Article 270
Article 270
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-
proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug
resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical
path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with
anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin,
ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic
acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline,
Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated
pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in
combined therapy and immunomodulation. In addition, the present review has extended to describe other promis-
ing compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and
cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodula-
tory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and
further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as
modulators of immune checkpoints.
Keywords: Cancer, Chinese herbal medicine, Natural products, Bioactive compounds, Traditional Chinese medicine
© The Author(s) 2019. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium,
provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license,
and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/
publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Luo et al. Chin Med (2019) 14:48 Page 2 of 58
(NF-κB), signal transducer and activator of transcrip- secretion from activated T cells, and enhances T cell-
tion and cell cycle-related proteins [36–46]. Curcumin is induced cytotoxicity in human esophageal adenocarci-
shown to induce anti-cancer activities through the dis- noma OE33 and OE19 cells, so it increases the sensitivity
ruption of mitochondrial membrane potential and block- of the cells to T cell-induced cytotoxicity [51]. The natu-
ade at G2/M phase of the cell cycle in human epidermoid ral killing (NK) cells can directly kill cancer cells, and
carcinoma A-431 cells [47]. In addition, mammalian tar- curcumin can enhance the cytotoxicity effect of NK cells
get of rapamycin (mTOR) plays a vital role in curcumin- when NK cells are co-cultured with human breast cancer
induced autophagy and apoptosis [30, 48–50]. Curcumin MDA-MB-231 cells, which is highly associated with sig-
induces apoptosis and autophagy through the inhibition nal transducer and activator of transcription 4 (STAT4)
of phosphoinositide 3-kinase (PI3K)/Akt/mTOR path- and signal transducer and activator of transcription 5
way in human NSCLC A549 cells [30], while it induces (STAT5) activation [52]. Besides, myeloid-derived sup-
autophagy by reducing Akt phosphorylation and mTOR pressor cells (MDSCs) are immune-suppressive cells
in human melanoma A375 and C8161 cells [49]. which are found in most cancer patients. Curcumin
Curcumin can also exert immunomodulatory effects decreases interleukin (IL)-6 levels in the tumor tissues
against cancer cells. Theracurmin, a highly bioavailable and serum of Lewis lung carcinoma (LLC)-bearing mice
form of curcumin, decreases pro-inflammatory cytokine to impair the growth of MDSCs, so targeting MDSCs is
Luo et al. Chin Med (2019) 14:48 Page 4 of 58
important for the treatment of lung cancer [13]. More- theracurmin every day with standard gemcitabine-
over, the anti-tumor immune response of curcumin is based chemotherapy. No new adverse effects and no
mediated through increased cluster of differentiation increase in the incidence of adverse effects were observed
(CD)8+ T cell population and decreased regulatory T among these patients. A pilot phase II study demon-
cell (Treg) population in tongue squamous cell carcinoma strated encouraging results for the combination of
[53–55]. docetaxel/prednisone and curcumin in patients with cas-
In order to overcome the solubility issues of curcumin tration-resistant prostate cancer. It was found that 59% of
and facilitate its intracellular delivery, a curcumin-loaded patients had prostate-specific antigen response and 40%
nanoparticle, curcumin-PLGA-NP, is synthesized. It has of patients achieved partial response. This study has pro-
a tenfold increase in water solubility compared to cur- vided additional evidence for a high response rate and
cumin, and shows threefold increased anti-cancer activi- better tolerability with the use of curcumin during cancer
ties in human breast cancer MDA-MB-231 and NSCLC therapy [77].
A549 cells [56]. Another curcumin-capped nanoparti-
cle exhibits promising anti-oxidative and selective anti-
cancer activities in human colorectal cancer HT-29 and Epigallocatechin gallate (EGCG)
SW-948 cells [57]. Moreover, a curcumin analog, WZ35, EGCG, also known as epigallocatechin-3-gallate (Fig. 2),
has high chemical stability, and higher efficacy in anti- is the main polyphenol in green tea (Camellia sinensis).
cancer effects compared to curcumin in human gastric Epidemiological studies have indicated that consumption
cancer SGC-7901 cells and SGC-7901 xenograft mice of green tea has potential impact of reducing the risk of
[20]. Another analog, B63, induces cell death and reduces many chronic diseases, such as cardiovascular diseases
tumor growth through ROS and caspase-independent and cancer [78, 79]. EGCG possesses various biological
paraptosis in human gastric cancer SGC-7901, BGC-823 effects including anti-obesity and anti-hyperuricemia,
and SNU-216 cells, 5-fluorouracil-resistant gastric cancer anti-oxidative, anti-viral, anti-bacterial, anti-infective,
cells, and SGC-7901 xenograft mice [58]. anti-angiogenic, anti-inflammatory and anti-cancer activ-
Curcumin can be used with other chemotherapeu- ities [80–84]. It is reported to present anti-cancer effects
tic agents to achieve synergistic effects, reduce adverse in variety of cancer cells, including lung, colorectal, pros-
effects and enhance sensitivity. Tamoxifen and curcumin tate, stomach, liver, cervical, breast, leukemia, gastric,
are packed into a diblocknanopolymer, and this nanopo- bladder cancers [85–90]. Among its anti-cancer activi-
lymer reduces the toxicity of tamoxifen in normal cells ties, EGCG exhibits multiple pharmacological actions,
and exhibits better anti-proliferative and pro-apoptotic including the suppression of cell growth, proliferation,
effects in human breast cancer tamoxifen-sensitive and metastasis and angiogenesis, induction of apoptosis, and
-resistant MCF-7 cells [59]. Triptolide has strong liver enhancement of anti-cancer immunity [85, 86, 91–94].
and kidney toxicities, and when combined with cur- EGCG can inhibit cell proliferation through multiple
cumin, they exert synergistic anti-cancer effects in ovar- ways in many types of cancer cells. It inhibits cell pro-
ian cancer, as well as reduce the side effects of triptolide liferation in human bladder cancer SW-780, breast can-
[60]. In addition, adriamycin, sildenafil, 5-fluorouracil, cer MDA-MB-231 and NSCLC A549 cells, and inhibits
irinotecan, doxorubicin, paclitaxel, sorafenib, Kruppel- tumor growth in gastric cancer SGC-7901 xenograft
like factor 4, emodin, docosahexaene acid and apigenin mice [89, 94, 95]. It also induces apoptosis in human oral
are shown to exhibit synergistic effects with curcumin cancer KB, head and neck cancer FaDu, NSCLC A549,
[61–71]. Similarly, copper supplementation significantly and breast cancer MCF-7 cells [96, 97]. Besides, EGCG
enhances the anti-tumor effects of curcumin in several induces autophagy, and inhibition of autophagy can
oral cancer cells [72], while epigallocatechin-3-gallic acid enhance EGCG-induced cell death in human mesothe-
ester (EGCG) increases the ability of curcumin to inhibit limoa ACC-meso, Y-meso, EHMES-10, EHMES-1 and
cell growth and induce apoptosis in human uterine leio- MSTO-211H, and primary effusion lymphoma BCBL-1
myosarcoma SKN cells [73]. and BC-1 cells [98, 99]. In contrast, it induces cell death
Clinical trials can confirm or reveal the effects, adverse via apoptosis and autophagy in oral squamous cell carci-
reactions and pharmacokinetics of the drugs. As the bio- noma SCC-4 cells [84], so autophagy plays a dual role in
availability of curcumin is very poor, many curcumin EGCG-induced cell death. It can also suppress metastasis
preparations are synthesized and tested in clinical tri- in human melanoma SK-MEL-5, SK-MEL-28, A375 and
als [74–76]. A phase I study was conducted to investi- G361, NSCLC CL1-5, A549 and H1299 cells, and lung
gate the safety and pharmacokinetics of theracurmin in metastasis mice [85, 93, 100]. In addition, EGCG sup-
pancreatic and biliary tract cancer patients who failed presses tumor angiogenesis in human NSCLC A549 cells
with standard chemotherapy [76]. They administered and A549 xenograft mice [101].
Luo et al. Chin Med (2019) 14:48 Page 5 of 58
EGCG mediates apoptosis which involves pro- and xenograft mice [111]. Besides, EGCG-DHA (docosahex-
anti-apoptotic proteins in various cancer cells. It up-reg- aenoic) ester, a lipophilic derivative of EGCG, shows
ulates pro-apoptotic proteins such as Bcl-2-associated X improved anti-oxidative effects compared to EGCG,
protein (Bax), and down-regulates anti-apoptotic pro- and suppresses colon carcinogenesis in mice [112, 113].
teins including B-cell lymphoma 2 (Bcl-2), B-cell lym- In the last decade, many studies were carried out using
phoma-extra large (Bcl-xL) and survivin [97, 102–104]. EGCG-loaded nanoparticles including FA-NPS-PEG and
ER stress also plays an important role in EGCG-induced FA-PEG-NPS (epigallocatechin gallate-β-lactoglobulin
cell death. EGCG inhibits endoplasmic reticulum (ER) nanoparticles), EGCG-SLN (solid lipid nanoparticle),
stress-induced protein kinase R-like endoplasmic reticu- DT-EGCG-nanoethosomes, FCS-EGCG-NPs (chi-
lum kinase (PERK) and eukaryotic translation-initiation tosan coated nanoparticles), EGCG-dispersed selenium
factor 2α (eIF2α) phosphorylation [105]. Besides, poly nanoparticles, 198AuNP-EGCg (gold nanoparticles),
(ADP-ribose) polymerase (PARP) 16 is shown to activate EGCG-loaded microspheres (EGCG/MS), and FCMPs
ER stress markers, PERK and inositol-requiring enzyme (ferritin-chitosan Maillard reaction products) [6, 110,
1α (IRE1α) [106]. ER stress-induced apoptosis, PERK 114–121]. These EGCG nanoparticles can improve the
and eIF2α phosphorylation by EGCG are suppressed in targeting ability and efficacy of EGCG, which greatly pro-
PARP16-deficient hepatocellular carcinoma QGY-7703 mote the clinical application and development of EGCG
cells, so EGCG mediates apoptosis through ER stress, analogs.
which is dependent on PARP16 [105]. Similarly, EGCG EGCG antagonizes toxicity induced by anti-cancer
causes 78-kDa glucose-regulated protein (GRP78) accu- chemotherapeutic agents, and sensitizes chemo-resist-
mulation in the ER, which up-regulates ER stress markers ant cancer cells. It also exerts synergistic effects with
such as activating transcription factor 4 (ATF-4), X-box anti-cancer agents in various cancer cells, such as cis-
binding protein 1 (XBP-1) and C/EBP homologous pro- platin, oxaliplatin, temozolomide, resveratrol, doxoru-
tein (CHOP), and shifts into pro-apoptotic ER stress, bicin, vardenafil, curcumin, erlotinib [122–129]. EGCG
leading to increased caspase-3 and -8 activities [107]. can enhance the sensitivity of cisplatin through copper
Furthermore, it suppresses cell migration and invasion transporter 1 (CTR1) up-regulation, which results in the
by blocking tumor necrosis factor (TNF) receptor-associ- accumulation of cellular cisplatin and cisplatin–DNA
ated factor 6 (TRAF6), MMP-2/c-Jun N-terminal kinase adducts in human ovarian cancer SKOV3 and OVCAR3
(JNK) and transforming growth factor-β (TGF-β) path- cells, and the combination of EGCG and cisplatin sup-
ways [85, 93, 100]. presses tumor growth in OVCAR3 xenograft mice [122].
In addition to anti-cancer effects, EGCG shows a sig- The combined low concentration of EGCG and curcumin
nificant inhibitory effect on interferon-γ (IFN-γ)-induced remarkably inhibits cell and tumor growth in human
indoleamine 2,3-dioxygenase (IDO) expression, an NSCLC A549 and NCI-H460 cells, and A549 xenograft
enzyme that guides cancer to regulate immune response, mice through cell cycle arrest [123].
in human colorectal cancer SW-837 cells [108], so this To evaluate the tolerance, safety, pharmacokinetics and
suggests that EGCG might be useful for chemopreven- efficacy of EGCG in humans, clinical trials have been or
tion and colorectal cancer treatment, and could be a are currently being conducted for cancer treatment. Dur-
potential agent for anti-tumor immunotherapy. EGCG is ing a phase I clinical trial for the treatment of radiation
also found to be a potential immune checkpoint inhibitor, dermatitis, patients with breast cancer received adjuvant
which down-regulates IFN-γ-induced B7 homolog 1 (B7- radiotherapy and EGCG solution. It was found that the
H1) levels, an immunoglobulin-like immune suppressive maximum dose (660 μM) of EGCG was well tolerated and
molecule, in human NSCLC A549 cells [109]. the maximum tolerated dose was undetermined [130].
Although EGCG has numerous biological activities It was concluded that EGCG was effective for treating
through different pathways, its efficacy demonstrated in radiation dermatitis. Moreover, a phase II clinical trial
in vivo studies is not always consistent with the results was conducted to investigate the benefits of EGCG as a
of in vitro studies. This can be due to its low oil solubil- treatment for acute radiation-induced esophagitis (ARIE)
ity, metabolic instability and poor bioavailability [110]. for patients with stage III lung cancer. The oral admin-
Therefore, EGCG analogs and EGCG-loaded nanopar- istration of EGCG was shown to be effective and phase
ticles by modifying EGCG are developed, and they have III clinical trial to study the potential effects of EGCG to
been reported to enhance anti-cancer effects [111–113]. ARIE treatment was anticipated [131].
The peracetate-protected (−)-EGCG, a prodrug of EGCG
obtained by modifying the reactive hydroxyl groups with Berberine
peracetate groups, is shown to increase the bioavailability Berberine (Fig. 2) is an isoquinoline alkaloid mainly
of EGCG and inhibit angiogenesis in endometrial cancer extracted from medicinal plants such as Coptidis
Luo et al. Chin Med (2019) 14:48 Page 6 of 58
chinensis Franch., Mahonia bealei (Fort.) Carr., and Phel- berberine, including down-regulation of cyclins A, D,
lodendron chinense Schneid. [132]. Berberine has diverse cyclin-dependent kinase (CDK) 1, CDK4, MMP-2 and
pharmacological effects and is normally used for the janus kinase 2 (Jak2)/vascular endothelial growth factor
treatment of gastroenteritis [133, 134]. It exhibits sig- (VEGF)/NF-κB/activator protein 1 (AP-1) pathway, and
nificant anti-cancer effects in a wide spectrum of cancers induction of autophagic cell death via mTOR signaling
including ovarian, breast, esophageal, and thyroid can- pathway [149, 155, 156]. Berberine also induces mito-
cers, leukemia, multiple myeloma, nasopharyngeal car- chondrial-mediated apoptosis through the loss of mito-
cinoma, and neuroblastoma, through inducing cell cycle chondrial membrane potential, cytochrome c release,
arrest and apoptosis, inhibiting metastasis and angiogen- caspase and PARP activation, up-regulation of pro-apop-
esis [135–143]. totic Bcl-2 family proteins, and down-regulation of anti-
Berberine can induce cell cycle arrest in various can- apoptotic Bcl-2 family proteins [150, 157–159]. It can
cer cells [137, 144, 145]. Berberine induces G1 and G2/M also activate apoptosis-inducing factor to induce ROS-
phase arrest in murine prostate cancer RM-1 cells, and mediated cell death in pancreatic, breast, and colon can-
G1 cell arrest by regulating cyclins D1 and E expressions cers [158, 160, 161].
in human HER2-overexpressed breast cancer cells [144, Immunotherapy has made great progress to cancer
145]. However, berberine induces G1 phase arrest in treatment over the past few years. Toll-like receptors
human estrogen receptor positive breast cancer MCF-7 (TLRs) can activate innate immune responses for host
cells but not in estrogen receptor negative MDA-MB-231 defense [162]. Berberine inhibits proto-oncogene tyros-
cells [137]. Besides, it inhibits cell proliferation by induc- ine kinase Src activation and TLR4-mediated chemotaxis
ing apoptosis in human colorectal cancer HCT-8 cells in lipopolysaccharide (LPS)-induced macrophages [163].
[146]. In p53-null leukemia EU-4 cells, berberine induces Besides, IDO1 inhibitors are promising candidates for
p53-independent and X-linked inhibitor of apoptosis cancer immunotherapy [164]. Berberine and its deriva-
protein (XIAP)-mediated apoptosis, which is associated tives are shown to exhibit anti-cancer activity through
with mouse double minute 2 homolog (MDM2) and pro- cell killing by NK cells via IDO1 [165]. IL-8 is associated
teasomal degradation [135]. Mitochondrial-mediated with metastasis, and berberine decreases IL-8 levels to
apoptosis with Bcl-2-like protein 11 (Bim) up-regulation inhibit cell growth and invasion in triple-negative breast
and Forkhead box O (FoxO) nuclear retention is vital cancer cells [166].
in berberine-induced apoptosis [147]. In addition, ber- Berberine has low oral bioavailability as well as poor
berine can induce autophagic cancer cell death through intestinal absorption [167]. As it has pronounced anti-
increased GRP78 levels and enhancing the binding ability microbial activity against gut microbiota, high dosage can
of GRP78 to VPS34 in human colorectal cancer HCT-116 translates into adverse events [168]. This limits the clini-
cells [148], whilst it induces autophagy through inhibiting cal use of berberine, and different approaches have been
AMPK/mTOR/UNC-51-like kinase 1 (ULK-1) pathway applied to improve the bioavailability of berberine. d-α-
in human glioma U251 and U87 cells [149]. In contrast, Tocopheryl polyethylene glycol 1000 succinate enhances
berberine induces protective autophagy in human the intestinal absorption of berberine by inhibiting P-gp
malignant pleural mesothelioma NCI-H2452 cells, and activity in rats [167]. A self-microemulsifying drug deliv-
inhibition of autophagy promotes berberine-induced ery system is developed to improve the bioavailability
apoptosis [150]. Therefore, autophagy plays a dual role of berberine, the bioavailability is increased by 2.42-fold
in berberine-induced apoptosis. Furthermore, berberine [169]. Ber8, a 9-alkylated derivative of berberine, has bet-
also inhibits tumor migration and invasion [143, 151]. It ter cytotoxicity and cellular uptake than berberine, and
up-regulates plasminogen activator inhibitor-1 (PAI-1), further inhibits cell proliferation and induces cell cycle
a tumor suppressor that down-regulates urokinase-type arrest in different cell lines, including SiHa, HL-60, and
plasminogen activator (uPA) and antagonizes uPA recep- A549 cells [170].
tor to suppress metastasis in human hepatocellular car- The combination of berberine and chemo- or radio-
cinoma Bel-7402 and SMMC-7721 cells [143]. Berberine therapies provides synergistic anti-cancer effects [171,
also inhibits epithelial mesenchymal transition through 172]. Taxol combined with berberine significantly slows
PI3K/Akt pathway in murine melanoma B16 cells, [151], down cell growth in human epidermal growth factor
and suppresses angiogenesis in glioblastoma U87 xeno- receptor 2 (HER2)-overexpressed breast cancer cells
graft mice and HUVECs [152, 153]. [145], while the combined administration of berberine
Berberine interacts with diverse molecular targets and caffeine enhances cell death through apoptosis and
as it binds to nucleic acids via specific deoxyribonu- necroptosis in human ovarian cancer OVCAR3 cells
cleic acid (DNA) sequences [154]. Several mechanisms [173]. The combination therapy of berberine and nira-
have been identified for the anti-proliferative effects of parib, a PARP inhibitor, markedly enhances apoptosis
Luo et al. Chin Med (2019) 14:48 Page 7 of 58
and inhibits tumor growth in ovarian cancer A2780 KKU-452, KKU-023 and KKU-100, and tongue squa-
xenograft mice [174]. Therefore, combination of berber- mous cell carcinoma Cal-27 cells [190, 198, 199], while
ine with other therapies is a promising treatment for the ART induces autophagy-mediated cell cycle arrest in
alternative cancer therapy. human ovarian cancer SKOV3 cells [200]. DHA is also
Previous pre-clinical research and animal studies shown to induce autophagy by suppressing NF-κB acti-
have demonstrated the anti-tumor action of berberine vation in several cancer cells including RPMI 8226, NB4,
hydrochloride. The people with a history of colorectal HCT-116, and HeLa cells [202]. Furthermore, ART and
cancer might be at higher risk for adenomas, thus they DHA can also inhibit metastasis in various cancer cells
are particularly suitable for the study of the chemopre- such as non-small-cell lung carcinoma (NSCLC), ovar-
ventive effects of berberine hydrochloride in adenomas. ian and lung cancer cells [184, 189, 203]. Apart from
A randomized, double-blind, placebo-controlled trial apoptosis and metastasis, the inhibition of angiogen-
was designed to determine whether the daily intake of esis is also a crucial approach in cancer treatment. ART
300 mg of berberine hydrochloride could decrease the inhibits angiogenesis through mitogen-activated protein
occurrence of new colorectal adenomas in patients with kinase (MAPK) activation in osteosarcoma [204], whilst
a history of colorectal cancer, and it is currently ongoing. DHA exerts strong anti-angiogenic effect by repressing
Another phase II clinical trial of berberine and gefitinib is extracellular signal–regulated kinase (ERK) and NF-κB
also ongoing in patients with advanced NSCLC and acti- pathways in human umbilical vein endothelial cells
vating EGFR mutations. (HUVECs) and pancreatic cancer, respectively [194, 195].
In the past decades, studies have been focused on stud-
Artemisinins ying the anti-cancer mechanisms of ARTs, but there are
Artemisinin (Fig. 2) is a sesquiterpene peroxide derived contentions. ARTs inhibit cancer cell proliferation mainly
from annual wormwood (Artemisia annua L.), which by the induction of apoptosis through mitochondrial-
was originally used as Traditional Chinese Medicine for dependent pathways [196, 205, 206]. ART mediates the
treating malaria and related symptoms such as fever and release of cytochrome c and caspase-9 cleavage, leading
chills [175]. Since the 2015 Nobel Prize in Physiology to increased apoptosis in human breast cancer MCF-7
or Medicine conferred to Chinese scientist, Youyou Tu, cells [196]. DHA induces apoptosis through Bcl-2 down-
artemisinin drew attention to worldwide [176]. Beside regulation in human cervical cancer HeLa and Caski
from their well-established anti-malarial effects, arte- cells [205], and via Bim-dependent intrinsic pathway in
misinin and its derivatives (ARTs), including dihydroar- human hepatocellular carcinoma HepG2 and Huh7 cells
temisinin (DHA), artesunate, artemether and arteether, [206]. Interestingly, ART is demonstrated to be an inhibi-
are also found to exhibit potent anti-cancer activities in tor of anti-cancer target, histone deacetylases (HDAC)
many studies [177–182]. DHA and artesunate are the [196]. In addition, another mechanism of killing tumor
most studied ART derivatives for cancer treatment, and cells by ARTs is iron-dependent cell death called ferrop-
artesunate will be discussed in a separate section. The tosis, a new form of cell death, so ferroptosis becomes an
anti-cancer effects of ARTs are demonstrated in a broad attractive strategy for cancer treatment [183, 207].
spectrum of cancer cells including lung, liver, pancreatic, DHA can enhance the anti-tumor cytolytic activity
colorectal, esophageal, breast, ovarian, cervical, head and of γδ T cells against human pancreatic cancer SW1990,
neck, and prostate cancers [183–191]. The anti-cancer BxPC-3 and Panc-1 cells [208], and ART also potenti-
activities of ARTs include induction of apoptosis and cell ates the cytotoxicity of NK cells to mediate anti-tumor
cycle arrest, inhibition of cell proliferation and growth, activity [209]. Similarly, ART inhibits tumor growth
metastasis and angiogenesis [189, 192–195]. through T cell activation and T reg suppression in breast
ART inhibits cell proliferation, migration and invasion, cancer 4T1 xenograft mice [188]. Therefore, this pro-
and induces apoptosis in human breast cancer MCF-7 vides a novel strategy for treating pancreatic cancer with
cells [193, 196], while DHA suppresses cell growth immunotherapy.
through cell cycle arrest and apoptosis in human hepa- ART has poor water solubility and bioavailability.
tocellular carcinoma HepG2 cells and HepG2 xenograft In order to solve this issue, ART is encapsulated into
mice [178]. Similarly, ART induces apoptosis in murine micelles by nanoprecipitation to form ART-loaded
mastocytome P815 cells and hamster kidney adenocarci- micelles [210]. The ART-loaded micelles enhance the
noma BSR cells, and inhibits tumor growth in P815 xeno- drug exposure time and accumulation in breast can-
graft mice [177]. Moreover, autophagy plays a vital role cer 4T1 xenograft mice, and shows specific toxicity in
in ART-mediated anti-cancer activities [190, 197–201]. human and murine breast cancer MCF-7 and 4T1 cells.
DHA can induce autophagy-dependent cell death in A mitochondrial-targeting analog of ART is also synthe-
human cervical cancer HeLa cells, cholangiocarcinoma sized to specifically target mitochondria for enhancing
Luo et al. Chin Med (2019) 14:48 Page 8 of 58
the inhibition of cell proliferation in various cancer cells 240], and inhibits cell migration in human colorectal can-
including HCT-116, MDA-MB-231, HeLa and SKBR3 cer LoVo, SW-620 and HCT-116 cells [240]. Ginsenoside
cells [211]. Moreover, dimmers of ART are also synthe- Rg3 can also modulate the tumor environment through
sized by polyamine linkers, and they further inhibit cell inhibiting angiogenesis and enhancing anti-tumor
proliferation in human breast cancer MCF-7 cells and immune responses [241]. Moreover, ginsenoside Rh2
angiogenesis in HUVECs [212]. exhibits anti-tumor activity in human NSCLC H1299
Many studies show the synergistic effects of ARTs cells and H1299 xenograft mice, through the induction
with other compounds or therapeutic approaches. The of ROS-mediated ER-stress-dependent apoptosis [242].
combined treatment of ART and resveratrol markedly It also suppresses cell proliferation and migration, and
inhibits cell proliferation and migration, and enhances induces cell cycle arrest in human hepatocellular car-
apoptosis and ROS production in human cervical can- cinoma HepG2 and Hep3B cells, and inhibits tumor
cer HeLa and hepatocellular carcinoma HepG2 cells growth in HepG2 xenograft mice [243]. Compound K,
[213]. Similarly, the use of combined DHA and gemcit- an intestinal bacterial metabolite of ginsenosides, also
abine exhibits strong synergistic effects on the loss of induces cell cycle arrest and apoptosis in human colorec-
mitochondrial membrane potential and induction of tal cancer HCT-116 cells, and suppresses tumor growth
apoptosis in human NSCLC A549 cells [214]. DHA also in HCT-116 xenograft mice [244]. It also efficiently
reinforces the anti-cancer activity of chemotherapeu- inhibits cell proliferation and induces apoptosis through
tic agent, cisplatin, in cisplatin-resistant ovarian cancer mitochondrial-related pathways in human hepatocel-
cells [215]. Studies also demonstrate the enhancement of lular carcinoma MHCC97-H cells [245]. Furthermore,
sensitivity by DHA in photodynamic therapy in esopha- 20(S)-ginsenoside Rg3 induces autophagy to mediate cell
geal cancer [182, 216]. Therefore, this suggests that ARTs migration and invasion in human ovarian cancer SKOV3
could be potential anti-cancer agents. cells [246]. In contrast, it sensitizes NSCLC cells to ico-
The population pharmacokinetic properties of DHA tinib and hepatocellular carcinoma cells to doxorubicin
were investigated using the plasma and saliva of breast through the inhibition of autophagy [247, 248]. Besides,
cancer patients for long-term treatment (> 3 weeks) ginsenoside Rh2 inhibits cell growth partially through the
[217]. The salivary DHA concentration was proportion- coordination of autophagy and β-cateninin signaling in
ally correlated with the plasma DHA concentration, so human heptocellular carcinoma HepG2 and Huh7 cells
saliva is a good use for monitoring DHA levels in the [249]. Compound K induces autophagy-mediated apop-
body. An artemisinin analog, Artenimol-R, was shown tosis through AMPK/mTOR and JNK pathways in human
to improve clinical symptoms and tolerability in patients NSCLC A549 and H1975 cells [250], while it also induces
with advanced cervical cancer [218]. autophagy and apoptosis through ROS and JNK path-
ways in human colorectal cancer HCT-116 cells [251].
Ginsenosides Therefore, autophagy plays a dual role in cancer via dif-
Ginsenosides (Fig. 2) are the main bioactive dammarane ferent signaling routes.
triterpenoids derived from the rhizomes of many plants In recent years, the potential anti-cancer mechanisms
including Panax notoginseng (Burk.) F. H. Chen, Panax of ginsenoside Rg3 have been demonstrated in various
ginseng and Cinnamomum cassia Presl., with various cancer models, which include the inhibition of cell pro-
biological effects including anti-oxidative, anti-inflamma- liferation and induction of apoptosis via down-regulating
tory, and anti-cancer activities [219–222]. Ginsenosides PI3K/Akt, and activation of caspase-3 and -9 and Bcl-2
mainly exert anti-cancer effects in colorectal, breast, liver family proteins [234, 252], induction of cell cycle arrest
and lung cancers, through inhibiting cell proliferation by regulating CDK pathway [240], inhibition of metas-
and migration, angiogenesis, and reversing drug resist- tasis through reducing the expressions of aquaporin
ance [7, 223–230]. Ginsenoside Rg3, ginsenoside Rh2, 1, C–X–C chemokine receptor type 4 (CXCR4) and
and compound K are the primary bioactive compounds hypoxia-inducible factor 1α (HIF-1α) [253–255]. More-
among ginsenosides for cancer prevention. over, 20(S)-ginsenoside Rh2 is shown to bind to recom-
Ginsenoside Rg3 inhibits cell viability and induces cell binant and intracellular annexin A2 directly, and this
apoptosis in human ovarian cancer HO8910 cells [231], inhibits the interaction between annexin A2 and NF-κB
hepatocellular carcinoma Hep1-6, HepG2 and SMMC- p50 subunit, which decreases NF-κB activation [256].
7721, breast cancer MCF-7, MDA-MB-231, MDA- NF-κB is important in cell survival, and 20(S)-ginseno-
MB-453 and BT-549, and NSCLC A549, H23 and Lewis side Rh2 can inhibit cell survival through NF-κB pathway.
lung carcinoma cells [232–238]. It induces cell cycle Furthermore, p53 also plays a vital role in ginsenoside-
arrest at G1 phase in human melanoma A375, and mul- induced anti-cancer activities [244, 257, 258]. Ginse-
tiple myeloma U266, RPMI 8226 and SKO-007 cells [239, noside Rh2 induces cell death through p53 activation
Luo et al. Chin Med (2019) 14:48 Page 9 of 58
in human colorectal cancer HCT-116 and SW-480 cells (TACE) were studied in patients with advanced hepa-
[257], while ginsenoside Rg3 and compound K induces tocellular carcinoma. The results showed that this ther-
apoptosis and cell cycle arrest through p53/p21 up-regu- apy ameliorated TACE-induced adverse effects and
lation in human colorectal cancer HCT-116, SW-480 and prolonged the overall survival compared to the use of
HT-29, and gallbladder cancer NOZ and GBC-SD cells, TACE alone [264].
respectively [244, 258].
For the promotion of immunity, ginsenoside Rg3 can Ursolic acid (UA)
enhance lymphocyte proliferation and T helper type 1 As an ursane-type pentacyclic triterpenic acid, UA
cell (Th1)-related cytokine secretion including IL-2 and (Fig. 2) can be found in the berries and leaves of a series
IFN-γ in hepatacellular carcinoma H22-bearing mice, of natural medicinal plants, including Vaccinium mac-
and inhibit tumor growth partly through the induc- rocarpon Ait. (cranberry), Arctostaphylos uva-ursi (L.)
tion this cellular immunity [259]. Ginsenoside Rg3 can Spreng (bearberry), Rhododendron hymenanthes Makino,
also down-regulate the levels of B7-H1 and B7 homolog Eriobotrya japonica, Rosemarinus officinalis, Calluna
3 (B7-H3), immunoglobulin-like immune suppressive vulgaris, Eugenia jambolana and Ocimum sanctum, as
molecules, to modulate tumor microenvironment and well as in the wax-like protective coatings of fruits such
enhance anti-tumor immunity, and these molecules are as pears, apples and prunes [265]. UA has numerous
negatively associated with overall survival in colorectal biochemical and pharmacological effects including anti-
cancer patients [241]. It also ameliorates cisplatin resist- inflammatory, anti-oxidative, anti-proliferative, anti-ath-
ance by down-regulating B7-H1 levels and resuming T erosclerotic, anti-leukemic, anti-viral, and anti-diabetic
cell cytotoxicity in human NSCLC A549 and A549/DDP effects [266–272]. It also exerts anti-cancer activities in
cells [260]. In addition, ginsenoside Rh2 can also enhance ovarian, breast, gastric, prostate, lung, liver, bladder, pan-
anti-tumor immunity in melanoma mice by promoting creatic, and colorectal cancers [273–281].
T cell infiltration in the tumor and cytotoxicity in spleen UA can be used as a potential therapeutic agent for
lymphocytes [261]. the treatment of various cancers [281–289]. It induces
The combination of ginsenosides with other chemo- apoptosis through both extrinsic death receptor and
therapeutic agents provides significant advantages for mitochondrial death pathways in human breast cancer
cancer treatment. Ginsenoside Rg3 alone demonstrates MDA-MB-231 cells [289], and inhibits cell prolifera-
modest anti-angiogenic effects, and displays additive tion and induces pro-apoptosis in human breast cancer
anti-angiogenic effects in B6 glioblastoma rats when MCF-7 cells by FoxM1 inhibition [282]. UA also inhib-
combined with temozolomide [262]. When it is com- its cell and tumor growth through suppressing NF-κB
bined with paclitaxel, it enhances cytotoxicity and apop- and STAT3 pathways in human prostate cancer DU-145
tosis through NF-κB inhibition in human triple-negative and LNCaP cells, and DU-145 xenograft mice [283], and
breast cancer MDA-MB-231, MDA-MB-453 and BT-549 induces apoptosis in human prostate cancer PC-3 cells
cells [233]. [284]. Similarly, UA induces apoptosis and inhibits cell
Ginsenosides have a long history of use as traditional proliferation in human colorectal cancer HCT-15, HCT-
medicine to treat many diseases in China. Relatively few 116, HT-29 and Caco-2 cells [286, 287]. UA is also shown
clinical studies have been performed in humans even- to induce autophagy to mediate cell death in murine
though ginseng products are widely recognized to have cervical cancer TC-1 cells [290], and promote cytotoxic
therapeutic effects when used alone or in combination autophagy and apoptosis in human breast cancer MCF-7,
with other chemotherapeutic agents. Therefore, clinical MD-MB-231 and SKBR3 cells [291]. It also inhibits cell
studies are needed to confirm the safety of such uses. A growth by inducing autophagy and apoptosis in human
phase II clinical trial is conducting to assess the safety breast cancer cells T47D, MCF-7 and MD-MB-231 cells
and efficacy of ginsenoside Rg3 in combination with first- [279]. In contrast, UA induces autophagy, but the inhi-
line chemotherapy in advanced gastric cancer. Patients bition of autophagy enhances UA-induced apoptosis in
with advanced NSCLC and epidermal growth factor human oral cancer Ca922 and SCC2095, and prostate
receptor-tyrosine kinase inhibitor (EGFR-TKI) muta- cancer PC-3 cells [265, 292]. Therefore, autophagy plays a
tion were recruited in a study that investigated the safety dual role in UA-induced apoptosis via different signaling
and efficacy of the combined therapy, ginsenoside Rg3 pathways. In addition, UA inhibits tumor angiogenesis
and EGFR-TKI. It was shown that this therapy increased through mitochondrial-dependent pathway in Ehrlich
progression-free survival, overall survival and objective ascites carcinoma xenograft mice [293].
response rate compared to EGFR-TKI alone [263]. In Increasing evidence has linked the anti-cancer activi-
another study, the safety and efficacy of combined ginse- ties of UA to the activation of mitochondrial-dependent
noside Rg3 and transcatheter arterial chemoembolization signaling pathways, including mitochondrial energy
Luo et al. Chin Med (2019) 14:48 Page 10 of 58
metabolism, oxidative stress and p53‑mediated mito- A human clinical study was conducted to investigate
chondrial pathways [289, 291, 293]. UA is demonstrated the toxicity and pharmacokinetics of UA-liposomes
to have apoptosis-promoting and anti-proliferative (UAL) including dose-limiting toxicity and maximum
capacities via modulating the expressions of mitochon- tolerated dose in healthy adult volunteers and patients
drial-related proteins such as Bax, Bcl-2, cytochrome with advanced solid tumors [309]. UAL had manageable
c and caspase-9 [289, 293]. It can also induce oxidative toxicities under the dose of 98 mg/m2, as well as a lin-
stress and disruption of mitochondrial membrane per- ear pharmacokinetic profile, so it was suggested that UA
meability to mediate apoptosis in human osteosarcoma could be developed as a potential and safe drug [309].
MG63 and cervical cancer HeLa cells [294, 295]. In
addition, p53 pathway also contributes to the anti-can- Silibinin
cer effects of UA. UA induces apoptosis and cell arrest Silibinin (Fig. 2), one of the flavonoids isolated from Sily-
through p21-mediated p53 activation in human colorec- bum marianum L. Gaertn, is commonly exploited for the
tal cancer SW-480 and breast cancer MCF-7 cells [296, treatment hepatic diseases in China, Germany and Japan.
297], and this p53 activation is through inhibiting nega- In addition, silibinin is also found to display various
tive regulators of p53, MDM2 and T-LAK cell-originated biological activities including anti-oxidative, anti‑pro-
protein kinase (TOPK) [297]. liferative, anti-bacterial, anti-fungal, neuro-protective,
Studies have reported the cancer immunomodulatory anti-leishmanial, anti-osteoclastic and anti-metastatic
activities of UA [279, 293]. UA down-regulates NF-κB to activities [310–317]. Previous studies have reported that
inhibit cell growth and suppress inflammatory cytokine silibinin exerts remarkable effects in numerous cancers
levels including TNF-α, IL-6, IL-1β, IL-18 and IFN-γ in such as renal, hepatocellular and pancreatic carcinoma,
human breast cancer T47D, MCF-7 and MDA-MB-231 bladder, breast, colorectal, ovarian, lung, salivary gland,
cells [279]. It also modulates the tumor environment prostate and gastric cancers, through the induction of
by modulating cytokine production such as TNF-α and apoptosis, inhibition of tumor growth, metastasis and
IL-12 in ascites Ehrlich tumor [293]. angiogenesis [318–328].
UA is insoluble in water, with poor pharmacokinetic Silibinin suppresses epidermal growth factor-induced
properties including poor oral bioavailability, low dis- cell adhesion, migration and oncogenic transformation
solution and weak membrane permeability [298]. Some through blocking STAT3 phosphorylation in triple nega-
new drug delivery technologies have been developed to tive breast cancer cells [329]. It strongly suppresses cell
overcome these problems including the uses of liposomes proliferation and induces apoptosis in human pancreatic
[280, 299–302], solid dispersions [303], niossomal gels cancer AsPC-1, BxPC-3 and Panc-1 cells, and induces
[304], and nanoliposomes [278]. Liposome is the most cell cycle arrest at G1 phase in AsPC-1 cells [330]. It can
commonly used drug delivery system. A chitosan-coated also induce apoptosis via non-steroidal anti-inflamma-
UA liposome is synthesized with tumor targeting and tory drug-activated gene-1 (NAG-1) up-regulation in
drug controlled release properties, and has fewer side human colorectal cancer HT-29 cells [331], and induces
effects [302]. It enhances the inhibition of cell prolifera- mitochondrial dysfunction to mediate apoptosis in
tion and tumor growth in human cervical cancer HeLa human breast cancer MCF-7 and MDA-MB-123 cells
cells and U14 xenograft mice. Besides, a pH-sensitive [332]. Moreover, silibinin induces autophagic cell death
pro-drug delivery system is also synthesized, and this via ROS-dependent mitochondrial dysfunction in human
pro-drug enhances cellular uptake and bioavailability of breast cancer MCF-7 cells [333]. In contrast, it induces
UA [305]. It further inhibits cell proliferation, cell cycle autophagy to exert protective effect against apoptosis in
arrest and induces apoptosis in human hepatocellular human epidermoid carcinoma A-431, glioblastoma A172
carcinoma HepG2 cells. and SR, and breast cancer MCF-7 cells [334–336], and
UA can also be used in combination with other drugs. autophagy inhibition enhances silibinin-induced apop-
The combined treatment of zoledronic acid and UA tosis in human prostate cancer PC-3 cells [337]. Silibinin
enhances the induction of apoptosis and inhibition of cell also induces autophagy to inhibit metastasis in human
proliferation through oxidative stress and autophagy in renal carcinoma ACHN and 786-O cells, and salivary
human osteosarcoma U2OS and MG63 cells [306], whilst gland adenoid cystic carcinoma cells [317]. Therefore,
the combination of UA and curcumin inhibits tumor autophagy plays a dual role in silibinin-induced anti-
growth compared to UA alone in skin cancer mice [307]. cancer effects. In addition, silibinin inhibits angiogenesis
Moreover, UA combined with doxorubicin enhances the in human prostate cancer PCa, LNCaP and 22Rv1 cells
cellular uptake of doxorubicin, and reverses multi-drug [327].
resistance (MDR) in human breast cancer MCF-7/ADR Silibinin exhibits anti-cancer activities mainly due to
cells [308]. the cell cycle arrest [330, 338–341]. It induces G1 phase
Luo et al. Chin Med (2019) 14:48 Page 11 of 58
arrest in human pancreatic cancer SW1990 and AsPC- Silibinin has been widely used as anti-cancer drug
1, and breast cancer MCF-7 and MCF-10A cells [330, in vitro and in vivo, and its combination with other thera-
339, 340], whilst it causes G2 phase arrest in human pies is a promising treatment for cancer, so clinical trials
cervical cancer HeLa, and gastric cancer MGC-803 and are needed to confirm its safety and efficacy in humans,
SGC-7901 cells [338, 341]. It also decreases the expres- and to develop as an anti-cancer drug.
sions of CDKs such as CDK1, CDK2, CDK4 and CDK6
that are involved in G1 and G2 progression [338, 339]. Emodin
Besides, silibinin suppresses metastasis through ERK1/2 Emodin (Fig. 2) is an anthraquinone derivative isolated
and MMP-9 down-regulation in human thyroid cancer from many plants including Rheum palmatum, Polygo-
TPC-1, breast cancer MCF-7, renal carcinoma ACHN, num cuspidatum, Polygonum multiflorum, and Cassia
OS-RC-2 and SW-839, and epidermoid carcinoma A-431 obtusifolia. It exhibits remarkable biological effects such
cells [342–344]. In addition, silibinin induces apopto- as anti-inflammation, anti-oxidant, prevention of intra-
sis and inhibits proliferation through the suppression hepatic fat accumulation and DNA damage [360–366].
of NF-κB activation [345–348]. On the other hand, sili- Many studies have shown that emodin can attenuate
binin is shown to induce apoptosis through the promo- numerous cancers including nasopharyngeal, gall blad-
tion of mitochondrial dysfunction, including increased der, lung, liver, colorectal, oral, ovarian, bladder, pros-
cytochrome c and Bcl-2 levels, the loss of mitochondrial tate, breast, stomach and pancreatic cancers, through the
membrane potential, and decreased adenosine triphos- inhibition of cell proliferation and growth, metastasis,
phate (ATP) levels [332, 333, 349, 350]. angiogenesis, and induction of apoptosis [367–379].
Silibinin has immunomodulatory effects in cancer and Emodin suppresses ATP-induced cell proliferation and
immunity. The MDSCs are associated with immunosup- migration through inhibiting NF-κB activation in human
pression in cancer, and silibinin increases the survival NSCLC A549 cells [380], and induces apoptosis through
rate in breast cancer 4T1 xenograft mice, and reduces cell cycle arrest and ROS production in human hepato-
the population of MDSCs in their blood and tumor [351]. cellular carcinoma HepaRG cells [381]. It also induces
There was also a reduction in macrophage infiltration autophagy to mediate apoptosis through ROS production
and neutrophil population in silibinin-treated prostate in human colorectal cancer HCT-116 cells [382]. Moreo-
cancer TRAMPC1 xenograft mice [352]. These studies ver, emodin can inhibit tumor growth and metastasis in
suggest a role of immunity in its anti-tumor effects. triple negative breast cancer cells, and human colorectal
Silibinin has poor water solubility and bioavailabil- cancer HCT-116 cells [383, 384], whilst it suppresses cell
ity, so it limits its efficacy in anti-cancer activities [353]. migration and invasion through microRNA-1271 up-reg-
Advanced technologies such as nanoprecipitation tech- ulation in human pancreatic cancer SW1990 cells [385].
It also down-regulates CXCR4 to suppress C–X–C motif Various effects have been disclosed as key contribu-
chemokine 12 (CXCL-12)-induced cell migration and tions to the anti-cancer effects of triptolide. Triptolide
invasion in hepatocellular carcinoma HepG2 and HepG3 is shown to exhibit pro-apoptosis effects in various
cells [395]. In addition, emodin inhibits the differentia- cancers [427–431]. It induces mitochondrial apoptotic
tion of maturation of DCs [396], and can modulate mac- pathway to mediate apoptosis in Burkitt’s lumphoma
rophage polarization to restore macrophage homeostasis Raji, NAMALWA and Daudi cells, and inhibits tumor
[397]. growth in Daudi xenograft mice [432], and inhibits cell
Aloe-emodin is a derivate of emodin, which exhib- proliferation through microRNA-181a up-regulation
its superior bioactivities in some cancers. It can inhibit in human neuroblastoma SH-SY5Y cells [433]. Moreo-
cell proliferation through caspase-3 and caspase-9 acti- ver, triptolide induces autophagy to induce apoptosis
vation in human oral squamous cell carcinoma SCC-15 and inhibit angiogenesis in human osteosarcoma MG63
cells [398], and induce apoptosis in human cervical can- cells, and breast cancer MCF-7 cells [431, 434]. In con-
cer HeLa and SiHa cells, which is associated with glucose trast, triptolide induces protective autophagy through
metabolism [399]. Another derivative of emodin, rhein, calcium (Ca2+)/calmodulin-dependent protein kinase
can also induce apoptosis in human pancreatic cancer kinase β (CaMKKβ)-AMPK pathway in human prostate
Panc-1 cells, and inhibit tumor growth in pancreatic can- cancer PC-3, LNCaP and C4-2 cells, and through Akt/
cer xenograft mice [400]. It also inhibits cell migration mTOR down-regulation in human cervical SiHa cells
and invasion through regulating Rac1/ROS/MAPK/AP-1 [420, 435]. Therefore, autophagy plays a dual role in
signaling pathway in human ovarian cancer SKOV3-PM4 triptolide-induced anti-cancer effects. In addition, trip-
cells [401]. tolide is able to inhibit cell migration and invasion in
The combination of emodin and other chemotherapies human prostate cancer PC-3 and DU-145 cells, and in
is widely used for cancer treatment. Emodin can pro- tongue squamous cell carcinoma SAS cells co-inoculated
mote the anti-tumor effects of gemcitabine in pancreatic with human monocytes U937 cells [417, 419]. Further-
cancer [402–404]. It enhances apoptosis in human pan- more, triptolide also possesses anti-angiogenic effect by
creatic cancer SW1990 cells, and further inhibits tumor inhibiting VEGFA expression in human breast cancer
growth in SW1990 xenograft mice, through suppressing MDA-MB-231 and Hs578T cells, and through COX-2
NF-κB pathway [402, 403]. The combination of emodin and VEGF down-regulation in human pancreatic cancer
and curcumin can also enhance the inhibition of cell pro- Panc-1 cells [436, 437].
liferation, survival, and invasion in human breast can- Triptolide is a natural substance, which exerts its anti-
cer MDA-MB-231, MDA-MB-435 and 184A1 cells [64]. cancer effects through multiple targets. Triptolide is
Moreover, emodin enhances cisplatin-induced cytotoxic- shown to induce mitochondrial-mediated apoptosis in
ity through ROS production and multi-drug resistance- various cancer cells, through decreased mitochondrial
associated protein 1 (MRP1) down-regulation in human membrane potential, Bax and cytochrome c accumula-
bladder cancer T24 and J82 cells [405]. tion, PARP and caspase-3 activation, decreased ATP lev-
Emodin has been shown to have remarkable anti-can- els, and Bcl-2 down-regulation [432, 438–441]. Moreover,
cer effects in vitro and in vivo, and its combination with ERK is also shown to be important in mediating trip-
other therapies is very effective in treating cancer, there- tolide-induced anti-cancer activities. Triptolide induces
fore it is important to evaluate the safety and efficacy of apoptosis through ERK activation in human breast can-
emodin as an anti-cancer drug as the next step. cer MDA-MB-231 and MCF-7 cells [434, 442], and ERK
activation leads to caspase activation, Bax up-regulation
Triptolide and Bcl-xL down-regulation [442]. On the other hand,
Triptolide (Fig. 2) is a natural constituent derived from it can also inhibit metastasis through ERK down-reg-
the root of a traditional Chinese medicine, Tripterygium ulation in esophageal squamous cell cancer KYSE180
wilfordii Hook. F., which possesses diverse effects includ- and KYSE150 cells, and murine melanoma B16F10 cells
ing anti-inflammatory, anti-oxidative, and anti-cancer [443, 444]. Interestingly, ERα is shown to be a potential
activities [60, 406, 407]. For cancer therapy, it has been binding protein of triptolide and its analogues [445]. In
used to treat breast, lung, bladder, liver, colorectal, pan- addition, triptolide-induced metastasis is shown to be
creatic, ovarian, stomach, prostate, cervical, and oral through MMP-2 and MMP-9 down-regulation in human
cancers, melanoma, myeloma, leukemia, neuroblastoma, neuroblastoma SH-SY5Y cells, via decreased MMP-3 and
osteosarcoma, lymphoma, renal, nasopharyngeal, and MMP-9 expressions in T-cell lymphoblastic lymphoma
endometrial carcinoma, through apoptosis, cell cycle cells, and through MMP-2, MMP-7 and MMP-9 down-
arrest, inhibition of cell proliferation, metastasis and regulation in human prostate cancer PC-3 and DU-145
angiogenesis [406, 408–426]. cells [417, 423, 433].
Luo et al. Chin Med (2019) 14:48 Page 13 of 58
Indeed, immunology has been frequently validated of its derivatives and analogs have been used in clini-
to be associated with cancer. The combined use of trip- cal studies to test the safety and efficacy on anti-cancer
tolide and cisplatin enhances the plasma levels of IL-2 effects [432, 455–457]. Omtriptolide, a derivative of trip-
and TNF-α in ovarian cancer SKOV3/DDP xenograft tolide, is highly water soluble, and a phase I clinical trial
mice, which can promote the differentiation of T cells was conducted in Europe with patients who had refrac-
and inhibit tumorigenesis respectively, thus resulting in tory and relapsed acute leukemia [432]. Another phase
an inflammatory microenvironment and leading to can- I clinical trial was completed in patients with refractory
cer cell death [446]. gastrointestinal malignancies to study the dose escalation
The derivatives of triptolide are always needed to and pharmacokinectics of minnelide, a pro-drug of trip-
improve its ant-cancer therapy. Triptolide derivative, tolide [457]. The doses used were 0.16 to 0.8 mg/m2 and
MRx102, shows positive effects on anti-proliferation they were well tolerated except from the common hema-
and anti-metastasis through Wnt inhibition in human tologic toxicity. LLDT-8, another triptolide derivative,
NSCLC H460 and A549 cells, and H460 xenograft mice has anti-cancer and immunosuppressive effects, and is
[447]. Minnelide, a water-soluble pro-drug of triptolide, going to proceed into phase II clinical trial to test its anti-
can inhibit tumor growth in pancreatic cancer MIA cancer effects in China [455, 456]. Moreover, minnelide is
PaCa-2 xenograft mice. Meanwhile, the combination of currently under phase II clinical trial to test anti-cancer
minnelide and oxaliplatin further inhibits tumor growth effects in patients with advanced pancreatic cancer [458].
[448]. Moreover, triptolide is poorly soluble in water
and exhibits hepatotoxicity and nephrotoxicity, selective Cucurbitacins
delivery is an effective strategy for further application in Cucurbitacins (Fig. 2) is a cluster of tetracyclic triter-
cancer treatment. Triptolide loaded onto a peptide frag- penoids originated from various plants like Bryonia,
ment (TPS-PF-A299–585) is specifically targeted to the Cucumis, Cucurbita and Lepidium sativum. Cucurbi-
kidney and with less toxicity [449]. Some modified trip- tacins A–T are twelve main curcurbitacins belonging
tolide-loaded liposomes are reported to contribute a tar- to this family. Cucurbitacins have multiple therapeutic
geted delivery with lower toxicity and better efficacy in effects such as anti-inflammation, anti-proliferation, anti-
lung cancer treatment [450]. Similarly, triptolide-loaded angiogenesis, and anti-cancer [452, 459–462]. Besides,
exosomes enhances apoptosis in human ovarian cancer cucurbitacins have also been elucidated as a potential
SKOV3 cells [451]. candidate for various cancer therapies, including oral cell
Triptolide has some side effects in various organs carcinoma, breast, ovarian, prostate, lung, gastric, blad-
because of excessive dosage, so researchers have been der, and thyroid cancers, neuroastoma, hepatoma, and
looking for alternative triptolide therapies, and combina- osteosarcoma [463–475]. Most of cucurbitacins have
tion therapy has become a hot spot. Triptolide combined been reported with various anti-cancer activities, such as
with gemcitabine markedly enhances pro-apoptosis pro-apoptosis, anti-angiogenesis, autophagy induction,
through Akt/glycogen synthase kinase 3β (GSK3β) path- and inhibition of metastasis [452, 460–462, 476].
way in human bladder cancer EJ and UMUC3 cells Cucurbitacin B is the most abundant source of cucur-
[452]. Triptolide plus ionizing radiation synergistically bitacins which can explain why it receives more atten-
enhances apoptosis and anti-angiogenic effects through tion from researchers than other cucurbitacins do. It
NF-κB p65 down-regulation in human nasopharyngeal suppresses cell proliferation and enhances apoptosis in
carcinoma cells and xenograft mice, which provides a human NSCLC A549 cells, colorectal cancer SW-480 and
new chemotherapy to advanced nasopharyngeal malig- Caco-2 cells [462, 477], and induces G1 phase cell cycle
nancy [425]. The combined therapy of triptolide and arrest in human colorectal cancer SW-480 and Caco-
5-fluorouracil further promotes apoptosis and inhib- 2, and gastric cancer MKN45 cells [477, 478]. Cucurbi-
its tumor growth through down-regulating vimentin tacin D inhibits cell survival in human gastric cancer
in human pancreatic cancer AsPC-1 cells and AsPC-1 AGS, SNU1 and Hs746T cells [479], while cucurbitacin E
xenograft mice [453]. Besides, low concentration of trip- induces cell cycle arrest at G2/M phase in triple negative
tolide potentiates cisplatin-induced apoptosis in human breast cancer cells [480]. Moreover, cucurbitacins B, E
lung cancer HTB-182, A549 and CRL-5810 and CRL- and I are shown to induce autophagy, however inhibition
5922 cells [454], and triptolide with cisplatin synergisti- of autophagy can enhance cucurbitacin-induced apopto-
cally enhances apoptosis and induces cell cycle arrest in sis [481–483]. They also inhibit cell migration and inva-
human bladder cancer cisplatin-resistant cells [409]. sion in human breast cancer MDA-MB-231 and SKBR3,
Triptolide has wide-spectrum activities in pre-clinical NSCLC H2030-BrM3 and PC9-BrM3, and colorectal
studies, but it has strong side effects and water insolu- cancer COLO-205 cells [484–487], as well as angiogen-
bility, so it is not used in clinical studies. However, some esis in HUVECs [461, 488].
Luo et al. Chin Med (2019) 14:48 Page 14 of 58
Various targets have been demonstrated to be respon- nanomicelles show a higher bioavailability and better
sible for the anti-cancer effects of cucurbitacins. STAT3 tumor inhibition [497].
signaling is a very common target for cancer treatment. For a better cancer therapy, some combinations
Cucurbitacins B and D are reported to inhibit prolifera- between cucurbitacins and other drugs have been
tion and induce apoptosis through STAT3 suppression employed. Low doses of cucurbitacin B or methotrexate
in human NSCLC A549 cells and doxorubicin-resistant cannot inhibit tumor growth in osteosarcoma xenograft
breast cancer MCF-7/ADR cells, respectively [462, 489]. mice, however when combined together, they synergisti-
On the other hand, cucurbitacin E induces cell arrest and cally inhibit tumor growth [498]. The combination ther-
apoptosis via STAT3 inhibition in human breast cancer apy of cucurbitacin B and curcumin enhances apoptosis
Bcap-37 and MDA-MB-231 cells [468], and cucurbi- and reverses MDR in human hepatocellular carcinoma
tacin I can inhibit STAT3 pathway to suppress cancer Bel-7402/5-Fu cells [499]. Recently, cucurbitacin B is sug-
stem cell properties in anaplastic thyroid cancer ATC– gested to be a potential candidate when it is applied with
CD133+ cells [463]. Besides, cucurbitacin E induces cell withanone, this combination can enhance cytotoxicity in
cycle arrest through cyclins B1 and D1 down-regulation human NSCLC A549 cells, and inhibit tumor growth and
[480, 490], while cucurbitacin D inhibits cyclin B expres- metastasis in A549 xenograft mice [500]. Cucurbitacin
sion [491]. Moreover, mitochondria and ER stress also I is also shown to be a STAT3 inhibitor to mediate cell
play an important role in cucurbitacin-induced anti- survival and proliferation, and when it is combined with
cancer effects. Cucurbitacins mediate apoptosis through irinotecan, and they further inhibit cell proliferation in
mitochondrial-related pathway, which is characterized human colorectal cancer SW-620 and LS174T cells [501].
by the loss of the mitochondrial membrane potential, The derivatives of cucurbitacins, cucurbitacin
Bcl-2 down-regulation, Bax up-regulation, cytochrome c B-nanomicelles, and the combination therapies show
release, that eventually leads to caspase activation [470, promising treatment for cancer in vitro and in vivo, so
492]. Cucurbitacin I induces cell death through ER stress, clinical trials are needed to confirm their safety and effi-
by up-regulating ER stress markers such as IRE1α and cacy in cancer treatment.
PERK in human ovarian cancer SKOV3 cells and pancre-
atic cancer Panc-1 cells [493]. Tanshinones
Cancer immunotherapy also plays a vital role in cucur- Tanshinone (Fig. 2) is a derivative of phenanthrenequi-
bitacin treatment. Cucurbitacins may influence the none isolated from the dried root or rhizomes of Salvia
production of cytokines and transcription factors that miltiorrhiza Bunge. Tanshinone IIA is the primary bioac-
suppress the immune system, and these mechanisms tive constituent of tanshinones [502], which has various
may help to prevent the development of cancer. Cucur- pharmacological effects, including anti-inflammatory,
bitacin B is able to promote DC differentiation and anti- anti-cancer and anti-atherosclerotic activities, and car-
tumor immunity in patients with lung cancer [494]. The diovascular protection [503–506]. Tanshinone exhibits
combined therapy of cucurbitacin I and recombinant anti-cancer activities in stomach, prostate, lung, breast,
IL-15 is also reported to exhibit immunologic anti-cancer and colon cancers, through inducing cell cycle arrest,
activities in lymphoma with increased C D4+ and C D8+ apoptosis, autophagy, and inhibiting cell migration
T cell differentiation, and promote DC function through [507–515].
TNF-α up-regulation [495]. Tanshinone IIA suppresses cell proliferation and apop-
Although cucurbitacin B has very effective anti-tumor tosis in numerous cancer cells, including human breast
effects, it is shown to exhibit high toxicity, which restricts cancer BT-20, MDA-MB-453, SKBR3, BT-474, MCF-7
its clinical application on cancer therapy. Therefore, and MD-MB-231 [508, 516, 517], and gastric cancer
studies have been focused on tackling this side effect, MKN45 and SGC-7901 cells [518]. It also induces cell
and some cucurbitacin B derivatives have been synthe- cycle arrest at G1 phase in human breast cancer BT-20
sized to screen for effective cancer therapy with safety cells [517], and inhibits cell migration in human gas-
and tolerability. Compound 10b, one of the derivatives tric cancer SGC-7901 cells [514], and cell migration
of cucurbitacin B, shows more potent anti-cancer activ- and invasion in cervix carcinoma stemness-likes cells
ity than cucurbitacin B [496]. The in vivo acute toxicity [519]. Tanshinone I and cryptotanshinone are two other
study also shows that compound 10b has better toler- major bioactive compounds, which also induce cytotox-
ability and safety than cucurbitacin B. In addition, some icity against cancer cells. Tanshinone I induces apopto-
other strategies have been applied to accelerate the clini- sis and pro-survival autophagy in human gastric cancer
cal use of cucurbitacin B. The collagen peptide-modified BGC-823 and SGC-7901 cells [510], while cryptotanshi-
nanomicelles with cucurbitacin B were synthesized to none suppresses cell proliferation and induces cell cycle
enhance the oral availability of cucurbitacin B, and these arrest at G1 phase in murine melanoma B16 cells, and
Luo et al. Chin Med (2019) 14:48 Page 15 of 58
G2/M phase in melanoma B16BL6 cells [520]. In addi- its leukemic activity in human leukemia NB4 cells [537],
tion, tanshinones I and IIA and cryptotanshinone also while the nanoparticles containing tanshinone IIA and
inhibit tumor angiogenesis in endothelial and cancer α-mangostin show increased cytotoxicity in human pros-
cells [521–525]. Furthermore, tanshinone IIA induces tate cancer PC-3 and DU-145 cells [538].
autophagy to inhibit cell growth in human osteosarcoma Tanshinone IIA is shown to enhance chemosensitiv-
143B and MG63 cells and tumor growth in NOD/SCID ity and its efficacy when combined with other therapeu-
mice [526], while it induces autophagy to mediate anti- tic agents. Tanshinone IIA can be an effective adjunctive
cancer activities through activating beclin-1 pathway and agent in cancer, and it enhances the chemosensitivity to
inhibiting PI3K/Akt/mTOR pathway in human oral squa- 5-fluorouracil therapy in human colorectal cancer HCT-
mous cell carcinoma SCC-9, melanoma A375, and gli- 1116 and COLO-205 cells through NF-κB inhibition
oma U251 cells [527–529]. Moreover, tanshinone IIA is [539]. The combination of tanshinone IIA with doxo-
shown to exhibit anti-cancer activities through the inter- rubicin does not only enhance the chemosensitivity of
play between autophagy and apoptosis in human prostate doxorubicin, but also reduces the toxic side effects of
cancer PC-3 cells, mesothelioma H28 and H2452 cells doxorubicin in human breast cancer MCF-7 cells [540].
[502, 530]. In addition, tanshinone IIA and cryptotanshinone syn-
Tanshinone IIA induces apoptosis through mitochon- ergistically enhance apoptosis in human leukemia K562
drial- and caspase-dependent pathways, which includes cells [541].
caspase-3, -9 and PARP activation, cytochrome c release, The anti-cancer effects of Tanshinone IIA have been
and increased ratio of Bax/Bcl-2 in human gastric cancer demonstrated in various cancers in vitro and in vivo, and
MKN45 and SGC-7901 cells, and tumor-bearing mice it can enhance chemosensitivity and its efficacy is very
[518]. It inhibits epithelial–mesenchymal transition by effective when combined with other therapeutic agents.
modulating STAT3-chemokine (C–C motif ) ligand 2 Up to now, the clinical trials of Tanshinone IIA are com-
(CCL2) pathway in human bladder cancer 5637, BFTC pleted only for the treatment of other diseases [542], so
and T24 cells [531], and suppresses cell proliferation and well-designed clinical trials should be done to further
migration via forkhead box protein M1 (FoxM1) down- confirm its safety and efficacy in cancer treatment.
regulation in human gastric cancer SGC-7901 cells [514].
On the other hand, tanshinone I induces apoptosis via Oridonin
Bcl-2 down-regulation in human gastric cancer BGC- Oridonin (Fig. 2) is an ent-kaurane diterpenoid iso-
823 and SGC-7901 cells [510], while cryptotanshinone lated from Rabdosia rubescens (Hemsl.) Hara, which is
induces apoptosis through mitochondrial-, cyclin- and also the main active constituent of Rabdosia rubescens
caspase-dependent pathways in human NSCLC A549 (Hemsl.) Hara [543]. As an orally available drug, oridonin
and NCI-H460 cells [532], as well as via ER stress in is demonstrated to have anti-cancer activities in multi-
human hepatocellular carcinoma HepG2 and breast can- ple cancers over the past decades, including leukemia,
cer MCF-7 cells [533]. lymphoma, osteosarcoma, myeloma, uveal melanoma,
Tanshinone IIA is also shown to exhibit immunomdu- neuroblastoma, hepatocellular, laryngeal, esophageal,
latory effects in cancer [534]. The combination of tanshi- and oral squamous cell carcinoma, lung, colorectal,
none IIA with cyclophosphamide increases C D4+ T cell, breast, gastric, pancreatic, and prostatic cancers [543–
+ +
CD4 /CD8 T cell and NK cell populations compared 558]. The anti-cancer effects of oridonin are shown in
to single treatment in NSCLC Lewis-bearing mice, so it many aspects, including the induction of cell apopto-
can improve the immunological function in lung cancer sis, autophagy, cell cycle arrest, and the suppression of
[534]. Furthermore, cryptotanshinone becomes a new angiogenesis, cell migration, invasion and adhesion [554,
promising anti-tumor immunotherapeutic agent [535]. 559–564].
It induces mouse DC maturation and stimulates IL-1β, Oridonin induces apoptosis in human hepatocellular
TNF-α, IL-12p70 secretion in DCs, and enhances T cell carcinoma HepG2 and Huh6, oral squamous cell carci-
infiltration and Th1 polarization in Lewis-bearing tumor noma WSU-HN4, WSU-HN6 and CAL27, and laryngeal
tissues [535]. cancer HEp-2 cells [550, 559, 561, 565]. It also induces
Tanshinone IIA has poor bioavailability, so a mixed G2/M cell cycle arrest in human oral squamous cell car-
micelle system is developed to form a tanshinone- cinoma WSU-HN4, WSU-HN6 and CAL27, gastric can-
encapsulated micelle [536]. This micelle has higher cer SGC-7901, prostate cancer PC-3 and DU-145, and
cytotoxicity and pro-apoptotic effects in human hepa- breast cancer MCF-7 cells [555, 561, 566, 567]. Oridonin
tocellular carcinoma HepG2 cells compared to tanshi- is also shown to induce autophagy in many cancer cells,
none IIA alone. The tanshinone IIA-loaded nanoparticles which is associated positively or negatively with apopto-
improve the bioavailability tanshinone IIA and enhance sis. It induces autophagy to mediate apoptosis in human
Luo et al. Chin Med (2019) 14:48 Page 16 of 58
NSCLC A549 and neuroblastoma SHSY-5Y cells [558, in human gastric cancer MGC-803 cells and MGC-
568]. On the other hand, autophagy provides a protective 803 xenograft mice [582]. Oridonin phosphate, another
role against oridonin-induced apoptosis, as autophagy derivative, is reported to induce autophagy, which
inhibitor enhances oridonin-induced apoptosis in human can enhance apoptosis in human breast cancer MDA-
cervical carcinoma HeLa, multiple myeloma RPMI 8266, MB-436 cells [583]. A novel analog of oridonin, CYD
laryngeal cancer HEp-2 and Tu212, and epidermoid car- 6-17, inhibits tumor growth in bladder cancer UMUC3
cinoma A-431 cells [569–572]. The anti-cancer effects of xenograft mice and renal carcinoma 786-O xenograft
oridonin are also shown to be through suppressing angi- mice [584, 585]. In addition, drug delivery system is also
ogenesis and metastasis, which are the primary causes of developed to improve the bioavailability of oridonin.
tumor growth and metastasis. It can inhibit cell migra- The inhalable oridonin-loaded microparticles exhibit
tion and invasion, and tube formation in human breast strong pro-apoptotic and anti-angiogenic effects through
cancer 4T1 and MDA-MB-231, human and murine mela- mitochondrial-related pathways in NSCLC rats [586],
noma A375 and B16F10, osteosarcoma MG63 and 143B, whilst the oridonin-loaded nanoparticles enhance cellu-
and HUVECs, as well as tumor metastasis in HepG2 xen- lar uptake and exert better anti-cancer effects in human
ograft zebrafish and mice, 4T1 xenograft mice, and 143B hepatocellular carcinoma HepG2 cells [587].
xenograft mice [554, 562–564, 573]. The combination of oridonin with other agents plays a
Proteomic and functional analyses reveal that ER potential role in cancer therapy. AG1478, a specific epi-
stress and poly(rC)-binding protein 1 (α-CP1) are poten- dermal growth factor receptor (EGFR) inhibitor, aug-
tial pathways involved in the anti-proliferative and pro- ments oridonin-induced apoptosis through oxidative
apoptotic activities of oridonin [546]. Oridonin inhibits stress and mitochondrial pathways in human epider-
cell growth and induces apoptosis through ER stress and moid carcinoma A-431 cells [588]. The combination of
ASK1/JNK signaling pathways in human hepatocellular γ-tocotrienol and oridonin exerts synergistic anti-can-
carcinoma Huh6 cells [559]. Besides, the mitochondrial cer effects in murine + SA mammary adenocarcinoma
redox change is proved to be a potential mediator for the epithelial cells, which are mainly through the induction
pro-apoptosis effect of oridonin [565]. The anti-prolif- of autophagy [589]. Moreover, oridonin can enhance
erative effect of oridonin is also shown to be associated the pro-apoptotic activity of NVP-BEZ235 in human
with mitochondrial-mediated apoptosis, which is charac- neuroblastoma SHSY-5Y and SK-N-MC cells through
terized by mitochondrial membrane potential reduction, autophagy [558], whilst the combination of oridonin and
subsequent cytochrome c release, PARP, caspase-3 and -9 cetuximab exhibits potent pro-apoptotic effect in human
activation, and decreased Bcl-2/Bax ratio [551, 565, 574, laryngeal cancer HEp-2 and Tu212 cells [572].
575]. Oridonin also inhibits cell proliferation through Clinical trials are essential to test the safety and effi-
bone morphogenetic protein 7 (BMP7)/p38 MAPK/ cacy of oridonin before drug approval. A derivative of
p53 pathway in human colorectal cancer HCT-116 and oridonin, HAO472, is currently under a phase I clinical
SW-620 cells [553, 576, 577], and induces apoptosis via trial for the treatment of acute myelogenous leukemia in
hydrogen peroxide (H2O2) production and glutathione China [590].
depletion in human colorectal cancer SW-1116 cells
[578]. Furthermore, the down-regulation of AP-1 is Shikonin
reported to be the initial response to oridonin treatment, Shikonin (Fig. 2) is an active naphthoquinone, which is
which decreases the expressions of NF-κB and MAPK to derived from the dried root of Lithospermum erythrorhi-
inhibit cell proliferation [579]. zon, Arnebia euchroma and Arnebia guttata, and it pos-
Oridonin possesses an immunosuppressive effect sesses anti-oxidative, anti-inflammatory, and anti-cancer
which modulates microglia activation, enhances T cell activities [591–594]. It is effective in treating different
proliferation, alters the balance of Th1-T helper type 2 kinds of cancers, including breast, prostate, ovarian and
cells (Th2), reduces inflammatory cytokine secretion thyroid cancers, Ewing sarcoma, and myelomonocytic
such as IL-2, IL-4, IL-6, IL-10 and TNF-α, and modulates lymphoma [595–600]. Shikonin exerts anti-cancer effects
an anti-inflammatory target, B lymphocyte stimulator mainly by inducing apoptosis, necroptosis, autophagy,
[580]. It also decreases inflammatory cytokine secre- cell cycle arrest, and by inhibiting cell proliferation,
tion in human pancreatic cancer BxPC-3 cells, including growth and metastasis [593, 601, 602].
IL-1β, IL-6 and IL-33 [581]. Shikonin is reported to inhibit cell growth by induc-
The derivatives and analogs of oridonin usually exhibit ing cell cycle arrest and promoting apoptosis in human
more potent anti-cancer activities than oridonin. Geri- NSCLC A549, gallbladder cancer NOZ and EHGB-1,
donin, a novel derivative of oridonin, inhibits cell growth esophageal cancer EC109, and epidermoid carcinoma
and induces G2/M phase arrest through ROS production A-431 cells [601, 603–605]. It can also induce necroptosis
Luo et al. Chin Med (2019) 14:48 Page 17 of 58
via autophagy inhibition in human NSCLC A549 cells naphthazarin ring of shikonin is modified to produce
[593], and through ROS overproduction in human naso- DMAKO-05, which can specifically target cancer cells
pharyngeal carcinoma 5-8F, and glioma SHG-44, U87 instead of normal cells [626]. DMAKO-05 can also sup-
and U251 cells [606, 607]. Moreover, shikonin induces press cell survival in human colorectal cancer HCT-116
autophagy in human melanoma A375, pancreatic can- cells, and inhibits tumor growth in colorectal cancer
cer BxPC-3, and hepatocellular carcinoma Bel-7402 and CT-26 xenograft mice [627]. Besides, it inhibits cell pro-
Huh7 cells [608–610]. However, autophagy provides a liferation and migration, and induces cell cycle arrest
protective role in shikonin-induced apoptosis in human and apoptosis in murine melanoma B16F0 cells [626].
melanoma A375 cells [608]. In addition, shikonin can Another novel shikonin derivative, cyclopropylacetyl-
suppress metastasis by the inhibition of tyrosine kinase shikonin, exhibits strong anti-tumor and pro-apoptotic
c-Met and integrin (ITG) β1 in human NSCLC A549 cells effects in human melanoma WM164 and MUG-MEL2
[602, 611]. cells [628]. In addition, drug delivery system is also
There are multiple mechanisms involved in the anti- developed to promote the intracellular delivery of shi-
cancer effects of shikonin, including ER stress, ROS konin. The shikonin-loaded nanogel enhances RIP1- and
generation, glutathione (GSH) depletion, mitochondrial RIP3-dependent necroptosis in human osteosarcoma
membrane potential disruption, p53, superoxide dis- 143B cells [629]. There is an increased accumulation of
mutase (SOD) and Bax up-regulation, PARP cleavage, shikonin-loaded nanogel in the tumor tissue, and this
catalase and Bcl-2 down-regulation [591, 612–614]. The nanogel can further inhibit tumor growth and metasta-
pro-apoptotic effect of shikonin is also caused by the dis- sis in 143B xenograft mice. Furthermore, the modified
a2+ homeostasis and mitochon-
ruption of intracellular C shikonin-loaded liposomes have higher cytotoxicity, and
drial dysfunction, which involves enhanced Ca2+ and inhibit cell proliferation, metastasis in human breast can-
+
potassium (K ) efflux, caspase-3, -8 and -9 activation, cer MDA-MB-231 cells [630].
and Bcl-2 family protein modulation [615, 616]. ERK The combination therapy is widely used to provide
pathway also plays a role in shikonin-induced anti-cancer synergistic effects of anti-cancer activities. Shikonin
effects. Shikonin induces apoptosis and inhibits metasta- can enhance the pro-apoptotic effect of taxol in human
sis through suppressing ERK pathway in human NSCLC breast cancer MBA-MD-231 cells, and this combination
NCI-H460 and A549 cells, respectively [611, 617]. c-Myc improves mice survival and inhibits tumor growth in
down-regulation along with inhibition of ERK/JNK/ MDA-MB-231 xenograft mice [631]. Besides, shikonin
MAPK and Akt pathways are also involved in shikonin- can also potentiate the anti-cancer effects of gemcitabine
induced apoptosis and anti-proliferation in acute and through NF-kB suppression and by regulating RIP1 and
chronic leukemia [618–620]. Moreover, the activation of RIP3 expressions in human pancreatic cancer [632, 633].
necroptosis initiators, receptor interacting serine-threo- Shikonin is also reported to promote the efficacy of adri-
nine protein kinase (RIP) 1 and RIP3, by shikonin does amycin in lung cancer and osteosarcoma [634, 635], and
not only contribute to DNA double strand breaks via enhance sensitization to cisplatin in colorectal cancer
ROS overproduction [621], but also facilitates glycolysis [636]. Apart from the synergistic effect of shikonin, the
suppression via intracellular H 2O2 production [622]. In combination of shikonin and paclitaxel reverses MDR in
addition, shikonin induces cell cycle arrest through p21 human ovarian cancer A2780 cells [10].
and p27 up-regulation, cyclin and CDK down-regulation The single or combined therapies with shikonin show
[605]. Therefore, numerous pathways involved in shi- promising anti-cancer effects in vitro and in vivo, so pre-
konin-induced anti-cancer effects may explain the broad clinical data has confirmed its therapeutic use in cancer
range of its activities. treatment, as a result, clinical trials will be carried out to
Shikonin is also shown to modulate the function of further to confirm its safety and efficacy in humans.
the immune system. It can enhance the proliferation of
NK cells and its cytotoxicity to human colorectal cancer Gambogic acid (GA)
Caco-2 cells by regulating ERK1/2 and Akt expressions GA (Fig. 2) is one of the major compounds derived
[623]. It can also bind directly to heterogeneous nuclear from gambogethe resin exuded from Garcinia spe-
ribonucleoprotein A1 to induce immunogenic cell death cies including G. hanburyi and G. Morella [637]. It has
in human breast cancer MDA-MB-231 cells [624]. Shi- multiple biological activities such as anti-oxidative,
konin is also reported to be used as an immunotherapy anti-inflammatory, and anti-cancer activities [638,
modifier in cell-based cancer vaccine systems, suggesting 639]. Plenty of evidence shows that GA inhibits cell
its potential application in cancer immunotherapy [625]. proliferation, invasion, survival, metastasis and chemo-
Derivatives are developed to enhance the anti- resistance, and induces angiogenesis in many types of
cancer and tumor targeting effects of shikonin. The cancers such as gastric and prostate cancers, leukemia,
Luo et al. Chin Med (2019) 14:48 Page 18 of 58
multiple myeloma, osteosarcoma, and renal carcinoma cytokine production including TNF-α, IL-1β and IL-6 by
through multiple signaling mechanisms [640–646]. suppressing p38 pathway in murine macrophage RAW
Many studies have reported the anti-cancer effects 264.7 cells [661].
of GA in human breast cancer [647–650]. GA at low GA has low solubility, instability and poor pharma-
concentrations (0.3–1.2 μM) can inhibit cell inva- cokinetic properties [662]. In order to increase its water
sion without affecting cell viability, while high con- solubility, GA is conjugated with a cell-penetrating pep-
centrations of GA (3 and 6 μM) can induce apoptosis tide, trans-activator of transcription, to form GA-TAT
via ROS accumulation and mitochondrial apoptotic [658]. This GA-TAT enhances apoptosis through ROS
pathway in human breast cancer MDA-MB-231 cells accumulation in human bladder cancer EJ cells. Another
[651]. GA also induces apoptosis via ROS production study uses a co-polymer to encapsulate GA to form GA
in human bladder T24 and UMUC3 cells [652]. At ear- micelles [639]. These GA micelles have better cellular
lier time points, GA induces ROS-mediated autophagy, uptake which can further enhance apoptosis in human
which produces a strong cell survival response. How- breast cancer MCF-7 cells and the anti-tumor effects in
ever, at later time points, caspases are activated which MCF-7 xenograft mice. Moreover, GA is encapsulated
degrade autophagic proteins and cell survival proteins, into the core of the nanoparticles to enhance the stability
and this eventually induces apoptosis. Similarly, GA- of GA and its circulation time [662]. These nanoparticles
induced autophagy via ROS provides a cytoprotective have tumor targeting properties, and enhance the anti-
effect to human pancreatic cancer Panc-1 and BxPC-3 tumor activities of GA without inducing higher toxicity.
cells [653], and ROS scavenger, N-acetylcysteine, can The combination of GA and other chemotherapy agents
reverse GA-induced autophagy in human NSCLC NCI- has been widely used to improve the therapeutic effects
H441 cells [654]. Moreover, GA inhibits cell invasion against various cancers such as osteosarcoma, pancreatic
and migration through reversion-inducing-cysteine- and lung cancers [639, 653, 663, 664]. Cisplatin resist-
rich protein with kazal motifs (RECK) up-regulation ance is a main clinical problem for the treatment of lung
in human NSCLC A549 cells and A549 xenograft cancer, and the treatment of cisplatin with GA is shown
mice [655], and prevents TNF-α-induced invasion in to enhance apoptosis and decrease the cisplatin resist-
human prostate cancer PC-3 cells [656]. It also inhibits ance index in human NSCLC cisplatin-resistance A549/
angiogenesis in HUVECs, and prevents tumor growth DDP cells [663]. Moreover, GA and retinoic acid chloro-
through the inhibition of tumor angiogenesis [657]. chalcone are loaded into glycol chitosan nanoparticles to
ROS-related pathways play a vital role in GA-induced form RGNP [639]. The RGNP exhibits synergistic effects
cell death [642, 646, 647, 651–654, 658]. GA induces to inhibit cell proliferation and induces apoptosis in oste-
apoptosis mainly through ROS accumulation in human osarcoma. The combination of GA with doxorubicin syn-
pancreatic cancer Panc-1 and BxPC-3, NSCLC NCI- ergistically reduces cell viability in human ovarian cancer
H441, castration-resistant prostate cancer PCAP-1, platinum-resistance SKOV3 cells, and this combination
melanoma A375, breast cancer MCF-7 cells [642, 646, also suppresses tumor growth in SKOV3 xenograft mice
647, 653, 654]. It also induces oxidative stress-dependent [665].
caspase activation to mediate apoptosis in human blad- The safety and efficacy of GA at different dosages in
der cancer T24 and UMUC3 cells [652]. Moreover, GA patients with advanced malignant tumors have been
increases the expressions of ER stress markers such as compared in a phase IIa clinical trial [666]. GA had a
GRP78, CHOP, activating transcription factor 6 (ATF-6) safety profile at a dosage of 45 mg/m2. The patients with
and caspase-12, and co-treatment with chemical chaper- GA administration on days 1–5 in a 2-week cycle showed
one, 4-PBA, significantly reduces these expressions and a greater disease control rate and only Grades I and II
apoptosis in human NSCLC A549 cells, so it is suggested adverse reactions. To further investigate the safety and
that GA induces ER stress to mediate apoptosis [659]. efficacy of GA, a phase IIb clinical trial involving a larger
Previous studies have shown some immunomodula- sample size of patients would be needed.
tory activities of GA [660, 661]. The activation of TLRs
is important to initiate immune responses, and TLR4 Artesunate
forms a complex with myeloid differentiation factor 2 Artesunate (Fig. 2) is a semi-synthetic compound derived
(MD2) to recognize its ligand, like LPS. GA is shown to from ART, which is widely used as an anti-malarial
reduce pro-inflammatory cytokine production in LPS- agent [667]. As an analog of ART, artesunate exerts bet-
primed primary macrophages such as TNF-α, IL-1β, ter water solubility and higher oral bioavailability, due
IL-6 and IL-12, and also inhibit the activation of TLR4 by to its special structure with an additional hemisuccinate
disrupting the interaction of TLR4/MD2 complex with group that makes it a better candidate for cancer treat-
LPS [660]. Similarly, it also reduces pro-inflammatory ment [668]. The anti-cancer effects of artesunate have
Luo et al. Chin Med (2019) 14:48 Page 19 of 58
been demonstrated in bladder, breast, cervical, colorec- to mediate apoptosis in murine ovarian cancer ID8 cells
tal, esophageal, gastric, ovarian and prostate cancer, renal [674]. It also exerts anti-tumor effects through suppress-
carcinoma, leukemia, melanoma and multiple myeloma ing NK killing activity and lymphocyte proliferation,
[179, 669–679]. Its anti-cancer effects include induction which results in decreased TGF-β1 and IL-10 levels in
of cell cycle arrest and apoptosis, inhibition of cell pro- colorectal cancer Colon-26 and RKO cells [691]. Besides,
liferation and growth, metastasis and angiogenesis [670, artesunate also exerts immunosuppression through
678, 680]. forkhead box P3 (Foxp3) down-regulation in T cells and
Artesunate can induce apoptosis in various cancers decreases prostaglandin E2 (PGE2) production in human
including human breast cancer MCF-7, MDA-MB-468 cervical cancer Caski and HeLa cells [671]. Moreover, it
and SKBR3 cells, gastric cancer SGC-7901 and HGC- enhances γδ T cell-mediated anti-cancer effect through
27, colorectal cancer HCT-116, and esophageal cancer augmenting γδ T cell cytotoxicity and decreasing TGF-β1
Eca109 and Ec9706 cells [670, 672, 673, 681–683]. It also levels to reverse immune escape in human hepatocellular
induces cell cycle arrest at ROS-dependent G2/M phase carcinoma HepG2 cells [692].
and ROS-independent G1 phase in human breast can- The treatment of artesunate with other therapies shows
cer MDA-MB-468 and SKBR3, and ovarian cancer HEY1 promising anti-cancer effects in several studies [693–
and HEY2 cells [670, 684], and induces G2/M cell cycle 697]. Artesunate and cisplatin synergistically induce
arrest through autophagy in human breast cancer MCF-7 DNA double-strand breaks and inhibit clonogenic forma-
and MDA-MB-231 cells [685]. Artesunate is also shown tion to mediate cytotoxic effects in human ovarian cancer
to induce autophagy to exert cytoprotective effects in A2780 and HO8910 cells [693]. The combined treatment
human colorectal cancer HCT-116 cells, and the inhibi- of artesunate and erlotinib enhances the inhibition of cell
tion of autophagy enhances artesunate-mediated apop- growth in human glioblastoma multiforme U87MG cells
tosis [179]. Similarly, artesunate-induced mitophagy [694].
provides a protective effects against cell death in human Clinical studies are carried out to investigate the safety
cervical cancer HeLa cells [686]. Moreover, it inhibits and efficacy of artesunate in patients with colorectal and
cell invasion and migration in human prostate cancer breast cancers, and advanced solid tumor malignancies
DU-145 and LNCaP, cervical cancer Caski and HeLa [698–701]. A phase I study is performed to evaluate the
cells, and uveal melanoma cells [675, 678, 687], and sup- safety and the maximum tolerated dose of artesunate in
presses tumor angiogenesis in HUVECs and renal carci- patients with metastatic breast cancer, the oral admin-
noma 786-O xenograft mice [676, 680]. istration of artesunate is safe and 2.2–3.9 mg/kg per day
In most cases, the inhibition effects of artesunate is well tolerated [701]. Another phase I study is assessed
against cancer cells are resulted from apoptosis. Artesu- in patients with advanced solid tumor malignancies, and
nate induces apoptosis through cyclooxygenase-2 the maximum tolerated dose of intravenous artesunate
(COX-2) down-regulation in human bladder cancer T24 is 18 mg/kg [698]. The tolerability and anti-proliferative
and RT4, and gastric cancer HGC-27 cells [669, 683]. properties of oral artesunate are also shown in patients
Mitochondrial pathways also play an important role in with colorectal cancer [699]. Moreover, a study of long
artesunate-mediated anti-cancer effects [673, 681, 683]. term treatment with oral artesunate is performed in
Artesunate inhibits tumor growth through ROS- and patients with metastatic breast cancer, 2.3–4.1 mg/kg per
p38 MAPK-mediated apoptosis in human rhabdomyo- day treatment for up to 1115 cumulative days does not
sarcoma TE671 cells [688]. It also exerts anti-tumor show any major safety concerns [700]. An ongoing phase
activities through the loss of mitochondrial membrane II clinical trial is carried out to study the safety and effec-
potential, Bcl-2 down-regulation, Bax up-regulation, tiveness of neoadjuvant artesunate in patients with stage
and caspase-3 activation in human gastric cancer SGC- II or III colorectal cancer awaiting surgical treatment.
7901 and HGC-27, esophageal cancer Eca109 and Ec9706
cells, and breast cancer MCF-7 xenograft mice [673, 681, Wogonin
683]. In addition, gene expression analysis identifies that Wogonin (Fig. 2) is a plant flavonoid extracted from roots
ER stress is the most relevant pathway for the anti-tumor of Scutellaria baicalensis, Scutellaria amoena and Scutel-
activity of artesunate in B-cell lymphoma [689]. Interest- laria rivularis, and stem of Anodendron affine Druce, and
ingly, artesunate selectively inhibits cell growth through has many pharmacological effects including anti-viral,
iron-dependent and ROS-mediated ferroptosis in human anti-oxidative, anti-inflammatory, anti-cancer and neuro-
head and neck cancer HN9 cells [690]. protective activities [702–705]. It has various anti-cancer
Immunomodulation also plays a vital role in artesu- effects in many cancers, including lung, breast, head and
nate-mediated anti-cancer effects [671, 674, 691, 692]. neck, gastric and colorectal cancers, glioma, leukemia,
Artesunate induces Th1 differentiation into C D4+ T cells lymphoma, and osteosarcoma, through the induction
Luo et al. Chin Med (2019) 14:48 Page 20 of 58
of apoptosis and cell cycle arrest, and inhibition of cell migration through modulating inflammatory microen-
growth, migration, invasion, and angiogenesis [706–716]. vironment via IL-6/STAT3 pathway in human NSCLC
Wogonin can induce apoptosis and inhibit cell prolif- A549 cells [723]. Moreover, immunization with wogonin-
eration in human neuroblastoma SK-N-BE2 and IMR-32, treated tumor cell vaccine effectively inhibits tumor
NSCLC A549, glioma U251 and U87, and hepatocellu- growth in MFC xenograft mice [732]. Targeting TNF
lar carcinoma HepG2 and Bel-7402 cells [704, 706, 711, receptor with wogonin is also suggested to be a potential
717]. It also induces cell cycle arrest in human colorectal strategy for the treatment of chronic lymphocytic leuke-
cancer HCT-116, NSCLC A549, chronic myelogenous mia [712].
leukemia imatinib-resistant K562, and ovarian cancer In order to enhance the accumulation and retention of
A2780 cells [716, 718–720]. Besides, wogonin induces wogonin in cancer cells, wogonin-conjugated Pt(IV) pro-
autophagy in human pancreatic cells Panc-1 and Colo- drug is developed [733]. This pro-drug enhances the anti-
357, and nasopharyngeal carcinoma NPC-TW076 and proliferative and pro-apoptotic effects through casein
NPC-TW039 cells [721, 722]. However, inhibition of kinase 2 (CK2)-mediated NF-κB pathway in human gas-
autophagy promotes wogonin-induced apoptosis in tric cancer SGC-7901 and cisplatin-resistant SGC-7901/
human nasopharyngeal carcinoma NPC-TW076 and cDDP cells, and reverses cisplatin resistance in cisplatin-
NPC-TW039 cells [722]. It also inhibits metastasis in resistant SGC-7901/cDDP xenograft mice. It also fur-
human hepatocellular carcinoma Bel-7402 and HepG2 ther induces cell cycle arrest, enhances ROS production
cells, and NSCLC A549 cells [717, 723], and through and apoptosis, and decreases mitochondrial membrane
MMP-9 suppression in human hepatocellular carcinoma potential compared to wogonin in SGC-7901 cells [734].
MHCC97-L and PLC/PRF/5 cells [724]. In addition, LW-213, a derivative of wogonin, inhibits cell prolifera-
wogonin also represses multiple myeloma-stimulated tion and induces cell cycle arrest in human breast cancer
angiogenesis through c-Myc/von Hippel-Lindau tumor MCF-7 and MDA-MB-231 cells, and suppresses tumor
suppressor (VHL)/HIF-1α signaling pathway [725], LPS- growth in MCF-7 xenograft mice [735]. A synthetic
and H2O2-induced angiogenesis through PI3K/Akt/ wogonin derivative, GL-V9, inhibits metastasis in human
NF-κB pathway [726, 727]. breast cancer MDA-MB-231 and MCF-7 cells [736], and
Mitochondrial dysfunction, oxidative stress and ER induces apoptosis and cell cycle arrest in human hepato-
stress play important roles in wogonin-induced anti- cellular carcinoma HepG2 and gastric cancer cells MGC-
cancer effects. Wogonin activates mitochondrial and 803 cells [737–739]. Moreover, targeting cancer cells
ER stress-related pathways including the modulation of specifically is an important strategy in cancer therapy, so
Bcl-2 family proteins, cytochrome c release, GRP78 and wogonin-loaded liposomes are synthesized [740]. These
94-kDa glucose-regulated protein (GRP94) accumula- liposomes accumulate in the liver and prolong its reten-
tion, and caspase activation in human neuroblastoma tion time and exert better inhibitory effects than wogonin
SK-N-BE2 and IMR-32 cells, and induces mitochondrial in human hepatocellular carcinoma HepG2 cells.
dysfunction through IRE1α-dependent pathway [704]. ER The combination therapy has been widely used to
stress markers and downstream pathways are also acti- enhance the anti-cancer effects of wogonin. The com-
vated following wogonin treatment in human leukemia bined treatment of wogonin and oxaliplatin syn-
HL-60 and osteosarcoma U2OS cells, including IRE1α, ergistically inhibits cell growth in human gastric
PERK-eIF2α, ATF-6, CHOP, GRP94 and GRP78 [714, cancer BGC-823 cells and BGC-823 xenograft zebrafish,
728]. Wogonin also enhances ROS production in human through nitrosative stress and disruption of mitochon-
glioma U251 and U87, pancreatic cancer Panc-1 and drial membrane potential [741]. Wogonin also suppresses
Colo-357, and NSCLC A549 cells [711, 721, 729]. Moreo- sorafenib-induced autophagy to exacerbate apoptosis in
ver, it inhibits cell growth and induces apoptosis through human hepatocellular carcinoma Hep3B and Bel-7402
NF-κB suppression in Epstein–Barr virus-positive lym- cells [742], and augments cisplatin-induced apoptosis
phoma cells [730], and suppresses cell proliferation and through H2O2 accumulation in human NSCLC A549 and
invasion through NF-κB/Bcl-2 and EGFR pathways in cervical cancer HeLa cells [743].
human hepatocellular carcinoma HepG2 and Bel-7402 As wogonin has various anti-cancer activities, it is cur-
cells [717]. rently under phase I clinical trial to test the safety and
Wogonin has immunomodulatory effects in cancer efficacy as an anti-cancer drug in China [734].
cells. It enhances the recruitment of DCs, T and NK cells
into the tumor tissues in gastric cancer MFC xenograft β‑Elemene
mice, and also down-regulates the level of B7-H1, an β-Elemene (Fig. 2) is a sesquiterpene mixture isolated
immunoglobulin-like immune suppressive molecule, to from various Chinese herbs such as Curcuma wenyujin
promote anti-tumor immunity [731]. It also inhibits cell Y. H. Chen et C. Ling, Rhizoma zedoariae, and Curcuma
Luo et al. Chin Med (2019) 14:48 Page 21 of 58
Zedoary. It has various pharmacological effects includ- A549 cells [747], and up-regulates HIF-1α expression
ing anti-oxidative, anti-inflammatory and anti-cancer via ROS to induce apoptosis in human osteosarcoma
activities [744–746]. It exerts anti-cancer effects in many MG63 and Saos-2 cells [751].
cancers, such as lung, gastric, cervical, breast and blad- β-Elemene has immunomodulatory effects in cancer
der cancers, osteosarcoma, through apoptosis, inhibition and immune cells. It inhibits LPS-induced IL-6, TNF-
of cell proliferation, migration and invasion, angiogenesis α, IL-1β and IL-10 secretion, as well as inducible nitric
[746–752]. oxide synthase in murine RAW264.7 marcophages
β-Elemene is shown to induce apoptosis in human [745]. M2 macrophages are regarded as tumor-associ-
cervical cancer SiHa, NSCLC A549 cells, primary blad- ated macrophages, which can promote tumorigenesis
der cancer cells, and Burkitt’s lumphoma, and inhibit [762]. β-Elemene can induce the polarization of M2
tumor growth in Lewis tumor-bearing mice [746, 747, to M1 macrophages, and can also suppress M2 mac-
749, 753, 754]. It up-regulates insulin-like growth factor- rophage-treated conditioned medium-induced cell pro-
binding protein 1 (IGFBP1) to induce a reciprocal inter- liferation, migration and invasion in mouse lung cancer
action between microRNA 155-5p and FoxO3a, which Lewis cells [762].
leads to the inhibition of cell growth in human NSCLC β-Elemene has poor water solubility, low oral bio-
A549 and H1975 cells [755]. β-Elemene also induces S availability and severe phlebitis, so different deliv-
phase arrest in human NSCLC A549 cells [754], while it ery systems have been developed to solve these issues
induces G0/G1 phase arrest in human glioblastoma U87 [763–765]. β-Elemene-loaded nanostructured lipid car-
cells [756]. Moreover, it induces protective autophagy riers are synthesized to enhance the intravenous deliv-
in human gastric cells MGC-803 and SGC-7901, and ery of β-elemene, and have higher bioavailiabity [763].
NSCLC A549 cells, as autophagy inhibition promotes They inhibit tumor growth compared to β-elemene in
β-elemene-induced anti-tumor effects [748, 757]. How- hepatocellular carcinoma H22 xenograft mice. ETME, a
ever, autophagy inhibition attenuates β-elemene-induced novel β-elemene derivative, synergizes with arsenic tri-
apoptosis in human NSCLC cisplatin-resistant SPC-A-1 oxide to induce cell cycle arrest and apoptosis in human
cells [758]. β-Elemene can also inhibit cell migration and hepatocellular carcinoma SMMC-7721 cells, which is
invasion in human cervical cancer SiHa, murine breast dependent on p53 [766]. Another β-elemene derivative,
cancer 4T1 and melanoma B16F10 cells [749, 752, 759], 13,14-bis(cis-3,5-dimethyl-1-piperazinyl)-β-elemene
whilst it inhibits cell growth and metastasis through (IIi), is shown to inhibit cell proliferation in human gas-
angiogenesis suppression in murine melanoma B16F10 tric cancer SGC-7901 and cervical cancer HeLa cells, and
cells [752]. In addition, β-elemene can reverse drug inhibit tumor growth in sarcoma S-180 xenograft mice
resistance in human NSCLC erlotinib-resistant A549/ER [767]. It also induces autophagy in human breast cancer
cells by inhibiting P-gp expression and P-gp dependent MCF-7 cells, so it can be a potential anti-tumor agent.
drug efflux [760]. The combination therapy is commonly used to
β-Elemene exerts anti-tumor effects through phos- enhance the efficacy of β-elemene for cancer treatment.
phatase and tensin homolog (PTEN) up-regulation and β-Elemene when combined with cisplatin synergisti-
Akt suppression in human primary bladder cancer cells cally enhances apoptosis and inhibits cell proliferation
[746]. It also inhibits cell proliferation and invasion, in human gingival squamous cell carcinoma YD-38 cells
and induces apoptosis via inhibition of Wnt/β‑catenin and YD-38 xenograft mice [768]. β-Elemene potenti-
signaling pathway in human cervical cancer SiHa cells ates the anti-proliferation effect of gefitinib as well as
[749]. β-elemene-induced apoptosis is also shown to be the induction of apoptosis and autophagy in human glio-
through mitochondrial-related pathways, including p21 blastoma multiforme U251 and U87MG cells, through
and Bax up-regulation, caspase-9 activation, Bcl-2 and inhibiting EGFR signaling pathway [769]. It also reverses
survivin down-regulation [754]. On the other hand, it drug resistance in chemo-resistant breast cancer cells by
reverses drug resistance through mitochondrial-medi- reducing resistance transmission via exosomes [770], and
ated apoptosis in human NSCLC cisplatin-resistant enhances the sensitivity to TNF‐related apoptosis‐induc-
A549/DDP cells, via cytochrome c release, caspase-3 ing ligand (TRAIL) partly through death-inducing signal-
activation, Bcl-2 associated agonist of cell death (Bad) ing complex formation in human gastric cancer BGC-823
up-regulation and Bcl-2 down-regulation [761]. ER and SGC-7901 cells [771].
stress also plays a role in β-elemene-induced apoptosis. The Elemene Emulsion mainly containing β-elemene
β-Elemene up-regulates ER stress markers to induce has been approved by China’s State Food and Drug
apoptosis in human NSCLC A549 cells, including Administration, and now it is prescribed as an oral or
PERK, IRE1α, ATF-6, ATF-4 and CHOP [747]. Moreo- injected drug to improve anti-cancer efficacy and reduce
ver, it also enhances ROS production in human NSCLC the side effects as adjuvant therapy.
Luo et al. Chin Med (2019) 14:48 Page 22 of 58
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
Curcumin Curcuma longa, Bladder cancer; breast T24, RT4, MDA- BxPC-3-GemR Anti-angio- Activates caspase-3, -9, PARP; 0–5 μM; 0–15 μM; Gemcitabine; [12, 21,
Curcuma cancer; cervical MB-231, HeLa, SiHa, xenograft mice; C4-2 genesis; Down-regulates Akt, Bcl-2, 0–16 μM; 0–20 μM; NVP-BEZ235; 795–814]
zedoaria, cancer; colorectal HCT-116, HT-29, xenograft mice; PC-3 anti-metastasis; Bcl-xL, CTGF, cyclin D1, cyclin 0–25 μM; 0–40 μM; α-Tomatine
(2019) 14:48
Acorus cancer; esophageal RKO, HCT-15, DLD- xenograft mice; RN5 anti-prolifera- E1, ERK1/2, EZH2, FoxM1, GLI1, 0–50 μM; 0–125 μM;
calamus L. squamous cell carci- 1, EC1, EC9706, xenograft mice; U87 tion; induces ITGA5, Jak1, JNK, MMP-2, Mcl-1, 10–40 μM; 15,
noma; gastrointestinal KYSE450, TE13, AGS, xenograft mice cell cycle NF-κB, Notch1, p15, p16, p62, 25 μM; 25 μM;
cancer; glioma; hepa- U87, T98G, HepG2, arrest; inhibits p70S6 K, ROCK1, RhoA, SHH, 30 μM; 0–6 μg/ml;
tocellular carcinoma; Tu212, A549, H1299, cell viability; SSAT, STAT1, STAT3, Suz12, 5 mg/kg; 60 mg/kg;
laryngeal cancer; lung H460, H292, NCI- pro-apoptosis TROP2, vimentin, WT1, XIAP, 200 mg/kg; 500 mg/
cancer; leukemia; liver H520, NCI-H1373, YAP/TAZ; Enhances cytochrome kg; 25 μg/mouse
cancer; mesothe- NCI-H2170, K562, c release, ROS accumulation;
lioma; neuroblastoma; HL-60, PLC/PRF5, Inhibits CDK2 activity, PI3K/Akt/
oral squamous cell WRL68, Huh7, mTOR, SHH/GLI1, STAT3, TGF-β
carcinoma; pancreatic KMCH, RN5, N2a, pathways; Up-regulates AIF,
cancer; prostate can- SCC-25, Patu8988, Bax, Bex-1, -2, -3, -4, -6, HIF-1α,
cer; renal carcinoma; Panc-1, C4-2, PC-3, microRNA-15a, microRNA-16-1,
retinoblastoma LNCaP, VCaP, Caki, microRNA-99a, p21, p53, p73,
O-Rb50, Y79 PKD1, SMOX
EGCG Camellia Biliary tract cancer; BDC, CCSW-1, EGI-1, 4T1 xenograft mice; Anti-angio- Activates caspase-3, -7, PARP; 0–20 µM; 0–40 µM; Bleomycin; Cispl- [93–95, 100,
sinensis bladder cancer; SkChA-1, TFK-1, A549 xenograft mice; genesis; Down-regulates ABCG2, Akt, 0–50 µM; 0–100 µM; atin; Curcumin; 101, 103,
breast cancer; cervical SW-780, MCF-7, 4T1, BCaPT10 xenograft anti-metastasis; AXL, Bcl-2, Bcl-xL, E-cadherin, 0–200 µM; Docetaxel; 123–125,
cancer; colorectal T47D, MDA-MB-231, mice; BCaPM-T10 anti-prolifera- β-catenin, CDK2, CDK4, COX-2, 0–400 μM; 5-Fluorouracil; 815–834]
cancer; gallbladder MDA-MB-436, xenograft mice; tion; induces CTTN, cyclin B1, cyclin D1, cyclin 2–100 μM; 10 μM; Oxaliplatin;
cancer; gastric cancer; SUM-149, SUM-190, CL1-5 xenograft autophagy, D2, cyclin D3, DNMT1, EGFR, 20 μM; 25, 50, Pterostilbene;
glioblastoma; head HeLa, DLD-1, HT-29, mice; Oral squamous cell cycle ERα, ERK1/2, FAK, FN1, GSK3β, 100 μM; 40 μM; 50, Temozolomide
and neck cancer; lung HCT-116, GBC, cell carcinoma arrest; inhibits HDAC1, HER2, HSP90, IKKα, JNK, 100 μM; 80 µM;
cancer; nasopharyn- MzChA-1, MzChA-2, xenograft mice; cell viability, MDR-1, MGMT, MMP-2, MMP-9, 0–60 μg/ml; 10 mg/
geal carcinoma; SGC-7901/FU, PC-12 xenograft epithelial– NANOG, NF-κB, Notch, Oct-4, kg; 10–20 mg/
NSCLC; oral cancer; MGC-803/FU, AGS, mice; SCG-7901/ mesenchymal u-PA, paxillin, P-gp, PI3K, Raf-1, kg; 15 mg/kg;
pancreatic cancer; C6, U251, SHG-44, FU xenograft mice; transition; pro- Snail, SOX2, Sp1, Src, STAT3, 16.5 mg/kg; 20 mg/
pheochromocytoma; U87, K3, K4, K5, SSC-9 xenograft apoptosis survivin, TFAP2A, Tyro3, VEGF, kg; 25 mg/kg;
prostate cancer; skin CL1-5, CL1-0, TW01, mice; SUM-149 xeno- vimentin; Enhances cytochrome 25–100 mg/kg;
cancer TW06, NCI-H1299, graft mice; SW-780 c release, ROS accumulation; 50 mg/kg; 0.025%,
A549, H460, SCC-9, xenograft mice Induces mitochondrial depo- 0.05%; 0.06%
MIA PaCa-2, Panc-1, larization; Inhibits MAPK/ERK,
PC-12, BCaPT1, PI3K/Akt pathways; Reduces ATP
BCaPT10, BCaPM- levels; Represses DNA replica-
T10, LNCaP, A431, tion; Up-regulates Bax, CK1α,
SCC13 endostatin, microRNA-16, p21,
p53, TIMP-1, TIMP-2
Page 23 of 58
Luo et al. Chin Med
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
Berberine Coptidis Breast cancer; cervical MCF-7, MCF-7/ 22RV1 xenograft mice; Anti-angio- Activates caspase-3, -7, -8, -9, 0–10 µM; 0–20 μM; Caffeine; Cetuxi- [139, 140, 144,
(2019) 14:48
cgubebsus cancer; cholangio- HER2, MCF-7/ A2780 xenograft genesis; anti- PARP; Decreases mitochondrial 0–25 μM; 0–40 μM; mab; Doxoru- 145, 147–
Franch., carcinoma; colorectal TAM, MDA-MB157, mice; A549 xenograft proliferation; membrane potential, catalase 0–50 µM; 0–80 μM; bicin; Erlotinib; 149, 153,
Mahonia cancer; endometrial MDA-MB231, mice; BGC-823 xeno- anti-metastasis; and superoxide dismutase 0–90 µM; 0–100 µM; d-limonene; 171, 174,
bealei (Fort.) carcinoma; esopha- MDA-MB453, BT20, graft mice; Eca109 enhances activities; Down-regulates Akt, 0–120 µM; Niraparib; 835–870]
Carr., Phel- geal squamous BT549, Hs578T, xenograft mice; H22 radiosensitiv- AR, Bcl-2, Bcl-xL, Bid, β-catenin, 0–150 μM; Tamoxifen;
lodendron cancer; gastric cancer; T47D, SKBR3, BT474, xenograft mice; ity; induces N-cadherin, CDK1, CDK2, 0–160 µM; Taxol; TRAIL
chinense glioblastoma; head HeLa, SiHa, QBC939, HONE1 xenograft autophagy, CDK4, COX-2, PLA2, cyclin A1, 0–200 µM;
Schneid and neck cancer; KKU-213, KKU-214, mice; LoVo xenograft cell cycle cyclin B1, cyclin D1, cyclin E, 0–250 μM;
hepatocellular carci- SW-480, SW-620, mice; LNCaP xeno- arrest; inhibits DHCR24, DHFR, E2F1, EBNA1, 0–350 μM;
noma; leukemia; lung HT-29, DLD-1; graft mice; MDA- cell viability, EGFR, EF-Tu, ERK, Ezrin, FAK, FN, 0–1000 μM;
cancer; medulloblas- HCT-116, LS174T, MB-231 xenograft epithelial– HER2, HIF-1α, HMGB1, HNF4α, 10–80 μM; 15 µM;
toma; melanoma; LoVo, Eca109, TE13, mice; Medulloblas- mesenchymal ITGβ1, Jak2, JNK, Mcl-1, MEK, 20 μM; 50 μM;
nasopharyngeal KYSE-70, EAC, toma xenograft mice; transition; pro- MMP-1, MMP-2, MMP-9, mTOR, 0–1 µg/ml; 0–80 μg/
carcinoma; oral squa- SKGT4, AN3 CA, MHCC97L xenograft apoptosis c-Myc, NANOG, NF-κB, iNOS, ml; 5 mg/kg; 10 mg/
mous cell carcinoma; HEC-1-A, KLE, MGC- mice; SGC-7901 occludin, Oct-4, p38, p50, p62, kg; 12.5–50 mg/
osteosarcoma; ovar- 803, SGC-7901, AGS, xenograft mice; p100, p105, p70S6 K, paxillin, kg; 20 mg/kg;
ian cancer; pancreatic BGC-823, MKN45, SW-620 xenograft u-PA, PCNA, PDK1, P GE2, PKC-α, 50, 100 mg/
cancer; prostate U87, U251, U118, mice; SiHa xenograft PSA, PTEN, PTTG-1, RAD51, kg; 50–200 mg/
cancer; skin cancer; SHG-44, FaDu, H22, mice; U87 xenograft b-Raf, c-Raf, Septin-8, Slug, Snail, kg; 200 mg/kg;
uterine leiomyoma Hepa1-6, HepG2, mice SOX2, Sp1, Src, STAT3, survivin, 0.01136 g/kg
Bel-7404, Huh7, UQCRC1, VEGF, vimentin, Wnt5α,
WRL68, MHCC97L, ZEBRA; Enhances cytochrome c
K562, A549, B16F10, release, ROS accumulation, SSAT
HONE1, HK1-EBV, activity; Induces DNA damage;
CNE-2, KB, U2OS, Inhibits Akt/mTOR/p70S6 K/
Panc-1, MIA PaCa-2, S6, arachidonic acid metabolic,
LNCaP, DU-145, androgen receptor pathways;
LAPC-4, PC-3, Reduces NO production; Sup-
22RV1, C4-2B, C42, presses Hedgehog signaling
RM-1, A-431 pathway; Up-regulates ACC, AIF,
AMPKα, Apaf-1, ATF-6, Bad, Bak,
Bax, Beclin-1, Bim, E-cadherin,
DR5, FasL, FoxO1, FoxO3a,
GRP78, HRK, Lig4, MST1, p21,
p27, p53, PHLPP2, SSAT, TIMP-2,
TRAIL, ULK1
Page 24 of 58
Luo et al. Chin Med
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
(2019) 14:48
Artemisinins Artemisia Breast cancer; cervical MCF-7, MDA-MB-231, A549 xenograft mice; Anti-metastasis; Activates caspase-3, -8, -9, PARP; 0–75 μM; 0–160 μM; 3CA; Halofugi- [184, 186, 213,
annua L. cancer; colorectal HeLa, HCT-116, BE(2)-C xenograft anti-prolifera- Decreases mitochondrial mem- 0–200 μM; none; 871–883]
cancer; gallbladder SW-480, SW-620, mice; C6 xenograft tion; induces brane potential, MMP activity; 0–250 μM; Holotransferrin;
cancer; gastric cancer; GBC-SD, NOZ, mice; GBC-SD apoptosis, Down-regulates Bcl-2, CDK2, 0–400 μM; Resveratrol
glioma; hepatocel- MGC-803, C6, xenograft mice; HCT- autophagy, cell CDK4, cyclin D1, cyclin E2, Dvl2, 0–500 μM;
lular carcinoma; HepG2, Hep3B, 116 xenograft mice; cycle arrest; ERK1/2, LRP6, MMP-2, NANOG, 0–1000 μM;
Ishikawa endometrial SMMC-7721, Ishi- HepG2 xenograft inhibits cell Oct-4, p38, p62, SOX2, vimentin, 0–1200 μM;
cancer; lung cancer; kawa, A375, A549, mice; NOZ xenograft viability Wnt5α/β; Enhances cytochrome 10–320 μM;
neuroblastoma; oral ASTC-a-1, H1299, mice c release, ROS accumulation; 40–160 μM;
carcinoma; pancreatic BE(2) -C, SHEP1, Induces DNA damage; Inhibits 0–40 μg/ml; 10 mg/
cancer SK-N-AS, SK-N-DZ, Wnt/β-catenin signaling path- kg; 50 mg/kg;
SCC25, RIN way; Up-regulates Axin2, Bax, 60 mg/kg; 100 mg/
E-cadherin, β-catenin, NKD2, kg
p16, TIMP-2
Ginsenoside Panax Breast cancer; colorectal BT549, MDA-MB-231, A375 xenograft mice; Anti-angio- Activates caspase-3, -8, -9, 12, 0–10 μM; 0–30 μM; Cisplatin; Cyclo- [227, 232–234,
Rg3 notoginseng cancer; esophageal MDA-MB-453, A549 xenograft mice; genesis; anti- PARP; Decreases mitochondrial 0–35 μM; 0–60 μM; phosphamide; 236–241,
(Burk.) F. H. carcinoma; gallblad- CT-26, HCT-116, BxPC-3 xenograft proliferation; membrane potential; Down- 0–80 μM; 0–100 μM; Erlotinib; 246, 252–
Chen, Panax der cancer; gastric LoVo, SW-480, mice; CT-26 xeno- anti-metastasis; regulates Akt, AQP1, B7-H1, 0–150 μM; 5-Fluorouracil; 255, 260,
ginseng, Cin- cancer; glioblastoma; SW-620, EC109, graft mice; GBC-SD enhances B7-H3, Bcl-2, Bcl-xL, VE-cadherin, 0–160 μM; Oxaliplatin; 884–900]
namomum glioma; hepatocel- KYSE170, TE1, xenograft mice; radiosensitiv- CDK2, COX-2, CXCR4, cyclin 0–200 μM; Paclitaxel
cassia Presl. lular carcinoma; leu- GBC-SD, Mz-ChA-1, HCT-116 xenograft ity; increases D1, cyclin E, DNMT3A, EGFR, 0–400 μM;
kemia; lung cancer; QBC939, SGC-7901, mice; Hep1-6 cell survival; EPHA2, ERK, FUT4, HDAC3, 0–600 μM;
melanoma; multiple U87MG, U87, Hep1- xenograft mice; H23 induces HIF-1α, HK2, IAP, JNK, LeY, 25 μM; 0–600 ng/
myeloma; ovarian 6, HepG2, Lewis, xenograft mice; autophagy, MMP-2, MMP-9, mTOR, c-Myc, ml; 0–80 μg/
cancer; pancreatic Jurkat, A549, A549/ Lewis tumor-bearing cell cycle NF-κB, p38, p53, PCNA, PD-L1, ml; 0–100 μg/
cancer; prostate DDP, H23, H1299, mice; LoVo xenograft arrest; inhibits PI3K, PKM2, Rb, STAT3, surviving, ml; 0–160 μg/ml;
cancer A375, C8161, SK- mice; MDA-MB-231 chemotaxis, VEGF; Enhances cytochrome c 0–200 μg/ml; 40,
MEL-28, RPMI 8226, xenograft mice; MCF- epithelial– release, ROS production; Inhibits 80 μg/ml; 50 μg/ml;
SKO-007, U266, 7 xenograft mice; mesenchymal the Warburg effect, Wnt/β- 80, 160 μg/ml; 80,
A2780, 3AO, SKOV3, SKOV3 xenograft transition; pro- catenin pathway; Up-regulates 160 mg/ml; 3 mg/
AcPC-1, BxPC-3, mice; SW1990 xeno- apoptosis Atg-5, Atg-7, Bax, CHOP, IRE1, kg; 5 mg/kg; 5, 10,
Panc-1, SW1990, graft mice; SW-620 microRNA-532-3p, p16, p21, 20 mg/kg; 6 mg/
PC-3 xenograft mice p27, p53, PERK kg; 7.5–30 mg/kg;
10 mg/kg; 20 mg/kg
Page 25 of 58
Luo et al. Chin Med
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
(2019) 14:48
Ursolic acid Vaccinium Bladder cancer; breast BIU-87, T24, MDA- 12-dimethylbenz[a] Anti-angio- Activates caspase-3, -7, -8, -9, 0–4 μM; 0–16 μM; Capecitabine; [274, 276, 281,
macrocarpon cancer; cervical MB-231, MCF-7, anthracene-induced genesis; Fas receptor, PARP; Decreases 0–17.5 μM; 5-Fluorouracil; 283–289,
Ait., Arcto- cancer; colorectal MCF-7/ADR, HeLa, mice; DU-145 xeno- anti-metastasis; mitochrondrial membrane 0–20 μM; 0–40 μM; Oxaliplatin; 293,
staphylos cancer; Ehrlick ascites HCT-8, HCT-116, graft mice; Ehrlich anti-prolifera- potential; Down-regulates 0–50 μM; 0–80 μM; Resveratrol; 901–907]
uva-ursi (L.) carcinoma; leukemia; HT-29, Caco-2, ascites carcinoma tion; enhances AEG-1, Akt, Bcl-2, Bcl-xL, Bid, 0–100 μM; 4 μM; TRAIL
Spreng, Rho- liver cancer; lung SW-480, SW-620, xenograft mice; HCT- chemosensitiv- β-catenin, CD31, cyclin D1, 20 μM; 0–400 μg/
dodendron cancer; melanoma; HCT-15, CO115, 116 xenograft mice; ity; induces EGFR, ERK, cFLIP, FN, HIF-1α, ml; 10 mg/kg;
hymenanthes ovarian cancer; HL-60, HL-60/ADR, HCT-15 xenograft apoptosis, cIAP-1, ICAM-1, IκBα, IKKα/β, IL-8, 25–100 mg/kg;
Makino, prostate cancer; skin Jurkat, K562, K562/ mice; U937 xenograft autophagy, cell Jak2, Ki-67, Mcl-1, MMP-2, MMP- 50 mg/kg; 75 mg/
Eriobotrya cancer ADR, U937, HL-60/ mice cycle arrest; 9, NF-κB, iNOS, p65, u-PA, P-gp, kg; 250 mg/kg;
japonica, ADR, Hep3B, Huh7, inhibits MDR S6 K, Src, STAT3, survivin, mTOR, 2 μmol/mouse
Rosemarinus HA22T, A549, TNF-α, VEGF, Wnt5α/β, XIAP;
officinalis, H3255, Calu-6, Enhances cytochrome c release,
Calluna M4Beu, SKOV3, PGE2 levels, ROS production;
vulgaris, DU-145, LNCaP, Inhibits NO production; Up-
Eugenia PC-3 regulates ACC, AMPK, ASK1, Bax,
jambolana, CHOP, DR4, DR5, eIF2α, GRP78,
Ocimum GSK3β, IL-12, JNK, c-Jun, NADPH,
sanctum p21, p52, p53, PERK
Silibinin Silybum Breast cancer; colorectal BT-20, MCF-7, MDA- 786-O xenograft mice; Anti-metastasis; Activates caspase-3, -8, -9, PARP; 0–75 μM; 0–100 μM; Curcumin; [318, 319,
marianum L. cancer; epidermoid MB-231, MDA- Azoxymethane- anti-prolifera- Down-regulates Akt, Bcl-2, EGFR, 0–200 μM; luteolin 329–331,
Gaertn carcinoma; glioblas- MB-468, SKBR3, induced rats; tion; induces ERK, GLI1, IL-1β, FN, MMP-2, 0–300 μM; 334,
toma; hepatocellular T47D, AsPC-1, BxPC- Diethylnitrosamine- apoptosis, MMP-7, MMP-9, NF-κB, iNOS, 0–800 μM; 25, 342–345,
carcinoma; osteo- 3, Panc-1, HT-29, induced mice autophagy, cell PLA2, TNF-α, mTOR; Enhances 50 μM; 120 μM; 357, 358,
sarcoma; pancreatic HCT-116, LoVo, cycle arrest; CYP2E1 activity, cytochrome c 125 μM; 200 mg/kg; 908–910]
cancer; prostate can- SW-480, Caco-2, inhibits cell release, ROS production; Up- 300 mg/kg; 0.5%
cer; renal carcinoma; A-431, LN18, SNB19, viability regulates AIF, Bax, Bid, calpain,
thyroid cancer U87MG, Hep3B, EGR1, ICAD, NAG-1, PTEN
HepG2, SK-Hep-1,
SaOS2, PC-3, 769-P,
786-O, ACHN,
OS-RC-2, SW839,
Caki, TPC-1
Page 26 of 58
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
Luo et al. Chin Med
effects agents
Emodin Rheum Bladder cancer; breast MBT2, T24, TSGH8301, 4T1 xenograft mice; Anti-metastasis; Activates caspase-3, -9, PARP, 0–10 μM; 0–40 μM; Cisplatin; [64, 367–378,
palmatum, cancer; colorectal 4T1, EO771, MCF-7, 7,12-dimethyl anti-prolifera- chloride currents; Decreases 0–50 μM; 0–60 μM; curcumin; 380, 382,
Polygonum cancer; gallbladder MDA-MB-231, benz(a)anthracene- tion; induces mitochondrial membrane 0–80 μM; 0–100 μM; 5-fluorouracil; 383, 385,
cuspidatum, cancer; gastric cancer; MDA-MB-435, induced golden apoptosis, potential; Down-regulates Akt, 0–250 μM; gemcitabine 389, 394,
Polygonum hepatocellular carci- MDA-MB-453, HCT- Syrian hamsters; autophagy, Bcl-2, Bcl-xL, Bim-1, β-catenin, 0–320 μM; 395, 402,
(2019) 14:48
multiflorum, noma; lung cancer; 116, LoVo, LS1034, EO771 xenograft cell cycle CDK1, CSF1, CSF2, CXCL12, 0–1000 μM; 20 μM; 403, 405,
Cassia nasopharyngeal SGC-996, MKN45, mice; HCCLM3 arrest; inhibits CXCR4, cyclin D1, ERα, ERK, 20–80 μM; 40 μM; 911]
obtusifolia carcinoma; oral car- C3A, Hep3B, tumor-bearing mice; cell viability, FABP4, bFGF, HBP17, HER2, ILK, 0.05 mM; 40 mg/
cinogenesis; ovarian HepG2, PLC/PRF/5, LS1034 xenograft epithelial- Jagged1, Jak1, Jak2, Ki-67, Mcl-1, ml; 20, 40 mg/kg;
cancer; pancreatic SMMC-7721, A549, mice; MDA-MB-231 mesenchymal MCP-1, MMP-2, MMP-9, MRP1, 20, 50 mg/kg; 25,
cancer; prostate CNE-2Z, A2780, xenograft mice; transition NF-κB, p38, p62, u-PA, u-PAR, 50 mg/kg; 40 mg/
cancer SKOV3, AsPC-1, SGC-996 xenograft Slug, Snail, Src, STAT3, survivin, kg; 50 mg/kg
BxPC-3, Panc-1, mice; SKOV3 xeno- Thy-1, VEGF, vimentin, XIAP,
SW1990, SW1990/ graft mice; SW1990 ZEB1; Enhances C a2+ levels,
GZ, PC-3 xenograft mice; T24 cytochrome c release, ROS
xenograft mice production; Up-regulates AIF,
Bax, Beclin-1, E-cadherin, GSK3β,
microRNA-34, Notch1, SHP-1
Triptolide Tripterygium Bladder cancer; breast UMUC3, MDA- 3LL xenograft mice; Anti-angio- Activates caspase-3, -7, -8, 0–10 nM; 0–40 nM; Cisplatin; [408, 410, 411,
wilfordii cancer; colorectal MB-231, MCF-7, A549 xenograft mice; genesis; -9, GSK3β, PARP; Decreases 0–50 nM; 0–80 nM; epirubicin; 414, 415,
Hook. F. cancer; endome- DLD-1, HCT-116, AsPC-1 xenograft anti-metastasis; mitochondrial membrane 0–100 nM; 5-fluorouracil; 417, 419,
trial carcinoma; HEC-1B, MHCC-97H, mice; BE(2)-C anti-prolifera- potential; Down-regulates Akt, 0–160 nM; gemcitabine; 422, 423,
liver cancer; lung HepaRG, HepG2, xenograft mice; tion; enhances AR, BCAR1, Bcl-2, β-catenin, 0–200 nM; hydroxycamp- 425–427,
cancer; lymphoma; H460, H358, CNE xenograft mice; radiosensitiv- Cav-1, CD147, CDK2, CHK1, COX 0–300 nM; tothecin 429, 431–
melanoma; myeloma; A549, A549/Taxol, Daudi xenograft ity; induces IV, CXCR4, cyclin A1, ERK, ETS2, 0–320 nM; 434, 438,
nasopharyngeal HTB182, BEAS-2B, mice; H358 xenograft autophagy, cell FAK, c-FLIP, GRB2, HIF-1α, HSF1, 0–400 nM; 444, 446,
carcinoma; neuroblas- H1299, NCI- mice; H460 xenograft cycle arrest; HSP70, IκBα, ITGβ1, ITGαVβ6, 0–500 nM; 453, 454,
toma; osteosarcoma; H2009, NCI-H460, mice; HEC-1B xeno- inhibits cell JMJD3, JMJD2B, NK, p38 MAPK, 0–0.1 μM; 0–25 μM; 912–925]
ovarian cancer; oral Jurkat, Molt-3, Raji, graft mice; Jurkat viability; pro- Mcl-1, MKP-1, MMP-2, MMP-3, 0–150 μM;
cancer; pancreatic NAMALWA, Daudi, xenograft mice; apoptosis MMP-7, MMP-9, MMP-14, MMP- 0–200 μM; 10 nM;
cancer; prostate B16F10, HS-sultan, MHCC-97H xenograft 19, c–Myc, NF-κB, iNOS, Nrf2, 50, 72 nM; 100 nM;
cancer IM9, RPMI 8226, mice; SAS + U937 p65, PCNA, PI3K, PYK2, ROCK1, 0–8 ng/ml; 0–36 ng/
U266, CNE, MG63, xenograft mice; RhoA, Slug, Snail, SOS1, Src, ml; 0–50 ng/ml;
BE(2)-C, SH-SY5Y, SKOV3/DDP xeno- survivin, mTOR, Twist, UTX, VEGF, 0–400 ng/ml; 5,
SAOS2, U2OS, graft mice; SW1990 vimentin, ZEB1; Enhances C a2+ 10 ng/ml; 5–160 ng/
SKOV3, SKOV3/DDP, xenograft mice levels, cytochrome c release, ml; 8 ng/ml; 250 μg/
A2780, SAS, Panc-1, ROS production; Inhibits Wnt/β- kg; 0–0.8 mg/kg;
AsPC-1, SW1990, Catenin pathway; Up-regulates 0.04–0.36 mg/
BxPC-3, LNCaP, ATM, Bax, Beclin-1, E-cadherin, kg; 0.075 mg/
PC-3, DU-145 cathepsin B, Fas, DKK1, DR5, kg; 0.15 mg/kg;
ENY2, FADD, FRZB, GSK3β, IL-2, 0.25 mg/kg; 0.4 mg/
γ-H2AX, LMP, LSD1, p53, PPARγ, kg; 1 mg/kg;
PTEN, SFRP1, SIRT3, Smac, 1.5 mg/kg; 2–4 μg/
SUV39H1, TNF-α, Wnt3α mouse
Page 27 of 58
Luo et al. Chin Med
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
(2019) 14:48
Cucurbitacin B Bryonia, Breast cancer; cervical 4T1, HCC1937, 4-(methylnitrosamino)- Anti-angiogen- Activates caspase-3, -8, -9, PARP; 0–100 nM; 0–200 nM; Curcumin; [452, 460–462,
Cucumis, cancer; hepatocellular MCF-7, MCF-7/ 1-(3-pyridyl)-1- esis; Anti- Decreases mitochondrial 0–1000 nM; 0.1– docetaxel; 472–475,
Cucurbita carcinoma; lung can- ADR, MDA-MB-231, butanone-induced metastasis; membrane potential; Down- 1000 nM; 0–0.1 μM; gefitinib; 485, 499,
and Lepidium cer; neuroblastoma; MDA-MB-436, mice; 4T-1 xenograft Anti-prolifera- regulates Akt, ACLY, BCAR1, 0–1 μM; 0–1.6 μM; gemcitabine 926–931]
sativum prostate cancer SKBR-3, HeLa, T47D, mice; Bel-7402 xeno- tion; Inducing Bcl-2, β-catenin, CD31, CDK1, 0–30 μM; 0–100 μM;
SK-Hep1, Hep3B, graft mice; MDA- apoptosis, CIP2A, cyclin B1, cyclin D1, EGFR, 0–128 μM; 0.02–
HepG2, Bel-7402, MB-231 xenograft cell cycle ERK, FAK, galectin-3, GSK3β, 62.5 μM; 0–100 μg/
Bel-7402/5-Fu, mice; NNK-induced arrest; Inhibits HER2, HIF-1α, ILK1, ITGA6, ITGB4, ml; 0.1–100 μg/ml;
A549, H1299, H23; mice; PC-3 xenograft epithelial- Jak2, MMP-2, MMP-9, MRP1, 0.1, 0.2 mg/kg; 0.1,
SH-SY5Y; LNCaP, mice mesenchymal c-Myc, nucleophosmin, P-gp, 0.25 mg/kg; 0.5,
PC-3 transition paxillin, RhoA, ROCK1, STAT3, 1 mg/kg; 1, 5 mg/
Src, survivin, TACE, TCF1, mTOR, kg; 2 mg/kg; 10 mg/
Twist, VEGF, VEGFR2, Wnt3; kg; 0.1 μmol/mouse
Enhances cytochrome c release,
PP2A activity, ROS production;
Inhibits Wnt/β-catenin pathway;
Up-regulates ATM, Bax, Bim,
E-cadherin, CDC25C, CHK1,
γ-H2AX, JNK, p21, p53
Tanshinone IIA Salvia miltior- Breast cancer; bladder BT-20, 5637, BFTC 905, HT-29 xenograft mice; Anti-angio- Activates caspase-3, -8, -9, -12, 0–8 μM; 0–20 µM; Adriamycin [514, 515, 517,
rhiza Bunge cancer; cervical can- T24, HeLa, C33 A, MKN45 xenograft genesis; PARP; Down-regulates ALDH1, 0–40 µM; 0–60 µM; 5-fluorouracil; 519, 523,
cer; colorectal cancer; HCT-116, COLO- mice; SGC-7901 anti-metastasis; Bcl-2, BIP, N-cadherin, β-catenin, 0–80 μM; 0–100 µM; TRAIL 531, 539,
esophageal carci- 205, LoVo, HT-29, xenograft mice; 143B anti-prolifera- CD31, COX-2, CTGF, FoxM1, 0–54.4 μM; 0–20 ng/ 932–935]
noma; gastric cancer; SW-620, Eca109, xenograft mice tion; enhances HIF-1α, Ki-67, LEF1, MCP-1, Mfn- ml; 0–4 µg/ml;
NSCLC; osteosarcoma; SGC-7901, MKN45, chemosensitiv- 1, Mfn-2, MMP-2, MMP-9, c-Myc, 0–8 μg/ml; 0–18 µg/
oral squamous A549, H596, H1299, ity, radiosensi- NANOG, Opa-1, p65, PCNA, ml; 0–60 µg/ml;
carcinoma Calu-1, H460, 143B, tivity; induces Slug, Snail, STAT3, survivin, TCF3, 1 mg/kg; 10, 30 mg/
SCC090 autophagy, VEGF, vimentin, YAP; Enhances kg; 20 mg/kg
cell cycle cytochrome c release, ROS
arrest; inhibits accumulation; Reduces mito-
cell viability, chondrial membrane potential;
epithelial– Up-regulates ATF-4, Bax, Bak,
mesenchymal Bad, E-cadherin, CHOP, Drp-1,
transition; pro- DR5, GRP78, p21
apoptosis
Page 28 of 58
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
Luo et al. Chin Med
Oridonin Rabdosia Breast cancer; cervical 4T1, MCF-7, 143B xenograft mice; Anti-angio- Activates caspase-3, -8, -9, PARP; 0–1000 nM; 0–1.5 μM; Cisplatin; [544–556,
rubescens cancer; colorectal MDA‑MB‑231, 4T1 xenograft mice; genesis; Decreases mitochondrial 0–4 μM; 0–9 μM; NVP-BEZ235; 558–567,
(Hemsl.) cancer; esophageal SW-48, SW-480, HCT-116 xenograft anti-metastasis; membrane potential; Down- 0–12 μM; 0–15 μM; valproic acid 573–576,
Hara cancer; gastric SW-620, SW-1116, mice; HepG2 anti-prolifera- regulates Akt, AMPK, AP-1, Bcl-2, 0–20 μM; 0–25 μM; 578, 579,
cancer; hepatocellular HeLa, LoVo, xenograft mice and tion; induces Bcl-xL, N-cadherin, CD31, CD44, 0–30 μM; 0–32 μM; 936–946]
carcinoma; laryngeal; HCT-116, HCT-15, zebrafish; HL-60 apoptosis, CDC25C, CDK1, CDK2, Claudin 1, 0–40 μM; 0–50 μM;
xenograft mice; HOS autophagy, cell Claudin 4, Claudin 7, α-CPI, cyc-
(2019) 14:48
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
(2019) 14:48
Gambogic acid G. hanburyi, G. Breast cancer; colorectal 4T1, MCF-7, MDA- 4T1 xenograft mice; Anti-angio- Activates caspase-3, -7, -8, -9, 200–400 nM; Chlorochalcone; [639, 644, 646,
Morella cancer; glioma; hepa- MB-231, HCT-15, A549 xenograft mice; genesis; PARP, JNK pathway; Decreases 0–1 μM; 0–2 μM; Cisplatin; 647, 650,
tocellular carcinoma; HCT-15R, HCT-116, B16F10 and MC38 anti-metastasis; mitochondrial membrane 0–3 μM; 0–5 μM; Doxorubicin; 656, 657,
NSCLC; osteosarcoma; HT-29, SW-480, xenograft mice; anti-prolifer- potential; Down-regulates Akt, 0–8 μM; 0–10 μM; 5–Fluorouracil; 663–665,
ovarian cancer; SW-620, LoVo/L- BxPC-3 xenograft ation; anti- ALDOA, ATG4B, Bcl-2, Bcl-xL, 0–40 μM; 0–50 μM; Gemcitabine; 953–966]
pancreatic cancer; OHP, LoVo/L-OHP/ mice; C26 xenograft tumor growth; β-catenin, cFLIPL, cyclin D1, 0–51.8 μM; 0.5 µM; Nal131; Oxalipl-
prostate cancer; renal GA, T98G, Hep3B, mice; SKOV3 xeno- enhances DLL1, DLL3, DLL4, ERK, Jagged1, 0–3 μg/ml; 2 mg/kg; atin; Retinoic
carcinoma Huh7, A549, A549/ graft mice chemosensitiv- Jagged2, LRP, p-53, P-gp, Mcl-1, 8 mg/kg acid; TRAIL
DDP, SPC-A-1, ity; induces MMP-2, MMP-9, MRP2, PI3K,
MG63, SKOV3, apoptosis, RRM2, SIRT1, survivin, TOPIIα,
BxPC-3, Capan-1, autophagy, VEGF, XIAP; Enhances ROS
Capan-2, Colo-357, cell cycle accumulation, cytochrome c
MIA PaCa-2, Panc-1, arrest; inhibits release; Inhibits ERK/E2F1/RRM2,
Suit-007, Suit-2, cell viability, MAPK, PI3K/Akt pathways, NF-κB
SW1990, B6WT, survival p65 binding activity, Trx activity;
DU-145, LAPC-4, Up-regulates AIF, Atg-5, Bax,
LNCaP, PC-3, PCAP- CHOP, DUSP1, DUSP5, FoxO3a,
1, PTEN−/−/p53−/−, c-Jun, p27, p53
Caki
Artesunate Artemisia B-cell lymphoma; blad- BL-41, Raji, Ramos, BL-41 xenograft mice; Anti-angio- Activates caspase-3, -9, p38 0.1–10 μM; 0–50 μM; Cisplatin; Con- [669, 673, 675,
annua L. der cancer; breast Rec-1, RT4, T24, A2780 xenograft genesis; MAPK pathway; Decreases 0–100 μM; nexin-43; 681, 683,
cancer; colorectal ACHN, BT-474, mice; HO8910 xeno- anti-metastasis; metabolic capacity, mitochon- 0–120 μM; Paclitaxel 688–690,
cancer; gastric cancer; MCF-7, MDA- graft mice; TE671 anti-prolifer- drial membrane potential, PGE2 0–200 μM; 693, 695,
head and neck MB-231, BGC-823, xenograft mice; ation; anti- production; Down-regulates 50 μM; 0–50 μg/ 967–971]
cancer; hepatocellular HGC-27, MGC-803, MCF-7 xenograft tumor growth; Bcl-2, CDC25A, COX-2, cyclin ml; 0–160 mg/L;
carcinoma; myelod- SGC-7901, HN3, mice induces B, cyclin D1, cyclin E2, γ-H2AX, 0–200 mg/kg;
ysplastic syndrome; HN4, HN9, SKM-1, apoptosis, cell IGF-1R, Keap1, c-Myc, PAX7, 50 mg/kg; 50,
ovarian cancer; HO8910, SKOV3, cycle arrest, RAD51, STAT3, UCA1, xCT; 150 mg/kg; 100 mg/
pancreatic cancer; AsPC-1, BxPC-3, DNA damage, Enhances ROS production; Up- kg; 200 mg/kg
prostate cancer; rhab- Colo-357, Panc-1, ferroptosis regulates ATF-4, ATM, ATR, Bax,
domyosarcoma DU-145, LNCaP, BRCA1, E-cadherin, CHK1, CHK2,
RD18, TE671 CHOP, HO-1, microRNA-16,
microRNA-133, microRNA-206,
Nrf2, p53
Page 30 of 58
Luo et al. Chin Med
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
Wogonin Scutellaria Breast cancer; gastric MDA-MB-231, BGC- MDA-MB-231 xenograft Anti-angio- Activates caspase-3, -4, -8, -9, -12, 0–20 μM; 0–40 μM; Cisplatin; [704, 708,
(2019) 14:48
baicalensis, cancer; head and 823, MFC, MGC-803, mice; Raji xenograft genesis; PARP, IRE1α-dependent path- 0–50 μM; 0–60 μM; Paclitaxel; 709, 716,
Scutellaria neck cancer; hepa- MKN45, SGC-7901, mice; AMC-HN4-cisR anti-metastasis; way; Decreases mitochondrial 0–80 μM; 0–100 μM; Oxaliplatin; 717, 719,
amoena, tocellular carcinoma; AMC-HN2, AMC- xenograft mice; anti-prolifer- membrane potential; Down- 0–150 μM; Sorafenib 721, 725,
Scutellaria leukemia; lymphoma; HN3, AMC-HN4, AMC-HN9-cisR xeno- ation; anti- regulates Akt, B7H1, Bcl-2, CDK4, 0–200 μM; 40 μM; 730, 731,
rivularis, melanoma; multiple AMC-HN5, AMC- graft mice; B16F10 tumor growth; CDK6, cyclin D1, cyclin E, EGFR, 50 μM; 0–40 μg/ 741, 742,
Anodendron myeloma; neuroblas- HN9, AMC-HN4- xenograft mice; induces ERK, HIF-1α, IL-8, IκB, IKKα, Ki-67, ml; 0–60 mg/kg; 972–976]
affine Druce toma; osteosarcoma; cisR, AMC-HN9-cisR, BGC-823 xenograft apoptosis, MMP-2, MMP-9, c-Myc, PDK1, 0–80 mg/kg; 8 mg/
ovarian cancer; SNU-1041, SNU- mice and zebrafish; autophagy, cell PI3K, Rac1, RAE-1ε, SGK1, ULK1, kg; 20 mg/kg;
pancreatic cancer; 1066, SNU-1076, MFC xenograft mice; cycle arrest, VEGF; Enhances calreticulin, 60 mg/kg; 12.5 ng/
NSCLC Bel-7402, Hep3B, RPMI 8226 xenograft ER stress, HMGB1, cytochrome c release, zebrafish
HepG2, SMMC- mice mitochondrial ROS accumulation; Inhibits
7721, K562, K562/ dysfunction; 5-LO/BLT2/ERK/IL-8/MMP-9,
A02, K562R, Raji, reverses drug NF-κB pathways; Up-regulates
B16F10, RPMI 8226, resistance ASK, Bax, Bid, GRP78, GRP94,
U266, IMR-32, IRE1α, JNK, p21, p53, PU.1,
SK-N-BE2, CD133+ PUMA
CAL72, A549,
A2780, Colo-357,
Panc-1
β-Elemene Curcuma Bladder cancer; bone PBC, Bcap37, A549 xenograft mice; Anti-angio- Activates caspase-3, -7, -8, -9, 0–25 μM; 0–1000 μM; Cisplatin; [746, 747, 749,
wenyujin neoplasms; breast MBA-MD-231, B16F10 xenograft genesis; -10; Down-regulates Akt, Bcl-2, 67.5–1000 μM; Paclitaxel; 752, 754,
Y. H. Chen cancer; cervical MCF-7, MCF-7/ADR, mice; BGC-823 xeno- anti-metastasis; β-catenin, CDC25C, CDK1, cyclin 0–40 μg/ Rapamycin 755, 762,
et C. Ling, cancer; gastric cancer; MCF-7/DOC, 5637, graft mice; Lewis anti-prolifer- B1, cyclin D1, endostatin, ERK, ml; 0–50 μg/ 977–987]
Rhizoma melanoma; NSCLC; SiHa, T-24, BGC-823, tumor-bearing mice; ation; anti- DNMT1, MMP-2, MMP-3, MMP- ml; 0–120 μg/
zedoariae, osteosarcoma; thyroid MKN45, SGC-7901, MG63 xenograft tumor growth; 9, MTA3, c-Myc, STAT3, Sp1, ml; 0–160 μg/
Curcuma cancer B16F10, A549, mice; U2OS xeno- enhances survivin, TCF7, TIMP-1, TIMP-2, ml; 0–200 μg/
Zedoary H358, H460, H1299, graft mice radiosensitiv- VEGF; Enhances ROS accumula- ml; 0–320 μg/
H1650, H1975, ity; induces tion; Induces polarization ml; 0–500 μg/
Lewis, PC9, MG63, apoptosis, from M2 to M1 macrophages; ml; 0–800 μg/ml;
U2OS, FTC-133 autophagy, cell Inhibits Wnt/β-catenin pathway; 0–0.16 mg/ml; 15,
cycle arrest; Up-regulates ATF-4, ATF-6, Bad, 30 μg/ml; 100 mg/
reverses chem- Bax, BTF, CHK2, CHOP, FoxO3a, ml; 1 mg/kg; 20 mg/
oresistance IGFBP1, IRE1α, p15, p21, p53, kg; 50 mg/kg;
Pak1, PAK1IP1, PERK, TOPIIα 75 mg/kg; 200 mg/
kg
Page 31 of 58
Luo et al. Chin Med
(2019) 14:48
Table 1 (continued)
Compounds Origins Cancer types In vitro models In vivo models Anti-cancer Underlying mechanisms Dosage Combinational References
effects agents
Cepharanthine Stephania Choroidal melanoma; MEL15-1, COLO-205, A549 xenograft mice; Anti-angio- Activates caspase-3, -9, PARP; 0–15 μM; 0–20 μM; Cisplatin; [777, 782–790,
cepharantha colorectal cancer; HCT-116 HT-29, NCI-H1975 xenograft genesis; Decreases mitochondrial mem- 0–80 μM; 0–100 μM; Dacomitinib; 794, 988]
Hayata, breast cancer; gastric SW-620, MCF-7, mice anti-metastasis; brane potential; Down-regulates 0–120 μM; 2–8 μM; Paclitaxel;
Stephania cancer; leukemia; MDA-MB-231, anti-prolifer- Akt, Bcl-2, Bcl-xL, CDK4, cyclin A, 4, 5 μM; 5–80 mM; TRAIL
japonica nasopharyngeal Jurkat T-cells, A549, ation; anti- cyclin D, c-FLIP, mTOR, p50, p52, 25 mg/kg; 50 mg/kg
carcinoma; NSCLC; H1299, HCC827, tumor growth; survivin; Enhances cytochrome
ovarian cancer; renal NCI-H1299, NCI- induces c release, ROS accumulation;
carcinoma H1650, NCI-H1975, apoptosis, Inhibits lysosomal cathepsin B
CNE-1, CNE-2, autophagy, cell and cathepsin D maturation,
A2780, A2780/Taxol, cycle arrest; Akt/mTOR, NF-κB, pathways;
CaOV-3, OVCAR3, Reverses multi- Up-regulates Atg-7, Bak, Bax,
Caki drug resistance Beclin1, DR5, p38 MAPK, Mcl-1,
p21Waf1/Cip1, p53
Page 32 of 58
Luo et al. Chin Med (2019) 14:48 Page 33 of 58
experimental models and conditions, pharmacological regulatory subunit 2; COX-2: cyclooxygenase-2; COX IV: cytochrome c oxidase
subunit 4; α-CP1: poly(rC)-binding protein 1; CSF: colony stimulating factor;
effects, as well as mechanistic actions of the natural com- CTGF: connective tissue growth factor; CTR1: copper transporter 1; CTTN:
pounds derived from Chinese herbal medicine. Despite cortactin; CXCL-12: C–X–C motif chemokine 12; CXCR4: C–X–C chemokine
the unique anti-cancer beneficial features of many com- receptor type 4; CYP2E1: cytochrome P450 2E1; DC: dendritic cell; DHA:
dihydroartemisinin; DHCR24: 24-dehydrocholesterol reductase; DHFR:
pounds derived from Chinese herbal medicine, their dihydrofolate reductase; DLL: delta-like canonical Notch ligand; DKK1:
clinical applications are disproportionally limited. As of Dickkopf-related protein 1; DNA: deoxyribonucleic acid; DNMT: DNA
2019, only preliminary clinical studies have been per- (cytosine-5)-methyltransferase; DR4: death receptor 4; DR5: death receptor 5;
Drp-1: dynamin-related protein 1; DUSP: dual-specificity phosphatase; Dvl2:
formed with artemisinins, emodin, cucurbitacins, tansh- dishevelled segment polarity protein 2; E2F1: E2F transcription factor 1;
iones, shikonin, and CEP in various cancers, without any EBNA1: Epstein–Barr nuclear antigen 1; EF-Tu: elongation factor thermo
approved clinical applications. The phase I safety studies unstable; EGCG: epigallocatechin gallate; EGFR: epidermal growth factor
receptor; EGFR-TKI: epidermal growth factor receptor-tyrosine kinase inhibitor;
of UA-liposomes, oridonin derivative (HAO472), and EGR1: early growth response protein 1; ENY2: enhancer of yellow 2 transcrip-
P-glycoprotein; PHLPP2: pH domain and leucine Rich repeat protein Competing interests
phosphatase 2; PLA2: phospholipase A2; PI3K: phosphoinositide 3-kinase; The authors declare that they have no competing interests.
PKC-α: protein kinase Cα; PKD1: polycystin 1; PKM2: pyruvate kinase isozyme
M2; PP2A: pyrophosphatase (inorganic) 2; PPARγ: peroxisome proliferator- Received: 20 August 2019 Accepted: 23 October 2019
activated receptor γ; PSA: prostate-specific antigen; PTEN: phosphatase and
tensin homolog; PTTG-1: pituitary tumor-transforming gene 1 protein; PU.1:
spleen focus forming virus proviral integration oncogene; PUMA: p53
upregulated modulator of apoptosis; PYK2: proline-rich tyrosine kinase 2; Rac1:
Ras-related C3 botulinum toxin substrate 1; Rac2: Ras-related C3 botulinum References
toxin substrate 2; RAE-1ε: ribonucleic acid export 1ε; Rb: retinoblastoma- 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global
associated protein; RECK: reversion-inducing-cysteine-rich protein with kazal cancer statistics 2018: GLOBOCAN estimates of incidence and
motifs; RECQL4: ATP-dependent DNA helicase Q4; RhoA: Ras homolog family mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin.
member A; RIP: receptor-interacting serine/threonine protein; RIPK1: 2018;68(6):394–424.
receptor-interacting serine/threonine protein kinase 1; RRM2: ribonucleotide 2. Wang S, Wu X, Tan M, Gong J, Tan W, Bian B, et al. Fighting fire with fire:
reductase regulatory subunit M2; ROCK1: Rho-associated protein kinase 1; poisonous Chinese herbal medicine for cancer therapy. J Ethnophar-
ROS: reactive oxygen species; S6: ribosomal protein S6; S6K: ribosomal protein macol. 2012;140(1):33–45.
S6 kinase; SERTAD1: SERTA domain-containing protein 1; SFRP1: secreted 3. Zhong Z, Yu H, Wang S, Wang Y, Cui L. Anti-cancer effects of Rhizoma
frizzled related protein 1; SFN: stratifin; SGK1: serum and glucocorticoid-regu- Curcumae against doxorubicin-resistant breast cancer cells. Chin Med.
lated kinase 1; SHH: sonic hedgehog; SHP-1: Src homology region 2 2018;13:44.
domain-containing phosphatase 1; SIRT: sirtuin; Smac: second mitochondria- 4. Sang W, Zhong Z, Linghu K, Xiong W, Tse AKW, Cheang WS, et al. Sieges-
derived activator of caspase; SMOX: spermine oxidase; SOS1: son of sevenless beckia pubescens Makino inhibits Pam3CSK4-induced inflammation
homolog 1; SOD: superoxide dismutase; SOX2: sex determining region in RAW 264.7 macrophages through suppressing TLR1/TLR2-mediated
Y-box 2; Sp1: specificity protein 1; SREBP1: sterol regulatory element-binding NF-kappaB activation. Chin Med. 2018;13:37.
protein 1; SSAT: spermidine/spermine N1-acetyltransferase; STAT: signal 5. Zhong Z, Zhang Q, Tao H, Sang W, Cui L, Qiang W, et al. Anti-inflamma-
transducer and activator of transcription; SUV39H1: suppressor of variegation tory activities of Sigesbeckia glabrescens Makino: combined in vitro
3-9 homolog 1; Suz12: suppressor of zeste 12 protein homolog; TACE: and in silico investigations. Chin Med. 2019;14:35.
TNF-α-converting enzyme; TAZ: tafazzin; TFAP2A: transcription factor 6. Shukla R, Chanda N, Zambre A, Upendran A, Katti K, Kulkarni RR, et al.
AP-2-alpha; TCF: T-cell factor; TGF-β: transforming growth factor-β; Th1: T Laminin receptor specific therapeutic gold nanoparticles (198AuNP-
helper type 1 cell; Th2: T helper type 2 cell; Thy-1: THYmocyte differentiation EGCg) show efficacy in treating prostate cancer. Proc Natl Acad Sci USA.
antigen 1; TIMP: TIMP metallopeptidase inhibitor; TLR: toll-like receptor; TNF: 2012;109(31):12426–31.
tumor necrosis factor; TOPK: T-LAK cell-originated protein kinase; TOPIIα: DNA 7. Zhang J, Zhou F, Wu X, Zhang X, Chen Y, Zha BS, et al. Cellular phar-
topoisomerase IIα; TRAF6: TNF receptor-associated factor 6; TRAIL: TNF‐related macokinetic mechanisms of adriamycin resistance and its modula-
apoptosis‐inducing ligand; TROP2: tumor-associated calcium signal transducer tion by 20(S)-ginsenoside Rh2 in MCF-7/Adr cells. Br J Pharmacol.
2; Treg: regulatory T cells; Trx: thioredoxin; TrxR: thioredoxin reductase; Tyro3: 2012;165(1):120–34.
tyrosine-protein kinase receptor; UA: ursolic acid; UAL: UA-lipsomes; UCA1: 8. Hsiao YT, Kuo CL, Chueh FS, Liu KC, Bau DT, Chung JG. Curcuminoids
urothelial cancer-associated 1; ULK-1: UNC-51-like autophagy activating kinase induce reactive oxygen species and autophagy to enhance apoptosis
1; UQCRC1: ubiquinol-cytochrome c reductase core protein 1; UTX: in human oral cancer cells. Am J Chin Med. 2018;46(5):1145–68.
ubiquitously transcribed tetratricopeptide repeat protein X-linked; VEGF: 9. Wang N, Tan HY, Li L, Yuen MF, Feng Y. Berberine and Coptidis Rhizoma
vascular endothelial growth factor; VEGFR2: vascular endothelial growth factor as potential anticancer agents: recent updates and future perspectives.
receptor 2; VHL: von Hippel–Lindau tumor suppressor; XBP-1: X-box binding J Ethnopharmacol. 2015;176:35–48.
protein 1; xCT: solute carrier family 7 member 11; XIAP: X-linked inhibitor of 10. Wang Z, Yin J, Li M, Shen J, Xiao Z, Zhao Y, et al. Combination of shikonin
apoptosis protein; WT1: Wilms tumor 1; YAP: Yes-associated protein 1; ZEB1: with paclitaxel overcomes multidrug resistance in human ovarian
zinc finger E-box binding homeobox 1; ZEBRA: BamHI Z Epstein–Barr virus carcinoma cells in a P-gp-independent manner through enhanced ROS
replication activator. generation. Chin Med. 2019;14:7.
11. Kharat M, Du Z, Zhang G, McClements DJ. Physical and chemical
Acknowledgements stability of curcumin in aqueous solutions and emulsions: impact of
We are grateful to the technical team of Institute of Chinese Medical Sciences pH, temperature, and molecular environment. J Agric Food Chem.
at University of Macau. 2017;65(8):1525–32.
12. Liu Y, Wang X, Zeng S, Zhang X, Zhao J, Zhang X, et al. The natural
Authors’ contributions polyphenol curcumin induces apoptosis by suppressing STAT3 signal-
YW and ZZ designed, organized, and supervised the study. HL, ZZ, HC, YG, PL, ing in esophageal squamous cell carcinoma. J Exp Clin Cancer Res.
LQ, MZ, QL, ZC, JZ, PY, and CG drafted the manuscript. CV, HL, and HC analyzed 2018;37(1):303.
the literature. CV and ZZ revised the manuscript. JW, CU, and SW participated 13. Liu D, You M, Xu Y, Li F, Zhang D, Li X, et al. Inhibition of curcumin on
in the revision. All authors read and approved the final manuscript. myeloid-derived suppressor cells is requisite for controlling lung cancer.
Int Immunopharmacol. 2016;39:265–72.
Funding 14. Samarghandian S, Azimi-Nezhad M, Farkhondeh T, Samini F. Anti-oxida-
This work was financially supported by the Macao Science and Technology tive effects of curcumin on immobilization-induced oxidative stress in
Development Fund (FDCT 071/2017/A2), and the Research Committee of the rat brain, liver and kidney. Biomed Pharmacother. 2017;87:223–9.
University of Macau (CPG2019-00006-ICMS). 15. Cai YY, Lin WP, Li AP, Xu JY. Combined effects of curcumin and
triptolide on an ovarian cancer cell line. Asian Pac J Cancer Prev.
Availability of data and materials 2013;14(7):4267–71.
Not applicable. 16. Jose A, Labala S, Venuganti VVK. Co-delivery of curcumin and STAT3
siRNA using deformable cationic liposomes to treat skin cancer. J Drug
Ethics approval and consent to participate Target. 2017;25(4):330–41.
Not applicable. 17. Ravindranathan P, Pasham D, Balaji U, Cardenas J, Gu JH, Toden S, et al.
A combination of curcumin and oligomeric proanthocyanidins offer
Consent for publication superior anti-tumorigenic properties in colorectal cancer. Sci Rep.
We declare that the Publisher has the Author’s permission to publish the 2018;8:13869.
relevant Contribution. 18. Siddiqui FA, Prakasam G, Chattopadhyay S, Rehman AU, Padder RA,
Ansari MA, et al. Curcumin decreases Warburg effect in cancer cells by
Luo et al. Chin Med (2019) 14:48 Page 35 of 58
down-regulating pyruvate kinase M2 via mTOR-HIF1 alpha inhibition. 38. Larasati YA, Yoneda-Kato N, Nakamae I, Yokoyama T, Meiyanto E, Kato
Sci Rep. 2018;8:8323. JY. Curcumin targets multiple enzymes involved in the ROS metabolic
19. Sivanantham B, Sethuraman S, Krishnan UM. Combinatorial effects of pathway to suppress tumor cell growth. Sci Rep. 2018;8(1):2039.
curcumin with an anti-neoplastic agent on head and neck squamous 39. Zhu GH, Dai HP, Shen Q, Ji O, Zhang Q, Zhai YL. Curcumin induces apop-
cell carcinoma through the regulation of EGFR-ERK1/2 and apoptotic tosis and suppresses invasion through MAPK and MMP signaling in
signaling pathways. Acs Comb Sci. 2016;18(1):22–35. human monocytic leukemia SHI-1 cells. Pharm Biol. 2016;54(8):1303–11.
20. Zou P, Zhang JR, Xia YQ, Kanchana K, Guo GL, Chen WB, et al. ROS 40. Liao H, Wang Z, Deng Z, Ren H, Li X. Curcumin inhibits lung cancer inva-
generation mediates the anti-cancer effects of WZ35 via activating sion and metastasis by attenuating GLUT1/MT1–MMP/MMP2 pathway.
JNK and ER stress apoptotic pathways in gastric cancer. Oncotarget. Int J Clin Exp Med. 2015;8(6):8948–57.
2015;6(8):5860–76. 41. Sun MX, Yu F, Gong ML, Fan GL, Liu CX. Effects of curcumin on the role
21. Guan F, Ding Y, Zhang Y, Zhou Y, Li M, Wang C. Curcumin sup- of MMP-2 in endometrial cancer cell proliferation and invasion. Eur Rev
presses proliferation and migration of MDA-MB-231 breast cancer Med Pharmacol Sci. 2018;22(15):5033–41.
cells through autophagy-dependent Akt degradation. PLoS ONE. 42. Chen J, Zhang L, Shu Y, Chen L, Zhu M, Yao S, et al. Curcumin analogue
2016;11(1):e0146553. CA15 exhibits anticancer effects on HEp-2 cells via targeting NF-
22. Khan MA, Zafaryab M, Mehdi SH, Ahmad I, Rizvi MM. Characterization kappaB. Biomed Res Int. 2017;2017:4751260.
and anti-proliferative activity of curcumin loaded chitosan nanoparti- 43. Li G, Wang Z, Chong T, Yang J, Li H, Chen H. Curcumin enhances the
cles in cervical cancer. Int J Biol Macromol. 2016;93(Pt A):242–53. radiosensitivity of renal cancer cells by suppressing NF-kappaB signal-
23. Silva G, Teixeira Lima F, Seba V, Mendes Lourenco AL, Lucas TG, de ing pathway. Biomed Pharmacother. 2017;94:974–81.
Andrade BV, et al. Curcumin analog CH-5 suppresses the proliferation, 44. Schwertheim S, Wein F, Lennartz K, Worm K, Schmid KW, Sheu-Grabel-
migration, and invasion of the human gastric cancer cell line HGC-27. lus SY. Curcumin induces G2/M arrest, apoptosis, NF-kappaB inhibition,
Molecules (Basel, Switzerland). 2018;23:2. and expression of differentiation genes in thyroid carcinoma cells. J
24. Starok M, Preira P, Vayssade M, Haupt K, Salome L, Rossi C. EGFR inhibi- Cancer Res Clin Oncol. 2017;143(7):1143–54.
tion by curcumin in cancer cells: a dual mode of action. Biomacromol. 45. Montazeri M, Pilehvar-Soltanahmadi Y, Mohaghegh M, Panahi A, Khodi
2015;16(5):1634–42. S, Zarghami N, et al. Antiproliferative and apoptotic effect of dendroso-
25. Gaikwad D, Shewale R, Patil V, Mali D, Gaikwad U, Jadhav N. Enhance- mal curcumin nanoformulation in P53 mutant and wide-type cancer
ment in in vitro anti-angiogenesis activity and cytotoxicity in lung can- cell lines. Anticancer Agents Med Chem. 2017;17(5):662–73.
cer cell by pectin-PVP based curcumin particulates. Int J Biol Macromol. 46. Fu H, Wang C, Yang D, Wei Z, Xu J, Hu Z, et al. Curcumin regulates pro-
2017;104(Pt A):656–64. liferation, autophagy, and apoptosis in gastric cancer cells by affecting
26. Liao W, Xiang W, Wang FF, Wang R, Ding Y. Curcumin inhibited growth PI3K and P53 signaling. J Cell Physiol. 2018;233(6):4634–42.
of human melanoma A375 cells via inciting oxidative stress. Biomed 47. Ciolac OA, Filippi A, Maru N, Popa M, Chifiriuc MC, Ganea C, et al. Reduc-
Pharmacother. 2017;95:1177–86. tion of the clonogenic potential and collapse of the mitochondrial
27. Chen P, Huang HP, Wang Y, Jin J, Long WG, Chen K, et al. Curcumin membrane potential in A-431 epidermoid carcinoma cell line induced
overcome primary gefitinib resistance in non-small-cell lung cancer by curcumin. Rom Biotechnol Lett. 2017;22(6):13068–75.
cells through inducing autophagy-related cell death. J Exp Clin Cancer 48. Liu F, Gao S, Yang Y, Zhao X, Fan Y, Ma W, et al. Curcumin induced
Res: CR. 2019;38(1):254. autophagy anticancer effects on human lung adenocarcinoma cell line
28. Zhu J, Zhao B, Xiong P, Wang C, Zhang J, Tian X, et al. Curcumin induces A549. Oncol Lett. 2017;14(3):2775–82.
autophagy via inhibition of yes-associated protein (YAP) in human 49. Zhao G, Han X, Zheng S, Li Z, Sha Y, Ni J, et al. Curcumin induces
colon cancer cells. Med Sci Monit. 2018;24:7035–42. autophagy, inhibits proliferation and invasion by downregulating
29. Hu P, Ke C, Guo X, Ren P, Tong Y, Luo S, et al. Both glypican-3/Wnt/beta- AKT/mTOR signaling pathway in human melanoma cells. Oncol Rep.
catenin signaling pathway and autophagy contributed to the inhibitory 2016;35(2):1065–74.
effect of curcumin on hepatocellular carcinoma. Digest Liver Dis. 50. Seo SU, Woo SM, Lee HS, Kim SH, Min KJ, Kwon TK. mTORC1/2 inhibitor
2019;51(1):120–6. and curcumin induce apoptosis through lysosomal membrane perme-
30. Liu F, Gao S, Yang Y, Zhao X, Fan Y, Ma W, et al. Antitumor activity of abilization-mediated autophagy. Oncogene. 2018;37(38):5205–20.
curcumin by modulation of apoptosis and autophagy in human lung 51. Milano F, Mari L, van de Luijtgaarden W, Parikh K, Calpe S, Krishnadath
cancer A549 cells through inhibiting PI3K/Akt/mTOR pathway. Oncol KK. Nano-curcumin inhibits proliferation of esophageal adenocarci-
Rep. 2018;39(3):1523–31. noma cells and enhances the T cell mediated immune response. Front
31. Tong WH, Wang Q, Sun DH, Suo J. Curcumin suppresses colon cancer Oncol. 2013;3:137.
cell invasion via AMPK-induced inhibition of NF-B, uPA activator and 52. Kang S, Oh SC, Min BW, Lee DH. Transglutaminase 2 regulates self-
MMP9. Oncol Lett. 2016;12(5):4139–46. renewal and stem cell marker of human colorectal cancer stem cells.
32. Chen QY, Zheng Y, Jiao DM, Chen FY, Hu HZ, Wu YQ, et al. Curcumin Anticancer Res. 2018;38(2):787–94.
inhibits lung cancer cell migration and invasion through Rac1-depend- 53. Zhao GJ, Lu ZQ, Tang LM, Wu ZS, Wang DW, Zheng JY, et al. Curcumin
ent signaling pathway. J Nutr Biochem. 2014;25(2):177–85. inhibits suppressive capacity of naturally occurring CD4 + CD25 + regu-
33. Cheng TS, Chen WC, Lin YY, Tsai CH, Liao CI, Shyu HY, et al. Curcumin- latory T cells in mice in vitro. Int Immunopharmacol. 2012;14(1):99–106.
targeting pericellular serine protease matriptase role in suppression 54. Luo F, Song X, Zhang Y, Chu Y. Low-dose curcumin leads to the inhibi-
of prostate cancer cell invasion, tumor growth, and metastasis. Cancer tion of tumor growth via enhancing CTL-mediated antitumor immu-
Prevent Res (Philadelphia, Pa). 2013;6(5):495–505. nity. Int Immunopharmacol. 2011;11(9):1234–40.
34. Hu C, Li M, Guo T, Wang S, Huang W, Yang K, et al. Anti-metastasis activ- 55. Liao F, Liu L, Luo E, Hu J. Curcumin enhances anti-tumor immune
ity of curcumin against breast cancer via the inhibition of stem cell-like response in tongue squamous cell carcinoma. Arch Oral Biol.
properties and EMT. Phytomedicine. 2019;58:152740. 2018;92:32–7.
35. Shimada K, Ushijima K, Suzuki C, Horiguchi M, Ando H, Akita T, et al. 56. Khan MN, Haggag YA, Lane ME, McCarron PA, Tambuwala MM.
Pulmonary administration of curcumin inhibits B16F10 melanoma Polymeric nano-encapsulation of curcumin enhances its anti-cancer
lung metastasis and invasion in mice. Cancer Chemother Pharmacol. activity in breast (MDA-MB231) and lung (A549) cancer cells through
2018;82(2):265–73. reduction in expression of HIF-1 alpha and nuclear p65 (Rel A). Curr
36. Liu GY, Sun YZ, Zhou N, Du XM, Yang J, Guo SJ. 3,3′-OH curcumin causes Drug Deliv. 2018;15(2):286–95.
apoptosis in HepG2 cells through ROS-mediated pathway. Eur J Med 57. Al-Ani LA, Yehye WA, Kadir FA, Hashim NM, AlSaadi MA, Julkapli NM,
Chem. 2016;112:157–63. et al. Hybrid nanocomposite curcumin-capped gold nanoparticle-
37. Wang L, Chen X, Du Z, Li G, Chen M, Chen X, et al. Curcumin suppresses reduced graphene oxide: anti-oxidant potency and selective cancer
gastric tumor cell growth via ROS-mediated DNA polymerase gamma cytotoxicity. PLoS ONE. 2019;14(5):e0216725.
depletion disrupting cellular bioenergetics. J Exp Clin Cancer Res: CR. 58. Chen X, Chen X, Zhang X, Wang L, Cao P, Rajamanickam V, et al.
2017;36(1):47. Curcuminoid B63 induces ROS-mediated paraptosis-like cell death by
targeting TrxR1 in gastric cells. Redox Biol. 2019;21:101061.
Luo et al. Chin Med (2019) 14:48 Page 36 of 58
59. Hajigholami S, Malekshahi ZV, Bodaghabadi N, Najafi F, Shirzad H, 78. Pang J, Zhang Z, Zheng TZ, Bassig BA, Mao C, Liu X, et al. Green tea
Sadeghizadeh M. Nano packaged tamoxifen and curcumin; effective consumption and risk of cardiovascular and ischemic related diseases: a
formulation against sensitive and resistant MCF-7 cells. Iran J Pharm meta-analysis. Int J Cardiol. 2016;202:967–74.
Res. 2018;17(1):1–10. 79. Shin CM, Lee DH, Seo AY, Lee HJ, Kim SB, Son WC, et al. Green tea
60. Liu L, Xiong XY, Shen M, Ru D, Gao P, Zhang XY, et al. Co-delivery of extracts for the prevention of metachronous colorectal polyps
triptolide and curcumin for ovarian cancer targeting therapy via mPEG- among patients who underwent endoscopic removal of colorectal
DPPE/CaP nanoparticle. J Biomed Nanotechnol. 2018;14(10):1761–72. adenomas: a randomized clinical trial. Clin Nutr (Edinburgh, Scotland).
61. Anirudhan TS, Nair AS, Bino SJ. Nanoparticle assisted solvent selective 2018;37(2):452–8.
transdermal combination therapy of curcumin and 5-flurouracil for 80. Zhan W, Liu Y, Li DP, Liu Y. Advancing insights on the anti-obesity
efficient cancer treatment. Carbohyd Polym. 2017;173:131–42. biochemical mechanism of (−)-epigallocatechin gallate (EGCG) by
62. Choudhury D, Ganguli A, Dastidar DG, Acharya BR, Das A, Chakrabarti G. inhibiting alpha-amylase activity. Rsc Adv. 2016;6(99):96918–27.
Apigenin shows synergistic anticancer activity with curcumin by bind- 81. Lee S, Al Razqan GS, Kwon DH. Antibacterial activity of epigallocate-
ing at different sites of tubulin. Biochimie. 2013;95(6):1297–309. chin-3-gallate (EGCG) and its synergism with beta-lactam antibiotics
63. Gao X, Wang B, Wu Q, Wei X, Zheng F, Men K, et al. Combined delivery sensitizing carbapenem-associated multidrug resistant clinical isolates
and anti-cancer activity of paclitaxel and curcumin using polymeric of Acinetobacter baumannii. Phytomedicine. 2017;24:49–55.
micelles. J Biomed Nanotechnol. 2015;11(4):578–89. 82. Zhu C, Xu Y, Liu ZH, Wan XC, Li DX, Tai LL. The anti-hyperuricemic effect
64. Guo J, Li W, Shi H, Xie X, Li L, Tang H, et al. Synergistic effects of cur- of epigallocatechin-3-gallate (EGCG) on hyperuricemic mice. Biomed
cumin with emodin against the proliferation and invasion of breast Pharmacother. 2018;97:168–73.
cancer cells through upregulation of miR-34a. Mol Cell Biochem. 83. Steinmann J, Buer J, Pietschmann T, Steinmann E. Anti-infective proper-
2013;382(1–2):103–11. ties of epigallocatechin-3-gallate (EGCG), a component of green tea. Br
65. Hu B, Sun D, Sun C, Sun YF, Sun HX, Zhu QF, et al. A polymeric nanopar- J Pharmacol. 2013;168(5):1059–73.
ticle formulation of curcumin in combination with sorafenib synergisti- 84. Irimie AI, Braicu C, Zanoaga O, Pileczki V, Gherman C, Berindan-Neagoe
cally inhibits tumor growth and metastasis in an orthotopic model I, et al. Epigallocatechin-3-gallate suppresses cell proliferation and pro-
of human hepatocellular carcinoma. Biochem Biophys Res Commun. motes apoptosis and autophagy in oral cancer SSC-4 cells. OncoTargets
2015;468(4):525–32. Ther. 2015;8:461–70.
66. Huang YF, Zhu DJ, Chen XW, Chen QK, Luo ZT, Liu CC, et al. Curcumin 85. Zhang JL, Lei Z, Huang ZN, Zhang X, Zhou YY, Luo ZL, et al. Epigallocat-
enhances the effects of irinotecan on colorectal cancer cells through echin-3-gallate(EGCG) suppresses melanoma cell growth and metasta-
the generation of reactive oxygen species and activation of the endo- sis by targeting TRAF6 activity. Oncotarget. 2016;7(48):79543–57.
plasmic reticulum stress pathway. Oncotarget. 2017;8(25):40264–75. 86. Tudoran O, Soritau O, Balacescu O, Balacescu L, Braicu C, Rus M, et al.
67. Ji J, Wang HS, Gao YY, Sang LM, Zhang L. Synergistic anti-tumor effect Early transcriptional pattern of angiogenesis induced by EGCG treat-
of KLF4 and curcumin in human gastric carcinoma cell line. Asian Pac J ment in cervical tumour cells. J Cell Mol Med. 2012;16(3):520–30.
Cancer Prevent. 2014;15(18):7747–52. 87. Manjegowda MC, Deb G, Kumar N, Limaye AM. Expression profiling of
68. Klippstein R, Bansal SS, Al-Jamal KT. Doxorubicin enhances curcumin’s genes modulated by estrogen, EGCG or both in MCF-7 breast cancer
cytotoxicity in human prostate cancer cells in vitro by enhancing its cells. Genomics Data. 2015;5:210–2.
cellular uptake. Int J Pharm. 2016;514(1):169–75. 88. Borutinskaite V, Virksaite A, Gudelyte G, Navakauskiene R. Green
69. Qian H, Yang Y, Wang X. Curcumin enhanced adriamycin-induced tea polyphenol EGCG causes anti-cancerous epigenetic modula-
human liver-derived Hepatoma G2 cell death through activation of tions in acute promyelocytic leukemia cells. Leuk Lymphoma.
mitochondria-mediated apoptosis and autophagy. Eur J Pharm Sci. 2018;59(2):469–78.
2011;43(3):125–31. 89. Yang CG, Du WF, Yang DG. Inhibition of green tea polyphenol
70. Roberts JL, Poklepovic A, Booth L. Curcumin interacts with sildenafil EGCG((−)-epigallocatechin-3-gallate) on the proliferation of gastric
to kill GI tumor cells via endoplasmic reticulum stress and reactive cancer cells by suppressing canonical wnt/beta-catenin signalling
oxygen/nitrogen species. Oncotarget. 2017;8(59):99451–69. pathway. Int J Food Sci Nutr. 2016;67(7):818–27.
71. Siddiqui RA, Harvey KA, Walker C, Altenburg J, Xu Z, Terry C, et al. Char- 90. Dettlaff K, Stawny M, Ogrodowczyk M, Jelinska A, Bednarski W,
acterization of synergistic anti-cancer effects of docosahexaenoic acid Watrobska-Swietlikowska D, et al. Formulation and characterization of
and curcumin on DMBA-induced mammary tumorigenesis in mice. EGCG for the treatment of superficial bladder cancer. Int J Mol Med.
BMC Cancer. 2013;13:418. 2017;40(2):329–36.
72. Lee HM, Patel V, Shyur LF, Lee WL. Copper supplementation amplifies 91. Ni J, Guo X, Wang H, Zhou T, Wang X. Differences in the effects of EGCG
the anti-tumor effect of curcumin in oral cancer cells. Phytomedicine. on chromosomal stability and cell growth between normal and colon
2016;23(12):1535–44. cancer cells. Molecules. 2018;23(4):788.
73. Kondo A, Takeda T, Li B, Tsuiji K, Kitamura M, Wong TF, et al. Epigallo- 92. Flores-Perez A, Marchat LA, Sanchez LL, Romero-Zamora D, Arechaga-
catechin-3-gallate potentiates curcumin’s ability to suppress uterine Ocampo E, Ramirez-Torres N, et al. Differential proteomic analysis
leiomyosarcoma cell growth and induce apoptosis. Int J Clin Oncol. reveals that EGCG inhibits HDGF and activates apoptosis to increase the
2013;18(3):380–8. sensitivity of non-small cells lung cancer to chemotherapy. Proteomics
74. Parsons HA, Baracos VE, Hong DS, Abbruzzese J, Bruera E, Kurzrock Clin Appl. 2016;10(2):172–82.
R. The effects of curcumin (diferuloylmethane) on body composi- 93. Deng YT, Lin JK. EGCG inhibits the invasion of highly invasive
tion of patients with advanced pancreatic cancer. Oncotarget. CL1-5 lung cancer cells through suppressing MMP-2 expression
2016;7(15):20293–304. via JNK signaling and induces G2/M arrest. J Agric Food Chem.
75. Greil R, Greil-Ressler S, Weiss L, Schonlieb C, Magnes T, Radl B, et al. A 2011;59(24):13318–27.
phase 1 dose-escalation study on the safety, tolerability and activity of 94. Luo KW, Wei C, Lung WY, Wei XY, Cheng BH, Cai ZM, et al. EGCG inhib-
liposomal curcumin (Lipocurc()) in patients with locally advanced or ited bladder cancer SW780 cell proliferation and migration both in vitro
metastatic cancer. Cancer Chemother Pharmacol. 2018;82(4):695–706. and in vivo via down-regulation of NF-kappaB and MMP-9. J Nutr
76. Kanai M, Otsuka Y, Otsuka K, Sato M, Nishimura T, Mori Y, et al. A phase Biochem. 2017;41:56–64.
I study investigating the safety and pharmacokinetics of highly bioavail- 95. Van Aller GS, Carson JD, Tang W, Peng H, Zhao L, Copeland RA, et al.
able curcumin (Theracurmin) in cancer patients. Cancer Chemother Epigallocatechin gallate (EGCG), a major component of green tea, is a
Pharmacol. 2013;71(6):1521–30. dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem Biophys Res
77. Mahammedi H, Planchat E, Pouget M, Durando X, Cure H, Guy L, et al. Commun. 2011;406(2):194–9.
The new combination docetaxel, prednisone and curcumin in patients 96. Shin YS, Kang SU, Park JK, Kim YE, Kim YS, Baek SJ, et al. Anti-cancer
with castration-resistant prostate cancer: a pilot phase II study. Oncol- effect of (−)-epigallocatechin-3-gallate (EGCG) in head and neck cancer
ogy. 2016;90(2):69–78. through repression of transactivation and enhanced degradation of
beta-catenin. Phytomedicine. 2016;23(12):1344–55.
Luo et al. Chin Med (2019) 14:48 Page 37 of 58
97. Huang CY, Han Z, Li X, Xie HH, Zhu SS. Mechanism of EGCG promot- 117. Liang J, Cao L, Zhang L, Wan XC. Preparation, characterization, and
ing apoptosis of MCF-7 cell line in human breast cancer. Oncol Lett. in vitro antitumor activity of folate conjugated chitosan coated EGCG
2017;14(3):3623–7. nanoparticles. Food Sci Biotechnol. 2014;23(2):569–75.
98. Satoh M, Takemura Y, Hamada H, Sekido Y, Kubota S. EGCG induces 118. Khan N, Bharali DJ, Adhami VM, Siddiqui IA, Cui HD, Shabana SM, et al.
human mesothelioma cell death by inducing reactive oxygen species Oral administration of naturally occurring chitosan-based nanoformu-
and autophagy. Cancer Cell Int. 2013;13(1):19. lated green tea polyphenol EGCG effectively inhibits prostate cancer
99. Tsai CY, Chen CY, Chiou YH, Shyu HW, Lin KH, Chou MC, et al. Epigal- cell growth in a xenograft model. Carcinogenesis. 2014;35(2):415–23.
locatechin-3-gallate suppresses human herpesvirus 8 replication and 119. Wu SS, Sun K, Wang X, Wang DX, Wan XC, Zhang JS. Protonation of
induces ROS leading to apoptosis and autophagy in primary effusion epigallocatechin-3-gallate (EGCG) results in massive aggregation and
lymphoma cells. Int J Mol Sci. 2017;19(1):16. reduced oral bioavailability of EGCG-dispersed selenium nanoparticles.
100. Liu LC, Tsao TC, Hsu SR, Wang HC, Tsai TC, Kao JY, et al. EGCG inhibits J Agric Food Chem. 2013;61(30):7268–75.
transforming growth factor-beta-mediated epithelial-to-mesen- 120. Onoue S, Ochi M, Yamada S. Development of (-)-epigallocatechin-3-gal-
chymal transition via the inhibition of Smad2 and Erk1/2 signaling late (EGCG)-loaded enteric microparticles with intestinal mucoadhesive
pathways in nonsmall cell lung cancer cells. J Agric Food Chem. property. Int J Pharm. 2011;410(1–2):111–3.
2012;60(39):9863–73. 121. Yang R, Liu YQ, Gao YJ, Wang YJ, Blanchard C, Zhou ZK. Ferritin glyco-
101. Sakamoto Y, Terashita N, Muraguchi T, Fukusato T, Kubota S. Effects of sylated by chitosan as a novel EGCG nano-carrier: structure, stability,
epigallocatechin-3-gallate (EGCG) on A549 lung cancer tumor growth and absorption analysis. Int J Biol Macromol. 2017;105:252–61.
and angiogenesis. Biosci Biotechnol Biochem. 2013;77(9):1799–803. 122. Wang XM, Jiang P, Wang PQ, Yang CS, Wang XR, Feng Q. EGCG enhances
102. Li M, Li JJ, Gu QH, An J, Cao LM, Yang HP, et al. EGCG induces lung cisplatin sensitivity by regulating expression of the copper and cisplatin
cancer A549 cell apoptosis by regulating Ku70 acetylation. Oncol Rep. influx transporter CTR1 in ovary cancer. PLoS ONE. 2015;10(4):e0125402.
2016;35(4):2339–47. 123. Zhou DH, Wang X, Yang M, Shi X, Huang W, Feng Q. Combination
103. Hu Q, Chang X, Yan R, Rong C, Yang C, Cheng S, et al. (−)-Epigallo- of low concentration of (-)-epigallocatechin gallate (EGCG) and
catechin-3-gallate induces cancer cell apoptosis via acetylation of curcumin strongly suppresses the growth of non-small cell lung cancer
amyloid precursor protein. Med Oncol (Northwood, London, England). in vitro and in vivo through causing cell cycle arrest. Int J Mol Sci.
2015;32(1):390. 2013;14(6):12023–36.
104. Onoda C, Kuribayashi K, Nirasawa S, Tsuji N, Tanaka M, Kobayashi 124. Zhang Y, Wang SX, Ma JW, Li HY, Ye JC, Xie SM, et al. EGCG inhibits
D, et al. (−)-Epigallocatechin-3-gallate induces apoptosis in gastric properties of glioma stem-like cells and synergizes with temozolomide
cancer cell lines by down-regulating survivin expression. Int J Oncol. through downregulation of P-glycoprotein inhibition. J Neurooncol.
2011;38(5):1403–8. 2015;121(1):41–52.
105. Wang J, Zhu C, Song D, Xia R, Yu W, Dang Y, et al. Epigallocatechin- 125. Hu F, Wei F, Wang Y, Wu B, Fang Y, Xiong B. EGCG synergizes the thera-
3-gallate enhances ER stress-induced cancer cell apoptosis by directly peutic effect of cisplatin and oxaliplatin through autophagic pathway
targeting PARP16 activity. Cell Death Discov. 2017;3:17034. in human colorectal cancer cells. J Pharmacol Sci. 2015;128(1):27–34.
106. Jwa M, Chang P. PARP16 is a tail-anchored endoplasmic reticulum pro- 126. Shin DM, Amin ARM, Wang D, Rahman MA, Nannapaneni S, Khuri FR,
tein required for the PERK- and IRE1alpha-mediated unfolded protein et al. 1187 molecular mechanism of synergistic anti-tumor activity by
response. Nat Cell Biol. 2012;14(11):1223–30. the combination of natural compounds Green tea (-)epigallocathetin-
107. Martinotti S, Ranzato E, Burlando B. (−)-Epigallocatechin-3-gallate 3-gallate (EGCG) and resveratrol for potential chemoprevention in head
induces GRP78 accumulation in the ER and shifts mesothe- and neck cancer (HNC). Eur J Cancer. 2012;48:S286.
lioma constitutive UPR into proapoptotic ER stress. J Cell Physiol. 127. Wang W, Chen D, Zhu K. SOX2OT variant 7 contributes to the synergis-
2018;233(10):7082–90. tic interaction between EGCG and Doxorubicin to kill osteosarcoma
108. Ogawa K, Hara T, Shimizu M, Nagano J, Ohno T, Hoshi M, et al. (−)-Epi- via autophagy and stemness inhibition. J Exp Clin Cancer Res: CR.
gallocatechin gallate inhibits the expression of indoleamine 2,3-dioxy- 2018;37(1):37.
genase in human colorectal cancer cells. Oncol Lett. 2012;4(3):546–50. 128. Kumazoe M, Tsukamoto S, Lesnick C, Kay NE, Yamada K, Shanafelt
109. Rawangkan A, Wongsirisin P, Namiki K, Iida K, Kobayashi Y, Shimizu Y, TD, et al. Vardenafil, a clinically available phosphodiesterase inhibi-
et al. Green tea catechin is an alternative immune checkpoint inhibitor tor, potentiates the killing effect of EGCG on CLL cells. Br J Haematol.
that inhibits PD-L1 expression and lung tumor growth. Molecules. 2015;168(4):610–3.
2018;23(8):12. 129. Haque A, Rahman MA, Chen ZG, Saba NF, Khuri FR, Shin DM, et al.
110. Zeng L, Yan J, Luo L, Ma M, Zhu H. Preparation and characterization Combination of erlotinib and EGCG induces apoptosis of head and
of (-)-Epigallocatechin-3-gallate (EGCG)-loaded nanoparticles and neck cancers through posttranscriptional regulation of Bim and Bcl-2.
their inhibitory effects on Human breast cancer MCF-7 cells. Sci Rep. Apoptosis. 2015;20(7):986–95.
2017;7:45521. 130. Zhao HX, Zhu WQ, Jia L, Sun XR, Chen GX, Zhao XG, et al. Phase I study
111. Wang JZ, Man GCW, Chan TH, Kwong J, Wang CC. A prodrug of green of topical epigallocatechin-3-gallate (EGCG) in patients with breast
tea polyphenol (-)-epigallocatechin-3-gallate (Pro-EGCG) serves as cancer receiving adjuvant radiotherapy. Br J Radiol. 2016;89(1058):10.
a novel angiogenesis inhibitor in endometrial cancer. Cancer Lett. 131. Zhao HX, Xie P, Li XL, Zhu WQ, Sun XD, Sun XR, et al. A prospective
2018;412:10–20. phase II trial of EGCG in treatment of acute radiation-induced esophagi-
112. Zhong Y, Chiou YS, Pan MH, Ho CT, Shahidi F. Protective effects of tis for stage III lung cancer. Radiother Oncol. 2015;114(3):351–6.
epigallocatechin gallate (EGCG) derivatives on azoxymethane-induced 132. Wang K, Feng X, Chai L, Cao S, Qiu F. The metabolism of berberine
colonic carcinogenesis in mice. J Funct Foods. 2012;4(1):323–30. and its contribution to the pharmacological effects. Drug Metab Rev.
113. Zhong Y, Shahidi F. Lipophilized epigallocatechin gal late (EGCG) deriva- 2017;49(2):139–57.
tives as novel antioxidants. J Agric Food Chem. 2011;59(12):6526–33. 133. Zhu XF, Yue HD, Guo XF, Yang JY, Liu JS, Liu JT, et al. The preconditioning
114. Wu M, Jin JC, Jin P, Xu YQ, Yin JF, Qin DK, et al. Epigallocatechin of berberine suppresses hydrogen peroxide-induced premature senes-
gallate-beta-lactoglobulin nanoparticles improve the antitumor cence via regulation of sirtuin 1. Oxid Med Cell Longevity. 2017;2017:9.
activity of EGCG for inducing cancer cell apoptosis. J Funct Foods. 134. Chuang TY, Wu HL, Min J, Diamond M, Azziz R, Chen YH. Berberine regu-
2017;39:257–63. lates the protein expression of multiple tumorigenesis-related genes in
115. Radhakrishnan R, Kulhari H, Pooja D, Gudem S, Bhargava S, Shukla R, hepatocellular carcinoma cell lines. Cancer Cell Int. 2017;17:8.
et al. Encapsulation of biophenolic phytochemical EGCG within lipid 135. Liu J, Zhang X, Liu A, Liu S, Zhang L, Wu B, et al. Berberine induces
nanoparticles enhances its stability and cytotoxicity against cancer. apoptosis in p53-null leukemia cells by down-regulating XIAP at the
Chem Phys Lipids. 2016;198:51–60. post-transcriptional level. Cell Physiol Biochem. 2013;32(5):1213–24.
116. Liao BW, Ying H, Yu CH, Fan ZY, Zhang WH, Shi J, et al. (-)-Epigallocat- 136. Park KS, Kim JB, Lee SJ, Bae J. Berberine-induced growth inhibition
echin gallate (EGCG)-nanoethosomes as a transdermal delivery system of epithelial ovarian carcinoma cell lines. J Obstetr Gynaecol Res.
for docetaxel to treat implanted human melanoma cell tumors in mice. 2012;38(3):535–40.
Int J Pharm. 2016;512(1):22–31.
Luo et al. Chin Med (2019) 14:48 Page 38 of 58
137. Wen CJ, Wu LX, Fu LJ, Yu J, Zhang YW, Zhang X, et al. Genomic screen- 157. Chidambara Murthy KN, Jayaprakasha GK, Patil BS. The natu-
ing for targets regulated by berberine in breast cancer cells. Asian Pac J ral alkaloid berberine targets multiple pathways to induce cell
Cancer Prev. 2013;14(10):6089–94. death in cultured human colon cancer cells. Eur J Pharmacol.
138. Luo X, Gu J, Zhu R, Feng M, Zhu X, Li Y, et al. Integrative analysis of 2012;688(1–3):14–21.
differential miRNA and functional study of miR-21 by seed-targeting 158. Xie J, Xu Y, Huang X, Chen Y, Fu J, Xi M, et al. Berberine-induced apop-
inhibition in multiple myeloma cells in response to berberine. BMC Syst tosis in human breast cancer cells is mediated by reactive oxygen
Biol. 2014;8:82. species generation and mitochondrial-related apoptotic pathway.
139. Wang J, Kang M, Wen Q, Qin YT, Wei ZX, Xiao JJ, et al. Berberine sensi- Tumour Biol. 2015;36(2):1279–88.
tizes nasopharyngeal carcinoma cells to radiation through inhibition of 159. Park GB, Park SH, Kim D, Kim YS, Yoon SH, Hur DY. Berberine induces
Sp1 and EMT. Oncol Rep. 2017;37(4):2425–32. mitochondrial apoptosis of EBV-transformed B cells through
140. Jiang SX, Qi B, Yao WJ, Gu CW, Wei XF, Zhao Y, et al. Berberine displays p53-mediated regulation of XAF1 and GADD45alpha. Int J Oncol.
antitumor activity in esophageal cancer cells in vitro. World J Gastroen- 2016;49(1):411–21.
terol. 2017;23(14):2511–8. 160. Park SH, Sung JH, Kim EJ, Chung N. Berberine induces apoptosis via
141. Naveen CR, Gaikwad S, Agrawal-Rajput R. Berberine induces neuronal ROS generation in PANC-1 and MIA-PaCa2 pancreatic cell lines. Braz J
differentiation through inhibition of cancer stemness and epithe- Med Biol Res. 2015;48(2):111–9.
lial–mesenchymal transition in neuroblastoma cells. Phytomedicine. 161. Wang L, Liu L, Shi Y, Cao H, Chaturvedi R, Calcutt MW, et al. Ber-
2016;23(7):736–44. berine induces caspase-independent cell death in colon tumor
142. Park KS, Kim JB, Bae J, Park SY, Jee HG, Lee KE, et al. Berberine inhibited cells through activation of apoptosis-inducing factor. PLoS ONE.
the growth of thyroid cancer cell lines 8505C and TPC1. Yonsei Med J. 2012;7(5):e36418.
2012;53(2):346–51. 162. Takeda K, Akira S. Toll-like receptors. Curr Protoc Immunol. 2015;109:142.
143. Wang XB, Wang N, Li HL, Liu M, Cao FJ, Yu XJ, et al. Up-regulation of 163. Cheng WE, Ying Chang M, Wei JY, Chen YJ, Maa MC, Leu TH. Berberine
PAI-1 and down-regulation of uPA are involved in suppression of reduces Toll-like receptor-mediated macrophage migration by suppres-
invasiveness and motility of hepatocellular carcinoma cells by a natural sion of Src enhancement. Eur J Pharmacol. 2015;757:1–10.
compound berberine. Int J Mol Sci. 2016;17(4):15. 164. Liu M, Wang X, Wang L, Ma X, Gong Z, Zhang S, et al. Targeting the
144. Wang Y, Liu Q, Liu Z, Li B, Sun Z, Zhou H, et al. Berberine, a genotoxic IDO1 pathway in cancer: from bench to bedside. J Hematol Oncol.
alkaloid, induces ATM-Chk1 mediated G2 arrest in prostate cancer cells. 2018;11(1):100.
Mutat Res. 2012;734(1–2):20–9. 165. Wang YX, Pang WQ, Zeng QX, Deng ZS, Fan TY, Jiang JD, et al. Synthesis
145. Kuo HP, Chuang TC, Yeh MH, Hsu SC, Way TD, Chen PY, et al. Growth and biological evaluation of new berberine derivatives as cancer
suppression of HER2-overexpressing breast cancer cells by berberine immunotherapy agents through targeting IDO1. Eur J Med Chem.
via modulation of the HER2/PI3K/Akt signaling pathway. J Agric Food 2018;143:1858–68.
Chem. 2011;59(15):8216–24. 166. Kim S, You D, Jeong Y, Yu J, Kim SW, Nam SJ, et al. Berberine down-regu-
146. Xu LN, Lu BN, Hu MM, Xu YW, Han X, Qi Y, et al. Mechanisms involved in lates IL-8 expression through inhibition of the EGFR/MEK/ERK pathway
the cytotoxic effects of berberine on human colon cancer HCT-8 cells. in triple-negative breast cancer cells. Phytomedicine. 2018;50:43–9.
Biocell. 2012;36(3):113–20. 167. Chen W, Miao YQ, Fan DJ, Yang SS, Lin X, Meng LK, et al. Bioavailability
147. Shukla S, Rizvi F, Raisuddin S, Kakkar P. FoxO proteins’ nuclear retention study of berberine and the enhancing effects of TPGS on intestinal
and BH3-only protein Bim induction evoke mitochondrial dysfunction- absorption in rats. AAPS PharmSciTech. 2011;12(2):705–11.
mediated apoptosis in berberine-treated HepG2 cells. Free Radical Biol 168. Alamzeb M, Khan MR, Mamoon Ur R, Ali S, Khan AA. A new isoquino-
Med. 2014;76:185–99. line alkaloid with anti-microbial properties from Berberis jaeschkeana
148. La X, Zhang L, Li Z, Yang P, Wang Y. Berberine-induced autophagic Schneid. var. jaeschkeana. Nat Prod Res. 2015;29(8):692–7.
cell death by elevating GRP78 levels in cancer cells. Oncotarget. 169. Zhu JX, Tang D, Feng L, Zheng ZG, Wang RS, Wu AG, et al. Develop-
2017;8(13):20909–24. ment of self-microemulsifying drug delivery system for oral bioavail-
149. Wang J, Qi Q, Feng Z, Zhang X, Huang B, Chen A, et al. Berberine ability enhancement of berberine hydrochloride. Drug Dev Ind Pharm.
induces autophagy in glioblastoma by targeting the AMPK/mTOR/ 2013;39(3):499–506.
ULK1-pathway. Oncotarget. 2016;7(41):66944–58. 170. Xiong YX, Su HF, Lv P, Ma Y, Wang SK, Miao H, et al. A newly identified
150. Yao Z, Wan Y, Li B, Zhai C, Yao F, Kang Y, et al. Berberine induces berberine derivative induces cancer cell senescence by stabilizing
mitochondrialmediated apoptosis and protective autophagy in endogenous G-quadruplexes and sparking a DNA damage response at
human malignant pleural mesothelioma NCIH2452 cells. Oncol Rep. the telomere region. Oncotarget. 2015;6(34):35625–35.
2018;40(6):3603–10. 171. Hashemi-Niasari F, Rabbani-Chadegani A, Razmi M, Fallah S. Synergy
151. Kou Y, Li L, Li H, Tan YH, Li B, Wang K, et al. Berberine suppressed epi- of theophylline reduces necrotic effect of berberine, induces cell cycle
thelial mesenchymal transition through cross-talk regulation of PI3K/ arrest and PARP, HMGB1, Bcl-2 family mediated apoptosis in MDA-
AKT and RAR alpha/RAR beta in melanoma cells. Biochem Biophys Res MB-231 breast cancer cells. Biomed Pharmacother. 2018;106:858–67.
Commun. 2016;479(2):290–6. 172. Palmieri A, Iapichino A, Cura F, Scapoli L, Carinci F, Mandrone M, et al.
152. Jin F, Xie T, Huang X, Zhao X. Berberine inhibits angiogenesis in glio- Pre-treatment with berberine enhances effect of 5-fluorouracil and cis-
blastoma xenografts by targeting the VEGFR2/ERK pathway. Pharm Biol. platin in HEP2 laryngeal cancer cell line. J Biol Regul Homeost Agents.
2018;56(1):665–71. 2018;32(2 Suppl. 1):167–77.
153. Chu SC, Yu CC, Hsu LS, Chen KS, Su MY, Chen PN. Berberine reverses 173. Liu L, Fan J, Ai G, Liu J, Luo N, Li C, et al. Berberine in combination with
epithelial-to-mesenchymal transition and inhibits metastasis and cisplatin induces necroptosis and apoptosis in ovarian cancer cells. Biol
tumor-induced angiogenesis in human cervical cancer cells. Mol Phar- Res. 2019;52(1):37.
macol. 2014;86(6):609–23. 174. Hou D, Xu G, Zhang C, Li B, Qin J, Hao X, et al. Berberine induces oxida-
154. Li X-L, Hu Y-J, Wang H, Yu B-Q, Yue H-L. Molecular spectroscopy evi- tive DNA damage and impairs homologous recombination repair in
dence of berberine binding to DNA: comparative binding and thermo- ovarian cancer cells to confer increased sensitivity to PARP inhibition.
dynamic profile of intercalation. Biomacromolecules. 2012;13(3):873–80. Cell Death Dis. 2017;8(10):e3070.
155. Li J, Liu F, Jiang S, Liu J, Chen X, Zhang S, et al. Berberine hydrochloride 175. Efferth T. From ancient herb to modern drug: artemisia annua and
inhibits cell proliferation and promotes apoptosis of non-small cell artemisinin for cancer therapy. Semin Cancer Biol. 2017;46:65–83.
lung cancer via the suppression of the MMP2 and Bcl-2/Bax signaling 176. Efferth T, Zacchino S, Georgiev MI, Liu L, Wagner H, Panossian AJ, et al.
pathways. Oncol Lett. 2018;15(5):7409–14. Nobel prize for artemisinin brings phytotherapy into the spotlight.
156. Lin YS, Chiu YC, Tsai YH, Tsai YF, Wang JY, Tseng LM, et al. Different Phytomedicine. 2015;22(13):1–4.
mechanisms involved in the berberine-induced antiproliferation 177. Tilaoui M, Mouse HA, Jaafari A, Zyad A. Differential effect of artemisinin
effects in triple-negative breast cancer cell lines. J Cell Biochem. against cancer cell lines. Nat Prod Bioprospect. 2014;4(3):189–96.
2019;120(8):13531–44.
Luo et al. Chin Med (2019) 14:48 Page 39 of 58
178. Zhang CZ, Zhang H, Yun J, Chen GG, San Lai PB. Dihydroartemisinin cell death in cholangiocarcinoma through a DAPK1-BECLIN1 pathway.
exhibits antitumor activity toward hepatocellular carcinoma in vitro Mol Carcinog. 2018;57(12):1735–50.
and in vivo. Biochem Pharmacol. 2012;83(9):1278–89. 199. Shi X, Wang L, Li X, Bai J, Li J, Li S, et al. Dihydroartemisinin induces
179. Jiang F, Zhou JY, Zhang D, Liu MH, Chen YG. Artesunate induces autophagy-dependent death in human tongue squamous cell
apoptosis and autophagy in HCT116 colon cancer cells, and autophagy carcinoma cells through DNA double-strand break-mediated oxidative
inhibition enhances the artesunateinduced apoptosis. Int J Mol Med. stress. Oncotarget. 2017;8(28):45981–93.
2018;42(3):1295–304. 200. Li B, Bu S, Sun J, Guo Y, Lai D. Artemisinin derivatives inhibit epithelial
180. Zhao X, Guo X, Yue W, Wang J, Yang J, Chen J. Artemether suppresses ovarian cancer cells via autophagy-mediated cell cycle arrest. Acta
cell proliferation and induces apoptosis in diffuse large B cell lym- Biochim Biophys Sin. 2018;50(12):1227–35.
phoma cells. Exp Ther Med. 2017;14(5):4083–90. 201. Odaka Y, Xu B, Luo Y, Shen T, Shang C, Wu Y, et al. Dihydroartemisinin
181. Azimi Mohamadabadi M, Hassan ZM, Zavaran Hosseini A, Gholamzad inhibits the mammalian target of rapamycin-mediated signaling path-
M, Noori S, Mahdavi M, et al. Arteether exerts antitumor activity and ways in tumor cells. Carcinogenesis. 2014;35(1):192–200.
reduces CD4 + CD25 + FOXP3 + T-reg cells in vivo. Iran J Immunol. 202. Hu W, Chen SS, Zhang JL, Lou XE, Zhou HJ. Dihydroartemisinin
2013;10(3):139–49. induces autophagy by suppressing NF-kappaB activation. Cancer Lett.
182. Li Y, Sui H, Jiang C, Li S, Han Y, Huang P, et al. Dihydroartemisinin 2014;343(2):239–48.
increases the sensitivity of photodynamic therapy via NF-kappaB/HIF- 203. Jiang J, Geng G, Yu X, Liu H, Gao J, An H, et al. Repurposing the anti-
1alpha/VEGF pathway in esophageal cancer cell in vitro and in vivo. Cell malarial drug dihydroartemisinin suppresses metastasis of non-small-
Physiol Biochem. 2018;48(5):2035–45. cell lung cancer via inhibiting NF-kappaB/GLUT1 axis. Oncotarget.
183. Lin R, Zhang Z, Chen L, Zhou Y, Zou P, Feng C, et al. Dihydroartemisinin 2016;7(52):87271–83.
(DHA) induces ferroptosis and causes cell cycle arrest in head and neck 204. Li Z, Ding X, Wu H, Liu C. Artemisinin inhibits angiogenesis by regulat-
carcinoma cells. Cancer Lett. 2016;381(1):165–75. ing p38 MAPK/CREB/TSP-1 signaling pathway in osteosarcoma. J Cell
184. Tong Y, Liu Y, Zheng H, Zheng L, Liu W, Wu J, et al. Artemisinin and Biochem. 2019. https://doi.org/10.1002/jcb.28424.
its derivatives can significantly inhibit lung tumorigenesis and 205. Hu CJ, Zhou L, Cai Y. Dihydroartemisinin induces apoptosis of cervical
tumor metastasis through Wnt/beta-catenin signaling. Oncotarget. cancer cells via upregulation of RKIP and downregulation of bcl-2.
2016;7(21):31413–28. Cancer Biol Ther. 2014;15(3):279–88.
185. Im E, Yeo C, Lee HJ, Lee EO. Dihydroartemisinin induced caspase- 206. Qin G, Zhao C, Zhang L, Liu H, Quan Y, Chai L, et al. Dihydroartemisinin
dependent apoptosis through inhibiting the specificity protein induces apoptosis preferentially via a Bim-mediated intrinsic pathway
1 pathway in hepatocellular carcinoma SK-Hep-1 cells. Life Sci. in hepatocarcinoma cells. Apoptosis. 2015;20(8):1072–86.
2018;192:286–92. 207. Ooko E, Saeed ME, Kadioglu O, Sarvi S, Colak M, Elmasaoudi K, et al.
186. Noori S, Hassan ZM, Farsam V. Artemisinin as a Chinese medicine, selec- Artemisinin derivatives induce iron-dependent cell death (ferroptosis)
tively induces apoptosis in pancreatic tumor cell line. Chin J Integr Med. in tumor cells. Phytomedicine. 2015;22(11):1045–54.
2014;20(8):618–23. 208. Zhou ZH, Chen FX, Xu WR, Qian H, Sun LQ, Lu XT, et al. Enhancement
187. Lu M, Sun L, Zhou J, Yang J. Dihydroartemisinin induces apoptosis in effect of dihydroartemisinin on human gammadelta T cell prolif-
colorectal cancer cells through the mitochondria-dependent pathway. eration and killing pancreatic cancer cells. Int Immunopharmacol.
Tumour Biol. 2014;35(6):5307–14. 2013;17(3):850–7.
188. Cao Y, Feng YH, Gao LW, Li XY, Jin QX, Wang YY, et al. Artemisinin 209. Houh YK, Kim KE, Park S, Hur DY, Kim S, Kim D, et al. The effects of arte-
enhances the anti-tumor immune response in 4T1 breast cancer cells misinin on the cytolytic activity of natural killer (NK) cells. Int J Mol Sci.
in vitro and in vivo. Int Immunopharmacol. 2019;70:110–6. 2017;18(7):1600.
189. Li X, Ba Q, Liu Y, Yue Q, Chen P, Li J, et al. Dihydroartemisinin selectively 210. Manjili HK, Malvandi H, Mousavi MS, Attari E, Danafar H. In vitro
inhibits PDGFRalpha-positive ovarian cancer growth and metastasis and in vivo delivery of artemisinin loaded PCL-PEG-PCL micelles
through inducing degradation of PDGFRalpha protein. Cell Discov. and its pharmacokinetic study. Artif Cells Nanomed Biotechnol.
2017;3:17042. 2018;46(5):926–36.
190. Wang L, Li J, Shi X, Li S, Tang PM, Li Z, et al. Antimalarial Dihydroar- 211. Zhang CJ, Wang J, Zhang J, Lee YM, Feng G, Lim TK, et al. Mechanism-
temisinin triggers autophagy within HeLa cells of human cervical guided design and synthesis of a mitochondria-targeting artemisinin
cancer through Bcl-2 phosphorylation at Ser70. Phytomedicine. analogue with enhanced anticancer activity. Angew Chem Int Ed Engl.
2019;52:147–56. 2016;55(44):13770–4.
191. Paccez JD, Duncan K, Sekar D, Correa RG, Wang Y, Gu X, et al. Dihydroar- 212. Magoulas GE, Tsigkou T, Skondra L, Lamprou M, Tsoukala P, Kokkin-
temisinin inhibits prostate cancer via JARID2/miR-7/miR-34a-depend- ogouli V, et al. Synthesis of nomicronvel artemisinin dimers with poly-
ent downregulation of Axl. Oncogenesis. 2019;8(3):14. amine linkers and evaluation of their potential as anticancer agents.
192. Li J, Feng W, Lu H, Wei Y, Ma S, Wei L, et al. Artemisinin inhibits breast Bioorg Med Chem. 2017;25(14):3756–67.
cancer-induced osteolysis by inhibiting osteoclast formation and breast 213. Li P, Yang S, Dou M, Chen Y, Zhang J, Zhao X. Synergic effects of
cancer cell proliferation. J Cell Physiol. 2019;234(8):12663–75. artemisinin and resveratrol in cancer cells. J Cancer Res Clin Oncol.
193. Tin AS, Sundar SN, Tran KQ, Park AH, Poindexter KM, Firestone GL. 2014;140(12):2065–75.
Antiproliferative effects of artemisinin on human breast cancer cells 214. Zhao C, Gao W, Chen T. Synergistic induction of apoptosis in A549 cells
requires the downregulated expression of the E2F1 transcription by dihydroartemisinin and gemcitabine. Apoptosis. 2014;19(4):668–81.
factor and loss of E2F1-target cell cycle genes. Anticancer Drugs. 215. Feng X, Li L, Jiang H, Jiang K, Jin Y, Zheng J. Dihydroartemisinin potenti-
2012;23(4):370–9. ates the anticancer effect of cisplatin via mTOR inhibition in cisplatin-
194. Dong F, Tian H, Yan S, Li L, Dong X, Wang F, et al. Dihydroartemisinin resistant ovarian cancer cells: involvement of apoptosis and autophagy.
inhibits endothelial cell proliferation through the suppression of the Biochem Biophys Res Commun. 2014;444(3):376–81.
ERK signaling pathway. Int J Mol Med. 2015;35(5):1381–7. 216. Li YJ, Zhou JH, Du XX, de Jia X, Wu CL, Huang P, et al. Dihydroartemisinin
195. Dong F, Zhou X, Li C, Yan S, Deng X, Cao Z, et al. Dihydroartemisinin tar- accentuates the anti-tumor effects of photodynamic therapy via inacti-
gets VEGFR2 via the NF-kappaB pathway in endothelial cells to inhibit vation of NF-kappaB in Eca109 and Ec9706 esophageal cancer cells. Cell
angiogenesis. Cancer Biol Ther. 2014;15(11):1479–88. Physiol Biochem. 2014;33(5):1527–36.
196. Kumari K, Keshari S, Sengupta D, Sabat SC, Mishra SK. Transcriptome 217. Ericsson T, Blank A, von Hagens C, Ashton M, Abelo A. Population phar-
analysis of genes associated with breast cancer cell motility in response macokinetics of artesunate and dihydroartemisinin during long-term
to artemisinin treatment. BMC Cancer. 2017;17(1):858. oral administration of artesunate to patients with metastatic breast
197. Wang Z, Hu W, Zhang JL, Wu XH, Zhou HJ. Dihydroartemisinin induces cancer. Eur J Clin Pharmacol. 2014;70(12):1453–63.
autophagy and inhibits the growth of iron-loaded human myeloid 218. Jansen FH, Adoubi I, Cnodder T, Jansen N, Tschulakow A, et al. First
leukemia K562 cells via ROS toxicity. FEBS open bio. 2012;2:103–12. study of oral Artenimol-R in advanced cervical cancer: clinical benefit,
198. Thongchot S, Vidoni C, Ferraresi A, Loilome W, Yongvanit P, Namwat N, tolerability and tumor markers. Anticancer Res. 2011;31(12):4417–22.
et al. Dihydroartemisinin induces apoptosis and autophagy-dependent
Luo et al. Chin Med (2019) 14:48 Page 40 of 58
219. He NW, Zhao Y, Guo L, Shang J, Yang XB. Antioxidant, antiproliferative, 239. Shan X, Fu YS, Aziz F, Wang XQ, Yan Q, Liu JW. Ginsenoside Rg3 inhibits
and pro-apoptotic activities of a saponin extract derived from the roots melanoma cell proliferation through down-regulation of histone
of Panax notoginseng (Burk.) F.H. Chen. J Med Food. 2012;15(4):350–9. deacetylase 3 (HDAC3) and increase of p53 acetylation. PLoS ONE.
220. Lu M, Fei Z, Zhang G. Synergistic anticancer activity of 20(S)-Ginseno- 2014;9(12):e115401.
side Rg3 and Sorafenib in hepatocellular carcinoma by modulating 240. Li Y, Yang T, Li J, Hao HL, Wang SY, Yang J, et al. Inhibition of multiple
PTEN/Akt signaling pathway. Biomed Pharmacother. 2018;97:1282–8. myeloma cell proliferation by ginsenoside Rg3 via reduction in the
221. Wang X, Sun YY, Zhao C, Qu FZ, Zhao YQ. 12-Chloracetyl-PPD, a novel secretion of IGF-1. Mol Med Rep. 2016;14(3):2222–30.
dammarane derivative, shows anti-cancer activity via delay the progres- 241. Tang YC, Zhang Y, Zhou J, Zhi Q, Wu MY, Gong FR, et al. Ginsenoside Rg3
sion of cell cycle G2/M phase and reactive oxygen species-mediate cell targets cancer stem cells and tumor angiogenesis to inhibit colorectal
apoptosis. Eur J Pharmacol. 2017;798:49–56. cancer progression in vivo. Int J Oncol. 2018;52(1):127–38.
222. Lee IS, Uh I, Kim KS, Kim KH, Park J, Kim Y, et al. Anti-inflammatory effects 242. Ge G, Yan Y, Cai H. Ginsenoside Rh2 inhibited proliferation by inducing
of ginsenoside Rg3 via NF-kappaB pathway in A549 cells and human ROS mediated ER stress dependent apoptosis in lung cancer cells. Biol
asthmatic lung tissue. J Immunol Res. 2016;2016:7521601. Pharm Bull. 2017;40(12):2117–24.
223. Lu Z, Xu H, Yu X, Wang Y, Huang L, Jin X, et al. 20(S)-Protopanaxadiol 243. Li Q, Li B, Dong C, Wang Y, Li Q. 20(S)-Ginsenoside Rh2 suppresses
induces apoptosis in human hepatoblastoma HepG2 cells by down- proliferation and migration of hepatocellular carcinoma cells by
regulating the protein kinase B signaling pathway. Exp Ther Med. targeting EZH2 to regulate CDKN2A-2B gene cluster transcription. Eur J
2018;15(2):1277–84. Pharmacol. 2017;815:173–80.
224. Liu Y, Fan D. Ginsenoside Rg5 induces apoptosis and autophagy via the 244. Zhang Z, Du GJ, Wang CZ, Wen XD, Calway T, Li Z, et al. Compound K, a
inhibition of the PI3K/Akt pathway against breast cancer in a mouse ginsenoside metabolite, inhibits colon cancer growth via multiple path-
model. Food Funct. 2018;9:5513–27. ways including p53-p21 interactions. Int J Mol Sci. 2013;14(2):2980–95.
225. Wu Q, Deng J, Fan D, Duan Z, Zhu C, Fu R, et al. Ginsenoside Rh4 245. Zheng ZZ, Ming YL, Chen LH, Zheng GH, Liu SS, Chen QX. Compound
induces apoptosis and autophagic cell death through activation of the K-induced apoptosis of human hepatocellular carcinoma MHCC97-H
ROS/JNK/p53 pathway in colorectal cancer cells. Biochem Pharmacol. cells in vitro. Oncol Rep. 2014;32(1):325–31.
2018;148:64–74. 246. Zheng X, Chen W, Hou H, Li J, Li H, Sun X, et al. Ginsenoside 20(S)-Rg3
226. Leem DG, Shin JS, Kim KT, Choi SY, Lee MH, Lee KT. Dammarane- induced autophagy to inhibit migration and invasion of ovarian cancer.
type triterpene ginsenoside-Rg18 inhibits human non-small cell Biomed Pharmacother. 2017;85:620–6.
lung cancer A549 cell proliferation via G1 phase arrest. Oncol Lett. 247. Wang XJ, Zhou RJ, Zhang N, Jing Z. 20(S)-ginsenoside Rg3 sensitizes
2018;15(4):6043–9. human non-small cell lung cancer cells to icotinib through inhibition of
227. Zeng D, Wang J, Kong P, Chang C, Li J, Li J. Ginsenoside Rg3 inhibits autophagy. Eur J Pharmacol. 2019;850:141–9.
HIF-1α and VEGF expression in patient with acute leukemia via inhibit- 248. Kim DG, Jung KH, Lee DG, Yoon JH, Choi KS, Kwon SW, et al. 20(S)-Ginse-
ing the activation of PI3K/Akt and ERK1/2 pathways. Int J Clin Exp noside Rg3 is a novel inhibitor of autophagy and sensitizes hepatocel-
Pathol. 2014;7(5):2172–8. lular carcinoma to doxorubicin. Oncotarget. 2014;5(12):4438–51.
228. Wang L, Gao S, Jiang W, Luo C, Xu M, Bohlin L, et al. Antioxidative 249. Yang Z, Zhao T, Liu H, Zhang L. Ginsenoside Rh2 inhibits hepatocellular
dietary compounds modulate gene expression associated with apop- carcinoma through beta-catenin and autophagy. Sci Rep. 2016;6:19383.
tosis, DNA repair, inhibition of cell proliferation and migration. Int J Mol 250. Li C, Dong Y, Wang L, Xu G, Yang Q, Tang X, et al. Ginsenoside metabo-
Sci. 2014;15(9):16226–45. lite compound K induces apoptosis and autophagy in non-small cell
229. Kim YJ, Choi WI, Jeon BN, Choi KC, Kim K, Kim TJ, et al. Stereospecific lung cancer cells via AMPK-mTOR and JNK pathways. Biochem Cell Biol.
effects of ginsenoside 20-Rg3 inhibits TGF-beta1-induced epithelial- 2018;97:406.
mesenchymal transition and suppresses lung cancer migration, inva- 251. Kim AD, Kang KA, Kim HS, Kim DH, Choi YH, Lee SJ, et al. A ginseng
sion and anoikis resistance. Toxicology. 2014;322:23–33. metabolite, compound K, induces autophagy and apoptosis via
230. Yang J, Yuan D, Xing T, Su H, Zhang S, Wen J, et al. Ginsenoside Rh2 generation of reactive oxygen species and activation of JNK in human
inhibiting HCT116 colon cancer cell proliferation through blocking colon cancer cells. Cell Death Dis. 2013;4:e750.
PDZ-binding kinase/T-LAK cell-originated protein kinase. J Ginseng Res. 252. Jiang J, Yuan Z, Sun Y, Bu Y, Li W, Fei Z. Ginsenoside Rg3 enhances the
2016;40(4):400–8. anti-proliferative activity of erlotinib in pancreatic cancer cell lines by
231. Wang J-H, Nao J-F, Zhang M, He P. 20(s)-ginsenoside Rg3 promotes downregulation of EGFR/PI3K/Akt signaling pathway. Biomed Pharma-
apoptosis in human ovarian cancer HO-8910 cells through PI3K/Akt cother. 2017;96:619–25.
and XIAP pathways. Tumor Biol. 2014;35(12):11985–94. 253. Pan XY, Guo H, Han J, Hao F, An Y, Xu Y, et al. Ginsenoside Rg3 attenu-
232. Zhang Y, Liu QZ, Xing SP, Zhang JL. Inhibiting effect of Endostar ates cell migration via inhibition of aquaporin 1 expression in PC-3M
combined with ginsenoside Rg3 on breast cancer tumor growth in prostate cancer cells. Eur J Pharmacol. 2012;683(1–3):27–34.
tumor-bearing mice. Asian Pac J Trop Med. 2016;9(2):180–3. 254. Chen XP, Qian LL, Jiang H, Chen JH. Ginsenoside Rg3 inhibits CXCR255
233. Yuan Z, Jiang H, Zhu X, Liu X, Li J. Ginsenoside Rg3 promotes cytotoxic- expression and related migrations in a breast cancer cell line. Int J Clin
ity of Paclitaxel through inhibiting NF-κB signaling and regulating Bax/ Oncol. 2011;16(5):519–23.
Bcl-2 expression on triple-negative breast cancer. Biomed Pharmaco- 255. Liu T, Zhao L, Hou H, Ding L, Chen W, Li X. Ginsenoside 20(S)-Rg3
ther. 2017;89:227–32. suppresses ovarian cancer migration via hypoxia-inducible fac-
234. Jiang JW, Chen XM, Chen XH, Zheng SS. Ginsenoside Rg3 inhibit tor 1 alpha and nuclear factor-kappa B signals. Tumour Biol.
hepatocellular carcinoma growth via intrinsic apoptotic pathway. World 2017;39(5):1010428317692225.
J Gastroenterol. 2011;17(31):3605–13. 256. Wang YS, Lin Y, Li H, Li Y, Song Z, Jin YH. The identification of molecular
235. Zhang C, Liu L, Yu Y, Chen B, Tang C, Li X. Antitumor effects of ginse- target of (20S) ginsenoside Rh2 for its anti-cancer activity. Sci Rep.
noside Rg3 on human hepatocellular carcinoma cells. Mol Med Rep. 2017;7(1):12408.
2012;5(5):1295–8. 257. Li B, Zhao J, Wang CZ, Searle J, He TC, Yuan CS, et al. Ginsenoside Rh2
236. Xie Q, Wen H, Zhang Q, Zhou W, Lin X, Xie D, et al. Inhibiting PI3K-AKt induces apoptosis and paraptosis-like cell death in colorectal cancer
signaling pathway is involved in antitumor effects of ginsenoside Rg3 cells through activation of p53. Cancer Lett. 2011;301(2):185–92.
in lung cancer cell. Biomed Pharmacother. 2017;85:16–21. 258. Zhang F, Li M, Wu X, Hu Y, Cao Y, Wang X, et al. 20(S)-ginsenoside Rg3
237. Sun HY, Lee JH, Han YS, Yoon YM, Yun CW, Kim JH, et al. Pivotal roles promotes senescence and apoptosis in gallbladder cancer cells via the
of ginsenoside Rg3 in tumor apoptosis through regulation of reactive p53 pathway. Drug Design Develop Ther. 2015;9:3969–87.
oxygen species. Anticancer Res. 2016;36(9):4647–54. 259. Wu R, Ru Q, Chen L, Ma B, Li C. Stereospecificity of ginsenoside Rg3 in
238. Kim BM, Kim DH, Park JH, Surh YJ, Na HK. Ginsenoside Rg3 inhibits the promotion of cellular immunity in hepatoma H22-bearing mice. J
constitutive activation of NF-kappaB signaling in human breast cancer Food Sci. 2014;79(7):H1430–5.
(MDA-MB-231) cells: ERK and Akt as potential upstream targets. J Can- 260. Jiang Z, Yang Y, Yang Y, Zhang Y, Yue Z, Pan Z, et al. Ginsenoside Rg3
cer Prev. 2014;19(1):23–30. attenuates cisplatin resistance in lung cancer by downregulating PD-L1
and resuming immune. Biomed Pharmacother. 2017;96:378–83.
Luo et al. Chin Med (2019) 14:48 Page 41 of 58
261. Wang M, Yan SJ, Zhang HT, Li N, Liu T, Zhang YL, et al. Ginsenoside 281. Shan JZ, Xuan YY, Ruan SQ, Sun M. Proliferation-inhibiting and apop-
Rh2 enhances the antitumor immunological response of a mela- tosis-inducing effects of ursolic acid and oleanolic acid on multi-drug
noma mice model. Oncol Lett. 2017;13(2):681–5. resistance cancer cells in vitro. Chin J Integr Med. 2011;17(8):607–11.
262. Sun C, Yu Y, Wang L, Wu B, Xia L, Feng F, et al. Additive antiangiogen- 282. Wang JS, Ren TN, Xi T. Ursolic acid induces apoptosis by suppressing
esis effect of ginsenoside Rg3 with low-dose metronomic temozolo- the expression of FoxM1 in MCF-7 human breast cancer cells. Med
mide on rat glioma cells both in vivo and in vitro. J Exp Clin Cancer Oncol (Northwood, London, England). 2012;29(1):10–5.
Res. 2016;35:32. 283. Shanmugam MK, Rajendran P, Li F, Nema T, Vali S, Abbasi T, et al. Ursolic
263. Li Y, Wang Y, Niu K, Chen X, Xia L, Lu D, et al. Clinical benefit from acid inhibits multiple cell survival pathways leading to suppression
EGFR-TKI plus ginsenoside Rg3 in patients with advanced non- of growth of prostate cancer xenograft in nude mice. J Mol Med.
small cell lung cancer harboring EGFR active mutation. Oncotarget. 2011;89(7):713–27.
2016;7(43):70535–45. 284. Park JH, Kwon HY, Sohn EJ, Kim KA, Kim B, Jeong SJ, et al. Inhibition of
264. Zhou B, Yan Z, Liu R, Shi P, Qian S, Qu X, et al. Prospective study of Wnt/beta-catenin signaling mediates ursolic acid-induced apoptosis in
transcatheter arterial chemoembolization (TACE) with ginsenoside PC-3 prostate cancer cells. Pharmacol Rep. 2013;65(5):1366–74.
Rg3 versus TACE alone for the treatment of patients with advanced 285. Shin SW, Park JW. Ursolic acid sensitizes prostate cancer cells to TRAIL-
hepatocellular carcinoma. Radiology. 2016;280(2):630–9. mediated apoptosis. Biochem Biophys Acta. 2013;1833(3):723–30.
265. Shin SW, Kim SY, Park JW. Autophagy inhibition enhances urso- 286. Xavier CP, Lima CF, Pedro DF, Wilson JM, Kristiansen K, Pereira-Wilson C.
lic acid-induced apoptosis in PC3 cells. Biochem Biophys Acta. Ursolic acid induces cell death and modulates autophagy through JNK
2012;1823(2):451–7. pathway in apoptosis-resistant colorectal cancer cells. J Nutr Biochem.
266. Kashyap D, Sharma A, Tuli HS, Punia S, Sharma AK. Ursolic acid and 2013;24(4):706–12.
oleanolic acid: pentacyclic terpenoids with promising anti-inflam- 287. Prasad S, Yadav VR, Sung B, Reuter S, Kannappan R, Deorukhkar A, et al.
matory activities. Recent Pat Inflammation Allergy Drug Discov. Ursolic acid inhibits growth and metastasis of human colorectal cancer
2016;10(1):21–33. in an orthotopic nude mouse model by targeting multiple cell signal-
267. Zhang Z, Zhang H, Chen R, Wang Z. Oral supplementation with urso- ing pathways: chemosensitization with capecitabine. Clin Cancer Res.
lic acid ameliorates sepsis-induced acute kidney injury in a mouse 2012;18(18):4942–53.
model by inhibiting oxidative stress and inflammatory responses. 288. Hassan L, Pinon A, Limami Y, Seeman J, Fidanzi-Dugas C, Martin F, et al.
Mol Med Rep. 2018;17(5):7142–8. Resistance to ursolic acid-induced apoptosis through involvement of
268. Zhao J, Chen J, Liu T, Fang J, Wan J, Zhao J, et al. Anti-viral effects of melanogenesis and COX-2/PGE2 pathways in human M4Beu mela-
urosolic acid on guinea pig cytomegalovirus in vitro. J Huazhong noma cancer cells. Exp Cell Res. 2016;345(1):60–9.
Univ Sci Technol Med Sci. 2012;32(6):883–7. 289. Kim KH, Seo HS, Choi HS, Choi I, Shin YC, Ko SG. Induction of apoptotic
269. Kaewthawee N, Brimson S. The effects of ursolic acid on cytokine cell death by ursolic acid through mitochondrial death pathway and
production via the MAPK pathways in leukemic T-cells. EXCLI J. extrinsic death receptor pathway in MDA-MB-231 cells. Arch Pharmacal
2013;12:102–14. Res. 2011;34(8):1363–72.
270. Wang CM, Yeh KL, Tsai SJ, Jhan YL, Chou CH. Anti-proliferative activity 290. Leng S, Hao Y, Du D, Xie S, Hong L, Gu H, et al. Ursolic acid promotes
of triterpenoids and sterols isolated from alstonia scholaris against cancer cell death by inducing Atg5-dependent autophagy. Int J Cancer.
non-small-cell lung carcinoma cells. Molecules (Basel, Switzerland). 2013;133(12):2781–90.
2017;22(12):2119. 291. Lewinska A, Adamczyk-Grochala J, Kwasniewicz E, Deregowska A,
271. Li Q, Zhao W, Zeng X, Hao Z. Ursolic acid attenuates atherosclerosis in Wnuk M. Ursolic acid-mediated changes in glycolytic pathway promote
ApoE(-/-) mice: role of LOX-1 Mediated by ROS/NF-kappaB pathway. cytotoxic autophagy and apoptosis in phenotypically different breast
Molecules (Basel, Switzerland). 2018;23(5):1101. cancer cells. Apoptosis. 2017;22(6):800–15.
272. Wang XT, Gong Y, Zhou B, Yang JJ, Cheng Y, Zhao JG, et al. Ursolic 292. Lin CW, Chin HK, Lee SL, Chiu CF, Chung JG, Lin ZY, et al. Ursolic acid
acid ameliorates oxidative stress, inflammation and fibrosis in dia- induces apoptosis and autophagy in oral cancer cells. Environ Toxicol.
betic cardiomyopathy rats. Biomed Pharmacother. 2018;97:1461–7. 2019. https://doi.org/10.1002/tox.22769.
273. Li J, Liang X, Yang X. Ursolic acid inhibits growth and induces apop- 293. Saraswati S, Agrawal SS, Alhaider AA. Ursolic acid inhibits tumor angio-
tosis in gemcitabine-resistant human pancreatic cancer via the JNK genesis and induces apoptosis through mitochondrial-dependent
and PI3K/Akt/NF-kappaB pathways. Oncol Rep. 2012;28(2):501–10. pathway in Ehrlich ascites carcinoma tumor. Chem Biol Interact.
274. Zheng QY, Li PP, Jin FS, Yao C, Zhang GH, Zang T, et al. Ursolic acid 2013;206(2):153–65.
induces ER stress response to activate ASK1-JNK signaling and 294. Guo JL, Han T, Bao L, Li XM, Ma JQ, Tang LP. Ursolic acid promotes the
induce apoptosis in human bladder cancer T24 cells. Cell Signal. apoptosis of cervical cancer cells by regulating endoplasmic reticulum
2013;25(1):206–13. stress. J Obstetr Gynaecol Res. 2019;45(4):877–81.
275. Liu T, Ma H, Shi W, Duan J, Wang Y, Zhang C, et al. Inhibition of STAT3 295. Wu CC, Cheng CH, Lee YH, Chang IL, Chen HY, Hsieh CP, et al. Ursolic
signaling pathway by ursolic acid suppresses growth of hepatocel- acid triggers apoptosis in human osteosarcoma cells via caspase
lular carcinoma. Int J Oncol. 2017;51(2):555–62. activation and the ERK1/2 MAPK pathway. J Agric Food Chem.
276. Huang CY, Lin CY, Tsai CW, Yin MC. Inhibition of cell proliferation, 2016;64(21):4220–6.
invasion and migration by ursolic acid in human lung cancer cell 296. Nam H, Kim MM. Ursolic acid induces apoptosis of SW480 cells via p53
lines. Toxicol In Vitro. 2011;25(7):1274–80. activation. Food Chem Toxicol. 2013;62:579–83.
277. Meng Y, Lin ZM, Ge N, Zhang DL, Huang J, Kong F. Ursolic acid 297. Zhang X, Song X, Yin S, Zhao C, Fan L, Hu H. p21 induction plays
induces apoptosis of prostate cancer cells via the PI3K/Akt/mTOR a dual role in anti-cancer activity of ursolic acid. Exp Biol Med.
pathway. Am J Chin Med. 2015;43(7):1471–86. 2016;241(5):501–8.
278. Zhu Z, Qian Z, Yan Z, Zhao C, Wang H, Ying G. A phase I pharmacoki- 298. Biswas S, Mukherjee PK, Harwansh RK, Bannerjee S, Bhattacharjee P.
netic study of ursolic acid nanoliposomes in healthy volunteers and Enhanced bioavailability and hepatoprotectivity of optimized ursolic
patients with advanced solid tumors. Int J Nanomed. 2013;8:129–36. acid-phospholipid complex. Drug Dev Ind Pharm. 2019;45(6):946–58.
279. Luo J, Hu YL, Wang H. Ursolic acid inhibits breast cancer growth by 299. Rocha TG, Lopes SC, Cassali GD, Ferreira E, Veloso ES, Leite EA, et al.
inhibiting proliferation, inducing autophagy and apoptosis, and sup- Evaluation of antitumor activity of long-circulating and pH-sensitive
pressing inflammatory responses via the PI3K/AKT and NF-kappaB liposomes containing ursolic acid in animal models of breast tumor
signaling pathways in vitro. Exp Ther Med. 2017;14(4):3623–31. and gliosarcoma. Integr Cancer Ther. 2016;15(4):512–24.
280. Qian Z, Wang X, Song Z, Zhang H, Zhou S, Zhao J, et al. A phase I trial 300. Zhao T, Liu Y, Gao Z, Gao D, Li N, Bian Y, et al. Self-assembly and cyto-
to evaluate the multiple-dose safety and antitumor activity of ursolic toxicity study of PEG-modified ursolic acid liposomes. Mater Sci Eng C
acid liposomes in subjects with advanced solid tumors. Biomed Res Mater Biol Appl. 2015;53:196–203.
Int. 2015;2015:7. 301. Lopes S, Novais M, Teixeira C, Honorato-Sampaio K, Pereira M, Ferreira
L, et al. Preparation, physicochemical characterization, and cell viability
Luo et al. Chin Med (2019) 14:48 Page 42 of 58
evaluation of long-circulating and pH-sensitive liposomes containing metastasis of mammalian breast cancer cells in vitro. Acta Pharmacol
ursolic acid. Biomed Res Int. 2013;2013:7. Sin. 2016;37(7):941–9.
302. Wang M, Zhao T, Liu Y, Wang Q, Xing S, Li L, et al. Ursolic acid liposomes 322. Nambiar D, Prajapati V, Agarwal R, Singh RP. In vitro and in vivo antican-
with chitosan modification: promising antitumor drug delivery and cer efficacy of silibinin against human pancreatic cancer BxPC-3 and
efficacy. Mater Sci Eng C Mater Biol Appl. 2017;71:1231–40. PANC-1 cells. Cancer Lett. 2013;334(1):109–17.
303. de Oliveira Eloy J, Saraiva J, de Albuquerque S, Marchetti JM. Solid 323. Akhtar R, Ali M, Mahmood S, Sanyal SN. Anti-proliferative action of
dispersion of ursolic acid in Gelucire 50/13: a strategy to enhance silibinin on human colon adenomatous cancer HT-29 cells. Nutr Hosp.
drug release and trypanocidal activity. AAPS PharmSciTech. 2014;29(2):388–92.
2012;13(4):1436–45. 324. Pashaei-Asl F, Pashaei-Asl R, Khodadadi K, Akbarzadeh A, Ebrahimie
304. Jamal M, Imam SS, Aqil M, Amir M, Mir SR, Mujeeb M. Transdermal E, Pashaiasl M. Enhancement of anticancer activity by silibinin and
potential and anti-arthritic efficacy of ursolic acid from niosomal gel paclitaxel combination on the ovarian cancer. Artif Cells Nanomed
systems. Int Immunopharmacol. 2015;29(2):361–9. Biotechnol. 2018;46(7):1483–7.
305. Li T, Chen X, Liu Y, Fan L, Lin L, Xu Y, et al. pH-Sensitive mesoporous silica 325. Amirsaadat S, Pilehvar-Soltanahmadi Y, Zarghami F, Alipour S, Ebrahimn-
nanoparticles anticancer prodrugs for sustained release of ursolic acid ezhad Z, Zarghami N. Silibinin-loaded magnetic nanoparticles inhibit
and the enhanced anti-cancer efficacy for hepatocellular carcinoma hTERT gene expression and proliferation of lung cancer cells. Artif Cells
cancer. Eur J Pharm Sci. 2017;96:456–63. Nanomed Biotechnol. 2017;45(8):1649–56.
306. Wu CC, Huang YF, Hsieh CP, Chueh PJ, Chen YL. Combined use of 326. Choi ES, Oh S, Jang B, Yu HJ, Shin JA, Cho NP, et al. Silymarin and its
zoledronic acid augments ursolic acid-induced apoptosis in human active component silibinin act as novel therapeutic alternatives for
osteosarcoma cells through enhanced oxidative stress and autophagy. salivary gland cancer by targeting the ERK1/2-Bim signaling cascade.
Molecules (Basel, Switzerland). 2016;21(12):1640. Cell Oncol (Dordrecht). 2017;40(3):235–46.
307. Tremmel L, Rho O, Slaga TJ, DiGiovanni J. Inhibition of skin tumor 327. Deep G, Kumar R, Nambiar DK, Jain AK, Ramteke AM, Serkova NJ, et al.
promotion by TPA using a combination of topically applied ursolic acid Silibinin inhibits hypoxia-induced HIF-1alpha-mediated signaling,
and curcumin. Mol Carcinog. 2019;58(2):185–95. angiogenesis and lipogenesis in prostate cancer cells: in vitro evidence
308. Zong L, Cheng G, Liu S, Pi Z, Liu Z, Song F. Reversal of multidrug and in vivo functional imaging and metabolomics. Mol Carcinog.
resistance in breast cancer cells by a combination of ursolic acid with 2017;56(3):833–48.
doxorubicin. J Pharm Biomed Anal. 2019;165:268–75. 328. Lu S, Zhang Z, Chen M, Li C, Liu L, Li Y. Silibinin inhibits the migration
309. Wang XH, Zhou SY, Qian ZZ, Zhang HL, Qiu LH, Song Z, et al. Evaluation and invasion of human gastric cancer SGC7901 cells by downregulat-
of toxicity and single-dose pharmacokinetics of intravenous ursolic acid ing MMP-2 and MMP-9 expression via the p38MAPK signaling pathway.
liposomes in healthy adult volunteers and patients with advanced solid Oncol Lett. 2017;14(6):7577–82.
tumors. Expert Opin Drug Metab Toxicol. 2013;9(2):117–25. 329. Kim S, Jeon M, Lee J, Han J, Oh SJ, Jung T, et al. Induction of fibronec-
310. Kim BR, Seo HS, Ku JM, Kim GJ, Jeon CY, Park JH, et al. Silibinin inhibits tin in response to epidermal growth factor is suppressed by silibinin
the production of pro-inflammatory cytokines through inhibition of through the inhibition of STAT3 in triple negative breast cancer cells.
NF-kappaB signaling pathway in HMC-1 human mast cells. Inflamma- Oncol Rep. 2014;32(5):2230–6.
tion Res. 2013;62(11):941–50. 330. Ge Y, Zhang Y, Chen Y, Li Q, Chen J, Dong Y, et al. Silibinin causes apop-
311. Bai D, Jin G, Yin S, Zou D, Zhu Q, Yang Z, et al. Antioxidative and anti- tosis and cell cycle arrest in some human pancreatic cancer cells. Int J
apoptotic roles of silibinin in reversing learning and memory deficits in Mol Sci. 2011;12(8):4861–71.
APP/PS1 mice. Neurochem Res. 2017;42(12):3439–45. 331. Woo SM, Min KJ, Kim S, Park JW, Kim DE, Chun KS, et al. Silibinin
312. Harati K, Behr B, Wallner C, Daigeler A, Hirsch T, Jacobsen F, et al. induces apoptosis of HT29 colon carcinoma cells through early
Antiproliferative activity of epigallocatechin3gallate and silibinin on soft growth response-1 (EGR-1)-mediated non-steroidal anti-inflammatory
tissue sarcoma cells. Mol Med Rep. 2017;15(1):103–10. drug-activated gene-1 (NAG-1) up-regulation. Chem Biol Interact.
313. Yun DG, Lee DG. Assessment of silibinin as a potential antifungal 2014;211:36–43.
agent and investigation of its mechanism of action. IUBMB Life. 332. Si L, Liu W, Hayashi T, Ji Y, Fu J, Nie Y, et al. Silibinin-induced apoptosis of
2017;69(8):631–7. breast cancer cells involves mitochondrial impairment. Arch Biochem
314. Vimalraj S, Rajalakshmi S, Saravanan S, Raj-Preeth D, Lav R, Shairam Biophys. 2019;671:42–51.
M, et al. Synthesis and characterization of zinc–silibinin complexes: 333. Jiang K, Wang W, Jin X, Wang Z, Ji Z, Meng G. Silibinin, a natural flavo-
a potential bioactive compound with angiogenic, and antibacte- noid, induces autophagy via ROS-dependent mitochondrial dysfunc-
rial activity for bone tissue engineering. Coll Surf B Biointerfaces. tion and loss of ATP involving BNIP3 in human MCF7 breast cancer cells.
2018;167:134–43. Oncol Rep. 2015;33(6):2711–8.
315. Fernandes V, Sharma D, Kalia K, Tiwari V. Neuroprotective effects of silib- 334. Liu W, Otkur W, Li L, Wang Q, He H, Ye Y, et al. Autophagy induced by
inin: an in silico and in vitro study. Int J Neurosci. 2018;128(10):935–45. silibinin protects human epidermoid carcinoma A431 cells from UVB-
316. Faridnia R, Kalani H, Fakhar M, Akhtari J. Investigating in vitro anti- induced apoptosis. J Photochem Photobiol, B. 2013;123:23–31.
leishmanial effects of silibinin and silymarin on Leishmania major. Ann 335. Bai ZL, Tay V, Guo SZ, Ren J, Shu MG. Silibinin induced human glioblas-
Parasitol. 2018;64(1):29–35. toma cell apoptosis concomitant with autophagy through simultane-
317. Jiang C, Jin S, Jiang Z, Wang J. Inhibitory effects of silibinin on prolifera- ous inhibition of mTOR and YAP. Biomed Res Int. 2018;2018:6165192.
tion and lung metastasis of human high metastasis cell line of salivary 336. Zheng N, Liu L, Liu WW, Li F, Hayashi T, Tashiro SI, et al. Crosstalk of ROS/
gland adenoid cystic carcinoma via autophagy induction. OncoTargets RNS and autophagy in silibinin-induced apoptosis of MCF-7 human
Ther. 2016;9:6609–18. breast cancer cells in vitro. Acta Pharmacol Sin. 2017;38(2):277–89.
318. Ma Z, Liu W, Zeng J, Zhou J, Guo P, Xie H, et al. Silibinin induces apop- 337. Kim SH, Kim KY, Yu SN, Park SK, Choi HD, Ji JH, et al. Autophagy inhibi-
tosis through inhibition of the mTOR-GLI1-BCL2 pathway in renal cell tion enhances silibinin-induced apoptosis by regulating reactive oxy-
carcinoma. Oncol Rep. 2015;34(5):2461–8. gen species production in human prostate cancer PC-3 cells. Biochem
319. Brandon-Warner E, Eheim AL, Foureau DM, Walling TL, Schrum LW, Biophys Res Commun. 2015;468(1–2):151–6.
McKillop IH. Silibinin (Milk Thistle) potentiates ethanol-dependent 338. Zhang Y, Ge Y, Chen Y, Li Q, Chen J, Dong Y, et al. Cellular and molecular
hepatocellular carcinoma progression in male mice. Cancer Lett. mechanisms of silibinin induces cell-cycle arrest and apoptosis on HeLa
2012;326(1):88–95. cells. Cell Biochem Funct. 2012;30(3):243–8.
320. Gandara L, Sandes E, Di Venosa G, Prack Mc Cormick B, Rodriguez L, 339. Zhang X, Liu J, Zhang P, Dai L, Wu Z, Wang L, et al. Silibinin induces
Mamone L, et al. The natural flavonoid silybin improves the response to G1 arrest, apoptosis and JNK/SAPK upregulation in SW1990 human
photodynamic therapy of bladder cancer cells. J Photochem Photobiol pancreatic cancer cells. Oncol Lett. 2018;15(6):9868–76.
B. 2014;133:55–64. 340. Jahanafrooz Z, Motameh N, Bakhshandeh B. Comparative evalu-
321. Sun HP, Su JH, Meng QS, Yin Q, Zhang ZW, Yu HJ, et al. Silibinin and ation of silibinin effects on cell cycling and apoptosis in human
indocyanine green-loaded nanoparticles inhibit the growth and breast cancer MCF-7 and T47D cell lines. Asian Pac J Cancer Prevent.
2016;17(5):2661–5.
Luo et al. Chin Med (2019) 14:48 Page 43 of 58
341. Zhang Y, Li Q, Ge Y, Chen Y, Chen J, Dong Y, et al. Silibinin triggers 360. Chen G, Zhang J, Zhang H, Xiao Y, Kao X, Liu Y, et al. Anti-inflammatory
apoptosis and cell-cycle arrest of SGC7901 cells. Phytother Res. effect of emodin on lipopolysaccharide-induced keratitis in Wistar rats.
2013;27(3):397–403. Int J Clin Exp Med. 2015;8(8):12382–9.
342. Liang L, Li L, Zeng J, Gao Y, Chen YL, Wang ZQ, et al. Inhibitory effect 361. Li Y, Xiong W, Yang J, Zhong J, Zhang L, Zheng J, et al. Attenuation of
of silibinin on EGFR signal-induced renal cell carcinoma progression inflammation by emodin in lipopolysaccharide-induced acute kidney
via suppression of the EGFR/MMP-9 signaling pathway. Oncol Rep. injury via inhibition of toll-like receptor 2 signal pathway. Iran J Kidney
2012;28(3):999–1005. Dis. 2015;9(3):202–8.
343. Liu W, Otkur W, Li L, Wang Q, He H, Zang L, et al. Interference of 362. Chen GL, Zhang JJ, Kao X, Wei LW, Liu ZY. Emodin ameliorates lipopol-
silibinin with IGF-1R signalling pathways protects human epidermoid ysaccharides-induced corneal inflammation in rats. Int J Ophthalmol.
carcinoma A431 cells from UVB-induced apoptosis. Biochem Biophys 2015;8(4):665–9.
Res Commun. 2013;432(2):314–9. 363. Park SY, Jin ML, Ko MJ, Park G, Choi YW. Anti-neuroinflammatory effect
344. Oh SJ, Jung SP, Han J, Kim S, Kim JS, Nam SJ, et al. Silibinin inhibits of emodin in LPS-stimulated microglia: involvement of AMPK/Nrf2
TPA-induced cell migration and MMP-9 expression in thyroid and activation. Neurochem Res. 2016;41(11):2981–92.
breast cancer cells. Oncol Rep. 2013;29(4):1343–8. 364. Yao WY, Zhou YF, Qian AH, Zhang YP, Qiao MM, Zhai ZK, et al. Emodin
345. Yousefi M, Ghaffari SH, Zekri A, Hassani S, Alimoghaddam K, has a protective effect in cases of severe acute pancreatitis via inhibi-
Ghavamzadeh A. Silibinin induces apoptosis and inhibits proliferation tion of nuclear factor-kappa B activation resulting in antioxidation. Mol
of estrogen receptor (ER)-negative breast carcinoma cells through Med Rep. 2015;11(2):1416–20.
suppression of nuclear factor kappa B activation. Arch Iran Med. 365. Alisi A, Pastore A, Ceccarelli S, Panera N, Gnani D, Bruscalupi G, et al.
2014;17(5):366–71. Emodin prevents intrahepatic fat accumulation, inflammation and
346. Yousefi M, Ghaffari SH, Soltani BM, Nafissi S, Momeny M, Zekri A, et al. redox status imbalance during diet-induced hepatosteatosis in rats. Int
Therapeutic efficacy of silibinin on human neuroblastoma cells: akt J Mol Sci. 2012;13(2):2276–89.
and NF-kappaB expressions may play an important role in silibinin- 366. Sharma R, Tiku AB. Emodin, an anthraquinone derivative, protects
induced response. Neurochem Res. 2012;37(9):2053–63. against gamma radiation-induced toxicity by inhibiting DNA damage
347. Raina K, Agarwal C, Agarwal R. Effect of silibinin in human colorectal and oxidative stress. Int J Radiat Biol. 2014;90(4):275–83.
cancer cells: targeting the activation of NF-kappaB signaling. Mol 367. Ma L, Yang Y, Yin Z, Liu M, Wang L, Chen L, et al. Emodin suppresses the
Carcinog. 2013;52(3):195–206. nasopharyngeal carcinoma cells by targeting the chloride channels.
348. Duan WJ, Li QS, Xia MY, Tashiro S, Onodera S, Ikejima T. Silibinin Biomed Pharmacother. 2017;90:615–25.
activated ROS-p38-NF-kappaB positive feedback and induced 368. Li XX, Dong Y, Wang W, Wang HL, Chen YY, Shi GY, et al. Emodin as an
autophagic death in human fibrosarcoma HT1080 cells. J Asian Nat effective agent in targeting cancer stem-like side population cells of
Prod Res. 2011;13(1):27–35. gallbladder carcinoma. Stem Cells Develop. 2013;22(4):554–66.
349. Zeng J, Sun Y, Wu K, Li L, Zhang G, Yang Z, et al. Chemopreven- 369. Haque E, Kamil M, Irfan S, Sheikh S, Hasan A, Nazir A, et al. Block-
tive and chemotherapeutic effects of intravesical silibinin against ing mutation independent p53 aggregation by emodin modulates
bladder cancer by acting on mitochondria. Mol Cancer Ther. autophagic cell death pathway in lung cancer. Int J Biochem Cell Biol.
2011;10(1):104–16. 2018;96:90–5.
350. Tiwari P, Kumar A, Balakrishnan S, Kushwaha HS, Mishra KP. Silibinin- 370. Zhang X, Chen Y, Zhang T, Zhang Y. Inhibitory effect of emodin on
induced apoptosis in MCF7 and T47D human breast carcinoma cells human hepatoma cell line SMMC-7721 and its mechanism. Afr Health
involves caspase-8 activation and mitochondrial pathway. Cancer Sci. 2015;15(1):97–100.
Invest. 2011;29(1):12–20. 371. Ma YS, Weng SW, Lin MW, Lu CC, Chiang JH, Yang JS, et al. Antitumor
351. Forghani P, Khorramizadeh MR, Waller EK. Silibinin inhibits accumula- effects of emodin on LS1034 human colon cancer cells in vitro and
tion of myeloid-derived suppressor cells and tumor growth of murine in vivo: roles of apoptotic cell death and LS1034 tumor xenografts
breast cancer. Cancer Med. 2014;3(2):215–24. model. Food Chem Toxicol. 2012;50(5):1271–8.
352. Ting H, Deep G, Kumar S, Jain AK, Agarwal C, Agarwal R. Beneficial 372. Manimaran A, Buddhan R, Manoharan S. Emodin downregulates
effects of the naturally occurring flavonoid silibinin on the prostate cell proliferation markers during DMBA induced oral carcinogenesis
cancer microenvironment: role of monocyte chemotactic protein-1 in Golden Syrian Hamsters. Afr J Tradit Complement Altern Med.
and immune cell recruitment. Carcinogenesis. 2016;37(6):589–99. 2017;14(2):83–91.
353. Gohulkumar M, Gurushankar K, Rajendra Prasad N, Krishnakumar N. 373. Lu J, Xu Y, Zhao Z, Ke X, Wei X, Kang J, et al. Emodin suppresses
Enhanced cytotoxicity and apoptosis-induced anticancer effect of proliferation, migration and invasion in ovarian cancer cells by down
silibinin-loaded nanoparticles in oral carcinoma (KB) cells. Mater Sci Eng regulating ILK in vitro and in vivo. OncoTargets Ther. 2017;10:3579–89.
C Mater Biol Appl. 2014;41:274–82. 374. Cha TL, Chuang MJ, Tang SH, Wu ST, Sun KH, Chen TT, et al. Emodin
354. Yazdi Rouholamini SE, Moghassemi S, Maharat Z, Hakamivala A, modulates epigenetic modifications and suppresses bladder carcinoma
Kashanian S, Omidfar K. Effect of silibinin-loaded nano-niosomal cell growth. Mol Carcinog. 2015;54(3):167–77.
coated with trimethyl chitosan on miRNAs expression in 2D and 3D 375. Deng G, Ju X, Meng Q, Yu ZJ, Ma LB. Emodin inhibits the proliferation of
models of T47D breast cancer cell line. Artif Cells Nanomed Biotechnol. PC3 prostate cancer cells in vitro via the Notch signaling pathway. Mol
2018;46(3):524–35. Med Rep. 2015;12(3):4427–33.
355. Vue B, Zhang S, Zhang X, Parisis K, Zhang Q, Zheng S, et al. Silibinin 376. Sun Y, Wang X, Zhou Q, Lu Y, Zhang H, Chen Q, et al. Inhibitory effect of
derivatives as anti-prostate cancer agents: synthesis and cell-based emodin on migration, invasion and metastasis of human breast cancer
evaluations. Eur J Med Chem. 2016;109:36–46. MDA-MB-231 cells in vitro and in vivo. Oncol Rep. 2015;33(1):338–46.
356. Pooja D, Babu Bikkina DJ, Kulhari H, Nikhila N, Chinde S, Raghavendra 377. Chihara T, Shimpo K, Beppu H, Yamamoto N, Kaneko T, Wakamatsu
YM, et al. Fabrication, characterization and bioevaluation of silibinin K, et al. Effects of aloe-emodin and emodin on proliferation of the
loaded chitosan nanoparticles. Int J Biol Macromol. 2014;69:267–73. MKN45 human gastric cancer cell line. Asian Pac J Cancer Prev.
357. Nasiri M, Zarghami N, Koshki KN, Mollazadeh M, Moghaddam MP, 2015;16(9):3887–91.
Yamchi MR, et al. Curcumin and silibinin inhibit telomerase expres- 378. Liu A, Chen H, Wei W, Ye S, Liao W, Gong J, et al. Antiproliferative and
sion in T47D human breast cancer cells. Asian Pac J Cancer Prevent. antimetastatic effects of emodin on human pancreatic cancer. Oncol
2013;14(6):3449–53. Rep. 2011;26(1):81–9.
358. Chakrabarti M, Ray SK. Synergistic anti-tumor actions of luteolin and 379. Qu W, Wang Y, Wu Q, Liu J, Hao D. Emodin inhibits HMGB1-induced
silibinin prevented cell migration and invasion and induced apoptosis tumor angiogenesis in human osteosarcoma by regulating SIRT1. Int J
in glioblastoma SNB19 cells and glioblastoma stem cells. Brain Res. Clin Exp Med. 2015;8(9):15054–64.
2015;1629:85–93. 380. Wang X, Li L, Guan R, Zhu D, Song N, Shen L. Emodin inhibits ATP-
359. Nafees S, Mehdi SH, Zafaryab M, Zeya B, Sarwar T, Rizvi MA. Synergistic induced proliferation and migration by suppressing P2Y recep-
interaction of rutin and silibinin on human colon cancer cell line. Arch tors in human lung adenocarcinoma cells. Cell Physiol Biochem.
Med Res. 2018;49(4):226–34. 2017;44(4):1337–51.
Luo et al. Chin Med (2019) 14:48 Page 44 of 58
381. Dong X, Ni B, Fu J, Yin X, You L, Leng X, et al. Emodin induces apoptosis 402. Wang ZH, Chen H, Guo HC, Tong HF, Liu JX, Wei WT, et al. Enhanced
in human hepatocellular carcinoma HepaRG cells via the mitochondrial antitumor efficacy by the combination of emodin and gemcitabine
caspasedependent pathway. Oncol Rep. 2018;40(4):1985–93. against human pancreatic cancer cells via downregulation of the
382. Wang Y, Luo Q, He X, Wei H, Wang T, Shao J, et al. Emodin induces expression of XIAP in vitro and in vivo. Int J Oncol. 2011;39(5):1123–31.
apoptosis of colon cancer cells via induction of autophagy in a ROS- 403. Liu A, Chen H, Tong H, Ye S, Qiu M, Wang Z, et al. Emodin potentiates
dependent manner. Oncol Res. 2017;5:889–99. the antitumor effects of gemcitabine in pancreatic cancer cells via
383. Song X, Zhou X, Qin Y, Yang J, Wang Y, Sun Z, et al. Emodin inhibits inhibition of nuclear factor-kappaB. Mol Med Rep. 2011;4(2):221–7.
epithelialmesenchymal transition and metastasis of triple negative 404. Chen H, Wei W, Guo Y, Liu A, Tong H, Wang Z, et al. Enhanced effect
breast cancer via antagonism of CCchemokine ligand 5 secreted from of gemcitabine by emodin against pancreatic cancer in vivo via
adipocytes. Int J Mol Med. 2018;42(1):579–88. cytochrome C-regulated apoptosis. Oncol Rep. 2011;25(5):1253–61.
384. Dai G, Ding K, Cao Q, Xu T, He F, Liu S, et al. Emodin suppresses growth 405. Li X, Wang H, Wang J, Chen Y, Yin X, Shi G, et al. Emodin enhances
and invasion of colorectal cancer cells by inhibiting VEGFR2. Eur J cisplatin-induced cytotoxicity in human bladder cancer cells through
Pharmacol. 2019;859:172525. ROS elevation and MRP1 downregulation. BMC Cancer. 2016;16(1):578.
385. Li N, Wang C, Zhang P, You S. Emodin inhibits pancreatic cancer 406. Li H, Pan GF, Jiang ZZ, Yang J, Sun LX, Zhang LY. Triptolide inhib-
EMT and invasion by upregulating microRNA1271. Mol Med Rep. its human breast cancer MCF-7 cell growth via downregulation
2018;18(3):3366–74. of the ERalpha-mediated signaling pathway. Acta Pharmacol Sin.
386. Lin SZ, Xu JB, Ji X, Chen H, Xu HT, Hu P, et al. Emodin inhibits angio- 2015;36(5):606–13.
genesis in pancreatic cancer by regulating the transforming growth 407. Bai S, Hu Z, Yang Y, Yin Y, Li W, Wu L, et al. Anti-inflammatory and
factor-beta/drosophila mothers against decapentaplegic pathway and neuroprotective effects of triptolide via the NF-kappaB signaling path-
angiogenesis-associated microRNAs. Mol Med Rep. 2015;12(4):5865–71. way in a Rat MCAO Model. Anatomical Record (Hoboken, NJ: 2007).
387. Lin SZ, Wei WT, Chen H, Chen KJ, Tong HF, Wang ZH, et al. Antitumor 2016;299(2):256–66.
activity of emodin against pancreatic cancer depends on its dual role: 408. Reno TA, Kim JY, Raz DJ. Triptolide inhibits lung cancer cell migration,
promotion of apoptosis and suppression of angiogenesis. PLoS ONE. invasion, and metastasis. Ann Thor Surg. 2015;100(5):1817–24.
2012;7(8):e42146. 409. Ho JN, Byun SS, Lee S, Oh JJ, Hong SK, Lee SE, et al. Synergistic anti-
388. Shi GH, Zhou L. Emodin suppresses angiogenesis and metastasis in tumor effect of triptolide and cisplatin in cisplatin resistant human
anaplastic thyroid cancer by affecting TRAF6mediated pathways in vivo bladder cancer cells. J Urol. 2015;193(3):1016–22.
and in vitro. Mol Med Rep. 2018;18(6):5191–7. 410. Wang H, Ma D, Wang C, Zhao S, Liu C. Triptolide inhibits invasion and
389. Sui JQ, Xie KP, Zou W, Xie MJ. Emodin inhibits breast cancer cell tumorigenesis of hepatocellular carcinoma MHCC-97H cells through
proliferation through the ERalpha-MAPK/Akt-cyclin D1/Bcl-2 signaling NF-kappaB signaling. Med Sci Monit. 2016;22:1827–36.
pathway. Asian Pac J Cancer Prev. 2014;15(15):6247–51. 411. Nakazato T, Sagawa M, Kizaki M. Triptolide induces apoptotic cell death
390. Saunders IT, Mir H, Kapur N, Singh S. Emodin inhibits colon cancer by of multiple myeloma cells via transcriptional repression of Mcl-1. Int J
altering BCL-2 family proteins and cell survival pathways. Cancer Cell Oncol. 2014;44(4):1131–8.
Int. 2019;19:98. 412. Chen J, Qiao Y, Tang B, Chen G, Liu X, Yang B, et al. Modulation of
391. Ying J, Xu H, Wu D, Wu X. Emodin induces apoptosis of human osteo- salmonella tumor-colonization and intratumoral anti-angiogenesis by
sarcoma cells via mitochondria- and endoplasmic reticulum stress- triptolide and its mechanism. Theranostics. 2017;7(8):2250–60.
related pathways. Int J Clin Exp Pathol. 2015;8(10):12837–44. 413. Liu Y, Xiao E, Yuan L, Li G. Triptolide synergistically enhances antitumor
392. Zhang L, He D, Li K, Liu H, Wang B, Zheng L, et al. Emodin targets mito- activity of oxaliplatin in colon carcinoma in vitro and in vivo. DNA Cell
chondrial cyclophilin D to induce apoptosis in HepG2 cells. Biomed Biol. 2014;33(7):418–25.
Pharmacother. 2017;90:222–8. 414. Ding X, Zhou X, Jiang B, Zhao Q, Zhou G. Triptolide suppresses prolifera-
393. Su J, Yan Y, Qu J, Xue X, Liu Z, Cai H. Emodin induces apoptosis of lung tion, hypoxia-inducible factor-1alpha and c-Myc expression in pancre-
cancer cells through ER stress and the TRIB3/NF-kappaB pathway. atic cancer cells. Mol Med Rep. 2015;12(3):4508–13.
Oncol Rep. 2017;37(3):1565–72. 415. Zhao H, Yang Z, Wang X, Zhang X, Wang M, Wang Y, et al. Triptolide
394. Iwanowycz S, Wang J, Hodge J, Wang Y, Yu F, Fan D. Emodin inhibits inhibits ovarian cancer cell invasion by repression of matrix metal-
breast cancer growth by blocking the tumor-promoting feedfor- loproteinase 7 and 19 and upregulation of E-cadherin. Exp Mol Med.
ward loop between cancer cells and macrophages. Mol Cancer Ther. 2012;44(11):633–41.
2016;15(8):1931–42. 416. Liu S, Zhang J, Zhang X-Z, Zhang H-H, Li X-W, Zhang S-J. Triptolide
395. Manu KA, Shanmugam MK, Ong TH, Subramaniam A, Siveen KS, Peru- induces cell apoptosis in human stomach cancer cell via caspase
mal E, et al. Emodin suppresses migration and invasion through the 3-dependent cascade pathway. Trop J Pharm Res. 2016;15(9):1853–8.
modulation of CXCR398 expression in an orthotopic model of human 417. Yuan S, Wang L, Chen X, Fan B, Yuan Q, Zhang H, et al. Triptolide inhibits
hepatocellular carcinoma. PLoS ONE. 2013;8(3):e57015. the migration and invasion of human prostate cancer cells via Caveo-
396. Zhang W, Li H, Bu H, Chen H, Tong H, Liu D, et al. Emodin inhibits the lin-1/CD147/MMPs pathway. Biomed Pharmacother. 2016;84:1776–82.
differentiation and maturation of dendritic cells and increases the 418. Chan SF, Chen YY, Lin JJ, Liao CL, Ko YC, Tang NY, et al. Triptolide induced
production of regulatory T cells. Int J Mol Med. 2012;29(2):159–64. cell death through apoptosis and autophagy in murine leukemia
397. Iwanowycz S, Wang J, Altomare D, Hui Y, Fan D. Emodin bidirection- WEHI-3 cells in vitro and promoting immune responses in WEHI-3
ally modulates macrophage polarization and epigenetically regulates generated leukemia mice in vivo. Environ Toxicol. 2017;32(2):550–68.
macrophage memory. J Biol Chem. 2016;291(22):11491–503. 419. Yang CY, Lin CK, Lin GJ, Hsieh CC, Huang SH, Ma KH, et al. Triptolide
398. Li Q, Wen J, Yu K, Shu Y, He W, Chu H, et al. Aloe-emodin induces represses oral cancer cell proliferation, invasion, migration, and
apoptosis in human oral squamous cell carcinoma SCC15 cells. BMC angiogenesis in co-inoculation with U937 cells. Clin Oral Invest.
Complement Altern Med. 2018;18(1):296. 2017;21(1):419–27.
399. Gao R, Wu X, Huang Z, Wang B, Li F, Xu H, et al. Anti-tumor effect of 420. Qin G, Li P, Xue Z. Triptolide induces protective autophagy and apop-
aloe-emodin on cervical cancer cells was associated with human tosis in human cervical cancer cells by downregulating Akt/mTOR
papillomavirus E6/E7 and glucose metabolism. OncoTargets Ther. activation. Oncol Lett. 2017;16(3):3929–34.
2019;12:3713–21. 421. Krosch TC, Sangwan V, Banerjee S, Mujumdar N, Dudeja V, Saluja AK,
400. Yang L, Lin S, Kang Y, Xiang Y, Xu L, Li J, et al. Rhein sensitizes human et al. Triptolide-mediated cell death in neuroblastoma occurs by both
pancreatic cancer cells to EGFR inhibitors by inhibiting STAT3 pathway. apoptosis and autophagy pathways and results in inhibition of nuclear
J Exp Clin Cancer Res: CR. 2019;38(1):31. factor-kappa B activity. Am J Surg. 2013;205(4):387–96.
401. Zhou G, Peng F, Zhong Y, Chen Y, Tang M, Li D. Rhein suppresses 422. Zhao L, Jiang BO, Wang D, Liu W, Zhang H, Liu W, et al. Triptolide
matrix metalloproteinase production by regulating the Rac1/ROS/ reduces the viability of osteosarcoma cells by reducing MKP-1 and
MAPK/AP-1 pathway in human ovarian carcinoma cells. Int J Oncol. Hsp70 expression. Exp Ther Med. 2016;11(5):2005–10.
2017;50(3):933–41. 423. Huang Y, Wu S, Zhang Y, Wang L, Guo Y. Antitumor effect of triptolide in
T-cell lymphoblastic lymphoma by inhibiting cell viability, invasion, and
Luo et al. Chin Med (2019) 14:48 Page 45 of 58
epithelial-mesenchymal transition via regulating the PI3K/AKT/mTOR 445. Liu X, Wang K, Duan N, Lan Y, Ma P, Zheng H, et al. Computational pre-
pathway. OncoTargets Ther. 2018;11:769–79. diction and experimental validation of low-affinity target of triptolide
424. Brincks EL, Kucaba TA, James BR, Murphy KA, Schwertfeger KL, Sangwan and its analogues. RSC Adv. 2015;5(44):34572–9.
V, et al. Triptolide enhances the tumoricidal activity of TRAIL against 446. Hu H, Huang G, Wang H, Li X, Wang X, Feng Y, et al. Inhibition effect
renal cell carcinoma. FEBS J. 2015;282(24):4747–65. of triptolide on human epithelial ovarian cancer via adjusting cellular
425. Zhang W, Kang M, Zhang T, Li B, Liao X, Wang R. Triptolide combined immunity and angiogenesis. Oncol Rep. 2018;39(3):1191–6.
with radiotherapy for the treatment of nasopharyngeal carcinoma via 447. Reno TA, Tong SW, Wu J, Fidler JM, Nelson R, Kim JY, et al. The triptolide
NF-kappaB-related mechanism. Int J Mol Sci. 2016;17(12):2139. derivative MRx102 inhibits Wnt pathway activation and has potent anti-
426. Ni J, Wu Q, Sun ZH, Zhong J, Cai Y, Huang XE. The inhibition effect of tumor effects in lung cancer. BMC Cancer. 2016;16:439.
triptolide on human endometrial carcinoma cell line HEC-1B: a in vitro 448. Modi S, Kir D, Giri B, Majumder K, Arora N, Dudeja V, et al. Minnelide
and in vivo studies. Asian Pac J Cancer Prevent. 2015;16(11):4571–6. overcomes oxaliplatin resistance by downregulating the DNA repair
427. You L, Dong X, Ni B, Fu J, Yang C, Yin X, et al. Triptolide induces apopto- pathway in pancreatic cancer. J Gastrointest Surg. 2016;20(1):13–23
sis through fas death and mitochondrial pathways in HepaRG cell line. (discussion-4).
Front Pharmacol. 2018;9:813. 449. Yuan ZX, Wu XJ, Mo J, Wang YL, Xu CQ, Lim LY. Renal targeted delivery
428. Wang Y, Guo SH, Shang XJ, Yu LS, Zhu JW, Zhao A, et al. Triptolide of triptolide by conjugation to the fragment peptide of human serum
induces Sertoli cell apoptosis in mice via ROS/JNK-dependent activa- albumin. Eur J Pharm Biopharm. 2015;94:363–71.
tion of the mitochondrial pathway and inhibition of Nrf2-mediated 450. Lin C, Zhang X, Chen H, Bian Z, Zhang G, Riaz MK, et al. Dual-ligand
antioxidant response. Acta Pharmacol Sin. 2018;39(2):311–27. modified liposomes provide effective local targeted delivery of lung-
429. Nardi I, Reno T, Yun X, Sztain T, Wang J, Dai H, et al. Triptolide inhibits cancer drug by antibody and tumor lineage-homing cell-penetrating
Wnt signaling in NSCLC through upregulation of multiple Wnt inhibi- peptide. Drug Deliv. 2018;25(1):256–66.
tory factors via epigenetic modifications to Histone H3. Int J Cancer. 451. Liu H, Shen M, Zhao X, Ru D, Duan Y, Ding C, et al. The effect of trip-
2018;143(10):2470–8. tolide-loaded exosomes on the proliferation and apoptosis of human
430. Mao X, Tong J, Wang Y, Zhu Z, Yin Y, Wang Y. Triptolide exhibits antitu- ovarian cancer SKOV3 cells. BioMed Res Int. 2019;2019:2595801.
mor effects by reversing hypermethylation of WIF1 in lung cancer cells. 452. Aribi A, Gery S, Lee DH, Thoennissen NH, Thoennissen GB, Alvarez
Mol Med Rep. 2018;18(3):3041–9. R, et al. The triterpenoid cucurbitacin B augments the antiprolifera-
431. Li X, Lu Q, Xie W, Wang Y, Wang G. Anti-tumor effects of triptolide on tive activity of chemotherapy in human breast cancer. Int J Cancer.
angiogenesis and cell apoptosis in osteosarcoma cells by inducing 2013;132(12):2730–7.
autophagy via repressing Wnt/beta-Catenin signaling. Biochem Bio- 453. Wang W, Wang Q, Wang L, Li X, Liu D. Enhanced antitumor effect via
phys Res Commun. 2018;496(2):443–9. combination of triptolide with 5-fluorouracil in pancreatic cancer. Trans-
432. Kong J, Wang L, Ren L, Yan Y, Cheng Y, Huang Z, et al. Triptolide induces lat Cancer Res. 2018;7(1):142–50.
mitochondria-mediated apoptosis of Burkitt’s lymphoma cell via 454. Wang G, Wang X, Xu X. Triptolide potentiates lung cancer cells to
deacetylation of GSK-3beta by increased SIRT3 expression. Toxicol Appl cisplatin-induced apoptosis by selectively inhibiting the NER activity.
Pharmacol. 2018;342:1–13. Biomarker Res. 2015;3:17.
433. Jiang J, Song X, Yang J, Lei K, Ni Y, Zhou F, et al. Triptolide inhibits 455. Qi X, Li C, Wu C, Yu C, Liu M, Gao M, et al. Dephosphorylation of Tak1 at
proliferation and migration of human neuroblastoma SH-SY5Y cells by Ser412 greatly contributes to the spermatocyte-specific testis toxicity
upregulating MicroRNA-181a. Oncol Res. 2018;26(8):1235–43. induced by (5R)-5-hydroxytriptolide in C57BL/6 mice. Toxicol Res
434. Gao H, Zhang Y, Dong L, Qu XY, Tao LN, Zhang YM, et al. Triptolide (Camb). 2016;5(2):594–601.
induces autophagy and apoptosis through ERK activation in human 456. Wang L, Xu Y, Fu L, Li Y, Lou L. (5R)-5-hydroxytriptolide (LLDT-8), a novel
breast cancer MCF-7 cells. Exp Ther Med. 2018;15(4):3413–9. immunosuppressant in clinical trials, exhibits potent antitumor activity
435. Zhao F, Huang W, Zhang Z, Mao L, Han Y, Yan J, et al. Triptolide induces via transcription inhibition. Cancer Lett. 2012;324(1):75–82.
protective autophagy through activation of the CaMKKbeta-AMPK sign- 457. Greeno E, Borazanci E, Gockerman J, Korn R, Saluja A, Von Hoff D.
aling pathway in prostate cancer cells. Oncotarget. 2016;7(5):5366–82. Abstract CT207: phase I dose escalation and pharmokinetic study
436. Liu H, Tang L, Li X, Li H. Triptolide inhibits vascular endothelial growth of 14-O-phosphonooxymethyltriptolide. Cancer Res. 2015;75(15
factor-mediated angiogenesis in human breast cancer cells. Exp Ther Supplement):CT207.
Med. 2018;16(2):830–6. 458. Noel P, Von Hoff DD, Saluja AK, Velagapudi M, Borazanci E, Han H.
437. Ma JX, Sun YL, Wang YQ, Wu HY, Jin J, Yu XF. Triptolide induces apoptosis Triptolide and its derivatives as cancer therapies. Trends Pharmacol Sci.
and inhibits the growth and angiogenesis of human pancreatic cancer 2019;40(5):327–41.
cells by downregulating COX-2 and VEGF. Oncol Res. 2013;20(8):359–68. 459. Wang Y, Zhao G-X, Xu L-H, Liu K-P, Pan H, He J, et al. Cucurbitacin IIb
438. Kumar A, Corey C, Scott I, Shiva S, D’Cunha J. Minnelide/triptolide exhibits anti-inflammatory activity through modulating multiple cel-
impairs mitochondrial function by regulating SIRT3 in P53-dependent lular behaviors of mouse lymphocytes. PLoS ONE. 2014;9(2):e89751.
manner in non-small cell lung cancer. PLoS ONE. 2016;11(8):e0160783. 460. Ma J, Zi Jiang Y, Shi H, Mi C, Li J, Xing Nan J, et al. Cucurbitacin B inhibits
439. Kwon HY, Kim KS, An HK, Moon HI, Kim HJ, Lee YC. Triptolide induces the translational expression of hypoxia-inducible factor-1alpha. Eur J
apoptosis through extrinsic and intrinsic pathways in human osteosar- Pharmacol. 2014;723:46–54.
coma U2OS cells. Indian J Biochem Biophys. 2013;50(6):485–91. 461. Piao XM, Gao F, Zhu JX, Wang LJ, Zhao X, Li X, et al. Cucurbitacin B
440. Kwon HY, Kim KS, Baik JS, Moon HI, Lee JW, Kim CH, et al. Triptolide- inhibits tumor angiogenesis by triggering the mitochondrial signaling
mediated apoptosis by suppression of focal adhesion kinase through pathway in endothelial cells. Int J Mol Med. 2018;42(2):1018–25.
extrinsic and intrinsic pathways in human melanoma cells. Evid Based 462. Zhang M, Bian ZG, Zhang Y, Wang JH, Kan L, Wang X, et al. Cucurbitacin
Complement Altern Med. 2013;2013:172548. B inhibits proliferation and induces apoptosis via STAT3 pathway inhibi-
441. Wu PP, Liu KC, Huang WW, Ma CY, Lin H, Yang JS, et al. Triptolide tion in A549 lung cancer cells. Mol Med Rep. 2014;10(6):2905–11.
induces apoptosis in human adrenal cancer NCI-H295 cells through a 463. Tseng L-M, Huang P-I, Chen Y-R, Chen Y-C, Chou Y-C, Chen Y-W, et al.
mitochondrial-dependent pathway. Oncol Rep. 2011;25(2):551–7. Targeting signal transducer and activator of transcription 3 pathway
442. Tan BJ, Chiu GN. Role of oxidative stress, endoplasmic reticulum by cucurbitacin I diminishes self-renewing and radiochemoresistant
stress and ERK activation in triptolide-induced apoptosis. Int J Oncol. abilities in thyroid cancer-derived CD133 + cells. J Pharmacol Exp Ther.
2013;42(5):1605–12. 2012;341(2):410–23.
443. Yanchun M, Yi W, Lu W, Yu Q, Jian Y, Pengzhou K, et al. Triptolide pre- 464. Huang W-W, Yang J-S, Lin M-W, Chen P-Y, Chiou S-M, Chueh F-S, et al.
vents proliferation and migration of Esophageal Squamous Cell Cancer Cucurbitacin E induces G(2)/M phase arrest through STAT3/p53/p21
via MAPK/ERK signaling pathway. Eur J Pharmacol. 2019;851:43–51. signaling and provokes apoptosis via Fas/CD95 and mitochondria-
444. Jao HY, Yu FS, Yu CS, Chang SJ, Liu KC, Liao CL, et al. Suppression of dependent pathways in human bladder cancer T24 cells. Evid Based
the migration and invasion is mediated by triptolide in B16F10 mouse Complement Alternat Med. 2012;2012:952762.
melanoma cells through the NF-kappaB-dependent pathway. Environ 465. Deng C, Zhang B, Zhang S, Duan C, Cao Y, Kang W, et al. Low nanomolar
Toxicol. 2016;31(12):1974–84. concentrations of Cucurbitacin-I induces G2/M phase arrest and
Luo et al. Chin Med (2019) 14:48 Page 46 of 58
apoptosis by perturbing redox homeostasis in gastric cancer cells suppresses brain metastasis of human nonsmall cell lung cancer in a
in vitro and in vivo. Cell Death Dis. 2016;7(2):e2106. murine model. Oncol Rep. 2019;42:697–707.
466. Liu J, Liu X, Ma W, Kou W, Li C, Zhao J. Anticancer activity of cucur- 487. Song J, Liu H, Li Z, Yang C, Wang C. Cucurbitacin I inhibits cell migra-
bitacin-A in ovarian cancer cell line SKOV3 involves cell cycle arrest, tion and invasion and enhances chemosensitivity in colon cancer.
apoptosis and inhibition of mTOR/PI3K/Akt signaling pathway. J BUON. Oncol Rep. 2015;33(4):1867–71.
2018;23(1):124–8. 488. Kim HJ, Kim J-K. Antiangiogenic effects of cucurbitacin-I. Arch Phar-
467. El-Senduny FF, Badria FA, El-Waseef AM, Chauhan SC, Halaweish F. macal Res. 2015;38(2):290–8.
Approach for chemosensitization of cisplatin-resistant ovarian cancer 489. Ku JM, Kim SR, Hong SH, Choi H-S, Seo HS, Shin YC, et al. Cucurbitacin
by cucurbitacin B. Tumor Biol. 2016;37(1):685–98. D induces cell cycle arrest and apoptosis by inhibiting STAT3 and
468. Lan T, Wang L, Xu Q, Liu W, Jin H, Mao W, et al. Growth inhibitory NF-κB signaling in doxorubicin-resistant human breast carcinoma
effect of Cucurbitacin E on breast cancer cells. Int J Clin Exp Pathol. (MCF7/ADR) cells. Mol Cell Biochem. 2015;409(1–2):33–43.
2013;6(9):1799–805. 490. Hsu YC, Huang TY, Chen MJ. Therapeutic ROS targeting of GADD-
469. Lopez-Haber C, Kazanietz MG. Cucurbitacin I inhibits Rac1 activation in 45gamma in the induction of G2/M arrest in primary human colorec-
breast cancer cells by a reactive oxygen species-mediated mechanism tal cancer cell lines by cucurbitacin E. Cell Death Dis. 2014;5:e1198.
and independently of Janus tyrosine kinase 2 and P-Rex1. Mol Pharma- 491. Spear SA, Burns SS, Oblinger JL, Ren Y, Pan L, Kinghorn AD, et al. Natu-
col. 2013;83(5):1141–54. ral compounds as potential treatments of NF2-deficient schwan-
470. Hung C-M, Chang C-C, Lin C-W, Ko S-Y, Hsu Y-C. Cucurbitacin E as noma and meningioma: cucurbitacin D and goyazensolide. Otol
inducer of cell death and apoptosis in human oral squamous cell Neurotol. 2013;34(8):1519–27.
carcinoma cell line SAS. Int J Mol Sci. 2013;14(8):17147–56. 492. Kausar H, Munagala R, Bansal SS, Aqil F, Vadhanam MV, Gupta RC.
471. Wang Y, Xu S, Wu Y, Zhang J. Cucurbitacin E inhibits osteosarcoma cells Cucurbitacin B potently suppresses non-small-cell lung cancer
proliferation and invasion through attenuation of PI3K/AKT/mTOR growth: identification of intracellular thiols as critical targets. Cancer
signalling pathway. Biosci Rep. 2016;36(6):e00405. Lett. 2013;332(1):35–45.
472. Gao Y, Islam MS, Tian J, Lui VW, Xiao D. Inactivation of ATP citrate lyase 493. Li H, Chen H, Li R, Xin J, Wu S, Lan J, et al. Cucurbitacin I induces
by Cucurbitacin B: a bioactive compound from cucumber, inhibits cancer cell death through the endoplasmic reticulum stress pathway.
prostate cancer growth. Cancer Lett. 2014;349(1):15–25. J Cell Biochem. 2018;10:758.
473. Shukla S, Sinha S, Khan S, Kumar S, Singh K, Mitra K, et al. Cucurbitacin B 494. Lu P, Yu B, Xu J. Cucurbitacin B regulates immature myeloid cell dif-
inhibits the stemness and metastatic abilities of NSCLC via downregula- ferentiation and enhances antitumor immunity in patients with lung
tion of canonical Wnt/beta-catenin signaling axis. Sci Rep. 2016;6:21860. cancer. Cancer Biother Radiopharm. 2012;27(8):495–503.
474. Zheng Q, Liu Y, Liu W, Ma F, Zhou Y, Chen M, et al. Cucurbitacin B 495. Hira SK, Mondal I, Manna PP. Combined immunotherapy with whole
inhibits growth and induces apoptosis through the JAK2/STAT3 and tumor lysate-pulsed interleukin-15-activated dendritic cells and
MAPK pathways in SHSY5Y human neuroblastoma cells. Mol Med Rep. cucurbitacin I promotes strong CD8(+) T-cell responses and cures
2014;10(1):89–94. highly aggressive lymphoma. Cytotherapy. 2015;17(5):647–64.
475. Zhou X, Yang J, Wang Y, Li W, Li-Ling J, Deng Y, et al. Cucurbitacin B 496. Ge W, Chen X, Han F, Liu Z, Wang T, Wang M, et al. Synthesis of
inhibits 12-O-tetradecanoylphorbol 13-acetate-induced invasion and Cucurbitacin B derivatives as potential anti-hepatocellular carcinoma
migration of human hepatoma cells through inactivating mitogen- agents. Molecules (Basel, Switzerland). 2018;23:12.
activated protein kinase and PI3K/Akt signal transduction pathways. 497. Tang L, Fu L, Zhu Z, Yang Y, Sun B, Shan W, et al. Modified mixed
Hepatol Res. 2012;42(4):401–11. nanomicelles with collagen peptides enhanced oral absorption
476. Zha Q-B, Zhang X-Y, Lin Q-R, Xu L-H, Zhao G-X, Pan H, et al. Cucurbita- of Cucurbitacin B: preparation and evaluation. Drug Delivery.
cin E induces autophagy via downregulating mTORC1 signaling and 2018;25(1):862–71.
upregulating AMPK activity. PLoS ONE. 2015;10(5):e0124355. 498. Lee DH, Thoennissen NH, Goff C, Iwanski GB, Forscher C, Doan NB,
477. Mao D, Liu AH, Wang ZP, Zhang XW, Lu H. Cucurbitacin B inhibits cell et al. Synergistic effect of low-dose cucurbitacin B and low-dose
proliferation and induces cell apoptosis in colorectal cancer by modu- methotrexate for treatment of human osteosarcoma. Cancer Lett (N
lating methylation status of BTG3. Neoplasma. 2019;66(4):593–602. Y, NY, U S). 2011;306(2):161–70.
478. Xie YL, Tao WH, Yang TX, Qiao JG. Anticancer effect of cucurbitacin B on 499. Sun Y, Zhang J, Zhou J, Huang Z, Hu H, Qiao M, et al. Synergistic
MKN-45 cells via inhibition of the JAK2/STAT3 signaling pathway. Exp effect of cucurbitacin B in combination with curcumin via enhancing
Ther Med. 2016;12(4):2709–15. apoptosis induction and reversing multidrug resistance in human
479. Zhang YZ, Wang CF, Zhang LF. Cucurbitacin D impedes gastric cancer hepatoma cells. Eur J Pharmacol. 2015;768:28–40.
cell survival via activation of the iNOS/NO and inhibition of the Akt 500. Garg S, Huifu H, Kaul SC, Wadhwa R, Garg S, Wadhwa R, et al.
signalling pathway. Oncol Rep. 2018;39(6):2595–603. Induction of senescence in cancer cells by a novel combination
480. Kong Y, Chen J, Zhou Z, Xia H, Qiu MH, Chen C. Cucurbitacin E induces of Cucurbitacin B and withanone: molecular mechanism and
cell cycle G2/M phase arrest and apoptosis in triple negative breast therapeutic potential. J Gerontol A Biol Sci Med Sci. 2019. https://doi.
cancer. PLoS ONE. 2014;9(7):e103760. org/10.1093/gerona/glz077.
481. Zhang T, Li Y, Park KA, Byun HS, Won M, Jeon J, et al. Cucurbitacin 501. Eyol E, Tanriverdi Z, Karakus F, Yilmaz K, Unuvar S. Synergistic anti-pro-
induces autophagy through mitochondrial ROS production which liferative effects of cucurbitacin i and irinotecan onhuman colorectal
counteracts to limit caspase-dependent apoptosis. Autophagy. cancer cell lines. Clin Exp Pharmacol. 2016;6(5):1000219.
2012;8(4):559–76. 502. Lee J, Sohn EJ, Yoon S, Won G, Kim CG, Jung JH, et al. Activation
482. Ma G, Luo W, Lu J, Ma DL, Leung CH, Wang Y, et al. Cucurbitacin E of JNK and IRE1 is critically involved in tanshinone I-induced p62
induces caspase-dependent apoptosis and protective autophagy dependent autophagy in malignant pleural mesothelioma cells:
mediated by ROS in lung cancer cells. Chem Biol Interact. 2016;253:1–9. implication of p62 UBA domain. Oncotarget. 2017;8(15):25032–45.
483. Yuan G, Yan SF, Xue H, Zhang P, Sun JT, Li G. Cucurbitacin I induces 503. Xu SW, Little PJ, Lan T, Huang Y, Le K, Wu XQ, et al. Tanshinone II-A
protective autophagy in glioblastoma in vitro and in vivo. J Biol Chem. attenuates and stabilizes atherosclerotic plaques in Apolipoprotein-E
2014;289(15):10607–19. knockout mice fed a high cholesterol diet. Arch Biochem Biophys.
484. Liang J, Zhang XL, Yuan JW, Zhang HR, Liu D, Hao J, et al. Cucurbitacin B 2011;515(1–2):72–9.
inhibits the migration and invasion of breast cancer cells by altering the 504. Gong Y, Li YL, Lu Y, Li LL, Abdolmaleky H, Blackburn GL, et al. Bioac-
biomechanical properties of cells. Phytother Res. 2019;33(3):618–30. tive tanshinones in Salvia Miltiorrhiza inhibit the growth of prostate
485. Sinha S, Khan S, Shukla S, Lakra AD, Kumar S, Das G, et al. Cucurbitacin cancer cells in vitro and in mice. Int J Cancer. 2011;129(5):1042–52.
B inhibits breast cancer metastasis and angiogenesis through VEGF- 505. Yin X, Yin Y, Cao FL, Chen YF, Peng Y, Hou WG, et al. Tanshinone IIA
mediated suppression of FAK/MMP-9 signaling axis. Int J Biochem Cell attenuates the inflammatory response and apoptosis after traumatic
Biol. 2016;77(Pt A):41–56. injury of the spinal cord in adult rats. Plos ONE. 2012;7(6):e38381.
486. Hsu PC, Tian B, Yang YL, Wang YC, Liu S, Urisman A, et al. Cucurbi-
tacin E inhibits the Yesassociated protein signaling pathway and
Luo et al. Chin Med (2019) 14:48 Page 47 of 58
506. Li ZM, Xu SW, Liu PQ. Salvia miltiorrhizaBurge (Danshen): a golden 526. Yen JH, Huang ST, Huang HS, Fong YC, Wu YY, Chiang JH, et al. HGK-
herbal medicine in cardiovascular therapeutics. Acta Pharmacologica sestrin 2 signaling-mediated autophagy contributes to antitumor
Sinica. 2018;39(5):802–24. efficacy of Tanshinone IIA in human osteosarcoma cells. Cell Death Dis.
507. Zhang RW, Liu ZG, Xie Y, Wang LX, Li MC, Sun X. In vitro inhibition of 2018;9(10):1003.
invasion and metastasis in colon cancer cells by TanIIA. Genetics Mol 527. Qiu Y, Li C, Wang Q, Zeng X, Ji P. Tanshinone IIA induces cell death via
Res Gmr. 2016;15(3):15039008. Beclin-1-dependent autophagy in oral squamous cell carcinoma SCC-9
508. Nicolin V, Fancellu G, Valentini R. Effect of tanshinone II on cell growth cell line. Cancer Med. 2018;7(2):397–407.
of breast cancer cell line type MCF-7 and MD-MB-231. Italian J Anat 528. Li X, Li Z, Li X, Liu B, Liu Z. Mechanisms of Tanshinone II a inhibits malig-
Embryol. 2014;119(1):38. nant melanoma development through blocking autophagy signal
509. Shin EA, Sohn EJ, Won G, Choi JU, Jeong M, Kim B, et al. Upregulation transduction in A375 cell. BMC Cancer. 2017;17(1):357.
of microRNA135a-3p and death receptor 5 plays a critical role in Tan- 529. Ding L, Wang S, Wang W, Lv P, Zhao D, Chen F, et al. Tanshinone IIA
shinone I sensitized prostate cancer cells to TRAIL induced apoptosis. affects autophagy and apoptosis of glioma cells by inhibiting phos-
Oncotarget. 2014;5(14):5624–36. phatidylinositol 3-Kinase/Akt/mammalian target of rapamycin signaling
510. Jing X, Xu Y, Cheng W, Guo S, Zou Y, He L. Tanshinone I induces apop- pathway. Pharmacology. 2017;99(3–4):188–95.
tosis and pro-survival autophagy in gastric cancers. Cancer Chemother 530. Li C, Han X, Hong Z, Wu J, Bao L. The interplay between autophagy and
Pharmacol. 2016;77(6):1171–81. apoptosis induced by tanshinone IIA in prostate cancer cells. Tumor
511. Yun SM, Jung JH, Jeong SJ, Sohn EJ, Kim B, Kim SH. Tanshinone IIA Biol. 2016;37(6):1–8.
induces autophagic cell death via activation of AMPK and ERK and 531. Huang SY, Chang SF, Liao KF, Chiu SC. Tanshinone IIA inhibits epithelial-
inhibition of mTOR and p70 S6K in KBM-5 leukemia cells. Phytother Res. mesenchymal transition in bladder cancer cells via modulation of
2014;28(3):458–64. STAT3-CCL2 signaling. Int J Mol Sci. 2017;18(8):1616.
512. Zhang K, Li J, Meng W, Xing H, Yang Y. Tanshinone IIA inhibits acute 532. Kim SA, Kang OH, Kwon DY. Cryptotanshinone induces cell cycle arrest
promyelocytic leukemia cell proliferation and induces their apoptosis and apoptosis of NSCLC cells through the PI3K/Akt/GSK-3beta pathway.
in vivo. Blood Cells Mol Dis. 2016;56(1):46–52. Int J Mol Sci. 2018;19(9):2739.
513. Wang L, Wu J, Lu J, Ma R, Sun D, Tang J. Regulation of the cell cycle and 533. Park IJ, Kim MJ, Park OJ, Choe W, Kang I, Kim SS, et al. Cryptotanshinone
PI3K/Akt/mTOR signaling pathway by tanshinone I in human breast induces ER stress-mediated apoptosis in HepG2 and MCF7 cells. Apop-
cancer cell lines. Mol Med Rep. 2015;11(2):931–9. tosis. 2012;17(3):248–57.
514. Jiao Y, Wang X, Li Y, Tang B. Tanshinone IIA suppresses gastric cancer cell 534. Li Q, He K, Tang S, Xu LF, Luo YC. Anti-tumor activity of tanshinone IIA
proliferation and migration by downregulation of FOXM1. Oncol Rep. in combined with cyclophosphamide against Lewis mice with lung
2017;37(3):1394. cancer. Asian Pac J Trop Med. 2016;9(11):1062–5.
515. Kim EO, Kang SE, Im CR, Lee JH, Ahn KS, Yang WM, et al. Tanshinone IIA 535. Liu S, Han Z, Trivett AL, Lin H, Hannifin S, Yang D, et al. Cryptotanshi-
induces TRAIL sensitization of human lung cancer cells through selec- none has curative dual anti-proliferative and immunotherapeutic
tive ER stress induction. Int J Oncol. 2016;48(5):2205–12. effects on mouse Lewis lung carcinoma. Cancer Immunol Immunother.
516. Guerram M, Jiang ZZ, Yousef BA, Hamdi AM, Hassan HM, Yuan ZQ, 2019;68(7):1059–71.
et al. The potential utility of acetyltanshinone IIA in the treatment of 536. Zhang J, Li Y, Fang X, Zhou D, Wang Y, Chen M. TPGS-g-PLGA/Pluronic
HER2-overexpressed breast cancer: induction of cancer cell death by F68 mixed micelles for tanshinone IIA delivery in cancer therapy. Int J
targeting apoptotic and metabolic signaling pathways. Oncotarget. Pharm. 2014;476(1–2):185–98.
2015;6(26):21865. 537. Li Z, Zhang Y, Zhang K, Wu Z, Feng N. Biotinylated-lipid bilayer coated
517. Yan MY, Chien SY, Kuo SJ, Chen DR, Su CC. Tanshinone IIA inhibits BT-20 mesoporous silica nanoparticles for improving the bioavailability and
human breast cancer cell proliferation through increasing caspase anti-leukaemia activity of Tanshinone IIA. Artif Cells Nanomed Biotech-
12, GADD153 and phospho-p38 protein expression. Int J Mol Med. nol. 2018;46(sup1):578–87.
2012;29(5):855–63. 538. Qiu S, Granet R, Mbakidi JP, Bregier F, Pouget C, Micallef L, et al. Delivery
518. Jian C, Dong-Yun S, Shan-Lin L, Liang Z. Tanshinone IIA induces growth of tanshinone IIA and alpha-mangostin from gold/PEI/cyclodextrin
inhibition and apoptosis in gastric cancer in vitro and in vivo. Oncol nanoparticle platform designed for prostate cancer chemotherapy.
Rep. 2012;27(2):523. Bioorg Med Chem Lett. 2016;26(10):2503–6.
519. Qin J, Shi H, Xu Y, Zhao F, Wang Q. Tanshinone IIA inhibits cervix carci- 539. Bai Y, Zhang L, Fang X, Yang Y. Tanshinone IIA enhances chemosensitiv-
noma stem cells migration and invasion via inhibiting YAP transcrip- ity of colon cancer cells by suppressing nuclear factor-kappaB. Exp Ther
tional activity. Biomed Pharmacother. 2018;105:758–65. Med. 2016;11(3):1085–9.
520. Chen L, Zheng SZ, Sun ZG, Wang AY, Huang CH, Punchard NA, et al. 540. Li K, Liu W, Zhao Q, Wu C, Fan C, Lai H, et al. Combination of tanshinone
Cryptotanshinone has diverse effects on cell cycle events in melanoma IIA and doxorubicin possesses synergism and attenuation effects
cell lines with different metastatic capacity. Cancer Chemother Pharma- on doxorubicin in the treatment of breast cancer. Phytother Res.
col. 2011;68(1):17–27. 2019;33(6):1658–69.
521. Wang Y, Li JX, Wang YQ, Miao ZH. Tanshinone I inhibits tumor angio- 541. Jung JH, Kwon TR, Jeong SJ, Kim EO, Sohn EJ, Yun M, et al. Apoptosis
genesis by reducing Stat3 phosphorylation at Tyr705 and hypoxia- induced by tanshinone IIA and cryptotanshinone is mediated by
induced HIF-1alpha accumulation in both endothelial and tumor cells. distinct JAK/STAT3/5 and SHP1/2 signaling in chronic myeloid leukemia
Oncotarget. 2015;6(18):16031–42. K562 cells. Evid Based Complement Altern Med. 2013;2013:805639.
522. Lee HP, Liu YC, Chen PC, Tai HC, Li TM, Fong YC, et al. Tanshinone IIA 542. Xu P, Ji L, Tian S, Li F. Clinical effects of tanshinone IIA sodium sulfonate
inhibits angiogenesis in human endothelial progenitor cells in vitro and combined with trimetazidine and levocarnitine in the treatment of
in vivo. Oncotarget. 2017;8(65):109217–27. AVMC and its effects on serum TNF-alpha, IL-18 and IL-35. Exp Ther
523. Sui H, Zhao J, Zhou L, Wen H, Deng W, Li C, et al. Tanshinone IIA inhibits Med. 2018;16(5):4070–4.
beta-catenin/VEGF-mediated angiogenesis by targeting TGF-beta1 543. Wang YY, Lv YF, Lu L, Cai L. Oridonin inhibits mTOR signaling and the
in normoxic and HIF-1alpha in hypoxic microenvironments in human growth of lung cancer tumors. Anticancer Drugs. 2014;25(10):1192–200.
colorectal cancer. Cancer Lett. 2017;403:86–97. 544. Zhang Y, Wang L, Zi Y, Zhang L, Guo Y, Huang Y. Oridonin effectively
524. Zhang L, Chen C, Duanmu J, Wu Y, Tao J, Yang A, et al. Cryptotanshi- reverses the drug resistance of cisplatin involving induction of cell
none inhibits the growth and invasion of colon cancer by suppressing apoptosis and inhibition of MMP expression in human acute myeloid
inflammation and tumor angiogenesis through modulating MMP/TIMP leukemia cells. Saudi J Biol Sci. 2017;24(3):678–86.
system, PI3K/Akt/mTOR signaling and HIF-1alpha nuclear translocation. 545. Huang HL, Weng HY, Wang LQ, Yu CH, Huang QJ, Zhao PP, et al.
Int Immunopharmacol. 2018;65:429–37. Triggering Fbw7-mediated proteasomal degradation of c-Myc by
525. Xu X, Wu L, Zhou X, Zhou N, Zhuang Q, Yang J, et al. Cryptotanshinone oridonin induces cell growth inhibition and apoptosis. Mol Cancer Ther.
inhibits VEGF-induced angiogenesis by targeting the VEGFR2 signaling 2012;11(5):1155–65.
pathway. Microvasc Res. 2017;111:25–31. 546. Wang H, Ye Y, Chu JH, Zhu GY, Fong WF, Yu ZL. Proteomic and functional
analyses reveal the potential involvement of endoplasmic reticulum
Luo et al. Chin Med (2019) 14:48 Page 48 of 58
stress and alpha-CP1 in the anticancer activities of oridonin in HepG2 568. Liu Y, Liu JH, Chai K, Tashiro S, Onodera S, Ikejima T. Inhibition of
cells. Integr Cancer Ther. 2011;10(2):160–7. c-Met promoted apoptosis, autophagy and loss of the mitochondrial
547. Lu Y, Sun Y, Zhu J, Yu L, Jiang X, Zhang J, et al. Oridonin exerts anticancer transmembrane potential in oridonin-induced A549 lung cancer
effect on osteosarcoma by activating PPAR-gamma and inhibiting Nrf2 cells. J Pharm Pharmacol. 2013;65(11):1622–42.
pathway. Cell Death Dis. 2018;9(1):15. 569. Zhang YH, Wu YL, Tashiro S, Onodera S, Ikejima T. Reactive oxygen
548. Zhao J, Zhang M, He P, Zhao J, Chen Y, Qi J, et al. Proteomic analysis of species contribute to oridonin-induced apoptosis and autophagy
oridonin-induced apoptosis in multiple myeloma cells. Mol Med Rep. in human cervical carcinoma HeLa cells. Acta Pharmacol Sin.
2017;15(4):1807–15. 2011;32(10):1266–75.
549. Gu Z, Wang X, Qi R, Wei L, Huo Y, Ma Y, et al. Oridonin induces apop- 570. Zeng R, Chen Y, Zhao S, Cui GH. Autophagy counteracts apoptosis
tosis in uveal melanoma cells by upregulation of Bim and down- in human multiple myeloma cells exposed to oridonin in vitro
regulation of Fatty Acid Synthase. Biochem Biophys Res Commun. via regulating intracellular ROS and SIRT1. Acta Pharmacol Sin.
2015;457(2):187–93. 2012;33(1):91–100.
550. Kang N, Cao SJ, Zhou Y, He H, Tashiro S, Onodera S, et al. Inhibition of 571. Yu Y, Fan SM, Song JK, Tashiro S, Onodera S, Ikejima T. Hydroxyl radi-
caspase-9 by oridonin, a diterpenoid isolated from Rabdosia rubescens, cal (·OH) played a pivotal role in oridonin-induced apoptosis and
augments apoptosis in human laryngeal cancer cells. Int J Oncol. autophagy in human epidermoid carcinoma A431 cells. Biol Pharm
2015;47(6):2045–56. Bull. 2012;35(12):2148–59.
551. Jiang JH, Pi J, Jin H, Cai JY. Oridonin-induced mitochondria-dependent 572. Cao S, Huang Y, Zhang Q, Lu F, Donkor PO, Zhu Y, et al. Molecular
apoptosis in esophageal cancer cells by inhibiting PI3K/AKT/mTOR and mechanisms of apoptosis and autophagy elicited by combined
Ras/Raf pathways. J Cell Biochem. 2019;120(3):3736–46. treatment with oridonin and cetuximab in laryngeal squamous cell
552. Yang J, Ren X, Zhang L, Li Y, Cheng B, Xia J. Oridonin inhibits oral carcinoma. Apoptosis. 2019;24(1–2):33–45.
cancer growth and PI3K/Akt signaling pathway. Biomed Pharmacother. 573. Sun Y, Jiang X, Lu Y, Zhu J, Yu L, Ma B, et al. Oridonin prevents epithe-
2018;100:226–32. lial–mesenchymal transition and TGF-beta1-induced epithelial–mes-
553. Ren CM, Li Y, Chen QZ, Zeng YH, Shao Y, Wu QX, et al. Oridonin inhibits enchymal transition by inhibiting TGF-beta1/Smad2/3 in osteosar-
the proliferation of human colon cancer cells by upregulating BMP7 to coma. Chem Biol Interact. 2018;296:57–64.
activate p38 MAPK. Oncol Rep. 2016;35(5):2691–8. 574. Zhu MIN, Hong DUN, Bao Y, Wang C, Pan W. Oridonin induces the
554. Xia S, Zhang X, Li C, Guan H. Oridonin inhibits breast cancer growth apoptosis of metastatic hepatocellular carcinoma cells via a mito-
and metastasis through blocking the Notch signaling. Saudi Pharm J. chondrial pathway. Oncol Lett. 2013;6(5):1502–6.
2017;25(4):638–43. 575. Gao S, Tan H, Zhu N, Gao H, Lv C, Gang J, et al. Oridonin induces
555. Gao SY, Li J, Qu XY, Zhu N, Ji YB. Downregulation of Cdk1 and cyclinB1 apoptosis through the mitochondrial pathway in human gastric
expression contributes to oridonin-induced cell cycle arrest at G2/M cancer SGC-7901 cells. Int J Oncol. 2016;48(6):2453–60.
phase and growth inhibition in SGC-7901 gastric cancer cells. Asian Pac 576. Wu QX, Yuan SX, Ren CM, Yu Y, Sun WJ, He BC, et al. Oridonin upregu-
J Cancer Prevent. 2014;15(15):6437–41. lates PTEN through activating p38 MAPK and inhibits proliferation in
556. Xu B, Shen W, Liu X, Zhang T, Ren J, Fan Y, et al. Oridonin inhibits BxPC-3 human colon cancer cells. Oncol Rep. 2016;35(6):3341–8.
cell growth through cell apoptosis. Acta Biochim Biophys Sin (Shang- 577. Liu RX, Ma Y, Hu XL, Ren WY, Liao YP, Wang H, et al. Anticancer effects
hai). 2015;47(3):164–73. of oridonin on colon cancer are mediated via BMP7/p38 MAPK/p53
557. Li J, Yang L, Wu H. Oridonin induced the apoptosis of PC-3 cells and its signaling. Int J Oncol. 2018;53(5):2091–101.
mechanism. J Central South Univ Med Sci. 2011;36(8):754–9. 578. Gao F-H, Liu F, Wei WEI, Liu L-B, Xu M-H, Guo Z-Y, et al. Oridonin
558. Zhang LD, Liu Z, Liu H, Ran DM, Guo JH, Jiang B, et al. Oridonin induces apoptosis and senescence by increasing hydrogen peroxide
enhances the anticancer activity of NVP-BEZ235 against neuro- and glutathione depletion in colorectal cancer cells. Int J Mol Med.
blastoma cells in vitro and in vivo through autophagy. Int J Oncol. 2012;29(4):649–55.
2016;49(2):657–65. 579. Jin H, Tan X, Liu X, Ding Y. Downregulation of AP-1 gene expression
559. Cai DT, Jin H, Xiong QX, Liu WG, Gao ZG, Gu GX, et al. ER stress and is an initial event in the oridonin-mediated inhibition of colorec-
ASK1-JNK activation contribute to oridonin-induced apoptosis and tal cancer: studies in vitro and in vivo. J Gastroenterol Hepatol.
growth inhibition in cultured human hepatoblastoma HuH-6 cells. Mol 2011;26(4):706–15.
Cell Biochem. 2013;379(1–2):161–9. 580. Wu QJ, Zheng XC, Wang T, Zhang TY. Effects of oridonin on immune
560. Yao Z, Xie F, Li M, Liang Z, Xu W, Yang J, et al. Oridonin induces cells, Th1/Th2 balance and the expression of BLys in the spleens of
autophagy via inhibition of glucose metabolism in p53-mutated colo- broiler chickens challenged with Salmonella pullorum. Res Vet Sci.
rectal cancer cells. Cell Death Dis. 2017;8(2):e2633. 2018;119:262–7.
561. Wang H, Zhu L, Feng X, Zhang H, Luo Q, Chen F. Oridonin induces G2/M 581. Chen RY, Xu B, Chen SF, Chen SS, Zhang T, Ren J, et al. Effect of
cell cycle arrest and apoptosis in human oral squamous cell carcinoma. oridonin-mediated hallmark changes on inflammatory pathways
Eur J Pharmacol. 2017;815:282–9. in human pancreatic cancer (BxPC-3) cells. World J Gastroenterol.
562. Tian L, Xie K, Sheng D, Wan X, Zhu G. Antiangiogenic effects of oridonin. 2014;20(40):14895–903.
BMC Complement Altern Med. 2017;17(1):192. 582. Wang SQ, Wang C, Wang JW, Yang DX, Wang R, Wang CJ, et al. Geri-
563. Li CY, Wang Q, Shen S, Wei XL, Li GX. Oridonin inhibits migration, inva- donin, a novel derivative of oridonin, inhibits proliferation of MGC 803
sion, adhesion and TGF-beta1-induced epithelial-mesenchymal transi- cells both in vitro and in vivo through elevating the intracellular ROS. J
tion of melanoma cells by inhibiting the activity of PI3K/Akt/GSK-3beta Pharm Pharmacol. 2017;69(2):213–21.
signaling pathway. Oncol Lett. 2018;15(1):1362–72. 583. Li Y, Wang Y, Wang S, Gao Y, Zhang X, Lu C. Oridonin phosphate-
564. Li C, Wang Q, Shen S, Wei X, Li G. Oridonin inhibits VEGF-A-associated induced autophagy effectively enhances cell apoptosis of human
angiogenesis and epithelial-mesenchymal transition of breast cancer breast cancer cells. Med Oncol. 2015;32(1):365.
in vitro and in vivo. Oncol Lett. 2018;16(2):2289–98. 584. Chen W, Zhou J, Wu K, Huang J, Ding Y, Yun EJ, et al. Targeting XBP1-
565. Liu X, Kang J, Wang H, Huang T. Mitochondrial ROS contribute to mediated beta-catenin expression associated with bladder cancer with
oridonin-induced HepG2 apoptosis through PARP activation. Oncol newly synthetic Oridonin analogues. Oncotarget. 2016;7(35):56842–54.
Lett. 2018;15(3):2881–8. 585. Zhou J, Yun EJ, Chen W, Ding Y, Wu K, Wang B, et al. Targeting
566. Lu J, Chen X, Qu S, Yao B, Xu Y, Wu J, et al. Oridonin induces G2/M 3-phosphoinositide-dependent protein kinase 1 associated with drug-
cell cycle arrest and apoptosis via the PI3K/Akt signaling path- resistant renal cell carcinoma using new oridonin analogs. Cell Death
way in hormone-independent prostate cancer cells. Oncol Lett. Dis. 2017;8(3):e2701.
2017;13(4):2838–46. 586. Zhu L, Li M, Liu X, Du L, Jin Y. Inhalable oridonin-loaded poly(lactic-co-
567. Zhang T, Tan Y, Zhao R, Liu Z. DNA damage induced by oridonin glycolic)acid large porous microparticles for in situ treatment of primary
involves cell cycle arrest at G2/M phase in human MCF-7 cells. Contem- non-small cell lung cancer. Acta Pharm Sinica B. 2017;7(1):80–90.
porary Oncol (Poznan, Poland). 2013;17(1):38–44.
Luo et al. Chin Med (2019) 14:48 Page 49 of 58
587. Wang Y, Liu X, Liu G, Guo H, Li C, Zhang Y, et al. Novel galactosylated via ROS-mediated ER stress and p38 pathways. Artif Cells Nanomed
biodegradable nanoparticles for hepatocyte-delivery of oridonin. Int J Biotechnol. 2019;47(1):626–35.
Pharm. 2016;502(1–2):47–60. 609. Shi S, Cao H. Shikonin promotes autophagy in BXPC-3 human pancre-
588. Yu Y, Fan SM, Ye YC, Tashiro S, Onodera S, Ikejima T. The tyrphostin atic cancer cells through the PI3K/Akt signaling pathway. Oncol Lett.
AG1478 augments oridonin-induced A431 cell apoptosis by blockage 2014;8(3):1087–9.
of JNK MAPK and enhancement of oxidative stress. Free Radic Res. 610. Gong K, Zhang Z, Chen Y, Shu HB, Li W. Extracellular signal-regulated
2012;46(11):1393–405. kinase, receptor interacting protein, and reactive oxygen species regu-
589. Tiwari RV, Parajuli P, Sylvester PW. Synergistic anticancer effects of com- late shikonin-induced autophagy in human hepatocellular carcinoma.
bined gamma-tocotrienol and oridonin treatment is associated with Eur J Pharmacol. 2014;738:142–52.
the induction of autophagy. Mol Cell Biochem. 2015;408(1–2):123–37. 611. Wang H, Wu C, Wan S, Zhang H, Zhou S, Liu G. Shikonin attenuates lung
590. Ding Y, Ding C, Ye N, Liu Z, Wold EA, Chen H, et al. Discovery and devel- cancer cell adhesion to extracellular matrix and metastasis by inhibiting
opment of natural product oridonin-inspired anticancer agents. Eur J integrin beta1 expression and the ERK1/2 signaling pathway. Toxicol-
Med Chem. 2016;122:102–17. ogy. 2013;308:104–12.
591. Chen CH, Lin ML, Ong PL, Yang JT. Novel multiple apoptotic 612. Huang CJ, Luo YA, Zhao JW, Yang FW, Zhao HW, Fan WH, et al. Shikonin
mechanism of shikonin in human glioma cells. Ann Surg Oncol. kills glioma cells through necroptosis mediated by RIP-1. PloS ONE.
2012;19(9):3097–106. 2013;8(6):e66326.
592. Lu L, Qin A, Huang H, Zhou P, Zhang C, Liu N, et al. Shikonin extracted 613. Yang JT, Li ZL, Wu JY, Lu FJ, Chen CH. An oxidative stress mechanism of
from medicinal Chinese herbs exerts anti-inflammatory effect via shikonin in human glioma cells. PloS ONE. 2014;9(4):e94180.
proteasome inhibition. Eur J Pharmacol. 2011;658(2–3):242–7. 614. Liu J, Wang P, Xue Y-X, Li Z, Qu C-B, Liu Y-H. Enhanced antitumor effect
593. Kim HJ, Hwang KE, Park DS, Oh SH, Jun HY, Yoon KH, et al. Shikonin- of shikonin by inhibiting endoplasmic reticulum stress via JNK/c-Jun
induced necroptosis is enhanced by the inhibition of autophagy in pathway in human glioblastoma stem cells. Biochem Biophys Res Com-
non-small cell lung cancer cells. J Transl Med. 2017;15(1):123. mun. 2015;466(1):103–10.
594. Zuo AR, Dong HH, Yu YY, Shu QL, Zheng LX, Yu XY, et al. The antityrosi- 615. Wang H, Liu Z, Li X, Zhao R, Pu Y, Wu H, et al. Shikonin causes apoptosis
nase and antioxidant activities of flavonoids dominated by the number by disrupting intracellular calcium homeostasis and mitochondrial
and location of phenolic hydroxyl groups. Chin Med. 2018;13:51. function in human hepatoma cells. Exp Ther Med. 2018;15(2):1484–92.
595. Jang SY, Lee JK, Jang EH, Jeong SY, Kim JH. Shikonin blocks migration 616. Liang W, Cui J, Zhang K, Xi H, Cai A, Li J, et al. Shikonin induces ROS-
and invasion of human breast cancer cells through inhibition of matrix based mitochondria-mediated apoptosis in colon cancer. Oncotarget.
metalloproteinase-9 activation. Oncol Rep. 2014;31(6):2827–33. 2017;8(65):109094–106.
596. Chen CM, Shanmugasundaram K, Rigby AC, Kung AL. Shikonin, a natu- 617. Qu D, Chen YU, Xu XM, Zhang M, Zhang YI, Li SQ. Cbl-b-regulated
ral product from the root of Lithospermum erythrorhizon, is a cytotoxic extracellular signal-regulated kinase signaling is involved in the
DNA-binding agent. Eur J Pharm Sci. 2013;49(1):18–26. shikonin-induced apoptosis of lung cancer cells in vitro. Exp Ther Med.
597. Chen Y, Zheng L, Liu J, Zhou Z, Cao X, Lv X, et al. Shikonin inhibits pros- 2015;9(4):1265–70.
tate cancer cells metastasis by reducing matrix metalloproteinase-2/-9 618. Zhao QL, Assimopoulou AN, Klauck SM, Damianakos H, Chinou I,
expression via AKT/mTOR and ROS/ERK1/2 pathways. Int Immunophar- Kretschmer N, et al. Inhibition of c-MYC with involvement of ERK/JNK/
macol. 2014;21(2):447–55. MAPK and AKT pathways as a novel mechanism for shikonin and its
598. Hao ZF, Qian J, Yang JS. Shikonin induces apoptosis and inhibits migra- derivatives in killing leukemia cells. Oncotarget. 2015;6(36):38934–51.
tion of ovarian carcinoma cells by inhibiting the phosphorylation of Src 619. Chen Y, Wang TT, Du J, Li YC, Wang X, Zhou Y, et al. The critical role
and FAK. Oncol Lett. 2015;9(2):629–33. of PTEN/PI3K/AKT signaling pathway in shikonin-induced apoptosis
599. Ni F, Huang X, Chen Z, Qian W, Tong X. Shikonin exerts antitumor and proliferation inhibition of chronic myeloid leukemia. Cell Physiol
activity in Burkitt’s lymphoma by inhibiting C-MYC and PI3K/AKT/ Biochem. 2018;47(3):981–93.
mTOR pathway and acts synergistically with doxorubicin. Sci Rep. 620. Shan ZL, Zhong L, Xiao CL, Gan LG, Xu T, Song H, et al. Shikonin
2018;8(1):3317. suppresses proliferation and induces apoptosis in human leukemia
600. Yang Q, Ji M, Guan H, Shi B, Hou P. Shikonin inhibits thyroid cancer cell NB4 cells through modulation of MAPKs and cMyc. Mol Med Rep.
growth and invasiveness through targeting major signaling pathways. J 2017;16(3):3055–60.
Clin Endocrinol Metab. 2013;98(12):E1909–17. 621. Zhou ZJ, Lu B, Wang C, Wang ZQ, Luo TF, Piao MH, et al. RIP1 and RIP3
601. Lan WJ, Wan SB, Gu WQ, Wang HY, Zhou SW. Mechanisms behind the contribute to shikonin-induced DNA double-strand breaks in glioma
inhibition of lung adenocarcinoma cell by shikonin. Cell Biochem cells via increase of intracellular reactive oxygen species. Cancer Lett.
Biophys. 2014;70(2):1459–67. 2017;390:77–90.
602. Hsieh YS, Liao CH, Chen WS, Pai JT, Weng MS. Shikonin inhibited 622. Lu B, Wang ZQ, Ding Y, Wang XZ, Lu S, Wang CC, et al. RIP1 and RIP3
migration and invasion of human lung cancer cells via suppression of contribute to shikonin-induced glycolysis suppression in glioma
c-Met-mediated epithelial-to-mesenchymal transition. J Cell Biochem. cells via increase of intracellular hydrogen peroxide. Cancer Lett.
2017;118(12):4639–51. 2018;425:31–42.
603. Zhai T, Hei Z, Ma Q, Liang H, Xu Y, Zhang Y, et al. Shikonin induces 623. Li Y, Lu H, Gu Y, Ning Z, Cao T, Chen C, et al. Enhancement of NK cells
apoptosis and G0/G1 phase arrest of gallbladder cancer cells via the proliferation and function by Shikonin. Immunopharmacol Immuno-
JNK signaling pathway. Oncol Rep. 2017;38(6):3473–80. toxicol. 2017;39(3):124–30.
604. Tang JC, Zhao J, Long F, Chen JY, Mu B, Jiang Z, et al. Efficacy of Shikonin 624. Yin SY, Efferth T, Jian FY, Chen YH, Liu CI, Wang AH, et al. Immunogenic-
against esophageal cancer cells and its possible mechanisms in vitro ity of mammary tumor cells can be induced by shikonin via direct
and in vivo. J Cancer. 2018;9(1):32–40. binding-interference with hnRNPA1. Oncotarget. 2016;7(28):43629–53.
605. Tian R, Li Y, Gao M. Shikonin causes cell-cycle arrest and induces 625. Chen HM, Wang PH, Aravindaram K, Chen YH, Yu HH, Yang WC, et al.
apoptosis by regulating the EGFR–NF-κB signalling pathway in human Shikonin enhances efficacy of a gene-based cancer vaccine via induc-
epidermoid carcinoma A431 cells. Biosci Rep. 2015;35(2):1–13. tion of RANTES. J Biomed Sci. 2012;19:42.
606. Liu T, Sun X, Cao Z. Shikonin-induced necroptosis in nasopharyngeal 626. Yang YY, He HQ, Cui JH, Nie YJ, Wu YX, Wang R, et al. Shikonin derivative
carcinoma cells via ROS overproduction and upregulation of RIPK1/ DMAKO-05 inhibits Akt signal activation and melanoma proliferation.
RIPK3/MLKL expression. OncoTargets Ther. 2019;12:2605–14. Chem Biol Drug Des. 2016;87(6):895–904.
607. Lu B, Gong X, Wang ZQ, Ding Y, Wang C, Luo TF, et al. Shikonin 627. Zhang X, Wang RB, Zhou W, Xiao S, Meng QQ, Li SS. Antitumor activity
induces glioma cell necroptosis in vitro by ROS overproduction and of DMAKO-05, a novel shikonin derivative, and its metabolism in rat
promoting RIP1/RIP3 necrosome formation. Acta Pharmacol Sin. liver microsome. AAPS PharmSciTech. 2015;16(2):259–66.
2017;38(11):1543–53. 628. Durchschein C, Hufner A, Rinner B, Stallinger A, Deutsch A, Lohberger
608. Liu Y, Kang X, Niu G, He S, Zhang T, Bai Y, et al. Shikonin induces B, et al. Synthesis of novel shikonin derivatives and pharmacological
apoptosis and prosurvival autophagy in human melanoma A375 cells effects of cyclopropylacetylshikonin on melanoma cells. Molecules
(Basel, Switzerland). 2018;23:11.
Luo et al. Chin Med (2019) 14:48 Page 50 of 58
629. Li S, Zhang T, Xu W, Ding J, Yin F, Xu J, et al. Sarcoma-targeting peptide- P-glycoprotein and suppressing survivin expression. Chem Biol Interact.
decorated polypeptide nanogel intracellularly delivers shikonin for 2015;235:76–84.
upregulated osteosarcoma necroptosis and diminished pulmonary 650. Wang S, Xu Y, Li C, Tao H, Wang A, Sun C, et al. Gambogic acid sensitizes
metastasis. Theranostics. 2018;8(5):1361–75. breast cancer cells to TRAIL-induced apoptosis by promoting the cross-
630. Wen X, Li J, Cai D, Yue L, Wang Q, Zhou L, et al. Anticancer efficacy of talk of extrinsic and intrinsic apoptotic signalings. Food Chem Toxicol.
targeted shikonin liposomes modified with RGD in breast cancer cells. 2018;119:334–41.
Molecules (Basel, Switzerland). 2018;23:2. 651. Li C, Qi Q, Lu N, Dai Q, Li F, Wang X, et al. Gambogic acid promotes
631. Li W, Liu J, Jackson K, Shi R, Zhao Y. Sensitizing the therapeutic efficacy apoptosis and resistance to metastatic potential in MDA-MB-231
of taxol with shikonin in human breast cancer cells. PLoS ONE. human breast carcinoma cells. Biochem Cell Biol. 2012;90(6):718–30.
2014;9(4):e94079. 652. Ishaq M, Khan MA, Sharma K, Sharma G, Dutta RK, Majumdar S.
632. Wang Y, Zhou Y, Jia G, Han B, Liu J, Teng Y, et al. Shikonin suppresses Gambogic acid induced oxidative stress dependent caspase activation
tumor growth and synergizes with gemcitabine in a pancreatic cancer regulates both apoptosis and autophagy by targeting various key mol-
xenograft model: involvement of NF-kappaB signaling pathway. Bio- ecules (NF-kappaB, Beclin-1, p62 and NBR1) in human bladder cancer
chem Pharmacol. 2014;88(3):322–33. cells. Biochim Biophys Acta. 1840;12:3374–84. https://doi.org/10.1016/j.
633. Chen C, Xiao W, Huang L, Yu G, Ni J, Yang L, et al. Shikonin induces bbagen.2014.08.019.
apoptosis and necroptosis in pancreatic cancer via regulating the 653. Wang H, Zhao Z, Lei S, Li S, Xiang Z, Wang X, et al. Gambogic acid
expression of RIP1/RIP3 and synergizes the activity of gemcitabine. Am induces autophagy and combines synergistically with chloroquine to
J Transl Res. 2017;9(12):5507–17. suppress pancreatic cancer by increasing the accumulation of reactive
634. Liu X, Sun G. Shikonin enhances Adriamycin antitumor effects by inhib- oxygen species. Cancer Cell Int. 2019;19:7.
iting efflux pumps in A549 cells. Oncol Lett. 2017;14(4):4270–6. 654. Ye L, Zhou J, Zhao W, Jiao P, Ren G, Wang S. Gambogic acid-
635. Yang Q, Li S, Fu Z, Lin B, Zhou Z, Wang Z, et al. Shikonin promotes induced autophagy in nonsmall cell lung cancer NCI-H441 cells
adriamycininduced apoptosis by upregulating caspase3 and caspase8 through a reactive oxygen species pathway. J Cancer Res Ther.
in osteosarcoma. Mol Med Rep. 2017;16(2):1347–52. 2018;14(Supplement):S942–7.
636. He G, He G, Zhou R, Pi Z, Zhu T, Jiang L, et al. Enhancement of 655. Qi Q, Lu N, Li C, Zhao J, Liu W, You Q, et al. Involvement of RECK in gam-
cisplatin-induced colon cancer cells apoptosis by shikonin, a natural bogic acid induced anti-invasive effect in A549 human lung carcinoma
inducer of ROS in vitro and in vivo. Biochem Biophys Res Commun. cells. Mol Carcinog. 2015;54(Suppl 1):E13–25. https://doi.org/10.1002/
2016;469(4):1075–82. mc.22138.
637. Wang X, Chen W. Gambogic acid is a novel anti-cancer agent that 656. Lu L, Tang D, Wang L, Huang LQ, Jiang GS, Xiao XY, et al. Gambogic acid
inhibits cell proliferation, angiogenesis and metastasis. Anticancer inhibits TNF-alpha-induced invasion of human prostate cancer PC3
Agents Med Chem. 2012;12(8):994–1000. cells in vitro through PI3K/Akt and NF-kappaB signaling pathways. Acta
638. Cascao R, Vidal B, Raquel H, Neves-Costa A, Figueiredo N, Gupta V, et al. Pharmacol Sin. 2012;33(4):531–41.
Potent anti-inflammatory and antiproliferative effects of gambogic 657. Wan L, Zhang Q, Wang S, Gao Y, Chen X, Zhao Y, et al. Gambogic acid
acid in a rat model of antigen-induced arthritis. Mediators Inflamm. impairs tumor angiogenesis by targeting YAP/STAT3 signaling axis.
2014;2014:195327. Phytother Res. 2019;33(5):1579–91.
639. Liu L, Qi XJ, Zhong ZK, Zhang EN. Nanomedicine-based combination of 658. Lyu L, Huang LQ, Huang T, Xiang W, Yuan JD, Zhang CH. Cell-penetrat-
gambogic acid and retinoic acid chlorochalcone for enhanced antican- ing peptide conjugates of gambogic acid enhance the antitumor effect
cer efficacy in osteosarcoma. Biomed Pharmacother. 2016;83:79–84. on human bladder cancer EJ cells through ROS-mediated apoptosis.
640. Zou Z, Xie L, Wei J, Yu L, Qian X, Chen J, et al. Synergistic anti-prolifera- Drug Design Develop Ther. 2018;12:743–56.
tive effects of gambogic acid with docetaxel in gastrointestinal cancer 659. Zhu M, Jiang Y, Wu H, Shi W, Lu G, Cong D, et al. Gambogic acid shows
cell lines. BMC Complement Altern Med. 2012;12:58. anti-proliferative effects on non-small cell lung cancer (NSCLC) cells by
641. Shi X, Chen X, Li X, Lan X, Zhao C, Liu S, et al. Gambogic acid induces activating reactive oxygen species (ROS)-induced endoplasmic reticu-
apoptosis in imatinib-resistant chronic myeloid leukemia cells via lum (ER) stress-mediated apoptosis. Med Sci Moni. 2019;25:3983–8.
inducing proteasome inhibition and caspase-dependent Bcr-Abl 660. Lee JY, Lee BH, Lee JY. Gambogic acid disrupts toll-like receptor4 activa-
downregulation. Clin Cancer Res. 2014;20(1):151–63. tion by blocking lipopolysaccharides binding to myeloid differentiation
642. Liang L, Zhang Z. Gambogic acid inhibits malignant melanoma cell factor 2. Toxicol Res. 2015;31(1):11–6.
proliferation through mitochondrial p66shc/ROS-p53/Bax-mediated 661. Ma J, Huang K, Ma Y, Chen S, Liu C, Shan Z, et al. Gambogic acid inhibits
apoptosis. Cell Physiol Biochem. 2016;38(4):1618–30. LPS-induced macrophage pro-inflammatory cytokine production
643. Pandey MK, Kale VP, Song C, Sung SS, Sharma AK, Talamo G, et al. mainly through suppression of the p38 pathway. Iran J Basic Med Sci.
Gambogic acid inhibits multiple myeloma mediated osteoclastogen- 2018;21(7):717–23.
esis through suppression of chemokine receptor CXCR651 signaling 662. Liu F, Huang X, Han L, Sang M, Hu L, Liu B, et al. Improved druggabil-
pathways. Exp Hematol. 2014;42(10):883–96. ity of gambogic acid using core-shell nanoparticles. Biomater Sci.
644. Jang JH, Kim JY, Sung EG, Kim EA, Lee TJ. Gambogic acid induces apop- 2019;7(3):1028–42.
tosis and sensitizes TRAIL-mediated apoptosis through downregulation 663. Zhang W, Zhou H, Yu Y, Li J, Li H, Jiang D, et al. Combination of
of cFLIPL in renal carcinoma Caki cells. Int J Oncol. 2016;48(1):376–84. gambogic acid with cisplatin enhances the antitumor effects on
645. Lu N, Hui H, Yang H, Zhao K, Chen Y, You QD, et al. Gambogic acid inhib- cisplatin-resistant lung cancer cells by downregulating MRP2 and LRP
its angiogenesis through inhibiting PHD2-VHL-HIF-1alpha pathway. Eur expression. OncoTargets Ther. 2016;9:3359–68.
J Pharm Sci. 2013;49(2):220–6. 664. Xia G, Wang H, Song Z, Meng Q, Huang X, Huang X. Gambogic acid
646. Pan H, Jansson KH, Beshiri ML, Yin J, Fang L, Agarwal S, et al. Gam- sensitizes gemcitabine efficacy in pancreatic cancer by reducing the
bogic acid inhibits thioredoxin activity and induces ROS-mediated expression of ribonucleotide reductase subunit-M2 (RRM2). J Exp Clin
cell death in castration-resistant prostate cancer. Oncotarget. Cancer Res. 2017;36(1):107.
2017;8(44):77181–94. 665. Wang J, Yuan Z. Gambogic acid sensitizes ovarian cancer cells to
647. Zhen YZ, Lin YJ, Li KJ, Yang XS, Zhao YF, Wei J, et al. Gambogic acid doxorubicin through ROS-mediated apoptosis. Cell Biochem Biophys.
lysinate induces apoptosis in breast cancer mcf-7 cells by increas- 2013;67(1):199–206.
ing reactive oxygen species. Evid Based Complement Altern Med. 666. Chi Y, Zhan XK, Yu H, Xie GR, Wang ZZ, Xiao W, et al. An open-labeled,
2015;2015:842091. randomized, multicenter phase IIa study of gambogic acid injection for
648. Li D, Song XY, Yue QX, Cui YJ, Liu M, Feng LX, et al. Proteomic and advanced malignant tumors. Chin Med J. 2013;126(9):1642–6.
bioinformatic analyses of possible target-related proteins of gambogic 667. Augustin Y, Krishna S, Kumar D, Pantziarka PJE. The wisdom of crowds
acid in human breast carcinoma MDA-MB-231 cells. Chin J Nat Med. and the repurposing of artesunate as an anticancer drug. Ecancermedi-
2015;13(1):41–51. calscience. 2015;9:ed50.
649. Wang S, Wang L, Chen M, Wang Y. Gambogic acid sensitizes 668. Ho WE, Peh HY, Chan TK, Wong WS. Artemisinins: pharmacological
resistant breast cancer cells to doxorubicin through inhibiting actions beyond anti-malarial. Pharmacol Ther. 2014;142(1):126–39.
Luo et al. Chin Med (2019) 14:48 Page 51 of 58
669. Zuo W, Wang ZZ, Xue J. Artesunate induces apoptosis of bladder 690. Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance
cancer cells by miR-16 regulation of COX-2 expression. Int J Mol Sci. of cisplatin-resistant head and neck cancer cells to artesunate-induced
2014;15(8):14298–312. ferroptosis. Redox Biol. 2017;11:254–62.
670. Greenshields AL, Fernando W, Hoskin DW. The anti-malarial drug 691. Cui C, Feng H, Shi X, Wang Y, Feng Z, Liu J, et al. Artesunate down-
artesunate causes cell cycle arrest and apoptosis of triple-negative regulates immunosuppression from colorectal cancer Colon26 and
MDA-MB-468 and HER2-enriched SK-BR-3 breast cancer cells. Exp RKO cells in vitro by decreasing transforming growth factor beta1 and
Mol Pathol. 2019;107:10–22. interleukin-10. Int Immunopharmacol. 2015;27(1):110–21.
671. Zhang LX, Liu ZN, Ye J, Sha M, Qian H, Bu XH, et al. Artesunate 692. Qian P, Zhang YW, Zhou ZH, Liu JQ, Yue SY, Guo XL, et al. Artesunate
exerts an anti-immunosuppressive effect on cervical cancer by enhances gammadelta T-cell-mediated antitumor activity through
inhibiting PGE2 production and Foxp3 expression. Cell Biol Int. augmenting gammadelta T-cell function and reversing immune escape
2014;38(5):639–46. of HepG2 cells. Immunopharmacol Immunotoxicol. 2018;40(2):107–16.
672. Liu L, Zuo LF, Zuo J, Wang J. Artesunate induces apoptosis and inhib- 693. Wang B, Hou D, Liu Q, Wu T, Guo H, Zhang X, et al. Artesunate sensitizes
its growth of Eca109 and Ec9706 human esophageal cancer cell lines ovarian cancer cells to cisplatin by downregulating RAD51. Cancer Biol
in vitro and in vivo. Mol Med Rep. 2015;12(1):1465–72. Ther. 2015;16(10):1548–56.
673. Wang L, Liu L, Wang J, Chen Y. Inhibitory effect of artesunate 694. Efferth T, Ramirez T, Gebhart E, Halatsch ME. Combination treatment of
on growth and apoptosis of gastric cancer cells. Arch Med Res. glioblastoma multiforme cell lines with the anti-malarial artesunate and
2017;48(7):623–30. the epidermal growth factor receptor tyrosine kinase inhibitor OSI-774.
674. Chen X, Zhang XL, Zhang GH, Gao YF. Artesunate promotes Th1 dif- Biochem Pharmacol. 2004;67(9):1689–700.
ferentiation from CD4 + T cells to enhance cell apoptosis in ovarian 695. Tran BN, Nguyen HT, Kim JO, Yong CS, Nguyen CN. Developing combi-
cancer via miR-142. Braz J Med Biol Res. 2019;52(5):e7992. nation of artesunate with paclitaxel loaded into poly-d, l-lactic-co-gly-
675. Zhou Y, Wang X, Zhang J, He A, Wang YL, Han K, et al. Artesunate colic acid nanoparticle for systemic delivery to exhibit synergic chemo-
suppresses the viability and mobility of prostate cancer cells through therapeutic response. Drug Dev Ind Pharm. 2017;43(12):1952–62.
UCA1, the sponge of miR-184. Oncotarget. 2017;8(11):18260–70. 696. Wang X, Du Q, Mao Z, Fan X, Hu B, Wang Z, et al. Combined treatment
676. Jeong DE, Song HJ, Lim S, Lee SJ, Lim JE, Nam DH, et al. Repurposing with artesunate and bromocriptine has synergistic anticancer effects in
the anti-malarial drug artesunate as a novel therapeutic agent for pituitary adenoma cell lines. Oncotarget. 2017;8(28):45874–87.
metastatic renal cell carcinoma due to its attenuation of tumor growth, 697. Osaki T, Uto Y, Ishizuka M, Tanaka T, Yamanaka N, Kurahashi T, et al.
metastasis, and angiogenesis. Oncotarget. 2015;6(32):33046–64. Artesunate enhances the cytotoxicity of 5-aminolevulinic acid-based
677. Drenberg CD, Buaboonnam J, Orwick SJ, Hu S, Li L, Fan Y, et al. Evalu- sonodynamic therapy against mouse mammary tumor cells in vitro.
ation of artemisinins for the treatment of acute myeloid leukemia. Molecules (Basel, Switzerland). 2017;22:4.
Cancer Chemother Pharmacol. 2016;77(6):1231–43. 698. Deeken JF, Wang H, Hartley M, Cheema AK, Smaglo B, Hwang JJ, et al. A
678. Zheng L, Pan J. The anti-malarial drug artesunate blocks Wnt/beta- phase I study of intravenous artesunate in patients with advanced solid
catenin pathway and inhibits growth, migration and invasion of uveal tumor malignancies. Cancer Chemother Pharmacol. 2018;81(3):587–96.
melanoma cells. Curr Cancer Drug Targets. 2018;18(10):988–98. 699. Krishna S, Ganapathi S, Ster IC, Saeed ME, Cowan M, Finlayson C, et al.
679. Chen H, Shi L, Yang X, Li S, Guo X, Pan L. Artesunate inhibiting angio- A randomised, double blind, placebo-controlled pilot study of oral
genesis induced by human myeloma RPMI8226 cells. Int J Hematol. artesunate therapy for colorectal cancer. EBioMedicine. 2015;2(1):82–90.
2010;92(4):587–97. 700. von Hagens C, Walter-Sack I, Goeckenjan M, Storch-Hagenlocher B,
680. Krusche B, Arend J, Efferth T. Synergistic inhibition of angiogenesis by Sertel S, Elsasser M, et al. Long-term add-on therapy (compassionate
artesunate and captopril in vitro and in vivo. Evid Based Complement use) with oral artesunate in patients with metastatic breast cancer
Altern Med. 2013;2013:454783. after participating in a phase I study (ARTIC M33/2). Phytomedicine.
681. Dong HY, Wang ZF. Antitumor effects of artesunate on human breast 2019;54:140–8.
carcinoma MCF-7 cells and IGF-IR expression in nude mice xenografts. 701. von Hagens C, Walter-Sack I, Goeckenjan M, Osburg J, Storch-Hagen-
Chin J Cancer Res. 2014;26(2):200–7. locher B, Sertel S, et al. Prospective open uncontrolled phase I study
682. Chen X, Wong YK, Lim TK, Lim WH, Lin Q, Wang J, et al. Artesunate acti- to define a well-tolerated dose of oral artesunate as add-on therapy in
vates the intrinsic apoptosis of HCT116 cells through the suppression patients with metastatic breast cancer (ARTIC M33/2). Breast Cancer Res
of fatty acid synthesis and the NF-kappaB pathway. Molecules (Basel, Treat. 2017;164(2):359–69.
Switzerland). 2017;22:8. 702. Seong RK, Kim JA, Shin OS. Wogonin, a flavonoid isolated from Scutel-
683. Zhang P, Luo HS, Li M, Tan SY. Artesunate inhibits the growth and laria baicalensis, has anti-viral activities against influenza infection via
induces apoptosis of human gastric cancer cells by downregulating modulation of AMPK pathways. Acta Virol. 2018;62(1):78–85.
COX-2. OncoTargets Ther. 2015;8:845–54. 703. Khan S, Zhang D, Zhang Y, Li M, Wang C. Wogonin attenuates diabetic
684. Greenshields AL, Shepherd TG, Hoskin DW. Contribution of reactive cardiomyopathy through its anti-inflammatory and anti-oxidative
oxygen species to ovarian cancer cell growth arrest and killing by the properties. Mol Cell Endocrinol. 2016;428:101–8.
anti-malarial drug artesunate. Mol Carcinog. 2017;56(1):75–93. 704. Ge W, Yin Q, Xian H. Wogonin induced mitochondrial dysfunction and
685. Chen K, Shou LM, Lin F, Duan WM, Wu MY, Xie X, et al. Artesunate endoplasmic reticulum stress in human malignant neuroblastoma cells
induces G2/M cell cycle arrest through autophagy induction in breast via ire1alpha-dependent pathway. J Mol Neurosci. 2015;56(3):652–62.
cancer cells. Anticancer Drugs. 2014;25(6):652–62. 705. Xu Y, Yang B, Hu Y, Lu L, Lu X, Wang J, et al. Wogonin prevents TLR4-
686. Zhang J, Sun X, Wang L, Wong YK, Lee YM, Zhou C, et al. Artesu- NF-kappaB-medicated neuro-inflammation and improves retinal
nate-induced mitophagy alters cellular redox status. Redox Biol. ganglion cells survival in retina after optic nerve crush. Oncotarget.
2018;19:263–73. 2016;7(45):72503–17.
687. Zhang L, Qian H, Sha M, Luan Z, Lin M, Yuan D, et al. Downregulation 706. Chen XM, Bai Y, Zhong YJ, Xie XL, Long HW, Yang YY, et al. Wogonin
of HOTAIR expression mediated anti-metastatic effect of artesu- has multiple anti-cancer effects by regulating c-Myc/SKP2/Fbw7alpha
nate on cervical cancer by inhibiting COX-2 expression. PLoS ONE. and HDAC1/HDAC2 pathways and inducing apoptosis in human lung
2016;11(10):e0164838. adenocarcinoma cell line A549. PLoS ONE. 2013;8(11):e79201.
688. Beccafico S, Morozzi G, Marchetti MC, Riccardi C, Sidoni A, Donato R, 707. Yao Y, Zhao K, Yu Z, Ren H, Zhao L, Li Z, et al. Wogonoside inhibits inva-
et al. Artesunate induces ROS- and p38 MAPK-mediated apoptosis and sion and migration through suppressing TRAF2/4 expression in breast
counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cancer. J Exp Clin Cancer Res: CR. 2017;36(1):103.
cells. Carcinogenesis. 2015;36(9):1071–83. 708. Kim EH, Jang H, Shin D, Baek SH, Roh JL. Targeting Nrf2 with wogonin
689. Vatsveen TK, Myhre MR, Steen CB, Walchli S, Lingjaerde OC, Bai B, et al. overcomes cisplatin resistance in head and neck cancer. Apoptosis.
Artesunate shows potent anti-tumor activity in B-cell lymphoma. J 2016;21(11):1265–78.
Hematol Oncol. 2018;11(1):23. 709. Wang T, Gao J, Yu J, Shen L. Synergistic inhibitory effect of wogonin and
low-dose paclitaxel on gastric cancer cells and tumor xenografts. Chin J
Cancer Res. 2013;25(5):505–13.
Luo et al. Chin Med (2019) 14:48 Page 52 of 58
710. Feng Q, Wang H, Pang J, Ji L, Han J, Wang Y, et al. Prevention of wogonin 731. Xiao W, Wu K, Yin M, Han S, Ding Y, Qiao A, et al. Wogonin inhibits
on colorectal cancer tumorigenesis by regulating p53 nuclear translo- tumor-derived regulatory molecules by suppressing STAT3 signaling
cation. Front Pharmacol. 2018;9:1356. to promote tumor immunity. J Immunother (Hagerstown, Md: 1997).
711. Tsai CF, Yeh WL, Huang SM, Tan TW, Lu DY. Wogonin induces reactive 2015;38(5):167–84.
oxygen species production and cell apoptosis in human glioma cancer 732. Yang Y, Li XJ, Chen Z, Zhu XX, Wang J, Zhang LB, et al. Wogonin
cells. Int J Mol Sci. 2012;13(8):9877–92. induced calreticulin/annexin A1 exposure dictates the immuno-
712. Durr C, Hanna BS, Schulz A, Lucas F, Zucknick M, Benner A, et al. Tumor genicity of cancer cells in a PERK/AKT dependent manner. PLoS ONE.
necrosis factor receptor signaling is a driver of chronic lymphocytic leu- 2012;7(12):e50811.
kemia that can be therapeutically targeted by the flavonoid wogonin. 733. Chen F, Qin X, Xu G, Gou S, Jin X. Reversal of cisplatin resistance in
Haematologica. 2018;103(4):688–97. human gastric cancer cells by a wogonin-conjugated Pt(IV) prodrug
713. Xu PP, Zuo HQ, Zhou RF, Chen B, Ouyang J. Wogonin inhibits growth of via attenuating Casein Kinase 2-mediated Nuclear Factor-kappaB
mantle cell lymphoma cells through nuclear factor-kappab signaling pathways. Biochem Pharmacol. 2017;135:50–68.
pathway. Chin Med J. 2018;131(4):495–7. 734. Qin X, Xu G, Chen F, Fang L, Gou S. Novel platinum(IV) complexes
714. Lin CC, Kuo CL, Lee MH, Lai KC, Lin JP, Yang JS, et al. Wogonin triggers conjugated with a wogonin derivative as multi-targeted anticancer
apoptosis in human osteosarcoma U-2 OS cells through the endoplas- agents. Bioorg Med Chem. 2017;25(8):2507–17.
mic reticulum stress, mitochondrial dysfunction and caspase-3-de- 735. Zhao L, Miao HC, Li WJ, Sun Y, Huang SL, Li ZY, et al. LW-213 induces
pendent signaling pathways. Int J Oncol. 2011;39(1):217–24. G2/M cell cycle arrest through AKT/GSK3beta/beta-catenin
715. Song X, Yao J, Wang F, Zhou M, Zhou Y, Wang H, et al. Wogonin inhibits signaling pathway in human breast cancer cells. Mol Carcinog.
tumor angiogenesis via degradation of HIF-1alpha protein. Toxicol Appl 2016;55(5):778–92.
Pharmacol. 2013;271(2):144–55. 736. Li L, Chen P, Ling Y, Song X, Lu Z, He Q, et al. Inhibitory effects of
716. Ruibin J, Bo J, Danying W, Chihong Z, Jianguo F, Linhui G. Therapy GL-V9 on the invasion of human breast carcinoma cells by downreg-
effects of wogonin on ovarian cancer cells. Biomed Res Int. 2017;2017:8. ulating the expression and activity of matrix metalloproteinase-2/9.
717. Liu X, Tian S, Liu M, Jian L, Zhao L. Wogonin inhibits the proliferation Eur J Pharm Sci. 2011;43(5):393–9.
and invasion, and induces the apoptosis of HepG2 and Bel7402 HCC 737. Li L, Lu N, Dai Q, Wei L, Zhao Q, Li Z, et al. GL-V9, a newly synthetic
cells through NFkappaB/Bcl-2, EGFR and EGFR downstream ERK/AKT flavonoid derivative, induces mitochondrial-mediated apoptosis and
signaling. Int J Mol Med. 2016;38(4):1250–6. G2/M cell cycle arrest in human hepatocellular carcinoma HepG2
718. He L, Lu N, Dai Q, Zhao Y, Zhao L, Wang H, et al. Wogonin induced G1 cells. Eur J Pharmacol. 2011;670(1):13–21.
cell cycle arrest by regulating Wnt/beta-catenin signaling pathway and 738. Zhao K, Li G, Yao Y, Zhou Y, Li Z, Guo Q, et al. Activation of phospho-
inactivating CDK8 in human colorectal cancer carcinoma cells. Toxicol- lipase C-gamma1 and translocation of phosphatidylinositol-3,4,5-
ogy. 2013;312:36–47. trisphosphate 3-phosphatase contribute to GL-V9-induced apoptosis
719. Shi G, Wang Q, Zhou X, Li J, Liu H, Gu J, et al. Response of human non- in human gastric cancer cells. Exp Cell Res. 2017;356(1):8–19.
small-cell lung cancer cells to the influence of Wogonin with SGK1 739. Zhang X, Kang Y, Huo T, Tao R, Wang X, Li Z, et al. GL-V9 induced
dynamics. Acta Biochim Biophys Sin. 2017;49(4):302–10. upregulation and mitochondrial localization of NAG-1 associates
720. Yang H, Hui H, Wang Q, Li H, Zhao K, Zhou Y, et al. Wogonin induces cell with ROS generation and cell death in hepatocellular carcinoma
cycle arrest and erythroid differentiation in imatinib-resistant K562 cells cells. Free Radical Biol Med. 2017;112:49–59.
and primary CML cells. Oncotarget. 2014;5(18):8188–201. 740. Tian J, Wang L, Wang L, Ke X. A wogonin-loaded glycyrrhetinic acid-
721. Li SJ, Sun SJ, Gao J, Sun FB. Wogonin induces Beclin-1/PI3K and reactive modified liposome for hepatic targeting with anti-tumor effects.
oxygen species-mediated autophagy in human pancreatic cancer cells. Drug Delivery. 2014;21(7):553–9.
Oncol Lett. 2016;12(6):5059–67. 741. Hong ZP, Wang LG, Wang HJ, Ye WF, Wang XZ. Wogonin exacerbates
722. Chow SE, Chen YW, Liang CA, Huang YK, Wang JS. Wogonin induces the cytotoxic effect of oxaliplatin by inducing nitrosative stress
cross-regulation between autophagy and apoptosis via a variety of Akt and autophagy in human gastric cancer cells. Phytomedicine.
pathway in human nasopharyngeal carcinoma cells. J Cell Biochem. 2018;39:168–75.
2012;113(11):3476–85. 742. Rong LW, Wang RX, Zheng XL, Feng XQ, Zhang L, Zhang L, et al. Com-
723. Zhao Y, Yao J, Wu XP, Zhao L, Zhou YX, Zhang Y, et al. Wogonin sup- bination of wogonin and sorafenib effectively kills human hepatocel-
presses human alveolar adenocarcinoma cell A549 migration in inflam- lular carcinoma cells through apoptosis potentiation and autophagy
matory microenvironment by modulating the IL-6/STAT3 signaling inhibition. Oncol Lett. 2017;13(6):5028–34.
pathway. Mol Carcinog. 2015;54(Suppl 1):E81–93. 743. He F, Wang Q, Zheng XL, Yan JQ, Yang L, Sun H, et al. Wogonin poten-
724. Hong M, Cheng H, Song L, Wang W, Wang Q, Xu D, et al. Wogonin tiates cisplatin-induced cancer cell apoptosis through accumulation
suppresses the activity of matrix metalloproteinase-9 and inhibits of intracellular reactive oxygen species. Oncol Rep. 2012;28(2):601–5.
migration and invasion in human hepatocellular carcinoma. Molecules. 744. Liu M, Chen X, Ma J, Hassan W, Wu H, Ling J, et al. beta-Elemene
2018;23(2):384. attenuates atherosclerosis in apolipoprotein E-deficient mice via
725. Fu R, Chen Y, Wang XP, An T, Tao L, Zhou YX, et al. Wogonin inhibits restoring NO levels and alleviating oxidative stress. Biomed Pharma-
multiple myeloma-stimulated angiogenesis via c-Myc/VHL/HIF-1alpha cother. 2017;95:1789–98.
signaling axis. Oncotarget. 2016;7(5):5715–27. 745. Fang Y, Kang Y, Zou H, Cheng X, Xie T, Shi L, et al. beta-elemene
726. Zhao K, Song X, Huang Y, Yao J, Zhou M, Li Z, et al. Wogonin inhibits attenuates macrophage activation and proinflammatory factor
LPS-induced tumor angiogenesis via suppressing PI3K/Akt/NF-kappaB production via crosstalk with Wnt/beta-catenin signaling pathway.
signaling. Eur J Pharmacol. 2014;737:57–69. Fitoterapia. 2018;124:92–102.
727. Zhou M, Song X, Huang Y, Wei L, Li Z, You Q, et al. Wogonin inhibits 746. Cai B, Ma L, Nong S, Wu Y, Guo X, Pu J. beta-elemene induced anti-
H2O2-induced angiogenesis via suppressing PI3K/Akt/NF-kappaB cancer effect in bladder cancer through upregulation of PTEN and
signaling pathway. Vascul Pharmacol. 2014;60(3):110–9. suppression of AKT phosphorylation. Oncol Lett. 2018;16(5):6019–25.
728. Hu C, Xu M, Qin R, Chen W, Xu X. Wogonin induces apoptosis and 747. Liu Y, Jiang ZY, Zhou YL, Qiu HH, Wang G, Luo Y, et al. beta-elemene
endoplasmic reticulum stress in HL-60 leukemia cells through inhibition regulates endoplasmic reticulum stress to induce the apoptosis of
of the PI3K-AKT signaling pathway. Oncol Rep. 2015;33(6):3146–54. NSCLC cells through PERK/IRE1alpha/ATF6 pathway. Biomed Pharma-
729. Yang L, Wang Q, Li D, Zhou Y, Zheng X, Sun H, et al. Wogonin enhances cother. 2017;93:490–7.
antitumor activity of tumor necrosis factor-related apoptosis-inducing 748. Liu J, Zhang Y, Qu J, Xu L, Hou K, Zhang J, et al. beta-Elemene-
ligand in vivo through ROS-mediated downregulation of cFLIPL and induced autophagy protects human gastric cancer cells from under-
IAP proteins. Apoptosis. 2013;18(5):618–26. going apoptosis. BMC Cancer. 2011;11:183.
730. Wu X, Liu P, Zhang H, Li Y, Salmani JM, Wang F, et al. Wogonin as a tar- 749. Wang L, Zhao Y, Wu Q, Guan Y, Wu X. Therapeutic effects of beta-
geted therapeutic agent for EBV (+) lymphoma cells involved in LMP1/ elemene via attenuation of the Wnt/beta-catenin signaling pathway
NF-kappaB/miR-155/PU1 pathway. BMC Cancer. 2017;17(1):147. in cervical cancer cells. Mol Med Rep. 2018;17(3):4299–306.
Luo et al. Chin Med (2019) 14:48 Page 53 of 58
750. Zhang X, Zhang Y, Li Y. beta-elemene decreases cell invasion by 770. Zhang J, Zhang HD, Yao YF, Zhong SL, Zhao JH, Tang JH. beta-elemene
upregulating E-cadherin expression in MCF-7 human breast cancer reverses chemoresistance of breast cancer cells by reducing resistance
cells. Oncol Rep. 2013;30(2):745–50. transmission via exosomes. Cell Physiol Biochem. 2015;36(6):2274–86.
751. Liang D, Yang M, Guo B, Yang L, Cao J, Zhang X. HIF-1alpha induced 771. Xu L, Guo T, Qu X, Hu X, Zhang Y, Che X, et al. beta-elemene increases
by beta-elemene protects human osteosarcoma cells from undergo- the sensitivity of gastric cancer cells to TRAIL by promoting the forma-
ing apoptosis. J Cancer Res Clin Oncol. 2012;138(11):1865–77. tion of DISC in lipid rafts. Cell Biol Int. 2018;42(10):1377–85.
752. Chen W, Lu Y, Wu J, Gao M, Wang A, Xu B. Beta-elemene inhibits 772. Matsuda K, Hattori S, Komizu Y, Kariya R, Ueoka R, Okada S. Cepharan-
melanoma growth and metastasis via suppressing vascular endothelial thine inhibited HIV-1 cell–cell transmission and cell-free infection
growth factor-mediated angiogenesis. Cancer Chemother Pharmacol. via modification of cell membrane fluidity. Bioorg Med Chem Lett.
2011;67(4):799–808. 2014;24(9):2115–7.
753. Hu T, Gao Y. beta-elemene against Burkitt’s lymphoma via activa- 773. Rogosnitzky M, Danks R. Therapeutic potential of the biscoclaurine alka-
tion of PUMA mediated apoptotic pathway. Biomed Pharmacother. loid, cepharanthine, for a range of clinical conditions. Pharmacol Rep.
2018;106:1557–62. 2011;63(2):337–47.
754. Wu Z, Wang T, Zhang Y, Zheng Z, Yu S, Jing S, et al. Anticancer effects 774. Desgrouas C, Chapus C, Desplans J, Travaille C, Pascual A, Baghdikian
of beta-elemene with hyperthermia in lung cancer cells. Exp Ther Med. B, et al. In vitro antiplasmodial activity of cepharanthine. Malaria J.
2017;13(6):3153–7. 2014;13:327.
755. Zheng F, Tang Q, Zheng XH, Wu J, Huang H, Zhang H, et al. Inactiva- 775. Paudel KR, Karki R, Kim DW. Cepharanthine inhibits in vitro VSMC prolif-
tion of Stat3 and crosstalk of miRNA155-5p and FOXO3a contribute to eration and migration and vascular inflammatory responses mediated
the induction of IGFBP1 expression by beta-elemene in human lung by RAW264.7. Toxicol Vitro. 2016;34:16–25.
cancer. Exp Mol Med. 2018;50(9):121. 776. Uthaisar K, Seubwai W, Srikoon P, Vaeteewoottacharn K, Sawanyawisuth
756. Zhu T, Zhao Y, Zhang J, Li L, Zou L, Yao Y, et al. ss-Elemene inhibits prolif- K, Okada S, et al. Cepharanthine suppresses metastatic potential of
eration of human glioblastoma cells and causes cell-cycle G0/G1 arrest human cholangiocarcinoma cell lines. Asian Pac J Cancer Prevent.
via mutually compensatory activation of MKK3 and MKK6. Int J Oncol. 2012;13(Suppl):149–54.
2011;38(2):419–26. 777. Lyu J, Yang EJ, Head SA, Ai N, Zhang B, Wu C, et al. Pharmacological
757. Liu J, Hu XJ, Jin B, Qu XJ, Hou KZ, Liu YP. beta-Elemene induces apop- blockade of cholesterol trafficking by cepharanthine in endothe-
tosis as well as protective autophagy in human non-small-cell lung lial cells suppresses angiogenesis and tumor growth. Cancer Lett.
cancer A549 cells. J Pharm Pharmacol. 2012;64(1):146–53. 2017;409:91–103.
758. Zhou K, Wang L, Cheng R, Liu X, Mao S, Yan Y. Elemene increases 778. Harada T, Harada K, Ueyama Y. The enhancement of tumor radiore-
autophagic apoptosis and drug sensitivity in human cisplatin (DDP)- sponse by combined treatment with cepharanthine is accompanied by
resistant lung cancer cell line SPC-A-1/DDP by inducing beclin-1 the inhibition of DNA damage repair and the induction of apoptosis in
expression. Oncol Res. 2017. https://doi.org/10.3727/096504017X14954 oral squamous cell carcinoma. Int J Oncol. 2012;41:565–72.
936991990. 779. Li H, Yan Z, Ning W, Xiao-Juan G, Cai-Hong Z, Jin-Hua J, et al. Using
759. Zhang Y, Sun X, Nan N, Cao KX, Ma C, Yang GW, et al. Elemene inhibits rhodamine 123 accumulation in CD8 + cells as a surrogate indicator to
the migration and invasion of 4T1 murine breast cancer cells via hep- study the P-glycoprotein modulating effect of cepharanthine hydro-
aranase. Mol Med Rep. 2017;16(1):794–800. chloride in vivo. J Biomed Biotechnol. 2011;2011:7.
760. Lin L, Li L, Chen X, Zeng B, Lin T. Preliminary evaluation of the potential 780. Zhou P, Zhang R, Wang Y, Xu D, Zhang L, Qin J, et al. Cepharanthine
role of beta-elemene in reversing erlotinib-resistant human NSCLC hydrochloride reverses the mdr1 (P-glycoprotein)-mediated esopha-
A549/ER cells. Oncol Lett. 2018;16(3):3380–8. geal squamous cell carcinoma cell cisplatin resistance through JNK and
761. Yao CC, Tu YR, Jiang J, Ye SF, Du HX, Zhang Y. beta-elemene reverses the p53 signals. Oncotarget. 2017;8(67):111144–60.
drug resistance of lung cancer A549/DDP cells via the mitochondrial 781. Han L, Wang Y, Guo X, Zhou Y, Zhang J, Wang N, et al. Downregulation
apoptosis pathway. Oncol Rep. 2014;31(5):2131–8. of MDR1 gene by cepharanthine hydrochloride is related to the activa-
762. Yu X, Xu M, Li N, Li Z, Li H, Shao S, et al. beta-elemene inhibits tumor- tion of c-Jun/JNK in K562/ADR cells. Biomed Res Int. 2014;2014:6.
promoting effect of M2 macrophages in lung cancer. Biochem Biophys 782. Tang ZH, Cao WX, Guo X, Dai XY, Lu JH, Chen X, et al. Identification of a
Res Commun. 2017;490(2):514–20. novel autophagic inhibitor cepharanthine to enhance the anti-cancer
763. Shi F, Yang G, Ren J, Guo T, Du Y, Feng N. Formulation design, prepara- property of dacomitinib in non-small cell lung cancer. Cancer Lett.
tion, and in vitro and in vivo characterizations of beta-Elemene-loaded 2018;412:1–9.
nanostructured lipid carriers. Int J Nanomed. 2013;8:2533–41. 783. Rattanawong A, Payon V, Limpanasittikul W, Boonkrai C, Mutirangura
764. Zeng Y-Y, Zeng Y-J, Zhang N-N, Li C-X, Xie T, Zeng Z-W. The prepara- A, Wonganan P. Cepharanthine exhibits a potent anticancer activity in
tion, determination of a flexible complex liposome co-loaded with p53-mutated colorectal cancer cells through upregulation of p21Waf1/
cabazitaxel and β-elemene, and animal pharmacodynamics on Cip1. Oncol Rep. 2018;39(1):227–38.
paclitaxel-resistant lung adenocarcinoma. Molecules (Basel, Switzer- 784. Zhu Q, Guo B, Chen L, Ji Q, Liang H, Wen N, et al. Cepharanthine exerts
land). 2019;24(9):1697. antitumor activity on choroidal melanoma by reactive oxygen spe-
765. Hu CJ, Zhao XL, Li JZ, Kang SM, Yang CR, Jin YH, et al. Preparation and cies production and c-Jun N-terminal kinase activation. Oncol Lett.
characterization of beta-elemene-loaded microemulsion. Drug Dev Ind 2017;13(5):3760–6.
Pharm. 2011;37(7):765–74. 785. Gao S, Li X, Ding X, Qi W, Yang Q. Cepharanthine induces autophagy,
766. Yu Z, Wu F, Chen L, Li Q, Wang C, Dong J, et al. ETME, a novel beta- apoptosis and cell cycle arrest in breast cancer cells. Cell Physiol Bio-
elemene derivative, synergizes with arsenic trioxide in inducing apop- chem. 2017;41(4):1633–48.
tosis and cell cycle arrest in hepatocarcinoma cells via a p53-depend- 786. Hua P, Sun M, Zhang G, Zhang Y, Tian X, Li X, et al. Cepharanthine
ent pathway. Acta Pharm Sinica B. 2014;4(6):424–9. induces apoptosis through reactive oxygen species and mitochon-
767. Ding XF, Shen M, Xu LY, Dong JH, Chen G. 13,14-bis(cis-3,5-dimethyl- drial dysfunction in human non-small-cell lung cancer cells. Biochem
1-piperazinyl)-beta-elemene, a novel beta-elemene derivative, shows Biophys Res Commun. 2015;460(2):136–42.
potent antitumor activities via inhibition of mTOR in human breast 787. Payon V, Kongsaden C, Ketchart W, Mutirangura A, Wonganan P. Mecha-
cancer cells. Oncol Lett. 2013;5(5):1554–8. nism of cepharanthine cytotoxicity in human ovarian cancer cells.
768. Huang C, Yu Y. Synergistic cytotoxicity of beta-elemene and cisplatin Planta Med. 2019;85(1):41–7.
in gingival squamous cell carcinoma by inhibition of STAT3 signaling 788. Huang CZ, Wang YF, Zhang Y, Peng YM, Liu YX, Ma F, et al. Cepharan-
pathway. Med Sci Monit. 2017;23:1507–13. thine hydrochloride reverses Pglycoprotein-mediated multidrug resist-
769. Mu L, Wang T, Chen Y, Tang X, Yuan Y, Zhao Y. beta-Elemene ance in human ovarian carcinoma A2780/Taxol cells by inhibiting the
enhances the efficacy of gefitinib on glioblastoma multiforme cells PI3K/Akt signaling pathway. Oncol Rep. 2017;38(4):2558–64.
through the inhibition of the EGFR signaling pathway. Int J Oncol. 789. Liu G, Wu D, Liang X, Yue H, Cui Y. Mechanisms and in vitro effects
2016;49(4):1427–36. of cepharanthine hydrochloride: classification analysis of the
Luo et al. Chin Med (2019) 14:48 Page 54 of 58
drug-induced differentially-expressed genes of human nasopharyngeal 811. Sundram V, Chauhan SC, Ebeling M, Jaggi M. Curcumin attenuates
carcinoma cells. Oncol Rep. 2015;34(4):2002–10. β-catenin signaling in prostate cancer cells through activation of
790. Xu W, Wang X, Tu Y, Masaki H, Tanaka S, Onda K, et al. Tetrandrine and protein kinase D1. PLoS ONE. 2012;7(4):e35368.
cepharanthine induce apoptosis through caspase cascade regulation, 812. Seo BR, Min K-J, Cho IJ, Kim SC, Kwon TK. Curcumin significantly
cell cycle arrest, MAPK activation and PI3K/Akt/mTOR signal modifica- enhances dual PI3K/Akt and mTOR inhibitor NVP-BEZ235-induced
tion in glucocorticoid resistant human leukemia Jurkat T cells. Chem- apoptosis in human renal carcinoma Caki cells through down-regula-
Biol Interactions. 2019;310:108726. tion of p53-dependent Bcl-2 expression and inhibition of Mcl-1 protein
791. Law BY, Chan WK, Xu SW, Wang JR, Bai LP, Liu L, et al. Natural small- stability. PLoS ONE. 2014;9(4):e95588.
molecule enhancers of autophagy induce autophagic cell death in 813. Li Y, Sun W, Han N, Zou Y, Yin D. Curcumin inhibits proliferation, migra-
apoptosis-defective cells. Sci Rep. 2014;4:5510. tion, invasion and promotes apoptosis of retinoblastoma cell lines
792. Villeneuve NF, Sun Z, Chen W, Zhang DD. Nrf2 and p21 regulate the fine through modulation of miR-99a and JAK/STAT pathway. BMC Cancer.
balance between life and death by controlling ROS levels. Cell Cycle 2018;18(1):1230.
(Georgetown, Tex). 2009;8(20):3255–6. 814. Amin ARMR, Haque A, Rahman MA, Chen ZG, Khuri FR, Shin DM. Cur-
793. Uto T, Nishi Y, Toyama M, Yoshinaga K, Baba M. Inhibitory effect of cumin induces apoptosis of upper aerodigestive tract cancer cells by
cepharanthine on dendritic cell activation and function. Int Immunop- targeting multiple pathways. PLoS One. 2015;10(4):e0124218.
harmacol. 2011;11(11):1932–8. 815. Tang H, Zeng L, Wang J, Zhang X, Ruan Q, Wang J, et al. Reversal of
794. Yu HH, Mi WN, Liu B, Zhao HP. In vitro and in vivo effect of paclitaxel and 5-fluorouracil resistance by EGCG is mediate by inactivation of TFAP2A/
cepharanthine co-loaded polymeric nanoparticles in gastric cancer. J VEGF signaling pathway and down-regulation of MDR-1 and P-gp
BUON. 2016;21(1):125–34. expression in gastric cancer. Oncotarget. 2017;8(47):82842–53.
795. Zhang LH, Yang GL, Zhang RY, Dong L, Chen HG, Bo JJ, et al. Curcumin 816. Zhu BH, Chen HY, Zhan WH, Wang CY, Cai SR, Wang Z, et al. (−)-Epigal-
inhibits cell proliferation and motility via suppression of TROP2 in blad- locatechin-3-gallate inhibits VEGF expression induced by IL-6 via Stat3
der cancer cells. Int J Oncol. 2018;53(2):515–26. in gastric cancer. World J Gastroenterol. 2011;17(18):2315–25.
796. Yoshida K, Toden S, Ravindranathan P, Han H, Goel A. Curcumin 817. Chen PN, Chu SC, Kuo WH, Chou MY, Lin JK, Hsieh YS. Epigallocat-
sensitizes pancreatic cancer cells to gemcitabine by attenuating echin-3 gallate inhibits invasion, epithelial-mesenchymal transi-
PRC2 subunit EZH2, and the lncRNA PVT1 expression. Carcinogenesis. tion, and tumor growth in oral cancer cells. J Agric Food Chem.
2017;38(10):1036–46. 2011;59(8):3836–44.
797. Thacker PC, Karunagaran D. Curcumin and emodin down-regulate 818. Sharma C, Nusri Qel A, Begum S, Javed E, Rizvi TA, Hussain A. (-)-Epi-
TGF-β signaling pathway in human cervical cancer cells. PLoS ONE. gallocatechin-3-gallate induces apoptosis and inhibits invasion and
2015;10(3):e0120045. migration of human cervical cancer cells. Asian Pac J Cancer Prevent.
798. Link A, Balaguer F, Shen Y, Lozano JJ, Leung H-CE, Boland CR, et al. 2012;13(9):4815–22.
Curcumin modulates DNA methylation in colorectal cancer cells. PLoS 819. Zhang X, Min KW, Wimalasena J, Baek SJ. Cyclin D1 degradation and
ONE. 2013;8(2):e57709. p21 induction contribute to growth inhibition of colorectal cancer
799. Lim T-G, Lee S-Y, Huang Z, Lim DY, Chen H, Jung SK, et al. Curcumin sup- cells induced by epigallocatechin-3-gallate. J Cancer Res Clin Oncol.
presses proliferation of colon cancer cells by targeting CDK2. Cancer 2012;138(12):2051–60.
Prevent Res (Philadelphia, Pa). 2014;7(4):466–74. 820. Kostin SF, McDonald DE, McFadden DW. Inhibitory effects of (-)-epigal-
800. Murray-Stewart T, Dunworth M, Lui Y, Giardiello FM, Woster PM, Casero locatechin-3-gallate and pterostilbene on pancreatic cancer growth
RA Jr. Curcumin mediates polyamine metabolism and sensitizes gastro- in vitro. J Surg Res. 2012;177(2):255–62.
intestinal cancer cells to antitumor polyamine-targeted therapies. PLoS 821. Min NY, Kim JH, Choi JH, Liang W, Ko YJ, Rhee S, et al. Selective death
ONE. 2018;13(8):e0202677. of cancer cells by preferential induction of reactive oxygen species in
801. Shao S, Duan W, Xu Q, Li X, Han L, Li W, et al. Curcumin suppresses response to (-)-epigallocatechin-3-gallate. Biochem Biophys Res Com-
hepatic stellate cell-induced hepatocarcinoma angiogenesis and mun. 2012;421(1):91–7.
invasion through downregulating CTGF. Oxid Med Cell Longevity. 822. Mineva ND, Paulson KE, Naber SP, Yee AS, Sonenshein GE. Epigallocate-
2019;2019:8148510. chin-3-gallate inhibits stem-like inflammatory breast cancer cells. PLoS
802. Du WZ, Feng Y, Wang XF, Piao XY, Cui YQ, Chen LC, et al. Curcumin sup- ONE. 2013;8(9):e73464.
presses malignant glioma cells growth and induces apoptosis by inhibi- 823. Jang JY, Lee JK, Jeon YK, Kim CW. Exosome derived from epigallocat-
tion of SHH/GLI1 signaling pathway in vitro and vivo. CNS Neurosci echin gallate treated breast cancer cells suppresses tumor growth by
Ther. 2013;19(12):926–36. inhibiting tumor-associated macrophage infiltration and M2 polariza-
803. Gao SM, Yang JJ, Chen CQ, Chen JJ, Ye LP, Wang LY, et al. Pure curcumin tion. BMC Cancer. 2013;13:421.
decreases the expression of WT1 by upregulation of miR-15a and miR- 824. Lee SH, Nam HJ, Kang HJ, Kwon HW, Lim YC. Epigallocatechin-
16-1 in leukemic cells. J Exp Clin Cancer Res: CR. 2012;31(1):27. 3-gallate attenuates head and neck cancer stem cell traits through
804. Marquardt JU, Gomez-Quiroz L, Camacho LOA, Pinna F, Hepatology SS. suppression of Notch pathway. Eur J Cancer (Oxford, England: 1990).
Curcumin effectively inhibits oncogenic NF-κB signaling and restrains 2013;49(15):3210–8.
stemness features in liver cancer. J Hepatol. 2015;63(3):661–9. 825. Singh T, Katiyar SK. Green tea polyphenol, (-)-epigallocatechin-3-gallate,
805. Wu G-Q, Chai K-Q, Zhu X-M, Jiang H, Wang X, Xue Q, et al. Anti-cancer induces toxicity in human skin cancer cells by targeting beta-catenin
effects of curcumin on lung cancer through the inhibition of EZH2 and signaling. Toxicol Appl Pharmacol. 2013;273(2):418–24.
NOTCH1. Oncotarget. 2016;7(18):26535–50. 826. Hwang YS, Park KK, Chung WY. Epigallocatechin-3 gallate inhibits
806. Zhang C, Hao Y, Wu L, Dong X, Jiang N, Cong B, et al. Curcumin induces cancer invasion by repressing functional invadopodia formation in oral
apoptosis and inhibits angiogenesis in murine malignant mesothe- squamous cell carcinoma. Eur J Pharmacol. 2013;715(1–3):286–95.
lioma. Int J Oncol. 2018;53(6):2531–41. 827. Wang S, Chen R, Zhong Z, Shi Z, Chen M, Wang Y. Epigallocatechin-
807. Sidhar H, Giri RK. Induction of Bex genes by curcumin is associated with 3-gallate potentiates the effect of curcumin in inducing growth
apoptosis and activation of p53 in N2a neuroblastoma cells. Sci Rep. inhibition and apoptosis of resistant breast cancer cells. Am J Chin Med.
2017;7:41420. 2014;42(5):1279–300.
808. Dudás J, Fullár A, Romani A, Pritz C, Kovalszky I, Hans Schartinger V, et al. 828. Kim KC, Lee C. Reversal of Cisplatin resistance by epigallocatechin
Curcumin targets fibroblast-tumor cell interactions in oral squamous gallate is mediated by downregulation of axl and tyro 3 expression in
cell carcinoma. Exp Cell Res. 2013;319(6):800–9. human lung cancer cells. Korean J Physiol Pharmacol. 2014;18(1):61–6.
809. Zhou X, Su J, Feng S, Wang L, Yin X, Yan J, et al. Antitumor activity of 829. Lin CH, Chao LK, Hung PH, Chen YJ. EGCG inhibits the growth and
curcumin is involved in down-regulation of YAP/TAZ expression in tumorigenicity of nasopharyngeal tumor-initiating cells through
pancreatic cancer cells. Oncotarget. 2016;7(48):79076–88. attenuation of STAT3 activation. Int J Clin Exp Pathol. 2014;7(5):2372–81.
810. Huang H, Chen X, Li D, He Y, Li Y, Du Z, et al. Combination of α-tomatine 830. Bimonte S, Leongito M, Barbieri A, Del Vecchio V, Barbieri M, Albino V,
and curcumin inhibits growth and induces apoptosis in human pros- et al. Inhibitory effect of (-)-epigallocatechin-3-gallate and bleomycin
tate cancer cells. PLoS ONE. 2015;10(12):e0144293.
Luo et al. Chin Med (2019) 14:48 Page 55 of 58
on human pancreatic cancer MiaPaca-2 cell growth. Infect Agents 851. Barzegar E, Fouladdel S, Movahhed TK, Atashpour S, Ghahremani MH,
Cancer. 2015;10:22. Ostad SN, et al. Effects of berberine on proliferation, cell cycle distribu-
831. Mayr C, Wagner A, Neureiter D, Pichler M, Jakab M, Illig R, et al. The tion and apoptosis of human breast cancer T47D and MCF7 cell lines.
green tea catechin epigallocatechin gallate induces cell cycle arrest Iran J Basic Med Sci. 2015;18(4):334–42.
and shows potential synergism with cisplatin in biliary tract cancer 852. Li J, Cao B, Liu X, Fu X, Xiong Z, Chen L, et al. Berberine suppresses
cells. BMC Complement Altern Med. 2015;15:194. androgen receptor signaling in prostate cancer. Mol Cancer Ther.
832. Moses MA, Henry EC, Ricke WA, Gasiewicz TA. The heat shock protein 2011;10(8):1346–56.
90 inhibitor, (-)-epigallocatechin gallate, has anticancer activity in a 853. Qi HW, Xin LY, Xu X, Ji XX, Fan LH. Epithelial-to-mesenchymal transition
novel human prostate cancer progression model. Cancer Prevent Res markers to predict response of Berberine in suppressing lung cancer
(Philadelphia, Pa). 2015;8(3):249–57. invasion and metastasis. J Transl Med. 2014;12:22.
833. De Amicis F, Russo A, Avena P, Santoro M, Vivacqua A, Bonofiglio D, 854. Kim S, Han J, Lee SK, Choi MY, Kim J, Lee J, et al. Berberine suppresses
et al. In vitro mechanism for downregulation of ER-alpha expression by the TPA-induced MMP-1 and MMP-9 expressions through the inhibition
epigallocatechin gallate in ER +/PR + human breast cancer cells. Mol of PKC-alpha in breast cancer cells. J Surg Res. 2012;176(1):e21–9.
Nutr Food Res. 2013;57(5):840–53. 855. Zhang XZ, Wang L, Liu DW, Tang GY, Zhang HY. Synergistic inhibitory
834. Pan X, Zhao B, Song Z, Han S, Wang M. Estrogen receptor-alpha36 effect of berberine and d-limonene on human gastric carcinoma cell
is involved in epigallocatechin-3-gallate induced growth inhibition line MGC803. J Med Food. 2014;17(9):955–62.
of ER-negative breast cancer stem/progenitor cells. J Pharmacol Sci. 856. Yip NK, Ho WS. Berberine induces apoptosis via the mitochondrial path-
2016;130(2):85–93. way in liver cancer cells. Oncol Rep. 2013;30(3):1107–12.
835. Li X, Zhang A, Sun H, Liu Z, Zhang T, Qiu S, et al. Metabolic characteri- 857. Yi T, Zhuang L, Song G, Zhang B, Li G, Hu T. Akt signaling is associated
zation and pathway analysis of berberine protects against prostate with the berberine-induced apoptosis of human gastric cancer cells.
cancer. Oncotarget. 2017;8(39):65022–41. Nutr Cancer. 2015;67(3):523–31.
836. Park JJ, Seo SM, Kim EJ, Lee YJ, Ko YG, Ha J, et al. Berberine inhibits 858. Kuo H-P, Lee Y-J, Hsu C-Y, Lee S-L, Hsu S-C, Chuang T-C, et al. Growth-
human colon cancer cell migration via AMP-activated protein kinase- suppressive effect of berberine on endometrial carcinoma cells: role of
mediated downregulation of integrin beta1 signaling. Biochem Biophys mitochondrial and PI3K/Akt pathway. J Funct Foods. 2015;17:600–9.
Res Commun. 2012;426(4):461–7. 859. Wen C, Wu L, Fu L, Zhang X, Zhou H. Berberine enhances the antitumor
837. Kim J-H, Ryu AR, Kang M-J, Lee M-Y. Berberine-induced changes in activity of tamoxifen in drugsensitive MCF7 and drugresistant MCF7/
protein expression and antioxidant enzymes in melanoma cells. Mol TAM cells. Mol Med Rep. 2016;14(3):2250–6.
Cell Toxicol. 2016;12(1):53–61. 860. Wang N, Wang X, Tan HY, Li S, Tsang CM, Tsao SW, et al. Berberine
838. Wu HL, Chuang TY, Al-Hendy A, Diamond MP, Azziz R, Chen YH. Berber- suppresses cyclin D1 expression through proteasomal degradation in
ine inhibits the proliferation of human uterine leiomyoma cells. Fertil human hepatoma cells. Int J Mol Sci. 2016;17(11):1899.
Steril. 2015;103(4):1098–106. 861. Liu Q, Xu X, Zhao M, Wei Z, Li X, Zhang X, et al. Berberine induces senes-
839. Pazhang Y, Ahmadian S, Mahmoudian M, Shafiezadeh M. Berberine- cence of human glioblastoma cells by downregulating the EGFR-MEK-
induced apoptosis via decreasing the survivin protein in K562 cell line. ERK signaling pathway. Mol Cancer Ther. 2015;14(2):355–63.
Med Oncol (Northwood, London, England). 2011;28(4):1577–83. 862. Yang X, Yang B, Cai J, Zhang C, Zhang Q, Xu L, et al. Berberine enhances
840. Ke R, Vishnoi K, Viswakarma N, Santha S, Das S, Rana A, et al. Involve- radiosensitivity of esophageal squamous cancer by targeting HIF-
ment of AMP-activated protein kinase and death receptor 5 in TRAIL- 1alpha in vitro and in vivo. Cancer Biol Ther. 2013;14(11):1068–73.
berberine-induced apoptosis of cancer cells. Sci Rep. 2018;8(1):5521. 863. Hu Q, Li L, Zou X, Xu L, Yi P. Berberine attenuated proliferation, invasion
841. Li J, Li O, Kan M, Zhang M, Shao D, Pan Y, et al. Berberine induces and migration by targeting the AMPK/HNF4alpha/WNT5A pathway in
apoptosis by suppressing the arachidonic acid metabolic pathway in gastric carcinoma. Front Pharmacol. 2018;9:1150.
hepatocellular carcinoma. Mol Med Rep. 2015;12(3):4572–7. 864. Seo YS, Yim MJ, Kim BH, Kang KR, Lee SY, Oh JS, et al. Berberine-induced
842. Jeong Y, You D, Kang HG, Yu J, Kim SW, Nam SJ, et al. Berberine sup- anticancer activities in FaDu head and neck squamous cell carcinoma
presses fibronectin expression through inhibition of c-Jun phospho- cells. Oncol Rep. 2015;34(6):3025–34.
rylation in breast cancer cells. J Breast Cancer. 2018;21(1):21–7. 865. Wang J, Yang S, Cai X, Dong J, Chen Z, Wang R, et al. Berberine inhibits
843. Kim JS, Oh D, Yim MJ, Park JJ, Kang KR, Cho IA, et al. Berberine induces EGFR signaling and enhances the antitumor effects of EGFR inhibitors
FasL-related apoptosis through p38 activation in KB human oral cancer in gastric cancer. Oncotarget. 2016;7(46):76076–86.
cells. Oncol Rep. 2015;33(4):1775–82. 866. Zhao Y, Jing Z, Lv J, Zhang Z, Lin J, Cao X, et al. Berberine activates
844. Marverti G, Ligabue A, Lombardi P, Ferrari S, Monti MG, Frassineti C, et al. caspase-9/cytochrome c-mediated apoptosis to suppress triple-
Modulation of the expression of folate cycle enzymes and polyamine negative breast cancer cells in vitro and in vivo. Biomed Pharmacother.
metabolism by berberine in cisplatin-sensitive and -resistant human 2017;95:18–24.
ovarian cancer cells. Int J Oncol. 2013;43(4):1269–80. 867. Yu R, Zhang ZQ, Wang B, Jiang HX, Cheng L, Shen LM. Berberine-
845. He W, Wang B, Zhuang Y, Shao D, Sun K, Chen J. Berberine inhibits induced apoptotic and autophagic death of HepG2 cells requires AMPK
growth and induces G1 arrest and apoptosis in human cholangiocarci- activation. Cancer Cell Int. 2014;14:49.
noma QBC939 cells. J Pharmacol Sci. 2012;119(4):341–8. 868. Li DX, Zhang J, Zhang Y, Zhao PW, Yang LM. Inhibitory effect of berber-
846. Wang J, Peng Y, Liu Y, Yang J, Ding N, Tan W. Berberine, a natural com- ine on human skin squamous cell carcinoma A431 cells. Genet Mol Res.
pound, suppresses Hedgehog signaling pathway activity and cancer 2015;14(3):10553–68.
growth. BMC Cancer. 2015;15:595. 869. Liu X, Ji Q, Ye N, Sui H, Zhou L, Zhu H, et al. Berberine inhibits invasion
847. Park SH, Sung JH, Chung N. Berberine diminishes side population and metastasis of colorectal cancer cells via COX-2/PGE2 mediated
and down-regulates stem cell-associated genes in the pancre- JAK2/STAT3 signaling pathway. PLoS ONE. 2015;10(5):e0123478.
atic cancer cell lines PANC-1 and MIA PaCa-2. Mol Cell Biochem. 870. Saxena S, Shukla S, Kakkar P. Berberine induced modulation of PHLPP2-
2014;394(1–2):209–15. Akt-MST1 kinase signaling is coupled with mitochondrial impairment
848. Wang C, Wang H, Zhang Y, Guo W, Long C, Wang J, et al. Berberine and hepatoma cell death. Toxicol Appl Pharmacol. 2018;347:92–103.
inhibits the proliferation of human nasopharyngeal carcinoma cells via 871. Li X, Gu S, Sun D, Dai H, Chen H, Zhang Z. The selectivity of artemisinin-
an Epstein–Barr virus nuclear antigen 1-dependent mechanism. Oncol based drugs on human lung normal and cancer cells. Environ Toxicol
Rep. 2017;37(4):2109–20. Pharmacol. 2018;57:86–94.
849. Puthdee N, Seubwai W, Vaeteewoottacharn K, Boonmars T, Cha’on U, 872. Chen J, Zhang L, Hao M. Effect of artemisinin on proliferation and
Phoomak C, et al. Berberine induces cell cycle arrest in cholangiocar- apoptosis-related protein expression in vivo and in vitro. Saudi J Biol Sci.
cinoma cell lines via inhibition of NF-kappaB and STAT3 pathways. Biol 2018;25(7):1488–93.
Pharm Bull. 2017;40(6):751–7. 873. Tran KQ, Tin AS, Firestone GL. Artemisinin triggers a G1 cell cycle arrest
850. Raghav D, Ashraf SM, Mohan L, Rathinasamy K. Berberine induces toxic- of human Ishikawa endometrial cancer cells and inhibits cyclin-
ity in HeLa cells through perturbation of microtubule polymerization by dependent kinase-4 promoter activity and expression by disrupting
binding to tubulin at a unique site. Biochemistry. 2017;56(20):2594–611.
Luo et al. Chin Med (2019) 14:48 Page 56 of 58
nuclear factor-kappaB transcriptional signaling. Anticancer Drugs. 894. He BC, Gao JL, Luo X, Luo J, Shen J, Wang L, et al. Ginsenoside Rg3
2014;25(3):270–81. inhibits colorectal tumor growth through the down-regulation of Wnt/
874. Suberu JO, Romero-Canelon I, Sullivan N, Lapkin AA, Barker GC. Com- ss-catenin signaling. Int J Oncol. 2011;38(2):437–45.
parative cytotoxicity of artemisinin and cisplatin and their interactions 895. Liu T, Duo L, Duan P. Ginsenoside Rg3 sensitizes colorectal cancer to
with chlorogenic acids in MCF7 breast cancer cells. ChemMedChem. radiotherapy through downregulation of proliferative and angiogenic
2014;9(12):2791–7. biomarkers. Evid Based Complement Altern Med. 2018;2018:1580427.
875. Gao W, Xiao F, Wang X, Chen T. Artemisinin induces A549 cell 896. Wang L, Li X, Song YM, Wang B, Zhang FR, Yang R, et al. Ginsenoside
apoptosis dominantly via a reactive oxygen species-mediated Rg3 sensitizes human non-small cell lung cancer cells to gamma-radi-
amplification activation loop among caspase-9, -8 and -3. Apoptosis. ation by targeting the nuclear factor-kappaB pathway. Mol Med Rep.
2013;18(10):1201–13. 2015;12(1):609–14.
876. Yang Y, Zhang X, Wang X, Zhao X, Ren T, Wang F, et al. Enhanced deliv- 897. Shan X, Aziz F, Tian LL, Wang XQ, Yan Q, Liu JW. Ginsenoside Rg3-
ery of artemisinin and its analogues to cancer cells by their adducts induced EGFR/MAPK pathway deactivation inhibits melanoma
with human serum transferrin. Int J Pharm. 2014;467(1–2):113–22. cell proliferation by decreasing FUT4/LeY expression. Int J Oncol.
877. Chen G, Gong R, Shi X, Yang D, Zhang G, Lu A, et al. Halofuginone and 2015;46(4):1667–76.
artemisinin synergistically arrest cancer cells at the G1/G0 phase by 898. Xia T, Wang YN, Zhou CX, Wu LM, Liu Y, Zeng QH, et al. Ginsenoside Rh2
upregulating p21Cip1 and p27Kip1. Oncotarget. 2016;7(31):50302–14. and Rg3 inhibit cell proliferation and induce apoptosis by increasing
878. Ganguli A, Choudhury D, Datta S, Bhattacharya S, Chakrabarti G. mitochondrial reactive oxygen species in human leukemia Jurkat cells.
Inhibition of autophagy by chloroquine potentiates synergistically Mol Med Rep. 2017;15(6):3591–8.
anti-cancer property of artemisinin by promoting ROS dependent 899. Yu Y, Zhang C, Liu L, Li X. Hepatic arterial administration of ginsenoside
apoptosis. Biochimie. 2014;107:338–49. Rg3 and transcatheter arterial embolization for the treatment of VX2
879. Zhu S, Liu W, Ke X, Li J, Hu R, Cui H, et al. Artemisinin reduces cell liver carcinomas. Exp Ther Med. 2013;5(3):761–6.
proliferation and induces apoptosis in neuroblastoma. Oncol Rep. 900. Joo EJ, Chun J, Ha YW, Ko HJ, Xu MY, Kim YS. Novel roles of ginsenoside
2014;32(3):1094–100. Rg3 in apoptosis through downregulation of epidermal growth factor
880. Weifeng T, Feng S, Xiangji L, Changqing S, Zhiquan Q, Huazhong Z, receptor. Chem Biol Interact. 2015;233:25–34.
et al. Artemisinin inhibits in vitro and in vivo invasion and metas- 901. Cho J, Rho O, Junco J, Carbajal S, Siegel D, Slaga TJ, et al. Effect of
tasis of human hepatocellular carcinoma cells. Phytomedicine. combined treatment with ursolic acid and resveratrol on skin tumor
2011;18(2–3):158–62. promotion by 12-O-tetradecanoylphorbol-13-acetate. Cancer Prevent
881. Deng XR, Liu ZX, Liu F, Pan L, Yu HP, Jiang JP, et al. Holotransferrin Res (Philadelphia, Pa). 2015;8(9):817–25.
enhances selective anticancer activity of artemisinin against human 902. Gao N, Cheng S, Budhraja A, Gao Z, Chen J, Liu EH, et al. Ursolic acid
hepatocellular carcinoma cells. J Huazhong Univ Sci Technol Med Sci. induces apoptosis in human leukaemia cells and exhibits anti-leukae-
2013;33(6):862–5. mic activity in nude mice through the PKB pathway. Br J Pharmacol.
882. Jia J, Qin Y, Zhang L, Guo C, Wang Y, Yue X, et al. Artemisinin inhibits 2012;165(6):1813–26.
gallbladder cancer cell lines through triggering cell cycle arrest and 903. Lin CC, Huang CY, Mong MC, Chan CY, Yin MC. Antiangiogenic potential
apoptosis. Mol Med Rep. 2016;13(5):4461–8. of three triterpenic acids in human liver cancer cells. J Agric Food
883. Xiao F, Gao W, Wang X, Chen T. Amplification activation loop between Chem. 2011;59(2):755–62.
caspase-8 and -9 dominates artemisinin-induced apoptosis of ASTC-a-1 904. Song YH, Jeong SJ, Kwon HY, Kim B, Kim SH, Yoo DY. Ursolic acid from
cells. Apoptosis. 2012;17(6):600–11. Oldenlandia diffusa induces apoptosis via activation of caspases and
884. Sin S, Kim SY, Kim SS. Chronic treatment with ginsenoside Rg3 induces phosphorylation of glycogen synthase kinase 3 beta in SK-OV-3 ovarian
Akt-dependent senescence in human glioma cells. Int J Oncol. cancer cells. Biol Pharm Bull. 2012;35(7):1022–8.
2012;41(5):1669–74. 905. Wu B, Wang X, Chi ZF, Hu R, Zhang R, Yang W, et al. Ursolic acid-induced
885. Wu K, Li N, Sun H, Xu T, Jin F, Nie J. Endoplasmic reticulum stress apoptosis in K562 cells involving upregulation of PTEN gene expres-
activation mediates Ginseng Rg3-induced anti-gallbladder cancer cell sion and inactivation of the PI3K/Akt pathway. Arch Pharmacal Res.
activity. Biochem Biophys Res Commun. 2015;466(3):369–75. 2012;35(3):543–8.
886. Li J, Liu T, Zhao L, Chen W, Hou H, Ye Z, et al. Ginsenoside 20(S)Rg3 906. Zhang Y, Huang L, Shi H, Chen H, Tao J, Shen R, et al. Ursolic acid
inhibits the Warburg effect through STAT3 pathways in ovarian cancer enhances the therapeutic effects of oxaliplatin in colorectal cancer by
cells. Int J Oncol. 2015;46(2):775–81. inhibition of drug resistance. Cancer Sci. 2018;109(1):94–102.
887. Zhou Y, Zheng X, Lu J, Chen W, Li X, Zhao L. Ginsenoside 20(S)- 907. Zheng QY, Jin FS, Yao C, Zhang T, Zhang GH, Ai X. Ursolic acid-induced
Rg3 inhibits the warburg effect via modulating DNMT3A/MiR- AMP-activated protein kinase (AMPK) activation contributes to growth
532-3p/HK2 pathway in ovarian cancer cells. Cell Physiol Biochem. inhibition and apoptosis in human bladder cancer T24 cells. Biochem
2018;45(6):2548–59. Biophys Res Commun. 2012;419(4):741–7.
888. Liu T, Zhao L, Zhang Y, Chen W, Liu D, Hou H, et al. Ginsenoside 20(S)- 908. Hagelgans A, Nacke B, Zamaraeva M, Siegert G, Menschikowski M. Silib-
Rg3 targets HIF-1alpha to block hypoxia-induced epithelial–mesenchy- inin down-regulates expression of secreted phospholipase A2 enzymes
mal transition in ovarian cancer cells. PLoS ONE. 2014;9(9):e103887. in cancer cells. Anticancer Res. 2014;34(4):1723–9.
889. Ge X, Zhen F, Yang B, Yang X, Cai J, Zhang C, et al. Ginsenoside Rg3 909. Kumar S, Raina K, Agarwal C, Agarwal R. Silibinin strongly inhibits
enhances radiosensitization of hypoxic oesophageal cancer cell lines the growth kinetics of colon cancer stem cell-enriched spheroids by
through vascular endothelial growth factor and hypoxia inducible fac- modulating interleukin 4/6-mediated survival signals. Oncotarget.
tor 1alpha. J Int Med Res. 2014;42(3):628–40. 2014;5(13):4972–89.
890. Luo Y, Zhang P, Zeng HQ, Lou SF, Wang DX. Ginsenoside Rg3 induces 910. Kauntz H, Bousserouel S, Gosse F, Marescaux J, Raul F. Silibinin, a
apoptosis in human multiple myeloma cells via the activation of Bcl- natural flavonoid, modulates the early expression of chemoprevention
2-associated X protein. Mol Med Rep. 2015;12(3):3557–62. biomarkers in a preclinical model of colon carcinogenesis. Int J Oncol.
891. Choi YJ, Lee HJ, Kang DW, Han IH, Choi BK, Cho WH. Ginsenoside Rg3 2012;41(3):849–54.
induces apoptosis in the U87MG human glioblastoma cell line through 911. Subramaniam A, Shanmugam MK, Ong TH, Li F, Perumal E, Chen L,
the MEK signaling pathway and reactive oxygen species. Oncol Rep. et al. Emodin inhibits growth and induces apoptosis in an orthotopic
2013;30(3):1362–70. hepatocellular carcinoma model by blocking activation of STAT3. Br J
892. Aziz F, Wang X, Liu J, Yan Q. Ginsenoside Rg3 induces FUT4-mediated Pharmacol. 2013;170(4):807–21.
apoptosis in H. pylori CagA-treated gastric cancer cells by regulating 912. Cheng X, Shi W, Zhao C, Zhang D, Liang P, Wang G, et al. Triptolide sensi-
SP1 and HSF1 expressions. Toxicol In Vitro. 2016;31:158–66. tizes human breast cancer cells to tumor necrosis factoralphainduced
893. Guo JQ, Zheng QH, Chen H, Chen L, Xu JB, Chen MY, et al. Ginsenoside apoptosis by inhibiting activation of the nuclear factorkappaB pathway.
Rg3 inhibition of vasculogenic mimicry in pancreatic cancer through Mol Med Rep. 2016;13(4):3257–64.
downregulation of VEcadherin/EphA2/MMP9/MMP2 expression. Int J 913. Jiang C, Fang X, Zhang H, Wang X, Li M, Jiang W, et al. Trip-
Oncol. 2014;45(3):1065–72. tolide inhibits the growth of osteosarcoma by regulating
Luo et al. Chin Med (2019) 14:48 Page 57 of 58
microRNA-181a via targeting PTEN gene in vivo and vitro. Tumour Biol. 933. Huang ST, Huang CC, Huang WL, Lin TK, Liao PL, Wang PW, et al.
2017;39(4):1010428317697556. Tanshinone IIA induces intrinsic apoptosis in osteosarcoma cells both
914. Li W, Liu BD, Liao K, Liu Y, Wan ZJ, Dong YF, et al. Alteration of androgen in vivo and in vitro associated with mitochondrial dysfunction. Sci Rep.
receptor protein stability by triptolide in LNCaP Cells. Medicina (Kaunas, 2017;7:40382.
Lithuania). 2018;54:3. 934. Ding L, Wang S, Qu X, Wang J. Tanshinone IIA sensitizes oral squamous
915. Meng G, Wang W, Chai K, Yang S, Li F, Jiang K. Combination treatment cell carcinoma to radiation due to an enhanced autophagy. Environ
with triptolide and hydroxycamptothecin synergistically enhances Toxicol Pharmacol. 2016;46:264–9.
apoptosis in A549 lung adenocarcinoma cells through PP2A- 935. Zhang Y, Li S, He H, Han Q, Wang B, Zhu Y. Influence of Tanshinone IIA
regulated ERK, p38 MAPKs and Akt signaling pathways. Int J Oncol. on apoptosis of human esophageal carcinoma Eca-109 cells and its
2015;46(3):1007–17. molecular mechanism. Thorac Cancer. 2017;8(4):296–303.
916. Qiao Z, He M, He MU, Li W, Wang X, Wang Y, et al. Synergistic antitumor 936. Ji ZHE, Tang Q, Zhang J, Yang YAN, Liu Y, Pan Y. Oridonin-induced apop-
activity of gemcitabine combined with triptolide in pancreatic cancer tosis in SW620 human colorectal adenocarcinoma cells. Oncol Lett.
cells. Oncol Lett. 2016;11(5):3527–33. 2011;2(6):1303–7.
917. Sangwan V, Banerjee S, Jensen KM, Chen Z, Chugh R, Dudeja V, 937. Jiang P, Jin H, Jiang J, Yang F, Cai H, Yang P, et al. Single molecule force
et al. Primary and liver metastasis-derived cell lines from KrasG12D; spectroscopy for in situ probing oridonin inhibited ROS-mediated
Trp53R172H; Pdx-1 Cre animals undergo apoptosis in response to EGF-EGFR interactions in living KYSE-150 cells. Pharmacol Res.
triptolide. Pancreas. 2015;44(4):583–9. 2017;119:479–89.
918. Tamgue O, Lei M. Triptolide promotes senescence of prostate cancer 938. Kwan HY, Yang Z, Fong WF, Hu YM, Yu ZL, Hsiao WL. The anticancer
cells through histone methylation and heterochromatin formation. effect of oridonin is mediated by fatty acid synthase suppression in
Asian Pac J Cancer Prevent. 2017;18(9):2519–26. human colorectal cancer cells. J Gastroenterol. 2013;48(2):182–92.
919. Wen L, Chen Y, Zeng LL, Zhao F, Li R, Liu Y, et al. Triptolide induces cell- 939. Li X, Li X, Wang J, Ye Z, Li JC. Oridonin up-regulates expression of P21
cycle arrest and apoptosis of human multiple myeloma cells in vitro and induces autophagy and apoptosis in human prostate cancer cells.
via altering expression of histone demethylase LSD1 and JMJD2B. Acta Int J Biol Sci. 2012;8(6):901–12.
Pharmacol Sin. 2012;33(1):109–19. 940. Liu JB, Yue JY. Preliminary study on the mechanism of oridonin-induced
920. Xie CQ, Zhou P, Zuo J, Li X, Chen Y, Chen JW. Triptolide exerts pro- apoptosis in human squamous cell oesophageal carcinoma cell line
apoptotic and cell cycle arrest activity on drug-resistant human lung EC9706. J Int Med Res. 2014;42(4):984–92.
cancer A549/Taxol cells via modulation of MAPK and PI3K/Akt signaling 941. Ming M, Sun FY, Zhang WT, Liu JK. Therapeutic effect of oridonin on
pathways. Oncol Lett. 2016;12(5):3586–90. mice with prostate cancer. Asian Pac J Trop Med. 2016;9(2):184–7.
921. Yan X, Ke XX, Zhao H, Huang M, Hu R, Cui H. Triptolide inhibits cell pro- 942. Shi M, Ren X, Wang X, Wang H, Liu G, Yuan X, et al. A novel combination
liferation and tumorigenicity of human neuroblastoma cells. Mol Med of oridonin and valproic acid in enhancement of apoptosis induction of
Rep. 2015;11(2):791–6. HL-60 leukemia cells. Int J Oncol. 2016;48(2):734–46.
922. Yang Y, Zhang LJ, Bai XG, Xu HJ, Jin ZL, Ding M. Synergistic antitumour 943. Weng H, Huang H, Dong B, Zhao P, Zhou H, Qu L. Inhibition of miR-17
effects of triptolide plus gemcitabine in bladder cancer. Biomed Phar- and miR-20a by oridonin triggers apoptosis and reverses chemoresist-
macother. 2018;106:1307–16. ance by derepressing BIM-S. Cancer Res. 2014;74(16):4409–19.
923. Zhao X, Zhang Q, Chen L. Triptolide induces the cell apoptosis 944. Yang I-H, Shin J-A, Lee K-E, Kim J, Cho N-P, Cho S-D. Oridonin induces
of osteosarcoma cells through the TRAIL pathway. Oncol Rep. apoptosis in human oral cancer cells via phosphorylation of histone
2016;36(3):1499–505. H2AX. Eur J Oral Sci. 2017;125(6):438–43.
924. Pandey P, Sayyed U, Tiwari RK, Siddiqui MH, Pathak N, Bajpai P. 945. Zheng M, Zhu Z, Zhao Y, Yao D, Wu M, Sun G. Oridonin promotes
Hesperidin induces ROS-Mediated apoptosis along with cell cycle G2/M arrest in A549 cells by facilitating ATM activation. Mol Med Rep.
arrest at G2/M phase in human gall bladder carcinoma. Nutr Cancer. 2017;15(1):375–9.
2019;71(4):676–87. 946. Ye LH, Li WJ, Jiang XQ, Chen YL, Tao SX, Qian WL, et al. Study on the
925. Zhu J, Wang H, Chen F, Lv H, Xu Z, Fu J, et al. Triptolide enhances autophagy of prostate cancer PC-3 cells induced by oridonin. Anatomi-
chemotherapeutic efficacy of antitumor drugs in non-small-cell lung cal Record (Hoboken, NJ: 2007). 2012;295(3):417–22.
cancer cells by inhibiting Nrf2-ARE activity. Toxicol Appl Pharmacol. 947. Han X, Kang KA, Piao MJ, Zhen AX, Hyun YJ, Kim HM, et al. Shikonin
2018;358:1–9. exerts cytotoxic effects in human colon cancers by inducing apoptotic
926. Cai F, Zhang L, Xiao X, Duan C, Huang Q, Fan C, et al. Cucurbitacin cell death via the endoplasmic reticulum and mitochondria-mediated
B reverses multidrug resistance by targeting CIP2A to reactivate pathways. Biomol Ther. 2019;27(1):41–7.
protein phosphatase 2A in MCF-7/adriamycin cells. Oncol Rep. 948. Liang W, Cai A, Chen G, Xi H, Wu X, Cui J, et al. Shikonin induces
2016;36(2):1180–6. mitochondria-mediated apoptosis and enhances chemotherapeutic
927. Dakeng S, Duangmano S, Jiratchariyakul W, Bogler O, Patmasiriwat P. sensitivity of gastric cancer through reactive oxygen species. Sci Rep.
Inhibition of Wnt signaling by cucurbitacin B in breast cancer cells: 2016;6:38267.
reduction of Wnt-associated proteins and reduced translocation of 949. Jeung YJ, Kim HG, Ahn J, Lee HJ, Lee SB, Won M, et al. Shikonin
galectin-3-mediated beta-catenin to the nucleus. J Cell Biochem. induces apoptosis of lung cancer cells via activation of FOXO3a/
2012;113(1):49–60. EGR1/SIRT1 signaling antagonized by p300. Biochem Biophys Acta.
928. Duangmano S, Sae-Lim P, Suksamrarn A, Domann FE, Patmasiriwat P. 2016;1863(11):2584–93.
Cucurbitacin B inhibits human breast cancer cell proliferation through 950. Jang SY, Hong D, Jeong SY, Kim JH. Shikonin causes apoptosis by
disruption of microtubule polymerization and nucleophosmin/B23 up-regulating p73 and down-regulating ICBP90 in human cancer cells.
translocation. BMC Complement Altern Med. 2012;12:185. Biochem Biophys Res Commun. 2015;465(1):71–6.
929. Guo J, Wu G, Bao J, Hao W, Lu J, Chen X. Cucurbitacin B induced ATM- 951. Wiench B, Eichhorn T, Paulsen M, Efferth T. Shikonin directly targets
mediated DNA damage causes G2/M cell cycle arrest in a ROS-depend- mitochondria and causes mitochondrial dysfunction in cancer cells.
ent manner. PLoS ONE. 2014;9(2):e88140. Evid Based Complement Altern Med. 2012;2012:15.
930. Gupta P, Srivastava SK. Inhibition of Integrin-HER2 signaling by Cucur- 952. Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X. Shikonin and its analogs
bitacin B leads to in vitro and in vivo breast tumor growth suppression. inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2.
Oncotarget. 2014;5(7):1812–28. Oncogene. 2011;30(42):4297–306.
931. Promkan M, Dakeng S, Chakrabarty S, Bogler O, Patmasiriwat P. The 953. Lee PN, Ho WS. Antiproliferative activity of gambogic acid isolated
effectiveness of cucurbitacin B in BRCA1 defective breast cancer cells. from Garcinia hanburyi in Hep3B and Huh7 cancer cells. Oncol Rep.
PLoS ONE. 2013;8(2):e55732. 2013;29(5):1744–50.
932. Chen GY, Shu YC, Chuang DY, Wang YC. Inflammatory and apoptotic 954. Zhang H, Lei Y, Yuan P, Li L, Luo C, Gao R, et al. ROS-mediated autophagy
regulatory activity of tanshinone IIA in Helicobacter pylori-infected cells. induced by dysregulation of lipid metabolism plays a protective role
Am J Chin Med. 2016;44(6):1187–206. in colorectal cancer cells treated with gambogic acid. PLoS ONE.
2014;9(5):e96418.
Luo et al. Chin Med (2019) 14:48 Page 58 of 58
955. Wei J, Yang P, Li W, He F, Zeng S, Zhang T, et al. Gambogic acid potenti- 974. Huynh DL, Kwon T, Zhang JJ, Sharma N, Gera M, Ghosh M, et al.
ates the chemosensitivity of colorectal cancer cells to 5-fluorouracil Wogonin suppresses stem cell-like traits of CD133 positive osteosar-
by inhibiting proliferation and inducing apoptosis. Exp Ther Med. coma cell via inhibiting matrix metallopeptidase-9 expression. BMC
2017;13(2):662–8. Complement Altern Med. 2017;17(1):304.
956. Gao G, Bian Y, Qian H, Yang M, Hu J, Li L, et al. Gambogic acid regulates 975. Huang B, Liu H, Huang D, Mao X, Hu X, Jiang C, et al. Apoptosis induc-
the migration and invasion of colorectal cancer via microRNA-21-me- tion and imaging of cadmium-telluride quantum dots with wogonin
diated activation of phosphatase and tensin homolog. Exp Ther Med. in multidrug-resistant leukemia K562/A02 cell. J Nanosci Nanotechnol.
2018;16(3):1758–65. 2016;16(3):2499–503.
957. Pan H, Lu LY, Wang XQ, Li BX, Kelly K, Lin HS. Gambogic acid induces 976. Zhao K, Wei L, Hui H, Dai Q, You QD, Guo QL, et al. Wogonin suppresses
cell apoptosis and inhibits MAPK pathway in PTEN(−/−)/p53(−/−) melanoma cell B16-F10 invasion and migration by inhibiting Ras-
prostate cancer cells in vitro and ex vivo. Chin J Integr Med. medicated pathways. PLoS ONE. 2014;9(9):e106458.
2018;24(2):109–16. 977. Wang Z, Li Y, Zhou F, Piao Z, Hao J. beta-elemene enhances anticancer
958. Wang Q, Wei J, Wang C, Zhang T, Huang D, Wei F, et al. Gambogic acid bone neoplasms efficacy of paclitaxel through regulation of GPR124 in
reverses oxaliplatin resistance in colorectal cancer by increasing intra- bone neoplasms cells. Oncol Lett. 2018;16(2):2143–50.
cellular platinum levels. Oncol Lett. 2018;16(2):2366–72. 978. Ding L, Zhang G, Hou Y, Chen J, Yin Y. Elemene inhibits osteosarcoma
959. Youns M, ElKhoely A, Kamel R. The growth inhibitory effect of gambogic growth by suppressing the reninangiotensin system signaling pathway.
acid on pancreatic cancer cells. Naunyn-Schmiedeberg’s Arch Pharma- Mol Med Rep. 2018;17(1):1022–30.
col. 2018;391(5):551–60. 979. Li P, Zhou X, Sun W, Sheng W, Tu Y, Yu Y, et al. Elemene induces
960. Zhu M, Wang M, Jiang Y, Wu H, Lu G, Shi W, et al. Gambogic acid apoptosis of human gastric cancer cell line BGC-823 via extracellular
induces apoptosis of non-small cell lung cancer (NSCLC) cells by sup- signal-regulated kinase (ERK) 1/2 signaling pathway. Med Sci Monit.
pressing notch signaling. Med Sci Monit. 2018;24:7146–51. 2017;23:809–17.
961. Zhao K, Zhang S, Song X, Yao Y, Zhou Y, You Q, et al. Gambogic acid sup- 980. Zhou J, He L-L, Ding X-F, Yuan Q-Q, Zhang J-X, Liu S-C, et al. Combinato-
presses cancer invasion and migration by inhibiting TGFbeta1-induced rial antitumor effect of rapamycin and β-elemene in follicular thyroid
epithelial-to-mesenchymal transition. Oncotarget. 2017;8(16):27120–36. cancer cells. Biomed Res Int. 2016;2016:8.
962. Wang S, Shao M, Zhong Z, Wang A, Cao J, Lu Y, et al. Co-delivery of 981. Zhao S, Wu J, Zheng F, Tang Q, Yang L, Li L, et al. beta-elemene inhibited
gambogic acid and TRAIL plasmid by hyaluronic acid grafted PEI-PLGA expression of DNA methyltransferase 1 through activation of ERK1/2
nanoparticles for the treatment of triple negative breast cancer. Drug and AMPKalpha signalling pathways in human lung cancer cells: the
Delivery. 2017;24(1):1791–800. role of Sp1. J Cell Mol Med. 2015;19(3):630–41.
963. Tang Q, Lu M, Zhou H, Chen D, Liu L. Gambogic acid inhibits the 982. Liu JS, Che XM, Chang S, Qiu GL, He SC, Fan L, et al. beta-elemene
growth of ovarian cancer tumors by regulating p65 activity. Oncol Lett. enhances the radiosensitivity of gastric cancer cells by inhibiting Pak1
2017;13(1):384–8. activation. World J Gastroenterol. 2015;21(34):9945–56.
964. Huang J, Zhu X, Wang H, Han S, Liu L, Xie Y, et al. Role of gambogic acid 983. Zhang J, Zhang H, Chen L, Sun DW, Mao C, Chen W, et al. beta-elemene
and NaI(131) in A549/DDP cells. Oncol Lett. 2017;13(1):37–44. reverses chemoresistance of breast cancer via regulating MDR-related
965. Thida M, Kim DW, Tran TTT, Pham MQ, Lee H, Kim I, et al. Gambogic acid microRNA expression. Cell Physiol Biochem. 2014;34(6):2027–37.
induces apoptotic cell death in T98G glioma cells. Bioorg Med Chem 984. Liu JS, He SC, Zhang ZL, Chen R, Fan L, Qiu GL, et al. Anticancer effects
Lett. 2016;26(3):1097–101. of beta-elemene in gastric cancer cells and its potential underlying
966. Wen C, Huang L, Chen J, Lin M, Li W, Lu B, et al. Gambogic acid inhibits proteins: a proteomic study. Oncol Rep. 2014;32(6):2635–47.
growth, induces apoptosis, and overcomes drug resistance in human 985. Guan C, Liu W, Yue Y, Jin H, Wang X, Wang XJ. Inhibitory effect of
colorectal cancer cells. Int J Oncol. 2015;47(5):1663–71. beta-elemene on human breast cancer cells. Int J Clin Exp Pathol.
967. Xu N, Zhou X, Wang S, Xu LL, Zhou HS, Liu XL. Artesunate induces 2014;7(7):3948–56.
SKM-1 cells apoptosis by inhibiting hyperactive beta-catenin signaling 986. Li QQ, Wang G, Liang H, Li JM, Huang F, Agarwal PK, et al. beta-Elemene
pathway. Int J Med Sci. 2015;12(6):524–9. promotes cisplatin-induced cell death in human bladder cancer and
968. Verma S, Das P, Kumar VL. Chemoprevention by artesunate in a other carcinomas. Anticancer Res. 2013;33(4):1421–8.
preclinical model of colorectal cancer involves down regulation of 987. Li QQ, Wang G, Huang F, Li JM, Cuff CF, Reed E. Sensitization of lung
beta-catenin, suppression of angiogenesis, cellular proliferation and cancer cells to cisplatin by beta-elemene is mediated through block-
induction of apoptosis. Chem Biol Interact. 2017;278:84–91. ade of cell cycle progression: antitumor efficacies of beta-elemene
969. Ilamathi M, Santhosh S, Sivaramakrishnan V. Artesunate as an anti- and its synthetic analogs. Med Oncol (Northwood, London, England).
cancer agent targets Stat-3 and favorably suppresses hepatocellular 2013;30(1):488.
carcinoma. Curr Top Med Chem. 2016;16(22):2453–63. 988. Shahriyar SA, Woo SM, Seo SU, Min KJ, Kwon TK. Cepharanthine
970. Eling N, Reuter L, Hazin J, Hamacher-Brady A, Brady NR. Identification enhances TRAIL-mediated apoptosis through STAMBPL1-mediated
of artesunate as a specific activator of ferroptosis in pancreatic cancer downregulation of survivin expression in renal carcinoma cells. Int J
cells. Oncoscience. 2015;2(5):517–32. Mol Sci. 2018;19(10):3280.
971. Raza A, Ghoshal A, Chockalingam S, Ghosh SS. Connexin-43 enhances
tumor suppressing activity of artesunate via gap junction-dependent
as well as independent pathways in human breast cancer cells. Sci Rep. Publisher’s Note
2017;7(1):7580. Springer Nature remains neutral with regard to jurisdictional claims in pub-
972. Go JH, Wei JD, Park JI, Ahn KS, Kim JH. Wogonin suppresses the LPSen- lished maps and institutional affiliations.
hanced invasiveness of MDAMB231 breast cancer cells by inhibiting the
5LO/BLT2 cascade. Int J Mol Med. 2018;42(4):1899–908.
973. Xu X, Zhang X, Zhang Y, Yang L, Liu Y, Huang S, et al. Wogonin reversed
resistant human myelogenous leukemia cells via inhibiting Nrf2 signal-
ing by Stat3/NF-kappaB inactivation. Sci Rep. 2017;7:39950.