Flavonoids from Achyrocline satureioides: promising biomolecules for anticancer therapy

Juliana Poglia Carini *a, Fábio Klamt bc and Valquiria Linck Bassani a
aPrograma de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Ipiranga 2752, 90610-000 Porto Alegre (RS), Brazil. E-mail: julianacarini@yahoo.com.br; Fax: +55 51 3308 5437; Tel: +55 51 3308 5231
bLaboratório de Bioquímica Celular, Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2600, 90035-003 Porto Alegre (RS), Brazil
cInstituto Nacional de Ciência e Tecnologia – Translacional em Medicina (INCT-TM), 90035-903 Porto Alegre (RS), Brazil

Received 15th July 2013 , Accepted 16th October 2013

First published on 17th October 2013


Abstract

Cancer is the major public health problem worldwide; consequently, the search for new chemotherapeutic drugs is constant. Most of these agents are derived from natural sources, which are the major consistent basis for the search for modern anticancer medicines. In this context, numerous studies indicate flavonoids as a potential new class of secondary metabolites for anticancer therapy. In this review, special attention was addressed to flavonoids present in Achyrocline satureioides, a widely used medicinal plant with several and a well established range of biological properties. Two of these flavonoids are extensively studied for anticancer therapy, quercetin and luteolin, followed by 3-O-methylquercetin. Achyrobichalcone, recently isolated from A. satureioides by our group, can also represent a promising chemotherapeutic biomolecule due to its similarity with other cytotoxic bichalcones to cancer cell lines. The anticancer properties of these flavonoids, specially quercetin and luteolin, type of cell death, mechanisms and molecular targets involved were described. In general, these effects were observed due to the inhibition of cell proliferation, cell cycle arrest, apoptosis, inhibition of angiogenesis, prevention of migration/metastasis and overcoming multidrug resistance, alone or in combination with commonly used chemotherapeutic drugs. All these successful findings in preclinical studies suggest that these flavonoids are promising biomolecules for the development of new anticancer drugs in the future.


image file: c3ra43627f-p1.tif

Juliana Poglia Carini

Juliana Poglia Carini received her Doctor degree in Pharmaceutical Sciences, with emphasis in Pharmaceutical Technology, from the Federal University of Rio Grande do Sul (UFRGS), Brazil, in 2013. Her doctoral thesis was developed in the research group of Professor Valquiria Linck Bassani, which focused on isolation of new phytopharmaceuticals from Achyrocline satureioides and their biological evaluation, with emphasis on anticancer activity.

image file: c3ra43627f-p2.tif

Fábio Klamt

Fábio Klamt is a Professor and Researcher at the Department of Biochemistry at the Federal University of Rio Grande do Sul (UFRGS), Brazil. He was a Postdoctoral fellow at the Division of Therapeutic Proteins/Food and Drug Administration (FDA), USA, studying molecular mechanisms of the elimination of tumor cells by oxidants. His experience is especially in the area of biochemistry and cell biology: development of in vitro models, study of the pathophysiology of oxidants in biological systems, cellular and molecular mechanisms involved in mitochondrial dysfunction and death by apoptosis in tumors and in neurons, and establishment of prognostic and predictive biomarkers in cancer. He is an Affiliate Member of the Brazilian Academy of Sciences.

image file: c3ra43627f-p3.tif

Valquiria Linck Bassani

Valquiria Linck Bassani is a Full Professor in Pharmaceutical Technology at the Federal University of Rio Grande do Sul (UFRGS), Brazil. She was the Dean of Pharmacy School and Vice-rector for Graduate Courses of the UFRGS. Her multidisciplinary research group is focused in phytopharmaceuticals with emphasis on phytopharmaceutical technology and biological activities of several species from South America, specially Achyrocline satureioides. The influence of the extraction or drying methods, cyclodextrin association and other technological tools on the biological activity of the herbal products have been studied as a strategy for exploring the pharmacological potential of the Brazilian plants.


1. Introduction

Cancer is a major public health problem in many countries of the world.1 Because of this, the disease receives the special attention of the World Health Organization from the International Agency for Research on Cancer. The GLOBOCAN 2008 project was one of the tools created by these international agencies that allowed estimation of the cancer incidence and mortality. The program indicated that about 14.9 million cancer cases and 8.9 million cancer deaths are estimated to occur in 2015 worldwide, with the majority of deaths occurring in the economically developing world.2 In recent decades, the disease has become more common in developing countries, such as Brazil, where it is estimated there has been more than 500[thin space (1/6-em)]000 new cases of cancer in 2013.3

Natural products are privileged structures created by strong biological and ecological pressure that are able to interact with a wide variety of biological targets, consequently originating effective drugs in a large variety of therapeutic indications.4,5 Throughout human evolution, the importance of natural products for medicine and health has been enormous, from the earliest civilizations until today.6 The accumulated experience, knowledge and research over the years makes the secondary metabolites from natural sources, like plants, the most consistently successful approach for obtaining modern medicines.6,7

According to an extensive work published by Newman and Cragg, in 2012, 175 small molecules were approved worldwide over the time frame from around the 1940s to 2010 for cancer treatment. Of this total, the majority (74.9%) were inspired by compounds found in nature.8 Since the discovery of successful natural anticancer drugs such as paclitaxel (Taxol®) (from Taxus brevifolia), camptothecins (from Camptotheca acuminata), vinca alkaloids, such as vincristine and vinblastine (from Catharanthus roseus) and many others,4 the clinical interest for secondary metabolites for cancer therapy increased over the years and currently receives major attention from researchers.

Flavonoids are an emerging potential class of secondary metabolites from plants for anticancer chemotherapy.9 The existence of a large number of recently published papers focused on the chemotherapeutic and/or chemopreventive properties of flavonoids9–22 reinforces the great interest and importance of this issue today. Thus, this work aims to review the potential application of flavonoids for anticancer therapy focusing on the compounds present in Achyrocline satureioides (Lam.) DC. Asteraceae.

2. Flavonoids and anticancer therapy

Flavonoids have probably existed in the plant kingdom for over one billion years and they are present in practically all dietary plants, vegetables and in several medicinal plants used in folk medicine around the world.11 Interest in the anticancer effects of flavonoids has emerged from in vitro and in vivo experimental evidence indicating they interfere with cancer processes such as proliferation, inflammation, angiogenesis, invasion and metastasis.18,22 In addition, a growing number of epidemiological studies suggests that high flavonoid intake may be correlated with a decreased risk of cancer.11 Usually, the molecular targets of flavonoids comprise the mitogen-activated protein kinase (MAPK), protein kinase C (PKC), phosphatidylinositol 3-kinase (PI3K or AKT) and β-catenin pathways, whose activity has been associated with malignant transformation and tumor promotion. Flavonoids can also interfere with the activation of the transcription factors such as nuclear factor kappa B (NFkB) and activator protein-1 (AP-1), while inducing cell cycle arrest and apoptosis.23

Two flavonoid derivatives reached close to approval and being launched in the market as new drugs for cancer treatment. Phenoxodiol, a synthetic analog of the plant isoflavone genistein, is one of these molecules, which is effective as anticancer agent and/or chemosensitizer for several traditional chemotherapeutic drugs in resistant tumors. The molecule is active against several human cancers, such as ovarian cancer,24–27 prostate cancer,28 melanoma,29,30 and squamous carcinoma,31 both in vitro and in vivo tests, reaching the step of clinical trials for some cancers.32 Phenoxodiol presented several promising and encouraging results, as the ability to increase the effectiveness of docetaxel in drug resistant ovarian carcinoma cells by at least 100 times,33 besides having an inhibitory effect against Topoisomerase 2 in the same range of etoposide, but stronger than genistein.34 Despite this, phenoxodiol failed in clinical trials for recurrent ovarian carcinoma after the “fast track” status for clinical use provided by FDA,9 the molecule (or its derivatives) still arouses great scientific35–38 and economical interest, especially from the pharmaceutical industry.39

Flavopiridol is another semisynthetic flavonoid with potential anticancer properties. The compound is a flavone derived from rohitukine, a natural anticancer agent isolated from an Indian tree.11 Due to its interesting biological properties both in vitro and in vivo, as cell cycle arrest in G1 and G2 phases, cytotoxicity against the majority of tumor cell lines in the nanomolar range, suppression of the tumor growth in animals and synergistic activity with most anticancer drugs and radiation, flavopiridol was the first cyclin-dependent kinase (CDK) inhibitor to be tested in human clinical trials.11,40,41 However, the molecule in Phase II clinical trials was ineffective against cancer and presented high toxicity to patients.40–42 Nevertheless, currently 60 anticancer clinical trials can be found with flavopiridol, alone or combined with other drugs, whereby 6 of them have shown to be active in the recruiting phase.43

2.1. Flavonoids from Achyrocline satureioides

Achyrocline satureioides (Fig. 1A), known as marcela, is a very popular medicinal plant in southern Brazil, being one of the most remembered and used by the local population.44 The species is widely used in folk medicine as an anti-inflammatory, antispasmodic, digestive and carminative agent.45–49 This popular use has motivated biological and ethnopharmacological studies of the species and revealed a wide range of pharmacological properties.45–84
image file: c3ra43627f-f1.tif
Fig. 1 Achyrocline satureioides inflorescences (A) and chemical structures of its major flavonoids: quercetin (QCT) (B); luteolin (LUT) (C); 3-O-methylquercetin (3OMQ) (D); and achyrobichalcone (ACB) (E). The main molecular mechanisms of anticancer activity for QCT and LUT are highlighted.

Some works have reported the ethnopharmacological use of Achyrocline genus (A. satureioides and A. lehmanii Heiron) for anticancer therapy.50,85 A work of Santos and Elisabetsky, in 1999, investigated the ethnopharmacological use of plants to treat signs and sympoms related to cancer. From these analyses, A. satureioides was one of the species with the highest degree of importance.58 Another study reported that the plant was used by cancer patients as a complementary therapy to radioterapy.86

The anticancer properties of A. satureioides extracts could also be observed in biological assays. Methanolic extracts of its flowers inhibited growth of two human cancer cell lines: oropharyngeal epidermoid carcinoma cells (KB cells)54 and hepatoblastoma cells (HepG2 cells).63 The inhibition growth of KB cells was 66.8% after 72 hours of treatment with 50 μg mL−1 of the methanolic extract.54 The IC50% of HepG2 cells after 48 hours of treatment was 237 μg mL−1 (SD 56 μg mL−1), with inhibition growth in a concentration-dependent manner.63 The difference between these results can be related to the types of cell line, but also due to variations in methodology, as methods of evaluation of the cell death, treatment times or lack of standardization of the tested extracts. In another report, an aqueous extract of A. satureioides significantly inhibited cell division in the Allium cepa system, a vegetal test which measures the mitotic and replication indexes.87 Since the main flavonoids are present in the plant as aglycone form, only low concentrations are present in aqueous extracts. Thus, the observed effect reported by Fachinetto et al., in 2007, can be related to other constituents of the plant rather than the flavonoid aglycones.

Polydoro et al., in 2004, noted that A. satureioides freeze dried extracts (prepared from hydroethanol extractive solutions using 80% or 40% v/v ethanol) were cytotoxic to Sertoli cells, at the concentrations of 0.25 and 0.125 mg mL−1, increasing cell death over 30%. It was also observed that an enriched flavonoid fraction, which contained the highest total flavonoid aglycone, caused increase in the cell death and the extracts with higher quercetin contents (prepared with 80% hidroethanol solution) were more cytotoxic.67 Nevertheless, another study showed that an aqueous extract of A. satureioides (2% infusion) had low acute toxicity when administered intraperitoneally and no toxicity upon oral administration,88 that could be related to the very low flavonoid aglycone content, which presents reduced water solubility.

Most of the biological properties ascribed to A. satureioides extracts are related to the presence of flavonoids in its inflorescences.89 Often, quercetin (QCT; Fig. 1B), luteolin (LUT; Fig. 1C) and 3-O-methylquercetin (3OMQ; Fig. 1D) are the main flavonoids found in extracts, mainly in hydroalcoholic preparations.45,49,90,91,92 More recently, our group identified a new flavonoid in A. satureioides, named achyrobichalcone (ACB; Fig. 1E).91,93 Bearing in mind that a better understanding of the pharmacological activity of flavonoids is a key factor to improve the comprehension of the mode of action of many plant extracts,94 the next topics will address a compilation of the main studies found in literature concerning the anticancer properties of flavonoids from A. satureioides. Noting that much information is found in scientific databases about this issue, the intention of this review is to describe the most relevant and recent findings.

2.2. Molecular targets and mechanisms of action

Effects of flavonoids from A. satureioides (mainly QCT and LUT) against cancer are multi-targeted and involve a wide range of biological activities, as antiproliferative action against several cancer cells of human origin, induction of apoptosis, cell cycle arrest, inhibition of cell migration/invasion and angiogenesis. Often, their anticancer molecular targets in the cell cycle are cyclins (cyclin A-cyclin T) and cyclin-dependent protein kinases (CDK1-CDK9).95 Other related proteins are mitogen-activated protein kinases (MAPK), originally called extracellular signal-regulated kinase (ERK), phosphatidylinositide 3-kinases (PI3K), protein kinase C (PKC), among others. These proteins activate specific cell signaling pathways culminating in cell death.96 The reviewed flavonoids induced apoptosis by activating the intrinsic or extrinsic pathways. In the intrinsic pathway or at mitochondrial level, the Bcl-2 family members were induced, as pro-apoptotic proteins (Bax, Bak, PUMA, NOXA, Bim, Bid) or suppressed, as anti-apoptotic proteins (Bcl-2, Mcl-1, Bcl-xL, Bfl-1/A1).97 Other signaling proteins were inhibited, as survivin (member of inhibitor of apoptosis-IAP family) and nuclear factor kappa B (NFkB), or activated, as caspases. Flavonoids also act in p53 regulation.98,99 The extrinsic apoptotic pathway is induced by extracellular ligands, such as CD95 and TRAIL, which bind to specific receptors on the membrane, for example, death receptors, and triggers a cascade of apoptotic signals culminating in activation of caspases. Caspases activation leads to the destruction and elimination of cancer cells.100,101 Other particular mechanisms and pathways for cell death were observed and will be described opportunely.

In general, the direct anticancer effects of flavonoids from A. satureioides were involved with cell proliferation, cell cycle arrest, apoptosis, angiogenesis, migration/metastasis and overcome multidrug resistance. These effects were observed for flavonoids alone or in combination with commonly used chemotherapeutic drugs.

2.3. Quercetin (QCT)

In the literature, numerous works are described evaluating the anticancer properties of QCT. Table 1 displays the major findings related to in vitro studies, with some of them correlated with in vivo or/and ex vivo experiments. QCT was active against many types of cancers and human cell lines, usually in concentrations between 50 and 100 μM, in a time and dose-dependent manner, reaching various cellular targets.
Table 1 Overview of the major molecular targets and biological effects of quercetin in anticancer therapy
Cancer Cell culture system Concentration used Molecular mechanism Biological effect Reference
Bladder 253J 50 μM Activation of BKCa channels Antiproliferative 102
T24 50–100 μM Inhibition of ecto-5′-NT/CD73 activity Antiproliferative 103
T24, EJ, J82 100 μM Increase in G0/G1 phase, decrease in mutant p53 and survivin Induction of apoptosis, cell cycle arrest 104
Neuro (glioma) U87, T98G 25–50 μM Blocker of IL-6/STAT3 Antiproliferative, inhibition of migration 105
U87 50 μM Inhibition of NFkB-induced by PLD1, decrease in MMP-2 Antiproliferative, anti-invasive 106
A172 50 μM Down-regulation of ERK, AKT and survivin Induction of apoptosis 107
Colon HT29 50–100 μM Increase in G1 phase, activation of AMPK, up-regulation of p53 and p21 Induction of apoptosis, cell cycle arrest 108
HT29/MCF-breast 25–200 μM Increase in G1 phase, activation of AMPK-COX-2 Induction of apoptosis, cell cycle arrest 109
HCT116 40 μM Induction of NAG-1 Induction of apoptosis 110
Liver HepG2 40–160 μM Increase in G0/G1 phase and p53, decrease in survivin and Bcl-2, activation of caspase-9 and -3 Induction of apoptosis, cell cycle arrest 111
Stomach BGC823 30–120 μM Increase in S phase, decrease in Bcl-2/Bax ratio, activation of caspase-3 Induction of apoptosis, cell cycle arrest 112
Blood (leukemia) CCRF-CEM, HL60, K562 12.5–100 μM Suppression of telomerase activity Induction of apoptosis 113
HL60 25–100 μM Induction of FasL, activation of caspase-8 Induction of apoptosis 114
Breast MCF7 25–200 μM Suppression of AMPK, decrease HIF-1α Induction of apoptosis 115
MCF7 10–175 μM Increase in S phase, decrease in Bcl-2 and ΔΨm, activation of caspase-6, -8 and -9 Induction of apoptosis, cell cycle arrest 116
Skin (melanoma) B16-BL6 330 μM Decrease in PKC and MMP-9 Anti-invasive 117
Prostate PC3 50–200 μM Increase in G0/G1 phase, decrease in Bcl-2 and ΔΨm, increase in Bax, activation of caspase-3, -8 and -9 Induction of apoptosis, cell cycle arrest 118
PC3 (and HUVECs) 20–40 μM Down-regulation of AKT, mTOR and P70S6K Induction of apoptosis, antiangiogenic 119
Lung A549 Increase in G0/G1 phase, decrease in survivin and Bcl-2, activation of caspase-3 Induction of apoptosis, cell cycle arrest 120
A549, H1299 20–80 μM Increase in G2/M phase and p53, decrease in survivin Induction of apoptosis, cell cycle arrest 121
A549 14.5–58 μM Activation of MEK–ERK Induction of apoptosis 122


In the bladder cancer cell lines T24, EJ and J82, QCT inhibited the cell growth by cell cycle arrest in G0/G1 and induced apoptosis by decrease in the expression of mutant p53 and surviving.104 Additionaly, Rockenbach et al., in 2013, observed the involvement of the ectonucleotidase pathway, by means of ecto-5′-NT/CD73, the enzyme responsible for AMP hydrolysis, in the antiproliferative effects of QCT on T24 cells.103 The large conductance Ca2+-activated K+ (BKCa) channel influenced the proliferation of 253J bladder cancer cells only in the presence of QCT, suggesting that this channel may represent a valuable target for antiproliferative effects of QCT in these cells.102

In U87 and T98G glioma cells, QCT exerted anticancer effects through abrogation of the IL-6/STAT3 signaling pathway, inhibiting IL-6 and consequently STAT3, a key player involved in cancer-related inflammation by activate cellular responses as cyclin D1, MMP2 and Bcl-2 responsible for cancer cell proliferation, invasion and survival, respectively.105 Other work demonstrated that QCT inhibited U87 cell proliferation by abolishing the phospholipase D (PLD1) expression, a regulator of cell proliferation and tumorigenesis.106 In A172 cells, QCT induced apoptosis through caspase-dependent mechanisms involving down-regulation of ERK, AKT and surviving.107

QCT promoted cell cycle arrest through the increase in G1 phase in HT29 colon cancer cells and upregulated apoptosis-related proteins, such as AMPK, p53 and p21. Additionaly, in vivo experiments showed that QCT resulted in a significant reduction in tumor volume over 6 weeks of treatment and apoptosis-related protein induction was significantly higher in the treated group compared to the control group.108 Other works observed that AMPK activation in HT29 colon cancer cells and MCF breast cancer cells seemed to be closely related to a decrease in COX-2 expression, suggesting that a possible mechanism involved in the anticancer activity of QCT is partly mediated through its anti-inflammatory action.109,123 The expression of NAG-1 (non-steroidal anti-inflammatory drug activated gene-1) was induced during treatment of HCT116 colon carcinoma cells with the QCT and apoptosis process was activated. Early growth response-1 (EGR-1) and p53 were required for QCT mediated activation of the NAG-1 promoter.110

The apoptotic effect and cell cycle arrest of QCT in HepG2 hepatoma cancer cells was promoted at G0/G1 phases, with suppression of survivin and Bcl-2, elevation of p53 and activation of caspase-9 and caspase-3.120,124 Similar results were found for the BGC-823 gastric cancer cell treated with QCT, where caspase-3 was activated and the Bcl-2/Bax ratio was decreased. Additionally, some morphological features of apoptosis were found, such as cell shrinkage or even the formation of an apoptotic body.112

Avci et al., in 2011, observed that QCT had antiproliferative and pro-apoptotic effects on acute lymphocytic leukemia (ALL) (CCRF-CEM), acute promyelocytic leukemia (HL60) and chronic myeloid leukemia (CML) (K562) cell lines by the suppression of the activity of telomerase, an enzyme that is one of the hallmarks of cancer because it is able to restore final sequences of DNA.113 QCT also activated the extrinsic apoptotic pathway in HL60 cells through interaction with death receptor Fasligand (FasL) in the plasma membrane, which is involved in ERK and JNK signaling pathways and consequent activation of caspase-8, induction of Bid cleavage and Bax conformation change.114

Despite its apoptotic effects, AMPK also can help the cancer cell in the adaptation and survival during hypoxic conditions. In MCF7 breast cancer cells under hypoxia, QCT was capable of inhibiting AMPK and decrease HIF-1α, which is a critical survival factor in hypoxia.115 Another study showed that QCT arrested the cell cycle in sub-G1 and S phases for MCF-7 cells. The arrest in S phase was mediated by decreasing the protein expression of CDK2, cyclins A and B while increasing the p53 and p57 proteins. Decreased levels of Bcl-2 protein and ΔΨm and increased activations of caspase-6, -8 and -9 also were observed.116 Similar results were found for metastatic breast cancer cells, as MDA-MB-231 (ref. 125 and 126) and MDA-MB-453 cells.127

Antiangiogenic activity of QCT was evaluated in vitro in PC3 prostate cancer cells and HUVEC endothelial cells, besides in vivo and ex vivo. QCT suppressed vascular endothelial growth factor (VEGF) that induced phosphorylation of VEGF receptor 2 and their downstream protein kinases AKT, mTOR and P70S6K in HUVECs, that are responsible by promoting proliferation, migration and tube formation of endothelial cells. QCT (20 mg kg−1) significantly reduced the volume and the weight of solid tumors in the prostate xenograft mouse model, inhibiting tumorigenesis by targeting angiogenesis. Apoptosis was induced in PC3 cells, which was correlated with the down-regulation of AKT, mTOR and P70S6K expressions.119 The actions of QCT suppressing tumor invasion, angiogenesis and metastasis in PC3 cells also were observed in other works128,129 and in B16-BL6 melanoma cells.117 QCT promoted cell cycle arrest, increasing G0/G1 phase, decreasing Bcl-2 and ΔΨm, increasing Bax and activating of caspase-3, -8 and -9 in PC3 cells.118 These findings are in agreement with the results of Senthilkumar et al., in 2010, that demonstrated the induction of both intrinsic and extrinsic pathways to mediate apoptosis in PC3 cells after treatment with QCT.130

QCT induced cytotoxicity and apoptosis in A549 and H1299 non-small lung carcinoma cells. Inhibition of cell growth was observed, with increase in G2/M phase and raised cyclin B1 and phospho-CDC2 proteins. Survivin proteins were decreased and levels of total p53, phospho-p53 and p21 proteins increased and translocated to the nuclei.121 Another work demonstrated that QCT reduced cell viability and DNA synthesis, with a rate of apoptosis equivalent, in A549 cells. QCT treatments led to increase in Bax and Bad, decrease in Bcl-2 and cleavage of caspase-3 and caspase-7. While Akt-1 was inhibited, ERK was phosphorylated following QCT treatment. The induction of MEK–ERK pathway in A549 cells led to activation of caspase-3 and apoptosis.122 However, QCT was not active against DMS114, a small lung carcinoma cell.131

Finally, some works pointed QCT as very potent inhibitor of topoisomerases (Top 1 and 2) having activity at concentrations similar to that of etoposide (50 μM).9

Co-treatments with QCT and traditional chemotherapeutics drugs. QCT effectively enhances the toxic effects of doxorubicin (DOX) in SMMC7721 and QGY7701 liver cancer cells and hepatoma xenografts in mices. Pro-apoptotic activity of QCT in DOX-treated liver cancer cells was mediated by p53, activation of the mitochondrial apoptotic pathway and cleavage of pro-caspases. In addition, QCT reduces hepatotoxicity of DOX in normal liver cells in vitro and in vivo.132 The synergistic action of QCT and DOX was also observed in the treatment of human breast cancer cells. QCT potentiated anticancer effects of DOX specifically in the highly invasive breast cancer cells and attenuated unwanted cytotoxicity by DNA damage in normal cells.133 These findings were confirmed in vivo in a work of Du et al., in 2010, where QCT combined to an intratumoral DOX injection induced potent rejection of 4T1 breast cancer and led to long-term, tumor-free survival in mice bearing established breast tumor. QCT or DOX alone failed to cure tumor-bearing mice. The response against the tumor was achieved by the induction of immune responses at the same time that QCT and DOX induced cell death.134

The resistance of death receptor to the fludarabine in leukemic cells isolated from chronic lymphocytic leukemia (CLL) patients was ameliorated by combined treatment with QCT.135 In another work, QCT cooperated with arsenic trioxide to induce apoptosis in U937, THP-1 and HL-60 leukemia cells, increasing the clinical efficacy of the antileukemic drug.136

QCT sensitized DB-1 melanoma (p53 wildtype) cells to temozolomide (TMZ) by a mechanism that involves the modulation of p53 family members. After treatment with TMZ, DB-1 cells demonstrated increased p53, however the cells were resistant to TMZ-induced apoptosis. QCT treatment in combination with TMZ abolished drug insensitivity and caused a more than additive induction of apoptosis compared to either treatment alone.137

The antiproliferative effect of QCT and cisplatin (CIS) alone or in combination was evaluated in SPC212 and SPC111 malignant mesothelioma cell lines. QCT significantly reduced the proliferation of cell lines, altered the cell cycle distribution and increased the level of caspase-9 and -3. Additionally, the combination of QCT and CIS was found to be more effective when compared to single-drug treatment.138 In another work, QCT exhibited synergistic effects with CIS against nasopharyngeal carcinoma cells. The dose reduction index found implied the possibility of reducing the CIS dosage required to treat with the addition of QCT, reducing the risk of CIS-associated toxicity.139 In vincristine (VCR) resistant A549 lung adenocarcinoma cells, QCT exerted increasing chemosensitivity when combined with CIS and VCR.140 It was also found that in lung cancer cells that QCT sensitizes, TRAIL-induced apoptosis and two independent pathways are involved: induction of DR5 through PKC and suppression of AKT mediated survivin expression.141

In the pancreatic cell line EPP85-181RDB, QCT seemed to sensitize resistant cells to daunorubicin (DAU). In parallel, the effect of both substances on the sensitive cell line was synergistic.142 QCT was an effective cytotoxic agent in gastric carcinoma cell lines. It had not only a synergistic effect with DAU on P-cells but also sensitized DOX-resistant cells. The analysis of p-glycoprotein (P-gp) activity showed that QCT may down-regulate this transporter.100

The combination treatment of 5-fluorouracil (5-FU) and QCT increased apoptosis in CO115 colorectal cells. Mitochondrial pathway and p53 were involved in cell death.143 In another work, QCT sensitized HCT116 colon cancer cells to CIS and etoposide (ETO) under hypoxic conditions by targeting AMPK.144

2.4. Luteolin (LUT)

The anticancer properties of LUT were extensively studied as those of QCT. The findings described in Table 2 exhibited in vitro studies, with some of them correlated with in vivo or/and ex vivo experiments. Usually, at concentrations near 50 μM, LUT was active against many types of cancers and human cell lines, in a time and dose-dependent manner, reaching various cellular targets.
Table 2 Overview of the major molecular targets and biological effects of luteolin in anticancer therapy
Cancer Cell culture system Concentration used Molecular mechanism/cellular targets Biological effect Reference
Bladder RT112 10–50 μM Increase in sub-G1 phase, poly(ADP-ribose) polymerase cleavage, DNA fragmentation Induction of apoptosis, cell cycle arrest 145
Colon COLO205, HCT116 40 μM Inhibition of NFkB, activation of JNK Induction of apoptosis 146
HT29 40–60 μM Increase in G1 and G2/M phases, poly(ADP-ribose) polymerase cleavage, decreased in p21CIP1/WAF1, survivin, Mcl-1, Bcl-xL, and Mdm-2, activation caspase-3, -7 and -9 Induction of apoptosis, cell cycle arrest 147
Liver HepG2 25–100 μM Activation of AMPK, inhibition of NFkB Induction of apoptosis 148
HLF 50 μM Blocker of STAT3 Antiproliferative, induction of apoptosis 149
Stomach AGS 20–80 μM Increase in G2/M phase, Bax and p53, decrease of Bcl-2, activation of caspase-3, -6, -9 Induction of apoptosis, cell cycle arrest 150
Blood (leukemia) H60 10–50 μM DNA fragmentation Induction of apoptosis 151
Breast MCF7, MDA-MB-231, SK-BR3 30 μM Activation of ERK and p38 Induction of apoptosis 152
Skin (melanoma) B16F10 5–50 μM Inhibition of β3 integrin/FAK Anti-invasive 153
Prostate PC3 (and HUVECs) 20–40 μM Suppression of VEGFR2, down-regulation of AKT, ERK, mTOR, P70S6K, MMP-2, MMP-9 Induction of apoptosis, antiangiogenic 154
PC3 10–40 μM Induction of E-cadherin Anti-invasive 155
Lung A549, H460 5–80 μM Production of superoxide, degradation of MKP-1, activation of JNK Induction of apoptosis 156
A549 20–80 μM Increase in G2/M phase, increase in Bax, activation of JNK, inhibition of NFkB, activation of caspase-3, -9 Induction of apoptosis, cell cycle arrest 157
A549 25–50 μM Increase in G1 phase, decrease in ΔΨm, disruption of actin Induction of apoptosis, cell cycle arrest, inhibition of migration 158


The cytotoxic effects of LUT on two different cancer cell lines, including RT112 bladder carcinoma and K562 chronic myeloid leukemia, was described by Kilani-Jaziri et al., in 2012. The apoptosis was induced, with minor cell-cycle perturbations, but some increase in sub-G1 fraction was observed. The poly(ADP-ribose) polymerase was cleaved and a typical ladder of DNA fragments was observed in treated cells.145

In COLO205 and HCT116 colorectal cancer cells and cervical cancer HeLa cells, LUT inhibited TNFα, a cell mediator that activates both cell death and cell survival pathways, which render most cancer cells resistant to its cytotoxicity. With the inhibition of TNFα, the activation of NFkB was decreased and consequently the induction of NFkB-targeted anti-apoptotic genes was inhibited. In addition, the inhibition of NFkB led to augmentation of JNK activation.146 Another work showed that LUT induced G1 and G2/M cell cycle arrest, which was mediated by CDK2, CDK4 and CDC2 in HT29 colon cancer cells. In addition, LUT promoted apoptosis through down-regulation of several anti-apoptotic proteins.147 The same group described that LUT down-regulates the activation of the PI3K/AKT and ERK1/2 pathways via reduction in insulin-like growth factor-I receptor (IGF-IR) signaling in HT29 cells.159

LUT strongly induced cell death in HepG2 hepatoma cells and dramatically reduced the tumor volume in a tumor xenograft model. Both effects were accompanied by AMPK activation and a strong inhibitory effect on NFkB. LUT treatment causes reactive oxygen species (ROS) production and these intracellular ROS in turn mediate AMPK-NFkB signaling.148 Selvendiran et al., in 2006, observed the pro-apoptotic effect of LUT on HLF hepatoma cells in vitro and in vivo. Apoptosis was found by caspase-8 activation and enhanced expression in Fas/CD95. Decrease of STAT3, a known negative regulator of Fas/CD95 transcription, lead to down-regulation on these target gene products, such as cyclin D1, survivin, Bcl-xL and vascular endothelial growth factor. An overexpression in STAT3 led to resistance to LUT, suggesting that STAT3 was a critical target of LUT. In nude mice with xenografted tumors using HAK-1B hepatoma cells, LUT significantly inhibited the growth of the tumors.149

Antiproliferative and chemosensitizing effects of LUT on AGS gastric cancer cells treated with LUT and/or other chemotherapeutic agents were evaluated by Wu et al., 2008. LUT induced cell cycle arrest by an increase of the G2/M phase, besides reducing protein levels of CDC2, cyclin B1 and CDC25C and increasing the cyclin-dependent kinase inhibitor p21 (cip1/Waf1). The pro-apoptotic proteins, including caspase-3,- 6, -9, Bax and p53 were increased and the levels of anti-apoptotic protein Bcl-2 was reduced, thus shifting the Bax/Bcl ratio in favor of apoptosis. The combined treatment of CIS and LUT induced more effective cell growth inhibition, compared to CIS treatment alone.150

In MCF7, MDA-MB-231 and SK-BR3 breast cancer cells, LUT induced nuclear translocation of AIF (apoptosis-inducing factor), which was mediated by activation of ERK and p38. LUT induces caspase dependent and independent apoptosis involving AIF nuclear translocation mediated by activation of ERK and p38.152 Another work with MDA-MB-231 cells showed that LUT arrested the cell cycle at the G2/M and S stages and activated the apoptosis by decreasing AKT, PLK1, cyclin B1, cyclin A, CDC2, CDK2 and Bcl-xL and increasing p21 and Bax expression. LUT-supplementation significantly reduced tumor burden in nude mice inoculated with MDA-MB-231 cells.160

LUT suppressed the hypoxia-induced changes in B16F10 melanoma cells, as increased cellular adhesion and invasion. Hypoxia significantly decreased the expression of E-cadherin while increasing the expression of N-cadherin in the cells, which was reversed by LUT. LUT up-regulated the E-cadherin at least partly via inhibiting the β3 integrin/FAK signal pathway. In experimental metastasis model mice, treatment with LUT (10–20 mg kg−1) reduced metastatic colonization in the lungs by 50%.153

The antiangiogenic activity of LUT was evaluated by Pratheeshkumar et al., in 2012, in HUVECs cells and PC3 prostate cancer cells, using in vitro, ex vivo and in vivo models. In vitro, LUT at non-toxic concentrations significantly inhibited microvessel sprouting and proliferation, migration, invasion and tube formation of endothelial cells, key events in the process of angiogenesis, which was also confirmed by ex vivo studies. LUT suppressed VEGF induced phosphorylation of VEGF receptor 2 and their downstream protein kinases AKT, ERK, mTOR, P70S6K, MMP-2 and MMP-9 in HUVECs. Proinflammatory cytokines such as IL-1b, IL-6, IL-8 and TNF-α were significantly reduced after treatment of PC3 cells with LUT. The apoptosis also was activated in PC3 cells. LUT (10 mg kg−1) significantly reduced the volume and the weight of solid tumors in the prostate xenograft mouse model.154 In another work, LUT inhibits invasion of PC3 prostate cancer cells through the induced expression of E-cadherin through MDM2. LUT inhibits MDM2 through down-regulation of AKT, suggesting that LUT regulates E-cadherin through the AKT/MDM2 pathway. In vivo experiments showed that LUT inhibited spontaneous lung metastasis of PC3 cells implanted onto the nude mice.155 LUT also acted as an antiproliferative and anti-invasion agent in LNCaP prostate cancer cells.161

In A549 and H460 non-small cell lung carcinoma, LUT induced apoptotic and non-apoptotic death mediated by the induction of superoxide radicals. JNK was potently activated after superoxide accumulation and suppression of superoxide completely blocked LUT-induced JNK activation. The induction of JNK suppressed MKP-1 and promoted cell death.156 Kim et al., in 2006, observed that caspase activation culminated in apoptosis in Lewis lung carcinoma cells treated with LUT.162 In another work, LUT blocked the NFkB pathway, sensitizing H23, H2009, H460 and A549 lung carcinoma cells to apoptosis.163

The antiproliferative effects of LUT in multi-drug resistance (MDR) breast cancer cells (ADR/RES and MCF-7/MitoR) that expressed high levels of P-glycoprotein and ABCG2 were evaluated by Rao et al., in 2012. LUT induces apoptosis in P-glycoprotein- and ABCG2-expressing MDR cancer cells without affecting the transport functions of these drug transporters. LUT induced apoptosis by ROS generation, DNA damage, activation of ATR → CHK2 → p53 signaling pathway, inhibition of NFkB signaling pathway, activation of p38 pathway and depletion of anti-apoptotic proteins.164

Co-treatments with LUT and traditional chemotherapeutic drugs. LUT sensitized the 4T1 and MCF7 breast cancer cells to DOX in vitro and in vivo. In vitro, LUT had only a slight effect on cell growth and cytotoxicity under normoxia, but it could reverse cancer resistance to DOX and promote death of cells under hypoxia. In vivo, there is no reduction in tumor growth when LUT was administered alone, but it could offer superior efficacy and lesser toxicity when administred with DOX in 4T1 and MCF-7 bearing mice. LUT was able to suppress glycolytic flux (but did not affect glucose uptake), P-glycoprotein, antioxidative enzymes under hypoxia in vitro, consequently keeping the ideal intratumoral DOX level in vivo.165

Tang et al., 2011, showed that LUT acted as a potent inhibitor of nuclear factor erythroid 2-related factor 2 (Nrf2). This transcription factor is redox-sensitive and regulates the expression of a battery of genes generally involved with cell defense, enhancing cancer cell survival and resistance to anticancer drugs. In A549 non-small lung cancer cells, which possess constitutively active Nrf2, LUT promoted a dramatic reduction in Nrf2, leading to decreased Nrf2 binding to antioxidant response elements (AREs), down-regulation of ARE-driven genes and depletion of reduced glutathione. Additionally, LUT significantly sensitized A549 cells to the anticancer drugs oxaliplatin (OXA), bleomycin (BLEO) and DOX.166

2.5. 3-O-Methylquercetin (3OMQ)

Some works evaluating anticancer properties of 3OMQ are found in literature. 3OMQ was tested in vitro, demonstrating activity against human cancer cells at low concentration (often 5–10 μM and not greater than 50 μM) in a time and dose-dependent manner. Some cellular targets were also identified in two of these papers.

Antiproliferative effect of 3OMQ was observed in mouse skin epidermal JB6P1 cells, with induction of cell cycle arrest at the G2/M phase. It also suppressed neoplastic cell transformation, with inhibitory efficacy greater than QCT. The activation of AP-1 was suppressed after the cell treatment with 3OMQ. The inhibition of ERK kinase activity and attenuated phosphorylation of ERKs were also observed.167

3OMQ caused significant growth inhibition of lapatinib (LAP) sensitive (SK–Br-3) and resistant (SK–Br-3–Lap R) breast cancer cells. 3OMQ had no effect on AKT or ERK signaling in resistant cells. However, it caused a pronounced G2/M arrest mainly through the CHK1–CDC25c–cyclin B1/CDK1 pathway in both cell types. Differently, LAP produced an accumulation of cells in G1 phase mediated through cyclin D1, but only in LAP sensitive cells. 3OMQ induced apoptosis by increasing levels of cleaved caspase-3, -7 and poly(ADP-ribose) polymerase (PARP) in both cell lines.168

Rubio et al., in 2006, evaluated the cytotoxicity of 3OMQ against several human cancer cell lines (HL60-leukemia, A431-epidermoid carcinoma, HeLa-cervical cancer, SKOV3-ovarian carcinoma and HOS-hosteosarcoma). The study also compared the cytotoxicity of 3OMQ and QCT in these cells. 3OMQ acted at a lower range and values of IC50% (15–50 μM) when compared to QCT (30–100 μM). Both were not active for HOS cells. The methylation of the hydroxyl group at position C3 of QCT yields 3OMQ, a molecule with a higher antiproliferative activity. The authors presuppose that methylated derivatives can pass through biological membranes more easily than QCT, consequently having higher intracellular concentration and lower IC50% values.169

3OMQ isolated from Inula viscosa showed antiproliferative activity against the MCF7 breast cancer cells and Hep2 hepatoma cells (IC50% values of 11.23 μg mL−1 and 26.12 μg mL−1, respectively). The toxicity was limited against Vero cells, with IC50% values above 150 μg mL−1. 3OMQ induced apoptosis in MCF7 cells and DNA fragmentation. The molecule increased the number of apoptotic cells with fragmented DNA, compared with untreated MCF7 cells. Cytomorphological alterations corresponding to typical morphology of apoptosis were detected in MCF7 cells treated with 3OMQ, that included cell shrinkage, membrane blebbing and formation of apoptotic bodies.170

2.6. Achyrobichalcone (ACB)

Achyrobichalcone (ACB) is a biflavonoid recently isolated from A. satureioides, found only in this plant so far.91,93 ACB arouses scientific interest due to its structural similarity to other bichalcones with anticancer properties.171 Furthermore, ACB integrates the class of chalcones, which are extensively studied for anticancer therapy. Some studies are running in our group with the aim of assessing the anticancer properties of the molecule. This work is reaching positive and promising results that will be published opportunely.

2.7. Clinical trials and future trends

Despite promising anticancer properties evidenced in preclinical studies for QCT and LUT, clinical trials should be conducted to confirm the chemotherapeutic properties of these molecules and to propose the possible use as adjuvants in anticancer therapy. In the literature, one Phase I clinical trial was performed for QCT.172 Their data suggested a safe dose injection of 1.4 g m−2 (about 35 mg kg−1) administered via intravenous infusion at three-week or weekly intervals. At higher doses, renal toxicity was detected. In this study, QCT inhibited the lymphocyte tyrosine kinase activity associated with the progression of cancer in some patients with hepatoma and ovarian cancer refractory to cisplatin, evidencing the antitumor activity of QCT in human. Currently, two ongoing clinical trials are evaluating the chemopreventive properties of QCT,173,174 but there is a lack of information about its chemotherapeutic properties. For LUT, no studies were found in clinical phase. Additionally, problems associated with the low bioavailability of QCT and LUT reported in pharmacokinetic studies172,175–178 in humans should also be investigated in future works. Although there is still a long run of research for the insertion of these flavonoids into the clinical routine, the overwhelming data obtained so far for QCT and LUT support the anticancer potential of these molecules.

3. Concluding remarks

Flavonoids from A. satureioides, especially QCT and LUT, are multi-target anticancer agents by modulation of multiple proteins, being advantageous by conducting to a higher clinical efficacy when compared with target-selective drugs. A wide range of cancers were affected after treatment with flavonoids, both when administered alone or combined to traditional chemotherapeutic drugs. In this sense, the successful anticancer preclinical studies with these flavonoids (especially QCT) encourage the clinical trials in human and, possibly, these molecules will emerge as strong candidates to anticancer drugs in the future.

Conflict of interest

The authors have declared no conflict of interest.

Abbreviations

MAPKMitogen-activated protein kinase
PKCProtein kinase C
PI3K or AKTPhosphatidylinositol 3-kinase
NFkBNuclear factor kappa B
CDKs(1–9)(1–9)Cyclin-dependent kinases
ERKExtracellular signal-regulated kinase
VEGFVascular endothelial growth factor
IGF-IRInsulin-like growth factor-I receptor
P-gpP-glycoprotein
Cip1/Waf1Cyclin-dependent kinase inhibitor p21
AIFApoptosis-inducing factor
Nrf2Nuclear factor erythroid 2-related factor 2
PARPPoly(ADP-ribose) polymerase
BaxBcl-2-associated X protein
PUMAp53 Upregulated modulator of apoptosis or BBC3, Bcl-2-binding component 3
NOXAPhorbol-12-myristate-13-acetate-induced protein 1
BakBcl-2 homologous antagonist/killer
BidBH3 interacting-domain death agonist
Bcl-2Derived from B-cell lymphoma 2
Bcl-xLDerived from B-cell lymphoma-extra large
Mcl-1Induced myeloid leukemia cell differentiation protein
IAPInhibitor of apoptosis
p53Protein 53 or tumor protein 53
TRAILTNF-related apoptosis-inducing ligand
survivin or BIRC5baculoviral inhibitor of apoptosis repeat-containing 5
ILInterleucine
STAT3Signal transducer and activator of transcription 3
MMPMatrix metalloproteinase
COX-2Cyclooxygenase-2
PLD1Phospholipase D1
p21Cyclin-dependent kinase inhibitor 1 or CDK-interacting protein 1
NAG-1Nonsteroidal anti-inflammatory drug-activated gene
EGR-1Early growth response protein 1
HIF-1αHypoxia-inducible factor 1-α
FasL or CD95LFas ligand
p57Cyclin-dependent kinase inhibitor 1C
ΔΨmMitochondrial transmembrane potential
mTORmammalian target of rapamycin
P70S6Kserine/threonine kinase
TNFαTumor necrosis factor
JNKc-Jun NH2-terminal kinase
PLK1Serine/threonine-protein kinase
ABCG2ATP-binding cassette sub-family G member 2
MKP-1Mitogen-activated protein kinase phosphatases
AREAntioxidant responsive element
ROSReactive oxygen species
QCTQuercetin
LUTLuteolin
3OMQ3-O-methylquercetin
ACBAchyrobichalcone
DOXDoxorubicin
TMZTemozolomide
CISCisplatin
VCRVincristine
DAUDaunorubicin
5-FU5-Fluorouracil
ETOEtoposide
MDRMulti-drug resistance
OXAOxaliplatin
BLEOBleomycin
LAPLapatinib.

Acknowledgements

The authors are grateful to Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) for the financial support and scholarship.

References

  1. R. Siegel, D. Naishadham and A. Jemal, Cancer Stat., 2013, 63, 11–30 Search PubMed.
  2. J. Ferlay, H. R. Shin, F. Bray, D. Forman, C. D. Mathers and D. Parkin, GLOBOCAN 2008, Cancer incidence and mortality worldwide: IARC Cancer Base n° 10, http://globocan.iarc.fr, International Agency for Research on Cancer, Lyon, France, 2010, last accessed 25/02/2013 Search PubMed.
  3. INCA, I. Nacional de Câncer José Alencar Gomes da Silva, Coordenação Geral de Ações Estratégicas, Coordenação de Prevenção e Vigilância, Estimativa 2012: incidência de câncer no Brasil, 2011, Rio de Janeiro.
  4. G. M. Cragg, D. G. I. Kingston and D. J. Newman, Anticancer agents from natural products, Taylor & Francis, Boca Raton, 2005 Search PubMed.
  5. F. E. Koehn and G. T. Carter, Nat. Rev. Drug Discovery, 2005, 4, 206–220 CrossRef CAS PubMed.
  6. H. Ji, X. Li and H. Zhang, EMBO J., 2009, 10, 194–200 CrossRef CAS PubMed.
  7. A. Harvey, Drug Discovery Today, 2000, 5, 294–300 CrossRef PubMed.
  8. D. J. Newman and G. M. Cragg, J. Nat. Prod., 2012, 75, 311–335 CrossRef CAS PubMed.
  9. P. Russo, A. Del Bufalo and A. Cesario, Curr. Med. Chem., 2012, 19, 5287–5293 CrossRef CAS PubMed.
  10. P. Knekt, R. Järvinen, R. Seppänen, M. Heliövaara, L. Teppo, E. Pukkala and A. Aromaa, Am. J. Epidemiol., 1997, 146, 223–230 CrossRef CAS PubMed.
  11. W. Ren, Z. Qiao, H. Wang, L. Zhu and L. Zhang, Med. Res. Rev., 2003, 23, 519–534 CrossRef CAS PubMed.
  12. A. Ghasemzadeh and N. Ghasemzadeh, J. Med. Plants Res., 2011, 5, 6697–6703 CAS.
  13. S. Cimino, G. Sortino, V. Favilla, T. Castelli, M. Madonia, S. Sansalone, G. I. Russo and G. Morgia, Oxid. Med. Cell. Longevity, 2012, 1–8 CrossRef PubMed.
  14. J. García-Tirado, C. Rieger-Reyes and P. Saz-Peiró, Medicina Clínica, 2012, 139, 358–363 CrossRef PubMed.
  15. A. G. Mercader and A. B. Pomilio, Curr. Med. Chem., 2012, 19, 4324–4347 CrossRef CAS PubMed.
  16. K. Michalak and O. Wesołowska, Anti-Cancer Agents Med. Chem., 2012, 12, 880–890 CrossRef CAS PubMed.
  17. E. Park and J. M. Pezzuto, Anti-Cancer Agents Med. Chem., 2012, 12, 836–851 CrossRef CAS PubMed.
  18. D. F. Romagnolo and O. I. Selmin, J. Nutr. Gerontol. Geriatr., 2012, 31, 206–238 CrossRef PubMed.
  19. C. Spatafora and C. Tringali, Anti-Cancer Agents Med. Chem., 2012, 12, 902–918 CrossRef CAS PubMed.
  20. D. Vira, S. K. Basak, M. S. Veena, M. B. Wang, R. K. Batra and E. S. Srivatsan, Cancer Metastasis Rev., 2012, 31, 733–751 CrossRef CAS PubMed.
  21. C. Weng and G. Yen, Cancer Metastasis Rev., 2012, 31, 323–351 CrossRef CAS PubMed.
  22. C. Weng and G. Yen, Cancer Treat. Rev., 2012, 38, 76–87 CrossRef CAS PubMed.
  23. Y. J. Surh, Nat. Rev. Cancer, 2003, 10, 768–780 CrossRef PubMed.
  24. M. Kamsteeg, T. Rutherford, E. Sapi, B. Hanczaruk, S. Shahabi, M. Flick, D. Brown and G. Mor, Oncogene, 2003, 22, 2611–2620 CrossRef CAS PubMed.
  25. A. B. Alvero, D. O'Malley, D. Brown, G. Kelly, M. Garg, W. Chen, T. Rutherford and G. Mor, Cancer, 2006, 106, 599–608 CrossRef CAS PubMed.
  26. P. L. De Souza, W. Liauw, M. Links, S. Pirabhahar, G. Kelly and L. G. Howes, Cancer Chemother. Pharmacol., 2006, 58, 427–433 CrossRef PubMed.
  27. J. R. Gamble, P. Xia, C. N. Hahn, J. J. Drew, C. J. Drogemuller, D. Brown and M. A. Vadas, Int. J. Cancer, 2006, 118, 2412–2420 CrossRef CAS PubMed.
  28. S. Mahoney, F. Arfuso, P. Rogers, S. Hisheh, D. Brown, M. Millward and A. Dharmarajan, J. Biosci., 2012, 37, 73–84 CrossRef CAS PubMed.
  29. F. Yu, R. N. Watts, X. D. Zhang, J. M. Borrow and P. Hersey, Anti-Cancer Drugs, 2006, 17, 1151–1161 CrossRef CAS PubMed.
  30. H. M. Kluger, M. M. McCarthy, A. B. Alvero, M. Sznol, S. Ariyan, R. L. Camp, D. L. Rimm and G. Mor, J. Transl. Med., 2007, 5, 1–15 CrossRef PubMed.
  31. M. F. Aguero, M. M. Facchinetti, Z. Sheleg and A. M. Senderowicz, Cancer Res., 2005, 65, 3364–3373 CAS.
  32. Clinical Trials, United State National Institutes of Health, 2013, http://clinicaltrials.gov/ct2/results?term=phenoxodiol%26Search=Search, last access: 28/02/2013 Search PubMed.
  33. E. Sapi, A. B. Alvero, W. Chen, D. O'Malley, X. Y. Hao, B. Dwipoyono, M. Garg, M. Kamsteeg, T. Rutherford and G. Mor, Oncol. Res., 2004, 14, 567–578 CAS.
  34. A. I. Constantinou and A. Husband, Anticancer Res., 2002, 22, 2581–2585 CAS.
  35. J. B. Howes, P. L. De Souza, L. West, L. J. Huang and L. G. Howes, BMC Clin. Pharmacol., 2011, 11, 3 CrossRef PubMed.
  36. M. G. Kelly, G. Mor, A. Husband, D. M. O'Malley, L. Baker, M. Azodi, P. E. Schwartz and T. Rutherford, Int. J. Gynecol. Cancer, 2011, 21, 633–639 CrossRef PubMed.
  37. L. Wu, T. De Luca, T. Watanabe, D. M. Morré and D. J. Morré, Biochim. Biophys. Acta, 2011, 1810, 784–789 CrossRef CAS PubMed.
  38. C. Yao, S. Wu, D. Li, H. Ding, Z. Wang, Y. Yang, S. Yan and Z. Gu, Mol. Oncol., 2012, 6, 392–404 CrossRef CAS PubMed.
  39. Marshall Edwards Pharma, 2013, available in: http://www.meipharma.com/, last acess: 28/02/2013.
  40. M. V. Blagosklonny, Cell cycle, 2004, 3, 1537–1542 CrossRef CAS PubMed.
  41. C. McInnes, Drug Discovery Today, 2008, 13, 875–881 CrossRef CAS PubMed.
  42. M. Guha, Nat. Rev. Drug Discovery, 2012, 11, 892–894 CrossRef PubMed.
  43. Clinical Trials, United State National Institutes of Health, 2013, http://www.clinicaltrials.gov/ct2/results?term=flavopiridol%26Search=Search, last access: 01/03/2013 Search PubMed.
  44. S. G. D. Oliveira, F. R. R. De Moura, F. F. Demarco, P. S. Nascente, F. A. B. Del Pino and R. G. Lund, J. Ethnopharmacol., 2012, 140, 428–437 CrossRef PubMed.
  45. C. M. O. Simões, E. P. Schenkel, L. Bauer and A. A. Langeloh, J. Ethnopharmacol., 1988, 22, 281–293 CrossRef.
  46. L. Silva and A. Langeloh, Lat. Am. J. Pharm., 1994, 13, 35–40 Search PubMed.
  47. C. M. O. Simões, L. A. Mentz, E. P. Schenkel, B. F. Irgang and J. R. Stehmann, Plantas da Medicina Popular no Rio Grande do Sul, Editora da Universidade Federal do Rio Grande do Sul, Porto Alegre, 1998 Search PubMed.
  48. C. Kadarian, A. M. Broussalis, J. Miño, P. López, S. Gorzalczany, G. Ferraro and C. Acevedo, Pharmacol. Res., 2002, 45, 57–61 CrossRef CAS PubMed.
  49. K. C. B. De Souza, V. L. Bassani and E. E. S. Schapoval, Phytomedicine, 2007, 14, 102–108 CrossRef CAS PubMed.
  50. J. F. Morton, Q. J. Crude Drug Res., 1975, 13, 97–121 Search PubMed.
  51. H. Wagner, A. Proksch, R. M. Vollmar, S. Odenthal, H. Stuppner, K. Jurcic, M. Le Turdu and J. N. Fang, Arzneimittelforschung, 1985, 35, 1069–1075 CAS.
  52. J. Puhlmann, U. Knaus, L. Tubaro, W. Schaefer and H. Wagner, Phytochemistry, 1992, 31, 2617–2621 CrossRef CAS PubMed.
  53. C. Anesini and C. Perez, J. Ethnopharmacol., 1993, 39, 119–128 CrossRef CAS PubMed.
  54. M. Arisawa, Nat. Med., 1994, 48, 338–347 Search PubMed.
  55. A. R. De Arias, E. Ferro, A. Inchausti, M. Ascurra, N. Acosta, E. Rodriguez and A. Fournet, J. Ethnopharmacol., 1995, 45, 35–41 CrossRef.
  56. C. Desmarchelier, J. Coussio and G. Ciccia, Braz. J. Med. Biol. Res., 1998, 31, 1163–1170 CrossRef CAS PubMed.
  57. A. L. G. Santos, D. Ripoll, N. Nardi and V. L. Bassani, Phytother. Res., 1999, 13, 65–66 CrossRef CAS PubMed.
  58. M. A. C. Santos and E. Elisabetsky, Rev. Bras. Plant. Med., 1999, 2, 7–17 Search PubMed.
  59. S. M. Zanon, F. S. Ceriatti, M. Rovera, L. J. Sabini and B. A. Ramos, Rev. Latinoam. Microbiol., 1999, 41, 59–62 CAS.
  60. J. G. Graham, M. L. Quinn, D. S. Fabricant and N. R. Farnsworth, J. Ethnopharmacol., 2000, 73, 347–377 CrossRef CAS PubMed.
  61. J. R. Carney, J. M. Krenisky, R. T. Williamson and J. Luo, J. Nat. Prod., 2002, 65, 203–205 CrossRef CAS.
  62. A. Gugliucci and T. Menini, Life Sci., 2002, 71, 693–705 CrossRef CAS PubMed.
  63. M. J. Ruffa, G. Ferraro, M. L. Wagner, M. L. Calcagno, R. H. Campos and L. Cavallaro, J. Ethnopharmacol., 2002, 79, 335–339 CrossRef CAS PubMed.
  64. M. F. Arredondo, F. Blasina, C. Echeverry, A. Morquio, M. Ferreira, J. A. Abin-Carriquiry, L. Lafon and F. Dajas, J. Ethnopharmacol., 2004, 91, 13–20 CrossRef CAS PubMed.
  65. J. M. R. Bettega, H. F. Teixeira, V. L. Bassani, C. R. M. Barardi and C. M. O. Simões, Phytother. Res., 2004, 18, 819–823 CrossRef CAS PubMed.
  66. O. Hnatyzyn, V. Moscatelli, R. Rondina, M. Costa, C. Arranz, A. Balaszczuck, J. Coussio and G. Ferraro, Phytomedicine, 2004, 11, 366–369 CrossRef PubMed.
  67. M. Polydoro, K. C. B. De Souza, M. E. Andrades, E. G. Da Silva, F. Bonatto, J. Heydrich, F. Dal-Pizzol, E. E. S. Schapoval, V. L. Bassani and J. C. F. Moreira, Life Sci., 2004, 74, 2815–2826 CrossRef CAS PubMed.
  68. M. C. T. Duarte, G. M. Figueira, A. Sartoratto, V. L. G. Rehder and C. Delarmelina, J. Ethnopharmacol., 2005, 97, 305–311 CrossRef PubMed.
  69. A. Morquio, F. Rivera-Megret and F. Dajas, Phytother. Res., 2005, 19, 486–490 CrossRef PubMed.
  70. D. Calvo, L. N. Cariddi, M. Grosso, M. S. Demo and A. M. Maldonado, Rev. Latinoam. Microbiol., 2006, 48, 247–255 CAS.
  71. M. L. Dickel, S. M. K. Rates and R. Ritter, J. Ethnopharmacol., 2007, 109, 60–71 CrossRef PubMed.
  72. M. Consentino, R. Bombelli, E. Carcano, A. Luini, M. Franca, F. Crema, F. Dajas and S. Lecchini, J. Ethnopharmacol., 2008, 116, 501–507 CrossRef PubMed.
  73. G. Ferraro, C. Anesini, A. Ouvina, D. Retta, R. Filip, M. Gattuso, S. Gattuso, O. Hnatyszyn and A. Bandoni, Lat. Am. J. Pharm., 2008, 27, 626–628 CAS.
  74. M. F. Blasina, L. Vaamonde, A. Morquio, C. Echeverry, F. Arredondo and F. Dajas, Phytother. Res., 2009, 23, 1263–1269 CrossRef CAS PubMed.
  75. C. L. C. Brandelli, R. B. Giordani, G. A. De Carli and T. Tasca, Parasitol. Res., 2009, 104, 1345–1349 CrossRef PubMed.
  76. M. J. González and J. M. Marioli, J. Invertebr. Pathol., 2010, 104, 209–213 CrossRef PubMed.
  77. J. R. Santin, M. Lemos, L. C. Klein Junior, R. Niero and S. F. De Andrade, J. Ethnopharmacol., 2010, 130, 334–339 CrossRef PubMed.
  78. M. B. Joray, M. R. Rollán, G. M. Ruiz, S. M. Palacios and M. C. Carpinella, Planta Med., 2011, 77, 95–100 CrossRef CAS PubMed.
  79. D. C. Sabaté, M. J. Gonzaléz, M. P. Porrini, M. J. Eguaras, M. C. Audisio and J. M. Marioli, World J. Microbiol. Biotechnol., 2012, 28, 1415–1422 CrossRef PubMed.
  80. D. C. Espiña, F. B. Carvalho, D. Zanini, J. B. Schlemmer, J. D. Coracini, M. A. Rubin, V. M. Morsch, M. R. C. Schetinger, D. B. R. Leal, C. R. Baiotto and J. A. S. Jaques, Cell Biochem. Funct., 2012, 30, 347–353 CrossRef PubMed.
  81. P. Faral-Tello, S. Mirazo, C. Dutra, A. Pérez, L. Geis-Asteggiante, S. Frabasile, E. Koncke, D. Davyt, L. Cavallaro, H. Heinzen and J. Arbiza, Sci. World J., 2012, 1–5 CrossRef PubMed , Article ID 174837.
  82. M. C. Sabini, F. M. Escobar, C. E. Tonn, S. M. Zanon, M. S. Contigiani and L. I. Sabini, Nat. Prod. Res., 2012, 26, 405–415 CrossRef CAS PubMed.
  83. M. Trojan-Rodrigues, T. L. S. Alves, G. L. G. Soares and M. R. Ritter, J. Ethnopharmacol., 2012, 139, 155–163 CrossRef CAS PubMed.
  84. M. B. Joray, S. M. Palacios and M. C. Carpinella, Phytomedicine, 2013, 20, 258–261 CrossRef CAS PubMed.
  85. H. Garcia-Barriga, Flora medicinal de Colombia, Universidad Nacional, Bogota, vol. 2, pp. 3, 1975 Search PubMed.
  86. M. Vanini, R. L. Barbieri, R. M. Heck and E. Schwartz, Enfermería Global, 2011, 21, 1–6 Search PubMed.
  87. J. M. Fachinetto, M. D. Bagatini, J. Durigon, A. C. F. Da Silva and S. B. Tedesco, Rev. Bras. Farmacogn., 2007, 17, 49–54 Search PubMed.
  88. F. Rivera, E. Gervaz, C. Sere and F. Dajas, J. Ethnopharmacol., 2004, 95, 359–362 CrossRef CAS PubMed.
  89. D. Retta, E. Dellacassa, J. Villamil, S. A. Suárez and A. L. Bandoni, Ind. Crops Prod., 2012, 38, 27–38 CrossRef.
  90. K. C. B. De Souza, E. E. S. Schapoval and V. L. Bassani, J. Pharm. Biomed. Anal., 2002, 28, 771–777 CrossRef CAS PubMed.
  91. M. H. Holzschuh, G. Gosmann, P. H. Schneider, E. E. S. Schapoval and V. L. Bassani, Pharmazie, 2010, 65, 650–656 CAS.
  92. J. P. Carini, S. Kaiser, G. G. Ortega and V. L. Bassani, Phytochem. Anal., 2013, 24, 193–200 CrossRef CAS PubMed.
  93. J. P. Carini, G. G. Leitão, P. H. Schneider, C. C. Santos, F. N. Costa and V. L. Bassani, J. Med. Food, 2013 Search PubMed , manuscript submitted.
  94. F. Dajas, J. Ethnopharmacol., 2012, 143, 383–396 CrossRef CAS PubMed.
  95. G. K. Schwartz and M. A. Shah, J. Clin. Oncol., 2005, 23, 9408–9421 CrossRef CAS PubMed.
  96. A. B. Granado-Serrano, M. A. Martin, L. Bravo, L. Goya and S. Ramos, J. Nutr., 2006, 136, 2715–2721 CAS.
  97. D. H. Lee, M. Szczepanski and Y. L. Lee, Biochem. Pharmacol., 2008, 75, 2345–2355 CrossRef CAS PubMed.
  98. R. V. Priyadarsini, R. S. Murugan, S. Maitreyi, K. Ramalingam, D. Karunagaran and S. Nagini, Eur. J. Pharmacol., 2010, 649, 84–91 CrossRef PubMed.
  99. G. Niu, S. Yin, S. Xie, Y. Li, D. Nie, L. Ma, X. Wang and Y. Wu, Acta Biochim. Biophys. Sin., 2011, 43, 30–37 CrossRef CAS PubMed.
  100. S. Borska, M. Chmielewska, T. Wysocka, M. Drag-Zalesinska, M. Zabel and P. Dziegiel, Food Chem. Toxicol., 2012, 50, 3375–3383 CrossRef CAS PubMed.
  101. C. Spagnuolo, M. Russo, S. Bilotto, I. Tedesco, B. Laratta and G. L. Russo, Ann. N. Y. Acad. Sci., 2012, 1259, 95–103 CrossRef CAS PubMed.
  102. Y. Kim, W. Kim and E. Cha, Korean J. Physiol. Pharmacol., 2011, 15, 279–283 CrossRef CAS PubMed.
  103. L. Rockenbach, L. Bavaresco, P. F. Farias, A. R. Cappellari, C. H. Barrios, F. B. Morrone and A. M. O. Battastini, Urol. Oncol., 2013, 31, 1204–1211 CrossRef CAS PubMed.
  104. L. Ma, J. M. Feugang, P. Konarski, J. Wang, J. Lu, S. Fu, B. Ma, B. Tian, C. Zou and Z. Wang, Front. Biosci., 2006, 11, 2275–2285 CrossRef CAS.
  105. J. Michaud-Levesque, N. Bousquet-Gagnon and R. Béliveau, Exp. Cell Res., 2012, 318, 925–935 CrossRef CAS PubMed.
  106. M. H. Park and D. S. Min, Biochem. Biophys. Res. Commun., 2011, 412, 710–715 CrossRef CAS PubMed.
  107. E. J. Kim, C. H. Choi, J. Y. Park, S. K. Kang and Y. K. Kim, Neurochem. Res., 2008, 33, 971–979 CrossRef CAS PubMed.
  108. H. Kim, S. Kim, B. Kim, S. Lee, Y. Park, B. Park, S. Kim, J. Kim, C. Choi, J. Kim, S. Cho, J. Jung, K. Roh, K. Kang and J. Jung, J. Agric. Food Chem., 2010, 58, 8643–8650 CrossRef CAS PubMed.
  109. Y. Lee, S. Y. Park, Y. Kim, W. S. Lee and O. J. Park, Exp. Mol. Med., 2009, 41, 201–207 CrossRef CAS PubMed.
  110. J. H. Lim, J.-W. Park, D. S. Min, J.-S. Chang, Y. H. Lee, Y. B. Park, K. S. Choi and T. K. Kwon, Apoptosis, 2007, 12, 411–421 CrossRef CAS PubMed.
  111. J. Tan, B. Wang and L. Zhu, Chem. Biodiversity, 2009, 6, 1101–1110 CAS.
  112. P. Wang, K. Zhang, Q. Zhang, J. Mei, C. Chen, Z. Feng and D. Yu, Toxicol. in Vitro, 2012, 26, 221–228 CrossRef CAS PubMed.
  113. C. B. Avci, S. Yilmaz, Z. O. Dogan, G. Saydam, Y. Dodurga, H. A. Ekiz, M. Karta, F. Sahin, Y. Baran and C. Gunduz, Hematology, 2011, 16, 303–307 CrossRef CAS PubMed.
  114. W. Lee, Y. Chen and T. Tseng, Oncol. Rep., 2011, 25, 583–591 CAS.
  115. Y. Lee and O. J. Park, Food Sci. Biotechnol., 2011, 20, 371–375 CrossRef CAS.
  116. C. Chou, J. Yang, H. Lu, S. Ip, C. Lo, C. Wu, J. Lin, N. Tang, J. Chung, M. Chou, Y. Teng and D. Chen, Arch. Pharmacal Res., 2010, 33, 1181–1191 CrossRef CAS PubMed.
  117. X. Zhang, S. Huang and Q. Xu, Cancer Chemother. Pharmacol., 2004, 53, 82–88 CrossRef CAS PubMed.
  118. K. Liu, C. Yen, R. S. Wu, J. Yang, H. Lu, K. Lu, C. Lo, H. Chen, N. Tang, C. Wu and J. Chung, Environ. Toxicol., 2012 DOI:10.1002/tox.21769.
  119. P. Pratheeshkumar, A. Budhraja, Y. Son, X. Wang, Z. Zhang, S. Ding, L. Wang, A. Hitron, J. Lee, M. Xu, G. Chen, J. Luo and X. Shi, PLoS One, 2012, 7, 1–10 Search PubMed.
  120. J. Tan, L.-C. Zhu and B.-C. Wang, Chinese Pharmacol. Bull., 2008, 24, 1220–1224 CAS.
  121. P. Kuo, H. Liu and J. Chao, J. Biol. Chem., 2004, 279, 55875–55885 CrossRef CAS PubMed.
  122. T. T. T. Nguyen, E. Tran, T. H. Nguyen, P. T. Do, T. H. Huynh and H. Huynh, Carcinogenesis, 2004, 25, 647–659 CrossRef CAS PubMed.
  123. R. Narayansingh and R. A. R. Hurta, J. Sci. Food Agric., 2009, 89, 542–547 CrossRef.
  124. S. Tanigawa, M. Fujii and D. Hou, Biosci., Biotechnol., Biochem., 2008, 72, 797–804 CrossRef CAS PubMed.
  125. C. M. J. Conklin, J. F. Bechberger, D. MacFabe, N. Guthrie, E. M. Kurowska and C. C. Naus, Carcinogenesis, 2007, 28, 93–100 CrossRef CAS PubMed.
  126. S. Chien, Y. Wu, J. Chung, J. Yang, H. Lu, M. Tsou, W. G. Wood, S. Kuo and D. Chen, Hum. Exp. Toxicol., 2009, 28, 493–503 CAS.
  127. E. J. Choi, S. M. Bae and W. S. Ahn, Arch. Pharmacal Res., 2008, 31, 1281–1285 CrossRef CAS PubMed.
  128. M. R. Noori-Daloii, M. Momeny, M. Yousefi, F. G. Shirazi, M. Yaseri, N. Motamed, N. Kazemialiakbar and S. Hashemi, Med. Pediatr. Oncol., 2011, 28, 1395–1404 CrossRef CAS PubMed.
  129. K. Senthilkumar, R. Arunkumar, P. Elumalai, G. Sharmila, D. N. Gunadharini, S. Banudevi, G. Krishnamoorthy, C. S. Benson and J. Arunakaran, Cell Biochem. Funct., 2011, 29, 87–95 CrossRef CAS PubMed.
  130. K. Senthilkumar, P. Elumalai, A. Arunkumar, S. Banudevi, N. D. Gunadharini, G. Sharmila, K. Selvakumar and J. Arunakaran, Mol. Cell. Biochem., 2010, 344, 173–184 CrossRef CAS PubMed.
  131. C. Tsimplouli, C. Demetzos, M. Hadzopoulou-Cladaras, P. Pantazis and K. Dimas, Eur. J. Nutr., 2012, 51, 181–190 CrossRef CAS PubMed.
  132. G. Wang, J. Zhang, L. Liu, S. Sharma and Q. Dong, PLoS One, 2012, 7, 1–12 Search PubMed.
  133. D. Staedler, E. Idrizi, B. H. Kenzaoui and L. Juillerat-Jeanneret, Cancer Chemother. Pharmacol., 2011, 68, 1161–1172 CrossRef CAS PubMed.
  134. G. Du, H. Lin, Y. Yang, S. Zhang, X. Wu, M. Wang, L. Ji, L. Lu, L. Yu and G. Han, Int. Immunopharmacol., 2010, 10, 819–826 CrossRef CAS PubMed.
  135. M. Russo, C. Spagnuolo, S. Volpe, A. Mupo, I. Tedesco and G.-L. Russo, Br. J. Cancer, 2010, 103, 642–648 CrossRef CAS PubMed.
  136. A. M. Ramos and P. Aller, Biochem. Pharmacol., 2008, 75, 1912–1923 CrossRef CAS PubMed.
  137. T. Thangasamy, S. Sittadjody, G. C. Mitchell, E. E. Mendoza, V. M. Radhakrishnan, K. H. Limesand and R. Burd, BMC Cancer, 2010, 10, 1–10 CrossRef PubMed.
  138. A. Demiroglu-Zergeroglu, B. Basara-Cigerim, E. Kilic and G. Yanikkaya-Demirel, J. Biomed. Biotechnol., 2010, 1–7 CrossRef PubMed , Article ID 851589.
  139. M. Daker, M. Ahmad and A. S. B. Khoo, Cancer Cell Int., 2012, 12, 1–8 CrossRef PubMed.
  140. X. Zhan, R. Zhang, Y. Xu, S. Yang, D. Xie and L. Tan, Chin.-Ger. J. Clin. Oncol., 2012, 11, 380–383 CrossRef CAS.
  141. W. Chen, X. Wang, J. Zhuang, L. Zhang and Y. Lin, Carcinogenesis, 2007, 28, 2114–2121 CrossRef CAS PubMed.
  142. S. Borska, M. Drag-Zalesinska, T. Wysocka, M. Sopel, M. Dumanska, M. Zabel and P. Dziegiel, Folia Histochem. Cytobiol., 2010, 48, 222–229 Search PubMed.
  143. C. P. R. Xavier, C. F. Lima, M. Rohde and C. Pereira-Wilson, Cancer Chemother. Pharmacol., 2011, 68, 1449–1457 CrossRef CAS PubMed.
  144. H. Kim, T. Wannatung, S. Lee, W. K. Yang, S. H. Chung, J. Lim, W. Choe, I. Kang, S. Kim and J. Há, Apoptosis, 2012, 17, 938–949 CrossRef CAS PubMed.
  145. S. Kilani-Jaziri, V. Frachet, W. Bhouri, K. Ghedira, L. Chekir-Ghedira and X. Ronot, Drug Chem. Toxicol., 2012, 35, 1–10 CrossRef PubMed.
  146. R. Shi, C. Ong and H. Shen, Oncogene, 2004, 23, 7712–7721 CrossRef CAS PubMed.
  147. D. Y. Lim, Y. Jeong, A. L. Tyner and J. H. Y. Park, Am. J. Physiol.: Gastrointest. Liver Physiol., 2007, 292, G66–G75 CrossRef CAS PubMed.
  148. J. Hwang, O. J. Park, Y. K. Lee, M. J. Sung, H. J. Hur, M. S. Kim, J. H. Ha and D. Y. Kwon, Int. J. Mol. Med., 2011, 28, 25–31 CAS.
  149. K. Selvendiran, H. Koga, T. Ueno, T. Yoshida, M. Maeyama, T. Torimura, H. Yano, M. Kojiro and M. Sata, Cancer Res., 2006, 66, 4826–4834 CrossRef CAS PubMed.
  150. B. Wu, Q. Zhang, W. Shen and J. Zhu, Mol. Cell. Biochem., 2008, 313, 125–132 CrossRef CAS PubMed.
  151. W. G. Ko, T. H. Kang, S. J. Lee, Y. C. Kim and B. H. Lee, Phytother. Res., 2002, 16, 295–298 CrossRef CAS PubMed.
  152. M. J. Kim, J. S. Woo, C. H. Kwon, J. H. Kim, Y. K. Kim and K. H. Kim, Cell Biol. Int., 2012, 36, 339–344 CrossRef CAS PubMed.
  153. J. Ruan, Y. Liu, L. Zhang, L. Yan, F. Fan, C. Shen, A. Wang, S. Zheng, S. Wang and Y. Lu, Acta Pharmacol. Sin., 2012, 33, 1325–1331 CrossRef CAS PubMed.
  154. P. Pratheeshkumar, Y. Son, A. Budhraja, X. Wang, S. Ding, L. Wang, A. Hitron, J. Lee, D. Kim, S. P. Divya, G. Chen, Z. Zhang, J. Luo and X. Shi, PLoS One, 2012, 7, 1–15 Search PubMed.
  155. Q. Zhou, B. Yan, X. Hu, X. Li, J. Zhang and J. Fang, Mol. Cancer Ther., 2009, 8, 1684–1691 CrossRef CAS PubMed.
  156. L. Bai, X. Xu, Q. Wang, S. Xu, W. Ju, X. Wang, W. Chen, W. He, H. Tang and Y. Lin, Mol. Pharmacol., 2012, 81, 549–555 CrossRef CAS PubMed.
  157. X. Cai, T. Ye, C. Liu, W. Lu, M. Lu, J. Zhang, M. Wang and P. Cao, Toxicol. In Vitro, 2011, 25, 1385–1391 CrossRef CAS PubMed.
  158. Y. Zhao, G. Yang, D. Ren, X. Zhang, Q. Yin and X. Sun, Mol. Biol. Rep., 2011, 38, 1115–1119 CrossRef CAS PubMed.
  159. D. Y. Lim, H. J. Cho, J. Kim, C. W. Nho, K. W. Lee and J. H. Y. Park, BMC Gastroenterol., 2012, 12, 1–10 CrossRef PubMed.
  160. E. Lee, S. Oh and M. Sung, Food Chem. Toxicol., 2012, 50, 4136–4143 CrossRef CAS PubMed.
  161. K. Tsui, L. Chung, T. Feng, P. Chang and H. Juang, Int. J. Cancer, 2012, 130, 281–282 CrossRef PubMed.
  162. J. Kim, E. Lee, H. Lee, J. Ku, M. Lee, D. Yang and S. Kim, Ann. N. Y. Acad. Sci., 2006, 1090, 147–160 CrossRef CAS PubMed.
  163. W. Ju, X. Wang, H. Shi, W. Chen, S. A. Belinsky and Y. Lin, Mol. Pharmacol., 2007, 71, 1381–1388 CrossRef CAS PubMed.
  164. P. S. Rao, A. Satelli, M. Moridani, M. Jenkins and U. S. Rao, Int. J. Cancer, 2012, 130, 2703–2714 CrossRef CAS PubMed.
  165. G. Du, Z. Song, H. Lin, X. Han, S. Zhang and Y. Yang, Biochem. Biophys. Res. Commun., 2008, 372, 497–502 CrossRef CAS PubMed.
  166. X. Tang, H. Wang, L. Fan, X. Wu, A. Xin, H. Ren and X. J. Wang, Free Radical Biol. Med., 2011, 50, 1599–1609 CrossRef CAS PubMed.
  167. J. Li, M. Mottama, H. Li, K. Liu, F. Zhu, Y. Cho, C. P. Sosa, K. Zhou, G. T. Bowden, A. M. Bode and Z. Dong, Carcinogenesis, 2012, 33, 459–465 CrossRef CAS PubMed.
  168. J. Li, F. Zhu, R. A. Lubet, A. De Luca, C. Grubbs, M. E. Ericson, A. D'Alessio, N. Normanno, Z. Dong and A. M. Bode1, Mol. Carcinog., 2013, 52, 134–143 CrossRef PubMed.
  169. S. Rubio, J. Quintana, M. López, J. L. Eiroa, J. Triana and F. Estévez, Eur. J. Pharmacol., 2006, 548, 9–20 CrossRef CAS PubMed.
  170. W. H. Talib, M. H. A. Zarga and A. M. Mahasneh, Molecules, 2012, 17, 3291–3303 CrossRef CAS PubMed.
  171. L. K. Mdee, S. O. Yeboah and B. M. Abegaz, J. Nat. Prod., 2003, 66, 559–604 CrossRef PubMed.
  172. D. R. Ferry, A. Smith, J. Malkhandi, D. W. Fyfe, P. G. deTakats, D. Anderson, J. Baker and D. J. Kerr, Clin. Cancer Res., 1996, 2, 659–668 CAS.
  173. Clinical Trials, United State National Institutes of Health, 2013, http://clinicaltrials.gov/ct2/show/NCT01538316?term=quercetin%26rank=9, last access: 03/10/2013 Search PubMed.
  174. Clinical Trials, United State National Institutes of Health, 2013, http://clinicaltrials.gov/ct2/show/NCT00003365?term=quercetin%26rank=19, last acess: 03/10/2013 Search PubMed.
  175. P. C. H. Hollman, M. V. D. Gaag, M. J. B. Mengelers, J. M. P. Van Trijp, J. H. M. De Wries and M. B. Katan, Free Radical Biol. Med., 1996, 21, 703–707 CrossRef CAS PubMed.
  176. P. C. Hollman and M. B. Katan, Biomed. Pharmacother., 1997, 51, 305–310 CrossRef CAS.
  177. E. U. Graefe, H. Derendorf and M. Veit, Int. J. Clin. Pharmacol. Ther., 1999, 37, 219–233 CAS.
  178. Y. J. Moon, L. Wang, R. DiCenzo and M. E. Morris, Biopharm. Drug Dispos., 2008, 29, 205–217 CrossRef CAS PubMed.

This journal is © The Royal Society of Chemistry 2014
Click here to see how this site uses Cookies. View our privacy policy here.