You are on page 1of 16

Talanta 140 (2015) 150–165

Contents lists available at ScienceDirect

Talanta
journal homepage: www.elsevier.com/locate/talanta

1
H NMR and HPLC/DAD for Cannabis sativa L. chemotype distinction,
extract profiling and specification
Wieland Peschel n,1, Matteo Politi
Centre for Pharmacognosy and Phytotherapy, Department for Pharmaceutical and Biological Chemistry, The School of Pharmacy, University College London,
29-39 Brunswick Square, London WC1N 1AX, United Kingdom

art ic l e i nf o a b s t r a c t

Article history: The medicinal use of different chemovars and extracts of Cannabis sativa L. requires standardization
Received 20 September 2014 beyond Δ9-tetrahydrocannabinol (THC) with complementing methods. We investigated the suitability of
Received in revised form 1
H NMR key signals for distinction of four chemotypes measured in deuterated dimethylsulfoxide
12 February 2015
together with two new validated HPLC/DAD methods used for identification and extract profiling based
Accepted 23 February 2015
on the main pattern of cannabinoids and other phenolics alongside the assayed content of THC, can-
Available online 5 March 2015
nabidiol (CBD), cannabigerol (CBG) their acidic counterparts (THCA, CBDA, CBGA), cannabinol (CBN) and
Keywords: cannflavin A and B. Effects on cell viability (MTT assay, HeLa) were tested. The dominant cannabinoid
Cannabis sativa L. pairs allowed chemotype recognition via assignment of selective proton signals and via HPLC even in
Extracts
cannabinoid-low extracts from the THC, CBD and CBG type. Substantial concentrations of cannabinoid
THC
acids in non-heated extracts suggest their consideration for total values in chemotype distinction and
CBD
CBG specifications of herbal drugs and extracts. Cannflavin A/B are extracted and detected together with
HPLC cannabinoids but always subordinated, while other phenolics can be accumulated via fractionation and
1
H NMR detected in a wide fingerprint but may equally serve as qualitative marker only. Cell viability reduction in
Analytical markers HeLa was more determined by the total cannabinoid content than by the specific cannabinoid profile.
Therefore the analysis and labeling of total cannabinoids together with the content of THC and 2–4 lead
cannabinoids are considered essential. The suitability of analytical methods and the range of compound
groups summarized in group and ratio markers are discussed regarding plant classification and phar-
maceutical specification.
& 2015 Elsevier B.V. All rights reserved.

1. Introduction highest affinity to cannabinoid receptors (CB1, CB2), has been


manifold tested pharmacologically and clinically [3,4]. Meanwhile
Alongside the development of synthetic cannabinergics, the other non-psychotropic, non-CB binding cannabinoids, mainly
authorized and the off-label medicinal use of cannabis regain cannabidiol (CBD) [5,6] but also cannabigerol (CBG) [7], are
popularity [1]. The chemical and pharmacological complexity of increasingly investigated showing partly distinct effects. Moreover
cannabis makes the pharmaceutical standardization challenging activities are reported for minor non-cannabinoid co-constituents
and requires complementing identity, purity and assay methods to such as the prenylated flavone cannflavin A [8] (CFL-A), common
characterize the starting material (plant/chemotype), the herbal flavonoids [9] or terpenes [10,11]. Despite or because of the
drug (Cannabis flos) and the preparation (extract). complexity some authors advocate the advantage of the natural
Approximately 70 phytocannabinoids—besides 419 other com- mixtures with combinations of cannabis constituents [12,13] pri-
pounds—are described for Cannabis sativa L.; classified chemically marily determined by the chemovar. Conventional plant classifi-
into 10 major groups, the Δ9-trans-tetrahydrocannabinol (THC), cations as drug-, intermediate or fiber type (hemp) are based on
cannabidiol (CBD), cannabigerol (CBG), and cannabinol (CBN)-type the THC and CBD content [14–16] while the nowadays available
being the most abundant [2]. The psychotropic THC, with the spectrum includes varieties with other lead compounds such as
CBG. Within those plants and derived materials the total and
n
relative amount of main constituents can vary considerably.
Corresponding author.
Besides plant distinction cannabis analysis served historically for-
E-mail address: wieland.peschel@ema.europa.eu (W. Peschel).
1
Present address: European Medicines Agency, 30 Churchill Place, Canary ensic/legal purposes to determine THC in biological fluids and con-
Wharf, London E14 5EU, United Kingdom. fiscated material. Originally the plant synthesizes and accumulates

http://dx.doi.org/10.1016/j.talanta.2015.02.040
0039-9140/& 2015 Elsevier B.V. All rights reserved.
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 151

carboxylated forms (e.g. Δ9-trans-tetrahydrocannabinolic acid-THCA) CBG; III: 0.01% THC, 0.68% CBD, 0.02% CBG; IV: 0.3% THC, 5.8% CBD,
which are converted into post-harvest ‘neutral’ derivatives accelerated 25.2% CBG). The II (III, IV) drugs obtained from non-standardized
by light and heat (e.g. THC) [17]. Others—such as CBN—are only growing and drying conditions contained 38% (42%, 42%) flowers,
degradation products of those derivatives. The common focus on 44% (51%, 28%) leaves, 13% (4%, 23%) stalks larger than 2 mm dia-
‘neutral’ cannabinoids can be explained by their activity, bioavailability meter and 5% (3%, 7%) seeds (all w/w), respectively. Stalks 43 mm
(traditional hot smoke inhalation), but also the heat conversion of the diameter were removed before extraction. The age of the materials
acids when traditionally analyzed by GC. However, in case of ‘cold’ at time of extraction was 18 months (I, II and III) and 3 months
preparation, analysis and application, herbal starting materials and (IV).
derived extracts contain the original carboxylated cannabinoids. Their
separate HPLC determination is reportedly more precise than ‘total 2.3. Extraction and fractionation
THC’ values via GC or derivatisation before chromatography [18].
Limitations of all chromatographic methods encouraged also testing The four drugs (I, II, III, IV) were extracted with ethyl acetate
other analytical methods including NMR spectroscopy [19–22]. and ethanol 40% using the classic drug–solvent ratio 1:10 for
Characterization beyond the THC content became more rele- tinctures. 10 g samples were macerated with 100 mL solvent in
vant with the increasing acceptance of medicinal use [23,24]. two passages (24 h each) at room temperature in the dark under
Specifications based on analytical markers vary now according to agitation (aluminum foil covered Erlenmeyer flask on an auto-
purpose i.e. not only to discriminate drug and ‘non-drug’ but mated shaker). After filtration, organic solvents were removed
guarantee identity and consistent quality of specific preparations. under vacuum ( o40 °C) followed by freeze drying in case of the
Even more important is in view of the multiple effects from sev- ethanolic 40% extracts (Et40, EtOAc). A test passage with drugs I
eral co-constituents the determination of prevailing active con- and III analyzed separately showed that 83–94% of cannabinoids
stituent groups that may contribute to the activity. had been extracted. Another 10 g of each drug was defatted with
We therefore used a new targeted 1H NMR profiling method heptane in order to remove a major proportion of cannabinoids.
and two newly developed and validated HPLC/DAD methods as The defatted residues were dried at room temperature and
complementary tools to distinguish chemotypes and identify exhaustively extracted in four passages using methanol (2  6 h)
extracts of different polarity. HPLC/DAD was further used to profile and methanol 70% (2  6 h) and ultrasonic treatment of 1 h. After
extracts as regards main cannabinoid pattern aside more polar reduction under vacuum or freeze drying extracts were dried
constituents such as flavonoids based on the quantification of under nitrogen until a constant weight had been reached (Me70).
main cannabinoids (THC, CBD, CBG and CBN), the corresponding Et40 and Me70 extracts were further fractionated in a liquid/
acids, and the cannabis-characteristic prenylated flavones CFL-A liquid system between water and organic solvents in several steps,
and cannflavin B (CFL-B). Group and ratio markers were derived first with dichloromethane, and secondly with ethyl acetate (Et40-
that are potentially useful in cannabis specifications and their diclo, Et40-etoac, Et40-wat, Me70-diclo, Me70-etoac, Me70-wat).
variation determined according to starting material and extrac- EtOAc extracts were fractionated into a hexane (EtOAc-hex), and
tion. As a simple activity test in relation to these markers we an aqueous (8% methanol) fraction (EtOAc-wat). All unified frac-
checked exemplarily their effect to reduce cell viability in HeLa tions were reduced and dried as described above. All extracts were
cells. stored at  20 °C in the dark.

1
2.4. H NMR procedures
2. Materials and methods
Stock solution with 100 mg/mL of dry extracts was prepared
2.1. Reference standards with deuterated dimethylsulfoxide (99.8% DMSO-d6, Sigma UK). In
some cases a minor amount of D2O (maximum 10%, Goss Scientific,
THC, CBD, CBG, CBN and THCA were purchased from THC UK) was added. Then 150 μL of this stock solution was diluted
Pharm GmbH (Frankfurt, Germany) and stored in the dark at with 1350 μL DMSO-d6 and filtered through a 0.45 mm Acrodiscs
20 °C. CFL-A/CFL-B were kindly provided by Giovanni Appen- syringe filter (Fisher Scientific, Loughborough, UK) and stored at
dino, (Novarra, Italy). As phenolic standards we used the canna- 4 °C. For analysis the 10 mg/mL solutions were thawed and 0.6 mL
binoid precursor olivetol, as common phenolcarbonic acid filled into WG-5 mm NMR tubes (Wilmad). 1H NMR spectra of
chlorogenic acid, and as flavonoids the aglycons quercetin and samples were obtained on Bruker AVANCE 400 MHz instruments
apigenin (all Sigma UK) and the glycosides orientin, homorientin, equipped with a multinuclear probehead with z-gradient. The
vitexin, isovitexin (all Extrasynthese S.A. Co., Genay-Sedex, Xwin-NMR 3.5 software was used for spectra acquisition and
France). processing. The size of all 1D spectra was 65 K and number of
transients varied for different type of spectra. The standard 1D 1H
2.2. Plant material NMR spectra are acquired with 30o pulse length, relaxation delay
of 2 s. The numbers of scans were 128 or 64. The spectra were
Four C. sativa L. dry herbal drugs varied in composition (leaf- recorded at 300 K.
flower ratio), genotype (THC-type, CBD-type, CBG-type, fiber
1
(CBD)-type) and production parameters such as cultivation, drying 2.5. H NMR experiments and analysis
conditions and age. THC-type drug I, a standardized indoor culti-
var from controlled cultivation for medicinal use was provided by We measured pure reference standards (at minimum three
TNO (Zeist, Netherlands) with specification certificate (18% THC concentrations usually 100, 10 and 1 mg/mL adapted to the natural
after heating, 0.8% CBD, o 1% CBN). The pure Cannabis flos drug occurrence in cannabis extracts) and combinations of reference
contained female flower tops without leaves and stalks from lower substances in different ratios. Spectra were recorded between
plant parts. Samples from a CBD-rich (labeled II), a low-cannabi- 0 and 14 ppm chemical shift (δ). After baseline correction, the
noid (III) and a CBG-rich (IV) variety were kindly provided by calibration was carried out on the residual DMSO-d6 solvent peak
Giampaolo Grassi (ISCI, Experimental Institute for Industrial Crops, in terms of the chemical shift (δ ¼ 2.5 ppm) and intensity (set as
Rovigo, Italy). They originate from three outdoor lines with former normalized basis for integration). The fingerprint 0 - 11 ppm and
batches specification (GC analysis: II: 0.7% THC, 13.7% CBD, 1.0% enlarged and expanded sections e.g. between 4 and 9 ppm were
152 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

selected as suitable areas for extract identification. Signal alloca- 2.6.3. Identification and qualification
tion (DMSO-d6) was performed by comparison with reference THC, THCA, CBD, CBG, CBN, CFL-A/B (Fig. 1), flavonoids and phe-
standards, extracts with known HPLC profile and previously pub- nolcarbonic acids were identified using reference standards (retention
lished assignments in deuterochloroform or deuteromethanol times in Table 1). The characteristic DAD-UV spectra (210–400 nm) of
(deuteroacetone for cannflavins) [19–21,25]. the standards and literature reports [24] allowed a classification of
THC: (1H NMR, 400 MHz in DMSO-d6,, all in ppm; in bold dis- compounds into neutral cannabinoids (THC/CBD/CBG pattern and
tinguishable diverse signals of THCA in extracts) δ: 0.85 (H-5”, 3H, CBN/CBC pattern), cannabinoid acids (THCA/CBDA/CBGA pattern or
t), 0.98 (H-9, 3H, s; THCA δ1.03), 1.25 (H-3”, H-4”, m), 1.30 (H-5, m,
THCA δ1.38), 1.31 (H-10, 3H, s), 1.48 (H-2”, 2H, m), 1.60-1 (H-6, H-7,
3H, s), 1.85 (H-5, 1H, m), 2.08 (H-4, 2H, m), 2.34 (H-1”, 2H, m, THCA 7
CH3
δ2.74 þ2.85), 3.08 (H-1, 1H, dm; THCA δ3.15), 6.01 (H-3’, 1H, m), 3
6.15 (H-5’, 1H, m), 6.37 (H-2, 1H, m; THCA δ6.31), 9.20 (2’-OH 1H, s) 4 2 (10)
CBD: (1H NMR, 400 MHz in DMSO-d6,, all in ppm; in bold OH
1 2’ (1)
distinguishable diverse signals of CBDA in extracts) δ: 0.86 (H- 5
6 1’ R
5”3H, t), 1.25 (H-3”, H-4”4H, m), 1.48 (H-2”, 2H, q), 1.60-61 (H-7, 3’ (2)
H3C
H-9, 3H, s), 1.67 (H-5, m; CBDA: δ1.72), 1.93 (H-4, 1H, m), 2.09 (H-4, 10 4’ 2’’ 4’’
8 6’
1H, m), 2.30 (H-1”, 2H, t, CBDA δ2.75), 3.04 (H-6, 1H, t), 3.82 (H-1, H3C O 5’(4) 1’’ 3’’
CH3
9 5’’
1H, d), 4.42 (H-10, 1H, s), 4.49 (H-10, 1H, s), 5.08 (H-2, 1H, s), 6.01
(H-3’, H-5’, 2H, s; CBDA H-5’ δ6.13), 8.62 (2’-OH) THC: R = H
CBG: (1H NMR, 400 MHz in DMSO-d6, all in ppm; in bold dis- THCA: R = COOH
tinguishable diverse signals of CBGA in extracts) δ: 0.85 (H-5”, 3H,
7
t), 1.25 (H-3” and H-4”, 4H, m), 1.48 (H-2”, 2H, m), 1.52 (H-10, 3H, CH3
s), 1.59 (H-9, 3H, s), 1.69 (H-7, 3H, s), 1.89 (H-5, 2H, m, CBGA δ1.99), 3

1.99 (H-4, 2H, m, CBGA: δ2.17), 2.32 (H-1”, 2H, t; CBGA: δ2.78), 4 2
OH
5.04 (H-6, 1H, m), 5.15 (H-2, 1H, m), 6.08 (H-3’ þH-5’, 2H, s, CBGA 5 1 2’
1’ R
H-5’: δ6.22), 8.86 (2’-OH, 2H, s) 6
3’
CFL-A: (1H NMR, 400 MHz in DMSO-d6, all in ppm) δ: 1.51 (H- H2C 2’’
4’ 4’’
8”, 3H, s), 1.58 (H-10”, 3H, s), 1.73 (H-9”, 3H, s), 1.92 (H-4”, 2H, t), 10
H3C HO 6’ CH3
2.0 (H-5” (2H, t), 3.23 (H-1” (2H, m), 3.89 (O-Me, 3H, s), 5.03 (H-6”, 9 5’ 1’’ 3’’ 5’’
t), 5.19 (H-2”, 1H, t), 6.55 (H-10, 1H, s), 6.89 (H-3, 1H, s), 6.94 (H-5’, CBD: R = H
1H, d), 7.55 (H-2’ and H-6’, 2H, m), 13.21 (5-OH, 1H, s) CBDA: R = COOH

2.6. HPLC 7
CH3 OH
1
2.6.1. Fingerprint 1’
2’
R
3
A HPLC Waters™ system 900, with a Waters™ 996 PDA detector 4
2 3’
and a Waters™ 717plus auto sampler device and EmPower soft- 4’ 2’’ 4’’
5
ware equipped with an Aces 5 Phenyl (25 cm x 4.6 mm) column 6
HO 6’ CH3
5’
(ACT, Aberdeen, UK) and a Nova-Paks C8 Guard Column 1’’ 3’’ 5’’

3.9  20 mm, 2/pkg (Waters UK, Elstree, UK) was used. Conditions CBG: R = H
H3C CH3 CBGA: R = COOH
were as follows: column temperature 25 °C, auto sampler 8 °C, 10 9
flow 0.9 mL/min, running time 80 min including a polar pre-phase
and a lipophilic washing post-phase (solvent C 63-71 min 100%);
solvent A water (TFA 0.1%), solvent B water-acetonitrile (65:35, TFA
0.1%) solvent C acetonitrile; gradient: solvent A 0 min 70%, 10 min
CH3
60%, 38 min 40%, 40 min 5%, 55 min 0%, 74 min 70%. For identifi- O
cation a triple fingerprint was recorded near absorbance maxima 3’
OH
of flavonoids (254 nm) cannabinoids (275 nm), and flavonoids/ 2’ 4’
8
1’ 5’
phenolcarbonic acids/cannabinoid acids (324). Detection at HO 9 O
7 2
214 nm provided the best levelled information for all compounds 6’
6 3
and was used for quantification. Samples (starting concentration 1’’
4
10
extracts 10 mg/mL, standards 1 mg/mL) were diluted in methanol, 5
OH
methanol/water mixtures and, prior to use, filtered through a H3C
2’’
4’’
0.45 mm Acrodiscs syringe filter (Fisher Scientific, Loughborough, 3’’

UK) before injection (10 μL or 30 μL). 6’’ 5’’

H3C
2.6.2. Cannabinoid profile 9’’ 7’’
The same equipment and methodology as described for the 8’’
CH3
fingerprint were used for cannabinoid analysis with modifications
10’ ’
in column (Agilent Zorbax RX-C18 column, 5 mm 4.6  250 mm
Fig. 1. (A) THC and THCA, CBD and CBDA, CBG and CBGA. The numbering follows
Highchrom, UK), solvents (only solvent B and C), running time and
adjusted p-cymene based monoterpene nomenclature of CBD after Choi et al. [19]
gradient (55 min, solvent B: 0 min 70%, 30 min 35%, 43 min 5%, for direct comparison in this study. For THC the other common nomenclature
48 min 70%). (dibenzopyran system) is indicated at key positions (in brackets). (B) cannflavin A.
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 153

Table 1
Retention times for standard references in two applied HPLC methods and their classification.

Reference standard tR FPa (min) tR CPb (min) UV λ max (nm) Classification for inclusion in group markers

chlorogenic acid 9.27 – 240–325 TPC


homorientin 21.78 – 256–348 TPC
orientin 22.12 – 256–348 TPC
isovitexin 25.62 – 268–337 TPC
vitexin 26.05 – 268–337 TPC
quercetin 31.42 – 255–372 TPC
apigenin 37.65 – 267–338 TPC
olivetol 44.19 5.98 274 –
cannflavin B (CFL-B) 54.54 11.49 273–242 CFL
cannflavin A (CFL-A) 59.18 20.17 274–342 CFL
cannabidiolic acid (CBDA) 59.54 20.36 269–307 CANA, CBD(A), CANtot
cannabigerolic acid (CBGA) 59.87 20.93 267–309 CANA, CBG(A), CANtot
cannabigerol (CBG) 60.19 21.57 272 CAN, CBG(A), CANtot
cannabidiol (CBD) 60.39 22.21 273 CAN, CBD(A), CANtot
cannabinol (CBN) 61.52 28.78 283 CAN, THC(A), THCtot, CANtot
Δ9-tetrahydrocannabinol (THC) 62.37 32.95 276 CAN, THC(A), THCtot, CANtot
tetrahydrocannabinolic acid A (THCA) 62.67 41.26 270–304 CANA, THC(A), THCtot, CANtot

a
FP: fingerprint (80 min).
b
CP: cannabinoid profile (55 min).

Table 2
Group markers and ratio markers for cannabis extract characterization. Description, relevance and determination from HPLC cannabinoid profile (CP) or fingerprint (FP).

Calculation Calculated as Description/Relevance

THCtot Σ THC, THCA, CBN THC, THCA, CBN (CP/FP) Total THC (‘potency marker’), dominant in classical plants and preparations,
constituent with highest CB1/CB2 receptor activity plus its instable parent
and main degradation product, psychotropic legal/forensic importance
THC(A) Σ THC, THCA THC, THCA (CP/FP) Sum of THC and THCA
CBD(A) Σ CBD, CBDA CBD (CP) Sum of CBD and CBDA, dominant in some chemotypes (intermediate, hemp),
main constituents without CB1/CB2 receptor affinity, non-psychotropic,
specific pharmacological effects
CBG(A) Σ CBG, CBGA CBG (CP) Sum of CBG and CBGA, dominant in some chemotypes, main constituents
without CB1/CB2 receptor affinity, non-psychotropic, specific pharmacologi-
cal effects
CAN Σ neutral cannabinoids THC, CBD, CBG, CBN (CP/FP) neutral (decarboxylated) cannabinoids - dominant in heated, aged and
excessively dried drugs
CANA Σ acidic cannabinoids THC, CBD, CBG, CBN (CP/FP) acidic (carboxylated) cannabinoids -dominant in fresh plants and
preparations without heating
CANtot Σ neutral and acidic cannabi- THC, CBD, CBG, CBN (CP/FP) total cannabinoid content
noids, CAN þ CANA
oCAN CANtot – (Σ THCtot þCBD(A) þ THC (in THC-type) CBD (in CBD- other cannabinoids than those usually found as main cannabinoids in
CBG(A)) type) CBG (in CBG-type) (CP) common chemotypes, (i.e. THCtot, CBD(A), CBG(A))
CFL CFL-A þCFL-B CFL-A, CFL-B(CP) cannflavins-cannabis specific prenylated flavones
TPC Σ flavonoids and phenolcarbonic vitexin, chlorogenic acid (FP) total phenolic content (without CFL)
acids
THCtot/ (CBD(A) þ Σ THC, THCA, CBN / Σ CBD(A), as for THC(A), CBG(A), CBD(A) ‘chemotype marker’, ratio main constituents in common chemotypes, ratio
CBG(A)) CBG(A) (CP/FP) main CB1/CB2 active vs. inactive constituents (plus acidic pro-drug), ratio
main psychotropic/ non-psychotropic constituents (plus acidic pro-drug),
legal/forensic importance
CANA / CAN Σ CANA/ Σ CAN as for CANA, CAN (CP/FP) ‘decarboxilation marker’, ratio acidic /neutral cannabinoids, indicator for
drug and extract quality and age
CANtot / TPC CANtot / TPC as for CANtot, TPC (FP) ‘polarity marker’, indicator for extract/solvent polarity, indicator for leaf and
flower portions, potential influence of phenolics on activity

cannabinolic acid (CBNA)/cannabichromenic acid (CBCA) pattern), resulting in 15–35 major peaks per extract. Peaks with a minimum
flavonoids and phenolcarbonic acids (Fig. A1). CBDA and CBGA were of 0.2% of the total peak area were integrated corresponding
identified as dominant peaks in extracts from CBD-type and CBG-type approximately to a 3:1 signal to noise ratio for the limit of
drugs and their conversion to their neutral forms. Unidentified peaks detection and 10:1 for the limit of quantification. Identified and
were qualified by the PDA spectrum (Fig. A1) as flavonoid, phe- qualified peaks were summarized to group markers ‘total THC’
nolcarbonic acid, cannabinoid, and cannabinoid acid (Tables 1 and 2). (THCtot ¼THC þTHCA þCBN), CBG(A) ( ¼CBG þCBGA) and CBD(A)
(¼CBD þCBDA), neutral cannabinoids (CAN), cannabinoid acids
2.6.4. Assay and marker calculation (CANA), total cannabinoids (CANtot), and total phenolics (TPC).
Standard curves reference substances as external standards From these group markers three ratio markers described as ‘che-
were established and standard mixtures injected with each ana- motype marker’ (THC(A)/(CBG(A) þCBD(A)), ‘decarboxilation
lytical run. For extract profiling all main peaks (above 0.05% of the marker’ (CANA/CAN) and a ‘polarity marker’ (CANtot/TPC) were
total peak area) were qualified according to their spectrum usually calculated (Table 2).
154 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

CFL-B

CBN
CFL-A
orientin
chlorogenic acid

THC
CBG
olivetol
10.00 20.00 30.00 40.00 50.00 60.00 70.00
Minutes

THC

THCA
CBGA
^^
CFL-B

CFL-A
vitexin

10.00 20.00 30.00 40.00 50.00 60.00 70.00

CBDA
CBD
*

THCA
**

THC
*
*
*

*
*

10.00
10.0 20.00 30.00 40.00 50.00 60.00 70.00
chlorogenic acid RS

+ CFL-A RS
**
* + vitexin RS
* + orientin RS

+ CFL-B RS

CBDA
CBD
^^

^^
^^
^
**

^
**
*

10.00 20.00 30.00 40.00 50.00 60.00 70.00


Minutes

Fig. 2. HPLC fingerprint. (A) selected standard references (λ: 254 nm), (B) hydroethanolic crude extract from THC-type, and (C) CBD-type drugs, (D) extract fraction from
fiber-type drug spiked with reference substances (RS) (all λ: 214 nm). Non-identified qualified peaks marked as: n flavonoid, nn phenolcarbonic acid, ^ cannabinoid,
^^ cannabinoid acid.

2.7. HPLC fingerprint – method validation 2.7.2. Linearity and range


In addition to common standard curves for single reference
The fingerprint covers cannabinoids and more polar con- standards (not shown), we tested linearity and range for the more
stituents (phenolcarbonic acids, flavonoid glycosides) separating complex group markers. Comparable results were obtained for
THC, THCA and CBN from other cannabinoids and closed flavo- different extract types in the range between 1 mg/mL and
noids previously described from each other such as vitexin/iso- 25 mg/mL extract (example Fig. 3A). The relative standard devia-
vitexin and orientin/homorientin. tion of the mean (RSD) was maximum 3–8% for group markers and
6–15% for ratio markers.

2.7.1. Selectivity and specificity 2.7.3. Accuracy


THC/THCA, CBD/CBDA or CBG/CBGA were always the promi- The accuracy for group markers was checked via the recovery
nent peaks within the cannabinoids (50 and 65 min) range largely rate of added reference standards (theoretical amount versus
separated from common phenolic co-constituents (8–40 min) found amount). We tested combinations of pure standards, com-
(Table 1, Fig. 2). Spiking of extracts confirmed peak allocation binations of one extract with different standards, and combina-
(example Fig. 2D). Despite overlapping of minor peaks qualifica- tions of three benchmark extracts (‘low flavonoid/cannabinoid’,
tion via UV spectra allowed the summation of AUC for calculation ‘high-cannabinoid’ and ‘high-phenolic’) with the same standard
of CANA, CAN, THCtot, CANtot and TPC. mixture (Table A1). The recovery rate for THCtot, CBD(A) þCBG(A),
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 155

3500 6000 400


THC(A) CBx(A) CAN THCA THC CBN CBDA CBD CBGA CBG

CANA CANtot TPC


3000
5000
y = 217.17x + 76.659
y = 112.92x + 57.828 300
2500 R = 0.9994
R = 0.9988 y = 13.969x + 1.2058
4000 R = 0.9998
y = 8.3066x + 1.2675
R = 0.9999
mg

mg

mg
2000
3000 y = 191.45x - 38.998 200
y = 36.99x + 23.56
R = 0.9969
R = 0.9998
1500
y = 7.2496x + 6.4821
y = 51.924x + 17.725 R = 0.9949
R = 0.9984 y = 31.663x - 5.9281 2000
1000 R = 0.9963 y = 1.3577x - 1.2049
100 R = 0.9693
y = 21.111x + 17.156
R = 0.983 1000 y = 0.8425x + 0.7867
500 R = 0.9921
y = 2.0963x + 0.9869
R = 0.9962
0 0 0
0 10 20 30 0 10 20 30 0 10 20 30

sample conc. (mg/mL) sample conc. (mg/mL) sample conc. (mg/mL)

Fig. 3. Examples for HPLC linearity tests. (A) Group markers calculated from the fingerprint of a CBD-type extract fraction, (B and C) cannabinoids calculated from can-
nabinoid profile of a THC-type extract. Samples were injected at 5 different concentrations between 1 and 25 mg/mL.

Fig. 4. HPLC cannabinoid profile (λ: 214 nm). (A) selected references standards; and extracts from (B) THC-type, (C) CBD-type, (D) CBG-type, (E) fiber-type extract alone and
(F) spiked with reference standards THC, CBD and CBG.

CANtot and TPC was between 89.9% and 106.9% (mixed standards), 2.8.1. Selectivity and specificity
79.8% and 126.4% (one extract plus different standards), and 92.8% THC, CBD, CBG (as well as THCA, CBDA, CBGA) share compar-
and 123.4% (different extracts plus one standard mix). Results able UV spectra with minor differences in the maximum absorp-
were considered acceptable for fingerprint sum parameters. tion (265–285 nm). Distinct UV spectra are obtained with CBN and
CBNA (equal to CBC and CBCA, Fig. A1) [24]. In extracts from THC-,
2.8. HPLC cannabinoid profile – method validation CBD- and CBG-type starting materials, the three main neutral
constituents and acidic forms were separated (Fig. 4B-E). Spiking
For cannabinoid and cannflavin assay, a 55 min HPLC analysis of different extracts did confirm peak allocations (example Fig. 4F).
was applied (Fig. 4). Both the THC and THCA peak covered in some extracts a minor
156 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

Fig. 5. 1H NMR spectra of cannabis constituents (DMSO-d6). THC and mixtures of THC þCBD (2:3), THCþ CBD (6:1) and CBDþ CFL-A (10:1) with proton signal assignments
for THC, CBD and CFL-A (in brackets).

peak with a similar spectrum-visible via a shoulder at low THC alongside detection of minor peaks was found with 10 mg/mL
(A) concentrations. sample concentrations.

2.8.2. Linearity and range


After establishing standard curves for pure substances, the 2.8.3. Accuracy
linearity was determined in different extracts. Fig. 3B and C shows We tested the recovery of single compounds when added
exemplarily the calculation of main cannabinoids in a THC-type combined to different types of extracts. THC-type extracts were
extract at five concentrations. The results were consistent at con- spiked with THC, CBD or CFL-A and three CBD-type extracts with
centrations 5–25 mg/mL with a relative confidence interval for 3 reference standards. Recovery rates were between 93.6–104.8%
single constituents between 1.4% and 5.7%. As only peaks with a (standard mixtures) and 70.8–129.5% (in extracts) for cannabi-
minimum signal to noise ratio of 1:10 (about 0.2% of the total peak noids and 91.6–118.6% (standard mixtures) and 77.3–93.2% (in
area) were integrated, the limit of quantification was approxi- extracts) for cannflavins, respectively. Percentage deviations
mately 0.5 mg in 1 g extract corresponding to about 0.25% of the higher than 15% resulted only from very low concentrated sub-
amount of the lead cannabinoid in cannabis preparations domi- stances. CFL-A determination was partially hampered when com-
nated by one compound pair. Satisfactory peak separation bined with a CBDA-rich extract and recovery rates of 60.5–88.2%
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 157

suggest that the method is not suitable when cannflavins are the volatile solvent for direct comparison. The same samples can also
main focus of analysis for CBD-type extracts. be used for further dilution in other solvents for chromatographic
analysis or in cell assays. Of advantage is also the use of the solvent
2.8.4. Precision signal for calibration and normalization (integration) without need
In addition to the instrument precision for single compounds for internal standards. Most cannabinoid proton signals in DMSO-
(e.g. RSD THCA 2.11%, THC 4.72%, CBN 1.75%, CBG 6.24%; n ¼6) we d6 were found comparable to those assignments previously made
tested the intra-assay precision when analyzing extracts over for deuterochloroform (with a general downward shift) and also
several hours. To detect whether mixtures decompose or instru- previous assignments with the protic solvent deuteromethanol
ment parameters shift, five times the same extract was injected at [19] (Figs. 1 and 5). While the discrimination of THC, CBD, CBG and
the beginning, the middle and the end of a 36 h analytical run. We their carboxylic counterparts in the ‘aliphatic area’ (0–4 ppm), is
obtained a satisfying precision with RSD below 4%. No trends were often hampered by overlaps of close signals, the ‘aromatic area’ in
observed. The inter-assay precision tested in different extract particular the H-5′ position and the two olefinic methins (H-2 and
types (measurement in duplicate at day 1, day 7 and day 12) was H-6) in CBD and CBG between 4 and 6.5 ppm are more suitable.
o5% RSD for compounds 4 20 mg/g extract concentration, o10% The hydroxyl groups provide additionally distinguishable signals
for compounds between 5 and 20 mg/g in the extract and o20% for main cannabinoids such as in 2′ position for THC at 9.2 ppm,
for compounds between LoD and 5 mg/g in the extract. A trend for CBD at 8.62 ppm, for CBG at 8.86 ppm, although somewhat
was observed regarding the conversion of acids into neutral forms unreliable due to temperature dependence as previously reported
within 12 days. [19,21]. We compared mixtures of pure compounds to estimate
the range of suitable ratios for simultaneous identification in
2.9. Cell culture and cell viability assay extracts. Fig. 5 illustrates the distinction of THC and CBD in two
combinations (0–11 ppm) as well as appearance of flavonoid
HeLa-IL-6 (HeLaluc) cells originating from Dr. M. L. Lienhard proton signals when CBD is combined with CFL-A. Fig. 6 shows
Schmitz (University of Giessen, Germany) were maintained in extracts from the four chemovars (4–9 ppm) demonstrating that in
DMEM (Invitrogen, UK) supplemented with 10% fetal bovine non-heated cannabis extracts usually two main substances have to
serum and antibiotics at 37 °C in a 5% CO2 humidified atmosphere be considered. In some cases, extracts with a balanced ratio
and split when confluent. Cells were allowed to grow in media to between two pairs, even four signal sets have to be taken into
60–80% confluence before harvesting. For the MTT assay cell sus- account. Nonetheless the per se selectivity of NMR by suppressing
pension was adjusted to 7.5  104 cells/mL and 96-well plates less intense signals allows in principle an easy identification of the
(200 μL per well) were incubated for 18–24 h at 37 °C with the 5% chemotype. The difference between benchmark extracts (canna-
CO2, 95% humidity. binoid-rich versus practically cannabinoid-devoid but containing
The MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium other phenolics) as well as the use of selected proton signals to
bromide] assay was performed as previously described [26]. Samples allow the distinction between neutral and acidic forms is shown in
were primarily dissolved in DMSO or methanol and diluted in media Fig. 7. Addressing both supports identification, indicates the level
for a starting concentration of 200 μg/mL (Et40 extracts) or 50 μg/mL of decarboxilation but also illustrates possible differences in the
(EtOAc extracts). Absorbance values were measured with an Anthos NMR fingerprint despite equal content in the prevailing pair such
Lucy 1 luminometer at 570 nm (reference filter at 620 nm, ASYS, as CBG(A). With our conditions, 1:10 ratios were found feasible to
Eugendorf, Austria). Absorbance values were converted into % growth recognize patterns of the lower concentrated compound in pure
values in comparison to the non-treated control. Toxic effects were compound mixtures. For extracts, only a maximum 1:5 ratio to the
expressed as maximum non-toxic concentration (MNTC¼85% of highest concentrated compound may still allow to address com-
control) and as IC50 value. fortably the subordinated compound. Below that, it depends case
by case on the actual concentrations and numbers of main co-
2.10. Statistical analysis constituents (e.g. THC plus THCA or THC plus THCA plus CBD plus
CBDA etc.).
After HPLC validation (see above) samples were injected in
duplicate and results expressed as mean 7SD. MTT assay: Samples 3.2. 1H NMR (DMSO-d6) key signals for chemotype distinction and
were tested in duplicate per plate and the median values from herbal drug identification
three independent experiments were used to calculate the mean
(n ¼3) 7SEM. Correlation between single group and ratio markers Spectra were manually checked for specific signals (qualitative
of cannabis extracts and cell viability reduction in HeLa cells (IC50 marker), for overlapping and selectivity (identification without
and MNTC) were tested using Pearson's correlation test. interference, potential for integration) and for intensity (detect-
able also at lower concentrations). We summarize in the following
signals for THC(A), CBD(A), CBG(A) differentiation and demarca-
3. Results and discussion tion between the acidic and neutral forms.

1
3.1. H NMR identification of cannabis extracts in DMSO-d6 3.2.1. THC(A)
44 ppm: For THC(A) the 6.15 ppm signal of the 5′ proton is the
After qualitative peak assignments of major cannabinoids and best recognizable one to distinguish from CBD(A)/CBG(A) with
cannflavins using 1H, 13C, 1H-1H COSY and HMBC [19], 1H NMR CBDA (H-5′) at 6.13 usually well separated. The THC 6.01 signal of
was favored to differentiate between chemovarieties followed by H-3′ (shared with CBD) is the most selective peak versus THCA,
proposals to quantify constituents in extracts [20,21] and to use although too weak in low concentrated extracts. For both demar-
metabolomics for chemovar distinction [30]. A direct analysis of cations, the H-2 signal (THC 6.37, THCA 6.31 ppm) represents the
tinctures with suppression of water signals using standard deut- marker of first choice.
erated solvents for extraction was previously reported by our o4 ppm: The THC-characteristic H-9 signal at 0.98 (THCA:
group [22]. Here, in contrast to commonly used chloroform, 1.03) is in mixtures within a large bulk of adjacent peaks. Although
methanol or water, the ‘DMSO-d6 fingerprint’ allows for extracts of both are not always completely separated, they may serve for
distinctive polarities a single sample preparation in only one non- demarcation to CBD(A)/CBG(A). Following signals are not shared
158 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

Fig. 6. 1H NMR spectra of extracts from four chemotypes (DMSO-d6, 10 mg/mL, 4–9 ppm, amplified 4  ). Extracts (Et40-EtOAc) from THC-type (A), CBD-type (B), Fiber-type
(C) and CBG-type (D) drugs with key proton signals for identification of main cannabinoids.

but can partially overlap with close signals from other cannabi- be found at 6.13 closed to THC(A) (6.15). The 2′-OH signal at 8.62 may
noids, i.e. useful for identification but less so for quantification: help distinguishing from THC and CBG. Only in extracts containing
1.31 (H-5; THCA 1.38), 2.34 (H-1″, CBD 2.31, CBG 2.32) and 3.08 (H- CBDA appeared another OH-group signal at 5.32 obviously due to
1, CBD 3.05). THCA-specific split H-1″ signals at 2.74 and 2.85 (THC hydrogen bonding of the carboxyl group. A sharp and intense signal
single at 2.34) are well detectable in THC-type extracts. However, is exhibited by the H-2 proton at 5.08, not interfering with
when less concentrated in extracts of other chemotypes, they are THC(A) but eventually with the 5.04 peak of CBG(A).
not sharp and too close to e.g. CBDA H-1″(2.75) for reliable iden- o4 ppm: The CBD-specific not very sharp 3.82 peak (H-1) is
tification. Also differences in the H-1 (THC 3.08, THCA 3.13-3.18) often too weak within mixtures. CBD characteristic signals at 2.30
may be disturbed by signals from other cannabinoids. (H-1″, CBDA 2.75) and 3.05 (H-6) can overlap with corresponding
THC signals (2.34, 3.01). The CBDA-characteristic H-1″ signal at
3.2.2. CBD(A): 2.75 distinguishes from CBD (2.3) but partially overlaps with THCA
44 ppm: Most characteristic are the split CBD H-10 proton sig- H-1″(2.75 þ2.85) in extracts with substantial THCA content or
nals appearing at 4.42 and 4.49 ppm. Both peaks are shared with with the corresponding CBGA signal at 2.78.
CBDA (4.41 and 4.47), but there are no THC(A) or CBG(A) signals
interfering. The prominent 6.01 signal of CBD is not only caused by H- 3.2.3. CBG(A):
3′ as in THC but also by H-5′, complicating its use for eventual 44 ppm: The most distinguishable to THC(A) and CBD(A) are
quantification of neutral cannabinoids. H-5′ of CBDA in contrast can the H-5′ signals at 6.08 ppm (CBG) and 6.22 (CBGA), that were also
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 159

Fig. 7. 1H NMR spectra of selected extracts in DMSO-d6. (all 10 mg/mL). (A) cannabinoid-rich extract (THC(A) 364.2 mg/g, CBG(A) 20.2 mg/g, HPLC; CANA/CAN ratio: 0.83)
with key signals to distinguish THC and THCA, (B) cannabinoid-reduced extract: THC(A) o 1 mg/g, CBD(A) o 1 mg/g, CBG(A) o 1 mg/g, (C1) extract from CBG-type drug
(CBG(A) 193.3 mg/g, CANA/CAN ratio 0.82), with key signals to distinguish CBG and CBGA (C2) extract from the same CBG-type drug with comparable CBG(A) content
(189.0 mg/g) but different CANA/CAN ratio (0.28).

Table 3
Key proton signals (δ in ppm) in DMSO-d6 for identification of characteristic compounds/groups in THC(A), CBD(A) or CBG(A) dominant cannabis chemotypes.

Specific signals for identificationa Most selective in extractsb

THC 0.98 (H-9, s), 2.34 (H-1″, m), 3.08 (H-1, dm), 6.37 (H-2, m), 9.21 (2′-OH, s) 6.37, 9.21
THCA 1.03 (H-9, s), 1.38 (H-5, m), 2.74/2.85 (H-1″, m), 6.31 (H-2, dm) 6.31, 2.74/2.85
THC(A) 1.85 (H-5, m), 6.15 (H-5′, m) 6.15, 0.98/1.03
(shared signals THC and THCA not shared with CBD(A) or CBG(A))

CBD 1.93/2.09 (H-4, m), 2.28 (H-1″, t), 3.05 (H-6, t), 3.82 (H-1, d), 5.08 (H-2, s), 8.62 (2′-OH, s) 5.08, 8.62
CBDA 2.18/2.27 (H-4, m), 2.75 (H-1″, m), 3.91 (H-1, d)
CBD(A) 4.41-4.42/4.47-9 (H-10) (shared signals CBD and CBDA not shared with THC(A) or CBG(A)) 4.41–4.49

CBG 1.89 (H-5, m), 2.32 (H-1″, t), 6.08 (H-5′, s), 8.82 (2′-OH, s) 6.08, 8.82
CBGA 2.78 (H-1″, t), 6.22 (H-5′, s) 6.22
CBG(A) 1.69 (H-7, s), 5.04/ 5.15 (H-6, m and H-2, m), (shared signals CBG and CBGA not shared with THC(A) or CBG(A)) 5.04/ 5.15

CAN 2.28-2.34 (H-1″) 2.28–2.34


(shared signals THC, CBG, CBD not shared with THCA, CBGA, CBDA)
CANA 2.75-2.78 (H-1″) 2.75–2.78
(shared signals THCA, CBGA, CBDA not shared with THC, CBG, CBD)
CFL-A 1.73 (H-9″, s), 6.55 (H-10, s), 6.89 (H-3, s), 6.95 (H-5″, d), 7.55 (H-2′, H-6′, m), 13.21 (5-OH, s) 6.55, 6.89, 7.55

a
Specific signals of the respective constituent not shared with other main cannabinoids in THC-type, CBD, type, fiber type, and CBG-type derived cannabis extracts.
b
Consideration of intensity and overlaps with other main compounds in THC-type, CBD, type, fiber type, and CBG-type derived cannabis extracts.

recognizable as minor peaks in THC-type extracts with a CBG mixtures. Equal to CBDA, a signal assigned to 6′-OH at 5.31 appears
(A) content around 1:10 vis-à-vis THC(A). CBG H-5′ overlaps with only in CBGA containing mixtures.
the H-3′ signal. Equally to THC and CBD, the 2′-OH signal is specific o4 ppm: Some signals of CBG(A) dominated extracts in the
and usually well recognizable at 8.82. The two CBG(A) olefinic aliphatic area are slightly distinct from THC(A) and CBD(A) but
methin protons H-6 and H-2 at 5.04 and 5.15 are characteristic as hampered by overlaps or low intensity such as those of H-4 / H-5
well as selective and sufficiently intense in most cases yet prone to or H-9/H-10. The H-9 signal at 1.59 is useful in CBG(A) dominant
overlaps with the CBD(A) 5.08 signal (H-2) in case of balanced materials but may overlap with the 1.61 signal of other
160 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

Table 4
Recognizable 1H NMR pattern of main cannabis constituents in cannabis extracts.Selective proton signals for discrimination of chemotypes for reference standards and THC-,
CBD,- and CBG-type extracts (all 10 mg/mL in DMSO-d6), □ ¼ strong signal, □ ¼ weak signal, □¼ no detected signal.

cannabinoids if CBGA is a minor constituent. The CBG H-1″ triplet was not identified. Signals most influenced by the carboxyl group
at 2.32 is only slightly different to CBD (2.29) but useful to dis- are mainly the aromatic H-5′, and to certain extent the hydroxyl-
tinguish from CBGA (2.78) that may be also identified by some and the methylen groups (H-1, H-5).
minor differences in methylene groups H-4 and H-5. An overview of suitable signals to identify THC-, CBD-, and
CBG-type extracts is compiled in Table 3. Because NMR identifi-
3.2.4. CFL-A: cation of multicompound mixtures is challenging due to over-
44 ppm: CFL-A exhibits some signals typical for the flavonoid lapping peaks of similar structures [27] it is usually combined with
structure and the prenyl moiety which are distinctive from main multivariate pattern recognition and principal component analysis
phytocannabinoids. The aromatic protons at 7.55, 6.94, 6.89, and (PCA) for metabolomic analysis but also industrial quality control
6.55 may not be selective in mixtures with other flavonoids but [28,29]. The targeted analysis of key signals as performed here, can
quite specific for more lipophilic cannabis extracts where non- amend PCA approaches in quality control because it provides in
prenylated flavonoids are unlikely to be found. The rather broad contrast to the mathematical data output of PCA a set of specific
prenyl peaks of the H-6″ and H-2″ position at 5.03 and 5.19 are signal allocation to ‘tick boxes’ for few compounds in particular
interfering with CBG(A) peaks. combined with HPLC information on major cannabinoids present.
o4 ppm: The most specific prenyl peak is the singlet at 1.73 Using the most selective signal in the aromatic and aliphatic area
(H-9″). The methoxy peak at 3.89 may be shared by other flavo- for each of the key compounds, a pattern of a few key signals can
noids but is distinguishable from main cannabinoids. Overall, be compiled for fast identification as demonstrated in Table 4 for
despite the possibility for identification, cannflavin detection in extracts of different polarity. It allows simplification avoiding the
cannabis extracts by 1H NMR appears limited due to low con- need to analyze the complete proton signal set of complex mix-
centration and signal intensity in relation to predominant canna- tures. An extension beyond THC, CBD and CBG type distinction
binoids. The same applies to lower concentrated common flavo- may be tested in with other chemovarieties.
noids. Exemplarily we had tested quercetin addition to cannabis
extracts (up to 20%), where typical signals such as at δ 1.90, 3.80, 3.3. HPLC/DAD fingerprint and cannabinoid profile
4.04 where not intense enough while minor peaks can be identi-
fied in the less crowded area 6.5–8.0 ppm (data not shown). Cannabis HPLC fingerprinting and quantitative analysis has
developed over years to improve the separation of very similar
3.2.5. Shared signals of THC(A), CBD(A), CBG(A): cannabinoid structures via specific columns, detection (e.g. flame/
Usually a standard cannabinoid pattern of the pentyl group electro ionization) or hyphenation with MS [23,31–33]. Compar-
signals is recognizable with approximate shifts of 0.85 (H-5″), 1.25 able to a more recent validated method, we show that conven-
(H-3″/4″), 1.48 (H-2″), whereof the signal at 0.85 interferes least tional HPLC/DAD provides valuable information for routine quality
with others. Also H-1″ is largely shared between THC, CBD and control despite limits for the simultaneous cannabinoid analysis
CBG around 2.28–2.34, but the influence of the neighboring car- [34]. The fingerprint signals the main cannabinoids and presence
boxyl group makes it an important marker to distinguish from the of more polar co-constituents and allows summarizing into groups
genuine acids (around 2.75). Another mutual signal of all main of qualified peaks for main extract pattern (Fig. 2). The advantage
acids (THCA, CBDA, CBGA) distinct to their neutral counterparts of a broad range is made on expense of baseline separation e.g.
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 161

Fig. 8. Comparison of 40% ethanol (A  D) and ethyl acetate (E  H) extracts from four drugs. (I THC type, II CBD type, III CBD fiber type, IV CBG-type) (Aþ E) Main compound
groups THCtot, CBx(A) ( ¼ CBG(A)þ CBD(A)), oCAN and TPC, (Bþ F) Total cannabinoid content and ratio markers, (C þG) cannabinoid profile, (D þH) Effect on cell viability in
HeLa (MTT assay, mean 7 SEM, n¼ 3).

CFL-A/B or CBD/CBG versus CBDA/CBGA. The cannabinoid profile the cannabinoid content (e.g. low in III) but also other drug char-
method allows in shorter time quantification of the main neutral acteristics, e.g. the leave portion higher in II and III than in I and IV
and acidic cannabinoids to THC, CBD and CBG type drugs plus CFL, leading to higher TPC values. (3) Fig. 8C, the cannabinoid profile,
the only non-cannabinoids that appeared within the cannabinoid focuses on the ratio of main cannabinoids to each other inde-
range (Fig. 4). Limits are baseline separation of minor cannabinoids pendent of total values. It illustrates in a different way the pre-
and in some cases overlapping peaks for excessive concentrations vailing cannabinoid pair, the ratio between the main acidic and
of one compound in the range 20–22 min (e.g. CBDA or CBGA) neutral cannabinoids and additionally CBN (confirming the
which can be solved by sample dilution. Extension to other rele- advanced age of I) and cannflavins (measurable amounts only in I:
vant cannabinoids of less common chemotypes such as Δ8 THC, 0.79 mg/g). Notably, I contains more CBG(A) than CBD(A) which
CBC/CBCA or tetrahydrocannabivarin (THCV) may be tested in the confirms other reports for THC-type drugs and raising doubts on
future. the suitability of the conventional focus on THC and CBD only [34].

3.4. HPLC based key markers for chemotype and herbal drug 3.5. Chemotype distinction
identification
HPLC analysis of the four varieties confirmed THC-type (I),
Main pattern for chemotype comparison and herbal drug CBD-type (II), CBD-type cannabinoid-low (III) and CBG-type (IV,
characteristics of non-heated material are presented as (1) abso- first described by Fournier et al. in 1987) [35] several CBG strains
lute values of main compound groups, (2) ratios of key compound are available [34]). In contrast, traditional terms may be used with
groups, and (3) relative proportion of cannabinoids (Fig. 8): (1) In caution when characterizing dry herbal drugs. Small and Beck-
line with traditionally important identification of ‘drug-type’ the stead had first differentiated between a chemotype with
absolute content in THC (plus THCA and CBN) is shown in relation THC4 0.3%/CBD o0.5%, an intermediate type with dominant CBD
the content of common main cannabinoids CBD(A) and CBG(A), but THC also present, and a particularly THC-low type [14]. The
accompanying minor cannabinoids and the total phenolic content still common classification drug type (THC 42%, CBD 0%), fiber
(Fig. 8A). (2) Fig. 8B provides at one glance the information ‘can- type (THC o 0.3%, CBD 4 0.5%, THC/CBD ratioo0.1) and an inter-
nabinoid-rich’ versus ‘cannabinoid-low’ plant (CANtot) alongside mediate type (THC 4 0.5%, CBD 40.5%, THC/CBD ratio 4 0.5)
indicators whether THC(A) prevail over non-psychotropic canna- reflects the importance distinguishing between Cannabis flos for
binoids, how fresh the material is (degree of decarboxilation) and recreational use (often425% THC) and low-THC industrial hemp
to which extent cannabinoids prevail over accompanying phe- without considering other possible inherited biosynthesis pattern
nolics. The THCtot / CBD(A)þ CBG(A) ratio indicates for I THC-type in plant populations [16,36–38]. CBD forming an essential part of
independent from absolute values. Even if very low amounts those classifications was supposed to be partially overestimated
would be found, e.g. in old material with low THC but high CBN due to non-selective methods, which became more relevant with
values, any upwards directed bar signals THC-type origin. The medicinal applications using plants optimized on CBD [3,13,39].
CANA/CAN ratio flagged progressed decarboxylation in all four For I–IV neither provider specification nor our results (EtOAc
drugs; most advanced in II. The CANtot /TPC ratio is influenced by extracts from specific batches of dry drugs of certain age) allows
162 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

Table 5
Classification of four cannabis chemotypes and proposed specification of herbal drugs and extracts.

I II III IV

Provider specification of Cannabis flosa:


THC (%) 18 (after heating) 0.7 0.01 0.3
CBD (%) 0.8 13.7 0.68 5.8
CBG (%) n.s. 1.0 0.02 25.2
THC/ CBD ratio 22.5 0.05 0.015 0.01

Conventional classification of plants (based on exhaustive analysis of Cannabis flos)b:


‘drug type’? ‘intermediate type’? ‘fiber type’ ‘intermediate type’?
THC (%) 42 40.5 o0.3 40.5
CBD (%) 0 40.5 40.5 40.5
CBG (%) n.s. n.s. n.s. n.s.
THC/ CBD ratio n.s. 40.5 o0.1 40.5

Proposed specification of Cannabis flosc:


‘THC-type’ ‘CBD-type’ ‘CBD-type’, low cannabinoid ‘CBG-type’
CANtot (%) 18.94 10.64 1.31 19.34
THCtot (%) 14.68 0.33 0.06 0.72
CBD(A) (%) o0.04 9.72 0.61 2.92
CBG(A) (%) 1.97 0.40 0.02 16.66
d
THCtot /CBx(A) 7.5 0.03 0.04 0.04
Solvent/Anal. Meth: (EtOAc/HPLC) (EtOAc/HPLC) (EtOAc/HPLC) (EtOAc/HPLC)
or
CANtot (mg/g) 448.8 273.5 120.6 344.2
THC(A)/CBx(A)d 7.5 0.03 0.04 0.04
CANA/CAN 0.47 0.23 0.63 0.64
CANtot/TPC 192.1 6.6 4.3 29.0
Solvent/Anal. Meth (EtOAc/HPLC) (EtOAc/HPLC) (EtOAc/HPLC) (EtOAc/HPLC)

Proposed specification of cannabis extracts:


Drug/extract ratio 2.4:1 2.6:1 9.2:1 1.7:1
Solvent EtOAc EtOAc EtOAc EtOAc
CANtot (mg/g) 448.8 273.5 120.6 344.2
THCtot (mg/g) 347.8 8.5 5.1 11.9
CBD(A) (mg/g) o1 249.7 56.3 48.2
CBG(A) (mg/g) 46.6 10.3 1.8 274.9
[TPC (mg/g)]e [2.3] [16.8] [13.4] [12.0]

a
As provided by the supplier at time of delivery I (HPLC), II-IV (GC).
b
Ref. [14].
c
Based on EtOAc extraction of specific batches available in this study.
d
CBx(A)¼ CBD(A) þ CBG(A).
e
Optional such as in case of hydroethanolic extracts from Cannabis folium.

unambiguous allocation (Table 5): the declared CBD-value of I extraction of not always well defined plant samples and total
would not match conventional ‘drug-type’ definition, II does nei- conversion of the acids such as via GC. Yet materials/extracts may
ther fit into ‘intermediate type’ nor ‘fiber type’ definition, and for differ and nowadays methods with separate acid detection prevail.
extracts derived from III the CBD-values were too low to comply
with fiber type criteria, which, however, CBG-type IV would 3.6. HPLC-based extract profiling – range and enrichment of com-
match. Nor does the three-type system consider varieties with pound groups
prevailing cannabichromene (CBC), enhanced Δ8-THC instead of
Δ9-THC, or the propyl homologs instead of the usual pentyl can- The profile of different extracts was investigated for the pos-
nabinoids (e.g. tetrahydrocannabivarin / tetrahydrocannabivarenic sibility to identify the original herbal drug and the extent com-
acid dominant clones often of South African origin) [40–42]. THC pound groups are extracted, which determines their relevance as
or CBD values (HPLC) alone without consideration of the acids potential markers for specification. Standard solvents to exhaus-
would not only be too low but also unreliable due to decarbox- tively extract cannabinoids are traditionally chloroform or
ylation according to the age and storage. As actual THC/ CBD ratio chloroform/methanol mixtures. We used EtOAc to macerate the
to indicate intermediate or fiber type we obtained 4529.6, 0.03, main part of the cannabinoids without enrichment of more lipo-
0.11 and 0.25, for I–IV respectively. The more real ratio philic substances (e.g. terpenoids, fatty acids). EtOAc extracts were
THCtot/CBx(A), independent from the actual main co-cannabinoids, compared with Et40 extracts (moderate cannabinoid with more
the decarboxilation progress, and the THC to CBN conversion, was: polar co-constituents), Me70 extracts of defatted material (‘low
7.5, 0.03, 0.04 and 0.04 for I–IV, respectively. CBx(A) summarizes cannabinoid’) and extract fractions (Fig. 8, Table 6).
the main non-CB1/CB2-active constituents: here CBD(A) and CBG Et40 extracts: Et40 extracts yielded only 6.2% (I), 16.6% (II), 15.3%
(A), in other cases eventually CBC plus CBCA or may even include (III) and 20.4% (IV) of CANtot extracted with EtOAc (Fig. 8E). TPC
all other cannabinoids (oCAN) to obtain information on the por- values varied between 4.3 and 33.2 mg/g in line with the portion
tion of psychotropic THCtot. In addition, such data are only com- of leaves in the drugs. Contrary, the relative ‘chemotype marker’
parable together with specification of the plant part (such as flos, (Fig. 8F 1st bar) indicates the chemotype solvent-independently.
folium or resinum), limits for admixtures (such as seeds, stalks, For I, a reduced extraction of THC(A) in relation to CBG(A) when
foreign matter), and the analytical method. It is assumed that using polar solvents instead of EtOAc is indicated by an only two-
traditional THC and CBD values are based on assumed exhaustive fold THCtot value compared to ’CBx(A)’. The CANA/CAN ratio
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 163

Table 6 Enrichment of cannabinoids: Fractionation allowed further


Group markers and ratio markers for six extract fractions from four cannabis concentration of total cannabinoids and affected slightly group
varieties. Marker maxima as obtained per chemotype are indicated in bold, minima
ratios such as THCtot/ CBG(A). Highest amounts in cannabinoids
underlined.
per extract where obtained in most cases with high lipophilic
EtOAc Ethanol 40% Methanol 70% fractions (dichloromethane) following polar first extraction (Et40).
Hexane Watera CH2Cl2 EtOAc CH2Cl2 EtOAc Lipophilic extraction throughout such as hexane fractions of EtOAc
extracts gave overall higher yields but did not substantially
(I)THC-type
THC(A) (mg/g) 372.5 14.0 245.5 87.4 33.3 1.2
increase CANtot compared to the crude extract. Although two-step
CBD(A) (mg/g) nd nd 5.3 nd nd nd extraction allows modifying the ratio of particular cannabinoid
CBG(A) (mg/g) 20.8 19.5 28.4 11.1 1.5 0.5 groups, purification of main compound pairs by simple solvent
CFL (mg/g) nd 3.7 nd 7.4 2.8 2.7 based fractionation (e.g. separation of CBG(A) from accompanying
TPC (mg/g) nd 30.0 nd 94.9 nd 27.4
THC(A) or CBD(A)) was not possible.
THCtot/CBxA 17.9 0.72 7.2 7.9 21.9 3.1
CANA/CAN 0.83 0.46 0.89 0.27 0.31 0.36 Enrichment of phenolics: We obtained fractions with a more
CANtot/TPC 4 846 4.8 4 786 1.4 4172 1.6 levelled content of flavonoids and cannabinoids via removal of
(II) CBD-type major parts of the cannabinoids. Et40-etoac/Me70-etoac and
THC(A) 10.7 nd 14.0 1.1 nd nd EtOAc-wat fractions had CANtot/TPC values between 1.4–4.8, 0.2-
CBD(A) 122.7 14.6 137.9 14.6 12.7 nd
CBG(A) nd nd 9.1 1.1 1.6 nd
1.8, 0.4–1.2, and 0.2-20.9 for varieties I, II, III and IV, respectively.
CFL nd nd nd nd 4.5 nd Cannflavins were often below the limit of quantification, did never
TPC nd 17.8 nd 168.3 125.1 290.4 reach more than 8 mg/g extract and remained usually ‘minor
THCtot/CBxA 0.09 o 0.03 0.09 0.07 o 0.04  constituents’ in relation to cannabinoids or other phenolic
CANA/CAN 0.51 0.78 0.75 0.34 1.46 
compounds.
CANtot/TPC 4 164 1.78 4 686 0.25 0.33 0.08
(III) Fiber-type In summary, fractionation of crude cannabis extracts influenced
THC(A) 12.2 nd 14.9 10.2 4.1 nd the CANtot/TPC ratios but less so THCtot/CBx(A) ratios. Cannabi-
CBD(A) 166.3 78.3 183.1 81.7 50.0 nd noids are chemically too similar to enrich separately via solvent
CBG(A) 20.0 nd 16.3 1.4 nd nd polarities without more sophisticated techniques. Thus chemotype
CFL nd nd nd 5.2 nd 5.1
TPC nd 118.9 45.1 86.1 5.6 55.4
identification is possible solvent-independently. On the other hand
THCtot/CBxA 0.07 0.01 0.08 0.12 0.08  it shows that mixing of specific herbal drugs may provide a better
CANA/CAN 2.40 1.78 1.24 2.20 1.79 0.62 path of tailoring preparations to a profile that differs substantially
CANtot/TPC 4 384 1.24 3.32 0.80 8.69 0.45 from the natural cannabinoid ratio in one chemovar.
(IV) CBG-type
THC(A)   13.8 10.1 1.6 nd
CBD(A)   69.1 41.8 21.8 5.2 3.7. Effect on the cell viability in HeLa
CBG(A)   219.0 193.4 163.9 35.1
CFL   nd 4.3 3.8 2.3 IC50 and MNTC levels were between 2.5-8.0 μg/mL and 0.3-
TPC   nd 14.7 6.6 24.3 2.2 μg/mL for EtOAc extracts compared to 6.1–25.2 μg/mL and 2.0–
THCtot/CBxA 0.05 0.04 0.01 o 0.01
CANA/CAN 0.41 1.01 1.57 1.12
12.7 μg/mL for Et40 extracts, respectively (Fig. 8D and H). With an
CANtot/TPC 4 744 20.9 18.9 0.24 assumed co-influence of the quantitative and the qualitative
composition, the cytotoxic effect in HeLa cells was less determined
ndo 0.5 mg/g (LoQ). by the dominant cannabinoids e.g. the THC(A) content but by the
a
Water containing 8% methanol. extraction solvent and the resulting CANtot value. For instance Et40
extracts of II, III, and IV with a 50-fold, 12.5-fold and 33-fold higher
(Fig. 8F 2nd bar) proved to be not only material but also extract- CBD(A) or CBG(A) content compared to THC(A) affected cell via-
dependent. The CANtot /TPC ratio (Fig. 8F 3rd bar) revealed that bility with an IC50 value that was maximum 2.5 times higher (Et40
Et40 extraction lowers the cannabinoid content more (10-fold extract of III) or even similar (Et40 extract of IV) to the THC(A)-rich
lower for the Et40 vs. EtOAc) than it accumulates phenolic co- Et40 extract of I. This was confirmed by testing additionally the
constituents (doubled). The relative cannabinoid profile (Fig. 8 G) fractions from all crude extracts. Over the complete matrix no
confirmed the genotype without substantial differences between clear correlation could be found between single constituents such
Et40 and EtOAc. In I Et40, however, the portion of other com- as THC, group or the defined ratio markers. The most likely rela-
pounds than the dominant cannabinoids were found relatively tionship between cannabis extract composition and resulting
increased compared to EtOAc. toxicity in the HeLa cells was between CANtot and log IC50 values
Exhaustive methanol/methanol 70% extraction from defatted from the MTT assay (Fig. 9).
material: Four-fold TPC values were obtained with exhaustive
Me70 extracts (I 4.3 and 19.4 mg/g, II 33.2 and 150.8 mg/g, III 20.4 3.8. Markers for standardization of herbal drugs and extracts
and 85.2 mg/g, respectively) compared to Et40. Despite removal of
the major part of cannabinoids they remained more concentrated For adequate specification of non-heated drugs (e.g. Cannabis
than phenolics in extracts from cannabinoid-rich material flos, C. resinum, C. folium) and drug preparations (including
(CANtot/TPC ratios: 6.2 (in I), 5.1 (in IV)). The cannabinoid profile extraction solvent and drug extract ratio) for pharmaceutical
still allowed the information about the original chemotype. The purposes [43], the preferred specification of absolute values of
enrichment of cannflavins in relation to cannabinoids could hardly CANtot, THCtot, and CBD(A) and other relevant main cannabinoids
be achieved. can be rationalized as follows (Table 5):
Recognition of the chemotype in extract fractions: Despite vari- CANtot: Due to the increased pharmacological knowledge on
able quantity of cannabinoids and their proportions the non-CB receptor mediated and non-THC caused effects, the overall
THCtot/CBxA ratio allowed for all fractions the identification of the amount of cannabinoids represents useful basic information which
dominant cannabinoid pair, as long as cannabinoids could still be should be further specified with regard to the main constituents.
detected. The THCtot/ CBx(A) ratio varied between 0.7 and 22 for Relevant specific markers may be different to classic main phyto-
THC-type, 0.03-0.09 for CBD-type, 0.01-0.05 for CBG-type, and cannabinoids [44,45]. The content of specific cannabinoids needs
0.07-0.12 for the fiber type derived extracts (Table 6). to be put into context, i.e. the relevance of 20 mg/g THC is different
164 W. Peschel, M. Politi / Talanta 140 (2015) 150–165

Me70 extracts, and 12% in fractions with two exceptions (I Me70-


etoac; II Me70-diclo) in which, however, other phenolics were at
least 10 fold more concentrated. Enrichment without parallel
concentration of cannabinoids (or other phenolics) may only be
achieved with more sophisticated fractionation and chromato-
graphic techniques. Despite some interesting biological activities
when applied alone8), cannflavins may play no direct role in the
activity of conventional extracts and serve as indicator for identity
without need for specification.

4. Conclusions

We newly developed one 1H NMR and two HPLC methods that


combined are useful to distinguish THC, CBD and CBG dominant
cannabis chemotypes and indicate phenolic co-constituents. A set
of potentially relevant markers was defined, their range detected
in a variety of extracts and discussed vis-à-vis plant classification
(e.g. with consequences for cultivation eligibility) and relevance
for pharmaceutical specification. If the focus is on the putative
psychotropic strength of ‘drug-type’ material like in conventional
chemotype distinction, for non-heated samples (CANA detection)
THC may be replaced by THCtot and THC/CBD ratios by
Fig. 9. Correlation between CANtot (mg/g) values in cannabis extract fractions and THCtot/CBx(A) with CBx(A) summarizing main non-CB1/CB2-active
cell viability reduction in HeLa cells (MTT assay, IC50 7 SEM, n¼ 3).
neutral and acidic cannabinoids. For pharmaceutical extract spe-
cification, CANtot and THCtot together with other lead cannabinoids
with overall amounts of 25 mg/g CANtot or 300 mg/g CANtot. Our that may vary according to purpose and preparation should be
cell viability test showed that effects did not correlate with THC declared alongside a defined herbal substance, solvent and ana-
but with CANtot when not only THC dominant samples are used. lytical method. The obtained data set as starting point for specific
THC, THCA, CBN versus THCtot: Fresh material contains mainly drug specification requires confirmation of applicability with an
THCA, if not converted via heating completely into THC. It has been extended sample set and may potentially be extended to other and
previously shown that both substances should be analyzed sepa- less common phytocannabinoids.
rately and summarized [18]. Thus in non-heated herbal drugs and
‘cold’ extracts, THCtot including THCA and CBN instead of THC
alone determines the putative ‘strength’ and pharmacological
Abbreviations
effects. CBN has a low to moderate CB1 affinity and psychotropic
effect [6]. The separated specification of CBN, THC and THCA may
For abbreviations of cannabis constituents and summarized
be useful in stability tests. Here, CBN was only detectable at
group markers see Tables 1 and 2.
advanced age in extracts from the THC-type.
CBD, CBDA, CBG, CBGA: In analogy to THCA/THC, the ‘pro-drug’
CBDA should be detected and added to CBD values in non-heated
Acknowledgments
preparations from fresh material. The explicit determination and
labeling of CBG(A) is suggested because of increasingly more CBG-
We thank Jose Maria Prieto for the scientific and technical
type plants on the market (identification), their importance as
support as well as Keith Helliwell (Ransom), Michael Heinrich and
main co-constituents in THC-type extracts, and own pharmacolo-
Andrew Constanti for the possibility to do this work and to use the
gical effects [7].
facilities at the School of Pharmacy London, the organizational
CANA/CAN: Values between 0.27 and 2.4 in our extracts
framework of the European research project COOP-CT-2004-
demonstrate a considerable amount of acids in all extracts when
512696 and partial funding by Ransom (Hitchin, UK). We thank
(relatively) fresh – prone to be influenced by age, light and tem-
also for the samples from the cannabis collection in Rovigo (IT) by
perature. Apart from the analytical importance, rather pharma-
Giampaolo Grassi and the provision of isolated cannflavins by
cokinetic than pharmacodynamic differences might be expected
Giovanni Appendino (Novarra, IT).
for the oral use of extracts, although differences between acids and
neutral cannabinoids have been reported from in-vitro tests [46].
This stability indicating parameter may be simplified to the
Appendix A. Supplementary material
dominant cannabinoid pair e.g. CBGA/CBG.
TPC: Phenolic co-constituents including orientin/ vitexin
Supplementary data associated with this article can be found in
reached at highest 16.8 mg/g in crude extracts with CANtot /TPC
the online version at http://dx.doi.org/10.1016/j.talanta.2015.02.
ratios of 4.3 (i.e. maximum 23% of cannabinoids), while in polar
040.
fractions maximum 290 mg/g were obtained mainly via removal of
cannabinoids. Although possible to manufacture extracts with a
significant portion of phenolics, TPC and CANtot /TPC ratio speci-
fication may not be necessary for more common lipophilic References
extracts. A check for phenolics in fingerprints may be useful to
indicate admixtures in Cannabis flos herbal drugs. [1] M. Summers, Independent (2010) 34–35.
[2] M.A. ElSohly, D. Slade, Life Sci. 78 (2005) 539–548.
CFL: CFL values reached at maximum 2.6% of the CANtot values [3] R. Mechoulam, L. Hanus, Chem. Phys. Lipids 108 (2000) 1–13.
in Et40 and EtOAc crude extracts, 13.9% in cannabinoid-reduced [4] F. Grotenthermen, Curr. Drug Targets CNS Neurol. Disord. 4 (2005) 507–530.
W. Peschel, M. Politi / Talanta 140 (2015) 150–165 165

[5] G. Esposito, D. De Filippis, M.C. Maiuri, D. De Stefano, R. Carnuccio, T. Iuvone, [29] U. Holzgrabe, R. Deubner, C. Schollmayer, B. Waibel, J. Pharm. Biomed. Anal. 38
Neurosci. Lett. 399 (2006) 91–95. (2005) 806–812.
[6] R.G. Pertwee, Br. J. Pharmacol. 15 (2008) 199–215. [30] J.T. Fischedick, A. Hazekamp, A. Erkelens, J.H. Choi, R. Verpoorte, Phytochem-
[7] F. Borrelli, I. Fasolino, B. Romano, R. Capasso, F. Maiello, D. Coppola, et al., istry 71 (2010) 2058–2073.
Biochem. Pharmacol. 85 (2013) 1306–1316. [31] A.K. Kovar, H. Lindner, Archiv der Pharmazie 324 (1991) 329–333.
[8] M.L. Barrett, D. Gordon, F.J. Evans, Biochem. Pharmacol. 34 (1985) 2019–2024. [32] V. Ferioli, C. Rustichelli, G. Pavesi, G. Gamberini, Chromatographia 52 (2000)
[9] G. Vanhoenacker, R.P. Van, K.D. De, P. Sandra, Nat. Prod. Lett. 16 (2002) 57–63. 39–44.
[10] K.W. Hillig, Biochem. Syst. Ecol. 32 (2004) 875–891. [33] O. Zoller, P. Rhyn, B. Zimmerli, J. Chromatogr. A 872 (2000) 101–110.
[11] J. Gertsch, M. Leonti, S. Raduner, I. Racz, J.Z. Chen, X.Q. Xie, K.H. Altmann, [34] B. De Backer, B. Debrus, P. Lebrun, L. Theunis, N. Dubois, L. Decock, et al., Anal.
K.M. Karsak, A. Zimmer, Proc. Natl. Acad. Sci. U.S.A 105 (2008) 9099–9104. Technol. Biomed. Life Sci. 877 (2009) 4115–4124.
[12] E.M. Williamson, Phytomedicine 8 (2001) 401–409. [35] G. Fournier, C. Richez-Dumanois, J. Duvezin, J.P. Mathieu, M. Paris, Planta med.
[13] E. Russo, G.W. Guy, Med. Hypotheses 66 (2006) 234–246. 53 (1987) 277–280.
[14] E. Small, H.D. Beckstead, Lloydia 36 (1973) 144–165. [36] J.A. Beutler, A.H. DerMarderosian, Econ. Bot. 32 (1978) 387–394.
[15] C. Rustichelli, V. Ferioli, M. Baraldi, P. Zanoli, G. Gamberini, Chromatographia [37] V.G. Virovets, J. Internat., J. Int. Hemp Assoc. 3 (1996) 13–15.
47 (1998) 215–222. [38] E. Small, D. Marcus, Econ. Bot. 57 (2003) 545–558.
[16] E.P.M. De Meijer, M. Bagatta, A. Carboni, P. Crucitti, V.M.C. Moliterni, P. Ranalli, [39] C. Slijkhuis, R. Hoving, L. Blok-Tip, D. de Kaste, [Kwaliteitsnormen Medicinale
G. Mandolino, Genetics 163 (2003) 335–346. Cannabis] Quality Standards for Medicinal Cannabis, 2004.
[17] M. Fellermeier, W. Eisenreich, A. Bacher, M.H. Zenk, Eur. J. Biochem. 268 [40] J.H. Holley, K.W. Hadley, C.E. Turner, J. Pharm. Sci. 64 (1975) 892–894.
(2001) 1596–1604. [41] P.B. Baker, T.A. Gough, S.I. Johncock, B.J. Taylor, L.T. Wyles, Bull. Narc. 34 (1982)
[18] F.E. Dussy, C. Hamberg, M. Luginbuhl, T. Schwerzmann, T.A. Briellmann, For- 101–108.
ensic Sci. Int. 149 (2005) 3–10. [42] J. McPartland, Online Proceedings of the 3rd International Symposium, Bior-
[19] Y.H. Choi, A. Hazekamp, A.M.G. Peltenburg-Looman, M. Frederich, C. Erkelens, esource Hemp, Wolfsburg (Germany), 13–16 September 2000, Available at:
A.W.M. Lefeber, R. Verpoorte, Phytochem. Anal. 15 (2004) 345–354. 〈www.Nova-institut.de〉.
[20] Y.H. Choi, H.K. Kim, A. Hazekamp, C. Erkelens, A.W.M. Lefeber, R. Verpoorte, [43] European Medicines Agency, Guideline on Declaration of Herbal Substances
J. Nat. Prod. 67 (2004) 953–957. and Herbal Preparations in Herbal Medicinal Products/Traditional Herbal
[21] A. Hazekamp, Y.H. Choi, R. Verpoorte, Chem. Pharm. Bull. 52 (2004) 718–721. Medicinal Products (EMA/HMPC/CHMP/CVMP/287539/2005 Rev.1), 11 March
[22] M. Politi, W. Peschel, N. Wilson, M. Zloh, J.M. Prieto, M. Heinrich, Phy- 2010, Available at: 〈http://www.ema.europa.eu〉.
tochemistry 69 (2008) 562–570. [44] M.A. ElSohly, H. deWit, S.R. Wachtel, S. Feng, T.P. Murphy, J. Anal. Toxicol. 25
[23] T. Lehmann, R. Brenneisen, J. Liq. Chromatogr. 18 (1995) 689–700. (2001) 565–571.
[24] A. Hazekamp, A. Peltenburg, R. Verpoorte, C. Giroud, J. Liq. Chromatogr. Relat. [45] A.J. Hill, S.E. Weston, N. Jones, I. Smith, S. Bevan, E.M. Williamson,
Technol. 28 (2005) 2361–2382. G.J. Stephens, C.M. Williams, B.J. Whalley, Epilepsia 51 (2010) 1522–1532.
[25] F. Taura, S. Morimoto, Y. Shoyama, Phytochemistry 39 (1995) 457–458. [46] K.C.M. Verhoeckx, H.A.A.J. Korthout, A.P. van Meeteren-Kreikamp, K.A. Ehlert,
[26] W. Peschel, A. Kump, J.M. Prieto, J. Pharm. Pharmacol. 63 (2011) 1483–1494. M. Wang, J. van der Greef, R.J.T. Rodenburgh, R.F. Witkamp, Int. Immunopharmacol.
[27] G.F. Pauli, B.U. Jaki, D.C. Lankin, J. Nat. Prod. 68 (2005) 133–149. 6 (2006) 656–665.
[28] M. Defernez, I.J. Colquhoun, Phytochemistry 62 (2003) 1009–1017.

You might also like