Open access peer-reviewed chapter

Natural Products as Cytotoxic Agents in Chemotherapy against Cancer

Written By

Abdelmajid Zyad, Inass Leouifoudi, Mounir Tilaoui, Hassan Ait Mouse, Mouna Khouchani and Abdeslam Jaafari

Reviewed: 27 November 2017 Published: 29 December 2017

DOI: 10.5772/intechopen.72744

From the Edited Volume

Cytotoxicity

Edited by Tülay Aşkin Çelik

Chapter metrics overview

1,770 Chapter Downloads

View Full Metrics

Abstract

Nature continues to produce a great wealth of natural molecules endowed with cytotoxic activity toward a large panel of tumor cells. Some of these molecules are used in chemotherapy, and others have shown great anti-tumor and anti-metastatic potential in preclinical trials. This review discusses some examples of these molecules that have been studied in our laboratory and others. We report a differential cytotoxic activity of some monoterpenes (carvacrol, tymol, carveol, carvone, and isopulegol) against a panel of tumor cell lines. The carvacrol was the most cytotoxic molecule both in vitro and in vivo as demonstrated by preclinical studies using the DBA2/P815 mice model. On the other hand, polyphenols were also studied with respect to their cytotoxic effects. Interestingly, these compounds showed a prominent cytotoxic activity toward a panel of cancer cells with differential molecular mechanisms. In addition, we report a very strong antitumor efficacy of artemisinin, a sesquiterpen lactone from Artemisia annua, together with an antimetastatic potential as demonstrated by preclinical experiments. Furthermore, some of the molecular mechanisms involved in these effects are described.

Keywords

  • natural products
  • monoterpenes
  • polyphenols
  • artemisinin
  • cytotoxicity

1. Introduction

Natural drugs have formed the basis of traditional medicine systems that have been used for centuries by different cultures [1]. An immense number of these natural sources and their isolated components have demonstrated beneficial therapeutic effects, such as anticancer, antioxidant, immunomodulatory, antimicrobial, and anti-inflammatory properties [2, 3]. Studies reported that plant-derived drugs represent about 25% of the American prescription drug market [4]. Also, natural products play an important role in the health care of 20% of the world’s people who mainly reside in developed countries and 119 chemicals compounds, derived from 90 plant species, can be considered as important drugs in many countries [5]. Based on a recent review, from 79 Food and Drug Administration anticancer and antiviral approved drugs from 1983 to 2002, 9 of them were isolated directly from plants and 21 among them were natural-products-based drug. Furthermore, between 39 conventional anticancer molecules, 13 of them were derived on a pharmacophore obtained from natural drugs [5, 6]. Actually, nature continues to be an attractive source of new molecules discovery due to important chemical diversity of the thousands of plant, animal, marine organisms, and micro-organism species. Today, about 60% of drugs are of natural origin [7] (Tables 13).

DrugUtilizationMechanism of actionSource
AspirineAnalgesic, anti-inflammatory, anti-pyrticInhibition of cyclo-oxygenasePlant
AtropinePupil dilatatorAnti-cholinergic on muscarinic receptorsPlant
CafeineStimulatingAntagonist of adenosine receptorsPlant
CodeineAnalgesic, anti-tussiveAntagonist of opoide receptorsPlant
DigoxineCardiotonicInhibition of membrane pump N+/K+ ATPasePlant
EugenolTouth painReduction of sensorial nerve excitabilityPlant
MorphineAnalgesicAntagonist of opoide receptorsPlant
PilocarpineGlaucomaAntagonist of muscarinic receptorsPlant
QuinineProphylaxis of malariaInhibition of protein synthesisPlant
TaxolAnticancerAntimitoticPlant
PenicillineAntibioticInhibition of cell membraneMicro-organism
TatracyclineAntibioticInhibition of protein synthesisMicro-organism
Cyclosporine AImmunosuppressorInhibition of lymphocytes T proliferationMicro-organism
AurantosidesAntifungalInhibition of tubulin polymerizationMarine organism
Spongistatine 1AntifungalInhibition of tubulin polymerizationMarine organism
ManoalideAnalgesic, anti-inflammatoryInhibition of phospholipase A2Marine organism

Table 1.

Some natural drugs derived from plants, micro-organisms, or marine organisms [8].

DrugUtilization
ActinomycineGerminale cells tumor, sarcoma
BléomycineCervix cancer, Germinale cells tumor, and neck
DaunomycineLeukemia
DoxorubicineLymphoma, breast, lung and ovarian cancer, sarcoma
EpirubicineBreast cancer
IdarubicineLeukemia and breast cancer
Mitomycine CColorectal, gastric, anal, and lung cancer
StreptozocineGastric and endocrine tumors

Table 2.

Some anticancer drugs derived from micro-organisms [8].

DrugUtilizationMechanism of action
CitarabineLeukemia, lymphomaInhibition of DNA synthesis
Bryostatine 1Experimental phaseActivation of protein kinase C
Dolastatine 10Experimental phaseInhibition of microtubules and pro-apoptotic effect
Ecteinascidine 743Experimental phaseAlkylation of DNA
AplidineExperimental phaseInhibition cell cycle progression
Halicondrine BExperimental phaseInteraction with tubuline
DiscodermolideExperimental phaseStabilization of tubuline
CryptophycineExperimental phaseHyperphosphorylation of Bcl-2

Table 3.

Some anticancer drugs derived from marine organisms [8].

DrugsUtilization
VincristineLeukemia, lymphoma, breast cancer, and lung cancer
VinblastineLymphoma, kidney cancer, germinal cells cancer, and breast cancer
PaclitaxelBreast cancer, ovarian, lung, and d’ovaire, de poumon, bladder, and neck cancer
DocetaxelBreast and lung cancer
TopotecanOvarian and lung cancer
IrinotecanColorectal and lung cancer

Table 4.

Anticancer drugs derived from plants [8].

Several molecules used as conventional chemotherapy are of natural origin. Some of these molecules and their use are described in Tables 2 and 3.

Advertisement

2. Phytotherapy and cancer

2.1. Generalities

There is a numerous plants involved in the prevention and/or treatment of cancer. As for other diseases, many anticancer drugs are derived from plants (Table 4). Studies reported that more than 200 drugs are of herbal origin. The vinca-alcaloids and the taxans are the main groups, which occupy an important place in anticancer chemotherapy.

2.2. Examples of natural products with important cytotoxic activity

2.2.1. Cytotoxic activity of some natural monoterpenes

The chemical composition of plant-extracts is known for being very rich and diversified. Thus, a single extract may contain more than hundreds of interactive biomolecules [9]. Therefore, finding and discovering those responsible for the biological Activity become essential. Many monoterpenes, such as eugenol, have been described in the literature to have a wide range of important biological activities [10]; it possesses in vitro and in vivo antiviral activity against human herpesvirus [11]. Carvone promoted protection of 75–87.5% against convulsions at 300–400 mg/kg [12]. Isopulegol and carvone showed significant bactericidal and fungicidal activities [13]. Also, the combination of these molecules between them or with conventional molecules could have a synergistic effect [14, 15]. Furthermore, carvacrol, extract of thyme essential oil, is one of natural products with important biological activities. It has been reported to have an important antitumor effect [9, 16]. Here, we present a summary of our findings [17] on the cytotoxic activity as well as their molecular mechanisms of six natural monoterpenes compounds (carvacrol, thymol, carveol, carvone, eugenol, and isopulegol).

2.2.1.1. In vitro cytotoxic effect of the products against a panel of target cells

The antitumor activity of the products was evaluated against the following five tumor cell lines: P-815, K-562, CEM, MCF-7, and MCF-7 resistant to gemcetabine (MCF-7-gem). The results are summarized in Figure 1, which shows that the cytotoxic effect depends on the nature of the products as well as on the target cell lines. In general, the effect of the products is dose-dependent. Moreover, the cytotoxic activity of carvacrol, thymol, carveol, carvone, eugenol, and isopulegol is more important against P-815 and CEM tumor cell lines compared to the other tested cell lines. The carvacrol is the most cytotoxic compared to other compounds. Against P-815, K-562 and CEM cancer cell lines, eugenol, carveol, and carvone exhibit also a strong cytotoxic activity. The IC50 values are ranging from 0.09 to 0.24 μM (Table 5). Nevertheless, those compounds showed a less effect toward MCF-7 and very lowest one against MCF-7-gem cancer cell lines as demonstrated by the IC50 values ranging from 0.26 to 0.87 μM. Comparing the activity of thymol and isopulegol on the tumor cell lines studied, it shows that P-815 is the most sensitive with an IC50 = 0.15 and 0.09 μM, respectively. Importantly, acquired resistance to gemcetabine by MCF-7 cell line was linked with a development of resistance to thymol, carveol, carvone, and eugenol but not to isopulegol or carvacrol (Table 5).

Figure 1.

Cytotoxic effect of carvacrol (A), thymol (B), carveol (C), carvone (D), eugenol (E) and isopulegol (F) against different tumor cell lines: P815 (♦), CEM (◾), K562 (_), MCF-7 (▴) and MCF-7 gem (*).

ProductP815CEMK-562MCF-7MCF-7/gem
Carvacrol0.0670.0420.0670.1250.067
Thymol0.150.310.440.48
Carveol0.110.110.130.260.45
Carvone0.160.110.170.630.91
Eugenol0.100.090.240.410.87
Isopulegol0.090.110.130.25

Table 5.

IC50 (μM) of the tested monoterpenes against different target cell lines.

2.2.1.2. Synergy

Our results demonstrate that the combination of natural monoterpene with MTX or Cis showed a synergistic effect at used concentrations (IC20) of each tested molecules (monoterpenes, cisplatine, and methotrexate). The interactions between these molecules exhibit a cell lysis ranging between 53 and 62%. Furthermore, a slight cytotoxicity was shown after the combinations between monoterpene-cisplatin and monoterpene-methotrexate (Table 6).

CombinationFaCI
C-MTX54.90.17
C-Cis56.60.01
T-MTX610.14
T-Cis57.60.01
Cl-MTX53.30.17
Cl-Cis57.90.01
Cn-MTX51.20.17
Cn-Cis58.50.01
E-MTX58.60.15
E-Cis55.90.01
I-MTX58.50.15
I-Cis62.30.01

Table 6.

Affected fraction (Fa) and combination index (CI) of molecule combinations.

2.2.1.3. Effect of carvacrol, thymol, carveol, carvone, eugenol, and isopulego on the cell cycle progression

At the molecular level, carveol- and carvacrol treatment-induced cell cycle arrest in S phase. Nevertheless, thymol and isopulegol stopped it in G0/G1 phase. Regarding the eugenol and carvone, they have no effect cell cycle progression (Figure 2).

Figure 2.

Effect of the tested products on cell cycle progression. The samples were analyzed using a FACStar plus flow cytometer and the WinMDI software. Results are the mean ± SEM of three tests. (A) Carvacrol, (B) thymol, (C) carveol, (D) carvone, (E) eugenol, and (F) isopulegol.

2.2.1.4. In vivo antitumor effect of carvacrol

Our experimental model was based on the use of the P-815 tumor-bearing DBA-2 mice to investigate the cell-killing induced by carvacrol. Experiments were carried out by oral administration (gavage) of carvacrol dissolved in vegetal oil to 6- to 8-week-old DbA-2/6 mice (6 mice for each group) (Orleans, France) weighting 18–22 g for 7 days. The tumor volume was measured for up to 30 days. The tumor volume at day n, (Tvn) was calculated using the formula: Tv = (l × W2)/2, where l equals the length of the tumor and W the width, as described by Yoshikawa [18]. Interestingly, during the first 18 days, there was no statistical difference (p < 0.94) in the volumes of the tumors in all the groups of mice, including the control group (0.4–0.5 ± 0.1 cm3). Nevertheless, after 18 days, the tumor volume was reduced for the treated groups; this decrease occurred more rapidly in the group “C” who received 100 mg/kg/day than the group “B” treated with 50 mg/kg/day (p < 0.05 at day 21th). Compared to untreated group, the tumor volume increased quickly reaching 1.5 cm3 at 23rd day. Furthermore and importantly, the tumor volume reduction was accompanied by a notable increase of mice survival (Figure 3). The antitumor activity of carvacrol has not been has not been widely discussed in the literature. At the best of our knowledge, this is the first study that reported the oral administration of carvacrol for successive 7 days significant decrease tumor volume, body weight loss, and delayed mortality (data not shown). These results corroborate those published by Karkabounas who demonstrated that carvacrol exhibited 30% reduction of 3,4-benzopyrene carcinogenic activity in vivo [19].

Figure 3.

In vivoantitumor effect of carvacrol.

Studies were carried out by gavage of carvacrol dissolved in vegetable oil to mice (6–8 week-old) for 7 days. Group “A” (untreated) treated with 100 μl/day of vegetal oil only. Groups “B” and “C” received 50 and 100 mg/kg/day of carvacrol dissolved in 100 μl of vegetal oil, respectively. Mice were weighted and the tumor volume was calculated by measurement of the width (W) and the length (l) for three times a week up to day 30. The tumor volume at day n (TVn) was measured using the following formula: TV = (l × W2)/2. The experiments are the mean ± SEM of two tests.

2.2.1.5. Discussion

Monoterpenes (carvacrol, thymol, carveol, carvone, eugenol, and isopulegol) have been found to exert antitumor effect. In fact, eugenol was described to exhibit cell death by apoptosis in mastocyte [20] and melanoma cells [21]. Also, it has been demonstrated not to be mutagenic neither carcinogenic [22]. Carveol has chemopreventive activity against mammary cancer when fed during the initiation phase [23]. Carvone prevents chemically induced lung and for stomach carcinoma development [24]. Carvacrol and thymol significantly reduced the level of DNA damage induced in K-562 cells by the strong oxidant H2O2 [25]. Furthermore, carvacrol has an important in vitro antitumor effect against tumor cell lines like Hep-2 [26] and A-549 [16, 27]. As shown in Table 5, the monoterpenes studied induced a differential cytotoxic activity against a panel of tumor cell lines. P-815 and CEM cell lines are the most sensitive targets to all tested molecules. Although the effects of these products are dose-dependent, the carvacrol is the most cytotoxic molecule as revealed by the IC50 values (Table 5). Importantly, unlike isopulegol and carvacrol, the acquired resistance to gemcitabine by MCF-7 tumor cell line was associated with a resistance to thymol, carveol, and carvone. Taken together, these results suggest that these compounds could have a similar pathway. The differential sensitivity of the studied monoterpenes toward MCF-7 and MCF-7-gem could be linked to the expression level of ribonucleotide reductase subunit R1 [28]. Furthermore, the cell cycle analysis showed that carveol- and carvacrol treatment-induced cell cycle arrest in S phase when thymol and isopulegol stopped it in G0/G1 phase. Nevertheless, the eugenol and carvone have no effect cell cycle progression. These results suggest that the molecular mechanistic pathway of the cytotoxicity exhibited by those molecules is more complicated and is not related only with the cell cycle. It was reported that monoterpenes decreased expression of cyclin-dependent kinase cdk4, cyclin D1, and cdk2 and increased expression of cyclin E and cdk inhibitor p21 [29]. Furthermore, geraniol, farnesol, and isoprenoids perillyl alcohol exhibited a G0/G1 cell cycle arrest by increasing in the expression level of p27 (Kip1) and the cyclin kinase inhibitor proteins p21 (Cip1) and a decreasing in cyclin B1, cyclin A, and cyclin-dependent kinase (Cdk2) expression [14]. Interestingly, our results demonstrate that the interaction of tested monoterpenes at lowest concentration (IC20) with the conventional anticancer molecules (cisplatine and methotrexate) exhibited a synergistic activity (Table 6). Thus, this combination may reduce the toxicity of the conventional chemotherapy drugs by reducing their doses. These results are supported by previous findings reporting that when combined to isoprenoids perillyl alcohol, farnesol, and geraniol showed an additive antiproliferative activity against the human pancreatic cancer cell line MIA PaCa-2 [14]. Also, Chander et al. reported that in chemotherapy of breast tumors, the combination of limonene, natural monoterpene, and 4-hydroxyandrostenedione, inhibitor of aromatase, was more effective than each drug used alone [30]. Interestingly, in our study, we reported a synergistic effect and not an additive one suggesting that only low doses of each monoterpene combined with tolerable low doses of methotrexate or cisplatine (IC20) showed an important effect (60% lysis).

2.2.2. Polyphenols: a potent cytotoxic molecules

Natural polyphenols have received increasing interest in the human health due to their benefit effects against several diseases attributed particularly to their antioxidant activity [31]. Beside their well-known and effective antioxidant activity [32, 33], several polyphenols shown a high cytotoxic effect against cancer cell lines through targeting cellular and molecular processes involved in cancer progression and metastasis. The antitumor potential of these active ingredients is due to their effect as modulators of oxidative stress [34], apoptosis inducers [35] cell proliferation inhibitor [36], tumor cell cycle blockers [37], and angiogenesis/metastasis suppressors [38]. These bioactive compounds have shown promising antitumor properties in both in vitro and in vivo interventions [39, 40]. These structural variations may be responsible for their various health benefits, including antioxidant [41], and anti-proliferative mechanisms, as well as regulation of key signaling protein and enzyme functions [42], and as promising immunostimulating effect on normal immune cells [43]. The relationship among natural polyphenols, antitumor activity, and cancer was identified by various studies on the ability of these compounds to act as cancer chemopreventive and/or chemotherapeutic agents [44]. In this purpose, a variety of natural polyphenols have been identified to interfere with carcinogenesis particularly through apoptosis induction and the modulation of oxidative stress [45, 46].

2.2.2.1. Polyphenols and apoptosis induction

Large number of studies has focused on the ability to introduce apoptosis on cancer therapy under cellular control conditions [47, 48]. The intrinsic and extrinsic molecular pathways involved in the regulation of the apoptotic process have recently been evaluated and give promising results. Several proapototic receptors have been selectively developed activating the intrinsic pathway, particularly including the antiapoptotic proteins, the Bcl-2 family proteins, and the p53 signaling pathways [49, 50, 51]. In this purpose, polyphenols could inhibit tumor cell proliferation via the programmed cell death (apoptosis) using both intrinsic and extrinsic cell pathways. As reported, polyphenols such as EGCG: (−)-epigallocatechin-3-gallate, resveratrol, naringenin, quercetin, hydroxytyrosol, and curcumin, through different intrinsic signaling pathways from mitochondrial intermembrane space, may inhibit NF-κB-dependent signal related to proliferation and survival [52], cause cell cycle arrest through upregulation of p53 [53], stabilize and activate the tumor suppressor gene p53 [54], and downregulate the expression of Bcl-2, and Bcl-XL anti-death proteins, favoring apoptosis induction via the activation of multiple caspases activity and cytochrome-c (cyt-c) [55, 56]. These polyphenols have been shown to promote apoptosis in different cancers particularly breast, lung, prostate, leukemia, colon, cervical, or melanoma [57, 58]. In breast cancer cells, naringenin demonstrated anti-estrogenic activity in estrogen-rich status and estrogenic activity in estrogen-deficient status [59]. Additionally, few early studies suggested that gavage of polyphenols in green tea (EGCG), even at low doses, prevented colon carcinogenesis by inhibiting metastasis and angiogenesis through apoptosis induction [60]. Few years ago, our research group has published an article [61] on natural polyphenols extracted from olive mill waste (OMW) and their implication in anticancer activity, where the in vitro cytotoxic and apoptotic assays involving several phenolic compounds found in those specific phenolic extracts (particularly including, quercetin, naringenin, apigenin, hydroxytyrosol, oleuropein, and its derivatives) have been discussed. The in vitro cytotoxic effect of olive mill waste extracts was evaluated using the MTT assay (methyl tetrazolium test). The IC50 values ranged from 4.8 to 7.6 μg/ml (Table 7), which demonstrate an effective cytoxicity of these phenolic compounds at low doses. We have demonstrated that the cytotoxic potential of these phenolic extracts was exhibited via apoptosis induction by DNA fragmentation test using agarose gel electrophoresis (Figure 4A). DNA isolated from MCF7 tumor cells was treated with OMW extracts at concentrations corresponding to the IC50 values and incubated for 24 h. To confirm the cell-death mechanism of these natural extracts, the apoptosis analysis was performed using the Annexin V biotin-streptavidin FITC test. We reported that phenolic extracts induced significantly apoptosis (Figure 4B) compared to untreated cells (Figure 4C). Interestingly, those polyphenols have not shown any cytotoxic effect against human normal cells (PBMC) (Figure 5). Hence, it triggered apoptosis in a dose-dependent manner on a breast cancer cell line (MCF-7) without any effects on normal cells by enhancing the viability with 12–16% in 48 h, compared to methotrexate (conventional cytotoxic drug), which suppressed 20% viability of these cells.

SamplesS1S2S3S4S5
IC50 (μg/ml)6.95 ± 0.155.3 ± 0.14.75 ± 0.057.75 ± 0.155.3 ± 0.2

Table 7.

IC50 values of the cytotoxicity of OMW polyphenolic extracts against MCF-7 breast cancer cell line.

Figure 4.

Apoptosis-induction analysis in MCF-7 cell line. (A) Phenolic extracts induced DNA fragmentation was detected by agarose gel electrophoresis of DNA isolated from MCF-7 tumor cells. Cells were incubated for 24 h with OCE (corresponding to IC50 concentrations). S1 to S5: Olive mill waste extracts samples. Positive control (WM): DNA weight marker. DNA of untreated cells was used as negative control (NC). (B) Annexin V biotin-streptavidin FITC test. MCF-7 tumor cells (2 × 106 cells) were treated with 25 μg/ml of OCE and incubated for 24 h. The assay is based on the ability of Annexin V (green fluorescence) to bind to the phosphatidylserine exposed on the surface of cells undergoing apoptosis. Cells cultured in a medium without serum were used as a positive control (C).

Figure 5.

Cytotoxic effect of OMW polyphenolic extracts on normal human peripheral blood mono mononuclear cells (PBMC) from normal donors.

Taken together, these data showed the differential and selective cytotoxic effect of natural polyphenols. In this sense, Miccadei et al. [43] have shown that polyphenolic extracts from the edible part of artichoke (Cynara scolymus L) may selectively inhibit the growth of human hepatoma cells with little or no toxicity against normal hepatocytes cells based on their differential redox status. Interestingly, the authors have shown that Artichoke extracts exhibit a pro-oxidant activity in breast cancer cells and an antioxidant effect on normal hepatocytes. Moreover, some flavanols may have a significant effect on cytokine release from both unstimulated and lipopolysaccharides-stimulated PBMCs [62]. Oral administration of naringenin suppressed breast cancer metastases after surgery by modulating the host immunity [63].

2.2.2.2. Role of polyphenols in therapy-induced senescence

Cellular senescence is a physiological process of irreversible cell-cycle arrest that contributes to various physiological and pathological processes of aging. It is an alternative and a novel therapeutic strategy to the cytotoxic treatment which leading to cytostasis approach target for aging and aging-related diseases [64]. Although senescence cells have irreversibly lost their capacity for cell division, they are still viable and remain metabolically active [65]. Prosenescence is usually associated with telomere erosion after repeated cell divisions and occurs in response to abnormal oncogenic signaling, oxidative stress, and DNA damage [66]. To this purpose, natural compounds targeting the epigenetic control of senescence are under investigations to develop additional prosenescence cancer therapeutic strategies [67]. Several anticancer polyphenolic compounds from fruit and vegetables have been shown to be potential chemopreventive and anticancer bioactive compounds [68] to induce cellular growth arrest through the induction of a ROS-dependent premature senescence. Among them, 20(S)-ginsenoside Rg3, a compound extracted from ginseng, and bisdemethoxycurcumin, a natural derivative of curcumin, caused senescence-like growth arrest and increased ROS production, respectively, in human glioma cells [69] (and human breast cancer cell [70]. Therefore, high doses of polyphenolic extracts from artichoke may induce apoptosis and decrease cell proliferation of the human breast cancer cell line, MDA-MB231 via the induction of premature senescence through epigenetic and ROS-mediated mechanisms [71]. Importantly, the authors have shown that Artichoke extracts have a pro-oxidant activity in breast cancer cells [72] and an antioxidant effect on normal hepatocytes [43]. Therefore, it has been hypothesized that Polyphenolic artichoke extracts could selectively inhibit the tumor cells growth with no cytotoxicity on healthy cells related on their differential cellular redox status. Furthermore, treatment with a low dose of resveratrol exhibits its chemopreventive and anticancer activities by induction of premature senescence in lung cancer cells. This event is associated with an increase in ROS generation and DNA double strands break through the up-regulation of NAPDH oxidase-5 expression [73]. The inhibitory effect of resveratrol was verified in vitro and in vivo, respectively, on gastric cells cancer and nude mice xenograft model. Low doses of resveratrol treatment arrested gastric cancer cells in the G1 phase and led to senescence instead of apoptosis and exerted inhibitory effect on gastric development and significantly decreased the fraction of Ki67-positive cells in the nude mice tumor specimens [74]. Interestingly, Resveratrol and quercetin administers in subapoptotic doses can induce senescence-like growth arrest in glioma tumors treatment [75]. The concept of prosenescence therapy has emerged over the past few years as a novel therapeutic approach to treat cancers, which may be viewed either as an independent anticancer approach or as a combined strategy with conventional chemo/radiotherapy [76]. In a neoadjuvant setting, prosenescence therapy could be also used with traditional treatments in order to reduce tumor mass before surgery; whereas in adjuvant therapy, the engagement of prosenescence could be helpful in reducing the statistical risk of cancer relapse [77]. Although the effective potential of polyphenol in anticancer therapy as well as their other various beneficial effects on human health, the poor bioavailibility of these active ingredients still a pending issue which limits their potential effects and their incorporation on western medicine. Further aimed challenging studies are needed to improve the absorption, distribution, and metabolism in order to develop the in vivo use and in clinical interventions.

2.2.3. Artemisinin: a cytotoxic molecule with medical interests

Artemisinin, the active component of Qinghao (Chinese name of Artemisia annua L.) was discovered in 1972 by Professor Tu’s team [78], a discovery that was recognized by her receipt of the Nobel Prize in medicine in 2015. Artemisinin belongs to the family of sesquiterpene lactone with an endoperoxide bridge found to be important for its activity. The yield of artemisinin that can be extracted from Artemisia annua ranges from 0.01 and 0.8% of the dry weight [79]. This amount of extraction represents a serious limitation on the drug commercialization. Consequently, genetic engineering techniques have been used with the aim to improve the production of artemisinin in cell plant cultures and in transgenic plants as well.

2.2.3.1. In vitro cytotoxic properties of artemisinin

A significant cytotoxicity of artemisinin against tumors has been recently documented. It suggests that artemisinin, commonly used against malaria, can be used to prevent and treat cancer [80, 81, 82, 83]. It is a relatively safe drug, with known pharmacokinetics and pharmacodynamics studies. In fact, in vitro work on the effects of artemisinin, at different concentrations, shows that artemisinin significantly inhibited growth and colony formation of human hepatocellular carcinoma cells through inducing apoptosis pathway [84]. Artemisinin at 20 μmol/l for 24 or 48 h of exposure inhibits growth and cell viability of human ovarian carcinoma cell lines (OVCAR-432 and SK-OV-3) [85]. Moreover, a recent study from our laboratory demonstrated that artemisinin has a differential effect on cancer cells. In fact, artemisinin induced lysis on the murin mastocytoma cancer cell line (P815) with IC50 = 12 μM and on kidney adeno-carcinoma cell line of hamster with IC50 = 52μM [86]. Furthermore, artemisinin was described to possess an anticancer effect on breast, lung, prostate, colon, leukemia, and other cancer cell types. Despite its efficacy, artemisinin has pharmacokinetic limitations such as poor bioavailability and low solubility in water or oil [87]. Thus, it was developed with semi-synthetic derivatives drugs to overcome some of these problems. So far, semi-synthetic derivative of artemisinin such as artesunate and dihydroartemisinin have been demonstrated to exert an important in vitro anticancer activity against different cancer cell lines. In breast cancer cells (MCF-7), artemisinin is less active, and the activity in these cells can be estrogen receptors-mediated (ERβ and ERα) which are implicated in cell proliferation [88]. In metastatic nasopharyngeal carcinoma cell lines (CNE-2 and CNE-1), the less sensitivity to artemisinin seems to be related to the over expression of polycomb complex protein BMI-1 [89]. Previously, Efferth et al. described a profound cytotoxic activity of artesunate, a semi-synthetic derivative of artemisinin, against 55 cancer cell lines of the U.S. National Cancer Institute with IC50 ranged from 246 nM to 100 μmM, by activating the expression of CDC25A and EGFR genes in cancer cells [83]. Another study by Efferth et al. showed that artesunate cytotoxicity on isogenic Saccharomyces cerevisiae with defined genetic defects, involved the implication of two putative target genes, BUB3 and CLN2 [82]. Furthermore, it was described that the anticancer activity of artesunate, arteether, and artemether (semi-synthetic derivative of artemisinin) is associated with the basal mRNA expression of 464 genes linked to proliferation of cells [90]. Generally, artemisinin molecules have been described to be more cytotoxic against cancer than normal healthy cells [86, 91], since normal cells contain significantly less free iron than cancer cells. In general, cancer cells, express more cell surface transferrin receptors and uptake significantly more iron than do normal cells [91].

Several studies have tried to explain, at molecular level, the mechanism of its anti-cancer action. A study on HL-60 cancer cell line demonstrated that rapid production of reactive oxygen species is associated with cell death by apoptosis after artemisinin treated cells [92]. Other factors such as endoplasmic reticulum stress and calcium metabolism can also be associated with the anticancer activity of artemisinins [93, 94]. Endoplasmic reticulum seems to be a possible site for artemisinin action, in HepG2 cancer cell line a derivative fluorescence accumulates preferentially in the endoplasmic reticulum as described by Crespo et al. [95].

Artemisinin has been described to induce apoptosis effect [86, 96, 97], as well as cell cycle arrest, especially at G0/G1 cell cycle transition phase [89, 98]. Multiple lines of evidence suggest that the apoptotic pathway could be due to intra and/or extra-mitochondrial mode of action, and the involvements of iron/heme as well [81, 99]. Two mechanistic pathways have been frequently described to explain the apoptotic effect of artemisinin, vascular endothelial growth factor decrease [100, 101, 102], and nuclear factor-kappa B inhibition [103, 104]. Recently, other processes have also been illustrated in different cancer cell types, by the involvement of NOXA [105], mitogen-activated protein kinase (MAPK) [106], Wnt/β-catenin [107], surviving [108], COX [109], c-MYC oncoprotein [93, 110], and epidermal growth factor [111]. Furthermore, it was also reported that the sensitivity to artemisinin action was related to the expression level of proapoptotic (Bax) and antiapoptotic (Bcl2) genes [112]. Also, artemisinin role in the inhibition of cancer is postulated to be associated with direct DNA damage [113] or indirectly in tumor cells involving a cascade of signaling pathways in many hallmarks of cancer [114]. Taken together, these results could explain the apoptotic pathway induction by artemisinin on tested cancer cells [101, 102, 115, 116]. However, we have also reported the possibility of the involvement of another cell death process of artemisinin; probably necrosis [86]. Artemsinin-induced necrosis remains not well documented and may be linked with the increasing level of ATP, defective apoptotic pathways, reactive oxygen species-independent mechanism of programmed cell death and cancer cell line type [86]. Furthermore, we have described that artemisinin interacted synergistically and additively with vincristin to reduce cancer cell proliferation [86], suggesting a possible use of artemisinin as an adjuvant to treat cancer.

2.2.3.2. In vivo anti-tumor and antimetastatic effects

Artemisinin treatment in oral route at 80 mg/kg considerably reduced the tumor volume growth of P815/DBA2 mice as described by our team [86]. In HepG2 and Hep3B human hepatoma mouse xenograft, artemisinin administered at 50 or 100 mg/kg/day delayed tumor onset, respectively, by 30 and 39.4% [117]. Also, in another study, artemisinin reduced tumor growth at 50% on day 20, when injected intraperitoneally at a concentration of 2.8 mg/kg/day on mammary gland ductal carcinoma in mice [118]. Inhibition of tumor growth and anti-angiogenic effect in MCF-7 mouse xenograft after subcutaneous treatment with artemisinin at dose 100 mg/kg/day for 2 weeks was reported [98]. Interestingly, artemisinin exhibited an anti-metastatic effect [116]. In fact, these authors showed that after orally artemisinin treatment with 50 mg/kg, a reduction of 63.5% of lung metastasis and lymph node metastases decrease in cervical and mediastinal lymph nodes, as well as an inhibition of lymphangiogenesis by 63% of mice. Artemisinin also exhibited inhibitory effects in lung tumor metastasis by 51.8 and 79.6% for 50 and 100 mg/kg/day, respectively. Furthermore, it was described that artesunate given in the drinking water at 167 mg/kg/day suppressed growth of Kaposi’s sarcoma-IMM xenograft tumors in nude mice [119]. The antimetastatic effect of artemisinin seems to be associated with the expression of metalloproteinase genes and their effect on αvβ3 integrins [120]. Moreover, the decrease of MMP2 with an increase of TIMP-2 in HepG2 and SMMC772 cancer cell lines after artemsinin treatment were reported [121]. Interestingly, the antimetastatic effect of artemisinin could be triggered by enhancing Cdc42 and E-cadherin activation [121]. However, in highly metastatic cancer such as nasopharyngeal cancer (CNE-1,CNE-2 cancer cell lines), artemisinin seems to have a low response due to the overexpression of BMI-1 gene that makes these cancer cells more sensitive to artemisinin drug [122]. In highly metastatic MDA-MB-231 breast tumor cells, artesunate induced resistance as described by Beatrice Bachmeier et al. (2011). This resistance was induced by the activation of transcription factors NF-κB and AP-1 [123]. Another study showed suppression of invasive and metastatic non-small cell lung cancer after artesunate treatment by the inhibition of urokinase-type plasminogen activator (u-PA), and matrix metalloproteinases (especially MMP-2 and MMP-7) transcription [10].

Advertisement

3. Conclusion

Nature continues to produce a great wealth of natural molecules endowed with cytotoxic activity towards a large panel of tumor cells. More than 60% of these molecules such as vinblastine, vincristine, etoposide, teniposide, taxol, navelbine, and camptothecin are used in chemotherapy and others have shown great anti-tumor and anti-metastatic potential in preclinical trials [124, 125]. Other natural product (i.e., Romidepsin 14, Omacetaxine mepesuccinate) [126] and natural product-derived drugs (i.e., metformin, metformin Polyphenon E, retinoids, soy isoflavones) [127] are in clinical trials. This chapter discusses some examples of these molecules (carvacrol, thymol, carveol, carvone, eugenol, isopulegol, and artemisinin) as well as polyphenols extract that have been studied in our laboratory. Other natural compounds are also under studies and remain promising. It is clear that if we understand the molecular mechanisms of the various interactions between these cytotoxic molecules on the one hand and the tumor cells in their tumoral environments on the other hand, we can develop new therapeutic modalities to overcome the side effects of these molecules and to fight cancer.

Advertisement

Acknowledgments

This work was supported by the Lalla Salma Foundation: Prevention and treatment of cancer-Rabat-Morocco (Research Project N° 09/AP 2013).

References

  1. 1. Cragg GM, Newman DJ. Natural products: A continuing source of novel drug leads. Biochimica et Biophysica Acta (BBA): General Subjects. 2013;1830(6):3670-3695
  2. 2. Tilaoui M, Mouse HA, Jaafari A, Zyad A. Comparative phytochemical analysis of essential oils from different biological parts of Artemisia herba alba and their cytotoxic effect on cancer cells. PLoS One. 2015;10(7):e0131799
  3. 3. Oliveira AH, de Oliveira GG, Carnevale Neto F, Portuondo DF, Batista-Duharte A, Carlos IZ. Anti-inflammatory activity of Vismia guianensis (Aubl.) Pers. extracts and antifungal activity against Sporothrix schenckii. Journal of Ethnopharmacology 2017;195:266-274
  4. 4. Grifo F, Newman D, Fairfield AS, Bhattacharya B, Grupenhoff JT. The origins of prescription drugs. In: Biodiversity and Human Health. 1997. pp. 131-163
  5. 5. Arvigo R, Balick MJ. Rainforest Remedies: One Hundred Healing Herbs of Belize. Wisconsin: Lotus Press; 1993
  6. 6. Cooper GM. The Cancer Book: A Guide to Understanding the Causes, Prevention, and Treatment of Cancer. MA, USA: Jones & Bartlett Learning; 1993
  7. 7. Cragg GM, Newman DJ. Discovery and development of antineoplastic agents from natural sources. Cancer Investigation. 1999;17(2):153-163
  8. 8. Da Rocha AB, Lopes RM, Schwartsmann G. Natural products in anticancer therapy. Current Opinion in Pharmacology. 2001;1(4):364-369
  9. 9. Jaafari A, Mouse HA, Rakib EM, Tilaoui M, Benbakhta C, Boulli A, Abbad A, Zyad A. Chemical composition and antitumor activity of different wild varieties of Moroccan thyme. Revista Brasileira de Farmacognosia. 2007;17(4):477-491
  10. 10. Rasheed SAK, Efferth T, Asangani IA, Allgayer H. First evidence that the antimalarial drug artesunate inhibits invasion and in vivo metastasis in lung cancer by targeting essential extracellular proteases. International Journal of Cancer. 2010;127(6):1475-1485
  11. 11. Benencia F, Courreges MC. In vitro and in vivo activity of eugenol on human herpesvirus. Phytotherapy Research. 2000;14(7):495-500
  12. 12. Zheng G-Q, Kenney PM, Lam LK. Sesquiterpenes from clove (Eugenia caryophyllata) as potential anticarcinogenic agents. Journal of Natural Products. 1992;55(7):999-1003
  13. 13. Naigre R, Kalck P, Roques C, Roux I, Michel G. Comparison of antimicrobial properties of monoterpenes and their carbonylated products. Planta Medica. 1996;62(03):275-277
  14. 14. Wiseman DA, Werner SR, Crowell PL. Cell cycle arrest by the isoprenoids perillyl alcohol, geraniol, and farnesol is mediated by p21Cip1 and p27Kip1 in human pancreatic adenocarcinoma cells. The Journal of Pharmacology and Experimental Therapeutics. 2007;320(3):1163-1170
  15. 15. Sobral MV, Xavier AL, Lima TC, de Sousa DP. Antitumor activity of monoterpenes found in essential oils. The Scientific World Journal. 2014;2014:953451
  16. 16. Zeytinoglu H, Incesu Z, Baser KHC. Inhibition of DNA synthesis by carvacrol in mouse myoblast cells bearing a human N-RAS oncogene. Phytomedicine. 2003;10(4):292-299
  17. 17. Jaafari A, Tilaoui M, Mouse HA, M’bark LA, Aboufatima R, Chait A, Lepoivre M, Zyad A. Comparative study of the antitumor effect of natural monoterpenes: Relationship to cell cycle analysis. Revista Brasileira de Farmacognosia. 2012;22(3):534-540
  18. 18. Yoshikawa T, Kokura S, Tainaka K, Naito Y, Kondo M. A novel cancer therapy based on oxygen radicals. Cancer Research. 1995;55(8):1617-1620
  19. 19. Karkabounas S, Kostoula OK, Daskalou T, Veltsistas P, Karamouzis M, Zelovitis I, Metsios A, Lekkas P, Evangelou AM, Kotsis N. Anticarcinogenic and antiplatelet effects of carvacrol. Experimental Oncology. 2006;28(2):121-125
  20. 20. Park BS, Song YS, Yee S-B, Lee BG, Seo SY, Park YC, Kim J-M, Kim HM, Yoo YH. Phospho-ser 15-p53 translocates into mitochondria and interacts with Bcl-2 and Bcl-xL in eugenol-induced apoptosis. Apoptosis. 2005;10(1):193-200
  21. 21. Ghosh R, Nadiminty N, Fitzpatrick JE, Alworth WL, Slaga TJ, Kumar AP. Eugenol causes melanoma growth suppression through inhibition of E2F1 transcriptional activity. The Journal of Biological Chemistry. 2005;280(7):5812-5819
  22. 22. Miller JA, Swanson AB, Miller EC. The metabolic activation of safrole and related naturally occurring alkenylbenzenes in relation to carcinogenesis by these agents. In: Proceedings International Symposium Princess Takamatsu Cancer Research Fund; 1979. pp. 111-125
  23. 23. Crowell PL. Monoterpenes in breast cancer chemoprevention. Breast Cancer Research and Treatment. 1997;46(2):191-197
  24. 24. Wattenberg LW, Sparnins VL, Barany G. Inhibition of N-nitrosodiethylamine carcinogenesis in mice by naturally occurring organosulfur compounds and monoterpenes. Cancer Research. 1989;49(10):2689-2692
  25. 25. Horvathova E, Turcaniova V, Slamenova D. Comparative study of DNA-damaging and DNA-protective effects of selected components of essential plant oils in human leukemic cells K562. Neoplasma. 2007;54(6):478-483
  26. 26. Stammati A, Bonsi P, Zucco F, Moezelaar R, Alakomi H-L, Von Wright A. Toxicity of selected plant volatiles in microbial and mammalian short-term assays. Food and Chemical Toxicology. 1999;37(8):813-823
  27. 27. Koparal AT, Zeytinoğlu M. Effects of carvacrol on a human non-small cell lung cancer (NSCLC) cell line, A549. In: Animal Cell Technology: Basic & Applied Aspects. Dordrecht: Springer; 2003. pp. 207-211
  28. 28. Jordheim LP, Guittet O, Lepoivre M, Galmarini CM, Dumontet C. Increased expression of the large subunit of ribonucleotide reductase is involved in resistance to gemcitabine in human mammary adenocarcinoma cells. Molecular Cancer Therapeutics. 2005;4(8):1268-1276
  29. 29. Bardon S, Foussard V, Fournel S, Loubat A. Monoterpenes inhibit proliferation of human colon cancer cells by modulating cell cycle-related protein expression. Cancer Letters. 2002;181(2):187-194
  30. 30. Chander SK, Lansdown AGB, Luqmani YA, Gomm JJ, Coope RC, Gould N, Coombes RC. Effectiveness of combined limonene and 4-hydroxyandrostenedione in the treatment of NMU-induced rat mammary tumours. British Journal of Cancer. 1994;69(5):879-882
  31. 31. Obrenovich ME, Nair NG, Beyaz A, Aliev G, Reddy VP. The role of polyphenolic antioxidants in health, disease, and aging. Rejuvenation Research. 2010;13(6):631-643
  32. 32. Leouifoudi I, Harnafi H, Zyad A. Olive mill waste extracts: Polyphenols content, antioxidant, and antimicrobial activities. Advances in Pharmacological Sciences. 2015;2015:714138
  33. 33. Obied HK, Prenzler PD, Konczak I, Rehman A, Robards K. Chemistry and bioactivity of olive biophenols in some antioxidant and antiproliferative in vitro bioassays. Chemical Research in Toxicology. 2008;22(1):227-234
  34. 34. Mileo AM, Miccadei S. Polyphenols as modulator of oxidative stress in cancer disease: New therapeutic strategies. Oxidative Medicine and Cellular Longevity. 2015;2016
  35. 35. Singh M, Bhui K, Singh R, Shukla Y. Tea polyphenols enhance cisplatin chemosensitivity in cervical cancer cells via induction of apoptosis. Life Sciences. 2013;93(1):7-16
  36. 36. Thawonsuwan J, Kiron V, Satoh S, Panigrahi A, Verlhac V. Epigallocatechin-3-gallate (EGCG) affects the antioxidant and immune defense of the rainbow trout, Oncorhynchus mykiss. Fish Physiology and Biochemistry. 2010;36(3):687-697
  37. 37. Ahmad N, Gupta S, Mukhtar H. Green tea polyphenol epigallocatechin-3-gallate differentially modulates nuclear factor κB in cancer cells versus normal cells. Archives of Biochemistry and Biophysics. 2000;376(2):338-346
  38. 38. Oak M-H, El Bedoui J, Schini-Kerth VB. Antiangiogenic properties of natural polyphenols from red wine and green tea. The Journal of Nutritional Biochemistry. 2005;16(1):1-8
  39. 39. Alexandre J, Batteux F, Nicco C, Chéreau C, Laurent A, Guillevin L, Weill B, Goldwasser F. Accumulation of hydrogen peroxide is an early and crucial step for paclitaxel-induced cancer cell death both in vitro and in vivo. International Journal of Cancer. 2006;119(1):41-48
  40. 40. Borin TF, Arbab AS, Gelaleti GB, Ferreira LC, Moschetta MG, Jardim-Perassi BV, Iskander ASM, Varma NRS, Shankar A, Coimbra VB. Melatonin decreases breast cancer metastasis by modulating rho-associated kinase protein-1 expression. Journal of Pineal Research. 2016;60(1):3-15
  41. 41. Danesi F, Kroon PA, Saha S, de Biase D, D’Antuono LF, Bordoni A. Mixed pro-and anti-oxidative effects of pomegranate polyphenols in cultured cells. International Journal of Molecular Sciences. 2014;15(11):19458-19471
  42. 42. Symonds EL, Konczak I, Fenech M. The Australian fruit Illawarra plum (Podocarpus elatus Endl., Podocarpaceae) inhibits telomerase, increases histone deacetylase activity and decreases proliferation of colon cancer cells. The British Journal of Nutrition. 2013;109(12):2117-2125
  43. 43. Miccadei S, Di Venere D, Cardinali A, Romano F, Durazzo A, Foddai MS, Fraioli R, Mobarhan S, Maiani G. Antioxidative and apoptotic properties of polyphenolic extracts from edible part of artichoke (Cynara scolymus L.) on cultured rat hepatocytes and on human hepatoma cells. Nutrition and Cancer. 2008;60(2):276-283
  44. 44. Katiyar SK, Matsui MS, Elmets CA, Mukhtar H. Polyphenolic antioxidant (−)-epigallocatechin-3-gallate from green tea reduces UVB-induced inflammatory responses and infiltration of leukocytes in human skin. Photochemistry and Photobiology. 1999;69(2):148-153
  45. 45. Giovannini C, Masella R. Role of polyphenols in cell death control. Nutritional Neuroscience. 2012;15(3):134-149
  46. 46. Kang NJ, Shin SH, Lee HJ, Lee KW. Polyphenols as small molecular inhibitors of signaling cascades in carcinogenesis. Pharmacology & Therapeutics. 2011;130(3):310-324
  47. 47. Rodríguez ML, Estrela JM, Ortega Á. Natural polyphenols and apoptosis induction in cancer therapy. Journal of Carcinognesis & Mutagenesis. 2013;6:1-10
  48. 48. Nooshinfar E, Bashash D, Safaroghli-Azar A, Bayati S, Rezaei-Tavirani M, Ghaffari SH, Akbari ME. Melatonin promotes ATO-induced apoptosis in MCF-7 cells: Proposing novel therapeutic potential for breast cancer. Biomedicine & Pharmacotherapy. 2016;83:456-465
  49. 49. Andersen MH, Svane IM, Kvistborg P, Nielsen OJ, Balslev E, Reker S, Becker JC, Straten P. Immunogenicity of Bcl-2 in patients with cancer. Blood. 2005;105(2):728-734
  50. 50. George J, Singh M, Srivastava AK, Bhui K, Roy P, Chaturvedi PK, Shukla Y. Resveratrol and black tea polyphenol combination synergistically suppress mouse skin tumors growth by inhibition of activated MAPKs and p53. PLoS One. 2011;6(8):e23395
  51. 51. Schneider G, Krämer OH. NFκB/p53 crosstalk—A promising new therapeutic target. Biochimica et Biophysica Acta (BBA): Reviews on Cancer. 2011;1815(1):90-103
  52. 52. Obregon DF, Rezai-Zadeh K, Bai Y, Sun N, Hou H, Ehrhart J, Zeng J, Mori T, Arendash GW, Shytle D. ADAM10 activation is required for green tea (−)-epigallocatechin-3-gallate-induced α-secretase cleavage of amyloid precursor protein. The Journal of Biological Chemistry. 2006;281(24):16419-16427
  53. 53. Arul D, Subramanian P. Naringenin (citrus flavonone) induces growth inhibition, cell cycle arrest and apoptosis in human hepatocellular carcinoma cells. Pathology Oncology Research. 2013;19(4):763-770
  54. 54. Singh N, Nigam M, Ranjan V, Sharma R, Balapure AK, Rath SK. Caspase mediated enhanced apoptotic action of cyclophosphamide-and resveratrol-treated MCF-7 cells. Journal of Pharmacological Sciences. 2009;109(4):473-485
  55. 55. Mukhtar H, Ahmad N. Tea polyphenols: Prevention of cancer and optimizing health. The American Journal of Clinical Nutrition. 2000;71(6):1698s-1702s
  56. 56. Fujiki H, Suganuma M, Okabe S, Sueoka E, Sueoka N, Fujimoto N, Goto Y, Matsuyama S, Imai K, Nakachi K. Cancer prevention with green tea and monitoring by a new biomarker, hnRNP B1. Mutation Research. Fundamental and Molecular Mechanisms of Mutagenesis. 2001;480:299-304
  57. 57. Roy P, Kalra N, Nigam N, George J, Ray RS, Hans RK, Prasad S, Shukla Y. Resveratrol enhances ultraviolet B-induced cell death through nuclear factor-κB pathway in human epidermoid carcinoma A431 cells. Biochemical and Biophysical Research Communications. 2009;384(2):215-220
  58. 58. Bernini R, Crisante F, Merendino N, Molinari R, Soldatelli MC, Velotti F. Synthesis of a novel ester of hydroxytyrosol and α-lipoic acid exhibiting an antiproliferative effect on human colon cancer HT-29 cells. European Journal of Medicinal Chemistry. 2011;46(1):439-446
  59. 59. Kim S, Park TI. Naringenin: A partial agonist on estrogen receptor in T47D-KBluc breast cancer cells. International Journal of Clinical and Experimental Medicine. 2013;6(10):890
  60. 60. Murakami A. Dose-dependent functionality and toxicity of green tea polyphenols in experimental rodents. Archives of Biochemistry and Biophysics. 2014;557:3-10
  61. 61. Leouifoudi I, Mbarki M, Tilaoui M, Amechrouq A, Rakib EM, Mouse HA, Zyad A. Study of the in vitro anticancer activity of Moroccan phenolic olive cake extracts. Journal of Pharmacognosy and Phytochemistry. 2014;2(6):154-165
  62. 62. Schepetkin IA, Ramstead AG, Kirpotina LN, Voyich JM, Jutila MA, Quinn MT. Therapeutic potential of polyphenols from Epilobium angustifolium (fireweed). Phytotherapy Research. 2016;30(8):1287-1297
  63. 63. Qin L, Jin L, Lu L, Lu X, Zhang C, Zhang F, Liang W. Naringenin reduces lung metastasis in a breast cancer resection model. Protein & Cell. 2011;2(6):507-516
  64. 64. Naylor RM, Baker DJ, Deursen J van. Senescent cells: A novel therapeutic target for aging and age-related diseases. Clinical Pharmacology and Therapeutics 2013;93(1):105-116
  65. 65. Jose Marin J, Vergel M, Carnero A. Targeting cancer by inducing senescence. The Open Enzyme Inhibition Journal. 2010 [cité 19 Nov. 2017];3(1):46-52
  66. 66. Lee M, Lee J-S. Exploiting tumor cell senescence in anticancer therapy. BMB Reports. 2014;47(2):51-59
  67. 67. Banerjee K, Mandal M. Oxidative stress triggered by naturally occurring flavone apigenin results in senescence and chemotherapeutic effect in human colorectal cancer cells. Redox Biology. 2015;5:153-162
  68. 68. Khan HY, Zubair H, Ullah MF, Ahmad A, Hadi SM. A prooxidant mechanism for the anticancer and chemopreventive properties of plant polyphenols. Current Drug Targets. 2012;13(14):1738-1749
  69. 69. Sin S, Kim SY, Kim SS. Chronic treatment with ginsenoside Rg3 induces Akt-dependent senescence in human glioma cells. International Journal of Oncology. 2012;41(5):1669-1674
  70. 70. Li Y-B, Gao J-L, Zhong Z-F, Hoi P-M, Lee SM-Y, Wang Y-T. Bisdemethoxycurcumin suppresses MCF-7 cells proliferation by inducing ROS accumulation and modulating senescence-related pathways. Pharmacological Reports. 2013;65(3):700-709
  71. 71. Mileo AM, Di Venere D, Linsalata V, Fraioli R, Miccadei S. Artichoke polyphenols induce apoptosis and decrease the invasive potential of the human breast cancer cell line MDA-MB231. Journal of Cellular Physiology. 2012;227(9):3301-3309
  72. 72. Mileo AM, Di Venere D, Abbruzzese C, Miccadei S. Long term exposure to polyphenols of artichoke (Cynara scolymus L.) exerts induction of senescence driven growth arrest in the MDA-MB231 human breast cancer cell line. Oxidative Medicine and Cellular Longevity. 2015;2015:1-11
  73. 73. Luo H, Yang A, Schulte BA, Wargovich MJ, Wang GY. Resveratrol induces premature senescence in lung cancer cells via ROS-mediated DNA damage. PLoS One. 2013;8(3):e60065
  74. 74. Yang Q, Wang B, Zang W, Wang X, Liu Z, Li W, Jia J. Resveratrol inhibits the growth of gastric cancer by inducing G1 phase arrest and senescence in a Sirt1-dependent manner. PLoS One. 2013;8(11):e70627
  75. 75. Zamin LL, Filippi-Chiela EC, Dillenburg-Pilla P, Horn F, Salbego C, Lenz G. Resveratrol and quercetin cooperate to induce senescence-like growth arrest in C6 rat glioma cells. Cancer Science. 2009;100(9):1655-1662
  76. 76. Cairney CJ, Bilsland AE, Evans TJ, Roffey J, Bennett DC, Narita M, Torrance CJ, Keith WN. Cancer cell senescence: A new frontier in drug development. Drug Discovery Today. 2012;17(5):269-276
  77. 77. Acosta JC, Gil J. Senescence: A new weapon for cancer therapy. Trends in Cell Biology. 2012;22(4):211-219
  78. 78. Tu Y. The discovery of artemisinin (qinghaosu) and gifts from Chinese medicine. Nature Medicine 2011;17(10):1217-20
  79. 79. Van Agtmael MA, Eggelte TA, van Boxtel CJ. Artemisinin drugs in the treatment of malaria: From medicinal herb to registered medication. Trends in Pharmacological Sciences. 1999;20(5):199-205
  80. 80. Woerdenbag HJ, Moskal TA, Pras N, Malingré TM, FS e-F, Kampinga HH, Konings AW. Cytotoxicity of artemisinin-related endoperoxides to Ehrlich ascites tumor cells. Journal of Natural Products. 1993;56(6):849-856
  81. 81. Lai H, Singh NP. Selective cancer cell cytotoxicity from exposure to dihydroartemisinin and holotransferrin. Cancer Letters. 1995;91(1):41-46
  82. 82. Efferth T, Dunstan H, Sauerbrey A, Miyachi H, Chitambar CR. The anti-malarial artesunate is also active against cancer. International Journal of Oncology. 2001;18(4):767-773
  83. 83. Efferth T, Sauerbrey A, Olbrich A, Gebhart E, Rauch P, Weber HO, Hengstler JG, Halatsch M-E, Volm M, Tew KD, et al. Molecular modes of action of artesunate in tumor cell lines. Molecular Pharmacology. 2003;64(2):382-394
  84. 84. Deng X, Liu Z, Liu F, Pan L, Yu H, Jiang J, Zhang J, Liu L, Yu J. Holotransferrin enhances selective anticancer activity of artemisinin against human hepatocellular carcinoma cells. Journal of Huazhong University of Science and Technology. Medical Sciences = Hua zhong ke ji da xue xue bao. Yi xue Ying De wen ban = Huazhong keji daxue xuebao. Yixue Yingdewen ban. 2013;33(6):862-865
  85. 85. Jiao Y, Ge C, Meng Q, Cao J, Tong J, Fan S. Dihydroartemisinin is an inhibitor of ovarian cancer cell growth. Acta Pharmacologica Sinica. 2007;28(7):1045
  86. 86. Tilaoui M, Mouse HA, Jaafari A, Zyad A. Differential effect of artemisinin against cancer cell lines. Natural Products and Bioprospecting. 2014;4(3):189-196
  87. 87. Li Q, Weina PJ, Milhous WK. Pharmacokinetic and pharmacodynamic profiles of rapid-acting artemisinins in the antimalarial therapy. Current Drug Therapy. 2007;2(3):210-223
  88. 88. Firestone GL, Sundar SN. Anticancer activities of artemisinin and its bioactive derivatives. Expert Reviews in Molecular Medicine. 2009;11:e32
  89. 89. Wu J, Hu D, Yang G, Zhou J, Yang C, Gao Y, Zhu Z. Down-regulation of BMI-1 cooperates with artemisinin on growth inhibition of nasopharyngeal carcinoma cells. Journal of Cellular Biochemistry. 2011;112(7):1938-1948
  90. 90. Efferth T, Davey M, Olbrich A, Rücker G, Gebhart E, Davey R. Activity of drugs from traditional Chinese medicine toward sensitive and MDR1-or MRP1-overexpressing multidrug-resistant human CCRF-CEM leukemia cells. Blood Cells, Molecules & Diseases. 2002;28(2):160-168
  91. 91. Lai H, Sasaki T, Singh NP, Messay A. Effects of artemisinin-tagged holotransferrin on cancer cells. Life Sciences. 2005;76(11):1267-1279
  92. 92. Zhou C, Pan W, Wang XP, Chen TS. Artesunate induces apoptosis via a Bak-mediated caspase-independent intrinsic pathway in human lung adenocarcinoma cells. Journal of Cellular Physiology. 2012;227(12):3778-3786
  93. 93. Lu J-J, Meng L-H, Shankavaram UT, Zhu C-H, Tong L-J, Chen G, Lin L-P, Weinstein JN, Ding J. Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells. Biochemical Pharmacology. 2010;80(1):22-30
  94. 94. Lu J-J, Chen S-M, Zhang X-W, Ding J, Meng L-H. The anti-cancer activity of dihydroartemisinin is associated with induction of iron-dependent endoplasmic reticulum stress in colorectal carcinoma HCT116 cells. Investigational New Drugs. 2011;29(6):1276-1283
  95. 95. Crespo-Ortiz MP, Wei MQ. Antitumor activity of artemisinin and its derivatives: From a well-known antimalarial agent to a potential anticancer drug. BioMed Research International. 2011;2012:e247597
  96. 96. Efferth T. Willmar Schwabe award 2006: Antiplasmodial and antitumor activity of artemisinin—From bench to bedside. Planta Medica. 2007;73(4):299-309
  97. 97. Hamacher-Brady A, Stein HA, Turschner S, Toegel I, Mora R, Jennewein N, Efferth T, Eils R, Brady NR. Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. The Journal of Biological Chemistry. 2011;286(8):6587-6601
  98. 98. Tin AS, Sundar SN, Tran KQ, Park AH, Poindexter KM, Firestone GL. Antiproliferative effects of artemisinin on human breast cancer cells requires the downregulated expression of the E2F1 transcription factor and loss of E2F1-target cell cycle genes. Anti-Cancer Drugs. 2012;23(4):370-379
  99. 99. Zhang S, Chen H, Gerhard GS. Heme synthesis increases artemisinin-induced radical formation and cytotoxicity that can be suppressed by superoxide scavengers. Chemico-Biological Interactions. 2010;186(1):30-35
  100. 100. Anfosso L, Efferth T, Albini A, Pfeffer U. Microarray expression profiles of angiogenesis-related genes predict tumor cell response to artemisinins. The Pharmacogenomics Journal. 2006;6(4):269-278
  101. 101. Chen H-H, Zhou H-J, Wu G-D, Lou X-E. Inhibitory effects of artesunate on angiogenesis and on expressions of vascular endothelial growth factor and VEGF receptor KDR/flk-1. Pharmacology. 2004;71(1):1-9
  102. 102. Lee J, Zhou H-J, Wu X-H. Dihydroartemisinin downregulates vascular endothelial growth factor expression and induces apoptosis in chronic myeloid leukemia K562 cells. Cancer Chemotherapy and Pharmacology. 2006;57(2):213-220
  103. 103. Thanaketpaisarn O, Waiwut P, Sakurai H, Saiki I. Artesunate enhances TRAIL-induced apoptosis in human cervical carcinoma cells through inhibition of the NF-κB and PI3K/Akt signaling pathways. International Journal of Oncology. 2011;39(1):279-285
  104. 104. Wang Y, Huang Z, Wang L, Meng S, Fan Y, Chen T, Cao J, Jiang R, Wang C. The anti-malarial artemisinin inhibits pro-inflammatory cytokines via the NF-κB canonical signaling pathway in PMA-induced THP-1 monocytes. International Journal of Molecular Medicine. 2011;27(2):233-241
  105. 105. Cabello CM, Lamore SD, Bair WB III, Qiao S, Azimian S, Lesson JL, Wondrak GT. The redox antimalarial dihydroartemisinin targets human metastatic melanoma cells but not primary melanocytes with induction of NOXA-dependent apoptosis. Investigational New Drugs. 2012;30(4):1289-1301
  106. 106. Hwang YP, Yun HJ, Kim HG, Han EH, Lee GW, Jeong HG. Suppression of PMA-induced tumor cell invasion by dihydroartemisinin via inhibition of PKCα/Raf/MAPKs and NF-κB/AP-1-dependent mechanisms. Biochemical Pharmacology. 2010;79(12):1714-1726
  107. 107. Li L-N, Zhang H-D, Yuan S-J, Tian Z-Y, Wang L, Sun Z-X. Artesunate attenuates the growth of human colorectal carcinoma and inhibits hyperactive Wnt/β-catenin pathway. International Journal of Cancer. 2007;121(6):1360-1365
  108. 108. Mu D, Chen W, Yu B, Zhang C, Zhang Y, Qi H. Calcium and survivin are involved in the induction of apoptosis by dihydroartemisinin in human lung cancer SPC-A-1 cells. Methods and Findings in Experimental and Clinical Pharmacology. 2007;29(1):33-38
  109. 109. WAng J, Hou L, YAng Y, TAng W, Li Y, Zuo J. SM905, an artemisinin derivative, inhibited NO and pro-inflammatory cytokine production by suppressing MAPK and NF-κB pathways in RAW 264.7 macrophages. Acta Pharmacologica Sinica. 2009;30(10):1428-1435
  110. 110. Sertel S, Eichhorn T, Simon CH, Plinkert PK, Johnson SW, Efferth T. Pharmacogenomic identification of c-Myc/max-regulated genes associated with cytotoxicity of artesunate towards human colon, ovarian and lung cancer cell lines. Molecules. 2010;15(4):2886-2910
  111. 111. Konkimalla VB, McCubrey JA, Efferth T. The role of downstream signaling pathways of the epidermal growth factor receptor for Artesunate’s activity in cancer cells. Current Cancer Drug Targets. 2009;9(1):72-80
  112. 112. Karnak D, Xu L. Chemosensitization of prostate cancer by modulating Bcl-2 family proteins. Current Drug Targets. 2010;11(6):699-707
  113. 113. Li Y, Wu YL. How Chinese scientists discovered qinghaosu (artemisinin) and developed its derivatives? What are the future perspectives? Médecine Trop Rev Corps Santé Colon. 1998;58(3 Suppl):9-12
  114. 114. O’neill PM, Barton VE, Ward SA. The molecular mechanism of action of artemisinin—The debate continues. Molecules. 2010;15(3):1705-1721
  115. 115. Wartenberg M, Wolf S, Budde P, Grünheck F, Acker H, Hescheler J, Wartenberg G, Sauer H. The antimalaria agent artemisinin exerts antiangiogenic effects in mouse embryonic stem cell-derived embryoid bodies. Laboratory Investigation. 2003;83(11):1647-1655
  116. 116. Wang J, Zhang B, Guo Y, Li G, Xie Q, Zhu B, Gao J, Chen Z. Artemisinin inhibits tumor lymphangiogenesis by suppression of vascular endothelial growth factor C. Pharmacology. 2008;82(2):148-155
  117. 117. Hou J, Wang D, Zhang R, Wang H. Experimental therapy of hepatoma with artemisinin and its derivatives: In vitro and in vivo activity, chemosensitization, and mechanisms of action. Clinical Cancer Research. 2008;14(17):5519-5530
  118. 118. Langroudi L, Hassan ZM, Ebtekar M, Mahdavi M, Pakravan N, Noori S. A comparison of low-dose cyclophosphamide treatment with artemisinin treatment in reducing the number of regulatory T cells in murine breast cancer model. International Immunopharmacology. 2010;10(9):1055-1061
  119. 119. Dell’Eva R, Pfeffer U, Vené R, Anfosso L, Forlani A, Albini A, Efferth T. Inhibition of angiogenesis in vivo and growth of Kaposi’s sarcoma xenograft tumors by the anti-malarial artesunate. Biochemical Pharmacology. 2004;68(12):2359-2366
  120. 120. Zhao F, Wang H, Kunda P, Chen X, Liu Q-L, Liu T. Artesunate exerts specific cytotoxicity in retinoblastoma cells via CD71. Oncology Reports. 2013;30(3):1473-1482
  121. 121. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini P-L. The immune hallmarks of cancer. Cancer Immunology, Immunotherapy. 2011;60(3):319-326
  122. 122. Ho WE, Peh HY, Chan TK, Wong WF. Artemisinins: Pharmacological actions beyond anti-malarial. Pharmacology & Therapeutics. 2014;142(1):126-139
  123. 123. Bachmeier B, Fichtner I, Killian PH, Kronski E, Pfeffer U, Efferth T. Development of resistance towards artesunate in MDA-MB-231 human breast cancer cells. PLoS One. 2011;6(5):e20550
  124. 124. Demain AL, Vaishnav P. Natural products for cancer chemotherapy. Microbial Biotechnology. 2011;4(6):687-699
  125. 125. Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25 years. Journal of Natural Products. 2007;70(3):461-477
  126. 126. Butler MS, Robertson AA, Cooper MA. Natural product and natural product derived drugs in clinical trials. Natural Product Reports. 2014;31(11):1612-1661
  127. 127. Cragg GM, Pezzuto JM. Natural products as a vital source for the discovery of cancer chemotherapeutic and chemopreventive agents. Medical Principles and Practice. 2016;25(Suppl. 2):41-59

Written By

Abdelmajid Zyad, Inass Leouifoudi, Mounir Tilaoui, Hassan Ait Mouse, Mouna Khouchani and Abdeslam Jaafari

Reviewed: 27 November 2017 Published: 29 December 2017