Next Article in Journal
Baseline MRI-Radiomics Can Predict Overall Survival in Non-Endemic EBV-Related Nasopharyngeal Carcinoma Patients
Next Article in Special Issue
Chemopreventive Potential of Caryophyllane Sesquiterpenes: An Overview of Preliminary Evidence
Previous Article in Journal
Quantification of Receptor Occupancy by Ligand—An Understudied Class of Potential Biomarkers
Previous Article in Special Issue
Carotenoids in Cancer Apoptosis—The Road from Bench to Bedside and Back
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An In Vitro Evaluation of the Molecular Mechanisms of Action of Medical Plants from the Lamiaceae Family as Effective Sources of Active Compounds against Human Cancer Cell Lines

by
Przemysław Sitarek
1,*,
Anna Merecz-Sadowska
2,
Tomasz Śliwiński
3,
Radosław Zajdel
2 and
Tomasz Kowalczyk
4
1
Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland
2
Department of Economic Informatics, University of Lodz, 90-214 Lodz, Poland
3
Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
4
Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
*
Author to whom correspondence should be addressed.
Submission received: 31 August 2020 / Revised: 5 October 2020 / Accepted: 9 October 2020 / Published: 13 October 2020
(This article belongs to the Special Issue Medicinal Plants and Their Active Ingredients in Cancer)

Abstract

:

Simple Summary

Plants have been used in folk medicine for thousands of years. The Lamiaceae family is one of the largest families of flowering plants and includes a wide variety of species with biological and medical uses. They are mainly herbs and shrubs with an aromatic scent and rich in valuable compounds of great value in medicine. The article focuses on the assessment of the anticancer properties of extracts, essential oils, and pure compounds derived from various species of the Lamiaceae family and their potential molecular mechanisms of action in in vitro studies against the four most common types of cancer in women and men: breast, lung, prostate, and colon.

Abstract

It is predicted that 1.8 million new cancer cases will be diagnosed worldwide in 2020; of these, the incidence of lung, colon, breast, and prostate cancers will be 22%, 9%, 7%, and 5%, respectively according to the National Cancer Institute. As the global medical cost of cancer in 2020 will exceed about $150 billion, new approaches and novel alternative chemoprevention molecules are needed. Research indicates that the plants of the Lamiaceae family may offer such potential. The present study reviews selected species from the Lamiaceae and their active compounds that may have the potential to inhibit the growth of lung, breast, prostate, and colon cancer cells; it examines the effects of whole extracts, individual compounds, and essential oils, and it discusses their underlying molecular mechanisms of action. The studied members of the Lamiaceae are sources of crucial phytochemicals that may be important modulators of cancer-related molecular targets and can be used as effective factors to support anti-tumor treatment.

Graphical Abstract

1. Introduction

Cancer is an important health problem and leading cause of death globally. The development of cancer is a multistage process that begins with genetic alteration and is followed by abnormal cell proliferation. Carcinogenesis is strictly related to the activation of oncogenes (induction of cell growth) and the inactivation of tumor suppressor genes (repression of cell growth), resulting in a loss of control of cell cycle progression. This initiation stage is followed by progression related to additive mutation within cells, some of which are implicated in even more rapid growth, and the suppression of cancer cell death. As a result of these changes, mature epithelial cancer cells may undergo epithelial–mesenchymal transition (EMT), which is characterized by the reduction of adhesion among cells and increased cell motility. Finally, tumor invasion and metastasis occur, and these are strictly related to angiogenesis, i.e., the process of new blood vessel formation. The progression of angiogenesis depends on the secretion of growth factors by cancer cells. The transformation of normal cells into malignant ones is a complex process regulated at every step by numerous factors, each of which may be a crucial target for anticancer agents [1,2].
The most prevalent form of cancer globally is lung cancer (11.6% of total cases), followed by breast cancer, among women (11.6%), prostate cancer, among men (7.1%), and colorectal cancer (6.1%) [3]. In 2018, more than 18 million cases of cancer were reported, and there were more than 9.6 million deaths globally [4]. In addition, the cost of medical care and economic value of nonmedical expense has been still rising [5].
Research into cancer therapy is currently directed toward the development of new therapeutic strategies and more effective chemotherapeutic factors [6]. An important source of novel effective agents with medicinal potential is the plant kingdom [7]. It is estimated that approximately 35 to 70,000 higher plant species produce secondary metabolites with diverse forms of bioactivity that are widely used in the regulation of signaling and metabolic pathways [8]. One family of particular interest is the Lamiaceae, which includes a number of herb and shrub genera with significant anticancer activities, such as Salvia sp., Thymus sp., Origanum sp., Melissa sp., Plectranthus sp., or Scutellaria sp.; all have been found to possess effective antiproliferative potential against lung, breast, prostate, and colon cancer cells in vitro. They commonly exert their cytotoxicity by promoting cancer cell death, especially via the apoptosis pathway, but they have also been found to influence angiogenesis [9]. Therefore, plant extracts, individual compounds, and essential oils from the Lamiaceae may support treatment as alternative or complementary cancer therapy.
The present paper focuses on the anticancer effects of plant extracts, purified single compounds, and essential oils from selected species of the Lamiaceae family. It discusses their in vitro cytotoxicity toward lung, colon, breast, and prostate cancer cell lines and the underlying mechanisms of action.

2. Criteria for Selection of Experimental Papers

This review was conducted to report work done previously to access the anticancer activity of plants from the Lamiaceae family published from 2015 to 2020. The studies were selected in the electronic databases PubMed/MEDLINE, Scopus, Web of Science, and Google Scholar. The search terms included Lamiaceae alone, and with the following: plant extract, derived compounds, essential oils, cancer, lung cancer cells, colon cancer cells, breast cancer cells, prostate cancer cells, mechanism of action. Published experimental studies reporting extracts, derived compounds, and essential oils from plants belonged to the Lamiaceae family with in vitro activity against lung, colon, breast, and prostate cancer cell lines were included. Research reporting review articles, published in languages other than English, abstract only or without full text access, lacking specific plant names with no reports of clear objective and methodologies, published more than five years ago, using plant species other than Lamiaceae, and cell lines other than lung, colon, breast, and prostate were excluded. The duplicates of articles obtained from the electronic databases were removed. After removal, inclusion/exclusion criteria were checked. Each selected document was examined and the following data were extracted and presented in the table: the scientific names of the species, parts of the plants used for extract preparation, types of extract, class of compounds, or compounds identified in extracts, cancer cell line, and reference. Articles with included mechanisms of action of interested plant extracts, single compounds, and essential oils were discussed in the main text.

3. Cancer

The term cancer is used to refer to a large group of diseases that can affect any part of the body. They are caused by uncontrolled cell proliferation that can take place in different tissues and spread into surrounding and distant organs [10,11]. Cancer occurs by a series of successive mutations in the relevant genes, leading to changes in cell function. Various physical and chemical factors play an obvious role in the formation of gene mutations and the appearance of cancer cells [12].
The first records of cancer date back to the ancient Egyptian and Greek civilizations, where the disease was treated mainly with radical surgery and often ineffective cauterization procedures, leading to the death of patients [13]. Currently, cancer is one of the most commonly occurring conditions and a major public health problem worldwide [14,15,16]. In 2018, cancer was responsible for approximately 9.6 million deaths [4]. Statistics show that high cancer morbidity and mortality are associated with an increasing incidence of risk factors such as overweight, alcohol abuse, smoking, unhealthy diets, urban air pollution, hepatitis and human papilloma virus, lack of physical activity, or sedentary lifestyle [17,18]. The World Health Organization (WHO) reports that in 2018, the highest percentages of cancer types in men occur in the lung, prostate, and colon, while the greatest prevalence in women is observed in the lung, breast, and colon. The most common types in both men and women were lung (2.09 million cases) and colon cancer (1.80 million cases). The second most common types of cancer were breast cancer in women (2.09 million cases) and prostate cancer in men (1.28 million cases) [14,19,20].
There are two main forms of lung cancer: non-small cell lung cancer (NSCLC; 85% of patients) and small cell lung cancer (SCLS, 15%). In 2018, this cancer caused 1.76 million deaths [14]. It is currently the most lethal malignant tumor in the world, often because it is not detected until the disease progresses significantly, leading to a significant reduction in the patient’s quality of life [21,22]. So far, genetic factors leading to increased susceptibility to lung cancer have been poorly studied. First-degree relatives of patients with lung cancer are at increased risk, even after adjusting for smoking habits. The most commonly accepted causes of lung cancer include active cigarette smoking, exposure to passive smoking, pipe and cigar smoking, indoor and outdoor air pollution, radiation, and occupational exposure to factors such as asbestos, nickel, or chromium [23,24,25]. Among others, important common driver gene mutations in patients with NSCLC are as follows: tumor suppressor genes TP53 and PTEN; EGFR, which encodes protein, is involved in cell growth and survival [26]. These genes constitute molecular targets for compounds from the Lamiaceae family.
Another common cancer among women, and one of the leading causes of death, is breast cancer [27]. In 2018, this cancer caused 627,000 deaths [14]. Breast cancer risk is related to many factors including a diet too high in calories leading to obesity, tobacco smoking, hormonal history, exposure to estrogen and progesterone, early first menstruation, and late menopause. All of these factors lead to shorter menstrual cycles and hormone exposure [28]. Genetic predisposition also plays an important role, as approximately 7% of breast cancer cases are hereditary [29]. In turn, mutations in the BRCA1 and BRCA2 genes cause as much as 80% of genetic breast cancer [30,31]. BRCAs proteins regulate genes linked to DNA repair, the cell cycle, and apoptosis [32]. Another crucial mutations is that in TP53, which is linked with histological grade in breast cancer patients [33].
Prostate cancer is the second most common malignant neoplasm after lung cancer in men around the world [34]. In 2018, this cancer caused 358,989 deaths, this being 3.8% of all deaths caused by cancer in men. About 60% of cases are diagnosed in men over 65. The mean age of diagnosis is 66 years. The disease rarely occurs in people under 40 years of age [35]. The main risk factors for the development of prostate cancer in men can be divided into non-modified and modified factors. Non-modified factors include age, family history, ethnicity, and genetic factors, while the modified ones are obesity, infectious diseases, smoking, hormones, or occupational and external exposure [36,37,38]. Inherited mutations in specific genes, such as BRCA1, BRCA2, and HOXB13, are the cause of some hereditary prostate cancer cases. HOXB13 protein regulates androgen receptor function [39]. Men with mutations in these genes are at high risk of developing prostate cancer and, in some cases, other cancers during their lifetime [40]. Additionally, other different genes have been implicated in prostate cancer by genetic alteration, depending on types (familial, sporadic, hereditary): TP53, PTEN, and protooncogenes MYC, which also constitute molecular targets for compounds from the Lamiaceae family [41].
Colorectal cancer is already the second most common cause of cancer death in the world (862,000 deaths) and 65% of all new incidences occur in high-income countries in Europe [14]. This incidence constitutes a 1.51% cumulative risk (assuming the absence of other competing causes of death) of colon cancer among men age 0–74 years, and a 1.12% risk among women. Their cumulative lifetime risks are 1.2% and 0.65%, respectively [3]. Colorectal cancer has been attributed to a number of mutations: mutational inactivation of tumor suppressor genes such as TP53, and activation of the oncogene pathway PI3K. These genes constitute molecular targets for compounds from the Lamiaceae family [42]. Genetically, colorectal cancer is divided into three categories: sporadic (about 60%) for patients with no family history, family (about 30%) for patients with at least one relative with colorectal cancer or adenoma, and hereditary forms (about 10%) due to the germline inheritance of mutations [43,44]. The development of colorectal cancer is believed to be supported by family history, lack of physical activity, old age, excessive alcohol consumption, high-fat diet, diabetes, and inflammatory bowel diseases, including ulcerative colitis and Crohn’s disease [45,46,47].

4. Cancer and Plants

Plants have been used in folk medicine for thousands of years [48]. The oldest written evidence of the use of medicinal plants in the preparation of medicines was found on a Sumerian Nagpur clay plate, approximately 5000 years old. It contained 12 prescriptions for the preparation of drugs, relating to over 250 different plants, including genera such as Papaver sp., Hyoscyamus sp., or Mandragora sp. containing alkaloids [49]. Medicinal plants are still used in developing countries as the source of healing due to their natural multidirectional action and low cost. The secondary metabolites contained in plants mainly do not play a direct role in their growth but serve as defence compounds against herbivores and microorganisms; therefore, they are required for survival in a specific environment. Most are polyphenols, alkaloids, terpenoids, or polyketides, among which there are a number of subclasses. Some have been found to demonstrate anticancer [50], anti-inflammatory [51], antioxidant [52], antimicrobial [53], antiviral [54] or anti-obesity effects [55]. In addition, while they can act as pure compounds, they also can show synergistic or antagonist effect with enhanced, or weaker, biological properties when administered as plant extracts [56,57,58].
According to the WHO, some nations still consider plant-based treatments to be the primary source of medicine and developing countries are exploiting the benefits of natural origin compounds for therapeutic purposes [59]. Numerous studies confirm the positive use of such natural compounds in the treatment of many diseases [60]. In turn, medicinal plants are also a valuable source of bioactive anticancer agents [61,62,63,64,65,66]. If plant-derived compounds can demonstrate selectivity in research, are non-toxic to normal cells, and show cytotoxicity in cancer cell lines, these compounds can be used in clinical trials for further therapeutic development. However, it is worth remembering that chemicals of plant origin may also exert toxic effects on both animals and human organisms in in vivo study [61,67].
The modern search for anticancer drugs from plants began in the 1950s, with the discovery of vinca alkaloids from the genus Vinca [68]. This highlights that medicinal plants remain a source of new drugs [69], e.g., vinblastine or vincristine obtained from the Vinca sp., paclitaxel from the Taxus sp., camptothecin from the Camptotheca sp., or podophyllotoxin from Podophyllum sp. [48,61,70,71]. These compounds often exert their anticancer activity by inhibiting the proliferation of cancer cells and inducing cell death [67]. Many in vivo and in vitro studies show the activation of apoptosis in various cancer cells, both under the influence of pure compounds and plant extracts [71]. Numerous plant species from different families (Lamiaceae, Fabaceae, Asteraceae, Papaveraceae, Apocynacea etc.) are traditionally used to treat or prevent the development of cancer.

5. The Lamiaceae Family Plants

The Lamiaceae family is one of the largest families of flowering plants and includes a wide variety of species with biological and medical uses. They are mainly herbs and shrubs with an aromatic scent and rich in valuable compounds of great value in natural medicine. Plants of this family are characterized by square stems and opposite leaves [72,73]. The most famous representatives are thyme, mint, oregano, basil, sage, savory, rosemary, hyssop, and lemon balm, which are used as aromatic spices, and some others with more limited uses [74]. Historically, species in the Lamiaceae family have enjoyed a long history of use in flavoring, preserving food, and for medicinal purposes. This family includes about 250 genera and about 7000 species, with the largest genera being Salvia, Scutellaria, Stachys, Plectranthus, Hyptis, Teucrium, Vitex, Thymus, or Nepeta (Figure 1). It is one of the most economically important families with great diversity and cosmopolitan distribution due to the aromatic properties of most of its members [75]. It is well known that each species produces a wide variety of secondary metabolites with strong antibacterial, antioxidant, anti-inflammatory, antiviral or anticancer properties; the oils comprise a complex mixture of bioactive compounds, in which each ingredient contributes to its overall bioactivity [56].

5.1. The Lamiaceae Family as a Source of Valuable Secondary Metabolites with Anti-Cancer Potential

The first and largest group of secondary metabolites occurring in the Lamiaceae is polyphenols, which are characterized by at least one aromatic ring having hydroxyl groups. Based on their numbers of phenolic groups and structural elements, polyphenols can be divided into phenolic acids, flavonoids, stilbenes, lignans, lignins, coumarins, anthraquinones, and xanthones [76,77,78]. In the human body, polyphenols exhibit antioxidant properties, which protect against chronic diseases caused by free radicals damaging of tissues and organs [72], as well as various anticancer [79] anti-diabetic [80], neuroprotective [81], anti-inflammatory [82], antiviral [83], antifungal [84] or antibacterial properties [85]. Polyphenols are believed to cause cancer cell death by apoptosis through several mechanisms, such as DNA fragmentation, alteration of the level of apoptotic proteins, and mitochondrial membrane potential and cell cycle arrest [86,87].
Another potent group of secondary compounds is the terpenes, in particular oxygenated terpenoids [88]. In turn, terpenoids are categorized into monoterpenes, sesquiterpenes, diterpenes, sesterpenes, and triterpenes depending on the number of their isoprene units. Diterpenes constitute a diverse class of plant metabolites with more than 10,000 different structures, isolated in the Lamiaceae present about 50 different skeletons [89,90]. Most diterpenes play a critical role in ecological interactions of plants and show interesting biological activities both in vitro and in vivo [91,92], such as antibacterial, antifungal, antiprotozoal, enzyme-inducing, anti-inflammatory, and the modulation of immune cell function and anticancer properties [92,93]. Studies have shown that some diterpenes have significant cytotoxic and cytostatic effects on various cell lines of human origin, interfere with the biochemical pathways of apoptosis and the cell cycle phase, and can influence the expression of proto-oncogenes such as avian myelocytomatosis viral oncogene homolog (c-Myc) and B-cell lymphoma 2 (Bcl-2) [93,94].
Many members of the Lamiaceae also produce alkaloids, which is an extremely diverse chemical group based on a ring structure including a nitrogen atom. Many alkaloids are toxic and are used by plants to protect themselves against aggression from other organisms [95,96]. Many such compounds have a strong cytotoxic effect and induce apoptosis through various pathways in different cell lines [97,98].
Essential oils (EO) are volatile and complex mixtures of diverse compounds, typically with a strong odor, synthesized as secondary metabolites by aromatic plants, mainly from the Lamiaceae family (132). Although EO are mainly contained in the leaves, they can be found in all the above-ground parts of plants. They are widely used in cosmetics, flavors, fragrances, perfumes, pesticides, and the pharmaceutical industry [99]. These phytocomplexes can be obtained by hydro or dry distillation. Their ingredients include sesquiterpenes, oxygenated sesquiterpenes, monoterpenes, oxygenated monoterpenes, and phenols, among others [100,101]. Many preclinical studies have found some anticancer, antibacterial, antioxidant, or anti-inflammatory effects in a range of cellular and animal models [102], and they have examined their mechanism of action and pharmacological targets [101].
The particular signaling pathways and related factors that constitute a target for natural anticancer modulators from Lamiaceae discussed in this paper are given in Figure 2.

5.2. The Anti-Cancer Activity of Plant Extracts from the Lamiaceae Family

Many members of the Lamiaceae have demonstrated considerable efficacy in inhibiting cancer cell growth through synergistic effects. Extracts from many species demonstrate cytotoxic properties against lung, colon, breast, and prostate cancer cells (Table 1). This section discusses their mechanisms of action, which are mainly based on inducing the apoptosis in cancer cells including lung, breast, prostate, and colon cancer.

5.2.1. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of Cell Cycle

The cell cycle events controlling cell duplication and arrest are highly dysregulated in cancer cells. Cell cycle arrest is closely related to the G1/S, G2/M and M phases’ checkpoints perturbations. Progression is mediated by the activation and deactivation of cyclin-dependent kinases (CDKs), while activation depends on the presence of activated subunits named cyclins. The levels of cyclins and CDKs are changed in human cancers; for example, the levels of CDK1 and cyclin B1 are reduced in human breast adenocarcinoma (MCF-7) and human colorectal cancer (HCT-116) cell lines following treatment with Micromeria fruticosa aerial part extract, resulting in G2/M arrest [143,144]. In addition, treatment with Melissa officinalis leaf extract blocked the expression of CDKs 2, 4, and 6 and cyclin D3 in the human colon carcinoma (HT-29 and T84) cell lines, and specifically activated an important CDK inhibitor named p18 [145]; in addition, treatment with Vitex rotundifolia fruit extract downregulated cyclin D1 and CDK 4 levels in HCT-116 and human colon adenocarcinoma (SW480) [146]. Manipulation of the cell cycle may induce an apoptotic response [147]. Ocimum basilicum leaf extract induced cell cycle arrest in MCF-7 cells [148], and Salvia miltiorrhiza root extract induced a G2/M phase arrest in human lung adenocarcinoma (Glc-82) cells [149], as did Melissa officinalis leaf extract in HT-29 and T84 cells and Prunella vulgaris root extract in human breast carcinoma MCF-5 cells [150]. Satureja khuzistanica extract enriched with rosmarinic acid [151] was found to increase the size of the apoptotic sub-G0/G1 population in MCF-7 cells.

5.2.2. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of Apoptosis Signaling

Apoptosis or programmed cell death is a crucial mechanism for maintaining cell homeostasis. The process can be triggered by various conditions, including immune reaction or reactive oxygen species (ROS). The study of Ocimum sanctum roots extract suggested that it may increase ROS production in the HCI-H460 lung carcinoma cell line and may decrease viability via apoptosis. The same phenomenon is observed for Melissa officinalis leaf extracts against HT-29 and T84 cell lines [145]. That strategy is confirmed by the occurrence of excessive numbers of apoptotic cells [152]. It was concluded that also stress originating from the endoplasmic reticulum may play an important role in triggering apoptosis; for example, Scutellaria barbata extract induced human lung cancer (CL1-5) cell death [153].
The signaling mechanisms of apoptotic cell death are divided into two pathways: an intrinsic pathway mediated by mitochondria and an extrinsic one mediated by death receptors. Cell size reduction, membrane blebbing, and apoptotic bodies were observed in HT-29 cells after Stachys pilifera leaf extract treatment; these were all signs of apoptosis [154]. MCF-7 cells showed a modified nucleus following incubation with Caryopteris x clandonensis stem extract [155], and changes in cell rounding, shrinkage, and detachment from other cells after treatment with Teucrium mascatense whole plant extract [156]. Another important future of apoptosis is the translocation of phosphatidylserine phospholipid from the inner to the outer plasma membrane, resulting inter alia in recognition by phagocytes [157]; this was observed against HT-29 and T84 cell lines after the administration of Teucrium mascatense extract [156] and Melissa officinalis leaf extract [145]. Teucrium flavum whole plant extracts were found to induce DNA fragmentation in breast carcinoma cells (MDA-MB-231) [158], as well as Vitex rotundifolia leaf extract in human breast cancer T-47D cells [159] and Ocimum sanctum leaf extract in human prostate adenocarcinoma LNCaP cells [160]; such DNA cleavage is a hallmark of apoptosis. Perovskia abrotanoides flower extract has also demonstrated proapoptotic effects against MCF-7 cells [161] and Salvia chorassanica root extract against MCF-7 and prostate cancer cells (DU-145) [162].
The treatment of human non-small cell lung carcinoma cells (NCI-H460) with Ocimum sanctum root extract [152] and MCF-7 cells with Teucrium sandrasicum leaf extract [163] resulted in increased mitochondrial membrane permeability, which is characteristic of the intrinsic pathway. The Bcl-2 protein family also induces a loss of mitochondrial membrane potential. This family is separated into two groups: one, including Bak and Bax, which possesses proapoptotic potential, and another, including Bcl-2 and Bcl-xL, which has antiapoptotic activities. The balance between the two groups influences the progression of apoptosis. The Bcl-2 protein family was found to be modulated by the Origanum compactum aerial part extract in A549 cells [164]. Changes in the level of Bcl-2 family members may result in the release of numerous pro-apoptotic molecules. The expression of Bcl-2 protein family members in different lines of human lung cancer cells became unregulated following the administration of extracts from Salvia milithoriza roots (Glc-82 cells) [149], Scutellaria baicalensis root (H358 and H2087 cells) [165], and Melissa officinalis leaves (A549) [166]. A similar result was observed for several human colon cancer cell lines treated with Coleus amboinicus leaves (WiDr cells) [167] and Vitex rotundifolia fruit (HCT-116, SW480, LoVo and HT-29 cells) extracts [146]. In addition, apoptotic signals have been triggered in breast cancer cell lines after incubation with extracts of Plectranthus amboinicus leaves (MCF-7 cells) [120], Orthosiphon stamineus leaves (MCF-7 cells) [168], Melissa officinalis leaves (MCF-7 cells) [166], Vitex rotundifolia leaves (T-47D cells) [159], and Prunella vulgaris root (MCF-5 cells) [150]. Changes in the expression level of Bcl-2 family protein are also induced in prostate cancer cell lines by extracts of Ocimum sanctum leaves (LNCaP cells) [160], Dracocephalum palmatum (PC-3 cells) [169], and Melissa officinalis leaves (PC-3) [166].
The insertion of Bax/Bak into the mitochondrial membrane results in the formation of a pore complex and release of cytochrome c into the cytosol from the intermembrane space. In contrast, Bcl-2 and Bcl-xl prevent the release of cytochrome c. Excessive levels of cytochrome c were observed in MCF-7 cells after Orthosiphon stamineus leaf extract treatment [168]. Cytochrome c binds to apoptotic protease-activating factor 1 (Apaf-1) to create an apoptosome that activates caspase-9. Caspase-9 was found to be upregulated following treatment with Coleus amboinicus leaf extract in WiDr cells [167], Teucrium sandrasicum leaf extract in MCF-7 cells [163], Vitex rotundifolia leaf extract in T-47D cells [159], Micromeria fruticose aerial part extract in MCF-7 cells [144], Stachys pilifera leaf extract in HT-29 cell line [154], and Teucrium chamaedrys aerial and flowering parts in SW480 cells [170].
Caspase-9 activity results in the activation of the effectors caspase-3 and caspase-7. Caspase-3 and/or caspase-7 activity were observed in the Glc-82, LNCaP, MCF-7, T-47D, MCF-7, and WiDr cell lines following treatment with, respectively, extracts of Salvia miltiorrhiza roots [149], Ocimum sanctum leaves [160], Plectranthus amboinicus leaves [120], Vitex rotundifolia leaves [159], Teucrium mascatense whole plant [156], and Coleus amboinicus leaves [167]. The induction of caspase-3 and/or caspase-7 cleavage was also observed for Melissa officinalis leaf extracts against HT-29 and T84 cells [145] and for Origanum majorana leaf extract against HT-29 cells [171]. Dracocephalum palmatum leaf extract also induced apoptosis according to the intrinsic pathway via the upregulation of activated caspase-3 in PC-3 cells [169]; the same was observed for Nepeta cataria aerial parts on PC-3 cells [172] and Scutellaria baicalensis whole body extract on H358 and H2087 cell lines [165].
In apoptosis, the detection of poly-ADP-ribose-polymerase (PARP) is an important diagnostic method because it produces specific patterns of proteolytic cleavage fragments [173]. Increased expression of PARP1, a hallmark of apoptotic death, was observed in Glc-82 cells treated with Salvia miltiorrhiza roots extract [149], LNCaP cells incubated with Ocimum sanctum leaf extract [160], and PC-3 cells interacted with Nepeta cataria aerial part extract [172]. PARP activation was also demonstrated after the treatment of Teucrium mascatense whole plant extract in MCF-7 cells [156], Rosmarinus officinalis leaf extract in A549 cells [174], and Scutellaria baicalensis root extract in H358 and H2087 cells [165], suggesting the involvement of mitochondria in the apoptotic signals.
The extrinsic pathway is initiated by the TNF (tumor necrosis factor), TRAIL (TNF-related apoptosis-inducing ligand), and Fas ligands binding to the extracellular domain of death receptors, such as type 1 TNF receptor (TNFR1), TRAIL, and Fas receptors. The Teucrium chamaedrys aerial flowering part extract initiated an excessive expression of Fas in SW480 cells [170]. The Fas ligand–receptor junction is inhibited by nucleolin, which is a protein that protects against apoptotic death [175]. Orthosiphon stamineus leaf extract treatment significantly decreased the nucleolin level in MCF-7 cells [168]. The attachment of a Fas to a specific receptor triggers the formation of specific death-inducing signaling complex (DISC) possessing a Fas-associated death domain (FADD); this complex can recruit an adaptor protein and then activate initiator caspase-8 and caspase-10. Caspase-8 induction was found to induce apoptosis in MCF-7 and HCT-116 cells following Micromeria fruticose aerial part extract administration [144], as well as treatment with Vitex rotundifolia leaf extract in T-47D cells [159], Stachys pilifera leaf extract in the HT-29 cell line [154], and for Teucrium chamaedrys aerial flowering part extract in SW480 cells [170]. In addition, it was speculated that Satureja khuzistanica extract enriched in rosmarinic acid can induce apoptosis through activation of the extrinsic pathway in MCF-7 cells [151]. Then, the activation of caspase-8 and caspase-10 is followed by the activation effector caspases 3/7, enzymatic cleavage of numerous downstream targets, and cell death [176,177]. The Fas-mediated apoptosis pathway is believed to play a role in Scutellaria barbata extract-induced CL1-5 cell cytotoxicity [153].
An important inhibitor of apoptosis is called survivin. This protein is under-expressed in cancer cells and is related to poor clinical outcome through the blockage of apoptosis by caspase inhibition [178]. Significant reductions of survivin levels were observed for Micromeria fruticosa aerial part extract in MCF-7 and HCT-116 cell lines [143,144], Origanum majorana leaf extract in HT-29 cells [171], and for Scutellaria barbata whole plant extract in HT-29 cells [179]. Another key molecule that plays a crucial role in the negative regulation of apoptosis is Her2, belonging to the human epidermal growth factor receptor family. Excessive Her2 expression is related to antiapoptotic signals via the activation of survivin and Bcl2 protein. Melissa officinalis extract shows significant potential to reduce Her2 levels in PC-3 cells [166].
Another regulator of apoptosis and autophagy is mammalian target of rapamycin kinase (mTOR), which also demonstrates pleiotropic features [180]. The activation of the mTOR pathway was promoted by Ocimum basilicum leaf extract in MCF-7 cells with p70S6K, which is a downstream target kinase [181]. Similar features were shown for Scutellaria baicalensis root extract in H358 and H2087 cell lines [165].

5.2.3. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of p53 Signaling

DNA damage occurs due to metabolic processes and environmental factors including ROS and results in an increase in the levels of tumor suppressor protein p53. The protein serves as a key regulator of the cellular response and triggers target genes including CDK and inhibitor p21, taking part in cell cycle arrest, and the proapoptotic protein Bax [182]. p53 expression was dysregulated in WiDr cells by Coleus amboinicus leaf extract [167], and the levels of both p53 and p21 increase in Glc-82 after treatment with Salvia miltiorrhiza root extract [149] and in MCF-7 cells after treatment with Plectranthus amboinicus leaf extract [120]. The level of p21 was induced in cell lines HT-29 and T84 after incubation with Melissa officinalis leaf extract [145]. p53 may be regulated by Sirtuin 1 (SIRT1), which is a member of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases. The inhibition of SIRT1 was found to enhance apoptosis in CL1-5 cells treated with Scutellaria barbata extract [153]. One of the outcomes of p53 activation is cell cycle arrest, and these mechanisms have potent antitumor effects.

5.2.4. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of PI3K/AKT Signaling

Apoptosis and cell cycle progression are mediated by phosphatidylinositol 3-kinase (PI3K) signal transduction. Activated PI3K is responsible for the conversion of phosphatidylinositol (4,5)-phosphate into phosphatidylinositol (3,4,5)-phosphate [183]. PI3K levels were significantly reduced in A549 cells after the administration of Nepeta cataria whole plant extract [184]. That PI3K signaling cascade induces protein kinase B, which is also known as PKB or Akt. Akt is a pro-oncogene and enables tumor proliferation. Akt has numerous downstream effects and controls several biological responses, including the phosphorylation of apoptosis signal proteins Bcl-2 and Bcl-xL followed by the suppression of apoptosis and also caspase-9 and p53. Prunella vulgaris root extract was found to modulate the PI3K/Akt signaling pathway in MCF-5 cells [150]. Akt may inactivate mammalian target of rapamycin complex 1 (mTORC1), p70S6 kinase, and p21, which are known to stimulate cell growth and proliferation. The levels of Akt, mTOR, and p70S6K were reduced in A549 cells after treatment with Rosmarinus officinalis leaf extract [174].
An important suppressor of PI3K is called PTEN: phosphatase and tensin homolog deleted on chromosome 10 [183]. PTEN expression was detected in A549 cells after Nepeta cataria whole plant extract [184]. An increased expression of PTEN was also observed in Glc-82 cells after Salvia miltiorrhiza root extract treatment, which then resulted in a reduced level of Akt [149]. The levels of phosphorylated Akt were reduced in human prostate cancer PC-3 cells following treatment with Dracocephalum palmatum leaf extract [169].

5.2.5. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of NF-κB Signaling

In addition, the stimulation of nuclear factor kappa B (NF-κB) signaling cascade is mainly related to antiapoptotic properties. The NF-κB protein family has five members: p50, p52, p65, RelB, and c-Rel. NF-KB pathways are divided into the canonical and non-canonical. The canonical one is activated by a range of cell stressor molecules such as TNF-α that interact with tumor necrosis factor (TNF) receptors. A first step is the induction of transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1), which promote stimulation of the IkB kinase (IKK) complex composed of IKKγ, IKKα, and IKKβ that phosphorylates IκB, which results in degradation. The remaining NF-κB (p50 and p65) migrate to the nucleus, bind to the DNA, and activate the transcription of numerous genes such as Bcl-2 and Bcl-xL. [185]. Hence, the suppression of NF-κB by Stachys pilifera leaf extract is believed to be responsible for the induction of apoptosis in HT-29 cells [154]. The NF-κB signaling cascade also plays a pivotal role in the inflammatory response by activating the transcription of several pro-inflammatory genes such as interleukin (IL) 1β, IL-8, and cyclooxygenase-2 (COX-2) [186]. Commercial standardized Thymus vulgaris extract (thymol 0.3% w/w) was found to downregulate the activity of p65 and modulate the release of IL-1β and IL-8, which play roles part in the inflammatory mechanisms in the H460 human lung cancer cell line [187]. COX-2 plays a part in numerous malignancies, including several human cancers. COX-2 is associated with apoptosis suppression followed by uncontrolled proliferation, metastasis, and angiogenesis as a consequence of tumor growth [188]. COX-2 expression was dysregulated by high concentrations of Origanum majorana extract in A549 cells [189].

5.2.6. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of Wnt/β-catenin Signaling

CDK/cyclin complexes control Wnt/β-catenin signaling [190]. Scutellaria barbata extract is related to decreased the expression of β-catenin in HT-29 cells [179], which is a component of the Wnt/β-catenin signaling pathway responsible for regulating cell growth. The signaling cascade is triggered by binding Wnt ligands to their receptors called Frizzled (Fz) and LDL receptor-related proteins 5 and 6 (LRP5 and LRP6). In the inactive state, a scaffolding protein Axin ensures β-catenin phosphorylation and promotes its degradation, but during the activation of the pathway, the Wnt ligand binds to Fz receptor, and it also allows β-catenin dephosphorylation and accumulation in the nucleus by Axin inhibition and the transcription of Wnt targeted genes such as c-Myc oncogene [191]. The suppression of Wnt signaling cascades induces apoptosis in SW480 and HCT-116 cells [192]. Additionally, Scutellaria barbata extract showed a potency to decrease the expression of the c-Myc in human colon adenocarcinoma HT-29 cells; excessive expression results in rapid cellular growth [193].

5.2.7. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of Autophagy Signaling

Another type of cell death connected with self-degradation is autophagy. That process may be caused by stress signals that originate from extracellular, intracellular, and endoplasmic reticulum such as growth factor deprivation, nutrient starvation, oxidative stress, protein aggregation, and pathogen infection. Upon autophagy, the organelles named autophagosomes capsules other organelles or a portion of cytosol and then fused them into lysosome and breakdown by lysosomal hydrolases. Nucleation of the phagophore is initiated by the activation of the Unc-51-like kinase 1 (ULK1) complex that triggered the phosphorylation the class IIIPI3K (PI3KC3) complex 1. This step is blocked by Bcl-2 proteins by direct association with becyclin-1, which is a component of that complex. The activated PI3KC complex mediates the production of phospatydyloinositol-3-posphate (PI3P). The first elongation step involves the enrollment of numerous autophagy-related (Atg) proteins by PI3P. The second phase of the elongation step is the formation of autophagosomal membrane-associated protein light chain 3 (LC3)-II from LC3-I via conjugation with phosphatidyl ethanolamine, which revealed the overexpression ratio of LC3-II/LC3-I in H358 and H2087 cells treated with Scutellaria baicalensis root extract [165]. Finally, autophagosomes fuse with lysosome and lysosomal enzymes with proteolytic activity degrade its cargo [194,195]. The autophagy is triggered in HT-29 cells by Origanum majorana leaf extract [171] and in CL1-5 cells by Scutellaria barbata extract [153].
The negative regulator of autophagy is mTOR. mTOR nucleates two distinct multi-protein complexes, mTORC1 and mTORC2. mTORC1 is composed of mTOR, regulatory-associated protein of mTOR (Raptor), mammalian lethal with Sec13 protein 8 (mLST8), proline-rich AKT substrate 40 kDa (PRAS40), and DEP-domain-containing mTOR-interacting protein (Deptor). mTORC1 positively regulates proliferation and cell growth. One of the most important factors involved in the regulation of mTORC1 activity is the tuberous sclerosis complex (TSC1/2). TSC1/2 functions as an activating protein for Rheb (Ras homolog enriched in brain). In turn, Rheb is able to activate mTORC1 [196]. One of the components of the mTOR pathway, Ras small GTPases—Rags, interact with mTORC1 and promote their translocation to a lysosomal membrane where Rheb resides [197].

5.2.8. The Activity of Plant Extracts from the Lamiaceae Family as Modulators of Necrosis Signaling

Necrosis is referred to as accidental cell death. It is frequently detected following harmful physical and chemical conditions, adverse stimuli, or deleterious mutations. The process is mediated by an imbalance of calcium flux, oxidative stress, and PARP. The physiological role of PARP is participation in DNA repair signaling in response to cell injury [198]. Excessive ROS levels, increased intracellular calcium, and the inhibition of PARP result in damage to cell components, degradation of proteins, and DNA damage. Necrosis is characterized by damage of the cell membrane and the release of its components into the extracellular space, resulting in inflammation and damage to the surrounding tissues [199,200]. This pathway is observed to take place in colon adenocarcinoma HT-29 and SW480 cell lines after Rosmarinus officinalis extract treatment. Rosemary extract possesses a strong antiproliferative activity, and its mechanism of action suggests the role of excessive intracellular ROS formation [201].

5.2.9. Plant Extracts from the Lamiaceae family and their Impact on Angiogenesis

The critical moment is tumor-induced angiogenesis. Melissa officinalis leaf extract halted neovascularization in breast adenocarcinoma MDA-MB-231 cells [202]. Antiangiogenic effects were also exerted by Prunella vulgaris root extract in MCF-5 cells [150]. The formation of new blood vessels from pre-existing ones may be regulated by numerous factors. One of them is epidermal growth factor (EGF) and its receptors (EGFR). It was observed that tumor cells demonstrate abnormally high EGFR activity and enhanced sensitivity to their ligands and the progression of tumor [203]. In turn, EGF was found to target vascular endothelial growth factor (VEGF) and induce their activity, whereas VEGF may modulate the EGFR signaling pathway [204]. VEGF demonstrated an ability to increase the permeability of existing blood vessels. The levels of both EGF and VEGF factors were found to be lowered in PC-3 cells and VEGF in DU-145, after Salvia triloba extract incubation [205]. Nepeta cataria whole plant extract exhibits a preventive effect against A549 cell invasiveness by reducing the level of VEGF [184], whereas Melissa officinalis leaf extract acts against PC-3, MCF-7, and A549 cells spread [166]. VEGF may be activated by human telomerase reverse transcriptase (hTERT), specific oncogene, catalytic subunit of the enzyme telomerase essential for chromosome termini replication. hTERT downregulation was observed in PC-3, MCF-7, and A549 cancer cell lines after Melissa officinalis leaf extract usage [166].
Other proangiogenic factors are angiogenin (ANG), IL-8 [206], leptin [207], and RANTES (regulated upon activation, normal T-cell expressed and secreted) [208]. ANG is a protein belonging to the RNase A superfamily, whose activity is related to the formation of blood vessels and tumor growth [209], ENA-78 is a chemokine associated with the activation of granulocytic immune cells and vascularity of the tumors, whereas leptin is an endocrine hormone produced by adipocytes. ENA-78 is strongly related to prostate cancer progression [210]. RANTES is an anti-inflammatory chemokine that recruits inflammatory cells and controls the secretion of growth factors included in the angiogenic process [208]. Salvia triloba extract was found to have the opposite effect on angiogenesis in PC-3 and DU-145 cells by reducing the levels of VEGF, ANG, ENA-78, IL-8, leptin, and RANTES [205].

5.3. The Anti-Cancer Activity of Plant-Derived Compounds from the Lamiaceae Family

Some of the compounds isolated from selected members of the Lamiaceae family and their cytotoxic effect against cancer cell lines are presented in Table 2. This section discusses the molecular mechanisms of action of these phytochemicals against lung, colon, breast, and prostate cancer cell lines.

5.3.1. The Anticancer Activity of Phenolics Compounds from the Lamiaceae Family

Rosmarinic acid (α-o-caffeoyl-3,4-dihydroxyphenyllactic acid; CAS 20283-92-5), an ester of caffeic acid, and 3,4-dihydroxyphenyllactic acid is commonly found in species of the Lamiaceae family. Rosmarinic acids and their derivatives possess a range of antioxidant, anti-inflammatory, antitumor, anti-angiogenic, and antimicrobial activities, among others [254]. Rosmarinic acid extracted from Salvia glabra demonstrates antitumor activity against breast cancer stem-like cells (BCSCs); this has been attributed to apoptosis induction by suppressing the expression of Bcl-2 and increasing that of Bax [255].
Wogonin (5,7-dihydroxy-8-methoxyflavone; CAS 632-85-9) is a flavone with numerous antitumor, anti-inflammatory, antiviral, and neuroprotective properties [256]. A natural source of wogonin is Scutellariae radix, which is the dried root of Scutellaria baicalensis. It has been shown to promote both autophagy and apoptosis processes in SW48 cells via the upregulation of autophagic factors including LC3II and Beclin 1 proteins and apoptotic factors such as caspase-3, caspase-8, caspase-9, and Bax proteins. The exposure to wogonin results in G2/M cell cycle arrest and the inhibition of PI3K/Akt signaling through attenuating PI3K protein expression [257]. Wogonin has been found to inhibit the invasiveness of MDA-MB-231 cells via suppressing the synthesis of two key factors related to new blood vessel formation: IL-8 and matrix metallopeptidase-9 (MMP-9) [258]. MMP-9 is critical for the progression of a pro-angiogenic outcome and the release of VEGF during carcinogenesis [259]. Scutellarein (5,6,7,4’-tetrahydroxyflavone), a flavone that is particularly present in the genus Scutellaria, has been found to be effective for the prevention and treatment of Helicobacter pylori infection, Alzheimer’s disease, and vascular complications of diabetes; it has also been found to inhibit certain carcinomas [260]. It was found that the exposure of the HCT116 cells to the scutellarein extracted from Scutellaria barbata induces apoptosis via ROS-mediated mitochondrial membrane permeability and cytochrome c release, and by downregulating the expression of Bcl-2, increasing Bax and cleaved-caspase-3 activity [261].
Another common flavanone is naringenin (4’,5,7-trihydroxyflavanone; CAS 480-41-1). Naringenin has shown antitumor, antioxidant, anti-inflammatory, antiviral, antibacterial, antiadipogenic, and cardioprotective potential [262]. Abaza et al. studied the effect of naringenin isolated from Thymus vulgaris whole plant on HTB26, HTB132, SW1116, and SW837 cells. Its administration was found to increase G1/S and G2/M phases cell cycle arrest and apoptotic cell death through the upregulation of p18, p21, Bak, Bax, activation of caspases 3, 7, 8, and 9, and the downregulation of CDK 4, 6, and Bcl2 in all cancer cell lines. It was also found to decrease cell survival factors including PI3K, Akt, and NFκBp65.

5.3.2. The Anticancer Activity of Terpenoids Compounds from the Lamiaceae Family

Arguably, the most important class of chemicals produced by the Lamiaceae family is the terpenoids; of these, the most numerous subclass of compounds with confirmed anticancer properties is that of the diterpenoids. Many possess pro-apoptotic properties, including 3-acetoxylteuvincenone from Ajuga ovalifolia whole plants [263], sahandone and sahandol II from Salvia chloroleuca roots [264], acetyl-macrocalin B from Isodon sylvatica [265], parvifloron D from Plectranthu ecklonii [227], and 7β-acetoxy-20-hydroxy-19,20-epoxyroyleanone from Salvia corrugate shoots [266]. 3-acetoxylteuvincenone activates Src homology phosphotyrosine phosphatase 2 (SHP2) and induces extracellular signal-regulated kinase (ERK) 1/2 and Akt pathways in A549 cells [263]. SHP2 controls the activation of Akt by insulin-like growth factor-1 (IGF-1), thus regulating the PI3K/Akt pathway related to the blockage of caspase 3-mediated apoptosis [267]. Moreover, SNP2 has been implicated in the activation of ERK 1/2, a member of mitogen-activated protein kinases (MAPKs) that restored the levels of c-Myc [268]. Sahandone and sahandol II change the ratio of Bax/Bcl-2 apoptotic proteins and activate PARP in the MCF-7, LNCaP, and PC-3 cell lines [264]. Acetyl-macrocalin B induces apoptosis in an ROS-dependent manner in A549 cells and then upregulates the p38 MAPK signaling pathway that mediates caspase-9 release. Additionally, the initiation of G2/M cell cycle delay throughout checkpoint kinase (Chks) Chk 1 and Chk 2 activation and cell division cycle 25C (Cdc25C) phosphatase degradation. Chk 1/2 are expressed during the cell cycle, where they regulate the spread of checkpoint signals and promote G2/M arrest by degrading Cdc25C phosphatase [269]. Cdc25C phosphatase dephosphorylates the cyclin B-CDK1 complex and enables cell entry into mitosis but inactivates it, preventing cell cycle progression [270]. Another diterpenoid, parvifloron D, induces apoptotic morphological changes in MDA-MB-231 cells [227]. 7β-acetoxy-20-hydroxy-19,20-epoxyroyleanone promotes G0/G1 cell cycle delay and ROS activation of ERK1/2 kinase in human breast cancer SKBR-3 and human breast carcinoma BT474 cell lines [266].
An important subgroup of terpenoid compounds is tanshinone diterpenoids, which are produced by Salvia miltiorrhiza roots; these are also connected with the induction of apoptosis in cancer cells. Increases in apoptosis were observed in colon cancer cells (human colorectal cancer DLD-1, human colon carcinoma COLO 205, and human colorectal adenocarcinoma Caco-2) exposed to trijuganone C [271], prostate cancer cells (PC3 and LNCaP) treated with tanshinone analog 2-(Glycine [methyl ester]methyl)-naphtho [272], and lung (human non-small cell lung cancer PC9 and A549) and breast cancer (MCF-7) cells administered by tanshinone [273]; these changes were attributed to the upregulation of numerous apoptotic factors including cytochrome c, pro- and antiapoptotic proteins ratio, caspases, PARP, p53, and p38. In addition, MCF-7 cells treated with three tanshinones isolated from the roots of Perovskia abrotanoides (cryptotanshinone, tanshinone 2A, and hydroxycryptotanshinone) exhibit high amounts of PARP protein cleavage, which is a hallmark of apoptosis [274].
Another diterpenoid, 11α, 12α-epoxyleukamenin E isolated from whole plants of Salvia cavaleriei show anticancer potential against HCT116 and SW480 cell lines [275]; this activity was attributed to suppression of the crucial Wnt signaling pathway, which regulates cell fate and the activation of targeted genes, including c-Myc and survivin. The diterpene clerodermic acid found in aerial parts of four Salvia species including S. spinosa, S. santolinifolia, S. syriaca, and S. nemorosa was also shown to be effective in suppressing Hypoxia-Inducible Factor (HIF) 1 alpha in A549 cells [276], which correlates with tumor metastasis and angiogenesis. Hypoxia is a common microenvironment in many types of solid tumors, and the HIF-1α pathway is crucial for their survival; therefore, the reduction of its expression may serve as a potential cancer therapy target [277].
It has been reported that phlomisoside F, a diterpene glycoside isolated from Phlomis younghusbandii root, promotes G0/G1 cell cycle delay and apoptosis induction, as confirmed by the overexpression of caspase-3, caspase-9, and Bax, and the underexpression of Bcl-2 and COX-2 in A549 cells [278]. 2α, 3α, 23-trihydroxy-13α, 27-cyclours-11-en-28-oic acid, a triterpene acid isolated from the aerial parts of Glechoma longituba, is responsible for NCI-H460 cell cycle arrest, the induction of ROS-mediated apoptosis, and the nullification of NF-κB activity [279]. The exposure of human lung cancer cell line SK-LU-1 cells to pogostemin A, a meroterpenoid isolated from the aerial parts of Pogostemon auricularius, triggered apoptosis and caspase-3 activation [280].

5.3.3. The Anticancer Activity of Polysaccharides Compounds from the Lamiaceae Family

Biologically active polysaccharides from natural plant sources also act as potent antitumor agents. They are also believed to be nontoxic and more preferred for living organisms [281]. SPS2p, a polysaccharide composed of carbohydrates, uronic acid, and proteins from Scutellaria barbata demonstrated pro-apoptotic activity against HT29 cells, probably by suppressing the PI3K/Akt pathway [282]. Whereas SBPW3, a polysaccharide containing rhamnose, arabinose, xylose, mannose, glucose, and galactose isolated from the same species also demonstrated anti-metastatic activity via the suppression of EMT in the same cell line [283].

5.4. The Anticancer Activity of Essential Oils from the Lamiaceae Family

The members of the Lamiaceae are sources of essential oils. Their cytotoxic potency against cancer cell lines are presented in Table 3. This section discusses their mechanisms of action against lung, colon, breast, and prostate cancer cell lines.
Nepeta rtanjensis essential oils rich in trans,cis-nepetalactone induced programmed cell death against A549 and MDA-MB-231 cells [319], as did Origanum onites essential oil, which is rich in carvacrol, against Ht-29 cells [320]. Ocimum viride essential oils, which have a high content of γ-terpinene, induced apoptosis as a consequence of DNA damage in HT-29 cells [321], while Thymus revolutus essential oil with a high content of γ-terpinene and p-cymene induced apoptosis in A549 cells [322]. Salvia aurea, S. judaica, and S. viscosa essential oils containing caryophyllene oxide as a main constituent induced apoptosis triggered by excessive ROS formation in DU-145 cells [323], as did Zataria multiflora essential oils in HCT-116 and SW48 cell lines [324].
G2/M cell cycle delay and apoptosis were observed in PC-3 cells after exposure to Lavandula angustifolia essential oil, with linalool and linalyl acetate as major components [325]. Plectranthus amboinicus essential oil, containing high amounts of carvacrol, dysregulated levels of pro- and antiapoptotic factors and induced caspase-9 and caspase-3 in A549 cells [326].
Patchouli alcohol, isolated from the essential oil of Pogostemon cablin, induced mitochondria-mediated apoptosis and caspase-9 and caspase-3 production in A549 cells [327]. The oil may also demonstrate its anticancer activity by dysregulating the MAPK pathway. Stachys viticina essential oils, with their main components being endo-borneol, eucalyptol, and epizonarene, inhibit mediators of apoptosis suppression COX-2 in COLO 205 cells [328]. The caspase-8 and caspase-9-dependent apoptosis pathways are triggered in HT-29 cells by treatment with Origanum majorana essential oil containing inter alia terpinen-4-ol, alpha-terpinol, alpha-pinene, camphene, p-cymol, B-caryophyllene, bicyclogermacrene, and neophytadiene [329]. Origanum majorana essential oil induces non-apoptotic cell death including autophagy and necrosis in HT-29 cells [329]; the same is observed in human lung cancer cell line Calu-3 treatment with Lavandula dentata essential oils rich in 1,8-cineole [330].

6. Conclusions

Cancer causes the greatest economic burden of any of the top 15 causes of death worldwide, both for the patient and society in general. Therefore, there is a great need to identify new active molecules with anticancer activities. Medicinal plants and their bioactive compounds are widely used in the treatment of numerous diseases. The species of the Lamiaceae are considered important because of their use in traditional medicine throughout the world. The Lamiaceae include a range of secondary metabolites including polyphenols, terpenoids, alkaloids, or essential oils that demonstrate promising cytotoxic activity against lung, breast, prostate, and colorectal cancer cell lines mainly via the apoptosis pathway by the modulation of cell cycle progression, changes in genes expression, and impact on various signaling cascades. These biologically active compounds represent promising candidates for supportive use in anticancer therapy; however, further extensive scientific and clinical investigations are required.

Author Contributions

Conceptualization, P.S.; writing—review and editing, P.S., A.M.-S., and T.K.; supervision, T.Ś., and R.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AktProtein Kinase B
ANGAngiogenin
ApafApoptotic Protease Activating Factor
AtgAutophagy-Related Protein
Cdc25CCell Division Cycle 25C Phosphatase
CDKsCyclin-Dependent Kinases
COXCyclooxygenase
DeptorDEP-Domain-Containing mTOR-Interacting Protein
DISCDeath-Inducing Signaling Complex
EGFEpidermal Growth Factor
EGFREpidermal Growth Factor Receptor
ERKExtracellular Signal-Regulated Kinase
FADDFas-Associated Death Domain
Fz receptorFrizzled Receptor
HIFHypoxia-Inducible Factor
hTERTHuman Telomerase Reverse Transcriptase
IKKIkB Kinase
ILInterleukin
LC3Autophagosomal Membrane-Associated Protein Light Chain 3
MAPKMitogen-Activated Protein Kinase
mLST8Mammalian Lethal with Sec13 Protein 8
MMPMatrix Metallopeptidase
mTORMammalian Target of Rapamycin
mTORC1Mammalian Target of Rapamycin Complex 1
NF-κBNuclear Factor Kappa B
PARPPoly(ADP-ribose) Polymerase
PI3KPhosphatidylinositol 3-Kinase
PI3KC3Class IIIPI3K Complex 1
PI3PPhospatydyloinositol-3-Posphate
PRAS40Proline-Rich AKT Substrate 40 kDa
PTENPhosphatase and Tensin Homolog
RANTESRegulated Upon Activation, Normal T-cell Expressed and Secreted
RaptorRegulatory-Associated Protein of mTOR
RhebRas Homolog Enriched in Brain
ROSReactive Oxygen Species
SHP2Src Homology Phosphotyrosine Phosphatase 2
SIRT1Sirtuin
TAKTGF-β-Activated Kinase
TNFTumor Necrosis Factor
TRAILTNF-Related Apoptosis-Inducing Ligand
ULK1Unc-51-like Kinase 1 Complex
VEGFVascular Endothelial Growth Factor

References

  1. Cooper, G.M. Cancer. In The Cell. A Molecular Approach, 2nd ed.; Sinauer Associates: Sunderland, MA, USA, 2000. [Google Scholar]
  2. Sarkar, S.; Horn, G.; Moulton, K.; Oza, A.; Byler, S.; Kokolus, S.; Longacre, M. Cancer development, progression, and therapy: An epigenetic overview. Int. J. Mol. Sci. 2013, 14, 21087–21113. [Google Scholar] [CrossRef] [PubMed]
  3. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  4. WHO. WHO—CancerReport—2020—Global Profile. Available online: https://www.paho.org/hq/index.php?option=com_docman&view=download&category_slug=4-cancer-country-profiles-2020&alias=51561-global-cancer-profile-2020&Itemid=270&lang=fr (accessed on 27 August 2020).
  5. Voda, A.I.; Bostan, I. Public Health Care Financing and the Costs of Cancer Care: A Cross-National Analysis. Cancers 2018, 10, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Pucci, C.; Martinelli, C.; Ciofani, G. Innovative approaches for cancer treatment: Current perspectives and new challenges. Ecancermedicalscience 2019, 13, 961. [Google Scholar] [CrossRef]
  7. Wang, H.; Khor, T.O.; Shu, L.; Su, Z.; Fuentes, F.; Lee, J.H.; Tony Kong, A.H. Plants Against Cancer: A Review on Natural Phytochemicals in Preventing and Treating Cancers and Their Druggability. Anticancer Agents Med. Chem. 2012, 12, 1281–1305. [Google Scholar] [CrossRef] [PubMed]
  8. Tiwari, R.; Rana, C.S. Plant secondary metabolites: A review. Int. J. Eng. Res. Gen. Sci. 2015, 3, 661–670. [Google Scholar]
  9. Iqbal, J.; Abbasi, B.A.; Mahmood, T.; Kanwal, S.; Ali, B.; Shah, S.A.; Khalil, A.T. Plant-derived anticancer agents: A green anticancer approach. Asian Pac. J. Trop. Biomed. 2017, 7, 1129–1150. [Google Scholar] [CrossRef]
  10. Hassanpour, S.H.; Dehghani, M. Review of cancer from perspective of molecular. J. Cancer Res. Pract. 2017, 4, 127–129. [Google Scholar] [CrossRef]
  11. Goodman, S.N.; Samet, J. Cause and Cancer Epidemiology. In Cancer Epidemiology and Prevention, 2nd ed.; Schottenfeld, D., Fraumeni, J.F., Eds.; Oxford University Press: New York, NY, USA, 2017. [Google Scholar]
  12. Baylin, S.B.; Jones, P.A. Epigenetic Determinants of Cancer. Cold Spring Harb. Perspect. Biol. 2016, 8, a019505. [Google Scholar] [CrossRef] [Green Version]
  13. Binder, M.; Roberts, C.; Spencer, N.; Antoine, D.; Cartwright, C. On the antiquity of cancer: Evidence for metastatic carcinoma in a young man from ancient Nubia (c. 1200 BC). PLoS ONE 2014, 9, e90924. [Google Scholar] [CrossRef] [Green Version]
  14. The Global Cancer Observatory. Available online: https://gco.iarc.fr/ (accessed on 27 August 2020).
  15. Vineis, P.; Wild, C.P. Global cancer patterns: Causes and prevention. Lancet 2014, 383, 549–557. [Google Scholar] [CrossRef]
  16. Nagai, H.; Kim, Y.H. Cancer prevention from the perspective of global cancer burden patterns. J. Thorac. Dis. 2017, 9, 448–451. [Google Scholar] [CrossRef] [PubMed]
  17. Thun, M.J.; DeLancey, J.O.; Center, M.M.; Jemal, A.; Ward, E.M. The global burden of cancer: Priorities for prevention. Carcinogenesis. 2010, 31, 100–110. [Google Scholar] [CrossRef] [Green Version]
  18. Bray, F.; Møller, B. Predicting the future burden of cancer. Nat. Rev. Cancer. 2006, 6, 63–74. [Google Scholar] [CrossRef]
  19. Siegel, R.L.; Miller, K.D.; Goding Sauer, A.; Fedewa, S.A.; Butterly, L.F.; Anderson, J.C.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 145–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. de Groot, P.M.; Wu, C.C.; Carter, B.W.; Munden, R.F. The epidemiology of lung cancer. Transl. Lung Cancer Res. 2018, 7, 220–233. [Google Scholar] [CrossRef]
  21. Barta, J.A.; Powell, C.A.; Wisnivesky, J.P. Global epidemiology of lung cancer. Ann. Glob. Health. 2019, 85, 1–16. [Google Scholar] [CrossRef] [Green Version]
  22. Malvezzi, M.; Bosetti, C.; Rosso, T.; Bertuccio, P.; Chatenoud, L.; Levi, F.; Romano, C.; Negri, E.; la Vecchia, C. Lung cancer mortality in European men: Trends and predictions. Lung Cancer 2013, 80, 138–145. [Google Scholar] [CrossRef]
  23. Kim, C.H.; Lee, Y.C.; Hung, R.J.; Boffetta, P.; Xie, D.; Wampfler, J.A.; Cote, M.L.; Chang, S.C.; Ugolini, D.; Neri, M.; et al. Secondhand Tobacco Smoke Exposure and Lung Adenocarcinoma In Situ/Minimally Invasive Adenocarcinoma (AIS/MIA). Cancer Epidemiol. Biomarkers Prev. 2015, 24, 1902–1906. [Google Scholar] [CrossRef] [Green Version]
  24. Boffetta, P.; Pershagen, G.; Jöckel, K.H.; Forastiere, F.; Gaborieau, V.; Heinrich, J.; Jahn, I.; Kreuzer, M.; Merletti, F.; Nyberg, F.; et al. Cigar and pipe smoking and lung cancer risk: A multicenter study from Europe. J. Natl. Cancer Inst. 1999, 91, 697–701. [Google Scholar] [CrossRef] [Green Version]
  25. Schwartz, A.G.; Cote, M.L. Epidemiology of Lung Cancer. Adv. Exp. Med. Biol. 2016, 893, 21–41. [Google Scholar] [CrossRef] [PubMed]
  26. Carper, M.B.; Claudio, P.P. Clinical potential of gene mutations in lung cancer. Clin. Transl. Med. 2015, 4, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zendehdel, M.; Niakan, B.; Keshtkar, A.; Rafiei, E.; Salamat, F. Subtypes of benign breast disease as a risk factor for breast cancer: A systematic review and meta-analysis protocol. Iran. J. Med. Sci. 2018, 43, 1–8. [Google Scholar] [PubMed]
  28. George, B.P.; Abrahamse, H. A Review on Novel Breast Cancer Therapies: Photodynamic Therapy and Plant Derived Agent Induced Cell Death Mechanisms. Anticancer Agents Med Chem. 2016, 16, 793–801. [Google Scholar] [CrossRef]
  29. Malone, K.E.; Daling, J.R.; Thompson, J.D.; O’Brien, C.A.; Francisco, L.V.; Ostrander, E.A. BRCA1 Mutations and Breast Cancer in the General Population. JAMA 1998, 279, 922–929. [Google Scholar] [CrossRef]
  30. Haber, D. Prophylactic oophorectomy to reduce the risk of ovarian and breast cancer in carriers of BRCA mutations. N. Engl. J. Med. 2002, 346, 1660–1662. [Google Scholar] [CrossRef]
  31. Armstrong, N.; Ryder, S.; Forbes, C.; Ross, J.; Quek, R.G.W. A systematic review of the international prevalence of BRCA mutation in breast cancer. Clin. Epidemiol. 2019, 11, 543–561. [Google Scholar] [CrossRef] [Green Version]
  32. Yoshida, K.; Miki, Y. Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage. Cancer Sci. 2004, 95, 11. [Google Scholar] [CrossRef]
  33. Li, X.; Chen, X.; Wen, L.; Wang, Y.; Chen, B.; Xue, Y.; Guo, L.; Liao, N. Impact of TP53 mutations in breast cancer: Clinicopathological features and prognosisImpact of TP53 mutations in breast CA. Thorac. Cancer 2020, 11, 1861–1868. [Google Scholar] [CrossRef]
  34. Taitt, H.E. Global Trends and Prostate Cancer: A Review of Incidence, Detection, and Mortality as Influenced by Race, Ethnicity, and Geographic Location. Am. J. Mens Health 2018, 12, 1807–1823. [Google Scholar] [CrossRef] [Green Version]
  35. Rawla, P. Epidemiology of Prostate Cancer. Med. Nucl. 2008, 32, 2–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cuzick, J.; Thorat, M.A.; Andriole, G.; Brawley, O.W.; Brown, P.W.; Culig, Z.; Eeles, R.A.; Ford, L.G.; Hamdy, F.C.; Holmberg, L.; et al. Prevention and early detection of prostate cancer. Lancet Oncol. 2014, 15, e484–e492. [Google Scholar] [CrossRef] [Green Version]
  37. Attard, G.; Parker, C.; Eeles, R.A.; Schröder, F.; Tomlins, S.A.; Tannock, I.; Drake, C.G.; de Bono, J.S. Prostate cancer. Lancet 2016, 387, 70–82. [Google Scholar] [CrossRef]
  38. Bashir, M.N.; Ahmad, M.R.; Malik, A. Risk factors of prostate cancer: A case-control study in Faisalabad, Pakistan. Asian Pac. J. Cancer Prev. 2014, 15, 10237–10240. [Google Scholar] [CrossRef] [Green Version]
  39. Yao, J.; Chen, Y.; Nguyen, D.T.; Thompson, Z.J.; Eroshkin, A.M.; Nerlakanti, N.; Patel, A.K.; Agarwal, N.; Teer, J.K.; Dhillon, J.; et al. The Homeobox gene, HOXB13, Regulates a Mitotic Protein-Kinase Interaction Network in Metastatic Prostate Cancers. Sci. Rep. 2019, 9, 9715. [Google Scholar] [CrossRef] [Green Version]
  40. Wallis, C.J.D.; Nam, R.K. Prostate Cancer Genetics: A Review. EJIFCC 2015, 26, 79–91. [Google Scholar]
  41. Dong, J.T. Prevalent mutations in prostate cancer. J. Cell. Biochem. 2006, 97, 433–447. [Google Scholar] [CrossRef]
  42. Markowitz, S.D.; Bertagnolli, M.M. Molecular basis of colorectal cancer. N. Engl. J. Med. 2009, 361, 2449–2460. [Google Scholar] [CrossRef] [Green Version]
  43. Kheirelseid, E.A.H.; Miller, N.; Kerin, M.J. Molecular biology of colorectal cancer: Review of the literature. Am. J. Mol. Biol. 2013, 3, 72–80. [Google Scholar] [CrossRef] [Green Version]
  44. Benarba, B.; Pandiella, A. Colorectal cancer and medicinal plants: Principle findings from recent studies. Biomed. Pharmacother. 2018, 107, 408–423. [Google Scholar] [CrossRef]
  45. Watson, A.J.M.; Collins, P.D. Colon cancer: A civilization disorder. Dig. Dis. 2011, 29, 222–228. [Google Scholar] [CrossRef] [PubMed]
  46. Johnson, C.M.; Wei, C.; Ensor, J.E.; Smolenski, D.J.; Amos, C.I.; Levin, B.; Berry, D.A. Meta-analyses of colorectal cancer risk factors. Cancer Causes Control 2013, 24, 1207–1222. [Google Scholar] [CrossRef] [PubMed]
  47. Gallagher, E.J.; LeRoith, D. Epidemiology and molecular mechanisms tying obesity, diabetes, and the metabolic syndrome with cancer. Diabetes Care 2013, 36 (Suppl. 2), S233–S239. [Google Scholar] [CrossRef] [Green Version]
  48. Petrovska, B.B. Historical review of medicinal plants’ usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Stephenson, W. History of Medicine. Br. Med. J. 1957, 2, 413. [Google Scholar] [CrossRef]
  50. Koul, B. Herbs for Cancer Treatment, 1st ed.; Springer: New York, NY, USA, 2019. [Google Scholar]
  51. Maione, F.; Russo, R.; Khan, H.; Mascolo, N. Medicinal plants with anti-inflammatory activities. Nat. Prod. Res. 2016, 30, 1243–1352. [Google Scholar] [CrossRef]
  52. Kasote, D.M.; Katyare, S.S.; Hegde, M.V.; Bae, H. Significance of antioxidant potential of plants and its relevance to therapeutic applications. Int. J. Biol. Sci. 2015, 1, 982–991. [Google Scholar] [CrossRef] [Green Version]
  53. Sitarek, P.; Merecz-Sadowska, A.; Kowalczyk, T.; Wieczfinska, J.; Zajdel, R.; Śliwiński, T. Potential synergistic action of bioactive compounds from plant extracts against skin infecting microorganisms. Int. J. Mol. Sci. 2020, 21, 5105. [Google Scholar] [CrossRef]
  54. Ben-Shabat, S.; Yarmolinsky, L.; Porat, D.; Dahan, A. Antiviral effect of phytochemicals from medicinal plants: Applications and drug delivery strategies. Drug Deliv. Transl. Res. 2020, 10, 354–367. [Google Scholar] [CrossRef] [Green Version]
  55. Zielinska-Blizniewska, H.; Sitarek, P.; Merecz-Sadowska, A.; Malinowska, K.; Zajdel, K.; Jablonska, M.; Sliwinski, T.; Zajdel, R. Plant extracts and reactive oxygen species as two counteracting agents with anti- and pro-obesity properties. Int. J. Mol. Sci. 2019, 20, 4556. [Google Scholar] [CrossRef] [Green Version]
  56. Seca, A.M.L.; Pinto, D.C.G.A. Biological Potential and Medical Use of Secondary Metabolites. Medicines 2019, 6, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wink, M. Modes of Action of Herbal Medicines and Plant Secondary Metabolites. Medicines (Basel) 2015, 2, 251–286. [Google Scholar] [CrossRef]
  58. Wink, M. Functions and Biotechnology of Plant Secondary Metabolites, 2nd ed.; Wiley-Blackwell: Oxford, UK, 2010. [Google Scholar]
  59. Rajeswara Rao, B.; Pandu Sastry, K.; Kumar Kothari, S. Cultivation technology for economically important medicinal plants. In Advances in Medicinal Plants, 1st ed.; Reddy, K.J., Bahadur, B., Bhadraiah, B., Rao, M.L.N., Eds.; Universities Press: Hyderabad, India, 2007. [Google Scholar]
  60. Seca, A.M.L.; Pinto, D.C.G.A. Plant secondary metabolites as anticancer agents: Successes in clinical trials and therapeutic application. Int. J. Mol. Sci. 2018, 19, 263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Greenwell, M.; Rahman, P.K.S.M. Medicinal Plants: Their Use in Anticancer Treatment. Int. J. Pharm. Sci. Res. 2015, 6, 4103–4112. [Google Scholar] [CrossRef] [PubMed]
  62. Sitarek, P.; Synowiec, E.; Kowalczyk, T.; Śliwiński, T.; Skała, E. An In Vitro Estimation of the Cytotoxicity and Genotoxicity of Root Extract from Leonurus sibiricus L. Overexpressing AtPAP1 against Different Cancer Cell Lines. Molecules 2018, 23, 2049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Skała, E.; Sitarek, P.; Toma, M.; Szemraj, J.; Radek, M.; Nieborowska-Skorska, M.; Skorski, T.; Wysokińska, H.; Śliwiński, T. Inhibition of human glioma cell proliferation by altered Bax/Bcl-2-p53 expression and apoptosis induction by Rhaponticum carthamoides extracts from transformed and normal roots. J. Pharm. Pharmacol. 2016, 68, 1454–1464. [Google Scholar] [CrossRef]
  64. Sitarek, P.; Kowalczyk, T.; Santangelo, S.; Białas, A.J.; Toma, M.; Wieczfinska, J.; Śliwiński, T.; Skała, E. The Extract of Leonurus sibiricus Transgenic Roots with AtPAP1 Transcriptional Factor Induces Apoptosis via DNA Damage and Down Regulation of Selected Epigenetic Factors in Human Cancer Cells. Neurochem. Res. 2018, 43, 1363–1370. [Google Scholar] [CrossRef] [Green Version]
  65. Kowalczyk, T.; Sitarek, P.; Skała, E.; Toma, M.; Wielanek, M.; Pytel, D.; Wieczfińska, J.; Szemraj, J.; Śliwiński, T. Induction of apoptosis by in vitro and in vivo plant extracts derived from Menyanthes trifoliata L. in human cancer cells. Cytotechnology 2019, 71, 165–180. [Google Scholar] [CrossRef] [Green Version]
  66. Kowalczyk, T.; Sitarek, P.; Toma, M.; Picot, L.; Wielanek, M.; Skała, E.; Śliwiński, T. An Extract of Transgenic Senna obtusifolia L. hairy roots with Overexpression of PgSS1 Gene in Combination with Chemotherapeutic Agent Induces Apoptosis in the Leukemia Cell Line. Biomolecules 2020, 10, 510. [Google Scholar] [CrossRef] [Green Version]
  67. Gali-Muhtasib, H.; Hmadi, R.; Kareh, M.; Tohme, R.; Darwiche, N. Cell death mechanisms of plant-derived anticancer drugs: Beyond apoptosis. Apoptosis 2015, 20, 1531–1562. [Google Scholar] [CrossRef]
  68. Moudi, M.; Go, R.; Yien, C.Y.S.; Nazre, M. Vinca alkaloids. Int. J. Prev. Med. 2013, 4, 1231–1235. [Google Scholar] [PubMed]
  69. Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Biophys. Acta. 2013, 1830, 3670–3695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Pan, L.; Chai, H.B.; Kinghorn, A.D. Discovery of new anticancer agents from higher plants. Front. Biosci. (Schol Ed.) 2012, 4, 142–156. [Google Scholar] [CrossRef] [PubMed]
  71. Lichota, A.; Gwozdzinski, K. Anticancer activity of natural compounds from plant and marine environment. Int. J. Mol. Sci. 2018, 19, 3533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Uritu, C.M.; Mihai, C.T.; Stanciu, G.D.; Dodi, G.; Alexa-Stratulat, T.; Luca, A.; Leon-Constantin, M.M.; Stefanescu, R.; Bild, V.; Melnic, S.; et al. Medicinal plants of the family Lamiaceae in pain therapy: A review. Pain Res. Manag. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Carović-Stanko, K.; Petek, M.; Grdiša, M.; Pintar, J.; Bedeković, D.; Ćustić, M.H.; Satovic, Z. Medicinal plants of the family Lamiaceae as functional foods-A review. Czech J. Food Sci. 2016, 34, 377–390. [Google Scholar] [CrossRef] [Green Version]
  74. Bekut, M.; Brkić, S.; Kladar, N.; Dragović, G.; Gavarić, N.; Božin, B.B. Potential of selected Lamiaceae plants in anti(retro) viral therapy. Pharmacol. Res. 2018, 133, 301–314. [Google Scholar] [CrossRef]
  75. Stankovic, M. Lamiaceae Species, 1st ed.; MDPI: Basel, Switzerland, 2020. [Google Scholar]
  76. Ganesan, K.; Xu, B. A critical review on polyphenols and health benefits of black soybeans. Nutrients 2017, 9, 455. [Google Scholar] [CrossRef] [Green Version]
  77. Rasouli, H.; Farzaei, M.H.; Khodarahmi, R. Polyphenols and their benefits: A review. Int. J. Food Prop. 2017, 20, 1700–1741. [Google Scholar] [CrossRef] [Green Version]
  78. Alam, M.N.; Almoyad, M.; Huq, F. Polyphenols in Colorectal Cancer: Current State of Knowledge including Clinical Trials and Molecular Mechanism of Action. Biomed. Res. Int. 2018, 2018, 4154185. [Google Scholar] [CrossRef] [Green Version]
  79. Luo, J.; Wei, Z.; Zhang, S.; Peng, X.; Huang, Y.; Zhang, Y.; Lu, J. Phenolic Fractions from Muscadine Grape “Noble” Pomace can Inhibit Breast Cancer Cell MDA-MB-231 Better than those from European Grape “Cabernet Sauvignon” and Induce S-Phase Arrest and Apoptosis. J. Food Sci. 2017, 82, 1254–1263. [Google Scholar] [CrossRef] [PubMed]
  80. Omodanisi, E.I.; Aboua, Y.G.; Oguntibeju, O.O. Assessment of the anti-hyperglycaemic, anti-inflammatory and antioxidant activities of the methanol extract of moringa oleifera in diabetes-induced nephrotoxic male wistar rats. Molecules 2017, 22, 439. [Google Scholar] [CrossRef] [PubMed]
  81. González-Sarrías, A.; Núñez-Sánchez, M.Á.; Tomás-Barberán, F.A.; Espín, J.C. Neuroprotective effects of bioavailable polyphenol-derived metabolites against oxidative stress-induced cytotoxicity in human neuroblastoma SH-SY5Y cells. J. Agric. Food Chem. 2017, 65, 752–758. [Google Scholar] [CrossRef] [Green Version]
  82. Franceschelli, S.; Pesce, M.; Ferrone, A.; Gatta, D.M.P.; Patruno, A.; De Lutiis, M.A.; Quiles, J.L.; Grilli, A.; Felaco, M.; Speranza, L. Biological effect of licochalcone C on the regulation of PI3K/Akt/eNOS and NF-κB/iNOS/NO signaling pathways in H9c2 cells in response to LPS stimulation. Int. J. Mol. Sci. 2017, 18, 690. [Google Scholar] [CrossRef] [PubMed]
  83. Alam, P.; Parvez, M.K.; Arbab, A.H.; Al-Dosari, M.S. Quantitative analysis of rutin, quercetin, naringenin, and gallic acid by validated RP- and NP-HPTLC methods for quality control of anti-HBV active extract of Guiera senegalensis. Pharm. Biol. 2017, 55, 1317–1323. [Google Scholar] [CrossRef] [Green Version]
  84. Ayub, M.A.; Hussain, A.I.; Hanif, M.A.; Chatha, S.A.S.; Kamal, G.M.; Shahid, M.; Janneh, O. Variation in Phenolic Profile, β-Carotene and Flavonoid Contents, Biological Activities of Two Tagetes Species from Pakistani Flora. Chem. Biodivers. 2017, 14, e1600463. [Google Scholar] [CrossRef]
  85. Miyamoto, T.; Zhang, X.; Ueyama, Y.; Apisada, K.; Nakayama, M.; Suzuki, Y.; Ozawa, T.; Mitani, A.; Shigemune, N.; Shimatani, K.; et al. Development of novel monoclonal antibodies directed against catechins for investigation of antibacterial mechanism of catechins. J. Microbiol. Methods 2017, 137, 6–13. [Google Scholar] [CrossRef]
  86. D’Archivio, M.; Santangelo, C.; Scazzocchio, B.; Varì, R.; Filesi, C.; Masella, R.; Giovannini, C. Modulatory effects of polyphenols on apoptosis induction: Relevance for cancer prevention. Int. J. Mol. Sci. 2008, 9, 213–228. [Google Scholar] [CrossRef]
  87. Thomas-Charles, C.; Fennell, H. Anti-Prostate Cancer Activity of Plant-Derived Bioactive Compounds: A Review. Curr. Mol. Biol. Rep. 2019, 5, 140–151. [Google Scholar] [CrossRef] [Green Version]
  88. Perveen, S. Introductory Chapter: Terpenes and Terpenoids. In Terpenes and Terpenoids, 1st ed.; IntechOpen: London, UK, 2018. [Google Scholar]
  89. Hanson, J.R. Diterpenoids of terrestrial origin. Nat. Prod. Rep. 2016, 33, 1227–1238. [Google Scholar] [CrossRef] [Green Version]
  90. Hanson, J.R.; Nichols, T.; Mukhrish, Y.; Bagley, M.C. Diterpenoids of terrestrial origin. Nat. Prod. Rep. 2019, 36, 1499–1512. [Google Scholar] [CrossRef] [PubMed]
  91. Topçu, G.; Yücer, R.; Şenol, H. Bioactive constituents of Anatolian Salvia species. In Salvia Biotechnology, 1st ed.; Georgiev, V., Atanas, P., Eds.; Springer: New York, NY, USA, 2018. [Google Scholar]
  92. Bisio, A.; Pedrelli, F.; D’Ambola, M.; Labanca, F.; Schito, A.M.; Govaerts, R.; De Tommasi, N.; Milella, L. Quinone diterpenes from Salvia species: Chemistry, botany, and biological activity. Phytochem. Rev. 2019, 18, 665–842. [Google Scholar] [CrossRef]
  93. Demetzos, C.; Dimas, K.S. Labdane-type diterpenes: Chemistry and biological activity. Stud. Nat. Prod. Chem. 2001, 25, 235–292. [Google Scholar] [CrossRef]
  94. Banerjee, A.; Laya, M.; Mora, H.; Cabrera, E. The Chemistry of Bioactive Diterpenes. Curr. Org. Chem. 2008, 12, 1050–1070. [Google Scholar] [CrossRef]
  95. Khattak, S.; Khan, H. Anti-cancer Potential of Phyto-alkaloids: A Prospective Review. Curr. Cancer Ther. Rev. 2016, 12, 66–75. [Google Scholar] [CrossRef]
  96. Debnath, B.; Singh, W.S.; Das, M.; Goswami, S.; Singh, M.K.; Maiti, D.; Manna, K. Role of plant alkaloids on human health: A review of biological activities. Mater. Today Chem. 2018, 9, 56–72. [Google Scholar] [CrossRef]
  97. Aniszewski, T. Alkaloids–Secrets of Life: Alkaloid Chemistry, Biological Significance, Applications and Ecological Role, 1st ed.; Elsevier Science: Amsterdam, The Netherlands, 2007. [Google Scholar]
  98. Burnet, M.W.; Goldmann, A.; Message, B.; Drong, R.; El Amrani, A.; Loreau, O.; Slightom, J.; Tepfer, D. The stachydrine catabolism region in Sinorhizobium meliloti encodes a multi-enzyme complex similar to the xenobiotic degrading systems in other bacteria. Gene 2000, 244, 151–161. [Google Scholar] [CrossRef]
  99. 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]
  100. Bhalla, Y.; Gupta, V.K.; Jaitak, V. Anticancer activity of essential oils: A review. J. Sci. Food Agric. 2013, 93, 3643–3653. [Google Scholar] [CrossRef]
  101. Sharifi-Rad, J.; Sureda, A.; Tenore, G.C.; Daglia, M.; Sharifi-Rad, M.; Valussi, M.; Tundis, R.; Sharifi-Rad, M.; Loizzo, M.R.; Oluwaseun Ademiluyi, A.; et al. Biological activities of essential oils: From plant chemoecology to traditional healing systems. Molecules 2017, 22, 70. [Google Scholar] [CrossRef]
  102. Sitarek, P.; Rijo, P.; Garcia, C.; Skała, E.; Kalemba, D.; Białas, A.J.; Szemraj, J.; Pytel, D.; Toma, M.; Wysokińska, H.; et al. Antibacterial, Anti-Inflammatory, Antioxidant, and Antiproliferative Properties of Essential Oils from Hairy and Normal Roots of Leonurus sibiricus L. and Their Chemical Composition. Oxid. Med. Cell Longev. 2017, 2017, 7384061. [Google Scholar] [CrossRef] [Green Version]
  103. Yousef, I.; Oran, S.; Bustanji, Y.; Al Eisawi, D.; Irmaileh, B.A. Cytotoxic Effect of Selected Wild Medicinal Plant Species from Jordan on Two Different Breast Cancer Cell Lines, MCF7 and T47D. Biol. Med. 2018, 10, 4. [Google Scholar] [CrossRef]
  104. Özdemir, A.; Yildiz, M.; Senol, F.S.; Şimay, Y.D.; Ibişoglu, B.; Gokbulut, A.; Orhan, I.E.; Ark, M. Promising anticancer activity of Cyclotrichium niveum L. extracts through induction of both apoptosis and necrosis. Food Chem. Toxicol. 2017, 109, 898–909. [Google Scholar] [CrossRef] [PubMed]
  105. Asnaashari, S.; Delazar, A.; Asgharian, P.; Lotfipour, F.; Moghaddam, S.B.; Afshar, F.H. In-vitro bioactivity and phytochemical screening of extracts from rhizomes of eremostachys azerbaijanica rech. F. Growing in Iran. Iran. J. Pharm. Res. 2017, 16, 306–314. [Google Scholar]
  106. Hoshyar, R.; Mostafavinia, S.E.; Zarban, A.; Hassanpour, M.; Partovfari, M.; Taheri, A.; Pouyan, M. Correlation of Anticancer Effects of 12 Iranian Herbs on Human breast Adenocarcinoma cells with antioxidant Properties. Free Radic. Antioxid. 2015, 5, 65–73. [Google Scholar] [CrossRef] [Green Version]
  107. Amalina, N.D.; Suzery, M.; Cahyono, B. Cytotoxic Activity of Hyptis Pectinate Extracts on MCF-7 Human Breast Cancer Cells. Indones. J. Cancer Chemoprevent. 2020, 11, 1–6. [Google Scholar] [CrossRef]
  108. Santana, F.R.; Luna-Dulcey, L.; Antunes, V.U.; Tormena, C.F.; Cominetti, M.R.; Duarte, M.C.; da Silva, J.A. Evaluation of the cytotoxicity on breast cancer cell of extracts and compounds isolated from Hyptis pectinata (L.) poit. Nat. Prod. Res. 2020, 34, 102–109. [Google Scholar] [CrossRef]
  109. Al-Sheddi, E.S. Cytotoxic potential of Petroleum ether, ethyl acetate, chloroform, and ethanol extracts of Lavandula Coronopifolia against human breast carcinoma cell line (MDA-MB-321). Asian Pac. J. Cancer Prev. 2019, 20, 2943–2949. [Google Scholar] [CrossRef] [PubMed]
  110. Yusufoglu, H.; Foudah, A.I.; Alqarni, M.; Alam, A.; Salkini, A.; Ahmed, E.O. Phenolic contents, cytotoxicity and antimicrobial activity of five medicinal plants of the lamiaceae family obtained from Saudi Arabia local markets. Indo Am. J. Pharm. Sci. 2019, 06, 14418–14425. [Google Scholar] [CrossRef]
  111. Jahanban-Esfahlan, A.; Modaeinama, S.; Abasi, M.; Abbasi, M.M.; Jahanban-Esfahlan, R. Anti proliferative properties of Melissa officinalis in different human cancer cells. Asian Pac. J. Cancer Prev. 2015, 16, 5703–5707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Magalhães, D.B.; Castro, I.; Lopes-Rodrigues, V.; Pereira, J.M.; Barros, L.; Ferreira, I.C.F.R.; Xavier, C.P.R.; Vasconcelos, M.H. Melissa officinalis L. ethanolic extract inhibits the growth of a lung cancer cell line by interfering with the cell cycle and inducing apoptosis. Food Funct. 2018, 9, 3134–3142. [Google Scholar] [CrossRef] [Green Version]
  113. Zengin, G.; Ferrante, C.; Gnapi, D.E.; Sinan, K.I.; Orlando, G.; Recinella, L.; Diuzheva, A.; Jekő, J.; Cziáky, Z.; Chiavaroli, A.; et al. Comprehensive approaches on the chemical constituents and pharmacological properties of flowers and leaves of American basil (Ocimum americanum L). Food Res. Int. 2019, 125, 108610. [Google Scholar] [CrossRef] [PubMed]
  114. Abdelhady, M.I.S.; Motaal, A.A. A cytotoxic C-glycosylated derivative of apigenin from the leaves of Ocimum basilicum var. thyrsiflorum. Braz. J. Pharmacogn. 2016, 26, 763–766. [Google Scholar] [CrossRef] [Green Version]
  115. Makrane, H.; El Messaoudi, M.; Melhaoui, A.; El Mzibri, M.; Benbacer, L.; Aziz, M. Cytotoxicity of the Aqueous Extract and Organic Fractions from Origanum majorana on Human Breast Cell Line MDA-MB-231 and Human Colon Cell Line HT-29. Adv. Pharmacol. Sci. 2018, 2018, 3297193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Aldisi, S.S.; Jaganjac, M.; Eid, A.H.; Goktepe, I. Evaluation of Apoptotic, Antiproliferative, and Antimigratory Activity of Origanum syriacum against Metastatic Colon Cancer Cells. J. Herbs Spices Med. Plants 2019, 25, 202–217. [Google Scholar] [CrossRef]
  117. Pobba, R.; Rama Kotaiah, M.; Chandra Sekar, K.B. Evaluation of antioxidant and anticancer activities of Orthosiphon aristatus (Blume). Int. J. Res. Pharm. Sci. 2015, 6, 193–198. [Google Scholar]
  118. Singh, M.K.; Dhongade, H.; Tripathi, D.K. Orthosiphon pallidus, a potential treatment for patients with breast cancer. J. Pharmacopunct. 2017, 20, 265–273. [Google Scholar] [CrossRef]
  119. Yarmolinsky, L.; Budovsky, A.; Ben-Shabat, S.; Khalfin, B.; Gorelick, J.; Bishitz, Y.; Miloslavski, R.; Yarmolinsky, L. Recent Updates on the Phytochemistry and Pharmacological Properties of Phlomis viscosa Poiret. Rejuvenation Res. 2019, 22, 282–288. [Google Scholar] [CrossRef]
  120. Yulianto, W.; Andarwulan, N.; Giriwono, P.E.; Pamungkas, J. HPLC-based metabolomics to identify cytotoxic compounds from Plectranthus amboinicus (Lour.) Spreng against human breast cancer MCF-7Cells. J. Chromatogr. 2016, 1039, 28–34. [Google Scholar] [CrossRef]
  121. Rai, V.; Pai, V.R.; Kedilaya, P. A preliminary evaluation of anticancer and antioxidant potential of two traditional medicinal plants from lamiaceae-pogostemon heyneanus and plectranthus amboinicus. J. Appl. Pharm. Sci. 2016, 6, 73–78. [Google Scholar] [CrossRef] [Green Version]
  122. Borrás-Linares, I.; Pérez-Sánchez, A.; Lozano-Sánchez, J.; Barrajón-Catalán, E.; Arráez-Román, D.; Cifuentes, A.; Micol, V.; Carretero, A.S. A bioguided identification of the active compounds that contribute to the antiproliferative/cytotoxic effects of rosemary extract on colon cancer cells. Food Chem. Toxicol. 2015, 80, 215–222. [Google Scholar] [CrossRef] [Green Version]
  123. El-burai, H.R. Cytotoxic and Antiproliferative Effects of Four Natural Plants Extracts on Colon Cancer Caco-2 Cell Line. Master’s Thesis, The Islamic University, Gaza, Palestine, 2017. [Google Scholar]
  124. Marrelli, M.; Cristaldi, B.; Menichini, F.; Conforti, F. Inhibitory effects of wild dietary plants on lipid peroxidation and on the proliferation of human cancer cells. Food Chem. Toxicol. 2015, 86, 16–24. [Google Scholar] [CrossRef] [PubMed]
  125. Shen, Y.; Han, J.; Zheng, X.; Ai, B.; Yang, Y.; Xiao, D.; Zheng, L.; Sheng, Z. Rosemary leaf extract inhibits glycation, breast cancer proliferation, and diabetes risks. Appl. Sci. 2020, 10, 2249. [Google Scholar] [CrossRef] [Green Version]
  126. Tundis, R.; Iacopetta, D.; Sinicropi, M.S.; Bonesi, M.; Leporini, M.; Passalacqua, N.G.; Ceramella, J.; Menichini, F.; Loizzo, M.R. Assessment of antioxidant, antitumor and pro-apoptotic effects of Salvia fruticosa Mill. subsp. thomasii (Lacaita) Brullo, Guglielmo, Pavone & Terrasi (Lamiaceae). Food Chem. Toxicol. 2017, 106, 155–164. [Google Scholar] [CrossRef] [PubMed]
  127. Campos-Xolalpa, N.; Alonso-Castro, Á.J.; Sánchez-Mendoza, E.; Zavala-Sánchez, M.Á.; Pérez-Gutiérrez, S. Cytotoxic activity of the chloroform extract and four diterpenes isolated from Salvia ballotiflora. Braz. J. Pharmacogn. 2017, 27, 302–305. [Google Scholar] [CrossRef]
  128. Eltawaty, S.I.; Yagoub, S.O.; Shouman, S.A.; Ahmed, A.; Omer, F.A. Anticancer Effects of Methanol Extract of Libyan Salvia Fruticosa Mill on Mcf7, T47D and (Mda-Mb-468) Breast Cells Lines. Eur. J. Pharm. Med. Res. 2020, 7, 165–169. [Google Scholar]
  129. Altay, A.; Kılıc Suloglu, A.; Sagdıcoglu Celep, G.; Selmanoglu, G.; Bozoglu, F. Anatolıan sage Salvıa frutıcosa ınhıbıts cytosolıc glutathıone-s-transferase actıvıty and colon cancer cell prolıferatıon. J. Food Meas. Charact. 2019, 13, 1390–1399. [Google Scholar] [CrossRef]
  130. Güzel, S.; Ülger, M.; Özay, Y. Antimicrobial and Antiproliferative Activities of Chia (Salvia hispanica L.) Seeds. Int. J. Second. Metab. 2020, 7, 174–180. [Google Scholar] [CrossRef]
  131. Kumar, D.G.; Perumal, P.C.; Kumar, K.; Muthusami, S.; Gopalakrishnan, V.K. Dietary evaluation, antioxidant and cytotoxic activity of crude extract from chia seeds (Salvia hispanica L.) against human prostate cancer cell line (PC-3). Int. J. Pharmacogn. Phytochem. Res. 2016, 8, 1358–1362. [Google Scholar]
  132. Garcia, C.S.C.; Menti, C.; Lambert, A.P.F.; Barcellos, T.; Moura, S.; Calloni, C.; Branco, C.S.; Salvador, M.; Roesch-Ely, M.; Henriques, J.A.P. Pharmacological perspectives from Brazilian Salvia officinalis (Lamiaceae): Antioxidant, and antitumor in mammalian cells. An. Acad. Bras. Cienc. 2016, 88, 281–292. [Google Scholar] [CrossRef] [Green Version]
  133. Darwish, A.; Hamad, G.; Sohaimy, S. Nutrients and Constituents Relevant to Antioxidant, Antimicrobial and Anti-Breast Cancer Properties of Salvia officinalis L. Int. J. Biochem. Res. Rev. 2018, 23, 1–13. [Google Scholar] [CrossRef]
  134. Yumrutaş, Ö.; Pehlivan, M.; Güven, C.; Bozgeyik, İ.; Bozgeyik, E.; Temiz, E.; Yumrutaş, P.; Üçkardeş, F. Investigation of Cytotoxic Effect of Salvia pilifera Extracts and Synthetic Chlorogenic and Caffeic Acids on DU145 Prostate Cancer Cells Line. KSU J. Agric Nat. 2018, 21, 141–147. [Google Scholar] [CrossRef]
  135. Güzel, S.; Kahraman, A.; Ülger, M.; Özay, Y.; Bozgeyik, İ.; Sarikaya, Ö. Morphology, myxocarpy, mineral content and in vitro antimicrobial and antiproliferative activities of mericarps of the vulnerable Turkish endemic Salvia pilifera. J. Res. Pharm. 2019, 23, 729–739. [Google Scholar] [CrossRef] [Green Version]
  136. Al-Zereini, W.A. Ononis natrix and Salvia verbenaca: Two Jordanian Medicinal Plants with Cytotoxic and Antibacterial Activities. J. Herbs Spices Med. Plants. 2017, 23, 18–25. [Google Scholar] [CrossRef]
  137. Yfanti, P.; Batistatou, A.; Manos, G.; Lekka, M.E. The Aromatic Plant Satureja horvatii ssp. macrophylla Induces Apoptosis and Cell Death to the A549 Cancer Cell Line. Am. J. Plant Sci. 2015, 6, 2092–2103. [Google Scholar] [CrossRef] [Green Version]
  138. Demirelma, H.; Gelinci, E. Determination of the cytotoxic effect on human colon cancer and phe nolic substance cont ent of the endemic species sideritis ozturkii Aytaç & Aksoy. Appl. Ecol. Environ. Res. 2019, 17, 7407–7419. [Google Scholar] [CrossRef]
  139. Yumrutas, O.; Oztuzcu, S.; Pehlivan, M.; Ozturk, N.; Eroz Poyraz, I.; Iğci, Y.Z.; Cevik, M.O.; Bozgeyik, I.; Aksoy, A.F.; Bagis, H.; et al. Cell viability, anti-proliferation and antioxidant activities of Sideritis syriaca, Tanacetum argenteum sub sp. argenteum and Achillea aleppica subsp. zederbaueri on human breast cancer cell line (MCF-7). J. Appl. Pharm. Sci. 2015, 5, 1–5. [Google Scholar] [CrossRef] [Green Version]
  140. Calderón-Montaño, J.M.; Martínez-Sánchez, S.M.; Burgos-Morón, E.; Guillén-Mancina, E.; Jiménez-Alonso, J.J.; García, F.; Aparicio, A.; López-Lázaro, M. Screening for Selective Anticancer Activity of 65 Extracts of Plants Collected in Western Andalusia, Spain. Preprints 2018, 2018060177. [Google Scholar] [CrossRef] [Green Version]
  141. Sadeghisamani, F.; Sazgar, H.; Ghasemi Pirbalouti, A. Investigation Cytotoxic Effect of Hydroalcholic Extract from Combination of Kelussia odaratissma Mozaff and Thymus daenesis Celak on MCF-7 Cancer Cells Line. J. Jahrom Univ. Med Sci. 2016, 14, 59–67. [Google Scholar] [CrossRef] [Green Version]
  142. Garbi, M.I.; Osman, E.E.; Kabbashi, A.S.; Saleh, M.S.; Yuosof, Y.S.; Mahmoud, S.A.; Salam, H.A.A. Cytotoxicity of Vitex trifolia leaf extracts on MCF-7 and Vero cell lines. J. Sci. Innov. Res. 2015, 4, 89–93. [Google Scholar]
  143. Abu-Gharbieh, E.; El-Huneidi, W.; Shehab, N.G.; Bajbouj, K.; Vinod, A.; El-Serafi, A.; Malhab, L.B.; Abdel-Rahman, W.M. Anti-tumor activity of the ethanolic extract of Micromeria fruticosa on human breast and colon cancer cells. FASEB J. 2020, 34. [Google Scholar] [CrossRef]
  144. El-Huneidi, W.; Shehab, N.G.; Bajbouj, K.; Vinod, A.; El-serafi, A.; Shafarin, J.; Boumalhab, L.J.; Abdel-rahman, W.M.; Abu-gharbieh, E. Micromeriafruticosa induces cell cycle arrest and apoptosis in breast and colorectal cancer cells. Pharmaceuticals 2020, 13, 115. [Google Scholar] [CrossRef] [PubMed]
  145. Weidner, C.; Rousseau, M.; Plauth, A.; Wowro, S.J.; Fischer, C.; Abdel-Aziz, H.; Sauer, S. Melissa officinalis extract induces apoptosis and inhibits proliferation in colon cancer cells through formation of reactive oxygen species. Phytomedicine 2015, 22, 262–270. [Google Scholar] [CrossRef] [PubMed]
  146. Song, H.M.; Park, G.H.; Park, S.B.; Kim, H.S.; Son, H.J.; Um, Y.; Jeong, J.B. Vitex rotundifolia Fruit Suppresses the Proliferation of Human Colorectal Cancer Cells through Down-regulation of Cyclin D1 and CDK4 via Proteasomal-Dependent Degradation and Transcriptional Inhibition. Am. J. Chin. Med. 2018, 46, 191–207. [Google Scholar] [CrossRef] [PubMed]
  147. Chen, J. The cell-cycle arrest and apoptotic functions of p53 in tumor initiation and progression. Cold Spring Harb. Perspect. Med. 2016, 6, a026104. [Google Scholar] [CrossRef]
  148. Elansary, H.O.; Mahmoud, E.A. In vitro antioxidant and antiproliferative activities of six international basil cultivars. Nat. Prod. Res. 2015, 29, 2149–2154. [Google Scholar] [CrossRef]
  149. Ye, Y.T.; Zhong, W.; Sun, P.; Wang, D.; Wang, C.; Hu, L.M.; Qian, J.Q. Apoptosis induced by the methanol extract of Salvia miltiorrhiza Bunge in non-small cell lung cancer through PTEN-mediated inhibition of PI3K/Akt pathway. J. Ethnopharmacol. 2017, 200, 107–116. [Google Scholar] [CrossRef]
  150. Gao, W.; Xu, H.L.Y.L.Y.L.Y. Root extract of Prunella vulgaris inhibits in vitro and in vivo carcinogenesis in MCF-5 human breast carcinoma via suppression of angiogenesis, induction of apoptosis, cell cycle arrest and modulation of PI3K/AKT signalling pathway. J. BUON 2019, 24, 549–554. [Google Scholar]
  151. Khojasteh, A.; Metón, I.; Camino, S.; Cusido, R.M.; Eibl, R.; Palazon, J. In Vitro Study of the Anticancer Effects of Biotechnological Extracts of the Endangered Plant Species Satureja Khuzistanica. Int. J. Mol. Sci. 2019, 20, 2400. [Google Scholar] [CrossRef] [Green Version]
  152. Sridevi, M.; Bright, J.; Yamini, K. Anti-cancer effect of ocimum-sanctum ethanolic extract in non-small cell lung carcinoma cell line. Int. J. Pharm. Pharm. Sci. 2016, 8, 8–20. [Google Scholar]
  153. Chen, C.C.; Kao, C.P.; Chiu, M.M.; Wang, S.H. The anti-cancer effects and mechanisms of Scutellaria barbata D. Don on CL1-5 lung cancer cells. Oncotarget 2017, 8, 109340–109357. [Google Scholar] [CrossRef] [PubMed]
  154. Kokhdan, E.P.; Sadeghi, H.; Ghafoori, H.; Sadeghi, H.; Danaei, N.; Javadian, H.; Aghamaali, M.R. Cytotoxic effect of methanolic extract, alkaloid and terpenoid fractions of Stachys pilifera against HT-29 cell line. Res. Pharm. Sci. 2018, 13, 404–412. [Google Scholar] [CrossRef] [PubMed]
  155. Sicora, O.; Naghi, M.A.; Soos, I.; Sicora, C. The ethanolic stem extract of Caryopteris x Clandonensis posseses antiproliferative potential by blocking breast cancer cells in mitosis. Farmacia 2019, 67, 1077–1082. [Google Scholar] [CrossRef] [Green Version]
  156. Panicker, N.G.; Balhamar, S.O.M.S.; Akhlaq, S.; Qureshi, M.M.; Rizvi, T.S.; AlHarrasi, A.; Hussain, J.; Mustafa, F. Identification and Characterization of the Caspase-Mediated Apoptotic Activity of Teucrium mascatense and an Isolated Compound in Human Cancer Cells. Molecules 2019, 24, 977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Fadok, V.A.; Bratton, D.L.; Frasch, S.C.; Warner, M.L.; Henson, P.M. The role of phosphatidylserine in recognition of apoptotic cells by phagocytes. Cell Death Differ. 1998, 5, 551–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Grujičić, D.; Marković, A.; Vukajlović, J.T.; Stanković, M.; Jakovljević, M.R.; Ćirić, A.; Djordjević, K.; Planojević, N.; Milutinović, M.; Milošević-Djordjević, O. Genotoxic and cytotoxic properties of two medical plants (Teucrium arduini L.and Teucrium flavum L.) in relation to their polyphenolic contents. Mutat. Res. 2020, 852, 503168. [Google Scholar] [CrossRef] [PubMed]
  159. Chaudhry, G.E.S.; Jan, R.; Mohamad, H.; Tengku Muhammad, T. Vitex rotundifolia fractions induce apoptosis in human breast cancer cell line, MCF-7, via extrinsic and intrinsic pathways. Res. Pharm. Sci. 2019, 14, 273–285. [Google Scholar] [CrossRef] [PubMed]
  160. Dhandayuthapani, S.; Azad, H.; Rathinavelu, A. Apoptosis Induction by Ocimum sanctum Extract in LNCaP Prostate Cancer Cells. J. Med. Food. 2015, 18, 776–785. [Google Scholar] [CrossRef]
  161. Geryani, M.A.; Mahdian, D.; Mousavi, S.H.; Hosseini, A. Ctotoxic and apoptogenic effects of Perovskia abrotanoides flower extract on MCF-7 and HeLa cell lines. Avicenna J. Phytomed. 2016, 6, 410–417. [Google Scholar]
  162. Golshan, A.; Amini, E.; Emami, S.A.; Asili, J.; Jalali, Z.; Sabouri-Rad, S.; Sanjar-Mousavi, N.; Tayarani-Najaran, Z. Cytotoxic evaluation of different fractions of Salvia chorassanica Bunge on MCF-7 and du 145 cell lines. Res. Pharm. Sci. 2016, 11, 73–80. [Google Scholar]
  163. Tarhan, L.; Nakipoğlu, M.; Kavakcıoğlu, B.; Tongul, B.; Nalbantsoy, A. The Induction of Growth Inhibition and Apoptosis in HeLa and MCF-7 Cells by Teucrium sandrasicum, Having Effective Antioxidant Properties. Appl. Biochem. Biotechnol. 2016, 178, 1028–1041. [Google Scholar] [CrossRef] [PubMed]
  164. Mohammed, H.; Arab, F. Antiproliferative activity of Origanum compactum extract on lung cancer and hepatoma cells. Arab. J. Med. Aromat. Plants. 2015, 1, 44–56. [Google Scholar] [CrossRef]
  165. Kim, H.I.; Hong, S.H.; Ku, J.M.; Lim, Y.S.; Lee, S.J.; Song, J.; Kim, T.Y.; Cheon, C.; Ko, S.G. Scutellaria radix promotes apoptosis in non-small cell lung cancer cells via induction of AMPK-dependent autophagy. Am. J. Chin. Med. 2019, 47, 691–705. [Google Scholar] [CrossRef] [PubMed]
  166. Jahanban-Esfahlan, R.; Seidi, K.; Monfaredan, A.; Shafie-Irannejad, V.; Abbasi, M.M.; Karimian, A.; Yousefi, B. The herbal medicine Melissa officinalis extract effects on gene expression of p53, Bcl-2, Her2, VEGF-A and hTERT in human lung, breast and prostate cancer cell lines. Gene 2017, 613, 14–19. [Google Scholar] [CrossRef] [PubMed]
  167. Laila, F.; Fardiaz, D.; Yuliana, N.D.; Damanik, M.R.M.; Nur Annisa Dewi, F. Methanol Extract of Coleus amboinicus (Lour) Exhibited Antiproliferative Activity and Induced Programmed Cell Death in Colon Cancer Cell WiDr. Int. J. Food Sci. 2020. [Google Scholar] [CrossRef] [Green Version]
  168. Saravanan, R.; Pemaiah, B.; Sridharan, S.; Narayanan, M.; Ramalingam, S. Enhanced cytotoxic potential of Orthosiphon stamineus extract in MCF-7 cells through suppression of nucleolin and BCL2. Bangladesh J. Pharmacol. 2017, 12, 268–275. [Google Scholar] [CrossRef] [Green Version]
  169. Lee, M.J.; Lee, S.; Choi, N.R.; Jo, S.H.; Cho, S. Dracocephalum palmatum Stephan on human-derived prostate cancer cell death. Kor. J. Herbology. 2018, 33, 69–76. [Google Scholar] [CrossRef]
  170. Milutinović, M.G.; Maksimović, V.M.; Cvetković, D.M.; Nikodijević, D.D.; Stanković, M.S.; Pešić, M.; Marković, S.D. Potential of Teucrium chamaedrys L. to modulate apoptosis and biotransformation in colorectal carcinoma cells. J. Ethnopharmacol. 2019, 240, 111951. [Google Scholar] [CrossRef]
  171. Benhalilou, N.; Alsamri, H.; Alneyadi, A.; Athamneh, K.; Alrashedi, A.; Altamimi, N.; Dhaheri, Y.A.; Eid, A.H.; Iratni, R. Origanum majorana ethanolic extract promotes colorectal cancer cell death by triggering abortive autophagy and activation of the extrinsic apoptotic pathway. Front. Oncol. 2019, 9, 795. [Google Scholar] [CrossRef]
  172. Emami, S.A.; Asili, J.; HosseinNia, S.; Yazdian-Robati, R.; Sahranavard, M.; Tayarani-Najaran, Z. Growth inhibition and apoptosis induction of essential oils and extracts of Nepeta cataria L. on human prostatic and breast cancer cell lines. Asian Pac. J. Cancer Prev. 2016, 17, 125–130. [Google Scholar] [CrossRef]
  173. Boulares, A.H.; Yakovlev, A.G.; Ivanova, V.; Stoica, B.A.; Wang, G.; Iyer, S.; Smulson, M. Role of Poly(ADP-ribose) Polymerase (PARP) Cleavage in Apoptosis. J. Biol. Chem. 1999, 274, 22932–22940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Moore, J.; Megaly, M.; MacNeil, A.J.; Klentrou, P.; Tsiani, E. Rosemary extract reduces Akt/mTOR/p70S6K activation and inhibits proliferation and survival of A549 human lung cancer cells. Biomed. Pharmacother. 2016, 83, 725–732. [Google Scholar] [CrossRef]
  175. Wise, J.F.; Berkova, Z.; Mathur, R.; Zhu, H.; Braun, F.K.; Tao, R.H.; Sabichi, A.L.; Ao, X.; Maeng, H.; Samaniego, F. Nucleolin inhibits Fas ligand binding and suppresses Fas-mediated apoptosis in vivo via a surface nucleolin-Fas complex. Blood 2013, 121, 4729–4739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Wattanathamsan, O.; Hayakawa, Y.; Pongrakhananon, V. Molecular mechanisms of natural compounds in cell death induction and sensitization to chemotherapeutic drugs in lung cancer. Phyther. Res. 2019, 33, 2531–2547. [Google Scholar] [CrossRef]
  177. Jan, R.; Chaudhry, G.e.S. Understanding apoptosis and apoptotic pathways targeted cancer therapeutics. Adv. Pharm. Bull. 2019, 9, 205–218. [Google Scholar] [CrossRef] [Green Version]
  178. Jaiswal, P.K.; Goel, A.; Mittal, R.D. Survivin: A molecular biomarker in cancer. Indian J. Med. Res. 2015, 141, 389–397. [Google Scholar] [CrossRef]
  179. Wei, L.H.; Lin, J.M.; Chu, J.F.; Chen, H.W.; Li, Q.Y.; Peng, J. Scutellaria barbata D. Don inhibits colorectal cancer growth via suppression of Wnt/β-catenin signaling pathway. Chin. J. Integr. Med. 2017, 23, 858–863. [Google Scholar] [CrossRef]
  180. Castedo, M.; Ferri, K.F.; Kroemer, G. Mammalian Target of Rapamycin (mTOR): Pro- and Anti-Apoptotic. Cell Death Differ. 2002, 9, 99–100. [Google Scholar] [CrossRef]
  181. Torres, R.G.; Casanova, L.; Carvalho, J.; Marcondes, M.C.; Costa, S.S.; Sola-Penna, M.; Zancan, P. Ocimum basilicum but not Ocimum gratissimum present cytotoxic effects on human breast cancer cell line MCF-7, inducing apoptosis and triggering mTOR/Akt/p70S6K pathway. J. Bioenerg. Biomembr. 2018, 50, 93–105. [Google Scholar] [CrossRef]
  182. Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef] [Green Version]
  183. Chang, F.; Lee, J.T.; Navolanic, P.M.; Steelman, L.S.; Shelton, J.G.; Blalock, W.L.; Franklin, R.A.; McCubrey, J.A. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: A target for cancer chemotherapy. Leukemia 2003, 17, 590–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Fan, J.; Bao, Y.; Meng, X.; Wang, S.; Li, T.; Chang, X.; Yang, G.; Bo, T. Mechanism of modulation through PI3K-AKT pathway about Nepeta cataria L.’s extract in non-small cell lung cancer. Oncotarget 2017, 8, 31395–31405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Lin, Y.; Bai, L.; Chen, W.; Xu, S. The NF-κB activation pathways, emerging molecular targets for cancer prevention and therapy. Expert Opin. Ther. Targets. 2010, 14, 45–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Oliviero, M.; Romilde, I.; Beatrice, M.M.; Matteo, V.; Giovanna, N.; Consuelo, A.; Claudio, C.; Giorgio, S.; Maggi, F.; Massimo, N. Evaluations of thyme extract effects in human normal bronchial and tracheal epithelial cell lines and in human lung cancer cell line. Chem. Biol. Interact. 2016, 256, 125–133. [Google Scholar] [CrossRef]
  188. Bakhle, Y.S.; Botting, R.M. Cyclooxygenase-2 and its regulation in inflammation. Mediators Inflamm. 1996, 5, 305–323. [Google Scholar] [CrossRef]
  189. Uzunhisarcikli, E.; Gürbüz, P.; Yerer, M.B. Investigation of Antiinflamatory Effects of Origanum majorana L. Extract in LPS-Induced Beas-2b and A549 Cells. Proceedings 2019, 40, 8. [Google Scholar] [CrossRef] [Green Version]
  190. Lim, S.; Kaldis, P. Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development 2013, 140, 3079–3093. [Google Scholar] [CrossRef] [Green Version]
  191. MacDonald, B.T.; Tamai, K.; He, X. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev. Cell. 2009, 17, 9–26. [Google Scholar] [CrossRef] [Green Version]
  192. He, B.; Reguart, N.; You, L.; Mazieres, J.; Xu, Z.; Lee, A.Y.; Mikami, I.; McCormick, F.; Jablons, D.M. Blockade of Wnt-1 signaling induces apoptosis in human colorectal cancer cells containing downstream mutations. Oncogene. 2005, 24, 3054–3058. [Google Scholar] [CrossRef] [Green Version]
  193. Miller, D.M.; Thomas, S.D.; Islam, A.; Muench, D.; Sedoris, K. c-Myc and cancer metabolism. Clin. Cancer Res. 2012, 18, 5546–5553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Yu, L.; Chen, Y.; Tooze, S.A. Autophagy pathway: Cellular and molecular mechanisms. Autophagy 2018, 14, 207–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Laplante, M.; Sabatini, D.M. mTOR signaling at a glance. J. Cell Sci. 2009, 122, 3589–3594. [Google Scholar] [CrossRef] [Green Version]
  197. Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [Green Version]
  198. Morales, J.C.; Li, L.; Fattah, F.J.; Dong, Y.; Bey, E.A.; Patel, M.; Gao, J.; Boothman, D.A. Review of poly (ADP-ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases. Crit. Rev. Eukaryot. Gene Expr. 2014, 21, 15–28. [Google Scholar] [CrossRef] [Green Version]
  199. Zong, W.X.; Thompson, C.B. Necrotic death as a cell fate. Genes Dev. 2006, 20, 1–15. [Google Scholar] [CrossRef] [Green Version]
  200. Henriquez, M.; Armisen, R.; Stutzin, A.; Quest, A.F.G. Cell Death by Necrosis, a Regulated Way to go. Curr. Mol. Med. 2008, 8, 187–206. [Google Scholar] [CrossRef]
  201. Pérez-Sánchez, A.; Barrajón-Catalán, E.; Ruiz-Torres, V.; Agulló-Chazarra, L.; Herranz-López, M.; Valdés, A.; Cifuentes, A.; Micol, V. Rosemary (Rosmarinus officinalis) extract causes ROS-induced necrotic cell death and inhibits tumor growth in vivo. Sci. Rep. 2019, 9, 808. [Google Scholar] [CrossRef]
  202. Ghiulai, R.; Avram, S.; Stoian, D.; Pavel, I.Z.; Coricovac, D.; Oprean, C.; Vlase, L.; Farcas, C.; Mioc, M.; Minda, D.; et al. Lemon Balm Extracts Prevent Breast Cancer Progression in Vitro and in Ovo on Chorioallantoic Membrane Assay. Evid. Based Complement. Altern. Med. 2020, 2020, 6489159. [Google Scholar] [CrossRef] [Green Version]
  203. Van Cruijsen, H.; Giaccone, G.; Hoekman, K. Epidermal growth factor receptor and angiogenesis: Opportunities for combined anticancer strategies. Int. J. Cancer. 2005, 117, 883–888. [Google Scholar] [CrossRef] [PubMed]
  204. Folkman, J. Tumor Angiogenesis: From Bench to Bedside. In Tumor Angiogenesis: Basic Mechanisms and Cancer Therapy, 1st ed.; Marmé, D., Fusenig, N., Eds.; Springer: New York, NY, USA, 2008; pp. 3–28. [Google Scholar]
  205. Atmaca, H.; Bozkurt, E. Apoptotic and anti-angiogenic effects of Salvia triloba extract in prostate cancer cell lines. Tumor Biol. 2016, 37, 3639–3646. [Google Scholar] [CrossRef] [PubMed]
  206. Li, A.; Dubey, S.; Varney, M.L.; Dave, B.J.; Singh, R.K. IL-8 Directly Enhanced Endothelial Cell Survival, Proliferation, and Matrix Metalloproteinases Production and Regulated Angiogenesis. J. Immunol. 2003, 170, 3369–3376. [Google Scholar] [CrossRef] [PubMed]
  207. Tahergorabi, Z.; Khazaei, M. Leptin and its cardiovascular effects: Focus on angiogenesis. Adv. Biomed. Res. 2015, 4, 79. [Google Scholar] [CrossRef]
  208. Suffee, N.; Richard, B.; Hlawaty, H.; Oudar, O.; Charnaux, N.; Sutton, A. Angiogenic properties of the chemokine RANTES/CCL5. Biochem. Soc. Trans. 2011, 39, 1649–1653. [Google Scholar] [CrossRef] [Green Version]
  209. Shestenko, O.P.; Nikonov, S.D.; Mertvetsov, N.P. Angiogenin and its functions in angiogenesis. Mol. Biol. 2001, 35, 294–314. [Google Scholar] [CrossRef]
  210. Begley, L.A.; Kasina, S.; Mehra, R.; Adsule, S.; Admon, A.J.; Lonigro, R.J.; Chinnaiyan, A.M.; Macoska, J.A. CXCL5 promotes prostate cancer progression. Neoplasia 2008, 10, 244–254. [Google Scholar] [CrossRef] [Green Version]
  211. Zhao, S.M.; Chou, G.X.; Yang, Q.S.; Wang, W.; Zhou, J.L. Abietane diterpenoids from Caryopteris incana (Thunb.) Miq. Org. Biomol. Chem. 2016, 14, 3510–3520. [Google Scholar] [CrossRef]
  212. Luo, Y.; Cheng, L.Z.; Luo, Q.; Yan, Y.M.; Wang, S.M.; Sun, Q.; Cheng, Y.X. New ursane-type triterpenoids from Clerodendranthus spicatus. Fitoterapia 2017, 119, 69–74. [Google Scholar] [CrossRef]
  213. Somwong, P.; Suttisri, R. Cytotoxic activity of the chemical constituents of Clerodendrum indicum and Clerodendrum villosum roots. J. Integr. Med. 2018, 16, 57–61. [Google Scholar] [CrossRef]
  214. Ba Vinh, L.; Thi Minh Nguyet, N.; Young Yang, S.; Hoon Kim, J.; Thi Vien, L.; Thi Thanh Huong, P.; Van Thanh, N.; Xuan Cuong, N.; Hoai Nam, N.; Van Minh, C.; et al. A new rearranged abietane diterpene from Clerodendrum inerme with antioxidant and cytotoxic activities. Nat. Prod. Res. 2018, 32, 2002–2007. [Google Scholar] [CrossRef] [PubMed]
  215. Xu, M.; Wang, S.; Jia, O.; Zhu, Q.; Shi, L. Bioactive diterpenoids from clerodendrum kiangsiense. Molecules 2016, 21, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Awouafack, M.D.; Aimaiti, S.; Tane, P.; Morita, H. Clerodendrumol, a new triterpenoid from Clerodendrum yaundense Gürke (Lamiaceae). Helv. Chim. Acta 2016, 99, 161–164. [Google Scholar] [CrossRef]
  217. Dai, L.P.; Li, C.; Yang, H.Z.; Lu, Y.Q.; Yu, H.Y.; Gao, H.M.; Wang, Z.M. Three new cytotoxic ent-kaurane diterpenes from isodon excisoides. Molecules 2015, 20, 17544–17556. [Google Scholar] [CrossRef] [Green Version]
  218. Liu, Y.L.; Zhang, L.X.; Wu, H.; Chen, S.Q.; Li, J.; Dai, L.P.; Wang, Z.M. Four New ent-Kaurane Diterpene Glycosides from Isodon henryi. Molecules 2019, 24, 2736. [Google Scholar] [CrossRef] [Green Version]
  219. Yang, J.; Wang, W.G.; Wu, H.Y.; Du, X.; Li, X.N.; Li, Y.; Pu, J.X.; Sun, H.D. Bioactive Enmein-Type ent-Kaurane Diterpenoids from Isodon phyllostachys. J. Nat. Prod. 2016, 79, 132–140. [Google Scholar] [CrossRef]
  220. Luo, G.Y.; Deng, R.; Zhang, J.J.; Ye, J.H.; Pan, L.T. Two cytotoxic 6,7-seco-spiro-lacton-ent-kauranoids from Isodon rubescens. J. Asian Nat. Prod. Res. 2018, 20, 227–233. [Google Scholar] [CrossRef]
  221. Wu, H.Y.; Wang, W.G.; Du, X.; Yang, J.; Pu, J.X.; Sun, H.D. Six new cytotoxic and anti-inflammatory 11, 20-epoxy-ent-kaurane diterpenoids from Iso Isodon wikstroemioides. Chin. J. Nat. Med. 2015, 13, 383–389. [Google Scholar] [CrossRef]
  222. Peng, W.; Huo, G.; Zheng, L.; Xiong, Z.; Shi, X.; Peng, D. Two new oleanane derivatives from the fruits of Leonurus japonicus and their cytotoxic activities. J. Nat. Med. 2019, 73, 252–256. [Google Scholar] [CrossRef]
  223. To, D.C.; Hoang, D.T.; Tran, M.H.; Pham, M.Q.; Huynh, N.T.; Nguyen, P.H. PTP1B Inhibitory Flavonoids From Orthosiphon stamineus Benth. and Their Growth Inhibition on Human Breast Cancer Cells. Nat. Prod. Commun. 2020, 15, 1934578X19899517. [Google Scholar] [CrossRef]
  224. Sajjadi, S.; Delazari, Z.; Aghaei, M.; Ghannadian, M. Flavone constituents of Phlomis bruguieri Desf. with cytotoxic activity against MCF-7 breast cancer cells. Res. Pharm. Sci. 2018, 13, 422–429. [Google Scholar] [PubMed]
  225. Le, D.D.; Nguyen, D.H.; Zhao, B.T.; Kim, J.A.; Kim, S.K.; Min, B.S.; Choi, J.S.; Woo, M.H. 28-Noroleanane-derived spirocyclic triterpenoids and iridoid glucosides from the roots of Phlomoides umbrosa (Turcz.) Kamelin & Makhm with their cytotoxic effects. Phytochemistry 2018, 153, 138–146. [Google Scholar] [CrossRef] [PubMed]
  226. Amina, M.; Alam, P.; Parvez, M.K.; Al-Musayeib, N.M.; Al-Hwaity, S.A.; Al-Rashidi, N.S.; Al-Dosari, M.S. Isolation and validated HPTLC analysis of four cytotoxic compounds, including a new sesquiterpene from aerial parts of Plectranthus cylindraceus. Nat. Prod. Res. 2018, 32, 804–809. [Google Scholar] [CrossRef] [PubMed]
  227. Saraiva, N.; Costa, J.G.; Reis, C.; Almeida, N.; Rijo, P.; Fernandes, A.S. Anti-migratory and pro-apoptotic properties of parvifloron D on triple-negative breast cancer cells. Biomolecules 2020, 10, 158. [Google Scholar] [CrossRef] [Green Version]
  228. Matias, D.; Nicolai, M.; Saraiva, L.; Pinheiro, R.; Faustino, C.; Diaz Lanza, A.; Pinto Reis, C.; Stankovic, T.; Dinic, J.; Pesic, M.; et al. Cytotoxic Activity of Royleanone Diterpenes from Plectranthus madagascariensis Benth. ACS Omega 2019, 4, 8094–8103. [Google Scholar] [CrossRef] [Green Version]
  229. Ito, T.; Rakainsa, S.K.; Nisa, K.; Morita, H. Three new abietane-type diterpenoids from the leaves of Indonesian Plectranthus scutellarioides. Fitoterapia 2018, 127, 146–150. [Google Scholar] [CrossRef]
  230. Nguyen, H.T.; Tran, L.T.T.; Ho, D.V.; Le, D.V.; Raal, A.; Morita, H. Pogostemins A-C, three new cytotoxic meroterpenoids from Pogostemon auricularius. Fitoterapia 2018, 130, 100–104. [Google Scholar] [CrossRef]
  231. Kim, K.H.; Beemelmanns, C.; Clardy, J.; Cao, S. A new antibacterial octaketide and cytotoxic phenylethanoid glycosides from Pogostemon cablin (Blanco) Benth. Bioorg. Med. Chem. Lett. 2015, 25, 2834–2836. [Google Scholar] [CrossRef]
  232. Elmaidomy, A.H.; Mohammed, R.; Hassan, H.M.; Owis, A.I.; Rateb, M.E.; Khanfar, M.A.; Krischke, M.; Mueller, M.J.; Abdelmohsen, U.R. Metabolomic profiling and cytotoxic tetrahydrofurofuran lignans investigations from Premna odorata Blanco. Metabolites 2019, 9, 223. [Google Scholar] [CrossRef] [Green Version]
  233. Wang, W.Q.; Xuan, L.J. Ent-6,7-Secokaurane diterpenoids from Rabdosia serra and their cytotoxic activities. Phytochemistry 2016, 122, 119–125. [Google Scholar] [CrossRef]
  234. Esquivel, B.; Bustos-Brito, C.; Sánchez-Castellanos, M.; Nieto-Camacho, A.; Ramírez-Apan, T.; Joseph-Nathan, P.; Quijano, L. Structure, absolute configuration, & antiproliferative activity of abietane & icetexane diterpenoids from salvia ballotiflora. Molecules 2017, 22, 1690. [Google Scholar] [CrossRef] [Green Version]
  235. Mirzaei, H.H.; Firuzi, O.; Baldwin, I.T.; Jassbi, A.R. Cytotoxic activities of different iranian solanaceae and lamiaceae plants and bioassay-guided study of an active extract from salvia lachnocalyx. Nat. Prod. Commun. 2017, 12, 1563–1566. [Google Scholar] [CrossRef] [Green Version]
  236. Farimani, M.M.; Taleghani, A.; Aliabadi, A.; Aliahmadi, A.; Esmaeili, M.A.; Sarvestani, N.N.; Khavasi, H.R.; Smieško, M.; Hamburger, M.; Ebrahimi, S.N. Labdane diterpenoids from Salvia leriifolia: Absolute configuration, antimicrobial and cytotoxic activities. Planta Med. 2016, 82, 1279–1285. [Google Scholar] [CrossRef] [PubMed]
  237. Li, L.W.; Qi, Y.Y.; Liu, S.X.; Wu, X.D.; Zhao, Q.S. Neo-clerodane and abietane diterpenoids with neurotrophic activities from the aerial parts of Salvia leucantha Cav. Fitoterapia 2018, 127, 367–374. [Google Scholar] [CrossRef]
  238. de Oliveira, P.F.; Munari, C.C.; Nicolella, H.D.; Veneziani, R.C.S.; Tavares, D.C. Manool, a Salvia officinalis diterpene, induces selective cytotoxicity in cancer cells. Cytotechnology 2016, 68, 2139–2143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Mofidi Tabatabaei, S.; Salehi, P.; Moridi Farimani, M.; Neuburger, M.; De Mieri, M.; Hamburger, M.; Nejad-Ebrahimi, S. A nor-diterpene from Salvia sahendica leaves. Nat. Prod. Res. 2017, 31, 1758–1765. [Google Scholar] [CrossRef] [PubMed]
  240. Jan, T.; Qadri, R.; Naqvi, B.; Adhikari, A.; Nadeem, S.; Muhammad, A. A novel Salvialactomine from the callus culture of Salvia santolinifolia Boiss. Nat. Prod. Res. 2018, 32, 749–754. [Google Scholar] [CrossRef]
  241. Eghbaliferiz, S.; Emami, S.A.; Tayarani-Najaran, Z.; Iranshahi, M.; Shakeri, A.; Hohmann, J.; Asili, J. Cytotoxic diterpene quinones from Salvia tebesana Bunge. Fitoterapia 2018, 128, 97–101. [Google Scholar] [CrossRef]
  242. Fan, M.; Bao, Y.; Zhang, Z.J.; Zhang, H.B.; Zhao, Q.S. New neo-clerodane diterpenoids with neurotrophic activity from the aerial parts of Salvia tiliifolia. Fitoterapia 2017, 123, 44–50. [Google Scholar] [CrossRef]
  243. Farimani, M.M.; Bahadori, M.B.; Koulaei, S.A.; Salehi, P.; Ebrahimi, S.N.; Khavasi, H.R.; Hamburger, M. New ursane triterpenoids from Salvia urmiensis Bunge: Absolute configuration and anti-proliferative activity. Fitoterapia 2015, 106, 1–6. [Google Scholar] [CrossRef]
  244. Wang, M.; Chen, Y.; Hu, P.; Ji, J.; Li, X.; Chen, J. Neoclerodane diterpenoids from Scutellaria barbata with cytotoxic activities. Nat. Prod. Res. 2020, 34, 1345–1351. [Google Scholar] [CrossRef] [PubMed]
  245. Yang, G.C.; Hu, J.H.; Li, B.L.; Liu, H.; Wang, J.Y.; Sun, L.X. Six New neo -Clerodane Diterpenoids from Aerial Parts of Scutellaria barbata and Their Cytotoxic Activities. Planta Med. 2018, 84, 1292–1299. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, M.; Ma, C.; Chen, Y.; Li, X.; Chen, J. Cytotoxic Neo-Clerodane Diterpenoids from Scutellaria barbata D.Don. Chem. Biodivers. 2019, 16, e1800499. [Google Scholar] [CrossRef]
  247. Hanh, T.T.H.; Anh, D.H.; Quang, T.H.; Trung, N.Q.; Thao, D.T.; Cuong, N.T.; An, N.T.; Cuong, N.X.; Nam, N.H.; Van Kiem, P.; et al. Scutebarbatolides A-C, new neo-clerodane diterpenoids from Scutellaria barbata D. Don with cytotoxic activity. Phytochem. Lett. 2019, 29, 65–69. [Google Scholar] [CrossRef]
  248. Kurimoto, S.I.; Pu, J.X.; Sun, H.D.; Shibata, H.; Takaishi, Y.; Kashiwada, Y. Acylated neo-clerodane type diterpenoids from the aerial parts of Scutellaria coleifolia Levl. (Lamiaceae). J. Nat. Med. 2016, 70, 241–252. [Google Scholar] [CrossRef] [PubMed]
  249. Dai, S.J.; Zhang, L.; Xiao, K.; Han, Q.T. New cytotoxic neo-clerodane diterpenoids from Scutellaria strigillosa. Bioorg. Med. Chem. Lett. 2016, 26, 1750–1753. [Google Scholar] [CrossRef] [PubMed]
  250. Elmasri, W.A.; Hegazy, M.E.F.; Mechref, Y.; Paré, P.W. Cytotoxic saponin poliusaposide from Teucrium polium. RSC Adv. 2015, 5, 27126–27133. [Google Scholar] [CrossRef]
  251. Ben Sghaier, M.; Mousslim, M.; Pagano, A.; Ammari, Y.; Luis, J.; Kovacic, H. β-eudesmol, a sesquiterpene from Teucrium ramosissimum, inhibits superoxide production, proliferation, adhesion and migration of human tumor cell. Environ. Toxicol. Pharmacol. 2016, 46, 227–233. [Google Scholar] [CrossRef] [Green Version]
  252. Gao, C.; Han, L.; Zheng, D.; Jin, H.; Gai, C.; Wang, J.; Zhang, H.; Zhang, L.; Fu, H. Dimeric abietane diterpenoids and sesquiterpenoid lactones from Teucrium viscidum. J. Nat. Prod. 2015, 78, 630–638. [Google Scholar] [CrossRef]
  253. 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]
  254. Kim, G.D.; Park, Y.S.; Jin, Y.H.; Park, C.S. Production and applications of rosmarinic acid and structurally related compounds. Appl. Microbiol. Biotechnol. 2015, 99, 2083–2092. [Google Scholar] [CrossRef] [PubMed]
  255. Li, H.; Zhang, Y.; Chen, H.-H.; Huang, E.; Zhuang, H.; Li, D.; Ni, F. Rosmarinic acid inhibits stem-like breast cancer through hedgehog and Bcl-2/Bax signaling pathways. Pharmacogn. Mag. 2019, 15, 600–606. [Google Scholar] [CrossRef]
  256. Tai, M.C.; Tsang, S.Y.; Chang, L.Y.F.; Xue, H. Therapeutic potential of wogonin: A naturally occurring flavonoid. CNS Drug Rev. 2005, 11, 141–150. [Google Scholar] [CrossRef] [PubMed]
  257. Tan, H.; Li, X.; Yang, W.H.; Yong, K. A flavone, Wogonin from Scutellaria baicalensis inhibits the proliferation of human colorectal cancer cells by inducing of autophagy, apoptosis and G2/M cell cycle arrest via modulating the PI3K/AKT and STAT3 signalling pathways. J. BUON 2019, 24, 1143–1149. [Google Scholar] [PubMed]
  258. Ji-Hyun, G.O.; Wei, J.D.; Park, J.I.; Ahn, K.S.; Kim, J.H. Wogonin suppresses the LPS-enhanced invasiveness of MDA-MB-231 breast cancer cells by inhibiting the 5-LO/BLT2 cascade. Int. J. Mol. Med. 2018, 42, 1899–1908. [Google Scholar] [CrossRef] [Green Version]
  259. Bergers, G.; Brekken, R.; McMahon, G.; Vu, T.H.; Itoh, T.; Tamaki, K.; Tanzawa, K.; Thorpe, P.; Itohara, S.; Werb, Z.; et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2000, 2, 737–744. [Google Scholar] [CrossRef]
  260. Chledzik, S.; Strawa, J.; Matuszek, K.; Nazaruk, J. Pharmacological Effects of Scutellarin, An Active Component of Genus Scutellaria and Erigeron: A Systematic Review. Am. J. Chin. Med. 2018, 46, 319–337. [Google Scholar] [CrossRef]
  261. Guo, F.; Yang, F.; Zhu, Y.H. Scutellarein from Scutellaria barbata induces apoptosis of human colon cancer HCT116 cells through the ROS-mediated mitochondria-dependent pathway. Nat. Prod. Res. 2019, 33, 2372–2375. [Google Scholar] [CrossRef]
  262. Salehi, B.; Fokou, P.V.T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The therapeutic potential of naringenin: A review of clinical trials. Pharmaceuticals 2019, 12, 11. [Google Scholar] [CrossRef] [Green Version]
  263. Liu, D.M.; Cao, Z.X.; Yan, H.L.; Li, W.; Yang, F.; Zhao, W.J.; Diao, Q.C.; Tan, Y. zhu A new abietane diterpenoid from Ajuga ovalifolia var. calantha induces human lung epithelial A549 cell apoptosis by inhibiting SHP2. Fitoterapia 2020, 141, 104484. [Google Scholar] [CrossRef]
  264. Shakeri, A.; Delavari, S.; Ebrahimi, S.N.; Asili, J.; Emami, S.A.; Tayarani-Najaran, Z. A new tricyclic abietane diterpenoid from Salvia chloroleuca and evaluation of cytotoxic and apoptotic activities. Braz. J. Pharmacogn. 2019, 29, 30–35. [Google Scholar] [CrossRef]
  265. Wang, J.N.; Zhang, Z.R.; Che, Y.; Yuan, Z.Y.; Lu, Z.L.; Li, Y.; Li, N.; Wan, J.; Sun, H.D.; Sun, N.; et al. Acetyl-macrocalin B, an ent-kaurane diterpenoid, initiates apoptosis through the ROS-p38-caspase 9-dependent pathway and induces G2/M phase arrest via the Chk1/2-Cdc25C-Cdc2/cyclin B axis in non-small cell lung cancer. Cancer Biol. Ther. 2018, 19, 609–621. [Google Scholar] [CrossRef] [Green Version]
  266. Cortese, K.; Marconi, S.; D’Alesio, C.; Calzia, D.; Panfoli, I.; Tavella, S.; Aiello, C.; Pedrelli, F.; Bisio, A.; Castagnola, P. The novel diterpene 7β-acetoxy-20-hydroxy-19,20-epoxyroyleanone from Salvia corrugata shows complex cytotoxic activities against human breast epithelial cells. Life Sci. 2019, 232, 116610. [Google Scholar] [CrossRef] [PubMed]
  267. Zito, C.I.; Kontaridis, M.I.; Fornaro, M.; Feng, G.S.; Bennett, A.M. SHP-2 Regulates the Phosphatidylinositide 3′-Kinase/Akt Pathway and Suppresses Caspase 3-Mediated Apoptosis. J. Cell. Physiol. 2004, 199, 227–236. [Google Scholar] [CrossRef] [PubMed]
  268. Cagnol, S.; Chambard, J.C. ERK and cell death: Mechanisms of ERK-induced cell death—Apoptosis, autophagy and senescence. FEBS J. 2010, 277, 2–21. [Google Scholar] [CrossRef]
  269. Bartek, J.; Lukas, J. Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell. 2003, 3, 421–429. [Google Scholar] [CrossRef] [Green Version]
  270. Cho, Y.C.; Park, J.E.; Park, B.C.; Kim, J.H.; Jeong, D.G.; Park, S.G.; Cho, S. Cell cycle-dependent Cdc25C phosphatase determines cell survival by regulating apoptosis signal-regulating kinase 1. Cell Death Differ. 2015, 22, 1605–1617. [Google Scholar] [CrossRef] [Green Version]
  271. Uto, T.; Tung, N.H.; Ohta, T.; Juengsanguanpornsuk, W.; Hung, L.Q.; Hai, N.T.; Long, D.D.; Thuong, P.T.; Okubo, S.; Hirata, S.; et al. Antiproliferative activity and apoptosis induction by trijuganone C isolated from the root of Salvia miltiorrhiza Bunge (Danshen). Phyther. Res. 2018, 32, 657–666. [Google Scholar] [CrossRef]
  272. Wang, M.; Zeng, X.; Li, S.; Sun, Z.; Yu, J.; Chen, C.; Shen, X.; Pan, W.; Luo, H. A novel tanshinone analog exerts anti-cancer effects in prostate cancer by inducing cell apoptosis, arresting cell cycle at G2 phase and blocking metastatic ability. Int. J. Mol. Sci. 2019, 20, 4459. [Google Scholar] [CrossRef] [Green Version]
  273. Shen, L.; Lou, Z.; Zhang, G.; Xu, G.; Zhang, G. Diterpenoid Tanshinones, the extract from Danshen (Radix Salviae Miltiorrhizae) induced apoptosis in nine human cancer cell lines. J. Tradit. Chin. Med. 2016, 36, 514–521. [Google Scholar] [CrossRef] [Green Version]
  274. Zaker, A.; Asili, J.; Abrishamchi, P.; Tayarani-Najaran, Z.; Mousavi, S.H. Cytotoxic and apoptotic effects of root extract and tanshinones isolated from Perovskia abrotanoides kar. Iran. J. Basic Med. Sci. 2017, 20, 1377–1384. [Google Scholar] [CrossRef] [PubMed]
  275. Ye, Q.; Yao, G.; Zhang, M.; Guo, G.; Hu, Y.; Jiang, J.; Cheng, L.; Shi, J.; Li, H.; Zhang, Y.; et al. A novel ent-kaurane diterpenoid executes antitumor function in colorectal cancer cells by inhibiting Wnt/β-catenin signaling. Carcinogenesis 2014, 36, 318–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Bahadori, M.B.; Eskandani, M.; De Mieri, M.; Hamburger, M.; Nazemiyeh, H. Anti-proliferative activity-guided isolation of clerodermic acid from Salvia nemorosa L.: Geno/cytotoxicity and hypoxia-mediated mechanism of action. Food Chem. Toxicol. 2018, 120, 155–163. [Google Scholar] [CrossRef] [PubMed]
  277. Masoud, G.N.; Li, W. HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm. Sin. B. 2015, 5, 378–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Lu, X.X.; Ji, X.X.; Bao, J.; Li, Q.Q.; Ji, D.D.; Luo, L. Inhibition of proliferation, migration and invasion of human non-small cell lung cancer cell line a549 by phlomisoside f from phlomis younghusbandii mukerjee. Trop. J. Pharm. Res. 2016, 15, 1413–1421. [Google Scholar] [CrossRef] [Green Version]
  279. Ouyang, X.L.; Qin, F.; Huang, R.Z.; Liang, D.; Wang, C.G.; Wang, H.S.; Liao, Z.X. NF-κB inhibitory and cytotoxic activities of hexacyclic triterpene acid constituents from Glechoma longituba. Phytomedicine 2019, 63, 153037. [Google Scholar] [CrossRef] [PubMed]
  280. Tran, L.T.T.; Ho, D.V.; Le, D.V.; Van Phan, K.; Nguyen, H.T.; Raal, A. Apoptosis-Inducing Effect of Pogostemin A Isolated from the Aerial Parts of Pogostemon auricularius Against the Human Lung Cancer Cells. J. Biol. Act. Prod. from Nat. 2019, 9, 320–327. [Google Scholar] [CrossRef]
  281. Ullah, S.; Khalil, A.A.; Shaukat, F.; Song, Y. Sources, extraction and biomedical properties of polysaccharides. Foods 2019, 8, 304. [Google Scholar] [CrossRef] [Green Version]
  282. Sun, P.; Sun, D.; Wang, X. Effects of Scutellaria barbata polysaccharide on the proliferation, apoptosis and EMT of human colon cancer HT29 Cells. Carbohydr. Polym. 2017, 167, 90–96. [Google Scholar] [CrossRef]
  283. Li, H.; Su, J.; Jiang, J.; Li, Y.; Gan, Z.; Ding, Y.; Li, Y.; Liu, J.; Wang, S.; Ke, Y. Characterization of polysaccharide from Scutellaria barbata and its antagonistic effect on the migration and invasion of HT-29 colorectal cancer cells induced by TGF-β1. Int. J. Biol. Macromol. 2019, 131, 886–895. [Google Scholar] [CrossRef]
  284. Basappa, G.; Kumar, V.; Sarojini, B.K.; Poornima, D.V.; Gajula, H.; Sannabommaji, T.K.; Rajashekar, J. Chemical composition, biological properties of Anisomeles indica Kuntze essential oil. Ind. Crops Prod. 2015, 77, 89–96. [Google Scholar] [CrossRef]
  285. Rigano, D.; Marrelli, M.; Formisano, C.; Menichini, F.; Senatore, F.; Bruno, M.; Conforti, F. Phytochemical profile of three Ballota species essential oils and evaluation of the effects on human cancer cells. Nat. Prod. Res. 2017, 31, 436–444. [Google Scholar] [CrossRef] [PubMed]
  286. Scharf, D.R.; Simionatto, E.L.; Carvalho, J.E.; Salvador, M.J.; Santos, É.P.; Stefanello, M.É.A. Chemical composition and cytotoxic activity of the essential oils of Cantinoa stricta (Benth.) Harley & J.F.B. Pastore (Lamiaceae). Rec. Nat. Prod. 2015, 10, 257–261. [Google Scholar]
  287. Zorzetto, C.; Sánchez-Mateo, C.C.; Rabanal, R.M.; Lupidi, G.; Bramucci, M.; Quassinti, L.; Iannarelli, R.; Papa, F.; Maggi, F. Antioxidant activity and cytotoxicity on tumour cells of the essential oil from Cedronella canariensis var. canariensis (L.) Webb & Berthel. (Lamiaceae). Nat. Prod. Res. 2015, 29, 1641–1649. [Google Scholar] [CrossRef] [PubMed]
  288. de Sousa, M.H.O.; Morgan, J.M.S.; Cesca, K.; Flach, A.; de Moura, N.F. Cytotoxic Activity of Cunila angustifolia Essential Oil. Chem. Biodivers. 2020, 17, e1900656. [Google Scholar] [CrossRef] [PubMed]
  289. Pudziuvelyte, L.; Stankevicius, M.; Maruska, A.; Petrikaite, V.; Ragazinskiene, O.; Draksiene, G.; Bernatoniene, J. Chemical composition and anticancer activity of Elsholtzia ciliata essential oils and extracts prepared by different methods. Ind. Crops Prod. 2017, 107, 90–96. [Google Scholar] [CrossRef]
  290. Donadu, M.G.; Usai, D.; Mazzarello, V.; Molicotti, P.; Cannas, S.; Bellardi, M.G.; Zanetti, S. Change in Caco-2 cells following treatment with various lavender essential oils. Nat. Prod. Res. 2017, 31, 2203–2206. [Google Scholar] [CrossRef]
  291. Damasceno, L.M.O.; Silva, A.L.N.; dos Santos, R.F.; Feitosa, T.A.; Viana, L.G.F.; de Oliveira, R.G.; e Silva, M.G.; Rolim, L.A.; Araújo, C.S.; Araújo, E.C.C.; et al. Cytotoxic activity of chemical constituents and essential oil from the leaves of leonotis nepetifolia (Lamiaceae). Rev. Virtual Quim. 2019, 11, 517–528. [Google Scholar] [CrossRef]
  292. Nikšić, H.; Durić, K.; Sijamić, I.; Korić, E.; Kusturica, J.; Omeragić, E.; Muratovic, S. In vitro antiproliferative activity of Melissa officinalis L. (Lamiaceae) leaves essential oil. Bol. Latinoam. Caribe Plantas Med. Aromat. 2019, 18, 480–491. [Google Scholar] [CrossRef]
  293. Ouakouak, H.; Benchikha, N.; Hassani, A.; Ashour, M.L. Chemical composition and biological activity of Mentha citrata Ehrh., essential oils growing in southern Algeria. J. Food Sci. Technol. 2019, 56, 5346–5353. [Google Scholar] [CrossRef]
  294. Bardaweel, S.K.; Bakchiche, B.; ALSalamat, H.A.; Rezzoug, M.; Gherib, A.; Flamini, G. Chemical composition, antioxidant, antimicrobial and Antiproliferative activities of essential oil of Mentha spicata L. (Lamiaceae) from Algerian Saharan atlas. BMC Complement. Altern. Med. 2018, 18, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Mothana, R.A.; Nasr, F.A.; Khaled, J.M.; AL-Zharani, M.; Noman, O.M.; Abutaha, N.; Al-Rehaily, A.J.; Almarfadi, O.M.; Kumar, A.; Kurkcuoglu, M. Analysis of chemical composition and assessment of cytotoxic, antimicrobial, and antioxidant activities of the essential oil of meriandra dianthera growing in Saudi Arabia. Molecules 2019, 24, 2647. [Google Scholar] [CrossRef] [Green Version]
  296. Kahkeshani, N.; Hadjiakhoondi, A.; Navidpour, L.; Akbarzadeh, T.; Safavi, M.; Karimpour-Razkenari, E.; Khanavi, M. Chemodiversity of Nepeta menthoides Boiss. & Bohse. essential oil from Iran and antimicrobial, acetylcholinesterase inhibitory and cytotoxic properties of 1,8-cineole chemotype. Nat. Prod. Res. 2018, 32, 2745–2748. [Google Scholar] [CrossRef] [PubMed]
  297. Sharifi-Rad, J.; Ayatollahi, S.A.; Varoni, E.M.; Salehi, B.; Kobarfard, F.; Sharifi-Rad, M.; Iriti, M.; Sharifi-Rad, M. Chemical composition and functional properties of essential oils from Nepeta schiraziana Boiss. Farmacia 2017, 65, 802–812. [Google Scholar]
  298. Shakeri, A.; Khakdan, F.; Soheili, V.; Sahebkar, A.; Shaddel, R.; Asili, J. Volatile composition, antimicrobial, cytotoxic and antioxidant evaluation of the essential oil from Nepeta sintenisii Bornm. Ind. Crops Prod. 2016, 84, 224–229. [Google Scholar] [CrossRef]
  299. Fitsiou, E.; Mitropoulou, G.; Spyridopoulou, K.; Tiptiri-Kourpeti, A.; Vamvakias, M.; Bardouki, H.; Panayiotidis, M.I.; Galanis, A.; Kourkoutas, Y.; Chlichlia, K.; et al. Phytochemical profile and evaluation of the biological activities of essential oils derived from the greek aromatic plant species Ocimum basilicum, Mentha spicata, Pimpinella anisum and Fortunella margarita. Molecules 2016, 21, 1069. [Google Scholar] [CrossRef] [Green Version]
  300. Hajlaoui, H.; Mighri, H.; Aouni, M.; Gharsallah, N.; Kadri, A. Chemical composition and in vitro evaluation of antioxidant, antimicrobial, cytotoxicity and anti-acetylcholinesterase properties of Tunisian Origanum majorana L. essential oil. Microb. Pathog. 2016, 95, 86–94. [Google Scholar] [CrossRef]
  301. Marrelli, M.; Conforti, F.; Formisano, C.; Rigano, D.; Arnold, N.A.; Menichini, F.; Senatore, F. Composition, antibacterial, antioxidant and antiproliferative activities of essential oils from three Origanum species growing wild in Lebanon and Greece. Nat. Prod. Res. 2016, 30, 735–739. [Google Scholar] [CrossRef]
  302. Elansary, H.O.; Abdelgaleil, S.A.M.; Mahmoud, E.A.; Yessoufou, K.; Elhindi, K.; El-Hendawy, S. Effective antioxidant, antimicrobial and anticancer activities of essential oils of horticultural aromatic crops in northern Egypt. BMC Complement. Altern. Med. 2018, 18, 214. [Google Scholar] [CrossRef]
  303. Mothana, R.A.; Khaled, J.M.; Noman, O.M.; Kumar, A.; Alajmi, M.F.; Al-Rehaily, A.J.; Kurkcuoglu, M. Phytochemical analysis and evaluation of the cytotoxic, antimicrobial and antioxidant activities of essential oils from three Plectranthus species grown in Saudi Arabia. BMC Complement. Altern. Med. 2018, 18, 237. [Google Scholar] [CrossRef]
  304. Zhang, H.Y.; Gao, Y.; Lai, P.X. Chemical composition, antioxidant, antimicrobial and cytotoxic activities of essential oil from premna microphylla turczaninow. Molecules 2017, 22, 381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Jardak, M.; Elloumi-Mseddi, J.; Aifa, S.; Mnif, S. Chemical composition, anti-biofilm activity and potential cytotoxic effect on cancer cells of Rosmarinus officinalis L. essential oil from Tunisia. Lipids Health Dis. 2017, 16, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Eftekhari, M.; Ardekani, M.R.S.; Amini, M.; Akbarzadeh, T.; Safavi, M.; Razkenari, E.K.; Khanavi, M. Biological activities of the essential oil and total extract of Salvia macrosiphon Boiss. J. Basic Clin. Pharm. 2017, 8, 82–86. [Google Scholar]
  307. Privitera, G.; Luca, T.; Castorina, S.; Passanisi, R.; Ruberto, G.; Napoli, E. Anticancer activity of Salvia officinalis essential oil and its principal constituents against hormone-dependent tumour cells. Asian Pac. J. Trop. Biomed. 2019, 9, 24–28. [Google Scholar] [CrossRef]
  308. Luca, T.; Napoli, E.; Privitera, G.; Musso, N.; Ruberto, G.; Castorina, S. Antiproliferative Effect and Cell Cycle Alterations Induced by Salvia officinalis Essential Oil and Its Three Main Components in Human Colon Cancer Cell Lines. Chem. Biodivers. 2020, 17, e2000309. [Google Scholar] [CrossRef] [PubMed]
  309. Alimpić, A.; Pljevljakušić, D.; Šavikin, K.; Knežević, A.; Ćurčić, M.; Veličković, D.; Stević, T.; Petrović, G.; Matevski, V.; Vukojević, J.; et al. Composition and biological effects of Salvia ringens (Lamiaceae) essential oil and extracts. Ind. Crops Prod. 2015, 76, 702–709. [Google Scholar] [CrossRef]
  310. Sharifi-Rad, J.; Sharifi-Rad, M.; Hoseini-Alfatemi, S.M.; Iriti, M.; Sharifi-Rad, M.; Sharifi-Rad, M. Composition, cytotoxic and antimicrobial activities of Satureja intermedia CA Mey essential oil. Int. J. Mol. Sci. 2015, 16, 17812–17825. [Google Scholar] [CrossRef] [Green Version]
  311. Fitsiou, E.; Anestopoulos, I.; Chlichlia, K.; Galanis, A.; Kourkoutas, I.; Panayiotidis, M.I.; Pappa, A. Antioxidant and antiproliferative properties of the essential oils of Satureja thymbra and Satureja parnassica and their major constituents. Anticancer Res. 2016, 36, 5757–5763. [Google Scholar] [CrossRef] [Green Version]
  312. Venditti, A.; Bianco, A.; Quassinti, L.; Bramucci, M.; Lupidi, G.; Damiano, S.; Papa, F.; Vittori, S.; Maleci Bini, L.; Giuliani, C.; et al. Phytochemical analysis, biological activity, and secretory structures of Stachys annua (L.) L. subsp. annua (Lamiaceae) from central Italy. Chem. Biodivers. 2015, 12, 1172–1183. [Google Scholar] [CrossRef]
  313. 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]
  314. 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]
  315. 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] [PubMed]
  316. Janitermi, M.; Nemati, F. Cytotoxic effect of Zataria multiflora on breast cancer cell line (MCF-7) and normal fibroblast cell. Sci. J. 2015, 36, 1895–1904. [Google Scholar]
  317. Mohammadpour, G.; Tahmasbpour, R.; Noureini, S.K.; Tahmasbpour, E.; Bagherpour, G. In vitro Antimicrobial and Cytotoxicity Assays of Satureja bakhtiarica and Zataria multiflora Essential Oils. Am. J. Phytomed. Clin. Ther. 2015, 3, 502–511. [Google Scholar]
  318. Saeidi, M.; Asili, J.; Emami, S.A.; Moshtaghi, N.; Malekzadeh-Shafaroudi, S. Comparative volatile composition, antioxidant and cytotoxic evaluation of the essential oil of Zhumeria majdae from south of Iran. Iran. J. Basic Med. Sci. 2019, 22, 80–85. [Google Scholar] [CrossRef]
  319. Skorić, M.; Gligorijević, N.; Čavić, M.; Todorović, S.; Janković, R.; Ristić, M.; Mišić, D.; Radulović, S. Cytotoxic activity of Nepeta rtanjensis Diklić & Milojević essential oil and its mode of action. Ind. Crops Prod. 2017, 100, 163–170. [Google Scholar] [CrossRef]
  320. Spyridopoulou, K.; Fitsiou, E.; Bouloukosta, E.; Tiptiri-Kourpeti, A.; Vamvakias, M.; Oreopoulou, A.; Papavassilopoulou, E.; Pappa, A.; Chlichlia, K. Extraction, Chemical Composition, and Anticancer Potential of Origanum onites L. Essential Oil. Molecules 2019, 24, 2612. [Google Scholar] [CrossRef] [Green Version]
  321. Bhagat, M.; Sangral, M.; Arya, K.; Rather, R. Chemical characterization, biological assessment and molecular docking studies of essential oil of Ocimum viride for potential antimicrobial and anticancer activities. bioRxiv. 2018, 390906. [Google Scholar] [CrossRef]
  322. Özkan, A.; Erdoğan, A. Evaluation of cytotoxic, membrane, and DNA damaging effects of Thymus revolutus célak essential oil on different cancer cells. Turk. J. Med. Sci. 2017, 47, 702–714. [Google Scholar] [CrossRef]
  323. Russo, A.; Cardile, V.; Graziano, A.C.E.; Avola, R.; Bruno, M.; Rigano, D. Involvement of Bax and Bcl-2 in induction of apoptosis by essential oils of three Lebanese Salvia species in human prostate cancer cells. Int. J. Mol. Sci. 2018, 19, 292. [Google Scholar] [CrossRef] [Green Version]
  324. Ahani, N.; Sangtarash, M.H.; Alipour Eskanda-Ni, M.; Houshmand, M. Zataria multiflora boiss. Essential oil induce apoptosis in two human colon cancer cell lines (HCT116 & SW48). Iran. J. Public Health 2020, 49, 753–762. [Google Scholar] [PubMed]
  325. Zhao, Y.; Chen, R.; Wang, Y.; Qing, C.; Wang, W.; Yang, Y. In Vitro and in Vivo Efficacy Studies of Lavender angustifolia Essential Oil and Its Active Constituents on the Proliferation of Human Prostate Cancer. Integr. Cancer Ther. 2017, 16, 215–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  326. Lipin Dev, M.S.; Menon, D.B. Essential Oil Extracted from Plectranthus amboinicus Induces Apoptosis in the Lung Cancer Cells via Mitochondrial Pathway. Int. J. Pharm. Sci. Drug Res. 2017, 9, 83–89. [Google Scholar] [CrossRef]
  327. Lu, X.; Yang, L.; Lu, C.; Xu, Z.; Qiu, H.; Wu, J.; Wang, J.; Tong, J.; Zhu, Y.; Shen, J. Molecular Role of EGFR-MAPK Pathway in Patchouli Alcohol-Induced Apoptosis and Cell Cycle Arrest on A549 Cells in Vitro and in Vivo. BioMed Res. Int. 2016, 2016, 4567580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  328. Jaradat, N.; Al-Maharik, N. Fingerprinting, antimicrobial, antioxidant, anticancer, cyclooxygenase and metabolic enzymes inhibitory characteristic evaluations of Stachys viticina Boiss. essential oil. Molecules 2019, 24, 3880. [Google Scholar] [CrossRef] [Green Version]
  329. Athamneh, K.; Alneyadi, A.; Alsamri, H.; Alrashedi, A.; Palakott, A.; El-Tarabily, K.A.; Eid, A.H.; Dhaheri, Y.A.; 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] [Green Version]
  330. Justus, B.; Kanunfre, C.C.; Budel, J.M.; de Faria, M.F.; Raman, V.; de Paula, J.P.; Farago, P.V. New insights into the mechanisms of French lavender essential oil on non-small-cell lung cancer cell growth. Ind. Crops Prod. 2019, 136, 28–36. [Google Scholar] [CrossRef]
Figure 1. Selected genera belonging to the Lamiaceae family.
Figure 1. Selected genera belonging to the Lamiaceae family.
Cancers 12 02957 g001
Figure 2. Selected molecular mechanisms of action of extracts, single derived compounds and essential oils from the Lamiaceae family against lung, breast, prostate, and colon cancer cell lines (created by BioRender). Abbreviations: Akt—protein kinase B, AMP—adenosine monophosphate, AMPK—AMP-activated protein kinase, Apaf—apoptotic protease activating factor, Atg—autophagy-related protein, Bak—Bcl-2 homologous antagonist/killer, Bax—Bcl-2 associated X protein, Bcl-2—B-cell lymphoma 2, Bcl-xL—B-cell lymphoma-extra large, CDK—cyclin-dependent kinase, c-Myc—avian myelocytomatosis viral oncogene homolog, COX—cyclooxygenase, DISC—death-inducing signaling complex, ERK—extracellular signal-regulated kinase, FADD—Fas-associated death domain, Her2—human epidermal growth factor receptor 2, IKK—IkB kinase, IL—interleukin, LC3—autophagosomal membrane-associated protein light chain 3, LPR 5/6—low-density lipoprotein receptor–related protein 5/6, MAP2K—MAPK kinase, MAP3K—MAP2K kinase, MAPK—mitogen-activated protein kinase, PARP—poly(ADP-ribose) polymerase, PI3K—phosphatidylinositol 3-kinase, PI3K-III—class III PI3K complex 1, PTEN—phosphatase and tensin homolog, Ras—rat sarcoma viral oncogene homolog, RHEB—Ras homolog enriched in brain, SIRT1—Sirtuin, TAK—TGF-β-activated kinase, TNF—tumor necrosis factor, TNFR—tumor necrosis factor receptor, TSC 1/2—tuberous sclerosis proteins 1 and 2, ULK—Unc-51 like autophagy activating kinase, Wnt—wingless-related integration site. Arrows: Activation— Cancers 12 02957 i001; Inhibition— Cancers 12 02957 i002.
Figure 2. Selected molecular mechanisms of action of extracts, single derived compounds and essential oils from the Lamiaceae family against lung, breast, prostate, and colon cancer cell lines (created by BioRender). Abbreviations: Akt—protein kinase B, AMP—adenosine monophosphate, AMPK—AMP-activated protein kinase, Apaf—apoptotic protease activating factor, Atg—autophagy-related protein, Bak—Bcl-2 homologous antagonist/killer, Bax—Bcl-2 associated X protein, Bcl-2—B-cell lymphoma 2, Bcl-xL—B-cell lymphoma-extra large, CDK—cyclin-dependent kinase, c-Myc—avian myelocytomatosis viral oncogene homolog, COX—cyclooxygenase, DISC—death-inducing signaling complex, ERK—extracellular signal-regulated kinase, FADD—Fas-associated death domain, Her2—human epidermal growth factor receptor 2, IKK—IkB kinase, IL—interleukin, LC3—autophagosomal membrane-associated protein light chain 3, LPR 5/6—low-density lipoprotein receptor–related protein 5/6, MAP2K—MAPK kinase, MAP3K—MAP2K kinase, MAPK—mitogen-activated protein kinase, PARP—poly(ADP-ribose) polymerase, PI3K—phosphatidylinositol 3-kinase, PI3K-III—class III PI3K complex 1, PTEN—phosphatase and tensin homolog, Ras—rat sarcoma viral oncogene homolog, RHEB—Ras homolog enriched in brain, SIRT1—Sirtuin, TAK—TGF-β-activated kinase, TNF—tumor necrosis factor, TNFR—tumor necrosis factor receptor, TSC 1/2—tuberous sclerosis proteins 1 and 2, ULK—Unc-51 like autophagy activating kinase, Wnt—wingless-related integration site. Arrows: Activation— Cancers 12 02957 i001; Inhibition— Cancers 12 02957 i002.
Cancers 12 02957 g002
Table 1. Cytotoxic properties of selected extracts from the Lamiaceae family against lung, colon, breast, and prostate cancer cells.
Table 1. Cytotoxic properties of selected extracts from the Lamiaceae family against lung, colon, breast, and prostate cancer cells.
Name of The SpeciesPart of the PlantType of ExtractClass of Compounds/Compounds Identified in ExtractCancer Cell LinesRef.
Ajuga chamaepitys subsp. chia (Schreb.) = synonym of Ajuga chia (Schreb)aerial partsethanolic-T-47D[103]
Cyclotrichium niveum (Boiss.) Manden. and
Scheng
aerial partsethyl acetategallic acid, protocatechuic acid, chlorogenic acid, caffeic acid, gentisic acid, p-coumaric acid, ferulic acid, rutin, luteolin-7 glucoside, quercetin, luteolin, apigeninMCF-7[104]
Eremostachys azerbaijanica Rech.f.rhizomesmethanolic, n-hexane,
dichloromethane
fatty acid derivatives and steroids
steroids and derivatives, heterocyclic hydrocarbons, sesquiterpenes, and linear alkanes
A549[105]
Eremostachys azerbaijanica Rech.f.rhizomesdichloromethanesteroids and derivatives, heterocyclic
hydrocarbons, sesquiterpenes,
and linear alkanes
HT-29[105]
Hymenocrater platystegius Rech.f.leaves and flowersaqueousphenolicsMCF-7[106]
Hyptis pectinata L.leavesethanolic-MCF-7[107]
Hyptis pectinate L.leaves, branches, rootethanolicpectinolide J, hyptolide, and pectinolide EMDA-MB-231[108]
Lavandula coronopifolia Poir.aerial partspetroleum ether, ethyl acetate,
chloroform, and ethanol
-MDA-MB-231[109]
Lavender angustifolia Millleavesmethanolicphenolics, flavonoids, glycosidesMCF-7[110]
Melissa officinalis L.leavesethanolicphenolics and flavonoidsMCF-7, PC-3 and A549[111]
Melissa officinalis L.leavesethanolicphenolics: 3-(3,4-dihydroxyphenyl)-lactic acid, caftaric acid, caffeic acid hexoside, fertaric acid, caffeic acid, sulphated rosmarinic acid, yunnaneic acid E, lithospermic acid A isomer, chicoric acid, yunnaneic acid F, salvianolic acid C derivative I, salvianolic acid C derivative II, rosmarinic acid hexoside, sagerinic acid, rosmarinic acid, salvianolic acid A, salvianolic acid C derivative III, lithospermic acid A, salvianolic acid A isomer, salvianolic acid C derivative IVNCI-H460[112]
Nepeta bracteata Benth.flowering shootaqueousphenolicsMCF-7[106]
Ocimum
americanum L.
leavesethyl acetatephenolics, flavonoids, flavanols, tannins, saponinsHCT-116[113]
Ocimum basilicum var. thyrsiflorum (L.) Benth.leavesethanolicphenolics and flavonoids:
cinnamic acid, gallic acid, methylgallate, ellagic acid, methyl
ellagic acid, apigenin, luteolin, vitexin, isovitexin
HCT-116[114]
Origanum dayi Post.aerial partsethanolic-MCF-7 and T47D[103]
Origanum majorana L.aerial partsethyl acetate2-(4-hydroxy phenyl) ethanol, vanillic acid, 4-hydroxybenzoic acid, syringic acid, caffeic acid, vanillin, trans-ferulic acid, luteolin, cinnamic acidMDA-MB-231 and HT-29[115]
Origanum syriacum L.leavesethanolic-LoVo and SW620[116]
Orthosiphon aristatus (Blume) Miq.leavesmethanolicphenolicsMCF-7[117]
Orthosiphon pallidus Royle ex Benth.whole plantsaqueous-MCF-7 and MDA-MB-231[118]
Phlomis viscosa Poiret.leaves, flowers and stemsethanolicdiosmin, isovaleraldehyde, 2,4-hexadienal, 2-hexenal, alpha terpinene, 1-octen-3-ol, himachala-2,4-diene, n-octanal, buorbonene, 1-propanal, 2-methyl, cubebeneMCF-7[119]
Plectranthus amboinicus
(Lour.) Spreng
leaveschloroformditerpene/7-acetoxy-6-hydroxyroyleanoneMCF-7[120]
Plectranthus amboinicus (Lour.) Spreng.leavesethanolic-MCF-7[121]
Pogostemon heyneanus Benth.leaveschloroform-MCF-7 and
MDA-MB-231
[121]
Rosmarinus officinalis L.leavesethanolicflavonoids, diterpenes, triterpenes:
apigenin, hispidulin, cirsiliol, diosmetin, cirsimaritin, rosmanol, epiisorosmanol, epirosmanol, genkwanin, miltipolone, carnosol, rosmadial, anemosapogenin, rosmaridiphenol, augustic acid, benthamic acid, carnosic acid, 12-methoxycarnosic acid, shogaol, micromeric acid, hinokione, betulinic acid, ursolic acid
HT-29 and SW480[122]
Rosmarinus officinalis L.leaves and flowersaqueousphenolicsMCF-7[106]
Rosmarinus officinalis L.aerial partsaqueous-Caco-2[123]
Rosmarinus officinalis L.leavesmethanolicterpenesLoVo[124]
Rosmarinus officinalis L.leavesmethanolicphenolics and flavonoids, alkaloids, tannins, glycosidesMCF-7[110]
Rosmarinus officinalis L.leavesethanolicphenolics: protocatechuic acid, caffeic acid, ellagic acid, ferulic acid, rosemarinic acid, carnosol, carnosic acidMCF-7[125]
Salvia
fruticosa Mill subsp. thomasii
aerial partsmethanolicluteolin, luteolin 7-O-glucoside, rutin, salvigeninMCF-7, MDA-MB-231, RKO and Caco-2[126]
Salvia ballotiflora Benth.aerial partschloroform-A549[127]
Salvia fruticosa Millbarkmethanolic-MCF-7, T-47D and MDA-468[128]
Salvia fruticosa Mill. (SF.)aerial partsaqueousphenolics and flavonoids:
rosmarinic and caffeic acid
Caco-2 and HT-29[129]
Salvia hispanica L.seedsethanolic-A549[130]
Salvia hispanica L.seedsethanolictannins, saponins, flavonoids, alkaloids, proteins, phenolsPC-3[131]
Salvia officinalis L.leavesethanoliceucalyptol (1,8-cineole), α-thujone, β-thujone, camphor, β-caryophyllene, α-caryophyllene (α-humulene), viridiflorol, manoolA-549, HT-29[132]
Salvia officinalis L.leavesaqueousphenolics: caffeic acid, syringic acid, rutin, coumaric acid, vanillin, quercetin, cinnamic acidMDA-MB-231[133]
Salvia pilifera Montbret and Aucher ex Benth.whole plantmethanolicfumaric acid, gallic acid, gallocatechin, catechin, oleorufein, 4-hydroxybenzoic acid, caffeic acid, syringic acid, ellagic acid, 3-hydroxy cinnamic acid and protocatechuic acidDU-145[134]
Salvia pilifera Montbret et Aucher ex Benth.mericarpsethanolic-A549[135]
Salvia verbenaca L.leavesethyl acetateflavonoids
and terpenes
MDA MB-231[136]
Satureja horvatii subsp. macrophylla (Halácsy) Baden.aerial partsmethanolicmonoterpene hydrocarbons, oxygenated monoterpens, sesquiterpene hydrocarbons, oxygenated sesquiterpenes, monoterpenes, sesquiterpenes: α-thujene, α-pinene, camphene, 1-octen-3-ol, α-myrcene, 3-octanol, α-phellandrene + δ 3-carene, α-terpinene, p-cymene, limonene, 1,8-cineole, cis-ocimene, γ-terpinene, trans sabinene hydrate, terpinolene, linalool, trans-pinocarveol, cis-verbenol, camphor, borneol, terpinene-4-ol, p-cymen-8-ol, α-terpineol, dihydrocarvone, thymol methyl ether, thymoquinone, thymol, carvacrol, caryophyllene, aromadendrene, α-humulene, alloaromadendrene, γ-muurolene, viridiflorene, γ-elemene, b-bisabolene, γ-cadinene, δ-cadinene, (-)-spathulenol, caryophyllene oxide, viridiflorolA549[137]
Sideritis ozturkii Aytaç and Aksoyflower and leafmethanolicgallic acid, protocatechuic acid, catechin, 4-hydroxybenzoic acid, caffeic acid, syringic acid, rutin trihydrate, trans-p-coumaric acid, trans-ferulic acid, myricetin, trans-resveratrol, quercetin, trans-cinnamic acid, naringenin, kaempferolDLD-11[138]
Sideritis syriaca L.leavesmethanolicphenolic acids: gallic acid, p-hydroxybenzoic acid, cafeic acid, chlorogenic acid, p-coumaric acid, ferulic acid, o-coumaric acid, rosmarinic acid and trans- cinnamic acidMCF-7[139]
Teucrium fruticans L.leavesethanol/ethyl acetate/water-A549[140]
Thyme vulgaris L.leavesmethanolicphenolics and flavonoids, tanninsMCF-7[110]
Thymus daenensis Celak.leaves and stemsethanolic-MCF-7[141]
Thymus mastichina L.whole plantethanol/ethyl acetate/water-A549[140]
Vitex trifolia L.leavesmethanol-MCF-7[142]
Table 2. Cytotoxic properties of isolated pure compounds from the Lamiaceae family against lung, colon, breast, and prostate cancer cells.
Table 2. Cytotoxic properties of isolated pure compounds from the Lamiaceae family against lung, colon, breast, and prostate cancer cells.
Name of The SpeciesPart of the PlantActive Compounds/
Class of Compounds
Cancer Cell LinesRef.
Caryopteris incana (Thunb.) Miq.,whole plantabietane diterpenes: caryopincaolide A, C, DA549, Calu-1[211]
Clerodendranthus spicatus (Thunb.) C. Y. Wuaerial partsursane-type triterpenoids: spicatusoids A–E and three known ones and a known oleanane-type triterpenoidA-549, MCF-7, SW480[212]
Clerodendrum indicum (L.) Kuntze and Clerodendrum villosum Blumeroots3b-hydroxy-D:B-friedo-olean-5-ene, oleanolic acid 3-acetate, taraxerol, lupeol, betulinic acid, (22E)-stigmasta-4,22,25-trien-3-one, stigmasta-4,25-dien-3-one, stigmasta-4,22-dien-3-one, 22-dehydroclerosterol, clerosterol, stigmasterol, b-sitosterol, pectolinarigenin, hispidulinSW620, ChaGo-K-1, BT474[213]
Clerodendrum inerme (L.) Gaertnleavesharwickiic acid, Crolerodendrum B-abietane diterpenes, uncinatone, acacetin, kaempferol 3,7,4′-trimethyl ether, 14,15-dihydro-15β-methoxy-3-epi-caryoptin, 5α,8α-epidioxyergosta-6,22-diene-3β-olHCT-116[214]
Clerodendrum kiangsiense Merrill ex H. L. Liherbabeo-abietane diterpenoid: 12-methoxy-6,11,14,16-tetrahydroxy-17(15Ñ16)-abeo5,8,11,13-abietatetraen-3,7-dione, cryptojaponol, fortunin EA549, MCF-7[215]
Clerodendrum yaundense
Gurke
twigslupane-type triterpene: (16a)-lupa-12,20(29)-dien-16-ol (clerodendrumol) and, O-Acetylclerodendrumol (16a)-Lupa-12,20(29)-dien-16-yl Ace-tateMDA-MB-231[216]
Isodon excisoides (Sun ex C. H. Hu) H. Hara, J. Japaerial parts1α,7α,14β-trihydroxy-20-acetoxy-ent-kaur-15-one, henryin, reniformin C; kamebacetal AHCT-116, NCI-H1650[217]
Isodon rubescens (Hemsl.) H. Haraaerial partsent-7,20-epoxy-kaur-16-en-1α,6β,7β,15β-tetrahydroxyl-11-O-β-d-glucopyranoside, ent-7,20-epoxy-kaur-16-en-6β,7β,14β,15β-tetrahydroxyl-1-O-β-d-glucopyranoside, ent-7,20-epoxy-kaur-16-en-6β,7β,15β-trihydroxyl-1-O-β-d-glucopyranoside, andent-7,20-epoxy-kaur-16-en-7β,11β,14α,15β-tetrahydr-oxyl-6-O-β-d-glucopyranoside, sodonterpene II, enmenol-1-β-glucoside, andenmenolHCT-116[218]
Isodon phyllostachys (Diels) Kudoaerial partsenmein-type diterpenoids enmein-type diterpenoids: 20-episerrin C, serrin C, isodocarpin, serrin BA549, MCF-7, SW480[219]
Isodon rubescens (Hemsl.) H.Haraleavesditerpenoids: 6-epi-11-O-acetylangustifolin and 11-O-acetylangustifolinA549[220]
Isodon wikstroemioides (Hand.–Mazz.) H. Haraaerial partsditerpenoids: 11, 20-epoxy-ent- kauranoids, isowikstroemins H–M, along with two known analogues, macrocalyxin BA549, MCF-7[221]
Leonurus japonicus Houtt.fruitsleonuronins A and BA549[222]
Ocimum basilicum var.thyrsiflorum (L.) BenthleavesC-glycosylated derivative of apigenin: 3′′-O-acetylvitexinHCT-116[114]
Ocimum sanctum L.aerial partslignans: (-)-rabdosiinMCF-7, SKBR3, HCT-116[201]
Orthosiphon stamineus Benth.aerial parts7,4′-dimethylkaempferol, 5,7,4′-trimethylkaempferol, 7,3′,4′-trimethylquercetin, 5,7,3′,4′-tetramethylquercetin, 5,7,4′-trimethylquercetin, 3,5,3′,4′-tetramethylquercetin, 5,7,3′,6′-tetramethoxyflavone, 5,7,3′,4′-tetramethoxyflavone, 2S-5,6,7,3′,4′-pentamethoxyflavanone, 2S-5′-hydroxy-5,7,3′,4′-tetramethoxyflavanoneMCF-7, MDA-MB-231[223]
Phlomis bruguieri Desfaerial parts4’-methoxy-luteolin-7-phosphateMCF-7[224]
Phlomoides umbrosa (Turcz.) Kamelin & Makhmroots28-noroleanane-derived spirocyclic triterpenoids: phlomisu D, phlomisu E, (2α,3α,17R,18β)-19(18→17)-abeo-28-norolean-12-ene-2,3,18,23,24-pentol, phlomispentaolMCF-7[225]
Plectranthus cylindraceus Hoechst. Ex. Benthaerial partssesquiterpene: plectranol A, maaliol, penduletin and chrysosplenol DMBD-MB-321[226]
Plectranthus ecklonii Benth.whole plantabietane diterpenoid: parvifloron DMDA-MB-231[227]
Plectranthus madagascariensis Benthwhole plantroyleanone diterpenes: 7α-formyloxy-6β-hydroxyroyleanon, 7α,6β-dihydroxyroyleanon, 7α-acetoxy-6β-hydroxyroy-leanoneMDA-MB-231, MCF-7, HCT-116, NCI-H460[228]
Plectranthus scutellarioides (L.) R. Br.leavesditerpenoids: spiroscutelones A–CMCF-7[229]
Pogostemon auricularius L. Hassk.aerial partsmeroterpenoids with pyrone-sesquiterpenoid hybrid skeletons: pogostemins A-CSW480, SK-LU-1[230]
Pogostemon cablin (Blanco) Benthaerial partsphenylethanoid glycosides: verbascoside, pedicularioside G ()A549, HCT-15[231]
Premna odorata Blancobarktetrahydrofurofuran lignin: 4β-hydroxyasarininHT-29, MCF-7[232]
Rabdosia serra (Maxim.) Haragrassesditerpenoids: rabdosins E–K, serrin B, serrin A, isodocarpin and lushanrubescensin JA549, NCI--H661[233]
Salvia ballotiflora Benthleavesditerpenoids: 7α-acetoxy-6,7-dihydroicetexone, anastomosineSK-LU-1[234]
Salvia ballotiflora Benth.aerial partsditerpenes: 19-deoxyicetexone,7,20-dihydroanastomosine, icetexoneand19-deoxyisoicetexone,A549, MCF-7[127]
Salvia lachnocalyx Hedgeshoots(2Z,6Z,10Z,14E)-geranylfarnesol and spathulenolMCF-7, HT-29[235]
Salvia leriifolia Benth.aerial partslabdane diterpenoids: 6β,13β-dihydroxylabd-8, 14-diene-19-oic acid, 13-hydroxylabd-8, 14-diene-6β,19-olide, 8,12E,14-labdatrien-6,19-olidMDA-MB-231, DU 145[236]
Salvia leucantha Cavaerial partsneoclerodane diterpenoids: leucansalvialins FeI (1–4), and one rare 18(4→3)-abeo-abietane diterpenoid, leucansalvialin JA549, MCF-7, SW480[237]
Salvia officinalis L.leavesditerpene: manoolMCF-7[238]
Salvia sahendica Boiss. & Buhsleavesterpenoid: sclareolMDA-MB-231[239]
Salvia santolinifolia Boisscallussalvialactomine, 5-MethylflavonePC-3[240]
Salvia tebesana Bungerootsditerpenoids: tebesinone A (1) and tebesinone B (2), aegyptinone A (3) andaegyptinone B (4)MCF-7, PC-[241]
Salvia tiliifolia L.aerial partsneo-clerodane diterpenoids: tiliifolins A–EA549, MCF-7, SW480[242]
Salvia urmiensis Bungeaerial partstriterpenoids: urmiensolide B and urmiensic acidMCF-7[243]
Scutellaria barbata D. Donwhole plantneoclerodane diterpenoids: barbatin F, barbatin G, scutebata A, scutebata B, scutebata C and scutebata PLoVo, MCF-7, HCT-116[244]
Scutellaria barbata D. Donaerial partsneo-clerodane diterpenoids: 13(R*)-1β,6α-dibenzoyloxy-7β-hydroxy-8β,13-epoxy-3-neo-cleroden-15,16-olide (scutebata C1), 2-oxo-6α-nicotinoyloxy-7β-benzoyloxy-8β-hy-droxy-3,11(E),13-neo-clerodatrien-15,16-olide.(scutebata X), 13(R*)-2-oxo-6α-acetoxy-7β-nicotinoyloxy-8β,13-epoxy-3-neo-cleroden-15,16-olide, (scutebata A1)MCF-7, A549[245]
Scutellaria barbata D. Donaerial partsneoclerodane diterpenoid: barbatin H, scutebata P, scutebarbatine F, 6-O-nicotinoylscutebarbatine G, scutebata G, scutebata E, scutebata D, barbatin C, scutebarbatine A, scutebartine G, scutebarbatine B, 6,7-Di-O-acetoxybarbatin A, scutebata C, scutebata A, scutebarbatine X, scutebata BLoVo, MCF-7, HCT-116[246]
Scutellaria barbata D.Donwhole plantneo-clerodane diterpenoids: scutebarbatolides A-C, 4-deoxy-11,12-didehydroandrographolide, scutehenanine H, 14β-hydroxyscutolideKLNCaP, MCF-7[247]
Scutellaria coleifolia Levl.aerial partsneo-clerodane type diterpenoids: scutefolides G1-SA549, MCF-7[248]
Scutellaria strigillosa Hemsley, J. Linn. Socwhole plantneo-clerodane diterpenoids: scutestrigillosins A-CMCF-7, HT-29[249]
Teucrium polium L.aerial partssaponin glycosides: poliusaposide A, poliusaposide B, poliusaposide CMDA-MB-468, HCC-2998, COLO 205[250]
Teucrium ramosissimum Desf.leavessesquiterpene: β-eudesmolA549, HT-29, Caco-2[251]
Teucrium viscidum Blum.whole plantsabietane diterpenoid: teuvisone, a pair of new dimeric abietane diterpenoid stereoisomers: biteuvisones A and B, and three sesquiterpenoid lactones, teuvislactones A−Cteuvisone and biteuvisones B showed cytotoxic effect against NCI-H460, HCT-116[252]
Thymus alternans Kloko.aerial partstriterpenes: 3a-hydroxy-urs-12,15-diene, a-amyrin, b-amyrin, isoursenol, epitaraxerol, and oleanolic acidMDA-MB-231, HCT-15 HCT-116, U1810[253]
Table 3. Cytotoxic properties of selected essential oils from the Lamiaceae family against lung, colon, breast, and prostate cancer cells.
Table 3. Cytotoxic properties of selected essential oils from the Lamiaceae family against lung, colon, breast, and prostate cancer cells.
Name of The SpeciesPart of the PlantCompounds Identified in Essential OilsCancer Cell LinesRef.
Anisomeles indica Kuntze.leavesThe major compounds: farnesyl acetone (10.67%), nootkatone (8.35%), phytol acetate (7.35%), jasmatone (7.81%)A549[284]
Ballota undulata, Ballota saxatilis Ballota nigra ssp. foetidaaerial partsBallota nigra ssp. foetida: germacrene D (23.1%), (E)-β-caryophyllene (20.3%) and caryophyllene oxide (6.2%),
Ballota saxatilis: linalool (11.2%), (E)-β-caryophyllene (8.8%), caryophyllene oxide (6.3%) and (E)-2-hexenal (5.6%),
Ballota undulata: germacrene D (16.0%), and bicyclogermacrene (10.4%)
MCF-7[285]
Cantinoa stricta (Benth.) Harley and J.F.B. Pastore (formely Hyptis stricta Benth.)leaves and flowersThe major compounds: caryophyllene oxide (leaf –31.6%; flower –21.7%) and cis-pinane (leaf –15.4%; flower –9.7%), α-pinene (9.4%) and β-pinene (9.1%)MCF-7, NCI-H460, PC-3[286]
Cedronella canariensis
var. canariensis (L.) Webb and Berthel.
aerial partsThe major compounds: pinocarvone (58.0%) and β-pinene (10.8%)MDA-MB-231, HCT-116[287]
Cunila angustifolia Benth.leavesThe major compounds: pulegone (29.5%), isomenthol (27.0%), menthone (8.6%), neomenthol (7.2%), menthyl acetate (2.5%), and caryophyllene oxide (2.0%)A-549, MCF-7[288]
Elsholtzia ciliata (Thunb.) Hylanderaerial partsThe major compounds: dehydroelsholtzia ketone, elsholtzia ketone, sesquiterpenes β-bourbonene, caryophyllene, α-caryophyllene, germacrene D, and α-farneseneMDA-MB-231[289]
Lavandula hybrid Rev., Lavandula latifolia Medikus., Lavandula vera D.C. and Lavandula angustifolia Miller.aerial partsTerpenes: linalool and linalyl acetate terpenoids: 1,8-cineoleCaco-2[290]
Leonotis nepetifolia (L.) R.Br.leavesThe major compounds: 3-octanone (3.75%), (E)-ocimene (15.85%), (Z)-ocimene (7.01%), linalool, caryophillene oxide, and 1-octen-3-ol (42.58%)HCT-116[291]
Melissa officinalis L.leavescitral (47.2%), caryophyllene oxide (10.2%), citronellal (5.4%), geraniol (6.6%), geranyl acetate (4.1%) and β- caryophyllene (3,8%)MCF-7, NCI-H460[292]
Mentha citrate Ehrh.aerial partThe major compounds: linalool (34.69%), linalyl acetate (35.75%)HCT-116[293]
Mentha spicata L.aerial partsThe major compounds: carvone (49.5%), limonene (16.1%), 1,8-cin-eole (8.7%), cis-dihydrocarvone (3.9%), β-caryophyllene (2.7%), germacrene D (2.1%), and β-pinene (1.1%)HCT-116, MCF-7[294]
Meriandra dianthera (Konig ex Roxb.) Benth.aerial partscamphor (54.3%), 1,8-cineole (12.2%), and camphene (10.4%)MCF-7, LoVo[295]
Nepeta menthoides Boiss. and Bohse.-The major compounds: 1,8-cineole (70.06%)MCF-7, T-47D, MDA-MB-231[296]
Nepeta schiraziana Boiss.aerial partsThe major compounds: 1,8-cineole (33.67%), germacrene D (11.45%), β-caryophyllene (9.88%), and caryophyllene oxide (7.34%)MCF-7[297]
Nepeta sintenisii Bornm.aerial parts4aα,7α7aβ nepetalactone (51.74%), β-farnesene (12.26%), 4aα,7α,7aα nepetalactone (8.01%), germacrene-D (5.01%), and 4aα7β,7aα-nepetalactone (3.71%)LS180, MCF-7[298]
Ocimum basilicum L., Mentha spicata L.chopped leaves and stemsMentha spicata: carvone (85.4%), limonene (8.4%), and β-Pinene (1.4%)
Ocimum basilicum: methyl chavicol (74.9%), linalol (18.4%)
MCF-7, Caco2[299]
Origanum majorana L.aerial partsThe major compounds: terpinen-4-ol (23.2%), Cis-sabinene hydrate (17.5%), γ-terpinene (10.5%), p-cymene (9%), α-terpineol (5.6%), α-terpinene (4.7%), and trans-sabinene hydrate (4.0%)HT-29[300]
Origanum dictamnus (L.) Kostel., Origanum libanoticum Boiss., and Origanum microphyllum (Bentham) T. Vogel-The major compounds: carvacrol, p-cymene, linalool, γ-terpinene, and terpinen-4-ol asLoVo[301]
Origanum vulgare L.leavespulegone (77.4%), menthone (4.8%), cis-Isopulegone
(2.2%), piperitenone (2.1%), limonene (1.0%), and
myrcene (0.6%)
MCF-7, HT-29[302]
Plectranthus. cylindraceus Hocst. ex Benth., Plectranthus. asirensis JRI Wood. and Plectranthus barbatus Andrews.leaves and branchesThe major compounds: α-pinene (46.2%), maaliol (42.8%), and β-caryophyllene (13.3%)HT-29[303]
Premna microphylla Turcz.aerial partsThe major compounds: blumenol C (49.7%), β-cedrene (6.1%), limonene (3.8%), α-guaiene (3.3%), cryptone (3.1%), and gurkeα-cyperone (2.7%)MCF-7[304]
Rosmarinus officinalis L.aerial partsThe major compounds: 1,8-cineole (23.56%), camphene (12.78%), camphor (12.55%), and β-pinene (12.3%)MCF-7[305]
Salvia macrosiphon Boiss.aerial partsThe major compounds: linalool (19%), β-cedrene (14.64%), and β-elemene (13.33%)MCF-7, MDA-MB-231, T-47D[306]
Salvia officinalis L.aerial partsThe major compounds: α-thujone, 1,8-cineole, and camphorLNCaP, MCF-7[307]
Salvia officinalis L.aerial partsα-thujone (29.39%), 1,8-cineole (eucalyptol 22.8%), and camphor (13.05%)Caco-2, HT-29, HCT-116[308]
Salvia ringens Sibth. and Sm.whole plantThe major compounds: 1.8-cineole (31.99%), cam-phene (17.06%), borneol (11.94%), and α-pinene (11.52%)HCT-116[309]
Satureja intermedia C.A.Meyaerial partsγ-terpinene (37.1%), thymol (30.2%), p-cymene (16.2%), limonene (3.9%), α-terpinene (3.3), myrcene (2.5%), germacrene B (1.4%), elemicine (1.1%), and carvacrol (0.5%)MCF-7[310]
Satureja thymbra L. and Satureja parnassica Heldr. and Sart. ex Boissaerial partsThe major compounds: carvacrol, thymol, γ-terpinene, and p-cymeneMCF-7, A549[311]
Stachys annua L. subsp. annuaaerial partsThe major compounds: phytol (9.8%), germacrene D (9.2%), and spathulenol (8.5%)HCT-116, MDA-MB-231[312]
Stachys annua L. subsp. annuaaerial partsphytol (9.8%), germacrene D (9.2%), and spathulenol (8.5%)MDA-MB 231, HCT-116[312]
Stachys parviflora L.aerial partsThe major compounds: α-terpenylacetate (23.6%), caryophyllene (16.8%), bicyclogermacrene (9.3%), spathulenol (4.9%), and α-pinene (4.2%)HCT-116[313]
Tetradenia riparia (Hochst.) Codd.leavesfenchone (6.1%), dronabinol (11.0%), aromadendrene oxide (14.7%), and (E,E)–farnesol (15.0%)HT-29, MCF-7[314]
Thymus alternans Klokov.aerial partsThe major compounds: (E)-nerolidol, neryl acetate, nerolMDA-MB-231, HCT-15, HCT-116, U1810[253]
Thymus munbyanus subsp. Coloratus (Boiss. and Reut.) Greuter and Burdetstems, leavesborneol (44.8 and 31.2%)
Other components occurring in noteworthy levels were camphor (5.7 and 13.6%), camphene (3.6 and 7.5%), 1,8-cineole (6.0 and 4.2%), and germacrene D (5.0 and 3.1%)
MDA-MB-231[315]
Zataria multiflora Boiss.aerial parts-MCF-7[316]
Zataria multiflora Boiss.
Satureja bakhtiarica Bunge.
leavesThe major compounds: phenol (56.35%, 37.4%), thymol (13.82%, 22.6%), P-cymene (8.79%, 19.3%), γ-terpinene (3.36%, 5.0%), β-myrcene (1.91%, 0.8%), β-caryophyllene (1.28%, 2.2%), α-terpinene (1.21%, 0.9%), caryophyllene oxide (0.47%, 2.0%), and carvacrole (2.88%, 0.2%)MDA-MB-231[317]
Zhumeria majdae Rech.f. and Wendelbo.aerial partsThe major compounds: linalool (24.4–34.6%), camphor (26.1–34.7%), and translinalool oxide (7.6–28.6%)MCF-7[318]

Share and Cite

MDPI and ACS Style

Sitarek, P.; Merecz-Sadowska, A.; Śliwiński, T.; Zajdel, R.; Kowalczyk, T. An In Vitro Evaluation of the Molecular Mechanisms of Action of Medical Plants from the Lamiaceae Family as Effective Sources of Active Compounds against Human Cancer Cell Lines. Cancers 2020, 12, 2957. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12102957

AMA Style

Sitarek P, Merecz-Sadowska A, Śliwiński T, Zajdel R, Kowalczyk T. An In Vitro Evaluation of the Molecular Mechanisms of Action of Medical Plants from the Lamiaceae Family as Effective Sources of Active Compounds against Human Cancer Cell Lines. Cancers. 2020; 12(10):2957. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12102957

Chicago/Turabian Style

Sitarek, Przemysław, Anna Merecz-Sadowska, Tomasz Śliwiński, Radosław Zajdel, and Tomasz Kowalczyk. 2020. "An In Vitro Evaluation of the Molecular Mechanisms of Action of Medical Plants from the Lamiaceae Family as Effective Sources of Active Compounds against Human Cancer Cell Lines" Cancers 12, no. 10: 2957. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12102957

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