Next Article in Journal
Research on Preparation Methods of Carbon Nanomaterials Based on Self-Assembly of Carbon Quantum Dots
Next Article in Special Issue
Cannabidiolic acid in Hemp Seed Oil Table Spoon and Beyond
Previous Article in Journal
Inhibitory Effect and Mechanism of Chitosan–Ag Complex Hydrogel on Fungal Disease in Grape
Previous Article in Special Issue
Hemp Seeds of the Polish ‘Bialobrzeskie’ and ‘Henola’ Varieties (Cannabis sativa L. var. sativa) as Prospective Plant Sources for Food Production
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Medicinal Natural Products of Cannabis sativa Linn.: A Review

by
Anwuli Endurance Odieka
1,
Gloria Ukalina Obuzor
2,
Opeoluwa Oyehan Oyedeji
3,
Mavuto Gondwe
4,
Yiseyon Sunday Hosu
5 and
Adebola Omowunmi Oyedeji
1,*
1
Department of Chemical and Physical Sciences, Walter Sisulu University, Mthatha 5099, South Africa
2
Department of Pure and Industrial Chemistry, University of Port Harcourt, Port Harcourt 500004, Rivers State, Nigeria
3
Department of Chemistry, Fort Hare University, Alice 5700, South Africa
4
Department of Human Biology, Walter Sisulu University, Mthatha 5099, South Africa
5
Department of Economics and Business Sciences, Walter Sisulu University, Mthatha 5099, South Africa
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(5), 1689; https://doi.org/10.3390/molecules27051689
Submission received: 31 December 2021 / Revised: 23 February 2022 / Accepted: 24 February 2022 / Published: 4 March 2022
(This article belongs to the Special Issue Industrial Hemp Chemistry and Nutraceutical Perspectives)

Abstract

:
Cannabis sativa is known among many cultures for its medicinal potential. Its complexity contributes to the historical application of various parts of the plant in ethno-medicines and pharmacotherapy. C. sativa has been used for the treatment of rheumatism, epilepsy, asthma, skin burns, pain, the management of sexually transmitted diseases, difficulties during child labor, postpartum hemorrhage, and gastrointestinal activity. However, the use of C. sativa is still limited, and it is illegal in most countries. Thus, this review aims to highlight the biological potential of the plant parts, as well as the techniques for the extraction, isolation, and characterization of C. sativa compounds. The plant produces a unique class of terpenophenolic compounds, called cannabinoids, as well as non-cannabinoid compounds. The exhaustive profiling of bioactive compounds and the chemical characterization and analysis of C. sativa compounds, which modern research has not yet fully achieved, is needed for the consistency, standardization, and the justified application of Cannabis sativa products for therapeutic purposes. Studies on the clinical relevance and applications of cannabinoids and non-cannabinoid phenols in the prevention and treatment of life-threatening diseases is indeed significant. Furthermore, psychoactive cannabinoids, when chemically standardized and administered under medical supervision, can be the legal answer to the use of C. sativa.

1. Introduction

The applications of plants as medicines predates human history. A medicinal plant refers to any plant which contains substances of therapeutic potential in one or more of its parts for the synthesis of plant-based drugs [1]. Active medicinal plant ingredients are referred to as bioactive phytochemicals. [2]. These bioactive compounds are believed to increase the ability of plants to survive or adapt to their surroundings [3] and are used as medicines, flavorings, and recreational drugs in humans. One notable medicinal plant that has continued to garner attention over the years, and in recent times, is Cannabis sativa.
Cannabis sativa L. is known for its medicinal uses since ancient times, because of its rich supply of phytochemicals [4], hence the quest for harnessing its pharmacological potential by scientists. The term “Cannabis” is used to define the products (drugs and essential oils) that are prepared or obtained from the annual herb C. sativa and its variants, which are of the family Cannabaceae [5]. The utilization of this multipurpose plant has been restrained for a long time because of the psychoactive effects of a specific cannabinoid (Δ9-tetrahydrocannabinol; C12H30O2) [6]. It was strongly prohibited in the twentieth century, and was removed from the British pharmacopeia. The plant was demonized due to its high abuse liability and supposedly insufficient health benefits [7]. Furthermore, due to the inability to prepare standardized preparations, and the diffusion of the recreational use of cannabis below therapeutic concentrations from the end of the 19th to the first half of the 20th century, the medical use of cannabis began to decline [7]. In 1937, the “Marihuana Tax Act”, a federal legislation in the United States, functionally ended all medical uses of cannabis and was removed from the “National Formulary and Pharmacopoeia” in 1941 [7]. In 1961, cannabis resin, extracts, and tinctures were listed in the Schedule I of the single Convention on Narcotic Drugs, which prohibits the use, possession, production, manufacture, export, import, and trade of cannabis, except for medical and scientific purposes [7].
However, in multiple countries today, its cultivation and usage are regulated by laws [6,8]. Recent decriminalization policies and new scientific evidence have increased the interest in the medicinal potential of cannabis and have paved the way for the release of marketing authorizations for cannabis-based products [7]. In 1985, the United States Food and Drugs Administration (US FDA) reconsidered the medical use of cannabinoids, and approved Marinol (dronabinol) and Cesamet (nabilone), two synthetic analogues of tetrahydrocannabinol (THC), for the management of nausea and vomiting associated with cancer chemotherapy [7]. The Office of Medical Cannabis Research (OMC), a Dutch government agency in Europe, became the first organization to obtain the exclusive right to supply medical cannabis to research institutes and pharmacies, and, under the Single Convention on Narcotic Drugs of 1961, to import and export cannabis extracts and resin for medical purposes. Several medical cannabis products, all of which are dried female flowering tops, except Bediol (which is ground into small pieces for its easy manipulation by patients with spasticity), are exported by the OMC, with the proper licenses, to other member states of the European Union [7]. In Italy, the Military Pharmaceutical Chemical Works of Florence became the official national settlement for cultivating and manufacturing medical cannabis with a standard cannabinoid content [7,9]. The Italian Ministry of Health in November 2015, in a Ministerial Degree, authorized the indoor cultivation of cannabis flowering tops at a fixed temperature and at fixed light-dark cycles, leading to a standardized composition of different cannabinoids [9]. Two Italian products (FM1 and FM2) are currently available for consumers, and their use was approved for the treatment of chronic pain, neurological disorders, and other diseases resistant to standard therapies [7,9,10]. Non-psychoactive compounds found in C. sativa are associated with fewer side effects and can be used for several industrial applications [6]. The hemp stem supplies both cellulosic and woody fibers. The woody fibers are used for animal beddings, while the cellulosic fibers (bast fibers) are used as a substitute for fiberglass, and to produce bioplastics [4]. Its use as an anti-bacterial finishing agent and in functionalized textiles have also been reported [4]. The inflorescence was used, traditionally, for acute pain, insomnia, coughing, and wounds. The leaves were used for malaria, panting, roundworm, scorpion stings, hair loss, and the greying of hair. The stem bark was used for physical injury and strangury. Vaginal discharge, difficult births, strangury, the retention of the placenta, and physical injuries were treated using the roots [11]. In addition, Cannabis sativa contains essential oils of a high value, which can also improve the effectiveness of cannabinoids in pharmaceutical formulations [6].
Despite the influx of chemical-based medicines for treatments, the relevance of medicinal plants in drug development cannot be overemphasized. In recent years, commercial medicinal cannabis products with several variations in the phytocannabinoid content have been licensed and produced in Canada [7,8] and in several other countries. Several synthetic and standardized products are currently available on the market; however, patients’ preferences lean towards herbal preparations, because they are easy to handle and self-administer [7]. Thus, this review intends to highlight the phytochemicals present in the different plant parts, which potentiates their pharmacological activities, as well as the techniques for the extraction, isolation, and characterization of C. sativa compounds.

2. Methods

Literature on the published works of Cannabis sativa was obtained using electronic search engines, such as Google Scholar, the WSU online database (PubChem), and Science Direct. The keywords included, namely, Cannabis sativa, medicinal plants, Cannabis phytochemicals, ethnopharmacology bioactivity, and medicinal potentials, were used to source for data. An extensive review of the literature from 2011 through to 2021 (the last ten years) on Cannabis sativa L. was used to summarize its medicinal potential. Conversely, an emphasis will be placed on the isolation and characterization techniques from 1970 to 2021 to have a broadened view of the advancements in analytical techniques over the past years. Overall, twenty-nine (29) papers relating to the areas of our focus were chosen and were reviewed by all authors. The results from the search were carefully sorted, based on a general understanding, the review questions, and the related objectives.

3. Origin and Botanical Description of C. sativa

The genus name Cannabis means “cane-like” while sativa means “sown”, which signifies that the plant is propagated from the seed and not from the roots [12]. It is believed to have originated in Asia and occurs widely in Africa [12,13]. Central and south-east Asia are the potential natural origins for the domestication of the Cannabis genus [14] and it is known by different common names in different languages (hemp, marihuana, kannabis sativa, ganja, bhang, and al-bhango) [15]. In South Africa, it is colloquially known, in Afrikaans, as “dagga”; in IsiXhoxa as “umfincafincane”; and in Isizulu as “umunyane” [16,17]. Taxonomically, Carl Linnaeus, a Swedish botanist, was the first to coin the name Cannabis sativa [18]. Other botanists stated that different types of Cannabis existed based on their size, shape, and resin content (breeding and selection). This review discusses, in particular, C. sativa.
The Cannabis phenotype (its observable traits or characteristics, such as its leaf shape and flower color) is based on two main factors: its genetic code (genotype) and the external environmental factors [19].
The roots are branched and are about 30–60 cm deep (Farag and Kayser, 2017) [12]. Cannabis inflorescence is made up of several flower heads found on long leafy stems from each leaf axil. A single brownish fruit, about 2-5mm long, is produced per flower, and it contains a single seed tightly covered with a hard shell [12]. The fruit is propagated by bird and the seed germinates after 8–12 days [18]. The leaves, bracts, and stems of the plant are rich in trichrome, which are a diverse set of structures containing the secondary metabolites (phytocannabinoids and terpenoids) responsible for the defense, plant interactions, and typical smell [18]. Figure 1, below, shows the plant parts of C. sativa.

4. Phytochemistry of C. sativa

4.1. Chemical Profile of C. sativa

Cannabis, as a herbal medicine, is a complex mixture of compounds, including cannabinoid phenols, non-cannabinoid phenols (stilbenoids, lignans, spiro-indans, and dihydrophenanthrenes), flavonoids, terpenoids, alcohols, aldehydes, n-alkanes, wax esters, steroids, and alkaloids [6,8,11]. Over 500 chemical compounds have been isolated from the cannabis plant and have been reported [13]. The several classes of secondary metabolites are present in different parts of the plant with a wide range of applications (nutraceuticals, cosmetics, aromatherapy, and pharmacotherapy) that are beneficial for humans. However, previous studies have focused mainly on the cannabinoids, Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) in particular; hence, the female flower top is only harvested, while other parts of the plant are discarded [11].
Cannabinoids are a class of terpenophenolic compounds obtained by the alkylation of an alkyl-resorcinol with a monoterpene unit [20,21]. They feature alkyl resorcinol and monoterpene moieties in their molecules [20,22]. This specific chemical class in Cannabis is present in the glandular trichomes, which are abundant in the female flower as phytocannabinoid acids, and in the vegetable matrix as neutral phytocannabinoids [6,13]. They are biosynthesized by the alkylation of olivetolic acid with geranyl-pyrophosphate by a prenyltransferase to produce cannabigerolic acid (CBGA). Decarboxylation, a chemical reaction, converts the acidic forms (Δ9 THCA, CBDA, CBCA, and CBGA) into their neutral forms, which are more active and efficient in terms of pharmacological activity [8,23]. To date, 125 cannabinoids have been identified and reported, in addition to five new cannabinoids reported in the past two years, 42 non-cannabinoid phenolics, 34 flavonoids, 120 terpenoids, 3 sterols, and 2 alkaloids [8,11,13]. Terpenoids are the second largest class of cannabis compounds and are responsible for their characteristic aroma [13]. Table 1 below summarizes the classes of compounds isolated from Cannabis sativa and the different plant parts in which they are present.
Figure 2, Figure 3 and Figure 4 below show the structures of the different classes of bioactive compounds isolated from Cannabis sativa [13,29,41].

4.2. Extraction, Isolation, and Chemical Characterization of C. sativa

Many methods have been reported for the extraction of Cannabis in the literature. These include direct maceration (DM), soxhlex extraction, ultrasound-assisted extraction (UAE), supercritical fluid extraction, and microwave-assisted extraction (MAE) [41]. However, two methods of extracting Cannabis are differentiated in the literature [41]. The first is the maceration of the plant material in an organic solvent (direct maceration) and the subsequent removal of the solvent by the concentration of the extract under reduced pressure [41]. The second is the innovative supercritical fluid extraction (SFE) method, which involves the use of pressurized solvents [41]. It is necessary for cannabinoid compounds to be extracted with organic solvents instead of water, because the active compounds are less soluble in polar solvents [41]. The most commonly used solvents are ethanol, ether, chloroform, and methanol [42]. When used for extraction, various compounds, including some undesired substances, dissolve together with the cannabinoids [42]. The high solvent power of ethanol for cannabinoid compounds is the reason why it is frequently used in home-made extracts of Cannabis [41]. However, non-desired compounds (chlorophyll, lipids, and waxy materials) are also extracted which, therefore, requires further steps to remove the co-extracted impurities for a high-purity medicinal product to be obtained [41]. A patent on the method for the isolation of herbal and cannabinoid medicinal extracts stated that the solubility of non-therapeutic substances (chlorophyll and waxy materials) is reduced when the solvent is selected from a group that includes acetonitrile, benzene, dichloromethane, diethyl ether, acetone, butanol, ethanol, chloroform, ethyl acetate, hexane, pentane, propanol, tetrahydrofuran, toluene, xylene, and various combinations of these solvents [41]. The International Conference on Harmonization (ICH) recommends the use of less toxic solvents in the manufacture of drug substances and dosage forms, and sets pharmaceutical limits for residual solvents in drug products [43]. Residual solvents pose risks to human health and are classified into three classes. Class 1 solvents (including carbon tetrachloride, benzene, and methyl chloroform) are regarded as human carcinogens and are environmentally hazardous [41]. Class 2 solvents include methanol and hexane, which are generally said to be limited, and they are possible causative agents of irreversible toxicity, such as neurotoxicity or teratogenicity [41]. Class 3 solvents (ethanol and ethyl acetate) are generally regarded as having a low toxic potential to humans [41]). Above all, ethanol is generally recognized as a safe (GRAS) solvent [41]. In a study by Brighenti et al., they compared the following four extraction techniques to obtain a high yield of medicinal cannabinoids: ultrasound-assisted extraction (UAE), microwave-assisted extraction (MAE), supercritical fluid extraction (SFE), and direct maceration (DM). They concluded that DM, with ethanol as the extraction solvent at room temperature for an overall time of 45 min, is the best extraction technique (in terms of a high yield) for non-psychoactive cannabinoids from hemp [44].
Over the last decade, compounds in Cannabis have been identified, isolated, and determined by various chromatographic techniques with different spectroscopic detection methods. C. sativa samples are analyzed for both legal and medicinal purposes [41]. Nevertheless, the knowledge of their exact composition remains very significant. In 2009, recommended methods for the identification and analysis of cannabis products were released by the United Nations Office on Drugs and Crime [45]. One notable technique that has been employed in identifying the diverse composition of the compounds found is high-performance liquid chromatography (HPLC) [41]. Spectroscopic approaches or methods are based on the variable absorbance or redirection of electromagnetic (EM) radiation by chemical bonds, resulting in the radiation or transition of the sample’s atoms to a higher energy state [46]. Some advantages are attributed to these spectroscopic methods, such as permitting spatial measurements of metabolites and offering a global metabolic fingerprint of a sample with rapid spectral acquisition [46]. Some of these approaches/methods include Fourier transform infrared spectroscopy (FTIR), nuclear magnetic resonance (NMR) spectroscopy, mass spectrometry (MS), HPLC, gas chromatography–mass spectrometry (GC–MS), and liquid chromatography–mass spectrometry (LS–MS) [41,46]. Taking into account the recommended methods and the mandatory requirement of the Ministerial decree to use only chromatographic techniques coupled with mass spectrometric detection, cannabinoid concentrations and its stability in cannabis tea and cannabis oil, prepared from standardized flowering tops obtained from the Military Pharmaceutical Chemical Works of Florence, were studied by Pacifici et al. using easy and fast ultra-high performance liquid chromatography–tandem mass spectrometry (UHPLC–MS/MS) [9,10]. Table 2 is a summary of the reported extraction solvents, as well as the identification, isolation, and characterization methods of Cannabis.

4.3. Biological Evaluation/Potentials of C. sativa

From the biological point of view, the psychoactive cannabinoids reported include Δ9 THC, cannabinol (CBN), and cannabinodiol (CBND), while cannabidiol (CBD) and other cannabinoids are non-psychoactive [8,11]. THC is the major psychoactive component and the toxicity of this metabolite of Cannabis is the most studied [11,28]. Its psychoactive component decreases in the order of inflorescence (the flower), leaves, stem, roots, and seeds, respectively [8]. The interest in the potential medical use of cannabis and cannabinoids rose significantly in the 1990s, following the discovery of the endocannabinoid (eCB) system in mammals [7]. The physiological effects of cannabinoids are exerted through various receptors, such as the cannabinoid receptors (CB1 and CB2), adrenergic receptors, and the recently discovered GPCRs (GPR55, GPR3 and GPR5) [8]. Historically, each part of the Cannabis plant is indicated mostly for pain killing, inflammation, and for mental illnesses. For example, the Cannabis root has been recommended for treating fever, inflammation, gout, arthritis, and joint pain, as well as skin burns, hard tumors, postpartum hemorrhage, difficult child labor, sexually transmitted diseases, gastrointestinal activity, and infections [40]. Cannabis has also been used to treat asthma, epilepsy, fatigue, glaucoma, insomnia, nausea, pain, and rheumatism, as well as being used as appetite stimulant and a digestive aid [7,11,13]. Since concentrations above 0.05% are pharmacologically interesting, Cannabis inflorescence and leaf material may contain sufficient cannabinoids, mono- and sesquiterpenoids, and flavonoids for therapeutic applications [11]. Cannabis terpenoids and flavonoids, mainly myrcene, limonene, pinene, β-caryophyllene, and cannflavin A, act in synergy with cannabinoids to induce pharmacological effects [7]. It was proven that these compounds, which are synthetized in the aerial parts of the plant, enhance CBD’s anti-inflammatory effects and antagonize THC dysphoric action [96]. Cannabidiol (CBD) and Cannabidavarin (CBDV) (neutral cannabinoids) have been reported to have the therapeutic potential for the treatment of epilepsy (focal seizures), as well as treating nausea and vomiting [97,98]. Conversely, THC and CBN have been found to be active in lowering intraocular pressure, and can be applied in all cases of glaucoma that are resistant to other therapies [9]. Cannflavin A and B are also notable flavonoids (prenylflavonoids) with medicinal potentials, such as their anti-inflammatory, anti-neoplastic, antioxidant, neuro-protective, anti-parasitic, and anti-viral effects [99]. Table 3, below, shows a summary of the reported bioactivities (biological potentials) of the bioactive compounds present in Cannabis sativa.
Cannabis female flowering tops can be simply administered through commercially available vaporizers (e.g., Micro Vape, G Pen Herbal Vaporizer, and Volcano), buccal sprays (e.g., Sativex), oral capsules (e.g., Cannador), decoctions, or oils [7]. Only cannabis use through oral or inhalatory administration is allowed. Smoking reduces the bioavailability of cannabis ingredients by 40%, and its complete combustion can cause lung diseases and airway obstructions [7]. Homemade decoctions and pharmacy oils are currently the most widespread cannabis formulations in Europe, making the standardization of preparation difficult [7]. Cannabis pharmacological action is dose-dependent and can induce many adverse effects (AEs), principally related to THC, due to unintentional overdosing [7]. The typical symptoms of cannabis acute intoxication that have been reported are dizziness, confusion, tachycardia, postural hypotension, dysphoria, panic depression, hallucinations, allergic reactions, vomiting, and diarrhea [7,137,138]. Furthermore, withdrawal symptoms, such as irritability, aggression, anxiety, insomnia, decreased appetite, tremors, sweating, and headaches may appear after the abrupt cessation of the long-term administration of high doses of cannabis [7]. According to the ICH efficacy and safety guidelines, it is recommended to start with low doses and increase quantities after a satisfactory period of clinic evaluation, depending on the pharmacological effects and the possible adverse effects [139].
In the current COVID-19 pandemic, scientists are repurposing medicines (identifying new therapeutic use(s) of existing drugs) known for their biological potential (anti-viral or anti-inflammatory properties) to tackle the global issue and similar future viruses [140]. They have hypothesized that CBD could be used as an anti-viral agent [141] or anti-inflammatory [142,143] tool, or to inhibit pulmonary fibrosis in COVID-19 patients [144]. In addition, the known growing evidence of the anxiolytic effects of CBD have also been hypothesized to be used as a therapeutic option to treat long-lasting COVID-19-related anxiety and PTSD [145], which is likely to be a significant issue of the pandemic.

5. Conclusions

With the recent evaluation, acceptance, and legalization of Cannabis products for therapeutic purposes, researchers, particularly in the field of natural products, are challenged to improve and standardize the extraction and characterization of the bioactive compounds from Cannabis sativa. Despite various reports of its economic and therapeutic values, it is legal in a handful of jurisdictions (Uruguay, Canada, some US states, and parts of Africa). Presently, Cannabis remains illegal in several countries. This review summarized the biological potential and the techniques for the extraction, isolation, and characterization of Cannabis sativa compounds, and it describes the effectiveness of the various parts of the herb in pharmacotherapy. The usage of C. sativa roots and stem barks in present-day medical research, and the development of new Cannabis-based medicines or products, in contrast to the flowering part only, is highly recommended because they can be exploited for medicine and other uses. In addition, Cannabis-based pharmaceutical products must undergo long purification processes to eliminate unwanted components such as chlorophyll and residual organic solvents. The use of standardized reagents is also very crucial in the analytical studies of C. sativa. Furthermore, future research should seek to clarify the factors responsible for the complexity of C. sativa extracts in terms of their chemical compositions, the physical properties of their active ingredients, and their liability to photochemical oxidation.

Author Contributions

Conceptualization, A.E.O., A.O.O., G.U.O., O.O.O., M.G. and Y.S.H.; methodology, A.E.O., G.U.O., O.O.O., M.G., Y.S.H. and A.O.O.; validation, A.E.O., M.G., Y.S.H. and A.O.O.; data curation, A.E.O.; writing—original draft preparation, AEO; writing—review and editing, A.E.O., A.O.O., O.O.O., G.U.O., M.G. and Y.S.H.; supervision, A.O.O., G.U.O., O.O.O., M.G. and Y.S.H.; project administration, A.O.O.; funding acquisition, A.E.O. and A.O.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Research Foundation (NRF), grant numbers 130205 and 137963.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the Directorate of Research and Innovation, Walter Sisulu University (WSU), South Africa and Govan Mbeki Research and Development Center, University of Fort Hare, South Africa.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sofowora, A.; Ogunbodede, E.; Onayade, A. The role and place of medicinal plants in the strategies for disease prevention. Afr. J. Tradit. Complement. Altern. Med. 2013, 10, 210–229. [Google Scholar] [CrossRef]
  2. Choudhary, N.; Siddiqui, M.; Bi, S.; Khatoon, S. Variation in preliminary phytochemicals screening of Cannabis sativa L. leaf, stem and root. Int. J. Pharmacogn. 2014, 1, 516–519. [Google Scholar]
  3. Bandar, H.; Hijazi, A.; Rammal, H.; Hachem, A.; Saad, Z.; Badran, B. Techniques for the extraction of bioactive compounds from Lebanese Urtica Dioica. Am. J. Phytomed. Clin. Ther. 2013, 1, 507–513. [Google Scholar]
  4. Andre, C.M.; Hausman, J.F.; Guerriero, G. Cannabis sativa: The plant of the thousand and one molecules. Front. Plant Sci. 2016, 7, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Elsohly, M.A.; Radwan, M.M.; Gul, W.; Chandra, S.; Galal, A. Phytochemistry of Cannabis sativa L. Phytocannabinoids 2017, 103, 1–36. [Google Scholar]
  6. Baldino, L.; Scognamiglio, M.; Reverchon, E. Supercritical fluid technologies applied to the extraction of compounds of industrial interest from Cannabis sativa L. and to their pharmaceutical formulations: A review. J. Pharm. Fluids 2020, 165, 104960. [Google Scholar] [CrossRef]
  7. Brunetti, P.; Pichini, S.; Pacifici, R.; Busardò, F.P.; del Rio, A. Herbal preparations of medical cannabis: A vademecum for prescribing doctors. Medicina 2020, 56, 237. [Google Scholar] [CrossRef]
  8. Lewis, M.M.; Yang, Y.; Wasilewski, E.; Clarke, H.A.; Kotra, L.P. Chemical profiling of medical Cannabis extracts. ACS Omega 2017, 2, 6091–6103. [Google Scholar] [CrossRef] [PubMed]
  9. Pacifici, R.; Marchei, E.; Salvatore, F.; Guandalini, L.; Busardò, F.P.; Pichini, S. Evaluation of cannabinoids concentration and stability in standardized preparations of cannabis tea and cannabis oil by ultra-high performance liquid chromatography tandem mass spectrometry. Clin. Chem. Lab. Med. 2017, 55, 1555–1563. [Google Scholar] [CrossRef]
  10. Pacifici, R.; Marchei, E.; Salvatore, F.; Guandalini, L.; Busardò, F.P.; Pichini, S. Evaluation of long-term stability of cannabinoids in standardized preparations of cannabis flowering tops and cannabis oil by ultra-high-performance liquid chromatography tandem mass spectrometry. Clin. Chem. Lab. Med. 2018, 56, 94–96. [Google Scholar] [CrossRef] [PubMed]
  11. **, D.; Dai, K.; **. Pharm. Ther. 2015, 97, 559–561. [Google Scholar] [CrossRef] [PubMed]
  12. Erridge, S.; Mangal, N.; Salazar, O.; Pacchetti, B.; Sodergren, M.H. Cannflavins—From plant to patient: A sco** review. Fitoterapia 2020, 146, 104712. [Google Scholar] [CrossRef] [PubMed]
  13. Pacher, P.; Mechoulam, R. Is lipid signaling through cannabinoid 2 receptors part of a protective system? Prog. Lipid Res. 2011, 50, 193–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Mcguire, P.; Robson, P.; Cubala, W.J.; Vasile, D.; Morrison, P.D.; Barron, R.; Taylor, A.; Wright, S.J. Cannabidiol (CBD) as an adjunctive therapy in schizophrenia: A multicenter randomized controlled trial. Am. J. Psychiatry 2018, 175, 225–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Parker, L.A.; Rock, E.M.; Limebeer, C.L. Regulation of nausea and vomiting by cannabinoids. Br. J. Pharmacol. 2011, 163, 1411–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Yeshurun, M.; Shpilberg, O.; Herscovici, C.; Shargian, L.; Dreyer, J.; Peck, A.; Israeli, M.; Levy-Assaraf, M.; Gruenewald, T.; Mechoulam, R.; et al. Cannabidiol for the prevention of graft-versus-host-disease after allogeneic hematopoietic cell transplantation: Results of a phase II study. Biol. Blood Marrow Transpl. 2015, 21, 1770–1775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Brierley, D.I.; Samuels, J.; Duncan, M.; Whalley, B.J.; Williams, C.M. Cannabigerol is a novel, well-tolerated appetite stimulant in pre-satiated rats. Psychopharmacology 2016, 233, 3603–3613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Rock, E.; Kopstick, R.L.; Limebeer, C.L.; Parker, L.A. Tetrahydrocannabinolic acid reduces nausea-induced conditioned ga** in rats and vomiting in S uncus murinus. Br. J. Pharmacol. 2013, 170, 641–648. [Google Scholar] [CrossRef] [Green Version]
  19. Rock, E.M.; Sticht, M.A.; Parker, L.A. Effect of Phytocannabinoids on Nausea and Vomiting; Oxford University Press: Oxford, UK, 2014. [Google Scholar]
  20. Tsien, R.; Whalley, B.J.; Devinsky, O. Cannabinoids and Epilepsy. Neurotherapeutics 2015, 12, 747–768. [Google Scholar]
  21. Moreno-Sanz, G. Can you pass the acid test? critical review and novel therapeutic perspectives of Δ9-tetrahydrocannabinolic acid A. Cannabis Cannabinoid Res. 2016, 1, 124–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Rock, E.; Parker, L.A. Effect of low doses of cannabidiolic acid and ondansetron on licl-induced conditioned ga** (a model of nausea-induced behaviour) in rats. Br. J. Pharmacol. 2013, 169, 685–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Rock, E.M.; Connolly, C.; Limebeer, C.L.; Parker, L.A. Effect of combined oral doses of Δ 9-tetrahydrocannabinol (THC) and cannabidiolic acid (CBDA) on acute and anticipatory nausea in rat models. Psychopharmacology 2016, 233, 3353–3360. [Google Scholar] [CrossRef] [PubMed]
  24. Russo, E.B. Taming THC: Potential cannabis synergy and phytocannabinoid-terpenoid entourage effects. Br. J. Pharmacol. 2011, 163, 1344–1364. [Google Scholar] [CrossRef] [PubMed]
  25. Cascone, P.; Iodice, L.; Maffei, M.E.; Bossi, S.; Arimura, G.I.; Guerrieri, E. Tobacco overexpressing β-ocimene induces direct and indirect responses against aphids in receiver tomato plants. J. Plant Physiol. 2015, 173, 28–32. [Google Scholar] [CrossRef] [PubMed]
  26. Bomfim, L.M.; Menezes, L.R.; Rodrigues, A.C.; Dias, R.B.; Gurgel Rocha, C.A.; Soares, M.B.; Neto, A.F.; Nascimento, M.P.; Campos, A.F.; Silva, L.C.; et al. Antitumour activity of the microencapsulation of Annona vepretorum essential oil. Basic Clin. Pharmacol. 2016, 118, 208–213. [Google Scholar] [CrossRef] [PubMed]
  27. De Oliveira Ramalho, T.R.; de Oliveira, M.T.; de Araujo Lima, A.L.; Bezerra-Santos, C.R.; Piuvezam, M.R. Gamma-terpinene modulates acute inflammatory response in mice. Planta Med. 2015, 81, 1248–1254. [Google Scholar]
  28. 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] [PubMed] [Green Version]
  29. Kasuya, H.; Okada, N.; Kubohara, M.; Satou, T.; Masuo, Y.; Koike, K. Expression of BDNF and TH mRNA in the Brain Following Inhaled Administration of A-Pinene. Phytother. Res. 2015, 29, 43–47. [Google Scholar] [CrossRef] [PubMed]
  30. Han, H.D.; Cho, Y.J.; Cho, S.K.; Byeon, Y.; Jeon, H.N.; Kim, H.S.; Kim, B.G.; Bae, D.S.; Lopez-Berestein, G.; Sood, A.K.; et al. Linalool-incorporated nanoparticles as a novel anticancer agent for epithelial ovarian carcinoma. Mol. Cancer Ther. 2016, 15, 618–627. [Google Scholar] [CrossRef] [Green Version]
  31. Russo, E.B. Cannabis and epilepsy: An ancient treatment returns to the fore. Epilepsy Behav. 2017, 70, 292–297. [Google Scholar] [CrossRef] [PubMed]
  32. Sulak, D.; Saneto, R.; Goldstein, B. The current status of artisanal Cannabis for the treatment of epilepsy in the United States. Epilepsy Behav. 2017, 70, 328–333. [Google Scholar] [CrossRef]
  33. Piccinelli, A.C.; Santos, J.A.; Konkiewitz, E.C.; Oesterreich, S.A.; Formagio, A.S.; Croda, J.; Ziff, E.B.; Kassuya, C. Antihyperalgesic and antidepressive actions of (R)-(+)-Limonene, A-phellandrene, and essential oil from schinus terebinthifolius fruits in a neuropathic pain model. Nutr. Neurosci. 2015, 18, 24. [Google Scholar] [CrossRef]
  34. Siqueira, H.D.; Neto, B.S.; Sousa, D.P.; Gomes, B.S.; da Silva, F.V.; Cunha, F.V.; Wanderley, C.W.; Pinheiro, G.; Cândido, A.G.; Wong, D.V.; et al. A-Phellandrene, a cyclic monoterpene, attenuates inflammatory response through neutrophil migration inhibition and mast cell degranulation. Life Sci. 2016, 160, 27–33. [Google Scholar] [CrossRef]
  35. Lima, D.F.; Brandão, M.S.; Moura, J.B.; Leitão, J.M.; Carvalho, F.A.; Miúra, L.M.; Leite, J.R.; Sousa, D.P.; Almeida, F.R. Antinociceptive activity of the monoterpene α-phellandrene in rodents: Possible mechanisms of action. J. Pharm. Pharmacol. 2012, 64, 283–292. [Google Scholar] [CrossRef] [PubMed]
  36. Aydin, E.; Türkez, H.; Taşdemir, Ş. Anticancer and antioxidant properties of terpinolene in rat brain cells. Arch. Ind. Hyg. Toxicol. 2013, 64, 415–424. [Google Scholar] [CrossRef] [PubMed]
  37. Turkez, H.; Aydın, E.; Geyikoglu, F.; Cetin, D. Genotoxic and oxidative damage potentials in human lymphocytes after exposure to terpinolene in vitro. Cytotechnology 2015, 67, 409–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Paula-Freire, L.I.; Andersen, M.; Gama, V.; Molska, G.; Carlini, E.L. The oral administration of trans-caryophyllene attenuates acute and chronic pain in mice. Phytomedicine 2014, 21, 356–362. [Google Scholar] [CrossRef] [PubMed]
  39. Varga, Z.V.; Matyas, C.; Erdelyi, K.; Cinar, R.; Nieri, D.; Chicca, A.; Nemeth, B.T.; Paloczi, J.; Lajtos, T.; Corey, L.; et al. Research Paper Themed Issue Β-Caryophyllene protects against alcoholic steatohepatitis by attenuating inflammation and metabolic dysregulation in mice. Br. J. Pharmacol. 2017, 175, 320–334. [Google Scholar] [CrossRef] [PubMed]
  40. Katsuyama, S.; Mizoguchi, H.; Kuwahata, H.; Komatsu, T.; Nagaoka, K.; Nakamura, H.; Bagetta, G.; Sakurada, T.; Sakurada, S. Involvement of peripheral cannabinoid and opioid receptors in β-caryophyllene-induced antinociception. Eur. J. Pain 2013, 17, 664–675. [Google Scholar] [CrossRef] [PubMed]
  41. Xu, H.B.; Zheng, L.P.; Li, L.; Xu, L.Z.; Fu, J. Elemene, one ingredient of a chinese herb, against malignant tumors: A literature-based meta-analysis. Cancer Invest. 2013, 31, 156–166. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, Q.; Wu, J.; Luo, Y.; Huang, N.; Zhen, N.; Zhou, Y.; Sun, F.; Li, Z.; Pan, Q.; Li, Y. (−)-Guaiol Regulates RAD51 stability via autophagy to induce cell apoptosis in non-small cell lung cancer. Oncotarget 2016, 7, 62585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chinsembu, K.C. Tuberculosis and nature’s pharmacy of putative anti-tuberculosis agents. Acta Tropica 2016, 153, 46–56. [Google Scholar] [CrossRef] [PubMed]
  44. Semenya, S.; Potgieter, M.; Tshisikhawe, M.; Shava, S.; Maroyi, A. Medicinal utilization of exotic plants by bapedi traditional healers to treat human ailments in Limpopo province, South Africa. J. Ethnopharmacol. 2012, 144, 646–655. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, H.H.; Son, J.K.; Jung, B.; Zheng, M.; Kim, J.R. Epifriedelanol from the root bark of Ulmus davidiana inhibits cellular senescence in human primary cells. Planta Medica 2011, 77, 441–449. [Google Scholar] [CrossRef] [Green Version]
  46. Zhou, X.; Wang, F.; Zhou, R.; Song, X.; **e, M. Apigenin: A current review on its beneficial biological activities. J. Food Biochem. 2017, 41, e12376. [Google Scholar] [CrossRef]
  47. He, M.; Min, J.W.; Kong, W.L.; He, X.H.; Li, J.X.; Peng, B.W. A review on the pharmacological effects of vitexin and isovitexin. Fitoterapia 2016, 115, 74–85. [Google Scholar] [CrossRef] [PubMed]
  48. Sharma, A.; Kashyap, D.; Sak, K.; Tuli, H.S.; Sharma, A.K. Therapeutic charm of quercetin and its derivatives: A review of research and patents. Pharm. Pat. Anal. 2018, 7, 15–32. [Google Scholar] [CrossRef] [PubMed]
  49. Ashaari, Z.; Hassanzadeh, G.; Alizamir, T.; Yousefi, B.; Keshavarzi, Z.; Mokhtari, T. The flavone luteolin improves central nervous system disorders by different mechanisms: A review. J. Mol. Neurosci. 2018, 65, 491–506. [Google Scholar] [CrossRef]
  50. Rudroff, T.; Sosnoff, J.J. Cannabidiol to improve mobility in people with multiple sclerosis. Front. Neurol. 2018, 9, 183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Wong, K.U.; Baum, C.R. Acute Cannabis Toxicity. Pediatr. Emerg. Care 2019, 35, 799–806. [Google Scholar] [CrossRef] [PubMed]
  52. International Conference on Harmonisation (ICH). All Guidelines. Available online: https://www.ich.org/page/ich-guidelines (accessed on 19 April 2020).
  53. Schlag, A.K.; O’sullivan, S.E.; Zafar, R.R.; Nutt, D.J. Current controversies in medical cannabis: Recent developments in human clinical applications and potential therapeutics. Neuropharmacology 2021, 191, 108586. [Google Scholar] [CrossRef]
  54. Hill, K.P. Cannabinoids and the coronavirus. Cannabis Cannabinoid Res. 2020, 5, 118–120. [Google Scholar] [CrossRef]
  55. Costiniuk, C.T.; Jenabian, M.A. Acute inflammation and pathogenesis of SARS-CoV-2 infection: Cannabidiol as a potential anti-inflammatory treatment? Cytokine Growth Factor Rev. 2020, 53, 63. [Google Scholar] [CrossRef] [PubMed]
  56. Byrareddy, S.N.; Mohan, M. SARS-CoV2 induced respiratory distress: Can cannabinoids be added to anti-viral therapies to reduce lung inflammation? Brain Behav. Immun. 2020, 87, 120. [Google Scholar] [CrossRef] [PubMed]
  57. Esposito, G.; Pesce, M.; Seguella, L.; Sanseverino, W.; Lu, J.; Corpetti, C.; Sarnelli, G. The potential of cannabidiol in the COVID-19 pandemic. Br. J. Pharmacol. 2020, 177, 4967–4970. [Google Scholar] [CrossRef] [PubMed]
  58. O’sullivan, S.E.; Stevenson, C.W.; Laviolette, S.R. Could cannabidiol be a treatment for coronavirus disease-19-related anxiety disorders? Cannabis Cannabinoid Res. 2021, 6, 7–18. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cannabis plant parts. (a) Male and female Cannabis flowering parts with fresh leaf and seed. (b) Fresh Cannabis root. (c) Fresh Cannabis inflorescence (flower). (d) Fresh Cannabis stem bark.
Figure 1. Cannabis plant parts. (a) Male and female Cannabis flowering parts with fresh leaf and seed. (b) Fresh Cannabis root. (c) Fresh Cannabis inflorescence (flower). (d) Fresh Cannabis stem bark.
Molecules 27 01689 g001
Figure 2. Chemical structures of major cannabinoids; Δ8 THC, tetrahydrocannabinol (a); Δ9-THC, tetrahydrocannabinol (b); THCA, tetrahydrocannabinolic acid (c); CBDA, cannabidiolic acid (d); CBD, cannabidiol (e); CBDV, cannabidivarin (f); THCV, tetrahydrocannabivarin (g); CBGA, cannabigerolic acid (h); CBG, cannabigerol (i); CBN, cannabinol (j); CBNA, cannabinolic acid (k); CBC, cannabichromene (l); CBCA, cannabichromenic acid (m); CBL, cannabicyclol (n); CBLA, cannabicyclolic acid (o). All structures drawn by Odieka, using ChemDraw Ultra 8.0.
Figure 2. Chemical structures of major cannabinoids; Δ8 THC, tetrahydrocannabinol (a); Δ9-THC, tetrahydrocannabinol (b); THCA, tetrahydrocannabinolic acid (c); CBDA, cannabidiolic acid (d); CBD, cannabidiol (e); CBDV, cannabidivarin (f); THCV, tetrahydrocannabivarin (g); CBGA, cannabigerolic acid (h); CBG, cannabigerol (i); CBN, cannabinol (j); CBNA, cannabinolic acid (k); CBC, cannabichromene (l); CBCA, cannabichromenic acid (m); CBL, cannabicyclol (n); CBLA, cannabicyclolic acid (o). All structures drawn by Odieka, using ChemDraw Ultra 8.0.
Molecules 27 01689 g002aMolecules 27 01689 g002b
Figure 3. Chemical structures of some Cannabis sativa terpenes (Monoterpenes, Sesquiterpenes, and Triterpenoids); Mycene (a), α-Pinene (b), D-Linalool (c), Limonene (d), α-Terpinene (e), α-Phellandrene (f), α-Terpinolene (g), β-Caryophyllene (h), α-Caryophyllene (i), β-Elemene (j), Guaiol (k), Friedelin (l), and Epifriedelanol (m). All structures drawn by Odieka, using ChemDraw Ultra 8.0.
Figure 3. Chemical structures of some Cannabis sativa terpenes (Monoterpenes, Sesquiterpenes, and Triterpenoids); Mycene (a), α-Pinene (b), D-Linalool (c), Limonene (d), α-Terpinene (e), α-Phellandrene (f), α-Terpinolene (g), β-Caryophyllene (h), α-Caryophyllene (i), β-Elemene (j), Guaiol (k), Friedelin (l), and Epifriedelanol (m). All structures drawn by Odieka, using ChemDraw Ultra 8.0.
Molecules 27 01689 g003aMolecules 27 01689 g003b
Figure 4. Chemical structures of some non-cannabinoid phenols (Spirans, phenanthrenes, flavonoids, alkaloids); Cannabispiran (a), Cannabispirone (b), Canniprene (c), Cannithrene I (d), Cannithrene II (e), Debinobin (f), Canniflavin A (g), Canniflavin B (h), Cannabisativine (i), and Anhydrocannabisativine (j). All structures drawn by Odieka, using ChemDraw Ultra 8.0.
Figure 4. Chemical structures of some non-cannabinoid phenols (Spirans, phenanthrenes, flavonoids, alkaloids); Cannabispiran (a), Cannabispirone (b), Canniprene (c), Cannithrene I (d), Cannithrene II (e), Debinobin (f), Canniflavin A (g), Canniflavin B (h), Cannabisativine (i), and Anhydrocannabisativine (j). All structures drawn by Odieka, using ChemDraw Ultra 8.0.
Molecules 27 01689 g004
Table 1. Chemical compounds Isolated from Cannabis sativa.
Table 1. Chemical compounds Isolated from Cannabis sativa.
S/N *Class of CompoundsPlant Part(s)Isolated CompoundsReferences
1Cannabinoids:

-Cannabinoid acids
-Neutral cannabinoids
-Cannabinoid derivatives
-Cannabinoid acid esters
Leaves, flowers, resin, stembarks, and rootsΔ9-tetrahydrocannabivarin, α/β-fenchyl Δ9-tetrahydrocannabinolate,
Δ9-tetrahydrocannabinol, α-terpenyl (−)-Δ9-trans-tetrahydrocannabinolate, γ-eudesmyl (−)Δ9-trans-tetrahydrocannabinolate,
8α-hydroxy-(−)-Δ9-trans-tetrahydrocannabinol, 8b-hydroxy-(−)- Δ9-trans-tetrahydro cannabinol, 8-oxo-(−)-Δ9-trans-tetrahydrocannabinol, Cannabisol, (−)-Δ9-trans-tetrahydrocannabiphorol, (−)-Δ9-trans-tetrahydrocannabihexol, (−)-Δ8-trans-tetrahydrocannabinol, Δ8- trans-tetrahydrocannabinolic acid, Cannabigerol, 6,7-trans/cis-epoxycannabigerolic acid, Sesquicannabigerol, Cannabigerolic acid, Cannnabigerovarin, Cannabidiol, C4-Cannabidiol, Cannabidivarin, C4-tetrahydrocannabinol, Cannabichromene, Cannabichromevarin, Cannabichromanone, -D4-acetoxycannabichromene, Cannabicitran, Cannabiripsol, Cannabicoumaronone, Cannabifuran, Cannabielsoin, Cannabielsoic acid, Cannabicyclol, Cannabinodiol, bornyl/epi-bornyl-Δ9-tetrahydrocannabinlate Cannabinol, Cannabitriol, Cannabimovone, and Cannabioxepane
[8,11,13,24,25,26,27,28,29,30,31]
Non-Cannabinoid Constituents
2Non-cannabinoid phenol:

-Stilbenoids
-Spiro-indans
-Phenanthrenes
-Lignans, lignanamides, and phenolic amides
Leaves, flowers, stem, hemp pectin, resin, fruit, seed, and rootDihydrostilbenes, Dihydrophenathrenes,
Cannabistilbene, Canniprene, Cannithrene, Denbinobin,
Phloroglucinol β-D-glucoside, Cannabispiran, Cannabispirone, Cannabispirenone,
Cannabispirol, Cannabispirketal, a-cannabispiranol-4′-O-β-glucopyranose, prenylspirodienone, 4,5-dihydroxy-2,3,6-trimethoxy-9,10-dihydrophenanthrene, Cannabisin A–O
4,7-dimethoxy-1,2,5-trihydroxyphenathrene,
5-methyl-4-pentyl-2,6,2-trihydroxybiphenyl,
5-methyl-4-pentylbiphenyl-2,2,6-triol, N-trans-coumaroyltyramine, N-trans-feruloyltyramine, Ntrans-caffeoyltyramine, 3,3′-demethylheliotropamide, and Grossamide.
[8,13,28,32,33,34]
3Terpenoids (Terpenes):

-Monoterpenes
-Sesquiterpenes
-Diterpenes
-Triterpene,
Essential oils of fresh and dried leaves, flowers, stembarks, and rootsα-pinene, β-pinene, linalool, linalool oxide, myrcene, limonene, camphene, α-terpinene, γ-terpinene, α-terpinolene, α-terpineol, terpinene-4-ol, sabinene, sabinene hydrate, cis-sabinene hydrate, α-phellandrene, β-phellandrene, 2-methyl-2-heptene-6-one, borneol, piperitenone, geraniol, carvacrol, carvone, cis-carveol, citronellol, bornyl acetate, ipsdienol, germacrene-B, clovandiol, α-bisabolol, β-eudesmol, γ-eudesmol, α-caryophyllene, β-caryophyllene oxide, α-Humulene, Phytol, neophytadiene, friedelin (friedelan-3-one), epifriedelanol, β-amyrin, Vomifoliol, dihydrovomifoliol, and dihydroactinidiolide
β-ionone
[8,11,13,28,29,35,36]
4Flavonoids:
-Methylated
-Glycosylated (C or O glycosides)
-Prenylated
-Geranylated
Leaves, flowers, seed, and fruitOrientin, Vitexin, Isovitexin, Apigenin, Luteolin, Kaempferol, Quercetin, Cytisoside, Cytisoside glucoside, Canniflavone (Cannflavin), Naringenin, and Naringin[8,11,13,28,29,35,37,38,39]
5SterolsStembarks, roots, and leavesCampsterol, Stigmasterol, and β- Sitosterol[11,40]
7AlkaloidsRoots, leaves, stembarkCannabisativine and Anhydrocannabisativine[13,40]
8Fatty acids:
-Saturated and unsaturated fatty acids and their esters
SeedsRoughanic acid, Stearidonic acid, α-linolenic acid, and oxylipins.[8,28]
9Hydrocarbons
(n-alkane)
-Δ9-Tetrahydrocannabiorcolic acid[8]
* S/N = Serial Number.
Table 2. Reported methods of identification, isolation, and characterization of C. sativa.
Table 2. Reported methods of identification, isolation, and characterization of C. sativa.
Extraction Solvent(s)Matrix and SpeciesIdentification, Isolation, and Purification MethodsElucidation/Analytical TechniquesAnalytesReferences
Phenolic Cannabinoids
Methanol/chloroform mixture.C. sativa inflorescenceQualitative, quantitative, and comparative derivatization study of cannabinoidsFast GC–MSCBDA, CBGA, CBG, CBD, THC, Δ8-THC, CBC, THCA, THC[47]
Supercritical fluid extractionPlant biomass and medicinal Cannabis resinQuantitative and qualitative analysis of cannabinoidsUHPLC–DAD and statistical analysisCBDA, THCA, CBD, CBN, CBC, THC[48]
Methanol/chloroform solvent mixtureCannabis flower samplesQualitative and quantitative measurement of cannabinoidsHPLC–DADΔ9-THC, CBD, CBDA, THCA, CBN, CBG, CBGA, Δ8-THC[49]
Ethanol/ethanolic extracts(i) Lebanese C. sativa
(ii) Cannabis
(iii) Decarbo-xylated hemp leaves
(i) Purified by counter-current distribution and silica gel chromatography
(ii) Florisil and silica gel column chromatography
GCMS, IR, and 1H NMR comparison with an authentic sampleCannabielsoin, (+)-trans-CBT and (−)-trans -CBT-OEt-C5(Cannibitriol), Cannabicitran, and Monomethylether of CBD[13,50,51,52]
Sequential extraction (hexanes, CH2Cl2, EtOAc, EtOH, EtOH/H2O, and H2O)(i) Bud and leaves of high-potency variety of C. sativa(i) Silica gel VLC, C18-solid phase extraction (SPE), and HPLC
(ii) VLC chromatography of hexane extract, TLC, flash silica gel, Sephadex LH-20 chromatography, and semipreparative reversed-phase (RP) and chiral HPLC
HRESIMS, 1D and 2D NMR, GC–MSepi-bornyl Δ9-tetrahydrocannabinolate, α-terpenyl Δ9-tetrahydrocannabinolate, 4-terpenyl Δ9-tetrahydro-cannabinolate, α-cadinyl Δ9-tetrahydrocannabinolate, γ-eudesmyl Δ9-tetrahydro-cannabinolate, γ-eudesmyl cannabigerolate, 4-terpenyl cannabinolate, bornyl Δ9-tetrahydrocannabinolate, α-fenchyl Δ9-tetrahydro-cannabinolate, α-cadinyl cannabigerolate, Δ9-tetrahydro-cannabinol (Δ9-THC), Δ9-tetrahydrocannabinolic acid A (Δ9-THCA), Cannabinolic acid A (CBNA), and Cannabigerolic acid (CBGA), (±)-6,7-trans-epoxycannabigerolic acid, (+)-6,7-cis-epoxycannabigerolic acid, (±)-6,7-cis-epoxycannabigerol, 5′-Methoxy-cannabigerolic acid, 5′-methyl-4-pentylbiphenyl-2, 2′, 6-triol,7-methoxy-cannabispirone, (±)-6,7- trans-epoxycannabigerol, 8α-hydroxy-(−)-Δ9-trans-tetrahydrocannabinol, 8b-hydroxy-(−)- Δ9-trans-tetrahydro cannabinol, 8-oxo-(−)-Δ9-trans-tetrahydrocannabinol, 10α-hydroxy-Δ8-tetra-hydrocannabinol, 10β-hydroxy- Δ8-tetra-hydrocannabinol, 10a-α-hydroxy-10-oxo-Δ8-tetrahydrocannabinol, (±)-4-acetoxycannabichromene, (±)-3″-hydroxy-Δ(4″, 5″)-cannabichromene,(−)-7-hydroxycannabichromane,(−)-7 R-cannabicoumarononic acid A, 5-acetyl-4-hydroxycannabigerol, 4-acetoxy-2-geranyl-5-hydroxy-3-n-pentylphenol, 8-hydroxycannabinol, 8-hydroxycannabinolic acid A, and 2-geranyl-5-hydroxy-3-n-pentyl-1, 4-benzoquinone, (±)-4-acetoxycannabichromene, (±)-3”-hydroxy- Δ4”-cannabichromene, (–)-7-hydroxycannabichromane, 8-hydroxycannabinol, 8-hydroxy cannabinolic acid, 10S-hydroxy-cannabinol, 9b,10b-epoxyhexahydrocannabinol, 9a-hydroxyhexahydrocannabinol, 10a-hydroxyhexahydrocannabinol, and 10aRhydroxyhexahydrocannabinol[31,53,54,55]
Hexane extract(i) C. sativa
(air-dried and powdered buds)
(ii) high-potency variety of C. sativa
(iii) Illicit Cannabis samples
(iv) C. sativa inflorescence (strain CINRO)
(v) Lebanese C. sativa
(vi) Hemp
(vii) Nepalese and Brazilian C. Sativa.
(i) Column chromatography using silica or alumina, TLC, then fractional distillation and preparative C18 HPLC
(ii) VLC (vacuum liquid chromatography) silica gel column chromatography, C18 HPLC and chiral HPLC
(iii) Flash silica gel chromatography
(iv) Florisil column chromatography
1H NMR, 13C NMR (2D NMR) HRESIMS, circular dichroism (CD), UV, LC-HRMS, MS/MS, GC–MS, and confirmation by phytochemical transformations.Δ9-THC, Δ9-THC aldehyde, Cannabinoid esters, Cannabisol, Δ9-trans-tetrahydrocannabiphorol, Δ9-trans-tetrahydrocannabihexol, Cannabidiorcol, Cannabidihexol (CBDH) and Cannabidiphorol (CBDP), Cannabitwinol, Cannabinodivirin and cannabinodiol (CBND), Cannabichromene (CBC), 9a-hydroxyhexahydrocannabinol, 7-oxo-9a-hydroxyhexa-hydrocannabinol, 10a-hydroxyhexahydrocannabinol, and 10a-R-hydroxyhexahydrocannabinol[53,56,57,58,59,60]
Ethyl acetate extractsCannabis resins, tinctures, and leaves-GC–MS and GC–FID analysisΔ9-THC and Δ9-THCA[61]
Petroleum ether(i) Cannabis tincture of Pakistani origin
(ii) Brazilian C. sativa
(iii) Cannabis leaves and flowers (Maryland and Czechoslovakian origin)
(iv) Congo C. sativa
(v) Hashish and Cannabis sativa
(i) Silicic acid column chromatography
(ii) Silica gel and Florisil chromatography, preparative TLC
IR, NMR, MS, GC–MS confirmed by synthesisΔ9-THCV, Δ9-THCO or Δ9-THC, Δ8-THC (Δ8-THCA), Cannabielsoin acid A (CBEAA,), Cannabielsoin acid B, and Cannabicyclovarin (CBLV)[61,62,63,64,65,66,67]
Benzene(i) Fresh C. sativa leaves from Thailand
(ii) Fresh tops and leaves of C. sativa
(iii) Hemp
Polyamide and silica gel column chromatographyIR, UV, NMR, and comparing UV spectrum with that of derivativesΔ9-THCVA, CBDV, THCV, CBCV, Cannabigerovarin CBGV, cannabigerovarinic acid (CBGVA), CBDA, cannabidivarinic acid (CBDVA), Cannabicyclolic acid (CBLA), and Cannabichromenic acid (CBCA)[49,68,69]
Acetone extract(i) Leaves of C. sativa (Mexican strain)
(ii) Wax of decarboxylated aerial parts of C. sativa (Carma strain)
(iii) Cannabis variety (carmagnola)
(i) Silica gel column chromatography
(ii) Silica and alumina column chromatography, followed by normal phase (NP)-HPLC.
(iii). Flash chromatography, over reverse-phased C18 silica gel followed by normal-phase HPLC
FAB–MS, 1H-NMR, 13C-NMR), and ESI–MS semisyn-thesis.Cannabigerolic acid (CBGA), dihydroxycannabigerol derivative (camagerol), Sesquicannabi-gerol, Cannabimovone, and Cannabioxepane[70,71,72,73]
Essential/volatile oils
Methanol dilutionsCannabis sativa oil samplesSeparation/quantitation of cannabinoidsFast-GC–FIDCBD, CBN, and THC[74]
Essential oilFresh C. sativa L. from IndiaFractional distillation and chromatography over alumina.GC–MS and physico–chemical analysesα-terpinene, β-phellandrene, γ-terpinene, α-terpinolene, α-pinene, β-pinene, camphene, linalool, α-terpineol, terpinene-4-ol, linalool oxide, and sabinene hydrate[13]
Volatile/essential oils(i) Cannabis
(Dutch and Turkish)
(ii) Fresh leaves of Cannabis sativa and Cannabis indica
(i) Hydrodistillation or through nitrogen extraction
(ii) Hydrodistillation, steam distillation, and supercritical fluid extraction
Capillary gas chromato-graphy,
GC–MS analysis
cis-β-ocimene, trans-β-ocimene, α-phellandrene, D3-carene, Δ4-carene, sabinene and α-thujene, caryophyllene, humulene, trans-β-bergamotene, cis-β–farnesene, δ–limonene, carophyllene oxide, linalool, trans-α- bergamotene, cis- β -farnesene, menthol, eucalyptol, and Carvone.[13,75]
Essential oilCannabis (marijuana fresh and dried buds)SteamdistillationGC–MS and GC–FIDIpsdienol, cis-carveol, and cis-sabinene hydrate[76]
Essential oilC. sativa resinMinor terpenic component analysisGC–MS and GC retention timeα-gurjunene, α-bisabolol, α-cedrene, α-cubebene, δ-cadinene, epi-β-santalene, farnesol, γ-cadinene, γ-elemene, γ-eudesmol, guaiol, (E,E)-α-farnesene, (Z)-β-farnesene, and farnesyl acetone[77]
Essential oilCannabisSteam distillation and silica gel chromatographyGC, GC–MSeugenol, methyleugenol, iso-eugenol, trans-anethol, and cis-anethol (simple phenols)[78]
Essential oilC. sativaColumn chromatography of the essential oilGC and GC–MS analysesIso-caryophyllene, β-selinene, selina-3,7(11)-diene, and selina-4(14),7(11)-diene[13,79]
Non-cannabinoid phenols
Ethanol/ethanolic extract(i) South African Cannabis variant
(ii) Saudi Arabia hashish
(iii) Leaves of C. sativa
(iv) High-potency Cannabis variety grown in Mississippi
(v) Hemp pectin
(vi). Cannabis roots
(vii) Roots, stem, and leaves of a Mexican variant of Cannabis sativa
(i) Partitioning and chromatography on silica and polyamide columns
(ii) Normal and reversed phase chromatographic techniques
(iii) Purification by macro reticular resin, silica gel column chromatography, and Sephadex-LH-20
(iv) TLC, chromatography over alumina, and recrystallization
(v) Partitioning and TLC eluted with chloroform:acetone:ammonia (1:1:1)
(vi) Series of acid–base extractions and silica-gel chromatography followed by crystallization of the alkaloid from acetone
IR, GCMS, UV, 1D NMR (1H NMR, 13C NMR) and 2DNMR (COSY, HSQC, HMBC, and ROESY), ESI–MS, comparison with authentic samples, X-ray crystallography, and semi-synthesisβ-cannabispiranol, b-cannabispirol, 5-hydroxy-7-methoxyindan-1-spiro-cyclohexane, 7-hydroxy-5-methoxyindan-1-spiro-cyclohexane, and 5,7-dihydroxyindan-1-spiro-cyclohexane, Cannabispirketal and the glycoside, a-cannabispiranol-4′-O-β-glucopyranose, 3,4′,5-trihydroxy-dihydrostilbene, 4,5-dihydroxy-2,3,7-trimethoxy-9,10-dihydrophenanthrene, 4-hydroxy-2,3,6,7-tetramethoxy-9,10-dihydrophenanthrene and 4,7-dimethoxy-1,2,5-trihydroxyphenanthrene, Rutin, friedelin (friedelan-3-one) and epifriedelanol, Anhydrocannabisativine and cannabisativine, α,α′-dihydro-3′,4,5′-trihydroxy-4′-methoxy-3-isopentenylstilbene, α,α′-dihydro-3,4′,5-trihydroxy-4-methoxy-2,6-diisopentenylstilbene, α,α′-dihydro-3′,4,5′-trihydroxy-4′-methoxy-2′,3-diisopentenylstilbene, α,α′-dihydro-3,4′,5-trihydroxy-4,5′-diisopentenylstilbene, and combretastatin B-2[32,33,55,80,81,82,83,84]
BenzeneDried leaves of Japanese cannabisChromatographed on a polyamide column followed by silica gel chromatographyIR, 1H NMR, MS, UV.Cannabispirol and acetyl Cannabispirol[85]
AcetoneC. sativa (CARMA)Gravity column chromatography on silica gel and purified by crystallization from ether and HPLCIdentified according to its physical and spectroscopic properties and synthesisDebinoben
(1,4-phenanthrenequinone)
[86]
Sequential extraction (Hexane, EtOAc, CH2Cl2, EtOH, EtOH/H2O, and H2OHigh-potency variety of C. sativa (Mississippi)VLC, silica gel column chromatography, and RP–HPLC1D and 2D NMR, IR analysisacetoxy-6-geranyl-3-n-pentyl-1,4-benzoquinone, 4,5-dihydroxy-2,3,6-trimethoxy-9,10-dihydrophenanthrene, 4-hydroxy-2,3,6,7-tetramethoxy-9,10-dihydrophenanthrene, 4,7-dimethoxy-1,2,5-trihydroxyphenanthrene, Cannflavin C and β-sitosteryl-3-O-β-d-glucopyranoside-2′-O-palmitate, α-cannabispiranol, Chrysoeriol, 6-prenylapigenin, and Cannflavin A and β-acetyl cannabispiranol[55]
Hexane extractLeaves of Cannabis sativaIsolation by normal-phase chromatography followed by C18-HPLCNMR and ESI–MS analysisPrenylspirodinone and 7-O-methyl-cannabispirone[55,87]
Dichloro-methane extract(i) Decarboxy-lated C. sativa hemp
(ii). Thai Cannabis sativa leaves
(iii) Panamanian variety of cannabis
C18 flash chromatography, followed by silica gel gravity column chromatography and HPLCHR–ESIMS and NMR (1H, 13C, HSQC, and HMBC) data, X-ray crystallography, and confirmation by hydrogenationIsocannabispiradienone and a-Cannabispiranol, Cannabispira-dienone, and
Cannabidihydro-phenanthrene (Cannithrene1 and Cannithrene2)
[88,89]
Methanol/methanolic extract(i) Branches and leaves of hemp
(ii) Pollen grains of Mexican variety of C. sativa
(iii) Dried leaves of South African and Indian cannabis sativa
(iv) Panamanian variant of C. sativa
(v) Leaves and branches of C. sativa
(i) TLC, silica gel column chromatography, normal-phase preparative HPLC, and Sephadex LH-20 column chromatography
(ii) Partitioning, silica gel chromatography, Sephadex LH-20 chromatography, semi preparative LC
MS, 1D and 2D NMR, UV experiments, IR, X-ray crystallography and confirmation by total synthesisRutin, Quercetin-3-O-α-L-rhamnoside, kaempferol-3-O-α-L-rhamnoside, apigenin-7-O-α-L-rhamnoside, apigenin-7-O-β-D-glucopyranoside, luteolin-7-O-β-D-glucopyranoside, 1,3,6,7-tetrahydroxyl-2-C-β-D-glucopyranosyl xanthone, vitexin, orientin, apigenin-6,8-di-C-β-D-glucopyranoside, vitexin-2″-O-α-L-rhamnoside, orientin-2″-O-β-D-glucopyranoside, quebrachitol, inositol and uracil, kaempferol-3-O-sophoroside (196) and quercetin-3-O-sophoroside, cannabispirone; cannabispirenone, Cannabispiran, Isocannabispiran Canniprene, Cannabistilbene I and Cannabistibene II, and 2,3,5,6-tetramethoxy 9,10-dihydrophenanthrenedione[90,91,92,93,94]
Mixture of hydro-alcoholic and organic solventsC. sativa inflorescence (Ferimon, Uso-31, Felina 32, and Fedora 17 cultivars)Metabolic and chemical profiling to identify and quantify compounds of different classesNMR, GC–MS, UHPLC, and HPLC–PDASugars, organic acids, amino acids, cannabinoids, terpenoids, phenols, tannins, flavonoids (Quercetin, Naringenin, and Naringin) and biogenic amines[35,37]
Diethyl etherStem exudate (greenhouse-grown C. sativa)TLC and acid hydrolysis of the exudate1H NMR and GC–MSPhloroglucinol β-D-glucoside[95]
Table 3. Summary of reported bioactivities associated with isolated compounds and essential oils from Cannabis sativa.
Table 3. Summary of reported bioactivities associated with isolated compounds and essential oils from Cannabis sativa.
Isolated Bioactive CompoundBioactivity/UsesReferences
Tetrahydrocannabinol THCAntioxidant, anti-pruritic, and anti-inflammatory effects[29,100]
Cannabidiol CBDAnti-convulsive, anti-inflammatory, immunosuppressive properties, antioxidant, and anti-psychotic effects[101,102,103]
Cannabigerol CBGAnti-fungal effects, anti-cancer, anti-depressant, mild anti-hypertensive agent, analgesic, and anti-erythemic effects[29,104]
Cannabichromene CBCAnti-inflammatory and analgesic[29]
Cannabinol CBNSedative, anti-convulsant, anti-inflammatory, antibiotic, and anti-MRSA activity[29]
Tetrahydrocannabivarin THCVAnti-convulsant[29]
Tetrahydrocannabinolic acid THCAImmunomodulatory, anti-inflammatory, neuroprotective, anti-neoplastic activity, and antiemetic effects[29,105]
Cannabidavarin CBDVAnti-convulsant (anti-epileptic) properties and anti-emetic properties[106,107]
Cannabidiolic acid CBDAAnti-emetic effects[104,108,109,110]
β-MyrceneAnti-inflammatory and analgesic sedative agent[7,29,111]
D-LimoneneStrongly anxiolytic, anti-depressant, antibiotic, and anti-cancer agent[7,29]
β-OcimeneAnti-convulsant activity, anti-fungal activity, anti-tumor activity, and pest resistance[112,113]
γ-Terpinene
Anti-inflammatory activity, antioxidant, and anti-proliferative activity[114,115]
α-TerpineneAntioxidant[29]
α-PineneAnti-inflammatory, bronchodilator, anti-microbial,
and anxiolytic effects
[7,11,29,116]
LinaloolAnalgesic and anticonvulsant, anxiolytic, anti-depressant, anti-glutamatergic, anti-leishmanial activity, anticancer agent, anti-nociceptive, and anti-depressant effects[111,117,118,119]
α-PhellandreneAnti-nociceptive, anti-depressant, anti-arthritic and allergic, and anti-hyperalgesic effects[120,121,122]
TerpinoleneAnti-fungal and larvicidal, anti-nociceptive, anti-inflammatory antioxidant, and anti-cancer effects[123,124]
β-CaryophylleneCardio-protective, hepato-protective, gastro-protective, neuro-protective, nephro-protective, antioxidant, anti-inflammatory, anti-microbial, anti-pruritic, and immunomodulatory activities[7,125,126,127]
Caryophyllene OxideAnti-fungal, insecticidal/anti-feedant, and anti-platelet effects[29]
β-ElemeneAnti-cancer and anti-tumor[128]
GuaiolAnti-inflammatory, antioxidant, anti-cancer
anti-rheumatic, antiseptic, diaphoretic, diuretic, and laxative effects
[29,129]
FriedelinAnti-inflammatory, anti-pyretic, and anti-tuberculosis agent[130,131]
EpifriedelanolAntioxidant[132]
Cannflavin A and BAnti-inflammatory, neoplastic, antioxidant, neuroprotective, anti-parasitic, and anti-viral agent[4,7,29,99]
ApigeninAnxiolytic and estrogenic properties, anti-tumor, antioxidant, anti-inflammatory, anti-osteoporosis, and immune regulation effects[4,133]
Vitexin and IsovitexinAntioxidant, anti-cancer, anti-inflammatory, anti-diabetic, anti-microbial, anti-viral, anti-hyperalgesic, and neuroprotective effects[134]
QuercetinAnti-cancer/anti-proliferator, antioxidative/anti-aging, anti-viral, anti-inflammatory, cardio-protective, skin-protective, anti-coagulant, and anti-platelet effects[135]
LuteolinNeuroprotective effects, anti-inflammatory, and antioxidant effects[136]
LignansAntioxidant, anti-viral, anti-diabetic, anti-tumorigenic, and anti-obesity activities[4]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Odieka, A.E.; Obuzor, G.U.; Oyedeji, O.O.; Gondwe, M.; Hosu, Y.S.; Oyedeji, A.O. The Medicinal Natural Products of Cannabis sativa Linn.: A Review. Molecules 2022, 27, 1689. https://doi.org/10.3390/molecules27051689

AMA Style

Odieka AE, Obuzor GU, Oyedeji OO, Gondwe M, Hosu YS, Oyedeji AO. The Medicinal Natural Products of Cannabis sativa Linn.: A Review. Molecules. 2022; 27(5):1689. https://doi.org/10.3390/molecules27051689

Chicago/Turabian Style

Odieka, Anwuli Endurance, Gloria Ukalina Obuzor, Opeoluwa Oyehan Oyedeji, Mavuto Gondwe, Yiseyon Sunday Hosu, and Adebola Omowunmi Oyedeji. 2022. "The Medicinal Natural Products of Cannabis sativa Linn.: A Review" Molecules 27, no. 5: 1689. https://doi.org/10.3390/molecules27051689

Article Metrics

Back to TopTop