ArticlePDF Available

Abstract and Figures

Purpose of Review This short review is aimed at providing an updated and comprehensive report on tannic acid biological activities and molecular mechanisms of action most important for cancer prevention and adjuvant therapy. Recent Findings Tannic acid (TA), a mixture of digallic acid esters of glucose, is a common ingredient of many foods. The early studies of its anti-mutagenic and anti-tumorigenic activity were mostly demonstrated in the mouse skin model. This activity has been explained by its ability to inhibit carcinogens activation, as well as antioxidant and anti-inflammatory properties. Recently, the cell cycle arrest, apoptosis induction, reduced rate of proliferation, and cell migration and adhesion of several cancer cell lines as a result of TA treatment were described. The underlining mechanisms include modulation of signaling pathways such as EGFR/Jak2/STATs, or inhibition of PKM2 glycolytic enzyme. Moreover, epithelial-to-mesenchymal transition prevention and decrease of cancer stem cells formation by TA were also reported. Besides, TA was found to be potent chemosensitizer overcoming multidrug resistance. Eventually, its specific physicochemical features were found useful for generation of drug-loaded nanoparticles. Summary TA was shown to be a very versatile molecule with possible application not only in cancer prophylaxis, as was initially thought, but also in adjuvant cancer therapy. The latter may refer to chemosensitization and its application as a part of drug delivery systems. More studies are required to better explore this subject. In addition, the effect of TA on normal cells and its bioavailability have to better characterized.
Content may be subject to copyright.
Tannic Acid: Specific Form of Tannins in Cancer Chemoprevention
and Therapy-Old and New Applications
Wanda Baer-Dubowska
1
&Hanna Szaefer
1
&Aleksandra Majchrzak-Celińska
1
&Violetta Krajka-Kuźniak
1
Published online: 27 March 2020
Abstract
Purpose of Review This short review is aimed at providing an updated and comprehensive report on tannic acid biological
activities and molecular mechanisms of action most important for cancer prevention and adjuvant therapy.
Recent Findings Tannic acid (TA), a mixture of digallic acid esters of glucose, is a common ingredient of many foods. The early
studies of its anti-mutagenic and anti-tumorigenic activity were mostly demonstrated in the mouse skin model. This activity has been
explained by its ability to inhibit carcinogens activation, as well as antioxidant and anti-inflammatory properties. Recently, the cell cycle
arrest, apoptosis induction, reduced rate of proliferation, and cell migration and adhesion of several cancer cell lines as a result of TA
treatment were described. The underlining mechanisms include modulation of signaling pathways such as EGFR/Jak2/STATs, or
inhibition of PKM2 glycolytic enzyme. Moreover, epithelial-to-mesenchymal transition prevention and decrease of cancer stem cells
formation by TA were also reported. Besides, TAwas found to be potent chemosensitizer overcoming multidrug resistance. Eventually,
its specific physicochemical features were found useful for generation of drug-loaded nanoparticles.
Summary TA was shown to be a very versatile molecule with possible application not only in cancer prophylaxis, as was
initially thought, but also in adjuvant cancer therapy. The latter may refer to chemosensitization and its application as a part
of drug delivery systems. More studies are required to better explore this subject. In addition, the effect of TA on normal
cells and its bioavailability have to better characterized.
Keywords Tann ic acid .Mouse skin model .Cancer cells .Chemosensitization .Nanomedicine
Introduction
Carcinogenesis is the process of transformation of a normal
cell into a neoplastic one. This transition involves several
steps starting with initiation, and followed by promotion and
progression. Genetic and epigenetic changes caused by exog-
enous agents or endogenous factors are driving these stages,
leading to the accumulation of mutations and epimutations in
genes responsible for cellular homeostasis. Thus, cancer de-
velopment involves gene-environment interactions.
Moreover, oxidative stress and inflammation play important
roles in carcinogenesis. Along with the epigenetic
modifications, they cause aberrant expression of a variety of
genes, both within the transforming cell population and the
cells within the surrounding lesion [1].
Since carcinogenesis is a complex and long multistep
process, intervention on its most early stages is considered
as the most logical and promising approach of combating
cancer. Chemoprevention is defined as the use of synthet-
ic or natural substances to reverse, suppress, or prevent
carcinogenic progression [2].
All of the above mentioned processes are targets for chemo-
preventive agents, and numerous reports of various bioactive
foods and their extracted compounds, including tannins, have
been shown to affect these hallmarks of carcinogenesis. Tannic
acid (TA) is a specific tannin that formally contains 10 galloyl
(3,4,5-trihydroxyphenyl) units surrounding a glucose center.
However, commercially available and often naturally occurring
TA consists of molecules with 212 galloyl moieties. TA con-
tains no carboxyl groups, but is weakly acidic because of the
multiplicity of phenolic hydroxyls. The hydroxyls also cause it
to be extremely soluble in water [https://www.acs.org/content/
This article is part of the Topical Collection on Redox Modulators
*Wan da Bae r-Du b o ws k a
baerw@ump.edu.pl
1
Department of Pharmaceutical Biochemistry, Poznan University of
Medical Sciences, Święcicki 4 Str, Poznań,Poland
Current Pharmacology Reports (2020) 6:2837
https://doi.org/10.1007/s40495-020-00211-y
REDOX MODULATORS (C JACOB, SECTION EDITOR)
#The Author(s) 2020
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
acs/en/molecule-of-the-week/archive/t/tannic-acid.html]. As
the others tannins, TA is found in a wide range of plants,
including fruits, green and black teas, nuts, and grains.
However, TA major limitation in biological systems might be
its relatively poor bioavailability. The available data indicate
that following oral administration, ~ 60% of tannic acid
remains unchanged, but some are hydrolyzed to gallic acid by
tannase in the intestine and are further metabolized to 4-O-
methylgallic acid, pyrogallol, and resorcinol [3].In vitro bio-
availability of tannic acid was evaluated in ligated rat small
intestine segments showing 50% uptake, but not complete
transfer through the gut wall [4]. One of the very first studies
about TA date back to 1989, when TA was found to inhibit skin,
lung, and forestomach tumors induced by chemical carcino-
gens [5••]. This activity was related to reduced carcinogens
activation resulting from inhibition of specific forms of cyto-
chrome P450, electrophile trapping, modulation of arachidonic
acid metabolism [6,7], and ultimately inhibition of DNA-
adducts formation [8,9,10]. Similarly, to other polyphenols,
TA possesses antioxidant activity [11]. However, it can act also
as pro-oxidant resulting in oxidative DNA damage [12].
In the recent years, growing number of reports describe
new mechanisms of TA activity and possible application not
only in primary chemoprevention, but also in sensitization to
conventional drugs used in anticancer therapy. Moreover, its
specific physicochemical features showed up to be useful for
nanomedicine purposes, including modern drug delivery sys-
tems (Fig. 1). This short review summarizes the current
knowledge about TA chemopreventive activity and its possi-
ble application in cancer prophylaxis and adjuvant cancer
therapy.
Early Studies: Tannic Acid Affects
the Processes Involved in Initiation
and Promotion of Carcinogenesis in Mouse
Skin Carcinogenesis Model
The mouse skin model of multistage chemical carcinogenesis
represents one of the best-established in vivo models for the
study of the sequential and stepwise development of tumors.
In addition, this model can be used to evaluate both novel skin
cancer prevention strategies and the impact of genetic back-
ground and genetic manipulation on tumor initiation, promo-
tion, and progression [13]. Therefore, the earliest data on the
anticarcinogenic activity of TA were demonstrated in this
model (Fig. 2).
In this regard, Muhtar et al. [14••] showed exceptional ac-
tivity of TA, among naturally occurring plant phenols, in the
protection against polycyclic aromatic hydrocarbons (PAH)
7,12-dimethylbenz[a]anthracene (DMBA), benzo[a]pyrene
(B[a]P), 3-methylcholanthrene (3-MC), and direct carcinogen
N-methyl-N-nitrosourea-induced skin tumorigenesis. The ini-
tiation of carcinogenesis by indirect carcinogens like PAH
requires metabolic activation to ultimate carcinogenic form,
which covalently binds to DNA. Cytochromes P450, mainly
Inhibition of cancer promotion
and progression
Induction of cell cycle arrrest
and apoptosis of cancer cells
Inhibition of cancer cells
migration, invasion and
colony formation
Modulation of phase I and II drug
metabolizin
g
enz
y
mes
Inhibition of cancer cell
proliferation and
angiogenesis
Inhibition of epithelial-to-
mesenchymal transition
Chemosenstization and impact
on multidrug resistance
An element of anti-cancer drug
delivery systems/carrier of anti-
cancer drugs
Fig. 1 The overview of mechanisms exerted by tannic acid important for cancer chemoprevention and/or therapy
Curr Pharmacol Rep (2020) 6:2837 29
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
CYP1A1, CYP1A2, CYP1B1, and CYP2E1, are involved in
this process. Several studies including ours have shown the
ability of TA to inhibit P450 monooxygenases or its specific
isoforms [1517]. Inhibitions of these enzymes led to the re-
duced carcinogen-DNA adduct formation in mouse epidermis
[8,18]. Interestingly, analysis of DMBA-DNA adducts
formed in vitro in the presence of 3-MC induced microsomes
and in mouse epidermis showed almost complete inhibition of
DMBA dAdo adducts formation [9,10]. This was an im-
portant observation since the adenine adducts induced by ul-
timate DMBA metabolites (bay-region diol-epoxides) lead to
mutation at the codon 61 of c-H-Ras and consequently initiate
tumorigenesis in the mouse skin [19].
Moreover, modulation of phase II enzymes involved in
ultimate carcinogens deactivation by TA was also observed
both in animal models and in cell cultures in vitro.In animal
models, this effect was to some extent tissue specific [16].
In tissue such as mouse epidermis, the role of modulation of
enzymes involved in carcinogens activation and deactivation in
the formation of DNA-adducts by TA depended on the type of
carcinogen. While the reduction of B[a]P-DNA seems to result
from decreased B[a]P activation, in case of DMBA-DNA ad-
ducts, the scavenging or masking of the binding sites to be oc-
cupied by DMBA reactive metabolites is more probable [15].
Following the initiation stage, the population of mutated
cells is promoted to clonally expand during the second stage,
referred to as promotion.Promoting agents, which are both
structurally and mechanistically diverse, stimulate cell signal-
ing, increase production of growth factors, and generate oxi-
dative stress and tissue inflammation [13]. Ours and the other
research groups demonstrated that TA in the mouse epidermis
stimulated by 12-O-tetradecanoyphorbol-13-acetate (TPA)
inhibited the activation of NF-κB transcription factor and sub-
sequently expression and activity of inducible isoform of ni-
tric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2)
enzymes the key players of inflammation process [20]. As
was mentioned above, TA through the reduction of DMBA,
dAdo adducts may protect against Ras activation, which is
required for chemical carcinogen-induced skin carcinogenesis
in mice. Tumor promoter such as TPA expends the population
of Ras initiated cells [13]. Thus, the compounds which affect
the initiation of carcinogenesis in the mouse epidermis by
reducing DNA adducts in critical genes like Ras family may
inhibit the activation of NF-κB. The results of our studies
demonstrated that TA may act in this way, i.e., by decreasing
carcinogen-dAdo adducts formation, TA may inhibit NF-κB
activation [20]. Moreover, TA affected also the activation of
epidermal growth factor receptor (EGFR), activator protein-1
(AP-1) transcription factor, and signal transducers and activa-
tors of transcription (STATs) signaling pathways [21].
Moreover, in the same model, TA down-regulated the expres-
sion and activity of protein kinase C (PKC) which is thought
to be a major intracellular receptor for the mouse skin tumor
promoter TPA [22].
More recently, anti-promotional activity of TA in the
mouse skin through the reduction of oxidative stress as well
as COX-2, iNOS, PCNA (proliferating cell nuclear antigen)
protein, and proinflammatory cytokine such as IL-6 release
was confirmed [23]. Anti-inflammatory activity of TA was
also shown in the context of house dust miteinduced atopic
Chemical, physical, biological
carcinogen
INITIATION PROMOTION PROGRESSION
Normal cells
Genetically and/or
epigenetically altered cell Selective clonal expansion Tum or c el ls
Fig. 2 Tannic acid can affect cancer initiation, promotion, and progression
30 Curr Pharmacol Rep (2020) 6:2837
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
dermatitis. As the underlying mechanism of this activity, the
induction of peroxisome proliferator-activated receptor
PPARγprotein was suggested [24].
Chemopreventive activity of very low dose dietary TA ad-
ministration in hepatoma bearing C3H male mice was also
demonstrated, but the studies on the mechanism of this activ-
ity in this model were not continued [25]. However, signifi-
cant hepatoprotective effects against acetaminophen-induced
hepatotoxicity were recently described [26]. The authors sug-
gested that hepatoprotective mechanisms of TA may be relat-
ed to antioxidation, anti-inflammation, and anti-apoptosis,
thus the same mechanisms which might be responsible for
TA chemopreventive activity observed in early study of hep-
atoma bearing mouse model.
Tannic Acid Induces Cell Cycle Arrest,
Apoptosis, and Limits Proliferation of Various
Cancer Cells
Promising data on the anticarcinogenic effect of TA in animal
models, particularly in two-stage mouse skin model, stimulat-
ed the studies on the effect of TA on vital cellular processes
such as apoptosis and proliferation in cancer cells of different
origin. The observed effects of TA treatment often were cell-
type dependent. The examples of TA influence on cancer cell
lines of different origin are presented below.
Breast Cancer Cells
Booth et al. [32] performed a series of experiments in which
ER-positive breast cancer cells, MCF7, and triple negative
(ER
,PR
,HER2
) MDA-MB-231 along with non-
tumorigenic MCF10A were exposed to TA-cross-linked col-
lagen type I beads. Growing cells remodeled collagen and
released TA into surrounding medium leading to caspase-
mediated apoptosis. MCF7 cells were more sensitive to the
pro-apoptotic effect of TA than MDA-MB-231. TA also in-
duced apoptosis through the same mechanism in HER-2 pos-
itive cell line BT474 [27,28,29].
Moreover, the same group successfully loaded adipocytes
onto collagen type I beads with TA cross-linked. As adipo-
cytes attached and grew on the TA cross-linked collagen
beads, they remodeled the collagen, releasing TA, which then
interacted with HER2 breast cancer cells, leading to its apo-
ptosis. Viability assays also revealed the higher toxicity of TA
to HER21 breast cancer cells as compared to normal human
breast epithelial cells and adipocytes.
In contrast to the results of the above mentioned studies, no
differences in the sensitivity to TA between MCF7 and MDA-
MB-231 breast cancer cells were observed in the investigation
of Nie et al. [30]. One reason of this discrepancy might be the
difference in TA concentrations applied in both studies which
make the two approaches difficult to compare. This group also
demonstrated that TA inhibits fatty acid synthase (FAS) activ-
ity. This key enzyme of fatty acids synthesis is overexpressed
in human breast cancer cells. The authors suggested that inhi-
bition of FAS may be one of the possible ways to induce
apoptosis in these cells. Interestingly, TA showed higher
cytotoxicity toward breast cancer cells than to FAS
overexpressed 3 T3-L1 adipocytes. Thus, it is possible that in
appropriate concentration, TA may induce apoptosis in cancer
cells, but not in the surrounding adipocytes.
Moreover, TA has been reported to have high tyrosine kinase
inhibition capacity. In this regard, strong inhibition of the tyrosine
kinase activity of epidermal growth factor receptor (EGFR) and a
weak inhibition of the P60 and insulin receptor tyrosine kinase
were observed as a result of the treatment of human hepatoma
HepG2 cells with TA. The molecular modeling study suggested
that TA could be docked into the ATP-binding pockets of either
EGFR or the insulin receptor [31]. EGFR-mediated phosphory-
lation of signal transducers and activators of transcription
(STATs) leads to their activation. STATs can be activated also
in EGFR-independent manner, involving phosphorylation by
Janus kinases (JAKs). It was found that TA modulates the
EGFR/Jak2/STAT3 pathway, inducing mitochondrial apoptosis
in breast cancer cells lines: MCF-7, T47D, SK-BR 3, and MDA-
MB-231. Moreover, both the enhancement of STAT1 ser727
phosphorylation and the inhibition of STAT1 tyr701 phosphory-
lation were discovered as the key factors leading to G1 arrest
upon TA treatment [32].
Prostate Cancer Cells
The effect of TA on proliferation, metastasis, and invasion was
investigated in prostate cancer PC-3 and LNCaP cell lines,
representing high and low metastatic potential, respectively.
Treatment with TA significantly inhibited migration, invasion
into matrigel, and ability to form colonies by prostate cancer
cells. Modulation of the expression of cytochromes CYP17A1,
CYP3A4, CYP2B6, and phase II enzymes NQO1, GSTM1, and
GSTP1 was also observed in these cells [33].
Head and Neck Cancer Cells
The effect of TA on hypopharyngeal FaDu cancer cells and
YD-38 gingival squamous cell carcinoma was investigated by
Ta et al. [34] and Darvin et al. [35], respectively. In FaDu cells
treatment with low dose (25 μM) of TA led to cell cycle arrest
in G2/M phase. As the dose of TA was increased, apoptosis
was induced with the increase of cell population at sub-G1
phase. Both intrinsic and extrinsic cell death pathways were
affected which was demonstrated by the evaluation of the
expressionof various cyclins and poly (ADP-ribose) polymer-
ase (PARP) as well as phosphorylation of kinases of ERK,
AKT and PKB [34]. In gingival squamous cell carcinoma
Curr Pharmacol Rep (2020) 6:2837 31
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
YD-38 cells, TA inhibited Jak2/STAT3 pathway by
preventing the expression as well as phosphorylation of its
elements. It was also proved that TA exerted an intense acti-
vation of p21
Waf 1 /C i p 1
,p27
Kip1
,and p53 genes confirming its
role in G1 phase inhibition [35].
Liver Cancer Cells
The effect of TA on liver cancer cells was investigated in
human hepatoma cell line HepG2. The results of our study
showed activation of Nrf2/ARE signaling pathway as a result
of the treatment with 2 and 10 μM of TA. Subsequent induc-
tion of phase II enzymes, particularly GST, as well as antiox-
idant enzymes, was observed [36]. In contrast, recent study of
Mhlanga et al. [37] showed increased levels of reactive oxy-
gen species (ROS) and reactive nitrogen species [RNS] and
down-regulation of antioxidant enzymes expression as a result
of the treatment with IC
50
and IC
25
concentrations of TA, i.e.,
29.4 and 14.7 μM, thus significantly higher than that applied
in our study. The results of both studies confirm earlier sug-
gestions [12] that TA may act as antioxidant or pro-oxidant
depending on concentration. On the other hand, GSTTwhich
was induced by TA in our study acts as a scavenger of elec-
trophiles, such as epoxides. However, it may also metaboli-
cally activate halogenated compounds, thus producing a vari-
ety of intermediates that can potentially damage DNA and
cells [36]. Therefore, its induction in cancer cells might be
considered as ambiguous. Moreover, in another study, TA
isolated from Caesalpinia coriaria induced G2/M phase cell
cycle arrest and triggered cell death by microtubule stabiliza-
tion in human hepatoma Hep3B cells [38].
Colon Cancer Cells
Interesting mechanism of inhibition of colorectal cancer
cells (CRC) proliferation was proposed by Yang et al.
[39••]. This group demonstrated that TA inhibits pyru-
vate kinase PKM2 activity and subsequently suppresses
cell proliferation. They proposed, as an underlying
mechanism of enzyme inhibition, binding of TA to ly-
sine residue 433, which triggers the dissociation of
PKM2 tetramers and blocks the activity of PKM2, not
affecting PKM1 isozyme. The non-allosteric isoform
PKM1 is constitutively active, and expressed in termi-
nally differentiated tissues. By contrast, PKM2 is
expressed in tissues with anabolic functions, and is sub-
ject to complex allosteric regulation. In the majority of
cancer cells, the expression of PKM2 is increased,
which suggests that PKM2 may be an attractive target
for cancer therapy [40]. Therefore, TA might be consid-
ered as one of the molecules acting as PKM2 inhibitor.
Glioma Cancer Cells
The effect of TA was studied in rat C6 and human T98G
glioma cell lines and verified in glioblastoma rat model. In
our study, no significant differences in cell cycle distribution
was observed in C6 glioma cells, but in T98G increased num-
ber of cells in S phase was found after incubation with TA at
the concentrations lower and higher than IC
50
. In both cell
lines, TA significantly increased the number of dead cells.
Induction of apoptosis resulted mostly from increased level
of caspase-3 [41]. In contrast, in the report of Bona et al.
[42], the increased sub-G1 population of C6 cells, as a result
of the treatment with TA in comparable concentrations, was
described, along with the induction of apoptosis and necrosis.
Moreover, TA reduced the formation and size of colonies, as
well as cell migration and adhesion. Importantly, anti-glioma
effect was also observed in vivo. TA decreased tumor volume
and increased the area of intra-tumoral necrosis and infiltra-
tion of lymphocytes without damage of the surrounding tis-
sue. These data suggest that TA may potentially support the
therapy of these highly aggressive tumors.
As the examples above show, TA may inhibit proliferation
and enhance, via different mechanisms, cell death of various
cancer cells. However, so far, the similar data on normal cells
or immortalized normal cells are scarce.
The Cellular Effect of Tannic Acid
Beyond Cancer Cell Death and Proliferation
Epithelial-to-mesenchymal transition (EMT) is a dynamic,
self-controlled, physiological process by which epithelial cells
lose their junctional architecture and apical-basal polarity, de-
tach from each other, and convert into a mesenchymal pheno-
type [43,44]. EMT is crucial during embryogenesis, wound
healing, and tissue regeneration; however, in noncontrolled
conditions, it may lead to fibrosis, angiogenesis, and tumor
progression with metastatic expansion [45]. It has recently
been reported that TA treatment prevents TGFβ-induced
EMT in breast cancer cells as well as in lung epithelial cells
[46••,47]. The direct interaction between TA and TGF-β1
was observed, attenuating the TGF-βsignaling [47]. In lung
epithelial cells, TA also decreased the expression of N-
cadherin, type-1-collagen, fibronectin, and vimentin.
Additionally, phosphorylation of Smad2 and 3, Akt,
ERK1/2, JNK1/2, and p38 also decreased after the treatment
with TA [47,48]. Moreover, in breast cancer cell line model,
TA not only led to EMT inhibition, but also prevented the
TGFβ-induced increase in cancer stem cells (CSC) formation.
Stemness-marker expression, including ALDH1 activity and
the CD44
high
/CD24
low
ratio was also decreased after the treat-
ment with only 10 μMTA[46••]. Moreover, TA attenuated
NF-κB signaling which is regarded as one of the most
32 Curr Pharmacol Rep (2020) 6:2837
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
important mechanisms leading to the alleviation of CSC for-
mation and EMT [46••]. In addition, blocking of NF-κBsig-
naling by TA in bone marrowderived macrophages inhibited
NLRP3 inflammasome activation [49]. Recent data suggests
that excessive NLRP3 inflammasome activation characterizes
different cancer cells including head and neck squamous cell
carcinoma and colorectal cancer cells. In U87 and GL261
xenograft mouse GBM model, NLRP3 inflammasome was
involved in the resistance to radiotherapy [50]. Thus TA, by
inhibiting NLRP3, may reduce cancer cell survival or improve
the outcome of therapy.
The antiangiogenic properties of TA along with migration
inhibition of MDA-MB-231 breast cancer cells were tested in
the early study of Chen et al. [51]. TA inhibited cell migration
induced by chemokine CXCL12. The effect of TA on the
angiogenic consequences of CXCL12/CXCR4 interaction
was studied using an in vitro assay of capillary tube out-
growth. Treatment with 0.5 g/ml of TA completely inhibited
tube formation induced by CXCL12, but not by basic fibro-
blast growth factor (bFGF) or endothelial cell growth supple-
ment (ECGS) in bovine aortic endothelial cells (BAEC), sug-
gesting that TA selectively antagonized the angiogenic activ-
ity of CXCL12. Chemokines, such as CXCL12 and their re-
ceptors, are now increasingly recognized as critical communi-
cation bridges between tumor cells and stromal cells to create
a permissive microenvironment for tumor growth and metas-
tasis [52]. Thus, both observations deserve further studies, but
so far were not continued.
Attempts to Apply Tannic Acid for Cancer
Cells Sensitization and Overcoming Multidrug
Resistance
The treatment of cancer with chemotherapeutic agents has two
major problems, time-dependent development of tumor resis-
tance to therapy and nonspecific toxicity toward normal cells.
A growing amount of studies indicate that plant polyphenols,
including TA, are able to sensitize drug-resistant tumors to
chemotherapy via various mechanisms, as well as to be pro-
tective from therapy-associated toxicities [53]. One of the
intracellular targets of polyphenols may be the proteasome.
This proteolytic enzyme complex, responsible for intracellular
protein degradation has been shown to play an important role
in tumor growth and the development of drug resistance.
Thus, inhibition of proteasome is considered as one of the
mechanisms to overcome drug resistance and chemosensitize
cancer cells to chemotherapy [54]. The ability of TA to inhibit
the proteasome activity was tested and verified in purified 20S
proteasome and cellular 26S proteasome in different cell types
as well as in tumor-bearing mouse models. Inhibition of the
proteasome function by TA resulted in increased p27 and Bax
expression, and impaired cell cycle progression [55].
However, no combination with anticancer drugs was tested
in this system.
Poly (ADP-ribose) glycohydrolase (PARG) is the main nucle-
ar enzyme, which digests poly (ADP-ribose) into ADP-ribose.
PARG inhibitors could also be considered as chemotherapeutic
agents, because of its involvement in DNA repair [56]. TA was
found to be PARG inhibitor, and through this mechanism, the
sensitivity of ovarian carcinoma cells to cisplatin was increased.
Combined treatment with TA and cisplatin induced apoptosis
and increased DNA damage in the human ovarian carcinomas
cisplatin-resistant SKOV-3 CDDP/R cell line and cisplatin-
sensitive SKOV-3 cell line [57].
The main mechanism leading to the multidrug resistance
(MDR) after the treatment with anticancer drugs is the over-
expression of ABC transporters in cancer cells. Among ABC
transporters, the major target of potential chemosensitizers is
P-glycoprotein (P-gp; MDR-1) [58]. P-gp is expressed in var-
ious cancers and mediates MDR by actively transporting a
wide range of anticancer drugs, including doxorubicin [59].
Early report of Naus et al. [60] described interactions between
TA and chemotherapeutic drugs in malignant human
cholangiocytes. TA inhibited cellular efflux pathways, as de-
termined by calcein retention assays by decreasing the expres-
sion of P-gp, MRP1, and MRP2 membrane efflux pumps.
Modulation of drug efflux pathways resulted in synergistic
effect to mitomycin C and 5-fluorouracil used in cholangio-
carcinoma therapy.
In more recent study, the P-gp overexpressing human colon
cancer cell line Caco-2 and human T-lymphoblastic leukemia
cell line CEM/ADR 5000 were used to evaluate the effect of
TA combination with doxorubicin (DOX). This combination
synergistically sensitized both types of cells to the treatment.
Decreased activity of P-gp as a result of the treatment with TA
indicated that the inhibition of this protein is responsible for
chemosensitization effect [58]. DOX is a highly effective drug,
but its toxicity to normal cells, particularly, cardiomyocytes,
restricts its therapeutic application. Thus, the use of phyto-
chemicals as a protective tactic to reverse DOX-induced
cardiotoxicity was the subject of several studies [61]. Zhang
et al. [62] reported that pretreatment of rats with TA weakened
DOX-induced cardiotoxicity by inhibiting oxidative stress, in-
flammation, and apoptotic damage. The possible protection of
normal human oral keratinocytes against DOX-induced cyto-
toxicity without mitigating its cytotoxic potential against oral
cancer cells was investigated in normal human oral
keratinocytes and HSC-2 human oral squamous cell carcinoma
cells. TA at the concentration above 50 μM mitigated the
DOX-induced keratinocyte toxicity without weakening DOX
effect in SSC cells. In contrast, combination of TA at the
concentration of 50 μM and 100 μMwithDOXalmost
completely inhibited their survival [63]. The above data indi-
cate that TA may be considered as a potential adjuvant in
cancer chemotherapy.
Curr Pharmacol Rep (2020) 6:2837 33
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Tannic Acid as a Carrier of Anticancer Drugs
Nanomedical approaches to drug delivery aim at developing
nanoscale particles or molecules to improve drug bioavailability
both at specific places in the body and over a period of time.
Different approaches have been applied to effectively load target
drugs to enhance delivery resulting in increasing number of
nanoparticles being carriers of anticancer drugs [64••]. Recently
TA attracted attention as a useful excipient for generation of
drug-loaded nanoparticles. TA, being an additive molecule, can
improve solubilization of various hydrophobic drugs intended
for parenteral applications. TA possesses additive feature due to
its low viscosity, good water solubility, and biocompatibility. TA
can bind to drug molecules via hydrophobic interactions, which
in turn form a self-assembled cross-linked network [67]. Such
approach was used to generate TA paclitaxel (PTX) self-
assembly nanoparticles (TAP NPs) in order to potentiate PTX
chemotherapeutic efficiency. The TAP NPs efficiently internal-
ized into the cytoplasm of breast cancer MDA-MB-231 cells
resultinginincreased,incomparisonwithpaclitaxelalone,inhi-
bition of cells proliferation, clones formation, and migration.
Moreover, TAP NPs increased beta-tubulin stabilization and ap-
optosis and limited P-gp-mediated drug efflux [65]. Moreover,
the study of PTX administered in a form of PTX-loaded tannic
acid/poly(N-vinylpyrrolidone) nanoparticles (PTX-NP) showed
that TA exhibits P-gp inhibitory function, whereas the intestinal
retention of a drug is prolonged and trans-epithelial transport
properties are improved. Oral administration of PTX-NP gener-
ated a high oral delivery efficiency and relative oral bioavailabil-
ity of 25.6% in rats, and further displayed a significant tumor-
inhibition effect in a xenograft breast tumor model. These find-
ings confirmed that PTX-NP might be a promising oral drug
formulation for chemotherapy [66].
In another study, an injectable drug delivery system was de-
veloped, involving oxaliplatin (OXA) and TA incorporated into
polymeric nanoparticles in a form of a thermo-sensitive hydrogel,
(OXA/TA NPs-H). Intraperitoneal application of OXA/TA NPs-
H restricted the growth of CT26 peritoneal colon cancer in vivo,
improved the quality of life, and prolonged the survival time of
the model mice, which suggests this drug delivery system can be
applied in colorectal cancer treatment [67].
Another form of nanoparticles with TA was formed by co-
assembling TA and polymer poly (2-oxazoline) and DOX as a
model drug [68]. These polymeric nanoparticles showed high sta-
bility, good biocompatibility, and the cellular uptake. Thus, they
can be considered as promising drug carriers for cancer therapy.
The major challenge in the design of anticancer drug carrier
is the drug release in response to tumor-specific microenvi-
ronments (TMEs) such as low pH, abnormal levels of ROS,
and hypoxic conditions [6972]. Hyaluronic acid, abundant in
synovial fluid and the extracellular matrix, owing specific
binding affinity for CD44-overexpressing cancer cells, has
been used to prepare amphiphilic derivatives, capable of
self-assembling into the nano-sized particles. However, these
nanoparticles (HANPs) were unstable in physiological condi-
tions and released a significant amount of drug into the blood-
stream. This problem has been overcome by preparing metal
(Fe
3+
)-phenolic (TA) network (MPN)-coated HANPs (MPN-
HANPs) as a pH-sensitive nano-carrier for hydrophobic
drugs. DOX-loaded MPN-HANPs exhibited a higher cy-
totoxicity for the squamous cell carcinoma (SCC7), sug-
gesting their potential use as a drug carrier in targeted
cancer therapy [73••].
Interesting application of TA in the treatment of lung can-
cer was described by Hatami et al. [74], who examined the TA
interaction with the lung fluid (LF) the major barrier for the
distribution of drugs to the lungs. They demonstrated that TA
binds to LF and forms self-assemblies, which profoundly en-
hance interaction with lung cancer cells. Thus, TA itself may
be considered as a novel carrier for pharmaceutical drugs such
as gemcitabine, carboplatin, and irinotecan. Therefore, TA,
when used to formulate effective, yet nontoxic anticancer
nanoparticles with drugs, has an excellent potential for trans-
lation from the bench to bedside cancer therapy.
Conclusions
TA showed up to be more versatile molecule than was initially
thought. While the earliest investigationsconcern was TA
chemopreventive potential related to its ability to inhibit car-
cinogenesis initiation and promotion in animal models, over
the years, the knowledge of its biological activities extended
beyond this aspect. It was demonstrated that TA may interfere
with the mechanisms, which might be important for cancer
therapy, e.g. prevention of EMT or decrease in CSC forma-
tion. Moreover, available data indicate that TA may increase
cancer cells sensitization to anticancer drugs and can help over-
coming multidrug resistance. However, TA chemosensitization
properties require more profound research on its effect on non-
tumorigenic cells. In addition, TA can be useful excipient for
generation of drug-loaded nanoparticles. Therefore, TA
certainly deserves further studies.
Funding Information This work is based upon work from COSTAction
NutRedOx-CA16112 supported by COST (European Cooperation in
Science and Technology).
Compliance with Ethical Standards
Conflict of Interest The authors received no financial support in the
writing of this manuscript.
Human and Animal Rights and Informed Consent This article does not
contain any studies with human or animal subjects performed by any of
the authors.
34 Curr Pharmacol Rep (2020) 6:2837
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing, adap-
tation, distribution and reproduction in any medium or format, as long as
you give appropriate credit to the original author(s) and the source, pro-
vide a link to the CreativeCommons licence, and indicate if changes were
made. The images or other third party material in this article are included
in the article's Creative Commons licence, unless indicated otherwise in a
credit line to the material. If material is not included in the article's
Creative Commons licence and your intended use is not permitted by
statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/.
References
Papers of particular interest, published recently, have been
highlighted as:
Of importance
•• Of major importance
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next genera-
tion. Cell. 2011;144:64674. https://doi.org/10.1016/j.cell.2011.02.
013.
2. Sporn MB. Approaches to prevention of epithelial cancer during the
preneoplastic period. Cancer Res. 1976;36(7 PT 2):2699702.
3. Nakamura Y, Tsuji S, Tonogai Y. Method for analysis of tannic acid
and its metabolites in biological samples: application to tannic acid
metabolism in the rat. J Agric Food Chem. 2003;51:3319. https://
doi.org/10.1021/jf020847+.
4. Carbonaro M, Grant G, Pusztai A. Evaluation of polyphenol bio-
availability in rat small intestine. Eur J Nutr. 2001;40:8490. https://
doi.org/10.1007/s003940170020.
5.•• Athar M, Khan WA, Mukhtar H. Effect of dietary tannic acid on
epidermal, lung, and forestomach polycyclic aromatic hydrocarbon
metabolism and tumorigenicity in SENCAR mice. Cancer Res.
1989;49:57848This is one of the first articles describing che-
mopreventive activity of tannic acid.
6. Das M, Mukhtar H, Bik DP, Bickers DR. Inhibition of Epidermal
Xenobiotic Metabolism in SENCAR Mice by Naturally Occurring
Plant Phenols. Cancer Res. 1987;47:760766.
7. Bartolí R, Fernández-Bañares F, Navarro E, Castellà E, Mañé J,
Alvarez M, et al. Effect of olive oil on early and late events of colon
carcinogenesis in rats: modulation of arachidonic acid metabolism
and local prostaglandin E(2) synthesis. Gut. 2000;46:1919. https://
doi.org/10.1136/gut.46.2.191.
8. Baer-Dubowska W, Gnojkowski J,Fenrych W. Effect of tannic acid
on benzo [a]pyrene-DNA adduct formation in mouse epidermis:
comparison with synthetic gallic acid esters. Nutr Cancer.
1997;29:4. https://doi.org/10.1080/01635589709514600.
9.Ignatowicz E, Balana B, Vulimiri SV, Szaefer H, Baer-Dubowska
W. The effect of plant phenolics on the formation of 7,12-
dimethylbenz [a]anthracene-DNA adducts and TPA-stimulated
polymorphonuclear neutrophils chemiluminescence in vitro.
Toxicology. 2003;189:199209. https://doi.org/10.1016/s0300-
483x(03)00148-3 This article reports the ability of tannic acid
to inhibit the formation of DMBA-dAdo adducts.
10. Szaefer H, Cichocki M, Brauze D, Baer-Dubowska W. Alteration in
phase I and II enzyme activities and polycyclic aromatic
hydrocarbons-DNA adduct formation by plant phenolics in mouse
epidermis. Nutr Cancer. 2004;48:707. https://doi.org/10.1207/
s15327914nc4801_10.
11. Andrade RG Jr, Dalvi LT, Silva JM Jr, Lopes GK, Alonso A,
Hermes-Lima M. The antioxidant effect of tannic acid on the
in vitro copper-mediated formation of free radicals. Arch
Biochem Biophys. 2005;437:19. https://doi.org/10.1016/j.abb.
2005.02.016.
12. Khan NS, Ahmad A, Hadi SM. Anti-oxidant, pro-oxidant proper-
ties of tannic acid and its binding to DNA. Chem Biol Interact.
2000;125:17789. https://doi.org/10.1016/s0009-2797(00)00143-
5.
13. Abel EL, Angel JM, Kiguchi K, DiGiovanni J. Multi-stage chem-
ical carcinogenesis in mouse skin: fundamentals and applications.
Nat Protoc. 2009;4:135062. https://doi.org/10.1038/nprot.2009.
120.
14.•• Mukhtar H, Das M, Khan WA, Wang ZY, Bik DP, Bickers DR.
Exceptional activity of tannic acid among naturally occurring plant
phenols in protecting against 7,12-dimethylbenz(a)anthracene-,
benzo(a)pyrene-, 3-methylcholanthrene-, and N-methyl-N-
nitrosourea-induced skin tumorigenesis in mice. Cancer Res.
1988;48:23615This the first report showing anti-carcinogenic
activity of tannic acid.
15. Szaefer H, Krajka-Kuźniak V, Baer-Dubowska W. The effect of
initiating doses of benzo [a] pyrene and 7,12-dimethylbenz [a] an-
thracene on the expression of PAH activating enzymes and its mod-
ulation by plant phenols. Toxicology. 2008;251:2834. https://doi.
org/10.1016/j.tox.2008.07.047.
16. Krajka-Kuźniak V, Baer-Dubowska W. The effects of tannic acid
on cytochrome P450 and phase II enzymes in mouse liver and
kidney. Toxicol Lett. 2003;143:20916. https://doi.org/10.1016/
s0378-4274(03)00177-2.
17. Mikstacka R, Gnojkowski J, Baer-Dubowska W. Effect of natural
phenols on the catalytic activity of cytochrome P450 2E1. Acta
Biochim Pol. 2002;49:91725.
18. Das M, Khan WA, Asokan P, Bickers DR, Mukhtar H. Inhibition of
polycyclic aromatic hydrocarbon-DNA adduct formation in epider-
mis and lungs of SENCAR mice by naturally occurring plant phe-
nols. Cancer Res. 1987;47:76773.
19. Tang MS, Vulimiri SV, Viaje A, Chen JX, Bilolikar DS, Morris RJ,
et al. Both (+/)syn- and (+/)anti-7,12-dimethylbenz [a]anthra-
cene-3,4-diol-1,2-epoxides initiate tumors in mouse skin that pos-
sess -CAA- to -CTA- mutations at codon 61 ofc-H-ras. Cancer Res.
2000;60:568895.
20. Cichocki M, Blumczyńska J, Baer-Dubowska W. Naturally occur-
ring phenolic acids inhibit 12-O-tetradecanoylphorbol-13-acetate
induced NF-kappaB, iNOS and COX-2 activation in mouse epider-
mis. Toxicology. 2010;268:11824. https://doi.org/10.1016/j.tox.
2009.12.013.
21. Cichocki M, Dałek M, Szamałek M, Baer-Dubowska W. Naturally
occurring phenolic acids modulate TPA-induced activation of
EGFR, AP-1, and STATs in mouse epidermis. Nutr Cancer.
2014;66:30814. https://doi.org/10.1080/01635581.2014.864419.
22. Szaefer H, Kaczmarek J, Rybczyńska M, Baer-Dubowska W. The
effect of plant phenols on the expression and activity of phorbol
ester-induced PKC in mouse epidermis. Toxicology. 2007;230:1
10. https://doi.org/10.1016/j.tox.2006.10.001.
23. Majed F, Rashid S, Khan AQ, Nafees S, Ali N, Ali R, et al. Tannic
acid mitigates the DMBA/croton oil-induced skin cancer progres-
sion in mice. Mol Cell Biochem. 2015;399:21728. https://doi.org/
10.1007/s11010-014-2248-3.
24.Karuppagounder V, Arumugam S, Thandavarayan RA,
Pitchaimani V, Sreedhar R, Afrin R, et al. Tannic acid modulates
NFκB signaling pathway and skin inflammation in NC/Nga mice
through PPARγexpression. Cytokine. 2015;76:20613. https://
doi.org/10.1016/j.cyto.2015.05.016 This article reports the
results of the studies showing the mechanism of anti-
inflammatory activity of tannic acid.
Curr Pharmacol Rep (2020) 6:2837 35
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
25. Nepka C, Sivridis E, Antonoglou O, Kortsaris A, Georgellis A,
Taitzoglou I, et al. Chemopreventive activity of very low dose die-
tary tannic acid administration in hepatoma bearing C3H male
mice. Cancer Lett. 1999;141:5762. https://doi.org/10.1016/
s0304-3835(99)00145-7.
26. Zhang J, Song Q, Han X, Zhang Y, Zhang Y, Zhang X, et al. Multi-
targeted protection of acetaminophen-induced hepatotoxicity in
mice by tannic acid. Int Immunopharmacol. 2017;47:95105.
https://doi.org/10.1016/j.intimp.2017.03.027.
27.Booth BW, Inskeep BD, Shah H, Park JP, Hay EJ, Burg KJ. Tannic
acid preferentially targets estrogen receptor-positive breast cancer.
Int J Breast Cancer. 2013;2013:369609. https://doi.org/10.1155/
2013/369609 This is the first of the series of articles describing
estrogen receptor depended effect of tannic acid.
28. Ngobili TA, Shah H, Park JP, Kwist KW, Inskeep B, Burg KJ, et al.
Remodeling of tannic acid crosslinked collagen type I induces ap-
optosis in ER+ breast cancer cells. Anticancer Res. 2015;35:1285
90.
29. Jordan LG, Booth BW. HER2
+
breast cancer cells undergo apopto-
sis upon exposure to tannic acid released from remodeled cross-
linked collagen type I. J Biomed Mater Res A. 2018;106:2632.
https://doi.org/10.1002/jbm.a.36205.
30. Nie F, Liang Y, Jiang B, Li X, Xun H, He W, et al. Apoptotic effect
of tannic acid on fatty acid synthase over-expressed human breast
cancer cells. Tumour Biol. 2016;37:213743. https://doi.org/10.
1007/s13277-015-4020-z.
31. Yang EB, Wei L, Zhang K, Chen YZ, Chen WN. Tannic acid, a
potent inhibitor of epidermal growth factor receptor tyrosine kinase.
J Biochem. 2006;139:495502. https://doi.org/10.1093/jb/mvj050.
32. Darvin P, Joung YH, Kang DY, Sp N, Byun HJ, Hwang TS, et al.
Tannic acid inhibits EGFR/STAT1/3 and enhances p38/STAT1 sig-
naling axis in breast cancer cells. J Cell Mol Med. 2017;21:72034.
https://doi.org/10.1111/jcmm.13015.
33. Karakurt S, Adali O. Tannic acid inhibits proliferation, migration,
invasion of prostate cancer and modulates drug metabolizing and
antioxidant enzymes. Anti Cancer Agents Med Chem. 2016;16:
7819. https://doi.org/10.2174/1871520616666151111115809.
34. Ta LT, Nguyen TTK, Yoo H. Tannic acid-induced apoptosis in
FaDu hypopharyngeal squamous cell carcinoma. Int J Oral Biol.
2019;44:439. https://doi.org/10.11620/IJOB.2019.44.2.43.
35. Darvin P, Baeg SJ, Joung YH, Sp N, Kang DY, Byun HJ, et al.
Tannic acid inhibits the Jak2/STAT3 pathway and induces G1/S
arrest and mitochondrial apoptosis in YD-38 gingival cancer cells.
Int J Oncol. 2015;47:111120. https://doi.org/10.3892/ijo.2015.
3098.
36. Krajka-Kuźniak V, Paluszczak J, Szaefer H, Baer-Dubowska W.
The activation of the Nrf2/ARE pathway in HepG2 hepatoma cells
by phytochemicals and subsequent modulation of phase II and an-
tioxidant enzyme expression. J Physiol Biochem. 2015;71:22738.
https://doi.org/10.1007/s13105-015-0401-4.
37. Mhlanga P, Perumal PO, Somboro AM, Amoako DG, Khumalo
HM, Khan RB. Mechanistic insights into oxidative stress and apo-
ptosis mediated by tannic acid in human liver hepatocellular carci-
noma cells. Int J Mol Sci. 2019;20. https://doi.org/10.3390/
ijms20246145.
38. Sánchez-Carranza JN, Alvarez L, Marquina-Bahena S, Salas-Vidal
E, Cuevas V, Jiménez EW, et al. Phenolic compounds isolated from
Caesalpinia coriaria induce S and G2/M phase cell cycle arrest
differentially and trigger cell death by interfering with microtubule
dynamics in cancer cell lines. Molecules. 2017;22. https://doi.org/
10.3390/molecules22040666.
39.•• Yang P, Ding GB, Liu W, Fu R, Sajid A, Li Z. Tannic acid directly
targets pyruvate kinase isoenzyme M2 to attenuate colon cancer cell
proliferation. Food Funct. 2018;9:554759. https://doi.org/10.
1039/c8fo01161c This article proposes the interesting
mechanism of the attenuation of cancer cell proliferation
through inhibition of glycolytic enzyme.
40. Dong G, Mao Q, Xia W, Xu Y, Wang J, Xu L, et al. PKM2 and
cancer: the function of PKM2 beyond glycolysis. Oncol Lett.
2016;11:19806. https://doi.org/10.3892/ol.2016.4168.
41. Zielińska-Przyjemska M, Kaczmarek M, Krajka-Kuźniak V,
Łuczak M, Baer-Dubowska W. The effect of resveratrol, its natu-
rally occurring derivatives and tannic acid on the induction of cell
cycle arrest and apoptosis in rat C6 and human T98G glioma cell
lines. Toxicol in Vitro. 2017;43:6975. https://doi.org/10.1016/j.
tiv.2017.06.004.
42. Bona NP, Pedra NS, Azambuja JH, Soares MSP, Spohr L,
Gelsleichter NE, et al. Tannic acid elicits selective antitumoral ac-
tivity in vitro and inhibits cancer cell growth in a preclinical model
of glioblastoma multiforme. Metab Brain Dis. 2019;26:28393.
https://doi.org/10.1007/s11011-019-00519-9.
43. Dongre A, Weinberg RA. New insights into the mechanisms of
epithelial-mesenchymal transition and implications for cancer. Nat
Rev Mol Cell Biol. 2019;20:6984. https://doi.org/10.1038/
s41580-018-0080-4.
44. Antony J, Thiery JP, Huang RY. Epithelial-to-mesenchymal transi-
tion: lessons from development, insights into cancer and the poten-
tial of EMT-subtype based therapeutic intervention. Phys Biol.
2019;16:041004. https://doi.org/10.1088/1478-3975/ab157a.
45. Roche J. The epithelial-to-mesenchymal transition in cancer. Cancers
(Basel). 2018;10. https://doi.org/10.3390/cancers10020052.
46.•• Kim DA, Choi HS, Ryu ES, Ko J, Shin HS, Lee JM, et al. Tannic
acid attenuates the formation of cancer stem cells by inhibiting NF-
κB-mediated phenotype transition of breast cancer cells. Am J
Cancer Res. 2019;9:166481 This article points out the possibil-
ity of attenuation of cancer stem cells formation by tannic acid
through inhibition of ETM.
47. Pattarayan D, Sivanantham A, Krishnaswami V, Loganathan L,
Palanichamy R, Natesan S, et al. Tannic acid attenuates TGF-β1-
induced epithelial-to-mesenchymal transition by effectively inter-
vening TGF-βsignaling in lung epithelial cells. J Cell Physiol.
2018;233:251325. https://doi.org/10.1002/jcp.26127.
48. Avila-Carrasco L, Majano P, Sánchez-Toméro JA, Selgas R, López-
Cabrera M, Aguilera A, et al. Natural plants compounds as modu-
lators of epithelial-to-mesenchymal transition. Front Pharmacol.
2019;10:715. https://doi.org/10.3389/fphar.2019.00715.
49. SongD,ZhaoJ,DengW,LiaoY,HongX,HouJ.Tannicacid
inhibits NLRP3 inflammasome-mediated IL-1βproduction via
blocking NF-κB signaling in macrophages. Biochem Biophys Res
Commun. 2018;503:307885. https://doi.org/10.1016/j.bbrc.2018.
08.096.
50. Moossavi M, Parsamanesh N, Bahrami A, Atkin SL, Sahebkar A.
Role of the NLRP3 inflammasome in cancer. Mol Cancer. 2018;17:
158. https://doi.org/10.1186/s12943-018-0900-3.
51. Chen X, Beutler JA, McCloud TG, Loehfelm A, Yang L, Dong HF,
et al. Tannic acid is an inhibitor of CXCL12 (SDF-1alpha)/CXCR4
with antiangiogenic activity. Clin Cancer Res. 2003;9:311523.
52. Guo F, Wang Y, Liu J, Mok SC, Xue F, Zhang W. CXCL12/
CXCR4: a symbiotic bridge linking cancer cells and their stromal
neighbors in oncogenic communication networks. Oncogene.
2016;35:81626. https://doi.org/10.1038/onc.2015.139.
53.Garg AK, Buchholz TA, Aggarwal BB. Chemosensitization and
radiosensitization of tumors by plant polyphenols. Antioxid
Redox Signal. 2005;7:163047. https://doi.org/10.1089/ars.2005.
7.1630 This article provides a comprehensive review on
chemo- and radiosensitization of tumor cells by plant
polyphenols.
54. Shen M, Chan TH, Dou QP. Targeting tumor ubiquitin-proteasome
pathway with polyphenols for chemosensitization. Anti Cancer
Agents Med Chem. 2012;12:891901. https://doi.org/10.2174/
187152012802649978.
36 Curr Pharmacol Rep (2020) 6:2837
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
55. Nam S, Smith DM, Dou QP. Tannic acid potently inhibits tumor
cell proteasome activity, increases p27 and Bax expression, and
induces G1 arrest and apoptosis. Cancer Epidemiol Biomark Prev.
2001;10:10838.
56. Sasaki Y, Hozumi M, Fujimori H, Murakami Y, Koizumi F, Inoue
K, et al. PARG inhibitors and functional PARG inhibition models.
Curr Protein Pept Sci. 2016;17:64153. https://doi.org/10.2174/
1389203717666160419145130.
57. Sun Y, Zhang T, Wang B, Li H, Li P. Tannic acid, an inhibitor of
poly (ADP-ribose) glycohydrolase, sensitizes ovarian carcinoma
cells to cisplatin. Anti-Cancer Drugs. 2012;23:97990. https://doi.
org/10.1097/CAD.0b013e328356359f.
58. Li H, Krstin S, Wink M. Modulation of multidrug resistant in cancer
cells by EGCG, tannic acid and curcumin. Phytomedicine.
2018;50:21322. https://doi.org/10.1016/j.phymed.2018.09.169.
59.Khan M, Maryam A, Mehmood T, Zhang Y, Ma T. Enhancing
activity of anticancer drugs in multidrug resistant tumors by mod-
ulating P-glycoprotein through dietary nutraceuticals. Asian Pac J
Cancer Prev. 2015;16:68319. https://doi.org/10.7314/apjcp.2015.
16.16.6831 This article presents the mechanism of overcoming
MDR by nutraceuticals.
60. Naus P, Henson R, Bleeker G, Wehbe H, Meng F, Patel T. Tannic
acid synergizes the cytotoxicity of chemotherapeutic drugs in hu-
man cholangiocarcinoma by modulating drug efflux pathways. J
Hepatol. 2007;46:2229. https://doi.org/10.1016/j.jhep.2006.08.
012.
61. Xiao J, Sun GB, Sun B, Wu Y, He L, Wang X, et al. Kaempferol
protects against doxorubicin-induced cardiotoxicity in vivo and
in vitro. Toxicology. 2012;292:5362. https://doi.org/10.1016/j.
tox.2011.11.018.
62. Zhang J, Cui L, Han X, Zhang Y, Zhang X, Chu X, et al. Protective
effects of tannic acid on acute doxorubicin-induced cardiotoxicity:
involvement of suppression in oxidative stress, inflammation, and
apoptosis. Biomed Pharmacother. 2017;93:125360. https://doi.
org/10.1016/j.biopha.2017.07.051.
63. Sheng H, Ogawa T, Niwano Y, Sasaki K, Tachibana K. Effects of
polyphenols on doxorubicin-induced oral keratinocyte cytotoxicity
and anticancer potency against oral cancer cells. J Oral Pathol Med.
2018;47:36874. https://doi.org/10.1111/jop.12685.
64.•• Chowdhury P, PKB N, Hatami E, Wagh S, Dan N, Tripathi MK,
et al. Tannic acid-inspired paclitaxel nanoparticles for enhanced
anticancer effects in breast cancer cells. J Colloid Interface Sci.
2019;535:13348. https://doi.org/10.1016/j.jcis.2018.09.072 This
article reports the results of a comprehensive study on tannic
acid as excipient of anti-cancer drug-paclitaxel.
65. Chen YN, Jiao C, Zhao Y, Zhang J, Wang H. Self-assembled poly-
vinyl alcohol-tannic acid hydrogels with diverse microstructures
and good mechanical properties. ACS Omega. 2018;3:1178895.
https://doi.org/10.1021/acsomega.8b02041.
66. Le Z, Chen Y, Han H, Tian H, Zhao P, Yang C, et al. Hydrogen-
bonded tannic acid-based anticancer nanoparticle for enhancement
of oral chemotherapy. ACS Appl Mater Interfaces. 2018;10:42186
97. https://doi.org/10.1021/acsami.8b18979.
67. Ren Y, Li X, Han B, Zhao N, Mu M, Wang C, et al. Improved anti-
colorectal carcinomatosis effect of tannic acid co-loaded with
oxaliplatin in nanoparticles encapsulated in thermosensitive hydro-
gel. Eur J Pharm Sci. 2019;128:27989. https://doi.org/10.1016/j.
ejps.2018.12.007.
68. Liu F, Kozlovskaya V, Zavgorodnya O, Martinez-Lopez C,
Catledge S, Kharlampieva E. Encapsulation of anticancer drug by
hydrogen-bonded multilayers of tannic acid. Soft Matter. 2014;10:
923747. https://doi.org/10.1039/c4sm01813c.
69. Wang S, Huang P, Chen X. Stimuli-responsive programmed specif-
ic targeting in nanomedicine. ACS Nano. 2016;10:29914. https://
doi.org/10.1021/acsnano.6b00870.
70. Son S, Deepagan VG, Shin S, Ko H, Min J, UmW, et al. Ultrasmall
gold nanosatellite-bearing transformable hybrid nanoparticles for
deep tumor penetration. Acta Biomater. 2018;79:294305. https://
doi.org/10.1016/j.actbio.2018.08.019.
71. Deepagan VG, Kwon S, You DG, Nguyen VQ, Um W, Ko H, et al.
In situ diselenide-crosslinked polymeric micelles for ROS-mediated
anticancer drug delivery. Biomaterials. 2016;103:5666. https://
doi.org/10.1016/j.biomaterials.2016.06.044.
72. Thambi T, Park JH, Lee DS. Hypoxia-responsive nanocarriers for
cancer imaging and therapy: recent approaches and future perspec-
tives. Chem Commun (Camb). 2016;52:8492500. https://doi.org/
10.1039/c6cc02972h.
73.•• Shin JM, Choi GH, Song SH, Ko H, Lee ES, Lee JA, et al. Metal-
phenolic network-coated hyaluronic acid nanoparticles for pH-
responsive drug delivery. Pharmaceutics. 2019;11. https://doi.org/
10.3390/pharmaceutics11120636 This article reports tannic acid
application in pH responsive drug delivery.
74. Hatami E, Nagesh PKB, Chowdhury P, Chauhan SC, Jaggi M,
Samarasinghe AE, et al. Tannic acid-lung fluid assemblies promote
interaction and delivery of drugs to lung cancer cells. Pharmaceutics.
2018;10. https://doi.org/10.3390/pharmaceutics10030111.
PublishersNoteSpringer Nature remains neutral with regard to jurisdic-
tional claims in published maps and institutional affiliations.
Curr Pharmacol Rep (2020) 6:2837 37
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... To date, the literature has not reported sufficient experimental results demonstrating the efficacy of a cell-permeable allosteric inhibitor of PKL, highlighting a significant gap in the therapeutic landscape for these liver conditions. Moreover, current studies have not conclusively determined whether polyphenolic compounds can penetrate human cells, a crucial aspect of their effectiveness in treating cellular-level disorders [13][14][15][16][17][18][19][20][21][22][23][24]. Therefore, The accumulation of fat in the liver is now known to be a mechanism that contributes to hepatic injury in the context of metabolic diseases, such as Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD). ...
... To date, the literature has not reported sufficient experimental results demonstrating the efficacy of a cell-permeable allosteric inhibitor of PKL, highlighting a significant gap in the therapeutic landscape for these liver conditions. Moreover, current studies have not conclusively determined whether polyphenolic compounds can penetrate human cells, a crucial aspect of their effectiveness in treating cellular-level disorders [13][14][15][16][17][18][19][20][21][22][23][24]. Therefore, recent studies have been conducted to develop PKLR inhibitors, with some showing active bioavailability in cell lysates of hepatic cancer lines [20]. ...
Article
Full-text available
Metabolic dysfunction-associated fatty liver disease (MAFLD) presents a significant global health challenge, characterized by the accumulation of liver fat and impacting a considerable portion of the worldwide population. Despite its widespread occurrence, effective treatments for MAFLD are limited. The liver-specific isoform of pyruvate kinase (PKL) has been identified as a promising target for developing MAFLD therapies. Urolithin C, an allosteric inhibitor of PKL, has shown potential in preliminary studies. Expanding upon this groundwork, our study delved into delineating the structure-activity relationship of urolithin C via the synthesis of sulfone-based urolithin analogs. Our results highlight that incorporating a sulfone moiety leads to substantial PKL inhibition, with additional catechol moieties further enhancing this effect. Despite modest improvements in liver cell lines, there was a significant increase in inhibition observed in HepG2 cell lysates. Specifically, compounds 15d, 9d, 15e, 18a, 12d, and 15a displayed promising IC50 values ranging from 4.3 µM to 18.7 µM. Notably, compound 15e not only demonstrated a decrease in PKL activity and triacylglycerol (TAG) content but also showed efficient cellular uptake. These findings position compound 15e as a promising candidate for pharmacological MAFLD treatment, warranting further research and studies.
... In recent years, the exploration of natural compounds with potential therapeutic benefits has gained considerable momentum, driven by the pursuit of novel and effective treatments with fewer side effects [1,2]. Plant possess numerous bioactive constituents and have been considered for new drug molecules. ...
... These free radicals, originating from both endogenous metabolic processes and exogenous sources, play a pivotal role in modifying various biomolecules such as proteins, lipids, and nucleic acids, thereby disrupting redox reactions and contributing to heightened oxidative stress. The latter has been implicated in a wide spectrum of health disorders including neurodegenerative conditions, respiratory ailments, diabetes, and in several forms of cancer (lung, colon, breast, bladder, and prostate cancers) [2,7]. Moreover, the anticancer potential of tannins, particularly in selectively inducing apoptosis in cancer cells while sparing normal cells, has garnered significant attention [8]. ...
Article
Full-text available
Citation: Ambreen M, Mirza SA, Bano Z, et al. Antioxidant and anticancer activity of tannins isolated from callus cultures of achyranthes aspera L. Sci Inq Rev. 2024;8(2):1-21. https://doi. ABSTRACT In the current study, the impact of oxidative stress induced by free radicals generated during metabolic processes was investigated. It was found to be linked to a variety of diseases due to their detrimental effects on nucleic acids and proteins. The primary focus remained on investigating the free radical-scavenging properties and potential anticancer activities of tannins found in callus cultures of A. aspera. It was observed that the induction of callus formation was notably successful when using leaf and root explants, resulting in a callus index as high as 160, as compared to stem explants in the presence of auxins. The extraction yield of callus tannins was the most abundant in chloroform extracts, although the overall antioxidant activity was comparable among chloroform, methanol, and petroleum ether callus extracts. Among these extracts, the chloroform extract from stem callus cultures grown on the MSDN medium showed the highest total antioxidant activity. Notably, tannins extracted from A. aspera leaf callus extracts displayed significant anticancer effects against the Jurket cell line. These effects were evaluated through measures such as cell viability and colony formation. The anticancer activity was notably higher in callus culture extracts as compared to the control. In summary, the results showed that in vitro biomass production can be a valuable approach for augmenting the production of bioactive compounds, such as tannins, from selected medicinal plants including A. aspera. Tannins extracted from A. aspera hold promise for their potential role in the development of new medicines.
... Encouragingly, the cumulative body of knowledge has firmly demonstrated the anticancer effects as well as the underline mechanisms of these class of compounds. 22,[57][58][59] The other phytochemical group, steroids, are also potential anticancer candidates due to their specific affinity toward various receptors and enzymes. 40,43 Importantly, corticosteroids have been widely used as supportive treatment for cancer patients to reduce the related complications and side effects of cancer treatments. ...
Article
Full-text available
Background Romdoul ( Sphaerocoryne affinis) is a flowering plant of the Annonaceae family and has been used customarily in folk medicine. The bioactivities of this plant, especially the anti-cancer effect, however, remain surprisingly few. Objective this study aimed to elucidate the anti-leukemic effect of romdoul fruit extracts and their underlining mechanisms. Methods The extracts were prepared from fresh fruits and the phytochemical contents were evaluated by biochemical assays and HPLC method. The promising extract was identified via the inhibition of HL60 as well as normal NIH-3T3 cell densities utilizing MTT assay. The underline mechanism of the extract’s effect was studied by accessing the treated HL60 cell population overtime (via MTT assay). The morphology of abnormal cells was examined by bright-field microscopic imaging. Hallmarks of apoptosis including nucleus characteristics and caspase 3 activation were analyzed by fluorescence imaging. The underline mechanisms of apoptosis and proliferation inhibition were accessed via RT-qPCR examination of involved genes. Results Our findings showed that the ethyl acetate extract of romdoul fruit (SA-EA) was found to be an exceptional anti-leukemic candidate (IC 50 was as low as 4.11 μg/mL). More interestingly, the treated HL60 cells expressed nuclear fragmentation and caspase 3 activation, indicating the effect could follow an apoptotic mechanism. Importantly, the transcription assessment of apoptotic and proliferative genes suggested that SA-EA might suppress the growth of HL60 cells and induce p21-dependent apoptotic pathway. Conclusion This study demonstrated one of the first scientific evidence for the anti-cancer activity of Sphaerocoryne affinis fruit-derived extract. Thus, our findings exhibited a novel and promising anti-leukemic candidate for future studies.
... Penelitian interaksi molekuler asam galat dengan target protein telah dilakukan oleh beberapa peneliti dengan metode molekuler docking, termasuk penelitian yang dilakukan oleh Yang et al 8 menunjukkan bahwa asam galat memiliki potensi sebagai kandidat obat untuk pengobatan kanker payudara dan hati dengan menghambat aktivitas epidermal growth factor receptor (EGFR) tirosin kinase. Derivat asam galat, yaitu aril-3,4,5trimethyl gallate juga berpotensi sebagai anti-inflamasi dengan menghambat aktivitas COX-1 dan COX-2. ...
Article
Full-text available
Gallic acid is a secondary metabolite compound found in several natural products and has several cytotoxic activities against some cell lines such as breast, colon, prostate, and cervical cancer. This research was aimed to study the stability, affinity, and interaction of gallic acid compounds and their derivatives as well as comparative ligand against androgen receptor through a computational approach, namely in silico molecular docking. The research was conducted in four stages, namely the preparation and optimization of ligand compounds and androgen receptor macromolecules, docking simulation process, and analysis and visualization of simulation results. Computational studies show that gallic acid derivatives provide better inhibitory activity than gallic acid and comparative ligand with an average ΔG value of-7,34 kcal/mol. Compound 3,4,5 tri-methoxy-phenyl-ethyl gallate had the best binding energy value of 8.20 kcal/mol. This was demonstrated by stability, affinity, and strong interactions towards active site amino acids at the androgen receptor. Besides, the formed hydrogen bond was strong enough with an average distance of <3.5 Ǻ which might lead to inhibition and apoptosis through activation of caspase 3 and 9 in prostate cancer cells. Thus, gallic acid derivatives have the potential as inhibitors of androgen receptors and become candidates for new drugs in prostate cancer. Abstrak Asam galat merupakan senyawa metabolit sekunder yang ditemukan pada beberapa produk alami dan memiliki beberapa aktivitas sitotoksik terhadap beberapa sel line seperti kanker payudara, kolon, prostat, dan serviks. Penelitian ini bertujuan mempelajari stabilitas, afinitas, dan interaksi senyawa asam galat dan turunannya, serta ligan pembanding terhadap reseptor androgen melalui pendekatan secara komputasi yaitu in silico molekuler docking. Penelitian dilakukan dengan empat tahapan yaitu preparasi dan optimasi senyawa ligan, serta makromolekul reseptor androgen, proses simulasi docking dan analisis, serta visualisasi hasil simulasi. Studi komputasi menunjukkan bahwa senyawa turunan asam galat memberikan aktivitas inhibisi yang lebih baik dibandingkan asam galat dan ligan pembanding dengan rata-rata nilai ΔG sebesar-7,34 kkal/mol. Senyawa 3,4,5 trimetoksi-feniletil galat memiliki nilai binding energy terbaik sebesar-8,20 kkal/mol. Hal ini ditunjukkan dengan stabilitas, afinitas, dan interaksi yang kuat terhadap situs aktif asam amino pada reseptor androgen. Selain itu, terbentuk ikatan hidrogen cukup kuat dengan rata-rata jarak ikatan sebesar <3,5Ǻ yang dapat menimbulkan inhibisi maupun apoptosis melalui aktivasi caspase 3 dan 9 pada sel kanker prostat. Dengan demikian, senyawa turunan asam galat memiliki potensi sebagai inhibitor terhadap reseptor androgen dan menjadi kandidat obat baru pada kanker prostat. Kata kunci: studi komputasi, molekular docking, asam galat dan turunannya, reseptor androgen, kanker prostat Pendahuluan Asam galat (AG; 3,4,5-trihydroxyl-benzoic acid) merupakan senyawa metabolit sekunder yang dapat ditemukan pada beberapa produk alami. 1 AG dilaporkan sebagai penangkap radikal bebas dan inducer dari diferensiasi dan apoptosis pada sel line kanker payudara,
... As an antagonist of CXCL12/CXCR4, it operates to preferentially target BC cells, sparing normal epithelial cells, and impede migration [45][46][47]. Furthermore, TA reduces the cardiotoxicity caused by doxorubicin and works in concert with paclitaxel, highlighting its potential as a pan-tumor suppressor in the treatment of BC [48]. In liver cancer, tannic acid (TA) demonstrates dual effects as an anti-fibrotic and anticancer drug. ...
Article
Full-text available
Cancer continues to be the leading cause of mortality worldwide, despite significant attempts to identify new risk factors, create earlier diagnostic indicators, and investigate alternative therapy options. This rising burden of cancer worldwide results in the advancement of innovative methods of cancer prevention and treatment leading to the developed idea of cancer chemoprevention, with an emphasis on employing natural substances included in the diet to inhibit tumor development. Newer techniques like immunotherapy and gene therapy are used in addition to conventional cancer therapies including radiation, chemotherapy, and surgery. Nonetheless, the potential of natural substances needs to be exploited. Natural products which can be proteins, carbohydrates, or nucleic acids in their forms or conjugated have tendencies to trigger innate and adaptive immunity, infusion reactions, inflammatory responses, hypersensitivity reactions, and other immunological responses. Numerous bioactive substances found in Cola nitida, such as flavonoids, catechin, tannins, alkaloids, and phenolics, have been shown to have promising medicinal qualities, such as the ability to prevent cancer, function as antioxidants, and stop the proliferation of cancer cells using some of the immunological mechanism. This study explores the molecular routes by which these bioactive chemicals cause apoptosis, inhibit angiogenesis, and modify signal transduction pathways to achieve their anticancer effects. The study concludes by highlighting the tremendous multimodal therapeutic potential of Cola nitida in the creation of safer and more effective cancer treatments.
... Although we cannot precisely explain these findings, they could be attributed, at least in part, to an intracellular downregulation of antioxidant enzyme transcription in a concentration-dependent manner. In addition, these results may suggest a potential biphasic response of antioxidant enzymes to NC-TA-ZN, where lower concentrations may induce an upregulation of antioxidant defense mechanisms, while higher concentrations may induce a compensatory downregulation, possibly due to cellular stress responses or regulatory feedback mechanisms [67]. ...
Article
Full-text available
Glioblastoma (GBM) is a highly aggressive brain tumor with a poor prognosis. Polyphenols, known for their antioxidant and antitumor properties, may be potential therapeutic targets in GBM. Advances in nanomedicine have enabled the development of nanoparticles to improve the pharmacokinetics of polyphenols like tannic acid (TA). In this study, we investigated the therapeutic effects of 1.5% (w/w) TA-loaded zein-coated nanocapsules (NC-TA-ZN) on glioma cell lines (C6, U87MG). The cells were exposed to concentrations of 100, 125, 150 and 175 µg/mL for 72 h. NC-TA-ZN demonstrated cytotoxic effects, as evidenced by a significant reduction in cell viability (up to 47.10% in C6, 41.02% in U87MG) and cell biomass (up to 49.52% in C6, 66.65% in U87MG). It also reduced reactive oxygen species (up to 79.02% in C6, 60.77% in U87MG) and increased total sulfhydryl content (up to 97.37% in C6, 330.7% in U87MG). Additionally, it increased superoxide dismutase (up to 147.4% in C6, 341.4% in U87MG) and catalase activity (up to 287.6% in C6, 544.4% in U87MG). These results demonstrated the potential antioxidant effect of NC-TA-ZN. Lastly, NC-TA-ZN inhibited cell migration in C6 cells by 40% and 65% at 25 and 50 µg/mL, respectively. These results are significant given the relationship between oxidative stress and GBM development and progression, and suggest the potential of NC-TA-ZN as a therapeutic approach to GBM. Future studies will focus on understanding the mechanisms of cell death and the signaling pathways involved in the antiglioma effect elicited by nanoparticles of TA.
Article
Coffee is not only a delicious beverage but also an important dietary source of natural antioxidants. We live in a world where it is impossible to avoid pollution, stress, food additives, radiation, and other sources of oxidants that eventually lead to severe health disorders. Fortunately, there are chemicals in our diet that counteract the hazards posed by the reactive species that trigger oxidative stress. They are usually referred to as antioxidants; some of them can be versatile compounds that exert such a role in many ways. This review summarizes, from a chemical point of view, the antioxidant effects of relevant molecules found in coffee. Their mechanisms of action, trends in activity, and the influence of media and pH in aqueous solutions, are analyzed. Structure-activity relationships are discussed, and the protective roles of these compounds are examined. A particular section is devoted to derivatives of some coffee components, and another one to their bioactivity. The data used in the analysis come from theoretical and computational protocols, which have been proven to be very useful in this context. Hopefully, the information provided here will pro-mote further investigations into the amazing chemistry contained in our morning coffee cup. Resumen. El café no solo es una bebida deliciosa, sino también una importante fuente dietética de antioxidantes naturales. Vivimos en un mundo donde es imposible evitar la contaminación, el estrés, los aditivos alimentarios, la radiación y otras fuentes de oxidantes que eventualmente conducen a trastornos de salud graves. Afortunadamente, existen sustancias químicas en nuestra dieta que contrarrestan los peligros planteados por las especies reactivas que desencadenan el estrés oxidativo. Por lo general, se les denomina antioxidantes; algunos de ellos pueden ser compuestos versátiles que ejercen dicho papel de muchas maneras. Este artículo de revisión resume, desde un punto de vista químico, los efectos antioxidantes de moléculas relevantes encontradas en el café. Se analizan sus mecanismos de acción, tendencias en la actividad y la influencia del medio y el pH en soluciones acuosas. Se discuten las relaciones estructura-actividad, y se examinan los roles protectores de estos compuestos. Se dedica una sección particular a los derivados de algunos componentes del café, y otra a su bioactividad. Los datos utilizados en el análisis provienen de protocolos teóricos y computacionales, que han demostrado ser muy útiles en este contexto. Se espera que la información proporcionada aquí promueva investigaciones futuras sobre la química contenida en nuestra taza de café matutina.
Article
Full-text available
The study investigated the cytotoxic effect of a natural polyphenolic compound Tannic acid (TA) on human liver hepatocellular carcinoma (HepG2) cells and elucidated the possible mechanisms that lead to apoptosis and oxidative stress HepG2 cell. The HepG2 cells were treated with TA for 24 h and various assays were conducted to determine whether TA could induce cell death and oxidative stress. The cell viability assay was used to determine the half maximal inhibitory concentration (IC 50), caspase activity and cellular ATP were determined by luminometry. Microscopy was employed to determine deoxyribonucleic acid (DNA) integrity, while thiobarbituric acid (TBARS) and nitric oxide synthase (NOS) assays were used to elucidate cellular reactive oxygen species (ROS) and reactive nitrogen species (RNS), respectively. Western blotting was used to confirm protein expression. The results revealed that tannic acid induced caspase activation and increased the presence of cellular ROS and RNS, while downregulating antioxidant expression. Tannic acid also showed increased cell death and increased DNA fragmentation. In conclusion, TA was able to induce apoptosis by DNA fragmentation via caspase-dependent and caspase-independent mechanism. It was also able to induce oxidative stress, consequently contributing to cell death.
Article
Full-text available
Although self-assembled nanoparticles (SNPs) have been used extensively for targeted drug delivery, their clinical applications have been limited since most of the drugs are released into the blood before they reach their target site. In this study, metal-phenolic network (MPN)-coated SNPs (MPN-SNPs), which consist of an amphiphilic hyaluronic acid derivative, were prepared to be a pH-responsive nanocarrier to facilitate drug release in tumor microenvironments (TME). Due to their amphiphilic nature, SNPs were capable of encapsulating doxorubicin (DOX), chosen as the model anticancer drug. Tannic acid and FeCl3 were added to the surface of the DOX-SNPs, which allowed them to be readily coated with MPNs as the diffusion barrier. The pH-sensitive MPN corona allowed for a rapid release of DOX and effective cellular SNP uptake in the mildly acidic condition (pH 6.5) mimicking TME, to which the hyaluronic acid was exposed to facilitate receptor-mediated endocytosis. The DOX-loaded MPN-SNPs exhibited a higher cytotoxicity for the cancer cells, suggesting their potential use as a drug carrier in targeted cancer therapy.
Article
Full-text available
Glioblastoma is a devastating tumor affecting the central nervous system with infiltrative capacity, high proliferation rate and chemoresistance. Therefore, it is urgent to find new therapeutic alternatives that improve this prognosis. Herein, we focused on tannic acid (TA) a polyphenol with antioxidant and antiproliferative activities. In this work, the antitumor and antioxidant effects of TA on rat (C6) glioblastoma cells and their cytotoxicity relative to primary astrocyte cultures were evaluated in vitro. Cells were exposed to TA of 6.25 to 75 μM for 24, 48 and/or 72 h. In addition, colony formation, migration and cell adhesion were analyzed and flow cytometry was used to analyze cell death and cell cycle. Next, the action of TA was evaluated in a preclinical glioblastoma model performed on Wistar rats. In this protocol, the animals were treated with a dose of 50 mg/kg/day TA for 15 days. Our results demonstrated that TA induced in vitro selective antiglioma activity, not demonstrating cytotoxicity in astrocyte culture. It induced cell death by apoptosis and cell cycle arrest, reducing formation and size of colonies, cell migration/adhesion and showing to be a potential antioxidant. Interestingly, the antiglioma effect was also observed in vivo, as TA decreased tumor volume by 55%, accompanied by an increase in the area of intratumoral necrosis and infiltration of lymphocytes without causing systemic damage. To the best of our knowledge, this is the first study to report TA activity in a GBM preclinical model. Thus, this natural compound is promising as a treatment for glioblastoma.
Article
Full-text available
Cancer stem cells (CSCs) are innately resistant to standard therapies, which positions CSCs in the focus of anti-cancer research. In this study, we investigated the potential inhibitory effect of tannic acid (TA) on CSCs. Our data demonstrated that TA (10 μM), at the concentration not inhibiting the proliferation of normal mammary cells (MCF10A), inhibited the formation and growth of mammosphere in MCF7, T47D, MDA-MB-231 cells shown as a decrease in mammosphere formation efficiency (MFE), cell number, diameter of mammosphere, and ALDH1 activity. NF-κB pathway was activated in the mammosphere indicated by an up-regulation of p65, a degradation of IκBα, and an increased IL-6. The inhibition of NF-κB pathway via gene silencing of p65 (sip65), NF-κB inhibitor (PDTC), or IKK inhibitor (Bay11-7082) alleviated MFE. Other CSCs markers such as an increase in ALDH1 and CD44high/CD24low ratio were ameliorated by sip65. TA also alleviated TGFβ-induced EMT, increase in MFE, and NF-κB activation. In murine xenograft model, TA reduced tumor volume which was associated with a decrease in CD44high/CD24low expression and IKK phosphorylation. These results suggest that TA negatively regulates CSCs by inhibiting NF-κB activation and thereby prevents cancer cells from undergoing EMT and CSCs formation, and may thus be a promising therapy targeting CSCs.
Article
Full-text available
Epithelial-to-mesenchymal transition (EMT) is a self-regulated physiological process required for tissue repair that, in non-controled conditions may lead to fibrosis, angiogenesis, loss of normal organ function or cancer. Although several molecular pathways involved in EMT regulation have been described, this process does not have any specific treatment. This article introduces a systematic review of effective natural plant compounds and their extract that modulates the pathological EMT or its deleterious effects, through acting on different cellular signal transduction pathways both in vivo and in vitro. Thereby, cryptotanshinone, resveratrol, oxymatrine, ligustrazine, osthole, codonolactone, betanin, tannic acid, gentiopicroside, curcumin, genistein, paeoniflorin, gambogic acid and Cinnamomum cassia extracts inhibit EMT acting on transforming growth factor-β (TGF-β)/Smads signaling pathways. Gedunin, carnosol, celastrol, black rice anthocyanins, Duchesnea indica, cordycepin and Celastrus orbiculatus extract downregulate vimectin, fibronectin and N-cadherin. Sulforaphane, luteolin, celastrol, curcumin, arctigenin inhibit β-catenin signaling pathways. Salvianolic acid-A and plumbagin block oxidative stress, while honokiol, gallic acid, piperlongumine, brusatol and paeoniflorin inhibit EMT transcription factors such as SNAIL, TWIST and ZEB. Plectranthoic acid, resveratrol, genistein, baicalin, polyphyllin I, cairicoside E, luteolin, berberine, nimbolide, curcumin, withaferin-A, jatrophone, ginsenoside-Rb1, honokiol, parthenolide, phoyunnanin-E, epicatechin-3-gallate, gigantol, eupatolide, baicalin and baicalein and nitidine chloride inhibit EMT acting on other signaling pathways (SIRT1, p38 MAPK, NFAT1, SMAD, IL-6, STAT3, AQP5, notch 1, PI3K/Akt, Wnt/β-catenin, NF-κB, FAK/AKT, Hh). Despite the huge amount of preclinical data regarding EMT modulation by the natural compounds of plant, clinical translation is poor. Additionally, this review highlights some relevant examples of clinical trials using natural plant compounds to modulate EMT and its deleterious effects. Overall, this opens up new therapeutic alternatives in cancer, inflammatory and fibrosing diseases through the control of EMT process.
Article
Full-text available
Abstract Inflammasomes are large intracellular multi-protein signalling complexes that are formed in the cytosolic compartment as an inflammatory immune response to endogenous danger signals. The formation of the inflammasome enables activation of an inflammatory protease caspase-1, pyroptosis initiation with the subsequent cleaving of the pro-inflammatory cytokines interleukin (IL)-1β and proIL-18 to produce active forms. The inflammasome complex consists of a Nod-like receptor (NLR), the adapter apoptosis-associated speck-like (ASC) protein, and Caspase-1. Dysregulation of NLRP3 inflammasome activation is involved tumor pathogenesis, although its role in cancer development and progression remains controversial due to the inconsistent findings described. In this review, we summarize the current knowledge on the contribution of the NLRP3 inflammasome on potential cancer promotion and therapy.
Article
Epithelial-to-mesenchymal transition (EMT) is a fundamental developmental process wherein polarized epithelial cells lose their junctional architecture and apical-basal polarity to become motile mesenchymal cells, and there is emerging evidence for its role in propagating tumor dissemination. While many multifaceted nodules converge onto the EMT program, in this review we will highlight the fundamental biology of the signaling schemas that enable EMT. In many cancers, the property of tumor dissemination and metastasis is closely associated with re-enabling developmental properties such as EMT. We discuss the molecular complexity of the tumor heterogeneity in terms of EMT-based gene expression molecular subtypes, and the rewiring of critical signaling nodules in the subtypes displaying higher degrees of EMT can be therapeutically exploited. Specifically in the context of a deadly malignancy such as ovarian cancer where there are no defined mutations or limited biomarkers for developing targeted therapy or personalized medicine, we highlight the importance of identifying EMT-based subtypes that will improve therapeutic intervention. In ovarian cancer, the poor prognosis mesenchymal 'Mes' subtype presents with amplified signaling of the receptor tyrosine kinase (RTK) AXL, extensive crosstalk with other RTKs such as cMET, EGFR and HER2, and sustained temporal activation of extracellular-signal regulated kinase (ERK) leading to induction of EMT transcription factor Slug, underscoring a pathway addiction in Mes that can be therapeutically targeted. We will further examine the emergence of therapeutic modalities in these EMT subtypes and finally conclude with potential interdisciplinary biophysical methodologies to provide additional insights in deciphering the mechanistic and biochemical aspects of EMT. This review intends to provide an overview of the cellular and molecular changes accompanying epithelial-to-mesenchymal transition (EMT) in development and the requisition of this evolutionarily conserved pathway in cancer progression and metastatic disease. Specifically, in a heterogeneous disease such as ovarian cancer lacking defined targetable mutations, the identification of EMT-based subtypes has opened avenues to tailor precision personalized medicine. In particular, using the oncogenic RTK AXL as an example, we will highlight how this classification enables EMT-subtype specific identification of targets that could improve treatment options for patients and how there is a growing need for biophysical approaches to model dynamic processes such as EMT.
Article
Tannic acid, a hydrolysable tannin, exists commonly in food plants. Tannic acid has already been shown various anticancer mechanisms such as inhibiting the proliferation, inducing a higher apoptotic rate and slowing down the metastasis of different cancers. Moreover, tannic acid was reported to reduce the side effects caused by chemotherapeutics on patients. But whether the tannic acid can improve the treatment of oxaliplatin on colorectal carcinomatosis has yet been studied. In this study, we developed an injectable drug delivery system by physical incorporation of oxaliplatin (OXA) and tannic acid (TA) polymeric nanoparticles (OXA/TA NPs) into a thermo-sensitive hydrogel, OXA/TA NPs-hydrogel (OXA/TA NPs-H). The OXA/TA NPs-H was injected into the peritoneal cavity for the treatment of colorectal peritoneal carcinoma. Firstly, a water-in-oil-in-water double-emulsion (w/o/w) method and solvent-evaporation procedure were used in the preparation of the biodegradable OXA/TA NPs. Then, we prepared the biodegradable thermo-sensitive poly(3-caprolactone) (PCL)-10R5-PCL (PCLR) hydrogel with a low critical solution temperature (LCST) which undergoes gelation process at body temperature. Transmission electron microscopy (TEM) showed the spherical profile of OXA/TA NPs. Fourier-transform infrared (FTIR) spectra demonstrated that OXA and TA were both encapsulated into the OXA/TA NPs. In this study, intraperitoneal application of OXA/TA NPs-H restricted the growth of CT26 peritoneal colon cancer in vivo, improved the quality of life and prolonged the survival time of the model mice. Our study suggested that OXA/TA NPs-H might have potential application in the treatment of colorectal cancer.
Article
Epithelial–mesenchymal transition (EMT) is a cellular programme that is known to be crucial for embryogenesis, wound healing and malignant progression. During EMT, cell–cell and cell–extracellular matrix interactions are remodelled, which leads to the detachment of epithelial cells from each other and the underlying basement membrane, and a new transcriptional programme is activated to promote the mesenchymal fate. In the context of neoplasias, EMT confers on cancer cells increased tumour-initiating and metastatic potential and a greater resistance to elimination by several therapeutic regimens. In this Review, we discuss recent findings on the mechanisms and roles of EMT in normal and neoplastic tissues, and the cell-intrinsic signals that sustain expression of this programme. We also highlight how EMT gives rise to a variety of intermediate cell states between the epithelial and the mesenchymal state, which could function as cancer stem cells. In addition, we describe the contributions of the tumour microenvironment in inducing EMT and the effects of EMT on the immunobiology of carcinomas.