Cisplatin-based chemotherapy is the first-line therapy for bladder cancer. However, cisplatin resistance has been associated with the recurrence of bladder cancer. Previous studies have shown that activation of FGFR and HER2 signaling are involved in bladder cancer cell proliferation and drug resistance. Smoking is the most common etiologic risk factor for bladder cancer, and there is emerging evidence that smoking is associated with cisplatin resistance. However, the underlying mechanism remains elusive. Acrolein, a highly reactive aldehyde, is abundant in tobacco smoke, cooking fumes, and automobile exhaust fumes. Our previous studies have shown that acrolein contributes to bladder carcinogenesis through the induction of DNA damage and inhibition of DNA repair. In this study, we found that acrolein induced cisplatin resistance and tumor progression in both non–muscle invasive bladder cancer (NMIBC) and muscle invasive bladder cancer (MIBC) cell lines RT4 and T24, respectively. Activation of HER2 and FGFR3 signaling contributes to acrolein-induced cisplatin resistance in RT4 and T24 cells, respectively. Furthermore, trastuzumab, an anti-HER2 antibody, and PD173074, an FGFR inhibitor, reversed cisplatin resistance in RT4 and T24 cells, respectively. Using a xenograft mouse model with acrolein-induced cisplatin-resistant T24 clones, we found that cisplatin combined with PD173074 significantly reduced tumor size compared with cisplatin alone. These results indicate that differential molecular alterations behind cisplatin resistance in NMIBC and MIBC significantly alter the effectiveness of targeted therapy combined with chemotherapy. This study provides valuable insights into therapeutic strategies for cisplatin-resistant bladder cancer.

Bladder cancer is the most common cancer of the urinary track and predominantly occurs in men worldwide (1). The majority of bladder cancers are urothelial cell carcinomas (UC), and more than 70% of patients are diagnosed with non–muscle invasive bladder cancer (NMIBC; ref. 2). Frequent recurrence in patients with NMIBC is a serious problem (3) that is observed in more than 50% of patients within 2 years, with 10% to 30% of recurrent patients progressing to muscle invasive bladder cancer (MIBC; ref. 4). Cisplatin-based chemotherapy is extensively used as a first-line treatment for many cancers, including bladder cancer (5); however, its efficacy is limited due to the development of drug resistance (6). Therefore, understanding the molecular mechanisms underlying cisplatin resistance is critical for improving bladder cancer treatment and prognosis.

The currently known mechanisms of bladder cancer cisplatin resistance are overactivation of growth factor signaling pathways, including FGFR and ErbB/HER, which results in diverse biological processes, including cell proliferation, survival, and tumorigenesis (7, 8). Members of the FGFR family are frequently activated by a mutation in bladder cancer and overexpressed in many bladder cancer cell lines, where they increase the activation of many survival pathways, such as MAPK/ERK (9). PD173074, a small molecular inhibitor, was reported to show both high affinity and selectivity for the FGFR family and was also shown to inhibit FGF-driven neoangiogenesis (10, 11). Previous studies have shown that PD173074 is extremely effective at potentiating the effect of cisplatin in lung cancer (12); however, similar work has not been carried out on bladder cancer thus far. Moreover, overexpression and/or amplification of the transmembrane tyrosine kinase (TK) receptor HER2, which is a member of the EGFR family, has been observed in bladder cancer (13), and HER2 overexpression is frequently detected in chemotherapy- and radiation-resistant bladder cancer (14). Anti-HER2 agents are able to induce cellular apoptosis in vitro and in vivo (15); however, multiple clinical trials of HER2-targeting agents generally in unselected patients with bladder cancer have not shown definitive clinical efficacy (16). Therefore, understanding the molecular alterations in bladder cancer has significant clinical implications in that they not only define radically different subgroups of patients with bladder cancer but also significantly affect tumor susceptibility to either conventional or target therapy and thus impact patient prognosis.

Smoking is the most common etiological risk factor for bladder cancer, and it accounts for up to 60% of all cases (17). Smoking intensity and duration have been shown to be closely associated with an increased risk of bladder cancer development (18). Furthermore, smoking cessation has also been suggested to be a cancer-specific benefit for bladder cancer prognosis (19, 20). Emerging evidence indicates that cigarette smoking is associated with the response to standard chemotherapeutic agents, including cisplatin (21, 22). Continued smoking during therapy is associated with low response rates to chemotherapy and radiation in many types of cancers, including bladder cancer (21, 23). However, little is known about the mechanism by which cigarette smoking enhances chemoresistance in bladder cancer. Acrolein, an α,β-unsaturated aldehyde, is abundant in cigarette smoking, cooking fumes, and automobile exhaust fumes (24). Acrolein can also be produced endogenously through lipid peroxidation, resulting from free radical damage to polyunsaturated fatty acids (25). Our previous studies showed that acrolein contributes to bladder carcinogenesis through the induction of DNA damage and inhibition of DNA repair (26). In addition, our current studies have shown that acrolein acts as an endogenous uremic toxin and contributes to urothelial carcinoma formation in patients with chronic kidney diseases (27). In this study, we investigated the molecular mechanisms by which acrolein induced cisplatin resistance in two well-established NMIBC and MIBC cell lines, RT4 and T24. Furthermore, the effectiveness of targeted therapies on cisplatin-resistant bladder cancer was analyzed in vitro and in vivo.

Chemicals

The following chemicals were purchased: acrolein from Sigma–Aldrich; cisplatin from Merck; trastuzumab from MedChemExpress; and PD173074 from MedChemExpress.

Cell culture and acrolein treatment

Human urinary bladder cancer cell lines RT4 (HTB-2) and T24 (HTB-4) were purchased from ATCC and maintained in McCoy 5A medium supplemented with 10% FBS and DMEM supplemented with 10% FBS, respectively. The absence of Mycoplasma in these cell lines was tested every month using an EZ-PCR Mycoplasma Detection Kit (Biological Industries). Acrolein stock solution was freshly prepared before use. Cells at 70% confluency were treated with acrolein (10 μmol/L) in complete culture medium for 1 month at 37°C in the dark, acrolein-containing medium was changed every 2 days, and selected clones were named Acr-clone#. For the dose or time effects, cells were treated with different concentrations of acrolein (0–10 μmol/L) for 24 hours or acrolein (7.5 μmol/L) for 3 to 24 hours, respectively.

Cell viability assay

Cell viability was determined using a modified 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma–Aldrich) assay and a lactate dehydrogenase (LDH) leakage assay, as previously described (28). Briefly, cells (5,000/well) were seeded in 96-well plates overnight and treated with cisplatin (0–30 μg/mL), trastuzumab (1 μg/mL), or PD173074 (5 μmol/L) for 24 hours. For the MTT assay, the resulting formazan dissolved in DMSO was measured at 570 nm, and the results are presented as the percentage of the control values. For the LDH leakage assay, LDH activities were measured using an LDH detection kit (Sigma–Aldrich) as described in the manufacturer's protocol. The extent of cellular damage was determined to be dependent on the percent LDH activity in the supernatants with respect to that in the cell lysates. These experiments were performed at least three times in triplicate.

Cell proliferation assay

Cell viability was determined using a modified MTT assay as described in the cell viability assay. Briefly, cells (1,000/well) were seeded in 96-well plates overnight and measured every day for 7 days. These experiments were performed at least three times in triplicate.

Soft agar colony formation assay

To evaluate anchorage-independent cell growth, a soft agar colony formation assay was performed as described previously (29). Briefly, bottom layers were formed with a 3-mL aliquot of 1.2% agar in culture medium in six-well plates. Then, the top layer containing 1,000 cells/well of parental cells or Acr-clones was mixed with 3 mL of 0.35% agar in medium and plated on solidified bottom agar. When the top agar solidified, the dishes were transferred to an incubator and cultured for 14 days. Two or three drops of the medium were added to each dish three times a week. Colonies were stained with 0.005% crystal violet, photographed, and counted. All of these experiments were performed in triplicate and repeated independently at least three times.

Cell migration assay

The cell migration assay was carried out in a Transwell apparatus with 8-μm pore size membranes (Corning) as previously described (30). Briefly, parental or Acr-clones (5 × 104/well, six-well plates) were added to the upper chamber, and the lower chamber contained growth medium supplemented with 10% FBS. After 24 hours of incubation at 37°C in a 5% CO2 incubator, cells on the upper filter surface were removed by wiping with a cotton swab. Filters were then fixed in methanol and stained with crystal violet. The number of cells was counted in six random fields under a microscope at 200× magnification. These experiments were performed at least three times in triplicate.

qRT-PCR

Total RNA were prepared and subsequent real-time RT-PCR analysis of cDNA was analyzed as described previously (31). The primers (5′-3′) were AAGCTTACCATGGAGCTGGCGGCCTTG and CTCGAGCACTGGCACGTCCAGACC for HER2; CCACTGTCTGGGTCAGGAT and AGGATGGAGCGTCTGTCAC for FGFR3; CCGTCTAGAAAAACCTGCC and GCCAAATTCGTTGTCATACC for GAPDH. To calculate the relative RNA expression, GAPDH was used as an internal control for all qRT-PCR reactions and compared with control groups. These experiments were performed at least three times in duplicate.

Western blot analysis

Cell lysates were prepared and analyzed as described previously (31). Briefly, blots were blocked with 5% nonfat milk and hybridized with primary antibodies overnight at 4°C. The antibodies against phospho-HER2/ErbB2 (Tyr1221/1222; 1:1000, Cell Signaling Technology #2249), HER2 (1:1000, Cell Signaling Technology #9212), phospho-FGFR3 (Tyr724; 1:1000, Abcam #ab155960), FGFR3 (1:1000, Novus Biologicals #JM81–10), phospho-p38 (Thr180/Tyr182; 1:1000, Cell Signaling Technology #9211), p38 (1:1000, Cell Signaling Technology #9212), phospho-p44/42 MAPK (Erk1/2; Thr202/Tyr204; 1:1000, Cell Signaling Technology #9101), p44/42 MAPK (Erk1/2; 1:1000, Cell Signaling Technology #9102), PARP-1 (1:1000, Cell Signaling Technology #9542), caspase-9 (1:1000, Cell Signaling Technology #9502), caspase-3 (1:1000, Cell Signaling Technology #9662), E-cadherin (1:1000, Cell Signaling Technology #3195), EpCAM (gifted from Prof. Ying Chih Chang from Genomic Research Center at Academia Sinica, Taipei, Taiwan), vimentin (1:1000, Taiclone # TCEA19744), Snail (1:1000, Cell Signaling Technology #C15D3), and GAPDH (1:5000, Cell Signaling Technology #5174). The immunodetection was performed using Enhanced Chemiluminescence (ECL; Millipore Corporation).

Xenograft mouse model

Forty 6-week-old male Balb/c nude mice weighing 20 to 25 g were used. All animal experiments were approved by the Institutional Animal Care and Use Committee (IACUC) of National Yang Ming Chiao Tung University (Taipei, Taiwan) and carried out according to the Guidelines for Animal Research of National Yang Ming Chiao Tung University (IACUC #1070208rr). T24 parental cells or T24 Acr-clone#1 cells (5 × 106 in 50% Matrigel, BD Bioscience) were subcutaneously inoculated into the right flank of mice. After 1 week, mice with palpable xenografts were randomly divided into four groups: vehicle (PBS, 50 μL, i.p., once weekly; and corn oil, 50 μL, i.p. once weekly), cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly), cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly) combined with PD173074 (10 mg/kg in corn oil, 50 μL, i.p. once weekly) and cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly) combined with PD173074 (10 mg/kg in corn oil, 50 μL, i.p. once weekly). To obtain the tumor growth curve, the tumor dimensions were monitored every 3 days. The perpendicular diameter was measured with a digital caliper, and the volume was calculated based on the (length × width2)/2. Body weight was also assessed twice weekly. The animals were sacrificed after 3 weeks of the drug treatment. Tumor samples were collected after sacrifice. Each sample was cut in half, with one half preserved in 4% paraformaldehyde and the other flash frozen in liquid nitrogen and stored at −80°C until further use.

Histological, terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling, and IHC analyses

For histologic staining, sections (4 µm) were deparaffinized in xylene, rehydrated in a descending ethanol series, and stained with hematoxylin and eosin (H&E). Cell death by apoptosis was analyzed using an in situ cell death detection kit (POD, Roche), as described in the manufacturer's protocol. Briefly, paraffin-embedded tumor sections were dewaxed by heating at 60°C, followed by washing in xylene and rehydration. Endogenous peroxidase activity was eliminated by incubation in hydrogen peroxide. Then, tumor sections were incubated with proteinase K solution (20 μg/mL in 10 mmol/L Tris-HCl, pH 7.4) for 20 minutes at room temperature. Following washing with PBS, tumor sections were permeabilized in 0.1% Triton X-100 and 0.1% sodium citrate for 8 minutes at room temperature and incubated in 50 µL of terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling (TUNEL) reaction mixture for 1 hour at 37°C in the dark. Tumor sections were washed with PBS and incubated with 50 µL Converter-POD for 30 minutes at 37°C. After rinsing in PBS, the sections were incubated for 10 minutes with 100 µL diaminobenzidine (DAB; Sigma–Aldrich) substrate in the dark. Finally, the samples were mounted and analyzed under a light microscope at 400× magnification, where apoptotic cells could be seen as condensed shrinking dark brown cells. For IHC analysis, sections were incubated in Tris–EDTA, pH 6.0, and boiled for 12 minutes to enhance immunoreactivity. Endogenous peroxidase activity was eliminated by incubation in hydrogen peroxide. Incubation with primary antibodies for FGFR3 antibody (1:300, Novus Biologicals #JM81–10) was performed overnight at 4°C in 1% BSA in PBS. Bound antibodies were visualized with DAB used as a chromogen, and omission of the primary antibody served as a negative control. Assessment of FGFR3 immunoexpression was performed by light microscopy at ×400 magnification.

Statistical analyses

Descriptive statistics are presented as the mean ± SD or as the number (percentage). All calculated P values were two-tailed. Statistical significance was defined as *P < 0.05. A minimum of three independent replicate experiments was performed to justify the use of statistical tests.

Acrolein induced cisplatin resistance in both the NMIBC cell line RT4 and MIBC cell line T24

To investigate whether cigarette smoke acrolein contributes to cisplatin resistance in human bladder cancer, we treated the NMIBC and MIBC cell lines, RT4 and T24, respectively, with a physiologically relevant dose of acrolein (10 μmol/L) for 1 month and selected three different clones, namely, RT4 Acr-clone# and T24 Acr-clone#. The results showed that RT4 Acr-clone#3 (IC50 = 25.13 ± 3.01 μg/mL) had a higher IC50 than the parental RT4 (IC50 = 12.04 ± 1.86 μg/mL; Fig. 1A and B), and T24 Acr-clone#1 (IC50 = 18.57 ± 3.95 μg/mL) had a higher IC50 than the parental T24 (IC50 = 9.46 ± 0.64 μg/mL; Fig. 1C and D) using MTT and LDH analysis. The results suggest that acrolein increased the IC50 of cisplatin by two-fold in both RT4 and T24 cells.

Figure 1.

Acrolein induces cisplatin resistance in both low-grade and high-grade bladder cancer cell lines RT4 and T24, respectively. RT4 and T24 cells were treated with acrolein (10 μmol/L) for 1 month and named RT3 Acr-clone# and T24 Acr-clone#. The cytotoxicity of cisplatin (0–30 μg/mL, 24 hours) in the absence of acrolein in RT4 Acr-clone#3 (A, B) and T24 Acr-clone#1 (C, D) cells was analyzed using MTT assays (A, C) and LDH assays (B, D) as described in the Materials and Methods. The dotted line indicates the IC50. Values are presented as the mean ± SD. Student t tests were used to determine statistical significance, and two-tailed P values are shown. **, P < 0.01; ***, P < 0.005 compared with parental cells.

Figure 1.

Acrolein induces cisplatin resistance in both low-grade and high-grade bladder cancer cell lines RT4 and T24, respectively. RT4 and T24 cells were treated with acrolein (10 μmol/L) for 1 month and named RT3 Acr-clone# and T24 Acr-clone#. The cytotoxicity of cisplatin (0–30 μg/mL, 24 hours) in the absence of acrolein in RT4 Acr-clone#3 (A, B) and T24 Acr-clone#1 (C, D) cells was analyzed using MTT assays (A, C) and LDH assays (B, D) as described in the Materials and Methods. The dotted line indicates the IC50. Values are presented as the mean ± SD. Student t tests were used to determine statistical significance, and two-tailed P values are shown. **, P < 0.01; ***, P < 0.005 compared with parental cells.

Close modal

We further analyzed cell proliferation, soft agar colony formation activity, and cell migration ability in acrolein-induced cisplatin-resistant RT4 and T24 clones. The results showed that increased cell proliferation (Fig. 2A and B), soft agar colony formation (Fig. 2C and D), and cell migration ability (Fig. 2E and F) were increased in RT4 Acr-clone#3 and T24 Acr-clone#1 cells compared with parental cells. We also analyzed epithelial–mesenchymal transition (EMT) markers in both cell clones. The results showed decreased E-cadherin but increased EpCAM in RT4 Acr-clone#3 cells compared with parental cells and increased vimentin and Snail in T24 Acr-clone#1 cells compared with the parental cells (Supplementary Fig. S1). These results suggest that acrolein induced cancer progression, including cisplatin resistance, in both high-grade and low-grade bladder cancer cell lines.

Figure 2.

Acrolein induces tumor progression in acrolein-induced cisplatin-resistant RT4 and T24 clones. Cell proliferation of RT4 Acr-clone#3 (A) and T24 Acr-clone#1 (B) cells was analyzed using MTT assays. Soft agar anchorage-dependent cell growth of RT4 Acr-clone#3 (C) and T24 Acr-clone#1 (D) was analyzed using a soft agar assay. The cell migration activity of RT4 Acr-clone#3 (E) and T24 Acr-clone#1 (F) was analyzed based on a Transwell migration analysis. Values were presented as the mean ± SD. Student t tests were used to determine statistical significance, and two-tailed P values are shown. *, P < 0.05; **, P < 0.01; ***, P < 0.005 compared with parental cells.

Figure 2.

Acrolein induces tumor progression in acrolein-induced cisplatin-resistant RT4 and T24 clones. Cell proliferation of RT4 Acr-clone#3 (A) and T24 Acr-clone#1 (B) cells was analyzed using MTT assays. Soft agar anchorage-dependent cell growth of RT4 Acr-clone#3 (C) and T24 Acr-clone#1 (D) was analyzed using a soft agar assay. The cell migration activity of RT4 Acr-clone#3 (E) and T24 Acr-clone#1 (F) was analyzed based on a Transwell migration analysis. Values were presented as the mean ± SD. Student t tests were used to determine statistical significance, and two-tailed P values are shown. *, P < 0.05; **, P < 0.01; ***, P < 0.005 compared with parental cells.

Close modal

Acrolein induced the expression of HER2 and downstream ERK and p38 signaling in RT4 cells, while acrolein induced the expression of FGFR3 and downstream ERK and p38 signaling in T24 cells

The currently known mechanisms of bladder cancer cisplatin resistance are overactivation of growth factor signaling pathways, including FGFR and ErbB/HER, which results in downstream MAPK/ERK activation and diverse biological processes, including cell proliferation differentiation, survival, and tumorigenesis (7, 8). We further analyzed the FGFR3 pathway and HER2 pathway in both acrolein-induced cisplatin-resistant RT4 and T24 clones. The results showed increased mRNA and protein expression of HER2 in RT4 Acr-clone#3 cells compared with the parental RT4 cells (Fig. 3A and C). Moreover, increased mRNA and protein expression of FGFR3 in T24 Acr-clone#1 cells were observed compared with that in the parental T24 cells (Fig. 3B and C). However, the phosphorylation of HER2 or FGFR3 was not increased in these cells (Supplementary Fig. S2). HER2- or FGFR3-downstream MAPK pathways, including the p38 and ERK pathways, were activated in RT4 Acr-clone#3 and T24 Acr-clone#1 cells compared with their parental cells (Fig. 3A and B). Furthermore, short-term treatment with acrolein induced protein expression of HER2 in both RT4 and T24 cells, while it increased protein expression of FGFR3 in T24 cells (Fig. 3D and E). These results suggest that acrolein induced cisplatin resistance in RT4 and T24 cells through HER2- or FGFR3-downstream MAPK pathways.

Figure 3.

Analysis of HER2 and FGFR3 signaling pathways in acrolein-induced cisplatin-resistant RT4 and T24 clones. Western blot analysis of HER2 and FGFR3 downstream signaling pathways was performed in RT4 Acr-clone#3 (A) and T24 Acr-clone#1 (B) compared with parental cells. mRNA expression of HER2 and FGFR3 was analyzed in RT4 Acr-clone#3 (upper panel of C) and T24 Acr-clone#1 (lower panel of C) compared with parental cells using qRT-PCR assay. Values were presented as the mean ± SD. Student's t tests were used to determine statistical significance, and two-tailed P values are shown. **, P < 0.01 compared with parental cells. Dose and time effects of acrolein on HER2 and FGFR3 expression, p38 activation, and ERK activation in RT4 Acr-clone#3 (D) and T24 Acr-clone#1 (E) cells were compared with that of parental cells using Western blot analysis. For the dose and time effects, the cells were treated with different concentrations of acrolein (0–10 μmol/L) for 24 hours or acrolein (7.5 μmol/L) for 3 to 24 hours, respectively. h, hour.

Figure 3.

Analysis of HER2 and FGFR3 signaling pathways in acrolein-induced cisplatin-resistant RT4 and T24 clones. Western blot analysis of HER2 and FGFR3 downstream signaling pathways was performed in RT4 Acr-clone#3 (A) and T24 Acr-clone#1 (B) compared with parental cells. mRNA expression of HER2 and FGFR3 was analyzed in RT4 Acr-clone#3 (upper panel of C) and T24 Acr-clone#1 (lower panel of C) compared with parental cells using qRT-PCR assay. Values were presented as the mean ± SD. Student's t tests were used to determine statistical significance, and two-tailed P values are shown. **, P < 0.01 compared with parental cells. Dose and time effects of acrolein on HER2 and FGFR3 expression, p38 activation, and ERK activation in RT4 Acr-clone#3 (D) and T24 Acr-clone#1 (E) cells were compared with that of parental cells using Western blot analysis. For the dose and time effects, the cells were treated with different concentrations of acrolein (0–10 μmol/L) for 24 hours or acrolein (7.5 μmol/L) for 3 to 24 hours, respectively. h, hour.

Close modal

Trastuzumab, an anti-HER2 antibody, reversed cisplatin resistance in acrolein-treated RT4 clones, while PD173074, an FGFR inhibitor, reversed cisplatin resistance in acrolein-treated T24 clones

To verify the activation of HER2 and FGFR3 signaling in RT4 Acr-clone#3 and T24 Acr-clone#1 cells, trastuzumab, an anti-HER2 mAb, and PD173074, an FGFR3 inhibitor, were utilized. The results showed that trastuzumab reversed cisplatin resistance in RT4 Acr-clone#3 cells (Fig. 4A). The combination of trastuzumab with cisplatin enhanced cellular apoptosis (Fig. 4B) and decreased the expression of HER2 in RT4 Acr-clone#3 cells compared with cisplatin alone (Fig. 4C). This phenomenon was not observed in the T24 Acr-clone#1 cells treated with the combination of trastuzumab and cisplatin (Fig. 4D–F). Moreover, PD173074 reversed cisplatin resistance in T24 Acr-clone#1 cells (Fig. 4G). The combination of PD173074 with cisplatin enhanced cellular apoptosis in T24 Acr-clone#1 cells (Fig. 4H) and decreased FGFR3 compared with vehicle control (Fig. 4I). However, this phenomenon was also observed in the combination treatment of PD173074 with cisplatin in RT4 Acr-clone#3 cells (Figs. 4JL). Since RT4 cells contain FGFR3 fusions, they have been shown to be more sensitive to FGFR inhibition (32, 33). Consistently, we found that PD173074 alone induced higher cytotoxicity and cellular apoptosis in both parental RT4 cells and Acr-clone#3 cells (Fig. 4J and K). These results further confirm that acrolein induced activation of HER2 signaling and FGFR3 signaling, resulting in cisplatin resistance in RT4 and T24 cells, respectively.

Figure 4.

Effect of trastuzumab or PD173074 combined with cisplatin in acrolein-induced cisplatin-resistant RT4 and T24 clones. RT4 Acr-clone#3 was treated with cisplatin (2.5 μg/mL), or cisplatin (2.5 μg/mL) with trastuzumab (1 μg/mL), or trastuzumab (1 μg/mL), or cisplatin (2.5 μg/mL) with PD173074 (5 μmol/L), or PD173074 (5 μmol/L) for 24 hours. T24 Acr-clone#1 was treated with cisplatin (1 μg/mL, 24 hours), or cisplatin (1 μg/mL) with PD173074 (5 μmol/L), or PD173074 (5 μmol/L), or cisplatin (1 μg/mL) with trastuzumab (1 μg/mL), or trastuzumab (1 μg/mL) for 24 hours. Cytotoxicity of trastuzumab combined with cisplatin in RT4 Acr-clone#3 (A) and in T24 Acr-clone#1 (D) and cytotoxicity of PD173074 combined with cisplatin in T24 Acr-clone#1 (G) and RT4 Acr-clone#3 (J) compared with that of the parental cells were analyzed using MTT assays. Values are presented as the mean ± SD. Student t tests were used to determine statistical significance, and two-tailed P values are shown. *, P < 0.05, compared with vehicle treatment, ##, P < 0.01; ###, P < 0.005 compared with cisplatin treatment group. Western blot analysis of apoptotic proteins, including cleavage of PARP, caspase-9, and caspase-3, in RT4 Acr-clone#3 (B, K) and T24 Acr-clone#1 (E, H) cells compared with that of the parental cells was performed. Western blot analysis of HER2 and FGFR3, p38 activation, and ERK activation in RT4 Acr-clone#3 (C, L) and T24 Acr-clone#1 (F, I) cells compared with that of the parental cells was performed. c-Cas.: cleavage form of caspase; pro-Cas.: proform of caspase.

Figure 4.

Effect of trastuzumab or PD173074 combined with cisplatin in acrolein-induced cisplatin-resistant RT4 and T24 clones. RT4 Acr-clone#3 was treated with cisplatin (2.5 μg/mL), or cisplatin (2.5 μg/mL) with trastuzumab (1 μg/mL), or trastuzumab (1 μg/mL), or cisplatin (2.5 μg/mL) with PD173074 (5 μmol/L), or PD173074 (5 μmol/L) for 24 hours. T24 Acr-clone#1 was treated with cisplatin (1 μg/mL, 24 hours), or cisplatin (1 μg/mL) with PD173074 (5 μmol/L), or PD173074 (5 μmol/L), or cisplatin (1 μg/mL) with trastuzumab (1 μg/mL), or trastuzumab (1 μg/mL) for 24 hours. Cytotoxicity of trastuzumab combined with cisplatin in RT4 Acr-clone#3 (A) and in T24 Acr-clone#1 (D) and cytotoxicity of PD173074 combined with cisplatin in T24 Acr-clone#1 (G) and RT4 Acr-clone#3 (J) compared with that of the parental cells were analyzed using MTT assays. Values are presented as the mean ± SD. Student t tests were used to determine statistical significance, and two-tailed P values are shown. *, P < 0.05, compared with vehicle treatment, ##, P < 0.01; ###, P < 0.005 compared with cisplatin treatment group. Western blot analysis of apoptotic proteins, including cleavage of PARP, caspase-9, and caspase-3, in RT4 Acr-clone#3 (B, K) and T24 Acr-clone#1 (E, H) cells compared with that of the parental cells was performed. Western blot analysis of HER2 and FGFR3, p38 activation, and ERK activation in RT4 Acr-clone#3 (C, L) and T24 Acr-clone#1 (F, I) cells compared with that of the parental cells was performed. c-Cas.: cleavage form of caspase; pro-Cas.: proform of caspase.

Close modal

Cisplatin combined with PD173074 inhibited tumor growth in acrolein-induced cisplatin-resistant T24 clones using a xenograft mouse model

We further investigated the potential of PD173074 combined with cisplatin in cisplatin-resistant T24 cells using a xenograft mouse model. T24 parental cells or T24 Acr-clone#1 cells in Matrigel were subcutaneously injected into the flanks of nude mice, and once the tumors were measurable, animals were randomized to receive carrier buffer in the absence or presence of PD172074 (10 mg/kg i.p.) once a week with or without cisplatin (5 mg/kg i.p.) once a week. After 3 weeks, the mice were sacrificed and tumors were collected for further analysis. The results showed that tumor volumes in the cisplatin group did not show a significant difference from those of the vehicle group, while PD173074 as a single agent was as efficient at decreasing the tumor size compared to vehicle treatment (Fig. 5A and B). Furthermore, PD173074 combined with cisplatin led to significantly better impairment of tumor growth than either agent alone (P < 0.01; Fig. 5A and B). However, this phenomenon was not observed in mice inoculated with parental T24 cells (Supplementary Fig. S3). Slight weight loss occurred in mice treated with PD173074 combined with cisplatin (Fig. 5C). In addition, the morphologic changes in the kidney and liver tissues were assessed using H&E staining. The results showed that kidneys from mice in either the vehicle control or PD173074 alone group displayed normal renal architecture. As expected, cisplatin treatment alone resulted in obvious renal tubular damage characterized by tubular degeneration and dilatation, extensive epithelial vacuolization and necrosis, and the formation of hyaline casts (Supplementary Fig. S4). However, cisplatin combined with PD173074 significantly attenuated the pathologic changes in cisplatin-treated mice. These results suggest that PD173074 may have a renoprotective effect in cisplatin-treated mice. In addition, there were no obvious morphologic changes in the liver among the four groups (Supplementary Fig. S4). We further analyzed apoptosis in the tumor samples using the TUNEL assay, and the results showed that PD173074 combined with cisplatin significantly increased the number of TUNEL-positive apoptotic cells in xenografts compared with either agent alone (Fig. 5D and E). In addition, we examined the expression of FGFR3 using IHC staining assay and found that PD173074 combined with cisplatin significantly decreased the FGFR3 expression in xenografts compared with vehicle control or cisplatin alone (Fig. 5D and F). This finding is consistent with the in vitro results (Fig. 4G–I), which suggests that the FGFR3 pathway contributes to cisplatin resistance in T24 Acr-clone#1 cells and combining PD173074 with cisplatin induced more cellular apoptosis in T24 Acr-clone#1 cells.

Figure 5.

Effect of PD173074 combined with cisplatin in acrolein-induced cisplatin-resistant T24 clones using an xenograft mouse model. The animal experimental protocol is described in the Materials and Methods. A, Overall view of tumors formed by T24 Acr-clone#1 cells treated with the vehicle (PBS, 50 μL, i.p. once weekly; and corn oil, 50 μL, i.p. once weekly), cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly), cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly) combined with PD173074 (10 mg/kg in corn oil, 50 μL, i.p. once weekly), or PD173074 (10 mg/kg in corn oil, 50 μL, i.p. once weekly). Tumor growth curves (B) and body weight (C) of nude mice in different experimental groups (n  =  5). D, Representative H&E (top), TUNEL staining (middle), and IHC staining of FGFR3 (bottom) in tumor sections from the vehicle, cisplatin, and/or PD173074 treatment groups. E, Quantification of the TUNEL-positive area shown in the middle panel of (D). The apoptotic tumor cells in the middle panel are stained brown. F, Quantification of the FGFR3-positive area shown in the lower panel of (D). Student t tests were used to determine statistical significance, and two-tailed P values are shown. **, P < 0.01; ***, P < 0.005 compared with vehicle group; #, P < 0.05; ##, P < 0.01 compared with cisplatin alone; &&, P < 0.01; &&&, P < 0.005 compared with cisplatin + PD173074.

Figure 5.

Effect of PD173074 combined with cisplatin in acrolein-induced cisplatin-resistant T24 clones using an xenograft mouse model. The animal experimental protocol is described in the Materials and Methods. A, Overall view of tumors formed by T24 Acr-clone#1 cells treated with the vehicle (PBS, 50 μL, i.p. once weekly; and corn oil, 50 μL, i.p. once weekly), cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly), cisplatin (5 mg/kg in PBS, 50 μL, i.p. once weekly) combined with PD173074 (10 mg/kg in corn oil, 50 μL, i.p. once weekly), or PD173074 (10 mg/kg in corn oil, 50 μL, i.p. once weekly). Tumor growth curves (B) and body weight (C) of nude mice in different experimental groups (n  =  5). D, Representative H&E (top), TUNEL staining (middle), and IHC staining of FGFR3 (bottom) in tumor sections from the vehicle, cisplatin, and/or PD173074 treatment groups. E, Quantification of the TUNEL-positive area shown in the middle panel of (D). The apoptotic tumor cells in the middle panel are stained brown. F, Quantification of the FGFR3-positive area shown in the lower panel of (D). Student t tests were used to determine statistical significance, and two-tailed P values are shown. **, P < 0.01; ***, P < 0.005 compared with vehicle group; #, P < 0.05; ##, P < 0.01 compared with cisplatin alone; &&, P < 0.01; &&&, P < 0.005 compared with cisplatin + PD173074.

Close modal

Cigarette smoking is among the most important risk factors for bladder cancer (17), and continued smoking during therapy is associated with lower response rates to chemotherapy or radiation in patients with bladder cancer (21). Cisplatin-based chemotherapy is the most effective therapeutic regimen for bladder cancer (5); however, the development of chemoresistance in patients with bladder cancer is a critical problem in the management of advanced bladder cancer (6). Our previous studies showed that cigarette smoke–containing acrolein contributes to bladder carcinogenesis through the induction of DNA damage and inhibition of DNA repair (26). In this study, our results showed that long-term exposure to acrolein induced cisplatin resistance and tumor progression in both the NMIBC and MIBC cell lines, RT4 and T24, respectively (Figs. 12). The physiologically relevant dosage of acrolein is approximately 10 μmol/L based on previous studies showing that the acrolein exposure level from an average smoker is approximately 1.7 to 2 mg/day (34). Assuming a 60-kg adult with 4 L of blood volume, the exposure concentration of acrolein in smokers is approximately 7.6 to 9 μmol/L. Differential molecular alterations, including HER2 and FGFR3, were observed in acrolein-induced cisplatin-resistant RT4 and T24 cells (Fig. 3). Furthermore, HER2- and FGFR3-targeted therapy reversed cisplatin resistance in RT4 and T24 cells, respectively (Figs. 4 and 5). Cisplatin-resistant bladder cancer models are typically established by treatment with increasing concentrations of cisplatin until they become tolerant to relatively high concentrations of cisplatin (generally approximately 12 µmol/L; ref. 7). Multiple mechanisms of cisplatin resistance have been reported, including the reduction of cellular cisplatin accumulation, changes in DNA repair mechanisms, and alterations in apoptotic cell death pathways, and growth factor signaling (35). This model was used to study the molecular mechanism of cisplatin resistance induced by long-term cisplatin treatment in bladder cancer; however, our study aimed to understand the molecular mechanism underlying cigarette smoking–induced chemoresistance in bladder cancer.

In previous studies, T24 cells were derived from a patient with undifferentiated grade 3 carcinoma, they were found to be representative cells of a cancer with a poor prognosis (36). In comparison, RT4 cells were derived from a patient with a well-differentiated grade 1 papillary tumor, and they were found to be representative cells of a cancer with good prognosis (36). In addition, RT4 cells showed strong expression of epithelial markers (E-cadherin and EpCAM), while T24 cells exhibited strong expression of mesenchymal markers (vimentin and Snail; Supplementary Fig. S1), which is consistent with previous studies (37). Therefore, these two cell lines could represent two discrete “epithelial” and “mesenchymal” subsets, with the latter consisting entirely of muscle-invasive tumors (38). Consequently, we would be able to differentiate distinct molecular mechanisms of cisplatin resistance induced by cigarette smoke–containing acrolein using these two cell models. Previous studies have shown that EMT coordinately regulates drug resistance and muscle invasion/metastasis in cancers (39). EMT is characterized by a loss of cell-to-cell adhesion and cell polarity and increases in metastasis and cell progression of bladder cancer (40). The most common change that occurs during EMT is downregulation of surface E-cadherin expression, increased expression of several transcriptional repressors of E-cadherin expression (Zeb-1, Zeb-2, Twist, Snail, and Slug) and increased expression of vimentin, fibronectin, and S100 proteins (41). Our results showed that E-cadherin is lost in RT4 Acr-clone#3 cells and vimentin and Snail are increased in T24 Acr-clone#1 cells compared with that in the parental cells (Supplementary Fig. S1). Consistently, we also found that cell migration activity was increased in both cisplatin-resistant clones (Fig. 2E and F). Interestingly, we found increased expression of EpCAM in RT4 Acr-clone#3 cells compared with that in its parental cells (Supplementary Fig. S1). EpCAM is an epithelial cell adhesion molecule (42), although it may also function to enhance the mobility of cells and tissues (43). Previous results have shown that EpCAM is overexpressed in many epithelial malignancies, including bladder carcinoma in situ and high-grade and advanced stage bladder cancers (44). Therefore, it is possible that EpCAM also contributes to increased cell proliferation and soft agar colony formation in RT4 Acr-clone#3 cells (Fig. 2A and C).

Overactivation of growth factor signaling pathways, including FGFR and ErbB/HER, result in diverse biological processes, including cell proliferation, survival, and tumorigenesis, and these pathways are known mechanisms of bladder cancer cisplatin resistance (7, 8). In this study, we found that acrolein induced distinct molecular alterations resulting in cisplatin resistance in two bladder cancer cell lines, RT4 and T24. Higher mRNA and protein expression levels of HER2 were observed in RT4 Acr-clone#3, while higher RNA and protein expression levels of FGFR3 were observed in T24 Acr-clone#1 (Fig. 3AC). Previous studies have shown that RT4 belongs to low-grade papillary bladder tumors containing FGFR3 fusion and is more sensitive to FGFR inhibition (32, 33). Therefore, it is postulated that RT4 becomes addicted to FGFR signaling during the process of tumor formation. Consistently, we found that PD173074 alone induced higher cytotoxicity and cellular apoptosis in both parental RT4 cells and Acr-clone#3 cells (Fig. 4J and K). In this study, we found increased HER2 expression but not FGFR3 expression in RT4 Acr-clone#3 cells compared with that in the parental cells (Fig. 3A and C). In addition, short-term treatment with acrolein induced HER2 expression and downstream ERK and p38 activation in RT4 cells (Fig. 3D), and trastuzumab reversed cisplatin resistance in RT4 cells (Fig. 4A). Moreover, acrolein upregulated FGFR3 in T24 cells, which are high-grade muscle invasive bladder tumors containing wild-type FGFR3 (Fig. 3B, C, and E). In addition, PD173074 reversed cisplatin resistance in T24 cells (Fig. 4G). These results indicate that acrolein induced distinct molecular alterations that underlie cisplatin resistance in RT4 and T24 cells with different genetic backgrounds. Our previous studies have shown that acrolein induces DNA damage and inhibits DNA repair activity, thus leading to bladder carcinogenesis (26). However, the molecular mechanisms by which acrolein induces upregulation of HER2 or FGFR3 expression still need further investigation. In addition, only a single cell line was utilized to represent each subset of NMIBC and MIBC in this study; hence, further studies are required to confirm the findings.

Previous studies have shown that HER2 overexpression is frequently detected in chemotherapy- and radiation-resistant bladder cancer (14, 45). Therefore, anti-HER2 agents are able to induce cellular apoptosis in vitro and in vivo (15). In this study, we found that the anti-HER2 antibody trastuzumab combined with cisplatin could induce more cellular apoptosis and reverse cisplatin resistance in RT4 Acr-clone#3 cells (Fig. 4A and B) and inhibit HER2 downstream signaling, including the activation of ERK and p38 (Fig. 4C). These results indicate that upregulation of HER2 contributes to cisplatin resistance in RT4 cells induced by acrolein. Since HER2 alterations are common in urothelial bladder cancer (13), HER2 has been targeted by numerous agents in multiple prospective clinical trials (16). Trastuzumab was suggested to have clinical efficacy in advanced urothelial cancer (46, 47). Furthermore, several clinical trials also showed that trastuzumab could be feasibly combined with chemotherapy for advanced urothelial cancer overexpressing HER2 (48, 49). A two-phase clinical trial showed that patients in the trastuzumab arm who received cisplatin-based chemotherapy had superior outcomes, although the higher than expected rate of cardiac toxicity was concerning (50). Therefore, our results may provide insights into the molecular mechanisms of cisplatin resistance in advanced bladder cancer.

Activating mutations of FGFR3 have been described in approximately 75% of low-grade papillary bladder tumors and 20% of MIBC (51). Furthermore, overexpression of FGFR3 is observed in approximately half of MIBC (52). Previous studies have shown that the FGFR3S249C mutation promotes chemoresistance in bladder cancer cells (53). Therefore, FGFR3 is a particularly promising target for therapy in bladder cancer. Previous studies have shown that FGFR inhibition (with the small molecule inhibitor PD173074) is extremely effective at potentiating the effect of cisplatin in lung cancer (12); however, similar work has not been carried out in bladder cancer thus far. In this study, we found that PD173074 combined with cisplatin could induce more cellular apoptosis, reverse cisplatin resistance in T24 Acr-clone#1 cells (Fig. 4G and H), and downregulate FGFR3 expression (Fig. 4I). Furthermore, we found that PD173074 induced intratumoral apoptosis and potentiated the effect of cisplatin in bladder cancer using a xenograft mouse model with acrolein-induced T24 Acr-clone#1 (Fig. 5). These results together suggest that upregulation of FGFR3 contributes to cisplatin resistance in T24 cells induced by acrolein and that PD173074 could reverse cisplatin resistance in vitro and in vivo.

In conclusion, our results show that long-term exposure to cigarette smoke–containing acrolein induced cisplatin resistance in NMIBC and MIBC cell lines, RT4 and T24, respectively. Distinct molecular mechanisms, including upregulation of HER2 or FGFR3 expression and downstream signaling pathways, were induced in these two bladder cancer cell lines. Our study indicates that different molecular alterations behind cisplatin resistance in two subsets of bladder cancer cell lines significantly alter the effectiveness of targeted therapy combined with chemotherapy.

No disclosures were reported.

J.-H. Hong: Funding acquisition, writing–original draft. Z.-J. Tong: Data curation, investigation, methodology. T.-E. Wei: Data curation, investigation, methodology. Y.-C. Lu: Data curation, methodology. C.-Y. Huang: Data curation, methodology. C.-Y. Huang: Resources, supervision, funding acquisition. C.-H. Chiang: Data curation, methodology. F.-S. Jaw: Data curation, methodology. H.-W. Cheng: Data curation, investigation, methodology. H.-T. Wang: Data curation, validation, methodology, writing–original draft, project administration, writing–review and editing.

We thank Prof. Y.C. Chang from Genomic Research Center at Academia Sinica for providing EpCAM antibody. This work was supported by National Health Research Institutes, Taiwan (under NHRI-EX110-11027PI and NHRI-EX111-11027PI to H.-T. Wang), Ministry of Science and Technology, Taiwan (under grant no. 109-2320-B-010-024, to H.-T. Wang), Yen Tjing Ling Medical Foundation (CI-110-11, to H.-T. Wang), Veterans General Hospitals and University System of Taiwan Joint Research Program (VGHUST111-G1-4-2, to H.-T. Wang), National Yang Ming Chiao Tung University Far Eastern Memorial Hospital Joint Research Program (NYCU-FEMH 110DN01 and 111DN01 to H.-T. Wang), and National Taiwan University Hospital (grant no. 110-N4915, to J.H. Hong), and by grants from Taiwan Health Foundation.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Cumberbatch
MGK
,
Jubber
I
,
Black
PC
,
Esperto
F
,
Figueroa
JD
,
Kamat
AM
, et al
.
Epidemiology of bladder cancer: a systematic review and contemporary update of risk factors in 2018
.
Eur Urol
2018
;
74
:
784
95
.
2.
Humphrey
PA
,
Moch
H
,
Cubilla
AL
,
Ulbright
TM
,
Reuter
VE
.
The 2016 WHO classification of tumours of the urinary system and male genital organs-part B: prostate and bladder tumours
.
Eur Urol
2016
;
70
:
106
19
.
3.
Sylvester
RJ
,
van der Meijden
AP
,
Oosterlinck
W
,
Witjes
JA
,
Bouffioux
C
,
Denis
L
, et al
.
Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials
.
Eur Urol
2006
;
49
:
466
5
.
4.
van Rhijn
BW
,
Burger
M
,
Lotan
Y
,
Solsona
E
,
Stief
CG
,
Sylvester
RJ
, et al
.
Recurrence and progression of disease in non-muscle-invasive bladder cancer: from epidemiology to treatment strategy
.
Eur Urol
2009
;
56
:
430
42
.
5.
Tchounwou
PB
,
Dasari
S
,
Noubissi
FK
,
Ray
P
,
Kumar
S
.
Advances in our understanding of the molecular mechanisms of action of cisplatin in cancer therapy
.
J Exp Pharmacol
2021
;
13
:
303
28
.
6.
Chang
JC
.
Cancer stem cells: Role in tumor growth, recurrence, metastasis, and treatment resistance
.
Medicine (Baltimore)
2016
;
95
:
S20
5
.
7.
Drayton
RM
,
Catto
JW
.
Molecular mechanisms of cisplatin resistance in bladder cancer
.
Expert Rev Anticancer Ther
2012
;
12
:
271
81
.
8.
Turner
N
,
Grose
R
.
Fibroblast growth factor signalling: from development to cancer
.
Nat Rev Cancer
2010
;
10
:
116
29
.
9.
Iyer
G
,
Milowsky
MI
.
Fibroblast growth factor receptor-3 in urothelial tumorigenesis
.
Urol Oncol
2013
;
31
:
303
11
.
10.
Dimitroff
CJ
,
Klohs
W
,
Sharma
A
,
Pera
P
,
Driscoll
D
,
Veith
J
, et al
.
Anti-angiogenic activity of selected receptor tyrosine kinase inhibitors, PD166285 and PD173074: implications for combination treatment with photodynamic therapy
.
Invest New Drugs
1999
;
17
:
121
35
.
11.
Mohammadi
M
,
Froum
S
,
Hamby
JM
,
Schroeder
MC
,
Panek
RL
,
Lu
GH
, et al
.
Crystal structure of an angiogenesis inhibitor bound to the FGF receptor tyrosine kinase domain
.
EMBO J
1998
;
17
:
5896
904
.
12.
Pardo
OE
,
Latigo
J
,
Jeffery
RE
,
Nye
E
,
Poulsom
R
,
Spencer-Dene
B
, et al
.
The fibroblast growth factor receptor inhibitor PD173074 blocks small cell lung cancer growth in vitro and in vivo
.
Cancer Res
2009
;
69
:
8645
51
.
13.
Isharwal
S
,
Huang
H
,
Nanjangud
G
,
Audenet
F
,
Chen
YB
,
Gopalan
A
, et al
.
Intratumoral heterogeneity of ERBB2 amplification and HER2 expression in micropapillary urothelial carcinoma
.
Hum Pathol
2018
;
77
:
63
9
.
14.
Koga
F
,
Yoshida
S
,
Tatokoro
M
,
Kawakami
S
,
Fujii
Y
,
Kumagai
J
, et al
.
ErbB2 and NFkappaB overexpression as predictors of chemoradiation resistance and putative targets to overcome resistance in muscle-invasive bladder cancer
.
PLoS One
2011
;
6
:
e27616
.
15.
Hayashi
T
,
Seiler
R
,
Oo
HZ
,
Jager
W
,
Moskalev
I
,
Awrey
S
, et al
.
Targeting HER2 with T-DM1, an antibody cytotoxic drug conjugate, is effective in HER2 over expressing bladder cancer
.
J Urol
2015
;
194
:
1120
31
.
16.
Koshkin
VS
,
O'Donnell
P
,
Yu
EY
,
Grivas
P
.
Systematic review: targeting HER2 in bladder cancer
.
Bladder Cancer
2019
;
5
:
1
12
.
17.
Burger
M
,
Catto
JW
,
Dalbagni
G
,
Grossman
HB
,
Herr
H
,
Karakiewicz
P
, et al
.
Epidemiology and risk factors of urothelial bladder cancer
.
Eur Urol
2013
;
63
:
234
41
.
18.
Boffetta
P
.
Tobacco smoking and risk of bladder cancer
.
Scand J Urol Nephrol Suppl
2008
:
45
54
.
19.
Chen
CH
,
Shun
CT
,
Huang
KH
,
Huang
CY
,
Tsai
YC
,
Yu
HJ
, et al
.
Stopping smoking might reduce tumour recurrence in nonmuscle-invasive bladder cancer
.
BJU Int
2007
;
100
:
281
6
.
20.
Rink
M
,
Zabor
EC
,
Furberg
H
,
Xylinas
E
,
Ehdaie
B
,
Novara
G
, et al
.
Impact of smoking and smoking cessation on outcomes in bladder cancer patients treated with radical cystectomy
.
Eur Urol
2013
;
64
:
456
64
.
21.
Boeri
L
,
Soligo
M
,
Frank
I
,
Boorjian
SA
,
Thompson
RH
,
Tollefson
M
, et al
.
Cigarette smoking is associated with adverse pathological response and increased disease recurrence amongst patients with muscle-invasive bladder cancer treated with cisplatin-based neoadjuvant chemotherapy and radical cystectomy: a single-centre experience
.
BJU Int
2019
;
123
:
1011
9
.
22.
Kim
PH
,
Kent
M
,
Zhao
P
,
Sfakianos
JP
,
Bajorin
DF
,
Bochner
BH
, et al
.
The impact of smoking on pathologic response to neoadjuvant cisplatin-based chemotherapy in patients with muscle-invasive bladder cancer
.
World J Urol
2014
;
32
:
453
9
.
23.
Browman
GP
,
Wong
G
,
Hodson
I
,
Sathya
J
,
Russell
R
,
McAlpine
L
, et al
.
Influence of cigarette smoking on the efficacy of radiation therapy in head and neck cancer
.
N Engl J Med
1993
;
328
:
159
63
.
24.
Moghe
A
,
Ghare
S
,
Lamoreau
B
,
Mohammad
M
,
Barve
S
,
McClain
C
, et al
.
Molecular mechanisms of acrolein toxicity: relevance to human disease
.
Toxicol Sci
2015
;
143
:
242
55
.
25.
Stevens
JF
,
Maier
CS
.
Acrolein: sources, metabolism, and biomolecular interactions relevant to human health and disease
.
Mol Nutr Food Res
2008
;
52
:
7
25
.
26.
Lee
HW
,
Wang
HT
,
Weng
MW
,
Hu
Y
,
Chen
WS
,
Chou
D
, et al
.
Acrolein- and 4-Aminobiphenyl-DNA adducts in human bladder mucosa and tumor tissue and their mutagenicity in human urothelial cells
.
Oncotarget
2014
;
5
:
3526
40
.
27.
Hong
JH
,
Lee
PAH
,
Lu
YC
,
Huang
CY
,
Chen
CH
,
Chiang
CH
, et al
.
Acrolein contributes to urothelial carcinomas in patients with chronic kidney disease
.
Urol Oncol
2020
;
38
:
465
75
.
28.
Wang
HT
,
Chen
TY
,
Weng
CW
,
Yang
CH
,
Tang
MS
.
Acrolein preferentially damages nucleolus eliciting ribosomal stress and apoptosis in human cancer cells
.
Oncotarget
2016
;
7
:
80450
64
.
29.
Borowicz
S
,
Van Scoyk
M
,
Avasarala
S
,
Karuppusamy Rathinam
MK
,
Tauler
J
,
Bikkavilli
RK
, et al
.
The soft agar colony formation assay
.
J Vis Exp
2014;92:
e51998
.
30.
Justus
CR
,
Leffler
N
,
Ruiz-Echevarria
M
,
Yang
LV
.
In vitro cell migration and invasion assays
.
J Vis Exp
2014
:
51046
.
31.
Chen
YT
,
Chen
JJ
,
Wang
HT
.
Targeting RNA polymerase I with hernandonine inhibits ribosomal RNA synthesis and tumor cell growth
.
Mol Cancer Res
2019
;
17
:
2294
305
.
32.
Lamont
FR
,
Tomlinson
DC
,
Cooper
PA
,
Shnyder
SD
,
Chester
JD
,
Knowles
MA
.
Small molecule FGF receptor inhibitors block FGFR-dependent urothelial carcinoma growth in vitro and in vivo
.
Br J Cancer
2011
;
104
:
75
82
.
33.
Williams
SV
,
Hurst
CD
,
Knowles
MA
.
Oncogenic FGFR3 gene fusions in bladder cancer
.
Hum Mol Genet
2013
;
22
:
795
803
.
34.
Carmella
SG
,
Chen
M
,
Zhang
Y
,
Zhang
S
,
Hatsukami
DK
,
Hecht
SS
.
Quantitation of acrolein-derived (3-hydroxypropyl)mercapturic acid in human urine by liquid chromatography-atmospheric pressure chemical ionization tandem mass spectrometry: effects of cigarette smoking
.
Chem Res Toxicol
2007
;
20
:
986
90
.
35.
Siddik
ZH
.
Cisplatin: mode of cytotoxic action and molecular basis of resistance
.
Oncogene
2003
;
22
:
7265
79
.
36.
O'Toole
C
,
Perlmann
P
,
Unsgaard
B
,
Moberger
G
,
Edsmyr
F
.
Cellular immunity to human urinary bladder carcinoma. I. Correlation to clinical stage and radiotherapy
.
Int J Cancer
1972
;
10
:
77
91
.
37.
Wu
K
,
Zeng
J
,
Zhou
J
,
Fan
J
,
Chen
Y
,
Wang
Z
, et al
.
Slug contributes to cadherin switch and malignant progression in muscle-invasive bladder cancer development
.
Urol Oncol
2013
;
31
:
1751
60
.
38.
McConkey
DJ
,
Choi
W
,
Marquis
L
,
Martin
F
,
Williams
MB
,
Shah
J
, et al
.
Role of epithelial-to-mesenchymal transition (EMT) in drug sensitivity and metastasis in bladder cancer
.
Cancer Metastasis Rev
2009
;
28
:
335
44
.
39.
Shibue
T
,
Weinberg
RA
.
EMT, CSCs, and drug resistance: the mechanistic link and clinical implications
.
Nat Rev Clin Oncol
2017
;
14
:
611
29
.
40.
Das
V
,
Bhattacharya
S
,
Chikkaputtaiah
C
,
Hazra
S
,
Pal
M
.
The basics of epithelial-mesenchymal transition (EMT): A study from a structure, dynamics, and functional perspective
.
J Cell Physiol
2019 Feb 5 [Epub ahead of print]
.
41.
Peinado
H
,
Olmeda
D
,
Cano
A
.
Snail Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype?
Nat Rev Cancer
2007
;
7
:
415
28
.
42.
Szala
S
,
Froehlich
M
,
Scollon
M
,
Kasai
Y
,
Steplewski
Z
,
Koprowski
H
, et al
.
Molecular cloning of cDNA for the carcinoma-associated antigen GA733–2
.
Proc Natl Acad Sci U S A
1990
;
87
:
3542
6
.
43.
Wang
MH
,
Sun
R
,
Zhou
XM
,
Zhang
MY
,
Lu
JB
,
Yang
Y
, et al
.
Epithelial cell adhesion molecule overexpression regulates epithelial-mesenchymal transition, stemness and metastasis of nasopharyngeal carcinoma cells via the PTEN/AKT/mTOR pathway
.
Cell Death Dis
2018
;
9
:
2
.
44.
Brunner
A
,
Prelog
M
,
Verdorfer
I
,
Tzankov
A
,
Mikuz
G
,
Ensinger
C
.
EpCAM is predominantly expressed in high grade and advanced stage urothelial carcinoma of the bladder
.
J Clin Pathol
2008
;
61
:
307
10
.
45.
Chakravarti
A
,
Winter
K
,
Wu
CL
,
Kaufman
D
,
Hammond
E
,
Parliament
M
, et al
.
Expression of the epidermal growth factor receptor and Her-2 are predictors of favorable outcome and reduced complete response rates, respectively, in patients with muscle-invading bladder cancers treated by concurrent radiation and cisplatin-based chemotherapy: a report from the Radiation Therapy Oncology Group
.
Int J Radiat Oncol Biol Phys
2005
;
62
:
309
17
.
46.
Salzberg
M
,
Borner
M
,
Bauer
JA
,
Morant
R
,
Rauch
D
,
Rochlitz
C
.
Trastuzumab (Herceptin) in patients with HER-2-overexpressing metastatic or locally advanced transitional cell carcinoma of the bladder: report on 7 patients
.
Eur J Cancer
2006
;
42
:
2660
1
.
47.
Peyromaure
M
,
Scotte
F
,
Amsellem-Ouazana
D
,
Vieillefond
A
,
Oudard
S
,
Beuzeboc
P
.
Trastuzumab (Herceptin) in metastatic transitional cell carcinoma of the urinary tract: report on six patients
.
Eur Urol
2005
;
48
:
771
5
.
48.
Hussain
MH
,
MacVicar
GR
,
Petrylak
DP
,
Dunn
RL
,
Vaishampayan
U
,
Lara
PN
Jr.
, et al
.
Trastuzumab, paclitaxel, carboplatin, and gemcitabine in advanced human epidermal growth factor receptor-2/neu-positive urothelial carcinoma: results of a multicenter phase II National Cancer Institute trial
.
J Clin Oncol
2007
;
25
:
2218
24
.
49.
Cimpean
AM
,
Tarlui
V
,
Cumpanas
AA
,
Bolintineanu
S
,
Cumpanas
A
,
Raica
M
.
Critical overview of HER2 assessement in bladder cancer: what is missing for a better therapeutic approach?
Anticancer Res
2017
;
37
:
4935
42
.
50.
Oudard
S
,
Culine
S
,
Vano
Y
,
Goldwasser
F
,
Theodore
C
,
Nguyen
T
, et al
.
Multicentre randomised phase II trial of gemcitabine+platinum, with or without trastuzumab, in advanced or metastatic urothelial carcinoma overexpressing Her2
.
Eur J Cancer
2015
;
51
:
45
54
.
51.
Billerey
C
,
Chopin
D
,
Aubriot-Lorton
MH
,
Ricol
D
,
Gil Diez de Medina
S
,
Van Rhijn
B
, et al
.
Frequent FGFR3 mutations in papillary non-invasive bladder (pTa) tumors
.
Am J Pathol
2001
;
158
:
1955
9
.
52.
Tomlinson
DC
,
Baldo
O
,
Harnden
P
,
Knowles
MA
.
FGFR3 protein expression and its relationship to mutation status and prognostic variables in bladder cancer
.
J Pathol
2007
;
213
:
91
8
.
53.
Xie
X
,
Lin
J
,
Zhong
Y
,
Fu
M
,
Tang
A
.
FGFR(3S249C) mutation promotes chemoresistance by activating Akt signaling in bladder cancer cells
.
Exp Ther Med
2019
;
18
:
1226
34
.

Supplementary data