Canna~Fangled Abstracts

Cannabis for Medical Use: Versatile Plant Rather Than a Single Drug

By April 25, 2022May 15th, 2022No Comments


Journal List > Front Pharmacol > PMC9081504

Abstract

Medical Cannabis and its major cannabinoids (−)-trans9-tetrahydrocannabinol (THC) and cannabidiol (CBD) are gaining momentum for various medical purposes as their therapeutic qualities are becoming better established. However, studies regarding their efficacy are oftentimes inconclusive. This is chiefly because Cannabis is a versatile plant rather than a single drug and its effects do not depend only on the amount of THC and CBD. Hundreds of Cannabis cultivars and hybrids exist worldwide, each with a unique and distinct chemical profile. Most studies focus on THC and CBD, but these are just two of over 140 phytocannabinoids found in the plant in addition to a milieu of terpenoids, flavonoids and other compounds with potential therapeutic activities. Different plants contain a very different array of these metabolites in varying relative ratios, and it is the interplay between these molecules from the plant and the endocannabinoid system in the body that determines the ultimate therapeutic response and associated adverse effects. Here, we discuss how phytocannabinoid profiles differ between plants depending on the chemovar types, review the major factors that affect secondary metabolite accumulation in the plant including the genotype, growth conditions, processing, storage and the delivery route; and highlight how these factors make Cannabis treatment highly complex.

Keywords: cannabis, chemovar, phytocannabinoids, terpenoids, secondary metabolites

Introduction

The past 2 decades have seen a major increase in the use of medical Cannabis as its therapeutic virtues are becoming better known and accepted (). These therapeutic qualities were attributed to a naturally-occurring unique family of secondary metabolites termed phytocannabinoids. The most abundant and best-known phytocannabinoids are the psychoactive (−)-trans9-tetrahydrocannabinol (THC), which was first isolated and structurally elucidated by Mechoulam and colleagues in 1964 (); and cannabidiol (CBD), which was extracted in 1940 () and its full chemical structure was elucidated in 1963 by the same Mechoulam (). CBD has been gaining interest since the 1980s when CBD oil was found to possess anti-epileptic properties (), and the CBD molecule was later shown to possess a wide range of therapeutic effects (). However, THC and CBD are just two of more than 140 distinctive phytocannabinoids that have been identified so far in different Cannabis plants ().

The isolation of phytocannabinoids from the Cannabis plant has led to the discovery of endogenous cannabinoids (endocannabinoids, eCBs) in vertebrates (). THC was found to bind a specific G-protein-coupled receptor, which was named cannabinoid receptor 1 (CB1) (). A second receptor, which was named CB2, was identified by homology (). Following the discovery of the receptors, their endogenous lipid ligands were identified. The first two and best-studied are N-arachidonoylethanolamine (anandamide) () and 2-arachidonoylglycerol (2-AG) (). These eCBs and their specific receptors, CB1 and CB2, form the classical endocannabinoid system (eCBS) (), a ubiquitous neuromodulatory signaling system that has widespread functions in the brain and throughout the body. Since its inception, the term eCBS was expanded and now additional cannabinoid receptors, additional eCBs and cannabimimetic lipids as well as the enzymes involved in their synthesis and degradation are recognized as part of the extended eCBS (). Many of the pharmacological and therapeutic properties of phytocannabinoids rely on their interactions with the eCBS. The numerous and versatile effects of Cannabis result from the involvement of the eCBS in multiple processes. It regulates many physiological processes in health and disease (). It is involved in the maintenance and homeostasis of many vital functions including immune response (), cardiovascular activity (), memory () and pain sensation (). This makes Cannabis treatment especially valuable since targeting the eCBS and its modulation by phytocannabinoids has been emerging as novel pharmacotherapy, with therapeutic potential suggested in a multitude of diseases affecting humans.

In the last decade, there has been a rapid growth in the discovery and use of pure THC, pure CBD and Cannabis-based extracts for various medical purposes. Results regarding the efficacy of Cannabis-based extracts are oftentimes inconclusive and sometimes even conflicting. That is because the effects of Cannabis extracts do not depend merely on the amount of THC and CBD (). Cannabis is a versatile plant rather than a single drug and importantly, studies involving pure THC or CBD do not reflect the potential benefits of full-spectrum extracts (). For example, THC and CBD were both effective in reducing neuropathic pain in various mice and rat models (). However, the pain-relieving effects were enhanced by their combination (). Moreover, a controlled high-CBD extract with additional secondary metabolites from the plant was more effective than purified CBD or THC at the same dose as in the extract (). In studies involving patients with multiple sclerosis, full-spectrum extracts demonstrated more beneficial effects for pain relief and reducing inflammation than pure THC and CBD (). We have recently shown that both high-THC and high-CBD extracts were effective in reducing chronic pain, however, specific phytocannabinoid compositions were associated with more adverse effects (). We also found Cannabis extracts effective in reducing migraine frequency, and here again, the presence of a few minor phytocannabinoids in the extracts made some more effective than others regardless of their THC or CBD content ().

Bioactive Secondary Metabolites From Cannabis as Therapeutic Agents

Phytocannabinoids are conventionally classified into 10 subclasses based on their chemical structure and an 11th miscellaneous types group (Figure 1) (). They are lipophilic compounds biosynthesized by the convergence of two main plant pathways: the polyketide and the plastidial non-mevalonate-dependent isoprenoid (MEP) pathways. Phytocannabinoids are made of a resorcinyl core with a carboxyl group (COOH) on the aromatic ring, an alkyl side-chain of varying length that typically contains an odd number of carbon atoms (one to seven carbons), and a terpene moiety (). The most abundant type of phytocannabinoids in Cannabis are those with a pentyl side-chain (five carbons), with cannabigerolic acid (CBGA) as the first cannabinoid compound, made by the prenylation of olivetolic acid with the isoprenoid geranyl pyrophosphate (GPP) (). Other phytocannabinoid subclasses, including (−)-trans9-tetrahydrocannabinolic acid (THCA), cannabidiolic acid (CBDA) and cannabichromenic acid (CBCA) are derived from CBGA-type phytocannabinoids via specific enzymatic reactions (). Thus, only these four subclasses are biosynthesized in the plant while the remaining subclasses are the result of different degradation routes and chemical processes such as oxidation, photochemical reaction, double bond isomerization, and others. The well-known neutral phytocannabinoids result from the decarboxylation of the acid compounds, where the carboxyl group is removed and carbon dioxide is released. In the less common cases, instead of olivetolic acid other molecules with different length alkyl side-chain serve as precursors. These undergo the same enzymatic and chemical reactions, resulting in a range of additional phytocannabinoids () such as the three-carbon cannabigerovarinic acid (CBGVA), (−)-trans9-tetrahydrocannabivarinic acid (THCVA) and cannabidivarinic acid (CBDVA), or the seven-carbon (-)-trans-Δ9-tetrahydrocannabiphorol (THCP) and cannabidiphorol (CBDP) () and others. Cannabinoid derivatives that were previously detected by MS methods are presented in Figure 1 (). Though initially considered unique to the Cannabis plant, other plant-derived natural products that are able to interact with ECS receptors were later discovered in other types of plants, such as Radula marginata and Piper nigrum ().

An external file that holds a picture, illustration, etc.
Object name is fphar-13-894960-g001.jpg

Phytocannabinoids are divided into subclasses according to their structure. Prenylation of olivetolic acid with the isoprenoid geranyl pyrophosphate forms CBGA. Less frequently, instead of olivetolic acid other molecules with different length alkyl side-chain serve as precursors. The acid forms THCA, CBDA and CBCA are synthesized in the Cannabis plant from CBGA. The neutral forms and other subclasses of phytocannabinoids are the result of chemical processes such as decarboxylation, isomerization and others. The shared core of olivetolic acid by the different subclasses is depicted in red. Subclass 11 includes phytocannabinoids identified by mass spectrometry in different Cannabis chemovars, whose structures have not been elucidated yet.

In addition to phytocannabinoids, the other major active secondary metabolites of Cannabis are terpenes and terpenoids (generally termed terpenoids). Terpenes are naturally occurring volatile unsaturated hydrocarbon biomolecules built up by branched 5-carbon isoprene units, sharing the same isoprenoid precursor as phytocannabinoids. Terpenoids are modified terpenes that contain additional functional groups, usually varying oxygen arrangements or oxidation states. Monoterpenoids are built by two isoprene units (10 carbons) and sesquiterpenoids are built up by three isoprene units (15 carbons) (). Monoterpenoids and phytocannabinoids share the common biosynthetic precursor GPP and are both biosynthesized in the plastid, while sesquiterpenoids are synthesized in the cytosol from farnesyl pyrophosphate (). Terpenoids are responsible for the fragrance and taste of plants as they are characterized by a strong and pleasant aroma (). Terpenoids are also suggested to have roles in protection from predation and attraction of pollinators. Terpenoids were shown to exert synergistic effects when combined with the phytocannabinoids in Cannabis and contribute crucially to its therapeutic effects (), and were also suggested to possess therapeutic effects of their own (). Terpenoids are widely distributed in plants and a few are also present in other species including some animals and microorganisms ().

Various flavonoids are also found in Cannabis and may give the plant some of its exclusive medicinal benefits (). Flavonoids are hydroxylated polyphenolic compounds consisting of two benzene rings linked via a heterocyclic pyran ring (). Three specific prenylated flavonoids, termed cannflavins A-C, are unique to Cannabis and show potent anti-inflammatory capabilities (). Cannabis plants produce additional kinds of secondary metabolites including various alkaloids, stilbenoids and others (), but little is known regarding their biosynthesis and regulation and whether they possess any therapeutic value remains to be elucidated.

New Analytical Approaches for Secondary Metabolites Profiling

It is the phytocannabinoids, terpenoids, flavonoids and other constituents in Cannabis, as well as their interplay, that determines the medicinal outcomes and adverse effects. As there is wide variability in their contents in different Cannabis plants (), there is a great need for their accurate chemical analyses that will help better understand the complexity and diversity of Cannabis compounds. Identification and quantification of phytocannabinoids and flavonoids can be achieved via gas chromatography (GC), either coupled to a flame ionization detector or a mass-spectrometer (MS). However, there are a few limitations to this method, as some analytes may not be sufficiently separated and decomposition is required for accurate quantification. Therefore, an alternative method using ultra-high-performance liquid chromatography with an ultraviolet detector (UHPLC/UV) and electrospray ionization-liquid chromatography/mass spectrometry (ESI-LC/MS) () allows for a high-resolution separation of components, without decomposition or derivatization prior to analysis. While UV detection is more appropriate for abundant components having analytical standards (such as THC, CBD and their corresponding acids), the use of mass spectrometry allows comprehensive identification and quantification of additional molecules, both abundant and rare. Additionally, MS and MS/MS analyses enable the identification of unknown molecules and their semi-quantification. Reference MS/MS data for identification of phytocannabinoids is available for labs and experts for putative identification (). Terpenoids can be detected using static headspace gas chromatography-tandem MS (SHS-GC/MS/MS) (). Similar to phytocannabinoids, terpenoids with no commercially available analytical standards can still be semi-quantified relying on the calibration curves of molecules with standards and relying on both similar MS spectral characteristics and similar retention times ().

Strains, Cultivars and Chemovars

Cannabis is one genus with one species, sativa L. (), which is sometimes divided into subspecies including in addition to sativa also indica and ruderalis. These Cannabis subspecies are divided into hundreds of different Cannabis cultivars and hybrids. Cultivar stands for cultivated variety, a plant that has been selected for cultivation. A Cannabis strain refers to plants reproduced asexually from a cultivar through clonal propagation. Cannabis cultivars worldwide vary significantly in their chemical compositions. Therefore, a Cannabis chemovar refers to the chemical profile of the plant and is considered a more useful classification in medicine (). Medical Cannabis has been divided into three phenotypic chemovar groups according to its content of THC and CBD: Type I which is THC-predominant, Type II in which the two are balanced and Type III which is CBD-predominant ().

From the genotypic perspective, Cannabis chemovar classification involves two codominant alleles on locus B, allele BT is specific to THCA and allele BD is specific to CBDA (). Thus, Type I chemovar is BT/BT, Type III is BD/BD and Type II is BT/BD. The nonfunctional allele B0 does not allow for the conversion of the precursor CBGA into THCA or CBDA, and is sometimes referred to as Type IV chemovar, which is CBGA-predominant. An independent gene at locus C codes for CBCA synthase that produces CBCA from CBGA (). Studies showed that type I chemovar dominates the markets, but often it is not as beneficial as the other chemovars in achieving the desired symptom relief (). Moreover, the minor phytocannabinoid are not randomly distributed between the different chemovar types. As is shown in the heatmap presented in Figure 2, phytocannabinoids from cannabitriol (CBT) and cannabinol (CBN) families are more abundant in Type I chemovars, as they are predominantly the degradation products of THC. They can also be found in type II chemovars, though their concentration would generally be lower due to limitation in the amount of available precursor. Similarly, phytocannabinoids from cannabielsoin (CBE) family are more abundant in Type III chemovars as they are the degradation products of CBD and can also be found in type II chemovars to a lesser extent. Type-IV chemovars contain unique phytocannabinoids from the cannabigerol (CBG) family and high levels of phytocannabinoids from the cannabichromene (CBC) family, as CBCA synthase is intact. These selective distributions among chemovars are the result of metabolic pathways unique to either THC or CBD, which are not found in type IV chemovars. The distribution of particular phytocannabinoids according to chemovar is presented in Figure 3. Variations in the minor phytocannabinoid contents of different Cannabis extracts lead to varied effects on the eCBS, stressing the importance of their characterization in assessing cannabis effectivity (). The high variability in the concentration of phytocannabinoid from 10 subclasses in their acidic and neutral forms in the inflorescences of 320 different cultivars is presented in Table 1.

An external file that holds a picture, illustration, etc.
Object name is fphar-13-894960-g002.jpg

Minor phytocannabinoids are associated with Type I, Type III and Type IV chemovars. Heatmap presenting the concentration of phytocannabinoids (% weight per weight) divided by chemovars. Type I chemovars defined THCA >20% (n = 13), Type III chemovars defined CBDA >15% (n = 9), Type II defined THCA >4% and CBDA >10% (n = 13), type IV defined CBGA >6% (n = 4). Groups of unique phytocannabinoids are depicted by a surrounding black square.

An external file that holds a picture, illustration, etc.
Object name is fphar-13-894960-g003.jpg

Venn diagram of the distribution of particular phytocannabinoids to specific chemovars. Examples of unique phytocannabinoids per chemovar type are shown in the appropriate subgroup.

TABLE 1

Variability of phytocannabinoids in 320 different cultivars.

Presented as concentration values (%w/w) Max Min Average Std dev
1. Cannabigerol (CBG) type
 Acids CBGA 6.182 0.012 0.400 0.464
CBGA-C4 0.028 0.000 0.001 0.002
CBGVA 0.024 0.000 0.000 0.002
CBGOA 0.004 0.000 0.000 0.000
CBGMA 0.003 0.000 0.000 0.000
Sesqui-CBGA 0.006 0.000 0.001 0.001
 Neutrals CBG 0.735 0.000 0.084 0.067
CBG-C4 0.001 0.000 0.000 0.000
CBGV 0.003 0.000 0.000 0.000
CBGO 0.000 0.000 0.000 0.000
CBGM 0.000 0.000 0.000 0.000
Sesqui-CBG 0.042 0.000 0.008 0.006
2. Δ 9 -trans-tetrahydrocannabinol (Δ 9 -THC) type
 Acids THCA 24.325 0.124 10.390 6.425
THCA-C4 0.192 0.000 0.044 0.036
THCVA 1.120 0.000 0.124 0.135
THCOA 0.113 0.000 0.021 0.021
THCMA 0.062 0.000 0.011 0.011
 Neutrals THC′ 7.058 0.000 0.948 1.076
THC-C4 0.062 0.000 0.003 0.008
THCV 0.147 0.000 0.007 0.016
THCO 0.000 0.000 0.000 0.000
THCM 0.000 0.000 0.000 0.000
3. Cannabidiol (CBD) type
 Acids CBDA 18.351 0.000 3.085 4.968
CBDA-C4 0.094 0.000 0.009 0.016
CBDVA 1.096 0.000 0.041 0.129
CBDOA 0.053 0.000 0.003 0.007
CBDMA 0.011 0.000 0.001 0.002
 Neutrals CBD 2.676 0.000 0.166 0.363
CBD-C4 0.068 0.000 0.001 0.004
CBDV 0.105 0.000 0.002 0.010
CBDO 0.003 0.000 0.000 0.000
CBDM 0.007 0.000 0.000 0.000
4. Cannabichromene (CBC) type
 Acids CBCA 2.835 0.003 0.251 0.284
CBCA-C4 0.006 0.000 0.001 0.001
CBCVA 0.083 0.000 0.003 0.010
CBCOA 0.018 0.000 0.001 0.002
 Neutrals CBC 0.830 0.000 0.034 0.055
CBC-C4 0.001 0.000 0.000 0.000
CBCV 0.014 0.000 0.000 0.001
CBCO 0.000 0.000 0.000 0.000
5. Cannabinol (CBN) type
 Acids CBNA 0.499 0.000 0.066 0.081
CBNA-C4 0.002 0.000 0.000 0.000
CBNVA 0.006 0.000 0.000 0.001
CBNOA 0.001 0.000 0.000 0.000
CBNA-8-OH 0.000 0.000 0.000 0.000
 Neutrals CBN 0.721 0.000 0.017 0.049
CBN-C4 0.001 0.000 0.000 0.000
CBNV 0.002 0.000 0.000 0.000
CBNO 0.000 0.000 0.000 0.000
CBNM 0.000 0.000 0.000 0.000
CBN-8-OH 0.001 0.000 0.000 0.000
6. Δ 8 -trans-tetrahydrocannabinol (Δ 8 -THC) type
 Neutral d8-THC 0.137 0.000 0.001 0.012
7. Cannabicyclol (CBL) type
 Neutral CBL 0.040 0.000 0.000 0.003
8. Cannabinodiol (CBND) type
 Acids CBNDA 0.014 0.000 0.001 0.002
CBNDVA 0.000 0.000 0.000 0.000
 Neutral CBND 0.127 0.000 0.002 0.011
9. Cannabielsoin (CBE) type
 Acids CBEA 0.056 0.000 0.004 0.008
CBEVA 0.001 0.000 0.000 0.000
 Neutrals CBE 0.007 0.000 0.000 0.001
CBEV 0.008 0.000 0.000 0.000
10. Cannabitriol (CBT) type
 Acids CBTA-1 0.203 0.000 0.005 0.013
CBTA-3 0.084 0.000 0.009 0.012
 Neutrals CBT-1 0.220 0.000 0.013 0.020
CBTV-1 0.011 0.000 0.000 0.001
CBT-3 0.172 0.000 0.009 0.015
CBTV-3 0.010 0.000 0.000 0.001
CBT-2 0.046 0.000 0.004 0.007

n = 320 inflorescences from cultivars; results are concentration values of phytocannabinoids per plant (%w/w).

In addition, the Cannabis plant contains an overwhelming milieu of terpenoids, but only a limited number are currently reported and used for metabolic analyses of Cannabis chemovars (). Terpenoids content in different cultivars of Cannabis is highly variable, with some terpenoids being more associated with specific cultivars (). Studies that assessed terpenoid metabolism found the monoterpenoids limonene, β-myrcene, terpinolene and α-pinene, and the sesquiterpenoids β-caryophyllene and humulene, were abundant in the majority of Cannabis chemovars (). Some terpenoids were predominantly found only in Type I chemovars and others only in Type III, suggesting joint metabolic pathways and chemovar-specific aroma and effects (). Table 2 summarizes the variability of monoterpenoids and sesquiterpenoids in 79 distinct Cannabis inflorescences (out of the 320 described for phytocannabinoids in Table 1).

TABLE 2

Variability of terpenoids in 79 different cultivars.

Compound Max (ppm) Min (ppm) Average (ppm) Std dev (ppm) V (%)
α-Pinene 1903.4 2.3 181.7 357.5 196.8
Camphene 161.6 1.7 18.3 34.0 186.3
Sabinene 3.7 0.0 0.9 1.1 126.3
β-Pinene 1705.3 2.2 132.1 259.9 196.8
β-Myrcene >2,706 5.1 444.3 706.4 159.0
3δ-Carene 530.3 0.0 10.1 62.7 622.3
α-Phellandrene 701.5 0.0 14.0 80.5 574.7
α-Terpinene 379.0 0.0 14.9 51.1 343.1
Limonene >2,760 2.7 247.5 577.7 233.5
β-Phellandrene 421.1 0.0 16.9 55.8 330.6
cis-Ocimene 101.6 0.0 4.3 12.9 302.4
Eucalyptol 63.6 0.0 7.3 11.9 162.9
p-Cymene 28.7 0.0 2.2 4.3 192.5
trans-Ocimene 1,648.5 0.0 62.9 237.6 377.8
γ-Terpinene 512.2 1.5 16.4 60.7 369.2
Terpinolene >2,433 2.4 96.2 394.4 410.1
Linalool 1,204.4 0.0 214.1 249.8 116.7
Fenchone 68.0 0.0 8.9 12.5 139.5
Fenchol 953.7 0.0 118.1 145.4 123.1
C10H18O-154 (99/93/79/121)-1* 222.6 0.0 25.8 43.3 168.3
C10H18O-154 (99/93/79/121)-2* 29.3 0.0 0.4 3.3 0.0
Menthol 62.1 0.0 5.6 12.9 230.7
Borneol 941.0 0.0 59.8 123.7 206.7
Camphor 20.4 0.0 1.0 2.6 257.5
Terpinen-4-ol 149.3 0.0 17.9 29.3 163.2
α-Terpineol 1,027.8 0.0 98.1 148.3 151.1
Citronellol 129.1 0.0 12.7 25.9 204.3
Nerol 26.2 0.0 2.5 5.4 218.6
Geraniol 93.8 0.0 4.1 14.1 347.1
Bornyl acetate 37.2 0.0 2.8 6.6 236.5
α-Cubebene* 8.2 0.0 2.2 1.9 110.8
Isoledene 7.7 0.0 0.1 0.9 885.1
Cyclosativene 11.6 0.0 0.2 1.3 728.5
Ylangene* 74.5 0.0 5.6 9.8 176.4
α-Copaene* 12.9 0.0 2.2 2.4 118.0
α-Funedrene 1.8 0.0 0.9 0.5 146.0
7-epi-Sesquithujene* 76.3 0.0 13.3 15.4 116.3
C15H24-204 (105/(120+119)/161)* 13.4 0.0 2.8 3.2 114.7
Sativene 2.5 0.0 1.0 1.1 106.4
β-Cubebene* 5.2 0.0 0.1 0.6 886.1
Sesquithujene* 116.0 0.0 14.9 17.6 118.4
β-Isocomene* 41.3 0.0 6.7 8.5 126.1
α-Santalene* 71.5 0.0 7.7 11.0 142.8
cis-α-Bergamotene* 23.6 0.0 3.9 4.4 113.2
α-Cedrene 3.9 0.0 1.1 1.1 104.7
trans-α-Bergamotene* 63.6 0.0 7.0 13.8 196.5
β-Caryophyllene >3,631.5 9.0 670.8 781.0 116.4
Geranyl acetate 1.8 0.0 0.4 0.6 144.7
β-Cedrene 18.5 0.0 1.0 2.9 284.5
α-Guaiene* 567.3 0.0 58.6 107.6 183.7
γ-Elemene* 161.1 0.0 11.9 24.1 202.5
Aromadendrene 8.3 0.0 1.9 2.2 120.3
β-Santalene* 39.2 0.0 3.0 5.7 187.8
Guaia-6,9-diene* 65.4 0.0 7.3 12.5 171.3
trans-β-Farnesene 617.3 3.2 44.5 71.7 161.4
C15H24-204 (69/91/105/161)* 25.7 0.0 3.9 6.3 160.6
C15H24-204 (91/105/161)* 302.3 0.0 12.3 34.2 279.0
C15H24-204 (161/105/133/91)* 44.4 0.0 9.1 11.0 121.5
C15H24-204 (105/91/133/161/189)* 44.2 0.0 9.5 11.0 115.9
α-Humulene 2,134.2 12.5 255.8 283.9 111.0
Alloaromadendrene 104.1 0.0 12.0 17.3 143.7
Acoradiene* 10.5 0.0 1.2 2.3 195.4
C15H24-204 (105)-1* 35.4 0.0 8.3 10.3 123.6
γ-Curcumene* 432.5 0.0 9.7 48.6 500.5
C15H24-204 (189/133)-1* 101.4 0.0 19.9 25.9 130.2
Sesquisabinene* 56.8 0.0 5.0 8.8 173.6
γ-Muurelene* 60.9 0.0 7.6 10.7 140.2
α-Amorphene* 27.0 0.0 6.7 7.5 112.5
Aristolochene* 14.6 0.0 1.8 2.7 152.4
Germacrene D* 28.3 0.0 4.3 7.7 180.4
β-Chamigrene 16.3 0.0 0.5 2.6 488.0
C15H24-204 (189/133)-2* 196.3 0.0 44.0 46.9 106.6
C15H24-204 (119/93/161)* 28.1 0.0 3.6 5.7 157.4
α-Selinene* 92.0 0.0 16.7 21.1 126.4
Ledene 6.0 0.0 0.1 0.7 688.9
α-Curcumene 69.9 0.0 9.8 17.6 180.2
Valencene 402.8 0.0 26.6 80.5 302.9
β-Selinene* 716.9 0.0 133.0 184.3 138.5
α-Farnesene* 88.7 0.0 6.8 13.4 196.4
β-Bisabolene* 663.2 0.0 42.7 87.0 204.0
δ-Guaiene* 560.0 0.0 47.2 96.5 204.3
C15H24-204 (119/161/105/134)* 32.6 0.0 6.2 7.8 125.4
β-Curcumene 27.9 0.0 5.0 6.6 130.0
Dihydroagarofuran* 15.3 0.0 2.2 3.2 145.7
C15H24-204 (similar Germarcene B)* 32.7 0.0 8.5 9.5 111.7
Sesquicineole* 135.1 0.0 9.4 16.5 175.2
Eremophilene* 38.6 0.0 9.0 11.5 128.6
β-Sesquiphellandrene* 77.7 0.0 9.3 14.0 149.9
γ-Cadinene* 22.3 0.0 4.5 5.8 128.7
δ-Cadinene* 27.4 0.0 7.0 6.7 95.8
C15H24-204 (105)-2* 28.9 0.0 7.3 8.7 118.6
α-Panasinsene* 31.3 0.0 1.3 3.9 288.6
trans-α-Bisabolene* 512.1 0.0 86.2 97.9 113.6
Selina-3,7 (11)-diene* >1,334.1 0.0 249.3 361.5 145.0
trans-Nerolidol 1,637.2 0.0 102.1 240.3 235.3
Germacrene B* 923.0 0.0 25.7 107.1 417.0
Globulol 31.2 0.0 0.5 3.6 698.6
Guaiol >2099 0.0 568.1 765.5 134.7
Caryophyllene oxide >1890 11.2 308.5 488.4 158.3
α-epi-7-epi-5-Eudesmol* 319.1 0.0 30.8 47.5 154.2
C15H26O-222 (similar γ-Eudesmol)* >2099 0.0 541.8 751.1 138.6
Selina-6-en-4-ol* 180.3 0.0 29.8 46.0 154.2
γ-Eudesmol* >1,588 0.0 296.1 478.4 161.6
Hinesol* 196.1 0.0 33.1 39.2 118.5
C15H26O-222 (105/161/59)-1* 496.1 0.0 63.8 108.8 170.5
Agarospirol* 158.1 0.0 14.7 26.2 178.5
C15H26O-222 (105/161/59)-2* 812.2 0.0 78.3 135.2 172.5
C15H26O-222 (59/81/107/149/161)* 566.6 0.0 66.1 96.2 145.6
α-Eudesmol* >1,588 0.0 377.3 541.5 143.5
β-Eudesmol >1,588 0.0 434.2 573.0 132.0
7-epi-α-Eudesmol* 573.7 0.0 61.0 108.5 177.9
Bulnesol* >2099 0.0 159.9 318.0 198.9
α-Bisabolol >3,791 0.0 1,515.7 1,592.2 105.0
Total monoterpenoids [ppm] 18,783.3 44.5 1842.0 2,896.2 157.2
Total monoterpenoids [%] 1.88 0.00 0.18 0.29 0.02
Total sesquiterpenoids [ppm] 25,135.2 147.6 6,678.2 5,089.5 76.2
Total sesquiterpenoids [%] 2.51 0.01 0.67 0.51 0.01
Total terpenoids [ppm] 26,501.4 196.1 8,520.2 6,047.6 71.0
Total terpenoids [%] 2.65 0.02 0.85 0.60 0.01

n = 79 inflorescences from cultivars; ppm–parts per million, > values above upper limit of detection, % represents concentration values of terpenoids per plant, * terpenoids that were semi-quantified.

Each Cannabis cultivar contains a different profile of more than 500 secondary metabolites (). The fact that hundreds of different Cannabis cultivars and hybrids exist worldwide, varying significantly in their chemical compositions, makes Cannabis treatment highly complex. Moreover, sometimes the outcome of treatment with medical Cannabis depends on the way its secondary metabolites act together synergistically, in a mechanism first described by Ben-Shabat and Mechoulam for eCBs () and later postulated by Russo as the ‘entourage effect’ for phytocannabinoids (). Thus, phytocannabinoids that are found together in a Cannabis chemovar modulate each other’s activity and thus the overall effect. The entourage effect postulates that the presence of minor phytocannabinoids, terpenoids and other plant metabolites contributes to the overall response in a way that significantly modulates the effects of the main active components, THC and CBD, and thereby produces more potent or more selective effects. Several studies have shown whole extracts or a combination of THC and CBD, with either each other, minor phytocannabinoids or terpenoids, are more effective than the corresponding major phytocannabinoid in producing the same response (). However, other studies did not find evidence that common terpenoids can bind eCBS receptors or modulate the effect of phytocannabinoids on the receptors (). A better understanding of the different components in Cannabis and the way they act together is required to fully utilize its therapeutic potential to the fullest.

Pre- and Post-Harvest Conditions

The concentrations of the different compounds in the plant depend on many factors. There is a strong genotypic influence on the composition of secondary metabolites in different Cannabis chemovars (). However, a very large variation exists also in the profiles of genetically identical plants grown under different conditions (). For example, we previously showed the differences in phytocannabinoids profiles of a high-CBD Cannabis chemovar that was used to treat refractory childhood epilepsy in Israel (). While the genetically identical plants from four different greenhouses were planted and harvested in the same way and at the same time, and considered as the same treatment, their CBDA contents were similar but they portrayed substantial differences in many other phytocannabinoids.

In addition to the genetic variety, many environmental factors affect the composition of the secondary metabolites in the Cannabis plant (). These include growth conditions such as humidity, light quality and intensity, CO2 concentration and mineral nutrition (). The tissue type is also an important factor as within the plant there is a location- and organ-specific distribution of the active secondary metabolites (). Phytocannabinoids are synthesized in glandular trichomes that are located in the highest density on the inflorescences of unfertilized female plants (), and their accumulation varies in the different aerial parts (flowers, fan leaves, inflorescence leaves, stalk and stem). Accumulation patterns also depend on the age of that part (). A study that tested phytocannabinoid and terpenoid content in the plant from the rooting until the end of the flowering stage () found that the accumulation of some major phytocannabinoids and monoterpenoids requires longer growth time in plants from Type II and Type III chemovars than in Type I. The functional roles of phytocannabinoids and terpenoids in planta are still not fully elucidated as well as the biosynthesis pathways involved in their production and the mechanisms of localization and secretion. Cannflavins accumulation also varies depending on the part of the plant, they are found in most parts, including the leaves and inflorescences, but are undetectable in roots and seeds (). Interestingly, all three cannflavins A-C were found in greater amounts in genetically identical Cannabis plants grown at a higher altitude ().

Importantly, the composition and concentration of the different secondary metabolites are also affected by harvest time () and change over time postharvest as a result of different degradation routes, depending on the storage conditions and its duration (). The concentrations of terpenoids rapidly decline in storage due to their volatile nature (). For phytocannabinoids, one of the main processes that occur during storage is decarboxylation. Over time due to heat and light, the acidic forms undergo spontaneous decarboxylation, but the extent of which is not uniform. For example, THC is the neutral counterpart of THCA. However, THCA is only partially converted to THC and to varying degrees (). THCA has different biological characteristics than THC, it is not psychoactive and has a distinctive pharmacological activity (). Several studies reported on the therapeutic activities of phytocannabinoids in their acidic form. For example, CBDA was found to be a more potent antiemetic and anticonvulsant agent than CBD in-vivo (), as well as a better inhibitor of breast cancer cell migration in-vitro (). Therefore, the relative ratio between THCA and THC, or between CBDA and CBD, has a therapeutic implication that has yet to be fully elucidated. For phytocannabinoids, the content of CBN is used as a marker for Cannabis aging, however, it is not a relevant marker in Type III chemovars () as it is formed mainly via the oxidation of THC or the decarboxylation of its acidic form cannabinolic acid (CBNA), which in turn rises from the oxidation of THCA.

In a study that tested the optimal postharvest processing, solvents and a range of temperatures, it was concluded that the conditions that best preserved the composition of the secondary metabolites relative to their pre-storage composition were unextracted whole inflorescences at 4°C (). The duration of storage, as well as of drying and curing before storage, varies greatly; as a consequence, a very large variation exists in the phytocannabinoid and terpenoid profiles of Cannabis chemovars that are considered the same.

Delivery Routes

As the active biomolecules in Cannabis such as phytocannabinoids are highly lipophilic and therefore present poor oral bioavailability, various administration routes have been investigated for the therapeutic use of Cannabis, including the pulmonary, sublingual, oral, dermal and rectal routes (). Currently, the common administration routes of whole-plant and plant-derived Cannabis products are either by inhalation (smoking or vaporization) or ingestion of edibles (). However, the pharmacokinetics and the effects observed with Cannabis administration vary significantly as a function of the delivery route, formulation, and the ratios between the multiple active compounds. For example, the acidic pH of the stomach further reduces bioavailability via the oral route (). Moreover, to be used via the oral or sublingual routes, the active secondary metabolites in the plant must be extracted. The extraction method and choice of extracting solvent affect the secondary metabolite profile (), a phenomenon which was shown for phytocannabinoids (), terpenoids () and flavonoids ().

Inhalation provides a rapid and efficient method of drug delivery. Symptom relief is immediate and effective, the dosage can be more controlled than via the alternative routes, and a lower dose can be used to get the desired effect (). However, inhalation has several considerable disadvantages; it leads to high and prompt peak plasma concentration of cannabinoids such as THC and CBD post inhalation (), causing a more intense and shorter-lasting effect than other routes, which in turn may be associated with higher toxicity (). Smoking is associated with health risks and the formation of toxic and carcinogenic substances during combustion (), vaporizers do not heat Cannabis to the point of combustion (i.e., less than 170–190°C), but still induce heat and expose to a variety of undesirable chemicals (). All the bioactive molecules of Cannabis are susceptible to degradation processes such as decarboxylation when Cannabis is heated above 120°C by smoking or vaping, as well as by cooking ().

The pharmacokinetics of the current consumption options modulates and limits the therapeutic bioavailability of Cannabis metabolites. For example, when THC is ingested rather than inhaled, it is metabolized by the liver before entering the bloodstream and hydroxylated to 11-hydroxy-THC, which is equally potent () or might be even more potent than THC (), and then further oxidized to the inactive metabolite 11-COOH-THC. This makes the consideration of the Cannabis delivery system vital for its effective administration and treatment ().

New analytical approaches now allow for more accurate profiling of Cannabis metabolites both in the plant itself and in the tissues they affect, allowing to better investigate their disposition over time by the body of the organism (). Many of the alternative routes to inhalation and digestion are aimed at improving the bioavailability via avoiding degradation with first-pass metabolism by the liver. Other delivery routes that have yet to be explored are intravenous, intramuscular and intranasal. Emulsions via nanotechnology advances are also aimed at improving the bioavailability of the active molecules in Cannabis ().

Discussion and Future Perspectives

The use of medical Cannabis is ever increasing in the treatment of numerous conditions as it has been proven to be both effective and safe, but the Cannabis plant contains more than 500 different components, each with potential therapeutic qualities. The components of Cannabis act together, hitting several targets at once and mutually enhancing each other’s activity so that the overall outcome is greater than that of their additive effect. The concentrations and combinations of the various secondary metabolites, including the way they complement each other, determine both the final medicinal response and adverse effects.

Cannabis can treat a multitude of very different conditions as it exerts its effects via the ECS, which is involved in many physiological processes. Cannabis treatment can be personalized to both the condition and the person to improve treatment outcomes while also reducing the drug load and minimizing the adverse effects. Most patients do not receive Cannabis-based medication but rather whole plants or extracts that contain many active bio-compounds in different proportions. Each has a different profile of components, undergoing different drug interactions. It is still unknown which molecules in the whole extract are responsible for its overall effect and via which ECS receptors, effectors and metabolic pathways. Further research is needed to find which whole extracts or specific molecules are best suited to treat a given condition.

Physicians and patients require more information to guide them in choosing the most appropriate cultivar or molecules, in the correct dose and via the optimal delivery route. The number of studies that tested different cannabinoids or tried to recognize the specific bioactive molecules from whole extracts is low and should be addressed to fulfill the full potential of Cannabis and improve human health.

Acknowledgments

We thank Almog Uziel for comments on the manuscript.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

  • Abraham A. D., Leung E. J. Y., Wong B. A., Rivera Z. M. G., Kruse L. C., Clark J. J., et al. (2020). Orally Consumed Cannabinoids Provide Long-Lasting Relief of Allodynia in a Mouse Model of Chronic Neuropathic PainNeuropsychopharmacology 45, 1105–1114. 10.1038/s41386-019-0585-3 [PMC free article] [PubMed] [CrossRef[]
  • Adams R., Hunt M., Clark J. H. (1940). Structure of Cannabidiol, a Product Isolated from the Marihuana Extract of Minnesota Wild Hemp. IJ. Am. Chem. Soc. 62, 196–200. 10.1021/ja01858a058 [CrossRef[]
  • Adusumilli N. C., Hazuka E. L., Friedman A. J. (2021). Nanotechnology to Deliver Cannabinoids in DermatologyPrecis. Nanomedicine 4, 787–794. 10.33218/001c.24597 [CrossRef[]
  • Aizpurua-Olaizola O., Soydaner U., Öztürk E., Schibano D., Simsir Y., Navarro P., et al. (2016). Evolution of the Cannabinoid and Terpene Content during the Growth of Cannabis Sativa Plants from Different ChemotypesJ. Nat. Prod. 79, 324–331. 10.1021/acs.jnatprod.5b00949 [PubMed] [CrossRef[]
  • Anderson L. L., Low I. K., Banister S. D., McGregor I. S., Arnold J. C. (2019). Pharmacokinetics of Phytocannabinoid Acids and Anticonvulsant Effect of Cannabidiolic Acid in a Mouse Model of Dravet SyndromeJ. Nat. Prod. 82, 3047–3055. 10.1021/acs.jnatprod.9b00600 [PubMed] [CrossRef[]
  • Andre C. M., Hausman J. F., Guerriero G. (2016). Cannabis Sativa: The Plant of the Thousand and One MoleculesFront. Plant Sci. 7, 19. 10.3389/fpls.2016.00019 [PMC free article] [PubMed] [CrossRef[]
  • Aviram J., Lewitus G. M., Vysotski Y., Uribayev A., Procaccia S., Cohen I., et al. (2020a). Short-term Medical Cannabis Treatment Regimens Produced Beneficial Effects Among Palliative Cancer PatientsPharmaceuticals (Basel) 13, 1–16. 10.3390/ph13120435 [CrossRef[]
  • Aviram J., Vysotski Y., Berman P., Lewitus G. M., Eisenberg E., Meiri D. (2020b). Migraine Frequency Decrease Following Prolonged Medical Cannabis Treatment: A Cross-Sectional StudyBrain Sci. 10, 360. 10.3390/brainsci10060360 [CrossRef[]
  • Aviram J., Lewitus G. M., Pud D., Procaccia S., Berman P., Yellin B., et al. (2021a). Specific Phytocannabinoid Compositions Are Associated with Analgesic Response and Adverse Effects in Chronic Pain Patients Treated with Medical CannabisPharmacol. Res. 169, 105651. 10.1016/j.phrs.2021.105651 [PubMed] [CrossRef[]
  • Aviram J., Pud D., Gershoni T., Schiff‐Keren B., Ogintz M., Vulfsons S., et al. (2021b). Medical Cannabis Treatment for Chronic Pain: Outcomes and Prediction of ResponseEur. J. Pain 25, 359–374. 10.1002/ejp.1675 [PubMed] [CrossRef[]
  • Baram L., Peled E., Berman P., Yellin B., Besser E., Benami M., et al. (2019). The Heterogeneity and Complexity of Cannabis Extracts as Antitumor AgentsOncotarget 10, 4091–4106. 10.18632/oncotarget.26983 [PMC free article] [PubMed] [CrossRef[]
  • Bautista J. L., Yu S., Tian L. (2021). Flavonoids in Cannabis Sativa: Biosynthesis, Bioactivities, and BiotechnologyACS Omega 6, 5119–5123. 10.1021/acsomega.1c00318 [PMC free article] [PubMed] [CrossRef[]
  • Belardo C., Iannotta M., Boccella S., Rubino R. C., Ricciardi F., Infantino R., et al. (2019). Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain InjuryFront. Pharmacol. 10, 352–411. 10.3389/fphar.2019.00352 [PMC free article] [PubMed] [CrossRef[]
  • Ben-Shabat S., Fride E., Sheskin T., Tamiri T., Rhee M. H., Vogel Z., et al. (1998). An Entourage Effect: Inactive Endogenous Fatty Acid Glycerol Esters Enhance 2-Arachidonoyl-Glycerol Cannabinoid ActivityEur. J. Pharmacol. 353, 23–31. 10.1016/S0014-2999(98)00392-6 [PubMed] [CrossRef[]
  • Berman P., Futoran K., Lewitus G. M., Mukha D., Benami M., Shlomi T., et al. (2018). A New ESI-LC/MS Approach for Comprehensive Metabolic Profiling of Phytocannabinoids in CannabisSci. Rep. 8, 14280–14315. 10.1038/s41598-018-32651-4 [PMC free article] [PubMed] [CrossRef[]
  • Berman P., Sulimani L., Gelfand A., Amsalem K., Lewitus G. M., Meiri D. (2020). Cannabinoidomics – An Analytical Approach to Understand the Effect of Medical Cannabis Treatment on the Endocannabinoid MetabolomeTalanta 219, 121336. 10.1016/j.talanta.2020.121336 [PubMed] [CrossRef[]
  • Bernstein N., Gorelick J., Koch S. (2019a). Interplay between Chemistry and Morphology in Medical Cannabis (Cannabis Sativa L.)Ind. Crops Prod. 129, 185–194. 10.1016/j.indcrop.2018.11.039 [CrossRef[]
  • Bernstein N., Gorelick J., Zerahia R., Koch S. (2019b). Impact of N, P, K, and Humic Acid Supplementation on the Chemical Profile of Medical Cannabis (Cannabis Sativa L)Front. Plant Sci. 10, 736. 10.3389/fpls.2019.00736 [PMC free article] [PubMed] [CrossRef[]
  • Blasco-Benito S., Seijo-Vila M., Caro-Villalobos M., Tundidor I., Andradas C., García-Taboada E., et al. (2018). Appraising the “entourage Effect”: Antitumor Action of a Pure Cannabinoid versus a Botanical Drug Preparation in Preclinical Models of Breast CancerBiochem. Pharmacol. 157, 285–293. 10.1016/j.bcp.2018.06.025 [PubMed] [CrossRef[]
  • Bolognini D., Rock E. M., Cluny N. L., Cascio M. G., Limebeer C. L., Duncan M., et al. (2013). Cannabidiolic Acid Prevents Vomiting in Suncus Murinus and Nausea-Induced Behaviour in Rats by Enhancing 5-HT1A Receptor ActivationBr. J. Pharmacol. 168, 1456–1470. 10.1111/bph.12043 [PMC free article] [PubMed] [CrossRef[]
  • Booth J. K., Yuen M. M. S., Jancsik S., Madilao L. L., Page J. E., Bohlmann J. (2020). Terpene Synthases and Terpene Variation in Cannabis SativaPlant Physiol. 184, 130–147. 10.1104/PP.20.00593 [PMC free article] [PubMed] [CrossRef[]
  • Bridgeman M. B., Abazia D. T. (2017). Medicinal Cannabis: History, Pharmacology, and Implications for the Acute Care SettingP T 42, 180–188. [PMC free article] [PubMed[]
  • Bruni N., Della Pepa C., Oliaro-Bosso S., Pessione E., Gastaldi D., Dosio F. (2018). Cannabinoid Delivery Systems for Pain and Inflammation TreatmentMolecules 23, 2478. 10.3390/molecules23102478 [CrossRef[]
  • Calzolari D., Magagnini G., Lucini L., Grassi G., Appendino G. B., Amaducci S. (2017). High Added-Value Compounds from Cannabis Threshing ResiduesInd. Crops Prod. 108, 558–563. 10.1016/j.indcrop.2017.06.063 [CrossRef[]
  • Casano S., Grassi G., Martini V., Michelozzi M. (2011). Variations in Terpene Profiles of Different Strains of Cannabis Sativa LActa Hortic. 925, 115–121. 10.17660/actahortic.2011.925.15 [CrossRef[]
  • Casey S. L., Atwal N., Vaughan C. W. (2017). Cannabis Constituent Synergy in a Mouse Neuropathic Pain ModelPain 158, 2452–2460. 10.1097/j.pain.0000000000001051 [PubMed] [CrossRef[]
  • Chandra S., Lata H., Khan I. A., Elsohly M. A. (2008). Photosynthetic Response of Cannabis Sativa L. To Variations in Photosynthetic Photon Flux Densities, Temperature and CO2 ConditionsPhysiol. Mol. Biol. Plants 14, 299–306. 10.1007/s12298-008-0027-x [PMC free article] [PubMed] [CrossRef[]
  • Chandra S., Lata H., Khan I. A., ElSohly M. A. (2017). “Cannabis Sativa L.: Botany and Horticulture,” in Cannabis Sativa L.-Botany and Biotechnology (Cham: Springer; ), 79–100. 10.1007/978-3-319-54564-6_3 [CrossRef[]
  • Christensen H. D., Freudenthal R. I., Gidley J. T., Rosenfeld R., Boegli G., Testino L., et al. (1971). Activity of delta8- and delta9-tetrahydrocannabinol and Related Compounds in the MouseScience 172, 165–167. 10.1126/science.172.3979.165 [PubMed] [CrossRef[]
  • Citti C., Linciano P., Panseri S., Vezzalini F., Forni F., Vandelli M. A., et al. (2019a). Cannabinoid Profiling of Hemp Seed Oil by Liquid Chromatography Coupled to High-Resolution Mass SpectrometryFront. Plant Sci. 10, 120–217. 10.3389/fpls.2019.00120 [PMC free article] [PubMed] [CrossRef[]
  • Citti C., Linciano P., Russo F., Luongo L., Iannotta M., Maione S., et al. (2019b). A Novel Phytocannabinoid Isolated from Cannabis Sativa L. With an In Vivo Cannabimimetic Activity Higher Than Δ9-tetrahydrocannabinol: Δ9-TetrahydrocannabiphorolSci. Rep. 9, 20335–20413. 10.1038/s41598-019-56785-1 [PMC free article] [PubMed] [CrossRef[]
  • Comelli F., Giagnoni G., Bettoni I., Colleoni M., Costa B. (2008). Antihyperalgesic Effect of a Cannabis Sativa Extract in a Rat Model of Neuropathic Pain: Mechanisms InvolvedPhytother Res. 22, 1017–1024. 10.1002/ptr.2401 [PubMed] [CrossRef[]
  • Consroe P., Benedito M. A., Leite J. R., Carlini E. A., Mechoulam R. (1982). Effects of Cannabidiol on Behavioral Seizures Caused by Convulsant Drugs or Current in MiceEur. J. Pharmacol. 83, 293–298. 10.1016/0014-2999(82)90264-3 [PubMed] [CrossRef[]
  • De Backer B., Debrus B., Lebrun P., Theunis L., Dubois N., Decock L., et al. (2009). Innovative Development and Validation of an HPLC/DAD Method for the Qualitative and Quantitative Determination of Major Cannabinoids in Cannabis Plant MaterialJ. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 877, 4115–4124. 10.1016/j.jchromb.2009.11.004 [CrossRef[]
  • de Fonseca F. R., del Arco I., Bermudez-Silva F. J., Bilbao A., Cippitelli A., Navarro M. (2005). The Endocannabinoid System: Physiology and PharmacologyAlcohol Alcohol 40, 2–14. 10.1093/alcalc/agh110 [PubMed] [CrossRef[]
  • De Meijer E. P., Bagatta M., Carboni A., Crucitti P., Moliterni V. M., Ranalli P., et al. (2003). The Inheritance of Chemical Phenotype in Cannabis Sativa LGenetics 163, 335–346. 10.1093/genetics/163.1.335 [PMC free article] [PubMed] [CrossRef[]
  • De Petrocellis L., Di Marzo V. (2009). An Introduction to the Endocannabinoid System: from the Early to the Latest ConceptsBest Pract. Res. Clin. Endocrinol. Metab. 23, 1–15. 10.1016/j.beem.2008.10.013 [PubMed] [CrossRef[]
  • De Petrocellis L., Cascio M. G., Di Marzo V. (2004). The Endocannabinoid System: A General View and Latest AdditionsBr. J. Pharmacol. 141, 765–774. 10.1038/sj.bjp.0705666 [PMC free article] [PubMed] [CrossRef[]
  • Delgado-Povedano M. M., Sánchez-Carnerero Callado C., Priego-Capote F., Ferreiro-Vera C. (2019). Untargeted Characterization of Extracts from Cannabis Sativa L. Cultivars by Gas and Liquid Chromatography Coupled to Mass Spectrometry in High Resolution ModeTalanta 208, 120384. 10.1016/j.talanta.2019.120384 [PubMed] [CrossRef[]
  • Devane W. A., Hanus L., Breuer A., Pertwee R. G., Stevenson L. A., Griffin G., et al. (1992). Isolation and Structure of a Brain Constituent that Binds to the Cannabinoid ReceptorScience 258, 1946–1949. 10.1126/science.1470919 [PubMed] [CrossRef[]
  • Di Marzo V., Bifulco M., De Petrocellis L. (2004). The Endocannabinoid System and its Therapeutic ExploitationNat. Rev. Drug Discov. 3, 771–784. 10.1038/nrd1495 [PubMed] [CrossRef[]
  • Dinis-oliveira R. J. (2016). Metabolomics of Δ9-tetrahydrocannabinol: Implications in ToxicityDrug Metab. Rev. 48, 80–87. 10.3109/03602532.2015.1137307 [PubMed] [CrossRef[]
  • Downer E. J. (2020). Anti-inflammatory Potential of Terpenes Present in Cannabis Sativa LACS Chem. Neurosci. 11, 659–662. 10.1021/acschemneuro.0c00075 [PubMed] [CrossRef[]
  • Dussy F. E., Hamberg C., Luginbühl M., Schwerzmann T., Briellmann T. A. (2005). Isolation of Delta9-THCA-A from Hemp and Analytical Aspects Concerning the Determination of Delta9-THC in Cannabis ProductsForensic Sci. Int. 149, 3–10. 10.1016/j.forsciint.2004.05.015 [PubMed] [CrossRef[]
  • ElSohly M. A., Slade D. (2005). Chemical Constituents of Marijuana: the Complex Mixture of Natural CannabinoidsLife Sci. 78, 539–548. 10.1016/j.lfs.2005.09.011 [PubMed] [CrossRef[]
  • Erridge S., Mangal N., Salazar O., Pacchetti B., Sodergren M. H. (2020). Cannflavins – from Plant to Patient: A Scoping ReviewFitoterapia 146, 104712. 10.1016/j.fitote.2020.104712 [PubMed] [CrossRef[]
  • Ferber S. G., Namdar D., Hen-Shoval D., Eger G., Koltai H., Shoval G., et al. (2020). The “Entourage Effect”: Terpenes Coupled with Cannabinoids for the Treatment of Mood Disorders and Anxiety DisordersCurr. Neuropharmacol. 18, 87–96. 10.2174/1570159X17666190903103923 [PMC free article] [PubMed] [CrossRef[]
  • Finlay D. B., Sircombe K. J., Nimick M., Jones C., Glass M. (2020). Terpenoids from Cannabis Do Not Mediate an Entourage Effect by Acting at Cannabinoid ReceptorsFront. Pharmacol. 11, 359. 10.3389/fphar.2020.00359 [PMC free article] [PubMed] [CrossRef[]
  • Flores-Sanchez I. J., Verpoorte R. (2008a). PKS Activities and Biosynthesis of Cannabinoids and Flavonoids in Cannabis Sativa L. PlantsPlant Cell Physiol. 49, 1767–1782. 10.1093/pcp/pcn150 [PubMed] [CrossRef[]
  • Flores-Sanchez I. J., Verpoorte R. (2008b). Secondary Metabolism in CannabisPhytochem. Rev. 7, 615–639. 10.1007/s11101-008-9094-4 [CrossRef[]
  • Foster B. C., Abramovici H., Harris C. S. (2019). Cannabis and Cannabinoids: Kinetics and InteractionsAm. J. Med. 132, 1266–1270. 10.1016/j.amjmed.2019.05.017 [PubMed] [CrossRef[]
  • Gaoni Y., Mechoulam R. (1964). Isolation, Structure, and Partial Synthesis of an Active Constituent of HashishJ. Am. Chem. Soc. 86, 1646–1647. 10.1021/ja01062a046 [CrossRef[]
  • Gates P., Jaffe A., Copeland J. (2014). Cannabis Smoking and Respiratory Health: Consideration of the LiteratureRespirology 19, 655–662. 10.1111/resp.12298 [PubMed] [CrossRef[]
  • Gershenzon J., Dudareva N. (2007). The Function of Terpene Natural Products in the Natural WorldNat. Chem. Biol. 3, 408–414. 10.1038/nchembio.2007.5 [PubMed] [CrossRef[]
  • Gertsch J., Pertwee R. G., Di Marzo V. (2010). Phytocannabinoids beyond the Cannabis Plant – Do They Exist? Br. J. Pharmacol. 160, 523–529. 10.1111/j.1476-5381.2010.00745.x [PMC free article] [PubMed] [CrossRef[]
  • Giupponi L., Leoni V., Pavlovic R., Giorgi A. (2020). Influence of Altitude on Phytochemical Composition of Hemp Inflorescence: A Metabolomic ApproachMolecules 25, 1381. 10.3390/molecules25061381 [CrossRef[]
  • Grotenhermen F. (2003). Pharmacokinetics and Pharmacodynamics of CannabinoidsClin. Pharmacokinet. 42, 327–360. 10.2165/00003088-200342040-00003 [PubMed] [CrossRef[]
  • Gülck T., Møller B. L. (2020). Phytocannabinoids: Origins and BiosynthesisTrends Plant Sci. 25, 985–1004. 10.1016/j.tplants.2020.05.005 [PubMed] [CrossRef[]
  • Hand A., Blake A., Kerrigan P., Samuel P. (2016). History of Medical CannabisJ. Pain Manag. 9, 387–394. []
  • Hanuš L. O., Hod Y. (2020). Terpenes/Terpenoids in Cannabis: Are They Important? Med. Cannabis Cannabinoids 3, 25–60. 10.1159/000509733 [PMC free article] [PubMed] [CrossRef[]
  • Hanuš L. O., Meyer S. M., Muñoz E., Taglialatela-Scafati O., Appendino G. (2016). Phytocannabinoids: A Unified Critical InventoryNat. Prod. Rep. 33, 1357–1392. 10.1039/c6np00074f [PubMed] [CrossRef[]
  • Happyana N., Kayser O. (2016). Monitoring Metabolite Profiles of Cannabis Sativa L. Trichomes during Flowering Period Using 1H NMR-Based Metabolomics and Real-Time PCRPlanta Med. 82, 1217–1223. 10.1055/s-0042-108058 [PubMed] [CrossRef[]
  • Happyana N., Agnolet S., Muntendam R., Van Dam A., Schneider B., Kayser O. (2013). Analysis of Cannabinoids in Laser-Microdissected Trichomes of Medicinal Cannabis Sativa Using LCMS and Cryogenic NMRPhytochemistry 87, 51–59. 10.1016/j.phytochem.2012.11.001 [PubMed] [CrossRef[]
  • Hazekamp A., Fischedick J. T. (2012). Cannabis – from Cultivar to ChemovarDrug Test. Anal. 4, 660–667. 10.1002/dta.407 [PubMed] [CrossRef[]
  • Hazekamp A., Ware M. A., Muller-Vahl K. R., Abrams D., Grotenhermen F. (2013). The Medicinal Use of Cannabis and Cannabinoids-Aan International Cross-Sectional Survey on Administration FormsJ. Psychoactive Drugs 45, 199–210. 10.1080/02791072.2013.805976 [PubMed] [CrossRef[]
  • Heblinski M., Santiago M., Fletcher C., Stuart J., Connor M., Mcgregor I. S., et al. (2020). Terpenoids Commonly Found in Cannabis Sativa Do Not Modulate the Actions of Phytocannabinoids or Endocannabinoids on TRPA1 and TRPV1 ChannelsCannabis Cannabinoid Res. 5, 305–317. 10.1089/can.2019.0099 [PMC free article] [PubMed] [CrossRef[]
  • Henry P., Hilyard A., Johnson S., Orser C. (2018). Predicting Chemovar Cluster and Variety Verification in Vegetative Cannabis Accessions Using Targeted Single Nucleotide PolymorphismsPeerJ Prepr 6, e27442v1. 10.7287/peerj.preprints.27442v1 [CrossRef[]
  • Hillig K. W. (2004). A Chemotaxonomic Analysis of Terpenoid Variation in CannabisBiochem. Syst. Ecol. 32, 875–891. 10.1016/j.bse.2004.04.004 [CrossRef[]
  • Holgado M. A., Martín-Banderas L., Álvarez-Fuentes J., Fernández-Arévalo M. (2017). Neuroprotective Effect of Cannabinoids Nanoplatforms in Neurodegenerative DiseasesJ. Drug Deliv. Sci. Technol. 42, 84–93. 10.1016/j.jddst.2017.04.023 [CrossRef[]
  • Hollister L. E. (1974). Structure-activity Relationships in Man of Cannabis Constituents, and Homologs and Metabolites of delta9-tetrahydrocannabinolPharmacology 11, 3–11. 10.1159/000136462 [PubMed] [CrossRef[]
  • Huestis M. A. (2005). “Pharmacokinetics and Metabolism of the Plant Cannabinoids, Δ 9-tetrahydrocannibinol, Cannabidiol and Cannabinol,” in Cannabinoids. Editor Pertwee R. G. (Berlin, Heidelberg: Springer; ), 657–690. []
  • Huestis M. A. (2007). Human Cannabinoid PharmacokineticsChem. Biodivers. 4, 1770–1804. 10.1002/cbdv.200790152 [PMC free article] [PubMed] [CrossRef[]
  • Isidore E., Karim H., Ioannou I. (2021). Extraction of Phenolic Compounds and Terpenes from Cannabis Sativa L. By-Products: From Conventional to Intensified ProcessesAntioxidants (Basel) 10, 942. 10.3390/antiox10060942 [PMC free article] [PubMed] [CrossRef[]
  • Jin D., Jin S., Chen J. (2019). Cannabis Indoor Growing Conditions, Management Practices, and Post-Harvest Treatment: A ReviewAm. J. Plant Sci. 10, 925–946. 10.4236/ajps.2019.106067 [CrossRef[]
  • Jung J., Meyer M. R., Maurer H. H., Neusüß C., Weinmann W., Auwärter V. (2009). Studies on the Metabolism of the Δ9-tetrahydrocannabinol Precursor Δ9-tetrahydrocannabinolic Acid A (Δ9-THCA-A) in Rat Using LC-MS/MS, LC-QTOF MS and GC-MS TechniquesJ. Mass. Spectrom. 44, 1423–1433. 10.1002/jms.1624 [PubMed] [CrossRef[]
  • King K. M., Myers A. M., Soroka-Monzo A. J., Tuma R. F., Tallarida R. J., Walker E. A., et al. (2017). Single and Combined Effects of Δ9 -tetrahydrocannabinol and Cannabidiol in a Mouse Model of Chemotherapy-Induced Neuropathic PainBr. J. Pharmacol. 174, 2832–2841. 10.1111/bph.13887 [PMC free article] [PubMed] [CrossRef[]
  • Křížek T., Bursová M., Horsley R., Kuchař M., Tůma P., Čabala R., et al. (2018). Menthol-based Hydrophobic Deep Eutectic Solvents: Towards Greener and Efficient Extraction of PhytocannabinoidsJ. Clean. Prod. 193, 391–396. 10.1016/j.jclepro.2018.05.080 [CrossRef[]
  • Lewis M. A., Russo E. B., Smith K. M. (2018). Pharmacological Foundations of Cannabis ChemovarsPlanta Med. 84, 225–233. 10.1055/s-0043-122240 [PubMed] [CrossRef[]
  • Linciano P., Citti C., Russo F., Tolomeo F., Laganà A., Capriotti A. L., et al. (2020). Identification of a New Cannabidiol N-Hexyl Homolog in a Medicinal Cannabis Variety with an Antinociceptive Activity in Mice: CannabidihexolSci. Rep. 10, 22019–22111. 10.1038/s41598-020-79042-2 [PMC free article] [PubMed] [CrossRef[]
  • Lipson Feder C., Cohen O., Shapira A., Katzir I., Peer R., Guberman O., et al. (2021). Fertilization Following Pollination Predominantly Decreases Phytocannabinoids Accumulation and Alters the Accumulation of Terpenoids in Cannabis InflorescencesFront. Plant Sci. 2426, 753847. 10.3389/fpls.2021.753847 [CrossRef[]
  • Lu H. C., Mackie K. (2016). An Introduction to the Endogenous Cannabinoid SystemBiol. Psychiatry 79, 516–525. 10.1016/j.biopsych.2015.07.028 [PMC free article] [PubMed] [CrossRef[]
  • Lunardi P., de Souza L. W., dos Santos B., Popik B., de Oliveira Alvares L. (2020). Effect of the Endocannabinoid System in Memory Updating and ForgettingNeuroscience 444, 33–42. 10.1016/j.neuroscience.2020.07.045 [PubMed] [CrossRef[]
  • Maayah Z. H., Takahara S., Ferdaoussi M., Dyck J. R. B. (2020a). The Anti-inflammatory and Analgesic Effects of Formulated Full-Spectrum Cannabis Extract in the Treatment of Neuropathic Pain Associated with Multiple SclerosisInflamm. Res. 69, 549–558. 10.1007/s00011-020-01341-1 [PubMed] [CrossRef[]
  • Maayah Z. H., Takahara S., Ferdaoussi M., Dyck J. R. B. (2020b). The Molecular Mechanisms that Underpin the Biological Benefits of Full-Spectrum Cannabis Extract in the Treatment of Neuropathic Pain and InflammationBiochim. Biophys. Acta Mol. Basis Dis. 1866, 165771. 10.1016/j.bbadis.2020.165771 [PubMed] [CrossRef[]
  • Maccarrone M. (2020). Phytocannabinoids and Endocannabinoids: Different in NatureRend. Fis. Acc. Lincei 31, 931–938. 10.1007/s12210-020-00957-z [CrossRef[]
  • Mackie K. (2008). Cannabinoid Receptors: Where They Are and what They DoJ. Neuroendocrinol. 20, 10–14. 10.1111/j.1365-2826.2008.01671.x [PubMed] [CrossRef[]
  • Maroso M., Szabo G. G., Kim H. K., Alexander A., Bui A. D., Lee S. H., et al. (2016). Cannabinoid Control of Learning and Memory through HCN ChannelsNeuron 89, 1059–1073. 10.1016/j.neuron.2016.01.023 [PMC free article] [PubMed] [CrossRef[]
  • Marsicano G., Lafenêtre P. (2009). “Roles of the Endocannabinoid System in Learning and Memory,” in Behavioral Neurobiology of the Endocannabinoid System (Berlin, Heidelberg: Springer; ), Vol. 1, 201–230. 10.1007/978-3-540-88955-7_8 [CrossRef[]
  • Matsuda L. A., Lolait S. J., Brownstein M. J., Young A. C., Bonner T. I. (1990). Structure of a Cannabinoid Receptor and Functional Expression of the Cloned cDNANature 346, 561–564. 10.1038/346561a0 [PubMed] [CrossRef[]
  • McGarvey P., Huang J., McCoy M., Orvis J., Katsir Y., Lotringer N., et al. (2020). De Novo assembly and Annotation of Transcriptomes from Two Cultivars of Cannabis Sativa with Different Cannabinoid ProfilesGene 762, 145026. 10.1016/j.gene.2020.145026 [PubMed] [CrossRef[]
  • Mechoulam R., Shvo Y. (1963). Hashish. I. The Structure of CannabidiolTetrahedron 19, 2073–2078. 10.1016/0040-4020(63)85022-x [PubMed] [CrossRef[]
  • Mechoulam R., Ben-Shabat S., Hanus L., Ligumsky M., Kaminski N. E., Schatz A. R., et al. (1995). Identification of an Endogenous 2-monoglyceride, Present in Canine Gut, that Binds to Cannabinoid ReceptorsBiochem. Pharmacol. 50, 83–90. 10.1016/0006-2952(95)00109-d [PubMed] [CrossRef[]
  • Mechoulam R., Peters M., Murillo-Rodriguez E., Hanus L. O. (2007). Cannabidiol–recent AdvancesChem. Biodivers. 4, 1678–1692. 10.1002/cbdv.200790147 [PubMed] [CrossRef[]
  • Milay L., Berman P., Shapira A., Guberman O., Meiri D. (2020). Metabolic Profiling of Cannabis Secondary Metabolites for Evaluation of Optimal Postharvest Storage ConditionsFront. Plant Sci. 11, 583605–583615. 10.3389/fpls.2020.583605 [PMC free article] [PubMed] [CrossRef[]
  • Montecucco F., Di Marzo V. (2012). At the Heart of the Matter: The Endocannabinoid System in Cardiovascular Function and DysfunctionTrends Pharmacol. Sci. 33, 331–340. 10.1016/j.tips.2012.03.002 [PubMed] [CrossRef[]
  • Moreno-Sanz G. (2016). Can You Pass the Acid Test? Critical Review and Novel Therapeutic Perspectives of Δ9-Tetrahydrocannabinolic Acid ACannabis Cannabinoid Res. 1, 124–130. 10.1089/can.2016.0008 [PMC free article] [PubMed] [CrossRef[]
  • Munro S., Thomas K. L., Abu-Shaar M. (1993). Molecular Characterization of a Peripheral Receptor for CannabinoidsNature 365, 61–65. 10.1038/365061a0 [PubMed] [CrossRef[]
  • Namdar D., Voet H., Ajjampura V., Nadarajan S., Mayzlish-Gati E., Mazuz M., et al. (2019). Terpenoids and Phytocannabinoids Co-produced in Cannabis Sativa Strains Show Specific Interaction for Cell Cytotoxic ActivityMolecules 24, 3031. 10.3390/molecules24173031 [CrossRef[]
  • Onaivi E. S., Ishiguro H., Gong J. P., Patel S., Perchuk A., Meozzi P. A., et al. (2006). Discovery of the Presence and Functional Expression of Cannabinoid CB2 Receptors in BrainAnn. N. Y. Acad. Sci. 1074, 514–536. 10.1196/annals.1369.052 [PubMed] [CrossRef[]
  • Pacher P., Steffens S. (2009). The Emerging Role of the Endocannabinoid System in Cardiovascular DiseaseSemin. Immunopathol. 31, 63–77. 10.1007/s00281-009-0145-8 [PMC free article] [PubMed] [CrossRef[]
  • Pandey R., Mousawy K., Nagarkatti M., Nagarkatti P. (2009). Endocannabinoids and Immune RegulationPharmacol. Res. 60, 85–92. 10.1016/j.phrs.2009.03.019 [PMC free article] [PubMed] [CrossRef[]
  • Perez-Reyes M., Timmons M. C., Lipton M. A., Davis K. H., Wall M. E. (1972). Intravenous Injection in Man of 9 -tetrahydrocannabinol and 11-OH- 9 -tetrahydrocannabinolScience 177, 633–635. 10.1126/science.177.4049.633 [PubMed] [CrossRef[]
  • Piper B. J. (2018). Mother of Berries, ACDC, or Chocolope: Examination of the Strains Used by Medical Cannabis Patients in New EnglandJ. Psychoactive Drugs 50, 95–104. 10.1080/02791072.2017.1390179 [PMC free article] [PubMed] [CrossRef[]
  • Russo E. B., McPartland J. M., ElSohly M. A., Wachtel S. R., Wit H. (2003). Cannabis Is More Than Simply delta(9)-tetrahydrocannabinolPsychopharmacology (Berl) 165, 431–434. 10.1007/s00213-002-1348-z [PubMed] [CrossRef[]
  • Russo E. B. (2011). Taming THC: Potential Cannabis Synergy and Phytocannabinoid-Terpenoid Entourage EffectsBr. J. Pharmacol. 163, 1344–1364. 10.1111/j.1476-5381.2011.01238.x [PMC free article] [PubMed] [CrossRef[]
  • Russo E. B. (2016). Beyond Cannabis: Plants and the Endocannabinoid SystemTrends Pharmacol. Sci. 37, 594–605. 10.1016/j.tips.2016.04.005 [PubMed] [CrossRef[]
  • Santiago M., Sachdev S., Arnold J. C., Mcgregor I. S., Connor M. (2019). Absence of Entourage: Terpenoids Commonly Found in Cannabis Sativa Do Not Modulate the Functional Activity of Δ9-THC at Human CB1 and CB2 ReceptorsCannabis Cannabinoid Res. 4, 165–176. 10.1089/can.2019.0016 [PMC free article] [PubMed] [CrossRef[]
  • Schwilke E. W., Schwope D. M., Karschner E. L., Lowe R. H., Darwin W. D., Kelly D. L., et al. (2009). Delta9-tetrahydrocannabinol (THC), 11-Hydroxy-THC, and 11-Nor-9-Carboxy-THC Plasma Pharmacokinetics during and after Continuous High-Dose Oral THCClin. Chem. 55, 2180–2189. 10.1373/clinchem.2008.122119 [PMC free article] [PubMed] [CrossRef[]
  • Shapira A., Berman P., Futoran K., Guberman O., Meiri D. (2019). Tandem Mass Spectrometric Quantification of 93 Terpenoids in Cannabis Using Static Headspace InjectionsAnal. Chem. 91, 11425–11432. 10.1021/acs.analchem.9b02844 [PubMed] [CrossRef[]
  • Shiplo S., Asbridge M., Leatherdale S. T., Hammond D. (2016). Medical Cannabis Use in Canada: Vapourization and Modes of DeliveryHarm Reduct. J. 13, 30–10. 10.1186/s12954-016-0119-9 [PMC free article] [PubMed] [CrossRef[]
  • Takeda S., Okajima S., Miyoshi H., Yoshida K., Okamoto Y., Okada T., et al. (2012). Cannabidiolic Acid, a Major Cannabinoid in Fiber-type Cannabis, Is an Inhibitor of MDA-MB-231 Breast Cancer Cell MigrationToxicol. Lett. 214, 314–319. 10.1016/j.toxlet.2012.08.029 [PMC free article] [PubMed] [CrossRef[]
  • Tang K., Struik P. C., Yin X., Thouminot C., Bjelková M., Stramkale V., et al. (2016). Comparing Hemp (Cannabis Sativa L.) Cultivars for Dual-Purpose Production under Contrasting EnvironmentsInd. Crops Prod. 87, 33–44. 10.1016/j.indcrop.2016.04.026 [CrossRef[]
  • Trofin I. G., Vlad C. C., Dabija G., Filipescu L. (2011). Influence of Storage Conditions on the Chemical Potency of Herbal CannabisRev. Chim. 62, 639–645. []
  • Turner S. E., Williams C. M., Iversen L., Whalley B. J. (2017). “Molecular Pharmacology of Phytocannabinoids,” in Phytocannabinoids (Cham: Springer; ), 61–101. 10.1007/978-3-319-45541-9_3 [CrossRef[]
  • Uziel A., Gelfand A., Amsalem K., Berman P., Lewitus G. M., Meiri D., et al. (2020). Full-Spectrum Cannabis Extract Microdepots Support Controlled Release of Multiple Phytocannabinoids for Extended Therapeutic EffectACS Appl. Mater. Inter. 12, 23707–23716. 10.1021/acsami.0c04435 [CrossRef[]
  • Velasco G., Sánchez C., Guzmán M. (2016). Anticancer Mechanisms of CannabinoidsCurr. Oncol. 23, S23–S32. 10.3747/co.23.3080 [CrossRef[]
  • Welling M. T., Liu L., Raymond C. A., Ansari O., King G. J. (2018). Developmental Plasticity of the Major Alkyl Cannabinoid Chemotypes in a Diverse Cannabis Genetic Resource CollectionFront. Plant Sci. 9, 1510. 10.3389/fpls.2018.01510 [PMC free article] [PubMed] [CrossRef[]
  • Woodhams S. G., Sagar D. R., Burston J. J., Chapman V. (2015). The Role of the Endocannabinoid System in PainHandb Exp. Pharmacol. 227, 119–143. 10.1007/978-3-662-46450-2_7 [PubMed] [CrossRef[]
  • Woodhams S. G., Chapman V., Finn D. P., Hohmann A. G., Neugebauer V. (2017). The Cannabinoid System and PainNeuropharmacology 124, 105–120. 10.1016/j.neuropharm.2017.06.015 [PMC free article] [PubMed] [CrossRef[]
  • Zamengo L., Bettin C., Badocco D., Di Marco V., Miolo G., Frison G. (2019). The Role of Time and Storage Conditions on the Composition of Hashish and Marijuana Samples: a Four-Year StudyForensic Sci. Int. 298, 131–137. 10.1016/j.forsciint.2019.02.058 [PubMed] [CrossRef[]
  • Zuardi A. W. (2008). Cannabidiol: from an Inactive Cannabinoid to a Drug with Wide Spectrum of ActionBraz. J. Psychiatry 30, 271–280. 10.1590/s1516-44462008000300015 [PubMed] [CrossRef[]

Articles from Frontiers in Pharmacology are provided here courtesy of Frontiers Media SA


Leave a Reply