Next Article in Journal
Erlotinib Promotes Ligand-Induced EGFR Degradation in 3D but Not 2D Cultures of Pancreatic Ductal Adenocarcinoma Cells
Next Article in Special Issue
Sulforaphane: A Broccoli Bioactive Phytocompound with Cancer Preventive Potential
Previous Article in Journal
Tackling Refractory Metastatic Colorectal Cancer: Future Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential of Bioactive Food Components against Gastric Cancer: Insights into Molecular Mechanism and Therapeutic Targets

1
College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Korea
2
Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Korea
3
Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
4
Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Korea
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(18), 4502; https://doi.org/10.3390/cancers13184502
Submission received: 26 July 2021 / Revised: 2 September 2021 / Accepted: 3 September 2021 / Published: 7 September 2021

Abstract

:

Simple Summary

Recently, it has been found that cancer of the gastrointestinal tract, especially gastric cancer (GC), is the second most leading cause of cancer-related death globally. Extensive research has shown that most epidemiological investigations indicated the increased intake of naturally-occurring bioactive food components could decrease the gastric cancer risk. Several experimental studies have explained that the molecular mechanisms of action to prevent GC comprise induction of apoptosis, inhibition of cell proliferation, suppression of angiogenesis and metastasis, and regulation of autophagy. To provide an updated understanding of relationships between naturally occurring bioactive food components and gastric cancer, this study will be helpful for guiding and preventing gastric cancer by natural bioactive food products.

Abstract

Gastric cancer, also known as stomach cancer, is a cancer that develops from the lining of the stomach. Accumulated evidence and epidemiological studies have indicated that bioactive food components from natural products play an important role in gastric cancer prevention and treatment, although its mechanism of action has not yet been elucidated. Particularly, experimental studies have shown that natural bioactive food products display a protective effect against gastric cancer via numerous molecular mechanisms, such as suppression of cell metastasis, anti-angiogenesis, inhibition of cell proliferation, induction of apoptosis, and modulation of autophagy. Chemotherapy remains the standard treatment for advanced gastric cancer along with surgery, radiation therapy, hormone therapy, as well as immunotherapy, and its adverse side effects including neutropenia, stomatitis, mucositis, diarrhea, nausea, and emesis are well documented. However, administration of naturally occurring bioactive phytochemical food components could increase the efficacy of gastric chemotherapy and other chemotherapeutic resistance. Additionally, several studies have suggested that bioactive food components with structural stability, potential bioavailability, and powerful bioactivity are important to develop novel treatment strategies for gastric cancer management, which may minimize the adverse effects. Therefore, the purpose of this review is to summarize the potential therapeutic effects of natural bioactive food products on the prevention and treatment of gastric cancer with intensive molecular mechanisms of action, bioavailability, and safety efficacy.

1. Introduction

The incidence and mortality of cancer is growing worldwide, with an estimated 19.3 million new cases and 10 million cancer deaths in 2020 [1]. Gastric cancer is the fifth most common neoplasm and the fourth leading cause of cancer death, which has led to over one million new cases and an estimated 769,000 deaths in 2020 [1]. Clinically, to offer pertinent treatment, gastric carcinoma is classified as early or advanced stage [2]. Gastric carcinoma has multiple risk factors: genetics, Helicobacter pylori infection, gastric ulcer, gastroesophageal reflux disease (GERD), tobacco, smoking, alcohol, chemical exposure, diet, obesity, and so forth [3,4]. Surgical resection, when possible, offers the best chances of cure for early gastric cancer [5]. Adjuvant or neoadjuvant chemotherapy may be beneficial in increasing the chance of successful resection or in decreasing the rate of recurrence and/or metastasis [6,7,8]. For patients with unresectable advanced gastric cancer, chemotherapy is a common choice. Conventional regimens are mostly based on cytotoxic agents including antimetabolites and platinum-based anticancer drugs. However, these regimens cause severe side effects such as chemotherapy-induced peripheral neuropathy (CIPN), neutropenia, stomatitis, mucositis, diarrhea, nausea, and emesis [9,10]. Moreover, failure of first-line chemotherapy due to resistance is also an obstacle of gastric cancer treatment hampering the novel and effective therapies and imposing significant economic costs to patients [11]. Moreover, exposure to unremovable toxins (not able to be removed or non-releasable), trauma, or infection lead to mutagenic chronic inflammatory responses, which cause dysplasia [12]. Considering gastric cancer, Helicobacter pylori infection is a major risk factor for developing deleterious tumor microenvironments [13]. Nuclear factor kappa-B (NF-κB), c-Jun N-terminal kinase (JNK), and signal transducer activator of transcription 3 (STAT3), inflammatory cytokines, tumor necrosis factor (TNF), interleukin (IL)-1/6, tumor-derived cytokines such as fasciclin (Fas) ligand, and vascular endothelial growth factor (VEGF) are major targets of regulation for the prevention and treatment of gastric cancer [14,15,16,17,18]. Therefore, novel drug development against gastric cancer is strongly needed to further improve survival rates of this disease and lower the side effects of conventional therapies.
Epidemiological studies have shown that natural dietary bioactive food components decrease the risks of gastric cancer [19,20,21,22]. Extensive research was conducted to measure the value of natural products for the prevention and treatment of gastric carcinoma, leading to the discovery of major bioactive phytochemicals with anti-cancer properties, such as quercetin, silymarin, taurine, berberine, curcumin, and so forth [23,24,25,26]. However, few review articles included agents from animal or marine sources, which are also being studied with growing expectation [27,28]. The same goes for traditional medicine, despite their wide use in clinical practice to combat various illnesses including cancer [29,30,31,32]. This review explores various bioactive compounds isolated from biological resources of bioactive food components and traditional medicine in the form of single compounds that show anti-cancer properties closely targeted to gastric cancer. Moreover, the use of bioactive food components could be a promising adjuvant remedy for gastric cancer treatment as well as in developing functional food components and drugs for the treatment and prevention of gastric cancer.

2. Methods

While there have been similar reviews highlighting the anti-neoplastic efficacies of bioactive food components, few of them were written with regards to the chemical classification of each bioactive compound. This review is not only a simple compilation of previous in vitro studies testing bioactive food components on gastric cancer but goes as far as to systematically organizing previous works depending on each cancer-related pathway, namely apoptosis, autophagy, metastasis, drug-resistant capability, and more. Literature-based online databases, Google Scholar, Web of Science, PubMed, Google, and Scopus were accessed to collect information on the published articles. As there is currently no golden standard for classifying phytochemicals, we adopted a comprehensive and clear method previously demonstrated in a literature highlighting the efficacies of bioactive food components on gastrointestinal diseases. This will help researchers rule out or select appropriate candidate species of natural bioactive food products for further studies. This review only included studies published from 2014 to 2021.

3. Apoptosis-Inducing Natural Bioactive Food Components in Gastric Cancer

Apoptosis is the process of programmed cell death, characterized by distinct morphology: cell shrinking, membrane blebbing, chromatin condensation, and nuclear fragmentation [33,34]. Several bioactive compounds showing apoptosis-inducing effects on gastric cancer cells and animal models are presented in Figure 1 and Table 1. Yang et al. reported that berberine could inhibit the proliferation of SGC-7901 cells and induce apoptosis [35]. In vitro models have demonstrated that cyclovirobuxine D originated from Buxux microphylla Richardii. Radix (Buxaceae) induced apoptosis in MGC-803 and MKN-28 cells [36]. Expressions of caspase-3, cytochrome c, endonuclease G (Endo G), apoptosis inducing factor (AIF), and Smac/Diablo were upregulated in melittin-treated SGC-7901 cells. Trifolirhizin, a compound isolated from Sophora flavescens Aiton Radix (Fabaceae), demonstrated apoptotic activity both in vitro and in vivo [37]. Trifolirhizin induced apoptosis of MKN-45 cells in vitro via EGFR-MAPK pathways and triggered G2/M phase cell cycle arrest by impacting the CDC2/Cyclin B complex. Qian et al. discovered that ginsenoside-Rh2 originated from Panax ginseng C.A. Mey, Radix (Araliaceae) inhibits proliferation and induces apoptosis of SGC-7901 cells by induction of the Bcl-like protein 4 (Bax) to Bcl-2 (Bax/Bcl-2) ratio [38].
Tanshinone IIA, originated from Salviae miltiorrhiza Bunge. Radix (Lamiaceae), suppressed AGS gastric tumor cells via activation of tumor necrosis factor-alpha (TNF-α), Fas, p38, JNK, p53, p21, caspase-3, and caspase-8 and inhibition of ERK [39]. [6]-gingerol treatment for 24 h to AGS cells generated ROS and decreased ΔΨm, leading to induction of apoptosis. Perturbations of ΔΨm were associated with deregulation of the Bax/Bcl-2 ratio at the protein level, which led to the upregulation of cytochrome c and triggered the caspase cascade. 2,7-dihydroxy-3-methylanthraquinone (DDMN), a flavone isolated from Hedyotis diffusa Willd. Herba, induced caspase-dependent apoptosis of SGC-7901 gastric cancer cells [40]. 6,7,30-trimethoxy-3,5,40-trihydroxy flavone (TTF), from Chrysosplenium nudicaule Ledeb. Herba, is a well-known traditional Chinese medicine for digestive diseases [41], which induced apoptosis on SGC-7901 cells. Sun et al. observed that curcumin, isolated from Curcuma longa L. Rhizoma (Zingiberaceae), induced apoptosis of SGC-7901 and BGC-823 cells by up-regulating microRNA-33b (miR-33b) expression [42]. Esculetin treatment triggered ROS formation, elevated caspase-3/9 activity, and induced poly (ADP-ribose) polymerase (PARP) cleavage [43]. Liu et al. reported that hydroxysafflor yellow A (HSYA) induces apoptosis of BGC-7901 gastric carcinoma cells via activation of the peroxisome proliferator-activated receptor gamma (PPARγ) signal through elevation of PPARγ and caspase-3 [44]. Kurarinone synergized TRAIL-induced apoptosis against gastric cancer cell line SGC-7901 [45]. Licochalcone A (LicA), a flavonoid isolated from licorice root, elucidated apoptosis by blocking the Akt signaling pathway and reducing hexokinase 2 (HK2) expression in MKN45 cells [46]. Curcuzedoalide, sesquiterpene bioactive components of Curcuma zedoaria Roscoe Rhizoma (Zingiberaceae), induced mitochondrial apoptosis induction with cleavage of PARP as well as caspase-8, caspase-9, and caspase-3 in AGS cells [47]. Thymol showed cytotoxicity on AGS cancer cells via the intrinsic mitochondrial pathway via upregulation of Bax and PARP expression, and also promoted cleavage of caspase-7, caspase-8, and caspase-9 and downregulated ΔΨm [48].
The apoptotic ability of ophiopogonin B, the active compound isolated from Ophiopogon japonicus Radix, against SGC-7901 cells were suspected to be relevant with the JNK 1/2 and ERK1/2 signaling pathways through upregulation of active caspase-3 and modulation of Bax/Bcl-2 expression [49]. It has been found that phloretin, a plant-derived natural bioactive product, is an important molecule for the treatment of AGS gastric cancer via expression of Bax and was increased in dose-dependently while the expression of Bcl-2 decreased [50]. Podophyllotoxin, isolated from Linum album Kotschy (Linaceae), induced apoptosis and downregulated zinc finger protein 703 oncogene expression [51]. Grifolin, isolated from the mushroom Albatrellus confluens (Alb. and Schwein) Kotl. and Pouzar (Albatrellaceae), inhibited growth and invasion of gastric cancer cells by inducing apoptosis and suppressing the ERK1/2 pathway [52]. Tsai et al. found that 7-acetylsinumaximol B (7-AB), discovered from Sinularia sandensis (Alcyoniidae), showed anti-proliferative effects through apoptosis against human gastric carcinoma NCI-N87 cells via the expression of Bad, Bcl-like protein 11 (Bim), Bax, and cytochrome c, and it decreased the expression levels of phosphorylated Bad (p-Bad), myeloid cell lukemia-1 (Mcl-1), Bcl-xL, and Bcl-2 proteins. [53] Crosolic acid, isolated from Actinidia valvata Dunn. Radix (Actinidiaceae), was reported to inhibit proliferation of BGC-823 cells by downregulating the NF-κB pathway [54]. Crosolic acid inhibited phosphorylation of nuclear factor kappa B-alpha (IκBα), expression of p65, and nuclear translocation and DNA-binding activity of NF-κB. Deacetylisovaltratum, derived from Patrinia heterophylla Bunge, induced mitochondrial and caspase-dependent apoptosis in AGS and HGC-27 cells [55]. Li et al. demonstrated that elemene, a sesquiterpenoid mixture isolated from a traditional herbal medicine, Curcuma zedoaria Roscoe Rhizoma (Zingiberaceae), countered gastric cancer via regulation of the ERK 1/2 signaling pathway [56]. Liao et al. reported that n-butylidenephthalide (BP), a bioactive compound of Angelica Sinensis Diels Radix, activated the intrinsic apoptotic pathway of human gastric cancer cells AGS, NCI-N87, and TSGH-9201 [57]. Paeonol treatment inhibited proliferation, invasion, migration, and induced apoptosis against BGC823 cells. The protein expression of matrix metalloproteinase (MMP)-2 and MMP-9 were attenuated in a concentration-dependent manner by paeonol [58]. Pseudolaric acid B, isolated from Pseudolarix amabilis, commonly called golden larch, inhibited cell proliferation and induced apoptosis of the multidrug-resistant SGC-7901/ADR gastric cancer cell line [59].
Thymol is a phenolic compound isolated from Thymus quinquecostatus Celak. (Lamiaceae) that possesses anti-inflammatory, anticancer, antibacterial, and more biological efficacies [48]. The anticancer potencies of toosendanin (TSN), a triterpenoid found in Melia toosendan Sieb et Zucc Cortex et Fructus (Meliaceae), was discussed in two studies. Wang et al. found that SGC-7901 cells treated with toosendanin (TSN) increased early apoptosis [60]. TSN inactivated the β-catenin pathway in SGC-7901 cells and subsequently induced apoptosis following facilitation of microRNA 200a [60]. It has been reported that peptic oligosaccharide, separated from Solanum lycopersicum L. (Solanaceae), induced apoptosis by suppressing galectin-3 expressions [61]. Additionally, several natural bioactive products retarded tumor growth in animal models, as presented in Table 2. Wu et al. revealed that phenolic alkaloids of Menispermum dauricum induced apoptosis and suppressed gastric tumor growth by inducing apoptosis and inhibiting oncogenic Kirsten Rat sarcoma viral oncogene homolog (K-RAS) expression [62]. When BALB/C mice grafted with MFC mouse gastric cancer cells were treated with curcumin solution every day for 60 days, expressions of interferon gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), granzyme B, and perforin were upregulated, while differentiated embryonic chondrocyte gene 1 (DEC1), hypoxia-inducible factor-1 alpha (HIF-1α), STAT3, and VEGF expression were downregulated in the experimental group [63]. When MKN45-treated BALB/ca mice were treated with LicA, tumor growth was significantly inhibited in contrast to the vehicle group without LicA treatment [46]. Elemene retarded tumor growth in nude mice and showed better efficacy when synergized with PD98059 [56]. In a xenograft mouse model, mice treated with grifolin survived for a longer period compared to the control group [52].
Table 1. Apoptosis-inducing bioactive food components in vitro. (↑ increase, ↓ decrease).
Table 1. Apoptosis-inducing bioactive food components in vitro. (↑ increase, ↓ decrease).
ClassificationCompoundSourceExperimental ModelDose; DurationEfficacyMechanismReferences
AlkaloidsBerberine(family: Ranunculaceae)
Coptidis japonica Makino Rhizoma
SGC-79015, 10, 20 µM; 24, 48 hInduction of apoptosis [35]
AlkaloidsCyclovirobuxine D(family: Buxaceae)
Buxus microphylla Richardii Radix
MGC-803, MKN-2830, 60, 120 μM/L; 48 hInduction of apoptosis↑c-caspase-3, Bax
↓Bcl-2
[36]
AlkaloidsGFG-3a(family: Meripilaceae)
Grifola frondose (Diks.)
Gray Mycelia
SGC-7901100, 200 μg/mL; 24, 48 hInduction of apoptosis↑RBBP4, caspase-3, -8, p53, Bax, Bad
↓RUVBL, NPM, Bcl-2, Bcl-xL, PI3K, Akt1
[64]
AlkaloidsMelittin(family: Apidae)
Apis cerena Fabricius venom
SGC-79014 μg/mL; 1, 2, 4 hInduction of apoptosis↑caspase-3, cyt c, Endo G, AIF, Smac/Diablo, ROS
↓ΔΨm
[65]
Alkaloids,
Terpenoids
Berberine,
d-Limonene
(1)
(family: Ranunculaceae)
Coptidis japonica Makino Rhizoma
(2) (family: Rutaceae)
Evodiae rutaecarpa Bentham. Fructus
MGC-803(1) 20 µM; 24, 36, 48 h
(2) 80 µM; 24, 36, 48 h
Induction of apoptosis↑ROS, caspase-3
↑ΔΨm, Bcl-2
[66]
FlavonoidsTrifolirhizin(family: Fabaceae) Sophora flavescens Aiton RadixMKN-4520, 30, 40 µg/mL; 48 hInduction of apoptosis↑caspase-9, -3, c-PARP, p53, p38
↓EGFR, CDC2, cyclin B, ΔΨm
[37]
PhytosterolsGinsenoside-Rh2(family: Araliaceae) Panax ginseng C.A. Mey RadixSGC-79015, 10, 20 μg/mL; 24, 48 hInduction of apoptosis↑Bax
↓Bcl-2
[38]
PhytosterolsPeriplocin(family: Apocynaceae) Periplocae sepium Bunge.SGC-7901, MGC-803, BGC-82350, 100, 200 ng/mL; 24, 48 hInduction of apoptosis↑Mcl-1, c-caspase-3, EGR 1
↓pro-Bid, p-ERK 1/2
[67]
PhytosterolsTanshinone IIA(family: Lamiaceae) Salviae miltiorrhiza Bunge. RadixAGS2.0, 3.7, 5.5 µg/mL; 24, 48 hInduction of apoptosis↑TNF-α, Fas, p-p38, p-JNK, p53, p21, caspase-8, -3
↓p-ERK, CDC2, cyclin A, cyclin B1
[39]
Polyphenols [6]-Gingerol(family: Zingiberaceae) Zingiber officinale Roscoe RhizomaAGS100, 250 µM; 24 hInduction of apoptosis↑cyt c, Bax
↓Bcl-2
[68]
Polyphenols2,7-dihydroxy-3-methylanthraquinone (DDMN)(family: Rubiaceae) Hedyotis diffusa Wild HerbaSGC-790110, 20, 40 µM; 48 hInhibition of proliferation↑Bax, Bad, caspase-3, -9, cyt c
↓Bcl-xL, Bcl-2
[40]
Polyphenols6, 7, 30-trimethoxy-3, 5, 40
-trihydroxy flavone (TTF)
(family: Saxifragaceae)
Chrysosplenium nudicaule Ledeb Herba
SGC-79012, 4, 8, 16, 32 µg/mL; 24, 48, 72 hInduction of apoptosis↑endogenous Ca2+/Mg2+ dependent endonuclease [41]
PolyphenolsCurcumin(family: Zingiberaceae)
Curcuma longa L. Rhizoma
SGC-7901, BGC-8235, 10, 15, 20, 40 μM/L; 24 hInduction of apoptosis↓XIAP
↑miR-33b
[42]
PolyphenolsEsculetin(family: Asteraceae) Artemesia scoparia Waldst. et Kit, Artemesia capillaris Thunb.))
(family: Plumbaginaceae)
SGC-7901, MGC-803, BGC-82312.5, 25, 50 μM; 24 hInduction of apoptosis↑ROS, c-caspase-9, -3, c-PARP, cyt c, Bak, Bax, CypD
↓Bcl-2, Bcl-xL, XIAP
[43]
PolyphenolsHydroxysafflor Yellow A(family: Asteraceae) Carthamus tinctorius L.BGC-823100 µM; 48 hInduction of apoptosis↑caspase-3, PPARγ [44]
PolyphenolsKurarinone(family: Fabaceae) Sophora flavescens Aiton RadixSGC-79015 μM; 24 hEnhancement ofTRAIL-induced apoptosis↓Mcl-1, c-FLIP, p-STAT3 [45]
PolyphenolsLicochalcone A(family: Fabaceae) Glycyrrhiza glabra L. RootMKN-45, SGC-790115, 30, 60 µM; 24 hInhibition of cell proliferation and tumor glycolysis↑c-caspase-3, c-PARP
↓Bcl-2, Mcl-1, HK2, p-Akt, p-ERK1/2, p-S6, p-GSK3β
[46]
PolyphenolsOphiopogonin B(family: Asparagaceae)
Ophiopogon japonicus Thunb Root
SGC-79015, 10, 20 μMInduction of apoptosis↑ROS, Bax, caspase-3
↓p-ERK 1/2, p-JNK 1/2, ΔΨm, Bcl-2
[49]
PolyphenolsPhloretin AGS4, 8, 16µM; 24 hInduction of apoptosis
Inhibition of invasion
↑Bax
↓Bcl-2
[50]
PolyphenolsPodophyllotoxin(family: Linaceae)
Linum album Kotschy
AGS200, 400, 600, 800, 1000 µg/mL; 24 hInduction of apoptosis↓ZNF703 [51]
Terpenoids7-Acetylsinumaximol B(family: Alcyoniidae) Sinularia sandensisNCI-N874, 8, 16 µM; 24 hInduction of apoptosis↑Bad, Bim, Bax, cyt c
↓p-Bad, Mcl-1, Bcl-xL, Bcl-2
[53]
TerpenoidsCrosolic Acid(family: Actinidiaceae)
Actinidia valvata Dunn Radix
BGC-82320, 40, 80 μg/mL; 72 hInduction of apoptosis↑Bax, smac, IκBα
↓Fas, Bcl-2, p65, p-IκBα, NF-κB
[54]
TerpenoidsCurcuzedoalide(family: Zingiberaceae)
Curcuma zedoaria Roscoe Rhizoma
AGS100, 200 µM; 24 hInduction of apoptosis↑c-caspase-8, -9, -3, c-PARP [47]
TerpenoidsDeacetylisovaltratum(family: Caprifoliaceae)
Patrinia heterophylla Bunge.
(1) AGS
(2) HGC-27
(1) 4, 8, 16 μM; 24 h
(2) 10, 20, 30 μM; 24 h
Induction of apoptosis↑p21, caspase-3, c-PARP
↓p-STAT3, pro-caspase-9, ΔΨm
[55]
TerpenoidsElemene(family: Zingiberaceae)
Curcuma zedoaria Roscoe Rhizoma
BGC-82320, 40, 80, 160 μg/mL: 24 hInduction of apoptosis↑Bax, p-ERK 1/2
↓Bcl-2
[56]
TerpenoidsGrifolin(family: Albatrellaceae)
Albatrellus confluens (Alb. and Schwein.) Kotl. and Pouzar
BGC-823, SGC-790110, 50 µM; 48 hInduction of apoptosis↑caspase-9, -3, CDKN2 ↓MEK1, MEKK3 MEK5 [52]
TerpenoidsN-butylidenephthalide(family: Apiaceae) Angelica Sinensis Diels RadixAGS25, 50, 75 µg/mL; 24 hInduction of apoptosis↑REDD1
↓mTOR
[57]
TerpenoidsPaeonol(family: Paeoniaceae) Paeonia suffruticosa Andr Root bark,
(family: Apocynaceae) Cynanchum paniculatum K. Schum Radix
BGC-8230.1, 0.2, 0.4 mg/mL; 24, 48 hInhibition of proliferation, invasion, and migration
Induction of apoptosis
↓MMP-2, -9 [58]
TerpenoidsPseudolaric acid B(family: Pinaceae) Pseudolarix kaempferi Gorden Root barkSGC-7901/ADR5, 10, 20 μM/L; 24 hInduction of apoptosis↑p53, Bax
↓P-gp, COX-2, Bcl-2, Bcl-xL
[59]
TerpenoidsThymol(family: Lamiaceae)
Thymus quinquecostatus Celak Essential oil
AGS100, 200, 400 µM; 6, 12, 24 hInduction of apoptosis↑Bax, c-PARP, caspase-8, caspase-7, caspase-9
↓ΔΨm
[48]
TerpenoidsToosendanin(family: Meliaceae)
Melia toosendan Sieb et zucc Cortex or Fructus
SGC-79010.5, 1 µM; 48 hInhibition of invasion, migration and EMT
Induction of apoptosis
↑E-cadherin
↓β-catenin
[60]
↑miR-200a
Table 2. Apoptosis-inducing bioactive food components in vivo. (↑ increase, ↓ decrease).
Table 2. Apoptosis-inducing bioactive food components in vivo. (↑ increase, ↓ decrease).
ClassificationCompoundSourceExperimental ModelDose; DurationEfficacyMechanismReferences
AlkaloidsPhenolic alkaloids(family: Menispermaceae)
Menispermum dauricum DC. Rhizoma
Nude mice/SGC-79015, 10, 20 mg/kg/week; 3 weeksSuppression of tumor growth [62]
FlavonoidsTrifolirhizin(family: Fabaceae)
Sophora flavescens Aiton. Radix
BALB/C nude mice/MKN-451–3 mg/kg; 3 weeksRetardation of tumor growth↑c-caspase-3
↓ΔΨm
[37]
Polyphenols2,7-dihydroxy-3-methylanthraquinone (DDMN)(family: Rubiaceae)
Hedyotis diffusa Wild. Herba
nude mice/SGC-790140 mg/kg; 5, 10, 15, 20 daysInhibition of gastric cancer cell growth↑Bax, Bad, c-caspase-3, -9, cyt c
↓Bcl-xL, Bcl-2
[40]
PolyphenolsCurcumin(family: Zingiberaceae)
Curcuma longa L. Rhizoma
BALB/C mice/MFC20, 40, 60 μM/L/day; 60 daysInhibition of tumor growth
Induction of apoptosis
Activation of immune cells
↑IFN-γ, TNF-α, granzyme B, perforin
↓DEC1, HIF-1α, STAT3, VEGF
[63]
PolyphenolsLicochalone A(family: Fabaceae)
Glycyrrhiza glabra L. Radix
BALB/ca nude mice/MKN-4510 mg/kg/day; 33 daysInhibition of tumor growth [46]
TerpenoidsElemene(family: Zingiberaceae)
Curcuma longa L. Rhizoma
BALB/c athymic nude mice/BGC-823200 mg/kg/day; 15 daysRetardation of tumor growth [56]
TerpenoidsGrifolin(family: Albatrellaceae)
Albatrellus confluens (Alb. and Schwein.) Kotl. and Pouzar
Balb/c nude mice/BGC-823, SGC-790115 mg/kg; 2 daysImprovement of survival time [52]

4. Role of Autophagy in Gastric Cancer Treatment Mediated by Natural Bioactive Food Products

Autophagy is a cellular process in which cytoplasmic contents are degraded within the lysosome/vacuole, and the resulting constituents are recycled [69,70]. Autophagy can be classified into macroautophagy, microautophagy, and chaperone-mediated autophagy (CMA) [71]. Among these, macroautophagy, which has been studied the most, is the process of forming autophagosomes that surround organelles and fuse with lysosomes, and natural products modulate autophagy [72,73]. Based on the isolation target, separate kinds of selective autophagy such as mitophagy, pexophagy, and xenophagy can be distinguished [74]. Macroautophagy consists of several sequential steps: initiation, nucleation, elongation, maturation, and fusion with the lysosome [73,75]. Phagosomes originate from omegasomes, subdomains of the ER, and associate with other organelles such as the mitochondria, golgi complex, plasma membrane, recycling endosome, etc., during its development. Four molecules, Unc-51-like kinase 1/2 (ULK1/2), autophagy-related gene 13 (ATG13), family 200-kD interacting protein (FIP200), and Atg101 form the ULK1/2 complex and initiate the process [73]. The mechanistic target of rapamycin complex 1 (mTORC1) is a major inhibitor of the ULK1/2 complex [69,76]. AMP-activated protein kinase (AMPK) inhibits mTORC1 and leads to the activation of the ULK1/2 complex [75]. The ULK1/2 complex phosphorylates the class III phosphatidylinositol-3-kinase (PI3K) vacuole protein sorting 34 (VPS34) complex consisting of VPS15, Beclin-1, and AtG14 complex, which promotes the formation of phosphatidylinositol-3-phosphate (PI3P), which is an essential lipid molecule required for the nucleation step of the phagophore [77]. Atg12 binds with Atg5 and composes a complex with Atg16L. The Atg12-5-16L1 complex lipidates LC3-I into LC3-II [78,79]. LC3-II, considered a marker of autophagy, is essential for phagosome elongation and fusion [80,81]. When the phagosome encloses and becomes a mature autophagosome, it fuses with a lysosome, and degradation and recycling processes follows. Bioactive food compounds were reported to induce autophagy along with apoptosis against gastric cancer cells, as presented in Figure 2.
It has been found that cinnamaldehyde, the bioactive ingredient in Cinnamomum cassia, suppressed tumor growth and the migratory and invasive abilities of gastric cancer [82]. Rottlerin, isolated from Mallotus philipensis Muell (Euphorbiaceae), induced autophagy and caspase-independent apoptosis against SGC-7901 and MGC-803 cells by downregulating mTOR and S-phase kinase-associated protein 2 (Skp2) [83]. Moreover, treatment of latcripin 1 protein, found in Lentinula edodes, activated autophagy of gastric cancer cell lines BGC-823 and SGC-7901 with autophagosome formation via the alteration of LC3-I into LC3-II expression [84]. Oxyresveratrol, found in grape, has been found to accumulate ROS production and initiated autophagic and apoptotic cell death via the FOXO-caspase-3 pathway [85,86]. Kaempferol, a natural bioactive flavonoid, induced autophagic cell death in gastric cancer via IRE1/JNK/CHOP and AMPK/ULK1 pathways [87]. It has demonstrated cytotoxic activity on AGS, MKN-45, and KATO-III human gastric cancer cells via induction of caspase activation and autophagy via the Akt/NF-κB pathway in AGS cells [22]. Pectolinarigenin, isolated from Cirsium chanroenicum, displayed anticancer activity through autophagy induction of human gastric cancer AGS and MKN-28 cells via the downregulation of the PI3K/Akt/mTOR pathway [88]. Perillaldehyde increased AMPK phosphorylation, leading to autophagy in human gastric cancer MFCs mouse and GC9811-P cells [89]. However, quercetin activated autophagy protection against the apoptosis in AGS and MKN-28 gastric cancer cells, which signified that autophagy might have contributed to the survival of cancer cells [90]. Therefore, autophagy induction by natural bioactive compounds might possibly be targeted as a potential therapeutic approach to control gastric cancer.

5. Role of Bioactive Natural Compounds to Arrest Cell Cycle in Gastric Cancer

The cell cycle is regulated through a series of control systems that in turn promote or inhibit cell division. Programmed cell death and cell cycle regulation occur together in many cancerous cells, since the tumor suppressor gene p53 and downstream proteins regulate both events [91]. A variety of natural bioactive components were described as causing cell death and inhibited cell proliferation by seizing the cell cycle according to the phase of cell cycle arrest (Table 3). Berberine, a traditional Chinese medicine normally used for gastroenteritis, inhibited proliferation of SGC-7901 gastric cancer cells in addition to inducing G1 arrest in the cell cycle phase and activated apoptosis [35]. Toosendanin, a triterpenoid, increased the proportion of cells in the G1 and S phase by activation of β-catenin signaling in gastric carcinoma [60,92]. Moreover, ginsenoside-Rh2 inhibited proliferation of SGC-7901 side population gastric cancer cells by the induction of cell cycle arrest, as well as cell apoptosis, and altered BAX/Bcl-2 protein expression [38]. Crosolic acid, isolated from Actinidia valvata Dunn. Radix, increased the sub G1 population of the cell cycle and decreased p65, bcl-2, Fas, and smac mRNA expression, and increased IκBα, bax, and survivin mRNA expression, which induced apoptosis of the human gastric cancer cell line BGC823 through down-regulation of the NF-κB pathway [54]. It has been found that rottlerin suppressed cell growth, induced autophagy as well as apoptosis, and reduced migration in addition to invasion in SGC-7901 and MGC-803 GC gastric cancer cells through mTOR and S-phase kinase-associated protein 2 downregulation [83]. Additionally, deacetylisovaltratum, a traditional Chinese herbal medicine Patrinia heterophylla Bunge, inhibited the cell viability of AGS and HGC-27 cells and induced G2/M cell cycle arrest via disruption of mitochondrial membrane potential as well as induction of caspase-dependent apoptosis [55].

6. Anti-Angiogenesis Effects of Natural Bioactive Products in Gastric Cancer

Angiogenesis is the most common pathway for new vessel formation in cancer [93]. Anti-angiogenic agents were studied and developed for anti-cancer therapies because angiogenesis can cause tumor growth [94]. The vascular endothelial growth factor (VEGF) signaling pathway plays an essential role in regulating tumor angiogenesis, which can be used as a therapeutic target in numerous types of human gastric cancers [95]. Inhibition of VEGF leads to anti-angiogenesis in various animal and cell line models [96]. VEGFs have an important role in forming new blood vessels, including angiogenesis and vasculogenesis (Figure 3). A dietary flavonoid, luteolin, has been found to prevent angiogenesis in gastric cancer cells of MGC-803 and Hs-746T via the suppression of Notch1)/VEGF signaling [22]. Cyperenoic acid, a sesquiterpene isolated from Croton crassifolius, reduced vascular endothelial growth factor A (Vegfa or VEGF-A) genes by targeting the Vegfa-Kdr and Angpt-Tie signaling pathways [97]. Moreover, zerumbone, a bioactive component of ginger, showed anti-angiogenesis activity in AGS cells by reducing VEGF expression and inhibiting NF-κB [98]. Plumbagin inhibits tumor angiogenesis of gastric carcinoma via reduction of VEGF, VEGRF2, and MVD expression in gastric carcinoma in mice by the modulating nuclear factor-kappa B pathway [99]. Moreover, nitidine chloride, Zanthoxylum nitidum (Roxb) DC, was found to inhibit the signal transducer as well as activator of transcription 3 (STAT3) signaling in SGC-7901 and AGS human gastric cancer cell lines, which is related to tumor angiogenesis [100]. Additionally, treatment of nitidine chloride decreased the tumor volume through angiogenesis inhibition via reduction of STAT3 and VEGF levels in a xenograft mouse model induced by SGC-7901 cells [100]. Therefore, natural bioactive compound can effectively use certain VEGF subtypes, including VEGFA156, VEGFA121, VEGFR1, and VEGFR2, for the treatment of gastric cancer.
Table 3. Cell cycle arrest by bioactive food components in gastric cancer. (↑ increase, ↓ decrease).
Table 3. Cell cycle arrest by bioactive food components in gastric cancer. (↑ increase, ↓ decrease).
Phase of Cell Cycle ArrestClassificationCompoundSourceExperimental ModelDose; DurationMechanismReferences
G0/G1AlkaloidsBerberine(family: Ranunculaceae)
Coptidis japonica Makino Rhizoma
SGC-79015, 10, 20 µM; 24, 48 h [35]
G0/G1PhytosterolsGinsenoside-Rh2(family: Araliaceae) Panax ginseng C.A. Mey RadixSGC-79015, 10, 20 μg/mL; 24, 48 h↑Bax
↓Bcl-2
[38]
G0/G1TerpenoidsCrosolic acid(family: Actinidiaceae)
Actinidia valvata Dunn Radix
BGC-82320, 40, 80 μg/mL; 72 h↑Bax, smac, IκBα
↓Fas, Bcl-2, p65, p-IκBα, NF-κB
[54]
G1PolyphenolsRottlerin(family: Euphorbiaceae)
Mallotus philipensis Muell.
SGC-7901, MGC-8032, 4, 8 µM; 24 h↑LC3-II
↓mTOR, Skp2
[83]
G1/STerpenoidsToosendanin(family: Meliaceae)
Melia toosendan Sieb et Zucc Cortex et Fructus
(1) AGS
(2) HGC-27
(1) 0.5, 1, 2 μM; 48 h
(2) 0.5, 1, 2 μM; 36 h
↑c-caspase-3, -8, -9, c-PARP, Bax, p-p38
↓Bcl-2, Bcl-xL, Mcl-1, survivin, XIAP
[92]
SAlkaloidsCyclovirobuxine D(family: Buxaceae)
Buxus microphylla Richardii Radix
MGC-803, MKN-2830, 60, 120 μM/L; 48 h↑c-caspase-3, Bax
↓Bcl-2
[36]
SAlkaloidsGFG-3a(family: Meripilaceae)
Grifola frondose (Diks.) Gray Mycelia
SGC-7901100, 200 μg/mL; 24, 48 h↑RBBP4, caspase-3, -8, p53, Bax, Bad
↓RUVBL, NPM, Bcl-2, Bcl-xL, PI3K, Akt1
[64]
G2/MFlavonoidsTrifolirhizin(family: Fabaceae)
Sophora flavescens Aiton. Radix
MKN-4520, 30, 40 µg/mL; 48 h↑caspase-9, -3, c-PARP, p53, p38
↓EGFR, CDC2, cyclin B, ΔΨm
[37]
G2/MPhytosterolsTanshinone IIA(family: Lamiaceae) Salviae miltiorrhiza Bunge. RadixAGS2.0, 3.7, 5.5 µg/mL; 24, 48 h↑TNF-α, Fas, p-p38, p-JNK, p53, p21, caspase-8, -3
↓p-ERK, CDC2, cyclin A, cyclin B1
[39]
G2/MTerpenoidsDeacetylisovaltratum(family: Caprifoliaceae)
Patrinia heterophylla Bunge.
(1) AGS
(2) HGC-27
(1) 4, 8, 16 μM; 24 h
(2) 10, 20, 30 μM; 24 h
↑p21, caspase-3, c-PARP
↓p-STAT3, pro-caspase-9, ΔΨm
[55]

7. Anti-Metastasis Effects of Bioactive Compounds in Gastric Cancer

Metastasis is a major contributor of death in cancer patients, arising from a growing tumor from which cells escape to distant organs of body [101]. Targeting metastasis is an attractive strategy in cancer treatment. Anti-metastatic ability is highlighted in diverse natural bioactive products in vitro and in vivo models. which are described below. Sulforaphane, an organosulfur compound isolated from Brassica oleracea var. italica Plenk (Brassicaceae), exerted anti-metastatic ability on AGS and MKN-45 cells [102]. Isoliquiritigenin, a phenol found in Glycyrrhiza glabra (Fabaceae), inhibited tumor migration and metastasis on MKN-28 cells [103]. Dehydroeffusol, a benzenoid derived from Juncus effusus L. Radix et Medulla (Juncaceae), inhibited matrix metalloproteinase 2 (MMP-2) and VE-cadherin expression, resulting in reduction of the cell-to-cell adherent junction in AGS and SGC-7901 cells [104]. Baicalein, a well-known flavone found in the roots of Scutellaria baicalensis Georgi Radix (Lamiaceae), restrains motility, migration, and invasion of AGS gastric cancer cells via downregulation of N-cadherin, vimentin, ZEB1, ZEB2, and TGF-β/Smad4 [105]. Andrographolide, a labdane diterpenoid from the herb Andrographis paniculata Nees Herba (Acanthaceae), inhibits proliferation and metastasis of gastric cancer SGC-7901 via cell cycle arrest; upregulation of Bax, Bik, and TIMP-1/2; and downregulation of Bcl-2, CD147, MMP-2, and MMP-9 [106]. Blockages of tumor proliferation and metastasis of several bioactive compounds are presented in Table 4 and Figure 4. It has been found that evodiamine, isolated from Evoida rutaecarpa (Rutaceae), suppressed the epithelial–mesenchymal transition (EMT) of AGS and SGC-7901 gastric cancer cells via inhibition of the Wnt/β-catenin signaling pathway [107]. A triterpenoid found from Melia toosendan Sieb et Zucc (Meiliaceae), named toosendanin, has anti-metastatic capability on SGC-7901 cells through inhibition of the epithelial–mesenchymal transition of gastric cancer by upregulating miR-200a and e-cadherin and suppressing β-catenin [60]. Low-molecular-weight citrus pectin (LCP), derived from tangerines, grapefruits, lemons, and oranges, demonstrated anti-metastatic effects by treatment on AGS cells [108]. N-butylidenephthalide inhibited tumor metastasis in AGS, NCI-N87, and TSGH-9201 cells. The compound promoted e-cadherin expression while downregulating n-cadherin and vimentin slug. The activity of e-cadherin was repressed on the other hand, which inhibited EGFR kinase activity [57]. The mechanism leads to downstream regulation of multiple growth factor-related activities, which is associated with anti-metastatic activities of such natural bioactive products. In other aspects, the Bcl-2 family of proteins was also found to play a role in anti-metastatic effects of natural bioactive products [109]. Many other factors including PI3K, Akt, Rac1, and CDX1/2 play a role in anti-metastatic activity of natural bioactive compounds, some of which are also related to apoptosis of tumor cells. As it is unclear whether natural products exert anti-metastatic effects in a multi-target manner, further study is therefore required to distinguish the specific mechanism.
Table 4. Metastasis-inhibiting bioactive food components in vitro in gastric cancer. (↑ increase, ↓ decrease).
Table 4. Metastasis-inhibiting bioactive food components in vitro in gastric cancer. (↑ increase, ↓ decrease).
ClassificationCompoundSourceExperimental ModelDosesEfficacyMechanismsReference
AlkaloidsEvodiamine(family: Rutaceae) Tetradium ruticarpumAGS, SGC-79012 µM; 48 hInhibition of EMT↓β-catenin, cyclin D1, c-Myc [107]
Organosulfur compoundsSulforaphane(family: Brassicaceae) Brassica oleracea var. italica PlenkAGS, MKN-4531.25, 62.5, 125, 250 μg/mL; 48 hInhibition of metastasis↑CDX1, CDX2 [102]
↑miR-326, miR-9
PolyphenolsIsoliquiritigenin(family: Fabaceae) Glycyrrhiza glabra RadixMKN-2820 µM; 24, 48, 72 hInhibition of migration, invasion, Induction of apoptosis and autophagy↓Caspase-3, Bax, Bcl-2, PI3K, Akt, mTOR [103]
PolyphenolsDehydroeffusol(family: Juncaceae) Juncus effusus L. Radix et MedullaAGS, SGC-790112, 24, 48 µM; 24 hReduction of cell–cell adherent junction↓VE-cadherin, MMP-2 [104]
PolyphenolsPaeonol(family: Paeoniaceae) Paeonia suffruticosa Andr. Cortex,
(family: Asclepiadaceae) Cynanchum paniculatum K. Schum Radix
BGC-8230.1, 0.2, 0.4 mg/mL; 24, 48 hInhibition of proliferation, invasion, and migration, Induction of apoptosis↓MMP-2, MMP-9 [58]
PolyphenolsBaicalein(Lamiaceae) Scutellaria baicalensis Georgi RadixAGS25, 50 µM; 24 hInhibition of motility, migration, invasion↓N-cadherin, vimentin, ZEB1, ZEB2, TGF-β/Smad4 [105]
TerpenoidsAndrographolide(family: Acanthaceae) Andrographis paniculata Nees HerbaSGC-79015, 20, 40 µg/mL; 24, 48, 72 hInhibition of proliferation, invasion, metastasis↑Bax, Bik, TIMP-1/2,
↓Bcl-2, CD147, MMP-2, MMP-9, survivin
[106]
TerpenoidsToosendanin(family: Meliaceae) Melia toosendan Sieb et Zucc Cortex et FructusSGC-79010.5, 1 µM; 48 hInhibition of invasion, migration, EMTInduction of apoptosis and cell cycle arrest↑E-cadherin
↓β-catenin
[60]
↑miR-200a

8. Chemotherapy Resistance and Natural Bioactive Products in Gastric Cancer

Drug resistance is an important issue in cancer treatment and is known as a primary factor limiting cancer treatment [110]. Several studies have indicated that natural bioactive compounds could be used along with the primary drug to overcome drug resistance and reinforce its efficacy. In vitro drug resistance-overcoming bioactive food components in gastric cancer and their target signals are presented in Figure 5. Isorhamnetin, a flavonoid metabolite of quercetin commonly found in onions, minimized the apoptotic effects of capecitabine via inhibition of NF-κB and various NF-κB regulated gene products in tumor cells [111]. Liquiritin, isolated from Glycyrrhiza uralensis Fischer. Radix (Leguminosae/Fabaceae/Fabaceae), could circumvent the resistance of cisplatin-based chemotherapy via suppression of cell proliferation and induce apoptosis, autophagy, and G0/G1 phase cell cycle arrest against DDP-resistant gastric cancer cells [112]. Astragalus polysaccharide and apatinib co-treatment were reported to enhance apoptosis compared to apatinib monotherapy [113]. The efficacy of astragalus polysaccharide, an active component derived from Astragalus mambranaceus Bunge Radix (Leguminosae/Fabaceae/Fabaceae), arises mainly from its ability to inhibit autophagy of apatinib-resistant cells, which serves as a survival mechanism. Tanshinone IIA solution combined with doxorubicin showed anticancer effects against doxorubicin-resistant cell lines, including SNU-638, SNU-668, SNU-216, and SNU-620 [114]. Apoptosis was mainly induced by inhibition of multidrug resistance-associated protein 1 (MRP1). Although specific targets vary, most natural bioactive compounds aim to prevent drug resistance by downregulating Akt and NF-κB and following pathways (Figure 5). Mineral isorhamnetin from quercetin inhibited cell viability and prevented drug resistance by downregulating NF-κB. Liquirtin from the Glycyrrhiza genus promoted p53 and p21 and caspase cleavages while inhibiting cyclin activities. The compound’s anti-resistant ability may be focused on apoptotic effects. Other factors such as Bax/Bcl-2 in mitochondria, and ERK1/2, MMP2, and PARP are broadly affected by many natural bioactive compounds.

9. Limitation and Future Perspectives of Natural Bioactive Food Products in Gastric Cancer Treatments

Gastric cancer is known to account for the fifth highest incidence and the fourth highest mortality among all cancers worldwide [1]. Chemotherapy is one of the methods typically used in advanced gastric cancer treatment, but it exerts severe side effects that limit the efficacies and decrease quality of life. Development of therapeutic remedies with less adverse effects and lower chemo-resistance is required. Natural bioactive food products are emerging as alternative resources to combat gastric carcinoma. Therefore, several natural bioactive resources obtained from dietary fruits and vegetables were discussed. Curcumin and oligosaccharide isolated from tomato, sulforaphane derived from broccoli, and citrus pectin originated from tangerine, grapefruit, lemon, and orange are good examples. These medicinal resources are still being extensively used in traditional medicine. Many natural bioactive food products were shown to exhibit multiple effects. The variety is attributed to the structural diversity and multi-target characteristic of natural compounds [115]. Additionally, clinical trials were excluded to focus on laboratory experiments highlighting specific biological pathways. Several investigations were insufficient to elucidate anti-cancer mechanisms at molecular levels in gastric cancer. They were generally focused on the cytotoxicity of the chemicals or the reporting of newly discovered compounds, which makes incisive research burdensome. By and large, more than half of the studies only carried out experiments in vitro. More in vivo studies are recommended to bridge the advance to clinical trials and therapeutic use.
Natural bioactive food products are indeed effective in the single compound to single target mechanistic perspective; however, it is worth highlighting the complex interactions between many compounds. While the importance of studying the interactions between multi-compound natural bioactive food products and other drugs was previously highlighted in many literatures, it is also important to further investigate the interactions between different natural bioactive food products, including herbal medicines, in a biochemical manner [116]. A systemic approach with a focus on structural similarities of several phytochemical compounds and human metabolites is a potential way of clearly highlighting the efficacies of multi compound drugs. Despite the value of natural bioactive food products as medicinal agents, it is important that users as well prescribers be aware of the potentially cross-reactivity and toxicity of natural bioactive food products. Indeed, it has often been stated that natural bioactive products are toxins that are taken at lower therapeutic doses. To avoid this problem, it is required to modify the natural chemical. Therefore, it is important to recognize that unmodified natural bioactive food products may have suboptimal efficacy or absorption, distribution, metabolism, excretion, as well as toxicity (ADMET) properties. Thus, for development of natural bioactive food products that lead to successful drugs, chemical modifications or combinations with other compounds are highly required. Furthermore, clinical development requires a sustainable and suitably economically viable compound supply with sufficient quantities of natural bioactive food products.

10. Conclusions

In this review, we summed up several natural bioactive food products that have anti-cancer efficacy against gastric cancer. Several epidemiological investigations have been recommended, namely that the consumption of bioactive dietary food products such as spices, vegetables, fruits, roots, bulk, and leafs are inversely related to the risk and control of gastric cancer. In vitro and in vivo studies have been exposed, namely that dietary bioactive products mainly induced cell death by apoptosis and autophagy, cell cycle arrest, inhibition of angiogenesis and metastasis, and circumvention of chemo-resistance against stomach cancer cells through various molecular mechanisms. Several compounds showed multiple efficacies, attributed to structural complexity and multiple target pathways and proteins of bioactive dietary food products. Thus, natural substances implicate possibilities of being used in nutrition or medications, which may lead to novel discoveries in alternative medicine in cancer treatment. Additionally, attention should be paid to the bioavailability and safety of dietary food product consumption and a promising approach for the management and prevention of gastric cancer. This review provides data for future research and clinical trials to develop novel drugs from natural bioactive food products for gastric cancer treatment.

Author Contributions

Methodology, S.Y.K.; validation, S.Y.K., D.H., S.S., and J.P.; investigation, S.Y.K., D.H., S.S., M.K., and J.P.; writing—original draft preparation, S.Y.K., D.H., S.S., M.K., and J.P.; writing—review and editing, S.Y.K., D.H., and B.K.; visualization, S.S., J.P., and D.H., and S.-G.K.; supervision, B.K.; project administration, B.K.; figure drawing and modification, M.H.R.; editing and reviewing, M.A.R.; funding acquisition, B.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1I1A2066868), the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. 2020R1A5A2019413), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HF20C0116), and a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HF20C0038).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Hu, B.; El Hajj, N.; Sittler, S.; Lammert, N.; Barnes, R.; Meloni-Ehrig, A. Gastric cancer: Classification, histology and application of molecular pathology. J. Gastrointest. Oncol. 2012, 3, 251–261. [Google Scholar] [CrossRef] [PubMed]
  3. Crew, K.D.; Neugut, A.I. Epidemiology of gastric cancer. World J. Gastroenterol. 2006, 12, 354–362. [Google Scholar] [CrossRef] [PubMed]
  4. Rawla, P.; Barsouk, A. Epidemiology of gastric cancer: Global trends, risk factors and prevention. Gastroenterol. Rev. 2019, 14, 26–38. [Google Scholar] [CrossRef]
  5. A Ajani, J.; D’Amico, T.A.; Almhanna, K.; Bentrem, D.J.; Chao, J.; Das, P.; Denlinger, C.S.; Fanta, P.; Farjah, F.; Fuchs, C.S.; et al. Gastric Cancer, Version 3.2016, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2016, 14, 1286–1312. [Google Scholar] [CrossRef] [PubMed]
  6. Ronellenfitsch, U.; Schwarzbach, M.; Hofheinz, R.; Kienle, P.; Kieser, M.; E Slanger, T.; Jensen, K. GE adenocarcinoma meta-analysis group Perioperative chemo(radio)therapy versus primary surgery for resectable adenocarcinoma of the stomach, gastroesophageal junction, and lower esophagus. Cochrane Database Syst. Rev. 2013, 2013, 008107. [Google Scholar] [CrossRef] [Green Version]
  7. Diaz-Nieto, R.; Orti-Rodríguez, R.; Winslet, M. Post-surgical chemotherapy versus surgery alone for resectable gastric cancer. Cochrane Database Syst. Rev. 2013, 2013, CD008415. [Google Scholar] [CrossRef]
  8. Oba, K.; Paoletti, X.; Alberts, S.; Bang, Y.-J.; Benedetti, J.; Bleiberg, H.; Catalano, P.; Lordick, F.; Michiels, S.; Morita, S.; et al. Disease-Free Survival as a Surrogate for Overall Survival in Adjuvant Trials of Gastric Cancer: A Meta-Analysis. J. Natl. Cancer Inst. 2013, 105, 1600–1607. [Google Scholar] [CrossRef] [Green Version]
  9. Gibson, R.J.; Keefe, D.M.K. Cancer chemotherapy-induced diarrhoea and constipation: Mechanisms of damage and prevention strategies. Support. Care Cancer 2006, 14, 890–900. [Google Scholar] [CrossRef] [PubMed]
  10. Staff, N.P.; Grisold, A.; Grisold, W.; Windebank, A.J. Chemotherapy-induced peripheral neuropathy: A current review. Ann. Neurol. 2017, 81, 772–781. [Google Scholar] [CrossRef] [PubMed]
  11. Yang, W.; Ma, J.; Zhou, W.; Cao, B.; Zhou, X.; Yang, Z.; Zhang, H.; Zhao, Q.; Fan, D.; Hong, L. Molecular mechanisms and theranostic potential of miRNAs in drug resistance of gastric cancer. Expert Opin. Ther. Targets 2017, 21, 1063–1075. [Google Scholar] [CrossRef]
  12. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121–126. [Google Scholar] [CrossRef] [PubMed]
  13. Lu, B.; Li, M. Helicobacter pylori eradication for preventing gastric cancer. World J. Gastroenterol. 2014, 20, 5660–5665. [Google Scholar] [CrossRef]
  14. Sokolova, O.; Naumann, M. NF-kappaB Signaling in Gastric Cancer. Toxins 2017, 9, 119. [Google Scholar] [CrossRef] [PubMed]
  15. Lee, H.; Jeong, A.J.; Ye, S.-K. Highlighted STAT3 as a potential drug target for cancer therapy. BMB Rep. 2019, 52, 415–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Naylor, M.S.; Stamp, G.W.; Foulkes, W.; Eccles, D.; Balkwill, F. Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. J. Clin. Investig. 1993, 91, 2194–2206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Wang, X.; Lin, Y. Tumor necrosis factor and cancer, buddies or foes? Acta Pharmacol. Sin. 2008, 29, 1275–1288. [Google Scholar] [CrossRef] [Green Version]
  18. Rabelo, A.C.S.; Camini, F.C.; Bittencourt, M.M.; Lacerda, K.; De Lima, W.G.; Costa, D.C. Baccharis trimera (carqueja) promotes gastroprotection on ethanol-induced acute gastric ulcer. Adv. Tradit. Med. 2020, 20, 563–570. [Google Scholar] [CrossRef]
  19. Bastos, J.; Lunet, N.; Peleteiro, B.; Lopes, C.; Barros, H. Dietary patterns and gastric cancer in a Portuguese urban population. Int. J. Cancer 2010, 127, 433–441. [Google Scholar] [CrossRef]
  20. Nagata, C.; Takatsuka, N.; Kawakami, N.; Shimizu, H. A prospective cohort study of soy product intake and stomach cancer death. Br. J. Cancer 2002, 87, 31–36. [Google Scholar] [CrossRef] [Green Version]
  21. Steevens, J.; Schouten, L.J.; Goldbohm, R.A.; Brandt, P.V.D. Vegetables and fruits consumption and risk of esophageal and gastric cancer subtypes in the Netherlands Cohort Study. Int. J. Cancer 2011, 129, 2681–2693. [Google Scholar] [CrossRef] [PubMed]
  22. Mao, Q.-Q.; Xu, X.-Y.; Shang, A.; Gan, R.-Y.; Wu, D.-T.; Atanasov, A.G.; Li, H.-B. Phytochemicals for the Prevention and Treatment of Gastric Cancer: Effects and Mechanisms. Int. J. Mol. Sci. 2020, 21, 570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Xu, J.; Long, Y.; Ni, L.; Yuan, X.; Yu, N.; Wu, R.; Tao, J.; Zhang, Y. Anticancer effect of berberine based on experimental animal models of various cancers: A systematic review and meta-analysis. BMC Cancer 2019, 19, 1–20. [Google Scholar] [CrossRef] [Green Version]
  24. Hassanalilou, T.; Ghavamzadeh, S.; Khalili, L. Curcumin and Gastric Cancer: A Review on Mechanisms of Action. J. Gastrointest. Cancer 2019, 50, 185–192. [Google Scholar] [CrossRef] [PubMed]
  25. Dutta, S.; Mahalanobish, S.; Saha, S.; Ghosh, S.; Sil, P.C. Natural products: An upcoming therapeutic approach to cancer. Food Chem. Toxicol. 2019, 128, 240–255. [Google Scholar] [CrossRef]
  26. Kim, H.-J.; Um, J.-Y.; Kim, Y.-K. Glutathione S-transferase gene polymorphism in Korean subjects with gastric and colorectal cancer. Orient. Pharm. Exp. Med. 2012, 12, 307–312. [Google Scholar] [CrossRef]
  27. Mann, J. Natural products in cancer chemotherapy: Past, present and future. Nat. Rev. Cancer 2002, 2, 143–148. [Google Scholar] [CrossRef]
  28. Wang, L.; Dong, C.; Li, X.; Han, W.; Su, X. Anticancer potential of bioactive peptides from animal sources (Review). Oncol. Rep. 2017, 38, 637–651. [Google Scholar] [CrossRef] [Green Version]
  29. Gras, M.; Vallard, A.; Brosse, C.; Beneton, A.; Sotton, S.; Guyotat, D.; Fournel, P.; Daguenet, E.; Magné, N.; Morisson, S. Use of Complementary and Alternative Medicines among Cancer Patients: A Single-Center Study. Oncology 2019, 97, 18–25. [Google Scholar] [CrossRef]
  30. Li, X.; Yang, G.; Li, X.; Zhang, Y.; Yang, J.; Chang, J.; Sun, X.; Zhou, X.; Guo, Y.; Xu, Y.; et al. Traditional Chinese medicine in cancer care: A review of controlled clinical studies published in chinese. PLoS ONE 2013, 8, e60338. [Google Scholar]
  31. Wode, K.; Henriksson, R.; Sharp, L.; Stoltenberg, A.; Nordberg, J.H. Cancer patients’ use of complementary and alternative medicine in Sweden: A cross-sectional study. BMC Complement. Altern. Med. 2019, 19, 1–11. [Google Scholar] [CrossRef] [Green Version]
  32. Kristoffersen, A.E.; Stub, T.; Broderstad, A.R.; Hansen, A.H. Use of traditional and complementary medicine among Norwegian cancer patients in the seventh survey of the Tromsø study. BMC Complement. Altern. Med. 2019, 19, 1–13. [Google Scholar] [CrossRef] [PubMed]
  33. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  34. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta Bioenerg. 2016, 1863, 2977–2992. [Google Scholar] [CrossRef] [PubMed]
  35. Yang, Y.; Zhang, N.; Li, K.; Chen, J.; Qiu, L.; Zhang, J. Integration of microRNA-mRNA profiles and pathway analysis of plant isoquinoline alkaloid berberine in SGC-7901 gastric cancers cells. Drug Des. Devel. Ther. 2018, 12, 393–408. [Google Scholar] [CrossRef] [Green Version]
  36. Wu, J.; Tan, Z.; Chen, J.; Dong, C. Cyclovirobuxine D Inhibits Cell Proliferation and Induces Mitochondria-Mediated Apoptosis in Human Gastric Cancer Cells. Molecules 2015, 20, 20659–20668. [Google Scholar] [CrossRef] [Green Version]
  37. Lu, X.; Ma, J.; Qiu, H.; Yang, L.; Cao, L.; Shen, J. Anti-proliferation effects of trifolirhizin on MKN45 cells and possible mechanism. Oncol. Rep. 2016, 36, 2785–2792. [Google Scholar] [CrossRef] [Green Version]
  38. Qian, J.; Li, J.; Jia, J.-G.; Jin, X.; Yu, D.-J.; Guo, C.-X.; Xie, B.; Qian, L.-Y. Ginsenoside-Rh2 Inhibits Proliferation and Induces Apoptosis of Human Gastric Cancer SGC-7901 Side Population Cells. Asian Pac. J. Cancer Prev. 2016, 17, 1817–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Su, C.-C. Tanshinone IIA inhibits gastric carcinoma AGS cells through increasing p-p38, p-JNK and p53 but reducing p-ERK, CDC2 and cyclin B1 expression. Anticancer. Res. 2014, 34, 7097–7110. [Google Scholar] [PubMed]
  40. Zhu, H.; Zheng, Z.; Zhang, J.; Liu, X.; Liu, Y.; Yang, W.; Liu, Y.; Zhang, T.; Zhao, Y.; Liu, Y.; et al. Anticancer effect of 2,7-dihydroxy-3-methylanthraquinone on human gastric cancer SGC-7901 cells in vitro and in vivo. Pharm. Biol. 2016, 54, 285–292. [Google Scholar] [CrossRef]
  41. Luo, Y.; Yu, H.; Yang, Y.; Tian, W.; Dong, K.; Shan, J.; Ma, X. A flavonoid compound from Chrysosplenium nudicaule inhibits growth and induces apoptosis of the human stomach cancer cell line SGC-7901. Pharm. Biol. 2016, 54, 1133–1139. [Google Scholar] [CrossRef] [Green Version]
  42. Sun, Q.; Zhang, W.; Guo, Y.; Li, Z.; Chen, X.; Wang, Y.; Du, Y.; Zang, W.; Zhao, G. Curcumin inhibits cell growth and induces cell apoptosis through upregulation of miR-33b in gastric cancer. Tumor Biol. 2016, 37, 13177–13184. [Google Scholar] [CrossRef]
  43. Pan, H.; Wang, B.-H.; Lv, W.; Jiang, Y.; He, L. Esculetin induces apoptosis in human gastric cancer cells through a cyclophilin D-mediated mitochondrial permeability transition pore associated with ROS. Chem. Interactions 2015, 242, 51–60. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, L.; Si, N.; Ma, Y.; Ge, D.; Yu, X.; Fan, A.; Wang, X.; Hu, J.; Wei, P.; Chen, J.; et al. Hydroxysafflor-Yellow A Induces Human Gastric Carcinoma BGC-823 Cell Apoptosis by Activating Peroxisome Proliferator-Activated Receptor Gamma (PPARgamma). Med. Sci. Monit. 2018, 24, 803–811. [Google Scholar] [CrossRef] [Green Version]
  45. Zhou, W.; Cao, A.; Wang, L.; Wu, D. Kurarinone Synergizes TRAIL-Induced Apoptosis in Gastric Cancer Cells. Cell Biophys. 2015, 72, 241–249. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, J.; Zhang, X.; Wang, Y.; Sun, Q.; Chen, M.; Liu, S.; Zou, X. Licochalcone A suppresses hexokinase 2-mediated tumor glycolysis in gastric cancer via downregulation of the Akt signaling pathway. Oncol. Rep. 2017, 39, 1181–1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Jung, E.B.; Trinh, T.A.; Lee, T.K.; Yamabe, N.; Kang, K.S.; Song, J.H.; Choi, S.; Lee, S.; Jang, T.S.; Kim, K.H.; et al. Curcuzedoalide contributes to the cytotoxicity of Curcuma zedoaria rhizomes against human gastric cancer AGS cells through induction of apoptosis. J. Ethnopharmacol. 2018, 213, 48–55. [Google Scholar] [CrossRef] [PubMed]
  48. Kang, S.-H.; Kim, Y.-S.; Kim, E.-K.; Hwang, J.-W.; Jeong, J.-H.; Dong, X.; Lee, J.-W.; Moon, S.-H.; Jeon, B.-T.; Park, P.-J. Anticancer Effect of Thymol on AGS Human Gastric Carcinoma Cells. J. Microbiol. Biotechnol. 2016, 26, 28–37. [Google Scholar] [CrossRef]
  49. Zhang, W.; Zhang, Q.; Jiang, Y.; Li, F.; Xin, H. Effects of ophiopogonin B on the proliferation and apoptosis of SGC-7901 human gastric cancer cells. Mol. Med. Rep. 2016, 13, 4981–4986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Xu, M.; Gu, W.; Shen, Z.; Wang, F. Anticancer Activity of Phloretin Against Human Gastric Cancer Cell Lines Involves Apoptosis, Cell Cycle Arrest, and Inhibition of Cell Invasion and JNK Signalling Pathway. Med. Sci. Monit. 2018, 24, 6551–6558. [Google Scholar] [CrossRef]
  51. Asl, E.A.; Mehrabadi, J.F.; Afshar, D.; Noorbazargan, H.; Tahmasebi, H.; Rahimi, A. Apoptotic Effects of Linum album Extracts on AGS Human Gastric Adenocarcinoma Cells and ZNF703 Oncogene Expression. Asian Pac. J. Cancer Prev. 2018, 19, 2911–2916. [Google Scholar] [CrossRef]
  52. Wu, Z.; Li, Y. Grifolin exhibits anti-cancer activity by inhibiting the development and invasion of gastric tumor cells. Oncotarget 2017, 8, 21454–21460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Tsai, T.C.; Lai, K.-H.; Su, J.-H.; Wu, Y.-J.; Sheu, J.-H. 7-Acetylsinumaximol B Induces Apoptosis and Autophagy in Human Gastric Carcinoma Cells through Mitochondria Dysfunction and Activation of the PERK/eIF2alpha/ATF4/CHOP Signaling Pathway. Mar. Drugs 2018, 16, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Cheng, Q.-L.; Li, H.-L.; Li, Y.-C.; Liu, Z.-W.; Guo, X.-H.; Cheng, Y.-J. CRA (Crosolic Acid) isolated from Actinidia valvata Dunn.Radix induces apoptosis of human gastric cancer cell line BGC823 in vitro via down-regulation of the NF-kappaB pathway. Food Chem. Toxicol. 2017, 105, 475–485. [Google Scholar] [CrossRef]
  55. Zhang, D.; Zhang, B.; Zhou, L.-X.; Zhao, J.; Yan, Y.-Y.; Li, Y.-L.; Zeng, J.-M.; Wang, L.-L.; Yang, B.; Lin, N.-M. Deacetylisovaltratum disrupts microtubule dynamics and causes G2/M-phase arrest in human gastric cancer cells in vitro. Acta Pharmacol. Sin. 2016, 37, 1597–1605. [Google Scholar] [CrossRef]
  56. Li, P.; Zhou, X.; Sun, W.; Sheng, W.; Tu, Y.; Yu, Y.; Dong, J.; Ye, B.; Zheng, Z.; Lu, M. Elemene Induces Apoptosis of Human Gastric Cancer Cell Line BGC-823 via Extracellular Signal-Regulated Kinase (ERK) 1/2 Signaling Pathway. Med. Sci. Monit. 2017, 23, 809–817. [Google Scholar] [CrossRef] [PubMed]
  57. Liao, K.-F.; Chiu, T.-L.; Huang, S.-Y.; Hsieh, T.-F.; Chang, S.-F.; Ruan, J.-W.; Chen, S.-P.; Pang, C.-Y.; Chiu, S.-C. Anti-Cancer Effects of Radix Angelica Sinensis (Danggui) and N-Butylidenephthalide on Gastric Cancer: Implications for REDD1 Activation and mTOR Inhibition. Cell. Physiol. Biochem. 2018, 48, 2231–2246. [Google Scholar] [CrossRef] [PubMed]
  58. Lyu, Z.-K.; Li, C.-L.; Jin, Y.; Liu, Y.-Z.; Zhang, X.; Zhang, F.; Ning, L.-N.; Liang, E.-S.; Ma, M.; Gao, W.; et al. Paeonol exerts potential activities to inhibit the growth, migration and invasion of human gastric cancer BGC823 cells via downregulating MMP-2 and MMP-9. Mol. Med. Rep. 2017, 16, 7513–7519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Yu, F.; Li, K.; Chen, S.; Liu, Y.; Li, Y. Pseudolaric Acid B Circumvents Multidrug Resistance Phenotype in Human Gastric Cancer SGC7901/ADR Cells by Downregulating Cox-2 and P-gp Expression. Cell Biophys. 2014, 71, 119–126. [Google Scholar] [CrossRef] [PubMed]
  60. Wang, G.; Huang, Y.-X.; Zhang, R.; Hou, L.-D.; Liu, H.; Chen, X.-Y.; Zhu, J.-S.; Zhang, J. Toosendanin suppresses oncogenic phenotypes of human gastric carcinoma SGC7901 cells partly via miR200amediated downregulation of beta-catenin pathway. Int. J. Oncol. 2017, 51, 1563–1573. [Google Scholar] [CrossRef] [Green Version]
  61. Kapoor, S.; Dharmesh, S.M. Pectic Oligosaccharide from tomato exhibiting anticancer potential on a gastric cancer cell line: Structure-function relationship. Carbohydr. Polym. 2017, 160, 52–61. [Google Scholar] [CrossRef]
  62. Zhang, H.; Wu, D.; Du, J.; Zhang, Y.; Su, Y. Anti-tumor effects of phenolic alkaloids of menispermum dauricum on gastric cancer in vivo and in vitro. J. Cancer Res. Ther. 2018, 14, 505. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, X.-P.; Wang, Q.-X.; Lin, H.-P.; Chang, N. Anti-tumor bioactivities of curcumin on mice loaded with gastric carcinoma. Food Funct. 2017, 8, 3319–3326. [Google Scholar] [CrossRef] [PubMed]
  64. Cui, F.; Zan, X.; Li, Y.; Sun, W.; Yang, Y.; Ping, L. Grifola frondosaGlycoprotein GFG-3a Arrests S phase, Alters Proteome, and Induces Apoptosis in Human Gastric Cancer Cells. Nutr. Cancer 2016, 68, 267–279. [Google Scholar] [CrossRef] [PubMed]
  65. Kong, G.-M.; Tao, W.-H.; Diao, Y.-L.; Fang, P.-H.; Wang, J.-J.; Bo, P.; Qian, F. Melittin induces human gastric cancer cell apoptosis via activation of mitochondrial pathway. World J. Gastroenterol. 2016, 22, 3186–3195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zhang, X.-Z.; Wang, L.; Liu, D.-W.; Tang, G.-Y.; Zhang, H.-Y. Synergistic Inhibitory Effect of Berberine and d-Limonene on Human Gastric Carcinoma Cell Line MGC803. J. Med. Food 2014, 17, 955–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Li, L.; Zhao, L.-M.; Dai, S.-L.; Cui, W.-X.; Lv, H.-L.; Chen, L.; Shan, B.-E. Periplocin Extracted from Cortex Periplocae Induced Apoptosis of Gastric Cancer Cells via the ERK1/2-EGR1 Pathway. Cell Physiol. Biochem. 2016, 38, 1939–1951. [Google Scholar] [CrossRef]
  68. Mansingh, D.P.; Oj, S.; Sali, V.K.; Vasanthi, H.R. [6]-Gingerol-induced cell cycle arrest, reactive oxygen species generation, and disruption of mitochondrial membrane potential are associated with apoptosis in human gastric cancer (AGS) cells. J. Biochem. Mol. Toxicol. 2018, 32, e22206. [Google Scholar] [CrossRef] [PubMed]
  69. Feng, Y.; He, D.; Yao, Z.; Klionsky, D.J. The machinery of macroautophagy. Cell Res. 2014, 24, 24–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Rahman, M.A.; Rhim, H. Therapeutic implication of autophagy in neurodegenerative diseases. BMB Rep. 2017, 50, 345–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Kang, R.; Zeh, H.J.; Lotze, M.T.; Tang, D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 2011, 18, 571–580. [Google Scholar] [CrossRef] [PubMed]
  72. Rahman, M.A.; Hannan, M.A.; Dash, R.; Rahman, M.H.; Islam, R.; Uddin, M.J.; Sohag, A.A.M.; Rahman, M.H.; Rhim, H. Phytochemicals as a Complement to Cancer Chemotherapy: Pharmacological Modulation of the Autophagy-Apoptosis Pathway. Front. Pharmacol. 2021, 12, 639628. [Google Scholar] [CrossRef]
  73. Rahman, M.A.; Rahman, M.S.; Rahman, M.H.; Rasheduzzaman, M.; Mamun-Or-Rashid, A.; Uddin, M.J.; Rahman, M.R.; Hwang, H.; Pang, M.G.; Rhim, H. Modulatory Effects of Autophagy on APP Processing as a Potential Treatment Target for Alzheimer’s Disease. Biomedicines 2021, 9, 5. [Google Scholar] [CrossRef]
  74. Onorati, A.V.; Dyczynski, M.; Ojha, R.; Amaravadi, R.K. Targeting autophagy in cancer. Cancer 2018, 124, 3307–3318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Mandhair, H.K.; Arambasic, M.; Novak, U.; Radpour, R. Molecular modulation of autophagy: New venture to target resistant cancer stem cells. World J. Stem Cells 2020, 12, 303–322. [Google Scholar] [CrossRef] [PubMed]
  76. Uddin, M.S.; Rahman, M.A.; Kabir, M.T.; Behl, T.; Mathew, B.; Perveen, A.; Barreto, G.E.; Bin-Jumah, M.N.; Abdel-Daim, M.M.; Ashraf, G.M. Multifarious roles of mTOR signaling in cognitive aging and cerebrovascular dysfunction of Alzheimer’s disease. Iubmb. Life 2020, 72, 1843–1855. [Google Scholar] [CrossRef]
  77. Rahman, M.A.; Cho, Y.; Nam, G.; Rhim, H. Antioxidant Compound, Oxyresveratrol, Inhibits APP Production through the AMPK/ULK1/mTOR-Mediated Autophagy Pathway in Mouse Cortical Astrocytes. Antioxidants 2021, 10, 408. [Google Scholar] [CrossRef]
  78. Tanida, I.; Ueno, T.; Kominami, E. LC3 conjugation system in mammalian autophagy. Int. J. Biochem. Cell Biol. 2004, 36, 2503–2518. [Google Scholar] [CrossRef]
  79. Dooley, H.C.; Razi, M.; Polson, H.E.J.; Girardin, S.E.; Wilson, M.I.; Tooze, S.A. WIPI2 Links LC3 Conjugation with PI3P, Autophagosome Formation, and Pathogen Clearance by Recruiting Atg12–5-16L1. Mol. Cell 2014, 55, 238–252. [Google Scholar] [CrossRef] [Green Version]
  80. Rahman, M.A.; Rahman, M.H.; Hossain, M.S.; Biswas, P.; Islam, R.; Uddin, M.J.; Rahman, M.H.; Rhim, H. Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention. Biomedicines 2020, 8, 517. [Google Scholar] [CrossRef]
  81. Rahman, M.A.; Cho, Y.; Hwang, H.; Rhim, H. Pharmacological Inhibition of O-GlcNAc Transferase Promotes mTOR-Dependent Autophagy in Rat Cortical Neurons. Brain Sci. 2020, 10, 958. [Google Scholar] [CrossRef] [PubMed]
  82. Pang, X.; Zhang, X.; Jiang, Y.; Su, Q.; Li, Q.; Li, Z. Autophagy: Mechanisms and Therapeutic Potential of Flavonoids in Cancer. Biomolecules 2021, 11, 135. [Google Scholar] [CrossRef]
  83. Song, J.; Zhou, Y.; Gong, Y.; Liu, H.; Tang, L. Rottlerin promotes autophagy and apoptosis in gastric cancer cell lines. Mol. Med. Rep. 2018, 18, 2905–2913. [Google Scholar] [CrossRef] [PubMed]
  84. Batool, S.; Joseph, T.P.; Hussain, M.; Vuai, M.S.; Khinsar, K.H.; Din, S.R.U.; Padhiar, A.A.; Zhong, M.; Ning, A.; Zhang, W.; et al. LP1 from Lentinula edodes C91-3 Induces Autophagy, Apoptosis and Reduces Metastasis in Human Gastric Cancer Cell Line SGC-7901. Int. J. Mol. Sci. 2018, 19, 2986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kwon, Y.H.; Bishayee, K.; Rahman, A.; Hong, J.S.; Lim, S.-S.; Huh, S.-O. Morus alba Accumulates Reactive Oxygen Species to Initiate Apoptosis via FOXO-Caspase 3-Dependent Pathway in Neuroblastoma Cells. Mol. Cells 2015, 38, 630–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Rahman, M.A.; Bishayee, K.; Sadra, A.; Huh, S.-O. Oxyresveratrol activates parallel apoptotic and autophagic cell death pathways in neuroblastoma cells. Biochim. Biophys Acta Gen. Subj. 2017, 1861, 23–36. [Google Scholar] [CrossRef] [PubMed]
  87. Kim, T.W.; Lee, S.Y.; Kim, M.; Cheon, C.; Ko, S.-G. Kaempferol induces autophagic cell death via IRE1-JNK-CHOP pathway and inhibition of G9a in gastric cancer cells. Cell Death Dis. 2018, 9, 875. [Google Scholar] [CrossRef] [PubMed]
  88. Lee, H.J.; Saralamma, V.V.G.; Kim, S.M.; Ha, S.E.; Raha, S.; Lee, W.S.; Kim, E.H.; Lee, S.J.; Heo, J.D.; Kim, G.S. Pectolinarigenin Induced Cell Cycle Arrest, Autophagy, and Apoptosis in Gastric Cancer Cell via PI3K/AKT/mTOR Signaling Pathway. Nutrients 2018, 10, 1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Zhang, Y.; Liu, S.; Feng, Q.; Huang, X.; Wang, X.; Peng, Y.; Zhao, Z.; Liu, Z. Perilaldehyde activates AMP-activated protein kinase to suppress the growth of gastric cancer via induction of autophagy. J. Cell. Biochem. 2019, 120, 1716–1725. [Google Scholar] [CrossRef]
  90. Wang, K.; Liu, R.; Li, J.; Mao, J.; Lei, Y.; Wu, J.; Zeng, J.; Zhang, T.; Wu, H.; Chen, L.; et al. Quercetin induces protective autophagy in gastric cancer cells: Involvement of Akt-mTOR- and hypoxia-induced factor 1alpha-mediated signaling. Autophagy 2011, 7, 966–978. [Google Scholar] [CrossRef] [Green Version]
  91. Mrakovcic, M.; Fröhlich, L. p53-Mediated Molecular Control of Autophagy in Tumor Cells. Biomolecules 2018, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  92. Zhou, Q.; Wu, X.; Wen, C.; Wang, H.; Wang, H.; Liu, H.; Peng, J. Toosendanin induces caspase-dependent apoptosis through the p38 MAPK pathway in human gastric cancer cells. Biochem. Biophys. Res. Commun. 2018, 505, 261–266. [Google Scholar] [CrossRef]
  93. Cao, Y.; Arbiser, J.; D’Amato, R.J.; D’Amore, P.A.; Ingber, D.E.; Kerbel, R.; Klagsbrun, M.; Lim, S.; Moses, M.A.; Zetter, B.; et al. Forty-Year Journey of Angiogenesis Translational Research. Sci. Transl. Med. 2011, 3, 114rv3. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, Z.; Dabrosin, C.; Yin, X.; Fuster, M.M.; Arreola, A.; Rathmell, W.K.; Generali, D.; Nagaraju, G.P.; El-Rayes, B.; Ribatti, D.; et al. Broad targeting of angiogenesis for cancer prevention and therapy. Semin. Cancer Biol. 2015, 35, S224–S243. [Google Scholar] [CrossRef] [PubMed]
  95. Yang, J.; Wang, Q.; Qiao, C.; Lin, Z.; Li, X.; Huang, Y.; Zhou, T.; Li, Y.; Shen, B.; Lv, M.; et al. Potent anti-angiogenesis and anti-tumor activity of a novel human anti-VEGF antibody, MIL60. Cell. Mol. Immunol. 2014, 11, 285–293. [Google Scholar] [CrossRef] [Green Version]
  96. Eklund, L.; Bry, M.; Alitalo, K. Mouse models for studying angiogenesis and lymphangiogenesis in cancer. Mol. Oncol. 2013, 7, 259–282. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, W.; Wang, J.; Liang, Y.; Ge, W.; Wang, G.; Li, Y.; Chung, H.Y. Potent anti-angiogenic component in Croton crassifolius and its mechanism of action. J. Ethnopharmacol. 2015, 175, 185–191. [Google Scholar] [CrossRef] [PubMed]
  98. Tsuboi, K.; Matsuo, Y.; Shamoto, T.; Shibata, T.; Koide, S.; Morimoto, M.; Guha, S.; Sung, B.; Aggarwal, B.B.; Takahashi, H.; et al. Zerumbone inhibits tumor angiogenesis via NF-kappaB in gastric cancer. Oncol. Rep. 2014, 31, 57–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Manu, K.A.; Shanmugam, M.K.; Rajendran, P.; Li, F.; Ramachandran, L.; Hay, H.S.; Kannaiyan, R.; Swamy, S.N.; Vali, S.; Kapoor, S.; et al. Plumbagin inhibits invasion and migration of breast and gastric cancer cells by downregulating the expression of chemokine receptor CXCR4. Mol. Cancer 2011, 10, 107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Chen, J.; Wang, J.; Lin, L.; He, L.; Wu, Y.; Zhang, L.; Yi, Z.; Chen, Y.; Pang, X.; Liu, M. Inhibition of STAT3 Signaling Pathway by Nitidine Chloride Suppressed the Angiogenesis and Growth of Human Gastric Cancer. Mol. Cancer Ther. 2012, 11, 277–287. [Google Scholar] [CrossRef] [Green Version]
  101. Suhail, Y.; Cain, M.P.; Vanaja, K.; Kurywchak, P.A.; Levchenko, A.; Kalluri, R. Kshitiz Systems Biology of Cancer Metastasis. Cell Syst. 2019, 9, 109–127. [Google Scholar] [CrossRef] [Green Version]
  102. Kiani, S.; Akhavan-Niaki, H.; Fattahi, S.; Kavoosian, S.; Jelodar, N.B.; Bagheri, N.; Zarrini, H.N. Purified sulforaphane from broccoli (Brassica oleracea var. italica) leads to alterations of CDX1 and CDX2 expression and changes in miR-9 and miR-326 levels in human gastric cancer cells. Gene 2018, 678, 115–123. [Google Scholar] [CrossRef]
  103. Zhang, X.; Wang, S.; Sun, W.; Wei, C. Isoliquiritigenin inhibits proliferation and metastasis of MKN28 gastric cancer cells by suppressing the PI3K/AKT/mTOR signaling pathway. Mol. Med. Rep. 2018, 18, 3429–3436. [Google Scholar] [CrossRef] [Green Version]
  104. Liu, W.; Meng, M.; Zhang, B.; Du, L.; Pan, Y.; Yang, P.; Gu, Z.; Zhou, Q.; Cao, Z. Dehydroeffusol effectively inhibits human gastric cancer cell-mediated vasculogenic mimicry with low toxicity. Toxicol. Appl. Pharmacol. 2015, 287, 98–110. [Google Scholar] [CrossRef]
  105. Chen, F.; Zhuang, M.; Peng, J.; Wang, X.; Huang, T.; Li, S.; Lin, M.; Lin, H.; Xu, Y.; Li, J.; et al. Baicalein inhibits migration and invasion of gastric cancer cells through suppression of the TGF-beta signaling pathway. Mol. Med. Rep. 2014, 10, 1999–2003. [Google Scholar] [CrossRef] [Green Version]
  106. Dai, L.; Wang, G.; Pan, W. Andrographolide Inhibits Proliferation and Metastasis of SGC7901 Gastric Cancer Cells. BioMed Res. Int. 2017, 2017, 1–10. [Google Scholar] [CrossRef] [PubMed]
  107. Wen, Z.; Feng, S.; Wei, L.; Wang, Z.; Hong, D.; Wang, Q. Evodiamine, a novel inhibitor of the Wnt pathway, inhibits the self-renewal of gastric cancer stem cells. Int. J. Mol. Med. 2015, 36, 1657–1663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Wang, S.; Li, P.; Lu, S.-M.; Ling, Z.-Q. Chemoprevention of Low-Molecular-Weight Citrus Pectin (LCP) in Gastrointestinal Cancer Cells. Int. J. Biol. Sci. 2016, 12, 746–756. [Google Scholar] [CrossRef] [PubMed]
  109. Kirkin, V.; Joos, S.; Zornig, M. The role of Bcl-2 family members in tumorigenesis. Biochim. Biophys. Acta 2004, 1644, 229–249. [Google Scholar] [CrossRef]
  110. Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Manu, K.A.; Shanmugam, M.K.; Ramachandran, L.; Li, F.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Arfuso, F.; Kumar, A.P.; et al. Isorhamnetin augments the anti-tumor effect of capecitabine through the negative regulation of NF-kappaB signaling cascade in gastric cancer. Cancer Lett. 2015, 363, 28–36. [Google Scholar] [CrossRef]
  112. Wei, F.; Jiang, X.; Gao, H.-Y.; Gao, S.-H. Liquiritin induces apoptosis and autophagy in cisplatin (DDP)-resistant gastric cancer cells in vitro and xenograft nude mice in vivo. Int. J. Oncol. 2017, 51, 1383–1394. [Google Scholar] [CrossRef] [Green Version]
  113. Wu, J.; Yu, J.; Wang, J.; Zhang, C.; Shang, K.; Yao, X.; Cao, B. Astragalus polysaccharide enhanced antitumor effects of Apatinib in gastric cancer AGS cells by inhibiting AKT signalling pathway. Biomed. Pharmacother. 2018, 100, 176–183. [Google Scholar] [CrossRef] [PubMed]
  114. Xu, Z.; Chen, L.; Xiao, Z.; Zhu, Y.; Jiang, H.; Jin, Y.; Gu, C.; Wu, Y.; Wang, L.; Zhang, W.; et al. Potentiation of the anticancer effect of doxorubicinin drug-resistant gastric cancer cells by tanshinone IIA. Phytomedicine 2018, 51, 58–67. [Google Scholar] [CrossRef] [PubMed]
  115. Guerra, A.R.; Duarte, M.F.; Duarte, I.F. Targeting Tumor Metabolism with Plant-Derived Natural Products: Emerging Trends in Cancer Therapy. J. Agric. Food Chem. 2018, 66, 10663–10685. [Google Scholar] [CrossRef] [PubMed]
  116. Roe, A.L.; Paine, M.F.; Gurley, B.J.; Brouwer, K.R.; Jordan, S.; Griffiths, J.C. Assessing Natural Product–Drug Interactions: An End-to-End Safety Framework. Regul. Toxicol. Pharmacol. 2016, 76, 1–6. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of natural bioactive food product-mediated apoptosis signaling pathways. FADD, Fas-associated proteins with death domain; TRAILR, TNF-related apoptosis-including ligand receptor; FASR, Fas receptor; tBid, truncated Bid; PARP, poly ADP-ribose polymerase; APAF1, apoptotic protase activating factor 1; MOMP, mitochondrial outer membrane permeabilization; PIP2, phosphatidylinositol-3,4-bisphosphate; PIP3, phosphatidylinositol-3,4,5-triphosphate; PI3K, phosphoinositide 3-kinase.
Figure 1. Schematic diagram of natural bioactive food product-mediated apoptosis signaling pathways. FADD, Fas-associated proteins with death domain; TRAILR, TNF-related apoptosis-including ligand receptor; FASR, Fas receptor; tBid, truncated Bid; PARP, poly ADP-ribose polymerase; APAF1, apoptotic protase activating factor 1; MOMP, mitochondrial outer membrane permeabilization; PIP2, phosphatidylinositol-3,4-bisphosphate; PIP3, phosphatidylinositol-3,4,5-triphosphate; PI3K, phosphoinositide 3-kinase.
Cancers 13 04502 g001
Figure 2. Bioactive compounds regulate molecular mechanisms of autophagy. Bioactive compounds initiate autophagy by the formation of a pre-autophagosomal structure via association of PI3K-AMPK, mammalian target of rapamycin (mTOR), ULK1, Vps34, and the Beclin-1 complex, which contribute to the formation of the pre-autophagosomal structure in addition to activating phagophore formation. Fusion of mature autophagosome as well as lysosome causes autolysosome formation. Lastly, elimination of molecules happens by acid hydrolases, which produce nutrients and recycle metabolites.
Figure 2. Bioactive compounds regulate molecular mechanisms of autophagy. Bioactive compounds initiate autophagy by the formation of a pre-autophagosomal structure via association of PI3K-AMPK, mammalian target of rapamycin (mTOR), ULK1, Vps34, and the Beclin-1 complex, which contribute to the formation of the pre-autophagosomal structure in addition to activating phagophore formation. Fusion of mature autophagosome as well as lysosome causes autolysosome formation. Lastly, elimination of molecules happens by acid hydrolases, which produce nutrients and recycle metabolites.
Cancers 13 04502 g002
Figure 3. Schematic diagram of angiogenesis signaling pathways. PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; S6K, S6 kinase; MEK1/2, mitogen-activated protein kinase kinase 1/2; ERK1/2, extracellular signal-regulated kinase 1/2; MNK, mitogen-activated protein kinase-interacting kinase; 4E-BP1, eIF4E-binding protein 1; elF4E1, eukaryotic initiation factor 4E 1; HIF-1α, hypoxia-inducible factor-1 alpha; HIF-1β, hypoxia-inducible factor-1 beta; CBP, CREB-binding protein; p300, CBP homolog; PDGF, platelet-derived growth factor; PDGFR, platelet-derived growth factor receptor; VEGF, vascular endothelial growth factor; NRP1, neuropilin-1; PlGF, placental growth factor; VEGFR-1, vascular endothelial growth factor receptor-1; VEGFR-2, vascular endothelial growth factor receptor-2; bFGF, basic fibroblast growth factor; FGFR, fibroblast growth factor receptors; TGF-β, transforming growth factor beta; TGF-R, transforming growth factor receptor; Dll4, delta-like ligands.
Figure 3. Schematic diagram of angiogenesis signaling pathways. PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; S6K, S6 kinase; MEK1/2, mitogen-activated protein kinase kinase 1/2; ERK1/2, extracellular signal-regulated kinase 1/2; MNK, mitogen-activated protein kinase-interacting kinase; 4E-BP1, eIF4E-binding protein 1; elF4E1, eukaryotic initiation factor 4E 1; HIF-1α, hypoxia-inducible factor-1 alpha; HIF-1β, hypoxia-inducible factor-1 beta; CBP, CREB-binding protein; p300, CBP homolog; PDGF, platelet-derived growth factor; PDGFR, platelet-derived growth factor receptor; VEGF, vascular endothelial growth factor; NRP1, neuropilin-1; PlGF, placental growth factor; VEGFR-1, vascular endothelial growth factor receptor-1; VEGFR-2, vascular endothelial growth factor receptor-2; bFGF, basic fibroblast growth factor; FGFR, fibroblast growth factor receptors; TGF-β, transforming growth factor beta; TGF-R, transforming growth factor receptor; Dll4, delta-like ligands.
Cancers 13 04502 g003
Figure 4. Schematic diagram of metastasis signaling pathways and regulation by bioactive compounds. Akt, protein kinase B; Bak, Bcl-2 antagonist/killer 1; Bax, Bcl-2-like protein 4; Bcl-2, B-cell lymphoma 2; CD44, homing cell adhesion molecule; COX-2, cyclooxygenase 2; EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; MDM2, murine double minute 2; MEK, matrix metalloproteinase-2/9; NF-κB, nuclear factor kappa-B; PI3K, phosphoinositide 3-kinase; PTEN, phosphatase and tensin homolog.
Figure 4. Schematic diagram of metastasis signaling pathways and regulation by bioactive compounds. Akt, protein kinase B; Bak, Bcl-2 antagonist/killer 1; Bax, Bcl-2-like protein 4; Bcl-2, B-cell lymphoma 2; CD44, homing cell adhesion molecule; COX-2, cyclooxygenase 2; EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; MDM2, murine double minute 2; MEK, matrix metalloproteinase-2/9; NF-κB, nuclear factor kappa-B; PI3K, phosphoinositide 3-kinase; PTEN, phosphatase and tensin homolog.
Cancers 13 04502 g004
Figure 5. Schematic diagram of resistance signaling pathway. RTK, receptor tyrosine kinase; IRS1, insulin receptor substrate 1; PI3K, phosphoinositide 3-kinases; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; AKT, protein kinase B (PKB); FOXO3a, forkhead box O 3; IKK-β, inhibitor of nuclear factor κB kinase subunit beta; TNF-α, tumor necrosis factor α; Ub, ubiquitin; KEAP1, Kelch-like ECH-associated protein 1; NRF2, nuclear factor erythroid 2-related factor 2.
Figure 5. Schematic diagram of resistance signaling pathway. RTK, receptor tyrosine kinase; IRS1, insulin receptor substrate 1; PI3K, phosphoinositide 3-kinases; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; AKT, protein kinase B (PKB); FOXO3a, forkhead box O 3; IKK-β, inhibitor of nuclear factor κB kinase subunit beta; TNF-α, tumor necrosis factor α; Ub, ubiquitin; KEAP1, Kelch-like ECH-associated protein 1; NRF2, nuclear factor erythroid 2-related factor 2.
Cancers 13 04502 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kang, S.Y.; Hwang, D.; Shin, S.; Park, J.; Kim, M.; Rahman, M.H.; Rahman, M.A.; Ko, S.-G.; Kim, B. Potential of Bioactive Food Components against Gastric Cancer: Insights into Molecular Mechanism and Therapeutic Targets. Cancers 2021, 13, 4502. https://doi.org/10.3390/cancers13184502

AMA Style

Kang SY, Hwang D, Shin S, Park J, Kim M, Rahman MH, Rahman MA, Ko S-G, Kim B. Potential of Bioactive Food Components against Gastric Cancer: Insights into Molecular Mechanism and Therapeutic Targets. Cancers. 2021; 13(18):4502. https://doi.org/10.3390/cancers13184502

Chicago/Turabian Style

Kang, Seog Young, Dongwon Hwang, Soyoung Shin, Jinju Park, Myoungchan Kim, MD. Hasanur Rahman, Md. Ataur Rahman, Seong-Gyu Ko, and Bonglee Kim. 2021. "Potential of Bioactive Food Components against Gastric Cancer: Insights into Molecular Mechanism and Therapeutic Targets" Cancers 13, no. 18: 4502. https://doi.org/10.3390/cancers13184502

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop