You are on page 1of 19

Pharmacological Research 130 (2018) 273–291

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Review

Targeting oncogenic transcription factors by polyphenols: A novel


approach for cancer therapy
Chitra Rajagopal a , Manendra Babu Lankadasari a , Jesil Mathew Aranjani b ,
K.B. Harikumar a,∗
a
Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
b
Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India

a r t i c l e i n f o a b s t r a c t

Article history: Inflammation is one of the major causative factor of cancer and chronic inflammation is involved in all
Received 31 July 2017 the major steps of cancer initiation, progression metastasis and drug resistance. The molecular mecha-
Received in revised form nism of inflammation driven cancer is the complex interplay between oncogenic and tumor suppressive
30 November 2017
transcription factors which include FOXM1, NF-kB, STAT3, Wnt/␤- Catenin, HIF-1␣, NRF2, androgen and
Accepted 31 December 2017
estrogen receptors. Several products derived from natural sources modulate the expression and activ-
Available online 4 January 2018
ity of multiple transcription factors in various tumor models as evident from studies conducted in cell
lines, pre-clinical models and clinical samples. Further combination of these natural products along with
Chemical compounds studied in this article:
4- Shogaol (PubChem CID: 9794897)
currently approved cancer therapies added an additional advantage and they considered as promising
6- Shogaol (PubChem CID: 5281794) targets for prevention and treatment of inflammation and cancer. In this review we discuss the application
Apigenin (PubChem CID: 5280443) of multi-targeting natural products by analyzing the literature and future directions for their plausible
Berberine (PubChem CID: 2353) applications in drug discovery.
Bergamottin (PubChem CID: 5471349) © 2018 Elsevier Ltd. All rights reserved.
Caffeic acid (PubChem CID:
689043)Capsaicin (PubChem CID: 1548943)
Cardamonin (PubChem CID: 641785)
Casticin(PubChem CID: 5315263)
Chrysin (PubChem CID: 5281607)
Cryptotanshinone (PubChem CID: 160254)
Cucurbitacin (PubChem CID: 5281319)
Curcumin (PubChem CID: 969516)
EGCG (PubChem CID: 65064)
Ellagic acid (PubChem CID: 5281855)
Escin (PubChem CID: 16211024)
Fistein (PubChem CID: 5281614)
Gambogic acid (PubChem CID: 16072310)
Gingerol (PubChem CID: 442793)
Gossypin (PubChem CID: 5281621)
Honokiol (PubChem CID: 72303)
Luteolin (PubChem CID: 5280445)

Abbreviations: AIF, apoptosis-inducing factor; Akt/PKA, protein kinase A; AR, androgen receptors; Bax, Bcl-2-associated X protein; Bcl2, B-cell lymphoma2; BIG3, brefeldin
A-inhibited guanine nucleotide-exchange protein3; CCL2, chemokine ligand 2; Cdc25B, cell division cycle 25B; Cdk 2, cyclin-dependent kinase 2; Cdk4, cyclin-dependent
kinase 4; c-FLIP, cellular FLICE- inhibitory protein; COX2, cyclooxygenase 2; CXCR2, C-X-C chemokine receptor type 2; DNMT1, DNA methyltransferase 1; EMT, epithelial
to mesenchymal transition; ERK1, extracellular signal-regulated kinase-1; ER␣, estrogen receptor alpha; FAK, focal adhesion kinase; FGF, fibroblast growth factors; FOXM1,
forkhead box protein M1; Foxo3a, forkhead box O3; HIF-1␣, hypoxia inducing factor-1␣; hTERT, telomerase reverse transcriptase; IAP, the inhibitors of apoptosis protein;
ICAM-1, intercellular adhesion molecule 1; IGF-1R, insulin-like growth factor 1; IKK, inhibitory kappa B kinase; iNOS, inducible nitric oxide synthase; I␬B␣, inhibitory kappa
B alpha; JAK, janus kinase; JNK, c-Jun N-terminal kinase; LRP4, LDL receptor related protein; Mcl-1, myeloid leukemia cell differentiation protein; MDM2, mouse double
minute 2 homolog; MMP, matrix metalloproteinase; mToR, mammalian target of rapamycin; NF-␬B, nuclear factor-␬B; PAIS3, protein inhibitor of activated STAT protein
3; PARP, poly (ADP-ribose) polymerase; PCNA, proliferative cell nuclear antigen; PGE2, prostaglandin E2; PHB, prohibitin; PI3K, phosphoinositide 3-kinase; PSA, prostate-
specific antigen; ROS, Reactive oxygen species; SHP1, src homology region 2 domain-containing phosphatase-1; SOCS3, suppressor of cytokine signaling 3; STAT, signal
transducer and activator of transcription; TGF␤, transforming growth factor beta; TNF-␣, tumor necrosis factor alpha; uPA, urokinase-type plasminogen activator; VEGF,
vascular endothelial growth factor; XIAP, X-linked inhibitor of apoptosis protein.
∗ Corresponding author.
E-mail address: harikumar@rgcb.res.in (K.B. Harikumar).

https://doi.org/10.1016/j.phrs.2017.12.034
1043-6618/© 2018 Elsevier Ltd. All rights reserved.
274 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

Mangiferin (PubChem CID: 5281647)


Piperlongumine (PubChem CID:
637858)
Pterostilbene (PubChem CID: 5281727)
Quercetin (PubChem CID: 5280343)
Resveratrol (PubChem CID: 445154)
Rutin (PubChem CID: 5280805)
Sesamin (PubChem CID: 72307)
Ursolic acid (PubChem CID: 64945)
Wogonin (PubChem CID: 5281703)
Xanthohumol (PubChem CID: 639665)

Keywords:
Polyphenols
Cancer
Inflammation
Metastasis
Natural compounds
Transcription factors

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 274
2. Natural products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 274
3. Transcription factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276
3.1. FOXM1 (Forkhead Box M1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276
3.2. Wnt/␤-catenin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276
3.3. Nrf2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 277
3.4. STAT3 (Signal transducer and activator of transcription 3) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 279
3.5. AR (androgen receptors) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 280
3.6. ER (estrogen receptors) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281
3.7. HIF1 (hypoxia inducing factor 1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283
3.8. NF-␬B (nuclear factor kappaB) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285

1. Introduction prises of three main subclasses namely flavonoids, phenolic acids,


and stilbenoids which include hydroxybenzoic acids, hydroxycin-
Cancer is a multifactorial disease caused by dysregulation of namic acids, anthocyanins, proanthocyanidins, flavonols, flavones,
numerous gene products and alteration of multiple cell signal- flavanols, flavanones, isoflavones, stilbenes, lignansetc [5]. Its his-
ing pathways. The key reasons for more than 90% of cancer tory of evolution as major class of phytochemicals in the field
are attributed to somatic mutations and environmental factors. of therapeutics started from leather industry. Before 20th cen-
Another major factor is epigenetic changes and research in recent tury, polyphenols were named as vegetable tannins used in
years clearly demonstrated that DNA methylation and other type of the conversion of animal skin to leather. Later, its co-evolution
histone modifications can lead to alterations in chromatin conden- along with molecular chemistry leads to its establishment as an
sation status, thereby regulating the expression of specific genes. important category of biomolecules in the field of therapeutics.
Any kind of perturbation in the expression of these genes can lead Structural elucidation of polyphenols shows them as the polymers
to cellular transformation eventually leading to cancer [1]. Several of phenol groups, poly-hydroxylated phytochemicals. Mode of
epidemiological, pre-clinical and clinical studies over the last sev- extraction varies according to its physicochemical properties. Dif-
eral decades established the relationship between the process of ferent extraction methods include solvent extraction, microwave
inflammation and cancer. The acute inflammatory response has assisted extraction, ultrasound assisted extractions etc which is
therapeutic advantages however chronic inflammation is associ- followed by its colorimetric quantification [3,9]. Polyphenols are
ated with a myriad of diseases [2]. dominated for their antioxidant property but the recent research
showed their ability to bind to certain proteins directly leading
2. Natural products to physiological changes. Their antioxidant properties make them
usable against different diseases like cancer, diabetes, cardiovas-
Natural products can be defined as structurally diverse func- cular diseases etc. [10]. They directly interacts with key enzymes,
tional entities with multiple biological functions derived from receptors, transcription factors and even with protein aggregates
natural sources such as plants [3]. Many of the currently using which changes different biochemical reactions and signaling path-
chemotherapeutics are derived from natural sources [4]. Polyphe- ways. These molecular level control mechanisms give their potency
nols are a class of natural products present in various plant as promising agents against different diseases including cancer
products like vegetables, fruits, seeds, legumes etc. Apart from [3,9]. The Table 1 describes the various details of polyphenols dis-
yielding colour, aroma and taste to plants, they can also act as cussed in the review which include their sources, scientifically
the major component of plant immune system [5–8]. It com- documented major biological functions and structure.
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 275

Table 1
Summary of different polyphenols with its plant sources, chemical structures and established biological functions.

SL No. Name of the compound Plant source Known activities Structure Reference

1 4- shogaol Ginger Anticancer [140,141]

2 6- Shogaol Ginger Anticancer, antiproliferative, [140,141]


anti-inflammatory and for hypertension

3 Apigenin Parsley, celery, chamomile tea Antioxidant, antistress, anti-inflammatory and [142–144]
anticancer

4 Berberine Berries, turmeric, orange grape, Anti-inflammatory, antidiabetic, anticancer, [145,146]


yellow root, cork tree antiviral, neuroprotective and antidepressant

5 Bergamottin Grapes, citrus Anticancer [147]

6 Caffeic acid Coffee Anticancer [148,149]

7 Capsaicin Chilli peppers Anticancer and antioxidant [150]

8 Cardamonin Cardamom Antioxidant, anticancer and anti-inflammatory [139,151]

9 Casticin Sweet worm wood Anticancer, inhibition of angiogenesis and [152]


chemosensitization

10 Chrysin Passion fruit Neuroprotective and anticancer [153]

11 Cryptotanshinone Salvia Anticancer [154]

12 Cucurbitacin Pumpkins, guards Anticancer [155]

13 Curcumin Turmeric, mango, ginger Anticancer, antidepressant, anti-inflammatory [121,122,156]


and anti- allergic

14 EGCG Tea leaves Antiviral, antidiabetic, anticancer and [157–159]


anti-inflammatory

15 Ellagic acid Berries, walnuts, pecans Anticancer and antioxidant [160,161]

16 Escin Horse chestnut Anticancer [162]

17 Fisetin Strawberries, apple, cucumber, Antioxidant, anticancer and antiproliferative [163] [164]
acacia, onion, persimmons

18 Gambogic acid Garcinia Anticancer [165,166]

19 Gingerol Ginger Anticancer and anti-inflammatory [167,168]

20 Gossypin Hibiscus Anticancer [169]

21 Honokiol Magnolia Anticancer, neuroprotective and cardiac [170,171]


hypertrophy

22 Icarisidell Horny goat weed Anticancer and for erectile dysfunctions [172]

23 Luteolin Orange, grape, lemon Anticancer [173,174]

24 Mangiferin Mango Antioxidant, anti infectious, antidiabetic, [175,176]


antiatherosclerotic and anticancer

25 Piperlongumine Long pepper Neuroprotective and anticancer [168,177]

26 Pterostilbene Almond, berries, grapes Antioxidant, anticancer and anti-inflammatory [5,178,179]

27 Quercetin Cherries, berries, tomato, Antioxidant, anticancer and anti-inflammatory [180–182]


spinach, pepper

28 Resveratrol Grapes, berries Anticancer, antiviral, neurodegenerative [62,183,184]


diseases and vascular diseases
276 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

Table 1 (Continued)

SL No. Name of the compound Plant source Known activities Structure Reference

29 Sesamin Sesame, long pepper, magnolia, Anticancer and antioxidant [185–187]


camellia, Pyrethrum

30 Ursolic acid Apple, berries, peppermint, Anticancer [188,189]


rosemary

31 Wogonin Baikal skullcap Anticancer. neuroprotective, antiviral and [190]


antioxidant

32 Xanthohumol Hops Antioxidant, anticancer, antiviral and [191]


anti-inflammatory

3. Transcription factors capacity and expression of stem cell markers along with FOXM1
and rescued the stem cells [22]. In addition, DFOG repressed
Transcription factors are gene expression regulators whose the stemness, EMT phenotype in SGC-7901 cells derived gastric
activity resides inside nucleus. Differential expression of genes cancer stem-like cells (GCSLCs), with an associated inhibition of
according to the demands of a cell is achieved with the action of FOXM1 [23]. Casticin, a polymethoxyflavone, inhibited FOXM1 and
specific transcription factors which have direct binding accessi- its downstream molecular targets (survivin and PLK1) in human
bility on the DNA [11]. Different categories of proteins are there hepatocellular and ovarian cancer cell lines [24,25]. The FOXM1
according to its mode of action and role in transcriptional regu- inhibition is associated with activation of FOXO3a and p27 which
lation. Alteration in the level of these proteins will result in the leads to growth arrest and cell cycle inhibition. Curcumin, an active
abnormal variations in its downstream effectors which affects the principle of turmeric also inhibited the FOXM1 expression in malig-
entire homeostatic balance of the system [12]. Transcription factors nant glioma and acute myeloid leukemia cells (AML). The siRNA
which include both oncogenes as well as tumor suppressor genes mediated silencing of FOXM1 in AML made them more susceptible
act as major link between the inflammation and tumorigenesis. The to doxorubicin mediated cell death. Combinatorial administration
expressions of several transcription factors are deregulated in most of curcumin and doxorubicin has shown more evident inhibitory
of the cancers [13,14]. effects in AML cells through the repression of FOXM1 expression
Transcription factors have emerged as the coupling link at both mRNA and protein levels. These studies clearly support the
between inflammation and tumorigenesis. The process of tumori- therapeutic potential in targeting FOXM1 with natural products in
genesis is multistep which can be activated by various environ- chemosensitization, apoptosis, cell proliferation and angiogenesis
mental carcinogens and inflammatory agents, obesity, stress etc in different carcinoma [26,27].
[15]. Most importantly all the above mentioned risk factors are pro-
inflammatory in nature. These agents are known to activate various
3.2. Wnt/ˇ-catenin
types of transcription factors to modulate different signal transduc-
tion cascades [16,17]. Chronic inflammatory conditions often lead
Wnt/␤-catenin signaling is an evolutionarily conserved and
to the process of initiation of tumorigenesis. The expression or acti-
versatile pathway involved in normal tissue homeostasis but
vation status of many of these factors is highly altered in tumor cells
its aberrant activation leads to wide variety of human diseases
providing survival advantages to them [18]. The functional ingre-
[28–30]. Wnt binds to its cell surface receptors, frizzled (Fz) and
dients derived from natural sources are mostly anti-inflammatory
LDL-receptor related proteins (LRP5/6), trigger a signaling cascade
in nature thereby it was postulated and later on proved by sev-
through ␤-catenin, a fascinating molecule in the field of cancer
eral scientific groups across the globe that these agents possess
research [31]. ␤-catenin is a multifunctional protein which is well
significant anticancer activity as well [14,19]. A critical analysis
studied for its structural and signaling roles and also acts as a tran-
of drug discovery studies in the last few decades established the
scription factor [32]. Targeting wnt signaling cascade proteins is
fact that natural product and/or lead molecule derived/synthesized
becoming a dynamic research zone in tumor therapy. In medul-
from natural products continue to play a vital role in the drug dis-
loblastoma cells, curcumin attenuated wnt/␤-catenin signaling and
covery and development process [4]. In this review, we discuss the
inhibited the cell proliferation [33]. Curcumin was found to silence
major transcription factors targeted by natural products in diverse
the TGF-␤ dependent ␤-catenin activation in human cervical cancer
models of cancer and their potential mechanism of action.
cells [34], attenuated wnt/␤-catenin signaling and cell proliferation
in medulloblastoma cells. Decreased population of CD133+ cells
3.1. FOXM1 (Forkhead Box M1) inhibited cancer stem cells and tumorsphere formation through
suppression of wnt and hedgehog signaling in lung cancer cells.
Forkhead Box M1 (FOXM1), an oncofetal transcription factor, The expression of various members of wnt family proteins such as,
is one of the members of the forkhead family of proteins and is LRP6, survivin and ␤-catenin in gastric carcinoma cells were also
required for normal cell division and its deletion is embryoni- down regulated [35].
cally lethal. Upregulation of FOXM1 is reported in a broad range Resveratrol, a phytoalexin has decreased ␤-catenin/TCF-
of cancers. In vitro studies showed that its increased expression mediated transcriptional activity. Resveratrol did not alter the
can lead to invasion and angiogenesis of cancer cells [20]. Silencing levels of ␤- catenin in either cytoplasm or nucleus but disrupted the
FOXM1 expression resulted in reduced tumor formation in immun- complex formation between TCF and ␤- catenin which terminates
odeficient animals [21]. Genistein, a major active component of the signal cascade [36]. In colorectal cancer cells, decreased ␤-
soybean and synthetic derivatives (7-Difluoromethoxyl-5, 4 -di- catenin nuclear translocation and its downstream cascade showed
n-octylgenistein, DFOG) attenuates the expression of FOXM1 and positive correlation with the MALAT-1 (a long non-coding RNA)
its downstream targets such as survivin, cdc25 B and cyclin B in expression linked with inhibition of cell invasion and metasta-
ovarian, gastric and small-cell lung cancer cells. In SKOV-3 ovar- sis [37]. However Liu et al. proposed yet another mechanism for
ian cancer stem cells, DFOG inhibited proliferation, self-renewal the action of resveratrol in CRC cells which is elucidated through
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 277

Fig. 1. Schematic representation of wnt/EGF-␤-catenin signaling and the effects of different polyphenols in its downstream pathway. Curcumin and resveratrol exhibits
direct inhibition of wnt-receptor interaction. Resveratrol disrupts ␤-catenin-TCF complex which prevents the gene expression along with the promotion of autophagy and
inhibition of CSC proliferation. EGCG promotes apoptosis in cancer cells.

increased level of PTEN and inhibition of PI3/Akt and wnt/␤-catenin inducer class of compounds will help in the detoxification of car-
signaling with concomitant decrease in the proliferation of CRC cinogens and environmental mutagens. They can decrease the level
cells [38]. Resveratrol is also known for its ability to decrease the of this transcription factor in tumor cells and make them sus-
survival of cancer stem cells (CSCs). In breast cancer, the effects ceptible for cancer therapy [44]. Tamoxifen resistant MCF7 breast
were mediated through the induction of autophagy in wnt/catenin cancer cells have shown evident response to a combination of EGCG
pathway dependent manner [39]. Similarly EGCG also induced (Epigallocatechin- 3 gallate) and siRNA against Nrf2. It reverses
apoptosis in colon [40] and lung cancer stem cells and inhibited chemoresistance and Nrf2 dependent gene expression [45]. EGCG
spheroid formation by suppressing ␤-catenin signaling [41]. These chemosensitizes triple negative breast cancer cells to cisplatin
studies are highly promising since non-toxic natural products dis- through activation of Nrf2 signaling [46]. Similar mechanism was
cussed here have the ability to suppress the proliferation of CSCs in observed in cervical cancer where EGCG enhanced cisplatin activity
metastasis, drug resistance and tumor recurrence (Fig. 1). and induced apoptosis [47]. A combination of copper and curcumin
was found to have better activity as compared with the agent alone
3.3. Nrf2 in decreasing the proliferation of oral squamous cell carcinoma
(OSCC) where the combination increased ROS levels and activation
The redox sensitive transcription factor, Nrf2 is a member of of Nrf2 and suppress the EMT associated changes [48]. In prostate
basic leucine-zipper family. Its expression is stimulated by the pres- cancer cells a curcumin analogue, FN1 reactivated Nrf2 and its
ence of electrophilic chemicals and ROS and leads to translocation downstream effectors, detoxification enzymes which inhibited the
into the nucleus where it binds to antioxidant-responsive element colony formation of cells [49]. Pterostilbene, a derivative of grape
(ARE) and activate a subset of cytoprotective genes. A cytoskeleton polyphenol resveratrol, decreased the proliferation of melanoma
binding protein keap-1 binds with Nrf2 and prevents the nuclear cells where it downregulated Nrf2 expression [50]. In an ultraviolet
translocation under normal physiological condition. Aberrant Nrf2 B driven skin carcinogens model, Pterostilbene was found to pro-
activation and mutations on keap-1 is reported in various types of tect the mice through the upregulationofNrf2 and Nrf2 dependent
malignancies. Currently there are two approaches (either induc- gene expression [51]. Resveratrol also induced its expression in rat
tion or inhibition) to target Nrf2 for cancer therapy [42,43]. Nrf2 breast cancer model along with the upregulation of Nrf2 depen-
278 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

Table 2
Possible mechanism of action of polyphenols on various tumor models through STAT3.

Polyphenols Type of cancer Mode of action Reference

In vitro

6-Shogaol Lung cancer (NCI-H1650, NCI-H520 and ⊥STAT3 activation, ↓CCl2, ⊥JAK2, caspase- 3, 8, [192–195]
NCI-H1975), Breast cancer(MDA-MB-231), 9 kinase, ⊥Bcl-2,⊥Bcl-XL, ⊥ Survivin, ⊥ c-Src
Prostate cancer (DU145, LNCaP, PC-3, HMVP2)
Acteoside Liver cancer (HepG2, HUH-7, Hep3b) ↓BCL2 [196]
Berberine Gastric cancer (AGS), Colorectal cancer ↓COX2/PGE2, ↓Survivin,↑Apoptosis, ↓MMP-2, [197–201]
(SW620, LoVo, AGS, SW620)0, Nasopharyngeal 9.
cancer (C666-1), Lung cancer (NCI-H460,
NCI-H1975), Skin cancer (A431)
Bergamottin Myeloma (U266), Prostate cancer (DU145), ⊥JAK activation, ↓COX-2, ↓Survivin, ↓VEGF, [202]
Breast cancer (MDA-MB-231) ↓Cyclin D1,↓IAP-1,↓Bcl-2,↓Bcl-XL,↓MMPs and
↑Caspase3
Capsaicin Liver cancer (HepG2) ↑Autophagy [203]
Cardamonin Prostate cancer (LNCaP), Glioblastoma (GSC) ↓STAT3, ⊥DNA binding [71,73]
Chrysin Lung cancer (A549), cervical cancer (HeLa) ↓MCL1, ⊥STAT3 [204]
Cryptotanshinone Liver cancer (BEL-S404, HepG2), pancreatic ↓STAT3, ↓BCL2, ↑Bax, ↑Cas3/9,↑ADP Ribose [205–208]
cancer (BxPC-3), Colorectal cancer (HCT116, polymerase,↓Cyclin-D1, ↓Survivin, ↓EGFR
SW480, LoVo), Glioblastoma (T98G, U8)
Cucurbitacin Lymphoma (Ramos, Raji, jeko), Breast cancer ⊥STAT3 activation, JAK inactivation, [209–213]
(MCF7, T47D, SKBR3, MDA-MB-468), Lung ↓Cyclin-B1, ↑ Apoptosis, ↑Cytochrome C, ↑p53,
cancer (A549),Neuroblastoma (SH-SY5Y), ↑p21
Pancreatic cancer(PC3), Umbilical vein
endothelial cancer (HUVEC)
Curcumin Lung cancer (NCI- H460), Squamous cell ⊥JAK2, ↓MMP2, ↓VEGF, ↓ERK, ↓MMP2, 9, [60,214–218]
carcinoma (Hep-2), Prostate cancer (LNCaP), ↓uPAR, ↓uPA, ↑ROS, Reactivation of RANK
Breast cancer (MCF-7), Bladder cancer
(253J-Bv, P24), Pancreatic cancer (Panc1,
BxPC-3), Tongue cancer (SCC-25), Glioblastoma
(U251, U87, GSC)
Epigallocatechin3- Gallate Leukemia (K562, KBM5), Breast cancer SHP1 activation, ⊥ BCR-ABL, ↓Pro-PARP [45,128]
(MDA-MB-231, MCF-7)
Ellagic acid Prostate cancer (PC3) ↓STAT3, ↑IL6, ↓ Mcl-1. [219]
Honokiol Lung cancer (BrM3, H2030-BrM3), Leukemia ↓p-STAT3, ↑SHP phosphatase, BAX/BCL2 ratio [220,221]
(HEL, THP1) disruption
Icariside II Lymphoma (U937) ⊥JAK, ↓Survivin, ↓Cox2, ↓Bcl2, ↓Bcl-xL [222]
Piperlongumine Gastric cancer (MKN45, AGS) ↓JAK phosphorylation, ↓MMP-9, ↓Twist [223]
Resveratrol Lung cancer (A549, SCC4, FaDu), Ovarian ↓STAT3, ⊥NF-kB, ↑SOCS3, ⊥JAK, ↓IL-6, ↑Beclin, [65–67,224–227]
cancer (OVCAR3, Caov-3), Cervical cancer ↑Autophagy, ↑PAIS3, ⊥Wnt, ⊥Notch
(SiHa,C33a),Leukemia (THP-1, HEL), Bone
marrow cancer (K562), Prostate cancer (LNCaP)
Sesamin Gall bladder cancer (SGC-996, GBC-SD),Liver ⊥NF-kB/AKT, ↓IL-6, ↑p53 [228,229]
cancer (HepG2)
Ursolic acid Colorectal cancer (HT29, HCT116, Sw480, ⊥STAT3 activation [70,230,231]
DLD1), Pancreatic cancer (MiaPaCa-2, AsPC-1)
Wogonin Lung cancer (A549) ⊥JAK activation, ↓Snail, ↓Twist,↓p-STAT3, [232]
↑E-Cadherin, ↓N-Cadherin.
Xanthohumol Bile duct cancer (CCA, M214, M139) ↓NF-kB/AKT, ↓STAT3 [233]

In vivo

Polyphenols Type of cancer Dose Mode of action Reference

6-Shogaol NCI-H1650,4T1, HMVP2Xenograft 10–40 mg/kg, 30 mg/kg, 50–100 mg/kg ↓Metastasis, ↑Caspase, ↑ PARP, [192,194,195]
↓Survivin, ↓p-STAT3
Acteoside DEN induced HCC model 0.1–0.3% of diet/animal ↑ Apoptosis [196]
Berberine C666 Xenograft 5 and 10 mg/kg ↓p-STAT3 [199]
Cryptotanshinone Bel-7404 Xenograft 100 mg/kg ↑Caspase3, 9 [205]
Cucurbitacin Ramos, PC3 cell Xenograft 250 ␮g/kg, 3 mg/kg ↓PCNA, ↓p-STAT3, ↓VEGF [209,213]
Curcumin NCI-H460 Xenograft model 40 mg/kg ↓p-JAK2, ↓p-STAT3, ↑Cleaved [60]
caspase 3, 9
Epigallocatechin3-Gallate K563 Xenograft mice model 15 mg/kg ↑SHP1, ↑Caspase-3, ↓Pro-PARP [234]
Honokiol H2030-BrM3 Orthotopic model 2 and 3 mg/kg ↓p-TrkA, ↓p-TrkB, ↓p-STAT3, [220]
↓p-VEGFR
Resveratrol RG2 Orthotopic model 50 ␮M per animal ↓Cyclin- D1, ↑Beclin, ↓p-STAT3 [64]
Sesamin GBC Xenograft 100 ␮M per animal ↓Drug resistance [228]
Ursolic acid TRAMP mice model, HCT116 1% of diet, 250 mg/kg, 250 mg/kg ↓CyclinD1, ↓Cox2, ↓VEGF, [68–70]
Xenograft, Panc-28 Orthotopic ↓Metastasis, ↓ Angiogenesis,
model ↓Gli-1, ↓VEGF, ↓FGF
Wogonin A549 cell Xenograft 30 and 60 mg/kg ↓p-STAT3, ↑E-Cadherin, [232]
↓N-Cadherin, ↓Vimentin
Xanthohumol KKU-M214 Xenograft 50 mg/animal ↓Ki-67 [233]
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 279

Fig. 2. Schematic representation of various polyphenols acting on STAT3 signaling and associated gene expression. Curcumin inhibits STAT3 activation by inhibiting JNK2
leading to blockage of AKT activation and increased PIAS3 levels. EGCG, cardamonin, resveratrol and ursolic acid directly blocks the STAT3 activation through its inhibition
of phosphorylation and in some cases along with inhibition of acetylation as well.

dent antioxidant genes, thereby reducing the oxidative stress and parallel reduction in p-STAT3 levels [58]. It also inhibited the prolif-
inflammation in mammary tissues [52]. eration of OSCC through decreasing the activation of Akt and STAT3
[59]. Wu et al. also reported that in lung cancer stem cells, curcumin
3.4. STAT3 (Signal transducer and activator of transcription 3) inhibited the growth and reduced tumor spheres through downreg-
ulation of JAK2/STAT3 signaling pathway [60]. Gersey et al. reported
STAT3 is a transcriptional activator of genes involved in cell the effect of curcumin on patient derived glioblastoma stem cell
proliferation, differentiation, inflammation etc. In response to the lines and found that this polyphenol was effective in decreasing
binding of various cytokines to tyrosine kinase receptors the active cell proliferation through increased ROS production and down reg-
dimerization and activation of STAT3 occurs which promotes its ulated STAT3 activation [61].
translocation in to the nucleus [53,54]. Being a candidate in cell Resveratrol, have shown effects on STAT3 proteins in both its
proliferation, it acts as an acute phase response factor which is asso- transcriptional and translational levels which ensure its regulatory
ciated with various processes of inflammation and tumorigenesis mechanisms on cancer [62]. Loss of endogenous STAT3 acetylation
and it is activated by several cytokines, growth factors and onco- resulted in the reactivation of tumor-suppressor genes associ-
genic proteins. A number of naturally derived agents have been ated with the demethylation at their promoters. Treatment with
shown to inhibit the activation of STAT3 and STAT3 dependent gene Resveratrol inhibited the tumor growth in mice and mechanism of
expression [55]. Curcumin inhibited dextran sodium sulphate (DSS) action is through the inhibition of STAT3 acetylation and demethy-
induced colitis (a chronic inflammation model of colon) murine lation of several tumor-suppressor gene promoters [63]. Studies
model through the inhibition of STAT3 pathway [56]. A novel cur- conducted in orthotopic rat glioblastoma models established the
cumin analogue, H-4073 enhanced the sensitivity of cisplatin in relevance of resveratrol as a potent antitumor agent. Capability to
head and neck squamous cell carcinoma (HNSCC) through the inhi- cross blood brain barrier facilitates its greater bio availability in
bition of STAT3 activation and increased expression of p21 and brain which helps to overcome a major problem in cancer thera-
cleaved caspase 3 [57]. Low expression of protein inhibitor of acti- peutics against brain tumors [64]. CAOV-3 and OVCAR-3, ovarian
vated STAT3 (PIAS3) is associated with increased STAT3 activity cancer cell lines exhibit increased autophagy with decreased cell
and poor patient survival in mesothelioma. Curcumin have shown proliferation through the regulation of STAT3 signaling. Resver-
effects in increasing the expression of PIAS3 in these cells with a atrol have shown prominent inhibitory effects on JAK2 protein
280 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

Fig. 3. Schematic representation of androgen receptor signaling and the effects of different polyphenols in its pathway. Resveratrol inhibits androgen receptor activation
with the down regulation of HIF1 and SIRT1 along with increased AR proteasomal degradation. EGCG and quercetin in combination with curcumin shows epigenetic level
regulation of ARs with the inhibition of DNMT and HAT. Curcumin alone prevents wnt dependent expression of androgen receptors.

phosphorylation and thereby indirectly suppress STAT3 activation. sion and IL-6 both at mRNA and protein level was downregulated
Resveratrol mediated indirect inhibition of STAT3 is also char- by cardamonin in breast cancer cells [72]. Wu et al. reported that
acterised by the increased expression of SOCS1 which is a well cardamonin induces apoptosis in glioblastoma stem cells through
explained regulatory protein involved in STAT3 signal desensitisa- suppression of STAT3 activation [73].
tion [65]. In prostate cancer cells elevated transcriptional activity EGCG enhanced therapeutic potential of gemcitabine and JAK3
of both androgen receptors (ARs) and STAT3 was decreased by inhibitor CP- 690550 (tasocitinib) in pancreatic cancer through
resveratrol leading to the inhibition of cellular proliferation [66]. the suppression of STAT3 activation and STAT3 dependent gene
Prominent reduction in ovarian cancer progression and cell survival expression [74]. In cholangiocarcinoma, a combination of EGCG and
with the effective down regulation or inhibition of STAT3 empha- quercetin inhibited the growth and migration of cancer cells and
sises the potential of resveratrol as an effective polyphenol in the abolished JAK/STAT cascade and it’s downstream signaling [75]. In
field of cancer therapeutics [67]. nasopharyngeal carcinoma, EGCG treatment eradicated stem cells
Studies performed using TRAMP mice revealed the protective like tumor cells and increased chemosensitivity through attenu-
role of ursolic acid (UA) in prostate cancer. UA inhibited the acti- ating STAT3 activation [76]. We summarized the observations in
vation of NF-kB, STAT3 and Akt in tumor as well as reduced the Table 2 and Fig. 2. All these reports provided the evidences that
levels of tumor necrosis factor (TNF) and interlukin-6 (IL-6) in STAT3 is involved in the process of inflammation and tumorigene-
serum. Further, UA downregulated the elevated cyclin D1 and COX- sis and there for considered as a common molecular hub for drug
2 and increased caspase 3 expression [68]. Similar effects had been targeting in the field of cancer therapeutics.
observed in orthotopic human colorectal cancer and pancreatic
cancer mice models, where UA inhibited the tumor volume and 3.5. AR (androgen receptors)
metastasis and enhanced the effects of capecitabine/gemcitabine
through downregulation of NF-kB and STAT3 [69,70]. AR is a member of nuclear receptor super family which mediates
Cardamonin, a chalcone inhibited growth of prostate cancer the biological effects of androgens. It binds to an androgen-
cells through the prevention of STAT3 activation. Further, compu- response element (ARE) and regulate gene expression [77]. Several
tational analysis revealed that cardamonin binds directly to the src lines of evidence indicate that AR and associated signaling are
homology 2 domain of STAT3 and thereby inhibits its dimerization implicated in the progression of different type of tumors especially
[71]. Chemotherapeutic drugs induced STAT3 and NF-kB expres- prostate and urothelial cancer. Several polyphenols are shown to
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 281

Fig. 4. Schematic representation of estrogen receptor signaling and polyphenol regulation in its pathway. Curcumin and EGCG show direct inhibition of estrogen receptors.
Xanthohumol disrupts BIG3- prohibitin complex leading to the arrest of activated ER in the cytoplasm. Resveratrol inhibits lGFR- ER signaling and prevents the transcription
of ER response genes. Resveratrol mediated AKT inhibition prevents cell cycle.

modulate AR function and AR dependent gene expression EGCG 3.6. ER (estrogen receptors)
has effects in prostate cancer cell lines with the inhibition of AR
acetylation along with successful blocking of histone acetyl trans- Estrogen receptors are members of nuclear receptor family pro-
ferase (HAT) activity [78]. A combination of curcumin and quercetin teins. ER-␣ and ER-␤ are the two types which are well known for
inhibited the proliferation of androgen refractory prostate cancer its nuclear translocation in response to 17␤-estradiol (estrogen)
cells through decreasing DNA methyl transferase (DNMT) expres- binding [85]. ER-␣ is one of the major culprits in the develop-
sion and methylation marks in AR promoter [79]. PLGA based ment of cancer. Many naturally occurring compounds found in diet
curcumin nanoparticle was effective in suppressing prostate can- which can inhibit the process of inflammation and breast carcino-
cer xenografts as compared to curcumin alone. The mechanism of genesis through suppression of ER signaling pathway [86,87]. ER-␣
action was by decreasing AR, Akt and STAT3 signaling [80]. Cur- has the capacity to shuttle between both nucleus and cytoplasm
cumin downregulated AR signaling through modulation of wnt and E2 binding can happen in any of the compartments. Xan-
pathway in human endometrial carcinoma leading to apoptosis of thohumol destabilizes the BIG3-prohibitn 2 complex and released
tumor cells [81]. prohibitin 2 binds with ER-␣ and inhibited the estrogen depen-
Resveratrol inhibited HIF-1 driven AR signaling and suppress dent gene expression in breast cancer cells [88]. Resveratrol can
the tumor formation in castration-resistant prostate cancer. The cause p53 dependent apoptosis in a dose dependent manner along
elevated AR expression on breast cancer is associated with shorter with ER␣ down regulation in T-47D breast cancer cell lines [89].
overall survival and over expression of BRCA1 in breast cancer cells Resveratrol inhibited the proliferative effects of estrogen and BPA
abrogates AR signaling through activation of SIRT1. Resveratrol is a (an endocrine disrupting chemical) in ovarian cancer cell line BG1
known SIRT1 agonist, which inhibited AR dependent proliferation by down regulating the expressions of ER-␣, IGF-1R, p-Akt1/2/3
of breast cancer cells [82]. It can mediate proteasomal degradation and cyclinD1 both at mRNA and protein levels [90]. Recently a
of ARV7, a splice variant of ARs with its increased ubiquitination; crystal structure of the ER␣ ligand-binding domain (LBD) as a com-
can keep its cellular level as very low in castration resistant prostate plex with resveratrol was reported. This study identified resveratrol
cancer cell lines[83]. Parallel to the AR suppression, resveratrol as a pathway-selective ER␣ ligand which modulate the inflam-
induces increased expression of caspase 3 and 7 which programs matory response surprisingly without affecting cell proliferation
cells to get in to its apoptotic fate [84] (Fig. 3). [91]. The study by Saluzzo et al. demonstrated the regulation of
p53 and ER-␣ by resveratrol at the molecular level [89]. Sev-
282 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

Table 3
Possible mechanism of action of polyphenols on various tumor models through NF-kB.

Polyphenols Type of cancer Mode of action Reference

In vitro
4- Shogaol Breast cancer (MDA-MB-231) ↓Snail, ↓ NF-kB [194]
6-Shogaol Prostate cancer (DU145), Breast cancer ↓ NF-kB, ↓ IkB, ↓ cyclin- D1, ↓ survivin, ↓ c- [195,235]
(MDA-MB-231) Myc, ↓MMP
Apigenin Prostate cancer (PC3, 22Rv1),Pancreatic cancer ↓CSC, ↓p50/p105, ↓p65, ↓IKK, ↓CXCR4, [236–240]
(BxPC-3, Panc1), Lung cancer (A549), Breast ↓Bcl2,↓Cox2, ↑Caspase 3, 8, ↓TNF-␣, ↓IL6,
cancer (MCF-7) ↓miR-16, ↓MMP-9, ↓Cyclin-D
Berberine Liver cancer (HepG2), Lung cancer(NSCLC), ↓P65, ↓P50, ↓IKK, ⊥cox2,↓HIF-1␣, [241,242]
Lymphoma (PEL) ↓VEGF,↓MMPs, ↓PARP and Caspase cleavage
Caffeic acid Skin cancer (HaCaT), Breast cancer(MCF-7) ↑Apoptosis, ↓ NF-kB, ↓ Snail, ↑P38, ↑FAS, ↑BAX, [243,244]
↑JNK
Capsaicin Bile duct carcinoma (HuCCT1) ↓ NF-kB, ⊥ MMP-9 [245]
Cardamonin Breast cancer(MDA-MB-231, 4T1), Liver cancer ↓ NOX-1, ⊥ TSP50 activation, ↓ IKB↓ IL6 and 8, [246]
(HepG2, H7402), Gastric cancer (SGC7901), ↓ MCP1, ↓ NF-kB,
Lung cancer (A549)
Curcumin Colorectal cancer (SW480, LoVo, HT-29, MC38, ↓ p65, ↓Cyclin-D1, ⊥IkB interaction, ↑ IkB, ↓ [127,197,218,247–268]
HCT116), Pancreatic cancer uPA, ↓ MMP2, 9, ROS, ↓ ␤-catenin, ⊥ Cox2, ↓
(BxPC-3,Panc-1,MIAPaCa-2), Breast cancer IKK, ↓MAPK, ↓c-jun, ↓Vimentin, ↓COX2, ↓
(MCF7, MDA-MB 231), Leukemia (Kasumi-1, CXCL1,↑ E-cadherin, ↓ c-PARP, ↓Survivin,
SHL-1), Liver cancer (Hep3B, ↑miR-146A, ↓ DNMT1, ↓ CSC, ⊥ERK, ↑ P38,
HepG2),Glioblastoma (U-87), Prostate cancer ↑JNK, ⊥BCL2,↑ Bax, ↓ Mcl-1, ↓ PI3 K, ↓ mTOR
(PC3, LNCaP,Melanoma (MV3, M14), kinase phosphorylation
Non-Hodgkin’s lymphoma (Namalwa,Ramos,
Raji),Osteoclastoma(GCT), Gastric cancer(AGS),
Oral squamous carcinoma(SAS), Non-Small
Celllung cancer (A549)
Delphinidin Breast cancer (MCF-7) ↓MAPK signaling,↓MMP-9 [269]
Escin Pancreatic cancer (Panc1) ↓ NF-kB [270]
Fisetin Cervical cancer (SiHa, CaSki,), Bladder cancer ↓ p50/p65, ↑Apoptosis, ↑ p21↓ IKKB, ↑ p53, ↓ [271–273]
(T24, J82) BAX, ↓uPA, ↓ Cyclin-D1, ↓ Cyclin-A, ↑ BAK,
↓CDK2, ↓CDK4, ↓Bcl-2
Gambogic acid Prostate cancer (PC3), Lukemia (K562), ↓CXCR4, ↓ NF-kB, ↓ c-Myc, ↓ IL-6, ⊥TNF-␣, ↓ [274–276]
Myeloma (MM.1S, RPMI 8226, UT66NCIH929) Bcl2, ↓ p-AKT
Genistein Bladder cancer (J82, SCaBER, ↓ERK, ↓Notch signaling, ↑miR-29b, ↓XIAP, [134–136,277–279]
TCCSUP),Leukemia(HUT102), Breast cancer ↓cIAP, ↓survivin, ↓PI3 K, ↓cyclin-B1, ↓Bcl-2,
(MDA-MB-231),Colorectal cancer (HT29,LoVo), ↓Bcl-xL
Myeloma (U266)
Gingerol Pancreatic cancer (Panc1) ↓ NF-kB, ⊥ ERK [280]
Gossypin Chronic myelogenous leukemia (KBM-5), ⊥IkB␣ kinase, ↓ Survivin, ↓ COX2, ↓ Cyclin-D1, [281]
Myeloma (U266), Lung cancer (H1299), ↓MMP-9, ↓c-Myc, ↓VEGF
Pancreatic cancer (BxPC-3), Urinary bladder
cancer (253J-BV)
Honokiol HTLV-1 infected Cell lines ↓NFkB,⊥I␬B␣, ⊥IKK␣, ⊥IKK␤, ⊥STAT3,↓Jun D, [282]
⊥ STAT5, ⊥ p-Akt, ↓ Jun B
Luteolin Ovarian cancer (X10, X22), ↓FAK, ↓ERK,↓ BACE1 [283–285]
Neuroblastoma(SH-SY5Y), Lung cancer (A549)
Mangiferin Lung cancer (A549), Prostate cancer (LNCaP) ↓NF-kB, ↓IKK, ↓Protein Kinase C, ↑PARP-1, [286,287]
↓Survivin, ↑Caspase, ↓MMP-9
Quercetin Glioma (C6), Tongue cancer(SAS) ↓ IKK, ↑ Caspase3, ↑Apoptosis, ↓ PI3 K, ↓MMP- [288,289]
2, 7, 9, 10, ↓ VEGF, ↓ IKB- ␣, ↓ COX2, ↓iNOS, ↓
uPA
Sesamin Prostate cancer (PC3), Gall bladder cancer ↓ NF-kB, ↓MMP-9,↓VEGF, ↓iCAM-1, ↓IL-6, ↓ [228,290]
(SGC-996, GBC-SD) Vimentin,↓Twist
Ursolic acid Skin melanoma (CRL-11147), Bladder cancer ↑Apoptosis, ↓p65, ↓ IkB␣,⊥AKT, ↓Bcl2 [291–293]
(T24), Pancreatic cancer (Panc1, Capan-1)
Xanthohumol Liver cancer (HepG2) ⊥IkB, ↑ P53, ↑ Cleaved PARP,↑ AIF, ↑ [294]
Cytochrome C

In vivo

Polyphenols Type of cancer Dose Mode of action Reference

6-Shogaol PC3 Allograft mice model 50 mg/kg ↓ NF-␬B, ↓ I␬B [195]


Apigenin Buccal Pouch carcinoma hamster 2.5 mg/kg ↓ PCNA, ↓ Cyclin-D, ↓ COX2 [295]
Berberine B16F-10 Melanoma mice model 10 mg/kg ↓ NF-␬B [296]
Cardamonin 4T1 Xenograft 25 mg/kg ⊥TSP50 activation, ↓ CSC [246]
Curcumin B[a] PDE induced lung cancer, SAS 2%of the diet, 70 mg/kg, ⊥NF-␬B/MAPK, ↓ Cox-2, ↓ Cyclin- [126,218,257,258,297–299]
Xenograft, GL261, leukemia cells mice 2 mg/animal,100–200 mg/kg, D1, ↓ p65, ↓ NF-␬B, ↓TNF- ␣, ↓ IL6,
model, H1975 ectopic tumor model, 0.6% of the diet, 0.6% of the ↓ CXCL1
MiaPaCa-2 orthotopic model, diet, 0.6% of the diet
MDA-MB-231 xenograft
Fisetin MNU induced bladder cancer model 200 mg/kg ↓ IKKB, ↓ NF-␬B [273]
Mangiferin A549 Xenograft mice model, Mouse 10, 50 and 100 mg/kg ↓NF-␬B, ↓ IKK, ↓ Protein Kinase C [286,300]
metastatic Melanomamodel (B16BL6) 50, 100 and 200 mg/kg
Rutin BaP induced lung cancer 40 and 80 mg/kg ↓ LDH activity, ↓ p65 [301]
Ursolic acid MiaPaCa-2 Xenograft 100 and 200 mg/kg ⊥p-I␬B [293]
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 283

Fig. 5. Schematic representation of various polyphenols and it derivatives acting on HIF-1␣ leading to decrease tumor growth, proliferation, metastasis etc. Curcumin and
EF24, a curcumin analogue inhibits EMT and chemosensitizes the cells to drugs. Tetrahydrocurcumin and EGCG inhibit VEGF and prevent angiogenesis. Resveratrol prevents
metastasis. Quercetin promotes autophagy of cancer cells.

eral studies conclusively established that ER- ␣ negative breast sion of genes involved in oxygen delivery to tissues and energy
cancer is more aggressive and less responsive to hormonal ther- metabolism. Increased expression of HIF1 is often associated with
apies. A combination of resveratrol and pterostilbene was found poor prognosis which is observed in several types of human can-
to restore the ER-␣ expression and thereby sensitizes the cancer cers. Several natural products have shown promising results in
cells to hormonal therapies [92]. These studies clearly demon- regulating HIF-1 synthesis and transcriptional activity thereby
strated the epigenetic regulatory activity of resveratrol. Curcumin bringing the potential role of HIF-1 inhibitors in cancer therapy
is yet another natural product which regulates ER-␣ expression [97,98]. Curcumin in combination with cisplatin reversed drug
and downstream signaling. This polyphenol inhibited the growth of resistance in lung cancer cells through HIF-1 degradation and cas-
several ER positive cancer cells in culture [93]. A curcumin analogue pase 3 activation and enhanced cisplatin mediated cell death [99].
PAC (4-hydroxy-3-methoxybenzylidene)-N-methyl-4-piperidone) A similar mechanism was executed by EF24, a novel curcumin
was found to be more stable than curcumin in blood which sup- analogue with improved pharmacokinetic activity as compared
pressed the ER-␣expression and induced apoptosis in different to parent curcumin. In hepatic cancer combinations EF24 with
breast cancer cells [94]. ER-␣36 is a variant of ER-␣ expressed sorafenib overcome the sorafenib resistance [100] and EMT reg-
in both hormonal positive and negative cancer cells. EGCG was ulated HIF-1 expression [101]. Tetrahydrocurcumin, a metabolite
found to abrogate to AR-␣36-EGFR loop and suppress the growth of curcumin was also found to suppress the expression of HIF-
and proliferation of breast cancer stem cells. EGCG regulates ER-␣ 1 and VEGF, thereby suppressing the process of angiogenesis
expression at promoter, mRNA and protein levels [95]. EGCG and [102]. Resveratrol inhibited hypoxia driven invasion and migra-
a histone deacetylase (HDAC) inhibitor, trichostatin A (TSA) syner- tion of pancreatic cells with decreased expression of HIF-1 [103]
gistically re-activate ER-␣ expression in ER-␣ negative cancer cells and hypoxia lead to EMT changes which reduced proliferation of
by altering the histone acetylation status in its promoter. This will osteosarcoma cells [104]. Mitani et al. reported that in prostate
be an ideal chemotherapeutic strategy in hormone resistant breast cancer, resveratrol was effective in suppressing hypoxia driven
cancer [96] (Fig. 4). accumulation of ␤ −catenin in nucleus and activation of AR thereby
contribute to regression of tumors [105]. EGCG abrogates lung
cancer angiogenesis through the inhibition of HIF-1 and VEGF
3.7. HIF1 (hypoxia inducing factor 1)
expression [106]. Honokiol a lignin class of compound was found to
have radio sensitizing activity in murine colorectal cancer models
HIF-1 is a transcription factor that mediates cellular response
by modulating hypoxia driven signaling pathways [107].
to reduced availability of oxygen through modulating the expres-
284 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

Fig. 6. Schematic representations of various polyphenols and it derivatives acting on NF-kB signaling and associated gene expression. Resveratrol through downregulation
of PI3 K and AKT pathways block the nuclear translocation of NF-kB. Curcumin modulates MAPK, NF-kB and chemokine signaling and inhibited cell and cancer stem cell
proliferation. EGCG inhibits both MAPK and NF-kB signaling and expression of NF-kB dependent genes VEGF, MMP 2 and 8. Both Cardamonin and Genistein reduce NF-kB
activation and increases apoptosis in cancer cells.

In gastric cancer cells the anti-proliferative effect of quercetin is resveratrol or EF24, was effective in promoting ionizing radiation
mediated through the induction of protective autophagy through (IR) induced apoptosis in hypoxic cancer cells through the suppres-
modulation of Akt/mTOR/HIF-1 pathway [108]. Interestingly, sion of IR induced NF-kB activation in breast cancer cells [117].
Isorhamnetin, a metabolite of quercetin was found to be a better Others have also shown that resveratrol inhibited the growth of dif-
inhibitor of HIF-1 than quercetin [109]. Currently, there is a search ferent tumor types through attenuating NF-kB signaling [118–120].
for agents that can effectively inhibit HIF-1 and VEGF1/2 which are Curcumin, its natural and synthetic analogues and different formu-
responsible for increased survivability for tumor cells in hypoxic lations were studied extensively in the regulation of NF-kB and
condition. Polyphenolic class of compounds can be potential source its dependent gene expression in multiple systems [121,122]. A
for such inhibitors (Fig. 5). liposomal formulation of EF24 was effectively down regulated NF-
kB cascade in pancreatic cancer cells and inhibited the growth
both in vitro and in vivo [123]. A combination of curcumin and
3.8. NF-B (nuclear factor kappaB) tolfenamic acid was found have synergistic activity in suppress-
ing the growth of pancreatic cancer cells [124]. This polyphenol
Nuclear factor-kB is a pro-inflammatory transcription factor also reversed the oxaliplatin resistance in colorectal cancer through
which regulates numerous pathways that has direct implications modulation of CXC-Chemokine/NF-␬B signaling pathway [125]. A
in inflammation and cancer [110]. Several in vitro, pre-clinical and diet containing curcumin was able to prevent benzopyrene induced
clinical studies showed that NF-kB and NF-kB dependent gene lung cancer in mice by reducing NF-kB activity and MAPK signal-
expressions play a major role in inflammation, cancer progression, ing [126]. In liver cancer, curcumin depleted the cells having cancer
metastasis and drug resistance [111]. Over the last few decades stem cell like properties through NF-kB dependent histone deacety-
several phytochemicals and their analogues were identified and lase (HDAC) inhibition [127]. A combination of EGCG and curcumin
demonstrated to have significant inhibitory effects on NF-signaling was suppressive in nature against breast cancer derived CSCs where
by targeting members of this cascade [112–114]. In colorectal can- both NF-kB and STAT3 pathways were downregulated [128] (Fig. 6).
cer cells, resveratrol attenuated the phosphorylation, acetylation EGCG, the tea polyphenol was also found to modulate NF-kB
and nuclear translocation of NF-kB [115]. This phytoalexin inhib- pathway in a variety of tumor cells. It blocked the proliferation and
ited adhesion, invasion and migration of glioblastoma-initiating invasion of colorectal cancer cells [129]. A combination of EGCG
cells (GICs) both in vitro and in vivo through suppression of and radiotherapy in breast cancer patients showed lower levels of
PI3 K/Akt/NF-␬B cascade signifying the therapeutic potential of NF-kB dependent genes such as VEGF, MMP-2 and 9 in serum [130].
resveratrol in glioblastoma cases [116]. The pre-treatment of either
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 285

In human bladder cancer, EGCG inhibited the invasion and migra- [6] V. Cheynier, F.A. Tomas-Barberan, K. Yoshida, Polyphenols from plants to a
tion of cells through suppression of PI3 K/Akt/NF-kB axis [131]. In variety of food and nonfood uses, J. Agric. Food Chem. 63 (35) (2015)
7589–7594.
nasopharyngeal cancer, this tea polyphenol reversed the process [7] F.A. Tomas-Barberan, C. Andres-Lacueva, Polyphenols and health: current
of EMT and inhibited the self-renewal of CSCs [132]. In lung can- state and progress, J. Agric. Food Chem. 60 (36) (2012) 8773–8775.
cer models EGCG modulate the expression of microRNAs that can [8] A.N. Li, S. Li, Y.J. Zhang, X.R. Xu, Y.M. Chen, H.B. Li, Resources and biological
activities of natural polyphenols, Nutrients 6 (12) (2014) 6020–6047.
regulate NF-kB and MAP kinase pathways [133]. [9] S. Quideau, D. Deffieux, C. Douat-Casassus, L. Pouysegu, Plant polyphenols:
Genistein is also explored for its potential pharmacological chemical properties, biological activities, and synthesis, Angew. Chem. Int.
effects. It induced cell death in human multiple myeloma with the Ed. Engl. 50 (3) (2011) 586–621.
[10] E. Brglez Mojzer, M. Knez Hrncic, M. Skerget, Z. Knez, U. Bren, Polyphenols:
possible mechanism of modulation of NF-kB and miR-29 expres-
extraction methods, antioxidative action, bioavailability and
sion [134]. GEN-27, a derivative of genistein was found to have anticarcinogenic effects, Molecules 21 (7) (2016).
therapeutic benefits in colitis associated cancer models and human [11] J.E. Darnell Jr., Transcription factors as targets for cancer therapy, Nat. Rev.
Cancer 2 (10) (2002) 740–749.
colorectal cancer and effects were mediated through suppression of
[12] A.H. Brivanlou, J.E. Darnell Jr., Signal transduction and the control of gene
p65-CDX2-␤-catenin axis [135]. Genistein was reported to induce expression, Science 295 (5556) (2002) 813–818.
apoptosis in T-cell leukemia [136], breast cancer [137] and gas- [13] S.J. Johnston, J.S. Carroll, Transcription factors and chromatin proteins as
tric cancer [138] with the suppression of NF-kB signaling. We therapeutic targets in cancer, Biochim. Biophys. Acta 1855 (2) (2015)
183–192.
have recently shown that cardamonin inhibited the azoxymethane [14] J.E. Yeh, P.A. Toniolo, D.A. Frank, Targeting transcription factors: promising
induced colorectal cancer in mice and one of the mechanisms was new strategies for cancer therapy, Curr. Opin. Oncol. 25 (6) (2013) 652–658.
found to be the inhibition of NF-kB signaling[139].We have sum- [15] B.B. Aggarwal, S. Shishodia, S.K. Sandur, M.K. Pandey, G. Sethi, Inflammation
and cancer: how hot is the link? Biochem. Pharmacol. 72 (11) (2006)
marized both in vitro and in vivo studies in Table 3 and Fig. 6. 1605–1621.
[16] F. Colotta, P. Allavena, A. Sica, C. Garlanda, A. Mantovani, Cancer-related
inflammation, the seventh hallmark of cancer: links to genetic instability,
4. Conclusion Carcinogenesis 30 (7) (2009) 1073–1081.
[17] P. Allavena, C. Garlanda, M.G. Borrello, A. Sica, A. Mantovani, Pathways
The process of tumorigenesis is a complex phenomenon where connecting inflammation and cancer, Curr. Opin. Genet. Dev. 18 (1) (2008)
3–10.
several perturbations of various signaling pathways elicited by sev- [18] B. Sung, S. Prasad, V.R. Yadav, B.B. Aggarwal, Cancer cell signaling pathways
eral transcription factors get altered leading to aberrant signaling targeted by spice-derived nutraceuticals, Nutr. Cancer 64 (2) (2012)
that cause cellular transformation. Currently, we are focusing more 173–197.
[19] J. Hagenbuchner, M.J. Ausserlechner, Targeting transcription factors by
on multi-targeted therapies which have several advantages over
small compounds–Current strategies and future implications, Biochem.
conventional therapies with less side effects and toxicity. Natu- Pharmacol. 107 (2016) 1–13.
ral products based drug discovery has significant potential against [20] H. Chen, Y. Zou, H. Yang, J. Wang, H. Pan, Downregulation of FoxM1 inhibits
various types of cancers as seen from various in vitro, ex vivo, pre- proliferation, invasion and angiogenesis of HeLa cells in vitro and in vivo,
Int. J. Oncol. 45 (6) (2014) 2355–2364.
clinical and clinical studies. In most of the cases, the target of natural [21] Q. Fan, Q. Cai, Y. Xu, FOXM1 is a downstream target of LPA and YAP
products is transcription factors. These agents derived from Mother oncogenic signaling pathways in high grade serous ovarian cancer,
Nature acts at different phases of signal transduction cascades initi- Oncotarget 6 (29) (2015) 27688–27699.
[22] Y.X. Ning, Q.X. Li, K.Q. Ren, M.F. Quan, J.G. Cao, 7-Difluoromethoxyl-5,
ated by transcription factors there by regulating three major steps 4’-di-n-octyl genistein inhibits ovarian cancer stem cell characteristics
in carcinogenesis viz. initiation, promotion and progression. The through the downregulation of FOXM1, Oncol. Lett. 8 (1) (2014) 295–300.
current need of the situation is to start appropriate randomized [23] X. Cao, K. Ren, Z. Song, D. Li, M. Quan, Y. Zheng, J. Cao, W. Zeng, H. Zou,
7-Difluoromethoxyl-5, 4’-di-n-octyl genistein inhibits the stem-like
clinical trials of natural products either alone or in combination characteristics of gastric cancer stem-like cells and reverses the phenotype
with existing standard of care for the benefit of cancer patients. of epithelial-mesenchymal transition in gastric cancer cells, Oncol. Rep. 36
(2) (2016) 1157–1165.
[24] L. Jiang, X.C. Cao, J.G. Cao, F. Liu, M.F. Quan, X.F. Sheng, K.Q. Ren, Casticin
Conflict of interest induces ovarian cancer cell apoptosis by repressing FoxM1 through the
activation of FOXO3a, Oncol. Lett. 5 (5) (2013) 1605–1610.
[25] L. He, X. Yang, X. Cao, F. Liu, M. Quan, J. Cao, Casticin induces growth
The authors showed no conflict of interest.
suppression and cell cycle arrest through activation of FOXO3a in
hepatocellular carcinoma, Oncol. Rep. 29 (1) (2013) 103–108.
Acknowledgements [26] W.Z. Du, Y. Feng, X.F. Wang, X.Y. Piao, Y.Q. Cui, L.C. Chen, X.H. Lei, X. Sun, X.
Liu, H.B. Wang, X.F. Li, D.B. Yang, Y. Sun, Z.F. Zhao, T. Jiang, Y.L. Li, C.L. Jiang,
Curcumin suppresses malignant glioma cells growth and induces apoptosis
This work was supported by the Ramalingaswami Re-entry fel- by inhibition of SHH/GLI1 signaling pathway in vitro and vivo, CNS Neurosci.
lowship of Department of Biotechnology, Government of India Ther. 19 (12) (2013) 926–936.
[27] J.R. Zhang, F. Lu, T. Lu, W.H. Dong, P. Li, N. Liu, D.X. Ma, C.Y. Ji, Inactivation of
(BT/RLF/Re-entry/38/2011) to KBH. CR and MBL acknowledge FoxM1 transcription factor contributes to curcumin-induced inhibition of
research fellowship from Council of Scientific & Industrial Research survival, angiogenesis, and chemosensitivity in acute myeloid leukemia
(CSIR) and University Grant Commission (UGC) respectively. We cells, J. Mol. Med. (Berl.) 92 (12) (2014) 1319–1330.
[28] B. Ma, M.O. Hottiger, Crosstalk between wnt/beta-catenin and NF-kappaB
also acknowledge the members of KBH lab for critical reading of signaling pathway during inflammation, Front. Immunol. 7 (2016) 378.
the manuscript. [29] B. Madan, D.M. Virshup, Targeting Wnts at the source–new mechanisms,
new biomarkers, new drugs, Mol. Cancer Ther. 14 (5) (2015) 1087–1094.
[30] R. Nusse, H. Clevers, Wnt/beta-catenin signaling, disease, and emerging
References therapeutic modalities, Cell 169 (6) (2017) 985–999.
[31] A. Karimaian, M. Majidinia, H. Bannazadeh Baghi, B. Yousefi, The crosstalk
[1] R.H. Wilting, J.H. Dannenberg, Epigenetic mechanisms in tumorigenesis, between Wnt/beta-catenin signaling pathway with DNA damage response
tumor cell heterogeneity and drug resistance, Drug Resist. Updat. 15 (1–2) and oxidative stress: implications in cancer therapy, DNA Repair (Amst.) 51
(2012) 21–38. (2017) 14–19.
[2] E. Elinav, R. Nowarski, C.A. Thaiss, B. Hu, C. Jin, R.A. Flavell, [32] H. Clevers, Wnt/beta-catenin signaling in development and disease, Cell 127
Inflammation-induced cancer: crosstalk between tumours, immune cells (3) (2006) 469–480.
and microorganisms, Nat. Rev. Cancer 13 (11) (2013) 759–771. [33] M. He, Y. Li, L. Zhang, L. Li, Y. Shen, L. Lin, W. Zheng, L. Chen, X. Bian, H.K. Ng,
[3] F. Bucar, A. Wube, M. Schmid, Natural product isolation–how to get from L. Tang, Curcumin suppresses cell proliferation through inhibition of the
biological material to pure compounds, Nat. Prod. Rep. 30 (4) (2013) Wnt/beta-catenin signaling pathway in medulloblastoma, Oncol. Rep. 32 (1)
525–545. (2014) 173–180.
[4] D.J. Newman, G.M. Cragg, Natural products as sources of new drugsfrom [34] P.C. Thacker, D. Karunagaran, Curcumin and emodin down-regulate
1981 to 2014, J. Nat. Prod. 79 (3) (2016) 629–661. TGF-beta signaling pathway in human cervical cancer cells, PLoS One 10 (3)
[5] R. Tsao, Chemistry and biochemistry of dietary polyphenols, Nutrients 2 (12) (2015) e0120045.
(2010) 1231–1246.
286 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

[35] R. Zheng, Q. Deng, Y. Liu, P. Zhao, Curcumin inhibits gastric carcinoma cell [60] L. Wu, L. Guo, Y. Liang, X. Liu, L. Jiang, L. Wang, Curcumin suppresses
growth and induces apoptosis by suppressing the wnt/beta-catenin stem-like traits of lung cancer cells via inhibiting the JAK2/STAT3 signaling
signaling pathway, Med. Sci. Monit. 23 (2017) 163–171. pathway, Oncol. Rep. 34 (6) (2015) 3311–3317.
[36] H.J. Chen, L.S. Hsu, Y.T. Shia, M.W. Lin, C.M. Lin, The beta-catenin/TCF [61] Z.C. Gersey, G.A. Rodriguez, E. Barbarite, A. Sanchez, W.M. Walters, K.C.
complex as a novel target of resveratrol in the Wnt/beta-catenin signaling Ohaeto, R.J. Komotar, R.M. Graham, Curcumin decreases malignant
pathway, Biochem. Pharmacol. 84 (9) (2012) 1143–1153. characteristics of glioblastoma stem cells via induction of reactive oxygen
[37] Q. Ji, X. Liu, X. Fu, L. Zhang, H. Sui, L. Zhou, J. Sun, J. Cai, J. Qin, J. Ren, Q. Li, species, BMC Cancer 17 (1) (2017) 99.
Resveratrol inhibits invasion and metastasis of colorectal cancer cells via [62] K.B. Harikumar, B.B. Aggarwal, Resveratrol: a multitargeted agent for
MALAT1 mediated Wnt/beta-catenin signal pathway, PLoS One 8 (11) age-associated chronic diseases, ABBV Cell Cycle 7 (8) (2008) 1020–1035.
(2013) e78700. [63] H. Lee, P. Zhang, A. Herrmann, C. Yang, H. Xin, Z. Wang, D.S. Hoon, S.J.
[38] Y.Z. Liu, K. Wu, J. Huang, Y. Liu, X. Wang, Z.J. Meng, S.X. Yuan, D.X. Wang, J.Y. Forman, R. Jove, A.D. Riggs, H. Yu, Acetylated STAT3 is crucial for methylation
Luo, G.W. Zuo, L.J. Yin, L. Chen, Z.L. Deng, J.Q. Yang, W.J. Sun, B.C. He, The of tumor-suppressor gene promoters and inhibition by resveratrol results in
PTEN/PI3 K/Akt and Wnt/beta-catenin signaling pathways are involved in demethylation, Proc. Natl. Acad. Sci. U. S. A. 109 (20) (2012) 7765–7769.
the inhibitory effect of resveratrol on human colon cancer cell proliferation, [64] S. Xue, S. Xiao-Hong, S. Lin, B. Jie, W. Li-Li, G. Jia-Yao, S. Shun, L. Pei-Nan, W.
Int. J. Oncol. 45 (1) (2014) 104–112. Mo-Li, W. Qian, C. Xiao-Yan, K. Qing-You, Z. Peng, L. Hong, L. Jia, Lumbar
[39] Y. Fu, H. Chang, X. Peng, Q. Bai, L. Yi, Y. Zhou, J. Zhu, M. Mi, Resveratrol puncture-administered resveratrol inhibits STAT3 activation, enhancing
inhibits breast cancer stem-like cells and induces autophagy via suppressing autophagy and apoptosis in orthotopic rat glioblastomas, Oncotarget 7 (46)
Wnt/beta-catenin signaling pathway, PLoS One 9 (7) (2014) e102535. (2016) 75790–75799.
[40] Y. Chen, X.Q. Wang, Q. Zhang, J.Y. Zhu, Y. Li, C.F. Xie, X.T. Li, J.S. Wu, S.S. Geng, [65] S.H. Baek, J.H. Ko, H. Lee, J. Jung, M. Kong, J.W. Lee, J. Lee, A. Chinnathambi,
C.Y. Zhong, H.Y. Han, (−)-Epigallocatechin-3-gallate inhibits colorectal M.E. Zayed, S.A. Alharbi, S.G. Lee, B.S. Shim, G. Sethi, S.H. Kim, W.M. Yang, J.Y.
cancer stem cells by suppressing wnt/beta-Catenin pathway, Nutrients 9 (6) Um, K.S. Ahn, Resveratrol inhibits STAT3 signaling pathway through the
(2017). induction of SOCS-1: Role in apoptosis induction and radiosensitization in
[41] J. Zhu, Y. Jiang, X. Yang, S. Wang, C. Xie, X. Li, Y. Li, Y. Chen, X. Wang, Y. Meng, head and neck tumor cells, Phytomedicine 23 (5) (2016) 566–577.
M. Zhu, R. Wu, C. Huang, X. Ma, S. Geng, J. Wu, C. Zhong, Wnt/beta-catenin [66] M.H. Lee, J.K. Kundu, Y.S. Keum, Y.Y. Cho, Y.J. Surh, B.Y. Choi, Resveratrol
pathway mediates (−)-epigallocatechin-3-gallate (EGCG) inhibition of lung inhibits IL-6-induced transcriptional activity of AR and STAT3 in human
cancer stem cells, Biochem. Biophys. Res. Commun. 482 (1) (2017) 15–21. prostate cancer LNCaP-FGC cells, Biomol. Ther. (Seoul) 22 (5) (2014)
[42] K. Taguchi, M. Yamamoto, The KEAP1-NRF2 system in cancer, Front. Oncol. 7 426–430.
(2017) 85. [67] L.X. Zhong, H. Li, M.L. Wu, X.Y. Liu, M.J. Zhong, X.Y. Chen, J. Liu, Y. Zhang,
[43] J. Zhu, H. Wang, F. Chen, J. Fu, Y. Xu, Y. Hou, H.H. Kou, C. Zhai, M.B. Nelson, Q. Inhibition of STAT3 signaling as critical molecular event in
Zhang, M.E. Andersen, J. Pi, An overview of chemical inhibitors of the resveratrol-suppressed ovarian cancer cells, J. Ovarian Res. 8 (2015) 25.
Nrf2-ARE signaling pathway and their potential applications in cancer [68] M.K. Shanmugam, T.H. Ong, A.P. Kumar, C.K. Lun, P.C. Ho, P.T. Wong, K.M.
therapy, Free Radic. Biol. Med. 99 (2016) 544–556. Hui, G. Sethi, Ursolic acid inhibits the initiation, progression of prostate
[44] G. Thiel, O.G. Rossler, Resveratrol regulates gene transcription via activation cancer and prolongs the survival of TRAMP mice by modulating
of stimulus-responsive transcription factors, Pharmacol. Res. 117 (2017) pro-inflammatory pathways, PLoS One 7 (3) (2012) e32476.
166–176. [69] S. Prasad, V.R. Yadav, B. Sung, S. Reuter, R. Kannappan, A. Deorukhkar, P.
[45] M.A. Esmaeili, Combination of siRNA-directed gene silencing with Diagaradjane, C. Wei, V. Baladandayuthapani, S. Krishnan, S. Guha, B.B.
epigallocatechin-3-gallate (EGCG) reverses drug resistance in human breast Aggarwal, Ursolic acid inhibits growth and metastasis of human colorectal
cancer cells, J. Chem. Biol. 9 (1) (2016) 41–52. cancer in an orthotopic nude mouse model by targeting multiple cell
[46] K. Foygel, T.V. Sekar, R. Paulmurugan, Monitoring the antioxidant mediated signaling pathways: chemosensitization with capecitabine, Clin. Cancer Res.
chemosensitization and ARE-signaling in triple negative breast cancer 18 (18) (2012) 4942–4953.
therapy, PLoS One 10 (11) (2015) e0141913. [70] S. Prasad, V.R. Yadav, B. Sung, S.C. Gupta, A.K. Tyagi, B.B. Aggarwal, Ursolic
[47] U. Kilic, K. Sahin, M. Tuzcu, N. Basak, C. Orhan, B. Elibol-Can, E. Kilic, F. Sahin, acid inhibits the growth of human pancreatic cancer and enhances the
O. Kucuk, Enhancement of Cisplatin sensitivity in human cervical cancer: antitumor potential of gemcitabine in an orthotopic mouse model through
epigallocatechin-3-gallate, Front. Nutr. 1 (2014) 28. suppression of the inflammatory microenvironment, Oncotarget 7 (11)
[48] H.M. Lee, V. Patel, L.F. Shyur, W.L. Lee, Copper supplementation amplifies (2016) 13182–13196.
the anti-tumor effect of curcumin in oral cancer cells, Phytomedicine 23 [71] J. Zhang, S. Sikka, K.S. Siveen, J.H. Lee, J.Y. Um, A.P. Kumar, A. Chinnathambi,
(12) (2016) 1535–1544. S.A. Alharbi, Basappa, K.S. Rangappa, G. Sethi, K.S. Ahn, Cardamonin
[49] W. Li, D. Pung, Z.Y. Su, Y. Guo, C. Zhang, A.Y. Yang, X. Zheng, Z.Y. Du, K. represses proliferation, invasion, and causes apoptosis through the
Zhang, A.N. Kong, Epigenetics reactivation of nrf2 in prostate TRAMP C1Cells modulation of signal transducer and activator of transcription 3 pathway in
by curcumin analogue FN1, Chem. Res. Toxicol. 29 (4) (2016) prostate cancer, Apoptosis 22 (1) (2017) 158–168.
694–703. [72] D. Jia, Y. Tan, H. Liu, S. Ooi, L. Li, K. Wright, S. Bennett, C.L. Addison, L. Wang,
[50] M. Benlloch, E. Obrador, S.L. Valles, M.L. Rodriguez, J.A. Sirerol, J. Alcacer, J.A. Cardamonin reduces chemotherapy-enriched breast cancer stem-like cells
Pellicer, R. Salvador, C. Cerda, G.T. Saez, J.M. Estrela, Pterostilbene decreases in vitro and in vivo, Oncotarget 7 (1) (2016) 771–785.
the antioxidant defenses of aggressive cancer cells in vivo: a physiological [73] N. Wu, J. Liu, X. Zhao, Z. Yan, B. Jiang, L. Wang, S. Cao, D. Shi, X. Lin,
glucocorticoids- and nrf2-dependent mechanism, Antioxid. Redox Signal. 24 Cardamonin induces apoptosis by suppressing STAT3 signaling pathway in
(17) (2016) 974–990. glioblastoma stem cells, Tumour Biol. 36 (12) (2015) 9667–9676.
[51] J.A. Sirerol, F. Feddi, S. Mena, M.L. Rodriguez, P. Sirera, M. Aupi, S. Perez, M. [74] S.N. Tang, J. Fu, S. Shankar, R.K. Srivastava, EGCG enhances the therapeutic
Asensi, A. Ortega, J.M. Estrela, Topical treatment with pterostilbene a natural potential of gemcitabine and CP690550 by inhibiting STAT3 signaling
phytoalexin, effectively protects hairless mice against UVB pathway in human pancreatic cancer, PLoS One 7 (2) (2012) e31067.
radiation-induced skin damage and carcinogenesis, Free Radic. Biol. Med. 85 [75] L. Senggunprai, V. Kukongviriyapan, A. Prawan, U. Kukongviriyapan,
(2015) 1–11. Quercetin and EGCG exhibit chemopreventive effects in
[52] B. Singh, R. Shoulson, A. Chatterjee, A. Ronghe, N.K. Bhat, D.C. Dim, H.K. Bhat, cholangiocarcinoma cells via suppression of JAK/STAT signaling pathway,
Resveratrol inhibits estrogen-induced breast carcinogenesis through Phytother. Res.: PTR 28 (6) (2014) 841–848.
induction of NRF2-mediated protective pathways, Carcinogenesis 35 (8) [76] C.H. Lin, L.K. Chao, P.H. Hung, Y.J. Chen, EGCG inhibits the growth and
(2014) 1872–1880. tumorigenicity of nasopharyngeal tumor-initiating cells through
[53] J.E. Darnell Jr., STATs and gene regulation, Science 277 (5332) (1997) attenuation of STAT3 activation, Int. J. Clin. Exp. Pathol. 7 (5) (2014)
1630–1635. 2372–2381.
[54] D.E. Levy, J.E. Darnell Jr., Stats: transcriptional control and biological impact, [77] S.N. Birrell, R.E. Hall, W.D. Tilley, Role of the androgen receptor in human
Nat. Rev. Mol. Cell Biol. 3 (9) (2002) 651–662. breast cancer, J. Mammary Gland Biol. Neoplasia 3 (1) (1998) 95–103.
[55] B.B. Aggarwal, A.B. Kunnumakkara, K.B. Harikumar, S.R. Gupta, S.T. [78] Y.H. Lee, J. Kwak, H.K. Choi, K.C. Choi, S. Kim, J. Lee, W. Jun, H.J. Park, H.G.
Tharakan, C. Koca, S. Dey, B. Sung, Signal transducer and activator of Yoon, EGCG suppresses prostate cancer cell growth modulating acetylation
transcription-3, inflammation, and cancer: how intimate is the relationship? of androgen receptor by anti-histone acetyltransferase activity, Int. J. Mol.
Ann. N. Y. Acad. Sci. 1171 (2009) 59–76. Med. 30 (1) (2012) 69–74.
[56] J.Y. Yang, X. Zhong, H.W. Yum, H.J. Lee, J.K. Kundu, H.K. Na, Y.J. Surh, [79] V. Sharma, L. Kumar, S.K. Mohanty, J.P. Maikhuri, S. Rajender, G. Gupta,
Curcumin inhibits STAT3 signaling in the colon of dextran sulfate Sensitization of androgen refractory prostate cancer cells to anti-androgens
sodium-treated mice, J Cancer Prev 18 (2) (2013) 186–191. through re-expression of epigenetically repressed androgen receptor –
[57] B. Kumar, A. Yadav, K. Hideg, P. Kuppusamy, T.N. Teknos, P. Kumar, A novel synergistic action of quercetin and curcumin, Mol. Cell. Endocrinol. 431
curcumin analog (H-4073) enhances the therapeutic efficacy of cisplatin (2016) 12–23.
treatment in head and neck cancer, PLoS One 9 (3) (2014) e93208. [80] M.M. Yallapu, S. Khan, D.M. Maher, M.C. Ebeling, V. Sundram, N. Chauhan, A.
[58] S. Dabir, A. Kluge, A. Kresak, M. Yang, P. Fu, B. Groner, G. Wildey, A. Dowlati, Ganju, S. Balakrishna, B.K. Gupta, N. Zafar, M. Jaggi, S.C. Chauhan,
Low PIAS3 expression in malignant mesothelioma is associated with Anti-cancer activity of curcumin loaded nanoparticles in prostate cancer,
increased STAT3 activation and poor patient survival, Clin. Cancer Res. 20 Biomaterials 35 (30) (2014) 8635–8648.
(19) (2014) 5124–5132. [81] W. Feng, C.X. Yang, L. Zhang, Y. Fang, M. Yan, Curcumin promotes the
[59] L. Zhen, D. Fan, X. Yi, X. Cao, D. Chen, L. Wang, Curcumin inhibits oral apoptosis of human endometrial carcinoma cells by downregulating the
squamous cell carcinoma proliferation and invasion via EGFR signaling expression of androgen receptor through Wnt signal pathway, Eur. J.
pathways, Int. J. Clin. Exp. Pathol. 7 (10) (2014) 6438–6446. Gynaecol. Oncol. 35 (6) (2014) 718–723.
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 287

[82] W. Zhang, J. Luo, F. Yang, Y. Wang, Y. Yin, A. Strom, J.A. Gustafsson, X. Guan, through downregulation of the HIF-1alpha protein, Mol. Med. Rep. 11 (3)
BRCA1 inhibits AR-mediated proliferation of breast cancer cells through the (2015) 1975–1981.
activation of SIRT1, Sci. Rep. 6 (2016) 22034. [105] T. Mitani, N. Harada, S. Tanimori, Y. Nakano, H. Inui, R. Yamaji, Resveratrol
[83] S. Wilson, L. Cavero, D. Tong, Q. Liu, K. Geary, N. Talamonti, J. Xu, J. Fu, J. inhibits hypoxia-inducible factor-1alpha-mediated androgen receptor
Jiang, D. Zhang, Resveratrol enhances polyubiquitination-mediated ARV7 signaling and represses tumor progression in castration-resistant prostate
degradation in prostate cancer cells, Oncotarget 8 (33) (2017) 54683–54693. cancer, J. Nutr. Sci. Vitaminol. (Tokyo) 60 (4) (2014) 276–282.
[84] A. Ferruelo, I. Romero, P.M. Cabrera, I. Arance, G. Andres, J.C. Angulo, Effects [106] X. Li, Y. Feng, J. Liu, X. Feng, K. Zhou, X. Tang, Epigallocatechin-3-gallate
of resveratrol and other wine polyphenols on the proliferation, apoptosis inhibits IGF-I-stimulated lung cancer angiogenesis through downregulation
and androgen receptor expression in LNCaP cells, Actas Urol. Esp. 38 (6) of HIF-1alpha and VEGF expression, J. Nutrigenet Nutrigenomics 6 (3)
(2014) 397–404. (2013) 169–178.
[85] R.E. Buller, B.W. O’Malley, The biology and mechanism of steroid hormone [107] K.L. Lan, K.H. Lan, M.L. Sheu, M.Y. Chen, Y.S. Shih, F.C. Hsu, H.M. Wang, R.S.
receptor interaction with the eukaryotic nucleus, Biochem. Pharmacol. 25 Liu, S.H. Yen, Honokiol inhibits hypoxia-inducible factor-1 pathway, Int. J.
(1) (1976) 1–12. Radiat. Biol. 87 (6) (2011) 579–590.
[86] A.C. Tecalco-Cruz, I.A. Perez-Alvarado, J.O. Ramirez-Jarquin, L. [108] K. Wang, R. Liu, J. Li, J. Mao, Y. Lei, J. Wu, J. Zeng, T. Zhang, H. Wu, L. Chen, C.
Rocha-Zavaleta, Nucleo-cytoplasmic transport of estrogen receptor alpha in Huang, Y. Wei, Quercetin induces protective autophagy in gastric cancer
breast cancer cells, Cell. Signal. 34 (2017) 121–132. cells: involvement of Akt-mTOR- and hypoxia-induced factor
[87] M.J. Bak, S. Das Gupta, J. Wahler, N. Suh, Role of dietary bioactive natural 1alpha-mediated signaling, Autophagy 7 (9) (2011) 966–978.
products in estrogen receptor-positive breast cancer, Semin. Cancer Biol. [109] S. Seo, K. Seo, S.H. Ki, S.M. Shin, Isorhamnetin inhibits reactive oxygen
40–41 (2016) 170–191. species-Dependent hypoxia inducible factor (HIF)-1alpha accumulation,
[88] T. Yoshimaru, M. Komatsu, E. Tashiro, M. Imoto, H. Osada, Y. Miyoshi, J. Biol. Pharm. Bull. 39 (11) (2016) 1830–1838.
Honda, M. Sasa, T. Katagiri, Xanthohumol suppresses oestrogen-signalling in [110] J.A. DiDonato, F. Mercurio, M. Karin, NF-kappaB and the link between
breast cancer through the inhibition of BIG3-PHB2 interactions, Sci. Rep. 4 inflammation and cancer, Immunol. Rev. 246 (1) (2012) 379–400.
(2014) 7355. [111] Q. Zhang, M.J. Lenardo, D. Baltimore, 30 Years of NF-kappaB: a blossoming of
[89] J. Saluzzo, K.M. Hallman, K. Aleck, B. Dwyer, M. Quigley, V. Mladenovik, A.E. relevance to human pathobiology, Cell 168 (1–2) (2017) 37–57.
Siebert, S. Dinda, The regulation of tumor suppressor protein, p53, and [112] S.C. Gupta, J.H. Kim, R. Kannappan, S. Reuter, P.M. Dougherty, B.B. Aggarwal,
estrogen receptor (ERalpha) by resveratrol in breast cancer cells, Genes Role of nuclear factor kappaB-mediated inflammatory pathways in
Cancer 7 (11–12) (2016) 414–425. cancer-related symptoms and their regulation by nutritional agents, Exp.
[90] N.H. Kang, K.A. Hwang, H.R. Lee, D.W. Choi, K.C. Choi, Resveratrol regulates Biol. Med. (Maywood) 236 (6) (2011) 658–671.
the cell viability promoted by 17beta-estradiol or bisphenol A via [113] M.K. Shanmugam, J.H. Lee, E.Z. Chai, M.M. Kanchi, S. Kar, F. Arfuso, A.
down-regulation of the cross-talk between estrogen receptor alpha and Dharmarajan, A.P. Kumar, P.S. Ramar, C.Y. Looi, M.R. Mustafa, V. Tergaonkar,
insulin growth factor-1 receptor in BG-1 ovarian cancer cells, Food Chem. A. Bishayee, K.S. Ahn, G. Sethi, Cancer prevention and therapy through the
Toxicol. 59 (2013) 373–379. modulation of transcription factors by bioactive natural compounds, Semin.
[91] J.C. Nwachukwu, S. Srinivasan, N.E. Bruno, A.A. Parent, T.S. Hughes, J.A. Cancer Biol. 40–41 (2016) 35–47.
Pollock, O. Gjyshi, V. Cavett, J. Nowak, R.D. Garcia-Ordonez, R. Houtman, P.R. [114] G. Sethi, V. Tergaonkar, Potential pharmacological control of the NF-kappaB
Griffin, D.J. Kojetin, J.A. Katzenellenbogen, M.D. Conkright, K.W. Nettles, pathway, Trends Pharmacol. Sci. 30 (6) (2009) 313–321.
Resveratrol modulates the inflammatory response via an estrogen [115] Z. Ren, L. Wang, J. Cui, Z. Huoc, J. Xue, H. Cui, Q. Mao, R. Yang, Resveratrol
receptor-signal integration network, Elife 3 (2014) e02057. inhibits NF-kB signaling through suppression of p65 and IkappaB kinase
[92] R. Kala, T.O. Tollefsbol, A novel combinatorial epigenetic therapy using activities, Pharmazie 68 (8) (2013) 689–694.
resveratrol and pterostilbene for restoring estrogen receptor-alpha [116] Y. Jiao, H. Li, Y. Liu, A. Guo, X. Xu, X. Qu, S. Wang, J. Zhao, Y. Li, Y. Cao,
(ERalpha) expression in ERalpha-negative breast cancer cells, PLoS One 11 Resveratrol inhibits the invasion of glioblastoma-Initiating cells via
(5) (2016) e0155057. down-Regulation of the PI3 K/Akt/NF-kappaB signaling pathway, Nutrients
[93] K. Hallman, K. Aleck, B. Dwyer, V. Lloyd, M. Quigley, N. Sitto, A.E. Siebert, S. 7 (6) (2015) 4383–4402.
Dinda, The effects of turmeric (curcumin) on tumor suppressor protein [117] S. Aravindan, M. Natarajan, T.S. Herman, V. Awasthi, N. Aravindan, Molecular
(p53) and estrogen receptor (ERalpha) in breast cancer cells, Breast Cancer basis of ‘hypoxic’ breast cancer cell radio-sensitization: phytochemicals
(Dove Med. Press.) 9 (2017) 153–161. converge on radiation induced Rel signaling, Radiat. Oncol. 8 (2013) 46.
[94] H.A. Al-Howail, H.A. Hakami, B. Al-Otaibi, A. Al-Mazrou, M.H. Daghestani, I. [118] X. Zhang, A. Jiang, B. Qi, Z. Ma, Y. Xiong, J. Dou, J. Wang, Resveratrol protects
Al-Jammaz, H.H. Al-Khalaf, A. Aboussekhra, PAC down-regulates estrogen against helicobacter pylori-associated gastritis by combating oxidative
receptor alpha and suppresses epithelial-to-mesenchymal transition in stress, Int. J. Mol. Sci. 16 (11) (2015) 27757–27769.
breast cancer cells, BMC Cancer 16 (2016) 540. [119] M.L. Tsai, C.S. Lai, Y.H. Chang, W.J. Chen, C.T. Ho, M.H. Pan, Pterostilbene, a
[95] F. De Amicis, A. Russo, P. Avena, M. Santoro, A. Vivacqua, D. Bonofiglio, L. natural analogue of resveratrol, potently inhibits
Mauro, S. Aquila, D. Tramontano, S.A. Fuqua, S. Ando, In vitro mechanism for 7,12-dimethylbenz[a]anthracene
downregulation of ER-alpha expression by epigallocatechin gallate in (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA)-induced mouse skin
ER+/PR+ human breast cancer cells, Mol. Nutr. Food Res. 57 (5) (2013) carcinogenesis, Food Funct. 3 (11) (2012) 1185–1194.
840–853. [120] J. Ryu, B.M. Ku, Y.K. Lee, J.Y. Jeong, S. Kang, J. Choi, Y. Yang, D.H. Lee, G.S. Roh,
[96] Y. Li, Y.Y. Yuan, S.M. Meeran, T.O. Tollefsbol, Synergistic epigenetic H.J. Kim, G.J. Cho, W.S. Choi, N. Kim, S.S. Kang, Resveratrol reduces
reactivation of estrogen receptor-alpha (ERalpha) by combined green tea TNF-alpha-induced U373MG human glioma cell invasion through regulating
polyphenol and histone deacetylase inhibitor in ERalpha-negative breast NF-kappaB activation and uPA/uPAR expression, Anticancer Res. 31 (12)
cancer cells, Mol. Cancer 9 (2010) 274. (2011) 4223–4230.
[97] G.L. Semenza, HIF-1 mediates metabolic responses to intratumoral hypoxia [121] S.C. Gupta, G. Kismali, B.B. Aggarwal, Curcumin, a component of turmeric:
and oncogenic mutations, J. Clin. Invest. 123 (9) (2013) 3664–3671. from farm to pharmacy, Biofactors 39 (1) (2013) 2–13.
[98] E. Minet, G. Michel, J. Remacle, C. Michiels, Role of HIF-1 as a transcription [122] A.B. Kunnumakkara, D. Bordoloi, C. Harsha, K. Banik, S.C. Gupta, B.B.
factor involved in embryonic development, cancer progression and Aggarwal, Curcumin mediates anticancer effects by modulating multiple
apoptosis (review), Int. J. Mol. Med. 5 (3) (2000) 253–259. cell signaling pathways, Clin. Sci. (Lond.) 131 (15) (2017) 1781–1799.
[99] M.X. Ye, Y.L. Zhao, Y. Li, Q. Miao, Z.K. Li, X.L. Ren, L.Q. Song, H. Yin, J. Zhang, [123] S. Bisht, M. Schlesinger, A. Rupp, R. Schubert, J. Nolting, J. Wenzel, S.
Curcumin reverses cis-platin resistance and promotes human lung Holdenrieder, P. Brossart, G. Bendas, G. Feldmann, A liposomal formulation
adenocarcinoma A549/DDP cell apoptosis through HIF-1alpha and of the synthetic curcumin analog EF24 (Lipo-EF24) inhibits pancreatic
caspase-3 mechanisms, Phytomedicine 19 (8–9) (2012) 779–787. cancer progression: towards future combination therapies, J.
[100] Y. Liang, T. Zheng, R. Song, J. Wang, D. Yin, L. Wang, H. Liu, L. Tian, X. Fang, X. Nanobiotechnology 14 (1) (2016) 57.
Meng, H. Jiang, J. Liu, L. Liu, Hypoxia-mediated sorafenib resistance can be [124] R. Basha, S.F. Connelly, U.T. Sankpal, G.P. Nagaraju, H. Patel, J.K.
overcome by EF24 through Von Hippel-Lindau tumor Vishwanatha, S. Shelake, L. Tabor-Simecka, M. Shoji, J.W. Simecka, B.
suppressor-dependent HIF-1alpha inhibition in hepatocellular carcinoma, El-Rayes, Small molecule tolfenamic acid and dietary spice curcumin
Hepatology 57 (5) (2013) 1847–1857. treatment enhances antiproliferative effect in pancreatic cancer cells via
[101] W. Duan, Y. Chang, R. Li, Q. Xu, J. Lei, C. Yin, T. Li, Y. Wu, Q. Ma, X. Li, suppressing Sp1, disrupting NF-kB translocation to nucleus and cell cycle
Curcumin inhibits hypoxia inducible factor1alphainduced phase distribution, J. Nutr. Biochem. 31 (2016) 77–87.
epithelialmesenchymal transition in HepG2 hepatocellular carcinoma cells, [125] V. Ruiz de Porras, S. Bystrup, A. Martinez-Cardus, R. Pluvinet, L. Sumoy, L.
Mol. Med. Rep. 10 (5) (2014) 2505–2510. Howells, M.I. James, C. Iwuji, J.L. Manzano, L. Layos, C. Buges, A. Abad, E.
[102] B. Yoysungnoen, P. Bhattarakosol, S. Patumraj, C. Changtam, Effects of Martinez-Balibrea, Curcumin mediates oxaliplatin-acquired resistance
tetrahydrocurcumin on hypoxia-inducible factor-1alpha and vascular reversion in colorectal cancer cell lines through modulation of
endothelial growth factor expression in cervical cancer cell-induced CXC-Chemokine/NF-kappaB signalling pathway, Sci. Rep. 6 (2016) 24675.
angiogenesis in nude mice, BioMed Res. Int. 2015 (2015) 391748. [126] V.T. Puliyappadamba, A.K. Thulasidasan, V. Vijayakurup, J. Antony, S.V. Bava,
[103] W. Li, L. Cao, X. Chen, J. Lei, Q. Ma, Resveratrol inhibits hypoxia-driven S. Anwar, S. Sundaram, R.J. Anto, Curcumin inhibits B[a]PDE-induced
ROS-induced invasive and migratory ability of pancreatic cancer cells via procarcinogenic signals in lung cancer cells, and curbs B[a]P-induced
suppression of the Hedgehog signaling pathway, Oncol. Rep. 35 (3) (2016) mutagenesis and lung carcinogenesis, Biofactors 41 (6) (2015) 431–442.
1718–1726. [127] J.U. Marquardt, L. Gomez-Quiroz, L.O. Arreguin Camacho, F. Pinna, Y.H. Lee,
[104] Y. Sun, H. Wang, M. Liu, F. Lin, J. Hua, Resveratrol abrogates the effects of M. Kitade, M.P. Dominguez, D. Castven, K. Breuhahn, E.A. Conner, P.R. Galle,
hypoxia on cell proliferation, invasion and EMT in osteosarcoma cells J.B. Andersen, V.M. Factor, S.S. Thorgeirsson, Curcumin effectively inhibits
288 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

oncogenic NF-kappaB signaling and restrains stemness features in liver [153] E.R. Kasala, L.N. Bodduluru, R.M. Madana, A.K.V, R. Gogoi, C.C. Barua,
cancer, J. Hepatol. 63 (3) (2015) 661–669. Chemopreventive and therapeutic potential of chrysin in cancer:
[128] S.S. Chung, J.V. Vadgama, Curcumin and epigallocatechin gallate inhibit the mechanistic perspectives, Toxicol. Lett. 233 (2) (2015) 214–225.
cancer stem cell phenotype via down-regulation of STAT3-NFkappaB [154] W. Chen, Y. Lu, G. Chen, S. Huang, Molecular evidence of cryptotanshinone
signaling, Anticancer Res. 35 (1) (2015) 39–46. for treatment and prevention of human cancer, Anticancer Agents Med.
[129] F. Zhou, H. Zhou, T. Wang, Y. Mu, B. Wu, D.L. Guo, X.M. Zhang, Y. Wu, Chem. 13 (7) (2013) 979–987.
Epigallocatechin-3-gallate inhibits proliferation and migration of human [155] E. Attard, M.G. Martinoli, E. Cucurbitacin, An experimental lead triterpenoid
colon cancer SW620 cells in vitro, Acta Pharmacol. Sin. 33 (1) (2012) with anticancer, immunomodulatory and novel effects against degenerative
120–126. diseases. A mini-review, Curr. Top. Med. Chem. 15 (17) (2015) 1708–1713.
[130] G. Zhang, Y. Wang, Y. Zhang, X. Wan, J. Li, K. Liu, F. Wang, Q. Liu, C. Yang, P. [156] T. Esatbeyoglu, P. Huebbe, I.M. Ernst, D. Chin, A.E. Wagner, G. Rimbach,
Yu, Y. Huang, S. Wang, P. Jiang, Z. Qu, J. Luan, H. Duan, L. Zhang, A. Hou, S. Jin, Curcumin–from molecule to biological function, Angew. Chem. Int. Ed. Engl.
T.C. Hsieh, E. Wu, Anti-cancer activities of tea epigallocatechin-3-gallate in 51 (22) (2012) 5308–5332.
breast cancer patients under radiotherapy, Curr. Mol. Med. 12 (2) (2012) [157] L. Chakrawarti, R. Agrawal, S. Dang, S. Gupta, R. Gabrani, Therapeutic effects
163–176. of EGCG: a patent review, Expert Opin. Ther. Pat. 26 (8) (2016) 907–916.
[131] J. Qin, Y. Wang, Y. Bai, K. Yang, Q. Mao, Y. Lin, D. Kong, X. Zheng, L. Xie, [158] B.N. Singh, S. Shankar, R.K. Srivastava, Green tea catechin,
Epigallocatechin-3-gallate inhibits bladder cancer cell invasion via epigallocatechin-3-gallate (EGCG): mechanisms, perspectives and clinical
suppression of NF-kappaBmediated matrix metalloproteinase-9 expression, applications, Biochem. Pharmacol. 82 (12) (2011) 1807–1821.
Mol. Med. Rep. 6 (5) (2012) 1040–1044. [159] C.S. Yang, H. Wang, G.X. Li, Z. Yang, F. Guan, H. Jin, Cancer prevention by tea:
[132] Y.J. Li, S.L. Wu, S.M. Lu, F. Chen, Y. Guo, S.M. Gan, Y.L. Shi, S. Liu, S.L. Li, evidence from laboratory studies, Pharmacol. Res. 64 (2) (2011) 113–122.
(−)-Epigallocatechin-3-gallate inhibits nasopharyngeal cancer stem cell [160] A. Shakeri, M.R. Zirak, A. Sahebkar, Ellagic acid: a logical lead for drug
self-renewal and migration and reverses the epithelial-mesenchymal development? Curr. Pharm. Des. (2017).
transition via NF-kappaB p65 inactivation, TumourBiol. 36 (4) (2015) [161] G. Derosa, P. Maffioli, A. Sahebkar, Ellagic acid and its role in chronic
2747–2761. diseases, Adv. Exp. Med. Biol. 928 (2016) 473–479.
[133] H. Zhou, J.X. Chen, C.S. Yang, M.Q. Yang, Y. Deng, H. Wang, Gene regulation [162] J.M.R. Patlolla, C.V. Rao, Anti-inflammatory and anti-cancer Properties of
mediated by microRNAs in response to green tea polyphenol EGCG in mouse ␤-Escin, a triterpene saponin, Curr. Pharmacol. Rep. 1 (2015) 170–178.
lung cancer, BMC Genomics 15 (Suppl 11) (2014) S3. [163] H.C. Pal, R.L. Pearlman, F. Afaq, Fisetin and its role in chronic diseases, Adv.
[134] J. Xie, J. Wang, B. Zhu, Genistein inhibits the proliferation of human multiple Exp. Med. Biol. 928 (2016) 213–244.
myeloma cells through suppression of nuclear factor-kappaB and [164] N. Khan, D.N. Syed, N. Ahmad, H. Mukhtar, Fisetin: a dietary antioxidant for
upregulation of microRNA-29b, Mol. Med. Rep. 13 (2) (2016) health promotion, Antioxid. Redox Signal. 19 (2) (2013) 151–162.
1627–1632. [165] M.K. Pandey, D. Karelia, S.G. Amin, Gambogic acid and its role in chronic
[135] Q. Du, Y. Wang, C. Liu, H. Wang, H. Fan, Y. Li, J. Wang, X. Zhang, J. Lu, H. Ji, R. diseases, Adv. Exp. Med. Biol. 928 (2016) 375–395.
Hu, Chemopreventive activity of GEN-27, a genistein derivative, in [166] X. Wang, W. Chen, Gambogic acid is a novel anti-cancer agent that inhibits
colitis-associated cancer is mediated by p65-CDX2-beta-catenin axis, cell proliferation, angiogenesis and metastasis, Anticancer Agents Med.
Oncotarget 7 (14) (2016) 17870–17884. Chem. 12 (8) (2012) 994–1000.
[136] M. Yamasaki, Y. Mine, M. Nishimura, S. Fujita, Y. Sakakibara, M. Suiko, K. [167] Y.A. Mohd Yusof, Gingerol and its role in chronic diseases, Adv. Exp. Med.
Morishita, K. Nishiyama, Genistein induces apoptotic cell death associated Biol. 929 (2016) 177–207.
with inhibition of the NF-kappaB pathway in adult T-cell leukemia cells, Cell [168] S. Prasad, A.K. Tyagi, Historical spice as a future drug: therapeutic potential
Biol. Int. 37 (7) (2013) 742–747. of piperlongumine, Curr. Pharm. Des. 22 (27) (2016) 4151–4159.
[137] H. Pan, W. Zhou, W. He, X. Liu, Q. Ding, L. Ling, X. Zha, S. Wang, Genistein [169] S. Bhaskaran, K.V. Dileep, S.S. Deepa, C. Sadasivan, M. Klausner, N.K.
inhibits MDA-MB-231 triple-negative breast cancer cell growth by Krishnegowda, R.R. Tekmal, J.L. VandeBerg, H.B. Nair, Gossypin as a novel
inhibiting NF-kappaB activity via the Notch-1 pathway, Int. J. Mol. Med. 30 selective dual inhibitor of V-RAF murine sarcoma viral oncogene homolog
(2) (2012) 337–343. B1 and cyclin-dependent kinase 4 for melanoma, Mol. Cancer Ther. 12 (4)
[138] Y.S. Li, L.P. Wu, K.H. Li, Y.P. Liu, R. Xiang, S.B. Zhang, L.Y. Zhu, L.Y. Zhang, (2013) 361–372.
Involvement of nuclear factor kappaB (NF-kappaB) in the downregulation of [170] A. Woodbury, S.P. Yu, L. Wei, P. Garcia, Neuro-modulating effects of
cyclooxygenase-2 (COX-2) by genistein in gastric cancer cells, J. Int. Med. honokiol: a review, Front. Neurol. 4 (2013) 130.
Res. 39 (6) (2011) 2141–2150. [171] R. Prasad, S.K. Katiyar, Honokiol, an active compound of magnolia plant,
[139] S. James, J.S. Aparna, A.M. Paul, M.B. Lankadasari, S. Mohammed, V.S. Binu, inhibits growth, and progression of cancers of different organs, Adv. Exp.
T.R. Santhoshkumar, G. Reshmi, K.B. Harikumar, Cardamonin inhibits Med. Biol. 928 (2016) 245–265.
colonic neoplasia through modulation of MicroRNA expression, Sci. Rep. 7 [172] M. Khan, A. Maryam, J.I. Qazi, T. Ma, Targeting apoptosis and multiple
(1) (2017) 13945. signaling pathways with icariside II in cancer cells, Int. J. Biol. Sci. 11 (9)
[140] R.B. Semwal, D.K. Semwal, S. Combrinck, A.M. Viljoen, Gingerols and (2015) 1100–1112.
shogaols. Important nutraceutical principles from ginger, Phytochemistry [173] M.J. Tuorkey, Molecular targets of luteolin in cancer, Eur. J. Cancer Prev. 25
117 (2015) 554–568. (1) (2016) 65–76.
[141] M.S. Baliga, R. Haniadka, M.M. Pereira, J.J. D’Souza, P.L. Pallaty, H.P. Bhat, S. [174] M. Lopez-Lazaro, Distribution and biological activities of the flavonoid
Popuri, Update on the chemopreventive effects of ginger and its luteolin, Mini Rev. Med. Chem. 9 (1) (2009) 31–59.
phytochemicals, Crit. Rev. Food Sci. Nutr. 51 (6) (2011) 499–523. [175] Jyotshna, P. Khare, K. Shanker, Mangiferin A review of sources and
[142] S.F. Nabavi, H. Khan, G. D’Onofrio, D. Samec, S. Shirooie, A.R. Dehpour, S. interventions for biological activities, Biofactors 42 (5) (2016) 504–514.
Arguelles, S. Habtemariam, E. Sobarzo-Sanchez, Apigenin as neuroprotective [176] O. Benard, Y. Chi, Medicinal properties of mangiferin, structural features,
agent: of mice and men, Pharmacol. Res. (2017). derivative synthesis, pharmacokinetics and biological activities, Mini Rev.
[143] S.A. Ganai, Plant-derived flavone apigenin: the small-molecule with Med. Chem. 15 (7) (2015) 582–594.
promising activity against therapeutically resistant prostate cancer, Biomed. [177] D.P. Bezerra, C. Pessoa, M.O. de Moraes, N. Saker-Neto, E.R. Silveira, L.V.
Pharmacother. 85 (2017) 47–56. Costa-Lotufo, Overview of the therapeutic potential of piplartine
[144] D. Tang, K. Chen, L. Huang, J. Li, Pharmacokinetic properties and drug (piperlongumine), Eur. J. Pharm. Sci. 48 (3) (2013) 453–463.
interactions of apigenin, a natural flavone, Expert Opin. Drug Metab. Toxicol. [178] J.M. Estrela, A. Ortega, S. Mena, M.L. Rodriguez, M. Asensi, Pterostilbene
13 (3) (2017) 323–330. biomedical applications, Crit. Rev. Clin. Lab. Sci. 50 (3) (2013) 65–78.
[145] T. Ahmed, A.U. Gilani, M. Abdollahi, M. Daglia, S.F. Nabavi, S.M. Nabavi, [179] D. McCormack, D. McFadden, A review of pterostilbene antioxidant activity
Berberine and neurodegeneration. A review of literature, Pharmacol. Rep. 67 and disease modification, Oxid. Med. Cell. Longev. 2013 (2013) 575482.
(5) (2015) 970–979. [180] G.S. Kelly, Quercetin. Monograph, Altern. Med. Rev. 16 (2) (2011) 172–194.
[146] S. Habtemariam, Berberine and inflammatory bowel disease: a concise [181] M. Russo, C. Spagnuolo, I. Tedesco, S. Bilotto, G.L. Russo, The flavonoid
review, Pharmacol. Res. 113 (Pt A) (2016) 592–599. quercetin in disease prevention and therapy: facts and fancies, Biochem.
[147] W.L. Hung, J.H. Suh, Y. Wang, Chemistry and health effects of Pharmacol. 83 (1) (2012) 6–15.
furanocoumarins in grapefruit, J. Food Drug Anal. 25 (1) (2017) 71–83. [182] K. Kawabata, R. Mukai, A. Ishisaka, Quercetin and related polyphenols: new
[148] T. Silva, C. Oliveira, F. Borges, Caffeic acid derivatives, analogs and insights and implications for their bioactivity and bioavailability, Food
applications: a patentreview (2009–2013), Expert Opin. Ther. Pat. 24 (11) Funct. 6 (5) (2015) 1399–1417.
(2014) 1257–1270. [183] A. Rauf, M. Imran, H.A.R. Suleria, B. Ahmad, D.G. Peters, M.S. Mubarak, A
[149] M. Touaibia, J. Jean-Francois, J. Doiron, Caffeic Acid, a versatile comprehensive review of the health perspectives of resveratrol, Food Funct.
pharmacophore: an overview, Mini Rev. Med. Chem. 11 (8) (2011) 695–713. (2017).
[150] K. Srinivasan, Biological activities of red pepper (Capsicum annuum) and its [184] H.Y. Tsai, C.T. Ho, Y.K. Chen, Biological actions and molecular effects of
pungent principle capsaicin: a review, Crit. Rev. Food Sci. Nutr. 56 (9) (2016) resveratrol, pterostilbene, and 3’-hydroxypterostilbene, J. Food. Drug Anal.
1488–1500. 25 (1) (2017) 134–147.
[151] L.M. Goncalves, I.M. Valente, J.A. Rodrigues, An overview on cardamonin, J. [185] A.F. Majdalawieh, M. Massri, G.K. Nasrallah, A comprehensive review on the
Med. Food 17 (6) (2014) 633–640. anti-cancer properties and mechanisms of action of sesamin, a lignan in
[152] A. Rasul, B.J. Zhao, J. Liu, B. Liu, J.X. Sun, J. Li, X.M. Li, Molecular mechanisms sesame seeds (Sesamum indicum), Eur. J. Pharmacol. 815 (2017) 512–521.
of casticin action: an update on its antitumor functions, Asian Pac. J. Cancer [186] A.A. Dar, N. Arumugam, Lignans of sesame: purification methods, biological
Prev. 15 (21) (2014) 9049–9058. activities and biosynthesis–a review, Bioorg. Chem. 50 (2013) 1–10.
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 289

[187] K. Yasuda, T. Sakaki, How is sesamin metabolised in the human liver to show [211] M. Zhang, Z.G. Bian, Y. Zhang, J.H. Wang, L. Kan, X. Wang, H.Y. Niu, P. He,
its biological effects? Expert Opin. Drug Metab. Toxicol. 8 (1) (2012) 93–102. Cucurbitacin B inhibits proliferation and induces apoptosis via STAT3
[188] D. Kashyap, H.S. Tuli, A.K. Sharma, Ursolic acid (UA): A metabolite with pathway inhibition in A549 lung cancer cells, Mol. Med. Rep. 10 (6) (2014)
promising therapeutic potential, Life Sci. 146 (2016) 201–213. 2905–2911.
[189] S.T. Cargnin, S.B. Gnoatto, Ursolic acid from apple pomace and traditional [212] Q. Zheng, Y. Liu, W. Liu, F. Ma, Y. Zhou, M. Chen, J. Chang, Y. Wang, G. Yang,
plants: a valuable triterpenoid with functional properties, Food Chem. 220 G. He, Cucurbitacin B inhibits growth and induces apoptosis through the
(2017) 477–489. JAK2/STAT3 and MAPK pathways in SHSY5Y human neuroblastoma cells,
[190] S. Chirumbolo, Anticancer properties of the flavone wogonin, Toxicology Mol. Med. Rep. 10 (1) (2014) 89–94.
314 (1) (2013) 60–64. [213] Y. Dong, B. Lu, X. Zhang, J. Zhang, L. Lai, D. Li, Y. Wu, Y. Song, J. Luo, X. Pang, Z.
[191] S. Venturelli, M. Burkard, M. Biendl, U.M. Lauer, J. Frank, C. Busch, Prenylated Yi, M. Liu, E. Cucurbitacin, a tetracyclic triterpenes compound from Chinese
chalcones and flavonoids for the prevention and treatment of cancer, medicine, inhibits tumor angiogenesis through VEGFR2-mediated
Nutrition 32 (11–12) (2016) 1171–1178. Jak2-STAT3 signaling pathway, Carcinogenesis 31 (12) (2010) 2097–2104.
[192] M.O. Kim, M.H. Lee, N. Oi, S.H. Kim, K.B. Bae, Z. Huang, D.J. Kim, K. Reddy, S.Y. [214] A. Hu, J.J. Huang, X.J. Jin, J.P. Li, Y.J. Tang, X.F. Huang, H.J. Cui, W.H. Xu, G.B.
Lee, S.J. Park, J.Y. Kim, H. Xie, J.K. Kundu, Z.Y. Ryoo, A.M. Bode, Y.J. Surh, Z. Sun, Curcumin suppresses invasiveness and vasculogenic mimicry of
Dong, [6]-Shogaol inhibits growth and induces apoptosis of non-small cell squamous cell carcinoma of the larynx through the inhibition of
lung cancer cells by directly regulating Akt1/2, Carcinogenesis 35 (3) (2014) JAK-2/STAT-3 signaling pathway, Am. J. Cancer. Res. 5 (1) (2015) 278–288.
683–691. [215] P. Wang, B. Wang, S. Chung, Y. Wu, S.M. Henning, J.V. Vadgama, Increased
[193] S.M. Kim, C. Kim, H. Bae, J.H. Lee, S.H. Baek, D. Nam, W.S. Chung, B.S. Shim, chemopreventive effect by combining arctigenin, green tea polyphenol and
S.G. Lee, S.H. Kim, G. Sethi, K.S. Ahn, 6-Shogaol exerts anti-proliferative and curcumin in prostate and breast cancer cells, RSC Adv. 4 (66) (2014)
pro-apoptotic effects through the modulation of STAT3 and MAPKs signaling 35242–35250.
pathways, Mol. Carcinog. 54 (10) (2015) 1132–1146. [216] B.H. Park, J.E. Lim, H.G. Jeon, S.I. Seo, H.M. Lee, H.Y. Choi, S.S. Jeon, B.C. Jeong,
[194] Y.L. Hsu, J.Y. Hung, Y.M. Tsai, E.M. Tsai, M.S. Huang, M.F. Hou, P.L. Kuo, Curcumin potentiates antitumor activity of cisplatin in bladder cancer cell
6-shogaol, an active constituent of dietary ginger, impairs cancer lines via ROS-mediated activation of ERK1/2, Oncotarget 7 (39) (2016)
development and lung metastasis by inhibiting the secretion of 63870–63886.
CC-chemokine ligand 2 (CCL2) in tumor-associated dendritic cells, J. Agric. [217] M. Fiala, Curcumin and omega-3 fatty acids enhance NK cell-induced
Food Chem. 63 (6) (2015) 1730–1738. apoptosis of pancreatic cancer cells but curcumin inhibits
[195] A. Saha, J. Blando, E. Silver, L. Beltran, J. Sessler, J. DiGiovanni, 6-Shogaol interferon-gamma production: benefits of omega-3 with curcumin against
from dried ginger inhibits growth of prostate cancer cells both in vitro and cancer, Molecules 20 (2) (2015) 3020–3026.
in vivo through inhibition of STAT3 and NF-kappaB signaling, Cancer Prev. [218] I.T. Chiang, Y.C. Liu, F.T. Hsu, Y.C. Chien, C.H. Kao, W.J. Lin, J.G. Chung, J.J.
Res. (Phila.) 7 (6) (2014) 627–638. Hwang, Curcumin synergistically enhances the radiosensitivity of human
[196] K.J. Peerzada, A.H. Faridi, L. Sharma, S.C. Bhardwaj, N.K. Satti, B. Shashi, S.A. oral squamous cell carcinoma via suppression of radiation-induced
Tasduq, Acteoside-mediates chemoprevention of experimental liver NF-kappaB activity, Oncol. Rep. 31 (4) (2014) 1729–1737.
carcinogenesis through STAT-3 regulated oxidative stress and apoptosis, [219] E. Eskandari, E. Heidarian, S.A. Amini, J. Saffari-Chaleshtori, Evaluating the
Environ. Toxicol. 31 (7) (2016) 782–798. effects of ellagic acid on pSTAT3, pAKT, and pERK1/2 signaling pathways in
[197] A. Pandey, K. Vishnoi, S. Mahata, S.C. Tripathi, S.P. Misra, V. Misra, R. prostate cancer PC3 cells, J. Cancer Res. Ther. 12 (4) (2016) 1266–1271.
Mehrotra, M. Dwivedi, A.C. Bharti, Berberine and curcumin target survivin [220] J. Pan, Y. Lee, Q. Zhang, D. Xiong, T.C. Wan, Y. Wang, M. You, Honokiol
and STAT3 in gastric cancer cells and synergize actions of standard decreases lung cancer metastasis through inhibition of the STAT3 signaling
chemotherapeutic 5-fluorouracil, Nutr. Cancer 67 (8) (2015) pathway, Cancer Prev. Res. (Phila.) 10 (2) (2017) 133–141.
1293–1304. [221] L. Bi, Z. Yu, J. Wu, K. Yu, G. Hong, Z. Lu, S. Gao, Honokiol inhibits constitutive
[198] X. Liu, Q. Ji, N. Ye, H. Sui, L. Zhou, H. Zhu, Z. Fan, J. Cai, Q. Li, Berberine inhibits and inducible STAT3 signaling via PU.1-induced SHP1 expression in acute
invasion and metastasis of colorectal cancer cells via COX-2/PGE2 mediated myeloid leukemia cells, Tohoku J. Exp. Med. 237 (3) (2015) 163–172.
JAK2/STAT3 signaling pathway, PLoS One 10 (5) (2015) e0123478. [222] S.H. Kang, S.J. Jeong, S.H. Kim, J.H. Kim, J.H. Jung, W. Koh, D.K. Kim, C.Y. Chen,
[199] C.M. Tsang, Y.C. Cheung, V.W. Lui, Y.L. Yip, G. Zhang, V.W. Lin, K.C. Cheung, Y. Icariside II induces apoptosis in U937 acute myeloid leukemia cells: role of
Feng, S.W. Tsao, Berberine suppresses tumorigenicity and growth of inactivation of STAT3-related signaling, PLoS One 7 (4) (2012) e28706.
nasopharyngeal carcinoma cells by inhibiting STAT3 activation induced by [223] B. Song, H. Zhan, Q. Bian, J. Gu, Piperlongumine inhibits gastric cancer cells
tumor associated fibroblasts, BMC Cancer 13 (2013) 619. via suppression of the JAK1, 2/STAT3 signaling pathway, Mol. Med. Rep. 13
[200] T. Zhu, L.L. Li, G.F. Xiao, Q.Z. Luo, Q.Z. Liu, K.T. Yao, G.H. Xiao, Berberine (5) (2016) 4475–4480.
increases doxorubicin sensitivity by suppressing STAT3 in lung cancer, Am. [224] X. Li, D. Wang, Q.C. Zhao, T. Shi, J. Chen, Resveratrol inhibited non-small cell
J. Chin. Med. 43 (7) (2015) 1487–1502. lung cancer through inhibiting STAT-3 signaling, Am. J. Med. Sci. 352 (5)
[201] D.X. Li, J. Zhang, Y. Zhang, P.W. Zhao, L.M. Yang, Inhibitory effect of (2016) 524–530.
berberine on human skin squamous cell carcinoma A431 cells, Genet. Mol. [225] L.X. Zhong, Y. Zhang, M.L. Wu, Y.N. Liu, P. Zhang, X.Y. Chen, Q.Y. Kong, J. Liu,
Res. 14 (3) (2015) 10553–10568. H. Li, Resveratrol and STAT inhibitor enhance autophagy in ovarian cancer
[202] S.M. Kim, J.H. Lee, G. Sethi, C. Kim, S.H. Baek, D. Nam, W.S. Chung, S.H. Kim, cells, Cell Death Discov. 2 (2016) 15071.
B.S. Shim, K.S. Ahn, Bergamottin, a natural furanocoumarin obtained from [226] P. Zhang, B. Yang, Y.Y. Yao, L.X. Zhong, X.Y. Chen, Q.Y. Kong, M.L. Wu, C. Li, H.
grapefruit juice induces chemosensitization and apoptosis through the Li, J. Liu, PIAS3, SHP2 and SOCS3 Expression patterns in cervical cancers:
inhibition of STAT3 signaling pathway in tumor cells, Cancer Lett. 354 (1) relevance with activation and resveratrol-caused inactivation of STAT3
(2014) 153–163. signaling, Gynecol. Oncol. 139 (3) (2015) 529–535.
[203] X. Chen, M. Tan, Z. Xie, B. Feng, Z. Zhao, K. Yang, C. Hu, N. Liao, T. Wang, D. [227] L.Q. Trung, J.L. Espinoza, D.T. An, N.H. Viet, K. Shimoda, S. Nakao, Resveratrol
Chen, F. Xie, C. Tang, Inhibiting ROS-STAT3-dependent autophagy enhanced selectively induces apoptosis in malignant cells with the JAK2V617F
capsaicin-induced apoptosis in human hepatocellular carcinoma cells, Free mutation by inhibiting the JAK2 pathway, Mol. Nutr. Food Res. 59 (11)
Radic. Res. 50 (7) (2016) 744–755. (2015) 2143–2154.
[204] K. Lirdprapamongkol, H. Sakurai, S. Abdelhamed, S. Yokoyama, S. [228] X. Kong, M.Z. Ma, Y. Zhang, M.Z. Weng, W. Gong, L.Q. Guo, J.X. Zhang, G.D.
Athikomkulchai, A. Viriyaroj, S. Awale, S. Ruchirawat, J. Svasti, I. Saiki, Wang, Q. Su, Z.W. Quan, J.R. Yang, Differentiation therapy: sesamin as an
Chrysin overcomes TRAIL resistance of cancer cells through Mcl-1 effective agent in targeting cancer stem-like side population cells of human
downregulation by inhibiting STAT3 phosphorylation, Int. J. Oncol. 43 (1) gallbladder carcinoma, BMC Complement. Altern. Med. 14 (2014) 254.
(2013) 329–337. [229] P. Deng, C. Wang, L. Chen, Y. Du, X. Yan, M. Chen, G. Yang, G. He, Sesamin
[205] L. Shen, G. Zhang, Z. Lou, G. Xu, Cryptotanshinone enhances the effect of induces cell cycle arrest and apoptosis through the inhibition of signal
Arsenic trioxide in treating liver cancer cell by inducing apoptosis through transducer and activator of transcription 3 signalling in human
downregulating phosphorylated- STAT3 in vitro and in vivo, BMC hepatocellular carcinoma cell line HepG2, Biol. Pharm. Bull. 36 (10) (2013)
Complement. Altern. Med. 17 (1) (2017) 106. 1540–1548.
[206] Y. Ge, B. Yang, Z. Chen, R. Cheng, Cryptotanshinone suppresses the [230] J. Lin, Y. Chen, L. Wei, Z. Hong, T.J. Sferra, J. Peng, Ursolic acid inhibits
proliferation and induces the apoptosis of pancreatic cancer cells via the colorectal cancer angiogenesis through suppression of multiple signaling
STAT3 signaling pathway, Mol. Med. Rep. 12 (5) (2015) 7782–7788. pathways, Int. J. Oncol. 43 (5) (2013) 1666–1674.
[207] W. Li, S.M. Saud, M.R. Young, N.H. Colburn, B. Hua, Cryptotanshinone, a Stat3 [231] W. Wang, C. Zhao, D. Jou, J. Lu, C. Zhang, L. Lin, J. Lin, Ursolic acid inhibits the
inhibitor, suppresses colorectal cancer proliferation and growth in vitro, growth of colon cancer-initiating cells by targeting STAT3, Anticancer Res.
Mol. Cell. Biochem. 406 (1–2) (2015) 63–73. 33 (10) (2013) 4279–4284.
[208] L. Lu, C. Li, D. Li, Y. Wang, C. Zhou, W. Shao, J. Peng, Y. You, X. Zhang, X. Shen, [232] Y. Zhao, J. Yao, X.P. Wu, L. Zhao, Y.X. Zhou, Y. Zhang, Q.D. You, Q.L. Guo, N. Lu,
Cryptotanshinone inhibits human glioma cell proliferation by suppressing Wogonin suppresses human alveolar adenocarcinoma cell A549 migration
STAT3 signaling, Mol. Cell. Biochem. 381 (1–2) (2013) 273–282. in inflammatory microenvironment by modulating the IL-6/STAT3 signaling
[209] X. Ding, J. Chi, X. Yang, J. Hao, C. Liu, C. Zhu, X. Wang, X. Liu, Y. Niu, W. Ji, D. pathway, Mol. Carcinog. 54 (Suppl 1) (2015) E81–93.
Chen, X. Wu, Cucurbitacin B synergistically enhances the apoptosis-inducing [233] H. Dokduang, P. Yongvanit, N. Namwat, C. Pairojkul, S. Sangkhamanon, M.S.
effect of arsenic trioxide by inhibiting STAT3 phosphorylation in lymphoma Yageta, Y. Murakami, W. Loilome, Xanthohumol inhibits STAT3 activation
Ramos cells, Leuk. Lymphoma 58 (10) (2017) 2439–2451. pathway leading to growth suppression and apoptosis induction in human
[210] J. Qi, G. Xia, C.R. Huang, J.X. Wang, J. Zhang, JSI-124 (Cucurbitacin I) inhibits cholangiocarcinoma cells, Oncol. Rep. 35 (4) (2016) 2065–2072.
tumor angiogenesis of human breast cancer through reduction of STAT3 [234] J.H. Jung, M. Yun, E.J. Choo, S.H. Kim, M.S. Jeong, D.B. Jung, H. Lee, E.O. Kim, N.
phosphorylation, Am. J. Chin. Med. 43 (2) (2015) 337–347. Kato, B. Kim, S.K. Srivastava, K. Kaihatsu, A derivative of
290 C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291

epigallocatechin-3-gallate induces apoptosis via SHP-1-mediated plasminogen activator by NF-kappa B signaling pathways, Mol. Biol. Rep. 39
suppression of BCR-ABL and STAT3 signalling in chronic myelogenous (4) (2012) 4803–4808.
leukaemia, Br. J. Pharmacol. 172 (14) (2015) 3565–3578. [257] S. Mukherjee, J. Baidoo, A. Fried, D. Atwi, S. Dolai, J. Boockvar, M. Symons, R.
[235] H. Ling, H. Yang, S.H. Tan, W.K. Chui, E.H. Chew, 6-Shogaol, an active Ruggieri, K. Raja, P. Banerjee, Curcumin changes the polarity of
constituent of ginger, inhibits breast cancer cell invasion by reducing matrix tumor-associated microglia and eliminates glioblastoma, Int. J. Cancer 139
metalloproteinase-9 expression via blockade of nuclear factor-kappaB (12) (2016) 2838–2849.
activation, Br. J. Pharmacol. 161 (8) (2010) 1763–1777. [258] M.A. Papiez, W. Krzysciak, K. Szade, K. Bukowska-Strakova, M. Kozakowska,
[236] S. Erdogan, O. Doganlar, Z.B. Doganlar, R. Serttas, K. Turkekul, I. Dibirdik, A. K. Hajduk, B. Bystrowska, J. Dulak, A. Jozkowicz, Curcumin enhances the
Bilir, The flavonoid apigenin reduces prostate cancer CD44(+) stem cell cytogenotoxic effect of etoposide in leukemia cells through induction of
survival and migration through PI3 K/Akt/NF-kappaB signaling, Life Sci. 162 reactive oxygen species, Drug Des. Devel. Ther. 10 (2016) 557–570.
(2016) 77–86. [259] F.T. Hsu, Y.C. Liu, T.T. Liu, J.J. Hwang, Curcumin sensitizes hepatocellular
[237] S. Shukla, R. Kanwal, E. Shankar, M. Datt, M.R. Chance, P. Fu, G.T. MacLennan, carcinoma cells to radiation via suppression of radiation-induced
S. Gupta, Apigenin blocks IKKalpha activation and suppresses prostate NF-kappaB activity, BioMed Res. Int. 2015 (2015) 363671.
cancer progression, Oncotarget 6 (31) (2015) 31216–31232. [260] H. Wu, Q. Liu, T. Cai, Y.D. Chen, Z.F. Wang, Induction of microRNA-146a is
[238] S. Shukla, E. Shankar, P. Fu, G.T. MacLennan, S. Gupta, Suppression of involved in curcumin-mediated enhancement of temozolomide cytotoxicity
NF-kappaB and NF-kappaB-regulated gene expression by apigenin through against human glioblastoma, Mol. Med. Rep. 12 (4) (2015) 5461–5466.
IkappaBalpha and IKK pathway in TRAMP mice, PLoS One 10 (9) (2015) [261] G.H. Zhu, H.P. Dai, Q. Shen, O. Ji, Q. Zhang, Y.L. Zhai, Curcumin induces
e0138710. apoptosis and suppresses invasion through MAPK and MMP signaling in
[239] J.L. Johnson, E.G. de Mejia, Flavonoid apigenin modified gene expression human monocytic leukemia SHI-1 cells, Pharm. Biol. 54 (8) (2016)
associated with inflammation and cancer and induced apoptosis in human 1303–1311.
pancreatic cancer cells through inhibition of GSK-3beta/NF-kappaB [262] A.J. Jiang, G. Jiang, L.T. Li, J.N. Zheng, Curcumin induces apoptosis through
signaling cascade, Mol. Nutr. Food Res. 57 (12) (2013) 2112–2127. mitochondrial pathway and caspases activation in human melanoma cells,
[240] H.S. Seo, H.S. Choi, S.R. Kim, Y.K. Choi, S.M. Woo, I. Shin, J.K. Woo, S.Y. Park, Mol. Biol. Rep. 42 (1) (2015) 267–275.
Y.C. Shin, S.G. Ko, Apigenin induces apoptosis via extrinsic pathway, [263] D.V. Singh, S. Agarwal, P. Singh, M.M. Godbole, K. Misra, Curcumin
inducing p53 and inhibiting STAT3 and NFkappaB signaling in conjugates induce apoptosis via a mitochondrion dependent pathway in
HER2-overexpressing breast cancer cells, Mol. Cell. Biochem. 366 (1–2) MCF-7 and MDA-MB-231 cell lines, Asian Pac. J. Cancer Prev. 14 (10) (2013)
(2012) 319–334. 5797–5804.
[241] M. Li, M. Zhang, Z.L. Zhang, N. Liu, X.Y. Han, Q.C. Liu, W.J. Deng, C.X. Liao, [264] J.M. Kim, E.M. Noh, K.B. Kwon, J.S. Kim, Y.O. You, J.K. Hwang, B.M. Hwang,
Induction of apoptosis by berberine in hepatocellular carcinoma HepG2Cells B.S. Kim, S.H. Lee, S.J. Lee, S.H. Jung, H.J. Youn, Y.R. Lee, Curcumin suppresses
via downregulation of NF-kappaB, Oncol. Res. 25 (2) (2017) 233–239. the TPA-induced invasion through inhibition of PKCalpha-dependent
[242] H. Goto, R. Kariya, M. Shimamoto, E. Kudo, M. Taura, H. Katano, S. Okada, MMP-expression in MCF-7 human breast cancer cells, Phytomedicine 19
Antitumor effect of berberine against primary effusion lymphoma via (12) (2012) 1085–1092.
inhibition of NF-kappaB pathway, Cancer Sci. 103 (4) (2012) 775–781. [265] J. Yu, Y. Peng, L.C. Wu, Z. Xie, Y. Deng, T. Hughes, S. He, X. Mo, M. Chiu, Q.E.
[243] M. Watabe, K. Hishikawa, A. Takayanagi, N. Shimizu, T. Nakaki, Caffeic acid Wang, X. He, S. Liu, M.R. Grever, K.K. Chan, Z. Liu, Curcumin down-regulates
phenethyl ester induces apoptosis by inhibition of NFkappaB and activation DNA methyltransferase 1 and plays an anti-leukemic role in acute myeloid
of Fas in human breast cancer MCF-7 cells, J. Biol. Chem. 279 (7) (2004) leukemia, PLoS One 8 (2) (2013) e55934.
6017–6026. [266] M. Shakibaei, A. Mobasheri, C. Lueders, F. Busch, P. Shayan, A. Goel,
[244] Y. Yang, Y. Li, K. Wang, Y. Wang, W. Yin, L. Li, P38/NF-kappaB/snail pathway Curcumin enhances the effect of chemotherapy against colorectal cancer
is involved in caffeic acid-induced inhibition of cancer stem cells-like cells by inhibition of NF-kappaB and Src protein kinase signaling pathways,
properties and migratory capacity in malignant human keratinocyte, PLoS PLoS One 8 (2) (2013) e57218.
One 8 (3) (2013) e58915. [267] Q. Qiao, Y. Jiang, G. Li, Curcumin enhances the response of non-Hodgkin’s
[245] G.R. Lee, S.H. Jang, C.J. Kim, A.R. Kim, D.J. Yoon, N.H. Park, I.S. Han, Capsaicin lymphoma cells to ionizing radiation through further induction of cell cycle
suppresses the migration of cholangiocarcinoma cells by down-regulating arrest at the G2/M phase and inhibition of mTOR phosphorylation, Oncol.
matrix metalloproteinase-9 expression via the AMPK-NF-kappaB signaling Rep. 29 (1) (2013) 380–386.
pathway, Clin. Exp. Metastasis 31 (8) (2014) 897–907. [268] P.H. Killian, E. Kronski, K.M. Michalik, O. Barbieri, S. Astigiano, C.P.
[246] X.G. Mi, Z.B. Song, L.G. Sun, Y.L. Bao, C.L. Yu, Y. Wu, Y.X. Li, Cardamonin Sommerhoff, U. Pfeffer, A.G. Nerlich, B.E. Bachmeier, Curcumin inhibits
inhibited cell viability and tumorigenesis partially through blockade of prostate cancer metastasis in vivo by targeting the inflammatory cytokines
testes-specific protease 50-mediated nuclear factor-kappaB signaling CXCL1 and −2, Carcinogenesis 33 (12) (2012) 2507–2519.
pathway activation, Int. J. Biochem. Cell Biol. 73 (2016) 63–71. [269] N.K. Im, W.J. Jang, C.H. Jeong, G.S. Jeong, Delphinidin suppresses
[247] W. Tong, Q. Wang, D. Sun, J. Suo, Curcumin suppresses colon cancer cell PMA-induced MMP-9 expression by blocking the NF-kappaB activation
invasion via AMPK-induced inhibition of NF-kappaB, uPA activator and through MAPK signaling pathways in MCF-7 human breast carcinoma cells,
MMP9, Oncol. Lett. 12 (5) (2016) 4139–4146. J. Med. Food 17 (8) (2014) 855–861.
[248] L. Cao, J. Liu, L. Zhang, X. Xiao, W. Li, Curcumin inhibits H2O2-induced [270] A. Rimmon, A. Vexler, L. Berkovich, G. Earon, I. Ron, S. Lev-Ari, Escin
invasion and migration of human pancreatic cancer via suppression of the chemosensitizes human pancreatic cancer cells and inhibits the nuclear
ERK/NF-kappaB pathway, Oncol. Rep. 36 (4) (2016) 2245–2251. factor-kappaB signaling pathway, Biochem Res Int 2013 (2013) 251752.
[249] O. Berrak, Y. Akkoc, E.D. Arisan, A. Coker-Gurkan, P. Obakan-Yerlikaya, N. [271] R.H. Chou, S.C. Hsieh, Y.L. Yu, M.H. Huang, Y.C. Huang, Y.H. Hsieh, Fisetin
Palavan-Unsal, The inhibition of PI3 K and NFkappaB promoted inhibits migration and invasion of human cervical cancer cells by
curcumin-induced cell cycle arrest at G2/M via altering polyamine down-regulating urokinase plasminogen activator expression through
metabolism in Bcl-2 overexpressing MCF-7 breast cancer cells, Biomed. suppressing the p38 MAPK-dependent NF-kappaB signaling pathway, PLoS
Pharmacother. 77 (2016) 150–160. One 8 (8) (2013) e71983.
[250] J.R. Tsai, P.L. Liu, Y.H. Chen, S.H. Chou, Y.J. Cheng, J.J. Hwang, I.W. Chong, [272] J. Li, Y. Cheng, W. Qu, Y. Sun, Z. Wang, H. Wang, B. Tian, Fisetin, a dietary
Curcumin inhibits non-small cell lung cancer cells metastasis through the flavonoid, induces cell cycle arrest and apoptosis through activation of p53
adiponectin/NF-kappab/MMPs signaling pathway, PLoS One 10 (12) (2015) and inhibition of NF-kappa B pathways in bladder cancer cells, Basic Clin.
e0144462. Pharmacol. Toxicol. 108 (2) (2011) 84–93.
[251] N. Bortel, S. Armeanu-Ebinger, E. Schmid, B. Kirchner, J. Frank, A. Kocher, C. [273] J. Li, W. Qu, Y. Cheng, Y. Sun, Y. Jiang, T. Zou, Z. Wang, Y. Xu, H. Zhao, The
Schiborr, S. Warmann, J. Fuchs, V. Ellerkamp, Effects of curcumin in pediatric inhibitory effect of intravesical fisetin against bladder cancer by induction of
epithelial liver tumors: inhibition of tumor growth and alpha-fetoprotein in p53 and down-regulation of NF-kappa B pathways in a rat bladder
vitro and in vivo involving the NFkappaB- and the beta-catenin pathways, carcinogenesis model, Basic Clin. Pharmacol. Toxicol. 115 (4) (2014)
Oncotarget 6 (38) (2015) 40680–40691. 321–329.
[252] J. Rao, R.Y. Zhang, G.A. Chen, F. Li, R.W. Huang, Inhibitory effect of curcumin [274] L. Lu, D. Tang, L. Wang, L.Q. Huang, G.S. Jiang, X.Y. Xiao, F.Q. Zeng, Gambogic
on proliferation of CD34(+) acute myeloid leukemia cells and its mechanism, acid inhibits TNF-alpha-induced invasion of human prostate cancer PC3
Zhongguo Shi Yan Xue Ye Xue Za Zhi 23 (4) (2015) 1005–1008. cells in vitro through PI3 K/Akt and NF-kappaB signaling pathways, Acta
[253] F. Cao, T. Liu, Y. Xu, D. Xu, S. Feng, Curcumin inhibits cell proliferation and Pharmacol. Sin. 33 (4) (2012) 531–541.
promotes apoptosis in human osteoclastoma cell through MMP-9, [275] M.K. Pandey, V.P. Kale, C. Song, S.S. Sung, A.K. Sharma, G. Talamo, S. Dovat,
NF-kappaB and JNK signaling pathways, Int J Clin Exp Pathol 8 (6) (2015) S.G. Amin, Gambogic acid inhibits multiple myeloma mediated
6037–6045. osteoclastogenesis through suppression of chemokine receptor CXCR4
[254] H. Guo, Y.M. Xu, Z.Q. Ye, J.H. Yu, X.Y. Hu, Curcumin induces cell cycle arrest signaling pathways, Exp. Hematol. 42 (10) (2014) 883–896.
and apoptosis of prostate cancer cells by regulating the expression of [276] J. Chen, M. Zhou, Q. Zhang, J. Xu, J. Ouyang, Anticancer effect and apoptosis
IkappaBalpha, c-Jun and androgen receptor, Pharmazie 68 (6) (2013) induction of gambogic acid in human leukemia cell line K562 In vitro, Med.
431–434. Sci. Monit. 21 (2015) 1604–1610.
[255] T. Huang, Z. Chen, L. Fang, Curcumin inhibits LPS-induced EMT through [277] Y. Wang, H. Wang, W. Zhang, C. Shao, P. Xu, C.H. Shi, J.G. Shi, Y.M. Li, Q. Fu,
downregulation of NF-kappaB-Snail signaling in breast cancer cells, Oncol. W. Xue, Y.H. Lei, J.Y. Gao, J.Y. Wang, X.P. Gao, J.Q. Li, J.L. Yuan, Y.T. Zhang,
Rep. 29 (1) (2013) 117–124. Genistein sensitizes bladder cancer cells to HCPT treatment in vitro and in
[256] H. Zong, F. Wang, Q.X. Fan, L.X. Wang, Curcumin inhibits metastatic vivo via ATM/NF-kappaB/IKK pathway-induced apoptosis, PLoS One 8 (1)
progression of breast cancer cell through suppression of urokinase-type (2013) e50175.
C. Rajagopal et al. / Pharmacological Research 130 (2018) 273–291 291

[278] Y. Luo, S.X. Wang, Z.Q. Zhou, Z. Wang, Y.G. Zhang, Y. Zhang, P. Zhao, induces cell death in glioma cells by modulating NF-kappaB nuclear
Apoptotic effect of genistein on human colon cancer cells via inhibiting the translocation and caspase-3 activation, Eur. J. Pharm. Sci. 84 (2016) 116–122.
nuclear factor-kappa B (NF-kappaB) pathway, Tumour Biol. 35 (11) (2014) [290] P. Xu, F. Cai, X. Liu, L. Guo, Sesamin inhibits lipopolysaccharide-induced
11483–11488. proliferation and invasion through the p38-MAPK and NF-kappaB signaling
[279] M.H. Chung, D.H. Kim, H.K. Na, J.H. Kim, H.N. Kim, G. Haegeman, Y.J. Surh, pathways in prostate cancer cells, Oncol. Rep. 33 (6) (2015) 3117–3123.
Genistein inhibits phorbol ester-induced NF-kappaB transcriptional activity [291] L. Gai, N. Cai, L. Wang, X. Xu, X. Kong, Ursolic acid induces apoptosis via
and COX-2 expression by blocking the phosphorylation of p65/RelA in Akt/NF-kappaB signaling suppression in T24 human bladder cancer cells,
human mammary epithelial cells, Mutat. Res. 768 (2014) 74–83. Mol. Med. Rep. 7 (5) (2013) 1673–1677.
[280] S.O. Kim, M.R. Kim, [6]-Gingerol prevents disassembly of cell junctions and [292] Y.H. Lee, E. Wang, N. Kumar, R.D. Glickman, Ursolic acid differentially
activities of MMPs in invasive human pancreas cancer cells through ERK/NF- modulates apoptosis in skin melanoma and retinal pigment epithelial cells
kappa B/snail signal transduction pathway, Evid Based Complement. exposed to UV–vis broadband radiation, Apoptosis 19 (5) (2014) 816–828.
Alternat. Med. 2013 (2013) 761852. [293] J. Li, X. Liang, X. Yang, Ursolic acid inhibits growth and induces apoptosis in
[281] A.B. Kunnumakkara, A.S. Nair, K.S. Ahn, M.K. Pandey, Z. Yi, M. Liu, B.B. gemcitabine-resistant human pancreatic cancer via the JNK and
Aggarwal, Gossypin, a pentahydroxy glucosyl flavone, inhibits the PI3 K/Akt/NF-kappaB pathways, Oncol. Rep. 28 (2) (2012) 501–510.
transforming growth factor beta-activated kinase-1-mediated NF-kappaB [294] X. Zhao, K. Jiang, B. Liang, X. Huang, Anticancer effect of xanthohumol
activation pathway, leading to potentiation of apoptosis, suppression of induces growth inhibition and apoptosis of human liver cancer through
invasion, and abrogation of osteoclastogenesis, Blood 109 (12) (2007) NF-kappaB/p53-apoptosis signaling pathway, Oncol. Rep. 35 (2) (2016)
5112–5121. 669–675.
[282] C. Ishikawa, J.L. Arbiser, N. Mori, Honokiol induces cell cycle arrest and [295] S. Silvan, S. Manoharan, Apigenin prevents deregulation in the expression
apoptosis via inhibition of survival signals in adult T-cell leukemia, Biochim. pattern of cell-proliferative, apoptotic, inflammatory and angiogenic
Biophys. Acta 1820 (7) (2012) 879–887. markers during 7,12-dimethylbenz[a]anthracene-induced hamster buccal
[283] V.P. Dia, P. Pangloli, Epithelial-to-mesenchymal transition in pouch carcinogenesis, Arch. Oral Biol. 58 (1) (2013) 94–101.
paclitaxel-resistant ovarian cancer cells is downregulated by luteolin, J. Cell. [296] T.P. Hamsa, G. Kuttan, Berberine inhibits pulmonary metastasis through
Physiol. 232 (2) (2017) 391–401. down-regulation of MMP in metastatic B16F-10 melanoma cells, Phytother.
[284] K.C. Chen, C.Y. Chen, C.R. Lin, T.Y. Yang, T.H. Chen, L.C. Wu, C.C. Wu, Luteolin Res.: PTR 26 (4) (2012) 568–578.
attenuates TGF-beta1-induced epithelial-mesenchymal transition of lung [297] S. Lev-Ari, A. Starr, S. Katzburg, L. Berkovich, A. Rimmon, R. Ben-Yosef, A.
cancer cells by interfering in the PI3 K/Akt-NF-kappaB-Snail pathway, Life Vexler, I. Ron, G. Earon, Curcumin induces apoptosis and inhibits growth of
Sci. 93 (24) (2013) 924–933. orthotopic human non-small cell lung cancer xenografts, J. Nutr. Biochem.
[285] N. Zheng, P. Yuan, C. Li, J. Wu, J. Huang, Luteolin reduces BACE1 expression 25 (8) (2014) 843–850.
through NF-kappaB and through estrogen receptor mediated pathways in [298] S. Bimonte, A. Barbieri, G. Palma, D. Rea, A. Luciano, M. D’Aiuto, C. Arra, F.
HEK293 and SH-SY5Y cells, J. Alzheimers Dis. 45 (2) (2015) 659–671. Izzo, Dissecting the role of curcumin in tumour growth and angiogenesis in
[286] W. Shi, J. Deng, R. Tong, Y. Yang, X. He, J. Lv, H. Wang, S. Deng, P. Qi, D. Zhang, mouse model of human breast cancer, BioMed Res. Int. 2015 (2015) 878134.
Y. Wang, Molecular mechanisms underlying mangiferin-induced apoptosis [299] S. Bimonte, A. Barbieri, G. Palma, A. Luciano, D. Rea, C. Arra, Curcumin
and cell cycle arrest in A549 human lung carcinoma cells, Mol. Med. Rep. 13 inhibits tumor growth and angiogenesis in an orthotopic mouse model of
(4) (2016) 3423–3432. human pancreatic cancer, BioMed Res. Int. 2013 (2013) 810423.
[287] M.G. Dilshara, C.H. Kang, Y.H. Choi, G.Y. Kim, Mangiferin inhibits tumor [300] T. Takeda, M. Tsubaki, K. Sakamoto, E. Ichimura, A. Enomoto, Y. Suzuki, T.
necrosis factor-alpha-induced matrix metalloproteinase-9 expression and Itoh, M. Imano, G. Tanabe, O. Muraoka, H. Matsuda, T. Satou, S. Nishida,
cellular invasion by suppressing nuclear factor-kappaB activity, BMB Rep. 48 Mangiferin, a novel nuclear factor kappa B-inducing kinase inhibitor,
(10) (2015) 559–564. suppresses metastasis and tumor growth in a mouse metastatic melanoma
[288] W.W. Lai, S.C. Hsu, F.S. Chueh, Y.Y. Chen, J.S. Yang, J.P. Lin, J.C. Lien, C.H. Tsai, model, Toxicol. Appl. Pharmacol. 306 (2016) 105–112.
J.G. Chung, Quercetin inhibits migration and invasion of SAS human oral [301] A. Shahid, R. Ali, N. Ali, S.K. Hasan, S. Rashid, F. Majed, S. Sultana, Attenuation
cancer cells through inhibition of NF-kappaB and matrix of genotoxicity, oxidative stress, apoptosis and inflammation by rutin in
metalloproteinase-2/-9 signaling pathways, Anticancer Res. 33 (5) (2013) benzo(a)pyrene exposed lungs of mice: plausible role of NF-kappaB,
1941–1950. TNF-alpha and Bcl-2, J. Complement. Integr. Med. 13 (1) (2016) 17–29.
[289] C.J. Kiekow, F. Figueiro, F. Dietrich, L.D. Vechia, E.N. Pires, E.H. Jandrey, S.C.
Gnoatto, C.G. Salbego, A.M. Battastini, G. Gosmann, Quercetin derivative

You might also like