histologic analysis of Pten pc−/− ; Trp53 pc−/− tumor reduction

after drug treatment did not reach statistical signifi cance

( Fig.  2G ), suggesting that single-agent olaparib treatment

is not suffi cient to induce a robust antitumor response in

these models. Interestingly, mass spectrometry analysis of ola-

parib in prostates revealed that only approximately 2 μmol/L of

the drug is delivered into the individual lobes, an amount that

is signifi cantly lower when compared with the dose used in

our in vitro studies (Supplementary Fig. S3C). This marked dif-

ference in drug concentration may in turn provide one possi-

ble explanation for the limited overall tumor response in vivo .

Hyperactivation of the PI3K–AKT Pathway Affects Sensitivity toward Olaparib

On the basis of our data in mice, we sought to determine

whether olaparib could trigger a similar response in human

prostate cancer cell lines. To this end, we engineered LNCaP

MEF PtenLx/Lx;Trp53Lx/Lx

MEF PtenLx/Lx

MEF PtenLx/Lx

MEF Trp53Lx/Lx

MEF PtenLx/Lx;Trp53Lx/Lx

PtenLx/Lx PtenLx/Lx;Trp53Lx/Lx

MEF WT

MEF PtenLx/Lx

MEF WT

ES

A-β

-Gala

cto

sid

ase (

%)

Cle

aved c

aspase-3

/7 (

fc)

3

1

2

2.5

1.5 ***

**

***

WB:PAR

WB:p53

WB:PTEN

WB:Actin

WB:γ-H2AX

WB:PARP

60

40

20

0

SA

-β-G

ala

cto

sid

ase (

%)

60

40

20

0

*

*

F

Olaparib (μmol/L)

0 1 3 10

Olaparib (μmol/L)

0 1 3 10

Olaparib (μmol/L)

Olaparib (μmol/L)

0

0 1 3 10 0 1 3 10

1 3 10

G

*

*

**

H

Figure 1. (Continued) E, MEFs upon increasing doses of olaparib at day 4 (*, P < 0.05). F, quantifi cation of SA-β-gal positivity in Pten Lx/Lx compared with Pten Lx/Lx ;Trp53 Lx/Lx MEFs upon increasing doses of olaparib at day 4 (*, P < 0.05). G, quantifi cation of caspase-3/7 activity after treatment with 10 μmol/L olaparib for 48 hours (**, P < 0.01; ***, P < 0.001). H, Western blot (WB) analysis of Pten Lx/Lx and Pten Lx/Lx ;Trp53 Lx/Lx MEFs after 3 days of olaparib treatment. WT, wild-type; fc, fold change.

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 4: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

AUGUST 2014�CANCER DISCOVERY | 899

PI3K and PARP Inhibition in PTEN-Defi cient Prostate Cancer RESEARCH BRIEF

Figure 2.   PARP inhibition induces a differential response in vivo with a modest effect on overall tumor response. A, SA-β-gal staining in prostates of 8-week-old Pten pc−/− mice upon olaparib ( n = 3) or vehicle ( n = 3) treatment for 2 weeks. B, quantifi cation of SA-β-gal positivity from A (*, P = 0.0419). C, histopathologic analysis of HGPIN status from A. D, hematoxylin and eosin (H&E) staining of DLP tumors from 4-month-old Pten pc−/− ;Trp53 pc−/− mice upon olaparib ( n = 3) or vehicle ( n = 3) treatment for 1 week. E, TUNEL staining to visualize apoptosis induction in D and F , its quantifi cation (***, P = 0.0006). DAPI, 4,6-diamidino-2-phenylindole. G, histopathologic analysis of HGPIN status from D. ns, not signifi cant.

D

A B

SA

-β-G

al sta

inin

g (

%)

100

0

80

60

40

*

20

Olaparib

50 mg/kg

Vehicle

E

Ptenpc–/–

Ptenpc–/–;Trp53pc–/–

Pte

npc–/–

;Trp

53pc–/–

F G

Vehic

leO

laparib 5

0 m

g/k

g

Grade 1

Vehicle Olaparib

50 mg/kg

100

80

60

40

20

0

Grade 3

Grade 2

HG

PIN

(%

)

ns

C

Olaparib

50 mg/kgVehicle

Grade 3

Grade 2

HG

PIN

(%

)

ns

Vehic

leO

laparib

50 m

g/k

g

H&

ES

A-β

-Gal

×4

×4

×4

×4

×20

×20

×20

×20

H&

ES

A-β

-Gal

TU

NE

L p

ositiv

ity (

%)

***

5

15

0

10

Olaparib

50 mg/kg

Vehicle

Vehicle Olaparib50 mg/kg

DA

PI

Flu

ore

scein

ME

RG

E×100

×100

×100

×100

×100

×100

×4

×4

×20

×20

×40

×40

0

20

40

60

80

100

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 5: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

900 | CANCER DISCOVERY�AUGUST 2014 www.aacrjournals.org

González-Billalabeitia et al.RESEARCH BRIEF

cells that lack PTEN to additionally lose p53 function by

overexpressing either a p53 short hairpin construct (Sup-

plementary Fig.  S4A) or dominant-negative p53 (p53-DN;

Supplementary Fig. S4B). Similar to MEFs, olaparib induced

senescence in a p53-profi cient setting that was blunted upon

loss of p53 ( Fig.  3A ). In contrast, LNCaP cells expressing

p53-DN did not show differential sensitivity to olaparib treat-

ment. Furthermore, only high olaparib concentrations (10

μmol/L) induced a strong growth inhibition overall ( Fig. 3B ),

suggesting mechanisms limiting olaparib effi cacy beyond the

limited uptake observed in vivo .

We therefore investigated whether classical survival sig-

naling such as the PI3K–AKT pathway might be superacti-

vated upon olaparib exposure and therefore analyzed AKT

activation upon olaparib treatment in vitro ( 7 , 14–16 ). Indeed,

Pten Lx/Lx ; Trp53 Lx/Lx MEFs ( Fig. 3C ) and LNCaP cells ( Fig. 3D )

hyperactivated AKT upon exposure to olaparib, suggesting

that AKT could affect the response to olaparib. Indeed,

Figure 3.   Hyperactivation of the PI3K–AKT pathway affects sensitivity toward olaparib. A, quantifi cation of SA-β-gal positivity toward increasing doses of olaparib after 4 days in LNCaP cells upon stable p53 knockdown ( n = 4; *, P = 0.01; **, P < 0.008; ***, P = 0.0006). B, quantifi cation of growth inhi-bition toward increasing doses of olaparib after 3 days in LNCaP cells overexpressing dominant-negative (DN) p53 ( n = 2). C, Western blot (WB) analysis of AKT activation in Pten Lx/Lx ;Trp53 Lx/Lx MEFs after 24 hours and D , LNCaP cells after 72 hours cells of olaparib treatment. E, quantifi cation of growth inhibition upon olaparib treatment (5 μmol/L) after AKT1 knockdown in LNCaP cells. F, Western blot analysis of apoptosis induction in LNCaP after AKT1 knockdown upon olaparib treatment for 72 hours. G, Western blot analysis of apoptosis induction in LNCaP upon olaparib, BKM120, or combination treat-ment for 72 hours; OL, olaparib; BK, BKM120.

A B

C

LNCap

WB: PARP

WB: Cleaved

PARP

WB: pAKT

(T308)

WB: AKT

WB: Actin

MEF Ptenlx/lx;Trp53 lx/lx

40

20

10

0

30

Olaparib (μmol/L)

0 1 3 10

LNCaP shp53LNCaP Mock

SA

-β-G

ala

cto

sid

ase (

%)

D E

G

1 1.5 3.2

1 2.6 3.6

LNCaP p53 DN

LNCaP empty

Norm

aliz

ed g

row

th (

%)

0 1 3 10

LNCap

DM

SO

DM

SO

OL

3

DM

SO

OL

3

OL

10

BK

0.5

BK

1

OL 3

DM

SO

BK

0.5

BK

1

OL 3

OL 5

OL 1

0B

K 0

.5/O

L 3

BK

0.5

/OL 5

DM

SO

BK

0.5

BK

1O

L 3

WB: Cleaved

PARP

WB: pAKT

(T308)

WB: AKT

WB: Actin

1 3.7

1 1.6

Olaparib (μmol/L)

Gro

wth

inhib

itio

n (

%) LNCaP shAKT1

LNCaP Mock

3 6

*P = 0.049

**P = 0.0027

Days

LNCapshControl shAKT1

WB: pAKT

(T308)

WB: AKT

WB: Actin

WB: Cleaved

PARP

F

*

1 0.9 3.2 2.9 1 6.0 8.3 7.8

WB: Cleaved

PARP

WB: pAKT

(T308)

WB: AKT

WB: Actin

1 3.7 7.6 2.1 4.9 8.9 8.9 13

****

*

***

0

50

100

150

0

10

20

30

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 6: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

AUGUST 2014�CANCER DISCOVERY | 901

PI3K and PARP Inhibition in PTEN-Defi cient Prostate Cancer RESEARCH BRIEF

knockdown of AKT1, the major AKT isoform, signifi cantly

increased growth inhibition upon PARP inhibition (Supple-

mentary Fig. S4C and 3E ) and induced apoptosis, as shown

by increased PARP cleavage ( Fig. 3F ).

To investigate the mechanism of AKT hyperactivation, we

made use of the dual PI3K inhibitor BKM120 that effi ciently

blocked AKT activation in LNCaP cells ( Fig. 3F ). Strikingly,

PI3K inhibition was able to restore pAKT to DMSO control

levels in olaparib-treated cells, suggesting that the major acti-

vator of AKT upon PARP inhibition is indeed PI3K ( Fig. 3G ).

Importantly, analysis of cleaved PARP demonstrated that the

combination of PARP and PI3K inhibition shows synergistic

apoptosis induction, suggesting effi cacy for a combinatorial

treatment strategy in prostate cancer ( Fig. 3G ).

Combination of PARP and PI3K Inhibitors as a Novel Therapeutic Approach

On the basis of our fi ndings, we next aimed to investi-

gate whether a combination treatment including PARP and

PI3K inhibition would be synergistic in vitro . To this end, we

treated Pten Lx/Lx ; Trp53 Lx/Lx MEFs and human prostate cancer

cells with either single or combination therapy (Supplemen-

tary Fig.  S4D–S4G). Indeed, the combination signifi cantly

increased growth suppression compared with the single treat-

ments in all tested cell lines.

These promising in vitro data prompted us to next evaluate

the effi cacy of the combinatorial inhibition of PARP and PI3K

in our Pten pc−/− ; Trp53 pc−/− model of highly aggressive prostate

cancer (Supplementary Fig. S5A). In vivo effi cacy of control,

single agent, and the combination arm was assessed by MRI

analysis for the dorso-lateral prostate (DLP) and the anterior

prostate (AP; Supplementary Fig. S5B and S5C), followed by

the analysis of survival and histopathologic differences.

To assess the effi cacy of PARP and PI3K inhibition in vivo ,

we fi rst followed tumor progression by MRI ( Fig. 4A ). Inter-

estingly, treatment with olaparib or BKM120 showed tumor

stabilization but eventually tumors regrew due to potential

resistance mechanisms. Strikingly, the combination of the

two compounds potentiated tumor stabilization and induced

robust tumor regression. These data indicate that the combi-

natorial treatment of PI3K and PARP inhibition is synergistic

to the single-compound arms in vivo . In contrast with the

DLP, only slight growth suppression was seen in APs that was

potentiated in the combination, suggesting a similar coop-

eration as seen in the DLP albeit with a much lower effi cacy

(Supplementary Fig. S6A).

We further analyzed tumor specimens of all treatment

arms after 1 to 4 weeks by histopathology ( Fig. 4B and C and

Supplementary S6B and S6C). Surprisingly, despite the initial

response toward olaparib observed at 1 week, histopatho-

logic analysis and cytokeratin 8 staining revealed no differ-

ence in the occurrence of HGPIN after 4 weeks compared

with the vehicle control. In contrast, the BKM120 treatment

arm triggered a clear decrease in HGPIN. Importantly, the

combinatorial treatment signifi cantly potentiated this effect,

clearly showing cooperation between the two compounds.

Furthermore, cytokeratin 14 staining in the DLPs treated

with the combination showed a clear reduction in the inter-

mediate basal cell population that is not apparent in the

vehicle- or single-arm–treated tumors. These data indicate a

trend toward normalization of the glands specifi cally in the

combination treatment.

In line with these data, progression-free survival (PFS)

was markedly improved in the combinatorial treatment arm

compared with the single and control arms ( Fig. 4D and Sup-

plementary S7A and S7B). However, and coherent with the

lack of response in the AP, overall survival (OS) analysis did

not reveal a striking difference between the single and combi-

nation treatment arms (Supplementary Fig. S7C).

Finally, to determine whether the data obtained in our

GEMMs could be confi rmed in human cell lines, we per-

formed xenograft studies using PC3 prostate cancer cell lines

( Fig.  4E ). Similar to our GEMMs, PC3 xenografts displayed

a signifi cant and synergistic response when treated with the

combination compared with the single agents. Collectively,

these data strongly support the notion of a marked coopera-

tive effect between the two drugs for the treatment of pros-

tate cancer.

DISCUSSION This study allowed us to reach a number of important

conclusions:

First, we demonstrated, in vitro as well as in vivo , that PTEN

defi ciency triggers sensitivity toward PARP inhibition with

differential responses. In a p53-profi cient setting, loss of

PTEN triggers a senescence response upon PARP inhibition

that is morphed into apoptosis upon loss of p53.

Second, a robust response toward PARP inhibition was

observed only at very high concentrations of olaparib in cell

lines, whereas mouse DLPs treated with olaparib displayed

no signifi cant difference in HGPIN grades. In line with these

observations, we fi nd that olaparib reaches only suboptimal

concentration in vivo in the prostate.

Third, we found that olaparib triggers the activation of

AKT, a classical cell survival mechanism in response to cellu-

lar stresses ( 7 , 14–16 ). These data demonstrated that olaparib

treatment on its own is insuffi cient to trigger a signifi cant

tumor response in the prostate due to a number of reasons.

Fourth, we showed that PI3K inhibition by BKM120 signif-

icantly increased sensitivity toward olaparib both in GEMMs

and xenograft models. Importantly, PC3 xenografts showed a

clear synergistic effect of growth inhibition in the combina-

tion arm in vivo , validating the effi cacy of the combination

in a human setting. Intriguingly, these data, especially in

PC3 xenografts, are in clear contrast with the in vitro data

that rather suggested an additive effect of the combination.

These results could be explained by the profound differences

in nutrients and growth factors in the two experimental

settings, or by the fact that the tumor microenvironment/

metabolism may also affect the effi cacy of the treatments, as

previously suggested ( 17–19 ).

Finally, despite these striking fi ndings, combinatorial PARP

and PI3K inhibition prolonged PFS and OS signifi cantly only

when compared to vehicle, but not to single-agent–treated

animals. However, these data are potentially infl uenced by the

general characteristics of Pten pc−/− ; Trp53 pc−/− prostate tumors

( 13 ). Cancers in the APs in this model are intrinsically resist-

ant to therapy, including androgen deprivation, and showed

only a slight growth inhibition in all of the treatment arms

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 7: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

902 | CANCER DISCOVERY�AUGUST 2014 www.aacrjournals.org

González-Billalabeitia et al.RESEARCH BRIEF

Figure 4.   Combination of PARP and PI3K inhibitors as a novel therapeutic approach. A, DLP tumor volume change of Pten pc−/− ;Trp53 pc−/− mice upon vehi-cle ( n = 7), olaparib ( n = 8), BKM120 ( n = 6), or combination ( n = 9) treatment. B, histopathologic analysis of HGPIN status from vehicle ( n = 3; 1 and 4 weeks pooled), olaparib ( n = 3), BKM120 ( n = 3), or combination ( n = 3) treatment for 4 weeks (*, P < 0.03). C, H&E staining from A. D, PFS of Pten pc−/− ;Trp53 pc−/− mice upon vehicle ( n = 7), olaparib ( n = 8), BKM120 ( n = 6), or combination ( n = 9) treatment. ns, not signifi cant. E, tumor growth of PC3 xenografts upon vehicle ( n = 5), olaparib ( n = 5), BKM120 ( n = 4), or combination ( n = 5; **, P < 0.009) treatment; olaparib, 50 mg/kg; BKM120, 30 mg/kg.

A

0

–50

50

–100

100

150

200

250

300

350D

LP

tum

or

volu

me c

hange (

%)

28 56 84 112 140

Days

BVehicle

BKM120

Olaparib + BKM120

Olaparib

C

Pte

npc–/–

;Trp

53pc–/–

**

0

50

100

150

Vehicle BKM120 Olaparib +

BKM120Olaparib

Perc

enta

ge o

f gla

nds (

%)

Normal

PIN I

PIN II

PIN III

PIN IV

0 50 100 150

50

100

0

PFS

(days) P

Vehicle 15

Olaparib 15 ns

BKM120 27 0.0016

Olaparib +

BKM120 40 <0.0001

Days1 3 5 8 10 12 15

Vehicle

Olaparib (50 mg/kg)

BKM120 (30 mg/kg)

Olaparib (50 mg/kg)/

BKM120 (30 mg/kg)

**

**

**

D E

Olaparib +

BKM120 BKM120Vehicle Olaparib

×40

×2

×10

×40

×10

×2

×40

×10

×2

×40

×10

×2

300

400

500

600

700

800

900

1,000

1,100

Days

PF

S (

%)

Tum

or

are

a (

cm

2)

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 8: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

AUGUST 2014�CANCER DISCOVERY | 903

PI3K and PARP Inhibition in PTEN-Defi cient Prostate Cancer RESEARCH BRIEF

( 20 ). However, the rapid growth of the APs strongly infl u-

ences the survival of Pten pc ; Trp53 pc−/− mice and, thus, strongly

affects the assessment and interpretation of OS.

In conclusion, we show that PTEN -defi cient prostate can-

cer is sensitive to PARP inhibition. Because of AKT hyperac-

tivation and poor pharmacokinetics, the in vivo effi cacy of

olaparib is modest, but concomitant suppression of PARP

and PI3K results in a cooperative antitumor effect. Thus,

the combination of PI3K and PARP inhibitors represents a

promising therapeutic approach for the treatment of PTEN -

defi cient prostate tumors.

METHODS Cells

Pten +/− , Pten Lx/Lx , and Pten Lx/Lx ; Trp53 Lx/Lx MEFs were prepared as

previously described. For the excision of target genes, MEFs were

retrovirally infected with pMSCV–Cre–PURO–IRES–GFP ( 13 ).

Human cell lines were obtained from the ATCC and cultured

according to the manufacturer’s instructions. Cell lines were

tested for mycoplasma (MycoAlert; Lonza), but not further authen-

ticated.

Cell Proliferation , Senescence , and Apoptosis Assays To study proliferation, cells were plated in a 12-well plate (MEFs at

1.7 × 10 4 cells/well and prostate cancer cells at 1 × 10 5 cells/well). After

2 to 9 days, cells were washed with PBS, fi xed with 10% paraformalde-

hyde, and stained with crystal violet. Crystal violet was extracted with

10% acetic acid and absorbance was detected at 595 nm.

To determine senescence, cells were plated in a 6-well plate (MEFs

at 3.4 × 10 4 cells/well and LNCaP at 1.7 × 10 4 cells/well). SA-β-Gal was

detected after 4 days with the Senescence Detection Kit (Calbiochem)

following the manufacturer’s instructions. For quantifi cation, more

than 200 cells per sample were counted.

For prostate tissue, frozen sections (6 mm) were stained for SA-β-

Gal (Calbiochem) following the manufacturer’s instructions. For

quantifi cation, slides were digitalized and analyzed with the Aperio

Spectrum ImageScope software.

Apoptosis was determined by the Apo-ONE caspase-3/7 assay

(Promega) and additionally analyzed by a dual Annexin V–APC/7-

AAD (BD Biosciences) staining using fl ow cytometry following the

manufacturer’s instructions.

For in vivo apoptosis, samples were analyzed with the In Situ Cell

Death Detection Kit (Roche) following the manufacturer’s instruc-

tions. For the quantifi cation of positive TUNEL staining, a total of

500 cells were counted from fi ve different fi elds.

Western Blot Analysis and Immunohistochemistry For Western blotting, cell lysates were prepared with RIPA buffer

or NP40 buffer (Boston BioProducts) supplemented with protease

(Roche) and HALT phosphatase inhibitor cocktail (Thermo Scien-

tifi c), and subsequently subjected to SDS–Gel separation (Invitrogen)

and Western blotting. The following antibodies were used from Cell

Signaling Technology: anti-γH2AX, anti-AKT1, anti-mouse PTEN, anti-

mouse p53, anti-AKT, anti-phospho threonine 308 and anti–phospho-

serine 473 anti-PARP, anti–cleaved PARP or human p53 (DO1; Santa

Cruz Biotechnology), anti-PAR (BD Biosciences), and anti–β-actin (AC-

74; Sigma). Western blots were quantifi ed using ImageJ software.

For IHC, tissues were fi xed in 4% paraformaldehyde and embedded

in paraffi n in accordance with standard procedures. Hematoxylin

and eosin (H&E) staining of sections was performed by the Histology

Core at the Beth Israel Deaconess Medical Center (BIDMC; Boston,

MA).

Drugs For in vitro studies, drugs were resuspended in DMSO (olaparib;

LC Laboratories, 10 −2 mol/L; BKM120, Novartis Pharma AG,

10 −2 mol/L). For in vivo studies, olaparib in DMSO (50 mg/mL) was

diluted in vehicle solution [10% 2-hydroxyl-propyl-βCyclodextrine

(Sigma) in PBS, 10 μL/g body] and administered daily by i.p. injection

at a dose of 50 mg/kg. BKM120 was resuspended in methylcelullose

solution at a fi nal concentration of 6 mg/mL and administered daily

at a dose of 30 mg/kg by oral gavage.

Mice , Xenografts , and In Vivo Drug Treatments All mice were maintained in the animal facilities of BIDMC/Har-

vard Medical School in accordance with institutional rules and ethi-

cal guidelines for experimental animal care. All animal experiments

were approved by the BIDMC Institutional Animal Care and Use

Committee protocol 066-2011 ( 8 , 13 , 21 ).

For drug treatments, Pten +/− (30-week-old) and Pten pc−/− (10-week-

old) mice were treated with olaparib over 2 weeks. Pten pc−/− ; Trp53 pc−/−

(4-month-old) mice were treated with olaparib over 1 week.

For the combination treatment of olaparib and BKM120,

Pten pc−/− ; Trp53 pc−/− (4-month-old) mice were evaluated by MRI for the

presence of prostate tumors and subsequently enrolled in the following

treatment arms: vehicle, olaparib, BKM120, or combination at the same

doses. Mice were imaged with MRI before treatment, monitored by MRI

until tumor progression, and followed for survival analysis. Tumor pro-

gression was defi ned as an increase of >50% in tumor volume.

Xenografts were performed with PC3 cells. A total of 1 × 10 6 cells

were injected subcutaneously in the fl anks NCr nude mice (Taconic).

Mice were enrolled at a tumor size of 300 m 3 in the following treat-

ment arms: vehicle, olaparib, BKM120, or combination at the same

doses. Mice were euthanized after 2 weeks of treatment, and tumors

were subjected to further analysis.

MRI Analysis Mouse prostate images were acquired on an ASPECT Model M2

1T tabletop MRI scanner (ASPECT Magnet Technologies Ltd.) and

imaged as previously described ( 20 ). Tumor volume quantifi cation

was performed as previously described ( 22 ).

Statistical and Histological Analyses For all statistical analyses, GraphPad Prism 6 software was used,

and values of P < 0.05 were considered statistically signifi cant. Sur-

vival was determined by Kaplan–Meier curves. The Mantel–Cox test

was used to determine signifi cance between survival curves. All other

statistical analyses were done by an unpaired Student t test.

Scoring of HGPIN status was performed as previously described

( 23 ). At least three samples were analyzed at the accorded time points.

For the preclinical assessment of the combinatorial treatment in

Pten pc−/− ; Trp53 pc−/− mice, at least three mice were analyzed after 4

weeks of treatment except the vehicle control, in which mice were

pooled from 1- and 4-week treatment.

Disclosure of Potential Confl icts of Interest L.C. Cantley has received a commercial research grant from Glaxo-

SmithKline and is a consultant/advisory board member for Novartis.

No potential confl icts of interest were disclosed by the other authors.

The Editor-in-Chief of Cancer Discovery (L.C. Cantley) is an author

of this article. In keeping with the AACR’s Editorial Policy, the paper

was peer reviewed and an AACR journal editor not affi liated with

Cancer Discovery rendered the decision concerning acceptability.

Authors’ Contributions Conception and design: E. González-Billalabeitia, N. Seitzer,

A. Patnaik, C. Nardella, L.C. Cantley, P.P. Pandolfi

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 9: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

904 | CANCER DISCOVERY�AUGUST 2014 www.aacrjournals.org

González-Billalabeitia et al.RESEARCH BRIEF

Development of methodology: E. González-Billalabeitia, A. Patnaik,

X.-S. Liu, A. Papa, M. Chen, M. Loda, J.M. Asara

Acquisition of data (provided animals, acquired and managed

patients, provided facilities, etc.): E. González-Billalabeitia, A. Pat-

naik, R.M. Hobbs, M. Chen, C. Ng, K.A. Webster, M. Loda, J.M. Asara,

C. Nardella, J.G. Clohessy

Analysis and interpretation of data (e.g., statistical analysis,

biostatistics, computational analysis): E. González-Billalabeitia,

N. Seitzer, M.S. Song, A. Patnaik, R.M. Hobbs, S. Signoretti, M. Loda,

J.M. Asara, J.G. Clohessy, P.P. Pandolfi

Writing, review, and/or revision of the manuscript: E. González-

Billalabeitia, N. Seitzer, M.S. Song, A. Patnaik, A. Lunardi, M. Loda,

J.M. Asara, L.C. Cantley, P.P. Pandolfi

Administrative, technical, or material support (i.e., reporting or

organizing data, constructing databases): E. González-Billalabei-

tia, M.S. Song, A. Patnaik, M.T. Epping

Study supervision: E. González-Billalabeitia, N. Seitzer, A. Patnaik,

J.G. Clohessy, P.P. Pandolfi

Carried out experiments: S.J. Song

Provided histology and immunohistochemistry: K.A. Webster

Acknowledgments The authors thank the current members of the Pandolfi laboratory

for critical discussion, Thomas Garvey for insightful editing, and

Min Yuan and Susanne Breitkopf for help with mass spectrometry

experiments.

Grant Support This work has been supported through a Mouse Models of Human

Cancers Consortium/NCI grant (RC2 CA147940-01), R01 GM41890,

and P01 CA089021 (to P.P. Pandolfi and L.C. Cantley), and NIH grant

5P01CA120964-04 and NIH Dana-Farber/HCC Cancer Center Sup-

port Grant 5P30CA006516-46 (to J.M. Asara). E. González-Billalabe-

itia has been supported by a Bolsa de ampliación de estudios (BAE)

from the Instituto de Salud Carlos III (Spain).

Received May 20, 2013; revised May 1, 2014; accepted May 19,

2014; published OnlineFirst May 27, 2014.

REFERENCES 1. Walsh PC , DeWeese TL , Eisenberger MA . Clinical practice. Localized

prostate cancer . N Engl J Med 2007 ; 357 : 2696 – 705 .

2. Carter HB , Kettermann A , Warlick C , Metter EJ , Landis P , Walsh PC ,

et al. Expectant management of prostate cancer with curative intent:

an update of the Johns Hopkins experience . J Urol 2007 ; 178 : 2359 – 65 .

3. Li J . PTEN, a putative protein tyrosine phosphatase gene mutated in

human brain, breast, and prostate cancer . Science 1997 ; 275 : 1943 – 7 .

4. Steck PA , Pershouse MA , Jasser SA , Yung WK , Lin H , Ligon AH , et al.

Identifi cation of a candidate tumour suppressor gene, MMAC1, at

chromosome 10q23.3 that is mutated in multiple advanced cancers .

Nat Genet 1997 ; 15 : 356 – 62 .

5. Song MS , Salmena L , Pandolfi PP . The functions and regulation of

the PTEN tumour suppressor . Nat Rev Mol Cell Biol 2012 ; 13 : 283 – 96 .

6. Engelman JA , Luo J , Cantley LC . The evolution of phosphatidylinosi-

tol 3-kinases as regulators of growth and metabolism . Nat Rev Genet

2006 ; 7 : 606 – 19 .

7. Manning BD , Cantley LC . AKT/PKB signaling: navigating down-

stream . Cell 2007 ; 129 : 1261 – 74 .

8. Di Cristofano A , Pesce B , Cordon-Cardo C , Pandolfi PP . Pten is essen-

tial for embryonic development and tumour suppression . Nat Genet

1998 ; 19 : 348 – 55 .

9. Wu X , Senechal K , Neshat MS , Whang YE , Sawyers CL . The PTEN/

MMAC1 tumor suppressor phosphatase functions as a negative regu-

lator of the phosphoinositide 3-kinase/Akt pathway . Proc Natl Acad

Sci U S A 1998 ; 95 : 15587 – 91 .

10. Puc J , Keniry M , Li H , Pandita T , Choudhury A , Memeo L , et al. Lack

of PTEN sequesters CHK1 and initiates genetic instability . Cancer

Cell 2005 ; 7 : 193 – 204 .

11. Mendes-Pereira AM , Martin SA , Brough R , McCarthy A , Taylor JR ,

Kim J-S , et al. Synthetic lethal targeting of PTEN mutant cells with

PARP inhibitors . EMBO Mol Med 2009 ; 1 : 315 – 22 .

12. Bryant HE , Schultz N , Thomas HD , Parker KM , Flower D , Lopez E ,

et al. Specifi c killing of BRCA2-defi cient tumours with inhibitors of

poly(ADP-ribose) polymerase . Nature 2005 ; 434 : 913 – 7 .

13. Chen Z , Trotman LC , Shaffer D , Lin H-K , Dotan ZA , Niki M , et al.

Crucial role of p53-dependent cellular senescence in suppression of

Pten-defi cient tumorigenesis . Nat Cell Biol 2005 ; 436 : 725 – 30 .

14. Marte BM , Downward J . PKB/Akt: connecting phosphoinositide

3-kinase to cell survival and beyond . Trends Biochem Sci 1997 ; 22 :

355 – 8 .

15. Hers I , Vincent EE , Tavaré JM . Akt signalling in health and disease .

Cell Signal 2011 ; 23 : 1515 – 27 .

16. Tapodi A . Pivotal role of Akt activation in mitochondrial protection

and cell survival by poly(ADP-ribose)polymerase-1 inhibition in oxi-

dative stress . J Biol Chem 2005 ; 280 : 35767 – 75 .

17. Juvekar A , Burga LN , Hu H , Lunsford EP , Ibrahim YH , Balmana J ,

et al. Combining a PI3K inhibitor with a PARP inhibitor provides

an effective therapy for BRCA1-related breast cancer . Cancer Discov

2012 ; 2 : 1048 – 63 .

18. Schnell CR , Stauffer F , Allegrini PR , O’Reilly T , McSheehy PMJ ,

Dartois C , et al. Effects of the dual phosphatidylinositol 3-kinase/

mammalian target of rapamycin inhibitor NVP-BEZ235 on the

tumor vasculature: implications for clinical imaging . Cancer Res

2008 ; 68 : 6598 – 607 .

19. Yuan TL , Choi HS , Matsui A , Benes C , Lifshits E , Luo J , et al. Class 1A

PI3K regulates vessel integrity during development and tumorigen-

esis . Proc Natl Acad Sci 2008 ; 105 : 9739 – 44 .

20. Lunardi A , Ala U , Epping MT , Salmena L , Clohessy JG , Webster KA ,

et al. A co-clinical approach identifi es mechanisms and potential

therapies for androgen deprivation resistance in prostate cancer

2013 ; 45 : 747 – 55 .

21. Trotman LC , Niki M , Dotan ZA , Koutcher JA , Di Cristofano A , Xiao

A , et al. Pten dose dictates cancer progression in the prostate . Plos

Biol 2003 ; 1 : e9 .

22. Nastiuk KL , Liu H , Hamamura M , Muftuler LT , Nalcioglu O ,

Krolewski JJ . In vivo MRI volumetric measurement of prostate regres-

sion and growth in mice . BMC Urol 2007 ; 7 : 12 .

23. Park J-H , Walls JE , Galvez JJ , Kim M , Abate-Shen C , Shen MM , et al.

Prostatic intraepithelial neoplasia in genetically engineered mice . Am

J Pathol 2010 ; 161 : 727 – 35 .

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230

Page 10: Vulnerabilities of PTEN TP53 -Defi cient Prostate Cancers ......896 | CANCER DISCOVERYAUGUST 2014 Vulnerabilities of PTEN – TP53 -Defi cient Prostate Cancers to Compound PARP–PI3K

2014;4:896-904. Published OnlineFirst May 27, 2014.Cancer Discovery   Enrique González-Billalabeitia, Nina Seitzer, Su Jung Song, et al.  

PI3K Inhibition−Compound PARP -Deficient Prostate Cancers toTP53−PTENVulnerabilities of

  Updated version

  10.1158/2159-8290.CD-13-0230doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerdiscovery.aacrjournals.org/content/suppl/2021/03/04/2159-8290.CD-13-0230.DC2 http://cancerdiscovery.aacrjournals.org/content/suppl/2014/05/28/2159-8290.CD-13-0230.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerdiscovery.aacrjournals.org/content/4/8/896.full#ref-list-1

This article cites 22 articles, 7 of which you can access for free at:

  Citing articles

  http://cancerdiscovery.aacrjournals.org/content/4/8/896.full#related-urls

This article has been cited by 8 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerdiscovery.aacrjournals.org/content/4/8/896To request permission to re-use all or part of this article, use this link

on March 20, 2021. © 2014 American Association for Cancer Research. cancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 27, 2014; DOI: 10.1158/2159-8290.CD-13-0230