Next Article in Journal
Plant Protease Inhibitors as Emerging Antimicrobial Peptide Agents: A Comprehensive Review
Previous Article in Journal
Vaccination with DC-SIGN-Targeting αGC Liposomes Leads to Tumor Control, Irrespective of Suboptimally Activated T-Cells
Previous Article in Special Issue
Antimutagenicity and Antioxidant Activity of Castanea sativa Mill. Bark Extract
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Effects of Essential Oils and Their Bioactive Compounds on Prostate Cancer Treatment

by
Leticia Santos Pimentel
*,
Luciana Machado Bastos
,
Luiz Ricardo Goulart
and
Lígia Nunes de Morais Ribeiro
*
Laboratory of Nanobiotechnology Professor Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, Bloco 2E, Sala 248, Uberlândia 38405-302, MG, Brazil
*
Authors to whom correspondence should be addressed.
Deceased author.
Pharmaceutics 2024, 16(5), 583; https://doi.org/10.3390/pharmaceutics16050583
Submission received: 25 March 2024 / Revised: 18 April 2024 / Accepted: 19 April 2024 / Published: 24 April 2024
(This article belongs to the Special Issue Natural Products for Anticancer Application)

Abstract

:
Since prostate cancer (PCa) relies on limited therapies, more effective alternatives are required. Essential oils (EOs) and their bioactive compounds are natural products that have many properties including anticancer activity. This review covers studies published between 2000 and 2023 and discusses the anti-prostate cancer mechanisms of the EOs from several plant species and their main bioactive compounds. It also provides a critical perspective regarding the challenges to be overcome until they reach the market. EOs from chamomile, cinnamon, Citrus species, turmeric, Cymbopogon species, ginger, lavender, Mentha species, rosemary, Salvia species, thyme and other species have been tested in different PCa cell lines and have shown excellent results, including the inhibition of cell growth and migration, the induction of apoptosis, modulation in the expression of apoptotic and anti-apoptotic genes and the suppression of angiogenesis. The most challenging aspects of EOs, which limit their clinical uses, are their highly lipophilic nature, physicochemical instability, photosensitivity, high volatility and composition variability. The processing of EO-based products in the pharmaceutical field may be an interesting alternative to circumvent EOs’ limitations, resulting in several benefits in their further clinical use. Identifying their bioactive compounds, therapeutic effects and chemical structures could open new perspectives for innovative developments in the field. Moreover, this could be helpful in obtaining versatile chemical synthesis routes and/or biotechnological drug production strategies, providing an accurate, safe and sustainable source of these bioactive compounds, while looking at their use as gold-standard therapy in the close future.

1. Introduction

1.1. Overview of Prostate Cancer

Cancer has been considered one of the most alarming diseases in the last few decades, worldwide. It is a term used to cover more than 200 multifactorial diseases characterized by the uncontrolled growth and invasion of abnormal cells leading to the formation of tumors in healthy tissues [1]. Prostate cancer (PCa) is the second most prevalent cancer in men across the world and the fourth overall [2]. Its risks factors include age, ethnicity, diet, genetic, environmental factors and others. The incidence rate for men under the age of 50 years is 1 in 350 men, but it increases to nearly 60% in men over the age of 65 years [3]. PCa is a highly heterogeneous disease whose subtypes remain poorly understood [4].
In the early stages, PCa’s primary treatment is based on radiation and surgery. However, up to 53% of these cases progress to recurrence. Recurrence or metastatic tumors are usually treated with androgen deprivation therapy (ADT) in the form of gonadotropin-releasing hormone (GnRH) and androgen receptor (AR) antagonists, so progression can be prevented and patient survival can be improved. However, in most cases, despite their initial response, the cells become therapy-resistant, progressing to castration-resistant PCa (CRPC) and metastatic CRPC (mCRPC), which lead to lethal disease [5]. CRPCs are characterized by the consecutive activation of AR signaling through the expression of AR variants, AR gene/enhancer amplification, AR mutations, the overexpression of AR coactivators, AR-independent pathways and other mechanisms [6,7].
In PCa cells, defective phosphatase and tensin homolog (PTEN) and the uncontrolled activation of phosphatidylinositol-3 kinase (PI3K)/AKT signaling frequently promotes cancer progression (Figure 1). The tumor suppressor PTEN is frequently mutated and shows a loss of function in PCa, allowing for the strong activation of the PI3K/AKT signaling pathway. Downstream of PI3K and AKT, the protein kinase mechanistic target of rapamycin (mTOR) plays an important role in cell growth regulation, and is often associated with tumorigenesis [8]. Other target molecules of downstream PI3K/AKT signaling, such as cyclin-dependent kinases (CDKs) and forkhead box subgroup O (FoxO), contribute to suppressing cell cycle control and apoptotic mechanisms, leading to PCa’s resistance to chemotherapeutic drugs [9].
The Ubiquitin-Proteasome (UPS) pathway plays a key role in the degradation of intracellular proteins involved in the regulation of several cellular processes, such as the cell cycle, DNA damage response, cell growth, apoptosis, angiogenesis and others (Figure 1). In PCa cells, the degradation of PTEN, cyclin-dependent kinase inhibitor p27 and Bcl2-associated x (Bax) by the UPS pathway are increased [10].
DNA damage repair (DDR) is another pathway altered in PCa cells. DDR deficiency induces cell dependence on poly (ADP-ribose) polymerase (PARP)-1 protein for DNA repair. The use of a PARP-1 inhibitor leads to PCa cell death [11,12].
Treatment options for PCa remain limited once chemotherapeutics present severe side effects, acting not only on cancer cells but also on normal tissues. Therefore, searching for new safe and efficient active compounds to treat PCa is necessary [13]. In this sense, natural products could be great potential therapeutics in cancer treatment, given that several pharmaceutical drugs have been developed from compounds derived from plants.

1.2. PCa Culture Cell Lines

Before proceeding with this review, it is important to understand the main PCa cell line models. DU145, PC3 and LNCaP were the first three PCa cell lines established from metastasized tumors and are the most frequently used to date, even with several other cell lines and sublines having been developed [14]. DU145 is an androgen-resistant cell line isolated in 1975 from the brain-metastatic PCa of a 69-year-old white man [15]. The androgen-resistant PC3 cell line was stablished in 1979 and derived from the lumbar-vertebral-metastatic PCa of a 62-year-old white man [16]. A subline was developed in 1984 from a PC3 xenograft tumor in an athymic mouse, called PC-3M, and revealed to be more aggressive than its parental PC3 cell line [17]. The androgen-responsive LNCaP cell line was established in 1980 and obtained from the needle aspiration biopsy of a lymph node metastatic lesion from a 50-year-old white man [18]. A subline from the vertebral metastasis of LNCaP xenografts in castrated mice was isolated in 1994 and called the C4-2 cell line. These cells became androgen-resistant upon interaction with bone fibroblasts [19]. Another subline (C4-2B) was developed from the bone metastasis of C4-2 xenografts in the same year [20]. The 22Rv1 is an androgen-responsive cell line isolated in 1999 from a CWR22R xenograft tumor in mice, which was obtainedfrom a patient with bone metastasis [21].

1.3. Essential Oils: Their Nature and Biological Activities

Essential oils (EOs) are natural lipid products present in various aromatic plants, derived from their secondary metabolites. They are usually obtained from leaves, flowers, fruits, seeds, buds, rhizomes, roots and/or bark [22,23], and are extracted by steam distillation and cold pressing processes [24]. EOs are defined as complex mixtures of approximately 20 to 60 components, with two or three of them found in high concentrations (20–70%) [25,26,27]. The chemical composition of their volatile fractions mainly includes mono- and sesquiterpenes, followed by several oxygenated derivatives, alcohols, aliphatic aldehydes and esters. On the other hand, non-volatile fractions comprise 1–10 wt% of EOs and are composed of carotenoids, fatty acids, flavonoids and waxes [26]. Generally, they are lipophilic, soluble in organic solvents and water-immiscible products [28].
Considering their wide chemical diversity, EOs have many properties and have been used since Ancient times as medicine. Among their activities, can be mentioned their antioxidant, anti-inflammatory, antibiotic, antiviral, antifungal, anti-parasitical, insecticidal, anticancer, wound healing, antihypertensive, analgesic and other clinical uses, such as a sedative, spasmolytic, analgesic, anesthetic or anxiolytic [29,30,31,32]. Furthermore, the global EO industry is valued at USD 18.6 billion and expected to reach USD 35.5 billion by 2028. This exponential growth can be attributed to increasing preference of people for a healthy lifestyle. The commercial use of EOs in aromatherapy, (mainly in spa and massage therapies) is their dominant application area, followed by the food and beverage industries [33]. Pharmaceutical fields also have a great interest in EOs’ properties, as they are rich sources of therapeutic compounds.
EOs are considered promising candidates for cancer treatments through their antioxidant, antimutagenic, antiangiogenic, antiproliferation effects; enhancement of the immune system; enzyme induction; and modulation of multidrug resistance mechanisms [34,35]. In PCa cells (Figure 1), EOs were demonstrated to act on PI3K/AKT signaling and the Ubiquitin-Proteasome (UPS) pathway, resulting in apoptosis. Moreover, EOs are able to induce mitochondrial stress, leading to changes in the expression of Bcl-2/Bax genes and membrane depolarization, resulting in the increased release of cytochrome-C to the cytoplasm and the induction of apoptosis. In addition, EOs are able to increase intracellular reactive oxygen species (ROS) levels, resulting in the apoptosis of PCa cells [35]. All of these signaling pathways may be targets for novel therapeutic drugs for PCa treatments.
In this context, the present work carried out a review of the anticancer effects of EOs from several plant species, while also focusing on their main bioactive compounds as effective treatments against PCa cells. Table 1 lists the successful in vitro studies of EOs extracted from several plants against different PCa cell lines. Table 2 displays the in vitro results of the major EOs compounds. Figure 2 shows the chemical structures of the bioactive compounds found in EOs that have been tested for anti-prostate cancer activity.

2. Methodology

This review follows the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) verification protocol [101]. The keywords used in the search for reports were “Prostate cancer”, “Essential oil”, “Basil”, “Chamomile”, “Citrus”, “Turmeric”, “Cymbopogon”, “Ginger”, “Jasmine”, “Lavender”, “Mentha”, “Myrtle”, “Oregano”, “Rose”, “Rosemary”, “Salvia” and “Thyme”. The search was performed in the Pubmed, ScienceDirect, WebOfScience and Scopus databases, focusing on the anti-prostate cancer activity of EOs and their bioactive compounds.
The works that were considered eligible have met the following criteria for inclusion: (1) retrospective experimental and clinical studies in human and mammal models and/or cell lines, (2) research investigating the anti-prostate cancer activity of EOs, (3) works containing quantitative and qualitative data, (4) without any language restrictions, (5) published between 2000 and 2023 and (6) manuscripts published in journals with impact indexes. On the other hand, the works that were not considered eligible met the following criteria for exclusion: (1) not meeting the objectives of this article, (2) university theses, (3) book chapters or books and (4) review articles.

3. EOs and Their Bioactive Compounds Tested in PCa Cell Lines

3.1. Chamomile

Chamomile is one of the most common and well-known herbs widely used in medicinal applications and is from the Asteraceae family. It has been traditionally used to treat several diseases due to its antioxidant, anti-inflammatory, antibacterial, antifungal, antiparasitic, insecticidal, antidiabetic and anti-tumoral activities. Chamomile EO is obtained from flowers by steam distillation [102,103]. German chamomile (Matricaria chamomilla) and Roman chamomile (Chamaemelum nobile, also called Anthemis nobilis) EOs mainly contain the terpenoid α-bisabolol, and its oxides A and B, and chamazulene [103,104].
In a previous report, the cytotoxicity of chamomile (Anthemis nobilis) EO was evaluated in the PC3 PCa cell line and it was found to generate a dose-dependent decrease in the survival of cells with an IC50 value of 0.071% (v/v). At a concentration of 0.2% (v/v) the EO exhibited strong cytotoxicity, showing cell viability lower than 4% [38]. A comparative study has evaluated the viability of PC3, DU145 and LNCaP human PCa cells exposed to aqueous and methanolic extracts of chamomile (Matricaria chamomilla). The aqueous extract treatment resulted in a time- and dose-dependent reduction in cell viability for all the cell lines studied, with IC50 values ranging from 2000 to 3000 µg/mL after 72 h of treatment. Similar results were found in all the cells lines after treatment with the methanolic extract, with IC50 values ranging from 100 to 200 µg/mL after 24 h treatments. In addition, the exposure to both chamomile extracts for 48 h led to apoptosis in all PCa cell lines with the formation of internucleosomal DNA fragments. Still, such treatments did not reduce the viability nor induce apoptosis in virally transformed normal human prostate epithelial PZ-HPV-7 cells [105].

3.2. Cinnamon

The Cinnamon genus belongs to the Lauraceae family and includes more than 250 species of evergreen trees. It is a common spice that possesses several biological properties such as antioxidant, anti-inflammatory, antibacterial, antifungal, anti-parasitic, antidiabetic and antitumor characteristics [106,107]. Cinnamon (Cinnamomum zeylanicum, also called Cinnamomum verum) EO is obtained by steam distillation, usually from the bark, and its main components are cinnamaldehyde and eugenol [108,109].
The viability of PC3 PCa cells treated with cinnamon EO was determined by an MTT essay. The results revealed a dose-dependent decrease in the survival of cells with an IC50 value of 0.012% (v/v) [38]. Another work demonstrated that aqueous cinnamon extract and procyanidin B2 (PCB2), a hydrophilic component, induced a time- and dose-dependent decrease in the viability of PC3 and LNCaP cells. Moreover, apoptosis occurred through the inhibition of 26S proteasome activity, the induction of a dose-dependent increase in caspase-3 activity, a decrease in Akt protein levels and a decrease in anti-apoptotic (Survivin and XIAP) and angiogenic (VEGF and VEGF receptor) gene markers’ expression in both cancer cells lines [110]. An HPLC analysis of this aqueous cinnamon extract revealed the presence of high amounts of cinnamaldehyde [86].
Other relevant compounds that have been isolated from cinnamon include eugenol and cinnamic acid. Cinnamaldehyde, eugenol and cinnamic acid compounds were able to inhibit proteasome activity in both PC3 and LNCaP cells, resulting in an increase in the expression of the chaperone protein (Hsp70), which negatively regulates the expression of FoxM1, an oncogenic transcription factor. In addition, cinnamon compounds downregulated the expression of angiogenic factors (VEGF and VEGFR), acting as antiangiogenic agents. The treatment of tumor cells with cinnamon compounds also led to a dose-dependent decrease in cell viability with IC50 values of 14.3 μM for cinnamaldehyde, 73.2 μM for eugenol and 6.2 μM for cinnamic acid [86]. Eugenol also inhibits the growth of DU145 cells [90]. Other authors reported that cinnamaldehyde inhibited the growth of PCa-associated fibroblasts (CAFs) in a dose-dependent manner, showing an IC50 value of 74.66 µM. Prostate CAFs play an important role in promoting carcinogenesis and the progression of prostate cancer. In addition to that, cinnamaldehyde induced apoptosis via cell cycle arrest at the G2/M phase, increased ROS generation and decreased Glutathione (GSH) levels and caspase activation on prostate CAFs [87]. It also relieved the immunosuppressive effects on prostate CAFs in a Toll-Like Receptor 4 (TLR4)-dependent manner [88].
β-Caryophyllene oxide, a bicyclic sesquiterpene isolated from several EOs including cinnamon EO, inhibited the growth of PC3 PCa cells in a time- and dose-dependent manner. The sesquiterpene has also induced apoptosis by inhibiting PI3K/AKT/mTOR/S6K1 signaling, which reduced mitochondrial membrane potential and cytochrome c release, activating caspase-3, the cleavage of PARP, ROS generation, the downregulation of anti-apoptotic proteins (Bcl-2, Bcl-xL, survivin, IAP-1 and IAP-2), cell cycle protein cyclin D1 and the upregulation of the tumor suppressor protein p53 and CDK inhibitor p21. In addition, β-Caryophyllene oxide downregulated proteins linked with metastasis and angiogenesis (COX-2 and VEGF) [78,79]. Another report showed that β-Caryophyllene oxide inhibited the cell growth, invasion and constitutive STAT3 activation in DU145 cells. The constitutive activation of STAT3 is often active and linked with proliferation, survival, invasion, metastasis and angiogenesis in tumor cells [80].

3.3. Citrus Species

The genus Citrus (Rutaceae family) is one of the most ancient and popular crops [111]. Citrus fruit are a significant source of vitamins such as ascorbic acid. The biological activities of citrus fruit include antioxidant, anti-inflammatory, antibacterial, antifungal, antitumor activities and others. The use of their EOs as natural preservatives in the food industry has been extensively studied [112]. Generally, the EOs of citrus fruit are obtained via a cold pressing method from different parts of the plants, such as fruit peel and leaves, and are mainly composed by limonene [109,112,113,114,115].
Mandarin (Citrus reticluata)-EO-treated PC3 PCa cells exhibited a dose-dependent decrease in viability with an IC50 value of 10.97 µg/mL. In addition, mandarin EO induced apoptotic DNA fragmentation, ROS generation and a decrease the expression of B-cell lymphoma 2 (Bcl2) and Murine Double Minute 2 (MDM2) genes, while it increased the expression of the p53 and Bax genes [47]. Lemon (Citrus limon) and grapefruit (Citrus paradisi) EO treatments in the PC3 cell line revealed a dose-dependent decrease in the survival of cells with IC50 values of 0.083% and 0.094% (v/v), respectively [38]. A navel orange (Citrus sinensis) EO treatment revealed a time- and dose-dependent inhibition of the proliferation of 22RV1 cells by inducing apoptosis. Its IC50 values at 24, 48 and 72 h were 45.74, 42.83, and 39.79 µg/mL, respectively [50]. Kumquat (Fortunella margarita) EO showed time- and dose-dependent antiproliferative activity against LNCaP cells by inducing apoptosis and the inhibition of inflammation. The treatment increased the Bax/Bcl2 ratio and induced the cleavage of caspase-3, indicating induction of apoptosis. Additionally, it decreased expression of inflammatory transcription factor NFκB and Cox-2, a downstream product of NFκB, indicating its inhibition of inflammation [42].
Limonene, a hydrocarbon monoterpene present in several citrus oils, inhibited the growth of DU145 cells in a dose-dependent manner with an IC50 value of 2.8 nM. In combination with docetaxel, limonene has enhanced cell growth inhibition. Moreover, this combined treatment induced ROS generation and enhanced apoptotic cell death by inducing cytoplasmic histone-associated DNA fragmentation; caspase-3 and caspase-9 cleavage; the upregulation of p21, p53 and Bad; and the downregulation of Bcl-xL and cleavage PARP protein. No effect on caspase-8 cleavage or on the expression of Bax and Bcl-2 were observed [94].

3.4. Turmeric

The genus Curcuma from the Zingiberaceae family is constituted by perennial rhizomatous herbs and includes about 100 species [116]. Turmeric (C. longa) has been widely explored as a coloring and flavoring agent, as well as in the pharmaceutical industry due to its antioxidant, anti-inflammatory, antimicrobial, antiviral, antitumor, antidiabetic, antiasthmatic, hypoglycemic, neuro- and dermoprotective and other properties. Its EO is obtained from its rhizome and is mainly composed of α and β-turmerone [116,117].
Turmeric EOs obtained from C. longa rhizomes collected from 20 different habitats were investigated for their cytotoxicity activity against LNCaP cells. It was revealed that they led to an inhibition of cell growth with IC50 values ranging from 16.41 to 124.27 µg/mL [55]. Turmeric-EO-treated PC3 prostate cancer cells revealed an inhibition of cell growth with an IC50 value of 97.94 µg/mL [52].
β-elemene reduced PC3 and DU145 cell growth in a time- and dose-dependent manner, with IC50 values of 105, 102 and 96 µg/mL at 24, 48 and 72 h; and 75, 70 and 66 µg/mL at 24, 48 and 72 h, respectively. Treatment with β-elemene also induced apoptosis in a time- and dose-dependent manner through cleaved caspase-3 and caspase-9, increased PARP and Bcl-2 downregulation [81].

3.5. Cymbopogon Species

The Cymbopogon genus belongs to the Andropoganeae family and includes more than 144 species of aromatic grass plants comprising lemon grass (C. citratus), tsauri grass (C. giganteus), citronella (C. nardus) and camel grass (C. schoenanthus). Cymbopogon species have several biological activities such as antioxidant, anti-inflammatory, antibacterial, antifungal, insecticidal and antitumor activities among others. Cymbopogon EOs are extensively used in the fragrance, cosmetics, food and flavor industries. They are obtained by the hydrodistillation of the species’ leaves and their main components are geraniol, citral and citronellal, depending on the species [118].
Treatments with lemon grass and tsauri grass EOs in PC3 and LNCaP cells showed that they act as antiproliferative agents. The IC50 values of lemon grass were 32.1 µg/mL for PC3 cells and 6.34 µg/mL for LNCaP. Tsauri grass exhibited values around 303.2 µg/mL for PC3 cells and 160.1 µg/mL for LNCaP [46]. Citronella EO also has antiproliferative activity in LNCaP cells (IC50 values of 58 µg/mL), caused by cell cycle arrest at the G2/M phase and changes in cell morphology [39]. Camel grass EO showed antiproliferative activity in LNCaP cells (IC50 values of 135.53 µg/mL), which was associated with its anti-migration property and cell cycle arrest in the G2/M phase [119].
Geraniol, an acyclic monoterpene, inhibited the cell growth of PC3 in the range of 0.25–1 mM. In the cells treated with geraniol, increased LDH and caspase-3 activity were observed, as were induced mitochondrial membrane depolarization and cell cycle arrest at the G1 phase. Additionally, the expressions of four cyclin isotypes (cyclin A, B, D, and E), two CDK family members (CDK1 and CDK4) and two anti-apoptotic Bcl-2 family members (Bcl-2 and Bcl-w) were reduced while the expressions of two CDK inhibitory proteins (p21 and p27) and two pro-apoptotic Bcl-2 family members (Bax and BNIP3) were noticeably elevated in cells treated with geraniol. In addition, the geraniol treatment, in a PC3 cell xenograft tumor in nude mice, efficiently suppressed tumor growth by inducing apoptosis and cell cycle arrest [91]. Geraniol-treated PC3 cells also experienced induced autophagy, inhibited AKT/mTOR signaling [92] and downregulated E2F8 transcription factor [93].
Citral, an acyclic monoterpene, when used as a treatment in PC3 and PC3M PCa cells, inhibited cell viability in a dose-dependent manner with IC50 values of 10 and 12.5 µg/mL. Citral treatment also reduced the cells’ clonogenic potential, induced morphological alterations and the expulsion of lipid droplets by the activation of AMPK protein expression and the subsequent downregulation of AMPK pathway genes such as SREBP1, ACC and HMGR. In addition, the Citral treatment induced apoptosis via DNA fragmentation, the upregulation of Bax and the downregulation of Bcl-2 expression [89].

3.6. Ginger

Ginger (Zingiber officinale), a member of the Zingiberaceae family, is a common spice used as a flavoring agent in beverages and food preparations. Ginger is also known to possess many therapeutic uses such as antioxidant, anti-inflammatory, anti-microbial and antitumor activities [120,121]. The EO of ginger is obtained from its rhizome and contains mainly zingiberene, curcumene and farnesene molecules, in its complex mixture [121].
A ginger EO treatment in PC3 cells exhibited a dose-dependent cytotoxicity, showing an IC50 value of 0.077% (v/v) [38]. Another work also showed the time- and dose-dependent antiproliferative activity of ginger EO in PC3 and LNCaP cell lines, with IC50 values of 0.42 mg/mL and 0.38 mg/mL, respectively [37]. Moreover, the ginger extract treatment was able to inhibit the proliferation of PC3 (IC50 of 250 µg/mL), LNCaP (IC50 of 75 µg/mL), DU145 (IC50 of 95 µg/mL), C4-2 (IC50 of 512 µg/mL) and C4-2B (IC50 of 240 µg/mL) PCa cells, while no effect was observed in normal prostate epithelial cells (PrEC). In addition, a treatment with ginger extract led to cell cycle arrest in PC3 cells by decreasing cyclin D1, cyclin E and CDK4 levels and increasing p21 and the CDK4 inhibitor. Additionally, a ginger extract induced apoptosis in PC3 cells by increasing Bax and decreasing Bcl2 expressions, releasing cytochrome c, increasing caspase-3 activity and cleaving PARP protein levels. In LNCaP cells, ginger extract also led to cell cycle arrest and increased caspase-3 activity. Mice treated with ginger extract had their tumor tissue suppressed and no detectable toxicity in their normal tissues was reported [122].

3.7. Lavender

The genus Lavandula (Lamiaceae family) comprises herbs popularly known for relieving stress, anxiety and depression. It also has other important therapeutic properties, such as antioxidant, anti-inflammatory, antibacterial and antitumor activities [33,44,45]. Lavender EO is obtained from flower heads and foliage by steam distillation and its main components are linalyl acetate and linalool molecules [123,124].
The cytotoxicity activity of lavender (L. stoechas) EO was evaluated in PC3 PCa cells. A dose-dependent decrease in the survival of cells was found, with an IC50 value of 0.05% (v/v) [38]. L. stoechas EO was also cytotoxic in LNCaP PCa cells [43]. PC3 cells were also treated with lavender (L. officialis) EO and showed a slight reduction in the viability of the cells [45]. A lavender (L. angustifolia) EO treatment in PC3 and DU145 cells revealed a time-dependent decrease in the viability of cells, with IC50 values of 0.037% and 0.199% (v/v), respectively. A treatment with the major components, linalyl acetate and linalool, also showed a potent cytotoxicity against both cell lines, with IC50 values of 4.98 µM and 11.74 µM in PC3 and 3.06 µM and 7.22 µM in DU145 cells, respectively. In addition, EO, linalyl acetate and linalool treatments inhibited migration and induced apoptosis via cell cycle arrest at the G2/M phase in PC3 and at the S phase in DU145 PCa cell lines. Lavender EO and its components have also inhibited the tumor growth of human PCa xenografts in mice [44]. Moreover, a linalool treatment inhibited the cell proliferation of 22Rvl PCa cells in a dose-dependent manner, showing an IC50 value of 3.38 mM. Linalool has also induced apoptosis, cell cycle arrest at the G0/G1 phase and a significant increase in expression of Bax, Bcl-2, p53, TRAIL receptors 1 (DR4), TRAIL receptors 2 (DR5) and cleaved caspases. Treatment with linalool suppressed significantly the tumor growth of human PCa xenografts in mice by inhibiting tumor cell proliferation and apoptosis [95].

3.8. Mentha Species

The genus Mentha belongs to the Lamiaceae family and possess more than 25 species including M. arvensis (wild mint), M. piperita (peppermint), M. longifolia (horsemint) and M. spicata (spearmint). The EOs and extracts from Mentha species have been used since ancient times for the treatment of several gastrointestinal system diseases. These EOs are obtained from the leaves and flowering aerial parts of these species and their main components are carvone, limonene, menthone and menthol [41,124,125].
The cytotoxicity of four Mentha species-based EOs has been assessed in LNCaP PCa cells. All of them showed strong cytotoxicity against the cancer cell line, with IC50 values of 55.7, 95.7, 52.0 and 90.0 µg/mL, respectively [41]. Another work has evaluated the cytotoxicity of spearmint EO in PC3 cells and it a dose-dependent decrease in the survival of cells was found, with an IC50 value of 0.088% (v/v) [38].
Menthol can evoke a cold sensation, mediated by cold-sensitive transient receptor potential melastatin 8 (TRPM8). This receptor was found to be expressed at high levels in several tumors, including PCa tissue and the PC3, LNCaP and DU145 culture cell lines [96]. Researchers have demonstrated a dose-dependent decrease in viability of PC3, LNCaP and DU145 cells treated with menthol [96,97,98,99]. Menthol also inhibits DU145 cell migration and induces cell cycle arrest at the G0/G1 phase [99]. On the other hand, the PC3 and LNCaP cell cycles were not affected by menthol treatments [97]. Although all three tested cell lines have expressed the TRPM8 receptor, menthol activity seems to not be mediated by the TRPM8 pathway [96,97].

3.9. Rosemary

Rosemary (Rosmarinus officinalis), a member of the Lamiaceae family, has been used as stimulant, analgesic and anti-inflammatory compound. Additionally, the pharmacological properties of rosemary include its antioxidant, antibacterial, antifungal, antiviral and antiproliferative activities. Its EO is mainly composed of eucalyptol, α-pinene and camphor [126].
A report has evaluated the cytotoxicity of rosemary EO in the LNCaP cell line and an IC50 value of 180.9 µg/mL has been found [53]. Rosemary extract treatments in LNCaP and 22Rv1 prostate cells inhibit cell growth (with IC50 values of 27 and 13.3 µg/mL, respectively), induced cell cycle arrest at the G1/G2 phase and G2 phase, respectively, and induced apoptosis by increasing the expression of Bax and cleaved caspase-3 [127]. Rosemary extract also inhibited cell growth, survival and migration, inducing apoptosis in PC3 PCa cells, while it had no significant effect on the proliferation of PNT1A normal PCa cells [128].
α-pinene, one of the most common terpenes of rosemary EO, possesses a strong inhibitory effect on the growth of PC3 and DU145 PCa cells, with IC50 values of 2.9 and 5.8 µM, respectively. In addition, α-pinene induced apoptosis and cell cycle arrest at the S phase in PC3 cells and at the G2/M phase in DU145. In xenograft tumors, α-pinene suppresses, significantly, their growth and induces apoptotic cells death [77].

3.10. Salvia Species

The Salvia genus (Lamiaceae family) comprises about 1000 species that are herbaceous, suffruticosus or shrubby perennial plants. Sage EO is commonly used as an analgesic, anti-inflammatory, anti-viral, or antitumor compound, in the treatment of cardiovascular and liver diseases, and in food and cosmetics industries, among other applications. Sage EO is obtained via a steam distillation process and its main components are linalyl acetate and linalool [129,130].
A group of researchers studied the effects of S. aurea, S. Judaica and S. viscosa EOs on DU145 PCs cells. Treatment with the three EOs in the range of 12.5–50 µg/mL induced a dose-dependent decrease in cell growth. However, only at 50 µg/mL was an increase in lactate dehydrogenase (LDH) observed, indicating the induction of necrosis, cell death, at this concentration. Furthermore, at 12.5 and 25 µg/mL, the EOs treatments caused an increase in DNA fragmentation, caspase activity and the Bax/Bcl-2 ratio and, at all three concentrations, it caused an increase in ROS generation and a decrease in GSH levels in a dose-dependent manner [54]. Although an S. officialis EO treatment in the range of 5–400 µg/mL in LNCaP cells has no significant effect, treatment with the sesquiterpene α-humulene showed high cytotoxicity with an IC50 value of 11.24 µg/mL [76].

3.11. Thyme

The Thymus genus of the Lamiaceae family contains about 400 species of perennial aromatic, evergreen or semi-evergreen herbaceous plants [131,132]. Thymus species have been used to treat several cardiorespiratory and gastrointestinal diseases due to their antioxidant, antibacterial, antifungal, antiviral and antispasmodic, among other, properties. Moreover, thyme EO is used in food and cosmetics as an antioxidant and preservative [133]. It is a product obtained from the flowering tops of Thymus vulgaris or Thymus zygis, or both species, by steam distillation. The main components found in thyme EO are thymol and carvacrol [131,134,135].
A previous work has evaluated the cytotoxicity of Thyme (T. vulgaris) EO in the PC3 cell line and found a dose-dependent decrease in the survival of cells with an IC50 value of 0.01% (v/v). At a concentration of 0.2% (v/v), the EOexhibited strong toxicity, showing a cell viability lower than 4%. Compared to the other nine EOs evaluated in this study, thyme EO showed the strongest cytotoxicity [38].
Studies have reported that thymol, a monoterpene phenol, has a cytotoxic effect on the viability of PC3 and DU145 PCa cells. Thymol-treated cells, after 24, 48 and 72 h, showed time- and dose-dependent decreases in cancer cell growth, exhibiting IC50 values of 711, 601 and 552 µM for PC3 and 799, 721 and 448 µM for DU145, respectively. Thymol has also induced apoptosis in both cell lines in a dose-dependent manner, but the mechanism of this action has not been elucidated yet. [100].
Carvacrol, a monoterpene phenol, was found to have antiproliferative and apoptotic action against human PCa DU145 cells in a time- and dose-dependent manner, showing IC50 values of 84.39 µM and 42.06 µM after 24 and 48 h of treatment, respectively. Carvacrol has also induced apoptosis via caspase-3 activation, an increase in ROS generation and cell cycle arrest at the G0/G1 phase [82]. In PC3 cells, carvacrol revealed similar effects, inhibiting proliferation, migration and invasion. Apoptosis acted in reducing cell viability with the induced activation of caspase-3, -8 and -9, induced high levels of ROS, disrupted the mitochondrial membrane potential, arrested the cell cycle at the G0/G1 phase, upregulated Bax and downregulated Bcl-2 expression. In addition, carvacrol decreased the expression of Notch1, Jagged-1, cyclin D1 and CDK4 and the increased expression of p21 [83].
Carvacrol also inhibits cell growth, migration and invasion in both PC3 and DU145 cell lines by decreasing the TRPM7-like current and reducing Matrix metalloproteinase-2 (MMP-2) protein expression and F-actin dynamics. In addition, both the PI3K/Akt and MEK/MAPK signaling pathways seems to be involved in the anticancer effects of carvacrol [84,85]. Trindade et al. successfully complexed carvacrol with β-cyclodextrin in order to enhance its solubility and anticancer activity. The resulting inclusion complex reduced the cell viability and migration of PC3 PCa cells in a dose-dependent manner [136].

3.12. Other Species

Jasmine (Jasminum grandiflorum)- and rose (Rosa centifolia)-EO-treated PC3 cells revealed a dose-dependent decrease in the survival of cells with IC50 values of 0.022% and 0.04% (v/v), respectively [38]. Oregano (Origanum vulgare) EO, which has a high content of carvacrol, cymene and linalool, was successfully encapsulated in a nanoemulsion and significantly inhibited the growth of PC3 PCa cells in a dose-dependent manner, with an IC50 value of 13.82 µg/mL. A treatment with an oregano-based nanoemulsion also induced apoptosis by causing cellular damage, DNA fragmentation, enhanced Bax expression, cytochrome c release, caspase-3 activation and decrease in Bcl2 expression [51]. Myrtle (Myrtus communis) EO induced a time- and dose-dependent decrease in the viability of PC3 and DU145 cells, while no effect was observed in normal PNT1A cells. In addition, myrtle EO showed antimigratory and proapoptotic properties [49].
Bayala et al. evaluated the cytotoxicity of several EOs towards PC3 and LNCaP cell lines. In addition to ginger, which has already been mentioned here, Ocimum basilicum, Lippia multiflora and Ageratum conyzoides EO treatments showed strong antiproliferative activity against both PCa cell lines. In PC3 cells, their IC50 values were 0.45, 0.30 and 0.49 mg/mL, respectively, and in LNCaP cells they were 0.46, 0.58 and 0.35 mg/mL, respectively. However, Ocimum americanum, Hyptis spicigera and Eucalyptus camaldulensis EOs have shown no antiproliferative effects [37]. Hyptis suaveolens-EO-treated LNCaP cells have showed a dose-dependent decrease in cell viability with an IC50 value of 163.01 µg/mL and cell cycle arrest at the G0/G1 phase [56].
Zataria Multiflora EO, which is mainly composed of carvacrol, terpinene, cymene and thymol, has inhibited the cell viability of PC3 PCa cells with an IC50 value of 26.3 µg/mL after 48 h of treatment. It also has induced apoptosis by increasing ROS generation, DNA fragmentation, cell cycle arrest at the G0/G1 phase, caspase activation, the upregulation of Bax and the downregulation of Bcl-2 expression. A combined treatment with this EO and doxorubicin improved the effects on PC3 cells in comparison to the pure drug [75].
Hedychium spicatum EO, mainly composed of β-pinene and eucalyptol, has inhibited the viability of PC3 cells in dose-dependent manner with an IC50 value of 21.88 µg/mL. It also induced apoptotic cell death, cell cycle arrest at the G2/M and S phases, intracellular ROS accumulation, mitochondria depolarization and increased caspase-3, -8, and -9 levels. In addition, an H. spicatum EO decreased Bcl-2 and Bcl-xL and increased Bax and Bak protein levels [66]. Other species, Hedychium genus, H. coccineum, H. gardnerianum, H. greenii and H. griffithianum, also showed antiproliferative activity against PC3 cells [67].
EOs obtained from the flowers and leaves of Artemisia arborescens have shown inhibitory growth activity against LNCaP and DU145 with IC50 values of 5.6–6.1 and 5.1–5.7 µg/mL, respectively. They have also induced DNA fragmentation and increased ROS levels [62].
An EO from Xylopia frutescens leaves, which is rich in E-caryophyllene and commonly known as embira, showed cytotoxic activity against PC-3M-metastatic PCa cells, with an IC50 value of 40 µg/mL [40]. An EO from the leaves of Guatteria pogonopus, which is mainly composed of γ-patchoulene, (E)-caryophyllene and β-pinene, showed similar results, with an IC50 value of 17.0 µg/mL [72].
Pinus mugo EO, which has a high content of β-caryophyllene, bornyl acetate and α-pinene, inhibited the viability of DU145 cells in time- and dose-dependent manner with an IC50 value less than 50 µg/mL. Their constitutive STAT3 activation signaling cascade was down-modulated, which decreased the expression of anti-proliferative as well as anti-apoptotic genes and proteins such as cyclin D1, Bcl-2, survivin, XIAP, Cox2 and IL-6. It also induced a quick decrease in GSH levels and an increase in ROS generation. In addition, apoptotic cell death was induced by caspase-3, and PARP cleavage induction and cell migration were inhibited in a dose-dependent manner [74].
Abies balsamea [36], Symphyopappus itatiayensis, Myrciaria floribundus, Talauma ovata, Psidium cattleyanum, Nectandra megapotamica [71], Amomum tsao-ko [59], Solanum erianthum, Solanum macranthum [48], Achillea wilhelmsii [57], Annona sylvatica [61], Hypericum hircinum [68], Anaxagorea brevipes [60], Guatteria elliptica [65], Bursera glabrifolia [63], Iryanthera polyneura [69], Aloysia polystachya [58], Euodia ruticarpa [64], Perralderia coronopifolia [73] and Liquidambar orientalis [70] EOs demonstrated antiproliferative activity in PC3 PCa cells. In contrast, Emani et al. evaluated a Nepeta cataria EO (15–500 µg/mL) treatment in PC3 and DU145 PCa cells and no significant effect was observed [137]. Clove (Syzygium aromaticum) EO, which has a high content of eugenol and β-caryophyllene, also showed no significant effect on the DU145 cell line in the range of 100–300 µL/mL [138]. The same was found for an Anemopsis californica EO treatment for PC3 cells [139].

4. Conclusions

PCa is the second most prevalent cancer in men worldwide and its treatment options remain limited. EOs are natural products that have been used in medicine since ancient times. Due to their wide chemical diversity, they possess several therapeutic properties, including anticancer activity.
Several EOs have been tested for their anti-prostate cancer property, such as chamomile, cinnamon, Citrus species, turmeric, Cymbopogon species, ginger, lavender, Mentha species, rosemary, Salvia species, thyme and other species, and shown to inhibit cell growth and migration, induce apoptosis, modulate the expression of apoptotic and anti-apoptotic genes and suppress angiogenesis. The major therapeutic compounds of these EOs have also been tested for anti-prostate cancer properties and showed similar results.
EOs and their constituents may be promising candidates for cancer treatments since they have potent therapeutic effects and are biocompatible, abundant and cheaper than current chemotherapeutics. In addition, novel anticancer compounds should be investigated in EOs’ molecular structures to contribute to the search for novel bioactives for innovative pharmaceutical preparations for anti-prostate cancer therapies.

5. Perspectives

There are many works that provide relevant results regarding EOs effects as a candidate treatment for several diseases, but clinical tests are still scarce. As observed, for PCa, there are very few in vivo tests and no clinical trials. The most challenging aspects of EOs, which limit their clinical use, are their highly lipophilic nature, physicochemical instability, photosensitivity and high volatility. The processing of EO-based products in the pharmaceutical field may be a good alternative to circumvent EOs limitations, resulting in the benefit of their therapeutic properties. New technologies for EO-based drug delivery system, such as nanoencapsulation, should be considered to ensure its high bioavailability and, consequently, its therapeutic effects. Different nanosystems could be developed, such as nanoemulsions, liposomes, lipid nanoparticles (SLN/NLC), polymer nanocapsulesand cyclodextrin. Although they are different nanocarriers, all of them have the ability to upload hydrophobic molecules with success. Therefore, they can offer physical protection to EO together with their nanometric particle sizes, which improve EOs solubility and prevent their degradation, hydrolysis and evaporation, allowing their safe and efficient administration via different routes.
However, more efforts are still necessary in order to develop stable, scaled-up, biocompatible and efficient EO-based nanosystems. Another challenge in the use of EOs in disease treatments is their composition variability, due to the difficulty of standardizing the composition of bioactive compounds whichcurrently limits their use to only as adjuvant therapy. Identifying the bioactive compound, its therapeutic effect and its chemical structure could open perspectives for novel research in this field. Chemical synthesis or biotechnological drug production strategies can provide accurate, safe and sustainable sources of these bioactive, allowing for their use in gold-standard therapies.
We hope that this review can stimulate further research in this field in order to provide promised PCa treatments in the close future.

Author Contributions

L.S.P.: Conceptualization and Writing—original draft. L.M.B.: Writing—review and editing. L.R.G.: Writing—review and editing. L.N.d.M.R.: Conceptualization and Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by INCT em Teranóstica e Nanobiotecnologia (INCT-TeraNano, CNPq Process n° 403193/2022-2 and FAPEMIG Process n° CBB—APQ-03613-17).

Acknowledgments

The authors thank Luiz Ricardo Goulart Filho (in memoriam), who passed away in 2021 due to complications from COVID-19. We are grateful and honored that we have worked with a brilliant and dedicated scientist who was passionate about his work.

Conflicts of Interest

The authors have declared no conflicts of interest.

References

  1. Vecchi, L.; Araújo, T.G.; Azevedo, F.V.P.d.V.; Mota, S.T.S.; Ávila, V.d.M.R.; Ribeiro, M.A.; Goulart, L.R. Phospholipase A2 Drives Tumorigenesis and Cancer Aggressiveness through Its Interaction with Annexin A1. Cells 2021, 10, 1472. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization Prostate Cancer Statistics. Available online: https://www.wcrf.org/cancer-trends/prostate-cancer-statistics (accessed on 21 July 2022).
  3. Rawla, P. Epidemiology of Prostate Cancer. World J. Oncol. 2019, 10, 63–89. [Google Scholar] [CrossRef] [PubMed]
  4. Kaffenberger, S.D.; Barbieri, C.E. Molecular Subtyping of Prostate Cancer. Curr. Opin. Urol. 2016, 26, 213–218. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, Y.A.; Sfakianos, J.; Tewari, A.K.; Cordon-cardo, C.; Kyprianou, N. Molecular Tracing of Prostate Cancer Lethality. Oncogene 2020, 39, 7225–7238. [Google Scholar] [CrossRef] [PubMed]
  6. Jernberg, E.; Bergh, A.; Wikström, P. Clinical Relevance of Androgen Receptor Alterations in Prostate Cancer. Endocr. Connect. 2017, 6, R146–R161. [Google Scholar] [CrossRef] [PubMed]
  7. Feng, Q.; He, B. Androgen Receptor Signaling in the Development of Castration-Resistant Prostate Cancer. Front. Oncol. 2019, 9, 858. [Google Scholar] [CrossRef] [PubMed]
  8. Wise, H.M.; Hermida, M.A.; Leslie, N.R. Prostate Cancer, PI3K, PTEN and Prognosis. Clin. Sci. 2017, 131, 197–210. [Google Scholar] [CrossRef] [PubMed]
  9. West, K.A.; Castillo, S.S.; Dennis, P.A. Activation of the PI3K/Akt Pathway and Chemotherapeutic Resistance. Drug Resist. Updates 2002, 6, 234–248. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, F.Z.; Zhao, X.K. Ubiquitin-Proteasome Pathway and Prostate Cancer. Onkologie 2013, 36, 592–596. [Google Scholar] [CrossRef]
  11. Risdon, E.N.; Chau, C.H.; Price, D.K.; Sartor, O.; Figg, W.D. PARP Inhibitors and Prostate Cancer: To Infinity and Beyond BRCA. Oncologist 2021, 26, e115–e129. [Google Scholar] [CrossRef]
  12. Teyssonneau, D.; Margot, H.; Cabart, M.; Anonnay, M.; Sargos, P.; Vuong, N.S.; Soubeyran, I.; Sevenet, N.; Roubaud, G. Prostate Cancer and PARP Inhibitors: Progress and Challenges. J. Hematol. Oncol. 2021, 14, 51. [Google Scholar] [CrossRef] [PubMed]
  13. Machado, T.Q.; da Fonseca, A.C.C.; Duarte, A.B.S.; Robbs, B.K.; de Sousa, D.P. A Narrative Review of the Antitumor Activity of Monoterpenes from Essential Oils: An Update. BioMed Res. Int. 2022, 2022, 6317201. [Google Scholar] [CrossRef]
  14. Namekawa, T.; Ikeda, K.; Horie-Inoue, K.; Inoue, S. Application of Prostate Cancer Models for Preclinical Study: Advantages and Limitations of Cell Lines, Patient-Derived Xenografts, and Three-Dimensional Culture of Patient-Derived Cells. Cells 2019, 8, 74. [Google Scholar] [CrossRef] [PubMed]
  15. Mickey, D.D.; Stone, K.R.; Wunderli, H.; Mickey, G.H.; Vollmer, R.T.; Paulson, D.F. Heterotransplantation of a Human Prostatic Adenocarcinoma Cell Line in Nude Mice. Cancer Res. 1977, 37, 4049–4058. [Google Scholar]
  16. Kaighn, M.E.; Narayan, K.S.; Ohnuki, Y.; Lechner, J.F.; Jones, L.W. Establishment and Characterization of a Human Prostatic Carcinoma Cell Line (PC-3). Investig. Urol. 1979, 17, 16–23. [Google Scholar]
  17. Kozlowski, J.M.; Fidler, I.J.; Campbell, D.; Xu, Z.L.; Kaighn, M.E.; Hart, I.R. Metastatic Behavior of Human Tumor Cell Lines Grown in the Nude Mouse. Cancer Res. 1984, 44, 3522–3529. [Google Scholar] [PubMed]
  18. Horoszewicz, J.; Leong, S.; Chu, T.; Wajsman, Z.; Friedman, M.; Papsidero, L.; Kim, U.; Chai, L.; Kakati, S.; Arya, S.; et al. The LNCaP Cell Line—A New Model for Studies on Human Prostatic Carcinoma. Prog. Clin. Biol. Res. 1980, 37, 115–132. [Google Scholar]
  19. Wu, H.C.; Hsieh, J.T.; Gleave, M.E.; Brown, N.M.; Pathak, S.; Chung, L.W. Derivation of Androgen-Independent Human LNCaP Prostatic Cancer Cell Sublines: Role of Bone Stromal Cells. Int. J. Cancer 1994, 57, 406–412. [Google Scholar] [CrossRef]
  20. Thalmann, G.N.; Anezinis, P.E.; Chang, S.M.; Zhau, H.E.; Kim, E.E.; Hopwood, V.L.; Pathak, S.; von Eschenbach, A.C.; Chung, L.W. Androgen-Independent Cancer Progression and Bone Metastasis in the LNCaP Model of Human Prostate Cancer. Cancer Res. 1994, 54, 2577–2581. [Google Scholar]
  21. Sramkoski, R.M.; Pretlow, T.G.; Giaconia, J.M.; Pretlow, T.P.; Schwartz, S.; Sy, M.S.; Marengo, S.R.; Rhim, J.S.; Zhang, D.; Jacobberger, J.W. A New Human Prostate Carcinoma Cell Line, 22Rv1. In Vitro Cell Dev. Biol. Anim. 1999, 35, 403–409. [Google Scholar] [CrossRef]
  22. Cascaes, M.M.; Carneiro, O.D.S.; do Nascimento, L.D.; de Moraes, Â.A.B.; de Oliveira, M.S.; Cruz, J.N.; Guilhon, G.M.S.P.; de Aguiar Andrade, E.H. Essential Oils from Annonaceae Species from Brazil: A Systematic Review of Their Phytochemistry, and Biological Activities. Int. J. Mol. Sci. 2021, 22, 12140. [Google Scholar] [CrossRef] [PubMed]
  23. Ferreira, O.O.; Cruz, J.N.; de Moraes, Â.A.B.; de Jesus Pereira Franco, C.; Lima, R.R.; dos Anjos, T.O.; Siqueira, G.M.; do Nascimento, L.D.; Cascaes, M.M.; de Oliveira, M.S.; et al. Essential Oil of the Plants Growing in the Brazilian Amazon: Chemical Composition, Antioxidants, and Biological Applications. Molecules 2022, 27, 4373. [Google Scholar] [CrossRef] [PubMed]
  24. De Groot, A.; Schmidt, E. Part II: General Aspects. Dermatitis 2016, 27, 43–49. [Google Scholar] [CrossRef] [PubMed]
  25. Dehsheikh, A.B.; Sourestani, M.M.; Dehsheikh, P.B.; Mottaghipisheh, J.; Vitalini, S.; Iriti, M. Monoterpenes: Essential Oil Components with Valuable Features. Mini-Rev. Med. Chem. 2020, 20, 958–974. [Google Scholar] [CrossRef]
  26. Aziz, Z.A.A.; Ahmad, A.; Setapar, S.H.M.; Karakucuk, A.; Azim, M.M.; Lokhat, D.; Rafatullah, M.; Ganash, M.; Kamal, M.A.; Ashraf, G.M. Essential Oils: Extraction Techniques, Pharmaceutical and Therapeutic Potential—A Review. Curr. Drug Metab. 2018, 19, 1100–1110. [Google Scholar] [CrossRef] [PubMed]
  27. Bakkali, F.; Averbeck, S.; Averbeck, D.; Idaomar, M. Biological Effects of Essential Oils—A Review. Food Chem. Toxicol. 2008, 46, 446–475. [Google Scholar] [CrossRef]
  28. El Asbahani, A.; Miladi, K.; Badri, W.; Sala, M.; Addi, E.H.A.; Casabianca, H.; El Mousadik, A.; Hartmann, D.; Jilale, A.; Renaud, F.N.R.; et al. Essential Oils: From Extraction to Encapsulation. Int. J. Pharm. 2015, 483, 220–243. [Google Scholar] [CrossRef] [PubMed]
  29. Ishfaq, P.M.; Shukla, A.; Beraiya, S.; Tripathi, S.; Mishra, S.K. Biochemical and Pharmacological Applications of Essential Oils in Human Health Especially in Cancer Prevention. Anticancer Agents Med. Chem. 2018, 18, 1815–1827. [Google Scholar] [CrossRef] [PubMed]
  30. Ramsey, J.T.; Shropshire, B.C.; Nagy, T.R.; Chambers, K.D.; Li, Y.; Korach, K.S.; Korach, S. Essential Oils and Health. Yale J. Biol. Med. 2020, 93, 291–305. [Google Scholar]
  31. Niculescu, A.G.; Grumezescu, A.M. Natural Compounds for Preventing Ear, Nose, and Throat-related Oral Infections. Plants 2021, 10, 1847. [Google Scholar] [CrossRef]
  32. Motelica, L.; Vasile, B.S.; Ficai, A.; Surdu, A.V.; Ficai, D.; Oprea, O.C.; Andronescu, E.; Mustățea, G.; Ungureanu, E.L.; Dobre, A.A. Antibacterial Activity of Zinc Oxide Nanoparticles Loaded with Essential Oils. Pharmaceutics 2023, 15, 2470. [Google Scholar] [CrossRef] [PubMed]
  33. Ridouh, I.; Hackshaw, K.V. Essential Oils and Neuropathic Pain. Plants 2022, 11, 1797. [Google Scholar] [CrossRef]
  34. Bhalla, Y.; Gupta, V.K.; Jaitak, V. Anticancer Activity of Essential Oils: A Review. J. Sci. Food Agric. 2013, 93, 3643–3653. [Google Scholar] [CrossRef] [PubMed]
  35. Gautam, N.; Mantha, A.K.; Mittal, S. Essential Oils and Their Constituents as Anticancer Agents: A Mechanistic View. Biomed. Res. Int. 2014, 2014, 154106. [Google Scholar] [CrossRef] [PubMed]
  36. Legault, J.; Dahl, W.; Debiton, E.; Pichette, A.; Madelmont, J.-C. Antitumor Activity of Balsam Fir Oil: Production of Reactive Oxygen Species Induced by Alpha-Humulene as Possible Mechanism of Action. Planta Med. 2003, 69, 402–407. [Google Scholar] [CrossRef] [PubMed]
  37. Bayala, B.; Bassole, I.H.N.; Gnoula, C.; Nebie, R.; Yonli, A.; Morel, L.; Figueredo, G.; Nikiema, J.B.; Lobaccaro, J.M.A.; Simpore, J. Chemical Composition, Antioxidant, Anti-Inflammatory and Anti-Proliferative Activities of Essential Oils of Plants from Burkina Faso. PLoS ONE 2014, 9, e92122. [Google Scholar] [CrossRef] [PubMed]
  38. Zu, Y.; Yu, H.; Liang, L.; Fu, Y.; Efferth, T.; Liu, X.; Wu, N. Activities of Ten Essential Oils towards Propionibacterium Acnes and PC-3, A-549 and MCF-7 Cancer Cells. Molecules 2010, 15, 3200–3210. [Google Scholar] [CrossRef] [PubMed]
  39. Bayala, B.; Coulibaly, A.Y.; Djigma, F.W.; Nagalo, B.M.; Baron, S.; Figueredo, G.; Lobaccaro, J.M.A.; Simpore, J. Chemical Composition, Antioxidant, Anti-Inflammatory and Antiproliferative Activities of the Essential Oil of Cymbopogon Nardus, a Plant Used in Traditional Medicine. Biomol. Concepts 2020, 11, 86–96. [Google Scholar] [CrossRef] [PubMed]
  40. Ferraz, R.P.C.; Cardoso, G.M.B.; Da Silva, T.B.; Fontes, J.E.D.N.; Prata, A.P.D.N.; Carvalho, A.A.; Moraes, M.O.; Pessoa, C.; Costa, E.V.; Bezerra, D.P. Antitumour Properties of the Leaf Essential Oil of Xylopia frutescens Aubl. (Annonaceae). Food Chem. 2013, 141, 542–547. [Google Scholar] [CrossRef]
  41. Hussain, A.I.; Anwar, F.; Nigam, P.S.; Ashraf, M.; Gilani, A.H. Seasonal Variation in Content, Chemical Composition and Antimicrobial and Cytotoxic Activities of Essential Oils from Four Mentha Species. J. Sci. Food Agric. 2010, 90, 1827–1836. [Google Scholar] [CrossRef]
  42. Jayaprakasha, G.; Murthy, K.C.; Demarais, R.; Patil, B. Inhibition of Prostate Cancer (LNCaP) Cell Proliferation by Volatile Components from Nagami Kumquats. Planta Med. 2012, 78, 974–980. [Google Scholar] [CrossRef] [PubMed]
  43. Gören, A.C.; Topçu, G.; Bilsel, G.; Bilsel, M.; Aydoǧmuş, Z.; Pezzuto, J.M. The Chemical Constituents and Biological Activity of Essential Oil of Lavandula Stoechas ssp. Stoechas. Z. Naturforschung—Sect. C J. Biosci. 2002, 57, 797–800. [Google Scholar] [CrossRef] [PubMed]
  44. Zhao, Y.; Chen, R.; Wang, Y.; Qing, C.; Wang, W.; Yang, Y. In Vitro and in Vivo Efficacy Studies of Lavender Angustifolia Essential Oil and Its Active Constituents on the Proliferation of Human Prostate Cancer. Integr. Cancer Ther. 2017, 16, 215–226. [Google Scholar] [CrossRef]
  45. Fahmy, M.A.; Farghaly, A.A.; Hassan, E.E.; Hassan, E.M.; Hassan, Z.M.; Mahmoud, K.; Omara, E.A. Evaluation of the Anti-Cancer/Anti-Mutagenic Efficiency of Lavandula Officinalis Essential Oil. Asian Pac. J. Cancer Prev. 2022, 23, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
  46. Bayala, B.; Bassole, I.H.N.; Maqdasy, S.; Baron, S.; Simpore, J.; Lobaccaro, J.M.A. Cymbopogon Citratus and Cymbopogon Giganteus Essential Oils Have Cytotoxic Effects on Tumor Cell Cultures. Identification of Citral as a New Putative Anti-Proliferative Molecule. Biochimie 2018, 153, 162–170. [Google Scholar] [CrossRef] [PubMed]
  47. Hussien, N.A.; Mohamed, H.R.H. Estimation of Mandarin Peel Oil–Induced Cytotoxicity and Genotoxicity in Human Normal Fibroblast and Cancerous Prostate Cell Lines. Toxicol. Mech. Methods 2021, 31, 100–106. [Google Scholar] [CrossRef] [PubMed]
  48. Essien, E.E.; Ogunwande, I.A.; Setzer, W.N.; Ekundayo, O. Chemical Composition, Antimicrobial, and Cytotoxicity Studies on S. Erianthum and S. Macranthum Essential Oils. Pharm. Biol. 2012, 50, 474–480. [Google Scholar] [CrossRef] [PubMed]
  49. Giuliani, C.; Moretti, R.M.; Bottoni, M.; Santagostini, L.; Fico, G.; Montagnani Marelli, M. The Leaf Essential Oil of Myrtus communis subsp. tarentina (L.) Nyman: From Phytochemical Characterization to Cytotoxic and Antimigratory Activity in Human Prostate Cancer Cells. Plants 2023, 12, 1293. [Google Scholar] [CrossRef]
  50. Yang, C.; Chen, H.; Chen, H.; Zhong, B.; Luo, X.; Chun, J. Antioxidant and Anticancer Activities of Essential Oil from Gannan Navel Orange Peel. Molecules 2017, 22, 1391. [Google Scholar] [CrossRef]
  51. Perumalsamy, H.; Shanmugam, R.; Kim, J.-R.; Anandapadmanaban, G.; Huq, M.A.; Dua, K.; Chellappan, D.K.; Yoon, T.H.; Balusamy, S.R. Nanoemulsion and Encapsulation Strategy of Hydrophobic Oregano Essential Oil Increased Human Prostate Cancer Cell Death via Apoptosis by Attenuating Lipid Metabolism. Bioinorg. Chem. Appl. 2022, 2022, 9569226. [Google Scholar] [CrossRef]
  52. Essien, E.; Newby, J.; Walker, T.; Setzer, W.; Ekundayo, O. Chemotaxonomic Characterization and In-Vitro Antimicrobial and Cytotoxic Activities of the Leaf Essential Oil of Curcuma longa Grown in Southern Nigeria. Medicines 2015, 2, 340–349. [Google Scholar] [CrossRef]
  53. Hussain, A.I.; Anwar, F.; Chatha, S.A.S.; Jabbar, A.; Mahboob, S.; Nigam, P.S. Rosmarinus officinalis Essential Oil: Antiproliferative, Antioxidant and Antibacterial Activities. Braz. J. Microbiol. 2010, 41, 1070–1078. [Google Scholar] [CrossRef] [PubMed]
  54. Russo, A.; Cardile, V.; Graziano, A.C.E.; Avola, R.; Bruno, M.; Rigano, D. Involvement of Bax and Bcl-2 in Induction of Apoptosis by Essential Oils of Three Lebanese Salvia Species in Human Prostate Cancer Cells. Int. J. Mol. Sci. 2018, 19, 292. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, L.; Yang, Z.; Chen, F.; Su, P.; Chen, D.; Pan, W.; Fang, Y.; Dong, C.; Zheng, X.; Du, Z. Composition and Bioactivity Assessment of Essential Oils of Curcuma longa L. Collected in China. Ind. Crops Prod. 2017, 109, 60–73. [Google Scholar] [CrossRef]
  56. Bayala, B.; Nadembega, C.; Guenné, S.; Buñay, J.; Zohoncon, T.M.; Djigma, F.W.; Yonli, A.; Baron, S.; Figueredo, G.; Lobaccaro, J.M.A.; et al. Chemical Composition, Antioxidant and Cytotoxic Activities of Hyptis suaveolens (L.) Poit. Essential Oil on Prostate and Cervical Cancers Cells. Pak. J. Biol. Sci. 2020, 23, 1184–1192. [Google Scholar] [CrossRef]
  57. Ahmadi-jouibari, T.; Nikbakhtb, M.R.; Mansouric, K.; Majnooni, M.B. Cytotoxic Effects of the Essential Oil from Achillea Wilhelmsii C. Koch. J. Rep. Pharm. Sci. 2013, 2, 98–102. [Google Scholar]
  58. Moller, A.C.; Parra, C.; Said, B.; Werner, E.; Flores, S.; Villena, J.; Russo, A.; Caro, N.; Montenegro, I.; Madrid, A. Antioxidant and Anti-Proliferative Activity of Essential Oil and Main Components from Leaves of Aloysia Polystachya Harvested in Central Chile. Molecules 2021, 26, 131. [Google Scholar] [CrossRef]
  59. Yang, Y.; Yue, Y.; Runwei, Y.; Guolin, Z. Cytotoxic, Apoptotic and Antioxidant Activity of the Essential Oil of Amomum Tsao-Ko. Bioresour. Technol. 2010, 101, 4205–4211. [Google Scholar] [CrossRef]
  60. De Alencar, D.C.; Pinheiro, M.L.B.; Pereira, J.L.D.S.; De Carvalho, J.E.; Campos, F.R.; Serain, A.F.; Tirico, R.B.; Hernández-Tasco, A.J.; Costa, E.V.; Salvador, M.J. Chemical Composition of the Essential Oil from the Leaves of Anaxagorea Brevipes (Annonaceae) and Evaluation of Its Bioactivity. Nat. Prod. Res. 2016, 30, 1088–1092. [Google Scholar] [CrossRef]
  61. Formagio, A.S.N.; Vieira, M.D.C.; Dos Santos, L.A.C.; Cardoso, C.A.L.; Foglio, M.A.; De Carvalho, J.E.; Andrade-Silva, M.; Kassuya, C.A.L. 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]
  62. Russo, A.; Graziano, A.; Bruno, M.; Cardile, V.; Rigano, D. Apoptosis Induction of Essential Oils from Artemisia arborescens L. in Human Prostate Cancer Cells. J. Ethnopharmacol. 2023, 303, 115929. [Google Scholar] [CrossRef] [PubMed]
  63. Villa-Ruano, N.; Becerra-Martínez, E.; Cruz-Durán, R.; Zarate-Reyes, J.A.; Landeta-Cortés, G.; Romero-Arenas, O. Volatile Profiling, Insecticidal, Antibacterial and Antiproliferative Properties of the Essential Oils of Bursera Glabrifolia Leaves. Chem. Biodivers. 2018, 15, e1800354. [Google Scholar] [CrossRef] [PubMed]
  64. Yeh, T.H.; Lin, J.Y. Active Ingredients from Euodia Ruticarpa Steam Distilled Essential Oil Inhibit Pc-3 Prostate Cancer Cell Growth via Direct Action and Indirect Immune Cells Conditioned Media in Vitro. Curr. Issues Mol. Biol. 2021, 43, 996–1018. [Google Scholar] [CrossRef] [PubMed]
  65. Rajca Ferreira, A.K.; Lourenço, F.R.; Young, M.C.M.; Lima, M.E.L.; Cordeiro, I.; Suffredini, I.B.; Lopes, P.S.; Moreno, P.R.H. Chemical Composition and Biological Activities of Guatteria elliptica R. E. Fries (Annonaceae) Essential Oils. J. Essent. Oil Res. 2018, 30, 69–76. [Google Scholar] [CrossRef]
  66. Ray, A.; Gadnayak, A.; Jena, S.; Sahoo, A.; Patnaik, J.; Chandra Panda, P.; Nayak, S. Hedychium Spicatum Rhizome Essential Oil Induces Apoptosis in Human Prostate Adenocarcinoma PC-3 Cells via Mitochondrial Stress and Caspase Activation. Heliyon 2023, 9, E13807. [Google Scholar] [CrossRef] [PubMed]
  67. Mohanty, S.; Ray, A.; Sahoo, C.; Sahoo, A.; Jena, S.; Panda, P.C.; Nayak, S. Volatile Profiling Coupled with Multivariate Analysis, Antiproliferative and Anti-Inflammatory Activities of Rhizome Essential Oil of Four Hedychium Species from India. J. Ethnopharmacol. 2023, 317, 116835. [Google Scholar] [CrossRef] [PubMed]
  68. Quassinti, L.; Lupidi, G.; Maggi, F.; Sagratini, G.; Papa, F.; Vittori, S.; Bianco, A.; Bramucci, M. Antioxidant and Antiproliferative Activity of Hypericum hircinum L. Subsp. Majus (Aiton) N. Robson Essential Oil. Nat. Prod. Res. 2013, 27, 862–868. [Google Scholar] [CrossRef] [PubMed]
  69. Martins, E.R.; Díaz, I.E.C.; Paciencia, M.L.B.; Fana, S.A.; Morais, D.; Eberlin, M.N.; Silva, J.S.; Silveira, E.R.; Barros, M.P.; Suffredini, I.B. Interference of Seasonal Variation on the Antimicrobial and Cytotoxic Activities of the Essential Oils from the Leaves of Iryanthera polyneura in the Amazon Rain Forest. Chem. Biodivers. 2019, 16, e1900374. [Google Scholar] [CrossRef]
  70. Cengiz Baloglu, M.; Yildiz Ozer, L.; Pirci, B.; Zengin, G.; Ibrahim Uba, A.; Celik Altunoglu, Y. Evaluation of the Potential Therapeutic Properties of Liquidambar orientalis Oil. Chem. Biodivers. 2023, 20, e202300291. [Google Scholar] [CrossRef]
  71. Apel, M.A.; Lima, M.E.L.; Souza, A.; Cordeiro, I.; Young, M.C.M.; Sobral, M.E.G.; Suffredini, I.B.; Moreno, P.R.H. Screening of the Biological Activity from Essential Oils of Native Species from the Atlantic Rain Forest (São Paulo-Brazil). Pharmacologyonline 2006, 3, 376–383. [Google Scholar]
  72. Fontes, J.E.d.N.; Ferraz, R.P.C.; Britto, A.C.S.; Carvalho, A.A.; Moraes, M.O.; Pessoa, C.; Costa, E.V.; Bezerra, D.P. Antitumor Effect of the Essential Oil from Leaves of Guatteria pogonopus (Annonaceae). Chem. Biodivers. 2013, 10, 722–729. [Google Scholar] [CrossRef] [PubMed]
  73. Boussaha, S.; Lassed, S.; Abdelwahab, A.B.; Krid, A.; Altun, M.; Chalard, P.P.; Chalchat, P.J.C.; Figueredo, G.; Zama, P.D.; Demirtas, P.I.; et al. Chemical Characterization, DNA-Damage Protection, Antiproliferative Activity and in Silico Studies of the Essential Oils from Perralderia coronopifolia Coss. Chem. Biodivers. 2024, 21, e202301535. [Google Scholar] [CrossRef] [PubMed]
  74. Thalappil, M.A.; Butturini, E.; Carcereri de Prati, A.; Bettin, I.; Antonini, L.; Sapienza, F.U.; Garzoli, S.; Ragno, R.; Mariotto, S. Pinus Mugo Essential Oil Impairs STAT3 Activation through Oxidative Stress and Induces Apoptosis in Prostate Cancer Cells. Molecules 2022, 27, 4834. [Google Scholar] [CrossRef] [PubMed]
  75. Zare, E.; Jamali, T.; Ardestani, S.K.; Kavoosi, G. Synergistic Effect of Zataria Multiflora Essential Oil on Doxorubicin-Induced Growth Inhibition of PC3 Cancer Cells and Apoptosis. Complement. Ther. Clin. Pract. 2021, 42, 101286. [Google Scholar] [CrossRef] [PubMed]
  76. Loizzo, M.R.; Tundis, R.; Menichini, F.; Saab, A.M.; Statti, G.A.; Menichini, F. Cytotoxic Activity of Essential Oils from Labiatae and Lauraceae Families Against In Vitro Human Tumor Models. Anticancer Res. 2007, 27, 3293–3300. [Google Scholar] [PubMed]
  77. Zhao, Y.; Chen, R.; Wang, Y.; Yang, Y. α-Pinene Inhibits Human Prostate Cancer Growth in a Mouse Xenograft Model. Chemotherapy 2018, 63, 1–7. [Google Scholar] [CrossRef]
  78. Park, K.R.; Nam, D.; Yun, H.M.; Lee, S.G.; Jang, H.J.; Sethi, G.; Cho, S.K.; Ahn, K.S. β-Caryophyllene Oxide Inhibits Growth and Induces Apoptosis through the Suppression of PI3K/AKT/MTOR/S6K1 Pathways and ROS-Mediated MAPKs Activation. Cancer Lett. 2011, 312, 178–188. [Google Scholar] [CrossRef]
  79. Delgado, C.; Mendez-Callejas, G.; Celis, C. Caryophyllene Oxide, the Active Compound Isolated from Leaves of Hymenaea Courbaril l. (Fabaceae) with Antiproliferative and Apoptotic Effects on Pc-3 Androgen-Independent Prostate Cancer Cell Line. Molecules 2021, 26, 6142. [Google Scholar] [CrossRef]
  80. Kim, C.; Cho, S.K.; Kapoor, S.; Kumar, A.; Vali, S.; Abbasi, T.; Kim, S.H.; Sethi, G.; Ahn, K.S. β-Caryophyllene Oxide Inhibits Constitutive and Inducible STAT3 Signaling Pathway through Induction of the SHP-1 Protein Tyrosine Phosphatase. Mol. Carcinog. 2014, 53, 793–806. [Google Scholar] [CrossRef]
  81. Li, Q.Q.; Wang, G.; Huang, F.; Banda, M.; Reed, E. Antineoplastic Effect of β-Elemene on Prostate Cancer Cells and Other Types of Solid Tumour Cells. J. Pharm. Pharmacol. 2010, 62, 1018–1027. [Google Scholar] [CrossRef]
  82. Khan, F.; Khan, I.; Farooqui, A.; Ansari, I.A. Carvacrol Induces Reactive Oxygen Species (ROS)-Mediated Apoptosis Along with Cell Cycle Arrest at G0/G1 in Human Prostate Cancer Cells. Nutr. Cancer 2017, 69, 1075–1087. [Google Scholar] [CrossRef]
  83. Khan, F.; Singh, V.K.; Saeed, M.; Kausar, M.A.; Ansari, I.A. Carvacrol Induced Program Cell Death and Cell Cycle Arrest in Androgen-Independent Human Prostate Cancer Cells via Inhibition of Notch Signaling. Anticancer Agents Med. Chem. 2019, 19, 1588–1608. [Google Scholar] [CrossRef] [PubMed]
  84. Luo, Y.; Wu, J.Y.; Lu, M.H.; Shi, Z.; Na, N.; Di, J.M. Carvacrol Alleviates Prostate Cancer Cell Proliferation, Migration, and Invasion through Regulation of PI3K/Akt and MAPK Signaling Pathways. Oxid. Med. Cell. Longev. 2016, 2016, 1469693. [Google Scholar] [CrossRef] [PubMed]
  85. Heidarian, E.; Keloushadi, M. Antiproliferative and Anti-Invasion Effects of Carvacrol on PC3 Human Prostate Cancer Cells through Reducing PSTAT3, PAKT, and PERK1/2 Signaling Proteins. Int. J. Prev. Med. 2019, 10, 156. [Google Scholar] [CrossRef] [PubMed]
  86. Gopalakrishnan, S.; Ismail, A. Aromatic Monophenols from Cinnamon Bark Act as Proteasome Inhibitors by Upregulating ER Stress, Suppressing FoxM1 Expression, and Inducing Apoptosis in Prostate Cancer Cells. Phytother. Res. 2021, 35, 5781–5794. [Google Scholar] [CrossRef] [PubMed]
  87. Han, L.; Mei, J.; Ma, J.; Wang, F.; Gu, Z.; Li, J.; Zhang, Z.; Zeng, Y.; Lou, X.; Yao, X.; et al. Cinnamaldehyde Induces Endogenous Apoptosis of the Prostate Cancer-Associated Fibroblasts via Interfering the Glutathione-Associated Mitochondria Function. Med. Oncol. 2020, 37, 91. [Google Scholar] [CrossRef] [PubMed]
  88. Mei, J.; Ma, J.; Xu, Y.; Wang, Y.; Hu, M.; Ma, F.; Qin, Z.; Xue, R.; Tao, N. Cinnamaldehyde Treatment of Prostate Cancer-Associated Fibroblasts Prevents Their Inhibitory Effect on T Cells through Toll-like Receptor 4. Drug Des. Dev. Ther. 2020, 14, 3363–3372. [Google Scholar] [CrossRef] [PubMed]
  89. Balusamy, S.R.; Perumalsamy, H.; Veerappan, K.; Huq, M.A.; Rajeshkumar, S.; Lakshmi, T.; Kim, Y.J. Citral Induced Apoptosis through Modulation of Key Genes Involved in Fatty Acid Biosynthesis in Human Prostate Cancer Cells: In Silico and in Vitro Study. BioMed Res. Int. 2020, 2020, 6040727. [Google Scholar] [CrossRef] [PubMed]
  90. Carrasco, H.; Espinoza, L.; Cardile, V.; Gallardo, C.; Cardona, W.; Lombardo, L.; Catalán, K.; Cuellar, M.; Russo, A. Eugenol and Its Synthetic Analogues Inhibit Cell Growth of Human Cancer Cells (Part I). J. Braz. Chem. Soc. 2008, 19, 543–548. [Google Scholar] [CrossRef]
  91. Kim, S.H.; Bae, H.C.; Park, E.J.; Lee, C.R.; Kim, B.J.; Lee, S.; Park, H.H.; Kim, S.J.; So, I.; Kim, T.W.; et al. Geraniol Inhibits Prostate Cancer Growth by Targeting Cell Cycle and Apoptosis Pathways. Biochem. Biophys. Res. Commun. 2011, 407, 129–134. [Google Scholar] [CrossRef]
  92. Kim, S.H.; Park, E.J.; Lee, C.R.; Chun, J.N.; Cho, N.H.; Kim, I.G.; Lee, S.; Kim, T.W.; Park, H.H.; So, I.; et al. Geraniol Induces Cooperative Interaction of Apoptosis and Autophagy to Elicit Cell Death in PC-3 Prostate Cancer Cells. Int. J. Oncol. 2012, 40, 1683–1690. [Google Scholar] [CrossRef]
  93. Lee, S.; Park, Y.R.; Kim, S.H.; Park, E.J.; Kang, M.J.; So, I.; Chun, J.N.; Jeon, J.H. Geraniol Suppresses Prostate Cancer Growth through Down-Regulation of E2F8. Cancer Med. 2016, 5, 2899–2908. [Google Scholar] [CrossRef]
  94. Rabi, T.; Bishayee, A. D-Limonene Sensitizes Docetaxel-Induced Cytotoxicity in Human Prostate Cancer Cells: Generation of Reactive Oxygen Species and Induction of Apoptosis. J. Carcinog. 2009, 8, 9. [Google Scholar] [CrossRef] [PubMed]
  95. Zhao, Y.; Cheng, X.; Wang, G.; Liao, Y.; Qing, C. Linalool Inhibits 22Rv1 Prostate Cancer Cell Proliferation and Induces Apoptosis. Oncol. Lett. 2020, 20, 289. [Google Scholar] [CrossRef] [PubMed]
  96. Nazıroğlu, M.; Blum, W.; Jósvay, K.; Çiğ, B.; Henzi, T.; Oláh, Z.; Vizler, C.; Schwaller, B.; Pecze, L. Menthol Evokes Ca2+ Signals and Induces Oxidative Stress Independently of the Presence of TRPM8 (Menthol) Receptor in Cancer Cells. Redox Biol. 2018, 14, 439–449. [Google Scholar] [CrossRef]
  97. Kim, S.H.; Nam, J.H.; Park, E.J.; Kim, B.J.; Kim, S.J.; So, I.; Jeon, J.H. Menthol Regulates TRPM8-Independent Processes in PC-3 Prostate Cancer Cells. Biochim. Biophys. Acta Mol. Basis Dis. 2009, 1792, 33–38. [Google Scholar] [CrossRef]
  98. Park, E.J.; Kim, S.H.; Kim, B.J.; Kim, S.Y.; So, I.; Jeon, J.H. Menthol Enhances an Antiproliferative Activity of 1α,25-Dihydroxyvitamin D3 in LNCaP Cells. J. Clin. Biochem. Nutr. 2009, 44, 125–130. [Google Scholar] [CrossRef]
  99. Wang, Y.; Wang, X.; Yang, Z.; Zhu, G.; Chen, D.; Meng, Z. Menthol Inhibits the Proliferation and Motility of Prostate Cancer DU145 Cells. Pathol. Oncol. Res. 2012, 18, 903–910. [Google Scholar] [CrossRef]
  100. Elbe, E.; Yigitturk, G.; Cavusoglu, T.; Uyanikgil, Y. Apoptotic Effects of Thymol, a Novel Monoterpene Phenol, on Different Types of Cancer. Bratisl. Med. J. 2020, 121, 122–128. [Google Scholar] [CrossRef]
  101. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, 25. [Google Scholar] [CrossRef]
  102. El Mihyaoui, A.; Esteves da Silva, J.C.G.; Charfi, S.; Candela Castillo, M.E.; Lamarti, A.; Arnao, M.B. Chamomile (Matricaria chamomilla L.): A Review of Ethnomedicinal Use, Phytochemistry and Pharmacological Uses. Life 2022, 12, 479. [Google Scholar] [CrossRef] [PubMed]
  103. Srivastava, J.K.; Shankar, E.; Gupta, S. Chamomile: A Herbal Medicine of the Past with a Bright Future (Review). Mol. Med. Rep. 2010, 3, 895–901. [Google Scholar] [CrossRef] [PubMed]
  104. Chauhan, R.; Singh, S.; Kumar, V.; Kumar, A.; Kumari, A.; Rathore, S.; Kumar, R.; Singh, S. A Comprehensive Review on Biology, Genetic Improvement, Agro and Process Technology of German Chamomile (Matricaria chamomilla L.). Plants 2022, 11, 29. [Google Scholar] [CrossRef] [PubMed]
  105. Srivastava, J.K.; Gupta, S. Antiproliferative and Apoptotic Effects of Chamomile Extract in Various Human Cancer Cells. J. Agric. Food Chem. 2007, 55, 9470–9478. [Google Scholar] [CrossRef] [PubMed]
  106. Ranasinghe, P.; Pigera, S.; Premakumara, G.S.; Galappaththy, P.; Constantine, G.R.; Katulanda, P. Medicinal Properties of “true” Cinnamon (Cinnamomum zeylanicum): A Systematic Review. BMC Complement. Altern. Med. 2013, 13, 275. [Google Scholar] [CrossRef]
  107. Yanakiev, S. Effects of Cinnamon (Cinnamomum spp.) in Dentistry: A Review. Molecules 2020, 25, 4184. [Google Scholar] [CrossRef] [PubMed]
  108. European Medicines Agency Assessment Report on Cinnamomum Verum J. S. Presl, Cortex and Corticis Aetheroleum. Available online: https://www.ema.europa.eu/en/documents/herbal-report/assessment-report-cinnamomum-verum-j-s-presl-cortex-corticis-aetheroleum_en.pdf (accessed on 20 July 2022).
  109. Denkova-Kostova, R.; Teneva, D.; Tomova, T.; Goranov, B.; Denkova, Z.; Shopska, V.; Slavchev, A.; Hristova-Ivanova, Y. Chemical Composition, Antioxidant and Antimicrobial Activity of Essential Oils from Tangerine (Citrus reticulata L.), Grapefruit (Citrus paradisi L.), Lemon (Citrus lemon L.) and Cinnamon (Cinnamomum zeylanicum Blume). Z. Naturforschung—Sect. C J. Biosci. 2021, 76, 175–185. [Google Scholar] [CrossRef] [PubMed]
  110. Gopalakrishnan, S.; Ediga, H.H.; Sreenivasa Reddy, S.; Bhanuprakash Reddy, G.; Ismail, A. Procyanidin-B2 Enriched Fraction of Cinnamon Acts as a Proteasome Inhibitor and Anti-Proliferative Agent in Human Prostate Cancer Cells. IUBMB Life 2018, 70, 445–457. [Google Scholar] [CrossRef]
  111. Gloria, A. Moore Oranges and Lemons: Clues to the Taxonomy of Citrus from Molecular Markers. Trends Genet. 2001, 17, 536–540. [Google Scholar] [CrossRef]
  112. Dosoky, N.S.; Setzer, W.N. Biological Activities and Safety of Citrus spp. Essential Oils. Int. J. Mol. Sci. 2018, 19, 966. [Google Scholar] [CrossRef]
  113. Ben Hsouna, A.; Ben Halima, N.; Smaoui, S.; Hamdi, N. Citrus lemon Essential Oil: Chemical Composition, Antioxidant and Antimicrobial Activities with Its Preservative Effect against Listeria Monocytogenes Inoculated in Minced Beef Meat. Lipids Health Dis. 2017, 16, 146. [Google Scholar] [CrossRef] [PubMed]
  114. De Groot, A.C.; Schmidt, E. Essential Oils, Part I: Introduction. Dermatitis 2016, 27, 39–42. [Google Scholar] [CrossRef] [PubMed]
  115. Deng, W.; Liu, K.; Cao, S.; Sun, J.; Zhong, B.; Chun, J. Chemical Composition, Antimicrobial, Antioxidant, and Antiproliferative Properties of Grapefruit Essential Oil Prepared by Molecular Distillation. Molecules 2020, 25, 217. [Google Scholar] [CrossRef] [PubMed]
  116. Dosoky, N.S.; Setzer, W.N. Chemical Composition and Biological Activities of Essential Oils of Curcuma Species. Nutrients 2018, 10, 1196. [Google Scholar] [CrossRef] [PubMed]
  117. Ibáñez, M.D.; Blázquez, M.A. Curcuma longa l. Rhizome Essential Oil from Extraction to Its Agri-Food Applications. A Review. Plants 2021, 10, 44. [Google Scholar] [CrossRef] [PubMed]
  118. Tibenda, J.J.; Yi, Q.; Wang, X.; Zhao, Q. Review of Phytomedicine, Phytochemistry, Ethnopharmacology, Toxicology, and Pharmacological Activities of Cymbopogon Genus. Front. Pharmacol. 2022, 13, 997918. [Google Scholar] [CrossRef] [PubMed]
  119. Bayala, B.; Coulibaly, L.L.; Djigma, F.; Bunay, J.; Yonli, A.; Traore, L.; Baron, S.; Figueredo, G.; Simpore, J.; Lobaccaro, J.M.A. Chemical Composition of Essential Oil of Cymbopogon schoenanthus (L.) Spreng from Burkina Faso, and Effects against Prostate and Cervical Cancer Cell Lines. Molecules 2023, 28, 4561. [Google Scholar] [CrossRef]
  120. Zheng, J.; Zhou, Y.; Li, Y.; Xu, D.P.; Li, S.; Li, H.B. Spices for Prevention and Treatment of Cancers. Nutrients 2016, 8, 495. [Google Scholar] [CrossRef] [PubMed]
  121. Shukla, Y.; Singh, M. Cancer Preventive Properties of Ginger: A Brief Review. Food Chem. Toxicol. 2007, 45, 683–690. [Google Scholar] [CrossRef]
  122. Karna, P.; Chagani, S.; Gundala, S.R.; Rida, P.C.G.; Asif, G.; Sharma, V.; Gupta, M.V.; Aneja, R. Benefits of Whole Ginger Extract in Prostate Cancer. Br. J. Nutr. 2012, 107, 473–484. [Google Scholar] [CrossRef]
  123. Cavanagh, H.M.A.; Wilkinson, J.M. Biological Activities of Lavender Essential Oil. Phytother. Res. 2002, 16, 301–308. [Google Scholar] [CrossRef]
  124. De Groot, A.; Schmidt, E. Essential Oils, Part V: Peppermint Oil, Lavender Oil, and Lemongrass Oil. Dermatitis 2016, 27, 325–332. [Google Scholar] [CrossRef]
  125. Mahboubi, M. Mentha spicata L. Essential Oil, Phytochemistry and Its Effectiveness in Flatulence. J. Tradit. Complement. Med. 2021, 11, 75–81. [Google Scholar] [CrossRef]
  126. Christopoulou, S.D.; Androutsopoulou, C.; Hahalis, P.; Kotsalou, C.; Vantarakis, A.; Lamari, F.N. Rosemary Extract and Essential Oil as Drink Ingredients: An Evaluation of Their Chemical Composition, Genotoxicity, Antimicrobial, Antiviral, and Antioxidant Properties. Foods 2021, 10, 3143. [Google Scholar] [CrossRef]
  127. Petiwala, S.M.; Berhe, S.; Li, G.; Puthenveetil, A.G.; Rahman, O.; Nonn, L.; Johnson, J.J. Rosemary (Rosmarinus officinalis) Extract Modulates CHOP/GADD153 to Promote Androgen Receptor Degradation and Decreases Xenograft Tumor Growth. PLoS ONE 2014, 9, e89772. [Google Scholar] [CrossRef] [PubMed]
  128. Jaglanian, A.; Termini, D.; Tsiani, E. Rosemary (Rosmarinus officinalis L.) Extract Inhibits Prostate Cancer Cell Proliferation and Survival by Targeting Akt and MTOR. Biomed. Pharmacother. 2020, 131, 110717. [Google Scholar] [CrossRef] [PubMed]
  129. Kumar Singh, V.; Das, S.; Kumar Dwivedy, A.; Kumar Chaudhari, A.; Upadhyay, N.; Dubey, N.K. Assessment of Chemically Characterized Salvia sclarea L. Essential Oil and Its Combination with Linalyl Acetate as Novel Plant Based Antifungal, Antiaflatoxigenic and Antioxidant Agent against Herbal Drugs Contamination and Probable Mode of Action. Nat. Prod. Res. 2021, 35, 782–787. [Google Scholar] [CrossRef]
  130. Ovidi, E.; Masci, V.L.; Zambelli, M.; Tiezzi, A.; Vitalini, S.; Garzoli, S. Laurus Nobilis, Salvia sclarea and Salvia Officinalis Essential Oils and Hydrolates: Evaluation of Liquid and Vapor Phase Chemical Composition and Biological Activities. Plants 2021, 10, 707. [Google Scholar] [CrossRef] [PubMed]
  131. Borugă, O.; Ianu, C.; Mişcă, C.; Goleţ, I.; Gruia, A.T.; Horhat, F.G. Thymus Vulgaris Essential Oil: Chemical Composition and Antimicrobial Activity. Med. Life 2014, 7, 56–60. [Google Scholar]
  132. Kowalczyk, A.; Przychodna, M.; Sopata, S.; Bodalska, A.; Fecka, I. Thymol and Thyme Essential Oil—New Insights into Selected Therapeutic Applications. Molecules 2020, 25, 4125. [Google Scholar] [CrossRef]
  133. Salehi, B.; Mishra, A.P.; Shukla, I.; Sharifi-Rad, M.; Contreras, M.d.M.; Segura-Carretero, A.; Fathi, H.; Nasrabadi, N.N.; Kobarfard, F.; Sharifi-Rad, J. Thymol, Thyme, and Other Plant Sources: Health and Potential Uses. Phytother. Res. 2018, 32, 1688–1706. [Google Scholar] [CrossRef]
  134. European Medicines Agency Assessment Report on Thymus vulgaris L., Thymus zygis L., Aetheroleum. Available online: https://www.ema.europa.eu/en/documents/herbal-report/final-assessment-report-thymus-vulgaris-l-thymus-zygis-l-aetheroleum-revision-1_en.pdf (accessed on 20 July 2022).
  135. Paiano, R.B.; Bonilla, J.; De Sousa, R.L.M.; Moreno, A.M.; Baruselli, P.S. Chemical Composition and Antibacterial Activity of Essential Oils against Pathogens Often Related to Cattle Endometritis. J. Infect. Dev. Ctries. 2020, 14, 177–183. [Google Scholar] [CrossRef] [PubMed]
  136. Trindade, G.G.G.; Thrivikraman, G.; Menezes, P.P.; França, C.M.; Lima, B.S.; Carvalho, Y.M.B.G.; Souza, E.P.B.S.S.; Duarte, M.C.; Shanmugam, S.; Quintans-Júnior, L.J.; et al. Carvacrol/β-Cyclodextrin Inclusion Complex Inhibits Cell Proliferation and Migration of Prostate Cancer Cells. Food Chem. Toxicol. 2019, 125, 198–209. [Google Scholar] [CrossRef] [PubMed]
  137. Emami, S.A.; Asili, J.; HosseinNia, S.; Yazdian-Robati, R.; Sahranavard, M.; Tayarani-Najaran, Z. Growth Inhibition and Apoptosis Induction of Essential Oils and Extracts of Nepeta cataria L. on Human Prostatic and Breast Cancer Cell Lines. Asian Pac. J. Cancer Prev. 2016, 17, 125–130. [Google Scholar] [CrossRef]
  138. Dwivedi, V.; Shrivastava, R.; Hussain, S.; Ganguly, C.; Bharadwaj, M. Comparative Anticancer Potential of Clove (Syzygium aromaticum)—An Indian Spice-Against Cancer Cell Lines of Various Anatomical Origin. Asian Pac. J. Cancer Prev. 2011, 12, 1989–1993. [Google Scholar]
  139. Medina-Holguín, A.L.; Omar Holguín, F.; Micheletto, S.; Goehle, S.; Simon, J.A.; O’Connell, M.A. Chemotypic Variation of Essential Oils in the Medicinal Plant, Anemopsis Californica. Phytochemistry 2008, 69, 919–927. [Google Scholar] [CrossRef]
Figure 1. EOs and their bioactive compounds have roles in multiple pathways in PCa cells. EOs have cell membrane permeability and interact with several cellular targets involved in different pathways. EOs are able to interact with different targets of the PI3K/Akt pathway, leading to cell cycle arrest, apoptosis, the inhibition of cell growth and proliferation. EOs can also play a role as proteasome inhibitors, resulting in apoptosis, a decrease in cell proliferation and the suppression of angiogenesis. EOs modulate DNA damage repair mechanisms by acting as DNA polymerase inhibitors, which leads to PARP cleavage, resulting in apoptosis. EOs were demonstrated to induce mitochondrial stress, leading to changes in the expression of BCL2/BAX genes and membrane depolarization, resulting in the increased release of cytochrome-C to the cytoplasm and the induction of apoptosis. In addition, EOs increase intracellular ROS levels, resulting in PCa cells’ apoptosis.
Figure 1. EOs and their bioactive compounds have roles in multiple pathways in PCa cells. EOs have cell membrane permeability and interact with several cellular targets involved in different pathways. EOs are able to interact with different targets of the PI3K/Akt pathway, leading to cell cycle arrest, apoptosis, the inhibition of cell growth and proliferation. EOs can also play a role as proteasome inhibitors, resulting in apoptosis, a decrease in cell proliferation and the suppression of angiogenesis. EOs modulate DNA damage repair mechanisms by acting as DNA polymerase inhibitors, which leads to PARP cleavage, resulting in apoptosis. EOs were demonstrated to induce mitochondrial stress, leading to changes in the expression of BCL2/BAX genes and membrane depolarization, resulting in the increased release of cytochrome-C to the cytoplasm and the induction of apoptosis. In addition, EOs increase intracellular ROS levels, resulting in PCa cells’ apoptosis.
Pharmaceutics 16 00583 g001
Figure 2. Representative illustration of the chemical structures of the bioactive compounds found in essential oils tested for anti-prostate cancer activity.
Figure 2. Representative illustration of the chemical structures of the bioactive compounds found in essential oils tested for anti-prostate cancer activity.
Pharmaceutics 16 00583 g002
Table 1. Essential oils investigated for in vitro anti-prostate cancer potential, and the major results reported.
Table 1. Essential oils investigated for in vitro anti-prostate cancer potential, and the major results reported.
Common NameScientific NameCell LineEffectsReferences
Balsam firAbies balsameaPC3Inhibition of cell growth[36]
BasilOcimum basilicumPC3 and LNCaPInhibition of cell growth[37]
ChamomileAnthemis nobilisPC3Inhibition of cell growth and induction of apoptosis[38]
Cinnamon Cinnamomum zeylanicumPC3Inhibition of cell growth[38]
CitronellaCymbopogon nardusLNCaPInhibition of cell growth, cell cycle arrest and changes to cell morphology[39]
EmbiraXylopia frutescensPC-3MInhibition of cell growth[40]
GingerZingiber officialePC3Inhibition of cell growth[38]
PC3 and LNCaPInhibition of cell growth[37]
Grapefruit Citrus paradisiPC3Inhibition of cell growth[38]
HorsemintMenthalongifoliaLNCaPInhibition of cell growth[41]
Jasmine Jasminum grandifloraPC3Inhibition of cell growth[38]
KumquatFortunella margaritaLNCaPInhibition of cell growth, induction of apoptosis, increased Bax/Bcl2 ratio, induced caspase-3 cleavage and inhibition of inflammation due to decreased expression of NFκB and Cox-2[42]
Lavender Lavandula stoechasPC3Inhibition of cell growth[38]
LNCaPInhibition of cell growth[43]
Lavandula angustifoliaPC3 and DU145Inhibition of cell growth and migration, induction of apoptosis and cell cycle arrest[44]
Lavandula officialisPC3Slight Inhibition of cell growth[45]
Lemon Citrus limonPC3Inhibition of cell growth[38]
Lemon grassCymbopogon citratusPC3 and LNCaPInhibition of cell growth[46]
LippiaLippia multifloraPC3 and LNCaPInhibition of cell growth[37]
MandarinCitrus reticluataPC3Inhibition of cell growth, induction of apoptotic DNA fragmentation and ROS generation; alterations in the expression of apoptotic and anti-apoptotic genes[47]
MentrastoAgeratum conyzoidesPC3 and LNCaPInhibition of cell growth[37]
Mullein nightshade Salanum erianthumPC3Inhibition of cell growth[48]
Myrtle Myrtus communisPC3 and DU145Inhibition of cell growth and migration and induction of apoptosis[49]
Navel orangeCitrus sinensis22RV1Inhibition of cell growth and induction of apoptosis[50]
OreganoOriganum vulgarePC3Inhibition of cell growth; induction of apoptosis; cellular damage; DNA fragmentation; enhanced Bax expression, cytochrome c release and caspase-3 activation; and decreased Bcl2 expression[51]
PeppermintMentha piperitaLNCaPInhibition of cell growth[41]
Potato treeSalanum macranthumPC3Inhibition of cell growth[52]
Rose Rosa centifoliaPC3Inhibition of cell growth[38]
RosemaryRosmarinus officinalisLNCaPInhibition of cell growth[53]
SageSalvia aureaDU145Inhibition of cell growth; induction of apoptosis; DNA fragmentation; increased LDH, caspase activity, Bax/Bcl-2 ratio and ROS generation; and decreased GSH[54]
Salvia JudaicaDU145Inhibition of cell growth; induction of apoptosis; DNA fragmentation; increased LDH, caspase activity, Bax/Bcl-2 ratio and ROS generation; and decreased GSH[54]
Salvia viscosaDU145Inhibition of cell growth; induction of apoptosis; DNA fragmentation; increased LDH, caspase activity, Bax/Bcl-2 ratio and ROS generation; and decreased GSH[54]
SpearmintMentha spicataLNCaPInhibition of cell growth[41]
PC3Inhibition of cell growth[38]
Thyme Thymus vulgarisPC3Inhibition of cell growth[38]
Tsauri grass Cymbopogon giganteusPC3 and LNCaPInhibition of cell growth[46]
TurmericCurcuma longaPC3Inhibition of cell growth[52]
LNCaPInhibition of cell growth[55]
Wild mintMenthaarvensisLNCaPInhibition of cell growth[41]
Wild spikenardHyptis suaveolensLNCaPInhibition of cell growth and cell cycle arrest[56]
Achillea wilhelmsiiPC3Inhibition of cell growth[57]
Aloysia polystachyaPC3Inhibition of cell growth[58]
Amomum tsao-koPC3Inhibition of cell growth[59]
Anaxagorea brevipesPC3Inhibition of cell growth[60]
Annona sylvaticaPC3Inhibition of cell growth[61]
Artemisia arborescensLNCaP and DU145Inhibition of cell growth, DNA fragmentation and ROS generation[62]
Bursera glabrifoliaPC3Inhibition of cell growth[63]
Euodia ruticarpaPC3Inhibition of cell growth[64]
Guatteria ellipticaPC3Inhibition of cell growth[65]
Hedychium spicatumPC3Inhibition of cell growth, induction of apoptosis, cell cycle arrest, increased caspase activity, Bax/Bcl-2 ratio and ROS generation[66]
Hedychium coccineum,
Hedychium gardnerianum,
Hedychium greenii and
Hedychium. griffithianum
PC3Inhibition of cell growth[67]
Hypericum hircinumPC3Inhibition of cell growth[68]
Iryanthera polyneuraPC3Inhibition of cell growth[69]
Liquidambar orientalisPC3Inhibition of cell growth[70]
Symphyopappus itatiayensis, Myrciaria floribundus, Talauma ovata, Psidium cattleyanum, Nectandra megapotamicaPC3Inhibition of cell growth[71]
Guatteria pogonopusPC-3MInhibition of cell growth[72]
Perralderia coronopifoliaPC3Inhibition of cell growth[73]
Pinus mugoDU145Inhibition of cell growth and constitutive STAT3 activation; decreased expression of cyclin D1, Bcl-2, survivin, XIAP, Cox2 and IL-6; decrease in GSH levels and increase in ROS generation; induced caspase-3 and PARP cleavage; and inhibition of cell migration[74]
Zataria MultifloraPC3Inhibition of cell growth, induction of apoptosis, DNA fragmentation, cell cycle arrest, increased ROS generation and caspase activation, upregulation of Bax and downregulation of Bcl-2 expression[75]
Table 2. Essential oils’ constituents investigated for their in vitro anti-prostate cancer potential and the major resultsreported.
Table 2. Essential oils’ constituents investigated for their in vitro anti-prostate cancer potential and the major resultsreported.
ConstituentPlantCell LineEffectsReferences
α-humulene Salvia speciesLNCaPInhibition of cell growth[76]
α-pineneRosemary, lavender and othersPC3 and DU145Inhibition of cell growth, induced apoptosis and cell cycle arrest[77]
β-Caryophyllene oxideCinnamon, oregano, clove and black pepperPC3Inhibition of cell growth; induced apoptosis by inhibiting PI3K/AKT/mTOR/S6K1 signaling; reduced mitochondrial membrane potential; cytochrome c release; activating caspase-3; cleavage of PARP; ROS generation; downregulation of Bcl-2, Bcl-xL, survivin, IAP-1, IAP-2 and cyclin D1; upregulation of p53 and p21; and downregulation of COX-2 and VEGF[78,79]
DU145Inhibition of cell growth and invasion and constitutive STAT3 activation[80]
β-elemeneCurcuma and Cymbopogon speciesPC3 and DU145Inhibition of cell growth; induced apoptosis through cleaved caspase-3, caspase-9 and increasing PARP levels; and downregulated and Bcl-2 [81]
CarvacrolThyme, oregano and othersDU145Inhibition of cell growth, induction of apoptosis, cell cycle arrest increased caspase-3 activation and ROS generation[82]
PC3Inhibition of cell growth; migration and invasion; induction of apoptosis increased caspase activation and ROS generation; disruption of mitochondrial membrane potential; cell cycle arrest; upregulation of Bax and downregulation of Bcl-2 expression; decreased expression of Notch1, Jagged-1, cyclin D1 and CDK4; and increased expression of p21 [83]
PC3 and DU145Inhibition of cell growth, migration, and invasion; decreased TRPM7-like current and reduced MMP-2 protein expression and F-actin dynamics; alterations of PI3K/Akt and MEK/MAPK signaling pathways[84,85]
CinnamaldehydeCinnamonPC3 and LNCaPInhibition of cell growth and proteasome activity; upregulated Hsp70 and downregulated VEGF and VEGFR expression[86]
Prostate CAFInhibition of cell growth, induction of apoptosis induction, cell cycle arrest, increased ROS generation and caspase activation and decreased GSH levels[87]
Prostate CAFRelieves the immunosuppressive effects in a TLR4-dependent manner[88]
Cinnamic acidCinnamonPC3 and LNCaPInhibition of cell growth and proteasome activity; upregulated Hsp70 and downregulated VEGF and VEGFR expression[86]
CitralCymbopogon speciesPC3 and PC3M Inhibition of cell growth, reduced clonogenic potential, induced morphological alterations, expulsion of lipid droplets, activation of AMPK protein expression, induction of apoptosis, DNA fragmentation, upregulation of Bax and downregulation of Bcl-2 expression [89]
EugenolCinnamon, clove and othersDU145Inhibition of cell growth[90]
PC3 and LNCaPInhibition of cell growth and proteasome activity; upregulated Hsp70 and downregulated VEGF and VEGFR expression[86]
GeraniolCymbopogon speciesPC3Inhibition of cell growth; increased LDH and caspase-3 activity; induced mitochondrial membrane depolarization and cell cycle arrest at the G1 phase; reduced expressions of cyclin A, B, D and E, CDK1 and CDK4, and Bcl-2 and Bcl-w; elevated expressions of p21 and p27 and Bax and BNIP3[91]
Inhibition of cell growth, induced autophagy and inhibited AKT-mTOR signaling [92]
Downregulated the transcription factor E2F8 [93]
LimoneneCitrus and Mentha speciesDU145Inhibition of cell growth, induction of apoptosis, ROS generation, DNA fragmentation, caspase-3 and caspase-9 cleavage, upregulation of p21, p53 and Bad, downregulation of Bcl-xL and cleavage of PARP protein[94]
LinaloolLavender, Salvia species and othersPC3 and DU145Inhibition of cell growth and migration, induction of apoptosis and cell cycle arrest[44]
22RvlInhibition of cell growth; induction of apoptosis; cell cycle arrest; increased expression of Bax, Bcl-2, p53, DR4, DR5; and cleaved caspases[95]
Linalyl acetateLavender, Salvia species and othersPC3 and DU145Inhibition of cell growth and migration, induction of apoptosis and cell cycle arrest[44]
MentholMentha speciesPC3, LNCaP and DU145 Inhibition of cell growth[96]
PC3 and LNCaPInhibition of cell growth[97]
LNCaPInhibition of cell growth[98]
DU145Inhibition of cell growth and migration and cell cycle arrest [99]
ThymolThymePC3 and DU145Inhibition of cell growth and induction of apoptosis[100]
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

Pimentel, L.S.; Bastos, L.M.; Goulart, L.R.; Ribeiro, L.N.d.M. Therapeutic Effects of Essential Oils and Their Bioactive Compounds on Prostate Cancer Treatment. Pharmaceutics 2024, 16, 583. https://doi.org/10.3390/pharmaceutics16050583

AMA Style

Pimentel LS, Bastos LM, Goulart LR, Ribeiro LNdM. Therapeutic Effects of Essential Oils and Their Bioactive Compounds on Prostate Cancer Treatment. Pharmaceutics. 2024; 16(5):583. https://doi.org/10.3390/pharmaceutics16050583

Chicago/Turabian Style

Pimentel, Leticia Santos, Luciana Machado Bastos, Luiz Ricardo Goulart, and Lígia Nunes de Morais Ribeiro. 2024. "Therapeutic Effects of Essential Oils and Their Bioactive Compounds on Prostate Cancer Treatment" Pharmaceutics 16, no. 5: 583. https://doi.org/10.3390/pharmaceutics16050583

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop