Skip to main content
PLOS One logoLink to PLOS One
. 2024 Nov 7;19(11):e0310770. doi: 10.1371/journal.pone.0310770

Chemical profiling and cytotoxicity screening of agarwood essential oil (Aquilaria sinensis) in brine shrimp nauplii and cancer cell lines

Sook Wah Chan 1,2, Valizadeh Lakeh Mahmoud 1, Xin Wang 1, Ming-Li Teoh 1,3, Kar Min Loh 4, Chun Howe Ng 4, Won Fen Wong 4, Chung Yeng Looi 1,5,*
Editor: Marcello Iriti6
PMCID: PMC11542896  PMID: 39509364

Abstract

Agarwood essential oil (AEO) has gained attention from healthcare industries due to its numerous pharmacological properties. However, a comprehensive understanding of the chemical composition and its cytotoxic property is lacking. The objective of this study was to investigate the chemical profile as well as the cytotoxic concentration range of AEO derived from Aquilaria sinensis agarwood. Gas chromatography-mass spectrometry (GC-MS) was employed to identify the AEO components. Results showed that sesquiterpenes and sesquiterpenoids constitute 95.85% of the AEO. Among the major compounds identified are allo-aromadendrene (13.04%), dihydro-eudesmol (8.81%), α-eudesmol (8.48%), bulnesol (7.63%), τ-cadinol (4.95%), dehydrofukinone (3.83%), valerenol (3.54%), cis-nerolidol (2.75%), agarospirol (2.72%), dehydrojinkoh-eremol (2.53%), selina-3,11-dien-9-al (2.36%), guaiol (2.12%) and caryophyllene oxide (2.0%). The presence of volatile quality marker compounds such as 10-epi-ϒ-eudesmol, aromadendrane, β-agarofuran, α-agarofuran, γ-eudesmol, agarospirol and guaiol, with no contaminants detected, indicates that the extracted AEO is of high purity. Interestingly, the AEO displayed moderate to high toxicity in brine shrimp lethality test (BLST). All studied tumor cell lines (MDA-MB-231, HepG2, B16F10) exhibited varying degrees of sensitivity to AEO, which resulted in time and dose-dependent reduction of cell proliferation. Moreover, flow cytometry analysis revealed that AEO could induce apoptosis in treated HepG2 cells. Our findings showed that AEO contains bioactive components that may be exploited in future studies for the development of anti-cancer therapeutics.

Introduction

Cancer is a pathological condition caused by uncontrolled cell proliferation, which is a hallmark of oncogenesis and progression [1]. According to the latest issue of the International Agency for Research on Cancer (IARC) in 2020, breast cancer in women has now become the most frequently diagnosed cancer, surpassing lung cancer. Approximately 2.3 million new cases, accounting for 11.7% of diagnoses, were reported, followed by lung (11.4%), colorectal (10.0%), prostate (7.3%) and stomach cancer (5.6%). Despite this shift, lung cancer remains the leading cause of cancer-related deaths, with an estimated 1.8 million fatalities (18%). Colorectal cancer stands at 9.4%, followed by liver (8.3%), stomach (7.7%) and breast cancer (6.9%) [2]. Among men, lung cancer is the leading cause of mortality [3]. The primary cause of cancer-related death in women is breast cancer [3]. On the other hand, acute lymphoblastic leukemia is the top hematological cancer in children and adolescents, representing 20% of all cancers diagnosed in persons aged < 20 years [4].

Depending on the type of cancer and how advanced it is, the standard treatments are surgery, chemotherapy and radiation. However, resistance to radiotherapy and chemotherapy may result in treatment failure or even cancer recurrence [5]. Thus, the development of new therapeutic agents to overcome these pressing issues is necessary. Chemotherapy is an important component of modern cancer therapy, which has evolved over centuries. While the historical use of plants in medicine dates back to the dawn of civilization, the World Health Organization (WHO) reports that people in Asia and some parts of Africa widely rely on herbal remedies for disease management [6]. At present, a significant amount of modern pharmaceutical drugs, particularly those used in cancer therapy, are derived from natural ingredients. Phytochemicals like vincristine, camptothecin, and paclitaxel have emerged as important as well as reliable chemotherapeutic agents in cancer treatment [7]. This long-standing reliance on natural sources highlights the ongoing pursuit of efficacious therapeutic agents sourced from plants.

Agarwood, also known as eaglewood, oud, aloeswood, and gaharu is a highly valuable fragrant heartwood used for a wide range of non-timber purposes in perfume, cosmetics industry, medicinal preparations as well as for religious ritual purposes [8]. The genus Aquilaria or simply known as agarwood from the family Thymelaeaceae, has different species including crassna, malaccensis, and sinensis (Fig 1). Agarwood is well known for its resinous secretion against injuries and this tree is commonly found in Southeast Asian countries and Southern China. Agarwood is rich in bioactive compounds, including sesquiterpenes, chromones, flavonoids, benzophenones, diterpenoids, triterpenoids and lignans, which show myriad pharmacological effects [911]. A number of studies reported that these chemicals extracted from Aquilaria wood exhibit acetylcholinesterase inhibition, anti-fungal, anti-bacterial, anti-inflammatory, anti-cancer and analgesic actions [1214].

Fig 1. Cultivated agarwood (A. sinensis) tree.

Fig 1

Previous study indicates that plant-derived essential oil possesses anti-tumor potential against leukemia as well as cancers of the mouth, breast, lung, prostate, liver, and colon [15]. Study revealed that specific components of essential oils might increase the cytotoxic action of chemotherapy drugs, such as docetaxel, paclitaxel, and 5-fluorouracil, on different cancer cell lines. This suggests that the dosage of these drugs may be reduced while still having the same effect [16]. Till date, the cytotoxic and chemical profile of AEO derived from A. sinensis is understudied, which hampered its feasibility to be used for potential medication or therapeutic agents. Hence, the present study was conducted to characterize the chemical constituents of AEO via GC-MS and to evaluate its cytotoxic profile using brine shrimp (Artemia salina) and cancer cell lines (MDA-MB-231, B16F10, HepG2).

Materials and methods

Plants material

Agarwood, the resinous heartwood from A. sinensis was harvested from a cultivated agarwood plantation in Tangkak, Johor, located at a longitude of 2.2951° N and latitude of 102.5756° E, which is registered with the Malaysian Timber Industry Board (MTIB) (Fig 2A). Taxonomic identification was performed by Forest Research Institute of Malaysia (FRIM). Voucher specimen (202312–1) is deposited at the Lab 2, Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya.

Fig 2.

Fig 2

Illustrations of (A) Cultivated agarwood (A. sinensis) plantation; (B) Agarwood powder; (C) Agarwood powder in distillation tank; (D) AEO collected at the bottom of the separation funnel; (E) Final AEO.

Extraction of agarwood essential oil

The agarwood was separated from the white wood, crushed into smaller pieces, and air-dried under the sun for 1–2 days until completely dry. Subsequently, the dried agarwood dust was ground into powder (Fig 2B) and stored in a bag at room temperature for further extraction. Before the extraction process, 20 kg of agarwood powder underwent a 14-day soaking period in filtered water to facilitate the release of AEO. The soaked powder and 200 L of filtered water were then introduced into a 250 L stainless steel tank of a novel designed industrial distiller (Lian Sin Enterprise Sdn. Bhd., Selangor, Malaysia) (Fig 2C) at a sample:water ratio of 1:10 for optimal yield and quality. The mixture was heated to a boil at 100°C, generating steam containing both water and AEO. The steam was condensed and collected in an essential oil separation funnel. The excess condensed water was recycled back to the distillation tank during the extraction process. After 120 h of hydro-distillation, the water will be removed, followed by the retrieval of the floating AEO through the bottom of the separation funnel, as illustrated in Fig 2D. The AEO was obtained as a pale yellow liquid (Fig 2E).

Chemical profiling by GC-MS

The volatile constituents of the sample were analyzed using a gas chromatography system, the Agilent 7890B (Agilent Technologies Inc., Santa Clara, CA, USA) coupled with an Agilent 5977A quadrupole mass spectrometer and Agilent DB-1ms capillary column (30 m × 0.25 mm x 0.25 μm film thickness) based on the method adopted from Eissa et al. [17] with modifications. GC was performed using the following conditions: autosampler injection (1 μL); injection temperature (230°C), carrier gas (helium) flow rate (1 mL/min); programmed oven temperature, 70°C held for 3 min, ramped from 70 to 230°C at a rate of 3°C/min, held for 3 min. The detector temperature was 240°C. The detected peaks from the total ion chromatography (TIC) and mass chromatograms were identified based on the National Institute of Standards and Technology (NIST) 2020 mass spectral library. The RI value of each component was determined relative to the retention times (RT) of a series of C7-C20 n-alkanes with linear interpolation on the Agilent DB-1ms column. The peak area (%) of each individual component of the essential oil was expressed as the percentage of the peak area relative to the total peak area.

Brine shrimp lethality test (BSLT)

The brine shrimp lethality test was performed according to the protocol of Asaduzzaman et al. [18]. The method estimates in vivo lethality in a simple zoological organism, using nauplii of the brine shrimp Artemia salina. Artificial seawater (ASW) was prepared by mixing 7.5 g of a commercially available salt mixture with 500 mL of distilled water according to the instructions. A. salina eggs were added to a small commercial nauplius hatching tank and incubated in an ASW under a halogen lamp providing direct light and heat. The hatched shrimp was allowed to mature for 24 h as nauplii. Hatched shrimps were attracted to light, and brine shrimp eggless nauplii were collected from illuminated sections of the aquarium. Ten nauplii were counted visually in the stem of a graduated Pasteur pipette against an illuminated background and transferred to a test tube. A stock solution (50 mg/mL) is prepared by dissolving the AEO in saline containing 15% propylene glycol and 1% Tween 80. This can be used to serially dilute it to produce a range of concentrations 31.25, 62.5, 125, 250, 500, 1000 μg/mL. Solvent alone was used as negative control. Testing was performed with three replicates for each dosage and 10 nauplii per replicate. Each test tube was illuminated for 24, 48 and 72 h. Survivors were counted visually by two independent enumerators. The LC50 values were determined with probit analysis using the Statistical Package for the Social Sciences (SPSS) application.

Cell culture

Human breast adenocarcinoma (MDA-MB-231), hepatocellular (HepG2) and murine melanoma (B16F10) cell lines were purchased from American Type Culture Collection (ATCC). The cells were grown in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% FBS and 1% penicillin-streptomycin and incubated at 37°C, 5% CO2 as described previously [19].

Cell viability assay

MTT assay was performed to measure cell viability, according to previous protocol [20]. The cells were seeded in 100 μL of complete culture media, at a density of 5 × 104 per mL in 96 well plates. The seeded cells were incubated for 12 h at 37°C and 5% CO2. The cells were then treated with AEO at different concentrations including 0.975, 1.95, 3.91, 7.81, 15.625, 31.25, 62.5, 125, 250, 500 μg/mL. A total of 20 μL of MTT solution was added to each well after the cells were treated with AEO for the indicated time point. After 3 h of incubation, the media was discarded and 100 μL of DMSO was added to each well. The absorption of produced formazan was measured at 570 nm using a BioTek EPOCH 2 microplate reader. The cell viability was calculated as following:

Cellviability(%)=(ODSample/ODControl)×100

Apoptosis assay

To quantify the percentages of apoptosis versus necrosis cell death, cells were seeded at 1×105 per ml on 25 cm2 flask overnight before treated with control solvent (15% propylene glycol and 1% Tween 80) and AEO at various concentrations (50 μg/mL and 100 μg/mL) for 16 h. Determination of apoptotic/necrotic cells by fluorescent staining was done as described previously [21]. Briefly, cells were incubated with FITC-annexin V and propidium iodide (PI) (BD Biosciences) in a binding buffer for 15 minutes in the dark. Stained cells were immediately subjected to flow cytometry analyses using Novocyte flow cytometer system (Agilent).

Statistical analysis

The statistical analysis was performed using GraphPad Prism 9.0. Two-way analysis of variance (ANOVA) was performed to determine the significant differences between means at a 95% confidence interval (p < 0.05).

Results and discussion

Yield, smells, and appearance of AEO

The agarwood used in this study originated from A. sinensis cultivated in Tangkak, Johor (Fig 2A). By adopting the ITS2, matK, and rbcL barcode sequences, the agarwood sample was identified as A. sinensis. The AEO was extracted from agarwood (A. sinensis) powder using hydro-distillation (HD) (Fig 2C). HD is the most conventional method employed by industries for the extraction of essential oil from agarwood using water, making it a relatively safe and environmentally-friendly process [22,23]. The AEO obtained had a clear, brown hue with a slightly golden tint and emitted a strong characteristic woody scent, accompanied by hints of bitter, sweet, and spicy aromas (Fig 2E).

The average yield of AEO through HD for 120 h at 100°C was reported as 1 g/kg of dried agarwood powder, equivalent to 0.1% (v/w). The yield of AEO in this experiment was similar to that of resinous heartwood and agarwood leaves reported in the previous literature. Sulaiman et al. [24] reported that the maximum extraction yield of AEO from A. malaccensis was 0.18% using the HD method for 72 h at 100°C. Other research done by Wu et al. [25] investigated the extraction of leaf aromatic oils from A. sinensis through HD, with a reported yield of 0.1% at 100°C for 5 h. Despite its practicability and safety, HD remains a time-consuming and low efficiency extraction method. In this work, soaking agarwood powder as a pre-treatment was performed to enhance the swelling of the cell wall of agarwood, thereby releasing more AEO [26].

Several new extraction methods, including solvent extraction (SE), enzyme-assisted extraction (EAE), microwave-assisted extraction (MAE), ultrasound-assisted extraction (UAE), and supercritical fluid extraction (SFE) have been explored for extracting AEO [2729]. In Radzi et al. [30] study, agarwood that underwent microwave pre-treatment demonstrated a threefold increase in yield compared to conventional HD. On the other hand, SFE exhibited a significant increase in AEO yield, rising from 0.061% (obtained by HD) to 0.317% [31]. Nevertheless, these extraction methods are still not widely utilized in the agarwood industry due to the safety concerns (SE), high costs (EAE, SFE), and upscaling issues (MAE, UAE) [23].

Chemical composition analysis

Profiling of chemical constituents in plant species is an important step for the investigation of their medical effects. The chemical constituents identified from the medicinal plants are the potential sources for drug development. As AEO is valued for its distinctive and pleasant fragrance, GC-MS is extensively employed to identify its compounds and assess its quality [32]. In this study, the AEO was eluted and analyzed by GC-MS using DB-1ms capillary column. The identification of the individual components of the AEO was based on the matching of their mass spectra with the NIST 2020 library. Fig 3 displays the GC-MS spectra of AEO, revealing the presence of 59 chemical compounds. The compounds were numbered in accordance with corresponding peaks. Visually, small peaks could be observed in large quantities, corresponding to the compounds that could not be determined (Fig 3). Chemical compounds showed up before retention time (RT) of 25.3 min were mainly sesquiterpenes and sesquiterpenoids. Most of the sesquiterpenoids and other compounds were detected between 25.3 and 40.0 min.

Fig 3. Chromatogram of the analysis by GC for AEO obtained from A. sinensis.

Fig 3

The chemical compounds found in AEO are listed in Table 1 according to their sequence of elution on a DB-1ms capillary column. A summary is provided in Table 1 for the distribution of chemical classes, retention time, % contents of the constituent components, as well as CAS number. Overall, the total percentage of sesquiterpenoids was the highest at 77.36%, followed by sesquiterpenes (18.49%) and other compounds (1.41%). This finding is in good agreement with previous studies which found sesquiterpenoids as the major component in AEO [3335]. However, the amount of sesquiterpenoids varied depending on the agarwood species, origin, and extraction methods. Other studies found varying amounts of sesquiterpenoids, ranging from 63.87% (India, A. malaccensis) to 67.40% (Malaysia, A. malaccensis) and 49.91% (Thailand, A. crassna) [36]. The findings in Tian et al. [37] showed that sesquiterpenoids were present in AEO (A. sinensis) at 68.68% through steam distillation, whereas the supercritical extraction method yielded sesquiterpenoids at 23.78%.

Table 1. Chemical compounds identified in AEO obtained from A. sinensis.

No Compounds Molecular formula Retention indices (RI) Retention time (min) Peak area (%) Phytochemical groups CAS no
1 4-Phenyl-2-butanone C10H12O 1207 12.841 0.53 Other compounds 2550-26-7
2 α-Gurjunene C15H24 1397 20.132 0.10 Sesquiterpene 489-40-7
3 Aromadendrene C15H24 1439 21.667 0.10 Sesquiterpene 489-39-4
4 allo-Aromadendrene C15H24 1463 22.585 13.04 Sesquiterpene 25246-27-9
5 α-Curcumene C15H22 1468 22.79 0.44 Sesquiterpene 644-30-4
6 γ-Selinene C15H24 1474 23.032 0.28 Sesquiterpene 515-17-3
7 β-Agarofuran C15H24O 1477 23.146 0.35 Sesquiterpenoid NA
8 β-Vetispirene C15H22 1480 23.259 0.52 Sesquiterpene 28908-27-2
9 Valencene C15H24 1484 23.436 0.56 Sesquiterpene 4630-07-3
10 α-Selinene C15H24 1488 23.574 0.55 Sesquiterpene 473-13-2
11 Dihydro-β-agarofuran C15H26O 1489 23.642 0.46 Sesquiterpenoid 5956-09-2
12 δ-Guaiene C15H24 1495 23.874 0.54 Sesquiterpene 3691-11-0
13 γ-Cadinene C15H24 1500 24.08 0.26 Sesquiterpene 39029-41-9
14 α-Elemene C15H24 1508 24.35 0.28 Sesquiterpene 5951-67-7
15 δ-Cadinene C15H24 1511 24.461 0.12 Sesquiterpene 483-76-1
16 Kessane C15H26O 1515 24.598 0.17 Sesquiterpenoid 3321-66-2
17 Selina-3.7(11)-diene C15H24 1524 24.925 0.34 Sesquiterpene 6813-21-4
18 α-Calacorene C15H20 1527 25.051 0.30 Sesquiterpene 21391-99-1
19 Dehydro-aromadendrene C15H22 1530 25.151 0.29 Sesquiterpene NA
20 α-Agarofuran C15H24O 1535 25.339 0.60 Sesquiterpenoid 5956-12-7
21 cis-Nerolidol C15H26O 1547 25.779 2.75 Sesquiterpenoid 142-50-7
22 nor-Ketoagarofuran C14H22O2 1560 26.264 1.75 Sesquiterpenoid 5986-25-4
23 β-Vetivenene C15H22 1571 26.668 0.77 Sesquiterpene 27840-40-0
24 Caryophyllene oxide C15H24O 1582 27.115 2.00 Sesquiterpenoid 1139-30-6
25 Tetradecanal C14H28O 1593 27.521 0.54 Other compounds 124-25-4
26 Guaiol C15H26O 1597 27.697 2.12 Sesquiterpenoid 489-86-1
27 10-epi-γ-Eudesmol C15H26O 1601 27.823 3.07 Sesquiterpenoid 15051-81-7
28 γ-Eudesmol C15H26O 1612 28.208 1.42 Sesquiterpenoid 1209-71-8
29 Agarospirol C15H26O 1617 28.379 2.72 Sesquiterpenoid 1460-73-7
30 Hinesol C15H26O 1620 28.475 1.41 Sesquiterpenoid 23811-08-7
31 τ-Cadinol C15H26O 1625 28.676 4.95 Sesquiterpenoid 5937-11-1
32 β-Eudesmol C15H26O 1629 28.801 1.14 Sesquiterpenoid 473-15-4
33 α-Eudesmol C15H26O 1634 28.971 8.48 Sesquiterpenoid 473-16-5
34 Jinkoh-eremol C15H26O 1640 29.174 1.21 Sesquiterpenoid 94201-17-9
35 Dihydro-eudesmol C15H28O 1651 29.58 8.81 Sesquiterpenoid 6770-16-7
36 Kusunol C15H26O 1655 29.716 1.07 Sesquiterpenoid 20489-45-6
37 Bulnesol C15H26O 1664 30.044 7.63 Sesquiterpenoid 22451-73-6
38 Dehydrojinkoh-eremol C15H24O 1671 30.297 2.53 Sesquiterpenoid NA
39 Cyperotundone C15H22O 1677 30.503 1.87 Sesquiterpenoid 3466-15-7
40 epi-α-Bisabolol C15H26O 1685 30.808 1.06 Sesquiterpenoid 78148-59-1
41 α-Bisabolol C15H26O 1688 30.942 0.50 Sesquiterpenoid 515-69-5
42 Selina-3,1 l-dien-9-one C15H22O 1697 31.273 1.24 Sesquiterpenoid NA
43 Rotundone C15H22O 1701 31.419 0.88 Sesquiterpenoid 18374-76-0
44 Valerenol C15H24O 1708 31.63 3.54 Sesquiterpenoid 101628-22-2
45 Selina-3,11-dien-9-ol C15H24O 1715 31.87 1.66 Sesquiterpenoid NA
46 Selina-4,11-dien-14-oic acid C15H22O2 1721 32.082 0.51 Sesquiterpenoid NA
47 α-Costol C15H22O 1727 32.259 0.73 Sesquiterpenoid 65018-15-7
48 Selina-3,11-dien-9-al C15H22O 1737 32.603 2.36 Sesquiterpenoid NA
49 9,l l-Eremophiladien-8-one C15H22O 1744 32.857 0.72 Sesquiterpenoid NA
50 Aristolone C15H22O 1749 33.023 0.70 Sesquiterpenoid 6831-17-0
51 Guaia-1 (10),11-dien-9-one C15H22O 1752 33.113 0.21 Sesquiterpenoid NA
52 Dehydrofukinone C15H22O 1769 33.693 3.83 Sesquiterpenoid 19598-45-9
53 Sinenofuranol C14H24O2 1783 34.196 0.86 Sesquiterpenoid NA
54 Dihydrokaranone C15H22O 1791 34.481 0.24 Sesquiterpenoid NA
55 Guaia-1(10),11-dien-15-oic acid C15H22O2 1815 35.278 0.46 Sesquiterpenoid NA
56 oxo-Agarospirol C15H24O2 1838 35.995 0.83 Sesquiterpenoid NA
57 Methyl palmitate C17H34O2 1899 38.016 0.18 Other compounds 112-39-0
58 Dihydrocolurnellarin C15H22O2 1907 38.262 0.52 Sesquiterpenoid 66873-38-9
59 1,5-Diphenyl-3-pentanone C17H18O 1964 40.03 0.16 Other compounds 5396-91-8

(NA—Not available).

The most abundant compound identified in AEO was allo-aromadendrene (13.04%), followed by dihydro-eudesmol (8.81%), α-eudesmol (8.48%), bulnesol (7.63%), τ-cadinol (4.95%) dehydrofukinone (3.83%), valerenol (3.54%), 10-epi-γ-eudesmol (3.07%), cis-nerolidol (2.75%), agarospirol (2.72%), dehydrojinkoh-eremol (2.53%), selina-3,11-dien-9-al (2.36%), guaiol (2.12%), caryophyllene oxide (2.00%), cyperotundone (1.87%), nor-Ketoagarofuran (1.75%), selina-3,11-dien-9-ol (1.66%), γ-eudesmol (1.42%), hinesol (1.41%), selina-3,1 l-dien-9-one (1.24%), jinkoh-eremol (1.21%), β-eudesmol (1.14%), kusunol (1.07%), and epi-α-Bisabolol (1.06%), all of which except allo-aromadendrene (sesquiterpenes) belong to sesquiterpenoids according to their structural characteristics (Table 1). Major compounds detected in this study were consistent with those found in previously published works [13,32,38,39]. Other common compounds that were detected belong to aromatic compounds. One study reported that the predominant compounds in the essential oil of A. sinensis were sesquiterpenes/sesquiterpenoids (63.4% and 58%, for natural and fungal inoculated agarwood, respectively) [39]. The differences in the chemical composition can arise due to several factors such as geographic origin, age of the tree, hydrodistillation parameters, induction methods and post-harvest handling. Most of these compounds exhibit various pharmacological activities [38]. According to literature, allo-aromadendrene contributes to the characteristic woody odor of AEO, while agarospirol gives out the smell of spicy, peppery, and woody [4042]. While most essential oils are volatile, AEO is relatively stable at room temperature. A study done by Syameera et al. [43] showed that the AEO is thermally stable at temperature of 80°C at 5-min and 10-min time points, without causing major loss in its terpene compounds, including monoterpenes, diterpenes sesquiterpenes, and sesquiterpenoids.

Different studies have highlighted different chemical constituents as the marker compounds for classifying the AEO. Ismail et al. [44] concluded 10-epi-ϒ-eudesmol, aromadendrane, β-agarofuran, α-agarofuran and γ-eudesmol as the marker compounds in high quality agarwood oils derived from A. malaccensis. The presence of four characteristic compounds (benzylacetone, agarospirol, aristolene, and guaiol) has been used as quality indicators for AEO (A. sinensis) in a study conducted by Tian et al. [38]. The majority of these compounds, except benzylacetone and aristolene, were present in substantial quantities in the AEO. In addition, there are no contaminants, namely dibutyl phthalate (DBP) and di(ethylhexyl) phthalate (DEHP), detected in the AEO, further ensuring that the studied sample is a high-value fragrant material. Nevertheless, as of now, there is no globally recognized standard for agarwood oil (AEO). The current grading system for AEO, which considers factors such as country-of-origin, aroma, duration of fragrance, color, thickness, and density, heavily relies on consumer perception and preference [45].

In addition, 2-(2-phenylethyl)chromone is the major aromatic constituent in agarwood, which has been extensively reported [32,46]. However, no chromone compounds, as well as their derivatives, were detected in the present study. They are rarely identified by GC-MS but are more commonly detected via LC-MS/MS [32,46].

Brine shrimp lethality test (BSLT)

The BSLT method has been widely used by researchers to assess the cytotoxic concentration range and toxicity of essential oils. The LC50 values obtained from brine shrimp lethality bioassay for AEO were 111.35, 66.93 and 54.05 μg/mL, for 24, 48 and 72 h, respectively (Table 2). The LC50 values were assessed at 95% confidence using probit analysis. The valorized essential oils should be considered toxic or medium to highly toxic by comparison to Meyer’s and Clarkson’s toxicity index as summarized in Table 2. The solvent group showed little toxicity towards brine shrimp with a survival rate of more than 90%. In contrast, AEO exhibited >90% mortality in the concentration range of 250–1000 μg/mL (Fig 4).

Table 2. LC50 value of AEO on brine shrimp lethality test (BSLT).

Time-point LC50 (μg/mL) 95% confidence interval Toxicity class (Meyer/Clarkson)
24 h 111.35 101.15–121.54 Toxic/Medium Toxic
48 h 66.93 53.77–87.42 Toxic/ Highly Toxic
72 h 54.05 45.60–66.49 Toxic/ Highly Toxic

Fig 4. Evaluation of acute toxicity effect of AEO on brine shrimp at different treatment time-points.

Fig 4

Elumba et al. [47], Del Socorro et al. [48] suggested that some of the tested extracts with LC50 below 100 µg/mL which are categorized as toxic, does not always indicate its danger or out-right toxicity toward humans, but may also suggest a potential anti-tumor or anti-cancer activity [49]. To the best of our knowledge, this is the first report on the toxicity effect of AEO in brine shrimp. We hypothesized that the lethality of brine shrimp larvae may be linked to the presence of bioactive compounds in the AEO, which can interfere with biochemical and physiological processes [50]. The toxic compounds can enter the digestive tracts of brine shrimp larvae, where the substances absorbed have the ability to interact with targets, such as lipids, enzymes, cell membranes, and nucleic acids, affecting the body’s processes and ultimately resulting in death [51,52].

Cell line viability test (MTT)

Table 3 and Fig 5A–5C show the growth inhibitory values determined from the dose-response curves of AEO and anti-cancer drugs used in cell lines MDA-MB-231, B16F10 and HepG2. The strongest anti-proliferative activity of AEO was observed against B16F10 cell line (IC50 48.9 ± 3.1 μg/mL), followed by HepG2 cells (IC50 56.2 ± 3.9 μg/mL) and MDA-MB-231 cell line (IC50 61.3 ± 3.2 μg/mL) after 24 h treatment. As shown in Fig 6, morphological analysis revealed that AEO-induced cell rounding, loss of cell-cell contact and cell detachment, an appearance normally seen in cells undergoing apoptosis. Nevertheless, AEO treatment at 48 and 72 h also showed growth inhibitory potential on all tested cancer cell-lines. These results suggest that AEO induced a time and dose-dependent inhibition of cell proliferation, although each cell-line showed different sensitivity, in accordance with the determined IC50. In addition, the anti-proliferative effect of the referent chemo-drug, doxorubicin is stated in Table 3.

Table 3. IC50 values of AEO and doxorubicin (Dox) on B16F10, MDA-MB-231 and HepG2 cell lines at different treatment time-points.


Cell line
AEO IC50 (μg/mL) Dox IC50 (μg/mL)
24 h 48 h 72 h 24 h 48 h 72 h
B16F10 48.9 ± 3.1 38.7 ± 5.0 30.5 ± 2.6 <10 <10 <5
MDA-MB-231 61.3 ± 3.2 54.4 ± 4.8 39.4 ± 3.7 <10 <10 <5
HepG2 56.2 ± 3.9 41.6 ± 2.4 36.8 ± 1.1 <10 <10 <5

Fig 5. Evaluation of effect of AEO on cancer cell lines by MTT assay.

Fig 5

Percentages of viable cells of (A) B16F10; (B) MDA-MB-231; (C) HepG2 at 24 h time-point after AEO treatments. Each experiment was repeated for a minimum of 3 times and plotted as bar graphs with error bars. Two-way ANOVA was conducted and the p value was calculated between control and AEO treated samples, where, *: p value < 0.05.

Fig 6. Morphology changes of B16F10, MDA-MB-231, HepG2 cells after AEO treatment (61.5 μg/mL) for 24 h.

Fig 6

Treated cells were rounded up and cell to cell adhesion was lost. Images showing 40 x magnification. Black bar in each image represents 100 μm.

At present, there is no study on AEO extracted from agarwood used against melanoma and hepatic cancer. Previous study reported that A. crassna essential oil mixture showed significant anti-proliferation activity against human cancer cell lines such as colorectal HCT 116 (IC50 28 µg/mL), pancreatic PANC-1 (IC50 32 µg/mL), prostate PC3 (IC50 79 µg/mL) and breast MCF-7 (IC50 110 µg/mL) [53]. The same group of researchers also reported that A. crassna essential oil exhibited potent cytotoxic activity against human pancreatic cancer MIA PaCa-2 cells with an IC50 (11 ± 2.18 μg/mL) [54]. They showed that cell migration and the colony formation ability of MIA PaCa-2 was effectively inhibited at 5 and 10 μg/mL, respectively. Furthermore, they discovered that AEO exhibited significant inhibition of HCT 116 colorectal cancer cell-induced subcutaneous tumors in nude mice [54]. It is interesting to note that these studies reported previously were using AEO derived from A crassna. Another scientific investigation found that AEO possesses anti-proliferative activity towards MCF-7 breast cancer cells with IC50 observed at 44 μg/mL [55]. However, the AEO used in this study was purchased directly from a Malaysian market, and it was unclear which type of agarwood they were using to produce AEO.

Recently, Zhang et al. [56] reported the cytotoxic activity of AEO derived from steam distillation (SD) and fungal fermentation-assisted SD (FFSD) on four different cancer-lines (human breast cancer MDA-MB-435, mouse gastric cancer MFC, human glioblastoma SNB19, mouse colon cancer MC38). They postulated that α-costal, aristolone, 10-epi-γ-eudesmol, γ-eudesmol, agarospirol, hinesol, guaiol, β-santalol, and 6-isopropenyl-4,8a-dimethyl-1,2,3,5,6,7,8,8a-octahydro- naphthalen-2-ol were antitumor ingredients of AEO. Interestingly, some of these compounds (aristolone, 10-epi-γ-eudesmol, γ-eudesmol, agarospirol, hinesol, guaiol) can be found in our AEO.

Besides sesquiterpenes, chromones, such as 2-(2-hydroxy-2-phenylethyl)chromone, demonstrated the ability to inhibit tumor development at non-cytotoxic concentrations, displaying half-inhibitory values ranging from 25 to 38 µM against different tumor cell lines [57]. Two novel chromone derivatives, namely 7-hydroxy-2-[2-(3’-methoxy-4’-hydroxyphenyl)-ethyl]chromone and 6,7-dimethoxy-2-[2-(3’-hydroxyphenyl)-ethyl]chromone, isolated from agarwood ethanol extract, exhibited weak cytotoxic activities (IC50: 18.82–37.95 mg/mL) against SMMC-7721, MGC-803, and OV-90 cell lines [58].

Apoptosis assessment

To investigate the anti-proliferative activity of AEO, treated HepG2 cells were stained with Annexin V and PI to examine the mechanism of cell death (necrosis and/or apoptosis). The populations of cells residing in the Annexin V+/PI- and the Annexin V+/PI+ quadrants were classified as early and late apoptotic cells, respectively. The Annexin V−/PI—and the Annexin V−/PI+ quadrants were designated as viable cells and necrotic cells, respectively. Control solvent treated cells showed the percentages of the apoptotic cells as < 5% after 16 h of incubation (Fig 7A). AEO-treated HepG2 samples showed varying degrees of percentages (20%-78%) of apoptotic cells (Fig 7B and 7C) (the percentages of the early apoptotic cells and the late apoptotic cells combined). Our results showed that AEO-induced apoptotic cell death in a dose-dependent manner. Recent evidence showed that activation of the apoptosis pathway is an effective strategy for cancer treatment [59]. Nevertheless, researchers have resorted to the use of essential oils as natural bioactive compounds that can elicit sensitive growth inhibition and death in cancer cells in the hope to find a more effective anticancer and chemopreventive methods [6062].

Fig 7. Effect of AEO on the induction of apoptosis.

Fig 7

HepG2 cells were cultured with (A) control solvent; (B) 50 μg/mL of AEO; (C) 100 μg/mL of AEO for 16 h. Treated cells were then stained with Annexin V and PI, followed by flow cytometry analysis.

The anticancer properties of essential oils can be attributed to their bioactive compounds and their effects on specific pathways involved in cancer cell growth and proliferation [63]. For example, bioactive compounds such as terpenes, phenolics, and other aromatic molecules may induce apoptosis, inhibit angiogenesis, or suppress the expression of genes involved in cancer progression. One study showed the growth inhibitory effect of AEO is attributed to the presence of sesquiterpene such as β-caryophyllene [53]. This compound has been shown to inhibit colorectal cancer cell proliferation, clonality, migration, invasion, and spheroid formation [53]. Further investigation revealed that the cell death mechanism induced by β-caryophyllene is related to the apoptotic properties such as DNA fragmentation and mitochondrial pathways. Moreover, studies show that volatility can be managed through appropriate formulation and application methods, allowing for controlled delivery and efficacy in anticancer treatments [64]. Research is ongoing to improve the delivery and stability of essential oils for therapeutic purposes [65]. Techniques such as encapsulation or nanoemulsion have been explored to enhance the stability and bioavailability of essential oil components [66]. Nevertheless, essential oils are often used in combination with other therapies (complementary medicine) rather than as standalone treatments [65]. These approaches can enhance their potential benefits while minimizing concerns about volatility.

Conclusion

The AEO was successfully extracted from agarwood (A. sinensis) powder using HD. The presence of key quality marker compounds and absence of contaminants suggest that the extracted AEO is of high purity. This study demonstrated for the first time the cytotoxic effects of AEO on brine shrimps as well as melanoma (B16F10), hepatocarcinoma (HepG2) and breast adenocarcinoma (MDA-MB-231) cancer cell lines. We showed that the anti-proliferative activity of the AEO is time and dose-dependent. Furthermore, our results indicated that the cytotoxic or growth inhibitory effect may be attributable to the presence of sesquiterpene and sesquiterpenoid, which is capable of inducing apoptotic cell death. Nevertheless, we also observed a positive correlation between BSLT and in vitro testing of cancer cell lines. Future studies should be directed to elucidate the cytotoxic agent(s) as well as the anti-cancer mechanism of AEO in order to enhance understanding and position the essential oil as a potential cancer remedy.

Supporting information

S1 File. Chemicals and reagents used in this study.

Dulbecco’s Modified Eagle’s medium (DMEM), trypsin, phosphate buffer saline (PBS) and fetal bovine serum (FBS) were purchased from Gibco, UK. Propylene glycol (PEG), standard drug doxorubicin (Dox) and 3-(4,5-dimethylthiazol-2-yl)-2-5-diphenyltetrazolium bromide (MTT) reagent were procured from Sigma-Aldrich, USA. Tween 80 was obtained from Acros Organics, USA.

(DOCX)

pone.0310770.s001.docx (13.3KB, docx)
S2 File. Link to dataset.

Toxicity data of AEO on brine shrimp at different treatment time-points. https://osf.io/pfwba/?view_only=f5b419159a024c97b463ef6d4c4b9626.

(DOCX)

pone.0310770.s002.docx (11.8KB, docx)
S3 File. Link to dataset.

Cell viability of B16F10 melanoma, MDA-MB-231 breast, HepG2 hepatocarcinoma cell-lines after 24h treatment and the IC50 data for 24h, 48h, 72h treatment. https://osf.io/tcm86/?view_only=77247d8859594c8cb9d53787db03661a.

(DOCX)

pone.0310770.s003.docx (11.9KB, docx)
S4 File. Link to data analysis.

Statistical analysis of MTT data. https://osf.io/hw59q/?view_only=dde0aa11171847e58fb183460c93dd88.

(DOCX)

pone.0310770.s004.docx (11.8KB, docx)
S5 File. Link to dataset.

Flow cytometry results of AEO treated HepG2 cells. https://osf.io/pywq7/?view_only=0d6a63a87f784b37b4e7aedfa6faf263.

(DOCX)

pone.0310770.s005.docx (11.9KB, docx)
S6 File

(DOCX)

pone.0310770.s006.docx (14.5KB, docx)
S7 File

(PDF)

pone.0310770.s007.pdf (294.5KB, pdf)

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

This research was supported by the Ministry of Higher Education (MOHE) through the Fundamental Research Grant Scheme (FRGS/1/2024/STG01/TAYLOR/02/2). This research is also funded by the industrial grant (Project Code: DADVANCE/2023/SBS/001). The funders did not play any role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. In addition, we would like to acknowledge the provision of research materials by staff of DAdvance Agarwood Solutions Sdn. Bhd. in particular Mr. Liew Kien Meng, Mr. Lim Kok Seong, Mr. Tan Chen Wee, and Mr. Gan Yong Hao.

References

  • 1.Choudhuri S, Chanderbhan R, Mattia A. Chapter 20-carcinogenesis: mechanisms and models. Vet. Toxicol. 2018; 339–354. 10.1016/B978-0-12-811410-0.00020-9. [DOI] [Google Scholar]
  • 2.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71(3): 209–249. doi: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
  • 3.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68(6): 394–424. doi: 10.3322/caac.21492 . [DOI] [PubMed] [Google Scholar]
  • 4.Ward E, DeSantis C, Robbins A, Kohlet B., Jemal A. Childhood and adolescent cancer statistics. CA Cancer J Clin. 2014; 64(2): 83–103. doi: 10.3322/caac.21219 . [DOI] [PubMed] [Google Scholar]
  • 5.Zhang CL, Huang T, Wu BL, He WX, Liu D. Stem cells in cancer therapy: Opportunities and challenges. Oncotarget. 2017; 8(43): 75756–75766. doi: 10.18632/oncotarget.20798 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.WHO. Traditional medicine has a long history of contributing to conventional medicine and continues to hold promise. 2023. Available from: https://www.who.int/news-room/feature-stories/detail/traditional-medicine-has-a-long-history-of-contributing-to-conventional-medicine-and-continues-to-hold-promise. [Google Scholar]
  • 7.Choudhari AS, Mandave PC, Deshpande M, Ranjekar P, Prakash O. Phytochemicals in cancer treatment: from preclinical studies to clinical practice. Front. Pharmacol. 2020; 10: 1614. doi: 10.3389/fphar.2019.01614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Liu Y, Chen HQ, Yang Y, Zhang Z, Wei JH, Meng H, et al. Whole-tree agarwood-inducing technique: an efficient novel technique for producing high-quality agarwood in cultivated Aquilaria sinensis trees. Molecules. 2013; 18(3): 3086–3106. doi: 10.3390/molecules18033086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hashim YZHY, Kerr PG, Abbas P, Salleh HM. Aquilaria spp. (agarwood) as source of health beneficial compounds: A review of traditional use, phytochemistry and pharmacology. J. Ethnopharmacol. 2016; 189: 331–360. doi: 10.1016/j.jep.2016.06.055 . [DOI] [PubMed] [Google Scholar]
  • 10.Kristanti AN, Tanjung M, Aminah NS. Review: Secondary metabolites of Aquilaria, a Thymelaeaceae genus. Mini Rev Org Chem. 2018; 15(1): 36–55. doi: 10.2174/1570193X14666170721143041 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yuan HW, Zhao JP, Liu YB, Qiu YX, Xie QL, Li MJ, et al. Advance in studies on chemical constituents, pharmacology and quality control of Aquilaria sinensis. Digit. Chin. Med. 2018; 1(4): 316–330. 10.1016/S2589-3777(19)30057-6. [DOI] [Google Scholar]
  • 12.Chen ST, Rao YK. An overview of agarwood, phytochemical constituents, pharmacological activities, and analyses. Tradit. Med. Res. 2022; 3(1): 1–71. 10.35702/Trad.10008. [DOI] [Google Scholar]
  • 13.Gogoi R, Sarma N, Begum T., Chanda SK, Lekhak HS, Sastry GH, et al. Agarwood (Aquilaria malaccensis L.) a quality fragrant and medicinally significant plant based essential oil with pharmacological potentials and genotoxicity. Ind Crops Prod. 2023; 197: 116535. 10.1016/j.indcrop.2023.116535. [DOI] [Google Scholar]
  • 14.Ma S, Qiao MJ, Fu YL, Wei PL, Li YJ, Liu ZG. Comparative analysis of biological activity of artificial and wild agarwood. Forests. 2021; 12(11): 1532. 10.3390/f12111532. [DOI] [Google Scholar]
  • 15.Gautam N., Mantha AK, Mittal S. Essential oils and their constituents as anticancer agents: a mechanistic view. Biomed Res. Int. 2014; 15410610. doi: 10.1155/2014/154106 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Carnesecchi S, Schneider Y, Ceraline J, Duranton B, Gosse F, Seiler N, et al. Geraniol, a component of plant essential oils, inhibits growth and polyamine biosynthesis in human colon cancer cells. J. Pharmacol. Exp. Ther. 2001; 298(1): 197–200. . [PubMed] [Google Scholar]
  • 17.Eissa MA, Hashim YZHY, El-Kersh DM, Abd-Azziz SSS Salleh HM, Isa MLM, et al. Metabolite profiling of Aquilaria malaccensis leaf extract using liquid chromatography-q-tof-mass spectrometry and investigation of its potential antilipoxygenase activity in-vitro. Processes. 2020; 8(2): 202. 10.3390/pr8020202. [DOI] [Google Scholar]
  • 18.Asaduzzaman M, Sohel Rana M, Raqibul Hasan SM, Monir Hossain M, Das N. Cytotoxic (brine shrimp lethality bioassay) and antioxidant investigation of Barringtonia acutangula (L.). Int J Pharm Sci Res. 2015; 6(8): 1179–1185. [Google Scholar]
  • 19.Hayyan M, Looi CY, Hayyan A, Wong WF, Hashim MA. In vitro and in vivo toxicity profiling of ammonium-based deep eutectic solvents. PloS one. 2015; 10(2): e0117934. 10.1371/journal.pone.0117934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Looi CY, Arya A, Cheah FK, Muharram B, Leong KH, Mohamad K, et al. Induction of apoptosis in human breast cancer cells via caspase pathway by vernodalin isolated from Centratherum anthelminticum (L.) seeds. PloS one. 2013; 8(2): e56643. doi: 10.1371/journal.pone.0056643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dai X, Wang L, Deivasigamni A, Looi CY, Karthikeyan C, Trivedi P, et al. A novel benzimidazole derivative, MBIC inhibits tumor growth and promotes apoptosis via activation of ROS-dependent JNK signaling pathway in hepatocellular carcinoma. Oncotarget. 2017; 8(8): 12831–12842. doi: 10.18632/oncotarget.14606 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hashin YZHY, Jamil MAM, Jamal P, Zainurin AAA, Azziz SSSA. Hydrodistillation and soxhlet extraction of agarwood leaf extract from Aquilaria malaccensis. Mal. J. Fund. Appl. Sci. 2019; 15(6): 842–846. 10.11113/mjfas.v15n6.1589. [DOI] [Google Scholar]
  • 23.Samadi M, Zainal Adibin Z, Yoshida H, Yunus R, Awang Biak DR. Towards higher oil yield and quality of essential oil extracted from Aquilaria malaccensis wood via the subcritical technique. Molecules. 2020; 25(17): 3872. doi: 10.3390/molecules25173872 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sulaiman N. Idayu MI, Ramlan AZ, Nur Fashya M, Nor Farahiyah AN, Mailina J, et al. Effects of extraction methods on yield and chemical compounds of Gaharu (Aquilaria malaccensis). J. Trop. For. Sci. 2015; 27(3): 413–419. [Google Scholar]
  • 25.Wu P, Sun L, Qi SY, Zeng TX, Hou ZY, Yang Y, et al. Extraction process, chemical profile, and biological activity of aromatic oil from agarwood leaf (Aquilaria sinensis) by supercritical carbon dioxide extraction. J. CO2 Util. 2023; 77: 102615. 10.1016/j.jcou.2023.102615. [DOI] [Google Scholar]
  • 26.Jok VA, Radzi NC, Hamid KHK. Agarwood oil yield as a result of changes in cell morphology due to soaking process. Procedia Soc Behav Sci. 2015; 195: 2443–2450. 10.1016/j.sbspro.2015.06.387. [DOI] [Google Scholar]
  • 27.Rahim AFA, Jamaluddin N, Kadri A, Idris SS, Rahman NA. Microwave-assisted hydrodistillation of Aquilaria subintegra. J. Adv. Res. Fluid Mech. Therm. Sci. 2020; 72(2): 1–12. 10.37934/arfmts.72.2.112. [DOI] [Google Scholar]
  • 28.Kusuma HS, Putri DKY, Triesty I, Mahfud M. Comparison of microwave hydrodistillation and solvent-free microwave extraction for extraction of agarwood oil. Chiang Mai Univ. J. Nat. Sci. 2019; 46(4): 741–755. [Google Scholar]
  • 29.Thuy DTT, Tuyen TT, Chien NQ, Thuy TTT, Bich HT, Toan TQ. Comparative study on volatile compounds of agarwood from khanh hoa province extracted by different methods. Vietnam J. Sci. Technol. 2018; 56(4A): 266–272. [Google Scholar]
  • 30.Radzi NC, Kasim FA, Effect of microwave pretreatment on gaharu essential oil using hydrodistillation method. Indones. J Chem. 2020; 20(4): 960–966. 10.22146/ijc.43191. [DOI] [Google Scholar]
  • 31.Pham TH, Dang QL, Le TT, Vu HD. Supercritical CO2 extraction and characterization of agarwood extract derived-from vietnamese Aquilaria crassna woodchips. Chiang Mai J. Sci. 2020; 47(5): 1001–1011. [Google Scholar]
  • 32.Yan TT, Yang S, Chen Y, Wang Q, Li GY. Chemical profiles of cultivated agarwood induced by different techniques. Molecules. 2019; 24(10): 1990; doi: 10.3390/molecules24101990 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Latib EHA, Najib MS, Che Mohd CMA, Tajuddin SN. Analysis of different quality agarwood oil (Aquilaria malaccensis) and sensory study. J. Telecommun. Electron. Comput. Eng. 2018; 10(1–3): 57–61. [Google Scholar]
  • 34.Latib EHA, Huq AKMM, Zareen S, Tajuddin SN. Gas chromatography mass spectrometry couple with quadrupole time-of-flight (GC-QTOF MS) as a powerful tool for profiling of oxygenated sesquiterpenes in agarwood oil. Arab. J. Chem. 2023; 16(9): 105025. 10.1016/j.arabjc.2023.105025. [DOI] [Google Scholar]
  • 35.Tan CS, Isa NM, Ismail I, Zainal Z. Agarwood induction: current developments and future perspectives. Front. Plant Sci. 2019; 10(122). doi: 10.3389/fpls.2019.00122 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Deep K, Tajuddin SN. King of scent–agarwood. Perfumer & Flavorist. 2019; 44(3): 42–56. [Google Scholar]
  • 37.Tian CP. Song YL, Xu HT, Niu SQ, Wu ZH, Shen LQ. Composition analysis, antioxidative and antibacterial activities comparison of agarwood oils extracted by supercritical and steam distillation. China J Chin Mater Med. 2019; 44(18): 4000–4008. doi: 10.19540/j.cnki.cjcmm.20190629.302 . [DOI] [PubMed] [Google Scholar]
  • 38.Tian CP, Yao XD, Lu JH, Shen LQ, Wu AQ, GC–MS fingerprints of essential oils from agarwood grown in wild and artificial environments. Trees. 2021; 35: 2105–2117. 10.1007/s00468-021-02177-w. [DOI] [Google Scholar]
  • 39.Zhang Z, Han X-M, Wei J-H Xue J, Yang Y, Liang L, Li X-J, et al. Compositions and antifungal activities of essential oils from agarwood of Aquilaria sinensis (Lour.) Gilg induced by Lasiodiplodia theobromae (Pat.) Griffon. & Maubl. J Braz Chem Soc. 2014; 25(20–26). https://doi.org.10.5935/0103-5053.20130263. [Google Scholar]
  • 40.Chung TY, Eiserich JP, Shibamoto T. Volatile compounds isolated from edible Korean chamchwi (Aster scaber Thunb). J. Agric. Food Chem. 1993; 41(10): 1693–1697. 10.1021/jf00034a033. [DOI] [Google Scholar]
  • 41.Ishihara M, Tsuneya T, Uneyama K. Components of the agarwood smoke on heating. J. Essent. Oil Res. 1993; 5(4): 419–423. 10.1080/10412905.1993.9698252. [DOI] [Google Scholar]
  • 42.Ishihara M, Tsuneya T, Uneyama K. Components of the volatile concentrate of agarwood. J. Essent. Oil Res. 1993; 5(3): 283–289. 10.1080/10412905.1993.9698221. [DOI] [Google Scholar]
  • 43.Syameera NA, Kaewdaungdee S, Tajuddin SN, Tanee T, Sudmoon R, Chaveerach A et al. Effects of heat treatment on the chemical composition, antioxidant activity, and toxicity of agarwood oil. J. King Saud Univ. Sci. 2024; 36(4): 103141. 10.1016/j.jksus.2024.103141. [DOI] [Google Scholar]
  • 44.Ismail N, Nor Azah MA, Jamil M, Rahiman MHF, Tajuddin SN, Taib MN. Analysis of high quality agarwood oil chemical compounds by means of spme/gc-ms and z-score technique. Malays. J. Anal. Sci. 2013; 17(3): 403–413. [Google Scholar]
  • 45.Antonopoulou M, Compton J, Perry LS, Al-Mubarak R. The trade and use of agarwood (oudh) in the United Arab Emirates. TRAFFIC Southeast Asia; 2010. [Google Scholar]
  • 46.Kim Ngan TT, Thu Thuy DT, Tuyen TT, Inh CT, Bich HT, Long PQ, et al. Chemical components of agarwood (Aquilaria crassna) essential oils grown in various regions of Asia. Asian Journal of Chemistry. 2020; 32(1): 36–40. 10.14233/ajchem.2020.22177. [DOI] [Google Scholar]
  • 47.Elumba Z, Teves F, Madamba MRS. DNA-binding and cytotoxic activities of supercritical-CO2 extracts of Ganoderma lucidum (Curt.:Fr.) P. Karst. collected from the wild of Bukidnon province, Philippines. Int. Res. J. Biol. Sci. 2013; 2(3): 62–68. [Google Scholar]
  • 48.Del Socorro MML, Bendoy CP, Dacayana CML. Cytotoxic effects of betel vine, Piper betle linn. leaf extracts using Artemia salina leach (brine shrimp lethality assay). J. Multidiscip. Stud. 2014; 3(1): 100–111. 10.7828/jmds.v3i1.629. [DOI] [Google Scholar]
  • 49.Moshi MJ, Innocent E, Magadula JJ, Otieno DF, Weisheit A, Mbabazi PK. et al. Brine shrimp toxicity of some plants used as traditional medicines in Kagera region, north western Tanzania. Tanzan J Health Res. 2010; 12(1): 63–67. doi: 10.4314/thrb.v12i1.56287 . [DOI] [PubMed] [Google Scholar]
  • 50.Sharififar F, Assadipour A, Moshafi MH, Alishahi F, Mahmoudvand H. Bioassay screening of the essential oil and various extracts of Nigella sativa L. seeds using brine shrimp toxicity assay. Herb. Med. J. 2017; 2(1): 26–31. 10.22087/HMJ.V1I2.578. [DOI] [Google Scholar]
  • 51.Arif MZ, Nik Zainuddin NAS, Zakaria IS, Wan Abdul Wahab WNA, Sul’ain MD. Phytochemical screening and toxicological evaluation of Pyrrosia piloselloides extracts. Int Med J. 2018; 25(3): 177–80. [Google Scholar]
  • 52.Jamil S, Khan RA, Afroz S, Ahmed S. Phytochemistry, brine shrimp lethality and mice acute oral toxicity studies on seed extracts of Vernonia anthelmintica. Pak J Pharm Sci. 2016; 29(6): 2053–2057. . [PubMed] [Google Scholar]
  • 53.Dahham SS. Tabana YM, Iqbal MA, Ahamed MB, Ezzat MO, Majid AS, et al. The anticancer, antioxidant and antimicrobial properties of the sesquiterpene β-caryophyllene from the essential oil of Aquilaria crassna. Molecules. 2015; 20(7): 11808–11829. 10.3390/molecules200711808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dahham SS, Ahmed Hassan LE, Khadeer Ahamed MB, Abdul Majid AS, Abdul Majid AMS, Zulkepli NN. In vivo toxicity and antitumor activity of essential oils extract from agarwood (Aquilaria crassna). BMC Complement Altern Med. 2016; 16(236): 1–11 doi: 10.1186/s12906-016-1210-1 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hashim YZHY, Phirdaous A, Azura A. Screening of anticancer activity from agarwood essential oil. Pharmacognosy Res. 2014; 6(3): 191–194. doi: 10.4103/0974-8490.132593 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang YL, Wu SH, Zhang BZ, Zhou X, Zhou WP, Zhang WM et al. Determination of antitumor active ingredients in agarwood essential oil by gas chromatography-mass spectrometry (GC-MS) and grey relational analysis. Anal. Lett. 2024; 1–23. 10.1080/00032719.2024.2325570. [DOI] [Google Scholar]
  • 57.Suzuki A, Miyake K, Saito Y, Rasyid FA, Tokuda H, Takeuchi M. et al. Phenylethylchromones with in vitro antitumor promoting activity from Aquilaria filaria. Planta Med. 2017; 83(3–04): 300–305. doi: 10.1055/s-0042-110858 . [DOI] [PubMed] [Google Scholar]
  • 58.Liu YY, Chen DL, Yu ZX, Wang CH, Feng J, Meng Y, et al. New 2-(2-phenylethyl) chromone derivatives from agarwood and their inhibitory effects on tumor cells. Nat Prod Res. 2020; 34(12): 1721–1727. doi: 10.1080/14786419.2018.1528591 . [DOI] [PubMed] [Google Scholar]
  • 59.Ferreira CG, Epping M, Kruyt FA, Giaccone G. Apoptosis target of cancer therapy. Clin Cancer Res. 2002; 8: 2024–2034. . [PubMed] [Google Scholar]
  • 60.Cha JD, Jeong MR, Kim HY, Lee JC, Lee KY. MAPK activation is necessary to the apoptotic death of KB cells induced by the essential oil isolated from Artemisia iwayomogi. J. Ethnopharmacol. 2009; 123: 308–314. doi: 10.1016/j.jep.2009.03.002 . [DOI] [PubMed] [Google Scholar]
  • 61.Sylvestre M, Pichette A, Longtin A, Nagau F, Legault J. Essential oil analysis and anticancer activity of leaf essential oil of Croton flavens L. from Guadeloupe. J. Ethnopharmacol. 2006; 103: 99–102. doi: 10.1016/j.jep.2005.07.011 . [DOI] [PubMed] [Google Scholar]
  • 62.Ahani N, Sangtarash MH, Alipour Eskandani M, Houshmand M. Zataria multiflora Boiss. essential oil induce apoptosis in two human colon cancer cell lines (HCT116 & SW48). Iran J Public Health. 2020; 49(4):753–762. . [PMC free article] [PubMed] [Google Scholar]
  • 63.Blowman K, Magalhães M, Lemos MFL, Cabral C, Pires IM. Anticancer properties of essential oils and other natural products. Evid Based Complement Alternat Med. 2018; 2018: 3149362. doi: 10.1155/2018/3149362 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Severino P, Andreani T, Chaud MV, Benites CI, Pinho SC, Souto EB. Essential oils as active ingredients of lipid nanocarriers for chemotherapeutic use. Curr Pharm Biotechnol. 2015; 16(4): 365–70. doi: 10.2174/1389201016666150206111253 . [DOI] [PubMed] [Google Scholar]
  • 65.Sharma M, Grewal K, Jandrotia R, Batish DR, Singh HP, Kohli RK. Essential oils as anticancer agents: Potential role in malignancies, drug delivery mechanisms, and immune system enhancement. Biomed Pharmacother. 2022; 146: 112514. doi: 10.1016/j.biopha.2021.112514 . [DOI] [PubMed] [Google Scholar]
  • 66.Jampilek J, Kralova K. Anticancer applications of essential oils formulated into lipid-based delivery nanosystems. Pharmaceutics. 2022; 14(12): 2681. doi: 10.3390/pharmaceutics14122681 . [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

S1 File. Chemicals and reagents used in this study.

Dulbecco’s Modified Eagle’s medium (DMEM), trypsin, phosphate buffer saline (PBS) and fetal bovine serum (FBS) were purchased from Gibco, UK. Propylene glycol (PEG), standard drug doxorubicin (Dox) and 3-(4,5-dimethylthiazol-2-yl)-2-5-diphenyltetrazolium bromide (MTT) reagent were procured from Sigma-Aldrich, USA. Tween 80 was obtained from Acros Organics, USA.

(DOCX)

pone.0310770.s001.docx (13.3KB, docx)
S2 File. Link to dataset.

Toxicity data of AEO on brine shrimp at different treatment time-points. https://osf.io/pfwba/?view_only=f5b419159a024c97b463ef6d4c4b9626.

(DOCX)

pone.0310770.s002.docx (11.8KB, docx)
S3 File. Link to dataset.

Cell viability of B16F10 melanoma, MDA-MB-231 breast, HepG2 hepatocarcinoma cell-lines after 24h treatment and the IC50 data for 24h, 48h, 72h treatment. https://osf.io/tcm86/?view_only=77247d8859594c8cb9d53787db03661a.

(DOCX)

pone.0310770.s003.docx (11.9KB, docx)
S4 File. Link to data analysis.

Statistical analysis of MTT data. https://osf.io/hw59q/?view_only=dde0aa11171847e58fb183460c93dd88.

(DOCX)

pone.0310770.s004.docx (11.8KB, docx)
S5 File. Link to dataset.

Flow cytometry results of AEO treated HepG2 cells. https://osf.io/pywq7/?view_only=0d6a63a87f784b37b4e7aedfa6faf263.

(DOCX)

pone.0310770.s005.docx (11.9KB, docx)
S6 File

(DOCX)

pone.0310770.s006.docx (14.5KB, docx)
S7 File

(PDF)

pone.0310770.s007.pdf (294.5KB, pdf)

Data Availability Statement

All relevant data are within the manuscript and its Supporting Information files.


Articles from PLOS ONE are provided here courtesy of PLOS

RESOURCES