Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways

  • Authors:
    • Ching-Wen Wu
    • Hsiao-Chun Liu
    • Yung-Luen Yu
    • Yu-Ting Hung
    • Chyou-Wei Wei
    • Giou-Teng Yiang
  • View Affiliations

  • Published online on: February 10, 2017     https://doi.org/10.3892/or.2017.5439
  • Pages: 2177-2184
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Methotrexate (MTX) is widely used as both an anticancer and anti-rheumatoid arthritis drug. Although MTX has been used to inhibit the growth of many cancer cells, it cannot effectively inhibit growth of triple-negative breast cancer cells (TNBC cells). Vitamin C is an antioxidant that can prevent oxidative stress. In addition, vitamin C has been applied as adjunct treatment for growth inhibition of cancer cells. Recent studies indicated that combined treatment with vitamin C and MTX may inhibit MCF-7 and MDA-MB-231 breast cancer cell growth through G2/M elongation. However, the mechanisms remain unknown. The aim of the present study was to determine whether combined treatment with low-dose vitamin C and MTX inhibits TNBC cell growth and to investigate the mechanisms of vitamin C/MTX-induced cytotoxicity. Neither low-dose vitamin C alone nor MTX alone inhibited TNBC cell growth. However, combined low-dose vitamin C and MTX had synergistic anti-proliferative/cytotoxic effects on TNBC cells. In addition, co-treatment increased H2O2 levels and activated both caspase-3 and p38 cell death pathways.

Introduction

Approximately 15–20% of breast cancer cells are triple-negative (TNBC cells) (1,2), lacking estrogen receptors (ERs), progesterone receptors (PRs) and epidermal growth factor receptor 2 (EGFR2). Expression of these receptors allows for treatment with endocrine or targeted therapies in clinical cases (35), which are not useful for clinical TNBC cell treatment (68). Therefore, it is important to develop new methods for suppressing TNBC cell growth and survival. Methotrexate (MTX) is a well-known antagonist of folic acid (9,10) and has been used widely for rheumatoid arthritis treatment (11,12). In addition, MTX has been applied for clinical cancer treatment (13,14). Previous studies demonstrated that MTX can inhibit the growth of various cancer cells, including hepatoma, leukemia, lymphoma and gastric cancer cells (1517). Nevertheless, MTX alone is not effective for breast cancer treatment. In order to enhance the anticancer activities of MTX on breast cancer cells, combining MTX with other agents has been considered. Currently, combined chemotherapy with MTX and other anticancer drugs, such as mitomycin C, cyclophosphamide and 5-fluorouracil, is used to treat breast cancer (1820). However, serious side-effects of these chemicals have been reported (2125). Therefore, drugs that can promote the anticancer activities of MTX with reduced side-effects are urgently needed.

Vitamin C, a common nutrient, has anti-oxidative (26,27) and anticancer activities (28,29). Previous studies have also demonstrated that combined treatment with vitamin C and conventional anticancer agents can enhance anticancer activities (15,30,31). Currently, vitamin C supplements are being applied for clinical cancer therapy (3234). However, vitamin C actually inhibits tamoxifen-induced cell death in ER-positive breast cancer (35). Alternatively, high-dose vitamin C alone can inhibit cancer cell growth, though the mechanisms remain elusive (3638). One study also showed that vitamin C can attenuate the incidence of ER-positive breast cancer cells (39). However, there is no evidence demonstrating that vitamin C alone is useful for TNBC treatment.

A recent study reported that vitamin C (30 µM to 4 mM) plus MTX can inhibit the growth of MCF-7 cells (an ER-positive breast cancer cell line) and MDA-MB-231 cells (a type of TNBC) through G2/M elongation and PI3K activation (30). However, the mechanisms of vitamin C/MTX-induced cytotoxicity on breast cancer cells are still unclear. Therefore, whether combined treatment with low-dose vitamin C (5 µM) and MTX can inhibit TNBC cell growth and the mechanisms of vitamin C/MTX-induced cytotoxicity were examined in the present study.

Materials and methods

Materials

Vitamin C, Luminol, Lucigenin and Hoechst 33342 were purchased from Sigma-Aldrich (St. Louis, MO, USA). Anti-tubulin (1:1,000; cat. no. BS1699), anti-p38 (1:400; cat. no. BS3567) and anti-p-p38 (1:400; cat. no. BS4766) primary rabbit polyclonal antibodies were acquired from Bioworld Technology, Inc., (Louis Park, MN, USA). Anti-cleaved PARP (1:2000; cat. no. 9544) and anti-caspase-3 (1:1000; cat. no. 9965) primary rabbit polyclonal antibodies and horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG secondary antibody (1:2,000, cat. no. 7074) were from Cell Signaling Technology (Danvers, MA, USA). Tarceva (Erlotinib) was purchased from Roche Ltd. (Kaiseraugst, Switzerland). An MTT assay kit was obtained from Bio Basic Inc. (Markham, ON, Canada). Fetal bovine serum (FBS), Dulbeccos modified Eagles medium (DMEM), non-essential amino acids, L-glutamine and penicillin/streptomycin were obtained from Gibco-BRL (Invitrogen Life Technologies, Carlsbad, CA, USA).

Cell lines and culture

Triple-negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) were purchased from the Bioresource Collection and Research Center (Hsin-chu, Taiwan). Tarceva-resistant MDA-MB-231 cells (MDA-MB-231 TR) were kindly provided by Dr Yung-Luen Yu (Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan). These cells were maintained in a humidified atmosphere containing 5% CO2 at 37°C and cultured with DMEM supplemented with 10% FBS, 0.1 mM non-essential amino acids, 2 mM L-glutamine and 100 IU/ml penicillin/streptomycin. In addition, 100 µM tarceva was added to the media for MDA-MB-231 TR culture.

Determination of cell viability

Cell viability was measured by the MTT assay described in previous studies (40,41). Briefly, cells were cultured into 96-well plates (5×103 cells/well). Every 24 h, the control and experimental groups were treated with MTT. After incubation for 3 h at 37°C, the formazan product was dissolved and absorbance measured at 570 nm (A570) using a Multiskan™ FC microplate photometer (Molecular Devices, Sunnyvale, CA, USA). The viable cell count (%) was calculated as (A570 experimental group)/(A570 control group) × 100%.

Measurements of intracellular H2O2 and O2−

Intracellular H2O2 and O2 were measured using the lucigenin-amplified chemiluminescence method (40,42). The samples (200 µl) were added to 0.2 mmol/ml of luminol solution (100 µl) for H2O2 measurement or to 0.1 mmol/ml lucigenin solution (500 µl) for O2 measurement. Next, all samples were analyzed using a chemiluminescence analyzing system (CLA-FSI; Tohoku Electronic Industrial, Co., Ltd., Sendai, Japan). The H2O2 and O2 were observed and incubated for 5 min.

Observation of DNA fragmentation and nuclear condensation

Nuclear condensation and DNA fragmentation, cardinal characteristics of apoptotic cells, were observed using Hoechst 33342 nuclear staining (40,41). Control and experimental (MTX and/or vitamin C-treated) cells were incubated in Hoechst 33342 (10 µg/ml) for 5 min. DNA fragmentation and nuclear condensation were observed under an Olympus DP71 fluorescence microscope (excitation, 352 nm; emission, 450 nm; Olympus Corp., Tokyo, Japan).

SDS electrophoresis and western blotting

Cells were lysed in radio-immunoprecipitation assay (RIPA) buffer (cat. no. 20-188; EMD Millipore, Billerica, MA, USA). Proteins were collected from the supernatant layer after centrifugation (16,000 × g; 4°C) for 20 min. The protein concentration was measured using a protein assay kit (cat. no. 23200; Thermo Fischer Scientific, Inc., Waltham, MA, USA). Equal quantities (40 µg) of protein were separated by SDS-PAGE using 13.3% gels (80 volts) and transferred onto polyvinylidene difluoride membranes (EMD Millipore). The membranes were blocked with 5% non-fat milk at room temperature for 2 h then washed with phosphate-buffered saline (PBS). After the incubation with primary antibodies for 4 h, the membranes were washed with PBS and treated with anti-rabbit HRP-conjugated secondary antibodies at room temperature for 1 h. Finally, the immunolabeled proteins were treated with Western Lightning® chemiluminescence Plus reagent (Perkin-Elmer, Inc., Waltham, MA, USA) and observed with a Luminescence Image Analysis system (LAS-4000; FujiFilm Electronic Materials Taiwan, Co., Ltd., Tainan, Taiwan).

Statistical analysis

All data were obtained from four independent experiments and presented as the mean ± SE. Means were compared by Students t-test using Microsoft Excel (http://microsoft-excel-2010.updatestar.com/zh-tw). A P<0.05 was considered statistically significant.

Results

Combined treatment with low-dose vitamin C and MTX effectively inhibits TNBC cell proliferation and viability

We first examined the effects of various concentrations of MTX on TNBC cell (MDA-MB-231) growth and survival. Low-dose MTX alone (0.1 and 0.01 µM) did not inhibit TNBC cell growth after 96-h treatment, and cell viability as measured by MIT assay was maintained at ~75–100% of control from 24 to 96 h (Fig. 1). Only 10 µM MTX reliably inhibited TNBC cell growth at 96 h, with cell viability <50% (Fig. 1). Next, combined treatment with low-dose vitamin C (5 µM) and MTX was examined. As shown in Fig. 2, compared to MTX-treated and vitamin C-treated groups, the MTX plus vitamin C-treated group exhibited significantly lower cell viability at 24 and 48 h. Cell viability was >70% in all MTX-treated and vitamin C-treated groups (Fig. 2), but <40% at 48 h in the 0.1 µM MTX plus vitamin C-treated group and 10 µM MTX plus vitamin C-treated group (Fig. 2). Overall, these data demonstrate that combined treatment with low-dose vitamin C and MTX effectively inhibits TNBC cell proliferation and survival.

Vitamin C enhances MTX-induced intracellular H2O2 accumulation

MTX can increase reactive oxygen species (ROS) accumulation in cells with ensuing cytotoxicity (15,43). In contrast, vitamin C is an anti-oxidant against ROS increase (26,27). Both H2O2 and O2 are major ROS species in cells. Intracellular H2O2 and O2 were compared among the control group, MTX-treated group, vitamin C-treated group, and MTX plus vitamin C-treated group (Fig. 3A). Intracellular H2O2 levels were increased in 0.1 and 10 µM MTX groups, in accordance with a previous study (15). Surprisingly, vitamin C did not reduce H2O2 levels in MTX-treated groups. Compared to MTX-treated groups, H2O2 levels were increased significantly in the vitamin C plus MTX-treated group. In contrast, O2 levels did not differ among treatment groups (Fig. 3B). Our data suggest that increased intracellular H2O2 may contribute to the decrease in cell viability induced by MTX plus vitamin co-treatment.

MTX-treatment and combined MTX plus vitamin C treatment induce apoptosis and caspase-3 activation

Induction of apoptosis by these treatments was assessed by nuclear staining. As shown in Fig. 4, nuclear condensation and DNA fragmentation were observed in MTX-treated and MTX plus vitamin C-treated groups. Apoptosis can be initiated by caspase-dependent and caspase-independent pathways (44,45). Therefore, caspase-3 activation was examined by western blotting. As shown in Fig. 5, compared to the control group, the ratio of cleaved (activated) caspase-3 to native caspase-3 was increased significantly in both the MTX-treated and MTX plus vitamin C-treated group. PARP is a downstream substrate of caspase-3, thus, cleaved PARP is a sign of caspase-3 activation. Indeed, cleaved PARP level was also increased in both MTX-treated and MTX plus vitamin C-treated groups. Taken together, these results indicate that combined MTX/vitamin C induces caspase-3-dependent apoptosis in TNBC cells.

p38 phosphorylation in MTX-treated and MTX plus vitamin C-treated cells

The MAPK family kinases ERK, JNK and p38 are involved in cell death, cell differentiation, and cell proliferation (4648). In the present study, expression levels of EKR, JNK, p38 and their phosphorylated (activated) forms (p-ERK, p-JNK and p-p38) were estimated by western blotting. The ratio of p-p38 to p38 was significantly increased in both MTX-treated and MTX plus vitamin C-treated groups (Fig. 7), while ERK and JNK expression levels did not differ significantly among groups (data not show). Thus, MTX/vitamin C-induced cytotoxicity of TNBC cells is associated with p38 activation.

Combined treatment with vitamin C and MTX inhibits growth of tarceva-sensitive, but not tarceva-resistant TNBC cells

As shown in Fig. 2, combined treatment with vitamin C and MTX effectively inhibited MDA-MB-231 cell growth. However, it is unclear whether MTX plus vitamin C is useful against other TNBC cells, thus, we examined the cell viability of TNBC cell lines MDA-MB-468 and MDA-MB-231 TR during MTX, vitamin C and combined treatment. Compared to the 10 µM MTX-treated group, the combined treatment group exhibited lower viability at 48, 72 and 96 h (Fig. 8B). Importantly, cell viability was <50% at 72 and 96 h in the 10 µM MTX plus vitamin C-treated group (Fig. 8B). Alternatively, cell viability did not differ among the control, 0.1 µM MTX-treated, and 0.1 µM MTX plus vitamin C-treated groups (Fig. 8A). These data indicate that 1 µM MTX plus vitamin C can effectively inhibit MDA-MB-468 proliferation/survival compared to MTX treatment. However, in similar assays of tarceva-resistant MDA-MB-231 TR cells, the MTX plus vitamin C-treated group exhibited significantly lower cell viability only at 96 h and was >60% for all other groups (Fig. 9). That is, neither MTX alone nor MTX plus vitamin C inhibited the growth of tarceva-resistant TNBC cells as effectively as tarceva-sensitive TNBC cells. Taken together, these findings (Figs. 2, 8 and 9) suggest that MTX plus vitamin C treatment can inhibit the growth of tarceva-responsive, but not tarceva-resistant TNBC cells.

Discussion

MTX alone is not useful for breast cancer treatment, but a recent study found that high-dose vitamin C (30 µM to 4 mM) enhanced the anticancer activities of MTX on breast cancer cells, including MCF-7 cells (ER-positive breast cancer) and MDA-MB-231 cells (30). That study also found G2/M elongation and PI3K pathway activation associated with vitamin C/MTX-induced cytotoxicity. The present study found that MTX had anti-proliferative/cytotoxic effects on TNBC cells only when combined with low-dose vitamin C (5 µM). The present study further suggests that increased intracellular H2O2 levels and activation of caspase-3 and p38 pathways are involved in vitamin C/MTX-induced cytotoxicity. In addition, combined treatment with low-dose vitamin C and MTX inhibited cell growth not only of MDA-MB-231 cells but also of MDA-MB-468 cells. Therefore, this study suggests that vitamin C plus MTX treatment may be effective for clinical suppression of TNBC cell growth.

Although a previous study (30) and the present study (Figs. 2 and 8) demonstrated that vitamin C plus MTX effectively inhibits TNBC cell growth, vitamin C plus MTX treatment did not effectively inhibit tarceva-resistant TNBC cells (Fig. 9). Tarceva (erlotinib) is an EGFR tyrosine kinase inhibitor (49,50) and has been applied for clinical treatment of lung and breast cancers (5154). These findings suggest that EGFR signaling may be involved in vitamin C/MTX-induced cytotoxicity.

Apoptosis can be induced via caspase-dependent and caspase-independent pathways (44,45). Vitamin C treatment can induce apoptosis of breast cancer cells and lung cancer cells via the caspase-independent pathway (36,55,56). On the other hand, vitamin C treatment can induce apoptosis of melanoma cells and hepatoma cells via the caspase-dependent pathway (15,57). In the present study, vitamin C plus MTX treatment activated caspase-3 in TNBC cells. Thus, whether apoptosis occurs via caspase-dependent or caspase-independent pathways may depend on the specific cancer cell type.

At low doses, vitamin C has anti-oxidant activities (26,27), and many studies have demonstrated that vitamin C supplements can decrease oxidative stress (15,58,59). However, high-dose (millimolar) vitamin C treatment can increase oxidative stress (6062), and previous studies showed that high-dose vitamin C can enhance intracellular H2O2, resulting in cancer cell death (37,63,64). Surprisingly, another study reported that low-dose (micromolar) vitamin C attenuated H2O2 levels but enhanced the anticancer activities of MTX on hepatoma cells (15). In addition, vitamin C/MTX only enhances H2O2 levels but not O2 levels in TNBC cells. These results indicated vitamin C/MTX does not inhibit the function of superoxide dismutase, while vitamin C/MTX may influence GSH levels or activity of glutathione peroxidase.

As shown in Fig. 9, the cell viability was not significantly different before 72 h between MTX alone group and MTX plus vitamin C group. The cell viability was significant different only at 96 h between MTX alone group and MTX plus vitamin C group. The results indicated that vitamin C is not efficient in enhancing MTX-induced cytotoxicity in tarceva-resistant TNBC cells. Previously, many studies showed that tarceva-resistant cells have EGFR gene mutations or additional bypass signaling pathways to activate downstream of EGFR (65,66). These mutation genes and bypass pathways are important factors for cell proliferation with EGFR inhibitor treatment. These factors may cause the vitamin C/MTX inefficiency in inducing cytotoxicity in tarceva-resistant cells. However, many bypass signals are related to tarceva-resistant cells, such as SOS1, NF-κB and Fas receptor, signals of which influence vitamin C/MTX-induced cytotoxicity, however, this remains to be studied in the future.

Collectively, we found that combined treatment with low-dose vitamin C and MTX enhanced intracellular H2O2 accumulation and suppressed TNBC cell growth. Therefore, we suggest that both vitamin C dose and cell type may influence cellular H2O2 levels during treatment.

Acknowledgements

The present study was supported by funding from the Ministry of Science and Technology, Taiwan (MOST103 2320-B-039-052-MY3; MOST104-2321-B-039-005), the Ministry of Health and Welfare, Taiwan (MOHW104-TDU-B-212-124-002) and the Taipei Tzu ChiHospital, Taiwan (TCRD-TPE-104-06; TCRD-TPE-104-34; TCRD-TPE-105-20; TCRD-TPE-105-02).

References

1 

Yu YL, Chou RH, Liang JH, Chang WJ, Su KJ, Tseng YJ, Huang WC, Wang SC and Hung MC: Targeting the EGFR/PCNA signaling suppresses tumor growth of triple-negative breast cancer cells with cell-penetrating PCNA peptides. PLoS One. 8:e613622013. View Article : Google Scholar : PubMed/NCBI

2 

Metzger-Filho O, Tutt A, de Azambuja E, Saini KS, Viale G, Loi S, Bradbury I, Bliss JM, Azim HA Jr, et al: Dissecting the heterogeneity of triple-negative breast cancer. J Clin Oncol. 30:1879–1787. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Ojo D, Wei F, Liu Y, Wang E, Zhang H, Lin X, Wong N, Bane A and Tang D: Factors promoting tamoxifen resistance in breast cancer via stimulating breast cancer stem cell expansion. Curr Med Chem. 22:2360–2374. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Spring L, Bardia A and Modi S: Targeting the cyclin D-cyclin-dependent kinase (CDK) 4/6-retinoblastoma pathway with selective CDK 4/6 inhibitors in hormone receptor-positive breast cancer: Rationale, current status, and future directions. Discov Med. 21:65–74. 2016.PubMed/NCBI

5 

Lumachi F, Chiara GB, Foltran L and Basso SM: Proteomics as a guide for personalized adjuvant chemotherapy in patients with early breast cancer. Cancer Genomics Proteomics. 12:385–390. 2015.PubMed/NCBI

6 

Williams CB, Soloff AC, Ethier SP and Yeh ES: Perspectives on epidermal growth factor receptor regulation in triple-negative breast cancer: Ligand-mediated mechanisms of receptor regulation and potential for clinical targeting. Adv Cancer Res. 127:253–281. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Hudis CA and Gianni L: Triple-negative breast cancer: An unmet medical need. Oncologist. 16:(Suppl 1). 1–11. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Schofield RC, Ramanathan LV, Murata K, Fleisher M, Pessin MS and Carlow DC: Development of an assay for methotrexate and its metabolites 7-hydroxy methotrexate and DAMPA in serum by LC-MS/MS. Methods Mol Biol. 1383:213–222. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Hara A, Taguchi A, Aoki H, Hatano Y, Niwa M, Yamada Y and Kunisada T: Folate antagonist, methotrexate induces neuronal differentiation of human embryonic stem cells transplanted into nude mouse retina. Neurosci Lett. 477:138–143. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Pavy S, Constantin A, Pham T, Gossec L, Maillefert JF, Cantagrel A, Combe B, Flipo RM, Goupille P, Le Loët X, et al: Methotrexate therapy for rheumatoid arthritis: clinical practice guidelines based on published evidence and expert opinion. Joint Bone Spine. 73:388–395. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Whittle SL and Hughes RA: Folate supplementation and methotrexate treatment in rheumatoid arthritis: A review. Rheumatology (Oxford). 43:267–271. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Cho KM, Kim YJ, Kim SH, Kim JW, Lee JO, Han JH, Lee KW, Kim JH, Kim CY, Bang SM, et al: Salvage treatment with intracerebrospinal fluid thiotepa in patients with leptomeningeal metastasis after failure of methotrexate-based treatment. Anticancer Res. 35:5631–5638. 2015.PubMed/NCBI

14 

van der Plas E, Nieman BJ, Butcher DT, Hitzler JK, Weksberg R, Ito S and Schachar R: Neurocognitive late effects of chemotherapy in survivors of acute lymphoblastic leukemia: Focus on methotrexate. J Can Acad Child Adolesc Psychiatry. 24:25–32. 2015.PubMed/NCBI

15 

Yiang GT, Chou PL, Hung YT, Chen JN, Chang WJ, Yu YL and Wei CW: Vitamin C enhances anticancer activity in methotrexate-treated Hep3B hepatocellular carcinoma cells. Oncol Rep. 32:1057–1063. 2014.PubMed/NCBI

16 

Shirao K, Boku N, Yamada Y, Yamaguchi K, Doi T, Goto M, Nasu J, Denda T, Hamamoto Y, Takashima A, et al: Gastrointestinal Oncology Study Group of the Japan Clinical Oncology Group: Randomized Phase III study of 5-fluorouracil continuous infusion vs. sequential methotrexate and 5-fluorouracil therapy in far advanced gastric cancer with peritoneal metastasis (JCOG0106). Jpn J Clin Oncol. 43:972–980. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Takemura Y and Jackman AL: Folate-based thymidylate synthase inhibitors in cancer chemotherapy. Anticancer Drugs. 8:3–16. 1997. View Article : Google Scholar : PubMed/NCBI

18 

Fukuda T, Tanabe M, Kobayashi K, Fukada I, Takahashi S, Iwase T and Ito Y: Combination chemotherapy with mitomycin C and methotrexate is active against metastatic HER2-negative breast cancer even after treatment with anthracycline, taxane, capecitabine, and vinorelbine. Springerplus. 4:3762015. View Article : Google Scholar : PubMed/NCBI

19 

Leone JP, Leone J, Vallejo CT, Pérez JE, Romero AO, Machiavelli MR, Romero Acuña L, Domínguez ME, Langui M, Fasce HM, et al: Sixteen years follow-up results of a randomized phase II trial of neoadjuvant fluorouracil, doxorubicin, and cyclophosphamide (FAC) compared with cyclophosphamide, methotrexate, and 5-fluorouracil (CMF) in stage III breast cancer: GOCS experience. Breast Cancer Res Treat. 143:313–323. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Wu CE, Chen SC, Lin YC, Lo YF, Hsueh S and Chang HK: Identification of patients with node-negative, triple-negative breast cancer who benefit from adjuvant cyclophosphamide, methotrexate, and 5-fluorouracil chemotherapy. Anticancer Res. 34:1301–1306. 2014.PubMed/NCBI

21 

Shea B, Swinden MV, Ghogomu ET, Ortiz Z, Katchamart W, Rader T, Bombardier C, Wells GA and Tugwell P: Folic acid and folinic acid for reducing side effects in patients receiving methotrexate for rheumatoid arthritis. J Rheumatol. 41:1049–1060. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Lee HJ, Hong SK, Seo JK, Lee D and Sung HS: A case of cutaneous side effect of methotrexate mimicking Behçets disease. Ann Dermatol. 23:412–414. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Colomer Gallardo A, Martínez Rodríguez R, Castillo Pacheco C, González Satue C and Ibarz Servio L: Dermatological side effects of intravesical mitomycin C: Delayed hypersensitivity. Arch Esp Urol. 69:89–91. 2016.PubMed/NCBI

24 

Elazzazy S, Mohamed AE and Gulied A: Cyclophosphamide-induced symptomatic hyponatremia, a rare but severe side effect: A case report. Onco Targets Ther. 7:1641–1645. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Sun W, Yan C, Jia S and Hu J: Correlation analysis of peripheral DPYD gene polymorphism with 5-fluorouracil susceptibility and side effects in colon cancer patients. Int J Clin Exp Med. 7:5857–5861. 2014.PubMed/NCBI

26 

Rietjens IM, Boersma MG, Haan L, Spenkelink B, Awad HM, Cnubben NH, van Zanden JJ, Woude H, Alink GM and Koeman JH: The pro-oxidant chemistry of the natural antioxidants vitamin C, vitamin E, carotenoids and flavonoids. Environ Toxicol Pharmacol. 11:321–333. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Mason SA, Gatta PA Della, Snow RJ, Russell AP and Wadley GD: Ascorbic acid supplementation improves skeletal muscle oxidative stress and insulin sensitivity in people with type 2 diabetes: Findings of a randomized controlled study. Free Radic Biol Med. 93:227–238. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Lee WJ: The prospects of vitamin C in cancer therapy. Immune Netw. 9:147–152. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Nagappan A, Park KI, Park HS, Kim JA, Hong GE, Kang SR, Lee DH, Kim EH, Lee WS, Won CK, et al: Vitamin C induces apoptosis in AGS cells by down-regulation of 14-3-3σ via a mitochondrial dependent pathway. Food Chem. 135:1920–1928. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Guerriero E, Sorice A, Capone F, Napolitano V, Colonna G, Storti G, Castello G and Costantini S: Vitamin C effect on mitoxantrone-induced cytotoxicity in human breast cancer cell lines. PLoS One. 9:e1152872014. View Article : Google Scholar : PubMed/NCBI

31 

Vetvicka V and Vetvickova J: Combination of glucan, resveratrol and vitamin C demonstrates strong anti-tumor potential. Anticancer Res. 32:81–87. 2012.PubMed/NCBI

32 

Nagao T, Warnakulasuriya S, Nakamura T, Kato S, Yamamoto K, Fukano H, Suzuki K, Shimozato K and Hashimoto S: Treatment of oral leukoplakia with a low-dose of beta-carotene and vitamin C supplements: A randomized controlled trial. Int J Cancer. 136:1708–1717. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Tokarski S, Rutkowski M, Godala M, Mejer A and Kowalski J: The impact of ascorbic acid on the concentrations of antioxidative vitamins in the plasma of patients with non-small cell lung cancer undergoing first-line chemotherapy. Pol Merkur Lekarski. 35:136–140. 2013.(In Polish). PubMed/NCBI

34 

Nechuta S, Lu W, Chen Z, Zheng Y, Gu K, Cai H, Zheng W and Shu XO: Vitamin supplement use during breast cancer treatment and survival: A prospective cohort study. Cancer Epidemiol Biomarkers Prev. 20:262–271. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Subramani T, Yeap SK, Ho WY, Ho CL, Omar AR, Aziz SA, Rahman NM and Alitheen NB: Vitamin C suppresses cell death in MCF-7 human breast cancer cells induced by tamoxifen. J Cell Mol Med. 18:305–313. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Hong SW, Jin DH, Hahm ES, Yim SH, Lim JS, Kim KI, Yang Y, Lee SS, Kang JS, Lee WJ, et al: Ascorbate (vitamin C) induces cell death through the apoptosis-inducing factor in human breast cancer cells. Oncol Rep. 18:811–815. 2007.PubMed/NCBI

37 

Uetaki M, Tabata S, Nakasuka F, Soga T and Tomita M: Metabolomic alterations in human cancer cells by vitamin C-induced oxidative stress. Sci Rep. 5:138962015. View Article : Google Scholar : PubMed/NCBI

38 

van der Reest J and Gottlieb E: Anti-cancer effects of vitamin C revisited. Cell Res. 26:269–270. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Mense SM, Singh B, Remotti F, Liu X and Bhat HK: Vitamin C and alpha-naphthoflavone prevent estrogen-induced mammary tumors and decrease oxidative stress in female ACI rats. Carcinogenesis. 30:1202–1208. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Yiang GT, Yu YL, Lin KT, Chen JN, Chang WJ and Wei CW: Acetaminophen induces JNK/p38 signaling and activates the caspase-9-3-dependent cell death pathway in human mesenchymal stem cells. Int J Mol Med. 36:485–492. 2015.PubMed/NCBI

41 

Yu YL, Yiang GT, Chou PL, Tseng HH, Wu TK, Hung YT, Lin PS, Lin SY, Liu HC, Chang WJ, et al: Dual role of acetaminophen in promoting hepatoma cell apoptosis and kidney fibroblast proliferation. Mol Med Rep. 9:2077–2084. 2014.PubMed/NCBI

42 

Lin BR, Yu CJ, Chen WC, Lee HS, Chang HM, Lee YC, Chien CT and Chen CF: Green tea extract supplement reduces D-galactosamine-induced acute liver injury by inhibition of apoptotic and proinflammatory signaling. J Biomed Sci. 16:352009. View Article : Google Scholar : PubMed/NCBI

43 

Kolli VK, Abraham P, Isaac B and Selvakumar D: Neutrophil infiltration and oxidative stress may play a critical role in methotrexate-induced renal damage. Chemotherapy. 55:83–90. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Ohgidani M, Komizu Y, Goto K and Ueoka R: Residual powders from Shochu distillation remnants induce apoptosis in human hepatoma cells via the caspase-independent pathway. J Biosci Bioeng. 114:104–109. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Yu VW and Ho WS: Tetrandrine inhibits hepatocellular carcinoma cell growth through the caspase pathway and G2/M phase. Oncol Rep. 29:2205–2210. 2013.PubMed/NCBI

46 

Wu Y, van der Schaft DW, Baaijens FP and Oomens CW: Cell death induced by mechanical compression on engineered muscle results from a gradual physiological mechanism. J Biomech. 49:1071–1077. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Chen J, Li Y, Hao H, Li C, Du Y, Hu Y, Li J, Liang Z, Li C, Liu J, et al: Mesenchymal stem cell conditioned medium promotes proliferation and migration of alveolar epithelial cells under Septic conditions in vitro via the JNK-P38 signaling pathway. Cell Physiol Biochem. 37:1830–1846. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Lee K, Chung YH, Ahn H, Kim H, Rho J and Jeong D: Selective regulation of MAPK signaling mediates RANKL-dependent osteoclast differentiation. Int J Biol Sci. 12:235–245. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Tan CS, Gilligan D and Pacey S: Treatment approaches for EGFR-inhibitor-resistant patients with non-small-cell lung cancer. Lancet Oncol. 16:e447–e459. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Sato F, Kubota Y, Natsuizaka M, Maehara O, Hatanaka Y, Marukawa K, Terashita K, Suda G, Ohnishi S, Shimizu Y, et al: EGFR inhibitors prevent induction of cancer stem-like cells in esophageal squamous cell carcinoma by suppressing epithelial-mesenchymal transition. Cancer Biol Ther. 16:933–940. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Li X, Qin N, Wang J, Yang X, Zhang X, Lv J, Wu Y, Zhang H, Nong J and Zhang Q: Clinical observation of icotinib hydrochloride for advanced non-small cell lung cancer patients with EGFR status identified. Zhongguo Fei Ai Za Zhi. 18:734–739. 2015.(In Chinese). PubMed/NCBI

52 

Osarogiagbon RU, Cappuzzo F, Ciuleanu T, Leon L and Klughammer B: Erlotinib therapy after initial platinum doublet therapy in patients with EGFR wild type non-small cell lung cancer: Results of a combined patient-level analysis of the NCIC CTG BR.21 and SATURN trials. Transl Lung Cancer Res. 4:465–474. 2015.PubMed/NCBI

53 

Montagna E, Cancello G, Bagnardi V, Pastrello D, Dellapasqua S, Perri G, Viale G, Veronesi P, Luini A, Intra M, et al: Metronomic chemotherapy combined with bevacizumab and erlotinib in patients with metastatic HER2-negative breast cancer: Clinical and biological activity. Clin Breast Cancer. 12:207–214. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Tolcher AW, LoRusso P, Arzt J, Busman TA, Lian G, Rudersdorf NS, Vanderwal CA, Kirschbrown W, Holen KD and Rosen LS: Safety, efficacy, and pharmacokinetics of navitoclax (ABT-263) in combination with erlotinib in patients with advanced solid tumors. Cancer Chemother Pharmacol. 76:1025–1032. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Evans MK, Tovmasyan A, Batinic-Haberle I and Devi GR: Mn porphyrin in combination with ascorbate acts as a pro-oxidant and mediates caspase-independent cancer cell death. Free Radic Biol Med. 68:302–314. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Ohtani S, Iwamaru A, Deng W, Ueda K, Wu G, Jayachandran G, Kondo S, Atkinson EN, Minna JD, Roth JA, et al: Tumor suppressor 101F6 and ascorbate synergistically and selectively inhibit non-small cell lung cancer growth by caspase-independent apoptosis and autophagy. Cancer Res. 67:6293–6303. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Lin SY, Lai WW, Chou CC, Kuo HM, Li TM, Chung JG and Yang JH: Sodium ascorbate inhibits growth via the induction of cell cycle arrest and apoptosis in human malignant melanoma A375.S2 cells. Melanoma Res. 16:509–519. 2006. View Article : Google Scholar : PubMed/NCBI

58 

Nagao N, Nakayama T, Etoh T, Saiki I and Miwa N: Tumor invasion is inhibited by phosphorylated ascorbate via enrichment of intracellular vitamin C and decreasing of oxidative stress. J Cancer Res Clin Oncol. 126:511–518. 2000. View Article : Google Scholar : PubMed/NCBI

59 

Liu JW, Nagao N, Kageyama K and Miwa N: Antimetastatic and anti-invasive ability of phospho-ascorbyl palmitate through intracellular ascorbate enrichment and the resultant antioxidant action. Oncol Res. 11:479–487. 1999.PubMed/NCBI

60 

Pires AS, Marques CR, Encarnação JC, Abrantes AM, Mamede AC, Laranjo M, Gonçalves AC, Sarmento-Ribeiro AB and Botelho MF: Ascorbic acid and colon cancer: An oxidative stimulus to cell death depending on cell profile. Eur J Cell Biol. 95:208–218. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Beck R, Pedrosa RC, Dejeans N, Glorieux C, Levêque P, Gallez B, Taper H, Eeckhoudt S, Knoops L, Calderon PB, et al: Ascorbate/menadione-induced oxidative stress kills cancer cells that express normal or mutated forms of the oncogenic protein Bcr-Abl. An in vitro and in vivo mechanistic study. Invest New Drugs. 29:891–900. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Venturelli S, Sinnberg TW, Berger A, Noor S, Levesque MP, Böcker A, Niessner H, Lauer UM, Bitzer M, Garbe C, et al: Epigenetic impacts of ascorbate on human metastatic melanoma cells. Front Oncol. 4:2272014. View Article : Google Scholar : PubMed/NCBI

63 

Ma Y, Chapman J, Levine M, Polireddy K, Drisko J and Chen Q: High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Sci Transl Med. 6:222ra182014. View Article : Google Scholar : PubMed/NCBI

64 

Cieslak JA and Cullen JJ: Treatment of pancreatic cancer with pharmacological ascorbate. Curr Pharm Biotechnol. 16:759–770. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Presutti D, Santini S, Cardinali B, Papoff G, Lalli C, Samperna S, Fustaino V, Giannini G and Ruberti G: MET gene amplification and MET receptor activation are not sufficient to predict efficacy of combined MET and EGFR inhibitors in EGFR TKI-resistant NSCLC cells. PLoS One. 10:e01433332015. View Article : Google Scholar : PubMed/NCBI

66 

De S, Dermawan JK and Stark GR: EGF receptor uses SOS1 to drive constitutive activation of NF-κB in cancer cells. Proc Natl Acad Sci USA. 111:11721–11726. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2017
Volume 37 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
Wu C, Liu H, Yu Y, Hung Y, Wei C and Yiang G: Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways. Oncol Rep 37: 2177-2184, 2017
APA
Wu, C., Liu, H., Yu, Y., Hung, Y., Wei, C., & Yiang, G. (2017). Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways. Oncology Reports, 37, 2177-2184. https://doi.org/10.3892/or.2017.5439
MLA
Wu, C., Liu, H., Yu, Y., Hung, Y., Wei, C., Yiang, G."Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways". Oncology Reports 37.4 (2017): 2177-2184.
Chicago
Wu, C., Liu, H., Yu, Y., Hung, Y., Wei, C., Yiang, G."Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways". Oncology Reports 37, no. 4 (2017): 2177-2184. https://doi.org/10.3892/or.2017.5439