ArticlePDF Available

Synthesis and Characterization of NESS 0327: A Novel Putative Antagonist of the CB1 Cannabinoid Receptor

Authors:

Abstract and Figures

The compound N-piperidinyl-[8-chloro-1-(2,4-dichlorophenyl)-1,4,5,6-tetrahydrobenzo [6,7]cyclohepta[1,2-c]pyrazole-3-carboxamide] (NESS 0327) was synthesized and evaluated for binding affinity toward cannabinoid CB1 and CB2 receptor. NESS 0327 exhibited a stronger selectivity for CB1 receptor compared with N-piperidinyl-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (SR 141716A), showing a much higher affinity for CB1 receptor (Ki = 350 +/- 5 fM and 1.8 +/- 0.075 nM, respectively) and a higher affinity for the CB2 receptor (Ki = 21 +/- 0.5 nM and 514 +/- 30 nM, respectively). Affinity ratios demonstrated that NESS 0327 was more than 60,000-fold selective for the CB1 receptor, whereas SR 141716A only 285-fold. NESS 0327 alone did not produce concentration-dependent stimulation of guanosine 5'-O-(3-[35S]thio)-triphosphate ([35S]GTPgammaS) binding in rat cerebella membranes. Conversely, NESS 0327 antagonized [R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrolol [1,2,3-de]-1,4-benzoxazin-yl]-(1-naphthalenyl)methanone mesylate] (WIN 55,212-2)-stimulated [35S]GTPgammaS binding. In functional assay, NESS 0327 antagonized the inhibitory effects of WIN 55,212-2 on electrically evoked contractions in mouse isolated vas deferens preparations with pA2 value of 12.46 +/- 0.23. In vivo studies indicated that NESS 0327 antagonized the antinociceptive effect produced by WIN 55,212-2 (2 mg/kg s.c.) in both tail-flick (ID50 = 0.042 +/- 0.01 mg/kg i.p.) and hot-plate test (ID50 = 0.018 +/- 0.006 mg/kg i.p.). These results indicated that NESS 0327 is a novel cannabinoid antagonist with high selectivity for the cannabinoid CB1 receptor.
Content may be subject to copyright.
JPET #49924
1
Journal of Pharmacology and Experimental Therapeutics
Synthesis and characterization of NESS 0327: a novel putative antagonist
of the CB
1
cannabinoid receptor
Stefania Ruiu, Gèrard A. Pinna
2
, Giorgio Marchese, Jean-Mario Mussinu
2
, Pierluigi Saba
3
,
Simone Tambaro, Paola Casti, Romina Vargiu
4
and Luca Pani
5
Neuroscienze S.c.a r.l., 09123 Cagliari, Italy;
Copyright 2003 by the American Society for Pharmacology and Experimental Therapeutics.
JPET Fast Forward. Published on March 27, 2003 as DOI:10.1124/jpet.103.049924
JPET #49924
2
NESS 0327: a novel putative CB1 cannabinoid receptor antagonist.
Correspondence to:
Luca Pani
Institute of Neurogenetic and Neuropharmacology
Via Boccaccio 8, 09047 Selargius (Ca), Italy.
Tel: +39 070 2548079 fax: +39 070 254275
e.mail: L.Pani@inn.cnr.it
Abbreviations: NESS 0327, N-piperidinyl-[8-chloro-1-(2,4-dichlorophenyl)-1,4,5,6-tetrahydrobenzo
[6,7]cyclohepta[1,2-c]pyrazole-3-carboxamide], SR 141716A, N-piperidinyl-5-(4-chlorophenyl)-1-
(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide, [
35
S]GTPγS, guanosine 5’-O-(3-
[
35
S]thio)-triphosphate, WIN 55,212-2, [R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl) methyl]
pyrolol [1,2,3-de]-1,4-benzoxazin-yl]-(1-naphthalenyl) methanone mesylate]
Text pages: 28
Tables:1
Figures: 4
References:36
Words in the Abstract: 214
Words in the Introduction : 369
Words in the Discussion: 749
JPET #49924
3
Abstract
The compound N-piperidinyl-[8-chloro-1-(2,4-dichlorophenyl)-1,4,5,6-tetrahydrobenzo
[6,7]cyclohepta[1,2-c]pyrazole-3-carboxamide] (NESS 0327) was synthesized and evaluated
for binding affinity towards cannabinoid CB
1
and CB
2
receptor. NESS 0327 exhibited a
stronger selectivity for CB
1
receptor when compared with N-piperidinyl-5-(4-chlorophenyl)-
1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (SR 141716A) showing a
much higher affinity for CB
1
receptor (K
i
= 350 ± 5 fM and 1.8 ± 0.075 nM, respectively)
and a higher affinity for the CB
2
receptor (K
i
= 21 ± 0.5 nM and 514 ± 30 nM, respectively).
Affinity ratios demonstrated that NESS 0327 was more than 60,000-fold selective for the
CB
1
receptor, while SR 141716A only 285 fold. NESS 0327 alone did not produce
concentration-dependent stimulation of guanosine 5’-O-(3-[
35
S]thio)-triphosphate
([
35
S]GTPγS) binding in rat cerebella membranes. Conversely, NESS 0327 antagonized
[R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl) methyl] pyrolol [1,2,3-de]-1,4-benzoxazin-
yl]-(1-naphthalenyl) methanone mesylate] (WIN 55,212-2)-stimulated [
35
S]GTPγS binding.
In functional assay, NESS 0327 antagonized the inhibitory effects of WIN 55,212-2 on
electrically evoked contractions in mouse isolated vas deferens preparations with pA
2
values
of 12.46 ± 0.23. In vivo studies indicated that NESS 0327 antagonized the antinociceptive
effect produced by WIN 55,212-2 (2 mg/kg, s.c.) in both tail flick (ID
50
= 0.042 ± 0.01
mg/kg i.p.) and hot plate test (ID
50
= 0.018 ± 0.006 mg/kg i.p.). These results indicated that
NESS 0327 is a novel cannabinoid antagonist with high selectivity for the cannabinoid CB1
receptor.
JPET #49924
4
Interest in the pharmacology of cannabinoids has rapidly increased after the cloning of
cannabinoid receptors and the discovery of their endogenous ligand:
arachidonylethanolamide (anandamide) (Devane et al., 1988, 1992; Munro et al., 1993). Two
types of cannabinoid receptors, CB
1
and CB
2,
have been characterized, both of which have
distinct anatomical distributions and ligand binding profiles. Cannabinoid CB
1
receptors are
present in the central nervous system (CNS) with the highest densities in the hippocampus,
cerebellum and striatum (Herkenham et al., 1990; Howlett, 1998) and, to a lesser extent, in
several peripheral tissues. Cannabinoid CB
2
receptors appear to be predominantly located in
peripheral tissues (Pertwee, 1997, 1999; Galiègue et al., 1995). Both receptors belong to the
G protein-coupled family of receptors that negatively regulate adenylate cyclase and control
the release of arachidonic acid (Howlett, 1995). Naturally occurring [
9
-
tetrahydrocannabinol (
9
-THC) and
8
-THC] and synthetic cannabinoid agonists (HU-210,
CP 55,940 and WIN 55,212-2) produce a number of effects in mice (hypoactivity, catalepsy,
hypothermia and antinociception) that are collectively known as the tetrad of cannabinoid-
induced behaviours (Abood and Martin, 1992; Compton et al., 1992, 1993). These behaviors
are of a central origin and are thought to be mediated via the cannabinoid CB
1
receptor
(Compton et al., 1996; Lichtman and Martin, 1997; Rinaldi Carmona et al., 1994), while the
CB
2
receptor may mediate some of the peripheral effects of
9
-THC, such as
immunosuppression (Martin, 1986).
The cloning of CB
1
and CB
2
receptors and the subsequent development of selective tools has
advanced the concept of therapeutically targeting cannabinoid receptors. Besides their
established clinical antiemetic action (Gralla 1999; Voth and Schwartz, 1997), cannabinoid
receptor agonists also possess appetite stimulant, anticonvulsant, antinociceptive,
hypothermic and antiglaucoma properties (Formukong et al., 1989; Mattes et al., 1994;
Pertwee, 1999; Porcella et al., 2001).
JPET #49924
5
Recently, several groups have become interested in the development of cannabinoid
antagonists, hoping to develop new drugs to cure diseases connected with possible
malfunctions of “cannabinoid/anandamide” system.
We report the synthesis of a putative cannabinoid ligand, code named NESS 0327, its
differential binding to CB
1
and CB
2
cannabinoid receptors, its ability to stimulate [
35
S]GTPγS
binding in rat brain, its effect on mouse vas deferens and its action on an in vivo assay known
to be affected by cannabinoids.
JPET #49924
6
Methods
(
Z
,
E
)-5-(3-Chlorophenyl)-pent-4-enoic acid (3)
: A solution of (3-carboxypropyl)
triphenylphosphonium bromide (2) (14 g, 32.61 mM) in anhydrous dimethylsulfoxide
(DMSO) (40 ml), with 2.6 M of the sodium salt of DMSO in anhydrous DMSO (24 ml,
62.25 mM), below 10°C, was added to a solution of 3-chlorobenzaldehyde (3.06 g, 21.74
mM) in anhydrous tetrahydrofuran (8 ml). The resulting solution was heated at 50°C for 18
h; subsequently, it was allowed to return to room temperature and poured into water. The
mixture was acidified with 6 N hydrochloric acid and extracted with ethyl acetate (3x25 ml).
The combined extracts were washed with brine, water and then dried over anhydrous sodium
sulphate, to provide a browning compound after evaporation. The crude compound was
purified by flash column chromatography on silica gel eluting with dichloromethane
/acetone
9/1 to afford the desired diastereomeric mixture 3 (42% yield); Rf 0.51
(dichloromethane/acetone 9/1); IR (nujol): 3200-2500 (OH), 1720 (C=O), 1590 (Ar);
1
H-
NMR: 2.40-2.75 (m, 8H), 5.60-5.75 (m, 2H), 6.15-6.45 (m, 2H), 7.10-7.28 (m, 6H), 7.32 (s,
2H), 9.50 (br s, 2H, exch. with D
2
O). Anal. C
11
H
11
ClO
2
(C, H, Cl).
5-(3-Chlorophenyl)-pentanoic acid (4): A suspension of the diastereomeric mixture of
pentenoic acid derivate 3 (1 g, 4.75 mM) was subjected to catalytic hydrogenation over PtO
2
(Adams‘ catalyst, 0.1 g, 10% w/w) in ethanol (EtOH) (50 ml) for 2.5 h at room temperature
and 45 psi of hydrogen pressure. The mixture was filtered through a paper filter and the
filtrate concentrated under reduced pressure to yield the desired acid 4 (100% yield) as a
yellow solid, m.p.56-58°C. Rf 0.84 (petroleum ether/ethyl acetate 1/1); IR(nujol): 3300
(OH), 1710 (C=O), 1600 (Ar);
1
H-NMR: 1.60-1.80 (m, 4H), 2.30-2.45 (m, 2H), 2.55-2.71
(m, 2H), 7.04 (d, 1H, J = 6.4 Hz), 7.12-7.35 (m, 3H), 9.65 (br s, 1H, exch. with D
2
O). Anal.
C
11
H
13
ClO
2
(C, H, Cl).
2-Chloro-6,7,8,9-tetrahydro-benzocyclohepten-5-one (5): A suspension of pentanoic acid
4 (0.5 g, 2.36 mM) and thionyl chloride (0.63 ml, 8.5 mM) was heated for 30 min at 50°C.
JPET #49924
7
Thionyl chloride in excess was subsequently removed under reduced pressure and the residue
was added for 3 times to dichloromethane (3 ml) which was evaporated under reduced
pressure. A solution of the crude acyl chloride in dichloromethane (3 ml) was added drop
wise to a magnetically stirred suspension of AlCl
3
(0.32 g, 2.36 mM) in dichloromethane (3
ml). The resulting mixture was stirred at room temperature overnight then poured into ice
and the whole extracted with dichloromethane (3x5 ml). The combined extracts were washed
with (5%) aqueous sodium bicarbonate solution, water and, after drying over anhydrous
sodium sulfate, filtered and evaporated to provide a brownish compound. The crude
compound was purified by flash chromatography on silica gel eluting with petroleum ether/
ethyl acetate 9/1 to afford the attempt compound 5 (77% yield) as a yellow orange oil, b.p.
94-97°C/0.05 mm Hg (lit
1
128-131/0.35 mmHg); Rf 0.65 (petroleum ether/ethyl acetate 9/1);
IR (film): 3350 (OH), 1680 (C=O), 1590 (Ar);
1
H-NMR: 1.72-1.98 (m, 4H), 2.73 (t, 2H, J =
6.2 Hz), 2.91 (t, 2H, J = 6.0 Hz), 7.21 (s, 1H), 7.28 (d, 1H, J = 8.8 Hz), 7.68 (d, 1H, J = 8.6
Hz). Anal. C
11
H
11
ClO (C, H, Cl).
(2-Chloro-5-oxo-6,7,8,9-tetrahydro-5H-benzocyclohepten-6-yl)-oxo-acetic acid ethyl
ester (6): A mixture of EtONa (7.5 mM) in absolute EtOH, (3.5 ml) and diethyl oxalate (0.51
ml, 3.75 mM) was stirred for 30 min at room temperature, and a solution of compound 5
(0.73 g, 3.75 mM) in absolute ethanol (27 ml) was added over 30 min. The resulting mixture
was reacted at room temperature for 9 h then poured onto crushed ice and the whole acidified
with 2 N hydrochloric acid and extracted with chloroform (3x15 ml). The combined extracts
were washed with water, dried over anhydrous sodium sulfate, filtered and evaporated to
afford the β-dichetoester 6 as an orange oil, which was used in the next step without further
purification, (84% yield); b.p. 95-98°C/0.05 mm Hg; Rf 0.78 (petroleum ether/ethyl acetate
9/1); IR (film): 3440 (OH), 1730 (C=O), 1680 (C=O), 1600 (Ar);
1
H-NMR: 1.41 (t, 3H, J = 7
Hz), 2.08 (quint, 2H), 2.32 (t, 2H, J = 7.2Hz), 2.72 (t, 2H, J = 7 Hz), 3.88 (q, 2H, J = 7 Hz),
7.23 (d, 1H, J = 1.8 Hz), 7.34 (dd, 1H), 7.58 (d, 1H, J = 8.2 Hz), 15.37 (br s, 1H, exch. with
D
2
O). Anal. C
15
H
15
ClO
4
(C, H, Cl).
JPET #49924
8
8-Chloro-1-(2,4-dichlorophenyl)-1,4,5,6-tetrahydrobenzo[6,7]cyclohepta[1,2-c]pyrazole-
3-carboxylic acid ethyl ester (8): 2,4-dichlorophenylhydrazine hydrochloride (7) (0.72 g,
3.38 mM) was added to a magnetically stirred solution of ester 6 (0.9 g, 3.05 mM) in EtOH
(21 ml) and the resulting mixture heated under reflux for 3 h; subsequently the solvent was
removed under reduced pressure to yield the crude ester. Purification by flash
chromatography on silica gel eluting with petroleum ether/ethyl acetate 8.5/1.5 gave the
attempt compound 8 as a yellow solid (58% yield); mp 160-161°C (crumbled with petroleum
ether); Rf 0.47 (petroleum ether/ethyl acetate 9/1); IR (nujol): 1725 (C=O), 1605 (Ar);
1
H-
NMR: 1.43 (t, 3H, J = 7 Hz), 2.13-2.40 (m, 2H), 2.67 (t, 2H, J = 6.4 Hz), 3.09-3.40 (m, 2H),
4.46 (q, 2H, J = 7 Hz), 6.60 (d, 1H, J = 8.2 Hz), 7.02 (dd, 1H), 7.31 (d, 1H, J = 2.2 Hz), 7.36-
7.49 (m, 2H), 7.54 (d, 1H, J = 9.2 Hz). Anal. C
21
H
17
Cl
3
N
2
O
2
(C, H, Cl, N).
8-Chloro-1-(2,4-dichlorophenyl)-1,4,5,6-tetrahydrobenzo[6,7]cyclohepta[1,2-c]pyrazole-
3-carboxylic acid (9) :A solution of potassium hydroxide (0.17 g, 2.94 mM) in methanol (5
ml) was added to a magnetically stirred solution of ester 8 (0.64 g, 1.47 mM) in methanol (7
ml), the mixture was refluxed for 9 h and the cooling reaction mixture poured onto crushed
ice and acidified with 1 M hydrochloric acid. The precipitate was filtered, washed with water
and dried under vacuum to yield the corresponding acid as a white solid. (97% yield); mp
270°C (EtOH); Rf 0.51 (chloroform/methanol 9/1); IR (nujol): 3410 (OH), 1690 (C=O);
1
H-
NMR: 2.20-2.39 (m, 2H), 2.50-3.35 (m, 4H), 6.61 (d, 1H, J = 8.2 Hz), 7.03 (dd, 1H), 7.32 (d,
1H, J = 1.8 Hz), 7.39-7.49 (m, 2H), 7.53 (d, 1H, J = 8.2 Hz), 13.25 (br s, 1H, exch. with
D
2
O). Anal. C
19
H
13
Cl
3
N
2
O
2
(C, H, Cl, N).
N-Piperidinyl-[8-chloro-1-(2,4-dichlorophenyl)-1,4,5,6 tetrahydrobenzo [6,7] cyclohepta
[1,2-c] pyrazole-3-carboxamide] (NESS 0327)
:
A solution of the acid 9 (0.50 g, 1.23 mM)
and thionyl chloride (0.24 ml, 3.69 mM) in toluene (10 ml) was refluxed for 3 h. Solvent was
evaporated under reduced pressure and the residue redissolved in toluene (3x5 ml) and
evaporated to yield the crude carboxylic chloride. A solution of the above carboxylic chloride
in dichloromethane (6 ml) was added dropwise to a solution of 1-aminopiperidine (10) (0.19
JPET #49924
9
ml, 1.65 mM) and triethylamine (0.23 ml, 1.65 mM) in dichloromethane (6.2 ml). After
stirring at room temperature for 1 h, the reaction mixture was added with brine and extracted
with dichloromethane (3x15 ml). The combined extracts were washed with brine, dried over
anhydrous sodium sulfate, filtered and evaporated to give a yellowish compound. The crude
compound was purified by flash chromatography on silica gel eluting with petroleum
ether/ethyl acetate 1/1 to afford the desired carboxamide NESS 0327 as a white solid, (93%
yield); mp 205-206°C (acetone), (lit
2
202°C); Rf 0.68 (petroleum ether/ethyl acetate 1/1);
IR(nujol): 3200 (NH), 1650 (C=O), 1600 (Ar);
1
H-NMR: 1.35-1.53 (m, 2H), 1.58-1.89 (m,
6H), 2.15-2.36 (m, 2H), 2.66 (t, 2H, J = 6.4 Hz), 2.87(t, 4H, J = 5.0 Hz), 6.56 (d, 1H, J = 8.2
Hz), 7.01 (dd, 1H), 7.31 (d, 1H, J = 1.8 Hz), 7.37-7.54 (m, 3H), 7.66 (br s, 1H, exch. with
D
2
O). Anal. C
24
H
23
Cl
3
N
4
O (C, H, Cl, N).
Radioligand Binding Methods. Male CD1 mice weighing 20-25 g (Charles River, Calco,
LC, Italy) were housed in the animal care quarters; temperatures were maintained at 22 ± 2°C
on a 12 h light/dark cycle and food and water were available ad libitum. All experimental
protocols were authorized by the Ethical Committee at the University of Cagliari and
performed in strict accordance with the E.C. regulations for care and use of experimental
animals (EEC N°86/609).
Mice were killed by cervical dislocation, brains (minus cerebellum) and spleens were rapidly
removed and placed on an ice-cold plate. After thawing, tissues were homogenated in 20 vol.
(w/v) of ice-cold TME buffer (50 mM Tris-HCl, 1 mM EDTA and 3.0 mM MgCl
2
, pH 7.4).
The homogenates were centrifuged at 1,086 x g for 10 min at 4°C, and the resulting
supernatants were centrifuged at 45,000 x g for 30 min at 4°C.
[
3
H]-CP 55,940 binding was performed by a modification of the method previously described
(Rinaldi-Carmona et al., 1994). Briefly, the membranes (30-80 µg of protein) were incubated
with 0.5 nM of [
3
H]-CP 55,940 for 1 h at 30 °C in a final volume of 0.5 ml of TME buffer
containing 5 mg/ml of fatty acid-free bovine serum albumin (BSA). Non-specific binding
was estimated in the presence of 1 µM of CP 55,940. All binding studies were performed in
JPET #49924
10
disposable glass tubes pre-treated with Sigma-Cote (Sigma Chemical Co. Ltd., Poole, UK),
in order to reduce non-specific binding. The reaction was terminated by rapid filtration
through Whatman GF/C filters pre-soaked in 0.5% polyethyleneimine using a Brandell 96-
sample harvester (Gaithersburg, MD, USA). Filters were washed five times with 4 ml
aliquots of ice-cold Tris HCl buffer (pH 7.4) containing 1 mg/ml BSA The filter bound
radioactivity was measured in a liquid scintillation counter (Tricarb 2900, Packard, Meridien,
USA) with 4 ml of scintillation fluid (Ultima Gold MV, Packard). Protein determination was
performed by means of Bradford (1976) protein assay using BSA as a standard, according to
the protocol of the supplier (Bio-Rad, Milan, Italy). Drugs were dissolved in DMSO. To
avoid possible undesired effects on radioligand binding, DMSO concentration in the different
assays never exceeded 0.1% (v/v). All experiments were performed in triplicate and results
were confirmed in at least four independent experiments. Data from radioligand inhibition
experiments were analyzed by non-linear regression analysis of a Sigmoid Curve using
Graph Pad Prism program (Graph Pad Software, Inc. San Diego, CA, USA). IC
50
values
were derived from the calculated curves and converted to K
i
values as described previously
(Cheng and Prusoff, 1973).
Mouse Vas Deferens Experiments. Vasa deferentia were obtained from albino CD1 mice
weighing 25-40 g. Tissue was mounted in 10 ml organ bath at an initial tension of 0.5 g using
the method described by Pertwee et al. (1993). The bath contained Krebs-Henseleit solution
(118.2 mM NaCl, 4.75 mM KCl, 1.19 mM KH
2
PO
4
,
25.0 mM NaHCO
3
, 11.0 mM glucose
and 2.54 mM CaCl
2
) which was kept at 37°C and bubbled with 95% O
2
and 5% CO
2
.
Isometric contractions were evoked by stimulation with 0.5 s trains of three pulses of 110%
maximal voltage (train frequency, 0.1 Hz; pulse duration, 0.5 ms) through platinum
electrodes attached to the upper end of each bath and a stainless steel electrode attached to
the lower end. Stimuli were generated by Grass S88K stimulator then amplified
(Multiplexing pulse booster 316S, Ugo Basile Comerio, Va, Italy) and divided to yield
separate outputs to four organ baths. Contractions were monitored by computer using a data
JPET #49924
11
recording and analysis system (PowerLab 400) linked via preamplifiers (QuadBridge) to an
F10 transducer (2Biological Instruments, Besozzo, Va, Italy).
Each tissue was subject to several periods of stimulation. The first of these began after the
tissue had equilibrated in the buffering medium but before drug administration, and
continued for 10 min. The stimulator was then switched off for 15 min, after which the
tissues were subjected to further periods of stimulation each lasting 5 min and separated by a
stimulation-free period. The drugs were added once the contractile responses to electrical
stimulation were reproducible. Preparations were exposed to cumulative increasing
concentrations of WIN 55,212-2 to obtain concentration-response curves either in the
absence (control) or in the presence of NESS 0327 (1 pM, 10 pM or 100 pM) added at a
fixed concentration 20 min before the first concentration of WIN 55,212-2. It was not
possible to reverse the inhibitory effect of cannabinoid on the twitch response by washing
them out of the organ bath. Consequently only one concentration-response curve was
constructed per tissue. DMSO was added instead of the drug. The control dose of DMSO was
the same as the dose added in combination with the highest dose of drug used. DMSO alone
did not inhibit the twitch response (n=6) at the maximum concentration used in the bath (4
µl/ml).
Drug additions were performed in volumes of 10 µl. The effects of the antagonists or
agonists were calculated as percentage of decrease in the pre-drug twitch force. Inhibition of
the electrically evoked twitch response is expressed in percentage terms and has been
calculated by comparing the amplitude of the twitch response after each addition of an
agonist with the amplitude immediately prior to the first addition of the agonist. The pA
2
values for competitive antagonists were calculated by Schild regression analysis
(Arunlakshana and Schild, 1959). Data were plotted as log antagonist concentrations (M) vs.
log (concentration-ratios,-1). It is assumed that when the slope value of the regression line in
the Schild plot does not differ statistically from unity, the pA
2
value represents the
dissociation constant of the antagonist (pK
B
). In each estimate eight isolated tissue
JPET #49924
12
preparations were used. Statistical significance was determined by use of Student’s test and
P<0.05 was considered significant.
[
35
S]GTP
γ
S Binding Assay. Male Sprague-Dawley rats (Charles River, Como, Italy),
weighing 200-250 g, were used in all experiments. Rats were killed by decapitation, their
brains rapidly removed and cerebella were dissected on ice. Cerebella tissue was suspended
in 20 volumes of cold centrifugation buffer (50 mM Tris-HCl, 3 mM MgCl
2
, 1 mM EDTA,
pH 7.4) and homogenated using a homogenizer system (Glas-Col, Terre Haute, IN, USA).
The homogenate was centrifuged at 48,000 x g for 10 min at 4°C. The pellet was then
resuspended in the same buffer, homogenized, and centrifuged as previously. The final P2
pellet was subsequently resuspended in assay buffer (50 mM Tris-HCl, 3 mM MgCl
2
, 0.2
mM EGTA, 100 mM NaCl, pH 7.4), homogenized and diluted to a concentration of
2
µgl with assay buffer. The protein concentration was determined by the method of
Bradford (1976) using bovine serum albumin as a standard according to the protocol of the
supplier (Bio-Rad, Milan, Italy). Membrane aliquots were then stored at –80°C until use.
[
35
S]GTPγS binding was measured as described by Selley et al. (1996). Briefly, rat cerebella
membranes (15 µg of protein) were incubated with drugs for 60 min at 30°C in assay buffer
containing 0.1% fatty acid free bovine serum albumin in the presence of 0.05 nM
[
35
S]GTPγS and 30 µM guanosine-5’-diphosphate (GDP), in a final volume of 1 ml. The
reaction was terminated by rapid filtration using a Packard Unifilter-GF/B, washed 2 times
with 1 ml of ice-cold 50 mM Tris-HCl, pH 7.4 buffer and dried 1 h at 30°C. The radioactivity
on the filters was counted in a liquid microplate scintillation counter (TopCount NXT,
Packard, Meridien, USA) using 50 µl of scintillation fluid (Microscint
TM
20, Packard,
Meridien, USA).
Stock solution of WIN 55,212-2 and NESS 0327 were prepared in DMSO and then diluted in
assay buffer. The final concentration of DMSO was < 0.01%, which had no effect either on
basal or stimulated [
35
S]GTP
γ
binding. WIN 55,212-2 concentration effect curves were
JPET #49924
13
determined by incubating membranes with various concentrations of WIN 55,212-2 (10-
10,000 nM) in the presence of 0.05 nM of [
35
S]GTPγS and 30 µM GDP.
Non specific binding was measured in the presence of 10 µM unlabeled GTPγS. Basal
binding was assayed in the absence of agonist and in the presence of GDP. The stimulation
by agonist was defined as a percentage increase above basal levels (i.e., {[dpm(agonist)-dpm
(no agonist)]/dpm (no agonist)}x 100).
Data are reported as mean
±
S.E.M. of three to six experiments, performed in triplicate.
Nonlinear regression analysis of concentration-response data was performed using Prism 2.0
software (GraphPad Prism Program, San Diego, CA, USA) to calculate E
max
and EC
50
values.
The resulting ED
50
values were used to determine K
e
values for antagonism of the agonist-
stimulated response by antagonist, using the relationship K
e
= [Ant]/(Dr-1), where [Ant] is
the concentration of antagonist, and DR is the ratio of ED
50
values in the presence and
absence of antagonist (Sim et al., 1995). Statistical analyses were carried out using one way
ANOVA followed by Newman-Keuls post-hoc test.
Determination of mouse antinociception. Male CD1 mice weighing 20-25 g (Charles
River, Calco, LC, Italy) were used to assess antinociception by means of the tail flick and hot
plate test. A tail flick meter (Ugo Basile Instruments, Italy) equipped with an irradiant heat
source that focused 2.5 cm of the distal tip of the tail was used. A 15 s cut-off time for heat
exposure was used to avoid cutaneous damage and the intensity of the thermal source was
adjusted to produce a 3-5 s latency in vehicle treated rats.
The effect of the compounds on the reaction time of mice placed on the hot plate (Ugo Basile
Instruments, Italy) (55 ± 0.8°C) was assessed determining the time at which animals first
displayed a nociceptive response (licking the front paws, fanning the hind paws or jumping).
To avoid skin damage, after 40 s (cut-off) the animal was removed from the hot plate. In both
tests each animal was tested prior to drug administration to determine control latency and the
JPET #49924
14
animals were used only in the determination of one time point. Data were transformed to the
% MPE by the following equation (Harris and Pierson, 1964); %MPE = [(test latency –
control latency) / (cut-off - basal latency)] X 100; where the latencies were expressed in
seconds and the cut-off varied depending on the test (tail flick = 15 s; hot plate = 40 s). To
establish the dose-dependent curves, at least four drug doses were used on 10 mice per each
dose and each animal group was used only in the determination of one time point. Mice were
tested 30 minutes after WIN 55,212-2 (2 mg/kg s.c.) or vehicle and up to 120 minutes. NESS
0327 (0.01-1 mg/kg, i.p.) or vehicle were given 20 min before WIN 55,212-2 administration.
WIN 55,212-2 was dissolved (5 ml/kg) in an emulsion of ethanol-cremophor-saline (1:1:18),
NESS 0327 was dissolved in two drops of Tween-80 diluted in distilled water to a volume of
5 ml/kg. Three independent experiments were carried out for ID
50
± S.E.M. determination.
Statistical analyses were carried out using two ways ANOVA followed by Newman-Keuls
post-hoc test.
Materials
. Unless otherwise stated, all materials were obtained from commercial suppliers
and used without purification. Anhydrous solvents such as ethanol, tetrahydrofuran and
DMSO were obtained from Sigma-Aldrich in sure-seal bottles. All reactions involving air- or
moisture-sensitive compounds were performed under a nitrogen atmosphere. Flash column
chromatography was carried out using Merck Silica gel 60 (230-400 mesh ASTM). Thin
layer chromatography was performed with Polygram
SIL N-HR-/HV
254
precoated plastic
sheet (0.2 mm).
1
H-NMR spectra were determined in CDCl
3
with super conducting FT-NMR
using a XL-200 Varian apparatus at 200 MHz. Chemical shifts are reported in
δ
(ppm)
relative to TMS as the internal standard and coupling constants in Hz. Significant
1
H-NMR
data are reported in the following order: multiplicity (s, singlet; d, doublet; t, triplet; q,
quartet; m, multiplet; dd, double doublet; br s, broad singlet), number of protons, coupling
constants (J) in Hz. IR spectra were recorded as thin films or nujol mulls on NaCl plates with
a Perkin-Elmer 781 IR spectrophotometer and are expressed in
ν
(cm
-1
). Melting points were
JPET #49924
15
determined on a K fler melting point apparatus and are uncorrected. Compounds are
indicated by the molecular formula followed by the symbols for the elements (C, H, N) and
were found to be within
±
0.4% of their theoretical values. [
3
H]-CP 55, 940 (180 Ci/mmol)
and [
35
S]GTPγS (1200-1350 Ci/mmole) were purchased from New England Nuclear (Boston,
MA, USA). CP 55,940 and WIN 55,212-2 were obtained from Tocris Cookson Ltd (Bristol,
UK). GDP and GTPγS were obtained from Sigma/RBI (St. Louis, MO, USA). SR 141716A
and SR 144528 were kindly provided by Sanofi-Syntlabo (Bagneux, France).
JPET #49924
16
Results
Chemistry
Target compound NESS 0327 was prepared as shown in fig 1. Acid 9, prepared via the ester
8 by saponification, was activated with thionyl chloride and, without isolation of the
intermediate acyl chloride, reacted with a stoichiometric amount of N-amino-piperidine, in
presence of triethylamine. Ester 8 was prepared starting from the aldehyde 1, which
submitted to a Wittig condensation with the phosphonium bromide 2 yielded the pentenoic
acid derivate 3. Reduction of the double bond of 3 to give 4 with H
2
over PtO
2
in EtOH
(Adams’ catalyst), followed by transformation into the corresponding acyl chloride with
thionyl chloride, and cyclization, with AlCl
3
in dichloromethane, afforded the
benzocycloheptanone 5. This benzocyclanone reacted with diethyl oxalate by means of
NaOEt in EtOH to provide the
α,γ
-diketoester 6, that was allowed to react with 2,4-
dichlorophenylhydrazine hydrochloride 7 to yield the educt 1H-pyrazole-3-carboxylic acid
ethyl ester 8. (fig 1)
Biology
The affinity of NESS 0327 for the cannabinoid CB
1
receptor in mouse forebrain membranes
was evaluated using competitive binding assay. As shown in fig. 2A the specific binding of
[
3
H]-CP 55,940 to its high affinity receptor in mouse brain synaptosomal membranes was
totally displaced by NESS 0327 in a concentration dependent manner with K
i
values of 350 ±
5 fM (n=4). Both SR 141716A and SR 144528 compete for CB
1
receptor with K
i
values of
1.8 ± 0.075 nM and 70 ± 7 nM (n=4) respectively, in close agreement with published values
(Rinaldi-Carmona et al., 1994, 1998). The affinities of NESS 0327, SR 141716A and SR
144528 for CB
2
receptor were determined in mouse spleen (fig. 2B). The concentration-
response gave K
i
values of 21 ± 0.5 nM, 514 ± 30 nM and 0.28 ± 0.04 nM (n=4) for NESS
0327, SR 141716A and SR 144528, respectively. These results show that NESS 0327 is over
60,000 fold selective for the CB
1
receptor versus CB
2
receptor. NESS 0327 was screened for
JPET #49924
17
cannabinoid agonist activity using mouse vas deferens model. Cannabinoid agonists inhibit
the electrically induced contractions of the mouse vas deferens via activation of inhibitory
CB
1
receptors present on the sympathetic nerve terminals (Pertwee, 1997). As shown in fig.
3, WIN 55,212-2 induced a concentration-dependent inhibition of the twitch contractions in
the mouse isolated vas deferens preparations, with pD
2
values of 8.45 ± 0.05. NESS 0327,
which alone had no effect up to 1 µM, produced a concentration-dependent rightward and
almost parallel shift of the concentration response-curve for WIN 55,212-2 showing that it
behaved as a competitive antagonist versus the synthetic cannabinoid agonist with pA
2
value
of 12.46 ± 0.23 and with a slope in the Schild plot not significantly different from unity (1.03
± 0.05, P>0.05).
Efficacy of the compound at the CB
1
receptor was measured using ligand stimulation of
[
35
S]GTPγ binding to cerebellar membranes. [
35
S]GTPγ binding was stimulated in a
concentration-dependent and saturable manner by WIN 55,212-2 with ED
50
and E
max
values
of 0.16 ± 0.01 µM and 286 ± 24% (stimulation above basal binding), respectively (table 1).
In order to determine the ability of NESS 0327 to antagonize CB
1
agonist-stimulated
activation of G-protein the effect of three concentrations of NESS 0327 (0.1, 1, 10 nM) on
the log concentration-response curve of WIN 55,212-2 was investigated. NESS 0327
produced concentration-dependent rightward shift of the WIN 55,212-2 concentration
response-curve (one way ANOVA: F(3,14) = 43.35, P<0.01) without affecting the E
max
of
the agonist (table 1). NESS 0327 at concentrations of 0.1, 1 and 10 nM shifted the dose
response curve for WIN 55,212-2 to the right with calculated K
e
values of 80.3 ± 20, 283 ±
11 and 2016 ± 226 pM, respectively. NESS 0327, at concentrations from 0.1 through 1 µM,
had no effect on [
35
S]GTPγS binding, while, in the same conditions, SR 141716A at
concentration of 1 µM produced an inhibition of 21 ± 2 % of basal [
35
S] GTPγS binding
(data not shown). The lack of effect on basal [
35
S]GTP
γ
S binding suggests that NESS 0327
had no appreciable negative intrinsic activity in brain under the conditions used in this study.
JPET #49924
18
The in vivo antagonism of NESS 0327 for the cannabinoid receptor was investigated in an
animal model classically used to study cannabinoid drug effects. As shown in fig. 4A and fig.
4B, NESS 0327 dose dependently reduced the analgesia induced by the cannabinoid agonist
WIN 55,212-2 (2 mg/kg s.c.) on both tail flick (Two ways ANOVA: F
dose
(6, 189) = 10.26,
P<0.01; F
time
(2, 189) = 7.22, P<0.01; F
interact
(12, 189) = 3.7, P<0.01 and hot plate (Two
ways ANOVA: F
dose
(6, 189) = 42.37, P<0.01; F
time
(2, 189) = 14.20, P<0.01; F
interact
(12,
189) = 5.4, P<0.01; a complete antagonism was reached at the dose of 0.1 mg/kg in the tail
flick test (P>0.05 vs. vehicle treated rats) and of 0.05 mg/kg in the hot plate test (P>0.05 vs.
vehicle treated rats). The ability of NESS 0327 to inhibit the antinociceptive effect induced
by WIN 55,212-2 was maintained during the observation period. 30 min after WIN 55,212-2
injection, NESS 0327 showed a ID
50
= 0.042 ± 0.01 mg/kg i.p. in the tail flick and ID
50
=
0.018 ± 0.006 mg/kg i.p.in the hot plate test. Furthermore, NESS 0327 did not show any
antinociception activity per se (data not shown), suggesting that it is devoid of inverse
agonist activity and it should be regarded as a pure antagonist.
JPET #49924
19
Discussion
Given the role of the endogenous cannabinoid system in different physiological responses
and its involvement in numerous pathological processes, the search of new and selective
agonists/antagonists of the CB
1
and CB
2
cannabinoid receptor will constitute an important
line of research in the forthcoming years. In this respect, NESS 0327 showed an high
selectivity for CB
1
vs. CB
2
receptors and the in vitro functional assays (isolated organ and
GTPγS) as well the in vivo antinociceptive studies indicated that the compound behaves as
antagonist of the CB
1
receptor. However since the relative binding affinity of NESS 0327 for
the CB1 receptor is about 5,000 times more than that of SR 141716A, the in vivo experiment
whereas the relative difference in activity is only ten times might suggest a poor central
bioavailability of NESS 0327.
NESS 0327 was selected as a lead compound from a series of potential cannabinoid receptor
antagonists (not shown) because it displayed the highest affinity for the CB
1
subtype of the
cannabinoid receptor. Structure relationship inferential reasoning would suggest that a proper
low-energy constrained conformation of the NESS 0327 semirigid tricyclic unit may relate to
its potent and selective affinity for the CB
1
receptor with respect to the parent compound SR
141716A. On the basis of the remarkable result further synthesis of analogues derived from
manipulation in the tricyclic 1,4,5,6-tetrahydrobenzo [6,7] cyclohepta [1,2-c] pyrazole
backbone and variation of substitution on either N
1
-aromatic ring and the aminopiperidine
carboxamide region, may facilitate the elucidation of the cannabinoid pharmacophore for
CB
1
selective antagonist.
Development of cannabinoid receptor selective antagonists will provide the tools necessary
for a better understanding of the cannabinoid receptor functions both in the central nervous
system and in the peripheral immune system. In this respect, considering the higher
selectivity for the CB1 receptor, NESS 0327 may prove to be more advantageous when
compared to the classical CB
1
receptor antagonist SR 141716A.
JPET #49924
20
Current views of the interaction between CB
1
/CB
2
receptors and signal transducting G-
proteins interaction are described in the general framework of allosteric modulation, in which
the receptor isomerizes between an active or inactive form (Samama et al., 1993; Nakamura-
Palacios et al., 1999). Therefore, more detailed studies will be needed to address whether
NESS 0327 may affect the distribution between the active or inactive states of the
cannabinoid receptor (as for a neutral-competitive antagonist) or, on the contrary, may
enhance the accumulation of the receptor in the inactive state (as for an inverse agonist). SR
141716A for instance, has been shown to stimulate cAMP production, providing evidence for
an inverse agonist effect (Mato et al., 2002). It has been further demonstrated that SR
141716A has a peculiar inverse-antagonist activity that is consistent with the stabilization of
an inactive receptor/Gi protein complex. Accordingly, SR 141716A could cause a depletion
of Gi and thus render the protein unavailable for the inhibitory action of other ligands
(Bouaboula et al., 1997). The availability of new and selective ligands, such as NESS 0327,
for the cannabinoid receptor CB
1
would allow a better conceptualisation of the rather
complex mode of cannabinoid receptor/ligand interaction, since
9
-THC itself has been
shown to be a weak but very selective antagonist for the cannabinoid receptor CB
2
(Bayewitch et al., 1996; Barth and Rinaldi-Carmona, 1999). Since recent data using SR
141716A seem to suggest a ligand-independent activity for cannabinoid receptor signalling
(Mato et al., 2002), NESS 0327 could be employed as a more selective antagonist for the
CB
1
receptors, to study the recent proposed ability of the CB1 receptor to sequester G-
proteins from a common pool and prevent other G-protein-coupled receptors from signalling
(Vasquez and Lewis, 1999).
The use of antagonists in studies investigating the biology of cannabinoid receptors may help
to distinguish between receptor-dependent and receptor-independent effects elicited by
cannabinoid agonists. A large arsenal of cannabinoid receptor antagonists will be
instrumental in characterizing both the well-known and eventually, newly discovered,
cannabinoid receptor subtypes. The availability of a compound such as NESS 0327
JPET #49924
21
displaying femtomolar affinity for the CB1 receptor would consequently allow radioactive
labelling of the latter, thus enabling the study of CB1 cellular and tissutal distribution in
further detail. Stringent screening techniques might also be of use in the characterization of
new cannabinoid receptors.
Additional in vivo experiments should provide further evidence for the clinical potential of
this powerful CB
1
antagonist. It should be determined whether NESS 0327 would show
better efficacy as a CB
1
antagonist in animal models of excessive food-intake, psychosis and
cognitive impairment, three area of possible interest for a novel CB
1
selective antagonist.
JPET #49924
22
JPET #49924
23
References
Abood ME and Martin BR (1992) Neurobiology of marijuana abuse. Trends Pharmacol Sci
13:201
¯
206.
Arunlakshana O and Schild HO (1959) Some quantitative uses of drug antagonists. Br J
Pharmacol 14:48-58.
Barth F and Rinaldi-Carmona M (1999) The development of cannabinoid antagonists. Curr
Med Chem 6(8):745-755.
Bayewitch M, Rhee MH, Avidor-Reiss T, Breuer A, Mechoulam R and Vogel Z (1996) (-)-
Delta9-tetrahydrocannabinol antagonizes the peripheral cannabinoid receptor-mediated
inhibition of adenylyl cyclase. J Biol Chem 271(17):9902-9905.
Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram
quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248-254.
Bouaboula M, Perrachon S, Milligan L, Canat X, Rinaldi-Carmona M, Portier M, Barth F,
Calandra B, Pecceu F, Lupker J, Maffrand JP, Le Fur G and Casellas P (1997) A selective
inverse agonist for central cannabinoid receptor inhibits mitogen-activated protein kinase
activation stimulated by insulin or insulin-like growth factor 1. Evidence for a new model of
receptor/ligand interactions. J Biol Chem 272(35):22330-22339.
Cheng Y and Prusoff WH (1973) Relationship between the inhibition constant (K
i
) and the
concentration of inhibitor which causes 50 per cent inhibition (IC
50
) of an enzymatic
reaction. Biochem Pharmacol 22:3099
¯
3108.
Compton DR, Jhonson MR, Melvin LS and Martin BR (1992) Pharmacological profile of a
series of bicyclic cannabinoid analogs: classification as cannabimimetic agents. J Pharmacol
Exp Ther 260:201
¯
209.
JPET #49924
24
Compton DR, Rice KC, De Costa BR, Razdan RK, Melvin LS, Johnson MR and Martin BR
(1993) Cannabinoid structure-activity relationships: correlation of receptor binding and in
vivo activities. J Pharmacol Exp Ther 265:218-226.
Compton DR, Aceto MD, Lowe J and Martin BR (1996) In vivo characterization of a
specific cannabinoid receptor antagonist (SR 141716A): inhibition of
9
-
tetrahydrocannabinol-induced responses and apparent agonist activity. J Pharmacol Exp Ther
277:586
¯
594.
Devane WA, Dysarz FA, Johnson MR, Melvin LS and Howlett AC (1988) Determination
and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol 34:605-613.
Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D,
Mandelbaum A, Etinger A and Mechoulam R (1992) Isolation and structure of a brain
constituent that binds to the cannabinoid receptor. Science 258:1946-1949.
Formukong EA, Evans AT and Evans FJ (1989) The medicinal uses of cannabis and its
constituents. Psytother Res 3:219-231.
Galiegue S, Mary S, Marchand J, Dussossoy D, Carriere D, Carayon P, Bouaboula M, Shire
D, Le Fur G and Casellas P (1995) Expression of central and peripheral cannabinoid
receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem 232:54-61.
Gralla RJ (1999) Cannabinoids and the control of chemotherapy-induced nausea and
vomiting, in Marijuana and Medicine (Nahas GG, Sutin KM, Harvey DJ and Agurell S eds)
pp 599-610, Human Press, Totowa, NJ.
Harris LS and Pierson AK (1964) Some narcotic antagonists in the benzomorphan series. J
Pharmacol Exp Ther 143:141-148.
Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, de Costa BR and Rice KC
(1990) Cannabinoid receptor localization in brain. Proc Natl Acad Sci 87:1932-1936.
JPET #49924
25
Howlett AC (1995) Pharmacology of cannabinoid receptors. Annu Rev Pharmacol Toxicol
35:607-634.
Howlett AC (1998) The CB1 cannabinoid receptor in the brain. Neurobiol Dis 5:405-416.
Lichtman AH and Martin BR (1997) The selective cannabinoid antagonist SR 141716A
blocks cannabinoid-induced antininoception in rats. Pharmacol Biochem Behav 57:7
¯
12.
Martin BR (1986) Cellular effects of cannabinoids. Pharmacol Rev 38:45-74.
Mato S, Pazos A and Valdizan EM (2002) Cannabinoid receptor antagonism and inverse
agonism in response to SR141716A on cAMP production in human and rat brain. Eur J
Pharmacol 443(1-3):43-46.
Mattes RD, Engelman K, Shaw LM and Elsohly MA (1994) Cannabinoids and appetite
stimulation. Pharmacol Biochem Behav 49:187-195.
Munro S, Thomas KL and Abu-Shaar M (1993) Molecular characterization of a peripheral
receptor for cannabinoids. Nature 365:61-65.
Nakamura-Palacios EM, Moerschbaecher JM and Barker LA (1999) The pharmacology of
SR 141716A: A Review. CNS Drug Review 5:43-58.
Pertwee RG, Stevenson LA and Griffin G (1993) Cross-tolerance between delta-9-
tetrahydrocannabinol and the cannabimimetic agents, CP 55,940, WIN 55,212-2 and
anandamide. Br J Pharmacol 110(4):1483-1490.
Pertwee RG (1997) Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther
74:129-180.
Pertwee RG (1999) Cannabis and cannabinoids: pharmacology and rationale for clinical use.
Fortsch Komplementarmed 3:12-15.
JPET #49924
26
Porcella A, Maxia C, Gessa GL, Pani L (2001) The synthetic cannabinoid WIN55212-2
decreases the intraocular pressure in human glaucoma resistant to conventional therapies. Eur
J Neurosci 13(2):409-412.
Rinaldi-Carmona M, Barth F, Heaulme M, Shire D, Calandra B, Congy C, Martinez S,
Maruani J, Neliat G, Caput D, Ferrara P, Soubriè P, Brelière JC and Le Fur G (1994)
SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett
350:240-244.
Rinaldi-Carmona M, Barth F, Millan J, Derocq JM, Casellas P, Congy C, Oustric D, Sarran
M, Bouaboula M, Calandra B, Portier M, Shire D, Breliere JC and Le Fur GL (1998) SR
144528, the first potent and selective antagonist of the CB2 cannabinoid receptor. J
Pharmacol Exp Ther 284(2):644-650.
Samama P, Cotecchia S, Costa T and Lefkowitz RJ(1993) A mutation-induced activated state
of the beta 2-adrenergic receptor. Extending the ternary complex model. J Biol Chem
268(7):4625-4636.
Selley DE, Stark S, Sim LS and Childers SR (1996) Cannabinoid receptor stimulation of
guanosine-5’-o-(3-[
35
S]thio) triphosphate binding in rat brain membranes. Life Sci 59(8):659-
668.
Sim LJ, Selley DE, Childers SR (1995) In vitro autoradiography of receptor-activated G
proteins in rat brain by agonist-stimulated guanylyl 5’-[γ-[35S]thio]triphospate binding. Proc
Natl Acad Sci USA: 92:7242-7246.
Vasquez C and Lewis DL (1999) The CB1 cannabinoid receptor can sequester G-proteins,
making them unavailable to couple to other receptors. J Neurosci 19(21):9271-9280.
Voth EA and Schwartz RH (1997) Medicinal applications of delta-9-tetrahydrocannabinol
and marijuana. Ann Intern Med 126:791-798.
JPET #49924
27
Footnotes
2
Dpt. Farmaco Chimico Tossicologico, University of Sassari, 07100 Sassari, Italy;
3
“B.B.
Brodie” Dpt of Neuroscience, University of Cagliari, 09124 Cagliari, Italy,
4
Section of
Human Physiology and Nutrition, Dpt. of Applied Sciences to Biosystems, University of
Cagliari, 09124 Cagliari, Italy,
5
C.N.R. Institute of Neurogenetics and Neuropharmacology
and Neuroscienze S.c.a r.l.
JPET #49924
28
Legends
Fig.1. Schematic synthesis of NESS 0327
Fig.2. Competitive inhibition of [
3
H]-CP 55,940 binding in mouse brain (A) and mouse
spleen (B) by NESS 0327, SR 141716A and SR 144528. Binding assays were carried out at
30°C using 0.5 nM [
3
H]-CP 55,940 and increasing concentrations of drugs. Data are mean ±
S.E.M. of at least four different experiments, each performed in triplicate.
Fig. 3. Cumulative concentration –response curves for WIN 55,212-2 on the amplitude of
twitch contractions elicited by electrical field stimulation of the mouse vas deferens obtained
in the presence of its vehicle, DMSO, (
) (control) and in the presence of NESS 0327 at 1
pM (
), 10 pM ( ) or 100 pM ( ). Assays were performed as described under Methods”.
Each symbol represents the mean value ± S.E.M of inhibition of electrically evoked
contractions of vasa deferentia expressed as a percentage of the amplitude of the twitch
response measured before the first addition of WIN 55,212-2 to the organ bath (n= 6-8
different preparations). NESS 0327 was added 20 min before the first addition of WIN
55,212-2.
Fig. 4. Inhibition by NESS 0327 of WIN 55,212-2 induced antinociception in the tail flick
(A) and in the hot plate test (B). Mice were tested after 30, 60 and 120 min after
administration of WIN 55,212-2 (2 mg/kg s.c.) or vehicle. NESS 0327 (0.01-1 mg/kg) or
vehicle were administered i.p. 20 minutes before WIN 55,212-2 injection. Each column
represents the mean
±
S.E.M of the %MPE obtained from ten animals. Statistical analysis
was carried out using two ways ANOVA followed by Newman Keuls post-hoc test (* P<0.05
and ** P<0.01).
JPET #49924
29
Table 1
ED
50
and E
max
values
of WIN 55,212-2 in stimulating [
35
S]GTP
γ
S binding in the presence or
absence of NESS 0327
Compounds ED
50
M) E
max
(% Basal Binding)
WIN 55,212-2
0.16
±
0.01
**
286
±
24
WIN 55,212-2 + NESS 0327 (0.1 nM)
0.36
±
0.07
**
269
±
42
WIN 55,212-2 + NESS 0327 (1 nM)
0.79
±
0.13
**
362
±
42
WIN 55,212-2+ NESS 0327 (10 nM)
1.11
±
0.03
**
200
±
21
[
35
S]GTP
γ
S binding was performed in rat cerebella membranes with 30 µM of GDP and 8-10
concentrations of WIN 55,212-2 in the absence and presence of 0.1, 1, and 10nM of NESS
0327. Data are expressed as percent of basal [
35
S]GTP
γ
S binding. The values represent mean
± S.E.M. of 4-6 separate experiments, each performed in triplicate. ED
50
and E
max
values
were calculated from non-linear
regression curve fitting using Graphpad prism program.
The statistical significance of differences between the groups was assessed by one-way
analysis of variance (ANOVA) following by the Neuman-Keuls test (*P<0.05 and
**
P<0.001
versus WIN 55,212-2)
... Differential physiological and behavioural processes have been ascribed to both forms of tonic eCB signalling suggesting that both pathways are potential mechanisms Meye et al., 2013;Sink et al., 2010). To address this question, we utilized a neutral antagonist, NESS0327, which only blocks agonist driven CB 1 receptor activation but does not influence constitutive receptor signalling (Ruiu et al., 2003). Similar to what was found following administration of AM251, NESS0327 increased Fos expression in the PVN, elevated CORT and increased grooming behaviour. ...
Article
Full-text available
Background and Purpose Endocannabinoid (eCB) signalling gates many aspects of the stress response, including the hypothalamic–pituitary–adrenal (HPA) axis. The HPA axis is controlled by corticotropin releasing hormone (CRH) producing neurons in the paraventricular nucleus of the hypothalamus (PVN). Disruption of eCB signalling increases drive to the HPA axis, but the mechanisms subserving this process are poorly understood. Experimental Approach Using an array of cellular, endocrine and behavioural readouts associated with activation of CRH neurons in the PVN, we evaluated the contributions of tonic eCB signalling to the generation of a stress response. Key Results The CB1 receptor antagonist/inverse agonist AM251, neutral antagonist NESS243 and NAPE PLD inhibitor LEI401 all uniformly increased Fos in the PVN, unmasked stress‐linked behaviours, such as grooming, and increased circulating CORT, recapitulating the effects of stress. Similar effects were also seen after direct administration of AM251 into the PVN, while optogenetic inhibition of PVN CRH neurons ameliorated stress‐like behavioural changes produced by disruption of eCB signalling. Conclusions and Implications These data indicate that under resting conditions, constitutive eCB signalling restricts activation of the HPA axis through local regulation of CRH neurons in the PVN.
... RIM is an inverse agonist, so effects of RIM may be because of increased constitutive activity of CB1Rs in CFA-treated rats (Ruiu et al., 2003). To determine whether the increased effect of RIM was a result of increased CB1R constitutive activity or eCB tone, we again tested for eCB tone but with the CB1R neutral antagonist NESS-0327 (0.5 mM). ...
Article
Full-text available
Cannabinoid-targeted pain therapies are increasing with the expansion of cannabis legalization, however, their efficacy may be limited by pain-induced adaptations in the cannabinoid system. Cannabinoid receptor subtype 1 (CB1Rs) inhibition of spontaneous, miniature and evoked GABAergic postsynaptic currents (mIPSCs and eIPSCs) in the ventrolateral periaqueductal gray (vlPAG) were compared in slices from naïve and inflamed male and female Sprague-Dawley rats. Complete Freund’s Adjuvant (CFA) injections into the hindpaw induced persistent inflammation. In naïve rats, exogenous cannabinoid agonists robustly reduce both eIPSCs and mIPSCs. After 5-7 days of inflammation, the effects of exogenous cannabinoids are significantly reduced due to CB1R desensitization via GRK2/3, as function is recovered in the presence of the GRK2/3 inhibitor, Compound 101 (Cmp101). Inhibition of GABA release by presynaptic mu opioid receptors (MORs) in the vlPAG does not desensitize with persistent inflammation. Unexpectedly, while CB1R desensitization significantly reduces inhibition produced by exogenous agonists, depolarization-induced suppression of inhibition (DSI) protocols that promote 2-AG synthesis exhibit prolonged CB1R activation after inflammation. 2-AG tone is detected in slices from CFA-treated rats when GRK2/3 is blocked suggesting an increase in 2-AG synthesis after persistent inflammation. Inhibiting 2-AG degradation with the monoacylglycerol lipase (MAGL) inhibitor JZL184 during inflammation results in desensitization of CB1Rs by eCBs that is reversed with Cmp101. Collectively, these data indicate that persistent inflammation primes CB1Rs for desensitization, and MAGL degradation of 2-AG protects CB1Rs from desensitization in inflamed rats. These adaptations with inflammation have important implications for the development of cannabinoid-based pain therapeutics targeting MAGL and CB1Rs. Significance statement: Presynaptic G protein-coupled receptors are resistant to desensitization. Here we find that persistent inflammation increases endocannabinoid levels, priming presynaptic cannabinoid 1 receptors for desensitization upon subsequent addition of exogenous agonists. Despite the reduced efficacy of exogenous agonists, endocannabinoids have prolonged efficacy after persistent inflammation. Endocannabinoids readily induce cannabinoid 1 receptor desensitization if their degradation is blocked, indicating that endocannabinoid concentrations are maintained at sub-desensitizing levels and that degradation is critical for maintaining endocannabinoid regulation of presynaptic GABA release in the ventrolateral periaqueductal gray during inflammatory states. These adaptations with inflammation have important implications for the development of cannabinoid-based pain therapies.
... The IC 50 for Win-2-mediated inhibition of [Ca 21 ] i spiking was 48 ± 13 nM, an 18-fold lower potency relative to that described for inhibition of synaptic activity in rodent models (Shen et al., 1996). The inhibition of [Ca 21 ] i spiking by Win-2 was completely blocked by the selective CB 1 R antagonist NESS 0327 (Ruiu et al., 2003) (Fig. 3, C and E), indicating that Win-2 acted on functional CB 1 Rs in these neurons. Application of 100 nM NESS 0327 did not consistently increase [Ca 21 ] i spiking activity (t (14) 5 1.9 P 5 0.07), suggesting that eCB tone was not sufficiently elevated under these conditions to suppress synaptic transmission. ...
Article
Full-text available
The endocannabinoid system (ECS) modulates synaptic function to regulate many aspects of neurophysiology. It adapts to environmental changes and is affected by disease. Thus, the ECS presents an important target for therapeutic development. Despite recent interest in cannabinoid-based treatments, few preclinical studies are conducted in human systems. Human induced pluripotent stem cells (hiPSCs) provide one possible solution to this issue. However, it is not known if these cells have a fully functional ECS. Here, we show that hiPSC-derived neuron/astrocyte cultures exhibit a complete ECS. Using Ca2+ imaging and a genetically encoded endocannabinoid sensor, we demonstrate that they not only respond to exogenously applied cannabinoids but also produce and metabolize endocannabinoids. Synaptically driven [Ca2+]i spiking activity was inhibited (EC50 = 48 ± 13 nM) by the efficacious agonist [R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrolol [1,2,3-de]-1,4-benzoxazin-yl]-(1-naphthalenyl)methanone mesylate] (Win 55,212-2) and by the endogenous ligand 2-arachidonoyl glycerol (2-AG; EC50 = 2.0 ± 0.6 µm). The effects of Win 55212-2 were blocked by a CB1 receptor-selective antagonist. Δ9-Tetrahydrocannabinol acted as a partial agonist, maximally inhibiting synaptic activity by 47 ± 14% (EC50 = 1.4 ± 1.9 µm). Carbachol stimulated 2-AG production in a manner that was independent of Ca2+ and blocked by selective inhibition of diacylglycerol lipase. 2-AG returned to basal levels via a process mediated by monoacylglycerol lipase as indicated by slowed recovery in cultures treated with 4-[Bis(1,3-benzodioxol-5-yl)hydroxymethyl]-1-piperidinecarboxylic acid 4-nitrophenyl ester (JZL 184). Win 55,212-2 markedly desensitized CB1 receptor function following a 1-day exposure, whereas desensitization was incomplete following 7-day treatment with JZL 184. This human cell culture model is well suited for functional analysis of the ECS and as a platform for drug development. SIGNIFICANCE STATEMENT: Despite known differences between the human response to cannabinoids and that of other species, an in vitro human model demonstrating a fully functional endocannabinoid system has not been described. Human induced pluripotent stem cells (hiPSCs) can be obtained from skin samples and then reprogrammed into neurons for use in basic research and drug screening. Here, we show that hiPSC-derived neuronal cultures exhibit a complete endocannabinoid system suitable for mechanistic studies and drug discovery.
... The copyright holder for this preprint this version posted September 28, 2022. ; https://doi.org/10.1101/2022.09.27.509585 doi: bioRxiv preprint question, we utilized a neutral antagonist, NESS0327, which only blocks agonist driven CB1 receptor activation but does not influence constitutive receptor signaling (Ruiu et al., 2003). Similar to what was found following administration of AM251, NESS0327 increased c-fos expression in the PVN, elevated CORT and increased grooming behavior. ...
Preprint
Full-text available
Endocannabinoid (eCB) signaling gates many aspects of the stress response, including the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis is controlled by corticotropin-releasing hormone (CRH) producing neurons in the paraventricular nucleus of the hypothalamus (PVN). Disruption of eCB signaling increases drive to the HPA axis, but the mechanisms subserving this process are poorly understood. Using an array of cellular, endocrine, and behavioral readouts associated with the activation of CRH neurons in the PVN, we evaluated the contributions of tonic eCB signaling to the generation of a stress response. The CB1 receptor antagonist/inverse agonist AM251, neutral antagonist NESS243, and NAPE PLD inhibitor LEI401 all uniformly increased c-fos in the PVN, unmasked stress-linked behaviors, such as grooming, and increased circulating CORT, recapitulating the effects of stress. Similar effects were also seen after direct administration of AM251 into the PVN, while optogenetic inhibition of PVN CRH neurons ameliorated stress-like behavioral changes produced by disruption of eCB signaling. These data indicate that under resting conditions, constitutive eCB signaling restricts activation of the HPA axis through local regulation of CRH neurons in the PVN.
... The ability of low efficacy agonists to antagonize the responses of higher efficacy compounds is well known-but the in vitro signaling data reported in this study suggests that some compounds are close to neutral antagonists. For the purposes of comparing the characteristics of analogue 157, cAMP assays were also performed for previously published CB 1 neutral antagonists NESS-0327 28,29 and AM4113, 30,31 in comparison to SR141716A (a well-known CB 1 inverse agonist). ...
Article
Full-text available
Neutral antagonists of GPCRs remain relatively rare-indeed, a large majority of GPCR antagonists are actually inverse agonists. The synthetic cannabinoid receptor agonist (SCRA) EG-018 was recently reported as a low efficacy cannabinoid receptor agonist. Here we report a comparative characterization of EG-018 and 13 analogues along with extant putative neutral antagonists of CB1 . In HEK cells stably expressing human CB1 , assays for inhibition of cAMP were performed by real-time BRET biosensor (CAMYEL), G protein cycling was quantified by [35 S]GTPγS binding, and stimulation of pERK was characterized by AlphaLISA (PerkinElmer). Signaling outcomes for the EG-018 analogues were highly variable, ranging from moderate efficacy agonism with high potency, to marginal agonism at lower potency. As predicted by differing pathway sensitivities to differences in ligand efficacy, most EG-018-based compounds were completely inactive in pERK alone. The lowest efficacy analogue in cAMP assays, 157, had utility in antagonism assay paradigms. Developing neutral antagonists of the CB1 receptor has been a long-standing research goal, and such compounds would have utility both as research tools and in therapeutics. Although these results emphasize again the importance of system factors in determining signaling outcomes, some compounds characterized in this study appear among the lowest efficacy agonists described to date and therefore suggest that development of neutral antagonists is an achievable goal for CB1 .
... Для выбора между двумя версиями тоничес кой активности -со стороны тонически высво бождаемых эндоканнабиноидов или конститу тивной (независимой от действия эндоканнаби ноидов) тонической активности пресинаптичес ких СВ1 -предпринимается использование жи вотных с нокаутированными генами липаз, ли митирующих синтез эндоканнабиноидов, что исключает их тоническую выработку и высво бождение [41], или используется сравнительный анализ эффектов обратных агонистов/антаго нистов СВ по сравнению с эффектами нейтраль ных антагонистов рецепторов (которые уже раз работаны и не вызывают эффектов, связанных с обратным агонизмом, например, NESS 0327, O 2654, O 2050) [37,42,43]. Еще одним экспе риментальным способом является тестирование утечки эндоканнабиноидов из проб тканей моз га, где также был установлен высокий уровень базальной активности эндоканнабиноидов и возможности их конститутивного синтеза и то нического выделения [44]. ...
... It has also been shown that stimulation of the activity of 2 AG and AEA degrading enzymes (monoacylglycerol lipase and fatty acid amide hydrolase, respectively) leads to attenuation of the tonic endocannabinoid mediated inhibition of the GABAergic system, which is supported by the fact that these enzymes that control basal production of endo cannabinoids, are tonically active in certain brain regions [40]. To choose between the two models of tonic endo cannabinoid activity, i.e., between the tonic mediated endocannabinoid release and constitutive tonic activity of presynaptic CB1 receptors in the absence of endo cannabinoids, the lipase knockout animals that have lim ited capacity of endocannabinoid production are used, which makes tonic endocannabinoid synthesis and release impossible [41], as well as comparative analysis of the effects produced by the inverse agonists/antagonists and by the neutral antagonists of CB receptors, which have been developed (NESS 0327, O 2654, O 2050) and do not cause the effects typical of inverse agonists [37,42,43]. Another experimental approach involves testing of the endocannabinoid leakage from the brain tissue samples, where basal activity of endocannabinoids and possibility of their tonic synthesis and release have been established [44]. ...
Article
Full-text available
This review focuses on new aspects of endocannabinoid functions and mechanisms of activity in central and peripheral synapses, different from the general viewpoint that endocannabinoids are retrograde signaling molecules, which inhibit neurotransmitter release by activating specific presynaptic endocannabinoid receptors CB1 and CB2. Biased agonism of the endogenous and synthetic cannabinoids as well as ability of the CB-receptors to couple not only with classical Gi-proteins, but also with Gs- and Gq-proteins and, moreover, with β-arrestins (thereby triggering additional signaling pathways in synapses) are described here in detail. Examples of noncanonical tonic activity of endocannabinoids and their receptors and their role in synaptic function are also presented. The role of endocannabinoids in short-term and long-term potentiation of neurotransmitter release in central synapses and their facilitating effect on quantal size and other parameters of acetylcholine release in mammalian neuromuscular junctions are highlighted in this review. In conclusion, it is stated that the endocannabinoid system has a wider range of various multidirectional modulating effects (both potentiating and inhibiting) on neurotransmitter release than initially recognized. Re-evaluation of the functions of endocannabinoid system with consideration of its noncanonical features will lead to better understanding of its role in the normal and pathological functioning of the nervous system and other systems of the body, which has an enormous practical value.
Article
Background and Purpose AKB48 is a synthetic cannabinoid illegally sold for its psychoactive cannabis‐like effects that has been associated to several acute intoxications and which effects are poorly known. Experimental Approach Using a behavioural, neurochemical and immunohistochemical approach we investigated the pharmaco‐toxicological effects, plasma pharmacokinetic and neuroplasticity at cannabinoid CB 1 receptor (CB 1 R) in the cerebellum and cortex induced by repeated AKB48 administration in male and female mice. Key Results The effects of AKB48 varied significantly depending on sex and length of treatment. The 1 st injection impaired sensorimotor responses and reduced body temperature, analgesia, and breath rate at a greater extent in females than in males, the 2 nd injection induced stronger effects in males while the 3 rd injection of AKB48 induced weaker responses in both sexes, suggesting the emergence of tolerance. The CB 1 R antagonist NESS‐0327 prevented the effects induced by repeated AKB48, confirming a CB 1 R‐mediated action of the drug. Blood AKB48 levels were higher in females than in males and repeated administration caused a progressive rise of AKB48 content in blood samples of both sexes, suggesting an inhibitory effect on cytochrome activity. Finally, immunohistochemical analysis revealed higher expression of CB 1 Rs in the cerebellum and cortex of females, and a rapid CB 1 R downregulation in cerebellar and cortical areas following repeated AKB48 injections, with neuroadaptation occurring generally more rapidly in females than in males. Conclusion and Implications We showed for the first time that AKB48 effects significantly vary with prolonged use and that sex affects the pharmacodynamic/pharmacokinetic responses to its repeated administration, suggesting a sex‐tailored approach in managing AKB48‐induced intoxication.
Article
Full-text available
Cannabinoid receptors (nomenclature as agreed by the NC-IUPHAR Subcommittee on Cannabinoid Receptors [119]) are activated by endogenous ligands that include N-arachidonoylethanolamine (anandamide), N-homo-γ-linolenoylethanolamine, N-docosatetra-7,10,13,16-enoylethanolamine and 2-arachidonoylglycerol. Potency determinations of endogenous agonists at these receptors are complicated by the possibility of differential susceptibility of endogenous ligands to enzymatic conversion [5].There are currently three licenced cannabinoid medicines each of which contains a compound that can activate CB1 and CB2 receptors [111]. Two of these medicines were developed to suppress nausea and vomiting produced by chemotherapy. These are nabilone (Cesamet®), a synthetic CB1/CB2 receptor agonist, and synthetic Δ9-tetrahydrocannabinol (Marinol®; dronabinol), which can also be used as an appetite stimulant. The third medicine, Sativex®, contains mainly Δ9-tetrahydrocannabinol and cannabidiol, both extracted from cannabis, and is used to treat multiple sclerosis and cancer pain.
Article
Full-text available
Heterocycles form by far the largest of classical divisions of organic chemistry and are of immense importance biologically and industrially, and about one half of over six million compounds recorded in chemical abstracts are heterocyclic. The majority of pharmaceuticals and biologically active agrochemicals are heterocyclic while countless additives and modifiers used in industrial applications ranging from cosmetics, reprography, information storage and plastics are heterocyclic in nature. One striking structural feature inherent to heterocycles, which continue to be exploited to great advantage by the drug industry, lies in their ability to manifest substituents around a core scaffold in defined three-dimensional representations. Among the approximately 20 million chemical compounds identified by the end of the second millennium, more than two-thirds are fully or partially aromatic and approximately half are heterocyclic. The presence of heterocycles in all kinds of organic compounds of interest in electronics, biology, optics, pharmacology, material sciences and so on is very well known. Between them, nitrogen-containing heterocyclic compounds have maintained the interest of researchers through decades of historical development of organic synthesis. In the present work, focus on the literature survey of chemical diversity (piperidine 1, pyridine 2, quinoline 3, [1,3,4]-thiadiazole 4, pyrazole 5 and [1,2,4]-triazole 6) in the molecular framework in order to get a complete information regarding pharmacologically interesting compounds of widely different composition.
Article
Full-text available
ABSTRACT Agonists stimulate guanylyl 5'- [y-[35S] thiol-triphosphate (GTP[y-35S]) binding to receptor-coupled guanine nucleotide binding protein (G proteins) in cell membranes as revealed in the presence of excess GDP. We now report that this reaction can be used to neuroanatomically localize receptor-activated G proteins in brain sections by in vitro autoradiography of GTP[y-35S] binding. Using the mu opioid-selective peptide [D-Ala2,N-MePhe4,Gly5-ol]enkephalin (DAMGO) as an agonist in rat brain sections and isolated thalamic membranes, agonist stimulation of GTP[y-35S] binding required the presence of excess GDP (1-2 mM GDP in sections vs. 10-30 µM GDP in membranes) to decrease basal G-protein activity and reveal agonist-stimulated GTP[y-35S] binding. Similar concentrations ofDAMGO were required to stimulate GTP[y-35S] binding in sections and membranes. To demonstrate the general applicability of the technique, agonist-stimulated GTP[v-35S] binding in tissue sections was assessed with agonists for the mu opioid (DAMGO), cannabinoid (WIN 55212-2), and y-aminobutyric acid type B (baclofen) receptors. For opioid and cannabinoid receptors, agonist stimulation of GTP[y-35S] binding was blocked by incubation with agonists in the presence of the appropriate antagonists (naloxone for mu opioid and SR-141716A for cannabinoid), thus demonstrating that the effect was specifically receptor mediated. The anatomical distribution of agonist-stimulated GTP[y-35S] binding qualitatively paralleled receptor distribution as determined by receptor binding autoradiography. However, quantitative differences suggest that variations in coupling efficiency may exist between different receptors in various brain regions. This technique provides a method of functional neuroanatomy that identifies changes in the activation of G proteins by specific receptors.
Article
Full-text available
(-)-Δ⁹-Tetrahydrocannabinol ((-)-Δ⁹-THC) is the major active psychotropic component of the marijuana plant, Cannabis sativa. The membrane proteins that have been found to bind this material or its derivatives have been called the cannabinoid receptors. Two GTP-binding protein-coupled cannabinoid receptors have been cloned. CB1 or the neuronal cannabinoid receptor is found mostly in neuronal cells and tissues while CB2 or the peripheral cannabinoid receptor has been detected in spleen and in several cells of the immune system. It has previously been shown that activation of CB1 or CB2 receptors by cannabinoid agonists inhibits adenylyl cyclase activity. Utilizing Chinese hamster ovary cells and COS cells transfected with the cannabinoid receptors we report that(-)-Δ⁹-THC binds to both receptors with similar affinity. However, in contrast to its capacity to serve as an agonist for the CB1 receptor, (-)-Δ⁹-THC was only able to induce a very slight inhibition of adenylyl cyclase at the CB2 receptor. Morever, (-)-Δ⁹-THC antagonizes the agonist-induced inhibition of adenylyl cyclase mediated by CB2. Therefore, we conclude that (-)-Δ⁹-THC constitutes a weak antagonist for the CB2 receptor.
Chapter
The major progress in controlling chemotherapy-induced emesis benefits thousands of patients each day. Antiemetic efficacy is found with agents of many different classes, including phenothiazines, butyrophenones, cannabinoids, corticosteroids, substituted benzamides, and serotonin receptor antagonists. To date, the most effective antiemetic regimens are combinations of serotoninreceptor antagonists with corticosteroids. Most antiemetic studies with cannabinoids have methodological difficulties and can be difficult to interpret. If one concentrates on the better-conducted trials, however, it is clear that cannabinoids possess antiemetic properties in patients receiving cancer chemotherapy. The degree of antiemetic activity demonstrated by cannabinoids is not as high as that seen with several other classes of agents. The side effects associated with cannabinoid use are tolerable, but greater that those seen with other classes of agents. Studies have not demonstrated an advantage of one tested cannabinoid over another. Results of trials with synthetic cannabinoids, such as levonantradol or nabilone, do not indicate a superior therapeutic index over naturally occurring cannabinoids. Only limited data from well-designed trials can be found comparing inhalant marihuana with other cannabinoids. From that which exists, there is no clear advantage for either inhalant marihuana or for oral THC. Additionally, there is no demonstration that the inhalant agent results in an improved pharmacokinetic profile, an advantage in self titration, or in a different pattern of side effects. Further studies with cannabinoids, using accepted methodology, could more accurately outline their activity. Decisions should be made whether or not the current moderate degree of efficacy and moderate amount of associated side effects with all tested cannabinoid antiemetics warrant such trials in the context of substantially more active agents that have more favorable toxicity profiles.
Article
The use of crude marijuana for herbal medicinal applications is now being widely discussed in both the medical and lay literature.Ballot initiatives in California and Arizona have recently made crude marijuana accessible to patients under certain circumstances. As medicinal applications of pure forms of delta-9-tetrahydrocannabinol (THC) and crude marijuana are being considered, the most promising uses of any form of THC are to counteract the nausea associated with cancer chemotherapy and to stimulate appetite. We evaluated the relevant research published between 1975 and 1996 on the medical applications, physical complications, and legal precedents for the use of pure THC or crude marijuana.Our review focused on the medical use of THC derivatives for nausea associated with cancer chemotherapy, glaucoma, stimulation of appetite, and spinal cord spasticity. Despite the toxicity of THC delivered in any form, evidence supports the selective use of pure THC preparations to treat nausea associated with cancer chemotherapy and to stimulate appetite. The evidence does not support the reclassification of crude marijuana as a prescribable medicine.
Article
Various applications of pAx measurements are discussed based on the hypothesis that drugs and drug antagonists compete for receptors according to the mass law. Examples are given illustrating the use of pAx measurements to identify agonists which act on the same receptors and to compare the receptors of different tissues. Tests of competitive and noncompetitive antagonism are considered in relation to the antagonisms acetylcholine-atropine, histamine-atropine and acetylcholine-cinchonidine. A new measure, pAh, is introduced to express the activity of unsurmountable antagonists.
Article
Recent work contributing towards an understanding of the mechanism of action of cannabis and its constituents is described and the known therapeutic and pharmacological activities of these substances reviewed, together with synthetic drugs derived from tetrahydrocannabinol (Δ1-THC). The medical uses discussed include the actions of cannabinoids in the treatment of glaucoma, asthma, emesis, hyperthermia, convulsion, muscle spasticity, anxiety, hypertension, pain and inflammation.
Article
JMM and LAB). Key Words: Cannabinoid agonists—Cannabinoid antagonists—Cannabinoid receptors— CB 1 —CB 2 —∆ 9 -THC (∆ 9 -Tetrahydrocannabinol)—Marijuana—SR 141716A [N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydro-chloride].
Article
A theoretical analysis has been made of the relationship between the inhibition constant (KI) of a substance and the (I50) value which expresses the concentration of inhibitor required to produce 50 per cent inhibition of an enzymic reaction at a specific substrate concentration. A comparison has been made of the relationships between KI and I50 for monosubstrate reactions when noncompetitive or uncompetitive inhibition kinetics apply, as well as for bisubstrate reactions under conditions of competitive, noncompetitive and uncompetitive inhibition kinetics. Precautions have been indicated against the indiscriminate use of I50 values in agreement with the admonitions previously described in the literature. The analysis described shows KI does not equal I50 when competitive inhibition kinetics apply; however, KI is equal to I50 under conditions of either noncompetitive or uncompetitive kinetics.