Anticancer effects of Ac‑Phe‑Lys‑PABC‑doxorubicin via mitochondria‑centered apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway in MGC‑803 cells

  • Authors:
    • Yan‑Jun Zhong
    • Shao‑Ping Liu
    • Raymond A. Firestone
    • Ya‑Ping Hong
    • Yan Li
  • View Affiliations

  • Published online on: July 19, 2013     https://doi.org/10.3892/or.2013.2629
  • Pages: 1681-1686
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ac‑Phe‑Lys‑PABC‑DOX (PDOX) is a smart doxorubicin (DOX) prodrug designed to decrease toxicities while maintaining the potent anticancer effects of DOX. The present study aimed to elucidate the molecular mechanisms of action of PDOX using MGC‑803 gastric cancer cells as a model. The cells were treated with both PDOX and DOX, and cytotoxicities, cell cycle analysis, reactive oxygen species (ROS) generation, mitochondrial damage and ERK1/2 signaling pathway alterations were studied. Abundant cathepsin B expression was observed in the MGC‑803 cells, and treatment with PDOX and DOX triggered dose‑dependent cytotoxicity and resulted in a significant reduction in cell viability. IC50 of PDOX and DOX was 14.9 and 4.9 µM, respectively. Both PDOX and DOX significantly decreased p‑ERK1/2, increased ROS generation, reduced mitochondrial membrane potential, caused mitochondrial swelling and arrested the cell cycle at the G2/S phase, and these effects were more pronounced for PDOX than for DOX. PDOX and DOX have different mechanisms of action, particularly the mitochondria‑centered intrinsic apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway.

Introduction

Doxorubicin (DOX, Adriamycin), an anthracycline isolated from Streptomyces strains, is one of the most efficacious anticancer drugs for the treatment of hematological malignancies and a broad range of solid tumors (1,2). However, the clinical application of DOX has long been limited by its dose-dependent toxicities to the heart, kidney, liver and bone marrow. To reduce these side effects, significant efforts have been made to develop DOX-prodrugs, such as PK1 and PK2, which remain inactive in blood and normal tissues but release DOX at the tumor site (3), and which have entered phase II/III clinical studies (4,5).

Cathepsin B (Cat B), a lysosomal cysteine protease in normal cells and tissues, is highly upregulated in malignant tumors and premalignant lesions at the mRNA, protein and activity levels (6). The active cleavage sites of Cat B cover a range of oligopeptides, including Arg-Arg, Ala-Leu, Phe-Arg, Phe-Lys, Ala-Phe-Lys, Gly-Leu-Phe-Gly, Gly-Phe-Leu-Gly and Ala-Leu-Ala-Leu (711). Therefore, several low- and high-molecular-weight DOX prodrugs through Cat B-cleavable oligopeptides, have been designed (4,5,12) and have demonstrated rapid and nearly quantitative DOX release in the presence of Cat B.

Based on the characteristics of Cat B, we designed and developed a smart DOX prodrug, Ac-Phe-Lys-PABC-DOX (PDOX), in which a Cat B-specific dipeptide (Phe-Lys) is introduced, containing a self-immolative spacer para-aminobenzyloxycarbonyl (PABC) to increase the distance between the dipeptide and DOX (1316), so that the dipeptide can be directly accessable to the Cat B′ active site. PDOX remains inactive and stable in blood circulation and normal tissues. When PDOX reaches Cat B-enriched areas such as tumor sites, the dipeptide Phe-Lys is cleaved by Cat B, exposing the PABC spacer that is hydrolyzed spontaneously, releasing free DOX.

An in vivo study in a nude mouse model of gastric cancer peritoneal carcinomatosis showed that, compared with free DOX, PDOX produced superior anti-metastasis effects in terms of the experimental peritoneal carcinomatosis index (ePCI) and body weight, and reduced toxicities to the liver, kidney and particularly the heart (16). However, the underlying mechanism remains unclear.

It has been reported that apoptosis plays a key role in the anticancer effects and toxicities of DOX (17), mainly via the mitochondria-centered intrinsic pathway (18,19), which involves the release of cytochrome c from the mitochondria (20,21). DOX promotes the generation of reactive oxygen species (ROS) in various cell types (22,23) and mitochondria are the major source of ROS production (24). The present study was aimed to investigate the in vitro mechanisms of action of PDOX with special focus on the mitochondria-centered intrinsic pathway.

Materials and methods

Drug preparation and cell culture

DOX (Pharmacia, Milan, Italy) and PDOX (synthesized by Y.-P.H.) were used. MGC-803 gastric cancer cells were cultured in 25 cm2 tissue culture flasks at 37°C in 5% CO2, 95% air and 100% humidity. Cells were grown in RPMI-1640 medium containing 10% newborn calf serum and 1% penicillin and streptomycin. Throughout the study, the medium was replaced every 2 to 3 days. The cells were passaged when grown to 90% confluence.

Immunocytochemistry

Cells were transferred onto a coverslip in 6-well culture plates, grown for 2 days to reach 90% confluence and then fixed by ice-cold 4% paraformaldehyde for 30 min. Immunocytochemistry for Cat B was then performed following a standard method (25). Briefly, cells were first blocked with 2% bovine serum albumin (BSA) at 37°C for 30 min. Next, cells were incubated with a primary rabbit anti-human Cat B antibody (cat no. 3190-100, dilution 1:200; BioVision, Mountain View, CA, USA) at 4°C overnight. Cells were washed with Tris-buffered saline-Tween (TBST; 0.05% Tween, 0.1 M Tris-base, 0.9% NaCl, pH 7.6) 3 times and then incubated with a peroxidase-labled goat anti-rabbit IgG at 37°C for 30 min. The antibody reaction products were visualized with diaminobenzidine (Dako, Denmark). The images were captured using an Olympus BX51 fluorescence microscope equipped with an Olympus Micro DP 72 camera.

Cell viability study by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay

Cells were passaged and 1×104 cells in 100 μl medium were transferred into 96-well culture plates. After 24 h, the MGC-803 cells were treated with 0, 0.86, 1.72, 3.44, 6.88 and 13.76 μM of PDOX, or 0, 0.86, 1.72, 3.44, 6.88 and 13.76 μM DOX in medium for 24, 48, 72 and 96 h, respectively; then immediately incubated with 100 μl 0.5 mg/ml MTT at 37°C in 5%CO2, 95% air and 100% humidity for 4 h. After medium removal, 150 μl DMSO was added and incubated for 10 min. OD560 was obtained by the ELISA (modulus microplate), and IC50 values were calculated.

Cell cycle analysis by flow cytometry

MGC-803 cells (2×105) in 2 ml of medium were transferred into 6-well culture plates and cultured for 24 h. The cells were treated with 0, 14.9 μM (IC50 of PDOX treated for 48 h), 4.9 μM PDOX and 14.9 and 4.9 μM (IC50 of DOX treated for 48 h) DOX for 48 h. The harvested cells were subjected to flow cytometry assay by Coulter® DNA PREP™ reagents kit according to the manufacturer’s protocol. Briefly, cells were harvested, washed twice with phosphate-buffered saline (PBS) and re-suspended in serum-free medium, then 50 μl DNA PREP™ LPR buffer was added and incubated in a light-protected moist chamber for 20 min. Next, 500 μl DNA PREP™ stain was added and incubated in the dark for 20 min. Finally, cell cycle analysis was performed by flow cytometry (FC 500; Beckman Coulter, USA).

ROS generation and mitochondrial membrane potential (ΔΨm) measurement

ROS generation was assayed using dichlorofluorescein diacetate (DCF-DA; Sigma, USA). The polar derivative from DCF-DA by intracellular esterases rapidly reacts with ROS to form a highly fluorescent compound (26). MGC-803 cells (2×105) in 2 ml of medium were transferred into 6-well culture plates for 24 h. The medium was replaced with serum-free medium the following day. Twelve hours later, cells were treated with 0, 4.9 and 14.9 μM PDOX or 4.9 and 14.9 μM DOX, respectively, for 48 h. For ROS generation, cells were immediately incubated with 100 μM DCF-DA at 37°C for 1 h in the dark. Images were captured using a laser confocal scanning microscope (Leica, Wetzlar, Germany) at 488 nm excitation and 525 nm emission.

For ΔΨm assay, MGC-803 cells were treated in the same way as above and then immediately incubated with 100 nM MitoTracker® Red CMXRos at 37°C for 30 min in the dark. Images were captured by a laser confocal scanning microscope at 579 nm excitation and 599 nm emission. For semi-quantitative analysis of ROS production and ΔΨm, total fluorescence intensity was analyzed with the LCS lite (Leica).

Mitochondrial morphology as determined by transmission electron microscopy

MGC-803 cells (2×106) in 8 ml of medium were transferred into 10-cm culture dishes. The cells were treated in the same way as above and harvested cells were initially fixed in 2.5% glutaraldehyde, then post-fixed in 1% osmic acid and embedded in epoxide resin. Ultrathin sections were cut with an ultramicrotome (LKB-V; Bromma, Sweden), stained with uranyl acetate and lead citrate, and examined using a transmission electron microscope (H-600; Hitachi, Japan) and photographed.

Western blotting

MGC-803 cells (2×105) in 2 ml of medium were transferred into 6-well culture plates and treated in the same way as above. The harvested cells were homogenated, centrifuged at 12,000 × g at 4°C for 30 min, and the protein concentration in supernatants was determined using the BCA assay.

A total of 100 μg proteins were separated by SDS-polyacrylamide gel electrophoresis (4% stacking and 10% separating gels) and then transferred overnight onto PVDF membranes. The membranes were blocked with 5% skimmed milk in 0.01 M PBS containing 0.05% (v/v) Tween. Next, they were immunoblotted with rabbit anti-human p-ERK1/2 antibody (cat no. 4370, dilution 1:2,000), rabbit anti-human ERK1/2 (cat no. 4695, di1ution 1:2,000; both from Cell Signaling Technology, Danvers, MA, USA), rabbit anti-human cytochrome c (cat no. ab133504, di1ution 1:1,000; Abcam, USA) for 2 h. Blots were then incubated with a peroxidase-conjugated sheep anti-rabbit IgG (dilution 1:8,000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) for 1 h and developed using chemiluminescent detection with a Supersignal West Pico Assay kit (Thermo, USA) and autoradiography film.

Results

Cat B expression in MGC-803 cells

As shown in Fig. 1, immunocytochemical analysis revealed abundant Cat B expression in the cytoplasm of the MGC-803 cells.

Cytotoxic effects of DOX and PDOX on MGC-803 cells

To investigate the cytotoxic effects of DOX and PDOX, MGC-803 cells were treated with the indicated concentrations of DOX and PDOX for 24, 48, 72 and 96 h, respectively. As confirmed by the MTT assay, DOX and PDOX triggered dose-dependent cytotoxicity and resulted in a significant reduction in cell viability (Fig. 2). At 48 h, the rates of proliferation inhibition following treatment with 0.86, 1.72, 3.44, 6.88 and 13.76 μM of PDOX were 13.17±12.12, 15.10±5.46, 18.33±4.85, 34.99±2.27 and 54.86±0.93%, respectively; and following treatment with the same concentrations of DOX were 32.30±5.47, 34.39±5.81, 41.50±2.58, 52.35±9.25 and 59.97±10.29%, respectively (Fig 2B). At 48 h of treatment, the IC50 concentration of PDOX (14.9 μM) was 3.04 times that of DOX (4.9 μM).

Effects of DOX and PDOX on the cell cycle distribution of MGC-803 cells

To study the effects of DOX and PDOX on the cell cycle, MGC-803 cells were treated with 4.9 and 14.9 μM PDOX or 4.9 and 14.9 μM DOX for 48 h, respectively. The percentages of cells at phases G1, G2 and S were 56.1, 18.7 and 25.5% in the control group; 37.0, 40.2 and 22.8% in the 4.9 μM DOX group; 29.2, 46.6 and 24.2% in the 4.9 μM PDOX group; 33.6, 41.7 and 24.7% in the 14.9 μM DOX group; and 14.4, 55.3 and 30.3% in the 14.9 μM PDOX group (Fig. 3). These results suggest that both PDOX and DOX arrested the cell cycle at the G2/S phase. Moreover, PDOX at IC50 arrested more cells at the G2/S phase compared with DOX at IC50.

Effects of DOX and PDOX on ROS generation of MGC-803 cells

ROS generation of MGC-803 cells was significantly increased by PDOX when compared with that by DOX (Fig. 4). Compared with the control, ROS levels were increased by 1.20-fold (P>0.05), 1.95-fold (P<0.05), 2.09-fold (P<0.05) and 2.15-fold (P<0.05) following treatment of 4.9 μM DOX or PDOX, 14.9 μM DOX or PDOX, respectively. Meanwhile, compared with DOX at IC50, PDOX at IC50 significantly increased ROS generation (1.53-fold, P<0.05).

Effects of DOX and PDOX on the mitochondria in MGC-803 cells

ΔΨm of MGC-803 cells was significantly decreased following treatment with both DOX and PDOX (Fig. 5A). Compared with the control, fluorescence intensity was increased by 1.04-fold (P<0.05), 1.07-fold (P<0.001), 1.08-fold (P<0.01) and 1.10-fold (P<0.001) after treatment with 4.9 μM DOX or PDOX and 14.9 μM DOX or PDOX, respectively. Meanwhile, compared with DOX at IC50, PDOX at IC50 significantly increased fluorescence values (1.53-fold, P<0.01).

Results of transmission electron microscopy revealed that PDOX and DOX induced mitochondrial swelling, particularly in cells treated with PDOX at IC50 (Fig. 5B).

Effects of DOX and PDOX on ERK1/2 phosphorylation and cytoplasmic cytochrome c release in MGC-803 cells

Phosphorylation of ERK1/2 was significantly reduced by both PDOX and DOX treatments, and the effect of PDOX was much more marked. Similarly, cytoplasmic cytochrome c was increased following both PDOX and DOX treatments, and the effect of PDOX was more obvious (Fig. 6).

Discussion

DOX is one of the most commonly used chemotherapeutic drugs with proven efficacy, but is associated with serious side effects. To resolve this issue, the ‘smart’ DOX-prodrug PDOX was designed with increased in vivo anti-metastasis effects and reduced toxicities in gastric cancer peritoneal carcinomatosis (16).

The present study aimed to elucidate the molecular mechanisms of action of PDOX using MGC-803 cells as a model, since immunocytochemical analysis demonstrated that this cell line is rich in Cat B. Our results showed that: i) the direct cytotoxicity of PDOX on cancer cells did not exceed that of DOX since the IC50 of PDOX was significantly higher than that of DOX; ii) PDOX induced a greater degree of mitochondria-centered intrinsic apoptosis than DOX, since more cytochrome c was released from the mitochondria into the cytoplasm following PDOX treatment; iii) PDOX promoted more ROS production and significantly inhibited p-ERK1/2 to a greater degree than DOX. Therefore, treatment with both PDOX and DOX caused mitochondrial swelling and arrested the cell cycle at the G2/S phase; moreover, the effects of PDOX on mitochondria and the cell cycle were more marked than DOX.

Previous studies found that the two N-(2-hydroxypropyl) methacrylamide copolymer-bound DOX-prodrugs (PK1 and HYD) and free DOX greatly differ in regards to their antiproliferative effect and cell death signals in EL-4 cancer cells; treatment with free DOX greatly increased p38 phosphorylation, while PK1 increased it slightly; PK1 also significantly increased ERK phosphorylation, while the free DOX slightly decreased it (27). Based on our study and on the results from the PK1 study, we conclude that antitumor mechanisms of action of a prodrug may be different from the original drug itself. Molecular modifications could bring new active groups, structural changes, drug metabolic alterations, which together may account for the different mechanisms of action from free DOX. This also suggests that the prodrugs may have a different antitumor spectrum. Therefore, more studies are needed to investigate the molecular mechanisms of action and the antitumor spectrum.

In conclusion, the present study found that PDOX has different mechanisms of action, particularly the mitochondria-centered intrinsic pathway involving reactive oxidative stress and the ERK1/2 signaling pathway. The new knowledge gained from this study may aid in the development of PDOX as a ‘smart’ molecular targeting drug against cancer metastasis.

Acknowledgements

The present study was supported by the State Key Research Project on Infectious Diseases (2012ZX10002012-012) and the National Natural Science Foundation of China (no. 81171396) and National University Students Innovation Training Project of China (101048639).

References

1 

Gianni L, Grasselli G, Cresta S, Locatelli A, Viganò L and Minotti G: Anthracyclines. Cancer Chemother Biol Response Modif. 21:29–40. 2003. View Article : Google Scholar

2 

Abu Ajaj K, Graeser R, Fichtner I and Kratz F: In vitro and in vivo study of an albumin-binding prodrug of doxorubicin that is cleaved by cathepsin B. Cancer Chemother Pharmacol. 64:413–418. 2009.PubMed/NCBI

3 

Kratz F, Warnecke A, Schmid B, Chung DE and Gitzel M: Prodrugs of anthracyclines in cancer chemotherapy. Curr Med Chem. 13:477–523. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Seymour LW, Ferry DR, Kerr DJ, Rea D, Whitlock M, Poyner R, et al: Phase II studies of polymer-doxorubicin (PK1, FCE28068) in the treatment of breast, lung and colorectal cancer. Int J Oncol. 34:1629–1636. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Seymour LW, Ferry DR, Anderson D, Hesslewood S, Julyan PJ, Poyner R, et al: Hepatic drug targeting: Phase I evaluation of polymer-bound doxorubicin. J Clin Oncol. 20:1668–1676. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Podgorski I and Sloane BF: Cathepsin B and its role(s) in cancer progression. Biochem Soc Symp. 70:263–276. 2003.PubMed/NCBI

7 

Calderón M, Graeser R, Kratz F and Haag R: Development of enzymatically cleavable prodrugs derived from dendritic polyglycerol. Bioorg Med Chem Lett. 19:3725–3728. 2009.PubMed/NCBI

8 

Kovár M, Strohalm J, Etrych T, Ulbrich K and Ríhová B: Star structure of antibody-targeted HPMA copolymer-bound doxorubicin: a novel type of polymeric conjugate for targeted drug delivery with potent antitumor effect. Bioconjug Chem. 13:206–215. 2002.PubMed/NCBI

9 

Thanou M and Duncan R: Polymer-protein and polymer-drug conjugates in cancer therapy. Curr Opin Investig Drugs. 4:701–709. 2003.PubMed/NCBI

10 

Mai J, Waisman DM and Sloane BF: Cell surface complex of cathepsin B/annexin II tetramer in malignant progression. Biochim Biophys Acta. 1477:215–230. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Kratz F, Müller IA, Ryppa C and Warnecke A: Prodrug strategies in anticancer chemotherapy. Chem Med Chem. 3:20–53. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Vasey PA, Kaye SB, Morrison R, Twelves C, Wilson P, Duncan R, et al: Phase I clinical and pharmacokinetic study of PK1 (N-(2-hydroxypropyl)methacrylamide copolymer doxorubicin): first member of a new class of chemotherapeutic agents-drug-polymer conjugates. Cancer Research Campaign Phase I/II Committee. Clin Cancer Res. 5:83–94. 1999.

13 

Dubowchik GM and Firestone RA: Cathepsin B-sensitive dipeptide prodrugs. 1. A model study of structural requirements for efficient release of doxorubicin. Bioorg Med Chem Lett. 8:3341–3346. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Dubowchik GM, Firestone RA, Padilla L, Willner D, Hofstead SJ, Mosure K, et al: Cathepsin B-labile dipeptide linkers for lysosomal release of doxorubicin from internalizing immunoconjugates: model studies of enzymatic drug release and antigen-specific in vitro anticancer activity. Bioconjug Chem. 13:855–869. 2002. View Article : Google Scholar

15 

Dubowchik GM, Mosure K, Knipe JO and Firestone RA: Cathepsin B-sensitive dipeptide prodrugs. 2. Models of anticancer drugs paclitaxel (Taxol), mitomycin C and doxorubicin. Bioorg Med Chem Lett. 8:3347–3352. 1998. View Article : Google Scholar : PubMed/NCBI

16 

Shao LH, Liu SP, Hou JX, Zhang YH, Peng CW, Zhong YJ, et al: Cathepsin B cleavable novel prodrug Ac-Phe-Lys-PABC-ADM enhances efficacy at reduced toxicity in treating gastric cancer peritoneal carcinomatosis: an experimental study. Cancer. 118:2986–2996. 2012. View Article : Google Scholar

17 

Liu LL, Li QX, Xia L, Li J and Shao L: Differential effects of dihydropyridine calcium antagonists on doxorubicin-induced nephrotoxicity in rats. Toxicology. 231:81–90. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Hancock JT, Desikan R and Neill SJ: Role of reactive oxygen species in cell signalling pathways. Biochem Soc Trans. 29:345–350. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Fruehauf JP and Meyskens FL Jr: Reactive oxygen species: a breath of life or death? Clin Cancer Res. 13:789–794. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Korsmeyer SJ, Yin XM, Oltvai ZN, Veis-Novack DJ and Linette GP: Reactive oxygen species and the regulation of cell death by the Bcl-2 gene family. Biochim Biophys Acta. 1271:63–66. 1995. View Article : Google Scholar : PubMed/NCBI

21 

Gupta S: Molecular signaling in death receptor and mitochondrial pathways of apoptosis (Review). Int J Oncol. 22:15–20. 2003.PubMed/NCBI

22 

Tsang WP, Chau SP, Kong SK, Fung KP and Kwok TT: Reactive oxygen species mediate doxorubicin induced p53-independent apoptosis. Life Sci. 73:2047–2058. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Wang S, Konorev EA, Kotamraju S, Joseph J, Kalivendi S and Kalyanaraman B: Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. intermediacy of H(2)O(2)- and p53-dependent pathways. J Biol Chem. 279:25535–25543. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Moungjaroen J, Nimmannit U, Callery PS, Wang L, Azad N, Lipipun V, et al: Reactive oxygen species mediate caspase activation and apoptosis induced by lipoic acid in human lung epithelial cancer cells through Bcl-2 down-regulation. J Pharmacol Exp Ther. 319:1062–1069. 2006. View Article : Google Scholar

25 

Peng CW, Liu XL, Chen C, Liu X, Yang XQ, Pang DW, et al: Patterns of cancer invasion revealed by QDs-based quantitative multiplexed imaging of tumor microenvironment. Biomaterials. 32:2907–2917. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Shinomol GK and Muralidhara: Effect of Centella asiatica leaf powder on oxidative markers in brain regions of prepubertal mice in vivo and its in vitro efficacy to ameliorate 3-NPA-induced oxidative stress in mitochondria. Phytomedicine. 15:971–984. 2008.

27 

Kovár L, Strohalm J, Chytil P, Mrkvan T, Kovár M, Hovorka O, et al: The same drug but a different mechanism of action: comparison of free doxorubicin with two different N-(2-hydroxypropyl)methacrylamide copolymer-bound doxorubicin conjugates in EL-4 cancer cell line. Bioconjug Chem. 18:894–902. 2007.

Related Articles

Journal Cover

October 2013
Volume 30 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhong YJ, Liu SP, Firestone RA, Hong YP and Li Y: Anticancer effects of Ac‑Phe‑Lys‑PABC‑doxorubicin via mitochondria‑centered apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway in MGC‑803 cells. Oncol Rep 30: 1681-1686, 2013
APA
Zhong, Y., Liu, S., Firestone, R.A., Hong, Y., & Li, Y. (2013). Anticancer effects of Ac‑Phe‑Lys‑PABC‑doxorubicin via mitochondria‑centered apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway in MGC‑803 cells. Oncology Reports, 30, 1681-1686. https://doi.org/10.3892/or.2013.2629
MLA
Zhong, Y., Liu, S., Firestone, R. A., Hong, Y., Li, Y."Anticancer effects of Ac‑Phe‑Lys‑PABC‑doxorubicin via mitochondria‑centered apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway in MGC‑803 cells". Oncology Reports 30.4 (2013): 1681-1686.
Chicago
Zhong, Y., Liu, S., Firestone, R. A., Hong, Y., Li, Y."Anticancer effects of Ac‑Phe‑Lys‑PABC‑doxorubicin via mitochondria‑centered apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway in MGC‑803 cells". Oncology Reports 30, no. 4 (2013): 1681-1686. https://doi.org/10.3892/or.2013.2629