IMR Press / FBL / Volume 24 / Issue 6 / DOI: 10.2741/4771
Review
Nano-engineered flavonoids for cancer protection
Show Less
1 Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
2 School of Pharmacy and Technology Management, Narsee Moonjee Institute of Management Studies, Shirpur, India
3 Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
4 Division of Reproductive Biology, Maternal and Child Health, Indian Council of Medical Research, New Delhi, India
5 Centre for Advanced Studies, University of Rajasthan, Jaipur, India
*Correspondence: pkm_8bh@yahoo.co.uk (Pradyumna Kumar Mishra)
Front. Biosci. (Landmark Ed) 2019, 24(6), 1097–1157; https://doi.org/10.2741/4771
Published: 1 March 2019
Abstract

Diet and environment are two critical regulators that influence an individual’s epigenetic profile. Besides the anterograde signaling, mitochondria act as a key regulator of epigenetic alterations in cancer either by controlling the concentration of the cofactors, activity of vital enzymes or by affecting the transcription of NF-kappaB and associated signaling molecules. As epigenetic modifications are the major drivers of aberrant gene expression, designing novel nutri-epigenomic strategies to modulate reversible epigenetic modifications will be important for effective cancer protection. In this regard, nutraceuticals such as flavonoids holds significant promise to modulate the epigenome through a network of interconnected anti-redox mechanisms. However, low solubility, rapid metabolism and poor absorption of flavonoids in gastrointestinal tract hinder their use in clinical settings. Therefore, it is imperative to develop nano-engineered systems which could considerably improve the targeted delivery of these bioactive compounds with better efficacy and pharmacokinetic properties. Concerted efforts in nano-engineering of flavonoids using polymer, lipid and complexation based approaches could provide successful bench-to-bedside translation of flavonoids as broad spectrum anti-cancer agents.

Keywords
Nutriepigenomics
Epigenetic modifiers
Anti-cancer agents
Nanomedicine
Translational research
Review
2. INTRODUCTION

Non-communicable diseases including cancer are the major cause of global deaths. According to a recent estimate, 18.1 million people have been newly diagnosed for cancer and 9.6 million deaths being recorded globally in 2018. This high rate of mortality can be prevented if detected early. As the treatment strategy varies with the pattern, type and form of cancer, management of this disease requires further development of better therapeutics and preventive strategies. Broadly, cancer shows the following seven characteristic features: uncontrolled proliferation, self-sufficient growth factors, contact-independent growth, absence of apoptosis, abnormal angiogenesis, increased inflammatory response and prevalence of metastasis and invasion. The abnormal cellular physiology of cancerous cells is due to altered molecular patterns at genomic, epigenomic, transcriptomic, proteomic and metabolomic levels. Unlike genetic control which was described as the trigger for evolutionary mechanisms, epigenomics deals with the study of a set of processes (above the DNA) involving DNA methylation, histone modifications and remodeling, and miRNA expressions. These epigenetic modifications play a vital role in chromatin remodeling for the regulation of gene expression. Altered epigenetic patterns have been strongly associated with cancer related events (1, 2). Recent studies have confirmed that mitochondria regulate epigenome either by controlling the concentration of the cofactors or by the redox-mediated alteration of the activities of epigenetic modifiers (3-5). However, finding potent epigenetic targets for therapeutic intervention intended for cancer prevention is an emerging aspect where different epigenetic inhibitors/modulators are being discovered, validated and verified (6-8). Over the centuries, many herbal and medicinal plants and their active constituents have been analyzed as possible remedies for several chronic and metabolic diseases. Secondary metabolites from plants, such as polyphenols including alkaloids, flavonoids, and stilbenes, which are present in small quantity have been tested for preventive management of several non-communicable diseases such as diabetes, cardiovascular diseases, asthma, neurological disorders and cancer (9-11). Interestingly, these molecules not only possess anti-oxidant, anti-inflammatory, cyto-protective and geno-protective properties but also modulate epigenomic re-programming through a mitochondrial mediated pathway (12-14).

Mitochondria not only control cellular oxidation via oxidative phosphorylation but also regulate programmed cell death, calcium concentration, metabolite concentrations, and diverse signaling pathways. These multifaceted roles make it dynamic controllers of cellular health and disease. Moreover, mitochondria may directly influence genome methylation, covalent histone modification, and expression of miRNA arrays to eventually alter the expression of target nuclear genes (15-17). However, the therapeutic outcome of flavonoids is limited due to reduced bioavailability as a result of limited solubility, poor permeability and pre-systematic metabolic effects. The other mechanisms involved in restricting the application of flavonoids include metabolism by gut micro flora, absorption across the intestinal wall, active efflux, and susceptibility to modification by environmental factors such as temperature, pH and light. In addition, recent studies have suggested that mitochondria are the central target of mechanistic basis of flavonoid’s mode of action, thus modulating the epigenetic machinery and complex cancer signaling, thereby suppressing the cancer progression programming. Therefore making a nanocarrier based system targeting mitochondria would offer a suitable strategy (Figure 1). Our previous studies have also explored numerous anti-cancer biological activities of the flavonoid-rich contents of an important medicinal herb Selaginella bryopteris (Sanjeevani) (11, 14). Our recent preclinical study demonstrated how nano-engineered flavonoid rich fraction isolated from S. bryopteris (NP.SB) act through mitochondria to alter the epigenetic landscape from the tumorigenesis route to normal cellular physiology (14). This contemporary approach offers a promising strategy to treat cancer with minimal side effects.

Figure 1.

An outline sketch of the proposed nano-engineered flavonoid based approach for effective mitochondrial targeting, maintenance of epigenomic machinery and induction of protective anti-cancer effects.

3. EPIGENETIC MODIFICATION: REVERSIBLE CODES

Epigenetic modifications play a vital role in gene regulation beyond the genetic material and exhibit stable meiotic and mitotic inheritance. Such epigenetic processes include covalent methylation of 5-cytosine of DNA, covalent modifications of N-terminal amino acid moieties of nucleosomal histone cores, and regulation through non-coding miRNA expressions. These modifications are important, intricate underlying mechanisms for maintenance of chromatin remodeling status. Epigenetic modifications occur commonly in all cell types which instruct genes either to turn off or on. Thus, cells with same genetic material respond differentially depending upon the internal and external environmental cues. Basically, epigenetic modifications are catalyzed by epigenetic modifiers known as writers (that add a specific modifying moiety); erasers (that remove a specific modifying moiety); and readers (that recognize and bind to specific modified moieties). Several distress points observed in the above mechanisms are implicated in various forms and types of cancers (18-22).

3.1. DNA methylation: Covalent cytosine methylation patterns

DNA methylation involves attaching a methyl group to the 5 carbon of cytosine nucleotide by DNA methyltransferases (DNMTs). On the other hand, mechanism of removal of a methyl group from cytosine either involves replication independent via TET demethylases or replication dependent through DNMT mediated demethylation (23). Methyl-CpG binding protein is the reader protein that specifically recognize and binds 5-methyl-cytosine nucleotides. Studies have confirmed that cancerous cells show aberrant promoter hypermethylation of tumor suppressor genes. In contrast, promoter hypomethylation of oncogenes increases the access of polymerase and transcription factors that mediate efficient transcription thereby causing increased expression of oncogenic proteins (24, 25).

Unlike nuclear genome, mitochondrial DNA is complexed with non-histone proteins and does not remain in a naked form. mtDNA methylation has been a subject of controversy so far, irrespective of existence of CpG and non-CpG sites (26-28). However, several studies have demonstrated that dysfunctional mtDNA methylation not only implicates the process of aging but also play an important role in onset and development of several age-associated degenerative diseases such as cancer (16, 29). It has been observed that several key enzymes such as DNMT1, DNMT3B and TET can translocate into the mitochondria and initiate the process of de novo mtDNA methylation (29).

3.2. Histone modification: Covalent tags on amino tails

Histone modifications are covalent modifications of N-terminal amino acid moieties of nucleosomal histone cores that include acetylation, methylation, phosphorylation, ubiquitination, and ribosylation. Several experimental investigations have documented the significant role of altered histone code and altered catalytic potentials of histone-modifying enzymes (21). Histone codes greatly influence gene expressions and significantly manipulate cellular processes by the inactivation or activation of tumor suppressor genes or oncogenes, respectively. For example, histone methylation of the tumor suppressor gene RASSF1A and histone de-acetylation of the transcription factor gene GATA4 silence the genes or more specifically reduces the binding of the transcription machinery, which increases the cancerous phenotypes (30) (Liu et al., 2016).

3.2.1. Histone acetylation

Histone acetyl transferase (HAT) is a writer for histone acetylation that uses acetyl-CoA as a cofactor to attach the acetyl group at the lysine or arginine amino acid residue of N-terminus of nucleosomal histone cores. There are four classes of histone deacetylases (Class I-IV) that function as erasers. Known mechanism for acetylation mediated regulation involves presence of electric charge mediated conformational change in nucleosome. For instance, highly acetylated nucleosomal histone cores create nucleosomes with loose configuration due to electric repulsion thus making euchromatin transcriptionally active. On the other hand, hypoacetylation marks the tight arrangement of several nucleosomes together, thus, producing heterochromatin or silent regions. Recent reports have shown acetyl transferase and deacetylases as the crucial players in the tumorigenesis of many cancers (31) (Su et al., 2016a). Aberrant expression and catalytic activity of different HATs cause acetylation of histone core tails, which generate distinct histone codes thus modulating normal encrypted language to the abnormal outcomes. For example, the acetylation of H4K16 is catalyzed by HATs such as males absent on the first (MOF) which belongs to MYST (Moz-Ybf2/Sas3-Sas2-Tip60) family. These modifications are associated with ovarian, breast, colorectal, gastric, lung, and renal cell cancers (31-33). Basically, silent MOF drive transcriptional gene inactivation and faulty DNA damage repair leading to genomic instability and lethality, thereby increasing the incidence of carcinogenesis.

3.2.2. Histone methylation

Similarly, histone methylation has lysine and arginine methyltransferase as writers, lysine and arginine demethylase as erasers and chromodomain as readers. In most diseases, including cancer, histone methylation of the bivalent mark as tri-methylated lysine 27 of histone 3/ di-methylated lysine 4 of histone 4 (H3K27me3/H3K4me2) is found repressed and correlates with a shorter survival time (34). Lysine methyltransferase enhancer of zeste homolog 2 (EZH2) is recognized as an important contributor in the progression of proliferative tumors (35). The H3K27 tri-methylation is catalyzed by EZH2 by involving the polycomb repressive complex 2 (PRC2). Enhanced levels of H3K9me3 were also observed in the stress-induced premature senescence of ovarian epithelial cells, indicating increased heterochromatinization in response to increased histone methylation (36). Further, upregulated expression of lysine-specific demethylase 1 (LSD1) is linked to various epithelial cancers (37). It is a histone demethylase that plays a major role in modifying epigenetic pattern of epithelial-mesenchymal transition (EMT) genes (37). Dysregulated expression of EMT genes promotes metastasis and invasion. For instance, demethylation of histone H3 lysine 4 (H3K4) at the E-cadherin promoter region downregulates its expression thereby enhances metastasis.

3.2.3. Histone phosphorylation

In addition, kinases and phosphatases control phosphorylation of histone, which is recognized by 14-3-3, BRCT, and BRCA1 proteins (38). Phosphorylation of histones provides essential chromatin configuration for the binding of transcription factors and DNA repair proteins. At molecular level, DNA damage is correlated with the concentration of phosphorylated histone 2AX (H2AX), which is a sensor protein for DNA damage and its phosphorylation activates the downstream protein ATM (39). H2AX and H3.3 phosphorylation have been linked with the high-grade tumors (40). Therefore, phosphorylated H2AX can be a potential marker to detect cancer at an early stage and to recognize relapse cases (41).

3.2.4. Histone ubiquitination

Along with the regulation of protein homeostasis by degradation, ubiquitination of histone plays a major role in transcription activation/inactivation, DNA repair, and condensation. Histone ubiquitination at the epigenomic level involves modification of lysine residue at the tails of nucleosomal histone core (13). Ubiquitin-protein ligase such as RING finger proteins RNF20 and RNF40 are the writers, and deubiquitinases (DUBs) such as USP7, USP22, USP44, and HAUSP are the erasers. Inverted ubiquitin interaction motif (IUIM) is a protein that binds at the ubiquitinated lysine (42, 43). Previous studies have shown that signaling pathways such as replication-dependent histone mRNA 3’-end processing, DNA repair response, transcriptional elongation and stem cell differentiation are controlled by CDK9 directed H2Bub1 modification (13). H2Bub1 has been implicated in tumor progression and prognosis of the aggressive cancers; thus, its loss can be an indicator of onset of the disease (44).

3.3. microRNA (miRNA) expression profiling

miRNAs are small non-coding RNAs transcripts of 21 to 23 nucleotides, which play a major role in gene expression regulation. Unlike regulation by DNA methylation and histone modifications at the genetic and epigenetic levels, microRNA functions at the post-transcriptional level. Molecular mechanisms of miRNAs functions involve binding to its complimentary mRNA where complete binding induces mRNA degradation and partial binding causes mRNA silencing (45). miRNAs are the important regulators of various fundamental processes such as growth, differentiation, stress response, and cellular homeostasis. miRNAs are known to interact with oncogenes and tumor suppressor genes and are thus involved in the control of tumor development and progression, invasion, metastasis, and in epithelial–mesenchymal transition of several cancers (46). Analysis of miRNA profiling suggests that some of miRNA are upregulated, and some are down-regulated in different sets of cancer (47).

4. FLAVONOIDS AS PROMISING NUTRACEUTICALS

Nutraceuticals are bioactive molecules with nutritional value such as dietary food components including phytochemicals. Phytochemicals are a group of secondary metabolites such as polyphenols, flavonoids, terpenoids, xanthones, organo-sulfur compounds, phytosterols, alkaloids, and carotenoids, which are known to have curative properties for diverse human ailments. Presence of hydroxyl groups and aromatic ring in polyphenols categorizes them into following subclasses: flavonoids, phenolic acids, stilbenes, and lignans. Flavonoids are widely distributed cluster of seven subgroups categorized on the basis of their chemical structures. There are several representative bioactive compounds in each subgroup of flavonoids (see Figure 2). These are synthesized via phenyl propanoid pathway, which converts phenylalanine into 4-coumaroyl-CoA to produce flavonoids. All subgroups of flavonoids consist of the same structural backbone of two phenolic rings A and B connected by the third oxygen-containing heterocyclic C ring (see the ring structures in Figure 3 and 4). These subgroups are named as flavones, flavonols, flavanones or dihydroflavones, flavanonols or dihydroflavonols, isoflavones or phytoestrogens, flavanols or catechins or anthocyanins, and proanthocyanins (Figure 3 and 4).

Figure 2.

The flow diagram showing detailed classification of polyphenols.

Figure 3.

A combined figure showing structures of different flavonoids (a) Flavones that consists of 4H-chromen-4-one with a phenyl substituent at position 2 and are found in fruits and vegetables, including onions, apples, broccoli, and berries, thyme, parsley, celery and capsicum pepper. (b) Flavanones or dihydroflavones which consists of flavan bearing an oxo substituent at position 4. It is derived from a hydride of a flavan and is generally found in grapes, orange and lemon juice. (c) Flavonols, a monohydroxyflavone that comprises a class of compounds with 3-hydroxy derivative of flavone. It is a conjugate acid of a flavonol (1-) and is found in green and black tea, yellow onions, apples, broccoli, black grapes, tomato, red wine, carrot, cherry, tomato, curly kale, leek, lettuce, nuts, walnuts, ginger, and blueberry.

Figure 4.

Figure showing structural representation of (a) flavanonols or dihydroflavonols are a class of flavonoids with 3-hydroxy-2,3-dihydro-2-phenylchromen-4-one backbone and are generally isolated from sources such as lemon, sour orange, cocoa, cocoa beverages, and chocolates. (b) Isoflavones or phytoestrogens are a class of polyphenolic compounds which consists of 4H-chromen-4-one ring in which the hydrogen at position 3 is replaced by a phenyl group and are generally found in sources such as soya bean, legumes, red clover chickpeas, peanuts, soy cheese, soy flour and tofu. (c) Flavanols or flavan-3-ol is a hydroxyflavonoid which are found in various sources such as green and black tea, apple, chocolate, beans, apricot, cherry, grapes, peach, red wine, cider, and blackberry. (d) Anthocyanidins, which are the salt derivatives of the 2-phenylchromenylium cation, also known as flavylium cation and most commonly found in blue berries, black grapes, and red wine, blackcurrant, cherry, rhubarb, plum, red cabbage, and cocoa.

These molecules have the potential to alter the health status as they have molecular targets within the cell, however, timing of food consumption, absorption, metabolism, and excretion are vital along with the presence of other factors like the use of tobacco and alcohol. Being a part of our ancient system of medicine, phytochemicals-based therapy is emerging as one of the most efficient therapies not only against cardiovascular and metabolic diseases, but also in cancer treatment (Table 1). Previous preclinical studies have demonstrated the therapeutic aspects of these bioactive molecules. Anti-oxidative, anti-inflammatory, anti-proliferative, cyto-protective and geno-protective effect are the potential therapeutic outcome of these flavonoids (11, 14, 47, 48). These therapeutic properties of flavonoids are due to their interaction with the intermediate of cell signaling proteins. Major regulatory mechanisms of phytochemicals involve modulation of redox reactions, enzyme activities, mitochondrial-retrograde responses, and cellular signaling pathways that significantly alter the metabolome and gene expression or vice-versa (Figure 5) (49, 50). Flavonoids possessing epigenetic restoration property have also been reported (51, 52).

Figure 5.

The diagrammatic representation of different mechanistic aspects of flavonoids

Table 1.The table shows several subclasses of flavonoids tested for the anti-carcinogenic potential using different preclinical cancer models
Sub-classes of flavonoids Examples Preclinical cancer model References
Flavones Apigenin B-cell lymphoma (148)
Flavones Apigenin Bladder (149)
Flavones Apigenin Breast (150)
Flavones Apigenin Cervical (151)
Flavones Apigenin Chronic lymphocytic leukemia (152)
Flavones Apigenin Colon (153)
Flavones Apigenin Colorectal (154)
Flavones Apigenin Esophageal (155)
Flavones Apigenin Gastric (156)
Flavones Apigenin Glioblastoma (157)
Flavones Apigenin Glioma (158)
Flavones Apigenin Hepatocellular (159)
Flavones Apigenin Leukemia (160)
Flavones Apigenin Lung (161)
Flavones Apigenin Melanoma (76)
Flavones Apigenin Neuroblastoma (162)
Flavones Apigenin Oral (163)
Flavones Apigenin Osteosarcoma (164, 165)
Flavones Apigenin Ovarian (166)
Flavones Apigenin Pancreatic (167)
Flavones Apigenin Prostate (168)
Flavones Apigenin Skin (169)
Flavones Apigenin Thyroid (170)
Flavones Baicalein Breast (171)
Flavones Baicalein Cervical (172)
Flavones Baicalein Colon (173)
Flavones Baicalein Colorectal (174)
Flavones Baicalein Gastric (175)
Flavones Baicalein Glioma (176)
Flavones Baicalein Hepatocellular (177, 178)
Flavones Baicalein Leukemia (179)
Flavones Baicalein Lung (180)
Flavones Baicalein Melanoma (181)
Flavones Baicalein Osteosarcoma (182)
Flavones Baicalein Ovarian (183)
Flavones Baicalein Pancreatic (184)
Flavones Baicalein Prostate (185)
Flavones Baicalein Skin (186)
Flavones Baicalein Sarcoma (187)
Flavones Baicalin B-cell lymphoma (188)
Flavones Baicalin Breast (189)
Flavones Baicalin Cervical (190)
Flavones Baicalin Colon (191)
Flavones Baicalin Colorectal (192)
Flavones Baicalin Glioma (193)
Flavones Baicalin Hepatocellular carcinoma (194)
Flavones Baicalin Leukemia (195)
Flavones Baicalin Lung (196)
Flavones Baicalin Osteosarcoma (197)
Flavones Baicalin Ovarian (198)
Flavones Baicalin Prostate (199)
Flavones Chrysin Breast (200)
Flavones Baicalin Cervical (201)
Flavones Baicalin Chronic lymphocytic leukemia (202)
Flavones Baicalin Colon (203)
Flavones Baicalin Colorectal (204)
Flavones Baicalin Gastric (205)
Flavones Baicalin Glioma (206)
Flavones Baicalin Hepatocellular (207)
Flavones Baicalin Lung (208)
Flavones Baicalin Leukemia (78)
Flavones Baicalin Melanoma (209)
Flavones Baicalin Osteosarcoma (210)
Flavones Baicalin Prostate (211)
Flavones Baicalin Skin (31)
Flavones Baicalin Thyroid (212)
Flavones Baicalin Ovarian (213)
Flavones Hispidulin Colon (214)
Flavones Hispidulin Gastric (215)
Flavones Hispidulin Renal cell (216)
Flavones Luteolin Breast (217)
Flavones Luteolin Cervical (218)
Flavones Luteolin Colon (219)
Flavones Luteolin Colorectal (220)
Flavones Luteolin Esophageal (221)
Flavones Luteolin Gastric (222)
Flavones Luteolin Glioblastoma (223)
Flavones Luteolin Hepatocellular (224)
Flavones Luteolin Leukemia (202)
Flavones Luteolin Lung (225)
Flavones Luteolin Neuroblastoma (226)
Flavones Luteolin Pancreatic (227)
Flavones Luteolin Prostate (228)
Flavones Luteolin Skin (229)
Flavones Luteolin Thyroid (230)
Flavones Luteolin Sarcoma (231)
Flavones Luteolin Urinary bladder (232)
Flavones Nobiletin Acute myeloid leukemia (233)
Flavones Nobiletin Breast (234)
Flavones Nobiletin Colon (235)
Flavones Nobiletin Colorectal (236)
Flavones Nobiletin Gastric (237)
Flavones Nobiletin Glioma (238)
Flavones Nobiletin Hepatocellular (239)
Flavones Nobiletin Lung (240)
Flavones Nobiletin Melanoma (241)
Flavones Nobiletin Neuroblastoma (242)
Flavones Nobiletin Ovarian (243, 244)
Flavones Nobiletin Prostate (245)
Flavones Pectolinarigenin Nasopharyngeal (246)
Flavones Pectolinarigenin Osteo-sarcoma (247)
Flavones Tangeretin Breast (248)
Flavones Tangeretin Colon (249)
Flavones Tangeretin Colorectal (250)
Flavones Tangeretin Gastric (251)
Flavones Tangeretin Glioma (252)
Flavones Tangeretin Lung (253)
Flavones Tangeretin Melanoma (254)
Flavones Tangeretin Ovarian (255)
Flavones Wogonin Breast (256)
Flavones Wogonin Melanoma (257)
Flavones Wogonin Glioma (258)
Flavones Wogonin Prostate (259)
Flavones Wogonin Leukemia (260)
Flavones Wogonin Lung adenocarcinoma cell (261)
Flavones Wogonin Gastric (262)
Flavones Wogonin Ovarian (263)
Flavones Wogonin Colorectal (264)
Flavonols Fisetin Acute myeloid leukemia (265)
Flavonols Fisetin Breast (266)
Flavonols Fisetin Cervical (267)
Flavonols Fisetin Colon (268)
Flavonols Fisetin Colorectal (269)
Flavonols Fisetin Gastric (270)
Flavonols Fisetin Glioma (271)
Flavonols Fisetin Hepatocellular (272)
Flavonols Fisetin Lung (273)
Flavonols Fisetin Melanoma (274)
Flavonols Fisetin Ovarian (275)
Flavonols Fisetin Prostate (276)
Flavonols Isorhamnetin Breast (277)
Flavonols Isorhamnetin Colon (278)
Flavonols Isorhamnetin Colorectal (279)
Flavonols Isorhamnetin Lung (280)
Flavonols Isorhamnetin Gastric (281)
Flavonols Isorhamnetin Skin (282)
Flavonols Kaempferol Breast (283)
Flavonols Kaempferol Cervical (284)
Flavonols Kaempferol Colon (285)
Flavonols Kaempferol Colorectal (286)
Flavonols Kaempferol Esophageal (287)
Flavonols Kaempferol Gastric (288)
Flavonols Kaempferol Glioma (289)
Flavonols Kaempferol Hepatocellular (290)
Flavonols Kaempferol Leukemia (291)
Flavonols Kaempferol Lung (292)
Flavonols Kaempferol Ovarian (293)
Flavonols Kaempferol Pancreatic (294)
Flavonols Kaempferol Prostate (295)
Flavonols Kaempferol Skin (296)
Flavonols Kaempferol Urinary bladder (297)
Flavonols Myricetin Breast (298)
Flavonols Myricetin Cervical (299)
Flavonols Myricetin Colon (300)
Flavonols Myricetin Colorectal (301)
Flavonols Myricetin Esophageal (302)
Flavonols Myricetin Gastric (303)
Flavonols Myricetin Glioblastoma (304)
Flavonols Myricetin Hepatocellular (305)
Flavonols Myricetin Lung (306)
Flavonols Myricetin Ovarian (307)
Flavonols Quercetin Breast (308)
Flavonols Quercetin Cervical (309)
Flavonols Quercetin Chronic lymphocytic leukaemia (310)
Flavonols Quercetin Colon (311)
Flavonols Quercetin Colorectal (312)
Flavonols Quercetin Esophageal (313)
Flavonols Quercetin Gastric (314)
Flavonols Quercetin Hepatocellular (315)
Flavonols Quercetin Osteosarcoma (316)
Flavonols Quercetin Ovarian (317)
Flavonols Quercetin Pancreatic (318)
Flavonols Quercetin Prostate (319)
Flavonols Quercetin Thyroid (320)
Flavonols Quercetin Urinary bladder (321)
Flavonols Rutin Colon (322)
Flavonols Rutin Lung (323)
Flavonols Rutin Neuroblastoma (324)
Flavonols Rutin Promyelocytic leukemia (325)
Flavonols Rutin Prostate (326)
Flavonols Rutin Skin (327)
Flavonols Alpinetin Lung (328)
Flavonols Alpinetin Hepatoma (329)
Flavonols Alpinetin Pancreatic (330)
Flavanones Hesperidin Acute myeloid leukemia (331)
Flavanones Hesperidin Breast (332)
Flavanones Hesperidin Colon (333)
Flavanones Hesperidin Hepatocellular (334)
Flavanones Hesperidin Lung (335)
Flavanones Hesperidin Urinary bladder (336)
Flavanones Hesperidin Ovarian (337)
Flavanones Hesperitin 5,7,3’-trihydroxyl-4’-methoxylflavanone Breast (338)
Flavanones Hesperitin Thyroid (339)
Flavanones 5,7,3’-trihydroxyl-4’-methoxylflavanone Prostate (340)
Flavanones Hesperitin Carcinoid (341)
Flavanones Naringenin Breast (342)
Flavanones Naringenin Colon (343)
Flavanones Naringenin Colorectal (344, 345)
Flavanones Naringenin Gastric (346)
Flavanones Naringenin Hepatocellular (347)
Flavanones Naringenin Lung (348)
Flavanones Naringenin Leukemia (349)
Flavanones Naringenin Pancreatic (350)
Flavanones Naringenin Prostate (351)
Flavanones Naringenin B-cell lymphoma (352)
Flavanones Furanoflavanone Gastric (353)
Flavanonols Taxifolin Prostate (354)
Flavanonols Taxifolin Skin (355)
Flavanonols Silibinin Acute myeloid leukemia (331)
Flavanonols Silibinin Breast (79)
Flavanonols Silibinin Cervical (356)
Flavanonols Silibinin Colon (357)
Flavanonols Silibinin Colorectal (358)
Flavanonols Silibinin Gastric (359)
Flavanonols Silibinin Glioblastoma (223)
Flavanonols Silibinin Lung (360)
Flavanonols Silibinin Esophageal (361)
Flavanonols Silibinin Ovarian (362)
Flavanonols Silibinin Pancreatic (363)
Flavanonols Silibinin Prostate (364)
Flavanonols Silibinin Skin (365)
Flavanonols Silibinin Bladder (366)
Flavanonols Silibinin Thyroid (367)
Flavanonols Silymarin Cervical (368)
Flavanonols Silymarin Colon (369)
Flavanonols Silymarin Hepatocellular (370)
Flavanonols Silymarin Lung (371)
Flavanonols Silymarin Neuroblastoma (372)
Flavanonols Silymarin Ovarian (373)
Flavanonols Silymarin Promyelocytic leukemia (374)
Flavanonols Silymarin Prostate (375)
Flavanonols Silymarin Bladder (376)
Flavanonols Pinobanksin B-cell lymphoma (377)
Isoflavones Genistein Acute myeloid leukemia (378)
Isoflavones Genistein Breast (379)
Isoflavones Genistein Cervical (380)
Isoflavones Genistein Colon (381)
Isoflavones Genistein Colorectal (382)
Isoflavones Genistein Esophageal (383)
Isoflavones Genistein Gastric (384)
Isoflavones Genistein Glioblastoma (385)
Isoflavones Genistein Hepatocellular (386)
Isoflavones Genistein Leukemia (387)
Isoflavones Genistein Lung (388)
Isoflavones Genistein Melanoma (389)
Isoflavones Genistein Oral (390)
Isoflavones Genistein Osteosarcoma (391)
Isoflavones Genistein Ovarian (392)
Isoflavones Genistein Pancreatic (393)
Isoflavones Genistein Prostate (394)
Isoflavones Genistein Uterine (395)
Isoflavones Genistein Bladder (396)
Isoflavones Biochanin A Breast (397)
Isoflavones Biochanin A Colon (398)
Isoflavones Biochanin A Glioma (399)
Isoflavones Biochanin A Hepatocellular (400)
Isoflavones Biochanin A Leukemia (401)
Isoflavones Biochanin A Lung (402)
Isoflavones Biochanin A Pancreatic (77)
Isoflavones Biochanin A Prostate (403)
Isoflavones Glycitein Breast (404)
Isoflavones Formononetin Colon (405)
Isoflavones Formononetin Cervical (406)
Isoflavones Formononetin Osteosarcoma (407)
Isoflavones Formononetin Prostate (408)
Isoflavones Formononetin NSCLC (409)
Isoflavones Formononetin Breast (61)
Isoflavones Formononetin Glioma (410)
Isoflavones Daidzein Breast (411)
Isoflavones Daidzein Colon (381)
Isoflavones Daidzein Hepatic (412)
Isoflavones Daidzein Melanoma (413)
Isoflavones Daidzein Neuroblastoma (414)
Isoflavones Daidzein Pancreatic (415)
Isoflavones Daidzein Prostate (416)
Isoflavones Daidzein Skin (417)
Flavanols Catechin B-cell lymphoma (418)
Flavanols Catechin Breast (419)
Flavanols Catechin Bladder (420)
Flavanols Catechin Cervical (421)
Flavanols Catechin Glioma (422)
Flavanols Catechin Hepatocellular (423)
Flavanols Catechin Lung (424)
Flavanols Catechin Melanoma (425)
Flavanols Catechin Pancreatic (426)
Flavanols Catechin Promyelocytic leukemia (427)
Flavanols Catechin Prostate (428)
Flavanols Epigallocatechin-3-gallate Acute myeloid leukemia (429)
Flavanols Epigallocatechin-3-gallate B-cell lymphoma (418)
Flavanols Epigallocatechin-3-gallate Urinary Bladder (430)
Flavanols Epigallocatechin-3-gallate Breast (431)
Flavanols Epigallocatechin-3-gallate Cervical (432)
Flavanols Epigallocatechin-3-gallate Chronic lymphocytic leukaemia (433)
Flavanols Epigallocatechin-3-gallate Colon (434)
Flavanols Epigallocatechin-3-gallate Colorectal (435)
Flavanols Epigallocatechin-3-gallate Esophageal (436)
Flavanols Epigallocatechin-3-gallate Gastric (437)
Flavanols Epigallocatechin-3-gallate Glioblastoma (438)
Flavanols Epigallocatechin-3-gallate Glioma (439)
Flavanols Epigallocatechin-3-gallate Hepatocellular (440)
Flavanols Epigallocatechin-3-gallate Lung (441)
Flavanols Epigallocatechin-3-gallate Melanoma (442)
Flavanols Epigallocatechin-3-gallate Neuroblastoma (443)
Flavanols Epigallocatechin-3-gallate Oral (444)
Flavanols Epigallocatechin-3-gallate Osteosarcoma (445)
Flavanols Epigallocatechin-3-gallate Ovarian (446)
Flavanols Epigallocatechin-3-gallate Pancreatic (447)
Flavanols Epigallocatechin-3-gallate Prostate (448)
Flavanols Epigallocatechin-3-gallate Skin (449)
Flavanols Epigallocatechin-3-gallate Thyroid (450)
Flavanols Epigallocatechin-3-gallate Sarcoma (451)
Anthocyanidins Cyanidin Breast (452)
Anthocyanidins Cyanidin Colon (453)
Anthocyanidins Cyanidin Lung (454)
Anthocyanidins Cyanidin Ovarian (455)
Anthocyanidins Cyanidin Prostate (456)
Anthocyanidins Delphinidine Breast (457)
Anthocyanidins Delphinidine Colon (453)
Anthocyanidins Delphinidine Glioblastoma (458)
Anthocyanidins Delphinidine Hepatocellular (459)
Anthocyanidins Delphinidine Lung (460)
Anthocyanidins Delphinidine Ovarian (461)
Anthocyanidins Delphinidine Prostate (462)
Anthocyanidins Pelargonidin Hepatic (463)
5. NUTRI-EPIGENETICS: FLAVONOIDS AS EPIGENETIC MODIFIERS

Epigenetic modifications are greatly affected with the change in the environmental conditions and act as interface between the gene and environment. Defined concentration of polyphenols in the diet can modulate epigenome-mediated genomic expressions essentially by controlling the metabolic fate of a cell hence influencing the mechanisms underlying human health and disease (9, 51, 53-55). Accumulative literature on epigenetically active polyphenols suggests that these molecules have anti-cancer effects as these can potentially manipulate the activities of epigenetic modifiers such as writers (DNMT, HAT, and HMT) and erasers (HDAC, KDM) (52). A diet containing epigenome-modifying and chemo preventive nutraceuticals influence the level of bioactive metabolites that in turn modulate cellular pools of methyl donor groups, acetyl-CoA, NAD+, and ATP, resulting in altered DNA methylation pattern and histone and non-histone protein acetylation/methylation/phosphorylation (3-5). For instance, alteration in DNA and histone methylation level is associated with the imbalance in the SAM/SAH ratio. A significant change in the SAM/SAH ratio level has been reported following ingestion of flavanol-rich diets (56-59). In vitro and in vivo tumor/cancer models treated with specific polyphenolic bioactive components have revealed efficient antitumor potential by stimulating the signaling pathway for programed cell death mediating epigenetic machinery (60). Such observations are enabling the modifiers of epigenetic mechanisms to emerge as anti-cancer therapeutics. Therefore, currently a powerful tenet is to explore how dietary molecules may influence prevention and treatment of various chronic diseases based on nutri-epigenomic intervention. Recently, the anti-cancer therapeutic potential of some bioactive compounds with the epigenetic reversal ability have been tested in clinical trials (see Table 2) (61-64).

Table 2.The table shows different sub-classes of flavonoids along with dose, duration and route of administration clinically tested for various cancers.
Clinical trial phase and type Flavonoids used Cancer type Participant number Dose amount, duration & mode Finding References
Pre-surgical trial Silybin-phosphatidylcholine Breast cancer 12 2.8 g daily for 4 weeks prior to surgery; oral High blood concentration of silybin-phosphatidylcholine ensures selective accumulation (464)
Phase Ib; Single site; Clinical study AXP107-11, a multi-component crystalline form of genistein Unresectable pancreatic cancer 16 400 mg-1600 mg daily in combination with 1000 mg/m(2) gemcitabine per week orally for 6 months Favorable PK-profile was reported (465)
Phase II; Randomized; Double-blind; Placebo-controlled Genistein Prostate cancer 47 30 mg genistein daily for 3-6 weeks Modulate genes related to cell cycle and androgen expression (466)
Phase II; Placebo-controlled; Block-randomized; Double-blind Genistein Localized prostate cancer 44 30 mg daily for 3 to 6 week prior to prostatectomy Well tolerated and reduced the level of serum PSA (467)
Phase II; Randomized Soy isoflavone Prostate cancer 32 2 slices of soy bread containing 34 mg isoflavones/slice or soy bread containing almond powder prescribed daily as a source of β-glucosidase for oral consumption for 56 days Suppressed pro-inflammatory cytokines expression and reduced immunosuppressive cells (64)
Phase II; Randomized crossover trial Isoflavone Asymptomatic prostate cancer 32 Crossover of two formulations as soy bread and soy almond bread to deliver approximately 60 mg aglycone equivalents of isoflavones per day for 20-weeks Metabolizing phenotypes of isoflavones to distinguish cancer patients resistance to preventive strategies were reported (62)
Randomized; Placebo-controlled study Isoflavones Breast Cancer 140 2 packets of 25.8g soy protein powder or 25.8g milk protein per day with water or juice until surgery High genistein level modulates gene expression prolife; and over-express cell proliferating genes (468)
Randomized; Double-blind; Placebo-controlled Isoflavone Prostate cancer 86 80 mg/day of total isoflavones & 51 mg/day aglucon units Serum hormone levels, total cholesterol, or PSA remain unchanged upon short-term intake of soy isoflavones (469)
Phase II; Randomized; Placebo-controlled Isoflavone Urothelial bladder cancer 59 300 or 600 mg/day as purified soy extract G-2535 for 14 to 21 days before surgery More effective responses at the lower dose in bladder cancer tissues were recorded (398)
Phase II; Randomized; Double-blind; Placebo-controlled Isoflavone Prostate cancer 158 60 mg/day isoflavones prescribe orally for 12 months Isoflavone reduces prostate cancer risk (470)
Phase I; Randomized; Dose-escalating Isoflavones Prostate cancer 45 40 -60-80 mg purified isoflavones from biopsy to prostatectomy Safe dose of purified isoflavones for future examinations were established (471)
Phase II Isoflavones Advanced pancreatic cancer 20 531 mg isoflavones twice daily from day 7 until the end of study; 1,000 mg/m² gemcitabine on days 1, 8, and 15; 150 mg erlotinib once daily on day 1 to day 28 Non-significant survival of patients (472)
Double-blind; Randomized; Placebo-controlled Isoflavones Breast cancer 205 Isoflavone tablet - 1 mg genistein, 26 mg biochanin A, 16 mg formononetin, and 0.5 mg daidzein Ineffective to increase mammographic breast density and has no effect on lymphocyte tyrosine kinase activity, oestradiol, gonadotrophins, or menopausal symptoms. (473)
Randomized; Placebo-controlled; Crossover Phytoestrogens Breast cancer 62 114 mg isoflavonoids as Phytoestrogen tablets for 3 months Non-significant to eliminatemenopausal symptoms in breast cancer patients (474)
Phase I Epigallocatechin-3-gallate (EGCG) Breast cancer 24 40 to 660 μmol l(-1) in 7 levels for 2 weeks; topical Well tolerated and but no maximum tolerated dose was detected. Effective treatment of radiation dermatitis was reported (475)
Phase II EGCG Stage III lung cancer. 37 440 μmol/L given orally three times a day during the radiation. Effective and safe method against acute radiation-induced esophagitis (476)
Phase II; Two-stage; Single-arm EGCG Advanced stage ovarian cancer 46 500 mL double-brewed green tea daily until recurrence or during a follow-up of 18 months Doesn’t show promising maintenance intervention in advanced stage ovarian cancer after standard treatment. (477)
Randomized controlled trial The Minnesota green tea trial; green tea catechin Breast cancer 1075 800 mg daily for one year Study verified breast cancer patients for the differences in tea catechins metabolism based on COMT genotype to propose biomarkers of breast cancer risk (478)
Placebo-controlled; Randomized clinical trial Green tea catechins Prostate Cancer 97 Polyphenon E® 400 mg/day for 1 year Well tolerated but not enough to overcome prostate cancer (428)
Phase II; Randomized clinical trial; Open label Brewed green and black tea Prostate cancer 113 1300 mg green tea polyphenol including 800 mg of EGCG Green tea modulates systemic oxidation and inflammatory arm through NFkB in prostate tissue (479)
Phase IB; Randomized; Placebo-controlled; Dose escalation Green tea extract (Polyphenon E) Breast cancer 34 400 mg; 600 mg; and 800 mg twice daily given orally for 6 months Promising role of tea polyphenols in signaling pathways of growth factors such as HGF and VEGF, angiogenesis and lipid metabolism was observed (63)
Phase II; Randomized; Double-blind; Placebo controlled Polyphenon E Low grade cervical intraepithelial neoplasia 98 800 mg epigallocatechin gallate daily for 4 months Well tolerated and safe but unable to resist high-risk HPV infections (480)
Phase II Polyphenon E Rai stage 0-II chronic lymphocytic leukemia 42 2000 mg twice daily for 6 months Well tolerated and its daily consumption reduces ALC and/or lymphadenopathy (481)
Randomized; Double-blind; Placebo controlled Polyphenon E Prostate cancer 50 800 mg epigallocatechin gallate in Polyphenon E daily for 3–6 weeks before surgery Low bioavailability and/or bioaccumulation reported in prostate tissue (482)
Phase I Polyphenon E Asymptomatic Rai stage 0 to II chronic lymphocytic leukemia 33 400 to 2,000 mg twice a day for up to 6 months; Orally Well tolerated and observed to decreased lymphadenopathy observed in the majority of patients (483)
Phase II; Multicenter study; Single-arm; Open-label P276-00, a flavone-derived molecule Refractory mantle cell lymphoma 13 Intravenous infusion of 185 mg/m2/day from days 1-5 of a 21-day cycle A novel well-tolerated CDK isoforms inhibitor evaluated in patients with relapsed or refractory MCL (484)
6. MOLECULAR MECHANISM OF FLAVONOIDS: EPIGENETIC MODULATION THROUGH MITOCHONDRIA

Flavonoids or mitochondria-targeted drugs act through redox-dependent and redox-independent regulation of nuclear gene expression. Although many studies have emphasized the anti-oxidative properties of flavonoids, their effect on mitochondria in a redox-independent manner has not been extensively studied (65). However, individual cancer type has different cellular and molecular disorientation due to abnormal expression of susceptible genes. Therefore, a tumor cannot be treated effectively by targeting a distinct gene or protein or a single signal transduction pathway (11, 14, 47, 66). Since, several intra- and extracellular signaling pathways are governed by mitochondria and therefore, it is the primary site of action for flavonoids and also promotes apoptosis and inhibition of cell growth (Figure 6). Various mitochondria-targeted flavonoid compositions have been analyzed for their anti-cancer effects. Numerous mitochondria-targeted drugs have also been identified and validated for their anti-cancer effects (67-69) and based on their mode of action, these mitochondria-targeted anti-cancer drugs as classified as mitocans. However, (68) Gorlach et al., described the molecular mechanisms of each mitocan underlying anti-cancer activity.

Figure 6.

Scheme illustrating the vital role of mitochondria in regulation of major cellular processes involved in signaling, cell cycle, autophagy, apoptosis, angiogenesis, and epigenetic modifications.

Further, harboring healthy mitochondria greatly impact the redox status and related processes. Most observation favors the flavonoid induced upregulation of mitochondrial biogenesis through increase in the PGC-1α level. Recent reports have shown that (-)-epicatechin increase biogenesis by G-protein coupled estrogen receptor, fisetin enhance biogenesis by downregulation of glycogen synthase kinase 3β activity, and digitoflavone upregulate biogenesis by enhancing AMPK phosphorylation (70-72). However, results are conflicting for quercetin, as it can both increase and attenuate this process (73).

Recent experiments have uncovered the intricate role of mitochondria in the epigenetic regulation of several processes involved in human health and disease (13, 16, 74). There are two important routes through which mitochondria regulate the epigenetic machinery. Firstly, as the hub for the metabolic processes, the mitochondrion holds several tiny co-factors and substrates that are utilized in the epigenetic modification processes, such as the methyl group from the SAM (indirect source of methionine) for DNA and histone methylation, the acetyl group from acetyl-CoA for histone acetylation, and ATP for histone phosphorylation (15). Second, being the center for the oxidative mechanisms, mitochondria can oxidatively modify the catalytic properties of the mitochondrial and cellular enzymes and proteins thereby affecting the epigenetic process and redox signaling. For instance, mitochondria alter the activity of sirtuins, which are a class of NAD-dependent deacetylases. Sirtuin 6 has been implicated in ageing, many metabolic diseases and explored for therapeutic interventions (75).

7. PRECLINICAL STUDIES OF MITOCHONDRIA TARGETED FLAVONOIDS FOR ANTI-CANCER ACTION

Flavones such as apigenin, chrysin, and baicalein are potential anti-neoplastic and anti-carcinogenic agents. The anti-cancer activity of these flavonoids is mediated through activation of mitochondria-dependent apoptotic pathway, which has been validated through in vitro and in vivo studies (76-81). Induction of mitochondria-dependent apoptotic pathway in peripheral blood lymphocytes from B-chronic lymphocytic leukemia patients and leukemia cell lines have been observed following chrysin treatment (78). Chrysin disturbs the mitochondrial membrane potential that causes release of cytochrome C, up-regulate the pro-apoptotic Bax and caspase-3 and down-regulate the anti-apoptotic Bcl-2 protein that in turn result in apoptosis/cell death (78). Additionally, silibinin, in breast cancer, triggers mitochondria-mediated cell death. Basically, silibinin induces ROS-dependent mitochondrial dysfunction leading to ATP depletion by involving BNIP3 to initiate autophagy (79). Similarly, apigenin also causes mitochondrial dysfunction leading to mitochondria-mediated apoptosis in hepatocytes of HCC rats (80). Apigenin-loaded PLGA nanoparticles targeted at the nucleus damage DNA and subsequently induce mitochondria-mediated apoptosis in tumor cells (76). Moreover, baicalein follows a similar path in the gastric cancer cell line where it induces S phase arrest, down-regulate Bcl-2, up-regulate Bax and disrupt mitochondrial membrane potential in dose dependent manner (81). Additionally, mitochondriotropic nanoemulsified genistein-loaded carriers have shown pro-apoptotic anti-cancer effects due to mitochondrial damage through depolarization-mediated cytochrome C release (82). Moreover, a pro-oxidant action upon treatment of colorectal cancer with 5-Hydroxy-7-methoxyflavone was observed due to ROS triggered mitochondria-mediated apoptosis (77).

8. CHALLENGES IN FLAVONOIDS MEDIATED-ANTI-CANCER THERAPEUTIC STRATEGY

Several preclinical evidences have suggested that flavonoids possess anti-cancer properties; however, proper exploitation of the associated health benefits is limited. Primarily, limitations are due to poor bioavailability as a result of low solubility, insignificant permeability, first-pass metabolic effects, metabolism by gut micro flora, and absorption across the intestinal wall, active efflux mechanism, and easy modification by environmental factors such as temperature, pH, and light. Moreover, differences in isolation, purification, treatment strategy, interpretation, and analysis create an additional level of challenge in using flavonoids as anti-cancer agents.

8.1. Isolation and purification challenges

Many challenges are associated with the extraction and isolation of flavonoids, that may include the presence of low levels of active compounds (ranging from few micrograms to milligrams per kg of plant), complexity of the compounds, difficulty in isolation, identification and purification of individual constituents, complicated analytical procedures, time-consuming processes, high costs, and low product yield. The variation in flavonoid composition decreases the predictive yield and their labile nature make them susceptible to a high degree of degradation or chemical modification during the purification (83).

8.2. Pharmacokinetic (PK) challenges

Flavonoids possess characteristic impaired solubility, compromised oral absorption, and vulnerability to high hepatic metabolism subsequently leading to unsuitable PK profile (i.e., ADME and toxicity). Moreover, complexation or precipitation of flavonoids when ingested with other food components as well as their degradation by intestinal micro flora contributes to reduced stability and poor bioavailability (83, 84).

9. APPROACHES TO SURMOUNT PHARMACOKINETIC/PHARMACODYNAMIC AND OTHER BARRIERS FOR FLAVONOIDS DELIVERY

To allow clinical applicability of dietary flavonoids, exploration of various approaches has been reviewed.

9.1. Improving isolation, purification and yield

Over the past many years, several attempts have been made to improve the yield of flavonoids by the means of modifications in extraction, purification, and isolation approaches. On large scale, numerous techniques that have shown distinctive advantages such as high caliber extraction yield of antioxidants, and good percentage of marker compounds have been employed. These techniques may include pressure based procedures, microwave assisted methods, resin based adsorption systems and many others like pre-fermentation treatment, semi-preparative high-speed counter-current chromatography, etc (85-93).

9.2. Overcoming PK challenges

In order to surmount the limited bioavailability issues of flavonoids employment of numerous approaches have been reported. For example, flavonoids can be formulated as certain types of glycosides complex, which serve as substrates for certain intestinal epithelial transporters, leading to their increased absorption and enhanced bioavailability (94, 95). The administration of quercetin-4-O-glucoside resulted in a 5-fold increase in the plasma level of flavonoids as compared to quercetin-3-Orutinoside; thus, offering a potential substitute for augmentation of bioavailability (95). Moreover, addition of natural bio-enhancers such as piperine can significantly inhibit phase II enzymes like UDP-glucuronosyltransferase preventing the metabolic degradation of flavonoids (84, 96, 97). In addition, use of specific ABC transporter blockers such as lapatinib and nilotinib may improve the flavonoid bioavailability (83).

9.3. Nanocarriers for efficient delivery of flavonoids

The numerous factors limiting the efficacy of the flavonoids can be overcome by application of nanocarrier based approaches that offers several advantages such as prevention against metabolic degradation in the gastrointestinal tract, improvement of solubilization potential, and alteration of absorption pathways (Table 3) (98).

Table 3. The table shows various nano-engineered flavonoids developed for evaluation in different cancer models with their findings.
Nanocarrier Flavonoid Model system Inference References
Lipid nanoparticle system Quercetin, Naringenin, and Hesperetin In vitro digestion Enhanced relative bio-accessibility as LNP preserved well during oral digestion (113)
Multifunctional solid lipid nanoparticles Baicalein and fisetin Colon adenocarcinoma cells Electroporation of SLN lead to increased anti-cancer effects (485)
anti-cancer
DQAsomes Curcumin A549 cell and Caco-2 cells Enhanced antioxidant activity and mitochondrial targeting ability due to improved stability of curcumin-loaded DQAsomes (126)
Cationic lipid-based nanocarriers Genistein CT26 and HepG2 cell lines Mitochondrial depolarization, cytosolic cytochrme c release and activated caspase-9 augmented anti-cancer activity (486)
TPP conjugates Cyanostilbene derivatives HeLa, MCF-7, Hec-1A, KGN, HCT116, A549, 293T, and CCD-18Co cellsImmunodeficientBALB/c nu/nu mice Selective mitochondrial accumulation generates intracellular reactive oxygen species, decreases mitochondrial membrane potential and upregulates anti-cancer activity (487)
Chemically modified gallic acid Mammary adenocarcinomaTA3/Ha cell line Increased uncoupling effect and decreased ATP levels lead to hiked anti-proliferative effect in vivo due to selective mitochondrial uptake (488)
Polymeric nanoparticles Apigenin Swiss Albino mice Stable and efficient encapsulation of nanoparticles result in ROS mediated mitochondrial apoptosis (169)
Hesperetin C6 glioma cells Delivery resulted in enhanced uptake, sustained drug release and therapeutic efficacy (489)
Carbon based nanostructures Quercetin Human lung epithelial carcinoma A549 line Increased stability and no cytotoxicity of nano graphene oxide for oral delivery is reported (490)
Gold nanoparticles Quercetin Hormone-dependent (MCF-7) and hormone-independent (MDA-MB-231) breast cancer cell lines Induced apoptosis and suppressed EGFR signaling for -anti-neoplastic effects is reported with increased therapeutic efficacy (491)
Baicalin MCF-7 cells Induction of apoptosis showed anti-cancer effectiveness of conjugated nanoparticles (492)
Morin MCF-7 breast cancer cells Reducing and stabilizing nanoparticles endocytosed by cells and stimulate cell death via promoted DNA damage, cell cycle arrest, apoptosis (493)
Hesperetin Hepatocellular carcinoma - Hep3B cells Polymer functionalized nanoparticles inhibit proliferation and induce apoptosis (494)
TiO2 nanoparticles Quercetin 3T3 mouse fibroblast cells High bioavailability and stability with maximum antioxidant capacity (495)
Iron oxide nanoparticles Baicalein Triple negative breast cancer MDA-MB-231 cells Controlled release profile, cell cycle arrest, significant down-regulation of anti-apoptotic protein Bcl-2 and up-regulation of pro-apoptotic proteins (496)
Superparamagnetic iron oxide nanoparticles Luteolin U87, MCF-7, HeLa, L929 and A549 cell lines Reduced cell viability and higher apoptosis of cancer cells is reported (497)
Mesoporous silica nanoparticles Quercetin MDA MB 231 and MCF-7 breast cancer cell lines Enhanced cellular uptake and bioavailability lead to cell cycle arrest and apoptosis via Akt and Bax signaling pathway (137)

Extensive research work has been conducted in the direction towards enhancement of various biopharmaceutical and pharmacokinetic attributes of flavonoids. Sandhu et al., demonstrated a 3.5 and 3.3-fold enhancement in the drug release rates and 7.32 and 11.45-fold escalation in Cmax and AUC signifying the improvement in oral bioavailability of both tamoxifen and naringenin from self nano-emulsifying drug delivery system to achieve a synergistic anti-proliferative effect (99). Ragelle et al., demonstrated an improvement in the pharmacokinetic profile, a 24-fold increase in the relative bioavailability and increase in the anti-tumor efficacy of fisetin nanoemulsion in comparison to the free fisetin (100). Aminoflavone (AF) exhibits significant growth inhibitory effects at low doses in human TNBC in vitro models and complex anti-cancer effects. However, the in vivo studies and clinical trials have revealed AF mediated dose-limiting pulmonary toxicity, halting its progress in the clinic. In order to curb this menace, Brinkman et al., developed a unimolecular micellar nanoformulation of AF that displayed an increased in vivo therapeutic efficacy and significant tumor growth inhibitory effect in the xenograft model for EGFR-overexpressing TNBC (101). An increase in the water solubility, bioavailability and permeability across intestinal epithelial cells and anti-tumoral efficacy of Silibinin via encapsulation in niosomal formulation was demonstrated (102). Similarly, the improvement in the anti-cancer efficacy and oral bioavailability by encapsulation in PLGA nanoparticles- hydroxyl propyl beta cyclodextrin complex has also been reported (103). Encapsulation within a liposomal formulation significantly enhanced the inhibitory effect of luteolin on the growth on the CT26 colorectal carcinoma cell line compared with free luteolin (104). Likewise, encapsulation of naringenin in soluthin-maltodextrin-based nanocarrier resulted into a 116-fold increase in oral bioavailability, a 21-fold reduction in the in vitro cytotoxicity along with significant in vivo tumor suppression (105). The in vitro studies of naringenin loaded eudragit nanopartricles exhibited bioavailability (∼96-fold; p <0.05) as well as cytotoxicity (∼16-fold; p <0.001) and in vivo studies demonstrated significant tumor suppression (p <0.01) with subsequent improvement in survival rate compared to free naringenin (106). An increase in the in vitro cytotoxic efficacy of (−)-epicatechin against breast cancer cell lines (MCF-7, MDA-MB-231, MDA-MB-436 and SK-Br3) was reported by Perez-Ruiz et al., due to encapsulation in lecithin–chitosan nanoparticles. The developed formulation displayed a four-fold lower IC50 (85 μM) compared to free (−)-epicatechin (350 μM) and high selectivity to cancerous cells (107).

9.3.1. Polymer-based nanocarriers
9.3.1.1 Polymeric nanoparticles (PNPs)

PNPs are nano-colloidal cargos made up of a wide variety of biodegradable natural or synthetic high molecular weight polymers with size ranging from 10-1000 nm (84). A synthetic polymer usually includes poly-α-cyanoacrylate alkyl esters, polyvinyl alcohol, polylactic acid, polyglycolic acid, and polylactic- glycolic acid. Natural polymers may be further categorized into two groups: proteins and polysaccharides of plant origin and microbial or animal origin. Polymeric nanoparticles have been evaluated as potential vectors for flavonoid delivery in the recent times because their colloidal nature that may help in overcoming diverse physiological barriers, including the gastrointestinal mucosa and blood brain barrier (76, 108-110).

9.3.1.2 Polymeric micelles

Polymeric micelles demonstrate promising delivery of hydrophilic drugs via oral route. They offer various advantages such as (i) ability to manipulate the hydrophobicity and hydrophilicity within the polymeric system to accommodate a wide variety of drug molecules (Table 3); (ii) guarantee a high degree of sustained release (iii) provides hydrophilicity, biodegradable and non-toxic delivery; (iv) high stability in the GI tract ; (v) preferential uptake by specialized Peyer’s patches (M cells) and isolated follicles of GALT; and (vi) protection from degradation along with economical processing (98).

9.3.2. Lipid-based nanocarriers

The past decades have witnessed several advancements in lipids-based nanocarriers. These nanocarriers exhibit better biocompatibility, commercial scalability and adaptability, as well as distinctive mechanisms of absorption contributing to fine-tuning of the oral bioavailability, and avoidance of various physiological barriers such as gastric and colonic pH, intestinal micro flora, hepatic environment, first pass metabolism etc. Monoglycerides, diglycerides, triglycerides, and oils constitute the lipid excipients along with various combinations of phospholipids, sphingolipids, and even high fat meal. Lipid based nanosystems include micelles, solid lipid nanoparticles, nanostructured lipid carriers, nanoemulsions, microemulsions and liposomes that vary from 10 nm to hundreds of nanometers. Stabilization (electrostatic, steric or elecrosteric) acts as a critical factor for the fabrication of the colloidal systems (98).

9.3.2.1 Micelles

Micelles are self-assembled colloidal nanostructures (10 to 100 nm in diameter) that are made up of amphiphilic molecules, arranged in core-shell architecture with hydrophobic chains directed inwards making up the inner core and hydrophilic portion making up the surrounding corona. Micelles may take up several forms depending on the conditions and composition of the system. Bioactive moieties can be confined or uniformly distributed either in the core or in the palisade of micelles. Moreover, micellar systems have several other features to be considered as an effective delivery system, such as small size (typically <20 nm), thermodynamic and colloidal stability (98).

9.3.2.2 Microemulsions, nanoemulsions and self-emulsifying drug delivery systems

The recent couple of decades have witnessed advancements in the emulsion based systems for the conveyance of hydrophobic drugs of which microemulsions and nanoemulsions has garnered significant attention owing to their potential attributes of super solvency, thermodynamic or kinetic stability, miniature droplet size and high industrial scalability (84). Microemulsion, also known as swollen micelle existing in the submicron range, are optically isotropic and thermodynamically stable systems (111) comprising of an oil phase, an aqueous phase, a surfactant, and a co-surfactant. In contrast, nano-emulsions (often called mini-emulsions) are 10 to 100 nm sized oil droplets (smaller than the droplets present in ordinary emulsions) dispersed in aqueous media. Their potential advantageous characteristics such as optical transparency, low viscosity, enhanced functionality, and physical stability has gathered considerable interest in the pharmaceutical field. Stabilization of the systems is achieved by a mixture of emulsifier and co-emulsifying agent. In addition to these, self-microemulsifying and self-nanoemulsifying drug delivery systems, have emerged as successful alternatives to current emulsion based systems for oral delivery of flavonoid. These are composed of homogeneous blend of oil, surfactant, co-surfactant, and bioactive molecule that result in the formation of an oil-in water microemulsion and nanoemulsion upon exposure to aqueous media (112).

9.3.3. Solid lipid nanoparticles (SLN)

SLNs are defined as nano size particles (50-1000 nm) comprising of a solid hydrophobic core, made up of lipids and lipid-like molecules such as triacylglycerols or waxes, surrounded by a phospholipid monolayer and stabilized by means of surfactants as stabilizers. The therapeutically active agents can be either homogeneously embedded in the core of SLNs or localized in shell. SLNs provide a wide array of benefits such as good stability, bio-compatibility, application versatility, target ability by appropriate chemical modification, effective protection of encapsulated bioactive molecules, sustained drug release, and economical scale-up strategies (98).

9.3.4. Nanostructured lipid carriers (NLC)

NLC are modified versions of SLNs with the core containing a combination of both solid (fat) and liquid (oil) lipids at room temperature. These were developed to overcome the problems associated with SLNs, such as improper drug loading, poor long-term stability, and subsequent leakage of drug during storage. NLC have therefore gathered much attention in recent times as potential alternative of SLN due to their attractive traits such as increased encapsulation efficiency, controlled release of therapeutic agents and henceforth improved bioavailability along with maintenance of physical and chemical stability (113-115).

9.3.5. Molecular inclusion complexes

Apart from the aforementioned approaches, better solubility and improved physical and chemical stability of therapeutically active moieties can also be achieved by physical complexation with other molecules. A molecular inclusion complex is characterized by a physical conjugation between the host and the guest (active ingredient) molecule (116, 117).

9.3.5.1. Cyclodextrins inclusion complexes

Cyclodextrins are natural oligosaccharides belonging to a family of molecules that consist of several glucopyranoses bound together to form a ring with a distinctive lipophilic cavity and hydrophilic outer surface that ensure improved aqueous solubility of the complex (enclosing hydrophobic moieties). The CD-complexation has brought about a drastic change in the delivery of therapeutically active lipophilic molecules by safeguarding against degradation and microbial contamination, reducing the incompatibility issues, and improving the availability of bioactive compound at the site of action (116, 118).

9.3.5.2. Phytosomes®

Phytosome®, a patented technology, is a vesicular drug delivery system developed to tackle various bioavailability issues and involves complexation between the phyto-constituents with naturally occurring phospholipid. Phytosome® is developed by forming a molecular complex between phosphatidylcholine and plant components (1:1 or a 2:1) through chemical bonds making them highly compatible and highly bioavailable (119). They can minimize the dose due to sustained drug release pattern (120-122).

10. MITOCHONDRIAL TARGETING NANO-ENGINEERED STRATEGIES FOR BIO-THERAPEUTICS

Extensive work in the recent times have been done for the development of a delivery system with ability to surpass biological barriers; avoidance of premature deactivation of bioactive molecules; and elevation of the intracellular availability of the drugs at their target site (123). Multifaceted cellular regulatory mechanisms mark mitochondria as a critical target for the cancer-prevention strategy. Specific characteristics of mitochondria include mitochondrial trans-membrane electrochemical gradient, protein import machinery, pH difference, and fusion process. These advantages are also being exploited for developing targeted strategies, which are known as mitochondria medicine (124). For mito-targeting of therapeutic agents, approaches involve either loading of drugs into surface-modified nanoparticles or conjugation of the targeting moiety with model drugs. In this context, specialized nano-delivery systems are being utilized such as delocalized lipophilic cations, polymeric NPs, mitochondrial targeting peptide, carbon nanostructures, blended nanoparticles, metal nanoparticles, and mesoporous silica nanoparticles (Figure 7). Uptake of mitochondria targeted nano-carrier is shown in the Figure 8.

Figure 7.

Detailed classification of different nanocarriers based strategies for effective flavonoid delivery and mitochondrial targeting.

Figure 8.

Schematic representation of the delivery mechanism of different mitochondria-targeted nanocarriers

10.1. Liposomal nanocarriers for mitochondrial targeting

Liposomes are effective antigen carriers that possess great translational potential for mitochondrial delivery owing to various favorable features such as biodegradability, non-toxicity and non-immunogenicity, and antigen loading properties. Further, these are one of the oldest drug delivery systems and most successful FDA approved therapeutics. Over a couple of decades, liposomes have emerged as the most investigated class of nano delivery systems for the mitochondrial targeted therapeutics (125).

10.2. DQAsomes

DQAsomes represent as the most standout systems amongst the most broadly reviewed nanocarriers. These are composed of dequalinium chloride molecules that are self-assembled into vesicle-like fashion in an aqueous suspension. These possess the ability to enter cells via endocytosis along with evading endosomal uptake (126).

10.3. MITO-Porter

MITO-Porter is a novel liposome-based approach possessing selective mitochondrial target ability that can encapsulate and deliver various types of cargo molecules irrespective of their size and physicochemical properties via membrane fusion (127). However, the delivery of bioactive natural moieties such as flavonoids by means of MITO-Porter is unexplored which require more focussed research (128).

10.4. Delocalized lipophilic cations (DLCs)

DLCs are small lipophilic, delocalised positive charge carrying molecules; whose mitochondrial targeting ability in the current scenario is under exploration. Given the lipophilic nature and resonance stabilized delocalization of the charge, they can easily pass through the phospholipid bilayers leading to increased accumulation in mitochondria and can be conjugated to various cargos (including antioxidants, nucleic acids, and spin traps) for mito-targeting purposes. Some of the commercialized DLCs based systems include MitoTracker™ and TPP (129).

10.4.1. TPP-conjugates

Among the available DLCs, triphenylphosphonium (TPP) conjugates stands out as the most extensively studied system for mitochondrial target ability. The TPP cation consists of three hydrophobic phenyl rings surrounding a cationic charged phosphorus atom thereby possessing intrinsic hydrophobicity and mitochondrial selective accumulating properties. Due to this particular attribute, the specificity and therapeutic efficacy can be drastically increased with a concomitant reduction of harmful effects, which is very important to overcome the resistance of some cancer cells (123).

10.5. Polymeric NPs

The mito-targeted engineered polymeric nanoparticles demonstrate a flexible application in therapeutics against diseases such as cancer, diabetes, and Alzheimer’s disease. Notably, FDA-approved poly (lacticco-glycolic acid) (PLGA) and polyethylene glycol (PEG)-based materials are used for synthesis of nanoparticle, providing a distinctive edge in terms of biocompatibility. A mitochondrial-targeted polymer based systems with TPP moiety at the terminus using polymer blending technology has been reported that ensure fine tuning of the size as well as surface charge (103).

10.6. Mitochondrial targeting peptide

A variety of amino acid- and peptides-based approaches provide several advantages over DLCs, such as avoiding endosomal and/or lysosomal sequestration leading to their accumulation in the mitochondria, better biocompatibility, as well as straightforward synthesis and amide coupling. These sequences are normally derived from cytosolic synthesized proteins destined for trafficking to mitochondria specified by their Arg, Ala, or Ser rich N-terminal sequence having alternating arrangement of hydrophobic and positively charged amino acids forming an amphiphilic α-helix. Even though MTPs shows noteworthy mitochondrial target ability but their applicability is limited due to retracted generation of soluble and cell permeable conjugates. Numerous synthetic peptides demonstrating mitochondrial targeting activity along with intrinsic pharmacological properties have been reported (123, 130).

10.7. Carbon nanostructures

In the current scenario, CNTs have emerged as highly versatile and effective nanocarriers for the conveyance of various drugs; but, their role in mitochondrial medicine is still under investigation. Intracellular components target ability of CNTs is currently at its infancy stage. Several carbon based nanostructures for the targeted delivery of bioactive molecules in mitochondria have been evidenced. In an exploratory study, fluorescein isothiocyanate (FITC)-labelled PL-PEG (PL-PEG-FITC)-functionalized SWCNTs (SWCNT-PL-PEG-FITC) was found to be localized at mitochondria in tumor and normal cells in a mitochondrial transmembrane potential-dependent manner (123, 131).

10.8. Blended nanoparticles

Blended biodegradable polymers is a promising polymer blending technique that provide effective delivery of bioactive molecules to the mitochondria (132).

10.8.1. Metal nanoparticles

These include gold, silver, platinum, iron oxide and titanium dioxide based nanosystems ranging within 10 nm size and provide an array of anti-oxidant capabilities along with ease of attachment of mitochondrial targeting ligands. However, their safety and efficacy is one of the major concerns among scientists. Several studies elucidating the toxicity profile of metal based nanocarriers have been reported (133-135). Lately, studies have reported the delivery of plant-based anti-cancer agents by means of metal nanoparticles with enhanced target ability (125).

10.8.2. Mesoporous silica nanoparticles

Mesoporous silica’s are solid porous structure with numerous empty capillaries (mesopores) arranged in a honeycomb-like arrangement that can accommodate bioactive molecules in massive manifolds. These nano-frameworks offer flexibility in tuning the pore size, chemical and mechanical stability, and subsequent biocompatibility; thereby gathering a great attention for the delivery of flavonoids (136, 137).

11. NANO-ENGINEERED FLAVONOIDS TARGETING MITOCHONDRIAL-INDUCED EPIGENETIC MODIFICATIONS

Several groups have illustrated the combinatorial effects of flavonoids with anti-cancer agents in reducing drug resistance, tumor recurrence and metastasis through nano-engineered systems. The mitochondrial targeting strategy through flavonoids may serve as an effective therapeutic approach to combat cancer stem cells (CSCs). For instance, one such combinatorial synergistic approach using doxorubicin and quercetin was demonstrated by Fang et al., using hyaluronic acid tethered mesoporous silica nanoparticles for gastric carcinoma chemotherapy (138). Signaling pathways such as Hh, Wnt/β-catenin and Notch pathways playing an important role in differentiation of normal stem cells offer an attractive approach to target CSCs. Wu et al., in their earlier study demonstrated anti-proliferative and apoptosis inducing activity of 2’ Hydroxyflavanone (2HF) via blockage of the Akt/STAT3 signaling pathway in prostate cancer. The subsequent studies further investigated the effect of 2HF on EMT, and cell migration and invasion using Wnt/β catenin signaling pathway by suppressing GSK 3 β phosphorylation, β catenin expression and transactivation (104). There have been several reports where the anti-tumor and anti-metastatic effects of flavonoids have been demonstrated. Dai et al., (139) reported the anti-carcinogenic effect of bacailin using tumor xenograft model reported antitumor activity of silybin in human HCC HepG2 using both in vitro and in vivo conditions (140). The results suggested a decrease in the expression of the Notch1 intracellular domain (NICD), RBP-Jκ, and Hes1 proteins, upregulation of apoptosis pathway-related protein Bax, and downregulation in Bcl2, survivin, and cyclin D1. Lim et al., illustrated caused a dose-dependent growth inhibition of various brain tumor cell cultures and neurospheres through curcumin encapsulated in polymeric nanoparticles, effectively blocking the CSC-associated Hh pathway and reducing IGF and STAT3 levels (141).

Several evidences suggesting the use of plant-derived phytoconstitutents in the treatment of chemo-resistant cells have reported. Mohammadian et al., reported significant inhibitory effect on AGS human gastric cell line than free chrysin (142). The study also has elucidated successful restoration of tumor suppressor miRNA expression levels. Minimization or reversion of drug resistance using MCF-7/ADR tumor was well demonstrated by Lv et al., using biotin conjugated doxorubicin and quercetin co-loaded polymeric nanoparticles. The resultant nanoparticle system reported a significant inhibition of the activity and expression of P-glycoprotein and in MCF-7/ADR cells both in vitro and in vivo conditions (143). A synergy in the combinantion therapy of paclitaxel and difluorinated curcumin was observed by Gawde et al., against gynaecological tumors (144).

Mitochondrial stress associated oxidative radical injury may alter cellular epigenome which in turn may initiate the process of carcinogenesis, therefore, an effective protective agent should be capable of maintaining epigenetic stability. Analysis of post-translational histone modifications showed that NP.SB potentially protects B/CMBA cells from epigenetic injury following exposure to pro-oxidants (14). In recent study, we observed that in addition to protecting mitochondria from pro-oxidants induced alterations, encapsulated SB fraction (NP.SB) significantly protects cellular epigenome. Exposure of cells to pro-oxidants resulted in significant epigenomic alterations, as evident from increased methylation of H3K9me1 and H4K20me3 and phosphorylation of H3 and H2A histones. While hyperubiquitination of uH2A/uH2B and hypoacetylation of AcH3 further highlighted the seriousness of these alterations following pro-oxidants exposure. In contrast, no such alterations were observed among cells pre-treated with NP.SB. Altered histone modification pattern modulates and affect DNA methylation machinery, which thus alters the expression of different genes important for cellular integrity and polarity. In addition, studies have shown that dysregulated mitochondrial functioning results in the activation of retrograde signalling cascade which consequently affects nuclear gene arrangements (145, 146). In a recent study, NP.SB pre-treatment regulated DNA methylation at p16 promoter regions even after pro-oxidants treatment. Furthermore, the un-altered levels of let-7a and let-7b in NP.SB pre-treated cells showed the ability of this encapsulated fraction to protect cells from transcriptional dysregulations due to altered histone profile, which were significantly reported among pro-oxidants alone treated cells.

12. CONCLUDING REMARKS

Nutri-epigenomics is an emerging field in terms of dosage, targeting, tissue distribution, bioavailability, molecular and/or cellular interactions that limits the clinical utility of several plant secondary metabolites including flavonoids. Since, cancer is characterized by both genetic and epigenetic instability; it requires multifaceted protective strategies for effective tumour cell targeting while conferring better protection of normal cells from adverse drug-induced toxicity. In this regard, nano-engineered systems encapsulating bioactive flavonoids holds great promise for improved delivery and selective tumour cell targeting with better efficacy and pharmacokinetic properties. Recently, several nanocarrier based approaches, including polymeric nanoparticles, SLNs, liquid crystals, liposomes, and microemulsions have been employed to overcome the limitations associated with clinical use of flavonoids. Overall, nano-engineered systems offer attractive opportunities for the safe, effective, sustained and targeted delivery of flavonoids to reshape the mitochondrial mediated epigenetic regulation for cancer therapeutics. However, challenges such as inquesting their targeting efficiency, meeting up the regulatory international standards and feasibility of scale-up are few concerns which warrants attention for effective bench-to bedside translation of flavonoids as broad spectrum cancer protective agents.

13. ACKNOWLEDGEMENT

The authors are thankful to Council of Scientific and Industrial Research (CSIR) and University Grant Commission (UGC), Government of India, New Delhi for providing necessary assistance. The award of Junior and Senior Research Fellowships to NB by Department of Biotechnology (DBT), Government of India, New Delhi is gratefully acknowledged.

Abbreviations: ADMET, absorption, distribution, metabolism, excretion, and toxicity; ATM, ataxia telangiectasia-mutated; sATP, adenosine triphosphate; Bcl-2, B-cell lymphoma 2; BNIP3, BCL2 Interacting Protein 3; BRCT, BRCA1 C Terminus; CMC, critical micelle concentration; CSCs, cancer stem cells; DLCs, Delocalized lipophilic cations; DNMTs, DNA methyltransferases; DUBs, deubiquitinases; EGCG, epigallocatechin gallate; EMT, epithelial-mesenchymal transition; EZH2, enhancer of zeste homolog 2; GALT, Gut-associated lymphoid tissue; GATA4, GATA Binding Protein 4; H2Bub, 1monoubiquitination of histone H2B at lysine 120; H3K4, histone H3 lysine 4; HAT, histone acetyl transferase; HDAC1, histone deacetylase 1; HMT, histone methyl transferase; IUIM, inverted ubiquitin interaction motif; KDM, histone methyl transferase; LSD1, lysine demethylates; M cells, specialized microfold cells; MeCP, methyl CpG binding protein; miRNA microRNA; MOF, males absent on the first; MYST, Moz-Ybf2/Sas3-Sas2-Tip60; NAD+, nicotinamide adenine dinucleotide; NICD, Notch1 intracellular domain; NLC, nanostructured lipid carriers; NP.SB, nano-engineered flavonoid rich fraction from Selaginella bryopteris; PARP-1, poly (ADP-ribose) polymerase 1; PD, Pharmacodynamics; PK, Pharmacokinetics; PLGA, poly(lactic-co-glycolic acid; PNP, polymeric nanoparticles; RASSF1A, Ras association domain family 1 isoform A; RNF20, RING finger proteins; RNF40, RING finger proteins; ROS, reactive oxygen species; SAH, S-adenosylhomocystein; SAM, S-adenosylmethionine; SAM/SAH ratio, SAM to SAH ratios; SB.Fr., flavonoid rich fraction from Selaginella bryopteris; SLN, solid lipid nanoparticles; TET, ten eleven translocation; γH2AX, phosphorylated histone 2AX

References
[1]
Feinberg , A.P. , Ohlsson , R. , and Henikoff , S. The epigenetic progenitor origin of human cancer. Nat Rev Genet 7 , 21 33 ( 2006) DOI: 10.1038/nrg1748
[2]
Conte , M. , and Altucci , L. Molecular pathways: the complexity of the epigenome in cancer and recent clinical advances. Clin Cancer Res 18 , 5526 5534 ( 2012) DOI: 10.1158/1078-0432.CCR-12-2037
[3]
Ladurner , A.G. Chromatin places metabolism center stage. Cell 138 , 18 20 ( 2009) DOI: 10.1016/j.cell.2009.06.025
[4]
Vaquero , A. , and Reinberg , D. Calorie restriction and the exercise of chromatin. Genes Dev 23 , 1849 1869 ( 2009) DOI: 10.1101/gad.1807009
[5]
Guarente , L. The logic linking protein acetylation and metabolism. Cell Metab 14 , 151 153 ( 2011) DOI: 10.1016/j.cmet.2011.07.007
[6]
Hardy , T.M. , and Tollefsbol , T.O. Epigenetic diet: impact on the epigenome and cancer. Epigenomics 3 , 503 518 ( 2011) DOI: 10.2217/epi.11.71
[7]
Godoy , L.D. , Lucas , J.E. , Bender , A.J. , Romanick , S.S. , and Ferguson , B.S. Targeting the epigenome: Screening bioactive compounds that regulate histone deacetylase activity. Mol Nutr Food Res 61 , 1 17 ( 2017) DOI: 10.1002/mnfr.201600744
[8]
Conte , M. , De Palma , R. , and Altucci , L. HDAC inhibitors as epigenetic regulators for cancer immunotherapy. Int J Biochem Cell Biol 98 , 65 74 ( 2018) DOI: 10.1016/j.biocel.2018.03.004
[9]
Thakur , V.S. , Deb , G. , Babcook , M.A. , and Gupta , S. Plant phytochemicals as epigenetic modulators: role in cancer chemoprevention. Aaps J 16 , 151 163 ( 2014) DOI: 10.1208/s12248-013-9548-5
[10]
Prieto-Dominguez , N. , Garcia-Mediavilla , M.V. , Sanchez-Campos , S. , Mauriz , J.L. , and Gonzalez-Gallego , J. Autophagy as a molecular target of flavonoids underlying their protective effects in human disease. Curr Med Chem 25 , 814 838 ( 2018) DOI: 10.2174/0929867324666170918125155
[11]
Putteeraj , M. , Lim , W.L. , Teoh , S.L. , and Yahaya , M.F. Flavonoids and its neuroprotective effects on brain ischemia and neurodegenerative diseases. Curr Drug Targets. 19 ( 14 ): 1710 1720 ( 2018) DOI: 10.2174/1389450119666180326125252
[12]
Mishra , P.K. , Raghuram , G.V. , Bhargava , A. , Ahirwar , A. , Samarth , R. , Upadhyaya , R. , Jain , S.K. , and Pathak , N. In vitro and in vivo evaluation of the anticarcinogenic and cancer chemopreventive potential of a flavonoid-rich fraction from a traditional Indian herb Selaginella bryopteris. Br J Nutr 106 , 1154 1168 ( 2011) DOI: 10.1017/S0007114511001498
[13]
Mishra , P.K. , Bunkar , N. , Raghuram , G.V. , Khare , N.K. , Pathak , N. , and Bhargava , A. Epigenetic dimension of oxygen radical injury in spermatogonial epithelial cells. Reprod Toxicol 52 , 40 56 ( 2015) DOI: 10.1016/j.reprotox.2015.02.006
[14]
Bhargava , A. , Pathak , N. , Seshadri , S. , Bunkar , N. , Jain , S.K. , Kumar , D.M. , Lohiya , N.K. , and Mishra , P.K. Pre-clinical validation of mito-targeted nano-engineered flavonoids isolated from Selaginella bryopteris (Sanjeevani) as a novel cancer prevention strategy. Anticancer Agents Med Chem 18 ( 13 ), 1860 1874 ( 2018) DOI: 10.2174/1871520618666171229223919
[15]
Naviaux , R.K. Mitochondrial control of epigenetics. Cancer Biol Ther 7 , 1191 1193 ( 2008) DOI: 10.4161/cbt.7.8.6741
[16]
Minocherhomji , S. , Tollefsbol , T.O. , and Singh , K.K. Mitochondrial regulation of epigenetics and its role in human diseases. Epigenetics 7 , 326 334 ( 2012) DOI: 10.4161/epi.19547
[17]
Mishra , P.K. , Raghuram , G.V. , Jain , D. , Jain , S.K. , Khare , N.K. , Pathak , N. Mitochondrial oxidative stress-induced epigenetic modifications in pancreatic epithelial cells. Int J Toxicol 33 ( 2 ), 116 129 ( 2014) http://ijt.sagepub.com/content/33/2/116
[18]
Robertson , K.D. DNA methylation and human disease. Nat Rev Genet 6 , 597 610 ( 2005) DOI: 10.1038/nrg1655
[19]
Portela , A. , and Esteller , M. Epigenetic modifications and human disease. Nat Biotechnol 28 , 1057 1068 ( 2010) DOI: 10.1038/nbt.1685
[20]
Morris , M.R. , and Latif , F. The epigenetic landscape of renal cancer. Nat Rev Nephrol 13 , 47 60 ( 2017) DOI: 10.1038/nrneph.2016.168
[21]
Ghosh , K. , O’neil , K. , and Capell , B.C. Histone modifiers: Dynamic regulators of the cutaneous transcriptome. J Dermatol Sci 89 , 226 232 ( 2018) DOI: 10.1016/j.jdermsci.2017.12.006
[22]
Mansouri , L. , Wierzbinska , J.A. , Plass , C. , and Rosenquist , R. Epigenetic deregulation in chronic lymphocytic leukemia: Clinical and biological impact. Semin Cancer Biol 51 , 1 11 ( 2018) DOI: 10.1016/j.semcancer.2018.02.001
[23]
Chen , C.C. , Wang , K.Y. , and Shen , C.K. DNA 5-methylcytosine demethylation activities of the mammalian DNA methyltransferases. J Biol Chem 288 , 9084 9091 ( 2013) DOI: 10.1074/jbc.M112.445585
[24]
Kim , N.H. , Sung , H.Y. , Choi , E.N. , Lyu , D. , Choi , H.J. , Ju , W. , and Ahn , J.H. Aberrant DNA methylation in the IFITM1 promoter enhances the metastatic phenotype in an intraperitoneal xenograft model of human ovarian cancer. Oncol Rep 31 , 2139 2146 ( 2014) DOI: 10.3892/or.2014.3110
[25]
Sung , H.Y. , Ju , W. , and Ahn , J.H. DNA hypomethylation-mediated overexpression of carbonic anhydrase 9 induces an aggressive phenotype in ovarian cancer cells. Yonsei Med J 55 , 1656 1663 ( 2014) DOI: 10.3349/ymj.2014.55.6.1656
[26]
Matilainen , O. , Quirós , P.M. , and Auwerxm J. Mitochondria and Epigenetics - Crosstalk in Homeostasis and Stress. Trends Cell Biol. 27 ( 6 ), 453 463 ( 2017) DOI: 10.1016/j.tcb.2017.02.004
[27]
Ghosh , S. , Ranawat , A.S. , Tolani , P. , Scaria , V. Mitoepigenome KB a comprehensive resource for human mitochondrial epigenetic data. Mitochondrion 42 , 54 58 ( 2018) DOI: 10.1016/j.mito.2017.11.001
[28]
Stimpfel , M. , Jancar , N. , Virant-Klun , I. New challenge: Mitochondrial epigenetics? Stem Cell Rev 14 , 13 26 . ( 2014) DOI: 10.1007/s12015-017-9771-z
[29]
van der Wijst , M.G. , Rots , M.G. Mitochondrial epigenetics: an overlooked layer of regulation? Trends Genet 31 ( 7 ), 353 356 ( 2015) DOI: 10.1016/j.tig.2015.03.009
[30]
Liu , B. , Song , J. , Luan , J. , Sun , X. , Bai , J. , Wang , H. , Li , A. , Zhang , L. , Feng , X. , and Du , Z. Promoter methylation status of tumor suppressor genes and inhibition of expression of DNA methyltransferase 1 in non-small cell lung cancer. Exp Biol Med (Maywood) 241 , 1531 1539 ( 2016) DOI: 10.1177/1535370216645211
[31]
Liu , H. , Liu , K. , Huang , Z. , Park , C.M. , Thimmegowda , N.R. , Jang , J.H. , Ryoo , I.J. , He , L. , Kim , S.O. , Oi , N. , Lee , K.W. , Soung , N.K. , Bode , A.M. , Yang , Y. , Zhou , X. , Erikson , R.L. , Ahn , J.S. , Hwang , J. , Kim , K.E. , Dong , Z. , and Kim , B.Y. A chrysin derivative suppresses skin cancer growth by inhibiting cyclin-dependent kinases. J Biol Chem 288 , 25924 25937 ( 2013) DOI: 10.1074/jbc.M113.464669
[32]
Su , J. , Wang , F. , Cai , Y. , and Jin , J. The Functional Analysis of Histone Acetyltransferase MOF in Tumorigenesis. Int J Mol Sci 17 ( 2016) DOI: 10.3390/ijms17010099
[33]
Cai , M. , Hu , Z. , Liu , J. , Gao , J. , Tan , M. , Zhang , D. , Zhu , L. , Liu , S. , Hou , R. , and Lin , B. Expression of hMOF in different ovarian tissues and its effects on ovarian cancer prognosis. Oncol Rep 33 , 685 692 ( 2015) DOI: 10.3892/or.2014.3649
[34]
Kampilafkos , P. , Melachrinou , M. , Kefalopoulou , Z. , Lakoumentas , J. , and Sotiropoulou-Bonikou , G. Epigenetic modifications in cutaneous malignant melanoma: EZH2, H3K4me2, and H3K27me3 immunohistochemical expression is enhanced at the invasion front of the tumor. Am J Dermatopathol 37 , 138 144 ( 2015) DOI: 10.1097/DAD.0b013e31828a2d54
[35]
Guo , J. , Cai , J. , Yu , L. , Tang , H. , Chen , C. , and Wang , Z. EZH2 regulates expression of p57 and contributes to progression of ovarian cancer in vitro and in vivo. Cancer Sci 102 , 530 539 ( 2011) DOI: 10.1111/j.1349-7006.2010.01836.x
[36]
Raghuram , G.V. , and Mishra , P.K. Stress induced premature senescence: a new culprit in ovarian tumorigenesis? Ind J Med Res 140 , S120 ( 2014)
[37]
Li , Y. , Wan , X. , Wei , Y. , Liu , X. , Lai , W. , Zhang , L. , Jin , J. , Wu , C. , Shao , Q. , Shao , G. , and Lin , Q. LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion. Oncol Rep 35 , 3586 3592 ( 2016) DOI: 10.3892/or.2016.4729
[38]
Dawson , M.A. , and Kouzarides , T. Cancer epigenetics: from mechanism to therapy. Cell 150 , 12 27 ( 2012) DOI: 10.1016/j.cell.2012.06.013
[39]
Bhargava , A. , Khare , N.K. , Bunkar , N. , Lenka , R.K. , and Mishra , P.K. Role of mitochondrial oxidative stress on lymphocyte homeostasis in patients diagnosed with extra-pulmonary tuberculosis. Cell Biol Int 40 ( 2 ), 166 176 ( 2016) DOI: 10.1002/cbin.10549
[40]
Chang , F.T. , Chan , F.L. , Jd , R.M. , Udugama , M. , Mayne , L. , Collas , P. , Mann , J.R. , and Wong , L.H. CHK1-driven histone H3.3 serine 31 phosphorylation is important for chromatin maintenance and cell survival in human ALT cancer cells. Nucleic Acids Res 43 , 2603 2614 ( 2015) DOI: 10.1093/nar/gkv104
[41]
Mei , L. , Hu , Q. , Peng , J. , Ruan , J. , Zou , J. , Huang , Q. , Liu , S. , and Wang , H. Phospho-histone H2AX is a diagnostic and prognostic marker for epithelial ovarian cancer. Int J Clin Exp Pathol 8 , 5597 5602 ( 2015)
[42]
Mcclurg , U.L. , and Robson , C.N. Deubiquitinating enzymes as oncotargets. Oncotarget 6 , 9657 9668 ( 2015) DOI: 10.18632/oncotarget.3922
[43]
Meas , R. , and Mao , P. Histone ubiquitylation and its roles in transcription and DNA damage response. DNA Repair (Amst) 36 , 36 42 ( 2015) DOI: 10.1016/j.dnarep.2015.09.016
[44]
Dickson , K.A. , Cole , A.J. , Gill , A.J. , Clarkson , A. , Gard , G.B. , Chou , A. , Kennedy , C.J. , Henderson , B.R. , Fereday , S. , Traficante , N. , Alsop , K. , Bowtell , D.D. , Defazio , A. , Clifton-Bligh , R. , and Marsh , D.J. The RING finger domain E3 ubiquitin ligases BRCA1 and the RNF20/RNF40 complex in global loss of the chromatin mark histone H2B monoubiquitination (H2Bub1) in cell line models and primary high-grade serous ovarian cancer. Hum Mol Genet 25 , 5460 5471 ( 2016) DOI: 10.1093/hmg/ddw362
[45]
Knowling , S. , and Morris , K.V. Non-coding RNA and antisense RNA. Nature’s trash or treasure? Biochimie 93 , 1922 1927 ( 2011) DOI: 10.1016/j.biochi.2011.07.031
[46]
Kong , Y.W. , Ferland-Mccollough , D. , Jackson , T.J. , and Bushell , M. microRNAs in cancer management. Lancet Oncol 13 , e249 258 ( 2012) DOI: 10.1016/S1470-2045(12)70073-6
[47]
Thorsson , V. , Gibbs , D.L. , Brown , S.D. , Wolf , D. , Bortone , D.S. , Ou Yang , T.H. , Porta-Pardo , E. , Gao , G.F. , Plaisier , C.L. , Eddy , J.A. , Ziv , E. , Culhane , A.C. , Paull , E.O. , Sivakumar , I.K.A. , Gentles , A.J. , Malhotra , R. , Farshidfar , F. , Colaprico , A. , Parker , J.S. , Mose , L.E. , Vo , N.S. , Liu , J. , Liu , Y. , Rader , J. , Dhankani , V. , Reynolds , S.M. , Bowlby , R. , Califano , A. , Cherniack , A.D. , Anastassiou , D. , Bedognetti , D. , Rao , A. , Chen , K. , Krasnitz , A. , Hu , H. , Malta , T.M. , Noushmehr , H. , Pedamallu , C.S. , Bullman , S. , Ojesina , A.I. , Lamb , A. , Zhou , W. , Shen , H. , Choueiri , T.K. , Weinstein , J.N. , Guinney , J. , Saltz , J. , Holt , R.A. , Rabkin , C.E. , Lazar , A.J. , Serody , J.S. , Demicco , E.G. , Disis , M.L. , Vincent , B.G. , and Shmulevich , L. The immune landscape of cancer. Immunity 48 , 812 830 e814 ( 2018) DOI: 10.1016/j.immuni.2018.03.023
[48]
Jain , D. , Pathak , N. , Khan , S. , Raghuram , G.V. , Bhargava , A. , Samarth , R. , and Mishra , P.K. Evaluation of cytotoxicity and anticarcinogenic potential of Mentha leaf extracts. Int J Toxicol 30 , 225 236 ( 2011) DOI: 10.1177/1091581810390527
[49]
Gibellini , L. , Bianchini , E. , De Biasi , S. , Nasi , M. , Cossarizza , A. , and Pinti , M. Natural Compounds Modulating Mitochondrial Functions. Evid Based Complement Alternat Med 2015, 527209 ( 2015) DOI: 10.1155/2015/527209
[50]
Singh , T. , Prasad , R. , and Katiyar , S.K. Therapeutic intervention of silymarin on the migration of non-small cell lung cancer cells is associated with the axis of multiple molecular targets including class 1 HDACs, ZEB1 expression, and restoration of miR-203 and E-cadherin expression. Am J Cancer Res 6 , 1287 1301 ( 2016)
[51]
Busch , C. , Burkard , M. , Leischner , C. , Lauer , U.M. , Frank , J. , and Venturelli , S. Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin Epigenetics 7 , 64 ( 2015) DOI: 10.1186/s13148-015-0095-z
[52]
Borutinskaitė , V. , Virkšaitė , A. , Gudelytė , G. , and Navakauskienė , R. Green tea polyphenol EGCG causes anti-cancerous epigenetic modulations in acute promyelocytic leukemia cells. Leuk Lymphoma 59 ( 2 ), 469 478 ( 2018) DOI: 10.1080/10428194.2017.1339881
[53]
Vanden Berghe , W. Epigenetic impact of dietary polyphenols in cancer chemoprevention: lifelong remodeling of our epigenomes. Pharmacol Res 65 , 565 576 ( 2012) DOI: 10.1016/j.phrs.2012.03.007
[54]
Gerhauser , C. Cancer chemoprevention and nutriepigenetics: state of the art and future challenges. Top Curr Chem 329 , 73 132 ( 2013) DOI: 10.1007/128_2012_360
[55]
Joseph , P.V. , Abey , S.K. , and Henderson , W.A. Emerging Role of Nutri-Epigenetics in Inflammation and Cancer. Oncol Nurs Forum 43 , 784 788 ( 2016) DOI: 10.1188/16.ONF.784-788
[56]
Ulrich , C.M. , Reed , M.C. , and Nijhout , H.F. Modeling folate, one-carbon metabolism, and DNA methylation. Nutr Rev 66 Suppl 1 , S27 30 ( 2008) DOI: 10.1111/j.1753-4887.2008.00062.x
[57]
Bistulfi , G. , Vandette , E. , Matsui , S. , and Smiraglia , D.J. Mild folate deficiency induces genetic and epigenetic instability and phenotype changes in prostate cancer cells. BMC Biol 8 , 6 ( 2010) DOI: 10.1186/1741-7007-8-6
[58]
Chen , N.C. , Yang , F. , Capecci , L.M. , Gu , Z. , Schafer , A.I. , Durante , W. , Yang , X.F. , and Wang , H. Regulation of homocysteine metabolism and methylation in human and mouse tissues. FASEB J 24 , 2804 2817 ( 2010) DOI: 10.1096/fj.09-143651
[59]
Zhang , H.P. , Wang , Y.H. , Ma , S.C. , Zhang , H. , Yang , A.N. , Yang , X.L. , Zhang , M.H. , Sun , J.M. , Hao , Y.J. , and Jiang , Y.D. Homocysteine inhibits endothelial progenitor cells proliferation via DNMT1-mediated hypomethylation of Cyclin A. Exp Cell Res 362 , 217 226 ( 2018) DOI: 10.1016/j.yexcr.2017.11.021
[60]
Srivastava , S. , Somasagara , R.R. , Hegde , M. , Nishana , M. , Tadi , S.K. , Srivastava , M. , Choudhary , B. , and Raghavan , S.C. Quercetin, a natural flavonoid interacts with DNA, arrests cell cycle and causes tumor regression by activating mitochondrial pathway of apoptosis. Sci Rep 6 , 24049 ( 2016) DOI: 10.1038/srep24049
[61]
Zhou , R. , Xu , L. , Ye , M. , Liao , M. , Du , H. , and Chen , H. Formononetin inhibits migration and invasion of MDA-MB-231 and 4T1 breast cancer cells by suppressing MMP-2 and MMP-9 through PI3K/AKT signaling pathways. Horm Metab Res 46 , 753 760 ( 2014) DOI: 10.1055/s-0034-1376977
[62]
Ahn- Jarvis , J.H. , Clinton , S.K. , Grainger , E.M. , Riedl , K.M. , Schwartz , S.J. , Lee , M.L. , Cruz-Cano , R. , Young , G.S. , Lesinski , G.B. , and Vodovotz , Y. Isoflavone pharmacokinetics and metabolism after consumption of a standardized soy and soy-almond bread in men with asymptomatic prostate cancer. Cancer Prev Res (Phila) 8 , 1045 1054 ( 2015) DOI: 10.1158/1940-6207.CAPR-14-0465
[63]
Crew , K.D. , Ho , K.A. , Brown , P. , Greenlee , H. , Bevers , T.B. , Arun , B. , Sneige , N. , Hudis , C. , Mcarthur , H.L. , Chang , J. , Rimawi , M. , Cornelison , T.L. , Cardelli , J. , Santella , R.M. , Wang , A. , Lippman , S.M. , and Hershman , D.L. Effects of a green tea extract, Polyphenon E, on systemic biomarkers of growth factor signalling in women with hormone receptor-negative breast cancer. J Hum Nutr Diet 28 , 272 282 ( 2015) DOI: 10.1111/jhn.12229
[64]
Lesinski , G.B. , Reville , P.K. , Mace , T.A. , Young , G.S. , Ahn-Jarvis , J. , Thomas-Ahner , J. , Vodovotz , Y. , Ameen , Z. , Grainger , E. , Riedl , K. , Schwartz , S. , and Clinton , S.K. Consumption of soy isoflavone enriched bread in men with prostate cancer is associated with reduced proinflammatory cytokines and immunosuppressive cells. Cancer Prev Res (Phila) 8 , 1036 1044 ( 2015) DOI: 10.1158/1940-6207.CAPR-14-0464
[65]
Calero , C.I. , Beltrán González , A.N. , Gasulla , J. , Alvarez , S. , Evelson , P. , and Calvo , D.J. Quercetin antagonism of GABAAρ₁ receptors is prevented by ascorbic acid through a redox-independent mechanism. Eur J Pharmacol 714 ( 1-3 ), 274 280 ( 2013) DOI: 10.1016/j.ejphar.2013.07.044
[66]
Baumann , S. , Fas , S.C. , Giaisi , M. , Muller , W.W. , Merling , A. , Gulow , K. , Edler , L. , Krammer , P.H. , and Li-Weber , M. Wogonin preferentially kills malignant lymphocytes and suppresses T-cell tumor growth by inducing PLCgamma1- and Ca2+-dependent apoptosis. Blood 111 , 2354 2363 ( 2008) DOI: 10.1182/blood-2007-06-096198
[67]
Neuzil , J. , Dong , L.F. , Rohlena , J. , Truksa , J. , and Ralph , S.J. Classification of mitocans, anti-cancer drugs acting on mitochondria. Mitochondrion 13 , 199 208 ( 2013) DOI: 10.1016/j.mito.2012.07.112
[68]
Gorlach , S. , Fichna , J. , and Lewandowska , U. Polyphenols as mitochondria-targeted drugs. Cancer Lett 366 , 141 149 ( 2015) DOI: 10.1016/j.canlet.2015.07.004
[69]
Pathak , N. , Khan , S. , Bhargava , A. , Raghuram , G.V. , Jain , D. , Panwar , H. , Samarth , R.M. , Jain , S.K. , Maudar , K.K. , Mishra , D.K. , and Mishra , P.K. Cancer chemopreventive effects of the flavonoid-rich fraction isolated from papaya seeds. Nutr Cancer 66 , 857 871 ( 2014) DOI: 10.1080/01635581.2014.904912
[70]
Yans , A. , Shahamati , S.Z. , Maghsoudi , A.H. , Maghsoudi , N. Digitoflavone provokes mitochondrial biogenesis in PC12 cells: A protective approach to oxidative stress. Pharm Biol 53 ( 12 ), 1727 1734 ( 2015) DOI: 10.3109/13880209.2015.1005749
[71]
Moreno- Ulloa , A. , Miranda-Cervantes , A. , Licea-Navarro , A. , Mansour , C. , Beltrán-Partida , E. , Donis-Maturano , L. , Delgado De la Herrán , H.C. , Villarreal , F. , Álvarez-Delgado , C. (-)-Epicatechin stimulates mitochondrial biogenesis and cell growth in C2C12 myotubes via the G-protein coupled estrogen receptor. Eur J Pharmacol 822 , 95 107 ( 2018) DOI: 10.1016/j.ejphar.2018.01.014
[72]
Shanmugam , K. , Ravindran , S. , Kurian , G.A. , Rajesh , M. Fisetin confers cardioprotection against myocardial ischemia reperfusion injury by suppressing mitochondrial oxidative stress and mitochondrial dysfunction and inhibiting glycogen synthase kinase 3 β activity. Oxid Med Cell Longev 2018, 9173436 ( 2018) DOI: 10.1155/2018/9173436
[73]
Qiu , L. , Luo , Y. , Chen , X. Quercetin attenufates mitochondrial dysfunction and biogenesis via upregulated AMPK/SIRT1 signaling pathway in OA rats. Biomed Pharmacother 103 , 1585 1591 ( 2018) DOI: 10.1016/j.biopha.2018.05.003
[74]
Bunkar , N. , Bhargava , A. , Khare , N.K. , and Mishra , P.K. Mitochondrial anomalies: driver to age associated degenerative human ailments. Front Biosci (Landmark Ed) 21 , 769 793 ( 2016) DOI: 10.2741/4420
[75]
Rahnasto-Rilla , M. , Tyni , J. , Huovinen , M. , Jarho , E. , Kulikowicz , T. , Ravichandran , S.A. , Bohr , V. , Ferrucci , L. , Lahtela-Kakkonen , M. , Moaddel , R. Natural polyphenols as sirtuin 6 modulators. Sci Rep 8 ( 1 ), 4163 ( 2018) DOI: 10.1038/s41598-018-22388-5
[76]
Das , S. , Das , J. , Samadder , A. , Paul , A. , and Khuda-Bukhsh , A.R. Strategic formulation of apigenin-loaded PLGA nanoparticles for intracellular trafficking, DNA targeting and improved therapeutic effects in skin melanoma in vitro. Toxicol Lett 223 , 124 138 ( 2013)
[77]
Bhardwaj , V. , Tadinada , S.M. , Jain , A. , Sehdev , V. , Daniels , C.K. , Lai , J.C. , and Bhushan , A. Biochanin A reduces pancreatic cancer survival and progression. Anticancer Drugs 25 , 296 302 ( 2014) DOI: 10.1097/CAD.0000000000000044
[78]
Zaric , M. , Mitrovic , M. , Nikolic , I. , Baskic , D. , Popovic , S. , Djurdjevic , P. , Milosavljevic , Z. , and Zelen , I. Chrysin induces apoptosis in peripheral blood lymphocytes isolated from human chronic lymphocytic leukemia. Anticancer Agents Med Chem 15 , 189 195 ( 2015) DOI: 10.2174/1871520614666140924123116
[79]
Jiang , K. , Wang , W. , Jin , X. , Wang , Z. , Ji , Z. , and Meng , G. Silibinin, a natural flavonoid, induces autophagy via ROS-dependent mitochondrial dysfunction and loss of ATP involving BNIP3 in human MCF7 breast cancer cells. Oncol Rep 33 , 2711 2718 ( 2015) DOI: 10.3892/or.2015.3915
[80]
Seydi , E. , Rasekh , H.R. , Salimi , A. , Mohsenifar , Z. , and Pourahmad , J. Selective Toxicity of Apigenin on Cancerous Hepatocytes by Directly Targeting their Mitochondria. Anticancer Agents Med Chem 16 , 1576 1586 ( 2016) DOI: 10.2174/1871520616666160425110839
[81]
Mu , J. , Liu , T. , Jiang , L. , Wu , X. , Cao , Y. , Li , M. , Dong , Q. , Liu , Y. , and Xu , H. The traditional chinese medicine baicalein potently inhibits gastric cancer cells. J Cancer 7 , 453 461 ( 2016) DOI: 10.7150/jca.13548
[82]
Pham , J. , Grundmann , O. , and Elbayoumi , T. Mitochondriotropic nanoemulsified genistein-loaded vehicles for cancer therapy. Methods Mol Biol 1265 , 85 101 ( 2015) DOI: 10.1007/978-1-4939-2288-8_7
[83]
Amawi , H. , Ashby , C.R. , Jr. , and Tiwari , A.K. Cancer chemoprevention through dietary flavonoids: what’s limiting? Chin J Cancer 36 , 50 ( 2017) DOI: 10.1186/s40880-017-0217-4
[84]
Gao , S. , and Hu , M. Bioavailability challenges associated with development of anti-cancer phenolics. Mini Rev Med Chem 10 , 550 567 ( 2010) DOI: 10.2174/138955710791384081
[85]
Peng , J. , Fan , G. , Chai , Y. , and Wu , Y. Efficient new method for extraction and isolation of three flavonoids from Patrinia villosa Juss. by supercritical fluid extraction and high-speed counter-current chromatography. J Chromatogr A 1102 , 44 50 ( 2006) DOI: 10.1016/j.chroma.2005.10.045
[86]
Hartonen , K. , Parshintsev , J. , Sandberg , K. , Bergelin , E. , Nisula , L. , and Riekkola , M.L. Isolation of flavonoids from aspen knotwood by pressurized hot water extraction and comparison with other extraction techniques. Talanta 74 , 32 38 . ( 2007) DOI: 10.1016/j.talanta.2007.05.040
[87]
Xiao , W. , Han , L. , and Shi , B. Microwave-assisted extraction of flavonoids from Radix Astragali. Sep Purif Technol 62 , 614 618 ( 2008) DOI: 10.1016/j.seppur.2008.03.025
[88]
Duan , M.-H. , Luo , M. , Zhao , C.-J. , Wang , W. , Zu , Y.-G. , Zhang , D.-Y. , Yao , X.-H. , and Fu , Y.-J. Ionic liquid-based negative pressure cavitation-assisted extraction of three main flavonoids from the pigeonpea roots and its pilot-scale application. Sep Purif Technol 107 , 26 36 ( 2013) DOI: 10.1016/j.seppur.2013.01.003
[89]
J. , Cao , F. , Su , E. , Wu , C. , Zhao , L. , and Ying , R. Improving flavonoid extraction from Ginkgo biloba leaves by prefermentation processing. J Agric Food Chem 61 , 5783 5791 ( 2013) DOI: 10.1021/jf400712n
[90]
Zhang , H.-F. , Zhang , X. , Yang , X.-H. , Qiu , N.-X. , Wang , Y. , and Wang , Z.-Z. Microwave assisted extraction of flavonoids from cultivated Epimedium sagittatum: Extraction yield and mechanism, antioxidant activity and chemical composition. Ind Crop Prod 50 , 857 865 ( 2013) DOI: 10.1016/j.indcrop.2013.08.017
[91]
Qi , X.-L. , Peng , X. , Huang , Y.-Y. , Li , L. , Wei , Z.-F. , Zu , Y.-G. , and Fu , Y.-J. Green and efficient extraction of bioactive flavonoids from Equisetum palustre L. by deep eutectic solvents-based negative pressure cavitation method combined with macroporous resin enrichment. Ind Crop Prod 70 , 142 148 ( 2015) DOI: 10.1016/j.indcrop.2015.03.026
[92]
Wei , Z.-F. , Wang , X.-Q. , Peng , X. , Wang , W. , Zhao , C.-J. , Zu , Y.-G. , and Fu , Y.-J. Fast and green extraction and separation of main bioactive flavonoids from Radix Scutellariae. Ind Crop Prod 63 , 175 181 ( 2015) DOI: 10.1016/j.indcrop.2014.10.013
[93]
Gu , H. , Chen , F. , Zhang , Q. , and Zang , J. Application of ionic liquids in vacuum microwave-assisted extraction followed by macroporous resin isolation of three flavonoids rutin, hyperoside and hesperidin from Sorbus tianschanica leaves. J Chromatogr B Analyt Technol Biomed Life Sci 1014 , 45 55 ( 2016) DOI: 10.1016/j.jchromb.2016.01.045
[94]
Olthof , M.R. , Hollman , P.C. , Vree , T.B. , and Katan , M.B. Bioavailabilities of quercetin-3-glucoside and quercetin-4’-glucoside do not differ in humans. J Nutr 130 , 1200 1203 ( 2000) DOI: 10.1093/jn/130.5.1200
[95]
Thilakarathna , S.H. , and Rupasinghe , H.P. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients 5 , 3367 3387 ( 2013) DOI: 10.3390/nu5093367
[96]
Shoba , G. , Joy , D. , Joseph , T. , Majeed , M. , Rajendran , R. , and Srinivas , P.S. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med 64 , 353 356 ( 1998) DOI: 10.1055/s-2006-957450
[97]
Vaidyanathan , J.B. , and Walle , T. Cellular uptake and efflux of the tea flavonoid (-)epicatechin-3-gallate in the human intestinal cell line Caco-2. J Pharmacol Exp Ther 307 , 745 752 ( 2003) DOI: 10.1124/jpet.103.054296
[98]
Bilia , A.R. , Isacchi , B. , Righeschi , C. , Guccione , C. , and Bergonzi , M.C. Flavonoids loaded in nanocarriers: an opportunity to increase oral bioavailability and bioefficacy. Food Nutri Sci 5 , 1212 ( 2014) DOI: 10.4236/fns.2014.513132
[99]
Sandhu , P.S. , Kumar , R. , Beg , S. , Jain , S. , Kushwah , V. , Katare , O.P. , Singh , B. Natural lipids enriched self-nano-emulsifying systems for effective co-delivery of tamoxifen and naringenin: Systematic approach for improved breast cancer therapeutics. Nanomedicine 13 , 1703 1713 ( 2017) DOI: 10.1016/j.nano.2017.03.003
[100]
Ragelle , H. , Crauste-Manciet , S. , Seguin , J. , Brossard , D. , Scherman , D. , Arnaud , P. , Chabot , G.G. Nanoemulsion formulation of fisetin improves bioavailability and antitumour activity in mice. Int J Pharm 427 , 452 459 ( 2012) DOI: 10.1016/j.ijpharm.2012.02.025
[101]
Brinkman , A.M. , Chen , G. , Wang , Y. , Hedman , C.J. , Sherer , N.M. , Havighurst , T.C. , Gong , S. , Xu , W. Aminoflavone-loaded EGFR-targeted unimolecular micelle nanoparticles exhibit anti-cancer effects in triple negative breast cancer. Biomaterials 101 , 20 31 ( 2016) DOI: 10.1016/j.biomaterials.2016.05.041
[102]
Yazdi Rouholamini , S.E. , Moghassemi , S. , Maharat , Z. , Hakamivala , A. , Kashanian , S. , Omidfar , K. Effect of silibinin-loaded nano-niosomal coated with trimethyl chitosan on miRNAs expression in 2D and 3D models of T47D breast cancer cell line. Artif Cells Nanomed Biotechnol 46 , 524 535 ( 2018) DOI: 10.1080/21691401.2017.1326928
[103]
Kadari , A. , Gudem , S. , Kulhari , H. , Bhandi , M.M. , Borkar , R.M. , Kolapalli , V.R. , Sistla , R. Enhanced oral bioavailability and anticancer efficacy of fisetin by encapsulating as inclusion complex with HPbetaCD in polymeric nanoparticles. Drug Deliv 24 , 224 232 ( 2017) DOI: 10.1080/10717544.2016.1245366
[104]
Wu , S. , Huang , J. , Hui , K. , Yue , Y. , Gu , Y. , Ning , Z. , Wang , X. , He , D. , and Wu , K. 2’Hydroxyflavanone inhibits epithelialmesenchymal transition, and cell migration and invasion via suppression of the Wnt/betacatenin signaling pathway in prostate cancer. Oncol Rep 40 , 2836 2843 ( 2018) DOI: 10.3892/or.2018.6678
[105]
Chaurasia , S. , Patel , R.R. , Vure , P. , Mishra , B. Oral naringenin nanocarriers: Fabrication, optimization, pharmacokinetic and chemotherapeutic efficacy assessments. Nanomedicine (Lond) 12 , 1243 1260 ( 2017) DOI: 10.2217/nnm-2016-0436
[106]
Chaurasia , S. , Patel , R.R. , Vure , P. , Mishra , B. Potential of cationic-polymeric nanoparticles for oral delivery of naringenin: in vitro and in vivo investigations. J Pharm Sci 107 , 706 716 ( 2018) DOI: 10.1016/j.xphs.2017.10.006
[107]
Perez-Ruiz , A.G. , Ganem , A. , Olivares-Corichi , I.M. , García-Sánchez , J.R. Lecithin–chitosan–TPGS nanoparticles as nanocarriers of (−)-epicatechin enhanced its anticancer activity in breast cancer cells. RSC Advances 8 , 34773 34782 ( 2018) DOI: 10.1039/C8RA06327C
[108]
Thanki , K. , Gangwal , R.P. , Sangamwar , A.T. , and Jain , S. Oral delivery of anticancer drugs: challenges and opportunities. J Control Release 170 , 15 40 ( 2013) DOI: 10.1016/j.jconrel.2013.04.020
[109]
Arya , A. , Khandelwal , K. , Ahmad , H. , Laxman , T.S. , Sharma , K. , Mittapelly , N. , Agrawal , S. , Bhatta , R.S. , and Dwivedi , A.K. Co-delivery of hesperetin enhanced bicalutamide induced apoptosis by exploiting mitochondrial membrane potential via polymeric nanoparticles in a PC-3 cell line. RSC Advances 6 , 5925 5935 ( 2016) DOI: 10.1039/C5RA23067E
[110]
Ghadi , R. , and Dand , N. BCS class IV drugs: highly notorious candidates for formulation development. J Control Release 248 , 71 95 ( 2017) DOI: 10.1016/j.jconrel.2017.01.014
[111]
Dong , X. , Ke , X. , and Liao , Z. The microstructure characterization of meloxicam microemulsion and its influence on the solubilization capacity. Drug Dev Ind Pharm 37 , 894 900 ( 2011) DOI: 10.3109/03639045.2010.548067
[112]
El-Haddad , A.E. , Sheta , N.M. , Boshra , S.A. Isolation, formulation, and efficacy enhancement of morin emulsified carriers against lung toxicity in rats. AAPS PharmSciTech 19 , 2346 2357 ( 2018) DOI: 10.1208/s12249-018-1072-6
[113]
Ban , C. , Park , S.J. , Lim , S. , Choi , S.J. , and Choi , Y.J. Improving flavonoid bioaccessibility using an edible oil-based lipid nanoparticle for oral delivery. J Agric Food Chem 63 ( 21 ), 5266 5272 ( 2015) DOI: 10.1021/acs.jafc.5b01495
[114]
Tsai , M.J. , Wu , P.C. , Huang , Y.B. , Chang , J.S. , Lin , C.L. , Tsai , Y.H. , Fang , J.Y. Baicalein loaded in tocol nanostructured lipid carriers (tocol NLCs) for enhanced stability and brain targeting. Int J Pharm 423 , 461 470 ( 2012) DOI: 10.1016/j.ijpharm.2011.12.009
[115]
Sun , M. , Wang , S. , Nie , S. , Zhang , J. Enhanced oral bioavailability of quercetin by nanostructured lipid carriers (1044.24). The FASEB J 28 , 1044 1024 ( 2014) DOI: 10.1096/fasebj.28.1_supplement.1044.24
[116]
Zhang , S. , Zhang , H. , Xu , Z. , Wu , M. , Xia , W. , and Zhang , W. Chimonanthus praecox extract/cyclodextrin inclusion complexes: Selective inclusion, enhancement of antioxidant activity and thermal stability. Ind Crop Prod 95 , 60 65 ( 2017) DOI: 10.1016/j.indcrop.2017.08.009
[117]
Bungaruang , L. , Gutmann , A. , and Nidetzky , B. β -Cyclodextrin improves solubility and enzymatic c-glucosylation of the flavonoid phloretin. Advanced Synthesis & Catalysis 358 , 486 493 ( 2015) DOI: 10.1002/adsc.201500838
[118]
Carneiro , S.B. , Costa Duarte , F.Í. , Heimfarth , L. , Siqueira Quintans , J.S. , Quintans-Júnior , L.J. , Veiga Júnior , V.F.D. , Neves de Lima , Á.A. Cyclodextrin-Drug Inclusion Complexes: In vivo and In vitro Approaches. Int J Mol Sci. 20 ( 3 ). pii: E642 ( 2019) DOI: 10.3390/ijms20030642
[119]
Jain , D. Phytosome: A novel drug delivery system for herbal medicine. Int J Pharm Sci Res 2 , 224 228 ( 2010)
[120]
Karthivashan , G. , Masarudin , M.J. , Kura , A.U. , Abas , F. , Fakurazi , S. Optimization, formulation, and characterization of multiflavonoids-loaded flavanosome by bulk or sequential technique. Int J Nanomedicine 11 , 3417 3434 ( 2016) DOI: 10.2147/IJN.S112045
[121]
Telange , D.R. , Patil , A.T. , Pethe , A.M. , Fegade , H. , Anand , S. , Dave , V.S. Formulation and characterization of an apigenin-phospholipid phytosome (APLC) for improved solubility, in vivo bioavailability, and antioxidant potential. Eur J Pharm Sci 108 , 36 49 ( 2017) DOI: 10.1016/j.ejps.2016.12.009
[122]
Riva , A. , Ronchi , M. , Petrangolini , G. , Bosisio , S. , Allegrini , P. Improved Oral Absorption of Quercetin from Quercetin Phytosome®, a New Delivery System Based on Food Grade Lecithin. Eur J Drug Metab Pharmacokinet ( 2018) DOI: 10.1007/s13318-018-0517-3
[123]
Zhang , X.Y. , and Zhang , P.Y. Mitochondria targeting nano agents in cancer therapeutics. Oncol Lett 12 , 4887 4890 ( 2016) DOI: 10.3892/ol.2016.5302
[124]
D’souza , G.G.M. , Wagle , M.A. , Saxena , V. , and Shah , A. Approaches for targeting mitochondria in cancer therapy. Biochimica et Biophysica Acta (BBA)-Bioenergetics 1807 , 689 696 ( 2011) DOI: 10.1016/j.bbabio.2010.08.008
[125]
Durazo , S.A. , and Kompella , U.B. Functionalized nanosystems for targeted mitochondrial delivery. Mitochondrion 12 , 190 201 ( 2012) DOI: 10.1016/j.mito.2011.11.001
[126]
Zupancic , S. , Kocbek , P. , Zariwala , M.G. , Renshaw , D. , Gul , M.O. , Elsaid , Z. , Taylor , K.M. , and Somavarapu , S. Design and development of novel mitochondrial targeted nanocarriers, DQAsomes for curcumin inhalation. Mol Pharm 11 , 2334 2345 ( 2014) DOI: 10.1021/mp500003q
[127]
Yamada , Y. , and Harashima , H. Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliv Rev 60 , 1439 1462 ( 2008) DOI: 10.1016/j.addr.2008.04.016
[128]
Yamada , Y. , Furukawa , R. , Yasuzaki , Y. , and Harashima , H. Dual function MITO-Porter, a nano carrier integrating both efficient cytoplasmic delivery and mitochondrial macromolecule delivery. Mol Ther 19 , 1449 1456 ( 2011) DOI: 10.1038/mt.2011.99
[129]
Lu , P. , Bruno , B.J. , Rabenau , M. , and Lim , C.S. Delivery of drugs and macromolecules to the mitochondria for cancer therapy. J Control Release 240 , 38 51 ( 2016) DOI: 10.1016/j.jconrel.2015.10.023
[130]
Farsinejad , S. , Gheisary , Z. , Ebrahimi Samani , S. , Alizadeh , A.M. Mitochondrial targeted peptides for cancer therapy. Tumour Biol 36 , 5715 5725 ( 2015) DOI: 10.1007/s13277-015-3719-1
[131]
Garcia , G. , Atilhan , M. , Aparicio , S. Flavonol-carbon nanostructure hybrid systems: a DFT study on the interaction mechanism and UV/Vis features. Phys Chem Chem Phys 18 , 4760 4771 ( 2016) DOI: 10.1039/c5cp07629c
[132]
Yue , C. , Yang , Y. , Zhang , C. , Alfranca , G. , Cheng , S. , Ma , L. , Liu , Y. , Zhi , X. , Ni , J. , Jiang , W. , Song , J. , De La Fuente , J.M. , and Cui , D. ROS-Responsive Mitochondria-Targeting blended nanoparticles: chemo- and photodynamic synergistic therapy for lung cancer with on-demand drug release upon irradiation with a single light source. Theranostics 6 , 2352 2366 ( 2016) DOI: 10.7150/thno.15433
[133]
Keelan , J.A. Nanotoxicology: nanoparticles versus the placenta. Nat Nanotechnol 6 , 263 264 ( 2011) DOI: 10.1038/nnano.2011.65
[134]
Rutberg , F.G. , Dubina , M.V. , Kolikov , V.A. , Moiseenko , F.V. , Ignat’eva , E.V. , Volkov , N.M. , Snetov , V.N. , Stogov , A.Y. Effect of silver oxide nanoparticles on tumor growth in vivo. Dokl Biochem Biophys 421 , 191 193 ( 2008)
[135]
Sharma , A. , Madhunapantula , S.V. , Robertson , G.P. Toxicological considerations when creating nanoparticle-based drugs and drug delivery systems. Expert Opin Drug Metab Toxicol 8 , 47 69 ( 2012)
[136]
Slowing, Ii, Vivero-Escoto , J.L. , Wu , C.W. , and Lin , V.S. Mesoporous silica nanoparticles as controlled release drug delivery and gene transfection carriers. Adv Drug Deliv Rev 60 , 1278 1288 ( 2008) DOI: 10.1016/j.addr.2008.03.012
[137]
Sarkar , A. , Ghosh , S. , Chowdhury , S. , Pandey , B. , and Sil , P.C. Targeted delivery of quercetin loaded mesoporous silica nanoparticles to the breast cancer cells. Biochim Biophys Acta 1860 , 2065 2075 ( 2016) DOI: 10.1016/j.bbagen.2016.07.001
[138]
Fang , J. , Zhang , S. , Xue , X. , Zhu , X. , Song , S. , Wang , B. , Jiang , L. , Qin , M. , Liang , H. , Gao , L. Quercetin and doxorubicin co-delivery using mesoporous silica nanoparticles enhance the efficacy of gastric carcinoma chemotherapy. Int J Nanomedicine 13 , 5113 5126 ( 2018) DOI: 10.2147/IJN.S170862
[139]
Dai , G. , Zheng , D. , Wang , Q. , Yang , J. , Liu , G. , Song , Q. , Sun , X. , Tao , C. , Hu , Q. , Gao , T. , Yu , L. , Guo , W. Baicalein inhibits progression of osteosarcoma cells through inactivation of the Wnt/beta-catenin signaling pathway. Oncotarget 8 , 86098 86116 ( 2017) DOI: 10.18632/oncotarget.20987
[140]
Zhang , S. , Yang , Y. , Liang , Z. , Duan , W. , Yang , J. , Yan , J. , Wang , N. , Feng , W. , Ding , M. , Nie , Y. , and Jin , Z. Silybin-Mediated inhibition of notch signaling exerts antitumor activity in human hepatocellular carcinoma cells. PLoS One 8 , e83699 ( 2013) DOI: 10.1371/journal.pone.0083699
[141]
Lim , K.J. , Bisht , S. , Bar , E.E. , Maitra , A. , Eberhart , C.G. A polymeric nanoparticle formulation of curcumin inhibits growth, clonogenicity and stem-like fraction in malignant brain tumors. Cancer Biol Ther 11 , 464 473 ( 2011) DOI: 10.4161/cbt.11.5.14410
[142]
Mohammadian , F. , Abhari , A. , Dariushnejad , H. , Nikanfar , A. , Pilehvar-Soltanahmadi , Y. , Zarghami , N. Effects of Chrysin-PLGA-PEG Nanoparticles on Proliferation and Gene Expression of miRNAs in Gastric Cancer Cell Line. Int J Cancer Manag 9 , e4190 ( 2016) DOI: 10.17795/ijcp-4190
[143]
Lv , L. , Liu , C. , Chen , C. , Yu , X. , Chen , G. , Shi , Y. , Qin , F. , Ou , J. , Qiu , K. , Li , G. Quercetin and doxorubicin co-encapsulated biotin receptor-targeting nanoparticles for minimizing drug resistance in breast cancer. Oncotarget 7 , 32184 32199 ( 2016) DOI: 10.18632/oncotarget.8607
[144]
Gawde , K.A. , Sau , S. , Tatiparti , K. , Kashaw , S.K. , Mehrmohammadi , M. , Azmi , A.S. , Iyer , A.K. Paclitaxel and di-fluorinated curcumin loaded in albumin nanoparticles for targeted synergistic combination therapy of ovarian and cervical cancers. Colloids Surf B Biointerfaces 167 , 8 19 ( 2018) DOI: 10.1016/j.colsurfb.2018.03.046
[145]
Butow , R.A. , and Avadhani , N.G. Mitochondrial signaling: the retrograde response. Mol Cell 14 , 1 15 ( 2004) DOI: 10.1016/S1097-2765(04)00179-0
[146]
Passos , J.F. , Saretzki , G. , Ahmed , S. , Nelson , G. , Richter , T. , Peters , H. , Wappler , I. , Birket , M.J. , Harold , G. , Schaeuble , K. , Birch-Machin , M.A. , Kirkwood , T.B. , and Von Zglinicki , T. Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol 5 , e110 ( 2007) DOI: 10.1371/journal.pbio.0050110
[147]
Hussain , A.R. , Khan , A.S. , Ahmed , S.O. , Ahmed , M. , Platanias , L.C. , Al-Kuraya , K.S. , and Uddin , S. Apigenin induces apoptosis via downregulation of S-phase kinase-associated protein 2-mediated induction of p27Kip1 in primary effusion lymphoma cells. Cell Prolif 43 , 170 183 ( 2010) DOI: 10.1111/j.1365-2184.2009.00662.x
[148]
Zhu , Y. , Mao , Y. , Chen , H. , Lin , Y. , Hu , Z. , Wu , J. , Xu , X. , Xu , X. , Qin , J. , and Xie , L. Apigenin promotes apoptosis, inhibits invasion and induces cell cycle arrest of T24 human bladder cancer cells. Cancer Cell Int 13 , 54 ( 2013) DOI: 10.1186/1475-2867-13-54
[149]
Seo , H.S. , Jo , J.K. , Ku , J.M. , Choi , H.S. , Choi , Y.K. , Woo , J.K. , Kim , H.I. , Kang , S.Y. , Lee , K.M. , Nam , K.W. , Park , N. , Jang , B.H. , Shin , Y.C. , and Ko , S.G. Induction of caspase-dependent extrinsic apoptosis by apigenin through inhibition of signal transducer and activator of transcription 3 (STAT3) signalling in HER2-overexpressing BT-474 breast cancer cells. Biosci Rep 35 ( 2015) DOI: 10.1042/BSR20150165
[150]
Souza , R.P. , Bonfim-Mendonca , P.S. , Gimenes , F. , Ratti , B.A. , Kaplum , V. , Bruschi , M.L. , Nakamura , C.V. , Silva , S.O. , Maria-Engler , S.S. , and Consolaro , M.E. Oxidative Stress Triggered by Apigenin Induces Apoptosis in a Comprehensive Panel of Human Cervical Cancer-Derived Cell Lines. Oxid Med Cell Longev 2017 , 1512745 ( 2017) DOI: 10.1155/2017/1512745
[151]
Hashemi , M. , Nouri Long , M. , Entezari , M. , Nafisi , S. , and Nowroozii , H. Anti-mutagenic and pro-apoptotic effects of apigenin on human chronic lymphocytic leukemia cells. Acta Med Iran 48 , 283 288 ( 2010)
[152]
Lee , Y. , Sung , B. , Kang , Y.J. , Kim , D.H. , Jang , J.Y. , Hwang , S.Y. , Kim , M. , Lim , H.S. , Yoon , J.H. , Chung , H.Y. , and Kim , N.D. Apigenin-induced apoptosis is enhanced by inhibition of autophagy formation in HCT116 human colon cancer cells. Int J Oncol 44 , 1599 1606 ( 2014) DOI: 10.3892/ijo.2014.2339
[153]
Xu , M. , Wang , S. , Song , Y.U. , Yao , J. , Huang , K. , and Zhu , X. Apigenin suppresses colorectal cancer cell proliferation, migration and invasion via inhibition of the Wnt/beta-catenin signaling pathway. Oncol Lett 11 , 3075 3080 ( 2016) DOI: 10.3892/ol.2016.4331
[154]
Zhu , H. , Jin , H. , Pi , J. , Bai , H. , Yang , F. , Wu , C. , Jiang , J. , and Cai , J. Apigenin induced apoptosis in esophageal carcinoma cells by destruction membrane structures. Scanning 38 , 322 328 ( 2016) DOI: 10.1002/sca.21273
[155]
Chen , J. , Chen , J. , Li , Z. , Liu , C. , and Yin , L. The apoptotic effect of apigenin on human gastric carcinoma cells through mitochondrial signal pathway. Tumour Biol 35 , 7719 7726 ( 2014) DOI: 10.1007/s13277-014-2014-x
[156]
Stump , T.A. , Santee , B.N. , Williams , L.P. , Kunze , R.A. , Heinze , C.E. , Huseman , E.D. , Gryka , R.J. , Simpson , D.S. , and Amos , S. The antiproliferative and apoptotic effects of apigenin on glioblastoma cells. J Pharm Pharmacol 69 , 907 916 ( 2017) DOI: 10.1111/jphp.12718
[157]
Wan , Y. , Fei , X. , Wang , Z. , Jiang , D. , Chen , H. , Wang , M. , Zhou , S. miR-423-5p knockdown enhances the sensitivity of glioma stem cells to apigenin through the mitochondrial pathway. Tumour Biology 39 ( 4 ), 1010428317695526 ( 2011) DOI: 10.1177/1010428317695526
[158]
Qin , Y. , Zhao , D. , Zhou , H.G. , Wang , X.H. , Zhong , W.L. , Chen , S. , Gu , W.G. , Wang , W. , Zhang , C.H. , Liu , Y.R. , Liu , H.J. , Zhang , Q. , Guo , Y.Q. , Sun , T. , and Yang , C. Apigenin inhibits NF-kappaB and snail signaling, EMT and metastasis in human hepatocellular carcinoma. Oncotarget 7 , 41421 41431 ( 2016) DOI: 10.18632/oncotarget.9404
[159]
Isoda , H. , Motojima , H. , Onaga , S. , Samet , I. , Villareal , M.O. , and Han , J. Analysis of the erythroid differentiation effect of flavonoid apigenin on K562 human chronic leukemia cells. Chem Biol Interact 220 , 269 277 ( 2014) DOI: 10.1016/j.cbi.2014.07.006
[160]
Lee , Y.M. , Lee , G. , Oh , T.I. , Kim , B.M. , Shim , D.W. , Lee , K.H. , Kim , Y.J. , Lim , B.O. , and Lim , J.H. Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress. Int J Oncol 48 , 399 408 ( 2016)
[161]
Chakrabarti , M. , Banik , N.L. , and Ray , S.K. Sequential hTERT knockdown and apigenin treatment inhibited invasion and proliferation and induced apoptosis in human malignant neuroblastoma SK-N-DZ and SK-N-BE2 cells. J Mol Neurosci 51 , 187 198 ( 2013) DOI: 10.1007/s12031-013-9975-x
[162]
Maggioni , D. , Garavello , W. , Rigolio , R. , Pignataro , L. , Gaini , R. , and Nicolini , G. Apigenin impairs oral squamous cell carcinoma growth in vitro inducing cell cycle arrest and apoptosis. Int J Oncol 43 , 1675 1682 ( 2013) DOI: 10.3892/ijo.2013.2072
[163]
Liu , X. , Li , L. , Lv , L. , Chen , D. , Shen , L. , Xie , Z. Apigenin inhibits the proliferation and invasion of osteosarcoma cells by suppressing the Wnt/β-catenin signaling pathway. Oncology Rep 34 ( 2 ), 1035 1041 ( 2015) DOI: 10.3892/or.2015.4022
[164]
Lin , T.H. , Hsu , W.H. , Tsai , P.H. , Huang , Y.T. , Lin , C.W. , Chen , K.C. , Tsai , I.H. , Kandaswami , C.C. , Huang , C.J. , Chang , G.D. , Lee , M.T. , and Cheng , C.H. Dietary flavonoids, luteolin and quercetin, inhibit invasion of cervical cancer by reduction of UBE2S through epithelial-mesenchymal transition signaling. Food Funct 8 , 1558 1568 ( 2017) DOI: 10.1039/C6FO00551A
[165]
Suh , Y.A. , Jo , S.Y. , Lee , H.Y. , and Lee , C. Inhibition of IL-6/STAT3 axis and targeting Axl and Tyro3 receptor tyrosine kinases by apigenin circumvent taxol resistance in ovarian cancer cells. Int J Oncol 46 , 1405 1411 ( 2015) DOI: 10.3892/ijo.2014.2808
[166]
Wu , D.G. , Yu , P. , Li , J.W. , Jiang , P. , Sun , J. , Wang , H.Z. , Zhang , L.D. , Wen , M.B. , and Bie , P. Apigenin potentiates the growth inhibitory effects by IKK-beta-mediated NF-kappaB activation in pancreatic cancer cells. Toxicol Lett 224 , 157 164 ( 2014) DOI: 10.1016/j.toxlet.2013.10.007
[167]
Shukla , S. , Kanwal , R. , Shankar , E. , Datt , M. , Chance , M.R. , Fu , P. , Maclennan , G.T. , and Gupta , S. Apigenin blocks IKKalpha activation and suppresses prostate cancer progression. Oncotarget 6 , 31216 31232 ( 2015) DOI: 10.18632/oncotarget.5157
[168]
Das , S. , Das , J. , Samadder , A. , Paul , A. , and Khuda-Bukhsh , A.R. Efficacy of PLGA-loaded apigenin nanoparticles in Benzo(a)pyrene and ultraviolet-B induced skin cancer of mice: mitochondria mediated apoptotic signalling cascades. Food Chem Toxicol 62 , 670 680 ( 2013) DOI: 10.1016/j.fct.2013.09.037
[169]
Kim , S.H. , Kang , J.G. , Kim , C.S. , Ihm , S.H. , Choi , M.G. , Yoo , H.J. , and Lee , S.J. Suppression of akt potentiates synergistic cytotoxicity of apigenin with trail in anaplastic thyroid carcinoma cells. Anticancer Res 35 , 6529 6537 ( 2015)
[170]
Shang , D. , Li , Z. , Zhu , Z. , Chen , H. , Zhao , L. , Wang , X. , and Chen , Y. Baicalein suppresses 17-beta-estradiol-induced migration, adhesion and invasion of breast cancer cells via the G protein-coupled receptor 30 signaling pathway. Oncol Rep 33 , 2077 2085 ( 2015) DOI: 10.3892/or.2015.3786
[171]
Ye , H. , Zhang , Y. , Wang , Y. , Xia , J. , Mao , X. , and Yu , X. The restraining effect of baicalein and U0126 on human cervical cancer cell line HeLa. Mol Med Rep 16 , 957 963 ( 2017) DOI: 10.3892/mmr.2017.6648
[172]
Havermann , S. , Chovolou , Y. , Humpf , H.U. , and Watjen , W. Modulation of the Nrf2 signalling pathway in Hct116 colon carcinoma cells by baicalein and its methylated derivative negletein. Pharm Biol 54 , 1491 1502 ( 2016) DOI: 10.3109/13880209.2015.1104703
[173]
Rui , X. , Yan , X.I. , and Zhang , K. Baicalein inhibits the migration and invasion of colorectal cancer cells via suppression of the AKT signaling pathway. Oncol Lett 11 , 685 688 ( 2016) DOI: 10.3892/ol.2015.3935
[174]
Chen , F. , Zhuang , M. , Zhong , C. , Peng , J. , Wang , X. , Li , J. , Chen , Z. , and Huang , Y. Baicalein reverses hypoxia-induced 5-FU resistance in gastric cancer AGS cells through suppression of glycolysis and the PTEN/Akt/HIF-1alpha signaling pathway. Oncol Rep 33 , 457 463 ( 2015) DOI: 10.3892/or.2014.3550
[175]
Zhang , Z. , Lv , J. , Lei , X. , Li , S. , Zhang , Y. , Meng , L. , Xue , R. , and Li , Z. Baicalein reduces the invasion of glioma cells via reducing the activity of p38 signaling pathway. PLoS One 9 , e90318 ( 2014) DOI: 10.1371/journal.pone.0090318
[176]
Chen , K. , Zhang , S. , Ji , Y. , Li , J. , An , P. , Ren , H. , Liang , R. , Yang , J. , and Li , Z. Baicalein inhibits the invasion and metastatic capabilities of hepatocellular carcinoma cells via down-regulation of the ERK pathway. PLoS One 8 , e72927 ( 2013) DOI: 10.1371/journal.pone.0072927
[177]
Li , J. , Duan , B. , Guo , Y. , Zhou , R. , Sun , J. , Bie , B. , Yang , S. , Huang , C. , Yang , J. , Li , Z. Baicalein sensitizes hepatocellular carcinoma cells to 5-FU and Epirubicin by activating apoptosis and ameliorating P-glycoprotein activity. Biomed Pharmacother 98 , 806 812 ( 2018) DOI: 10.1016/j.biopha.2018.01.002
[178]
Chen , Y.J. , Wu , C.S. , Shieh , J.J. , Wu , J.H. , Chen , H.Y. , Chung , T.W. , Chen , Y.K. , and Lin , C.C. Baicalein triggers mitochondria-mediated apoptosis and enhances the antileukemic effect of vincristine in childhood acute lymphoblastic leukemia ccrf-cem cells. Evid Based Complement Alternat Med 2013 , 124747 ( 2013) DOI: 10.1155/2013/124747
[179]
Cathcart , M.C. , Useckaite , Z. , Drakeford , C. , Semik , V. , Lysaght , J. , Gately , K. , O’byrne , K.J. , and Pidgeon , G.P. Anti-cancer effects of baicalein in non-small cell lung cancer in-vitro and in-vivo. BMC Cancer 16 , 707 ( 2016) DOI: 10.1186/s12885-016-2740-0
[180]
Choi , E.O. , Cho , E.J. , Jeong , J.W. , Park , C. , Hong , S.H. , Hwang , H.J. , Moon , S.K. , Son , C.G. , Kim , W.J. , and Choi , Y.H. Baicalein inhibits the migration and invasion of b16f10 mouse melanoma cells through inactivation of the pi3k/akt signaling pathway. Biomol Ther (Seoul) 25 , 213 221 ( 2017) DOI: 10.4062/biomolther.2016.094
[181]
He , N. , and Zhang , Z. Baicalein suppresses the viability of MG-63 osteosarcoma cells through inhibiting c-MYC expression via Wnt signaling pathway. Mol Cell Biochem 405 , 187 196 ( 2015) DOI: 10.1007/s11010-015-2410-6
[182]
Yan , H. , Xin , S. , Wang , H. , Ma , J. , Zhang , H. , and Wei , H. Baicalein inhibits MMP-2 expression in human ovarian cancer cells by suppressing the p38 MAPK-dependent NF-kappaB signaling pathway. Anticancer Drugs 26 , 649 656 ( 2015) DOI: 10.1097/CAD.0000000000000230
[183]
Donald , G. , Hertzer , K. , and Eibl , G. Baicalein--an intriguing therapeutic phytochemical in pancreatic cancer. Curr Drug Targets 13 , 1772 1776 ( 2012) DOI: 10.2174/138945012804545470
[184]
Guo , Z. , Hu , X. , Xing , Z. , Xing , R. , Lv , R. , Cheng , X. , Su , J. , Zhou , Z. , Xu , Z. , Nilsson , S. , and Liu , Z. Baicalein inhibits prostate cancer cell growth and metastasis via the caveolin-1/AKT/mTOR pathway. Mol Cell Biochem 406 , 111 119 ( 2015) DOI: 10.1007/s11010-015-2429-8
[185]
Wu , B. , Li , J. , Huang , D. , Wang , W. , Chen , Y. , Liao , Y. , Tang , X. , Xie , H. , and Tang , F. Baicalein mediates inhibition of migration and invasiveness of skin carcinoma through Ezrin in A431 cells. BMC Cancer 11 , 527 ( 2011) DOI: 10.1186/1471-2407-11-527
[186]
Ye , C. , Yu , X. , Zeng , J. , Dai , M. , and Zhang , B. Effects of baicalein on proliferation, apoptosis, migration and invasion of Ewing’s sarcoma cells. Int J Oncol 51 , 1785 1792 ( 2017) DOI: 10.3892/ijo.2017.4148
[187]
Huang , Y. , Hu , J. , Zheng , J. , Li , J. , Wei , T. , Zheng , Z. , and Chen , Y. Down-regulation of the PI3K/Akt signaling pathway and induction of apoptosis in CA46 Burkitt lymphoma cells by baicalin. J Exp Clin Cancer Res 31 , 48 ( 2012) DOI: 10.1186/1756-9966-31-48
[188]
Chung , H. , Choi , H.S. , Seo , E.K. , Kang , D.H. , and Oh , E.S. Baicalin and baicalein inhibit transforming growth factor-beta1-mediated epithelial-mesenchymal transition in human breast epithelial cells. Biochem Biophys Res Commun 458 , 707 713 ( 2015) DOI: 10.1016/j.bbrc.2015.02.032
[189]
Zhang , J. , Liu , S. , Xu , B. , Li , G. , Li , G. , Huang , A. , Wu , B. , Peng , L. , Song , M. , Xie , Q. , Lin , W. , Xie , W. , Wen , S. , Zhang , Z. , Xu , X. , and Liang , S. Study of baicalin on sympathoexcitation induced by myocardial ischemia via P2X3 receptor in superior cervical ganglia. Auton Neurosci 189 , 8 15 ( 2015) DOI: 10.1016/j.autneu.2014.12.001
[190]
Wang , C.Z. , Zhang , C.F. , Chen , L. , Anderson , S. , Lu , F. , and Yuan , C.S. Colon cancer chemopreventive effects of baicalein, an active enteric microbiome metabolite from baicalin. Int J Oncol 47 , 1749 1758 ( 2015) DOI: 10.3892/ijo.2015.3173
[191]
Yang , B.L. , Chen , H.J. , Chen , Y.G. , Gu , Y.F. , Zhang , S.P. , Lin , Q. , and Zhu , P. Effects of Baicalin on an orthotopic transplantation mouse model of mismatch repair gene deficient colorectal cancer. Zhonghua Wai Ke Za Zhi 50 , 843 847 ( 2012)
[192]
Kyo , R. , Nakahata , N. , Sakakibara , I. , Kubo , M. , and Ohizumi , Y. Baicalin and baicalein, constituents of an important medicinal plant, inhibit intracellular Ca2+ elevation by reducing phospholipase C activity in C6 rat glioma cells. J Pharm Pharmacol 50 , 1179 1182 ( 1998) DOI: 10.1111/j.2042-7158.1998.tb03331.x
[193]
Yu , Z. , Luo , X. , Wang , C. , Ye , J. , Liu , S. , Xie , L. , Wang , F. , and Bao , J. Baicalin promoted site-2 protease and not site-1 protease in endoplasmic reticulum stress-induced apoptosis of human hepatocellular carcinoma cells. FEBS Open Bio 6 , 1093 1101 ( 2016) DOI: 10.1002/2211-5463.12130
[194]
Ren , X. , Zhang , Y. , Li , C. , Wang , H. , Jiang , Z. , Zhang , Z. , Guo , Q. , Song , G. , Bi , K. , and Jiang , G. Enhancement of baicalin by hexamethylene bisacetamide on the induction of apoptosis contributes to simultaneous activation of the intrinsic and extrinsic apoptotic pathways in human leukemia cells. Oncol Rep 30 , 2071 2080 ( 2013) DOI: 10.3892/or.2013.2684
[195]
Du , G. , Han , G. , Zhang , S. , Lin , H. , Wu , X. , Wang , M. , Ji , L. , Lu , L. , Yu , L. , and Liang , W. Baicalin suppresses lung carcinoma and lung metastasis by SOD mimic and HIF-1alpha inhibition. Eur J Pharmacol 630 , 121 130 ( 2010) DOI: 10.1016/j.ejphar.2009.12.014
[196]
Wan , D. , and Ouyang , H. Baicalin induces apoptosis in human osteosarcoma cell through ROS-mediated mitochondrial pathway. Nat Prod Res, 1 5 . ( 2017) DOI: 10.1080/14786419.2017.1359173
[197]
Gao , C. , Zhou , Y. , Li , H. , Cong , X. , Jiang , Z. , Wang , X. , Cao , R. , and Tian , W. Antitumor effects of baicalin on ovarian cancer cells through induction of cell apoptosis and inhibition of cell migration in vitro. Mol Med Rep 16 , 8729 8734 ( 2017) DOI: 10.3892/mmr.2017.7757
[198]
Chen , S. , Ruan , Q. , Bedner , E. , Deptala , A. , Wang , X. , Hsieh , T.C. , Traganos , F. , and Darzynkiewicz , Z. Effects of the flavonoid baicalin and its metabolite baicalein on androgen receptor expression, cell cycle progression and apoptosis of prostate cancer cell lines. Cell Prolif 34 , 293 304 ( 2001) DOI: 10.1046/j.0960-7722.2001.00213.x
[199]
Huang , C. , Chen , Y.J. , Chen , W.J. , Lin , C.L. , Wei , Y.X. , and Huang , H.C. Combined treatment with chrysin and 1,2,3,4,6-penta-O-galloyl-beta-D-glucose synergistically inhibits LRP6 and Skp2 activation in triple-negative breast cancer and xenografts. Mol Carcinog 54 , 1613 1625 ( 2015) DOI: 10.1002/mc.22234
[200]
Laishram , S. , Moirangthem , D.S. , Borah , J.C. , Pal , B.C. , Suman , P. , Gupta , S.K. , Kalita , M.C. , and Talukdar , N.C. Chrysin rich Scutellaria discolor Colebr. induces cervical cancer cell death via the induction of cell cycle arrest and caspase-dependent apoptosis. Life Sci 143 , 105 113 ( 2015) DOI: 10.1016/j.lfs.2015.10.035
[201]
Sak , K. , Kasemaa , K. , and Everaus , H. Potentiation of luteolin cytotoxicity by flavonols fisetin and quercetin in human chronic lymphocytic leukemia cell lines. Food Funct 7 , 3815 3824 ( 2016) DOI: 10.1039/C6FO00583G
[202]
Leon , I.E. , Cadavid-Vargas , J.F. , Tiscornia , I. , Porro , V. , Castelli , S. , Katkar , P. , Desideri , A. , Bollati-Fogolin , M. , and Etcheverry , S.B. Oxidovanadium(IV) complexes with chrysin and silibinin: anticancer activity and mechanisms of action in a human colon adenocarcinoma model. J Biol Inorg Chem 20 , 1175 1191 ( 2015) DOI: 10.1007/s00775-015-1298-7
[203]
Ronnekleiv-Kelly , S.M. , Nukaya , M. , Diaz-Diaz , C.J. , Megna , B.W. , Carney , P.R. , Geiger , P.G. , and Kennedy , G.D. Aryl hydrocarbon receptor-dependent apoptotic cell death induced by the flavonoid chrysin in human colorectal cancer cells. Cancer Lett 370 , 91 99 ( 2016) DOI: 10.1016/j.canlet.2015.10.014
[204]
Xia , Y. , Lian , S. , Khoi , P.N. , Yoon , H.J. , Joo , Y.E. , Chay , K.O. , Kim , K.K. , and Do Jung , Y. Chrysin inhibits tumor promoter-induced MMP-9 expression by blocking AP-1 via suppression of ERK and JNK pathways in gastric cancer cells. PLoS One 10 , e0124007 ( 2015) DOI: 10.1371/journal.pone.0124007
[205]
Jia , W.Z. , Zhao , J.C. , Sun , X.L. , Yao , Z.G. , Wu , H.L. , and Xi , Z.Q. Additive anticancer effects of chrysin and low dose cisplatin in human malignant glioma cell (U87) proliferation and evaluation of the mechanistic pathway. J Buon 20 , 1327 1336 . ( 2015)
[206]
Rehman , M.U. , Ali , N. , Rashid , S. , Jain , T. , Nafees , S. , Tahir , M. , Khan , A.Q. , Lateef , A. , Khan , R. , Hamiza , O.O. , Kazim , S. , Qamar , W. , and Sultana , S. Alleviation of hepatic injury by chrysin in cisplatin administered rats: probable role of oxidative and inflammatory markers. Pharmacol Rep 66 , 1050 1059 ( 2014) DOI: 10.1016/j.pharep.2014.06.004
[207]
Samarghandian , S. , Afshari , J.T. , and Davoodi , S. Chrysin reduces proliferation and induces apoptosis in the human prostate cancer cell line pc-3. Clinics (Sao Paulo) 66 , 1073 1079 ( 2011) DOI: 10.1590/S1807-59322011000600026
[208]
Pichichero , E. , Cicconi , R. , Mattei , M. , and Canini , A. Chrysin-induced apoptosis is mediated through p38 and Bax activation in B16-F1 and A375 melanoma cells. Int J Oncol 38 , 473 483 ( 2011) DOI: 10.3892/ijo.2010.876
[209]
Leon , I.E. , Cadavid-Vargas , J.F. , Resasco , A. , Maschi , F. , Ayala , M.A. , Carbone , C. , and Etcheverry , S.B. n vitro and in vivo antitumor effects of the VO-chrysin complex on a new three-dimensional osteosarcoma spheroids model and a xenograft tumor in mice. J Biol Inorg Chem 21 , 1009 1020 ( 2016) DOI: 10.1007/s00775-016-1397-0
[210]
Samarghandian , S. , Nezhad , M.A. , and Mohammadi , G. Role of caspases, Bax and Bcl-2 in chrysin-induced apoptosis in the A549 human lung adenocarcinoma epithelial cells. Anticancer Agents Med Chem 14 , 901 909 ( 2014) DOI: 10.2174/1871520614666140209144042
[211]
Yu , X.M. , Phan , T. , Patel , P.N. , Jaskula-Sztul , R. , and Chen , H. Chrysin activates Notch1 signaling and suppresses tumor growth of anaplastic thyroid carcinoma in vitro and in vivo. Cancer 119 , 774 781 ( 2013) DOI: 10.1002/cncr.27742
[212]
Li , H.Z. , Chen , Y.H. , Fang , Y.L. , Zhong , L.Y. , Yuan , Q.Q. , Xu , X.Y. , and Cao , J.G. Effects of chrysin on sphere formation and CK2alpha expression of ovarian cancer stem-like cells derived from SKOV3 cell line. Zhonghua Yi Xue Za Zhi 96 , 2013 2016 ( 2016) DOI: 10.3760/cma.j.issn.0376-2491.2016.25.012
[213]
Xie , J. , Gao , H. , Peng , J. , Han , Y. , Chen , X. , Jiang , Q. , and Wang , C. Hispidulin prevents hypoxia-induced epithelial-mesenchymal transition in human colon carcinoma cells. Am J Cancer Res 5 , 1047 1061 ( 2015)
[214]
Yu , C.Y. , Su , K.Y. , Lee , P.L. , Jhan , J.Y. , Tsao , P.H. , Chan , D.C. , and Chen , Y.L. Potential therapeutic role of hispidulin in gastric cancer through induction of apoptosis via nag-1 signaling. Evid Based Complement Alternat Med 2013 , 518301 ( 2013) DOI: 10.1155/2013/518301
[215]
Gao , H. , Gao , M.Q. , Peng , J.J. , Han , M. , Liu , K.L. , and Han , Y.T. Hispidulin mediates apoptosis in human renal cell carcinoma by inducing ceramide accumulation. Acta Pharmacol Sin 38 , 1618 1631 ( 2017) DOI: 10.1038/aps.2017.154
[216]
Jeon , Y.W. , Ahn , Y.E. , Chung , W.S. , Choi , H.J. , and Suh , Y.J. Synergistic effect between celecoxib and luteolin is dependent on estrogen receptor in human breast cancer cells. Tumour Biol 36 , 6349 6359 ( 2015) DOI: 10.1007/s13277-015-3322-5
[217]
Lin , C.C. , Chuang , Y.J. , Yu , C.C. , Yang , J.S. , Lu , C.C. , Chiang , J.H. , Lin , J.P. , Tang , N.Y. , Huang , A.C. , and Chung , J.G. Apigenin induces apoptosis through mitochondrial dysfunction in U-2 OS human osteosarcoma cells and inhibits osteosarcoma xenograft tumor growth in vivo. J Agric Food Chem 60 , 11395 11402 ( 2012) DOI: 10.1021/jf303446x
[218]
Abdel Hadi , L. , Di Vito , C. , Marfia , G. , Ferraretto , A. , Tringali , C. , Viani , P. , and Riboni , L. Sphingosine kinase 2 and ceramide transport as key targets of the natural flavonoid luteolin to induce apoptosis in colon cancer cells. PLoS One 10 , e0143384 ( 2015) DOI: 10.1371/journal.pone.0143384
[219]
Krifa , M. , Leloup , L. , Ghedira , K. , Mousli , M. , and Chekir-Ghedira , L. Luteolin induces apoptosis in BE colorectal cancer cells by downregulating calpain, UHRF1, and DNMT1 expressions. Nutr Cancer 66 , 1220 1227 ( 2014) DOI: 10.1080/01635581.2014.951729
[220]
Wang , T.T. , Wang , S.K. , Huang , G.L. , and Sun , G.J. Luteolin induced-growth inhibition and apoptosis of human esophageal squamous carcinoma cell line Eca109 cells in vitro. Asian Pac J Cancer Prev 13 , 5455 5461 ( 2012) DOI: 10.7314/APJCP.2012.13.11.5455
[221]
Wu , H. , Huang , M. , Liu , Y. , Shu , Y. , and Liu , P. Luteolin induces apoptosis by up-regulating miR-34a in human gastric cancer cells. Technol Cancer Res Treat 14 , 747 755 ( 2015) DOI: 10.7785/tcrt.2012.500434
[222]
Chakrabarti , M. , and Ray , S.K. Anti-tumor activities of luteolin and silibinin in glioblastoma cells: overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo. Apoptosis 21 , 312 328 ( 2016) DOI: 10.1007/s10495-015-1198-x
[223]
Xu , H. , Yang , T. , Liu , X. , Tian , Y. , Chen , X. , Yuan , R. , Su , S. , Lin , X. , and Du , G. Luteolin synergizes the antitumor effects of 5-fluorouracil against human hepatocellular carcinoma cells through apoptosis induction and metabolism. Life Sci 144 , 138 147 ( 2016) DOI: 10.1016/j.lfs.2015.12.002
[224]
Kasala , E.R. , Bodduluru , L.N. , Barua , C.C. , and Gogoi , R. Antioxidant and antitumor efficacy of Luteolin, a dietary flavone on benzo(a)pyrene-induced experimental lung carcinogenesis. Biomed Pharmacother 82 , 568 577 ( 2016) DOI: 10.1016/j.biopha.2016.05.042
[225]
Wang , F. , Gao , F. , Pan , S. , Zhao , S. , and Xue , Y. Luteolin induces apoptosis, G0/G1 cell cycle growth arrest and mitochondrial membrane potential loss in neuroblastoma brain tumor cells. Drug Res (Stuttg) 65 , 91 95 ( 2015) DOI: 10.1055/s-0034-1372648
[226]
Huang , X. , Dai , S. , Dai , J. , Xiao , Y. , Bai , Y. , Chen , B. , and Zhou , M. Luteolin decreases invasiveness, deactivates STAT3 signaling, and reverses interleukin-6 induced epithelial-mesenchymal transition and matrix metalloproteinase secretion of pancreatic cancer cells. Onco Targets Ther 8 , 2989 3001 ( 2015) DOI: 10.2147/OTT.S91511
[227]
Sakurai , M.A. , Ozaki , Y. , Okuzaki , D. , Naito , Y. , Sasakura , T. , Okamoto , A. , Tabara , H. , Inoue , T. , Hagiyama , M. , Ito , A. , Yabuta , N. , and Nojima , H. Gefitinib and luteolin cause growth arrest of human prostate cancer PC-3 cells via inhibition of cyclin G-associated kinase and induction of miR-630. PLoS One 9 , e100124 ( 2014) DOI: 10.1371/journal.pone.0100124
[228]
Weng , Z. , Patel , A.B. , Vasiadi , M. , Therianou , A. , and Theoharides , T.C. Luteolin inhibits human keratinocyte activation and decreases NF-kappaB induction that is increased in psoriatic skin. PLoS One 9 , e90739 ( 2014) DOI: 10.1371/journal.pone.0090739
[229]
Xia , N. , Chen , G. , Liu , M. , Ye , X. , Pan , Y. , Ge , J. , Mao , Y. , Wang , H. , Wang , J. , and Xie , S. Anti-inflammatory effects of luteolin on experimental autoimmune thyroiditis in mice. Exp Ther Med 12 , 4049 4054 ( 2016) DOI: 10.3892/etm.2016.3854
[230]
Liao , Y.X. , Kong , G.M. , Wu , K.Y. , Tao , W.H. , and Bo , P. (Bilateral regulation of luteolin on spleen cells and sarcoma S180 cells of ICR mice: an experimental study). Zhongguo Zhong Xi Yi Jie He Za Zhi 34 , 1374 1378 ( 2014)
[231]
Shen , X.F. , Teng , Y. , Sha , K.H. , Wang , X.Y. , Yang , X.L. , Guo , X.J. , Ren , L.B. , Wang , X.Y. , Li , J. , and Huang , N. Dietary flavonoid luteolin attenuates uropathogenic Escherichia. Coli invasion of the urinary bladder. Biofactors 42 , 674 685 ( 2016) DOI: 10.1002/biof.1314
[232]
Hsiao , P.C. , Lee , W.J. , Yang , S.F. , Tan , P. , Chen , H.Y. , Lee , L.M. , Chang , J.L. , Lai , G.M. , Chow , J.M. , and Chien , M.H. Nobiletin suppresses the proliferation and induces apoptosis involving MAPKs and caspase-8/-9/-3 signals in human acute myeloid leukemia cells. Tumour Biol 35 , 11903 11911 ( 2014) DOI: 10.1007/s13277-014-2457-0
[233]
Chen , C. , Ono , M. , Takeshima , M. , and Nakano , S. Antiproliferative and apoptosis-inducing activity of nobiletin against three subtypes of human breast cancer cell lines. Anticancer Res 34 , 1785 1792 ( 2014)
[234]
Wu , X. , Song , M. , Wang , M. , Zheng , J. , Gao , Z. , Xu , F. , Zhang , G. , and Xiao , H. Chemopreventive effects of nobiletin and its colonic metabolites on colon carcinogenesis. Mol Nutr Food Res 59 , 2383 2394 ( 2015) DOI: 10.1002/mnfr.201500378
[235]
Kawabata , K. , Murakami , A. , and Ohigashi , H. Nobiletin, a citrus flavonoid, down-regulates matrix metalloproteinase-7 (matrilysin) expression in HT-29 human colorectal cancer cells. Biosci Biotechnol Biochem 69 , 307 314 ( 2005) DOI: 10.1271/bbb.69.307
[236]
Moon , J.Y. , Cho , M. , Ahn , K.S. , and Cho , S.K. Nobiletin induces apoptosis and potentiates the effects of the anticancer drug 5-fluorouracil in p53-mutated SNU-16 human gastric cancer cells. Nutr Cancer 65 , 286 295 ( 2013) DOI: 10.1080/01635581.2013.756529
[237]
Lien , L.M. , Wang , M.J. , Chen , R.J. , Chiu , H.C. , Wu , J.L. , Shen , M.Y. , Chou , D.S. , Sheu , J.R. , Lin , K.H. , and Lu , W.J. Nobiletin, a polymethoxylated flavone, inhibits glioma cell growth and migration via arresting cell cycle and suppressing mapk and akt pathways. Phytother Res 30 , 214 221 ( 2016) DOI: 10.1002/ptr.5517
[238]
Ma , X. , Jin , S. , Zhang , Y. , Wan , L. , Zhao , Y. , and Zhou , L. Inhibitory effects of nobiletin on hepatocellular carcinoma in vitro and in vivo. Phytother Res 28 , 560 567 ( 2014) DOI: 10.1002/ptr.5024
[239]
Gao , X.J. , Liu , J.W. , Zhang , Q.G. , Zhang , J.J. , Xu , H.T. , and Liu , H.J. Nobiletin inhibited hypoxia-induced epithelial-mesenchymal transition of lung cancer cells by inactivating of Notch-1 signaling and switching on miR-200b. Pharmazie 70 , 256 262 ( 2015)
[240]
Yoon , H.S. , Lee , S.R. , Ko , H.C. , Choi , S.Y. , Park , J.G. , Kim , J.K. , and Kim , S.J. Involvement of extracellular signal-regulated kinase in nobiletin-induced melanogenesis in murine B16/F10 melanoma cells. Biosci Biotechnol Biochem 71 , 1781 1784 ( 2007) DOI: 10.1271/bbb.70088
[241]
Ikeda , A. , Nemoto , K. , Yoshida , C. , Miyata , S. , Mori , J. , Soejima , S. , Yokosuka , A. , Mimaki , Y. , Ohizumi , Y. , and Degawa , M. Suppressive effect of nobiletin, a citrus polymethoxyflavonoid that downregulates thioredoxin-interacting protein expression, on tunicamycin-induced apoptosis in SK-N-SH human neuroblastoma cells. Neurosci Lett 549 , 135 139 ( 2013) DOI: 10.1016/j.neulet.2013.06.004
[242]
Chen , J. , Chen , A.Y. , Huang , H. , Ye , X. , Rollyson , W.D. , Perry , H.E. , Brown , K.C. , Rojanasakul , Y. , Rankin , G.O. , Dasgupta , P. , and Chen , Y.C. The flavonoid nobiletin inhibits tumor growth and angiogenesis of ovarian cancers via the Akt pathway. Int J Oncol 46 , 2629 2638 ( 2015) DOI: 10.3892/ijo.2015.2946
[243]
Jiang , Y.P. , Guo , H. , Wang , X.B. Nobiletin (NOB) suppresses autophagic degradation via over-expressing AKT pathway and enhances apoptosis in multidrug-resistant SKOV3/TAX ovarian cancer cells. Biomed Pharmacother 103 , 29 37 ( 2018) DOI: 10.1016/j.biopha.2018.03.126
[244]
Chen , J. , Creed , A. , Chen , A.Y. , Huang , H. , Li , Z. , Rankin , G.O. , Ye , X. , Xu , G. , and Chen , Y.C. Nobiletin suppresses cell viability through AKT pathways in PC-3 and DU-145 prostate cancer cells. BMC Pharmacol Toxicol 15 , 59 ( 2014) DOI: 10.1186/2050-6511-15-59
[245]
Wang , C. , Cheng , Y. , Liu , H. , Xu , Y. , Peng , H. , Lang , J. , Liao , J. , Liu , H. , Liu , H. , and Fan , J. Pectolinarigenin suppresses the tumor growth in nasopharyngeal carcinoma. Cell Physiol Biochem 39 , 1795 1803 ( 2016) DOI: 10.1159/000447879
[246]
Zhang , T. , Li , S. , Li , J. , Yin , F. , Hua , Y. , Wang , Z. , Lin , B. , Wang , H. , Zou , D. , Zhou , Z. , Xu , J. , Yi , C. , and Cai , Z. Natural product pectolinarigenin inhibits osteosarcoma growth and metastasis via SHP-1-mediated STAT3 signaling inhibition. Cell Death Dis 7 , e2421 ( 2016) DOI: 10.1038/cddis.2016.305
[247]
Periyasamy , K. , Sivabalan , V. , Baskaran , K. , Kasthuri , K. , and Sakthisekaran , D. Cellular metabolic energy modulation by tangeretin in 7,12-dimethylbenz(a) anthracene-induced breast cancer. J Biomed Res 30 , 134 141 ( 2016) DOI: 10.7555/JBR.30.20150060
[248]
Morley , K.L. , Ferguson , P.J. , and Koropatnick , J. Tangeretin and nobiletin induce G1 cell cycle arrest but not apoptosis in human breast and colon cancer cells. Cancer Lett 251 , 168 178 ( 2007) DOI: 10.1016/j.canlet.2006.11.016
[249]
Pan , M.H. , Chen , W.J. , Lin-Shiau , S.Y. , Ho , C.T. , and Lin , J.K. Tangeretin induces cell-cycle G1 arrest through inhibiting cyclin-dependent kinases 2 and 4 activities as well as elevating Cdk inhibitors p21 and p27 in human colorectal carcinoma cells. Carcinogenesis 23 , 1677 1684 ( 2002) DOI: 10.1093/carcin/23.10.1677
[250]
Zhang , X. , Zheng , L. , Sun , Y. , Wang , T. , and Wang , B. Tangeretin enhances radiosensitivity and inhibits the radiation-induced epithelial-mesenchymal transition of gastric cancer cells. Oncol Rep 34 , 302 310 ( 2015) DOI: 10.3892/or.2015.3982
[251]
Ma , L.L. , Wang , D.W. , Yu , X.D. , and Zhou , Y.L. Tangeretin induces cell cycle arrest and apoptosis through upregulation of PTEN expression in glioma cells. Biomed Pharmacother 81 , 491 496 ( 2016) DOI: 10.1016/j.biopha.2016.04.006
[252]
Li , Y.R. , Li , S. , Ho , C.T. , Chang , Y.H. , Tan , K.T. , Chung , T.W. , Wang , B.Y. , Chen , Y.K. , and Lin , C.C. Tangeretin derivative, 5-acetyloxy-6,7,8,4’-tetramethoxyflavone induces G2/M arrest, apoptosis and autophagy in human non-small cell lung cancer cells in vitro and in vivo. Cancer Biol Ther 17 , 48 64 ( 2016) DOI: 10.1080/15384047.2015.1108491
[253]
Yoon , H.S. , Ko , H.C. , Kim , S.S. , Park , K.J. , An , H.J. , Choi , Y.H. , Kim , S.J. , Lee , N.H. , and Hyun , C.G. Tangeretin triggers melanogenesis through the activation of melanogenic signaling proteins and sustained extracellular signal- regulated kinase in B16/F10 murine melanoma cells. Nat Prod Commun 10 , 389 392 ( 2015) DOI: 10.1016/j.bmcl.2012.10.130
[254]
Arafa El , S.A. , Zhu , Q. , Barakat , B.M. , Wani , G. , Zhao , Q. , El-Mahdy , M.A. , and Wani , A.A. Tangeretin sensitizes cisplatin-resistant human ovarian cancer cells through downregulation of phosphoinositide 3-kinase/Akt signaling pathway. Cancer Res 69 , 8910 8917 ( 2009) DOI: 10.1158/0008-5472.CAN-09-1543
[255]
Yu , J.S. , and Kim , A.K. Wogonin induces apoptosis by activation of ERK and p38 MAPKs signaling pathways and generation of reactive oxygen species in human breast cancer cells. Mol Cells 31 , 327 335 ( 2011) DOI: 10.1007/s10059-011-0041-7
[256]
Zhao , K. , Wei , L. , Hui , H. , Dai , Q. , You , Q.D. , Guo , Q.L. , and Lu , N. Wogonin suppresses melanoma cell B16-F10 invasion and migration by inhibiting Ras-medicated pathways. PLoS One 9 , e106458 ( 2014) DOI: 10.1371/journal.pone.0106458
[257]
Wang , Y. , Zhang , Y. , Qian , C. , Cai , M. , Li , Y. , Li , Z. , You , Q. , Wang , Q. , Hu , R. , and Guo , Q. GSK3beta/beta-catenin signaling is correlated with the differentiation of glioma cells induced by wogonin. Toxicol Lett 222 , 212 223 ( 2013) DOI: 10.1016/j.toxlet.2013.07.013
[258]
Zhang , H. , Liu , X. , Wu , F. , Qin , F. , Feng , P. , Xu , T. , Li , X. , and Yang , L. A novel prostate-specific membrane-antigen (PSMA) targeted micelle-encapsulating wogonin inhibits prostate cancer cell proliferation via inducing intrinsic apoptotic pathway. Int J Mol Sci 17 ( 2016) DOI: 10.3390/ijms17050676
[259]
Peng , M.X. , Wang , X.Y. , Wang , F. , Wang , L. , Xu , P.P. , and Chen , B. Apoptotic Mechanism of Human Leukemia K562/A02 Cells Induced by Magnetic Ferroferric Oxide Nanoparticles Loaded with Wogonin. Chin Med J (Engl) 129 , 2958 2966 ( 2016) DOI: 10.4103/0366-6999.195466
[260]
Chen , X.M. , Bai , Y. , Zhong , Y.J. , Xie , X.L. , Long , H.W. , Yang , Y.Y. , Wu , S.G. , Jia , Q. , and Wang , X.H. Wogonin has multiple anti-cancer effects by regulating c-Myc/SKP2/Fbw7alpha and HDAC1/HDAC2 pathways and inducing apoptosis in human lung adenocarcinoma cell line A549. PLoS One 8 , e79201 ( 2013) DOI: 10.1371/journal.pone.0079201
[261]
Wang , T. , Gao , J. , Yu , J. , and Shen , L. Synergistic inhibitory effect of wogonin and low-dose paclitaxel on gastric cancer cells and tumor xenografts. Chin J Cancer Res 25 , 505 513 ( 2013) DOI: 10.3978%2Fj.issn.1000-9604.2013.08.14
[262]
Ruibin , J. , Bo , J. , Danying , W. , Chihong , Z. , Jianguo , F. , and Linhui , G. Therapy effects of wogonin on ovarian cancer cells. Biomed Res Int 2017 , 9381513 ( 2017) DOI: 10.1155/2017/9381513
[263]
Yao , J. , Zhao , L. , Zhao , Q. , Zhao , Y. , Sun , Y. , Zhang , Y. , Miao , H. , You , Q.D. , Hu , R. , and Guo , Q.L. NF-kappaB and Nrf2 signaling pathways contribute to wogonin-mediated inhibition of inflammation-associated colorectal carcinogenesis. Cell Death Dis 5 , e1283 ( 2014) DOI: 10.1038/cddis.2014.221
[264]
Adan , A. , and Baran , Y. The pleiotropic effects of fisetin and hesperetin on human acute promyelocytic leukemia cells are mediated through apoptosis, cell cycle arrest, and alterations in signaling networks. Tumour Biol 36 , 8973 8984 . ( 2015) DOI: 10.1007/s13277-015-3597-6
[265]
Smith , M.L. , Murphy , K. , Doucette , C.D. , Greenshields , A.L. , and Hoskin , D.W. The dietary flavonoid fisetin causes cell cycle arrest, caspase-dependent apoptosis, and enhanced cytotoxicity of chemotherapeutic drugs in triple-negative breast cancer cells. J Cell Biochem 117 , 1913 1925 ( 2016) DOI: 10.1002/jcb.25490
[266]
Chou , R.H. , Hsieh , S.C. , Yu , Y.L. , Huang , M.H. , Huang , Y.C. , and Hsieh , Y.H. Fisetin inhibits migration and invasion of human cervical cancer cells by down-regulating urokinase plasminogen activator expression through suppressing the p38 MAPK-dependent NF-kappaB signaling pathway. PLoS One 8 , e71983 ( 2013) DOI: 10.1371/journal.pone.0071983
[267]
Chen , Y. , Wu , Q. , Song , L. , He , T. , Li , Y. , Li , L. , Su , W. , Liu , L. , Qian , Z. , and Gong , C. Polymeric micelles encapsulating fisetin improve the therapeutic effect in colon cancer. ACS Appl Mater Interfaces 7 , 534 542 ( 2015) DOI: 10.1021/am5066893
[268]
Leu , J.D. , Wang , B.S. , Chiu , S.J. , Chang , C.Y. , Chen , C.C. , Chen , F.D. , Avirmed , S. , and Lee , Y.J. Combining fisetin and ionizing radiation suppresses the growth of mammalian colorectal cancers in xenograft tumor models. Oncol Lett 12 , 4975 4982 ( 2016) DOI: 10.3892/ol.2016.5345
[269]
Sabarwal , A. , Agarwal , R. , and Singh , R.P. Fisetin inhibits cellular proliferation and induces mitochondria-dependent apoptosis in human gastric cancer cells. Mol Carcinog 56 , 499 514 ( 2017) DOI: 10.1002/mc.22512
[270]
Chen , C.M. , Hsieh , Y.H. , Hwang , J.M. , Jan , H.J. , Hsieh , S.C. , Lin , S.H. , and Lai , C.Y. Fisetin suppresses ADAM9 expression and inhibits invasion of glioma cancer cells through increased phosphorylation of ERK1/2. Tumour Biol 36 , 3407 3415 ( 2015) DOI: 10.1007/s13277-014-2975-9
[271]
Maurya , B.K. , and Trigun , S.K. Fisetin modulates antioxidant enzymes and inflammatory factors to inhibit aflatoxin-b1 induced hepatocellular carcinoma in rats. Oxid Med Cell Longev 2016 , 1972793 ( 2016) DOI: 10.1155/2016/1972793
[272]
Ravichandran , N. , Suresh , G. , Ramesh , B. , Manikandan , R. , Choi , Y.W. , and Vijaiyan Siva , G. Fisetin modulates mitochondrial enzymes and apoptotic signals in benzo(a)pyrene-induced lung cancer. Mol Cell Biochem 390 , 225 234 ( 2014) DOI: 10.1007/s11010-014-1973-y
[273]
Syed , D.N. , Lall , R.K. , Chamcheu , J.C. , Haidar , O. , and Mukhtar , H. Involvement of ER stress and activation of apoptotic pathways in fisetin induced cytotoxicity in human melanoma. Arch Biochem Biophys 563 , 108 117 ( 2014) DOI: 10.1016/j.abb.2014.06.034
[274]
Meng , Y.B. , Xiao , C. , Chen , X.L. , Bai , P. , Yao , Y. , Wang , H. , and Xiao , X. (The Antitumor Effects of Fisetin on Ovarian Cancer in vitro and in vivo.). Sichuan Da Xue Xue Bao Yi Xue Ban 47 , 830 836 ( 2016)
[275]
Mukhtar , E. , Adhami , V.M. , Sechi , M. , and Mukhtar , H. Dietary flavonoid fisetin binds to beta-tubulin and disrupts microtubule dynamics in prostate cancer cells. Cancer Lett 367 , 173 183 ( 2015) DOI: 10.1016/j.canlet.2015.07.030
[276]
Hu , S. , Huang , L. , Meng , L. , Sun , H. , Zhang , W. , and Xu , Y. Isorhamnetin inhibits cell proliferation and induces apoptosis in breast cancer via Akt and mitogenactivated protein kinase kinase signaling pathways. Mol Med Rep 12 , 6745 6751 ( 2015) DOI: 10.3892/mmr.2015.4269
[277]
Li , C. , Yang , X. , Chen , C. , Cai , S. , and Hu , J. Isorhamnetin suppresses colon cancer cell growth through the PI3KAktmTOR pathway. Mol Med Rep 9 , 935 940 ( 2014) DOI: 10.3892/mmr.2014.1886
[278]
Saud , S.M. , Young , M.R. , Jones-Hall , Y.L. , Ileva , L. , Evbuomwan , M.O. , Wise , J. , Colburn , N.H. , Kim , Y.S. , and Bobe , G. Chemopreventive activity of plant flavonoid isorhamnetin in colorectal cancer is mediated by oncogenic Src and beta-catenin. Cancer Res 73 , 5473 5484 ( 2013) DOI: 10.1158/0008-5472.CAN-13-0525
[279]
Li , Q. , Ren , F.Q. , Yang , C.L. , Zhou , L.M. , Liu , Y.Y. , Xiao , J. , Zhu , L. , and Wang , Z.G. Anti-proliferation effects of isorhamnetin on lung cancer cells in vitro and in vivo. Asian Pac J Cancer Prev 16 , 3035 3042 ( 2015) DOI: 10.7314/APJCP.2015.16.7.3035
[280]
Ramachandran , L. , Manu , K.A. , Shanmugam , M.K. , Li , F. , Siveen , K.S. , Vali , S. , Kapoor , S. , Abbasi , T. , Surana , R. , Smoot , D.T. , Ashktorab , H. , Tan , P. , Ahn , K.S. , Yap , C.W. , Kumar , A.P. , and Sethi , G. Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor gamma activation pathway in gastric cancer. J Biol Chem 287 , 38028 38040 ( 2012) DOI: 10.1074/jbc.M112.388702
[281]
Kim , J.E. , Lee , D.E. , Lee , K.W. , Son , J.E. , Seo , S.K. , Li , J. , Jung , S.K. , Heo , Y.S. , Mottamal , M. , Bode , A.M. , Dong , Z. , and Lee , H.J. Isorhamnetin suppresses skin cancer through direct inhibition of MEK1 and PI3-K. Cancer Prev Res (Phila) 4 , 582 591 ( 2011) DOI: 10.1158/1940-6207.CAPR-11-0032
[282]
Kim , S.H. , Hwang , K.A. , and Choi , K.C. Treatment with kaempferol suppresses breast cancer cell growth caused by estrogen and triclosan in cellular and xenograft breast cancer models. J Nutr Biochem 28 , 70 82 ( 2016) DOI: 10.1016/j.jnutbio.2015.09.027
[283]
Tu , L.Y. , Bai , H.H. , Cai , J.Y. , and Deng , S.P. The mechanism of kaempferol induced apoptosis and inhibited proliferation in human cervical cancer SiHa cell: From macro to nano. Scanning 38 , 644 653 ( 2016) DOI: 10.1002/sca.21312
[284]
Lee , H.S. , Cho , H.J. , Kwon , G.T. , and Park , J.H. Kaempferol downregulates insulin-like growth factor-i receptor and erbb3 signaling in ht-29 human colon cancer cells. J Cancer Prev 19 , 161 169 ( 2014) DOI: 10.15430/JCP.2014.19.3.161
[285]
Nirmala , P. , and Ramanathan , M. Effect of kaempferol on lipid peroxidation and antioxidant status in 1,2-dimethyl hydrazine induced colorectal carcinoma in rats. Eur J Pharmacol 654 , 75 79 ( 2011) DOI: 10.1016/j.ejphar.2010.11.034
[286]
Li , R.J. , Mei , J.Z. , and Liu , G.J. Kaempferol-induced apoptosis of human esophageal squamous carcinoma Eca-109 cells and the mechanism. Nan Fang Yi Ke Da Xue Xue Bao 31 , 1440 1442 ( 2011)
[287]
Song , H. , Bao , J. , Wei , Y. , Chen , Y. , Mao , X. , Li , J. , Yang , Z. , and Xue , Y. Kaempferol inhibits gastric cancer tumor growth: An in vitro and in vivo study. Oncol Rep 33 , 868 874 ( 2015) DOI: 10.3892/or.2014.3662
[288]
Jeong , J.C. , Kim , M.S. , Kim , T.H. , and Kim , Y.K. Kaempferol induces cell death through ERK and Akt-dependent down-regulation of XIAP and survivin in human glioma cells. Neurochem Res 34 , 991 1001 ( 2009) DOI: 10.1007/s11064-008-9868-5
[289]
Huang , W.W. , Tsai , S.C. , Peng , S.F. , Lin , M.W. , Chiang , J.H. , Chiu , Y.J. , Fushiya , S. , Tseng , M.T. , and Yang , J.S. Kaempferol induces autophagy through AMPK and AKT signaling molecules and causes G2/M arrest via downregulation of CDK1/cyclin B in SK-HEP-1 human hepatic cancer cells. Int J Oncol 42 , 2069 2077 ( 2013) DOI: 10.3892/ijo.2013.1909
[290]
Moradzadeh , M. , Tabarraei , A. , Sadeghnia , H.R. , Ghorbani , A. , Mohamadkhani , A. , Erfanian , S. , and Sahebkar , A. Kaempferol increases apoptosis in human acute promyelocytic leukemia cells and inhibits multidrug resistance genes. J Cell Biochem 119 , 2288 2297 ( 2018) DOI: 10.1002/jcb.26391
[291]
Jo , E. , Park , S.J. , Choi , Y.S. , Jeon , W.K. , and Kim , B.C. Kaempferol Suppresses Transforming Growth Factor-beta1-Induced Epithelial-to-Mesenchymal Transition and Migration of A549 Lung Cancer Cells by Inhibiting Akt1-Mediated Phosphorylation of Smad3 at Threonine-179. Neoplasia 17 , 525 537 ( 2015) DOI: 10.1016/j.neo.2015.06.004
[292]
Luo , H. , Jiang , B. , Li , B. , Li , Z. , Jiang , B.H. , and Chen , Y.C. Kaempferol nanoparticles achieve strong and selective inhibition of ovarian cancer cell viability. Int J Nanomedicine 7 , 3951 3959 ( 2012) DOI: 10.2147/IJN.S33670
[293]
Lee , J. , and Kim , J.H. Kaempferol inhibits pancreatic cancer cell growth and migration through the blockade of egfr-related pathway in vitro. PLoS One 11 , e0155264 ( 2016) DOI: 10.1371/journal.pone.0155264
[294]
Halimah , E. , Diantini , A. , Destiani , D.P. , Pradipta , I.S. , Sastramihardja , H.S. , Lestari , K. , Subarnas , A. , Abdulah , R. , and Koyama , H. Induction of caspase cascade pathway by kaempferol-3-O-rhamnoside in LNCaP prostate cancer cell lines. Biomed Rep 3 , 115 117 ( 2015) DOI: 10.3892/br.2014.385
[295]
Yao , K. , Chen , H. , Liu , K. , Langfald , A. , Yang , G. , Zhang , Y. , Yu , D.H. , Kim , M.O. , Lee , M.H. , Li , H. , Bae , K.B. , Kim , H.G. , Ma , W.Y. , Bode , A.M. , Dong , Z. , and Dong , Z. Kaempferol targets RSK2 and MSK1 to suppress UV radiation-induced skin cancer. Cancer Prev Res (Phila) 7 , 958 967 ( 2014) DOI: 10.1158/1940-6207.CAPR-14-0126
[296]
Dang , Q. , Song , W. , Xu , D. , Ma , Y. , Li , F. , Zeng , J. , Zhu , G. , Wang , X. , Chang , L.S. , He , D. , and Li , L. Kaempferol suppresses bladder cancer tumor growth by inhibiting cell proliferation and inducing apoptosis. Mol Carcinog 54 , 831 840 ( 2015) DOI: 10.1002/mc.22154
[297]
Jiao , D. , and Zhang , X.D. Myricetin suppresses p21-activated kinase 1 in human breast cancer MCF-7 cells through downstream signaling of the beta-catenin pathway. Oncol Rep 36 , 342 348 ( 2016) DOI: 10.3892/or.2016.4777
[298]
Yi , J.L. , Shi , S. , Shen , Y.L. , Wang , L. , Chen , H.Y. , Zhu , J. , and Ding , Y. Myricetin and methyl eugenol combination enhances the anticancer activity, cell cycle arrest and apoptosis induction of cis-platin against HeLa cervical cancer cell lines. Int J Clin Exp Pathol 8 , 1116 1127 ( 2015)
[299]
Kim , M.E. , Ha , T.K. , Yoon , J.H. , and Lee , J.S. Myricetin induces cell death of human colon cancer cells via BAX/BCL2-dependent pathway. Anticancer Res 34 , 701 706 ( 2014)
[300]
Ko , C.H. , Shen , S.C. , Lee , T.J. , and Chen , Y.C. Myricetin inhibits matrix metalloproteinase 2 protein expression and enzyme activity in colorectal carcinoma cells. Mol Cancer Ther 4 , 281 290 ( 2005)
[301]
Zang , W. , Wang , T. , Wang , Y. , Li , M. , Xuan , X. , Ma , Y. , Du , Y. , Liu , K. , Dong , Z. , and Zhao , G. Myricetin exerts anti-proliferative, anti-invasive, and pro-apoptotic effects on esophageal carcinoma EC9706 and KYSE30 cells via RSK2. Tumour Biol 35 , 12583 12592 ( 2014) DOI: 10.1007/s13277-014-2579-4
[302]
Feng , J. , Chen , X. , Wang , Y. , Du , Y. , Sun , Q. , Zang , W. , and Zhao , G. Myricetin inhibits proliferation and induces apoptosis and cell cycle arrest in gastric cancer cells. Mol Cell Biochem 408 ( 1-2 ), 163 170 ( 2015) DOI: 10.1007/s11010-015-2492-1
[303]
Wang , G. , Wang , J.J. , Tang , X.J. , Du , L. , and Li , F. In vitro and in vivo evaluation of functionalized chitosan-Pluronic micelles loaded with myricetin on glioblastoma cancer. Nanomedicine 12 , 1263 1278 ( 2016) DOI: 10.1016/j.nano.2016.02.004
[304]
Iyer , S.C. , Gopal , A. , and Halagowder , D. Myricetin induces apoptosis by inhibiting P21 activated kinase 1 (PAK1) signaling cascade in hepatocellular carcinoma. Mol Cell Biochem 407 , 223 237 ( 2015) DOI: 10.1007/s11010-015-2471-6
[305]
Zhang , S. , Wang , L. , Liu , H. , Zhao , G. , and Ming , L. Enhancement of recombinant myricetin on the radiosensitivity of lung cancer A549 and H1299 cells. Diagn Pathol 9 , 68 ( 2014) DOI: 10.1186/1746-1596-9-68
[306]
Xu , Y. , Xie , Q. , Wu , S. , Yi , D. , Yu , Y. , Liu , S. , Li , S. , and Li , Z. Myricetin induces apoptosis via endoplasmic reticulum stress and DNA double-strand breaks in human ovarian cancer cells. Mol Med Rep 13 , 2094 2100 ( 2016) DOI: 10.3892/mmr.2016.4763
[307]
Sun , M. , Nie , S. , Pan , X. , Zhang , R. , Fan , Z. , and Wang , S. Quercetin-nanostructured lipid carriers: characteristics and anti-breast cancer activities in vitro. Colloids Surf B Biointerfaces 113 , 15 24 ( 2014) DOI: 10.1016/j.colsurfb.2013.08.032
[308]
Luo , C.L. , Liu , Y.Q. , Wang , P. , Song , C.H. , Wang , K.J. , Dai , L.P. , Zhang , J.Y. , and Ye , H. The effect of quercetin nanoparticle on cervical cancer progression by inducing apoptosis, autophagy and anti-proliferation via JAK2 suppression. Biomed Pharmacother 82 , 595 605 ( 2016) DOI: 10.1016/j.biopha.2016.05.029
[309]
Russo , M. , Spagnuolo , C. , Volpe , S. , Tedesco , I. , Bilotto , S. , and Russo , G.L. ABT-737 resistance in B-cells isolated from chronic lymphocytic leukemia patients and leukemia cell lines is overcome by the pleiotropic kinase inhibitor quercetin through Mcl-1 down-regulation. Biochem Pharmacol 85 , 927 936 ( 2013) DOI: 10.1016/j.bcp.2013.01.011
[310]
Refolo , M.G. , D’alessandro , R. , Malerba , N. , Laezza , C. , Bifulco , M. , Messa , C. , Caruso , M.G. , Notarnicola , M. , and Tutino , V. Anti proliferative and pro apoptotic effects of flavonoid quercetin are mediated by cb1 receptor in human colon cancer cell lines. J Cell Physiol 230 , 2973 2980 ( 2015) DOI: 10.1002/jcp.25026
[311]
Cho , S.Y. , Kim , M.K. , Park , K.S. , Choo , H. , and Chong , Y. Quercetin-POC conjugates: Differential stability and bioactivity profiles between breast cancer (MCF-7) and colorectal carcinoma (HCT116) cell lines. Bioorg Med Chem 21 , 1671 1679 ( 2013) DOI: 10.1016/j.bmc.2013.01.057
[312]
Zheng , N.G. , Mo , S.J. , Li , J.P. , and Wu , J.L. Anti-CSC effects in human esophageal squamous cell carcinomas and Eca109/9706 cells induced by nanoliposomal quercetin alone or combined with CD 133 antiserum. Asian Pac J Cancer Prev 15 , 8679 8684 ( 2014) DOI: 10.7314/APJCP.2014.15.20.8679
[313]
Zhang , J.Y. , Lin , M.T. , Zhou , M.J. , Yi , T. , Tang , Y.N. , Tang , S.L. , Yang , Z.J. , Zhao , Z.Z. , and Chen , H.B. Combinational treatment of curcumin and quercetin against gastric cancer MGC-803 cells in vitro. Molecules 20 , 11524 11534 ( 2015) DOI: 10.3390/molecules200611524
[314]
Maurya , A.K. , and Vinayak , M. Anticarcinogenic action of quercetin by downregulation of phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC) via induction of p53 in hepatocellular carcinoma (HepG2) cell line. Mol Biol Rep 42 , 1419 1429 ( 2015) DOI: 10.1007/s11033-015-3921-7
[315]
Zhang , X. , Guo , Q. , Chen , J. , and Chen , Z. Quercetin enhances cisplatin sensitivity of human osteosarcoma cells by modulating microRNA-217-KRAS axis. Mol Cells 38 , 638 642 ( 2015) DOI: 10.14348/molcells.2015.0037
[316]
Wang , Y. , Han , A. , Chen , E. , Singh , R.K. , Chichester , C.O. , Moore , R.G. , Singh , A.P. , and Vorsa , N. The cranberry flavonoids PAC DP-9 and quercetin aglycone induce cytotoxicity and cell cycle arrest and increase cisplatin sensitivity in ovarian cancer cells. Int J Oncol 46 , 1924 1934 ( 2015) DOI: 10.3892/ijo.2015.2931
[317]
Angst , E. , Park , J.L. , Moro , A. , Lu , Q.Y. , Lu , X. , Li , G. , King , J. , Chen , M. , Reber , H.A. , Go , V.L. , Eibl , G. , and Hines , O.J. The flavonoid quercetin inhibits pancreatic cancer growth in vitro and in vivo. Pancreas 42 , 223 229 ( 2013) DOI: 10.1097/MPA.0b013e318264ccae
[318]
Bhat , F.A. , Sharmila , G. , Balakrishnan , S. , Arunkumar , R. , Elumalai , P. , Suganya , S. , Raja Singh , P. , Srinivasan , N. , and Arunakaran , J. Quercetin reverses EGF-induced epithelial to mesenchymal transition and invasiveness in prostate cancer (PC-3) cell line via EGFR/PI3K/Akt pathway. J Nutr Biochem 25 , 1132 1139 ( 2014) DOI: 10.1016/j.jnutbio.2014.06.008
[319]
Quagliariello , V. , Armenia , E. , Aurilio , C. , Rosso , F. , Clemente , O. , De Sena , G. , Barbarisi , M. , and Barbarisi , A. New Treatment of medullary and papillary human thyroid cancer: biological effects of hyaluronic acid hydrogel loaded with quercetin alone or in combination to an inhibitor of aurora kinase. J Cell Physiol 231 , 1784 1795 ( 2016) DOI: 10.1002/jcp.25283
[320]
Su , Q. , Peng , M. , Zhang , Y. , Xu , W. , Darko , K.O. , Tao , T. , Huang , Y. , Tao , X. , and Yang , X. Quercetin induces bladder cancer cells apoptosis by activation of AMPK signaling pathway. Am J Cancer Res 6 , 498 508 ( 2016)
[321]
Guon , T.E. , and Chung , H.S. Hyperoside and rutin of Nelumbo nucifera induce mitochondrial apoptosis through a caspase-dependent mechanism in HT-29 human colon cancer cells. Oncol Lett 11 , 2463 2470 ( 2016) DOI: 10.3892/ol.2016.4247
[322]
Wu , F. , Chen , J. , Fan , L.M. , Liu , K. , Zhang , N. , Li , S.W. , Zhu , H. , and Gao , H.C. Analysis of the effect of rutin on GSK-3beta and TNF-alpha expression in lung cancer. Exp Ther Med 14 , 127 130 ( 2017) DOI: 10.3892/etm.2017.4494
[323]
Chen , H. , Miao , Q. , Geng , M. , Liu , J. , Hu , Y. , Tian , L. , Pan , J. , and Yang , Y. Anti-tumor effect of rutin on human neuroblastoma cell lines through inducing G2/M cell cycle arrest and promoting apoptosis. Scientific World Journal 2013 , 269165 ( 2013) DOI: 10.1155/2013/269165
[324]
Chuang , C.H. , Huang , C.S. , and Hu , M.L. Vitamin E and rutin synergistically inhibit expression of vascular endothelial growth factor through down-regulation of binding activity of activator protein-1 in human promyelocytic leukemia (HL-60) cells. Chem Biol Interact 183 , 434 441 ( 2010) DOI: 10.1016/j.cbi.2009.12.007
[325]
Ahmad , M. , Sahabjada, Akhtar , J. , Hussain , A. , Badaruddeen, Arshad , M. , and Mishra , A. Development of a new rutin nanoemulsion and its application on prostate carcinoma PC3 cell line. Excli J 16 , 810 823 ( 2017) DOI: 10.1016/j.redox.2017.04.014
[326]
Gegotek , A. , Bielawska , K. , Biernacki , M. , Dobrzynska , I. , and Skrzydlewska , E. Time-dependent effect of rutin on skin fibroblasts membrane disruption following UV radiation. Redox Biol 12 , 733 744 ( 2017) DOI: 10.3892/or.2011.1580
[327]
Wu , L. , Yang , W. , Zhang , S.N. , and Lu , J.B. Alpinetin inhibits lung cancer progression and elevates sensitization drug-resistant lung cancer cells to cis-diammined dichloridoplatium. Drug Des Devel Ther 9 , 6119 6127 ( 2015) DOI: 10.2147/DDDT.S92702
[328]
Tang , B. , Du , J. , Wang , J. , Tan , G. , Gao , Z. , Wang , Z. , and Wang , L. Alpinetin suppresses proliferation of human hepatoma cells by the activation of MKK7 and elevates sensitization to cis-diammined dichloridoplatium. Oncol Rep 27 , 1090 1096 ( 2012) DOI: 10.3892/or.2011.1580
[329]
Du , J. , Tang , B. , Wang , J. , Sui , H. , Jin , X. , Wang , L. , and Wang , Z. Antiproliferative effect of alpinetin in BxPC-3 pancreatic cancer cells. Int J Mol Med 29, 607-612. ( 2012) DOI: 10.3892/ijmm.2012.884
[330]
Desai , U.N. , Shah , K.P. , Mirza , S.H. , Panchal , D.K. , Parikh , S.K. , and Rawal , R.M. Enhancement of the cytotoxic effects of Cytarabine in synergism with Hesperidine and Silibinin in Acute Myeloid Leukemia: An in-vitro approach. J Cancer Res Ther 11 , 352 357 ( 2015) DOI: 10.4103/0973-1482.157330
[331]
Nandakumar , N. , Rengarajan , T. , Balamurugan , A. , and Balasubramanian , M.P. Modulating effects of hesperidin on key carbohydrate-metabolizing enzymes, lipid profile, and membrane-bound adenosine triphosphatases against 7,12-dimethylbenz(a)anthracene-induced breast carcinogenesis. Hum Exp Toxicol 33 , 504 516 ( 2014) DOI: 10.1177/0960327113485252
[332]
Saiprasad , G. , Chitra , P. , Manikandan , R. , and Sudhandiran , G. Hesperidin induces apoptosis and triggers autophagic markers through inhibition of Aurora-A mediated phosphoinositide-3-kinase/Akt/mammalian target of rapamycin and glycogen synthase kinase-3 beta signalling cascades in experimental colon carcinogenesis. Eur J Cancer 50 , 2489 2507 ( 2014) DOI: 10.1016/j.ejca.2014.06.013
[333]
Banjerdpongchai , R. , Wudtiwai , B. , Khaw-on , P. , Rachakhom , W. , Duangnil , N. , and Kongtawelert , P. Hesperidin from Citrus seed induces human hepatocellular carcinoma HepG2 cell apoptosis via both mitochondrial and death receptor pathways. Tumour Biol 37 , 227 237 ( 2016) DOI: 10.1007/s13277-015-3774-7
[334]
Birsu Cincin , Z. , Unlu , M. , Kiran , B. , Sinem Bireller , E. , Baran , Y. , and Cakmakoglu , B. Anti-proliferative, apoptotic and signal transduction effects of hesperidin in non-small cell lung cancer cells. Cell Oncol (Dordr) 38 , 195 204 ( 2015) DOI: 10.1007/s13402-015-0222-z
[335]
Yang , M. , Tanaka , T. , Hirose , Y. , Deguchi , T. , Mori , H. , and Kawada , Y. Chemopreventive effects of diosmin and hesperidin on N-butyl-N-(4-hydroxybutyl)nitrosamine-induced urinary-bladder carcinogenesis in male ICR mice. Int J Cancer 73 , 719 724 ( 1997) DOI: 10.1002/(SICI)1097-0215(19971127)73:5<719::AID-IJC18>3.0.CO;2-0
[336]
Zhao , J. , Li , Y. , Gao , J. , and De , Y. Hesperidin inhibits ovarian cancer cell viability through endoplasmic reticulum stress signaling pathways. Oncol Lett 14 , 5569 5574 ( 2017) DOI: 10.3892/ol.2017.6873
[337]
Yang , Y. , Wolfram , J. , Boom , K. , Fang , X. , Shen , H. , and Ferrari , M. Hesperetin impairs glucose uptake and inhibits proliferation of breast cancer cells. Cell Biochem Funct 31 , 374 379 ( 2013) DOI: 10.1002/cbf.2905
[338]
Patel , P.N. , Yu , X.M. , Jaskula-Sztul , R. , and Chen , H. Hesperetin activates the Notch1 signaling cascade, causes apoptosis, and induces cellular differentiation in anaplastic thyroid cancer. Ann Surg Oncol 21 Suppl 4 , S497 504 ( 2014) DOI: 10.1245/s10434-013-3459-7
[339]
Shirzad , M. , Heidarian , E. , Beshkar , P. , and Gholami-Arjenaki , M. Biological Effects of Hesperetin on Interleukin-6/Phosphorylated Signal Transducer and Activator of Transcription 3 Pathway Signaling in Prostate Cancer PC3 Cells. Pharmacognosy Res 9 , 188 194 ( 2017) DOI: 10.4103/0974-8490.204655
[340]
Zarebczan , B. , Pinchot , S.N. , Kunnimalaiyaan , M. , and Chen , H. Hesperetin, a potential therapy for carcinoid cancer. Am J Surg 201 , 329 332 ; discussion 333 ( 2011) DOI: 10.1016/j.amjsurg.2010.08.018
[341]
Abaza , M.S. , Orabi , K.Y. , Al-Quattan , E. , and Al-Attiyah , R.J. Growth inhibitory and chemo-sensitization effects of naringenin, a natural flavanone purified from Thymus vulgaris, on human breast and colorectal cancer. Cancer Cell Int 15 , 46 ( 2015) DOI: 10.1186/s12935-015-0194-0
[342]
Yoon , H. , Kim , T.W. , Shin , S.Y. , Park , M.J. , Yong , Y. , Kim , D.W. , Islam , T. , Lee , Y.H. , Jung , K.Y. , and Lim , Y. Design, synthesis and inhibitory activities of naringenin derivatives on human colon cancer cells. Bioorg Med Chem Lett 23 , 232 238 ( 2013) DOI: 10.1016/j.bmcl.2012.10.130
[343]
Song , H.M. , Park , G.H. , Eo , H.J. , Lee , J.W. , Kim , M.K. , Lee , J.R. , Lee , M.H. , Koo , J.S. , and Jeong , J.B. Anti-Proliferative Effect of Naringenin through p38-Dependent Downregulation of Cyclin D1 in Human Colorectal Cancer Cells. Biomol Ther (Seoul) 23 , 339 344 ( 2015) DOI: 10.4062/biomolther.2015.024
[344]
Li , H. , Zhu , F. , Chen , H. , Cheng , K.W. , Zykova , T. , Oi , N. , Lubet , R.A. , Bode , A.M. , Wang , M. , and Dong , Z. 6-C-(E-phenylethenyl)-naringenin suppresses colorectal cancer growth by inhibiting cyclooxygenase-1. Cancer Res 74 , 243 252 ( 2014) DOI: 10.1158/0008-5472.CAN-13-2245
[345]
Bao , L. , Liu , F. , Guo , H.B. , Li , Y. , Tan , B.B. , Zhang , W.X. , and Peng , Y.H. Naringenin inhibits proliferation, migration, and invasion as well as induces apoptosis of gastric cancer SGC7901 cell line by downregulation of AKT pathway. Tumour Biol 37 , 11365 11374 ( 2016) DOI: 10.1007/s13277-016-5013-2
[346]
Yen , H.R. , Liu , C.J. , and Yeh , C.C. Naringenin suppresses TPA-induced tumor invasion by suppressing multiple signal transduction pathways in human hepatocellular carcinoma cells. Chem Biol Interact 235 , 1 9 ( 2015) DOI: 10.1016/j.cbi.2015.04.003
[347]
Kumar , S.P. , Birundha , K. , Kaveri , K. , and Devi , K.T. Antioxidant studies of chitosan nanoparticles containing naringenin and their cytotoxicity effects in lung cancer cells. Int J Biol Macromol 78 , 87 95 ( 2015) DOI: 10.1016/j.ijbiomac.2015.03.045
[348]
Li , R.F. , Feng , Y.Q. , Chen , J.H. , Ge , L.T. , Xiao , S.Y. , and Zuo , X.L. Naringenin suppresses K562 human leukemia cell proliferation and ameliorates Adriamycin-induced oxidative damage in polymorphonuclear leukocytes. Exp Ther Med 9 , 697 706 ( 2015) DOI: 10.3892/etm.2015.2185
[349]
Lou , C. , Zhang , F. , Yang , M. , Zhao , J. , Zeng , W. , Fang , X. , Zhang , Y. , Zhang , C. , and Liang , W. Naringenin decreases invasiveness and metastasis by inhibiting TGF-beta-induced epithelial to mesenchymal transition in pancreatic cancer cells. PLoS One 7 , e50956 ( 2012) DOI: 10.1371/journal.pone.0050956
[350]
Torricelli , P. , Ricci , P. , Provenzano , B. , Lentini , A. , and Tabolacci , C. Synergic effect of alpha-tocopherol and naringenin in transglutaminase-induced differentiation of human prostate cancer cells. Amino Acids 41 , 1207 1214 ( 2011) DOI: 10.1007/s00726-010-0788-8
[351]
Kim , J.H. , and Lee , J.K. Naringenin enhances NK cell lysis activity by increasing the expression of NKG2D ligands on Burkitt’s lymphoma cells. Arch Pharm Res 38 , 2042 2048 ( 2015) DOI: 10.1007/s12272-015-0624-5
[352]
Minakawa , T. , Toume , K. , Ahmed , F. , Sadhu , S.K. , Ohtsuki , T. , Arai , M.A. , and Ishibashi , M. Constituents of Pongamia pinnata isolated in a screening for activity to overcome tumor necrosis factor-related apoptosis-inducing ligand-resistance. Chem Pharm Bull (Tokyo) 58 , 1549 1551 ( 2010) DOI: 10.1248/cpb.58.1549
[353]
Zhang , Z.R. , Al Zaharna , M. , Wong , M.M. , Chiu , S.K. , and Cheung , H.Y. Taxifolin enhances andrographolide-induced mitotic arrest and apoptosis in human prostate cancer cells via spindle assembly checkpoint activation. PLoS One 8 , e54577 ( 2013) DOI: 10.1371/journal.pone.0054577
[354]
Oi , N. , Chen , H. , Ok Kim , M. , Lubet , R.A. , Bode , A.M. , and Dong , Z. Taxifolin suppresses UV-induced skin carcinogenesis by targeting EGFR and PI3K. Cancer Prev Res (Phila) 5 , 1103 1114 ( 2012) DOI: 10.1158/1940-6207.CAPR-11-0397
[355]
Garcia- Maceira , P. , and Mateo , J. Silibinin inhibits hypoxia-inducible factor-1alpha and mTOR/p70S6K/4E-BP1 signalling pathway in human cervical and hepatoma cancer cells: implications for anticancer therapy. Oncogene 28 , 313 324 ( 2009) DOI: 10.1038/onc.2008.398
[356]
Bhatia , V. , and Falzon , M. Restoration of the anti-proliferative and anti-migratory effects of 1, 25-dihydroxyvitamin D by silibinin in vitamin D-resistant colon cancer cells. Cancer Lett 362 , 199 207 ( 2015) DOI: 10.1016/j.canlet.2015.03.042
[357]
Raina , K. , Agarwal , C. , Wadhwa , R. , Serkova , N.J. , and Agarwal , R. Energy deprivation by silibinin in colorectal cancer cells: a double-edged sword targeting both apoptotic and autophagic machineries. Autophagy 9 , 697 713 ( 2013) DOI: 10.4161/auto.23960
[358]
Wang , Y.X. , Cai , H. , Jiang , G. , Zhou , T.B. , and Wu , H. Silibinin inhibits proliferation, induces apoptosis and causes cell cycle arrest in human gastric cancer MGC803 cells via STAT3 pathway inhibition. Asian Pac J Cancer Prev 15 , 6791 6798 ( 2014) DOI: 10.7314/APJCP.2014.15.16.6791
[359]
Sadava , D. , and Kane , S.E. Silibinin reverses drug resistance in human small-cell lung carcinoma cells. Cancer Lett 339 , 102 106 ( 2013) DOI: 10.1016/j.canlet.2013.07.017
[360]
Li , J. , Li , B. , Xu , W.W. , Chan , K.W. , Guan , X.Y. , Qin , Y.R. , Lee , N.P. , Chan , K.T. , Law , S. , Tsao , S.W. , and Cheung , A.L. Role of AMPK signaling in mediating the anticancer effects of silibinin in esophageal squamous cell carcinoma. Expert Opin Ther Targets 20 , 7 18 ( 2016) DOI: 10.1517/14728222.2016.1121236
[361]
Momeny , M. , Ghasemi , R. , Valenti , G. , Miranda , M. , Zekri , A. , Zarrinrad , G. , Javadikooshesh , S. , Yaghmaie , M. , Alimoghaddam , K. , Ghavamzadeh , A. , and Ghaffari , S.H. Effects of silibinin on growth and invasive properties of human ovarian carcinoma cells through suppression of heregulin/HER3 pathway. Tumour Biol 37 , 3913 3923 ( 2016) DOI: 10.1007/s13277-015-4220-6
[362]
Shukla , S.K. , Dasgupta , A. , Mehla , K. , Gunda , V. , Vernucci , E. , Souchek , J. , Goode , G. , King , R. , Mishra , A. , Rai , I. , Nagarajan , S. , Chaika , N.V. , Yu , F. , and Singh , P.K. Silibinin-mediated metabolic reprogramming attenuates pancreatic cancer-induced cachexia and tumor growth. Oncotarget 6 , 41146 41161 ( 2015) DOI: 10.18632/oncotarget.5843
[363]
Prajapati , V. , Kale , R.K. , and Singh , R.P. Silibinin combination with arsenic strongly inhibits survival and invasiveness of human prostate carcinoma cells. Nutr Cancer 67 , 647 658 ( 2015) DOI: 10.1080/01635581.2015.1019635
[364]
Khan , A.Q. , Khan , R. , Tahir , M. , Rehman , M.U. , Lateef , A. , Ali , F. , Hamiza , O.O. , Hasan , S.K. , and Sultana , S. Silibinin inhibits tumor promotional triggers and tumorigenesis against chemically induced two-stage skin carcinogenesis in Swiss albino mice: possible role of oxidative stress and inflammation. Nutr Cancer 66 , 249 258 ( 2014) DOI: 10.1080/01635581.2014.863365
[365]
Wu , K. , Ning , Z. , Zeng , J. , Fan , J. , Zhou , J. , Zhang , T. , Zhang , L. , Chen , Y. , Gao , Y. , Wang , B. , Guo , P. , Li , L. , Wang , X. , and He , D. Silibinin inhibits beta-catenin/ZEB1 signaling and suppresses bladder cancer metastasis via dual-blocking epithelial-mesenchymal transition and stemness. Cell Signal 25 , 2625 2633 ( 2013) DOI: 10.1016/j.cellsig.2013.08.028
[366]
Oh , S.J. , Jung , S.P. , Han , J. , Kim , S. , Kim , J.S. , Nam , S.J. , Lee , J.E. , and Kim , J.H. Silibinin inhibits TPA-induced cell migration and MMP-9 expression in thyroid and breast cancer cells. Oncol Rep 29 , 1343 1348 ( 2013) DOI: 10.3892/or.2013.2252
[367]
Yu , H.C. , Chen , L.J. , Cheng , K.C. , Li , Y.X. , Yeh , C.H. , and Cheng , J.T. Silymarin inhibits cervical cancer cell through an increase of phosphatase and tensin homolog. Phytother Res 26 , 709 715 ( 2012) DOI: 10.1002/ptr.3618
[368]
Scavo , M.P. , Gentile , E. , Wolfram , J. , Gu , J. , Barone , M. , Evangelopoulos , M. , Martinez , J.O. , Liu , X. , Celia , C. , Tasciotti , E. , Vilar , E. , and Shen , H. Multistage vector delivery of sulindac and silymarin for prevention of colon cancer. Colloids Surf B Biointerfaces 136 , 694 703 ( 2015) DOI: 10.1016/j.colsurfb.2015.10.005
[369]
Yurtcu , E. , Darcansoy Iseri , O. , and Iffet Sahin , F. Effects of silymarin and silymarin-doxorubicin applications on telomerase activity of human hepatocellular carcinoma cell line HepG2. J Buon 20 , 555 561 ( 2015)
[370]
Singh , T. , Prasad , R. , and Katiyar , S.K. Therapeutic intervention of silymarin on the migration of non-small cell lung cancer cells is associated with the axis of multiple molecular targets including class 1 HDACs, ZEB1 expression, and restoration of miR-203 and E-cadherin expression. Am J Cancer Res 6 , 1287 1301 ( 2016)
[371]
Chtourou , Y. , Trabelsi , K. , Fetoui , H. , Mkannez , G. , Kallel , H. , and Zeghal , N. Manganese induces oxidative stress, redox state unbalance and disrupts membrane bound ATPases on murine neuroblastoma cells in vitro: protective role of silymarin. Neurochem Res 36 , 1546 1557 ( 2011) DOI: 10.1007/s11064-011-0483-5
[372]
Fan , L. , Ma , Y. , Liu , Y. , Zheng , D. , and Huang , G. Silymarin induces cell cycle arrest and apoptosis in ovarian cancer cells. Eur J Pharmacol 743 , 79 88 ( 2014) DOI: 10.1016/j.ejphar.2014.09.019
[373]
Invernizzi , R. , Bernuzzi , S. , Ciani , D. , and Ascari , E. Silymarine during maintenance therapy of acute promyelocytic leukemia. Haematologica 78 , 340 341 ( 1993)
[374]
Snima , K.S. , Arunkumar , P. , Jayakumar , R. , and Lakshmanan , V.K. Silymarin encapsulated poly(D,L-lactic-co-glycolic acid) nanoparticles: a prospective candidate for prostate cancer therapy. J Biomed Nanotechnol 10 , 559 570 ( 2014) DOI: 10.1166/jbn.2014.1735
[375]
Tyagi , A. , Raina , K. , Singh , R.P. , Gu , M. , Agarwal , C. , Harrison , G. , Glode , L.M. , and Agarwal , R. Chemopreventive effects of silymarin and silibinin on N-butyl-N-(4-hydroxybutyl) nitrosamine induced urinary bladder carcinogenesis in male ICR mice. Mol Cancer Ther 6 , 3248 3255 ( 2007) DOI: 10.1158/1535-7163.MCT-07-2006
[376]
Alday , E. , Valencia , D. , Carreno , A.L. , Picerno , P. , Piccinelli , A.L. , Rastrelli , L. , Robles-Zepeda , R. , Hernandez , J. , and Velazquez , C. Apoptotic induction by pinobanksin and some of its ester derivatives from Sonoran propolis in a B-cell lymphoma cell line. Chem Biol Interact 242 , 35 44 ( 2015) DOI: 10.1016/j.cbi.2015.09.013
[377]
Sanchez , Y. , Amran , D. , De Blas , E. , and Aller , P. Regulation of genistein-induced differentiation in human acute myeloid leukaemia cells (HL60, NB4) Protein kinase modulation and reactive oxygen species generation. Biochem Pharmacol 77 , 384 396 ( 2009) DOI: 10.1016/j.bcp.2008.10.035
[378]
Pons , D.G. , Nadal-Serrano , M. , Blanquer-Rossello , M.M. , Sastre-Serra , J. , Oliver , J. , and Roca , P. Genistein modulates proliferation and mitochondrial functionality in breast cancer cells depending on ERalpha/ERbeta ratio. J Cell Biochem 115 , 949 958 ( 2014) DOI: 10.1002/jcb.24737
[379]
Zhang , H. , Liu , G. , Zeng , X. , Wu , Y. , Yang , C. , Mei , L. , Wang , Z. , and Huang , L. Fabrication of genistein-loaded biodegradable TPGS-b-PCL nanoparticles for improved therapeutic effects in cervical cancer cells. Int J Nanomedicine 10 , 2461 2473 ( 2015) DOI: 10.2147/IJN.S78988
[380]
Lepri , S.R. , Zanelatto , L.C. , Da Silva , P.B. , Sartori , D. , Ribeiro , L.R. , and Mantovani , M.S. Effects of genistein and daidzein on cell proliferation kinetics in HT29 colon cancer cells: the expression of CTNNBIP1 (beta-catenin), APC (adenomatous polyposis coli) and BIRC5 (survivin). Hum Cell 27 , 78 84 ( 2014) DOI: 10.1007/s13577-012-0051-6
[381]
Qin , J. , Chen , J.X. , Zhu , Z. , and Teng , J.A. Genistein inhibits human colorectal cancer growth and suppresses miR-95, Akt and SGK1. Cell Physiol Biochem 35 , 2069 2077 ( 2015) DOI: 10.1159/000374013
[382]
Fang , M.Z. , Jin , Z. , Wang , Y. , Liao , J. , Yang , G.Y. , Wang , L.D. , and Yang , C.S. Promoter hypermethylation and inactivation of O(6)-methylguanine-DNA methyltransferase in esophageal squamous cell carcinomas and its reactivation in cell lines. Int J Oncol 26 , 615 622 ( 2005) DOI: 10.3892/ijo.26.3.615
[383]
Huang , W. , Wan , C. , Luo , Q. , Huang , Z. , and Luo , Q. Genistein-inhibited cancer stem cell-like properties and reduced chemoresistance of gastric cancer. Int J Mol Sci 15 , 3432 3443 ( 2014) DOI: 10.3390/ijms15033432
[384]
Liu , X. , Liu , K. , Qin , J. , Hao , L. , Li , X. , Liu , Y. , Zhang , X. , Liu , X. , Li , P. , Han , S. , Mao , Z. , and Shen , L. C/EBPbeta promotes angiogenesis through secretion of IL-6, which is inhibited by genistein, in EGFRvIII-positive glioblastoma. Int J Cancer 136 , 2524 2534 ( 2015) DOI: 10.1002/ijc.29319
[385]
Li , S. , Li , J. , Dai , W. , Zhang , Q. , Feng , J. , Wu , L. , Liu , T. , Yu , Q. , Xu , S. , Wang , W. , Lu , X. , Chen , K. , Xia , Y. , Lu , J. , Zhou , Y. , Fan , X. , Mo , W. , Xu , L. , and Guo , C. Genistein suppresses aerobic glycolysis and induces hepatocellular carcinoma cell death. Br J Cancer 117 , 1518 1528 ( 2017) DOI: 10.1038/bjc.2017.323
[386]
Kim , I.G. , Kim , J.S. , Lee , J.H. , and Cho , E.W. Genistein decreases cellular redox potential, partially suppresses cell growth in HL60 leukemia cells and sensitizes cells to gammaradiationinduced cell death. Mol Med Rep 10 , 2786 2792 ( 2014) DOI: 10.3892/mmr.2014.2611
[387]
Cheng , J. , Qi , J. , Li , X.T. , Zhou , K. , Xu , J.H. , Zhou , Y. , Zhang , G.Q. , Xu , J.P. , and Zhou , R.J. ATRA and Genistein synergistically inhibit the metastatic potential of human lung adenocarcinoma cells. Int J Clin Exp Med 8 , 4220 4227 ( 2015)
[388]
Danciu , C. , Borcan , F. , Bojin , F. , Zupko , I. , and Dehelean , C. Effect of the isoflavone genistein on tumor size, metastasis potential and melanization in a B16 mouse model of murine melanoma. Nat Prod Commun 8 , 343 346 ( 2013)
[389]
Park , S.J. , Kim , M.J. , Kim , Y.K. , Kim , S.M. , Park , J.Y. , and Myoung , H. Combined cetuximab and genistein treatment shows additive anti-cancer effect on oral squamous cell carcinoma. Cancer Lett 292 , 54 63 ( 2010) DOI: 10.1016/j.canlet.2009.11.004
[390]
Song , M. , Tian , X. , Lu , M. , Zhang , X. , Ma , K. , Lv , Z. , Wang , Z. , Hu , Y. , Xun , C. , Zhang , Z. , and Wang , S. Genistein exerts growth inhibition on human osteosarcoma MG-63 cells via PPARgamma pathway. Int J Oncol 46 , 1131 1140 ( 2015) DOI: 10.3892/ijo.2015.2829
[391]
Arzuman , L. , Beale , P. , Proschogo , N. , Yu , J.Q. , and Huq , F. Combination of genistein and cisplatin with two designed monofunctional platinum agents in human ovarian tumour models. Anticancer Res 35 , 6027 6039 ( 2015)
[392]
Xia , J. , Cheng , L. , Mei , C. , Ma , J. , Shi , Y. , Zeng , F. , Wang , Z. , and Wang , Z. Genistein inhibits cell growth and invasion through regulation of miR-27a in pancreatic cancer cells. Curr Pharm Des 20 , 5348 5353 ( 2014) DOI: 10.2174/1381612820666140128215756
[393]
Hirata , H. , Hinoda , Y. , Shahryari , V. , Deng , G. , Tanaka , Y. , Tabatabai , Z.L. , and Dahiya , R. Genistein downregulates onco-miR-1260b and upregulates sFRP1 and Smad4 via demethylation and histone modification in prostate cancer cells. Br J Cancer 110 , 1645 1654 ( 2014) DOI: 10.1038/bjc.2014.48
[394]
Yeh , C.C. , Fan , Y. , Jiang , L. , Yang , Y.L. , He , B. , You , L. , and Mann , M. Genistein suppresses growth of human uterine sarcoma cell lines via multiple mechanisms. Anticancer Res 35 , 3167 3173 ( 2015)
[395]
Messing , E. , Gee , J.R. , Saltzstein , D.R. , Kim , K. , Disant’agnese , A. , Kolesar , J. , Harris , L. , Faerber , A. , Havighurst , T. , Young , J.M. , Efros , M. , Getzenberg , R.H. , Wheeler , M.A. , Tangrea , J. , Parnes , H. , House , M. , Busby , J.E. , Hohl , R. , and Bailey , H. A phase 2 cancer chemoprevention biomarker trial of isoflavone G-2535 (genistein) in presurgical bladder cancer patients. Cancer Prev Res (Phila) 5 , 621 630 ( 2012) DOI: 10.1158/1940-6207.CAPR-11-0455
[396]
Chen , J. , Ge , B. , Wang , Y. , Ye , Y. , Zeng , S. , and Huang , Z. Biochanin A promotes proliferation that involves a feedback loop of microRNA-375 and estrogen receptor alpha in breast cancer cells. Cell Physiol Biochem 35, 639-646. ( 2015) DOI: 10.1159/000369725
[397]
Puthli , A. , Tiwari , R. , and Mishra , K.P. Biochanin A enhances the radiotoxicity in colon tumor cells in vitro. J Environ Pathol Toxicol Oncol 32 , 189 203 ( 2013) DOI: 10.1615/JEnvironPatholToxicolOncol.2013007280
[398]
Jain , A. , Lai , J.C. , and Bhushan , A. Biochanin A inhibits endothelial cell functions and proangiogenic pathways: implications in glioma therapy. Anti-cancer Drugs 26 , 323 330 ( 2015) DOI: 10.1097/CAD.0000000000000189
[399]
Youssef , M.M. , Tolba , M.F. , Badawy , N.N. , Liu , A.W. , El-Ahwany , E. , Khalifa , A.E. , Zada , S. , and Abdel-Naim , A.B. Novel combination of sorafenib and biochanin-A synergistically enhances the anti-proliferative and pro-apoptotic effects on hepatocellular carcinoma cells. Sci Rep 6 , 30717 ( 2016) DOI: 10.1038/srep30717
[400]
Chung , M.J. , Sohng , J.K. , Choi , D.J. , and Park , Y.I. Inhibitory effect of phloretin and biochanin A on IgE-mediated allergic responses in rat basophilic leukemia RBL-2H3 cells. Life Sci 93 , 401 408 ( 2013) DOI: 10.1016/j.lfs.2013.07.019
[401]
Lee , Y.S. , Seo , J.S. , Chung , H.T. , and Jang , J.J. Inhibitory effects of biochanin A on mouse lung tumor induced by benzo(a)pyrene. J Korean Med Sci 6 , 325 328 ( 1991) DOI: 10.3346/jkms.1991.6.4.325
[402]
Szliszka , E. , Czuba , Z.P. , Mertas , A. , Paradysz , A. , and Krol , W. The dietary isoflavone biochanin-A sensitizes prostate cancer cells to TRAIL-induced apoptosis. Urol Oncol 31 , 331 342 ( 2013) DOI: 10.1016/j.urolonc.2011.01.019
[403]
Zhang , B. , Su , J.P. , Bai , Y. , Li , J. , and Liu , Y.H. Inhibitory effects of O-methylated isoflavone glycitein on human breast cancer SKBR-3 cells. Int J Clin Exp Pathol 8 , 7809 7817 ( 2015)
[404]
Auyeung , K.K. , Law , P.C. , and Ko , J.K. Novel anti-angiogenic effects of formononetin in human colon cancer cells and tumor xenograft. Oncol Rep 28 , 2188 2194 ( 2012) DOI: 10.3892/or.2012.2056
[405]
Jin , Y.M. , Xu , T.M. , Zhao , Y.H. , Wang , Y.C. , and Cui , M.H. In vitro and in vivo anti-cancer activity of formononetin on human cervical cancer cell line HeLa. Tumour Biol 35 , 2279 2284 ( 2014) DOI: 10.1007/s13277-013-1302-1
[406]
Liu , Y. , He , J. , Chen , X. , Li , J. , Shen , M. , Yu , W. , Yang , Y. , and Xiao , Z. The proapoptotic effect of formononetin in human osteosarcoma cells: involvement of inactivation of ERK and Akt pathways. Cell Physiol Biochem 34 , 637 645 ( 2014) DOI: 10.1159/000363029
[407]
Zhang , X. , Bi , L. , Ye , Y. , and Chen , J. Formononetin induces apoptosis in PC-3 prostate cancer cells through enhancing the Bax/Bcl-2 ratios and regulating the p38/Akt pathway. Nutr Cancer 66 , 656 661 ( 2014) DOI: 10.1080/01635581.2014.894098
[408]
Yang , Y. , Zhao , Y. , Ai , X. , Cheng , B. , and Lu , S. Formononetin suppresses the proliferation of human non-small cell lung cancer through induction of cell cycle arrest and apoptosis. Int J Clin Exp Pathol 7 , 8453 8461 ( 2014)
[409]
Liu , Q. , Sun , Y. , Zheng , J.M. , Yan , X.L. , Chen , H.M. , Chen , J.K. , and Huang , H.Q. Formononetin sensitizes glioma cells to doxorubicin through preventing EMT via inhibition of histone deacetylase 5. Int J Clin Exp Pathol 8 , 6434 6441 ( 2015)
[410]
Hu , Y.C. , Wu , C.T. , Lai , J.N. , and Tsai , Y.T. Detection of a negative correlation between prescription of Chinese herbal products containing coumestrol, genistein or daidzein and risk of subsequent endometrial cancer among tamoxifen-treated female breast cancer survivors in Taiwan between 1998 and 2008: A population-based study. J Ethnopharmacol 169 , 356 362 ( 2015) DOI: 10.1016/j.jep.2015.04.028
[411]
Park , H.J. , Jeon , Y.K. , You , D.H. , and Nam , M.J. Daidzein causes cytochrome c-mediated apoptosis via the Bcl-2 family in human hepatic cancer cells. Food Chem Toxicol 60 , 542 549 ( 2013) DOI: 10.1016/j.fct.2013.08.022
[412]
Miyazaki , K. Novel approach for evaluation of estrogenic and anti-estrogenic activities of genistein and daidzein using B16 melanoma cells and dendricity assay. Pigment Cell Res 17 , 407 412 ( 2004) DOI: 10.1111/j.1600-0749.2004.00167.x
[413]
Lo , F.H. , Mak , N.K. , and Leung , K.N. Studies on the anti-tumor activities of the soy isoflavone daidzein on murine neuroblastoma cells. Biomed Pharmacother 61 , 591 595 ( 2007) DOI: 10.1016/j.biopha.2007.08.021
[414]
Guo , J.M. , Xiao , B.X. , Dai , D.J. , Liu , Q. , and Ma , H.H. Effects of daidzein on estrogen-receptor-positive and negative pancreatic cancer cells in vitro. World J Gastroenterol 10 , 860 863 ( 2004) DOI: 10.3748/wjg.v10.i6.860
[415]
Adjakly , M. , Ngollo , M. , Boiteux , J.P. , Bignon , Y.J. , Guy , L. , and Bernard-Gallon , D. Genistein and daidzein: different molecular effects on prostate cancer. Anticancer Res 33 , 39 44 ( 2013)
[416]
Lee , D.E. , Lee , K.W. , Byun , S. , Jung , S.K. , Song , N. , Lim , S.H. , Heo , Y.S. , Kim , J.E. , Kang , N.J. , Kim , B.Y. , Bowden , G.T. , Bode , A.M. , Lee , H.J. , and Dong , Z. . 7,3’,4’-Trihydroxyisoflavone, a metabolite of the soy isoflavone daidzein, suppresses ultraviolet B-induced skin cancer by targeting Cot and MKK4. J Biol Chem 286 , 14246 14256 ( 2011) DOI: 10.1074/jbc.M110.147348
[417]
Renno , W.M. , Al-Maghrebi , M. , Rao , M.S. , and Khraishah , H. (-)-Epigallocatechin-3-gallate modulates spinal cord neuronal degeneration by enhancing growth-associated protein 43, B-cell lymphoma 2, and decreasing B-cell lymphoma 2-associated x protein expression after sciatic nerve crush injury. J Neurotrauma 32 , 170 184 ( 2015) DOI: 10.1089/neu.2014.3491
[418]
Luo , J. , Gao , Y.T. , Chow , W.H. , Shu , X.O. , Li , H. , Yang , G. , Cai , Q. , Rothman , N. , Cai , H. , Shrubsole , M.J. , Franke , A.A. , Zheng , W. , and Dai , Q. Urinary polyphenols and breast cancer risk: results from the Shanghai Women’s Health Study. Breast Cancer Res Treat 120 , 693 702 ( 2010) DOI: 10.1007/s10549-009-0487-x
[419]
Chen , Z. , Yu , T. , Zhou , B. , Wei , J. , Fang , Y. , Lu , J. , Guo , L. , Chen , W. , Liu , Z.P. , and Luo , J. Mg(II)-Catechin nanoparticles delivering siRNA targeting EIF5A2 inhibit bladder cancer cell growth in vitro and in vivo. Biomaterials 81 , 125 134 ( 2016) DOI: 10.1016/j.biomaterials.2015.11.022
[420]
Al-Hazzani , A.A. , and Alshatwi , A.A. Catechin hydrate inhibits proliferation and mediates apoptosis of SiHa human cervical cancer cells. Food Chem Toxicol 49 , 3281 3286 ( 2011) DOI: 10.1016/j.fct.2011.09.023
[421]
Shervington , A. , Pawar , V. , Menon , S. , Thakkar , D. , and Patel , R. The sensitization of glioma cells to cisplatin and tamoxifen by the use of catechin. Mol Biol Rep 36 , 1181 1186 ( 2009) DOI: 10.1007/s11033-008-9295-3
[422]
Butler , L.M. , Huang , J.Y. , Wang , R. , Lee , M.J. , Yang , C.S. , Gao , Y.T. , and Yuan , J.M. Urinary biomarkers of catechins and risk of hepatocellular carcinoma in the Shanghai Cohort Study. Am J Epidemiol 181 , 397 405 ( 2015) DOI: 10.1093/aje/kwu304
[423]
Yoon , J.W. , Lee , J.S. , Kim , B.M. , Ahn , J. , and Yang , K.M. Catechin-7-O-xyloside induces apoptosis via endoplasmic reticulum stress and mitochondrial dysfunction in human non-small cell lung carcinoma H1299 cells. Oncol Rep 31 , 314 320 ( 2014) DOI: 10.3892/or.2013.2840
[424]
Di Leo , N. , Battaglini , M. , Berger , L. , Giannaccini , M. , Dente , L. , Hampel , S. , Vittorio , O. , Cirillo , G. , and Raffa , V. A catechin nanoformulation inhibits WM266 melanoma cell proliferation, migration and associated neo-angiogenesis. Eur J Pharm Biopharm 114 , 1 10 ( 2017) DOI: 10.1016/j.ejpb.2016.12.024
[425]
Appari , M. , Babu , K.R. , Kaczorowski , A. , Gross , W. , and Herr , I. Sulforaphane, quercetin and catechins complement each other in elimination of advanced pancreatic cancer by miR-let-7 induction and K-ras inhibition. Int J Oncol 45 , 1391 1400 ( 2014) DOI: 10.3892/ijo.2014.2539
[426]
Zhang , L. , Chen , Q.S. , Xu , P.P. , Qian , Y. , Wang , A.H. , Xiao , D. , Zhao , Y. , Sheng , Y. , Wen , X.Q. , and Zhao , W.L. Catechins induced acute promyelocytic leukemia cell apoptosis and triggered PML-RARalpha oncoprotein degradation. J Hematol Oncol 7 , 75 ( 2014) DOI: 10.1186/s13045-014-0075-3
[427]
Kumar , N.B. , Pow-Sang , J. , Egan , K.M. , Spiess , P.E. , Dickinson , S. , Salup , R. , Helal , M. , Mclarty , J. , Williams , C.R. , Schreiber , F. , Parnes , H.L. , Sebti , S. , Kazi , A. , Kang , L. , Quinn , G. , Smith , T. , Yue , B. , Diaz , K. , Chornokur , G. , Crocker , T. , and Schell , M.J. Randomized, placebo-controlled trial of green tea catechins for prostate cancer prevention. Cancer Prev Res (Phila) 8 , 879 887 ( 2015) DOI: 10.1158/1940-6207.CAPR-14-0324
[428]
Britschgi , A. , Simon , H.U. , Tobler , A. , Fey , M.F. , and Tschan , M.P. Epigallocatechin-3-gallate induces cell death in acute myeloid leukaemia cells and supports all-trans retinoic acid-induced neutrophil differentiation via death-associated protein kinase 2. Br J Haematol 149 , 55 64 ( 2010) DOI: 10.1111/j.1365-2141.2009.08040.x
[429]
Chen , N.G. , Lu , C.C. , Lin , Y.H. , Shen , W.C. , Lai , C.H. , Ho , Y.J. , Chung , J.G. , Lin , T.H. , Lin , Y.C. , and Yang , J.S. Proteomic approaches to study epigallocatechin gallate-provoked apoptosis of TSGH-8301 human urinary bladder carcinoma cells: roles of AKT and heat shock protein 27-modulated intrinsic apoptotic pathways. Oncol Rep 26 , 939 947 ( 2011) DOI: 10.3892/or.2011.1377
[430]
Mineva , N.D. , Paulson , K.E. , Naber , S.P. , Yee , A.S. , and Sonenshein , G.E. Epigallocatechin-3-gallate inhibits stem-like inflammatory breast cancer cells. PLoS One 8 , e73464 ( 2013) DOI: 10.1371/journal.pone.0073464
[431]
Khan , M.A. , Hussain , A. , Sundaram , M.K. , Alalami , U. , Gunasekera , D. , Ramesh , L. , Hamza , A. , and Quraishi , U. (-)-Epigallocatechin-3-gallate reverses the expression of various tumor-suppressor genes by inhibiting DNA methyltransferases and histone deacetylases in human cervical cancer cells. Oncol Rep 33 , 1976 1984 ( 2015) DOI: 10.3892/or.2015.3802
[432]
Lemanne , D. , Block , K.I. , Kressel , B.R. , Sukhatme , V.P. , and White , J.D. A case of complete and durable molecular remission of chronic lymphocytic leukemia following treatment with epigallocatechin-3-gallate, an extract of green tea. Cureus 7 , e441 ( 2015) DOI: 10.7759/cureus.441
[433]
Moseley , V.R. , Morris , J. , Knackstedt , R.W. , and Wargovich , M.J. Green tea polyphenol epigallocatechin 3-gallate, contributes to the degradation of DNMT3A and HDAC3 in HCT 116 human colon cancer cells. Anticancer Res 33 , 5325 5333 ( 2013)
[434]
D’angelo , L. , Piazzi , G. , Pacilli , A. , Prossomariti , A. , Fazio , C. , Montanaro , L. , Graziani , G. , Fogliano , V. , Munarini , A. , Bianchi , F. , Belluzzi , A. , Bazzoli , F. , and Ricciardiello , L. A combination of eicosapentaenoic acid-free fatty acid, epigallocatechin-3-gallate and proanthocyanidins has a strong effect on mTOR signaling in colorectal cancer cells. Carcinogenesis 35 , 2314 2320 ( 2014) DOI: 10.1093/carcin/bgu173
[435]
Liu , L. , Hou , L. , Gu , S. , Zuo , X. , Meng , D. , Luo , M. , Zhang , X. , Huang , S. , and Zhao , X. Molecular mechanism of epigallocatechin-3-gallate in human esophageal squamous cell carcinoma in vitro and in vivo. Oncol Rep 33 , 297 303 ( 2015) DOI: 10.3892/or.2014.3555
[436]
Ma , J. , Shi , M. , Li , G. , Wang , N. , Wei , J. , Wang , T. , Ma , J. , and Wang , Y. Regulation of Id1 expression by epigallocatechin3gallate and its effect on the proliferation and apoptosis of poorly differentiated AGS gastric cancer cells. Int J Oncol 43 , 1052 1058 ( 2013) DOI: 10.3892/ijo.2013.2043
[437]
Chen , T.C. , Wang , W. , Golden , E.B. , Thomas , S. , Sivakumar , W. , Hofman , F.M. , Louie , S.G. , and Schonthal , A.H. Green tea epigallocatechin gallate enhances therapeutic efficacy of temozolomide in orthotopic mouse glioblastoma models. Cancer Lett 302 , 100 108 ( 2011) DOI: 10.1016/j.canlet.2010.11.008
[438]
Li , H. , Li , Z. , Xu , Y.M. , Wu , Y. , Yu , K.K. , Zhang , C. , Ji , Y.H. , Ding , G. , and Chen , F.X. Epigallocatechin-3-gallate induces apoptosis, inhibits proliferation and decreases invasion of glioma cell. Neurosci Bull 30 , 67 73 ( 2014) DOI: 10.1007/s12264-013-1394-z
[439]
Shen , X. , Zhang , Y. , Feng , Y. , Zhang , L. , Li , J. , Xie , Y.A. , and Luo , X. Epigallocatechin-3-gallate inhibits cell growth, induces apoptosis and causes S phase arrest in hepatocellular carcinoma by suppressing the AKT pathway. Int J Oncol 44 , 791 796 ( 2014) DOI: 10.3892/ijo.2014.2251
[440]
Shi , J. , Liu , F. , Zhang , W. , Liu , X. , Lin , B. , and Tang , X. Epigallocatechin-3-gallate inhibits nicotine-induced migration and invasion by the suppression of angiogenesis and epithelial-mesenchymal transition in non-small cell lung cancer cells. Oncol Rep 33 , 2972 2980 ( 2015) DOI: 10.3892/or.2015.3889
[441]
Siddiqui , I.A. , Bharali , D.J. , Nihal , M. , Adhami , V.M. , Khan , N. , Chamcheu , J.C. , Khan , M.I. , Shabana , S. , Mousa , S.A. , and Mukhtar , H. Excellent anti-proliferative and pro-apoptotic effects of (-)-epigallocatechin-3-gallate encapsulated in chitosan nanoparticles on human melanoma cell growth both in vitro and in vivo. Nanomedicine 10 , 1619 1626 ( 2014) DOI: 10.1016/j.nano.2014.05.007
[442]
Nishimura , N. , Hartomo , T.B. , Pham , T.V. , Lee , M.J. , Yamamoto , T. , Morikawa , S. , Hasegawa , D. , Takeda , H. , Kawasaki , K. , Kosaka , Y. , Yamamoto , N. , Kubokawa , I. , Mori , T. , Yanai , T. , Hayakawa , A. , Takeshima , Y. , Iijima , K. , Matsuo , M. , and Nishio , H. Epigallocatechin gallate inhibits sphere formation of neuroblastoma BE(2)-C cells. Environ Health Prev Med 17 , 246 251 ( 2012) DOI: 10.1007/s12199-011-0239-5
[443]
Lee , J.C. , Chung , L.C. , Chen , Y.J. , Feng , T.H. , Chen , W.T. , and Juang , H.H. Upregulation of B-cell translocation gene 2 by epigallocatechin-3-gallate via p38 and ERK signaling blocks cell proliferation in human oral squamous cell carcinoma cells. Cancer Lett 360 , 310 318 ( 2015) DOI: 10.1016/j.canlet.2015.02.034
[444]
Tang , G. , Zhang , Z. , Qian , H. , Chen , J. , Wang , Y. , Chen , X. , Chen , B. , and Chen , Y. (-)-Epigallocatechin-3-gallate inhibits osteosarcoma cell invasiveness by inhibiting the MEK/ERK signaling pathway in human osteosarcoma cells. J Environ Pathol Toxicol Oncol 34, 85 93 ( 2015) DOI: 10.1615/JEnvironPatholToxicolOncol.2015011925
[445]
Harpole , J.L. , Tucci , M. , and Benghuzzi , H. Pathophysiological effects of thymoquinone and epigallocatechin-3-gallate on SK-OV-3 ovarian cancer like cell line. Biomed Sci Instrum 51 , 31 39 ( 2015)
[446]
Bimonte , S. , Leongito , M. , Barbieri , A. , Del Vecchio , V. , Barbieri , M. , Albino , V. , Piccirillo , M. , Amore , A. , Di Giacomo , R. , Nasto , A. , Granata , V. , Petrillo , A. , Arra , C. , and Izzo , F. Inhibitory effect of (-)-epigallocatechin-3-gallate and bleomycin on human pancreatic cancer MiaPaca-2 cell growth. Infect Agent Cancer 10 , 22 ( 2015) DOI: 10.1186/s13027-015-0016-y
[447]
Mukherjee , S. , Siddiqui , M.A. , Dayal , S. , Ayoub , Y.Z. , and Malathi , K. Epigallocatechin-3-gallate suppresses proinflammatory cytokines and chemokines induced by Toll-like receptor 9 agonists in prostate cancer cells. J Inflamm Res 7 , 89 101 ( 2014) DOI: 10.2147/JIR.S61365
[448]
Singh , T. , and Katiyar , S.K. Green tea polyphenol, (-)-epigallocatechin-3-gallate, induces toxicity in human skin cancer cells by targeting beta-catenin signaling. Toxicol Appl Pharmacol 273 , 418 424 ( 2013) DOI: 10.1016/j.taap.2013.09.021
[449]
De Amicis , F. , Perri , A. , Vizza , D. , Russo , A. , Panno , M.L. , Bonofiglio , D. , Giordano , C. , Mauro , L. , Aquila , S. , Tramontano , D. , and Ando , S. Epigallocatechin gallate inhibits growth and epithelial-to-mesenchymal transition in human thyroid carcinoma cell lines. J Cell Physiol 228 , 2054 2062 ( 2013) DOI: 10.1002/jcp.24372
[450]
Sun , Y. , Wang , H. , Lin , F. , Hua , J. , and Zhou , G. Inhibition of proliferation and gene expression regulation by (-)-epigallocatechin-3-gallate in human synovial sarcoma cells. Med Oncol 28 , 1463 1468 ( 2011) DOI: 10.1007/s12032-010-9560-x
[451]
Wang , L. , Li , H. , Yang , S. , Ma , W. , Liu , M. , Guo , S. , Zhan , J. , Zhang , H. , Tsang , S.Y. , Zhang , Z. , Wang , Z. , Li , X. , Guo , Y.D. , and Li , X. Cyanidin-3-o-glucoside directly binds to ERalpha36 and inhibits EGFR-positive triple-negative breast cancer. Oncotarget 7 , 68864 68882 ( 2016) DOI: 10.18632/oncotarget.12025
[452]
Cvorovic , J. , Tramer , F. , Granzotto , M. , Candussio , L. , Decorti , G. , and Passamonti , S. Oxidative stress-based cytotoxicity of delphinidin and cyanidin in colon cancer cells. Arch Biochem Biophys 501 , 151 157 ( 2010) DOI: 10.1016/j.abb.2010.05.019
[453]
Chen , P.N. , Chu , S.C. , Chiou , H.L. , Kuo , W.H. , Chiang , C.L. , and Hsieh , Y.S. Mulberry anthocyanins, cyanidin 3-rutinoside and cyanidin 3-glucoside, exhibited an inhibitory effect on the migration and invasion of a human lung cancer cell line. Cancer Lett 235 , 248 259 ( 2006) DOI: 10.1016/j.canlet.2005.04.033
[454]
Zeng , L. , Gao , J. , and Zhang , R. Study on anti-tumor effect of cyanidin-3-glucoside on ovarian cancer. Zhongguo Zhong Yao Za Zhi 37 , 1651 1654 ( 2012) DOI: 10.4268/cjcmm20121131
[455]
Sorrenti , V. , Vanella , L. , Acquaviva , R. , Cardile , V. , Giofre , S. , and Di Giacomo , C. Cyanidin induces apoptosis and differentiation in prostate cancer cells. Int J Oncol 47 , 1303 1310 ( 2015) DOI: 10.3892/ijo.2015.3130
[456]
Im , N.K. , Jang , W.J. , Jeong , C.H. , and Jeong , G.S. Delphinidin suppresses PMA-induced MMP-9 expression by blocking the NF-kappaB activation through MAPK signaling pathways in MCF-7 human breast carcinoma cells. J Med Food 17 , 855 861 ( 2014) DOI: 10.1089/jmf.2013.3077
[457]
Chakrabarti , M. , and Ray , S.K. Direct transfection of miR-137 mimics is more effective than DNA demethylation of miR-137 promoter to augment anti-tumor mechanisms of delphinidin in human glioblastoma U87MG and LN18 cells. Gene 573 , 141 152 ( 2015) DOI: 10.1016/j.gene.2015.07.034
[458]
Feng , R. , Wang , S.Y. , Shi , Y.H. , Fan , J. , and Yin , X.M. Delphinidin induces necrosis in hepatocellular carcinoma cells in the presence of 3-methyladenine, an autophagy inhibitor. J Agric Food Chem 58 , 3957 3964 ( 2010) DOI: 10.1021/jf9025458
[459]
Pal , H.C. , Sharma , S. , Strickland , L.R. , Agarwal , J. , Athar , M. , Elmets , C.A. , and Afaq , F. Delphinidin reduces cell proliferation and induces apoptosis of non-small-cell lung cancer cells by targeting EGFR/VEGFR2 signaling pathways. PLoS One 8 , e77270 ( 2013) DOI: 10.1371/journal.pone.0077270
[460]
Lim , W. , Jeong , W. , and Song , G. Delphinidin suppresses proliferation and migration of human ovarian clear cell carcinoma cells through blocking AKT and ERK1/2 MAPK signaling pathways. Mol Cell Endocrinol 422 , 172 181 ( 2016) DOI: 10.1016/j.mce.2015.12.013
[461]
Jeong , M.H. , Ko , H. , Jeon , H. , Sung , G.J. , Park , S.Y. , Jun , W.J. , Lee , Y.H. , Lee , J. , Lee , S.W. , Yoon , H.G. , and Choi , K.C. Delphinidin induces apoptosis via cleaved HDAC3-mediated p53 acetylation and oligomerization in prostate cancer cells. Oncotarget 7 , 56767 56780 ( 2016) DOI: 10.18632/oncotarget.10790
[462]
Kamenickova , A. , Anzenbacherova , E. , Pavek , P. , Soshilov , A.A. , Denison , M.S. , Anzenbacher , P. , and Dvorak , Z. Pelargonidin activates the AhR and induces CYP1A1 in primary human hepatocytes and human cancer cell lines HepG2 and LS174T. Toxicol Lett 218 , 253 259 ( 2013) DOI: 10.1016/j.toxlet.2013.01.020
[463]
Lazzeroni , M. , Guerrieri-Gonzaga , A. , Gandini , S. , Johansson , H. , Serrano , D. , Cazzaniga , M. , Aristarco , V. , Puccio , A. , Mora , S. , Caldarella , P. , Pagani , G. , Pruneri , G. , Riva , A. , Petrangolini , G. , Morazzoni , P. , Decensi , A. , and Bonanni , B. A presurgical study of oral silybin-phosphatidylcholine in patients with early breast cancer. Cancer Prev Res (Phila) 9 , 89 95 ( 2016) DOI: 10.1158/1940-6207.CAPR-15-0123
[464]
Löhr , J.-M. , Karimi , M. , Omazic , B. , Kartalis , N. , Verbeke , C.S. , Berkenstam , A. , and Frödin , J.-E . A phase I dose escalation trial of AXP107-11, a novel multi-component crystalline form of genistein, in combination with gemcitabine in chemotherapy-naive patients with unresectable pancreatic cancer. Pancreatology 16 , 640 645 ( 2016) DOI: 10.1016/j.pan.2016.05.002
[465]
Lazarevic , B. , Boezelijn , G. , Diep , L.M. , Kvernrod , K. , Ogren , O. , Ramberg , H. , Moen , A. , Wessel , N. , Berg , R.E. , Egge-Jacobsen , W. , Hammarstrom , C. , Svindland , A. , Kucuk , O. , Saatcioglu , F. , Tasken , K.A. , and Karlsen , S.J. Efficacy and safety of short-term genistein intervention in patients with localized prostate cancer prior to radical prostatectomy: a randomized, placebo-controlled, double-blind Phase 2 clinical trial. Nutr Cancer 63 , 889 898 ( 2011) DOI: 10.1080/01635581.2011.582221
[466]
Lazarevic , B. , Hammarstrom , C. , Yang , J. , Ramberg , H. , Diep , L.M. , Karlsen , S.J. , Kucuk , O. , Saatcioglu , F. , Tasken , K.A. , and Svindland , A. The effects of short-term genistein intervention on prostate biomarker expression in patients with localised prostate cancer before radical prostatectomy. Br J Nutr 108 , 2138 2147 ( 2012) DOI: 10.1017/S0007114512000384
[467]
Shike , M. , Doane , A.S. , Russo , L. , Cabal , R. , Reis-Filho , J.S. , Gerald , W. , Cody , H. , Khanin , R. , Bromberg , J. , and Norton , L. The effects of soy supplementation on gene expression in breast cancer: a randomized placebo-controlled study. J Natl Cancer Inst 106 ( 2014) DOI: 10.1093/jnci/dju189
[468]
Hamilton-Reeves , J.M. , Banerjee , S. , Banerjee , S.K. , Holzbeierlein , J.M. , Thrasher , J.B. , Kambhampati , S. , Keighley , J. , and Van Veldhuizen , P. Short-term soy isoflavone intervention in patients with localized prostate cancer: a randomized, double-blind, placebo-controlled trial. PLoS One 8 , e68331 ( 2013) DOI: 10.1371/journal.pone.0068331
[469]
Miyanaga , N. , Akaza , H. , Hinotsu , S. , Fujioka , T. , Naito , S. , Namiki , M. , Takahashi , S. , Hirao , Y. , Horie , S. , Tsukamoto , T. , Mori , M. , and Tsuji , H. Prostate cancer chemoprevention study: an investigative randomized control study using purified isoflavones in men with rising prostate-specific antigen. Cancer Sci 103 , 125 130 ( 2012) DOI: 10.1111/j.1349-7006.2011.02120.x
[470]
Kumar , N.B. , Kang , L. , Pow-Sang , J. , Xu , P. , Allen , K. , Riccardi , D. , Besterman-Dahan , K. , and Krischer , J.P. Results of a randomized phase I dose-finding trial of several doses of isoflavones in men with localized prostate cancer: administration prior to radical prostatectomy. J Soc Integr Oncol 8 , 3 13 ( 2010)
[471]
El-Rayes , B.F. , Philip , P.A. , Sarkar , F.H. , Shields , A.F. , Ferris , A.M. , Hess , K. , Kaseb , A.O. , Javle , M.M. , Varadhachary , G.R. , Wolff , R.A. , and Abbruzzese , J.L. A phase II study of isoflavones, erlotinib, and gemcitabine in advanced pancreatic cancer. Invest New Drugs 29 , 694 699 ( 2011) DOI: 10.1007/s10637-010-9386-6
[472]
Atkinson , C. , Warren , R.M. , Sala , E. , Dowsett , M. , Dunning , A.M. , Healey , C.S. , Runswick , S. , Day , N.E. , and Bingham , S.A. Red-clover-derived isoflavones and mammographic breast density: a double-blind, randomized, placebo-controlled trial (ISRCTN42940165). Breast Cancer Res 6 , R170 179 ( 2004) DOI: 10.1186/bcr773
[473]
Nikander , E. , Kilkkinen , A. , Metsa-Heikkila , M. , Adlercreutz , H. , Pietinen , P. , Tiitinen , A. , and Ylikorkala , O. A randomized placebo-controlled crossover trial with phytoestrogens in treatment of menopause in breast cancer patients. Obstet Gynecol 101 , 1213 1220 ( 2003) DOI: 10.1016/S0029-7844(03)00232-1
[474]
Zhao , H. , Zhu , W. , Jia , L. , Sun , X. , Chen , G. , Zhao , X. , Li , X. , Meng , X. , Kong , L. , Xing , L. , and Yu , J. Phase I study of topical epigallocatechin-3-gallate (EGCG) in patients with breast cancer receiving adjuvant radiotherapy. Br J Radiol 89 , 20150665 ( 2016) DOI: 10.1259/bjr.20150665
[475]
Zhao , H. , Xie , P. , Li , X. , Zhu , W. , Sun , X. , Sun , X. , Chen , X. , Xing , L. , and Yu , J. A prospective phase II trial of EGCG in treatment of acute radiation-induced esophagitis for stage III lung cancer. Radiother Oncol 114 , 351 356 ( 2015) DOI: 10.1016/j.radonc.2015.02.014
[476]
Trudel , D. , Labbe , D.P. , Araya-Farias , M. , Doyen , A. , Bazinet , L. , Duchesne , T. , Plante , M. , Gregoire , J. , Renaud , M.C. , Bachvarov , D. , Tetu , B. , and Bairati , I. A two-stage, single-arm, phase II study of EGCG-enriched green tea drink as a maintenance therapy in women with advanced stage ovarian cancer. Gynecol Oncol 131 , 357 361 ( 2013) DOI: 10.1016/j.ygyno.2013.08.019
[477]
Samavat , H. , Dostal , A.M. , Wang , R. , Bedell , S. , Emory , T.H. , Ursin , G. , Torkelson , C.J. , Gross , M.D. , Le , C.T. , Yu , M.C. , Yang , C.S. , Yee , D. , Wu , A.H. , Yuan , J.M. , and Kurzer , M.S. The Minnesota Green Tea Trial (MGTT), a randomized controlled trial of the efficacy of green tea extract on biomarkers of breast cancer risk: study rationale, design, methods, and participant characteristics. Cancer Causes Control 26 , 1405 1419 ( 2015) DOI: 10.1007/s10552-015-0632-2
[478]
Henning , S.M. , Wang , P. , Said , J.W. , Huang , M. , Grogan , T. , Elashoff , D. , Carpenter , C.L. , Heber , D. , and Aronson , W.J. Randomized clinical trial of brewed green and black tea in men with prostate cancer prior to prostatectomy. Prostate 75 , 550 559 ( 2015) DOI: 10.1002/pros.22943
[479]
Garcia , F.A. , Cornelison , T. , Nuno , T. , Greenspan , D.L. , Byron , J.W. , Hsu , C.H. , Alberts , D.S. , and Chow , H.H. Results of a phase II randomized, double-blind, placebo-controlled trial of Polyphenon E in women with persistent high-risk HPV infection and low-grade cervical intraepithelial neoplasia. Gynecol Oncol 132 , 377 382 ( 2014) DOI: 10.1016/j.ygyno.2013.12.034
[480]
Shanafelt , T.D. , Call , T.G. , Zent , C.S. , Leis , J.F. , Laplant , B. , Bowen , D.A. , Roos , M. , Laumann , K. , Ghosh , A.K. , Lesnick , C. , Lee , M.J. , Yang , C.S. , Jelinek , D.F. , Erlichman , C. , and Kay , N.E. Phase 2 trial of daily, oral Polyphenon E in patients with asymptomatic, Rai stage 0 to II chronic lymphocytic leukemia. Cancer 119 , 363 370 ( 2013) DOI: 10.1002/cncr.27719
[481]
Nguyen , M.M. , Ahmann , F.R. , Nagle , R.B. , Hsu , C.H. , Tangrea , J.A. , Parnes , H.L. , Sokoloff , M.H. , Gretzer , M.B. , and Chow , H.H. Randomized, double-blind, placebo-controlled trial of polyphenon E in prostate cancer patients before prostatectomy: evaluation of potential chemopreventive activities. Cancer Prev Res (Phila) 5 , 290 298 ( 2012) DOI: 10.1158/1940-6207.CAPR-11-0306
[482]
Shanafelt , T.D. , Call , T.G. , Zent , C.S. , Laplant , B. , Bowen , D.A. , Roos , M. , Secreto , C.R. , Ghosh , A.K. , Kabat , B.F. , Lee , M.J. , Yang , C.S. , Jelinek , D.F. , Erlichman , C. , and Kay , N.E. Phase I trial of daily oral Polyphenon E in patients with asymptomatic Rai stage 0 to II chronic lymphocytic leukemia. J Clin Oncol 27 , 3808 3814 ( 2009) DOI: 10.1200/JCO.2008.21.1284
[483]
Cassaday , R.D. , Goy , A. , Advani , S. , Chawla , P. , Nachankar , R. , Gandhi , M. , and Gopal , A.K. A phase II, single-arm, open-label, multicenter study to evaluate the efficacy and safety of P276-00, a cyclin-dependent kinase inhibitor, in patients with relapsed or refractory mantle cell lymphoma. Clin Lymphoma Myeloma Leuk 15 , 392 397 ( 2015) DOI: 10.1016/j.clml.2015.02.021
[484]
Kulbacka , J. , Pucek , A. , Kotulska , M. , Dubinska-Magiera , M. , Rossowska , J. , Rols , M.P. , and Wilk , K.A. Electroporation and lipid nanoparticles with cyanine IR-780 and flavonoids as efficient vectors to enhanced drug delivery in colon cancer. Bioelectrochemistry 110 , 19 31 ( 2016) DOI: 10.1016/j.bioelechem.2016.02.013
[485]
Pham , J. , Brownlow , B. , and Elbayoumi , T. Mitochondria-specific pro-apoptotic activity of genistein lipidic nanocarriers. Mol Pharm 10 , 3789 3800 ( 2013) DOI: 10.1021/mp4004892
[486]
Kim , K.Y. , Jin , H. , Park , J. , Jung , S.H. , Lee , J.H. , Park , H. , Kim , S.K. , Bae , J. , and Jung , J.H. Mitochondria-targeting self-assembled nanoparticles derived from triphenylphosphonium-conjugated cyanostilbene enable site-specific imaging and anti-cancer drug delivery. Nano Research 11 , 1082 1098 ( 2017) DOI: 10.1007/s12274-017-1728-7
[487]
Jara , J.A. , Castro-Castillo , V. , Saavedra-Olavarria , J. , Peredo , L. , Pavanni , M. , Jana , F. , Letelier , M.E. , Parra , E. , Becker , M.I. , Morello , A. , Kemmerling , U. , Maya , J.D. , and Ferreira , J. Antiproliferative and uncoupling effects of delocalized, lipophilic, cationic gallic acid derivatives on cancer cell lines. Validation in vivo in singenic mice. J Med Chem 57 , 2440 2454 ( 2014) DOI: 10.1021/jm500174v
[488]
Ersoz , M. , Erdemir , A. , Duranoglu , D. , Uzunoglu , D. , Arasoglu , T. , Derman , S. , Mansuroglu , B. Comparative evaluation of hesperetin loaded nanoparticles for anticancer activity against C6 glioma cancer cells. Artif Cells Nanomed Biotechnol 47 ( 1 ), 319 329 ( 2019) DOI: 10.1080/21691401.2018.1556213
[489]
Rahmanian , N. , Hamishehkar , H. , Dolatabadi , J.E. , and Arsalani , N. Nano graphene oxide: a novel carrier for oral delivery of flavonoids. Colloids Surf B Biointerfaces 123 , 331 338 ( 2014) DOI: 10.1016/j.colsurfb.2014.09.036
[490]
Balakrishnan , S. , Mukherjee , S. , Das , S. , Bhat , F.A. , Raja Singh , P. , Patra , C.R. , and Arunakaran , J. Gold nanoparticles-conjugated quercetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated pathway in breast cancer cell lines (MCF-7 and MDA-MB-231). Cell Biochem Funct 35 , 217 231 ( 2017) DOI: 10.1002/cbf.3266
[491]
2016a Lee , D. , Ko , W.K. , Hwang , D.S. , Heo , D.N. , Lee , S.J. , Heo , M. , Lee , K.S. , Ahn , J.Y. , Jo , J. , and Kwon , I.K. Use of baicalin-conjugated gold nanoparticles for apoptotic induction of breast cancer cells. Nanoscale Res Lett 11 , 381 ( 2016a) DOI: 10.1186/s11671-016-1586-3
[492]
Kondath , S. , Srinivas Raghavan , B. , Anantanarayanan , R. , and Rajaram , R. Synthesis and characterisation of morin reduced gold nanoparticles and its cytotoxicity in MCF-7 cells. Chem Biol Interact 224 , 78 88 ( 2014) DOI: 10.1016/j.cbi.2014.09.025
[493]
Dhas , G. , Subramaniyan , J. , Subramani , P. , and Thiruvengadam , D. Nanochemopreventive effect of polymer functionalized gold nanoparticles containing hesperetin drug inhibited proliferation and induced apoptosis in Hep3B cells. JAPS 6 , 114 123 ( 2016) DOI: 10.7324/JAPS.2016.601216
[494]
Birinci , Y. , Niazi , J. , and Basaga , H. Enhancing antioxidant cellular defense by using quercetin loaded tio2 nanoparticles in swiss 3t3 albino mouse fibroblast cells. J Nanomedic Nanotechnol S8 , 1 10 ( 2017) DOI: 10.4172/2157-7439.S8-001
[495]
Kavithaa , K. , Paulpandi , M. , Padma , P.R. , and Sumathi , S. Induction of intrinsic apoptotic pathway and cell cycle arrest via baicalein loaded iron oxide nanoparticles as a competent nano-mediated system for triple negative breast cancer therapy. RSC Advances 6 , 64531 64543 ( 2016) DOI: 10.1039/C6RA11658B
[496]
Alpsoy , L. , Baykal , A. , and Ü . Akal , Z. Luteolin-loaded spion as a drug carrier for cancer cell in vitro. J Supercond Nov Magn 31 , 467 474 ( 2017) DOI: 10.1007/s10948-017-4199-x
Share
Back to top