ArticlePDF AvailableLiterature Review

Preventive and therapeutic use of herbal compounds against doxorubicin induced hepatotoxicity: a comprehensive review

Authors:

Abstract and Figures

Doxorubicin (DOX) is associated with numerous acute and chronic dose-related toxicities including hepatotoxicity. This adverse reaction may limit the use of other chemotherapeutic agents with hepatic excretion, and so, its prevention is an important issue. The aim of this study was to conduct a comprehensive review of in vitro, in vivo and human studies regarding the protective effects of synthetic and naturally-occurring compounds against DOX-induced liver injury. The search was conducted in Embase, PubMed, and Scopus databases using the following keywords: “doxorubicin,” “Adriamycin,” “hepatotoxicity,” “liver injury,” “liver damage,” and “hepatoprotective,” and all articles published in English were included without time restriction. Forty eligible studies to the end of May 2022 finally were reviewed. Our results demonstrated that all of these drugs, except acetylsalicylic acid, had considerable hepatoprotective effects against DOX. In addition, none of the studied compounds attenuated the antitumor efficacy of DOX treatment. Silymairn was the only compound which is assessed in human studies and showed promising preventive and therapeutic effects. Altogether, our results demonstrated that most of compounds with antioxidant, anti-apoptosis, and anti-inflammatory properties are efficacious against DOX-induced hepatotoxicity and may be considered as a potential adjuvant agent for prevention of hepatotoxicity in cancer patients, after fully been assessed in well-designed large clinical trials.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Naunyn-Schmiedeberg's Archives of Pharmacology
https://doi.org/10.1007/s00210-023-02429-1
REVIEW
Preventive andtherapeutic use ofherbal compounds
againstdoxorubicin induced hepatotoxicity: acomprehensive review
FaezehMahmoudi1· OmidArasteh1· SepidehElyasi1
Received: 19 August 2022 / Accepted: 16 February 2023
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2023
Abstract
Doxorubicin (DOX) is associated with numerous acute and chronic dose-related toxicities including hepatotoxicity. This
adverse reaction may limit the use of other chemotherapeutic agents with hepatic excretion, and so, its prevention is an
important issue. The aim of this study was to conduct a comprehensive review of invitro, invivo and human studies regard-
ing the protective effects of synthetic and naturally-occurring compounds against DOX-induced liver injury. The search
was conducted in Embase, PubMed, and Scopus databases using the following keywords: “doxorubicin,” “Adriamycin,
“hepatotoxicity,” “liver injury,” “liver damage,” and “hepatoprotective,” and all articles published in English were included
without time restriction. Forty eligible studies to the end of May 2022 finally were reviewed. Our results demonstrated that
all of these drugs, except acetylsalicylic acid, had considerable hepatoprotective effects against DOX. In addition, none of
the studied compounds attenuated the antitumor efficacy of DOX treatment. Silymairn was the only compound which is
assessed in human studies and showed promising preventive and therapeutic effects. Altogether, our results demonstrated that
most of compounds with antioxidant, anti-apoptosis, and anti-inflammatory properties are efficacious against DOX-induced
hepatotoxicity and may be considered as a potential adjuvant agent for prevention of hepatotoxicity in cancer patients, after
fully been assessed in well-designed large clinical trials.
Keywords Doxorubicin· Adriamycin· Hepatotoxicity· Herbal compounds· Liver injury· Hepatoprotective
Abbreviations
DOX Doxorubicin
ROS Reactive oxygen species
NADPH Nicotinamide adenine dinucleo-
tide phosphate
AST Aspartate aminotransaminase
ALT Alanine aminotransaminase
SD rats Sprague-Dawley rats
PI3K Phosphoinositide3-kinase
TNBC cells Triple negative breast cancer cells
G10 10-Gingerol
Cdk-6 Cyclin-dependent kinase 6
GGT γ Glutamyl transferase
NM Nutrient mixture
RCD Regular chow diet
VCO Virgin coconut oil
ATP Adenosine triphosphate
LDH Lactate dehydrogenase
CkMB Creatine kinase myocardial band
DLA Daltone’s lymphoma ascites
GAE Ganoderma applantum Extract
LDL Low density lipoprotein
HDL High density lipoprotein
ALP Alkaline phosphatase
TG Triglycerides
PRV Pravastatin
LNE Lipid nanoemulsion
EAC-challenged mice Mice which injected with tumor
cells
TBARS Thiobarbituric acid reactive
substances
GSH Glutathione
CAT Catalase
SOD Superoxide dismutase
LD50 Median lethal dose
PARP Poly-ADP-ribose polymerase
ECG Electrocardiogram
* Sepideh Elyasi
elyasis@mums.ac.ir
1 Department ofClinical Pharmacy, School ofPharmacy,
Mashhad University ofMedical Sciences, P.O.
Box91775-1365, Mashhad, Iran
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
CGA Chlorgenic acid
FAC Fluorouracil/doxorubicin/
cyclophosphamide
TE Transient elastography
RCT Randomized clinical trial
AC-T Doxorubicin/
cyclophosphamide-paclitaxel
AKBA Acetyl 11-keto-b-boswellic acid
Nrf2 NF-E2-related factor 2
BOS Boswellic acid
HO-1 Oxygenase-1
Sirt1 Silent information regulator 1
FOXO1 Forkhead box protein O1
Keap1 Kelch-like ECH-associated
protein-1
PCNA Proliferating cell nuclear antigen
GPx Glutathione peroxidase
GR Glutathione reductase
GST Glutathione-S-transferase
EC50 Half maximal effective
concentration
Cr Creatine
FN-1 Fibronectin
NIN Naringin
LA DL-alpha lipoic acid
GSSE Grape seed and skin extract
MDA Malondialdehyde
G6PD Glucose-6-phosphate
dehydrogenase
ATS Artemisinin
ATX Astaxanthin
AA Asiatic acid
ALP Alkaline phosphatase
LPO Lipid peroxidation
ZM Zataria multiflora
AHE Acacia hydaspica
POD Peroxidase
QR Quinone reductase
pCA P-Coumaric acid
ALB Albumin
ZJ Ziziphus jujuba
HSP70 Heat shock protein 70
IGF Insulin-like growth factor
IGFBP-3 Insulin-like growth factor binding
protein
ETC Electron transport chain
Introduction
Cancer is one of the most common human diseases with the
incidence rate of over 440 per 100,000 men and women,
and a total number of 19.3 million people are diagnosed
annually. This devastating disease is the second leading
cause of human mortality after cardiovascular disease,
accounting for up to 10 million deaths in 2020, worldwide
(Ferlay etal. 2021). Chemotherapeutic agents generally tar-
get the uncontrolled growth and proliferation of cancer cells
(Sak 2012).
Doxorubicin (DOX) is a chemotherapeutic agent from the
anthracyclines family, which is first extracted from Strep-
tomyces peucetius (Thorn etal. 2011). DOX acts against
cancer cells through multiple mechanisms, including (I)
intercalation into the DNA double helix and interference
with topoisomerase II-mediated DNA repair and (II) genera-
tion of free radicals which can damage cell membrane, DNA
molecules, and proteins. The latter mechanism is mediated
through oxidization of DOX to an unstable metabolite, semi-
quinone, which is then converted back to DOX in a process
that generate reactive oxygen species (ROS) (Thorn etal.
2011). Currently, DOX is routinely used for the treatment
of a wide range of human malignancies, such as acute leu-
kemia and lymphomas, carcinomas, sarcomas, breast, blad-
der, ovarian, gastric, bone, and also some pediatric cancers
(Kalyanaraman 2020). However, high doses and prolonged
use of this drug for cancer treatment are associated with
several side effects and post-treatment debacles, e.g., hepa-
totoxicity and cardiotoxicity, resulting in chemotherapy dis-
continuation or dose reduction (Shivakumar etal. 2012).
An increasing body of evidence demonstrates that differ-
ent synthetic drugs and naturally-occurring flavonoids may
be effective against DOX-induced hepatotoxicity. The ame-
liorative effects of such compounds include, but not limited
to the reduction of the inflammatory cytokines and oxida-
tive stress markers, down-regulation of pro-apoptotic pro-
teins and up-regulation of antiapoptotic proteins. These can
lead to reduction of liver enzymes serum level and hepatic
collagen fibers deposition scores (Superfin etal. 2007).
However, in spite of the extensive studies on the protective
effects of these compounds against DOX, the full potentials
and benefits and also the detailed mechanisms remain to
be elucidated. The objective of this study was to conduct a
review of published preclinical and clinical studies regarding
the protective effects of various herbal compounds against
DOX-induced liver injuries and to define the cellular and
molecular mechanisms underlying this hepatoprotection.
Materials andmethods
In May 2022, we explored scientific databases including
Embase, PubMed, and Scopus from the inception until now
with the following keywords: “doxorubicin,” “adriamycin,”
“hepatotoxicity,” “liver injury,” “liver damage,” and “hepato-
protective.” We considered no restriction on the availability
of the full text, if enough data could be accessed from the
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
abstract. The Boolean operators and nesting were as follows:
(TS = (Doxorubicin) OR TS = (adriamycin)) AND (TS = (hepa-
totoxicity) OR TS = (“liver injury”) OR TS = (“liver damage”)
OR TS = (“liver toxicity”) OR TS = (hepatoprotective) OR
TS = (“liver dysfunction’) OR TS = (“drug-induced hepatitis”)
OR TS = (“fatty liver disease”) OR TS = (“liver failure”)). We
chose the related articles based on their title and abstract. Our
inclusion criteria were papers written in English that assessed
the protective effect of various herbal compounds against DOX-
induced hepatotoxicity. All invitro, invivo, and clinical studies
which were accordant with our criteria were included. Dupli-
cated or unrelated articles, or articles published in languages
other than English, were excluded, if they did not have English
abstract. Data collection was carried out between May 5, 2021
and April 15, 2022. The search process and initial selection
of eligible studies were done by the first author. Finally, in the
result section, we gathered and compared the effectiveness in
ameliorating DOX-induced hepatotoxicity.
Results
In present paper, 885 articles were found by searching
abovementioned databases. Among these articles, 647,
151, 23, 3, and 21 articles were omitted because of being
non-relevant, duplication, review articles, papers in other
languages, and using non-herbal compounds, respectively.
Finally, 40 studies were eligible for this review (Fig.2). The
included studies are summarized in Tables1 and 2. Thirty
compounds are assessed as hepatoprotective against DOX
and related studies are reviewed in the following sections.
Mechanisms ofdoxurobicin‑induced liver injury
A great deal of available studies on doxorubicin adverse
effects is focused on its cardiotoxicity. However, the admin-
istration of DOX is also frequently associated with toxic
injury to the liver and may intensify the rate of transient
increase in serum liver enzyme and acute liver injury with
jaundice that can be severe and even life-threatening (Dos
Santos Arruda etal. 2019). The production of ROS is the
major consequence of its redox cycling profile and is con-
sidered as a double-edged sword, which acts not only on
cancer cells but also on several normal body cells. Various
enzymes, including xanthine oxidases, NADPH oxidases,
nitric oxide synthases, and peroxidases, which are located
in several subcellular compartments, e.g., mitochondria,
endoplasmic reticulum, and cytoplasm, account as major
sources of ROS. These alterations may result in various
histopathological changes as well as induction of apoptosis
(Moher etal. 2009). The probable mechanisms of hepato-
toxicity are summarized in Fig.1. Furthermore, pharma-
cokinetics studies of DOX in patients with abnormal liver
biochemistry tests have discovered that patients with reduced
DOX clearance not only have an increased bilirubin level,
but also elevated levels of liver enzymes, such as aspartate
aminotransaminase (AST) and alanine aminotransaminase
(ALT) (Twelves etal. 1998). Approaches to doxorubicin
dosing in patients with impaired liver function are variable
now. The United States Prescribing Information for doxo-
rubicin and pegylated liposomal doxorubicin recommend a
50 percent dose reduction for bilirubin 1.2 to 3mg/dL, and
a 75 percent dose reduction for bilirubin 3.1 to 5mg/dL.
Others suggest omission of the drug for bilirubin > 5mg/dL
(Superfin etal. 2007; Sahlan etal 2021) (Fig.2).
Hepatoprotective effect ofherbal compounds
Citronellal
Citronellal is a well-known metabolite of Cymbopogon spp.
plants and is demonstrated to exhibit many beneficial prop-
erties such as insect repellent or antimicrobial activities (Wei
& Wee 2013). Given the various potentials of this drug in
health issues, Liu etal. investigated the effect of citronellal
(200mg/kg b.w./day orally) on DOX-induced hepatotoxic-
ity (2.5mg/kg b.w./week, intraperitoneally) in SD rats (Liu
etal. 2021). It attenuated DOX-induced pathological liver
tissue changes and improved liver function biomarkers.
In particular, citronellal administration resulted in signifi-
cant decrease in the levels of AST, ALT, glutamic pyruvic
transaminase, and albumin in serum and modulated the lev-
els of malondialdehyde, superoxide dismutase, and reduced
glutathione in liver. In addition, citronellal diminished the
Bax/Bcl-2 ratio and caspase-3 expression, thereby inhibiting
cell apoptosis. This drug also enhanced the levels of PI3K
signaling members and CD31 in the liver, further highlight-
ing the role of citronellal in the protection of liver against
DOX. So, it seems that anti-apoptosis and antioxidant activ-
ity is the most important proposed mechanism of action for
citronellal.
[10]‑Gingerol andginger extract
10-Gingerol is a member of gingerols, which are the major
pungent constituents in ginger (Zingiber officinale), and
comprises a series of homologues varying in the length
of their alkyl chains. 10-Gingerol is believed to inhibit the
anti-serotonin 3 receptor function leading to anti-emetic
properties (Semwal etal. 2015). In a study conducted by
Bapitsa etal. on 4T1Br4 murine TNBC cells bearing Balb/c
mice, concomitant use of 10-Gingerol (10mg/kg, five times
a week) and DOX (3mg/kg, twice a week) can increase
the levels of active caspase-3 and γH2AX and to decrease
the level of Cdk-6 cyclin (Baptista Moreno Martin etal.
2020). Likewise, combined regimen led to decreased tumor
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 Summarized hepatoprotective natural-occuring compounds against doxorubicin hepatotoxicity; preclinical studies
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Liu etal. 2021 CTN In vivo 40 SD rats
4 groups:
CTRL: NS p.o
CTN: 200mg/kg/day p.o
DOX: 2.5mg/kg/week i.p
CT + DOX
6weeks
Sig liver histopathological changes
Sig serum ALB and sig serum levels of AST and ALT
Sig MDA levels and sig the hepatic levels of GSH and
SOD
Sig anti-apoptosis effect ( Bax/Bcl2 pr and caspase-3 pr
through activating PI3k/Akt signaling pathway)
Sig pro-angiogenesis effect ( CD-31 pr) in liver tissue
Baptista Moreno
Martin etal. 2020
G10 In vivo 60 4T1Br4 murine TNBC cells bearing Balb/c mice
4 groups:
CTRL: NS
G10: 10mg/kg 5times a week
DOX: 3mg/kg twice a week
G10 + DOX
28days
Sig BW in G10 + DOX group
Sig of survival rate in DOX group compared to others
Prevention of DOX-induced AST and ALT serum levels
Histological effect: liver inflammatory and liver damages
with G10 treatment
Sakr etal. 2007 GIN In vivo 60 albino rats
4 groups:
CTRL
DOX: 2mg/kg/week i.p
GIN: 24mg p.o. 3 times a week
DOX + GIN
6weeks
Sig in BW in DOX + GIN compared to DOX group
Sig in DOX-induced ALT, AST and MDA elevations
Sig in DOX-induced SOD reduction
Sig anti-inflammatory effects in treatment group
Ahmed etal. 2013 GIN In vivo 40 Albino Wistar rats
4 groups:
CTRL: NS p.o
GIN: 250mg/kg/day p.o
DOX: 2.5mg/kg for a total cumulative dose of 15mg/kg
DOX + GIN
14days
Sig offset of DOX-induced weight loss with GIN treatment
Sig DOX-induced severe liver damages, degeneration
and necrosis score, and hepatic collagen fibers deposi-
tion scores
Kocahan etal. 2017 QE In vivo 36 Wistar rats
6 groups: CTRL, QE, CPY, DOX, CPY + QE, DOX + QE
1.8mg/kg DOX i.p. once every 3weeks
10mg/kg QE i.p. daily
10weeks
Sig MDA levels in DOX + QE group
No sig changes in GSH levels
No sig differences in SOD levels
No sig changes in CAT levels
Sig GPx levels in the DOX group compared with
QE + DOX
Wang etal. 2012 QE In vivo Acute hepatotoxicity study on 24 C57BL/6 mice
4 groups:
CTRL: NS
DOX: 20mg/kg i.p. on day 5
QE: 100mg/kg/day p.o. for 4 consecutive days
DOX + QE
10days
Subchronic hepatic damages study on nude mice with SMMC7721
tumor xenografts: 4mg/kg/week DOX i.p. for 3weeks
Co-administration of DOX and QE: partial reversed of
DOX-induced of serum ALT and AST levels
Sig in liver histological damages in co-administration of
QE with DOX
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Ahmed etal. 2019 CSI and
EPI
In vivo 40 Wistar rats
4 groups:
CTRL: NS i.p. weekly and 1% CMC solution p.o. QOD
DOX: 4mg/kg i.p. weekly and 1% CMC solution p.o. QOD
DOX + CSI: DOX + 200mg/kg CSI p.o. QOD and 1% CMC solution
DOX + EPI: DOX + 25mg/5ml 1% CMC/kg EPI QOD
6weeks
CSI: sig in ALT, AST, ALP, GGT, AFP and total BL.
Non-sig serum ALB
EPI: sig in ALT, AST, ALP and AFP. Non-sig serum
ALB. Non-sig in total BL. Sig serum GGT activity
CSI: non-sig in SOD activity and sig LPO
EPI: sig in SOD activity and sig LPO
Sig in hepatic GSH and GPx activity with both CSI and
EPI. Non-sig effect on GST
Sig in p53 mRNA expression, Bcl2 mRNA expression
and liver NF-κB mRNA expression with both CSI and
EPI
Sig amelioration of DOX-induced liver histological injuri-
ous alterations with both CSI and EPI treatments
Feeble expression of COX-2 and caspase-3 with both CSI
and EPI treatments
Kalender etal. 2005 VitE and
CTH
In vivo 38 SD rats
6 groups:
CTRL: 1ml/week water i.v
DOX: 5mg/kg/week i.v
VitE: 200IU/kg/week i.v
CTH: 200mg/kg/week i.p
VitE + DOX
CTH + DOX:
Non-sig difference in SOD activity between DOX,
VitE + DOX and CTH + DOX groups
Sig in GPx, CAT, MDA activity in VitE + DOX and
CTH + DOX groups compared to the DOX group
Sig amelioration of hepatocyte changes in VitE + DOX
and CTH + DOX groups compared with DOX group
Somparn etal. 2015 CUR or
THC
In vitro HeLa (Chang liver) cells
CUR or THC 1 or 6μM for 24h
Sig. in cell survival to 80 or 90%, respectively with THC
or CUR (p < 0.05)
Associated with suppressed DOX superoxide formation
and induction of GCLC and NQO1 expression
Yenny etal. 2020 CURL and
VCO
In vivo 15 Wistar rats
5 groups:
CTRL: Na-CMC
DOX: 5mg/kg/week
DOX + VCO: DOX + 6ml VCO
DOX + CURL: DOX + 100mg/kg CURL
VCO + CURL + DOX:
3weeks
Sig in ALT and AST levels in all treated groups
CURL + DOX was more effective than VCO + DOX
Lowest ALT and AST levels in VCO + CURL + DOX
group
Prevention of DOX-induced production of blood lipid
peroxides in rat liver by secondary metabolites of VCO
(flavonoids and phenolics) with antioxidant features
Neutralization of superoxide radicals with antioxidant
effects of curcuminoid compounds in CURL
Khazdair etal. 2016 CUR and
vit C
In vivo 48 male Wistar rat
6 groups (n = 8):
CTRL: 0.5ml NS IV on day 1
DOX: 5mg/kg IV on day 1
Vit C: 100mg/kg in drinking water for 28 consecutive days
DOX + vit C
CUR: 1000mg/kg in drinking water for 28 consecutive days
CUR + DOX
Sig in DOX-induced MDA elevation in DOX + CUR or
Vit C groups
Sig in DOX induced reduction of CAT activity in liver in
Vit C + DOX group but not CUR group
No sig. changes in thiol level by co-administration of CUR
or Vit C
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Mohebbati etal. 2017 CUR, vit
C and
Nigella
sativa
In vivo 64 male Wistar rat
8 groups (n = 8):
CTRL: 0.5ml NS IV on day 1
DOX: 5mg/kg IV on day 1
Vit C: 100mg/kg in drinking water for 28 consecutive days
DOX + vit C
CUR: 1000mg/kg in drinking water for 28 consecutive days
CUR + DOX
Nigella sativa 200mg/kg in drinking water for 28 consecutive days
Nigella sativa + DOX
Sig in DOX-induced MDA elevation in DOX + Vit C
group
Sig in DOX induced reduction of CAT activity and thiol
level in liver in Vit C + DOX and CUR + DOX groups
(in Nigella sativa group, just for CAT activity)
Sadat-Hoseini and DabidiRoshan
2017
Nano-
CUR
In vivo Male Wistar rat
10 groups (n = 8)
DOX 1mg/mL/kg/day, i.p. for 15d, Nanomicellar curcumin soft
gels (Sinacurcumin ®) 100mg/kg gavage for 14days
Insig. in DOX-induced reduction of SOD and also on
apoptosis induing factors
Lalmuansangi etal. 2020 GAP In vivo 42 DLA bearing swiss albino mice (except CTRL group)
7 groups:
CTRL: 0.5ml NS i.p. on day 1
DLA: 0.5ml NS i.p. on day 1 + 0.5ml/day 1% ethanol p.o
DOX: 20mg/kg i.p. on day 1 + 0.5ml/day vehicle p.o
DOX + GAP: DOX + GAP at a dosage of 50, 100 and 150mg/kg
respectively p.o. daily
GAP: 0.5ml NS i.p. on day 1 + 100mg/kg/day GAP p.o
Sig in ALT, AST and LDH in DOX + GAP (150mg/kg)
Sig DOX-induced reduction of antioxidant parameters
levels (GSH, GST, CAT and SOD) in DOX + GAP
groups treated for 7days
Dose-dependent hepatoprotective effect of GAP
Sig in DOX-induced MDA elevation in DOX + GAP
groups with different doses
Zhao etal. 2012 BRB In vivo 40 mice
4 groups:
CTRL: 10mg/kg NS i.p. QOD
DOX: 2.5mg/kg i.p. QOD
DOX + BRB: 60mg/kg BRB i.p. 1h before DOX
BRB: 60mg/kg i.p. QOD
14days
Sig in BW in DOX compared to CTRL, BRB and
BRB + DOX groups
Sig liver weight in BRB + DOX compared to DOX
group
in mortality rates in DOX and BRB + DOX groups
respectively
Sig DOX-induced ALT and AST elevations in
BRB + DOX group; hence, efficient protection against
hepatotoxicity
Sig prevention of DOX-induced hepatic injuries and
inflammation
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Patel etal. 2010 SMN In vivo Male ICR mice
4 groups
CTRL: NS i.p. daily
DOX: 60mg/kg DOX (LD50) on day 12 + NS i.p daily
SMN: 16mg/kg/day p.o
DOX + SMN
-14days
in BW in DOX + SMN group
Sig in mortality rate in DOX + SMN group
Sig in DOX-induced elevation of serum ALT level in
DOX + SMN
Sig in liver histopathological changes in DOX + SMN
group
Sig prevention of DOX-induced oxidative stress ( MDA
levels) in DOX + SMN group
Sig in DOX-induced liver DNA fragmentation due to
blocked DOX-mediated initiation of caspase-activated
DNAse in DOX + SMN group
Sig amelioration of DOX-induced Bcl-xL and PARP in
SMN + DOX group
Little effect of SMN pre-treatment on DOX-induced P53
Sig in DOX- induced increase of DNA cleavage with
SMN pre-treatment
Rašković etal. 2011 SMN In vivo 30 Wistar rats
5 groups:
CTRLS: 1ml/kg p.o
CTRL: 1ml/kg olive oil p.o
SMN: 60mg/kg/day p.o
DOX: 1.66mg/kg i.p. QOD
DOX + SMN
12days
Sig in BW in DOX + SMN group
Sig in DOX-induced LPO in DOX + SMN group
Sig in XOD, CAT and GPx activities in DOX group
No sig changes in GSH level in DOX + SMN group
Sig in AST and LDH levels in DOX + SMN group. No
sig changes in ALT levels
Sig protection against DOX-induced histopathological
alterations
Abbas etal. 2020 SMN and
CGA
In vivo 30 albino rats
5 groups:
CTRL: 0.5ml/kg NS i.p
DOX: 1.5mg/kg i.p. QOD
SMN: 1.5mg/kg DOX i.p. QOD + 206.7mg/kg SMN p.o. QOD
CGA: 1.5mg/kg DOX i.p. QOD + 124mg/kg CGA p.o. QOD
SMN + CGA: DOX + SMN + CGA
Sig liver enzyme activities (AST, ALT and GGT) in
SMN and SMN + CGA than CGA group. More statisti-
cally sig enzymes activities in SMN + CGA group
Sig in DOX-induced caspase-3 elevation in SMN and
CGA groups. More statistically sig caspase-3 and
NF-kB levels in SMN + CGA group
Sig in P-AKt level in SMN and SMN + CGA groups in
comparison with DOX and CGA groups
Sig in MDA level and sig SOD and AMPK levels in
SMN and CGA groups compared to DOX. More statisti-
cally sig amelioration of MDA, SOD and AMPK in
SMN + CGA group compared to others
Sig in hepatic inflammatory reactions (TNF α and IL-1β)
in CGA compared to CTRL group
Sig in inflammatory reactions in SMN group; more sig
TNF α and IL-1β in SMN + CGA group compared to
each one individually
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Barakat etal. 2018 BOS In vivo 40 Swiss albino mice
5 groups:
CTRL: 16mg/kg/week NS i.p. and 12ml/kg/day water p.o
DOX: 6mg/kg/week i.p. + water p.o
BOS125: 125mg/kg/day BOS p.o. + DOX
BOS250: 250mg/kg/day BOS p.o. + DOX
BOS500: 500mg/kg/day BOS p.o. + DOX
3weeks
Sig in ALT and AST in a dose-dependent manner
Sig in MDA levels in all BOS treated groups
Sig in expression of NRF-2 and HO-1 in a dose-depend-
ent manner
Sig in expression of caspase-3 in a dose-dependent
manner
Sig in apoptosis and DNA fragmentations in BOS
treated groups
Sig in DOX-induced histopathological changes in a
dose-dependent manner
Song etal. 2019 DIS In vitro and
invivo
In vitro: AML-12 cells + DIS (50, 100 and 200ng/ml) 6, 12 and
24h before DOX treatment (5ng/ml) for 24h
In vivo: 40 male mice
5 groups:
CTRL: 0.5% Na-CMC
DOX: 15mg/kg i.p. on the 7th day
DIS60: 60mg/kg DIS p.o. + DOX
DIS30: 30mg/kg DIS + DOX
DIS15: 15mg/kg DIS + DOX
14days
Sig protection of DOX-induced cell injuries in a dose- and
time-dependent manner
Sig reverse of DOX-induced morphological changes
invitro and invivo experiments
Sig in ALT and AST levels
Sig in oxidative stress ( ROS and MDA levels)
Sig in antioxidant parameters (GSH, GPx and SOD
levels)
Sig in DOX-induced apoptosis invitro and invivo
experiments ( TUNEL positive cells)
Sig in pr levels of NRF-2 and HO-1 and pr levels of
FOXO1 and Keap1 invitro and invivo studies
Sig in DOX-induced inflammation ( expression of
NF-kB and mRNA levels of IL-1β, IL-6 and TNF-α)
invitro and vivo experiments
Sig in expression levels of P53 and Bax and Bcl -2 in a
dose dependent manner
Sig in oxidative stress, inflammation and apoptosis via
Sirt 1 expression levels
Jung etal. 2014 EDI In vitro Hepatocytes isolated from SD rats
Adding 10µl of 5µM DOX to each well and 2days incubation
Sig protective effects of EtOAc and n-BuOH fractions of
EtOH extract of EDI with EC50 values of 3.0µg/ml and
8.4µg/ml respectively as compared with positive CTRL
SMN (EC50 value = 8.0µg/ml)
Concurrent cytotoxic effects at hepatoprotective concen-
tration observed with CH2Cl2 fraction
No sig protective effects observed in n-hexane and H2O
fractions
Sig protective effect of EtOAc fraction’s plorotanins
respectively:
dioxinodehydroeckol > phlorophucophuroeckol
A > dieckol > eckol > triphloroethol-A
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Rashid etal. 2013 CH In vivo 30 Wistar rats
5 groups:
Negative CTRL: 10mg/kg/day corn oil and NS single i.p
Positive CTRL: 40mg/kg DOX i.p. on day 14
DOX + CH40: 40mg/kg/day CH p.o. + DOX
DOX + CH80: 80mg/kg/day CH p.o. + DOX
CH: 80mg/kg/day p.o
16days
Sig of DOX-induced hepatic toxicity markers (serum
ALT, AST and LDH activities) with CH treatment at
both doses
Sig of DOX-induced reduction of hepatic GSH levels in
CH treatment with both doses
Sig of DOX-induced reduction of hepatic antioxidant
enzymes (GPx, GR, CAT and SOD) in CH pre- and
post-treatment
Sig of MDA formation by mending membrane integrity
and DOX-induced LPO in DOX + CH treatments
CH pre-treatments retained the structure of liver cells near
to that of CTRL group
Liu etal. 2017 NIN In vitro and
invivo
In vitro: Hela cells seeded on a 96-well plate and then added 20µl
of DOX (1.6 to 10.0µg mL−1) and NIN (200 to 2000µmol L1)
at different concentrations
In vivo: 32 Hela cells bearing athymic nude mice
4 groups: CTRL: NS; DOX: 5mg/kg; NIN: 20mg/kg;
DOX + NIN
DOX and NIN administration with 3-day interval for 5 times
Sig anti-tumor efficacy in DOX + NIN group compared to
DOX and NIN groups invitro and invivo
Sig in DOX-induced weight loss and systemic sides
effects in DOX + NIN group
Sig in hepatocyte necrosis in DOX + NIN compared to
DOX group
Wali etal. 2020 NIGN In vivo 24 Wistar rats
4 groups:
CTRL: vehicle p.o. daily
DOX: 20mg/kg i.p. on the 20th day
NIGN50: 50mg/kg/day NIGN p.o. + DOX
NIGN100: 100mg/kg/day NIGN p.o. + DOX
20days
Sig in oxidative stress (ROS and MDA levels)
Sig in antioxidant enzymes (GSH, GPx, GR, SOD and
CAT) in NIGN treatments in a dose dependent manner
Sig of H2O2 in liver tissue
Sig in DOX-induced serum toxicity markers (ALT,
AST, ALP, LDH and total pr)
Sig in inflammatory mediators (NF-kB, TNF-α, IL-1β,
PGE-2, TGF-β and IL-6)
Sig in DOX-induced elevated NO levels
Sig protection of DOX-induced liver damages in NIGN
treatment at higher dose
Sig in expression of COX-2 in NIGN treatments con-
cludes anti-inflammatory potential of NIGN
Anandakumar etal. 2007 LA In vivo 24 Wistar rats
4 groups:
CTRL: NS
DOX: 15mg/kg DOX single injection i.p
LA: 75mg/kg LA single injection i.p
DOX + LA: single injection of LA 24h before DOX
4days
Sig of DOX-induced elevation of serum enzymes (ALT,
AST and BL) and Sig of DOX-induced reduction of
liver enzymes (ALP, LDH, AST and ALT) in DOX + LA
group
Sig of lipid per-oxidations (basal, ascorbate and ferrous-
sulfate-induced) in DOX + LA group
Sig of antioxidants (GSH, CAT, SOD, GPx, GR, GST
and G6PD) in DOX + LA group
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Mokni etal. 2015 GSSE In vivo 24 Wistar rats
4 groups:
1.CTRL: ethanol i.p
2. GSSE: 500mg/kg/day i.p
3. DOX: 20mg/kg single i.p. on day 4 till day 8
4. DOX + GSSE
8days
Sig in DOX-induced increase of AST and ALT levels
Sig in MDA level and pro-oxidative effect of DOX in
DOX + GSSE group
Sig amelioration of DOX-induced liver antioxidant
enzymes alterations (CAT, POD and SOD) in GSSE
pre- and post-treatment
Sig restore of DOX-induced intracellular mediator disor-
ders ( free Fe, H2O2 and Ca) nearly to CTRL levels
Sig protection of liver structure against DOX-induced liver
damages
Aktaş etal. 2020 ATS In vivo 49 SD rats
7 groups:
1.CTRL: untreated
2. DOX: 10mg/kg single i.p. on first day
3.DOX + ATS7: DOX + 7mg/kg/day ATS p.o
4. DOX + ATS35: DOX + 35mg/kg/day ATS p.o
5. Negative CTRL: 1ml/kg/day NS p.o
14days
Sig in liver degenerative changes and histological score
in DOX group
Caspase-3 and expression of TNF-α in hepatocytes in
DOX group; no expression in other groups
expression of iNOS in DOX group and in
DOX + ATS7 group; no expression in other groups
NF-kB expression in DOX group and in ATS treat-
ments with either dose
Kuzu etal. 2019 Morin In vivo 35 Wistar albino rats
5 groups:
CTRL: NS p.o. + NS i.p. on the 8th day
Morin: 100mg/kg/day p.o + NS i.p. on the 8th day
DOX + morin50: 40mg/kg DOX single i.p. on the 8th day + 50mg/
kg/day morin
DOX + morin100
10days
Sig in MDA levels in morin pre-treatment groups in a
dose dependent manner
Sig in antioxidant markers (SOD, CAT and GPx)
Sig improving of liver function by ALT and AST activi-
ties
Sig regulatory effect on TNF-α, IL-1β and NF-κB levels
Sig anti-apoptotic effect Bcl-2 level and Bax level in a
dose dependent manner
Sig preservation of liver structure against DOX damages
in DOX + morin100 group
Ma etal. 2020 ATX In vivo 30 ICR mice
5 groups:
CTRL: 6mg/kg NS i.p
DOX: 6mg/kg single i.p
Solvent: DOX + 50mg/kg/day corn oil
ATX50: DOX + 50mg/k/day ATX
ATX100: DOX + 100mg/k/day ATX
3weeks
Sig in BW and food intake in ATX groups compared to
DOX group
Sig in liver function by DOX-induced elevation of
ALT, AST, ALP and total BL in ATX groups with either
doses
Sig preservation of hepatocytes against DOX-induced
injuries
Sig anti-apoptosis features of ATX treatments with both
doses
Sig in antioxidant markers levels (SOD, CAT and GPx)
in ATX groups
Sig in ROS and MDA levels in ATX groups
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Kamble and Patil 2018 AA In vivo 36 Wistar rats
6 groups:
CTRL: 1ml/day NS
STD: 200mg/kg VitE p.o
DOX: 65.75mg/kg i.v
AA5: 5mg/kg/day p.o
AA10: 10mg/kg/day p.o
AA20: 20mg/kg/day p.o
DOX administration in all groups (except CTRL) 48h before the
end of study
7days
Sig in DOX-induced upsurge of injury marker enzymes
(LDH, ALT and AST)
Sig in antioxidant markers (SOD and GSH) in AA20
group
Sig in DOX-induced elevation of LPO in AA20 group
Sig protection of liver architecture against DOX-induced
alterations
Roomi etal. 2014 NM In vivo 24 Balb/c mice
4 groups:
CTRL: RCD
NM: RCD + 20mg/day NM
DOX: RCD for 3weeks prior to 20mg/kg DOX i.p
DOX + NM: RCD + DOX + NM
3weeks
No sig effect of DOX on liver weight in both groups
Sig in food intake in mice treated with NM
Sig protection of liver function against DOX-induced
serum enzyme levels (ALT, AST and GGT)
Afsar etal. 2019 AHE In vivo 36 SD rats
6 groups:
CTRL: 0.4ml/week NS i.p
DOX: 3mg/kg/week i.p
AHE: 400mg/kg/day p.o
DOX + AHE200
DOX + AHE400
DOX + SMN: DOX + 100mg/kg SMN 2 times a week
6weeks
Sig amelioration of DOX-induced BW and liver weight
with AHE treatments in a dose dependent manner
Sig in ALT, AST, ALP, LDH, total and direct BL with
AHE treatments in a dose dependent manner
Sig in chol, TG and LDL with AHE treatments
Sig in HDL in DOX + AHE400 group
Sig in antioxidant parameters levels (POD, SOD, CAT,
QR, GSH, GR, GST, GGT and GPx) with AHE treat-
ments dose dependently
Sig in oxidative stress markers (H2O2 and NO) and LPO
(TBARs) with AHE treatments dose dependently
Sig in DOX-induced liver injuries with AHE high dose
Rafiee etal. 2021 pCA In vivo 32 Wistar rats
4 groups:
CTRL: 10% PG p.o
pCA: 100mg/kg/day p.o
DOX: 15mg/kg i.p. on day 5
DOX + pCA
5days
Sig in serum ALT, AST, ALP, TG, chol, LDL, and total
BL and sig in HDL and serum ALB with pCA treat-
ment
Sig in LPO (MDA) and sig antioxidant enzymes
activities (GPx, SOD and CAT)
Sig prevention of DOX-induced liver histopathological
changes
Sig in IL-1β expressions in liver tissues
Khajavi Rad 2021 ZJ In vivo Wistar rats
5groups: CTRL, DOX and 3 orally doses of ZJ
1month
Sig in ALT, AST, LDH, ALP, total and direct BL
Sig in oxidative stress
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Table 1 (continued)
Study (year)
[reference]
Drug Type of study The way of administration and the dose of DOX Results/mechanisms
Husna etal. 2022 MNG In vivo 25 SD rats
5 groups:
Normal: corn oil and NaCl 0.9% QOD fo 2weeks
DOX: 2.5mg/kg DOX QOD for 2weeks
MNG50: DOX + 50mg/kg MNG PO for 5weeks
MNG100: DOX + 100mg/kg MNG PO for 5weeks
Positive CTRL: DOX (2weeks) + 50mg/kg SMN (5weeks)
Sig in ALT and AST in MNG50 and 100 groups
Sig in MDA level and sig GSH and SOD in MNG50
and 100 groups
Slight liver damage in MNG goups compared to DOX
group (not assessed statistically
Improvement of liver function and oxidative stress with
MNG treatment
Ahmed etal. 2022 TYM and
TML
In vivo 24 wistar rats
4 groups:
Normal: 0.9% NaCl 1ml/kg i.p. every week
+ 1% CMC 5ml/kg QD p.o. for 7weeks
DOX: 2mg/kg i.p. every week + 1% CMC p.o. QOD for 7weeks
TYM: 2mg/kg DOX i.p. every week + 250mg TYM p.o. QOD for
7weeks
TML: 2mg/kg DOX QOD + 100mg TML p.o. QOD for 7weeks
Sig in DOX induced elevated AST, ALT, ALP and total
BL in TML group
Distinguished but non-sig ALB level in TYM group
(TML more effective in ameliorating liver function than
TYM exception for ALB level)
Sig in AFP and CA19.9 (tumor marker), serum TNFα
and IL-4 in both treated groups (TYM more effective)
Sig in LPO and GSH and activities GST and GPx
Sig in DOX-induced reduction of Bcl-2 mRNA expres-
sion (TML more effective)
Sig in liver p53 expression
Sig in liver histopathological changes including inflam-
mation, apoptosis and fibrosis
Improvement of liver function, inflammation, oxidative
stress, apoptosis, and liver structure
Mohebbati etal. 2018 ZM and
CAR
In vivo 24 wistar rats
4 groups:
Normal: 0.9% NaCl
DOX: 5mg/kg IV 28 d + NS
DOX + ZM: 200mg/kg IV 28 d
DOX + CAR: 20mg/kg IV 28 d
Sig. in serum ALP, ALT and AST in ZM and CAR
groups compared to those of the ADR group
Sig. in MDA in ZM and CAR groups compared to those
of the ADR group
Sig in CAT activity in ZM and CAR groups compared to
those of the ADR group
Sig in thiol levels in ZM group compared to the ADR group
CTN, citronellal; SD, Sprague–Dawley; CTRL, control; NS, normal saline; p.o., orally; DOX, doxorubicin; i.p., intraperitoneal; sig, significant; ALB, albumin; AST, aspartate aminotransferase;
ALT, alanine aminotransferase; MDA, malondialdehyde; GSH, glutathione; SOD, superoxide dismutase; Bax, Bcl-2 associated X protein; Bcl-2, B-cell lymphoma 2; pr, protein; PI3k, phospho-
inositide 3-kinase; Akt, protein kinase B; G10, [10]-Gingerol; TNBC, triple negative breast cancer; GIN, Zingiber officinale; QE, quercetin; GPx, glutathione peroxidase; CSI, Camellia sinensis;
EPI, epicatechin; CMC, carboxymethyl cellulose; QOD, every other day; GGT , Gamma-glutamyl transferase; AFP, alpha-fetopotein; BL, bilirubin; LPO, lipid peroxidation; GST, glutathione-
S-transferase; NF-kB, nuclear factor-kappa B cells; COX-2, cyclooxygenase-2; VitE, vitamin E; CTH, catechin; CUR , curcumin; THC, tetrahydrocurcumin; GCLC, glutamylcysteine ligase cata-
lytic subunit; NQO1: NADP (H), quinone oxidoreductase1; CURL, Curcuma longa; VCO, virgin coconut oil; Na-CMC, sodium carboxymethyl cellulose; GAP, Ganoderma applantum; DLA,
Dalton’s lymphoma ascites; BRB, berberine; SMN, silymarin; Bcl-xL, B-cell lymphoma-extra large; PARP, poly-ADP-ribose polymerase; P53, tumor protin P53; XOD, xanthine oxidase; CGA
,chlorgenic acid; P-AKt, protein kinase B; AMPK, AMP-activated protein kinase; TNF-α, tumor necrosis factor-α; IL-1β, interleukin-1β; BOS, boswellic acid; NRF-2, nuclear factor erythroid2-
related factor 2; HO-1, heme oxygenase 1; DIS, dioscin; AML-12 cells, alpha mouse liver 12 cells; ROS, reduced reactive oxygen species; TUNEL, terminal deoxynucleotidyl transferase dUTP
nick end labeling; FOXO1, forkhead box rotein O1; keap1, kelch like ECH associated protein 1; IL-6, interleukin-6; EDI, edible brown alga Ecklonia stolonifera; CH, chrysin; Cr, creatine; STD,
standard; FN-1, fibronectin; 8-OHdG, 8-hydroxydeoxyguanosine; MCP-1, chemoattractant protein-1; NIN, naringin; NIGN, naringenin; PGE-2, prostaglandin-E2; TGF-β, transforming growth
factor beta; LA, DL-alpha lipoic acid; G6PD, glucose-6-phosphate dehydrogenase; GSSE, grape seed and skin extract; POD, peroxidase; ATS, artemisinin; ATX, astaxanthin; AA, asiatic acid;
NM, nutrient mixture; RCD, regular chow diet; ZM, zataria multiflora; CAR , carvecrol; AHE, acacia hydaspica; QR, quinone reductase; pCA, p-coumaric acid; ZJ, Ziziphus jujuba; EXR, short
term exercise; NrF-1, nuclear respiratory factor 1; HSP70, heat shock protein 70; Sirt1, sirtuin 1; AIR, regular aerobic training; IGF-1, insulin-like growth factor-1; IGFBP-3, insulin-like growth
factor binding protein-3; MNG, mangiferin; TYM, thyme oil; TML, thymolol
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
dissemination and inhibited chemotherapy-induced weight
loss and hepatotoxicity. The combination of these drugs also
resulted in decreasing the numbers of circulating tumor cells
in the peripheral blood of the tumor-bearing animals.
In addition, two other studies evaluated the hepatoprotec-
tive effects of aqueous extract of ginger on DOX in albino
Wistar rats. In the first study, Sakr etal. mentioned that adri-
amycin (2mg/kg/w) induced various histological changes in
the liver and significantly increased the serum levels of ALT
and AST after 4 and 6weeks of treatment. It also caused an
increase in malondialdehyde (lipid peroxidation marker) and
depletion of the antioxidant enzyme; superoxide dismutase.
On the other hand, treatment of the rats with aqueous extract
of ginger (24mg/kg thrice weekly for 6weeks) beside adria-
mycin led to histological and functional improvement of the
liver. The ginger extract also reduced the level of malon-
dialdehyde and increased the activity of superoxide dis-
mutase. In the second study, Ahmed etal. also showed that
adriamycin (2.5mg/kg/d) resulted in the destruction of the
hepatic cords and adverse pathological changes in the liver
of animals. However, these hepatotoxic effects were par-
tially ameliorated when treatment was combined with ginger
(250mg/kg/d) for 2weeks (Ahmed etal. 2013). Moreover,
considering the dose of ginger and DOX in two abovemen-
tioned studies, it seems that the protective effect of ginger is
dose dependent, and the cumulative dose of both ginger and
DOX is almost similar in both studies. So, gingerol possess
antioxidant and anti-inflammatory features and can prevent
the liver damage by these mechanisms.
Quercetin
Quercetin is a flavonoid which is found in several fruits and
vegetables and receives an increasing attention as an anti-
oxidant and a pro-apoptotic flavonoid. Two invitro studies
have evaluated its hepatoprotective role against DOX. The
first study was conducted by Wang etal. on liver in C57BL/6
mice. Their investigations, using the MTT and Annexin V/
PI staining assay, revealed that that quercetin (100mg/kg/d
4days) significantly stimulated the cytotoxicity of DOX
Table 2 Summarized hepatoprotective natural-occuring compounds against doxorubicin hepatotoxicity; clinical studies
CAF, cyclophosphamide 600mg/m2 + doxorubicin 60 mg/m2 + 5-FU 600 mg/m2; TB, total bilirubin; RCT, randomized clinical-trial; AC-T regi-
men: doxorubicin, cyclophosphamide, and paclitaxel regimen; CPY, cyclophosphamide; DOX, doxorubicin
Combination Type of study Subject Dose Result Ref
SMN/CAF Open label RCT 74 patients with breast
cancer
Treatment group 1
(n = 25): 210mg/d
SMN
Treatment group 2
(m = 25): 420mg/d
SMN
Control group
(n = 24): no inter-
vention
for three course of
21days
-Sig. elevation of AST
and ALT in group
1, sig. reduction
with SMN, in a time
and dose dependent
manner
-No sig. change in TB
after treatment with
SMN
Mshemish etal. (2011)
SMN / DOX, epi-
rubicin, CPY, and
taxanes
placebo-controlled
RCT
99 patients with
invasive breast
carcinoma
Study group (n = 49):
70mg (Livergol®)
PO three times daily
Control group
(n = 50): Placebo
-Sig. lower increase
in AST ad ALT in
treatment group
Mohaghegh etal.
(2015)
SMN /AC-T Triple blind, placebo-
controlled RCT
30 breast cancer
patients who
received AC-T
regimen
Treatment group
(n = 15): 140mg
(Livergol®) three
times a day for
1month
Placebo group (n = 15)
- Non-sig trend
toward more severe
liver involvement in
placebo group based
on ultrasonography
- Sig. hepatic involve-
ment grade improve-
ment based on
ultrasonography in
treatment group
-No sig. different
between two groups
based on FibroScan
and liver function
tests
Askarpour etal.2021
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Fig. 1 Probable mechanisms of doxorubicin induced hepatotoxicity
Fig. 2 Diagram of the study
selection process
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
(20mg/kg on day 5) in liver cancer cells while protecting
normal liver cells. This pro-apoptotic effect on hepatoma
cells was p53-dependent and occurred by downregulating
Bcl-xl expression. Moreover, quercetin reversed the DOX-
induced pathological changes and decreased the serum lev-
els of ALT and AST (Wang etal. 2012).
In the other study, Kocahan etal. evaluated the effects
of quercetin (10mg/kg/day) on doxorubicin (1.8mg/kg IP
every 3weeks, for 10weeks) (Kocahan etal. 2017). Querce-
tin diminished the DOX-induced production of malondial-
dehyde and also increased glutathione peroxidase levels. So,
it seems that quercetin with anti-apoptotic and antioxidant
activity can be effective for prevention of DOX liver injury.
Vitamin E, catechin, epicatechin, andgreen tea
Vitamin E is an antioxidant with endogenous origin which
can suppress the propagation of lipid peroxidation and is
helpful in scavenging free radicals. Catechin is a type of
natural phenols with abundant antioxidant activity and is
a plant secondary metabolite, belonging to the group of
flavan-3-ols. Kalender etal. showed promising hepatopro-
tective effects of vitamin E (200IU/kg/week) and catechin
(200mg/kg/week) in SD rats against DOX (5mg/kg/week)
by obtaining the maximum levels of glutathione peroxidase
(Kalender etal. 2005), whereas malondialdehyde levels
considerably decreased in both vitamin E + DOX and cate-
chin + DOX groups. Electron microscopic studies supported
biochemical hepatoprotective findings.
Also, Ahmed etal.’s treatment of DOX-injected Wistar
rats (4mg/kg i.p. weekly) with aqueous extracts of Camel-
lia sinensis (green tea) leaf (200mg/kg) and epicatechin
(25mg/kg) resulted in significant correction of albumin,
alpha-fetoprotein, bilirubin, ALT, AST, alkaline phos-
phatase, and gamma glutamyl transferase (GGT) levels. The
treatments also diminished the serum levels of tumor necro-
sis factor (TNF)-α and interleukin-4, liver lipid peroxidation,
and glutathione levels as well as liver superoxide dismutase,
glutathione peroxidase, and glutathione-S-transferase activi-
ties. The expressions of liver nuclear factor-kappa B cells,
p53, and caspase-3 were also highly decreased, whereas the
expression of Bcl-2 was increased, and the liver histologi-
cal architecture was remarkably ameliorated. The authors
concluded that Camellia sinensis aqueous extract and epi-
catechin may have great hepatoprotective effects against
DOX which seems to be through reinforcement of antioxi-
dant defense system and attenuation of the inflammation and
apoptosis (Ahmed etal. 2019).
In another study, Roomi etal. evaluated the preventive
effect of a unique nutrient mixture (NM) (20mg/kg), contain-
ing primarily ascorbic acid, lysine, proline, N-acetyl cysteine,
and green tea extract against adriamycin (20mg/kg)-induced
hepatic and renal toxicity in male BALB/c mice. This extract
exhibits a broad spectrum of pharmacological activities,
including antioxidant activity. NM significantly increased
food intake in the mice and significantly protected the liver
function against DOX-induced elevation of serum liver
enzyme levels (ALT, AST, and GGT) (Roomi etal. 2014).
Curcumin
Curcumin (CUR) is a natural antioxidant agent which is
nontoxic even up to 12g/day based on human clinical trials.
Various studies focused on CUR antioxidant, anti-inflam-
matory, and anti-tumor activities. This bioactive molecule
is isolated from the rhizomes of Curcuma longa (turmeric).
Lots of studies have highlighted the role of CUR in inhibi-
tion of ROS generation in numerous cell lines and organ tis-
sues of animals and shown beneficial effects in management
of different DOX-induced toxicity including heart, CNS,
kidney, and liver. Moreover, it is proposed that curcumin
may suppress tumor growth, by scavenging free radicals and
inhibiting ABC drug transporters which bring it up as a host
of research studies for improving chemotherapeutic agents’
efficacy (Mohajeri & Sahebkar 2017). Regarding its hepato-
protective effects against DOX, several invitro and invivo
researches are available. An invitro study on Chang liver
cells, pre-treatment with tetrahydrocurcumin or CUR (1 or
6μM) for 24h significantly increased cell survival to 80 and
90%, respectively via suppression of DOX-induced superox-
ide formation and induction of lutamylcysteine ligase cata-
lytic subunit (GCLC) and NADP (H): quinone oxidoreduc-
tase1 (NQO1) expression (Somparn etal. 2015). In Khazdair
etal.’s study, Curcuma longa in dose of 1000mg/kg PO
for 28 consecutive days could be protective against hepa-
totoxicity induced by DOX 5mg/kg in rats, via antioxidant
mechanism. It significantly increased MDA level but no con-
siderable changes in thiol level and also catalase activity in
liver tissue (Khazdair etal. 2016). An additional study also
used curucumin with same dosing alone or in combination
with Nigella sativa (200mg/kg) beside the same dose of
DOX, and they found that thiol level and also CAT activity
increased in treatment groups (Mohebbati etal. 2017). In
another study, DOX was administrated for 15days (1mg/
mL/kg/day, i.p.), and nanocurcumin supplement was admin-
istrated for 14days (100mg/kg/day orally). However, it did
not show significant antioxidant and anti-apoptosis effect
(Sadat-Hoseini & DabidiRoshan 2017).
Virgin coconut oil andCurcuma longa
The pure coconut oil like virgin coconut oil (VCO) is a strong
antioxidant and has several beneficial characteristics which
make it effective against the liver injury caused by chemo-
therapeutic agents (Famurewa etal. 2017). Recently, Yenni
etal. conducted a study comparing the hepatoprotective
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
activity of VCO and Curcuma longa by evaluating the AST
and ALT level in Wistar rats. They named the treatments as
group I (DOX 5mg/kg + 6ml VCO), group II (DOX 5mg/
kg + Curcuma longa 100mg/kg), and group III (DOX 5mg/
kg + 6ml VCO + Curcuma longa 100mg/kg). Their results
demonstrated that DOX greatly increased the AST and ALT
and caused significant liver injuries, but treatment with a
combination of the VCO and Curcuma longa resulted in
significant hepatoprotective effects (Yenny etal. 2020). The
probable mechanism of its action is related to its antioxidant
properties but is not assessed in this study.
Ganoderma applanatum extract
In 2014, Lalmuansangi etal. explored the ability of the medic-
inal fungus Ganoderma applanatum (GAP) in providing
protective effects against DOX-induced toxicity in Dalton’s
lymphoma ascites (DLA)-bearing mice. Their study demon-
strated that treatment of DLA mice with DOX (20mg/kg i.p.
on day 1 + 0.5ml/day vehicle p.o.) significantly increased the
serum levels of AST, ALT, and LDH. However, co-treatment
of DOX with intraperitoneal injection and G. applanatum
(0.5ml NS i.p. on day 1 + 0.5ml/day 1% ethanol p.o.) resulted
in diminished levels of GSH and decreased activities of glu-
tathione-s-transferase, catalase, and superoxide dismutase,
and antioxidant property is its main proposed mechanism of
action. Their results also revealed that among various sol-
vent extracts of G. applanatum, methanolic extract showed
the highest phenolic and flavonoid contents compared to the
aqueous and chloroform extracts (Lalmuansangi etal. 2020).
Berberine
Berberine is an isoquinoline alkaloid, which is extracted
from the traditional Chinese plant Coptis chinensis and is
used for treatment of various infectious diseases. However,
recent studies have established that this alkaloid has a wide
range of pharmacological activities, including antidiabetic,
anti-inflammatory, and antitumor effects (Wang etal. 2020).
In particular, both animal and clinical investigations have
reported that berberine is beneficial in protection against
ROS formation (Cho etal. 2005). In a study, Zhao etal.
observed an increased mortality rate, a declined body
weight, and increased plasma ALT and AST level in DOX-
treated mice (2.5mg/kg i.p.). These changes were largely
prevented by pretreatment with berberine (60mg/kg i.p.)
(Zhao etal. 2012). Its mechanism of action did not define in
this study and should be determined in the future.
Silymarin andchlorogenic acid
Silymarin is an extract of Silybum marianum, which is
famous historically for its hepatoprotective properties
via apoptotic and antioxidant mechanisms (Gillessen &
Schmidt. 2020). Patel etal. evaluated the effects of sily-
marin pretreatments at the dose of 16mg/kg/day p.o. on
DOX-induced liver injuries in male ICR rats. The results
demonstrated an increased body weight in DOX + silymarin.
In addition, it diminished undesirable histopathological and
functional changes and mortality rate and prevented DOX-
induced oxidative stress (marked by decreasing malondial-
dehyde). Significant amelioration of DOX-induced Bcl-xL
decrease and PARP increase was also observed in this group
(Patel etal. 2010).
In the second study, Rašković etal. used a higher dose
of silymarin (60mg/kg/day p.o) for a time span of 12days
beside DOX (1.66mg/kg i.p. QOD) in male Wistar rats.
Their study showed a significant change in body weight,
ECG, oxidative stress parameters, serum ALT, AST, LDH,
and creatine kinase levels and histopathological preparations
of heart and liver specimens. They concluded that silymarin,
at the examined dose, had a protective influence on the liver
(and heart) tissues against DOX-induced toxicity (Rašković
etal. 2011).
The third study, conducted by Abbas etal. evaluated the
protective effects of silymarin in combination with chloro-
genic acid (CGA) 1.5mg/kg, on DOX-induced hepatotox-
icity (206.7mg/kg) every other day for 4weeks in Albino
rats. The results demonstrated that CGA caused a significant
decrease in apoptosis in hepatic cells (marked by caspase-3
and nuclear factor-κB levels), significant amelioration of
hepatic oxidant status (marked by decreasing malondialde-
hyde and superoxide dismutase), and significant decrease in
hepatic inflammatory markers (TNF-α and IL1β) compared
with DOX treatment alone (Abbas etal. 2020).
It should be mentioned that the administered dose of sily-
marin in abovementioned studies varied too much, and daily
dose of 16- 103mg/kg was administered in these three stud-
ies. The proposed mechanisms of action were anti-inflam-
matory, antioxidant, and anti-apoptotic ones.
Following these three animal studies, three clinical trials
were also performed on silymarin. In first study, Mshemish
etal. patients were randomly assigned to one of three groups
of fluorouracil/doxorubicin/cyclophosphamide (FAC) alone
(n = 24), FAC plus low-dose silymarin (210mg per day,
n = 25), and FAC plus high-dose silymarin (420mg per day,
n = 25), for three courses of 21days. Silymarin showed a
dose and time-dependent prophylactic effect at the end of
the first, second and third chemotherapy courses. But no sig-
nificant change in total bilirubin serum level was found after
treatment with silymarin (Mshemish etal. 2011). However,
they only assessed liver function tests with no ultrasonogra-
phy or transient elastography (TE) assessment, and silymarin
was used as a preventive measure.
In the second randomized clinical trial (RCT), 99 patients
with invasive breast carcinoma receiving chemotherapy that
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
contained Adriamycin or epirubicin, cyclophosphamide,
docetaxel, or paclitaxel randomly received silymarin 70mg
PO three times daily or the placebo during their treatment
course. The patients had mild rises in AST, ALT, and biliru-
bin in both groups, but the changes were less notable in the
study group, with significant difference between two groups
(Mohaghegh etal. 2015).
In third study, Askarpour Moezian etal. assessed the
therapeutic effect of silymarin, 140mg three times a day on
DOX-induced hepatotoxicity in patients with non-metastatic
breast cancer who received doxorubicin/cyclophosphamide-
paclitaxel (AC-T) regimen; in contrast to the previous stud-
ies which focused on preventive effects, they found a non-
significant trend toward more severe hepatic injury in the
placebo based on ultrasonography. Besides, in silymarin
group, hepatic involvement grade based on ultrasonogra-
phy considerably reduced after intervention. However, no
difference was found between two groups based on TE and
liver function tests (Askarpour etal. 2021). These three stud-
ies are summarized in Table2. As TE has been repeatedly
validated and has shown overall good accuracy in evaluat-
ing fibrosis and cirrhosis in different settings, the promising
effect of silymarin based on ultrasonography should be con-
firmed in future well-designed RCT using TE as a measur-
ing tool. Moreover, the mechanism of action is better to be
assessed in human studies.
Boswellic acid
Acetyl 11-keto-b-boswellic acid (AKBA) is a pentacyclic
triterpenoid compound, which is known as the most effective
component of Boswellia serrata resin (Safayhi etal. 1992).
The antioxidant protective effect of AKBA is thought to
be mediated through activating the NF-E2-related factor 2
(Nrf2) pathways (Zhang etal. 2016). In the study of Barakat
etal., the Swiss albino rats received DOX (6mg/kg/w), with
or without protective doses of BOS (125, 250, and 500mg/
kg/day). The results demonstrated that treatment with BOS
significantly reduced ALT, AST, and malondialdehyde.
These findings were coupled with significant improvement
in histological features of the liver. BOS increased the Nrf2
and heme oxygenase-1 (HO-1) expression, which further
confirmed a possible mechanism for protective effects of this
agent, which is antioxidant property (Barakat etal. 2018).
Dioscin
Dioscin is a naturally derived triterpenoid saponin which has
protective activities against organ damage and acts vigorously
against metabolic diseases. Recent studies have shown the
therapeutic effects of dioscin on acute liver injury through
diminishing oxidative stress and inhibition of inflammation
through suppression of various pathways including NF-κB
(Liu etal. 2015). Based on these considerations, Song etal.
investigated the protective effects of dioscin on DOX-induced
liver injury by invitro (in AML-12 cells) and invivo (in male
mice) studies. For the invitro study, they used dioscin for 6, 12,
and 24h before DOX treatment (5mg/ml for 24h). For invivo
study, they used 15, 30, and 60mg/kg of dioscin. Their results
showed that dioscin significantly decreased DOX-induced
cell injury and ROS level and suppressed DOX-induced cell
apoptosis in AML-12 cells. Regarding the results from invivo
investigations, dioscin evidently decreased the levels of ALT,
AST, and malondialdehyde and increased the levels of super-
oxide dismutase, glutathione, and glutathione peroxidase. In
pursuit of the mechanism of this liver protection, they found
that dioscin significantly up-regulated the expression of silent
information regulator 1 (Sirt1) and HO-1 through increase of
Nrf2 and inhibited the expression of forkhead box protein O1
(FOXO1) and kelch-like ECH-associated protein-1 (Keap1),
resulting in the inhibition of oxidative stress. Dioscin also obvi-
ously decreased the NF-κB protein level and the mRNA levels
of TNF-α, IL-1β, and IL-6, which resulted in suppression of
inflammation. On the other hand, dioscin inhibited apoptosis
through regulation of p53 and BCL-2. These results demon-
strated that dioscin inhibited DOX-induced hepatotoxicity via
Sirt1/FOXO1/NF-κB signal (Song etal. 2019). So, all three
anti-apoptotic, anti-inflammatory, and antioxidant mechanisms
could be considered for its action.
Edible brown alga Ecklonia stolonifera
Species of Ecklonia have been reported to be potent phlo-
rotannin-rich raw materials. Phlorotannins are secondary
metabolites of phloroglucinol which have been polymer-
ized through different solvents including ether, phenyl, or
1,4-dibenzodioxin. These polyphenolic compounds show a
variety of bioactivities, including anti-diabetic, anti-inflam-
matory, and hepatoprotective activities (Kim etal. 2005).
Jung etal. investigated the protective capacity of 14 edible
varieties of Korean seaweed against DOX-induced liver
injury invitro in primary rat hepatocytes (Jung etal. 2014).
This study was carried out by adding 10µl of 5-µM DOX to
each well and 2days incubation and half maximal effective
concentration (EC50) values of 2.0, 2.5, 3.0, and 15.0µg/
ml, respectively. The results demonstrated significant pro-
tective effects of EtOAc and n-BuOH fractions of ethanol
extract of Ecklonia stolonifera with EC50 values of 3.0µg/
ml and 8.4µg/ml, respectively, as compared with positive
control silymarin (EC50 value = 8.0µg/ml). In addition, they
observed concurrent cytotoxic effects at hepatoprotective
concentration observed with CH2Cl2 fraction. However, no
significant protective effect was observed in N-hexane and
H2O fractions (Jung etal. 2014). Its mechanism of action
is not well defined.
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Chrysin
Chrysin (5,7-dihydroxyflavone) is a natural flavonoid which
has been isolated from different plants, as well as honey and
propolis. This flavonoid has important pharmacological prop-
erties, including antidiabetic, antioxidant, and anti-inflamma-
tory effects and can prevent chemoresistance through inhibi-
tion of efflux proteins (Pushpavalli etal. 2010). Rashid etal.
evaluated the protective efficacy of chrysin (CH) (40 and
80mg/kg PO) against DOX-induced oxidative stress and kid-
ney and liver injuries (40mg/kg) in male Wistar rats. Treat-
ment with CH significantly diminished serum levels of ALT,
AST, and LDH. These observations were also confirmed by
the histopathological findings and antioxidant enzyme level
changes, which were highly reversed by chrysin via antioxi-
dant mechanism (Rashid etal. 2013).
Naringin andnaringenin
Naringin (NIN) is a flavonoid extracted from grapefruit or
other citrus fruits. This flavonoid is stated to have a variety of
pharmacological characteristics, including antioxidant, antia-
therogenic, and anti-inflammatory activities (Nie etal. 2012).
In 2017, Liu etal. investigated the role of NIN in DOX-induced
toxicity invitro and invivo. For invitro study, they used Hela
cells seeded onto a 96-well plate and then added 20µl of DOX
(1.6 to 10.0µg/mL) and NIN (200 to 2000µmol/L) at different
concentrations. For invivo purpose, they used 32 Hela cells-
bearing athymic nude mice in 4 groups: control; DOX (5mg/
kg); NIN (20mg/kg) and DOX + NIN. DOX and NIN were
administrated with 3-day interval for 5 times. A significant
antitumor efficacy was observed in DOX + NIN group in both
invitro and invivo studies, in addition to significant inhibition
of DOX-induced weight loss and other systemic sides effects
and diminishing hepatocyte necrosis (Liu etal. 2017).
Naringenin (NIGN) has long been considered as an
important ingredient of traditional Chinese medicine. It has
been reported that this flavonoid has exceptional biological
properties ranging from antioxidant to anticancer (Rehman
etal. 2018). Wali etal. investigated the role of naringenin
(50 and 100mg/kg/day p.o. 20 d) on DOX-induced hepato-
toxicity (20mg/kg i.p. on 20th day) after 20days of concomi-
tant use. Naringenin diminished ROS production and ROS-
induced lipid peroxidation and replenished the suppressed
antioxidant armory, marked by catalase, glutathione reduc-
tase, superoxide dismutase, glutathione, and glutathione
peroxidase. Furthermore, naringenin significantly dimin-
ished DOX-induced hepatotoxicity biochemichal (ALT,
AST, ALP, LDH) as well as inflammatory markers (NF-kB,
TNF-α, IL-1β, PGE-2, TGF-β, and IL-6) (Wali etal. 2020).
Its antioxidant and anti-inflammatory effects for prevention
of liver injury should be addressed in future studies, using
lower doses of DOX.
DL‑alpha lipoic acid
Lipoic acid is a naturally-occurring compound which has a
great potential against oxidative metabolism and participates
as protein bound lipoamide in alpha-keto acid dehydrogenase
complexes in mitochondria. It can be converted to a more
active antioxidant molecule, dihydroplipoic acid (Rochette
etal. 2013). In a study by Anandakumar etal., protective
efficacy of DL-alpha lipoic acid (75mg/kg LA single injec-
tion i.p., 24h before DOX) on DOX-induced hepatotoxicity
(15mg/kg i.p.) was investigated in Wistar rats. The adminis-
tration of DOX resulted in an elevation in ALT, AST, ALP,
and LDH level and malondialdehyde, which may be amelio-
rated by DL-alpha lipolic acid (Anandakumar etal. 2007).
Grape seed andskin extract
Grape seed and skin extract (GSSE) is a mixture of various
polyphenolic compounds which is considered to be protec-
tive against DOX-induced cardiotoxicity. The cardioprotective
effects of GSSE have been closely attributed to its iron chelat-
ing property (Quiles etal. 2002). However, there is paucity of
studies regarding its hepatoprotective effects against DOX. In
the study of Monki etal., Wistar rats were treated with GSSE
(500mg/kg/day) for 8days. At the 4th day of treatment, they
received DOX (20mg/kg), and at the 9th day, the rat’s liv-
ers were collected and assessed for oxidative stress status.
According to the results, treatment with DOX alone resulted
in more than 900% increase in MDA and significant increases
in peroxidase and superoxide dismutase activities. However,
GSSE significantly decreased DOX-induced elevation of
serum enzymes (ALT, AST, and bilirubin) and stimulated
lipid peroxidation and the levels of antioxidant markers (GSH,
CAT, SOD, GPx, GR, GST, and G6PD) (Mokni etal. 2015).
Artemisinin
Artemisinin (ATS) is a sesquiterpene lactone which is iso-
lated from Artemisia annua and is known as an antimalarial
agent. However, studies have also demonstrated that this
compound is an effective anti-inflammatory, anti-oxidative,
and anti-cancer agent. Recently, Aktaş etal. evaluated the
efficacy of ATS (7mg/kg/day (ATS7) and 35mg/kg/day
(ATS35) for 14days) on DOX-induced liver damage (10mg/
kg, single dose). DOX, when used alone, resulted in sig-
nificant degeneration of liver and increasing histological
involvement scores and Caspase-3 and TNF-α expression
in hepatocytes. However, treatment with DOX + ATS7 and
DOX + ATS35 led to decrease in the expression of iNOS,
apoptotic markers and NF-kB and diminishment in the liver
damages caused by DOX (Aktaş etal. 2020). So, antioxidant
and anti-apoptotic and anti-inflammatory potentials are key
proposed mechanisms of action.
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Morin
Morin is a flavonoid which is presented in members of the
Moraceae family, and white berry and cranberry branches
are rich in this compound. It inhibits the production of ROS
in high-level glucose-treated rats and inhibits apoptosis
through stimulation of Bcl-2 and antioxidant genes expres-
sion (Kapoor & Kakkar 2012). Recently, Kuzu etal. inves-
tigated the protective effects of Morin (100mg/kg, on the
8th day) against DOX-induced hepatotoxicity (40mg/kg, 10
d) in Wistar albino rats. Adminstration of morin along with
DOX decreased MDA levels in a dose-dependent manner
and significantly increased antioxidant markers (SOD, CAT,
and GPx). Furthermore, this combination had regulatory
effect on TNF-α, IL-1β, and NF-κB levels. DOX + Morin
treatment also significantly increased the expression of
anti-apoptotic marker Bcl-2 and decreased Bax levels dose-
dependently and resulted in significant preservation of liver
structure against DOX damages. So, all three antioxidant,
anti-apoptosis, and anti-inflammatory mechanisms could be
involved in its preventive effects (Kuzu etal. 2019).
Astaxanthin
Astaxanthin (ATX) is a carotenoid which is derived from
Haematococcus pluvialis and has strong antioxidant activi-
ties. ATX structurally can act as a free radical scavenger by
attracting free radicals to its unpaired electrons (Visioli &
Artaria 2017). Ma etal. assessed the hepatoprotective effects
of ATX (50 and 100mg/kg/day, administrated for 3weeks)
against DOX (6mg/kg single i.p.). A significant increase
in body weight and food intake in ATX groups was found
in comparison with the DOX group. Significant preserva-
tion of hepatocytes against DOX-induced injuries was also
observed in ATX group. In addition, significant increase
in anti-apoptotic and antioxidant markers (SOD, CAT and
GPx) was observed in ATX groups (Ma etal. 2020).
Asiatic acid
Asiatic acid (AA) is the effective constituent of Centella
asiatica, which is known because of its anti-cancer, anti-
diabetic, and anti-inflammatory properties (Matsuda etal.
2001). Kamble etal. evaluated the ameliorative effects of
AA (5, 10, and 20mg/kg/day PO, 7d) on DOX-induced car-
diac and hepato-renal toxicities (65.75mg/kg IV, 2d before
the end of the study) with Nrf2 transcriptional factor acti-
vation in Wistar rats. According to their report, significant
DOX-induced upsurge of injury marker enzymes (LDH,
ALT and AST) was observed. However, significant increase
in antioxidant markers (SOD and GSH) was observed in
AA20 group, and it decreased DOX-induced elevation of
LPO and protected liver architecture (Kamble & Patil 2018).
Zataria multiflora andcarvacrol
Zataria multiflora (ZM), which is also known as Avishan
Shirazi, is a member of Lamiaceae family. This plant has dif-
ferent effective ingredients, such as p-cymene (10%), thymol
(16%), and carvacrol (CAR) (52%) (Kavoosi etal. 2012).
ZM is proposed to be effective against various conditions,
including asthma and cancer, and has different pharmaco-
logical potentials such as anti-inflammatory and antioxidant
effects (Silva etal. 2012). Given these properties, Mohebbati
etal. investigated the effects of ZM (200mg/kg/d IV) and
CAR (20mg/kg/d IV) against hepatic injuries caused by
DOX (5mg/kg IV) in Wistar rats for 28days. A significant
increase in body weight of the rats in DOX + CAR group
was found compared to the DOX group. In addition, a sig-
nificant reduction in serum ALP, AST, and ALT levels was
observed in DOX + CAR group. The catalase activity was
increased in both DOX + CAR and DOX + ZM groups, but
no significant differences in SOD activities were observed
among all groups (Mohebbati etal. 2018). It seems that car-
vacrol is the main active compound of ZM for prevention
of DOX induced hepatotoxicity via antioxidant mechanism.
Acacia hydaspica ethyl acetate fraction
Acacia hydaspica R. Parker (AHE) is a member of Legumi-
nosae family, which has important pharmacological activities.
Its bark and seeds are rich in tannins, have strong antioxidant,
anti-inflammatory, and anti-cancer properties, and protect
against cisplatin-induced DNA damage (Afsar etal. 2017).
Afshar etal. evaluated the effect of AHE extract (200 and
400mg/kg/day, 6weeks) on lipid peroxidation, antioxidant
status, and liver function and integrity in doxorubicin (3mg/
kg/week) treated SD rats. Their results demonstrated signifi-
cant amelioration of DOX-induced body weight loss and an
increase in liver weight with AHE in a dose dependent man-
ner. Significant diminishment in ALT, AST, ALP, LDH, and
total and direct BL with AHE was also observed dose depend-
ently. The lipid profile, including total cholesterol, triglyceride
(TG), and low density lipoprotein (LDL)-c decreased with
AHE treatments in both concentrations of AHE, but increase
in HDL was only observed in AHE 400 group. Antioxidant
parameters (POD, SOD, CAT, QR, GSH, GR, GST, GGT, and
GPx) were also decreased with AHE treatments dose depend-
ently. AHE at both doses also significantly decreased liver
injuries, determined by histopathological parameters (Afsar
etal. 2019). So, antioxidant action may be the main mecha-
nism of this herbal compound hepato-protection.
p‑Coumaric acid
p-Coumaric acid (pCA) is a hydroxy derivative of cinnamic
acid, which is presented in a variety of edible plants such
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
as barley grains, peanuts, tomatoes, and garlic. This com-
pound is an antioxidant with anti-inflammatory and antitu-
mor effects and is efficacious against cardiovascular disease
(Walker etal. 2011). Recently, Rafiee etal. assessed the
effects of pCA (100mg/kg/day, 5days) on adriamycin-
induced hepatotoxicity (15mg/kg on the 5th day) in Wistar
rats. pCA pretreatment resulted in significant decrease in
serum ALT, AST, ALP, TG, total cholesterol, LDL-c, and
total BL and an increase in HDL and serum ALB. In addi-
tion, pCA decreased lipid peroxidation (marked by MDA)
and significantly increased antioxidant enzymes activities
(GPx, SOD, and CAT), while prevented DOX-induced liver
histopathological changes (Rafiee etal. 2021). So, antioxi-
dant action may be the main mechanism of this herbal com-
pound hepato-protection.
Ziziphus jujuba Mill
Ziziphus jujuba Mill (ZJ) has been long traditionally used
for biliousness, obesity, cough, headache, and some other
indications. In the last two decades, ZJ has been considered
as a potential treatment of cancer, leukorrhea, tuberculosis,
and cardiovascular disorders (Chen etal. 2017). Also, ZJ can
play an antibacterial, hypotensive, and anti-inflammatory
role. Khajavi Rad etal. investigated the function of a stand-
ardized extract of ZJ against adriamycin-induced liver, heart,
and brain toxicity (20mg/kg PO, thrice daily) in Wistar rats.
Administration of ZJ significantly decreased the biochemical
enzymes (AST, ALT, LDH) and total and direct bilirubin.
Oxidative condition of ZJ treated rats improved considerably
compared to the DOX group (Khajavi Rad 2021).
Thyme oil andthymol
Thymol is a monoterpene phenol which is found in some
plants such as Thymus vulgaris, and it has been reported to
exhibit antioxidant, anti-bacterial, anti-inflammatory, and
anti-apoptosis properties (Tsai etal. 2011, El-Sayed etal.
2015). In recent years, Ahmed etal. study on Wistar rats
demonstrated that the combination of thyme oil (250mg/
kg) or thymol (100mg/kg) every other day for 5weeks
with 2mg/kg DOX once a week for 2weeks could improve
liver structure and integrity, hepatic necrosis, apoptosis,
and fibrosis in treated rats. Moreover, thyme oil and thymol
were effective in reversal of DOX-induced rise in serum
ALT, AST, ALP, total BL, and serum albumin levels. Also,
they observed an increase in serum level of AFP (CA 19.9),
TNFα, and IL-4 in DOX-treated rats which was returned to
normal values in rats treated with thyme oil and thymol. An
assay on oxidant and antioxidant factors revealed that thyme
oil and thymol can ameliorate DOX-induced increase of
LPO and decrease content of GSH and activities of GST and
GPx. Besides, they inhibited apoptosis through increasing
Bcl-2 mRNA expression and decreasing p53 expression
(Ahmed etal. 2022). So, it may show preventive effects via
antioxidant and anti-apoptosis properties.
Mangiferins
Mangiferin is a bioactive extract of an Indonesian mango,
called Mangifera foetida (Shah etal. 2010). This compound
possesses antioxidant and free radical scavenging properties
(Pardo-Andereu etal. 2006). Based on these considerations,
Husna etal. investigated the protective effect of mangiferin
(50 and 100mg/kg PO, 5weeks) on liver damage of DOX
(2.5mg/kg every 2days for 2weeks) in SD rats. Their result
demonstrated that the treatment with mangiferin resulted in
significant reduction of ALT and AST serum levels. Besides,
mangiferin declined lipid peroxidation (marked by MDA
level) and increased content of GSH and SOD activity. So,
it has antioxidant property for preventing hepatotoxicity
(Husna etal. 2022).
Conclusion
The aim of the current study was to appraise the ameliorative
effects of various herbal compounds on the hepatotoxicity
of DOX. Anthracyclines, including doxurobicin, have been
routinely used in treatment of various cancers. However, the
clinical use of these medications is limited because of severe
toxic effects on the various body tissues, including heart,
liver, kidneys, and nervous system particularly in high cumu-
lative dose. The exact mechanism of these severe toxicities of
DOX is not completely understood, but ROS is thought to be
a key factor, interfering in the mitochondrial electron trans-
port chain (ETC) and apoptosis. Besides, abundant evidence
is suggesting the role of inflammation in DOX-induced organ
damage. Considering the abovementioned probable mecha-
nisms of DOX toxicity various invitro and invivo studies
focused on compounds with antioxidant, anti-inflammatory,
and anti-apoptotic properties for prevention of this complica-
tion. In this review, 40 related studies are included, 35 studies
were invivo, two studies were invitro researches (2 stud-
ies both invivo and invitro), and three others were clinical
trials. Most of the studies focused on biochemical (n = 26)
and histopathological (n = 30) aspects of hepatotoxicity and
showed that the assessed compounds considerably improved
these aspects in DOX treated animals.
It is important to mentioned that these compounds mostly
have demonstrated promising efficacy on liver injury his-
topathology (necrosis, fibrosis…) and biochemical factors
(AST, ALT, ALP, LDH…) and reducing oxidative stress
markers (MDA, ROS, CAT, SOD, GSH…), inflammatory
cytokines (IL6, IL-1B, TNFα, …), and apoptotic factors
(caspase3, Bax, Bcl2…) in cellular and animal studies.
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Actually, the most common proposed mechanism of
action for these compounds in reviewed articles was anti-
oxidant effect which was defined in 30 studies. Anti-
inflammatory and anti-apoptotic mechanisms were in
second and third place, proposed in 14 and 10 studies,
respectively, and anti-angiogenic effect was also sug-
gested in one study. Moreover, in 12 studies, significant
increment of animals’ body weight was noteworthy,
and in 4 studies, reduction of mortality was a promis-
ing finding. Antioxidant effect was the common mecha-
nisms defined for all these four compounds; however, the
mechanism should be confirmed in human studies which
is lacking. Further human studies particularly on these
compounds including berberine, silymarin, dioscin and
gingerol could be logical. Only three available clinical
trials assessed silymarin hepatoprotective effect. Two
studies showed its significant preventive effect, and
the other one focused on its therapeutic effect on DOX-
induced liver injury, which was just confirmed based
on ultrasound assessment but not TE or liver enzyme
changes. It is also noticeable that naringin and narin-
genin, beside hepatoprotective effect, showed some anti-
tumor abilities which may be an interesting finding if it
could be proven in future well-designed human studies.
It is noteworthy that some other compounds which are
considered in this review like silymarin and curcumin
also showed promising anti-tumor effects in previous
researches, and considering this evidence beside their
hepatoprotective effects makes them good targets for
future studies in various cancers.
It also should be mentioned that in lots of reviewed
invivo studies (QE, GAP, CH, NIGN, GSSE, morin, AA,
and NM), doxorubicin is administered in considerably
higher than therapeutic dose (if converted to human equiva-
lent dose), and it was much more than LD50 of DOX which
is about 12–15mg/kg for mice (Aston etal. 2017). So, the
reported hepatoprotective efficacy did not make sense in
practice before being confirmed in future studies on DOX
with therapeutic doses.
In conclusion, the promising findings of preclinical
studies should be confirmed in well-designed clinical
trials for better judgment. Actually, invivo studies are
performed under controlled condition on healthy animals.
So, their findings could not be generalized to humans.
Besides, pharmacodynamic and pharmacokinetic interac-
tions between these compounds and chemotherapy regi-
mens should be assessed in human studies. Definition of
the proper safe dose of these compounds for human use is
another challenge. Some of these compounds were previ-
ously used in human for other indications, and their safe
doses are determined; however, for some other agents, this
safe dose should be calculated based on animal adminis-
tered dose which may be difficult.
Author contribution Faezeh Mahmoudi searched the databases and
wrote the manuscript. Sepideh Elyasi defined the manuscript’s subject
and edited the manuscript. Omid Arasteh searched the databases and
edited the manuscript.
Availability of data and materials Not applicable.
Declarations
Ethics approval and consent to participate Not applicable.
Consent for publication All authors approved manuscript in present
format for publication.
Competing interests The authors declare no competing interests.
References
Abbas NAT, Awad MM, Nafea OE (2020) Silymarin in combination
with chlorogenic acid protects against hepatotoxicity induced
by doxorubicin in rats: possible role of adenosine monophos-
phate-activated protein kinase pathway. Toxicol Res (Camb)
9(6):771–777
Afsar T, Razak S, Khan MR, Almajwal A (2017) Acacia hydaspica
ethyl acetate extract protects against cisplatin-induced DNA
damage, oxidative stress and testicular injuries in adult male rats.
BMC Cancer 17(1):883
Afsar T, Razak S, Almajwal A (2019) Effect of Acacia hydaspica R.
Parker extract on lipid peroxidation, antioxidant status, liver func-
tion test and histopathology in doxorubicin treated rats. Lipids
Health Dis 18(1):126
Ahmed MA etal (2013) The protective effect of ginger (Zingiber offici-
nale) against adriamycin-induced hepatotoxicity in rats. Life Sci-
encejournal 10(1):1412–1422
Ahmed O, Abdul-Hamid M, El-bakry A, Mohamed HM, Rahman FE-
ZSA (2019) Camellia sinensis and epicatechin abate doxorubicin-
induced hepatotoxicity in male Wistar rats via their modulatory
effects on oxidative stress, inflammation and apoptosis. J Appl
Pharm Sci 9:30–44
Ahmed OM, Galaly SR, Mostafa M-A, Eed EM, Ali TM, Fahmy
AM, Zaky M (2022) Thyme oil and thymol counter doxorubicin-
induced hepatotoxicity via modulation of inflammation, apoptosis
and oxidative stress. Oxid Med Cell Longev 2022
Aktaş İ, Özmen Ö, Tutun H, Yalçın A, Türk A (2020) Artemisinin
attenuates doxorubicin induced cardiotoxicity and hepatotoxicity
in rats. Biotech Histochem 95(2):121–128
Anandakumar PP, Malarkodi SP, Sivaprasad TR, Saravanan GD
(2007) Antioxidant DL-alpha lipoic acid as an attenuator of adri-
amycin induced hepatotoxicity in rat model. Indian J Exp Biol
45(12):1045–1049
Askarpour GS, Javadinia SA, Shahid S, Fanipakdel A, Elyasi S, Karimi
G (2021) Oral silymarin formulation efficacy in management of
AC-T protocol induced hepatotoxicity in breast cancer patients: a
randomized, triple blind, placebo-controlled clinical trial. J Oncol
Pharm Pract 1–9
Aston WJ, Hope DE, Nowak AK, Robinson BW, Lake RA, Lesterhuis
WJ (2017) A systematic investigation of the maximum tolerated
dose of cytotoxic chemotherapy with and without supportive
care in mice. BMC Cancer 17(1):684. https:// doi. org/ 10. 1186/
s12885- 017- 3677-7
Baptista Moreno Martin AC, Tomasin R, Luna-Dulcey L, Graminha
AE, Araújo Naves M, Teles RHG etal (2020) [10]-Gingerol
improves doxorubicin anticancer activity and decreases its
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
side effects in triple negative breast cancer models. Cell Oncol
(Dordrecht) 43(5):915–29
Barakat BM, Ahmed HI, Bahr HI, Elbahaie AM (2018) Protective
effect of boswellic acids against doxorubicin-induced hepato-
toxicity: impact on Nrf2/HO-1 defense pathway. Oxid Med Cell
Longev 2018:8296451
Chen J, Liu X, Li Z (2017) A review of dietary Ziziphus jujuba Fruit
(Jujube): developing health food supplements for brain protection.
Evid Based Complement Alternat Med 2017:3019568
Cho BJ, Im EK, Kwon JH, Lee KH, Shin HJ, Oh J etal (2005) Berber-
ine inhibits the production of lysophosphatidylcholine-induced
reactive oxygen species and the ERK1/2 pathway in vascular
smooth muscle cells. Mol Cells 20(3):429–434
Dos Santos Arruda F, Tomé FD, Miguel MP, de Menezes LB, Nagib
PRA, Campos EC etal (2019) Doxorubicin-induced cardiotoxicity
and cardioprotective agents: classic and new players in the game.
Curr Pharm Des 25(2):109–118
El-Sayed EM, Abd-Allah AR, Mansour AM, el-Arabey AA (2015)
Thymol and carvacrol prevent cisplatin-induced nephrotoxicity
by abrogation of oxidative stress, inflammation and apoptosis in
rats. J Biochem Mol Toxicol 29(4):165–172
Famurewa AC, Ufebe OG, Egedigwe CA, Nwankwo OE, Obaje GS
(2017) Virgin coconut oil supplementation attenuates acute chem-
otherapy hepatotoxicity induced by anticancer drug methotrexate
via inhibition of oxidative stress in rats. Biomed Pharmacother
87:437–42
Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor
A etal (2021) Cancer statistics for the year 2020: an overview. Int J
Cancer 149(4):778–789
Gillessen A, Schmidt HHJ (2020) Silymarin as supportive treatment
in liver diseases: a narrative review. Adv Ther 37(4):1279–1301
Husna F, Arozal W, Yusuf H, Safarianti S (2022) Protective effect of
mangiferin on doxorubicin-induced liver damage in animal model.
Journal of Research in Pharmacy 26(1):44–51
Jung HA, Kim JI, Choung SY, Choi JS (2014) Protective effect of the
edible brown alga Ecklonia stolonifera on doxorubicin-induced
hepatotoxicity in primary rat hepatocytes. J Pharm Pharmacol
66(8):1180–1188
Kalender Y, Yel M, Kalender S (2005) Doxorubicin hepatotoxicity and
hepatic free radical metabolism in rats. The effects of vitamin E
and catechin. Toxicology 209(1):39–45
Kalyanaraman B (2020) Teaching the basics of the mechanism of
doxorubicin-induced cardiotoxicity: have we been barking up the
wrong tree? Redox Biol 29:101394
Kamble SM, Patil CR (2018) Asiatic acid ameliorates doxorubicin-
induced cardiac and hepato-renal toxicities with Nrf2 transcrip-
tional factor activation in rats. Cardiovasc Toxicol 18(2):131–141
Kapoor R, Kakkar P (2012) Protective role of morin, a flavonoid,
against high glucose induced oxidative stress mediated apoptosis
in primary rat hepatocytes. PLoS ONE 7(8):e41663
Kavoosi G, Teixeira da Silva JA, Saharkhiz MJ (2012) Inhibitory
effects of Zataria multiflora essential oil and its main com-
ponents on nitric oxide and hydrogen peroxide production in
lipopolysaccharide-stimulated macrophages. J Pharm Pharmacol
64(10):1491–500
Khajavi Rad A, Entezari Heravi N (2021) A standardized extract of
Ziziphus jujuba Mill protects against adriamycin-induced liver,
heart, and brain toxicity: an oxidative stress and biochemical
approach. J Food Biochem 45(4):e13698
Khazdair MR, Mhebbati R, Karimi S, Abbasnezhad A, Haghshenas M
(2016) The protective effects of Curcuma longa extract on oxida-
tive stress markers in the liver induced by Adriamycin in rats.
Physiol Pharmacol 20(1):31–37
Kim YC, An RB, Yoon NY, Nam TJ, Choi JS (2005) Hepatoprotec-
tive constituents of the edible brown alga Ecklonia stolonifera on
tacrine-induced cytotoxicity in Hep G2 cells. Arch Pharmacal Res
28(12):1376–1380
Kocahan S, Dogan Z, Erdemli E, Taskin E (2017) Protective effect of
quercetin against oxidative stress-induced toxicity associated with
doxorubicin and cyclophosphamide in rat kidney and liver tissue.
Iran J Kidney Dis 11(2):124–131
Kuzu M, Yıldırım S, Kandemir FM, Küçükler S, Çağlayan C, Türk E
etal (2019) Protective effect of morin on doxorubicin-induced
hepatorenal toxicity in rats. Chem Biol Interact 308:89–100
Lalmuansangi C, Zosangzuali M, Lalremruati M, Tochhawng L, Siama
Z (2020) Evaluation of the protective effects of Ganoderma appla-
natum against doxorubicin-induced toxicity in Dalton's lymphoma
ascites (DLA) bearing mice. Drug Chem Toxicol 1–11
Liu M, Xu Y, Han X, Yin L, Xu L, Qi Y etal (2015) Dioscin alle-
viates alcoholic liver fibrosis by attenuating hepatic stellate cell
activation via the TLR4/MyD88/NF-κB signaling pathway. Sci
Rep 5:18038
Liu X, Yang X, Chen F, Chen D (2017) Combined application of doxo-
rubicin and naringin enhances the antitumor efficiency and attenu-
ates the toxicity of doxorubicin in HeLa cervical cancer cells. Int
J Clin Exp Pathol 10(7):7303–7311
Liu X, Qiu Y, Liu Y, Huang N, Hua C, Wang Q etal (2021) Citronellal
ameliorates doxorubicin-induced hepatotoxicity via antioxidative
stress, antiapoptosis, and proangiogenesis in rats. J Biochem Mol
Toxicol 35(2):e22639
Ma H, Chen S, Xiong H, Wang M, Hang W, Zhu X etal (2020) Asta-
xanthin from Haematococcus pluvialis ameliorates the chemo-
therapeutic drug (doxorubicin) induced liver injury through the
Keap1/Nrf2/HO-1 pathway in mice. Food Funct 11(5):4659–4671
Matsuda H, Morikawa T, Ueda H, Yoshikawa M (2001) Medicinal
foodstuffs. XXVII. Saponin constituents of gotu kola (2): struc-
tures of new ursane- and oleanane-type triterpene oligoglycosides,
centellasaponins B, C, and D, from Centella asiatica cultivated in
Sri Lanka. Chem Pharm Bull 49(10):1368–71
Mohaghegh F, Solhi H, Kazemifar AM (2015) Silymarin (milk thistle)
can revoke liver enzyme changes during chemotherapy of breast
cancer with taxanes. Eur J Integr Med 7(6):650–652
Mohajeri M, Sahebkar A (2017) Protective effect of curcumin against
doxorubicin-induced toxicity and resistance: a review. Crit Rev
Oncol/Hematol 122:30–51
Mohebbati R, Khazdair MR, Karimi S, Abbasnezhad A (2017) Hepato-
protective effects of combination hydroalcoholic extracts of
Nigella sativa and Curcuma longa on Adriamycin induced oxida-
tive stress in rat. J Rep Pharm Sci 6(2):93–102
Mohebbati R, Paseban M, Soukhtanloo M, Jalili-Nik M, Shafei MN,
Yazdi AJ etal (2018) Effects of standardized Zataria multiflora
extract and its major ingredient, carvacrol, on Adriamycin-
induced hepatotoxicity in rat. Biomed J 41(6):340–347
Moher D, Liberati A, Tetzlaff J, Altman DG (2009) Preferred report-
ing items for systematic reviews and meta-analyses: the PRISMA
statement. PLoS Med 6(7):e1000097
Mokni M, Hamlaoui S, Kadri S, Limam F, Amri M, Marzouki L
etal (2015) Efficacy of grape seed and skin extract against dox-
orubicin-induced oxidative stress in rat liver. Pak J Pharm Sci
28(6):1971–1978
Mshemish BAR, Al-Khazragy KA, Al-Nakaash MAI, Abdul FW (2011)
Effect of silymarin against CAF protocol hepatotoxicity. Al-Mustan-
siriyah J Pharm Sci 9:52–62
Nie YC, Wu H, Li PB, Luo YL, Long K, Xie LM etal (2012) Anti-
inflammatory effects of naringin in chronic pulmonary neutro-
philic inflammation in cigarette smoke-exposed rats. J Med Food
15(10):894–900
Pardo-Andereu G, Delgado R, Nunez-Selles AJ, Vercesi AE (2006)
Dual mechanism of mangiferin protection against iron-induced
damageto 2-deoxyribose and ascorbate oxidation. Pharmacol Res
53(3):253–260
Naunyn-Schmiedeberg's Archives of Pharmacology
1 3
Patel N, Joseph C, Corcoran GB, Ray SD (2010) Silymarin modulates
doxorubicin-induced oxidative stress, Bcl-xL and p53 expression
while preventing apoptotic and necrotic cell death in the liver. Toxi-
col Appl Pharmacol 245(2):143–152
Pushpavalli G, Kalaiarasi P, Veeramani C, Pugalendi KV (2010) Effect
of chrysin on hepatoprotective and antioxidant status in D-galac-
tosamine-induced hepatitis in rats. Eur J Pharmacol 631(1–3):36–41
Quiles JL, Huertas JR, Battino M, Mataix J, Ramírez-Tortosa MC
(2002) Antioxidant nutrients and adriamycin toxicity. Toxicology
180(1):79–95
Rafiee Z, Zare Moaiedi M, Valizadeh Gorji A, Mansouri E (2021)
p-Coumaric acid alleviates adriamycin-induced hepatotoxicity in
rats. Asian Pac J Trop Biomed 11:115–121
Rashid S, Ali N, Nafees S, Ahmad ST, Arjumand W, Hasan SK etal
(2013) Alleviation of doxorubicin-induced nephrotoxicity and
hepatotoxicity by chrysin in Wistar rats. Toxicol Mech Methods
23(5):337–345
Rašković A, Stilinović N, Kolarović J, Vasović V, Vukmirović S, Mikov
M (2011) The protective effects of silymarin against doxorubicin-
induced cardiotoxicity and hepatotoxicity in rats. Molecules (Basel,
Switzerland) 16(10):8601–8613
Rehman MU, Rahman Mir MU, Farooq A, Rashid SM, Ahmad B, Bilal
Ahmad S (2018) Naringenin (4,5,7-trihydroxyflavanone) suppresses
the development of precancerous lesions via controlling hyperpro-
liferation and inflammation in the colon of Wistar rats. Environ
Toxicol 33(4):422–35
Rochette L, Ghibu S, Richard C, Zeller M, Cottin Y, Vergely C (2013)
Direct and indirect antioxidant properties of α-lipoic acid and thera-
peutic potential. Mol Nutr Food Res 57(1):114–125
Roomi MW, Kalinovsky T, Roomi NW, Rath M, Niedzwiecki A (2014)
Prevention of Adriamycin-induced hepatic and renal toxicity in male
BALB/c mice by a nutrient mixture. Exp Ther Med 7(4):1040–1044
Sahlan M, Rizka Alia Hapsari N, Diah Pratami K, Cahya Khayrani A,
Lischer K, Alhazmi A etal (2021) Potential hepatoprotective effects
of flavonoids contained in propolis from South Sulawesi against
chemotherapy agents. Saudi J Biol Sci 2021
Sadat-Hoseini SK, DabidiRoshan V (2017) The intractive effects of two
forced and voluntary exercise training method and nanocurcumin
supplement on doxorubicin-induced hepatotoxicity in aging induced
by D-galactose. Tehran Univ Med J 74(11):807–816
Safayhi H, Mack T, Sabieraj J, Anazodo MI, Subramanian LR, Ammon
HP (1992) Boswellic acids: novel, specific, nonredox inhibitors of
5-lipoxygenase. J Pharmacol Exp Ther 261(3):1143–1146
Sak K (2012) Chemotherapy and dietary phytochemical agents. Chem-
other Res Pract. 2012:282570
Sakr SA, Mahran HA, Lamfon HA (2007) Protective effect of ginger
(Zingiber officinale) on adriamycin - induced hepatotoxicity in
albino rats. J Med Plants Res 5:133–140
Semwal RB, Semwal DK, Combrinck S, Viljoen AM (2015) Gingerols
and shogaols: important nutraceutical principles from ginger. Phy-
tochemistry 117:554–568
Shah KA, Patel MB, Patel RJ, Parmar PK (2010) Mangifera indica
(mango). Pharmacogn Rev 4(7):42–48
Shivakumar P, Rani MU, Reddy AG, Anjaneyulu Y (2012) A study on
the toxic effects of doxorubicin on the histology of certain organs.
Toxicol Int 19(3):241–244
Silva FV, Guimarães AG, Silva ER, Sousa-Neto BP, Machado FD,
Quintans-Júnior LJ etal (2012) Anti-inflammatory and anti-ulcer
activities of carvacrol, a monoterpene present in the essential oil of
oregano. J Med Food 15(11):984–991
Somparn N, Kukongviriyapan V, Kukongviriyapan U, Senggunprai L,
Prawan A (2015) Protective effects of tetrahydrocurcumin and cur-
cumin against doxorubicin and cadmium-induced cytotoxicity in
Chang liver cells. Trop J Pharm Res 14(5):769–776
Song S, Chu L, Liang H, Chen J, Liang J, Huang Z etal (2019) Protective
effects of dioscin against doxorubicin-induced hepatotoxicity via
regulation of Sirt1/FOXO1/NF-κb signal. Front Pharmacol 10:1030
Superfin D, Iannucci AA, Davies AM (2007) Commentary: oncologic
drugs in patients with organ dysfunction: a summary. Oncologist
12(9):1070
Thorn CF, Oshiro C, Marsh S, Hernandez-Boussard T, McLeod H, Klein TE
etal (2011) Doxorubicin pathways: pharmacodynamics and adverse
effects. Pharmacogenet Genomics 21(7):440–446
Tsai ML, Lin CC, Lin WC, Yang CH (2011) Antimicrobial, antioxidant and
anti-inflammatory activities of essential oils from five selected herbs.
Biosci Biotechnol Biochem 75(10):1977–1983
Twelves CJ, Dobbs NA, Gillies HC, James CA, Rubens RD, Harper PG
(1998) Doxorubicin pharmacokinetics: the effect of abnormal liver
biochemistry tests. Cancer Chemother Pharmacol 42(3):229–234
Visioli F, Artaria C (2017) Astaxanthin in cardiovascular health and dis-
ease: mechanisms of action, therapeutic merits, and knowledges.
Food Funct 8(1):39–63
Walker JR, Sharma A, Lytwyn M, Bohonis S, Thliveris J, Singal PK etal
(2011) The cardioprotective role of probucol against anthracycline
and trastuzumab-mediated cardiotoxicity. J Am Soc Echocardiogr
24(6):699–705
Wali AF, Rashid S, Rashid SM, Ansari MA, Khan MR, Haq N etal
(2020) Naringenin regulates doxorubicin-induced liver dysfunc-
tion: Impact on oxidative stress and inflammation. Plants (Basel,
Switzerland) 9(4)
Wang G, Zhang J, Liu L, Sharma S, Dong Q (2012) Quercetin potentiates
doxorubicin mediated antitumor effects against liver cancer through
p53/Bcl-xl. PLoS ONE 7(12):e51764
Wang Y, Liu Y, Du X, Ma H, Yao J (2020) The anti-cancer mechanisms
of berberine: a review. Cancer Manag Res 12:695–702
Wei LS, Wee W (2013) Chemical composition and antimicrobial activ-
ity of Cymbopogon nardus citronella essential oil against systemic
bacteria of aquatic animals. Iran J Microbiol 5(2):147–152
Yenny, Girsang E, Nasution AN, Lister INE, Eds (2020) Comparison
hepatoprotective effect of virgin coconut oil and Curcuma longa
Linn against doxorubicin induced hepatotoxicity in Wistar rat. 2020
3rd International Conference on Mechanical, Electronics, Computer,
and Industrial Technology (MECnIT); pp 25–27, June 2020
Zhang Y, Jia J, Ding Y, Ma Y, Shang P, Liu T etal (2016) Alpha-boswel-
lic acid protects against ethanol-induced gastric injury in rats:
involvement of nuclear factor erythroid-2-related factor 2/heme
oxygenase-1 pathway. J Pharm Pharmacol 68(4):514–522
Zhao X, Zhang J, Tong N, Chen Y, Luo Y (2012) Protective effects of ber-
berine on doxorubicin-induced hepatotoxicity in mice. Biol Pharm
Bull 35(5):796–800
Publisher's note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
... The results of the in vivo drug distribution experiment demonstrated predominant accumulation of flavokawain C in the liver tissue of mice following oral administration (Fig. 1A). In additional to targeting tumor tissues, hepatotoxicity poses a significant challenge in the development and utilization of therapy drug for liver cancer (Mahmoudi et al. 2023;Vigano et al. 2023), particularly those derived from herbal products (Lee et al. 2015;He et al. 2019). Accordingly, the potential impact of flavokawain C on liver function was assessed through quantification of serum levels of alanine transaminase (ALT) and aspartate transaminase (AST) (Juan et al. 2023). ...
Article
Full-text available
Purpose This study investigated the potential applicability and the underlying mechanisms of flavokawain C, a natural compound derived from kava extracts, in liver cancer treatment. Methods Drug distribution experiment used to demonstrate the preferential tissues enrichment of flavokawain C. Cell proliferation, apoptosis and migration effect of flavokawain C were determined by MTT, colony formation, EdU staining, cell adhesion, transwell, flow cytometry and western blot assay. The mechanism was explored by comet assay, immunofluorescence assay, RNA-seq-based Kyoto encyclopedia of genes and genomes analysis, molecular dynamics, bioinformatics analysis and western blot assay. The anticancer effect of flavokawain C was further confirmed by xenograft tumor model. Results The studies first demonstrated the preferential enrichment of flavokawain C within liver tissues in vivo. The findings demonstrated that flavokawain C significantly inhibited proliferation and migration of liver cancer cells, induced cellular apoptosis, and triggered intense DNA damage along with strong DNA damage response. The findings from RNA-seq-based KEGG analysis, molecular dynamics, bioinformatics analysis, and western blot assay mechanistically indicated that treatment with flavokawain C notably suppressed the FAK/PI3K/AKT signaling pathway in liver cancer cells. This effect was attributed to the induction of gene changes and the binding of flavokawain C to the ATP sites of FAK and PI3K, resulting in the inhibition of their phosphorylation. Additionally, flavokawain C also displayed the strong capacity to inhibit Huh-7-derived xenograft tumor growth in mice with minimal adverse effects. Conclusions These findings identified that flavokawain C is a promising anticancer agent for liver cancer treatment.
... While N-acetylcysteine remains the mainstay of treatment for acute acetaminophen overdose, significant research efforts continue into the prevention of liver injury using natural products, as recently reviewed by Liao and colleagues [89] and Chilvery et al. [90]. Mahmoudi et al. reviewed the recent literature regarding the use of a variety of natural products in the prevention of liver injury from doxorubicin [91] The interested reader is referred to these and other reviews describing efforts in this particular area. ...
Article
Full-text available
Herbal-induced liver injury (HILI) continues to increase in prevalence each year due to the ongoing popularity of herbal supplements and complementary and alternative medicines. A detailed literature review of case reports and clinical studies published from March 2021 to March 2023 was performed. We discuss the epidemiology and diagnosis of HILI as well as the current and proposed laws and regulations. The 2021 ACG guidelines and 2022 AASLD practice guidelines for the diagnosis and management of drug and herbal-induced liver injury are discussed. We describe updates to previously reported etiologies of HILI such as ayurveda, ashwagandha, turmeric, kratom, green tea extract, and garcinia cambogia. Newly described supplements resulting in HILI, such as tinospora cordifolia, horse chestnut, alkaline water, and more, are described. We discuss newly and previously identified hepatoprotective herbal supplements as they have been reported in the study of animal models and human liver cells. This review suggests the need for ongoing research on the causes and mechanisms of HILI to ensure its proper diagnosis, prevention, and treatment in the future. The goal of this review is to provide novice and expert readers with knowledge regarding the possible etiologies of HILI and a general overview.
... The active ingredients of TCM, including flavonoids, alkaloids, terpenoids, etc., can not only enhance the body's immune function, and induce apoptosis of the tumor cells, but also reverse the multidrug resistance. [37][38][39] Therefore, treating physicians prefer CTX in combination with Chinese herbal medicine to improve the efficacy and reduce the adverse effects in patients with heavy symptom loads. Furthermore, we discovered that the resilience in the moderate class was not as great as that in the low class. ...
Article
Full-text available
Objective This study aims to identify distinct subgroups among gastric cancer patients undergoing chemotherapy (CTX), delineate associated symptom networks, and ascertain the clinical and sociodemographic variables contributing to diverse symptom patterns. Methods Conducted in eastern China, our investigation involved gastric cancer patients receiving CTX. We gathered data using the M.D. Anderson Symptom Inventory Gastrointestinal Cancer Module along with clinical and sociodemographic variables. Subgroups were discerned based on symptom severity through latent profile analysis, and subsequent comparisons were made regarding the symptom networks in different subgroups. Results The analysis encompassed 677 eligible gastric cancer patients, revealing three profiles: “Profile 1: low class” (n = 354, 52.3%), “Profile 2: moderate class” (n = 222, 32.8%), and “Profile 3: all high class” (n = 101, 14.9%). Nausea—vomiting exhibited robust associations in the symptom networks of all subgroups, whereas sadness—distress, and taste change—lack of appetite were notably linked with Profile 1 and Profile 2. Distress emerged as a core symptom in Profile 1, lack of appetite dominated the symptom network in Profile 2, and fatigue attained the highest strength in Profile 3. Distinct symptom profiles were influenced by variables such as education level, CTX combined with surgical or herbal treatment, psychological resilience, and social support. Conclusions Patients within different subgroups manifest individualized patterns of symptom profiles. Analyzing demographics, disease characteristics, and psychosocial information among diverse subgroups facilitates healthcare providers in devising more personalized and targeted symptom management strategies, thereby alleviating the symptom burden on patients.
... Prominent signs of hepatotoxicity are jaundice, exhaustion, abdominal pain, nausea and vomiting, loss of appetite, and dark urine. In some situations, liver poisoning can result in liver failure, which can be fatal (Mahmoudi et al., 2023;Sharma et al., 2019c). As a result, dose recommendations are determined to minimize hepato-toxicity during cancer treatment (King and Perry, 2001). ...
Article
The liver is a well-known player in the metabolism and removal of drugs. Drug metabolizing enzymes in the liver detoxify drugs and xenobiotics, ultimately leading to the acquisition of homeostasis. However, liver toxicity and cell damage are not only related to the nature and dosage of a particular drug but are also influenced by other factors such as aging, immune status, environmental contaminants, microbial metabolites, gender, obesity, and expression of individual genes Furthermore, factors such as drugs, alcohol, and environmental contaminants could induce oxidative stress, thereby impairing the regenerative potential of the liver and causing several diseases. Persons suffering from other ailments and those with comorbidities are found to be more prone to drug-induced toxicities. Moreover, drug composition and drug-drug interactions could further aggravate the risk of drug-induced hepatotoxicity. A plethora of mechanisms are responsible for initiating liver cell damage and further aggravating liver cell injury, followed by impairment of homeostasis, ultimately leading to the generation of reactive oxygen species, immune-suppression, and oxidative stress.
Article
Full-text available
Doxorubicin (DOX) is an effective anticancer agent with a wide spectrum of activities. However, it has many adverse effects on various organs especially on the liver. Thymol, one of the major components of thyme oil, has biological properties that include anti-inflammatory and antioxidant activities. Thus, this study was designed to examine thyme oil and thymol for their ability to prevent doxorubicin-induced hepatotoxicity in Wistar rats. Hepatotoxicity was induced by an intraperitoneal injection of doxorubicin, at a dose of 2 mg/kg bw/week, for seven weeks. Doxorubicin-injected rats were supplemented with thyme oil and thymol at doses 250 and 100 mg/kg bw, respectively, four times/week by oral gavage for the same period. Treatment of rats with thyme oil and thymol reversed the high serum activities of AST, ALT, and ALP and total bilirubin, AFP, and CA19.9 levels, caused by doxorubicin. Thyme oil and thymol also reduced the high levels of TNF-α and the decreased levels of both albumin and IL-4. These agents ameliorated doxorubicin-induced elevation in hepatic lipid peroxidation and associated reduction in GSH content and GST and GPx activities. Further, the supplementation with thyme oil and thymol significantly augmented mRNA expression of the level of antiapoptotic protein Bcl-2 and significantly downregulated nuclear and cytoplasmic levels of the hepatic apoptotic mediator p53. Thus, thyme oil and thymol successfully counteracted doxorubicin-induced experimental hepatotoxicity via their anti-inflammatory, antioxidant, and antiapoptotic properties.
Article
Full-text available
The use of doxorubicin and epirubicin as chemotherapy agent causes side effects such as liver damage due to oxidative stress by reactive oxygen species (ROS) that cause increased of ALT and AST level as liver parameter. One source of natural antioxidants as ROS neutralizer comes from flavonoid that contain in propolis. Most researchers claim that flavonoid can be used to protect the liver. The aim of this study was to test the hepatoprotective effect of flavonoid in propolis from South Sulawesi against doxorubicin and epirubicin. The experiment included male Sprague dawley rats divided into nine groups. The rats received the microcapsule propolis or the quercetin orally for 15 days. The hepatotoxicity was promoted by injection epirubicin and doxorubicin (i.v.) with a cumulative dose of 9 mg/kg. In this study, total polyphenol and flavonoid tests of propolis have been carried out, there were 1.1% polyphenols and 2.7% flavonoids, the antioxidant activity tests showed IC50 value of 9,849 ppm and LCMS/MS tests supported the presence of phenolic compounds in propolis from South Sulawesi. Liver parameter was measured and the results showed that the propolis 200 mg/kg group produced the lowest ALT and had potential protective effect against doxorubicin and epirubicin-induced hepatotoxicity.
Article
Full-text available
Our study briefly reviews the data sources and methods used in compiling the International Agency for Research on Cancer (IARC) GLOBOCAN cancer statistics for the year 2020 and summarises the main results. National estimates were calculated based on the best available data on cancer incidence from population‐based cancer registries (PBCR) and mortality from the World Health Organization mortality database. Cancer incidence and mortality rates for 2020 by sex and age groups were estimated for 38 cancer sites and 185 countries or territories worldwide. There were an estimated 19.3 million (95% uncertainty interval [UI]: 19.0‐19.6 million) new cases of cancer (18.1 million excluding non‐melanoma skin cancer) and almost 10.0 million (95% UI: 9.7‐10.2 million) deaths from cancer (9.9 million excluding non‐melanoma skin cancer) worldwide in 2020. The most commonly diagnosed cancers worldwide were female breast cancer (2.26 million cases), lung (2.21) and prostate cancers (1.41); the most common causes of cancer death were lung (1.79 million deaths), liver (830000) and stomach cancers (769000).
Article
Full-text available
Due to the antioxidant effects of the Ziziphus jujuba Mill (Z. jujuba), we investigated the liver, heart, and brain‐protective effects of this herb against toxicity induced by adriamycin (ADR). In this study, Wistar rats were divided into 1) control, 2) ADR and 3, 4, and 5) treated groups orally administrated three doses of Z. jujuba hydroalcoholic extract for 1 month. The liver, heart, and brain were removed for evaluation of the oxidative markers. Blood samples were evaluated to determine the levels of Lactate dehydrogenase, total and direct bilirubin, alkaline phosphatase, Aspartate transaminase, and Alanine aminotransferase. Administration of Z. jujuba significantly decreased the biochemical enzymes compared to the ADR. Oxidative condition in treated rats with different doses of Z. jujuba was improved compared to the ADR group. Z. jujuba could decrease the oxidative injury through invigoration of the tissues antioxidant system. The mentioned hepatic and cardiac parameters levels improved during extract administration. Practical applications In the first stage, our findings and other supplementary works have shown that administration of jujube extract has prevented the effects of histotoxicity caused by adriamycin, so it seems that in the next stage, the effects of this herbal plant on patients with tissue toxicity caused by adriamycin should be evaluated and if the results are positive in pharmacological studies, it should be used as a complementary drug in the treatment of these patients.
Article
Full-text available
Objective: To evaluate the effect of p-coumaric acid against adriamycin-induced hepatotoxicity in rats. Methods: The rats were divided into 4 groups. The control group received solvent; the p-coumaric acid group was treated with 100 mg/kg of p-coumaric acid orally for five consecutive days; the adriamycin group was administered with a single dose of adriamycin (15 mg/kg, i.p.), and the p-coumaric acid + adriamycin group was given p-coumaric acid five days before adriamycin administration. Twenty-four hours after the last administration, blood samples were collected for biochemical analysis, and liver tissues were removed for histopathological and immunohistochemistrical studies. Moreover, the levels of tissue lipid peroxidation and enzyme activities of glutathione peroxidase, superoxide dismutase, and catalase in liver tissue were measured. Results: Treatment with p-coumaric acid protected the liver from the toxicity of adriamycin by attenuating the increase in alkaline phosphatase, alanine transaminase, aspartate transaminase, total bilirubin, total cholesterol, triglyceride, and low-density lipoprotein cholesterol and lessening the decrease in high-density lipoprotein cholesterol and albumin. p-Coumaric acid also raised the levels of glutathione peroxidase, superoxide dismutase, and catalase, as well as decreased lipid peroxidation in liver tissue and hepatic IL- 1β expression. Additionally, histopathological study confirmed the protective effect of p-coumaric acid against liver damage. Conclusions: p-Coumaric acid can alleviate adriamycin-induced hepatotoxicity.
Article
Breast cancer became the commonest type of cancers among Iraqi women since the last two decades. The main underlying cause is thought to be DNAdamage; much of which is oxidative in nature. CAF protocol (Cyclophosphamide + Adriamycin + 5-FU) associated with toxic effects in several body organs (likeliver), mainly through production of free radicals and reactive oxygen species. Silymarin, the dried extract of a ripe seeds of the plant silybium marianum, wasfound to be a powerful antioxidant agent against toxin -induced tissue damage. Aim of the study is to evaluate the possible time and dose-dependent protective effect of the orally administered silymarin as antioxidant agent against oxidative stress-related hepatotoxicity induced by CAF protocol in breast cancer wome.Seventy four breast cancer women randomly distributed and allocated into three groups:Group (A): 24 patients received CAF protocol by I.V infusion once every 21 days and for 63 days.Group (B): 25 patients received 210mg/day of silymarin along with the same CAF protocol of group (A).Group (C): 25 patients received 420mg/day of silymarin along with the same CAF protocol of group (A).Indices of liver function (AST, ALT,TSB) were measured at baseline, after 21, 42, and 63 days of treatment. The levels of AST and ALT, which were significantly elevated due to CAF protocol therapy, showed significant reduction when silymarin used with CAFprotocol, in a time and dose-dependent manner. Mean while, TSB levels significantly reduced by CAF protocol but dose not show any significant changeafter treatment with silymarin.Use of antioxidant agent (silymarin) in this study can ameliorate, in a time and dose-dependant manner, liver damage that induced by oxidative stress.Keywords: Breast cancer, CAF protocol (cyclophosphamide+Adriamycin+5- fluorouracil), hepatotoxicity, silymarin.
Article
Background Chemotherapeutic agents, with or without other drugs and radiation, may cause indirect or direct hepatotoxicity. Doxorubicin-induced hepatotoxicity (DIH) is a major health concern in cancer patients receiving this cytotoxic drug that is mostly resulted from the production of reactive oxygen species leading to transient or permanent liver damages. Silymarin, a flavonoid extracted from the Silybum marianum, exhibits antioxidant and anti-inflammatory activities. Purpose This study aimed to investigate the clinical efficacy of systemic administration of silymarin in management of chemotherapy induced hepatotoxicity in patients with non-metastatic breast cancer who received doxorubicin/cyclophosphamide-paclitaxel (AC-T) regimen. Material: In this randomized, triple blind, placebo-controlled clinical trial, 30 patients who received AC-T who fulfilled the inclusion criteria were randomly allocated to silymarin (n = 15) or placebo (n = 15) groups to receive oral silymarin 140 mg three times a day or placebo tablets, respectively. Fatty liver severity was assessed by liver ultrasound imaging and FibroScan® and also measurement of liver function tests before and after the intervention. Results There was a non-significant trend toward more severe liver involvement in placebo group comparing to the silymarin group after intervention based on ultrasonography (p = 0.083). Besides, in silymarin group, hepatic involvement grade based on ultrasonography considerably reduced after intervention (p = 0.012). However, no difference was found between two groups based on FibroScan and liver function tests. Conclusion Oral administration of silymarin could significantly reduce hepatotoxicity severity after 1 month of treatment in non-metastatic breast cancer patients treated with AC-T regimen.
Article
Many xenobiotics are known to cause hepatic damage with subsequent significant morbidity and mortality. Doxorubicin (DOX) is a broad-spectrum antineoplastic agent. DOX is reported to cause hepatocellular damage. Previous studies verified the promising role of many natural antioxidant products against various models of hepatic dysfunction. We conducted this study to evaluate the possible hepatoprotective effect of silymarin (SILY) and/or chlorogenic acid (CGA) in a rat model of DOX-induced hepatotoxicity. For this purpose, we randomly divided 30 adult male rats into five equal groups as control, DOX, co-treated DOX with SILY, co-treated DOX with GCA and co-treated DOX with SILY and CGA groups. All treatments were administered every second day for 4 weeks. Our results showed that simultaneous SILY and CGA administration caused a significant decrease in hepatic apoptosis biomarkers (hepatic caspase-3 and nuclear factor-κB levels), a significant improvement in hepatic oxidant/antioxidant status (malondialdehyde and superoxide dismutase) and significant decrease in hepatic pro-inflammatory biomarkers (tumor necrosis factor-alpha and interlukin-1β) compared with DOX treatment. We concluded that adding CGA to SILY acts as a hepatoprotective agent against DOX-induced liver injury through inhibiting apoptosis biomarkers, maintaining antioxidant enzyme levels, decreasing pro-inflammatory cytokines as well as regulating liver adenosine monophosphate-activated protein kinase signaling.