You are on page 1of 13

Biomedicine & Pharmacotherapy 117 (2019) 109142

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Natural products for treating colorectal cancer: A mechanistic review T


a,1 b,1 a a a,⁎ b,⁎
Xuan-mei Huang , Zhi-jie Yang , Qing Xie , Zi-kang Zhang , Hua Zhang , Jun-ying Ma
a
Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Laboratory Medicine, Marine Biomedical Research Institute, Guangdong Medical
University, Dongguan 523808, China
b
CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, RNAM Center for Marine Microbiology,
South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China

A R T I C LE I N FO A B S T R A C T

Keywords: Colorectal cancer (CRC) is one of the most common types of cancers in humans and is closely linked to the global
Natural products cancer-related mortalities worldwide. Molecular pathological epidemiology studies can reveal the risk factors of
Colorectal cancer CRC and contribute to biomarker research and precision medicine. The current clinical treatment for CRC mainly
Anti-cancer effects involves surgery and chemotherapy. However, because of the occurrence of side effects and the emergence of
Molecular mechanisms
drug resistance, there is an urgent need to find new and more effective drugs for CRC treatment. An increasing
Regulation networks
number of studies have demonstrated that many natural products possess effective anti-CRC effects and may
serve as alternative chemotherapy agents for CRC treatment. In this review, we summarize the natural products
with anti-CRC effects from different sources based on the chemical structures such as alkaloids, polysaccharides,
polyphenols, terpenoid, unsaturated fatty acids, and discuss the natural product-derived drugs used clinically for
colorectal cancer treatment. Furthermore, natural products of marine origin are also discussed for their en-
ormous potential to serve as the candidate drugs. Notably, we generalize the experiment-based molecular me-
chanisms and the regulatory networks whereby natural products exert anticancer effects on cell proliferation,
metastasis, apoptosis, autophagy, and angiogenesis.

1. Introduction loss has been identified as a promising prognostic marker for CRC pa-
tients due to its significant association with worse clinical outcomes,
Colorectal cancer (CRC) is the fourth leading cause of malignant chemoresistance, and reduced immune infiltration [7]. Previous studies
tumor-related deaths worldwide [1]. According to data from GLOBO- have shown that smoking and obesity are risk factors for CRC; however,
CAN in 2012, there are 1.6 million new cases of CRC and 694,000 MPE studies have revealed that smoking is a risk factor for the CpG
people that die from CRC every year [1]. The occurrence and pro- island methylator phenotype-high or MSI-high CRC subtype, while
gression of colorectal cancer are caused by a combination of multiple obesity is a risk factor for the non-MSI-high CRC subtype [5,8,9]. The
factors, among which age, family history, gender, region, and personal dietary pattern is also an important factor in CRC. It has been found that
history are the major risk factors [2–4]. calcium intake is inversely proportional to the risk of CRC; calcium
Recently, molecular pathological epidemiology (MPE), a transdis- functions in the immune prevention of CRC by regulating T cell func-
ciplinary research field that reveals the relationship between the risk tion [10–12]. However, alcohol intake has been found to increase the
factors (germline genetics, lifestyle, dietary pattern, microbiome, etc.) risk of the KRAS-wild/BRAF-wild and KRAS mutated CRC subtypes
and molecular characteristics, occurrence, development and prognosis [13]. Fusobacterium nucleatum is a pathogenic microorganism involved
of diseases, has been predominantly applied for epidemiological studies in the development and prognosis of CRC [14,15]. Intensive me-
on cancer to provide new insights into, and strategies for, cancer pre- chanism-based studies have shown that it plays a carcinogenic role by
vention and treatment. Genetic changes including microsatellite in- regulating signaling pathways such as the E-cadherin/β-catenin
stability (MSI), BRAF mutations, PIK3CA mutations, KRAS mutations pathway and is involved in the metastasis of CRC [16,17]. MPE studies
and high-degree CpG island methylator phenotype are the main factors have also revealed that aspirin could reduce the risk and patient mor-
closely related to the development of CRC [5,6]. More recently, SMAD4 tality of the PTGS2-positive CRC subtype but not the PTGS2-negative


Corresponding authors.
E-mail addresses: 1837766853@qq.com (X.-m. Huang), yzjie6@126.com (Z.-j. Yang), 1048271414@qq.com (Q. Xie), 1119129766@qq.com (Z.-k. Zhang),
huazhang@gdmu.edu.cn (H. Zhang), majunying@scsio.ac.cn (J.-y. Ma).
1
These authors contributed equally.

https://doi.org/10.1016/j.biopha.2019.109142
Received 20 March 2019; Received in revised form 14 June 2019; Accepted 14 June 2019
0753-3322/ © 2019 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

CRC subtype [18]. In addition, compared to the PI3KCA-wild subtype, endothelial growth factor (VEGF) and hypoxia-inducible factor 1 alpha
aspirin has been verified to exert strong anticancer effect against the expression. SN38 has dual inhibitory effects, including the suppression
PI3KCA-mutated CRC subtype [19,20]. There is no doubt that the ap- of endothelial cell proliferation or lumen formation, as well as the in-
plication of MPE for studying cancer will provide a more precise and hibition of the angiogenic activity of glioma cells. Kamiyama H has
effective strategy for the prevention and treatment of CRC. reported that SN38 can selectively inhibit three-dimensional tubular
The conventional treatments for CRC include surgery and che- structure formation and endothelial cell proliferation at the con-
motherapy. Chemotherapeutics can lead to cancer cell death by indu- centration of 0.01 μM [45].
cing DNA damage or initiating multiple signaling pathways, including Although CPT-11 and SN38 have broad-spectrum anti-tumor ac-
cell cycle arrest, inhibition of global translation, DNA repair, etc. [21]. tivity against various tumors, CPT-11 and its analogs have significant
However, as many studies, including MPE studies, have shown, the toxicity including cholinergic syndrome, neutropenia and delayed
treatment outcome of chemotherapeutic drugs in CRC patients varies diarrhea [46–48]. An increasing number of studies have been carried
with the subtype of cancer. The effects of cytotoxicity, drug resistance out in recent years to improve the efficacy and safety of these com-
and adverse reactions are the main problems associated with che- pounds through chemical structure modification and combination
motherapy. chemotherapy [44].
Complex and diverse natural products have yielded numerous ef-
fective lead compounds for the discovery of new drugs. Many natural 2.1.2. Polysaccharides
products have exhibited excellent biological activities against in- PSK (Krestin) (Fig. 1C) is protein-bound polysaccharide K isolated
flammation, viruses, bacteria, tumors, etc. [22,23]. The application of from Coriolus versicolor; it has a molecular weight of approximately
natural product-derived drugs has made a tremendous contribution to 100 kDa and a protein content of 18–38% [49]. Tsutsumi et al. have
human health, for example, penicillin for the treatment of infectious found that PSK effectively reduced the mortality induced associated
bacterial diseases [24], streptomycin for the long-term treatment of with dextran sodium sulfate-induced colitis and inhibited the devel-
tuberculosis [25], the significant anti-inflammatory plant-derived opment of CRC, suggesting that it shows dual antagonism against both
monoterpene glycoside, paeoniflorin [26], the natural plant-derived inflammation and CRC [50]. The efficacy of PSK against cancer is
anti-HIV compound, patentiflorin A [27], cyclosporin, which has im- mediated via several pathways, including recovery from im-
munomodulatory effects [28], lovastatin, which has with hypolipi- munosuppression that is induced by TGF-β and anti-tumor immune
demic function [29] and ecteinascidin-743, which has an anti-tumor responses, and enhanced efficacy of chemotherapy by inducing apop-
activity [30]. tosis and inhibiting the metastasis of cancer cells [49].
Natural products that are well tolerated and have less toxicity will Fucoidan (Fig. 1D) is a sulfated polysaccharide isolated from brown
help patients to achieve better treatment outcomes and improve their seaweeds; it has been shown to exhibit a wide range of biological ac-
quality of life. Many chemotherapeutic agents have been identified by tivities [51]. A previous study has indicated that fucoidan induces
investigating potential compounds from plants, animals, and micro- apoptosis in HT-29 and HCT116 cells [52]. Double-blind, randomized
organisms including marine organisms, and these have been found to clinical trials have demonstrated that low-molecular-weight fucoidan
exert anti-cancer effects against a variety of tumors [31–35]. Accumu- has significant activity against CRC in humans, with relatively few side
lating evidences from previous studies have shown that natural pro- effects [53]
duct-derived drugs have broad application prospects for the treatment
of CRC [36–38]. 2.1.3. Polyphenols
Resveratrol (Fig. 1E) is a polyphenolic compound synthesized by
2. Natural products serving as drugs against CRC in clinical plants following attack by pathogens, such as bacteria or fungi; peanuts,
setting grapes, and red wine are highly rich in resveratrol. Evidence from
previous studies has suggested that resveratrol participates in the reg-
2.1. Natural products as anti-CRC drugs ulation of the average life cycle of organisms as the silent information
regulation 2 homolog 1 (SIRT1), and it has significant inhibitory effects
Natural products have become prolific sources of new anticancer against hepatocellular carcinoma, breast cancer, gastric cancer, leu-
drugs, and approximately 50% of the currently used anticancer drugs kemia, and other tumor cells [54–58]. Previous research has shown that
have been directly or indirectly derived from natural products; these resveratrol exerts apoptotic and anti-proliferative effects on CRC via the
products, including alkaloids, polysaccharides, polyphenols, diterpe- induction of Fas redistribution and inhibition of Wnt signaling [59].
noid and unsaturated fatty acids possess various structures [39]. The Engelbrecht et al. have reported that grape seed extract could inhibit
continuous discovery of such products has opened up a novel direction the PI3K pathway, to induce apoptosis and suppress the viability of
for the prevention and therapy of cancers; the natural product-based CaCo-2 cells [60].
drugs used clinically for CRC treatment are summarized in Fig. 1 and Curcumin (Fig. 1F), a polyphenol isolated from Curcuma longa, has
Table 1. been shown to have significant anti-angiogenic properties, and can
reduce the secretory level of TNF-α in the serum, enhance the expres-
2.1.1. Alkaloids sion of the p53 and cip1/p21, and induce the apoptosis of CRC cells
Irinotecan (CPT-11) (Fig. 1A), a water-soluble camptothecin deri- with little effect on the quality of life of the patients [61]. Watson et al.
vative, functions as a DNA topoisomerase I inhibitor that blocks DNA have found that curcumin induced p21-independent apoptosis in
replication and inhibits RNA synthesis [40]. Although it has been used HCT116 cells [62].
for first-line therapy or second-line therapy after the failure of fluor-
ouracil treatment, CPT-11 has been shown to have an interesting anti- 2.1.4. Terpenoids
tumor activity against metastatic colorectal cancer (mCRC). CPT-11 has Andrographolide (Fig. 1G) is a diterpenoid and the major bioactive
been utilized for standard treatment against CRC worldwide, and clin- component of the plant Andrographis paniculata, which has been used as
ical studies have reported that CPT-11 has better anti-tumor effect a traditional herbal medicine in Asia for thousands of years. Andro-
when combinated with 5-fluorouracil (5-FU)/formyltetrahydrofolate grapholide has been reported to suppress NF-κB activation and DNA-
(CF) for the treatment of advanced mCRC [41–43]. binding activity [63,64], and it thus, has various biological activities,
SN38 (Fig. 1B), the active analog of irinotecan, has an inhibitory such as anti-inflammatory functions [65,66] and anti-cancer properties
effect against the growth of a variety of malignant cells [44]. The anti- [67,68]. Combination treatment of andrographolide and 5-FU can in-
angiogenic activity of SN38 is mainly due to the inhibition of vascular duce the apoptosis of HCT116 cells via the binding of andrographolide

2
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

Fig. 1. Structures of previously discovered natural products drugs used in colorectal cancer.

to the BAX protein and the upregulation of BAX expression [69]. Su treatment had failed in the early stage of the disease. As a natural
et al. have evidenced the safety and efficacy of the combination of product-derived drug, irinotecan remains the backbone of combination
andrographolides and capecitabine for the treatment of elderly patients chemotherapies for metastatic CRC, including FOLFIRI (levoleucovorin
with locally advanced CRC [70]. (LV) and 5-FU plus irinotecan), IROX (irinotecan plus oxaliplatin), and
XELIRI (capecitabine plus irinotecan).
2.1.5. Unsaturated fatty acids
Marine microalgae are a rich source of polyunsaturated fatty acids
2.2.1. FOLFIRI
(PUFAs) (> 80%), mainly eicosapentaenoic acid (EPA) and doc-
FOLFIRI, a combination therapy consisting of levoleucovorin, 5-FU,
osahexaenoic acid (DHA) [71]. EPA and DHA inhibit important an-
and irinotecan, is one of the current standard chemotherapy regimens
giogenic factors, such as platelet growth factor, VEGF, and endothelial
for advanced CRC. In the study by Tournigand et al, the objective re-
cell growth factor, and are thus, effective against CRC, breast cancer,
sponse rate obtained with FOLFIRI as the first-line therapy (consisting
and pre-existing adenocarcinoma [72]. DHA and EPA (in the form of
of 200 mg/m2 l-LV or 400 mg/m2 dl-LV as a 2 h infusion, and 180 mg/
acid and phospholipids) have been shown to inhibit the growth of CRC
m2 irinotecan administered as a 90-minute infusion in 500 mL of 5%
cells, and PUFAs have been shown to have stronger growth-inhibitory
dextrose, followed by 400 mg/m2 FU and a 46 -h infusion of 2400 mg/
effects in the HT-29 cell line than in the Caco-2 and DLD-1 cell lines
m2 FU for two cycles, with the dose being increased to 3000 mg/m2
[73].
from cycle 3 in case of no toxicity, repeated every 2 weeks) was 56%.
Sastre J et al. have administered the FOLFIRI regimen to patients with
2.2. Application of natural product-based drugs for combination advanced CRC for whom treatment with 5-FU or FOLFOX (oxaliplatin
chemotherapy in clinical setting and fluorouracil plus formyltetrahydrofolate) had failed, and achieved
satisfactory results: 8.3% complete response rate, 20.8% partial effi-
Metastatic CRC is generally considered to be the terminal stage of ciency, and 29% objective efficiency [79]. The side effects of FOLFIRI
CRC [74–77]. In previous clinical trials, irinotecan treatment has been treatment mainly include mucositis, nausea, vomiting, and gastro-
reported to markedly increase the survival rate and reduce the in- intestinal toxicity [80]. Antibody-based drugs, including panitumumab
cidence of adverse events in patients with advanced mCRC [78]. Since and bevacizumab, have been shown to effectively increase the safety
then, irinotecan has been regarded as a standard chemotherapeutic and efficacy of mCRC treatment when administered in combination
drug, especially in patients with advanced mCRC, for whom 5-FU with FOLFIRI [81–83].

Table 1
Clinical anti-colorectal cancer natural products and derivatives.
Natural Products Source Chemical class Mechanism Phase NCT ID References

EGCG Plant Polyphenol Apoptosis Ⅱ 01360320 [162]


SN-38 Plant Alkaloid Anti-Proliferation Ⅱ 00311610 [44]
Resveratrol Plant Polyphenol Apoptosis, Anti-Proliferation Ⅰ 00256334 [163]
Curcumin Plant Polyphenol Apoptosis, Anti-Angiogenesis, Cell cycle arrest Ⅰ 00973869 [164]
PSK Fungi Polysaccharide Apoptosis, Anti-Proliferation Ⅲ 00497107 [49]
Topotecan Plant Alkaloid Anti-Proliferation Ⅱ 00193167 [165]
Metformin Plant Alkaloid Anti-Proliferation, Anti-Metastasis Ⅱ 01941953 [166]
Everolimus Bacterial Macrolide Anti-Angiogenesis, Anti-Metastasis Ⅱ 00419159 [167,168]
Methotrexate Plant Alkaloid Anti-Proliferation, Cell cycle arrest Ⅱ 00952016 [169]
Andrographolide Plant Diterpenoid Apoptosis, Anti-Proliferation, Cell cycle arrest Ⅱ 01993472 [170]
Irofulven Fungi Sesquiterpene Anti-Proliferation Ⅱ 00003786 [171,172]
Epothilone D Bacterial Macrolide Anti-Proliferation Ⅱ 00077259 [173]
Silymarin Plant Flavonoid Apoptosis, Anti-Proliferation Ⅳ 03130634 [174]
Berberine Plant Alkaloid Anti-Proliferation, Cell cycle arrest Ⅱ/Ⅲ 02226185 [175]
Mitomycin C Streptomyces Ethyleneimines Anti-Proliferation, Cell cycle arrest Ⅱ 03073694 [176]

EGCG: Epigallocatechin Gallate; PSK: Polysaccharide-K; DHA: docosahexaenoic acid; PUFAs: Polyunsaturated fatty acids.

3
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

2.2.2. IROX induction of apoptosis and inhibition of cell invasion have been ob-
Oxaliplatin (OXA), a platinum-based drug, forms a cross-link with served following methylferulate treatment [104]. Sulforaphane, an
DNA, thereby antagonizing its replication and transcription [84]. In isothiocyanate, has been found to induce G2/M phase arrest in HCT116
vitro studies have shown that OXA inhibits the growth of cisplatin- and CRC cells by increasing the expression levels of WEE1, CDK2, cyclin A,
carboplatin-resistant colorectal tumor cell lines [85]. The efficacy of and cyclin B, while decreasing the levels of cdc25C and CDK1 [105].
IROX (irinotecan plus OXA) in the treatment of CRC has been supported Tripolinolate A, a novel compound isolated from Tripolium vulgare, has
by numerous clinical phase I/II studies [86–89]. Haller et al. have de- been confirmed to induce apoptosis and G2/M phase arrest in SW620
monstrated that IROX therapy (85 mg/m2 OXA, intravenous infusion cells, and inhibit tumor growth in animal models [106].
for 120 min, and 200 mg/m2 irinotecan, intravenous infusion for Lawsonaringenin, a flavonoid extracted from the leaves of Lawsonia
30 min or 90 min every three weeks) is more effective than the therapy alba Lam, has been shown to trigger S phase arrest and apoptosis via
with irinotecan alone after the failure of 5-FU therapy in advanced CRC multiple pathways [107]. Another study has shown that Ent-kaur-2-
patients [90]. However, compared to the therapy with irinotecan alone, one-16β, 17-dihydroxy-acetone-ketal (DEK), a novel ent-kaurane di-
IROX therapy resulted in a greater incidence of grade 3–4 diarrhea terpenoid isolated from Rubus corchorifolius L. f. leaves, can result in S
(28% versus 23%) and severe neuropathy (5% versus 0%) [91]. The phase arrest by decreasing the CDK2, CDK4, and cyclin D1 levels in
incidence of vomiting, diarrhea, nausea, and febrile neutropenia has HCT116 cells [108]. The extract of Iberis amara (IAE) has been shown to
been found to be significantly higher in patients treated with IROX than exhibit an ability to inhibit cell growth and induce G2/M phase arrest in
in those treated with FOLFOX [92]. In general, IROX is preferred HT-29 cells by decreasing the levels of regulatory proteins, such as
whenever patients are insensitive to 5-FU, indicating that it is a suitable cyclin A2, cyclin D3, CDK2, CDK4, and CDK6 [109]. Other natural
second-line therapy for these advanced CRC patients. compounds that exert similar effects are shown in Table 2.
Since combination therapy is a strategy to overcome chemother-
2.2.3. XELIRI apeutic resistance for CRC treatment, many studies focused on finding
Capecitabine, a highly active oral fluoropyrimidine, is an appealing natural drugs for adjuvant therapy are being performed. OXA, a third-
alternative to 5-FU in the treatment of CRC [93]. Compared with 5-FU/ generation platinum agent, has been used to treat CRC in recent dec-
LV, capecitabine has a higher response rate and better safety [94,95]. ades and has been used in combination with leucovorin and 5-FU
Recent clinical trials have shown that the XELIRI regimen (capecitabine [110–112]. However, resistance to OXA has been eventually reported in
plus irinotecan), as a second-line treatment, exhibited definite efficacy the treatment of mCRC [113,114]. A previous study has shown that co-
and safety for the treatment of patients with advanced CRC [96,97]. In treatment with oxymatrine (OMT) and OXA induced G0/G1 phase ar-
2018, Xu et al. performed a clinical phase III trial involving a modified rest via the upregulation of p27 and p21, and the downregulation of
XELIRI (mXELIRI)-AXEPT regimen (NCT01996306) [98]. Compared to cyclin D; thus, OMT and OXA were combined to explore the efficiency
FOLFIRI, mXELIRI (200 mg/m2 irinotecan administered intravenously of this combination therapy for treating colon cancer [115].
on day 1, plus 800 mg/m2 capecitabine administered orally twice daily
on days 1–14, repeated every 21 days) has been reported to show highly 3.2. Migration- and invasion-based regulation
satisfactory results: the survival time median of patients in the mXELIRI
group was 16.8 months and that of those in the FOLFIRI group was 15.4 Metastasis is one of the most serious features of cancer, and it de-
months, with a lower probability of neutropenia; this indicates that termines the prognosis and clinical stage of the disease [116]. The
mXELIRI can be used as a potential substitute for FOLFIRI, and as the process of metastasis comprises multiple steps, involving adhesion,
standard second-line therapy for mCRC, thus providing a new option migration, invasion of cancer cells, and degradation of the extracellular
for the treatment of mCRC patients [98]. matrix (ECM) surrounding the target organ [117]. Epithelial-me-
senchymal transition (EMT) usually occurs during the progression of
3. Mechanisms underlying the action of potential natural product- various epithelial cell carcinomas and is closely related to the invasion
based drugs against CRC and metastasis of various tumors including CRC [118]. The activation
of signaling pathways, such as the TGF-β/Smad and JAK2/STAT3
In this part of the review, we summarize the most recent and closely pathways, plays a key role during the growth, progression, and me-
related natural products potentially used for CRC treatment. Many tastasis of a tumor [119–121]. Recently, many experimental studies
studies have indicated that natural product-derived drugs can exert have shown that natural products can exert an anti-cancer activity
anticancer effects by acting on metastasis, invasion, apoptosis, and against the metastasis and invasion of CRC (Table 3).
angiogenesis, which are the major hallmarks in human cancers α-Hederin, which is extracted from the leaves of Hedera helix, is a
[99–102]. In recent years, drugs derived from natural products have monodesmosidic triterpenoid saponin that can suppress the IL-6-in-
been widely used for cancer treatment due to their wide range of duced EMT associated with the disruption of JAK2/STAT3 signaling, to
sources, better bioactivities, and low toxicity. Here, we generalize the inhibit the migration and invasion of SW620 cells [122]. Zerumbone, a
potential functions of natural products showing anti-CRC effects and cyclic sesquiterpene from Zingiber zerumbet, has the ability to suppress
provide a mechanism-based foundation for the development of natural the migration of, and trigger cell cycle arrest and apoptosis in, SW480
product-derived drugs against CRC (Fig. 2). cells [123]. Tea polysaccharide (TPS), which is the major bioactive
component in green tea, has been found to inhibit cancer cell pro-
3.1. Proliferation-based regulation liferation and invasion, and increase cancer cell apoptosis partially via
the reduction of the expression of cyclin D1, MMP-2, and MMP-9 both
The cell cycle is tightly controlled by checkpoints to ensure that in vitro and in vivo [124]. Curcumin, which is isolated from turmeric,
cells replicate in sequential and correctly oriented steps; the dysregu- has been shown to suppress metastasis and tumor growth in case of
lation of the cell cycle can cause the abnormal proliferation of cancer both CRC cells and an orthotopic mouse model by regulating signaling
cells. Many natural products have been shown to inhibit cancer cell pathways, including the STAT3, FAK/Src, ErK, and Akt pathways,
proliferation by controlling mitosis and the cell cycle, and are thus, suggesting its promising use for treating mCRC patients [125].
important potential replacements for chemotherapeutic drugs [103].
Methylferulate, which is extracted from Tamarix aucheriana, has 3.3. Apoptosis-based regulation
been confirmed to upregulate the expression of p19, p21, and p27 and
downregulate the levels of CDK1 and CDK2, leading to the induction of Apoptosis is mainly grouped into two pathways: the intrinsic and
S and G2/M phase arrest in SW1116 and SW837 cells. Moreover, the the extrinsic pathway. The intrinsic pathway (mitochondrial pathway)

4
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

Fig. 2. The experiment-based molecular me-


chanisms and regulation networks of natural
products in colorectal cancer. Natural products
are numbered in the white boxes and listed on
the right of the figure. Natural products from
different sources exert anti-colorectal cancer
effects on apoptosis, proliferation, metastasis,
autophagy and angiogenesis by regulating
various proteins and pathways.

Table 2
Natural products with anti-proliferative effect on colorectal cancer cells.
Compound Source Cell line Mechanism Cell cycle References
phase

Lawsonaringenin Plant HT-29 Downregulate β-catenin and c-Myc S [107]


Ribonucleic acid fraction Fungus HT-29 Increase p21 and p27, decrease Cyclin D1 and Cyclin A S [177]
Ent-kaur-2-one-16β,17-dihydroxy-acetone- Plant HCT116 Increase p21, decrease Cyclin D, CDK2 and CDK4 S [108]
ketal
Tripolinolate A Plant SW620 – G2/M [106]
Extracts of Iberis amara Plant HT-29 Decrease Cyclin A2, Cyclin D3, CDK2, CDK4 and CDK6 G2/M [108]
Kahweol Plant HCT116, SW480 Decrease Cyclin D1 – [178]
Indigocarpan Plant LS174T Increase p21, decrease Cyclin B1 and Cyclin D1 G2/M [179]
Sulforaphane Plant HCT116 Increase CDK1, CDK2, Cyclin B and WEE1, decrease cdc 25C and G2/M [105]
Cyclin A
Methylferulate Plant SW1116, SW837 Increase p19 and p27, decrease CDK1 and CDK2 S and G2/M [104]
Yuanhuacine Plant HCT116 Increase p21 G2/M [180]

can trigger the caspase cascade and leads to apoptosis via the release of downregulate the levels of Bcl2 and XIAP, and upregulate the levels of
cytochrome C [126]. The extrinsic pathway initiates the activation of pro-apoptotic factors including cytochrome C, caspase 3/9, and PUMA
apoptosis by triggering cell death receptors (DRs), such as FasL/CD95 L in both HCT116 and HT-29 cells [129]. Quercetrin from Toona sinensis
and TRAIL, on the cell surface [127,128]. leaves (QTL) has been found to promote apoptosis by upregulating
Previous literature has suggested that many natural products exert cytochrome C, Bax, Apaf-1, caspase-3, and caspase-9. In addition, QTL
an anticancer effect that is mediated by the intrinsic apoptotic pathway has also been shown to trigger ROS generation and induce the loss of
(Table 4). Natural products induce apoptosis mainly by inducing the mitochondrial membrane potential in the human CRC cell line SW620
generation of ROS, decreasing the mitochondrial membrane potential [130]. Cardol, which is a major bioactive compound isolated from
and the Bcl2/Bax ratio, and activating the related caspase proteins, Trigona incisa propolis, can activate caspase-3, caspase-9, and PARP,
which ultimately results in DNA fragmentation and apoptosis. Alkyl- and induce ROS generation in SW620 cells [131]. Additionally, 6-bro-
resorcinols (ARs) that were isolated from wheat bran have been found moisatin, which was isolated and identified from an Australian marine
to induce cell cycle arrest and mitochondrial apoptosis by activating gastropod, Dicathais orbita, has been shown to induce apoptosis by in-
and accumulating p53. AR C15 and AR C17 have been found to creasing the expression of caspase-3 and -7 in both HT-29 and Caco-2

Table 3
Natural products with anti-migration and anti-invasion effects on colorectal cancer cells.
Compound Source Cell line Mechanism References

α-Hederin Plant SW620 Suppress EMT and JAK2/STAT3 [122]


Zerumbone Plant SW480 – [123]
Tea Polysaccharide Plant CT26(mouse) Decrease Cyclin D1, MMP-2 and MMP-9 [124]
Curcumin Plant Colon 26-M01 Inhibit FAK/Src, STAT3, Erk and Akt pathways [125]
Esculetin Plant HCT116 Target Axin2/Ecadherin axis [181]
Polysaccharides Plant Colon 26-M3.1 – [182]
Luteolin Plant HCT116 Inhibit CREB1 and block EMT [183]
Cairicoside E Plant SW480, HCT116 Inhibit the EMT through down-regulation of AQP5 and suppression of p-Smad2/3 [184]
Resveratrol Plant HCT116, SW480 Inhibit FAK activity [185]
Ganoderma lucidum polysaccharides Plant LoVo – [186]
Gypenosides Plant SW620 – [187]
Gypenosides Plant SW480 – [188]

5
X.-m. Huang, et al.

Table 4
Natural products inducing apoptosis in colorectal cancer cells.
Compound Source Cell line Mechanism References

Zerumbone Plant SW480 Induce ROS, activate caspase-3/8/9 and downgregulate Bcl2 [123]
Alkylresorcinols Plant HCT116, HT-29 Decrease Bcl2 and XIAP, increase PUMA, cytochrome C, cleaved caspase-9 and -3 [129]
Quercetrin Plant SW620 Upregulate Bax, caspase-9, caspase-3 and Apaf-1 [130]
Pteisolic acid G Plant HCT116 Downregualte NF-κB p65 activity, stimulate p53 and promote ROS production [135]
Oblongifolin C and guttiferone K Plant HCT116 Increase cleavage of caspase-3, PARP and JNK phosphorylation and enhance cellular ROS production [189]
Cardol Plant SW620 Increase caspase-3, caspase-9, cleavage of pro-caspase-3 and pro-caspase-9, PARP and ROS [131]
Cuminaldehyde Plant COLO 205 Activate caspase-3 and -9 [190]
BG-4 Plant HCT116, HT-29 Reduce Bcl-2, increase Bax and caspase-3 [191]
Deoxyelephantopin Plant HCT116 Activate caspase-3 and PARP cleavage [192]
Isoledene Plant HCT116 Increase ROS, caspase-8, -9 and -3, upregulate Bid, Bim and cytochrome C, downregulate Bcl-2, Bcl-W, survivn, [193]
xlAP and HSPs pro-survival proteins

6
Gambogic acid Plant HCT-15 Activation of JNK signaling pathway [194]
Hederagenin Plant LoVo Increase ROS, up regulate Bax and decrease Bcl-2, Bcl-xL, procaspase-9, procaspase-3, PARP and survivin [195]
Goniothalamin Plant LoVo Increase death receptor DR5, decrease cFLIP, cleavage of PARP, caspase-3, caspase-8, caspase-9, Bid, Bax and [142]
Bcl2
Procyanidins Plant SW480 –TRAIL-sensitive and SW620- Activate caspase-8 [139]
TRAIL -resistant
Oplopantriol A Plant HCT116, SW480 Up regulate TNFRSF10A, TNF, TNFSF8, CRADD, FADD, TRADD, caspase-3, -7 and -8 [140]
Maslinic Acid Plant Caco-2 p53-Deficient cells Activate caspase-3 and caspase-8 [196]
Calotroposid A Plant WiDr cells Increase caspase-8 [141]
Propolis cinnamic acid derivatives Plant DLD-1 Regulate TRAIL/DR4/5 and/or FasL/Fas death-signaling loops and increase miR-143 [197]
Mertensene Plant HT29 Activate caspase-3 and PARP cleavage and increase TRADD [143]
2-(Pro-1-ynyl)-5-(5,6-dihydroxypenta-1,3diynyl) Plant SW620 ROS-Mediated JNK Activation [133]
Thiophene
Riccardin D Plant HT-29 Upregulate cleaved caspase-3, -9 and the ratio of Bax/Bcl-2, block the NF-κB signaling pathway [134]
Icariin Plant HCT116, HT-29 Suppress the NF-κB activity [198]
Oxymatrine Plant HT-29, SW480 Activate PI3K/AKT/mTOR pathway [115]
Biomedicine & Pharmacotherapy 117 (2019) 109142
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

Table 5
Natural products regulating autophagy on colorectal cancer cells.
Compound Source Cell line Mechanism References

Thymoquinone Plant Irinotecan-resistant (CPT-11-R) LoVo colon cancer cell line Activate JNK and P38 and MOMP [150]
Peiminine Plant HCT116 Repress the phosphorylation of mTOR [149]
Tanshinones Plant multidrug-resistant colon cancer cells SW620 Ad300 Induce LC3B-Ⅱaccumulation [151]
Epigallocatechin gallate Plant DLD-1 and HT-29 Induce LC3-Ⅱaccumulation [199]
Cardamonin Plant HCT116 Increase p53/JNK [152]
Isothiocyanates Plant CoLo 205 Decrease the phosphorylation of Akt and mTOR [200]
Salvianolic Acid B Plant HCT116 and HT-29 Suppress Akt/mTOR pathway [153]

Table 6
Basic information for marine-derived anti-colorectal cancer natural products.
Natural products Marine organism Chemical class Mechanism Cancer type/cell lines References

Axidjiferosides Axinyssa djiferi β-Galactosylceramide Anti-Angiogenesis, Anti-Proliferation HCT116 [201]


Calothrixins Calothrix sp. Quinonoid Anti-Proliferation HCT116 [202]
Hierridin B Cyanobium sp. Ketone Apoptosis HT-29, HCT116 [203]
Violaxanthin Chlorella ellipsoidea Carotenoid Anti-Proliferation HCT116 [204]
Fucoxanthin Undaria pinnatifida Carotenoid Apoptosis CaCo-2, HT-29, DLD-1 [205]
Astaxanthin Haematococcus pluvialis Terpene Apoptosis, Anti-Proliferation, Cell cycle arrest HCT116 [206]
Salternamide A Streptomyces sp. Alkaloid Cell cycle arrest HCT116 [207]
Milnamide D Cymbastela sp. Peptide Unknown HCT116 [208]
Monactin Streptomyces sp. Macrolide Unknown HCT116 [209]
6-Bromoisatin Dicathais orbita Isatin Apoptosis, Anti-Proliferation HT-29 [210]
Nortopsentin Spongosorites ruetzleri Alkaloid Anti-Proliferation, Cell cycle arrest HCT116 [211]
Peridinin Gonyaulax polyedra Carotenoids Apoptosis, Cell cycle arrest DLD-1 [212]
Stichoposide C Thelenota anax Triterpene glycosides Apoptosis, Anti-Proliferation SNU-C4, HT-29, CT-26 [213]
Caulerpin Caulerpa cylindracea Alkaloid Anti-Proliferation HCT-116, HT-29 [214]
Phorbaketals Phorbas sp. Hydrobenzopyran unknown HT-29 [215]
Sipholenone A Siphonochalina sp. Triterpenoid Cell cycle arrest CaCo-2, HT-29 [216]
Sipholenol A Siphonochalina sp. Triterpenoid Anti-Proliferation, Cell cycle arrest CaCo-2, HT-29 [216]
Sipholenol L Siphonochalina sp. Triterpenoid Anti-Proliferation, Cell cycle arrest HT-29 [216]
Candidaspongiolide Candidaspongia sp. Polyketide Apoptosis HCT116 [217]
Physcion Penicillium janthinellum Anthraquinone Apoptosis HCT116 [218]
Gliotoxin Neosartorya pseufofischeri Epipolythiodioxopiperazine Apoptosis, Anti-Proliferation HCT116 [219]
Caldoramide Caldora penicillata Pentapeptide unknown HCT116 [220]
Renieramycin M Xestospongia sp. Tetrahydroisoquinoline Apoptosis HCT116 [221]
Manzamine A Acanthostrongylophora sp. Alkaloid Apoptosis HCT116 [222]

cells [132]. 2-(Pro-1-ynyl)-5-(5,6-dihydroxypenta-1,3diynyl) thiophene signaling pathways [140]. Calotroposid A, which was isolated from
(PYDDT), a thiophene isolated from the roots of Echinops grijsii, has Calotropis gigantean and determined to be a terpenoid glycoside, has
been found to induce mitochondrial-mediated apoptosis by regulating been found to trigger the extrinsic apoptosis pathway by increasing the
the levels of the related proteins, producing ROS, and activating ERK1/ caspase-8 expression in WiDr colon cancer cells [141].
2 and JNK in SW460 CRC cells [133]. A novel compound extracted from In addition, some natural products can trigger apoptosis via both the
the Chinese liverwort plant, Riccardin D, has been found to suppress the intrinsic and extrinsic pathways (Table 4). Zerumbone, a cyclic ses-
proliferation of HT-29 cells and induce their apoptosis via the NF-κB quiterpene from Zingiber zerumbet, can suppress cell growth and en-
signaling pathway [134]. Pteisolic acid G, which is a novel ent-kaurane hance cell apoptosis via the induction of ROS and activation of caspase-
diterpenoid isolated from Pteris semipinnata, has not only been found to 3, -8, and -9 [123]. Goniothalamin, a major bioactive styryl-lactone
inhibit the viability of HCT116 cancer cells, but also induce apoptosis compound that is present in plants, has been found to increase the DR5
via the generation of intracellular ROS, stimulation of p53, and expression and reduce cFILP expression, thereby enhancing TRAIL-in-
downregulation of NF-κB p65 activity [135]. The PI3K/Akt/mTOR duced apoptosis in CRC LoVo cells. Moreover, the activation of apop-
signaling pathway is an important pathway; its dysregulation has been tosis pathways, including the caspase-8, Bid-mediated death receptor
reported to be associated with the proliferation, survival, metabolism, apoptosis pathway and the caspase-9, Bcl-2, Bax-mediated mitochon-
and drug resistance of CRC cells [136–138]. It has been confirmed that drial apoptosis pathway, has been verified to promote apoptosis via the
OMT and OXA are not only able to inhibit the proliferation of CRC cells extrinsic and intrinsic pathways [142]. Mertensene, which is isolated
and lead to cell cycle arrest, as mentioned above, but are also able to from the red alga Pterocladiella capillacea, has been demonstrated to
induce apoptosis via the PI3K/Akt/mTOR pathway [115]. trigger apoptosis by regulating the expression of caspases, PARP, and
There are many natural products that exert anticancer effects TRADD in HT-29 and LS174 cells [143].
mediated by the extrinsic apoptotic pathway (Table 4). Proanthocya-
nidins (Pcys) extracted from 11 berry species have been shown to
trigger the extrinsic apoptosis pathway by activating caspase-8 in 3.4. Autophagy-based regulation
SW620-TRAIL-resistant and SW480-TRAIL-sensitive CRC cells, which
illustrate its potential to serve as an alternative to chemotherapeutic Autophagy, which is also known as type II programmed cell death,
agents for CRC treatment [139]. Oplopantriol A, which is a compound is a conserved catabolic process to eliminate excess long-lived proteins,
isolated from Oplopanax horridus, has been shown to induce apoptosis damaged organelles, and invasive pathogens mediated by autophago-
by the regulation of the related genes and TNF-mediated cell death somes [144,145]. Autophagy can be activated in response to various
pathways, including the TNF, FADD, TRADD, and caspase-3, -7, and -8 cellular stresses, including hypoxia, endoplasmic reticulum stress, ac-
cumulation of ROS, and drug stimulation [146–149]. Here, we

7
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

summarize some natural products that regulate the process of autop- therefore, they have become a treasure of lead compounds for the de-
hagy in CRC (Table 5). velopment of new drugs. Many anti-tumor compounds, including pep-
Peiminine, which is extracted from Fritillaria thunbergii, can enhance tides, macrolides, alkaloids, and polyphenols, derived from marine or-
the autophagic flux by inhibiting the phosphorylation of mTOR in ganisms have entered the preclinical and clinical research stages
HCT116 cells [149]. Thymoquinone, a phytochemical from Nigella sa- [30,160,161], highlighting that natural products from marine organ-
tiva, has been shown to cause caspase-independent autophagic cell isms are a constant source of new drugs against cancers including CRC
death via mitochondrial dysfunction and activate p38 and JNK in ir- (Table 6). In recent years, MPE has been used to elucidate the etiology
inotecan-resistant LoVo colon cancer cells, thereby exerting an anti- of diseases at the molecular, individual, and population levels, pro-
cancer function [150]. Hu et al. have confirmed that cryptotanshinone viding a more comprehensive understanding of CRC prevention and
(CTS) and dihydrotanshinone (DTS), which were isolated from the treatment. With the development of precision medicine, the combina-
traditional Chinese medicine Salvia miltiorrhiza, had the ability to in- tion of pharmacoepidemiology and MPE can make drugs safer and more
duce autophagic cell death in apoptosis-resistant colon cancer cells effective in clinical treatment. Furthermore, advanced applications in-
[151]. Cardamonin, which was isolated from Alpinia katsumadai volving genome mining, combinatorial biosynthesis, molecular mod-
Hayata, has been shown to not only suppress the proliferation of, and ification, and the high-throughput screening (HTS) technology will
cause G2/M phase arrest in, HCT116 cells, but also induce autophagy in provide strong supports for the development of novel promising and
these cells through the activation of p53/JNK [152]. Salvianolic acid B effective anti-CRC drugs and CRC treatment strategies.
(Sal B), isolated from the Chinese herb Salvia miltiorrhiza, has been
found to trigger autophagy via the suppression of the Akt/mTOR Funding
pathway in HT-29 and HCT116 cells [153]. Coroglaucigenin, which is a
natural product extracted from the roots of Calotropis gigantean, disrupts This work was supported by the National Natural Science
the association of Hsp90 with Akt and CDK4, leading to Akt depho- Foundation of China (31870046 and 81741166), the Natural Science
sphorylation and CDK4 degradation, finally resulting in senescence and Foundation of Guangdong Province (2016A030312014,
autophagy [154]. 2018A0303130005 and 2015A03313528), the Special Support
Program for Training High-Level Talents in Guangdong (201528018)
3.5. Angiogenesis-based regulation and Fund of the School of Laboratory Medicine of Guangdong Medical
University.
Tumor growth and metastasis are inseparable from cell migration,
invasion, and angiogenesis. Angiogenesis is a normal physiological References
process that provides oxygen and nutrients to tissues and involves en-
dothelial cell migration and morphogenesis during neovascularization [1] J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo,
D.M. Parkin, D. Forman, F. Bray, Cancer incidence and mortality worldwide:
[155]. During the process of angiogenesis, VEGF and its receptor play
sources, methods and major patterns in GLOBOCAN 2012, Int. J. Cancer 136
essential roles, and extracellular factors, such as interleukin-8 (IL-8), (2015) E359–86, https://doi.org/10.1002/ijc.29210.
tumor necrosis factor (TNF), and platelet-derived growth factor (PDGF), [2] L. Peng, K. Weigl, D. Boakye, H. Brenner, Risk scores for predicting advanced
colorectal neoplasia in the average-risk population: a systematic review and meta-
are also involved in the process of angiogenesis [156,157]. Conferone, analysis, Am. J. Gastroenterol. 113 (2018) 1788–1800, https://doi.org/10.1038/
an active compound isolated from plants in the genus Ferula, has been s41395-018-0209-2.
shown to inhibit the angiogenic ability of HT-29 cells by regulating the [3] T. Shen, L.D. Cai, Y.H. Liu, S. Li, W.J. Gan, X.M. Li, J.R. Wang, P. Da Guo, Q. Zhou,
X.X. Lu, L.N. Sun, J.M. Li, Ube2v1-mediated ubiquitination and degradation of
expression of VEGF and the growth factors angiopoietin-1 and -2. Sirt1 promotes metastasis of colorectal cancer by epigenetically suppressing au-
Moreover, its ability to suppress the migration of HT-29 cells has also tophagy, J. Hematol. Oncol. 11 (2018) 1–16, https://doi.org/10.1186/s13045-
018-0638-9.
been reported [158]. [4] M.M. Center, A. Jemal, R.A. Smith, E. Ward, Worldwide variations in colorectal
cancer, CA Cancer J. Clin. 59 (2009) 366–378, https://doi.org/10.3322/caac.
4. Conclusions and prospects 20038.
[5] S. Ogino, A.T. Chan, C.S. Fuchs, E. Giovannucci, Molecular pathological epide-
miology of colorectal neoplasia: an emerging transdisciplinary and inter-
Natural products have been confirmed to exert undoubted ad- disciplinary field, Gut. 60 (2011) 397–411, https://doi.org/10.1136/gut.2010.
vantages and have potential use for cancer treatment. In this review, we 217182.
[6] M. Yamauchi, T. Morikawa, A. Kuchiba, Y. Imamura, Z.R. Qian, R. Nishihara,
summarized the recently explored natural products that have been used X. Liao, L. Waldron, Y. Hoshida, C. Huttenhower, A.T. Chan, E. Giovannucci,
or have great potential for use in the treatment of CRC, and we have C. Fuchs, S. Ogino, Assessment of colorectal cancer molecular features along bowel
subsites challenges the conception of distinct dichotomy of proximal versus distal
presented experiment-based molecular mechanisms and regulatory colorectum, Gut. 61 (2012) 847–854, https://doi.org/10.1136/gutjnl-2011-
networks whereby natural products exert effects on the proliferation, 300865.
metastasis, apoptosis, autophagy, and angiogenesis in CRC. [7] I. Wasserman, L.H. Lee, S. Ogino, M.R. Marco, C. Wu, X. Chen, J. Datta, E. Sadot,
B. Szeglin, J.G. Guillem, P.B. Paty, M.R. Weiser, G.M. Nash, L. Saltz, A. Barlas,
Drug resistance and side effects remain the major problems asso- K. Manova-Todorova, S.P.B. Uppada, A.E. Elghouayel, P. Ntiamoah,
ciated with CRC treatment. Some natural products have been combined J.N. Glickman, T. Hamada, K. Kosumi, K. Inamura, A.T. Chan, R. Nishihara,
A. Cercek, K. Ganesh, N.E. Kemeny, P. Dhawan, R. Yaeger, C.L. Sawyers, J. Garcia-
with classical chemotherapeutic agents, such as 5-FU, to enhance the
Aguilar, M. Giannakis, J. Shia, J.J. Smith, SMAD4 loss in colorectal cancer patients
susceptibility of the cancer cells to conventional therapy, finally re- correlates with recurrence, loss of immune infiltrate, and chemoresistance, Clin.
inforcing the combined effect; this represents a new strategy for the Cancer Res. 25 (2019) 1948–1956, https://doi.org/10.1158/1078-0432.ccr-18-
1726.
treatment of CRC. However, with the identification of more natural [8] S. Ogino, P. Lochhead, A.T. Chan, R. Nishihara, E. Cho, B.M. Wolpin,
products, it has become very difficult to isolate novel active substances J.A. Meyerhardt, A. Meissner, E.S. Schernhammer, C.S. Fuchs, E. Giovannucci,
based on the discovery of traditional natural products from terrestrial Molecular pathological epidemiology of epigenetics: emerging integrative science
to analyze environment, host, and disease, Mod. Pathol. 26 (2013) 465–484,
animals, plants, or microorganisms. In particular, an increasing number https://doi.org/10.1038/modpathol.2012.214.
of known natural products have been repeatedly identified; this ser- [9] H. Suzuki, E. Yamamoto, R. Maruyama, T. Niinuma, M. Kai, Biological significance
of the CpG island methylator phenotype, Biochem. Biophys. Res. Commun. 455
iously interferes with the discovery of novel compounds. Thus, the (2014) 35–42, https://doi.org/10.1016/j.bbrc.2014.07.007.
discovery of new structural compounds remains a major challenge. [10] X. Zhang, N. Keum, K. Wu, S.A. Smith-Warner, S. Ogino, A.T. Chan, C.S. Fuchs,
Marine environments are a new resource that comprise a diversity E.L. Giovannucci, Calcium intake and colorectal cancer risk: results from the
nurses’ health study and health professionals follow-up study, Int. J. Cancer 139
of marine life; several organisms from marine environments have been (2016) 2232–2242, https://doi.org/10.1002/ijc.30293.
reported to produce novel secondary metabolites [159]. Marine-derived [11] N. Keum, D. Aune, D.C. Greenwood, W. Ju, E.L. Giovannucci, Calcium intake and
natural products have attracted a great amount of interest due to their colorectal cancer risk: dose-response meta-analysis of prospective observational
studies, Int. J. Cancer 135 (2014) 1940–1948, https://doi.org/10.1002/ijc.28840.
novel structures, diverse bioactivities, and new mechanisms of action;

8
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

[12] W. Yang, L. Liu, N. Keum, Z.R. Qian, J.A. Nowak, T. Hamada, M. Song, Y. Cao, doi.org/10.1186/s13045-016-0373-z.
K. Nosho, S.A. Smith-Warner, S. Zhang, Y. Masugi, K. Ng, K. Kosumi, Y. Ma, [34] S.H. Sharma, J. Suresh, D. Raj, S. Nagarajan, Biomedicine & pharmacotherapy
W.S. Garrett, M. Wang, H. Nan, M. Giannakis, J.A. Meyerhardt, A.T. Chan, molecular chemoprevention by morin – a plant flavonoid that targets nuclear
C.S. Fuchs, R. Nishihara, K. Wu, E.L. Giovannucci, S. Ogino, X. Zhang, Calcium factor kappa B in experimental colon cancer, Biomed. Pharmacother. 100 (2018)
intake and risk of colorectal cancer according to tumor-infiltrating T Cells, Cancer 367–373, https://doi.org/10.1016/j.biopha.2018.02.035.
Prev. Res. (Phila) 12 (2019) 283–294, https://doi.org/10.1158/1940-6207.CAPR- [35] J. Mao, S. Liu, M. Ai, Z. Wang, D. Wang, X. Li, K. Hu, X. Gao, A novel melittin
18-0279. nano-liposome exerted excellent anti-hepatocellular carcinoma efficacy with
[13] H. Jayasekara, R.J. MacInnis, E.J. Williamson, A.M. Hodge, M. Clendenning, better biological safety, J. Hematol. Oncol. (2017) 4–7, https://doi.org/10.1186/
C. Rosty, R. Walters, R. Room, M.C. Southey, M.A. Jenkins, R.L. Milne, s13045-017-0442-y.
J.L. Hopper, G.G. Giles, D.D. Buchanan, D.R. English, Lifetime alcohol intake is [36] J.J. Johnson, H. Mukhtar, Curcumin for chemoprevention of colon cancer, Cancer
associated with an increased risk of KRAS+ and BRAF-/KRAS- but not BRAF+ Lett. 255 (2007) 170–181, https://doi.org/10.1016/j.canlet.2007.03.005.
colorectal cancer, Int. J. Cancer 140 (2017) 1485–1493, https://doi.org/10.1002/ [37] B. Benarba, A. Pandiella, Colorectal cancer and medicinal plants: principle find-
ijc.30568. ings from recent studies, Biomed. Pharmacother. 107 (2018) 408–423, https://
[14] Y. Yamaoka, Y. Suehiro, S. Hashimoto, T. Hoshida, M. Fujimoto, M. Watanabe, doi.org/10.1016/j.biopha.2018.08.006.
D. Imanaga, K. Sakai, T. Matsumoto, M. Nishioka, T. Takami, N. Suzuki, [38] V. Aires, E. Limagne, A.K. Cotte, N. Latruffe, F. Ghiringhelli, D. Delmas,
S. Hazama, H. Nagano, I. Sakaida, T. Yamasaki, Fusobacterium nucleatum as a Resveratrol metabolites inhibit human metastatic colon cancer cells progression
prognostic marker of colorectal cancer in a Japanese population, J. Gastroenterol. and synergize with chemotherapeutic drugs to induce cell death, Mol. Nutr. Food
53 (2018) 517–524, https://doi.org/10.1007/s00535-017-1382-6. Res. 57 (2013) 1170–1181, https://doi.org/10.1002/mnfr.201200766.
[15] T. Hamada, J.A. Nowak, D.A. Milner, M. Song, S. Ogino, Integration of micro- [39] D.J. Newman, G.M. Cragg, Natural products as sources of new drugs from 1981 to
biology, molecular pathology, and epidemiology: a new paradigm to explore the 2014, J. Nat. Prod. 79 (2016) 629–661, https://doi.org/10.1021/acs.jnatprod.
pathogenesis of microbiome-driven neoplasms, J. Pathol. 247 (2019) 615–628, 5b01055.
https://doi.org/10.1002/path.5236. [40] A. Font, J.M. Sanchez, R. Rosell, M. Taron, E. Martinez, M. Guillot, J.L. Manzano,
[16] M.R. Rubinstein, X. Wang, W. Liu, Y. Hao, G. Cai, Y.W. Han, Fusobacterium nu- M. Margeli, A. Barnadas, A. Abad, Phase I study of weekly CPT-11 (irinotecan)/
cleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-ca- docetaxel in patients with advanced solid tumors, Lung Cancer. 37 (2002)
tenin signaling via its FadA adhesin, Cell Host Microbe 14 (2013) 195–206, 213–218.
https://doi.org/10.1016/j.chom.2013.07.012. [41] J.Y. Douillard, D. Cunningham, A.D. Roth, M. Navarro, R.D. James, P. Karasek,
[17] S. Bullman, C.S. Pedamallu, E. Sicinska, T.E. Clancy, X. Zhang, D. Cai, D. Neuberg, P. Jandik, T. Iveson, J. Carmichael, M. Alakl, G. Gruia, L. Awad, P. Rougier,
K. Huang, F. Guevara, T. Nelson, O. Chipashvili, T. Hagan, M. Walker, Irinotecan combined with fluorouracil compared with fluorouracil alone as first-
A. Ramachandran, B. Diosdado, G. Serna, N. Mulet, S. Landolfi, Y.C.S. Ramon, line treatment for metastatic colorectal cancer: a multicentre randomised trial,
R. Fasani, A.J. Aguirre, K. Ng, E. Elez, S. Ogino, J. Tabernero, C.S. Fuchs, Lancet 355 (2000) 1041–1047 https://www.sciencedirect.com/science/article/
W.C. Hahn, P. Nuciforo, M. Meyerson, Analysis of Fusobacterium persistence and pii/S0140673600020341?via%3Dihub.
antibiotic response in colorectal cancer, Science 358 (2017) 1443–1448, https:// [42] J.A. Conti, N.E. Kemeny, L.B. Saltz, Y. Huang, W.P. Tong, T.C. Chou, M. Sun,
doi.org/10.1126/science.aal5240 (80-.). S. Pulliam, C. Gonzalez, Irinotecan is an active agent in untreated patients with
[18] R. Benelli, R. Vene, N. Ferrari, Prostaglandin-endoperoxide synthase 2 (cycloox- metastatic colorectal cancer, J. Clin. Oncol. 14 (1996) 709–715, https://doi.org/
ygenase-2), a complex target for colorectal cancer prevention and therapy, Transl. 10.1200/jco.1996.14.3.709.
Res. 196 (2018) 42–61, https://doi.org/10.1016/j.trsl.2018.01.003. [43] P. Rougier, R. Bugat, J.Y. Douillard, S. Culine, E. Suc, P. Brunet, Y. Becouarn,
[19] M. Gu, R. Nishihara, Y. Chen, W. Li, Y. Shi, Y. Masugi, T. Hamada, K. Kosumi, M. Ychou, M. Marty, J.M. Extra, J. Bonneterre, A. Adenis, J.F. Seitz, G. Ganem,
L. Liu, A. da Silva, J.A. Nowak, T. Twombly, C. Du, H. Koh, W. Li, J.A. Meyerhardt, M. Namer, T. Conroy, S. Negrier, Y. Merrouche, F. Burki, M. Mousseau, P. Herait,
B.M. Wolpin, M. Giannakis, A.J. Aguirre, A.J. Bass, D.A. Drew, A.T. Chan, M. Mahjoubi, Phase II study of irinotecan in the treatment of advanced colorectal
C.S. Fuchs, Z.R. Qian, S. Ogino, Aspirin exerts high anti-cancer activity in PIK3CA- cancer in chemotherapy-naive patients and patients pretreated with fluorouracil-
mutant colon cancer cells, Oncotarget. 8 (2017) 87379–87389, https://doi.org/ based chemotherapy, J. Clin. Oncol. 15 (1997) 251–260, https://doi.org/10.
10.18632/oncotarget.20972. 1200/jco.1997.15.1.251.
[20] X. Liao, T. Morikawa, P. Lochhead, Y. Imamura, A. Kuchiba, M. Yamauchi, [44] M.F. Al-Kasspooles, S.K. Williamson, D. Henry, J. Howell, F. Niu, C.J. Decedue,
K. Nosho, Z.R. Qian, R. Nishihara, J.A. Meyerhardt, C.S. Fuchs, S. Ogino, K.F. Roby, Preclinical antitumor activity of a nanoparticulate SN38, Invest. New
Prognostic role of PIK3CA mutation in colorectal cancer: cohort study and lit- Drugs 31 (2013) 871–880, https://doi.org/10.1007/s10637-012-9919-2.
erature review, Clin. Cancer Res. 18 (2012) 2257–2268, https://doi.org/10.1158/ [45] H. Kamiyama, S. Takano, K. Tsuboi, A. Matsumura, Anti-angiogenic effects of
1078-0432.ccr-11-2410. SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1
[21] D. Woods, J.J. Turchi, Chemotherapy induced DNA damage response: con- alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of
vergence of drugs and pathways, Cancer Biol. Ther. 14 (2013) 379–389, https:// glioma and growth of endothelial cells, J. Cancer Res. Clin. Oncol. 131 (2005)
doi.org/10.4161/cbt.23761. 205–213, https://doi.org/10.1007/s00432-004-0642-z.
[22] A.F. Oliveira, R.R. Teixeira, A.S. Oliveira, A.P. Souza, M.L. Silva, S.O. Paula, [46] H. Iihara, H. Fujii, C. Yoshimi, R. Kobayashi, N. Matsuhashi, T. Takahashi,
Potential antivirals: natural products targeting replication enzymes of dengue and K. Yoshida, A. Suzuki, Prophylactic effect of scopolamine butylbromide, a com-
Chikungunya viruses, Molecules. 22 (2017), https://doi.org/10.3390/ petitive antagonist of muscarinic acetylcholine receptor, on irinotecan-related
molecules22030505. cholinergic syndrome, Cancer Chemother. Pharmacol. 83 (2019) 393–398,
[23] T. Miyata, Pharmacological basis of traditional medicines and health supplements https://doi.org/10.1007/s00280-018-3736-z.
as curatives, J. Pharmacol. Sci. 103 (2007) 127–131. [47] H. Hamano, M. Mitsui, Y. Zamami, K. Takechi, T. Nimura, N. Okada,
[24] C.H. Rammelkamp, C.S. Keefer, Penicillin: its antibacterial effect in whole blood K. Fukushima, M. Imanishi, M. Chuma, Y. Horinouchi, Y. Izawa-Ishizawa,
and serum for the hemolytic Streptococcus and Staphylococcus aureus, J. Clin. Y. Kirino, T. Nakamura, K. Teraoka, Y. Ikeda, H. Fujino, H. Yanagawa, T. Tamaki,
Invest. 22 (1943) 649–657, https://doi.org/10.1172/jci101437. K. Ishizawa, Irinotecan-induced neutropenia is reduced by oral alkalization drugs:
[25] I.M. Bondarev, A.A. Ertli, M.M. Khandulaev, Method of maximum concentration of analysis using retrospective chart reviews and the spontaneous reporting database,
antibacterial drugs in the treatment of destructive pulmonary tuberculosis, Probl. Support. Care Cancer 27 (2019) 849–856, https://doi.org/10.1007/s00520-018-
Tuberk. (1980) 36–39. 4367-y.
[26] L.L. Zhang, W. Wei, N.P. Wang, Q.T. Wang, J.Y. Chen, Y. Chen, H. Wu, X.Y. Hu, [48] F. Saliba, R. Hagipantelli, J.L. Misset, G. Bastian, G. Vassal, M. Bonnay, P. Herait,
Paeoniflorin suppresses inflammatory mediator production and regulates G pro- C. Cote, M. Mahjoubi, D. Mignard, E. Cvitkovic, Pathophysiology and therapy of
tein-coupled signaling in fibroblast-like synoviocytes of collagen induced arthritic irinotecan-induced delayed-onset diarrhea in patients with advanced colorectal
rats, Inflamm. Res. 57 (2008) 388–395, https://doi.org/10.1007/s00011-007- cancer: a prospective assessment, J. Clin. Oncol. 16 (1998) 2745–2751, https://
7240-x. doi.org/10.1200/jco.1998.16.8.2745.
[27] H.J. Zhang, E. Rumschlag-Booms, Y.F. Guan, D.Y. Wang, K.L. Liu, W.F. Li, [49] Y. Maehara, S. Tsujitani, H. Saeki, E. Oki, K. Yoshinaga, Y. Emi, M. Morita,
V.H. Nguyen, N.M. Cuong, D.D. Soejarto, H.H.S. Fong, L. Rong, Potent Inhibitor of S. Kohnoe, Y. Kakeji, T. Yano, H. Baba, Biological mechanism and clinical effect of
Drug-Resistant HIV-1 Strains Identified from the Medicinal Plant Justicia gen- protein-bound polysaccharide K (KRESTIN((R))): review of development and fu-
darussa, J. Nat. Prod. 80 (2017) 1798–1807, https://doi.org/10.1021/acs. ture perspectives, Surg. Today 42 (2012) 8–28, https://doi.org/10.1007/s00595-
jnatprod.7b00004. 011-0075-7.
[28] B.D. Kahan, Cyclosporine, N. Engl. J. Med. 321 (1989) 1725–1738, https://doi. [50] N. Tsutsumi, S. Kohnoe, H. Sonoda, A. Guntani, T. Rikimaru, K.I. Taguchi,
org/10.1056/nejm198912213212507. M. Tomikawa, Y. Kakeji, H. Nakashima, Y. Maehara, Protein-bound poly-
[29] R.S. Rosenson, C.C. Tangney, Antiatherothrombotic properties of statins: im- saccharide-K reduces colitic tumors and improves survival of inflammatory bowel
plications for cardiovascular event reduction, J. Am. Med. Assoc. 279 (1998) disease in vivo, Oncol. Lett. 2 (2011) 791–796, https://doi.org/10.3892/ol.2011.
1643–1650, https://doi.org/10.1001/jama.279.20.1643. 336.
[30] F. Grosso, R.L. Jones, G.D. Demetri, I.R. Judson, J.Y. Blay, A. Le Cesne, [51] M. Ikeguchi, M. Yamamoto, Y. Arai, Y. Maeta, K. Ashida, K. Katano, Y. Miki,
R. Sanfilippo, P. Casieri, P. Collini, P. Dileo, C. Spreafico, S. Stacchiotti, T. Kimura, Fucoidan reduces the toxicities of chemotherapy for patients with
E. Tamborini, J.C. Tercero, J. Jimeno, M. D’Incalci, A. Gronchi, J.A. Fletcher, unresectable advanced or recurrent colorectal cancer, Oncol. Lett. 2 (2011)
S. Pilotti, P.G. Casali, Efficacy of trabectedin (ecteinascidin-743) in advanced 319–322, https://doi.org/10.3892/ol.2011.254.
pretreated myxoid liposarcomas: a retrospective study, Lancet Oncol. 8 (2007) [52] E.J. Kim, S.Y. Park, J.Y. Lee, J.H. Park, Fucoidan present in brown algae induces
595–602, https://doi.org/10.1016/s1470-2045(07)70175-4. apoptosis of human colon cancer cells, BMC Gastroenterol. 10 (2010) 96, https://
[31] R. Singh, M. Sharma, P. Joshi, D.S. Rawat, Clinical status of anti-cancer agents doi.org/10.1186/1471-230x-10-96.
derived from marine sources, Anticancer Agents Med. Chem. 8 (2008) 603–617. [53] H.L. Tsai, C.J. Tai, C.W. Huang, F.R. Chang, J.Y. Wang, Efficacy of low-molecular-
[32] G. Li, H.X. Lou, Strategies to diversify natural products for drug discovery, Med. weight Fucoidan as a supplemental therapy in metastatic colorectal cancer pa-
Res. Rev. 38 (2018) 1255–1294, https://doi.org/10.1002/med.21474. tients: a double-blind randomized controlled trial, Mar. Drugs 15 (2017), https://
[33] P. Angulo, G. Kaushik, D. Subramaniam, P. Dandawate, K. Neville, K. Chastain, doi.org/10.3390/md15040122.
S. Anant, Natural compounds targeting major cell signaling pathways: A novel [54] A. Puissant, G. Robert, N. Fenouille, F. Luciano, J.P. Cassuto, S. Raynaud,
paradigm for osteosarcoma therapy, J. Hematol. Oncol. 10 (2017) 1–13, https:// P. Auberger, Resveratrol promotes autophagic cell death in chronic myelogenous

9
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

leukemia cells via JNK-mediated p62/SQSTM1 expression and AMPK activation, [77] E. Ellebaek, M.H. Andersen, I.M. Svane, P.T. Straten, Immunotherapy for meta-
Cancer Res. 70 (2010) 1042–1052, https://doi.org/10.1158/0008-5472.can-09- static colorectal cancer: present status and new options, Scand. J. Gastroenterol.
3537. 47 (2012) 315–324, https://doi.org/10.3109/00365521.2012.640831.
[55] H. Nakagawa, Y. Kiyozuka, Y. Uemura, H. Senzaki, N. Shikata, K. Hioki, [78] D. Cunningham, S. Pyrhonen, R.D. James, C.J. Punt, T.F. Hickish, R. Heikkila,
A. Tsubura, Resveratrol inhibits human breast cancer cell growth and may miti- T.B. Johannesen, H. Starkhammar, C.A. Topham, L. Awad, C. Jacques, P. Herait,
gate the effect of linoleic acid, a potent breast cancer cell stimulator, J. Cancer Res. Randomised trial of irinotecan plus supportive care versus supportive care alone
Clin. Oncol. 127 (2001) 258–264. after fluorouracil failure for patients with metastatic colorectal cancer, Lancet 352
[56] L. Mouchiroud, L. Molin, N. Dalliere, F. Solari, Life span extension by resveratrol, (1998) 1413–1418, https://doi.org/10.1016/s0140-6736(98)02309-5.
rapamycin, and metformin: the promise of dietary restriction mimetics for an [79] C. Tournigand, T. Andre, E. Achille, G. Lledo, M. Flesh, D. Mery-Mignard,
healthy aging, Biofactors. 36 (2010) 377–382, https://doi.org/10.1002/biof.127. E. Quinaux, C. Couteau, M. Buyse, G. Ganem, B. Landi, P. Colin, C. Louvet, A. de
[57] A. Bishayee, T. Politis, A.S. Darvesh, Resveratrol in the chemoprevention and Gramont, FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced
treatment of hepatocellular carcinoma, Cancer Treat. Rev. 36 (2010) 43–53, colorectal cancer: a randomized GERCOR study, J. Clin. Oncol. 22 (2004)
https://doi.org/10.1016/j.ctrv.2009.10.002. 229–237, https://doi.org/10.1200/jco.2004.05.113.
[58] M.J. Atten, E. Godoy-Romero, B.M. Attar, T. Milson, M. Zopel, O. Holian, [80] J. Sastre, R. Alfonso, J.A. Macias, I. Manrique, L. Flores, E. Diaz-Rubio, 5-fluor-
Resveratrol regulates cellular PKC alpha and delta to inhibit growth and induce ouracil as a protracted continuous infusion plus irinotecan (CPT-11) in patients
apoptosis in gastric cancer cells, Invest. New Drugs 23 (2005) 111–119, https:// with advanced colorectal cancer treated with fluoropyrimidine-based regimens as
doi.org/10.1007/s10637-005-5855-8. first line, Anticancer Drugs 14 (2003) 543–547, https://doi.org/10.1097/01.cad.
[59] Q. Ji, X. Liu, X. Fu, L. Zhang, H. Sui, L. Zhou, J. Sun, J. Cai, J. Qin, J. Ren, Q. Li, 0000086653.37889.7f.
Resveratrol inhibits invasion and metastasis of colorectal cancer cells via MALAT1 [81] S. Xie, G. Han, Z. Fan, L. He, W. Xu, Z. Qin, Safety and efficacy of second-line
mediated Wnt/β-catenin signal pathway, PLoS One 8 (2013), https://doi.org/10. treatment with folinic acid, 5-fluorouracil and irinotecan (FOLFIRI) in combina-
1371/journal.pone.0078700. tion of panitumumab and bevacizumab for patients with metastatic colorectal
[60] A.M. Engelbrecht, M. Mattheyse, B. Ellis, B. Loos, M. Thomas, R. Smith, S. Peters, cancer, Med. Oncol. 31 (2014) 35, https://doi.org/10.1007/s12032-014-0035-3.
C. Smith, K. Myburgh, Proanthocyanidin from grape seeds inactivates the PI3- [82] Y. Liu, L. Luan, X. Wang, A randomized Phase II clinical study of combining pa-
kinase/PKB pathway and induces apoptosis in a colon cancer cell line, Cancer Lett. nitumumab and bevacizumab, plus irinotecan, 5-fluorouracil, and leucovorin
258 (2007) 144–153, https://doi.org/10.1016/j.canlet.2007.08.020. (FOLFIRI) compared with FOLFIRI alone as second-line treatment for patients with
[61] C.C. Su, J.G. Lin, T.M. Li, J.G. Chung, J.S. Yang, S.W. Ip, W.C. Lin, G.W. Chen, metastatic colorectal cancer and KRAS mutation, Onco. Ther. 8 (2015)
Curcumin-induced apoptosis of human colon cancer colo 205 cells through the 1061–1068, https://doi.org/10.2147/ott.s81442.
production of ROS, Ca2+ and the activation of caspase-3, Anticancer Res. 26 [83] J.W. Holch, I. Ricard, S. Stintzing, L. Fischer von Weikersthal, T. Decker, A. Kiani,
(2006) 4379–4389 http://ar.iiarjournals.org/content/26/6B/4379.full.pdf. U. Vehling-Kaiser, T. Heintges, C. Kahl, F. Kullmann, W. Scheithauer, M. Moehler,
[62] J.L. Watson, R. Hill, P.W. Lee, C.A. Giacomantonio, D.W. Hoskin, Curcumin in- I. Jelas, D.P. Modest, C.B. Westphalen, J.C. von Einem, M. Michl, V. Heinemann,
duces apoptosis in HCT-116 human colon cancer cells in a p21-independent Relevance of baseline carcinoembryonic antigen for first-line treatment against
manner, Exp. Mol. Pathol. 84 (2008) 230–233, https://doi.org/10.1016/j.yexmp. metastatic colorectal cancer with FOLFIRI plus cetuximab or bevacizumab (FIRE-3
2008.02.002. trial), Eur. J. Cancer 106 (2019) 115–125, https://doi.org/10.1016/j.ejca.2018.
[63] T. Zhu, D.X. Wang, W. Zhang, X.Q. Liao, X. Guan, H. Bo, J.Y. Sun, N.W. Huang, 10.001.
J. He, Y.K. Zhang, J. Tong, C.Y. Li, Andrographolide protects against LPS-induced [84] J.M. Woynarowski, W.G. Chapman, C. Napier, M.C. Herzig, P. Juniewicz,
acute lung injury by inactivation of NF-kappaB, PLoS One 8 (2013) e56407, , Sequence- and region-specificity of oxaliplatin adducts in naked and cellular DNA,
https://doi.org/10.1371/journal.pone.0056407. Mol. Pharmacol. 54 (1998) 770–777 http://molpharm.aspetjournals.org/content/
[64] M.A. Hidalgo, A. Romero, J. Figueroa, P. Cortes, I.I. Concha, J.L. Hancke, 54/5/770.long.
R.A. Burgos, Andrographolide interferes with binding of nuclear factor-kappaB to [85] O. Rixe, W. Ortuzar, M. Alvarez, R. Parker, E. Reed, K. Paull, T. Fojo, Oxaliplatin,
DNA in HL-60-derived neutrophilic cells, Br. J. Pharmacol. 144 (2005) 680–686, tetraplatin, cisplatin, and carboplatin: spectrum of activity in drug-resistant cell
https://doi.org/10.1038/sj.bjp.0706105. lines and in the cell lines of the national cancer institute’s anticancer drug screen
[65] T. Shen, W.S. Yang, Y.S. Yi, G.H. Sung, M.H. Rhee, H. Poo, M.Y. Kim, K.W. Kim, panel, Biochem. Pharmacol. 52 (1996) 1855–1865 https://www.sciencedirect.
J.H. Kim, J.Y. Cho, AP-1/IRF-3 targeted anti-inflammatory activity of andro- com/science/article/pii/S0006295297814906?via%3Dihub.
grapholide isolated from Andrographis paniculata, Evid. Based Complement. [86] E. Wasserman, C. Cuvier, F. Lokiec, F. Goldwasser, S. Kalla, D. Mery-Mignard,
Altern. Med. 2013 (2013) 210736, , https://doi.org/10.1155/2013/210736. M. Ouldkaci, A. Besmaine, G. Dupont-Andre, M. Mahjoubi, M. Marty, J.L. Misset,
[66] K.C. Lee, H.H. Chang, Y.H. Chung, T.Y. Lee, Andrographolide acts as an anti-in- E. Cvitkovic, Combination of oxaliplatin plus irinotecan in patients with gastro-
flammatory agent in LPS-stimulated RAW264.7 macrophages by inhibiting intestinal tumors: results of two independent phase I studies with pharmacoki-
STAT3-mediated suppression of the NF-kappaB pathway, J. Ethnopharmacol. 135 netics, J. Clin. Oncol. 17 (1999) 1751–1759, https://doi.org/10.1200/jco.1999.
(2011) 678–684, https://doi.org/10.1016/j.jep.2011.03.068. 17.6.1751.
[67] J. Li, C. Zhang, H. Jiang, J. Cheng, Andrographolide inhibits hypoxia-inducible [87] F. Goldwasser, M. Gross-Goupil, J.M. Tigaud, M. Di Palma, J. Marceau-Suissa,
factor-1 through phosphatidylinositol 3-kinase/AKT pathway and suppresses E. Wasserman, A. Yovine, J.L. Misset, E. Cvitkovic, Dose escalation of CPT-11 in
breast cancer growth, Onco. Ther. 8 (2015) 427–435, https://doi.org/10.2147/ combination with oxaliplatin using an every two weeks schedule: a phase I study
ott.s76116. in advanced gastrointestinal cancer patients, Ann. Oncol. 11 (2000) 1463–1470,
[68] C.Y. Chao, C.K. Lii, Y.T. Hsu, C.Y. Lu, K.L. Liu, C.C. Li, H.W. Chen, Induction of https://doi.org/10.1023/a:1026535824044.
heme oxygenase-1 and inhibition of TPA-induced matrix metalloproteinase-9 ex- [88] E. Bajetta, E. Beretta, M. Di Bartolomeo, D. Cortinovis, E. Ferrario, G. Dognini,
pression by andrographolide in MCF-7 human breast cancer cells, Carcinogenesis. L. Toffolatti, R. Buzzoni, Efficacy of treatment with irinotecan and oxaliplatin
34 (2013) 1843–1851, https://doi.org/10.1093/carcin/bgt131. combination in FU-resistant metastatic colorectal cancer patients, Oncology 66
[69] W. Wang, W. Guo, L. Li, Z. Fu, W. Liu, J. Gao, Y. Shu, Q. Xu, Y. Sun, Y. Gu, (2004) 132–137, https://doi.org/10.1159/000077439.
Andrographolide reversed 5-FU resistance in human colorectal cancer by elevating [89] E. Timotheadou, P. Papakostas, D. Tsavdaridis, G. Basdanis, H. Kalofonos,
BAX expression, Biochem. Pharmacol. 121 (2016) 8–17, https://doi.org/10.1016/ G. Aravantinos, D. Bafaloukos, G. Fountzilas, Irinotecan and oxaliplatin combi-
j.bcp.2016.09.024. nation, as second-line treatment, in fluoropyrimidine-pretreated advanced color-
[70] M. Su, B. Qin, F. Liu, Y. Chen, R. Zhang, Andrographolide enhanced 5-fluorouracil- ectal cancer. A phase II study by the Hellenic Cooperative Oncology Group
induced antitumor effect in colorectal cancer via inhibition of c-MET pathway, (HeCOG), Tumori 91 (2005) 309–313.
Drug Des. Devel. Ther. 11 (2017) 3333–3341, https://doi.org/10.2147/DDDT. [90] D.G. Haller, M.L. Rothenberg, A.O. Wong, P.M. Koralewski, W.H. Miller Jr.,
S140354. G. Bodoky, N. Habboubi, C. Garay, L.O. Olivatto, Oxaliplatin plus irinotecan
[71] R.E. Go, K.A. Hwang, G.T. Park, H.M. Lee, G.A. Lee, C.W. Kim, S.Y. Jeon, J.W. Seo, compared with irinotecan alone as second-line treatment after single-agent
W.K. Hong, K.C. Choi, Effects of microalgal polyunsaturated fatty acid oil on body fluoropyrimidine therapy for metastatic colorectal carcinoma, J. Clin. Oncol. 26
weight and lipid accumulation in the liver of C57BL/6 mice fed a high fat diet, J. (2008) 4544–4550, https://doi.org/10.1200/jco.2008.17.1249.
Biomed. Res. 30 (2016) 234–242, https://doi.org/10.7555/jbr.30.2016k0004. [91] A. Sobrero, Medical oncology: IROX as second-line therapy for metastatic color-
[72] J.M. Nauroth, Y.C. Liu, M. Van Elswyk, R. Bell, E.B. Hall, G. Chung, ectal cancer, Nat. Rev. Clin. Oncol. 6 (2009) 248–250, https://doi.org/10.1038/
L.M. Arterburn, Docosahexaenoic acid (DHA) and docosapentaenoic acid (DPAn- nrclinonc.2009.43.
6) algal oils reduce inflammatory mediators in human peripheral mononuclear [92] R.M. Goldberg, D.J. Sargent, R.F. Morton, C.S. Fuchs, R.K. Ramanathan,
cells in vitro and paw edema in vivo, Lipids. 45 (2010) 375–384, https://doi.org/ S.K. Williamson, B.P. Findlay, H.C. Pitot, S.R. Alberts, A randomized controlled
10.1007/s11745-010-3406-3. trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in
[73] Z. Hossain, M. Hosokawa, K. Takahashi, Growth inhibition and induction of patients with previously untreated metastatic colorectal cancer, J. Clin. Oncol. 22
apoptosis of colon cancer cell lines by applying marine phospholipid, Nutr. Cancer (2004) 23–30, https://doi.org/10.1200/jco.2004.09.046.
61 (2009) 123–130, https://doi.org/10.1080/01635580802395725. [93] Y.Z. Patt, F.C. Lee, J.E. Liebmann, D. Diamandidis, S.G. Eckhardt, M. Javle,
[74] S.J. Winawer, R.H. Fletcher, L. Miller, F. Godlee, M.H. Stolar, C.D. Mulrow, G.R. Justice, W. Keiser, J.R. Salvatore, A. Bexon, E. Lin, Capecitabine plus 3-
S.H. Woolf, S.N. Glick, T.G. Ganiats, J.H. Bond, L. Rosen, J.G. Zapka, S.J. Olsen, weekly irinotecan (XELIRI regimen) as first-line chemotherapy for metastatic
F.M. Giardiello, J.E. Sisk, R. Van Antwerp, C. Brown -Davis, D.A. Marciniak, colorectal cancer: phase II trial results, Am. J. Clin. Oncol. 30 (2007) 350–357,
R.J. Mayer, Colorectal cancer screening: clinical guidelines and rationale the https://doi.org/10.1097/COC.0b013e31804b40bb.
adenoma-carcinoma sequence, Gastroenterology 112 (1997) 594–642. [94] D.W. Rea, J.W. Nortier, W.W. Ten Bokkel Huinink, S. Falk, D.J. Richel,
[75] Y. Horita, Y. Yamada, K. Kato, Y. Hirashima, K. Akiyoshi, K. Nagashima, T. Maughan, G. Groenewegen, J.M. Smit, N. Steven, J.M. Bakker, D. Semiond,
T. Nakajima, T. Hamaguchi, Y. Shimada, Phase II clinical trial of second-line D.J. Kerr, C.J. Punt, A phase I/II and pharmacokinetic study of irinotecan in
FOLFIRI plus bevacizumab for patients with metastatic colorectal cancer: AVASIRI combination with capecitabine as first-line therapy for advanced colorectal cancer,
trial, Int. J. Clin. Oncol. 17 (2012) 604–609, https://doi.org/10.1007/s10147- Ann. Oncol. 16 (2005) 1123–1132, https://doi.org/10.1093/annonc/mdi227.
011-0331-2. [95] S.J. Shin, J.B. Ahn, H.J. Choi, B.C. Cho, H.C. Jeung, S.Y. Rha, H.C. Chung,
[76] R.M. Lee, K. Cardona, M.C. Russell, Historical perspective: two decades of progress J.K. Roh, The combination of capecitabine and irinotecan in treating 5-fluorour-
in treating metastatic colorectal cancer, J. Surg. Oncol. 119 (2019) 549–563, acil- and oxaliplatin-pretreated metastatic colorectal cancer, Cancer Chemother.
https://doi.org/10.1002/jso.25431. Pharmacol. 61 (2008) 75–81, https://doi.org/10.1007/s00280-007-0447-2.

10
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

[96] P.M. Hoff, R. Ansari, G. Batist, J. Cox, W. Kocha, M. Kuperminc, J. Maroun, enhances antitumor activity of oxaliplatin in colon carcinoma through PI3K/AKT/
D. Walde, C. Weaver, E. Harrison, H.U. Burger, B. Osterwalder, A.O. Wong, mTOR pathway, Apoptosis. 21 (2016) 1398–1407, https://doi.org/10.1007/
R. Wong, Comparison of oral capecitabine versus intravenous fluorouracil plus s10495-016-1297-3.
leucovorin as first-line treatment in 605 patients with metastatic colorectal cancer: [116] B.R. Zetter, Angiogenesis and tumor metastasis, Annu. Rev. Med. 49 (1998)
results of a randomized phase III study, J. Clin. Oncol. 19 (2001) 2282–2292, 407–424, https://doi.org/10.1146/annurev.med.49.1.407.
https://doi.org/10.1200/jco.2001.19.8.2282. [117] E. Ondrouskova, L. Sommerova, R. Hrstka, (2019) [Multistep Process of
[97] E. Van Cutsem, C. Twelves, J. Cassidy, D. Allman, E. Bajetta, M. Boyer, R. Bugat, Establishing Carcinoma Metastases], Klin. Onkol. 29 Suppl 4 (n.d.) 12–17.
M. Findlay, S. Frings, M. Jahn, J. McKendrick, B. Osterwalder, G. Perez-Manga, [118] X. Lin, S. Wang, M. Sun, C. Zhang, C. Wei, C. Yang, R. Dou, Q. Liu, B. Xiong, miR-
R. Rosso, P. Rougier, W.H. Schmiegel, J.F. Seitz, P. Thompson, J.M. Vieitez, 195-5p/NOTCH2-mediated EMT modulates IL-4 secretion in colorectal cancer to
C. Weitzel, P. Harper, Oral capecitabine compared with intravenous fluorouracil affect M2-like TAM polarization, J. Hematol. Oncol. 12 (2019) 1–14, https://doi.
plus leucovorin in patients with metastatic colorectal cancer: results of a large org/10.1186/s13045-019-0708-7.
phase III study, J. Clin. Oncol. 19 (2001) 4097–4106, https://doi.org/10.1200/ [119] R.Y. Liu, Y. Zeng, Z. Lei, L. Wang, H. Yang, Z. Liu, J. Zhao, H.T. Zhang, JAK/STAT3
jco.2001.19.21.4097. signaling is required for TGF-beta-induced epithelial-mesenchymal transition in
[98] R.H. Xu, K. Muro, S. Morita, S. Iwasa, S.W. Han, W. Wang, M. Kotaka, lung cancer cells, Int. J. Oncol. 44 (2014) 1643–1651, https://doi.org/10.3892/
M. Nakamura, J.B. Ahn, Y.H. Deng, T. Kato, S.H. Cho, Y. Ba, H. Matsuoka, ijo.2014.2310.
K.W. Lee, T. Zhang, Y. Yamada, J. Sakamoto, Y.S. Park, T.W. Kim, Modified XELIRI [120] L. Cheng, T.S. Xia, Y.F. Wang, W. Zhou, X.Q. Liang, J.Q. Xue, L. Shi, Y. Wang,
(capecitabine plus irinotecan) versus FOLFIRI (leucovorin, fluorouracil, and ir- Q. Ding, M. Wang, The anticancer effect and mechanism of alpha-hederin on
inotecan), both either with or without bevacizumab, as second-line therapy for breast cancer cells, Int. J. Oncol. 45 (2014) 757–763, https://doi.org/10.3892/ijo.
metastatic colorectal cancer (AXEPT): a multicentre, open-label, randomised, non- 2014.2449.
inferiority, phase 3 trial, Lancet Oncol. 19 (2018) 660–671, https://doi.org/10. [121] S.W. Wang, Y.M. Sun, The IL-6/JAK/STAT3 pathway: potential therapeutic stra-
1016/s1470-2045(18)30140-2. tegies in treating colorectal cancer (Review), Int. J. Oncol. 44 (2014) 1032–1040,
[99] C.A. Neoh, W.T. Wu, G.F. Dai, J.H. Su, C.I. Liu, T.R. Su, Y.J. Wu, Flaccidoxide-13- https://doi.org/10.3892/ijo.2014.2259.
acetate extracted from the soft coral cladiella kashmani reduces human bladder [122] D. Sun, W. Shen, F. Zhang, H. Fan, C. Xu, L. Li, J. Tan, Y. Miao, H. Zhang, Y. Yang,
cancer cell migration and invasion through reducing activation of the FAK/PI3K/ H. Cheng, Alpha-Hederin inhibits interleukin 6-induced epithelial-to-mesench-
AKT/mTOR signaling pathway, Molecules 23 (2017), https://doi.org/10.3390/ ymal transition associated with disruption of JAK2/STAT3 signaling in colon
molecules23010058. cancer cells, Biomed. Pharmacother. 101 (2018) 107–114, https://doi.org/10.
[100] S.H. Sharma, J. Suresh, D. Raj, S. Nagarajan, Biomedicine & pharmacotherapy 1016/j.biopha.2018.02.062.
molecular chemoprevention by morin – a plant fl avonoid that targets nuclear [123] T. Sithara, B.P. Dhanya, K.B. Arun, S. Sini, M. Dan, R. Kokkuvayil Vasu, P. Nisha,
factor kappa B in experimental colon cancer, Biomed. Pharmacother. 100 (2018) Zerumbone, a cyclic sesquiterpene from Zingiber zerumbet induces apoptosis, cell
367–373, https://doi.org/10.1016/j.biopha.2018.02.035. cycle arrest, and antimigratory effects in SW480 colorectal Cancer cells, J. Agric.
[101] J.B. Morgan, Y. Liu, V. Coothankandaswamy, F. Mahdi, M.B. Jekabsons, Food Chem. 66 (2018) 602–612, https://doi.org/10.1021/acs.jafc.7b04472.
W.H. Gerwick, F.A. Valeriote, Y.D. Zhou, D.G. Nagle, Kalkitoxin inhibits angio- [124] L.Q. Liu, H.S. Li, S.P. Nie, M.Y. Shen, J.L. Hu, M.Y. Xie, Tea polysaccharide pre-
genesis, disrupts cellular hypoxic signaling, and blocks mitochondrial electron vents colitis-associated carcinogenesis in mice by inhibiting the proliferation and
transport in tumor cells, Mar. Drugs 13 (2015) 1552–1568, https://doi.org/10. invasion of tumor cells, Int. J. Mol. Sci. 19 (2018), https://doi.org/10.3390/
3390/md13031552. ijms19020506.
[102] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70. [125] M. Li, G.G. Yue, S.K. Tsui, K.P. Fung, C.B. Lau, Turmeric extract, with absorbable
[103] A. Amin, P.A. Karpowicz, T.E. Carey, J. Arbiser, R. Nahta, Z.G. Chen, J.T. Dong, curcumin, has potent anti-metastatic effect in vitro and in vivo, Phytomedicine. 46
O. Kucuk, G.N. Khan, G.S. Huang, S. Mi, H.Y. Lee, J. Reichrath, K. Honoki, (2018) 131–141, https://doi.org/10.1016/j.phymed.2018.03.065.
A.G. Georgakilas, A. Amedei, A. Amin, B. Helferich, C.S. Boosani, M.R. Ciriolo, [126] G. Kroemer, L. Galluzzi, C. Brenner, Mitochondrial membrane permeabilization in
S. Chen, S.I. Mohammed, A.S. Azmi, W.N. Keith, D. Bhakta, D. Halicka, E. Niccolai, cell death, Physiol. Rev. 87 (2007) 99–163, https://doi.org/10.1152/physrev.
H. Fujii, K. Aquilano, S.S. Ashraf, S. Nowsheen, X. Yang, A. Bilsland, D.M. Shin, 00013.2006.
Evasion of anti-growth signaling: a key step in tumorigenesis and potential target [127] S. Fulda, K.M. Debatin, Extrinsic versus intrinsic apoptosis pathways in anticancer
for treatment and prophylaxis by natural compounds, Semin. Cancer Biol. 35 chemotherapy, Oncogene. 25 (2006) 4798–4811, https://doi.org/10.1038/sj.onc.
(Suppl. (S55–S77)) (2015), https://doi.org/10.1016/j.semcancer.2015.02.005. 1209608.
[104] M.S. Abaza, M. Afzal, R.J. Al-Attiyah, R. Guleri, Methylferulate from Tamarix [128] G. Dranoff, Cytokines in cancer pathogenesis and cancer therapy, Nat. Rev. Cancer
aucheriana inhibits growth and enhances chemosensitivity of human colorectal 4 (2004) 11–22, https://doi.org/10.1038/nrc1252.
cancer cells: possible mechanism of action, BMC Complement. Altern. Med. 16 [129] J. Fu, D.N. Soroka, Y. Zhu, S. Sang, Induction of apoptosis and cell-cycle arrest in
(2016) 384, https://doi.org/10.1186/s12906-016-1358-8. human colon-cancer cells by whole-grain alkylresorcinols via activation of the p53
[105] K.C. Liu, T.Y. Shih, C.L. Kuo, Y.S. Ma, J.L. Yang, P.P. Wu, Y.P. Huang, K.C. Lai, pathway, J. Agric. Food Chem. 66 (2018) 11935–11942, https://doi.org/10.1021/
J.G. Chung, Sulforaphane induces cell death through G2/M phase arrest and acs.jafc.8b04442.
triggers apoptosis in HCT 116 human colon cancer cells, Am. J. Chin. Med. (Gard [130] Y. Zhang, Y. Guo, M. Wang, H. Dong, J. Zhang, L. Zhang, Quercetrin from Toona
City N Y) 44 (2016) 1289–1310, https://doi.org/10.1142/s0192415x16500725. sinensis leaves induces cell cycle arrest and apoptosis via enhancement of oxida-
[106] L. Chen, W.L. Wang, T.F. Song, X. Xie, X.W. Ye, Y. Liang, H.C. Huang, S.L. Yan, tive stress in human colorectal cancer SW620 cells, Oncol. Rep. 38 (2017)
X.Y. Lian, Z.Z. Zhang, Anti-colorectal cancer effects of tripolinolate A from tri- 3319–3326, https://doi.org/10.3892/or.2017.6042.
polium vulgare, Chin. J. Nat. Med. 15 (2017) 576–583, https://doi.org/10.1016/ [131] P.M. Kustiawan, K. Lirdprapamongkol, T. Palaga, S. Puthong, P. Phuwapraisirisan,
s1875-5364(17)30085-7. J. Svasti, C. Chanchao, Molecular mechanism of cardol, isolated from Trigona
[107] A. Anwar, N. Uddin, B.S. Siddiqui, R.A. Siddiqui, S. Begum, M.I. Choudhary, A incisa stingless bee propolis, induced apoptosis in the SW620 human colorectal
natural flavonoid lawsonaringenin induces cell cycle arrest and apoptosis in HT-29 cancer cell line, BMC Pharmacol. Toxicol. 18 (2017) 32, https://doi.org/10.1186/
colorectal cancer cells by targeting multiple signalling pathways, Mol. Biol. Rep. s40360-017-0139-4.
45 (2018) 1339–1348, https://doi.org/10.1007/s11033-018-4294-5. [132] B. Esmaeelian, K. Benkendorff, M.R. Johnston, C.A. Abbott, Purified brominated
[108] X. Chen, X. Wu, W. Ouyang, M. Gu, Z. Gao, M. Song, Y. Chen, Y. Lin, Y. Cao, indole derivatives from Dicathais orbita induce apoptosis and cell cycle arrest in
H. Xiao, Novel ent-kaurane diterpenoid from rubus corchorifolius L. F. inhibits colorectal cancer cell lines, Mar Drugs. 11 (2013) 3802–3822, https://doi.org/10.
human colon cancer cell growth via inducing cell cycle arrest and apoptosis, J. 3390/md11103802.
Agric. Food Chem. 65 (2017) 1566–1573, https://doi.org/10.1021/acs.jafc. [133] D.G. Xu, W. Lv, C.Y. Dai, F.F. Zhu, G.H. Xu, Z.J. Ma, Z. Chen, 2-(Pro-1-ynyl)-5-
6b05376. (5,6-dihydroxypenta-1,3-diynyl) thiophene induces apoptosis through reactive
[109] C. Weidner, M. Rousseau, A. Plauth, S.J. Wowro, C. Fischer, H. Abdel-Aziz, oxygen species-mediated jnk activation in human colon cancer SW620 cells, Anat.
S. Sauer, Iberis amara extract induces intracellular formation of reactive oxygen Rec. Hoboken (Hoboken) 385 (2015) 376–385, https://doi.org/10.1002/ar.
species and inhibits colon cancer, PLoS One 11 (2016) e0152398, , https://doi. 23045.
org/10.1371/journal.pone.0152398. [134] H. Liu, G. Li, B. Zhang, D. Sun, J. Wu, F. Chen, F. Kong, Y. Luan, W. Jiang,
[110] L. Kelland, The resurgence of platinum-based cancer chemotherapy, Nat. Rev. R. Wang, X. Xue, Suppression of the NFkappaB signaling pathway in colon cancer
Cancer 7 (2007) 573–584, https://doi.org/10.1038/nrc2167. cells by the natural compound Riccardin D from Dumortierahirsute, Mol. Med.
[111] Y.L. Lin, J.Y. Liau, S.C. Yu, D.L. Ou, L.I. Lin, L.H. Tseng, Y.L. Chang, K.H. Yeh, Rep. 17 (2018) 5837–5843, https://doi.org/10.3892/mmr.2018.8617.
A.L. Cheng, KRAS mutation is a predictor of oxaliplatin sensitivity in colon cancer [135] S. Qiu, X. Wu, H. Liao, X. Zeng, S. Zhang, X. Lu, X. He, X. Zhang, W. Ye, H. Wu,
cells, PLoS One 7 (2012) e50701, https://doi.org/10.1371/journal.pone.0050701. X. Zhu, Pteisolic acid G, a novel ent-kaurane diterpenoid, inhibits viability and
[112] R. Shiragami, S. Murata, C. Kosugi, T. Tezuka, M. Yamazaki, A. Hirano, induces apoptosis in human colorectal carcinoma cells, Oncol. Lett. 14 (2017)
Y. Yoshimura, M. Suzuki, K. Shuto, K. Koda, Enhanced antitumor activity of cer- 5540–5548, https://doi.org/10.3892/ol.2017.6889.
ulenin combined with oxaliplatin in human colon cancer cells, Int. J. Oncol. 43 [136] Y. Li, C. Zeng, J. Hu, Y. Pan, Y. Shan, B. Liu, L. Jia, Long non-coding RNA-SNHG7
(2013) 431–438, https://doi.org/10.3892/ijo.2013.1978. acts as a target of miR-34a to increase GALNT7 level and regulate PI3K/Akt/
[113] R.M. Goldberg, D.J. Sargent, R.F. Morton, C.S. Fuchs, R.K. Ramanathan, mTOR pathway in colorectal cancer progression, J. Hematol. Oncol. 11 (2018)
S.K. Williamson, B.P. Findlay, H.C. Pitot, S.R. Alberts, A randomized controlled 1–17, https://doi.org/10.1186/s13045-018-0632-2.
trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in [137] C. Porta, C. Paglino, A. Mosca, Targeting PI3K/Akt/mTOR signaling in Cancer,
patients with previously untreated metastatic colorectal cancer, J. Clin. Oncol. 22 Front. Oncol. 4 (2014) 64, https://doi.org/10.3389/fonc.2014.00064.
(2004) 23–30, https://doi.org/10.1200/jco.2004.09.046. [138] A. Bahrami, M. Khazaei, M. Hasanzadeh, S. ShahidSales, M. Joudi Mashhad,
[114] D. Fiore, M.C. Proto, S. Pisanti, P. Picardi, A.C. Pagano Zottola, S. Butini, M. Farazestanian, H.R. Sadeghnia, M. Rezayi, M. Maftouh, S.M. Hassanian,
S. Gemma, A. Casagni, C. Laezza, M. Vitale, A. Ligresti, V. Di Marzo, D.M. Zisterer, A. Avan, Therapeutic potential of targeting PI3K/AKT pathway in treatment of
S. Nathwani, D.C. Williams, G. Campiani, P. Gazzerro, M. Bifulco, Antitumor effect colorectal Cancer: rational and progress, J. Cell. Biochem. 119 (2018) 2460–2469,
of pyrrolo-1,5-benzoxazepine-15 and its synergistic effect with Oxaliplatin and 5- https://doi.org/10.1002/jcb.25950.
FU in colorectal cancer cells, Cancer Biol. Ther. 17 (2016) 849–858, https://doi. [139] C. Minker, L. Duban, D. Karas, P. Jarvinen, A. Lobstein, C.D. Muller, Impact of
org/10.1080/15384047.2015.1078028. procyanidins from different berries on caspase 8 activation in Colon Cancer, Oxid.
[115] Y. Liu, T. Bi, Z. Wang, G. Wu, L. Qian, Q. Gao, G. Shen, Oxymatrine synergistically Med. Cell. Longev. 2015 (2015) 154164, , https://doi.org/10.1155/2015/154164.

11
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

[140] Z. Zhang, C. Yu, C.F. Zhang, X.H. Wu, X.D. Wen, S. Anderson, W. Du, W.H. Huang, E. Bonmassar, Effect of resveratrol on proliferation and telomerase activity of
S.P. Li, C.Z. Wang, C.S. Yuan, Chemopreventive effects of oplopantriol A, a novel human colon cancer cells in vitro, J. Exp. Clin. Cancer Res. 25 (2006) 189–193.
compound isolated from Oplopanax horridus, on colorectal cancer, Nutrients 6 [164] Y.H. Li, Y.B. Niu, Y. Sun, F. Zhang, C.X. Liu, L. Fan, Q.B. Mei, Role of phyto-
(2014) 2668–2680, https://doi.org/10.3390/nu6072668. chemicals in colorectal cancer prevention, World J. Gastroenterol. 21 (2015)
[141] R. Mutiah, A. Widyawaruyanti, S. Sukardiman, Calotroposid A: a glycosides ter- 9262–9272, https://doi.org/10.3748/wjg.v21.i31.9262.
penoids from Calotropis gigantea induces apoptosis of colon cancer WiDr cells [165] G.J. Creemers, Topotecan in advanced colorectal cancer, Semin. Oncol. 24 (1997)
through cell cycle arrest G2/M and caspase 8 expression, Asian Pac. J. Cancer S20-42-s20-48.
Prev. 19 (2018) 1457–1464, https://doi.org/10.22034/apjcp.2018.19.6.1457. [166] M.S. Han, H.J. Lee, S.J. Park, S.P. Hong, J.H. Cheon, W.H. Kim, T.I. Kim, The effect
[142] T. Sophonnithiprasert, S. Nilwarangkoon, Y. Nakamura, R. Watanapokasin, of metformin on the recurrence of colorectal adenoma in diabetic patients with
Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells previous colorectal adenoma, Int J Color. Dis. 32 (2017) 1223–1226, https://doi.
through DR5 upregulation and cFLIP downregulation, Int. J. Oncol. 47 (2015) org/10.1007/s00384-017-2782-z.
2188–2196, https://doi.org/10.3892/ijo.2015.3204. [167] I. Altomare, H. Hurwitz, Everolimus in colorectal cancer, Expert Opin.
[143] S. Tarhouni-Jabberi, O. Zakraoui, E. Ioannou, I. Riahi-Chebbi, M. Haoues, Pharmacother. 14 (2013) 505–513, https://doi.org/10.1517/14656566.2013.
V. Roussis, R. Kharrat, K. Essafi-Benkhadir, Mertensene, a halogenated mono- 770473.
terpene, induces G2/M cell cycle arrest and caspase dependent apoptosis of human [168] C. Chu, M.S. Noel-Hudson, V. Boige, D. Goere, S. Marion, M. Polrot, L. Bigot,
colon adenocarcinoma HT29 cell line through the modulation of ERK-1/-2, AKT P. Gonin, R. Farinotti, L. Bonhomme-Faivre, Therapeutic efficiency of everolimus
and NF-Kappab signaling, Mar. Drugs 15 (2017), https://doi.org/10.3390/ and lapatinib in xenograft model of human colorectal carcinoma with KRAS mu-
md15070221. tation, Fundam. Clin. Pharmacol. 27 (2013) 434–442, https://doi.org/10.1111/j.
[144] C. He, D.J. Klionsky, Regulation mechanisms and signaling pathways of autop- 1472-8206.2012.01035.x.
hagy, Annu. Rev. Genet. 43 (2009) 67–93, https://doi.org/10.1146/annurev- [169] G. Palmieri, C. Gridelli, G. Airoma, R. Pepe, P. Incoronato, F. Caponigro,
genet-102808-114910. A.R. Bianco, Second-line chemotherapy of advanced colorectal cancer with se-
[145] G.J. Yoshida, Therapeutic strategies of drug repositioning targeting autophagy to quential high-dose methotrexate and 5-fluorouracil, J. Chemother. 3 (1991)
induce cancer cell death: from pathophysiology to treatment, J. Hematol. Oncol. 55–60.
10 (2017) 67, https://doi.org/10.1186/s13045-017-0436-9. [170] M.D. Shi, H.H. Lin, Y.C. Lee, J.K. Chao, R.A. Lin, J.H. Chen, Inhibition of cell-cycle
[146] T. Yorimitsu, D.J. Klionsky, Endoplasmic reticulum stress: a new pathway to in- progression in human colorectal carcinoma Lovo cells by andrographolide, Chem.
duce autophagy, Autophagy 3 (2007) 160–162. Biol. Interact. 174 (2008) 201–210, https://doi.org/10.1016/j.cbi.2008.06.006.
[147] W.S. Lee, W.H. Yoo, H.J. Chae, ER stress and autophagy, Curr. Mol. Med. 15 [171] V. Poindessous, F. Koeppel, E. Raymond, E. Cvitkovic, S.J. Waters, A.K. Larsen,
(2015) 735–745. Enhanced antitumor activity of irofulven in combination with 5-fluorouracil and
[148] H.L. Yang, R.W. Lin, P. Karuppaiya, D.C. Mathew, T. Der Way, H.C. Lin, C.C. Lee, cisplatin in human colon and ovarian carcinoma cells, Int. J. Oncol. 23 (2003)
Y.C. Hseu, Induction of autophagic cell death in human ovarian carcinoma cells by 1347–1355 https://www.spandidos-publications.com/ijo/23/5/1347.
Antrodia salmonea through increased reactive oxygen species generation, J. Cell. [172] A.E. Escargueil, V. Poindessous, D.G. Soares, A. Sarasin, P.R. Cook, A.K. Larsen,
Physiol. 234 (2019) 10747–10760, https://doi.org/10.1002/jcp.27749. Influence of irofulven, a transcription-coupled repair-specific antitumor agent, on
[149] Q. Lyu, F. Tou, H. Su, X. Wu, X. Chen, Z. Zheng, The natural product peiminine RNA polymerase activity, stability and dynamics in living mammalian cells, J.
represses colorectal carcinoma tumor growth by inducing autophagic cell death, Cell. Sci. 121 (2008) 1275–1283, https://doi.org/10.1242/jcs.023259.
Biochem. Biophys. Res. Commun. 462 (2015) 38–45, https://doi.org/10.1016/j. [173] A. Kolman, Epothilone d (Kosan/Roche), Curr. Opin. Investig. Drugs 5 (2004)
bbrc.2015.04.102. 657–667.
[150] M.C. Chen, N.H. Lee, H.H. Hsu, T.J. Ho, C.C. Tu, D.J. Hsieh, Y.M. Lin, L.M. Chen, [174] H.J. Eo, G.H. Park, J.B. Jeong, Inhibition of wnt signaling by silymarin in human
W.W. Kuo, C.Y. Huang, Thymoquinone induces caspase-independent, autophagic colorectal cancer cells, Biomol. Ther. (Seoul) 24 (2016) 380–386, https://doi.org/
cell death in CPT-11-resistant lovo colon cancer via mitochondrial dysfunction and 10.4062/biomolther.2015.154.
activation of JNK and p38, J. Agric. Food Chem. 63 (2015) 1540–1546, https:// [175] Y. Cai, Q. Xia, R. Luo, P. Huang, Y. Sun, Y. Shi, W. Jiang, Berberine inhibits the
doi.org/10.1021/jf5054063. growth of human colorectal adenocarcinoma in vitro and in vivo, J. Nat. Med. 68
[151] T. Hu, L. Wang, L. Zhang, L. Lu, J. Shen, R.L. Chan, M. Li, W.K. Wu, K.K. To, (2014) 53–62, https://doi.org/10.1007/s11418-013-0766-z.
C.H. Cho, Sensitivity of apoptosis-resistant colon cancer cells to tanshinones is [176] A. Marinelli, F.R. Dijkstra, J.H. van Dierendonck, P.J. Kuppen, C.J. Cornelisse,
mediated by autophagic cell death and p53-independent cytotoxicity, C.J. van de Velde, Effectiveness of isolated liver perfusion with mitomycin C in the
Phytomedicine 22 (2015) 536–544, https://doi.org/10.1016/j.phymed.2015.03. treatment of liver tumours of rat colorectal cancer, Br. J. Cancer 64 (1991) 74–78.
010. [177] M.K. Lemieszek, M. Ribeiro, G. Marques, F.M. Nunes, P. Pozarowski, W. Rzeski,
[152] Y.J. Kim, K.S. Kang, K.C. Choi, H. Ko, Cardamonin induces autophagy and an New insights into the molecular mechanism of Boletus edulis ribonucleic acid
antiproliferative effect through JNK activation in human colorectal carcinoma fraction (BE3) concerning antiproliferative activity on human colon cancer cells,
HCT116 cells, Bioorg. Med. Chem. Lett. 25 (2015) 2559–2564, https://doi.org/10. Food Funct. 8 (2017) 1830–1839, https://doi.org/10.1039/c6fo01626j.
1016/j.bmcl.2015.04.054. [178] G.H. Park, H.M. Song, J.B. Jeong, The coffee diterpene kahweol suppresses the cell
[153] Z. Jing, W. Fei, J. Zhou, L. Zhang, L. Chen, X. Zhang, X. Liang, J. Xie, Y. Fang, proliferation by inducing cyclin D1 proteasomal degradation via ERK1/2, JNK and
X. Sui, W. Han, H. Pan, Salvianolic acid B, a novel autophagy inducer, exerts GKS3beta-dependent threonine-286 phosphorylation in human colorectal cancer
antitumor activity as a single agent in colorectal cancer cells, Oncotarget. 7 (2016) cells, Food Chem. Toxicol. 95 (2016) 142–148, https://doi.org/10.1016/j.fct.
61509–61519, https://doi.org/10.18632/oncotarget.11385. 2016.07.008.
[154] Y.H. Huang, J. Lei, G.H. Yi, F.Y. Huang, Y.N. Li, C.C. Wang, Y. Sun, H.F. Dai, [179] P. Mahajan, R. Gnana Oli, S.M. Jachak, S.B. Bharate, B. Chaudhuri, Antioxidant
G.H. Tan, Coroglaucigenin induces senescence and autophagy in colorectal cancer and antiproliferative activity of indigocarpan, a pterocarpan from Indigofera as-
cells, Cell Prolif. 51 (2018) e12451, https://doi.org/10.1111/cpr.12451. palathoides, J. Pharm. Pharmacol. 68 (2016) 1331–1339, https://doi.org/10.
[155] D.R. Bielenberg, B.R. Zetter, The contribution of angiogenesis to the process of 1111/jphp.12609.
metastasis, Cancer J. 21 (2015) 267–273, https://doi.org/10.1097/ppo. [180] R. Zhang, Y. Wang, J. Li, H. Jin, S. Song, C. Huang, The Chinese herb isolate
0000000000000138. yuanhuacine (YHL-14) induces G2/M arrest in human cancer cells by up-reg-
[156] S. Zhang, Z. Cao, H. Tian, G. Shen, Y. Ma, H. Xie, Y. Liu, C. Zhao, S. Deng, Y. Yang, ulating p21 protein expression through an p53 protein-independent cascade, J.
R. Zheng, W. Li, N. Zhang, S. Liu, W. Wang, L. Dai, S. Shi, L. Cheng, Y. Pan, Biol. Chem. 289 (2014) 6394–6403, https://doi.org/10.1074/jbc.M113.513960.
S. Feng, X. Zhao, H. Deng, S. Yang, Y. Wei, SKLB1002, a novel potent inhibitor of [181] W.K. Kim, W.S. Byun, H.J. Chung, J. Oh, H.J. Park, J.S. Choi, S.K. Lee, Esculetin
VEGF receptor 2 signaling, inhibits angiogenesis and tumor growth in vivo, Clin. suppresses tumor growth and metastasis by targeting Axin2/E-cadherin axis in
Cancer Res. 17 (2011) 4439–4450, https://doi.org/10.1158/1078-0432.ccr-10- colorectal cancer, Biochem. Pharmacol. 152 (2018) 71–83, https://doi.org/10.
3109. 1016/j.bcp.2018.03.009.
[157] W. Sun, Angiogenesis in metastatic colorectal cancer and the benefits of targeted [182] M.S. Shin, S.H. Hwang, T.J. Yoon, S.H. Kim, K.S. Shin, Polysaccharides from
therapy, J. Hematol. Oncol. 5 (2012) 63, https://doi.org/10.1186/1756-8722- ginseng leaves inhibit tumor metastasis via macrophage and NK cell activation,
5-63. Int. J. Biol. Macromol. 103 (2017) 1327–1333, https://doi.org/10.1016/j.
[158] O. Cheraghi, G. Dehghan, M. Mahdavi, R. Rahbarghazi, A. Rezabakhsh, ijbiomac.2017.05.055.
H.N. Charoudeh, M. Iranshahi, S. Montazersaheb, Potent anti-angiogenic and cy- [183] Y. Liu, T. Lang, B. Jin, F. Chen, Y. Zhang, R.W. Beuerman, L. Zhou, Z. Zhang,
totoxic effect of conferone on human colorectal adenocarcinoma HT-29 cells, Luteolin inhibits colorectal cancer cell epithelial-to-mesenchymal transition by
Phytomedicine. 23 (2016) 398–405, https://doi.org/10.1016/j.phymed.2016.01. suppressing CREB1 expression revealed by comparative proteomics study, J.
015. Proteomics 161 (2017) 1–10, https://doi.org/10.1016/j.jprot.2017.04.005.
[159] M.S. Kumar, K.M. Adki, Biomedicine & pharmacotherapy marine natural products [184] C. Chen, T. Ma, C. Zhang, H. Zhang, L. Bai, L. Kong, J. Luo, Down-regulation of
for multi-targeted cancer treatment: a future insight, Biomed. Pharmacother. 105 aquaporin 5-mediated epithelial-mesenchymal transition and anti-metastatic ef-
(2018) 233–245, https://doi.org/10.1016/j.biopha.2018.05.142. fect by natural product Cairicoside E in colorectal cancer, Mol. Carcinog. 56
[160] D.J. Newman, G.M. Cragg, Marine natural products and related compounds in (2017) 2692–2705, https://doi.org/10.1002/mc.22712.
clinical and advanced preclinical trials, J. Nat. Prod. 67 (2004) 1216–1238, [185] C. Buhrmann, P. Shayan, A. Goel, M. Shakibaei, Resveratrol regulates colorectal
https://doi.org/10.1021/np040031y. Cancer cell invasion by modulation of focal adhesion molecules, Nutrients. 9
[161] E. Hansen, J.H. Andersen, Screening for marine natural products with potential as (2017), https://doi.org/10.3390/nu9101073.
chemotherapeutics for acute myeloid leukemia, Curr. Pharm. Biotechnol. 17 [186] T. Yamamoto, K. Uemura, K. Moriyama, K. Mitamura, A. Taga, Inhibitory effect of
(2016) 71–77. maple syrup on the cell growth and invasion of human colorectal cancer cells,
[162] J.C. Stingl, T. Ettrich, R. Muche, M. Wiedom, J. Brockmoller, A. Seeringer, Oncol. Rep. 33 (2015) 1579–1584, https://doi.org/10.3892/or.2015.3777.
T. Seufferlein, Protocol for minimizing the risk of metachronous adenomas of the [187] H. Yan, X. Wang, Y. Wang, P. Wang, Y. Xiao, Antiproliferation and anti-migration
colorectum with green tea extract (MIRACLE): a randomised controlled trial of induced by gypenosides in human colon cancer SW620 and esophageal cancer
green tea extract versus placebo for nutriprevention of metachronous colon ade- Eca-109 cells, Hum. Exp. Toxicol. 33 (2014) 522–533, https://doi.org/10.1177/
nomas in the elderly population, BMC Cancer 11 (2011) 360, https://doi.org/10. 0960327113497771.
1186/1471-2407-11-360. [188] H. Yan, X. Wang, J. Niu, Y. Wang, P. Wang, Q. Liu, Anti-cancer effect and the
[163] M.P. Fuggetta, G. Lanzilli, M. Tricarico, A. Cottarelli, R. Falchetti, G. Ravagnan, underlying mechanisms of gypenosides on human colorectal cancer SW-480 cells,

12
X.-m. Huang, et al. Biomedicine & Pharmacotherapy 117 (2019) 109142

PLoS One 9 (2014) e95609, https://doi.org/10.1371/journal.pone.0095609. 1675 (2004) 113–119, https://doi.org/10.1016/j.bbagen.2004.08.012.


[189] H. Li, X.X. Meng, L. Zhang, B.J. Zhang, X.Y. Liu, W.W. Fu, H.S. Tan, Y.Z. Lao, [206] P. Palozza, C. Torelli, A. Boninsegna, R. Simone, A. Catalano, M.C. Mele, N. Picci,
H.X. Xu, Oblongifolin C and guttiferone K extracted from Garcinia yunnanensis Growth-inhibitory effects of the astaxanthin-rich alga Haematococcus pluvialis in
fruit synergistically induce apoptosis in human colorectal cancer cells in vitro, human colon cancer cells, Cancer Lett. 283 (2009) 108–117, https://doi.org/10.
Acta Pharmacol. Sin. 38 (2017) 252–263, https://doi.org/10.1038/aps.2016.101. 1016/j.canlet.2009.03.031.
[190] K.D. Tsai, Y.H. Liu, T.W. Chen, S.M. Yang, H.Y. Wong, J. Cherng, K.S. Chou, [207] D.H. Bach, S.H. Kim, J.Y. Hong, H.J. Park, D.C. Oh, S.K. Lee, Salternamide A
J.M. Cherng, Cuminaldehyde from Cinnamomum verum induces cell death Suppresses Hypoxia-Induced Accumulation of HIF-1alpha and Induces Apoptosis
through targeting topoisomerase 1 and 2 in human colorectal adenocarcinoma in Human Colorectal Cancer Cells, Mar. Drugs 13 (2015) 6962–6976, https://doi.
COLO 205 cells, Nutrients 8 (2016), https://doi.org/10.3390/nu8060318. org/10.3390/md13116962.
[191] V.P. Dia, H.B. Krishnan, BG-4, a novel anticancer peptide from bitter gourd [208] C. Chevallier, A.D. Richardson, M.C. Edler, E. Hamel, M.K. Harper, C.M. Ireland, A
(Momordica charantia), promotes apoptosis in human colon cancer cells, Sci. Rep. new cytotoxic and tubulin-interactive milnamide derivative from a marine sponge
6 (2016) 33532, https://doi.org/10.1038/srep33532. Cymbastela sp, Org. Lett. 5 (2003) 3737–3739, https://doi.org/10.1021/
[192] C.K. Chan, G. Chan, K. Awang, H. Abdul Kadir, Deoxyelephantopin from ol035476c.
Elephantopus scaber inhibits HCT116 human colorectal carcinoma cell growth [209] A. Becerril-Espinosa, G. Guerra-Rivas, N. Ayala-Sánchez, I.E. Soria-Mercado,
through apoptosis and cell cycle arrest, Molecules 21 (2016) 385, https://doi.org/ Antitumor activity of actinobacteria isolated in marine sediment from Todos
10.3390/molecules21030385. Santos Bay, Baja California, Mexico, Rev. Biol. Mar. Oceanogr. 47 (2012)
[193] M. Asif, A. Shafaei, S.F. Jafari, S.K. Mohamed, M.O. Ezzat, A.S. Abdul Majid, 317–325, https://doi.org/10.4067/s0718-19572012000200013.
C.E. Oon, S.H. Petersen, K. Kono, A.M. Abdul Majid, Isoledene from Mesua ferrea [210] B. Esmaeelian, C.A. Abbott, R.K. Le Leu, K. Benkendorff, 6-bromoisatin found in
oleo-gum resin induces apoptosis in HCT 116 cells through ROS-mediated mod- muricid mollusc extracts inhibits colon cancer cell proliferation and induces
ulation of multiple proteins in the apoptotic pathways: a mechanistic study, apoptosis, preventing early stage tumor formation in a colorectal cancer rodent
Toxicol. Lett. 257 (2016) 84–96, https://doi.org/10.1016/j.toxlet.2016.05.027. model, Mar. Drugs 12 (2013) 17–35, https://doi.org/10.3390/md12010017.
[194] C. Wen, L. Huang, J. Chen, M. Lin, W. Li, B. Lu, Z.J. Rutnam, A. Iwamoto, Z. Wang, [211] A. Carbone, B. Parrino, G. Di Vita, A. Attanzio, V. Spano, A. Montalbano,
X. Yang, H. Liu, Gambogic acid inhibits growth, induces apoptosis, and overcomes P. Barraja, L. Tesoriere, M.A. Livrea, P. Diana, G. Cirrincione, Synthesis and an-
drug resistance in human colorectal cancer cells, Int. J. Oncol. 47 (2015) tiproliferative activity of thiazolyl-bis-pyrrolo[2,3-b]pyridines and indolyl-thia-
1663–1671, https://doi.org/10.3892/ijo.2015.3166. zolyl-pyrrolo[2,3-c]pyridines, nortopsentin analogues, Mar. Drugs 13 (2015)
[195] B.X. Liu, J.Y. Zhou, Y. Li, X. Zou, J. Wu, J.F. Gu, J.R. Yuan, B.J. Zhao, L. Feng, 460–492, https://doi.org/10.3390/md13010460.
X.B. Jia, R.P. Wang, Hederagenin from the leaves of ivy (Hedera helix L.) induces [212] T. Sugawara, K. Yamashita, S. Sakai, A. Asai, A. Nagao, T. Shiraishi, I. Imai,
apoptosis in human LoVo colon cells through the mitochondrial pathway, BMC T. Hirata, Induction of apoptosis in DLD-1 human colon cancer cells by peridinin
Complement. Altern. Med. 14 (2014) 412, https://doi.org/10.1186/1472-6882- isolated from the dinoflagellate, Heterocapsa triquetra, Biosci. Biotechnol.
14-412. Biochem. 71 (2007) 1069–1072, https://doi.org/10.1271/bbb.60597.
[196] F.J. Reyes-Zurita, E.E. Rufino-Palomares, L. Garcia-Salguero, J. Peragon, [213] S.H. Yun, E.S. Park, S.W. Shin, Y.W. Na, J.Y. Han, J.S. Jeong, V.V. Shastina,
P.P. Medina, A. Parra, M. Cascante, J.A. Lupianez, Maslinic acid, a natural tri- V.A. Stonik, J.I. Park, J.Y. Kwak, Stichoposide C induces apoptosis through the
terpene, induces a death receptor-mediated apoptotic mechanism in Caco-2 p53- generation of ceramide in leukemia and colorectal cancer cells and shows in vivo
deficient colon adenocarcinoma cells, PLoS One 11 (2016) e0146178, , https:// antitumor activity, Clin. Cancer Res. 18 (2012) 5934–5948, https://doi.org/10.
doi.org/10.1371/journal.pone.0146178. 1158/1078-0432.ccr-12-0655.
[197] M. Kumazaki, H. Shinohara, K. Taniguchi, N. Yamada, S. Ohta, K. Ichihara, [214] H. Yu, H. Zhang, M. Dong, Z. Wu, Z. Shen, Y. Xie, Z. Kong, X. Dai, B. Xu, Metabolic
Y. Akao, Propolis cinnamic acid derivatives induce apoptosis through both ex- reprogramming and AMPKalpha1 pathway activation by caulerpin in colorectal
trinsic and intrinsic apoptosis signaling pathways and modulate of miRNA ex- cancer cells, Int. J. Oncol. 50 (2017) 161–172, https://doi.org/10.3892/ijo.2016.
pression, Phytomedicine 21 (2014) 1070–1077, https://doi.org/10.1016/j. 3794.
phymed.2014.04.006. [215] J.R. Rho, B.S. Hwang, C.J. Sim, S. Joung, H.Y. Lee, H.J. Kim, A. Phorbaketals, B,
[198] Y. Zhang, Y. Wei, Z. Zhu, W. Gong, X. Liu, Q. Hou, Y. Sun, J. Chai, L. Zou, T. Zhou, and C, sesterterpenoids with a spiroketal of hydrobenzopyran moiety isolated from
Icariin enhances radiosensitivity of colorectal cancer cells by suppressing NF- the marine sponge Phorbas sp, Org. Lett. 11 (2009) 5590–5593, https://doi.org/
kappaB activity, Cell Biochem. Biophys. 69 (2014) 303–310, https://doi.org/10. 10.1021/ol902223m.
1007/s12013-013-9799-x. [216] A. Abdel-Lateff, A.M. Al-Abd, A.M. Alahdal, W.M. Alarif, S.E. Ayyad, S.S. Al-
[199] F. Hu, F. Wei, Y. Wang, B. Wu, Y. Fang, B. Xiong, EGCG synergizes the therapeutic Lihaibi, M.E. Hegazy, A. Al Mohammadi, T.M. Abdelghany, A.B. Abdel-Naim,
effect of cisplatin and oxaliplatin through autophagic pathway in human color- M.A. Moustafa, Z.M. Banjer, A.S. Azhar, Antiproliferative effects of triterpenoidal
ectal cancer cells, J. Pharmacol. Sci. 128 (2015) 27–34, https://doi.org/10.1016/j. derivatives, obtained from the marine sponge Siphonochalina sp., on human he-
jphs.2015.04.003. patic and colorectal cancer cells, Z. Naturforsch. C 71 (2016) 29–35, https://doi.
[200] S.W. Hsuan, C.C. Chyau, H.Y. Hung, J.H. Chen, F.P. Chou, The induction of org/10.1515/znc-2015-0160.
apoptosis and autophagy by Wasabia japonica extract in colon cancer, Eur. J. Nutr. [217] D. Trisciuoglio, B. Uranchimeg, J.H. Cardellina, T.L. Meragelman, S. Matsunaga,
55 (2016) 491–503, https://doi.org/10.1007/s00394-015-0866-5. N. Fusetani, D. Del Bufalo, R.H. Shoemaker, G. Melillo, Induction of apoptosis in
[201] F. Farokhi, P. Grellier, M. Clement, C. Roussakis, P.M. Loiseau, E. Genin-Seward, human cancer cells by candidaspongiolide, a novel sponge polyketide, J. Natl.
J.M. Kornprobst, G. Barnathan, G. Wielgosz-Collin, Antimalarial activity of ax- Cancer Inst. 100 (2008) 1233–1246, https://doi.org/10.1093/jnci/djn239.
idjiferosides, new beta-galactosylceramides from the African sponge Axinyssa [218] X. Chen, H. Gao, Y. Han, J. Ye, J. Xie, C. Wang, Physcion induces mitochondria-
djiferi, Mar. Drugs 11 (2013) 1304–1315, https://doi.org/10.3390/md11041304. driven apoptosis in colorectal cancer cells via downregulating EMMPRIN, Eur. J.
[202] N. Hatae, R. Satoh, H. Chiba, T. Osaki, T. Nishiyama, M. Ishikura, T. Abe, Pharmacol. 764 (2015) 124–133, https://doi.org/10.1016/j.ejphar.2015.07.008.
S. Hibino, T. Choshi, C. Okada, E. Toyota, N-Substituted calothrixin B derivatives [219] J. Chen, C. Wang, W. Lan, C. Huang, M. Lin, Z. Wang, W. Liang, A. Iwamoto,
inhibited the proliferation of HL-60 promyelocytic leukemia cells, Med. Chem. X. Yang, H. Liu, Gliotoxin inhibits proliferation and induces apoptosis in colorectal
Res. 23 (2014) 4956–4961, https://doi.org/10.1007/s00044-014-1061-6. Cancer cells, Mar. Drugs 13 (2015) 6259–6273, https://doi.org/10.3390/
[203] S. Freitas, R. Martins, A. Campos, J. Azevedo, H. Osorio, M. Costa, P. Barros, md13106259.
V. Vasconcelos, R. Urbatzka, Insights into the potential of picoplanktonic marine [220] S.P. Gunasekera, L. Imperial, C. Garst, R. Ratnayake, L.H. Dang, V.J. Paul,
cyanobacteria strains for cancer therapies - cytotoxic mechanisms against the RKO H. Luesch, Caldoramide, a Modified Pentapeptide from the Marine
colon cancer cell line, Toxicon. 119 (2016) 140–151, https://doi.org/10.1016/j. Cyanobacterium Caldora penicillata, J. Nat. Prod. 79 (2016) 1867–1871, https://
toxicon.2016.05.016. doi.org/10.1021/acs.jnatprod.6b00203.
[204] W. Soontornchaiboon, S.S. Joo, S.M. Kim, Anti-inflammatory effects of violax- [221] T. Matsubara, M. Yokoya, N. Sirimangkalakitti, N. Saito, Asymmetric synthesis and
anthin isolated from microalga Chlorella ellipsoidea in RAW 264.7 macrophages, cytotoxicity evaluation of right-half models of antitumor renieramycin marine
Biol. Pharm. Bull. 35 (2012) 1137–1144 https://www.jstage.jst.go.jp/article/bpb/ natural products, Mar. Drugs 17 (2018), https://doi.org/10.3390/md17010003.
35/7/35_b12-00187/_pdf. [222] L.C. Lin, T.T. Kuo, H.Y. Chang, W.S. Liu, S.M. Hsia, T.C. Huang, Manzamine a
[205] M. Hosokawa, M. Kudo, H. Maeda, H. Kohno, T. Tanaka, K. Miyashita, exerts anticancer activity against human colorectal Cancer cells, Mar. Drugs 16
Fucoxanthin induces apoptosis and enhances the antiproliferative effect of the (2018), https://doi.org/10.3390/md16080252.
PPARgamma ligand, troglitazone, on colon cancer cells, Biochim. Biophys. Acta

13

You might also like