Omcl2021 9068850
Omcl2021 9068850
Omcl2021 9068850
Review Article
Roles of Therapeutic Bioactive Compounds in
Hepatocellular Carcinoma
Divya Jain,1 Yogesh Murti,2 Wasi Ullah Khan,3 Rajib Hossain,4 Mohammad Nabil Hossain,5
Krishn Kumar Agrawal,6 Rana Azeem Ashraf,7 Muhammad Torequl Islam,4
Pracheta Janmeda ,1 Yasaman Taheri ,8 Mohammed M. Alshehri ,9
Sevgi Durna Daştan,10,11 Balakyz Yeskaliyeva,12 Aliya Kipchakbayeva,12
Javad Sharifi-Rad ,8 and William C. Cho 13
1
Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan, India
2
Institute of Pharmaceutical Research, GLA University, Mathura, India
3
Key Laboratory for Sustainable Utilization of Tropical Bioresource, College of Tropical Crops Hainan University, Haikou, China
4
Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University,
Dhaka, Bangladesh
5
College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
6
Faculty of Pharmacy, R.B.S. Engineering Technical Campus, Bichpuri, Agra, India
7
School of Pharmaceutical Science and Technology (SPST), Tianjin University, China
8
Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
9
Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
10
Department of Biology, Faculty of Science, Sivas Cumhuriyet University, 58140 Sivas, Turkey
11
Beekeeping Development Application and Research Center, Sivas Cumhuriyet University, 58140 Sivas, Turkey
12
Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, 050040 Almaty, Kazakhstan
13
Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, SAR, China
Copyright © 2021 Divya Jain et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Hepatocellular carcinoma (HCC) is due to poor prognosis and lack of availability of effective treatment. Novel therapeutic
strategies will be the fine tuning of intracellular ROS signaling to effectively deprive cells of ROS-induced tumor-promoting
events. This review discusses the generation of ROS, the major signaling their modulation in therapeutics. We explore
some of the major pathways involved in HCC, which include the VEGF, MAPK/ERK, mTOR, FGF, and Ser/Thr kinase
pathways. In this review, we study cornerstone on natural bioactive compounds with their effect on hepatocarcinomas.
Furthermore, we focus on oxidative stress and FDA-approved signaling pathway inhibitors, along with chemotherapy and
radiotherapy enhancers which with early evidence of success. While more in vivo testing is required to confirm the
findings presented here, our findings will aid future nonclinical, preclinical, and clinical studies with these compounds, as
well as inspire medicinal chemistry scientists to conduct appropriate research on this promising natural compound and their
derivatives.
2 Oxidative Medicine and Cellular Longevity
Extracellular matrix
Vascular permeability
VEGF-2 Rho GTPase Cellular migration
Invasion
VEGF-2-P
PLC-𝛾 PKC
MAPK No
ANGIOGENESIS
Figure 1: VEGF signalling mechanism. VEGF: vascular endothelial growth factor; PLC-γ: phospholipase C gamma; PKC: protein kinase C;
MAPK: mitogen-activated protein kinase; NO: nitric oxide.
ing their catalytic cycle alters the redox reactions and disrupts
FGF monomer
H
P
S
the normal P450 catalytic cycle, which results in oxidative
stress leading to development of various kinds of disease.
FGFR
FGFR
FGFR
Cell membrane
FGFR
FGFR
GRB 2.8. Mitochondrial Dysfunction and Signaling. Mitochondria
FRS2
2 PLCΥ regulate the urea cycle, amino acid, iron, and fat metabolism
SO
S and produce energy required for the cell to perform all impor-
P P tant functions [80–82]. In cells, the major site for the produc-
RAS tion of ROS is mitochondrion [83]. Increased levels of ROS
production act as a clear death threat to the cells because it
PI3K
RAF directly affects the defense mechanism, the most exclusive
IP3-DAG
MAPK AKT autophagy, and plays their role as signaling molecules which
mTOR PKC-Ca+2 ultimately results in cell death either by autophagy or apopto-
ERK1/2 tic pathway (Figure 8). In each case, mobilization of various
Cell proliferation&differentiation H2O2 sensitive pathways is initiated [84]. Moreover, in starved
conditions, autophagy process increases due to elevated ROS
ANGIOGENESIS
production by mitochondria [85].
Figure 4: Fibroblast growth factor signaling pathway. GBR: growth Similar studies in obese (ob/ob) mice have also shown
factor receptor-bound; SOS: Son of Sevenless; PLCγ: phospholipase increased production of FFAs from glucose, elevated mt
C gamma; MAPK: mitogen-activated protein kinase; ERK1/2: ROS productions, and elevated levels of triglycerides [86],
extracellular signal-regulated kinase; PI3K: phospho-inositide-3- higher oxidative stress due to increased lipid peroxidation
kinase; AKT: protein kinase B; mTOR: mammalian target of while decreased hepatic mitochondrial components of
rapamycin; PKC-Ca2+: protein kinase C-Ca2+. MRC and decreased ATP levels [87]. All these mitochon-
drial changes require alterations in mitochondrial ROS
In the cytochrome chain, free radicals are formed when the levels, changes in mitophagy, biogenesis, and relevant signal-
electrons, donated by FADH and NADH, react with oxygen ing pathways of ROS.
and other electron acceptors [76] as shown in Figure 5. It also requires changes in cholesterol and GSH levels in
mitochondria. Changes in FFAs, lipid peroxidation prod-
2.7. Enzymatic Cycle of P450. Human cytochrome P450s ucts, and TNF are observed as well [88]. Oxidative stress
(CYP) are a superfamily of monooxygenases that are pri- causes ROS generation which results in the activation of cas-
marily known for the oxidation of the vast majority of xeno- cades involving PKCβ-dependent phosphorylation of
biotics in phase I metabolism helping in increasing substrate pp66shc and its movement to the matrix of mitochondria,
polarity and helping in excretion [77]. CYP generates ROS and these mitochondria are also the main target of ROS.
and how they contribute to an increase in oxidative stress.
In the very first step, the substrate (R-H) binds to the active 2.9. Serine/Threonine Kinase (AKT) Pathway. It is also known
site of the CYP enzyme via ferric iron (Fe3+) of the heme as protein kinase B (PKB) play an important role in angiogen-
thiolate group (Figure 6). In the second step, the heme thio- esis in pathological condition and tumor growth via different
late group receives one electron from the NADPH regener- Ser/Thr kinase family members like liver kinase B1 (LKB1),
ating system and CPR cytochrome peroxidase reductase calcium/calmodulin-dependent protein kinase IV (CAM-
(redox partner of CYP enzyme) and gets reduced to Fe+2. KIV), and sulfatase (SULF2). LKB1 has multiple phenotypic
This is the time when molecular oxygen binds to O2 and expressions for the regulation of cell polarity, metabolism,
CPR, then donates the second electron and reduces the Fe2 proliferation, and apoptosis. In HCC, LKB1 phosphorylate
+-
O2 complex which activates oxygen in the complex (Fe2 at Ser428 that phosphorylates AMP-activated protein kinase
+-
O2−). In the next step (H+), ions get into the active site (AMPK) microtubule affinity-regulating kinase (MARK)
by some special ion channels and cleave the O-O bond and phosphorylation leads to activation and localization of cofac-
release water. The complex (FeO2+3) then removes a proton tors like pseudokinase Ste20-related adaptor (STRADα) and
in the step from the substrate and leaves an intermediate the scaffolding protein MO25. The STRADα and MO25 form
RFe3+.OH−. In the last step, the –OH hydroxyl group is a complex that binds with the LKB1 and relocalize it from
transferred to the substrate radical and the oxidized sub- nucleus to cytoplasm and stimulate its cell proliferation and
strate is released at the end (Figure 7). The latest research angiogenesis [89]. Calcium (Ca+2) regulates various biologi-
is underway to specifically highlight the role of intermediate cal processes as a second messenger via a variety of signaling
species in various types of CYP-mediated oxidation reac- pathways. It binds to downstream effector calmodulin
tions; these intermediate species are formed during steps of (CAM) and increases the affinity toward calmodulin-kinase
the CYP 450 catalytic cycle [78]. Oxygen concentration like Ca+2±/CAM-protein kinase-IV (CAMKIV). CAMKIV
and pH are the two major factors that play an important role expression is increased in HCC and shows cell proliferation
in CYP-mediated coupling reactions [79]. and cell cycle regulation [90].
In this way, the CYP-mediated ROS-generated reaction Overexpression of calcium in HCC binds with the calmod-
through their catalytic cycle modifies the cellular compo- ulin that forms a complex with upregulated Ca+2/CAM-
nents which lead to various diseases. It is clear how CYP dur- dependent protein kinase kinase-2 (CAMKK2). Further, this
6 Oxidative Medicine and Cellular Longevity
NOs
ONOO– 2O2
Arginine NO- O2
xo Uric acid
Xanthine H2O
O2
0
P45
NADPH
NADP + O2
+
NADP + H
NADPH
O2 NADPH Oxidase
O2
H+ H+ H+
I III IV V
II
NADPH FAD O2
NADP + FADH2 H2O ADP = Pi
ONOO– SOD
H2O2 OH– ATP H+
O2
NO
Substrate
Human diseases:
(R)
–Cancer
Oxidized
substrate –Cardiovascular
(R-0H)
–Neuro degenrative
P450 reaction
cycle
Alcoholism effects:
–Oxidative damage
Figure 6: Cytochrome P450 contribution to human diseases caused by ROS and produced as a result of substrate metabolism by CYP 450s
which cause elevations in protein and nucleic acid levels and cause lipid modifications. These modified products further lead to lipid
peroxidation processes and also cause DNA damage which in turn causes cancer.
Oxidative Medicine and Cellular Longevity 7
1 2 3
RH ⁎
+RH 4
Fe+3 Fe+3RH +e– Fe+2RH
+O2
Fe+2–O2 +e
⁎
O2
9 RH
+2H+ +2
–ROH H2O2 Fe –O–O–
RH
+ H+
R. RH
+3 +3–OH +3 +2
+ H+ Fe 5
Fe Fe Fe –O –O–OH
ROH –H2O
8 7 6
Figure 7: Summarized CYP 450 catalytic cycle. Catalytic cycle of P450s showing some critical steps where ROS are generated (shown in
red). These ROS can further cause damage to cellular components that lead to various diseases.
P-p66
P-p66
Endoplasmic reticulum
Ca+2 Mitopathy
Bnip3
UIK1
Ca+2 PTP
Parkin
Fis1 (fusion)
Mitochondria
Table 1: Clinical trials and FDA approved molecules that exert inhibitory effect for each signaling pathway.
Clinical
Compounds/ trial/ Receptor/
Chemical structure Description Inhibitor References
drugs FDA target
approved
Cl
VEGFR1,
Vatalanib N NH VEGFR2, Small-molecule tyrosine VEGF
N N Phase
(PTK787/ZK VEGFR3, kinase receptor signaling [93]
-III
222584) PDGFR-β, inhibitor pathway
c-Kit
Vatalanib
VEGF–
VEGFR- VEGF
AE-941 Phase Shark-cartilage
Structure not available binding signaling [93, 94]
(Neovastat®) -III component
MMP2, pathway
MMP9
F
F
NH NH
F
O
O Cl
VEGFR-2, Small-molecule Raf VEGF
Phase
Sorafenib PDGFR-β, kinase and tyrosine signaling [93]
-III
FLT3, c-Kit kinase inhibitor pathway
NH
N C H3
Sorafenib
O N O
F
H3 C NH N NH
Allosteric, non-ATP
FDA MAPK
Trametinib BRAF competitive small- [95, 96]
O N approved pathway
H3 C C H3 I molecule inhibitors
O
Trametinib
H3 C O
N O
NH OH
N NH
BRAFV600E Allosteric, non-ATP
F F FDA MAPK
Binimetinib or competitive small- [96]
approved pathway
BRAFV600K molecule inhibitors
Br
Binimetinib
Inactivate Wnt
Wnt/β-
signaling by
Phase I- catenin
Genistein GSK3-β upregulating the [97]
II signalling
expression of GSK3-β
pathway
and E-cadherin
Oxidative Medicine and Cellular Longevity 9
Table 1: Continued.
Clinical
Compounds/ trial/ Receptor/
Chemical structure Description Inhibitor References
drugs FDA target
approved
HO O
OH O
OH
Genistein
OH
HO P O
O
O
Blocks the interaction
Wnt/β-
O
N between β-catenin and
catenin
PRI-724 Phase 1 β-Catenin its transcriptional [98, 99]
N N signalling
N CH3 coactivator CREB-
pathway
N CH3 binding protein (CBP)
O NH
PRI-724
N
HN
N
HN N Capable of inducing
N C H3 apoptosis and inhibit PI3K
USFDA
Idelalisib PI3K-δ AKT phosphorylation signaling [100, 101]
approved
N and downstream pathway
effectors
F O
Idelalisib
N
HN
N
HN N Capable of inducing
apoptosis and inhibit PI3K
USFDA PI3K-γ and
Duvelisib C H3 AKT phosphorylation signaling [101, 102]
approved PI3K-δ
N and downstream pathway
effectors
Cl O
Duvelisib
Table 1: Continued.
Clinical
Compounds/ trial/ Receptor/
Chemical structure Description Inhibitor References
drugs FDA target
approved
CH3
O O
CH3
CH3
CH3 N
N
N N
H3 C NH
Erdafitinib
O O N Serine/
threonine
H3C CH3 NH USFDA ROCK1/2 Inhibits the enzyme rho
Netarsudil kinase [104]
approved nonreceptor kinase
H2N (AKT)
pathway
Netarsudil
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
O 1. Liver biochemical
OH
parameters
Andrographolide (labdane Swiss albino In vivo (diethylnitrosamine) 2. Increased MDA and NO
1. [130]
diterpene) mice antioxidant assay O level
3. Decreased level of Gal-3
and IL-6
OH
OH
HO OH
OH
11
Table 2: Continued.
12
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
O N+
Berberine (benzyl- Hep3B, BEL- Cell counting kit-8 assay and EdU It suppressed the glutamine
5. O [134]
isoquinoline alkaloid) 7404 assay uptake by inhibiting SLC1A5
O
OH
1. Induction of apoptosis
2. Increased the Bax and
NOD/SCID
OH cleaved caspase-3
mice, HepG2, MTT assay, pulmonary metastasis
7. Betulinic acid (triterpene) 3. Decreased the level of Bcl-2 [136]
LM3, model
4. Decreased the level of ROS
MHCC97H
O 5. Inhibit metastasis via
MMP-2, MMP-9, and TIMP2
HO
1. Decreased expression of
SCLC (NCI- MTT assay, BrdU and PCNA O E2F-responsive proliferative
H69, NCI- proliferation assays, CAM assay, genes (cyclin E, thymidylate
Capsaicin (homovanillic
8. H82, DMS53, nude mice models, Chromatin synthase, cdc25A, and cdc6, [137]
acid alkaloid) O OH
DMS114), immunoprecipitation (ChIP) both at mRNA and protein
chicken eggs assay NH levels)
2. G1 phase arrest
Hoechst 33258 staining, MAPK 1. Inhibited the cell
9. Caffeine (purine alkaloid) [138]
activity, flow cytometry proliferation
Oxidative Medicine and Cellular Longevity
Table 2: Continued.
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
N 2. Activated the MERK-
regulating kinase and ERK
N pathway
3. Downregulation of EGRF
HepG2, HLF, N
Huh7, PLC/ O
PRF/5
N
OH
HO OH
Oxidative Medicine and Cellular Longevity
OH O O
HO OH
1. Antiproliferative
Male albino, HO O 2. Induced proapoptotic body
In vivo (DEN) induced hepato- OH
10. Crocin (apocarotenoid) Wistar rats, 3. Arresting the cell cycle at S [139]
carcinogenesis
HepG2 O HO OH and G2/M phases
OH
4. Induced apoptosis
O O OH
O O
O
O OH
HO
H
N N
N O O
S S
N
H
1. Induced apoptosis
O O 2. Upregulation of the Bax
Coumarin-6-sulfonamides Sulforhodamine B (SRB) method, 3. Downregulation of the Bcl-
11. HepG2 [140]
(sulfonamide derivative) annexin V–FITC apoptosis assay 2
H
N N 4. Increased caspase-3 levels
N O O
O 5. Arrest in the G2-M phase
S S
O N
H
HO O O
B16F10 cell
Carnosic acid MTT assay, BrdU incorporation 1. Arrested G0/G1 phase
12. xenograft [141]
(polyphenolicditerpene) assay 2. Enhances p21 expression
model
13
Table 2: Continued.
14
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH 3. Reduces the values of AST
and ALT
HO
C OOH
O O
1. Increased expression of
HO O prdx-3
2. Decreased ROS level
3. Upregulation of Bak protein
Daidzein (7- 4. Downregulation of Bcl-2
14. SK-HEP-1 TUNEL assay [143]
hydroxyisoflavones) and BclxL proteins
5. Increased the release of
O mitochondrial cytochrome c
OH 6. Activated the APAF-1,
caspase 9, and caspase 3
O
Male Swiss OH
1. Decreased incidence of
mice, Wistar preneoplastic foci
In vivo (N-nitrosodiethylamine,
15. Embelin (benzoquinone) albino rats, 2. Decreased biochemical [144]
CCl4)
Sprague HO
markers (SGOT, SGPT, ALP,
Dawley rats GGT, GST, and LPO)
O
1. Inhibited proliferation of
C57BL/6J
Esculetin (coumarin HCC cells
16. mice In vivo MTT assay [145]
derivative) 2. Arrest cell cycle at S phase
Hepa1-6 cells
3. Induced apoptosis
Oxidative Medicine and Cellular Longevity
Table 2: Continued.
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH 4.Increased caspase-3 and
caspase-9 activity
5. Increased Bax expression
6. Decreased Bcl-2 expression
O O OH
OH O OH
1. Attenuated cholesterol
HepG2 Western blotting, quantitative
synthesis and oncogenic AKT
Hep3B real-time PCR, tumor xenograft
signaling
17. Emodin (anthraquinone) Huh7 assay, Ki67 cell proliferation assay, [146]
2. Inactivated STAT3
Oxidative Medicine and Cellular Longevity
OH
OH
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
HO O
OH O
OH
HO
1. Decreased the size of
O tumors
Wistar albino HO
2. Decreased the levels of
21. Gallic acid (phenolic acid) In vivo (diethylnitrosamine) [150]
rats marker enzymes in serum
OH 3. Decreased the levels of
AgNORs and PCNA
HO
OH
H
O
18?-Glycyrrhetinic acid Decreased cell viability in
22. HepG2, H22 MTT assay [151]
(triterpene) higher concentration
HO
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH
HO OH
OH
HO
O O CH3
O O
HO CH3
O O
OH
Oxidative Medicine and Cellular Longevity
OH O
HO
1. Inhibited cell proliferation
2. Arrested G0/G1 cell cycle
CCA cell lines phase
HO
Honokiol (biphenol (KKU-100 3. Induced apoptosis
24. MTT assay [153]
neolignans) and KKU- 4. Suppressed the adhesion
213) and migration of cell
5. Inhibited the MMP-9 and
MMP-2 activity
OH
HO O
OH
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
(BALB/c-nu/ 2. Downregulated CD133
nu) expression
3. Sensitize (PTEN)-Akt-
ABCG2 pathway
H
H
HO
H
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH
HO O
OH O
OH
Oxidative Medicine and Cellular Longevity
OH
1. Suppressed expression of
activated AKT
Cell counting kit 8, flow
Oleuropein 2. Inhibited cell growth
30. HepG2, Huh7 cytometric analysis, cell viability O [159]
(monoterpenoid) O
3. Induced cell apoptosis
assay, luciferase assay
4. Inhibited PI3K/AKT
H
O O
signaling pathway
HO
O O
HO OH
OH O
HO
H
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
monodansylcadaverine (MDC), 2. Reduced expression of Bcl-2
AO staining 3. Increased expression of Bax,
H p53, and caspase-3 and 9
4. Induced autophagy
5. Inhibited the expression of
O the Ki-67 gene
O
H3C
1. Induced apoptosis
2. Regulated the NF-kB
H3C
H
signaling of NSCLC cells
NSCLC N H
N N 3. Significantly reduced the
(A549, NCI- O
Phycocyanin MTT assay, annexin V-FITC and O expression of MMP-2 and
33. H1299, NCI- N [162]
(phycobiliproteins) 7AAD staining H MMP-9
H460, and H3C CH3
CH3 4. Suppressed the proliferation
H3C
LTEP-A2)
of NSCLC cells
COOH COOH
5. Arrested G1 and S phase of
cell cycle
OH
OH O
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH
HO
HO
HO OH
O
O
HO
O
HO OH
O
OH O
HO
Oxidative Medicine and Cellular Longevity
OH
HO O
O
O 1. Decreased p65
phosphorylation
Rosmarinic acid (coumaric H22 tumor- ELISA, Western blotting, qRT-
36. 2. Inhibited the tumor growth [165]
acid derivative) bearing mice PCR HO
3. Decreased the elevated level
OH of cytokines
OH
OH
OH
1. Inhibited cell proliferation
2. Arrested G1 phase
MTT assay, flow cytometric 3. Downregulated the
37. Resveratrol (polyphenol) HepG2 analysis, Western blot analysis, expression of cyclin D1, p38 [166]
laser confocal microscopy MAP kinase, Akt, and Pak1
HO
4. Increased ERK activity
OH 5. Induced apoptosis
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH
OH
HO OH
OH
S. Animal/cell
Phytomolecules Methods Chemical structure Mechanisms Ref.
no. lines
OH
H
O O
H
O
H
Oxidative Medicine and Cellular Longevity
H H
O
OH
H
MTT assay, cell viability assay,
1. Reduced cell viability
flow cytometry, annexin-V/PI
HepG2, Bel- 2. Arrested S phase cell cycle
Xanthatin (sesquiterpene double staining assay, Western
41. 7402, SMMC- O O 3. Induced apoptosis [170]
lactone) blotting, immunofluorescence
7721 4. Induced ERS and activated
staining, dual-luciferase reporter O UPR pathway
gene assay
H
23
24 Oxidative Medicine and Cellular Longevity
phytochemicals has represented such as, andrographolide, Table 3: Medicinal plants and their bioactive compounds used in
berberine, capsaicin, curcumin, genistein, ursolic acid, and radiotherapy and chemotherapy.
withaferin A.
Andrographolide restrains tumor development by Plants as radioprotector/radiosensitizer in HCC
obstructing tumor adjustment to hypoxic conditions. The Decreases the depleted level of endogenic
Amaranthus
detected effects of andrographolide (100 mg/kg) were attrib- antioxidant enzymes during radiotherapy
paniculatus Linn.
of mice liver.
uted to the restriction of the hypoxia-inducible figure (HIF)
[176]. The recovery rate of different myeloma patients has Coronopus didymus Enhance the level of antioxidant enzymes
made strides through the use of andrographolide in a clinical (L.) in the liver of mice.
trial [177, 178]. While investigations are undertaken via the Augmented the SOD, CAT, and GSH
Grewia asiatica L.
large quantity of information on preclinical efficacy, clinical levels in the liver of irradiated mice.
studies are limited in the evaluation of berberines and It protected plasmid DNA and reduced
andrographolide genuine potential as a carcinoma opera- Glycyrrhiza glabra L. the liver microsomal LPO level in rat from
tor [179]. irradiation.
Capsaicin supplementation significantly reduced the In vitro and in vivo studies show the
Hypericum
establishment of preneoplastic foci in a rat model of hepato- increased level of SOD, CAT, GSH-Px,
perforatum L.
carcinogenesis caused by diethylnitrosamine, HCC cell lines and GSH during radiation therapy
were likewise stopped from proliferating, and apoptosis was Pilea microphylla (L.) Increased level of endogenous antioxidant
triggered on a dose-based manner [180, 181]. Furthermore, Liebm. enzyme levels in the liver of mice.
HCC cells were shown to be more sensitive than normal Augmented the SOD, CAT, and GSH
Rosmarinus officinalis
hepatocytes to capsaicin induced cytotoxicity suggesting that levels in blood and liver of mice during
L.
it might have a chemotherapeutic effect [182]. the radiation therapy.
Curcumin is a chemopreventive agent with a lot of opti- Xylopia aethiopica
Protect the liver of rat from γ-radiation
mism. This has prompted clinical practices to investigate the (Dunal) A.Rich.
pharmacokinetics and effectiveness of curcumin in patients. Bioactive phytomolecules as adjunct with chemotherapy
It was shown to be safe and nontoxic in phase I clinical stud- Used as an adjuvant with vinorelbine
ies, even at large dosages (8 g/day). However, it had limited Curcumin chemotherapy and enhances the
absorption individuals [183, 184]. Their clinical trials, either antiproliferative effect of drugs.
alone or as anticancer agent combinations, demonstrated Used as an adjunct in doxorubicin,
efficacy, despite challenges to bioavailability, in several dis- busulfan, and cisplatin chemotherapy. It
ease sites [185–187]. Quercetin also increased cytotoxic effects of these
The medication with genistein (140 mg/kg) is by pre- drugs and protect from drug-induced
venting aberrant nuclear β-catenin harvests and concealing nephrotoxicity.
WNT signaling features [188]. Ursolic acid (UA) was rep- Used as an adjunct with cisplatin and 5-
resented to upgrade the restorative impacts of oxaliplatin Ginsenosides FU chemotherapy and enhanced
in the mouse model of CRC by restraining the tumor antiproliferative effect.
and expanding the survival rate. Tumor shape is lessened
by the UA nanoparticles by focusing on caspases and
p53 with downregulation of Bcl-2 and cIAP, instigating 5.4. Modification of Genomic Stability. Phytochemicals tar-
apoptosis and driving to cervical cancer cell distortion gets both DNA repair and their damage mechanism, where
[189], whereas the tumor development of human colorec- genomic stability within cells plays an important role.
tal carcinoma (HCT-116) cells which overexpress AKT Besides, this genomic stability also helps a lot when chemo-
and microvessel arrangement is hindered through the ver- preventive agents trigger a selective number of cancer cells
bal organization of Withaferin A (5 mg/kg) in a mouse [194, 195]. There are some therapeutic agents which gener-
model [190]. ate the DNA repair pathways in normal cells to modulate
the stress conditions within the cells. In a contradicting
5.3. Detoxification of Enzymes. Xenobiotic compounds are way, DNA damage response can also be increased when can-
responsible for putting impacts on humans affecting tis- cer cells are exposed in a large number. As a result, apoptosis
sues. To lessen that impact, there are several responses can happen, and cells can be dead permanently [196].
or potentials [191]. Among them, the initialization of
some detoxifying enzymes is important, especially for the 5.5. Cancer Cell Metabolism. Cell metabolism in tumors
liver [192]. Detoxified enzymes can be induced by antiox- plays an important role in the stimulation process in proto-
idative phytochemicals found in plants. These phytochem- oncogenes by involving ROS production [197, 198]. The sur-
icals mainly target antioxidant response or electrophile vival and growth rate of tumor cells largely depend on their
response elements (ARE/EpRE) to modulate the molecular metabolic requirements which are adjusted by themselves
pathways. These pathways mainly depend on three main [199]. Energy requirements are supplied by glucose, and
components like ARE, nuclear factor erythroid 2 p45- thus, it initiates tumor growth. Besides glucose, glutamine
related factors 2 (Nrf2), and Kelchlike ECH-associated also plays an important role in tumor growth by providing
protein 1 (Keap1) [193]. nitrogen for the biosynthesis process. Phytochemicals can
Oxidative Medicine and Cellular Longevity 25
obstruct basal transport of glucose [200], e.g., curcumin can [4] F. X. Bosch, J. Ribes, M. Díaz, and R. Cléries, “Primary liver
convert glucose to glutathione [201]. cancer: worldwide incidence and trends,” Gastroenterology,
vol. 127, no. 5, pp. S5–S16, 2004.
5.6. Chemotherapy and Radiotherapy Enhancers. Cancer and [5] M. E. Norhaizan, S. K. Ng, M. S. Norashareena, and M. A.
complications that are associated with it are treated by che- Abdah, “Antioxidant and cytotoxicity effect of rice bran phytic
acid as an anticancer agent on ovarian, breast and liver cancer
motherapy and radiotherapy for decades. Radiotherapy
cell lines,” Malaysian journal of nutrition, vol. 17, no. 3, 2011.
began in the twentieth century before chemotherapy, as the
[6] G. S. Oh, H. O. Pae, H. Oh et al., “In vitro anti-proliferative
primary treatment of cancer. Approximately 8% of total can-
effect of 1, 2, 3, 4, 6-penta-O-galloyl-beta-D-glucose on
cer patients need radiotherapy, but unfortunately, it causes human hepatocellular carcinoma cell line, SK-HEP-1 cells,”
acute toxicity even at low doses of radiation. To overcome Cancer Letters, vol. 174, no. 1, pp. 17–24, 2001.
these side effects and prevention of resistance to chemother- [7] Y. L. Hsu, P. L. Kuo, C. Y. Cho et al., “Antrodiacinnamomea
apy, a promising new approach is developed by the scientist fruiting bodies extract suppresses the invasive potential of
by using the medicinal plant-derived drugs like taxol to human liver cancer cell line PLC/PRF/5 through inhibition
combat against cancer; many researches is going on radio- of nuclear factor κB pathway,” Food and Chemical Toxicol-
protectors and radiosensitizers [202]. Radioprotectors are ogy, vol. 45, no. 7, pp. 1249–1257, 2007.
the compounds that are used to protect the normal cell dur- [8] S. L. Yan, C. Y. Huang, S. T. Wu, and M. C. Yin, “Oleanolic
ing radiotherapy sessions; on the other hand, radiosensiti- acid and ursolic acid induce apoptosis in four human liver
zers are the molecule that is used to sensitize the tumor cancer cell lines,” Toxicology in Vitro, vol. 24, no. 3,
cell and increases the efficiency of cancer therapy. Some pp. 842–848, 2010.
plants like Pilea microphylla act as a radioprotector and pre- [9] A. Ito, H. Watanabe, and N. Basaran, “Effects of soy products
vent the depleting SOD, GSH, CAT, and thiol levels during in reducing risk of spontaneous and neutron-induced liver-
the radiotherapy in HCC. Table 3 shows the effect of medic- tumors in mice,” International Journal of Oncology, vol. 2,
inal plants and their bioactive compounds in chemotherapy no. 5, pp. 773–776, 1993.
and radiotherapy [203–206]. [10] X. G. Wu, D. H. Zhu, and X. Li, “Anticarcinogenic effect of
red ginseng on the development of liver cancer induced by
diethylnitrosamine in rats,” Journal of Korean Medical Sci-
6. Conclusion and Future Perspective ence, vol. 16, p. S61, 2001.
[11] M. Zabala, J. J. Lasarte, C. Perret et al., “Induction of immu-
The scavenging property of plant-derived bioactive com- nosuppressive molecules and regulatory T cells counteracts
pounds should be the pathway to treat HCC as they block the antitumor effect of interleukin-12-based gene therapy in
the propagation stage in oxidative chain reactions. Further- a transgenic mouse model of liver cancer,” Journal of Hepa-
more, these compounds inhibit cancer cell growth and act tology, vol. 47, no. 6, pp. 807–815, 2007.
as potent anticancer agents via different cellular and molec- [12] S. Demir, I. Turan, and Y. Aliyazicioglu, “Selective cytotoxic
ular mechanisms. At present, a plethora of kinase inhibitors effect of Rhododendron luteum extract on human colon
against specific molecular targets are being investigated in and liver cancer cells,” Journal of the Balkan Union of Oncol-
HCC, which initiate differential networks that consequently ogy, vol. 21, no. 4, pp. 883–888, 2016.
result in HCC cell cycle promotion. It offers hope that other [13] S. Grabley and R. Thiericke, Drug Discovery from Nature,
effective therapies will eventually be developed. Springer Science & Business Media, 1998.
[14] J. M. Pezzuto, “Plant-derived anticancer agents,” Biochemical
Pharmacology, vol. 53, no. 2, pp. 121–133, 1997.
Data Availability
[15] J. Uzoigwe and E. R. Sauter, “Cancer prevention and treat-
The data used to support the findings of this study are avail- ment using combination therapy with plant-and animal-
able from the corresponding author upon request. derived compounds,” Expert Review of Clinical Pharmacol-
ogy, vol. 5, no. 6, pp. 701–709, 2012.
[16] T. L. Simmons, E. Andrianasolo, K. McPhail, P. Flatt, and
Conflicts of Interest W. H. Gerwick, “Marine natural products as anticancer drugs,”
Molecular Cancer Therapeutics, vol. 4, no. 2, pp. 333–342, 2005.
There are no conflicts of interest to declare. [17] N. Sithranga Boopathy and K. Kathiresan, “Anticancer drugs
from marine flora: an overview,” Journal of oncology,
References vol. 2010, Article ID 214186, 18 pages, 2010.
[18] G. M. Cragg, P. G. Grothaus, and D. J. Newman, “Impact of
[1] M. Von Meyenfeldt, “Cancer-associated malnutrition: an natural products on developing new anti-cancer agents,”
introduction,” European Journal of Oncology Nursing, vol. 9, Chemical Reviews, vol. 109, no. 7, pp. 3012–3043, 2009.
pp. S35–S38, 2005. [19] V. Sharma and P. Janmeda, “Protective assessment of
[2] H. Van Zandwijk, G. Reid, and A. L. Frank, “Asbestos-related Euphorbia neriifolia and its isolated flavonoid against N-
cancers: the ‘Hidden Killer’ remains a global threat,” Expert nitrosodiethylamine-induced hepatic carcinogenesis in male
Review of Anticancer Therapy, vol. 20, no. 4, pp. 271–278, 2020. mice: a histopathological analysis,” Toxicology International,
[3] S. Darvesh, A. B. Aggarwal, and A. Bishayee, “Curcumin and vol. 21, no. 1, p. 37, 2014.
liver cancer: a review,” Current Pharmaceutical Biotechnol- [20] T. Choedon, S. K. Shukla, and V. Kumar, “Chemopreventive
ogy, vol. 13, no. 1, pp. 218–228, 2012. and anti-cancer properties of the aqueous extract of flowers
26 Oxidative Medicine and Cellular Longevity
of Butea monosperma,” Journal of Ethnopharmacology, [36] M. B. Thomas, R. Chadha, K. Glover et al., “Phase 2 study of
vol. 129, no. 2, pp. 208–213, 2010. erlotinib in patients with unresectable hepatocellular carci-
[21] J. S. Yoon, H. M. Kim, A. K. Yadunandam et al., “Neferine noma,” Cancer, vol. 110, no. 5, pp. 1059–1067, 2007.
isolated from Nelumbo nucifera enhances anti-cancer [37] M. J. Hawkins, “Clinical trials of antiangiogenic agents,” Cur-
activities in Hep3B cells: molecular mechanisms of cell rent Opinion in Oncology, vol. 7, no. 1, pp. 90–93, 1995.
cycle arrest, ER stress induced apoptosis and anti- [38] Y. C. Shen, C. Hsu, and A. L. Cheng, “Molecular targeted
angiogenic response,” Phytomedicine, vol. 20, no. 11, therapy for advanced hepatocellular carcinoma: current sta-
pp. 1013–1022, 2013. tus and future perspectives,” Journal of Gastroenterology,
[22] E. Brâkenhielm, R. Cao, and Y. Cao, “Suppression of angio- vol. 45, no. 8, pp. 794–807, 2010.
genesis, tumor growth, and wound healing by resveratrol, a [39] S. M. Wilhelm, C. Carter, L. Y. Tang et al., “BAY 43-9006
natural compound in red wine and grapes,” The FASEB Jour- exhibits broad spectrum oral antitumor activity and targets
nal, vol. 15, no. 10, pp. 1798–1800, 2001. the RAF/MEK/ERK pathway and receptor tyrosine kinases
[23] I. Ali and D. P. Braun, “Resveratrol enhances mitomycin C- involved in tumor progression and angiogenesis,” Cancer
mediated suppression of human colorectal cancer cell prolif- Research, vol. 64, no. 19, pp. 7099–7109, 2004.
eration by up-regulation of p21WAF1/CIP1,” Anticancer [40] E. K. Kim and E. J. Choi, “Pathological roles of MAPK signal-
Research, vol. 34, no. 10, pp. 5439–5446, 2014. ing pathways in human diseases,” Biochimica et Biophysica
[24] D. L. Aminin, E. S. Menchinskaya, E. A. Pisliagin, A. S. Sil- Acta (BBA)-Molecular Basis of Disease, vol. 1802, no. 4,
chenko, S. A. Avilov, and V. I. Kalinin, “Anticancer activity pp. 396–405, 2010.
of sea cucumber triterpene glycosides,” Marine Drugs, [41] J. A. Gollob, S. Wilhelm, C. Carter, and S. L. Kelley, “Role of
vol. 13, no. 3, pp. 1202–1223, 2015. Raf kinase in cancer: therapeutic potential of targeting the
[25] I. Yıldırım and T. Kutlu, “Anticancer agents: saponin and Raf/MEK/ERK signal transduction pathway,” Seminars in
tannin,” International Journal of Biological Chemistry, Oncology, vol. 33, no. 4, pp. 392–406, 2006.
vol. 9, pp. 332–340, 2015. [42] W. X. Schulze, L. Deng, and M. Mann, “Phosphotyrosine
[26] X. B. Sun, S. M. Wang, T. Li, and Y. Q. Yang, “Anticancer interactome of the ErbB-receptor kinase family,” Molecular
activity of linalool terpenoid: apoptosis induction and cell Systems Biology, vol. 1, no. 1, p. 2005, 2005.
cycle arrest in prostate cancer cells,” Tropical Journal of Phar- [43] N. Zarich, J. L. Oliva, N. Martínez et al., “Grb2 is a negative
maceutical Research, vol. 14, no. 4, pp. 619–625, 2015. modulator of the intrinsic Ras-GEF activity of hSos1,” Molec-
[27] D. M. Parkin, F. Bray, J. Ferlay, and P. Pisani, “Estimating the ular Biology of the Cell, vol. 17, no. 8, pp. 3591–3597, 2006.
world cancer burden: Globocan, 2000,” International Journal [44] J. Avruch, A. Khokhlatchev, J. M. Kyriakis et al., “Ras activa-
of Cancer, vol. 94, no. 2, pp. 153–156, 2001. tion of the Raf kinase: tyrosine kinase recruitment of the
[28] H. B. El-Serag and A. C. Mason, “Rising incidence of hepato- MAP kinase cascade,” Recent Progress in Hormone Research,
cellular carcinoma in the United States,” New England Jour- vol. 56, pp. 127–155, 2001.
nal of Medicine, vol. 340, no. 10, pp. 745–750, 1999. [45] C. Huang, K. Jacobson, and M. D. Schaller, “MAP kinases
[29] H. B. El-Serag, “Hepatocellular carcinoma: recent trends in and cell migration,” Journal of Cell Science, vol. 117, no. 20,
the United States,” Gastroenterology, vol. 127, no. 5, pp. 4619–4628, 2004.
pp. S27–S34, 2004. [46] K. Balmanno and S. J. Cook, “Tumour cell survival signalling
[30] A. O. Kaseb, A. Hanbali, M. Cotant, M. M. Hassan, by the ERK1/2 pathway,” Cell Death & Differentiation,
I. Wollner, and P. A. Philip, “Vascular endothelial growth vol. 16, no. 3, pp. 368–377, 2009.
factor in the management of hepatocellular carcinoma: a [47] K. J. Schmitz, J. Wohlschlaeger, H. Lang et al., “Activation of
review of literature,” Cancer: Interdisciplinary International the ERK and AKT signalling pathway predicts poor progno-
Journal of the American Cancer Society, vol. 115, no. 21, sis in hepatocellular carcinoma and ERK activation in cancer
pp. 4895–4906, 2009. tissue is associated with hepatitis C virus infection,” Journal
[31] R. Roskoski Jr., “Vascular endothelial growth factor (VEGF) of Hepatology, vol. 48, no. 1, pp. 83–90, 2008.
signaling in tumor progression,” Critical Reviews in Oncolo- [48] D. F. Calvisi, F. Pinna, F. Meloni et al., “Dual-specificity phos-
gy/Hematology, vol. 62, no. 3, pp. 179–213, 2007. phatase 1 ubiquitination in extracellular signal-regulated
[32] R. van der Meel, M. H. Symons, R. Kudernatsch et al., “The kinase–mediated control of growth in human hepatocellular
VEGF/Rho GTPase signalling pathway: a promising target carcinoma,” Cancer Research, vol. 68, no. 11, pp. 4192–
for anti-angiogenic/anti-invasion therapy,” Drug Discovery 4200, 2008.
Today, vol. 16, no. 5-6, pp. 219–228, 2011. [49] Y. Keshet and R. Seger, “The MAP kinase signaling cascades:
[33] R. Yamaguchi, H. Yano, A. Iemura, S. Ogasawara, a system of hundreds of components regulates a diverse array
M. Haramaki, and M. Kojiro, “Expression of vascular endo- of physiological functions,” in MAP Kinase Signaling Proto-
thelial growth factor in human hepatocellular carcinoma,” cols, Springer, 2010.
Hepatology, vol. 28, no. 1, pp. 68–77, 1998. [50] Q. Chang, Y. Zhang, K. J. Beezhold et al., “Sustained JNK1
[34] J. Zhou, Z. Y. Tang, J. Fan et al., “Expression of platelet- activation is associated with altered histone H3 methylations
derived endothelial cell growth factor and vascular endothe- in human liver cancer,” Journal of Hepatology, vol. 50, no. 2,
lial growth factor in hepatocellular carcinoma and portal vein pp. 323–333, 2009.
tumor thrombus,” Journal of Cancer Research and Clinical [51] M. Das, D. S. Garlick, D. L. Greiner, and R. J. Davis, “The role
Oncology, vol. 126, no. 1, pp. 57–61, 2000. of JNK in the development of hepatocellular carcinoma,”
[35] S. Whittaker, R. Marais, and A. X. Zhu, “The role of signaling Genes & Development, vol. 25, no. 6, pp. 634–645, 2011.
pathways in the development and treatment of hepatocellular [52] L. Hui, L. Bakiri, A. Mairhorfer et al., “p38α suppresses nor-
carcinoma,” Oncogene, vol. 29, no. 36, pp. 4989–5005, 2010. mal and cancer cell proliferation by antagonizing the JNK–
Oxidative Medicine and Cellular Longevity 27
c-Jun pathway,” Nature Genetics, vol. 39, no. 6, pp. 741–749, oxygenase, free radical-catalyzed mechanism,” Proceedings of
2007. the National Academy of Sciences, vol. 87, no. 23, pp. 9383–
[53] M. Soares-Silva, F. F. Diniz, G. N. Gomes, and D. Bahia, “The 9387, 1990.
mitogen-activated protein kinase (MAPK) pathway: role in [72] G. L. Milne, Q. Dai, and L. J. Roberts II, “The isoprostanes–25
immune evasion by trypanosomatids,” Frontiers in Microbi- years later,” Biochimica et Biophysica Acta (BBA)-Molecular
ology, vol. 7, p. 183, 2016. and Cell Biology of Lipids, vol. 1851, no. 4, pp. 433–445, 2015.
[54] P. W. Finch, X. He, M. J. Kelley et al., “Purification and [73] K. Linhart, H. Bartsch, and H. K. Seitz, “The role of reactive
molecular cloning of a secreted, Frizzled-related antagonist oxygen species (ROS) and cytochrome P-450 2E1 in the gen-
of Wnt action,” Proceedings of the National Academy of Sci- eration of carcinogenic etheno-DNA adducts,” Redox Biol-
ences, vol. 94, no. 13, pp. 6770–6775, 1997. ogy, vol. 3, pp. 56–62, 2014.
[55] P. Fedi, A. Bafico, A. N. Soria et al., “Isolation and biochem- [74] R. S. Balaban, S. Nemoto, and T. Finkel, “Mitochondria, oxi-
ical characterization of the human Dkk-1 homologue, a novel dants, and aging,” Cell, vol. 120, no. 4, pp. 483–495, 2005.
inhibitor of mammalian Wnt signaling,” Journal of Biological [75] F. J. Gonzalez, “Role of cytochromes P450 in chemical toxic-
Chemistry, vol. 274, no. 27, pp. 19465–19472, 1999. ity and oxidative stress: studies with CYP2E1,” Mutation
[56] H. Clevers, “Wnt/β-catenin signaling in development and Research/Fundamental and Molecular Mechanisms of Muta-
disease,” Cell, vol. 127, no. 3, pp. 469–480, 2006. genesis, vol. 569, no. 1-2, pp. 101–110, 2005.
[57] M. Shtutman, J. Zhurinsky, I. Simcha et al., “The cyclin D1 [76] K. A. Salman and S. Ashraf, “Reactive oxygen species: a link
gene is a target of the β-catenin/LEF-1 pathway,” Proceedings between chronic inflammation and cancer,” Asia-Pacific
of the National Academy of Sciences, vol. 96, no. 10, pp. 5522– Journal of Molecular Biology and Biotechnology., vol. 22,
5527, 1999. pp. 42–49, 2015.
[58] W. J. Nelson and R. Nusse, “Convergence of Wnt, ß-catenin, [77] F. P. Guengerich, “Common and uncommon cytochrome
and cadherin pathways,” Science, vol. 303, no. 5663, P450 reactions related to metabolism and chemical toxicity,”
pp. 1483–1487, 2004. Chemical Research in Toxicology, vol. 14, no. 6, pp. 611–650,
[59] J. Behari, “The Wnt/β-catenin signaling pathway in liver biol- 2001.
ogy and disease,” Expert Review of Gastroenterology & Hepa- [78] A. R. Modi and J. H. Dawson, “Oxidizing intermediates in
tology, vol. 4, no. 6, pp. 745–756, 2010. P450 catalysis: a case for multiple oxidants,” Monooxygenase,
[60] B. Vanhaesebroeck and M. D. Waterfield, “Signaling by dis- Peroxidase and Peroxygenase Properties and Mechanisms of
tinct classes of phosphoinositide 3-kinases,” Experimental Cytochrome, vol. P450, pp. 63–81, 2015.
Cell Research, vol. 253, no. 1, pp. 239–254, 1999. [79] A. Veith and B. Moorthy, “Role of cytochrome P450s in the
[61] A. Villanueva, D. Y. Chiang, P. Newell et al., “Pivotal role of generation and metabolism of reactive oxygen species,” Cur-
mTOR signaling in hepatocellular carcinoma,” Gastroenter- rent Opinion in Toxicology, vol. 7, pp. 44–51, 2018.
ology, vol. 135, no. 6, pp. 1972–1983, 2008. [80] D. Pessayre, A. Berson, B. Fromenty, and A. Mansouri,
[62] J. Paez and W. R. Sellers, “PI3K/PTEN/Akt pathway,” in Sig- “Mitochondria in steatohepatitis,” Seminars in Liver Disease,
nal Transduction in Cancer, Springer, 2004. vol. 21, no. 1, pp. 57–70, 2001.
[63] S. Wullschleger, R. Loewith, and M. N. Hall, “TOR signaling [81] M. Vacca, M. Allison, J. L. Griffin, and A. Vidal-Puig, “Fatty
in growth and metabolism,” Cell, vol. 124, no. 3, pp. 471–484, acid and glucose sensors in hepatic lipid metabolism: impli-
2006. cations in NAFLD,” Seminars in Liver Disease, vol. 35,
[64] A. Beenken and M. Mohammadi, “The FGF family: biology, no. 3, pp. 250–261, 2015.
pathophysiology and therapy,” Nature Reviews Drug Discov- [82] A. M. Gusdon, K. X. Song, and S. Qu, “Nonalcoholic fatty
ery, vol. 8, no. 3, pp. 235–253, 2009. liver disease: pathogenesis and therapeutics from a
[65] N. Turner and R. Grose, “Fibroblast growth factor signalling: mitochondria-centric perspective,” Oxidative Medicine and
from development to cancer,” Nature Reviews Cancer, vol. 10, Cellular Longevity, vol. 2014, Article ID 637027, 20 pages,
no. 2, pp. 116–129, 2010. 2014.
[66] D. M. Ornitz, J. Xu, J. S. Colvin et al., “Receptor specificity of [83] G. Tell, C. Vascotto, and C. Tiribelli, “Alterations in the redox
the fibroblast growth factor family,” Journal of Biological state and liver damage: hints from the EASL Basic School of
Chemistry, vol. 271, no. 25, pp. 15292–15297, 1996. Hepatology,” Journal of Hepatology, vol. 58, no. 2, pp. 365–
[67] V. P. Eswarakumar, I. Lax, and J. Schlessinger, “Cellular sig- 374, 2013.
naling by fibroblast growth factor receptors,” Cytokine & [84] T. Finkel, “Signal transduction by mitochondrial oxidants,”
Growth Factor Reviews, vol. 16, no. 2, pp. 139–149, 2005. Journal of Biological Chemistry, vol. 287, no. 7, pp. 4434–
[68] N. Zheng, W. Wei, and Z. Wang, “Emerging roles of FGF sig- 4440, 2012.
naling in hepatocellular carcinoma,” Translational Cancer [85] R. Scherz-Shouval, E. Shvets, E. Fass, H. Shorer, L. Gil, and
Research, vol. 5, no. 1, pp. 1–6, 2016. Z. Elazar, “Reactive oxygen species are essential for autoph-
[69] K. M. Holmström and T. Finkel, “Cellular mechanisms and agy and specifically regulate the activity of Atg4,” The EMBO
physiological consequences of redox-dependent signalling,” Journal, vol. 26, no. 7, pp. 1749–1760, 2007.
Nature Reviews Molecular Cell Biology, vol. 15, no. 6, [86] K. Begriche, A. Igoudjil, D. Pessayre, and B. Fromenty, “Mito-
pp. 411–421, 2014. chondrial dysfunction in NASH: causes, consequences and
[70] H. Sies, C. Berndt, and D. P. Jones, “Oxidative stress,” Annual possible means to prevent it,” Mitochondrion, vol. 6, no. 1,
Review of Biochemistry, vol. 86, pp. 715–748, 2017. pp. 1–28, 2006.
[71] J. D. Morrow, K. E. Hill, R. F. Burk, T. M. Nammour, K. F. [87] K. Begriche, J. Massart, and B. Fromenty, “Effects of β-ami-
Badr, and L. J. Roberts, “A series of prostaglandin F2-like noisobutyric acid on leptin production and lipid homeostasis:
compounds are produced in vivo in humans by a non-cyclo- mechanisms and possible relevance for the prevention of
28 Oxidative Medicine and Cellular Longevity
obesity,” Fundamental & Clinical Pharmacology, vol. 24, [102] J. R. Brown, “Phosphatidylinositol 3 kinase δ inhibitors: pres-
no. 3, pp. 269–282, 2010. ent and future,” Cancer Journal, vol. 25, no. 6, pp. 394–400,
[88] H. Z. Lin, S. Q. Yang, C. Chuckaree, F. Kuhajda, G. Ronnet, 2019.
and A. M. Diehl, “Metformin reverses fatty liver disease in [103] R. Roskoski Jr., “Properties of FDA-approved small molecule
obese, leptin-deficient mice,” Nature Medicine, vol. 6, no. 9, protein kinase inhibitors: a 2020 update,” Pharmacological
pp. 998–1003, 2000. Research, vol. 152, article 104609, 2020.
[89] T. C. Delgado, F. L. Otsoa, and M. L. Martinez-Chantar, [104] R. Roskoski, “Properties of FDA-approved small molecule
“Post-translational modifiers of liver kinase B1/Serine/threo- protein kinase inhibitors: a 2021 update,” Pharmacological
nine kinase II in hepatocellular carcinoma,” Journal of Hepa- Research, vol. 165, article 105463, 2021.
tocellular Carcinoma, vol. 6, pp. 85–91, 2019. [105] B. N. Ames, “Micronutrients prevent cancer and delay
[90] J. Li, C. Zhang, H. Jiang, and J. Cheng, “Andrographolide aging,” Toxicology Letters, vol. 102, pp. 5–18, 1998.
inhibits hypoxia-inducible factor-1 through phos- [106] D. A. Cox and M. L. Cohen, “Effects of oxidized low-density
phatidylinositol 3-kinase/AKT pathway and suppresses lipoprotein on vascular contraction and relaxation: clinical
breast cancer growth,” OncoTargets and Therapy, vol. 8, and pharmacological implications in atherosclerosis,” Phar-
pp. 427–435, 2015. macological Reviews, vol. 48, no. 1, pp. 3–19, 1996.
[91] H. Naz, M. Tarique, S. Ahmad et al., “Hesperidin-CAMKIV [107] T. Finkel and N. J. Holbrook, “Oxidants, oxidative stress and
interaction and its impact on cell proliferation and apoptosis the biology of ageing,” Nature, vol. 408, no. 6809, pp. 239–
in the human hepatic carcinoma and neuroblastoma cells,” 247, 2000.
Journal of Cellular Biochemistry, vol. 120, no. 9, pp. 15119– [108] V. Sharma, P. Janmeda, R. Paliwal, and S. Sharma, “Antihe-
15130, 2019. patotoxic activity of Euphorbia neriifolia extract against N-
[92] R. M. Carr, P. A. R. Duran, E. J. Tolosa et al., “The extracellu- nitrosodiethylamine-induced hepatocarcinogenesis in mice,”
lar sulfatase SULF2 promotes liver tumorigenesis by stimulat- Zhong xi yijie he xue bao= Journal of Chinese integrative med-
ing assembly of a promoter looping GLI1-STAT3 icine, vol. 10, no. 11, pp. 1303–1309, 2012.
transcriptional complex,” Journal of Biological Chemistry, [109] S. Sharma, “Chemoprotective activity of hydro-ethanolic
vol. 295, no. 9, pp. 2698–2712, 2020. extract of Euphorbia neriifolia Linn leaves against DENA-
[93] R. K. Jain, D. G. Duda, J. W. Clark, and J. S. Loeffler, “Lessons induced liver carcinogenesis in mice,” Biology and Medicine,
from phase III clinical trials on anti-VEGF therapy for can- vol. 3, pp. 36–44, 2011.
cer,” Nature Clinical Practice Oncology, vol. 3, no. 1, pp. 24– [110] S. V. Pracheta, R. Paliwal, and S. Sharma, “In vitro free radical
40, 2006. scavenging and antioxidant potential of ethanolic extract of
[94] S. Y. Lee and S. M. Chung, “Neovastat (Æ-941) inhibits the Euphorbia neriifolia Linn,” International Journal of Pharmacy
airway inflammation via VEGF and HIF-2α suppression,” and Pharmaceutical Sciences, vol. 3, no. 1, pp. 238–242, 2011.
Vascular Pharmacology, vol. 47, no. 5-6, pp. 313–318, 2007. [111] R. Paliwal, V. Sharma, and S. S. Pracheta, “Hepatoprotective
[95] K. T. Flaherty, J. R. Infante, A. Daud et al., “Combined BRAF and antioxidant potential of Moringa oleifera pods against
and MEK inhibition in melanoma with BRAF V600 muta- DMBA-induced hepatocarcinogenesis in male mice,” Inter-
tions,” New England Journal of Medicine, vol. 367, no. 18, national Journal of Drug Development & Research, vol. 3,
pp. 1694–1703, 2012. no. 2, pp. 128–138, 2011.
[96] J. Wei, J. Hu, L. Wang et al., “Discovery of a first-in-class [112] D. D. Kitts, A. N. Wijewickreme, and C. Hu, “Antioxidant
mitogen-activated protein kinase kinase 1/2 degrader,” Jour- properties of a North American ginseng extract,” Molecular
nal of Medicinal Chemistry, vol. 62, no. 23, pp. 10897– and Cellular Biochemistry, vol. 203, no. 1, pp. 1–10, 2000.
10911, 2019. [113] K. G. Lee and T. Shibamoto, “Antioxidant properties of
[97] A. Sebio, M. Kahn, and H. J. Lenz, “The potential of targeting aroma compounds isolated from soybeans and mung beans,”
Wnt/β-catenin in colon cancer,” Expert Opinion on Thera- Journal of Agricultural and Food Chemistry, vol. 48, no. 9,
peutic Targets, vol. 18, no. 6, pp. 611–615, 2014. pp. 4290–4293, 2000.
[98] K. H. Emami, C. Nguyen, H. Ma et al., “A small molecule [114] H. Jiao and S. Y. Wang, “Correlation of antioxidant capacities
inhibitor of β-catenin/cyclic AMP response element- to oxygen radical scavenging enzyme activities in blackberry,”
binding protein transcription,” Proceedings of the National Journal of Agricultural and Food Chemistry, vol. 48, no. 11,
Academy of Sciences, vol. 101, no. 34, pp. 12682–12687, pp. 5672–5676, 2000.
2004. [115] S. V. Pracheta, R. Paliwal, and S. Sharma, “Preliminary phy-
[99] J. Harb, P. J. Lin, and J. Hao, “Recent development of Wnt tochemical screening and in vitro antioxidant potential of
signaling pathway inhibitors for cancer therapeutics,” Cur- hydro-ethanolic extract of Euphorbia neriifolia Linn,” Inter-
rent Oncology Reports, vol. 21, no. 2, p. 12, 2019. national Journal of PharmTech Research, vol. 3, no. 1,
[100] S. E. Herman, A. L. Gordon, A. J. Wagner et al., “Phos- pp. 124–132, 2011.
phatidylinositol 3-kinase-δ inhibitor CAL-101 shows prom- [116] A. Prakash, D. Jain, and R. Tripathi, “Pharmacognostical
ising preclinical activity in chronic lymphocytic leukemia by analysis of different parts of Cyperus rotundus L.,” Plant Sci-
antagonizing intrinsic and extrinsic cellular survival signals,” ence Today, vol. 6, no. sp1, pp. 607–612, 2019.
Blood, vol. 116, no. 12, pp. 2078–2088, 2010. [117] D. Jain, P. Chaudhary, A. Kotnala, R. Hossain, K. Bisht, and
[101] D. A. Sabbah, R. Hajjo, S. K. Bardaweel, and H. A. Zhong, M. N. Hossain, “Hepatoprotective activity of medicinal
“Phosphatidylinositol 3-kinase (PI3K) inhibitors: a recent plants: a mini review,” Journal of Medicinal Plants, vol. 8,
update on inhibitor design and clinical trials (2016-2020),” no. 5, pp. 183–188, 2020.
Expert Opinion on Therapeutic Patents, vol. 31, no. 10, [118] D. Jain, N. Uniyal, D. Mitra, and P. Janmeda, “Traditional
pp. 1–16, 2021. resources and use of aromatic and ethnomedicinal plants in
Oxidative Medicine and Cellular Longevity 29
Uttarakhand: compliment of nature,” International Journal [134] P. Zhang, Q. Wang, Z. Lin, P. Yang, K. Dou, and R. Zhang,
of Herbal Medicine, vol. 8, no. 5, pp. 88–95, 2020. “Berberine inhibits growth of liver cancer cells by suppressing
[119] K. S. B. Razzak, D. Jain, M. N. Hossain, and A. Bushra, “Plant glutamine uptake,” OncoTargets and Therapy, vol. 12,
edible vaccines: a natural way of vaccination,” Vigyan Varta, p. 11751, 2019.
vol. 1, no. 5, pp. 21–24, 2020. [135] N. Subramaniam, P. Kannan, and D. Thiruvengadam,
[120] S. P. Hussain, L. J. Hofseth, and C. C. Harris, “Radical causes “Hepatoprotective effect of boldine against
of cancer,” Nature Reviews Cancer, vol. 3, no. 4, pp. 276–285, diethylnitrosamine-induced hepatocarcinogenesis in Wistar
2003. rats,” Journal of Biochemical and Molecular Toxicology,
vol. 33, no. 12, article e22404, 2019.
[121] R. Olinski, A. Siomek, R. Rozalski et al., “Oxidative damage to
DNA and antioxidant status in aging and age-related dis- [136] W. Wang, Y. Wang, M. Liu et al., “Betulinic acid induces apo-
eases,” Acta Biochimica Polonica, vol. 54, no. 1, pp. 11–26, ptosis and suppresses metastasis in hepatocellular carcinoma
2007. cell lines in vitro and in vivo,” Journal of Cellular and Molec-
ular Medicine, vol. 23, no. 1, pp. 586–595, 2019.
[122] P. Abraham, V. K. Kolli, and S. Rabi, “Melatonin attenuates
methotrexate-induced oxidative stress and renal damage in [137] K. C. Brown, T. R. Witte, W. E. Hardman et al., “Capsaicin
rats,” Cell Biochemistry and Function, vol. 28, no. 5, displays anti-proliferative activity against human small cell
pp. 426–433, 2010. lung cancer in cell culture and nude mice models via the
E2F pathway,” PLoS One, vol. 5, no. 4, article e10243, 2010.
[123] S. Rashid, N. Ali, S. Nafees, S. K. Hasan, and S. Sultana, “Mit-
igation of 5-fluorouracil induced renal toxicity by chrysin via [138] J. I. Okano, T. Nagahara, K. Matsumoto, and Y. Murawaki,
targeting oxidative stress and apoptosis in Wistar rats,” Food “Caffeine inhibits the proliferation of liver cancer cells and
and Chemical Toxicology, vol. 66, pp. 185–193, 2014. activates the MEK/ERK/EGFR signalling pathway,” Basic &
Clinical Pharmacology & Toxicology, vol. 102, no. 6,
[124] D. N. Tripathi and G. B. Jena, “Intervention of astaxanthin pp. 543–551, 2008.
against cyclophosphamide-induced oxidative stress and
DNA damage: a study in mice,” Chemico-Biological Interac- [139] A. Amin, A. Hamza, S. Daoud et al., “Saffron-based crocin
tions, vol. 180, no. 3, pp. 398–406, 2009. prevents early lesions of liver cancer: in vivo, in vitro and net-
work analyses,” Recent Patents on Anti-Cancer Drug Discov-
[125] S. Nafees, S. Rashid, N. Ali, S. K. Hasan, and S. Sultana, ery, vol. 11, no. 1, pp. 121–133, 2016.
“Rutin ameliorates cyclophosphamide induced oxidative
[140] A. Sabt, O. M. Abdelhafez, R. S. El-Haggar et al., “Novel
stress and inflammation in Wistar rats: role of NFκB/MAPK
coumarin-6-sulfonamides as apoptotic anti-proliferative
pathway,” Chemicobiological Interactions, vol. 231, pp. 98–
agents: synthesis, in vitro biological evaluation, and QSAR
107, 2015.
studies,” Journal of Enzyme Inhibition and Medicinal Chem-
[126] S. Li, H. Y. Tan, N. Wang et al., “The role of oxidative stress istry, vol. 33, no. 1, pp. 1095–1107, 2018.
and antioxidants in liver diseases,” International Journal of
[141] K. I. Lin, C. C. Lin, S. M. Kuo et al., “Carnosic acid impedes
Molecular Sciences, vol. 16, no. 11, pp. 26087–26124, 2015.
cell growth and enhances anticancer effects of carmustine
[127] V. Sharma, S. Sharma, and S. S. Pracheta, “Lead induced hep- and lomustine in melanoma,” Bioscience Reports, vol. 38,
atotoxicity in male Swiss albino mice: the protective potential no. 4, 2018.
of the hydromethanolic extract of Withania somnifera,”
[142] J. Wang, C. Wang, and G. Bu, “Curcumin inhibits the growth
International Journal of Pharmaceutical Sciences and
of liver cancer stem cells through the phosphatidylinositol 3-
Research, vol. 7, pp. 116–121, 2011.
kinase/protein kinase B/mammalian target of rapamycin sig-
[128] B. Mayer and B. Hemmens, “Biosynthesis and action of nitric naling pathway,” Experimental and Therapeutic Medicine,
oxide in mammalian cells,” Trends in Biochemical Sciences, vol. 15, no. 4, pp. 3650–3658, 2018.
vol. 22, no. 12, pp. 477–481, 1997. [143] H. J. Park, Y. K. Jeon, D. H. You, and M. J. Nam, “Daidzein
[129] D. Jain, P. Chaudhary, N. Varshney, and P. Janmeda, “Carci- causes cytochrome c-mediated apoptosis via the Bcl-2 family
nogenic effects of N-nitroso compounds in the environment,” in human hepatic cancer cells,” Food and Chemical Toxicol-
Environment Conservation Journal, vol. 21, no. 3, pp. 25–41, ogy, vol. 60, pp. 542–549, 2013.
2020. [144] R. Poojari, S. Gupta, G. Maru, B. Khade, and S. Bhagwat,
[130] F. A. Yassen, A. T. Keshta, and N. A. Ghonaim, “Androgra- “Chemopreventive and hepatoprotective effects of embelin
pholide and resveratrol effect against hepatocellular carci- on N-nitrosodiethylamine and carbon tetrachloride induced
noma induced in male albino rats,” Biochemistry Letters, preneoplasia and toxicity in rat liver,” Asian Pacific Journal
vol. 11, no. 1, pp. 89–101, 2015. of Cancer Prevention, vol. 11, no. 4, pp. 1015–1020, 2010.
[131] X. Zou, J. Liang, J. Sun et al., “Allicin sensitizes hepatocellular [145] J. Wang, M. L. Lu, H. L. Dai, S. P. Zhang, H. X. Wang, and
cancer cells to anti-tumor activity of 5-fluorouracil through N. Wei, “Esculetin, a coumarin derivative, exerts in vitro
ROS-mediated mitochondrial pathway,” Journal of Pharma- and in vivo antiproliferative activity against hepatocellular
cological Sciences, vol. 131, no. 4, pp. 233–240, 2016. carcinoma by initiating a mitochondrial-dependent apopto-
[132] X. Dong, J. Fu, X. Yin et al., “Induction of apoptosis in sis pathway,” Brazilian Journal of Medical and Biological
HepaRG cell line by aloe-emodin through generation of reac- Research, vol. 48, no. 3, pp. 245–253, 2015.
tive oxygen species and the mitochondrial pathway,” Cellular [146] Y. S. Kim, Y. M. Lee, T. I. Oh et al., “Emodin sensitizes hepa-
Physiology and Biochemistry, vol. 42, no. 2, pp. 685–696, tocellular carcinoma cells to the anti-cancer effect of sorafe-
2017. nib through suppression of cholesterol metabolism,”
[133] S. Nadi, M. Elahi, S. Moradi, and A. Banaei, “Radioprotective International Journal of Molecular Sciences, vol. 19, no. 10,
effect of arbutin in megavoltage therapeutic X-irradiated p. 3127, 2018.
mice using liver enzymes assessment,” Journal of Biomedical [147] H. Khan, W. Jia, Z. Yu et al., “Emodin succinyl ester inhibits
Physics & Engineering, vol. 9, no. 5, p. 533, 2019. malignant proliferation and migration of hepatocellular
30 Oxidative Medicine and Cellular Longevity
carcinoma by suppressing the interaction of AR and EZH2,” [161] J. Sun, C. Zhang, Y. L. Bao et al., “Parthenolide-induced apo-
Biomedicine & Pharmacotherapy, vol. 128, article 110244, ptosis, autophagy and suppression of proliferation in HepG2
2020. cells,” Asian Pacific Journal of Cancer Prevention, vol. 15,
[148] J. Chen, L. Chen, T. Lu et al., “ERα36 is an effective target of no. 12, pp. 4897–4902, 2014.
epigallocatechin-3-gallate in hepatocellular carcinoma,” [162] S. Hao, Y. Yan, S. Li et al., “The in vitro anti-tumor activity of
International Journal of Clinical and Experimental Pathology, phycocyanin against non-small cell lung cancer cells,”
vol. 12, no. 9, p. 3222, 2019. Marine Drugs, vol. 16, no. 6, p. 178, 2018.
[149] M. Sanaei, F. Kavoosi, A. Valiani, and M. A. Ghobadifar, [163] L. Wu, J. Li, T. Liu et al., “Quercetin shows anti-tumor effect
“Effect of genistein on apoptosis and proliferation of hepato- in hepatocellular carcinoma LM3 cells by abrogating JAK2/-
cellular carcinoma Hepa1-6 cell line,” International Journal STAT3 signaling pathway,” Cancer Medicine, vol. 8, no. 10,
of Preventive Medicine, vol. 9, 2018. pp. 4806–4820, 2019.
[150] S. Jagan, G. Ramakrishnan, P. Anandakumar, S. Kamaraj, [164] M. Alía, R. Mateos, S. Ramos, E. Lecumberri, L. Bravo, and
and T. Devaki, “Antiproliferative potential of gallic acid L. Goya, “Influence of quercetin and rutin on growth and
against diethylnitrosamine-induced rat hepatocellular carci- antioxidant defense system of a human hepatoma cell line
noma,” Molecular and Cellular Biochemistry, vol. 319, no. 1, (HepG2),” European Journal of Nutrition, vol. 45, no. 1,
pp. 51–59, 2008. pp. 19–28, 2006.
[151] P. Kuang, W. Zhao, W. Su et al., “18β-glycyrrhetinic acid [165] W. Cao, C. Hu, L. Wu, L. Xu, and W. Jiang, “Rosmarinic acid
inhibits hepatocellular carcinoma development by reversing inhibits inflammation and angiogenesis of hepatocellular car-
hepatic stellate cell-mediated immunosuppression in mice,” cinoma by suppression of NF-κB signaling in H22 tumor-
International Journal of Cancer, vol. 132, no. 8, pp. 1831– bearing mice,” Journal of pharmacological sciences, vol. 132,
1841, 2013. no. 2, pp. 131–137, 2016.
[152] S. Yumnam, H. S. Park, M. K. Kim et al., “Hesperidin induces [166] P. Parekh, L. Motiwale, N. Naik, and K. V. K. Rao, “Downreg-
paraptosis like cell death in hepatoblatoma, HepG2 cells: ulation of cyclin D1 is associated with decreased levels of p38
involvement of ERK1/2 MAPK,” PloS One, vol. 9, no. 6, arti- MAP kinases, Akt/PKB and Pak1 during chemopreventive
cle e101321, 2014. effects of resveratrol in liver cancer cells,” Experimental and
[153] W. Songjang and A. Jiraviriyakul, “Honokiol and magnolol Toxicologic Pathology, vol. 63, no. 1-2, pp. 167–173, 2011.
inhibit growth, metastasis and induce apoptosis in human
[167] X. Hu, S. Lin, D. Yu, S. Qiu, X. Zhang, and R. Mei, “A prelim-
cholangiocarcinoma,” Biomedical and Pharmacology Journal,
inary study: the anti-proliferation effect of salidroside on dif-
vol. 12, no. 2, pp. 759–773, 2019.
ferent human cancer cell lines,” Cell biology and Toxicology,
[154] G. Zhu, X. Liu, H. Li, Y. Yan, X. Hong, and Z. Lin, “Kaemp- vol. 26, no. 6, pp. 499–507, 2010.
ferol inhibits proliferation, migration, and invasion of liver
[168] H. S. Son, H. Y. Kwon, E. J. Sohn et al., “Activation of AMP-
cancer HepG2 cells by down-regulation of microRNA-21,”
activated protein kinase and phosphorylation of glycogen
International Journal of Immunopathology and Pharmacol-
synthase kinase 3 β mediate ursolic acid induced apoptosis
ogy, vol. 32, 2018.
in HepG2 liver cancer cells,” Phytotherapy Research, vol. 27,
[155] T. K. W. Lee, A. Castilho, V. C. H. Cheung, K. H. Tang, S. Ma, no. 11, pp. 1714–1722, 2013.
and I. O. L. Ng, “Lupeol targets liver tumor-initiating cells
through phosphatase and tensin homolog modulation,” [169] Y. X. Wang, W. B. Ding, and C. W. Dong, “Withaferin A sup-
Hepatology, vol. 53, no. 1, pp. 160–170, 2011. presses liver tumor growth in a nude mouse model by down-
regulation of cell signaling pathway leading to invasion and
[156] Y. D. Wang, X. J. Sun, W. J. Yang, J. Li, and J. J. Yin, “Magno-
angiogenesis,” Tropical Journal of Pharmaceutical Research,
lol exerts anticancer activity in hepatocellular carcinoma cells
vol. 14, no. 6, pp. 1005–1011, 2015.
through regulating endoplasmic reticulum stress-mediated
apoptotic signaling,” OncoTargets and Therapy, vol. 11, [170] T. L. Shi, L. Zhang, Q. Y. Cheng et al., “Xanthatin induces
p. 5219, 2018. apoptosis by activating endoplasmic reticulum stress in hep-
atoma cells,” European Journal of Pharmacology, vol. 843,
[157] P. B. Pal, K. Sinha, and P. C. Sil, “Mangiferin, a natural xan-
pp. 1–11, 2019.
thone, protects murine liver in Pb (II) induced hepatic damage
and cell death via MAP kinase, NF-κB and mitochondria depen- [171] P. Arulselvan, M. T. Fard, W. S. Tan et al., “Role of antioxi-
dent pathways,” PloS One, vol. 8, no. 2, article e56894, 2013. dants and natural products in inflammation,” Oxidative med-
[158] D. Arul and P. Subramanian, “Naringenin (citrus flavonone) icine and cellular longevity, vol. 2016, Article ID 5276130, 15
induces growth inhibition, cell cycle arrest and apoptosis in pages, 2016.
human hepatocellular carcinoma cells,” Pathology & Oncol- [172] I. Sakaida, M. Tsuchiya, K. Kawaguchi, T. Kimura, S. Terai,
ogy Research, vol. 19, no. 4, pp. 763–770, 2013. and K. Okita, “HerbalmedicineInchin-ko-to (TJ-135) pre-
[159] C. M. Yan, E. Q. Chai, H. Y. Cai, G. Y. Miao, and W. Ma, vents liver fibrosis and enzyme-altered lesions in rat liver cir-
“Oleuropein induces apoptosis via activation of caspases and rhosis induced by a choline-deficient L-amino acid-defined
suppression of phosphatidylinositol 3-kinase/protein kinase B diet,” Journal of Hepatology, vol. 38, no. 6, pp. 762–769, 2003.
pathway in HepG2 human hepatoma cell line,” Molecular [173] H. J. Lin, J. Y. Chen, C. F. Lin et al., “Hepatoprotective effects
Medicine Reports, vol. 11, no. 6, pp. 4617–4624, 2015. of Yi Guan Jian, an herbal medicine, in rats with
[160] Y. Y. Zhu, H. Y. Huang, and Y. L. Wu, “Anticancer and apo- dimethylnitrosamine-induced liver fibrosis,” Journal of Eth-
ptotic activities of oleanolic acid are mediated through cell nopharmacology, vol. 134, no. 3, pp. 953–960, 2011.
cycle arrest and disruption of mitochondrial membrane [174] X. Lin, S. Zhang, Q. Huang et al., “Protective effect of Fufang-
potential in HepG2 human hepatocellular carcinoma cells,” Liu-Yue-Qing, a traditional Chinese herbal formula, on CCl4
Molecular Medicine Reports, vol. 12, no. 4, pp. 5012–5018, induced liver fibrosis in rats,” Journal of Ethnopharmacology,
2015. vol. 142, no. 2, pp. 548–556, 2012.
Oxidative Medicine and Cellular Longevity 31
[175] J. Lv, Z. Zhao, Y. Chen et al., “The Chinese herbal decoction tumor growth in colorectal cancer cells,” Oncotarget, vol. 7,
danggui buxue tang inhibits angiogenesis in a rat model of no. 12, pp. 13854–13864, 2016.
liver fibrosis,” Evidence-Based Complementary and Alterna- [191] D. Mansuy, “Metabolism of xenobiotics: beneficial and
tive Medicine, vol. 2012, Article ID 284963, 11 pages, 2012. adverse effects,” Biologieaujourd'hui, vol. 207, no. 1, pp. 33–
[176] F. Lin, K. L. Marcelo, K. Rajapakshe et al., “The CaMKK2/- 37, 2013.
CaMKIV is an essential regulator of hepatic cancer,” Hepatol- [192] P. Jancova, P. Anzenbacher, and E. Anzenbacherova, “Phase
ogy, vol. 62, no. 2, pp. 505–520, 2015. II drug metabolizing enzymes,” Biomedical Papers, vol. 154,
[177] H. Gao and J. Wang, “Andrographolide inhibits multiple no. 2, pp. 103–116, 2010.
myeloma cells by inhibiting the TLR4/NF-kappa B signaling [193] V. Kaur, M. Kumar, A. Kumar, K. Kaur, V. S. Dhillon, and
pathway,” Molecular Medicine Reports, vol. 13, pp. 1827– S. Kaur, “Pharmacotherapeutic potential of phytochemicals:
1832, 2016. implications in cancer chemoprevention and future perspec-
[178] E. J. Gunn, J. T. Williams, D. T. Huynh et al., “The natural tives,” Biomedicine & Pharmacotherapy, vol. 97, pp. 564–586,
products parthenolide and andrographolide exhibit anti- 2018.
cancer stem cell activity in multiple myeloma,” Leukemia & [194] A. Azqueta and A. Collins, “Polyphenols and DNA damage: a
Lymphoma, vol. 52, pp. 1085–1097, 2011. mixed blessing,” Nutrients, vol. 8, no. 12, p. 785, 2016.
[179] Y. Zhang, X. Li, D. Zou et al., “Treatment of type 2 diabetes [195] M. R. Kelly, J. Xu, K. E. Alexander, and G. Loo, “Disparate
and dyslipidemia with the natural plant alkaloid berberine,” effects of similar phenolic phytochemicals as inhibitors of
The Journal of Clinical Endocrinology & Metabolism, oxidative damage to cellular DNA,” Mutation Research/DNA
vol. 93, no. 7, pp. 2559–2565, 2008. Repair, vol. 485, no. 4, pp. 309–318, 2001.
[180] J. J. Jang, K. J. Cho, Y. S. Lee, and J. H. Bae, “Different mod-
[196] P. Rajendran, E. Ho, D. E. Williams, and R. H. Dashwood,
ifying responses of capsaicin in a widespectrum initiation
“Dietary phytochemicals, HDAC inhibition, and DNA dama-
model of F344 rat,” Journal of Korean Medical Science,
ge/repair defects in cancer cells,” Clinical Epigenetics, vol. 3,
vol. 6, pp. 31–36, 1991.
no. 1, pp. 1–23, 2011.
[181] J. A. Kim, Y. S. Kang, and Y. S. Lee, “A phospholipase C-
[197] S. Reuter, S. C. Gupta, M. M. Chaturvedi, and B. B. Aggarwal,
dependent intracellular Ca2+ release pathway mediates the
“Oxidative stress, inflammation, and cancer: how are they
capsaicin-induced apoptosis in HepG2 human hepatoma
linked?,” Free Radical Biology and Medicine, vol. 49, no. 11,
cells,” Archives of Pharmacal Research, vol. 28, pp. 73–80, 2005.
pp. 1603–1616, 2010.
[182] G. Galati and P. J. O’Brien, “Cytoprotective and anticancer
[198] G. Waris and H. Ahsan, “Reactive oxygen species: role in the
properties of coenzyme Q vs. capsaicin,” Biofactors, vol. 18,
development of cancer and various chronic conditions,” Jour-
pp. 195–205, 2003.
nal of Carcinogenesis, vol. 5, p. 14, 2006.
[183] R. A. Sharma, S. A. Euden, S. L. Platton et al., “Phase I clinical
[199] R. J. DeBerardinis, “Is cancer a disease of abnormal cellular
trial of oral curcumin: biomarkers of systemic activity and
metabolism? New angles on an old idea,” Genetics in Medi-
compliance,” Clinical Cancer Research, vol. 10, no. 20,
cine, vol. 10, no. 11, pp. 767–777, 2008.
pp. 6847–6854, 2004.
[184] M. Kanai, Y. Otsuka, K. Otsuka et al., “A phase I study inves- [200] W. Zhang, Y. Liu, X. Chen, and S. C. Bergmeier, “Novel
tigating the safety and pharmacokinetics of highly bioavailable inhibitors of basal glucose transport as potential anticancer
curcumin (Theracurmin) in cancer patients,” Cancer Chemo- agents,” Bioorganic & Medicinal Chemistry Letters, vol. 20,
therapy and Pharmacology, vol. 71, no. 6, pp. 1521–1530, 2013. no. 7, pp. 2191–2194, 2010.
[185] M. Bayet-Robert, F. Kwiatkowski, M. Leheurteur et al., [201] M. Bayet-Robert and D. Morvan, “Metabolomics reveals met-
“Phase I dose escalation trial of docetaxel plus curcumin in abolic targets and biphasic responses in breast cancer cells
patients with advanced and metastatic breast cancer,” Cancer treated by curcumin alone and in association with docetaxel,”
Biology & Therapy, vol. 9, no. 1, pp. 8–14, 2010. PloS One, vol. 8, no. 3, article e57971, 2013.
[186] H. Mahammedi, E. Planchat, M. Pouget et al., “The new com- [202] H. Zaid, M. Silbermann, A. Amash, D. Gincel, E. Abdel-Sat-
bination docetaxel, prednisone and curcumin in patients with tar, and N. B. Sarikahya, “Medicinal plants and natural active
castration-resistant prostate cancer: a pilot phase II study,” compounds for cancer chemoprevention/chemotherapy,”
Oncology, vol. 90, no. 2, pp. 69–78, 2016. Evidence-Based Complementary and Alternative Medicine,
vol. 2017, Article ID 7952417, 2 pages, 2017.
[187] R. Epelbaum, M. Schaffer, B. Vizel, V. Badmaev, and G. Bar-
Sela, “Curcumin and gemcitabine in patients with advanced [203] B. Hazra, S. Ghosh, A. Kumar, and B. N. Pandey, “The pro-
pancreatic cancer,” Nutrition and Cancer, vol. 62, no. 8, spective role of plant products in radiotherapy of cancer: a
pp. 1137–1141, 2010. current overview,” Frontiers in Pharmacology, vol. 2, no. 94,
pp. 1–13, 2012.
[188] Y. Zhang, Q. Li, D. Zhou, and H. Chen, “Genistein, a soya iso-
flavone, prevents azoxymethane-induced up-regulation of [204] V. Kuruba and P. Gollapalli, “Natural radioprotectors and
WNT/beta-catenin signalling and reduces colon pre- their impact on cancer drug discovery,” Radiation Oncology
neoplasia in rats,” British Journal of Nutrition, vol. 109, Journal, vol. 36, no. 4, pp. 265–275, 2018.
no. 1, pp. 33–42, 2013. [205] C. P. Wu, S. Ohnuma, and S. V. Ambudkar, “Discovering
[189] Y. Wang, Z. Wei, and X. Wang, “Enhancement of anti-tumor natural product modulators to overcome multidrug resis-
activity of ursolic acid by nanostructured lipid carriers,” tance in cancer chemotherapy,” Current Pharmaceutical Bio-
Nanomedicine: Nanotechnology, Biology and Medicine, technology, vol. 12, no. 4, pp. 609–620, 2011.
vol. 14, pp. 1743–1885, 2018. [206] K. Sak, “Chemotherapy and dietary phytochemical agents,”
[190] S. Suman, T. P. Das, S. Sirimulla, H. Alatassi, M. K. Ankem, Chemotherapy Research and Practice, vol. 2012, Article ID
and C. Damodaran, “Withaferin-A suppress AKT induced 282570, 11 pages, 2012.