Next Article in Journal
Virtual Evolution of HVEM Segment for Checkpoint Inhibitor Discovery
Next Article in Special Issue
Bioactive Phytochemicals from Mulberry: Potential Anti-Inflammatory Effects in Lipopolysaccharide-Stimulated RAW 264.7 Macrophages
Previous Article in Journal
Mitochondrial Function Are Disturbed in the Presence of the Anticancer Drug, 3-Bromopyruvate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Phytochemicals Derived from Gymnosperms in the Prevention and Treatment of Cancers

1
Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 15731, Iran
2
Student Research Committee, Tabriz University of Medical Sciences, Tabriz 15731, Iran
3
School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
4
Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz 15731, Iran
5
Research Center for Evidence based Medicine, Tabriz University of Medical Sciences, Tabriz 15731, Iran
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(12), 6636; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126636
Submission received: 31 May 2021 / Revised: 16 June 2021 / Accepted: 17 June 2021 / Published: 21 June 2021

Abstract

:
The incidence of various types of cancer is increasing globally. To reduce the critical side effects of cancer chemotherapy, naturally derived compounds have been considered for cancer treatment. Gymnosperms are a group of plants found worldwide that have traditionally been used for therapeutic applications. Paclitaxel is a commercially available anticancer drug derived from gymnosperms. Other natural compounds with anticancer activities, such as pinostrobin and pinocembrin, are extracted from pine heartwood, and pycnogenol and enzogenol from pine bark. Gymnosperms have great potential for further study for the discovery of new anticancer compounds. This review aims to provide a rational understanding and the latest developments in potential anticancer compounds derived from gymnosperms.

1. Introduction

Natural plant products have been used as medicines throughout human history for different purposes. Natural compounds are complex chemical molecules found in various parts of plants with pharmacological or biological activities and are used for the treatment of cancer, inflammation, infections, and other diseases [1]. In the past 30 years, approximately 61% of bioactive natural compounds have been used for cancer treatment, and 49% of these have also been used for infectious disease treatment [2]. Cancer is the second leading cause of death globally, with more than six million deaths each year [3].
The use of natural components for cancer prevention and treatment has been widely studied. Curcumin is a well-known anticancer compound found in the rhizome of Curcuma longa. Curcumin can promote autophagy through the AMP-activated protein kinase signaling pathway, which is related to cell death [4,5]. Another potential candidate for anticancer agents is resveratrol, a polyphenol found in peanuts, soy, and grapes. Resveratrol induces autophagy through the ceramide/Akt/mTOR pathway [6]. 6-Shogaol, an active component of ginger, inhibits cancer cell invasion by blocking the nuclear factor-κB pathway (NFκB) [7]. In addition, flavonoids have different mechanisms for cancer treatment. Cyclin-dependent kinases (CDKs) are inhibited by flavonoids, such as quercetin, silymarin, genistein, luteolin, apigenin, and kaempferol. Different types of cancers are related to the activation of CDKs due to mutations in CDK genes. Therefore, CDK inhibitors are novel agents for cancer treatment [8,9,10]. Epigallocatechin gallate is a polyphenol found in the dried leaves of green tea that initiates cancer cell apoptosis through the activation of the p38 MAP kinase [11]. Many natural products have been commercialized for cancer treatment. One of the most effective natural anticancer agents for the treatment of various cancers is paclitaxel (Taxol®), which was discovered in the bark of Pacific yew, Taxus brevifolia. Paclitaxel is a potent inhibitor of mitosis by binding to tubulin and preventing its depolymerization during cell division [12].
At present, interest in the use of traditional medicine is increasing, and some important anticancer drugs have an herbal origin. Gymnosperms consist of an estimated 12 families, 83 genera, and more than 1000 living species, including those utilized in the production of commercially available drugs and dietary supplements, such as paclitaxel, pycnogenol, enzogenol, pinostrobin, and pinocembrin. Gymnosperms can be a source of effective cancer drugs. Various natural compounds derived from gymnosperms are considered as potential candidates for cancer therapy. Leelamine, α-pinene, and β-pinene have been studied for their ability to inhibit the growth of cancer cell lines [13,14,15]. Other terpenoids in gymnosperms, such as limonene, α-phellandrene, β-caryophyllene oxide, γ-terpinene, longifolene, D-germacrene, verbenol, and β-ocimene, have anticancer effects [16,17,18,19,20]. Due to the anticancer potential of gymnosperms and the relatively few studies that have been conducted in this field, we provide here a comprehensive summary of natural phytochemicals derived from gymnosperms in the prevention and treatment of cancers.

2. Gymnosperms in Cancer Therapy

Gymnosperms are defined as a group of plants that produce seeds that are not enclosed within an ovary or fruit. Gymnosperms include Cycadophyta (cycads), Ginkgophyta (ginkgo), Gnetophyta, and Coniferophyta (conifers). The main group of living gymnosperms is conifers, such as pines, yews, cedars, and redwoods, which are cone-bearing trees and shrubs. More than 600 species have been reported and are dispersed worldwide [21]. Conifers are one of the oldest groups of plants that are ecologically and economically the most important plant group. The leaves of many conifers, such as pines, are needle-like, but yews, cedars, and redwoods have flat, scale-like, and triangular leaves. Cycads are the next most abundant group of gymnosperms. Cycads are palm-like woody plants that make up approximately 338 species, most of which are native to tropical climates. Gnetales is the other living group of gymnosperms, consisting of 95–100 species comprising Gnetum, Ephedra, and Welwitschia. The smallest genus of gymnosperms is Ginkgo, which comprises one extant species [22].
Gymnosperms are reservoirs of terpenoids, phenolic components, alkanes, and alkenes. In Asia, gymnosperms are traditionally used as medicinal plants for various disease treatments. Most are used to treat stomach disorders, arthritis, fever, diabetes, cold, ulcers, and even cancer. Paclitaxel is an anticancer drug obtained from the bark of yew trees. Paclitaxel is used to treat ovarian, breast, lung, and prostate cancers. The structures of the selected commercially available compounds discussed in this review are shown in Figure 1. Leelamine from pine bark shuts off phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinase (MAPK), and signal transducer and activator of transcription 3 (STAT3) pathways in cancer cells [13]. α-Pinene from pine needle oil causes a reduction in the levels of cyclin B protein in hepatoma cell lines in vitro [14]. β-Pinene is a major component of various pine essential oils and inhibits the growth of cancer cell lines [15]. Some commercially available drugs and dietary supplements [12,23,24,25,26,27,28,29,30,31,32,33] are derived from gymnosperms and their physicochemical properties (Table 1). The anticancer effects of stilbenoids in Welwitschia and five species of Gnetaceae were studied. The results showed that these stilbenoids had cytotoxic activity against HL60 cells [34]. Limonene prevents the assembly of microtubules in dividing cells [35]. β-caryophyllene oxide is a sesquiterpene isolated from the essential oils of various species of pine. It has an anticancer effect through the inhibition of c-Src kinase and Janus kinase 2 (JAK2) [36].

3. Clinically Available Natural Products

3.1. Paclitaxel

Paclitaxel (Taxol®) is a well-known chemotherapeutic agent with a taxane structure. It is extracted from the bark of the North American Pacific yew tree, T. brevifolia, with wide anticancer uses, including for small-cell lung cancer, refractory ovarian cancer, Kaposi sarcoma, metastatic breast cancer, and melanoma [37]. Paclitaxel comprises a taxane ring (Figure 1) in the C4 and C5 positions and has a four-membered oxetane side ring in C13, which is an ester side chain that plays a key role in the formation of the paclitaxel-tubulin complex, blocks the progression of the cell cycle in mitosis, and stabilizes microtubules. Paclitaxel binds to the β-subunit of tubulin on the inner surface of microtubules and prevents the dissociation rate of tubulin dimers in a guanosine triphosphate-independent manner. As a result, this complex cannot be disassembled and affects the cell dynamics that are necessary for intra-cell transportation and chromosome movement during mitosis. During the metaphase of mitosis, incomplete formation of the metaphase plate leads to cell cycle arrest. Paclitaxel blocks cells in the G2/M phase of the cell cycle without disrupting the synthesis phase [38,39]. Therefore, paclitaxel is a potent cell replication inhibitor in eukaryotic cells, which ultimately causes apoptosis [40,41]. Paclitaxel induces apoptosis in prostate and breast cancer cell lines by anti-apoptotic B cell lymphoma 2 (Bcl-2) phosphorylation, consequently arresting its function [23]. Recently, paclitaxel was reported to upregulate mitogen-activated protein kinase (MAPK) in ovarian cancer [42]. Among cancer cells, p53 has a pro-apoptotic role in HCT116 colon cancer cells. However, in the presence of paclitaxel, acetylation of p53 was observed, which is more susceptible to cell death [43]. In the human breast adenocarcinoma cell line, paclitaxel induced apoptosis via higher G2/M cell-cycle arrest, reactive oxygen species (ROS) production, and mitochondrial membrane potential disruption. ROS generation and increased hydroperoxide production by nicotinamide adenine dinucleotide phosphate oxidase is considered to play a key role in the anticancer activity of paclitaxel. Upregulation of p21, Bcl-2 associated X protein (Bax), caspase-3, and caspase-9 suggests that apoptosis occurs through intrinsic pathways [23]. Sun et al. reported that paclitaxel induces apoptosis through the activation of Toll-like receptor 4 (TLR4) via the MyD88-independent or -dependent pathway and the NFκB pathway in ovarian carcinoma cells [44]. Phosphorylation of interleukin (IL)-1 receptor-associated kinases by MyD88 initiates a signaling pathway that results in the activation of MAPKs. Furthermore, TLR4, through the TIR-domain-containing adapter-inducing interferon-β adaptors, activates receptor-interacting protein, resulting in NF-κB activation. Paclitaxel treatment induces cytotoxicity, apoptosis, and growth inhibition in the cells, which was confirmed by decreased cell viability. Low doses of paclitaxel induce the enhanced expression of p53 protein without disturbing the cell cycle and its nuclear translocation with well-performed apoptotic events in breast cancer cells [45]. The mechanisms underlying the anticancer activity of paclitaxel are summarized in Figure 2.

3.2. Pinostrobin

Dietary flavonoids are a large group of polyphenolic molecules with low molecular weights obtained from plant sources. Several phenolic compounds have numerous health benefits, including anti-carcinogenic, antioxidant, anti-inflammatory, and antimicrobial activities. Pinostrobin, 5-hydroxy-7-methoxy flavanone (Figure 1), is a natural flavonoid that exists in Pinus strobus, Alpinia zerumbet, Salvia texana, and other plants [46]. Pinostrobin exhibits many therapeutic activities, such as anti-Helicobacter pylori and anti-herpes simplex virus-1 activity, demonstrating its antibacterial and anti-viral activity [47]. Furthermore, the ability of pinostrobin to prevent the cyclooxygenase enzyme pathway indicates anti-inflammatory potential [27]. In addition, pinostrobin has significant effects on cell cycle arrest, growth inhibition, and apoptosis in many cancers [48]. Jaudan et al. demonstrated that HeLa cells are more sensitive to pinostrobin than other cancer cell lines in vitro [27]. They also found that pinostrobin induced ROS generation and apoptosis in HeLa cells via extrinsic and intrinsic pathways. In HepG2 liver cancer cell lines, pinostrobin demonstrated moderate cytotoxicity [49]. Among the various active components isolated from Cajanus cajan, pinostrobin showed maximum toxicity on the T lymphoblastoid cell line derived from patients with acute leukemia, whereas in other cell lines, such as lung cancer, melanoma, and breast cancer cell lines, toxicity was lower [50]. Based on the results of these studies, pinostrobin appears to have anticancer potential.

3.3. Pycnogenol

Pycnogenol® is a formulation of Pinus pinaster (French maritime pine) bark aqueous extract with a composition of a mixture of flavonoids, mainly procyanidin compounds. Procyanidins are chain-like structures comprising catechin, epicatechin, phenolic acids, taxifolin, and cinnamic acids [51]. Pycnogenol is similar to cinnamon, grape seed extract, green tea, and cocoa bean polyphenols, which are the four most common sources of procyanidins. Procyanidins may play an important role in preventing and treating cancer [52]. Procyanidins exhibit inhibitory effects on the proliferation of certain tumor cells in vitro and in vivo. The yields of catechin from P. pinaster bark extract increase as the ethanol concentration of the extract increases. Catechins, major components of pycnogenol, have anti-proliferative and cytotoxic effects on pancreatic, breast, and colorectal cancer cell lines [53]. Furthermore, catechins inhibit protein kinase C and telomerase. In colon adenocarcinoma and monoblastoid leukemia cells, the presence of catechins causes telomere shortening, and chromosomal abnormalities result in life-span reduction [54]. The expression of telomerase in most tumors is a key parameter for explaining the proliferative ability of cancer cells enabled by the preservation of the tips of the chromosomes [55]. Thus, telomerase inhibition could be one of the major mechanisms underlying the anticancer effects of catechins present in pycnogenol [56]. The molecular basis of pycnogenol activity depends on the scavenging of reactive oxygen and reactive nitrogen species and it participates in the cellular antioxidant system [57,58]. Pycnogenol has cardioprotective effects by increasing blood flow through a mechanism associated with increased nitric oxide levels; it also aids in improving blood glucose control, thereby exhibiting anti-diabetic properties [59,60]. Pycnogenol can arrest hydroxyl, superoxide, and free oxygen radicals and reduce lipid peroxidation in red blood cells [61]. The role of pycnogenol in inhibiting NFκB activation, and vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) expression suggests that this antioxidant compound may play a role in cancer prevention and atherogenic processes [62,63]. Abnormal activation of NF-κB has been observed in many cancers. Furthermore, the suppression of NF-κB decreases the proliferation of cancer cells. NF-κB also plays a major role in inflammatory diseases. David et al. demonstrated that pycnogenol reduced lipid peroxidation and carbonyl proteins in ascitic fluid [56]. This action may be related to pycnogenol-mediated effects that cause NF-κB inhibition, reduce IL-1β production, and decrease protein kinase B (Akt) phosphorylation (Figure 3). This study suggests that P. pinaster procyanidins could be a candidate for future studies on multifunctional diet-based cancer prevention approaches.

3.4. Enzogenol

Enzogenol® is a water-soluble proanthocyanidin-rich bioflavonoid extract derived from the bark of Pinus radiata that contains different flavonoids and phenolic acids. Enzogenol is rich in proanthocyanidins, 80% of which include catechin, epicatechin, taxifolin, quercetin, dihydroquercetin, procyanidin dimers, trimers, oligomers, and polymers, and other phenolic acids [64,65]. In vitro and in vivo research has shown that enzogenol has high potential as an antioxidant and anti-inflammatory agent; studies have shown it to effect a decrease in the levels of cardiovascular disease markers and to protect experimental mice against tumor growth [66,67,68,69]. One study demonstrated that enzogenol is much more effective in antioxidant activity than catechin and ascorbic acid under the same conditions [70]. O’Callaghan et al. found that enzogenol elevated caspase-3 activity in a dose-dependent manner [71]. In addition, in the presence of enzogenol, the expression of the anti-apoptotic protein Bcl-2 decreased, leading to apoptosis [33]. Enzogenol has anti-inflammatory and anti-atherosclerotic effects. The anti-inflammatory effects of enzogenol are related to the downregulation of inflammatory cytokines and the inhibition of TNF-α-induced VCAM-1 and ICAM expression [68]. Enzogenol also has cholesterol-lowering properties that can prevent atherosclerosis [72].

3.5. Pinocembrin

Pinocembrin (5,7-dihydroxyflavanone) is a flavonoid that has been isolated from several plants (Figure 1), and Pinus pinastar heartwood is the main source of this compound. Other sources of pinocembrin extraction are Eucalyptus, Euphorbia, Populus, chilca, and honey [73,74]. Pinocembrin is the most significant phytochemical among flavonoids. Its pharmacological activities include anticancer, anti-inflammatory, antioxidant, and antimicrobial properties [75]. Pinocembrin can prevent cancer or reverse disease onset by delaying or stopping the growth of cancer cells. Pinocembrin has cytotoxic effects against breast, colon, cervical, and prostate cancer cell lines [76,77]. In human colorectal adenocarcinoma (HT29) and colon cancer cell lines (HCT116), pinocembrin increases the expression of Bax protein, caspase-3 and -9, and heme oxygenase activity, as well as decreasing the superoxide anion radical and nitric oxide levels [78,79,80]. In prostate cancer cell lines (LNCaP), pinocembrin causes cell cycle arrest at the G2/M phase and is involved in the depletion of mitochondrial membrane potential before apoptosis. Subsequently, cytochrome c is released from the mitochondria and binds to apoptotic protease-activating factor 1 to activate caspase-9, which consequently activates caspase-3 [81,82]. Pinocembrin can activate the PI3K/Akt/mTOR pathway in melanoma cell lines (B16F10 and A375), which may suggest a link between pinocembrin’s pro-apoptotic effect and autophagy [83]. Phosphorylation of phosphatidylinositol-4, 5-bisphosphate by PI3K catalyzes the production of phosphatidylinositol-3, 4, 5-triphosphate (PIP3). PIP3 localized in the inner layer of the plasma membrane promotes the activation of AKT activation, followed by that of the mammalian target of rapamycin (mTOR). Subsequently, mTOR directly phosphorylates unc-51-like kinase 1 during autophagy initiation and autophagosome formation [84]. Zheng et al. demonstrated that pinocembrin can induce apoptosis in melanoma cell lines in a mitochondria-independent manner in vitro and in vivo, as well as create severe endoplasmic reticulum (ER) stress conditions in vitro. The inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (Xbp1)/C/EBP-homologous protein (CHOP) pathway activates ER stress after pinocembrin treatment. First, IRE1 expression increases, followed by an increase in the expression of Xbp1, activating transcription factor 6, and CHOP, which are key factors in ER stress-mediated apoptosis [83]. On the other hand, IRE1 binds with TNF receptor-associated factor 2, which activates caspase-12 and -3, which leads to apoptosis. Phosphorylation of apoptosis signal-regulating kinase 1 after IRE1 activation causes c-Jun-N terminal kinase initiation of Bax, Bcl-2 homologous antagonist/killer and Bcl-2-like protein 11 activation, and B-cell lymphoma-extra-large (Bcl-xL) and Bcl-2 inhibition (Figure 4) [85]. Eventually, the Fas-associated protein death domain, as an extrinsic pathway for apoptosis, activates downstream processes, including the activation of caspases-8,-7,-6, and -3 [86]. These studies showed that pinocembrin is a strong candidate as an anticancer agent with pharmacological potential.

4. The Future Role of Gymnosperms in Cancer Therapy

In recent years, many studies have focused on the anticancer effects of plant derivatives. Gymnosperms have a wide variety of chemical compounds that could play a vital role in cancer treatment in the future. Some of these compounds have strong anticancer properties (Table 2).

4.1. Leelamine

Leelamine (dehydroabietylamine) is a natural diterpene molecule found in the bark of Pinus species. Leelamine targets several signaling pathways involved in cancer because of the disruption of intracellular cholesterol homeostasis [91]. Cholesterol is abundant in the membrane of the mitochondria, ER, and Golgi, and plays a critical role in transport and intracellular signaling systems [92]. When leelamine accumulates in the lysosomes due to its lysosomotropic properties, translocation of cholesterol from the lysosomes to the cytoplasm is blocked. Thus, cholesterol is not available for cancer cell activities and subsequently, receptor-mediated endocytosis and autophagic flux are inhibited. This inhibition is related to receptor tyrosine kinase (RTK) signaling pathways, which can affect the PI3K/AKT, STAT3, ERK, and MAPK signaling cascades [13,93,94]. Kuzu et al. recently reported that leelamine can interrupt important signaling pathways through the phosphorylation of several cell signaling proteins in melanoma cells [13]. There was an alteration in the RTK/AKT signaling pathway and the most important pathways, the PI3K/AKT and AKT/mTOR pathways, were modified after leelamine treatment (Figure 5). Indeed, the hypoactivation of RTKs followed by the effect of extracellular factors inactivates the PI3K/AKT, STAT, and MAPK signaling cascades. Additionally, the reduction of mutant V600E BRAF protein can inactivate the MAPK cascade without RTK effects [95,96]. Mutant Ras and BRAF genes have been found in some cancers, such as melanomas, and play a key role in the growth and spread of cancer cells [97]. STAT3 activity is significantly reduced by leelamine and subsequently, Bcl-2 and Bcl-xL expression levels decrease [13]. Leelamine is a lysosomotropic diterpene that causes autophagic flux disruption and the inhibition of signaling pathways in malignant cancers.

4.2. Stilbenoids

Stilbenoids are formed by the heterogeneous oligomerization of stilbene monomers, such as resveratrol, oxyresveratrol, and isorhapontigenin, and sometimes their glucosides with a molecular backbone, consisting of 1,2-diphenylethylene units [98,99]. Stilbenoids are extracted from various plants, such as those of the families of Welwitschiaceae and Gnetaceae. The compounds have unique structures, and stilbene glucosides and stilbene oligomers are found in Gnetum latifolium and Welwitschia mirabilis. Stilbenoids have several biological activities, such as anticancer, antibacterial, anti-inflammatory, antioxidant, and antiviral activities [100,101]. Previous studies have reported the growth inhibition activity of stilbenoids against human cancer cell lines. In human leukemia HL60 cells, DNA fragmentation and nuclear condensation have been observed. Among the 56 stilbenoids, gnemonol G and gnetin I demonstrated high anticancer activity that was approximately two-fold higher than that of resveratrol [34]. Among stilbenoids, resveratrol is the most well-known and commercially available product. It has demonstrated potent antioxidant, anticancer, anti-inflammatory, and atherosclerosis prevention properties [102]. However, other stilbenoids, such as viniferin, have stronger anticancer activity than resveratrol [103,104]. Figure 6 shows the mechanism of action of stilbenoids; the NF-κB signaling pathway plays a vital role in inflammation; resveratrol has been studied inhibiting this signaling pathway in the treatment of several diseases [105]. Resveratrol also has antioxidant properties through activation of the PI3K/Akt/Nrf2 intracellular signaling pathway and restored SOD and GSH levels [106]. Apoptosis induction by resveratrol has been associated with caspase induction and downregulation of Bcl-2. Recent studies have shown that stilbenoids induced tumor autophagy is an hsp70-dependent mechanism by lysosomal membrane permeabilization [107].

4.3. Pinenes

Pinenes (α- and β-enantiomers) are organic compounds derived from monoterpenes with the molecular formula C10H16. They are the most abundant terpenoids in nature and are mainly found in the essential oils of Pinus species, as well as in various other plants. α- and β-pinenes are extracted from different parts of pines, such as from the needles, nuts, pollen, and bark oils [20,108]. Pinenes have anti-inflammatory and antibacterial activities [109,110]. Both α- and β-pinenes have anticancer activity, and can inhibit the proliferation of human cancer cell lines, including breast, prostate, liver, and colon cancer cell lines [14,111]. Pinenes have good anticancer activity, arrest the cell cycle in the G2/M phase, and inhibit hepatoma cell proliferation [14]. Pinus koraiensis essential oils, mainly consisting of α-pinene, stop the proliferation and migration of human gastric carcinoma cells (MGC-803) and induce apoptosis [89]. This is due to its effect on the Hippo signaling pathway, known as the tumor suppressor, which has been extensively studied [112]. In this pathway, the phosphorylation of YAP occurs and inhibits the migration and proliferation of tumor cells [113]. α-Pinene can decrease the expression of YAP, leading to the inhibition of cancer cells and the induction of apoptosis [89].

4.4. Caryophyllenes

Sesquiterpenes have multiple biological activities, including anticancer, antibacterial, anti-inflammatory, and antiviral activities [20,114]. α-Caryophyllene, β-caryophyllene, and β-caryophyllene oxide are sesquiterpenes from terpenoids naturally isolated from the essential oils of pine species and many other aromatic plants. Caryophyllenes inhibit the proliferation of different types of cancer cells, such as those of liver, breast, lung, and prostate adenocarcinoma [90,115,116]. β-Caryophyllene suppresses STAT3 activation in human breast and prostate carcinoma and in multiple myeloma cell lines via the inactivation of IL-6 [36]. Src, JAK1, and JAK2 may be involved in β-caryophyllene-induced STAT3 inactivation. β-caryophyllene suppresses the PI3K/AKT signaling pathway, and therefore acts as a strong anticancer agent [90].

5. Conclusions and Future Development

For centuries, plants have been used as medicines for the treatment of human diseases in most regions worldwide. Recently, partly due to renewed public interest, scientific research efforts and medical communities around the world have produced considerable information regarding the pharmacological use, effects, and future of herbal medicine development and therapeutic phytochemicals for cancer treatment [117]. While considerable efforts have been made to promote and confirm the efficacy of many traditional therapies or various herbal formulations, well-defined clinical trials and systematic standardized research are still quite limited and should be carried out more extensively to accelerate the development of new phytomedicines [118]. In recent years, the FDA has approved botanical drugs, such as Veregen® (a fraction of the green tea leaf water extract), for the treatment of patients with external genital warts and Crofelemer® (a proanthocyanidin from the Croton lechleri tree latex) for diarrhea treatment in patients with HIV/AIDS [119,120]. This review summarizes the potential role of gymnosperms in the development of anticancer compounds (Table 2). Some potentially pharmacological compounds derived from gymnosperms could serve as new botanical drugs for cancer treatment.

Author Contributions

Conceptualization, H.H. and K.H.K.; Formal Analysis, T.G., J.-H.H., S.A., A.D. and S.F.; Writing—Original Draft Preparation, T.G., J.-H.H., H.H. and K.H.K.; Writing—Review and Editing, T.G. and K.H.K.; Project Administration, H.H. and K.H.K.; Funding Acquisition, H.H. and K.H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported financially by the Drug Applied Research Center, Tabriz University of Medical Science, Tabriz, Iran (Grant No. 58972), and the Iran National Science Foundation (Grant No. 97008952). This work was also supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (grant numbers 2019R1A5A2027340 and 2021R1A2C2007937). This study was also supported by a biological toxicity research grant (KNA 1-3-2, 19-5) of the National Institute of Forest Science affiliated with the Korea Forest Service.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Aktprotein kinase B
ASK1apoptosis signal-regulating kinase 1
ATF6activating transcription factor 6
BakB-cell lymphoma-2 homologous antagonist/killer
BaxBcl-2 associated X protein
Bcl-2B cell lymphoma 2
Bcl-xLB-cell lymphoma-extra-large
BIMB-cell lymphoma-2-like protein 11
Cascellular apoptosis susceptibility protein
caspasecysteine-aspartic proteases
CDKsCyclin-dependent kinases
CHOPC/EBP-homologous protein
Cyt ccytochrome c
ERendoplasmic reticulum
ERKextracellular signal-regulated kinase
FADDFas-associated protein death domain
G1Gap 1 phase for cell enlargement
G2Gap 2 phase
Hsp 70heat shock protein 70
ICAM-1intercellular adhesion molecule 1
IKKNF-κB kinase
IRAKinterleukin-1 receptor associated kinase
IRE1inositol-requiring enzyme 1
JNKc-Jun-N terminal kinase
KSRkinase suppressor of Ras
MAPKmitogen-activated protein kinase
MEKmitogen-activated protein kinase kinase
M phasemitosis phase
mTORmammalian target of rapamycin
MYD88myeloid differentiation primary response 88
NFκBNuclear factor-κB pathway
Nrf2Nuclear factor erythroid 2-related factor 2
PI3Kphosphoinositide 3-kinase
PIP2phosphatidylinositol-4, 5-bisphosphate
PIP3phosphatidylinositol-3, 4, 5-triphosphate
Raf-1Raf kinase family
ROSreactive oxygen species
RTKreceptor tyrosine kinase
S phasesynthesis phase
STAT3signal transducer and activator of transcription 3
TAT3tyrosine aminotransferase 3
tBidtruncated Bid
TLR4Toll-like receptor 4
TRAFTNF receptor associated factor
TRIFTIR-domain-containing adapter-inducing interferon-β
TRAMTRIF-related adapter molecule
ULK1unc-51-like kinase 1
VCAM-1vascular cell adhesion molecule 1
Xbp1X-box binding protein 1

References

  1. Sheikh, I.; Sharma, V.; Tuli, H.S.; Aggarwal, D.; Sankhyan, A.; Vyas, P.; Sharma, A.K.; Bishayee, A. Cancer chemoprevention by flavonoids, dietary polyphenols and terpenoids. Biointerface Res. Appl. Chem. 2020, 11, 8502–8537. [Google Scholar]
  2. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [Green Version]
  3. Loizzo, M.R.; Tundis, R.; Menichini, F.; Saab, A.M.; Statti, G.A.; Menichini, F. Antiproliferative effects of essential oils and their major constituents in human renal adenocarcinoma and amelanotic melanoma cells. Cell Prolif. 2008, 41, 1002–1012. [Google Scholar] [CrossRef]
  4. Aoki, H.; Takada, Y.; Kondo, S.; Sawaya, R.; Aggarwal, B.B.; Kondo, Y. Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: Role of akt and extracellular signal-regulated kinase signaling pathways. Mol. Pharmacol. 2007, 72, 29–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Zhuang, W.; Long, L.; Zheng, B.; Ji, W.; Yang, N.; Zhang, Q.; Liang, Z. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Sci. 2012, 103, 684–690. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, M.; Yu, T.; Zhu, C.; Sun, H.; Qiu, Y.; Zhu, X.; Li, J. Resveratrol triggers protective autophagy through the ceramide/Akt/mTOR pathway in melanoma B16 cells. Nutr. Cancer 2014, 66, 435–440. [Google Scholar] [CrossRef]
  7. Ling, H.; Yang, H.; Tan, S.H.; Chui, W.K.; Chew, E.H. 6-Shogaol, an active constituent of ginger, inhibits breast cancer cell invasion by reducing matrix metalloproteinase-9 expression via blockade of nuclear factor-κB activation. Br. J. Pharmacol. 2010, 161, 1763–1777. [Google Scholar] [CrossRef] [Green Version]
  8. Zi, X.; Feyes, D.K.; Agarwal, R. Anticarcinogenic effect of a flavonoid antioxidant, silymarin, in human breast cancer cells MDA-MB 468: Induction of G1 arrest through an increase in Cipl/p21 concomitant with a decrease in kinase activity of cyclin- dependent kinases and associated cyclins. Clin. Cancer Res. 1998, 4, 1055–1064. [Google Scholar]
  9. Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo, Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest and apoptosis in human breast cancer cells by quercetin. Int. J. Oncol. 2001, 19, 837–844. [Google Scholar] [CrossRef]
  10. Casagrande, F.; Darbon, J.M. Effects of structurally related flavonoids on cell cycle progression of human melanoma cells: Regulation of cyclin-dependent kinases CDK2 and CDK1. Biochem. Pharmacol. 2001, 61, 1205–1215. [Google Scholar] [CrossRef]
  11. Manohar, M.; Fatima, I.; Saxena, R.; Chandra, V.; Sankhwar, P.L.; Dwivedi, A. (-)-Epigallocatechin-3-gallate induces apoptosis in human endometrial adenocarcinoma cells via ROS generation and p38 MAP kinase activation. J. Nutr. Biochem. 2013, 24, 940–947. [Google Scholar] [CrossRef]
  12. Kampan, N.C.; Madondo, M.T.; McNally, O.M.; Quinn, M.; Plebanski, M. Paclitaxel and its evolving role in the management of ovarian cancer. BioMed Res. Int. 2015, 2015, 413076. [Google Scholar] [CrossRef] [PubMed]
  13. Kuzu, O.F.; Gowda, R.; Sharma, A.; Robertson, G.P. Leelamine mediates cancer cell death through inhibition of intracellular cholesterol transport. Mol. Cancer Ther. 2014, 13, 1690–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Chen, W.; Liu, Y.; Li, M.; Mao, J.; Zhang, L.; Huang, R.; Jin, X.; Ye, L. Anti-tumor effect of α-pinene on human hepatoma cell lines through inducing G2/M cell cycle arrest. J. Pharmacol. Sci. 2015, 127, 332–338. [Google Scholar] [CrossRef] [Green Version]
  15. Li, Y.L.; Yeung, C.M.; Chiu, L.C.M.; Cen, Y.Z.; Ooi, V.E.C. Chemical composition and antiproliferative activity of essential oil from the leaves of a medicinal herb, schefflera Heptaphylla. Phytother. Res. 2009, 23, 140–142. [Google Scholar] [CrossRef]
  16. Lin, J.J.; Lu, K.W.; Ma, Y.S.; Tang, N.Y.; Wu, P.P.; Wu, C.C.; Lu, H.F.; Lin, J.G.; Chung, J.G. Alpha-phellandrene, a natural active monoterpene, influences a murine WEHI-3 leukemia model in vivo by enhancing macrophague phagocytosis and natural killer cell activity. In Vivo 2014, 28, 583–588. [Google Scholar]
  17. Shapira, S.; Pleban, S.; Kazanov, D.; Tirosh, P.; Arber, N. Terpinen-4-ol: A novel and promising therapeutic agent for human gastrointestinal cancers. PLoS ONE 2016, 11, e0156540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Sajid, A.; Manzoor, Q.; Iqbal, M.; Tyagi, A.K.; Sarfraz, R.A.; Sajid, A. Pinus roxburghii essential oil anticancer activity and chemical composition evaluation. EXCLI J. 2018, 17, 233–245. [Google Scholar]
  19. Tilaoui, M.; Mouse, H.A.; Jaafari, A.; Aboufatima, R.; Chait, A.; Zyad, A. Chemical composition and antiproliferative activity of essential oil from aerial parts of a medicinal herb Artemisia herba-alba. Braz. J. Pharmacogn. 2011, 21, 781–785. [Google Scholar] [CrossRef] [Green Version]
  20. Ghaffari, T.; Kafil, H.S.; Asnaashari, S.; Farajnia, S.; Delazar, A.; Baek, S.C.; Hamishehkar, H.; Kim, K.H. Chemical Composition and Antimicrobial Activity of Essential Oils from the Aerial Parts of Pinus eldarica Grown in Northwestern Iran. Molecules 2019, 24, 3203. [Google Scholar] [CrossRef] [Green Version]
  21. Simpson, M. Plant Systematics, 2nd ed.; Academic Press: Cambridge, MA, USA, 2010; pp. 1–741. [Google Scholar]
  22. Christenhusz, M.J.M.; Reveal, J.L.; Farjon, A.; Gardner, M.F.; Mill, R.R.; Chase, M.W. A new classification and linear sequence of extant gymnosperms. Phytotaxa 2011, 19, 55–70. [Google Scholar] [CrossRef]
  23. Meher, J.G.; Dixit, S.; Pathan, D.K.; Singh, Y.; Chandasana, H.; Pawar, V.K.; Sharma, M.; Bhatta, R.S.; Konwar, R.; Kesharwani, P.; et al. Paclitaxel-loaded TPGS enriched self-emulsifying carrier causes apoptosis by modulating survivin expression and inhibits tumour growth in syngeneic mammary tumours. Artif. Cells Nanomed. Biotechnol. 2018, 46 (Suppl. S3), S344–S358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Feroz, S.R.; Mohamad, S.B.; Bakri, Z.S.D.; Malek, S.N.A.; Tayyab, S. Probing the Interaction of a Therapeutic Flavonoid, Pinostrobin with Human Serum Albumin: Multiple Spectroscopic and Molecular Modeling Investigations. PLoS ONE 2013, 8, e76067. [Google Scholar] [CrossRef] [Green Version]
  25. Le Bail, J.C.; Aubourg, L.; Habrioux, G. Effects of pinostrobin on estrogen metabolism and estrogen receptor transactivation. Cancer Lett. 2000, 156, 37–44. [Google Scholar] [CrossRef]
  26. Wu, N.; Kong, Y.; Zu, Y.; Fu, Y.; Liu, Z.; Meng, R.; Liu, X.; Efferth, T. Activity investigation of pinostrobin towards herpes simplex virus-1 as determined by atomic force microscopy. Phytomedicine 2011, 18, 110–118. [Google Scholar] [CrossRef] [PubMed]
  27. Jaudan, A.; Sharma, S.; Abd Malek, S.N.; Dixit, A. Induction of apoptosis by pinostrobin in human cervical cancer cells: Possible mechanism of action. PLoS ONE 2018, 13, e0191523. [Google Scholar] [CrossRef] [Green Version]
  28. Rasul, A.; Millimouno, F.M.; Ali Eltayb, W.; Ali, M.; Li, J.; Li, X. Pinocembrin: A novel natural compound with versatile pharmacological and biological activities. BioMed Res. Int. 2013, 2013, 379850. [Google Scholar] [CrossRef]
  29. Enseleit, F.; Sudano, I.; Périat, D.; Winnik, S.; Wolfrum, M.; Flammer, A.J.; Fröhlich, G.M.; Kaiser, P.; Hirt, A.; Haile, S.R.; et al. Effects of Pycnogenol on endothelial function in patients with stable coronary artery disease: A double-blind, randomized, placebo-controlled, cross-over study. Eur. Heart J. 2012, 33, 1589–1597. [Google Scholar] [CrossRef] [Green Version]
  30. Virgili, F.; Kobuchi, H.; Packer, L. Procyanidins extracted from pinus Maritima (Pycnogenol®): Scavengers of free radical species and modulators of nitrogen monoxide metabolism in activated murine raw 264.7 macrophages. Free Radic. Biol. Med. 1998, 24, 1120–1129. [Google Scholar] [CrossRef]
  31. Nelson, A.B.; Lau, B.H.S.; Ide, N.; Rong, Y. Pycnogenol inhibits macrophage oxidative burst, lipoprotein oxidation, and hydroxyl radical-induced DNA damage. Drug Dev. Ind. Pharm. 1998, 24, 139–144. [Google Scholar] [CrossRef]
  32. Wei, Z.H.; Peng, Q.L.; Lau, B.H.S. Pycnogenol enhances endothelial cell antioxidant defenses. Redox Rep. 1997, 3, 219–224. [Google Scholar] [CrossRef]
  33. Li, Y.Y.; Feng, J.; Zhang, X.L.; Cui, Y.Y. Pine bark extracts: Nutraceutical, pharmacological, and toxicological evaluation. J. Pharmacol. Exp. Ther. 2015, 353, 9–16. [Google Scholar] [CrossRef] [Green Version]
  34. Iliya, I.; Akao, Y.; Matsumoto, K.; Nakagawa, Y.; Zulfiqar, A.; Ito, T.; Oyama, M.; Murata, H.; Tanaka, T.; Nozawa, Y.; et al. Growth inhibition of stilbenoids in Welwitschiaceae and Gnetaceae through induction of apoptosis in human leukemia HL60 cells. Biol. Pharm. Bull. 2006, 29, 1490–1492. [Google Scholar] [CrossRef] [Green Version]
  35. Chidambara Murthy, K.N.; Jayaprakasha, G.K.; Patil, B.S. D-limonene rich volatile oil from blood oranges inhibits angiogenesis, metastasis and cell death in human colon cancer cells. Life Sci. 2012, 91, 429–439. [Google Scholar] [CrossRef]
  36. Kim, C.; Cho, S.K.; Kapoor, S.; Kumar, A.; Vali, S.; Abbasi, T.; Kim, S.H.; Sethi, G.; Ahn, K.S. β-Caryophyllene oxide inhibits constitutive and inducible STAT3 signaling pathway through induction of the SHP-1 protein tyrosine phosphatase. Mol. Carcinog. 2014, 53, 793–806. [Google Scholar] [CrossRef] [PubMed]
  37. Singh, S.; Dash, A.K. Paclitaxel in cancer treatment: Perspectives and prospects of its delivery challenges. Crit. Rev. Ther. Drug Carr. Syst. 2009, 26, 333–372. [Google Scholar] [CrossRef] [PubMed]
  38. Barbuti, A.M.; Chen, Z.S. Paclitaxel through the ages of anticancer therapy: Exploring its role in chemoresistance and radiation therapy. Cancers 2015, 7, 2360–2371. [Google Scholar] [CrossRef] [PubMed]
  39. Horwitz, S.B.; Cohen, D.; Rao, S.; Ringel, I.; Shen, H.J.; Yang, C.P. Taxol: Mechanisms of action and resistance. J. Natl. Cancer Inst. Monogr. 1993, 55–61. [Google Scholar] [CrossRef]
  40. Guénard, D.; Guéritte-Voegelein, F.; Dubois, J.; Potier, P. Structure-activity relationships of Taxol and Taxotere analogues. J. Natl. Cancer Inst. Monogr. 1993, 79–82. [Google Scholar]
  41. Kingston, D.G.I. Taxol: The chemistry and structure-activity relationships of a novel anticancer agent. Trends Biotechnol. 1994, 12, 222–227. [Google Scholar] [CrossRef]
  42. Li, Z.; Tang, X.; Luo, Y.; Chen, B.; Zhou, C.; Wu, X.; Tang, Z.; Qi, X.; Cao, G.; Hao, J.; et al. NK007 helps in mitigating paclitaxel resistance through p38MAPK activation and HK2 degradation in ovarian cancer. J. Cell. Physiol. 2019, 234, 16178–16190. [Google Scholar] [CrossRef] [PubMed]
  43. Kim, J.H.; Yoon, E.K.; Chung, H.J.; Park, S.Y.; Hong, K.M.; Lee, C.H.; Lee, Y.S.; Choi, K.; Yang, Y.; Kim, K.; et al. P53 acetylation enhances Taxol-induced apoptosis in human cancer cells. Apoptosis 2013, 18, 110–120. [Google Scholar] [CrossRef]
  44. Sun, N.K.; Huang, S.L.; Chang, T.C.; Chao, C.C.K. TLR4 and NFκB signaling is critical for taxol resistance in ovarian carcinoma cells. J. Cell. Physiol. 2018, 233, 2489–2501. [Google Scholar] [CrossRef] [PubMed]
  45. Panno, M.L.; Giordano, F.; Mastroianni, F.; Morelli, C.; Brunelli, E.; Palma, M.G.; Pellegrino, M.; Aquila, S.; Miglietta, A.; Mauro, L.; et al. Evidence that low doses of Taxol enhance the functional transactivatory properties of p53 on p21 waf promoter in MCF-7 breast cancer cells. FEBS Lett. 2006, 580, 2371–2380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Roman Junior, W.A.; Gomes, D.B.; Zanchet, B.; Schönell, A.P.; Diel, K.A.P.; Banzato, T.P.; Ruiz, A.L.T.G.; Carvalho, J.E.; Neppel, A.; Barison, A.; et al. Antiproliferative effects of pinostrobin and 5,6-dehydrokavain isolated from leaves of alpinia zerumbet. Braz. J. Pharmacogn. 2017, 27, 592–598. [Google Scholar] [CrossRef]
  47. Bankova, V.; Galabov, A.S.; Antonova, D.; Vilhelmova, N.; Di Perri, B. Chemical composition of Propolis Extract ACF® and activity against herpes simplex virus. Phytomedicine 2014, 21, 1432–1438. [Google Scholar] [CrossRef] [PubMed]
  48. Jadaun, A.; Sharma, S.; Verma, R.; Dixit, A. Pinostrobin inhibits proliferation and induces apoptosis in cancer stem-like cells through a reactive oxygen species-dependent mechanism. RSC Adv. 2019, 9, 12097–12109. [Google Scholar] [CrossRef] [Green Version]
  49. Abd El-Hady, F.K.; Shaker, K.H.; Imhoff, J.F.; Zinecker, H.; Salah, N.M.; Ibrahim, A.M. Bioactive metabolites from propolis inhibit superoxide anion radical, acetylcholinesterase and phosphodiesterase (PDE4). Int. J. Pharm. Sci. Rev. Res. 2013, 21, 338–344. [Google Scholar]
  50. Ashidi, J.S.; Houghton, P.J.; Hylands, P.J.; Efferth, T. Ethnobotanical survey and cytotoxicity testing of plants of South-western Nigeria used to treat cancer, with isolation of cytotoxic constituents from Cajanus cajan Millsp. leaves. J. Ethnopharmacol. 2010, 128, 501–512. [Google Scholar] [CrossRef]
  51. Cheah, K.Y.; Howarth, G.S.; Bindon, K.A.; Kennedy, J.A.; Bastian, S.E.P. Low molecular weight procyanidins from grape seeds enhance the impact of 5-Fluorouracil chemotherapy on Caco-2 human colon cancer cells. PLoS ONE 2014, 9, e98921. [Google Scholar] [CrossRef]
  52. Minker, C.; Duban, L.; Karas, D.; Järvinen, P.; Lobstein, A.; Muller, C.D. Impact of procyanidins from different berries on caspase 8 activation in colon cancer. Oxidative Med. Cell. Longev. 2015, 2015, 154164. [Google Scholar] [CrossRef]
  53. Silva, C.; Correia-Branco, A.; Andrade, N.; Ferreira, A.C.; Soares, M.L.; Sonveaux, P.; Stephenne, J.; Martel, F. Selective pro-apoptotic and antimigratory effects of polyphenol complex catechin:lysine 1:2 in breast, pancreatic and colorectal cancer cell lines. Eur. J. Pharmacol. 2019, 859, 172533. [Google Scholar] [CrossRef]
  54. Naasani, I.; Seimiya, H.; Tsuruo, T. Telomerase inhibition, telomere shortening, and senescence of cancer cells by tea catechins. Biochem. Biophys. Res. Commun. 1998, 249, 391–396. [Google Scholar] [CrossRef]
  55. Fusi, J.; Bianchi, S.; Daniele, S.; Pellegrini, S.; Martini, C.; Galetta, F.; Giovannini, L.; Franzoni, F. An in vitro comparative study of the antioxidant activity and SIRT1 modulation of natural compounds. Biomed. Pharmacother. 2018, 101, 805–819. [Google Scholar] [CrossRef] [PubMed]
  56. David, I.M.B.; de Souza Fernandes, F.; dos Santos Silva Ferreira, J.B.; Lüdtke, D.D.; Martins, D.F.; Bobinski, F.; da Silva, T.B.G.C.; Buffon, L.D.; Kopper, M.B.R.; da Silva, G.S.; et al. Dietary supplementation with procyanidin-rich Pinus pinaster extract is associated with attenuated Ehrlich tumor development in mice. Nutr. Res. 2019, 62, 41–50. [Google Scholar] [CrossRef] [PubMed]
  57. Packer, L.; Rimbach, G.; Virgili, F. Antioxidant activity and biologic properties of a procyanidin-rich extract from pine (pinus maritima) bark, pycnogenol. Free Radic. Biol. Med. 1999, 27, 704–724. [Google Scholar] [CrossRef]
  58. Kim, Y.G.; Park, H.Y. The effects of pycnogenol on DNA damage in vitro and expression of superoxide dismutase and HP1 in Escherichia coli SOD and catalase deficient mutant cells. Phytother. Res. 2004, 18, 900–905. [Google Scholar] [CrossRef]
  59. Aydın, S.; Bacanlı, M.; Anlar, H.G.; Çal, T.; Arı, N.; Ündeğer Bucurgat, Ü.; Başaran, A.A.; Başaran, N. Preventive role of Pycnogenol® against the hyperglycemia-induced oxidative stress and DNA damage in diabetic rats. Food Chem. Toxicol. 2019, 124, 54–63. [Google Scholar] [CrossRef]
  60. Tokaç, M.; Bacanli, M.; Dumlu, E.G.; Aydin, S.; Engin, M.; Bozkurt, B.; Yalçin, A.; Erel, Ö.; Kiliç, M.; Başaran, N. The ameliorative effects of pycnogenol® on liver ischemia-reperfusion injury in rats. Turk. J. Pharm. Sci. 2017, 14, 257–263. [Google Scholar] [CrossRef]
  61. Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-inflammatory effects of flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef]
  62. Xia, R.; Ji, C.; Zhang, L. Neuroprotective Effects of Pycnogenol Against Oxygen-Glucose Deprivation/Reoxygenation-Induced Injury in Primary Rat Astrocytes via NF-κB and ERK1/2 MAPK Pathways. Cell. Physiol. Biochem. 2017, 42, 987–998. [Google Scholar] [CrossRef] [PubMed]
  63. Peng, Q.; Wei, Z.; Lau, B.H.S. Pycnogenol inhibits tumor necrosis factor-α-induced nuclear factor kappa B activation and adhesion molecule expression in human vascular endothelial cells. Cell. Mol. Life Sci. 2000, 57, 834–841. [Google Scholar] [CrossRef]
  64. Ramos, V.; Bocalandro, C.; Riquelme, S.; Sanhueza, V.; Aspé, E.; Roeckel, M.; Fernández, K. Effect of the bench scale extraction conditions on Pinus radiata bark extract yield, antioxidant properties and composition. Maderas Cienc. Tecnol. 2013, 15, 31–44. [Google Scholar] [CrossRef] [Green Version]
  65. Walter, A.; Finelli, K.; Bai, X.; Arnett, P.; Bream, T.; Seidenberg, P.; Lynch, S.; Johnson, B.; Slobounov, S. Effect of Enzogenol® Supplementation on Cognitive, Executive, and Vestibular/Balance Functioning in Chronic Phase of Concussion. Dev. Neuropsychol. 2017, 42, 93–103. [Google Scholar] [CrossRef]
  66. Yazaki, Y. Utilization of flavonoid compounds from bark and wood: A review. Nat. Prod. Commun. 2015, 10, 513–520. [Google Scholar] [CrossRef] [Green Version]
  67. Shen, Q.; Hiebert, J.B.; Hartwell, J.; Thimmesch, A.R.; Pierce, J.D. Systematic Review of Traumatic Brain Injury and the Impact of Antioxidant Therapy on Clinical Outcomes. Worldviews Evid. Based Nurs. 2016, 13, 380–389. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, D.S.; Kim, M.S.; Kang, S.W.; Sung, H.Y.; Kang, Y.H. Pine bark extract enzogenol attenuated tumor necrosis factor-α- induced endothelial cell adhesion and monocyte transmigration. J. Agric. Food Chem. 2010, 58, 7088–7095. [Google Scholar] [CrossRef] [PubMed]
  69. Bahramsoltani, R.; Ebrahimi, F.; Farzaei, M.H.; Baratpourmoghaddam, A.; Ahmadi, P.; Rostamiasrabadi, P.; Rasouli Amirabadi, A.H.; Rahimi, R. Dietary polyphenols for atherosclerosis: A comprehensive review and future perspectives. Crit. Rev. Food Sci. Nutr. 2019, 59, 114–132. [Google Scholar] [CrossRef] [PubMed]
  70. Wood, J.E.; Senthilmohan, S.T.; Peskin, A.V. Antioxidant activity of procyanidin-containing plant extracts at different pHs. Food Chem. 2002, 77, 155–161. [Google Scholar] [CrossRef]
  71. O’Callaghan, Y.C.; Drummond, E.; O’Gorman, D.M.; Obrien, N.M. Antioxidant and pro-Apoptotic effects of marine-derived, multi-mineral Aquamin supplemented with a pine bark extract, Enzogenol, and a green tea extract, Sunphenon. J. Med. Food 2013, 16, 920–926. [Google Scholar] [CrossRef]
  72. Sato, M.; Yamada, Y.; Matsuoka, H.; Nakashima, S.; Kamiya, T.; Ikeguchi, M.; Imaizumi, K. Dietary pine bark extract reduces atherosclerotic lesion development in male apoE-deficient mice by lowering the serum cholesterol level. Biosci. Biotechnol. Biochem. 2009, 73, 1314–1317. [Google Scholar] [CrossRef]
  73. Diaz Napal, G.N.; Carpinella, M.C.; Palacios, S.M. Antifeedant activity of ethanolic extract from Flourensia oolepis and isolation of pinocembrin as its active principle compound. Bioresour. Technol. 2009, 100, 3669–3673. [Google Scholar] [CrossRef]
  74. Mandal, M.; Jaganathan, S.K. Antiproliferative effects of honey and of its polyphenols: A review. J. Biomed. Biotechnol. 2009, 2009, 830616. [Google Scholar]
  75. García Forero, A.; Villamizar Mantilla, D.A.; Núñez, L.A.; Ocazionez, R.E.; Stashenko, E.E.; Fuentes, J.L. Photoprotective and Antigenotoxic Effects of the Flavonoids Apigenin, Naringenin and Pinocembrin. Photochem. Photobiol. 2019, 95, 1010–1018. [Google Scholar] [CrossRef]
  76. Aryappalli, P.; Shabbiri, K.; Masad, R.J.; Al-Marri, R.H.; Haneefa, S.M.; Mohamed, Y.A.; Arafat, K.; Attoub, S.; Cabral-Marques, O.; Ramadi, K.B.; et al. Inhibition of Tyrosine-Phosphorylated STAT3 in Human Breast and Lung Cancer Cells by Manuka Honey is Mediated by Selective Antagonism of the IL-6 Receptor. Int. J. Mol. Sci. 2019, 20, 4340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Barbarić, M.; Mišković, K.; Bojić, M.; Lončar, M.B.; Smolčić-Bubalo, A.; Debeljak, Z.; Medić-Šarić, M. Chemical composition of the ethanolic propolis extracts and its effect on HeLa cells. J. Ethnopharmacol. 2011, 135, 772–778. [Google Scholar] [CrossRef] [PubMed]
  78. Kumar, M.A.S.; Nair, M.; Hema, P.S.; Mohan, J.; Santhoshkumar, T.R. Pinocembrin triggers Bax-dependent mitochondrial apoptosis in colon cancer cells. Mol. Carcinog. 2007, 46, 231–241. [Google Scholar] [CrossRef] [PubMed]
  79. Punvittayagul, C.; Wongpoomchai, R.; Taya, S.; Pompimon, W. Effect of pinocembrin isolated from Boesenbergia pandurata on xenobiotic-metabolizing enzymes in rat liver. Drug Metab. Lett. 2011, 5, 1–5. [Google Scholar] [CrossRef] [PubMed]
  80. Žižić, J.B.; Vuković, N.L.; Jadranin, M.B.; Andelković, B.D.; Tešević, V.V.; Kacaniova, M.M.; Sukdolak, S.B.; Marković, S.D. Chemical composition, cytotoxic and antioxidative activities of ethanolic extracts of propolis on HCT-116 cell line. J. Sci. Food Agric. 2013, 93, 3001–3009. [Google Scholar] [CrossRef] [PubMed]
  81. Ricci, M.S.; Zong, W.X. Chemotherapeutic approaches for targeting cell death pathways. Oncologist 2006, 11, 342–357. [Google Scholar] [CrossRef] [Green Version]
  82. Li, J.; Li, X.; Chen, Z.; Rasul, A.; Zhao, C.; Millimouno, F.M.; Tsuji, I.; Yamamura, T.; Iqbal, R.; Malhi, M. Antiproliferative and apoptotic effects of pinocembrin in human prostate cancer cells. Bangladesh J. Pharmacol. 2013, 8, 255–262. [Google Scholar]
  83. Zheng, Y.; Wang, K.; Wu, Y.; Chen, Y.; Chen, X.; Hu, C.W.; Hu, F. Pinocembrin induces ER stress mediated apoptosis and suppresses autophagy in melanoma cells. Cancer Lett. 2018, 431, 31–42. [Google Scholar] [CrossRef]
  84. Zachari, M.; Ganley, I.G. The mammalian ULK1 complex and autophagy initiation. Essays Biochem. 2017, 61, 585–596. [Google Scholar]
  85. Yu, C.; Minemoto, Y.; Zhang, J.; Liu, J.; Tang, F.; Bui, T.N.; Xiang, J.; Lin, A. JNK Suppresses Apoptosis via Phosphorylation of the Proapoptotic Bcl-2 Family Protein BAD. Mol. Cell 2004, 13, 329–340. [Google Scholar] [CrossRef]
  86. Fulda, S.; Debatin, K.M. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 2006, 25, 4798–4811. [Google Scholar] [CrossRef] [Green Version]
  87. Merarchi, M.; Jung, Y.Y.; Fan, L.; Sethi, G.; Ahn, K.S. A Brief Overview of the Antitumoral Actions of Leelamine. Biomedicines 2019, 7, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Martins, A.P.; Salgueiro, L.R.; Gonçalves, M.J.; Proença Da Cunha, A.; Vila, R.; Cañigueral, S. Essential oil composition and antimicrobial activity of Santiria trimera bark. Planta Med. 2003, 69, 77–79. [Google Scholar] [CrossRef]
  89. Zhang, Y.; Xin, C.; Qiu, J.; Wang, Z. Essential oil from Pinus koraiensis pinecones inhibits gastric cancer cells via the Hippo/YAP signaling pathway. Molecules 2019, 24, 3851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Park, K.R.; Nam, D.; Yun, H.M.; Lee, S.G.; Jang, H.J.; Sethi, G.; Cho, S.K.; Ahn, K.S. β-Caryophyllene oxide inhibits growth and induces apoptosis through the suppression of PI3K/AKT/mTOR/S6K1 pathways and ROS-mediated MAPKs activation. Cancer Lett. 2011, 312, 178–188. [Google Scholar] [CrossRef] [PubMed]
  91. Gowda, R.; Madhunapantula, S.V.; Kuzu, O.F.; Sharma, A.; Robertson, G.P. Targeting multiple key signaling pathways in melanoma using leelamine. Mol. Cancer Ther. 2014, 13, 1679–1689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lars Bastiaanse, E.M.; Höld, K.M.; Van Der Laarse, A. The effect of membrane cholesterol content on ion transport processes in plasma membranes. Cardiovasc. Res. 1997, 33, 272–283. [Google Scholar] [CrossRef] [Green Version]
  93. Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The role of resveratrol in cancer therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kallunki, T.; Olsen, O.D.; Jäättelä, M. Cancer-associated lysosomal changes: Friends or foes? Oncogene 2013, 32, 1995–2004. [Google Scholar] [CrossRef] [Green Version]
  95. Sun, J.; Carr, M.J.; Khushalani, N.I. Principles of Targeted Therapy for Melanoma. Surg. Clin. N. Am. 2020, 100, 175–188. [Google Scholar] [CrossRef] [PubMed]
  96. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  97. Bae, J.S.; Kim, Y.; Jeon, S.; Kim, S.H.; Kim, T.J.; Lee, S.; Kim, M.H.; Lim, D.J.; Lee, Y.S.; Jung, C.K. Clinical utility of TERT promoter mutations and ALK rearrangement in thyroid cancer patients with a high prevalence of the BRAF V600E mutation. Diagn. Pathol. 2016, 11, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Shen, T.; Wang, X.N.; Lou, H.X. Natural stilbenes: An overview. Nat. Prod. Rep. 2009, 26, 916–935. [Google Scholar] [CrossRef]
  99. Rivière, C.; Pawlus, A.D.; Mérillon, J.M. Natural stilbenoids: Distribution in the plant kingdom and chemotaxonomic interest in Vitaceae. Nat. Prod. Rep. 2012, 29, 1317–1333. [Google Scholar] [CrossRef]
  100. Akinwumi, B.C.; Bordun, K.-A.M.; Anderson, H.D. Biological activities of stilbenoids. Int. J. Mol. Sci. 2018, 19, 792. [Google Scholar] [CrossRef] [Green Version]
  101. Wahedi, H.M.; Ahmad, S.; Abbasi, S.W. Stilbene-based natural compounds as promising drug candidates against COVID-19. J. Biomol. Struct. Dyn. 2020, 39, 1–10. [Google Scholar] [CrossRef] [PubMed]
  102. Lambert, C.; Lemaire, J.; Auger, H.; Guilleret, A.; Reynaud, R.; Clément, C.; Courot, E.; Taidi, B. Optimize, modulate, and scale-up resveratrol and resveratrol dimers bioproduction in Vitis Labrusca L. Cell suspension from flasks to 20 L bioreactor. Plants 2019, 8, 567. [Google Scholar] [CrossRef] [Green Version]
  103. Colin, D.; Lancon, A.; Delmas, D.; Lizard, G.; Abrossinow, J.; Kahn, E.; Jannin, B.; Latruffe, N. Antiproliferative activities of resveratrol and related compounds in human hepatocyte derived HepG2 cells are associated with biochemical cell disturbance revealed by fluorescence analyses. Biochimie 2008, 90, 1674–1684. [Google Scholar] [CrossRef] [Green Version]
  104. Hyo, J.K.; Eun, J.C.; Song, J.B.; Sun, M.S.; Heui, D.P.; Chang, H.R.; Jun, H.P.; Sang, W.C. Cytotoxic and antimutagenic stilbenes from seeds of Paeonia lactiflora. Arch. Pharmacal Res. 2002, 25, 293–299. [Google Scholar]
  105. Xu, L.; Botchway, B.O.; Zhang, S.; Zhou, J.; Liu, X. Inhibition of NF-κB signaling pathway by resveratrol improves spinal cord injury. Front. Neurosci. 2018, 12, 690. [Google Scholar] [CrossRef]
  106. Hui, Y.; Chengyong, T.; Cheng, L.; Haixia, H.; Yuanda, Z.; Weihua, Y. Resveratrol attenuates the cytotoxicity induced by amyloid-β 1–42 in PC12 cells by upregulating heme oxygenase-1 via the PI3K/Akt/Nrf2 pathway. Neurochem. Res. 2018, 43, 297–305. [Google Scholar] [CrossRef] [PubMed]
  107. Sirerol, J.A.; Rodríguez, M.L.; Mena, S.; Asensi, M.A.; Estrela, J.M.; Ortega, A.L. Role of natural stilbenes in the prevention of cancer. Oxid. Med. Cell. Longev. 2016, 2016, 3128951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Russo, E.B. Taming THC: Potential cannabis synergy and phytocannabinoid-terpenoid entourage effects. Br. J. Pharmacol. 2011, 163, 1344–1364. [Google Scholar] [CrossRef]
  109. Marques, A.P.S.; Bonfim, F.P.G.; Dantas, W.F.C.; Puppi, R.J.; Marques, M.O.M. Chemical composition of essential oil from Varronia curassavica Jacq. accessions in different seasons of the year. Ind. Crop. Prod. 2019, 140, 11656. [Google Scholar] [CrossRef]
  110. Hennia, A.; Nemmiche, S.; Dandlen, S.; Miguel, M.G. Myrtus communis essential oils: Insecticidal, antioxidant and antimicrobial activities: A review. J. Essent. Oil Res. 2019, 31, 487–545. [Google Scholar] [CrossRef]
  111. El-Abid, H.; Amaral, C.; Cunha, S.C.; Augusto, T.V.; Fernandes, J.O.; Correia-da-Silva, G.; Teixeira, N.; Moumni, M. Chemical composition and anti-cancer properties of Juniperus oxycedrus L. essential oils on estrogen receptor-positive breast cancer cells. J. Funct. Foods 2019, 59, 261–271. [Google Scholar] [CrossRef]
  112. Azad, T.; Nouri, K.; Janse van Rensburg, H.J.; Maritan, S.M.; Wu, L.; Hao, Y.; Montminy, T.; Yu, J.; Khanal, P.; Mulligan, L.M.; et al. A gain-of-functional screen identifies the Hippo pathway as a central mediator of receptor tyrosine kinases during tumorigenesis. Oncogene 2020, 39, 334–355. [Google Scholar] [CrossRef] [PubMed]
  113. Bi, L.; Ma, F.; Tian, R.; Zhou, Y.; Lan, W.; Song, Q.; Cheng, X. AJUBA increases the cisplatin resistance through hippo pathway in cervical cancer. Gene 2018, 644, 148–154. [Google Scholar] [CrossRef]
  114. Sarvmeili, N.; Jafarian-Dehkordi, A.; Zolfaghari, B. Cytotoxic effects of Pinus eldarica essential oil and extracts on HeLa and MCF-7 cell lines. Res. Pharm. Sci. 2016, 11, 476–483. [Google Scholar] [PubMed] [Green Version]
  115. Zheng, G.Q.; Kenney, P.M.; Lam, L.K.T. Sesquiterpenes from clove (eugenia caryophyllata) as potential anticarcinogenic agents. J. Nat. Prod. 1992, 55, 999–1003. [Google Scholar] [CrossRef]
  116. Xiao, J.B.; Chen, X.Q.; Zhang, Y.W.; Jiang, X.Y.; Xu, M. Cytotoxicity of Marchantia convoluta leaf extracts to human liver and lung cancer cells. Braz. J. Med. Biol. Res. 2006, 39, 731–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Lagoa, R.; Silva, J.; Rodrigues, J.R.; Bishayee, A. Advances in phytochemical delivery systems for improved anticancer activity. Biotechnol. Adv. 2020, 38, 107382. [Google Scholar] [CrossRef]
  118. Wishart, D.S.; Feunang, Y.D.; Guo, A.C.; Lo, E.J.; Marcu, A.; Grant, J.R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082. [Google Scholar] [CrossRef] [PubMed]
  119. Scheinfeld, N. Sinecatechins: Treatment of genital warts. Drugs Future 2008, 33, 27–30. [Google Scholar] [CrossRef]
  120. Tradtrantip, L.; Namkung, W.; Verkman, A.S. Crofelemer, an antisecretory antidiarrheal proanthocyanidin oligomer extracted from Croton lechleri, targets two distinct intestinal chloride channels. Mol. Pharmacol. 2010, 77, 69–78. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Chemical structures of the commercially available products displayed in Table 1.
Figure 1. Chemical structures of the commercially available products displayed in Table 1.
Ijms 22 06636 g001
Figure 2. Possible mechanism of paclitaxel action. Paclitaxel targets microtubules and inhibits the depolymerization of microtubules by binding to β-tubulin and leading to cell death. Solute carrier organic anion transporter family member 1B3, which is expressed in various tumors, is the most effective influx transporter for paclitaxel. Paclitaxel induces apoptosis via reactive oxygen species production as well as p21, B-cell lymphoma-2 associated X protein, and caspase overexpression and also activates the Toll-like receptor 4/nuclear factor kappa B pathway. (adopted with modification from [12]).
Figure 2. Possible mechanism of paclitaxel action. Paclitaxel targets microtubules and inhibits the depolymerization of microtubules by binding to β-tubulin and leading to cell death. Solute carrier organic anion transporter family member 1B3, which is expressed in various tumors, is the most effective influx transporter for paclitaxel. Paclitaxel induces apoptosis via reactive oxygen species production as well as p21, B-cell lymphoma-2 associated X protein, and caspase overexpression and also activates the Toll-like receptor 4/nuclear factor kappa B pathway. (adopted with modification from [12]).
Ijms 22 06636 g002
Figure 3. Mechanism of pycnogenol anticancer and antioxidant action. Pycnogenol can inhibit PKC and telomerase in cancer cells for life-span reduction. Furthermore, by reducing NFκB activation, VCAM-1 and ICAM-1 can prevent cancer. Pycnogenol has anti-diabetic and cardioprotective effects through increasing nitric oxide levels and reducing lipid peroxidation.
Figure 3. Mechanism of pycnogenol anticancer and antioxidant action. Pycnogenol can inhibit PKC and telomerase in cancer cells for life-span reduction. Furthermore, by reducing NFκB activation, VCAM-1 and ICAM-1 can prevent cancer. Pycnogenol has anti-diabetic and cardioprotective effects through increasing nitric oxide levels and reducing lipid peroxidation.
Ijms 22 06636 g003
Figure 4. Mechanism of pinocembrin induction of apoptosis in cancer cells. In the intrinsic pathway, the expression of the pro-apoptotic proteins B-cell lymphoma-2 (Bcl-2) associated X protein, Bcl-2 homologous antagonist/killer, and Bcl-2-like protein 11 increases and that of the anti-apoptotic proteins Bcl-2 and Bcl-extra-large decreases. Cytochrome c translocates from the mitochondria to the cytosol, leading to apoptosis. In the extrinsic pathway, pinocembrin leads to apoptosis via the Fas-associated protein death domain/caspase-8/caspase-3 signaling pathway.
Figure 4. Mechanism of pinocembrin induction of apoptosis in cancer cells. In the intrinsic pathway, the expression of the pro-apoptotic proteins B-cell lymphoma-2 (Bcl-2) associated X protein, Bcl-2 homologous antagonist/killer, and Bcl-2-like protein 11 increases and that of the anti-apoptotic proteins Bcl-2 and Bcl-extra-large decreases. Cytochrome c translocates from the mitochondria to the cytosol, leading to apoptosis. In the extrinsic pathway, pinocembrin leads to apoptosis via the Fas-associated protein death domain/caspase-8/caspase-3 signaling pathway.
Ijms 22 06636 g004
Figure 5. Mechanism of action of leelamine. Leelamine disrupts receptor tyrosine kinase signaling pathways, leading to a decrease in the phosphoinositide 3-kinase/protein kinase B, mitogen-activated protein kinase, and signal transducer and activator of transcription 3 signaling cascades, resulting in the reduction of B cell lymphoma (Bcl) 2 and Bcl-extra-large expression levels.
Figure 5. Mechanism of action of leelamine. Leelamine disrupts receptor tyrosine kinase signaling pathways, leading to a decrease in the phosphoinositide 3-kinase/protein kinase B, mitogen-activated protein kinase, and signal transducer and activator of transcription 3 signaling cascades, resulting in the reduction of B cell lymphoma (Bcl) 2 and Bcl-extra-large expression levels.
Ijms 22 06636 g005
Figure 6. Mechanism of action of stilbenoids. Stilbenoids induces apoptosis through the caspase activation pathway; they also suppress Bcl-2 expression and apoptosome production. Stilbenoids induce autophagy in cancer cells in an hsp-70 dependent mechanism. These compounds suppress the NF-κB signaling pathway that plays an important role in inflammation. They also activate the Nrf2 antioxidant defense system (adopted with modification from [107]).
Figure 6. Mechanism of action of stilbenoids. Stilbenoids induces apoptosis through the caspase activation pathway; they also suppress Bcl-2 expression and apoptosome production. Stilbenoids induce autophagy in cancer cells in an hsp-70 dependent mechanism. These compounds suppress the NF-κB signaling pathway that plays an important role in inflammation. They also activate the Nrf2 antioxidant defense system (adopted with modification from [107]).
Ijms 22 06636 g006
Table 1. Gymnosperm-derived commercially available products with therapeutic effects.
Table 1. Gymnosperm-derived commercially available products with therapeutic effects.
Commercially Available Product aParental PlantsCommon NameBiological Activity Mechanism of Anticancer ActivityReferences
PaclitaxelTaxus brevifoliaPacific yewAnticancer effectsMicrotubule inhibitor, induces apoptosis, Bcl-2 inhibitor[12,23]
PinostrobinPinus strobusEastern white pineAnti-oxidative, anticancer, anti-inflammatory, and antimicrobial effectsInduces apoptosis, ROS generation in cancer cells, DNA fragmentation[24,25,26,27]
PinocembrinPinus pinasterFrench maritime pineAnticancer, antimicrobial, anti-inflammatory, and antioxidant effectsIncreases the activity of heme oxygenase, caspase-3 and 9, and Bax[28]
PycnogenolPinus pinasterFrench maritime pineAnticancer, anti-inflammatory, antioxidant, blood clotting reduction, and LDL cholesterol-lowering effectsIncreases nitric oxide levels in serum, inhibits NF-kB activity[29,30,31,32]
EnzogenolPinus radiataMonterey pineAnticancer, anti-inflammatory, antioxidant, cardioprotective, and neuroprotective effectsInduces apoptosis, increases the activity of caspase-3, Bcl-2 inhibitor[33]
a Chemical structures of the commercially available products are shown in Figure 1.
Table 2. Gymnosperm-derived chemical compounds with therapeutic effects.
Table 2. Gymnosperm-derived chemical compounds with therapeutic effects.
Chemical Compound *Parental PlantsBiological ActivityMechanism of Anticancer ActivityReferences
LeelaminePinus speciesAnticancerDisruption of cholesterol homeostasis and autophagic flux inhibitor[13,87]
StilbenoidsWelwitschia mirabilisAnticancer, antibacterial, anti-inflammatory, and antioxidantApoptosis and growth inhibition[34]
PinenesPinus speciesAnticancer, antibacterial, and anti-fungalCell-cycle arrest in the G2/M phase and induction of apoptosis[14,88,89]
CaryophyllenesPinus speciesAnticancer, antibacterial, and anti-inflammatorySuppression of STAT3 activation and suppression of the PI3K/AKT signaling pathway[36,90]
* All of chemicals are terpenoids and phenolic compounds.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ghaffari, T.; Hong, J.-H.; Asnaashari, S.; Farajnia, S.; Delazar, A.; Hamishehkar, H.; Kim, K.H. Natural Phytochemicals Derived from Gymnosperms in the Prevention and Treatment of Cancers. Int. J. Mol. Sci. 2021, 22, 6636. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126636

AMA Style

Ghaffari T, Hong J-H, Asnaashari S, Farajnia S, Delazar A, Hamishehkar H, Kim KH. Natural Phytochemicals Derived from Gymnosperms in the Prevention and Treatment of Cancers. International Journal of Molecular Sciences. 2021; 22(12):6636. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126636

Chicago/Turabian Style

Ghaffari, Tayyebeh, Joo-Hyun Hong, Solmaz Asnaashari, Safar Farajnia, Abbas Delazar, Hamed Hamishehkar, and Ki Hyun Kim. 2021. "Natural Phytochemicals Derived from Gymnosperms in the Prevention and Treatment of Cancers" International Journal of Molecular Sciences 22, no. 12: 6636. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126636

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