Effect of docetaxel on the regulation of proliferation and apoptosis of human prostate cancer cells

  • Authors:
    • Chongyi Yang
    • Weijie Zhang
    • Jie Wang
    • Pengpeng Chen
    • Jiangjiang Jin
  • View Affiliations

  • Published online on: March 1, 2019     https://doi.org/10.3892/mmr.2019.9998
  • Pages: 3864-3870
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer is a common type of malignancy. Given the complexity of prostate cancer and the pressing challenge of chemoresistance, the current study was conducted to investigate the effect of docetaxel (Doc) on androgen receptor (AR)‑dependent and AR‑independent prostate cancers cells. Subsequent experiments were designed to explore the mechanism underlying the Doc‑induced apoptosis. Three different human prostate cancer cell lines, namely PC‑3, LNCaP and DU‑145, were exposed to various concentrations of Doc. The cytotoxic effects of Doc were evaluated by an MTT assay, while apoptosis and cell cycle distribution were determined by flow cytometric analysis of cells stained with Annexin V‑FITC and propidium iodide. Western blot assay was also used to measure the protein levels of B‑cell lymphoma 2 (Bcl‑2), Bcl‑2‑associated death promoter (Bad), total protein kinase B (Akt), phospho‑Akt and caspase‑3/9. Doc induced cytotoxicity in all three cell lines in a dose‑dependent manner. The half maximal inhibitory concentration values for the effect of Doc on PC‑3, DU‑145 and LNCaP cells were 3.72, 4.46 and 1.13 nM, respectively. Furthermore, the results indicated a significant difference in Doc sensitivity between AR‑dependent and AR‑independent prostate cancer cells. Evaluation of key gene expression at protein levels revealed a notable decrease in antiapoptotic Bcl‑2 and p‑Akt levels, along with a significant increase in pro‑apoptotic Bad, caspase‑3 and caspase‑9 levels. Therefore, Doc may induce cell apoptosis in prostate cancer via various pathways.

Introduction

Prostate cancer is the most frequently diagnosed non-cutaneous cancer in males in western countries (1). The majority of prostate cancer cases are diagnosed at a local stage and have a 5-year survival rate of almost 100% (2). However, prostate cancer remains a major cause of cancer-associated mortality due to its heterogeneous nature, ranging from asymptomatic to a rapidly fatal systemic malignancy. One of the most challenging aspects of prostate cancer is that the androgen receptor-dependent tumors inevitably progress to highly aggressive castration-resistant tumors following initial androgen-ablation therapy.

Only a limited number of effective therapeutic options are available for advanced prostate cancer. Docetaxel (Doc), a semi-synthetically taxane analogue, has displayed promising therapeutic potential in treating advanced-stage prostate cancer. In clinical practice, it is recommended that combination of Doc and prednisone should be used in the treatment of prostate cancer patients to improve overall survival and disease control (3). However, progression is ultimately observed after 6–8 months in patients treated with Doc due to inherent or acquired drug resistance (4,5).

Doc is considered to function as a microtubule inhibitor by binding to β-tubulin and preventing microtubule disassembly. Hence, Doc is able to arrest cells in the G2/M-phase of the cell cycle and induce cell death. A number of studies have suggested that Doc induces phosphorylation of B-cell lymphoma 2 (Bcl-2) and Bcl-xL members, thus inactivating their anti-apoptotic capacities (6). In addition, the mitogen-activated protein kinase pathway is considered to be involved in the development of drug resistance in prostate cancer cells (7,8). Nevertheless, the association between the expression of Bcl-2 family members and Doc resistance remains controversial (9). Overexpression of pro-apoptotic proteins caspase-9 and Bcl-2 interacting protein 3 in resistant prostate cancer cells was observed in previous studies (10,11). However, few studies have carefully examined the regulation of these mechanisms in response to initial treatment with Doc. Thus, identification of the mechanistic basis of Doc-induced cell death in prostate cancer will improve our understanding on the resistance mechanism.

In the present study, the antiproliferative effect of Doc on androgen-dependent and androgen-independent human prostate cancer cells was investigated. In addition, we also investigated signaling pathways involved in Doc-induced cell apoptosis.

Materials and methods

Reagents

MTT was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Dulbecco's modified Eagle's medium (DMEM) and Annexin V-FITC/propidium iodide (PI) were purchased from Beyotime Institute of Biotechnology (Hangzhou, China). Fetal bovine serum (FBS) was obtained from Sijingqing Biotechnology Co., Ltd. (Hangzhou, China). Monoclonal antibodies against total protein kinase B (Akt; 1:1,000; cat. no. ab179463; Abcam, Cambridge, UK), phospho-Akt (1:1,000; cat. no. ab133458; Abcam), Bcl-2 (1:1,000; cat. no. ab59348; Abcam), Bcl-2-associated death promoter (Bad; 1:1,000; cat. no. ab32445; Abcam), caspase-3 (1:1,000; cat. no. ab4051; Abcam), caspase-9 (1:1,000; cat. no. ab52298; Abcam) and β-actin (1:1,000; cat. no. ab16039; Abcam) were used as primary antibodies. Horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G (1:1,000; cat. no. 31460; Thermo Fisher Scientific, Inc., Waltham, MA, USA) was used as secondary antibody.

Cell culture

Three human prostate cancer cell lines, namely PC-3 [androgen receptor (AR)-negative], DU-145 (AR-negative) and LNCaP (AR-sensitive) were purchased from The American Type Culture Collection (Manassas, VA, USA) and were maintained in DMEM supplemented with 10% (v/v) FBS and antibiotics at 37°C with 5% CO2. After 3–4 days of culture, the cells were detached from the surface by trypsinization and reseeded into the appropriate plates for use in further experiments.

MTT assay

Cell proliferation was evaluated by an MTT assay. Briefly, cells were seeded into 96-well plates at a density of 4×103 cells per well in 100 µl medium and cultured for ~24 h. Next, PC-3, DU-145 and LNCaP cells were treated with Doc at the doses of 0–64, 0–40 and 0–8 nM, respectively. Six replicated samples were assayed for each concentration. Cells were cultured for 48 h, followed by the addition of 20 µl MTT in each well. After 4 h, the supernatant were discarded, 200 µl dimethyl sulfoxide (DMSO) was added to each well, and the plate was placed on a shaker for 10 min to completely dissolve DMSO. Absorbance was then measured at wavelengths of 490 and 630 nM. The half maximal inhibitory concentration (IC50) was calculated using Origin software, version 8.5 (OriginLab Corporation, Northampton, MA, USA).

Cell apoptosis assay

Cells in the logarithmic growth phase were seeded in 6-well plates at a density of 2×105 cells per well. The three cell lines were divided into three groups each and then incubated with two different concentrations of Doc (4 and 10 nM) as the positive controls and in the absence of Doc as the negative control. After 48 h, cells were harvested, washed in cold phosphate-buffered saline (PBS), and then treated with Annexin V-FITC and PI using an Apoptosis Detection kit with PI, according to the manufacturer's protocol. Annexin V-FITC and PI binding was detected with an Attune flow cytometer (Thermo Fisher Scientific, Inc.), and flow cytometric analysis was performed using BD CellQuest software (BD Biosciences, Franklin Lakes, NJ, USA).

Cell cycle analysis

The DNA content during the cell cycle phases was evaluated by flow cytometry. Briefly, 4×105 PC-3, DU-145 and LNCaP cells were seeded and were treated with Doc at concentrations of 4, 10 and 2 nM, respectively. Cells were then incubated for 48 h, and cell pellets were washed and fixed in cold 70% ethanol at 4°C overnight. On the following day, the cell pellets were washed three times and resuspended in PBS, followed by treatment with RNase (50 µg/ml) for 1 h. Next, 5 µl PI (1 mg/ml) was added to a final concentration of 50 µg/ml and incubated for 1 h at room temperature in the dark. Subsequent to staining of DNA by PI, samples were evaluated using the Attune flow cytometer, and analysis was performed using the BD CellQuest software.

Protein extraction and western blotting

Following treatment for 24 or 48 h, total protein was extracted using a cell lysis buffer (Thermo Fisher Scientific, Inc.). Protein concentration in the supernatant was determined using a Bio-Rad protein assay kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Next, protein (20 µg) was subjected to 12% SDS-PAGE and transferred to a polyvinylidene difluoride membrane. Subsequent to blocking in non-fat dry milk for 2 h at room temperature, the membrane was incubated overnight at 4°C with total Akt, phospho-Akt, Bcl-2, Bad, caspase-3 and caspase-9 primary antibodies. The membrane was then washed three times with Tris-buffered saline (10 min each time), followed by incubation with secondary antibody for 2 h at room temperature. Signal development was performed using an enhanced chemiluminescence kit. Signals were captured and band densities were quantified using Bandscan software, version 5.0 (ProZyme; Agilent Technologies, Inc., Santa Clara, CA, USA).

Statistical analysis

Data are presented as the mean ± standard deviation. Analysis of variance with Tukey-Kramer test adjustment was used for comparisons among multiple groups, while Student' test was used to examine comparisons between two groups. All analyses were performed using SPSS software, version 11.0 (SPSS, Inc., Chicago, IL, USA). Probability values of P<0.05 were considered to denote statistically significant differences.

Results

Effects of Doc on the growth of human prostate cancer cells

Following treatment for 48 h, the inhibitory effects of Doc on the three prostate cancer cell lines PC-3, DU-145 and LNCaP were explored by an MTT assay. The results presented in Fig. 1 demonstrate that, upon treatment with Doc, cell viability in the three cell lines decreased in a concentration-dependent manner. The IC50 values of the effect of Doc on PC-3, DU-145 and LNCaP cells were 3.72, 4.46 and 1.13 nM, respectively. Among the three cell lines, LNCaP is an androgen-dependent prostate cancer cell line, while the other two cell lines are androgen-independent (12). The study results indicated that PC-3 and DU-145 cells were more resistant to Doc treatment, as their IC50 values were approximately threefold and fourfold higher in comparison with that of LNCaP cells, respectively.

Effects of Doc on the apoptosis of human prostate cancer cells

To examine the mechanism underlying the antitumor effect of Doc, flow cytometric analysis with Annexin V/PI staining was performed. Cells were exposed to low and high concentrations of Doc, which were determined according to the IC50 value of each cell line. The low Doc dose for PC-3, DU-145 and LNCaP cells was 0.75, 2 and 0.5 nM, respectively, while the high dose was 3, 4 and 1 nM, respectively. As presented in Fig. 2, low concentrations of Doc had no effect on cell death in all cells lines. Compared with the negative control and with the cells treated with a low concentration of Doc, treatment with high dose of Doc significantly increased the proportion of Annexin V+ apoptotic cells.

Effect of Doc on cell cycle phase distribution

Cultured human prostate cancer PC-3, DU-145 and LNCaP cells were exposed to Doc at the concentrations of 4, 10 and 2 nM, respectively. As presented in Fig. 3, it was observed that treatment with Doc led to marked cell cycle arrest at G2/M phase, as observed by the significant increase in the percentage of cells at this phase in PC-3 and DU145 cells, as compared with the untreated cells.

Effects of Doc on the protein levels of Bcl-2, Bad, phospho-Akt and caspase-3/9

The study next explored the expression of proteins associated with the development of Doc resistance in prostate cancer cells. Western blot assay was used to assess the expression of Bcl-2, total Akt, phospho-Akt, Bcl-2, Bad, caspase-3 and caspase-9 at the protein levels after 24 and 48 h of Doc treatment. As shown in Fig. 4, compared with the control group, treatment with Doc for 24 h led to significantly decreased expression of anti-apoptotic Bcl-2 protein. The activity of p-Akt was decreased significantly, while total Akt level remained stable. Treatment of PC-3, DU-145 and LNCaP cell lines with Doc also resulted in significant increase in the levels of caspase-3, caspase-9 and pro-apoptotic protein Bad. Protein expression was further examined at 48 h, and similar results were obtained (Fig. 5).

Discussion

In spite of the significantly improved survival, prostate cancer remains a clinical challenge due to the fact that, in a certain portion of patients, prostate cancer may progress to metastatic castration-resistant prostate cancer with no curative treatment options (2,3). Doc is an effective cytotoxic agent that provides survival benefits for patients with castration-resistant tumors (13). However, clinical resistance to Doc remains a challenge in clinical practice. Growing evidence has demonstrated the complexity of the mechanism underlying Doc resistance, which includes cellular anti-apoptotic, AR-mediated redox signaling pathways (1416). In the current study, the inhibitory and apoptotic effects of Doc on androgen-dependent or androgen-independent prostate cancer cells were initially examined. The study then attempted to explore the regulation of Bcl-2 and phosphoinositide 3-kinase (PI3K)/Akt signaling pathway in Doc-induced apoptosis.

The results reported in the present study confirmed that Doc exerted an inhibitory effect on the growth of cultured prostate cancer cells in a dose-independent manner. The IC50 values for the effect of Doc in PC-3, DU-145 and LNCaP cells were 3.72, 4.46 and 1.13 nM, respectively. These results indicated that Doc had a stronger inhibitory effect on the AR-dependent LNCaP cells, as compared with that on AR-independent prostate cancer cells PC-3 and DU-145. In respect to Doc-induced apoptosis, the current data are in agreement with previous findings indicating that a low dose of Doc causes no apoptotic cell death, which is accompanied by senescence necrosis and mitotic catastrophe (1719). AR signaling is known to serve a crucial role in the progression of prostate cancer. Dysregulation of AR signaling and transcriptional activity stimulates resistance to Doc (20). Aberrant AR pathway activation and various splice variant isoforms are frequently observed in advanced prostate cancer (21). At present, the molecular mechanism of the differences in Doc sensitivity is unknown. In a small scale clinical trial, Doc was proven to exert a therapeutic effect on early hormone-sensitive prostate cancer, without affecting testosterone levels (22). In future studies, it would be of interest to explore the impact of low-dose Doc treatment on hormone-sensitive prostate cancer compared with the effect of traditional anti-androgen therapy. It would also be interesting to explore the mechanical differences between inherent and therapy-induced AR activation upon initial treatment with Doc.

To examine whether the strong effects of Doc treatment on the three prostate cancer cell lines were mediated through apoptosis-associated signaling pathways, the expression levels of associated genes were assessed at the protein levels in the present study. Previous studies have demonstrated that taxanes inhibit the anti-apoptotic protein Bcl-2 by stabilization of microtubules, leading to apoptosis (20,23). Bcl-2, a proto-oncogene, is one of the most widely studied negative regulators of apoptotic cascades. It is considered that Bcl-2 prevents the release of cytochrome c and consequently blocks the activation of the caspase cascade (24). In the present study, it was observed that treatment of prostate cancer cells lines PC-3, DU-145 and LNCaP with Doc significantly inhibited Bcl-2 activity, and further upregulated the pro-apoptotic Bad, caspase-3 and caspase-9 proteins. Recent studies suggested that activation of Akt directly affects the apoptosis pathway by targeting and downregulating the levels of Bcl-2 family members, including Bad and Bcl-2-associated X protein, resulting in cell survival (25,26). Emerging evidence revealed a negative feedback link between PI3K/Akt and AR (25,26). Furthermore, activation of Akt induced the phosphorylation of AR, resulting in inhibition of the AR-induced apoptotic pathway, whereas AR inhibition was reported to activate the PI3K/Akt signaling pathway (25,26). The results of the present study revealed that treatment of the three prostate cancer cell lines with Doc markedly decreased the level of phospho-Akt. A certain decrease, although not significant, was also observed in AR-dependent prostate cancer cells (namely LNCaP), as compared with that detected in AR-independent prostate cancer cells, implying a potential interplay between AR and PI3K/Akt when prostate cancer cells are treated with Doc. Given the central role of PI3K/Akt signaling pathway on the growth, proliferation, motility, survival and angiogenesis of tumor cells (27,28), there is great need for understanding the associations between PI3K/Akt and AR pathway in Doc-induced apoptosis. In addition, other pathways may participate in Doc-induced apoptosis through AR-dependent or AR-independent pathways; however, further studies are required to investigate the involvement of other pathways.

It has been reported that the development of Doc resistance in prostate cancer is associated with AR activation (29), which is consistent with the findings of the present study, further confirming the reliability of our data. Nevertheless, compared with a 2D cell culture system, a 3D cell culture system that exhibits a more similar behavior to in vivo conditions (30) should be performed to further confirm the conclusions of the present study. Doc has been widely used in the clinical treatment of prostate cancer (31), and multiple pathways have been proven to be involved in the Doc-mediated therapeutic response in this disease, such as p53/p21WAF1/CIP1, p27KIP1 and Notch pathways (32,33). However, the involvement of Akt signaling in this process is rarely studied. Thus, the present study systemically investigated the role of Akt signaling in Doc-mediated therapeutic responses in prostate cancer. A number of pathways, such as type I insulin-like growth factor (34), have been proven to be involved in androgen-dependent and androgen-independent prostate cancer, and our future studies will focus on these pathways.

In conclusion, the present study demonstrated that Doc strongly inhibited the growth and induced the apoptosis of human prostate cancer cells. AR-dependent prostate cancer cells were more sensitive to Doc in comparison with androgen-independent cells. The effects of Doc on growth inhibition and apoptosis in prostate cancer cells were associated with inhibition of PI3K/Akt activation, decreased levels of Bcl-2 and increased caspase-3/9 activation.

Acknowledgements

Not applicable.

Funding

The present study was funded by The Agricultural and Social Class Science and Technology Plan Project of Ninghai County (grant no. 2013B11).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JJ and CY designed and conceived the study. CY and WZ performed experiments and collected the data. JW and PC analyzed and interpreted the data. JJ wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Eyre H, Kahn R and Robertson RM; ACS/ADA/AHA Collaborative Writing Committee, : Preventing cancer, cardiovascular disease, and diabetes: A common agenda for the American cancer society, the American diabetes association, and the American heart association. CA Cancer J Clin. 54:190–207. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Brunelli A, Kim AW, Berger KI and Addrizzo-Harris DJ: Physiologic evaluation of the patient with lung cancer being considered for resectional surgery: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 143 (Suppl 5):e166S–e190S. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Lee JL, Kim JE, Ahn JH, Lee DH, Lee J, Kim CS, Hong JH, Hong B, Song C and Ahn H: Efficacy and safety of docetaxel plus prednisolone chemotherapy for metastatic hormone-refractory prostate adenocarcinoma: Single institutional study in Korea. Cancer Res Treat. 42:12–17. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Petrylak DP, Tangen CM, Hussain MH, Lara PN Jr, Jones JA, Taplin ME, Burch PA, Berry D, Moinpour C, Kohli M, et al: Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. 351:1513–1520. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Montero A, Fossella F, Hortobagyi G and Valero V: Docetaxel for treatment of solid tumours: A systematic review of clinical data. Lancet Oncol. 6:229–239. 2005. View Article : Google Scholar : PubMed/NCBI

7 

West KA, Castillo SS and Dennis PA: Activation of the PI3K/Akt pathway and chemotherapeutic resistance. Drug Resist Updat. 5:234–248. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Pritchard AL and Hayward NK: Molecular pathways: Mitogen-activated protein kinase pathway mutations and drug resistance. Clin Cancer Res. 19:2301–2309. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Mohammadian J, Sabzichi M, Molavi O, Shanehbandi D and Samadi N: Combined treatment with stattic and docetaxel alters the Bax/Bcl-2 gene expression ratio in human prostate cancer cells. Asian Pac J Cancer Prev. 17:5031–5035. 2016.PubMed/NCBI

10 

Yoo NJ, Kim MS and Park SW: Expression analysis of caspase-6, caspase-9 and BNIP3 in prostate cancer. Tumori. 96:138–142. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Winter RN, Kramer A, Borkowski A and Kyprianou N: Loss of caspase-1 and caspase-3 protein expression in human prostate cancer. Cancer Res. 61:1227–1232. 2001.PubMed/NCBI

12 

Webber MM, Bello D and Quader S: Immortalized and tumorigenic adult human prostatic epithelial cell lines: Characteristics and applications Part 2. Tumorigenic cell lines. Prostate. 30:58–64. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Basch EM, Somerfield MR, Beer TM, Carducci MA, Higano CS, Hussain MH and Scher HI; American Society of Clinical Oncology, : American Society of Clinical Oncology endorsement of the cancer care ontario practice guideline on nonhormonal therapy for men with metastatic hormone-refractory (castration-resistant) prostate cancer. J Clin Oncol. 25:5313–5318. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Hughes C, Murphy A, Martin C, Sheils O and O'Leary J: Molecular pathology of prostate cancer. J Clin Pathol. 58:673–684. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Muenchen HJ, Poncza PJ and Pienta KJ: Different docetaxel-induced apoptotic pathways are present in prostate cancer cell lines LNCaP and PC-3. Urology. 57:366–370. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Hwang C: Overcoming docetaxel resistance in prostate cancer: A perspective review. Ther Adv Med Oncol. 4:329–340. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Morse DL, Gray H, Payne CM and Gillies RJ: Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol Cancer Ther. 4:1495–1504. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Fabbri F, Amadori D, Carloni S, Brigliadori G, Tesei A, Ulivi P, Rosetti M, Vannini I, Arienti C, Zoli W and Silvestrini R: Mitotic catastrophe and apoptosis induced by docetaxel in hormone-refractory prostate cancer cells. J Cell Physiol. 217:494–501. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Fabbri F, Carloni S, Brigliadori G, Zoli W, Lapalombella R and Marini M: Sequential events of apoptosis involving docetaxel, a microtubule-interfering agent: A cytometric study. BMC Cell Biol. 7:62006. View Article : Google Scholar : PubMed/NCBI

20 

Kahn B, Collazo J and Kyprianou N: Androgen receptor as a driver of therapeutic resistance in advanced prostate cancer. Int J Biol Sci. 10:588–595. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Attar RM, Takimoto CH and Gottardis MM: Castration-resistant prostate cancer: Locking up the molecular escape routes. Clin Cancer Res. 15:3251–3255. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Gravis G, Fizazi K, Joly F, Oudard S, Priou F, Esterni B, Latorzeff I, Delva R, Krakowski I, Laguerre B, et al: Androgen-deprivation therapy alone or with docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15): A randomised, open-label, phase 3 trial. Lancet Oncol. 14:149–158. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Loriot Y and Fizazi K: Taxanes: Still a major weapon in the armamentarium against prostate cancer. Eur Urol. 63:983–985. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Akinleye A, Avvaru P, Furqan M, Song Y and Liu D: Phosphatidylinositol 3-kinase (PI3K) inhibitors as cancer therapeutics. J Hematol Oncol. 6:882013. View Article : Google Scholar : PubMed/NCBI

25 

Morgan TM, Koreckij TD and Corey E: Targeted therapy for advanced prostate cancer: Inhibition of the PI3K/Akt/mTOR pathway. Curr Cancer Drug Targets. 9:237–249. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Lin HK, Yeh S, Kang HY and Chang C: Akt suppresses androgen-induced apoptosis by phosphorylating and inhibiting androgen receptor. Proc Natl Acad Sci USA. 98:7200–7205. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Thomas C, Lamoureux F, Crafter C, Davies BR, Beraldi E, Fazli L, Kim S, Thaper D, Gleave ME and Zoubeidi A: Synergistic targeting of PI3K/AKT pathway and androgen receptor axis significantly delays castration-resistant prostate cancer progression in vivo. Mol Cancer Ther. 12:2342–2355. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Osaki M, Oshimura M and Ito H: PI3K-Akt pathway: Its functions and alterations in human cancer. Apoptosis. 9:667–676. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Komura K, Jeong SH, Hinohara K, Qu F, Wang X, Hiraki M, Azuma H, Lee GS, Kantoff PW and Sweeney CJ: Resistance to docetaxel in prostate cancer is associated with androgen receptor activation and loss of KDM5D expression. Proc Natl Acad Sci USA. 113:6259–6264. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Souza AG, Silva IBB, Campos-Fernandez E, Barcelos LS, Souza JB, Marangoni K, Goulart LR and Alonso-Goulart V: Comparative assay of 2D and 3D cell culture models: Proliferation, gene expression and anticancer drug response. Curr Pharm Des. 24:1689–1694. 2018. View Article : Google Scholar : PubMed/NCBI

31 

James ND and Mason M: Docetaxel and/or zoledronic acid for hormone-naïve prostate cancer: First survival results from STAMPEDE. J Clin Oncol. 33:50012015. View Article : Google Scholar

32 

Singh SK, Banerjee S, Acosta EP, Lillard JW and Singh R: Resveratrol induces cell cycle arrest and apoptosis with docetaxel in prostate cancer cells via a p53/p21WAF1/CIP1 and p27KIP1 pathway. Oncotarget. 8:17216–17228. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Cui D, Dai J, Keller JM, Mizokami A, Xia S and Keller ET: Notch pathway inhibition using PF-03084014, a γ-secretase inhibitor (GSI), enhances the antitumor effect of docetaxel in prostate cancer. Clin Cancer Res. 21:4619–4629. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Wu JD, Odman A, Higgins LM, Haugk K, Vessella R, Ludwig DL and Plymate SR: In vivo effects of the human type I insulin-like growth factor receptor antibody A12 on androgen-dependent and androgen-independent xenograft human prostate tumors. Clin Cancer Res. 11:3065–3074. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 19 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang C, Zhang W, Wang J, Chen P and Jin J: Effect of docetaxel on the regulation of proliferation and apoptosis of human prostate cancer cells. Mol Med Rep 19: 3864-3870, 2019
APA
Yang, C., Zhang, W., Wang, J., Chen, P., & Jin, J. (2019). Effect of docetaxel on the regulation of proliferation and apoptosis of human prostate cancer cells. Molecular Medicine Reports, 19, 3864-3870. https://doi.org/10.3892/mmr.2019.9998
MLA
Yang, C., Zhang, W., Wang, J., Chen, P., Jin, J."Effect of docetaxel on the regulation of proliferation and apoptosis of human prostate cancer cells". Molecular Medicine Reports 19.5 (2019): 3864-3870.
Chicago
Yang, C., Zhang, W., Wang, J., Chen, P., Jin, J."Effect of docetaxel on the regulation of proliferation and apoptosis of human prostate cancer cells". Molecular Medicine Reports 19, no. 5 (2019): 3864-3870. https://doi.org/10.3892/mmr.2019.9998