ArticlePDF Available

Anticancer Activity of Natural Flavonoids: Inhibition of HIF-1α Signaling Pathway

Authors:
  • 西安电子科技大学

Abstract and Figures

Rapid tumor growth is dependent on the capability of tumor blood vessels and glycolysis to provide oxygen and nutrients. Tumor hypoxia is a common characteristic of many solid tumors, and it essentially happens when the growth of the tumor exceeds the concomitant angiogenesis. Hypoxia-inducible factor 1 (HIF-1) as the critical transcription factor in hypoxia regulation is activated to adapting to this hypoxia situation. Flavonoids widely distribute in plants comprise many polyphenolic secondary metabolites, possessing broadspectrum pharmacological activities including their potentiality as anticancer agents. Due to their low toxicity, the intense effort has been made in investigating natural flavonoids and its derivatives that can be used as HIF-1α inhibitors for cancer therapy during the past few decades. In this review, we sum up the findings concerning inhibition of HIF-1α by natural flavonoids in the last few years and propose the idea of designing tumor vascular and glycolytic multi-target inhibitors with HIF-1α as one of the targets.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.net
Current Organic Chemistry, 2019, 23, 2945-2959 2945
MINI-REVIEW ARTICLE
1385-2728/19 $58.00+.00 © 2019 Bentham Science Publishers
Anticancer Activity of Natural Flavonoids: Inhibition of HIF-1α Signaling Pathway
Xiangping Deng1, Yijiao Peng1, Jingduo Zhao1, Xiaoyong Lei1, Xing Zheng1, Zhizhong Xie1 and Guotao
Tang1,*
1Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study,
University of South China, Hengyang, P.R. China
A R T I C L E H I S T O R Y
Received: October 10, 2019
Revised: November 02, 2019
Accepted: November 20, 2019
DOI:
10.2174/1385272823666191203122030
Abstract: Rapid tumor growth is dependent on the capability of tumor blood vessels and
glycolysis to provide oxygen and nutrients. Tumor hypoxia is a common characteristic of
many solid tumors, and it essentially happens when the growth of the tumor exceeds the
concomitant angiogenesis. Hypoxia-inducible factor 1 (HIF-1) as the critical transcription
factor in hypoxia regulation is activated to adapt to this hypoxia situation. Flavonoids,
widely distributed in plants, comprise many polyphenolic secondary metabolites, possess-
ing broadspectrum pharmacological activities, including their potentiality as anticancer
agents. Due to their low toxicity, intense efforts have been made for investigating natural
flavonoids and their derivatives that can be used as HIF-1α inhibitors for cancer therapy
during the past few decades. In this review, we sum up the findings concerning the inhibi-
tion of HI F-1α by natural flavonoids in the last few years and propose the idea of designing tumor vascular and
glycolytic multi-target inhibitors with HIF-1α as on e of the targets.
Keywords: HIF-1α, natural flavonoids, anticancer, inhibition, signaling pathway, hypoxia situation.
1. INTRODUCTION
Tumor hypoxia is a common characteristic of many solid tu-
mors, and it essentially happens when the growth of the tumor ex-
ceeds the concomitant angiogenesis [1, 2]. Therefore, it is not sur-
prising that many parts of developing tumor are hypoxic and ne-
crotic [3]. Hypoxia-inducible factor 1 (HIF-1) is the critical tran-
scription factor in the regulation of hypoxia. HIF-1 is closely re-
lated to the occurren ce and development of tumors, such as angio-
genesis, glycolysis, metastasis, cell survival and apoptosis [4-8].
HIF-1 is a heterodimer composed of two subunits: HIF-1α, an oxy-
gen-regulated protein, and HIF-1β that is constitutively expressed.
HIF-1α subunit is degraded fleetly in normoxic conditions and sta-
bilized under hypoxic conditions, while HIF-1β is constantly ex-
pressed as a nuclear protein. HIF-1 activity in tumors depends on
the availability of the HIF-1α subunit, the levels of which increase
under hypoxic conditions and through activation of oncogenes
and/or inactivation of tumor suppressor genes [9-12]. The prolyl
hydroxylation of HIF-1α at the oxygen-dependent degradation do-
main (ODD) is pivotal in the regulation of HIF-1α steady-state
levels. Under normoxic conditions, the proline residue on the HIF-
1α is hydroxylated, promoting the degradation of the HIF-1α. This
hydroxylation process is mediated by prolyl hydroxylase (PHD), a
series of enzymes that are dependent on oxygen, iron ions o r ascor-
bic acid and a dioxygenase superfamily of ketoglutarate. Von Hip-
pel Lindau (VHL) tumor suppressor protein facilitates the hydroxy-
lation of the HIF-1α and degrades the HIF-1α in a ubiquitinated
*Address correspondence to this author at the Guotao Tang, Institute of Pharmacy and
Pharmacology, University of South China, Hengyang, Hunan, 421001, P.R. China;
E-mail: tgtzq@163.com
form. Under hypoxic conditions, the velocity of hydroxylation and
ubiquitination descends, leading to the accumulation of HIF-1α.
HIF-1α enters the nucleus and dimerizes with HIF-1β to form a
HIF-1 transcription factor. Within the nucleus, HIF-1 binds to the
A/GCGTG sequence on the promoter region of the target gene, the
hypoxia response element (HRE), to initiate gene transcription
(Fig. 1) [13-17]. An increasing number of anticancer agents such as
flavonoids have been shown to inhibit HIF-1 activity. For many of
them, the mechanism of action has been ful ly established and in-
volves signaling pathways linked to HIF-1 mRNA expression, HIF-
1 protein translation, HIF-1 protein degradation or HIF-1 transcrip-
tional activity/DNA binding [10]. Considering that HIF-1α is over-
expressed in a majority of human cancers, natural flavonoids are
promising anticancer therapeutic agents targeting HIF-1α.
Hypoxia, CoCl2-, H2O2- or IGF-treatment can increase HIF-1α
levels. Moreover, in many cancers, growth factors, cytokines and
other signaling molecules stimulate HIF-1α synthesis by activating
the phosphatidylinositol 3 -kinase (PI3K) or mitogen-activated pro-
tein kinase (MAPK) pathways. mTOR activity is an important de-
terminant of the rate of HIF-1α protein synthesis. The constitutive
activation of receptor tyrosine kinases (such as HER2neu, BCR-
ABL, and EGFR) and/or the downstream PI3K/AKT and
RAS/MAP kinase signal transduction pathways in cancer cells lead
to increased mTOR activity and the induction of HIF-1 activity.
MAPK pathways involved in HIF-1α regulation results in the acti-
vation of ERK1-2 (also called p44/42) after activation of the up-
stream molecules Ras/Raf/MEK-1/ERK1-2. Furthermore, the chap-
erone HSP90 interacts with HIF-1α and is required for HIF-1 tran-
scriptional activity and HIF -1 protein stability [10, 11]. In addition
to some of the common and important signaling molecules men-
Guotao Tang
2946 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
tioned above, there are many signaling pathways involved in the
regulation of HIF-1.
Flavonoids are a series of compounds in which two benzene
rings (A- and B-ring s) are linked via a central 3-carbon bridge,
forming a C6-C3-C6 system. The skeleton of flavonoids consists of
2-phenyl-4H-chromen-4-one (Fig. 2). Flavonoids belong to a kind
of secondary metabolites with a wide distribution in nature. Over
5000 naturally occurring flavonoids have been characterized by
various plants. Flavonoids have been classified according to their
chemical structure and are usu ally subdivided into several sub-
groups, including anthoxanthins, flavanones, flavanonols, flavans,
anthocyanidins and iso flavonoids (Table 1) [18-21]. They are one
of the main active ingredients in medicinal plants and have a wide
range of pharmacological activities, such as anti-inflammatory ac-
tivity [22-26], antioxidant activity [27, 28], antiviral activity [29,
30], antibacterial activity [31-33], anti-platelet aggregation effects
[34, 35], cardiovascular disease protection [26, 36] and so on. Due
to their great anti-tumor potential, flavonoids have drawn a lot of
attention in the development of anticancer drugs [26, 37-40]. It has
been reported that flavonoids have anticancer and preventive effects
against several common human cancer such as stomach, prostate,
colon, breast, lung and ovarian cancers [41-45]. Flavonoids exert
their anticancer activity with different and multiple signal transduc-
tion pathways [36, 45, 46]. So far, accumulating evidence concerns
flavonoids as HIF-1 inhibitors for drug therapy [47, 48]. In the past
decade, several studies have summarized the anticancer activity and
structure-activity relationship of natural flavonoids, but few reviews
have outlined the role of natural flavonoids on HIF-1. In this re-
view, we hope to provide a more comprehensive overview of the
inhib ition of flavonoids on HIF -1 (Tables 2-7) to provide researchers
with a thought to design potent and low-toxic anticancer drug candi-
dates.
O
O
1
2
3
4
5
6
7
81'
2'
3'
4'
5'
6'
A
B
C
Fig. (2). The basic core structure of flavonoids.
2. ANTHOXANTHINS
Anthoxanthins, including flavones and flavonols, are a type of
flavonoid pigments in plants. Chrysin (5,7-Dihydroxy-2-phenyl-
4H-chromen-4-one) reduces the stability of HIF-1α by increasing
prolyl hydroxylation of the ODD, resulting in inhibiting insulin-
induced expression of HIF-1α in human prostate cancer DU145
cells [49]. Binding of HIF-1α to Hsp90 prevents HIF-1α degrada-
tion. Moreover, the expression of HIF-1α is inhibited by chrysin
through interfering with the interaction between HIF-1α and heat
shock protein 90 (Hsp90) but also the AKT signaling pathway [49].
Baicalein (Fig. 3) (5,6,7-Trihydroxy-2-phenyl-chromen-4-one) is
originally isolated from the roots of Scutellaria baicalensis and
Scutellaria lateriflora. Baicalein inhibited the cell viability of two
kinds of human ovarian cancer (OVCAR-3 and A2780/CP-70) cell
Fig. (1). Regulation of HIF-1α activity under normoxia and hypoxia. ADM: adrenomedullin; EG-VEGF: endocrine-gland-derived vascular endothelial growth
factor; GLUT: Glucose transporter; HK: Hexokinase; HRE: hypoxia response element; LDHA: lactate dehydrogenase A; LOX: lysyl oxidase; MET: Met
proto-oncogene/hepatocyte growth factor receptor; MMP-2, MMP-9 and MMP-14: matrix metalloproteinase 2, 9 and 14; OCT4: octamer binding protein 4;
PDK1: pyruvate dehydrogenase kinase 1 ; PFKL: 6-phosphofructokinase, liver type; PGM: phosphoglycerate mutase; PHD: prolyl hydroxylase; Pr: Proline;
TERT: telomerase; TGFA: transforming growth factor α; TGF-β3: transforming growth factor beta-3; Ub: ubiquitin; VEGF: vascular endothelial growth fac-
tor; VEGFR: vascular endothelial growth factor receptor; VHL: von Hippel Lindau tumor suppressor protein.
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2947
Table 1. Classification of flavonoids.
Group
Structural Fo rmula
Description
Examples
Flavone
O
O
2-phen ylchrom en-4-one
Chrysin , Baicalein, Apigenin, Luteo-
lin, Wogonin, Acacetin, Vitexin
Anthoxanthins
Flavonol
O
O
OH
3-hydro xy-2-phenylchromen-4-one
Galangin, Kaempferol,
Fisetin, Querc etin, Myri cetin,
Isorhamnetin, Rutin
Flavanones
Flavanone
O
O
2,3-dihydro-2-phenylchromen-4-one
Liquiritigenin, Naringin, Blumeatin,
Butin, Er iodictyol, Hesperetin
Flavanonols
Flavanonol
O
O
OH
3-hydro xy-2,3-dihydro-2-
phenylchromen-4-one
Taxifolin, Aromadedrin
Flavan-3-ol
O
OH
2-phen yl-3,4-dihydro-2H-chromen-3-ol
Epigallocatechin, Epigallocatechin
gallate, Proanthocyanidin, Theafla-
vin digallate
Flavan-4-ol
O
OH
2-phen yl-3,4-dihydro-2H-chromen-4-ol
Apiforol, Luteoforol
Flavans
Flavan-3,4-diol
O
OH
OH
2-phen yl-3,4-dihydro-2H-chromen-3,4-
diol
Leucocyanidin, Leucodelphinidin,
Leucofisetinidin, Leucomalvidin
Anthocyanidins
Anthocyanidin
O
2-phen ylchrom enylium ion
Delphinidin, Cyanidin, Aurantin idin,
Europinidin
Isoflavonoids
Isoflavanes
Isoflavenes
isoflavonones
rotenoids
Pterocarpans
O
O
3-phen ylchrom en-4-one
Daidzein, Genistein, Biochan in A,
Alpinumisoflavone, Deguelin
2948 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
lines in a concentration-dependent manner (LD50=39.4 µM for
OVCAR-3 at 24 h, LD50=24.3 µM for A2780/CP70 at 24 h), while
exhibited lower cytotoxicity in normal ovarian cell line IOSE-364
(LD50=68 µM), suggesting that baicalein had a certain extent of
selectivity to human ovarian cancer cells. The potential mech anism
for inhibition of human ovarian cancer cell viability by baicalein
treatment is the suppression of the cancer-promoting genes expres-
sion including HIF-1α [50]. Baicalein (IC50=78.0 µM at 48 h) at the
concentration of 50 µM inhibits HIF-1α stabilization and also sup-
presses its transcription activity in human breast cancer MCF-7
cells in vitro [51]. Baicalein also suppresses hypoxia-enhanced
glycolytic flux and the key glycolysis-associated enzymes (HK2,
LDH-A and PDK1) in the human gastric adenocarcinoma AGS
cells, and reduces HIF -1α expression via inhibiting hypoxia-
induced Akt phosphorylation. Baicalein also is found to suppress
hypoxia-induced Akt phosphorylation through the promotion of
PTEN (an upstream negative regulator of Akt) accumulation in
AGS cells, suggesting that baicalein may inhibit hypoxia-dependent
HIF-1α expression via the PTEN/Akt pathway. The research dem-
onstrates that baicalein reverses hypoxia-induced 5-FU resistance
through the suppression of glycolysis via regulation of th e
PTEN/Akt/HIF-α signaling pathway [52]. The in vivo, treatment
with baicalein (20 and 40 mg/kg/day, i.p.) has been found to sig-
nificantly suppress the intracerebral tumor growth and lengthen the
survival in the U87 orthotopic glioma models. Moreover, treatment
with baicalein decreased the expression of HIF-1α in U87 gliomas
[53]. Apigenin (Fig. 3) (5,7-dihydroxy-2-(4-hydroxyphenyl)-4H-1-
benzopyran-4-one), a natural product belonging to the flavone
class, significantly down-regulates the expression of HIF-1α mRNA
at the concentration of 50 µM in human pancreatic cancer CD18
and S2-013 cells in both normoxic and hypoxic conditions [54].
Apigenin has also been found to inhibit HIF-1α expression via in-
terfering with th e binding of HIF-1α and Hsp90 in human hepatoma
Hep3B cells, leading to the reduction of VEGF expression. In addi-
tion, apigenin degrades HIF-1α via a pVHL-independent pathway
[55, 56]. Importantly, apigenin significantly suppresses tumor angi-
ogenesis in vivo in human prostate carcinoma PC-3 cells and human
ovarian cancer cells OVCAR-3 cells which are linked to the de-
crease of HIF-1 and VEGF, via using the chicken chorioallantoic
membrane (CAM), Matrigel plug assays and immunoblotting [56].
GSK-3b is a well-characterized downstream substrate of Akt. Inhi-
bition of HIF-1α by apigenin treatment in human prostate carci-
noma PC3-M cells is accompanied by the phosphorylation (activa-
tion) of Akt and phosphorylation (inhib ition) of GSK -3b, suggest-
ing the involvement of PI3K/Akt/GSK-3 pathway in HIF-1 inhibi-
tion [57]. p70S6K1 is a downstream target of PI3K/AKT. p53 func-
tions to promote the degradation of HIF-1. HDM2 is the protein
that mediates p53 degradation by binding p53 and stimulating the
addition of ubiquitin to the carboxyl terminus of p53 for degrada-
tion.Inhibition of AKT activation impaired the HDM2 phosphoryla-
tion leading to its destabilization and degradation, which accounts
for the induction of the p53 protein. Apigenin also inhibits HIF-1
expression via PI3K/Akt/p70S6K1 and HDM2/p53 pathways in
human ovarian cancer cells OVCAR-3 and A2780/CP70 [58].
Luteolin (Fig. 3) (2-(3,4-Dihydroxyphenyl)- 5,7-dihydroxy-4-
chromenone) has a strong antican cer effect and its mechanism is
associated with the inhibition of HIF-1α in mouse macrophage
RAW264.7 cells under hypoxic conditions [59]. IC50 values of lute-
olin after 24, 48 and 78 h of incubation were 140.73, 64.94, and
44.45 µM for A375 cells and 143.89, 67.34, and 55.09 µM for B16-
F10 cells, respectively. Luteolin shows anti-metastasis activity by
reducing the p-Akt, HIF-1α, VEGF-A, p-VEGFR-2, MMP-2, and
MMP-9 proteins expression in human A375 melanoma cells and
mouse B16-F10 melanoma cells [60]. Treatment of luteolin plus
radiation reduces the expression levels of VEGF and HIF-1α in the
gastric cancer cell line SGC-7901 [61]. Luteolin reduces HIF-1α
nuclear accumulation in human cervical carcinoma (HeLa) cells,
and also inhibits HIF-1α phosphorylation and its transcriptional
activity via interfering with the MAPK pathway. Wogonin (Fig. 3)
(5,7-Dihydroxy-8-methoxy-2-phenyl-4H-chromen-4-one) is a fla-
vone with a remarkable antitumor activity. Wogonin has anti-
proliferative activity against SGC-7901 and A549 cells by a time-
and concentration-dependent manner. The inhibitory rates of SGC-
7901 and A549 cells following wogonin treatment (15 µg/mL) were
35.00±0.12 and 54.17±0.24%, respectively. Wogonin has the inhi-
bition effect of the energy metabolism, cell proliferation and angio-
genesis in SGC-7901 and human carcinoma A549 cells via decreas-
ing HIF-1α protein expression [62]. Wogonin can induce the activa-
tion of caspase-3 by inhibiting the expression of HIF-1α and sur-
vivin in human eosinophilic leukemia EoL-1 cells, thereby promot-
ing cell apoptosis [63]. Wogonin can repress H IF-1α expression and
glycolysis by suppressing PI3K/Akt signaling pathway in human
colon cancer HCT116 cells in vitro and vivo, which may be the
potential mechanism of the reversion of wogonin resistance [64].
Proto-oncogene c-Myc enforces cellular proliferation and growth in
tumors [11] and cooperates with HIF-1 in inducing VEGF expres-
sion. As c-Myc/HIF-1α signaling axis is reported to play a critical
role in angiogenesis via controlling VEGF expression and secretion,
treatment by wogonin increases HIF-1α-VHL interaction, and pro-
motes HIF-1α degradation through proteasome/ubiquitinatio n
pathway without affecting HIF-1α mRNA expression level. It has
been found that the inhibition of multiple myeloma-stimulated
angiogenesis and tumor progression by wogonin in the human mul-
tiple myeloma cell lines RPMI 8226 is associated with
c-Myc/VHL/HIF-1α signaling axis [65]. Acacetin (Fig. 3) (5,7-
dihydroxy-2-(4-methoxyphenyl)-4H-chromen-4-one)is a flavone
compound and has anti-cancer activity. Acacetin promotes the deg-
radation of HIF-1α protein and decreases the stability of HIF-1α
protein, resulting in the attenuation of HIF-1α expression. In addi-
tion , acacetin inhibits HIF-1α expression and AKT activation to
restrain VEGF expression, thereby significantly suppressing
OVCAR-3 cell-induced angiogenesis and tumor growth in vivo
[66]. Vitexin (Fig. 3) (5, 7, 4-trihydroxyflavone-8-glucoside) as an
HIF-1α inhibitor reduces the mRNA level of transcriptional genes
(VEGF, etc.) controlled by HIF-1α [67, 68], and inhibits the protein
levels HIF-1α in rat pheochromocytoma (PC12) [67]. Vitexin re-
duced HIF-1α expression in MCF-7 cells [69]. Baicalin (Fig. 3)
((2S,3S,4S,5R, 6S)-6-(5,6-dihydroxy-4-oxo-2-phenyl-chromen-7-
yl)oxy-3,4,5-trihydroxy-tetrahydropyran-2-carboxylic acid) is the
glucuronide of baicalein. LD50 is calculated for two cancer cell lines
and the normal ovarian cell line: 44.6 µM for OVCAR-3, 55.2 µM
for A2780/CP70, and 177.0 µM for IOSE-364 treated by baicalin.
Baicalin exerts antiproliferative activities in human lung carcinoma
A549 cell line and mouse Lewis lung cancer (LLC) cell line by a
concentration- and time-dependent manner [70]. It has been re-
ported that baicalin enhances SOD activity and decreases HIF-1α
stabilization to suppress cancer cell proliferation and metastasis
[70-72].
Flavonols are a class of flavonoids that have the 3-
hydroxyflavone backbone. Galangin (Fig. 4) (3,5,7-Trihydroxy-2-
phenyl-chromen-4-one), a flavonol, down-regulates the level
of HIF-1α protein in A2780/CP70 and OVCAR-3 cells [73]. The
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2949
O
O
HO
OH
Chrysin
O
O
HO
OH
Baicalein
HO
O
O
HO
OH
Apigenin
O
O
HO
OH
Luteolin
O
O
HO
OH
Wogonin
O
O
HO
OH
Acacetin
OH
OH
OCH3
OH
OCH3
O
O
O
O
HO
OH OH
OH
HO
O
HO
OH
HO OH
O
OH
OH
OH
HO
O
Vitexin Baicalin
Fig. (3). The structure of flavones inhibiting HIF-1α signaling pathway.
Table 2. Natural flavones that affect HIF-1α signaling pathway and function.
Compounds
Target
Mechanism of HIF-1 Inhibition
Cancer Cell Line
Reference
Chrysin
ODD
HIF-1α stabilization
DU145 cells
[49]
Hsp90 and AKT signaling pathway
HIF-1α expression
DU145 cells
[49]
Baicalein
Akt phosphorylation and PTEN/Akt
pathway
HIF-1α expression
OVCAR-3cells ; A2780/CP-
70 cells ; AGS cells; U87
cells
[50,52,53]
HIF-1α stabilization and transcription
activity
MCF-7 cells
[51]
Apigenin
Hsp90 or PI3K/Akt/GSK-3,
PI3K/Akt/p70S6K1 and HDM2/p53
pathways
HIF-1α expression
Hep3B cells; PC-3 cells;
PC3-M cells; OVCAR-3
cells; A2780/CP70 cells
[55-58]
HIF-1α mRNA level
CD18 cel ls; S2-013 cells
[54]
HIF-1α degradation
Hep3B cells
[55]
Luteolin
HIF-1α expression
RAW264.7 cell; A375 cells;
B16-F10 ce lls; SGC-7901
cells
[59-61]
MAPK pathway
HIF-1α phosphorylation and its transcrip-
tional activity
HeLa cells
[70]
Wogonin
PI3K/Akt signaling pathway
HIF-1α expression
SGC-7901 cells; A549 cells;
EoL-1 cells; HCT116 cells
[62-64]
proteasome/ubiquitination pathway
HIF-1α degradation
RPMI 8226 cells
[65]
Acacetin
HIF-1α degradation and HIF-1α stabiliza-
tion
OVCAR-3 cell
[66]
Vitexin
HIF-1α expression
PC12 cells; MCF-7 cells
[67-69]
Baicalin
HIF-1α stabilization
A549 cell; LLC cells
[70-72]
dietary flavonol kaempferol (Fig. 4) (3,5,7-trihydroxy-2-(4-
hydroxyphenyl)-4H-1-benzopyran-4-one) has been shown to inhibit
HIF-1 activity concentration-dependently with an IC50 value of 5.16
µM on human hepatoma (Huh7) cancer cells under hypoxic condi-
tions (1% oxygen). The mechanism is involved in the mislocaliza-
tion of HIF-1 into the cytoplasm caused by the inactivation of
2950 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
p44/42 MAPK by kaempferol (IC50=4.75 µM) [74]. Treatment of
two ovarian cancer cells (OVCAR-3 and A2780/CP70) with
kaempferol down-regulates the expression of HIF-1α and inhibits
AKT phosphorylation in a concentration-dependent manner at 5 to
20 µM concentrations. Kaempferol is shown to repress angiogene-
sis and VEGF expression on human ovarian cancer cells via HIF-
dependent (Akt/HIF) and HIF-independent (ESRRA, estrogen-
related receptor alpha) signaling path [75]. Quercetin (Fig. 4) (2-
(3,4-dihydroxyphenyl)-3,5,7-trihydroxy-4H-chrom en-4-one), a
plant flavonol, attenuates the activity of HIF-1 and suppresses tu-
mor growth in an HCT116 cancer xenograft mod el [76]. Compared
with doxorubicin alone, treatment combined with doxorubicin and
quercetin (0.7 µM) significantly represses cell proliferation and
invasion on MCF-7 and MCF-7/dox cells and inhibits HIF-1α and
permeability glycoprotein (P-gp) expression [77]. Under normoxic
conditions, a slight effect of treatment with quercetin on cell viabil-
ity and DOX-induced cytotoxicity is observed in 4T1 breast cancer
cells. Conversely, quercetin protects spleen cells from DOX-
induced toxic side effects in vitro under hypoxic conditions. Quer-
cetin inhibits HIF-1α in tumors in a hypoxia-dependent manner,
while increases its accumulation in normal cells. These results sug-
gest that quercetin can enhance the treatment index of DOX
through its opposite effect on HIF-1α in normal cells and tumors
[78]. Quercetin inhibits the accumulation of HIF-1α by suppression
of protein synthesis in a concentration-dependent manner under
hypoxic conditions on human prostate cancer LNCaP, colon cancer
CX-1, and breast cancer SkBr3 cells [79]. Quercetin also signifi-
cantly represses the transcriptional activation of HIF-1 by hypoxia
in the RIF cell line (derived from a radiation-induced fibrosarcoma)
[80]. Quercetin down-regulates the expression of HIF-1α in human
neuroblastoma SK-N-MC cell line resulting in the decrease of
Foxo3a and Notch intracellular domain (NICD) contents, accompa-
nying up-regulation of p53 and Bax as pro-apoptotic factors, and
further to cause cell death induced by oxidative stress [81].
Myricetin (Fig. 4) (3,5,7-trihydroxy-2-(3,4,5-trihydroxyphenyl)-4-
chromenone) has demonstrated potent anti-neoplastic properties
[82, 83]. Myricetin suppresses the expression of UVB-induced HIF-
1α in an SKH-1 hairless mouse skin tumorigenesis model [84].
Myricetin down-regulates the level of HIF-1α and represses the
expression of VEGF by th e Akt/p70S6K/ HIF-1α and p21/HIF-
1α/VEGF pathway in OVCAR-3 cells [73]. Isorhamnetin (Fig. 4)
(3,5,7-trihydroxy-2-(4-hydroxy-3-methoxyphenyl) chromen-4-one),
a flavonol metabolite of quercetin, markedly inhibits CoCl2-, H2O2-
or hypoxia-induced HIF-1α accumulation in colorectal cancer cells
(HCT116 and HT29). Isorhamnetin has been found to be a better
inhibitor of HIF-1 than quercetin [85].
3. FLAVANONES
Liquiritigenin (Fig. 5) ((2S)-7-Hydroxy-2-(4-hydroxyphenyl)-
2,3-dihy-dro-4H-chromen-4-one) is a dietary flav anone with vari-
ous anti-cancer activities. Liquiritigenin could block its kinase ac-
tivity in breast cancer cells (MCF-7 and MDA-MB-231) via pro-
moting HIF-1α proteasome degradation and directly interacting
with VEGFR-2, thereby remarkably inhibiting the VEGF expres-
sion [86]. Liquiritigenin significantly represses both serum - and
mimicked hypoxia-induced HIF-1α protein accumulation but does
not affect the HIF-1α mRNA level. Moreover, liquiritigenin de-
creases the stability and synthesis of HIF-1α to suppress serum-
induced HIF-1α expression in a concentration-dependent manner.
Mechanistically, liquiritigenin inhibits serum-induced HIF-1α and
VEGF expression in HeLa cells and the mechanisms correlate with
the AKT/mTOR-p70S6K signaling pathway [87]. Naringin (Fig. 5)
(7-[[2-O-(6-Deoxy-α-L-mannopyranosyl)-β-D-glucopyranosyl]ox-
y]-2,3-dihydro-5-hydro-xy-2-(4-hydroxyphenyl)-4H-1-benzopyran
-4-one) is a flavanone-7-O-glycoside and shows its antitumor ef-
fects [88, 89] via regulating cell cycle progression, promoting tu-
mor cell apoptosis, repressing angiogenesis, and inhibiting metasta-
sis and invasion. Naringin suppresses the expression of HIF-1α on
human melanoma cell line A375 and A875 cells [90].
4. FLAVANS
The flavans are benzopyran derivatives with the 2-phenyl-3,4-
dihydro-2H-chromene skeleton, including the flavan-3-ols, flavan-
O
O
OH
HO O
O
OH
HO O
O
OH
HO
O
O
OH
HO O
O
OH
HO
OH
OH
OCH3
OH
OH
OH
OH
Myricetin Isorhamnetin
Quercetin
Galangin Kaempferol
OH
OH
OH
OH
OH
OH
Fig. (4). The structure of flavonols inhibiting HIF-1α signaling pathway.
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2951
Table 3. Natural flavonols that affect HIF-1α signaling pathway and function.
Compounds
Target
Mechanism of HIF-1 Inhibition
Cancer Cell Line
reference
Galangin
HIF-1α expression
A2780/CP70 cells;
OVCAR-3 cells
[73]
Kaempferol
p44/42 MAPK pathway
HIF-1 activity
Huh7 cells
[74]
AKT phosphorylation
HIF-1α expression
OVCAR-3cells;
A2780/CP70 cells
[75]
Quercetin
HIF-1 activity
HCT116 cells
[76]
HIF-1α expression
4T1 cells; SK-N-MC cells
[77,78,81]
protein synthesis
HIF-1α accumulation
LNCaP cells; CX-1 cells;
SkBr3 cells
[79]
HIF-1 transcription activity
RIF cells
[80]
Myricetin
Akt/p70S6K/ HIF-1α and p21/HIF-
1α/VEGF pathways
HIF-1α expression
OVCAR-3 cells; SKH-1
hairless mouse skin tu-
morigenesis model
[73,84]
Isorhamnetin
HIF-1α accumulation
HCT116 cells; HT29 cells
[85]
O
O
HO
OH
O
O
O
OH
O
O
O
OH
HO
HO
HO
H3C
OHOH
OH
Liquiritigenin Naringin
Fig. (5). The structure of flavanones inhibiting HIF-1α signaling pathway.
Table 4. Natural flavanones that affect HIF-1α signaling pathway and function.
Compounds
Target
Mechanism of HIF-1 Inhibition
Cancer Cell Line
Reference
Liquiritigenin
HIF-1α proteasome
HIF-1α expression
MDA-MB-231 cells
[86]
AKT/mTOR-p70S6K pathways
HIF-1α accumulation, the stability and
synthesis of HIF-1α
HeLa cells
[87]
Naringin
HIF-1α expression
A375 cells; A875 cells
[90]
4-ols and flavan-3,4-diols (leucoanthocyanidin). Epigallocatechin
(Fig. 6) (5,7-triol,3,4-dihydro-2-(3,4,5-trihydroxyphenyl)-2H-
1-benzo-pyran-(2r-cis)) is a flavan-3-ol. Epigallocatechin as a
critical compound for LDH-A-inhibition activity of spatholobus
suberectus suppresses lactate dehydrogenase A (LDH-A) activity in
MCF-7 and MDA-MB-231 cells, owing to the disassociation of
Hsp90 from HIF-1α and subsequent accelerated HIF-1α proteasome
degradation. Epigallocatechin can significantly suppress the growth
of breast cancer and the expression of HIF-1α/LDH-A, and induce
apoptosis with no toxic effects [91]. Epigallocatechin gallate
(EGCG) ([(2R,3R)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)chro-
man-3-yl]3,4,5-trihydroxybenzoate), also called as epigalloc-
atechin-3-gallate, is the ester of epigallocatechin and gallic acid,
and is a type of catechin. EGCG inhibits cell growth and prolifera-
tion ag ainst breast cancer cells MCF-7, possibly due to the suppres-
sion of the HIF-1α and VEGF protein expression [92]. Addition-
ally, EGCG is found to observably inhibit the HIF-1α protein ex-
pression upregulated by IGF-I induction in A549 cells. EGCG re-
presses IGF-I-stimulated lung cancer angiogenesis via down-
regulating HIF-1α and VEGF expression [93]. HIF-1α-dependent
angiogenesis in vitro and in vivo is promoted by overexpression of
human papillomavirus (HPV)-16 E6 and E7 oncoproteins, which is
repressed by EGCG. Mechanistically, EGCG suppresses HPV-16
oncoprotein-induced HIF-1α protein expression while it does not
affect HIF-1α mRNA expression in non-small cell lung cancer
(NSCLC) cells (A549 and NCI-H460). Meanwhile, EGCG sup-
presses HIF-1α-dependent activation of Akt mediated by HPV-16
oncoproteins in A549 cells. Moreover, HPV-16 oncoprotein-
induced HIF-1α and HIF-1α-dependent VEGF and CD31 expres-
sions are suppressed by EGCG in A549 xenografted tumors [94].
2952 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
Treatment of human cervical carcinoma cells C-33A with EGCG
significantly represses th e HPV-16 oncoproteins-induced expres-
sion of HIF -1α, and VEGF protein and mRNA concentration-
dependently [95]. In the human pancreatic carcinoma cell line
PANC-1, EGCG inhibits the expression of the HIF-1α protein in a
concentration-dependent manner under hypoxic conditions but has
no effects on HIF-1α mRNA [96]. EGCG suppresses the accumula-
tion of HIF-1α protein in HeLa and hepatoma (HepG2) cells while
having no effects on HIF-1α mRNA expression, probably due to the
blocking of both PI3K/Akt and extracellular signal-regulated kinase
(ERKs) 1/2 signaling pathways and the acceleration of HIF-1α
protein degradation [97]. Chemically, proanthocyanidin (Fig. 6)
(2-(3,4-Dihydroxyphenyl)-2-((2-(3,4-dihydroxyphenyl)-3,4-dihy-
dro-5,7-di-hydroxy-2H-1-benzopyran-3-yl)oxy)-3,4-dihydro-2H-
1-benzopyr-an-3,4,5,7-tetrol) is oligomeric flavonoids, constituted
by oligomers of catechin and epicatechin and their gallic acid esters
or forming the group of tannins via the same polymeric building
block. Grape seed extract-standardized preparation, comprising at
least 85% (wt/wt) proanthocyanidin, decreases HIF-1α protein syn-
thesis by inhibiting Akt activation in human glioma U251 cells or
human breast cancer MDA-MB-231 cells [98]. Theaflavin digallate
(Fig. 6) ([1-[(2R,3R)-3,5-Dihydroxy-7-(3,4,5-trihydroxybenzoyl)
oxychroman-2-yl]-3,5-dihydroxy-6-oxo-8-[(3R)-3,5,7-trihydroxy-
chroman -2-yl] benzo[7]annulen-4-yl] 3,4,5-trihydroxybenzoate),
also known as theaflavin-3, 3'-digallate, is a theaflavin derivative
found in black tea. Theaflavin digallate is demonstrated to inhibi t
human ovarian carcinoma OVCAR-3 cell-induced tumor angio-
genesis by down-regulating HIF-1α and VEGF and is more effec-
tive than EGCG. The involved mechanism is that theaflavin di-
gallate inhibits activation of the Akt and Notch-1 pathway [99].
5. ANTHOCYANIDINS
Anthocyanidins are common plant pigments. They are the
sugar-free counterparts of anthocyanins based on the flavylium ion
or 2-phenylchromenylium, which is a type of oxonium ion
(chromenylium is also referred to as benzopyrylium). Delphinidin
(Fig. 7) (also named as delphinidin) (2-(3,4,5-Trihydroxyphenyl)
chromenylium-3,5,7-triol) is a kind of an anthocyanidin and pos-
sesses anti-proliferative, and anticancer activities against many
cancer cells [100-102]. Delphinidin specifically down-regulates the
expression of CoCl2- and epidermal growth factor (EGF)-induced
HIF-1α protein in A549 cells through inhibiting th e PI3K/
Akt/mTOR/p70S6K and ERK signaling pathways [103]. Besid es,
delphinidin modulates the intracellular level of Cdk inhibitor
(CDKI) protein p27 via modulating HIF-1 level and PI3K/
Akt/mTOR signaling pathway [104].
6. ISOFLAVONOIDS
Isoflavone is a type of naturally occurring isoflavonoids. Soy-
beans are the most common source of isoflavones in human food
and the major isoflavones in soybean are daidzein and genistein.
Daidzein (Fig. 8) (7-hydroxy-3-(4-hydroxyphenyl)-4H-chromen-4-
one) and genistein (Fig. 8) (5,7-Dihydroxy-3-(4-hydroxy-
O
OH
OH
OH
OH
OH
HO
O
OH
OH
OH
OH
HO
O
OOH
OH
OH
O
O
HO
HO
O
OH
OH
OH
HO
OH
OH
OH
OH
O
OH
HO
O
O
HO
OH
OH
O
HO
OH
HO
O
OH
OH
OO
OH
OH
HO
Epigallocatechin Epigallocatechin gallate
Proanthocyanidin Theaflavin digallate
Fig. (6). The structure of flavans inhibiting HIF-1α signaling path way.
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2953
phenyl)chromen-4-one), are known isoflavones that are described
as radiosensitizers for prostat e cancer [105-108]. Daidzein sup-
presses cell growth and enhances radiation in vitro, and inhibits the
expression of HIF-1α in PC-3 and C4-2B cells. Compared with
daidzein, genistein is more potent [105]. However, daidzein does
not promote metastasis to lymph nodes and guards against genis-
tein-induced metastasis [105, 107]. The expression levels of HIF-1α
and VEGF protein in SW480 cell s were markedly lower in the
normoxia group (P < 0.01, P < 0.05, respectively) and hy-
poxia/genistein group (P < 0.01, P < 0.05, respectively) than in the
hypoxia group, suggesting that genistein inhibits the expression of
hypoxia-induced HIF-1α [109, 110]. Genistein decreases the ex-
pression of VEGF mRNA by inhibiting the DNA-binding activity
of HIF-1 in human pancreatic carcinoma cell lines Capan-1 and
Mia PaCa-2 [111]. Genistein can inhibit CoCl2-induced HIF-1α
expression in a concentration-dependent manner in K562 cells but
has no effect on HIF-1α mRNA [112]. Genistein induces mitochon-
drial apoptosis through directly down-regulating HIF-1α protein in
aerobic g lycolytic hepatocellular carcinoma (HCC) cells, thereb y
inactivating GLUT1 and HK2 for inhibiting aerobic glycoly sis
[113]. Genistein as an inhibitor of tyrosine kinases can completely
inhibit the synthesis of HIF-1 subunits and HIF-1 DNA-binding
activity in Hep3B cells [114]. Biochanin A (Fig. 8) (5,7-Dihydroxy-
3-(4-methoxyphenyl)chromen-4-one) is classified as an isoflavone
known for its potential anticancer activity. Biochanin A inhibits the
expression of CoCl2-induced HIF-1α and VEGF in rat brain tumor
C6 cells, which contributes to the inhibition of angiogen esis [115].
Alpinumisoflavone (Fig. 8) (5-hydroxy-3-(4-hydroxyphenyl)-8,8-
dimethylpyrano[3,2-g]chromen-4-one), a pyranoisoflavone, inhibits
hypoxia-induced HIF-1 activation with IC50 values of 5 µm in hu-
man breast tumor T47D cells [116]. Deguelin (Fig. 8) ((7aS,13aS)-
13,13a-Dihydro-9,10-dimethoxy-3,3-dimethyl-3H-bis [1]benzopy-
rano[3,4-b:6’,5’-3]pyran-7(7aH)-one) is a derivative of rotenone
which belongs to isoflavonoid. Deguelin exhibits inhibitory effects
on the HIF transcriptional activity and the HIF-1α expression by
blocking protein synthesis and by inducing the ubiquitin-mediated
proteasome degradation pathway in H1299 lung cancer cells [117,
118]. Further evidence indicates that deguelin only marginally in-
hibits HIF-1 mRNA expression, whereas significantly inhibits the
expression of HIF-1α protein in H1299 cells under normoxic IGF-
stimulated and hypoxic conditions which is independent of ROS,
PHD, pVHL and PI3K-Akt pathways. Moreover, Hsp90 is involved
in the deguelin-mediated HIF-1α ubiquitination and destabilization.
[119]. Furthermore, HIF-1α degradation was found to be correlated
with Hsp90 binding. Inhibition of Hsp90 function by deguelin in-
hibits the increase in HIF-1α/Hsp90 interaction and HIF-1α expres-
sion in the radioresistant lung cancer cells (H1299, H226B,
H226Br, and H460) [120].
CONCLUSION AND FUTURE PERSPECTIVES
It is worth noting that global cancer mortality has not declined
over the past few decades in spite of new anticancer drugs and more
sensitive means for early detection of malignancies. Thus, there is
an urgent need for discovering new and rel iable anticancer drugs to
improve the survival rates of cancer patients. Rapid tumor growth is
dependent on the capability of tumor blood vessels and glycolysis
O
OH
OH
OH
OH
OH
HO
Delphinidin
Fig. (7). The structure of anthocyanidins inhibiting HIF-1α signaling path-
way.
Table 5. Natural flavans that affect HIF-1α signaling pathway and function.
Compounds
Target
Mechanism of HIF-1 Inhibition
Cancer Cell Line
Reference
Epigallocatechin
HIF-1α expression
MCF-7 cells; MDA-MB-
231 cells
[91]
Epigallocatechin gallate
Akt
HIF-1α expression
MCF-7 cells;
A549 cells;
C-33A cells; PANC-1 cells
[92-96]
PI3K/Akt and ERKs 1/2 signalin g
pathways
HIF-1α accumulation
HeLa cells; HepG2 cells
[97]
Proanthocyanidin
Akt activation
HIF-1α protein synthesis
U251 cells; MDA-MB-231
cells
[98]
Theaflavin digallate
Akt activation and Notch-1 path-
way
HIF-1α expression
OVCAR-3 cells
[99]
Table 6. Natural anthocyanidins that affect HIF-1α signaling pathway and function.
Compounds
Target
Mechanism of HIF-1 Inhibition
Cancer Cell Line
Reference
Delphinidin
PI3K/Akt/mTOR/p70S6K and ERK
signaling pathways
HIF-1α expression
A549 cells
[103, 104]
2954 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
O
O
HO
OH
O
O
HO
OH
O
O
HO
OCH3
OH OH
O
O
OH
O
OH
O
O
O
O
O
O
H
H
Daidzein Genistein Biochanin A
Alpinumisoflavone Deguelin
Fig. (8). The structure of isoflavonoids inhibiting HIF-1α signaling pathway.
Table 7. Natural isoflavonoids that affect HIF-1α signaling pathway and function.
Compounds
Target
Mechanism of HIF-1 Inhibition
Cancer Cell Line
Reference
Daidzein
HIF-1α expression
PC-3 cells; C4-2B cells
[105]
Genistein
HIF-1α expression
SW480 cells; K56 2 cells;
PC-3 cells; C4-2B cells;
HCC cells
[109, 110, 112, 105,
113]
HIF-1 subunits synthesis and HI F-1
DNA-binding activity
Capan-1 cells; Mia PaCa-2
cell; Hep3B cells
[111, 114]
Biochan in A
HIF-1α expression
C6 cells
[115]
Alpinumisoflavone
HIF-1 activation
T47D cells
[116]
Deguelin
protein synthesis and the ubiquitin-
mediated proteasome degradation
pathway
the HIF transcriptio nal activity and
the HIF-1α expression
H1299 cells
[117]
Hsp90
HIF-1α protein expression, HIF-1α
ubiquitination and destabilization
H1299 cells
[119]
PI3K/Akt/mTOR-mediated de novo
protein synthesis
HIF-1α/Hsp90 interaction and HIF-1α
expression
H1299 cells; H226B cells;
H226Br cells; H460 cells
[120]
to provide oxygen and nutrients. Moreover, the discrepancy be-
tween the swift rate of tumor growth and the ability of established
blood vessels to provide oxygen and nutrients make the adaptation
to hypoxia environment the foundation for the survival and growth
of tumor cells [48, 121, 122]. Most flavonoids have an inhibitory
effect on HIF -1α, and a few have stable and induced HIF-1α ef-
fects. The m ain objective of this review is focused on the inhibitio n
of HIF-1α by natural flavonoids. HIF-1α plays an important role in
hypoxia regulation as well as in cancer development. Fig. (9) repre-
sents the HIF-1α signaling pathway involved in the inhibition of
tumor vasculature, glycolysis and metastasis by natural flavonoids.
Interestingly, in addition to the inhibition of tumor angiogenesis
and glycolysis by inhibiting HIF-1α, flavonoids also block the es-
tablished tumor vascular [123]. Herein, our research group has p ro-
posed an idea of whether the flavonoids can be structurally modi-
fied to simultaneously inhibit tumor blood vessels and glycolysis,
completely severing the source of oxygen and nutrients of tumor
cells. Currently, a pharmacophore combination approach has been
proposed to design new anti-tumor molecules that act synchro-
nously on more than one target by combining two/more active
pharmacophores covalently in a single-hybrid molecule with
dual/multiple anti-tumor functions [124-126]. Moreover, most natu-
ral flavonoids have th e disadvantages of poor fat solubility or poor
water solubility, low bioavailability, and poor stability. The natural
flavonoids can be structurally modified to improve the solubility of
flavonoids and enhance physiological activity and stability. Our
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2955
group introduced amino acids or amino groups into the flavonoid
structure to improve water solubility and bioavailability. Th e phar-
macophore combination approach is feasible for the development of
flavonoids as multi-target inhibitors for cancer therapy. Under-
standing the effects of anti-cancer flavonoids on HIF-1α may be
useful for developing new compounds with enhanced anti-cancer
properties.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
Financial support from the Hunan Province Cooperative Inno-
vation Center for Molecular Target New Drug Study (0223-0002-
0002000-49) and Graduate student science foundation of the Uni-
versity of South China (2018479) are gratefully acknowledged.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or oth-
erwis e.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Folkman, J. What is the evidence that tu mors are a ngiogenesis dep endent? J.
Natl. Cancer Inst., 1990, 82(1), 4-6.
[http://dx.doi.org/10.1093/jnci/82.1.4] [PMID: 1688381]
[2] Otrock, Z.K.; Hatoum, H.A.; Awada, A.H.; Ishak, R.S.; Shamseddine, A.I.
Hypoxia-inducible factor in cancer angiogenesis: structure, regulation and
clinical perspectives. Crit. Rev. On col. Hema tol., 2009, 70(2), 93-102.
[http://dx.doi.org/10.1016/j.critrevonc.2009.01.001] [PMID: 19186072]
[3] Talks, K.L.; Turley, H.; Gatter, K.C.; Maxwell, P.H.; Pugh, C.W.; Ratcliffe,
P.J.; Harris, A .L. The expression and distribution of the h ypoxia-inducible
factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and
tumor-associated macrophages. Am. J. Pathol., 2000, 157(2), 411-42 1.
[http://dx.doi.org/10.1016/S0002-9440(10)64554-3] [PMI D: 10934146]
[4] Wang, R.; Zhou, S.; Li, S. Cancer thera peutic agents targeting hypoxia-
inducible factor-1. Curr. Med. Chem., 2011, 18(21), 3168-3189.
[http://dx.doi.org/10.2174/092986711796391606] [PMID: 21671859]
[5] Semenza, G.L. Development of novel therapeutic strateg ies that target HIF-
1. Expert Opin. Ther. Targets, 2006, 10(2), 267-280.
[http://dx.doi.org/10.1517/14728222.10.2.267] [PMID: 16548775]
[6] Denko, N.C. Hypoxia, HIF1 and gluco se metabolism in the solid tumour.
Nat. Rev. Ca ncer, 2008, 8(9), 705-713.
[http://dx.doi.org/10.1038/nrc2468] [PMID: 19143055]
[7] Giaccia, A.; Siim, B.G.; Johnson, R.S. HIF-1 as a target for drug develop-
ment. Nat. Rev. Drug Discov., 2003, 2(10), 803-811.
[http://dx.doi.org/10.1038/nrd1199] [PMID: 14526383]
[8] Melillo, G. Targeting hypoxia cell signaling for cancer therapy. Cancer
Metastasis Rev., 2007, 26(2), 341-352.
[http://dx.doi.org/10.1007/s10555-007-9059-x] [PMID: 17415529]
[9] Yewalkar, N.; Deore, V.; Padgaonkar, A.; Manohar, S.; Sahu, B.; Kumar, P.;
Jalota-Bad hwar, A.; Joshi, K.S.; Sharma, S.; Kumar, S. Dev elopment of
novel inhibitors targeting HIF-1α towards anticancer drug discovery. Bioorg.
Med. Chem. Lett., 2010, 20(22), 6426-6429.
Fig. (9). The HIF-1α signaling pathway involved in the inhibition of tumor vasculature, glycolysis and metastasis by natural flavonoids. 1:Chrysin;
2:Baicalein; 3:Apigenin; 4:Luteolin; 5:Wogonin; 6:Kaempferol; 7:Quercetin; 8:Myricetin; 9:Liquiritigenin; 10:Epigallocatechin; 11:Epigallocatechin gallate;
12:Proanthocyanidin; 13:Theaflavin digallate; 14:Delphinidin; 15 :Deguelin; AcCoA: acetyl coenzyme A; Akt: protein kinase B, also known as PKB; c-Myc: ;
CoCl2: cobalt(II) chloride; ERK: extracellular regulated protein kinase; Glucose-6-P: GLUT-1: glucose transporter 1; GSK-3: Glycogen synthase kinase 3;
HDM-2: human double minute 2; HRE: hypoxia Response Element; Hsp90: heat shock protein 90; MAPK: mitogen-activated protein kinase; MCT-4: mono-
carboxylate transporter 4 ; MMP-2 and MMP-9: matrix metalloproteinase 2 and 9; mTOR: mammalian target of rapamycin; VEGF: vascular endothelial
growth factor; VEGFR: vascular endothelial growth factor receptor; VHL: von Hippel Lindau tumor suppressor protein; P21: cyclin-dependent kinase inhibi-
tor 1; P53: tumor protein p53; p70S6K: Ribosomal protein S6 kinase beta-1; P-gp: P-glycoprotein 1; PI3K: phosphatidylinositol-3-kinase; PTEN: gene of
phosphate and tension homology de leted on chromosome ten.
2956 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
[http://dx.doi.org/10.1016/j.bmcl.2010.09.083] [PMID: 20932758]
[10] Semenza, G.L. Evaluation of HIF-1 inhibitors as anticancer agents. Drug
Discov. Today, 2007, 12(19-20), 853-859.
[http://dx.doi.org/10.1016/j.drudis.2007.08.006] [PMID: 17933687]
[11] Xia, Y.; Choi, H.K.; Lee, K. Recent ad vances in h ypoxia-inducible factor
(HIF)-1 inhibitors. Eur. J. Med. Chem., 2012, 49, 24-40.
[http://dx.doi.org/10.1016/j.ejmech.2012.01.033] [PMID: 22305612]
[12] Semenza, G.L. Hypoxia-in ducib le fact ors: me diato rs of can cer pro gressi on
and targets for cancer therapy. Trends Pha rmacol. Sci., 2012, 33(4) , 207-
214.
[http://dx.doi.org/10.1016/j.tips.2012.01.005] [PMID: 22398146]
[13] Epstein, A.C.; Gleadle, J.M.; McNeill, L.A.; Hewitson, K.S.; O’Rourke, J.;
Mole, D.R.; Mukherji, M.; Metzen, E.; Wilson, M.I.; Dhanda, A.; Tian,
Y.M.; Masson, N.; Hamilton, D.L.; Jaakk ola, P.; Barstead, R.; Hodgkin, J.;
Maxwell, P.H.; Pugh, C.W. Schofie l,d C.J.; Ratcliffe, P.J. Celegans EGL-9
and mammalian homologs define a family of d ioxygenases that regulate HIF
by prolyl hydroxylation. Cell, 2001, 10743-10 754.
[14] Huang, L.E.; Gu, J.; Schau, M.; Bunn, H.F. Regulation of hypoxia-inducib le
factor 1alpha is mediated by an O2-dependent degradation domain via the
ubiquitin-protea some p athway . Proc. Natl. Acad. Sci. USA, 1998, 95(14),
7987-7992.
[http://dx.doi.org/10.1073/pnas.95.14.7987] [PMID: 9653127]
[15] Ivan, M.; Kondo, K.; Yang, H.; Kim, W.; Valiando, J.; Ohh, M.; Salic, A.;
Asara, J.M.; Lane, W.S.; Kaelin, W.G., Jr HIFalpha targeted for VHL-
mediated destruction by proline hydroxylation: implications for O2 sensing.
Science, 2001, 292(5516), 464-468.
[http://dx.doi.org/10.1126/science.1059817] [PMID: 11292862]
[16] Salceda, S.; Caro , J. Hy poxia-inducible factor 1alpha (HIF-1alph a) pro tein is
rapidly degraded by the ubiquitin-proteasome system under normoxic condi-
tions. Its stabilization by hypox ia depends on redox-induced changes. J. Biol.
Chem., 1997, 272(36), 22642-22647.
[http://dx.doi.org/10.1074/jbc.272.36.22642] [PMID: 9278421]
[17] Kim, S.Y.; Yang, E.G. Recent advances in developing inhibitors for hypoxia-
inducible factor prolyl h ydroxylases and their ther apeutic implications.
Molecules, 2015, 20(11), 20551-20568.
[http://dx.doi.org/10.3390/molecules201119717] [PMID: 26610437]
[18] Chahar, M .K.; Sharm a, N.; Dobhal, M.P.; Josh i, Y.C. Flavon oids: A v ersatile
source o f anticancer drugs. Pharmacogn. Rev., 2011, 5(9), 1-12.
[http://dx.doi.org/10.4103/0973-7847.7909 3] [PMID: 22 096313]
[19] Nema, R.; Jain, P.; Khare, S.; Pradhan, A. Flavonoid and cancer prevention-
mini review. Res Pharm., 2015, 2, 46-50.
[20] Ververidis, F.; Trantas, E.; Douglas, C.; Vo llmer, G.; Kretzschmar, G.;
Panopoulos, N. Biotechnology of flavonoids and other phenylpropanoid-
derived natural products. Part I: Chemical diversity, impacts on plant biology
and human health. Biotechnol. J., 2007, 2(10), 12 14-1234.
[http://dx.doi.org/10.1002/biot.200700084] [PMID: 17935117]
[21] Raffa, D.; Mag gio, B.; Raim ondi, M.V.; Plescia , F.; Daidon e, G. Recent
discoveries of anticancer flavonoids. Eur. J. Med. Chem., 2017, 142, 21 3-
228.
[http://dx.doi.org/10.1016/j.ejmech.2017.07.034] [PMID: 28793973]
[22] Marzocchella, L.; Fantini, M.; Benvenuto, M.; Masuelli, L.; Tresoldi, I.;
Modesti, A.; Bei, R. Dietary flavonoids: molecular mechanisms of action as
anti- inflammatory agents. Recent Pat. Inflamm. Allergy Drug Discov., 2011,
5(3), 200-220.
[http://dx.doi.org/10.2174/187221311797264 937] [P MID: 2 1827399 ]
[23] Spagnuolo, C.; Moccia, S.; Russo, G.L. Anti-inflammatory effects o f flavon-
oids in neurodegenerative disorders. Eur. J. Med. Chem., 2018, 153, 105-
115.
[http://dx.doi.org/10.1016/j.ejmech.2017.09.001] [PMID: 28923363]
[24] Antunes-Ricardo, M.; Guti érrez-Ur ibe, J .; Sern a-Saldívar, S.O. Anti-
inflammatory glycosylated flavonoid s as therapeutic agents for treatment of
diabetes-impaired wounds. Curr. Top. Med. Chem., 2015, 15(23), 2456-
2463.
[http://dx.doi.org/10.2174/1568026615666150619141702] [PMID:
26088354]
[25] Shu, J.; Li, L.; Zhou, M.; Yu, J.; Peng , C.; Shao, F. ; Liu, R.; Zh u, G.; Huang,
H. Three new flavonoid glycosides from Smilax glabra and their anti-
inflammatory activ ity. Nat. Prod. R es., 2018, 32(15), 1760-1768.
[http://dx.doi.org/10.1080/14786419.2017.1402314] [PMID: 29149807]
[26] Benavente-García, O.; Castillo, J. Update on u ses and properties of citrus
flavonoids: new findings in anticancer, cardiovascular, and anti-
inflammatory activ ity. J. Agric. Food Chem., 2008, 56(15), 6185-6205.
[http://dx.doi.org/10.1021/jf8006568] [PMID: 18593176]
[27] Pietta, P.G. Flavonoids as antioxid ants. J. Nat. Prod., 2000, 63(7), 103 5-
1042.
[http://dx.doi.org/10.1021/np9904509] [PMID: 10924197]
[28] Agati, G.; Azzarello, E.; Po llastri, S .; Tattini, M. Flavonoids as antioxidants
in plants: location and functional significance. Plant Sci., 2012, 196, 67-76.
[http://dx.doi.org/10.1016/j.plantsci.2012.07.014] [PMID: 23017900]
[29] Min, N.; Leong, P.T.; Lee, R.C.H.; Khuan, J.S.E.; Chu, J.J.H. A flavonoid
compound library screen revealed potent antiviral activity of plant-derived
flavonoids on human enterovirus A71 replication. Antiviral Res., 2018, 150,
60-68.
[http://dx.doi.org/10.1016/j.antiviral.2017.12.003] [PMID: 29233744]
[30] Mateeva, N.; Eyunni, S.V.K.; Redda, K.K.; Ononuju, U.; Hansberry, T.D.,
II; Aikens, C.; Nag, A. Functional evaluation of synthetic flavonoids and
chalcones for potential antiviral and an ticancer properties. Bioorg. Med.
Chem. Lett., 2017, 27(11), 2350 -2356.
[http://dx.doi.org/10.1016/j.bmcl.2017.04.034] [PMID: 28442256]
[31] Xie, Y .; Yang, W.; Tang, F.; Chen, X.; Ren, L. An tibacterial activities of
flavonoids: structure-activity relationship and mechanism. Curr. Me d.
Chem., 2015, 22(1), 132 -149.
[http://dx.doi.org/10.2174/092 9867321666140916113443] [PMID:
25245513]
[32] Cushnie, T.P.; Lamb, A.J. An timicro bial activity of f lavonoids. Int. J. Antim-
icrob. Agents, 2005, 26(5), 343-35 6.
[http://dx.doi.org/10.1016/j.ijantimicag.2005.09.002] [PMID: 16323269]
[33] Cushnie, T .P.; Lam b, A.J. Recent advanc es in un derstan ding the antibacterial
properties of flavonoids. Int. J. Antimicrob. Agents, 2011, 38(2), 99-107.
[http://dx.doi.org/10.1016/j.ijantimicag.2011.02.014] [PMID: 21514796]
[34] Faggio, C.; Su reda, A.; Morabito, S.; Sanches-Silva, A.; Mocan, A .; Nabavi,
S.F.; Nabavi, S.M. Flav onoids and platelet aggregation: a brief review. Eur.
J. Pharmacol., 2017, 807, 91-101.
[http://dx.doi.org/10.1016/j.ejphar.2017.04.009] [PMID: 28412372]
[35] Wright, B.; Spencer, J.P.; Lo vegrove, J.A.; Gibb ins, J.M. Insights into die-
tary flavonoids as molecular templates for th e design of anti-platelet drugs.
Cardiov asc. Res., 2013, 97(1 ), 13-22.
[http://dx.doi.org/10.1093/cvr/cvs304] [PMID: 23024269]
[36] Grassi, D.; Desideri, G.; Cro ce, G.; T iberti, S.; Aggio , A.; Ferri, C. Flavon-
oids, vascular function and cardiovascular protection. Curr. Pharm. Des.,
2009, 15(10), 1072-1084.
[http://dx.doi.org/10.2174/138161209787846982] [PMID: 19355949]
[37] Katyal, P.; Bhardwaj, N.; Khaju ria, R. Flavonoids and their therapeutic
potential as anticancer agents: biosynthesis, metabolism and regulation.
World J. Pharm. Pharm. Sci., 2014, 3, 21 88-2216.
[38] Pathak, N.; Khan, S.; Bhargava, A.; Raghuram, G.V.; Jain , D.; Panwar, H.;
Samarth, R.M.; Jain, S.K.; Maudar, K.K.; Mishra, D.K.; Mishra, P.K. Cancer
chemopreventive effects of the flavonoid-rich fraction isolated from papaya
seeds. Nutr . Cancer, 2014, 66(5), 857-871.
[http://dx.doi.org/10.1080/01635581.2014.904912] [PMID: 24820939]
[39] Batra, P.; Sharma, A.K. Anti-can cer potential of flavonoids: recent trends
and future perspectives. 3 Biotech., 2013, 3, 439-459.
[40] Li, Y.; Fang, H.; Xu, W. Recent advance in the research of flavonoids as
anticancer agents. Mini Rev. Med. Chem., 2007, 7(7), 663-678.
[http://dx.doi.org/10.2174/138955707781024463] [PMID: 17627579]
[41] Neuhouser, M.L. Dietary flavonoids and cancer risk: evidence from human
population studies. Nutr. Cancer, 2004, 50(1 ), 1-7.
[http://dx.doi.org/10.1207/s15327914nc5001_1] [PMID: 15572291]
[42] Deng, X.; Wang, Z.; Liu, J.; Xiong, S.; Xiong, R.; Cao, X.; Chen, Y.; Zheng,
X.; Tang, G. Design, syn thesis and biolog ical evaluation of f lavonoid salicy-
late derivatives as potential antitumor ag ents. RSC Advances, 2017, 7, 38171-
38178.
[http://dx.doi.org/10.1039/C7RA07235J]
[43] Wang, Z.; Den g, X.; Xiong, R.; Xi ong, S.; Liu, J.; Cao, X.; Lei, X.; Ch en,
Y.; Zheng, X.; Tang, G. De sign, synthesis and bio logical evaluation of
3,4,5-trimethoxy flavonoid benzimidazole derivatives as potential anti-
tumor agents. MedChemComm, 2017, 9(2), 305-31 5.
[http://dx.doi.org/10.1039/C7MD00578D] [PMID: 30108924]
[44] Vue, B.; Zhang, S.; Chen, Q.H. Flavonoids with therapeutic potential in
prostate cancer. Anticancer Agents Med. Chem., 2016, 16(10), 1205-1229.
[http://dx.doi.org/10.2174/1871520615666151008122622] [PMID:
26446382]
[45] Han, D.; Tachibana, H.; Yamada, K. Inhibition of environmental estrogen-
induced proliferation of human breast car cinoma MCF-7 cells by flavonoids.
In Vitro Cell. Dev. Biol. A nim., 2001, 37(5) , 275-28 2.
[PMID: 11513082]
[46] Kandaswami, C.; Lee, L.T.; Lee, P.P.; Hwang, J.J.; Ke, F .C.; Huan g, Y.T.;
Lee, M.T. The antitumor activities of flavonoids. In Vivo, 2005, 19(5), 895-
909.
[PMID: 16097445]
[47] Hu, Y.; Liu, J.; Huang, H . Recent agents targ eting HI F-1α for cancer therapy.
J. Cell. Biochem., 2013, 114(3), 498-509.
[http://dx.doi.org/10.1002/jcb.24390] [PMID: 22961911]
[48] Manolescu, B.; Oprea, E.; Busu, C.; Cercasov, C. Natural compounds and
the hypo xia-inducib le f actor (HIF) signalling pathway . Biochimie, 2009,
91(11-12), 1347-1358.
[http://dx.doi.org/10.1016/j.biochi.2009.08.005] [PMID: 19703512]
[49] Fu, B.; Xue , J.; Li, Z.; Shi, X.; Jiang, B .H.; Fang , J. Chrysin inhib its expres-
sion of hypo xia-ind uci ble factor-1alpha through reducing hypoxia-inducible
factor-1alpha stability and inhibitin g its protein synthesis. Mol. Canc er Ther.,
2007, 6(1), 220-226.
[http://dx.doi.org/10.1158/1535-7163.MCT-06-0526] [PMID: 17237281]
[50] Chen, J.; Li, Z.; Chen, A.Y.; Ye, X.; Luo, H.; Rankin, G.O.; Chen, Y.C.
Inhibitory effect of baicalin and baicalein on ovarian cancer cells. Int. J. Mol.
Sci., 2013, 14(3), 6012-6025.
[http://dx.doi.org/10.3390/ijms14036012] [PMID: 23502466]
[51] Gade, S.; Gandhi, N.M. Baicalein inhib its MCF-7 cell proliferation in vitro,
induces radiosensitivity, and inhibits hypoxia inducible factor. J. En viron.
Pathol. Toxicol. Oncol., 2015, 34(4), 299-308.
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2957
[http://dx.doi.org/10.1615/JEnvironPatholToxicolOncol.2015013806]
[PMID: 26756423]
[52] Chen, F.; Zhuang, M.; Zhong, C.; Peng, J.; Wang, X.; Li, J.; Chen, Z.;
Huang, Y. Baicalein reverses hypoxia-indu ced 5 -FU resistance in gastric
cancer AGS cells through suppression of glycolysis and the PTEN/Akt/HIF-
1α signaling pathway. Oncol. Rep., 2015, 33(1), 4 57-463.
[http://dx.doi.org/10.3892/or.2014.3550] [PMID: 25333894]
[53] Wang, F.R.; Jiang, Y.S. Ef fect of treatme nt with baic alein on th e intrace re-
bral tumor growth and survival of orthotopic glioma models. J. Neurooncol.,
2015, 124(1), 5-11.
[http://dx.doi.org/10.1007/s11060-015-1804-3] [PMID: 25968345]
[54] Melstrom, L.G.; Salabat, M.R.; Ding, X.Z.; Strouch, M.J.; Grippo, P.J.;
Mirzoeva, S.; Pelling, J.C.; Bentrem, D.J. Apigenin down-regulates the hy-
poxia response genes: HIF-1α, GLUT-1, and VEGF in human pancreatic
cancer cells. J. Surg. Res., 2011, 167(2), 1 73-181.
[http://dx.doi.org/10.1016/j.jss.2010.10.041] [PMID: 21227456]
[55] Osada, M.; Imaoka, S.; Funae, Y. Apigenin suppresses th e expression of
VEGF, an important factor for angiogenesis, in endothelial cells via degrada-
tion of HIF-1alpha pr otein . FEBS Lett., 2004, 575(1-3), 59-63.
[http://dx.doi.org/10.1016/j.febslet.2004.08.036] [PMID: 15388333]
[56] Fang, J.; Zhou, Q.; Liu, L.Z.; Xia, C.; Hu, X.; Shi, X.; Jiang, B.H. Apigenin
inhibits tumor angiogenesis through decreasing HIF-1alpha and VEGF ex-
pression. Carcinogenesis, 2007, 28(4 ), 858-864.
[http://dx.doi.org/10.1093/carcin/bgl205] [PMID: 17071632]
[57] Mirzoeva, S.; Kim, N.D.; Chiu, K.; Franzen, C.A.; Bergan, R.C.; Pelling,
J.C. Inhibition of HIF-1 alpha and VEGF expression by the chemopreventive
bioflavonoid apigenin is accompanied by Akt inhibition in human prostate
carcinoma PC3-M cells. Mol. Carcino g., 2008, 47(9), 686-700.
[http://dx.doi.org/10.1002/mc.20421] [PMID: 18240292]
[58] Fang, J.; Xia, C.; Cao, Z.; Zhen g, J.Z.; Reed , E.; Jian g, B.H. Apig enin inhib-
its VEGF and HIF-1 expression via PI3K/AKT/p70S6K1 and HDM2/p53
pathways. FASEB J., 2005, 19(3), 342-353.
[http://dx.doi.org/10.1096/fj.04-2175com] [PMID: 15746177]
[59] Fang, B.; Chen, X.; Wu, M.; Kong, H.; Chu, G.; Zhou, Z .; Zhang, C.; Chen ,
B. Lu teolin inh ibits angiogenesis of the M2 like TAM s via th e downregu la-
tion of hypoxia inducib le factor 1α and the STAT 3 signalling pathway under
hypoxia. Mol. Med. Rep., 2018, 18(3), 2914-2922.
[http://dx.doi.org/10.3892/mmr.2018.9250] [PMID: 30015852]
[60] Li, C.; Wang, Q.; Shen, S.; Wei, X.; Li, G. HIF-1α/VEGF signaling-
mediated epithelial-mesenchymal transition and angiogenesis is critically in-
volved in anti-metastasis effect of luteolin in melanoma cells. Phytother.
Res., 2019, 33(3), 798-807.
[http://dx.doi.org/10.1002/ptr.6273] [PMID: 30653763]
[61] Zhang, Q.; Wan, L.; Guo, Y.; Cheng, N.; Cheng, W.; Sun, Q.; Zhu, J. Radio-
sensitization effect of luteolin on human gastric cancer SGC-7901 cells. J.
Biol. Regul. Homeost. Agents, 2009, 23(2), 71 -78.
[PMID: 19589287]
[62] Wang, S.J.; Zhao, J.K.; Ren , S.; Sun, W.W.; Zha ng, W.J.; Zh ang, J.N.
Wogon in affects prolifer ation and the energy metabolism of SGC-7901 and
A549 cells. Exp. Ther. Med., 2019, 17(1), 911-918.
[PMID: 30651880]
[63] Khalmuratova, R.; Lee, M.; Mo, J.H.; Jung, Y.; Park, J.W.; Shin, H.W.
Wogon in attenuate s na sal p olyp fo rmation by inducing eosinophil apoptosis
through HIF-1α and survivin suppression. Sci. Rep., 2018, 8(1), 6201.
[http://dx.doi.org/10.1038/s41598-018-24356-5] [PMID: 29670184]
[64] Wang, H.; Zhao, L.; Zhu, L.T.; Wang, Y.; Pan, D .; Yao, J.; You , Q.D.; Guo,
Q.L. Wogonin r everses hypoxia resistance of human colon cancer HCT116
cells via downregulation of HIF-1α and glycolysis, by inhibiting PI3K/Akt
signalin g pathway . Mol. Carcinog., 2014, 53(Suppl. 1), E107-E118.
[http://dx.doi.org/10.1002/mc.22052] [PMID: 23761018]
[65] Fu, R.; Chen , Y.; Wang, X.P.; An, T.; Tao, L.; Zhou, Y.X.; Huang, Y.J.;
Chen, B.A.; Li, Z.Y.; You, Q.D.; Guo, Q.L.; Wu, Z.Q. Wogonin inhibits
multiple myeloma-stimulated ang iogenesis via c-Myc/VHL/HIF-1α signaling
axis. Oncotarget, 2016, 7(5), 5715-5727.
[http://dx.doi.org/10.18632/oncotarget.6796] [PMID: 26735336]
[66] Liu, L.Z.; Jing, Y.; Jiang, L.L.; Jiang, X.E.; Jiang, Y.; Rojanasakul, Y.; Jiang,
B.H. Acacetin inhibits VEG F expression, tum or angiogenesis and growth
through AKT/HIF-1α pathway. Biochem. Biophys. Res. Commun., 2011,
413(2), 299-305.
[http://dx.doi.org/10.1016/j.bbrc.2011.08.091] [PMID: 21893035]
[67] Choi, H.J.; Eun, J.S.; Kim, B.G.; Kim, S.Y.; Jeon, H.; Soh, Y. Vitexin, an
HIF-1alpha inhibitor, has anti-metastatic potential in PC12 cells. Mol. Cells,
2006, 22(3), 291-299.
[PMID: 17202857]
[68] Scarpa, E .S.; Antonini, E.; Palma, F.; Ma ri, M .; Ninfali, P. Antipro liferativ e
activity of vitexin-2-O-xyloside and avenanthramides on CaCo-2 and HepG2
cancer cells occurs through apoptosis induction and reduction of pro-surviv al
mechanisms. Eur. J. Nutr., 2018, 57(4), 1381-1395.
[http://dx.doi.org/10.1007/s00394-017-1418-y] [PMID: 28283822]
[69] Epstein Shochet, G.; Drucker, L.; Pasmanik-Chor, M.; Po meranz, M.; Fish-
man, A.; Tartakover Matalon, S.; Lishner, M. First trimester human placental
factors induce breast cancer cell autophagy. Breast Cancer Res. Treat., 2015,
149(3), 645-654.
[http://dx.doi.org/10.1007/s10549-015-3266-x] [PMID: 25656679]
[70] Triantafyllou, A.; Mylonis, I.; Simos, G.; Bonanou, S.; Tsakalof, A. Flavon-
oids induce HIF-1alpha but impair its nuclear accumulation and activity.
Free Radic. Biol. Med., 2008, 44(4), 657-670.
[http://dx.doi.org/10.1016/j.freeradbiomed.2007.10.050] [PMID: 18061585]
[71] Du, G.; Han, G.; Zhang, S.; Lin, H.; Wu, X.; Wang, M.; Ji , L.; Lu, L.; Yu,
L.; Liang, W. Baicalin suppresses lung carcinoma and lung metastasis by
SOD mimic and HIF-1alpha inhibition. Eur. J. Pharmacol., 2010, 630(1-3) ,
121-130.
[http://dx.doi.org/10.1016/j.ejphar.2009.12.014] [PMID: 20036231]
[72] Liu, L.L.; Gong, L.K.; Wang, H.; Xiao, Y.; Wu, X.F.; Zhang, Y.H.; Xue, X.;
Qi, X.M.; Ren, J. Baicalin inhibits macrophage activation by lipopolysaccha-
ride and protects mice from endotoxin shock. Biochem. Pharmacol., 2008,
75(4), 9 14-922.
[http://dx.doi.org/10.1016/j.bcp.2007.10.009] [PMID: 18191816]
[73] Huang, H.; Chen, A.Y.; Rojanasakul, Y.; Ye, X.; Rankin, G.O.; Chen, Y.C.
Dietary compounds galangin and my ricetin suppress ovarian cancer cell an-
giogenesis. J. Funct. Foods, 2015, 15, 464-475.
[http://dx.doi.org/10.1016/j.jff.2015.03.051] [PMID: 26113875]
[74] Mylonis, I.; Lakka, A.; Tsakalof, A.; Simos, G. The dietary flavonoid
kaempferol effectively inhibits HIF-1 activity and hepatoma cancer cell vi-
ability under hypoxic conditions. Biochem. Biophys. Res. Commun., 2010,
398(1), 74-78.
[http://dx.doi.org/10 .1016 /j.bbrc.2 010.0 6.038] [PMID: 2055 8139]
[75] Luo, H.; Rankin, G.O.; Liu, L.; Daddysman, M.K.; Jiang, B.H.; Chen, Y.C.
Kaempferol inhibits ang iogenesis and VEGF expression through both HIF
dependent and independent pat hways in hu man ovar ian cancer cells. Nutr.
Cancer, 2009, 61 (4), 554-563.
[http://dx.doi.org/10.1080/01635580802666281] [PMID: 19838928]
[76] Kim, H.S.; Wannatung, T.; Lee, S.; Yang, W.K.; Chung, S.H.; Lim, J.S.;
Choe, W.; Kang, I.; Kim, S.S.; Ha , J. Que rcetin enhances hypoxia-mediated
apoptosis via direct inhibition of AMPK activity in HCT116 colon cancer.
Apoptosis, 2012, 17(9), 938-949.
[http://dx.doi.org/10.1007/s10495-012-0719-0] [PMID: 22684842]
[77] Li, S.Z.; Li, K.; Zhang, J.H.; Dong, Z. The effect of quercetin on doxorubicin
cytotoxicity in human breast cancer cells. Anticancer Agents Med. Chem.,
2013, 13(2), 352-355.
[http://dx.doi.org/10.2174/1871520611313020020] [PMID: 22721393]
[78] Du, G.; Lin, H.; Wang, M.; Zhan g, S.; Wu, X.; Lu, L.; Ji , L.; Yu, L . Quer-
cetin greatly improved therapeutic index of doxorubicin against 4T1 breast
cancer by its opposing effects on HIF-1α in tumor and normal cells. Cancer
Chemoth er. Pharmacol., 2010, 65(2), 277-287.
[http://dx.doi.org/10.1007/s00280-009-1032-7] [PMID: 19466611]
[79] Lee, D.H.; Lee, Y.J. Qu ercetin supp resses hypox ia-induced accumulation of
hypoxia-induci ble factor -1alpha (HIF-1alpha) through inhibiting protein syn-
thesis. J. Cell. Biochem., 2008, 105(2), 546-553.
[http://dx.doi.org/10.1002/jcb.21851] [PMID: 18655183]
[80] Baek, S.H.; Lee, U.Y.; Park, E.M.; Han, M.Y.; Lee, Y.S.; Park, Y.M. Role of
protein kinase Cdelta in transmitting hypoxia signal to HSF and HIF-1. J.
Cell. Physiol., 2001, 188(2) , 223-235.
[http://dx.doi.org/10.1002/jcp.1117] [PMID: 11424089]
[81] Roshanzamir, F.; Yazdanparast, R. Quercetin attenuates cell apoptosis of
oxidant-stressed SK-N-MC cells while suppressing up-regulation of the de-
fensive element, HIF-1α. Ne uroscien ce, 2014, 277, 7 80-793.
[http://dx.doi.org/10.1016/j.neuroscience.2014.07.036] [PMID: 25108166]
[82] Devi, K.P.; Rajavel, T.; Habtemariam, S.; Nabavi, S.F.; Nabavi, S.M. Mo-
lecular mech anisms underlying an ticancer effects of myricetin. Life Sci.,
2015, 142, 19-25.
[http://dx.doi.org/10.1016/j.lfs.2015.10.004] [PMID: 26455550]
[83] Mondal, S.; Jana, J.; Sengupta, P.; Jana, S.; Chatterjee, S. Myricetin arrests
human telomeric G-quadr uplex structur e: a new mechanistic ap proach as a n
anticancer agent. Mol. Biosyst., 2016, 12(8), 2506-2518.
[http://dx.doi.org/10.1039/C6MB00218H] [PMID: 27249025]
[84] Jung, S.K.; Lee, K.W.; Byun , S.; L ee, E.J.; Kim, J.E .; Bode, A.M.; Dong, Z.;
Lee, H.J. Myricetin inhibits UVB-induced angiogenesis by regulating PI-3
kinase in vivo. Carcinogenesis, 2010, 31(5), 911-917.
[http://dx.doi.org/10.1093/carcin/bgp221] [PMID: 20008033]
[85] Seo, S.; Seo, K.; Ki , S.H.; Shin, S.M. Isorhamn etin inhibits reactive o xygen
species-dependent hypoxia inducible factor (HIF)-1α accumulation. Biol.
Pharm. Bull., 2016, 39(11), 1830-1838.
[http://dx.doi.org/10.1248/bpb.b16-00414] [PMID: 27803454]
[86] Wang, Z.; Wan g, N.; Han, S.; Wang, D.; Mo, S.; Yu, L .; Huang , H.; Tsui,
K.; Shen, J.; Chen, J. Dietary compound isoliquiritigenin inhibits breast can-
cer neoangiogenesis via VEGF/VEGFR-2 signaling pathway. PLoS One,
2013, 8(7)e68566
[http://dx.doi.org/10.1371/journal.pone.0068566] [PMID: 23861918]
[87] Xie, S.R.; Wang, Y.; Liu, C.W.; Luo, K.; Cai, Y.Q. Liquiritigenin inhibits
serum-induced HIF-1α and VEGF expression via the AKT/mTOR-p70S6K
signallin g pathway in HeLa c ells. Phytother. Res., 2012, 26(8), 1 133-1141.
[http://dx.doi.org/10.1002/ptr.3696] [PMID: 22170854]
[88] Mohamed, E.A.; Abu Hashim, I.I.; Yusif, R.M.; Shaaban, A.A.A.; El-
Sheakh, A.R.; Hamed, M.F.; Badria, F.A.E. Polym eric micelles for potenti-
ated antiulcer and anticancer activities of naringin. Int. J. Nan omedici ne,
2018, 13, 1009-1027.
[http://dx.doi.org/10.2147/IJN.S154325] [PMID: 29497294]
2958 Current Organic Chemistry, 2019, Vol. 23, No. 26 Deng et al.
[89] Bharti, S.; Rani, N. ; Kr ishnam urthy, B.; Arya, D .S. Preclinical eviden ce f or
the pharmacological actions of naringin: a review. Planta Med., 2014, 80(6),
437-451.
[http://dx.doi.org/10.1055/s-0034-1368351] [PMID: 24710903]
[90] Guo, B.; Zhang, Y.; Hui, Q.; Wang, H.; Tao, K. Naringin suppresses the
metabolism of A375 cells by inhibiting the phosphorylation of c-Src. Tumour
Biol., 2016, 37(3), 3841-3850.
[http://dx.doi.org/10.1007/s13277-015-4235-z] [PMID: 2647 6533]
[91] Wang, Z.; Wang, D.; Ha n, S.; Wang, N.; M o, F.; L oo, T.Y .; Shen, J .; Huang,
H.; Chen, J. Bioactiv ity-guided identification and cell signaling technology
to delineate the lactate dehydrogenase A inh ibition effects of Spatholobus
suberectu s on bre ast cancer. PLoS One, 2013, 8(2)e56 631
[http://dx.doi.org/10.1371/journal.pone.0056631] [PMID: 23457597]
[92] Luo, H.Q.; Xu, M.; Zhong, W.T.; Cui, Z.Y.; Liu, F.M.; Zhou, K.Y.; Li, X.Y.
EGCG decreases the expression of HIF-1α and VEGF and cell growth in
MCF-7 breast cancer cells. J. BUON, 2014, 19(2), 435 -439.
[PMID: 24965403]
[93] Li, X.; Feng, Y.; Liu, J.; Feng, X.; Zhou, K.; Tang , X. Epigallocatechin-3-
gallate inhibits IGF-I-stimulated lung ca ncer angiog enesis thro ugh down-
regulation of HIF-1α and VEGF expression. J. Nutrigenet. Nutrigenomics,
2013, 6(3), 169-178.
[http://dx.doi.org/10.1159/000354402] [PMID: 24008975]
[94] He, L.; Zhang, E.; Shi, J.; Li, X.; Zho u, K.; Zhang, Q.; Le, A.D.; Tang, X. (-
)-Epigallocatechin-3-gallate inhibits human papillomavirus (HPV)-16 onco-
protein-induced angiogenesis in non-small cell l ung cancer cells by targeting
HIF-1α. Cancer Chemother. Pha rmacol., 2013, 71(3), 713-725.
[http://dx.doi.org/10.1007/s00280-012-2063-z] [PMID: 2329 2117]
[95] Tang, X.D.; Zhou, X.; Zhang, Q.Z.; Le, A.D.; Zhou, K.Y. Effects of green
tea extract on expression of human papillomavirus type 16 oncoproteins-
induced hypoxia-in ducib le f actor -1alpha and vascular endothelial growth
factor in human cervical carcinoma cells. Zhonghu a Yi Xue Za Zh i, 2008,
88(40) , 2872-2877.
[PMID: 19080502]
[96] Zhu, Z.; Wang, Y.; Liu, Z.; Wang, F.; Zhao, Q. Inhibitory effects of epigal-
locatechin-3-gallate on cell proliferation and the expression of HIF-1α and P-
gp in the human pancreatic carcinoma cell line PANC-1. Oncol. Rep., 2012,
27(5), 1 567-1572.
[http://dx.doi.org/10.3892/or.2012.1697] [PMID: 22367292]
[97] Zhang, Q.; Tang, X.; Lu, Q.; Zhang, Z.; Rao, J.; Le, A.D. Green tea extract
and (-)-epigall ocate chi n-3-gallate inhibit hypoxia- and serum-induced HIF-
1alpha protein accumulation and VEGF expression in human cervical carci-
noma and hepatoma cells. Mol. Cancer Ther., 2006, 5(5), 1227-1238.
[http://dx.doi.org/10.1158/1535-7163.MCT-05-0490] [PMID: 16731755]
[98] Lu, J.; Zhang, K.; Chen, S.; Wen, W. Grape seed extract inhibits VEGF
expression via reducing HIF-1alpha protein expression. Carcin ogenesis,
2009, 30(4), 636-644.
[http://dx.doi.org/10.1093/carcin/bgp009] [PMID: 19131542]
[99] Gao, Y.; Rankin, G.O.; Tu, Y.; Chen, Y.C. Theaflavin-3, 3-digallate de-
creases h uman ovarian carcinoma OVCAR-3 cell-induced angiogenesis via
Akt and Notch-1 pathways, not via MAPK pathways. Int. J. Oncol., 2016,
48(1), 2 81-292.
[http://dx.doi.org/10.3892/ijo.2015.3257] [PMID: 26648098]
[100] Pal, H.C.; Sharma, S.; Str ickland, L.R.; Agarwal, J.; Atha r, M.; Elmets, C.A.;
Afaq, F. Delphinidin reduces cell proliferation and induces apoptosis of non-
small-cell lung cancer cells by targeting EGFR/VEGFR2 signaling pathways.
PLoS One, 2013, 8(10), e77 270
[http://dx.doi.org/10.1371/journal.pone.0077270] [PMID: 24124611]
[101] Tsai, T.C.; Huang, H.P.; Chang, K.T.; Wang, C.J.; Chang, Y.C. Anthocyan-
ins from roselle extract arrest cell cycle G2/M phase transition via ATM/Chk
pathway in p53-deficient leukemia HL-60 cell s. Environ. Toxicol., 2017,
32(4), 1 290-1304.
[http://dx.doi.org/10.1002/tox.22324] [PMID: 27444805]
[102] Martin, S.; Favot, L.; Matz, R.; Lugnier, C.; Andriantsitohaina, R. Del-
phinidin inhibits endothelial cell proliferation and cell cycle progression
through a transien t activation of ERK-1/-2. biochem Pharmacol., 2003, 65,
669-675.
[103] Kim, M.H.; Jeong, Y.J.; Cho, H.J.; Hoe, H.S.; Park, K .K.; Park, Y.Y.; Choi,
Y.H.; Kim, C.H .; Chan g, H.W.; Park , Y.J.; Chung, I.K.; Chang, Y.C. Del-
phinidin inhibits angiogenesis through the suppression of HIF-1α and VEGF
expression in A549 lung cancer cells. Oncol. Rep., 2017, 37(2), 777-784.
[http://dx.doi.org/10.3892/or.2016.5296] [PMID: 27 959445]
[104] Quintos, L.; Lee, I.A.; Kim, H.J.; Lim, J.S.; Park, J.; Sung, M.K.; Seo, Y.R.;
Kim, J.S. Significance of p27 as potential biomarker for intracellular oxida-
tive status. Nutr. R es. Pract., 2010, 4(5), 351-355.
[http://dx.doi.org/10.4162/nrp.2010.4.5.351] [PMID: 21103079]
[105] Singh-Gup ta, V.; Zhan g, H.; Yun ker, C.K.; Ah mad, Z.; Z wier, D.; Sarkar ,
F.H.; Hillman, G.G . Daidzein eff ect on hormone refractory prostate cancer in
vitro and in vivo compared to genistein and soy extract: potentiation of radio-
therapy. Pharm. Res., 2010, 27(6), 1115 -1127.
[http://dx.doi.org/10.1007/s11095-010-0107-9] [PMID: 20309614]
[106] Raffoul, J.J.; Ban erjee, S.; Che, M.; Kn oll, Z.E.; Doerge, D.R.; Abrams, J.;
Kucuk, O.; Sarkar, F.H.; Hillman, G.G. Soy iso flavones e nhance radiother-
apy in a m etastatic prostate cancer model. Int. J. Cancer, 2007, 120(11),
2491-2498.
[http://dx.doi.org/10.1002/ijc.22548] [PMID: 17304503]
[107] Hillman, G.G.; Wang, Y.; Kucuk, O.; Che, M.; Doerge, D.R.; Yudelev, M.;
Joiner, M.C.; Marples, B.; Forman, J.D.; Sark ar, F.H. Gen istein potentiates
inhibition o f tumor growth by radiation in a prostate cancer orthotopic
model. Mol. Cancer Ther., 2004, 3(10), 1271-1279.
[PMID: 15486194]
[108] Singh-Gup ta, V.; Zh ang, H.; Ban erjee, S .; Kong, D. ; Raffou l, J.J.; Sar kar,
F.H.; Hil lman, G.G. Radiation-induced HIF-1alpha cell survival pathway is
inhibited by soy isoflavones in prostate cancer cells. Int. J. Cancer, 2009,
124(7), 1675-1684.
[http://dx.doi.org/10.1002/ijc.24015] [PMID: 19101986]
[109] Jiang, C.Q.; Fan, L.F .; Liu, Z.S.; Q ian, Q.; Xia, D.; Diao, L.M.; He, Y.M.;
Ai, Z.L. Ex pression levels and significance o f hypoxia inducible factor-1 al-
pha and vascular endothelial growth factor in human colorectal adenocarci-
noma. Chin. Med. J. (Engl.), 2004, 117(10), 1541-1546.
[PMID: 15498380]
[110] Fan, L.F.; Diao , L.M.; Jiang, C.Q.; Tang, Z.J .; Xia, D.; Liu, M.Q.; Liu , Z.S.;
Ai, Z.L. [Expression and pathobiological implication of hypoxia-inducible
factor-1alpha in human colorectal carcinoma]. Zhonghua Bing Li Xue Za Zhi,
2004, 33(3), 242-246.
[PMID: 15256117]
[111] Büchler, P.; Reber, H.A.; Büc hler, M. W.; Friess, H.; Lavey, R.S.; Hines, O.J .
Antiangiogenic activity of genistein in pancreatic carcinoma cells is medi-
ated by the inhibition of hypoxia-inducible factor-1 and the down-regulation
of VEGF gene expression. Ca ncer, 2004, 100(1), 201-210.
[http://dx.doi.org/10.1002/cncr.11873] [PMID: 14692041]
[112] Li, G.Q.; Zhang, Y.; Shen, W.G.; Zhou, W.; Gao, N.; Gu, J. [Inhibitory
effect of genistein on hypoxia-inducible factor-1alpha expression induced by
cobalt ch loride in leukem ia cell line K562]. Zhongguo Shi Yan Xu e Ye Xue
Za Zhi, 2008, 16(1), 38-43.
[PMID: 18315897]
[113] Li, S.; Li, J.; Dai, W.; Zhang, Q.; Feng, J.; Wu, L.; Liu, T.; Yu , Q.; Xu, S.;
Wang, W.; Lu, X.; Ch en, K.; Xia, Y.; Lu , J.; Z hou, Y.; Fa n, X.; Mo, W.; Xu,
L.; Guo, C. Genistein suppresses aerobic glycolysis and induces hepatocellu-
lar carcinoma cell death . Br. J. Cancer, 2017, 117(10), 1518-1528.
[http://dx.doi.org/10.1038/bjc.2017.323] [PMID: 289265 27]
[114] Wang, G.L.; Jian g, B.H.; Semenza, G.L. Effect of protein k inase and ph os-
phatase inhibitors on expression of hypoxia-inducible factor 1. Biochem.
Biophys. Res. Commun., 1995, 216(2), 6 69-675.
[http://dx.doi.org/10.1006/bbrc.1995.2674] [PMID: 7488163]
[115] Jain, A .; Lai, J. C.; Bhushan, A. Biochanin A inhibits endothelial cell f unc-
tions and proangiogenic pathways: implications in glioma th erapy. Antican-
cer Drugs, 2015, 26(3), 323 -330.
[http://dx.doi.org/10.1097/CAD.0000000000000189] [PMID: 25501542]
[116] Liu, Y.; Veena, C.K.; Morgan, J.B.; Mohammed, K.A.; Jekabsons, M.B.;
Nagle, D.G.; Zhou, Y.D. Methylalpinumisoflavone inhibits hypoxia-
inducible factor-1 (HIF-1) activation by simultaneously targeting multiple
pathways. J. Biol. Chem., 2009, 284(9), 5 859-5868.
[http://dx.doi.org/10.1074/jbc.M806744200] [PMID: 19091749]
[117] Oh, S.H.; Woo, J.K.; Jin, Q.; Kang, H.J.; Jeong, J.W.; Kim, K.W.; Hong,
W.K.; Lee, H.Y. Identifica tion of novel antiangiogenic a nticancer activities
of deguelin targeting hypoxia-indu cible fa ctor-1 alpha. Int. J. Cancer, 2008,
122(1), 5-14.
[http://dx.doi.org/10.1002/ijc.23075] [PMID: 17764071]
[118] Wang, Y.; Ma, W.; Zheng , W. Deguelin, a nov el anti-tum origenic agent
targeting apoptosis, cell cycle arrest and anti-angiogenesis for cancer chemo-
prevention. Mol. Clin . Oncol., 2013, 1(2), 215-219.
[http://dx.doi.org/10.3892/mco.2012.36] [PMID: 24649149]
[119] Oh, S.H.; Woo , J.K.; Yazici, Y.D.; Myers, J.N.; Kim, W .Y.; Jin, Q.; Hong ,
S.S.; Park, H.J.; Su h, Y.G.; Kim, K.W.; Hong, W.K.; Lee, H.Y. Stru ctural
basis for depletion of heat shock protein 90 client proteins by deguelin. J.
Natl. Cancer Inst., 2007, 99(12), 949-961.
[http://dx.doi.org/10.1093/jnci/djm007] [PMID: 17565155]
[120] Kim, W .Y.; Oh, S.H.; Woo, J.K.; Hong, W.K.; Lee, H.Y. Targeting heat
shock protein 90 overrides th e resistance of lung cancer cells by blocking ra-
diation-induce d stab ilization o f h ypoxia-indu cible f actor -1alpha. Cancer
Res., 2009, 69(4), 1624-1632.
[http://dx.doi.org/10.1158/0008-5472.CAN-08 -0505] [PMID: 19176 399]
[121] Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: the next generation. Cell,
2011, 144(5), 646-674.
[http://dx.doi.org/10.1016/j.cell.2011.02.013] [PMID: 21376230]
[122] Semenza, G.L. Regulation of metabolism by hypoxia-inducible factor 1.
Cold Sp ring Ha rb. Symp. Quant. B iol., 2011, 76, 347-353 .
[http://dx.doi.org/10.1101/sqb.2011.76.010678] [PMID: 21785006]
[123] Harris, S.R.; Panaro, N.J.; Thorgeirsson, U.P. Oxidative stress contributes to
the anti-prolif erative eff ects of flav one acetic acid on endoth elial cells. Anti-
cancer Res., 2000, 20(4), 2249-2254.
[PMID: 10953282]
[124] Conesa-Milián, L.; Falomir, E.; Murga, J.; Carda, M.; Meyen, E.; Liekens,
S.; Alb erto Marco, J. Synthesis and biological evaluation of carbamates de-
rived from aminocombretastatin A-4 as vascular disrupting agents. Eur. J.
Med. Chem., 2018, 147, 183-19 3.
[http://dx.doi.org/10.1016/j.ejmech.2018.01.058] [PMID: 29432949]
[125] Mita, M.M.; Sargsyan, L.; Mita, A.C.; Spear, M. Vascular-disrupting agents
in oncology. Expert Opin. Investig. Drugs, 2013, 22(3), 317-328.
[http://dx.doi.org/10.1517/13543784.2013.759557] [PMID: 23316880]
Anticance r Activity of Natural Fla vonoid s Current Organic Chem istry, 2019, Vol. 23 , No. 26 2959
[126] Pérez-Pérez, M.J.; Priego, E.M.; Bueno, O.; Mar tins, M.S.; Canela, M.D.;
Liekens, S. Blocking blood flow to solid tumors by destabilizing tubulin: an
approach to targeting tumor growth. J. Med. Chem., 2016, 59(19), 868 5-
8711.
[http://dx.doi.org/10.1021/acs.jmedchem.6b00463] [PMID: 27348355]
... The uncontrolled and rapid proliferation of tumor cells and insufficient formation of new blood vessels lead to inadequate oxygen supply to tumor tissues. Therefore, it is not unusual for developing malignant tissue to possess hypoxic and necrotic areas [4]. A cell's adaptation to a lower level of oxygen in hypoxic regions is accompanied by the activation of several survival pathways [5]. ...
... Flavonoids, a class of naturally occurring plantderived compounds, exert numerous beneficial human health attributes [10]. Flavonoids modulate various signaling pathways associated with cancer initiation, promotion, and progression, both in vitro and in vivo [4,[11][12][13]. The effects of flavonoids on the regulatory cascade connected to HIF-1 and glucose metabolism constitute a promising way to inhibit metabolic reprogramming via the regulation of HIF-1 activity, as well as critical components of glycolysis. ...
... HIF-1 is a critical transcription factor targeting essential genes contributing to the Warburg phenotype. Therefore, it is necessary to comprehensively discuss the effects of secondary plant metabolites on HIF-1 activity [4]. In this regard, we summarize the current status of experimental studies that focus on the modulation of HIF-1 activity by flavonoids ...
Article
Full-text available
Tumor hypoxia is described as an oxygen deprivation in malignant tissue. The hypoxic condition is a consequence of an imbalance between rapidly proliferating cells and a vascularization that leads to lower oxygen levels in tumors. Hypoxia-inducible factor 1 (HIF-1) is an essential transcription factor contributing to the regulation of hypoxia-associated genes. Some of these genes modulate molecular cascades associated with the Warburg effect and its accompanying pathways and, therefore, represent promising targets for cancer treatment. Current progress in the development of therapeutic approaches brings several promising inhibitors of HIF-1. Flavonoids, widely occurring in various plants, exert a broad spectrum of beneficial effects on human health, and are potentially powerful therapeutic tools against cancer. Recent evidences identified numerous natural flavonoids and their derivatives as inhibitors of HIF-1, associated with the regulation of critical glycolytic components in cancer cells, including pyruvate kinase M2(PKM2), lactate dehydrogenase (LDHA), glucose transporters (GLUTs), hexokinase II (HKII), phosphofructokinase-1 (PFK-1), and pyruvate dehydrogenase kinase (PDK). Here, we discuss the results of most recent studies evaluating the impact of flavonoids on HIF-1 accompanied by the regulation of critical enzymes contributing to the Warburg phenotype. Besides, flavonoid effects on glucose metabolism via regulation of HIF-1 activity represent a promising avenue in cancer-related research. At the same time, only more-in depth investigations can further elucidate the mechanistic and clinical connections between HIF-1 and cancer metabolism.
... Flavonoids' anti-cancer properties are partially mediated by inhibiting HIF-1. Consequently, it is essential to go into detail about how secondary plant metabolites affect HIF-1 function [81]. Secondary metabolites of plants having distinct phenolic structures are known as flavonoids. ...
Article
Full-text available
Hypoxia-inducible Factor 1 (HIF-1) is a transcriptional activator that intervenes in versatile reactions to hypoxia. Natural drugs, widely distributed in plants, comprise many metabolites, possessing their potential as anti-cancer agents. Studies have highlighted HIF-1 as a potential pharmacological target for cancer therapy. Secondary metabolites derived from natural products (plant-derived or marine organisms) with unique chemical structures have demonstrated HIF-1 inhibition. Ganetespib, topotecan, PX-478, aminoflavone, fluorine-19-fluoroded xyglucose, etc. have entered clinical phases of evaluation for different types of cancer. The present work describes role of HIF-1 in tumor progression, summarizes plant based inhibitors such as berberine, rohitukine, harringtonine, acronycin, nuciferine, evodiamine, matrine, etc. Some of the major categories with HIF-1 inhibitory potential are alkaloids, flavonoids, steroids, etc. The manuscript aims to benefit the discovery and development of anti-cancer therapeutics from natural compounds.
... This may be suggestive of the presence of non-cannabinoid active molecules, selectively extracted by the polar solvent ethanol and not by the non-polar solvent hexane, occurring in five different cannabis varieties (Moniek, Magda, Mati, Pilar and Theresa). Several flavonoids have been shown to actively inhibit HIF-1a transcription [33]. Flavonoids are more polar than cannabinoids and, therefore, more likely to be extracted by ethanol than hexane, thus providing a plausible explanation as to why ETH extracts from the same cannabis cultivar are active, but not their HEX counterparts. ...
Article
Full-text available
Minor cannabinoid and non-cannabinoid molecules have been proposed to significantly contribute to the pharmacological profile of cannabis extracts. Phytoplant Research has developed highly productive cannabis cultivars with defined chemotypes, as well as proprietary methods for the extraction and purification of cannabinoids. Here, we investigate the effect of solvent selection and decarboxylation on the composition and pharmacological activity of cannabis extracts. A library of forty cannabis extracts was generated from ten different cannabis cultivars registered by Phytoplant Research at the EU Community Plant Variety Office. Plant material was extracted using two different solvents, ethanol and hexane, and crude extracts were subsequently decarboxylated or not. Cannabinoid content in the resulting extracts was quantified, and biological activity was screened in vitro at three molecular targets involved in hypoxia and inflammation (NF-κB, HIF-1α and STAT3). Changes in transcriptional activation were strongly associated to solvent selection and decarboxylation. Two decarboxylated extracts prepared with hexane were the most potent at inhibiting NF-κB transcription, while HIF-1α activation was preferentially inhibited by ethanolic extracts, and decarboxylated extracts were generally more potent at inhibiting STAT3 induction. Our results indicate that solvent selection and proper decarboxylation represent key aspects of the standardized production of cannabis extracts with reproducible pharmacological activity.
... Elevated expression of HIF-1 promotes tumour-associated angiogenesis, proliferation and progression through the modulation of glycolytic cascades [156]. Current evidence suggests that flavonoids may play a considerable role in cancer treatment and have multiple potential targets in tumourigenesis, including HIF-1 [157]. Baicalein (BA; 5,6,7-trihydroxyflavone), a flavonoid isolated from Scutellaria baicalensis, exerts strong anticancer activity on various tumours [158][159][160]. ...
Article
Full-text available
The Warburg effect is characterised by increased glucose uptake and lactate secretion in cancer cells resulting from metabolic transformation in tumour tissue. The corresponding molecular pathways switch from oxidative phosphorylation to aerobic glycolysis, due to changes in glucose degradation mechanisms known as the ‘Warburg reprogramming’ of cancer cells. Key glycolytic enzymes, glucose transporters and transcription factors involved in the Warburg transformation are frequently dysregulated during carcinogenesis considered as promising diagnostic and prognostic markers as well as treatment targets. Flavonoids are molecules with pleiotropic activities. The metabolism-regulating anticancer effects of flavonoids are broadly demonstrated in preclinical studies. Flavonoids modulate key pathways involved in the Warburg phenotype including but not limited to PKM2, HK2, GLUT1 and HIF-1. The corresponding molecular mechanisms and clinical relevance of ‘anti-Warburg’ effects of flavonoids are discussed in this review article. The most prominent examples are provided for the potential application of targeted ‘anti-Warburg’ measures in cancer management. Individualised profiling and patient stratification are presented as powerful tools for implementing targeted ‘anti-Warburg’ measures in the context of predictive, preventive and personalised medicine.
Article
Background Hypoxia is a characteristic feature of many solid tumors. As an adaptive response to hypoxia, tumor cells activate hypoxia-inducible factor-1α (HIF-1α). Under hypoxic conditions, angiogenesis mediated by HIF-1α is involved in the growth and metastasis of tumor cells. During the angiogenic process, differentiated tip endothelial cells (ECs) characterized by high expression of DLL4 promote angiogenic germination through filopodia. Inhibitors of HIF-1α or DLL4 have been widely studied Purpose We tried to find inhibitors targeting both HIF-1α and DLL4 in tumor which have not yet been developed. Study design In this study, we examined a natural compound that inhibits sprouting angiogenesis and tumor growth by targeting both HIF-1α and DLL4 under hypoxic conditions. Methods After examining cell viability of 70 selected natural compounds, we assessed the effects of compounds on HIF-1α and DLL4 transcriptional activity using a dual-luciferase reporter assay. Western blot analysis, immunofluoresecnt assay and real-time qPCR were performed to identify expression of proteins, such as HIF-1α and DLL4, as well as HIF-1α target genes under hypoxic conditions. In vitro angiogenesis assay and in vivo allograft tumor experiment were performed to investigate inhibition of tumor growth through anti-angiogenic activity. Results Among these compounds, steppogenin, which is extracted from the root bark of Morus alba L, respectively inhibited the transcriptional activity of HIF-1α under hypoxic conditions in HEK293T cells and vascular endothelial growth factor (VEGF)-induced DLL4 expression in vascular ECs in a dose-dependent manner. In tumor cells and retinal pigment epithelial cells, steppogenin significantly suppressed HIF-1α protein levels under hypoxic conditions as well as VEGF-induced DLL4 expression in ECs. Furthermore, steppogenin suppressed hypoxia-induced vascular EC proliferation and migration as well as VEGF-induced sprouting of EC spheroids. Conclusion These results suggest that the natural compound steppogenin could potentially be used to treat angiogenic diseases, such as those involving solid tumors, because of its dual inhibition of HIF-1α and DLL4.
Article
Full-text available
Inhibition of matrix metalloproteinase-9 (MMP-9) can be an emerging strategy for a cancer cure as overexpression of MMP-9 is associated with several types of malignancies, including cancers related to lungs, breasts, and prostate. The present work was proposed to design and synthesize some novel cinnamic acid derivatives as potential anticancer agents. Novel cinnamic acid derivatives were designed using a structure-based drug design approach, and a series of 16 newer cinnamic acid analogs was prepared and evaluated for in vitro cytotoxicity (lung cancer cell line, A-549), followed by docking studies to explore binding interactions of designed compounds in the binding site of MMP-9. In the in vitro cytotoxicity assay, compound 5 was found to be most potent, with an IC50 value of 10.36 µM amongst the synthesized analogs. The designed molecules showed appreciable docking interactions and binding patterns with MMP-9 protein supporting the in vitro cytotoxicity outcomes. Compound 5 can be further explored as a possible lead molecule to develop potent and selective MMP-9 inhibitors as potential antineoplastic agents.
Article
Full-text available
Metabolic alterations are one of the hallmarks of cancer, which has recently gained great attention. Increased glucose absorption and lactate secretion in cancer cells are characterized by the Warburg effect, which is caused by the metabolic changes in the tumor tissue. Cancer cells switch from oxidative phosphorylation (OXPHOS) to aerobic glycolysis due to changes in glucose degradation mechanisms, a process known as “metabolic reprogramming”. As a result, proteins involved in mediating the altered metabolic pathways identified in cancer cells pose novel therapeutic targets. Hypoxic tumor microenvironment (HTM) is anticipated to trigger and promote metabolic alterations, oncogene activation, epithelial-mesenchymal transition, and drug resistance, all of which are hallmarks of aggressive cancer behaviour. Angiogenesis, erythropoiesis, glycolysis regulation, glucose transport, acidosis regulators have all been orchestrated through the activation and stability of a transcription factor termed hypoxia-inducible factor-1 (HIF-1), hence altering crucial Warburg effect activities. Therefore, targeting HIF-1 as a cancer therapy seems like an extremely rational approach as it is directly involved in the shift of cancer tissue. In this mini-review, we present a brief overview of the function of HIF-1 in hypoxic glycolysis with a particular focus on novel therapeutic strategies currently available.
Article
Breast cancer is the most diagnosed invasive cancer and the second leading cause of cancer in women. Standard medicinal protocol for treatment of this disease implies surgery and use of chemotherapeutic drugs. It is known that these drugs are associated with resistance, cancer relapse, and numerous side effects. Researchers give great importance to natural bioactive compounds because of their conformed health beneficial effects. Ocimum minimum L. is a well-known plant in folk medicine for treating various diseases and conditions, whit a large specter of pharmaceutical activities. The aim of this study is to evaluate anticancer properties and impact on fatty acid synthesis process under the influence of O. minimum L. flowers in triple-negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) and normal fetal fibroblasts (MRC-5). The impact of O. minimum L. on fatty acid synthesis process in breast cancer cells has been evaluated through cytotoxic activity by MTT assay, pro-apoptotic activity by AO/EB staining, concentrations of redox status parameters (O2.-, lipid peroxidation, GSH) by colorimetric methods. The expression of genes involved in fatty acid synthesis (ACLY, FASN, MAGL, HIF-1α) were determined by the qPCR method, while the protein expression (FASN) was analyzed by immunocytochemistry. Based on the obtained results, synergic action between bioactive compounds identified from O. minimum L. showed cytotoxic, proapoptotic, and prooxidant effects on breast cancer cell lines, as well as suppression of genes and protein (FASN) involved in fatty acid synthesis process. This plant showed significant pharmacological effects whit potential use as nutritional supplement in anticancer therapy.
Article
A series of flavonoid derivatives bearing trimethoxyphenyl and benzimidazole were designed, synthesized and evaluated as potential anti-tumor agents. Compound 5g showed remarkable inhibitory activity against SGC-7901, HGC-27 and MCF-7 (IC50 values of 20.5 ± 5.93, 3.3 ± 2.53 and 16.6 ± 0.75 μM, respectively), and had great selectivity to cancer cells. The results of Hoechst 33258 staining and Annexin V-FITC/PI dual staining confirmed that compound 5 g induced apoptosis of the HGC-27 cells in a concentration-dependent manner. After treatment with compound 5 g, the expression of HIF-1α, VEGF and PKM2 were down-regulated, accompanied by weakened the levels of lactate. Further research revealed that compound 5 g could obviously inhibit tubulin assembly. In summary, compound 5 g possessed a promising potential for further development into anti-tumor drug candidates.
Article
Full-text available
Tumor metastasis is still the leading cause of melanoma mortality. Luteolin, a natural flavonoid, is found in fruits, vegetables, and medicinal herbs. The pharmacological action and mechanism of luteolin on the metastasis of melanoma remain elusive. In this study, we investigated the effect of luteolin on A375 and B16‐F10 cell viability, migration, invasion, adhesion, and tube formation of human umbilical vein endothelial cells. Epithelial–mesenchymal transition (EMT) markers and pivotal molecules in HIF‐1α/VEGF signaling expression were analysed using western blot assays or quantitative real‐time polymerase chain reaction. Results showed that luteolin inhibits cellular proliferation in A375 and B16‐F10 melanoma cells in a time‐dependent and concentration‐dependent manner. Luteolin significantly inhibited the migratory, invasive, adhesive, and tube‐forming potential of highly metastatic A375 and B16‐F10 melanoma cells or human umbilical vein endothelial cells at sub‐IC50 concentrations, where no significant cytotoxicity was observed. Luteolin effectively suppressed EMT by increased E‐cadherin and decreased N‐cadherin and vimentin expression both in mRNA and protein levels. Further, luteolin exerted its anti‐metastasis activity through decreasing the p‐Akt, HIF‐1α, VEGF‐A, p‐VEGFR‐2, MMP‐2, and MMP‐9 proteins expression. Overall, our findings first time suggests that HIF‐1α/VEGF signaling‐mediated EMT and angiogenesis is critically involved in anti‐metastasis effect of luteolin as a potential therapeutic candidate for melanoma.
Article
Full-text available
Chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) is an inflammatory sinonasal disorder characterized by eosinophilic inflammation and T-helper 2 skewing. Eosinophil accumulation in sinonasal mucosa comprises a major feature of CRSwNP. The study aimed to investigate the effect of the flavone wogonin in nasal polyposis by assessing its ability to induce eosinophil apoptosis in vitro and attenuate eosinophilic CRSwNP in mice. Double immunofluorescence, immunohistochemistry, flow cytometry, and immunoblotting were performed to evaluate hypoxia-inducible factor (HIF)-1α, survivin, and apoptotic markers in the human eosinophilic EoL-1 cell line or sinonasal tissues from patients with CRS with or without NPs. In sinonasal specimens from patients with CRS, HIF-1α and survivin were up-regulated in eosinophils from patients with NPs compared with levels in patients without NPs. Under hypoxia, HIF-1α and survivin expression was up-regulated in EoL-1 cells. Wogonin down-regulated both HIF-1α and survivin in EoL-1 cells. In addition, overexpression of survivin protected EoL-1 cells against apoptosis in response to wogonin. Moreover, wogonin attenuated nasal polyp formation in a murine model. Our findings suggest that wogonin could induce caspase-3 activation by suppressing HIF-1α and survivin expression in EoL-1 cells. Further studies regarding novel therapeutic options for CRSwNP targeting eosinophil apoptosis are needed.
Article
Full-text available
Naringin is one of the most interesting phytopharmaceuticals that has been widely investigated for various biological actions. Yet, its low water solubility, limited permeability, and suboptimal bioavailability limited its use. Therefore, in this study, polymeric micelles of naringin based on pluronic F68 (PF68) were developed, fully characterized, and optimized. The optimized formula was investigated regarding in vitro release, storage stability, and in vitro cytotoxicity vs different cell lines. Also, cytoprotection against ethanol-induced ulcer in rats and antitumor activity against Ehrlich ascites carcinoma in mice were investigated. Nanoscopic and nearly spherical 1:50 micelles with the mean diameter of 74.80±6.56 nm and narrow size distribution were obtained. These micelles showed the highest entrapment efficiency (EE%; 96.14±2.29). The micelles exhibited prolonged release up to 48 vs 10 h for free naringin. The stability of micelles was confirmed by insignificant changes in drug entrapment, particle size, and retention (%) (91.99±3.24). At lower dose than free naringin, effective cytoprotection of 1:50 micelles against ethanol-induced ulcer in rat model has been indicated by significant reduction in mucosal damage, gastric level of malondialdehyde, gastric expression of tumor necrosis factor-alpha, caspase-3, nuclear factor kappa-light-chain-enhancer of activated B cells, and interleukin-6 with the elevation of gastric reduced glutathione and superoxide dismutase when compared with the positive control group. As well, these micelles provoked pronounced antitumor activity assessed by potentiated in vitro cytotoxicity particularly against colorectal carcinoma cells and tumor growth inhibition when compared with free naringin. In conclusion, 1:50 naringin–PF68 micelles can be represented as a potential stable nanodrug delivery system with prolonged release and enhanced antiulcer as well as antitumor activities.
Article
Full-text available
Background: Genistein is a natural isoflavone with many health benefits, including antitumour effects. Increased hypoxia-inducible factor 1 α (HIF-1α) levels and glycolysis in tumour cells are associated with an increased risk of mortality, cancer progression, and resistance to therapy. However, the effect of genistein on HIF-1α and glycolysis in hepatocellular carcinoma (HCC) is still unclear. Methods: Cell viability, apoptosis rate, lactate production, and glucose uptake were measured in HCC cell lines with genistein incubation. Lentivirus-expressed glucose transporter 1 (GLUT1) or/and hexokinase 2 (HK2) and siRNA of HIF-1α were used to test the direct target of genistein. Subcutaneous xenograft mouse models were used to measure in vivo efficacy of genistein and its combination with sorafenib. Results: Genistein inhibited aerobic glycolysis and induced mitochondrial apoptosis in HCC cells. Neither inhibitors nor overexpression of HK2 or GLUTs enhance or alleviate this effect. Although stabiliser of HIF-1α reversed the effect of genistein, genistein no longer has effects on HIF-1α siRNA knockdown HCC cells. In addition, genistein enhanced the antitumour effect of sorafenib in sorafenib-resistant HCC cells and HCC-bearing mice. Conclusions: Genistein sensitised aerobic glycolytic HCC cells to apoptosis by directly downregulating HIF-1α, therefore inactivating GLUT1 and HK2 to suppress aerobic glycolysis. The inhibitory effect of genistein on tumour cell growth and glycolysis may help identify effective treatments for HCC patients at advanced stages.British Journal of Cancer advance online publication 19 September 2017; doi:10.1038/bjc.2017.323 www.bjcancer.com.
Article
Many studies have focused on the identification of therapeutic targets for the treatment of certain types of cancer. Wogonin is a natural flavonoid compound that exhibits a potent anti-cancer effect. The underlying mechanism of wogonin may therefore reveal an effective way to identify novel therapeutic targets. In the current study, growth curves and MTT assays were performed to determine the effects of wogonin in human gastric cancer cells (SGC-7901) and human lung adenocarcinoma cells (A549), respectively. Changes in morphology were observed using hematoxylin and eosin (H&E) staining. The activities of key enzymes in the glycolysis and tricarboxylic acid cycle were measured using spectropho-tometry. Western blot analysis was performed to determine the expression levels of hypoxia inducible factor-1α (HIF-1α) and monocarboxylate transporter-4 (MCT-4). Wogonin inhibited cell proliferation in a time- and dose-dependent manner in SGC-7901 and A549 cells. H&E staining suggested that wogonin induced cell morphology changes. In SGC-7901 cells, lactate dehydrogenase (LDH) and succinate dehydrogenase (SDH) activities and adenosine triphosphate (ATP) generation were decreased significantly by wogonin treatment compared with the untreated control. In A549 cells, wogonin significantly reduced LDH activity, but exhibited no significant effects on kinase activities or ATP generation. Furthermore, wogonin significantly decreased HIF-1α and MCT-4 protein expression in SGC-7901 cells, but not in A549 cells. The results demonstrated that wogonin inhibited the energy metabolism, cell proliferation and angiogenesis in SGC-7901 and A549 cells by negatively regulating HIF-1α and MCT-4 expression. The differential regulatory roles of wogonin in metabolism-associated enzymes in human gastric cancer and lung adenocarcinoma cells indicated its various antitumor mechanisms. The different metabolic regulatory mechanisms exhibited by wogonin in different tumor tissues should therefore be considered for antitumor therapy.
Article
The imbalance between angiogenic inducers and inhibitors appears to be a critical factor in tumour pathogenesis. Angiogenesis serves a key role in the occurrence, invasion and metastasis of tumours. Macrophages are a major cellular component of human and rodent tumours, where they are usually termed tumour‑associated macrophages (TAMs). In malignant tumours, TAMs tend to resemble alternatively activated macrophages (M2‑like), promote TA angiogenesis, strengthen tumour migration and invasive abilities, and simultaneously inhibit antitumor immune responses. In our previous study, luteolin, commonly found in a wide variety of plants, had a strong antitumor effect under normoxia; however, it is unknown whether luteolin serves a similar role under hypoxia. In the present study, cobalt chloride (CoCl2) was used to simulate hypoxia. Hypoxia‑inducible factor‑1α (HIF‑1α), which is difficult to detect under normoxic conditions, was significantly increased. Additionally, vascular endothelial growth factor (VEGF) was also significantly increased in response to CoCl2 treatment. Subsequently, luteolin was applied with CoCl2 to examine the effects of luteolin. Luteolin decreased the expression of VEGF and matrix metalloproteinase‑9, which promote angiogenesis. In addition, luteolin also suppressed the activation of HIF‑1 and phosphorylated‑signal transducer and activator of transcription 3 (STAT3) signalling, particularly within the M2‑like TAMs. The results of the present study provide novel evidence that luteolin, under hypoxic conditions, has a strong anticancer effect via the HIF‑1α and STAT3 signalling pathways.
Article
A series of twenty-six carbamates derived from aminocombretastatin A-4 (AmCA-4) were synthesized and evaluated for their capacity to affect cell proliferation, tubulin polymerization, mitotic cell arrest, microtubule network organization, apoptosis and endothelial tubular structures in vitro. The anti-proliferative activity of the synthetic carbamates was measured on several human tumor cell lines (i.e. HT-29, MCF-7, HeLa, A-549, MDA-MB-231, HL-60) as well as on the endothelial cell line HMEC-1 and the non-tumor cell line HEK-293. The compounds showed anti-proliferative activity in the nanomolar range thereby exceeding by far the activity of combretastatin A-4 (CA-4) and, in some cases, the activity of AmCA-4. The most active compounds proved to be the carbamates bearing chloro, bromo or methoxy groups in the meta position of the phenyl ring. Moreover, all carbamates inhibited in vitro tubulin polymerization, in a similar manner to that of CA-4 and AmCA-4 by interacting with the colchicine binding site in tubulin. The synthetic carbamates proved as active as AmCA-4 in causing mitotic arrest, as assessed in A549 human lung cancer cells, and disruption of the microtubule cellular network. Some selected carbamates induced apoptosis at concentrations as low as 10 nM, being more active than AmCA-4. Finally, these selected carbamates displayed a vascular disrupting activity on endothelial cells in a dose-dependent manner. In conclusion, our data indicate that carbamates derived from aminocombretastatin A-4 represent interesting lead compounds for the design of vascular disrupting agents.
Article
A series of 3’,4’,5’-trimethoxy flavonoids with benzimidazole linked by different chain alkanes have been designed and synthesized. The potential activity of these compounds as anti-tumor agents was evaluated by cytotoxicity assay in MGC-803 (Human gastric cancer cell line), MCF-7 (Human breast cancer cell line), HepG-2 (Human hepatoma cell line) and MFC (Mouse gastric cancer cell line) tumor cell lines. Among them, compound 15 7-(3-(2-chloro-1H-benzo[d]imidazol-1-yl)propoxy)-2-(3,4,5-trimethoxyphenyl)-4H-chromen-4-one displayed the most potent antiproliferative activity with IC50 values of 20.47 ± 2.07, 43.42 ± 3.56, 35.45 ± 2.03 μM and 23.47 ± 3.59 μM, respectively. The flow cytometry (FCM) results showed that the compound 15 caused the cell cycle to be arrested in G1 phase and induced apoptosis of MFC cells in a dose dependent manner. In addition, compound 15 exhibited a significant inhibitory effect on tumor growth in vivo. All the results outlined the great potential of compound 15 for further exploitation as anti-tumor agent.
Article
Hand Foot Mouth Disease (HFMD), resulting from human enterovirus A71 (HEVA71) infection can cause severe neurological complications leading to fatality in young children. Currently, there is no approved antiviral for therapeutic treatment against HEVA71 infection. In this study, a 500-compound flavonoid library was screened to identify potential inhibitors of HEVA71 using high-throughput immunofluorescence-based phenotypic screening method. Two lead flavonoid compounds, ST077124 and ST024734 at the non-cytotoxic concentration of 50 μM were found to be effective antivirals that inhibited replication of HEVA71, reducing infectious viral titers by 3.5 log10 PFU/ml and 2.5 log10 PFU/ml respectively. Our study revealed that ST077124 is a specific antiviral compound that inhibits human enteroviruses while ST024734 exhibited antiviral activity against human enteroviruses as well as dengue virus type-2. We also identified that both compounds affected the viral RNA transcription and translation machinery of HEVA71 but did not interfere with the viral internal ribosomal entry site (IRES) activity. Hence, our findings strongly suggest that ST077124 and ST024734 are effective antiviral compounds of minimal cytotoxicity and could serve as promising therapeutic agents against HEVA71 infection.
Article
Three new flavonoid glycosides, 2(S)-5-hydroxy-6,8-dimethoxyflavonone-7-O-β-D-glucopyranosyl-(1→6)-O-β-D-glucopyranoside (1), 5-hydroxy-3,8-dimethoxyflavone-7-O-β-D-glucopyranosyl-(1→6)-O-β-D-glucopyranoside (2) and 3,7-dihydroxy-8-methoxyflavone-6-O-β-D-glucopyranosyl-(1→6)-O-β-D-glucopyranoside (3), together with five known flavonoids (4–8) were isolated from the roots of Smilax glabra Roxb. Their structures were elucidated on the basis of chemical and spectral evidence, as well as by comparison with literature data. Three new flavonoids were subjected to evaluate anti-inflammatory activity. Compounds 1–3 inhibited the NF κB induction by 32.2, 55.8 and 61.7%, respectively.