Next Article in Journal
Cellular and Molecular Progression of Prostate Cancer: Models for Basic and Preclinical Research
Next Article in Special Issue
Molecular Insights and Emerging Strategies for Treatment of Metastatic Uveal Melanoma
Previous Article in Journal
Erratum: Lee, H.H., et al. Histone 2A Family Member J Drives Mesenchymal Transition and Temozolomide Resistance in Glioblastoma Multiforme. Cancers 2020, 12, 98
Previous Article in Special Issue
Loss of Two-Pore Channel 2 (TPC2) Expression Increases the Metastatic Traits of Melanoma Cells by a Mechanism Involving the Hippo Signalling Pathway and Store-Operated Calcium Entry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Essential Oils and Their Main Chemical Components: The Past 20 Years of Preclinical Studies in Melanoma

1
Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
2
Department of Chemistry and Technologies of Drugs, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy
3
Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2020, 12(9), 2650; https://doi.org/10.3390/cancers12092650
Submission received: 31 July 2020 / Revised: 7 September 2020 / Accepted: 14 September 2020 / Published: 16 September 2020
(This article belongs to the Special Issue Advances and Novel Treatment Options in Metastatic Melanoma)

Abstract

:

Simple Summary

In the last years, targeted therapy and immunotherapy modified the landscape for metastatic melanoma treatment. These therapeutic approaches led to an impressive improvement in patients overall survival. Unfortunately, the emergence of drug resistance and side effects occurring during therapy strongly limit the long-term efficacy of such treatments. Several preclinical studies demonstrate the efficacy of essential oils as antitumoral agents, and clinical trials support their use to reduce side effects emerging during therapy. In this review we have summarized studies describing the molecular mechanism through which essential oils induce in vitro and in vivo cell death in melanoma models. We also pointed to clinical trials investigating the use of essential oils in reducing the side effects experienced by cancer patients or those undergoing anticancer therapy. From this review emerged that further studies are necessary to validate the effectiveness of essential oils for the management of melanoma.

Abstract

The last two decades have seen the development of effective therapies, which have saved the lives of a large number of melanoma patients. However, therapeutic options are still limited for patients without BRAF mutations or in relapse from current treatments, and severe side effects often occur during therapy. Thus, additional insights to improve treatment efficacy with the aim to decrease the likelihood of chemoresistance, as well as reducing side effects of current therapies, are required. Natural products offer great opportunities for the discovery of antineoplastic drugs, and still represent a useful source of novel molecules. Among them, essential oils, representing the volatile fraction of aromatic plants, are always being actively investigated by several research groups and show promising biological activities for their use as complementary or alternative medicine for several diseases, including cancer. In this review, we focused on studies reporting the mechanism through which essential oils exert antitumor action in preclinical wild type or mutant BRAF melanoma models. We also discussed the latest use of essential oils in improving cancer patients’ quality of life. As evidenced by the many studies listed in this review, through their effect on apoptosis and tumor progression-associated properties, essential oils can therefore be considered as potential natural pharmaceutical resources for cancer management.

1. Introduction

Melanoma is the third most common cutaneous malignancy and one of the most dangerous forms of skin cancer. It is increasing worldwide and is caused by several factors, including environmental and genetic ones. In primary tumors, the most frequent driver oncogenic mutations (BRAF, NRAS, KIT), as well as mutations responsible for intrinsic resistance, have been identified with genomic analyses. Genetic alterations developed during the acquired resistance and genetic lesions leading to metastasis spreading were also identified [1].
The use of surgery in case of primary tumors is considered curative. With the advent of targeted therapy (MAPK pathway inhibitors) and immunotherapy (immune checkpoint inhibitors), treatment of metastatic melanoma has changed dramatically in recent years. These therapeutic strategies generated unprecedented improvement in patients’ survival [2,3]. Targeted therapy, mostly represented by BRAF inhibitors, shows efficacy in the treatment of BRAF mutated metastatic melanoma when administered as single agents or in combination with MEK inhibitors [4]. Immune checkpoint inhibitors, such as anti-CTLA4 and anti-PD1 monoclonal antibodies, are able to activate the immune response in the host through their ability to unmask the inhibition of the host immune cells [5]. Targeted therapy in combination with immune checkpoint inhibitors has also showed encouraging results in the last few years [6]. Patients who are not eligible candidates for targeted therapy or immunotherapy are treated with systemic chemotherapy, such as dacarbazine, taxanes or temozolomide, while radiation therapy is used for brain metastases or for the treatment of oligometastatic disease [7]. Despite this clinical success, treatment of metastatic melanoma is still ineffective in about half of the treated patients and is often limited by numerous side effects and by the emergence of resistance to both targeted therapy and immunotherapy [8,9,10].
The diagnosis of melanoma and the therapeutic approaches used to contrast the disease, strongly affect the patient’s quality of life. In fact, melanoma patients often experience the risk of disease progression, or even of new primary disease, for several years after diagnosis. Often, anxiety, depression and fear, leading to impaired social functioning, are evidenced in melanoma survivors. For these reasons, there is an urgent need to identify new therapeutic strategies for melanoma patients who do not respond or relapse after therapy, as well as to define new options to improve their quality of life.
In the last two decades, many new anticancer agents have been discovered from natural products as an alternative option to current cancer therapy. This is due to high cost, emergence of drug resistance and the side effects of standard therapies. Several plant-derived drugs, such as camptothecin, vinblastine, vincristine, etoposide, taxol, and paclitaxel have found wide applications in cancer therapy [11,12]. Current cancer research has brought about many promising preclinical results regarding the antiproliferative, anti-inflammatory, antioxidant, antiangiogenic and antimetastatic effect of essential oils (EOs), which are relevant components of aromatic plants. In addition, chemoprevention by EOs could represent a potentially effective option in the fight against cancer and, in particular, against melanoma. At present, EOs are used as a non-invasive therapy with minimal risk intervention that could potentially improve the quality of life of cancer patients and could alleviate the severity of treatment-related liver injuries or, more generally, symptoms in cancer patients undergoing chemotherapy. Alleviation of collateral effects would enhance prognosis status and patients’ survival.
From a survey in scopus [13] with the “essential oil” and “melanoma” keywords, a list of almost 170 references were retrieved and analyzed. Focusing our attention on papers published in the last 20 years, we will summarize and discuss the chemical composition of EOs used in melanoma models, and the molecular mechanism through which EOs and their main components exert an antitumor effect in preclinical melanoma models carrying wild type or mutated BRAF. One section is dedicated to the use of EOs in clinical trials for managing cancer symptoms. This comprehensive summary could be a useful source for a better understanding of EOs’ mechanism of action. It could also help researchers to appreciate and consider the importance of EOs being a potential adjuvant to enhance the efficacy of current available therapy, as well as, for improving patients’ quality of life.
Taking into consideration the studies presented in this review, EOs showing antitumor activity could represent a good opportunity for combination therapy or, particularly, for reducing those side effects caused by current treatments.

2. Essential Oils

A wide number of studies have reported that plants contain valuable compounds, including bioactive EOs [14]. EOs are a complex combination of chemical aromatic-smelling low-molecular weight compounds. They are derived from the plants’ secondary metabolism [15], leading predominately to monoterpenes [16], sesquiterpenes [17] and their oxygenated derivatives [18]. They are biosynthesized in different plant organs and parts such as flowers, leaves, fruits and roots [15] and are industrially produced mainly by hydro- or steam-distillation [19,20]. Chemical qualitative and quantitative composition of EOs, which are composed of volatile compounds, is determined using a combination of Gas Chromatography/Flame Ionization Detection, Gas Chromatography/Mass Spectrometry and determination of their Kovats index [21] and/or Linear Retention Indices [22]. The chemical composition of aromatic plants furnishing EOs active against melanoma models is reported in Table 1. Common and family names of the plants, as well as the percentage of the main components identified in each EO, are also included.
Anticancer activities of EO mixtures as a whole and only a hypothetical correlation between the chemical profile and the anticancer activity have been described for some EOs. In some cases, EOs’ bioactive properties were related to the anticancer activity of specific components. Due to EOs’ complex chemical composition, the additive, synergistic or anti-synergistic roles of individual EO constituents are currently being investigated to establish the possible pharmacological activity thereof [92,93]. This categorization is even more complicated due to the ‘chemotype’ concept, in which the same plant could produce different EOs characterized by different chemical composition profiles and, hence, different biological properties [94]. Ocimum tenuiflorum (holy basil), Thymus vulgaris (thyme), Lavandula angustifolia (lavender) and Mentha piperita (peppermint) are examples of plants with several chemotypes [95]. Despite this mix up, an effort to characterize EOs is currently taking place in medical and pharmaceutical fields. This characterization could help to obtain a clearer indication of EOs’ uses in traditional medicine, chemical or pharmaceutical, as witnessed by almost 5000 articles published in PubMed (http://pubmed.ncbi.nlm.gov/). In the last decade, an average positive increment of more than 7% per year was observed in this field [96].
When referring to EOs, their chemical composition and biological activities strictly depend on habitat, climate condition, season, agronomic practices, soil type, extraction procedures, as well as the harvesting stages and storage conditions of plants [31,36,37,51,75,97,98,99,100,101,102,103]. All these elements should be taken into account. By analyzing gene expression patterns and metabolic fingerprints, recently Spring’s group identified environmental factors as regulatory factors of biosynthetic pathways [104]. Substantial variability was also reported according to the part of plant used for extraction of EOs [83]. Examples are EOs from Helichrysum microphyllum [45] and from Liriodendron tulipifera, and their main components β-elemene and (E)-nerolidol, showing antiproliferative activity in human melanoma cells strictly depending on harvesting period [51]. Moreover, EOs from Chrysanthemum boreale Makino, showed different levels of their component contents and bioactivities among the harvesting stages [31], while phytoconstituents and bioactivities of EOs from Curcuma kwangsiensis, strictly depended on the natural habitat [37]. Another example is provided by chemotaxonomical analysis of Artemisia absinthium, Salvia officinalis, Tanacetum vulgare and Thuja occidentalis, the amount of thujones (α-thujone and β-thujone) present in the EOs of the four species being strictly related to the plant organ and to its developmental phase [105]. Moreover, exposure to ultraviolet (UV) light was reported to induce deterioration of EOs’ biochemical profiles [49] or activation of some EOs [34]. In this regard, the antiproliferative effect of both Citrus medica and Citrus bergamia EOs and their constituent bergapten, was observed in human melanoma cells after exposure to UV irradiation, thus indicating UV irradiation’s ability to activate EOs [34]. In some cases, derivatives of EOs are designed to increase EOs’ half-life. This is the case of farnesyl-O-acetylhydroquinone, geranyl-O-acetylhydroquinone, geranyl ester and farnesyl ester derived from geraniol and farnesol. Structure–activity relationship studies reported the ability of these derivatives to reduce proliferation of mouse melanoma cells more efficiently than the parental compounds [106,107]. Furthermore, 6-(menthoxybutyryl)thymoquinone, the terpene conjugate derivative of thymoquinone, was shown to be more active in human melanoma cells than its parental compound [108]. A cumulative impact of some EOs’ components was also evidenced: farnesol and nerolidol in combination showed an enhanced antiproliferative effect in mouse melanoma cells when compared to exposure to single treatments [109].
For all their biological effects, EOs can be considered as an interesting source for therapeutic, food preservation and/or nutraceutical uses [110,111,112,113,114].

3. Mechanism of Action of EOs in Melanoma

EOs and some of their components exert antitumor activity in melanoma models by affecting multiple pathways, including inhibition of in vitro cell proliferation, alteration of cell distribution in the different cell cycle phases, induction of apoptosis, inhibition of in vitro cell invasion and migration, in vivo tumor growth and metastasization and in vitro/in vivo angiogenesis. Several EOs also act as chemopreventive agents in melanoma, reduce melanogenesis and show antioxidant properties.
The most frequently used human melanoma models are represented by M14, A2058, A375 cells and SK-MEL variants. Murine melanoma B16 cells, that originate in the syngeneic C57BL/6 (H-2b) mouse strain, and its derivatives B16-F1, B16-F10, B16-F10-Nex2, B16-Bl6, B164A5 [115,116,117,118] represent the most used in vivo models. They are employed to evaluate the effect of EOs on tumor growth and metastasization, as well as tumor angiogenesis, after subcutaneous or intravenous (lateral tail vein) cell injection [119]. In some cases subretinal, intradermal, intracerebral injection of melanoma cells is employed [38,86,120,121]. Routes of EO administration include oral, intraperitoneal, intravitreal, peritumoral, topical as well as inhalation (fragrant environmental).

3.1. Inhibition of Cell Proliferation

Cell viability and proliferation can be detected by a wide range of assays based on several cell functions, including mitochondrial enzyme and cellular uptake activity, cell membrane permeability and ATP production. The most used assay is a colorimetric test that evaluates the reduction of yellow 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide by mitochondrial succinate dehydrogenase [122]. Analysis of cell proliferation can also be performed by alamarBlue and PrestoBlue assays, both using reduction of resazurin as an indicator of cell viability [122]. Detection of 5-bromo-2′-deoxyuridine (BrdU)- or 5-ethynyl-2′-deoxyuridine (EdU)- labeled DNA also represents a valid method to detect viable cells: BrdU or EdU are efficiently incorporated into DNA of replicating cells by substituting thymidine and their binding to DNA can be detected with specific antibodies [123]. Moreover, the thymidine incorporation assay, based on measuring the incorporation of methyl-[3H]-thymidine into the DNA of dividing cancer cells, is frequently used to estimate cell proliferation [124].
A high number of EOs and their components have been found to reduce in vitro proliferation/viability of melanoma cells, and in some cases the cytotoxic potential has been predicted by in silico studies [125].
EOs and their main components demonstrated to reduce in vitro proliferation of melanoma cells are reported in Table 2. The used melanoma models are also indicated in the Table.

3.2. Alteration of Cell Cycle Distribution

Cell cycle transition is a process involving multiple checkpoints, which control growth signals, cell size and DNA integrity. It is regulated by active forms of cyclin-dependent kinases, which control the passage of cells from one phase of the cell cycle to another. Cyclin-dependent kinases act as cell cycle regulators to elicit cell cycle arrest in response to DNA damage [153]. Cytofluorimetric analysis of cells stained with the DNA intercalator, propidium iodide, represents the most used methods to analyze cell cycle transition.
Several authors demonstrated the ability of EOs or their constituents to induce DNA damage in melanoma cells, and consequently inducing delay/arrest in the different cell cycle phases. Recently, Ramadam et al. reported the Melaleuca alternifolia EO (Tea tree oil, TTO) ability to induce cell cycle arrest at the G2/M phase of A375 cells [129], while in a previously published study, Beilharz’s group described the TTO ability to elicit G1 cell cycle arrest in B16 cells [130]. In addition, α-santalol, the main component of Santalum album (Sandalwood oil) [154], in UACC-62 cells induced G2/M phase arrest through down-regulation of proteins critical for G2/M transition, such as cyclin A/Cdk2 and cyclin B/Cdc2 complexes, as well as microtubule depolymerisation. It also increased expression of wild-type p53 [155]. Eugenol, a component present in many EOs including Syzygium aromaticum (Clove oil) and Cinnamomum zeylanicum [156], abrogated the G2/M phase in A2058 but not in SK-MEL-28 cells. It was also reported to arrest WM1205Lu cells in the S phase of the cell cycle through the inhibition of E2F1 transcription factor activity. E2F1 is a key cell cycle regulator, targeting genes that encode proteins involved in G1/S transition [134,135]. Eugenol also reduced the expression of proliferation cell nuclear antigen in A2058 cells [134]. Given the role of E2F1 in melanoma progression and resistance to therapy [157,158], the authors also suggested that eugenol could be developed as an E2F-targeted agent for melanoma treatment. Farnesol (a mixture of trans, trans and cis, trans isomers) and nerolidol, present in different EOs including that from Psidium guajava [159], induced an increase in cells in the G0/G1 phase, concomitant with a reduction in the S phase, and a cumulative impact of the two compounds was evidenced in B16 cells [109].
The different response between cell lines sometimes observed after exposure to the same EO indicate a non-generalizable and cell-type specific effect or is due to differences in the composition of the EOs’ used in the distinct studies. The genetic background of the cell lines tested should be also considered: some melanoma cells harbor activating BRAFV600E mutations (A375, M14, A2058, SK-MEL-5, SK-MEL-19, SK-MEL-28, UACC-62, UACC-257, 518A2, G-361, WM266) not present in other ones (B16, Sbcl2, CHL1, WM3211, SK-MEL-147, MeWo, RPMI-7932) (https://web.expasy.org/cellosaurus/), or harbor different mutations (i.e., NRAS, CDKN2A, TP53, TERT). A better understanding of these cell-to-cell differences is crucial for a deeper comprehension of the EO mechanism of action.
EOs and their main components demonstrated to induce cell cycle perturbation of melanoma cells are reported in Table 3. The used melanoma models are also listed in the Table.

3.3. Induction of Apoptosis

Apoptosis is a programmed cell death characterized by the activation of a group of intracellular caspases leading to a cascade of events connected with various substrates, including poly(ADP-ribose) polymerase-I (PARP), and internalization by phagocytes [163,164]. Hallmarks of apoptosis include cell shrinkage, formation of apoptotic bodies, DNA fragmentation, heterochromatin aggregation and activation of caspases and substrates such as PARP. Exposure of phosphatidyl serine on the cell surface, change in the mitochondrial membrane potential, as well as change in the ratio of mRNA expression of pro- and anti-apoptotic proteins, also represent features of apoptotic cells. The presence of a sub-G0/G1 population in the cell cycle is considered indicative of DNA damage and apoptosis [165]. Comet assay, a genotoxic test, can be used as an indicator of early apoptosis, since cells entering apoptosis undergo DNA fragmentation resulting in the characteristic images. In these images the tail and the head indicate, respectively, fragmented and intact DNA [166].
Most EOs have been reported to cause cell death of melanoma cells, primarily inducing apoptosis, and hallmarks of apoptosis were recognized in melanoma cells treated with a huge number of EOs or their components. TTO induced apoptosis in A375 cells through the activation of caspases 3, 7 and 9, upregulation of p53 and Bax proapoptotic proteins and downregulation of bcl-2 antiapoptotic protein [129]. TTO and its main active component, terpinen-4-ol, also induced apoptosis in both adriamycin-sensitive and -resistant M14 cells: interaction with plasma membrane and subsequent reorganization of membrane lipid architecture has been identified as a possible mechanism through which TTO induced caspases dependent apoptosis [52,167]. Moreover, EO from Salvia verbenaca showed proapoptotic effects in M14 cells, and the EOs from cultivated plants exhibited major effects when compared to those growing in natural sites [75], thus further confirming the relevance of cultivar conditions on EOs activity. The same group also pointed a proapoptotic activity of Salvia officinalis EO in A375, M14 and A2058 cells, the percentages of main components depending on environmental factors [101]. The authors also suggested activation of apoptosis by Salvia rubifolia and Salvia bracteata EOs in M14 cells using a Comet assay [72,166]. Boswellia carterii EO (frankincense oil) was found to induce apoptosis through downregulation of Bcl-2 and up regulation of Bax proteins in B16-F10 cells, while inducing down regulation of Mcl-1 and cleavage of caspase 3 and 9 and PARP in human melanoma FM94 cells. On the contrary, proliferation of normal human epithelial melanocytes was not affected [146]. EOs extracted from Pituranthos tortuosus and Annona Vepretorum and their major constituents spathulenol, o-cymene and α-pinene, induced apoptosis of B16-F10 cells [25,66].
Hallmarks of apoptosis were also recognized in cells treated with several EO components, including camphene, α-pinene, eugenol, linalool, zerumbone, carvacrol, thujone, curzerene, citral, thymoquinone, isoegomaketone and menthol. Camphene isolated from Piper cernuum EO, induced apoptosis in B16-F10-Nex2 cells through loss of mitochondrial membrane potential, activation of caspases 3, endoplasmic reticulum stress, release of calcium, increased expression of high mobility group box 1 (HMGB1) and cell surface calreticulin [62]. The induction of HMGB1 and calreticulin after treatment with camphene could elicit immunogenic cell death, a relevant pathway for the activation of the immune system [168]. In B16-F10-Nex2 cells, α-pinene, a component present in many EOs including those from Schinus terebinthifolius, Tridax procumbens, Pituranthos tortuosus, Annona Vepretorum and Boswellia carterii [169], induced disruption of the mitochondrial potential, production of reactive oxygen species (ROS), activation of caspase 3, aggregation of heterochromatin, fragmentation of DNA and exposure of phosphatidyl serine on the cell surface [149,169]. Experiments performed to identify the structure/activity relationship, indicated the presence of a double bond in the α-pinene structure as crucial for its cytotoxic potential against both B16-F10-Nex2 and A2058 cells [78].
Proapoptotic properties, in terms of DNA fragmentation, phosphatidylserine exposure, and mitochondrial damage were reported by eugenol and the 6,6′-dibromo-dehydrodieugenol (S) enantiomeric form, in established and primary melanoma cells from patient tissue samples, with no effect on fibroblasts. Clastogenesis analysis and clonogenic assay also pointed out the ability of eugenol, respectively, to induce DNA breaks and to reduce colony forming potential, through a direct cytotoxicity or a lingering antiproliferative effect, in A2058 and SK-MEL-28 cells [134,136,170]. Scanning electron microscopy and transmission electron microscopy demonstrated that linalool, present in several EOs including those from Citrus bergamia, induced morphological changes and apoptosis in RPMI-7932 human melanoma cells while not affecting proliferation of normal keratinocytes [160].
The proapoptotic effect of zerumbone, one of the main constituents of EOs from Zingiber zerumbet and Cheilocostus speciosus [171] was proved in human melanoma CHL-1 cells through induction of ROS, reduction of mitochondrial matrix potential and mitochondrial biogenesis mediated by reduced mitochondrial ATP synthesis, mitochondrial DNA levels, and mRNA expression of mitochondrial transcription factor A level, a mitochondrial biogenesis factor [138]. Activation of apoptosis through downregulation of Bcl-2 and upregulation of Bax and cytochrome c gene and protein levels, as well as activation of caspases 3, was also observed in A375 cells after treatment with zerumbone [139]. Morphological and biochemical features of apoptosis in A375 cells were also induced by carvacrol, the main constituent isolated from Coleus aromaticus and present in other EOs, such as those from Origanum ehrenbergii, Origanum syriacum and Satureja hortensis [127]. EO fractions rich in thujone, isolated from Thuja occidentalis [172], induced apoptosis in A375 cells, with minimal growth inhibitory responses when exposed to normal cells [131]. Curzerene from Eugenia uniflora activated apoptosis in SK-MEL-19 cells [43].
Induction of apoptosis, necrosis, and autophagy was observed in B16-F10 cells after treatment with citral, a key component of EOs from Cymbopogon citrates, Melissa officinalis and Verbena officinalis. Apoptosis was associated with reduction of extracellular signal-regulated kinase-1 (ERK-1) and -2 (ERK-2), AKT and nuclear factor kappa B (NF-kB), as well as, induction of oxidative stress, DNA lesions, ROS and lipid peroxidation. SK-MEL-147 and UACC-257 cells showed a lower sensitivity to citral, when compared to B16-F10 cells [133].
Data pointing toward a proapoptotic activity of thymoquinone, a constituent of the EOs from Nigella sativa and Thymus species, have been also reported in B16-F10 cells through JAK2/STAT signal transduction [121]. Isoegomaketone from Perilla frutescens trigged ROS-mediated caspase-dependent and -independent apoptosis in B16 cells. In vitro studies were supported by in vivo experiments demonstrating that oral gavage of isoegomaketone in mice subcutaneously carrying B16 melanoma inhibited tumor growth, induced apoptosis, as well as increased Bax/Bcl-2 ratio [60].
Menthol, a compound present in EOs such as peppermint and mint has been reported to exert in vitro cytotoxic effect in A375 cells and to induce morphological changes, such as cell shrinkage and ruptured membranes, indicative of apoptosis. Decrease in transient receptor potential melastatin 8 (TRPM8), at the transcript level, was also evidenced following treatment with menthol. TRPM8 is a membrane receptor involved in the regulation of calcium ion influx and melanocytic behavior, and upregulated in melanoma [173]. The authors also hypothesized that the effect of menthol on TRPM8 expression could be linked to both decrease in cell proliferation and increase in cell death [137,161]. Menthol induced cytotoxicity was also pointed out in G-361 melanoma cells through a TRPM8-dependent mechanism only when using high doses of menthol [162], thus indicating a significant difference between A375 and G-361 cells in the sensitivity to menthol.
EOs and their main components demonstrated to induce apoptosis in melanoma cells are reported in Table 3. The in vitro and in vivo melanoma models used are also listed in the Table.

3.4. Induction of Necrosis or Modulation of Autophagy

Necrosis is a non-programmed cell death that, contrary to apoptosis, does not use a highly regulated intracellular program. Necrotic cells have usually lost cell membrane integrity and release products and enzymes in the extracellular space, with consequent activation of an inflammatory response. They are taken up and internalized by macropinocytotic mechanisms [163].
Autophagy is a recycling process playing a relevant role in cell survival and maintenance. Through the analysis of LC3II protein expression, presence of dot-like formations of endogenous LC3 protein and its colocalization with the lysosome marker LAMP-1, degradation of the specific autophagy substrate p62, use of early and late autophagy inhibitors, it is possible to analyze whether a particular compound affects autophagy, inducing autophagy rather than decreasing autophagosomal turnover [174,175].
Only a few EOs have been reported to induce necrosis in melanoma cells. Among them, TTO and its major active component, terpinen-4-ol, induced necrotic cell death coupled with low level apoptotic cell death in B16 cells. Necrosis was evidenced by ultrastructural features, including cell and organelle swelling, identified by video time lapse microscopy and transmission electron microscopy [130]. Measurement of lactate dehydrogenase (LDH) release from the cytosol into the supernatant demonstrated cellular membrane damage associated with necrosis in B16-F10 cells exposed to citral. Apoptosis induction and signs of autophagy were also evident after treatment with citral [133]. EOs distilled from Salvia bracteata, Salvia rubifolia, Salvia aurea, Salvia judaica and Salvia viscosa induced apoptosis and necrosis in M14 cells [71,72].
Regarding autophagy, some EOs or their components, have been found to affect basal autophagy or to trigger autophagy induced by serum starvation and/or rapamycin in cancer cells from different origin, indicating an mTOR independent mechanism [176]. α-Thujone, D-limonene, terpinel-4-ol and β-elemene are all components of EOs reported to affect in vitro and/or in vivo melanoma growth or melanogenesis [120,131,144,152,177] and to induce autophagy, respectively, in glioblastoma [178], neuroblastoma [179], leukemic [180] and breast carcinoma [181] cells. To the best of our knowledge, no studies exist regarding the ability of these or other EO components to induce autophagy or to affect autophagic flux in melanoma cells. Generation of autophagic vacuoles in B16-F10 cells was observed after treatment with citral. The authors suggested that B16-F10 cells shifted cellular metabolism, trying to recycle damaged structures by oxidative stress under treatment with citral, but no further characterization of autophagy was provided [133].
EOs and their main components demonstrated to induce necrosis or to modulate autophagy in melanoma cells are reported in Table 3. The in vitro melanoma models used are also listed in the Table.

3.5. Inhibition of Angiogenesis and Lymphangiogenesis

Angiogenesis, the formation of new blood vessels from existing microvessels, plays a relevant role in the growth and metastasization of many tumors, including melanoma [182]. Lesions at the beginning stage do not grow in the absence of angiogenesis or inflammation [183]. Furthermore, vasculogenic mimicry (VM) and lymphangiogenesis contribute to the metastatic spread of melanoma [184]. VM and lymphangiogenesis represent, respectively, the ability of cells to form networks of vessel-like channels and the formation of lymphatic vessels from pre-existing ones. Thus, inhibition of angiogenesis, VM or lymphangiogenesis, could represent a valid strategy for melanoma prevention and treatment. Vascular endothelial growth factor (VEGF) is one of the most important angiogenic factors that, through its binding to the receptor tyrosine kinase VEGFR, and the formation of a VEGF–VEGFR complex, induces angiogenesis (VEGF-A) or lymphangiogenesis (VEGF-C, VEGF-D) [185,186].
The ability to reduce angiogenesis has been highlighted in several EOs and their components. In particular, in vitro endothelial cells and in vivo/ex vivo assays such as, rat aortic ring, matrigel plug and chick embryo chorioallantoic membrane (CAM), have been used to study the effect of EOs on the formation of new blood vessels from pre-existing ones. Figure 1A summarizes pro- and anti- angiogenic factors regulated by EOs or their components.
EO from Pistacia lentiscus (Mastic oil) has been extensively studied due to its antiangiogenic effect attributed to both its activities on in vitro endothelial cell proliferation and in vivo microvessel formation. It also inhibited VEGF release by B16 cells. Investigation of underlying mechanism by Pistacia lentiscus EOs in endothelial cells demonstrated activation of RhoA, a regulator of neovessel organization [64]. Tridax procumbens EO, when administered intraperitoneally, inhibited capillary formation in B16-F10 injected intradermally on the shaven ventral skin of C57BL/6 mice [86]. Using the same experimental model, the Plectranthus amboinicus EO ability to reduce tumor-directed blood vessel formation was also evidenced [67]. In addition, EO from Curcuma zedoaria was reported to suppress in vitro proliferation of human umbilical vein endothelial cells, sprouting vessels of aortic ring and formation of microvessels in CAM. The antiangiogenic effect was also confirmed by immunohistochemical analysis of tumors showing a reduced expression of the endothelial marker CD34 after oral administration of Curcuma zedoaria EO in C57BL/6 mice carrying B16-Bl6 melanoma [38].
Regarding EO components, several groups reported that β-elemene, one of the most active constituents of Curcuma zedoaria and Curcuma wenyujin EOs, inhibited the VEGF-induced sprouting vessel of rat aortic ring, microvessel formation of CAM, as well as CD34 and VEGF expression in C57BL/6 mice carrying B16-F10 melanoma after subretinal injection. VEGF expression in serum and lung of mice was also inhibited following treatment with β-elemene [120,152]. In vitro and in vivo studies carried out by Jung’s group highlighted the ability of dietary β-caryophyllene, a component found in many EOs (basil, black pepper, cinnamon, cannabis, lavender, rosemary, cloves, oregano), to suppress high fat diet (HDF)-induced in vitro and in vivo angiogenesis and lymphangiogenesis. In particular, dietary β-caryophyllene reduced the expression at transcriptional and protein level of hypoxia inducible factors 1α, VEGF-A, CD31 and VE-cadherin observed in the tumors of HFD-fed mice. In addition, the HFD-stimulated expression of VEGF-C, VEGF-D, VEGF-R3 and lymphatic vessel endothelial receptor was prevented by β-caryophyllene supplementation in the diet. The authors indicate the effect of β-caryophyllene in angiogenesis as one of the most important mechanisms for reduced tumor growth in β-caryophyllene-fed mice [140].
Even if a direct effect on melanoma angiogenesis has not yet been reported, several EOs or their components have been found to affect in vitro endothelial functions and/or in vivo neovascularisation. Among them, EOs from Hypericum perforatum [187] and Myristica fragrans [56] should be mentioned, together with coated-dacarbazine eugenol liposomes, for their ability to reduce proliferation and migration of endothelial-like cells [188]. Citrus lemon EO nanoemulsions have been found to decrease angiogenesis in CAM [189]. Interestingly, when analyzing the effect of the ointment prepared from Salvia officinalis EO on the healing process of an infected wound mouse model, Farahpour et al. demonstrated that, in addition to antioxidant and anti-inflammatory properties, Salvia officinalis EO accelerates the wound healing process. This process requires coordination of overlapping distinct cellular activities including angiogenesis. Promotion of the healing process by Salvia officinalis EO was attributed to enhanced angiogenesis through upregulation of VEGF and fibroblast growth factor-2 (FGF-2) expression. An increase in the number of blood vessels and fibroblasts, through cyclin-D1 pathway activation and enhanced expression of Bcl-2, was also observed [190]. A positive effect on wound healing process has also been described for other EOs such as lavender EO, TTO, Alpinia zerumbet and Chrysantemum boreale Makino EO and their use has been suggested for the treatment of wounds, burns, abscesses or diseases such as diabetes [191,192,193,194]. Thus, the antiangiogenic property or the positive effect on wound healing showed by some EOs should be considered when proposing them for treatment of cancer or other diseases.
The ability to interfere with the process of angiogenesis has also been attributed to zerumbone, perillyl alcohol and curcumol, three components of many EOs. Zerumbone, has been found to inhibit proliferation, migration, and morphogenesis, but not viability of endothelial cells, as well as the outgrowth of new blood vessels in rat aortic rings, vessel formation in the matrigel plug and CAM assays. All these effects were mediated by downregulation of phosphorylation of VEGFR2 and fibroblast growth factor receptor-1, two essential signaling pathways for angiogenesis [195,196]. Perillyl alcohol, a component of Anethum graveolens, Conyza newii and Citrus limon EOs [197], blocked the growth of new blood vessel in the in vivo CAM assay and inhibited the in vitro morphogenic differentiation of endothelial cells. Perillyl alcohol also reduced proliferation and induced both apoptosis and the expression of the antiangiogenic molecule, angiopoietin 2, in endothelial cells, indicating that it exerted its effect through both vessel regression and neovascularization suppression [198]. Curcumol, a representative component for the quality control of the EO of Curcuma wenyujin, inhibited angiogenesis by reducing PD-L1 expression in endothelial cells. In particular, addition of curcumol reduced the expression of VEGF and metalloproteinase-9 (MMP-9) and tube formation induced by PD-L1. It also cooperated with the ability of PD-L1 silencing to downregulate VEGF and MMP-9 expression and morphogenesis of endothelial cells [199].
EOs and their main components demonstrated to affect angiogenesis by using both endothelial and melanoma models are reported in Table 4. The used in vitro and in vivo models are also listed.

3.6. Alteration of In Vitro Tumor Progression-Associated Functions and Inhibition of In Vivo Tumor Growth and Metastasization

Tumor metastasization, the spread of tumor cells from the primary site to distant organs, represents the main cause of death of cancer patients, including those affected by melanoma. Thus, new therapeutic approaches, which are able to block functions associated to tumor progression, or even to prevent metastasization, represent a big turning point for cancer therapy. Several studies reported that the antimetastatic potential of EOs goes across the regulation of inflammatory cytokines and chemokines. Figure 1B,C summarizes factors regulated by EOs and responsible for tumor dissemination and tumor-promoting inflammation.
Several studies demonstrated EOs’ ability to affect in vitro tumor progression-associated functions and in vivo tumor growth and metastasization. EO from Alpinia zerumbet was shown to inhibit transforming growth factor (TGF)-β1-induced endothelial-to-mesenchymal transition in endothelial cells through regulation of Krüppel-like factor 4. Activation of endothelial-to-mesenchymal transition, a process in which endothelial cells switch from the endothelial to a mesenchymal-like phenotype, cell marker and functions, contribute to cancer progression [141].
Treatment of A375 cells with EOs from Satureja hortensis inhibited the in vitro cell migration process, while not affecting migration of normal keratinocytes and fibroblasts [77]. TTO and its main active component, terpinen-4-ol, interfered with in vitro cell migration and invasion of adriamycin-sensitive and -resistant M14 cells by inhibiting the intracellular pathway induced by the multidrug transporter p170 glycoprotein [142]. In vivo results also reported that topical TTO formulation was able to slow the growth of B16-F10 melanoma subcutaneously injected in C57BL/6J mice. The treatment was accompanied by a quick and complete disappearance of skin irritation together with recruitment of neutrophils and other immune effector cells in the treated area, while it did not induce systemic toxicity [147]. Both studies highlighted the potential of TTO in topical formulations as a promising chemopreventive candidate or as an alternative topical antitumor treatment against melanoma.
Through its component β-ursolic acid, Salvia officinalis EO, inhibited proteases implicated in the mechanisms by which tumor cells metastasize, such as serine proteases (trypsin, thrombin and urokinase) and the cysteine protease cathepsin B. In vivo inhibition of lung colonization of B16 mouse melanoma cells by intraperitoneal administration of β-ursolic acid was also highlighted [148]. After oral administration, Curcuma zedoaria EO was reported to suppress in vivo growth of B16-Bl6 tumors after subcutaneous cell injection into the left oxter of C57BL/6 mice, and their metastasization to the lung. A reduced expression of MMP-2 and MMP-9 in serum of treated mice was also evidenced after treatment with Curcuma zedoaria EO [38].
Intraperitoneal treatment with Boswellia carterii EO reduced the tumor burden in C57BL/6 mice carrying the B16-F10 model, while it did not elicit a detrimental effect on body weight. The authors also reported hepatoprotection by the EO [146]. Considering that liver injury is a frequent consequence of melanoma drug treatments [200,201], this represents an important remark. In the same experimental model, EO from Pituranthos tortuosus inhibited in vitro cell migration and invasion, focal adhesion and invadopodia formation. It also induced downregulation of kinases, and molecules involved in cell movement and migration, such as FAK, Src, ERK, p130Cas and paxillin. A decreased expression of p190RhoGAP and Grb2, which impaired cell migration and actin assembly, was also induced by the Pituranthos tortuosus EO. In vivo treatment of B16-F10 carrying mice with intraperitoneal administered Pituranthos tortuosus EO led to impaired tumor growth with no sign of abnormal behavior or adverse toxicity [66].
Zornia brasiliensis [91] and Annona vepretorum [25] EOs intraperitoneally administered in C57BL/6 mice subcutaneously carrying B16-F10 melanoma elicited antitumor activity. Importantly, microencapsulation of the Annona vepretorum EO with β-cyclodextrin, used to form inclusion complexes with EO and to improve their characteristics, further increased in vivo tumor growth inhibition with respect to free-EO to the level induced by dacarbazine [202]. While not showing any lethal effect/abnormality on mice when injected intraperitoneally, EO from Tridax procumbens elicited a reduction of tumor lung nodules of B16-F10 cells injected through the tail vein. Increased apoptosis, associated with enhanced p53 expression, was also observed after treatment. Decrease in body weight, increase in white blood cells and decrease in haemoglobin observed in untreated group, were almost normalized in the EO treated group [86]. Using the same experimental model, the same group also evidenced the ability of EO from Plectranthus amboinicus to decrease experimental metastase formation [67].
Moreover, some EO components were reported to inhibit tumor progression-associated properties and in vivo metastasization. In this regard, zerumbone [138] and curzerene from Eugenia uniflora EO [43] inhibited in vitro migration of CHL-1 and SK-MEL-19 cells, respectively.
In the Section 3.5, we reported that β-caryophyllene reduced angiogenesis and lymphangiogenesis. In addition to these effects, β-caryophyllene in tumor tissues also reduced M2 macrophages and macrophage mannose receptors. Reduction of cytokines promoting macrophage recruitment and differentiation toward M2 type, such as keratinocyte chemoattractant, monocyte chemoattractant protein-1 (MCP-1) and macrophage colony stimulating factor, were also observed. β-Caryophyllene also increased the number of apoptotic cells and the expression of apoptosis related proteins Bax and activated caspases 3. In the adipose tissues surrounding the lymphnode, β-caryophyllene reduced M2 macrophages and blocked the CCL19-CCL21/CCR7 axis, a signaling pathway important for recruitment of CCR7-expressing cancer cells or leukocytes to lymphnodes. The authors suggested the use of β-caryophyllene for people with high risk of melanoma and/or consuming a high-fat diet regimen [140].
The antimetastatic potential of thujone was demonstrated after injection of B16-F10 cells in the lateral tail vein of C57BL/6 mice. In addition to the reduction of lung nodules, thujone administration also reduced expression of MMP-2, MMP-9, ERK-1, ERK-2, and VEGF and upregulated the expression of nm-23, tissue inhibitor of metalloproteinase-1 (TIMP-1), and TIMP-2 in the lung tissues and the production of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-2 and granulocyte-monocyte colony-stimulating factor. In the same model, thujone also inhibited in vitro secretion of MMP-2 and MMP-9 and the adhesion of tumor cells to the collagen-coated plate, as well as cell invasion and migration [144].
Intraperitoneally administered β-elemene was reported to inhibit in vivo growth and metastasization of C57BL/6 mice carrying B16-F10 melanoma through downregulation of tumor promoting factors such as MMP-2, MMP-9, VEGF, urokinase-type plasminogen activator (uPA) and uPA receptor. Reduction of melanin content in lung confirmed the antimetastatic effect of β-elemene [120,152]. The ability of intravitreal administered β-elemene to block the growth of subretinal injected B16-F10 cells in C57BL/6J mice was also reported [120].
Intraperitoneal administration of limonene and perillic acid remarkably reduced the experimental metastatic tumor nodule formation of C57BL/6 mice intravenously injected with B16-F10 cells and increased the life span of animals. Limonene and perillic acid treatment also induced an increased expression in lung tissues and an enhanced serum content of sialic acid and uronic acid, two biochemical markers playing important roles in tumor growth and metastasis, including cell–cell communication and tumor cell escape from immune surveillance [150,203,204]. An antimetastatic effect by α-pinene from Schinus terebinthifolius Raddi was reported when B16-F10-Nex2 cells were injected intravenously and C57BL/6 mice treated intraperitoneally [149]. Recent findings provided important insights into the mechanism through which α-pinene induced tumor regression in melanoma models. Some authors supposed the relevance of environment in minimizing cancer growth and reported that α-pinene has no inhibitory effect on melanoma cell proliferation in vitro, but indicated activation in the hypothalamus/sympathetic nerve/leptin axis tumor growth (decreased plasma leptin concentration) and in the immune system (increased the numbers of B cells, CD4+ T cells, CD8+ T cells, and NK cells) as possible mechanisms through which exposure to a fragrant environment containing α-pinene suppressed B16 tumor growth in C57BL/6 mice [205,206].
Camphene derived from Piper cernuum EO, when injected peritumorally, exerted antitumor activity in vivo by inhibiting subcutaneous growth of B16-F10-Nex2 in C57BL/6 mice, while it did not induce toxic effects, weight loss, or behavior alterations in mice [62]. Oral administration of thymoquinone reduced experimental metastases of B16-F10 model through destabilization of the oncogene MUC4 mRNA by tristetraprolin, a RNA binding protein regulating the MUC4 transcript [151], while intraperitoneal administration inhibited the growth of the B16-F10 intracerebral model and increased the median overall survival of C57BL/6J mice. Reduction by thymoquinone of inflammatory cytokines, such as MCP-1, TGF-β1, and RANTES, as well as induction of apoptosis, were identified as possible causes of tumor inhibition [121]. Another study evidenced thymoquinone’s ability to inhibit the in vitro migration of both human (A375) and mouse (B16-F10) melanoma cells and to suppress B16-F10 metastasis in C57BL/6 mice by inhibition of NLRP3 inflammasome and NF-κB activity, thus indicating thymoquinone ability to act as a potential immunotherapeutic agent [145].
Intraperitoneal administration of eugenol in B6D2F1 mice bearing B16 melanoma, reduced tumor sizes, extended the mice median survival and reduced metastasis [135]. Myrtenal, one of the most abundant components in the Teucrium polium EO, reduced in vitro invasion and migration of both murine (B16-F0 and B16-F10) and human (SK-MEL-5) melanoma cells and metastasis induced in C57BL/6 mice bearing B16-F10 melanoma, through inhibition of the proton pump V-ATPases [143].
EOs and their main components demonstrated to affect in vitro tumor progression-associated functions and in vivo tumor growth and metastasization are reported in Table 2. The in vitro and in vivo models used are also listed in the Table.

3.7. Sensitization of Antitumor Agents

In addition to their ability to affect in vitro and in vivo tumor growth, several EO constituents have been reported to act synergistically with conventional chemotherapy and radiotherapy [207]. While some particular EO constituents, such as eugenol, geraniol, β-elemene, limonene, β-caryophyllene, have been shown to synergize with chemotherapy or radiotherapy in leukemia [208] or solid tumors [207,208], their efficacy in combination therapy for melanoma models are rare.
β-Elemene, one of the most active constituents of EOs from Curcuma zedoaria and Curcuma wenyujin, remarkably decreased A375 cell proliferation and enhanced apoptosis induced by radiation [209]. The effect of thymoquinone on the sensitivity of human 518A2 melanoma cells to doxorubicin was also analyzed. The authors demonstrated a cell line-dependent effect of thymoquinone on doxorubicin sensitivity inducing a synergistic proapoptotic effect on leukemia and multi-drug-resistant breast cancer cells, but not in 518A2 cells where the combination did not affect caspase kinetics or mitochondrial membrane potential but induced an antagonistic effect [208]. The authors indicate alteration in the apoptotic machinery of 518A2 cells [210] as a possible explanation of their response to thymoquinone/doxorubicin combination, thus indicating that further studies are needed to explore the effect of thymoquinone on doxorubicin sensitivity of melanoma cells. Surprisingly, the authors also reported a significant growth inhibitory effect of thymoquinone/doxorubicin combination in normal foreskin fibroblasts [208]. The ability of thymoquinone to further enhance the in vitro apoptotic effect of Gamma Knife irradiation has been reported in B16-F10, while it did not add any survival benefit to Gamma Knife treatment in C57BL/6J mice with intracerebral B16-F10 melanoma [121].
Liposomes loaded with dacarbazine and eugenol, and coated with hyaluronic acid in order to enable the active targeting of the CD44 receptor that is overexpressed by tumor cells [211], have been reported to inhibit proliferation of both human (SK-MEL-28) and mouse (B16-F10) melanoma cells and to induce both apoptosis and necrosis. The authors suggested the use of this liposome formulation to reduce dacarbazine dose during chemotherapy and consequently toxicity on normal cells [188].
EOs and their main components demonstrated to sensitize antitumor agents are reported in Table 5. The used melanoma models are also listed in the Table.

3.8. Chemopreventive Activity

Exposure to artificial or natural UV rays are among the major risks for the development of both non-melanoma and melanoma skin cancer. Other risk factors for melanoma development include the number of naevi and dysplastic naevi, phenotype characteristics and family history [227,228,229]. Chemopreventive agents prevent formation of cancer by multiple mechanisms, interfering with the initiation, promotion, or progression steps. Although mouse models that spontaneously develop melanoma are extremely rare, different chemically- or genetically-induced melanoma mouse models have been developed to evaluate the chemopreventive potential of compounds. Among them, the two-stage skin carcinogenesis 7,12-dimethylbenz[a]anthracene (DMBA)/12-O-tetradecanoylphobol-13-acetate (TPA) model, that fully recapitulates the multistage tumorigenesis of the skin, is the most commonly used to evaluate the chemopreventive potential of several compounds, including EOs. In particular, tumor initiation in BALB/c, CD1, ICR, SENCAR and Swiss albino models can be obtained with a single topical application of DMBA, whereas tumor promotion can be triggered by repeated applications of TPA [230]. To decrease the latency of melanoma appearance, these compounds are often administered in combination with other agents such as UV rays, or used in genetically engineered mice that harbor activating mutations in BRAF and NRAS, two oncogenes frequently mutated in human melanoma [230]. However, a drawback of DMBA/TPA models exists in the evidence that they induce the development of papillomas and small naevi more frequently than that of melanoma, making their use more accurate in the study of skin cancer on the whole, rather than of cutaneous melanoma. Thus, the chemopreventive effect of EOs or their constituents in skin cancer is discussed in this paragraph.
The chemopreventive potential of Santalum album EO and its major constituent α-santalol has been demonstrated in DMBA/TPA-induced skin tumors in CD1 mice: topical application of Santalum album EO reduced tumor incidence and multiplicity in animals. Furthermore, the chemopreventive potential of α-santalol was similar to that of Santalum album in DMBA/TPA-induced skin tumors in CD1 and SENCAR mice [76]. Topical application of Salvia libanotica EO (sage oil) delayed tumor appearance and inhibited tumor incidence and yield in DMBA/TPA in BALB/c mice, whereas decreasing EO concentration reduced only tumor yield [73]. By using the same DMBA/TPA model, Mentha aquatica var. Kenting Water Mint EO has been identified as a chemopreventive agent against cutaneous side effects induced by vemurafenib, a BRAF inhibitor used for treatment of melanoma patients carrying the BRAFV600 mutation. The results of this study evidenced that Mentha aquatica EO induces G2/M cell-cycle arrest and apoptosis, reduces cell viability, colony formation and the invasive and migratory functions of the mouse keratinocyte bearing HRASQ61L mutation. This mutation is found in melanoma patients with a higher probability of developing keratoacanthomas and squamous cell carcinoma after treatment with vemurafenib. In vivo treatment with Mentha aquatica EO decreased the formation of cutaneous papilloma and the expression of keratin14 and COX-2 observed in FVB/NJ mice exposed to DMBA/TPA and treated with vemurafenib [54].
Several studies investigated the chemopreventive potential of some components of EOs. Among them, Pal and colleagues demonstrated that oral administration of eugenol produced a reduction in the incidence and size of skin cancer in Swiss albino mice treated with DMBA and croton oil, along with an increase in the overall survival of mice. The carcinogenic process prevention by eugenol was due to reduction in cell proliferation and induction of apoptosis through the downregulation of bcl-2, c-Myc and H-ras expression along with the upregulation of active caspase 3, Bax and p53 in the skin lesions [213]. In the same year, Kaur and colleagues confirmed the inhibitory effect of eugenol in DMBA/TPA-induced skin cancer in Swiss albino mice. In particular, eugenol treatment delayed tumor onset, incidence and multiplicity when applied during the initiation, as well as during the promotion phase. The chemopreventive effect of eugenol was due to the induction of apoptosis, prevention of oxidative stress, decrease in ornitine decarboxylase activity, attenuation of tumor inflammation caused by reduction in NF-kB pathway, COX-2 and iNOS in tumor samples and in pro-inflammatory cytokines in mice serum (e.g., IL-6, TNF-α and PGE2) [212]. However, both groups demonstrated that mice treated with DMBA and TPA developed squamous cell carcinoma of the skin in their experimental system [212,213].
Perillyl alcohol topical treatment showed the ability to delay development and incidence of DMBA-induced melanoma in transgenic mice harboring a mutated HaRas gene driven by the tyrosinase promoter (TPras). Moreover, perillyl alcohol treatment reduced the UV-induced ROS, the levels of Ras protein and inhibited the activation of MAPK and AKT pathways in a cell line derived from DMBA-induced melanoma of TPras mice [217]. A few years later, another two studies by Chaudhary and colleagues confirmed the chemopreventive effect of perillyl alcohol and its precursor D-limonene in DMBA/TPA-induced skin cancer in Swiss albino mice. In particular, topical application of perillyl alcohol or D-limonene elicited a significant reduction in tumor incidence and tumor burden with extension of the latency period of tumor development. In agreement with Prevatt et al., perillyl alcohol or D-limonene treatment effectively reduced the skin tumorigenesis by inducing apoptosis, reducing ROS production, inflammation, Ras/Raf/ERK1/2 pathway and Bcl-2 expression along with an increase in Bax levels [177,218]. Moreover, a phase 2a clinical trial showed a modest reduction in nuclear chromatin abnormality caused by twice-daily topical application of perillyl alcohol in participants with sun-damaged skin [231]. A phase 1 clinical trial demonstrated that daily topical application of perillyl alchol cream for 30 days was well tolerated in participants with normal appearing skin [232].
The chemopreventive potential of farnesol, geraniol and menthol was analyzed by several investigators using DMBA/TPA-promoted skin tumorigenesis in Swiss albino or in ICR, a strain of albino mice. These monoterpenes reduced tumor incidence and tumor volume with an extension of latency period during the promotion phase. The mechanism of action of these three components is the same as is described for perillyl alchol, D-limonene and geraniol. In particular, their chemopreventive effects occurring through alteration of phase II detoxification agents [215], reduction in inflammation and ROS production, suppression of the Ras/Raf/ERK1/2, p38 and NF-kB signaling pathways, reduction in Bcl-2 and induction of Bax expression [177,214].
EOs and their main components demonstrated to show chemopreventive potential are reported in Table 5. The used melanoma models are also listed in the Table.

3.9. Antioxidant Effect

Oxidative stress producing ROS, such as superoxide, hydrogen peroxide, and hydroxyl radical, are associated with many cancer types, including non-melanoma and melanoma skin cancer, where ROS are reported to cause free radical damage to the skin [233]. Oxidative stress is involved in all stages of melanoma development, and modulates intracellular pathways related to cellular proliferation and death. Several factors, including inadequate lifestyle and/or diet or UV-irradiation lead to the formation of ROS, which are often associated with alterations in the DNA, proteins and lipids, and consequent induction of cellular aging, mutagenicity and carcinogenicity. By chelating oxidation-catalytic metals or by scavenging free radicals and ROS, natural enzymatic and non-enzymatic antioxidant defense counteracts the dangerous effects of free radicals and other oxidants. Hence the relevance of antioxidant compounds to decrease oxidative stress or damage. In this context, many plant-derived components, including EOs, are reported as a useful antioxidant source able to remove excessive free radicals and to prevent free radical-induced damage.
To evaluate the antioxidant activity of a compound of interest, several test procedures should be carried out including cell-free methods, such as 2,2′-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid (ABTS) and 1,1-diphenyl-2-picrylhydrazyl (DPPH) that are the most popular and commonly employed. Other cell-free assays include hydrogen peroxide, nitric oxide, peroxynitrite radical, superoxide radical, hydroxyl radical scavenging activity, metal-ion chelating assay, lipid peroxidation and the xanthine oxidase method. Analysis of oxidation status performed in in vitro cell systems or in vivo animal models (blood or tissues) include glutathione, glutathione peroxidase, glutathione-S-transferase, superoxide dismutase and catalase activity [234].
When performing a search on PubMed, about 2800 results in the last 20 years are shown for “essential oil” and “antioxidant”. Analysis of the PubMed results identified only few research articles reporting studies on the evaluation of EO antioxidant activity in melanoma models. Among these studies, the antioxidant activity of Melaleuca quinquenervia EO and its constituents 1,8 cineole, α-pinene and α-terpineol [53,235], Vetiveria zizanioides EO and its most abundant compound, cedr-8-en-13-ol [87], Cinnamomum cassia EO and its major component cinnamaldehyde [32] and Achillea Millefolium EO with its main component linalyl acetate [23], was reflected in the recovered activities of glutathione peroxidase, superoxide dismutase, and catalase in α-MSH stimulated B16 cells. Protection from cell oxidative damage by Lavandula Angustifolia EO [49] and by high concentrations of Eucalyptus camaldulensis EO [41] was also observed in B16-F10 cells. Worthy of mention, is the in vivo study demonstrating that Wedelia chinensis EO (Osbeck oil) showed in vivo antioxidant activity by scavenging free radicals (lipid peroxidation and nitric oxide) and enhancing the level of endogenous antioxidants (catalase, superoxide dismutase, glutathione peroxidase, glutathione) in lung, liver and blood tissues of B16-F10-carrying C57BL/6 mice [90]. Protective effects against oxidative stress and apoptosis also in bovine aortic endothelial cells was induced by Crocus Sativus EO (Saffron oil) through SAPK/JNK and ERK1/2 signaling pathways, supporting its use in endothelial dysfunctions [236].
Table 6 shows studies in which the antioxidant effects of EOs and their components have been analyzed using cell-free assays or melanoma models.

3.10. Antimelanogenic Activity

Several EOs and their components have been reported to suppress melanogenesis, the secretion of melanin by epidermal melanocytes, and their use in skin-whitening materials has been suggested. Melanin is synthesized as a normal defense to diverse stimuli. An excessive production of melanin can be associated with hyperpigmentation and melanoma. Melanogenesis is stimulated by the melanogenic factors α-Melanocyte Stimulating Hormone (α-MSH) and Stem Cell Factor. Through the α-MSH/MC1R binding and the cyclic adenosine monophosphate-protein kinase A-cAMP response element binding protein (cAMP-PKA-CREB) signaling pathway, α-MSH stimulation activates microphthalmia-associated transcription factor (MITF), that in turn increases the expression of its target melanogenic genes, such as tyrosinase, tyrosinase-related protein-1 (TRP-1) and -2 (TRP-2). Furthermore, ERK1/2 are involved in the regulation of MITF expression through their ability to promote MITF phosphorylation and degradation [240,241]. α-MSH-induced melanogenesis is associated with ROS generation [242]. This is the reason why oxidation and melanogenesis are strictly interconnected, and EOs-induced decrease in melanin production is often attributed to EOs antioxidant property [243,244]. Figure 2 shows the effect of EOs and their components in melanogenesis and oxidation.
Several EOs, including those from Pomelo Peel, Glechoma heredacea, Eucalyptus camaldulensis, Melaleuca quinquenervia, Chrysanthemum boreale Makino, Alpinia zerumbert, Achillea millefolium, Cinnamomum cassia, Artemisia argyi, Cryptomeria japonica and Vetiveria zizanioides showed antimelanogenic activity, alongside their antioxidant properties. For these properties, they can be widely used in food, pharmaceutical, as well as in cosmetic industries as natural compounds.
The antimelanogenic properties of EO from Pomelo Peel have demonstrated in B16 cells where it blocked the synthesis pathway of melanin through the decrease of expression and activity of intracellular tyrosinase, without affecting cell viability and morphology [68]. Moreover, EOs from Cryptomeria japonica (Yakushima native cedar), Syzygium aromaticum (Clove oil) and Cinnamomum zeylanicum demonstrated antimelanogenesis activity in the same experimental model [33,81,245]. The last two EOs contained a high level of eugenol, which was also found to inhibit tyrosinase and melanogenesis in the same model [81]. An antimelanogenic characteristic of Eucalyptus camaldulensis EO was evidenced by its ability to inhibit the activity of mushroom tyrosinase, often used as the target enzyme in screening potential inhibitors of melanogenesis, and to decrease tyrosinase activity and intracellular melanin content of B16-F10 cells. It also reduced the expression level of MC1R, tyrosinase, TRP-1 and TRP-2, and of MAPK, JNK, PKA and ERK signaling pathways, thus suggesting the involvement of these pathways in Eucalyptus camaldulensis EO-mediated inhibition of melanogenesis [41]. In addition, EOs from Psiadia terebinthina, Citrus grandis, Citrus hystrix, and Citrus reticulata inhibited both intracellular and extracellular melanin production when tested against the B16-F10 model [219]. EOs from Glechoma hederacea [44], Vetiveria zizanioides [87], Cinnamomum cassia and its main component cinnamaldehyde [32], and Melaleuca quinquenervia and its main constituents, 1,8-cineole, α-pinene, and α-terpineol [53], showed potent anti-tyrosinase and antimelanogenic activities in α-MSH-stimulated B16 cells.
Other EOs showing antimelanogenic properties, in terms of reduction of melanin synthesis, intracellular tyrosinase activity and MITF expression, when tested in B16-F10 cells, include those extracted from Vitex negundo [88], Origanum syriacum and Origanum ehrenbergii and their main component, carvacrol [59], Artemisia argyi [28], Chrysanthemum boreale Makino [31], two varieties of Alpinia Zerumbet EOs, shima and tairin oils [24], Achillea millefolium and its component lynalyl acetate blocking melanin production through the regulation of the JNK and ERK signaling pathways [23], Mentha aquatica (lime mint oil) and one of its main compounds, β-caryophyllene, [221], Vitex Trifolia and its main component abietatriene [89].
Regarding the antimelanogenic effect of EO components, the study of Lin JH’s group reported the ability of zerumbrone to decrease melanin accumulation in B16-F10 cells, and to suppress the expression of MITF and its target genes, TYRP1 and TYRP2, after MSH stimulation with a mechanism involving ERK1/2, but not the PKA-CREB signaling pathway [226]. Antimelanogenic activity in the same melanoma model through ubiquitination and proteasomal degradation of MITF, in a ROS/ERK-dependent way, was also reported for phytol [223]. Valencene one of the major constituents of Ocotea dispersa EO decreased melanin content after UVB irradiation in B16-F10 cells [225]. A zebrafish embryo experimental model was also employed to demonstrate antimelanogenic activity of some EOs or their components, such as that from Dalbergia pinnata, which reduced tyrosinase activity and body pigmentation in zebrafish embryos [39], and thymoquinone which blocked melanogenesis also in B16-F10 mouse melanoma cells through the inhibition of the glycogen synthase kinase 3β (GSK3β)/β-catenin pathway [224]. Contrasting results were reported by the Zaidi KU group using the same experimental model and the same assays (tyrosinase activity and melanin production). The authors demonstrated that thymoquinone plays a protective role for melanogenesis [246].
Table 5 shows studies demonstrating the antimelanogenic activity of EOs and their active components in melanoma models. The used melanoma models are also listed.

4. Clinical Use of EOs for Cancer Patients

The side effects experienced by patients diagnosed with cancer or undergoing radio- or chemotherapy can be debilitating and can be challenging in the management of the disease. In the last few years, EOs have gained popularity as supportive therapies for cancer patients [247]. Some EOs have been reported to improve the quality of life of patients affected by cancer and showed efficacy in several side effects, such as chemotherapy-induced nausea and vomiting, mucositis, ulcer of skin, distress, depression and anxiety.
A descriptive systematic review, carried out by Boehm K et al. in 2012, highlighted short-term effects of aromatherapy, the use of EOs, on depression, anxiety, and overall wellbeing. Minimal adverse effects were reported for EO use and potential risks, including ingesting large amounts, local skin irritation, allergic contact dermatitis and phototoxicity [248]. A small feasibility study performed to evaluate the effects of mouthwash with EOs from Leptospermum scoparium and Kunzea ericoides, on mucositis of the oropharyngeal area induced by radiation during treatment for head and neck carcinoma, provided a positive effect on the development of radiation induced mucositis [249]. Topical application of the Boswellia carterii EO demonstrated its efficacy as supportive therapy for cancer-related fatigue in a case study [250]. Inhalation of Citrus aurantium EO exhibited an anxiolytic effect and reduced the symptoms associated with anxiety in patients with chronic myeloid leukaemia [251]. A cool damp washcloth with Mentha x piperita EO was reported to be effective in decreasing the intensity of nausea experienced by patients receiving chemotherapy [252]. Antiemetic activity of volatile oil from Mentha spicata and Mentha piperita has been also reported in chemotherapy-treated patients [253]. A recent randomized controlled trial with 120 patients evidenced the efficacy of aromatherapy with lavender and peppermint EOs in improving the sleep quality of cancer patients [254]. Linalool, linalyl acetate and menthol present in lavender or peppermint EOs [255,256] can be responsible of EOs effect on sleep due to their sedative effects. A previously performed study by M. Lisa Blackburn showed the positive effect of aromatherapy with lavender, peppermint and chamomile on insomnia and other common symptoms among 50 patients with acute leukaemia [257].
Some studies also reported the lack of effect of aromatherapy in improving sleep quality in cancer patients or in reducing chemotherapy side effects. In this regard, while non-toxic, non-invasive and well received and tolerated, the inhaled Zingiber officinale EO was not an effective complementary therapy for chemotherapy-induced nausea and vomiting and health-related quality of life neither in children with cancer [258] nor in women with breast cancer [259]. While not showing any harm or adverse events, the study by Sasano’s group evidenced no effects of aromatherapy on quality of life, sleep quality, and vital sign during perioperative periods of breast cancer patients [260]. Meta-analysis of three randomized controlled trials including a total of 278 participants did not show any clinical effect of aromatherapy massage on reducing pain in cancer patients [261]. Inhalation of Lavandula angustifolia, Citrus bergamia, Cedrus atlantica EOs administered during radiotherapy did not reduce anxiety [262]. Sample size, duration of intervention, tools used for measuring the different symptoms, use of different chemotherapies and cancer stage could account for the differences observed in the different studies.
At present, several EOs are used in clinical trials to evaluate their efficacy for symptom management in patients during cancer therapy. In order to evaluate the effect of inhaled EOs on common quality of life issues during chemotherapy, targeted therapy, and/or immunotherapy administered intravenously, a single blind, randomized controlled trial study was completed few months ago. In particular, the effect of inhalated Zingiber officinale, German chamomile, or Citrus bergamia EOs on nausea, anxiety, loss of appetite, and fatigue has been evaluated with no available information yet about the results (NCT03858855). Two clinical trials are currently open and actively recruiting patients undergoing chemotherapy to assess the perceived effectiveness to relieve symptoms of nausea or vomiting and/or anxiety, by using peppermint and lavender EOs (NCT02163369, NCT03449511). A clinical trial is actively recruiting breast cancer patients in order to test the hypothesis that an EOs blend composed of Curcuma longa, Piper nigrum, Pelargonium asperum, Zingiber officinale, Mentha x piperita, and Rosmarinus officinalis, could reduce symptoms of chemotherapy induced peripheral neuropathy, a painful, debilitating consequence of cancer treatment considered the most adverse of non-hematologic events (NCT03449303). A randomized, controlled, single blind and longitudinal study is recruiting women with breast cancer undergoing chemotherapy in order to evaluate the impact of a hedonic aroma (inhalation) on the clinical, emotional and neurocognitive variables that contribute to reduce the side effects of chemotherapy and promote quality of life (NCT03585218). A not yet recruiting clinical trial aimed at evaluating (1) the ability of peppermint and lavender aromatherapy (sniff) to promote, respectively, relief of nausea or anxiety, in an outpatient oncology setting (NCT04449315); (2) the effects of inhaled peppermint and ginger EOs, or pure vanilla extract on chemotherapy induced nausea and vomiting in men and women with breast cancer (NCT04478630) is going to start in August/September of the current year 2020.

5. Conclusions

Natural products have always played a pivotal role in drug discovery and in the development of many potent anticancer agents. It is, thus, desirable to continue the efforts aiming at identifying new antitumor compounds from natural sources. Based on the studies mentioned in this review, different EOs and some of their constituents appear to be suitable as a part of effective melanoma prevention, prevention of metastatic melanoma, or as complementary therapies to supplement patient care. Preclinical data also indicate the possibility of using some EO components as adjuvant agents to reduce the toxicity of drugs used in cancer prevention or therapy, such as statin [263]. Pharmacokinetic studies are needed to validate the safety and efficacy of EOs and their bioactive compounds. In fact, even if several papers indicated EOs and their components as safe and not toxic [258,259,260], hepatotoxicity described for monoterpenes and sesquiterpenes, major components of many EOs, should be also considered and should deserve more attention [264]. Further studies on terpene metabolism and toxicity need to be performed to avoid the risk of eventual liver injury. In addition, the relevance of the BRAF status of melanoma cells in response to EOs is worthy of further investigation to shed light on this issue. In fact, to the best of our knowledge, no studies have evaluated whether the antitumor effect of EOs is related with BRAF status.
Since EOs composition is affected by several factors, including geographical position and agricultural practices, an important issue to be considered is the standardization of their composition. In this context, multidisciplinary applications, including machine learning, can constitute a possible tool able to predict the bioactivity of complex mixtures and to design EOs characterized by high antineoplastic efficacy and low toxicity [93,96,265].
The studies presented in this review hold promise for further analysis of EOs as new anticancer drugs and as a source of potential anticancer supplement against melanoma. Further investigations in this area are certainly necessary, desirable, and warranted to validate the results, to ascertain the therapeutic spectrum of biological studies and to determine the clinical efficacy and safety of EOs on patients affected by melanoma.

Author Contributions

Conceptualization, M.D.M., R.R. and D.D.B.; Figures and Tables preparation: M.D.M.; Writing—original draft preparation, M.D.M. and D.D.B.; Analysis of essential oils composition, S.G. and R.R. All authors have read and agreed to the published version of the manuscript.

Funding

M.D.M. is recipient of a fellowship from Italian Foundation for Cancer Research. The manuscript was supported by grants from Italian Association for Cancer Research (DDB, IG 18560); IRCCS Regina Elena National Cancer Institute (MDM, Ricerca Corrente 2018–2019); Progetti di Rilevante Interesse Nazionale 2017 (RR, prot. 2017JL8SRX); Ateneo Sapienza 2019 (RR, prot. RM11916B8876093E) and Ateneo Sapienza 2018 (RR, prot. RM118164361B425B).

Acknowledgments

This review is dedicated to the memory of our marvelous and joyful colleague and friend Marianna Desideri, who enthusiastically worked on our project on essential oils before she passed away. We thank Adele Petricca for preparation of the manuscript and Virginia Filacchione for English revision.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tímár, J.; Vizkeleti, L.; Doma, V.; Barbai, T.; Rásó, E. Genetic progression of malignant melanoma. Cancer Metastasis Rev. 2016, 35, 93–107. [Google Scholar] [CrossRef] [PubMed]
  2. Nogrady, B. Game-changing class of immunotherapy drugs lengthens melanoma survival rates. Nature 2020, 580, S14–S16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ambrosi, L.; Khan, S.; Carvajal, R.D.; Yang, J. Novel Targets for the Treatment of Melanoma. Curr. Oncol. Rep. 2019, 21, 97. [Google Scholar] [CrossRef] [PubMed]
  4. Subbiah, V.; Baik, C.; Kirkwood, J.M. Clinical Development of BRAF plus MEK Inhibitor Combinations. Trends Cancer 2020. [Google Scholar] [CrossRef] [PubMed]
  5. Leonardi, G.C.; Candido, S.; Falzone, L.; Spandidos, D.A.; Libra, M. Cutaneous melanoma and the immunotherapy revolution (Review). Int. J. Oncol. 2020. [Google Scholar] [CrossRef]
  6. Shin, M.H.; Kim, J.; Lim, S.A.; Kim, J.; Lee, K.M. Current Insights into Combination Therapies with MAPK Inhibitors and Immune Checkpoint Blockade. Int. J. Mol. Sci. 2020, 21, 2531. [Google Scholar] [CrossRef] [Green Version]
  7. Becco, P.; Gallo, S.; Poletto, S.; Frascione, M.P.M.; Crotto, L.; Zaccagna, A.; Paruzzo, L.; Caravelli, D.; Carnevale-Schianca, F.; Aglietta, M. Melanoma Brain Metastases in the Era of Target Therapies: An Overview. Cancers 2020, 12, 1640. [Google Scholar] [CrossRef]
  8. Ishizuka, J.J.; Manguso, R.T.; Cheruiyot, C.K.; Bi, K.; Panda, A.; Iracheta-Vellve, A.; Miller, B.C.; Du, P.P.; Yates, K.B.; Dubrot, J.; et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 2019, 565, 43–48. [Google Scholar] [CrossRef]
  9. Mangan, B.L.; McAlister, R.K.; Balko, J.M.; Johnson, D.B.; Moslehi, J.J.; Gibson, A.; Phillips, E.J. Evolving Insights into the Mechanisms of Toxicity Associated with Immune Checkpoint Inhibitor Therapy. Br. J. Clin. Pharmacol. 2020. [Google Scholar] [CrossRef]
  10. Lu, H.; Liu, S.; Zhang, G.; Bin, W.; Zhu, Y.; Frederick, D.T.; Hu, Y.; Zhong, W.; Randell, S.; Sadek, N.; et al. PAK signalling drives acquired drug resistance to MAPK inhibitors in BRAF-mutant melanomas. Nature 2017, 550, 133–136. [Google Scholar] [CrossRef]
  11. Cheung, M.K.; Yue, G.G.L.; Chiu, P.W.Y.; Lau, C.B.S. A Review of the Effects of Natural Compounds, Medicinal Plants, and Mushrooms on the Gut Microbiota in Colitis and Cancer. Front. Pharmacol. 2020, 11, 744. [Google Scholar] [CrossRef] [PubMed]
  12. Pradhan, D.; Biswasroy, P.; Sahu, A.; Sahu, D.K.; Ghosh, G.; Rath, G. Recent advances in herbal nanomedicines for cancer treatment. Curr. Mol. Pharmacol. 2020. [Google Scholar] [CrossRef] [PubMed]
  13. Burnham, J.F. Scopus database: A review. Biomed. Digit. Libr. 2006, 3, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zuzarte, M.; Salgueiro, L. Essential Oils Chemistry. In Bioactive Essential Oils and Cancer; de Sousa, D.o.P., Ed.; Springer International Publishing: Cham, Switzerland, 2015; pp. 19–61. [Google Scholar] [CrossRef]
  15. Rehman, R.; Hanif, M.A.; Mushtaq, Z.; Al-Sadi, A.M. Biosynthesis of essential oils in aromatic plants: A review. Food Rev. Int. 2016, 32, 117–160. [Google Scholar] [CrossRef]
  16. 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. [Google Scholar] [CrossRef]
  17. De Cássia Da Silveira e Sá, R.; Andrade, L.N.; De Sousa, D.P. Sesquiterpenes from Essential Oils and Anti-Inflammatory Activity. Nat. Prod. Commun. 2015, 10. [Google Scholar] [CrossRef] [Green Version]
  18. Dhifi, W.; Bellili, S.; Jazi, S.; Bahloul, N.; Mnif, W. Essential Oils’ Chemical Characterization and Investigation of Some Biological Activities: A Critical Review. Medicines 2016, 3, 25. [Google Scholar] [CrossRef] [Green Version]
  19. Tongnuanchan, P.; Benjakul, S. Essential Oils: Extraction, Bioactivities, and Their Uses for Food Preservation. J. Food Sci. 2014, 79, R1231–R1249. [Google Scholar] [CrossRef]
  20. Masango, P. Cleaner production of essential oils by steam distillation. J. Clean. Prod. 2005, 13, 833–839. [Google Scholar] [CrossRef]
  21. Kováts, E. Gas-chromatographische Charakterisierung organischer Verbindungen. Teil 1: Retentionsindices aliphatischer Halogenide, Alkohole, Aldehyde und Ketone. Helv. Chim. Acta 1958, 41, 1915–1932. [Google Scholar] [CrossRef]
  22. Zellner, B.D.; Bicchi, C.; Dugo, P.; Rubiolo, P.; Dugo, G.; Mondello, L. Linear retention indices in gas chromatographic analysis: A review. Flavour Frag. J. 2008, 23, 297–314. [Google Scholar] [CrossRef]
  23. Peng, H.Y.; Lin, C.C.; Wang, H.Y.; Shih, Y.; Chou, S.T. The melanogenesis alteration effects of Achillea millefolium L. Essential oil and linalyl acetate: Involvement of oxidative stress and the JNK and ERK signaling pathways in melanoma cells. PLoS ONE 2014, 9, e95186. [Google Scholar] [CrossRef] [PubMed]
  24. Tu, P.T.; Tawata, S. Anti-Oxidant, Anti-Aging, and Anti-Melanogenic Properties of the Essential Oils from Two Varieties of Alpinia zerumbet. Molecules 2015, 20, 16723–16740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bomfim, L.M.; Menezes, L.R.A.; Rodrigues, A.C.B.C.; Dias, R.B.; Gurgel Rocha, C.A.; Soares, M.B.P.; Neto, A.F.S.; Nascimento, M.P.; Campos, A.F.; Silva, L.C.R.C.E.; et al. Antitumour Activity of the Microencapsulation of Annona vepretorum Essential Oil. Basic Clin. Pharmacol. Toxicol. 2016, 118, 208–213. [Google Scholar] [CrossRef]
  26. Conforti, F.; Menichini, F.; Formisano, C.; Rigano, D.; Senatore, F.; Bruno, M.; Rosselli, S.; Çelik, S. Anthemis wiedemanniana essential oil prevents LPS-induced production of NO in RAW 264.7 macrophages and exerts antiproliferative and antibacterial activities invitro. Nat. Prod. Res. 2012, 26, 1594–1601. [Google Scholar] [CrossRef]
  27. Zhao, J.; Zheng, X.; Newman, R.A.; Zhong, Y.; Liu, Z.; Nan, P. Chemical composition and bioactivity of the essential oil of Artemisia anomala from China. J. Essent. Oil Res. 2013, 25, 520–525. [Google Scholar] [CrossRef]
  28. Huang, H.C.; Wang, H.F.; Yih, K.H.; Chang, L.Z.; Chang, T.M. Dual bioactivities of essential oil extracted from the leaves of Artemisia argyi as an antimelanogenic versus antioxidant agent and chemical composition analysis by GC/MS. Int. J. Mol. Sci. 2012, 13, 14679–14697. [Google Scholar] [CrossRef] [Green Version]
  29. Rodriguez, S.A.; Murray, A.P. Antioxidant activity and chemical composition of essential oil from Atriplex undulata. Nat. Prod. Commun. 2010, 5, 1841–1844. [Google Scholar] [CrossRef] [Green Version]
  30. Salvador, M.J.; de Carvalho, J.E.; Wisniewski, A., Jr.; Kassuya, C.A.L.; Santos, E.P.; Riva, D.; Stefanello, M.E.A. Chemical composition and cytotoxic activity of the essential oil from the leaves of Casearia lasiophylla. Rev. Bras. Farmacogn. 2011, 21, 864–868. [Google Scholar] [CrossRef] [Green Version]
  31. Kim, D.Y.; Won, K.J.; Hwang, D.I.; Park, S.M.; Kim, B.; Lee, H.M. Chemical Composition, Antioxidant and Anti-melanogenic Activities of Essential Oils from Chrysanthemum boreale Makino at Different Harvesting Stages. Chem. Biodivers. 2018, 15, e1700506. [Google Scholar] [CrossRef]
  32. Chou, S.T.; Chang, W.L.; Chang, C.T.; Hsu, S.L.; Lin, Y.C.; Shih, Y. Cinnamomum cassia essential oil inhibits α-MSH-induced melanin production and oxidative stress in murine B16 melanoma cells. Int. J. Mol. Sci. 2013, 14, 19186–19201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Fiocco, D.; Arciuli, M.; Arena, M.P.; Benvenuti, S.; Gallone, A. Chemical composition and the anti-melanogenic potential of different essential oils. Flavour Frag. J. 2016, 31, 255–261. [Google Scholar] [CrossRef]
  34. Menichini, F.; Tundis, R.; Loizzo, M.R.; Bonesi, M.; Provenzano, E.; Cindio, B.D.; Menichini, F. In vitro photo-induced cytotoxic activity of Citrus bergamia and C. medica L. cv. Diamante peel essential oils and identified active coumarins. Pharm. Biol. 2010, 48, 1059–1065. [Google Scholar] [CrossRef] [PubMed]
  35. Hajlaoui, H.; Mighri, H.; Noumi, E.; Snoussi, M.; Trabelsi, N.; Ksouri, R.; Bakhrouf, A. Chemical composition and biological activities of Tunisian Cuminum cyminum L. essential oil: A high effectiveness against Vibrio spp. strains. Food Chem. Toxicol. 2010, 48, 2186–2192. [Google Scholar] [CrossRef] [PubMed]
  36. Xiang, H.; Zhang, L.; Yang, Z.; Chen, F.; Zheng, X.; Liu, X. Chemical compositions, antioxidative, antimicrobial, anti-inflammatory and antitumor activities of Curcuma aromatica Salisb. essential oils. Ind. Crop. Prod. 2017, 108, 6–16. [Google Scholar] [CrossRef]
  37. Zhang, L.; Yang, Z.; Huang, Z.; Zhao, M.; Li, P.; Zhou, W.; Zhang, K.; Zheng, X.; Lin, L.; Tang, J.; et al. Variation in Essential Oil and Bioactive Compounds of Curcuma kwangsiensis Collected from Natural Habitats. Chem. Biodivers. 2017, 14, e1700020. [Google Scholar] [CrossRef]
  38. Chen, W.; Lu, Y.; Gao, M.; Wu, J.; Wang, A.; Shi, R. Anti-angiogenesis effect of essential oil from Curcuma zedoaria in vitro and in vivo. J. Ethnopharmacol. 2011, 133, 220–226. [Google Scholar] [CrossRef]
  39. Zhou, W.; He, Y.; Lei, X.; Liao, L.; Fu, T.; Yuan, Y.; Huang, X.; Zou, L.; Liu, Y.; Ruan, R.; et al. Chemical composition and evaluation of antioxidant activities, antimicrobial, and anti-melanogenesis effect of the essential oils extracted from Dalbergia pinnata (Lour.) Prain. J. Ethnopharmacol. 2020, 254, 112731. [Google Scholar] [CrossRef]
  40. Cianfaglione, K.; Blomme, E.E.; Quassinti, L.; Bramucci, M.; Lupidi, G.; Dall’Acqua, S.; Maggi, F. Cytotoxic Essential Oils from Eryngium campestre and Eryngium amethystinum (Apiaceae) Growing in Central Italy. Chem. Biodivers. 2017, 14, e1700096. [Google Scholar] [CrossRef]
  41. Huang, H.C.; Ho, Y.C.; Lim, J.M.; Chang, T.Y.; Ho, C.L.; Chang, T.M. Investigation of the anti-melanogenic and antioxidant characteristics of Eucalyptus camaldulensis flower essential oil and determination of its chemical composition. Int. J. Mol. Sci. 2015, 16, 10470–10490. [Google Scholar] [CrossRef] [Green Version]
  42. Aranha, E.S.P.; de Azevedo, S.G.; dos Reis, G.G.; Silva Lima, E.; Machado, M.B.; de Vasconcellos, M.C. Essential oils from Eugenia spp.: In vitro antiproliferative potential with inhibitory action of metalloproteinases. Ind. Crop. Prod. 2019, 141, 111736. [Google Scholar] [CrossRef]
  43. Figueiredo, P.L.B.; Pinto, L.C.; da Costa, J.S.; da Silva, A.R.C.; Mourão, R.H.V.; Montenegro, R.C.; da Silva, J.K.R.; Maia, J.G.S. Composition, antioxidant capacity and cytotoxic activity of Eugenia uniflora L. chemotype-oils from the Amazon. J. Ethnopharmacol. 2019, 232, 30–38. [Google Scholar] [CrossRef] [PubMed]
  44. Chou, S.T.; Lai, C.C.; Lai, C.P.; Chao, W.W. Chemical composition, antioxidant, anti-melanogenic and anti-inflammatory activities of Glechoma hederacea (Lamiaceae) essential oil. Ind. Crop. Prod. 2018, 122, 675–685. [Google Scholar] [CrossRef]
  45. Ornano, L.; Venditti, A.; Sanna, C.; Ballero, M.; Maggi, F.; Lupidi, G.; Bramucci, M.; Quassinti, L.; Bianco, A. Chemical composition and biological activity of the essential oil from Helichrysum microphyllum cambess. ssp. tyrrhenicum bacch., brullo e giusso growing in la maddalena archipelago, Sardinia. J. Oleo Sci. 2015, 64, 19–26. [Google Scholar] [CrossRef] [Green Version]
  46. Maggi, F.; Quassinti, L.; Bramucci, M.; Lupidi, G.; Petrelli, D.; Vitali, L.A.; Papa, F.; Vittori, S. Composition and biological activities of hogweed [Heracleum sphondylium L. subsp. ternatum (Velen.) Brummitt] essential oil and its main components octyl acetate and octyl butyrate. Nat. Prod. Res. 2014, 28, 1354–1363. [Google Scholar] [CrossRef]
  47. 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]
  48. 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, 3293–3299. [Google Scholar]
  49. Gismondi, A.; Canuti, L.; Grispo, M.; Canini, A. Biochemical composition and antioxidant properties of Lavandula angustifolia Miller essential oil are shielded by propolis against UV radiations. Photochem. Photobiol. 2014, 90, 702–708. [Google Scholar] [CrossRef]
  50. Ferraz, R.P.; Bomfim, D.S.; Carvalho, N.C.; Soares, M.B.; da Silva, T.B.; Machado, W.J.; Prata, A.P.; Costa, E.V.; Moraes, V.R.; Nogueira, P.C.; et al. Cytotoxic effect of leaf essential oil of Lippia gracilis Schauer (Verbenaceae). Phytomedicine 2013, 20, 615–621. [Google Scholar] [CrossRef] [Green Version]
  51. Quassinti, L.; Maggi, F.; Ortolani, F.; Lupidi, G.; Petrelli, D.; Vitali, L.A.; Miano, A.; Bramucci, M. Exploring new applications of tulip tree (Liriodendron tulipifera L.): Leaf essential oil as apoptotic agent for human glioblastoma. Environ. Sci. Pollut. Res. 2019, 26, 30485–30497. [Google Scholar] [CrossRef]
  52. Calcabrini, A.; Stringaro, A.; Toccacieli, L.; Meschini, S.; Marra, M.; Colone, M.; Salvatore, G.; Mondello, F.; Arancia, G.; Molinari, A. Terpinen-4-ol, the main component of Melaleuca alternifolia (tea tree) oil inhibits the in vitro growth of human melanoma cells. J. Investig. Dermatol. 2004, 122, 349–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Chao, W.W.; Su, C.C.; Peng, H.Y.; Chou, S.T. Melaleuca quinquenervia essential oil inhibits α-melanocyte-stimulating hormone-induced melanin production and oxidative stress in B16 melanoma cells. Phytomedicine 2017, 34, 191–201. [Google Scholar] [CrossRef] [PubMed]
  54. Chang, C.T.; Soo, W.N.; Chen, Y.H.; Shyur, L.F. Essential Oil of Mentha aquatica var. Kenting water mint suppresses two-stage skin carcinogenesis accelerated by BRAF inhibitor vemurafenib. Molecules 2019, 24, 2344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Stefanello, M.É.A.; Riva, D.; De Carvalho, J.E.; Ruiz, A.L.T.G.; Salvador, M.J. Chemical Composition and Cytotoxic Activity of Essential Oil from Myrcia laruotteana Fruits. J. Essent. Oil Res. 2011, 23, 7–10. [Google Scholar] [CrossRef]
  56. Piaru, S.P.; Mahmud, R.; Abdul Majid, A.M.; Mahmoud Nassar, Z.D. Antioxidant and antiangiogenic activities of the essential oils of Myristica fragrans and Morinda citrifolia. Asian Pac. J. Trop. Med. 2012, 5, 294–298. [Google Scholar] [CrossRef] [Green Version]
  57. Grecco Sdos, S.; Martins, E.G.; Girola, N.; de Figueiredo, C.R.; Matsuo, A.L.; Soares, M.G.; Bertoldo Bde, C.; Sartorelli, P.; Lago, J.H. Chemical composition and in vitro cytotoxic effects of the essential oil from Nectandra leucantha leaves. Pharm. Biol. 2015, 53, 133–137. [Google Scholar] [CrossRef]
  58. Trevisan, M.T.; Vasconcelos Silva, M.G.; Pfundstein, B.; Spiegelhalder, B.; Owen, R.W. Characterization of the volatile pattern and antioxidant capacity of essential oils from different species of the genus Ocimum. J. Agric. Food Chem. 2006, 54, 4378–4382. [Google Scholar] [CrossRef]
  59. El Khoury, R.; Michael-Jubeli, R.; Bakar, J.; Dakroub, H.; Rizk, T.; Baillet-Guffroy, A.; Lteif, R.; Tfayli, A. Origanum essential oils reduce the level of melanin in B16-F1 melanocytes. Eur. J. Dermatol. 2019, 29, 596–602. [Google Scholar] [CrossRef]
  60. Kwon, S.J.; Lee, J.H.; Moon, K.D.; Jeong, I.Y.; Ahn, D.U.K.; Lee, M.K.; Seo, K.I. Induction of apoptosis by isoegomaketone from Perilla frutescens L. in B16 melanoma cells is mediated through ROS generation and mitochondrial-dependent, -independent pathway. Food Chem. Toxicol. 2014, 65, 97–104. [Google Scholar] [CrossRef]
  61. Da Silva, J.K.R.; Pinto, L.C.; Burbano, R.M.R.; Montenegro, R.C.; Guimarães, E.F.; Andrade, E.H.A.; Maia, J.G.S. Essential oils of Amazon Piper species and their cytotoxic, antifungal, antioxidant and anti-cholinesterase activities. Ind. Crop. Prod. 2014, 58, 55–60. [Google Scholar] [CrossRef]
  62. Girola, N.; Figueiredo, C.R.; Farias, C.F.; Azevedo, R.A.; Ferreira, A.K.; Teixeira, S.F.; Capello, T.M.; Martins, E.G.A.; Matsuo, A.L.; Travassos, L.R.; et al. Camphene isolated from essential oil of Piper cernuum (Piperaceae) induces intrinsic apoptosis in melanoma cells and displays antitumor activity in vivo. Biochem. Biophys. Res. Commun. 2015, 467, 928–934. [Google Scholar] [CrossRef] [PubMed]
  63. Lima, R.N.; Ribeiro, A.S.; Santiago, G.M.P.; D’S. Costa, C.O.; Soares, M.B.; Bezerra, D.P.; Shanmugam, S.; Freitas, L.D.S.; Alves, P.B. Antitumor and Aedes aegypti Larvicidal Activities of Essential Oils from Piper klotzschianum, P. hispidum, and P. arboreum. Nat. Prod. Commun. 2019, 14. [Google Scholar] [CrossRef] [Green Version]
  64. Loutrari, H.; Magkouta, S.; Pyriochou, A.; Koika, V.; Kolisis, F.N.; Papapetropoulos, A.; Roussos, C. Mastic oil from Pistacia lentiscus var. chia inhibits growth and survival of human K562 leukemia cells and attenuates angiogenesis. Nutr. Cancer 2006, 55, 86–93. [Google Scholar] [CrossRef] [PubMed]
  65. Krifa, M.; El Mekdad, H.; Bentouati, N.; Pizzi, A.; Ghedira, K.; Hammami, M.; El Meshri, S.E.; Chekir-Ghedira, L. Immunomodulatory and anticancer effects of Pituranthos tortuosus essential oil. Tumor Biol. 2015, 36, 5165–5170. [Google Scholar] [CrossRef] [PubMed]
  66. Krifa, M.; El Meshri, S.E.; Bentouati, N.; Pizzi, A.; Sick, E.; Chekir-Ghedira, L.; Rondé, P. In Vitro and in Vivo Anti-Melanoma Effects of Pituranthos tortuosus Essential Oil Via Inhibition of FAK and Src Activities. J. Cell. Biochem. 2016, 117, 1167–1175. [Google Scholar] [CrossRef]
  67. Manjamalai, A.; Grace, V.M.B. The chemotherapeutic effect of essential oil of Plectranthus amboinicus (Lour) on lung metastasis developed by B16F-10 cell line in C57BL/6 mice. Cancer Investig. 2013, 31, 74–82. [Google Scholar] [CrossRef]
  68. He, W.; Li, X.; Peng, Y.; He, X.; Pan, S. Anti-oxidant and anti-melanogenic properties of essential oil from peel of Pomelo cv. Guan XI. Molecules 2019, 24, 242. [Google Scholar] [CrossRef] [Green Version]
  69. Da Silva, E.B.; Matsuo, A.L.; Figueiredo, C.R.; Chaves, M.H.; Sartorelli, P.; Lago, J.H. Chemical constituents and cytotoxic evaluation of essential oils from leaves of Porcelia macrocarpa (Annonaceae). Nat. Prod. Commun. 2013, 8, 277–279. [Google Scholar] [CrossRef] [Green Version]
  70. Dutra, R.C.; Pittella, F.; Dittz, D.; Marcon, R.; Pimenta, D.S.; Lopes, M.T.P.; Raposo, N.R.B. Chemical composition and cytotoxicity activity of the essential oil of Pterodon emarginatus. Rev. Bras. Farmacogn. 2012, 22, 971–978. [Google Scholar] [CrossRef] [Green Version]
  71. Russo, A.; Formisano, C.; Rigano, D.; Cardile, V.; Arnold, N.A.; Senatore, F. Comparative phytochemical profile and antiproliferative activity on human melanoma cells of essential oils of three lebanese Salvia species. Ind. Crop. Prod. 2016, 83, 492–499. [Google Scholar] [CrossRef]
  72. Cardile, V.; Russo, A.; Formisano, C.; Rigano, D.; Senatore, F.; Arnold, N.A.; Piozzi, F. Essential oils of Salvia bracteata and Salvia rubifolia from Lebanon: Chemical composition, antimicrobial activity and inhibitory effect on human melanoma cells. J. Ethnopharmacol. 2009, 126, 265–272. [Google Scholar] [CrossRef] [PubMed]
  73. Gali-Muhtasib, H.U.; Affara, N.I. Chemopreventive effects of sage oil on skin papillomas in mice. Phytomedicine 2000, 7, 129–136. [Google Scholar] [CrossRef]
  74. Alexa, E.; Sumalan, R.M.; Danciu, C.; Obistioiu, D.; Negrea, M.; Poiana, M.A.; Rus, C.; Radulov, I.; Pop, G.; Dehelean, C. Synergistic antifungal, allelopatic and anti-proliferative potential of Salvia officinalis L., and Thymus vulgaris L. Essential oils. Molecules 2018, 23, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Russo, A.; Cardile, V.; Graziano, A.C.E.; Formisano, C.; Rigano, D.; Canzoneri, M.; Bruno, M.; Senatore, F. Comparison of essential oil components and in vitro anticancer activity in wild and cultivated Salvia verbenaca. Nat. Prod. Res. 2015, 29, 1630–1640. [Google Scholar] [CrossRef] [PubMed]
  76. Santha, S.; Dwivedi, C. Anticancer Effects of Sandalwood (Santalum album). Anticancer Res. 2015, 35, 3137–3145. [Google Scholar]
  77. Popovici, R.A.; Vaduva, D.; Pinzaru, I.; Dehelean, C.A.; Farcas, C.G.; Coricovac, D.; Danciu, C.; Popescu, I.; Alexa, E.; Lazureanu, V.; et al. A comparative study on the biological activity of essential oil and total hydro-alcoholic extract of Satureja hortensis L. Exp. Ther. Med. 2019, 18, 932–942. [Google Scholar] [CrossRef] [PubMed]
  78. Santana, J.S.; Sartorelli, P.; Guadagnin, R.C.; Matsuo, A.L.; Figueiredo, C.R.; Soares, M.G.; Da Silva, A.M.; Lago, J.H.G. Essential oils from Schinus terebinthifolius leaves chemical composition and in vitro cytotoxicity evaluation. Pharm. Biol. 2012, 50, 1248–1253. [Google Scholar] [CrossRef] [Green Version]
  79. Conforti, F.; Menichini, F.; Formisano, C.; Rigano, D.; Senatore, F.; Arnold, N.A.; Piozzi, F. Comparative chemical composition, free radical-scavenging and cytotoxic properties of essential oils of six Stachys species from different regions of the Mediterranean Area. Food Chem. 2009, 116, 898–905. [Google Scholar] [CrossRef]
  80. Shakeri, A.; D’Urso, G.; Taghizadeh, S.F.; Piacente, S.; Norouzi, S.; Soheili, V.; Asili, J.; Salarbashi, D. LC-ESI/LTQOrbitrap/MS/MS and GC–MS profiling of Stachys parviflora L. and evaluation of its biological activities. J. Pharm. Biomed. Anal. 2019, 168, 209–216. [Google Scholar] [CrossRef]
  81. Arung, E.T.; Matsubara, E.; Kusuma, I.W.; Sukaton, E.; Shimizu, K.; Kondo, R. Inhibitory components from the buds of clove (Syzygium aromaticum) on melanin formation in B16 melanoma cells. Fitoterapia 2011, 82, 198–202. [Google Scholar] [CrossRef]
  82. De Oliveira, P.F.; Alves, J.M.; Damasceno, J.L.; Oliveira, R.A.M.; Júnior Dias, H.; Crotti, A.E.M.; Tavares, D.C. Cytotoxicity screening of essential oils in cancer cell lines. Rev. Bras. Farmacogn. 2015, 25, 183–188. [Google Scholar] [CrossRef] [Green Version]
  83. Venditti, A.; Frezza, C.; Sciubba, F.; Serafini, M.; Bianco, A.; Cianfaglione, K.; Lupidi, G.; Quassinti, L.; Bramucci, M.; Maggi, F. Volatile components, polar constituents and biological activity of tansy daisy (Tanacetum macrophyllum (Waldst. et Kit.) Schultz Bip.). Ind. Crop. Prod. 2018, 118, 225–235. [Google Scholar] [CrossRef]
  84. Dall’Acqua, S.; Peron, G.; Ferrari, S.; Gandin, V.; Bramucci, M.; Quassinti, L.; Mártonfi, P.; Maggi, F. Phytochemical investigations and antiproliferative secondary metabolites from Thymus alternans growing in Slovakia. Pharm. Biol. 2017, 55, 1162–1170. [Google Scholar] [CrossRef] [Green Version]
  85. Bendif, H.; Boudjeniba, M.; Miara, M.D.; Biqiku, L.; Bramucci, M.; Lupidi, G.; Quassinti, L.; Vitali, L.A.; Maggi, F. Essential Oil of Thymus munbyanus subsp. coloratus from Algeria: Chemotypification and in vitro Biological Activities. Chem. Biodivers. 2017, 14, e1600299. [Google Scholar] [CrossRef]
  86. Manjamalai, A.; Mahesh Kumar, M.J.; Berlin Grace, V.M. Essential oil of tridax procumbens L induces apoptosis and suppresses angiogenesis and lung metastasis of the B16F-10 cell line in C57BL/6 mice. Asian Pac. J. Cancer Prev. 2012, 13, 5887–5895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Peng, H.Y.; Lai, C.C.; Lin, C.C.; Chou, S.T. Effect of Vetiveria zizanioides essential oil on melanogenesis in melanoma cells: Downregulation of tyrosinase expression and suppression of oxidative stress. Sci. World J. 2014, 2014. [Google Scholar] [CrossRef]
  88. Huang, H.C.; Chang, T.Y.; Chang, L.Z.; Wang, H.F.; Yih, K.H.; Hsieh, W.Y.; Chang, T.M. Inhibition of melanogenesis Versus antioxidant properties of essential oil extracted from leaves of vitex negundo linn and chemical composition analysis by GC-MS. Molecules 2012, 17, 3902–3916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Lee, H.G.; Kim, T.Y.; Jeon, J.H.; Lee, S.H.; Hong, Y.K.; Jin, M.H. Inhibition of melanogenesis by abietatriene from Vitex trifolia leaf oil. Nat. Prod. Sci. 2016, 22, 252–258. [Google Scholar] [CrossRef]
  90. Manjamalai, A.; Berlin Grace, V.M. Antioxidant activity of essential oils from Wedelia chinensis (Osbeck) in vitro and in vivo lung cancer bearing C57BL/6 mice. Asian Pac. J. Cancer Prev. 2012, 13, 3065–3071. [Google Scholar] [CrossRef] [Green Version]
  91. Costa, E.V.; Menezes, L.R.A.; Rocha, S.L.A.; Baliza, I.R.S.; Dias, R.B.; Rocha, C.A.G.; Soares, M.B.P.; Bezerra, D.P. Antitumor properties of the leaf essential oil of Zornia brasiliensis. Planta Med. 2015, 81, 563–567. [Google Scholar] [CrossRef]
  92. Harris, R. Synergism in the essential oil world. Int. J. Aromather. 2002, 12, 179–186. [Google Scholar] [CrossRef]
  93. Patsilinakos, A.; Artini, M.; Papa, R.; Sabatino, M.; Bozovic, M.; Garzoli, S.; Vrenna, G.; Buzzi, R.; Manfredini, S.; Selan, L.; et al. Machine Learning Analyses on Data including Essential Oil Chemical Composition and In Vitro Experimental Antibiofilm Activities against Staphylococcus Species. Molecules 2019, 24, 890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Freitas, J.V.B.; Alves Filho, E.G.; Silva, L.M.A.; Zocolo, G.J.; de Brito, E.S.; Gramosa, N.V. Chemometric analysis of NMR and GC datasets for chemotype characterization of essential oils from different species of Ocimum. Talanta 2018, 180, 329–336. [Google Scholar] [CrossRef] [PubMed]
  95. Clarke, S. Chapter 8-Handling, safety and practical applications for use of essential oils. In Essential Chemistry for Aromatherapy, 2nd ed.; Clarke, S., Ed.; Churchill Livingstone: Edinburgh, UK, 2008; pp. 231–264. [Google Scholar] [CrossRef]
  96. Sabatino, M.; Fabiani, M.; Bozovic, M.; Garzoli, S.; Antonini, L.; Marcocci, M.E.; Palamara, A.T.; De Chiara, G.; Ragno, R. Experimental Data Based Machine Learning Classification Models with Predictive Ability to Select in Vitro Active Antiviral and Non-Toxic Essential Oils. Molecules 2020, 25, 2452. [Google Scholar] [CrossRef]
  97. Delfine, S.; Marrelli, M.; Conforti, F.; Formisano, C.; Rigano, D.; Menichini, F.; Senatore, F. Variation of Malva sylvestris essential oil yield, chemical composition and biological activity in response to different environments across Southern Italy. Ind. Crop. Prod. 2017, 98, 29–37. [Google Scholar] [CrossRef]
  98. Djarri, L.; Medjroubi, K.; Akkal, S.; Elomri, A.; Seguin, E.; Groult, M.L.; Verite, P. Variability of two essential oils of Kundmannia sicula (L.) DC., a traditional Algerian medicinal plant. Molecules 2008, 13, 812–817. [Google Scholar] [CrossRef] [Green Version]
  99. Sefidkon, F.; Abbasi, K.; Khaniki, G.B. Influence of drying and extraction methods on yield and chemical composition of the essential oil of Satureja hortensis. Food Chem. 2006, 99, 19–23. [Google Scholar] [CrossRef]
  100. Mohtashami, S.; Rowshan, V.; Tabrizi, L.; Babalar, M.; Ghani, A. Summer savory (Satureja hortensis L.) essential oil constituent oscillation at different storage conditions. Ind. Crop. Prod. 2018, 111, 226–231. [Google Scholar] [CrossRef]
  101. Russo, A.; Formisano, C.; Rigano, D.; Senatore, F.; Delfine, S.; Cardile, V.; Rosselli, S.; Bruno, M. Chemical composition and anticancer activity of essential oils of Mediterranean sage (Salvia officinalis L.) grown in different environmental conditions. Food Chem. Toxicol. 2013, 55, 42–47. [Google Scholar] [CrossRef]
  102. Garzoli, S.; Pirolli, A.; Vavala, E.; Di Sotto, A.; Sartorelli, G.; Bozovic, M.; Angiolella, L.; Mazzanti, G.; Pepi, F.; Ragno, R. Multidisciplinary Approach to Determine the Optimal Time and Period for Extracting the Essential Oil from Mentha suaveolens Ehrh. Molecules 2015, 20, 9640–9655. [Google Scholar] [CrossRef] [Green Version]
  103. Bozovic, M.; Navarra, A.; Garzoli, S.; Pepi, F.; Ragno, R. Esential oils extraction: A 24-hour steam distillation systematic methodology. Nat. Prod. Res. 2017, 31, 2387–2396. [Google Scholar] [CrossRef] [PubMed]
  104. Padilla-González, G.F.; Frey, M.; Gómez-Zeledón, J.; Da Costa, F.B.; Spring, O. Metabolomic and gene expression approaches reveal the developmental and environmental regulation of the secondary metabolism of yacón (Smallanthus sonchifolius, Asteraceae). Sci. Rep. 2019, 9, 1–15. [Google Scholar] [CrossRef] [PubMed]
  105. Zámboriné Németh, É.; Thi Nguyen, H. Thujone, a widely debated volatile compound: What do we know about it? Phytochem. Rev. 2020, 19, 405–423. [Google Scholar] [CrossRef]
  106. McAnally, J.A.; Jung, M.; Mo, H. Farnesyl-O-acetylhydroquinone and geranyl-O-acetylhydroquinone suppress the proliferation of murine B16 melanoma cells, human prostate and colon adenocarcinoma cells, human lung carcinoma cells, and human leukemia cells. Cancer Lett. 2003, 202, 181–192. [Google Scholar] [CrossRef] [PubMed]
  107. Mo, H.; Tatman, D.; Jung, M.; Elson, C.E. Farnesyl anthranilate suppresses the growth, in vitro and in vivo, of murine B16 melanomas. Cancer Lett. 2000, 157, 145–153. [Google Scholar] [CrossRef]
  108. Effenberger, K.; Breyer, S.; Schobert, R. Terpene conjugates of the Nigella sativa seed-oil constituent thymoquinone with enhanced efficacy in cancer cells. Chem. Biodivers. 2010, 7, 129–139. [Google Scholar] [CrossRef]
  109. Tatman, D.; Mo, H. Volatile isoprenoid constituents of fruits, vegetables and herbs cumulatively suppress the proliferation of murine B16 melanoma and human HL-60 leukemia cells. Cancer Lett. 2002, 175, 129–139. [Google Scholar] [CrossRef]
  110. Fabbri, J.; Maggiore, M.A.; Pensel, P.E.; Denegri, G.M.; Elissondo, M.C. In vitro efficacy study of Cinnamomum zeylanicum essential oil and cinnamaldehyde against the larval stage of Echinococcus granulosus. Exp. Parasitol. 2020, 214, 107904. [Google Scholar] [CrossRef]
  111. Mediouni, S.; Jablonski, J.A.; Tsuda, S.; Barsamian, A.; Kessing, C.; Richard, A.; Biswas, A.; Toledo, F.; Andrade, V.M.; Even, Y.; et al. Oregano Oil and Its Principal Component, Carvacrol, Inhibit HIV-1 Fusion into Target Cells. J. Virol. 2020, 94. [Google Scholar] [CrossRef]
  112. Liu, Q.; Meng, X.; Li, Y.; Zhao, C.N.; Tang, G.Y.; Li, H.B. Antibacterial and Antifungal Activities of Spices. Int. J. Mol. Sci. 2017, 18, 1283. [Google Scholar] [CrossRef] [Green Version]
  113. Kokoska, L.; Kloucek, P.; Leuner, O.; Novy, P. Plant-Derived Products as Antibacterial and Antifungal Agents in Human Health Care. Curr. Med. Chem. 2019, 26, 5501–5541. [Google Scholar] [CrossRef] [PubMed]
  114. Deyno, S.; Mtewa, A.G.; Abebe, A.; Hymete, A.; Makonnen, E.; Bazira, J.; Alele, P.E. Essential oils as topical anti-infective agents: A systematic review and meta-analysis. Complement. Ther. Med. 2019, 47, 102224. [Google Scholar] [CrossRef] [PubMed]
  115. Fidler, I.J. Selection of successive tumour lines for metastasis. Nat. New Biol. 1973, 242, 148–149. [Google Scholar] [CrossRef] [PubMed]
  116. Danciu, C.; Oprean, C.; Coricovac, D.E.; Andreea, C.; Cimpean, A.; Radeke, H.; Soica, C.; Dehelean, C. Behaviour of four different B16 murine melanoma cell sublines: C57BL/6J skin. Int. J. Exp. Pathol. 2015, 96, 73–80. [Google Scholar] [CrossRef]
  117. Teicher, B.A. Tumor Models in Cancer Research; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2010. [Google Scholar]
  118. Paschoalin, T.; Carmona, A.K.; Rodrigues, E.G.; Oliveira, V.; Monteiro, H.P.; Juliano, M.A.; Juliano, L.; Travassos, L.R. Characterization of thimet oligopeptidase and neurolysin activities in B16F10-Nex2 tumor cells and their involvement in angiogenesis and tumor growth. Mol. Cancer 2007, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  119. Nakamura, K.; Yoshikawa, N.; Yamaguchi, Y.; Kagota, S.; Shinozuka, K.; Kunitomo, M. Characterization of mouse melanoma cell lines by their mortal malignancy using an experimental metastatic model. Life Sci. 2002, 70, 791–798. [Google Scholar] [CrossRef]
  120. Shi, H.; Liu, L.; Liu, L.M.; Geng, J.; Chen, L. Inhibition of tumor growth by beta-elemene through downregulation of the expression of uPA, uPAR, MMP-2, and MMP-9 in a murine intraocular melanoma model. Melanoma Res. 2015, 25, 15–21. [Google Scholar] [CrossRef]
  121. Hatiboglu, M.A.; Kocyigit, A.; Guler, E.M.; Akdur, K.; Nalli, A.; Karatas, E.; Tuzgen, S. Thymoquinone Induces Apoptosis in B16-F10 Melanoma Cell Through Inhibition of p-STAT3 and Inhibits Tumor Growth in a Murine Intracerebral Melanoma Model. World Neurosurg. 2018, 114, e182–e190. [Google Scholar] [CrossRef]
  122. Xu, M.; McCanna, D.J.; Sivak, J.G. Use of the viability reagent PrestoBlue in comparison with alamarBlue and MTT to assess the viability of human corneal epithelial cells. J. Pharmacol. Toxicol. 2015, 71, 1–7. [Google Scholar] [CrossRef]
  123. Mead, T.J.; Lefebvre, V. Proliferation assays (BrdU and EdU) on skeletal tissue sections. Methods Mol. Biol. 2014, 1130, 233–243. [Google Scholar]
  124. Griffiths, M.; Sundaram, H. Drug design and testing: Profiling of antiproliferative agents for cancer therapy using a cell-based methyl-[3H]-thymidine incorporation assay. Methods Mol. Biol. 2011, 731, 451–465. [Google Scholar] [PubMed]
  125. Alsaraf, S.; Hadi, Z.; Al-Lawati, W.M.; Al Lawati, A.A.; Khan, S.A. Chemical composition, in vitro antibacterial and antioxidant potential of Omani Thyme essential oil along with in silico studies of its major constituent. J. King Saud Univ. Sci. 2020, 32, 1021–1028. [Google Scholar] [CrossRef]
  126. Mitropoulou, G.; Fitsiou, E.; Spyridopoulou, K.; Tiptiri-Kourpeti, A.; Bardouki, H.; Vamvakias, M.; Panas, P.; Chlichlia, K.; Pappa, A.; Kourkoutas, Y. Citrus medica essential oil exhibits significant antimicrobial and antiproliferative activity. LWT Food Sci. Technol. 2017, 84, 344–352. [Google Scholar] [CrossRef]
  127. Govindaraju, S.; Arulselvi, P.I. Characterization of Coleus aromaticus essential oil and its major constituent carvacrol for in vitro antidiabetic and antiproliferative activities. J. Herbs Spices Med. Plants 2018, 24, 37–51. [Google Scholar] [CrossRef]
  128. Melo, J.O.; Fachin, A.L.; Rizo, W.F.; Jesus, H.C.R.; Arrigoni-Blank, M.F.; Alves, P.B.; Marins, M.A.; França, S.C.; Blank, A.F. Cytotoxic effects of essential oils from three lippia gracilis schauer genotypes on HeLa, B16, and MCF-7 cells and normal human fibroblasts. Genet. Mol. Res. 2014, 13, 2691–2697. [Google Scholar] [CrossRef]
  129. Ramadan, M.A.; Shawkey, A.E.; Rabeh, M.A.; Abdellatif, A.O. Expression of P53, BAX, and BCL-2 in human malignant melanoma and squamous cell carcinoma cells after tea tree oil treatment in vitro. Cytotechnology 2019, 71, 461–473. [Google Scholar] [CrossRef]
  130. Greay, S.J.; Ireland, D.J.; Kissick, H.T.; Levy, A.; Beilharz, M.W.; Riley, T.V.; Carson, C.F. Induction of necrosis and cell cycle arrest in murine cancer cell lines by Melaleuca alternifolia (tea tree) oil and terpinen-4-ol. Cancer Chemother. Pharmacol. 2010, 65, 877–888. [Google Scholar] [CrossRef]
  131. Biswas, R.; Mandal, S.K.; Dutta, S.; Bhattacharyya, S.S.; Boujedaini, N.; Khuda-Bukhsh, A.R. Thujone-Rich Fraction of Thuja occidentalis Demonstrates Major Anti-Cancer Potentials: Evidences from In Vitro Studies on A375 Cells. Evid. Based Complement. Altern. Med. 2011, 2011, 568148. [Google Scholar] [CrossRef] [Green Version]
  132. Rajput, J.; Bagul, S.; Tadavi, S.; Bendre, R. Comparative Anti-Proliferative Studies of Natural Phenolic Monoterpenoids on Human Malignant Tumour Cells. Med. Aromat. Plants 2016, 5, 1–4. [Google Scholar] [CrossRef] [Green Version]
  133. Sanches, L.J.; Marinello, P.C.; Panis, C.; Fagundes, T.R.; Morgado-Diaz, J.A.; de-Freitas-Junior, J.C.; Cecchini, R.; Cecchini, A.L.; Luiz, R.C. Cytotoxicity of citral against melanoma cells: The involvement of oxidative stress generation and cell growth protein reduction. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [Green Version]
  134. Junior, P.L.; Camara, D.A.; Costa, A.S.; Ruiz, J.L.; Levy, D.; Azevedo, R.A.; Pasqualoto, K.F.; de Oliveira, C.F.; de Melo, T.C.; Pessoa, N.D.; et al. Apoptotic effect of eugenol envolves G2/M phase abrogation accompanied by mitochondrial damage and clastogenic effect on cancer cell in vitro. Phytomedicine 2016, 23, 725–735. [Google Scholar] [CrossRef] [PubMed]
  135. Ghosh, R.; Nadiminty, N.; Fitzpatrick, J.E.; Alworth, W.L.; Slaga, T.J.; Kumar, A.P. Eugenol causes melanoma growth suppression through inhibition of E2F1 transcriptional activity. J. Biol. Chem. 2005, 280, 5812–5819. [Google Scholar] [CrossRef] [Green Version]
  136. Pisano, M.; Pagnan, G.; Loi, M.; Mura, M.E.; Tilocca, M.G.; Palmieri, G.; Fabbri, D.; Dettori, M.A.; Delogu, G.; Ponzoni, M.; et al. Antiproliferative and pro-apoptotic activity of eugenol-related biphenyls on malignant melanoma cells. Mol. Cancer 2007, 6, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Kijpornyongpan, T.; Sereemaspun, A.; Chanchao, C. Dose-dependent cytotoxic effects of menthol on human malignant melanoma A-375 cells: Correlation with TRPM8 transcript expression. Asian Pac. J. Cancer Prev. 2014, 15, 1551–1556. [Google Scholar] [CrossRef] [Green Version]
  138. Yan, H.; Ren, M.Y.; Wang, Z.X.; Feng, S.J.; Li, S.; Cheng, Y.; Hu, C.X.; Gao, S.Q.; Zhang, G.Q. Zerumbone inhibits melanoma cell proliferation and migration by altering mitochondrial functions. Oncol. Lett. 2017, 13, 2397–2402. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, S.D.; Wang, Z.H.; Yan, H.Q.; Ren, M.Y.; Gao, S.Q.; Zhang, G.Q. Chemotherapeutic effect of Zerumbone on melanoma cells through mitochondria-mediated pathways. Clin. Exp. Dermatol. 2016, 41, 858–863. [Google Scholar] [CrossRef]
  140. Jung, J.I.; Kim, E.J.; Kwon, G.T.; Jung, Y.J.; Park, T.; Kim, Y.; Yu, R.; Choi, M.S.; Chun, H.S.; Kwon, S.H.; et al. β-Caryophyllene potently inhibits solid tumor growth and lymph node metastasis of B16F10 melanoma cells in high-fat diet-induced obese C57BL/6N mice. Carcinogenesis 2015, 36, 1028–1039. [Google Scholar] [CrossRef] [Green Version]
  141. Zhang, Y.; Li, C.; Huang, Y.; Zhao, S.; Xu, Y.; Chen, Y.; Jiang, F.; Tao, L.; Shen, X. EOFAZ inhibits endothelialtomesenchymal transition through downregulation of KLF4. Int. J. Mol. Med. 2020, 46, 300–310. [Google Scholar]
  142. Bozzuto, G.; Colone, M.; Toccacieli, L.; Stringaro, A.; Molinari, A. Tea tree oil might combat melanoma. Planta Med. 2011, 77, 54–56. [Google Scholar] [CrossRef]
  143. Martins, B.X.; Arruda, R.F.; Costa, G.A.; Jerdy, H.; de Souza, S.B.; Santos, J.M.; de Freitas, W.R.; Kanashiro, M.M.; de Carvalho, E.C.Q.; Sant’Anna, N.F.; et al. Myrtenal-induced V-ATPase inhibition-A toxicity mechanism behind tumor cell death and suppressed migration and invasion in melanoma. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 1–12. [Google Scholar] [CrossRef]
  144. Siveen, K.S.; Kuttan, G. Thujone inhibits lung metastasis induced by B16F-10 melanoma cells in C57BL/6 mice. Can. J. Physiol. Pharmacol. 2011, 89, 691–703. [Google Scholar] [CrossRef] [PubMed]
  145. Ahmad, I.; Muneer, K.M.; Tamimi, I.A.; Chang, M.E.; Ata, M.O.; Yusuf, N. Thymoquinone suppresses metastasis of melanoma cells by inhibition of NLRP3 inflammasome. Toxicol. Appl. Pharmacol. 2013, 270, 70–76. [Google Scholar] [CrossRef] [PubMed]
  146. Hakkim, F.L.; Bakshi, H.A.; Khan, S.; Nasef, M.; Farzand, R.; Sam, S.; Rashan, L.; Al-Baloshi, M.S.; Abdo Hasson, S.S.A.; Jabri, A.A.; et al. Frankincense essential oil suppresses melanoma cancer through down regulation of Bcl-2/Bax cascade signaling and ameliorates heptotoxicity via phase I and II drug metabolizing enzymes. Oncotarget 2019, 10, 3472–3490. [Google Scholar] [CrossRef] [Green Version]
  147. Greay, S.J.; Ireland, D.J.; Kissick, H.T.; Heenan, P.J.; Carson, C.F.; Riley, T.V.; Beilharz, M.W. Inhibition of established subcutaneous murine tumour growth with topical Melaleuca alternifolia (tea tree) oil. Cancer Chemother. Pharmacol. 2010, 66, 1095–1102. [Google Scholar] [CrossRef]
  148. Jedinak, A.; Muckova, M.; Kost’alova, D.; Maliar, T.; Masterova, I. Antiprotease and antimetastatic activity of ursolic acid isolated from Salvia officinalis. Z. Naturforsch. C J. Biosci. 2006, 61, 777–782. [Google Scholar] [CrossRef] [Green Version]
  149. Matsuo, A.L.; Figueiredo, C.R.; Arruda, D.C.; Pereira, F.V.; Scutti, J.A.; Massaoka, M.H.; Travassos, L.R.; Sartorelli, P.; Lago, J.H. Alpha-Pinene isolated from Schinus terebinthifolius Raddi (Anacardiaceae) induces apoptosis and confers antimetastatic protection in a melanoma model. Biochem. Biophys. Res. Commun. 2011, 411, 449–454. [Google Scholar] [CrossRef] [Green Version]
  150. Raphael, T.J.; Kuttan, G. Effect of naturally occurring monoterpenes carvone, limonene and perillic acid in the inhibition of experimental lung metastasis induced by B16F-10 melanoma cells. J. Exp. Clin. Cancer Res. 2003, 22, 419–424. [Google Scholar]
  151. Lee, S.R.; Mun, J.Y.; Jeong, M.S.; Lee, H.H.; Roh, Y.G.; Kim, W.T.; Kim, M.H.; Heo, J.; Choi, Y.H.; Kim, S.J.; et al. Thymoquinone-Induced Tristetraprolin Inhibits Tumor Growth and Metastasis through Destabilization of MUC4 mRNA. Int. J. Mol. Sci. 2019, 20, 2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Chen, W.; Lu, Y.; Wu, J.; Gao, M.; Wang, A.; Xu, B. Beta-elemene inhibits melanoma growth and metastasis via suppressing vascular endothelial growth factor-mediated angiogenesis. Cancer Chemother. Pharmacol. 2011, 67, 799–808. [Google Scholar] [CrossRef]
  153. Lim, S.; Kaldis, P. Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development 2013, 140, 3079. [Google Scholar] [CrossRef] [Green Version]
  154. Bommareddy, A.; Brozena, S.; Steigerwalt, J.; Landis, T.; Hughes, S.; Mabry, E.; Knopp, A.; VanWert, A.L.; Dwivedi, C. Medicinal properties of alpha-santalol, a naturally occurring constituent of sandalwood oil. Nat. Prod. Res. 2019, 33, 527–543. [Google Scholar] [CrossRef]
  155. Zhang, X.; Dwivedi, C. Skin cancer chemoprevention by α-santalol. Front. Biosci. Schol. Ed. 2011, 3, S777–S787. [Google Scholar]
  156. Barboza, J.N.; da Silva Maia Bezerra Filho, C.; Silva, R.O.; Medeiros, J.V.R.; de Sousa, D.P. An overview on the anti-inflammatory potential and antioxidant profile of eugenol. Oxid. Med. Cell. Longev. 2018, 2018. [Google Scholar] [CrossRef] [PubMed]
  157. Liu, X.; Mi, J.; Qin, H.; Li, Z.; Chai, J.; Li, M.; Wu, J.; Xu, J. E2F1/IGF-1R Loop Contributes to BRAF Inhibitor Resistance in Melanoma. J. Investig. Dermatol. 2020, 140, 1295–1299. [Google Scholar] [CrossRef]
  158. Meng, P.; Bedolla, R.G.; Yun, H.; Fitzpatrick, J.E.; Kumar, A.P.; Ghosh, R. Contextual role of E2F1 in suppression of melanoma cell motility and invasiveness. Mol. Carcinog. 2019, 58, 1701–1710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Delmondes, G.A.; Santiago Lemos, I.C.; Dias, D.Q.; Cunha, G.L.D.; Araújo, I.M.; Barbosa, R.; Coutinho, H.D.M. Pharmacological applications of farnesol (C(15)H(26)O): A patent review. Expert Opin. Ther. Pat. 2020, 30, 227–234. [Google Scholar] [CrossRef]
  160. Cerchiara, T.; Straface, S.V.; Brunelli, E.; Tripepi, S.; Gallucci, M.C.; Chidichimo, G. Antiproliferative effect of linalool on RPMI 7932 human melanoma cell line: Ultrastructural studies. Nat. Prod. Commun. 2015, 10, 547–549. [Google Scholar] [CrossRef] [Green Version]
  161. Slominski, A. Cooling skin cancer: Menthol inhibits melanoma growth. Focus on “TRPM8 activation suppresses cellular viability in human melanoma”. Am. J. Physiol. Cell Physiol. 2008, 295, C293–C295. [Google Scholar] [CrossRef] [Green Version]
  162. Yamamura, H.; Ugawa, S.; Ueda, T.; Morita, A.; Shimada, S. TRPM8 activation suppresses cellular viability in human melanoma. Am. J. Physiol. Cell Physiol. 2008, 295, C296–C301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
  164. Krysko, D.V.; Berghe, T.V.; Parthoens, E.; D’Herde, K.; Vandenabeele, P. Methods for distinguishing apoptotic from necrotic cells and measuring their clearance. Method. Enzymol. 2008, 442, 307–341. [Google Scholar]
  165. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
  166. Choucroun, P.; Gillet, D.; Dorange, G.; Sawicki, B.; Dewitte, J.D. Comet assay and early apoptosis. Mutat. Res. 2001, 478, 89–96. [Google Scholar] [CrossRef]
  167. Giordani, C.; Molinari, A.; Toccacieli, L.; Calcabrini, A.; Stringaro, A.; Chistolini, P.; Arancia, G.; Diociaiuti, M. Interaction of tea tree oil with model and cellular membranes. J. Med. Chem. 2006, 49, 4581–4588. [Google Scholar] [CrossRef] [PubMed]
  168. Galluzzi, L.; Vitale, I.; Warren, S.; Adjemian, S.; Agostinis, P.; Martinez, A.B.; Chan, T.A.; Coukos, G.; Demaria, S.; Deutsch, E.; et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [Green Version]
  169. Salehi, B.; Upadhyay, S.; Orhan, I.E.; Jugran, A.K.; Jayaweera, S.L.D.; Dias, D.A.; Sharopov, F.; Taheri, Y.; Martins, N.; Baghalpour, N.; et al. Therapeutic potential of α-and β-pinene: A miracle gift of nature. Biomolecules 2019, 9, 738. [Google Scholar] [CrossRef] [Green Version]
  170. Jaganathan, S.K.; Supriyanto, E. Antiproliferative and molecular mechanism of eugenol-induced apoptosis in cancer cells. Molecules 2012, 17, 6290–6304. [Google Scholar] [CrossRef]
  171. Girisa, S.; Shabnam, B.; Monisha, J.; Fan, L.; Halim, C.E.; Arfuso, F.; Ahn, K.S.; Sethi, G.; Kunnumakkara, A.B. Potential of zerumbone as an anti-cancer agent. Molecules 2019, 24, 734. [Google Scholar] [CrossRef] [Green Version]
  172. Pelkonen, O.; Abass, K.; Wiesner, J. Thujone and thujone-containing herbal medicinal and botanical products: Toxicological assessment. Regul. Toxicol. Pharm. 2013, 65, 100–107. [Google Scholar] [CrossRef]
  173. Guo, H.; Carlson, J.A.; Slominski, A. Role of TRPM in melanocytes and melanoma. Exp. Dermatol. 2012, 21, 650–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid. Redox. Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [Green Version]
  175. Kuma, A.; Komatsu, M.; Mizushima, N. Autophagy-monitoring and autophagy-deficient mice. Autophagy 2017, 13, 1619–1628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Athamneh, K.; Alneyadi, A.; Alsamri, H.; Alrashedi, A.; Palakott, A.; El-Tarabily, K.A.; Eid, A.H.; Al Dhaheri, Y.; Iratni, R. Origanum majorana Essential Oil Triggers p38 MAPK-Mediated Protective Autophagy, Apoptosis, and Caspase-Dependent Cleavage of P70S6K in Colorectal Cancer Cells. Biomolecules 2020, 10, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Chaudhary, S.C.; Siddiqui, M.S.; Athar, M.; Alam, M.S. D-Limonene modulates inflammation, oxidative stress and Ras-ERK pathway to inhibit murine skin tumorigenesis. Hum. Exp. Toxicol. 2012, 31, 798–811. [Google Scholar] [CrossRef] [PubMed]
  178. Pudełek, M.; Catapano, J.; Kochanowski, P.; Mrowiec, K.; Janik-Olchawa, N.; Czyż, J.; Ryszawy, D. Therapeutic potential of monoterpene α-thujone, the main compound of Thuja occidentalis L. essential oil, against malignant glioblastoma multiforme cells in vitro. Fitoterapia 2019, 134, 172–181. [Google Scholar] [CrossRef]
  179. Russo, R.; Cassiano, M.G.; Ciociaro, A.; Adornetto, A.; Varano, G.P.; Chiappini, C.; Berliocchi, L.; Tassorelli, C.; Bagetta, G.; Corasaniti, M.T. Role of D-Limonene in autophagy induced by bergamot essential oil in SH-SY5Y neuroblastoma cells. PLoS ONE 2014, 9, e113682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Banjerdpongchai, R.; Khaw-On, P. Terpinen-4-ol induces autophagic and apoptotic cell death in human leukemic HL-60 cells. Asian Pac. J. Cancer Prev. 2013, 14, 7537–7542. [Google Scholar] [CrossRef] [Green Version]
  181. Ding, X.F.; Shen, M.; Xu, L.Y.; Dong, J.H.; Chen, G. 13,14-bis(cis-3,5-dimethyl-1-piperazinyl)-beta-elemene, a novel beta-elemene derivative, shows potent antitumor activities via inhibition of mTOR in human breast cancer cells. Oncol. Lett. 2013, 5, 1554–1558. [Google Scholar] [CrossRef] [Green Version]
  182. Streit, M.; Detmar, M. Angiogenesis, lymphangiogenesis, and melanoma metastasis. Oncogene 2003, 22, 3172–3179. [Google Scholar] [CrossRef] [Green Version]
  183. Albini, A.; Tosetti, F.; Li, V.W.; Noonan, D.M.; Li, W.W. Cancer prevention by targeting angiogenesis. Nat. Rev. Clin. Oncol. 2012, 9, 498–509. [Google Scholar] [CrossRef]
  184. Mabeta, P. Paradigms of vascularization in melanoma: Clinical significance and potential for therapeutic targeting. Biomed. Pharmacother. 2020, 127, 110135. [Google Scholar] [CrossRef]
  185. Stacker, S.A.; Caesar, C.; Baldwin, M.E.; Thornton, G.E.; Williams, R.A.; Prevo, R.; Jackson, D.G.; Nishikawa, S.; Kubo, H.; Achen, M.G. VEGF-D promotes the metastatic spread of tumor cells via the lymphatics. Nat. Med. 2001, 7, 186–191. [Google Scholar] [CrossRef] [PubMed]
  186. Jour, G.; Ivan, D.; Aung, P.P. Angiogenesis in melanoma: An update with a focus on current targeted therapies. J. Clin. Pathol. 2016, 69, 472–483. [Google Scholar] [CrossRef] [PubMed]
  187. Kiyan, H.T.; Demirci, B.; Baser, K.H.; Demirci, F. The in vivo evaluation of anti-angiogenic effects of Hypericum essential oils using the chorioallantoic membrane assay. Pharm. Biol. 2014, 52, 44–50. [Google Scholar] [CrossRef] [PubMed]
  188. Mishra, H.; Mishra, P.K.; Iqbal, Z.; Jaggi, M.; Madaan, A.; Bhuyan, K.; Gupta, N.; Gupta, N.; Vats, K.; Verma, R.; et al. Co-Delivery of Eugenol and Dacarbazine by Hyaluronic Acid-Coated Liposomes for Targeted Inhibition of Survivin in Treatment of Resistant Metastatic Melanoma. Pharmaceutics 2019, 11, 163. [Google Scholar] [CrossRef] [Green Version]
  189. Yousefian Rad, E.; Homayouni Tabrizi, M.; Ardalan, P.; Seyedi, S.M.R.; Yadamani, S.; Zamani-Esmati, P.; Haghani Sereshkeh, N. Citrus lemon essential oil nanoemulsion (CLEO-NE), a safe cell-depended apoptosis inducer in human A549 lung cancer cells with anti-angiogenic activity. J. Microencapsul. 2020, 37, 394–402. [Google Scholar] [CrossRef]
  190. Farahpour, M.R.; Pirkhezr, E.; Ashrafian, A.; Sonboli, A. Accelerated healing by topical administration of Salvia officinalis essential oil on Pseudomonas aeruginosa and Staphylococcus aureus infected wound model. Biomed. Pharmacother. 2020, 128, 110120. [Google Scholar] [CrossRef]
  191. Santos-Júnior, L.; Oliveira, T.V.C.; Cândido, J.F.; Santana, D.S.; Pereira, R.N.F.; Pereyra, B.B.S.; Gomes, M.Z.; Lima, S.O.; Albuquerque-Júnior, R.L.C.; Cândido, E.A.F. Effects of the essential oil of Alpinia zerumbet (Pers.) B.L. Burtt & R.M. Sm. on healing and tissue repair after partial Achilles tenotomy in rats. Acta Cir. Bras. 2017, 32, 449–458. [Google Scholar] [PubMed]
  192. Kim, D.Y.; Won, K.J.; Yoon, M.S.; Hwang, D.I.; Yoon, S.W.; Park, J.H.; Kim, B.; Lee, H.M. Chrysanthemum boreale Makino essential oil induces keratinocyte proliferation and skin regeneration. Nat. Prod. Res. 2015, 29, 562–564. [Google Scholar] [CrossRef]
  193. Avola, R.; Granata, G.; Geraci, C.; Napoli, E.; Eleonora Graziano, A.C.; Cardile, V. Oregano (Origanum vulgare L.) essential oil provides anti-inflammatory activity and facilitates wound healing in a human keratinocytes cell model. Food Chem. Toxicol. 2020. [Google Scholar] [CrossRef] [PubMed]
  194. Labib, R.M.; Ayoub, I.M. Appraisal on the wound healing potential of Melaleuca alternifolia and Rosmarinus officinalis L. essential oil-loaded chitosan topical preparations. PLoS ONE 2019, 14, e0219561. [Google Scholar] [CrossRef] [PubMed]
  195. Park, J.H.; Park, G.M.; Kim, J.K. Zerumbone, Sesquiterpene Photochemical from Ginger, Inhibits Angiogenesis. Korean J. Physiol. Pharmacol. 2015, 19, 335–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Samad, N.A.; Abdul, A.B.; Rahman, H.S.; Rasedee, A.; Tengku Ibrahim, T.A.; Keon, Y.S. Zerumbone Suppresses Angiogenesis in HepG2 Cells through Inhibition of Matrix Metalloproteinase-9, Vascular Endothelial Growth Factor, and Vascular Endothelial Growth Factor Receptor Expressions. Pharmacogn. Mag. 2018, 13, S731–S736. [Google Scholar] [PubMed]
  197. Shojaei, S.; Kiumarsi, A.; Moghadam, A.R.; Alizadeh, J.; Marzban, H.; Ghavami, S. Perillyl Alcohol (Monoterpene Alcohol), Limonene. Enzymes 2014, 36, 7–32. [Google Scholar] [PubMed]
  198. Loutrari, H.; Hatziapostolou, M.; Skouridou, V.; Papadimitriou, E.; Roussos, C.; Kolisis, F.N.; Papapetropoulos, A. Perillyl alcohol is an angiogenesis inhibitor. J. Pharmacol. Exp. Ther. 2004, 311, 568–575. [Google Scholar] [CrossRef]
  199. Zuo, H.X.; Jin, Y.; Wang, Z.; Li, M.Y.; Zhang, Z.H.; Wang, J.Y.; Xing, Y.; Ri, M.H.; Jin, C.H.; Xu, G.H.; et al. Curcumol inhibits the expression of programmed cell death-ligand 1 through crosstalk between hypoxia-inducible factor-1alpha and STAT3 (T705) signaling pathways in hepatic cancer. J. Ethnopharmacol. 2020, 257, 112835. [Google Scholar] [CrossRef]
  200. Tanaka, R.; Fujisawa, Y.; Sae, I.; Maruyama, H.; Ito, S.; Hasegawa, N.; Sekine, I.; Fujimoto, M. Severe hepatitis arising from ipilimumab administration, following melanoma treatment with nivolumab. Jpn. J. Clin. Oncol. 2017, 47, 175–178. [Google Scholar] [CrossRef] [Green Version]
  201. Scarpati, G.D.; Fusciello, C.; Perri, F.; Sabbatino, F.; Ferrone, S.; Carlomagno, C.; Pepe, S. Ipilimumab in the treatment of metastatic melanoma: Management of adverse events. Oncotargets Ther. 2014, 7, 203–209. [Google Scholar] [CrossRef] [Green Version]
  202. Marques, H.M.C. A review on cyclodextrin encapsulation of essential oils and volatiles. Flavour Frag. J. 2010, 25, 313–326. [Google Scholar] [CrossRef]
  203. Zhou, X.; Yang, G.; Guan, F. Biological Functions and Analytical Strategies of Sialic Acids in Tumor. Cells 2020, 9, 273. [Google Scholar] [CrossRef] [Green Version]
  204. Wang, X.; Liu, R.; Zhu, W.; Chu, H.; Yu, H.; Wei, P.; Wu, X.; Zhu, H.; Gao, H.; Liang, J.; et al. UDP-glucose accelerates SNAI1 mRNA decay and impairs lung cancer metastasis. Nature 2019, 571, 127–131. [Google Scholar] [CrossRef] [PubMed]
  205. Kusuhara, M.; Urakami, K.; Masuda, Y.; Zangiacomi, V.; Ishii, H.; Tai, S.; Maruyama, K.; Yamaguchi, K. Fragrant environment with alpha-pinene decreases tumor growth in mice. Biomed. Res. 2012, 33, 57–61. [Google Scholar] [CrossRef] [Green Version]
  206. Kusuhara, M.; Maruyama, K.; Ishii, H.; Masuda, Y.; Sakurai, K.; Tamai, E.; Urakami, K. A Fragrant Environment Containing α-Pinene Suppresses Tumor Growth in Mice by Modulating the Hypothalamus/Sympathetic Nerve/Leptin Axis and Immune System. Integr. Cancer Ther. 2019, 18, 1534735419845139. [Google Scholar] [CrossRef] [PubMed]
  207. Lesgards, J.F.; Baldovini, N.; Vidal, N.; Pietri, S. Anticancer activities of essential oils constituents and synergy with conventional therapies: A review. Phytother. Res. 2014, 28, 1423–1446. [Google Scholar] [CrossRef]
  208. Effenberger-Neidnicht, K.; Schobert, R. Combinatorial effects of thymoquinone on the anti-cancer activity of doxorubicin. Cancer Chemother. Pharmacol. 2011, 67, 867–874. [Google Scholar] [CrossRef] [Green Version]
  209. Balavandi, Z.; Neshasteh-Riz, A.; Koosha, F.; Eynali, S.; Hoormand, M.; Shahidi, M. The Use of ss-Elemene to Enhance Radio Sensitization of A375 Human Melanoma Cells. Cell J. 2020, 21, 419–425. [Google Scholar] [PubMed]
  210. Benimetskaya, L.; Lai, J.C.; Khvorova, A.; Wu, S.; Hua, E.; Miller, P.; Zhang, L.M.; Stein, C.A. Relative Bcl-2 independence of drug-induced cytotoxicity and resistance in 518A2 melanoma cells. Clin. Cancer Res. 2004, 10, 8371–8379. [Google Scholar] [CrossRef] [Green Version]
  211. Senbanjo, L.T.; Chellaiah, M.A. CD44: A Multifunctional Cell Surface Adhesion Receptor Is a Regulator of Progression and Metastasis of Cancer Cells. Front. Cell Dev. Biol. 2017, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  212. Kaur, G.; Athar, M.; Alam, M.S. Eugenol precludes cutaneous chemical carcinogenesis in mouse by preventing oxidative stress and inflammation and by inducing apoptosis. Mol. Carcinog. 2010, 49, 290–301. [Google Scholar] [CrossRef]
  213. Pal, D.; Banerjee, S.; Mukherjee, S.; Roy, A.; Panda, C.K.; Das, S. Eugenol restricts DMBA croton oil induced skin carcinogenesis in mice: Downregulation of c-Myc and H-ras, and activation of p53 dependent apoptotic pathway. J. Dermatol. Sci. 2010, 59, 31–39. [Google Scholar] [CrossRef]
  214. Chaudhary, S.C.; Alam, M.S.; Siddiqui, M.S.; Athar, M. Chemopreventive effect of farnesol on DMBA/TPA-induced skin tumorigenesis: Involvement of inflammation, Ras-ERK pathway and apoptosis. Life Sci. 2009, 85, 196–205. [Google Scholar] [CrossRef] [PubMed]
  215. Manoharan, S.; Selvan, M.V. Chemopreventive potential of geraniol in 7,12-dimethylbenz(a) anthracene (DMBA) induced skin carcinogenesis in Swiss albino mice. J. Environ. Biol. 2012, 33, 255–260. [Google Scholar] [PubMed]
  216. Liu, Z.; Shen, C.; Tao, Y.; Wang, S.; Wei, Z.; Cao, Y.; Wu, H.; Fan, F.; Lin, C.; Shan, Y.; et al. Chemopreventive efficacy of menthol on carcinogen-induced cutaneous carcinoma through inhibition of inflammation and oxidative stress in mice. Food Chem. Toxicol. 2015, 82, 12–18. [Google Scholar] [CrossRef] [PubMed]
  217. Lluria-Prevatt, M.; Morreale, J.; Gregus, J.; Alberts, D.S.; Kaper, F.; Giaccia, A.; Powell, M.B. Effects of perillyl alcohol on melanoma in the TPras mouse model. Cancer Epidemiol. Biomark. Prev. 2002, 11, 573–579. [Google Scholar]
  218. Chaudhary, S.C.; Alam, M.S.; Siddiqui, M.S.; Athar, M. Perillyl alcohol attenuates Ras-ERK signaling to inhibit murine skin inflammation and tumorigenesis. Chem. Biol. Interact. 2009, 179, 145–153. [Google Scholar] [CrossRef]
  219. Aumeeruddy-Elalfi, Z.; Lall, N.; Fibrich, B.; Blom van Staden, A.; Hosenally, M.; Mahomoodally, M.F. Selected essential oils inhibit key physiological enzymes and possess intracellular and extracellular antimelanogenic properties in vitro. J. Food Drug Anal. 2018, 26, 232–243. [Google Scholar] [CrossRef] [Green Version]
  220. Horiba, H.; Nakagawa, T.; Zhu, Q.; Ashour, A.; Watanabe, A.; Shimizu, K. Biological Activities of Extracts from Different Parts of Cryptomeria japonica. Nat. Prod. Commun. 2016, 11, 1337–1342. [Google Scholar] [CrossRef] [Green Version]
  221. Yang, C.H.; Huang, Y.C.; Tsai, M.L.; Cheng, C.Y.; Liu, L.L.; Yen, Y.W.; Chen, W.L. Inhibition of melanogenesis by β-caryophyllene from lime mint essential oil in mouse B16 melanoma cells. Int. J. Cosmet. Sci. 2015, 37, 550–554. [Google Scholar] [CrossRef]
  222. El Khoury, R.; Michael Jubeli, R.; El Beyrouthy, M.; Baillet Guffroy, A.; Rizk, T.; Tfayli, A.; Lteif, R. Phytochemical screening and antityrosinase activity of carvacrol, thymoquinone, and four essential oils of Lebanese plants. J. Cosmet. Dermatol. 2019, 18, 944–952. [Google Scholar] [CrossRef]
  223. Ko, G.A.; Cho, S.K. Phytol suppresses melanogenesis through proteasomal degradation of MITF via the ROS-ERK signaling pathway. Chem. Biol. Interact. 2018, 286, 132–140. [Google Scholar] [CrossRef]
  224. Jeong, H.; Yu, S.M.; Kim, S.J. Inhibitory effects on melanogenesis by thymoquinone are mediated through the betacatenin pathway in B16F10 mouse melanoma cells. Int. J. Oncol. 2020, 56, 379–389. [Google Scholar] [PubMed]
  225. Nam, J.H.; Nam, D.Y.; Lee, D.U. Valencene from the Rhizomes of Cyperus rotundus Inhibits Skin Photoaging-Related Ion Channels and UV-Induced Melanogenesis in B16F10 Melanoma Cells. J. Nat. Prod. 2016, 79, 1091–1096. [Google Scholar] [CrossRef] [PubMed]
  226. Oh, T.I.; Jung, H.J.; Lee, Y.M.; Lee, S.; Kim, G.H.; Kan, S.Y.; Kang, H.; Oh, T.; Ko, H.M.; Kwak, K.C.; et al. Zerumbone, a Tropical Ginger Sesquiterpene of Zingiber officinale Roscoe, Attenuates alpha-MSH-Induced Melanogenesis in B16F10 Cells. Int. J. Mol. Sci. 2018, 19, 3149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Gandini, S.; Sera, F.; Cattaruzza, M.S.; Pasquini, P.; Abeni, D.; Boyle, P.; Melchi, C.F. Meta-analysis of risk factors for cutaneous melanoma: I. Common and atypical naevi. Eur. J. Cancer 2005, 41, 28–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Gandini, S.; Sera, F.; Cattaruzza, M.S.; Pasquini, P.; Picconi, O.; Boyle, P.; Melchi, C.F. Meta-analysis of risk factors for cutaneous melanoma: II. Sun exposure. Eur. J. Cancer 2005, 41, 45–60. [Google Scholar] [CrossRef] [PubMed]
  229. Gandini, S.; Sera, F.; Cattaruzza, M.S.; Pasquini, P.; Zanetti, R.; Masini, C.; Boyle, P.; Melchi, C.F. Meta-analysis of risk factors for cutaneous melanoma: III. Family history, actinic damage and phenotypic factors. Eur. J. Cancer 2005, 41, 2040–2059. [Google Scholar] [CrossRef]
  230. McKinney, A.J.; Holmen, S.L. Animal models of melanoma: A somatic cell gene delivery mouse model allows rapid evaluation of genes implicated in human melanoma. Chin. J. Cancer 2011, 30, 153–162. [Google Scholar] [CrossRef] [Green Version]
  231. Stratton, S.P.; Alberts, D.S.; Einspahr, J.G.; Sagerman, P.M.; Warneke, J.A.; Curiel-Lewandrowski, C.; Myrdal, P.B.; Karlage, K.L.; Nickoloff, B.J.; Brooks, C.; et al. A phase 2a study of topical perillyl alcohol cream for chemoprevention of skin cancer. Cancer Prev. Res. (Phila) 2010, 3, 160–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Stratton, S.P.; Saboda, K.L.; Myrdal, P.B.; Gupta, A.; McKenzie, N.E.; Brooks, C.; Salasche, S.J.; Warneke, J.A.; Ranger-Moore, J.; Bozzo, P.D.; et al. Phase 1 study of topical perillyl alcohol cream for chemoprevention of skin cancer. Nutr. Cancer 2008, 60, 325–330. [Google Scholar] [CrossRef]
  233. Nishigori, C.; Hattori, Y.; Toyokuni, S. Role of reactive oxygen species in skin carcinogenesis. Antioxid. Redox. Signal. 2004, 6, 561–570. [Google Scholar] [CrossRef]
  234. Alam, M.N.; Bristi, N.J.; Rafiquzzaman, M. Review on in vivo and in vitro methods evaluation of antioxidant activity. Saudi Pharm. J. 2013, 21, 143–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Cui, X.; Gong, J.; Han, H.; He, L.; Teng, Y.; Tetley, T.; Sinharay, R.; Chung, K.F.; Islam, T.; Gilliland, F.; et al. Relationship between free and total malondialdehyde, a well-established marker of oxidative stress, in various types of human biospecimens. J. Thorac. Dis. 2018, 10, 3088–3097. [Google Scholar] [CrossRef] [PubMed]
  236. Rahiman, N.; Akaberi, M.; Sahebkar, A.; Emami, S.A.; Tayarani-Najaran, Z. Protective effects of saffron and its active components against oxidative stress and apoptosis in endothelial cells. Microvasc. Res. 2018, 118, 82–89. [Google Scholar] [CrossRef] [PubMed]
  237. Onyebuchi, C.; Kavaz, D. Chitosan And N, N, N-Trimethyl Chitosan Nanoparticle Encapsulation Of Ocimum Gratissimum Essential Oil: Optimised Synthesis, In Vitro Release And Bioactivity. Int. J. Nanomed. 2019, 14, 7707–7727. [Google Scholar] [CrossRef] [Green Version]
  238. Do Nascimento, K.F.; Moreira, F.M.F.; Alencar Santos, J.; Kassuya, C.A.L.; Croda, J.H.R.; Cardoso, C.A.L.; Vieira, M.D.C.; Gois Ruiz, A.L.T.; Ann Foglio, M.; de Carvalho, J.E.; et al. Antioxidant, anti-inflammatory, antiproliferative and antimycobacterial activities of the essential oil of Psidium guineense Sw. and spathulenol. J. Ethnopharmacol. 2018, 210, 351–358. [Google Scholar] [CrossRef]
  239. Manjamalai, A.; Grace, B. Chemotherapeutic effect of essential oil of Wedelia chinensis (Osbeck) on inducing apoptosis, suppressing angiogenesis and lung metastasis in C57BL/6 mice model. J. Cancer Sci. Ther. 2013, 5, 271–281. [Google Scholar] [CrossRef]
  240. Lambert, M.W.; Maddukuri, S.; Karanfilian, K.M.; Elias, M.L.; Lambert, W.C. The physiology of melanin deposition in health and disease. Clin. Dermatol. 2019, 37, 402–417. [Google Scholar] [CrossRef]
  241. Jackett, L.A.; Scolyer, R.A. A Review of Key Biological and Molecular Events Underpinning Transformation of Melanocytes to Primary and Metastatic Melanoma. Cancers 2019, 11, 2041. [Google Scholar] [CrossRef] [Green Version]
  242. Liu, G.S.; Peshavariya, H.; Higuchi, M.; Brewer, A.C.; Chang, C.W.; Chan, E.C.; Dusting, G.J. Microphthalmia-associated transcription factor modulates expression of NADPH oxidase type 4: A negative regulator of melanogenesis. Free Radic. Biol. Med. 2012, 52, 1835–1843. [Google Scholar] [CrossRef]
  243. Gillbro, J.M.; Olsson, M.J. The melanogenesis and mechanisms of skin-lightening agents--existing and new approaches. Int. J. Cosmet. Sci. 2011, 33, 210–221. [Google Scholar] [CrossRef]
  244. Pillaiyar, T.; Manickam, M.; Jung, S.H. Downregulation of melanogenesis: Drug discovery and therapeutic options. Drug Discov. Today 2017, 22, 282–298. [Google Scholar] [CrossRef] [PubMed]
  245. Nakagawa, T.; Zhu, Q.; Ishikawa, H.; Ohnuki, K.; Kakino, K.; Horiuchi, N.; Shinotsuka, H.; Naito, T.; Matsumoto, T.; Minamisawa, N. Multiple uses of essential oil and by-products from various parts of the Yakushima native Cedar (Cryptomeria Japonica). J. Wood Chem. Technol. 2016, 36, 42–55. [Google Scholar] [CrossRef]
  246. Zaidi, K.U.; Khan, F.N.; Ali, S.A.; Khan, K.P. Insight into Mechanistic Action of Thymoquinone Induced Melanogenesis in Cultured Melanocytes. Protein Pept. Lett. 2019, 26, 910–918. [Google Scholar] [CrossRef] [PubMed]
  247. Dyer, J.; Cleary, L.; Ragsdale-Lowe, M.; McNeill, S.; Osland, C. The use of aromasticks at a cancer centre: A retrospective audit. Complement. Ther. Clin. Pract. 2014, 20, 203–206. [Google Scholar] [CrossRef]
  248. Boehm, K.; Büssing, A.; Ostermann, T. Aromatherapy as an adjuvant treatment in cancer care—A descriptive systematic review. Afr. J. Tradit. Complement. Altern. Med. 2012, 9, 503–518. [Google Scholar] [CrossRef] [PubMed]
  249. Maddocks-Jennings, W.; Wilkinson, J.M.; Cavanagh, H.M.; Shillington, D. Evaluating the effects of the essential oils Leptospermum scoparium (manuka) and Kunzea ericoides (kanuka) on radiotherapy induced mucositis: A randomized, placebo controlled feasibility study. Eur. J. Oncol. Nurs. 2009, 13, 87–93. [Google Scholar] [CrossRef]
  250. Reis, D.; Jones, T.T. Frankincense Essential Oil as a Supportive Therapy for Cancer-Related Fatigue: A Case Study. Holist. Nurs. Pract. 2018, 32, 140–142. [Google Scholar] [CrossRef]
  251. Pimenta, F.C.; Alves, M.F.; Pimenta, M.B.; Melo, S.A.; de Almeida, A.A.; Leite, J.R.; Pordeus, L.C.; Diniz Mde, F.; de Almeida, R.N. Anxiolytic Effect of Citrus aurantium L. on Patients with Chronic Myeloid Leukemia. Phytother. Res. 2016, 30, 613–617. [Google Scholar] [CrossRef]
  252. Mapp, C.P.; Hostetler, D.; Sable, J.F.; Parker, C.; Gouge, E.; Masterson, M.; Willis-Styles, M.; Fortner, C.; Higgins, M. Peppermint Oil: Evaluating Efficacy on Nausea in Patients Receiving Chemotherapy in the Ambulatory Setting. Clin. J. Oncol. Nurs. 2020, 24, 160–164. [Google Scholar] [CrossRef] [PubMed]
  253. Tayarani-Najaran, Z.; Talasaz-Firoozi, E.; Nasiri, R.; Jalali, N.; Hassanzadeh, M. Antiemetic activity of volatile oil from Mentha spicata and Mentha x piperita in chemotherapy-induced nausea and vomiting. Ecancermedicalscience 2013, 7, 290. [Google Scholar]
  254. Hamzeh, S.; Safari-Faramani, R.; Khatony, A. Effects of Aromatherapy with Lavender and Peppermint Essential Oils on the Sleep Quality of Cancer Patients: A Randomized Controlled Trial. Evid. Based Complement. Altern. Med. 2020, 2020, 7480204. [Google Scholar] [CrossRef] [PubMed]
  255. Re, L.; Barocci, S.; Sonnino, S.; Mencarelli, A.; Vivani, C.; Paolucci, G.; Scarpantonio, A.; Rinaldi, L.; Mosca, E. Linalool modifies the nicotinic receptor–ion channel kinetics at the mouse neuromuscular junction. Pharmacol. Res. 2000, 42, 177–181. [Google Scholar] [CrossRef] [PubMed]
  256. Gedney, J.J.; Glover, T.L.; Fillingim, R.B. Sensory and affective pain discrimination after inhalation of essential oils. Psychosom. Med. 2004, 66, 599–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Blackburn, L.; Achor, S.; Allen, B.; Bauchmire, N.; Dunnington, D.; Klisovic, R.B.; Naber, S.J.; Roblee, K.; Samczak, A.; Tomlinson-Pinkham, K.; et al. The Effect of Aromatherapy on Insomnia and Other Common Symptoms Among Patients With Acute Leukemia. Oncol. Nurs. Forum 2017, 44, E185–E193. [Google Scholar] [CrossRef]
  258. Evans, A.; Malvar, J.; Garretson, C.; Pedroja Kolovos, E.; Baron Nelson, M. The Use of Aromatherapy to Reduce Chemotherapy-Induced Nausea in Children With Cancer: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Pediatr. Oncol. Nurs. 2018, 35, 392–398. [Google Scholar] [CrossRef]
  259. Lua, P.L.; Salihah, N.; Mazlan, N. Effects of inhaled ginger aromatherapy on chemotherapy-induced nausea and vomiting and health-related quality of life in women with breast cancer. Complement. Ther. Med. 2015, 23, 396–404. [Google Scholar] [CrossRef]
  260. Tamaki, K.; Fukuyama, A.K.; Terukina, S.; Kamada, Y.; Uehara, K.; Arakaki, M.; Yamashiro, K.; Miyashita, M.; Ishida, T.; McNamara, K.M. Randomized trial of aromatherapy versus conventional care for breast cancer patients during perioperative periods. Breast Cancer Res. Treat. 2017, 162, 523–531. [Google Scholar] [CrossRef]
  261. Chen, T.H.; Tung, T.H.; Chen, P.S.; Wang, S.H.; Chao, C.M.; Hsiung, N.H.; Chi, C.C. The Clinical Effects of Aromatherapy Massage on Reducing Pain for the Cancer Patients: Meta-Analysis of Randomized Controlled Trials. Evid. Based Complement. Altern. Med. 2016, 2016, 9147974. [Google Scholar] [CrossRef] [Green Version]
  262. Graham, P.H.; Browne, L.; Cox, H.; Graham, J. Inhalation aromatherapy during radiotherapy: Results of a placebo-controlled double-blind randomized trial. J. Clin. Oncol. 2003, 21, 2372–2376. [Google Scholar] [CrossRef]
  263. Mo, H.; Jeter, R.; Bachmann, A.; Yount, S.T.; Shen, C.L.; Yeganehjoo, H. The Potential of Isoprenoids in Adjuvant Cancer Therapy to Reduce Adverse Effects of Statins. Front. Pharmacol. 2018, 9, 1515. [Google Scholar] [CrossRef] [Green Version]
  264. Zárybnický, T.; Boušová, I.; Ambrož, M.; Skálová, L. Hepatotoxicity of monoterpenes and sesquiterpenes. Arch. Toxicol. 2018, 92, 1–13. [Google Scholar] [CrossRef] [PubMed]
  265. Artini, M.; Patsilinakos, A.; Papa, R.; Bozovic, M.; Sabatino, M.; Garzoli, S.; Vrenna, G.; Tilotta, M.; Pepi, F.; Ragno, R.; et al. Antimicrobial and Antibiofilm Activity and Machine Learning Classification Analysis of Essential Oils from Different Mediterranean Plants against Pseudomonas aeruginosa. Molecules 2018, 23, 482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. EOs and their components reduce tumor angiogenesis, lymphangiogenesis (A) and tumor metastasization, by targeting proteins responsible for tumor dissemination (B) and tumor-promoting inflammation (C). Angiopoietin 2 (ANGPT2), vascular endothelial growth factor (VEGF), hypoxia responsive factor 1α (HIF-1α), lymphatic vessel endothelial receptor (LYVE-1), cluster of differentiation 31 (CD31), cluster of differentiation 34 (CD34), vascular endothelial cadherin (VE-cadherin), fibroblast growth factor 2 (FGF-2), phospho vascular endothelial growth factor receptor 2 (pVEGFR2), phospho fibroblast growth factor receptor-1 (pFGFR1), krüppel-like factor 4 (KFL4), programmed death-ligand 1 (PD-L1), tissue inhibitor of metalloproteinase-1 (TIMP-1) and -2 (TIMP-2), extracellular signal-regulated kinase-1 (ERK-1) and -2 (ERK-2), focal adhesion kinase (FAK), growth factor receptor-bound protein 2 (Grb2), urokinase-type plasminogen activator (uPA), uPA receptor (uPAR), metalloproteinases-2 (MMP-2) and -9 (MMP-9), nuclear factor kappa B (NF-kB), NLR family pyrin domain containing 3 (NLRP3), interleukin 1β (IL-1 β), interleukin 6 (IL-6), interleukin 2 (IL-2), tumor necrosis factor-α (TNF-α), transforming growth factor β1 (TGF-β1), macrophage colony stimulating factor (M-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), monocyte chemoattractant protein-1 (MCP-1), keratinocyte chemoattractant (KC). Proteins that are upregulated by EOs or their components are reported in green, proteins that are downregulated by EOs or their components are reported in red. Parts of the figure are drawn using pictures from Servier Medical Art (https://smart.servier.com).
Figure 1. EOs and their components reduce tumor angiogenesis, lymphangiogenesis (A) and tumor metastasization, by targeting proteins responsible for tumor dissemination (B) and tumor-promoting inflammation (C). Angiopoietin 2 (ANGPT2), vascular endothelial growth factor (VEGF), hypoxia responsive factor 1α (HIF-1α), lymphatic vessel endothelial receptor (LYVE-1), cluster of differentiation 31 (CD31), cluster of differentiation 34 (CD34), vascular endothelial cadherin (VE-cadherin), fibroblast growth factor 2 (FGF-2), phospho vascular endothelial growth factor receptor 2 (pVEGFR2), phospho fibroblast growth factor receptor-1 (pFGFR1), krüppel-like factor 4 (KFL4), programmed death-ligand 1 (PD-L1), tissue inhibitor of metalloproteinase-1 (TIMP-1) and -2 (TIMP-2), extracellular signal-regulated kinase-1 (ERK-1) and -2 (ERK-2), focal adhesion kinase (FAK), growth factor receptor-bound protein 2 (Grb2), urokinase-type plasminogen activator (uPA), uPA receptor (uPAR), metalloproteinases-2 (MMP-2) and -9 (MMP-9), nuclear factor kappa B (NF-kB), NLR family pyrin domain containing 3 (NLRP3), interleukin 1β (IL-1 β), interleukin 6 (IL-6), interleukin 2 (IL-2), tumor necrosis factor-α (TNF-α), transforming growth factor β1 (TGF-β1), macrophage colony stimulating factor (M-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), monocyte chemoattractant protein-1 (MCP-1), keratinocyte chemoattractant (KC). Proteins that are upregulated by EOs or their components are reported in green, proteins that are downregulated by EOs or their components are reported in red. Parts of the figure are drawn using pictures from Servier Medical Art (https://smart.servier.com).
Cancers 12 02650 g001
Figure 2. EOs and their components reduce melanogenesis and oxidation through interconnected mechanisms. Superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase (CAT), glutathione (GSH), reactive oxygen species (ROS), α-melanocyte stimulating hormone (α-MSH), melanocortin 1 receptor (MC1R), adenylyl cyclase (AC), stem cell factor (SCF), microphthalmia-associated transcription factor (MITF), tyrosinase (TYR), tyrosinase-related protein -1 (TRP-1) and -2 (TRP-2), glycogen synthase kinase 3β (GSK3β), c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK). Proteins that are upregulated by EOs or their components are reported in green, proteins that are downregulated by EOs or their components are reported in red. Parts of the figure are drawn using pictures from Servier Medical Art (https://smart.servier.com).
Figure 2. EOs and their components reduce melanogenesis and oxidation through interconnected mechanisms. Superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase (CAT), glutathione (GSH), reactive oxygen species (ROS), α-melanocyte stimulating hormone (α-MSH), melanocortin 1 receptor (MC1R), adenylyl cyclase (AC), stem cell factor (SCF), microphthalmia-associated transcription factor (MITF), tyrosinase (TYR), tyrosinase-related protein -1 (TRP-1) and -2 (TRP-2), glycogen synthase kinase 3β (GSK3β), c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK). Proteins that are upregulated by EOs or their components are reported in green, proteins that are downregulated by EOs or their components are reported in red. Parts of the figure are drawn using pictures from Servier Medical Art (https://smart.servier.com).
Cancers 12 02650 g002
Table 1. Chemical composition of essential oils (EOs) active against melanoma models.
Table 1. Chemical composition of essential oils (EOs) active against melanoma models.
Plant Name from Which EOs Were ExtractedPlant Common Name Plant Family NameMain EO Chemical Components Reference
Achillea millefoliumYarrow, common yarrow, thousand-leafAsteraceaeArtemisia ketone (14.92%), camphor (11.64%), linalyl acetate (11.51%), 1,8-cineole (10.15%)[23]
Alpinia zerumbetLight Galangal, shell gingerZingiberaceaeγ-Terpinene (14.5%), cineole (13.8%), p-cymene (13.5%), sabinene (12.5%), terpinen-4-ol (11.9%), caryophyllene oxide (4.96%), methyl cinnamate (4.24%), caryophyllene (2.4%), γ-terpineol (1.28%)[24]
Annona vepretorumAraticum, pinha da caatinga, araticum-da-BahiaAnnonaceaeBicyclogermacrene (35.7%), spathulenol (18.89%), α-phellandrene (8.08%), α-pinene (2.18%), o-cymene (6.24%)[25]
Anthemis wiedemanniana-Asteraceae9,12-Octadecadienoic acid (12.2%), hexadecanoic acid (10.5%), hexahydrofarnesyl acetone (8.3%), 1,8-cineol (6.2%), carvacrol (5.8%)[26]
Artemisia anomala-Asteraceae or CompositaeCamphor (18.3%), 1,8-cineole (17.3%), β-caryophyllene oxide (12.7%), borneol (9.5%)[27]
Artemisia argyi-Asteraceae or CompositaeCaryophyllene (10.19%), eucalyptol (23.66%)[28]
Atriplex undulata-Chenopodiaceaep-Acetanisole (28.1%), β-damascenone (9.3%), β-ionone (5.1%), viridiflorene (4.7%), 3-oxo-α-ionol (2.2%)[29]
Casearia lasiophylla-SalicaceaeGermacrene D (18.6%), E-caryophyllene (14.7%), δ-cadinene (6.2%), α-cadinol (5.4%)[30]
Chrysanthemum boreale Makino-AsteraceaeGermacrene D (10.6–34.9%), β-caryophyllene (10.8%), (–)-camphor (10.8–18.0%), β-thujone (11.7%), α-thujone (9.8%)[31]
Cinnamomum cassia-LauraceaeCis-2-methoxycinnamic acid (43.06%), cinnamaldehyde (42.37%)[32]
Cinnamomum zeylanicum-LauraceaeEugenol (70%), β-caryophyllene (2.4%)[33]
Citrus bergamiaAcid lemonRutaceaeLimonene (38.1%), linalyl acetate (28.9%), γ-terpinene (7.3%), linalool (6.4%), β-pinene (5.4%), bergapten (1.7%)[34]
Citrus medicaCitronRutaceaeLimonene (35.4%), γ-terpinene (24.5%), geranial (5.5%), neral (4.4%), β-pinene (2.6%), α-pinene (2.5%), β-myrcene (2.1%), terpinen-4-ol (1.5%)[34]
Cuminum cyminumCumin-jeeraApiaceae or UmbelliferaeCuminaldehyde (39.48%), γ-terpinene (15.21%), O-cymene (11.82%), β-pinene (11.13%), 2-caren-10-al (7.93%), trans-carveol (4.49%), and myrtenal (3.5%)[35]
Curcuma aromaticaWild turmericZingiberaceae8,9-Dehydro-9- formyl-cycloisolongifolene (2.66–36.83%), germacrone (4.31–16.53%), ar-turmerone (2.52–17.69%), turmerone (2.62–18.38%), ermanthin (0.75–13.26%), β-sesquiphyllandrene (0.33–11.32%), ar-curcumene (0.29–10.52%)[36]
Curcuma kwangsiensisMango-gingerZingiberaceae8,9-Dehydro-9-formyl-cycloisolongifolene (2.37–42.59%), germacrone (6.53–22.20%), L-camphor (0.19–6.12%)[37]
Curcuma zedoariaKua-zedoaryZingiberaceae8,9-Dehydro-9-formyl-cycloisolongifolene (60%), 6-ethenyl-4,5,6,7-tetra-hydro-3,6-dimethyl-5-isopropenyl-trans-benzofuran (12%)[38]
Dalbergia pinnataLaleng-chaliFabaceaeElemicin (91.06%), methyl eugenol (3.69%), 4-allyl-2,6-dimethoxyphenol (1.16%), whiskey lactone (0.55%)[39]
Eryngium amethystinum-ApiaceaeGermacrene D (56.7%), β-elemene (4.7%), bicyclogermacrene (3.3%), α-copaene (2.2%), (E)-caryophyllene (1.9%), germacrene B (1.8%), germacra-4(15),5,10(14)-trien-1-α-ol (1.7%), cadin-4-en-10-ol (1.6%)[40]
Eryngium campestreEryngo,
field eryngo,
sea-holly
ApiaceaeGermacrene D (13.8%), allo-aromadendrene (7.7%), spathulenol (7.0%), ledol (5.7%), cadin-4-en-10-ol (3.9%), γ-cadinene (3.6%), epi-α-muurolol (2.1%), germacra-4(15),5,10(14)-trien-1-α-ol (2.0%), δ-cadinene (1.9%), caryophyllene oxide (1.5%)[40]
Eucalyptus camaldulensisMurray red gum, red gum, red river gumMyrtaceae1,8-Cineole (23.9%), α-eudesmol (11.6%), γ-eudesmol (8.0%), and elemol (5.0%)[41]
Eugenia cuspidifolia-MyrtaceaeCaryophyllene oxide (57.46%), α-copaene (3.75%)[42]
Eugenia tapacumensis-MyrtaceaeCaryophyllene oxide (55.95%), α-copaene (13.67%)[42]
Eugenia unifloraBrazil cherryMyrtaceaeCurzerene (13.4–50.6%), selina-1,3,7(11)-trien-2-one (18.1–43.1%), selina-1,3,7(11)-trien-2-one epoxidem(16.0–30.4%), germacrene B (5.0–18.4%), caryophyllene oxide(1.2–18.1%), (E)-caryophyllene (0.3–9.1%), β-elemene (3.5–8.9%), γ-elemene (2.0–7.8%)[43]
Glechoma hederaceaGround ivy, field balm, gill over the ground, runaway robinLamiaceae or LabiataeTrans-3-pinanone (41.4%), 4,5,6,7-tetrahydro-5-isopropenyl-3,6-β-dimethyl-6-α-vinylbenzofuran (10.8%), β-caryophyllene (10.2%), and spathulenol (4.3%)[44]
Helichrysum microphyllum-Asteraceae or CompositaeNeryl acetate (18.2%), rosifoliol (11.3%), δ-cadinene (8.4%), γ-cadinene (6.7%)[45]
Heracleum sphondyliumCow parsnip, eltrotApiaceae or UmbelliferaeOctyl acetate (54.9–60.2%), octyl butyrate (10.1–13.4%)[46]
Hypericum hircinum-HypericaceaeCis-β-guaiene (29.3%), δ-selinene (11.3%), isolongifolan-7-α-ol (9.8%), (E)-caryophyllene (7.2%)[47]
Laurus nobilisBay Tree, sweet bay, Grecian Laurel, true laurelLauraceae1,8-cineole (35.15%)[48]
Lavandula augustifoliaEnglish lavender, true lavenderLamiaceae or Labiataeα-Pipene, β-pipene, camphene, eucalyptol, D-limonene[49]
Lippia gracilis-VerbenaceaeThymol (55.50%), p-cymene (10.80%), γ-terpinene (5.53%), myrcene (4.03%)[50]
Liriodendron tulipiferaTulip tree, tulip poplar, yellow poplar, canary whitewoodMagnoliaceae(Z)-β-Ocimene (12.5–25.2%), (E)-β-ocimene (3.7–6.8%), β-elemene (16.4–17.1%), germacrene D (18.9–27.2%)[51]
Melaleuca alternifoliaTea TreeMyrtaceaeTerpinen-4-ol (42.35%), γ-terpinene (20.65%), α-terpinene (9.76%)[52]
Melaleuca quinquenervia-Myrtaceae1,8-Cineole (21.06%), α-pinene (15.93%), viridiflorol (14.55%), α-terpineol (13.73%)[53]
Mentha aquatica-Lamiaceae or Labiataeβ-Ocimene (22.18%), β-pinene (15.41%), 1,8-cineole (12.87%), α-pinene (10.49%)[54]
Myrcia laruotteana-Myrtaceaeα-Bisabolol (23.6%), α-bisabolol oxide B (11.5%)[55]
Myristica fragransMace, nutmegMyristicaceaeMyristicin, limonene, eugenol and terpinen-4-ol[56]
Nectandra leucantha-LauraceaeBicyclogermacrene (28.44%), germacrene A (7.34%)[57]
Ocimum basilicumSweet basil, common basil, thai basil, tropical basilLamiaceae or Labiatae1,8 Cineole (11.0%), linalool (42.5%), estragole (33.1%)[58]
Ocimum gratissimumAfrican basil,
east Indian basil,
russian basil, shrubby basil
Lamiaceae or LabiataeEugenol (54.0%), 1,8 cineole (21.6%), β-selinene (5.5%), β-caryophyllene (5.3%), (Z)-ocimene (4.0%)[58]
Ocimum micranthum-Lamiaceae or LabiataeEugenol (64.8%), β-caryophyllene (14,3%), bicyclogermacrene (8.1%)[58]
Ocimum tenuiflorumSacred basilLamiaceae or LabiataeEugenol (59.4%), β-caryophyllene (29.4%), germacrene A (8.1%)[58]
Origanum ehrenbergii-Lamiaceae or LabiataeCarvacrol, thymoquinone[59]
Origanum syriacumBible hyssopLamiaceae or LabiataeCarvacrol, thymoquinone[59]
Perilla frutescensShiso, beefsteakplant, spreading beefsteak plantLamiaceae or Labiataeisoegomaketone[60]
Piper aleyreanum-Piperaceaeβ-Elemene (16.3%), bicyclogermacrene (9.2%), δ-elemene (8.2%), germacrene D (6.9%), β-caryophyllene (6.2%), spathulenol (5.2%)[61]
Piper cernuum-Piperaceaeα-Pinene, camphene, limonene, carvacrol, tymol, myrcene, p-cymene, aterpineol, linalol[62]
Piper klotzschianum-PiperaceaeGermacrene D (7.3–22.8%), bicyclogermacrene (13.4–21.6%), E)-caryophyllene (11.9–16.8%), β-pinene (2.3–27.2%), α-pinene (1.4–7.2%)[63]
Pistacia lentiscusChios mastictree, aroeira,
lentiscus,
lentisk,
mastic,
mastictree
AnacardiaceaePerillyl alcohol[64]
Pituranthos tortuosus-ApiaceaeSabinene (24.24%), α-pinene (17.98%), limonene (16.12%), and terpinen-4-ol (7.21%)[65,66]
Plectranthus amboinicusCountry borage, Indian borageLamiaceae or LabiataeCarvacrol thymol, cis-caryophyllene, trans-caryophyllene, and p-cymene[67]
Pomelo peel-RutaceaeLimonene (55.92%), β-myrcene (31.17%), β-pinene (3.16%), ocimene (1.42%), β-copaene (1.24%)[68]
Porcelia macrocarpa-AnnonaceaeGermacrene D (47%), bicyclogermacrene (37%), verbanyl acetate (0.5%), phytol (1.2%)[69]
Pterodon emarginatusFaveiro, sucupira, sucupira-brancaFabaceaeβ-Elemene (15.3%), trans-caryophyllene (35.9%), α-humulene (6.8%), germacrene-D (9.8%), bicyclogermacrene (5.5%), spathulenol (5.9%)[70]
Salvia aurea-Lamiaceae or LabiataeCaryophyllene oxide (12.5%), α-amorphene (12.0%), aristolone (11.4%), aro-madendrene (10.7%), elemenone (6.0%)[71]
Salvia breacteata-Lamiaceae or LabiataeCaryophyllene oxide (16.6%)[72]
Salvia judaica-Lamiaceae or LabiataeCaryophyllene oxide (12.8%)[71]
Salvia libanotica-Lamiaceae or LabiataeCineole (57.4%), camphor (8.4%), β-pinene (5.1%), α-pinene (3.9%), camphene (3.0%)[73]
Salvia officinalisSage, kitchen sage, small leaf sage, garden sageLamiaceae or LabiataeCaryophyllene (25.634%), camphene (14.139%), eucalyptol (13.902%)[74]
Salvia rubifolia-Lamiaceae or Labiataeγ-Muurolene (11.8%)[72]
Salvia verbenacaWild claryLamiaceae or LabiataeHexadecanoic acid (11–23.1%), Z)-9-octadecenoic acid (5.6–11.1%), benzaldehyde (1.1–7.3%)[75]
Salvia viscosa-Lamiaceae or LabiataeCaryophyllene oxide (12.7%)[71]
Santalum albumWhite sandal tree, sandalwood, sandal tree, sandalSantalaceaeα-Santalol (61%), β-santalol (28%)[76]
Satureja hortensisSummer savoryLamiaceae or Labiataeγ-Terpinene (37.862%), o-cymene (15.113%), thymol (13.491%), carvacrol (13.225%)[77]
Schinus terebinthifolius RaddiBrazilian pepper treeAnacardiaceaeβ-Longipinene (8.1%), germacrene D (23.8%), biclyclogermacrene (15.0%), α-pinene (5.7%), β-pinene (9.1%)[78]
Stachys germanicaDowny woundwort, German hedgenettleLamiaceae or Labiatae(Z,Z,Z)-9,12,15-octa-decatrienoic acid methyl ester (33.3%), exadecanoic acid (22.1%)[79]
Stachys parviflora-Lamiaceae or Labiataeα-Terpenyl acetate (23.6%), β-caryophyllene (16.8%), bicyclogermacrene (9.3%), spathulenol (4.9%), α-pinene (4.2%)[80]
Syzygium aromaticumClove, Zanzibar redheadMyrtaceaeEugenol (61%), (β-carophillene 5.7%)[33,81]
Tagetes erectaAfrican marigold, Aztec marigold, big marigold, American marigoldAsteraceae or CompositaeLimonene (10.4%), α-terpinolene (18.1%), (E)-ocimenone (13.0%), dihydrotagetone (11.8%)[82]
Tanacetum macrophyllumTansy, rayed tansy, tansy chrysanthemumAsteraceae or CompositaeGermacrene D (6.9–30.9%), 10-epi-γ-eudesmol (3.9–13.5%), camphor (11.1%), linalool (0.6–10.8%), 1,8-cineole (5.5–8.8%)[83]
Thymus alternans-Lamiaceae or Labiatae(E)-Nerolidol (15.8–31.4%), germacrene D (6.7–7.4%), geranial (6.8–7.7%), (E)-β-ocimene (2.6–7.0%), linalool (1.7–6.4%), geraniol (3.3–6.2%), neral (4.9–5.4%)[84]
Thymus munbyanus-Lamiaceae or LabiataeBorneol (31.2–44.8%), camphor (5.7–13.6%), camphene (3.6–7.5%), 1,8-cineole (4.2–6.0%), germacrene D (3.1–5.0%)[85]
Thymus vulgarisEnglish thyme,
French thyme, garden thyme, thyme
Lamiaceae or Labiataeγ-Terpinene (68.415%), thymol (24.721%), caryophyllene (5.5%),
α-pinene (4.816%)
[74]
Tridax procumbensCoat buttons, coat-button, Mexican daisyAsteraceae or Compositaeα-Pipene, β-pinene, phellandrene, sabinene[86]
Vetiveria zizanioidesCuscus grass, khus-khus, khas-khas, vetiverPoaceaeCedr-8-en-13-ol (12.4%), α-amorphene (7.80%), β-vatirenene (5.94%), α-gurjunene (5.91%), dehydroaromadendrene (5.45%)[87]
Vitex NegundoCommon chaste tree, negundo, five keaved chaste tree, negundo chastetree, chaste treeLamiaceae or LabiataeSabinene (19.04%), caryophyllene (18.27%)[88]
Vitex TrifoliaIndian privet, Arabian lilac, Indian three-leaf vitex, hand of maryLamiaceae or Labiataeα-Pinene (11.38%), β-pinene (2.84%), sabinene (10.25%), eucaluptol (8.60%), camphene (12.69%), manoyl oxide (16.11%), abietatriene (9.03%)[89]
Wedelia chinensisChinese wedeliaAsteraceae or CompositaeCarvocrol, trans-caryophyllene[90]
Zornia brasiliensis-FabaceaeTrans-nerolidol (48.0%), germacrene D (13.9%), α-humulene (9.3%), trans-caryophyllene (8.4%), and (Z,E)-α-farnesene (7.3%)[91]
where not indicated, some plant common names and EOs composition were not available from the cited paper. EOs from Boswellia carterii, Citrus grandis, Citrus hystix, Citrus reticulate, Psiadia terebinthina were not included in the table as their chemical compositions were not reported in the corresponding cited articles.
Table 2. EOs and their components that have been demonstrated to affect in vitro or in vivo melanoma growth and metastasization.
Table 2. EOs and their components that have been demonstrated to affect in vitro or in vivo melanoma growth and metastasization.
Pathway AffectedPlant Name from Which EOs Were ExtractedEO Active ComponentsIn Vitro and In Vivo ModelsReference
In vitro cell proliferationAnnona VepretorumSpathulenol, o-cymene, α-pineneB16-F10[25]
Anthemis wiedemanniana-C32[26]
Artemisia anomala-BRO[27]
Casearia lasiophylla-UACC-62[30]
Citrus bergamiaBergaptenA375[34]
Citrus medicaLimoneneA375[34,126]
Coleus aromaticusCarvacrolA375[127]
Curcuma aromatica-B16[36]
Curcuma kwangsiensis-B16[37]
Curcuma zedoaria-B16-Bl6[38]
Eryngium amethystinum-A375[40]
Eryngium campestre-A375[40]
Eugenia cuspidifolia-SK-MEL-19[42]
Eugenia tapacumensis-SK-MEL-19[42]
Eugenia unifloraCurzereneSK-MEL-19[43]
Helichrysum microphyllum-A375[45]
Heracleum sphondyliumOctyl butyrateA375[46]
Hypericum hircinum-B16-F1[47]
Laurus nobilis-C32[48]
Lippia gracilis-B16-F10[50,128]
Liriodendron tulipiferaβ-ElemeneA375[51]
Melaleuca alternifoliaTerpinen-4-olA375, M14, B16-F10[52,129,130]
Melaleuca quinquenervia1,8-Cineole, α-Pipene, α-TerpineolB16[53]
Myrcia laruotteana-UACC-62[55]
Nectandra leucanthaBicyclogermacreneB16-F10-Nex2[57]
Perilla frutescensIsoegomaketoneB16[60]
Piper aleyreanum-SK-MEL-19[61]
Piper cernuumCampheneB16-F10-Nex2[62]
Piper klotzschianum-B16-F10[63]
Porcelia macrocarpa-B16-F10-Nex2[69]
Pterodon emarginatus-MeWo[70]
Salvia aurea-M14, A375, A2058[71]
Salvia bracteata-M14[72]
Salvia judaica-M14, A375, A2058[71]
Salvia officinalis-A375, M14, A2058, B164A5[74,101]
Salvia rubifolia-M14[72]
Salvia verbenaca-M14[75]
Salvia viscosa-M14, A375, A2058[71]
Satureja hortensis-B164A5, A375[77]
Schinus terebinthifolius Raddiα-Pipene, β-pipene, pipaneB16-F10-Nex2, A2058[78]
Stachys germanica-C32[79]
Stachys parviflora-B16-F10[80]
Syzygium aromaticumEugenolB16[81]
Tagetes erecta-B16-F10[82]
Tanacetum macrophyllum-A375[83]
Thuja occidentalisThujoneA375[131]
Thymus alternans-A375[84]
Thymus munbyanus-A375[85]
Thymus vulgaris-B164A5, A375[74]
Vitex TrifoliaAbietatrieneB16-F10[89]
CarvacrolSK-MEL-2[132]
CitralB16-F10, SK-MEL-147, UACC-257[133]
EugenolSK-MEL-2, A2058, SK-MEL-28, Sbcl2, WM3211, WM98-1, WM1205Lu, LCM-MEL
GR-MEL, 13443
[132,134,135,136]
FarnesolB16, B16-F10[106,109]
Farnesyl anthranilateB16[106,107]
Farnesyl-O-acetylhydroquinoneB16[106]
MentholA375[137]
NeridolB16[109]
ThymolSK-MEL-2[132]
ZerumboneCHL-1, A375[138,139]
β-CaryophylleneB16-F10[140]
In vitro tumor progression-associated functionsAlpinia zerumbet-HUVEC[141]
Eugenia unifloraCurzereneSK-MEL-19[43]
Melaleuca alternifoliaTerpinen-4-OlM14[142]
Pituranthos tortuosus-B16-F10[66]
Satureja hortensis-B164A5, A375[77]
MyrtenalB16-F0, B16-F10, SK-MEL-5[143]
ThujoneB16-F10[144]
ThymoquinoneB16-F10, A375[145]
ZerumboneCHL-1[138]
In vivo tumor growth and metastasizationAnnona VepretorumSpathulenol, o-cymene, α-pineneB16-F10 (C57BL/6J)[25]
Boswellia carterii-B16-F10 (C57BL/6)[146]
Curcuma zedoaria-B16-Bl6 (C57BL/6)[38]
Melaleuca alternifolia-B16-F10 (C57BL/6J)[147]
Perilla frutescensIsoegomaketoneB16 (C57BL/6N)[60]
Piper cernuumCampheneB16-F10-Nex2 (C57BL/6)[62]
Pituranthos tortuosus-B16-F10 (BALB/c)[65,66]
Plectranthus amboinicus-B16-F10 (C57BL/6)[67]
Salvia officinalisβ-Ursolic acidB16 (C57BL/6)[148]
Schinus terebinthifolius Raddiα-PipeneB16-F10-Nex2 (C57BL/6)[149]
Tridax procumbens-B16-F10 (C57BL/6)[86]
Zornia brasiliensis-B16-F10 (C57BL/6)[91]
EugenolB16 (B6D2F1)[135]
LimoneneB16-F10 (C57BL/6)[150]
MyrtenalB16-F10 (C57BL/6)[143]
Perillic AcidB16-F10 (C57BL/6)[150]
ThujoneB16-F10 (C57BL/6)[144]
ThymoquinoneB16-F10 (C57BL/6)[121,151]
α-PineneB16-F10-Nex2 (C57BL/6)[149]
β-CaryophylleneB16-F10 (C57BL/6N)[140]
β-ElemeneB16-F10 (C57BL/6)[120,152]
where not indicated, EO active components were not available from the cited articles. When referred to in vivo studies, the murine strain used is indicated in brackets. Human Umbilical Vein Endothelial Cells (HUVEC).
Table 3. EOs and their components that were demonstrated to affect cell cycle distribution, apoptosis, necrosis and autophagy of melanoma cells.
Table 3. EOs and their components that were demonstrated to affect cell cycle distribution, apoptosis, necrosis and autophagy of melanoma cells.
Pathway AffectedPlant Name from Which EOs Were ExtractedEO Active ComponentsIn Vitro and In Vivo Melanoma ModelsReference
Cell cycleMelaleuca alternifoliaTerpinen-4-olA375, B16[129,130]
Santalum albumα-SantolUACC-62[155]
EugenolA2058, WM1205Lu, Sbcl2, WM3211[134,135]
FarnesolB16[109]
NeridolB16[109]
ApoptosisAnnona VepretorumSpathulenol, o-cymene, α-pineneB16-F10[25]
Boswellia carterii B16-F10, FM94[146]
Coleus aromaticusCarvacrolA375[127]
Eugenia unifloraCurzereneSK-MEL-19[43]
Melaleuca alternifoliaTerpinen-4-OlA375,
M14
[52,129]
Perilla frutescensIsoegomaketoneB16[60]
Piper cernuumCampheneB16-F10-Nex2[62]
Pituranthos tortuosus-B16-F10[66]
Salvia aurea-M14[71]
Salvia bracteata-M14[72]
Salvia judaica-M14[71]
Salvia officinalis-A375, M14, A2058[101]
Salvia verbenaca-M14[75]
Salvia viscosa-M14[71]
Salvia rubifolia-M14[72]
Schinus terebinthifolius Raddiα-PipeneB16-F10-Nex2, A2058[78,149]
Thuja occidentalisThujoneA375[131]
Tridax procumbens-B16-F10 (C57BL/6)[86]
CitralB16-F10, SK-MEL-147, UACC-257[133]
EugenolA2058, SK-MEL-28[134,135,136]
LinaloolRPMI-7932[160]
MentholA375, G-361[161,162]
ThymoquinoneB16-F10[121]
ZerumboneCHL-1, A375[138,139]
β-CarophylleneB16-F10 (C57BL/6N)[140]
Necrosis and autophagyMelaleuca alternifoliaTerpinen-4-OlB16[130]
Salvia aurea-M14[71]
Salvia bracteata-M14[72]
Salvia judaica-M14[71]
Salvia viscosa-M14[71]
Salvia rubifolia-M14[72]
CitralB16-F10[133]
where not indicated, EO active components were not available from the cited articles. When referred to in vivo studies, the murine strain used is indicated in brackets.
Table 4. EOs and their components that have been demonstrated to affect in vitro and in vivo angiogenesis and lymphangiogenesis.
Table 4. EOs and their components that have been demonstrated to affect in vitro and in vivo angiogenesis and lymphangiogenesis.
Plant Name from Which EOs Were ExtractedEO Active ComponentsIn Vitro and In Vivo ModelsReference
Citrus lemon-CAM[189]
Curcuma zedoaria-HUVEC, CAM, rat aortic ring assay, B16-Bl6 (C57BL/6)[38]
Hypericum perforatum-EAhy.926[187]
Myristica fragrans-EAhy.926[56]
Pistacia lentiscus-B16[64]
Plectranthus amboinicus-B16-F10 (C57BL/6)[67]
Salvia officinalis-Infected wound model (BALB/c)[190]
Tridax procumbens-B16-F10 (C57BL/6)[86]
CurcumolHUVEC[199]
EugenolEAhy.926[188]
Perillyl AlcoholBLMVEC, HUVEC, B16-F10[198]
ZerumboneHUVEC, CAM, rat aortic ring assay[195,196]
β-CaryophylleneB16-F10 (C57BL/6N)[140]
β-ElemeneCAM, rat aortic ring assay, B16-F10 (C57BL/6)[120,152]
Human Umbilical Vein Endothelial Cells (HUVEC), transformed human umbilical vein endothelial cells produced by fusion of A549/8 lung adenocarcinoma with human umbilical endothelial cells (EAhy.926), Bovine Lung Microvascular Endothelial Cells (BLMVEC), chick embryo chorioallantoic membrane (CAM). Where not reported, EO active components were not available in the cited paper. When referrring to in vivo studies, the murine strain used is indicated in brackets.
Table 5. Effect of EOs and their components in the sensitization of antitumor agents, chemoprevention and melanogenesis.
Table 5. Effect of EOs and their components in the sensitization of antitumor agents, chemoprevention and melanogenesis.
Pathway AffectedPlant Name from Which EOs Were ExtractedEO Active ComponentsIn Vitro and In Vivo ModelsReference
Sensitization of antitumor agents EugenolSK-MEL-28, B16-F10[188]
ThymoquinoneB16-F10[121]
β-ElemeneA375[209]
ChemopreventionMentha aquatica-DMBA/TPA (FVB/NJ)[54]
Salvia libanotica-DMBA/TPA (BALB/c)[73]
Santalum albumα-SantolDMBA/TPA (CD1, SENCAR)[76]
EugenolDMBA/TPA, DMBA/croton oil (Swiss albino)[212,213]
FarnesolDMBA/TPA (Swiss albino)[214]
GeraniolDMBA/TPA (Swiss albino)[215]
LimoneneDMBA/TPA (Swiss albino)[177]
MentholDMBA/TPA (ICR)[216]
Perillyl AlcoholDMBA/TPras mut, DMBA/TPA (Swiss albino)[217,218]
MelanogenesisAchillea millefoliumLinalyl AcetateB16[23]
Alpinia zerumbet-B16-F10[24]
Artemisia argyi-B16-F10[28]
Cinnamomum cassiaCinnamaldehydeB16[32]
Cinnamomum zeylanicum-B16[33]
Citrus grandis-B16-F10[219]
Citrus hystrix-B16-F10[219]
Citrus reticulata-B16-F10[219]
Cryptomeria japonica-B16[220]
Chrysanthemum boreale MakinoCuminaldehydeB16-Bl6[31]
Dalbergia pinnata-Zebrafish embryos[39]
Eucalyptus camaldulensis-B16-F10[41]
Glechoma hederacea-B16[44]
Melaleuca quinquenervia1,8-Cineole, α-pipene, α-terpineolB16[53]
Mentha aquaticaβ-CaryophylleneB16-F10[221]
Origanum syriacumCarvacrolB16-F1[59,222]
Origanum ehrenbergiiCarvacrolB16-F1[59]
Pomelo peel-B16[68]
Psiadia terebinthina-B16-F10[219]
Syzygium aromaticumEugenolB16[81]
Vetiveria zizanioidesCedr-8-En-13-OlB16[87]
Vitex Negundo-B16-F10[88]
Vitex TrifoliaAbietatrieneB16-F10[89]
PhytolB16-F10[223]
ThymoquinoneB16-F10[224]
ValenceneB16-F10[225]
ZerumboneB16-F10[226]
where not indicated, EO active components were not available from the cited articles. When referred to in vivo studies, the murine strain used is indicated in brackets. 7,12-dimethylbenz[a]anthracene (DMBA), 12-O-tetradecanoylphobol-13-acetate (TPA), HaRas gene driven by the tyrosinase promoter (TPras).
Table 6. EOs and their components showing antioxidant effect in cell-free assay or in melanoma models.
Table 6. EOs and their components showing antioxidant effect in cell-free assay or in melanoma models.
Plant Name from Which EOs Were ExtractedEO Active ComponentsCell Free Assay and Melanoma ModelsReference
Achillea millefoliumLinalyl AcetateB16[23]
Alpinia zerumbet-DPPH, ABTS, nitric oxide, hydroxyl radical scavenging activity, xanthine oxidase[24]
Artemisia argyi-DPPH, ABTS, metal-ion chelation[28]
Atriplex undulata-Crocin bleaching inhibition, DPPH[29]
Cinnamomum cassiaCinnamaldehydeB16[32]
Chrysanthemum boreale Makino-DPPH, ABTS[31]
Cryptomeria japonica-B16[220]
Cuminum Cyminum-DPPH, superoxide anion radical-scavenging activity, β-carotene/linoleic acid[35]
Curcuma aromatica-DPPH[36]
Curcuma kwangsiensis-DPPH[37]
Dalbergia pinnata-DPPH, ABTS[39]
Eucalyptus camaldulensis-DPPH, ABTS, B16-F10[41]
Eugenia uniflora-DPPH, β-carotene/linoleic acid[43]
Glechoma hederacea-β-carotene/linoleic acid, B16[44]
Helichrysum microphyllum-DPPH, ABTS[45]
Hypericum hircinum-DPPH, ABTS[47]
Lavandula augustifolia-B16-F10[49]
Melaleuca quinquenervia1,8-Cineole, α-pipene, α-terpineolB16[53]
Ocimum basilicum-DPPH, hypoxanthine/xanthine oxidase[58]
Ocimum gratissimum-DPPH, hypoxanthine/xanthine oxidase[58,237]
Ocimum micranthum-DPPH, hypoxanthine/xanthine oxidase[58]
Ocimum tenuiflorum-DPPH, hypoxanthine/xanthine oxidase[58]
Piper aleyreanum-DPPH[61]
Psidium guineense-DPPH, ABTS[238]
Pomelo peel-DPPH, ABTS[68]
Satureja hortensis-DPPH[77]
Stachys cretica-DPPH[79]
Stachys hydrophila-DPPH[79]
Stachys palustri-DPPH[79]
Stachys parviflora-DPPH, β-carotene/linoleic acid[80]
Tanacetum macrophyllum-DPPH, ABTS, FRAP[83]
Thymus munbyanus-DPPH, ABTS, FRAP[85]
Thymus vulgaris-DPPH[125]
Vetiveria zizanioides-β-carotene/linoleic acid, B16[87]
Vitex Negundo-DPPH, ABTS, metal-ion chelation[88]
Wedelia chinensis-B16-F10 (C57BL/6)[90,239]
EugenolA2058,
SK-MEL 28
[134]
where not indicated, EO active components were not available from the cited articles. When referred to in vivo studies, the murine strain used is indicated in brackets. 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,2′-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid (ABTS), ferric reducing/antioxidant power (FRAP).

Share and Cite

MDPI and ACS Style

Di Martile, M.; Garzoli, S.; Ragno, R.; Del Bufalo, D. Essential Oils and Their Main Chemical Components: The Past 20 Years of Preclinical Studies in Melanoma. Cancers 2020, 12, 2650. https://doi.org/10.3390/cancers12092650

AMA Style

Di Martile M, Garzoli S, Ragno R, Del Bufalo D. Essential Oils and Their Main Chemical Components: The Past 20 Years of Preclinical Studies in Melanoma. Cancers. 2020; 12(9):2650. https://doi.org/10.3390/cancers12092650

Chicago/Turabian Style

Di Martile, Marta, Stefania Garzoli, Rino Ragno, and Donatella Del Bufalo. 2020. "Essential Oils and Their Main Chemical Components: The Past 20 Years of Preclinical Studies in Melanoma" Cancers 12, no. 9: 2650. https://doi.org/10.3390/cancers12092650

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