targeting dna damage response in prostate ......et al., 2003; yoshida et al., 2010), which inhibits...

20
Article Targeting DNA Damage Response in Prostate Cancer by Inhibiting Androgen Receptor-CDC6-ATR- Chk1 Signaling Graphical Abstract Highlights d CDC6 expression is increased during prostate cancer (PCa) progression d AR or CDC6 knockdown, together with AZD7762, suppresses TopBP1-ATR-Chk1 signaling d AR or CDC6 knockdown sensitizes PCa cells to AZD7762 d Enzalutamide and AZD7762 combination treatment generates synergistic therapeutic effects Authors Styliani Karanika, Theodoros Karantanos, Likun Li, ..., Wei Zhang, Shuhua Li, Timothy C. Thompson Correspondence [email protected] In Brief CDC6 is an androgen receptor (AR) target gene and an essential regulator of DNA replication and checkpoint activation. Karanika et al. show that combined inhibition of the AR and Chk1 signaling promotes DNA damage accumulation in prostate cancer cells to induce cell death, regardless of p53 status. Karanika et al., 2017, Cell Reports 18, 1970–1981 February 21, 2017 ª 2017 The Author(s). http://dx.doi.org/10.1016/j.celrep.2017.01.072

Upload: others

Post on 17-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Article

Targeting DNA Damage Re

sponse in ProstateCancer by Inhibiting AndrogenReceptor-CDC6-ATR-Chk1 Signaling

Graphical Abstract

Highlights

d CDC6 expression is increased during prostate cancer (PCa)

progression

d AR or CDC6 knockdown, together with AZD7762, suppresses

TopBP1-ATR-Chk1 signaling

d AR or CDC6 knockdown sensitizes PCa cells to AZD7762

d Enzalutamide and AZD7762 combination treatment

generates synergistic therapeutic effects

Karanika et al., 2017, Cell Reports 18, 1970–1981February 21, 2017 ª 2017 The Author(s).http://dx.doi.org/10.1016/j.celrep.2017.01.072

Authors

Styliani Karanika, Theodoros Karantanos,

Likun Li, ..., Wei Zhang, Shuhua Li,

Timothy C. Thompson

[email protected]

In Brief

CDC6 is an androgen receptor (AR) target

gene and an essential regulator of DNA

replication and checkpoint activation.

Karanika et al. show that combined

inhibition of the AR and Chk1 signaling

promotes DNA damage accumulation in

prostate cancer cells to induce cell death,

regardless of p53 status.

Page 2: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Cell Reports

Article

Targeting DNA Damage Response in ProstateCancer by Inhibiting AndrogenReceptor-CDC6-ATR-Chk1 SignalingStyliani Karanika,1,4 Theodoros Karantanos,1,4 Likun Li,1,4 Jianxiang Wang,1 Sanghee Park,1 Guang Yang,1 Xuemei Zuo,1

Jian H. Song,1 Sankar N. Maity,1 Ganiraju C. Manyam,2 Bradley Broom,2 Ana M. Aparicio,1 Gary E. Gallick,1

Patricia Troncoso,3 Paul G. Corn,1 Nora Navone,1 Wei Zhang,1 Shuhua Li,1 and Timothy C. Thompson1,5,*1Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA2Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA3Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA4Co-first author5Lead Contact

*Correspondence: [email protected]://dx.doi.org/10.1016/j.celrep.2017.01.072

SUMMARY

Cell division cycle 6 (CDC6), an androgen receptor(AR) target gene, is implicated in regulating DNAreplication and checkpoint mechanisms. CDC6expression is increased during prostate cancer(PCa) progression and positively correlates with ARin PCa tissues. AR or CDC6 knockdown, togetherwithAZD7762, aChk1/2 inhibitor, results indecreasedTopBP1-ATR-Chk1signaling andmarkedly increasedataxia-telangiectasia-mutated (ATM) phosphoryla-tion, a biomarker of DNA damage, and synergisticallyincreases treatment efficacy. Combination treatmentwith the AR signaling inhibitor enzalutamide (ENZ)and the Chk1/2 inhibitor AZD7762 demonstratessynergy with regard to inhibition of AR-CDC6-ATR-Chk1 signaling, ATM phosphorylation induction,and apoptosis in VCaP (mutant p53) and LNCaP-C4-2b (wild-type p53) cells. CDC6 overexpressionsignificantly reduced ENZ- and AZD7762-inducedapoptosis. Additive or synergistic therapeutic activ-ities are demonstrated in AR-positive animal xeno-graft models. These findings have important clinicalimplications, since they introduce a therapeutic strat-egy for AR-positive, metastatic, castration-resistantPCa, regardless of p53 status, through targeting AR-CDC6-ATR-Chk1 signaling.

INTRODUCTION

Metastatic prostate cancer remains an incurable disease with

variable prognosis (Wu et al., 2014). After an initial period of

response to systemic hormone therapy, the disease inexorably

progresses to a state known as metastatic, castration-resistant

prostate cancer (mCRPC) (Karanika et al., 2014). The therapeutic

armamentarium for mCRPC is limited to chemotherapy and

1970 Cell Reports 18, 1970–1981, February 21, 2017 ª 2017 The AutThis is an open access article under the CC BY-NC-ND license (http://

novel inhibitors of androgen receptor (AR) signaling, such as

abiraterone acetate and enzalutamide (ENZ), which provide

only moderate survival benefits (Ryan et al., 2013; Scher et al.,

2012).

DNA damage response (DDR) refers to coordinated cellular

mechanisms that prevent DNA damage accumulation and main-

tain genomic integrity (Karanika et al., 2014), and it plays a cen-

tral role in prostate cancer (PCa) initiation, development, and

progression (Tapia-Laliena et al., 2014). AR signaling in PCa cells

has been associated with numerous aspects of DDR pathways,

including regulation of ATM-Chk2 signaling for the initiation of

DDR (Ide et al., 2012), poly(ADP-ribose) polymerase function

(Schiewer et al., 2012), and non-homologous end joining recom-

bination (Polkinghorn et al., 2013). AR was reported to regulate

TopBP1-ATR-Chk1 signaling (Li et al., 2014), whereas ENZ

decreases CHEK1 expression in PCa cells (L.L., S.K., G.Y.,

J.W., S.P., B.B., G.C.M., J.H.S., G.E.G., T.K., P.G.C., P.T.,

X.Z., T.C.T., unpublished data).

One of the main types of DNA damage is DNA strand

breakage, which activates a cascade of intracellular events

that promote cell-cycle arrest and DDR, ensuring genomic integ-

rity, which can be particularly critical for cell survival in patients

with aggressive malignancies accumulating a myriad of genetic

errors (Karanika et al., 2014). DNA strand breaks activate ATR

via upstreammediators such as TopBP1, leading to Chk1-medi-

ated checkpoint activation and cell-cycle arrest. Cells are

thereby able to repair DNA damage, alleviating replication stress

and genomic instability. Chk1 pathway inhibition results in DNA

damage accumulation and, thus, increased ATM auto-phos-

phorylation, which mediates apoptosis of cells with incompletely

replicated DNA (d’Adda di Fagagna, 2008; Sarmento et al.,

2015). These findings demonstrate that ATR-Chk1 signaling is

crucial for the prevention of DNA-damage-induced cell death

associated with increased ATM phosphorylation/activation.

CDC6 is an essential regulator of DNA replication in eukaryotic

cells, and its best characterized function is pre-replicative com-

plex assembly at origins of replication during G1 phase (Borlado

and Mendez, 2008). Furthermore, CDC6 overexpression during

G2 phase blocks mitotic entry by activating Chk1 (Clay-Farrace

hor(s).creativecommons.org/licenses/by-nc-nd/4.0/).

Page 3: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Figure 1. CDC6 Is Induced during PCa Progression and Is Positively Correlated with AR Expression

(A) Immunohistochemical analysis of AR, CDC6, and P-CDC6 (S54) in normal prostate, primary prostate tumor, and bone metastases. NL, normal prostate; PCa,

prostate cancer; Bone met, bone metastasis. Treatment information of patient with metastasis is available in Table S1.

(B) qRT-PCR analysis of CDC6 mRNA levels. VCaP and C4-2b cells were transfected with 20 nM ARsi or NCsi for 48 hr. **p < 0.01; ***p < 0.0001.

(C and D) Protein stability analysis of Cdc6. VCaP (C) and C4-2b (D) cells were transfected with 20 nMArsi-1 or NCsi for 48 hr prior to the treatment with 100 mg/mL

cycloheximide (CHX) for indicated time. Left panels: western blotting analysis; right panels: densitometry analysis for Cdc6 protein stability in ARsi (red) and

NCsi (blue) transfected cells.

See also Table S1.

et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-

sion. CDC6 is required for Chk1 activation upon replication inhi-

bition (Oehlmann et al., 2004), and human CDC6 interacts with

ATR, promoting activation of the replication checkpoint (Yoshida

et al., 2010). Consequently, CDC6 decreases genomic insta-

bility, which is vital for cancer cell survival. CDC6 can also man-

ifest oncogenic activities via regulation of DNA replication and

repression of tumor suppressors (Gonzalez et al., 2006). Target-

ing ATR-Chk1 signaling increases the sensitivity to treatment

with DNA-damaging agents (Bartucci et al., 2012), making this

approach particularly attractive for the development of cancer

therapies. Notably, CHEK1 knockdown increases the sensitivity

of PCa stem cells to radiotherapy through increased DNA dam-

age (Fokas et al., 2012). AZD7762, a Chk1/2 inhibitor, synergizes

with DNA-damaging agents and radiation to induce apoptosis

via DNA double-strand breaks mediated by ATM activation in

many cell types (Mitchell et al., 2010; Sausville et al., 2014).

Furthermore, Brooks et al. found that Chk1 inhibition can selec-

tively induce apoptosis in melanoma cells in proportion to the

level of endogenous DNA damage related to replicative stress

without further induction of DNA damage by chemotherapy

(Brooks et al., 2013).

Taking the results of these studies into account, a potential

approach to treatment of mCRPC is the inhibition of more

than one level of a specific DDR signaling cascade, with the

goal of completely abolishing a specific signaling pathway.

This approach would take advantage of the fact that cancer cells

eventually accumulate more DNA damage than normal cells do,

eluding adverse effects of chemotherapy. Targeting the ATR-

Chk1 pathway at multiple levels to inhibit the repair of DNA dam-

age induced by replication stress in cancer cells may represent

an effective strategy for more complete DDR pathway inhibition.

Because CDC6 is an AR target gene (Jin and Fondell, 2009; Bai

et al., 2005) and is also involved with ATR-Chk1 signaling, this is

a particularly intriguing strategy for AR-positive PCa. In addition,

this approach may be effective under conditions of wild-type

p53, which is involved in multiple DDR pathways and can miti-

gate the response to some DNA-damaging agents (Ma et al.,

2012).

The aimof the present studywas to determine the role of CDC6

in regulation of ATR-Chk1 signaling and to test the combined in-

hibition of AR and Chk1 signaling as a therapeutic approach for

AR-positive mCRPC. Through this study, we also aimed to intro-

duce a therapeutic approach that effectively targets a DDR

pathway that promotes sufficient DNA damage accumulation in

PCa cells to induce cell death, regardless of p53 status.

RESULTS

CDC6 Is Induced during PCa Progression and IsPositively Correlated with AR ExpressionWe first evaluated the CDC6 expression and phosphorylation

and analyzed its correlation with AR expression in normal human

prostates and in primary and metastatic prostate tumor

specimens. In our immunohistochemistry (IHC) analysis, we

found that AR expression (p = 0.0057) and CDC6 expression

(p < 0.001) and phosphorylation (p = 0.0083) increased signifi-

cantly during PCa development and progression (Figure 1A). It

Cell Reports 18, 1970–1981, February 21, 2017 1971

Page 4: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

was reported that AR regulates CDC6 expression (Jin and Fon-

dell, 2009; Bai et al., 2005) and protein stability (Bai et al.,

2005), indicating functional associations between these twomol-

ecules. To determine themechanism of AR regulation of CDC6 in

our experimental models, we performed qRT-PCR and protein

stability analysis for CDC6 using AR siRNA (ARsi)- and nega-

tive-control siRNA (NCsi)-transfected VCaP and LNCaP C4-2b

(C4-2b) cells. Our results demonstrated that AR regulation of

CDC6 occurs at the mRNA level and not through regulation of

protein stability (Figures 1B–1D).

AR orCDC6Knockdown Increases the Sensitivity of PCaCells to theChk1/2 Inhibitor AZD7762 through Inhibitionof TopBP1-ATR-Chk1 SignalingTargeting Chk1/2 with AZD7762 increases the efficacy of DNA-

damaging modalities such as chemotherapy and radiation ther-

apy in patients with multiple malignancies (Mitchell et al., 2010;

Sausville et al., 2014). Thus, we tested our hypothesis that the

combination treatment with AR or CDC6 downregulation and

AZD7762 results in synergistic activities in VCaP and C4-2b.

The combination of ARsi and AZD7762 reduced protein levels

of CDC6, TopBP1, ATR, and Chk1, and it reduced phosphoryla-

tion of ATR and Cdc25C in VCaP and C4-2b (Figures 2A and 2B).

These signaling effects were accompanied by synergistically

increased ATM S1981 phosphorylation, a marker of DNA dam-

age. Phosphorylation of Chk1 S317, the site phosphorylated

by ATR or ATM (Canman, 2001; Kastan and Lim, 2000; Gatei

et al., 2003), was markedly elevated by AZD7762. Interestingly,

Chk1 S317 phosphorylation was positively correlated with

AZD7762-mediated, DNA-damage-induced phosphorylation of

ATM S1981, yet phosphorylation of Cdc25C S216, a Chk1

target, was substantially reduced (Figures 2A and 2B). To further

evaluate the contribution of ATM and ATR to the phosphorylation

of Chk1 at S317 in these specific cell contexts, we treated VCaP

and C4-2b cells with ATM inhibitor KU-60019 or ATR inhibitor

VE-821 in the absence and presence of AZD7762. The results

showed that both ATM inhibitor and ATR inhibitor can reduce

basal and AZD7762-induced phosphorylation of Chk1 S317

(Figures 2C and 2D). Since ATR and phosphorylated ATR

(P-ATR) were unchanged or reduced, it is unlikely that ATR

activities are related to the substantially increased Chk1 S317

phosphorylation in response to AZD7762; instead, the increased

Chk1 phosphorylation at S317 is most likely due to DNA-dam-

age-induced ATM phosphorylation of Chk1 S317.

Flow cytometric analysis demonstrated that the combination

treatment with AR knockdown and AZD7762 resulted in a

greater apoptotic effect than did AR knockdown (p < 0.001 in

both cell lines) or AZD7762 alone (VCaP, p = 0.01; C4-2b,

p < 0.001) (Figures 2E and 2F). DNA fragmentation analysis

demonstrated that ARsi and AZD7762 combination increased

the rate of apoptosis over AR knockdown (VCaP, p < 0.001;

C4-2b, p < 0.001) and AZD7762 (VCaP, p = 0.013; C4-2b,

p < 0.001) alone (Figures 2G and 2H). The differing responses

of VCaP and C4-2b cells to AZD7762 treatment are notable. In

addition to higher treatment efficacy from the combination of

ARsi and AZD7762, inhibition of AR with ARsi caused G1 arrest

and reduction of cells in S phase in both cell lines; however,

inhibition of Chk1 by AZD7762 led to significantly increased

1972 Cell Reports 18, 1970–1981, February 21, 2017

sub-G1 cells and DNA fragmentation in VCaP but had very little

effect on C4-2b (Figures 2E–2H), which may be largely due to

different p53 status in the two cell lines (VCaP, p53 mutant;

C4-2b, p53wild-type). It was reported that p53-mediated G1 ar-

rest in response to DNA damage can spare cells from AZD7762

action that predominantly occurs during G2 (Castedo et al.,

2004; Zhou and Bartek, 2004; Benada and Macurek, 2015). To

address this possibility, we knocked down p53 using small inter-

fering RNA (siRNA) in p53wild-type C4-2b cells and analyzed the

effect on cell-cycle distribution. As expected, knockdown of p53

significantly reduced the G0-G1 cell fraction and the combina-

tion of p53 knockdown and AZD7762 resulted in significantly

reduced G0-G1, S, and G2-M cells and significantly increased

sub-G1 cells (Figures 2I and 2J; Table S3).

Interestingly, we found that CDC6 knockdown and combina-

tion treatment with AZD7762 also markedly reduced TopBP1

protein levels and ATR S428 and Cdc25C S216 phosphorylation

in both cell lines (Figures 3A and 3B). We also found that the

combination treatment resulted in markedly greater ATM auto-

phosphorylation than did treatment with both agents alone,

suggesting a synergistic increase in DNA damage (Figures 3A

and 3B). Additionally, the combination treatment increased

Chk1 S317 phosphorylation in both cell lines (Figures 3A and 3B).

To determine the biological effects of combination treatment

with CDC6si and AZD7762, we examined apoptotic activities

using flow cytometry and a DNA fragmentation assay. Combina-

tion treatment increased the percentage of sub-G1 (apoptotic)

cells more than CDC6 knockdown did (VCaP, p = 0.01; C4-2b,

p<0.001) or AZD7762 (VCaP, p =0.0048;C4-2b, p<0.001) alone

(Figures 3C and 3D). The combination treatment also resulted in

greater DNA fragmentation/apoptosis than CDC6 knockdown

did (VCaP, p < 0.001; C4-2b, p = 0.004) and AZD7762 (VCaP,

p = 0.03; C4-2b, p = 0.03) alone (Figures 3E and 3F).

TopBp1 is an essential activator for the ATR-Chk1 signaling

pathway (Cimprich and Cortez, 2008; Wardlaw et al., 2014).

Combination of AR knockdown or CDC6 knockdown with

AZD7762 led to markedly reduced TopBP1 protein levels (Fig-

ures 2A, 2B, 3A, and 3B). These observations prompt us to

test whether knockdown of TOPBP1 could also synergize with

AZD7762 in the induction of PCa apoptosis and cell death.West-

ern blotting (WB) analysis showed that TOPBP1 knockdown

reduced Chk1 S317 phosphorylation, Cdc25C expression, and

Cdc25C S216 phosphorylation and that TOPBP1 knockdown,

together with AZD7762, further reduced Cdc25C S216 phos-

phorylation (Figures 4A and 4B). Flow cytometric analysis

demonstrated that the combination treatment with TOPBP1

knockdown and AZD7762 also resulted in a greater apoptotic

effect than TOPBP1 knockdown did (in VCaP: TOPBP1si_sc,

p = 0.0023; and TOPBP1si_3 p = 0.0066; in C4-2b: TOPBP1si_

sc, p = 0.0142; and TOPBP1si_3, p = 0.0037) and AZD7762

(in VCaP: TOPBP1si_sc, p = 0.0014; and TOPBP1si_3,

p = 0.0063; in C4-2b: TOPBP1si_sc, p = 0.0006; and

TOPBP1si_3, p = 0.0002) alone (Figures 4C and 4D). DNA

fragmentation analysis demonstrated that this combination

increased the rate of apoptosis over TOPBP1 knockdown

(in VCaP: TOPBP1si_sc, p < 0.0001; and TOPBP1si_3,

p = 0.0047; in C4-2b: TOPBP1si_sc, p = 0.0028; and

TOPBP1si_3, p = 0.0368) and AZD7762 (in VCaP: TOPBP1si_sc,

Page 5: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Figure 2. Effect of AR Knockdown and Chk1/2 Inhibitor AZD7762 on TopBP1-ATR-Chk1 Signaling and PCa Cell Survival

(A–J) In (A), (B), (E), and (H), VCaP and C4-2b cells were transfected with 20 mM ARsi or NCsi 24 hr prior to the treatment with 200 nM AZD7762 (AZD) for 48 hr.

(A and B) Western blotting analysis of Cdc6 and TopBP1-ATR-Chk1 signaling molecules in ARsi-, AZD-, and ARsi+AZD-treated VCaP (A) and C4-2b (B) cells.

(C and D) Western blotting analysis of P-Chk1 (S317) in VCaP and C4-2b cells that were treated with 200 nM AZD; 5 and 10 mMKU-60019 (KU), an ATM inhibitor;

1 and 3 mM VE-821 (VE), an ATR inhibitor; or a combination of AZD and KU or VE for 48 hr. (E and F) Cell-cycle analyses of ARsi-, AZD-, and ARsi+AZD-treated

VCaP (E) and C4-2b (F) cells. (G and H) DNA fragmentation analyses of ARsi-, AZD-, and ARsi+AZD-treated VCaP (G) and C4-2b (H) cells. (I) p53wild-type C4-2b

cells were transfected with 20 nMp53si or NCsi for 48 hr prior to western blotting analysis. (J) C4-2b cells were transfected with 20 nMp53si or NCsi for 48 hr prior

to the treatment with 200 nM AZD for 48 hr, followed by cell-cycle analysis.

*p < 0.05, statistically significant in sub-G1 cell distribution (E and F) or in DNA fragmentation (G and H) comparing the combination of ARsi and AZD to ARsi or

AZD alone; in G0-G1 and S cell distribution comparing p53si + DMSO to NCsi + DMSO (J); or in sub-G1, G0-G1, S, and G2-M cell distribution comparing p53si +

AZD to NCsi + AZD (J). Detailed statistical information on (J) is available in Table S2.

See also Table S2.

p < 0.0001; and TOPBP1si_3, p = 0.0027; in C4-2b: TOPBP1si_

sc, p = 0.0003; and TOPBP1si_3, p = 0.0011) alone (Figures 4E

and 4F).

Treatment with ENZ and AZD7762 Inhibits CDC6-TopBP1-ATR-Chk1 Signaling and Promotes DNADamage and Apoptosis in PCa CellsENZ is a potent AR signaling inhibitor approved for treatment of

mCRPC (Scher et al., 2012), and by combining it with AZD7762,

we can translate our findings into a viable therapeutic approach

for prostate cancer. We initially treated VCaP and C4-2b with

ENZ and/or AZD7762. We found that the combination treatment

markedly reduced Cdc6 phosphorylation and protein levels,

TopBP1 protein levels, and ATR and Chk1 phosphorylation

levels (Figure 5A). Importantly, the combination treatment syner-

gistically increased ATM phosphorylation and reduced Cdc25C

phosphorylation, markers for DNAdamage and the abrogation of

G2/M checkpoint, respectively (Figure 5A).

Next, we examined the apoptotic effect of combination treat-

ment with ENZ and AZD7762 using flow cytometry and a DNA

fragmentation assay. The results of flow cytometric analysis

demonstrated that the combination treatment significantly

Cell Reports 18, 1970–1981, February 21, 2017 1973

Page 6: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Figure 3. CDC6Knockdown Increases the Sensitivity of PCaCells to Treatmentwith Chk1/2 Inhibitor AZD through Inhibition of TopBP1-ATR-

Chk1 Signaling

VCaP and C4-2b cells were transfected with 20 mM CDC6si or NCsi 24 hr prior to the treatment with 200 nM AZD for 48 hr.

(A and B) WB analysis of TopBP1-ATR-Chk1 signaling proteins in VCaP (A) and C4-2b (B) cells.

(C and D) Flow cytometry analysis of CDC6si7- and AZD-treated VCaP (C) and C4-2b (D) cells. Top panels: representative cell-cycle profiles. Bottom panels:

quantitative analysis of cell-cycle distribution. Red arrows point to sub-G1, and blue arrows point to S phase.

(E and F) DNA fragmentation analysis of CDC6si7- and AZD-treated VCaP (E) and C4-2b (F) cells.

*p < 0.05, statistically significant in sub-G1 cell distribution (B and E) or in DNA fragmentation (C and F) when comparing the combination of ARsi and AZD7762 to

ARsi or AZD7762 alone.

increased the percentage of apoptotic (sub-G1) cells over that

resulting from ENZ (VCaP, p = 0.0053; C4-2b, p = 0.009) and

AZD7762 (VCaP, p = 0.012; C4-2b, p = 0.008) alone (Figures

5B and 5C). The results of DNA fragmentation analysis demon-

strated that the combination treatment increased the apoptotic

effect over that induced by ENZ (VCaP, p < 0.001; C4-2b,

p = 0.018) and AZD7762 (VCaP, p < 0.001; C4-2b, p = 0.0046)

alone (Figures 5D and 5E).

To strengthen our finding regarding the role of CDC6 in

the combination therapy using ENZ and AZD7762, we tested

1974 Cell Reports 18, 1970–1981, February 21, 2017

whether overexpression of CDC6 can overcome ENZ- and

AZD7762-induced PCa cell death. Our data demonstrated

that overexpression of CDC6 (Figure 5F) significantly reduced

sub-G1 cells (Figure 5G) and apoptotic DNA fragmentation (Fig-

ure 5H) in both VCaP and C4-2b.

Overall, our data demonstrated that combination treatment

with ENZ and AZD7762 downregulated CDC6 and TopBP1-

ATR-Chk1 signaling, released Cdc25C from inactivating phos-

phorylation by Chk1, and abolished G2/M checkpoint, resulting

in increased DNA damage and apoptosis (Figure 5I).

Page 7: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Figure 4. TOPBP1 Knockdown Increases the Sensitivity of PCa Cells to Treatment with Chk1/2 Inhibitor AZD

VCaP and C4-2b cells were transfected with 20 mM TOPBP1si or NCsi 24 hr prior to the treatment with 200 nM AZD for 48 hr.

(A and B) WB analysis of TopBP1, Chk1, P-Chk1 (S317), Cdc25C, and P-Cdc25C after TOPBP1 knockdown and treatment of AZD7762 in VCaP (A) and C4-2b

(B) cells.

(C and D) Flow cytometry analysis for sub-G1 cell distribution in TOPBP1si- and AZD-treated VCaP (C) and C4-2b (D) cells.

(E and F) DNA fragmentation analysis of TOPBP1si- and AZD-treated VCaP (E) and C4-2b (F) cells.

*p < 0.05, statistically significant in sub-G1 cell distribution (C and D) or in DNA fragmentation (E and F) when comparing the combination of TOPBP1si and

AZD7762 to TOPBP1si or AZD alone.

Combination Treatment with ENZ and AZD7762 Inhibitsthe Growth of Prostate Tumor XenograftsTo test our hypothesis that combination treatment with ENZ and

AZD7762 is a potential therapeutic approach for mCRPC, we

used three different animal models: VCaP, C4-2b xenografts,

and patient-derived xenograft (PDX) MDA-133-4, which harbors

amissensep53mutation (Leeet al., 2011).Weadministered treat-

ment to mice with orthotopic VCaP xenografts and monitored tu-

mor progression. Treatment with ENZ alone and the combination

of ENZ and AZD7762 reduced tumor growth compared to control

mice (p = 0.05 and p = 0.02, respectively), but the differences be-

tween combination and single-agent treatment were not statisti-

cally significant, according to assessment using an in vitro imag-

ing system (IVIS) (Figures 6A and S2A). However, ENZ is an

inducer of CYP450; it increases luciferinmetabolism and, through

this activity, reduces bioluminescence. In comparison, AZD7762

is an ATP-competitive Chk1/2 inhibitor; it binds to their respective

ATP-binding sites and increases the availability of ATP, which

is free to react with D-luciferin to produce light and, through this

activity, increase bioluminescence, leading to false increased

signal. When we evaluated tumor wet weights, we found that sin-

gle-agent treatment reduced tumor growth significantly (ENZ,

p = 0.009; AZD7762, p < 0.001), whereas the combination treat-

ment significantly reduced weights more than that resulting from

treatment with ENZ (p < 0.001) or AZD7762 (p = 0.008) alone.

Remarkably, the combination treatment was synergistic with re-

gard to tumor wet weight (p = 0.0097) (Figure 6B), according to

two-way ANOVA (Slinker, 1998).

To further validate our in vivo data and establish associations

with our WB data, we analyzed CDC6 and ATM expression and

phosphorylation in mice. Our in vitro studies demonstrated that

combination treatment with ENZ and AZD7762 significantly

reduced CDC6 expression and phosphorylation and increased

ATM phosphorylation. IHC analysis of VCaP xenografts demon-

strated that ENZ reduced CDC6 phosphorylation significantly,

compared to control (p = 0.036), whereas AZD7762 did not

produce significant effects (p = 0.06) (Figure S3A). However,

the combination treatment significantly reduced CDC6 phos-

phorylation compared to control (p = 0.012), ENZ-treated

(p = 0.0366), and AZD776-treated (p = 0.036) mice (Figures 6C

and S3A). Similarly, CDC6 expression was reduced to a greater

extent in ENZ-treated mice than in control mice (p = 0.036),

whereas the difference in AZD7762-treated and control mice

was not statistically significant (p = 0.4) (Figure S3A). The

combination treatment reduced CDC6 expression significantly,

compared to the control (p = 0.012), ENZ (p = 0.0214), and

AZD7762 (p = 0.0214). Treatment with ENZ or AZD7762 alone

did not have a significant effect on ATM phosphorylation,

whereas the combination treatment significantly increased this

phosphorylation over that in control (p = 0.021), ENZ-treated

(p = 0.036), and AZD7762-treated (p = 0.036) mice (Figures 6C

and S3A).

In subcutaneous C4-2b xenografts, we found that ENZ and

AZD7762, as single agents, did not significantly affect tumor vol-

ume or wet weight, compared to the control treatment (Figures

6D and 6E). In contrast, the combination treatment produced

Cell Reports 18, 1970–1981, February 21, 2017 1975

Page 8: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Figure 5. Combination Treatment with ENZ and AZD Inhibits CDC6-TopBP1-ATR-Chk1 Signaling, Promoting DNA Damage and Apoptosis in

PCa Cells

(A–E) VCaP and C4-2b cells were transfected with DMSO, 1 mM ENZ, and 200 nM AZD or ENZ+AZD for 48 hr. (A) ENZ and AZD combination treatment reduced

phosphorylation and protein levels of CDC6; TopBP1 protein levels; and phosphorylations of ATR, Chk1, and Cdc25C, leading to increased ATMphosphorylation

in both VCaP and C4-2b cells. (B and C) Flow cytometric analysis. ENZ and AZD combination treatment increased the percentage of apoptotic (sub-G1) cells in

VCaP (B) more than treatment with ENZ (p = 0.0053) or AZD7762 (p = 0.012) alone did, and it also increased the percentage of apoptotic (sub-G1) cells in C4-2b

(C) more than treatment with ENZ (p = 0.009) or AZD7762 (p = 0.008) alone did. (D and E) DNA fragmentation assays. The combination treatment increased

apoptosis in VCaP cells (D)more than treatment with ENZ alone (p < 0.001) or AZD7762 (p < 0.001) alone did, and it also increased apoptosis in C4-b cells (E) more

than treatment with ENZ (p = 0.018) or AZD7762 (p = 0.0046) alone did.

(F–H) Overexpression of CDC6 reduces ENZ- and/or AZD-induced apoptotic cell death. VCaP and C4-2b cells were transfected with 1 mg of CDC6 plasmid DNA

or control empty vector DNA for 24 hr prior to the treatment with DMSO, 1 mMENZ, 200 nMAZDor ENZ+AZD for 48 hr. (F) CDC6protein levelsmarkedly increased

after enforcedCDC6 expression in VCaP and C4-2b cells. (G) Flow cytometric analysis for apoptotic cell death (Sub-G1 cells). (H) DNA fragmentation assay. Data

in (H) are presented as fold of DMSO control. *p < 0.05, statistically significant when comparing combination treatment to single-agent treatment (B–E) and

comparing CDC6 overexpression to empty vector (G and H).

(I) Proposed signaling schema.

significantly lower tumor volumes at 8 days than in control

(p = 0.004) and ENZ-treated (p = 0.047) mice and significantly

lower tumor volumes at 11 days than in AZD7762-treated mice

(p = 0.04). The differences continued to be statistically significant

throughout the 21-day treatment period (Figure 6D). Moreover,

the combination treatment produced significantly lower tumor

wet weights than those in the control (p < 0.001), ENZ-treated

(p = 0.02), and AZD7762-treated (p = 0.022) mice (Figures 6E

and S2B).

1976 Cell Reports 18, 1970–1981, February 21, 2017

IHC analysis of C4-2b xenograft tumors showed that single-

agent treatment did not significantly reduce CDC6 phosphoryla-

tion, whereas the combination treatment significantly reduced it

to a greater extent than that in control (p = 0.012), ENZ-treated

(p = 0.021), and AZD7762-treated (p = 0.021) mice (Figures 6F

and S3B). CDC6 protein expression was reduced by single-

agent treatment, but differences did not reach statistical signifi-

cance compared to control mice (p = 0.4). In comparison, the

combination treatment significantly reduced CDC6 expression

Page 9: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Figure 6. Combination Treatment with ENZ and AZD7762 Inhibited the Growth of Prostate Tumor Xenografts

VCaP orthotopic xenografts, C4-2b subcutaneous xenografts, and MDA-133-4 PDX model were treated with vehicle control (C), enzalutamide (E), AZD7762 (A),

or enzalutamide + AZD7762 (E+A) for 35, 21, and 28 days, respectively.

(A–C) VCaP xenografts. (A) ENZ alone and combined with AZD7762 reduced tumor growth more than the control treatment (p = 0.05 and p = 0.02, respectively)

via IVIS measurements. (B) Treatment with ENZ and AZD7762 as single agents had significant effects (p = 0.009 and p < 0.001, respectively) on tumor wet

weights, and mice given the combination had tumors with significantly lower wet weight than mice given ENZ (p < 0.001) or AZD7762 (p = 0.008) alone did. The

combination treatment had synergistic effects on wet weights as determined using two-way ANOVA (p = 0.0097, indicated by a pound sign). (C) IHC analysis

demonstrated that the combination of ENZ and AZD7762 significantly decreased CDC6 phosphorylation (p = 0.01208) and Cdc6 protein levels (p = 0.012) and

significantly increased ATM phosphorylation (p = 0.01208) compared to the control treatment in VCaP xenografts. Full IHC analysis results, including comparison

of combination treatment with single-agent treatment, and quantitative analysis results can be found in Figure S2.

(D–F) C4-2b xenografts. Neither of the single agents had a significant effect on tumor volume, whereas the combination treatment resulted in significantly lower

tumor volumes at 8 days than the control treatment (p = 0.004) and enzalutamide (p = 0.047) and at 11 days than AZD7762 (p = 0.04). (D) These differences

continued to be statistically significant over 21 days. (E) Neither of the single agents had a significant effect on tumor wet weights, but the combination treatment

produced significantly lower tumor wet weights than the control treatment (p < 0.001), ENZ (p = 0.02), or AZD7762 (p = 0.022) did. (F) IHC analysis demonstrated

that the combination of ENZ and AZD7762 significantly decreased CDC6 phosphorylation (p = 0.01208) and CDC6 protein levels (p = 0.01208) and significantly

increased ATM phosphorylation (p = 0.01208) compared to the control treatment. Full IHC analysis results, including comparison of combination treatment with

single-agent treatment and quantitative analysis results can be found in Figure S2.

(G and H) The subcutaneous MDA-133-4 PDX model. (G) The combination treatment had a greater effect on tumor volume than ENZ alone at 21 (p = 0.016),

24 (p = 0.004), and 27 (p = 0.015) days or AZD7762 alone at 9 (p = 0.04), 14 (p = 0.014), 17 (p = 0.005), 21 (p = 0.009), 24 (p = 0.006), and 27 (p < 0.001) days. (H) The

combination treatment also had a greater effect on tumor wet weights than did ENZ (p = 0.036) and AZD7762 (p = 0.034) alone did.

Synergism was evaluated using ANOVA (p = 0.0004 for tumor volume, and p = 0.0107 for tumor wet weight, indicated by a pound sign in the panels). *p < 0.05.

See also Figure S2.

compared to the control (p = 0.012), ENZ-treated (p = 0.036),

and AZD7762-treated (p = 0.012) mice (Figures 6F and S3B).

Moreover, we found that neither of the single-agent treatments

had a greater effect on ATM phosphorylation than the control

treatment did. However, the combination treatment signifi-

cantly increased ATM phosphorylation over that with control

(p = 0.012), ENZ treatment (p = 0.036), and AZD7762 treatment

(p = 0.036) (Figure 6F), indicating that this combination treatment

Cell Reports 18, 1970–1981, February 21, 2017 1977

Page 10: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

synergistically increases the incidence of DNA damage in PCa

cells.

We also used the MDA-133-4 PDX, which was shown to

harbor a frameshift mutation of p53 that results in a truncated

protein, as an additional model (Lee et al., 2011; data not shown).

We found that the combination treatment had a greater effect

than ENZ did at 21 (p = 0.016), 24 (p = 0.004), and 28

(p = 0.015) days; and than AZD7762 did at 9 (p = 0.04), 14

(p = 0.014), 17 (p = 0.005), 21 (p = 0.009), 24 (p = 0.006), and

27 (p < 0.001) days (Figure 6G). Similar to the results for mice

with VCaP xenografts, single-agent treatment had a greater ef-

fect on tumor wet weight than the control treatment did, but

the combination treatment further reduced wet weights more

than ENZ (p = 0.036) and AZD7762 (p = 0.034) alone did (Fig-

ure 6H). These data suggested that ENZ and AZD7762 can syn-

ergistically inhibit tumor growth in a mCRPC model such as

MDA-133-4 PDX, which we confirmed via two-way ANOVA

(tumor volume, p = 0.0004; and wet weight, p = 0.0107). No sig-

nificant differences in body weight were found between drug-

treated and vehicle-control-treated mice in all three models.

DISCUSSION

For this study, we hypothesized that the targeting of ATR-Chk1

signaling in PCa cells is an effective approach. To maximize the

therapy effect, we evaluated the impact of targeting CDC6 on

ATR-Chk1 signaling. Gonzalez at al. claimed that CDC6 exerts

oncogenic activity though repression of the INK4/ARF locus

(Gonzalez et al., 2006). Additionally, Sideridou et al. showed

that not only p14, p15, and p16 but also E-cadherin is downregu-

lated at the transcriptional level by increased CDC6 (Sideridou

et al., 2011). Interestingly, p14ARF can repress AR trans-

activation in prostate cancer cells (Lu et al., 2013). CDC6 canpro-

tect genomic integrity via activation of DDR (Clay-Farrace et al.,

2003; Yoshida et al., 2010; Oehlmann et al., 2004), yet deregu-

lated overexpression of CDC6 can lead to rereplication, a form

of replication stress that can result in genomic instability (Liontos

et al., 2007).Within this context, the role of CDC6 in PCa is partic-

ularly intriguing, given that CDC6 is a direct AR target gene (Jin

and Fondell, 2009; Bai et al., 2005) and that AR upregulates the

expression of genes involved in DNA repair and DDR (Polking-

horn et al., 2013; Li et al., 2014). Indeed, we found that CDC6

was upregulated during PCa progression and CDC6 downregu-

lation synergized with the dual Chk1/2 inhibitor, AZD7762, to

inhibit TopBP1-ATR-Chk1 signaling in VCaP and C4-2b cells

and to increase its cytotoxic effects. It was notable that, in

marked contrast to treatmentwith single agents, the combination

of CDC6 knockdown and AZD7762 markedly affected the

expression of TopBP1, ATR (Figures 3A and 3B), and down-

stream biological effects (Figures 3C–3F). To monitor increased

DNA damage, we utilized the ATM S1981 phosphorylation as

a DNA damage marker, which is increased under Chk1 inhibi-

tion-mediated accumulation of DNA damage (Sarmento et al.,

2015). Experimentally, we demonstrated that ATM S1981 phos-

phorylation was increased following CDC6 knockdown and

further increased by a combination treatment of CDC6si

together with AZD7762 (Figures 3A and 3B). Importantly, CDC6

knockdown and AZD7762 combination treatment significantly

1978 Cell Reports 18, 1970–1981, February 21, 2017

increased the apoptotic response toDNAdamage inC4-2b cells.

Although it was not a major focus of this study, we infer that, in

p53 wild-type C4-2b cells, the combination ofCDC6 knockdown

andAZD7762 treatment will increase ATM-dependent p53 phos-

phorylation/activation, leading to increased Bax and p21 protein

expressions and subsequent p53-dependent G1 arrest, which

may spare C4-2b cells from AZD7762-mediated DNA-damage-

induced cell death. We used p53 knockdown to analyze the

role of p53 in C4-2b cells following AZD7762 treatment and

demonstrated that reduction of p53 levels significantly reduced

G0-G1 and S cell fractions and significantly increased sub-G1

cells in AZD7762-treated cells (Figures 2I and 2J; Table S3).

Previous publications have shown that TopBp1 plays an

important role in the activation of the ATR-Chk1 pathway (Cim-

prich and Cortez, 2008; Wardlaw et al., 2014; Li et al.,

2014). Our results show that AR or CDC6 knockdown combined

with AZD7762 treatment coordinately downregulated TopBP1

in both VCaP and C4-2b models and that, similar to AR or

CDC6 knockdown, the knockdown of TOPBP1 synergizes with

AZD7762 in the induction of apoptotic cell death in VCaP and

C4-2b PCa cells. These data, together with the results of ENZ

and AZD7762 combination experiments, demonstrated that a

synergistic therapeutic effect can be reached by targeting AR

and Chk1 simultaneously through inhibition/downregulation of

TopBP1-ATR-Chk1 signaling.

To validate our in vitro results, we selected VCaP and C4-2b

xenograft and human MDA-133-4 PDX AR-positive models for

our studies on the basis of their phenotypic characteristics,

which allow us to evaluate specific drug activities within the

context of androgen dependence and variable p53 status. We

found that ENZ andAZD7762 synergistically inhibit tumor growth

in VCaP xenografts compared to single agents. Regarding

C4-2b xenografts, we found that AZD7762 was not as effective

as it was in the othermodels, likely owing to its p53wild-type sta-

tus (Ma et al., 2012). Reduced response to ENZ and to the ENZ

and AZD7762 combination treatment was anticipated, since this

model is AR positive but androgen independent (Nguyen et al.,

2014). However, even though this model is androgen indepen-

dent, the combination of AR and Chk1/2 inhibition exhibited

marked activity. This suggests that PCa that is AR positive yet

fails to respond to ENZ treatment alone (Nguyen et al., 2014)

will respond to this combination therapeutic approach. Similar

to VCaP, in the MDA-PCa-133-4 model, combination treatment

of ENZ and AZD7762 synergistically inhibited tumor growth in

this model, despite the marked activities of single-agent treat-

ment. IHC analysis demonstrated that this combination reduced

CDC6 phosphorylation and expression and increased the levels

of ATM phosphorylation, a DNA damage biomarker, in VCaP and

C4-2b xenografts.

In summary, we demonstrated that CDC6 is upregulated

during progression of PCa and is positively associated with AR

expression. Our results indicated that targeting of AR-CDC6

via gene knockdown or ENZ, together with inhibition of Chk1/2

signaling by AZD7762, resulted in maximal suppression of

TopBP1-ATR-Chk1 signaling and the induction of DNA damage

and apoptosis in vitro. Furthermore, this therapeutic strategy ex-

hibited additive or synergistic therapeutic activities in xenograft

and PDX models in vivo. Importantly, one of the models we

Page 11: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

used, C4-2b, is androgen positive but androgen independent

and wild-type for p53. Its marked response to the combination

treatment indicates that the combined inhibition of androgen

signaling and Chk1/2 can be effective in the absence of intact

AR signaling and the presence of wild-type p53. Additional

studies are required to confirm the efficacy of this approach

and evaluate alternative methods of targeting DDR using AR

signaling inhibitors combined with DDR-targeted agents in

patients with mCRPC.

EXPERIMENTAL PROCEDURES

Cell Lines and Reagents

The human PCa cell line VCaP was validated as described previously (Li

et al., 2014), and LNCaP C4-2b (C4-2b) was recently validated in the MD

Anderson Characterized Cell Line Core Facility using the same method.

Cycloheximide (239763) was purchased from Calbiochem; and AZD7762,

enzalutamide (MDV3100), KU-60019, and VE-821 were purchased from

SelleckChem.

RNAi

CDC6si6 (SI04218389), CDC6si7 (SI04254782), TOPBP1si3 (SI00749553),

TP53si_7 (SI026233764), TP53si_8 (SI02623754), TP53si_9 (SI02655170),

TP53si_13 (SI4384079), and NCsi (1022076) were purchased from QIAGEN;

ARsi1 (s1538) and ARsi2 (s1540) were purchased from Life Technologies;

and TOPBP1si_sc (sc-41068) was purchased from Santa Cruz Biotechnology.

Gene knockdown experiments were performed using the Lipofectamine

RNAiMax transfection reagent (Life Technologies). PCa cells were seeded at

desired densities (VCaP: 1.0 3 106 per well; and C4-2b: 5 3 105 per well in

six-well plates, 1/5 or 1/30 of these cell numbers in 24- or 96-well plates,

respectively). Cells were transfected with 20 nM siRNA in the following day.

24 hr later, VCaP and C4-2b cells were treated with DMSO or AZD772, either

for 24 hr for WB analysis or for 48 hr for DNA fragmentation assay or flow cy-

tometric analysis.

WB Analysis

For WB analysis of the effects of CDC6si, ARsi, TOPBP1si, AZD7762, and

combinations of them, cells were transfected with siRNA for 24 hr and then

treated with DMSO or AZD7762 for 24 hr. Afterward, cells were treated with

a serum-free medium overnight and then with full serum for 4 hr (synchroniza-

tion) before protein extract preparation. For WB analysis of ENZ and AZD7762

effects, cells were treatedwith DMSOor ENZ for 24 hr. DMSOor AZD7762was

then added for 24 hr. Synchronization was achieved as described earlier.

Antibodies against CDC6 (3387), ATR (2790), P-ATR Ser428 (2853), Chk1

(2360), P-Chk1Ser317 (12302), Cdc25C (4688), P-Cdc25C Ser216 (9528),

ATM (2873), and P-ATM Ser1981 (13050) were purchased from Cell Signaling

Technology. Antibodies against P-CDC6 Ser54 (ab75809), P-Chk1ser296

(ab79758), and TopBp1 (ab2402) were purchased from Abcam. Antibodies

against GAPDH (365062) and AR (816) were purchased from Santa Cruz

Biotechnology. When indicated, densitometric analysis was performed, and

quantification of integrated density was assessed using the NIS-Elements-

AR software program (version 3.0; Nikon), followed by GAPDH normalization.

DNA Fragmentation Assay

A DNA fragmentation assay was performed for apoptotic evaluation of siRNA

and/drug effects. Cells were transfected with CDC6si, ARsi, TOPBP1si, or

NCsi; treated with DMSO or ENZ for 24 hr; and then treated with DMSO,

AZD7762, or ENZ+AZD7762 for 48 hr. DNA fragmentation analysis was per-

formed according to manufacturer’s instructions. Data are presented as

mean ± SE.

Flow Cytometry Analysis

Cells were treated as described earlier and were prepared for flow cyto-

metric analysis as described previously (Li et al., 2014). Data are presented

as mean ± SD.

Protein Stability Assay

VCaP and C4-2b cells were treated with NCsi or ARsi for 48 hr and then

incubated with 100 mg/mL cycloheximide for the indicated time periods. Cell

extracts were obtained via lysis in a modified RIPA buffer.

Animal Studies

All animal experiments were conducted in accordance with accepted stan-

dards of humane animal care approved by MDACC IACUC. Data are pre-

sented as mean ± SE.

Orthotopic VCaP Xenografts

VCaP xenografts were generated in mice as described previously (Li et al.,

2014). The experimental groups received DMSO, ENZ (10 mg/kg daily),

AZD7762 (25 mg/kg, twice daily every third day) or a combination of ENZ

and AZD7762 for 35 days. Tumor size was monitored weekly according to

luminescence signal, using the IVIS 200 Imaging System (PerkinElmer). The

mice were sacrificed, and their tumors were collected.

Subcutaneous C4-2b Xenografts

Aliquots of 6 3 106 C4-2b cells in 100 mL of 10% fetal calf serum (FCS) and

T medium + 50% Matrigel were injected subcutaneously into the right flanks

of athymic nude male mice (Taconic) to induce subcutaneous tumors. Tumors

were allowed to grow for 24 days before treatment. The experimental groups

received treatment for 21 days in doses as described earlier. Tumor size was

monitored by measuring three dimensions and using the following formula:

length/2 3 width/2 3 height/2 3 p 3 4/3. The mice were sacrificed, and their

tumors were collected.

MDA-PCa-133-4 PDXs

Subcutaneous PDXs of MDA-PCa-133-4 were generated by implanting

0.125-cm3 tumor fragment into the left flanks of previously castrated severe

combined immunodeficiency mice (SCID) mice (Charles River Laboratories).

RNA sequencing (RNA-seq) analysis showed that this tumor harbors a

missense p53 mutation (P72R). Tumors were allowed to grow until they

reached a volume of 50 mm3. The experimental groups received treatment

for 28 days in doses as described earlier. Tumor size was monitored as

described for the C4-2b model. The mice were sacrificed, and tumors were

collected.

Patient-Derived Xenografts

The MD Anderson Cancer Center PCa PDXs were developed, as described

previously (Li et al., 2014).

Immunohistochemical Analysis

Eleven human normal prostate specimens, 28 human primary PCa specimens

(not previously treated) obtained after radical prostatectomy, and 9 metastatic

PCa specimens (submitted to various previous treatments; Table S1) were

obtained after patients provided informed consent and were used to analyze

phosphorylated CDC6 (P-CDC6) and CDC6 expression. Antibodies against

AR (816), P-CDC6 S54 (12920), and CDC6 (8341) obtained from Santa Cruz

Biotechnology were used for IHC. CDC6 and AR immunostainings were also

performed and scored on tissue microarray slides composed of 34 tumor

xenografts generated from different histologic types of PCa (Table S2). For

IHC analysis of tumor xenografts, specimens were prepared as described

previously (Li et al., 2014). Antibodies against P-CDC6 S54 and CDC6 (Santa

Cruz Biotechnology), P-ATM S1981 (32420), and ATM (1292; Abcam) were

used. Immunostaining scoring was performed as described previously (Li

et al., 2014).

Statistical Analysis

The results are presented as the mean ± SE or mean ± SD from at least

three independent experiments. Comparisons of groups were appropriately

analyzed using the Student’s t test, the Mann-Whitney U test, Spearman’s

rho, or the Kruskal-Wallis rank test. p values less than 0.05 were considered

statistically significant, and all tests were two-tailed. Synergism was deter-

mined using two-way ANOVA (Slinker, 1998; Li et al., 2014).

Cell Reports 18, 1970–1981, February 21, 2017 1979

Page 12: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

SUPPLEMENTAL INFORMATION

Supplemental Information includes three figures and three tables can be found

with this article online at http://dx.doi.org/10.1016/j.celrep.2017.01.072.

AUTHOR CONTRIBUTIONS

T.C.T., S.K., T.K., and L.L. conceived and designed the study and wrote the

paper. In vitro studies, including siRNA and drug treatments, WB analysis,

flow cytometric assays, DNA fragmentation experiments, protein stability as-

says, statistic and synergy analysis: L.L., S.K., T.K., J.W., X.Z., W.Z., and

S.L.; IHC and tissue microarray analysis: G.Y., S.K., and T.K.; xenograft model

studies: S.P., S.K., T.K., J.W., J.H.S., and G.G. P.G.C. contributed to manu-

script preparation. S.N.M., A.M.A., P.T., and N.N. contributed to pathological

analysis of human samples and the establishment of PDX lines. B.B and

G.C.M. performed bioinformatics analyses of RNA-seq data.

ACKNOWLEDGMENTS

We thank XinhaiWan and Jun Yang for guiding in animal experiments and Don-

ald Norwood and Linda Bohannon for editing the manuscript. Funding: this

work was supported in part by National Cancer Institute grant R0150588 (to

T.C.T.); National Cancer Institute grant P50140388; the Prostate Cancer

Specialized Program of Research Excellence at The University of Texas MD

Anderson Cancer Center; National Cancer Institute grant CA16672; MD An-

derson Cancer Center Support Grant; and Tony’s Prostate Cancer Research

Foundation.

Received: January 20, 2015

Revised: November 11, 2016

Accepted: January 26, 2017

Published: February 21, 2017

REFERENCES

Bai, V.U., Cifuentes, E., Menon, M., Barrack, E.R., and Reddy, G.P. (2005).

Androgen receptor regulates Cdc6 in synchronized LNCaP cells progressing

from G1 to S phase. J. Cell. Physiol. 204, 381–387.

Bartucci, M., Svensson, S., Romania, P., Dattilo, R., Patrizii, M., Signore, M.,

Navarra, S., Lotti, F., Biffoni, M., Pilozzi, E., et al. (2012). Therapeutic targeting

of Chk1 in NSCLC stem cells during chemotherapy. Cell Death Differ. 19,

768–778.

Benada, J., and Macurek, L. (2015). Targeting the checkpoint to kill cancer

cells. Biomolecules 5, 1912–1937.

Borlado, L.R., and Mendez, J. (2008). CDC6: from DNA replication to cell cycle

checkpoints and oncogenesis. Carcinogenesis 29, 237–243.

Brooks, K., Oakes, V., Edwards, B., Ranall, M., Leo, P., Pavey, S., Pinder, A.,

Beamish, H., Mukhopadhyay, P., Lambie, D., and Gabrielli, B. (2013). A potent

Chk1 inhibitor is selectively cytotoxic in melanomas with high levels of replica-

tive stress. Oncogene 32, 788–796.

Canman, C.E. (2001). Replication checkpoint: preventing mitotic catastrophe.

Curr. Biol. 11, R121–R124.

Castedo, M., Perfettini, J.L., Roumier, T., Andreau, K., Medema, R., and

Kroemer, G. (2004). Cell death by mitotic catastrophe: a molecular definition.

Oncogene 23, 2825–2837.

Cimprich, K.A., and Cortez, D. (2008). ATR: an essential regulator of genome

integrity. Nat. Rev. Mol. Cell Biol. 9, 616–627.

Clay-Farrace, L., Pelizon, C., Santamaria, D., Pines, J., and Laskey, R.A.

(2003). Human replication protein Cdc6 prevents mitosis through a checkpoint

mechanism that implicates Chk1. EMBO J. 22, 704–712.

d’Adda di Fagagna, F. (2008). Living on a break: cellular senescence as a DNA-

damage response. Nat. Rev. Cancer 8, 512–522.

Fokas, E., Prevo, R., Pollard, J.R., Reaper, P.M., Charlton, P.A., Cornelissen,

B., Vallis, K.A., Hammond, E.M., Olcina, M.M., Gillies McKenna, W., et al.

1980 Cell Reports 18, 1970–1981, February 21, 2017

(2012). Targeting ATR in vivo using the novel inhibitor VE-822 results in selec-

tive sensitization of pancreatic tumors to radiation. Cell Death Dis. 3, e441.

Gatei, M., Sloper, K., Sorensen, C., Syljuasen, R., Falck, J., Hobson, K., Sav-

age, K., Lukas, J., Zhou, B.B., Bartek, J., and Khanna, K.K. (2003). Ataxia-tel-

angiectasia-mutated (ATM) and NBS1-dependent phosphorylation of Chk1 on

Ser-317 in response to ionizing radiation. J. Biol. Chem. 278, 14806–14811.

Gonzalez, S., Klatt, P., Delgado, S., Conde, E., Lopez-Rios, F., Sanchez-Ces-

pedes, M., Mendez, J., Antequera, F., and Serrano, M. (2006). Oncogenic ac-

tivity of Cdc6 through repression of the INK4/ARF locus. Nature 440, 702–706.

Ide, H., Lu, Y., Yu, J., China, T., Kumamoto, T., Koseki, T., Yamaguchi, R.,

Muto, S., and Horie, S. (2012). Testosterone promotes DNA damage response

under oxidative stress in prostate cancer cell lines. Prostate 72, 1407–1411.

Jin, F., and Fondell, J.D. (2009). A novel androgen receptor-binding element

modulates Cdc6 transcription in prostate cancer cells during cell-cycle pro-

gression. Nucleic Acids Res. 37, 4826–4838.

Karanika, S., Karantanos, T., Li, L., Corn, P.G., and Thompson, T.C. (2014).

DNA damage response and prostate cancer: defects, regulation and thera-

peutic implications. Oncogene 34, 2815–2822.

Kastan, M.B., and Lim, D.S. (2000). The many substrates and functions of

ATM. Nat. Rev. Mol. Cell Biol. 1, 179–186.

Lee, Y.C., Cheng, C.J., Bilen, M.A., Lu, J.F., Satcher, R.L., Yu-Lee, L.Y., Gal-

lick, G.E., Maity, S.N., and Lin, S.H. (2011). BMP4 promotes prostate tumor

growth in bone through osteogenesis. Cancer Res. 71, 5194–5203.

Li, L., Chang, W., Yang, G., Ren, C., Park, S., Karantanos, T., Karanika, S.,

Wang, J., Yin, J., Shah, P.K., et al. (2014). Targeting poly(ADP-ribose) polymer-

ase and the c-Myb-regulated DNA damage response pathway in castration-

resistant prostate cancer. Sci. Signal. 7, ra47.

Liontos, M., Koutsami, M., Sideridou, M., Evangelou, K., Kletsas, D., Levy, B.,

Kotsinas, A., Nahum, O., Zoumpourlis, V., Kouloukoussa, M., et al. (2007). De-

regulated overexpression of hCdt1 and hCdc6 promotes malignant behavior.

Cancer Res. 67, 10899–10909.

Lu, W., Xie, Y., Ma, Y., Matusik, R.J., and Chen, Z. (2013). ARF represses

androgen receptor transactivation in prostate cancer. Mol. Endocrinol. 27,

635–648.

Ma, C.X., Cai, S., Li, S., Ryan, C.E., Guo, Z., Schaiff, W.T., Lin, L., Hoog, J.,

Goiffon, R.J., Prat, A., et al. (2012). Targeting Chk1 in p53-deficient triple-

negative breast cancer is therapeutically beneficial in human-in-mouse tumor

models. J. Clin. Invest. 122, 1541–1552.

Mitchell, J.B., Choudhuri, R., Fabre, K., Sowers, A.L., Citrin, D., Zabludoff,

S.D., and Cook, J.A. (2010). In vitro and in vivo radiation sensitization of human

tumor cells by a novel checkpoint kinase inhibitor, AZD7762. Clin. Cancer Res.

16, 2076–2084.

Nguyen, H.G., Yang, J.C., Kung, H.J., Shi, X.B., Tilki, D., Lara, P.N., Jr., DeVere

White, R.W., Gao, A.C., and Evans, C.P. (2014). Targeting autophagy over-

comes Enzalutamide resistance in castration-resistant prostate cancer cells

and improves therapeutic response in a xenograft model. Oncogene 33,

4521–4530.

Oehlmann, M., Score, A.J., and Blow, J.J. (2004). The role of Cdc6 in ensuring

complete genome licensing and S phase checkpoint activation. J. Cell Biol.

165, 181–190.

Polkinghorn, W.R., Parker, J.S., Lee, M.X., Kass, E.M., Spratt, D.E., Iaquinta,

P.J., Arora, V.K., Yen, W.F., Cai, L., Zheng, D., et al. (2013). Androgen receptor

signaling regulates DNA repair in prostate cancers. Cancer Discov. 3, 1245–

1253.

Ryan, C.J., Molina, A., and Griffin, T. (2013). Abiraterone in metastatic prostate

cancer. N. Engl. J. Med. 368, 1458–1459.

Sarmento, L.M., Povoa, V., Nascimento, R., Real, G., Antunes, I., Martins, L.R.,

Moita, C., Alves, P.M., Abecasis,M.,Moita, L.F., et al. (2015). CHK1 overexpres-

sion in T-cell acute lymphoblastic leukemia is essential for proliferation and sur-

vival by preventing excessive replication stress. Oncogene 34, 2978–2990.

Sausville, E., Lorusso, P., Carducci, M., Carter, J., Quinn, M.F., Malburg, L.,

Azad, N., Cosgrove, D., Knight, R., Barker, P., et al. (2014). Phase I dose-esca-

lation study of AZD7762, a checkpoint kinase inhibitor, in combination with

Page 13: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

gemcitabine in US patients with advanced solid tumors. Cancer Chemother.

Pharmacol. 73, 539–549.

Scher, H.I., Fizazi, K., Saad, F., Taplin, M.E., Sternberg, C.N., Miller, K., deWit,

R., Mulders, P., Chi, K.N., Shore, N.D., et al.; AFFIRM Investigators (2012).

Increased survival with enzalutamide in prostate cancer after chemotherapy.

N. Engl. J. Med. 367, 1187–1197.

Schiewer, M.J., Goodwin, J.F., Han, S., Brenner, J.C., Augello, M.A., Dean,

J.L., Liu, F., Planck, J.L., Ravindranathan, P., Chinnaiyan, A.M., et al. (2012).

Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov.

2, 1134–1149.

Sideridou, M., Zakopoulou, R., Evangelou, K., Liontos, M., Kotsinas, A., Ram-

pakakis, E., Gagos, S., Kahata, K., Grabusic, K., Gkouskou, K., et al. (2011).

Cdc6 expression represses E-cadherin transcription and activates adjacent

replication origins. J. Cell Biol. 195, 1123–1140.

Slinker, B.K. (1998). The statistics of synergism. J. Mol. Cell. Cardiol. 30,

723–731.

Tapia-Laliena, M.A., Korzeniewski, N., Hohenfellner, M., and Duensing, S.

(2014). High-risk prostate cancer: a disease of genomic instability. Urol. Oncol.

32, 1101–1107.

Wardlaw, C.P., Carr, A.M., and Oliver, A.W. (2014). TopBP1: a BRCT-scaffold

protein functioning in multiple cellular pathways. DNA Repair (Amst.) 22,

165–174.

Wu, J.N., Fish, K.M., Evans, C.P., Devere White, R.W., and Dall’Era, M.A.

(2014). No improvement noted in overall or cause-specific survival formen pre-

senting with metastatic prostate cancer over a 20-year period. Cancer 120,

818–823.

Yoshida, K., Sugimoto, N., Iwahori, S., Yugawa, T., Narisawa-Saito, M.,

Kiyono, T., and Fujita, M. (2010). CDC6 interaction with ATR regulates activa-

tion of a replication checkpoint in higher eukaryotic cells. J. Cell Sci. 123,

225–235.

Zhou, B.B., and Bartek, J. (2004). Targeting the checkpoint kinases: chemo-

sensitization versus chemoprotection. Nat. Rev. Cancer 4, 216–225.

Cell Reports 18, 1970–1981, February 21, 2017 1981

Page 14: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Cell Reports, Volume 18

Supplemental Information

Targeting DNA Damage Response in Prostate

Cancer by Inhibiting Androgen

Receptor-CDC6-ATR-Chk1 Signaling

Styliani Karanika, Theodoros Karantanos, Likun Li, Jianxiang Wang, SangheePark, Guang Yang, Xuemei Zuo, Jian H. Song, Sankar N. Maity, Ganiraju C.Manyam, Bradley Broom, Ana M. Aparicio, Gary E. Gallick, Patricia Troncoso, Paul G.Corn, Nora Navone, Wei Zhang, Shuhua Li, and Timothy C. Thompson

Page 15: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Fig. S1

P-Cdc6 S54 P-Cdc6 S54 pre-absorbed with a P-Cdc6 S54 peptide

Fig. S1. Verification of the specificity of P-Cdc6 (Ser54) antibody (related to Figure 1). The specificity of P-Cdc6 (Ser54) antibody was verified by incubating human bone met tissue sections with P-Cdc6 (Ser54) antibody (1 µg/ml) (left panel) or with the same antibody pre-absorbed with a phosphor-Cdc6 (Ser54) peptide (100 µg/ml) (right panel). Both antibody and peptide were from Santa Cruz Biotech. The staining in the tissue slide incubated with the pre-absorbrd antibody was mostly abolished as compared with that incubated with the specific P-Cdc6 (Ser54) antibody.

Page 16: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

A B

C E

A E+A

Fig. S2

C E

A E+A

Figure S2. Combination treatment with enzalutamide and AZD7762 inhibited the growth of prostate tumor xenografts (Related to Figure 6). A. Representative IVIS images showing that in VCaP xenografts, combination treatment reduced tumor growth more than the control treatment, enzalutamide or AZD7762 treatment alone. B. Representative images showing that in C4-2b subcutaneous model, combination treatment reduced tumor growth more than the control treatment, enzalutamide or AZD7762 treatment alone. C, control; E, enzalutamide; A, AZD7762, E+A, enzalutamide+AZD7762.

Page 17: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Fig. S3

Figure S3. Combination treatment with enzalutamide and AZD7762 decreased the expression and phosphorylation of CDC6 and increased the phosphorylation of ATM in prostate tumor xenografts (Related to Figure 6). A. IHC analysis demonstrated that the combination of enzalutamide and AZD7762 significantly decreased CDC6 phosphorylation more so than the control treatment (P=0.01208), enzalutamide (P=0.03662) and AZD7762 (P=0.03662) did in VCaP xenografts. Furthermore, the combination of two drugs reduced CDC6 protein levels more so than the control treatment (P=0.012), enzalutamide (P=0.021) and AZD7762 (P=0.021) did in these xenografts. B. The combination of enzalutamide and AZD7762 significantly increased ATM phosphorylation more so than did the control treatment (P=0.01208), enzalutamide (P=0.03662) and AZD7762 (P=0.03662) in VCaP xenografts. No significant differences in ATM protein levels with the different therapies where observed in VCaP xenografts. C. The combination of enzalutamide and AZD7762 significantly decreased s CDC6 phosphorylation more so than the control treatment (P=0.01208), enzalutamide (P=0.02144) and AZD7762 (P=0.02144) did in C4-2b xenografts. In addition, the combination treatment decreased CDC6 protein levels more so than did control treatment (P=0.01208), enzalutamide (P=0.03662) and AZD7762 (P=0.01208) in these xenografts. D. The combination of enzalutamide and AZD7762 significantly increased ATM phosphorylation more so than control treatment (P=0.01208), enzalutamide (P=0.03662) and AZD7762 (P=0.03662) did in C4-2b xenografts.

Page 18: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Table S1. Treatment information for patients with metastatic disease.

(Related to Figure 1)

Sample ID# ADT-yes/no ADT therapy received Date on Date off

TR08-16-40 yes Casodex Aug.,1999 7/28/2000

Lupron 10/1/1999 10/14/2001

DES 10/14/2001 4/18/2003

TR08-16-42 yes Lupron 5/26/2004 11/16/2004

Xopondex 10/12/2004 5/9/2005

Casodex 4/13/2004 5/26/2004

DES 3/20/2005 5/9/2005

TR12-10-42 yes Casodex 10/26/2000 unk

Lupron 10/16/2000

TR08-16-44 yes Flutamide 10/16/2000 unk

Lupron 10/16/2000 1/26/2006

TR08-16-45 yes Casodex 6/29/2004 unk 2004

Lupron 6/29/2004 unk 2004

Casodex Jul. 2000 Apr. 2001

Lupron Jul. 2000 Apr. 2001

TR08-16-46 yes Casodex 6/24/2004 3/1/2005

Lupron 7/6/2004 12/21/2004

TR08-16-47 yes Lupron unk 1999 1/30/2003

Casodex unk 1999 4/18/2001

TR12-10-45 yes Lupron Jun. 2000 unk 2001

Casodex Jun. 1999 unk 2001

TR08-16-49 yes Lupron unk 1999 unk 2006

Flutamide unk unk 2005

Casodex Nov. 2005 unk 2006

Page 19: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Table S2. Cell cycle analysis (Related Figure 2E-F, Figure 3C-D and Figure 5B-C). VCaP and C4-2b Cells were treated with negative control siRNA (NCsi), ARsi, or CDC6si 24 h prior to AZD7762 (AZD) treatment for 48 h, or pretreated with 1 µM enzalutamide (ENZ) or vehicle control DMSO, followed by treatment with DMSO, ENZ, AZD or ENZ+AZD for 48 h. Cells were stained with propidium iodide, and analyzed on a FACS Canto II flow cytometer (BD Biosciences). Cell cycle profiles and quantitative data were obtained using FlowJo software (Tree Star Inc.)

VCaP C4-2b Sub-G1 G0-G1 S G2-M

NCsi+DMSO NCsi+AZD ARsi+DMSO ARsi+AZD NCsi+DMSO NCsi+AZD ARsi+DMSO ARsi+AZD 3.77 10.65 10.20 15.56 65.04 60.43 79.36 71.24 7.89 7.48 2.30 2.87 16.70 15.15 6.51 8.30

4.51 5.11 4.54 8.44 63.81 67.02 74.86 69.24 7.89 7.65 2.98 2.45 16.90 18.12 10.23 9.63

Sub-G1 G0-G1 S G2-M

NCsi+DMSO NCsi+AZD CDC6si+DMSO CDC6si+AZD NCsi+DMSO NCsi+AZD CDC6si+DMSO CDC6si+AZD 3.78 16.13 6.86 26.53 69.47 50.97 64.40 48.53 3.60 5.37 4.72 3.64 16.53 21.37 17.63 15.47

4.38 4.76 8.13 20.83 65.53 67.97 70.23 62.83 8.75 8.44 3.77 3.40 16.97 18.60 13.67 10.86

Sub-G1 G0-G1 S G2-M

DMSO ENZ AZD ENZ+AZD DMSO ENZ AZD ENZ+AZD 5.70 10.69 11.77 18.70 72.53 71.67 66.87 65.63 3.62 2.94 7.56 3.36 15.00 12.23 9.67 9.87

4.82 6.49 6.08 12.11 63.93 66.70 67.17 62.70 7.98 4.02 6.03 3.41 16.93 14.50 19.00 13.80

Page 20: Targeting DNA Damage Response in Prostate ......et al., 2003; Yoshida et al., 2010), which inhibits G2/M progres-sion. CDC6 is required for Chk1 activation upon replication inhi-bition

Table S3. Statistical analysis of cell cycle data in p53si and AZD treated C4-2b cells (Related to figure 2J). C4-2b Cells were treated with 20 nM negative control siRNA (NCsi), or p53si 24 h prior to AZD7762 (AZD) treatment for 48 h. Cells were stained with propidium iodide, and analyzed on a FACS Canto II flow cytometer (BD Biosciences). Cell cycle profiles and quantitative data were obtained using FlowJo software (Tree Star Inc.). Statistical analysis was performed using two-tailed Student t-test.

Condition Sub-G1 (%) G0-G1 (%) S (%) G2-M (%)

NCsi+DMSO

NCsi+AZD

P53si_7+DMSO

P53si_7+AZD

P53si_9+DMSO

P53si_9+AZD

7.07 55.81 9.81 17.06 8.93 57.00 9.09 16.52 8.47 47. 84 7.78 17.94 25.8 40.83 5.98 13.72 9.17 47.57 7.94 17.56 24.3 39.06 5.87 13.76

Comparison p-value

NCsi+DMSO: p53si_7+DMSO

NCsi+DMSO: p53si_9+DMSO

NCsi+AZD:p53si_7+AZD

NCsi+AZD:p53si_9+AZD

Sub-G1 G0-G1 S G2-M

0.6615 0.0296 0.0041 0.2821

0.4304 0.0429 0.0094 0.4605

0.0008 0.0023 0.0096 0.0251

0.0141 0.0022 0.0108 0.0021