Next Article in Journal
Mono-(Ni, Au) and Bimetallic (Ni-Au) Nanoparticles-Loaded ZnAlO Mixed Oxides as Sunlight-Driven Photocatalysts for Environmental Remediation
Previous Article in Journal
Gastrointestinal Digestion Impact on Phenolics and Bioactivity of Tannat Grape Pomace Biscuits
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro and In Vivo Efficacy of the Essential Oil from the Leaves of Annona amazonica R.E. Fries (Annonaceae) Against Liver Cancer

by
Maria V. L. de Castro
1,†,
Milena C. F. de Lima
2,†,
Gabriela A. da C. Barbosa
1,
Sabrine G. Carvalho
1,
Amanda M. R. M. Coelho
1,
Luciano de S. Santos
1,
Valdenizia R. Silva
1,
Rosane B. Dias
1,3,
Milena B. P. Soares
1,4,
Emmanoel V. Costa
2,5,* and
Daniel P. Bezerra
1,*
1
Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
2
Department of Chemistry, Institute of Exact Sciences, Federal University of Amazonas (UFAM), Manaus 69080-900, AM, Brazil
3
Department of Biological Sciences, State University of Feira de Santana, Feira de Santana 44036-900, BA, Brazil
4
SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador 41650-010, BA, Brazil
5
Postgraduate Program in Chemistry, Institute of Exact Sciences, Federal University of Amazonas (UFAM), Manaus 69080-900, AM, Brazil
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(15), 3248; https://doi.org/10.3390/molecules30153248 (registering DOI)
Submission received: 25 June 2025 / Revised: 28 July 2025 / Accepted: 29 July 2025 / Published: 2 August 2025
(This article belongs to the Special Issue Advances and Opportunities of Natural Products in Drug Discovery)

Abstract

Annona amazonica R.E. Fries (synonyms Annona amazonica var. lancifolia R.E. Fries), popularly known in Brazil as “envireira”, is a tropical tree belonging to the Annonaceae family and is traditionally used as a food source. In this work, the in vitro and in vivo anti-liver cancer effects of essential oil (EO) from A. amazonica leaves were investigated for the first time. The chemical composition of the EO was evaluated via GC–MS and GC–FID. The alamar blue assay was used to evaluate the cytotoxicity of EOs against different cancerous and noncancerous cell lines. Cell cycle analyses, YO-PRO-1/PI staining, and rhodamine 123 staining were performed via flow cytometry in HepG2 cells treated with EO. The in vivo antitumor activity of EO was evaluated in NSG mice that were xenografted with HepG2 cells and treated with EO at a dose of 60 mg/kg. The major constituents (>5%) of the EO were (E)-caryophyllene (32.01%), 1,8-cineole (13.93%), α-copaene (7.77%), α-humulene (7.15%), and α-pinene (5.13%). EO increased apoptosis and proportionally decreased the number of viable HepG2 cells. The induction of DNA fragmentation and cell shrinkage together with a significant reduction in the ΔΨm in EO-treated HepG2 cells confirmed that EO can induce apoptosis. A significant 39.2% inhibition of tumor growth in vivo was detected in EO-treated animals. These data indicate the anti-liver cancer potential of EO from A. amazonica leaves.

1. Introduction

Liver cancer is a global health issue, with hepatocellular carcinoma being the most common type of primary liver cancer. It is the sixth most diagnosed cancer, with an estimated 865,269 new cases, and the third leading cause of cancer-related deaths, accounting for approximately 757,948 deaths worldwide in 2022 [1]. In advanced stages of the disease, systemic therapy is commonly employed. This includes combinations of tyrosine kinase inhibitors (e.g., sorafenib, regorafenib, lenvatinib, and cabozantinib) and immune checkpoint inhibitors, such as anti-PD-1 agents (e.g., nivolumab and pembrolizumab), anti-PD-L1 agents (e.g., atezolizumab), and/or anti-CTLA-4 agents (e.g., ipilimumab) [2,3,4].
In contrast to recent advances in liver cancer treatment, the American Cancer Society reported that from 2014 to 2020, the 5-year relative survival rates for patients with liver cancer were 37% for those with localized tumors, 13% for those with regional tumors, and 3% for those with distant tumors, resulting in an overall survival rate of only 22% [5]. These data reinforce the need to explore new therapeutic options for the treatment of liver cancer.
Essential oils (EOs) are complex mixtures of volatile molecules isolated from aromatic plants that exhibit promising cytotoxic effects against various types of cancer, including breast, liver, colon, and lung cancer cell lines, through diverse mechanisms of action [6,7,8,9,10]. Owing to their natural origin and multitarget effects, EOs are promising candidates for complementary cancer treatments.
Annona amazonica R.E. Fries (synonyms Annona amazonica var. lancifolia R.E. Fries), popularly known in Brazil as “envireira”, is a tropical tree belonging to the Annonaceae family. It is a tree that is 20 to 25 m tall and is distinguished from other species by having very small flowers, with congenital outer and inner petals and glaucous fruits. This species is native to a region stretching from Costa Rica to northern Brazil, where it is traditionally used as a food source [11,12,13].
Previous phytochemical investigations of A. amazonica have reported the presence of cyanogenic compounds in its leaves, seeds, and fruits [14]. Pinheiro et al. [15] successfully isolated acanthoic acid in high yield from the stem of A. amazonica and demonstrated its trypanocidal activity. More recently, Alcantara et al. [16] identified (E)-caryophyllene and linalool as the major constituents of EOs extracted from the leaves and branches of A. amazonica, respectively. However, the antitumor potential of this species remains unexplored.
This work aimed to determine the chemical composition of A. amazonica leaf EO and investigate its anticancer potential. In the present study, the in vitro and in vivo anti-liver cancer effects of the EO derived from A. amazonica leaves were reported for the first time.

2. Results and Discussion

2.1. Chemical Constituents of the EO from the Leaves of A. amazonica

The EO obtained from the dried leaves of A. amazonica by hydrodistillation exhibited a light green color, a pronounced aroma, and an average yield of 0.18 ± 0.02% relative to the dry weight of the analyzed samples. Figure S1 illustrates the experimental workflow performed. The chemical composition of the oil was characterized via gas chromatography–mass spectrometry (GC–MS) and gas chromatography–flame ionization detection (GC–FID) (Figure S2). The identification of the chemical constituents was based on comparisons of the obtained mass spectra (Figures S3–S7), retention indices, and previously reported data.
As a result of the qualitative and quantitative analyses, 49 compounds were identified, representing 98.17% of the total EO composition. All the detected constituents belonged to the terpenoid class, with a predominance of sesquiterpenes, which accounted for 68.50% of the total composition, whereas monoterpenes constituted 29.67% of the total composition. The major constituents (>5%) of the EO were (E)-caryophyllene (32.01%), 1,8-cineole (13.93%), α-copaene (7.77%), α-humulene (7.15%), and α-pinene (5.13%) (Table 1 and Figure 1).
These findings are consistent with those reported by Alcantara et al. [16], who identified (E)-caryophyllene as the major constituent of the EO extracted from the leaves of A. amazonica. In other Annona species, (E)-caryophyllene has also been identified among the principal components of the EOs from A. cherimola, A. foetida, A. emarginata, A. glabra, atemoya/Abdel Razek (hybrid of A. squamosa and A. cherimola), A. reticulata, A. squamosa, A. leptopetala, A. neoinsignis, A. pickelii, and A. salzmannii [19,20,21,22,23,24].
In addition, α-copaene, α-humulene, and/or α-pinene have also been previously reported as constituents of EOs from other Annona species [19,20,21,22,23,24,25,26,27]. These results suggest that the EO of A. amazonica shares similar chemical constituents with those of other Annona species. Nonetheless, variations in EO composition are common and may be influenced by various factors, including soil characteristics, climatic conditions, and plant age [28].

2.2. In Vitro Cytotoxic Effects of EO from the Leaves of A. amazonica

The in vitro cytotoxic activities of the EO from A. amazonica leaves were investigated via the alamar blue assay after 72 h of incubation. Table 2 displays the half-maximal inhibitory concentration (IC50) values found. EO had IC50 values ranging from 14.72 to 44.07 μg/mL for the human liver cancer cell line HepG2 and the human breast cancer cell line MCF-7. Doxorubicin was used as a positive control and had IC50 values ranging from 0.04 to 0.62 μg/mL for the human liver cancer cell line HepG2 and the human breast cancer cell line MDA-MB-231. For the noncancerous cell line MRC-5, EO had an IC50 value of 39.41 μg/mL, whereas doxorubicin had an IC50 value of 0.43 μg/mL.
The cytotoxic properties of EOs from Annona species have been previously reported and include those of EOs derived from A. cherimola, A. glabra, Atemoya/Abdel Razek, A. squamosa, A. leptopetala, A. senegalensis, A. sylvatica, A. pickelii, A. salzmannii, A. vepretorum, A. neoinsignis, and A. muricata [20,21,23,24,25,26,27,29,30,31]. Like the EO from A. amazonica leaves, (E)-caryophyllene is among the major constituents of the oils extracted from A. cherimola, A. glabra, Atemoya/Abdel Razek, A. squamosa, A. leptopetala, A. pickelii, A. neoinsignis, and A. salzmannii [20,21,23,24].
In contrast, the cytotoxic activity of the EO extracted from A. senegalensis leaves was related to the presence of caryophyllene oxide, as reported by Ahmed et al. [29]. Furthermore, spathulenol was identified as one of those responsible for the cytotoxicity observed in EO obtained from A. vepretorum leaves [25]. Moreover, the mixture of minor and major constituents of each EO may contribute to its pharmacological activity.

2.3. Apoptosis Induction by A. amazonica Leaf EO

In a new set of experiments, the induction of apoptosis by EO from A. amazonica leaves was evaluated via different flow cytometry assays at concentrations of 10, 5, and 2.5 μg/mL after 24 and 48 h of incubation with the liver cancer cell line HepG2, which is the cell line most sensitive to EO cytotoxicity.
First, the viability of HepG2 cells was assessed by staining with YO-PRO-1 together with propidium iodide (PI) (Figure 2). YO-PRO-1 staining is used as an early marker of apoptosis, whereas PI staining is used as a marker of cell membrane integrity [32]. In this assay, YO-PRO-1/PI were considered viable; YO-PRO-1+/PI were considered apoptotic cells; and YO-PRO-1+/PI+ plus YO-PRO-1/PI+ were considered dead cells, without identification of the type of cell death. Interestingly, EO increased apoptosis after 24 h of incubation, whereas dead cells predominated after 48 h of incubation. The number of viable cells decreased proportionally.
The size and internal complexity of the HepG2 cells were also assessed via flow cytometry at 24 and 48 h after EO treatment (Figure 3). Forward scatter (FSC) was used as a cell size parameter and side scatter (SSC) was used as a cell granularity parameter. The reduction in FSC after 24 and 48 h of incubation and the increase in SSC after 48 h of incubation indicated cell shrinkage and nuclear condensation, characteristics of apoptotic cell death. Furthermore, the mitochondrial membrane potential (ΔΨm) was quantified in HepG2 cells via rhodamine-123 staining after 24 h of incubation with EO (Figure 4). A significant reduction in the ΔΨm was detected at a concentration of 10 μg/mL, confirming that the EO can induce apoptosis.
Next, the cell cycle and internucleosomal DNA fragmentation were detected in EO-treated HepG2 cells. For this purpose, PI staining was used to determine the cellular DNA content, and the G0/G1 (2n), S (2–4n), and G2/M (4n) cell phases were quantified (Figure 5). All cells with a DNA content less than 2n (sub-G0/G1 cells) were considered to have fragmented DNA. At concentrations of 10, 5, and 2.5 μg/mL, EO increased the percentage of HepG2 cells with fragmented DNA by 43.3, 30.3, and 24.3% after 24 h (vs. 12.6% of the control) and by 46.8, 33.1, and 25.9% after 48 h (vs. 17.4% of the control), respectively. The cell cycle phases (G0/G1, S, and G2/M) were proportionally reduced in EO-treated HepG2 cells. These data are also in agreement with cell death caused by apoptosis.
Previous studies revealed that EO extracted from A. squamosa pericarp induced apoptosis in SMMC-7721 hepatocellular carcinoma cells, as reported by Chen et al. [31], whereas the EO obtained from A. neoinsignis leaves caused apoptosis in HepG2 cells, as reported by Souza et al. [24]. Similarly, EO obtained from A. vepretorum leaves promoted apoptosis in B16-F10 melanoma cells [25]. In this study, we demonstrated for the first time that the EO extracted from A. amazonica causes apoptotic cell death in HepG2 cells.
(E)-Caryophyllene, the main constituent of the EO of A. amazonica leaves, has been previously reported as a potential antitumor agent [33]. It induces apoptosis and decreases angiogenesis in colorectal cancer cells [34]. This compound also causes S-phase cell cycle arrest and death in ovarian cancer cells [35] and reduces STAT-3/mTOR/AKT signaling in bladder cancer cells [36]. (E)-Caryophyllene also causes G1 cell cycle arrest, reduces sphingosine kinase 1, and potentiates the effects of cisplatin in lung cancer cells [37,38,39].

2.4. In Vivo Effects of A. amazonica Leaf EO on Liver Cancer Development in a Xenograft Model

The in vivo efficacy of the EO from A. amazonica leaves on liver cancer development was evaluated in a xenograft model. NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice xenografted with HepG2 cells were treated with 60 mg/kg EO for two weeks (Figure 6A). After treatment, the mean tumor weight was 1.9 ± 0.3 g, while the EO-treated animals had a mean weight of 1.1 ± 0.2 (Figure 6B), indicating significant tumor inhibition of 39.2% (Figure 6C).
Histological analysis revealed that the morphological characteristics of the tumors were consistent with those of moderately differentiated hepatocellular carcinoma. The extracellular intratumoral matrix was predominantly composed of collagen fibers and exhibited limited vascularization. In some areas, neoplastic cells were arranged in island-like clusters embedded within a collagen-rich stroma. Extensive regions of coagulative necrosis were observed throughout the tumor mass and were frequently associated with the peripheral infiltration of inflammatory cells. Areas of granulation tissue and focal dystrophic calcification were also identified. Moreover, evidence of tumor invasion into adjacent muscle, adipose, and cartilaginous tissues was observed (Figure 6D).
Systemic toxicology evaluation was also performed in EO-treated NGS mice xenografted with HepG2 cells. All groups presented 100% survival rates, and no significant alterations in body or organ weights (liver, kidney, lung, or heart) were detected in any group (Figure S8).
The systemic toxicity of EO was evaluated through histological analysis of the heart, liver, lungs, and kidneys. The tissue architecture of the heart and kidneys remained preserved in both groups, with only mild vascular hyperemia observed as the sole alteration. In contrast, the hepatic parenchyma was partially preserved, with mild hydropic degeneration of hepatocytes. Vascular hyperemia was identified in the central venules and vessels of the portal triad, accompanied by infiltration of inflammatory cells, predominantly mononuclear, in these regions as well as in the perisinusoidal space. The pulmonary parenchyma exhibited marked structural alterations, including areas of atelectasis secondary to alveolar septal thickening caused by pneumocyte hyperplasia and hypertrophy. Moderate hyperemia was also observed in the pulmonary capillaries, along with inflammatory infiltrates predominantly composed of alveolar macrophages. Additionally, focal areas of necrosis were identified in the bronchial epithelium, along with foci of hemorrhage in the lung tissue (Figure S9).
In previous studies, A. vepretorum leaf EO complexed with β-cyclodextrin reduced the development of B16-F10 melanoma cells, and no signs of systemic toxicity were observed [25]. Similarly, A. leptopetala EO demonstrated antitumor activity in a 180 sarcoma tumor model, with no genotoxicity and only moderate gastrointestinal toxicity [23]. EO obtained from A. neoinsignis leaves also reduced HepG2 cell growth in a xenograft model [24]. The major chemical constituent of A. amazonica leaf EO, (E)-caryophyllene, also significantly reduces the tumor development of HCT116 cells in xenograft nude mice [34]. (E)-Caryophyllene also inhibited tumor growth and metastasis in a B16-F10 melanoma mouse model [40].

3. Conclusions

In summary, this study demonstrates, for the first time, the antitumor potential of A. amazonica leaf EO against liver cancer both in vitro and in vivo. The chemical composition of the EO revealed a predominance of bioactive compounds such as (E)-caryophyllene, 1,8-cineole, α-copaene, α-humulene, and α-pinene, which may contribute to its cytotoxic activity. EO treatment induced apoptosis in HepG2 cells through DNA fragmentation and loss of ΔΨm. In a xenograft model, EO reduced tumor development by 39.2%. These findings demonstrate that A. amazonica EO has promising properties as a natural therapeutic agent against liver cancer and justify further studies to characterize its pharmacological action and elucidate its molecular mechanisms of action, as well as its combination with clinically approved chemotherapeutics.

4. Materials and Methods

4.1. Botanical Material

Leaves of A. amazonica were collected in December 2021 at the Adolpho Ducke Forest Reserve, which is located near Manaus, Amazonas, Brazil (coordinates: 2°54′49.9” S, 59°58′43.6” W). The species was taxonomically identified by Professor Antônio Carlos Webber of the Federal University of Amazonas (UFAM). A voucher specimen was deposited in the Herbarium of the Department of Biology at UFAM (HUAM) under registration number #012098. The genetic material was registered in Brazil’s National System for the Management of Genetic Heritage and Associated Traditional Knowledge (SISGEN) under the registration code #A70EDCD.

4.2. EO Extraction

A. amazonica leaves were dried in a forced-air oven at 40 °C for 24 h and subsequently ground via a four-blade mill. EO extraction was performed by hydrodistillation via a Clevenger-type apparatus connected to a 4 L round-bottom flask. The mixture was heated to 100 °C, and extractions were conducted in triplicate, each with 300 g of plant material and sufficient distilled water to fill half the volume of the flask. The hydrodistillation process lasted for 3 h. The resulting EO was collected and dehydrated with anhydrous sodium sulfate to remove residual moisture and then stored in amber glass bottles under refrigeration to prevent chemical degradation. The EO yield was calculated as a percentage via the following formula: EO yield (%) = (mass of EO/mass of plant material) × 100.

4.3. Chemical Analysis of the EO

The chemical composition of the EO was analyzed via a TRACE GC ULTRA/ISQ gas chromatograph–mass spectrometer (Thermo Scientific) equipped with a TriPlus RSH autosampler operating in both gas chromatography–mass spectrometry and gas chromatography–flame ionization detection modes. Separation was achieved via the use of a DB-5MS fused silica capillary column (30 m × 0.25 mm × 0.25 μm) coated with 5% phenylarylene and 95% dimethylpolysiloxane. Helium was employed as the carrier gas at a constant flow rate of 1.0 mL/min. The oven temperature program was as follows: initial temperature of 40 °C (held for 4 min), increased at 4 °C/min to 240 °C, ramped at 10 °C/min to 280 °C, and held for 2 min. The injector temperature was set at 250 °C, and the detector temperature was 280 °C, following the method described by Silva et al. [41].
The samples were prepared by dissolving 10 mg of EO in 1 mL of dichloromethane to obtain pesticide residue. A 1 μL aliquot was injected in split mode (1:25 ratio). The retention indices were calculated via a homologous series of n-alkanes (C8–C20) and the equation proposed by van den Dool and Kratz [17]. Chromatographic data, including retention times and peak areas, were acquired and processed electronically. Mass spectra were recorded at 70 eV with a scan interval of 0.5 s and a mass range of 40–550 Da. The same column and stationary phase used for GC–FID were applied in the GC–MS analysis.
The EO constituents were identified by comparing the obtained mass spectra and calculated retention indices with those reported in the literature [18]. The relative abundance of each compound was expressed as a percentage of the total chromatographic peak area.

4.4. Cells

This study used a panel of cancer cell lines that included HCT116 (human colorectal cancer, ATCC code #CCL-247), HepG2 (human liver cancer, ATCC code #HB-8065), MDA-MB-231 (human breast cancer, ATCC code #HTB-26), MCF-7 (human breast cancer, ATCC code #HTB-22), 4T1 (mouse breast cancer, ATCC code #CRL-2539), and B16-F10 (mouse melanoma, ATCC code #CRL-6475) cells, along with the noncancerous human lung fibroblast line MRC-5 (ATCC code #CCL-171). All cell lines were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA) and cultured according to standard ATCC protocols for animal cell maintenance [42], and only mycoplasma-free cells were used.

4.5. Alamar Blue Assay

Cell viability was evaluated via the alamar blue assay, following the protocol described by Ahmed et al. [43]. The cells were seeded into 96-well plates and treated with the EO, which had been previously dissolved in dimethyl sulfoxide (DMSO; Vetec Química Fina Ltd.a., Duque de Caxias, RJ, Brazil). Eight different EO concentrations were tested, and the cells were incubated for 72 h. Doxorubicin hydrochloride (IMA S.A.I.C., Buenos Aires, Argentina) was used as a positive control. After the incubation period, 20 μL of a 1 mM resazurin solution (Sigma–Aldrich, St. Louis, MO, USA) was added to each well. The absorbance readings were taken at 570 and 600 nm via a SpectraMax 190 microplate reader (Molecular Devices, Sunnyvale, CA, USA). IC50 values and corresponding 95% confidence intervals (95% CIs) were calculated via nonlinear regression analysis.

4.6. Flow Cytometry Assays

The quantification of apoptotic cells was performed via the use of the fluorescent dyes YO-PRO-1 (Sigma–Aldrich Co.) and PI (BD Biosciences, San Jose, CA, USA), adapted from Idziorek et al. [32]. The cells were stained with a solution containing 0.1 µM YO-PRO-1 and 1.5 µM PI, and fluorescence was measured via flow cytometry.
The ΔΨm was assessed via rhodamine-123 staining, as described by Sureda et al. [44]. The cells were incubated with 1 µg/mL rhodamine-123 (Sigma–Aldrich Co.) for 15 min at 37 °C in the dark. After washing, the fluorescence intensity was analyzed via flow cytometry.
To assess internucleosomal DNA fragmentation and cell cycle distribution, the cells were stained with PI following the methods of Nicoletti et al. [45]. Briefly, the cells were suspended in permeabilization buffer containing 0.1% Triton X-100, 2 μg/mL PI, 0.1% sodium citrate, and 100 μg/mL RNase (all from Sigma–Aldrich Co.), followed by fluorescence analysis via flow cytometry.
All flow cytometric analyses were performed via a BD LSRFortessa cytometer, with data acquisition and analysis performed via BD FACSDiva (BD Biosciences) and FlowJo 10 (FlowJo LLC; Ashland, OR, USA). For each sample, at least 104 events were recorded. Cellular debris was excluded from the analysis, and individual cell populations were gated based on forward scatter height vs. area (FSC-H vs. FSC-A) and/or side scatter height vs. area (SSC-H vs. SSC-A) parameters.

4.7. Animals

Twenty NSG mice (specific pathogen-free, male and female, 8–10-week-old, 20–25 g) were obtained and housed in the animal facility of Instituto Gonçalo Moniz—FIOCRUZ (Salvador, Bahia, Brazil). The animals were kept under control in polyacrylic cages (with a maximum of five animals per cage), with ad libitum access to food and water. A 12 h light/dark cycle was applied (lights on at 7:00 AM). All procedures were approved by the Institutional Animal Care and Use Committee of the Instituto Gonçalo Moniz—FIOCRUZ (protocol number 01/2021), which are in accordance with international regulations.

4.8. Human Liver Tumor Xenograft Model

To establish the human liver tumor xenograft model, HepG2 cells (107 cells in 500 µL per animal) were injected subcutaneously into the left axillary region. Twenty-four hours after inoculation, the mice received daily intraperitoneal treatments (200 µL per animal) for two weeks. The animals were randomly divided into two groups: (1) the control group receiving vehicle (5% DMSO; n = 10) and (2) the treatment group receiving EO at a dose of 60 mg/kg (n = 10). At the end of the treatment period, the animals were euthanized by an anesthetic overdose (ketamine-xylazine), and the tumors were excised and weighed. Tumor growth inhibition was calculated via the following formula: inhibition ratio (%) = [(A − B)/A] × 100, where A is the mean tumor weight of the control group and B is the mean tumor weight of the treated group.
Toxicological evaluations included monitoring body weight at the beginning and end of the experiment. The animals were also observed daily for clinical signs of toxicity. Major organs (liver, kidneys, lungs, and heart) were removed, weighed, and examined macroscopically for signs of pathological changes such as lesions, discoloration, or hemorrhage. Histopathological analysis of tumors and organs was conducted via light microscopy after fixation in 4% formaldehyde. The tissue sections were stained with hematoxylin and eosin (H&E) for gross morphology and with periodic acid-Schiff (PAS) for evaluation of the liver and kidney. Microscopic examinations were conducted at magnifications of ×4, ×100, ×200, and ×400. Histological findings were scored semiquantitatively as negative (0), mild (+1), moderate (+2), or severe (+3). Representative images were acquired via a Leica LMD6500 microscope with LAS v4 image acquisition software.

4.9. Statistical Analysis

The data are expressed as the means ± standard errors of the means (SEMs) or as IC50 values accompanied by 95% CIs and are based on a minimum of three independent experiments (biological replicates) conducted in duplicate or triplicate (technical replicates). Statistical analysis between groups was performed via two-tailed unpaired Student’s t test or one-way analysis of variance (ANOVA) followed by Dunnett’s post hoc test. Differences were considered statistically significant at p < 0.05. All the statistical analyses were carried out via GraphPad Prism software version 8 (GraphPad Software, San Diego, CA, USA).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules30153248/s1, Figure S1: Illustration of the experimental workflow; Figure S2: Chromatogram of the total ions; Figure S3: Mass spectrum of α-pinene; Figure S4: Mass spectrum of 1,8-cineole; Figure S5: Mass spectrum of α-copaene; Figure S6: Mass spectrum of (E)-caryophyllene; Figure S7: Mass spectrum of α-humulene; Figure S8: The body weight and relative organ weight; Figure S9: Representative photomicrographs of organs.

Author Contributions

Conceptualization, M.V.L.d.C., M.C.F.d.L., M.B.P.S., E.V.C. and D.P.B.; methodology, M.V.L.d.C., M.C.F.d.L., G.A.d.C.B., S.G.C., A.M.R.M.C., L.d.S.S., V.R.S., R.B.D., M.B.P.S., E.V.C. and D.P.B.; formal analysis, M.V.L.d.C., M.C.F.d.L., L.d.S.S., V.R.S., R.B.D., E.V.C. and D.P.B.; investigation, M.V.L.d.C., M.C.F.d.L., G.A.d.C.B., S.G.C., A.M.R.M.C., L.d.S.S., V.R.S., R.B.D., M.B.P.S., E.V.C. and D.P.B.; writing—original draft preparation, D.P.B.; writing—review and editing, M.V.L.d.C., M.C.F.d.L., R.B.D., E.V.C. and D.P.B.; visualization, M.V.L.d.C., M.C.F.d.L., G.A.d.C.B., S.G.C., A.M.R.M.C., L.d.S.S., V.R.S., R.B.D., M.B.P.S., E.V.C. and D.P.B.; supervision, E.V.C. and D.P.B.; project administration, E.V.C. and D.P.B.; funding acquisition, E.V.C., M.B.P.S. and D.P.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Brazilian agencies Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, code 001), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), National Institutes of Science and Technology (INCT/CNPq Grant: 465357/2014-8), FitoAmazônia Research Network (Pró-Amazônia/CNPq Grant: 445615/2024—9), and Fundação de Amparo à Pesquisa do Estado do Amazonas (FAPEAM).

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Ethics Committee of the Oswaldo Cruz Foundation (Salvador, Bahia, Brazil) (protocol code: 01/2021; date of approval: 6 April 2021). All methods were performed in accordance with the relevant international guidelines and regulations.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the Supplementary Materials.

Acknowledgments

The authors are grateful to the histotechnology and flow cytometry cores of FIOCRUZ-Bahia for performing the histological techniques and collecting the flow cytometric data and to Central Analítica of the Universidade Federal do Amazonas (CA/UFAM) for the GC–MS analysis. The authors are also grateful to the Herbarium of the Department of Biology of Universidade Federal do Amazonas (HUAM) for collecting and identifying the botanical materials.

Conflicts of Interest

The authors declare that they have no conflicts of interest regarding the publication of this paper.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef]
  3. Brown, Z.J.; Tsilimigras, D.I.; Ruff, S.M.; Mohseni, A.; Kamel, I.R.; Cloyd, J.M.; Pawlik, T.M. Management of hepatocellular carcinoma: A review. JAMA Surg. 2023, 158, 410–420. [Google Scholar] [CrossRef]
  4. Shin, H.; Yu, S.J. A concise review of updated global guidelines for the management of hepatocellular carcinoma: 2017-2024. J. Liver Cancer 2025, 25, 19–30. [Google Scholar] [CrossRef]
  5. American Cancer Society. Cancer Facts & Figures 2025. Atlanta, Ga: American Cancer Society; 2025. Available online: https://www.cancer.org/cancer/types/liver-cancer/detection-diagnosis-staging/survival-rates.html (accessed on 20 June 2025).
  6. Sharma, M.; Grewal, K.; Jandrotia, R.; Batish, D.R.; Singh, H.P.; Kohli, R.K. Essential oils as anticancer agents: Potential role in malignancies, drug delivery mechanisms, and immune system enhancement. Biomed. Pharmacother. 2022, 146, 112514. [Google Scholar] [CrossRef] [PubMed]
  7. Santos, A.C.D.; Nogueira, M.L.; Oliveira, F.P.; Costa, E.V.; Bezerra, D.P. Essential oils of Duguetia species A. St. Hill (Annonaceae): Chemical diversity and pharmacological potential. Biomolecules 2022, 12, 615. [Google Scholar] [CrossRef] [PubMed]
  8. Abdoul-Latif, F.M.; Ainane, A.; Aboubaker, I.H.; Mohamed, J.; Ainane, T. Exploring the potent anticancer activity of essential oils and their bioactive compounds: Mechanisms and prospects for future cancer therapy. Pharmaceuticals 2023, 16, 1086. [Google Scholar] [CrossRef] [PubMed]
  9. de Sousa, D.P.; Damasceno, R.O.S.; Amorati, R.; Elshabrawy, H.A.; de Castro, R.D.; Bezerra, D.P.; Nunes, V.R.V.; Gomes, R.C.; Lima, T.C. Essential oils: Chemistry and pharmacological activities. Biomolecules 2023, 13, 1144. [Google Scholar] [CrossRef]
  10. Alabrahim, O.A.A.; Lababidi, J.M.; Fritzsche, W.; Azzazy, H.M.E. Beyond aromatherapy: Can essential oil loaded nanocarriers revolutionize cancer treatment? Nanoscale Adv. 2024, 6, 5511–5562. [Google Scholar] [CrossRef]
  11. Maas, P.J.M.; Maas, H.; Miralha, J.M.S.; Junikka, L. Flora da reserva Ducke, Amazonas, Brasil: Annonaceae. Rodriguésia 2007, 58, 617. [Google Scholar] [CrossRef]
  12. The Herbarium Catalogue, Royal Botanic Gardens, Kew. Published on the Internet. Available online: http://www.kew.org/herbcat (accessed on 2 June 2025).
  13. Mendes-Silva, I.; Lopes, J.C.; Silva, L.V.; Bazante, M.L. Annona in flora e funga do Brasil. Jardim Botânico do Rio de Janeiro. Available online: https://floradobrasil.jbrj.gov.br/FB117059 (accessed on 9 June 2025).
  14. Thomsen, K.; Brimer, L. Cyanogenic constituents in woody plants in natural lowland rain forest in Costa Rica. Bot. J. Linnean Soc. 1997, 124, 273–294. [Google Scholar] [CrossRef]
  15. Pinheiro, M.L.B.; Xavier, C.M.; De Souza, A.D.; Rabelo, D.D.M.; Batista, C.L.; Batista, R.L.; Costa, E.V.; Campos, F.R.; Barison, A.; Valdez, R.H.; et al. Acanthoic acid and other constituents from the stem of Annona amazonica (Annonaceae). J. Braz. Chem. Soc. 2009, 20, 1095–1102. [Google Scholar] [CrossRef]
  16. Alcântara, J.M.; Lucena, J.M.V.M.; Marques, M.O.M.; Lima, J.A.O.; Lima, M.P. Chemical composition and antibacterial activity of the essential oils of the Amazon Annona species. Orbital: Electron. J. Chem. 2024, 16, 263–266. [Google Scholar] [CrossRef]
  17. Van Den Dool, H.A.N.D.; Kratz, P.D. A generalization of the retention index system including linear temperature programmed gas-liquid partition chromatography. J. Chromatogr. 1963, 11, 463–471. [Google Scholar] [CrossRef]
  18. Adams, R.P. 2007Identification of Essential oil Components by Gas Chromatography/Mass Spectroscopy, 4th ed.; Allured Publishing, Corp: Carol Stream, IL, USA, 2007; 803p. [Google Scholar]
  19. Costa, E.V.; Pinheiro, M.L.B.; Silva, J.R.A.; Maia, B.H.L.N.S.; Duarte, M.C.T.; Amaral, A.C.F.; Machado, G.M.C.; Leon, L.L. Antimicrobial and antileishmanial activity of essential oil from the leaves of Annona foetida (Annonaceae). Quim. Nova 2009, 32, 78–81. [Google Scholar] [CrossRef]
  20. Costa, E.V.; Dutra, L.M.; Salvador, M.J.; Ribeiro, L.H.; Gadelha, F.R.; de Carvalho, J.E. Chemical composition of the essential oils of Annona pickelii and Annona salzmannii (Annonaceae), and their antitumour and trypanocidal activities. Nat. Prod. Res. 2013, 27, 997–1001. [Google Scholar] [CrossRef]
  21. Elhawary, S.S.; El Tantawy, M.E.; Rabeh, M.A.; Fawaz, N.E. DNA fingerprinting, chemical composition, antitumor and antimicrobial activities of the essential oils and extractives of four Annona species from Egypt. J. Nat. Sci. Res. 2013, 3, 59–68. [Google Scholar]
  22. Campos, F.G.; Baron, D.; Marques, M.O.M.; Ferreira, G.; Boaro, C.S.F. Caracterização do perfil químico do óleo essencial de Annona emarginata (Schltdl.) H. Rainer ‘terra-fria’ and Annona squamosa L. Rev. Bras. Frutic. 2014, 36, 202–208. [Google Scholar] [CrossRef]
  23. Brito, M.T.; Ferreira, R.C.; Beltrao, D.M.; Moura, A.P.G.; Xavier, A.L.; Pita, J.C.L.R.; Batista, T.M.; Longato, G.B.; Ruiz, A.L.T.G.; Carvalho, J.E.D.; et al. Antitumor activity and toxicity of volatile oil from the leaves of Annona leptopetala. Rev. Bras. Farmacogn. 2018, 28, 602–609. [Google Scholar] [CrossRef]
  24. Souza, M.P.; de Castro, M.V.L.; Barbosa, G.A.d.C.; Carvalho, S.G.; Coelho, A.M.R.M.; Dias, R.B.; Soares, M.B.P.; Costa, E.V.; Bezerra, D.P. Essential oil from the leaves of Annona neoinsignis H. Rainer (Annonaceae) against liver cancer: In vitro and in vivo studies. Molecules 2025, 30, 2971. [Google Scholar] [CrossRef]
  25. Bomfim, L.M.; Menezes, L.R.; Rodrigues, A.C.; Dias, R.B.; Rocha, C.A.; Soares, M.B.; Neto, A.F.; Nascimento, M.P.; Campos, A.F.; Silva, L.C.; et al. Antitumour activity of the microencapsulation of Annona vepretorum essential oil. Basic Clin. Pharmacol. Toxicol. 2016, 118, 208–213. [Google Scholar] [CrossRef]
  26. Rabelo, S.V.; Oliveira, F.G.D.S.; Lira, M.M.C.; Dutra, L.M.; Sartoratto, A.; Duarte, M.C.T.; Luciano, M.C.D.S.; Silva, M.F.S.; Pessoa, C.D.O.; Moraes Filho, M.O.; et al. Non-polar chemical constituents of atemoya and evaluation of the cytotoxic and antimicrobial activity. Phyton 2021, 90, 921–931. [Google Scholar] [CrossRef]
  27. Mohammed, M.A.; Elzefzafy, N.; El-Khadragy, M.F.; Alzahrani, A.; Yehia, H.M.; Kachlicki, P. Comprehensive tools of alkaloid/volatile compounds-metabolomics and DNA profiles: Bioassay-role-guided differentiation process of six Annona sp. Grown in Egypt as anticancer therapy. Pharmaceuticals 2024, 17, 103. [Google Scholar] [CrossRef]
  28. Soares, E.R.; Almeida, R.A.; Lima, B.R.; Pereira Junior, R.C.; Freitas, F.A.; Mafra, H.R.; Araujo, N.F.; Maciel, J.B.; Leão, L.Q.S.; Souza, A.D.L.; et al. Chemical composition of essential oils of three species of the genus Bocageopsis (Annonaceae) Amazon region. Rev. Vir. Quím 2022, 14, 947–953. [Google Scholar] [CrossRef]
  29. Ahmed, A.L.; Bassem, S.E.; Mohamed, Y.H.; Gamila, M.W. Cytotoxic essential oil from Annona sengalensis Pers. leaves. Pharmacogn. Res. 2010, 2, 211–214. [Google Scholar] [CrossRef] [PubMed]
  30. Formagio, A.S.; Vieira, M.C.; Dos Santos, L.A.; Cardoso, C.A.; Foglio, M.A.; de Carvalho, J.E.; Andrade-Silva, M.; Kassuya, C.A. Composition and evaluation of the anti-inflammatory and anticancer activities of the essential oil from Annona sylvatica A. St.-Hil. J. Med. Food. 2013, 16, 20–25. [Google Scholar] [CrossRef] [PubMed]
  31. Chen, Y.Y.; Peng, C.X.; Hu, Y.; Bu, C.; Guo, S.C.; Li, X.; Chen, Y.; Chen, J.W. Studies on chemical constituents and anti-hepatoma effects of essential oil from Annona squamosa L. pericarps. Nat. Prod. Res. 2017, 31, 1305–1308. [Google Scholar] [CrossRef] [PubMed]
  32. Idziorek, T.; Estaquier, J.; De Bels, F.; Ameisen, J.C. YOPRO-1 permits cytofluorometric analysis of programmed cell death (apoptosis) without interfering with cell viability. J. Immunol. Methods 1995, 185, 249–258. [Google Scholar] [CrossRef]
  33. Ahmed, E.A. The Potential therapeutic role of beta-caryophyllene as a chemosensitizer and an inhibitor of angiogenesis in cancer. Molecules 2025, 30, 1751. [Google Scholar] [CrossRef]
  34. Dahham, S.S.; Tabana, Y.; Asif, M.; Ahmed, M.; Babu, D.; Hassan, L.E.; Ahamed, M.B.K.; Sandai, D.; Barakat, K.; Siraki, A.; et al. β-Caryophyllene induces apoptosis and inhibits angiogenesis in colorectal cancer models. Int. J. Mol. Sci. 2021, 22, 10550. [Google Scholar] [CrossRef]
  35. Arul, S.; Rajagopalan, H.; Ravi, J.; Dayalan, H. Beta-caryophyllene suppresses ovarian cancer proliferation by inducing cell cycle arrest and apoptosis. Anticancer Agents Med. Chem. 2020, 20, 1530–1537. [Google Scholar] [CrossRef] [PubMed]
  36. Yu, X.; Liao, B.; Zhu, P.; Cheng, S.; Du, Z.; Jiang, G. β-Caryophyllene induces apoptosis and inhibits cell proliferation by deregulation of STAT-3/mTOR/AKT signaling in human bladder cancer cells: An in vitro study. J. Biochem. Mol. Toxicol. 2021, 35, e22863. [Google Scholar] [CrossRef] [PubMed]
  37. Chung, K.S.; Hong, J.Y.; Lee, J.H.; Lee, H.J.; Park, J.Y.; Choi, J.H.; Park, H.J.; Hong, J.; Lee, K.T. β-Caryophyllene in the essential oil from Chrysanthemum Boreale induces G1 phase cell cycle arrest in human lung cancer cells. Molecules 2019, 24, 3754. [Google Scholar] [CrossRef] [PubMed]
  38. Lei, J.; Wang, Q.; Li, G.; Li, Y.; Zhang, P.; Xu, G. β-Caryophyllene from chilli pepper inhibits the proliferation of non-small cell lung cancer cells by affecting miR-659-3p-targeted sphingosine kinase 1 (SphK1). Int. J. Gen. Med. 2021, 14, 9599–9613. [Google Scholar] [CrossRef]
  39. Ahmed, E.A.; Abu Zahra, H.; Ammar, R.B.; Mohamed, M.E.; Ibrahim, H.M. Beta-caryophyllene enhances the anti-tumor activity of cisplatin in lung cancer cell lines through regulating cell cycle and apoptosis signaling molecules. Molecules 2022, 27, 8354. [Google Scholar] [CrossRef]
  40. Jung, J.I.; Kim, E.J.; Kwon, G.T.; Jung, Y.J.; Park, T.; Kim, Y.; Yu, R.; Choi, M.S.; Chun, H.S.; Kwon, S.H.; et al. β-Caryophyllene potently inhibits solid tumor growth and lymph node metastasis of B16F10 melanoma cells in high-fat diet-induced obese C57BL/6N mice. Carcinogenesis 2015, 36, 1028–1039. [Google Scholar] [CrossRef]
  41. Silva, T.B.; Menezes, L.R.A.; Sampaio, M.F.C.; Meira, C.S.; Guimaraes, E.T.; Soares, M.B.P.; Prata, A.P.N.; Nogueira, P.C.L.; Costa, E.V. Chemical composition and anti-Trypanosoma cruzi activity of essential oils obtained from leaves of Xylopia frutescens and X. laevigata (Annonaceae). Nat. Prod. Commun. 2013, 8, 403–406. [Google Scholar] [CrossRef]
  42. ATCC Animal Cell Culture Guide: Get Time-Tested Tips for Culturing ATCC Animal Cells. Available online: https://www.atcc.org/resources/culture-guides/animal-cell-culture-guide (accessed on 9 June 2024).
  43. Ahmed, S.A.; Gogal, R.M.; Walsh, J.E. A new rapid and simple non-radioactive assay to monitor and determine the proliferation of lymphocytes an alternative to [3H] thymidine incorporation assay. J. Immunol. Methods 1994, 170, 211–224. [Google Scholar] [CrossRef]
  44. Sureda, F.X.; Escubedo, E.; Gabriel, C.; Comas, J.; Camarasa, J.; Camins, A. Mitochondrial membrane potential measurement in rat cerebellar neurons by flow cytometry. Cytometry 1997, 28, 74–80. [Google Scholar] [CrossRef]
  45. Nicoletti, I.; Migliorati, G.; Pagliacci, M.C.; Grignani, F.; Riccardi, C. A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry. J. Immunol. Methods 1991, 139, 271–279. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Principal constituents identified in the EO hydrodistillation from the leaves of A. amazonica.
Figure 1. Principal constituents identified in the EO hydrodistillation from the leaves of A. amazonica.
Molecules 30 03248 g001
Figure 2. Apoptotic response of HepG2 cells to A. amazonica leaf EO after 24 and 48 h of treatment. (A) Representative flow cytometry dot plots showing the distribution of viable, apoptotic, and dead cells. (B) Quantitative analysis of the following cell populations: viable cells (YO-PRO-1/PI), apoptotic cells (YO-PRO-1+/PI), and dead cells (YO-PRO-1+/PI+ plus YO-PRO-1/PI+). The cells treated with 0.2% DMSO served as the vehicle control (CTL). The results are expressed as the mean ± SEM of three independent experiments performed in triplicate. * p < 0.05 vs. the control group (CTL), as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Figure 2. Apoptotic response of HepG2 cells to A. amazonica leaf EO after 24 and 48 h of treatment. (A) Representative flow cytometry dot plots showing the distribution of viable, apoptotic, and dead cells. (B) Quantitative analysis of the following cell populations: viable cells (YO-PRO-1/PI), apoptotic cells (YO-PRO-1+/PI), and dead cells (YO-PRO-1+/PI+ plus YO-PRO-1/PI+). The cells treated with 0.2% DMSO served as the vehicle control (CTL). The results are expressed as the mean ± SEM of three independent experiments performed in triplicate. * p < 0.05 vs. the control group (CTL), as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Molecules 30 03248 g002
Figure 3. Effects of A. amazonica leaf EO on the size and internal complexity of HepG2 cells, as assessed by flow cytometry at 24 and 48 h after treatment. (A) Representative dot plots showing forward scatter (FSC) and side scatter (SSC) profiles, which reflect cell size and granularity, respectively. (B) Quantitative analysis of FSC and SSC values. The cells treated with 0.2% DMSO served as the vehicle control (CTL). The data are expressed as the means ± SEMs of three independent experiments performed in triplicate. * p < 0.05 vs. CTL, as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Figure 3. Effects of A. amazonica leaf EO on the size and internal complexity of HepG2 cells, as assessed by flow cytometry at 24 and 48 h after treatment. (A) Representative dot plots showing forward scatter (FSC) and side scatter (SSC) profiles, which reflect cell size and granularity, respectively. (B) Quantitative analysis of FSC and SSC values. The cells treated with 0.2% DMSO served as the vehicle control (CTL). The data are expressed as the means ± SEMs of three independent experiments performed in triplicate. * p < 0.05 vs. CTL, as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Molecules 30 03248 g003
Figure 4. The ΔΨm in HepG2 cells after 24 h of treatment with A. amazonica leaf EO assessed via rhodamine-123 staining and flow cytometry. (A) Representative histograms obtained by flow cytometry. (B) Quantitative analysis of ΔΨm. The cells treated with 0.2% DMSO served as the vehicle control (CTL). The results are expressed as the mean fluorescence intensity (MFI) and represent the mean ± SEM of three independent experiments conducted in triplicate. * p < 0.05 vs. CTL, as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Figure 4. The ΔΨm in HepG2 cells after 24 h of treatment with A. amazonica leaf EO assessed via rhodamine-123 staining and flow cytometry. (A) Representative histograms obtained by flow cytometry. (B) Quantitative analysis of ΔΨm. The cells treated with 0.2% DMSO served as the vehicle control (CTL). The results are expressed as the mean fluorescence intensity (MFI) and represent the mean ± SEM of three independent experiments conducted in triplicate. * p < 0.05 vs. CTL, as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Molecules 30 03248 g004
Figure 5. Cell cycle distribution of HepG2 cells after treatment with A. amazonica leaf EO for 24 and 48 h. (A) Representative histograms obtained by flow cytometry. (B) Quantitative analysis of the percentage of cells in the sub-G0/G1 (cells with fragmented DNA), G0/G1, S, and G2/M fractions. The cells treated with 0.2% DMSO served as the vehicle control (CTL). The data are expressed as the means ± SEMs of three independent experiments performed in triplicate. * p < 0.05 vs. CTL, as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Figure 5. Cell cycle distribution of HepG2 cells after treatment with A. amazonica leaf EO for 24 and 48 h. (A) Representative histograms obtained by flow cytometry. (B) Quantitative analysis of the percentage of cells in the sub-G0/G1 (cells with fragmented DNA), G0/G1, S, and G2/M fractions. The cells treated with 0.2% DMSO served as the vehicle control (CTL). The data are expressed as the means ± SEMs of three independent experiments performed in triplicate. * p < 0.05 vs. CTL, as determined by one-way ANOVA followed by Dunnett’s post hoc test.
Molecules 30 03248 g005
Figure 6. Antitumor effects of A. amazonica leaf EO in NSG mice xenografted with HepG2 cells. (A) Liver tumor xenograft model design. (B) Tumor weight and (C) tumor growth inhibition in animals treated intraperitoneally with EO (60 mg/kg) for two weeks. (D) Representative histological images of HepG2 tumor tissues. Arrows highlight regions of dystrophic calcification, necrotic areas are demarcated with circles, and fibrotic scar tissue is indicated by an asterisk. The mice that received 5% DMSO served as the vehicle control (CTL). The data are presented as the means ± SEMs for groups of 10 animals. * p < 0.05 compared with CTL, as assessed by two-tailed unpaired Student’s t test.
Figure 6. Antitumor effects of A. amazonica leaf EO in NSG mice xenografted with HepG2 cells. (A) Liver tumor xenograft model design. (B) Tumor weight and (C) tumor growth inhibition in animals treated intraperitoneally with EO (60 mg/kg) for two weeks. (D) Representative histological images of HepG2 tumor tissues. Arrows highlight regions of dystrophic calcification, necrotic areas are demarcated with circles, and fibrotic scar tissue is indicated by an asterisk. The mice that received 5% DMSO served as the vehicle control (CTL). The data are presented as the means ± SEMs for groups of 10 animals. * p < 0.05 compared with CTL, as assessed by two-tailed unpaired Student’s t test.
Molecules 30 03248 g006
Table 1. Chemical composition of the EO obtained from the leaves of A. amazonica.
Table 1. Chemical composition of the EO obtained from the leaves of A. amazonica.
CompoundsAI aAI bPeak Area (%)
1α-pinene9299325.13 ± 1.72
2α-fenchene9439450.32 ± 0.20
3sabinene9699690.11 ± 0.02
4β-pinene9719741.72 ± 0.38
5myrcene9909880.62 ± 0.24
6α-terpinene101310140.12 ± 0.03
7O-cymene102110220.18 ± 0.02
81,8-cineole1027102613.93 ± 0.84
9(Z)-β-ocimene103810320.11 ± 0.05
10(E)-β-ocimene104810440.21 ± 0.08
11γ-terpinene105610540.15 ± 0.02
12terpinolene108510861.04 ± 0.42
13linalool109910952.91 ± 0.79
14terpinen-4-ol117411740.23 ± 0.09
15α-terpineol118811862.16 ± 0.76
16geraniol125412490.73 ± 0.41
17δ-elemene133613350.19 ± 0.08
18α-cubebene134913480.32 ± 0.04
19cyclosativene136513690.31 ± 0.04
20α-copaene137513747.77 ± 0.36
21β-elemene139113890.60 ± 0.36
22(E)-caryophyllene1420141732.01 ± 3.98
23β-copaene142714301.10 ± 0.18
24γ-elemene143314340.15 ± 0.01
25trans-α-bergamotene143514320.40 ± 0.19
26aromadendrene143714390.56 ± 0.03
276,9-guaiadiene144214420.24 ± 0.10
28α-humulene145214527.15 ± 1.10
29allo-aromadendrene145914580.12 ± 0.02
30γ-muurolene147514780.76 ± 0.07
31germacrene D148014801.86 ± 1.43
32β-selinene148414890.36 ± 0.03
33cis-β-guaiene149014920.12 ± 0.05
34bicyclogermacrene149515002.07 ± 0.35
35α-muurolene149915000.77 ± 0.05
36γ-cadinene151215130.29 ± 0.04
37δ-cadinene152215221.17 ± 0.05
38α-calacorene154115440.14 ± 0.03
39elemol154815480.88 ± 0.09
40germacrene B155515590.37 ± 0.08
41caryophyllene oxide157915820.52 ± 0.09
42guaiol159616001.40 ± 0.30
43eremoligenol162716290.43 ± 0.14
44γ-eudesmol163016301.54 ± 0.48
45hinesol163716400.12 ± 0.07
46cubenol164016450.17 ± 0.11
47β-eudesmol164816491.76 ± 0.62
48α-eudesmol165116522.59 ± 0.90
49bulnesol166516700.26 ± 0.09
Monoterpenes 29.67
Sesquiterpenes 68.50
Total not identified 1.83
Total identified 98.17
Note: The data are presented as the means ± S.D. of two analyses. RI (retention indices): This index was calculated on a TR-5MS capillary column (30 m × 0.25 mm × 0.25 µm) according to Van Den Dool and Kratz [17], which is based on a homologous series of normal alkanes, according to Adams [18].
Table 2. In vitro cytotoxic effects of A. amazonica leaf EO.
Table 2. In vitro cytotoxic effects of A. amazonica leaf EO.
CellsHistological TypeIC50 and 95% CIs (in μg/mL)
DOXEO
Cancer cells
HCT116Human colon cancer0.20
0.11–0.35
21.42
17.60–26.07
HepG2Human liver cancer0.04
0.03–0.05
14.72
7.54–28.75
MDA-MB-231Human breast cancer0.62
0.37–1.02
16.20
6.59–29.85
MCF-7Human breast cancer0.36
0.20–0.63
44.07
39.37–48.52
4T1Mouse breast cancer0.60
0.31–0.96
21.65
16.41–29.90
B16-F10Mouse melanoma0.06
0.03–0.09
32.16
27.46–38.34
Noncancerous cells
MRC-5Human lung fibroblast0.43
0.18–1.03
39.41
30.88–50.29
Note: Doxorubicin (DOX) was used as a positive control.
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

de Castro, M.V.L.; de Lima, M.C.F.; da C. Barbosa, G.A.; Carvalho, S.G.; Coelho, A.M.R.M.; de S. Santos, L.; Silva, V.R.; Dias, R.B.; Soares, M.B.P.; Costa, E.V.; et al. In Vitro and In Vivo Efficacy of the Essential Oil from the Leaves of Annona amazonica R.E. Fries (Annonaceae) Against Liver Cancer. Molecules 2025, 30, 3248. https://doi.org/10.3390/molecules30153248

AMA Style

de Castro MVL, de Lima MCF, da C. Barbosa GA, Carvalho SG, Coelho AMRM, de S. Santos L, Silva VR, Dias RB, Soares MBP, Costa EV, et al. In Vitro and In Vivo Efficacy of the Essential Oil from the Leaves of Annona amazonica R.E. Fries (Annonaceae) Against Liver Cancer. Molecules. 2025; 30(15):3248. https://doi.org/10.3390/molecules30153248

Chicago/Turabian Style

de Castro, Maria V. L., Milena C. F. de Lima, Gabriela A. da C. Barbosa, Sabrine G. Carvalho, Amanda M. R. M. Coelho, Luciano de S. Santos, Valdenizia R. Silva, Rosane B. Dias, Milena B. P. Soares, Emmanoel V. Costa, and et al. 2025. "In Vitro and In Vivo Efficacy of the Essential Oil from the Leaves of Annona amazonica R.E. Fries (Annonaceae) Against Liver Cancer" Molecules 30, no. 15: 3248. https://doi.org/10.3390/molecules30153248

APA Style

de Castro, M. V. L., de Lima, M. C. F., da C. Barbosa, G. A., Carvalho, S. G., Coelho, A. M. R. M., de S. Santos, L., Silva, V. R., Dias, R. B., Soares, M. B. P., Costa, E. V., & Bezerra, D. P. (2025). In Vitro and In Vivo Efficacy of the Essential Oil from the Leaves of Annona amazonica R.E. Fries (Annonaceae) Against Liver Cancer. Molecules, 30(15), 3248. https://doi.org/10.3390/molecules30153248

Article Metrics

Back to TopTop