Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Inhibition of the EGFR/STAT3/CEBPD axis reverses cisplatin cross-resistance with paclitaxel in the urothelial carcinoma of the urinary bladder

Wei-Jan Wang1, Chien-Feng Li6, Yu-Yi Chu4, Yu-Hui Wang4, Tzyh-Chyuan Hour8,

Chia-Jui Yen5, Wen-Chang Chang2, 7 and Ju-Ming Wang2, 3, 4, 7*

1Institute of Basic Medical Science, 2Infectious Disease and Signaling Research Center, 3Center of molecular inflammation, and 4Institute of Bioinformatics and 5 Biosignal Transduction, Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 701, 6Department of Pathology, Chi-Mei Medical Center, Tainan 701, 7Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, 8Department of Biochemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan R. O. C.

*To whom all correspondence should be addressed: Ju-Ming Wang Institute of Bioinformatics and Biosignal Transduction College of Bioscience and Biotechnology National Cheng Kung University Tainan 701, Taiwan Phone: Fax: E-mail:

CONFLICT OF INTEREST No potential conflicts of interest were disclosed.

RUNNING TITLE: Targeting CEBPD prevents drug resistance in UCUB

1

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

Purpose: Cisplatin (CDDP) is frequently used in combination chemotherapy with

paclitaxel (PTX) for treating urothelial carcinoma of the urinary bladder (UCUB).

CDDP cross-resistance has been suggested to develop with PTX, thus hindering

successful UCUB treatment. Therefore, elucidating the mechanisms underlying

CDDP-induced anticancer drug resistance is imperative and may provide an insight in

developing novel therapeutic strategy.

Experimental Design: Loss-of-function assays were performed to elucidate the

role of the epidermal growth factor (EGFR) and transducer and activator of

transcription 3 (STAT3) in CDDP-induced CCAAT/enhancer-binding delta

(CEBPD) expression in UCUB cells. Reporter and in vivo DNA-binding assays were

employed to determine whether CEBPD directly regulates ATP binding cassette

subfamily B member 1 (ABCB1) and ATP binding cassette subfamily C member 2

(ABCC2) activation. Finally, a xenograft animal assay was used to examine the

abilities of gefitinib and S3I-201 (a STAT3 inhibitor) to reverse CDDP and PTX

sensitivity.

Results: CEBPD expression was maintained in postoperative

chemotherapy patients, and this expression was induced by CDDP even in

CDDP-resistant UCUB cells. Upon CDDP treatment, CEBPD activated ABCB1 and

ABCC2. Furthermore, the EGFR/STAT3 pathway contributed to CDDP-induced

CEBPD expression in UCUB cells. Gefitinib and S3I-201 treatment significantly

reduced the expression of CEBPD and enhanced the sensitivity of CDDP-resistant

UCUB cells to CDDP and PTX.

Conclusions: Our results revealed the risk of CEBPD activation in

CDDP-resistant UCUB cells and suggested a therapeutic strategy for patients with

2

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

UCUB or UCUB resisted to CDDP and PTX by combination with either gefitinib or

S3I-201.

3

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Translational Relevance

Despite extensive investigation, urothelial carcinoma of the urinary bladder (UCUB)

remains a relatively understudied disease, and nonsurgical treatments have undergone

little development in recent decades. We demonstrated that epidermal growth factor

receptor (EGFR)-induced CEBPD promotes cisplatin (CDDP) and paclitaxel (PTX)

cross-resistance in CDDP-resistant UCUB by elevating ATP binding cassette

subfamily B member 1 (ABCB1) and ATP binding cassette subfamily C member 2

(ABCC2) levels, respectively. In addition, our results validated suppression of

CEBPD by gefitinib or S3I-201 to enhance the apoptotic effects in UCUB and

CDDP-resistant UCUB cells in vitro and in vivo. The study also describes the first

preclinical evaluation of S3I-201 in UCUB and supports that CEBPD inhibition can

reduce drug resistance and promote therapeutic efficiency in CDDP-treated UCUB.

4

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

UCUB is the fourth and tenth most common malignancy in men and women,

respectively. At present, CDDP is the most common agent for treating locally

advanced and metastatic UCUB. However, the intrinsic and acquired CDDP

resistance of UCUB is a major clinical problem, which eventually results in treatment

failure (1). In addition, PTX, a microtubule dissociation inhibitor, is a widely accepted

combination agent for use in CDDP in UCUB therapy. However, CDDP-resistant

tumors (including UCUB) are not completely responsive to salvage combination

chemotherapy involving PTX (2). Therefore, novel therapeutic combinations or

treatments are required to reverse CDDP-induced PTX cross-resistance.

Approximately 50% of bladder tumors exhibit strong immunohistochemical

staining for EGFR, which is also associated with muscle invasion and poor tumor

differentiation (3, 4). In addition, a subgroup of muscle-invasive bladder carcinoma

displaying a basal-like phenotype is sensitive to EGFR inhibitor-erlotinib (5).

However, the underlying mechanisms remained unknown. Gefitinib, also known as

ZD1839, selective EGFR tyrosine kinase antagonist that inhibits tumor cell growth (6),

attenuates UCUB cell proliferation and enhances apoptosis (7). However, the detailed

mechanisms underlying the effects of EGFR in UCUB cells drug resistance, remain

largely unknown.

Under normal physiological conditions, the expression levels of transcription

factor CEBPD is relatively low, but the levels can be upregulated by various

extracellular stimuli (8). CEBPD is also responsive to several anticancer drugs,

including vitamin D3 (9) and

1-(2-hydroxy-5-methylphenyl)-3-phenyl-1,3-propanedione (HMDB) (10). Several

studies have suggested that the p38/CREB (cAMP responsive element binding protein)

5

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and JAK (Janus kinase)/STAT3 (signal transducer and activator of transcription 3)

pathways play crucial roles in CEBPD transcriptional activation (10, 11). Previous

studies have also suggested that CEBPD plays a vital role in inflammation and

inflammatory disease processes (12, 13). However, unlike the inflammatory effectors

nuclear factor-B and STAT3 that are consistently activated in cancer cells, CEBPD

inactivation has been observed in several types of cancer, including cervical and

hepatocellular carcinoma (10, 14), breast (15), prostate cancer (9), and leukemia (16).

In addition to acting as a tumor suppressor, several recent reports have suggested that

CEBPD plays an oncogenic role under certain conditions (17, 18). Furthermore,

CEBPD attenuation can sensitize CDDP-induced cell death in CDDP-resistant UCUB

cells (19). However, the details remain largely unclear, and the improvement in

CEBPD activation following CDDP and PTX cross-resistance remains

uninvestigated.

Multidrug resistance (MDR) transporter are most known for their

contribution to chemoresistance through the cellular efflux of anticancer drugs.

Several members of the ABC transporter family can induce MDR. To date, tumor

tissue studies have consistently revealed that in most cancer cells, the major

mechanism underlying MDR involves ABCB1, ABCC1, or ABCG2. ABCC2 endows

tumor cells with resistance to various anticancer drugs, including CDDP (20).

Furthermore, the ABC transporter family proteins have well-known specific

chemotherapy substrates: ABCC2 effluxes CDDP and mitoxantrone, whereas ABCC1

effluxes mitoxantrone and PTX (21). Currently, most evidence for additional roles is

correlative, and definitive studies are required to confirm the causality.

In this study, we showed that CDDP-resistant UCUB cells also exhibited

considerable PTX resistance, CEBPD responded to CDDP treatment, and CEBPD

6

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

activation contributed to ABCB1 and ABCC2 expression in UCUB cells. Furthermore,

CDDP-induced EGFR phosphorylation and gefitinib markedly inhibited CEBPD

levels in UCUB cells. We observed that STAT3 activation was EGF responsive and

sustained in CDDP-resistant NTUB1 (NTUB1/P) cells. Moreover, CEBPD expression

correlated with pEGFR and pSTAT3 levels in specimens from patients with UCUB

receiving postoperative chemotherapy. The ABC transporters ABCB1 and ABCC2

were upregulated by CDDP-induced CEBPD through transcriptional regulation and

contributed to CDDP resistance and CDDP-induced PTX cross-resistance. Finally,

CDDP administered in combination with either gefitinib or S3I-201 significantly

inhibited viability of CDDP-resistant UCUB cells and tumor growth. Overall, our

results suggest that gefitinib and S3I-201 are useful adjuvants for treating

CDDP-resistant UCUB, particularly for patients who are administered these drugs in

combination with CDDP or PTX.

7

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Materials and Methods

Patients and Tumor Specimens

This study was approved by the Institutional Review Board (approval number

IRB10302015) of the Chi Mei Medical Center, Tainan City, Taiwan. All samples

were obtained from the Chi Mei Medical Center BioBank and were previously

collected according to the official ethical guidelines. We retrieved UCUB cases

(1996–2004) from the aforementioned medical center archives for

immunohistochemical and survival analysis, as previously described (22). To evaluate

the significance of CEBPD expression with respect to a CDDP-based chemotherapy

response, we enrolled 79 UCUB cases with pT2 to pT4 tumors or with nodal

involvement in which CDDP-based postoperative adjuvant chemotherapy was applied.

To closely validate the significant findings, 60 independent cases were collected from

Liou Ying Campus of Chi Mei Healthcare system. The clinicopathological evaluation

criteria were essentially identical to those used in our previous study (23). An expert

pathologist (C.F.L) reevaluated hematoxylin and eosin-stained sections from each

case.

Cell Lines and Culture Conditions

J82, TSGH8301, and TCCSUP cells were maintained in Dulbecco’s modified

Eagle’s medium supplemented with 10% fetal bovine serum (FBS), 100 units/mL

penicillin, and 100 mg/mL streptomycin. The J82 and TCCSUP cells were purchased

from American Type Culture Collection. NTUB1 (24) and the TSGH8301 (25)

cell lines were obtained from and authenticated by Dr. Tzyh-Chyuan Hour. The

NTUB1 and nasopharyngeal cancer HONE1 cells were maintained in RPMI 1640

media supplemented with 10% FBS, 100 units/mL penicillin, and 100 mg/mL

8

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

streptomycin. Stable CDDP-resistant NTUB1/P and HONE1 (HONE1/R) clones were

maintained in the aforementioned media with 10 μM CDDP (Sigma).

Cell Viability and Cell Death Assays

Cell survival was measured using the Cell Counting Kit-8 (Sigma). Experimental

cells were treated with either CDDP (20 M; BioVision) or PTX (1 M; BioVision)

for 24 h. For the combination treatment, the cells were treated with CDDP or PTX

plus either gefitinib (5 M; BioVision) or S3I-201 (10 M; BioVision) for 24 h. The

percentages of cell viability and death were determined for each treatment through

comparisons with the untreated control group. For cell death assays, the cells were

plated and cultured in regular media as previously mentioned for 16 h and then treated

with CDDP alone or in combination with either gefitinib or S3I-201 for another 24 h.

The cells were stained with propidium iodide (Sigma) and analyzed through flow

cytometry.

Chromatin Immunoprecipitation–PCR Assay

The chromatin immunoprecipitation assay was performed, as previously described

(13). In brief, the experimental cells were treated with 1% formaldehyde for 15 min,

and the cross-linked chromatin was prepared and sonicated to an average size of 500

bp. The DNA fragments were immunoprecipitated using either specific antibodies

recognizing CEBPD or control rabbit immunoglobulin G (IgG) at 4 °C for 12–16 h.

Following cross-link reversal, quantitative real-time PCR assays were performed

using the precipitated DNA and primers corresponding to specific target regions.

The primers were as follows:

CEBPD-A (A), 5ʹ-GCCAAGTCCTGGTTTTGATT-3ʹ; CEBPD (AS), 5ʹ

9

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

-GCCCCCTCTCAGTTCCTC-3ʹ; CEBPD-B (A),

5ʹ-GGTTTCACCATGTTGACCAG-3ʹ; CEBPD-B (AS),

5ʹ-AGAATGGGCTTTGTCATGTG-3ʹ; ABCB1-A (A),

5ʹ-CTCTGGTACTGGGATAAACACTTGTA-3ʹ; ABCB1-A (AS),

5ʹ-TGATTGCAAACTTCTAGTCAAGACA-3ʹ; ABCB1-B (A),

5ʹ-CCTGGACCCAGCTTTCACTA-3ʹ; and ABCB1-B (AS),

5ʹ-AGCACAAATTGAAGGAAGGAG-3ʹ. The amplified products were resolved

using agarose gel electrophoresis and confirmed using sequencing.

Lentivirus Knockdown Assays

Viruses were produced in Phoenix cells by cotransfecting various small hairpin

RNA (shRNA) expression vectors with pMD2.G and psPAX2. The lentiviral

knockdown expression vectors were obtained from the National RNAi Core Facility

(Genomic Research Center of the Institute of Molecular Biology, Academia Sinica,

Taiwan). After the viral infection efficiency was determined, lentiviruses containing

sh-galactosidase (shLacZ) or shCEBPD were used for infecting NTUB1/P cells for

72 h at a multiplicity of infection. For all lentiviral experiments, media containing

uninfected viruses were removed before further analysis. The shRNA sequences in the

lentiviral expression vectors were as follows: shLacZ,

5ʹ-CCGGTGTTCGCATTATCCGAACCATCTCGAGATGGTTCGGATAATGCGA

ACATTTTTG-3ʹ; shCEBPD, 5ʹ-CCGGGCCGACCTCTTCAACAGCAATC

TCGAGATTGCTGTTGAAGAGGTCGGCTTTTT-3ʹ; shABCB1,

5ʹ-CCGGGCTGCTTTCCTGCTGATCTATCTCGAGATAGATCAGCAGGAAAGC

AGCTTTTTG-3ʹ and shABCC2,

5ʹ-CCGGCCTGGTGGATAGCAACAATATCTCGAGATATTGTTGCTATCCACC

10

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

AGGTTTTTG-3ʹ.

MDR Pump Activity Assays

The experimental cells were plated overnight in a growth medium at a

concentration of 60000 cells/well/90 μL in 96-well plates. The following day, the

cells were treated with CDDP and gefitinib or S3I-201 for 24 h. Furthermore, the cells

were mixed with 100 μL/well of MDR dye-loading solution (Abcam) and incubated

for 4 h at room temperature. Sample fluorescence intensity was then measured at 525

nm by using a fluorescence reader (Thermo).

Immunohistochemical Staining

Tissue sections (4 μm) were cut from paraffin-embedded tissue blocks, placed

onto precoated slides, deparaffinized, and rehydrated. Their antigens were retrieved,

and endogenous peroxidase was blocked, as previously described (23). Moreover, the

slides were incubated for 1 h with primary antibodies for CEBPD (1:200; ab65081;

Abcam), pEGFR (1:25; Tyrosine 1086-specific, Zymed), pSTAT3 (1:25; Tyrosine

705-specific, Santa Cruz), and ABCB1 (1:100, C-19, Santa Cruz). We then detected

the primary antibodies by using the DAKO ChemMate EnVision kit (K5001;

Carpinteria, CA, USA). To ensure immunostaining quality, a sample prepared without

the primary antibody served as the negative control.

Immunohistochemistry Interpretation and Scoring

An expert pathologist (C.F.L), blinded to clinical and follow-up data, evaluated

the immunohistochemistry results. Immunoreactivity was evaluated using both the

percentage and intensity of positively stained tumors, illustrating representative

11

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

staining patterns (pEGFR, membranous and cytoplasmic; pSTAT3, nuclear; CEBPD,

nuclear; ABCB1, cytoplasmic) to calculate the H-score according to the following

equation: H-score = ∑Pi(i +1), where i is the intensity of the stained tumor cells (0–3),

and Pi is the percentage of stained tumor cells for each intensity (0%–100%). This

formula generates a score between 100 and 400; 100 and 400 indicate that 100% of

the tumor cells were negative (0) and strongly stained (3+), respectively (23).

Animal Studies

Male 6–8-weeks-old nonobese diabetic/severe combined immunodeficient

(NOD/SCID) mice were purchased from the Laboratory Animal Center of the

National Cheng Kung University. NTUB1/P cells (1 × 107) in 100 μL of PBS were

subcutaneously inoculated into the right flanks of the mice. Once macroscopic tumors

(100–125 mm3) formed, the animals (n = 4 per group) were randomly separated into 4

groups. The groups were then administered intraperitoneal treatments as follows: the

control group received the vehicle (PBS), the CDDP group received CDDP (5

mg/kg/2 d) for 3 weeks, the CDDP plus gefitinib group received CDDP (5 mg/kg/2 d)

and gefitinib (10 mg/kg/2 d) for 3 weeks, and the CDDP plus S3I-201 group received

CDDP (5 mg/kg/2 d) and S3I-201 (10 mg/kg/2 d) for 3 weeks. Animal weights and

tumor dimensions were measured every 3 d by using calipers, and tumor volumes

were estimated from the tumor length and width by using the formula [L x W2]/2,

where L and W are the length and width, respectively.

Statistical Analysis

Statistical analysis was performed using SPSS V.14.0 software (SPSS Inc.

Chicago, IL, USA). The median immunohistochemistry H-scores for CEBPD, pEGFR,

pSTAT3, and ABCB1 were used as cutoffs for separating the cases into high and low 12

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

expression groups. Furthermore, a chi-square test was used to assess the associations

among CEBPD, pEGFR, pSTAT3, and ABCB1 expression. The endpoint analyzed

was metastasis-free survival (MeFS), which was calculated from the dates of curative

surgery and development of distal spreading. Patients lost to follow-up were censored

on the final follow-up date. We plotted survival curves by using the Kaplan–Meier

method and evaluated predictive differences between the groups by using the log-rank

test. For all analyses, 2-sided tests of significance were conducted, with P < 0.05

considered significant. The statistical significance of the variations between the mean

values was estimated using the SigmaPlot software package and independent Student

t test for unequal variances. The data were expressed as the mean ± standard error of

the mean, and P < 0.05 was considered statistically significant.

13

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Results Significance of CEBPD immunoreactivity in UCUB

UCUB cells exhibited nuclear CEBPD. After dichotomizing the tumors into low

and high CEBPD expression groups, we found that increased CEBPD expression was

significantly associated with nodal involvement (P = 0.013; Supplementary Table 1).

Notably, UCUB patients with high CEBPD levels had significantly worse MeFS (P =

0.0077; Supplementary Fig. 1A), suggesting a potential role of CEBPD in UCUB

progression following postoperative adjuvant chemotherapy. Of note, the

aforementioned findings remained significant in our independent cohort

(Supplementary Fig. 1B)

CEBPD expression is increased in CDDP-resistant UCUB cell lines and contributes to

CDDP resistance and CDDP-induced PTX cross-resistance

To investigate the involvement of CEBPD in the development of CDDP resistance,

we evaluated the association of CEBPD expression and CDDP sensitivity in UCUB

cell lines, including J82, TSGH8301, TCCSUP, and NTUB1. CEBPD abundance was

correlated with UCUB cell resistance to CDDP-induced cell death (Fig. 1A). Our

previous studies suggested that CEBPD is responsive to anticancer drugs (26, 27). We

further assessed whether CDDP affected CEBPD expression in UCUB cells. In

addition to the increased basal CEBPD levels being observed in CDDP-resistant

NTUB1/P cells, further CEBPD induction was observed after CDDP treatment in

NTUB1/P cells as well as other UCUB cells (Fig. 1B). These results suggested not

only that CEBPD expression is associated with CDDP resistance in UCUB cells but

also that CDDP-induced CEBPD expression leads to drug resistance of UCUB cells.

Cancer cells evade death and exhibit increased drug efflux that contributes to drug

resistance. As previously mentioned, CDDP treatment could lead to PTX 14

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cross-resistance; however, the underlying mechanisms in UCUB cells remain unclear.

We assessed whether CDDP-resistant NTUB1/P cells were also PTX insensitive.

NTUB1/P cells exhibited attenuated PTX sensitivity compared with NTUB1 cells

(Fig. 1C). Furthermore, we examined the involvement of CEBPD in CDDP resistance

and CDDP-induced PTX resistance in our system. The loss-of-function assays using

shLacZ and shCEBPD were performed to verify the role of CEBPD in CDDP

resistance and CDDP-induced PTX resistance. The results revealed that the

attenuation of CEBPD levels by shCEBPD led to increased CDDP sensitivity and

CDDP-induced PTX resistance in NTUB1/P cells (Fig. 1D). We also assessed

whether CEBPD contributes to an increased drug efflux. In NTUB1/P cells, the loss

of CEBPD enhanced cellular fluorescence intensity in the MDR pump activity assay

(Fig. 1E), suggesting that CEBPD contributes to the increase in drug efflux.

EGFR inhibitor significantly enhances CDDP sensitivity in drug-resistant UCUB cells

by reducing the CEBPD activity

A recent study reported the involvement of EGFR in drug-resistant UCUB (28);

however, the mechanisms of EGFR-mediated drug resistance in UCUB remain

unclear. We observed that CDDP induced EGFR expression and activity in NTUB1/P

cells but not in NTUB1 cells (Fig. 2A). We then examined the EGFR loss of function

by treating UCUB cells with acquired resistance with an EGFR inhibitor. This assay

revealed that gefitinib treatment alone significantly induced cell death in NTUB1/P

cells; however, it only marginally affected NTUB1 cells (Fig. 2B). Furthermore, we

analyzed the anticancer effects of gefitinib and CDDP cotreatment. In the combined

treatment, gefitinib increased NTUB1/P cell sensitivity to CDDP (Fig. 2C, compare

lanes 4 and 8) and promoted NTUB1 cell death (Fig. 2C, compare lanes 3 and 7).

15

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

These results imply that EGFR plays a critical role in the CDDP resistance of

NTUB1/P cells.

Abrogating STAT3 activation inhibits CEBPD expression and enhances CDDP

efficiency in CDDP-resistant UCUB

CDDP induced EGFR signaling in NTUB1/P cells (Fig. 2), and activated p38

MAPK and STAT3 have been suggested to play crucial roles in CEBPD

transcriptional activation (8). Therefore, we analyzed STAT3 and p38 activation in

the EGF-treated NTUB1/P cells, revealing that EGF induced the activation of STAT3

(pY705) but not p38 MAPK in NTUB1/P cells (Fig. 2D). In addition to CEBPD

suppression through attenuated STAT3 activation, CEBPD was suppressed in

gefitinib-treated NTUB1/P cells (Supplementary Fig. 2A). To further determine

whether STAT3 mediated the EGF-induced CEBPD expression in UCUB cells, we

examined S3I-201 (29, 30). We first determined the effects of S3I-201 on NTUB1/P

cells treated with either CDDP or EGF. Similar to our findings in gefitinib treatment

in CDDP-treated NTUB1/P and J82 cells, S3I-201 inhibited CDDP- or EGF-induced

STAT3 activation and CEBPD expression (Fig. 2E and Supplementary Fig. 2B).

Moreover, an in vivo DNA binding assay revealed that the binding of activated

STAT3 (pY705) in response to CDDP treatment was attenuated in NTUB1/P cells

treated with both CDDP and S3I-201 (Fig. 2F).

CEBPD mediates CDDP-induced ABCB1 expression

As previously mentioned, ABC transporters contribute to drug resistance by

increasing drug efflux, thereby reducing toxicity. Therefore, we assessed which ABC

transporters could respond to CEBPD induction in NTUB1/P cells. Among the ABC

transporters, ABCB1 and ABCC2 demonstrated specific transcript upregulation in 16

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

response to CEBPD treatment (Supplementary Fig. 3A). High ABCB1 levels were

observed in patients with UCUB (31, 32) and have been suggested to be involved in

PTX resistance. We observed that ABCB1 transcripts were present at higher levels in

NTUB1/P cells and were further activated in NTUB1/P cells treated with CDDP;

however, this activation did not occur in NTUB1 cells (Fig. 3A). Furthermore,

exogenously expressed CEBPD induced ABCB1 transcription in NTUB1/P cells (Fig.

3B), and CEBPD knockdown attenuated the CDDP-induced ABCB1 reporter activity

(Fig. 3C). RT-PCR was used for determining whether exon 1 of ABCB1 could be

transcribed in NTUB1/P cells (Supplementary Fig. 3B). Various 5ʹ-flanking regions

and ABCB1 fragments were cloned into a reporter vector for CEBPD-responsive

region identification (Fig. 3D, left panel, exon and intron mapping according to

ABCB1 cDNA clone NM_000927.4). Six putative CEBPD binding motifs within the

aforementioned fragments were predicted using the TFSEARCH program

(http://www.cbrc.jp/research/db/TFSEARCH.html), and a reporter assay revealed that

the −21/+386 bp region was significantly responsive to exogenous CEBPD expression

(Fig. 3D). To determine whether CEBPD can directly bind the CEBPD-responsive

motifs within the −21/+386 bp region, we performed a ChIP assay. The results

showed CEBPD binding, which was responsive to CDDP treatment in NTUB1/P cells

(Fig. 3E). These results suggested that CDDP-induced CEBPD directly binds to

ABCB1 exon 1 and upregulates its transcription.

Gefitinib and S3I-201 reduce ABCB1 and ABCC2 transcript levels and enhance

CDDP and PTX sensitivity in CDDP-resistant NTUB1/P and J82 cells

We next assessed ABCC2 and ABCB1 mRNA levels following the treatment with

either CDDP and gefitinib or CDDP and S3I-201 in NTUB1/P and J82 cells. The

17

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

ABCC2 and ABCB1 transcript levels were responsive to CDDP treatment; however,

they were suppressed in NTUB1/P (Fig. 4A) and J82 cells (Supplementary Figs. 4A

and 4B), following the treatment with either gefitinib or S3I-201. In addition, an

efflux assay assessing the ABCB1 transporter activity was used for examining

CDDP-induced PTX resistance. The results revealed that gefitinib and S3I-201 inhibit

fluorescent MDR indicator efflux in NTUB1/P and J82 cells (Fig. 4B and

Supplementary Fig. 4C). These results suggested that gefitinib- and

S3I-201-attenuated ABCB1 enhanced CDDP cytotoxicity in CDDP-resistant UCUB

cells.

Our results suggested that the EGF/pSTAT3 signaling axis mediates

CDDP-induced CEBPD transcription and that increased CEBPD contributes to

ABCB1 transcription and expression in CDDP-resistant UCUB cells. We further

demonstrated that gefitinib and S3I-201 increased NTUB1/P and J82 cell sensitivity

to CDDP and PTX (Figs. 4C and 4D and Supplementary Fig. 4D, respectively).

These results suggested that gefitinib and S3I-201 recover CDDP and PTX sensitivity

in CDDP-resistant cells. In addition, CDDP and PTX are the substrate of ABCC2 and

ABCB1, respectively (21). To further assess the specific involvement of ABCB1 and

ABCC2 in cross-resistance, a cell viability assay was conducted in ABCB1 or

ABCC2 knockdown NTUB1/P cells. The results showed that the loss of ABCC2 in

NTUB1/P cells is sensitive to CDDP but miner to PTX treatment (Fig. 4E). Moreover,

the loss of ABCB1 in NTUB1/P cells is specifically sensitive to PTX but not to

CDDP treatment (Fig. 4F). In addition, to verify that CEBPD expression is correlated

with pEGFR, pSTAT3, and ABCB1 expression in patients with UCUB, we performed

immunohistochemical assays. The results revealed that CEBPD expression was

significantly associated with EGFR and STAT3 activation (P = 0.010 and <0.001,

18

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

respectively). Moreover, ABCB1 expression in UCUB was significantly associated

with high CEBPD and pSTAT3 levels (P < 0.001; Table 1 and Supplementary

Table 2 and Fig. 5).

As previously mentioned, S3I-201 can abolish CEBPD activation to promote the

CDDP sensitivity of CDDP-resistant UCUB cells. To determine whether CEBPD and

ABCB1 were also responsive to CDDP treatment and were suppressed by gefitinib

and S3I-201, we studied a CDDP-resistant nasopharyngeal HONE1 cancer cell line,

HONE1/R (Supplementary Fig. 4E). CEBPD and ABCB1 expression was higher in

HONE1/R cells than in parental HONE1 cells (Supplementary Fig. 4F). As observed

for NTUB1/P cells, S3I-201 also inhibited ABCB1 transporter efflux activity

(Supplementary Fig. 4G) and sensitized HONE1/R cells to CDDP (Supplementary

Fig. 4H).

Gefitinib and S3I-201 significantly enhance CDDP therapeutic efficacy in

CDDP-resistant UCUB

Our in vitro results suggested that gefitinib and S3I-201 treatment significantly

sensitized CDDP-resistant UCUB cells to CDDP. To further evaluate these results in

vivo, the effects of CDDP, gefitinib, S3I-2-1, combinations of CDDP/gefitinib and

CDDP/S3I-201 treatments were examined in a NTUB1/P-xenografted NOD/SCID

mouse model. In accordance with previous results, gefitinib and S3I-201 have a fewer

effect on NTUB1/P tumor burden (58% and 52% inhibition, respectively, at day 9).

Importantly, CDDP/gefitinib and CDDP/S3I-201 treatments significantly reduced the

tumor burden (82% and 81% inhibition, respectively, at day 9) and weight (Fig. 5).

Furthermore, pSTAT3, CEBPD, and ABCB1 levels in NTUB1/P tumors were

verified. As shown in Supplementary Figs. 6A and 6B, pSTAT3, CEBPD, and

19

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

ABCB1 levels and ABCC2 mRNA levels were reduced in the CDDP/gefitinib- and

CDDP/S3I-201-treated NTUB1/P xenografted NOD/SCID mice; this result was

consistent with the reduction in tumor size.

20

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Discussion CDDP and PTX are widely used in UCUB therapy. CDDP ultimately triggers cell

death and is commonly used in the clinical therapy of many cancers, including UCUB.

Resistance can be pre-existing (intrinsic) or drug induced (acquired). As previously

mentioned, UCUB therapy remains associated with persistent high rates of local and

distant failure resulting from the acquisition of chemoresistance (33). CEBPD

expression was higher in patients with UCUB than in patients with normal urothelial

tissue (34). Our findings indicated that CEBPD upregulated ABCB1 and ABCC2

expression, revealing an increased drug efflux activity in CEBPD-abundant UCUB

cells. These results suggested not only that CEBPD plays a functional role in intrinsic

and acquired resistance but also explained why patients with UCUB tend to develop

drug resistance and become desensitized to CDDP and PTX (35, 36).

CEBPD is a CDDP-responsive gene. In this study, we demonstrated that the

EGFR/STAT3 signaling axis participates in CDDP-induced CEBPD transcription in

UCUB cells, contributing to CDDP and CDDP-induced cross-resistance to PTX by

activating ABCC2 and ABCB1 expression (Fig. 3). This study also provided a novel

insight into the mechanism underlying acquired resistance and linked EGFR signaling

with ABCB1-mediated PTX cross-resistance in CDDP-resistant UCUB. Furthermore,

treatment with either gefitinib or S3I-201 reduced ABCB1 expression in

CDDP-resistant HONE1 cells and in CDDP-resistant UCUB cells. Thus, CEBPD

plays a functional role in promoting drug resistance, and CEBPD inhibition may be an

effective approach for reducing the occurrence of drug resistance.

Forced CEBPD overexpression induces growth arrest and death in cancer cells (9).

Increased CEBPD levels have been observed in response to treatment with HMDB

(10), dexamethasone (37), retinoic acid (37), and 5-azacytidine (38). Furthermore,

CEBPD have been suggested to contribute to apoptosis. It has been recently suggested 21

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

to play dual roles in pro and antitumor processes under certain conditions (39-41).

Furthermore, upregulated caspase 3 and caspase 8 activity has been reported in

response to CEBPD induction in prostate cancer (41), and CEBPD was involved in

antiapoptosis in astroglioma U373MG cells by upregulating the antiapoptotic gene

ZNF179 (40). These observations imply that CEBPD may serve dual roles in

controlling cell survival and regulating cell fate in a cell-specific manner. Herein, we

revealed that the EGFR/STAT3 pathway contributes to CEBPD activation and confers

a drug-resistant activity that includes cross-resistance. In addition, a previous study

showed that the overexpression of EGFR is sensitive to CDDP treatment through a

ligand-independent pathway in U87 glioma and MB468 breast cancer cells (42). In

our system, the basal levels of pEGFR and pSTAT3 are higher and coincident with

basal CEBPD abundance in NTUB1/P cells (Fig. 1A and Supplementary Fig. 7).

Therefore, treatment of CDDP could further enhance the activities of pEGFR and

pSTAT3 and coordinately CEBPD abundance in NTUB1/P cells (Fig. 2A), indicating

a unique but unknown protein/pathway is responsive to CDDP in the CDDP-resistant

cells to strengthen the activation of pEGFR and pSTAT3. More importantly, it also

provided a motivation for initiation of clinical studies with inhibition of the

EGFR/STAT3 signaling pathway when combined with CDDP to prevent the

development of therapeutic resistance and thus significantly impact overall survival in

patients with UCUB.

P-gp was the first factor known to modulate MDR and is the most widely studied

ATP-dependent drug efflux pump. P-gp is encoded by ABCB1 and is considered to

mediate MDR by reducing the intracellular accumulation of cytotoxic drugs and

compounds. Many types of cancers express high ABCB1 levels and are unresponsive

to chemotherapy (43, 44); ABCB1 expression has been considered a predictor of poor

22

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

response to chemotherapy and overall survival (45, 46). Our results also indicated that

UCUB cells with high expression of CEBPD are strongly resistant to CDDP and PTX

because of ABCC2 and ABCB1 activation (Fig. 4), particularly in CDDP-resistant

UCUB cells. Thus, we speculated that CEBPD activation should, at least in part,

contribute to the proapoptotic activity in response to anticancer drugs; however, its

activation conferred acquired resistance via ABCB1 and ABCC2, promoting the

resistance of anticancer drug-induced cytotoxicity. In addition to forming a

homodimer, CEBPD can heterodimerize with other C/EBP family members, including

CEBPA and CEBPB (47). Moreover, ABCB1 is upregulated by CEBPD and CEBPB

in anticancer drug-resistant breast cancer cells (48). In accordance with this study, our

results also suggested the importance of examining the response and involvement of

CEBPB in CDDP-treated and -resistant UCUB cells in the future. The association

between ABC transporters with bladder cancers was previously established. Recent

studies have reported the overexpression of ABC transporters in chemotherapeutic

patients with bladder cancer (32, 49). CEBPD is also responsive to stimuli involving

the activation of either p38 MAPK or STAT3. Therefore, it is reasonable to speculate

that anticancer treatments, particularly those promoting CEBPD overexpression and

p38 MAPK and STAT3 activation, tend to lead to the development of drug-resistant

cancers.

Chemoresistance in several cancer types has been linked to STAT3 activation.

Recently, constitutively active STAT3 was shown to lead to cellular transformation,

suggesting an oncogenic role. STAT3 has also been suggested to confer an enhanced

cellular survival following genotoxic treatments (11). Moreover, STAT3 pathway

inhibition has been shown to result in growth arrest, apoptosis, and chemosensitivity

in several human malignancy models (50). We previously demonstrated that CEBPD

23

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

inhibition increased UCUB sensitivity to CDDP (19). In this study, we demonstrated

that CEBPD attenuation in UCUB and CDDP-resistant UCUB cells sensitized them to

CDDP and CDDP-induced PTX resistance (Fig. 1). We further revealed the novel

finding that ABCB1 responds to CEBPD upon CDDP treatment by activating EGFR

and STAT3 in UCUB and CDDP-resistant UCUB cells. Therefore, inactivating either

EGFR or STAT3 can sensitize UCUB and CDDP-resistant UCUB cells to CDDP and

even to PTX. In addition to gefitinib, we demonstrated that S3I-201 inhibited

CDDP-induced CEBPD and augmented CDDP and PTX sensitivity.

24

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Grant Support

This work was supported from the grant MOST103-2321-B-006-031,

MOST103-2320-B-006-034-MY3 from the Ministry of Science and Technology and

MOHW104- TDU-B-212–124-003 from the Ministry of Health and Welfare, Health

and welfare surcharge of tobacco products.

25

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References 1. Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4:307-20. 2. Bellmunt J, Guillem V, Paz-Ares L, Gonzalez-Larriba JL, Carles J, Albanell J, et al. Gemcitabine/paclitaxel-based three-drug regimens in advanced urothelial cancer. Eur J Cancer. 2000;36 Suppl 2:17-25. 3. Colquhoun AJ, Mellon JK. Epidermal growth factor receptor and bladder cancer. Postgrad Med J. 2002;78:584-9. 4. Neal DE, Sharples L, Smith K, Fennelly J, Hall RR, Harris AL. The epidermal growth factor receptor and the prognosis of bladder cancer. Cancer. 1990;65:1619-25. 5. Rebouissou S, Bernard-Pierrot I, de Reynies A, Lepage ML, Krucker C, Chapeaublanc E, et al. EGFR as a potential therapeutic target for a subset of muscle-invasive bladder cancers presenting a basal-like phenotype. Sci Transl Med. 2014;6:244ra91. 6. Ciardiello F, Caputo R, Bianco R, Damiano V, Pomatico G, De Placido S, et al. Antitumor effect and potentiation of cytotoxic drugs activity in human cancer cells by ZD-1839 (Iressa), an epidermal growth factor receptor-selective tyrosine kinase inhibitor. Clin Cancer Res. 2000;6:2053-63. 7. Shrader M, Pino MS, Lashinger L, Bar-Eli M, Adam L, Dinney CP, et al. Gefitinib reverses TRAIL resistance in human bladder cancer cell lines via inhibition of AKT-mediated X-linked inhibitor of apoptosis protein expression. Cancer Res. 2007;67:1430-5. 8. Wang JM, Tseng JT, Chang WC. Induction of human NF-IL6beta by epidermal growth factor is mediated through the p38 signaling pathway and cAMP response element-binding protein activation in A431 cells. Mol Biol Cell. 2005;16:3365-76. 9. Ikezoe T, Gery S, Yin D, O'Kelly J, Binderup L, Lemp N, et al. CCAAT/enhancer-binding protein delta: a molecular target of 1,25-dihydroxyvitamin D3 in androgen-responsive prostate cancer LNCaP cells. Cancer Res. 2005;65:4762-8. 10. Pan YC, Li CF, Ko CY, Pan MH, Chen PJ, Tseng JT, et al. CEBPD reverses RB/-mediated gene repression and participates in HMDB-induced apoptosis of cancer cells. Clin Cancer Res. 2010;16:5770-80. 11. Sanford DC, DeWille JW. C/EBPdelta is a downstream mediator of IL-6 induced growth inhibition of prostate cancer cells. Prostate. 2005;63:143-54. 12. Ko CY, Chang LH, Lee YC, Sterneck E, Cheng CP, Chen SH, et al. CCAAT/enhancer binding protein delta (CEBPD) elevating PTX3 expression inhibits -mediated phagocytosis of dying neuron cells. Neurobiol Aging. 2012;33:422. 11-25.

26

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

13. Wang JM, Ko CY, Chen LC, Wang WL, Chang WC. Functional role of NF-IL6beta and its sumoylation and acetylation modifications in promoter activation of cyclooxygenase 2 gene. Nucleic Acids Res. 2006;34:217-31. 14. Ko CY, Hsu HC, Shen MR, Chang WC, Wang JM. Epigenetic silencing of CCAAT/enhancer-binding protein delta activity by YY1/polycomb group/DNA methyltransferase complex. J Biol Chem. 2008;283:30919-32. 15. Tang D, Sivko GS, DeWille JW. Promoter methylation reduces C/EBPdelta (CEBPD) in the SUM-52PE human breast cancer cell line and in primary breast tumors. Breast Cancer Res Treat. 2006;95:161-70. 16. Gery S, Tanosaki S, Hofmann WK, Koppel A, Koeffler HP. C/EBPdelta expression in a BCR-ABL-positive cell line induces growth arrest and myeloid differentiation. Oncogene. 2005;24:1589-97. 17. Wu SR, Li CF, Hung LY, Huang AM, Tseng JT, Tsou JH, et al. CCAAT/enhancer-binding protein delta mediates alpha-induced Aurora kinase C transcription and promotes genomic instability. J Biol Chem. 2011;286:28662-70. 18. Balamurugan K, Wang JM, Tsai HH, Sharan S, Anver M, Leighty R, et al. The tumour suppressor C/EBPdelta inhibits FBXW7 expression and promotes mammary tumour metastasis. EMBO J. 2010;29:4106-17. 19. Hour TC, Lai YL, Kuan CI, Chou CK, Wang JM, Tu HY, et al. Transcriptional up-regulation of SOD1 by CEBPD: a potential target for cisplatin resistant human urothelial carcinoma cells. Biochem Pharmacol. 2010;80:325-34. 20. Taniguchi K, Wada M, Kohno K, Nakamura T, Kawabe T, Kawakami M, et al. A human canalicular multispecific organic anion transporter (cMOAT) gene is overexpressed in cisplatin-resistant human cancer cell lines with decreased drug accumulation. Cancer Res. 1996;56:4124-9. 21. Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer. 2010;10:147-56. 22. Li CF, Wu WJ, Wu WR, Liao YJ, Chen LR, Huang CN, et al. The cAMP responsive element binding protein 1 transactivates epithelial membrane protein 2, a potential tumor suppressor in the urinary bladder urothelial carcinoma. Oncotarget. 2015;6:9220-39. 23. Fan EW, Li CC, Wu WJ, Huang CN, Li WM, Ke HL, et al. FGF7 Over Expression Is an Independent Prognosticator in Patients with Urothelial Carcinoma of the Upper Urinary Tract and Bladder. J Urol. 2015;194:223-9 24. Yu HJ, Tsai TC, Hsieh TS, Chiu TY. Characterization of a newly established human bladder carcinoma cell line, NTUB1. J Formos Med Assoc. 1992;91:608-13.

27

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

25. Yeh MY, Yu DS, Chen SC, Lin MS, Chang SY, Ma CP, et al. Establishment and characterization of a human urinary bladder carcinoma cell line (TSGH-8301). J Surg Oncol. 1988;37:177-84. 26. Chi JY, Hsiao YW, Li CF, Lo YC, Lin ZY, Hong JY, et al. Targeting chemotherapy-induced PTX3 in tumor stroma to prevent the progression of drug-resistant cancers. Oncotarget. 2015;6:23987-4001. 27. Li CF, Tsai HH, Ko CY, Pan YC, Yen CJ, Lai HY, et al. HMDB and 5-AzadC Combination Reverses Tumor Suppressor CCAAT/Enhancer-Binding Protein Delta to Strengthen the Death of Liver Cancer Cells. Mol Cancer Ther. 2015;14:2623-33. 28. Kim WT, Kim J, Yan C, Jeong P, Choi SY, Lee OJ, et al. S100A9 and EGFR gene signatures predict disease progression in muscle invasive bladder cancer patients after chemotherapy. Ann Oncol. 2014;25:974-9. 29. Lin L, Deangelis S, Foust E, Fuchs J, Li C, Li PK, et al. A novel small molecule inhibits STAT3 phosphorylation and DNA binding activity and exhibits potent growth suppressive activity in human cancer cells. Mol Cancer. 2010;9:217. 30. Siddiquee K, Zhang S, Guida WC, Blaskovich MA, Greedy B, Lawrence HR, et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc Natl Acad Sci U S A. 2007;104:7391-6. 31. Clifford SC, Thomas DJ, Neal DE, Lunec J. Increased mdr1 gene transcript levels in high-grade carcinoma of the bladder determined by quantitative PCR-based assay. Br J Cancer. 1994;69:680-6. 32. Tada Y, Wada M, Kuroiwa K, Kinugawa N, Harada T, Nagayama J, et al. MDR1 gene overexpression and altered degree of methylation at the promoter region in bladder cancer during chemotherapeutic treatment. Clin Cancer Res. 2000;6:4618-27. 33. Piulats JM, Jimenez L, Garcia del Muro X, Villanueva A, Vinals F, Germa-Lluch JR. Molecular mechanisms behind the resistance of cisplatin in germ cell tumours. Clin Transl Oncol. 2009;11:780-6. 34. Wang YH, Wu WJ, Wang WJ, Huang HY, Li WM, Yeh BW, et al. CEBPD amplification and overexpression in urothelial carcinoma: a driver of tumor metastasis indicating adverse prognosis. Oncotarget. 2015;6:31069-84. 35. Yagoda A. Chemotherapy of Urothelial Tract Tumors. Cancer. 1987;60:574-85. 36. Gosland M, Lum B, Schimmelpfennig J, Baker J, Doukas M. Insights into mechanisms of cisplatin resistance and potential for its clinical reversal. Pharmacotherapy. 1996;16:16-39.

28

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

37. Yeh WC, Cao Z, Classon M, McKnight SL. Cascade regulation of terminal adipocyte differentiation by three members of the C/EBP family of proteins. Dev. 1995;9:168-81. 38. Weiland T, Weiller M, Kunstle G, Wendel A. Sensitization by 5-azacytidine toward death receptor-induced hepatic apoptosis. J Pharmacol Exp Ther. 2009;328:107-15. 39. Balamurugan K, Sterneck E. The many faces of C/EBPdelta and their relevance for inflammation and cancer. Int J Biol Sci. 2013;9:917-33. 40. Wang SM, Lee YC, Ko CY, Lai MD, Lin DY, Pao PC, et al. Increase of Protein 179 in Response to CCAAT/Enhancer Binding Protein Delta Conferring an Antiapoptotic Effect in Astrocytes of Alzheimer's Disease. Mol Neurobiol. 2015;51:370-82. 41. Chuang CH, Wang WJ, Li CF, Ko CY, Chou YH, Chuu CP, et al. The combination of the prodrugs perforin-CEBPD and perforin-granzyme B efficiently enhances the activation of caspase signaling and kills prostate cancer. Cell Death Dis. 2014;5:e1220. 42. Benhar M, Engelberg D, Levitzki A. Cisplatin-induced activation of the EGF receptor. Oncogene. 2002;21:8723-31. 43. Weinstein RS, Jakate SM, Dominguez JM, Lebovitz MD, Koukoulis GK, Kuszak JR, et al. Relationship of the expression of the multidrug resistance gene product (P-glycoprotein) in human colon carcinoma to local tumor aggressiveness and lymph node metastasis. Cancer Res. 1991;51:2720-6. 44. Fojo AT, Shen DW, Mickley LA, Pastan I, Gottesman MM. Intrinsic drug resistance in human kidney cancer is associated with expression of a human multidrug-resistance gene. J Clin Oncol. 1987;5:1922-7. 45. Melguizo C, Prados J, Luque R, Ortiz R, Rama AR, Caba O, et al. Modulation of multidrug resistance gene expression in peripheral blood mononuclear cells of lung cancer patients and evaluation of their clinical significance. Cancer Chemother Pharmacol. 2013;71:537-41. 46. Campa D, Muller P, Edler L, Knoefel L, Barale R, Heussel CP, et al. A comprehensive study of polymorphisms in ABCB1, ABCC2 and ABCG2 and lung cancer chemotherapy response and prognosis. Int J Cancer. 2012;131:2920-8. 47. Takiguchi M. The C/EBP family of transcription factors in the liver and other organs. Int J Exp Pathol. 1998;79:369-91. 48. Conze D, Weiss L, Regen PS, Bhushan A, Weaver D, Johnson P, et al. Autocrine production of causes multidrug resistance in breast cancer cells. Cancer Res. 2001;61:8851-8.

29

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

49. Nakagawa M, Emoto A, Nasu N, Hanada T, Kuwano M, Cole SP, et al. Clinical significance of multi-drug resistance associated protein and P-glycoprotein in patients with bladder cancer. J Urol. 1997;157:1260-4; discussion 4-5. 50. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798-809.

30

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends

Figure 1. CEBPD expression is responsive to CDDP and is associated with CDDP

and PTX cross-resistance

A, CDDP affects cell viability and CEBPD expression in various UCUB cells. UCUB

cell lines were treated with various concentrations of CDDP for 24 h. CEBPD

expression levels were evaluated using Western blotting with the indicated antibodies,

and cell viability was examined using the CCK8 assay. TSGH8301 (TSG), TCCSUP

(TCC), NTUB1 (NTU), and CDDP-resistant NTUB1/P (NTP) cells.

B, CEBPD expression is responsive to CDDP treatment and sustained in all UCUB

cells. A Western blot was conducted with lysates harvested from CDDP-treated all

UCUB cell lines for 24 h. Western blot analysis of CDDP-treated UCUB cell lysates.

C, NTP cells are insensitive to PTX treatment. NTU and NTP cells were treated with

or without PTX and assessed for cell viability by using the CCK8 assay after 24 h of

incubation.

D, CEBPD attenuation in NTP cells sensitizes them to CDDP and PTX. NTP cells

were pretreated with lentiviruses containing either shLacZ or shCEBPD (shCD1 and

shCD2). Following 48 h of incubation, experimental cells were treated with CDDP or

PTX for 24 h and then assessed for cell viability by using the CCK8 assay.

E, CEBPD affects the ABC transporter-mediated pump activity in NTP cells. NTP

cells were pretreated with lentiviruses containing either shLacZ or shCEBPD (shCD2).

The cells were then treated with or without CDDP. After 24 h, an MDR dye-loading

solution was incubated in each well for 4 h, and the resulting fluorescence intensity

was detected using an ELISA reader. In this figure, *** and ** denote a significant

difference with P < 0.001 and <0.05, respectively. CTL: control

31

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2. Abrogation of EGFR and STAT3 activation inhibits EGF and

CDDP-induced CEBPD expression in CDDP-resistant NTUB1/P cells

A, EGFR and STAT3 expression and activity levels increased in NTUB1/P (NTP)

cells. Western blot analysis was performed using lysates from NTUB1 (NTU) and

NTP cells treated with CDDP for 24 h and the indicated specific pEGFR (pY1085),

pSTAT3 (pY705), EGFR, STAT3, and α-tubulin antibodies.

B, gefitinib attenuated NTU and NTP cell survival. NTU and NTP cells were treated

with gefitinib at the indicated doses for 24 h, and cell death was examined using

propidium iodine staining.

C, gefitinib enhances NTU and NTP cell sensitization to CDDP. NTU and NTP cells

were treated with either gefitinib or CDDP for 24 h, and cell death was assayed using

PI staining.

D, CEBPD expression in NTP cells is associated with STAT3 activity following EGF

treatment. NTP cells were treated with EGF for the indicated periods, and lysates

were harvested at the indicated time points. Antibodies recognizing pSTAT3 (pY705),

STAT3, pp38 (T182/Y180), p38, CEBPD, and α-tubulin were used for Western blot

analysis.

E, a STAT3 inhibitor, S3I-201, inhibits EGF and CDDP-activated CEBPD expression.

Western blot analysis was performed using the indicated antibodies and lysates of

cells pretreated with S3I-201 for 1 h and then stimulated with EGF or CDDP for 24 h.

F, CDDP enhances binding of STAT3 to the CEBPD promoter in NTP cells.

Sonicated chromatin was employed in ChIP assays by using STAT3 or control IgG

antibodies and then subjected to PCR analysis after eliminating DNA-binding proteins,

as described in the Materials and Methods. In this figure, ***, **, and * denote a

significant difference with P < 0.001, <0.01, and <0.05, respectively.

32

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 3. CEBPD upregulates ABCB1 expression following CDDP treatment

A, ABCB1 transcripts are abundant in NTUB1/P (NTP) cells. qPCR assays were

performed using total RNA harvested from NTUB1 (NTU) or NTP cells treated with

CDDP for 24 h.

B, CEBPD induces ABCB1 transcription. qPCR assays were performed using the total

RNA harvested from NTP cells transfected with expression vectors with and without

CEBPD cDNA (CD and control [CTL], respectively).

C, Loss of CEBPD attenuates CDDP-induced ABCB1 reporter activity. NTP cells

were pretreated with lentiviruses containing shLacZ or shCEBPD (shCD2),

transfected with ABCB1 reporter (AB-I, diagram shown as follows), and treated with

or without CDDP. Luciferase assays were performed after 24 h by using lysates

harvested from the experimental cells.

D, Identification of the CEBPD-responsive region within the ABCB1 5ʹ-flanking

region and intron 1. Schematic of the putative CEBPD motifs (oval) within the human

ABCB1 5ʹ-flanking region and exon/intron 1 (−1000 to +917 bp) and various ABCB1

reporters. For reporter assays, NTP cells were transfected with the indicated ABCB1

reporters and pCDNA3/HA with or without CEBPD cDNA (CD and CTL,

respectively).

E, CDDP enhances CEBPD binding to the ABCB1 region containing putative CEBPD

binding motifs in NTP cells. Sonicated chromatin was subjected to ChIP–PCR

analysis by using CEBPD or control IgG antibodies, as described in the Materials and

Methods. In this figure, *** denotes a significant difference (P < 0.001).

33

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 4. EGFR/STAT3 inhibition attenuates transporter efflux and promotes

CDDP- or PTX-induced death in NTUB1/P cells

A, gefitinib and S3I-201 inhibit CDDP-induced ABCC2 and ABCB1 transcription.

NTUB1/P (NTP) cells were treated with CDDP alone or in combination with either

S3I-201 or gefitinib for 24 h. qPCR assays were used for measuring ABCC2 and

ABCB1 transcript levels.

B, gefitinib and S3I-201-mediated the inhibition of ABCB1 transporter pump activity

in NTP cells. NTP cells were treated with CDDP alone or in combination with either

gefitinib or S3I-201 for 24 h. An MDR dye-loading solution was then added to each

well and incubated for 4 h. Fluorescence intensity was then detected using an ELISA

reader.

C and D, gefitinib and S3I-201 enhance NTP cells sensitivity to CDDP and PTX.

NTP cells were treated with CDDP or PTX alone or combination with or without

either gefitinib or S3I-201 for 24 h. Cell viability was measured using the CCK8

assay. In this figure, *** denotes a significant difference (P < 0.001).

E and F, attenuation of ABCC2 and ABCB1 in NTP cells sensitizes to CDDP and

PTX, respectively. NTP cells were pretreated with lentiviruses containing shLacZ

(LacZ) or shABCC2 or shABCB1. After 48 h of incubation, experimental cells were

treated with CDDP or PTX for 24 h and then examined for cell viability by CCK8

assay. In this figure, ***, significant difference (P < 0.001). **, significant difference

(P < 0.05).

34

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 5. Antitumor effects of CDDP treatment alone or in combination with

either gefitinib or S3I-201 on NTUB1/P-xenografted NOD/SCID mice

Effects of the intraperitoneal administration of CDDP, gefitinib, S3I-201,

CDDP/gefitinib, or CDDP/S3I-201 on NTUB1/P (NTP)-xenografted NOD/SCID

mice. A total of 24 NOD/SCID mice were subcutaneously inoculated with NTP cells

(1 × 107) and divided into 6 groups. Following solid tumor formation, the animals

were intraperitoneally injected with a control vehicle or CDDP (5 mg/kg/2 d),

gefitinib (10 mg/kg/2 d), S3I-201 (10 mg/kg/2 d), CDDP and gefitinib, or CDDP and

S3I-201.

35

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 1. Correlations between expression status of pEGFR, pSTAT3, CEBPD, and ABCB1 in muscle invasive UCUB Parameter Expression Case pSTAT3 Exp. p-value CEBPD Exp. p-value ABCB1 Exp. p-value status No. Low High Low High Low High pEGFR Low Exp. 39 23 16 0.092 25 14 0.010* 25 14 0.010* High Exp. 40 16 24 14 26 14 26 pSTAT3 Low Exp. 39 - - - 29 10 <0.001* 28 11 <0.001* High Exp. 40 - - - 10 30 11 29 CEBPD Low Exp. 39 ------32 7 <0.001* High Exp. 40 ------7 33 * Statistically significant

1

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2016; DOI: 10.1158/1078-0432.CCR-15-1169 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Inhibition of EGFR/STAT3/CEBPD axis reverses cisplatin cross-resistance with paclitaxel in urothelial carcinoma of urinary bladder

Wei-Jan Wang, Chien-Feng Li, Yu-Yi Chu, et al.

Clin Cancer Res Published OnlineFirst July 19, 2016.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-15-1169

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2016/07/19/1078-0432.CCR-15-1169.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet Manuscript been edited.

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2016/07/19/1078-0432.CCR-15-1169. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research.