Next Article in Journal
Interferon Lambda: The Next Frontier in Antiviral Therapy?
Previous Article in Journal
Influence of a Zombie-like State of the Liver on Drugs and Its Medico-Legal Implications: A Scoping Review
Previous Article in Special Issue
Elderberry Leaves with Antioxidant and Anti-Inflammatory Properties as a Valuable Plant Material for Wound Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent HPLC-UV Approaches for Cannabinoid Analysis: From Extraction to Method Validation and Quantification Compliance

by
Eduarda M. P. Silva
1,2,
Antonella Vitiello
3,
Agnese Miro
3 and
Carlos J. A. Ribeiro
1,2,4,*
1
Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
2
UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
3
Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
4
Avextra Portugal SA, 7570-003 Grândola, Portugal
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(6), 786; https://doi.org/10.3390/ph18060786
Submission received: 30 April 2025 / Revised: 21 May 2025 / Accepted: 22 May 2025 / Published: 24 May 2025

Abstract

Since the 1990s, cannabis has experienced a gradual easing of access restrictions, accompanied by the expansion of its legalization and commercialization. This shift has led to the proliferation of cannabis-based products, available as cosmetics, food supplements, and pharmaceutical dosage forms. Consequently, there has been a growing demand for reliable and reproducible extraction techniques alongside precise analytical methods for detecting and quantifying cannabinoids, both of which are essential for ensuring consumer safety and product quality. Given the variability in extraction and quantification techniques across laboratories, significant attention has recently been directed toward method validation. Validated methods ensure precise cannabinoid measurement in cannabis-based products, supporting compliance with dosage guidelines and legal limits. Thus, this review highlights recent advancements in these areas, with a particular focus on High-Performance Liquid Chromatography (HPLC) coupled with Ultraviolet (UV) detection, as it is considered the gold standard for cannabinoid analysis included in cannabis monographs present in several pharmacopeias. The research focused on studies published between January 2022 and December 2024, sourced from PubMed, Scopus, and Web of Science, that employed an HPLC-UV analytical technique for the detection of phytocannabinoids. Additionally, the review examines cannabinoid extraction techniques and the validation methodologies used by the authors in the selected papers. Notably, ultrasound extraction has emerged as the most widely utilized technique across various matrices, with Deep Eutectic Solvents (DESs) offering a promising, efficient, and environmentally friendly extraction alternative. Analytical chromatographic separations continue to be predominantly conducted using C18 reversed-phase columns. Nevertheless, in recent years, researchers have explored various stationary phases, particularly to achieve the enantioseparation of cannabinoids.

1. Introduction

Cannabis is a versatile plant with a long history of medicinal, recreational, and industrial use, garnering significant scientific and commercial interest in recent years. Due to its pronounced therapeutic properties, it is considered a highly promising medicinal plant, despite its common use as a substance of abuse [1]. The legalization of cannabis in various regions has catalyzed an expansion of research efforts, focusing on its chemical composition, therapeutic potential, and safety profile. From a taxonomic point of view, there is no general agreement regarding the classification of this plant. However, three species of the Cannabis genus have been described: Cannabis sativa L., Cannabis indica Lam., and Cannabis ruderalis [2]. Among its many constituents, including terpenes, terpenoids, stilbenoids, lignanamides, carotenoids, flavonoids, and alkaloid compounds, the most notable class of compound in cannabis remains the phytocannabinoids [3]. In addition to these phytochemicals, terpenes are thought to interact synergistically with cannabinoids, potentially enhancing their therapeutic effects by contributing to the entourage effect [4].
Medical cannabis has swiftly gained significant importance in contemporary society, fueled by renewed interest in its use as a therapeutical tool. Two cannabinoids, Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD), are currently available on the market in formulations approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) [5]. These drugs represent a significant shift in how cannabinoids are perceived and used in medical practice, representing a departure from traditional herbal preparations to standardized pharmaceutical formulations backed by scientific evidence and regulatory approval. Beyond its use as pharmaceuticals, cannabis is also becoming increasingly popular in the food supplement and cosmetics industries. The expansion of commercial cannabinoid-based products is prompting revisions of cannabis regulatory frameworks to permit their legal use [6]. At the same time, efforts are being made to curb recreational usage, as cannabis remains the most widely used illegal drug worldwide. A legal limit has been established to distinguish between drug-type and non-drug-type cannabis. In most European countries, this classification relies on the content of Δ9-THC, the primary psychoactive compound in cannabis. Cannabis plants with a Δ9-THC content of 0.2% or less are classified as hemp and are primarily cultivated for industrial uses, such as fiber production. In contrast, cannabis plants with a Δ9-THC content greater than 0.2% are classified as marijuana and are subject to stricter regulations due to their psychoactive properties [7].
Over the past decade, numerous review articles have been published outlining the various analytical methods available for detecting phytocannabinoids [8,9,10,11,12,13,14,15,16,17,18], with some specifically focusing on Gas Chromatography (GC) [19,20] or Liquid Chromatography (LC) [21,22,23,24]. Due to the widespread use of High-Performance Liquid Chromatography (HPLC) coupled with Ultraviolet (UV) and Diode Array Detection (DAD), which has been featured in an astonishing 169 publications from 2022 to 2024, this review will comprehensively compile information regarding this technique. Furthermore, since recent reviews often fail to provide a thorough discussion of the approaches used to validate analytical techniques, the present review will analyze key validation parameters, such as precision, accuracy, and lower limits, utilized by the authors to achieve accurate and reliable quantification of cannabinoids. In addition, this review will briefly examine, within these manuscripts, the types of extraction methods and matrices selected for HPLC–UV analysis.

2. Cannabinoids

The term “cannabinoids” refers to a complex family of herbal, endogenous, and synthetic compounds that bind to specific receptors within the body’s endocannabinoid system, causing a medicinal response. These are classified into three groups: endocannabinoids, synthetic cannabinoids, and phytocannabinoids (Figure 1) [25]. Endocannabinoids play critical roles in regulating various physiological processes such as pain, mood, appetite, and memory by interacting with cannabinoid receptors (CB1 and CB2) in the endocannabinoid system [26]. Synthetic cannabinoids consist of a diverse range of chemical structures designed to interact with this endogenous system to mimic and enhance the effects of natural cannabinoids, particularly Δ9-THC [27]. Phytocannabinoids are primarily responsible for the plant’s medicinal and psychoactive properties. To date, more than 100 distinct phytocannabinoids have been identified from Cannabis sativa L. [28]. Most phytocannabinoids occur in two forms: as acids (dominant form in plants) and as neutral, decarboxylated compounds (commonly found in processed plant material). Acidic cannabinoids (AC) are thermally unstable and can undergo decarboxylation to neutral cannabinoids (NC) when subjected to light or heat (e.g., extraction at high temperatures, smoking) [29]. The two major cannabinoids in cannabis-related products are Δ9-THC and CBD. In this section, only phytocannabinoids with current therapeutic interest will be described.

2.1. Δ9-Tetrahydrocannabinol (Δ9-THC)

Among all cannabinoids, Δ9-THC shows the highest potency in both CB1 and CB2. It acts as a moderate partial agonist, resulting in a mixed agonist–antagonist behavior [30]. This interaction underpins many of its psychoactive effects, as well as its therapeutic properties as an analgesic, muscle relaxant, and antispasmodic agent [31]. Clinical trials have shown positive effect in treating neuropathic pain [32,33], opioid withdrawal [34,35], stress disorders [36,37], and sleep apnea [38]. Dronabinol (synthetic Δ9-THC) is already used in clinic for HIV/AIDS-induced loss of appetite [39] and to treat chemotherapy-induced nausea and vomiting (CINV) [40].

2.2. Cannabidiol (CBD)

CBD and its precursor, cannabidiolic acid (CBDA), are the most common nonpsychoactive cannabinoids found in cannabis-related products. CBD acts as an antagonist of CB1/CB2 agonists and a negative allosteric modulator of CB receptors [41,42]. Due to CBDA’s high content in certain cultivars, its non-psychoactive nature, coupled with its diverse pharmacological properties, CBD has gained significant attention in recent years, leading to a remarkable increase in the development of CBD-based product [43]. Several clinical trials involving CBD have revealed promising effects in epileptic seizures [44,45,46], Parkinson’s disease tremors [47], depression symptoms [48], anxiety [49,50], psychosis [51,52], substance use-disorder [53], and insomnia [54]. In addition, association of CBD and Δ9-THC has been studied in clinical trials for the management of multiple sclerosis-related spasticity [55,56], and CINV [57]. Sativex® (oromucosal spray containing standardized extract with 2.7 mg Δ9-THC and 2.5 mg CBD per 100 µL spray) and Epidyolex® (100 mg CBD per 1 mL oral solution) are already approved drugs to treat symptoms of multiple sclerosis and epilepsy, respectively [10].

2.3. Cannabinol (CBN)

CBN is formed by non-enzymatic oxidative aromatization of Δ9-THC [58]. Its levels in plant material are initially low but can increase over time as Δ9-THC is exposed to light, heat, and oxygen, resulting in higher concentrations of CBN in aged cannabis preparations [59]. CBN has lower binding affinities for the CB1 and CB2 receptors compared to Δ9-THC, which means it does not produce the same psychoactive effects. Recent clinical trial suggested that CBN may help reduce overall sleep disturbances [60] and possibly attenuate chronic itch in Epidermolysis Bullosa when applied in a cream form [61].

2.4. Cannabigerol (CBG)

CBG is a minor cannabinoid that displays weak partial agonist activity at both the CB1 and CB2 receptors [62]. Pre-clinical studies suggest several potential therapeutic interests, such as analgesic, anti-inflammatory [63], and antibacterial [64] effects, as well as a positive outcome in inflammatory bowel disease [65]. A patient survey revealed that consumers used CBG-dominant products to manage anxiety, depression, chronic pain, and insomnia [66], and a recent published clinical trial revealed positive effect of CBG in reducing anxiety and stress in healthy adults [67].

2.5. Tetrahydrocannabivarin (THCV)

THCV is a n-propyl analog of THC that lacks its psychoactive effects [68]. It binds to both CB1 and CB2 receptors acting as an antagonist or reverse agonist [69]. In rodent studies, THCV has been shown to reduce inflammatory pain [70], epileptic activity [71], and food intake [72]. In a clinical trial, significative weight loss was observed in subjects using THCV and CBD-infused mucoadhesive strips [73]. In addition, THCV has shown potential protective role against psychological and physiological effects induced by Δ9-THC in healthy volunteers [74].

2.6. Cannabidivarin (CBDV)

CBDV is a n-propyl homologue of CBD that displays low binding affinity for both CB1 and CB2 receptors [75]. CBDV has been link to intestinal anti-inflammatory [76], and anti-convulsant activity in animal models [77]. Recent human trials revealed a positive correlation between CBDV and seizure control in adults with focal seizures [78] and girls with Rett syndrome [79].

3. Extraction of Cannabinoids

An overview and discussion of the extraction methods described in the manuscripts collected in Table 1 [80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162], Table 2 [163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216], Table S2 [217,218,219,220,221,222,223,224,225,226,227,228], and Table S3 [109,223,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251], concerning original manuscripts published between 2022 and 2024, will be presented herein. Most of the studies are focused on the extraction of cannabinoids from plant materials (Figure 2), namely, inflorescence, leaves, and stems (Table S1, entries SE1–97). Nineteen manuscripts were published regarding the extraction of cannabinoids from resins and oils, natural and commercially obtained (entries SE98–116), while twenty-six considered pharmaceutical and other formulated products (entries SE132–157). Several research groups focused their research efforts on the extraction of cannabinoids incorporated into food, beverages, and tablets (entries SE117–131). Finally, a smaller number of publications considered the extraction of cannabinoids from biological matrices such as plasma and tissues (entries SE158–162). It is important to note that the authors do not intend to revise all extraction techniques applied to the analysis of cannabinoids, but rather to discuss those applied in recent research, considering the manuscripts collected in Table S1. For more detailed reviews on the topic, a considerable number have been published recently, namely by Sainz Martinez et al. [252], Al Ubeed et al. [253], Liu et al. [254], Nahar et al. [255], and Szijj [256].

3.1. Extraction of Cannabinoids from Plant Material

The flowering parts of the female cannabis plant (Table S1, entries SE1–70) are the most frequently used for cannabinoid extraction, as they contain the highest concentrations of cannabinoids [257,258]. Analysis of Table S1 indicate that solid–liquid extraction (SLE) is the most common technique used for extracting phytocannabinoids from plant material. To assist with this extraction methodology and increase the yield of extracted cannabinoids, various physical techniques are usually employed (Figure 3a). Dynamic maceration (DM), due to its simplicity, is an often-used procedure, normally accompanied by mechanical or manual stirring (entries SE10, SE11, SE13, SE45, SE47, SE50–52, SE54, SE67, and SE87) or shaking (entries SE1–7, SE9, SE24, SE26, SE52, SE65, SE71, SE73, SE76, and SE82). Vortexing is frequently used simply to allow thorough mixing between solvent and plant material; only in three manuscripts is vortexing used for extraction purposes (entries SE25, SE68, and SE83). Shaking-assisted extraction usually involves a broader and slower movement, often in a linear or circular fashion, accommodating larger volumes, making it more suitable for scaled-up extractions of up to 100 g of plant material and 500 mL of extraction solvent [83,125,183]. In comparison, vortexing creates a higher-speed circular motion, usually in small containers, suited only for small-batch extractions of around 100 mg of plant material [114,221]. In contrast to the simplicity of DM, supercritical fluid extraction (SFE) requires high-pressure solvents, especially supercritical CO2 (scCO2) and therefore entails specific automated equipment [259]. SFE is commonly used for cannabis extraction-separation in batch processes (Figure 3a) and is considered efficient for extracting both neutral and acidic cannabinoids, as it does not induce the degradation of acidic cannabinoid. Moreover, in SFE the co-extraction of undesirable compounds, like chlorophyll, is minimized [252,260,261]. SFE has recently been used to extract cannabinoids from ground plant material, namely inflorescences, leaves, and stalks, using scCO2 (entries SE48–51, SE53, SE67, and SE95). Fernández and co-workers [115] (entry SE50), performed the extraction of THC and CBD from two different cannabis inflorescences chemotypes containing high total THC and high total CBD content. Their strategy involved the addition of small amounts of ethanol (EtOH) as co-solvent to address the low polarity of scCO2, thereby enhancing the extraction process and increasing the total THC and CBD recovery. To optimize the extraction of the cannabinoids of interest, the authors also studied extraction temperature and pressure and performed a decarboxylation process. For the THC-dominant chemotype, the best extraction conditions were 70 °C, 300 bar using 10% of EtOH. For the CBD-rich chemotype, which is best extracted at lower temperatures, the optimum extraction conditions were 60 °C, 300 bar using 10% of EtOH. Furthermore, higher yields were obtained for both chemotypes using scCO2 extraction compared with EtOH DM at room temperature. Mileti et al. [221] (entry SE67) investigated various extraction techniques, including Soxhlet extraction (SE) and DM using EtOH as solvent, and compared their performance against SFE using scCO2. Although a direct comparison of these techniques is challenging due to significant differences in sample amount, temperature, time, and solvent volume (SE: 15 ± 1 g, 400 mL EtOH, 78 °C, 8 h; DM: 1 g, 100 mL EtOH (total volume), RT, 45 min (15 min each extraction); SFE: 25 ± 1 g, 40 °C, 140 min), the authors concluded that, under these specific conditions, SE yielded the highest amount of extracted material. Regarding CBD yield, DM performed best. They also noted the importance of considering cannabinoid degradation during longer extraction times and at higher temperatures, particularly with techniques like Soxhlet. SFE did not lead to higher cannabinoid yields, likely due to the non-polar nature of the supercritical fluid. It would have been important to study the use of a co-solvent, such as EtOH, as previously described, and to investigate whether comparable extraction times and temperatures across the different techniques would improve cannabinoid yield.
Mastellone et al. [93] (entry SE94) explored the use of natural eutectic solvents (ESs) in a dispersive solid–liquid microextraction (DSLME) method, complemented with sonication, for the extraction of flavonoids and cannabinoids from the aerial parts, mainly stem and leaves, of the cannabis plant. By integrating ultrasound with liquid–liquid microextraction, US-LLME enhances analyte transfer between immiscible phases while reducing the amount of solvent needed to enrich the final extract with analyte. The ESs used in the extraction process were prepared as a 1:1 mixture of hydrogen bond donor (HBD) and acceptor (HBA) natural compounds, namely menthol, linalool, thymol, carvacrol, terpinene-4-ol, and eugenol. Two years later, the group applied this methodology using different ESs (entry SE66), including an ammonium bromide salt (dimethyl-(2-hydroxy)ethyl-hexadecyl ammonium bromide) as the HBA and thymol as the HBD, as well as one based on natural terpenoids (menthol:linalool, 1:1) [220]. This DSLME ultrasound-assisted extraction (UAE) required only 100 μL of ESs and 2 mL of water as co-solvent per 100 mg of plant material (entry SE66). According to the authors, the ESs combining hydrophobic properties and hydrophilic moieties (ammonium bromide salt:thymol, 1:2), facilitated the interaction with both polar and less polar compounds, resulting in an enriched cannabinoid extraction from inflorescences. However, it slightly reduced the sustainability of the process compared to the natural ESs pair. It is important to note that the US-DSLME technique was used to analyze cannabinoids in three different hemp products—aerial parts of Cannabis sativa L., plant inflorescences, and CBD oil—and proved to be easily adaptable to these different matrices.
Of all the methods used for extraction of bioactive compounds, UAE is the most widely employed for the extraction of cannabinoids from plant materials (Table S1 and Figure 3a). The most used solvents in US-SLE are methanol (MeOH) (entries SE27–35, SE37, SE56, SE57, SE65, SE70, SE76, SE79, SE84, and SE97), EtOH (entries SE14–20, SE37, SE52, SE76, and SE85) and acetonitrile (ACN) (entries SE36, SE37, SE58, SE59, and SE80). Interestingly, a 9:1 mixture of MeOH:chloroform (CHCl3) (entries SE41, SE42, SE44, SE63, SE81, SE92, and SE93) is often used, even though CHCl3 has been associated with CBD instability [262]. Chlorinated solvents and MeOH are not considered safe for consumer application and are also discouraged for laboratory safety reasons, as they are not classified as low-toxicity solvents and have a significant environmental impact [263]. Hence, EtOH is recommended for cannabinoid extraction because of its safety, effectiveness, and regulatory acceptance. In 2024, an extraction protocol using 96% (v/v) EtOH was included as the official cannabis flower extraction method in the European Pharmacopoeia [264].
Several parameters and factors can be critical in optimizing the efficiency and effectiveness of the extraction process, namely the amount of sample relative to solvent volume, particle size, time taken for the extraction, number of consecutive extractions, and temperature [252,255]. These experimental conditions were studied in several manuscripts gathered in Table S1 and exhibit significant variability depending on the purpose of the research and the desired selectivity during the extraction process. For example, this process can involve extraction times ranging from as little as 1 min to as much as 1 h (entries SE91 and SE80) for the extraction of, respectively, 50 mg of cannabis samples using 10 mL of an EtOH:ACN (1:1, v/v) mixture and 150 mg of leaves and vegetative shoots using 1 mL of ACN at a temperature of 55 °C [95,172]. Concerning the number of consecutive extractions or the use of extended extraction times, de Souza et al. [141] (entry SE28) found that the latter led to a decrease in THCA and Δ9-THC, accompanied by a corresponding increase in CBN. This suggests, as mentioned previously, that prolonged extraction times may lead to the degradation of these cannabinoids. Coherently, THCA and Δ9-THC recoveries were slightly lower under conditions involving two extraction steps with solvent renewal compared to a single-step extraction, supporting the hypothesis of degradation during extended extraction. These findings align with a previous study by Mudge et al. [263], which demonstrated that UAE is superior to shaking, but that extended sonication time can lead to the degradation of cannabinoids. However, de Souza et al. also noted that a complete cannabinoid recovery might not be attainable with just a single extraction step. Design of Experiments (DoE) studies considering the effect of variables such as granulometry, time, and the number of extractions on cannabinoids recovery allowed the authors to conclude that the ideal compromise among these factors involves granulometries of 180 to 250 µm of plant material and two extractions of 10 min each (entry SE28). Recently, DoE has been applied in the optimization of cannabinoids extraction protocols, constituting a valuable tool for systematically studying the relationship among multiple input factors (such as solvent type and concentration, temperature, extraction time, particle size of plant material, solid-to-liquid ratio, etc.) and the desired output, namely yield, purity, or efficiency of the process.

3.2. Extraction of Cannabinoids from Oils and Resins

Concerning the study of hemp or cannabis concentrates, the terms ‘resin’ or ‘oil’ are freely applied and refer often to different types of extracts, namely full-spectrum extracts, CBD isolates, distillates, crude extracts, etc. Therefore, careful attention is required when assessing the results. Ultrasound is the technique generally employed for the extraction of cannabinoids from oils and resins (Table S1, entries SE98–116 and Figure 3b). The extraction procedures are primarily defined by the type of matrices used and secondarily by the extracting solvent. For example, when extracting bioactives from oils, extraction procedures are frequently conducted in polar solvents such as MeOH (entries SE99, SE103, and SE107), EtOH (entries SE102 and SE105), isopropanol (entry 104), or by partitioning in a more complex biphasic solvent system, namely n-Hexane:Ethyl acetate:ACN:MeOH (8:2:8:2, v/v) (entry SE106). When considering resins as matrices, cannabinoid extraction is primarily performed using US-SLE with polar solvents such as MeOH (entry SE115) or a combination of MeOH with CHCl3 (entries SE112 and SE113).

3.3. Extraction of Cannabinoids from Food Products

The number and variety of commercially available cannabis-infused food products have increased globally. These products cover a wide range of food matrices, including juices, tea and coffee to ice cream, honey, cookies, candies, gummies, and chocolates (Table S1, entries SE117–131). Such diversity in food matrices necessarily involves adapting extraction protocols for the efficient recovery of cannabinoids. Several manuscripts focused their research on, for example, gummies matrices. In these cases, ultrasound or shaking extraction methods were employed, using water or a mixture of this solvent with MeOH, MTBE, or ACN (entries SE126–128, SE130, and SE131). Melchert et al. analyzed CBD-fortified gummy bears (entry SE126) using a US-SLE procedure with a MeOH:H2O (1:1, v/v) mixture [121], while Song et al. examined several hemp-infused edibles (entry SE131) using the same solvent mixture but with a higher organic solvent content (95%). In this last example, the extraction was performed by first forming a suspension of the pulverized sample in water, followed by the addition of MeOH and ultrasonication [184].

3.4. Extraction of Cannabinoids from Pharmaceutical and Other Formulated Products

Cannabinoids in pharmaceutical and other formulated products of medicinal interest come in diverse forms, including pills, powders, ointments, oil-in-gel emulsion, lotions and creams, bandages, vaping products, and nanoparticles (Table S1, entries SE132–157). Each type of formulation presents unique analytical challenges due to its distinct matrices, and tailored testing methods are applied to ensure accurate measurement and identification of cannabinoids within these complex matrices. Nevertheless, ultrasonication was the technique most often used in these matrices (Figure 3d). Regarding cosmetic products, the reported studies utilize ACN or MeOH in combination with either ultrasound or stirring for extraction (entries SE135–137, and SE141). Pires et al. [208] (entry SE135) studied 31 commercially available cannabis-based products in Portugal, of which nine were for cosmetic use (creams, shower gel, shampoo, and balm). The extraction procedure for cosmetic creams comprised agitating 0.5 g of the sample in ACN for 15 min, followed by centrifugation and supernatant filtration. Mouton et al. [104] also used ACN as the extraction solvent, employing ultrasonication when analyzing a total of five cosmetic products. The extraction protocol was optimized for several types of matrices, with longer times (45 min) required for the topical ones (entry SE141).

3.5. Extraction of Cannabinoids from Biological Materials

Cannabinoids can be extracted from various biological matrices, including urine, blood, oral fluid, plasma, hair, and different tissues (Table S1, entries SE158–162). As new cannabis-based medicinal products and technologies are developed and tested, the number of types of matrices increases due to the rise in both in vitro and in vivo studies. Analyses of Table S1 indicate that classic protein precipitation followed by LLE using immiscible solvents like hexane and ethyl acetate, along with vortexing, was applied to human and mouse plasma samples, as well as mouse lung, liver, and brain tissues (entries SE158 and SE161) [82,235]. SLE was applied using a bead-beating system at lower temperature for the extraction of cannabinoids from zebrafish larvae tissue (entry SE162) [177]. Cheng et al. [234] developed a transdermal drug-delivery system based on a CBD nanosuspension that was incorporated into dissolving microneedles (entry SE161). In this example, the authors used a rat model to test the efficacy and safety of this delivery platform, and it was necessary to quantify the amount of CBD retained in the rat skin. The processing of skin samples involved their removal, weighing, and extraction in MeOH for 24 h with continuous agitation in a shaker, followed by two periods of 30 min using ultrasound. As can be seen in Table S1, depending on the assay performed and the specific aims of the study, adequate extraction protocols need to be designed.

In summary, regarding the manuscripts collected in Table S1, advanced methods such as SFE, UAE, and the use of DESs to extract cannabinoids were the most commonly applied extraction methods. It is important to note that while cannabinoids can be extracted from different matrices using a diverse array of techniques, each method has unique limitations. SFE offers clean, solvent-free extracts with high selectivity and efficiency; however, its accessibility is limited by high equipment costs and complexity. Similarly, techniques like UAE, though effective and rapid, can lead to the thermal degradation of heat-sensitive compounds if not carefully optimized, and may have poor scalability. UAE is clearly the most widely applied technique across all matrices as can be seen in Figure 3. EtOH extraction assisted by ultrasound is the most used technique for plant material matrices, as it is efficient at extracting a wide range of cannabinoids. Additionally, maceration is also often used, although it is a slower and less efficient process. The use of ESs in the extraction of cannabinoids is at an early stage of research. However, they are gaining popularity for the extraction of natural products due to their compatibility with most extraction techniques, lower toxicity, enhanced biodegradability compared to conventional solvents, and the fact that they are often derived from natural and less expensive materials. Regarding the extraction of cannabinoids from complex matrices, namely biological samples such as plasma and tissues, the traditional LLE method using immiscible solvents combined with vortexing is commonly employed. This technique is preceded, in the studies reviewed, by protein precipitation, which is used as a pre-treatment step before the extraction process. Furthermore, adding steps such as solid-phase extraction (SPE), can be beneficial, if applicable, resulting in cleaner samples, albeit at a higher cost. For example, Mastellone et al. [93] used SPE to remove chlorophylls from the plant extracts (entry SE94).

4. HPLC-UV Analysis

Among the several separation techniques available, LC is generally the preferred method for determining the cannabinoid profile of the cannabis plant. Its advantage, for instance, over GC lies in its ability to directly analyze all cannabinoids, including acidic precursors, without the need for derivatization. Derivatization is essential in GC because the high temperatures used cause acidic cannabinoids (AC) to decarboxylate into their neutral forms. Moreover, thermal degradation (e.g., oxidation, isomerization) of cannabinoids can potentially occur in the GC injector port and column, leading to inaccurate results [10,265].
For the analysis of cannabinoid content, Ultraviolet (UV) and Mass Spectrometry (MS) are widely used [24]. MS enhances the sensitivity and specificity of analyses, which is crucial for quantifying minor cannabinoids, especially in biological matrices [9]. However, HPLC-UV remains the favored method for analyzing cannabis plant samples due to its cost-effectiveness and operational benefits [17,21], making it more accessible for cannabis growers and commercial suppliers [184]. In 2024, with the addition of a cannabis monograph to the European Pharmacopoeia [264], an official HPLC-UV method was established for the quantification of CBD, CBDA, CBN, Δ9-THC, and THCA.
This section describes what has been done with LC coupled with UV detection for the identification and quantification of phytocannabinoids from 2022 to 2024 (Table 1 (entries 1–65), Table 2 (entries 66–104), Table S2 (entries SH01–12), and Table S3 (entries SH13–34).

4.1. Stationary Phases

Almost all studies used reversed-phase (RP) octadecyl (C18) packed columns, with five studies employing C18-Pentafluorophenyl (PFP) columns (entries 15, 101, and SH18), and one a column with mixed aromatic functionality (C18-AR) (entry SH28). In addition, two studies used octyl (C8) columns (entries 68 and 73). The most frequently used columns, each featured in at least five studies, were the following:
  • (i) particle size > 2 μm: Ascentis Express C18 (150 × 3.0 mm, 2.7 μm), Waters Cortecs Shield RP18 (150 × 4.6 mm, 2.7 μm), InfinityLab Poroshell 120 EC-C18 (150 × 3 mm, 2.7 μm), NexLeaf CBX for Potency C18 (150 × 4.6 mm, 2.7 μm), Phenomenex Kinetex C18 (150 × 4.6 mm, 2.6 μm), and Phenomenex Luna C18(2) (150 × 4.6 mm, 5.0 μm), all with a length of 150 mm.
  • (ii) particle size < 2 μm: Waters Cortecs UPLC C18 (100 × 2.1 mm, 1.6 μm) and Phenomenex Kinetex C18 (150 × 2.1 mm, 1.7 μm).
While using a guard column with the same stationary phase is recommended, especially when analyzing complex matrices [266], only about a quarter of the studies reported its use.
Since its introduction in 2004, the Ultra High-Performance Liquid Chromatography (UHPLC) system has gained significant popularity, offering numerous advantages over HPLC, including higher sensitivity, enhanced resolution and efficiency, reduced analysis time, and lower solvent consumption [9,267]. These benefits are particularly crucial when analyzing complex matrices, such as those found in cannabis analysis. Among the previously cited columns, UHPLC systems can operate with columns that have smaller particle sizes (1.7 µm) and smaller inner diameters (2.1 mm). Another notable advancement in column technology is the use of superficially porous particles, often referred to as core-shell or fused-core particles, which enhance performance [267,268]. This column chemistry merges the advantages of both fully porous and nonporous particles and currently represents the majority of columns utilized for cannabinoid separation. Furthermore, although most of the columns discussed are C18 types, there are subtle differences in their stationary phases: Poroshell 120 EC-C18, Kinetex C18, and Ascentis Express C18 contain dimethyl-octadecyl stationary phases, while Raptor ARC-18 features a diisobutyl-octadecyl phase, and Cortecs Shield RP-18 and Ascentis Express RP-Amide incorporate embedded polar functional groups. These minor variations affect column polarity, which, in turn, influences cannabinoid separation and may be beneficial depending on the analysis objectives. A recent study by Song et al. [269] evaluated four of these column types (Poroshell 120 EC-C18, Raptor ARC-18, Cortecs Shield RP-18, and Ascentis Express RP-Amide), all sharing the same dimensions (150 mm × 2.1 mm) and core-shell particle size (2.7 μm) for the separation of 18 cannabinoids. Their results indicated that when using HPLC-DAD for cannabinoid quantification, the two conventional RP stationary phases (Poroshell 120 EC-C18 and Raptor ARC-18) provided the best results, offering shorter run times under similar conditions. Szijj et al. [243] (entry SH28) used a C18-AR column. The choice of a column with mixed aromatic functionality was based on a prior study conducted by Ciolino et al. [262], which investigated four columns of the same dimensions (250 × 4.6 mm, 5 µm) with different derivatized silica stationary phases: Luna C18(2), phenylhexyl (Luna), and two columns featuring mixed C18/aromatic functionality (ACE 5 C18-AR and Siliachrom C18/phenyl). They reported that the ACE 5 C18 column was the only one capable of resolving all nine tested cannabinoids under the chromatographic conditions examined.
As an alternative to reversed-phase liquid chromatography, De Luca et al. [270] investigated several polar-bonded stationary phases (Si, CN, Diol, and NH2 absorbents) and their impact on cannabinoid separation. A promising condition was achieved when normal phase with NH2 adsorbent was employed for the separation of five neutral cannabinoids (CBC, CBD, CBG, CBN, and Δ9-THC) using as mobile phase a mixture of heptane/isopropanol 95:5% (v/v). The same group also investigated different polysaccharide-based (cellulose and amylose) chiral stationary phases. They achieved the best results using CHIRALPAK IC and IF columns for the separation of CBD, CBDA, THCA, (-)-Δ9-THC, and both CBC enantiomers, with ACN and phosphoric acid mixtures as the eluent [271]. In addition, Russo et al. [272] were able to achieve enantioseparation of trans-CBD, trans-CBDA, trans9-THC, and trans9-THCA. The best separation was obtained using a CHIRALPAK AD-RH column with different percentages of organic modifiers in the mobile phase. Other CHIRALPAK columns (IA, ID, IE, and IG) were also shown to deliver good separations of CBD enantiomers [273]. Huang et al. [216] conducted a comparative study using two HPLC-DAD-MS methods for the separation of CBD, Δ(4)8-iso-THC, Δ8-iso-THC, Δ8-THC, and Δ9-THC. One method employed a reversed-phase column (entry 83), but only the method using a normal-phase silica-Ag(I) column (entry 104) effectively separated these five isomers.

4.2. Mobile Phases and Type of Elution

Separating the main cannabinoids via LC can be challenging; therefore, several studies favor gradient elution (Table 1 and Table S2) over isocratic elution (Table 2 and Table S3). Generally, gradient methods allow for shorter run times, higher flow rates, and better peak resolution [14]. The most common solvent system was ACN and water acidified with 0.1% formic acid.
Regarding the choice of organic solvent (B), ACN was, as expected, the preferred option by far (entries 1–47, 66–99, SH01–10, and SH13–29) over MeOH (entries 48–60, 100–102, SH11, SH12, and SH30–33). ACN offers a few advantages in comparison to alcohols: (a) its lower viscosity results in less system backpressure, consequently enabling higher flows; and (b) its lower UV absorbance at wavelengths most frequently used for cannabinoid analysis can lead to increased method sensitivity and baseline uniformity [274]. Nevertheless, each organic modifier offers distinct characteristics that may be valuable to explore; for instance, MeOH has some acidic properties, while ACN exhibits dipolar properties [275]. As an example, Franklin et al. [276] were able to obtain a better separation of Δ9-THC, Δ8-THC, and CBL when a mixture of 50:50 ACN and MeOH was used. Six studies reported the use of both ACN and MeOH (entries 61–65, and 103).
The most used aqueous phase (A) was water acidified with 0.1% formic acid (entries 7–26, 48–55, 62, 63, 71–82, SH03–06, and SH20–22). Adding pH-modifying agents to the mobile phase can influence the selectivity factor by increasing ionic strength and altering the pH [117]. AC will be more affected, showing greater retention at lower pH values due to the prevalence of their non-ionized form, whereas NC are much less impacted by mobile phase pH changes [141]. Moreover, acid additives play an important role in achieving optimal peak shape with minimum tailing, especially for AC [276,277]. The choice of acid modifier can also affect baseline uniformity in the resulting chromatogram. Ryu et al. [278] tested 0.10% formic acid, 0.05% trifluoroacetic acid, and 0.10% phosphoric acid under gradient conditions and reported that formic acid produced a more pronounced raised baseline than trifluoroacetic acid, while phosphoric acid had minimal impact. In addition to formic acid, other researchers have employed different agents to adjust the pH of the aqueous phase, including trifluoroacetic acid (entries 60, 96–99, 102, SH10, and SH24), ammonium formate (entries 28–36, 56, 65, 84–94, SH11, and SH23), phosphoric acid (entries 37–45, 57, 64), acetic acid (entries 46, 47, 103, SH08, SH09, SH25–28), and ammonium acetate (entries 58 and 59). Only a few studies did not incorporate a pH modifier (entries 1–3, 66–69, SH01, SH13–19, SH30–32, and SH34).
Furthermore, the mobile phase is also optimized to ensure the best separation of the target cannabinoids without interference from other matrix components. It is important to note that in some of these investigations, the goal was to characterize the matrix composition and not only to assess cannabinoids, leading to long runs covering a wide range of solvent gradient (e.g., entries 9, and 10).

4.3. UV Detection

Detection depends on the UV absorption capability of the analyte. In cannabinoids, the primary chromophore is a substituted phenolic ring (Figure 1). Further contributions to the UV spectrum occur when cyclization of the non-phenolic part is present, leading to a conjugated double bond (CBC and CBCA) or the formation of a second phenyl ring (CBN and CBNA) [17,265]. Consequently, many cannabinoids exhibit similar UV spectra, which can result in low specificity. To address this limitation, diode array detection (DAD) can be valuable for peak characterization, as the absorption spectra differ, at least between neutral and acidic cannabinoids. Additionally, exploring these UV spectral differences can also enhance sensitivity by enabling the measurement of various cannabinoids at distinct wavelengths. For example, CBD, CBG, Δ9-THC, and NC in general, exhibit a single absorbance peak around 210 nm, while their corresponding acids show a primary peak near 220 nm, along with two smaller peaks around 260–270 nm and 300 nm [157,158,194,279,280]. These absorption profile differences have been applied to enhance method sensitivity for NC and AC, as demonstrated by the following examples: NC-210 nm, AC-220 nm (entries 7, 14, and 28), NC-210 nm, AC-221 nm (entry 17), NC-210 nm, AC-225 nm (entry 15), NC-210 nm, AC-228 nm (entry 88), NC-210 nm, AC-284 nm (entry 73), NC-225 nm, AC-306 nm (entry 43), and NC-228 nm, AC-306 nm (entry 45). For the main NC, exceptions to this absorbance spectrum trend are seen with CBN and CBC. They display two absorbance peaks, one around 220–230 and a second around 280–285 nm [129,180,194], enabling them to be also analyzed at the same wavelengths as AC (entries 7, 15, and 73). Their corresponding acid (CBCA and CBNA) also exhibit a different absorbance profile, with only one major peak around 254–262 [130,180,194]. Song et al. [157,182,184] used 262 nm for quantification of CBCA and CBNA (entry 61) or, individually, at 251 nm and 261 nm, respectively (entries 84 and 86), and 284 or 285 nm for CBC and CBN. These two NC were also detected at 283 nm by Carona et al. [82] (entry 3).
When a single wavelength was used, it was typically set between 210 and 240 nm with 220 nm and 228 nm being the most frequently used. A few exceptions were reported: 269 nm (entry 75), 270 nm (entries 36, SH04, and SH05), 272 nm (entry 68), 275 nm (entry 94), and 280 nm (entries 71 and 77). Barhdadi et al. [173] stated that by using 269 nm they were able to decrease the UV-absorbance of selected interfering terpenes; Križman [281] reported improved sensitivity (tenfold reduction in signal noise) at 275 nm compared to 228 nm, and Benkirane et al. [90] selected 280 nm since it is a typical maximum absorption wavelength for phenolic compounds. In addition, Song et al. [182] quantified cannabinoids using two wavelengths to increase method specificity (entry 84).
To validate and/or improve specificity, some authors used MS for cannabinoid assignment and qualitative evaluation, and UV for quantification (e.g., entries 14, 25, 29, 70, 75, 88, 98, 100, and 102). MS was also used to quantify cannabinoids below the limit of quantification (LOQ) obtained by UV (e.g., entry 88). In recent years, hyphenated dual techniques such as LC-UV-MS have become increasingly popular due to their enhanced sensitivity and specificity [24].

4.4. Retention Time and Difficult Resolutions

Retention times will differ depending on the solvent system and gradient employed. However, since almost all publications included in this review used reversed-phase columns, general considerations can be made based mainly on lipophilicity [270,282,283]:
  • CBDV(A) and THCV(A) have shorter retention times than their homologous CBD(A) and Δ9-THC(A) due to their shorter C3-side chains (n = 3 vs. 5, respectively), which leads to decreased lipophilicity.
  • Cannabinoids with two hydroxyl groups are more polar than their one-hydroxyl chromane or chromene counterpart, leading to shorter retention times (e.g., CBD, CBDV, and CBG vs. Δ9-THC, THCV, and CBC, respectively).
  • Δ9-THC tends to elute slower than its oxidative product, CBN, due to a decrease in lipophilicity caused by the two extra double bonds formed during ring aromatization.
  • CBC(A), which has two aliphatic side chains, is slightly less polar than Δ9-THC(A), resulting in longer retention times.
The retention times of NC compared to their acidic counterparts vary depending on several factors, such as pH, eluent system, and gradient. For instance, Büttenbender et al. [154] (entry 58) used a gradient of 10 mM ammonium acetate (pH 5.2) and MeOH, resulting in all AC eluting before any NC. Song et al. [213] (entry 102) noted that an elution system of water and MeOH exhibited a stronger retention for AC, leading to a significantly altered elution order of cannabinoids compared to the water and ACN elution system they tested.
When optimizing chromatographic conditions, among the most frequently detected cannabinoids, particular attention should be given to the CBD/CBG and Δ8-THC/Δ9-THC pairs. The CBD/CBG pair differs by a closed versus open ring side chain, while Δ8-THC and Δ9-THC vary by the position of a single double bond, resulting in similar polarity in both cases. These structural similarities can lead to co-elution, and their comparable UV spectra make accurate identification and quantification more challenging. Furthermore, as previously noted, these NC are less responsive to changes in the mobile phase pH. De Souza et al. [141] (entry 46), using DoE-assisted development, found that, within their design parameters, the CBD/CBG pair achieved better resolution at lower temperatures and with a flow rate close to 0.8 mL/min, while the Δ8-THC/Δ9-THC pair benefited from higher temperatures and flow rates. In both cases, extending the initial gradient (without altering the starting ACN percentage) improved resolution. Büttenbender et al. [154] (entry 58) also reported improved resolution between CBD and CBG when the temperature was decreased. Roussel et al. [105] (entry 18), applying prediction intervals for robustness, observed increased CBD/CBG resolution with higher temperatures and lower flow rates for their solvent system. Song et al. [182] (entry 84), in preliminary studies, reported improving the resolution of both pairs to the required minimum acceptable value of 1.5 by increasing the stationary phase length. In another work, Song et al. [213] (entry 102) found that the resolution for Δ9-THC/Δ8-THC was improved in a water/MeOH mobile phase, while the resolution for CBD/CBG was inferior compared to a water/ACN eluent. Focusing on the Δ9-THC/Δ8-THC pair, Pittiglio et al. [134] (entry 41) developed a method that allows baseline resolution at 500 ppm.
Additional cases of co-elution can also pose challenges, depending on the specific analytical method developed. For instance, Song et al. [168] (entry 70) were not able to separate THCVA and CBG under the primary conditions stablished. However, a second run of the sample with a different column temperature (45 vs. 30 °C) and detection at 269 nm instead of 230 nm, was sufficient to obtain good THCVA/CBG resolution.
Al-Maraseemi et al. [236] (entry SH19) evaluated retention time, peak areas, and tailing factors of CBD using Box–Behnken design experiments. They identified ACN composition as the most critical parameter affecting retention time, while the flow rate and oven temperature were the most significant factors influencing the tailing factor and peak area.

5. Analytical Method Validation

The validation of an analytical method is the process of proving that a specific method is suitable for its intended purpose by evaluating its performance characteristics under a variety of conditions [284]. Defining its intended use will have a major impact on the validation protocol and the established parameter limits. For instance, surveillance methods will prioritize speed and may tolerate lower precision and a higher rate of false positives, while dispute resolution methods will demand higher accuracy, precision, and reproducibility, making assay time less critical [285]. Nevertheless, method validation, which ensures the reliability, accuracy, and reproducibility of data, is crucial in regulated industries where product safety and quality are the highest priority [286].

5.1. Validation Guidelines

These guidelines provide a framework for analyzing key analytical performance characteristics for method validation such as specificity, linearity, reportable range, precision, accuracy, lower range limits, robustness, and system suitability. Several guidelines and compendiums were mentioned in the selected papers for validation analysis (Table S4, entries SV01-50), such as the following:
  • International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use—ICH guidelines: Validation of analytical procedures Q2 (R1) [287] and Q2 (R2) [288]; M10 on bioanalytical method validation and study sample analysis [289]—22 papers.
  • Association of Official Analytical Collaboration (AOAC) International guidelines: Appendix F: Guidelines for Standard Method Performance Requirements [285]; Appendix K: Guidelines for Dietary Supplements and Botanicals [290]; Standard Method Performance Requirements (SMPRs®) for Quantitation of Cannabinoids in Plant Materials of Hemp (Low THC Varieties Cannabis sp.) [291]—10 papers.
  • International Organization for Standardization (ISO) 17025: General requirements for the competence of testing and calibration laboratories [292]—9 papers.
  • United States Food and Drug Administration (FDA): Analytical Procedures and Methods Validation for Drugs and Biologics—Guidance for Industry [293]; ORA Lab Manual Vol. II—Methods, Method Verification and Validation (ORALAB.5.4.5) [294]—6 papers.
  • Agência Nacional de Vigilância Sanitária (ANVISA) RDC Nº 166/2017 [295]—3 papers.
  • Eurachem group—The Fitness for Purpose of Analytical Methods [296]—3 papers.
  • United States Pharmacopeia (USP) (e.g., [297])—3 papers.
  • American Academy of Forensic Science: Standard Practices for Method Validation in Forensic Toxicology (ANSI/ASB Standard 036) [298]—2 papers.
  • SANTE 11312/2021—Analytical Quality Control and Method Validation Procedures for Pesticide Residues Analysis in Food and Feed [299]—2 papers.
  • United Nations Office on Drugs and Crime (UNODC): Guidance For the Implementation of a Quality Management System in Drug Testing Laboratories [300]—1 paper.
  • Scientific Working Group for Forensic Toxicology (SWGTOX): standard practices for method validation in forensic toxicology [301]—1 paper.
Notably, several papers combined multiple guidelines, with ICH being the most frequently included. Although these guidelines share common principles, they vary in their scope, focus, and application, reflecting the diverse needs of different sectors. In this section special attention will be given to three validation parameters—accuracy, precision, and lower limits—and to the most cited guidelines: ICH and AOAC.

5.2. Precision

The precision of an analytical procedure reflects the closeness of agreement, or degree of scatter, between a series of measurements obtained from multiple samplings of the same homogeneous sample under the specified conditions [299]. Precision can be assessed in terms of repeatability, intermediate precision, and reproducibility.
Repeatability, also referred to as intra-assay precision, refers to the precision of measurements obtained under identical operating conditions within a short timeframe. Several authors also report repeatability as intra-day precision. ICH Q2 recommends at least nine determinations covering the established linear range or a minimum of six determinations when analyzing the target concentration at 100%. Most frequently, the reviewed papers used three quality control (QC) concentration levels of the selected calibration curve: low, medium, and high, analyzed in triplicate. ICH M10 recommends a fourth QC corresponding to LOQ to evaluate precision and accuracy. However, overall, the investigated papers used between one to six concentrations, and between three to twelve injections per concentration to investigate repeatability.
Intermediate precision reflects the variability observed within a laboratory. Common variations that can be evaluated include different days (inter-day), analysts, equipment, and environmental conditions. In most studies, inter-day precision was assessed over a period of three days with a few papers using two days (entries SV01, SV13, SV31, and SV42); five days (entry SV38); six days (entries SV14 and SV37); and ten days (entry SV41). Less frequently, intermediate precision was also evaluated using different analysts (entry SV01 and SV02). Lastly, reproducibility involves evaluating inter-laboratory precision using the same method.
In most works, the precision of the method was expressed as relative standard deviation (RSD%) or coefficient of variation (CV%) and calculated according to Equation (1), where “SD” represents the standard deviation and “μ” the average of the data [298].
Depending on the scope of the papers, the procedure to assess precision ranged from merely evaluating the instrument precision using standard solutions of the analyte to incorporating complete sample preparation with different analysts, operating conditions, and equipment. Furthermore, the scope of the analysis determines the variability expected and accepted; precision typically decreases as concentration diminishes and as matrix complexity increases, especially in biological samples [284]. For that reason, the ICH M10 guideline established that precision should be within ±15% at each level of the linear range, but allowing ±20% at LOQ. The ANSI/ASB Standard 036 guideline establishes a maximum RSD of 20%, but notes that certain methods require RSD limits of 10% or lower. The AOAC SMPR® 2019.003 guideline, which is specific for cannabinoids in plant materials, recommends different limits depending on the concentration of the analyte (% on dry weight basis): 0.05–0.5%: RSD ≤ 5% and ≤10%; >0.5–5%: RSD ≤ 3% and ≤8%; 5–35% (just for CBD and CBDA): RSD ≤ 2% and ≤6% for repeatability and reproducibility, respectively. Conversely, for drug products and drug active pharmaceutical ingredients, the FDA establishes a repeatability RSD of less than or equal to 3.0% and 2.0%, respectively [294]. In the analyzed papers, some authors also defined distinct limits for different precisions: RSD not exceeding 5% and 10% (entries SV30 and SV31) or 15% and 30% (entry SV36) for repeatability and intermediate precision, respectively.
For assay method validation, some authors include, as system suitability tests, other precision parameters such as the precision for retention time, peak area, peak height, or tailing factor (e.g., entries SV01, SV02, SV04, and SV05):
RSD% = SD/μ × 100

5.3. Accuracy

As described by ICH Q2, accuracy, sometimes referred to as trueness, in an analytical procedure refers to how closely the results obtained agree with the true value or an accepted reference value [287,288]. Related to accuracy, the AOAC guideline uses the term “bias” to describe the difference between the expected test result and an accepted reference value [285]. Accuracy should be determined across the analytical procedure’s entire reportable range and demonstrated under standard analytical procedure conditions, including, when possible, sample matrix effects and the sample preparation protocol. As an example, ICH Q2 suggests testing accuracy using three concentration levels in triplicate across the established range. There are a few ways that accuracy can be determined:
  • Comparison to a reference standard or to an already validated procedure.
Accuracy can be assessed by applying the analytical method to a known-purity analyte, such as a reference standard. Ideally, to prevent biased results unrelated to the method’s performance, calibration standards and QC samples should be prepared from different stock solutions [302].
Some studies used QCs prepared solely in solvent to assess validation parameters. However, for analytes in products, recovery experiments from matrices before and/or after extraction should also be performed to investigate potential matrix effects. For example, Song et al. [157,168,182,184,213] (entries SV19-23) used QCs prepared in MeOH containing an internal standard to assess accuracy and precision. However, all their analyzed hemp-infused samples were spiked with the non-naturally occurring cannabinoid ACBD to track recovery for every sample preparation. Likewise, Correia et al. [172] (entry SV34) verified the complete cannabinoid extraction recovery from a surrogate matrix to ensure that validation parameters obtained from neat standard solutions were valid.
  • Recovery of the analyte spiked into blank matrix or surrogate matrix.
The procedure involves spiking a formulation blank (containing all non-analytes) with a precisely measured quantity of the pure analyte. Ideally, the spiked sample is then subjected to both sample preparation and HPLC analysis, and the obtained results are compared with the predicted values [284]. However, the presence of cannabis extracts in formulations complicates recovery experiments due to the challenge of obtaining a suitable cannabinoid-free cannabis blank matrix, leading several authors to resort to surrogate matrices. In the reviewed papers, for the cannabis inflorescence matrix, Hall et al. [162] (entry SV02) used chamomile as a surrogate matrix, reporting it as an inexpensive floral material without cannabinoids that shares similar phytochemicals (terpenes and flavonoids) with cannabis. Correia et al. [172] (entry SV34) used hops from Humulus lupulus, a member of the Cannabaceae family like the cannabis plant, and Pires et al. [208] (entry SV38) used flowers from Urtica dioica to mimic cannabis flowers and herbal tea samples to perform spiking experiments before and after extraction.
A few studies employed matrices with residual levels of cannabinoids as blank matrices. Leersnijder et al. [174,175] (entry SV17) used fiber-type herbal cannabis, with negligible cannabinoids, as a blank matrix for recovery testing. Chan [164] (entry S41) used a sample matrix obtained by extracting only a small amount of cannabis samples, with subsequent dilutions before the spiking experiments if the analyte concentration was still elevated. Other authors used cannabis plant waste material obtained after extraction procedures as the blank matrix for validation studies [151,169,180] (entries SV03, SV32, and SV37). In these cases, during spike recovery experiments, if necessary, residual background cannabinoids were subtracted from the results obtained, as in the standard addition method.
Several oils were employed as surrogates for cannabis oil products: coconut oil [218] (entry SV26), MCT oil [243] (entry SV08), olive oil [162,181,244] (entries SV02, SV11, and SV13), sunflower oil [181] (entry SV13), and sunflower and coconut oil (60:40) [237] (entry SV28). For formulated products with cannabis extracts/cannabinoids, the spiking experiments were performed in cannabis-free formulations comparable to those examined [186,216,246] (entries SV25, SV29, and SV50). In addition, Melchert et al. [121] (entry SV45) used honey (rapeseed flower) as a surrogate matrix for formulated products, and Carona et al. [82] (entry SV09) used human plasma and mouse matrices without the investigated analytes as blank matrices for validation studies.
  • Standard addition method.
Given the inherent presence of cannabinoids within the plant matrix, a spiking methodology using standard addition was commonly applied. In these cases, spiked and unspiked samples are analyzed, and the cannabinoid recovery corresponds to the sum of the added and naturally occurring amounts (entries SV05-07, SV14, SV16, SV27, SV30, SV31, SV35, SV36, and SV40). Chaiwangrach et al. [192] (entry SV16) used enriched cannabinoid mixtures prepared from cannabis samples for accuracy studies instead of pure standards.
When reported, accuracy was calculated using Equation (2), where “CD” is the concentration determined, and “CA” is the known concentration. For standard addition methodology, recovery was determined using Equation (3) where “CS” is the concentration determined for the standard added plus the endogenous amount of the cannabinoid (spiked sample), “CU” is the determined endogenous amount in the sample (unspiked sample), and “CA” is the known concentration of the standard solution added [99,103,111,133].
A smaller subset of studies reported accuracy as bias % (entries SV08, SV09, SV25, SV34, SV37, and SV50), calculated using the Equation (4) [169,243] or as mean relative error % (entry SV38). Furthermore, some studies evaluated accuracy/trueness using a total error approach, with relative bias, accuracy profiles, and β-expectation tolerance intervals (entries SV17 and SV18) [173,174,175]:
Accuracy% = CD/CA ∗ 100
Accuracy% = (CS − CU)/CA ∗ 100
Bias% = (CD − CA)/CA ∗ 100
As observed with precision, the AOAC SMPR® 2019.003 guideline recommends limits as a function of cannabinoid concentration (% on dry weight basis): 0.05–0.5%: 85–118%; >0.5–5%: 90–111%; 5–35% (just for CBD and CBDA): 95–105% for recovery experiments using the whole procedure (limits used by entry SV30). Some papers used AOAC appendix F limits for recovery: 0.1%: 90–107% (entry SV29) and 0.01%: 80–110% (entry SV28). ICH M10 establishes that accuracy should be within ±15% of the nominal concentration at any level of the linear range, except at the LOQ, which allows variation within ±20% (limits used by entries SV09 and SV38). Some authors established limits of 80–120% (entries SV07, SV12, SV37, and SV47), 90–110% (entry SV10), 80–125% (entry SV13) for recovery and ≤20% (entry SV25) or ≤15% (entry SV08, and SV34) for % bias or error. Song et al. [184] (entry 21) used 80–120% for quality control at low concentration and 85–115% for medium and high concentration controls. The widest interval deemed acceptable by the authors was 70–120% (entry SV26), as described in SANTE guideline as acceptable during initial validation stages.

5.4. Lower Range Limits

When the analysis requires the analytical procedure to operate close to its lower range, establishing a limit of detection (LOD) or detection limit (DL), and especially a limit of quantification (LOQ) or quantitation limit (QL), is paramount. LOD represents the lowest concentration of an analyte that can be reliably detected, though it remains too small to be measured with certainty under the specified experimental conditions. Conversely, LOQ is the lowest concentration of an analyte that can be quantitatively measured with a previously defined level of acceptable precision and accuracy [284]. They can be estimated using different methods:
  • Based on signal-to-noise ratio (S/N).
The S/N is determined by comparing signals from samples containing low concentrations of the analyte to signals from blank samples. Alternatively, the peak-to-peak noise amplitude can be measured around the analyte retention time and compared with the analyte signal height. A S/N of at least three and ten is considered suitable for estimating LOD and LOQ, respectively [218,288]. Twenty-four papers reported these S/N ratios to assess LOD and LOQ. In one case, LOD was also established as S/N ≥ 2 [218] (entry SV26).
  • Based on visual evaluation/Based on accuracy and precision at lower range limits.
The limits are found through analysis of known low concentration samples, identifying the lowest level at which the analyte can be reliably detected or quantitated. In these cases, LOQ can be directly validated by accuracy and precision measurements [164,172,208,244] (entries SV11, SV34, SV38, and SV41). In fact, as mentioned before, the ICH M10 guideline recommends that one of the concentrations used during precision and accuracy validation should be the LOQ [289]. Carvalho et al. [151] (entry SV32) established the LOD as the lowest concentration that yielded a signal greater than noise with RSD ≤ 10%, while adopting LOQ as the lowest concentration of the calibration curve. Likewise, Song et al. [168,182] (entries SV19 and SV23) also established LOQ as the lowest concentration of the linear range. Meanwhile, Chan [164] (entry SV41) reported LOD as the lowest concentration tested even though lower concentrations were likely detected, and Barhdadi et al. [173] (entry SV18) simply established LOQ as the lowest validated concentration level. In addition, entries SV34 and SV41 also evaluated the S/N of the signals obtained in these experiments. Although some papers did not directly specify the method used to determine LOQ, they used this concentration for precision and accuracy experiments (e.g., [82] entry SV09).
  • Based on standard deviation of a linear response and a slope.
The lower limits can also be estimated by applying Equations (5) and (6), where “σ” represents the standard deviation of the response and “S” the slope of the calibration curve. In the analyzed papers, when specified, “σ” was calculated as follows: (i) standard deviation of the blank (entries SV05 and SV50); (ii) standard deviation of the lowest linear concentration (entry SV02); standard deviation of the lowest response compound that yielded the most conservative value (for LOD) (entry SV25); (iii) standard deviation of the slope (entry SV12); (iv) standard deviation of y-intercepts (entry SV28); and (v) residual standard deviation of the regression line (entries SV06, SV07, and SV36). Furthermore, in other works, the type of standard deviation “σ” was not specified (entries SV10, SV14, SV29, SV31, SV33, SV40, and SV44). ICH Q2 describes “σ” as the standard deviation of (i), (iv), and (v) and recommends that when the estimation is based on a calibration curve, it should be constructed with samples with analyte concentrations that fall within the lower limits range. When LOD and LOQ are obtained by estimation from analytical curves, ICH also recommends further validation by analyzing an appropriate number of samples that are known to be at or near the LOD and LOQ [288]. In some studies, experiments to evaluate the S/N ratio (entries SV10, SV12, SV25, and SV31) or to evaluate accuracy and precision (entries SV28, SV33, and SV50) were also performed to verify the estimated values:
LOD = 3.3σ/S
LOQ = 10σ/S

5.5. General Matrix Interference Considerations

When working with a new matrix, two questions must be addressed: (i) How effectively can the analytes be extracted from the sample matrix? (ii) Do the co-extracted components of the matrix interfere with the detection and quantification of the analytes?
As discussed in Section 4.1, careful selection of HPLC conditions is essential to ensure method specificity and achieve good resolution between the cannabinoids of interest, as well as between them and other matrix interferences. Additionally, general strategies to mitigate matrix effects during analysis can be employed, such as adjusting the retention time of the analytes to avoid the solvent front, where polar and non-retained compounds elute, and the end of the chromatographic gradient, where a higher concentration of organic solvent leads to the elution of the strongly retained nonpolar compounds [303,304].
Furthermore, understanding the impact of the sample matrix on analyte recovery is crucial for developing extraction methodologies. This knowledge will help elucidate how incorporating clean-up steps—if needed to mitigate matrix effects during analysis—will influence overall efficiency. These studies are particularly important when multiple analyte quantifications are targeted, as extraction may impact each analyte differently. For example, Dawson et al. [305] studied the impact of changes in chocolate matrices on cannabinoid extraction efficiency and, consequently, quantification using the same extraction protocol. They reported that the extent of interference from the chocolate matrix on cannabinoid analysis is influenced by both the quantity of chocolate present and the chemical structure of the cannabinoid analyte. Increased amounts of chocolate in the sample correlate with decreased percent recovery of the analyte, and this effect is much more pronounced as cannabinoid polarity decreases.
When analyzing various oil samples, it is essential to consider the distinct properties of different carrier oils, especially if they are also used as solvents during cannabinoid extraction from the cannabis plant, as these factors can influence extraction efficiency and matrix complexity [256,306]. Given that several papers reviewed employed surrogate oils for the validation of these preparations, it is crucial to conduct proper matrix validation studies to ensure that the selected surrogate matrix accurately represents all the oil preparations analyzed. These studies become increasingly important as the matrix complexity increases, such as in cannabis edibles.
Due to the potential interference of the matrix, certain performance parameters should be determined when validating a new analytical procedure for analyte quantification: the matrix effect (ME), the recovery (RE) of the extraction procedure, and the overall process efficiency (PE). These can be calculated using Equations (7)–(9), where A represents the peak response of the analyte in the solvent standard, B represents the peak response of the analyte in the matrix spiked after extraction, and C represents the peak response of the analyte in the matrix spiked pre-extraction [307,308]. The ME calculated in this manner is known as absolute ME. To evaluate MEs across various matrices, it is important to assess relative ME by measuring the peak response of the analyte in different matrix lots that have been spiked after extraction, with the results expressed as RSD (%). Additionally, assessing relative MEs is especially important when employing matrix-matched calibration or background subtraction calibration techniques to mitigate matrix interference [309,310]:
ME (%) = B/A ∗ 100
RE (%) = C/B ∗ 100
PE (%) = C/A ∗ 100

6. Green Analytical Chemistry Considerations

The 12 principles of green analytical chemistry proposed by Gałuszka et al. [311] emphasize the importance of minimizing environmental impact and enhancing analytical efficiency in scientific practices while guaranteeing operator safety. Key practices should include minimizing sample treatment and sample sizes. Researchers should adopt HPLC techniques that promote faster and more efficient cannabinoid separation while minimizing waste production. For example, regarding column optimization, researchers should consider reducing the size (diameter and length) of the columns, using core-shell particles, employing columns with smaller particle sizes, and, if possible, adopting more polar reversed-phase columns instead of C18 [312,313].
In the discussed reversed-phase HPLC analytical protocols, ACN and MeOH were used as the preferred organic modifiers in all referenced papers. However, their toxic and flammable nature presents significant hazards. ACN can be metabolized in the liver into toxic compounds such as hydrogen cyanide and formaldehyde, and chronic exposure to it can lead to adverse health effects. In addition, even low-level exposure to MeOH, whether through inhalation or skin contact, can harm the nervous system, liver, and kidneys [313]. Therefore, it is of primary importance to seek alternative organic solvents for mobile phase to promote a greener LC separation of cannabinoids.
Regarding the extraction of cannabinoids from various matrices, the most commonly used technique in the reviewed papers is UAE. This method is considered a green extraction technique due to its enhanced efficiency and reduced solvent consumption. Additionally, UAE shortens extraction time, lowers analysis costs, and minimizes the potential degradation of cannabinoids. However, the use of UAE should be paired with green solvents, such as EtOH, which is commonly used in most protocols involving plant material matrices, along with MeOH. Researchers should aim to avoid solvents like hexane, chloroform, acetone, and petroleum ether due to their toxicity and environmental impact. The increasing application of DESs in the extraction of natural products, particularly cannabinoids, shows promise for developing more sustainable methodologies due to their biodegradability, non-toxicity, and recyclability.
To assist researchers in evaluating the environmental impact of their analytical methods, several tools are available in the literature that can be utilized [314,315,316,317,318,319,320]. For example, the Analytical GREEnness calculator translates each of the 12 principles of green analytical chemistry into scores ranging from 0 to 1. Values close to 1, represented by a dark green color, indicate that the evaluated procedure is more environmentally friendly. Additionally, the width of each segment in the resulting pictogram reflects the importance of its corresponding principle [318].

7. Materials and Methods

The literature search was performed up to 6 January 2025, across three online databases: PubMed, Web of Science, and Scopus, to include all indexed papers published from 2022 to 2024. The search terms were categorized into three groups (1st group: “Cannabis”, “THC”, “CBD”, “Hemp”; 2nd group: “HPLC”, “LC”, “UPLC”, UHPLC”, “Chromatography”; 3rd group: “UV”, “DAD”, “PDA”, “Diode”, “Photodiode”, “Ultraviolet”). The search utilized Boolean operators to ensure results included at least one term from each group. When permitted by the search engine, the Boolean operator “Asterisk*” was applied before and after specific terms, such as “*cannabi*” and “*LC”, to capture additional variations of the keywords. For PubMed and Web of Science, the search was performed to include “All Fields” of the database, while in Scopus it was confined to the “Title”, “Abstract”, and “Keywords”. A second, less restrictive search in Scopus (only with terms from the first two groups) was also performed (Figure 4). After removing duplicates, a total of 1081 studies were identified. The initial screening involved excluding papers not written in English and eliminating reviews, book chapters, conference papers, and grey literature. This process resulted in the selection of 956 entries for title and abstract analysis, ultimately narrowing the list down to 172 papers for full evaluation. A study was considered eligible if it was original experimental research that included analytical HPLC-UV analysis of phytocannabinoids, resulting in 146 papers. Upon reviewing the selected papers and relevant review articles, an additional 23 papers were considered eligible to this review, leading to a total of 169 papers (included in the HPLC-UV section, Table 1, Table 2 and Tables S2 and S3).
For the extraction section, a total of 148 manuscripts were analyzed and organized in Table S1 according to the matrices studied and the details of the protocol and extraction techniques employed. Twenty-one manuscripts were excluded from this analysis because important details regarding the extraction method were missing, a dilute-and-shoot protocol was used without actual extraction of cannabinoids from samples, or standards were used instead. Concerning the method-validation section, only papers that reported at least two of the three validation parameters discussed (precision, accuracy, and lower limits) were included in Table S4 (57 papers).

8. Conclusions and Future Directions

The growing market for medicinal cannabis and cannabis-based products requires a corresponding enhancement in rigorous quality control, standardization, and innovation to meet regulatory standards and consumer expectations. Reliable quantification of cannabinoids in these diverse cannabis-related products depends on key processes, such as extraction methodologies, sample preparation, and thorough analytical method validation.
In the pharmaceutical industry, the effects of matrix interference on analyte extraction and detection are well-documented, resulting in highly accurate and precise analyses. However, similar research in cannabis analytical testing remains limited, and specific molecular interactions that could impact accuracy are still largely unexplored. Matrix interference in these products is complex, arising from variations in chemovars, differences in cannabinoid extraction procedures, and the effects of food additives and drug formulations on cannabinoid quantification. Furthermore, introducing clean-up steps during extractions can affect extraction efficiency and have varying impacts on structurally distinct cannabinoids. To effectively standardize cannabis analytical methods, scientific efforts must be grounded in comprehensive studies of both the analytes and the matrices involved. Advancements in cannabis testing are essential for the industry to progress beyond the black market and ensure long-term stability.
In the reviewed papers, the most prominent extraction technologies used for cannabinoids have been SFE, UAE, and DES. UAE is the preferred technique across a diverse range of matrices and has been successfully applied to the extraction of cannabinoids from complex sources such as formulated products, plasma, and tissue samples. While DESs show promise for future green extractions, their applicability have thus far been limited to cannabis plants and oil matrices.
Analytical chromatographic separations are still primarily performed using C18 reversed-phase columns. However, in recent years, researchers have begun investigating a variety of stationary phases, including normal phases with NH2 adsorbents and polysaccharide-based chiral stationary phases. The analysis of chiral cannabinoids is becoming increasingly important, as different enantiomers and diastereomers may have distinct pharmacological effects.
One of the most significant challenges in the detection and quantification of cannabinoids is the standardization of analytical methods and their associated validation parameters across laboratories to ensure consistent and reliable results on a global scale. To achieve this, the scientific community needs to actively participate in this conversation by reporting in published papers how analytical method validation was performed, specifying how each parameter was calculated, and detailing the protocols involved, including the preparation of quality controls and the determination of matrix effects, analyte recovery, and overall process efficiency. The present review highlights the lack of consistency in the presented validation parameters.
Guidelines should be established for each cannabis-related product to adequately address questions regarding sample preparation, analytical methods, and validation parameters necessary for establishing robust standardized methods, while also investigating the potential for using green techniques, or more environmentally friendly methods.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ph18060786/s1, Table S1: Extraction methods for the analysis of cannabinoids in different matrices; Table S2: Summary of LC parameters used in gradient chromatographic runs for detection of 1 or 2 cannabinoids; Table S3: Summary of LC parameters used in isocratic chromatographic runs for detection of 1 or 2 cannabinoids; Table S4: Summary of validation parameters: Linearity, Precision, Accuracy, LOD, and LOQ.

Author Contributions

Conceptualization, E.M.P.S. and C.J.A.R.; methodology, A.V. and C.J.A.R.; writing—original draft preparation, E.M.P.S., A.V., and C.J.A.R.; writing—review and editing, E.M.P.S., A.V., A.M., and C.J.A.R.; supervision, C.J.A.R.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Author Carlos J. A. Ribeiro was employed by the company Avextra Portugal SA. The remaining authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ACAcidic cannabinoids
ACNAcetonitrile
AOACAssociation of Official Analytical Collaboration
C18Octadecyl
C8Octyl
CBCannabinoid receptor
CBCCannabichromene
CBCACannabichromenic acid
CBCOCannabichromeorcin
CBCTCannabicitran
CBCVCannabichromevarin
CBCVACannabichromivarinic acid
CBDCannabidiol
CBDACannabidiolic acid
CBDA-MECannabidiolic acid methyl ester
CBDPCannabidiphorol
CBDVCannabidivarin
CBDVACannabidivarinic acid
CBECannabielsoin
CBEACannabielsoic acid
CBGCannabigerol
CBGACannabigerolic acid
CBGQACannabigerol quinone acid
CBGVCannabigerovarin
CBGVACannabigerovarinic acid
CBLCannabicyclol
CBLACannabicyclolic acid
CBNCannabinol
CBNACannabinolic acid
CBTCannabitriol
CVCoefficient of variation
CINVChemotherapy-induced nausea and vomiting
DADDiode array detector
DESDeep eutectic solvent
DMDynamic maceration
DoEDesign of Experiments
DSLMEDispersive solid–liquid microextraction
Δ4(8)-iso-THCΔ4(8)-iso-tetrahydrocannabinol
Δ8-iso-THCΔ8-iso-tetrahydrocannabinol
Δ8-THC8-tetrahydrocannabinol
Δ9-THC9-tetrahydrocannabinol
Δ8-THCA8-tetrahydrocannabinolic acid
DHCDihydrocannabinol
ESEutectic solvent
EtOHEthanol
FAFormic acid
GCGas chromatography
HBAHydrogen bond acceptor
HBDHydrogen bond donor
HPLCHigh-performance liquid chromatography
ICHInternational Council of Harmonisation
LCLiquid chromatography
LODLimit of detection
LOQLimit of quantification
MeOHMethanol
MSMass spectrometry
MTBEMethyl tert-butyl ether
MWDMultiple wavelength detector
NCNeutral cannabinoids
NRNot reported
PFPPentafluorophenyl
QCQuality Control
RPReversed-phase
RSDRelative standard deviation
RTRoom temperature
scCO2Supercritical carbon dioxide
SESoxhlet extraction
SFESupercritical fluid extraction
SLESolid–liquid extraction
S/NSignal to noise ratio
SPESolid phase extraction
THCA9-tetrahydrocannabinolic acid
THCB9-tetrahydrocannabutol
THCH9-tetrahydrocannabihexol
THCP9-tetrahydrocannabiphorol
THCV9-Tetrahydrocannabivarin
THCVA9-Tetrahydrocannabivarinic acid
TUVTunable ultraviolet
UAEUltrasound-assisted extraction
USUltrasound
US-LLEUltrasound-assisted liquid–liquid extraction
US-SLEUltrasound-assisted solid–liquid extraction
UVUltraviolet
VWDVariable wavelength detector

References

  1. Berning, A.; Compton, R.; Wochinger, K. Results of the 2013–2014 National Roadside Study of Alcohol and Drug Use by Drivers [Traffic Safety Facts]; United States. Department of Transportation. National Highway Traffic Safety Administration: Washington, DC, USA, 2015. [Google Scholar]
  2. Farag, S.; Kayser, O. The Cannabis Plant: Botanical Aspects. In Handbook of Cannabis and Related Pathologies; Preedy, V.R., Ed.; Academic Press: San Diego, CA, USA, 2017; pp. 3–12. ISBN 978-0-12-800756-3. [Google Scholar]
  3. 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]
  4. Ferber, S.G.; Namdar, D.; Hen-Shoval, D.; Eger, G.; Koltai, H.; Shoval, G.; Shbiro, L.; Weller, A. The “Entourage Effect”: Terpenes Coupled with Cannabinoids for the Treatment of Mood Disorders and Anxiety Disorders. Curr. Neuropharmacol. 2020, 18, 87–96. [Google Scholar] [CrossRef] [PubMed]
  5. Bilbao, A.; Spanagel, R. Medical Cannabinoids: A Pharmacology-Based Systematic Review and Meta-Analysis for All Relevant Medical Indications. BMC Med. 2022, 20, 259. [Google Scholar] [CrossRef] [PubMed]
  6. Sznitman, S.R.; Zolotov, Y. Cannabis for Therapeutic Purposes and Public Health and Safety: A Systematic and Critical Review. Int. J. Drug Policy 2015, 26, 20–29. [Google Scholar] [CrossRef]
  7. Fiorini, D.; Molle, A.; Nabissi, M.; Santini, G.; Benelli, G.; Maggi, F. Valorizing Industrial Hemp (Cannabis sativa L.) by-Products: Cannabidiol Enrichment in the Inflorescence Essential Oil Optimizing Sample Pre-Treatment Prior to Distillation. Ind. Crops Prod. 2019, 128, 581–589. [Google Scholar] [CrossRef]
  8. Duchateau, C.; Stévigny, C.; Waeytens, J.; Deconinck, E. Chromatographic and Spectroscopic Analyses of Cannabinoids: A Narrative Review Focused on Cannabis Herbs and Oily Products. Molecules 2025, 30, 490. [Google Scholar] [CrossRef]
  9. Inamassu, C.H.; Raspini E Silva, L.; Marchioni, C. Recent Advances in the Chromatographic Analysis of Endocannabinoids and Phytocannabinoids in Biological Samples. J. Chromatogr. A 2024, 1732, 465225. [Google Scholar] [CrossRef]
  10. Madden, O.; Walshe, J.; Kishore Patnala, P.; Barron, J.; Meaney, C.; Murray, P. Phytocannabinoids—An Overview of the Analytical Methodologies for Detection and Quantification of Therapeutically and Recreationally Relevant Cannabis Compounds. Crit. Rev. Anal. Chem. 2023, 53, 211–231. [Google Scholar] [CrossRef]
  11. Antunes, M.; Barroso, M.; Gallardo, E. Analysis of Cannabinoids in Biological Specimens: An Update. Int. J. Environ. Res. Public Health 2023, 20, 2312. [Google Scholar] [CrossRef]
  12. Stefkov, G.; Cvetkovikj Karanfilova, I.; Stoilkovska Gjorgievska, V.; Trajkovska, A.; Geskovski, N.; Karapandzova, M.; Kulevanova, S. Analytical Techniques for Phytocannabinoid Profiling of Cannabis and Cannabis-Based Products—A Comprehensive Review. Molecules 2022, 27, 975. [Google Scholar] [CrossRef]
  13. Deidda, R.; Dispas, A.; De Bleye, C.; Hubert, P.; Ziemons, É. Critical Review on Recent Trends in Cannabinoid Determination on Cannabis Herbal Samples: From Chromatographic to Vibrational Spectroscopic Techniques. Anal. Chim. Acta 2022, 1209, 339184. [Google Scholar] [CrossRef]
  14. Micalizzi, G.; Vento, F.; Alibrando, F.; Donnarumma, D.; Dugo, P.; Mondello, L. Cannabis sativa L.: A Comprehensive Review on the Analytical Methodologies for Cannabinoids and Terpenes Characterization. J. Chromatogr. A 2021, 1637, 461864. [Google Scholar] [CrossRef] [PubMed]
  15. Pourseyed Lazarjani, M.; Torres, S.; Hooker, T.; Fowlie, C.; Young, O.; Seyfoddin, A. Methods for Quantification of Cannabinoids: A Narrative Review. J. Cannabis Res. 2020, 2, 35. [Google Scholar] [CrossRef]
  16. Karschner, E.L.; Swortwood-Gates, M.J.; Huestis, M.A. Identifying and Quantifying Cannabinoids in Biological Matrices in the Medical and Legal Cannabis Era. Clin. Chem. 2020, 66, 888–914. [Google Scholar] [CrossRef]
  17. Citti, C.; Braghiroli, D.; Vandelli, M.A.; Cannazza, G. Pharmaceutical and Biomedical Analysis of Cannabinoids: A Critical Review. J. Pharm. Biomed. Anal. 2018, 147, 565–579. [Google Scholar] [CrossRef] [PubMed]
  18. Leghissa, A.; Hildenbrand, Z.L.; Schug, K.A. A Review of Methods for the Chemical Characterization of Cannabis Natural Products. J. Sep. Sci. 2018, 41, 398–415. [Google Scholar] [CrossRef]
  19. Nahar, L.; Gavril, G.-L.; Sarker, S.D. Application of Gas Chromatography in the Analysis of Phytocannabinoids: An Update (2020–2023). Phytochem. Anal. 2023, 34, 903–924. [Google Scholar] [CrossRef] [PubMed]
  20. Nahar, L.; Guo, M.; Sarker, S.D. Gas Chromatographic Analysis of Naturally Occurring Cannabinoids: A Review of Literature Published during the Past Decade. Phytochem. Anal. 2020, 31, 135–146. [Google Scholar] [CrossRef]
  21. Nahar, L.; Onder, A.; Sarker, S.D. A Review on the Recent Advances in HPLC, UHPLC and UPLC Analyses of Naturally Occurring Cannabinoids (2010–2019). Phytochem. Anal. 2020, 31, 413–457. [Google Scholar] [CrossRef]
  22. Felletti, S.; De Luca, C.; Buratti, A.; Bozza, D.; Cerrato, A.; Capriotti, A.L.; Laganà, A.; Cavazzini, A.; Catani, M. Potency Testing of Cannabinoids by Liquid and Supercritical Fluid Chromatography: Where We Are, What We Need. J. Chromatogr. A 2021, 1651, 462304. [Google Scholar] [CrossRef]
  23. Nicolaou, A.G.; Christodoulou, M.C.; Stavrou, I.J.; Kapnissi-Christodoulou, C.P. Analysis of Cannabinoids in Conventional and Alternative Biological Matrices by Liquid Chromatography: Applications and Challenges. J. Chromatogr. A 2021, 1651, 462277. [Google Scholar] [CrossRef] [PubMed]
  24. Nahar, L.; Chaiwut, P.; Sangthong, S.; Theansungnoen, T.; Sarker, S.D. Progress in the Analysis of Phytocannabinoids by HPLC and UPLC (or UHPLC) during 2020–2023. Phytochem. Anal. 2024, 35, 927–989. [Google Scholar] [CrossRef] [PubMed]
  25. ElSohly, H.N.; ElSohly, M.A. Marijuana Smoke Condensate: Chemistry and Pharmacology. In Marijuana and the Cannabinoids; Springer: Berlin/Heidelberg, Germany, 2007; pp. 67–96. [Google Scholar]
  26. Castillo, P.E.; Younts, T.J.; Chávez, A.E.; Hashimotodani, Y. Endocannabinoid Signaling and Synaptic Function. Neuron 2012, 76, 70–81. [Google Scholar] [CrossRef]
  27. Roque-Bravo, R.; Silva, R.S.; Malheiro, R.F.; Carmo, H.; Carvalho, F.; da Silva, D.D.; Silva, J.P. Synthetic Cannabinoids: A Pharmacological and Toxicological Overview. Annu. Rev. Pharmacol. Toxicol. 2023, 63, 187–209. [Google Scholar] [CrossRef] [PubMed]
  28. Radwan, M.M.; Chandra, S.; Gul, S.; ElSohly, M.A. Cannabinoids, Phenolics, Terpenes and Alkaloids of Cannabis. Molecules 2021, 26, 2774. [Google Scholar] [CrossRef]
  29. Wang, M.; Wang, Y.-H.; Avula, B.; Radwan, M.M.; Wanas, A.S.; Van Antwerp, J.; Parcher, J.F.; ElSohly, M.A.; Khan, I.A. Decarboxylation Study of Acidic Cannabinoids: A Novel Approach Using Ultra-High-Performance Supercritical Fluid Chromatography/Photodiode Array-Mass Spectrometry. Cannabis Cannabinoid Res. 2016, 1, 262–271. [Google Scholar] [CrossRef]
  30. Morales, P.; Hurst, D.P.; Reggio, P.H. Molecular Targets of the Phytocannabinoids: A Complex Picture. In Phytocannabinoids: Unraveling the Complex Chemistry and Pharmacology of Cannabis sativa; Kinghorn, A.D., Falk, H., Gibbons, S., Kobayashi, J., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 103–131. ISBN 978-3-319-45541-9. [Google Scholar]
  31. Pagano, C.; Navarra, G.; Coppola, L.; Avilia, G.; Bifulco, M.; Laezza, C. Cannabinoids: Therapeutic Use in Clinical Practice. Int. J. Mol. Sci. 2022, 23, 3344. [Google Scholar] [CrossRef]
  32. Weizman, L.; Dayan, L.; Brill, S.; Nahman-Averbuch, H.; Hendler, T.; Jacob, G.; Sharon, H. Cannabis Analgesia in Chronic Neuropathic Pain Is Associated with Altered Brain Connectivity. Neurology 2018, 91, e1285–e1294. [Google Scholar] [CrossRef]
  33. Van Amerongen, G.; Kanhai, K.; Baakman, A.C.; Heuberger, J.; Klaassen, E.; Beumer, T.L.; Strijers, R.L.M.; Killestein, J.; Van Gerven, J.; Cohen, A.; et al. Effects on Spasticity and Neuropathic Pain of an Oral Formulation of Δ9-Tetrahydrocannabinol in Patients with Progressive Multiple Sclerosis. Clin. Ther. 2018, 40, 1467–1482. [Google Scholar] [CrossRef]
  34. Bisaga, A.; Sullivan, M.A.; Glass, A.; Mishlen, K.; Pavlicova, M.; Haney, M.; Raby, W.N.; Levin, F.R.; Carpenter, K.M.; Mariani, J.J.; et al. The Effects of Dronabinol during Detoxification and the Initiation of Treatment with Extended Release Naltrexone. Drug Alcohol Depend. 2015, 154, 38–45. [Google Scholar] [CrossRef]
  35. Lofwall, M.R.; Babalonis, S.; Nuzzo, P.A.; Elayi, S.C.; Walsh, S.L. Opioid Withdrawal Suppression Efficacy of Oral Dronabinol in Opioid Dependent Humans. Drug Alcohol Depend. 2016, 164, 143–150. [Google Scholar] [CrossRef] [PubMed]
  36. Childs, E.; Lutz, J.A.; De Wit, H. Dose-Related Effects of Delta-9-THC on Emotional Responses to Acute Psychosocial Stress. Drug Alcohol Depend. 2017, 177, 136–144. [Google Scholar] [CrossRef] [PubMed]
  37. Zabik, N.L.; Rabinak, C.A.; Peters, C.A.; Iadipaolo, A. Cannabinoid Modulation of Corticolimbic Activation during Extinction Learning and Fear Renewal in Adults with Posttraumatic Stress Disorder. Neurobiol. Learn. Mem. 2023, 201, 107758. [Google Scholar] [CrossRef]
  38. Carley, D.W.; Prasad, B.; Reid, K.J.; Malkani, R.; Attarian, H.; Abbott, S.M.; Vern, B.; Xie, H.; Yuan, C.; Zee, P.C. Pharmacotherapy of Apnea by Cannabimimetic Enhancement, the PACE Clinical Trial: Effects of Dronabinol in Obstructive Sleep Apnea. Sleep 2018, 41, zsx184. [Google Scholar] [CrossRef] [PubMed]
  39. DeJesus, E.; Rodwick, B.M.; Bowers, D.; Cohen, C.J.; Pearce, D. Use of Dronabinol Improves Appetite and Reverses Weight Loss in HIV/AIDS-Infected Patients. J. Int. Assoc. Provid. AIDS Care 2007, 6, 95–100. [Google Scholar] [CrossRef]
  40. Bathula, P.P.; Maciver, M.B. Cannabinoids in Treating Chemotherapy-Induced Nausea and Vomiting, Cancer-Associated Pain, and Tumor Growth. Int. J. Mol. Sci. 2023, 25, 74. [Google Scholar] [CrossRef]
  41. Yau, G.T.Y.; Tai, W.; Arnold, J.C.; Chan, H.-K.; Kwok, P.C.L. Cannabidiol for the Treatment of Brain Disorders: Therapeutic Potential and Routes of Administration. Pharm. Res. 2023, 40, 1087–1114. [Google Scholar] [CrossRef]
  42. Singh, K.; Bhushan, B.; Chanchal, D.K.; Sharma, S.K.; Rani, K.; Yadav, M.K.; Porwal, P.; Kumar, S.; Sharma, A.; Virmani, T.; et al. Emerging Therapeutic Potential of Cannabidiol (CBD) in Neurological Disorders: A Comprehensive Review. Behav. Neurol. 2023, 2023, 8825358. [Google Scholar] [CrossRef]
  43. Rapin, L.; Gamaoun, R.; El Hage, C.; Arboleda, M.F.; Prosk, E. Cannabidiol Use and Effectiveness: Real-World Evidence from a Canadian Medical Cannabis Clinic. J. Cannabis Res. 2021, 3, 19. [Google Scholar] [CrossRef]
  44. Scheffer, I.E.; Halford, J.J.; Miller, I.; Nabbout, R.; Sanchez-Carpintero, R.; Shiloh-Malawsky, Y.; Wong, M.; Zolnowska, M.; Checketts, D.; Dunayevich, E.; et al. Add-on Cannabidiol in Patients with Dravet Syndrome: Results of a Long-Term Open-Label Extension Trial. Epilepsia 2021, 62, 2505–2517. [Google Scholar] [CrossRef]
  45. Patel, A.D.; Mazurkiewicz-Bełdzińska, M.; Chin, R.F.; Gil-Nagel, A.; Gunning, B.; Halford, J.J.; Mitchell, W.; Scott Perry, M.; Thiele, E.A.; Weinstock, A.; et al. Long-term Safety and Efficacy of Add-On Cannabidiol in Patients with Lennox–Gastaut Syndrome: Results of a Long-term Open-label Extension Trial. Epilepsia 2021, 62, 2228–2239. [Google Scholar] [CrossRef]
  46. Thiele, E.A.; Bebin, E.M.; Bhathal, H.; Jansen, F.E.; Kotulska, K.; Lawson, J.A.; O’Callaghan, F.J.; Wong, M.; Sahebkar, F.; Checketts, D.; et al. Add-on Cannabidiol Treatment for Drug-Resistant Seizures in Tuberous Sclerosis Complex: A Placebo-Controlled Randomized Clinical Trial. JAMA Neurol. 2021, 78, 285. [Google Scholar] [CrossRef]
  47. Leehey, M.A.; Liu, Y.; Hart, F.; Epstein, C.; Cook, M.; Sillau, S.; Klawitter, J.; Newman, H.; Sempio, C.; Forman, L.; et al. Safety and Tolerability of Cannabidiol in Parkinson Disease: An Open Label, Dose-Escalation Study. Cannabis Cannabinoid Res. 2020, 5, 326–336. [Google Scholar] [CrossRef]
  48. Solowij, N.; Broyd, S.J.; Beale, C.; Prick, J.-A.; Greenwood, L.; Van Hell, H.; Suo, C.; Galettis, P.; Pai, N.; Fu, S.; et al. Therapeutic Effects of Prolonged Cannabidiol Treatment on Psychological Symptoms and Cognitive Function in Regular Cannabis Users: A Pragmatic Open-Label Clinical Trial. Cannabis Cannabinoid Res. 2018, 3, 21–34. [Google Scholar] [CrossRef]
  49. Bergamaschi, M.M.; Queiroz, R.H.C.; Chagas, M.H.N.; De Oliveira, D.C.G.; De Martinis, B.S.; Kapczinski, F.; Quevedo, J.; Roesler, R.; Schröder, N.; Nardi, A.E.; et al. Cannabidiol Reduces the Anxiety Induced by Simulated Public Speaking in Treatment-Naïve Social Phobia Patients. Neuropsychopharmacology 2011, 36, 1219–1226. [Google Scholar] [CrossRef]
  50. Berger, M.; Li, E.; Rice, S.; Davey, C.G.; Ratheesh, A.; Adams, S.; Jackson, H.; Hetrick, S.; Parker, A.; Spelman, T.; et al. Cannabidiol for Treatment-Resistant Anxiety Disorders in Young People: An Open-Label Trial. J. Clin. Psychiatry 2022, 83, 21m14130. [Google Scholar] [CrossRef] [PubMed]
  51. Leweke, F.M.; Piomelli, D.; Pahlisch, F.; Muhl, D.; Gerth, C.W.; Hoyer, C.; Klosterkötter, J.; Hellmich, M.; Koethe, D. Cannabidiol Enhances Anandamide Signaling and Alleviates Psychotic Symptoms of Schizophrenia. Transl. Psychiatry 2012, 2, e94. [Google Scholar] [CrossRef]
  52. McGuire, P.; Robson, P.; Cubala, W.J.; Vasile, D.; Morrison, P.D.; Barron, R.; Taylor, A.; Wright, S. Cannabidiol (CBD) as an Adjunctive Therapy in Schizophrenia: A Multicenter Randomized Controlled Trial. Am. J. Psychiatry 2018, 175, 225–231. [Google Scholar] [CrossRef] [PubMed]
  53. Hurd, Y.L.; Spriggs, S.; Alishayev, J.; Winkel, G.; Gurgov, K.; Kudrich, C.; Oprescu, A.M.; Salsitz, E. Cannabidiol for the Reduction of Cue-Induced Craving and Anxiety in Drug-Abstinent Individuals with Heroin Use Disorder: A Double-Blind Randomized Placebo-Controlled Trial. Am. J. Psychiatry 2019, 176, 911–922. [Google Scholar] [CrossRef]
  54. Shannon, S.; Lewis, N.; Lee, H.; Hughes, S. Cannabidiol in Anxiety and Sleep: A Large Case Series. Perm. J. 2019, 23, 18–41. [Google Scholar] [CrossRef]
  55. Jones, É.; Vlachou, S. A Critical Review of the Role of the Cannabinoid Compounds Δ9-Tetrahydrocannabinol (Δ9-THC) and Cannabidiol (CBD) and Their Combination in Multiple Sclerosis Treatment. Molecules 2020, 25, 4930. [Google Scholar] [CrossRef] [PubMed]
  56. Keating, G.M. Delta-9-Tetrahydrocannabinol/Cannabidiol Oromucosal Spray (Sativex®): A Review in Multiple Sclerosis-Related Spasticity. Drugs 2017, 77, 563–574. [Google Scholar] [CrossRef]
  57. Sukpiriyagul, A.; Chartchaiyarerk, R.; Tabtipwon, P.; Smanchat, B.; Prommas, S.; Bhamarapravatana, K.; Suwannarurk, K. Oral Tetrahydrocannabinol (THC):Cannabinoid (CBD) Cannabis Extract Adjuvant for Reducing Chemotherapy-Induced Nausea and Vomiting (CINV): A Randomized, Double-Blinded, Placebo-Controlled, Crossover Trial. Int. J. Women’s Health 2023, 15, 1345–1352. [Google Scholar] [CrossRef]
  58. Hanuš, L.O.; Meyer, S.M.; Muñoz, E.; Taglialatela-Scafati, O.; Appendino, G. Phytocannabinoids: A Unified Critical Inventory. Nat. Prod. Rep. 2016, 33, 1357–1392. [Google Scholar] [CrossRef] [PubMed]
  59. Rosenthaler, S.; Pöhn, B.; Kolmanz, C.; Nguyen Huu, C.; Krewenka, C.; Huber, A.; Kranner, B.; Rausch, W.-D.; Moldzio, R. Differences in Receptor Binding Affinity of Several Phytocannabinoids Do Not Explain Their Effects on Neural Cell Cultures. Neurotoxicol. Teratol. 2014, 46, 49–56. [Google Scholar] [CrossRef]
  60. Bonn-Miller, M.O.; Feldner, M.T.; Bynion, T.M.; Eglit, G.M.L.; Brunstetter, M.; Kalaba, M.; Zvorsky, I.; Peters, E.N.; Hennesy, M. A Double-Blind, Randomized, Placebo-Controlled Study of the Safety and Effects of CBN with and without CBD on Sleep Quality. Exp. Clin. Psychopharmacol. 2024, 32, 277–284. [Google Scholar] [CrossRef] [PubMed]
  61. Mancini, A.; Stratigos, A.; Guez, S.; Bourrat, E.; Kiritsi, D.; Samuelov, L.; Mazereeuw-Hautier, J.; Didona, B.; Bodemer, C.; Laimer, M.; et al. INM-755 Cannabinol Cream Demonstrates Anti-Itch Activity in Patients with Epidermolysis Bullosa. In Proceedings of the 12th World Congress on Itch (WCI), Miami, FL, USA, 5–7 November 2023. [Google Scholar]
  62. Zagzoog, A.; Mohamed, K.A.; Kim, H.J.J.; Kim, E.D.; Frank, C.S.; Black, T.; Jadhav, P.D.; Holbrook, L.A.; Laprairie, R.B. In Vitro and in Vivo Pharmacological Activity of Minor Cannabinoids Isolated from Cannabis sativa. Sci. Rep. 2020, 10, 20405. [Google Scholar] [CrossRef]
  63. Formukong, E.A.; Evans, A.T.; Evans, F.J. Analgesic and Antiinflammatory Activity of Constituents of Cannabis sativa L. Inflammation 1988, 12, 361–371. [Google Scholar] [CrossRef]
  64. Appendino, G.; Gibbons, S.; Giana, A.; Pagani, A.; Grassi, G.; Stavri, M.; Smith, E.; Rahman, M.M. Antibacterial Cannabinoids from Cannabis sativa: A Structure-Activity Study. J. Nat. Prod. 2008, 71, 1427–1430. [Google Scholar] [CrossRef]
  65. Borrelli, F.; Fasolino, I.; Romano, B.; Capasso, R.; Maiello, F.; Coppola, D.; Orlando, P.; Battista, G.; Pagano, E.; Di Marzo, V.; et al. Beneficial Effect of the Non-Psychotropic Plant Cannabinoid Cannabigerol on Experimental Inflammatory Bowel Disease. Biochem. Pharmacol. 2013, 85, 1306–1316. [Google Scholar] [CrossRef]
  66. Russo, E.B.; Cuttler, C.; Cooper, Z.D.; Stueber, A.; Whiteley, V.L.; Sexton, M. Survey of Patients Employing Cannabigerol-Predominant Cannabis Preparations: Perceived Medical Effects, Adverse Events, and Withdrawal Symptoms. Cannabis Cannabinoid Res. 2022, 7, 706–716. [Google Scholar] [CrossRef] [PubMed]
  67. Cuttler, C.; Stueber, A.; Cooper, Z.D.; Russo, E. Acute Effects of Cannabigerol on Anxiety, Stress, and Mood: A Double-Blind, Placebo-Controlled, Crossover, Field Trial. Sci. Rep. 2024, 14, 16163. [Google Scholar] [CrossRef] [PubMed]
  68. Abioye, A.; Ayodele, O.; Marinkovic, A.; Patidar, R.; Akinwekomi, A.; Sanyaolu, A. Δ9-Tetrahydrocannabivarin (THCV): A Commentary on Potential Therapeutic Benefit for the Management of Obesity and Diabetes. J. Cannabis Res. 2020, 2, 6. [Google Scholar] [CrossRef] [PubMed]
  69. Dennis, I.; Whalley, B.J.; Stephens, G.J. Effects of Δ9-Tetrahydrocannabivarin on [35S]GTPγS Binding in Mouse Brain Cerebellum and Piriform Cortex Membranes. Br. J. Pharmacol. 2008, 154, 1349–1358. [Google Scholar] [CrossRef]
  70. Bolognini, D.; Costa, B.; Maione, S.; Comelli, F.; Marini, P.; Di Marzo, V.; Parolaro, D.; Ross, R.A.; Gauson, L.A.; Cascio, M.G.; et al. The Plant Cannabinoid Δ9-Tetrahydrocannabivarin Can Decrease Signs of Inflammation and Inflammatory Pain in Mice. Br. J. Pharmacol. 2010, 160, 677–687. [Google Scholar] [CrossRef]
  71. Hill, A.J.; Weston, S.E.; Jones, N.A.; Smith, I.; Bevan, S.A.; Williamson, E.M.; Stephens, G.J.; Williams, C.M.; Whalley, B.J. Δ9-Tetrahydrocannabivarin Suppresses In Vitro Epileptiform and In Vivo Seizure Activity in Adult Rats. Epilepsia 2010, 51, 1522–1532. [Google Scholar] [CrossRef]
  72. Riedel, G.; Fadda, P.; McKillop-Smith, S.; Pertwee, R.G.; Platt, B.; Robinson, L. Synthetic and Plant-Derived Cannabinoid Receptor Antagonists Show Hypophagic Properties in Fasted and Non-Fasted Mice. Br. J. Pharmacol. 2009, 156, 1154–1166. [Google Scholar] [CrossRef]
  73. Smith, G. Weight Loss and Therapeutic Metabolic Effects of Tetrahydrocannabivarin (THCV)-Infused Mucoadhesive Strips. Cannabis 2024, 8, 1. [Google Scholar] [CrossRef]
  74. Englund, A.; Atakan, Z.; Kralj, A.; Tunstall, N.; Murray, R.; Morrison, P. The Effect of Five Day Dosing with THCV on THC-Induced Cognitive, Psychological and Physiological Effects in Healthy Male Human Volunteers: A Placebo-Controlled, Double-Blind, Crossover Pilot Trial. J. Psychopharmacol. 2016, 30, 140–151. [Google Scholar] [CrossRef]
  75. Alves, P.; Amaral, C.; Teixeira, N.; Correia-da-Silva, G. Cannabis sativa: Much More beyond Δ9-Tetrahydrocannabinol. Pharmacol. Res. 2020, 157, 104822. [Google Scholar] [CrossRef]
  76. Pagano, E.; Romano, B.; Iannotti, F.A.; Parisi, O.A.; D’Armiento, M.; Pignatiello, S.; Coretti, L.; Lucafò, M.; Venneri, T.; Stocco, G.; et al. The Non-Euphoric Phytocannabinoid Cannabidivarin Counteracts Intestinal Inflammation in Mice and Cytokine Expression in Biopsies from UC Pediatric Patients. Pharmacol. Res. 2019, 149, 104464. [Google Scholar] [CrossRef] [PubMed]
  77. Hill, A.; Mercier, M.; Hill, T.; Glyn, S.; Jones, N.; Yamasaki, Y.; Futamura, T.; Duncan, M.; Stott, C.; Stephens, G.; et al. Cannabidivarin Is Anticonvulsant in Mouse and Rat. Br. J. Pharmacol. 2012, 167, 1629–1642. [Google Scholar] [CrossRef]
  78. Brodie, M.J.; Czapinski, P.; Pazdera, L.; Sander, J.W.; Toledo, M.; Napoles, M.; Sahebkar, F.; Schreiber, A.; Nezadal, T.; Slonkova, J.; et al. A Phase 2 Randomized Controlled Trial of the Efficacy and Safety of Cannabidivarin as Add-on Therapy in Participants with Inadequately Controlled Focal Seizures. Cannabis Cannabinoid Res. 2021, 6, 528–536. [Google Scholar] [CrossRef]
  79. Hurley, E.N.; Ellaway, C.J.; Johnson, A.M.; Truong, L.; Gordon, R.; Galettis, P.; Martin, J.H.; Lawson, J.A. Efficacy and Safety of Cannabidivarin Treatment of Epilepsy in Girls with Rett Syndrome: A Phase 1 Clinical Trial. Epilepsia 2022, 63, 1736–1747. [Google Scholar] [CrossRef]
  80. Santos Álvarez, I.; Pérez Lloret, P.; González Soriano, J.M.; Pérez Moreno, M. An Approach to the Evaluation of the Potency of Cannabis Resin in Madrid: A Health Hazard? Adicciones 2023, 35, 279–288. [Google Scholar] [CrossRef] [PubMed]
  81. Zeger, V.R.; Bell, D.S.; Herrington, J.S.; Anderson, J.L. Selective Isolation of Pesticides and Cannabinoids Using Polymeric Ionic Liquid-Based Sorbent Coatings in Solid-Phase Microextraction Coupled to High-Performance Liquid Chromatography. J. Chromatogr. A 2022, 1680, 463416. [Google Scholar] [CrossRef]
  82. Carona, A.; Bicker, J.; Fonseca, C.; da Graça Campos, M.; Falcão, A.; Fortuna, A. Development and Validation of an HPLC-DAD Method for the Quantification of Cannabigerol, Cannabidiol, Cannabinol and Cannabichromene in Human Plasma and Mouse Matrices. Analyst 2024, 149, 3815–3827. [Google Scholar] [CrossRef]
  83. Szalata, M.; Dreger, M.; Zielińska, A.; Banach, J. Simple Extraction of Cannabinoids from Female Inflorescences of Hemp (Cannabis sativa L.). Molecules 2022, 27, 5868. [Google Scholar] [CrossRef]
  84. Bartkowiak-Wieczorek, J.; Mądry, E.; Książkiewicz, M.; Winkler-Galicki, J.; Szalata, M.; Szalata, M.; Jiménez, U.E.; Wielgus, K.; Grześkowiak, E.; Słomski, R.; et al. THC-Reduced Cannabis sativa L.—How Does the Solvent Determine the Bioavailability of Cannabinoids Given Orally? Nutrients 2023, 15, 2646. [Google Scholar] [CrossRef]
  85. Schadich, E.; Kaczorová, D.; Béres, T.; Džubák, P.; Hajdúch, M.; Tarkowski, P.; Ćavar Zeljković, S. Secondary Metabolite Profiles and anti-SARS-CoV-2 Activity of Ethanolic Extracts from Nine Genotypes of Cannabis sativa L. Arch. Pharm. 2024, 358, e2400607. [Google Scholar] [CrossRef]
  86. Skala, T.; Kahánková, Z.; Tauchen, J.; Janatová, A.; Kloucek, P.; Hubka, V.; Franková, A. Medical Cannabis Dimethyl Ether, Ethanol and Butane Extracts Inhibit the In Vitro Growth of Bacteria and Dermatophytes Causing Common Skin Diseases. Front. Microbiol. 2022, 13, 953092. [Google Scholar] [CrossRef]
  87. Acquavia, M.A.; Tesoro, C.; Pascale, R.; Ostuni, A.; Matera, I.; Bianco, G.; Scrano, L.; Bufo, S.A.; Ciriello, R.; Di Capua, A. Legal Cannabis sativa L. Dried Inflorescences: Cannabinoids Content and Cytotoxic Activity against Human HepG2 Cell Line. Appl. Sci. 2023, 13, 4960. [Google Scholar] [CrossRef]
  88. Boonyayothin, W.; Kobtrakul, K.; Khositanon, P.; Vimolmangkang, S.; Phoolcharoen, W. Development of a Plant-Produced Recombinant Monoclonal Antibody against Δ-9-Tetrahydrocannabinol (Δ9-THC) for Immunoassay Application. Biotechnol. Rep. 2022, 34, e00725. [Google Scholar] [CrossRef]
  89. Muhammad Zen, N.A.; Kobtrakul, K.; Khositanon, P.; Sanookpan, K.; Buranasudja, V.; Vimolmangkang, S. Vegetable Oil-Based Cannabis: Its Cannabinoid Profiling and Photoprotective Effect on UVA-Irradiated Human Skin Keratinocytes. Thai J. Pharm. Sci. 2023, 46, 720–733. [Google Scholar] [CrossRef]
  90. Benkirane, C.; Ben Moumen, A.; Fauconnier, M.-L.; Belhaj, K.; Abid, M.; Caid, H.S.; Elamrani, A.; Mansouri, F. Bioactive Compounds from Hemp (Cannabis sativa L.) Seeds: Optimization of Phenolic Antioxidant Extraction Using Simplex Lattice Mixture Design and HPLC-DAD/ESI-MS2 Analysis. RSC Adv. 2022, 12, 25764–25777. [Google Scholar] [CrossRef]
  91. Benkirane, C.; Mansouri, F.; Ben Moumen, A.; Taaifi, Y.; Melhaoui, R.; Caid, H.S.; Fauconnier, M.; Elamrani, A.; Abid, M. Phenolic Profiles of Non-industrial Hemp (Cannabis sativa L.) Seed Varieties Collected from Four Different Moroccan Regions. Int. J. Food Sci. Technol. 2023, 58, 1367–1381. [Google Scholar] [CrossRef]
  92. Mastellone, G.; Marengo, A.; Sgorbini, B.; Scaglia, F.; Capetti, F.; Gai, F.; Peiretti, P.G.; Rubiolo, P.; Cagliero, C. Characterization and Biological Activity of Fiber-Type Cannabis sativa L. Aerial Parts at Different Growth Stages. Plants 2022, 11, 419. [Google Scholar] [CrossRef]
  93. Mastellone, G.; Marengo, A.; Sgorbini, B.; Rubiolo, P.; Cagliero, C. Development of a Dispersive Solid-Liquid Microextraction Method Using Natural Eutectic Solvents for a Greener Extraction of Phytochemicals from Fiber-Type Cannabis sp. Ind. Crops Prod. 2022, 187, 115476. [Google Scholar] [CrossRef]
  94. Ohtsuki, T.; Friesen, J.B.; Chen, S.-N.; McAlpine, J.B.; Pauli, G.F. Selective Preparation and High Dynamic-Range Analysis of Cannabinoids in “CBD Oil” and Other Cannabis sativa Preparations. J. Nat. Prod. 2022, 85, 634–646. [Google Scholar] [CrossRef]
  95. Kim, A.L.; Yun, Y.J.; Choi, H.W.; Hong, C.-H.; Shim, H.J.; Lee, J.H.; Kim, Y.-C. Profiling Cannabinoid Contents and Expression Levels of Corresponding Biosynthetic Genes in Commercial Cannabis (Cannabis sativa L.) Cultivars. Plants 2022, 11, 3088. [Google Scholar] [CrossRef]
  96. Judžentienė, A.; Garjonytė, R.; Būdienė, J. Phytochemical Composition and Antioxidant Activity of Various Extracts of Fibre Hemp (Cannabis sativa L.) Cultivated in Lithuania. Molecules 2023, 28, 4928. [Google Scholar] [CrossRef] [PubMed]
  97. Monari, A.; Cantalù, S.; Zanfrognini, B.; Brighenti, V.; Verri, P.; Zanardi, C.; Pellati, F.; Pigani, L. An Electrochemical Approach for the Prediction of Δ9-Tetrahydrocannabinolic Acid and Total Cannabinoid Content in Cannabis sativa L. Analyst 2023, 148, 4688–4697. [Google Scholar] [CrossRef]
  98. Anceschi, L.; Codeluppi, A.; Brighenti, V.; Tassinari, R.; Taglioli, V.; Marchetti, L.; Roncati, L.; Alessandrini, A.; Corsi, L.; Pellati, F. Chemical Characterization of Non-Psychoactive Cannabis sativa L. Extracts, In Vitro Antiproliferative Activity and Induction of Apoptosis in Chronic Myelogenous Leukaemia Cancer Cells. Phytother. Res. 2022, 36, 914–927. [Google Scholar] [CrossRef]
  99. Tzimas, P.S.; Petrakis, E.A.; Halabalaki, M.; Skaltsounis, L.A. Extraction Solvent Selection for Cannabis sativa L. by Efficient Exploration of Cannabinoid Selectivity and Phytochemical Diversity. Phytochem. Anal. 2024, 35, 163–183. [Google Scholar] [CrossRef]
  100. Tzimas, P.S.; Beteinakis, S.; Petrakis, E.A.; Papastylianou, P.T.; Kakabouki, I.; Small-Howard, A.L.; Skaltsounis, L.A.; Halabalaki, M. Uncovering the Metabolite Complexity and Variability of Cultivated Hemp (Cannabis sativa L.): A First Phytochemical Diversity Mapping in Greece. Phytochemistry 2024, 222, 114076. [Google Scholar] [CrossRef] [PubMed]
  101. Salazar-Bermeo, J.; Moreno-Chamba, B.; Martínez-Madrid, M.C.; Valero, M.; Rodrigo-García, J.; Hosseinian, F.; Martín-Bermudo, F.; Aguado, M.; De La Torre, R.; Martí, N.; et al. Preventing Mislabeling: A Comparative Chromatographic Analysis for Classifying Medical and Industrial Cannabis. Molecules 2023, 28, 3552. [Google Scholar] [CrossRef]
  102. Moreno-Chamba, B.; Salazar-Bermeo, J.; Hosseinian, F.; Martin-Bermudo, F.; Aguado, M.; De La Torre, R.; Martínez-Madrid, M.C.; Valero, M.; Martí, N.; Saura, D. Aromatic and Cannabinoid Profiles of Cannabis Inflorescences and Seed Oils: A Comprehensive Approach for Variety Characterization. Ind. Crops Prod. 2024, 210, 118143. [Google Scholar] [CrossRef]
  103. Mouton, M.; Gerber, M.; Van der Kooy, F. Validation of a Fast LC-PDA Method for the Quantification of Cannabinoids in Commercial Tea Samples. Rev. Bras. Farmacogn. 2024, 34, 197–201. [Google Scholar] [CrossRef]
  104. Mouton, M.; Gerber, M.; Van Der Kooy, F. Cure-All Cannabidiol? The Cannabidiol Content of Commercial Products. Phytomed. Plus 2024, 4, 100520. [Google Scholar] [CrossRef]
  105. Roussel, J.-M.; Schelling, C.; Righezza, M.; Veuthey, J.-L. Application of Prediction Intervals to the Interpretation of the Robustness Study of a UHPLC Method for the Separation of Cannabinoids. J. Pharm. Biomed. Anal. 2022, 220, 114977. [Google Scholar] [CrossRef]
  106. Coppey, F.; Schelling, C.; Veuthey, J.; Esseiva, P. Cloud-Enabled Handheld NIR Spectroscopy: A Transformative Approach for Real-Time Forensic Analysis of Cannabis Specimens. Helv. Chim. Acta 2023, 106, e202300052. [Google Scholar] [CrossRef]
  107. Majumdar, C.G.; ElSohly, M.A.; Ibrahim, E.A.; Elhendawy, M.A.; Stanford, D.; Chandra, S.; Wanas, A.S.; Radwan, M.M. Effect of Gamma Irradiation on Cannabinoid, Terpene, and Moisture Content of Cannabis Biomass. Molecules 2023, 28, 7710. [Google Scholar] [CrossRef]
  108. Piani, B.; Ferfuia, C.; Bortolomeazzi, R.; Verardo, G.; Baldini, M. Development and Optimization of an HPLC-PDA Method for the Determination of Major Cannabinoids in Hemp (Cannabis sativa L.) Essential Oil Obtained by Hydrodistillation. Food Anal. Methods 2022, 15, 1677–1686. [Google Scholar] [CrossRef]
  109. Yu, C.; Long, R.; Cao, F.; Zhao, X.; Lan, T.; Xu, D. Development of Pure Certified Reference Material of Cannabidiol. Molecules 2024, 29, 921. [Google Scholar] [CrossRef]
  110. Aviram, J.; Atzmony, D.; Frenklakh, A.; Kroll, A.; Zaks, I.; Hazekamp, A. THC Degradation Does Not Impair the Accuracy of THC Doses Aerosolized by the Metered-Dose SyqeAir Inhaler: A 24-Month Stability Trial. J. Cannabis Res. 2022, 4, 55. [Google Scholar] [CrossRef]
  111. Tran, J.; Elkins, A.C.; Spangenberg, G.C.; Rochfort, S.J. High-Throughput Quantitation of Cannabinoids by Liquid Chromatography Triple-Quadrupole Mass Spectrometry. Molecules 2022, 27, 742. [Google Scholar] [CrossRef]
  112. Treyer, A.; Reinhardt, J.K.; Eigenmann, D.E.; Oufir, M.; Hamburger, M. Phytochemical Comparison of Medicinal Cannabis Extracts and Study of Their CYP-Mediated Interactions with Coumarinic Oral Anticoagulants. Med. Cannabis Cannabinoids 2023, 6, 21–31. [Google Scholar] [CrossRef] [PubMed]
  113. Luca, S.V.; Wojtanowski, K.; Korona-Głowniak, I.; Skalicka-Woźniak, K.; Minceva, M.; Trifan, A. Spent Material Extractives from Hemp Hydrodistillation as an Underexplored Source of Antimicrobial Cannabinoids. Antibiotics 2024, 13, 485. [Google Scholar] [CrossRef]
  114. Huang, S.; Qiu, R.; Fang, Z.; Min, K.; Van Beek, T.A.; Ma, M.; Chen, B.; Zuilhof, H.; Salentijn, G.I. Semiquantitative Screening of THC Analogues by Silica Gel TLC with an Ag(I) Retention Zone and Chromogenic Smartphone Detection. Anal. Chem. 2022, 94, 13710–13718. [Google Scholar] [CrossRef]
  115. Fernández, S.; Carreras, T.; Castro, R.; Perelmuter, K.; Giorgi, V.; Vila, A.; Rosales, A.; Pazos, M.; Moyna, G.; Carrera, I.; et al. A Comparative Study of Supercritical Fluid and Ethanol Extracts of Cannabis Inflorescences: Chemical Profile and Biological Activity. J. Supercrit. Fluids 2022, 179, 105385. [Google Scholar] [CrossRef]
  116. Fernández, S.; Castro, R.; López-Radcenco, A.; Rodriguez, P.; Carrera, I.; García-Carnelli, C.; Moyna, G. Beyond Cannabinoids: Application of NMR-Based Metabolomics for the Assessment of Cannabis sativa L. Crop Health. Front. Plant Sci. 2023, 14, 1025932. [Google Scholar] [CrossRef] [PubMed]
  117. Durante, C.; Anceschi, L.; Brighenti, V.; Caroli, C.; Afezolli, C.; Marchetti, A.; Cocchi, M.; Salamone, S.; Pollastro, F.; Pellati, F. Application of Experimental Design in HPLC Method Optimisation for the Simultaneous Determination of Multiple Bioactive Cannabinoids. J. Pharm. Biomed. Anal. 2022, 221, 115037. [Google Scholar] [CrossRef] [PubMed]
  118. Brighenti, V.; Marani, M.; Caroli, C.; Bertarini, L.; Gaggiotti, A.; Pollastro, F.; Durante, C.; Cannazza, G.; Pellati, F. A New HPLC Method with Multiple Detection Systems for Impurity Analysis and Discrimination of Natural versus Synthetic Cannabidiol. Anal. Bioanal. Chem. 2024, 416, 4555–4569. [Google Scholar] [CrossRef]
  119. Trovato, E.; Arena, K.; La Tella, R.; Rigano, F.; Laganà Vinci, R.; Dugo, P.; Mondello, L.; Guarnaccia, P. Hemp Seed-Based Food Products as Functional Foods: A Comprehensive Characterization of Secondary Metabolites Using Liquid and Gas Chromatography Methods. J. Food Compos. Anal. 2023, 117, 105151. [Google Scholar] [CrossRef]
  120. MacLaughlin, L.L.; MacDonald, M.T. Is Nitrogen-Modified Atmosphere Packaging a Tool for Retention of Volatile Terpenes and Cannabinoids in Stored Cannabis sativa Inflorescence? J. Cannabis. Res. 2024, 6, 42. [Google Scholar] [CrossRef]
  121. Melchert, D.; Schaare, F.; Winterhalter, P.; Beuerle, T. CBD Products: Labeling Accuracy of an Obscure Niche Market. Food Control 2024, 160, 110375. [Google Scholar] [CrossRef]
  122. Gardener, H.; Wallin, C.; Bowen, J. Heavy Metal and Phthalate Contamination and Labeling Integrity in a Large Sample of US Commercially Available Cannabidiol (CBD) Products. Sci. Total Environ. 2022, 851, 158110. [Google Scholar] [CrossRef]
  123. Osiripun, V.; Labua, S. Effects of Thermal Processing on Cannabidiol Degradation in Cannabidiol-Infused Pomegranate Juice and Evaluation of Its Antioxidant Property. J. Curr. Sci. Technol. 2023, 13, 107–117. [Google Scholar]
  124. Vaclavik, L.; Benes, F.; Fenclova, M.; Hricko, J.; Krmela, A.; Svobodova, V.; Hajslova, J.; Mastovska, K. Quantitation of Cannabinoids in Cannabis Dried Plant Materials, Concentrates, and Oils Using Liquid Chromatography–Diode Array Detection Technique with Optional Mass Spectrometric Detection: Single-Laboratory Validation Study, First Action 2018.11. J. AOAC Int. 2019, 102, 1822–1833. [Google Scholar] [CrossRef]
  125. Wongumpornpinit, V.; Temkitthawon, P.; Paenkaew, S.; Saesong, T.; Waranuch, N.; Ingkaninan, K. Preparation of Cannabis Biomass and Crude Extracts with High Neutral Cannabinoid Content. Sci. Technol. Asia 2023, 28, 216227. [Google Scholar] [CrossRef]
  126. Putthong, C.; Temkitthawon, P.; Phimnuan, P.; Viyoch, J.; Tongpoolsomjit, K. Formulation of Chewable Nutraceuticals Prepared from Banana Fruit Pulp and Hemp Seed Oil. ScienceAsia 2024, 50, 2024056. [Google Scholar] [CrossRef]
  127. Musetti, B.; Kun, A.; Menchaca, D.; Rodríguez-Haralambides, A.; Varela, J.; Thomson, L.; Bahnson, E.M. Cannabis sativa Extracts Inhibit LDL Oxidation and the Formation of Foam Cells In Vitro, Acting as Potential Multi-Step Inhibitors of Atherosclerosis Development. PLoS ONE 2024, 19, e0310777. [Google Scholar] [CrossRef] [PubMed]
  128. Kursa, W.; Jamiołkowska, A.; Wyrostek, J.; Kowalski, R. Attempts to Use Hemp (Cannabis sativa L. var. Sativa) Inflorescence Extract to Limit the Growth of Fungi Occurring in Agricultural Crops. Appl. Sci. 2024, 14, 1680. [Google Scholar] [CrossRef]
  129. Wilson, W.B.; Abdur-Rahman, M. Determination of 11 Cannabinoids in Hemp Plant and Oils by Liquid Chromatography and Photodiode Array Detection. Chromatographia 2022, 85, 115–125. [Google Scholar] [CrossRef]
  130. Wilson, W.B.; Urbas, A.A.; Abdur-Rahman, M.; Romares, A.; Mistek-Morabito, E. Determination of Δ9-THC, THCA, Δ8-THC, and Total Δ9-THC in 53 Smokable Hemp Plant Products by Liquid Chromatography and Photodiode Array Detection. Forensic Chem. 2024, 37, 100550. [Google Scholar] [CrossRef]
  131. Wilson, W.B.; Urbas, A.A.; Jensen, H.; Sander, L.C. High-Throughput LC-PDA Method for Determination of Δ9-THC and Related Cannabinoids in Cannabis sativa. Forensic Sci. 2024, 41, 100610. [Google Scholar] [CrossRef]
  132. Casiraghi, A.; Gentile, A.; Selmin, F.; Gennari, C.G.M.; Casagni, E.; Roda, G.; Pallotti, G.; Rovellini, P.; Minghetti, P. Ultrasound-Assisted Extraction of Cannabinoids from Cannabis sativa for Medicinal Purpose. Pharmaceutics 2022, 14, 2718. [Google Scholar] [CrossRef]
  133. Silva Sofrás, F.M.; Alonso, R.; Retta, D.S.; Di Leo Lira, P.; Desimone, M.F.; van Baren, C.M. Development and Validation of a Simple, Fast, and Accessible HPLC-UV Method for Cannabinoids Determination in Cannabis sativa L. Extracts and Medicinal Oils. Curr. Pharm. Des. 2023, 29, 1918–1928. [Google Scholar] [CrossRef] [PubMed]
  134. Pittiglio, M.K.; Ramirez, G.A.; Tesfatsion, T.T.; Ray, K.P.; Cruces, W. HPLC Method for Better Separation of THC Isomers to Ensure Safety and Compliance in the Hemp Market. ACS Omega 2024, 9, 25390–25394. [Google Scholar] [CrossRef]
  135. Cuchiaro, J.; Baumgartner, J.; Reynolds, M.M. Modeling a Pesticide Remediation Strategy for Preparative Liquid Chromatography Using High-Performance Liquid Chromatography. J. Cannabis Res. 2023, 5, 13. [Google Scholar] [CrossRef]
  136. Gigopulu, O.; Geskovski, N.; Stefkov, G.; Stoilkovska Gjorgievska, V.; Slaveska Spirevska, I.; Huck, C.W.; Makreski, P. A Unique Approach for In-Situ Monitoring of the THCA Decarboxylation Reaction in Solid State. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 267, 120471. [Google Scholar] [CrossRef]
  137. Gjorgievska, V.S.; Karanfilova, I.C.; Trajkovska, A.; Karapandzova, M.; Petrovska, B.B.; Kulevanova, S.; Stefkov, G. Monitoring of Cannabis Cultivar Technological Maturity by Trichome Morphology Analysis and HPLC Phytocannabinoid Content. Pharmacogn. Res. 2023, 15, 94–100. [Google Scholar] [CrossRef]
  138. Oliveira, G.D.A.R.; Estrada-Semprun, O.E.; Arantes, L.C.; Rodrigues, P.M.; Ribeiro, R.A.; Fagg, C.W.; Magalhães, P.O.; Fonseca-Bazzo, Y.M.; Silveira, D. Does Trema micranthum (L.) Blume Produce Cannabinoids? Plants 2024, 13, 1951. [Google Scholar] [CrossRef]
  139. Loxterkamp, L.; Monks, K. Analyzing Cannabis Flowers According to the German Pharmacopeia—Monograph 2018. Available online: https://www.knauer.net/document/download/354/3dffc74b-b035-4c99-9983-078b9c3ffaba/11569?preview=True (accessed on 28 February 2025).
  140. Spadafora, N.D.; Felletti, S.; Chenet, T.; Sirangelo, T.M.; Cescon, M.; Catani, M.; De Luca, C.; Stevanin, C.; Cavazzini, A.; Pasti, L. The Influence of Drying and Storage Conditions on the Volatilome and Cannabinoid Content of Cannabis sativa L. Inflorescences. Anal. Bioanal. Chem. 2024, 416, 3797–3809. [Google Scholar] [CrossRef]
  141. de Souza, M.R.; Koetz, M.; Limberger, R.P.; Henriques, A.T. DoE-Assisted Development and Validation of a Stability-Indicating HPLC-DAD Method for Simultaneous Determination of Five Cannabinoids in Cannabis sativa L. Based on Analytical Quality by Design (AQbD) Concept. Phytochem. Anal. 2022, 33, 999–1017. [Google Scholar] [CrossRef] [PubMed]
  142. Lajoie, C.; Doyen, A.; Feutry, P.; Gagnon, D.; Brisson, G. Impact of Emulsifiers for the Nanoencapsulation with Maltodextrin of Cannabis Oil by Spray Drying on the Physicochemical Properties and Bioaccessibility of Cannabinoids. Food Funct. 2022, 13, 10320–10332. [Google Scholar] [CrossRef] [PubMed]
  143. Mano-Sousa, B.J.; Alves, B.C.; Pedrosa, A.M.; Lima, P.L.; de Andrade, F.P.; Duarte-Almeida, J.M. Validation of Analytical Method of Cannabinoids: Novel Approach Using Turbo-Extraction. Talanta 2023, 254, 124108. [Google Scholar] [CrossRef] [PubMed]
  144. Dreger, M.; Szalata, M.; Górska-Paukszta, M.; Mańkowska, G.; Oleszak, G.; Kwiatkowska, E.; Ożarowski, M. Content of Cannabinoids in Clonally Propagated Industrial Hemp. J. Nat. Fibers 2023, 20, 2245968. [Google Scholar] [CrossRef]
  145. Jokić, S.; Jerković, I.; Pavić, V.; Aladić, K.; Molnar, M.; Kovač, M.J.; Vladimir-Knežević, S. Terpenes and Cannabinoids in Supercritical CO2 Extracts of Industrial Hemp Inflorescences: Optimization of Extraction, Antiradical and Antibacterial Activity. Pharmaceuticals 2022, 15, 1117. [Google Scholar] [CrossRef]
  146. Bongiorno, P.; Lopalco, A.; Casiraghi, A.; Spennacchio, A.; Pitruzzella, A.; Lopedota, A.A.; Minghetti, P.; Denora, N. Digital Technologies Applied to Control the One-Step Process of Cannabis Olive Oil Preparations. Pharmaceutics 2023, 15, 870. [Google Scholar] [CrossRef]
  147. Cuchiaro, J.; DeRoo, J.; Thai, J.; Reynolds, M.M. Evaluation of the Adsorption-Accessible Surface Area of MIL-53(Al) Using Cannabinoids in a Closed System. ACS Appl. Mater. Interfaces 2022, 14, 12836–12844. [Google Scholar] [CrossRef] [PubMed]
  148. Rylands, M.; Kusza, D.A.; Hlabisa, N.; Gwampa, T.; Ravenscroft, N. Development of Physicochemical Methods for the Quantification of CBD in South African Cannabis-Based Consumer Goods. S. Afr. J. Chem. 2024, 78, 36–42. [Google Scholar]
  149. Kim, E.-S.; Park, S.-H.; Kinney, C.A.; Olejar, K.J.; Corredor-Perilla, I.C. Comparison of Decarboxylation Rates of Acidic Cannabinoids between Secretory Cavity Contents and Air-Dried Inflorescence Extracts in Cannabis sativa cv. ‘Cherry Wine’. Sci. Rep. 2024, 14, 16411. [Google Scholar] [CrossRef] [PubMed]
  150. Park, S.-H.; Pauli, C.S.; Gostin, E.L.; Staples, S.K.; Seifried, D.; Kinney, C.; Vanden Heuvel, B.D. Effects of Short-Term Environmental Stresses on the Onset of Cannabinoid Production in Young Immature Flowers of Industrial Hemp (Cannabis sativa L.). J. Cannabis Res. 2022, 4, 1–13. [Google Scholar] [CrossRef]
  151. Carvalho, V.M.; de Almeida, F.G.; de Aguiar, A.F.L.; Pereira, F.C.S.; Rocha, E.D. Facing the Forensic Challenge of Cannabis Regulation: A Methodology for the Differentiation between Hemp and Marijuana Samples. Braz. J. Anal. Chem. 2022, 9, 162–176. [Google Scholar] [CrossRef]
  152. Qamar, S.; Torres, Y.J.M.; Parekh, H.S.; Falconer, J.R. Fractional Factorial Design Study for the Extraction of Cannabinoids from CBD-Dominant Cannabis Flowers by Supercritical Carbon Dioxide. Processes 2022, 10, 93. [Google Scholar] [CrossRef]
  153. Qamar, S.; Manrique, Y.J.; Parekh, H.S.; Falconer, J.R. Development and Optimization of Supercritical Fluid Extraction Setup Leading to Quantification of 11 Cannabinoids Derived from Medicinal Cannabis. Biology 2021, 10, 481. [Google Scholar] [CrossRef]
  154. Büttenbender, S.; Carlos, G.; Steppe, M.; Ortiz, R.S.; Limberger, R.P.; Mendez, A.S.L. Fast and Reliable Profiling of Cannabinoids in Seized Samples Using the Method of HPLC–DAD Followed by Chemometrics. Forensic Toxicol. 2022, 40, 407–413. [Google Scholar] [CrossRef]
  155. Sedan, D.; Vaccarini, C.; Demetrio, P.; Morante, M.; Montiel, R.; Saurí, A.; Andrinolo, D. Cannabinoid Content in Cannabis Flowers and Homemade Cannabis-Based Products Used for Therapeutic Purposes in Argentina. Cannabis Cannabinoid Res. 2023, 8, 197–206. [Google Scholar] [CrossRef]
  156. Liebling, J.P.; Clarkson, N.J.; Gibbs, B.W.; Yates, A.S.; O’Sullivan, S.E. An Analysis of Over-the-Counter Cannabidiol Products in the United Kingdom. Cannabis Cannabinoid Res. 2022, 7, 207–213. [Google Scholar] [CrossRef]
  157. Song, L.; Provis, J.; Al-Bataineh, A.M.; Fabien, K.J.; Kotler, M. Development of a Liquid Chromatographic Method with a Different Selectivity for the Quantification of Eighteen Phytocannabinoids in Hemp. Talanta Open 2024, 10, 100336. [Google Scholar] [CrossRef]
  158. Czauderna, M.; Taubner, T.; Wojtak, W. Comparative Study of Gas and Liquid Chromatography Methods for the Determination of Underivatised Neutral and Acidic Cannabinoids and Cholesterol. Molecules 2024, 29, 2165. [Google Scholar] [CrossRef] [PubMed]
  159. Mazzara, E.; Torresi, J.; Fico, G.; Papini, A.; Kulbaka, N.; Dall’Acqua, S.; Sut, S.; Garzoli, S.; Mustafa, A.M.; Cappellacci, L.; et al. A Comprehensive Phytochemical Analysis of Terpenes, Polyphenols and Cannabinoids, and Micromorphological Characterization of 9 Commercial Varieties of Cannabis sativa L. Plants 2022, 11, 891. [Google Scholar] [CrossRef] [PubMed]
  160. Boumrah, Y.; Baroudi, S.; Kecir, M.; Bouanani, S. Characterization of Algerian-seized Hashish over Eight Years (2011–2018). Part II: Chemical Categorization. J. Forensic Sci. 2020, 65, 1845–1851. [Google Scholar] [CrossRef]
  161. Boumrah, Y.; Kermouche, W.; Zarita, B.; Drardja, E.D.; Bouanani, S. Hashish Seizures in Algeria over the 2019–2020 Period: The Rise of Potent Hashish Hybrids. J. Forensic Sci. 2022, 67, 889–898. [Google Scholar] [CrossRef] [PubMed]
  162. Hall, D.R.; Sinclair, J.S.; Bhuyan, D.J.; Khoo, C.; Li, C.G.; Sarris, J.; Low, M. Quality Control of Cannabis Inflorescence and Oil Products: Response Factors for the Cost-Efficient Determination of Ten Cannabinoids by HPLC. Talanta Open 2022, 5, 100112. [Google Scholar] [CrossRef]
  163. Nalli, Y.; Bashir Mir, K.; Amin, T.; Gannedi, V.; Jameel, E.; Goswami, A.; Ali, A. Divergent Synthesis of Fractionated Cannabis sativa Extract Led to Multiple Cannabinoids C-&O-Glycosides with Anti-Proliferative/Anti-Metastatic Properties. Bioorg. Chem. 2024, 143, 107030. [Google Scholar] [CrossRef]
  164. Chan, K.-W. Kick Starting Method Development for Hemp Analysis Using Cannabis as Equivalent Material. Prob. Forensic Sci. 2023, 132, 271–283. [Google Scholar] [CrossRef]
  165. Tapia-Tapia, E.; Aránguiz, P.; Diaz, R.; Espinoza, L.; Weinstein-Oppenheimer, C.R.; Cuellar, M. Effect of Cannabis sativa L. Extracts, Phytocannabinoids and Their Acetylated Derivates on the SHSY-5Y Neuroblastoma Cells’ Viability and Caspases 3/7 Activation. Biol. Res. 2024, 57, 33. [Google Scholar] [CrossRef]
  166. Darigh, F.; Iranbakhsh, A.; Oraghi Ardebili, Z.; Ebadi, M.; Hassanpour, H. Simulated Microgravity Contributed to Modification of Callogenesis Performance and Secondary Metabolite Production in Cannabis indica. Plant Physiol. Biochem. 2022, 186, 157–168. [Google Scholar] [CrossRef]
  167. Darigh, F.; Iranbakhsh, A.; Oraghi Ardebili, Z.; Ebadi, M. Non-Thermal Plasma Improved Callogenesis Performance and Elicited the Production of Cannabinoids by Modifying DNA Methylome, Expression of WRKY1 and ERF1B Transcription Factors, and Expression of Genes That Contributed to the Biosynthesis of Cannabinoids. Protoplasma 2023, 260, 159–170. [Google Scholar] [CrossRef]
  168. Song, L.; Carlson, S.; Valenzuela, G.; Chao, M.; Pathipaka, S.B. Development of a Validated Method for Rapid Quantification of up to Sixteen Cannabinoids Using Ultra-High-Performance Liquid Chromatography Diode-Array Detector with Optional Electrospray Ionization Time-of-Flight Mass Spectrometry Detection. J. Chromatogr. A 2022, 1670, 462953. [Google Scholar] [CrossRef] [PubMed]
  169. Lazarjani, M.; Seyfoddin, A.; Le, T.T.; Chen, T. Development and Validation of HPLC and GC Methods for Quantification of Cannabinoids and Terpenes Extracted by Ultrasound Assisted Extraction Technique. Drug Anal. Res. 2024, 8, 33–45. [Google Scholar] [CrossRef]
  170. Mastinu, A.; Ascrizzi, R.; Ribaudo, G.; Bonini, S.A.; Premoli, M.; Aria, F.; Maccarinelli, G.; Gianoncelli, A.; Flamini, G.; Pistelli, L.; et al. Prosocial Effects of Nonpsychotropic Cannabis sativa in Mice. Cannabis Cannabinoid Res. 2022, 7, 170–178. [Google Scholar] [CrossRef] [PubMed]
  171. Tsujikawa, K.; Okada, Y.; Segawa, H.; Yamamuro, T.; Kuwayama, K.; Kanamori, T.; Iwata, Y.T. Evaluation of Decarboxylation Efficiency of Δ9-Tetrahydrocannabinolic Acid and Cannabidiolic Acid by UNODC Method. Forensic Toxicol. 2023, 41, 105–113. [Google Scholar] [CrossRef] [PubMed]
  172. Correia, B.; Ahmad, S.M.; Quintas, A. Determination of Phytocannabinoids in Cannabis Samples by Ultrasound-Assisted Solid-Liquid Extraction and High-Performance Liquid Chromatography with Diode Array Detector Analysis. J. Chromatogr. A 2023, 1705, 464191. [Google Scholar] [CrossRef]
  173. Barhdadi, S.; Courselle, P.; Deconinck, E.; Vanhee, C. The Analysis of Cannabinoids in E-Cigarette Liquids Using LC-HRAM-MS and LC-UV. J. Pharm. Biomed. Anal. 2023, 230, 115394. [Google Scholar] [CrossRef]
  174. De Leersnijder, C.; Duchateau, C.; De Braekeleer, K.; Deconinck, E. Relative Response Factors and Multiple Regression Models in Liquid Chromatography to Quantify Low-Dosed Components Using Alternative Standards—Proof of Concept: Total Δ9-THC Content in Cannabis Flowers Using CBD as Reference. Anal. Bioanal. Chem. 2022, 414, 6507–6520. [Google Scholar] [CrossRef]
  175. Duchateau, C.; De Leersnijder, C.; Barhdadi, S.; Canfyn, M.; De Braekeleer, K.; Deconinck, E. Discrepancies between Validated GC-FID and UHPLC-DAD Methods for the Analysis of Δ9-THC and CBD in Dried Hemp Flowers. Drug Test. Anal. 2022, 14, 1732–1743. [Google Scholar] [CrossRef]
  176. Toloza, H.; Buitrago, O.Y.; Orjuela, A.; Santaella, M.A.; Hurtado, A.M.; Arturo, D.E. Solvent Extraction of Cannabis sativa under Cryogenic Conditions. Sep. Purif. Technol. 2024, 329, 124906. [Google Scholar] [CrossRef]
  177. Kollipara, R.; Langille, E.; Tobin, C.; French, C.R. Phytocannabinoids Reduce Seizures in Larval Zebrafish and Affect Endocannabinoid Gene Expression. Biomolecules 2023, 13, 1398. [Google Scholar] [CrossRef] [PubMed]
  178. Gilmore, A.M.; Elhendawy, M.A.; Radwan, M.M.; Kidder, L.H.; Wanas, A.S.; Godfrey, M.; Hildreth, J.B.; Robinson, A.E.; ElSohly, M.A. Absorbance-Transmittance Excitation Emission Matrix Method for Quantification of Major Cannabinoids and Corresponding Acids: A Rapid Alternative to Chromatography for Rapid Chemotype Discrimination of Cannabis sativa Varieties. Cannabis Cannabinoid Res. 2023, 8, 911–922. [Google Scholar] [CrossRef]
  179. Hajrulai-Musliu, Z.; Dimitreska Stojkovikj, E.; Gusheski, D.; Musliu, D.; Velkovski, D. High-Performance Liquid Chromatography with DAD Detection for the Determination of Cannabinoids in Commercial Veterinary CBD Oil. Pharmacy 2024, 12, 181. [Google Scholar] [CrossRef]
  180. Elhendawy, M.A.; Radwan, M.M.; Ibrahim, E.A.; Wanas, A.S.; Chandra, S.; Godfrey, M.; ElSohly, M.A. Validation and Quantitation of Fifteen Cannabinoids in Cannabis and Marketed Products Using High-Performance Liquid Chromatography-Ultraviolet/Photodiode Array Method. Cannabis Cannabinoid Res. 2024, 9, e1091–e1107. [Google Scholar] [CrossRef]
  181. Fries, A.; Moldes, C.A.; Mazzaferro, L.S. Cost-Efficient Analysis of Cannabinoids in Therapeutic Oils Using HPLC with UV and Mass Spectrometry Detection. Nat. Prod. Res. 2024, 13, 1–9. [Google Scholar] [CrossRef]
  182. Song, L.; Valenzuela, G.; Carlson, S.; Dodson, Z.; Adisa, M. Potency Testing of up to Twenty Cannabinoids by Liquid Chromatography Diode Array Detector with Optional Electrospray Ionization Time-of-Flight Mass Spectrometry. Anal. Chim. Acta 2022, 1207, 339827. [Google Scholar] [CrossRef] [PubMed]
  183. Todd, J.; Song, H.; Van Acker, R. Does Pollination Alter the Cannabinoid Composition and Yield of Extracts from Hemp (Cannabis sativa L. cv. Finola) Flowers? Ind. Crops Prod. 2022, 183, 114989. [Google Scholar] [CrossRef]
  184. Song, L.; Meyer, G.; Adejumo, E.; Jovanovich, E.; LeBlanc, L.; Provis, J. Potency Testing of up to Sixteen Cannabinoids in Hemp-Infused Edibles Using Liquid Chromatography Diode Array Detector with Optional Confirmation of Identity by Electrospray Ionization Time-of-Flight Mass Spectrometry. Food Chem. 2023, 417, 135819. [Google Scholar] [CrossRef] [PubMed]
  185. Quiñones, R.; Casiano-Negroni, A.; Pijor, H.; Moreno, S.; Suarez, K.; Westfall, T.D.; Sullins, C.; Ivey, S.; Buxó, J.A. Analysis of Cannabinoids in Lotions Using High-Performance Liquid Chromatography. J. Chem. Educ. 2022, 99, 3558–3565. [Google Scholar] [CrossRef]
  186. Quiñones, R.; Moreno, S.; Smythers, A.L.; Sullins, C.; Pijor, H.; Brown, G.; Trouten, A.; Richards-Waugh, L.L.; Siddig, A. Quantification of Cannabis in Infused Consumer Products and Their Residues on Skin. ACS Pharmacol. Transl. Sci. 2022, 5, 642–651. [Google Scholar] [CrossRef]
  187. Glinn, M.A.; Michaud, G.P. Potency Levels of Regulated Cannabis Products in Michigan 2021–2022. J. Forensic Sci. 2023, 68, 1894–1905. [Google Scholar] [CrossRef]
  188. Namera, A.; Ota, S.; Tomioka, Y.; Saito, T.; Nagao, M. Facile Determination of Natural Cannabinoids in Cannabis Products Using a Conventional Fully Porous Particle Column and Isocratic High-Performance Liquid Chromatography with Diode-Array Detector. Forensic Toxicol 2022, 40, 417–421. [Google Scholar] [CrossRef]
  189. Jaidee, W.; Siridechakorn, I.; Nessopa, S.; Wisuitiprot, V.; Chaiwangrach, N.; Ingkaninan, K.; Waranuch, N. Kinetics of CBD, Δ9-THC Degradation and Cannabinol Formation in Cannabis Resin at Various Temperature and pH Conditions. Cannabis Cannabinoid Res. 2022, 7, 537–547. [Google Scholar] [CrossRef]
  190. Klinsang, T.; Charoensit, P.; Phimnuan, P.; Luangpraditkun, K.; Ross, G.M.; Viennet, C.; Ross, S.; Viyoch, J. In Vitro Wound Healing Potential of a Fibroin Film Incorporating a Cannabidiol/2-Hydroxypropyl-β-Cyclodextrin Complex. Pharmaceutics 2023, 15, 2682. [Google Scholar] [CrossRef] [PubMed]
  191. Luangpraditkun, K.; Pimjuk, P.; Phimnuan, P.; Wisanwattana, W.; Wisespongpand, C.; Waranuch, N.; Viyoch, J. Anti-Aging Properties of Cannabis sativa Leaf Extract against UVA Irradiation. Cosmetics 2024, 11, 45. [Google Scholar] [CrossRef]
  192. Chaiwangrach, N.; Waranuch, N.; Temkitthawon, P.; Wongwad, E.; Nuengchamnong, N.; Usuwanthim, K.; Saesong, T.; Rakkhetkorn, Y.; Pisutthanan, S.; Ingkaninan, K. Quality Assessment, Stability and In-Vitro Anti-Inflammatory Activities of a Topical Ointment Containing Cannabis and Turmeric Extracts for Haemorrhoids and Skin Diseases. Phytomed. Plus 2024, 4, 100635. [Google Scholar] [CrossRef]
  193. Birenboim, M.; Chalupowicz, D.; Maurer, D.; Barel, S.; Chen, Y.; Fallik, E.; Paz-Kagan, T.; Rapaport, T.; Sadeh, A.; Kengisbuch, D.; et al. Multivariate Classification of Cannabis Chemovars Based on Their Terpene and Cannabinoid Profiles. Phytochemistry 2022, 200, 113215. [Google Scholar] [CrossRef]
  194. Birenboim, M.; Fallik, E.; Kengisbuch, D.; Shimshoni, J.A. Quantitative and Qualitative Spectroscopic Parameters Determination of Major Cannabinoids. J. Lumin. 2022, 252, 119387. [Google Scholar] [CrossRef]
  195. Birenboim, M.; Kenigsbuch, D.; Shimshoni, J.A. Novel Fluorescence Spectroscopy Method Coupled with N-PLS-R and PLS-DA Models for the Quantification of Cannabinoids and the Classification of Cannabis Cultivars. Phytochem. Anal. 2023, 34, 280–288. [Google Scholar] [CrossRef]
  196. Birenboim, M.; Rinnan, Å.; Kengisbuch, D.; Shimshoni, J.A. Novel Fluorescence Spectroscopy Coupled with PARAFAC Modeling for Major Cannabinoids Quantification and Identification in Cannabis Extracts. Chemom. Intell. Lab. Syst. 2023, 232, 104717. [Google Scholar] [CrossRef]
  197. Birenboim, M.; Brikenstein, N.; Duanis-Assaf, D.; Maurer, D.; Chalupowicz, D.; Kenigsbuch, D.; Shimshoni, J.A. In Pursuit of Optimal Quality: Cultivar-Specific Drying Approaches for Medicinal Cannabis. Plants 2024, 13, 1049. [Google Scholar] [CrossRef]
  198. Birenboim, M.; Chalupowicz, D.; Kenigsbuch, D.; Shimshoni, J.A. Improved Long-Term Preservation of Cannabis Inflorescence by Utilizing Integrated Pre-Harvest Hexanoic Acid Treatment and Optimal Post-Harvest Storage Conditions. Plants 2024, 13, 992. [Google Scholar] [CrossRef] [PubMed]
  199. Brikenstein, N.; Birenboim, M.; Kenigsbuch, D.; Shimshoni, J.A. Optimization of Trimming Techniques for Enhancing Cannabinoid and Terpene Content in Medical Cannabis Inflorescences. Med. Cannabis Cannabinoids 2024, 7, 111–118. [Google Scholar] [CrossRef] [PubMed]
  200. Malík, M.; Praus, L.; Tlustoš, P. Comparison of Recirculation and Drain-to-Waste Hydroponic Systems in Relation to Medical Cannabis (Cannabis sativa L.) Plants. Ind. Crops Prod. 2023, 202, 117059. [Google Scholar] [CrossRef]
  201. Velechovský, J.; Malík, M.; Šenkyřík, J.B.; Tlustoš, P. Effect of Augmented Nutrient Composition and Fertigation System on Biomass Yield and Cannabinoid Content of Medicinal Cannabis (Cannabis sativa L.) Cultivation. Front. Plant Sci. 2024, 15, 1322824. [Google Scholar] [CrossRef]
  202. Malikova, L.; Malik, M.; Pavlik, J.; Ulman, M.; Pechouckova, E.; Skrivan, M.; Kokoska, L.; Tlustos, P. Anti-Staphylococcal Activity of Soilless Cultivated Cannabis across the Whole Vegetation Cycle under Various Nutritional Treatments in Relation to Cannabinoid Content. Sci. Rep. 2024, 14, 4343. [Google Scholar] [CrossRef]
  203. Kim, J.; Kim, D.-G.; Kim, J.-B.; Ryu, J.; Lee, Y.-J.; Woo, N.M.; Sik, J.J.; Kim, S.H. Simple Method for Detecting Tetrahydrocannabinolic Acid and Tetrahydrocannabinol in Cannabis Tissues Using Urine-Based Drug Diagnostic Device. Ind. Crops Prod. 2024, 219, 119007. [Google Scholar] [CrossRef]
  204. Lumu, S.; Hatterman-Valenti, H.; Hammed, A.; Monono, E. Effect of Short-Term Storage on Cannabinoid Content of Dried Floral Hemp (Cannabis sativa L.) Inflorescence. J. Appl. Res. Med. Aromat. Plants 2024, 42, 100567. [Google Scholar] [CrossRef]
  205. Stasiłowicz-Krzemień, A.; Sip, S.; Szulc, P.; Cielecka-Piontek, J. Determining Antioxidant Activity of Cannabis Leaves Extracts from Different Varieties—Unveiling Nature’s Treasure Trove. Antioxidants 2023, 12, 1390. [Google Scholar] [CrossRef]
  206. Stasiłowicz-Krzemień, A.; Sip, S.; Szulc, P.; Walkowiak, J.; Cielecka-Piontek, J. The Antioxidant and Neuroprotective Potential of Leaves and Inflorescences Extracts of Selected Hemp Varieties Obtained with scCO2. Antioxidants 2023, 12, 1827. [Google Scholar] [CrossRef]
  207. Sip, S.; Stasiłowicz-Krzemień, A.; Sip, A.; Szulc, P.; Neumann, M.; Kryszak, A.; Cielecka-Piontek, J. Development of Delivery Systems with Prebiotic and Neuroprotective Potential of Industrial-Grade Cannabis sativa L. Molecules 2024, 29, 3574. [Google Scholar] [CrossRef] [PubMed]
  208. Pires, B.; Oliveira, P.; Simão, A.Y.; Reis, J.; Ramos, S.; Duarte, A.P.; Margalho, C.; Rosado, T.; Barroso, M.; Gallardo, E. Characterisation of Cannabis-Based Products Marketed for Medical and Non-Medical Use Purchased in Portugal. Molecules 2024, 29, 2737. [Google Scholar] [CrossRef] [PubMed]
  209. Chauhan, J.; Bastia, B.K.; Kohli, K.; Chaudhary, B.; Chikara, G.; Gupta, A.; Kumar, A. Phytocannabinoid Profile and Potency of Cannabis Resin (Hashish) of Northwest Himalayas of India. J. Forensic Sci. 2024, 69, 1918–1925. [Google Scholar] [CrossRef]
  210. Li, H.; Zhao, Q.; Chang, S.; Wang, L.; Zhao, B. Phytochemical Analysis and Bioactivity of Different Ethanolic Extracts from Cannabidiol Full-Spectrum Oil. J. Mol. Liq. 2023, 372, 121173. [Google Scholar] [CrossRef]
  211. Monton, C.; Chankana, N.; Leelawat, S.; Suksaeree, J.; Songsak, T. Optimization of Supercritical Carbon Dioxide Fluid Extraction of Seized Cannabis and Self-Emulsifying Drug Delivery System for Enhancing the Dissolution of Cannabis Extract. J. Supercrit. Fluids 2022, 179, 105423. [Google Scholar] [CrossRef]
  212. Monton, C.; Tanpao, T.; Navakul, C.; Pengkum, T.; Santasanasuwan, S.; Suksaeree, J.; Charoenchai, L.; Songsak, T. Cannabidiol, ∆9-Tetrahydrocannabinol, and Cannabinol Contents of Cannabis sativa L. Inflorescences Claimed to Be Hang Kra Rog Phu Phan Cultivar Cultivated Outdoors in Various Locations of Thailand. Phytochem. Lett. 2023, 57, 126–132. [Google Scholar] [CrossRef]
  213. Song, L.; LeBlanc, L.; Jovanovich, E.; Mohammad Al-Bataineh, A.; Jervelle Fabien, K. A Rapid and Accurate Liquid Chromatographic Method for Hemp Compliance Testing. Forensic Chem. 2024, 40, 100592. [Google Scholar] [CrossRef]
  214. Mehdi, J.; Husain, D.; Srivastava, A.; Gupta, N.; Shanker, K.; Talha, M.; Lal, R.K.; Kumar, B.; Gupta, A.K. The Genetic Diversity and Chemical Profiling of Natural Populations of Indian Cannabis. Ind. Crops Prod. 2023, 203, 117187. [Google Scholar] [CrossRef]
  215. Ghosh, D.; Chaudhary, N.; Shanker, K.; Kumar, B.; Kumar, N. Monoecious Cannabis sativa L. Discloses the Organ-Specific Variation in Glandular Trichomes, Cannabinoids Content and Antioxidant Potential. J. Appl. Res. Med. Aromat. Plants 2023, 35, 100476. [Google Scholar] [CrossRef]
  216. Huang, S.; Van Beek, T.A.; Claassen, F.W.; Janssen, H.-G.; Ma, M.; Chen, B.; Zuilhof, H.; Salentijn, G.I. Comprehensive Cannabinoid Profiling of Acid-Treated CBD Samples and Δ8-THC-Infused Edibles. Food Chem. 2024, 440, 138187. [Google Scholar] [CrossRef]
  217. Pathompak, P.; Chankana, N.; Vipunngeun, N.; Phonkrathok, S. HPLC Method Development and Validation for Quantitation of CBD and THC in Suk-SaiYas Pills. J. Curr. Sci. 2022, 12, 538546. [Google Scholar] [CrossRef]
  218. Patirathnan, N.; Tummarintra, P.; Phakdeenukoolkitja, S. Analysis of THC and CBD in Cannabis Commercial Oil: Preliminary Study. KKU Res. J. (Grad. Stud.) 2024, 24, 143–154. [Google Scholar]
  219. Erukainure, O.L.; Oyenihi, O.R.; Amaku, J.F.; Chukwuma, C.I.; Nde, A.L.; Salau, V.F.; Matsabisa, M.G. Cannabis sativa L. Modulates Altered Metabolic Pathways Involved in Key Metabolisms in Human Breast Cancer (MCF-7) Cells: A Metabolomics Study. Heliyon 2023, 9, e16156. [Google Scholar] [CrossRef] [PubMed]
  220. Mastellone, G.; Marengo, A.; Sgorbini, B.; Rubiolo, P.; Anderson, J.L.; Cagliero, C. Ultrasound-Assisted Dispersive Solid-Liquid Microextraction with Eutectic Solvents for the Determination of Cannabinoids in Different Hemp Products. J. Chromatogr. B 2024, 1232, 123967. [Google Scholar] [CrossRef]
  221. Mileti, O.; Baldino, N.; Paleologo, M.F.O.; Lupi, F.R.; Marra, M.; Iacopetta, D.; Gabriele, D. Oil Extraction from Hemp Plant as a Potential Source of Cannabidiol for Healthy Protein Foods. Antioxidants 2023, 12, 1950. [Google Scholar] [CrossRef]
  222. Genovese, S.; Epifano, F.; Palumbo, L.; Collevecchio, C.; Fiorito, S. An Easy Way for the Removal of Residual Hydrocarbon Fractions from Crystallized Cannabigerol and Cannabidiol. Food Bioprod. Process 2023, 142, 50–58. [Google Scholar] [CrossRef]
  223. Fu, Y.; Zhu, S.; Duan, S.; Liu, L. Bioassay-Guided Isolation and Identification of Antibacterial Components against Escherichia coli from Industrial Hemp Leaves. Separations 2023, 10, 35. [Google Scholar] [CrossRef]
  224. Kalinowska, M.; Płońska, A.; Trusiak, M.; Gołębiewska, E.; Gorlewska-Pietluszenko, A. Comparing the Extraction Methods, Chemical Composition, Phenolic Contents and Antioxidant Activity of Edible Oils from Cannabis sativa and Silybum marianu Seeds. Sci. Rep. 2022, 12, 20609. [Google Scholar] [CrossRef]
  225. Kalinowska, M.; Gryko, K.; Wróblewska, A.M.; Jabłońska-Trypuć, A.; Karpowicz, D. Phenolic Content, Chemical Composition and Anti-/Pro-Oxidant Activity of Gold Milenium and Papierowka Apple Peel Extracts. Sci. Rep. 2020, 10, 14951. [Google Scholar] [CrossRef]
  226. Fućak, T.; Kreft, S.; Svedružić, Ž.M.; Tavčar, E. Mechanism and Kinetics of CBDA Decarboxylation into CBD in Hemp. J. Plant Biochem. Biotechnol. 2023, 32, 608–621. [Google Scholar] [CrossRef]
  227. Vida, R.G.; Strauss, L.V.; Bajtel, Á.; Kiss, T.; Csupor, D.; Fittler, A. Safety and Risks of CBD Oils Purchased Online: Unveiling Uncertain Quality and Vague Health Claims. Front. Pharmacol. 2023, 14, 1273540. [Google Scholar] [CrossRef]
  228. Picco, A.; Segale, L.; Miletto, I.; Pollastro, F.; Aprile, S.; Locatelli, M.; Bari, E.; Torre, M.L.; Giovannelli, L. Spray-Dried Powder Containing Cannabigerol: A New Extemporaneous Emulgel for Topical Administration. Pharmaceutics 2023, 15, 2747. [Google Scholar] [CrossRef]
  229. Jarén, C.; Zambrana, P.C.; Pérez-Roncal, C.; López-Maestresalas, A.; Ábrego, A.; Arazuri, S. Potential of NIRS Technology for the Determination of Cannabinoid Content in Industrial Hemp (Cannabis sativa L.). Agronomy 2022, 12, 938. [Google Scholar] [CrossRef]
  230. Recommended Methods for the Identification and Analysis of Cannabis and Cannabis Products: Manual for Use by National Drug Analysis Laboratories; United Nations Office on Drugs and Crime, Ed.; Rev. and updated; United Nations: New York, NY, USA, 2009; ISBN 978-92-1-148242-3. [Google Scholar]
  231. Ediz, E.F.; Demirel Kars, M.; Çetin, K. Development and Characterization of New Cannabidiol-Loaded Polycaprolactone Nanoparticles for Increased Bioavailability. ChemistrySelect 2024, 9, e202403609. [Google Scholar] [CrossRef]
  232. Analakkattillam, S.; Langsi, V.K.; Hanrahan, J.P.; Moore, E. Analytical Method Validation for Assay Determination of Cannabidiol and Tetrahydrocannabinol in Hemp Oil Infused Products by RP-HPLC. Sci. Rep. 2022, 12, 12453. [Google Scholar] [CrossRef] [PubMed]
  233. Sumontri, S.; Eiamart, W.; Tadtong, S.; Samee, W. Ultra-High-Performance Liquid Chromatography–Tandem Mass Spectrometry Analysis of Δ9-Tetrahydrocannabinol and Cannabidiol in Commercial Suk-Saiyasna Herbal Remedy: Applying Hansen Solubility Parameters for Sample Extraction to Ensure Regulatory Compliance. Pharmaceuticals 2024, 17, 1502. [Google Scholar] [CrossRef]
  234. Cheng, A.; Zhang, S.; Meng, F.; Xing, M.; Liu, H.; Yang, G.; Gao, Y. Nanosuspension-Loaded Dissolving Microneedle Patches for Enhanced Transdermal Delivery of a Highly Lipophilic Cannabidiol. Int. J. Nanomed. 2024, 19, 4061–4079. [Google Scholar] [CrossRef]
  235. Muresan, P.; McCrorie, P.; Smith, F.; Vasey, C.; Taresco, V.; Scurr, D.J.; Kern, S.; Smith, S.; Gershkovich, P.; Rahman, R. Development of Nanoparticle Loaded Microneedles for Drug Delivery to a Brain Tumour Resection Site. Eur. J. Pharm. Biopharm. 2023, 182, 53–61. [Google Scholar] [CrossRef] [PubMed]
  236. Hameed al-maraseemi, B.; Usmani, Z.; Rizwanullah, M.; Amin, S.; Mir, S.R. Box-Behnken Design Assisted Development and Optimization of RP-HPLC-PDA Technique for Determination of Cannabidiol in the Bulk and Nanoformulation. Egypt. J. Chem. 2022, 65, 1–2. [Google Scholar] [CrossRef]
  237. Raslan-Jaramillo, J.J.; Ríos-Gajardo, G.A.; Avello, M.A.; de Diego, M.G. Determination of Cannabinoids in Cannabis sativa Oil and Infused Ice Cream by LC-DAD Method. J. AOAC Int. 2024, 107, 140–145. [Google Scholar] [CrossRef]
  238. Abeysekera, S.K.; Robinson, A.; Ooi, M.P.-L.; Kuang, Y.C.; Manley-Harris, M.; Holmes, W.; Hirst, E.; Nowak, J.; Caddie, M.; Steinhorn, G.; et al. Sparse Reproducible Machine Learning for near Infrared Hyperspectral Imaging: Estimating the Tetrahydrocannabinolic Acid Concentration in Cannabis sativa L. Ind. Crops Prod. 2023, 192, 116137. [Google Scholar] [CrossRef]
  239. Johnson, D.A.; Hogan, M.; Marriot, R.; Heaney, L.M.; Bailey, S.J.; Clifford, T.; James, L.J. A Comparison of Advertised versus Actual Cannabidiol (CBD) Content of Oils, Aqueous Tinctures, e-Liquids and Drinks Purchased in the UK. J. Cannabis Res. 2023, 5, 28. [Google Scholar] [CrossRef] [PubMed]
  240. Khabir, Z.; Partalis, C.; Panchal, J.V.; Deva, A.; Khatri, A.; Garcia-Bennett, A. Enhanced Skin Penetration of Cannabidiol Using Organosilane Particles as Transdermal Delivery Vehicles. Pharmaceutics 2023, 15, 798. [Google Scholar] [CrossRef] [PubMed]
  241. Shreiber-Livne, I.; Sulimani, L.; Shapira, A.; Procaccia, S.; Meiri, D.; Sosnik, A. Poly(ethylene glycol)-b-Poly(epsilon-caprolactone) Nanoparticles as a Platform for the Improved Oral Delivery of Cannabidiol. Drug Deliv. Transl. Res. 2023, 13, 3192–3203. [Google Scholar] [CrossRef]
  242. Zheng, H.; Chen, B.; Rao, J. Nutraceutical Potential of Industrial Hemp (Cannabis sativa L.) Extracts: Physicochemical Stability and Bioaccessibility of Cannabidiol (CBD) Nanoemulsions. Food Funct. 2022, 13, 4502–4512. [Google Scholar] [CrossRef] [PubMed]
  243. Vella Szijj, J.; Gallo, L.L.; Buhagiar, P.I.; Szyrner, K.; Sammut Bartolo, N.; Ronsisvalle, S.; Serracino Inglott, A.; Azzopardi, L.M. Analysis of Cannabinoids in Oil. Integr. Med. Rep. 2024, 3, 122–131. [Google Scholar] [CrossRef]
  244. Tejada Rodriguez, E.; Vella Szijj, J.; Cachia, M.; Falzon, P.; Axisa, K.; Serracino-Inglott, A.; Azzopardi, L.M. An Efficient HPLC-UV Method for Determination of Tetrahydrocannabinol in Oil. Asian J. Pharm. Clin. Res. 2023, 16, 110–115. [Google Scholar] [CrossRef]
  245. Morakul, B.; Junyaprasert, V.B.; Sakchaisri, K.; Teeranachaideekul, V. Cannabidiol-Loaded Nanostructured Lipid Carriers (NLCs) for Dermal Delivery: Enhancement of Photostability, Cell Viability, and Anti-Inflammatory Activity. Pharmaceutics 2023, 15, 537. [Google Scholar] [CrossRef]
  246. Sucontphunt, A.; Sueree, L.; Jongrungruangchok, S.; Maha, A.; Madaka, F.; Chankana, N.; Chaiyabutr, K.; Leelawat, S.; Songsak, T.; Pradubyat, N. Validation and Comparison of Method Using Different Wavelength Detection at 210 and 228 Nm for Quantitative Analysis of CBD in Sublingual Cannabis Oil Products. Interprof. J. Health Sci. 2022, 20, 28–35. [Google Scholar]
  247. Mostafaei Dehnavi, M.; Ebadi, A.; Peirovi, A.; Taylor, G.; Salami, S.A. THC and CBD Fingerprinting of an Elite Cannabis Collection from Iran: Quantifying Diversity to Underpin Future Cannabis Breeding. Plants 2022, 11, 129. [Google Scholar] [CrossRef]
  248. Tajik, T.; Baghaei, K.; Moghadam, V.E.; Farrokhi, N.; Salami, S.A. Extracellular Vesicles of Cannabis with High CBD Content Induce Anticancer Signaling in Human Hepatocellular Carcinoma. Biomed. Pharmacother. 2022, 152, 113209. [Google Scholar] [CrossRef] [PubMed]
  249. Babayeva, M.; Srdanovic, I. Non-Linear Plasma Protein Binding of Cannabidiol. J. Cannabis Res. 2024, 6, 27. [Google Scholar] [CrossRef] [PubMed]
  250. Tai, W.; Yau, G.T.Y.; Arnold, J.C.; Chan, H.-K.; Kwok, P.C.L. High-Loading Cannabidiol Powders for Inhalation. Int. J. Pharm. 2024, 660, 124370. [Google Scholar] [CrossRef] [PubMed]
  251. Tai, W.; Arnold, J.C.; Chan, H.-K.; Kwok, P.C.L. Spray Freeze Dried Cannabidiol with Dipalmitoylphosphatidylcholine (DPPC) for Inhalation and Solubility Enhancement. Int. J. Pharm. 2024, 659, 124235. [Google Scholar] [CrossRef]
  252. Sainz Martinez, A.; Lanaridi, O.; Stagel, K.; Halbwirth, H.; Schnürch, M.; Bica-Schröder, K. Extraction Techniques for Bioactive Compounds of Cannabis. Nat. Prod. Rep. 2023, 40, 676–717. [Google Scholar] [CrossRef]
  253. Al Ubeed, H.M.S.; Bhuyan, D.J.; Alsherbiny, M.A.; Basu, A.; Vuong, Q.V. A Comprehensive Review on the Techniques for Extraction of Bioactive Compounds from Medicinal Cannabis. Molecules 2022, 27, 604. [Google Scholar] [CrossRef]
  254. Liu, Y.; Liu, H.-Y.; Li, S.-H.; Ma, W.; Wu, D.-T.; Li, H.-B.; Xiao, A.-P.; Liu, L.-L.; Zhu, F.; Gan, R.-Y. Cannabis sativa Bioactive Compounds and Their Extraction, Separation, Purification, and Identification Technologies: An Updated Review. TrAC Trends Anal. Chem. 2022, 149, 116554. [Google Scholar] [CrossRef]
  255. Nahar, L.; Uddin, S.J.; Alam, M.A.; Sarker, S.D. Extraction of Naturally Occurring Cannabinoids: An Update. Phytochem. Anal. 2021, 32, 228–241. [Google Scholar] [CrossRef]
  256. Vella Szijj, J. Challenges of Extracting and Determining Cannabinoids in Different Matrices. Cannabis Cannabinoid Res. 2024, 9, 1470–1477. [Google Scholar] [CrossRef]
  257. Tanney, C.A.S.; Backer, R.; Geitmann, A.; Smith, D.L. Cannabis Glandular Trichomes: A Cellular Metabolite Factory. Front. Plant Sci. 2021, 12, 721986. [Google Scholar] [CrossRef]
  258. Tahir, M.N.; Shahbazi, F.; Rondeau-Gagné, S.; Trant, J.F. The Biosynthesis of the Cannabinoids. J. Cannabis Res. 2021, 3, 7. [Google Scholar] [CrossRef] [PubMed]
  259. Reverchon, E.; De Marco, I. Supercritical Fluid Extraction and Fractionation of Natural Matter. J. Supercrit. Fluids 2006, 38, 146–166. [Google Scholar] [CrossRef]
  260. Pilařová, V.; Hadysová, Z.; Švec, F.; Nováková, L. Supercritical Fluids in Analysis of Cannabinoids in Various Cannabis Products. Anal. Chim. Acta 2022, 1232, 340452. [Google Scholar] [CrossRef]
  261. Rovetto, L.J.; Aieta, N.V. Supercritical Carbon Dioxide Extraction of Cannabinoids from Cannabis sativa L. J. Supercrit. Fluids 2017, 129, 16–27. [Google Scholar] [CrossRef]
  262. Ciolino, L.A.; Ranieri, T.L.; Taylor, A.M. Commercial Cannabis Consumer Products Part 2: HPLC-DAD Quantitative Analysis of Cannabis Cannabinoids. Forensic Sci. Int. 2018, 289, 438–447. [Google Scholar] [CrossRef]
  263. Mudge, E.M.; Murch, S.J.; Brown, P.N. Leaner and Greener Analysis of Cannabinoids. Anal. Bioanal. Chem. 2017, 409, 3153–3163. [Google Scholar] [CrossRef]
  264. Cannabis Flower Monography 3028 (07/2024). In European Pharmacopoeia 11.5; Council of Europe: Strasbourg, France, 2024.
  265. Hazekamp, A.; Peltenburg, A.; Verpoorte, R.; Giroud, C. Chromatographic and Spectroscopic Data of Cannabinoids from Cannabis sativa L. J. Liq. Chromatogr. Relat. Technol. 2005, 28, 2361–2382. [Google Scholar] [CrossRef]
  266. Raharjo, T.J.; Verpoorte, R. Methods for the Analysis of Cannabinoids in Biological Materials: A Review. Phytochem. Anal. 2004, 15, 79–94. [Google Scholar] [CrossRef]
  267. Fekete, S.; Schappler, J.; Veuthey, J.-L.; Guillarme, D. Current and Future Trends in UHPLC. TrAC Trends Anal. Chem. 2014, 63, 2–13. [Google Scholar] [CrossRef]
  268. D’Atri, V.; Fekete, S.; Clarke, A.; Veuthey, J.-L.; Guillarme, D. Recent Advances in Chromatography for Pharmaceutical Analysis. Anal. Chem. 2019, 91, 210–239. [Google Scholar] [CrossRef]
  269. Owolabi, A.; Ogunsola, O.; Joens, E.; Kotler, M.; Song, L. A Systematic Study of Liquid Chromatography in Search of the Best Separation of Cannabinoids for Potency Testing of Hemp-Based Products. Molecules 2025, 30, 758. [Google Scholar] [CrossRef]
  270. De Luca, C.; Buratti, A.; Krauke, Y.; Stephan, S.; Monks, K.; Brighenti, V.; Pellati, F.; Cavazzini, A.; Catani, M.; Felletti, S. Investigating the Effect of Polarity of Stationary and Mobile Phases on Retention of Cannabinoids in Normal Phase Liquid Chromatography. Anal. Bioanal. Chem. 2022, 414, 5385–5395. [Google Scholar] [CrossRef] [PubMed]
  271. De Luca, C.; Buratti, A.; Umstead, W.; Franco, P.; Cavazzini, A.; Felletti, S.; Catani, M. Investigation of Retention Behavior of Natural Cannabinoids on Differently Substituted Polysaccharide-Based Chiral Stationary Phases under Reversed-Phase Liquid Chromatographic Conditions. J. Chromatogr. A 2022, 1672, 463076. [Google Scholar] [CrossRef] [PubMed]
  272. Russo, F.; Tolomeo, F.; Angela Vandelli, M.; Biagini, G.; Laganà, A.; Laura Capriotti, A.; Cerrato, A.; Carbone, L.; Perrone, E.; Cavazzini, A.; et al. Enantioseparation of Chiral Phytocannabinoids in Medicinal Cannabis. J. Chromatogr. B 2023, 1221, 123682. [Google Scholar] [CrossRef] [PubMed]
  273. Umstead, W.J. The Chiral Separation of the (+) and (−)Enantiomers of Cannabidiol. Cannabis Sci. Technol. 2022, 5, 30–34. [Google Scholar]
  274. Welch, C.J.; Brkovic, T.; Schafer, W.; Gong, X. Performance to Burn? Re-Evaluating the Choice of Acetonitrile as the Platform Solvent for Analytical HPLC. Green Chem. 2009, 11, 1232–1238. [Google Scholar] [CrossRef]
  275. Dolan, J.W. Selectivity in Reversed-Phase LC Separations (Part 1): Solvent-Type Selectivity. LCGC Eur. 2010, 23, 581–584. [Google Scholar]
  276. Franklin, E.; Wilcox, M. Gradient HPLC-UV Method for Cannabinoid Profiling. In Chromatography Today; 2019; pp. 34–37. Available online: https://www.chromatographytoday.com/article/hplc-uhplc/31/regis-technologies-inc/gradient-hplc-uv-method-for-cannabinoid-profiling/2599 (accessed on 28 February 2025).
  277. Tzimas, P.S.; Petrakis, E.A.; Halabalaki, M.; Skaltsounis, L.A. Effective Determination of the Principal Non-Psychoactive Cannabinoids in Fiber-Type Cannabis sativa L. by UPLC-PDA Following a Comprehensive Design and Optimization of Extraction Methodology. Anal. Chim. Acta 2021, 1150, 338200. [Google Scholar] [CrossRef]
  278. Ryu, B.R.; Islam, M.J.; Azad, M.O.K.; Go, E.-J.; Rahman, M.H.; Rana, M.S.; Lim, Y.-S.; Lim, J.-D. Conversion Characteristics of Some Major Cannabinoids from Hemp (Cannabis sativa L.) Raw Materials by New Rapid Simultaneous Analysis Method. Molecules 2021, 26, 4113. [Google Scholar] [CrossRef]
  279. Clifford, B.; Young, C.; Owens, A.; Mott, J.; Lieberman, R. How to Choose the Right Instrumentation for Cannabinoid and Terpene Analysis. Cannabis Sci. Technol. 2020, 3, 34–42. [Google Scholar]
  280. De Backer, B.; Debrus, B.; Lebrun, P.; Theunis, L.; Dubois, N.; Decock, L.; Verstraete, A.; Hubert, P.; Charlier, C. Innovative Development and Validation of an HPLC/DAD Method for the Qualitative and Quantitative Determination of Major Cannabinoids in Cannabis Plant Material. J. Chromatogr. B 2009, 877, 4115–4124. [Google Scholar] [CrossRef]
  281. Križman, M. A Simplified Approach for Isocratic HPLC Analysis of Cannabinoids by Fine Tuning Chromatographic Selectivity. Eur. Food Res. Technol. 2020, 246, 315–322. [Google Scholar] [CrossRef]
  282. Berman, P.; Futoran, K.; Lewitus, G.M.; Mukha, D.; Benami, M.; Shlomi, T.; Meiri, D. A New ESI-LC/MS Approach for Comprehensive Metabolic Profiling of Phytocannabinoids in Cannabis. Sci. Rep. 2018, 8, 14280. [Google Scholar] [CrossRef] [PubMed]
  283. Thomas, B.F.; Compton, D.R.; Martin, B.R. Characterization of the Lipophilicity of Natural and Synthetic Analogs of Delta 9-Tetrahydrocannabinol and Its Relationship to Pharmacological Potency. J. Pharmacol. Exp. Ther. 1990, 255, 624–630. [Google Scholar] [CrossRef]
  284. Chandran, S.; Singh, R.S.P. Comparison of Various International Guidelines for Analytical Method Validation. Pharmazie 2007, 62, 4–14. [Google Scholar] [CrossRef]
  285. AOAC Official Methods of Analysis. Appendix F: Guidelines for Standard Method Performance Requirements; AOAC International: Rockville, MD, USA, 2016; Available online: https://www.aoac.org/resources/guidelines-for-standard-method-performance-requirements/ (accessed on 7 March 2025).
  286. Ojha, A.; Bhargava, S. Chapter 3—International Council for Harmonisation (ICH) Guidelines. In Regulatory affairs in the Pharmaceutical Industry; Elsevier: Amsterdam, The Netherlands, 2022; pp. 47–74. [Google Scholar]
  287. International Conference for Harmonisation. ICH Q2(R1) Validation of Analytical Procedures: Text and Methodology; European Medicines Agency: Amsterdam, The Netherlands, 1995.
  288. International Conference for Harmonisation. ICH Q2(R2) Guideline on Validation of Analytical Procedures; European Medicines Agency: Amsterdam, The Netherlands, 2024. Available online: https://www.ema.europa.eu/en/ich-q2r2-validation-analytical-procedures-scientific-guideline (accessed on 7 March 2025).
  289. International Conference for Harmonisation. ICH Guideline M10 on Bioanalytical Method Validation and Study Sample Analysis; European Medicines Agency: Amsterdam, The Netherlands, 2023. Available online: https://www.ema.europa.eu/en/ich-m10-bioanalytical-method-validation-scientific-guideline (accessed on 7 March 2025).
  290. AOAC Official Methods of Analysis. Appendix K: Guidelines for Dietary Supplements and Botanicals; AOAC International: Rockville, MD, USA, 2013. [Google Scholar]
  291. AOAC SMPR® 2019.003; Standard Method Performance Requirements (SMPRs®) for Quantitation of Cannabinoids in Plant Materials of Hemp (Low THC Varieties Cannabis sp.). AOAC International: Rockville, MD, USA, 2019. Available online: https://www.aoac.org/resources/smpr-2019003/ (accessed on 7 March 2025).
  292. ISO/IEC 17025:2017; General Requirements for the Competence of Testing and Calibration Laboratories. International Organization for Standardization: Geneva, Switzerland, 2017.
  293. Analytical Procedures and Methods Validation for Drugs and Biologics—Guidance for Industry; Food and Drug Administration: Silver Spring, MD, USA, 2015. Available online: https://www.fda.gov/media/87801/download (accessed on 7 March 2025).
  294. ORA Lab Manual Vol. II—Methods, Method Verification and Validation (ORALAB.5.4.5); Food and Drug Administration: Silver Spring, MD, USA, 2023. Available online: https://www.fda.gov/media/73920/download (accessed on 7 March 2025).
  295. Agência Nacional de Vigilância Sanitária. RDC No 166, de 24 de Julho de 2017. Dispõe Sobre a Validação de Métodos Analíticos e Dá Outras Providências. 2017. Available online: https://bvsms.saude.gov.br/bvs/saudelegis/anvisa/2017/rdc0166_24_07_2017.pdf (accessed on 7 March 2025).
  296. Magnusson, B.; Örnemark, U. (Eds.) Eurachem Guide: The Fitness for Purpose of Analytical Methods—A Laboratory Guide to Method Validation and Related Topics, 2nd ed.; Eurochem: Zug, Switzerland, 2014; ISBN 978-91-87461-59-0. [Google Scholar]
  297. Weitzel, M.L.J. The Estimation and Use of Measurement Uncertainty for a Drug Substance Test Procedure Validated According to USP. Accred. Qual. Assur. 2012, 17, 139–146. [Google Scholar] [CrossRef]
  298. Standard Practices for Method Validation in Forensic Toxicology. ANSI/ASB Standard 036, 1st ed.; American Academy of Forensic Science: Colorado Springs, CO, USA, 2019; Available online: https://www.aafs.org/asb-standard/standard-practices-method-validation-forensic-toxicology (accessed on 7 March 2025).
  299. Analytical Quality Control and Method Validation Procedures for Pesticide Residues Analysis in Food and Feed. SANTE 11312/2021; European Reference Laboratories for Residues in Pesticides, 2021; Available online: https://www.eurl-pesticides.eu/docs/public/tmplt_article.asp?CntID=727 (accessed on 7 March 2025).
  300. Guidance for the Implementation of a Quality Management System in Drug Testing Laboratories; United Nations Office on Drugs and Crime: Vienna, Austria, 2009; Available online: https://www.unodc.org/unodc/en/scientists/guidance-for-the-implementation-of-a-quality-management-system-in-drug-testing-laboratories_new.html (accessed on 7 March 2025).
  301. Scientific Working Group for Forensic Toxicology. Scientific Working Group for Forensic Toxicology (SWGTOX) Standard Practices for Method Validation in Forensic Toxicology. J. Anal. Toxicol. 2013, 37, 452–474. [Google Scholar] [CrossRef]
  302. Peters, F.T.; Drummer, O.H.; Musshoff, F. Validation of New Methods. Forensic Sci. Int. 2007, 165, 216–224. [Google Scholar] [CrossRef] [PubMed]
  303. Trufelli, H.; Palma, P.; Famiglini, G.; Cappiello, A. An Overview of Matrix Effects in Liquid Chromatography–Mass Spectrometry. Mass Spectrom. Rev. 2011, 30, 491–509. [Google Scholar] [CrossRef]
  304. Choi, S.-I.; Han, X.; Lee, S.-J.; Men, X.; Oh, G.; Lee, D.-S.; Lee, O.-H. Validation of an Analytical Method for the Determination of Thiabendazole in Various Food Matrices. Separations 2022, 9, 135. [Google Scholar] [CrossRef]
  305. Dawson, D.D.; Martin, R.W. Investigation of Chocolate Matrix Interference on Cannabinoid Analytes. J. Agric. Food Chem. 2020, 68, 5699–5706. [Google Scholar] [CrossRef] [PubMed]
  306. Ramella, A.; Roda, G.; Pavlovic, R.; Dei Cas, M.; Casagni, E.; Mosconi, G.; Cecati, F.; Minghetti, P.; Grizzetti, C. Impact of Lipid Sources on Quality Traits of Medical Cannabis-Based Oil Preparations. Molecules 2020, 25, 2986. [Google Scholar] [CrossRef]
  307. Williams, M.L.; Olomukoro, A.A.; Emmons, R.V.; Godage, N.H.; Gionfriddo, E. Matrix Effects Demystified: Strategies for Resolving Challenges in Analytical Separations of Complex Samples. J. Sep. Sci. 2023, 46, 2300571. [Google Scholar] [CrossRef]
  308. Matuszewski, B.K.; Constanzer, M.L.; Chavez-Eng, C.M. Strategies for the Assessment of Matrix Effect in Quantitative Bioanalytical Methods Based on HPLC−MS/MS. Anal. Chem. 2003, 75, 3019–3030. [Google Scholar] [CrossRef]
  309. Cortese, M.; Gigliobianco, M.R.; Magnoni, F.; Censi, R.; Di Martino, P. Compensate for or Minimize Matrix Effects? Strategies for Overcoming Matrix Effects in Liquid Chromatography-Mass Spectrometry Technique: A Tutorial Review. Molecules 2020, 25, 3047. [Google Scholar] [CrossRef] [PubMed]
  310. Nasiri, A.; Jahani, R.; Mokhtari, S.; Yazdanpanah, H.; Daraei, B.; Faizi, M.; Kobarfard, F. Overview, Consequences, and Strategies for Overcoming Matrix Effects in LC-MS Analysis: A Critical Review. Analyst 2021, 146, 6049–6063. [Google Scholar] [CrossRef] [PubMed]
  311. Gałuszka, A.; Migaszewski, Z.; Namieśnik, J. The 12 Principles of Green Analytical Chemistry and the SIGNIFICANCE Mnemonic of Green Analytical Practices. TrAC Trends Anal. Chem. 2013, 50, 78–84. [Google Scholar] [CrossRef]
  312. Safaei, Z.; Bocian, S.; Buszewski, B. Green Chromatography-Carbon Footprint of Columns Packed with Core–Shell Materials. RSC Adv. 2014, 4, 53915–53920. [Google Scholar] [CrossRef]
  313. Kalisz, O.; Tobiszewski, M.; Nowaczyk, A.; Bocian, S. Exploring the Potential of Green Chemistry in Reversed-Phase Liquid Chromatography: A Review of Sustainable Solvents. TrAC Trends Anal. Chem. 2024, 181, 118007. [Google Scholar] [CrossRef]
  314. Nowak, P.M.; Wietecha-Posłuszny, R.; Pawliszyn, J. White Analytical Chemistry: An Approach to Reconcile the Principles of Green Analytical Chemistry and Functionality. TrAC Trends Anal. Chem. 2021, 138, 116223. [Google Scholar] [CrossRef]
  315. Prat, D.; Wells, A.; Hayler, J.; Sneddon, H.; McElroy, C.R.; Abou-Shehada, S.; Dunn, P.J. CHEM21 Selection Guide of Classical- and Less Classical-Solvents. Green Chem. 2016, 18, 288–296. [Google Scholar] [CrossRef]
  316. Keith, L.H.; Gron, L.U.; Young, J.L. Green Analytical Methodologies. Chem. Rev. 2007, 107, 2695–2708. [Google Scholar] [CrossRef] [PubMed]
  317. Gałuszka, A.; Migaszewski, Z.M.; Konieczka, P.; Namieśnik, J. Analytical Eco-Scale for Assessing the Greenness of Analytical Procedures. TrAC Trends Anal. Chem. 2012, 37, 61–72. [Google Scholar] [CrossRef]
  318. Pena-Pereira, F.; Wojnowski, W.; Tobiszewski, M. AGREE—Analytical GREEnness Metric Approach and Software. Anal. Chem. 2020, 92, 10076–10082. [Google Scholar] [CrossRef]
  319. Płotka-Wasylka, J. A New Tool for the Evaluation of the Analytical Procedure: Green Analytical Procedure Index. Talanta 2018, 181, 204–209. [Google Scholar] [CrossRef]
  320. Sammut Bartolo, N.; Gallo, L.L.; Szyrner, K.; Buhagiar, P.I.; Vella Szijj, J. Greenness Assessment of Analytical Methods for Determination of Cannabinoids in Oils Using NEMI, Analytical Eco-Scale, AGREE and GAPI. Anal. Methods 2024, 16, 5931–5942. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of key cannabinoids.
Figure 1. Chemical structures of key cannabinoids.
Pharmaceuticals 18 00786 g001
Figure 2. Distribution of the publications reported in Table S1, categorized by the types of matrices used and analyzed.
Figure 2. Distribution of the publications reported in Table S1, categorized by the types of matrices used and analyzed.
Pharmaceuticals 18 00786 g002
Figure 3. (a) Distribution of the reported extraction techniques based on the matrices employed: (a) Plant materials; (b) Oils and resins; (c) Food products; (d) Pharmaceutical and other formulated products. DSLME: dispersive solid–liquid microextraction; US: ultrasound; US-LLE: ultrasound liquid–liquid extraction; US-SLE: ultrasound solid–liquid extraction; SFE: supercritical fluid extraction (scCO2).
Figure 3. (a) Distribution of the reported extraction techniques based on the matrices employed: (a) Plant materials; (b) Oils and resins; (c) Food products; (d) Pharmaceutical and other formulated products. DSLME: dispersive solid–liquid microextraction; US: ultrasound; US-LLE: ultrasound liquid–liquid extraction; US-SLE: ultrasound solid–liquid extraction; SFE: supercritical fluid extraction (scCO2).
Pharmaceuticals 18 00786 g003
Figure 4. Flow diagram illustrating the review methodology, including the database searches, the number of abstracts analyzed, and the full texts assessed. HPLC: High-Performance Liquid Chromatography; Prep: Preparative.
Figure 4. Flow diagram illustrating the review methodology, including the database searches, the number of abstracts analyzed, and the full texts assessed. HPLC: High-Performance Liquid Chromatography; Prep: Preparative.
Pharmaceuticals 18 00786 g004
Table 1. Summary of LC parameters used in gradient chromatographic runs.
Table 1. Summary of LC parameters used in gradient chromatographic runs.
EntryMethod aMobile Phase bColumnFlow Rate
(mL/min) c
Run Time
(min) d
Injection Vol. (µL)Temp. (°C)UV Detection
(nm) e
Cannabinoids
Analyzed f
Ref.
Aqueous Phase (A) Organic Phase (B)Gradient
% B
1HPLC-UV/Vis ACN10–90Agilent C18 (150 × 46 mm, 5 μm)NR201035NR5: CBD, CBDA, CBN, Δ9-THC, THCA[80]
2HPLC-VWD ACN65–75Restek Raptor ARC-18 (150 × 4.6 mm, 5 μm) with a guard col115 dNRNR2286: CBC, CBD, CBG, CBN, Δ8-THC, Δ9-THC[81]
3HPLC-DAD ACN70–80InfinityLab Poroshell 120 C18 (100 × 4.6 mm, 2.7 μm) with a guard col1162040220: CBD, Δ9-THC
283: CBC, CBN
4: CBC, CBD, CBG, CBN[82]
4HPLC-DAD0.01% FAACN-0.01% FA65–100Accucore C18 (100 × 2.1 mm, 2.6 μm)0.312 d1502304: CBD, CBDA, Δ9-THC, THCA[83,84]
5UHPLC-UV0.05% FAACN-0.05% FA70–100Waters Cortecs UPLC C18 (100 × 2.1 mm, 1.6 μm)0.312.5103522817: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, CBNA, CBL, CBLA, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[85]
6HPLC-DAD0.075% FAACN-0.5% FA70–100Excel Super C18 (250 × 4.6 mm, 3 μm)114 d104021012: CBC, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, CBNA, Δ9-THC, THCA, THCV[86]
7HPLC-DAD0.1% FAACN65–95Luna C18 (150 × 4.6 mm, 5 μm) with a guard col1282025210: CBD, Δ9-THC
220: CBDA, THCA, CBN
5: CBD, CBDA, CBN, Δ9-THC, THCA[87]
8HPLC-DAD0.1% FAACN70–95InfinityLab Poroshell EC-C18 (150 × 3.0 mm, 2.7 μm) with a guard column0.5255352287: CBD, CBDA, CBG, CBGA, CBN, Δ9-THC, THCA[88,89]
9HPLC-DAD0.1% FAACN-0.1% FA2–100Eclipse XDB-C18 (150 × 4.6 mm, 3.5
μm)
0.65110NR2804: CBD, CBDA, CBEA, DHC[90,91]
10UHPLC-DAD0.1% FAACN-0.1% FA5–100Ascentis Express RP Amide (100 × 2.1 mm, 2.7 μm)0.465NRNR220/269/306: CBDA
220/268/305: CBGA
220/271/304: THCA
3: CBDA, CBGA, THCA f[92,93]
11UHPLC-DAD0.1% FAACN-0.1% FA5–100Phenomenex XB-C18 (50 × 2.1 mm, 1.7 μm)NR18 dNRNR210, 2737: CBC, CBD, CBDV, CBG, CBN, cis-Δ9-THC, Δ9-THC f[94]
12HPLC-UV/Vis0.1% FAACN-0.1% FA25–100µ-Bondapak C18 (300 × 3.9 mm)24510NR22514: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, Δ8-THC, ∆9-THC, THCA, THCV, THCVA[95]
13HPLC-DAD0.1% FAACN-0.1% FA40–90Zorbax Eclipse XDB C18 (150 × 4.6 mm, 5 μm)0.73510–1535NR4: CBD, CBDA, CBN, ∆9-THC[96]
14HPLC-UV f0.1% FAACN-0.1% FA60–90Ascentis Express C18 (150 × 3.0 mm, 2.7 μm)0.440 or 45 d3NR210: NC
220: AC
7: CBD, CBDA, CBG, CBGA, CBN, Δ9-THC, THCA[97,98]
15UHPLC-DAD0.1% FAACN-0.1% FA60–90Fortis SpeedCore C18-PFP (100 mm × 2.1 mm, 2.6 μm)0.411330210: 6 NC
225: AC, CBC, CBN
12: CBC, CBD, CBDA, CBCT, CBDV, CBDVA, CBE, CBGA, CBN, Δ9-cis-THC, Δ9-THC, THCA[99,100]
16HPLC-DAD0.1% FAACN-0.1% FA65–95Poroshell 120 SB-C18, (150 × 4.6 mm, 2.7 μm)0.5182NR2149: CBC, CBD, CBDA, CBG, CBGA, CBN, Δ9-THC, THCA, THCV[101,102]
17HPLC-DAD0.1% FAACN-0.1% FA65–100Zorbax Eclipse Plus C18 (50 × 2.1 mm, 1.8 μm)0.47.5240210: CBD, Δ9-THC
221: CBDA, THCA
4: CBD, CBDA, Δ9-THC, THCA[103,104]
18UHPLC-DAD0.1% FAACN-0.1% FA68–95InfinityLab Poroshell 120 EC-C18 (150 × 2.1 mm, 2.7 μm) with a guard col0.51813021411: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[105,106]
19HPLC-DAD0.1% FAACN-0.1% FA70–77Phenomenex Luna C18(2) (150 × 4.6 mm, 3 μm) with a guard col1.222.210NR22011: CBC, CBCA, CBD, CBDA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[107]
20HPLC-DAD0.1% FAACN-0.1% FA70–90Poroshell 120 EC-C18 (150 × 4.6 mm, 2.7 μm)0.46010252205: CBD, CBDA, CBN, Δ9-THC, THCA[108]
21HPLC-DAD0.1% FAACN-0.1% FA70–90Supersil ODS2 (250 × 4.6 mm, 5 μm)13010352205: CBD, CBDA, CBN, Δ9-THC, THCA[109]
22UHPLC-UV0.1% FAACN-0.1% FA70–100Waters Acquity UPLC C18 (150 × 2.1 mm, 1.7 μm)0.414NR30NR6: CBD, CBDA, CBGA, CBN, Δ9-THC, THCA[110]
23HPLC-DAD0.1% FAACN-0.1% FA70–100Phenomenex Luna Omega C18 (150 × 2.1 mm × 1.6 μm)0.48540214, 280 e16: CBC, CBCA, CBD, CBDA, CBDV CBDVA, CBG, CBGA, CBL, CBN, CBNA, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[111]
24HPLC-DAD0.1% FAACN-0.1% FA70–100Waters SunFire C18 (150 × 3.0 mm, 3.5 μm) with a guard colNR29 d2NR2253: CBD, CBG, Δ9-THC f[112]
25HPLC-UV/Vis0.1% FAACN-0.1% FA72–90Zorbax XDB-C18 (150 mm × 4.6 mm, 3.5 μm)1.512 d103022813: CBC, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBN, Δ8-THC, Δ9-THC, THCA, THCV f[113]
26HPLC-DAD0.1% FAACN-0.1% FA80–100Ultimate LP-C18 (150 × 4.6 mm, 2.5 μm)12810NR2286: CBD, CBDA, CBG, CBN, Δ9-THC, THCA f[114]
27HPLC-DAD1 mM FA
(pH 3.53)
ACN70–99Hypersil C18 (150 × 4.6 mm, 3 μm) with a guard col1181025NR14: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[115,116]
28HPLC-DADAF 2 mM–0.1% FAACN-0.1% FA70–90Ascentis Express C18 (150 × 3.0 mm, 2.7 μm)0.320625210: NC
220: AC
12: CBC, CBCA, CBCV, CBD, CBDA, CBDV, CBGA, CBGV, CBN, Δ8-THC, Δ9-THC, THCA f[117]
29HPLC-VWDAF 2 mM–0.1% FAACN-0.1% FA70–90Ascentis Express C18 (150 × 3.0 mm, 2.7 μm)0.2353NR2109: CBC, CBCV, CBD, CBDB, CBDV, CBG, CBGV, Δ8-THC, Δ9-THC[118]
30UHPLC-DADAF 5 mM–0.1% FAACN-0.1% FA67–95Ascentis Express C18 (150 × 3.0 mm, 2.7 μm)18 d54022816: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBN, CBNA, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[119]
31UHPLC-UVAF 5 mM–0.1% FAACN-0.1% FA70–98Ascenti Express C18 (150 × 2.1 mm, 2 μm)0.48253022817: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[120]
32HPLC-DADAF 5 mM–0.1% FAACN-0.1% FA75–95Luna C18(2) (150 × 2 mm, 3 μm) with a guard col0.25355RT22010: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ9-THC, THCA, THCVA[121]
33HPLC-DADAF 10 mM (pH 3.6)ACN-0.1% FA70–100InfinityLab Poroshell 120 EC-C18, (100 × 2.1 mm, 1.9 μm)0.811140NR16: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, Δ9-THC, THCA, THCV, THCVA[122]
34HPLC-DADAF 20 mM (pH 3.2)ACN60–95XBridge C18 (150 × 4.6 mm, 3.5 μm)1NR25252403: CBD, CBN, Δ9-THC[123,124]
35HPLC-DADAF 20 mM (pH 3.6)ACN-0.1% FANRInfinityLab Poroshell 120 EC-C18 (150 × 4.6 mm, 2.7 μm) with a guard col115 d5402206: CBD, CBDA, CBN, Δ8-THC, Δ9-THC, THCA[125,126]
36UHPLC-DADAF 50 mM, 10% ACN (pH 3.75)ACN-0.1% FA55–90Cortecs UPLC Waters C18 (100 × 2.1 mm, 1.6 μm) with a guard col0.31033027010: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA[127]
37HPLC-UV0.085% PAACN-0.085% PA70–95NexLeaf CBX Potency RP C18 (150 × 4.6 mm, 2.7 μm) with a guard col1105352209: CBC, CBD, CBDA, CBDV, CBG CBGA, CBN, Δ9-THC, THCA[128]
38HPLC-DAD0.085% PAACN-0.085% PA70–95NexLeaf CBX for Potency C18 (150 × 4.6 mm, 2.7 μm) with a guard col1.61054022011: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV f[129,130,131]
39HPLC-UV0.085% PAACN-0.085% PA70–95NexLeaf CBX for Potency C18 (150 × 4.6 mm, 2.7 μm) with a guard col1.610NR352205: CBD, CBDA, CBN, Δ9-THC, THCA[132]
40HPLC-MWD0.085% PAACN-0.085% PA75–95Phenomenex Kinetex XB-C18 (250 × 4.6 mm × 5 μm) with a guard col1.625NR3522011: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[133]
41HPLC-DAD0.1% PAACN-0.1% PA55–60Restek Raptor C18 (150 × 4.6 mm, 2.7 μm)1.5275452208: CBD, CBDV, CBG, CBN, Δ8-THC, Δ9-THC, THCA[134]
42HPLC-DAD0.1% PAACN-0.1% PA60–100InfinityLab Poroshell 120 EC-C18 (50 × 3.0 mm, 2.7 μm)NR191NR208, 220, 230, and 24026: CBC, CBCO, CBCV, CBCVA, CBD, CBDA, CBDA-ME, CBDP, CBDV, CBE, CBG, CBGA, CBGQA, CBGV, CBGVA, CBL, CBN, CBT, Δ8-THC, Δ9-THC, THCA, THCB, THCH, THCP, THCV, THCVA[135]
43HPLC-DADPA 8.64 g/LACN64–82InfinityLab Poroshell 120 EC-C18 (150 × 3 mm, 2.7 μm)0.7301040225: NC
306: AC
5: CBD, CBDA, CBN, Δ9-THC, THCA[136,137]
44HPLC-DADPA 8.6 g/LACN64–82Purospher RP-18 (250 × 4.6 mm, 5 μm) with a guard col1251040225, 3063: CBD, CBN, Δ9-THC f[138,139]
45UHPLC-DADPA 85%
(pH 2.2)
ACN68–90Raptor ARC-18 (150 × 2.1 mm, 2.7 μm)0.413125228: NC
306: AC
8: CBC, CBD, CBDA, CBDV, CBG, CBGA, Δ9-THC, THCA[140]
46HPLC-DAD0.05% AcOH (pH 4.40)ACN56–75Phenomenex Kinetex C18 (150 × 4.6 mm, 2.6 μm) with a guard col0.83552522012: CBC, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[141]
47UHPLC-TUV0.1% AcOHACN53–95Cortecs RP C18 (150 × 2.1 mm, 1.6 μm)0.4352402303: CBD, CBN, Δ9-THC[142]
48HPLC-DAD0.1% FAMeOH60–90Phenomenex Kinetex C18 (100 mm × 2.1 mm, 5.0 μm)0.575142206: CBD, CBDA, CBN, Δ8-THC, Δ9-THC, THCA[143]
49HPLC-DAD0.1% FAMeOH-0.05% FA60–95Dr Maisch ReproSil XR 120 (50 × 3.5 mm, 3 μm)1101402287: CBC, CBD, CBDA, CBG, CBN, Δ9-THC, THCA[144]
50HPLC-DAD0.1% FAMeOH-0.05% FA60–95InfinityLab Poroshell 120 EC-C18 (150 × 4.6 mm, 4 μm)1773550210, 23011: CBC, CBCA, CBD, CBDA, CBDVA, CBG, CBGA, CBN, Δ9-THC, THCA, THCVA[145]
51HPLC-VWD0.1% FAMeOH-0.05% FA60–95InfinityLab Poroshell 120 EC-C18, (50 × 3 mm, 2.7 μm)1115502305: CBD, CBDA, CBN, Δ9-THC, THCA[146]
52HPLC-DAD0.1% FAMeOH-0.05% FA60–100InfinityLab Poroshell 120 EC-C18 (50 × 3.0 mm, 2.7 μm)NR12.55NR2203: CBD, CBN, Δ9-THC[147]
53HPLC-VWD0.1% FAMeOH-0.05% FA75–95InfinityLab Poroshell 120 EC-C18, (150 × 4.6 mm)112.55502303: CBD, CBN, Δ9-THC[148]
54HPLC-DAD0.1% FAMeOH-0.1% FA62–80Accucore aQ C18 Polar Endcapped (100 × 2.1 mm, 2.6 μm)0.452725022813: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, Δ9-THC, THCA, THCV, THCVA[149]
55HPLC-UV0.1% FAMeOH67–95Accucore aQ C18 Polar Endcapped (100 × 2.1 mm, 2.6 μm)0.4523NRNR2107: CBD, CBDA, CBG, CBGA, CBN, Δ9-THC, THCA[150]
56HPLC-DAD50 mM AF
(pH 5.19)
MeOH68–95C-18 (250 × 4.6 mm, 5.0 μm)130NR302405: CBD, CBDA, CBN, Δ9-THC, THCA[151]
57UHPLC-DAD0.07% PAMeOH-0.07% PA65–95Shim-pack XR-ODSII C18 (2.2 μm)1451050NR11: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[152,153]
58HPLC-DAD10 mM AA
(pH 5.2)
MeOH72–92Phenomenex Kinetex C18 (100 × 4.6 mm, 2.6 μm)0.72810302208: CBD, CBDA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA[154]
59HPLC-DAD25 mM AAMeOH75–95Thermo Hypersil BDS C18 (150 × 4.6 mm, 5 μm)125NRNR2055: CBD, CBDA, CBN, Δ9-THC, THCA[155]
60HPLC-DAD0.1%TFAMeOH-0.1% TFA79–100Waters Cortecs C18 (150 × 4.6 mm, 2.7 μm0.721 dNR5422614: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[156]
61HPLC-DAD0.015% FAMeOH:ACN (75:25)74.5–80.5 bRestek Raptor ARC-18 (150 × 2.1 mm, 2.7 μ) with a guard col0.3, 0.5 c32330230: 8 NC;
262: CBCA, CBNA; 271: 6 AC;
284: CBC, CBN
18: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, CBT, Δ8-THC, Δ9-THC, THCA, THCV, THCVA f[157]
62HPLC-DAD0.1% FAB: ACN-0.1% FA, C: MeOH a55%B,0%C–0%B,100%C3 columns: Shim-Pak C18 (75 × 3 mm, 2.2 μm), and 2 Phenomenex Synergy C18 (100 × 3 mm, 2.5 μm) with a guard col0.3–0.41 c885–3040absorbance maxima of the analyzed CBs e17: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[158]
63HPLC-DAD0.1% FAB: ACN-0.1% FA, C: MeOH a70%B,0%C–
0%B,100%C
Agilent XDB (250 × 4.6 mm, 5.0 μm)NR33 dNRNR200–4008: CBC, CBD, CBDA, CBG, CBGA, CBN, Δ9-THC, THCA[159]
64HPLC-DAD0.1% PAB: ACN
C: MeOH a
38%B,33%C–53%B,47%CPhenomenex C18 (150 × 4.6 mm, 5 μm)1.226.510302206: CBD, CBDA, CBN, CBNA, Δ9-THC, THCA[160,161]
65HPLC-DADAF 20 mM
0.1% FA
B: ACN, C: MeOH 10 mM AF, 0.05% FA a50%B,0%C–45%B,45%CPhenomenex Luna C18(2) (150 × 4.6 mm, 5 μm) with a guard col2.534104023210: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA[162]
AA: ammonium acetate; AC: acidic cannabinoids; AcOH: acetic acid; ACN: acetonitrile; AF: ammonium formate; Col: column; DAD: diode array detector; FA: formic acid; HPLC: high-performance liquid chromatography; MeOH: methanol; MWD: multiple wavelengths detector; NC: neutral cannabinoids; NR: not reported; PA: phosphoric acid; RT: room temperature; TFA: trifluoroacetic acid; TUV: tunable ultraviolet; UHPLC: ultra high-performance liquid chromatography; UV: ultraviolet; Vis: visible; VWD: variable wavelength detector. Cannabinoids Abbreviations: CBD: cannabidiol; CBDA: cannabidiolic acid; CBDA-ME: cannabidiolic acid methyl ester; CBDP: cannabidiphorol; CBE: cannabielsoin; CBEA: cannabielsoic acid; CBDV: cannabidivarin; CBC: cannabichromene; CBCA: cannabichromenic acid; CBCO: cannabichromeorcin; CBCV: cannabichromevarin; CBCVA: cannabichromevarinic acid; CBG: cannabigerol; CBGA: cannabigerolic acid; CBGV: cannabigerovarin; CBGVA: cannabigerovarinic acid; CBL: cannabicyclol; CBLA: cannabicyclolic acid; CBN: cannabinol; CBT: cannabicitran; Δ8-THC: Δ8-tetrahydrocannabinol; Δ9-THC: Δ9-tetrahydrocannabinol; THCA: Δ9-tetrahydrocannabinolic acid; THCB: Δ9-tetrahydrocannabutol; THCH: Δ9-tetrahydrocannabihexol; THCP: Δ9-tetrahydrocannabiphorol; THCV: Δ9-tetrahydrocannabivarin; THCVA: Δ9-tetrahydrocannabivarinic acid. a Type of liquid chromatography apparatus and UV detector used; when not specified, the UV term is used; Entry 14: HPLC-VWD [98] HPLC-DAD [97]. b Entry 1–3: Only water was used as aqueous phase; Entry 61: Method used a four-step isocratic mobile phase; Entry 62–65: Method used a ternary solvent system, with two separate organic phases (B and C). c Entry 61: 0–7 min: F = 0.3 mL/min, 17.5–30 min: F = 0.5 mL/min; Entry 62: 0–17 min: 0.3 mL/min, 19–30 min: 0.35 mL/min, 32–46 min: 0.4 mL/min; 70 min: 0.41 mL/min. d When reported, run time includes column equilibration; Entry 2, 35: As indicated in the published chromatograms; Entry 4, 6, 11, 24, 25, 30, 60, and 63: No column equilibration was reported; Entry 14: Run time: 45 min [98] or 40 min [97]. e When reported, the wavelengths described correspond to those selected for quantification. Entry 23: Only CBDA and THCA were quantified by UV at 280 nm; Entry 62: DAD detection (nm): CBC: 230, 279; CBCA: 199, 254; CBD: 205, 274; CBDA: 221, 268; CBDV: 205, 273; CBDVA: 221, 268; CBG: 205, 273; CBGA: 220, 268; CBL: 210, 278; CBLA: 227, 273; CBN: 215, 284; CBNA: 262, 326; Δ8-THC: 207, 279; Δ9-THC: 209, 279; THCA: 220, 271; THCV: 205, 278; THCVA: 220, 269. f Additional cannabinoids were assigned by MS: Entry 10, 25, 26, 44, 61; Entry 9: Cannabinoids were identified by MS and semi-quantified by UV; Entry 28: The developed method enabled the separation of 12 out of 14 cannabinoids. The additional two (CBG and CBNR) were assigned by MS. Entry 38: An additional 9 cannabinoid were examined in relation to the separation of Δ8-THC, Δ9-THC, and THCA [130].
Table 2. Summary of LC parameters used in isocratic chromatographic runs.
Table 2. Summary of LC parameters used in isocratic chromatographic runs.
EntryMethod aMobile Phase bColumnFlow Rate
(mL/min) c
Run Time
(min) d
Injection Vol. (µL) Temp. (°C) eUV Detection
(nm) f
Cannabinoids
Analyzed g
Ref.
Aqueous Phase (A)Organic Phase (B)Ratio A:B
66HPLC-DAD ACN20:80Chromolith RP18 (150 × 4.6 mm, 5 μm)145NRRT2545: CBD, CBDV, CBN, Δ9-THC, THCV[163]
67HPLC-DAD ACN20:80Kinetex C18 (150 × 4.6 mm, 5 μm)1925352104: CBD, CBN, Δ9-THC, THCA[164]
68HPLC-UV ACN20:80Kromasil C8 (150 × 4.6 mm, 5 μm)11520NR2723: CBN, Δ9-THC, THCA[165]
69HPLC-DAD ACN50:50C18 (250 × 4.6 mm, 5 μm)14020NR2285: CBC, CBD, CBG, CBN, Δ9-THC[166,167]
70UHPLC-DAD0.028% FA (pH 3.6)ACN27:73Luna Omega Polar C18 (150 × 2.1 mm, 1.6 μm) with a guard col0.315330223, 230, 251, 269, 285 f15: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBLA, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[168]
71HPLC-UV0.1% FAACN16:84Zobax Eclipse Plus C18NR265NR28012: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBL, CBN, Δ8-THC, Δ9-THC, THCA, THCV[169]
72HPLC-VWD0.1% FAACN20:80Zorbax Eclipse XDB C18 (250 × 3 mm, 5 μm)0.525NR302204: CBD, CBDA, Δ9-THC, THCA[170]
73HPLC-DAD0.1% FAACN25:75Mightysil RP-8GP (150 × 2 mm, 5 μm)0.216540210: CBD, Δ9-THC
284: CBDA, CBN, THCA
5: CBD, CBDA, CBN, Δ9-THC, THCA[171]
74HPLC-DAD0.1% FAACN25:75Phenomenex Kinetex C18 (150 × 2.1 mm, 2.6 μm) with guard col0.53010202306: CBD, CBDA, CBN, Δ8-THC, Δ9-THC, THCA[172]
75UHPLC-DAD0.1% FAACN41:59Cortecs UPLC Shield RP18 (100 × 2.1 mm, 1.6 μm)0.5132352694: CBD, CBDA, Δ9-THC, THCA[173]
76UHPLC-DAD0.1% FAACN48:52Cortecs UPLC Shield RP18 (100 × 2.1 mm, 1.6 μm)0.7185352284: CBD, CBDA, Δ9-THC, THCA[174,175]
77UHPLC-DAD0.1% FAACN49–51Hypersyl Gold RP (150 × 2.1 mm, 1.9 μm)0.45255352805: CBD, CBDA, CBN, Δ9-THC, THCA[176]
78HPLC-DAD0.1% FAACN-0.1% FA17.5:82.5two HALO C18 (100 × 4.6 mm, 2.7 μm) (200 mm total length)11220NR222: CBN
230: CBC
210: other 4 NC
6: CBC, CBD, CBG, CBN, Δ8-THC, Δ9-THC[177]
79HPLC-DAD0.1% FAACN-0.1% FA23:77Phenomenex Luna C18(2) (150 × 4.6 mm, 3 μm)1.22010NR2204: CBD, CBDA, Δ9-THC, THCA[178]
80HPLC-DAD0.1% FAACN-0.1% FA25:75InfinityLab Poroshell 120 EC-18 (150 × 3.0 mm, 2.7 μm)0.612103022812: CBC, CBD, CBDA, CBDV, CBG, CBGA, CBL, CBN, Δ8-THC, Δ9-THC, THCA, THCV[179]
81HPLC-DAD0.1% FAACN-0.1% FA25:75Phenomenex Luna C18(2) (250 × 4.6 mm, 3 μm) with a guard col1, 1.2 c30104022015: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[180]
82HPLC-UV0.1% FAACN-0.1% FA30:70Phenomenex Aeris peptide XB-C18 (250 × 2.1 mm, 2.6 μm)0.352010352355: CBD, CBDA, CBN, Δ9-THC, THCA[181]
83UHPLC-DAD5 mM FAACN42:58Zorbax Eclipse Plus C18 (100 × 2.1 mm, 1.8 μm)0.5258NR2155: CBD, Δ(4)8-iso-THC Δ8-iso-THC, Δ8-THC, Δ9-THC[216]
84HPLC-DADAF 0.5 mM
0.02% FA
(pH 3.0)
ACN25:75Raptor ARC-18 (150 × 2.1 mm, 2.7 μm) with a guard col0.420430223, 230, 251, 261, 269, 285 f20: CBC, CBCA, CBCV, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, CBT, Δ8-THC, Δ9-THC, Δ8-THCA, THCA, THCV, THCVA[182]
85HPLC-VWDAF 0.5 mM–
0.1% FA
ACN-
AF 0.5 mM
0.1% FA
25:75Restek Raptor ARC18 (150 × 4.6 mm, 2.7 μm)1.59NRNR22016: CBC, CBCA, CBD, CBDA, CBDV CBDVA, CBG, CBGA, CBL CBN, CBNA, Δ8-THC, Δ9-THC THCA, THCV, THCVA[183]
86HPLC-DADAF 2 mM–
0.011% FA
(pH 3.6)
ACN27:73Luna Omega Polar C18 (150 × 2.1 mm, 1.6 μm) with guard col0.318330223, 230, 251, 269, 285 f15: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBLA, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[184]
87HPLC-UVAF 5 mM–0.1% FAACN-0.1% FA20:80ES Industries Harmony Secure RP18 (150 × 4.6 mm, 3.5 μm) with a guard col1102040210, 2283: CBD, CBN, Δ9-THC[185]
88HPLC-UVAF 5 mM–0.1% FAACN-0.1% FA20:80Harmony Secure RP18 (150 × 4.6 mm, 3.5 μm)1155040210: CBD, Δ9-THC, CBN
228: THCA, CBDA
5: CBD, CBDA, CBN, Δ9-THC, THCA[186]
89HPLC-UVAF 5 mM–0.1% FAACN-0.1% FA20:80Shimadzu Nexcol C18 (50 × 3.0 mm, 5 μm)0.385402283: CBD, CBN, Δ9-THC[185]
90UHPLC-DADAF 5 mM–0.1% FAACN-0.1% FA25:75Raptor ARC-18 (100 × 3.0 mm, 1.8 μm) with a guard col1624022815: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[187]
91HPLC-DADAF 10 mM–0.1% FAACN-0.1% FA30:70Inertsil ODS-HL (150 × 2.1 mm, 3 μm)0.335NR4022013: CBC, CBD, CBDA, CBDV
CBG, CBGA, CBL, CBN, Δ8-THC, Δ9-THC, THCA, THCV, THCVA
[188]
92HPLC-DADAF 20 mM (pH 3.6)ACN25:75Phenomenex Kinetex C18 (150 × 4.6 mm, 2.6 μm) with a guard col0.81554022010: CBC, CBD, CBDA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[189,190,191,192]
93HPLC-DADAF 20 mM–FA (pH 2.9)ACN-0.1% FA30:70Phenomenex Kinetex XB-C18 (150 × 2.1 mm, 1.7 μm)0.3162NR22816: CBC, CBCA, CBD, CBDA, CBDVA, CBG, CBGA, CBL, CBN, CBNA, Δ8-THC, Δ9-THC, THCA, THCA-C4, THCV, THCVA[193,194,195,196,197,198,199]
94HPLC-DADAF 0.1 M–
0.1% FA
ACN-AF 0.1 M, 0.1% FA22.5:77.5Phenomenex Luna C18 (2) (250 × 3 mm, 3 μm)0.552183727517: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[200,201,202]
95HPLC-UVformate bufferACN20:80CTInstruments C18, (150 × 4.6 mm, 5 μm)1.213.270302204: CBDA, CBD, Δ9-THC, THCA[203]
96HPLC-DAD0.1% TFAACN25:75Cortecs C18 (150 × 4.6 mm, 2.7 μm)115NRNR2288: CBC, CBD, CBDA, CBDV, CBDVA, CBL, Δ9-THC, THCA[204]
97UHPLC-DAD0.1% TFAACN41:59Cortecs Shield RP18 (150 × 4.6 mm, 2.7 μm)25010352288: CBC, CBD, CBDA, CBG, CBGA, CBN, Δ9-THC, THCA[205,206,207]
98HPLC-DAD0.1% TFAACN41:59Cortecs Shield RP18 (150 × 4.6 mm, 2.7 μm)15020352289: CBCA, CBD, CBDA, CBG, CBGA, CBN, Δ8-THC, Δ9-THC THCA[208]
99HPLC-DAD0.1% TFAACN41:59Reliant C18 (250 × 4.6 mm, 5 μm)120203523510: CBC, CBD, CBDA, CBDV, CBG, CBN, Δ8-THC, Δ9-THC, THCA THCV[209]
100HPLC-VWD MeOH13:87MultoHigh 100 RP18 (125 × 4.6 mm, 3 μm)0.24010352123: CBD, CBDV, CBN[210]
101HPLC-UV MeOH17:83ACE 3 C18-PFP (150 × 3.0 mm, 3 μm) with guard col0.4205252223: CBD, CBN, Δ9-THC g[211,212]
102HPLC-DAD0.1% TFAMeOH15:85Raptor ARC-18 (150 × 2.1 mm, 2.7 μm) with guard col0.310330230, 269 f18: CBC, CBCA, CBD, CBDA, CBDV, CBDVA, CBG, CBGA, CBL, CBLA, CBN, CBNA, CBT, Δ8-THC, Δ9-THC, THCA, THCV, THCVA[213]
103HPLC-DADAcOH 0.25%ACN:MeOH (15:1)20:80Phenomenex Luna C18(2) (150 × 4.6 mm, 5.0 μm)1301030 or 35 e2287: CBD, CBDA, CBN, Δ8-THC, Δ9-THC, THCA, THCV[214,215]
104HPLC-DAD1% ACN in hexaneNANucleosil Ag(I) phase (100 × 4.6 mm, 3 μm)1405NR2155: CBD, Δ(4)8-iso-THC Δ8-iso-THC, Δ8-THC, Δ9-THC[216]
AC: acidic cannabinoids; AcOH: acetic acid; ACN: acetonitrile; AF: ammonium formate; Col: column; DAD: diode array detector; FA: formic acid; HPLC: high-performance liquid chromatography; MeOH: methanol; NA: non-applicable; NC: neutral cannabinoids; NR: not reported;TFA: trifluoroacetic acid; UHPLC: ultra high-performance liquid chromatography; RT: room temperature; UV: ultraviolet; VWD: variable wavelength detector. Cannabinoids Abbreviations: CBD: cannabidiol; CBDA: cannabidiolic acid; CBDV: cannabidivarin; CBC: cannabichromene; CBCA: cannabichromenic acid; CBCV: cannabichromevarin; CBG: cannabigerol; CBGA: cannabigerolic acid; CBL: cannabicyclol; CBLA: cannabicyclolic acid; CBN: cannabinol; CBT: cannabicitran; Δ(4)8-iso-THC: Δ4(8)-iso-tetrahydrocannabinol; Δ8-iso-THC: Δ8-iso-tetrahydrocannabinol; Δ8-THC: Δ8-tetrahydrocannabinol; Δ9-THC: Δ9-tetrahydrocannabinol; THCA: Δ9-tetrahydrocannabinolic acid; THCV: Δ9-tetrahydrocannabivarin; THCVA: Δ9-tetrahydrocannabivarinic acid. a Type of liquid chromatography apparatus and UV detector used; when not specified, the UV term is used. b Entry 66–69, 100, and 101: Only water was used as aqueous phase (A). c Entry 81: 0–10 min: F = 1 mg/mL, 10.1–30 min: F = 1.2 mg/mL. d When not clearly stated in the paper, run time is indicated as presented in the published chromatograms. e Entry 103: 30 °C [214], 35 °C [215]. f When reported, the wavelengths described correspond to those selected for quantification. Entry 70: THCVA was determined at 269 nm and oven temperature of 45 °C. Entry 70, 84, 86: DAD wavelength (nm) used for specific cannabinoids alongside 230: 223 (CBT), 251 (CBCA), 261 (CBNA), 269 (the other AC), 285 (CBN, CBC). Entry 102: THCA: 269 nm. g Entry 100: Additional cannabinoids were assigned by MS.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Silva, E.M.P.; Vitiello, A.; Miro, A.; Ribeiro, C.J.A. Recent HPLC-UV Approaches for Cannabinoid Analysis: From Extraction to Method Validation and Quantification Compliance. Pharmaceuticals 2025, 18, 786. https://doi.org/10.3390/ph18060786

AMA Style

Silva EMP, Vitiello A, Miro A, Ribeiro CJA. Recent HPLC-UV Approaches for Cannabinoid Analysis: From Extraction to Method Validation and Quantification Compliance. Pharmaceuticals. 2025; 18(6):786. https://doi.org/10.3390/ph18060786

Chicago/Turabian Style

Silva, Eduarda M. P., Antonella Vitiello, Agnese Miro, and Carlos J. A. Ribeiro. 2025. "Recent HPLC-UV Approaches for Cannabinoid Analysis: From Extraction to Method Validation and Quantification Compliance" Pharmaceuticals 18, no. 6: 786. https://doi.org/10.3390/ph18060786

APA Style

Silva, E. M. P., Vitiello, A., Miro, A., & Ribeiro, C. J. A. (2025). Recent HPLC-UV Approaches for Cannabinoid Analysis: From Extraction to Method Validation and Quantification Compliance. Pharmaceuticals, 18(6), 786. https://doi.org/10.3390/ph18060786

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

Article Metrics

Back to TopTop