Amlodipine inhibits proliferation, invasion, and colony formation of breast cancer cells

  • Authors:
    • Mohammad A.Y. Alqudah
    • Raneem Al‑Samman
    • Marwah Azaizeh
    • Karem H. Alzoubi
  • View Affiliations

  • Published online on: May 6, 2022     https://doi.org/10.3892/br.2022.1533
  • Article Number: 50
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Calcium channel upregulation has been implicated in cancer cell proliferation and progression including in breast cancer. Fortunately, the function of calcium channels can be manipulated pharmacologically using calcium channel blockers (CCBs). Amlodipine, a dihydropyridine CCB, has been demonstrated to exert cytotoxic effects in several types of cancers. The present study evaluated the effects of amlodipine on proliferation, caspase activation, colony formation, and invasion of human breast cancer cells. Cell viability was assessed using a colorimetric MTT assay. An Apo‑ONE® caspase‑3/7 assay was used to measure caspase‑3/7 levels. Cell invasion was evaluated using Matrigel invasion chambers. The expression of phospho‑(p‑)ERK1/2, Bcl‑2, and integrin β1 proteins were analyzed using western blotting. A one‑way ANOVA with a post‑hoc Tukey's multiple comparison tests was used for statistical analysis. Amlodipine significantly inhibited the growth of both MDA‑MB‑231 and MCF‑7 human breast cancer cells in a dose‑dependent manner and inhibited colony formation of MCF‑7 cells, and this was accompanied by the downregulation of p‑ERK1/2 in MDA‑MB‑231 cells. In addition, treatment with amlodipine resulted in increased caspase‑3/7 levels in MDA‑MB‑231 cells, which was accompanied by the downregulation of the anti‑apoptotic protein, Bcl‑2. Moreover, amlodipine impaired the invasive abilities of MDA‑MB‑231 cells, and integrin β1 expression was concurrently downregulated. The present study illustrates the anticancer effects of amlodipine on breast cancer proliferation, colony formation, and invasion in vitro and highlights the potential value of amlodipine as an anticancer agent.

Introduction

Globally, breast cancer is the most prevalent cancer, accounting for 11.7% of all cancer cases and 6.9% of cancer deaths (1). It has been estimated that 284,200 new breast cancer cases were diagnosed in 2021 in the US (2). In Jordan, 2,403 new breast cancer cases were diagnosed last year accounting for 38.5% of all cases of cancer in women (3). Tumor metastasis is a major cause of breast cancer mortality (4). This multi-step process encompasses local tumor invasion, migration of primary cells, and colonization at distal sites (5). Metastatic breast cancer (MBC) represents 6% of newly diagnosed breast cancer cases. However, 20-30% of early-stage breast cancer cases eventually develop into MBC (6). Systemic therapies are the primary treatment options for MBC including chemotherapy, endocrine therapy, and targeted therapy (7). Although combination chemotherapy is commonly used in MBC, it has been associated with increased toxicity (8,9). Furthermore, the emergence of chemotherapeutic resistance limits the effectiveness of breast cancer treatments, thereby increasing disease relapse and death (10). Therefore, the identification of novel strategies targeting the primary tumor with enhanced efficacy and reduced toxicity are needed to improve patient outcomes.

Calcium channel blockers (CCB) have been implicated as anti-cancer molecules in several types of human cancers. For example, amlodipine, a dihydropyridine CCB, has been shown to induce apoptosis, resulting in cell cycle arrest, and suppress the proliferation of cancerous cells in several studies (11-13). In addition, Ji et al (14) showed that p-glycoprotein-mediated multidrug resistance could be ameliorated in leukemic cells when an amlodipine derivative was used, which in turn prevented doxorubicin efflux and thus enhanced its efficacy. Moreover, in vitro and in vivo studies on human epidermoid cancerous cells have shown that several CCBs can inhibit cancer cell growth including amlodipine, nicardipine, and nimodipine (15). However, the exact cellular and molecular anticancer mechanisms of amlodipine have not been studied in breast cancer cells. In the present study, the effects of amlodipine treatment on breast cancer cell proliferation, apoptosis, colony formation, and invasion were evaluated, and the protein expression levels of the downstream targets were determined as well.

Materials and methods

Cell culture and drug treatment

Triple-negative MDA-MB-231 and luminal MCF-7 breast cell lines were purchased from the American Type Culture Collection (ATCC). Cells were cultured in RPMI-1640 media supplemented with 10% FBS and 1% penicillin/streptomycin in a humidified incubator with 5% CO2 at 37˚C. DMSO was used as a solvent to prepare amlodipine stocks (Tocris Bioscience), with a final DMSO concentration of <0.1% in all experiments.

Cell viability assay

To evaluate the effects of amlodipine on breast cancer cell viability, the colorimetric MTT cell proliferation assay (ATCC) was performed as described previously (16). Briefly, 1x104 cells/well were cultured in a 96-well plate and incubated overnight. Then, cells were treated with several concentrations of amlodipine or with DMSO as a control. After 48 h of treatment, cells were incubated at 37˚C for 4 h with MTT solution at a final concentration of 500 µg/ml. To solubilize formazan crystals, 100 µl DMSO was added to each well. The optical density was measured at 490 nm on a microplate reader (BioTek Instruments, Inc.). The results are expressed as a percentage of viable cells normalized to vehicle-treated cells using the following equations: % Of viable cells in each well=(Absorbance treatment/Average of Absorbance vehicle in 4 replicates)x100; and % of viable cells for each treatment concentration=Average of normalized % of viable cells in 4 treatment replicates.

Caspase-3/7 assay

An Apo-ONE® homogeneous caspase-3/7 assay (Promega Corporation) was used to assess the effects of amlodipine on the induction of caspase-3/7 activities in MDA-MB-231 cells as described previously (17). Briefly, cells were plated at a density of 1x104 cells/well in a 96-well black plate. After attachment, cells were treated with several concentrations of amlodipine or with DMSO as a control. After 48 h of treatment, the caspase-3/7 reagent was added to each well in a 1:1 ratio with the sample volume at room temperature. After 3 h of incubation, enzyme activity was analyzed using a synergy 2 multi-mode microplate reader (Biotek Instruments, Inc.) at excitation and emission wavelengths of 499 nm and 521 nm, respectively.

Colony formation assay

Assessing anchorage-dependent growth of breast cancer cells was performed using colony formation assays as previously described (18,19). MCF-7 cells were plated at a low density (2x103 cells/flask) in T25 flasks and incubated for 24 h, then treated with amlodipine or DMSO as a control. Culture media was replaced every 3 days. Following 3 weeks of incubation, PBS was used to wash the cells before fixing them with pre-cooled (1:1) methanol/acetone at -20˚C for 15 min. After staining with 0.1% crystal violet for 5 min at room temperature, the colonies that had formed were visualized using a light microscope (x4 magnification).

Invasion assay

Invasion assays were performed using Corning BioCoat Matrigel Invasion Chambers (Corning Inc.) as described previously (20,21). Cells were resuspended in serum-free media with various concentrations of amlodipine (0, 5 or 10 µM), and a chemotactic serum gradient was generated by placing media supplemented with 10% FBS in the bottom chambers. After 24 h, the invading cells were fixed with ice-cold ethanol at -20˚C for 15 min, stained with 0.1% crystal violet for 5 min at room temperature, visualized using a light microscope (x4 magnification) and counted using ImageJ (version 1.53q; National Institutes of Health). The results are expressed as a percentage of invading cells in treatment groups relative to the control group.

Western blotting

To assess the effects of amlodipine on the protein expression levels of downstream targets, western blotting was performed as previously described (21). Briefly, cells were plated at a density of 5x104 cells/well into a 6-well plate. The following day, the cells were treated with amlodipine (1-25 µM) or DMSO as a control for 48 h. After cell washing with ice-cold PBS, cells were lysed in RIPA buffer (Thermo Fisher Scientific, Inc.) containing protease inhibitor (150 µl/well for 30 min on ice). Cell lysates were transferred into Eppendorf tubes and then centrifuged at 13,000 x g for 5 min at 4˚C. Proteins were quantified in all collected supernatants using a BCA assay. Protein samples were loaded in equal amounts (35 µg per lane) with one lane for 10 µl of the protein ladder into 10% polyacrylamide gels in tris-glycine buffer and run at 200 mv for 40 min at room temperature. After the proteins had been resolved, they were transferred to nitrocellulose membranes, incubated with primary antibodies (all 1:1,000 dilution) for 2 h at room temperature against phospho (p-)ERK1/2 (Cell Signaling Technology, Inc.; cat. no. 5726), ERK1/2 (Cell Signaling Technology, Inc.; cat. no. 9102), Bcl-2 (Cell Signaling Technology, Inc.; cat. no. 3498), and integrin β1 (Cell Signaling Technology, Inc.; cat. no. 4706). GAPDH was used as the loading control (Cell Signaling Technology, Inc.; cat. no. 5174). After washing with TBST buffer, membranes were incubated with the secondary horseradish peroxidase-conjugated antibodies (1:1,000) for 1 h at room temperature [anti-rabbit IgG (cat. no. 7074) or anti-mouse IgG (cat. no. 7076)]. An enhanced chemiluminescent detection kit was used to visualize the immunoreactive protein bands using the Montreal Biotech Fusion Pulse 6 imaging system (Montreal Biotech Inc.). All experiments were repeated three times.

Statistical analysis

Data were analyzed using GraphPad Prism version 9 (GraphPad Software, Inc.). A one-way ANOVA followed by a Tukey's multiple comparison test was used to compare the difference between multiple groups. The half-maximal inhibitory concentration (IC50) values were obtained by applying a nonlinear regression curve fit analysis. P<0.05 was considered to indicate a statistically significant difference. Data are presented as the mean ± SEM.

Results

Cytotoxic effects of amlodipine on breast cancer cells

The in vitro biological effects of amlodipine treatment on MDA-MB-231 and MCF-7 cell proliferation are shown in Fig. 1. Amlodipine treatment reduced MDA-MB-231 and MCF-7 cell viability in a dose-dependent manner. In MDA-MB-231 cells, treatment with 2.5-50 µM amlodipine significantly reduced cell viability compared with the control-treated cells (P<0.05, Fig. 1A). In MCF-7 cells, 5-50 µM amlodipine significantly reduced cell viability compared with the control-treated cells (P<0.05, Fig. 1B). The IC50 values for amlodipine in MDA-MB-231 and MCF-7 cells were 8.66 and 12.60 µM, respectively. These findings suggest a cytotoxic effect of amlodipine on breast cancer cells.

Since amlodipine reduced breast cancer cell viability, the potential underlying mechanisms of the growth suppression were assessed by analyzing caspase-3/7 activity, which is a well-established marker of apoptosis (22). The results revealed that amlodipine treatment (2.5-10 µM) significantly increased caspase-3/7 activity compared with the control treatment in MDA-MB-231 cells (P<0.05, Fig. 1C) and thus highlighted caspase activation as a potential mechanism underlying the cytotoxic effects of amlodipine. However, caspase-3/7 activity was not assessed in the MCF-7 cells since they do not express caspase-3, and thus caspase activation may be underestimated (23,24).

To further determine the anticancer effects of amlodipine in cell proliferation, whether amlodipine could modulate anchorage-dependent growth was assessed using colony formation assays. Although it may be considered a limitation of the present study for MCF-7 cells to have a high capacity to form colonies compared to MDA-MB-231 cells (25), the MCF-7 clonogenic ability was still assessed following amlodipine treatment. The effect of amlodipine on colony formation of MCF-7 cells is shown in Fig. 2A and B. Amlodipine markedly inhibited colony formation in a dose-dependent manner in MCF-7 cells. Treatment with 2.5-10 µM amlodipine significantly decreased the colony size compared with the control (P<0.05). These findings indicate the ability of amlodipine to suppress the clonogenic proliferation of breast cancer cells over a prolonged period of time.

Effect of amlodipine on the invasion of breast cancer cells

To mimic the in vivo process of cell invasion through the extracellular matrix, the effects of amlodipine on the invasive abilities of MDA-MB-231 cells were evaluated using Matrigel invasion chambers. As shown in Fig. 2C and D, amlodipine significantly suppressed MDA-MB-231 cell invasion in a dose-dependent manner. Treatment with 5 and 10 µM amlodipine significantly reduced MDA-MB-231 invasiveness by >60 and 90% compared with the control-treated cells, respectively. Since MCF-7 cells are not highly invasive cells (25), invasion assays were not performed using these cells, which is considered a limitation of our study. These results provide robust evidence of the anti-invasive effects of amlodipine on breast cancer cells in vitro.

Anticancer effects of amlodipine may be mediated via ERK1/2, integrin β1, and Bcl-2 inhibition

To further shed light on the potential signaling molecules driving the anticancer effects of amlodipine on breast cancer cells, the expression levels of key proteins involved in cell proliferation, apoptosis, and invasion were evaluated using western blotting. The results indicated that amlodipine reduced the protein expression levels of the anti-apoptotic protein Bcl-2, in both MDA-MB-231 and MCF-7 cells compared with the control (Fig. 3). As MCF-7 cells are not highly invasive cells, p-ERK1/2 and integrin β1 protein expression levels were only assessed in MDA-MB-231 cells. Amlodipine treatment reduced ERK1/2 phosphorylation in MDA-MB-231 cells compared with the control treatment. Moreover, amlodipine reduced the protein expression levels of integrin-β1 in MDA-MB-231 cells compared with the control treatment. These findings may partially explain the possible molecular drivers of the anti-cancer effects of amlodipine.

Discussion

Voltage-activated calcium channels are widely distributed in all types of human cells (26). Several studies have shown that calcium channel expression is altered as an adaptive mechanism in human cancers such as in breast, prostate, and colorectal cancer (26-28). Interestingly, recent studies have implicated calcium channels in cancer cell proliferation, invasion, and metastasis (29-31). Recent studies have also shown that calcium channel expression is upregulated in breast cancer cells (31,32). In the present study, treatment of breast cancer cells with the CCB, amlodipine, resulted in a dose-dependent reduction in breast cancer cell viability. Similarly, recent studies have shown that silencing calcium channel expression inhibited breast cancer cell growth both in vitro and in vivo (31,32).

To ascertain the underlying mechanism(s) of amlodipine-induced growth suppression, breast cancer cellular apoptosis was assessed by measuring caspase-3/7 activity. The results showed that amlodipine induced caspase-3/7 activity in MDA-MB-231 cells, which may contribute to caspase-dependent apoptosis. Although this finding was limited by a lack of flow cytometry analysis to confirm the occurrence of apoptosis, activation of caspase-3/7 pathways was accompanied by downregulation of the anti-apoptotic protein Bcl-2, which strongly indicated that caspase-dependent apoptosis occurred in the breast cancer cells following amlodipine treatment. The Bcl-2 gene promotes cell survival and protects cells against apoptosis. High expression of Bcl-2 is associated with lower apoptosis-mediated death and contributes to resistance to chemotherapy. Moreover, Bcl-2 protein expression is typically altered in breast cancer cells (33,34). In agreement with the findings of the present study, a previous study demonstrated that amlodipine treatment induced apoptosis in MDA-MB-231 cells via downregulation of Bcl-2 protein expression (35). In addition, activation of caspase-dependent apoptosis has been reported with other dihydropyridine CCBs (36). Moreover, Wong et al (36) reported that treating cancer cells with calcium channel inhibitors may also lead to caspase-independent apoptosis. In the present study, amlodipine treatment of breast cancer cells resulted in caspase-dependent apoptosis as shown by the activation of caspase3/7. However, the increase in caspase3/7 activity appeared to decrease at higher concentrations (10-25 µM), which could be due to the dominance of caspase-independent apoptosis at higher concentrations.

To further illustrate the antiproliferative effect of amlodipine, the tumorigenic ability of breast cancer cells was assessed using colony formation assays whilst being treated with amlodipine. The inhibitory effects of amlodipine on colony formation were notable in MCF-7 cells and in agreement with previous findings in gastric cancer (37). To the best of our knowledge, this is the first study to provide proof of the inhibitory effects of amlodipine on breast cancer colony formation. In the present study, the effects of amlodipine on breast cancer cell proliferation and colony formation were accompanied by a reduction in ERK1/2 phosphorylation. Recent studies have also shown the inhibitory effects of amlodipine and other CCBs on the ERK1/2 pathway in gastric cancer (38), hepatic cancer (39), ovarian cancer (40), and melanoma (41). Taken together, the current and previous studies highlight the suppressive effects of amlodipine on cell proliferation, resistance to apoptosis, and tumorigenic potential via inhibition of major signaling proteins such as ERK1/2 and Bcl-2.

Previous studies have implicated calcium channels in breast cancer cell adhesion and invasion (30,31,42). For example, silencing calcium channel expression in breast cancer cells has been associated with a reduction in cell motility and adhesion (31). In the present study, amlodipine significantly reduced MDA-MB-231 breast cancer cell invasion. Filopodia structures are finger-like cytoplasmic projections that extend beyond the cell's edge and promote cancer cell invasion (42,43). The results of the present study are consistent with a recent study that showed the ability of several CCBs, including amlodipine, in inhibit filopodia formation and thus impairing breast cancer cell invasion (30). In the present study, the anti-invasive effects of amlodipine were accompanied by downregulation in p-ERK1/2 and integrin β1 protein expression. These proteins are well-established key players in breast cancer cell migration, invasion, and metastasis (44-46). Together, these findings suggest that the anti-invasive effects of amlodipine are mediated via at least the inhibition of p-ERK1/2 and integrin β1 expression.

In conclusion, the results of the present study showed that amlodipine exerted anticancer effects on cell proliferation, colony formation, and invasion, and they were, at least in part, achieved by the inhibition of p-ERK1/2, integrin β1, and Bcl-2 expression and activation of caspase-3/7, indicating the induction of caspase-dependent apoptosis. This study highlights amlodipine as a potential therapeutic agent for the management of breast cancer and may provide novel insights for future research on the effects of amlodipine in the sensitization of breast cancer cells to chemotherapy.

Acknowledgements

We would like to acknowledge Jordan University of Science and Technology for providing sabbatical leave to Dr. Mohammad A. Y. Alqudah. We are grateful to Dr. Moh'd Shara for his kind revision of the language proficiency of this manuscript and to Prof. Omar Khabour for providing us with the total ERK1/2 antibody (Jordan University of Science and Technology, Jordan).

Funding

Funding: This project was funded by Jordan University of Science and Technology (Deanship of Research, grant no. 238/2019).

Availability of data and materials

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

Authors' contributions

MAYA was involved in the study conception and design. MAYA, RAS and MA performed the experiments. MAYA, RAS, and KHA conducted the data analysis. MAYA and RAS wrote the first draft of the manuscript. All authors revised 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 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

2 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021.PubMed/NCBI View Article : Google Scholar

3 

World Health Organization. Jordan-Global Cancer Observatory [Fact sheet] 2020, December. Available from: https://gco.iarc.fr/today/data/factsheets/populations/400-jordan-fact-sheets.pdf.

4 

Seyfried TN and Huysentruyt LC: On the origin of cancer metastasis. Crit Rev Oncog. 18:43–73. 2013.PubMed/NCBI View Article : Google Scholar

5 

van Zijl F, Krupitza G and Mikulits W: Initial steps of metastasis: Cell invasion and endothelial transmigration. Mutat Res. 728:23–34. 2011.PubMed/NCBI View Article : Google Scholar

6 

Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR (eds), et al: SEER cancer statistics review, 1975-2017, National Cancer Institute. Bethesda, MD, 2020. Available from: https://seer.cancer.gov/csr/1975_2017/.

7 

American Cancer Society. Breast cancer facts and figures 2019-2020: American Cancer Society, 2020. Available from: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/breast-cancer-facts-and-figures/breast-cancer-facts-and-figures-2019-2020.pdf.

8 

Carrick S, Parker S, Thornton CE, Ghersi D, Simes J and Wilcken N: Single agent versus combination chemotherapy for metastatic breast cancer. Cochrane Database Syst Rev: Apr 15, 2009 (Epub ahead of print). doi: 10.1002/14651858.CD003372.pub3.

9 

Boster BL, Patel NK and Michaud LB: Breast cancer. In: Pharmacotherapy: A Pathophysiologic Approach, 11e. DiPiro JT, Yee GC, Posey LM, Haines ST, Nolin TD and Ellingrod V (eds). McGraw-Hill Education, New York, NY, 2020.

10 

Velaei K, Samadi N, Barazvan B and Soleimani Rad J: Tumor microenvironment-mediated chemoresistance in breast cancer. Breast. 30:92–100. 2016.PubMed/NCBI View Article : Google Scholar

11 

Wilson LE, D'Aloisio AA, Sandler DP and Taylor JA: Long-term use of calcium channel blocking drugs and breast cancer risk in a prospective cohort of US and Puerto Rican women. Breast Cancer Res. 18(61)2016.PubMed/NCBI View Article : Google Scholar

12 

Lee AR, Seo MJ, Kim J, Lee DM, Kim IY, Yoon MJ, Hoon H and Choi KS: Lercanidipine synergistically enhances bortezomib cytotoxicity in cancer cells via enhanced endoplasmic reticulum stress and mitochondrial Ca2+ overload. Int J Mol Sci. 20(6112)2019.PubMed/NCBI View Article : Google Scholar

13 

Alqudah MAY, Alrababah BA and Mhaidat NM: Amlodipine inhibits cell proliferation and induces cell cycle arrest in colorectal cancer cells. Jordan J Pharm Sci. 10:189–197. 2017.

14 

Ji BS, He L and Liu GQ: Reversal of p-glycoprotein-mediated multidrug resistance by CJX1, an amlodipine derivative, in doxorubicin-resistant human myelogenous leukemia (K562/DOX) cells. Life Sci. 77:2221–2232. 2005.PubMed/NCBI View Article : Google Scholar

15 

Yoshida J, Ishibashi T and Nishio M: Antitumor effects of amlodipine, a Ca2+ channel blocker, on human epidermoid carcinoma A431 cells in vitro and in vivo. Eur J Pharmacol. 492:103–112. 2004.PubMed/NCBI View Article : Google Scholar

16 

Alqudah MA, Agarwal S, Al-Keilani MS, Sibenaller ZA, Ryken TC and Assem M: NOTCH3 is a prognostic factor that promotes glioma cell proliferation, migration and invasion via activation of CCND1 and EGFR. PLoS One. 8(e77299)2013.PubMed/NCBI View Article : Google Scholar

17 

Al-Oudat BA, Alqudah MA, Audat SA, Al-Balas QA, El-Elimat T, Hassan MA, Frhat IN and Azaizeh MM: Design, synthesis, and biologic evaluation of novel chrysin derivatives as cytotoxic agents and caspase-3/7 activators. Drug Des Devel Ther. 13:423–433. 2019.PubMed/NCBI View Article : Google Scholar

18 

Siragusa M, Dall'Olio S, Fredericia PM, Jensen M and Groesser T: Cell colony counter called CoCoNut. PLoS One. 13(e0205823)2018.PubMed/NCBI View Article : Google Scholar

19 

Ayoub NM, Alkhalifa AE, Ibrahim DR and Alhusban A: Combined crizotinib and endocrine drugs inhibit proliferation, migration, and colony formation of breast cancer cells via downregulation of MET and estrogen receptor. Med Oncol. 38(8)2021.PubMed/NCBI View Article : Google Scholar

20 

Ayoub NM, Al-Shami KM, Alqudah MA and Mhaidat NM: Crizotinib, a MET inhibitor, inhibits growth, migration, and invasion of breast cancer cells in vitro and synergizes with chemotherapeutic agents. Onco Targets. 10:4869–4883. 2017.PubMed/NCBI View Article : Google Scholar

21 

Alqudah MAY, Azaizeh M, Zayed A and Asaad L: Calcium-sensing receptor antagonist NPS-2143 inhibits breast cancer cell proliferation, migration and invasion via downregulation of p-ERK1/2, Bcl-2 and integrin β1 and induces caspase 3/7 activation. Adv Pharm Bull. 12:383–388. 2022.

22 

Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E and Boise LH: Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 14(32)2013.PubMed/NCBI View Article : Google Scholar

23 

Jänicke RU: MCF-7 breast carcinoma cells do not express caspase-3. Breast Cancer Res Treat. 117:219–221. 2009.PubMed/NCBI View Article : Google Scholar

24 

Kottke TJ, Blajeski AL, Meng XW, Svingen PA, Ruchaud S, Mesner PW Jr, Boerner SA, Samejima K, Henriquez NV, Chilcote TJ, et al: Lack of correlation between caspase activation and caspase activity assays in paclitaxel-treated MCF-7 breast cancer cells. J Biol Chem. 277:804–815. 2002.PubMed/NCBI View Article : Google Scholar

25 

Comşa Ş, Cîmpean AM and Raica M: The story of MCF-7 breast cancer cell line: 40 Years of experience in research. Anticancer Res. 35:3147–3154. 2015.PubMed/NCBI

26 

Yamakage M and Namiki A: Calcium channels-basic aspects of their structure, function and gene encoding; anesthetic action on the channels-a review. Can J Anaesth. 49:151–164. 2002.PubMed/NCBI View Article : Google Scholar

27 

Phan NN, Wang CY, Chen CF, Sun Z, Lai MD and Lin YC: Voltage-gated calcium channels: Novel targets for cancer therapy. Oncol Lett. 14:2059–2074. 2017.PubMed/NCBI View Article : Google Scholar

28 

Prevarskaya N, Skryma R and Shuba Y: Ion channels and the hallmarks of cancer. Trends Mol Med. 16:107–121. 2010.PubMed/NCBI View Article : Google Scholar

29 

Varghese E, Samuel SM, Sadiq Z, Kubatka P, Liskova A, Benacka J, Pazinka P, Kruzliak P and Büsselberg D: Anti-cancer agents in proliferation and cell death: The calcium connection. Int J Mol Sci. 20(3017)2019.PubMed/NCBI View Article : Google Scholar

30 

Jacquemet G, Baghirov H, Georgiadou M, Sihto H, Peuhu E, Cettour-Janet P, He T, Perälä M, Kronqvist P, Joensuu H and Ivaska J: L-type calcium channels regulate filopodia stability and cancer cell invasion downstream of integrin signalling. Nat Commun. 7(13297)2016.PubMed/NCBI View Article : Google Scholar

31 

Kanwar N, Carmine-Simmen K, Nair R, Wang C, Moghadas-Jafari S, Blaser H, Tran-Thanh D, Wang D, Wang P, Wang J, et al: Amplification of a calcium channel subunit CACNG4 increases breast cancer metastasis. EBioMedicine. 52(102646)2020.PubMed/NCBI View Article : Google Scholar

32 

Ji Y, Han Z, Shao L and Zhao Y: Ultrasound-targeted microbubble destruction of calcium channel subunit α 1D siRNA inhibits breast cancer via G protein-coupled receptor 30. Oncol Rep. 36:1886–1892. 2016.PubMed/NCBI View Article : Google Scholar

33 

Zhang GJ, Kimijima I, Tsuchiya A and Abe R: The role of bcl-2 expression in breast carcinomas (Review). Oncol Rep. 5:1211–1216. 1998.PubMed/NCBI View Article : Google Scholar

34 

Pratt MA, Niu M and White D: Differential regulation of protein expression, growth and apoptosis by natural and synthetic retinoids. J Cell Biochem. 90:692–708. 2003.PubMed/NCBI View Article : Google Scholar

35 

Lan L, Xinghua X, Wenjuan S and Liying D: Effect of amlodipine on apoptosis of human breast carcinoma MDA-MB-231 cells. J Med Coll PLA. 23:358–363. 2008.PubMed/NCBI View Article : Google Scholar

36 

Wong BS, Chiu LY, Tu DG, Sheu GT and Chan TT: Anticancer effects of antihypertensive L-type calcium channel blockers on chemoresistant lung cancer cells via autophagy and apoptosis. Cancer Manag Res. 12:1913–1927. 2020.PubMed/NCBI View Article : Google Scholar

37 

Shiozaki A, Katsurahara K, Kudou M, Shimizu H, Kosuga T, Ito H, Arita T, Konishi H, Komatsu S, Kubota T, et al: Amlodipine and verapamil, voltage-gated Ca2+ channel inhibitors, suppressed the growth of gastric cancer stem cells. Ann Surg Oncol. 28:5400–5411. 2021.PubMed/NCBI View Article : Google Scholar

38 

Panneerpandian P, Rao DB and Ganesan K: Calcium channel blockers lercanidipine and amlodipine inhibit YY1/ERK/TGF-β mediated transcription and sensitize the gastric cancer cells to doxorubicin. Toxicol In Vitro. 74(105152)2021.PubMed/NCBI View Article : Google Scholar

39 

Li Y, Liu S, Lu F, Zhang T, Chen H, Wu S and Zhuang H: A role of functional T-type Ca2+ channel in hepatocellular carcinoma cell proliferation. Oncol Rep. 22:1229–1235. 2009.PubMed/NCBI View Article : Google Scholar

40 

Lee H, Kim JW, Kim DK, Choi DK, Lee S, Yu JH, Kwon OB, Lee J, Lee DS, Kim JH and Min SH: Calcium channels as novel therapeutic targets for ovarian cancer stem cells. Int J Mol Sci. 21(2327)2020.PubMed/NCBI View Article : Google Scholar

41 

Granados K, Hüser L, Federico A, Sachindra S, Wolff G, Hielscher T, Novak D, Madrigal-Gamboa V, Sun Q, Vierthaler M, et al: T-type calcium channel inhibition restores sensitivity to MAPK inhibitors in de-differentiated and adaptive melanoma cells. Br J Cancer. 122:1023–1036. 2020.PubMed/NCBI View Article : Google Scholar

42 

Jacquemet G, Hamidi H and Ivaska J: Filopodia in cell adhesion, 3D migration and cancer cell invasion. Curr Opin Cell Biol. 36:23–31. 2015.PubMed/NCBI View Article : Google Scholar

43 

Jacquemet G, Green DM, Bridgewater RE, von Kriegsheim A, Humphries MJ, Norman JC and Caswell PT: RCP-driven α5β1 recycling suppresses Rac and promotes RhoA activity via the RacGAP1-IQGAP1 complex. J Cell Biol. 202:917–935. 2013.PubMed/NCBI View Article : Google Scholar

44 

Hou S, Isaji T, Hang Q, Im S, Fukuda T and Gu J: Distinct effects of β1 integrin on cell proliferation and cellular signaling in MDA-MB-231 breast cancer cells. Sci Rep. 6(18430)2016.PubMed/NCBI View Article : Google Scholar

45 

Yin HL, Wu CC, Lin CH, Chai CY, Hou MF, Chang SJ, Tsai HP, Hung WC, Pan MR and Luo CW: β1 integrin as a prognostic and predictive marker in triple-negative breast cancer. Int J Mol Sci. 17(1432)2016.PubMed/NCBI View Article : Google Scholar

46 

Thibaudeau L, Taubenberger AV, Theodoropoulos C, Holzapfel BM, Ramuz O, Straub M and Hutmacher DW: New mechanistic insights of integrin β1 in breast cancer bone colonization. Oncotarget. 6:332–344. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2022
Volume 16 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Alqudah MA, Al‑Samman R, Azaizeh M and Alzoubi KH: Amlodipine inhibits proliferation, invasion, and colony formation of breast cancer cells. Biomed Rep 16: 50, 2022
APA
Alqudah, M.A., Al‑Samman, R., Azaizeh, M., & Alzoubi, K.H. (2022). Amlodipine inhibits proliferation, invasion, and colony formation of breast cancer cells. Biomedical Reports, 16, 50. https://doi.org/10.3892/br.2022.1533
MLA
Alqudah, M. A., Al‑Samman, R., Azaizeh, M., Alzoubi, K. H."Amlodipine inhibits proliferation, invasion, and colony formation of breast cancer cells". Biomedical Reports 16.6 (2022): 50.
Chicago
Alqudah, M. A., Al‑Samman, R., Azaizeh, M., Alzoubi, K. H."Amlodipine inhibits proliferation, invasion, and colony formation of breast cancer cells". Biomedical Reports 16, no. 6 (2022): 50. https://doi.org/10.3892/br.2022.1533