Open Access

Anticancer and apoptosis‑inducing effects of quercetin in vitro and in vivo

  • Authors:
    • Mahmoud Hashemzaei
    • Amin Delarami Far
    • Arezoo Yari
    • Reza Entezari Heravi
    • Kaveh Tabrizian
    • Seyed Mohammad Taghdisi
    • Sarvenaz Ekhtiari Sadegh
    • Konstantinos Tsarouhas
    • Dimitrios Kouretas
    • George Tzanakakis
    • Dragana Nikitovic
    • Nikita Yurevich Anisimov
    • Demetrios   A. Spandidos
    • Aristides M. Tsatsakis
    • Ramin Rezaee
  • View Affiliations

  • Published online on: June 28, 2017     https://doi.org/10.3892/or.2017.5766
  • Pages: 819-828
  • Copyright: © Hashemzaei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study focused on the elucidation of the putative anticancer potential of quercetin. The anticancer activity of quercetin at 10, 20, 40, 80 and 120 µM was assessed in vitro by MMT assay in 9 tumor cell lines (colon carcinoma CT‑26 cells, prostate adenocarcinoma LNCaP cells, human prostate PC3 cells, pheocromocytoma PC12 cells, estrogen receptor‑positive breast cancer MCF‑7 cells, acute lymphoblastic leukemia MOLT‑4 T‑cells, human myeloma U266B1 cells, human lymphoid Raji cells and ovarian cancer CHO cells). Quercetin was found to induce the apoptosis of all the tested cancer cell lines at the utilized concentrations. Moreover, quercetin significantly induced the apoptosis of the CT‑26, LNCaP, MOLT‑4 and Raji cell lines, as compared to control group (P<0.001), as demonstrated by Annexin V/PI staining. In in vivo experiments, mice bearing MCF‑7 and CT‑26 tumors exhibited a significant reduction in tumor volume in the quercetin‑treated group as compared to the control group (P<0.001). Taken together, quercetin, a naturally occurring compound, exhibits anticancer properties both in vivo and in vitro.

Introduction

The development or identification of compounds capable of killing transformed or cancer cells, without being toxic to their normal counterparts, is of utmost importance, and has gained the increasing interest of scientists worldwide. Since antiquity, plants have been considered rich sources of chemicals, with immense therapeutic potential. During recent years, some of these plant-derived compounds or phytochemicals have been shown to be highly competent anticancer agents, in addition to being effective against many other diseases (14).

Cancer, following cardiovascular diseases, is the main cause of mortality and morbidity in Europe. The key characteristics of this aggressive disease are uncontrolled growth and the spread of transformed cells (5). Each year, millions of individuals are diagnosed with cancer, whereas approximately 3.5 million cancer-related deaths are annually recorded worldwide (6,7). Specifically, only in Europe, approximately 3.45 million new cases of cancer were reported in 2012, excluding non-melanoma skin cancer, whereas approximately 1.75 million deaths occurred (8).

Throughout history, plant extracts and their purified active components have been the backbone of cancer chemotherapeutics (9). It is estimated that over 70% of anticancer compounds are either natural products, or natural product-derived substances (10).

Natural polyphenols are a large and abundant group of phytochemicals found in herbal beverages and food (1113). The plethora of these compounds is highlighted by the fact that up to date, over 8,000 polyphenols have been identified. Structurally, polyphenols consist of aromatic rings which are functionalized with one or more hydroxyl groups (12). Flavonoids and phenolics in particular, represent an important component of a normal human diet (14,15). The average daily flavonoid intake varies from approximately 1–2 g per day (16), depending on the type and the amount of fruit, vegetables or beverages consumed. The existence of a negative correlation between a diet rich in polyphenols and various diseases, such as cancer, cardiovascular and degenerative diseases has been well established (4,17). The total phenolic content (TPC) and the total flavonoid content (TFC) seem to correlate positively with in vivo and in vitro anticancer and antioxidant properties (18). Indeed, The American Institute of Cancer Research has reported that a high intake of fruit and vegetables correlates with a low risk of tumor occurrence (19). In addition, The National Academy of Sciences of the United States (1982) has also laid stress on the importance of fruits and vegetables in cancer prevention by including respective guidelines in its report on diet and cancer (2022). The polyphenolic compounds have been reported to have many pharmacological activities, such as antioxidant, anti-inflammatory, anticarcinogenic, antiviral, or antiallergic effects (3,4,2325).

Among anticancer and cancer preventing drugs, flavonoids are the most studied ones. These compounds can interfere with specific stages of the carcinogenic process, inhibit cell proliferation and induce apoptosis in several types of cancer cells (2630). Flavonoids demonstrate a notable antioxidant activity, not only by inhibiting ROS generation, but also by affecting the activity of sundry detoxifying enzymes, such as cyclooxygenases, lipoxygenases and inducible nitric oxide synthase (3,3133). This antioxidant capacity of flavonoids could possibly account for their anticancer potency. Flavonoids have also been found to influence epigenetic alterations by chromatin remodeling (34,35).

Quercetin (3,3′,4′,5,7-pentahydroxyflavone) belongs to polyphenolic flavonoids which are abundantly found in apples, red grapes, onions, raspberries, honey, cherries, citrus fruits and green leafy vegetables, and exerts various biological effects, including antioxidant, anticancer, antiviral, apoptosis-inducing, protein kinase C-inhibitory, cell cycle modulatory and angiogenesis inhibitory effects. Indeed, quercetin is a unique compound due to its potential to combat cancer-related diseases in a multi-targeted manner (3638). A number of studies have investigated the anticancer activity of quercetin (3941). In particular, it has been reported that quercetin at various concentrations, suppresses tumor growth of various cancer cell lines, including breast, colorectal, stomach, head and neck, lung, ovarian, melanoma and leukemia (4250). In addition, quercetin has been shown to inhibit the release of P-glycoprotein in the MCF-7 cell line and to enhance in vitro anticancer activity of adriamycin in breast cancer cell line (44).

Furthermore, it has been suggested that the chronic administration and daily intake of quercetin may be useful for prevention of some cancer types (51). However, cancer treatment in many cases is not effective, with disease recurrence and/or spreading leading to poor outcomes (52). Thus, development of novel therapeutic strategies remains an important goal in the on-going battle against cancer.

The aim of this study was to investigate the anticancer effects of quercetin in vitro and in vivo. The in vitro experiments were performed using a cohort of 9 representative cell lines. We examined the effects of quercetin on cancer cell proliferation (using MTT assay) and apoptosis [utilizing flow cytometer Annexin V/propidium iodide (PI) and TUNEL assay]. In mice bearing MCF-7 and CT-26 xenografts, we also performed an in vivo evaluation of the effects of quercetin.

Materials and methods

Cell culture

We used the following cancer cell lines: CT-26 which is a mouse colon carcinoma cell line that is widely used for drug development (53); PC-12 cell line which is derived from pheocromocytoma and has been widely used as a model of neuronal differentiation (54); LNCaP (androgen-sensitive cancer line) and PC-3 (androgen-insensitive cancer line) utilized as prostate cancer models; MOLT-4 [the T-cell line that causes acute lymphoblastic leukemia (ALL)], U266B1 (human myeloma cell line) and the Raji cell line (derived from human lymphoid) utilized as blood cancer models; we also used MCF-7 cells (estrogen receptor-positive breast cancer cells) as a breast cancer cell model; and CHO (ovarian cancer cell line) utilized to study ovarian cancer responses. The cancer cell lines (Pasteur Institute, Tehran, Iran) were grown under the following conditions: the CT-26, MOLT-4, U266B and PC12 in RPMI-1640 (Sigma, St. Louis, MO, USA); the PC3 cells in Ham's F12 (Sigma) and the LNCaP, MCF-7, Raji and CHO cells in DMEM (Sigma) supplied with 10% FBS (Gibco, Grand Island, NY, USA) and penicillin-streptomycin (Sigma) at 37°C in a humidified incubator containing 5% CO2. All experiments were performed when the cells reached 85–90% confluence.

Measurement of cell viability by MTT assay

Dilution series (10, 20, 40, 80 and 120 µM) of quercetin were prepared and used for 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. All 9 cancer cell lines were seeded at a density of 1.5×104 cells/well and treated with a range of concentrations in triplicate in 96-well cell culture plates, whereupon cell proliferation was assessed using a standard MTT assay. Specifically, the growth inhibitory activity of quercetin was determined using MTT, which correlates the cell number with the mitochondrial reduction of MTT to a blue formazan precipitate. In brief, the cells were plated in 96-well plates and allowed to attach overnight. The medium was then replaced with serum-free medium containing the test compounds and cells were incubated at 37°C for 72 h. The medium was then replaced with fresh medium containing 1 mg/ml MTT. Following incubation at 37°C for 2–4 h, the wells were aspirated, the dye was solubilized in DMSO and the absorbance was measured at 570 nm using a BioTek Instruments EL800 Microplate Reader (BioTek Instruments, Inc., Winooski, VT, USA). The viability of cells was compared with that of the control cells.

Detection of necrosis and apoptosis by flow cytometry

For the evaluation of apoptosis using the Annexin V/PI method, the LNCaP, CT-26, MOLT-4 and Raji cells were seeded in 12-well plates (2×105 cells/well). The cells were cultured and incubated (with 5% CO2 and 95% air) at 37°C. Various concentrations of quercetin (10, 20, 40, 80 and 120 µM were dissolved in DMSO and incubated with the cells for 48 h. DMSO in culture medium never exceeded 0.1% (v/v), the concentration known not to affect cell proliferation. The Annexin V-FITC/PI apoptosis kit (Abcam, Cambridge, MA, USA) was used. For this purpose, the cells were incubated with 5 µl Annexin V-FITC and 5 µl PI for 5 min in the dark. The treated cells were analyzed using a Partec PAS flow cytometer (Sysmex Partec GmbH, Gorlitz, Germany).

In vivo experiments

Female BALB/c mice, aged 6–8 weeks (weighing, 20–25 g) were obtained from Zabol University of Medical Sciences (Zabol, Iran). The animals were maintained in a temperature and humidity-controlled room. On day 1, the animals were shaved on the back flank. In the shaved right flank of each mouse, 3×105 CT-26 or MCF-7 cells in 50 µl PBS were injected subcutaneously, as previously described (55). Eight days after tumor implantation, the animals were randomly assigned into 8 groups as follows (5 mice in each group): i) CT-26 tumor-bearing mice treated with dextrose 5% (CT-26 control group); ii) CT-26 tumor-bearing mice treated with 50 mg/kg [intraperitoneally (i.p.)] quercetin; iii) CT-26 tumor-bearing mice treated with 100 mg/kg (i.p.) quercetin; iv) CT-26 tumor-bearing mice treated with 200 mg/kg (i.p.) quercetin; v) MCF-7 tumor-bearing mice treated with dextrose 5% (MCF-7 control group); vi) MCF-7 tumor-bearing mice treated with 50 mg/kg (i.p.) quercetin; vii) MCF-7 tumor-bearing mice treated with 100 mg/kg (i.p.) quercetin; and viii) MCF-7 tumor-bearing mice treated with 200 mg/kg (i.p.) quercetin. The tumor volume was measured and calculated based on the following formula: [(length)x(width)2]/2, as previously described (56,57).

In addition, the animal survival rate was evaluated up to 40 days. Furthermore, for apoptosis evaluation in the animals, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay was performed. The study protocol was approved by the Ethics Committee of Zabol University of Medical Sciences. All experimental procedures conformed to the declaration of Helsinki and were conducted in accordance with recent legislation of National Institutes of Health guide for the care and use of laboratory animals.

In situ apoptosis assay

Implanted tumor tissues were collected and apoptosis was detected using TUNEL assay. Based on the instructions of the manufacturer (Roche Diagnostics, Basel, Switzerland), paraformaldehyde-fixed blocks were embedded in paraffin, cut into 4-µm thick slices, and incubated with TUNEL reaction mixture containing TdT and fluorescein-dUTP. Prior to incubation of the slices with TUNEL mixture, their permeability was enhanced by proteinase solution. The TUNEL signal was then detected by an anti-fluorescein antibody conjugated with alkaline phosphatase (in situ Cell Death Detection kit; 11684809910 Roche; Sigma), a reporter enzyme, which catalytically generates a colored product. Three slides from each block and four slide fields were evaluated for the percentage of apoptotic cells. Four fields were randomly selectd on each slice and the number of apoptotic myocardial cells/total myocardial cells was calculated, at ×200 magnification. At the end of this procedure, the apoptotic index (number of myocardial nuclei labeled by the TUNEL method/number of total myocardial nuclei) was calculated, as previously described (58).

Statistical analysis

Statistical analysis was performed using the Student's t-test. Data are presented as the means ± standard deviation of 3 independent treatments. A value of P<0.05 was considered to indicate a statistically significant difference.

Results

Cell viability

MTT assay was used to evaluate the viability of all 9 cancer cell lines following 24, 48 and 72 h of treatment with quercetin at 10, 20, 40, 80 and 120 µM (Table I). All utilized concentrations of quercetin were inhibitory with the exception of the PC3 and CHO cells where at the 24 h time point, no inhibition was evident even at the highest concentration (120 µΜ) as presented in Table II. Furthermore, MTT assay revealed that the inhibitory effect of each concentration of quercetin on the viability of all cancer cell lines was enhanced by an increase in the incubation time. In addition, we calculated the IC50 values as previously described by Entezari Heravi et al (59). Therefore, the inhibitory effect of quercetin on the growth of utilized cancer cell lines was dose- and time-dependent, as demonstrated by the obtained IC50 values of quercetin (Table II). It is noteworthy, based on our cancer cell line panel (Table II), that we selected cell lines of lower and higher sensitivity to quercetin.

Table I.

Data obtained from MTT assay on the cell viability of different cell lines treated with quercetin (10, 20, 40, 80 and 120 µM) for 24, 48 and 72 h.

Table I.

Data obtained from MTT assay on the cell viability of different cell lines treated with quercetin (10, 20, 40, 80 and 120 µM) for 24, 48 and 72 h.

A, Cells treated for 24 h

Cell line 10µM 20 µM 40 µM 80 µM 120 µM
CT-26 94.2±4.4 83.5±3.7 75.1±4.2 65.8±5.5 49.7±5.9
LNCaP 96.8±5.4 90.3±4.6 71.7±2.2 61.5±3.4 45.1±5.0
PC3 96.4±5.0 87.6±4.9 80.1±4.6 75±4.4 73.2±4.1
PC12 91.1±6.5 84.5±6.0 68.5±6.8 57.1±6.3 40.3±4.4
MCF-7 94.8±6.1 90.2±5.9 77.5±5.1 66.4±4.7 47.1±4.2
MOLT-4 88.6±3.6 70.2±4.1 57.5±4.0 46±2.8 42.8±3
U266B1 95.1±4.9 73.1±5 53.8±4.6 47.5±3.3 33.8±4.7
Raji 85.6±4.1 80.1±3.2 68.1±2.8 42.9±3.2 29.4±4.6
CHO 97.7±5.2 91.3±5.5 74.4±4.1 75.4±4.1 70.5±5.2

B, Cells treated for 48 h

Cell line 10 µM 20 µM 40 µM 80 µM 120 µM

CT-26 87.4±5.4 77.7±5.9 70.3±4.1 61.5±3.2 42.1±3
LNCaP 91.5±6.3 84.2±5.1 66.6±5.7 46.7±4.9 38.5±3.8
PC3 89.9±3.6 77.6±3.2 70.7±2.8 52.2±3.3 28.5±3.4
PC12 94.4±5 80.8±3.4 62.5±4.6 44.5±3.2 30.7±3.8
MCF-7 81.3±4.1 70.2±3.1 55.5±3.4 39.6±3.7 25.2±2.1
MOLT-4 70.6±2.8 52.5±2.6 43.1±1.9 33.3±2.5 21.6±1.4
U266B1 68.5±2.3 53.4 ±1.8 37.2±2 33.4±1.6 20.4±2.1
Raji 60.6±3.6 49.5±2.3 30.3±2.4 26.4±2.3 14.6±3.3
CHO 97.4±4.4 81.3±3.4 64.6±2.8 45.8±2.6 21.9±3.5

C, Cells treated for 72 h

Cell line 10 µM 20 µM 40 µM 80 µM 120 µM

CT-26 65.5±1.5 55.8±1.9 35.9±0.83 29.7±1.1 25±2.3
LNCaP 58.4±2.9 51.4±2.6 39±1.9 36.1±2.2 30.7±2
PC3 61.7±2.1 57.3±1.8 46.9±1.4 36.2±0.9 31.5±3.7
PC12 50.4±3.6 47.2±2.6 40.8±1.8 31.9±2.3 22.1±1.1
MCF-7 51.6±3.2 48.5±2.9 35.7±2.5 30.8±3 19.1±1.4
MOLT-4 11.5±0.5 10.2±0.45 10±0.37 5.2±0.48 2.1±0.9
U266B1 15.9±0.8 13.3±0.65 4.8±0.72 6.8±1.1 5.5±0.38
Raji 5.5±0.4 2.7±0.8 1.3±0.25 0.25±0.12 0.18±0.09
CHO 57.8±3.9 52.4±3.2 39.2±2.7 28.5±2.1 20.7±3.7

Table II.

IC50 values (in µM) for the studied cell lines following treatment with various concentrations of quercetin (10, 20, 40, 80, 120 µM) for 24, 48 and 72 h.

Table II.

IC50 values (in µM) for the studied cell lines following treatment with various concentrations of quercetin (10, 20, 40, 80, 120 µM) for 24, 48 and 72 h.

Cell line 24 h 48 h 72 h
CT-26 118.1±5.55 97.5±4.31 27.2±1.52
LNCaP 110.7±4.30 72.6±5.15 21.7±2.31
PC3 >120 81.9±3.27 36±1.98
PC12   99.3±6.11 65.2±4 11.8±2.27
MCF-7 105.4±5.2 52.5±3.28 13.7±2.61
MOLT-4 64.9±3.5 28.6±2.23 2.91±0.54
U266B1 54.3±4.5 25±1.96 6.13±0.73
Raji   66.5±3.57 19.2±2.83 3.52±0.46
CHO >120 70.7±3.44 23.4±3.11

[i] IC50 values were calculated as previously described by Entezari Heravi et al (59).

Determination of the apoptosis of CT-26, LNCaP, MOLT-4 and Raji cell lines following treatment with quercetin

In continuation, we examined the apoptotic rate in a panel of 4 cell lines of high and low sensitivity to quercetin (i.e., CT-26 and LNCaP, as well as MOLT-4 and Raji, respectively). Our results revealed that quercetin initiated the apoptotic process in these cells in a dose-dependent manner (Fig. 2). The effect of quercetin at 120 µΜ on the apoptotic rate of the CT-26, LNCaP, MOLT-4 and Raji cell lines in comparison to the control group (P<0.001) is depicted in Fig. 1. The results of MTT assay were compatible with the results of Annexin V/PI assay, as the cell lines with a higher sensitivity to quercetin exhibited a higher apoptotic rate (P< 0.001; Figs. 1 and 2).

Effect of quercetin treatment on tumor volume

In continuation, we evaluated the in vivo effect of quercetin on CT-26 and MCF-7 tumors; these cells lines exhibited a relatively lower sensitivity to quercetin in in vitro experiments (Table II). Therefore, mice bearing CT-26 and MCF-7 tumors were treated with various concentrations of quercetin (50, 100 and 200 mg/kg; i.p.) This approach revealed that the administration of quercetin significantly reduced the tumor volume at all 3 utilized doses on day 18 post-treatment for CT26 tumors and on day 20 post-treatment for MCF-7 tumors (P<0.001; Fig. 3).

Animal survival rate

In vivo experiments demonstrated that the survival rate for mice bearing CT-26 and MCF-7 tumors that were treated with the two higher concentrations of quercetin (100 and 200 mg/kg) was significantly higher compared to the control group (P<0.05 and P<0.01, respectively). No significant difference in the survival rate was evident between the group treated with quercetin at 50 mg/kg and the control group (P>0.05; Fig. 4).

TUNEL assay

At the end of the experiment (36 days following treatment), all surviving animals were sacrificed, and tumors from all animals were dissected and the poly-D-lysine-coated coverslips for TUNEL assay were positioned. An increase in the percentage of apoptotic cells in the treated as compared to the control groups was observed; however, it did not reach statistical significance (P>0.05; Fig. 5).

Discussion

In spite of many advances in cancer therapy, cancer is still one of the major causes of mortality worldwide. Natural products, particularly flavones found in the human diet, have been found to exert anti-proliferative and apoptosis-promoting effects against cancer cells (60,61). The current study demonstrated that quercetin induces the apoptosis of various cancer cell lines. Furthermore, a significant increase in the survival rate and a significant reduction in tumor volume was observed in tumor-bearing animals treated with quercetin.

Previous studies have shown that grape stem extracts have an ability to inhibit the growth of colon (HT29), breast (MCF-7), renal (Caki-1) and thyroid (K1) cancer cell lines (62,63). These extracts are rich in flavonols, particularly quercetin and rutin (63). Importantly, quercetin was found to be in both aglycon and glycoside forms (64). The inhibitory effect of quercetin on cancer cell growth is attributed to the inhibition of survival signaling proteins, such as protein kinase C (PKC-α) and the activation of death signals, such as PKC-δ (63). Moreover, grape stem extracts seem to present an anti-angiogenic potential evident by VEGF downregulation (65).

Furthermore, quercetin induces pro-apoptotic effects via different mechanisms involving antioxidant effects and the suppression of p53 gene and BCL-2 protein (66). The suppression of BCL-2 gene transcription diminishes the inhibitory effects on BAD protein in the mitochondria, which is considered as the initiator of apoptosis for the intrinsic pathway (67).

The role of quercetin in apoptosis mediated by p53 has been studied in many cancer cell lines. When p53 is inhibited, cells become more susceptible toward cytotoxicity induced by quercetin (68). Apart from cell cycle regulation and the induction of apoptosis, p53 acts as a modulator of intracellular levels of ROS. In this regard, p53 exerts antioxidant effects in cells with no or low stress through the regulation of genes involved in such activity, which comprises microsomal GSH transferase homolog PIG12 (69), aldehyde dehydrogenase 4 family member A1 (ALDH4A1) (70), Gpx1, manganese superoxide dismutase (SOD2) (71) and catalase (72). Some studies, however, have suggested that the effect of quercetin may be independent of p53. Although apoptotic cell death caused by DNA damage is often mediated by p53, there are other proteins, such as p63 and p73, which may be involved in this mechanism (73). Chien et al demonstrated that quercetin-induced apoptotic cell death was accompanied by a decrease in p53 expression in breast cancer cells (74). Additionally, quercetin inhibited the metabolic activity and induced cell death by apoptosis, followed by an increase in BAX expression with a concomitant decrease in the expression of anti-apoptotic proteins.

Quercetin has also been found to modulate the PI3K/Akt/mTOR pathway (75). The flavonoid has structural homology to the PI3K inhibitor, LY294002 (LY) and as expected, the phytochemical was found to inhibit the PI3K-Akt pathway in a similar manner to the inhibition elicited by LY in the breast cancer cell lines, HCC1937 and T47D (76). Breast cancer invasion was found to be suppressed by quercetin through the inactivation of the PKC/ERK/AP-1-dependent MMP-9 activation (76).

Quercetin exerts anticancer effects through the cell death domain mechanism at the cell surface (67). Quercetin activated the cell death domain which leads to FAS and FADD activation, and the induction of cell death in a cancer cell line via activation of caspase 8 (67). The above-mentioned findings are in accordance with the findings of the present study, which demonstrated the apoptotic-inducing properties of quercetin as assessed by the Annexin V/PI method.

The expression of heat shock proteins (HSPs) in almost all forms of cancer is elevated (58). Badziul et al showed that quercetin decreased the transcription and translation of HSP27 and 72 in the T98G cell line (58). HSPs are involved in cell proliferation and the inhibition of their production leads to cell apoptosis (77). This is well in accordance with our study, as we demonstrated that apoptosis was induced in quercetin-treated cancer cell lines. HSP27 has been reported to promote the development of leukemia by protecting tumor cells from apoptosis through various mechanisms. Another study investigated the effects of small hairpin (sh)RNA-mediated HSP27 knockdown on the anticancer effects of quercetin in U937 human leukemia cells. The results indicated that shHSP27 and quercetin synergistically inhibited U937 cell proliferation and induced apoptosis by reducing the Bcl2/Bax ratio. Moreover, this combined treatment significantly suppressed the infiltration of tumor cells and the expression of the angiogenesis-associated proteins, hypoxia-inducible factor 1α (HIF1α) and vascular endothelial growth factor (VEGF). In comparison with shHSP27 or quercetin separately, shHSP27 and quercetin together, notably decreased the expression of cyclin D1, and thus the cell cycle was arrested at the G1 phase (78). Furthermore, the Notch/AKT/mTOR signaling pathway contributes to tumor aggressiveness; quercetin plus shHSP27 has been shown to significantly decrease Notch 1 expression and the phosphorylation levels of the downstream signaling proteins, mTOR and AKT (79).

The anticancer effects of quercetin have been confirmed in many studies (36,42,44). Specifically, in vitro and in vivo studies have suggested that quercetin possesses anticancer activity against different tumors; e.g. colon, lung, breast and prostate cancer (78,8084). Quercetin can exert its anticancer effects through different mechanisms, including the inhibition of DNA topoisomerase I/II, the release of cytochrome c, the activation of caspase 3, and HSP27 and 72 elevation (37,58,85).

An in vivo examination of the effects of quercetin in mice bearing CT-26 tumors was performed for the first time in this study, at least to the best of our knowledge. The results revealed that quercetin significantly reduced the tumor volume and increased animal survival. Previous studies have provided evidence for the anticancer effects of quercetin on breast and prostate cancers in vivo (86,87), an observation which was verified in the current study using the MCF-7 breast cancer in vivo model.

Importantly, quercetin can exert tumor suppressive effects by interfering with the cell cycle. The molecular targets of this flavonoid include p27, topoisomerase II, p21 and cyclin B (88,89). Depending on the tumor origin, quercetin is able to block the cell cycle at G2/M or at the G1/S transition (90,91). In breast cancer cell lines, a low dose of quercetin has been shown to induce mild DNA damage and Chk2 activation, which is the main regulator of p21 expression (92). In addition, quercetin has been shown to downregulate cyclin B1 and CDK1, essential components of G2/M cell cycle progression (92). Moreover, quercetin can inhibit the recruitment of NF-Y, a key transcription factor, which binds to the cyclin B1 gene promoter and leads to transcriptional cessation. In the human hepatoma cell line (HepG2), quercetin upregulated p21, p27 and p53, and consequently the cells were arrested at the G1 phase (93).

Furthermore, quercetin has been shown to inhibit NF-κB-evoked pathways of cell survival and reduce pro-inflammatory cytokine expression that finally leads to cancer formation (94). Notably, quercetin inhibits the production of tumor necrosis factor (TNF)-α, a major pro-inflammatory molecule involved in chronic inflammatory diseases, which may develop into tumors. The quercetin-induced suppression of TNF-α results in the stimulation of anti-inflammatory cytokines through the inhibition of NF-κB activation (95). TNF-α is a major regulator of the cellular release of other chemokines, cytokines and other inflammatory mediators, and thus can be considered as a potential target for the treatment of inflammatory diseases and inflammation-driven cancer. In the current study, quercetin was able to suppress tumor growth and improve animal survival (independent of its action on the induction of apoptosis).

Quercetin seems to play an inhibitory role in angiogenesis in human prostate tumor growth. A recent study using an animal model, indicated that low doses of quercetin inhibited the following angiogenic stages: proliferation and migration, as well as the invasion and tube formation of endothelial cells. Protein expression analysis of prostate cancer cells treated with quercetin revealed the inhibition of the VEGF-induced phosphorylation of VEGFR-2 and its downstream targets, such as mTOR, Akt, and ribosomal S6 kinase (86). Thus, quercetin can decrease tumor volume and increase animal survival rate following systemic administration, a finding which is consistent with the results of the present study.

The oral administration of quercetin is recommended for cancer prevention. It has been shown that a diet supplemented with 2% quercetin significantly reduced the onset of colorectal cancer (96). In addition, in vitro studies have proved its potency in inhibiting the proliferation of colon cancer cells of different lineages (97,98). However, in a phase-1 clinical study performed at the University of Birmingham for the evaluation of the non-toxic and anticancer efficacy of quercetin in terminally ill cancer patients, no patient achieved conventional radiological response according to the WHO criteria, despite favorable indications of its anticancer activity (99).

In conclusion, our results demonstrate that quercetin inhibits the growth of a panel of 9 cancer cell lines with various IC50 values. Cell growth inhibition was attributed to the induction of apoptosis, evident in the CT-26, PC-12, LNCaP and PC-3 cancer cell lines. Furthermore, our results demonstrated that quercetin reduced CT-26 and MCF-7 tumor volume in a mouse model and increased animal survival; however, we did not verify increased in situ apoptosis in the induced tumors. The current study results strongly suggest that quercetin has potential for therapeutic application in neoplasia; however, further studies are required to confirm these findings.

Acknowledgements

Special thanks to Zabol University of Medical Sciences for their financial support.

References

1 

Schnekenburger M, Dicato M and Diederich M: Plant-derived epigenetic modulators for cancer treatment and prevention. Biotechnol Adv. 32:1123–1132. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Butler MS, Robertson AA and Cooper MA: Natural product and natural product derived drugs in clinical trials. Nat Prod Rep. 31:1612–1661. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Hashemzaei M, Barani AK, Iranshahi M, Rezaee R, Tsarouhas K, Tsatsakis AM, Wilks MF and Tabrizian K: Effects of resveratrol on carbon monoxide-induced cardiotoxicity in rats. Environ Toxicol Pharmacol. 46:110–115. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Hashemzaei M, Heravi Entezari R, Rezaee R, Roohbakhsh A and Karimi G: Regulation of autophagy by some natural products as a potential therapeutic strategy for cardiovascular disorders. Eur J Pharmacol. 802:44–51. 2017. View Article : Google Scholar : PubMed/NCBI

5 

DeVita VT Jr, Young RC and Canellos GP: Combination versus single agent chemotherapy: A review of the basis for selection of drug treatment of cancer. Cancer. 35:98–110. 1975. View Article : Google Scholar : PubMed/NCBI

6 

Pisani P, Bray F and Parkin DM: Estimates of the world-wide prevalence of cancer for 25 sites in the adult population. Int J Cancer. 97:72–81. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: The impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D and Bray F: Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur J Cancer. 49:1374–1403. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Fridlender M, Kapulnik Y and Koltai H: Plant derived substances with anti-cancer activity: From folklore to practice. Front Plant Sci. 6:7992015. View Article : Google Scholar : PubMed/NCBI

10 

Karikas GA: Anticancer and chemopreventing natural products: Some biochemical and therapeutic aspects. J BUON. 15:627–638. 2010.PubMed/NCBI

11 

Katz DL, Doughty K and Ali A: Cocoa and chocolate in human health and disease. Antioxid Redox Signal. 15:2779–2811. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Pandey KB and Rizvi SI: Current understanding of dietary polyphenols and their role in health and disease. Curr Nutr Food Sci. 5:249–263. 2009. View Article : Google Scholar

13 

Pandey KB and Rizvi SI: Plant polyphenols as dietary antioxidants in human health and disease. Oxid Med Cell Longev. 2:270–278. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Kühnau J: The Flavonoids. A Class of Semi-Essential Food Components: Their Role in Human Nutrition. World Review of Nutrition and Dietetics. Bourne GH: Basel: Karger; pp. 117–191. 1976, View Article : Google Scholar : PubMed/NCBI

15 

Scalbert A and Williamson G: Dietary intake and bioavailability of polyphenols. J Nutr. 130:(Suppl). 2073S–2085S. 2000.PubMed/NCBI

16 

Benbrook CM: Elevating Antioxidant Levels in Food through Organic Farming and Food Processing. An Organic Center, State of Science Review. The Organic Center for Education and Promotion. 2005.

17 

Hashemzaei M, Karami SP, Delaramifar A, Sheidary A, Tabrizian K, Rezaee R, Shahsavand S, Arsene AL, Tsatsakis AM and Mohammad S: Anticancer effects of co-administration of daunorubicin and resveratrol in MOLT-4, U266 B1 and RAJI cell lines. Farmacia. 64:36–42. 2016.

18 

Ramkissoon JS, Mahomoodally MF, Ahmed N and Subratty AH: Antioxidant and anti-glycation activities correlates with phenolic composition of tropical medicinal herbs. Asian Pac J Trop Med. 6:561–569. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Glade MJ: Food, nutrition, and the prevention of cancer: a global perspective. American Institute for Cancer Research/World Cancer Research Fund, American Institute for Cancer Research, 1997. Nutrition. 15:523–526. 1999.PubMed/NCBI

20 

Murillo G and Mehta RG: Cruciferous vegetables and cancer prevention. Nutr Cancer. 41:17–28. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Cronin FJ, Krebs-Smith SM, Wyse BW and Light L: Characterizing food usage by demographic variables. J Am Diet Assoc. 81:661–673. 1982.PubMed/NCBI

22 

Block G, Dresser CM, Hartman AM and Carroll MD: Nutrient sources in the American diet: quantitative data from the NHANES II survey. II. Macronutrients and fats. Am J Epidemiol. 122:27–40. 1985. View Article : Google Scholar : PubMed/NCBI

23 

Hashemzaei M, SadeghiBonjar MA, Tabrizian K, Iranshahi M, Iranshahy M and Rezaee R: Evaluation of the analgesic effect of Umbelliprenin and Umbelliprenin-morphine co-administration on the acute, chronic and neuropathic pain. IJPER. 49:121–125. 2015. View Article : Google Scholar

24 

Kumar S and Pandey AK: Chemistry and biological activities of flavonoids: An overview. Sci World J. 2013:1627502013. View Article : Google Scholar

25 

Tabrizian K, Yaghoobi NS, Iranshahi M, Shahraki J, Rezaee R and Hashemzaei M: Auraptene consolidates memory, reverses scopolamine-disrupted memory in passive avoidance task, and ameliorates retention deficits in mice. Iran J Basic Med Sci. 18:1014–1019. 2015.PubMed/NCBI

26 

Nikitovic D, Tsatsakis AM, Karamanos NK and Tzanakakis GN: The effects of genistein on the synthesis and distribution of glycosaminoglycans/proteoglycans by two osteosarcoma cell lines depends on tyrosine kinase and the estrogen receptor density. Anticancer Res. 23:(1A). 459–464. 2003.PubMed/NCBI

27 

Yamaguchi M, Murata T, El-Rayes BF and Shoji M: The flavonoid p-hydroxycinnamic acid exhibits anticancer effects in human pancreatic cancer MIA PaCa-2 cells in vitro: Comparison with gemcitabine. Oncol Rep. 34:3304–3310. 2015.PubMed/NCBI

28 

Cárdenas M, Marder M, Blank VC and Roguin LP: Antitumor activity of some natural flavonoids and synthetic derivatives on various human and murine cancer cell lines. Bioorg Med Chem. 14:2966–2971. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Chan FL, Choi HL, Chen ZY, Chan PS and Huang Y: Induction of apoptosis in prostate cancer cell lines by a flavonoid, baicalin. Cancer Lett. 160:219–228. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Kawaii S, Tomono Y, Katase E, Ogawa K and Yano M: Antiproliferative activity of flavonoids on several cancer cell lines. Biosci Biotechnol Biochem. 63:896–899. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Lin P, Tian XH, Yi YS, Jiang WS, Zhou YJ and Cheng WJ: Luteolin-induced protection of H2O2-induced apoptosis in PC12 cells and the associated pathway. Mol Med Rep. 12:7699–7704. 2015.PubMed/NCBI

32 

Leung HW, Kuo CL, Yang WH, Lin CH and Lee HZ: Antioxidant enzymes activity involvement in luteolin-induced human lung squamous carcinoma CH27 cell apoptosis. Eur J Pharmacol. 534:12–18. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Wu TH, Yen FL, Lin LT, Tsai TR, Lin CC and Cham TM: Preparation, physicochemical characterization, and antioxidant effects of quercetin nanoparticles. Int J Pharm. 346:160–168. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Busch C, Burkard M, Leischner C, Lauer UM, Frank J and Venturelli S: Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin Epigenetics. 7:642015. View Article : Google Scholar : PubMed/NCBI

35 

Gilbert ER and Liu D: Flavonoids influence epigenetic-modifying enzyme activity: Structure - function relationships and the therapeutic potential for cancer. Curr Med Chem. 17:1756–1768. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Erlund I: Review of the flavonoids quercetin, hesperetin, and naringenin. Dietary sources, bioactivities, bioavailability, and epidemiology. Nutr Res. 24:851–874. 2004. View Article : Google Scholar

37 

Jakubowicz-Gil J, Paduch R, Piersiak T, Głowniak K, Gawron A and Kandefer-Szerszeń M: The effect of quercetin on pro-apoptotic activity of cisplatin in HeLa cells. Biochem Pharmacol. 69:1343–1350. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Ramos S: Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. J Nutr Biochem. 18:427–442. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Ren MX, Deng XH, Ai F, Yuan GY and Song HY: Effect of quercetin on the proliferation of the human ovarian cancer cell line SKOV-3 in vitro. Exp Ther Med. 10:579–583. 2015.PubMed/NCBI

40 

Deng XH, Song HY, Zhou YF, Yuan GY and Zheng FJ: Effects of quercetin on the proliferation of breast cancer cells and expression of survivin in vitro. Exp Ther Med. 6:1155–1158. 2013.PubMed/NCBI

41 

Ren KW, Li YH, Wu G, Ren JZ, Lu HB, Li ZM and Han XW: Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. Int J Oncol. 50:1299–1311. 2017.PubMed/NCBI

42 

Chen J and Kang JH: Quercetin and trichostatin A cooperatively kill human leukemia cells. Pharmazie. 60:856–860. 2005.PubMed/NCBI

43 

Priego S, Feddi F, Ferrer P, Mena S, Benlloch M, Ortega A, Carretero J, Obrador E, Asensi M and Estrela JM: Natural polyphenols facilitate elimination of HT-29 colorectal cancer xenografts by chemoradiotherapy: A Bcl-2- and superoxide dismutase 2-dependent mechanism. Mol Cancer Ther. 7:3330–3342. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Scambia G, Ranelletti FO, Panici PB, De Vincenzo R, Bonanno G, Ferrandina G, Piantelli M, Bussa S, Rumi C, Cianfriglia M, et al: Quercetin potentiates the effect of adriamycin in a multidrug-resistant MCF-7 human breast-cancer cell line: P-glycoprotein as a possible target. Cancer Chemother Pharmacol. 34:459–464. 1994. View Article : Google Scholar : PubMed/NCBI

45 

Yoshida M, Sakai T, Hosokawa N, Marui N, Matsumoto K, Fujioka A, Nishino H and Aoike A: The effect of quercetin on cell cycle progression and growth of human gastric cancer cells. FEBS Lett. 260:10–13. 1990. View Article : Google Scholar : PubMed/NCBI

46 

Sharma H, Sen S and Singh N: Molecular pathways in the chemosensitization of cisplatin by quercetin in human head and neck cancer. Cancer Biol Ther. 4:949–955. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Yang JH, Hsia TC, Kuo HM, Chao PD, Chou CC, Wei YH and Chung JG: Inhibition of lung cancer cell growth by quercetin glucuronides via G2/M arrest and induction of apoptosis. Drug Metab Dispos. 34:296–304. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Gao X, Wang B, Wei X, Men K, Zheng F, Zhou Y, Zheng Y, Gou M, Huang M, Guo G, et al: Anticancer effect and mechanism of polymer micelle-encapsulated quercetin on ovarian cancer. Nanoscale. 4:7021–7030. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Cao HH, Tse AK, Kwan HY, Yu H, Cheng CY, Su T, Fong WF and Yu ZL: Quercetin exerts anti-melanoma activities and inhibits STAT3 signaling. Biochem Pharmacol. 87:424–434. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Spagnuolo C, Russo M, Bilotto S, Tedesco I, Laratta B and Russo GL: Dietary polyphenols in cancer prevention: the example of the flavonoid quercetin in leukemia. Ann N Y Acad Sci. 1259:95–103. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Araújo JR, Gonçalves P and Martel F: Chemopreventive effect of dietary polyphenols in colorectal cancer cell lines. Nutr Res. 31:77–87. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Maira F, Catania A, Candido S, Russo AE, McCubrey JA, Libra M, Malaponte G and Fenga C: Molecular targeted therapy in melanoma: A way to reverse resistance to conventional drugs. Curr Drug Deliv. 9:17–29. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Castle JC, Loewer M, Boegel S, de Graaf J, Bender C, Tadmor AD, Boisguerin V, Bukur T, Sorn P, Paret C, et al: Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma. BMC Genomics. 15:1902014. View Article : Google Scholar : PubMed/NCBI

54 

Westerink RHS and Ewing AG: The PC12 cell as model for neurosecretion. Acta Physiol (Oxf). 192:273–285. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Mayhew EG, Lasic D, Babbar S and Martin FJ: Pharmacokinetics and antitumor activity of epirubicin encapsulated in long-circulating liposomes incorporating a polyethylene glycol-derivatized phospholipid. Int J Cancer. 51:302–309. 1992. View Article : Google Scholar : PubMed/NCBI

56 

Gao F, Li L, Liu T, Hao N, Liu H, Tan L, Li H, Huang X, Peng B, Yan C, et al: Doxorubicin loaded silica nanorattles actively seek tumors with improved anti-tumor effects. Nanoscale. 4:3365–3372. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Tang L, Gabrielson NP, Uckun FM, Fan TM and Cheng J: Size-dependent tumor penetration and in vivo efficacy of monodisperse drug-silica nanoconjugates. Mol Pharm. 10:883–892. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Bądziul D, Jakubowicz-Gil J, Langner E, Rzeski W, Głowniak K and Gawron A: The effect of quercetin and imperatorin on programmed cell death induction in T98G cells in vitro. Pharmacol Rep. 66:292–300. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Heravi Entezari R, Hadizadeh F, Sankian M, Afshari Tavakol J, Taghdisi SM, Jafarian H and Behravan J: Novel selective Cox-2 inhibitors induce apoptosis in Caco-2 colorectal carcinoma cell line. Eur J Pharm Sci. 44:479–86. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Chahar MK, Sharma N, Dobhal MP and Joshi YC: Flavonoids: A versatile source of anticancer drugs. Pharmacogn Rev. 5:1–12. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Batra P and Sharma AK: Anti-cancer potential of flavonoids: recent trends and future perspectives. 3 Biotech. 3:439–459. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Apostolou A, Stagos D, Galitsiou E, Spyrou A, Haroutounian S, Portesis N, Trizoglou I, Hayes Wallace A, Tsatsakis AM and Kouretas D: Assessment of polyphenolic content, antioxidant activity, protection against ROS-induced DNA damage and anticancer activity of Vitis vinifera stem extracts. Food Chem Toxicol. 61:60–68. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Sahpazidou D, Geromichalos GD, Stagos D, Apostolou A, Haroutounian SA, Tsatsakis AM, Tzanakakis GN, Hayes AW and Kouretas D: Anticarcinogenic activity of polyphenolic extracts from grape stems against breast, colon, renal and thyroid cancer cells. Toxicol Lett. 230:218–224. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Erlund I, Kosonen T, Alfthan G, Mäenpää J, Perttunen K, Kenraali J, Parantainen J and Aro A: Pharmacokinetics of quercetin from quercetin aglycone and rutin in healthy volunteers. Eur J Clin Pharmacol. 56:545–553. 2000. View Article : Google Scholar : PubMed/NCBI

65 

Stagos D, Apostolou A, Poulios E, Kermeliotou E, Mpatzilioti A, Kreatsouli K, Koulocheri SD, Haroutounian SA and Kouretas D: Antiangiogenic potential of grape stem extract through inhibition of vascular endothelial growth factor expression. J Physiol Pharmacol. 65:843–852. 2014.PubMed/NCBI

66 

Hsu CL and Yen GC: Phenolic compounds: Evidence for inhibitory effects against obesity and their underlying molecular signaling mechanisms. Mol Nutr Food Res. 52:53–61. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Vargas AJ and Burd R: Hormesis and synergy: Pathways and mechanisms of quercetin in cancer prevention and management. Nutr Rev. 68:418–428. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Kuo P-C, Liu H-F and Chao J-I: Survivin and p53 modulate quercetin-induced cell growth inhibition and apoptosis in human lung carcinoma cells. J Biol Chem. 279:55875–55885. 2004. View Article : Google Scholar : PubMed/NCBI

69 

Polyak K, Xia Y, Zweier JL, Kinzler KW and Vogelstein B: A model for p53-induced apoptosis. Nature. 389:300–305. 1997. View Article : Google Scholar : PubMed/NCBI

70 

Yoon KA, Nakamura Y and Arakawa H: Identification of ALDH4 as a p53-inducible gene and its protective role in cellular stresses. J Hum Genet. 49:134–140. 2004. View Article : Google Scholar : PubMed/NCBI

71 

Hussain SP, Amstad P, He P, Robles A, Lupold S, Kaneko I, Ichimiya M, Sengupta S, Mechanic L, Okamura S, et al: p53-induced up-regulation of MnSOD and GPx but not catalase increases oxidative stress and apoptosis. Cancer Res. 64:2350–2356. 2004. View Article : Google Scholar : PubMed/NCBI

72 

O'Connor JC, Wallace DM, O'Brien CJ and Cotter TG: A novel antioxidant function for the tumor-suppressor gene p53 in the retinal ganglion cell. Invest Ophthalmol Vis Sci. 49:4237–4244. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Ozaki T and Nakagawara A: p73, a sophisticated p53 family member in the cancer world. Cancer Sci. 96:729–737. 2005. View Article : Google Scholar : PubMed/NCBI

74 

Chien SY, Wu YC, Chung JG, Yang JS, Lu HF, Tsou MF, Wood WG, Kuo SJ and Chen DR: Quercetin-induced apoptosis acts through mitochondrial- and caspase-3-dependent pathways in human breast cancer MDA-MB-231 cells. Hum Exp Toxicol. 28:493–503. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Shen X, Si Y, Wang Z, Wang J, Guo Y and Zhang X: Quercetin inhibits the growth of human gastric cancer stem cells by inducing mitochondrial-dependent apoptosis through the inhibition of PI3K/Akt signaling. Int J Mol Med. 38:619–626. 2016.PubMed/NCBI

76 

Lin CW, Hou WC, Shen SC, Juan SH, Ko CH, Wang LM and Chen YC: Quercetin inhibition of tumor invasion via suppressing PKC δ/ERK/AP-1-dependent matrix metalloproteinase-9 activation in breast carcinoma cells. Carcinogenesis. 29:1807–1815. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Lee C-H, Hong H-M, Chang Y-Y and Chang W-W: Inhibition of heat shock protein (Hsp) 27 potentiates the suppressive effect of Hsp90 inhibitors in targeting breast cancer stem-like cells. Biochimie. 94:1382–1389. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Catanzaro D, Ragazzi E, Vianello C, Caparrotta L and Montopoli M: Effect of quercetin on cell cycle and cyclin expression in ovarian carcinoma and osteosarcoma cell lines. Nat Prod Commun. 10:1365–1368. 2015.PubMed/NCBI

79 

Chen X, Dong XS, Gao HY, Jiang YF, Jin YL, Chang YY, Chen LY and Wang JH: Suppression of HSP27 increases the anti tumor effects of quercetin in human leukemia U937 cells. Mol Med Rep. 13:689–696. 2016.PubMed/NCBI

80 

Neuhouser ML: Dietary flavonoids and cancer risk: Evidence from human population studies. Nutr Cancer. 50:1–7. 2004. View Article : Google Scholar : PubMed/NCBI

81 

Li W, Liu M, Xu YF, Feng Y, Che JP, Wang GC and Zheng JH: Combination of quercetin and hyperoside has anticancer effects on renal cancer cells through inhibition of oncogenic microRNA-27a. Oncol Rep. 31:117–124. 2014.PubMed/NCBI

82 

Moon JH, Eo SK, Lee JH and Park SY: Quercetin-induced autophagy flux enhances TRAIL-mediated tumor cell death. Oncol Rep. 34:375–381. 2015.PubMed/NCBI

83 

Seo HS, Ku JM, Choi HS, Choi YK, Woo JK, Kim M, Kim I, Na CH, Hur H, Jang BH, et al: Quercetin induces caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells. Oncol Rep. 36:31–42. 2016.PubMed/NCBI

84 

Yang FQ, Liu M, Li W, Che JP, Wang GC and Zheng JH: Combination of quercetin and hyperoside inhibits prostate cancer cell growth and metastasis via regulation of microRNA21. Mol Med Rep. 11:1085–1092. 2015.PubMed/NCBI

85 

Kandaswami C, Lee LT, Lee PP, Hwang JJ, Ke FC, Huang YT and Lee MT: The antitumor activities of flavonoids. In Vivo. 19:895–909. 2005.PubMed/NCBI

86 

Pratheeshkumar P, Budhraja A, Son YO, Wang X, Zhang Z, Ding S, Wang L, Hitron A, Lee JC, Xu M, et al: Quercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR- 2 regulated AKT/mTOR/P70S6K signaling pathways. PLoS One. 7:e475162012. View Article : Google Scholar : PubMed/NCBI

87 

Steiner J, Davis J, McClellan J, Enos R, Carson J, Fayad R, Nagarkatti M, Nagarkatti P, Altomare D, Creek K, et al: Dose-dependent benefits of quercetin on tumorigenesis in the C3(1)/SV40Tag transgenic mouse model of breast cancer. Cancer Biol Ther. 15:1456–1467. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Prasad S, Phromnoi K, Yadav VR, Chaturvedi MM and Aggarwal BB: Targeting inflammatory pathways by flavonoids for prevention and treatment of cancer. Planta Med. 76:1044–1063. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Yang H, Landis-Piwowar KR, Chen D, Milacic V and Dou QP: Natural compounds with proteasome inhibitory activity for cancer prevention and treatment. Curr Protein Pept Sci. 9:227–239. 2008. View Article : Google Scholar : PubMed/NCBI

90 

Gibellini L, Pinti M, Nasi M, Montagna JP, De Biasi S, Roat E, Bertoncelli L, Cooper EL and Cossarizza A: Quercetin and cancer chemoprevention. Evid Based Complement Alternat Med. 2011:5913562011. View Article : Google Scholar : PubMed/NCBI

91 

Zhou J, Fang L, Yao WX, Zhao X, Wei Y, Zhou H, Xie H, Wang LY and Chen LJ: Effect of quercetin on heat shock protein expression in HepG2 cells determined by SILAC. Zhonghua Zhong Liu Za Zhi. 33:737–741. 2011.(In Chinese). PubMed/NCBI

92 

Jeong JH, An JY, Kwon YT, Rhee JG and Lee YJ: Effects of low dose quercetin: Cancer cell-specific inhibition of cell cycle progression. J Cell Biochem. 106:73–82. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Mu C, Jia P, Yan Z, Liu X, Li X and Liu H: Quercetin induces cell cycle G1 arrest through elevating Cdk inhibitors p21 and p27 in human hepatoma cell line (HepG2). Methods Find Exp Clin Pharmacol. 29:179–183. 2007. View Article : Google Scholar : PubMed/NCBI

94 

Crespo I, García-Mediavilla MV, Gutiérrez B, Sánchez-Campos S, Tuñón MJ and González-Gallego J: A comparison of the effects of kaempferol and quercetin on cytokine-induced pro-inflammatory status of cultured human endothelial cells. Br J Nutr. 100:968–976. 2008. View Article : Google Scholar : PubMed/NCBI

95 

Nair MP, Mahajan S, Reynolds JL, Aalinkeel R, Nair H, Schwartz SA and Kandaswami C: The flavonoid quercetin inhibits proinflammatory cytokine (tumor necrosis factor alpha) gene expression in normal peripheral blood mononuclear cells via modulation of the NF-kappa beta system. Clin Vaccine Immunol. 13:319–328. 2006. View Article : Google Scholar : PubMed/NCBI

96 

Matsukawa Y, Nishino H, Okuyama Y, Matsui T, Matsumoto T, Matsumura S, Shimizu Y, Sowa Y and Sakai T: Effects of quercetin and/or restraint stress on formation of aberrant crypt foci induced by azoxymethane in rat colons. Oncology. 54:118–121. 1997. View Article : Google Scholar : PubMed/NCBI

97 

Shan BE, Wang MX and Li RQ: Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D1 and survivin expression through Wnt/beta-catenin signaling pathway. Cancer Invest. 27:604–612. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Yang K, Lamprecht SA, Liu Y, Shinozaki H, Fan K, Leung D, Newmark H, Steele VE, Kelloff GJ and Lipkin M: Chemoprevention studies of the flavonoids quercetin and rutin in normal and azoxymethane-treated mouse colon. Carcinogenesis. 21:1655–1660. 2000. View Article : Google Scholar : PubMed/NCBI

99 

Ferry DR, Smith A, Malkhandi J, Fyfe DW, deTakats PG, Anderson D, Baker J and Kerr DJ: Phase I clinical trial of the flavonoid quercetin: Pharmacokinetics and evidence for in vivo tyrosine kinase inhibition. Clin Cancer Res. 2:659–668. 1996.PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 38 Issue 2

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
Hashemzaei M, Delarami Far A, Yari A, Heravi RE, Tabrizian K, Taghdisi SM, Sadegh SE, Tsarouhas K, Kouretas D, Tzanakakis G, Tzanakakis G, et al: Anticancer and apoptosis‑inducing effects of quercetin in vitro and in vivo. Oncol Rep 38: 819-828, 2017
APA
Hashemzaei, M., Delarami Far, A., Yari, A., Heravi, R.E., Tabrizian, K., Taghdisi, S.M. ... Rezaee, R. (2017). Anticancer and apoptosis‑inducing effects of quercetin in vitro and in vivo. Oncology Reports, 38, 819-828. https://doi.org/10.3892/or.2017.5766
MLA
Hashemzaei, M., Delarami Far, A., Yari, A., Heravi, R. E., Tabrizian, K., Taghdisi, S. M., Sadegh, S. E., Tsarouhas, K., Kouretas, D., Tzanakakis, G., Nikitovic, D., Anisimov, N. Y., Spandidos, D. A., Tsatsakis, A. M., Rezaee, R."Anticancer and apoptosis‑inducing effects of quercetin in vitro and in vivo". Oncology Reports 38.2 (2017): 819-828.
Chicago
Hashemzaei, M., Delarami Far, A., Yari, A., Heravi, R. E., Tabrizian, K., Taghdisi, S. M., Sadegh, S. E., Tsarouhas, K., Kouretas, D., Tzanakakis, G., Nikitovic, D., Anisimov, N. Y., Spandidos, D. A., Tsatsakis, A. M., Rezaee, R."Anticancer and apoptosis‑inducing effects of quercetin in vitro and in vivo". Oncology Reports 38, no. 2 (2017): 819-828. https://doi.org/10.3892/or.2017.5766