Next Article in Journal
Nickel-Rich Cathodes for Solid-State Lithium Batteries: Comparative Study Between PVA and PIB Binders
Previous Article in Journal
Editorial to the Special Issue “Gulliver in the Country of Lilliput: An Interplay of Noncovalent Interactions (Volume II)”
Previous Article in Special Issue
Polysorbate 80 Differentially Impacts Erinacine Production Profiles in Submerged Cultures of Hericium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Essential Oil from the Leaves of Annona neoinsignis H. Rainer (Annonaceae) Against Liver Cancer: In Vitro and In Vivo Studies

by
Melissa P. Souza
1,2,†,
Maria V. L. de Castro
3,†,
Gabriela A. da C. Barbosa
3,
Sabrine G. Carvalho
3,
Amanda M. R. M. Coelho
3,
Rosane B. Dias
3,4,
Milena B. P. Soares
3,5,
Emmanoel V. Costa
1,2,* and
Daniel P. Bezerra
3,*
1
Postgraduate Program in Chemistry, Institute of Exact Sciences, Federal University of Amazonas (UFAM), Manaus 69080-900, Amazonas, Brazil
2
Department of Chemistry, Institute of Exact Sciences, Federal University of Amazonas (UFAM), Manaus 69080-900, Amazonas, Brazil
3
Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
4
Department of Biological Sciences, State University of Feira de Santana, Feira de Santana 44036-900, Bahia, Brazil
5
SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador 41650-010, Bahia, Brazil
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(14), 2971; https://doi.org/10.3390/molecules30142971
Submission received: 10 June 2025 / Revised: 7 July 2025 / Accepted: 10 July 2025 / Published: 15 July 2025

Abstract

Annona neoinsignis H. Rainer (Annonaceae) is a tree native to the Amazon rainforest. Its fruits are also suitable for human consumption in their natural state or are processed to make desserts. In this work, we characterized the chemical composition of the essential oil (EO) from the leaves of A. neoinsignis and evaluated its anti-liver-cancer potential via in vitro and in vivo approaches. Chemical composition analysis revealed β-elemene, (E)-caryophyllene, germacrene D, and germacrene B as the main constituents. The EO had IC50 values ranging from 12.28 to 37.50 μg/mL for B16-F10 cells and MCF-7 cells, whereas an IC50 value of >50 μg/mL was found for noncancerous MRC-5 cells. DNA fragmentation, YO-PRO-1 staining, and loss of mitochondrial transmembrane potential were detected in EO-treated HepG2 cells, indicating the induction of apoptosis. Significant in vivo growth inhibition of 53.7% was observed in mice bearing HepG2 cell xenografts treated with EO at a dosage of 40 mg/kg. These data suggest that EO from A. neoinsignis leaves is a drug source for liver cancer.

1. Introduction

Liver cancer is the third deadliest cancer and the sixth most common cancer worldwide, with an estimated 757,948 deaths and 865,269 new cases in 2022 [1]. By 2040, there will be an estimated 1,392,474 new cases of liver cancer each year [2].
In recent years, several new therapies have been approved to treat advanced liver cancer, including immune checkpoint inhibitors (e.g., atezolizumab, durvalumab, nivolumab, and pembrolizumab), multiple kinase inhibitors (e.g., sorafenib, regorafenib, lenvatinib, and cabozantinib), and angiogenesis inhibitors (bevacizumab and ramucirumab) [3,4,5,6]. On the other hand, although new therapeutic options have been recently approved, current treatments for patients with liver cancer have limited success, so new drugs are urgently needed.
Annona neoinsignis H. Rainer (synonyms Rollinia insignis R. E. Fr. and Rollinia insignis var. pallida R. E. Fr.) (Annonaceae), popularly known in Peru as ‘loreto’ or in Brazil as ‘envira-bobó’, ‘envireira-bobó’, ‘araticum do mato’, ‘cortição’, and ‘cortiça crespa’, is a tree native to the Amazon rainforest. This tree is suitable for both urban landscaping and forest restoration, as its fruits serve as food for birds and other terrestrial animals. Its fruits are also suitable for human consumption in their natural state or are processed to make mousses, ice creams, doughs, or fillings for cakes and cookies [7,8,9,10,11]. Despite this, only one scientific study was conducted with this plant, in which naphthalene was detected as the main volatile constituent of its flowers [12].
Essential oils (EOs) from different Annona species, including Annona senegalensis Pers., Annona pickelii (Diels) H. Rainer, Annona salzmannii A. DC., Annona vepretorum Mart., Annona glabra L., Annona leptopetala (R.E.Fr) H. Rainer, Annona muricata L., Annona squamosa L., Annona cherimola Mill., and Atemoya/Abdel Razek (hybrid of A. squamosa and A. cherimola) have been reported as cytotoxic agents [13,14,15,16,17,18,19,20]. On the other hand, the EO of A. neoinsignis has never been evaluated for its antitumor potential. In this work, we characterized the chemical composition of the EO from the leaves of A. neoinsignis and evaluated its anti-liver-cancer potential via in vitro and in vivo approaches.

2. Results and Discussion

2.1. Chemical Composition of the EO from the Leaves of A. neoinsignis

The chemical components of the EO extracted from the leaves of A. neoinsignis were determined and quantified via gas chromatograph connected to a mass spectrometer (GC–MS) and flame ionization detector (GC–FID). The volatile compounds were identified by their mass spectra and retention indices, which were then compared with previously published data. The oil had a greenish color and a yield of 0.18% in relation to the weight of the dry material and a density of 0.80 g/cm3. The EO is mainly composed of hydrocarbons of the monoterpene and sesquiterpene groups, as well as their derivatives. A total of 44 compounds were detected, representing 99.84% (Table 1).
The main compounds identified in the EO extracted from the leaves of A. neoinsignis were β-elemene (29.61%), (E)-caryophyllene (18.23%), germacrene D (15.34%), and germacrene B (6.80%), which together accounted for approximately 69.98% of the total identified oil (Figure 1 and Figures S1–S45). Other compounds identified at concentrations above 1.5% included bicyclogermacrene (4.61%), γ-elemene (4.47%), α-humulene (3.33%), α-copaene (2.52%), δ-elemene (2.27%), δ-amorphene (1.81%), and β-selinene (1.69%).
These findings corroborate what was reported in previous studies with EOs from Annonaceae species, particularly those belonging to the genus Annona. The prevalence of sesquiterpene hydrocarbons is common in the leaf EOs of plants of this family [23], and the main compounds found in the EO of A. neoinsignis resemble those identified in several species of the genus Annona, including the EOs extracted from the leaves of A. coriacea [24], A. vepretorum [25], and A. squamosa [26].
Furthermore, the occurrence of germacrene D, bicyclogermacrene, and (E)-caryophyllene as major compounds has also been detected in the chemical composition of the leaf EOs of A. atemoya, A. senegalensis, A. pickelii, A. glabra, and A. foetida [27]. Factors that can affect the chemical composition of EOs include temperature, seasonal changes, water supply, soil nutrients, circadian rhythm, and plant age [28].

2.2. Cytotoxicity of the EO Extracted from the Leaves of A. neoinsignis

The cytotoxicity of A. neoinsignis leaf EO was evaluated against six (HepG2, HCT116, MCF-7, MDA-MB-231, 4T1, and B16-F10) cancer cell lines and one (MRC-5) noncancerous cell line by the Alamar blue assay after 72 h of incubation (Figure S46). Table 2 shows the half maximum inhibitory concentration (IC50) values found. A. neoinsignis leaf EO had the lowest IC50 value (12.28 μg/mL) in the mouse melanoma cell line B16-F10, while the highest IC50 value (37.50 μg/mL) was found in the human breast cancer cell line MCF-7. Doxorubicin was used as a positive control, and the IC50 values ranged from 0.03 to 0.84 μg/mL for the liver cancer cell line HepG2 and the human breast cancer cell line MCF-7. For noncancer cell lines, A. neoinsignis leaf EO had IC50 values > 50 μg/mL for the human lung fibroblast line. Doxorubicin had an IC50 value of 1.96 μg/mL for the same cell line.
As mentioned previously, many EOs derived from Annona species have been shown to be cytotoxic. Among them, the cytotoxicity of EO extracted from A. senegalensis leaves was associated with the presence of caryophyllene oxide [13], whereas spathulenol was attributed, at least in part, to the cytotoxicity of EO extracted from A. vepretorum leaves [16]. In the EO from A. neoinsignis leaves, the main constituents are β-elemene, (E)-caryophyllene, germacrene D, and germacrene B. Importantly, these major components of A. neoinsignis leaf EO are known cytotoxic agents [29,30,31,32]. In any case, the cytotoxicity of this EO can be attributed to the combination of major and minor volatile compounds.

2.3. Induction of Apoptosis by A. neoinsignis Leaf EO in Liver Cancer Cells

The liver cancer cell line HepG2 is sensitive to A. neoinsignis leaf EO and was selected for further experiments. Next, cell cycle analysis was also performed on HepG2 cells treated with EO from A. neoinsignis leaves at concentrations of 5, 10, and 15 μg/mL after 24 and 48 h of incubation (Figure 2). Propidium iodide (PI) staining was used to measure the DNA content of cells to identify sub-G0/G1, G0/G1, S, and G2/M cells via flow cytometry, and all cells with sub-G0/G1 (<2n) DNA content were considered to have fragmented DNA. Curiously, EO from A. neoinsignis leaves caused DNA fragmentation in HepG2 cells and proportionally reduced all phases of the cell cycle.
We then detected the viability of HepG2 cells treated with EO from A. neoinsignis leaves at concentrations of 5, 10, and 15 μg/mL via YO-PRO-1/PI double-staining after 24 and 48 h of incubation (Figure 3). For this purpose, viable cells (YO-PRO-1/PI double-negative cells), apoptotic cells (YO-PRO-1-positive/PI-negative cells), and dead cells (YO-PRO-1/PI double-positive cells plus YO-PRO-1-negative/PI-positive cells) were quantified via flow cytometry. In this case, dead cells indicate cells with unidentified types of cell death. Interestingly, A. neoinsignis leaf EO decreased the number of viable cells while increasing the number of apoptotic cells, followed by dead cells.
The cell size and granularity/complexity were also determined via flow cytometry via forward scatter (FSC) and side scatter (SSC), respectively. Treatment of HepG2 cells with EO from A. neoinsignis leaves reduced the FSC (Figure 4) after 24 and 48 h of incubation, indicating a reduction in cell size and corroborating cell death by apoptosis. Furthermore, the mitochondrial transmembrane potential was also measured via rhodamine-123 staining in HepG2 cells treated with EO from A. neoinsignis leaves at concentrations of 5, 10, and 15 μg/mL after 24 h of incubation. A significant reduction in the mitochondrial transmembrane potential was also detected in HepG2 cells treated with EO from A. neoinsignis (Figure 5), confirming that this EO can cause apoptosis in liver cancer cells.
Apoptosis is a type of regulated cell death characterized by a series of unique morphological and biochemical changes, such as decreased cell size, blebbing, DNA fragmentation, phosphatidylserine externalization, loss of the mitochondrial membrane potential, and caspase activation [33,34,35]. In this study, we found that A. neoinsignis leaf EO induced apoptosis in liver cancer cells, as evidenced by DNA fragmentation, loss of mitochondrial transmembrane potential, and YO-PRO-1/PI double-staining. Previously, Bomfim et al. [16] reported that EO from A. vepretorum leaves causes apoptosis in B16-F10 melanoma cells, as observed by the induction of phosphatidylserine externalization without affecting cell membrane integrity. Similarly, EO extracted from A. squamosa pericarp also caused apoptosis in SMMC-7721 liver cancer cells, as indicated by shrinkage or fragmentation of the cell nucleus [17].
β-Elemene and (E)-caryophyllene, two of the main constituents of A. neoinsignis leaf EO, have also been previously reported as inducers of apoptosis. β-Elemene caused G2/M cell cycle arrest and apoptotic cell death in non-small cell lung cancer cells, as observed by the induction of caspase-3, -7, and -9 activities; cytochrome c release; decreased Bcl-2 expression; and increased levels of DNA fragmentation [36]. β-Elemene also promoted apoptosis in colorectal cancer cells, as observed by nuclear chromatin condensation and phosphatidylserine externalization, decreased mitochondrial membrane potential, and cleavage of the caspase-3/9 and PARP proteins [37]. (E)-Caryophyllene induced apoptosis in liver cancer cells, as observed by the externalization of phosphatidylserine and the cleavage of caspase-3 and PARP [38].

2.4. Effect of A. neoinsignis Leaf EO on Mice Xenografted with Liver Cancer Cells

The in vivo antitumor effect of A. neoinsignis leaf EO was measured in C.B.17 SCID mice bearing HepG2 xenografts. One day after tumor inoculation, the animals were treated daily for two weeks with vehicle (5% DMSO, control group) or A. neoinsignis leaf EO at a dosage of 40 mg/kg. At the end of treatment, the mean tumor weight in the control group was 0.76 ± 0.15 g, whereas in the mice treated with EO, the mean tumor weight was 0.35 ± 0.05 g (Figure 6A), indicating a significant decrease of 53.7% (Figure 6B). These data indicate that A. neoinsignis leaf EO can effectively inhibit tumor growth in vivo.
Histological analysis revealed tumors with morphologies consistent with moderately differentiated hepatocellular carcinoma, delineated by a connective tissue capsule. The intratumor extracellular matrix is predominantly composed of collagen fibers, with sparse vascularization. Extensive areas of coagulative necrosis were observed throughout the tumor, generally accompanied by inflammatory cell infiltration in the peripheral regions. In certain areas, neoplastic cells are arranged in clusters resembling islands surrounded by a collagenous matrix or located near necrotic zones. Additionally, foci of invasion into adipose and cartilaginous tissues were identified. Tumors in the EO group presented granulation tissue adjacent to the necrotic areas (Figure 6C).
All the animals were weighed at the beginning and end of the experiment to assess weight gain or loss. The wet weights of the liver, heart, lungs, and kidneys were also determined to assess the toxicological potential of A. neoinsignis leaf EO. Interestingly, no significant changes in animal or organ weight were detected in the mice treated with A. neoinsignis leaf EO for two weeks at a dosage of 40 mg/kg (Figure S47).
The systemic toxicity of the EO was assessed through histological analysis of various organs. The tissue architecture of the heart and kidneys remained preserved in both groups, with only mild vascular hyperemia observed. In contrast, the hepatic parenchyma was partially preserved, with moderate hydropic degeneration of hepatocytes. Vascular hyperemia was also noted in the central venules and vessels of the portal triad, accompanied by inflammatory cell infiltration in these regions. The pulmonary parenchyma exhibited significant alterations, including areas of atelectasis resulting from thickening of the alveolar septa caused by hyperplasia and hypertrophy of pneumocytes, as well as hyperemia of the pulmonary capillaries. Additionally, focal areas of necrosis were identified in the bronchial epithelium, along with moderate active hyperemia and focal hemorrhages in the lung tissue (Figure S48).
In previous studies, A. vepretorum leaf EO, when microencapsulated with β-cyclodextrin and administered intraperitoneally at a dose of 50 mg/kg for 11 consecutive days, reduced the growth of B16-F10 melanoma cells in C57BL/6 mice by 62.66%, with no systemic toxicity detected [16]. Similarly, A. leptopetala EO inhibited sarcoma 180 tumor growth in Swiss mice by 59.29% and 58.77% after 7 days of intraperitoneal treatment at doses of 100 and 150 mg/kg, respectively. Moderate gastrointestinal toxicity was observed, but no genotoxicity was found at a dose of 350 mg/kg [18].
In conclusion, A. neoinsignis leaf EO demonstrated potent cytotoxicity to cancer cells, with liver cancer cells being sensitive. Furthermore, this EO caused apoptotic cell death in liver cancer cells and had the ability to reduce tumor growth in a human liver cancer xenograft model. These data suggest that A. neoinsignis leaf EO is an important source of anti-liver cancer drugs. Additionally, these results are described for the first time in A. neoinsignis, which suggests the need for further studies on its chemical and cytotoxic activity against tumor cell lines, as well as other biological activities, including mechanism of action and safety studies.

3. Materials and Methods

3.1. Botanical Material

The leaves of A. neoinsignis were collected in July 2024 at coordinates 3°05′51.2″ S and 59°58′34.7″ W on the campus of the Federal University of Amazonas (UFAM) in the metropolitan region of Manaus, Amazonas, Brazil. The species was identified by the biologist Deisy Pereira Saraiva from UFAM. An exsiccate of the species was deposited in the herbarium of the Department of Biology of UFAM (HUAM) under number 12,577. The accession received registration number A6E79F4 from the National System for the Management of Genetic Heritage and Associated Traditional Knowledge (SISGEN, Brazil).

3.2. EO Extraction

The leaves of A. neoinsignis were dried for 24 h in an oven with air circulation at 40 °C and then ground in a four-blade mill. The EO was extracted via the hydrodistillation method via a Clevenger system coupled to a 4 L round-bottom flask and boiled at 100 °C. The extractions were performed in triplicate, with 300 g of plant material added to each one, followed by distilled water to half the level of the flask. The EO was extracted for 3 h. Then, the oil was collected and filtered with anhydrous sodium sulfate to eliminate residual water and then stored in an amber bottle under refrigeration to avoid loss or degradation of chemicals. The oil yield, expressed as a percentage, was calculated via the following formula: Oil yield (%) = (mass of oil/mass of plant material) × 100.

3.3. Analysis of the Chemical Constituents

EO analysis was performed with a TRACE GC ULTRA/ISQ gas chromatograph (Thermo Scientific, Waltham, MA, USA) connected to an ISQ mass spectrometer equipped with a Tri Plus RSH autosampler (GC–MS) and flame ionization detector (GC–FID). A DB-5MS fused silica capillary column (film thickness of 30 m × 0.25 mm × 0.25 μm) coated with 5% phenylarylene-95% dimethylpolysiloxane was used for compound separation. Helium was employed as the retention gas, with a flow rate of 1.0 mL/min. The initial temperature was set at 40 °C for 4 min, followed by a flow rate of 4 °C/min up to 240 °C, then 10 °C/min up to 280 °C, and finally 280 °C for 2 min. The injector temperature was 250 °C, while the detector temperature was 280 °C [39]. The samples were produced by dissolving 10 mg in 1 mL of HPLC-grade ethyl acetate and injecting 1 μL of the solution in split mode at a ratio of 1:25. A typical solution of n-alkanes (C8–C20) was used to establish the retention indices, and calculations were performed via the van den Dool and Kratz equation [21]. The peak areas and retention times agreed electronically with an integrator. The stationary phase for GC–MS analysis was a DB-5MS fused silica capillary column (30 m × 0.25 mm × 0.25 μm film thickness) coated with 5% phenylarylene-95% dimethylpolysiloxane. Mass spectra were obtained at 70 eV with 0.5 s scan intervals and fragments ranging in size from 40 to 550 Da. Other analysis settings were similar to those used for the GC–FID analysis.
To identify the volatile constituents, the collected mass spectra were compared with those available in the literature [22], and retention indices were used. The proportion of each substance was calculated by dividing its area by the total area of all the substances in the sample and multiplying by 100.

3.4. Cytotoxicity Assay

The cancer cell lines HepG2 (human liver cancer), HCT116 (human colon cancer), MCF-7 (human breast cancer), MDA-MB-231 (human breast cancer), 4T1 (mouse breast cancer), and B16-F10 (mouse melanoma), together with the noncancerous cell line MRC-5 (human lung fibroblast), were used in this study. All cell lines were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA) and maintained following the ATCC animal cell culture guidelines [40]. To confirm the use of mycoplasma-free cells, all the cells were tested for mycoplasma with a mycoplasma staining kit (Sigma–Aldrich, St. Louis, MO, USA).
The Alamar blue assay was used to quantify the cell viability as described previously [41]. The cells were cultured in 96-well plates for each experiment at a concentration of 3 × 104 cells/well for nonadherent cells or 7 × 103 cells/well for adherent cells. EO was dissolved in pure dimethyl sulfoxide (DMSO, Vetec Química Fina Ltda., Duque de Caxias, RJ, Brazil), diluted in culture medium, and subsequently added (at eight different concentrations) to each well prior to a 72 h incubation. The DMSO concentration did not exceed 0.5%. Doxorubicin (IMA S.A.I.C., Buenos Aires, Argentina) served as a positive control, while untreated cells served as negative control. At the end of the treatment, 20 μL of 1 mM resazurin (Sigma–Aldrich, St. Louis, MO, USA) was added to each well. A SpectraMax 190 microplate reader was used to measure the absorbance at 570 and 600 nm (Molecular Devices, Sunnyvale, CA, USA). The percentage of cell inhibition was normalized to that of the negative control group. The IC50 values with 95% confidence intervals were obtained via nonlinear regression.

3.5. DNA Fragmentation and Cell Cycle Distribution Analysis

Cells stained with PI were used to quantify the cellular DNA content and measure internucleosomal DNA fragmentation and the cell cycle distribution [42]. Briefly, the cells were collected in a permeabilization solution containing 100 μg/mL RNase, 2 μg/mL PI, 0.1% Triton X-100, and 0.1% sodium citrate (all from Sigma–Aldrich, St. Louis, MO, USA), and flow cytometry was used to assess cellular fluorescence. A minimum of 104 events were acquired for each sample. A BD LSRFortessa cytometer was used in conjunction with BD FACSDiva (BD Biosciences, San Jose, CA, USA) and FlowJo 10 (FlowJo LLC; Ashland, OR, USA) software. Cell debris was excluded from the analysis, whereas single cells were selected via H-FSCs vs. A-FSCs and/or H-SSCs vs. A-SSCs.

3.6. Apoptosis Staining Assay

YO-PRO-1 (Sigma–Aldrich, St. Louis, MO, USA) and PI (BD Biosciences, San Jose, CA, USA) dyes were used to quantify the percentage of apoptotic cells in culture, where YO-PRO-1-positive/PI-negative cells were considered apoptotic cells (early stage) [43]. Briefly, the cells were stained with a solution containing 1.5 µM PI plus 0.1 µM YO-PRO-1, and the cellular fluorescence was assessed via flow cytometry as described above.

3.7. Mitochondrial Transmembrane Potential Analysis

Cells stained with rhodamine-123 dye were used to evaluate the mitochondrial transmembrane potential [44]. The cells were incubated with 1 μg/mL rhodamine-123 (Sigma–Aldrich, St. Louis, MO, USA) for 15 min at 37 °C in the dark and then rinsed, after which the cellular fluorescence was quantified via flow cytometry as described above.

3.8. Human Liver Cancer Xenograft Model

Sixteen male specific pathogen-free C. B-17 SCID mice (8 weeks old and weighing 20–25 g) were acquired and housed in the vivarium of Fiocruz-Bahia (Salvador, Bahia, Brazil). The animals were kept in polyacrylic cages, with a maximum of five mice per unit, receiving food and water ad libitum. The environment was controlled with a 12 h light–dark cycle, with the light phase starting at 7 am. All experimental procedures were previously approved by the institution’s animal ethics committee (protocol no. 01/2021).
To establish the human liver cancer xenograft model, HepG2 cells (107 cells/500 µL/animal) were injected subcutaneously into the left frontal axilla of the mice. After a single day, the animals received intraperitoneal treatment (200 µL/animal) daily for two weeks. The animals were divided into two groups: 1. Animals that received injections of vehicle (5% DMSO solution) used to dilute the EO (n = 8); and 2. Animals that received injections of EO at 40 mg/kg (n = 8). One day after the end of treatment, an overdose of anesthetic (ketamine–xylazine) was administered to euthanize the animals, after which the tumors were removed and weighed. The inhibition ratio (percentage) was determined via the following formula: inhibition ratio (percentage) = [(A − B)/A] × 100, where A represents the mean tumor weight of the negative control and B represents the tumor weight of the treatment group.
To evaluate the toxicological effects, the mice were weighed at the beginning and end of the experiment. Throughout the experimental period, the animals were monitored for the presence of behavioral or physiological changes. After euthanasia, the liver, kidneys, lungs and heart were removed and examined for significant lesions, color changes and/or evidence of hemorrhage. Histological analyses of the tumors and organs were performed via light microscopy after they were stained with hematoxylin–eosin (H and E) and periodic acid–Schiff (PAS), the latter being applied to the liver and kidneys. The tissues were previously fixed in 4% formaldehyde solution. Histopathological evaluation was performed via light microscopy at magnifications of 4×, 100×, 200×, and 400×. The distributed parts were classified as absent (0), mild (+1), moderate (+2), and intense (+3). Representative photomicrographs were acquired via LAS v4 image acquisition software (LMD6500), adapted to the Leica specification.

3.9. Statistical Analysis

The data are presented as the means ± standard errors of the means (S.E.M.) or as IC50 values with 95% confidence intervals (95% CI) from at least three independent experiments performed in duplicate or triplicate. Two-tailed unpaired Student’s t test or one-way ANOVA followed by Dunnett’s test were used to compare differences among experimental groups (p < 0.05). GraphPad Prism version 8 was used to perform all the statistical analyses (Intuitive Software for Science; San Diego, CA, USA).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules30142971/s1, Figure S1: chromatogram of the total ions; Figure S2–S45: Mass spectrum of compounds. Figure S46: Concentration–response curves; Figure S47: The body weight and relative organ weight; Figure S48: Representative photomicrographs of organs.

Author Contributions

Conceptualization, M.P.S., M.V.L.d.C., M.B.P.S., E.V.C. and D.P.B.; methodology, M.P.S., M.V.L.d.C., G.A.d.C.B., S.G.C., A.M.R.M.C., R.B.D., E.V.C., M.B.P.S. and D.P.B.; formal analysis, M.P.S., M.V.L.d.C., E.V.C. and D.P.B.; investigation, M.P.S., M.V.L.d.C., G.A.d.C.B., S.G.C., A.M.R.M.C., R.B.D., E.V.C., M.B.P.S. and D.P.B.; data curation M.P.S., M.V.L.d.C., R.B.D., E.V.C., M.B.P.S. and D.P.B.; writing—original draft preparation, D.P.B.; writing—review and editing, M.P.S., M.V.L.d.C., G.A.d.C.B., R.B.D., E.V.C. and D.P.B.; visualization, M.P.S., M.V.L.d.C., G.A.d.C.B., S.G.C., A.M.R.M.C., R.B.D., E.V.C., M.B.P.S. 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, 001), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), FitoAmazônia Research Network (Pró-Amazônia/CNPq process 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; and date of approval: 6 April 2021). All methods were performed in accordance with the relevant 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 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 botanical materials. Melissa P. Souza would like to thank FAPEAM for the research grant.

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 Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. Guo, Q.; Zhu, X.; Beeraka, N.M.; Zhao, R.; Li, S.; Li, F.; Mahesh, P.A.; Nikolenko, V.N.; Fan, R.; Liu, J. Projected epidemiological trends and burden of liver cancer by 2040 based on GBD, CI5plus, and WHO data. Sci. Rep. 2024, 14, 28131. [Google Scholar] [CrossRef] [PubMed]
  3. 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] [PubMed]
  4. Ducreux, M.; Abou-Alfa, G.K.; Bekaii-Saab, T.; Berlin, J.; Cervantes, A.; de Baere, T.; Eng, C.; Galle, P.; Gill, S.; Gruenberger, T.; et al. The management of hepatocellular carcinoma. Current expert opinion and recommendations derived from the 24th ESMO/World Congress on Gastrointestinal Cancer, Barcelona, 2022. ESMO Open 2023, 8, 101567. [Google Scholar] [CrossRef] [PubMed]
  5. Ntellas, P.; Chau, I. Updates on Systemic Therapy for Hepatocellular Carcinoma. Am. Soc. Clin. Oncol. Educ. Book. 2024, 44, e430028. [Google Scholar] [CrossRef] [PubMed]
  6. Zheng, J.; Wang, S.; Xia, L.; Sun, Z.; Chan, K.M.; Bernards, R.; Qin, W.; Chen, J.; Xia, Q.; Jin, H. Hepatocellular carcinoma: Signaling pathways and therapeutic advances. Signal Transduct. Target Ther. 2025, 10, 35. [Google Scholar] [CrossRef] [PubMed]
  7. Rainer, H. Monographic studies in the genus Annona L. (Annonaceae): Inclusion of the genus Rollinia A.ST.-HIL. Ann. Des. Naturhistorischen Mus. Wien. 2007, 108, 191–205. [Google Scholar]
  8. Oliveira, A.N.; Amaral, I.L.; Ramos, M.B.P.; Nobre, A.D.; Couto, L.B.; Sahdo, R.M. Composition and floristic-structural diversity of a hectare of terra firme dense forest in Central Amazonia, Amazonas, Brazil. Acta Amaz. 2008, 38, 627–642. [Google Scholar] [CrossRef]
  9. Leite, D.O.D.; de FA Nonato, C.; Camilo, C.J.; de Carvalho, N.K.G.; da Nobrega, M.G.L.A.; Pereira, R.C.; da Costa, J.G.M. Annona Genus: Traditional Uses, Phytochemistry and Biological Activities. Curr. Pharm. Des. 2020, 26, 4056–4091. [Google Scholar] [CrossRef] [PubMed]
  10. Leal, F.; Paull, R.E. The genus Annona: Botanical characteristics, horticultural requirements and uses. Crop Sci. 2023, 63, 1030–1049. [Google Scholar] [CrossRef]
  11. 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/FB110253 (accessed on 9 May 2025).
  12. Jürgens, A.; Webber, A.C.; Gottsberger, G. Floral scent compounds of Amazonian Annonaceae species pollinated by small beetles and thrips. Phytochemistry 2000, 55, 551–558. [Google Scholar] [CrossRef]
  13. 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]
  14. 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]
  15. 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]
  16. 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]
  17. 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]
  18. 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]
  19. 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-Int. J. Exp. Bot. 2021, 90, 921. [Google Scholar] [CrossRef]
  20. 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]
  21. 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] [PubMed]
  22. Adams, R.P. Identification of Essential Oil Components by Gas Chromatography/Mass Spectroscopy, 4th ed.; Allured Publishing: Carol Stream, IL, USA, 2007; p. 803. [Google Scholar]
  23. Souza, T.J.T.; Zanetti, G.D.; Apel, M.A.; Henriques, A.T.; Manfron, M.P. Characterization of seasonal and chemotypical variability in the essential oil from leaves of Annona neosalicifolia H. Rainer (Annonaceae). Nat. Volatiles Essent. Oils 2021, 8, 81–91. [Google Scholar] [CrossRef]
  24. Siqueira, C.A.T.; Oliani, J.; Sartoratto, A.; Queiroga, C.L.; Moreno, P.R.H.; Reimão, J.Q.; Tempone, A.G.; Fischer, D.C.H. Chemical constituents of the volatile oil from leaves of Annona coriacea and in vitro antiprotozoal activity. Rev. Bras. Farmacog. 2011, 21, 33–40. [Google Scholar] [CrossRef]
  25. Costa, E.V.; Dutra, L.M.; Nogueira, P.C.L.; Moraes, V.R.S.; Salvador, M.J.; Ribeiro, L.H.G.; Gadelha, F.R. Essential oil from the leaves of Annona vepretorum: Chemical composition and bioactivity. Nat. Prod. Commun. 2012, 7, 265–266. [Google Scholar] [CrossRef] [PubMed]
  26. Verma, R.S.; Joshi, N.; Padalia, R.C.; Singh, V.R.; Goswami, P.; Chauhan, A. Characterization of the leaf essential oil composition of Annona squamosa L. from foothills of north India. Med. Arom. Plants 2016, 5, 5–9. [Google Scholar] [CrossRef]
  27. Joseph, S.M.; Dev, A.R.A.; Kanchana, A. Unveiling the chemical variations of Annona essential oils and its associated pharmacological activities. J. Mol. Struct. 2023, 1292, 136082. [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. Virtual Quim. 2022, 14, 1–7. [Google Scholar] [CrossRef]
  29. Palazzo, M.C.; Agius, B.R.; Wright, B.S.; Haber, W.A.; Moriarity, D.M.; Setzer, W.N. Chemical compositions and cytotoxic activities of leaf essential oils of four Lauraceae tree species from Monteverde, Costa Rica. Rec. Nat. Prod. 2009, 3, 32–37. [Google Scholar]
  30. Loizzo, M.R.; Tundis, R.; Menichini, F.; Saab, A.M.; Statti, G.A.; Menichini, F. Antiproliferative effects of essential oils and their major constituents in human renal adenocarcinoma and amelanotic melanoma cells. Cell Prolif. 2008, 41, 1002–1012. [Google Scholar] [CrossRef] [PubMed]
  31. Feng, Y.; An, Q.; Zhao, Z.; Wu, M.; Yang, C.; Liang, W.; Xu, X.; Jiang, T.; Zhang, G. Beta-elemene: A phytochemical with promise as a drug candidate for tumor therapy and adjuvant tumor therapy. Biomed. Pharmacother. 2024, 172, 116266. [Google Scholar] [CrossRef] [PubMed]
  32. Chaturvedula, V.S.; Schilling, J.K.; Miller, J.S.; Andriantsiferana, R.; Rasamison, V.E.; Kingston, D.G. New cytotoxic terpenoids from the wood of Vepris punctata from the Madagascar Rainforest. J. Nat. Prod. 2004, 67, 895–898. [Google Scholar] [CrossRef] [PubMed]
  33. Vitale, I.; Pietrocola, F.; Guilbaud, E.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostini, M.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; et al. Apoptotic cell death in disease-Current understanding of the NCCD 2023. Cell Death Differ. 2023, 30, 1097–1154. [Google Scholar] [CrossRef] [PubMed]
  34. Park, W.; Wei, S.; Kim, B.S.; Kim, B.; Bae, S.J.; Chae, Y.C.; Ryu, D.; Ha, K.T. Diversity and complexity of cell death: A historical review. Exp. Mol. Med. 2023, 55, 1573–1594. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, Y.; Li, X.; Yang, M.; Liu, S.B. Research progress on morphology and mechanism of programmed cell death. Cell Death Dis. 2024, 15, 327. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, G.; Li, X.; Huang, F.; Zhao, J.; Ding, H.; Cunningham, C.; Coad, J.E.; Flynn, D.C.; Reed, E.; Li, Q.Q. Antitumor effect of beta-elemene in non-small-cell lung cancer cells is mediated via induction of cell cycle arrest and apoptotic cell death. Cell. Mol. Life Sci. 2005, 62, 881–893. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, G.Y.; Zhang, L.; Geng, Y.D.; Wang, B.; Feng, X.J.; Chen, Z.L.; Wei, W.; Jiang, L. β-Elemene induces apoptosis and autophagy in colorectal cancer cells through regulating the ROS/AMPK/mTOR pathway. Chin. J. Nat. Med. 2022, 20, 9–21. [Google Scholar] [CrossRef] [PubMed]
  38. Basheer, I.; Wang, H.; Li, G.; Jehan, S.; Raza, A.; Du, C.; Ullah, N.; Li, D.; Sui, G. β-caryophyllene sensitizes hepatocellular carcinoma cells to chemotherapeutics and inhibits cell malignancy through targeting MAPK signaling pathway. Front. Pharmacol 2024, 15, 1492670. [Google Scholar] [CrossRef] [PubMed]
  39. 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] [PubMed]
  40. 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 22 January 2024).
  41. 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] [PubMed]
  42. 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]
  43. 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] [PubMed]
  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]
Figure 1. Main compounds identified in A. neoinsignis leaf EO.
Figure 1. Main compounds identified in A. neoinsignis leaf EO.
Molecules 30 02971 g001
Figure 2. DNA fragmentation (sub-G0/G1 cells) and cell cycle phases (G0/G1, S, and G2/M) of HepG2 cells treated with A. neoinsignis leaf EO for 24 and 48 h. (A) Representative flow cytometric histograms. (B) Percentages of cells in sub-G0/G1, G0/G1, S, and G2/M. Vehicle (0.2% DMSO) was used as a negative control (CTL). The data are shown as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test.
Figure 2. DNA fragmentation (sub-G0/G1 cells) and cell cycle phases (G0/G1, S, and G2/M) of HepG2 cells treated with A. neoinsignis leaf EO for 24 and 48 h. (A) Representative flow cytometric histograms. (B) Percentages of cells in sub-G0/G1, G0/G1, S, and G2/M. Vehicle (0.2% DMSO) was used as a negative control (CTL). The data are shown as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test.
Molecules 30 02971 g002
Figure 3. Effects of A. neoinsignis leaf EO on the induction of apoptosis in HepG2 cells after 24 and 48 h of treatment. (A) Representative flow cytometric dot plots. (B) Quantification of viable (YO-PRO-1/PI double-negative cells, blue bars), apoptotic (YO-PRO-1-positive/PI-negative cells, red bars), and dead (YO-PRO-1/PI double-positive cells plus YO-PRO-1-negative/PI-positive cells, green bars) cells. Vehicle (0.2% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test.
Figure 3. Effects of A. neoinsignis leaf EO on the induction of apoptosis in HepG2 cells after 24 and 48 h of treatment. (A) Representative flow cytometric dot plots. (B) Quantification of viable (YO-PRO-1/PI double-negative cells, blue bars), apoptotic (YO-PRO-1-positive/PI-negative cells, red bars), and dead (YO-PRO-1/PI double-positive cells plus YO-PRO-1-negative/PI-positive cells, green bars) cells. Vehicle (0.2% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test.
Molecules 30 02971 g003
Figure 4. Effect of A. neoinsignis leaf EO on the cell size and granularity/complexity of HepG2 cells, as assessed by light scattering characteristics (FSC: forward scatter; SSC: side scatter) detected by flow cytometry after 24 and 48 treatments. (A) Representative flow cytometric dot plots. (B) Quantification of FSC and SSC. Vehicle (0.2% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test.
Figure 4. Effect of A. neoinsignis leaf EO on the cell size and granularity/complexity of HepG2 cells, as assessed by light scattering characteristics (FSC: forward scatter; SSC: side scatter) detected by flow cytometry after 24 and 48 treatments. (A) Representative flow cytometric dot plots. (B) Quantification of FSC and SSC. Vehicle (0.2% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test.
Molecules 30 02971 g004
Figure 5. Mitochondrial depolarization in HepG2 cells treated with EO from A. neoinsignis leaves for 24 h. Mitochondrial potential was assessed with rhodamine-123-stained cells via flow cytometry. Vehicle (0.5% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test. MFI = Mean fluorescence intensity.
Figure 5. Mitochondrial depolarization in HepG2 cells treated with EO from A. neoinsignis leaves for 24 h. Mitochondrial potential was assessed with rhodamine-123-stained cells via flow cytometry. Vehicle (0.5% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of three biological replicates performed in triplicate. * p < 0.05 compared with CTL by one-way ANOVA followed by Dunnett’s multiple comparisons test. MFI = Mean fluorescence intensity.
Molecules 30 02971 g005
Figure 6. Effects of treatment with A. neoinsignis leaf EO on tumor weight (A) and tumor inhibition (B) in C.B.17 SCID mice bearing HepG2 cell xenografts. (C) Representative photomicrographs of HepG2 tumors. The treatments (40 mg/kg EO) were injected intraperitoneally into the mice daily for two weeks. Vehicle (5% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of 8 animals. * p < 0.05 compared with CTL by two-tailed unpaired Student’s t test.
Figure 6. Effects of treatment with A. neoinsignis leaf EO on tumor weight (A) and tumor inhibition (B) in C.B.17 SCID mice bearing HepG2 cell xenografts. (C) Representative photomicrographs of HepG2 tumors. The treatments (40 mg/kg EO) were injected intraperitoneally into the mice daily for two weeks. Vehicle (5% DMSO) was used as a negative control (CTL). The data are presented as the means ± S.E.M.s of 8 animals. * p < 0.05 compared with CTL by two-tailed unpaired Student’s t test.
Molecules 30 02971 g006
Table 1. Chemical composition of the EO extracted from the leaves of A. neoinsignis.
Table 1. Chemical composition of the EO extracted from the leaves of A. neoinsignis.
Compounds RI aRI bPeak Area %
1β-Pinene9709740.04 ± 0.00
2Myrcene9889880.11 ± 0.01
3Limonene102710240.07 ± 0.01
4(Z)-β-Ocimene103510320.04 ± 0.00
5(E)-β-Ocimene104610440.20 ± 0.02
6Terpinolene108310860.02 ± 0.00
7Linalool109810950.39 ± 0.02
8Terpinen-4-ol117911740.05 ± 0.00
9α-Terpineol119311860.24 ± 0.05
10Nerol122312270.01 ± 0.00
11Geraniol125012490.04 ± 0.00
12δ-Elemene133413352.27 ± 0.20
13α-Cubebene134613480.64 ± 0.07
14α-Ylangene136713730.41 ± 0.10
15α-Copaene137413742.52 ± 0.25
16β-Elemene1389138929.61 ± 3.80
17(E)-Caryophyllene1419141718.23 ± 0.43
18γ-Elemene142814344.47 ± 0.04
19α-trans-Bergamotene143214320.87 ± 0.02
20Aromadendrene143714390.05 ± 0.00
21cis-Muurola-3,5-diene144814480.10 ± 0.00
22α-Humulene145414523.33 ± 0.06
23cis-Cadina-1(6),4-diene146114610.05 ± 0.00
24γ-Muurolene147314780.73 ± 0.02
25Germacrene D14.80148015.34 ± 2.13
26β-Selinene148814891.69 ± 0.07
27Bicyclogermacrene149415004.61 ± 0.15
28α-Muurolene149715000.20 ± 0.01
29Germacrene A150615080.82 ± 0.08
30γ-Cadinene151115130.28 ± 0.00
31δ-Amorphene151615111.81 ± 0.06
32trans-Calamenene152015210.12 ± 0.04
33(E)-γ-Bisabolene152515290.57 ± 0.01
34trans-Cadina-1(2),4-diene153115330.21 ± 0.01
35α-Cadinene153515370.32 ± 0.01
36Selina-3,7(11)-diene154015450.15 ± 0.00
37Germacrene B155815596.80 ± 0.39
38Spathulenol157515770.28 ± 0.02
39Caryophyllene oxide158015820.65 ± 0.02
40Globulol158415900.10 ± 0.00
411-epi-Cubenol162616270.33 ± 0.00
42Cubenol164216450.25 ± 0.01
43α-Cadinol165316520.27 ± 0.01
44neo-Intermedeol165716580.55 ± 0.01
Monoterpenes 1.22%
Sesquiterpenes 98.63%
Total Identified 99.84%
Total Not Identified (N.I.)0.15%
Note: The data are presented as the means ± S.D.s of three analyses. RI a (experimental 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 [21], which is based on a homologous series of normal alkanes. RI b (literature retention indices): according to Adams [22]. N.I. = Not identified.
Table 2. Cytotoxicity of A. neoinsignis leaf EO.
Table 2. Cytotoxicity of A. neoinsignis leaf EO.
CellsHistological TypeIC50 and 95% CI (in μg/mL)
DOXEO
Cancer cells
HepG2Human liver cancer0.03
0.02–0.04
27.90
20.44–36.66
HCT116Human colon cancer0.05
0.04–0.06
24.35
16.86–37.23
MCF-7Human breast cancer0.84
0.65–1.12
37.50
30.07–55.21
MDA-MB-231Human breast cancer0.61
0.47–0.82
31.78
24.42–44.90
4T1Mouse breast cancer0.60
0.46–0.81
21.64
17.05–27.32
B16-F10Mouse melanoma0.06
0.04–0.08
12.28
10.08–14.85
Noncancerous cells
MRC-5Human lung fibroblast1.94
1.40–2.88
>50
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

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. https://doi.org/10.3390/molecules30142971

AMA Style

Souza MP, de Castro MVL, Barbosa GAdC, Carvalho SG, Coelho AMRM, Dias RB, Soares MBP, Costa EV, Bezerra DP. Essential Oil from the Leaves of Annona neoinsignis H. Rainer (Annonaceae) Against Liver Cancer: In Vitro and In Vivo Studies. Molecules. 2025; 30(14):2971. https://doi.org/10.3390/molecules30142971

Chicago/Turabian Style

Souza, Melissa P., Maria V. L. de Castro, Gabriela A. da C. Barbosa, Sabrine G. Carvalho, Amanda M. R. M. Coelho, Rosane B. Dias, Milena B. P. Soares, Emmanoel V. Costa, and Daniel P. Bezerra. 2025. "Essential Oil from the Leaves of Annona neoinsignis H. Rainer (Annonaceae) Against Liver Cancer: In Vitro and In Vivo Studies" Molecules 30, no. 14: 2971. https://doi.org/10.3390/molecules30142971

APA Style

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. (2025). Essential Oil from the Leaves of Annona neoinsignis H. Rainer (Annonaceae) Against Liver Cancer: In Vitro and In Vivo Studies. Molecules, 30(14), 2971. https://doi.org/10.3390/molecules30142971

Article Metrics

Back to TopTop