Next Article in Journal
Overexpression of OsRbohH Enhances Heat and Drought Tolerance through ROS Homeostasis and ABA Mediated Pathways in Rice (Oryza sativa L.)
Previous Article in Journal
Development and Characterization of a New TILLING Population for Forward and Reverse Genetics in Barley (Hordeum vulgare L.)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unveiling Drimenol: A Phytochemical with Multifaceted Bioactivities

by
Zhongming Yang
1,
Kim Wei Chan
1,
Md Zuki Abu Bakar
1,2,* and
Xi Deng
1,*
1
Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
2
Department of Veterinary Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia
*
Authors to whom correspondence should be addressed.
Plants 2024, 13(17), 2492; https://doi.org/10.3390/plants13172492
Submission received: 9 August 2024 / Revised: 2 September 2024 / Accepted: 4 September 2024 / Published: 5 September 2024
(This article belongs to the Section Phytochemistry)

Abstract

:
Drimenol, a phytochemical with a distinct odor is found in edible aromatic plants, such as Polygonum minus (known as kesum in Malaysia) and Drimys winteri. Recently, drimenol has received increasing attention owing to its diverse biological activities. This review offers the first extensive overview of drimenol, covering its sources, bioactivities, and derivatives. Notably, drimenol possesses a wide spectrum of biological activities, including antifungal, antibacterial, anti-insect, antiparasitic, cytotoxic, anticancer, and antioxidant effects. Moreover, some mechanisms of its activities, such as its antifungal effects against human mycoses and anticancer activities, have been investigated. However, there are still several crucial issues in the research on drimenol, such as the lack of experimental understanding of its pharmacokinetics, bioavailability, and toxicity. By synthesizing current research findings, this review aims to present a holistic understanding of drimenol, paving the way for future studies and its potential utilization in diverse fields.

1. Introduction

Plants are among the most versatile and proficient chemists of nature. Utilizing basic molecules, such as carbon dioxide, water, and inorganic ions, they synthesize sugars and subsequently produce an endless array of structurally diverse chemical compounds [1]. Throughout the existence of humans and animals, they have relied on plants for sustenance and various health benefits [2]. It is frequently asserted that every plant possesses medicinal properties. Holistically speaking, a medicinal plant is defined as one that contains bioactive substances (metabolites) in one or more of its organs, which can be utilized for therapeutic purposes or serve as precursors (templates) for the semi-synthesis of drug-like molecules [1]. Recent efforts have focused on developing innovative therapeutic compounds derived from natural products for various applications, such as in the agricultural, food, medicinal, pharmaceutical, cosmetic, and fragrance industries [3]. This means that finding bioactive molecules in natural products remains a great requirement. Furthermore, a shortcut to the discovery of active compounds is from medicinal and ethnopharmacological plants, especially aromatic plants [4].
Aromatic plants have been used for food, spice, and medicinal purposes since the beginning of human history [5]. Over time, they began to be used worldwide in various foods to flavor them and also for preservative purposes [6]. They are used in different forms, such as extracts, essential oils, ground leaves, or powders, to improve flavor and color, and provide antimicrobial and antioxidant effects [6]. The aroma compounds in aromatic plants are responsible for their distinctive smell and taste flavor [7]. Also, these compounds are typically stored in specialized structures, such as glands or secretory cells, and serve various ecological functions, including deterring herbivores, deterring pests, and attracting pollinators [8,9]. These compounds have diverse applications across various industries, including perfumery, aromatherapy, food, traditional medicine, pharmaceuticals, and cosmetics [10,11]. Given this, many scientists have been persistently looking for aromatic compounds with various biological activities from aromatic plants, and have made many gratifying discoveries [12,13]. For example, these aromatic compounds have anti-diabetic, hypolipidemic, antioxidant, anti-inflammatory, anti-tumor, anticonvulsant, antidepressant, anti-nociceptive, and antibiotic activities [7,14].
According to our previous review, Polygonum minus (PM) is a dietary aromatic plant that is frequently used as a food and flavoring agent, and is also a botanical treasure with a wide range of applications and functional properties [15,16]. By studying the composition of its phytochemicals, it was found that the compound named drimenol, presented here, was detected in its different parts, i.e., roots, stems, and leaves [17,18,19,20,21]. Moreover, in a previous article, the bioactivity of drimenol was briefly described in one sentence, including its anti-allergic, cytotoxic, insecticidal, antibacterial, antifungal, molluscicidal, piscicidal, and antifeedant activities, as well as plant growth modulation [22]. Although the biological effects of drimenol have been extensively reported, the majority of previous reports is scattered without systematic summarization [23,24,25]. In this review, we provided a comprehensive and up-to-date summary of the sources, synthesis, and derivatives of drimenol, drawing from the current literature. We emphasized the potential mechanisms underlying its reported bioactivities and discussed future application prospects. This will pave the way for a more informed exploration and utilization of drimenol, fostering its development and application.

2. Drimenol

2.1. Structure and Chemical Properties of Drimenol

Drimenol is a sesquiterpene alcohol with diverse natural bioactivities [26]. Sesquiterpenoids are natural compounds with a 15-carbon frame skeleton that are considered to be important for plant protection and for humans, presenting many biological activities, such as anti-inflammatory, antioxidant, antimicrobial, antitumor, and cytotoxic properties [27,28,29]. Drimenol was first isolated in 1948 from the bark of the aromatic plant Drimys winteri Forst (DW), which is used as a food flavoring and pepper replacement in Argentina and Chile [30]. Its absolute configuration was identified by Appel et al. [31], with the chemical structure displayed in Figure 1. The molecular formula of drimenol is C15H2O6, with a molecular weight of 222.37 g/mol and an appearance of colorless to pale-yellow liquid. Its density is 0.92 g/cm3, boiling point is 260–262 °C at 760 mmHg, and flash point is 110 °C. It was the first bicyclic sesquiterpene with the structure and absolute configuration characteristic of the A, B ring system of many di- and triterpenes [32] and sesquiterpenoid primary alcohol, being methanol in which one of the methyl hydrogens is substituted by a 2,5,5,8a-tetramethyl-1,4,4a,5,6,7,8,8a-octahydronaphthalen-1-yl group. It is also known as [(1S,4aS,8aS)-2,5,5,8a-Tetramethyl-1,4,4a,5,6,7,8,8a-octahydro-1-naphthalenyl] methanol [ACD/IUPAC name]. This unique structure is responsible for its characteristic odor, often described as sweet, woody, and slightly earthy [33]. Drimenol is soluble in various organic solvents, such as ethanol, methanol, dimethyl sulfoxide, and dimethyl formamide. However, drimenol is insoluble in water.

2.2. Plant Sources of Drimenol

Drimenol, a secondary metabolite produced during plant metabolism, is widely distributed across various plants globally, with PM being the primary and most accessible source. All parts of PM (its leaves, stems, and even roots) and its essential oils are rich sources of drimenol [18,19,21,34,35,36,37,38,39]. In addition, drimenol can be isolated from other plants, as listed in Table 1.

2.3. Extraction, Isolation, and Characterization of Drimenol

The methods for extracting drimenol have been extensively researched and well-documented [93,125,126,127,128,129]. The conventional technique for drimenol extraction is maceration [130]. These extraction techniques are primarily simple without complex experimental configurations and are implemented on many plant materials, such as DW bark, Bazzania trilobata, and Polygonum acuminatum Kunth [131,132,133,134,135]. The extraction process typically begins with drying the plant material and grinding it into a fine powder. The solvents used for extraction include ethyl acetate, n-hexane, a mixture of ethyl acetate-n-hexane, and dichloromethane. The concentrated drimenol-containing extract is prepared by evaporating the crude extract in a rotary evaporator. Drimenol was isolated and further purified using preparative column chromatography on silica gel. Primary fractionation of the crude extract using solvents of increasing polarities from hexane to ethyl acetate was analyzed by thin-layer chromatography (TLC). A crystalline compound with different retention times on TLC was then produced and identified as drimenol by NMR.
Nevertheless, traditional extraction methods often require large amounts of solvents, longer extraction times, and are generally less efficient [136]. To address these limitations, some researchers have explored alternative methods [106]. Specifically, branches of Drimys angustifolia were dried in the dark at room temperature and pulverized into fine powder. The branch oils were extracted through hydrodistillation for 4 h using a modified Clevenger-type apparatus under a nitrogen atmosphere. Hexane was added to the crystallized branch essential oil from Drimys angustifolia in a 1:1 volume ratio. Upon heating, a homogeneous mixture was formed, which was subsequently cooled in a refrigerator for several hours, resulting in the separation of drimenol as colorless crystals [106]. It is not difficult to see that they need less extraction solvent, less extraction time, and have a higher extraction efficiency, compared with traditional approaches. Nonetheless, a specially designed experimental setup is required, which may be expensive during installation. These technologies are still in the stage that needs further development.
Once drimenol is extracted and purified from the plant material, it must be characterized to analyze its properties. The productivity of the entire extraction procedure depends on the correct identification of drimenol. Therefore, scientists used Fourier-transform infrared spectroscopy (FTIR) and nuclear magnetic resonance (NMR) and compared the results with those of pure standards by spectroscopic methods [133]. Pure compounds were identified based on the optical rotation, micro-melting point, and spectroscopic data from 1H and 13C NMR [52], and were compared with the literature data for drimenol [137].

2.4. Synthesis of Drimenol

2.4.1. Biosynthesis of Drimenol

A decade ago, Kwon et al. [26] investigated the biosynthesis of drimenol in the roots of the valerian plant (Valeriana officinalis). They identified a novel sesquiterpene synthase cDNA (VoTPS3) that catalyzes the formation of drimenol from farnesyl diphosphate (FPP). NMR analyses, following the purification of the terpene produced by VoTPS3 and the characterization of the VoTPS3 enzyme, confirmed that VoTPS3 synthesizes drimenol. Regarding the mechanism of drimenol synthesis, the researchers suggested that drimenol synthase (DMS) might utilize protonation-initiated cyclization. These findings indicate that VoTPS3 can be used to produce drimenol in plants [26].
In a separate study, researchers identified and characterized both a DMS and a cytochrome P450 drimenol oxidase (PhDOX1) from Persicaria hydropiper, which is involved in the biosynthesis and conversion of drimenol. The expression of DMS alone resulted in the production of drimenol, whereas the co-expression with PhDOX1 primarily yielded drimendiol and cinnamolide in yeast [138]. These results highlight the critical role of DMS in drimenol production.
Consequently, other researchers have focused on DMS, discovering five DMSs of marine bacterial origin [24]. These include two recombinant proteins (Aquimarina spongiae DMS and Rhodobacteraceae KLH11 DMS) and three candidates (Aquimarina spongiae AU474 DMS, Aquimarina spongiae AU119 DMS, and Flavivirga eckloniae DMS), all of which catalyze the biosynthesis of drimenol from FPP [24]. The overall biosynthesis mechanism of drimenol is illustrated in Figure 2.

2.4.2. Chemical Synthesis of Drimenol

Besides biosynthesis pathways, drimenol can be obtained by chemical synthesis. The relatively low content of drimenol in natural sources has spurred studies on its synthetic production [139]. Several methods for drimenol synthesis have been developed (Figure 3). For instance, Akita et al. [140] used lipase ‘PL-266’ from Alcaligenes sp. to perform the enantioselective acetylation of albicanol with isopropenyl acetate, resulting in enantiomerically pure albicanyl acetate and albicanol. Subsequent deprotection of albicanyl acetate produced natural albicanol, which was then converted into drimenol [140].
A convenient and efficient method for drimenol synthesis starting from drimane-8α,11-diol 11-monoacetate was proposed by Kuchkova et al. [141]. This process is highlighted due to its mild conditions and good yield. The key step involves treating drimane-8α,11-diol 11-monoacetate with sulfuric acid in ethanol. The reaction proceeds under mild conditions (20 °C for 18 h), yielding a mixture of drimenol and its isomer drim-8(12)-en-11-ol in approximately a 10:1 ratio. The mixture is then recrystallized from hexane to obtain pure drimenol, with a yield of 52.8%, and its physicochemical properties match those of an authentic sample [141].
Additionally, Aricu [139] summarized several compounds that can be used for drimenol synthesis, including ambreinolide, labdanoid gispanolone, larixol, sclareol, monoacetate, and driman-8α,11-diol [139]. In another study, Rihak et al. [142] successfully isolated gram-scale quantities of highly pure polygodial from Tasmannia lanceolata in a few hours. They employed polygodial for the semi-syntheses of several structurally related natural products, including drimenol [142,143].

3. Biological Activities of Drimenol

As mentioned above, drimenol possesses various biological activities. The elucidation of its biological activities has aroused huge interest in the scientific community. Numerous research reports published to date have explored the varied beneficial biological activities and applications of drimenol. In this section, we will focus on the general biological activities and applications of drimenol, as well as provide a brief description of its mechanism of action.

3.1. Antifungal Activity of Drimenol

Drimenol is a broad-spectrum antifungal agent effective against a wide range of pathogenic organisms, as illustrated in Figure 4. Previous studies have reported its antifungal activity against various pathogenic fungi. It has been determined that the presence of a Δ7,8-double bond in the drimane skeleton of drimenol is a critical structural feature for its antifungal efficacy. Additionally, the aldehyde group at C-9 appears to be non-essential for its activity [127]. To have a comprehensive understanding of its antifungal potential, we summarized the effects of drimenol on various fungi pathogenic to humans, animals, and plants, as well as its mechanisms of action in several fungal species.

3.1.1. Phytopathogenic Fungi

The agricultural industry places significant emphasis on managing plant diseases due to the substantial economic and biosecurity risks posed by plant pathogens [144]. Among these, fungal infections are particularly threatening to food production [145], evidenced by historical events, like the Irish Potato Famine, and contemporary challenges, such as rice blast and wheat rust, which jeopardize food security and incur significant economic losses [146]. Although conventional fungicides have played a crucial role in enhancing food security and controlling agricultural diseases, their associated risks have prompted the exploration of alternative fungal control methods [147]. These alternatives should ideally be environmentally friendly and have minimal adverse effects on animal health when applied exogenously. Over millions of years, plants have evolved diverse defense mechanisms against fungal infections, providing biodegradable, generally non-toxic solutions antagonistic to harmful microorganisms [148]. In this section, we summarize and discuss drimenol as a promising candidate for the future development of antifungal agents for agricultural purposes, as depicted in Figure 4.
The effects of drimenol on the mycelial growth of Botrytis cinerea were evaluated by Robles-Kelly et al. [149]. The results indicated that drimenol inhibited the growth of Botrytis cinerea with an EC50 value of 80 ppm. Moreover, at concentrations of 40 and 80 ppm, the germination rate of Botrytis cinerea was reduced to nearly half of the control value [149,150,151].
Monsálvez et al. [131] investigated the effects of an n-hexane extract from the bark of DW on wheat seedlings inoculated with Gaeumannomyces graminis var. tritici. The study found that a dose of 250 mg/kg of the n-hexane extract effectively controlled Gaeumannomyces graminis, resulting in significantly greater plant height, biomass, chlorophyll content, and stomatal conductance compared to the inoculated control, while also markedly reducing disease severity. Chemical fractionation and analysis of the n-hexane extract revealed that the antifungal activity was primarily associated with compounds such as polygodial, drimenin, drimenol, and isodrimenol. Consequently, the application of a complex mixture of these components can reduce fungal damage severity and protect the growth of wheat seedlings infected by Gaeumannomyces graminis [131,151,152,153].
Scher et al. [134] found that drimenol exhibited moderate antifungal activity against Septoria tritici (IC50: 80.1 μg/mL) and strong activity against Cladosporium cucumerinum (IC50: 6.6 μg/mL). Furthermore, they detected the weak activity of drimenol against Botrytis cinerea and Pyricularia oryzae [134,154,155,156,157,158,159,160].
To develop new antifungal agents based on drimane sesquiterpenes, Edouarzin et al. [161] investigated the antifungal activity of synthetic drimane terpenes, drimenol, and albicanol, along with six analogs. Their results demonstrated that drimenol is a potent fungicide and caused 100% death of various fungi at concentrations of 8–64 µg/mL, such as Rhizopus and Apophysomyces [161,162].
The ethyl acetate crude extracts of endophytic fungi isolated from Litsea petiolata leaves were evaluated for their antifungal activity against two isolates (THL084 and THL861) of Magnaporthe oryzae, the causative agent of rice blast disease determined by Pripdeevech et al. [163]. In a disc diffusion assay, the crude extract from Fusarium sp. MFLUCC16-1462 demonstrated antifungal activity against the THL084 isolate. In addition, after 96 h dual cultures with Fusarium sp. MFLUCC16-1462, the mycelium growth of THL084 and THL 861 was inhibited by 61.96% and 31.74%, respectively. Moreover, the major components of Litsea petiolate crude extracts were pregeijerene B, callitrin, drimenol, and angustione [163].
Among the pathogens that significantly impact global tomato production are Clavibacter michiganensis subsp. michiganensis and Pseudomonas syringae pv. tomato, responsible for bacterial canker and bacterial speck, respectively; the fungus Fusarium oxysporum f. sp. lycopersici, which induces Fusarium wilt; and Phytophthora spp., which affects both potato and tomato cultivation. Montenegro et al. [126] studied the effects of drimenol against these four phytopathogenic microorganisms. Most promisingly, study results displayed that drimenol presented inhibition activity against Fusarium oxysporum f. sp. lycopersici, with MIC and MFC values in the range of 128–256 µg/mL [126,164].

3.1.2. Human or Animal Mycoses

In recent years, the risk of opportunistic mycotic infections has increased in immunocompromised patients, such as those patients receiving organ transplantation, cancer chemotherapy, and with human immunodeficiency virus [165,166]. In immunocompromised patients, the fungus most often causing these infections is Candida albicans, which causes 90% of candidal vaginitis in these patients and healthy women [167]. Despite great advances in drugs to treat mycotic infections, their use is limited by their side effects and the growing resistance of Candida albicans to antifungal drugs [168]. Amphotericin B, considered the “gold standard” antifungal drug, is extensively used for treating severe fungal infections. Nevertheless, its use can lead to nephrotoxicity and infusion-related adverse reactions [169]. Additionally, azole antifungal mediations can generate resistant strains of Candida species. Studies have shown that the resistance rate to fluconazole in isolates collected from women with candidal vaginitis ranges from 3.6% to 7.2% [170]. Moreover, surveys in the United States indicate that approximately 2 million people experience fungal and bacterial infections annually, with 65% of these patients showing resistance to at least one antimicrobial drug [171]. The continuous use of antibiotics and inadequate infection control measures have contributed to instances of drug failure in treating fungal infections [172]. Consequently, there is an increasing need for research into alternative anti-infective therapies and the development of new treatments [173,174].
Medicinal plants, long used in traditional medicine systems to treat fungal infections in humans and animals, are considered valuable sources for discovering new antifungal drugs [175]. Therefore, natural products show great potential in the discovery of new antifungal drugs [176]. Drimenol, a sesquiterpenoid primary alcohol derived from natural sources, offers a promising foundation for the development of novel antimycotic agents [177].
M. G. Derita et al. [135] conducted a study to assess the antifungal properties of the aerial parts of Polygonum acuminatum to validate its traditional use as an antifungal agent and to isolate the compound(s) responsible for its antifungal activity. The study revealed that drimenol was effective against Trichophyton rubrum, Microsporum gypseum, and Trichophyton mentagrophytes (MIC = 62.5 μg/mL), but showed no activity against Aspergillus spp, Saccharomyces cerevisiae, and Candida albicans [135,178].
Edouarzin et al. [161] synthesized drimenol from sclareolide and evaluated its antifungal activities. The results demonstrated that drimenol possessed a strong inhibitory effect on Candida albicans. It not only had a bactericidal effect on Candida albicans (MIC: 32 μg/mL), but also inhibited other fungi, such as Paecilomyces variotii (MIC: 16 μg/mL), Cryptococcus neoformans (MIC: 8 μg/mL), Aspergillus fumigatus (MIC: 8 μg/mL), Fusarium (MIC: 32 μg/mL), Scedosporium (MIC: 16 μg/mL), Saksenaea (MIC: 4 μg/mL), Blastomyces (MIC: 4 μg/mL), fluconazole-resistant strains of Candida parapsilosis (MIC: 32 μg/mL), Candida krusei (MIC: 30 μg/mL), Candida glabrata (MIC: 30 μg/mL), Candida albicans (MIC: 30 μg/mL), and Candida auris (MIC: 50 μg/mL). These findings suggest that drimenol is a broad-spectrum antifungal compound. Furthermore, at concentrations increasing up to 100 μg/mL, drimenol caused the cell wall/membrane disruption of fungi, such as Candida albicans and Cryptococcus spp. [161].
Moreover, Candida auris is an emerging multidrug-resistant strain associated with nosocomial infections, and it has been increasingly reported worldwide [179]. Bioscreen-based growth curve monitoring of drimenol indicated that it had superior activity compared to fluconazole, suggesting its potential usefulness against Candida auris and other drug-resistant fungal pathogens. Since drimenol is active against antifungally resistant strains of Candida auris, Candida albicans, and certain Cryptococcus neoformans, and the mechanism of action of drimenol is different from fluconazole or other clinical antifungal drugs, drimenol could be a useful additional antifungal drug with a novel target. Drimenol demonstrated synergistic activity with fluconazole (FICI < 0.5) against Candida albicans in a checkerboard assay, indicating its potential in combination antifungal therapies [161,180,181].
To elucidate the main characteristics required for drimenol to exhibit antifungal activity, M. Derita et al. [127] tested its efficacy against a unique set of nine fungal strains using standardized methods. The results showed that drimenol was moderately active against Cryptococcus neoformans (MIC100: 125 μg/mL), Microsporum gypseum (MIC100: 62.5 μg/mL, MFC100: 125 μg/mL), Trichophyton rubrum (MIC100: 62.5 μg/mL, MFC100: 125 μg/mL), and Trichophyton mentagrophytes (MIC100: 62.5 μg/mL, MFC100: 125 μg/mL). They also observed that the presence of aldehydes at C-9 or C-8, or a CH2OH group at C-8, was not necessarily required for activity. The Δ7,8-double bond within the drimane skeleton was considered a crucial structural feature for antifungal activity. The fact that both structural types of drimane exhibited antifungal properties strongly indicated that their mechanism of action did not involve a Michael addition. Moreover, the electronic properties of drimanes significantly influenced their antifungal behavior, suggesting that the electronic distribution surrounding the Δ7,8 was pivotal for activity [127].

3.1.3. Antifungal Mechanisms of Drimenol

Fungal cells possess distinct structures, such as cell walls, cell membranes, and nuclei, which antifungal agents can target to inhibit their functions or directly kill fungi [182]. Currently, several antifungal drugs, such as itraconazole, voriconazole, ketoconazole, and fluconazole, are available. These drugs are sometimes used in combination with amphotericin B to treat infections caused by Candida species and ringworm. Nevertheless, these drugs are primarily synthetic or semi-synthetic and, despite their efficacy against fungi, can also harm normal human cells [183]. In contrast, certain natural products offer advantages over synthetic compounds. They often exhibit specific binding to fungal targets and have reduced toxicity to human cells. Natural products can target fungal cell walls, cell membranes and various organelles, disrupting internal processes and impeding fungal cell reproduction [184]. Additionally, the structural optimization of natural products has enhanced their antifungal efficacy, making them competitive with synthetic drugs. Drimenol, a key biosynthetic precursor of various naturally occurring drimane sesquiterpenes, is a potent broad-spectrum antifungal drug [24]. Therefore, this section summarizes the natural antifungal product drimenol with therapeutic efficacy and its molecular targets.
Understanding the mechanism of action of a substance is crucial for predicting potential side effects, anticipating the development of resistance, and guiding the synthesis of novel bioactive compounds [185]. The antifungal mechanisms of drimenol have been extensively studied, as illustrated in Figure 5.

Cytoplasmic Membrane

The plasma membrane of fungi plays a critical role in cell morphogenesis, viability, and pathogenicity [186]. Membrane damage can result in the efflux of cytoplasmic molecules, which in turn can cause fungal cell death [187]. Numerous plant bioactive compounds produce cytotoxicity through targeting the fungal plasma membrane. Notable examples include Thymus vulgaris CT thymol EOs, star anise EOs, tea tree, Palmarosa, esterified p-coumarates, tea tree oil, hinokitiol, mentha piperita, mentha spicata, cuminic acid, Thymus vulgaris CT carvacrol, and the phenanthroindolizidine alkaloid antofine [188,189,190,191,192,193,194,195]. Robles-Kelly et al. [149] used a SYTOX Green uptake assay to assess whether drimenol affects the plasma membrane integrity of Botrytis cinerea [196]. SYTOX Green is a high-affinity nucleic acid dye that is impermeable to the membrane of live cells, but readily penetrates cells with compromised plasma membranes [197]. Their findings showed that methanol, used as a negative control, did not result in fluorescence in the hyphal nuclei. In contrast, ethanol (70% v/v), which causes cell membrane dehydration, served as a positive control, with fluorescent nuclei observed, indicating membrane disruption. Drimenol treatment at concentrations of 40 and 80 ppm for 1 h resulted in clear fluorescence in the conidia of Botrytis cinerea. These results indicate that drimenol disrupts the plasma membrane of Botrytis cinerea, increasing its permeability to SYTOX Green and suggesting that drimenol inhibits fungal growth by compromising membrane integrity [149,196].

Reactive Oxygen Species (ROS) Metabolism

ROS is essential for fungi development. Nonetheless, excessive ROS accumulation can lead to irreversible oxidative damage to cellular components, including DNA, lipids, and proteins [198]. Growing evidence suggests that oxidative damage modulated by ROS is associated with the antifungal activity of plant bioactives [199]. Robles-Kelly et al. [149] examined the effects of drimenol on ROS production by incubating Botrytis cinerea conidia in the presence of drimenol at 21 °C for 2 h. They used the ROS-Glo™ hydrogen peroxide (H2O2) assay, a rapid, homogeneous, and sensitive luminescent assay that directly measured the levels of the H2O2 in cell cultures, to assess ROS production [200]. The results showed that drimenol significantly increased luminescence, thereby increasing ROS production [149].

Expression of Specific Genes

A study identified that the protective effect of drimenol against Botrytis cinerea is linked to the expression levels of specific genes associated with cellular damage [149]. To elucidate the mechanism of action of drimenol, Robles-Kelly et al. [149] investigated the expression changes in genes related to cellular damage, focusing on bchex, which encodes a key protein of the Woronin body, and bcnma and cas-1, which are involved in programmed death cells (PDCs). Bioinformatics analysis revealed these genes in Botrytis cinerea. The cas-1 gene is an ortholog of cas-A, corresponding to a metacaspase, while bcnma, a homolog of the yeast NMA11 gene, belongs to the high-temperature-requirement (HtrA) family of serine proteases and is homologous to the human HtrA2/Omi, a mitochondrial protein with pro-apoptotic functions [201]. The expression of bchex increases in response to hyphae damage, as the Woronin bodies appear to occlude septal pores within minutes [202,203]. Moreover, the gene bcaox1, which is related to oxidative stress in fungi, was analyzed [204,205]. The results showed that, in the presence of drimenol, there was no increase in the transcript levels of genes associated with PCD, suggesting that drimenol negatively modulates their transcription. However, bchex transcripts increased in the presence of drimenol compared to control conditions, indicating hyphal damage. Consistent with these findings, bcaox1 expression was also elevated in the presence of drimenol, indicating mitochondrial dysfunction. This enzyme can accept electrons from the ubiquinone pool and directly reduce oxygen, particularly when complexes III and IV in the inner membrane are impaired [204,205].

Cell Wall and Cell Membrane

The cell wall is vital for pathogenic fungi, serving as a permeability barrier and playing a key role in survival, adaptation, and signaling under stress during infection [206,207]. The ability to grow filamentously is essential for biofilm formation, with hyphae contributing to the structure integrity and multilayered architecture of mature biofilms [208,209]. Hyphae are significant in the pathogenicity of Candida albicans [210,211,212]. Research has shown that Candida albicans can proliferate in either yeast or hyphae forms, with the hyphae form exhibiting greater virulence [213,214]. CRK1, a member of the Cdc2 kinase subfamily, is crucial for hyphal development [215]. The deletion of CRK1 severely impaired hyphal formation under various inducing conditions, whereas the ectopic expression of its catalytic domain enhanced hyphal colony formation, even in conditions conducive to yeast growth [216]. Thus, the CRK1/CRK1 null mutant showed significant deficits in filamentous growth and the transcriptional activation of hyphal-specific genes, highlighting the importance of CRK1 in Candida albicans virulence [217]. To understand the broad-spectrum antifungal potential, Edouarzin et al. [161] evaluated the effects of drimenol against several human pathogenic fungi and analyzed its mechanisms of action in Saccharomyces cerevisiae and Candida albicans. Their yeast mutant screening and spot assay results indicated that drimenol functioned as a fungicidal agent by disrupting cellular processes related to protein trafficking between the Golgi apparatus and the ER, protein secretion (Sec system), and cell signaling, potentially involving CRK1.
Genetic methods have been employed to analyze the mechanisms of action of antifungal compounds through drug-induced hypersensitivity assays [218,219]. Edouarzin et al. [161] employed a similar methodology to demonstrate that drimenol inhibited Candida albicans heterozygous mutants of CDC37, Orf19.4382, Orf19.1672, and Orf19.759—known or putative targets of CRK1 kinase targets—at sub-MIC concentrations [161]. Based on Nelson [220], Cdc2 kinase (of which CRK1 is a member) plays a critical role in regulating retrograde membrane transport from the Golgi to the ER during mitosis, either independently or in conjunction with another kinase, such as MEK1. It is speculated that drimenol may disrupt the interaction between CRK1 and one or more of these gene products [220].
Current research on drimenol highlights its broad-spectrum antifungal activity and potential therapeutic applications, yet significant gaps remain. While drimenol shows promise against various fungal pathogens, including both phytopathogens and human mycoses, the precise molecular mechanisms of its action are not fully understood. Future research should focus on elucidating precise molecular mechanisms of drimenol, its interactions with fungal cellular pathways, and its potential synergies with existing antifungal agents. Additionally, studies could investigate the structural modifications of drimenol to enhance its efficacy and reduce resistance, aiming to optimize its application in both agricultural and clinical settings.

3.2. Antibacterial Activity of Drimenol

The proliferation of bacterial infections poses a significant threat to human life globally. Bacterial diseases represent a foremost concern for human health, ranking as the second leading cause of death as early as 2019 [221]. In recent years, antibiotic resistance has emerged as a primary concern associated with microbial infections. The World Health Organization has emphasized that infections such as pneumonia, septicemia, and foodborne illnesses are increasingly challenging to treat due to this phenomenon [222]. Since the introduction of antibiotics in the 20th century [223], bacteria have developed defense mechanisms that reduce or completely negate the effectiveness of these drugs. Consequently, there is an urgent imperative to explore complementary or alternative approaches to traditional infection treatment protocols. Numerous studies have demonstrated that compounds derived from plants in specific groups exhibited significant antibacterial activity, which can be highly beneficial [224,225,226,227].
Drimenol is one of the most promising sources of bioactive compounds showing antibacterial activity [228,229]. Drimenol and its derivatives have been shown to have antibacterial activity in various studies, such as against Staphylococcus aureus, Pseudomonas aeruginosa, and Mycobacterium vaccae [25]. Several approaches have been employed to assess the potential of drimenol as an antibacterial agent (Figure 6).
The research results of Santos et al. [106] demonstrated that drimenol exhibited MIC values of 67 µg/mL, 1333 µg/mL, 583 µg/mL, 667 µg/mL, and 667 µg/mL against Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumanii, Bacillus cereus, and Staphylococcus aureus, respectively. This indicates that drimenol possesses moderate antibacterial activity against Pseudomonas aeruginosa, Acinetobacter baumanii, Bacillus cereus, and Staphylococcus aureus [106,162].
Moreover, certain herbal plants, such as Drimys granadensis and Polygonum hydropiper, which contain drimenol, are widely used as antibacterial agents. Numerous studies have revealed that drimenol exhibits significant antibacterial effects, even though it is present at varying levels.
Tuberculosis is part of a group of infectious diseases responsible for approximately 90% of global deaths [230]. Mycobacterium tuberculosis, the bacterium causing tuberculosis, infects around eight million new individuals annually and causes a death every 10 s. Despite the designation of tuberculosis by the World Health Organization as a global health emergency, challenges persist, including prolonged treatment durations, limited diagnostic access, and the presence of multidrug-resistant strains of Mycobacterium tuberculosis [231]. Moreover, there has been a notable increase in infections caused by non-tuberculous mycobacteria, such as Mycobacterium kansasii and Mycobacterium avium, which can affect the lungs, lymphatic system, skin, and joints, leading to severe complications if untreated [232]. Therefore, discovering new active molecules targeting mycobacteria is of urgent importance. Alves et. al. [233] examined the antimycobacterial properties of 18 commercially available plant-derived essential oils by evaluating their efficacy against Mycobacterium kansasii, Mycobacterium avium, and Mycobacterium tuberculosis through MIC measurements. The majority of these essential oils exhibited minimal to no activity against these mycobacteria, with MIC values ranging from 1000 to 2000 µg/mL [233]. However, Amyris balsamifera demonstrated the highest activity against Mycobacterium kansasii, with an MIC of 250 µg/mL. Subsequent gas chromatography-mass spectrometry (GC-MS) analysis of Amyris balsamifera revealed that its major constituents were the sesquiterpenes 7-epi-eudesmol (23.6%), agarospirol (14.0%), eudesmol (12.3%), hedycaryol (10.9%), and drimenol (5.3%) [234].
The antibacterial properties of Drimys granadensis leaf essential oil were assessed against eight bacteria strains, including three Gram-negative and five Gram-positive strains. The results indicated that the Gram-negative bacteria (Escherichia coli, Pseudomonas aeruginosa, and Salmonella enteritidis) exhibited no sensitivity to the essential oil. Among the Gram-positive bacteria, Staphylococcus aureus CAMP (+), Staphylococcus epidermidis, Bacillus cereus, and multiresistant Staphylococcus aureus were sensitive to the oil. Staphylococcus epidermidis was the most sensitive, showing the widest inhibition zone (19 mm), although the zone was not as well-defined as that for Bacillus cereus (16 mm). Listeria monocytogenes did not exhibit an inhibition zone and was the only Gram-positive bacterium tested that was resistant to the oil. Thus, Drimys granadensis essential oil demonstrated antibacterial activity against half of the tested bacteria with varying degrees of effectiveness. The chemical composition of Drimys granadensis essential oil, obtained by the hydrodistillation of the leaves, was analyzed using GC and GC/MS, identifying 85 components. The major compounds were germacrene D (14.7%), sclarene (9.5%), α-cadinol (7.3%), longiborneol acetate (6.3%), drimenol (4.2%), (Z)-β-ocimene (4.2%), α-pinene (3.2%), and β-elemene (2.7%) [112].
Kipanga et al. [235] found that the biofilm inhibitory concentration (BIC) of drimenol required to inhibit 50% of developing biofilms in Staphylococcus aureus and Staphylococcus epidermidis was 14.7 ± 2 μg/mL and 16.4 ± 3 μg/mL, respectively [235].
Proteus mirabilis, a common cause of urinary tract infections, especially among elderly individuals with catheters, is the second most prevalent cause of such infections after Escherichia coli [236]. In the current study, the hexane fraction of Polygonum hydropiper exhibited the highest activity against Proteus mirabilis, generating inhibitory zones of 28 mm at a concentration of 100 μg/mL in the disc diffusion assay. In the well-diffusion assay, the hexane fraction produced an inhibition zone of 25 mm at the same concentration. Furthermore, the Polygonum hydropiper hexane fraction demonstrated significant activity, with an MIC of 100 μg/mL. GC followed by GC-MS analyses identified 124 compounds in the hexane fraction of Polygonum hydropiper, with the most abundant being 9,12,15-octadecatrienoic acid, drimenol (7.26%); methyl palmitate (7.68%), caryophyllene oxide (7.7%), methyl ester (8.85%), and humulene oxide (13.79%) were the compounds found abundantly [96].
Drimenol shows promise as an antibacterial agent, yet significant research gaps remain. Its precise antibacterial mechanisms are not fully understood, and its effectiveness against multidrug-resistant strains is underexplored. Future studies should focus on uncovering its molecular targets, evaluating its synergy with current antibiotics, and assessing its safety and pharmacokinetics to better establish its therapeutic potential.

3.3. Anti-Insect Activity of Drimenol

According to [237], plants have evolved various defense mechanisms to protect themselves from natural enemies. In response, pests have developed strategies to overcome these defenses. Plants typically produce compounds known as allelochemicals, which serve as protective agents against predators and microbes. These allelochemicals also help defend against vertebrates, given the similarity in neuronal signaling pathways across the animal kingdom. Moreover, combinations of secondary metabolites may offer more sustained protection against herbivores and pests than individual compounds.
Synthetic pesticides have long been employed as a prominent method for pest control. However, chemical pesticides, such as methyl bromide, phosphine, ethane dinitrile, sulfuryl fluoride, ethyl formate, and carbonyl sulfide, are associated with adverse effects on human health and the environment. Consequently, botanical products have garnered significant interest as potential alternatives. In the search for bio-insecticides, drimenol has been extensively studied as a promising substitute for conventional insecticides.
The effectiveness of insecticides varies across insect species, depending on the physiological traits of the insects and the type of insecticidal plant used. The components of various botanical insecticides can be classified into six categories: attractants, chemosterilants, growth retardants, toxicants, feeding deterrents/antifeedants, and repellents [238].

3.3.1. Toxicant

Insect toxicants are substances that are harmful or lethal to insects upon exposure, leading to their incapacitation or death [239]. Some researchers found that drimenol was toxic and caused death to insects [128,132,240].
Cereals serve as a primary source of dietary protein for humans [241]; however, they are frequently infested by various stored food pests, mainly Coleoptera. These infestations lead to both quantitative and qualitative losses and can adversely affect food safety [242]. Cereal losses during storage can reach up to 50% of total production [243]. Effective grain handling and storage practices are essential to mitigate damage caused by insects, especially the wheat weevil, Sitophilus granarius L. (Coleoptera: Curculionidae), a persistent and destructive pest of stored grains. This insect significantly impacts the quality and yield of maize, wheat, and rice. Although several chemical fumigants with a broad spectrum of activity are employed to combat stored food pests, concerns about their adverse effects persist. These concerns include pesticide residues, pest resistance, ozone depletion (especially from halogenated fumigants), toxicity to non-target organisms, and environmental contamination [244,245]. Consequently, there is growing interest in discovering new bioactive compounds to address insect infestations. Plant-derived natural products are often preferred over traditional fumigants due to their low toxicity and biodegradability.
Paz et al. [132] assessed the dose-dependent toxicity of drimenol against Sitophilus granaries. They assessed acute mortality rates by incorporating varying concentrations of drimenol into the diet over six days, ranging from 0.25% to 3% (w/w). At the highest concentration, drimenol exhibited complete insect mortality within six days, with an LC50 value of 0.31% (w/w). Grain damage analysis after six days revealed that grains treated with 0.5% (w/w) drimenol experienced fewer attacks (13% incidence) compared to those treated with 0.25% (w/w) drimenol (15% incidence) [132]. This trend was consistent with the emergence of new insects relative to the initial population after 21 days. Specifically, grains treated with 0.5% (w/w) drimenol showed a 7% emergence rate of new insects, compared to a 9% emergence rate in those treated with 0.25% (w/w) drimenol. These findings highlight the potential of drimenol as a promising candidate for developing more effective derivatives against storage pests [240].
Drosophila melanogaster, commonly known as the vinegar fly or fruit fly, is a small dipterous insect typically measuring 3–5 mm long, with yellow–red coloring and red eyes. This insect possesses a rapid development cycle, thriving on access to sugary liquids. Its primary impact lies in the transmission of diseases such as sour rot, identifiable by the distinct vinegar odor emanating from contaminated substances. Montenegro et al. [128] assessed mortality over 168 h and determined EC50 values within the initial six days through no-choice test experiments (insecticidal bioassay) targeting first instar larvae of Drosophila melanogaster. Surviving larvae subsequently underwent pupation and emerged as normal insects. The larvicidal test lasted for six days, as the biological cycle from larva to pupa in Drosophila melanogaster takes seven days. The results showed that drimenol presented moderate activity, with an EC50 of >100 mg/L and 15.0% mortality over 168 h [128].

3.3.2. Feeding Deterrents/Antifeedants

In plants, certain compounds can provoke a range of insect behavioral responses, ranging from stimulating to deterring. Deterrents, also known as antifeedants, often indicate the unsuitability of a plant by inhibiting or disrupting feeding. These compounds render treated plant materials unattractive or unpalatable, leading insects to avoid these materials and thereby preventing the ingestion of potentially toxic compounds [246,247,248]. Generally, insect antifeedants possess qualities that are highly desirable for environmentally friendly crop protection agents, as they can be effective at low concentrations and their ability to target specific insect pests without harming beneficial insects or other species [249,250]. Additionally, the likelihood of insect species developing heritable resistance to these antifeedants is considered low [250,251]. Being of natural origin, these compounds are not expected to persist in the environment, offering reduced toxicity compared to synthetic pesticides and a more targeted bioactivity against specific insect pests [252,253,254]. Consequently, the investigation of insect antifeedants as crop protectants has garnered significant attention from researchers [255,256,257,258]. Unlike conventional pesticides, the active components in antifeedants do not directly cause pest mortality. Instead, they work by inducing starvation or making pests more susceptible to predation by their natural enemies. Therefore, innovative strategies are required to effectively utilize antifeedants in the field under varying environmental conditions [256].
Studies have shown that drimenol can be synthesized from drimenyl pyrophosphate through fermentation processes. This compound can subsequently be converted into antifeedants, such as warburganal and 9-hydroxydrimenal, using cost-effective methods [59,259]. Montenegro et al. (2013) aimed to identify more potent compounds derived from DW, some of which exhibited feeding dissuasive activity against Droshophila melanogaster larvae in choice assays. Drimenol demonstrated antifeedant effects, even at concentrations as low as 5 ppm, with an inhibition rate of 51.82%. An antifeedant index above 75% is typically considered significant, while moderate inhibition falls within the 50% to 75% range. Larvae treated with drimenol showed moderate feeding inhibition compared to the control, as evidenced by the reduced weight at lower concentrations of drimenol [128].
Current research on drimenol anti-insect activity reveals its potential as a bio-insecticide, but highlights several gaps. Although drimenol has demonstrated insecticidal and antifeedant properties, its mechanisms of action remain poorly understood, particularly in terms of its impact on various insect species and resistance development. Moreover, the effectiveness of drimenol in diverse environmental conditions and its safety profile for non-target organisms have not been thoroughly investigated. Future research should focus on elucidating the molecular targets of drimenol in insects, optimizing its formulation for different pest species, and conducting comprehensive ecological impact assessments to ensure its viability as a sustainable alternative to synthetic pesticides.

3.4. Antiparasitic Activity of Drimenol

Chagas Disease (CD) is a severe and potentially fatal condition caused by the protozoan parasite Trypanosoma cruzi, transmitted by blood-feeding triatomine insects from the Reduviidae family [260]. In addition to an estimated 6–8 million people currently infected and approximately 50,000 deaths annually, 65–100 million individuals reside in regions at risk of infection [261,262]. The only drugs available for CD treatment, nifurtimox and benznidazole, have been in use since the 1960s [263]. However, these medications can cause severe side effects [264]. Moreover, nifurtimox has been discontinued in several countries due to its toxicity [265]. As a result, there is an urgent need for effective anti-Trypanosoma compounds with lower toxicity, driving the exploration of natural products as potential new drug candidates [266,267,268]. Muñoz et al. [269,270,271] assessed the in vitro activity of several Chilean plant extracts against the trypomastigote forms of Trypanosoma cruzi. Drimenol was isolated from active DW extract, demonstrating activity comparable to nifurtimox and benznidazole, with an IC50 value of 25.1 µM against Trypanosoma cruzi trypomastigotes [269,270,271].
Parasitic helminths remain a significant concern in human and veterinary medicine, as well as in agriculture [272,273]. Approximately one-third of the global human population, particularly in developing regions, is infected with one or more nematodes, affecting over 2 billion people [274]. Intestinal nematode parasites, such as Ascaris lumbricoides and Trichuris trichiura, can cause a range of symptoms, including intestinal disturbances, systemic discomfort, and weakness, which can impair physical development and hinder the ability to work and study [275,276,277]. In addition, parasitic nematodes contribute significantly to economic losses in the livestock and crop industries worldwide. In the United States alone, nematodes account for an estimated USD 2 billion in annual losses in the livestock industry, due to reduced productivity and increased operational costs [278]. In the absence of vaccines against intestinal nematodes, chemotherapy remains the primary method of controlling infections. However, resistance to anthelmintics has been widely reported among livestock parasites and occasionally in human parasites, with the potential for becoming more common in human infections [272,279,280,281,282]. This situation underscores the urgent need for new anthelmintic compounds with novel mechanisms of action [283]. Medicinal plants offer a promising source of effective anthelmintic drugs due to their traditional use, demonstrated efficacy, and safety [284]. Anthelmintic metabolites derived from these plants may serve as potential drug candidates [285]. Some natural products from medicinal plants have shown efficacy against nematodes in both in vitro and in vivo models of Trichuris muris and Schistosoma mansoni [286,287,288]. However, many effective compounds from medicinal plants have yet to be identified. The free-living nematode Caenorhabditis elegans has proven to be a valuable model for discovering new anthelmintic drugs and elucidating their mechanisms of action or resistance [285,289,290,291]. Due to its ease of maintenance in the laboratory, small size, and short generation time, Caenorhabditis elegans is well-suited for testing the anthelmintic effects of crude plant extracts or pure compounds without the need for host infection experiments [292,293,294]. Liu et al. [295] utilized Caenorhabditis elegans as a model system to identify novel anthelmintic compounds from medicinal plants by assessing the motility of the nematode.
As polygodial exhibited potent activity against Caenorhabditis elegans, Liu et al. [295] investigated the anthelmintic activity of 18 polygodial analogs to elucidate the structural features that influence their bioactivity. Most of these analogs contained the drimane sesquiterpene skeleton. The findings revealed that drimenol had potent activity, with an IC50 value of 49.1 ± 10.1 μM [295]. Thus, drimenol may be a promising candidate for anthelmintic agent development and warrants further exploration.
Current research on drimenol antiparasitic activity shows promise but has significant gaps. While it exhibits notable in vitro effects against Trypanosoma cruzi and nematodes, its in vivo efficacy, safety, and pharmacokinetics are not well understood. Additionally, the mechanisms of its antiparasitic action remain unclear. Future research should focus on in vivo evaluations and uncovering its molecular targets and resistance mechanisms to improve its potential as an antiparasitic treatment.

3.5. Cytotoxic Activity of Drimenol

Cytotoxicity refers to the extent to which a chemical compound or substance can damage or destroy cells [296]. The toxicity of a compound to cells typically depends on extrinsic factors, including its physiochemical properties, such as structure, shape, surface, size, solubility, aggregation, and chemical nature. Toxic compounds can compromise cell membranes, leading to reduced cell viability and proliferation [296,297]. Therefore, investigating the cytotoxic properties of compounds is valuable for screening and preliminary assessing their biological properties [298].
Numerous studies have documented the cytotoxic effects of drimenol. Kahlos et al. [299] investigated the volatile constituents of Gloeophyllum odoratum. Using GC and GC-MS analyses, they identified the primary volatiles as linalool, citronellol, geraniol, and drimenol. These volatile oils demonstrated toxicity to brine shrimp larvae (Artemia salina), suggesting potential insecticidal and cytotoxic properties [299]. Moreover, Montenegro et al. [300] proposed that drimenol exhibits diverse biological activities, including antifeedant, cytotoxic, antibacterial, and antifungal effects. Similarly, Melo et al. [301] reported the multifaceted properties of drimenol, derived from Canellaceae species, which exhibited anti-inflammatory, cytotoxic, antifungal, and antibacterial effects [301].
Additionally, Mahnashi et al. [302] identified various compounds from Polygonum hydropiper, including warburganal and drimane-type sesquiterpenoids, such as confertifolin, drimenol, isopolygodial, polygodial, and isodrimeninol, which were found to possess cytotoxic properties [302]. Some researchers assert that drimenol, present in various species of the Polygonum (Polygonaceae) and Drimys (Winteraceae), is characterized by a bicyclic farnesane-type skeleton. This structural feature contributes to its diverse biological activities, which include antifungal, antibacterial, and cytotoxic effects [23,125].
The research suggests that antibacterial properties are often associated with cytotoxic effects [303]. As mentioned above in Section 3.2 (antibacterial), since drimenol possesses antibacterial properties, it is unsurprising that it also exhibits cytotoxic effects.
Current research on drimenol cytotoxic activity reveals a promising spectrum of effects, yet significant gaps remain. While drimenol has demonstrated cytotoxicity across various cell types and biological systems, the mechanisms underlying its cytotoxic action are not well understood. Additionally, studies have often focused on broad assessments of drimenol activity, without detailed investigations into its selectivity and potential off-target effects. Future research should prioritize elucidating the specific cellular pathways and molecular targets affected by drimenol to better understand its cytotoxic mechanisms.

3.6. Anticancer Activity of Drimenol

Cancer is a progressive disease characterized by uncontrolled and abnormal cell proliferation [304]. Annually, cancer accounts for approximately 9.8 million deaths globally, making it the second leading cause of death worldwide [304,305]. Extensive research has focused on developing natural medicines for cancer treatment, leading to the discovery of several anticancer drugs derived from medicinal plant compounds, such as podophyllotoxin, vinca alkaloids, and taxanes [304]. Drimenol is a very interesting plant secondary metabolite with tremendous biological activities [22], with anticancer activity being particularly noteworthy. Given the global burden of cancer, the cytotoxicity of drimenol against various cancer types has garnered considerable attention from natural product chemists. This section reviews recent studies on the cytotoxic effects of drimenol against cancer cells.

3.6.1. In Vitro Study

Russo et al. [125] investigated the effects of an ethyl acetate extract from the bark of DW, which includes sesquiterpenoids such as polygodial, isonordrimenone, nordrimenone, and drimenol, on human melanoma cells. Drimenol treatment resulted in a significant decrease in cell viability in A2058 and A375 melanoma cells, with IC50 values of approximately 33.50 ± 0.03 μM and 31.25 ± 0.05 μM, respectively. Notably, drimenol showed no cytotoxic effects on normal human buccal fibroblasts at higher concentrations. Furthermore, drimenol (12–25 μM) induced DNA damage in A375 cells in a dose-dependent manner. The TUNEL assay confirmed that treatment with drimenol (12–25 μM) for 72 h led to a significant increase in green fluorescence indicative of DNA fragmentation. Additionally, drimenol treatment resulted in the reduced expression of the heat shock protein Hsp70 in cancer cells. Drimenol (25 μM) was also found to inhibit the anti-apoptotic protein Bcl-2, activate the pro-apoptotic protein Bax, and increase caspase-9 levels. These findings suggest that drimenol downregulates Hsp70 expression and may play a role in the apoptotic process, indicating its potential as a drug candidate for combination therapy in melanoma treatment [125].

3.6.2. In Vivo Study

Essential oil from Siparuna guianensis, which contains 13.7 ± 0.2% drimenol, demonstrated antitumoral activity. Treatment with this essential oil resulted in a significant reduction in tumor cell counts (59.76 ± 12.33) compared to the untreated control group (96.88 ± 19.15). Additionally, the essential oil decreased MDA levels and increased SOD levels in liver tissue. These results suggest that the essential oil, due to its drimenol content, exhibits both antitumor and antioxidant properties by mitigating oxidative stress [306].

3.6.3. Anticancer Mechanisms of Drimenol

Researchers have extensively explored the anticancer mechanism of drimenol (Figure 7). The transient receptor potential cation channel subfamily V member 1 (TRPV1) is a transmembrane protein that can be activated by various physical and chemical stimuli related to pain transduction. Recent findings have highlighted the significant roles of TRPV1 in cancer tumorigenesis and progression, as its expression levels are altered in various cancer cell types. Numerous studies have identified direct links between TRPV1 and cancer cell proliferation, apoptosis, and metastasis. Consequently, there is increased interest in examining the impacts of TRPV1 agonists and antagonists on cancer development. Both types of compounds may exhibit anticancer effects, either through TRPV1 or via alternative mechanisms [307]. Natural compounds that influence TRPV1 activity include dialdehyde terpenes, such as polygodial and drimenol [308].
Additionally, the modulation of Ca2+ signaling in cancer cells has emerged as a novel therapeutic target [309]. During carcinogenesis, Ca2+ signaling is significantly altered, disrupting normal physiological functions and conferring advantages that facilitate uncontrolled proliferation, resistance to apoptosis, angiogenesis, and adaptation to nutrient-poor conditions. These changes also enhance the ability of cells to invade and metastasize [310]. The literature suggests that plants are a valuable source of phytochemicals that may combat cancer through targeted modulation of Ca2+ signaling. Numerous plants with bioactive compounds have been identified as inhibitors of tumor progression and development [311]. Studies have shown that various natural ligands, including drimenol, can modulate Ca2+ channels [312].
These research perspectives are complementary rather than contradictory. TRPV1, like the related TRPV2–TRPV4 channels and other transient receptor potential (TRP) channels, features a pore that is non-selective for cations and exhibits significant permeability to Ca2+ [313]. The activation of the TRPV1 channel induces a flux of Ca2+ ions into cells. Intracellular Ca2+ overload leads to cell death [314].
Drimenol shows potential as an anticancer agent, but research gaps remain. While it exhibits anticancer effects through TRPV1 modulation and Ca2+ signaling interference, its impact on different cancer types of cells is not fully explored. Future studies should focus on detailed mechanisms of action, toxicity profiles, and potential synergy with other treatments to better assess drimenol therapeutic potential in clinical settings.

3.7. Antioxidant Activity of Drimenol

Natural antioxidants inhibit the propagation of free radical reactions, thereby protecting the human body from diseases and slowing the oxidative rancidity of lipids in food. This role helps to replace potentially harmful synthetic additives [315]. Consequently, the search for natural antioxidants is of significant importance. The antioxidant activity of drimenol has been explored in recent studies. One study analyzed the principal components of Siparuna guianensis essential oil, which include curzerenone (16.4 ± 1.5%), drimenol (13.7 ± 0.2%), and spathulenol (12.4 ± 0.8%). This oil demonstrated antioxidant activity by inhibiting 11.1 % of DPPH radicals (95.7 mg TE/g) and 15.5 % of β-carotene peroxidation [306]. Numerous other researchers shared the same perspective, asserting that drimenol exhibited antioxidant properties [316,317].
However, contrasting results were found in another study. The researchers detected 39 constituents within the essential oil extracted from Cinnamodendron dinisii, with drimenol comprising 0.2% of the oil. This oil showed low antioxidant activity in the β-carotene/linoleic acid test and was not detectable in the DPPH test [55]. The variability in drimenol antioxidant activity across studies may be attributed to differences in drimenol concentration and extraction methods [318]. To address these gaps, future research should focus on standardizing drimenol extraction and testing methods, as well as investigating its antioxidant mechanisms in diverse biological systems and potential interactions with other antioxidants.

3.8. Other Activities of Drimenol

Studies have investigated additional pharmacological properties of drimenol. Burgos et al. [319] reported that drimenol has potential as a molecular scaffold in drug development for inflammatory vascular diseases. Specifically, drimenol at a concentration of 10 μg/mL inhibited vascular cell adhesion molecule 1 and intercellular adhesion molecule 1 and reduced the adhesion of monocytes cells (THP1s) to human umbilical vein endothelial cells (HUVECs). Eser and Yoldas found that drimenol had anti-inflammatory, anti-allergic, and antiasthmatic effects [42]. In another report, drimenol, the main constituent of Warburgia salutaris bark, was used to treat skin and respiratory ailments [320].

4. Derivatives of Drimenol

Natural products and their derivatives are crucial sources for the development of new drugs [321]. In the past two decades, over 70% of new drugs have been derived from specialized plant metabolites [322]. Between 1981 and 2018, natural products and their synthetically modified analogs constituted approximately 70–80% of bioactive agents used in clinical settings [323]. Therefore, chemically modifying natural products to enhance their biological profiles and address pharmacokinetic issues, such as poor solubility, is highly advantageous [324,325,326]. Natural products are instrumental in identifying new scaffolds with diverse biological activities that can be applied directly [327,328].
Sesquiterpenes, among the most prevalent secondary metabolites derived from plants, have demonstrated significant medicinal value through extensive basic research and clinical applications. Previous studies have highlighted that sesquiterpenes and their derivatives exhibit substantial therapeutic potential against various cancers and are frequently investigated as drug candidates in clinical trials to replace traditional chemotherapeutics [28,329,330].
Drimenol, with its numerous chiral centers, including drimenol polymers, exhibits a wide range of activities and stereoselectivities. As a fundamental framework for active lead compounds, drimenol can enhance structural diversity and serve as a reference for the development of small-molecule drugs. Consequently, research efforts have focused on drimenol and its natural and synthetic derivatives to identify compounds with improved pharmacological properties. Table 2 provides an overview of the findings from different studies on drimenol derivatives, highlighting their biological activities and applications.

5. Conclusions and Perspectives

Naturally occurring compounds have emerged as crucial reservoirs for novel drug development. Recently, they have attracted increased attention due to their proven therapeutic efficacy and minimal toxicity, as demonstrated by compounds such as artemisinin and paclitaxel [390]. Drimenol, a significant natural compound obtainable from various herbal medicines, like PM and DW, stands out in this regard. This study provides a comprehensive overview of drimenol, covering its structure, chemical properties, plant origins, synthesis, derivatives, and biological activities. It shows that drimenol has various biological activities, including antioxidant, antifungal, antibacterial, insecticide, antiparasitic, cytotoxic, and anticancer activities. The summarized information could be valuable for guiding future research and development efforts related to drimenol.
Certainly, the journey toward the practical application of drimenol is still in progress. In the future, we should pay attention to several aspects of the research on drimenol. On the one hand, for the application of drimenol, we still need to conduct a large number of experiments, including in vitro and in vivo studies, to explore and confirm its diverse bioactivities, such as different cancer cell lines, microbial strains, and insect species, to identify its most promising therapeutic applications. Further studies are necessary to fully investigate its mechanism of action and understand its beneficial effects in humans. Elucidating the molecular mechanisms of drimenol action through advanced techniques, like omics, is essential for understanding its effects at the cellular and molecular levels. In particular, animal research and clinical trials must be encouraged. On the other hand, alongside its biological activities, ensuring the safety of drimenol remains a crucial factor constraining its potential applications. Despite reported beneficial effects, as a sesquiterpenoid, there is a need to carefully consider its potential toxicity to both humans and the environment. Presently, there have been no apparent signs of toxicity associated with drimenol. However, toxicity assessments have been limited to cellular studies. Rigorous in vivo experiments are imperative to comprehensively assess any potential side effects or toxicity, thus ensuring its safe utilization. Moreover, the absence of studies on the pharmacokinetics and bioavailability of drimenol poses a limitation. However, approaches like nanotechnology or microparticle delivery systems, which have proven effective in overcoming the poor bioavailability challenges of natural alkaloids such as berberine, could offer potential solutions to this limitation [391,392]. This includes the development of advanced drug delivery systems, such as nanoparticles and liposomes, to enhance bioavailability and ensure targeted delivery [393,394,395]. Moreover, chemical modification and drug combinations represent viable strategies for enhancing the bioavailability of bioactive compounds [396,397]. In summary, drimenol exhibits a broad spectrum of biological activities. To advance its development and applications, future research should focus on the following areas: (1) conducting additional studies to further elucidate its biological activities; (2) applying in vivo and in vitro studies to clarify the molecular mechanisms underlying its effects; (3) investigating its pharmacokinetics and bioavailability to assess biological activities; and (4) performing comprehensive toxicity assessments to determine its safety profile. By focusing on these research directions, drimenol’s promising bioactivities can be effectively translated into safe and effective clinical applications.

Author Contributions

Z.Y.: Writing—original draft, Conceptualization, and Funding acquisition. K.W.C.: Writing—review and editing. M.Z.A.B.: Funding acquisition. X.D.: Writing—original draft and Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funding from Universiti Putra Malaysia, Geran Putra IPS (GP-IPS/2023/9748400).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sharma, A.; Tiwari, P.; Arora, R.; Sankaranarayanan, A. Madagascar Periwinkle Alkaloids: Biosynthesis, Ethnobotanical Attributes, and Pharmacological Functions. S. Afr. J. Bot. 2022, 151, 108–115. [Google Scholar] [CrossRef]
  2. Abdallah, E.M. Black Pepper Fruit (Piper nigrum L.) as Antibacterial Agent: A Mini-Review. J. Bacteriol. Mycol. Open Access 2018, 6, 141–145. [Google Scholar] [CrossRef]
  3. Pereira, I.; Severino, P.; Santos, A.C.; Silva, A.M.; Souto, E.B. Linalool Bioactive Properties and Potential Applicability in Drug Delivery Systems. Colloids Surf. B Biointerfaces 2018, 171, 566–578. [Google Scholar] [CrossRef] [PubMed]
  4. Chaachouay, N.; Zidane, L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. Drugs Drug Candidates 2024, 3, 184–207. [Google Scholar] [CrossRef]
  5. Mercanoglu Taban, B.; Stavropoulou, E.; Kretli Winkelströter, L.; Bezirtzoglou, E. Value-Added Effects of Using Aromatic Plants in Foods and Human Therapy. Food Sci. Technol. 2022, 42, e43121. [Google Scholar] [CrossRef]
  6. Vlaic, R.A.M.; Mureșan, V.; Mureșan, A.E.; Mureșan, C.C.; Tanislav, A.E.; Pușcaș, A.; Petruţ, G.S.M.; Ungur, R.A. Spicy and Aromatic Plants for Meat and Meat Analogues Applications. Plants 2022, 11, 960. [Google Scholar] [CrossRef]
  7. Fierascu, R.C.; Fierascu, I.; Baroi, A.M.; Ortan, A. Selected Aspects Related to Medicinal and Aromatic Plants as Alternative Sources of Bioactive Compounds. Int. J. Mol. Sci. 2021, 22, 1521. [Google Scholar] [CrossRef]
  8. Markiewicz-Gospodarek, A. Aromatic Plants: Nature’s Fragrant Bounty and Their Diverse Applications. Ann. Clin. Trials Vaccines Res. 2023, 13, 156–157. [Google Scholar] [CrossRef]
  9. Kieliszek, M.; Edris, A.; Kot, A.M.; Piwowarek, K. Biological Activity of Some Aromatic Plants and Their Metabolites, with an Emphasis on Health-Promoting Properties. Molecules 2020, 25, 2478. [Google Scholar] [CrossRef]
  10. Kliszcz, A.; Danel, A.; Puła, J.; Barabasz-Krasny, B.; Możdżeń, K. Fleeting Beauty—The World of Plant Fragrances and Their Application. Molecules 2021, 26, 2473. [Google Scholar] [CrossRef]
  11. Chomchalow, N. Production of Aromatic Plants in Asia: An Overview. AU J. Technol. 2005, 5, 7–21. [Google Scholar]
  12. Costa, D.C.; Costa, H.S.; Albuquerque, T.G.; Ramos, F.; Castilho, M.C.; Sanches-Silva, A. Advances in Phenolic Compounds Analysis of Aromatic Plants and Their Potential Applications. Trends Food Sci. Technol. 2015, 45, 336–354. [Google Scholar] [CrossRef]
  13. Samarth, R.M.; Samarth, M.; Matsumoto, Y. Medicinally Important Aromatic Plants with Radioprotective Activity. Futur. Sci. OA 2017, 3, FSO247. [Google Scholar] [CrossRef] [PubMed]
  14. Christaki, E.; Bonos, E.; Giannenas, I.; Florou-Paneri, P. Aromatic Plants as a Source of Bioactive Compounds. Agriculture 2012, 2, 228–243. [Google Scholar] [CrossRef]
  15. Yang, Z.; Deng, X.; Yang, Z.; Han, M.; Chan, K.W.; Abdull Razis, A.F.; Esa, N.M.; Ho, K.L.; Abu Bakar, M.Z. Polygonum minus: A Tropical Medicinal Herb with Vast Applications in Food, Agricultural, and Medicinal Fields. Food Biosci. 2024, 60, 104511. [Google Scholar] [CrossRef]
  16. Han, M.; Kasim, S.; Yang, Z.; Deng, X.; Uddin, M.K.; Saidi, N.B.; Shuib, E.M. Application of Polygonum minus Extract in Enhancing Drought Tolerance in Maize by Regulating Osmotic and Antioxidant System. Phyton-Int. J. Exp. Bot. 2024, 93, 213–226. [Google Scholar] [CrossRef]
  17. Vikram, P.; Chiruvella, K.K.; Ripain, I.H.A.; Arifullah, M. A Recent Review on Phytochemical Constituents and Medicinal Properties of Kesum (Polygonum minus Huds.). Asian Pac. J. Trop. Biomed. 2014, 4, 430–435. [Google Scholar] [CrossRef]
  18. Baharum, S.N.; Bunawan, H.; Ghani, M.A.; Mustapha, W.A.W.; Noor, N.M. Analysis of the Chemical Composition of the Essential Oil of Polygonum minus Huds. Using Two-Dimensional Gas Chromatography-Time-of-Flight Mass Spectrometry (GC-TOF MS). Molecules 2010, 15, 7006–7015. [Google Scholar] [CrossRef]
  19. Rusdi, N.A.; Goh, H.H.; Baharum, S.N. GC-MS/Olfactometric Characterisation and Aroma Extraction Dilution Analysis of Aroma Active Compounds in Polygonum minus Essential Oil. Plant Omics 2016, 9, 289–294. [Google Scholar] [CrossRef]
  20. Bashir, M.I.; Aziz, N.H.K.A.; Noor, D.A.M. Possible Antidepressant Potential of a Cognitive Enhancer Polygonum minus Based on Its Major Chemical Constituents in Leaf Part. Drug Invent. Today 2020, 13, 549–557. [Google Scholar]
  21. Ahmad, R.; Baharum, S.N.; Bunawan, H.; Lee, M.; Noor, N.M.; Rohani, E.R.; Ilias, N.; Zin, N.M. Volatile Profiling of Aromatic Traditional Medicinal Plant, Polygonum minus in Different Tissues and Its Biological Activities. Molecules 2014, 19, 19220–19242. [Google Scholar] [CrossRef] [PubMed]
  22. Silva, T.d.M.; Miranda, R.R.S.; Ferraz, V.P.; Pereira, M.T.; de Siqueira, E.P.; Alcântara, A.F.C. Changes in the Essential Oil Composition of Leaves of Echinodorus Macrophyllus Exposed to γ-Radiation. Rev. Bras. Farmacogn. 2013, 23, 600–607. [Google Scholar] [CrossRef]
  23. Araque, I.; Ramírez, J.; Vergara, R.; Mella, J.; Aránguiz, P.; Espinoza, L.; Vera, W.; Montenegro, I.; Salas, C.O.; Villena, J.; et al. Cytotoxic Activity, Topoisomerase I Inhibition and In Silico Studies of New Sesquiterpene-Aryl Ester Derivatives of (-) Drimenol. Molecules 2023, 28, 3959. [Google Scholar] [CrossRef]
  24. Vo, N.N.Q.; Nomura, Y.; Kinugasa, K.; Takagi, H.; Takahashi, S. Identification and Characterization of Bifunctional Drimenol Synthases of Marine Bacterial Origin. ACS Chem. Biol. 2022, 17, 1226–1238. [Google Scholar] [CrossRef]
  25. Kreuzenbeck, N.B.; Dhiman, S.; Roman, D.; Burkhardt, I.; Conlon, B.H.; Fricke, J.; Guo, H.; Blume, J.; Görls, H.; Poulsen, M.; et al. Isolation, (Bio)Synthetic Studies and Evaluation of Antimicrobial Properties of Drimenol-Type Sesquiterpenes of Termitomyces Fungi. Commun. Chem. 2023, 6, 79. [Google Scholar] [CrossRef]
  26. Kwon, M.; Cochrane, S.A.; Vederas, J.C.; Ro, D.K. Molecular Cloning and Characterization of Drimenol Synthase from Valerian Plant (Valeriana officinalis). FEBS Lett. 2014, 588, 4597–4603. [Google Scholar] [CrossRef] [PubMed]
  27. Yao, P.; Liu, Y. Terpenoids: Natural Compounds for Non-Alcoholic Fatty Liver Disease (NAFLD) Therapy. Molecules 2023, 28, 272. [Google Scholar] [CrossRef]
  28. Abu-Izneid, T.; Rauf, A.; Shariati, M.A.; Khalil, A.A.; Imran, M.; Rebezov, M.; Uddin, M.S.; Mahomoodally, M.F.; Rengasamy, K.R.R. Sesquiterpenes and Their Derivatives-Natural Anticancer Compounds: An Update. Pharmacol. Res. 2020, 161, 105165. [Google Scholar] [CrossRef] [PubMed]
  29. Shu, H.Z.; Peng, C.; Bu, L.; Guo, L.; Liu, F.; Xiong, L. Bisabolane-Type Sesquiterpenoids: Structural Diversity and Biological Activity. Phytochemistry 2021, 192, 112927. [Google Scholar] [CrossRef]
  30. Philcox, D.; Moore, D.M. Flora of Tierra Del Fuego. Kew Bull. 1984, 39, 848–849. [Google Scholar] [CrossRef]
  31. Appel, H.H.; Brooks, C.J.W.; Overton, K.H. The Constitution and Stereochemistry of Drimenol, a Novel Bicyclic Sesquiterpenoid. J. Chem. Soc. 1959, 673, 3322. [Google Scholar] [CrossRef]
  32. Jansen, B.J.M.; De Groot, A. The Occurrence and Biological Activity of Drimane Sesquiterpenoids. Nat. Prod. Rep. 1991, 8, 309–318. [Google Scholar] [CrossRef]
  33. Riad, N.; Zahi, M.R.; Bouzidi, N.; Daghbouche, Y.; Touafek, O.; El Hattab, M. Occurrence of Marine Ingredients in Fragrance: Update on the State of Knowledge. Chemistry 2021, 3, 1437–1463. [Google Scholar] [CrossRef]
  34. Ahmad, R.; Bunawan, H.; Normah, M.N.; Baharum, S.N. Chemical Composition in Different Tissues of Polygonum minus by Using GC X GC-TOF MS and Direct Discrimination by Multivariate Analysis of Fourier Transform Infrared Spectroscopy Data. Int. J. Pharmacogn. Phytochem. Res. 2016, 8, 1986–1992. [Google Scholar]
  35. Hassim, N.; Markom, M.; Anuar, N.; Dewi, K.H.; Baharum, S.N.; Mohd Noor, N. Antioxidant and Antibacterial Assays on Polygonum minus Extracts: Different Extraction Methods. Int. J. Chem. Eng. 2015, 2015, 826709. [Google Scholar] [CrossRef]
  36. Syaiful, A.J. Harlia Pengaruh Waktu Distilasi Terhadap Komponen Minyak Atsiri Pada Daun Kesum (Polygonum minus Huds). J. Kim. Khatulistiwa 2015, 4, 18–23. [Google Scholar]
  37. Ridzuan, P.M.; Hamzah, H.A.; Shah, A.; Hassan, N.M.; Roesnita, B. Synergistic Effects of Persicaria Odorata (Daun Kesom) Leaf Extracts with Standard Antibiotics on Pathogenic Bacteria. IIUM Med. J. Malays. 2017, 16, 27–32. [Google Scholar] [CrossRef]
  38. Ain, N.; Kiraman, S.; Yusof, H. A Mini Review on the Methods for the Extraction, Isolation, and Determination of P. Odorata’s Bioactive Compounds. Healthscope 2022, 5, 38–44. [Google Scholar]
  39. Chanprapai, P.; Kubo, I.; Chavasiri, W. Anti-Rice Pathogenic Microbial Activity of Persicaria sp. Extracts. Sci. Technol. Asia 2018, 23, 32–41. [Google Scholar] [CrossRef]
  40. Adaeze, B.-C.A.; Amadi, P.U. Evaluation of Bioactive Component, Free Radical Scavenging Potentials and Protein Qualities of Gomphrena Celosoides and Zea Mays Leaves. Curr. Bioact. Compd. 2019, 16, 1108–1115. [Google Scholar] [CrossRef]
  41. Juliani, H.R.; Zygadlo, J.A.; Scrivanti, R.; de la Sota, E.; Simon, J.E. The Essential Oil of Anemia tomentosa (Savigny) Sw. Var. anthriscifolia (Schrad.) Mickel. Flavour Fragr. J. 2004, 19, 541–543. [Google Scholar] [CrossRef]
  42. Eser, N.; Yoldas, A. Identification of Heat-Resistant Chemical Components of Ferula Elaeochytris Root Extracts by Gas Chromatography-Mass Spectrometry. Trop. J. Pharm. Res. 2019, 18, 55–60. [Google Scholar] [CrossRef]
  43. Murray, A.F.; Satooka, H.; Shimizu, K.; Chavasiri, W.; Kubo, I. Polygonum Odoratum Essential Oil Inhibits the Activity of Mushroom Derived Tyrosinase. Heliyon 2019, 5, e02817. [Google Scholar] [CrossRef] [PubMed]
  44. Fujita, K.I.; Chavasiri, W.; Kubo, I. Anti-Salmonella Activity of Volatile Compounds of Vietnam Coriander. Phyther. Res. 2015, 29, 1081–1087. [Google Scholar] [CrossRef]
  45. Tuberoso, C.I.G.; Kowalczyk, A.; Coroneo, V.; Russo, M.T.; Dessì, S.; Cabras, P. Chemical Composition and Antioxidant, Antimicrobial, and Antifungal Activities of the Essential Oil of Achillea Ligustica All. J. Agric. Food Chem. 2005, 53, 10148–10153. [Google Scholar] [CrossRef]
  46. Sharma, R.K.; Kaur, H.; Singh, M.; Kumar, M.; Sharma, R.; Shah, G.C.; Sharma, P. Chemical Composition and Antimicrobial Properties of Essential Oil Anaphalis Triplinervis from Western Himalaya. Chem. Nat. Compd. 2019, 55, 751–753. [Google Scholar] [CrossRef]
  47. Saroglou, V.; Dorizas, N.; Kypriotakis, Z.; Skaltsa, H.D. Analysis of the Essential Oil Composition of Eight Anthemis Species from Greece. Proc. J. Chromatogr. A 2006, 1104, 313–322. [Google Scholar] [CrossRef]
  48. Sharma, V.; Sharma, N.; Pathania, V.; Mishra, T.; Bhatia, A.; Singh, B.; Gupta, R.C. Essential Oil Analysis and Antimicrobial Activity of Artemisia Parviflora from Losar. J. Essent. Oil-Bear. Plants 2011, 14, 309–315. [Google Scholar] [CrossRef]
  49. Liu, X.J.; Li, Y.; Su, S.L.; Wei, D.D.; Yan, H.; Guo, S.; Shang, E.X.; Sun, X.D.; Duan, J.A. Comparative Analysis of Chemical Composition and Antibacterial and Anti-Inflammatory Activities of the Essential Oils from Chrysanthemum Morifolium of Different Flowering Stages and Different Parts. Evid.-Based Complement. Altern. Med. 2022, 2022, 5954963. [Google Scholar] [CrossRef]
  50. Lawal, O.A.; Ogunwande, I.A.; Kasali, A.A.; Opoku, A.R.; Oyedeji, A.O. Chemical Composition, Antibacterial and Cytotoxic Activities of Essential Oil from the Leaves of Helichrysum Odoratissimum Grown in South Africa. J. Essent. Oil-Bear. Plants 2015, 18, 236–241. [Google Scholar] [CrossRef]
  51. Kim, T.H.; Truong, V.L.; Jeong, W.S. Phytochemical Composition and Antioxidant and Anti-Inflammatory Activities of Ligularia Fischeri Turcz: A Comparison between Leaf and Root Extracts. Plants 2022, 11, 3005. [Google Scholar] [CrossRef]
  52. Ying, B.P.; Peiser, G.; Ji, Y.Y.; Mathias, K.; Tutko, D.; Hwang, Y.S. Phytotoxic Sesquiterpenoids from Canella winterana. Phytochemistry 1995, 38, 909–915. [Google Scholar] [CrossRef]
  53. Pino, J.A.; Rosado, A.; Fuentes, V. Leaf Oil of Canella winterana (L.) Gaertn. From Cuba. J. Essent. Oil Res. 1998, 10, 311–312. [Google Scholar] [CrossRef]
  54. Torres, E.; Wisniewski, A.; Simionatto, E.L. Composição Química Dos Componentes Voláteis de Capsicodendron dinisii Schwancke (Canellaceae). Quim. Nova 2010, 33, 130–132. [Google Scholar] [CrossRef]
  55. Andrade, M.A.; Cardoso, M.D.G.; De Andrade, J.; Silva, L.F.; Teixeira, M.L.; Resende, J.M.V.; Figueiredo, A.C.D.S.; Barroso, J.G. Chemical Composition and Antioxidant Activity of Essential Oils from Capsicodendron dinisii Schwacke and Siparuna guianensis Aublet. Antioxidants 2013, 2, 384–397. [Google Scholar] [CrossRef]
  56. Lawal, O.A.; Ogunwande, I.A.; Opoku, A.R.; Kasali, A.A.; Oyedeji, A.O. Chemical Composition and Antibacterial Activities of Essential Oil of Warburgia salutaris (Bertol. f.) Chiov. from South Africa. J. Biol. Act. Prod. Nat. 2014, 4, 272–277. [Google Scholar] [CrossRef]
  57. Maroyi, A. The Genus Warburgia: A Review of Its Traditional Uses and Pharmacology. Pharm. Biol. 2014, 52, 378–391. [Google Scholar] [CrossRef] [PubMed]
  58. Dharani, N. A Review of Phytochemical Constituents and Phar- Macological Activities of Ethnomedicinal Warburgia ugandensis Sprague Ssp. Ugandensis in East Africa. Pharm. J. Kenya 2020, 24, 67–71. [Google Scholar]
  59. Leonard, C.M.; Viljoen, A.M. Warburgia: A Comprehensive Review of the Botany, Traditional Uses and Phytochemistry. J. Ethnopharmacol. 2015, 165, 260–285. [Google Scholar] [CrossRef] [PubMed]
  60. Bos, R.; Woerdenbag, H.J.; Hendriks, H.; Scheffer, J.J.C. Composition of the Essential Oils from Underground Parts of Valeriana officinalis L. s.l. and Several Closely Related Taxa. Flavour Fragr. J. 1997, 12, 359–370. [Google Scholar] [CrossRef]
  61. Sati, S.; Mathela, C.S. Essential Oil Composition of Valeriana hardwickii Var. Arnottiana from the Himalayas. Flavour Fragr. J. 2005, 20, 299–301. [Google Scholar] [CrossRef]
  62. Gränicher, F.; Christen, P.; Kapetanidis, I. Essential Oils from Normal and Hairy Roots of Valeriana officinalis Var. Sambucifolia. Phytochemistry 1995, 40, 1421–1424. [Google Scholar] [CrossRef]
  63. Samaneh, E.T.; Tayebeh, R.; Hassan, E.; Niknam, V. Composition of Essential Oils in Subterranean Organs of Three Species of Valeriana L. Nat. Prod. Res. 2010, 24, 1834–1842. [Google Scholar] [CrossRef]
  64. Demetzos, C.; Angelopoulou, D.; Perdetzoglou, D. A Comparative Study of the Essential Oils of Cistus Salviifolius in Several Populations of Crete (Greece). Biochem. Syst. Ecol. 2002, 30, 651–665. [Google Scholar] [CrossRef]
  65. Rguez, S.; Djébali, N.; Ben Slimene, I.; Abid, G.; Hammemi, M.; Chenenaoui, S.; Bachkouel, S.; Daami-Remadi, M.; Ksouri, R.; Hamrouni-Sellami, I. Cupressus Sempervirens Essential Oils and Their Major Compounds Successfully Control Postharvest Grey Mould Disease of Tomato. Ind. Crops Prod. 2018, 123, 135–141. [Google Scholar] [CrossRef]
  66. Liu, H.; Huang, J.; Yang, S.; Li, J.; Zhou, L. Chemical Composition, Algicidal, Antimicrobial, and Antioxidant Activities of the Essential Oils of Taiwania Flousiana Gaussen. Molecules 2020, 25, 967. [Google Scholar] [CrossRef]
  67. Nassar, M.I.; Yassine, Y.M.; Elshamy, A.I.; El-Beih, A.A.; El-Shazly, M.; Singab, A.N.B. Essential Oil and Antimicrobial Activity of Aerial Parts of Cyperus leavigatus L. (Family: Cyperaceae). J. Essent. Oil-Bear. Plants 2015, 18, 416–422. [Google Scholar] [CrossRef]
  68. Samejo, M.Q.; Memon, S.; Bhanger, M.I.; Khan, K.M. Chemical Composition of Essential Oils from Alhagi maurorum. Chem. Nat. Compd. 2012, 48, 898–900. [Google Scholar] [CrossRef]
  69. Loizzo, M.R.; Rashed, K.; Said, A.; Bonesi, M.; Menichini, F.; Tundis, R. Antiproliferative and Antioxidant Properties of Alhagi maurorum Boiss (Leguminosae) Aerial Parts. Ind. Crops Prod. 2014, 53, 289–295. [Google Scholar] [CrossRef]
  70. Al-Qudah, M.A.; Al-Ghoul, A.M.; Trawenh, I.N.; Al-Jaber, H.I.; Al Shboul, T.M.; Abu Zarga, M.H.; Abu orabi, S.T. Antioxidant Activity and Chemical Composition of Essential Oils from Jordanian Ononis natrix L. and Ononis sicula Guss. J. Biol. Act. Prod. Nat. 2014, 4, 52–61. [Google Scholar] [CrossRef]
  71. Aboaba, S.A.; Ogunwande, I.A.; Walker, T.M.; Setzer, W.N.; Oladosu, I.A.; Ekundayo, O. Essential Oil Composition, Antibacterial Activity and Toxicity of the Leaves of Tetrapleura tetraptera (Schum. & Thonn.) Taubert from Nigeria. Nat. Prod. Commun. 2009, 4, 287–290. [Google Scholar] [CrossRef]
  72. Kraut, L.; Mues, R.; Sim-Sim, M. Sesquiterpene Lactones and 3-Benzylphthalides from Frullania Muscicola. Phytochemistry 1994, 37, 1337–1346. [Google Scholar] [CrossRef]
  73. Öǧütçü, H.; Sökmen, A.; Sökmen, M.; Polissiou, M.; Serkedjieva, J.; Daferera, D.; Şahin, F.; Bariş, Ö.; Güllüce, M. Bioactivities of the Various Extracts and Essential Oils of Salvia Limbata C.A.Mey. and Salvia sclarea L. Turk. J. Biol. 2008, 32, 181–192. [Google Scholar]
  74. Karimov, A.M.; Drenin, A.A.; Vasina, N.A.; Botirov, E.K. GC-MS Study of Nonpolar Constituents from Scutellaria Comosa. Chem. Nat. Compd. 2015, 51, 1188–1190. [Google Scholar] [CrossRef]
  75. Komaitis, M.E.; Falirea, A.; Voudouris, E.C. Constituents of the Essential Oil of Sideritis Cretica Boiss. J. Sci. Food Agric. 1985, 36, 970–972. [Google Scholar] [CrossRef]
  76. Mujawah, A.A.H.; Abdallah, E.M.; Alshoumar, S.A.; Alfarraj, M.I.; Alajel, S.M.I.; Alharbi, A.L.; Alsalman, S.A.; Alhumaydhi, F.A. GC-MS and in Vitro Antibacterial Potential of Cinnamomum Camphora Essential Oil against Some Clinical Antibiotic-Resistant Bacterial Isolates. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 5372–5379. [Google Scholar] [CrossRef]
  77. Toyota, M.; Asakawa, Y. Sesquiterpenoids from the Liverwort Bazzania Fauriana. Phytochemistry 1988, 27, 2155–2159. [Google Scholar] [CrossRef]
  78. Rakotosaona, R.; Nicoletti, M.; Papa, F.; Randrianarivo, E.; Rasoanaivo, P.; Maggi, F. Chemical Composition of the Essential Oil of Kaliphora madagascariensis Hook. F. Nat. Prod. Res. 2016, 30, 960–966. [Google Scholar] [CrossRef]
  79. Ogunwande, I.A.; Flamini, G.; Adefuye, A.E.; Lawal, N.O.; Moradeyo, S.; Avoseh, N.O. Chemical Compositions of Casuarina equisetifolia L., Eucalyptus toreliana L. and Ficus elastica Roxb. Ex Hornem Cultivated in Nigeria. S. Afr. J. Bot. 2011, 77, 645–649. [Google Scholar] [CrossRef]
  80. Ogunwande, I.A.; Sonibare, M.A.; Thang, T.D.; Dung, N.X.; Soladoye, M.O.; Morohunfolu, O.O. Comparative Analysis of the Oils of Three Ficus Species from Nigeria. J. Essent. Oil Res. 2008, 20, 386–389. [Google Scholar] [CrossRef]
  81. Costa, M.D.S.; Araújo, N.J.S.; de Freitas, T.S.; da Cunha, F.A.B.; Do Amaral, W.; Deschamps, C.; Confortin, C.; da Silva, L.E.; Coutinho, H.D.M. GC-FID Analysis and Antibacterial Activity of the Calyptranthes Concinna Essential Oil against MDR Bacterial Strains. Separations 2020, 7, 10. [Google Scholar] [CrossRef]
  82. Sabiu, S.; Ajani, E.O.; Sunmonu, T.O.; Balogun, F.O.; Ashafa, A.O.T.; Othman, R.B.; Olowa, S.K. Mechanism of Hepatoprotective Potential of Aqueous Leaves Extract of Eucalyptus obliqua (Myrtaceae) in Carbon Tetrachloride Intoxicated Wistar Rats. J. Appl. Pharm. Sci. 2017, 7, 183–190. [Google Scholar] [CrossRef]
  83. El-Ghorab, A.H.; Fadel, H.M.; El-Massry, K.F. The Egyptian Eucalyptus Camaldulensis Var. Brevirostris: Chemical Compositions of the Fruit Volatile Oil and Antioxidant Activity. Flavour Fragr. J. 2002, 17, 306–312. [Google Scholar] [CrossRef]
  84. Sabiu, S.; Ashafa, A.O.T. Membrane Stabilization and Kinetics of Carbohydrate Metabolizing Enzymes (α-Amylase and α-Glucosidase) Inhibitory Potentials of Eucalyptus obliqua L.Her. (Myrtaceae) Blakely Ethanolic Leaf Extract: An In Vitro Assessment. S. Afr. J. Bot. 2016, 105, 264–269. [Google Scholar] [CrossRef]
  85. Sousa, R.M.F.; de Morais, S.A.L.; Vieira, R.B.K.; Napolitano, D.R.; Guzman, V.B.; Moraes, T.S.; Cunha, L.C.S.; Martins, C.H.G.; Chang, R.; de Aquino, F.J.T.; et al. Chemical Composition, Cytotoxic, and Antibacterial Activity of the Essential Oil from Eugenia Calycina Cambess. Leaves against Oral Bacteria. Ind. Crops Prod. 2015, 65, 71–78. [Google Scholar] [CrossRef]
  86. Zoghbi, M.G.B.; Guilhon, G.M.S.P.; Sarges, F.N.; Pereira, R.A.; Oliveira, J. Chemical Variability of the Volatiles from the Leaves of Eugenia protenta McVaugh (Myrtaceae) Growing Wild in the North of Brazil. Biochem. Syst. Ecol. 2011, 39, 660–665. [Google Scholar] [CrossRef]
  87. Youssef, F.S.; Labib, R.M.; Gad, H.A.; Eid, S.; Ashour, M.L.; Eid, H.H. Pimenta Dioica and Pimenta Racemosa: GC-Based Metabolomics for the Assessment of Seasonal and Organ Variation in Their Volatile Components, in Silico and in Vitro Cytotoxic Activity Estimation. Food Funct. 2021, 12, 5247–5259. [Google Scholar] [CrossRef]
  88. 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]
  89. Samejo, M.Q.; Memon, S.; Bhanger, M.I.; Khan, K.M. Essential Oil Constituents in Fruit and Stem of Calligonum polygonoides. Ind. Crops Prod. 2013, 45, 293–295. [Google Scholar] [CrossRef]
  90. Samejo, M.Q.; Memon, S.; Bhanger, M.I.; Khan, K.M. Chemical Composition of Essential Oil from Calligonum polygonoides Linn. Nat. Prod. Res. 2013, 27, 619–623. [Google Scholar] [CrossRef]
  91. Ahmad, S.; Akram, M. Antifungal Activity in the Methanolic, Aqueous and Hexane Extracts of Calligonum Polygonoides. Int. J. Immunopathol. Pharmacol. 2019, 33, 2058738418821275. [Google Scholar] [CrossRef]
  92. Prota, N.; Mumm, R.; Bouwmeester, H.J.; Jongsma, M.A. Comparison of the Chemical Composition of Three Species of Smartweed (Genus Persicaria) with a Focus on Drimane Sesquiterpenoids. Phytochemistry 2014, 108, 129–136. [Google Scholar] [CrossRef]
  93. Derita, M.; Zacchino, S. Chemotaxonomic Importance of Sesquiterpenes and Flavonoids in Five Argentinian Species of Polygonum Genus. J. Essent. Oil Res. 2011, 23, 11–14. [Google Scholar] [CrossRef]
  94. Fukuyama, Y.; Sato, T.; Asakawa, Y.; Takemoto, T. A Potent Cytotoxic Warburganal and Related Drimane-Type Sesquiterpenoids from Polygonum hydropiper. Phytochemistry 1980, 21, 2895–2898. [Google Scholar] [CrossRef]
  95. Kong, Y.D.; Qi, Y.; Cui, N.; Zhang, Z.H.; Wei, N.; Wang, C.F.; Zeng, Y.N.; Sun, Y.P.; Kuang, H.X.; Wang, Q.H. The Traditional Herb Polygonum Hydropiper from China: A Comprehensive Review on Phytochemistry, Pharmacological Activities and Applications. Pharm. Biol. 2023, 61, 799–814. [Google Scholar] [CrossRef]
  96. Ayaz, M.; Junaid, M.; Ullah, F.; Sadiq, A.; Shahid, M.; Ahmad, W.; Ullah, I.; Ahmad, A.; Syed, N.i.H. GC-MS Analysis and Gastroprotective Evaluations of Crude Extracts, Isolated Saponins, and Essential Oil from Polygonum hydropiper L. Front. Chem. 2017, 5, 58. [Google Scholar] [CrossRef] [PubMed]
  97. Miyazawa, M.; Tamura, N. Components of the Essential Oil from Sprouts of Polygonum hydropiper L. (‘Benitade’). Flavour Fragr. J. 2007, 22, 188–190. [Google Scholar] [CrossRef]
  98. Potter, T.L.; Fagerson, I.S.; Craker, L.E. Composition of Vietnamese Coriander Leaf Oil. Acta Hortic. 1993, 34, 305–311. [Google Scholar] [CrossRef]
  99. Zeng, Y.X.; Zhao, C.X.; Liang, Y.Z.; Yang, H.; Fang, H.Z.; Yi, L.Z.; Zeng, Z. Da Comparative Analysis of Volatile Components from Clematis Species Growing in China. Anal. Chim. Acta 2007, 595, 328–339. [Google Scholar] [CrossRef]
  100. Andonova, T.; Dimitrova-Dyulgerova, I.; Slavov, I.; Muhovski, Y.; Stoyanova, A. A Comparative Study of Koelreuteria Paniculata Laxm. Aerial Parts Essential Oil Composition. J. Essent. Oil-Bear. Plants 2020, 23, 1363–1370. [Google Scholar] [CrossRef]
  101. Toyota, M.; Ooiso, Y.; Kusuyama, T.; Asakawa, Y. Drimane-Type Sesquiterpenoids from the Liverwort Diplophyllum serrulatum. Phytochemistry 1994, 35, 1263–1265. [Google Scholar] [CrossRef]
  102. Asakawa, Y.; Toyota, M.; Cheminat, A. Terpenoids from the French Liverwort Targionia hypophylla. Phytochemistry 1986, 25, 2555–2556. [Google Scholar] [CrossRef]
  103. Essien, E.E.; Walker, T.M.; Newby, J.S.; Setzer, W.N.; Ekundayo, O. Characterization and Antimicrobial Activity of Essential Oils of Characterization and Antimicrobial Activity of Essential Oils of Recommended Citation Recommended Citation. J. Med. Act. Plants 2017, 5, 47–52. [Google Scholar]
  104. Limberger, R.P.; Scopel, M.; Sobral, M.; Henriques, A.T. Comparative Analysis of Volatiles from Drimys brasiliensis Miers and D. angustifolia Miers (Winteraceae) from Southern Brazil. Biochem. Syst. Ecol. 2007, 35, 130–137. [Google Scholar] [CrossRef]
  105. Gomes, M.R.F.; Schuh, R.S.; Jacques, A.L.B.; Augustin, O.A.; Bordignon, S.A.L.; Dias, D.O.; Kelmann, R.G.; Koester, L.S.; Gehring, M.P.; Morrone, F.B.; et al. Citotoxic Activity Evaluation of Essential Oils and Nanoemulsions of Drimys angustifolia and D. Brasiliensis on Human Glioblastoma (U-138 MG) and Human Bladder Carcinoma (T24) Cell Lines in Vitro. Rev. Bras. Farmacogn. 2013, 23, 259–267. [Google Scholar] [CrossRef]
  106. Santos, T.G.; Dognini, J.; Begnini, I.M.; Rebelo, R.A.; Verdi, M.; De Gasper, A.L.; Dalmarco, E.M. Chemical Characterization of Essential Oils from Drimys angustifolia Miers (Winteraceae) and Antibacterial Activity of Their Major Compounds. J. Braz. Chem. Soc. 2013, 24, 164–170. [Google Scholar] [CrossRef]
  107. Ribeiro, V.L.S.; Rolim, V.; Bordignon, S.; Henriques, A.T.; Dorneles, G.G.; Limberger, R.P.; Von Poser, G. Chemical Composition and Larvicidal Properties of the Essential Oils from Drimys brasiliensis Miers (Winteraceae) on the Cattle Tick Rhipicephalus (Boophilus) Microplus and the Brown Dog Tick Rhipicephalus sanguineus. Parasitol. Res. 2008, 102, 531–535. [Google Scholar] [CrossRef]
  108. Lago, J.H.G.; Carvalho, L.A.C.; Da Silva, F.S.; Toyama, D.D.O.; Fávero, O.A.; Romoff, P. Chemical Composition and Anti-Inflammatory Evaluation of Essential Oils from Leaves and Stem Barks from Drimys brasiliensis Miers (Winteraceae). J. Braz. Chem. Soc. 2010, 21, 1760–1765. [Google Scholar] [CrossRef]
  109. Zem, L.M.; Zuffellato-Ribas, K.C.; Koehler, H.S.; Radomski, M.I.; Deschamps, C. Drimys brasiliensis Essential Oil as a Source of Drimenol. HOLOS 2016, 2, 68. [Google Scholar] [CrossRef]
  110. Cicció, J.F. Essential Oils of Drimys granadensis (Interaceae) Leaves and Green Fruits. Rev. Biol. Trop. 1997, 44, 29–33. [Google Scholar]
  111. Cicció, J.F. Aceites Esenciales de Las Hojas y de Los Frutos Verdes de Drimys granadensis (Winteraceae). Rev. Biol. Trop. 1995, 44, 29–33. [Google Scholar]
  112. Gaviria, M.; Quijano, C.; Pino, J.; Madriñán, S. Chemical Composition and Antibacterial Activity of the Essential Oil of Drimys granadensis L.f. Leaves from Colombia. Chem. Biodivers. 2011, 8, 532–539. [Google Scholar] [CrossRef] [PubMed]
  113. Sierra, J.R.; López, J.T.; Cortés, M.J. (-)-3β-Acetoxydrimenin from the Leaves of Drimys Winteri. Phytochemistry 1985, 25, 253–254. [Google Scholar] [CrossRef]
  114. Paz, C.; Cárcamo, G.; Silva, M.; Becerra, J.; Urrutia, H.; Sossa, K. Drimendiol, a Drimane Sesquiterpene with Quorum Sensing Inhibition Activity. Nat. Prod. Commun. 2013, 8, 147–148. [Google Scholar] [CrossRef]
  115. Negri, L.M.; Spontón, E.A.; Salgado, O.; Sancho, A.; Denoya, G.I. Effect of Different Drying Conditions on Volatile Compounds of Canelo Pepper (Drimys Winteri). J. Phytol. 2020, 12, 56–61. [Google Scholar] [CrossRef]
  116. Barrero, A.F.; Herrador, M.M.; Arteaga, P.; Lara, A.; Cortés, M. Chemical Composition of the Essential Oil from Drimys winteri Forst. Wood. J. Essent. Oil Res. 2000, 12, 685–688. [Google Scholar] [CrossRef]
  117. Céspedes, C.L.; Marín, J.C.; Domínguez, M.; Avila, J.G.; Serrato, B. Plant Growth Inhibitory Activities by Secondary Metabolites Isolated from Latin American Flora. Adv. Phytomed. 2006, 2, 373–410. [Google Scholar] [CrossRef]
  118. Muñoz, O.; Christen, P.; Cretton, S.; Barrero, A.F.; Lara, A.; Herrador, M.M. Comparison of the Essential Oils of Leaves and Stem Bark from Two Different Populations of Drimys winteri a Chilean Herbal Medicine. Nat. Prod. Commun. 2011, 6, 879–882. [Google Scholar] [CrossRef]
  119. Brown, G.D. Drimendiol, a Sesquiterpene from Drymis winterii. Phytochemistry 1994, 35, 975–977. [Google Scholar] [CrossRef]
  120. Paul, A.; Bondhon, T.A.; Afrose, S.; Hasan, A.; Jannat, K.; Paul, A.; Jahan, R.; Rahmatullah, M. In Silico Binding Studies with Compounds Present in Essential Oil of Tasmannia Lanceolata Leaves to 3C-like Protease of SARS-CoV-2. J. Med. Plants Stud. 2020, 8, 108–116. [Google Scholar] [CrossRef]
  121. Sethi, S.; Prakash, O.; Pant, A.K.; Kumar, M. Phytochemical Analysis and Pharmacological Activities of Methanolic Extract and Essential Oil from Rhizomes of Alpinia malaccensis (Burm. f.) Roscoe. J. Essent. Oil-Bear. Plants 2017, 20, 1018–1029. [Google Scholar] [CrossRef]
  122. Iweala, E.J.; Uche, M.E.; Dike, E.D.; Etumnu, L.R.; Dokunmu, T.M.; Oluwapelumi, A.E.; Okoro, B.C.; Dania, O.E.; Adebayo, A.H.; Ugbogu, E.A. Curcuma longa (Turmeric): Ethnomedicinal Uses, Phytochemistry, Pharmacological Activities and Toxicity Profiles—A Review. Pharmacol. Res.-Mod. Chin. Med. 2023, 6, 100222. [Google Scholar] [CrossRef]
  123. Al-Dhahli, A.S.; Al-Hassani, F.A.; Mohammed Alarjani, K.; Mohamed Yehia, H.; Al Lawati, W.M.; Najmul Hejaz Azmi, S.; Alam Khan, S. Essential Oil from the Rhizomes of the Saudi and Chinese Zingiber officinale Cultivars: Comparison of Chemical Composition, Antibacterial and Molecular Docking Studies. J. King Saud Univ.-Sci. 2020, 32, 3343–3350. [Google Scholar] [CrossRef]
  124. Premram, A.S.; Parki, A.; Chaubey, P.; Prakash, O.; Kumar, R.; Punetha, H.; Pant, A.K. Phytochemical Diversity Among Parts of Zingiber roseum Rosc. Extracts With Their Antioxidant and Antifungal Activity. J. Biol. Act. Prod. Nat. 2018, 8, 255–264. [Google Scholar] [CrossRef]
  125. Russo, A.; Cardile, V.; Graziano, A.C.E.; Avola, R.; Montenegro, I.; Cuellar, M.; Villena, J.; Madrid, A. Antigrowth Activity and Induction of Apoptosis in Human Melanoma Cells by Drymis winteri Forst Extract and Its Active Components. Chem. Biol. Interact. 2019, 305, 79–85. [Google Scholar] [CrossRef]
  126. Montenegro, I.; Madrid, A.; Cuellar, M.; Seeger, M.; Alfaro, J.F.; Besoain, X.; Martínez, J.P.; Ramirez, I.; Olguín, Y.; Valenzuela, M. Biopesticide Activity from Drimanic Compounds to Control Tomato Pathogens. Molecules 2018, 23, 2053. [Google Scholar] [CrossRef]
  127. Derita, M.; Montenegro, I.; Garibotto, F.; Enriz, R.D.; Fritis, M.C.; Zacchino, S.A. Structural Requirements for the Antifungal Activities of Natural Drimane Sesquiterpenes and Analogues, Supported by Conformational and Electronic Studies. Molecules 2013, 18, 2029–2051. [Google Scholar] [CrossRef]
  128. Montenegro, I.; Pino, L.; Werner, E.; Madrid, A.; Espinoza, L.; Moreno, L.; Villena, J.; Cuellar, M. Comparative Study on the Larvicidal Activity of Drimane Sesquiterpenes and Nordrimane Compounds against Drosophila Melanogaster Til-Til. Molecules 2013, 18, 4192–4208. [Google Scholar] [CrossRef]
  129. Montenegro, I.; Tomasoni, G.; Bosio, C.; Quiñones, N.; Madrid, A.; Carrasco, H.; Olea, A.; Martinez, R.; Cuellar, M.; Villena, J. Study on the Cytotoxic Activity of Drimane Sesquiterpenes and Nordrimane Compounds against Cancer Cell Lines. Molecules 2014, 19, 18993–19006. [Google Scholar] [CrossRef]
  130. Pereira-Torres, D.; Gonçalves, A.T.; Ulloa, V.; Martínez, R.; Carrasco, H.; Olea, A.F.; Espinoza, L.; Gallardo-Escárate, C.; Astuya, A. In Vitro Modulation of Drimys winteri Bark Extract and the Active Compound Polygodial on Salmo salar Immune Genes after Exposure to Saprolegnia parasitica. Fish Shellfish Immunol. 2016, 59, 103–108. [Google Scholar] [CrossRef]
  131. Monsálvez, M.; Zapata, N.; Vargas, M.; Berti, M.; Bittner, M.; Hernández, V. Antifungal Effects of N-Hexane Extract and Essential Oil of Drimys winteri Bark against Take-All Disease. Ind. Crops Prod. 2010, 31, 239–244. [Google Scholar] [CrossRef]
  132. Paz, C.; Burgos, V.; Iturra, A.; Rebolledo, R.; Ortiz, L.; Baggio, R.; Becerra, J.; Cespedes-Acuña, C.L. Assessment of Insecticidal Responses of Extracts and Compounds of Drimys Winteri, Lobelia Tupa, Viola Portalesia and Vestia Foetida against the Granary Weevil Sitophilus Granarius. Ind. Crops Prod. 2018, 122, 232–238. [Google Scholar] [CrossRef]
  133. Paz, C.; Viscardi, S.; Iturra, A.; Marin, V.; Miranda, F.; Barra, P.J.; Mendez, I.; Duran, P. Antifungal Effects of Drimane Sesquiterpenoids Isolated from Drimys winteri against Gaeumannomyces graminis Var. Tritici. Appl. Environ. Microbiol. 2020, 86, e01834-20. [Google Scholar] [CrossRef] [PubMed]
  134. Scher, J.M.; Speakman, J.B.; Zapp, J.; Becker, H. Bioactivity Guided Isolation of Antifungal Compounds from the Liverwort Bazzania trilobata (L.) S.F. Gray. Phytochemistry 2004, 65, 2583–2588. [Google Scholar] [CrossRef] [PubMed]
  135. Derita, M.G.; Leiva, M.L.; Zacchino, S.A. Influence of Plant Part, Season of Collection and Content of the Main Active Constituent, on the Antifungal Properties of Polygonum acuminatum Kunth. J. Ethnopharmacol. 2009, 124, 377–383. [Google Scholar] [CrossRef] [PubMed]
  136. Rai, S.; Acharya-Siwakoti, E.; Kafle, A.; Devkota, H.P.; Bhattarai, A. Plant-Derived Saponins: A Review of Their Surfactant Properties and Applications. Sci 2021, 3, 44. [Google Scholar] [CrossRef]
  137. Rodríguez, B.; Zapata, N.; Medina, P.; Viñuela, E. A Complete 1H and 13C NMR Data Assignment for Four Drimane Sesquiterpenoids Isolated from Drimys winterii. Magn. Reson. Chem. 2005, 43, 82–84. [Google Scholar] [CrossRef]
  138. Henquet, M.G.L.; Prota, N.; van der Hooft, J.J.J.; Varbanova-Herde, M.; Hulzink, R.J.M.; de Vos, M.; Prins, M.; de Both, M.T.J.; Franssen, M.C.R.; Bouwmeester, H.; et al. Identification of a Drimenol Synthase and Drimenol Oxidase from Persicaria hydropiper, Involved in the Biosynthesis of Insect Deterrent Drimanes. Plant J. 2017, 90, 1052–1063. [Google Scholar] [CrossRef]
  139. Aricu, A. (-)-Drimenol—A Valuable Starting Compound for the Synthesis of Drimanic Sesquiterpenoids. Chem. J. Mold. 2009, 4, 14–23. [Google Scholar] [CrossRef]
  140. Akita, H.; Nozawa, M.; Mitsuda, A.; Ohsawa, H. A Convenient Synthesis of (+)-Albicanol Based on Enzymatic Function: Total Syntheses of (+)-Albicanyl Acetate, (−)-Albicanyl 3,4-Dihydroxycinnamate, (−)-Drimenol, (−)-Drimenin and (−)-Ambrox. Tetrahedron Asymmetry 2000, 11, 1375–1388. [Google Scholar] [CrossRef]
  141. Kuchkova, K.I.; Aricu, A.N.; Dragalin, I.P.; Vlad, P.F. Convenient Synthesis of Drimenol and Its Oxidation with Selenium Dioxide. Russ. Chem. Bull. 2004, 53, 2862–2865. [Google Scholar] [CrossRef]
  142. Rihak, K.J.; Bissember, A.C.; Smith, J.A. Polygodial: A Viable Natural Product Scaffold for the Rapid Synthesis of Novel Polycyclic Pyrrole and Pyrrolidine Derivatives. Tetrahedron 2018, 74, 1167–1174. [Google Scholar] [CrossRef]
  143. Just, J.; Jordan, T.B.; Paull, B.; Bissember, A.C.; Smith, J.A. Practical Isolation of Polygodial from Tasmannia Lanceolata: A Viable Scaffold for Synthesis. Org. Biomol. Chem. 2015, 13, 11200–11207. [Google Scholar] [CrossRef]
  144. Waage, J.K.; Mumford, J.D. Agricultural Biosecurity. Philos. Trans. R. Soc. B Biol. Sci. 2008, 363, 863–876. [Google Scholar] [CrossRef]
  145. Pandey, A.K.; Samota, M.K.; Kumar, A.; Silva, A.S.; Dubey, N.K. Fungal Mycotoxins in Food Commodities: Present Status and Future Concerns. Front. Sustain. Food Syst. 2023, 7, 1162595. [Google Scholar] [CrossRef]
  146. Fones, H.N.; Bebber, D.P.; Chaloner, T.M.; Kay, W.T.; Steinberg, G.; Gurr, S.J. Threats to Global Food Security from Emerging Fungal and Oomycete Crop Pathogens. Nat. Food 2020, 1, 332–342. [Google Scholar] [CrossRef] [PubMed]
  147. Chiu, T.; Poucet, T.; Li, Y. The Potential of Plant Proteins as Antifungal Agents for Agricultural Applications. Synth. Syst. Biotechnol. 2022, 7, 1075–1083. [Google Scholar] [CrossRef]
  148. Zaker, M. Natural Plant Products as Eco-Friendly Fungicides for Plant Diseases Control—A Review. Agriculturists 2016, 14, 134. [Google Scholar] [CrossRef]
  149. Robles-Kelly, C.; Rubio, J.; Thomas, M.; Sedán, C.; Martinez, R.; Olea, A.F.; Carrasco, H.; Taborga, L.; Silva-Moreno, E. Effect of Drimenol and Synthetic Derivatives on Growth and Germination of Botrytis cinerea: Evaluation of Possible Mechanism of Action. Pestic. Biochem. Physiol. 2017, 141, 50–56. [Google Scholar] [CrossRef]
  150. Mendoza, L.; Espinoza, P.; Urzua, A.; Vivanco, M.; Cotoras, M. In Vitro Antifungal Activity of the Diterpenoid 7α-Hydroxy-8(17)-Labden-15-Oic Acid and Its Derivatives against Botrytis cinerea. Molecules 2009, 14, 1966–1979. [Google Scholar] [CrossRef]
  151. Jiménez-Reyes, M.F.; Carrasco, H.; Olea, A.F.; Silva-Moreno, E. Natural Compounds: A Sustainable Alternative to the Phytopathogens Control. J. Chil. Chem. Soc. 2019, 64, 4459–4465. [Google Scholar] [CrossRef]
  152. Prasad, W.; Wani, A.D.; Khamrui, K.; Hussain, S.A.; Khetra, Y. Green Solvents, Potential Alternatives for Petroleum Based Products in Food Processing Industries. Clean. Chem. Eng. 2022, 3, 100052. [Google Scholar] [CrossRef]
  153. Jiménez-Reyes, M.F.; Carrasco, H.; Olea, A.; Silva-Moreno, E. Natural Compounds: A Sustainable Alternative for Controlling Phytopathogens. PeerJ Prepr. 2018, 6, e26664v1. [Google Scholar]
  154. Ludwiczuk, A.; Asakawa, Y. Bryophytes as a Source of Bioactive Volatile Terpenoids—A Review. Food Chem. Toxicol. 2019, 132, 110649. [Google Scholar] [CrossRef] [PubMed]
  155. Asakawa, Y.; Ludwiczuk, A.; Nagashima, F. Phytochemical and Biological Studies of Bryophytes. Phytochemistry 2013, 91, 52–80. [Google Scholar] [CrossRef] [PubMed]
  156. Asakawa, Y.; Ludwiczuk, A.; Nagashima, F. Biologically Active Compounds of the Marchantiophyta and Bryophyta; Springer: Berlin/Heidelberg, Germany, 2013. [Google Scholar]
  157. Asakawa, Y.; Ludwiczuk, A. Chemical Constituents of Bryophytes: Structures and Biological Activity. J. Nat. Prod. 2018, 81, 641–660. [Google Scholar] [CrossRef]
  158. Chen, F.; Ludwiczuk, A.; Wei, G.; Chen, X.; Crandall-Stotler, B.; Bowman, J.L. Terpenoid Secondary Metabolites in Bryophytes: Chemical Diversity, Biosynthesis and Biological Functions. CRC Crit. Rev. Plant Sci. 2018, 37, 210–231. [Google Scholar] [CrossRef]
  159. Aqil, F.; Zahin, M.; Ahmad, I.; Owais, M.; Khan, M.S.A.; Bansal, S.S.; Farooq, S. Antifungal Activity of Medicinal Plant Extracts and Phytocompounds: A Review; Springer: Berlin/Heidelberg, Germany, 2010; ISBN 978-3-642-12173-9. [Google Scholar]
  160. Commisso, M.; Guarino, F.; Marchi, L.; Muto, A.; Piro, A.; Degola, F. Bryo-Activities: A Review on How Bryophytes Are Contributing to the Arsenal of Natural Bioactive Compounds against Fungi. Plants 2021, 10, 203. [Google Scholar] [CrossRef] [PubMed]
  161. Edouarzin, E.; Horn, C.; Paudyal, A.; Zhang, C.; Lu, J.; Tong, Z.; Giaever, G.; Nislow, C.; Veerapandian, R.; Hua, D.H.; et al. Broad-Spectrum Antifungal Activities and Mechanism of Drimane Sesquiterpenoids. Microb. Cell 2020, 7, 146–159. [Google Scholar] [CrossRef]
  162. Anyamele, T.G.; Onwuegbuchu, P.N.; Ugbogu, E.A.; Ibe, C. Phytochemical Composition, Bioactive Properties, and Toxicological Profile of Tetrapleura tetraptera. Bioorg. Chem. 2023, 131, 106288. [Google Scholar] [CrossRef]
  163. Pripdeevech, P.; Tanapichatsakul, C.; Chimthai, S. Antagonistic Effect of Endophytic Fungal, Fusarium sp. MFLUCC16-1462, Against Magnaporthe Oryzae. Thai J. Sci. Technol. 2018, 7, 581–591. [Google Scholar] [CrossRef]
  164. Di Liberto, M.G.; Seimandi, G.M.; Fernández, L.N.; Ruiz, V.E.; Svetaz, L.A.; Derita, M.G. Botanical Control of Citrus Green Mold and Peach Brown Rot on Fruits Assays Using a Persicaria Acuminata Phytochemically Characterized Extract. Plants 2021, 10, 425. [Google Scholar] [CrossRef] [PubMed]
  165. Rayens, E.; Norris, K.A.; Cordero, J.C.D.S.F. Mortality Trends in Risk Conditions and Invasive Mycotic Disease in the United States, 1999–2018. Clin. Infect. Dis. 2022, 74, 309–318. [Google Scholar] [CrossRef] [PubMed]
  166. McCarthy, M.; Rosengart, A.; Schuetz, A.N.; Kontoyiannis, D.P.; Walsh, T.J. Mold Infections of the Central Nervous System. N. Engl. J. Med. 2014, 371, 150–160. [Google Scholar] [CrossRef]
  167. De Bernardis, F.; Amacker, M.; Arancia, S.; Sandini, S.; Gremion, C.; Zurbriggen, R.; Moser, C.; Cassone, A. A Virosomal Vaccine against Candidal Vaginitis: Immunogenicity, Efficacy and Safety Profile in Animal Models. Vaccine 2012, 30, 4490–4498. [Google Scholar] [CrossRef]
  168. Nakama, T.; Yamashita, S.; Hirahara, T.; Okamoto, S.; Honda, S.; Watanabe, M.; Kimura, E.; Uchino, M.; Yano, S.; Kuratsu, J.I.; et al. Usefulness of Intraventricular Infusion of Antifungal Drugs through Ommaya Reservoirs for Cryptococcal Meningitis Treatment. J. Neurol. Sci. 2015, 358, 259–262. [Google Scholar] [CrossRef]
  169. Mendonça, D.V.C.; Lage, L.M.R.; Lage, D.P.; Chávez-Fumagalli, M.A.; Ludolf, F.; Roatt, B.M.; Menezes-Souza, D.; Faraco, A.A.G.; Castilho, R.O.; Tavares, C.A.P.; et al. Poloxamer 407 (Pluronic® F127)-Based Polymeric Micelles for Amphotericin B: In Vitro Biological Activity, Toxicity and in Vivo Therapeutic Efficacy against Murine Tegumentary Leishmaniasis. Exp. Parasitol. 2016, 169, 34–42. [Google Scholar] [CrossRef]
  170. Bachtiar, E.W.; Dewiyani, S.; Surono Akbar, S.M.; Bachtiar, B.M. Inhibition of Candida Albicans Biofilm Development by Unencapsulated Enterococcus Faecalis Cps2. J. Dent. Sci. 2016, 11, 323–330. [Google Scholar] [CrossRef]
  171. Sobel, J.D.; Fisher, J.F.; Kauffman, C.A.; Newman, C.A. Candida Urinary Tract Infections—Epidemiology. Clin. Infect. Dis. 2011, 52, S433–S436. [Google Scholar] [CrossRef]
  172. Pilmis, B.; Puel, A.; Lortholary, O.; Lanternier, F. New Clinical Phenotypes of Fungal Infections in Special Hosts. Clin. Microbiol. Infect. 2016, 22, 681–687. [Google Scholar] [CrossRef]
  173. Zida, A.; Bamba, S.; Yacouba, A.; Ouedraogo-Traore, R.; Guiguemdé, R.T. Anti-Candida albicans Natural Products, Sources of New Antifungal Drugs: A Review. J. Mycol. Med. 2017, 27, 1–19. [Google Scholar] [CrossRef]
  174. Soliman, G.M. Nanoparticles as Safe and Effective Delivery Systems of Antifungal Agents: Achievements and Challenges. Int. J. Pharm. 2017, 523, 15–32. [Google Scholar] [CrossRef]
  175. Raghavendra, R.; Mahadevan, G.D. In Vitro Antimicrobial Activity of Various Plant Latex against Resistant Human Pathogens. Int. J. Pharm. Pharm. Sci. 2011, 3, 70–72. [Google Scholar]
  176. Khan, H.; Khan, Z.; Amin, S.; Mabkhot, Y.N.; Mubarak, M.S.; Hadda, T.B.; Maione, F. Plant Bioactive Molecules Bearing Glycosides as Lead Compounds for the Treatment of Fungal Infection: A Review. Biomed. Pharmacother. 2017, 93, 498–509. [Google Scholar] [CrossRef]
  177. Kainz, K.; Bauer, M.A.; Madeo, F.; Carmona-Gutierrez, D. Fungal Infections in Humans: The Silent Crisis. Microb. Cell 2020, 7, 143. [Google Scholar] [CrossRef] [PubMed]
  178. Wei, Y.-X.; Xu, X.-Y.; Song, X. A Review of Antifungal Natural Products Against the Pathogenic Fungi Causing Athletes’ Foot Disease. Curr. Org. Chem. 2017, 21, 1907–1919. [Google Scholar] [CrossRef]
  179. Kean, R.; Ramage, G. Combined Antifungal Resistance and Biofilm Tolerance: The Global Threat of Candida auris. Msphere 2019, 4, e00458-19. [Google Scholar] [CrossRef] [PubMed]
  180. Bandara, N.; Samaranayake, L. Emerging and Future Strategies in the Management of Recalcitrant Candida auris. Med. Mycol. 2022, 60, myac008. [Google Scholar] [CrossRef]
  181. Billamboz, M.; Fatima, Z.; Hameed, S.; Jawhara, S. Promising Drug Candidates and New Strategies for Fighting against the Emerging Superbug Candida auris. Microorganisms 2021, 9, 634. [Google Scholar] [CrossRef]
  182. Garcia-Rubio, R.; de Oliveira, H.C.; Rivera, J.; Trevijano-Contador, N. The Fungal Cell Wall: Candida, Cryptococcus, and Aspergillus Species. Front. Microbiol. 2020, 10, 492056. [Google Scholar] [CrossRef]
  183. Lepesheva, G.I.; Waterman, M.R. Sterol 14α-Demethylase Cytochrome P450 (CYP51), a P450 in All Biological Kingdoms. Biochim. Biophys. Acta-Gen. Subj. 2007, 1770, 467–477. [Google Scholar] [CrossRef]
  184. Jakubczyk, D.; Dussart, F. Selected Fungal Natural Products with Antimicrobial Properties. Molecules 2020, 25, 911. [Google Scholar] [CrossRef] [PubMed]
  185. De Moraes, D.C. Current Scenario of the Search for New Antifungal Agents to Treat Candida Auris Infections: An Integrative Review. J. Med. Mycol. 2022, 32, 101232. [Google Scholar] [CrossRef] [PubMed]
  186. Juvvadi, P.R.; Lamoth, F.; Steinbach, W.J. Calcineurin as a Multifunctional Regulator: Unraveling Novel Functions in Fungal Stress Responses, Hyphal Growth, Drug Resistance, and Pathogenesis. Fungal Biol. Rev. 2014, 28, 56–69. [Google Scholar] [CrossRef]
  187. Gifford, J.L.; Hunter, H.N.; Vogel, H.J. Lactoferricin: A Lactoferrin-Derived Peptide with Antimicrobial, Antiviral, Antitumor and Immunological Properties. Cell. Mol. Life Sci. 2005, 62, 2588. [Google Scholar] [CrossRef] [PubMed]
  188. Da Rocha Neto, A.C.; Navarro, B.B.; Canton, L.; Maraschin, M.; Di Piero, R.M. Antifungal Activity of Palmarosa (Cymbopogon martinii), Tea Tree (Melaleuca alternifolia) and Star Anise (Illicium verum) Essential Oils against Penicillium Expansum and Their Mechanisms of Action. LWT-Food Sci. Technol. 2019, 105, 385–392. [Google Scholar] [CrossRef]
  189. Li, W.; Yuan, S.; Sun, J.; Li, Q.; Jiang, W.; Cao, J. Ethyl P-Coumarate Exerts Antifungal Activity in Vitro and in Vivo against Fruit Alternaria alternata via Membrane-Targeted Mechanism. Int. J. Food Microbiol. 2018, 278, 26–35. [Google Scholar] [CrossRef]
  190. Yuan, S.; Li, W.; Li, Q.; Wang, L.; Cao, J.; Jiang, W. Defense Responses, Induced by p-Coumaric Acid and Methyl p-Coumarate, of Jujube (Ziziphus jujuba Mill.) Fruit against Black Spot Rot Caused by Alternaria Alternata. J. Agric. Food Chem. 2019, 67, 2801–2810. [Google Scholar] [CrossRef]
  191. Xu, Y.; Wei, J.; Wei, Y.; Han, P.; Dai, K.; Zou, X.; Jiang, S.; Xu, F.; Wang, H.; Sun, J.; et al. Tea Tree Oil Controls Brown Rot in Peaches by Damaging the Cell Membrane of Monilinia Fructicola. Postharvest Biol. Technol. 2021, 175, 111474. [Google Scholar] [CrossRef]
  192. Wang, Y.; Liu, X.; Chen, T.; Xu, Y.; Tian, S. Antifungal Effects of Hinokitiol on Development of Botrytis Cinerea in Vitro and in Vivo. Postharvest Biol. Technol. 2020, 159, 111038. [Google Scholar] [CrossRef]
  193. Yan, J.; Wu, H.; Shi, F.; Wang, H.; Chen, K.; Feng, J.; Jia, W. Antifungal Activity Screening for Mint and Thyme Essential Oils against Rhizopus Stolonifer and Their Application in Postharvest Preservation of Strawberry and Peach Fruits. J. Appl. Microbiol. 2021, 130, 1993–2007. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, Y.; Qiao, Y.; Zhang, M.; Ma, Z.; Xue, Y.; Mi, Q.; Wang, A.; Feng, J. Potential Value of Small-Molecule Organic Acids for the Control of Postharvest Gray Mold Caused by Botrytis Cinerea. Pestic. Biochem. Physiol. 2021, 177, 104884. [Google Scholar] [CrossRef] [PubMed]
  195. Xin, Z.; OuYang, Q.; Wan, C.; Che, J.; Li, L.; Chen, J.; Tao, N. Isolation of Antofine from Cynanchum atratum BUNGE (Asclepiadaceae) and Its Antifungal Activity against Penicillium digitatum. Postharvest Biol. Technol. 2019, 157, 110961. [Google Scholar] [CrossRef]
  196. Mendoza, L.; Ribera, A.; Saavedra, A.; Silva, E.; Araya-Maturana, R.; Cotoras, M. Action Mechanism for 3β-Hydroxykaurenoic Acid and 4,4-Dimethylanthracene-1,9,10(4H)-Trione on Botrytis cinerea. Mycologia 2015, 107, 661–666. [Google Scholar] [CrossRef]
  197. Lebaron, P.; Catala, P.; Parthuisot, N. Effectiveness of SYTOX Green Stain for Bacterial Viability Assessment. Appl. Environ. Microbiol. 1998, 64, 2697–2700. [Google Scholar] [CrossRef]
  198. Tian, S.; Qin, G.; Li, B. Reactive Oxygen Species Involved in Regulating Fruit Senescence and Fungal Pathogenicity. Plant Mol. Biol. 2013, 82, 593–602. [Google Scholar] [CrossRef]
  199. Chen, Y.; Xing, M.; Chen, T.; Tian, S.; Li, B. Effects and Mechanisms of Plant Bioactive Compounds in Preventing Fungal Spoilage and Mycotoxin Contamination in Postharvest Fruits: A Review. Food Chem. 2023, 415, 135787. [Google Scholar] [CrossRef]
  200. Kelts, J.L.; Cali, J.J.; Duellman, S.J.; Shultz, J. Altered Cytotoxicity of ROS-Inducing Compounds by Sodium Pyruvate in Cell Culture Medium Depends on the Location of ROS Generation. Springerplus 2015, 4, 269. [Google Scholar] [CrossRef]
  201. Shlezinger, N.; Minz, A.; Gur, Y.; Hatam, I.; Dagdas, Y.F.; Talbot, N.J.; Sharon, A. Anti-Apoptotic Machinery Protects the Necrotrophic Fungus Botrytis Cinerea from Host-Induced Apoptotic-like Cell Death during Plant Infection. PLoS Pathog. 2011, 7, e1002185. [Google Scholar] [CrossRef]
  202. Aguayo, C.; Riquelme, J.; Valenzuela, P.D.T.; Hahn, M.; Moreno Silva, E. Bchex Virulence Gene of Botrytis Cinerea: Characterization and Functional Analysis. J. Gen. Plant Pathol. 2011, 77, 230–238. [Google Scholar] [CrossRef]
  203. Collinge, A.J.; Markham, P. Woronin Bodies Rapidly Plug Septal Pores of Severed Penicillium chrysogenum Hyphae. Exp. Mycol. 1985, 9, 80–85. [Google Scholar] [CrossRef]
  204. Magnani, T.; Soriani, F.M.; Martins, V.P.; Nascimento, A.M.; Tudella, V.G.; Curti, C.; Uyemura, S.A. Cloning and Functional Expression of the Mitochondrial Alternative Oxidase of Aspergillus Fumigatus and Its Induction by Oxidative Stress. FEMS Microbiol. Lett. 2007, 271, 230–238. [Google Scholar] [CrossRef] [PubMed]
  205. Honda, Y.; Hattori, T.; Kirimura, K. Visual Expression Analysis of the Responses of the Alternative Oxidase Gene (Aox1) to Heat Shock, Oxidative, and Osmotic Stresses in Conidia of Citric Acid-Producing Aspergillus Niger. J. Biosci. Bioeng. 2012, 113, 338–342. [Google Scholar] [CrossRef]
  206. Latgé, J.P.; Beauvais, A.; Chamilos, G. The Cell Wall of the Human Fungal Pathogen Aspergillus Fumigatus: Biosynthesis, Organization, Immune Response, and Virulence. Annu. Rev. Microbiol. 2017, 71, 99–116. [Google Scholar] [CrossRef]
  207. Corrêa, J.L.; Veiga, F.F.; Jarros, I.C.; Costa, M.I.; Castilho, P.F.; de Oliveira, K.M.P.; Rosseto, H.C.; Bruschi, M.L.; Svidzinski, T.I.E.; Negri, M. Propolis Extract Has Bioactivity on the Wall and Cell Membrane of Candida albicans. J. Ethnopharmacol. 2020, 256, 112791. [Google Scholar] [CrossRef]
  208. Du, H.; Guan, G.; Xie, J.; Sun, Y.; Tong, Y.; Zhang, L.; Huang, G. Roles of Candida Albicans Gat2, a GATA-Type Zinc Finger Transcription Factor, in Biofilm Formation, Filamentous Growth and Virulence. PLoS ONE 2012, 7, e29707. [Google Scholar] [CrossRef] [PubMed]
  209. Ramage, G.; Saville, S.P.; Thomas, D.P.; López-Ribot, J.L. Candida biofilms: An Update. Eukaryot. Cell 2005, 4, 633–638. [Google Scholar] [CrossRef] [PubMed]
  210. Modrzewska, B.; Kurnatowski, P. Selected Pathogenic Characteristics of Fungi from the Genus Candida. Ann. Parasitol. 2013, 59, 57–66. [Google Scholar]
  211. Gow, N.A.R.; Van De Veerdonk, F.L.; Brown, A.J.P.; Netea, M.G. Candida Albicans Morphogenesis and Host Defence: Discriminating Invasion from Colonization. Nat. Rev. Microbiol. 2012, 10, 112–122. [Google Scholar] [CrossRef]
  212. Whiteway, M.; Bachewich, C. Morphogenesis in Candida albicans. Annu. Rev. Microbiol. 2007, 61, 529–553. [Google Scholar] [CrossRef]
  213. Baillie, G.S.; Douglas, L.J. Role of Dimorphism in the Development of Candida albicans Biofilms. J. Med. Microbiol. 1999, 48, 671–679. [Google Scholar] [CrossRef] [PubMed]
  214. Leng, P.; Lee, P.R.; Wu, H.; Brown, A.J.P. Efg1, a Morphogenetic Regulator in Candida albicans, Is a Sequence-Specific DNA Binding Protein. J. Bacteriol. 2001, 183, 4090–4093. [Google Scholar] [CrossRef] [PubMed]
  215. Chen, J.; Zhou, S.; Wang, Q.; Chen, X.; Pan, T.; Liu, H. Crk1, a Novel Cdc2-Related Protein Kinase, Is Required for Hyphal Development and Virulence in Candida Albicans. Mol. Cell. Biol. 2000, 20, 8696–8708. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, Q.; Zhou, S.; Chen, J.Y. Functions of CRK1 Gene of Candida albicans as Studied by Gene Knock-Out. Acta Biochim. Biophys. Sin. 1999, 31, 545–552. [Google Scholar]
  217. Ni, J.; Gao, Y.; Liu, H.; Chen, J. Candida albicans Cdc37 Interacts with the Crk1 Kinase and Is Required for Crk1 Production. FEBS Lett. 2004, 561, 223–230. [Google Scholar] [CrossRef]
  218. Xu, D.; Jiang, B.; Ketela, T.; Lemieux, S.; Veillette, K.; Martel, N.; Davison, J.; Sillaots, S.; Trosok, S.; Bachewich, C.; et al. Genome-Wide Fitness Test and Mechanism-of-Action Studies of Inhibitory Compounds in Candida albicans. PLoS Pathog. 2007, 3, e92. [Google Scholar] [CrossRef]
  219. Giaever, G.; Shoemaker, D.D.; Jones, T.W.; Liang, H.; Winzeler, E.A.; Astromoff, A.; Davis, R.W. Genomic Profiling of Drug Sensitivities via Induced Haploinsufficiency. Nat. Genet. 1999, 21, 278–283. [Google Scholar] [CrossRef]
  220. Nelson, W.J. W(h)Ither the Golgi during Mitosis? J. Cell Biol. 2000, 149, 243–248. [Google Scholar] [CrossRef]
  221. Abbafati, C.; Abbas, K.M.; Abbasi, M.; Abbasifard, M.; Abbasi-Kangevari, M.; Abbastabar, H.; Abd-Allah, F.; Abdelalim, A.; Abdollahi, M.; Abdollahpour, I.; et al. Global Burden of 369 Diseases and Injuries in 204 Countries and Territories, 1990–2019: A Systematic Analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef]
  222. Menichetti, A.; Mavridi-Printezi, A.; Mordini, D.; Montalti, M. Effect of Size, Shape and Surface Functionalization on the Antibacterial Activity of Silver Nanoparticles. J. Funct. Biomater. 2023, 14, 244. [Google Scholar] [CrossRef]
  223. Cohen, M.L. Changing Patterns of Infectious Disease. Nature 2000, 406, 762–767. [Google Scholar] [CrossRef] [PubMed]
  224. Scalbert, A. Antimicrobial Properties of Tannins. Phytochemistry 1991, 30, 3875–3883. [Google Scholar] [CrossRef]
  225. Abbasoglu, U.; Sener, B.; Günay, Y.; Temizer, H. Antimicrobial Activity of Some Isoquinoline Alkaloids. Arch. Pharm. 1991, 324, 379–380. [Google Scholar] [CrossRef] [PubMed]
  226. Guimarães, A.C.; Meireles, L.M.; Lemos, M.F.; Guimarães, M.C.C.; Endringer, D.C.; Fronza, M.; Scherer, R. Antibacterial Activity of Terpenes and Terpenoids Present in Essential Oils. Molecules 2019, 24, 2471. [Google Scholar] [CrossRef] [PubMed]
  227. Cushnie, T.P.T.; Lamb, A.J. Antimicrobial Activity of Flavonoids. Int. J. Antimicrob. Agents 2005, 26, 343–356. [Google Scholar] [CrossRef]
  228. Seimandi, G.; Álvarez, N.; Stegmayer, M.I.; Fernández, L.; Ruiz, V.; Favaro, M.A.; Derita, M. An Update on Phytochemicals and Pharmacological Activities of the Genus Persicaria and Polygonum. Molecules 2021, 26, 5956. [Google Scholar] [CrossRef]
  229. Lim, V.; Gorji, S.G.; Daygon, V.D.; Fitzgerald, M. Untargeted and Targeted Metabolomic Profiling of Australian Indigenous Fruits. Metabolites 2020, 10, 114. [Google Scholar] [CrossRef]
  230. De Souza, M.V.N.; Vasconcelos, T.R.A. Drugs against Tuberculose: Past, Present and Future. Quim. Nova 2005, 28, 678–682. [Google Scholar] [CrossRef]
  231. Palanisamy, G.S.; Smith, E.E.; Shanley, C.A.; Ordway, D.J.; Orme, I.M.; Basaraba, R.J. Disseminated Disease Severity as a Measure of Virulence of Mycobacterium tuberculosis in the Guinea Pig Model. Tuberculosis 2008, 88, 295–306. [Google Scholar] [CrossRef]
  232. Franco-Paredes, C.; Marcos, L.A.; Henao-Martínez, A.F.; Rodríguez-Morales, A.J.; Villamil-Gómez, W.E.; Gotuzzo, E.; Bonifaz, A. Cutaneous Mycobacterial Infections. Clin. Microbiol. Rev. 2019, 32, e00069-18. [Google Scholar] [CrossRef]
  233. Holetz, F.B.; Pessini, G.L.; Sanches, N.R.; Cortez, D.A.G.; Nakamura, C.V.; Dias Filho, B.P. Screening of Some Plants Used in the Brazilian Folk Medicine for the Treatment of Infectious Diseases. Mem. Inst. Oswaldo Cruz 2002, 97, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
  234. Alves, J.A.; Mantovani, A.L.L.; Martins, M.H.G.; Abrao, F.; Lucarini, R.; Crotti, A.E.M.; Martins, C.H.G. Antimycobacterial Activity of Some Commercially Available Plant-Derived Essential Oils. Chem. Nat. Compd. 2015, 51, 353–355. [Google Scholar] [CrossRef]
  235. Kipanga, P.N.; Liu, M.; Panda, S.K.; Mai, A.H.; Veryser, C.; Van Puyvelde, L.; De Borggraeve, W.M.; Van Dijck, P.; Matasyoh, J.; Luyten, W. Biofilm Inhibiting Properties of Compounds from the Leaves of Warburgia ugandensis Sprague Subsp ugandensis against Candida and Staphylococcal Biofilms. J. Ethnopharmacol. 2020, 248, 112352. [Google Scholar] [CrossRef]
  236. Mohammed, S.O.; Ahmed, E.S.; Hafez, E.E.; Khalid, A.; Elshahaby, O.A. Characterization and Purification of Urease Enzyme from New Proteus Mirabilis Strain. J. Adv. Sci. Res. 2014, 5, 8–11. [Google Scholar]
  237. Rattan, R.S. Mechanism of Action of Insecticidal Secondary Metabolites of Plant Origin. Crop Prot. 2010, 29, 913–920. [Google Scholar] [CrossRef]
  238. Bezabih, G.; Satheesh, N.; Workneh Fanta, S.; Wale, M.; Atlabachew, M. Reducing Postharvest Loss of Stored Grains Using Plant-Based Biopesticides: A Review of Past Research Efforts. Adv. Agric. 2022, 2022, 6946916. [Google Scholar] [CrossRef]
  239. Mossa, A.T.H.; Mohafrash, S.M.M.; Chandrasekaran, N. Safety of Natural Insecticides: Toxic Effects on Experimental Animals. Biomed Res. Int. 2018, 2018, 4308054. [Google Scholar] [CrossRef]
  240. Tampe, J.; Espinoza, J.; Chacón-Fuentes, M.; Quiroz, A.; Rubilar, M. Evaluation of Drimys winteri (Canelo) Essential Oil as Insecticide against Acanthoscelides obtectus (Coleoptera: Bruchidae) and Aegorhinus superciliosus (Coleoptera: Curculionidae). Insects 2020, 11, 335. [Google Scholar] [CrossRef]
  241. Shewry, P.R. Improving the Protein Content and Composition of Cereal Grain. J. Cereal Sci. 2007, 46, 239–250. [Google Scholar] [CrossRef]
  242. Athanassiou, C.G.; Kavallieratos, N.G.; Sciarretta, A.; Palyvos, N.E.; Trematerra, P. Spatial Associations of Insects and Mites in Stored Wheat. J. Econ. Entomol. 2011, 104, 1752–1764. [Google Scholar] [CrossRef]
  243. Fornal, J.; Jeliński, T.; Sadowska, J.; Grundas, S.; Nawrot, J.; Niewiada, A.; Warchalewski, J.R.; Błaszczak, W. Detection of Granary Weevil Sitophilus granarius (L.) Eggs and Internal Stages in Wheat Grain Using Soft X-ray and Image Analysis. J. Stored Prod. Res. 2007, 43, 142–148. [Google Scholar] [CrossRef]
  244. Benhalima, H.; Chaudhry, M.Q.; Mills, K.A.; Price, N.R. Phosphine Resistance in Stored-Product Insects Collected from Various Grain Storage Facilities in Morocco. J. Stored Prod. Res. 2004, 40, 241–249. [Google Scholar] [CrossRef]
  245. Bughio, F.M.; Wilkins, R.M. Influence of Malathion Resistance Status on Survival and Growth of Tribolium castaneum (Coleoptera: Tenebrionidae), When Fed on Flour from Insect-Resistant and Susceptible Grain Rice Cultivars. J. Stored Prod. Res. 2004, 40, 65–75. [Google Scholar] [CrossRef]
  246. Simmonds, M.S.J. The Search for Plant-Derived Compounds with Antifeedant Activity. Adv. Phytomed. 2006, 3, 291–324. [Google Scholar] [CrossRef]
  247. Rajashekar, Y.; Bakthavatsalam, N.; Shivanandappa, T. Botanicals as Grain Protectants. Psyche A J. Entomol. 2012, 2012, 646740. [Google Scholar] [CrossRef]
  248. Talukder, F.A. Plant Products as Potential Stored-Product Insect Management Agents—A Mini Review. Emir. J. Food Agric. 2006, 18, 17–32. [Google Scholar] [CrossRef]
  249. van Beek, T.A.; de Groot, A.E. Terpenoid Antifeedants, Part I. An Overview of Terpenoid Antifeedants of Natural Origin. Recl. Des. Trav. Chim. Des. Pays-Bas 1986, 105, 513–527. [Google Scholar] [CrossRef]
  250. Jermy, T. Prospects of Antifeedant Approach to Pest Control—A Critical Review. J. Chem. Ecol. 1990, 16, 3151–3166. [Google Scholar] [CrossRef]
  251. Schoonhoven, L.M. Biological Aspects of Antifeedants. Entomol. Exp. Appl. 1982, 31, 57–69. [Google Scholar] [CrossRef]
  252. Lengai, G.M.W.; Muthomi, J.W.; Mbega, E.R. Phytochemical Activity and Role of Botanical Pesticides in Pest Management for Sustainable Agricultural Crop Production. Sci. Afr. 2020, 7, e00239. [Google Scholar] [CrossRef]
  253. Damalas, C.A.; Koutroubas, S.D. Current Status and Recent Developments in Biopesticide Use. Agriculture 2018, 8, 13. [Google Scholar] [CrossRef]
  254. Sharma, A.; Shukla, A.; Attri, K.; Kumar, M.; Kumar, P.; Suttee, A.; Singh, G.; Barnwal, R.P.; Singla, N. Global Trends in Pesticides: A Looming Threat and Viable Alternatives. Ecotoxicol. Environ. Saf. 2020, 201, 110812. [Google Scholar] [CrossRef]
  255. Isman, M.B. Botanical Insecticides in the Twenty-First Century-Fulfilling Their Promise? Annu. Rev. Entomol. 2020, 65, 233–249. [Google Scholar] [CrossRef]
  256. Isman, M. Insect Antifeedants. Pestic. Outlook 2002, 13, 152–157. [Google Scholar] [CrossRef]
  257. Isman, M.B. Botanical Insecticides, Deterrents, and Repellents in Modern Agriculture and an Increasingly Regulated World. Annu. Rev. Entomol. 2006, 51, 45–66. [Google Scholar] [CrossRef]
  258. Miresmailli, S.; Isman, M.B. Botanical Insecticides Inspired by Plant-Herbivore Chemical Interactions. Trends Plant Sci. 2014, 19, 29–35. [Google Scholar] [CrossRef] [PubMed]
  259. Ogwuru, N.; Adamczeski, M. Bioactive Natural Products Derived from Polygonum Species of Plants: Their Structures and Mechanisms of Action. Stud. Nat. Prod. Chem. 2000, 22, 607–642. [Google Scholar] [CrossRef]
  260. Magalhães, L.M.D.; Gollob, K.J.; Zingales, B.; Dutra, W.O. Pathogen Diversity, Immunity, and the Fate of Infections: Lessons Learned from Trypanosoma cruzi Human–Host Interactions. Lancet Microbe 2022, 3, e711–e722. [Google Scholar] [CrossRef]
  261. Ferri, G.; Edreira, M.M. All Roads Lead to Cytosol: Trypanosoma Cruzi Multi-Strategic Approach to Invasion. Front. Cell. Infect. Microbiol. 2021, 11, 634793. [Google Scholar] [CrossRef]
  262. Liu, Z.; Ulrich vonBargen, R.; McCall, L.I. Central Role of Metabolism in Trypanosoma Cruzi Tropism and Chagas Disease Pathogenesis. Curr. Opin. Microbiol. 2021, 63, 204–209. [Google Scholar] [CrossRef]
  263. Pérez-Molina, J.A.; Crespillo-Andújar, C.; Bosch-Nicolau, P.; Molina, I. Trypanocidal Treatment of Chagas Disease. Enferm. Infecc. Microbiol. Clin. 2021, 39, 458–470. [Google Scholar] [CrossRef] [PubMed]
  264. Vela, A.; Coral-Almeida, M.; Sereno, D.; Costales, J.A.; Barnabé, C.; Brenière, S.F. In Vitro Susceptibility of Trypanosoma Cruzi Discrete Typing Units (Dtus) to Benznidazole: A Systematic Review and Meta-Analysis. PLoS Negl. Trop. Dis. 2021, 15, e0009269. [Google Scholar] [CrossRef] [PubMed]
  265. Crespillo-Andújar, C.; Chamorro-Tojeiro, S.; Norman, F.; Monge-Maillo, B.; López-Vélez, R.; Pérez-Molina, J.A. Toxicity of Nifurtimox as Second-Line Treatment after Benznidazole Intolerance in Patients with Chronic Chagas Disease: When Available Options Fail. Clin. Microbiol. Infect. 2018, 24, P1344.E1–P1344.E4. [Google Scholar] [CrossRef]
  266. Nekoei, S.; Khamesipour, F.; Habtemariam, S.; de Souza, W.; Mohammadi Pour, P.; Hosseini, S.R. The Anti-Trypanosoma Activities of Medicinal Plants: A Systematic Review of the Literature. Vet. Med. Sci. 2022, 8, 2738–2772. [Google Scholar] [CrossRef]
  267. Tomar, R.; Tiwari, R.; Gupta, R.; Bhargava, S.; Bisht, D.; Rana, V.S.; Sethiya, N.K. Natural Products in the Management of Trypanosomiasis. In Natural Products in Vector-Borne Disease Management; Elsevier: Amsterdam, The Netherlands, 2023; pp. 379–411. [Google Scholar]
  268. Cockram, P.E.; Smith, T.K. Active Natural Product Scaffolds against Trypanosomatid parasites: A Review. J. Nat. Prod. 2018, 81, 2138–2154. [Google Scholar] [CrossRef] [PubMed]
  269. Muñoz, O.M.; Maya, J.D.; Ferreira, J.; Christen, P.; San Martin, J.; López-Muñoz, R.; Morello, A.; Kemmerling, U. Medicinal Plants of Chile: Evaluation of Their Anti-Trypanosoma Cruzi Activity. Z. für Naturforsch. C 2013, 68, 198–202. [Google Scholar] [CrossRef]
  270. García-Huertas, P.; Cardona-Castro, N. Advances in the Treatment of Chagas Disease: Promising New Drugs, Plants and Targets. Biomed. Pharmacother. 2021, 142, 112020. [Google Scholar] [CrossRef]
  271. Barbosa, H.; Thevenard, F.; Quero Reimão, J.; Tempone, A.G.; Honorio, K.M.; Lago, J.H.G. The Potential of Secondary Metabolites from Plants as Drugs or Leads against Trypanosoma Cruzi-An Update from 2012 to 2021. Curr. Top. Med. Chem. 2023, 23, 159–213. [Google Scholar] [CrossRef]
  272. Taman, A.; Azab, M. Present-Day Anthelmintics and Perspectives on Future New Targets. Parasitol. Res. 2014, 113, 2425–2433. [Google Scholar] [CrossRef]
  273. Loukas, A.; Hotez, P.J.; Diemert, D.; Yazdanbakhsh, M.; McCarthy, J.S.; Correa-Oliveira, R.; Croese, J.; Bethony, J.M. Hookworm Infection. Nat. Rev. Dis. Prim. 2016, 2, 16088. [Google Scholar] [CrossRef]
  274. Rana, A.K.; Misra-Bhattacharya, S. Current Drug Targets for Helminthic Diseases. Parasitol. Res. 2013, 112, 1819–1831. [Google Scholar] [CrossRef] [PubMed]
  275. Norhayati, M.; Fatmah, M.S.; Yusof, S.; Edariah, A.B. Intestinal Parasitic Infections in Man: A Review. Med. J. Malays. 2003, 58, 296–305. [Google Scholar]
  276. Bajin ba Ndob, I.; Mengome, L.E.; Bourobou Bourobou, H.P.; Lossangoye Banfora, Y.; Bivigou, F. Ethnobotanical Survey of Medicinal Plants Used as Anthelmintic Remedies in Gabon. J. Ethnopharmacol. 2016, 191, 360–371. [Google Scholar] [CrossRef] [PubMed]
  277. Brindley, P.J.; Loukas, A. Helminth Infection–Induced Malignancy. PLoS Pathog. 2017, 13, e1006393. [Google Scholar] [CrossRef]
  278. Li, R.W.; Rinaldi, M.; Capuco, A.V. Characterization of the Abomasal Transcriptome for Mechanisms of Resistance to Gastrointestinal Nematodes in Cattle. Vet. Res. 2011, 42, 114. [Google Scholar] [CrossRef]
  279. Bachaya, H.A.; Iqbal, Z.; Khan, M.N.; Sindhu, Z.u.D.; Jabbar, A. Anthelmintic Activity of Ziziphus nummularia (Bark) and Acacia nilotica (Fruit) against Trichostrongylid nematodes of Sheep. J. Ethnopharmacol. 2009, 123, 325–329. [Google Scholar] [CrossRef]
  280. Kaewintajuk, K.; Cho, P.Y.; Kim, S.Y.; Lee, E.S.; Lee, H.K.; Choi, E.B.; Park, H. Anthelmintic Activity of KSI-4088 against Caenorhabditis Elegans. Parasitol. Res. 2010, 107, 27–30. [Google Scholar] [CrossRef] [PubMed]
  281. Geurden, T.; Chartier, C.; Fanke, J.; di Regalbono, A.F.; Traversa, D.; von Samson-Himmelstjerna, G.; Demeler, J.; Vanimisetti, H.B.; Bartram, D.J.; Denwood, M.J. Anthelmintic Resistance to Ivermectin and Moxidectin in Gastrointestinal Nematodes of Cattle in Europe. Int. J. Parasitol. Drugs Drug Resist. 2015, 5, 163–171. [Google Scholar] [CrossRef] [PubMed]
  282. Laing, R.; Maitland, K.; Lecová, L.; Skuce, P.J.; Tait, A.; Devaney, E. Analysis of Putative Resistance Gene Loci in UK Field Populations of Haemonchus Contortus after 6 Years of Macrocyclic Lactone Use. Int. J. Parasitol. 2016, 46, 621–630. [Google Scholar] [CrossRef]
  283. Epe, C.; Kaminsky, R. New Advancement in Anthelmintic Drugs in Veterinary Medicine. Trends Parasitol. 2013, 29, 129–134. [Google Scholar] [CrossRef]
  284. Romero-Benavides, J.C.; Ruano, A.L.; Silva-Rivas, R.; Castillo-Veintimilla, P.; Vivanco-Jaramillo, S.; Bailon-Moscoso, N. Medicinal Plants Used as Anthelmintics: Ethnomedical, Pharmacological, and Phytochemical Studies. Eur. J. Med. Chem. 2017, 129, 209–217. [Google Scholar] [CrossRef]
  285. Kumarasingha, R.; Karpe, A.V.; Preston, S.; Yeo, T.C.; Lim, D.S.L.; Tu, C.L.; Luu, J.; Simpson, K.J.; Shaw, J.M.; Gasser, R.B.; et al. Metabolic Profiling and in Vitro Assessment of Anthelmintic Fractions of Picria fel-terrae Lour. Int. J. Parasitol. Drugs Drug Resist. 2016, 6, 171–178. [Google Scholar] [CrossRef] [PubMed]
  286. Edwards, J.; Brown, M.; Peak, E.; Bartholomew, B.; Nash, R.J.; Hoffmann, K.F. The Diterpenoid 7-Keto-Sempervirol, Derived from Lycium Chinense, Displays Anthelmintic Activity against Both Schistosoma Mansoni and Fasciola Hepatica. PLoS Negl. Trop. Dis. 2015, 9, e0003604. [Google Scholar] [CrossRef] [PubMed]
  287. Wangchuk, P.; Giacomin, P.R.; Pearson, M.S.; Smout, M.J.; Loukas, A. Identification of Lead Chemotherapeutic Agents from Medicinal Plants against Blood Flukes and Whipworms. Sci. Rep. 2016, 6, 32101. [Google Scholar] [CrossRef]
  288. Wangchuk, P.; Pearson, M.S.; Giacomin, P.R.; Becker, L.; Sotillo, J.; Pickering, D.; Smout, M.J.; Loukas, A. Compounds Derived from the Bhutanese Daisy, Ajania nubigena, Demonstrate Dual Anthelmintic Activity against Schistosoma mansoni and Trichuris muris. PLoS Negl. Trop. Dis. 2016, 10, e0004908. [Google Scholar] [CrossRef] [PubMed]
  289. Burns, A.R.; Luciani, G.M.; Musso, G.; Bagg, R.; Yeo, M.; Zhang, Y.; Rajendran, L.; Glavin, J.; Hunter, R.; Redman, E.; et al. Caenorhabditis Elegans Is a Useful Model for Anthelmintic Discovery. Nat. Commun. 2015, 6, 7485. [Google Scholar] [CrossRef] [PubMed]
  290. Salas, A.L.; Alberto, M.R.; Zampini, I.C.; Cuello, A.S.; Maldonado, L.; Ríos, J.L.; Schmeda-Hirschmann, G.; Isla, M.I. Biological Activities of Polyphenols-Enriched Propolis from Argentina Arid Regions. Phytomedicine 2016, 23, 27–31. [Google Scholar] [CrossRef]
  291. Weaver, K.J.; May, C.J.; Ellis, B.L. Using a Health-Rating System to Evaluate the Usefulness of Caenorhabditis Elegans as a Model for Anthelmintic Study. PLoS ONE 2017, 12, e0179376. [Google Scholar] [CrossRef]
  292. Katiki, L.M.; Ferreira, J.F.S.; Zajac, A.M.; Masler, C.; Lindsay, D.S.; Chagas, A.C.S.; Amarante, A.F.T. Caenorhabditis Elegans as a Model to Screen Plant Extracts and Compounds as Natural Anthelmintics for Veterinary Use. Vet. Parasitol. 2011, 182, 264–268. [Google Scholar] [CrossRef]
  293. Kumarasingha, R.; Palombo, E.A.; Bhave, M.; Yeo, T.C.; Lim, D.S.L.; Tu, C.L.; Shaw, J.M.; Boag, P.R. Enhancing a Search for Traditional Medicinal Plants with Anthelmintic Action by Using Wild Type and Stress Reporter Caenorhabditis Elegans Strains as Screening Tools. Int. J. Parasitol. 2014, 44, 291–298. [Google Scholar] [CrossRef]
  294. Piña-Vázquez, D.M.; Mayoral-Peña, Z.; Gómez-Sánchez, M.; Salazar-Olivo, L.A.; Arellano-Carbajal, F. Anthelmintic Effect of Psidium Guajava and Tagetes Erecta on Wild-Type and Levamisole-Resistant Caenorhabditis Elegans Strains. J. Ethnopharmacol. 2017, 202, 92–96. [Google Scholar] [CrossRef] [PubMed]
  295. Liu, M.; Kipanga, P.; Mai, A.H.; Dhondt, I.; Braeckman, B.P.; De Borggraeve, W.; Luyten, W. Bioassay-Guided Isolation of Three Anthelmintic Compounds from Warburgia ugandensis Sprague Subspecies Ugandensis, and the Mechanism of Action of Polygodial. Int. J. Parasitol. 2018, 48, 833–844. [Google Scholar] [CrossRef] [PubMed]
  296. Liu, J.; Pope, C.N. Intrinsic and Extrinsic Factors That Can Modify Toxicity. In An Introduction to Interdisciplinary Toxicology: From Molecules to Man; Elsevier: Amsterdam, The Netherlands, 2020; pp. 285–293. [Google Scholar]
  297. Lim, G.P.; Soon, C.F.; Ma, N.L.; Morsin, M.; Nayan, N.; Ahmad, M.K.; Tee, K.S. Cytotoxicity of MXene-Based Nanomaterials for Biomedical Applications: A Mini Review. Environ. Res. 2021, 201, 111592. [Google Scholar] [CrossRef] [PubMed]
  298. Godlewska-żyłkiewicz, B.; Świsłocka, R.; Kalinowska, M.; Golonko, A.; Świderski, G.; Arciszewska, Ż.; Nalewajko-Sieliwoniuk, E.; Naumowicz, M.; Lewandowski, W. Biologically Active Compounds of Plants: Structure-Related Antioxidant, Microbiological and Cytotoxic Activity of Selected Carboxylic Acids. Materials 2020, 13, 4454. [Google Scholar] [CrossRef]
  299. Kahlos, K.; Kiviranta, J.L.J.; Hiltunen, R.V.K. Volatile Constituents of Wild and in Vitro Cultivated Gloeophyllum Odoratum. Phytochemistry 1994, 36, 917–922. [Google Scholar] [CrossRef]
  300. Montenegro, I.J.; del Corral, S.; Diaz Napal, G.N.; Carpinella, M.C.; Mellado, M.; Madrid, A.M.; Villena, J.; Palacios, S.M.; Cuellar, M.A. Antifeedant Effect of Polygodial and Drimenol Derivatives against Spodoptera Frugiperda and Epilachna Paenulata and Quantitative Structure-Activity Analysis. Pest Manag. Sci. 2018, 74, 1623–1629. [Google Scholar] [CrossRef]
  301. Melo, R.; Armstrong, V.; Navarro, F.; Castro, P.; Mendoza, L.; Cotoras, M. Characterization of the Fungitoxic Activity on Botrytis Cinerea of N-Phenyl-Driman-9-Carboxamides. J. Fungi 2021, 7, 902. [Google Scholar] [CrossRef]
  302. Mahnashi, M.H.; Alqahtani, Y.S.; Alyami, B.A.; Alqarni, A.O.; Ullah, F.; Wadood, A.; Sadiq, A.; Shareef, A.; Ayaz, M. Cytotoxicity, Anti-Angiogenic, Anti-Tumor and Molecular Docking Studies on Phytochemicals Isolated from Polygonum hydropiper L. BMC Complement. Med. Ther. 2021, 21, 239. [Google Scholar] [CrossRef]
  303. Shahed, C.A.; Ahmad, F.; Günister, E.; Foudzi, F.M.; Ali, S.; Malik, K.; Harun, W.S.W. Antibacterial Mechanism with Consequent Cytotoxicity of Different Reinforcements in Biodegradable Magnesium and Zinc Alloys: A Review. J. Magnes. Alloy. 2023, 11, 3038–3058. [Google Scholar] [CrossRef]
  304. Aumeeruddy, M.Z.; Mahomoodally, M.F. Global Documentation of Traditionally Used Medicinal Plants in Cancer Management: A Systematic Review. S. Afr. J. Bot. 2021, 138, 424–494. [Google Scholar] [CrossRef]
  305. Esther Lydia, D.; Khusro, A.; Immanuel, P.; Esmail, G.A.; Al-Dhabi, N.A.; Arasu, M.V. Photo-Activated Synthesis and Characterization of Gold Nanoparticles from Punica Granatum L. Seed Oil: An Assessment on Antioxidant and Anticancer Properties for Functional Yoghurt Nutraceuticals. J. Photochem. Photobiol. B Biol. 2020, 206, 111868. [Google Scholar] [CrossRef] [PubMed]
  306. Barbosa, L.G.V.; de Jesus, E.N.S.; Jerônimo, L.B.; Costa, J.S.d.; Silva, R.C.; Setzer, W.N.; da Silva, J.K.R.; da Silva Freitas, J.J.; Figueiredo, P.L. Siparuna guianensis Essential Oil Antitumoral Activity on Ehrlich Model and Its Effect on Oxidative Stress. Chem. Biodivers. 2023, 20, e202301120. [Google Scholar] [CrossRef] [PubMed]
  307. Li, L.; Chen, C.; Chiang, C.; Xiao, T.; Chen, Y.; Zhao, Y.; Zheng, D. The Impact of Trpv1 on Cancer Pathogenesis and Therapy: A Systematic Review. Int. J. Biol. Sci. 2021, 17, 2034. [Google Scholar] [CrossRef] [PubMed]
  308. Benso, B.; Bustos, D.; Zarraga, M.O.; Gonzalez, W.; Caballero, J.; Brauchi, S. Chalcone Derivatives as Non-Canonical Ligands of TRPV1. Int. J. Biochem. Cell Biol. 2019, 112, 18–23. [Google Scholar] [CrossRef]
  309. Bong, A.H.L.; Monteith, G.R. Calcium Signaling and the Therapeutic Targeting of Cancer Cells. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2018, 1865, 1786–1794. [Google Scholar] [CrossRef]
  310. Humeau, J.; Bravo-San Pedro, J.M.; Vitale, I.; Nuñez, L.; Villalobos, C.; Kroemer, G.; Senovilla, L. Calcium Signaling and Cell Cycle: Progression or Death. Cell Calcium 2018, 70, 3–15. [Google Scholar] [CrossRef]
  311. Basu, P.; Maier, C. Phytoestrogens and Breast Cancer: In Vitro Anticancer Activities of Isoflavones, Lignans, Coumestans, Stilbenes and Their Analogs and Derivatives. Biomed. Pharmacother. 2018, 107, 1648–1666. [Google Scholar] [CrossRef]
  312. Singh, J.; Hussain, Y.; Luqman, S.; Meena, A. Targeting Ca2+ Signalling through Phytomolecules to Combat Cancer. Pharmacol. Res. 2019, 146, 104282. [Google Scholar] [CrossRef]
  313. Vangeel, L.; Voets, T. Transient Receptor Potential Channels and Calcium Signaling. Cold Spring Harb. Perspect. Biol. 2019, 11, a035048. [Google Scholar] [CrossRef]
  314. Li, L.; Chen, C.; Xiang, Q.; Fan, S.; Xiao, T.; Chen, Y.; Zheng, D. Transient Receptor Potential Cation Channel Subfamily V Member 1 Expression Promotes Chemoresistance in Non-Small-Cell Lung Cancer. Front. Oncol. 2022, 12, 773654. [Google Scholar] [CrossRef]
  315. Escobar, A.; Pérez, M.; Romanelli, G.; Blustein, G. Thymol Bioactivity: A Review Focusing on Practical Applications. Arab. J. Chem. 2020, 13, 9243–9269. [Google Scholar] [CrossRef]
  316. Liu, M.; Li, W.; Ma, H.; Yang, X.; Liu, A.; Ji, C. Formulation of a Novel Anti-Leukemia Drug and Evaluation of Its Therapeutic Effects in Comparison with Cytarabine. Arab. J. Chem. 2022, 15, 103690. [Google Scholar] [CrossRef]
  317. Chinnathambi, A.; Alahmadi, T.A. Zinc Nanoparticles Green-Synthesized by Alhagi maurorum Leaf Aqueous Extract: Chemical Characterization and Cytotoxicity, Antioxidant, and Anti-Osteosarcoma Effects. Arab. J. Chem. 2021, 14, 103083. [Google Scholar] [CrossRef]
  318. Jha, A.K.; Sit, N. Extraction of Bioactive Compounds from Plant Materials Using Combination of Various Novel Methods: A Review. Trends Food Sci. Technol. 2022, 119, 579–591. [Google Scholar] [CrossRef]
  319. Burgos, V.; Paz, C.; Saavedra, K.; Saavedra, N.; Foglio, M.A.; Salazar, L.A. Drimenol, Isodrimeninol and Polygodial Isolated from Drimys Winteri Reduce Monocyte Adhesion to Stimulated Human Endothelial Cells. Food Chem. Toxicol. 2020, 146, 111775. [Google Scholar] [CrossRef]
  320. Pandey, S.; Pant, P. Possibilities and Challenges for Harnessing Tree Bark Extracts for Wood Adhesives and Green Chemicals and Its Prospects in Nepal. For. Sci. Technol. 2023, 19, 68–77. [Google Scholar] [CrossRef]
  321. Patridge, E.; Gareiss, P.; Kinch, M.S.; Hoyer, D. An Analysis of FDA-Approved Drugs: Natural Products and Their Derivatives. Drug Discov. Today 2016, 21, 204–207. [Google Scholar] [CrossRef]
  322. De Luca, V.; Salim, V.; Atsumi, S.M.; Yu, F. Mining the Biodiversity of Plants: A Revolution in the Making. Science 2012, 336, 1658–1661. [Google Scholar] [CrossRef]
  323. Zhang, H.; Boghigian, B.A.; Armando, J.; Pfeifer, B.A. Methods and Options for the Heterologous Production of Complex Natural Products. Nat. Prod. Rep. 2011, 28, 125–151. [Google Scholar] [CrossRef]
  324. Stuurman, F.E.; Nuijen, B.; Beijnen, J.H.; Schellens, J.H.M. Oral Anticancer Drugs: Mechanisms of Low Bioavailability and Strategies for Improvement. Clin. Pharmacokinet. 2013, 52, 399–414. [Google Scholar] [CrossRef]
  325. Jana, S.; Mandlekar, S.; Marathe, P. Prodrug Design to Improve Pharmacokinetic and Drug Delivery Properties: Challenges to the Discovery Scientists. Curr. Med. Chem. 2010, 17, 3874–3908. [Google Scholar] [CrossRef] [PubMed]
  326. Cragg, G.M.; Newman, D.J. Natural Products: A Continuing Source of Novel Drug Leads. Biochim. Biophys. Acta-Gen. Subj. 2013, 1830, 3670–3695. [Google Scholar] [CrossRef]
  327. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Barreca, D.; Weckwerth, W.; Bauer, R.; Bayer, E.A.; et al. Natural Products in Drug Discovery: Advances and Opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  328. Parthasarathy, A.; Mantravadi, P.K.; Kalesh, K. Detectives and Helpers: Natural Products as Resources for Chemical Probes and Compound Libraries. Pharmacol. Ther. 2020, 216, 107688. [Google Scholar] [CrossRef] [PubMed]
  329. Babaei, G.; Aliarab, A.; Abroon, S.; Rasmi, Y.; Aziz, S.G.G. Application of Sesquiterpene Lactone: A New Promising Way for Cancer Therapy Based on Anticancer Activity. Biomed. Pharmacother. 2018, 106, 239–246. [Google Scholar] [CrossRef] [PubMed]
  330. Ghantous, A.; Gali-Muhtasib, H.; Vuorela, H.; Saliba, N.A.; Darwiche, N. What Made Sesquiterpene Lactones Reach Cancer Clinical Trials? Drug Discov. Today 2010, 15, 668–678. [Google Scholar] [CrossRef]
  331. Maturana, H.; Sierra, J.; Lopez, J.; Cortes, M. Synthesis of 1.3 Dioxans Related with Ambergris. Synth. Commun. 1984, 14, 661–674. [Google Scholar] [CrossRef]
  332. Kuchkova, K.I.; Arycu, A.N.; Vlad, P.F. Synthesis of 11-Aminodrim-7-Ene from Drimenol. Chem. Nat. Compd. 2009, 45, 367–370. [Google Scholar] [CrossRef]
  333. Hu, N.; Wang, X.; Sun, S.; Yang, J.; Li, S. Practical Synthesis and Divergent Optimization of Halichonine B for the Discovery of Novel Pharmaceutical Leads. Org. Chem. Front. 2023, 11, 709–716. [Google Scholar] [CrossRef]
  334. Ramirez, H.E.; Manuel Cortes, M.; Agosin, E. Bioconversion of Drimenol into 3′-Hydroxydrimanes by Aspergillus Niger. Effect of Culture Additives. J. Nat. Prod. 1993, 56, 762–764. [Google Scholar] [CrossRef]
  335. Gonzalez-Coloma, A.; Reina, M.; Diaz, C.E.; Fraga, B.M. Natural Product-Based Biopesticides for Insect Control. In Comprehensive Natural Products II: Chemistry and Biology; Elsevier: Amsterdam, The Netherlands, 2010; Volume 3, pp. 237–268. [Google Scholar]
  336. Aminudin, N.I.; Ridzuan, M.; Susanti, D.; Zainal Abidin, Z.A. Biotransformation of Sesquiterpenoids: A Recent Insight. J. Asian Nat. Prod. Res. 2022, 24, 103–145. [Google Scholar] [CrossRef]
  337. Cortés, M.; Lόpez, J. Synthesis Of (-)-8-Epiambreinolide. Formal Synthesis Ofambraoxide. Nat. Prod. Lett. 1994, 5, 183–186. [Google Scholar] [CrossRef]
  338. Armstrong, V.; Barrero, A.F.; Alvarez-Manzaneda, E.J.; Cortés, M.; Sepúlveda, B. An Efficient Stereoselective Synthesis of Cytotoxic 8-Epipuupehedione. J. Nat. Prod. 2003, 66, 1382–1383. [Google Scholar] [CrossRef] [PubMed]
  339. Maiti, S.; Sengupta, S.; Giri, C.; Achari, B.; Banerjee, A.K. Enantiospecific Synthesis of 8-Epipuupehedione from (R)-(-)-Carvone. Tetrahedron Lett. 2001, 42, 2389–2391. [Google Scholar] [CrossRef]
  340. Gonzalez-Sierra, M.; los Angeles Laborde, M.d.; Ruveda, E.A. Alternative and Stereoselective Synthesis of Εβ(H)-Drimane, a Bicyclic Sesquiterpane of Widespread Occurrence in Petroleums. Synth. Commun. 1987, 17, 431–441. [Google Scholar] [CrossRef]
  341. Cortés, M.; Armstrong, V.; Reyes, M.E.; Lopez, J.; Madariaga, E. Formal Synthesis of Ambrox® and 9-Epiambrox. Synth. Commun. 1996, 26, 1995–2002. [Google Scholar] [CrossRef]
  342. Paquette, L.A.; Maleczka, R.E. Enantioselective Total Synthesis of (-)-9-Epi-Ambrox, a Potent Ambergris-Type Olfactory Agent. J. Org. Chem. 1991, 56, 912–913. [Google Scholar] [CrossRef]
  343. Delgado, V.; Armstrong, V.; Cortés, M.; Barrero, A.F. Synthesis of Racemic and Chiral Albicanol, Albicanyl Acetate and Cyclozonarone: Cytotoxic Activity of Ent-Cyclozonarone. J. Braz. Chem. Soc. 2008, 19, 1258–1263. [Google Scholar] [CrossRef]
  344. Lihui, X.; Xiaojie, Q.; Hao, Y.; Jialiang, C.; Jinming, G.; Ying, C. Albicanol Modulates Oxidative Stress and the P53 Axis to Suppress Profenofos Induced Genotoxicity in Grass Carp Hepatocytes. Fish Shellfish Immunol. 2022, 122, 325–333. [Google Scholar] [CrossRef]
  345. Guan, Y.; Zhao, X.; Song, N.; Cui, Y.; Chang, Y. Albicanol Antagonizes Cd-Induced Apoptosis through a NO/INOS-Regulated Mitochondrial Pathway in Chicken Liver Cells. Food Funct. 2021, 12, 1757–1768. [Google Scholar] [CrossRef]
  346. Ito, H.; Muranaka, T.; Mori, K.; Jin, Z.X.; Tokuda, H.; Nishino, H.; Yoshida, T. Ichthyotoxic Phloroglucinol Derivatives from Dryopteris Fragrans and Their Anti-Tumor Promoting Activity. Chem. Pharm. Bull. 2000, 48, 1190–1195. [Google Scholar] [CrossRef]
  347. Xuan, L.; Guo, J.; Xia, D.; Li, L.; Wang, D.; Chang, Y. Albicanol Antagonizes PFF-Induced Mitochondrial Damage and Reduces Inflammatory Factors by Regulating Innate Immunity. Ecotoxicol. Environ. Saf. 2023, 259, 115014. [Google Scholar] [CrossRef]
  348. Chen, L.L.; Zhang, D.R.; Li, J.; Wang, H.M.; Song, C.H.; Tang, X.; Guan, Y.; Chang, Y.; Wang, W.F. Albicanol Alleviates D-Galactose-Induced Aging and Improves Behavioral Ability Via by Alleviating Oxidative Stress-Induced Damage. Neurochem. Res. 2021, 46, 1058–1067. [Google Scholar] [CrossRef]
  349. Shishido, K.; Tokunaga, Y.; Omachi, N.; Hiroya, K.; Fukumoto, K.; Kametani, T. Total Synthesis of (+)-Albicanol and (+)-Albicanyl Acetate via a Highly Diastereoselective Intramolecular [3 + 2] Cycloaddition. J. Chem. Soc. Perkin Trans. 1 1990, 1, 2481–2486. [Google Scholar] [CrossRef]
  350. Suryani, F.; Aulia Bakhtra, D.D.; Fajrina, A. Cytotoxic Activity of Endophytic Fungus against HeLa Cells (Cervical Cancer Cells): A Article Review. Asian J. Pharm. Res. Dev. 2022, 10, 25–28. [Google Scholar] [CrossRef]
  351. Ncube, E.N.; Steenkamp, L.; Dubery, I.A. Ambrafuran (AmbroxTM) Synthesis from Natural Plant Product Precursors. Molecules 2020, 25, 3851. [Google Scholar] [CrossRef] [PubMed]
  352. Maturana, H.; López, J.; Cortes, M. Synthesis of Ambraoxide from Drimenol. Synth. Commun. 1991, 21, 1533–1543. [Google Scholar] [CrossRef]
  353. He, N.; Li, D.F.; Yu, H.W.; Ye, L.D. Construction of an Artificial Microbial Consortium for Green Production of (−)-Ambroxide. ACS Sustain. Chem. Eng. 2023, 11, 1939–1948. [Google Scholar] [CrossRef]
  354. Benites, J.; Lopez, J.; Farias, J.G.; Cortes, M. The Preparation of Oxygenated Derivatives of Ambrox and Isoambrox from Drimenol. J. Chil. Chem. Soc. 2006, 51, 979–981. [Google Scholar] [CrossRef]
  355. Eichhorn, E.; Locher, E.; Guillemer, S.; Wahler, D.; Fourage, L.; Schilling, B. Biocatalytic Process for (−)-Ambrox Production Using Squalene Hopene Cyclase. Adv. Synth. Catal. 2018, 360, 2339–2351. [Google Scholar] [CrossRef]
  356. Coulerie, P.; Poullain, C. New Caledonia: A ‘Hot Spot’ for Valuable Chemodiversity. Part 1: Gymnosperms. Chem. Biodivers. 2015, 12, 841–858. [Google Scholar] [CrossRef] [PubMed]
  357. Kurata, K.; Taniguchi, K.; Suzuki, M. Cyclozonarone, a Sesquiterpene-Substituted Benzoquinone Derivative from the Brown Alga Dictyopteris Undulata. Phytochemistry 1996, 41, 749–752. [Google Scholar] [CrossRef]
  358. Laube, T.; Beil, W.; Seifert, K. Total Synthesis of Two 12-Nordrimanes and the Pharmacological Active Sesquiterpene Hydroquinone Yahazunol. Tetrahedron 2005, 61, 1141–1148. [Google Scholar] [CrossRef]
  359. Cuellar, M.A.; Salas, C.; Cortés, M.J.; Morello, A.; Maya, J.D.; Preite, M.D. Synthesis and in Vitro Trypanocide Activity of Several Polycyclic Drimane-Quinone Derivatives. Bioorg. Med. Chem. 2003, 11, 2489–2497. [Google Scholar] [CrossRef] [PubMed]
  360. Sobarzo, N.Q.; Venegas, I.M.; Sánchez, C.S.; Catalán, L.E.; Rojas, C.C.; Valdivia, V.U.; García, J.V.; Fritis, M.C. Synthesis of a New Ent-Cyclozonarone Angular Analog, and Comparison of Its Cytotoxicity and Apoptotic Effects with Ent-Cyclozonarone. Molecules 2013, 18, 5517–5530. [Google Scholar] [CrossRef]
  361. Zhang, S.; Wang, X.; Hao, J.; Li, D.; Csuk, R.; Li, S. Expediently Scalable Synthesis and Antifungal Exploration of (+)-Yahazunol and Related Meroterpenoids. J. Nat. Prod. 2018, 81, 2010–2017. [Google Scholar] [CrossRef]
  362. Göhl, M.; Seifert, K. Total Synthesis of 3-Oxo- and 3β-Hydroxytauranin via Negishi Coupling of a Bis(Ortho-Oxy)-Functionalized Benzyl Chloride. Eur. J. Org. Chem. 2015, 2015, 6249–6258. [Google Scholar] [CrossRef]
  363. Parshikov, I.A.; Sutherland, J.B. The Use of Aspergillus niger Cultures for Biotransformation of Terpenoids. Process Biochem. 2014, 49, 2086–2100. [Google Scholar] [CrossRef]
  364. Muñoz, O.; Tapia-Merino, J.; Nevermann, W.; San-Martín, A. Phytochemistry and Biological Properties of Drimys Winteri Jr et g. Forster Var Chilensis (Dc) A. Bol. Latinoam. Y Del Caribe Plantas Med. Y Aromat. 2021, 20, 443–462. [Google Scholar] [CrossRef]
  365. Vlad, P.; Gorincioi, E.; Aricu, A.; Barba, A.; Manzocchi, A.; Santaniello, E. Asymmetric Dihydroxylation of Drim-7-En-11-Ol: Synthesis of Diastereomerically Pure Driman-7α,8α,11-Triol and Its Elaboration into Novel Chlorinated Norlabdanic Compounds. Tetrahedron Asymmetry 2010, 21, 2108–2116. [Google Scholar] [CrossRef]
  366. Ammon, H.P.T.; Muller, A.B. Forskolin: From an Ayurvedic Remedy to a Modern Agent. Planta Med. 1985, 51, 473–477. [Google Scholar] [CrossRef] [PubMed]
  367. Kreutner, W.; Chapman, R.W.; Gulbenkian, A.; Tozzi, S. Bronchodilator and Antiallergy Activity of Forskolin. Eur. J. Pharmacol. 1985, 111, 1–8. [Google Scholar] [CrossRef] [PubMed]
  368. Chang, J.; Hand, J.M.; Schwalm, S.; Dervinis, A.; Lewis, A.J. Bronchodilating Activity of Forskolin in Vitro and in Vivo. Eur. J. Pharmacol. 1984, 101, 271–274. [Google Scholar] [CrossRef] [PubMed]
  369. Burra, S.; Voora, V.; Rao, C.P.; Vijay Kumar, P.; Kancha, R.K.; David Krupadanam, G.L. Synthesis of Novel Forskolin Isoxazole Derivatives with Potent Anti-Cancer Activity against Breast Cancer Cell Lines. Bioorg. Med. Chem. Lett. 2017, 27, 4314–4318. [Google Scholar] [CrossRef]
  370. Cristóbal, I.; Garcia-Orti, L.; Cirauqui, C.; Alonso, M.M.; Calasanz, M.J.; Odero, M.D. PP2A Impaired Activity Is a Common Event in Acute Myeloid Leukemia and Its Activation by Forskolin Has a Potent Anti-Leukemic Effect. Leukemia 2011, 25, 606–614. [Google Scholar] [CrossRef]
  371. Seamon, K.B.; Daly, J.W.; Metzger, H.; de Souza, N.J.; Reden, J. Structure-Activity Relationships for Activation of Adenylate Cyclase by the Diterpene Forskolin and Its Derivatives. J. Med. Chem. 1983, 26, 436–439. [Google Scholar] [CrossRef]
  372. Marín, V.; Bart, B.; Cortez, N.; Jiménez, V.A.; Silva, V.; Leyton, O.; Cabrera-Pardo, J.R.; Schmidt, B.; Heydenreich, M.; Burgos, V.; et al. Drimane Sesquiterpene Aldehydes Control Candida Yeast Isolated from Candidemia in Chilean Patients. Int. J. Mol. Sci. 2022, 23, 11753. [Google Scholar] [CrossRef]
  373. Razmilic, I.; López, J.; Sierra, J.; Cortés, M. An Alternative Partial Synthesis of (-)-Polygodial. Synth. Commun. 1987, 17, 95–103. [Google Scholar] [CrossRef]
  374. Kubo, I.; Fujita, K.I.; Lee, S.H.; Ha, T.J. Antibacterial Activity of Polygodial. Phyther. Res. 2005, 19, 1013–1017. [Google Scholar] [CrossRef]
  375. Kubo, I.; Fujita, K.; Lee, S.H. Antifungal Mechanism of Polygodial. J. Agric. Food Chem. 2001, 49, 1607–1611. [Google Scholar] [CrossRef]
  376. Kang, R.; Helms, R.; Stout, M.J.; Jaber, H.; Chen, Z.; Nakatsu, T. Antimicrobial Activity of the Volatile Constituents of Perilla Frutescens and Its Synergistic Effects with Polygodial. J. Agric. Food Chem. 1992, 40, 2328–2330. [Google Scholar] [CrossRef]
  377. Moreno-Osorio, L.; Cortés, M.; Armstrong, V.; Bailén, M.; González-Coloma, A. Antifeedant Activity of Some Polygodial Derivatives. Z. fur Naturforsch.-Sect. C J. Biosci. 2008, 63, 215–220. [Google Scholar] [CrossRef] [PubMed]
  378. Mendes, G.L.; Santos, A.R.S.; Malheiros, A.; Cechinel Filho, V.; Yunes, R.A.; Calixto, J.B. Assessment of Mechanisms Involved in Antinociception Caused by Sesquiterpene Polygodial. J. Pharmacol. Exp. Ther. 2000, 292, 164–172. [Google Scholar] [PubMed]
  379. Da Cunha, F.M.; Fröde, T.S.; Mendes, G.L.; Malheiros, A.; Filho, V.C.; Yunes, R.A.; Calixto, J.B. Additional Evidence for the Anti-Inflammatory and Anti-Allergic Properties of the Sesquiterpene Polygodial. Life Sci. 2001, 70, 159–169. [Google Scholar] [CrossRef] [PubMed]
  380. Corrêa, D.S.; Tempone, A.G.; Reimão, J.Q.; Taniwaki, N.N.; Romoff, P.; Fávero, O.A.; Sartorelli, P.; Mecchi, M.C.; Lago, J.H.G. Anti-Leishmanial and Anti-Trypanosomal Potential of Polygodial Isolated from Stem Barks of Drimys brasiliensis Miers (Winteraceae). Parasitol. Res. 2011, 109, 231–236. [Google Scholar] [CrossRef]
  381. Cahill, P.L.; Kuhajek, J.M. Polygodial: A Contact Active Antifouling Biocide. Biofouling 2014, 30, 1035–1043. [Google Scholar] [CrossRef]
  382. Pongpiriyadacha, Y.; Matsuda, H.; Morikawa, T.; Asao, Y.; Yoshikawa, M. Protective Effects of Polygodial on Gastric Mucosal Lesions Induced by Necrotizing Agents in Rats and the Possible Mechanisms of Action. Biol. Pharm. Bull. 2003, 26, 651–657. [Google Scholar] [CrossRef]
  383. Urones, J.G.; Marcos, I.S.; Gómez Pérez, B.; Díez, D.; Lithgow, A.M.; Gómez, P.M.; Basabe, P.; Garrido, N.M. Chemistry of Zamoranic Acid. Part V Homochiral Semisyntheses of Active Drimanes: Pereniporin B, Polygodial and Warburganal. Tetrahedron 1994, 50, 10995–11012. [Google Scholar] [CrossRef]
  384. Gerard, P.J.; Ruf, L.D.; Perry, N.B.; Foster, L.B. Insecticidal Properties of the Terpenoids Polygodial, 9-Deoxymuzigadial and Azadirachtin. In Proceedings of the New Zealand Plant Protection Conference, Wellington, New Zealand, 11–13 August 1992; Volume 45, pp. 239–242. [Google Scholar]
  385. Pillai, A.P.; Dasari, S.; Patel, B.; Banerjee, S.; Kornienko, A.V.; Munirathinam, G. Polygodial: A Potential Sesquiterpene Dialdehyde for Castration-Resistant Prostate Cancer Treatment. Cancer Res. 2016, 76, 4804. [Google Scholar] [CrossRef]
  386. Martínez, A.R.; Rodríguez-García, I. Marine Puupehenone and Puupehedione: Synthesis and Future Perspectives. Mar. Drugs 2023, 21, 322. [Google Scholar] [CrossRef]
  387. Martínez-Poveda, B.; Quesada, A.R.; Medina, M.Á. Pleiotropic Role of Puupehenones in Biomedical Research. Mar. Drugs 2017, 15, 325. [Google Scholar] [CrossRef] [PubMed]
  388. Kohmoto, S.; McConnell, O.J.; Wright, A.; Koehn, F.; Thompson, W.; Lui, M.; Snader, K.M. Puupehenone, a Cytotoxic Metabolite from a Deep Water Marine Sponge, Stronglyophora Hartmani. J. Nat. Prod. 1987, 50, 336. [Google Scholar] [CrossRef]
  389. Barrero, A.F.; Alvarez-Manzaneda, E.J.; Chahboun, R.; Cortés, M.; Armstrong, V. Synthesis and Antitumor Activity of Puupehedione and Related Compounds. Tetrahedron 1999, 55, 15181–15208. [Google Scholar] [CrossRef]
  390. Xu, T.; Kuang, T.; Du, H.; Li, Q.; Feng, T.; Zhang, Y.; Fan, G. Magnoflorine: A Review of Its Pharmacology, Pharmacokinetics and Toxicity. Pharmacol. Res. 2020, 152, 104632. [Google Scholar] [CrossRef]
  391. Mirhadi, E.; Rezaee, M.; Malaekeh-Nikouei, B. Nano Strategies for Berberine Delivery, a Natural Alkaloid of Berberis. Biomed. Pharmacother. 2018, 104, 465–473. [Google Scholar] [CrossRef]
  392. Zhu, J.X.; Tang, D.; Feng, L.; Zheng, Z.G.; Wang, R.S.; Wu, A.G.; Duan, T.T.; He, B.; Zhu, Q. Development of Self-Microemulsifying Drug Delivery System for Oral Bioavailability Enhancement of Berberine Hydrochloride. Drug Dev. Ind. Pharm. 2013, 39, 499–506. [Google Scholar] [CrossRef] [PubMed]
  393. Rehman, K.U.; Gouda, M.; Zaman, U.; Tahir, K.; Khan, S.U.; Saeed, S.; Khojah, E.; El-Beltagy, A.; Zaky, A.A.; Naeem, M.; et al. Optimization of Platinum Nanoparticles (PtNPs) Synthesis by Acid Phosphatase Mediated Eco-Benign Combined with Photocatalytic and Bioactivity Assessments. Nanomaterials 2022, 12, 1079. [Google Scholar] [CrossRef] [PubMed]
  394. Gouda, M.; Lv, J.M.; Huang, Z.; Chen, J.C.; He, Y.; Li, X. Bioprobe-RNA-Seq-MicroRaman System for Deep Tracking of the Live Single-Cell Metabolic Pathway Chemometrics. Biosens. Bioelectron. 2024, 261, 116504. [Google Scholar] [CrossRef]
  395. Deng, X.; Yang, Z.; Chan, K.W.; Ismail, N.; Abu Bakar, M.Z. 5-Fluorouracil in Combination with Calcium Carbonate Nanoparticles Loaded with Antioxidant Thymoquinone against Colon Cancer: Synergistically Therapeutic Potential and Underlying Molecular Mechanism. Antioxidants 2024, 13, 1030. [Google Scholar] [CrossRef]
  396. Cosme, P.; Rodríguez, A.B.; Espino, J.; Garrido, M. Plant Phenolics: Bioavailability as a Key Determinant of Their Potential Health-Promoting Applications. Antioxidants 2020, 9, 1263. [Google Scholar] [CrossRef]
  397. Deng, X.; Yang, Z.; Chan, K.W.; Abu Bakar, M.Z. Exploring the Therapeutic Potential of 5-Fluorouracil-Loaded Calcium Carbonate Nanoparticles Combined with Natural Compound Thymoquinone for Colon Cancer Treatment. Pharmaceutics 2024, 16, 1011. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) Chemical structure of drimenol; (B) model of cylindrical bonds; (C) model of sticks; (D) model of ball and stick.
Figure 1. (A) Chemical structure of drimenol; (B) model of cylindrical bonds; (C) model of sticks; (D) model of ball and stick.
Plants 13 02492 g001
Figure 2. Biosynthesis pathways of drimenol.
Figure 2. Biosynthesis pathways of drimenol.
Plants 13 02492 g002
Figure 3. Chemical synthesis of drimenol.
Figure 3. Chemical synthesis of drimenol.
Plants 13 02492 g003
Figure 4. Antifungal activity of drimenol.
Figure 4. Antifungal activity of drimenol.
Plants 13 02492 g004
Figure 5. Proposed antifungal mechanisms of drimenol.
Figure 5. Proposed antifungal mechanisms of drimenol.
Plants 13 02492 g005
Figure 6. Antibacterial activity of drimenol.
Figure 6. Antibacterial activity of drimenol.
Plants 13 02492 g006
Figure 7. Anticancer mechanisms of drimenol.
Figure 7. Anticancer mechanisms of drimenol.
Plants 13 02492 g007
Table 1. Main plant sources of drimenol.
Table 1. Main plant sources of drimenol.
FamilyPlant SpeciesUsed PartContentsReferences
AmaranthaceaeGomphrena celosioidesLeaves1.62%[40]
AnemiaceaeAnemia tomentosa (Savigny) Sw. var. Aerial parts0.20%[41]
ApiaceaeFerula elaeochytris KorovinRoots0.96%[42]
AsparagaceaePolygonum odoratumLeaves0.39%[43]
Leaves0.39%[44]
AsteraceaeAchillea santolinaAerial parts2.33%[45]
Anaphalis triplinervisAerial parts0.60%[46]
Anthemis werneri L.\0.10%[47]
Artemisia parvifloraAerial parts4.31%[48]
Chrysanthemum morifolium Ramat.Roots0.85%[49]
Hetichrysum odoratissimumLeaves0.70%[50]
Ligularia fischeri TurczRoots0.09–1.33%[51]
CanellaceaeCanella winteranaLeaves\[52]
Leaves4.70%[53]
Capsicodendron dinisiiStem bark0.70%[54]
Cinnamodendron dinisiiLeaves0.20%[55]
Warburgia salutarisLeaves0.70%[56]
\\[57]
Warburgia ugandensisBark or other parts\[58]
Heartwood\[59]
CaprifoliaceaeValeriana angustifolia TauschSeeds\[60]
Valeriana celtica ssp. norica Vierh.Seeds\[60]
Valeriana hardwickii var. arnottianaRoots5.40%[61]
Valeriana officinalis var. sambucifoliaHairy roots\[62]
Valeriana phu L.Seeds\[60]
Valeriana salina PleijelSeeds\[60]
Valeriana sisymbriifolia Vahl\4.02–4.45%[63]
CistaceaeCistus salviifoliusAerial parts0.6–1.9%[64]
CupressaceaeCupressus sempervirensGround material0.37–0.4%[65]
Taiwania flousiana GaussenStem bark0.04%[66]
CyperaceaeCyperus leavigatusAerial parts0.71%[67]
FabaceaeAlhagi maurorumLeaves23.20%[68]
Stems0.60%[68]
Leaves23.20%[69]
Ononis Sicula GussAerial parts0.32%[70]
Tetrapleura TetrapteraLeaves0.70%[71]
FrullaniaceaeFrullania muscicola\\[72]
LamiaceaeSalvia limbataLeaves0.10%[73]
Scutellaria comosaRoots1.44%[74]
Sideritis cretica boiss\0.92%[75]
LauraceaeCinnamomum camphoraLeaves0.23%[76]
LepidoziaceaeBazzania fauriana\\[77]
MontiniaceaeKaliphora madagascariensis Leaves0.70%[78]
MoraceaeFicus elasticaLeaves1%[79]
Ficus polita Vahl\5.80%[80]
MyrtaceaeCalyptranthes concinnaLeaves2.60%[81]
Eucalyptuc obliquaLeaves6.97%[82]
Eucalyptus camaldulensis var. brevirostrisFruits12.35%[83]
Eucalyptus obliquaLeaves9.33%[84]
Eugenia calycina CambessLeaves0.65–0.69%[85]
Eugenia protentaLeaves0.1–0.7%[86]
Pimenta racemosaLeaves0.01–0.03%[87]
PinaceaePinus eldaricaBark13.20%[88]
PolygonaceaeCalligonum polygonoidesStems0.70%[89]
Roots29.19–29.65%[90]
Calligonum PolyoidesRoots29.42% [91]
Persicaria hydropiperFlowers\[92]
Polygonum acuminatumLeaves\[93]
Polygonum hydropiperLeaves\[94]
\\[95]
Whole plant7.26%[96]
Sprouts4.00%[97]
Polygonum hydropiperoides var. hydropiperoidesLeaves\[93]
Polygonum lapathifoliumLeaves\[93]
Polygonum persicariaLeaves\[93]
Polygonum punctatumLeaves\[93]
Vietnamese corianderLeaves0.03%[98]
RanunculaceaeClematis chinensis OsbeckRoots0.20%[99]
SapindaceaeKoelreuteria paniculataStem bark16.03–16.35%[100]
ScapaniaceaeDiplophyllum serrulatum\\[101]
TargioniaceaeTargionia hypophyllaGround material\[102]
VerbenaceaeStachytarpheta indicaRoots, shoots, and inflorescences0.30%[103]
WinteraceaeDrimys angustifolia MiersFresh leaves1.40%[104]
Stem bark26.20%[104]
Leaves1.20%[105]
Leaves1.60%[106]
Branch50%[106]
Drimys brasiliensis MiersLeaves and stem barks4.40%[107]
Leaves9.96%[108]
Fresh leaves0.4–11.3%[104]
Dried leaves0.10%[104]
Stem bark3.7–14.6%[104]
Unripe fruits0.20%[104]
Leaves0.80%[105]
Green leaves9.30%[109]
Dried leaves11.60%[109]
Drimys granadensisUnripe fruits10%[110]
Unripe fruits10%[111]
Leaves4.30%[112]
Drimys winteriLeaves\[113]
\\[114]
Fresh fruits0.4–0.6%[115]
Mild dried (green) fruits0.49–0.81%[115]
Strong dried (brown) fruits0.61%[115]
\3.30%[116]
\12.10%[117]
Stem bark 2–5.8%[118]
\\[119]
Tasmannia lanceolataLeaves\[120]
ZingiberaceaeAlpinia malaccensisRhizomes0.10%[121]
Curcuma longa\\[122]
GingerFresh ginger 0.51%[123]
Zingiber roseumSeeds1.30%[124]
Table 2. Derivatives of drimenol and their biological activities or applications.
Table 2. Derivatives of drimenol and their biological activities or applications.
Name of CompoundBiological Activities/ApplicationReferences
1.3 Dioxans\[331]
11-Aminodrim-7-eneAntifungal activity[332,333]
3β/-HydroxydrimanesInsecticidal activity[334,335]
3β-Hydroxy-7α,8α-epoxydrimenol\[336]
3β-Hydroxydrimenol\[336]
6α-Hydroxydrimenol\[336]
8-Epiambreinolide\[337]
8-EpipuupehedioneAntitumor and cytotoxic activities[338,339]
8β(H)-Drimane\[340]
9-EpiambroxPerfumes[341,342]
AlbicanolCytotoxic, fish antifeedant, antifungal, anti-inflammatory, antiaging, and antioxidant activities, and antagonistic activity against heavy metal toxicity[161,343,344,345,346,347,348,349,350]
AmbrafuranPerfumes[351]
AmbraoxidePerfumes and flavoring agents in food[352,353]
AmbroxPerfumes[354,355]
IsoambroxPerfumes[354,356]
CyclozonaroneCytotoxic, cytostatic, anticancer, antileishmanial, and feeding-deterrent activities[343,357,358,359,360,361,362]
Drimenyl acetateAntifungal activity[149,363,364]
ForskolinAnti-leukemic, antiproliferative, bronchodilator, anti-allergy, and hypotensive activities, and cardiac adenylate cyclase activation[365,366,367,368,369,370,371]
Monoaldehyde drimenalFungistatic and fungicidal activities[372]
PolygodialAntibacterial, antifungal, antifeedant, antimicrobial, antinociception, anti-inflammatory, anti-allergic, anti-leishmanial, anti-trypanosomal, antifouling biocide, gastromucosal protection, cytotoxic, and insecticidal activities[373,374,375,376,377,378,379,380,381,382,383,384,385]
PuupehenoneAntiangiogenic, antioxidant, antimicrobial, cytotoxic, antitumor, immunomodulatory, antimalarial, antiviral, antibiotic, antiatherosclerotic, and antitubercular activities[386,387,388]
PuupehedioneAntitumor, cytotoxic, antimicrobial, and antifungal activities[386,389]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, Z.; Chan, K.W.; Abu Bakar, M.Z.; Deng, X. Unveiling Drimenol: A Phytochemical with Multifaceted Bioactivities. Plants 2024, 13, 2492. https://doi.org/10.3390/plants13172492

AMA Style

Yang Z, Chan KW, Abu Bakar MZ, Deng X. Unveiling Drimenol: A Phytochemical with Multifaceted Bioactivities. Plants. 2024; 13(17):2492. https://doi.org/10.3390/plants13172492

Chicago/Turabian Style

Yang, Zhongming, Kim Wei Chan, Md Zuki Abu Bakar, and Xi Deng. 2024. "Unveiling Drimenol: A Phytochemical with Multifaceted Bioactivities" Plants 13, no. 17: 2492. https://doi.org/10.3390/plants13172492

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