Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Dec 5.
Published in final edited form as: Eur J Pharmacol. 2023 Oct 29;960:176168. doi: 10.1016/j.ejphar.2023.176168

Effects of cannabinoid agonists and antagonists in male rats discriminating the synthetic cannabinoid AM2201

Dalal AlKhelb 1,2, Emily L Burke 1, Alexander Zvonok 3, Christos Iliopoulos-Tsoutsouvas 1, Markos-Orestis Georgiadis 1, Shan Jiang 1, Thanh C Ho 1, Spyros P Nikas 1, Alexandros Makriyannis 1,3, Rajeev I Desai 1,4
PMCID: PMC10704044  NIHMSID: NIHMS1944030  PMID: 38059442

Abstract

The synthetic forms of delta-9-tetrahydrocannabinol (Δ9-THC), dronabinol or nabilone, have been approved to treat several indications. However, due to safety concerns their clinical utility remains limited. Consequently, there is a need for developing cannabinoid (CB) ligands that display behavioral pharmacological profiles better than Δ9-THC. Here, we utilized drug discrimination methods to compare the interoceptive effects of CB ligands that vary in potency, efficacy, and selectivity at the CB receptors, including two ligands, AM411 and AM4089, that show CB1 partial agonist-like actions in vitro. Male rats were trained to discriminate 0.1 mg/kg AM2201 from saline under a fixed-ratio (FR) 10 response schedule of food reinforcement. After establishing AM2201’s discriminative-stimulus effects, pretreatment tests with the CB1 antagonist/inverse agonist rimonabant blocked AM2201’s effects, whereas the peripherally-restricted antagonist AM6545 had no effect. Next, the generalization profiles of AM411 and AM4089 with CB1 full (JWH-018, CP-55,940, AM8936), partial agonist (Δ9-THC) and non-cannabinoids (fentanyl, atropine) were compared. The CBs either fully (AM2201, CP-55,940, JWH-018, AM8936, Δ9-THC) or partially (AM411, AM4089) substituted for AM2201, whereas fentanyl and atropine did not produce AM2201-like effects. All CB drugs were more potent than Δ9-THC and correlation analysis confirmed the relative behavioral potencies of CBs corresponded strongly with their relative affinities at the CB1 but not CB2 receptors. Together, our results further demonstrate that AM411 and AM4089 exhibit a better pharmacological profile compared to Δ9-THC, in that, they are more potent and display in vivo partial agonist-like actions that are centrally mediated via CB1 receptors.

Keywords: Cannabinoid Agonists and Antagonists, Drug Discrimination, Cannabinoid Receptors, AM2201 Discrimination, Δ9-THC, Partial Agonists

1. Introduction

Increasing evidence suggests that the cannabinoid (CB) G-protein-coupled CB1 and CB2 receptors play an important role in a variety of biological functions (e.g., Lowe et al 2021). Consequently, considerable research has focused on developing drugs that target CB receptors to either modulate various physiological functions or as potential treatments for pathological conditions (e.g., Schurman et al. 2020; Finn et al. 2021; Lowe et al 2021). Currently, dronabinol or nabilone, the synthetic forms of delta-9-tetrahydrocannabinol (Δ9-THC) have been approved to treat nausea and vomiting following chemotherapy or as an appetite stimulant for AIDS-related anorexia (Coronado-Álvarez et al. 2021). Moreover, both Δ9-THC (combined with the noradrenergic agonist lofexidine) and nabilone have produced favorable results in cannabis withdrawal studies in humans (Haney et al., 2004), strongly supporting the potential therapeutic utility of partial CB1 agonist-based medications for managing cannabis use disorders. Unfortunately, their clinical utility remains limited due to safety concerns related to the high lipophilic properties of Δ9-THC causing unpredictable pharmacokinetics, the production of active metabolites (Grotenhermen, 2003; Klumpers et al., 2012; Huestis 2007), and unwanted physiological/subjective effects that overlap with those of smoked cannabis (Hart et al., 2002; Sachs et al. 2015).

Additionally, Δ9-THC has a relatively low potency at CB receptors, thus higher concentration or repeated administration is needed to achieve a desirable therapeutic effect, which results in psychoactive and/or toxic effects (Coronado-Álvarez et al. 2021; Sachs et al. 2015). While in vitro studies have shown evidence of Δ9-THC’s partial CB1 agonist activity (e.g., Dutta et al. 2022; Sim et al. 1996), such effects have not been consistently reported in in vivo laboratory animal studies. Unlike CB1 full agonists, Δ9-THC only partially reduces body temperature in mice (McMahon and Koek 2007; Tai et al. 2015) and, in pretreatment studies, blocks the hypothermic effects of the full CB1 agonist AM2389 in mice (Paronis et al. 2012). Studies have also suggested that a greater degree of pharmacological tolerance with Δ9-THC compared to the full CB1 agonists may reflect the partial agonist-like effects of Δ9-THC (Singh et al. 2011, Hruba et al. 2012; Desai et al., 2013). However, in other studies Δ9-THC produces antinociceptive, cataleptic, and locomotor suppressant effects that are comparable to the bicyclic, cyclohexylphenol, aminoalkylindol, and naphthoylindoles full CB1 agonists (e.g., Compton et al. 1992; Fan et al. 1994; Ginsburg et al. 2012; McMahon and Koek 2007; Wang et al. 2020). Moreover, in drug discrimination studies, full CB1 agonists substitute for Δ9-THC’s discriminative-stimulus effects in rats or monkeys and in synthetic CB1 agonists-trained subjects, Δ9-THC and other full CB1 agonists generalize for their stimulus effects (e.g., Gatch and Forster 2014; Ginsburg et al. 2012; Hruba and McMahon 2017; Järbe et al. 2014; 2016a; 2011a; Wiley et al. 2012; 2014; Kangas et al. 2013; Rodriguez and McMahon 2014). Collectively, such observations have hindered our ability to identify CB1-based pharmacotherapies with partial agonist-like activity.

We have previously synthesized and evaluated the structure activity relationship of a series of the 3-(1-adamantyl) substituted CB compounds that have a relatively similar chemical structure to Δ9-THC (Thakur et al., 2013). Unlike the full CB1 agonist AM4054, two CBs, AM411 and AM4089, displayed promising in vitro (cyclic adenosine monophosphate assay cAMP) and in vivo (tetrad assays of antinociception and hypothermia) evidence of CB1 partial agonists-like activity (Thakur et al. 2013). The present studies were conducted to further characterize the partial CB1-agonist-like behavioral effects of AM411 and AM4089 in rats trained to discriminate the full synthetic CB1 agonist AM2201. First, pretreatment studies with rimonabant (inverse agonist/antagonist) and AM6545 (peripherally-restricted) CB1 antagonists were conducted to establish that AM2201’s discriminative-stimulus effects are centrally-mediated by CB1 receptors. Next, to confirm that AM411 and AM4089 produce partial CB1 agonist-like behavioral effects, we compared their substitution profiles with other standard CB (JWH-018, CP-55,940, AM8936, Δ9-THC) and non-cannabinoid (fentanyl and atropine) drugs. Finally, to further show that the discriminative-stimulus effects of CBs are mediated mainly through CB1 receptors, correlation analysis was conducted to examine the correspondence between the behavioral effects of CBs in the present experiments and their affinities for the different CB receptors.

2. Materials and Methods:

2.1.1. Subjects.

Eight experimentally naive adult male Sprague-Dawley rats (Charles River Laboratories, Inc., Wilmington MA) weighing 275–350g were used. All rats were group-housed in a temperature and humidity-controlled room with a 12-h light/dark cycle (lights on 7:00 AM). Discrimination experiments were conducted five days/week during the light phase of the light/dark cycle between 9:00 AM and 5:00 PM. Rats were maintained at 85% of the free-feeding weight and were fed standard laboratory chows (~15 g) one hour after the behavioral session. Water was freely accessible except during experimental sessions. Rats were maintained per guidelines provided by the Guide for Care and Use of Laboratory Animals (National Academy of Sciences, 2011). Drug discrimination protocol (no. 20-0519R) was approved by the Institutional Animal Care and Use Committee at Northeastern University, Boston, MA, USA.

2.1.2. Apparatus.

Standard two-lever operant conditioning chambers that contain light and sound attenuating enclosures and supplied with continuous white noise and a fan to mask external noise were used (Med Associates, St. Albans, VT). The operant chamber was equipped with a feeder, and a food delivery tray centered on the front wall in between the two response levers set 17 cm apart in each chamber. A downward force of ~ 0.4 N was required to produce an audible click in the chamber and recorded as a response. Light-emitting diodes (LEDs) located directly above the response levers served as stimulus lights, and a house light located on the back panel of the chamber provided ambient illumination. During the behavioral sessions, experimental variables and data collection were controlled by computers with Med Associate interfacing equipment and operating software (Med Associates, St. Albans, VT).

2.1.3. AM2201 Discrimination.

The training procedure used to establish AM2201 discrimination has been described previously (Desai et al., 2003; Desai and Bergman 2010). Subjects were first trained to press both response levers under the fixed-ratio (FR) 10 response schedule of food reinforcement. Completing the FR-10 response requirement resulted in the delivery of one 45-mg pellet (BioServe, Frenchtown, NJ) into the food tray. Subsequently, subjects were trained to discriminate intraperitoneal (i.p.) injections of 0.1 mg/kg AM2201 from i.p. injections of saline. After AM2201 injection, responses only on one lever were reinforced; after saline injection, responses only on the other lever were reinforced. The assignment of AM2201-associated and saline-associated levers were counterbalanced across rats. Training sessions with AM2201 and saline were conducted five days per week and occurred in a double alternation sequence (i.e., saline, saline, 0.1 mg/kg AM2201, 0.1 mg/kg AM2201, saline, …etc.). Each experimental session was started by placing the rat inside the chamber immediately after the i.p. injection, all lights were extinguished, and responding had no scheduled consequences. After a 10-min timeout (TO), the session started and all lights in the experimental chamber were illuminated. Only completion of the FR-10 response requirement on the injection-associated (correct) lever was reinforced; responses on the other lever (incorrect) reset the lever response for food delivery. The presentation of each food pellet was followed by a 20-s TO period during which all lights were off, and lever responses had no scheduled consequences. Sessions ended after each rat obtained 20-reinforcements or after 15-min, whichever occurred first. Testing was initiated when the overall and the first FR-10 (1st FR) injection-associated responding criteria of at least 80% during drug sessions and 20% or less during saline sessions were met over four successive training sessions. After subjects met this initial criterion, test sessions were only conducted over this course of these studies whenever rats met the above criteria for two consecutive training days in the sequence saline/0.1 mg/kg AM2201 or 0.1 mg/kg AM2201/saline. To determine the time course of action, the discriminative-stimulus effects of 0.1 mg/kg AM2201 were determined in single test session over multiple test days at 10, 30, 120, 360 min and 24 hours after drug administration. Following initial AM2201 dose-response and time course determinations, doses of each drug were tested in a random order in all subjects. Test sessions lasted 25-min (10-min timeout plus 15-min test session), during which the FR-10 schedule was in effect, and a single dose of each test drug was administered at the start of the 25-min cycle. The CB1 antagonist’s interaction studies were conducted by administering 1.0 or 3.2 mg/kg of rimonabant or AM6545 20-min before injections of the training dose of 0.1 mg/kg AM2201. Doses of each antagonist were selected based on previous studies conducted to evaluate the effects of these CB1 antagonists in rodents (e.g., Järbe et al. 2011b, Järbe et al. 2016a). All schedule parameters and contingencies during test sessions were identical to the training sessions, except that the 10 consecutive responses on either lever resulted in the delivery of a food reinforcer.

2.1.4. Drugs.

AM2201, JWH-018, AM8936, AM411, AM4089, rimonabant (SR141716A), and AM6545 were synthesized for these studies in the Center for Drug Discovery at Northeastern University, Boston, MA. Δ9-THC, CP-55,940, and fentanyl were provided by the National Institution of Drug Abuse drug supply program (Division of Therapeutics and Medical Consequences, NIDA). Atropine sulphate was purchased from Sigma-Aldrich (St. Louis, MO). AM2201, JWH-018, AM411, AM4089, and CP-55,940 were prepared in a 20:20:30:30 mixture of 95% ethanol, 100% dimethyl sulfoxide (DMSO), Alkamuls (EL-620), and saline. Δ9-THC and rimonabant were prepared in a 20:20:60 mixture of 95% ethanol, Tween-80, and saline. Fentanyl and atropine were dissolved in 0.9% saline. All drugs were administered via i.p. injections in volumes of 1.0 ml/kg or less. All tested drugs were given 30-minutes before the session except for AM2201, fentanyl and atropine which was given 10-minutes before the start of the session.

2.1.5. Data Analysis.

The percentage of AM2201-associated responses was calculated by dividing the number of responses on the drug lever by the total number of responses on both levers. The overall response rates were calculated for each session by dividing the total number of responses by the session duration without the timeout periods. Data for any rat that failed to emit at least 10 responses on either lever was excluded from the calculation of mean drug-appropriate responding at that dose. If fewer than three of the rats met the response rate requirement at a particular dose, no mean value was calculated for percentage of drug appropriate responding, but the subject’s data were included in the group mean for overall response rate analysis (Desai et al., 2003; Desai and Bergman 2010; Katz et al., 2004; Li et al., 2011; Wiley et al. 1998; 2014) Mean results for saline and each drug dose were calculated by averaging data from all subjects. Complete, partial, and no substitution for 0.1 mg/kg AM2201 was defined as ≥ 80%, 21%–79%, and ≤ 20% of total responses on the drug-associated lever, respectively. Repeated measures oneway ANOVA followed by Dunnett’s test was used to determine differences in overall response rates from saline values and the student’s paired t-test was used to determine the significant differences in responses in the antagonist studies. The functional in vivo half-life of AM2201 and its 95% confidence limits were determined using nonlinear regression (GraphPad Prism, version 9.1.3). The dose of drug needed to produce 50% AM2201-associated responding (ED50 values and their 95% confidence limits) was determined using the linear portion of the dose-response curve (GraphPad Prism, version 9.1.3). The behavioral relative potency of CBs was determined using the ED50 shift equation to evaluate the potency difference relative to AM2201 ED50 (GraphPad Prism, version 9.1.3).

Correlation between the effects of drugs on behavior and competitive receptor binding (in vitro Ki values) studies were examined by comparing the calculated behavioral relative potency of each CB and their previously published relative in-vitro affinities for binding CB1 and CB2 receptors, i.e., Ki values of the tested drug divided by the Ki value for AM2201 alone (Table 1). Cannabinoid CB1 receptor affinity values were obtained from previously published radioligand binding experiments in rat brains, whereas affinity values for cannabinoid CB2 receptors were obtained from radioligand binding experiments in human CB2 receptors due to the insufficient published data from rodents CB2 receptors. Where appropriate, affinity values from multiple studies were averaged. Pearson’s product-moment correlation coefficient of these values was calculated using the correlation equation (GraphPad Prism, version 9.1.3) to determine the relationship between relative behavioral potency and relative binding affinity at cannabinoid CB1 and CB2 receptors.

Table.1:

ED50 values (95% C.I) and relative behavioral potencies with which CBs produce increases in AM2201-associated responding in rats [AM2201 = 1] and their relative affinity at CB1 and CB2 receptors [AM2201 = 1].

Cannabinoid Ligand %Drug-associated responding doses %Drug-associated responding ED50
(95% C.I)
Relative Behavioral Potency
(95% C.I)
rCB1 in-vitro affinity
(Ki value)
Relative Affinity at CB1 hCB2 in-vitro affinity
(Ki value)
Relative Affinity at CB2

ms/kg ms/kg ED50 ratio nM nM
AM2201 (Start) 0.0032- 0.1 0.031
(0.017- 0.057)
1.0 1.0 a) 1.0 25 a) 1.0
AM2201 (End) 0.0032- 0.1 0.035
(0.02-0.06)
1.2
(0.7-1.8)
1.0 a) 1.0 25 a) 1.0
CP-55,940 0.0032- 0.32 0.042
(0.021- 0.084)
1.38
(0.68- 2.78)
0.97 b) 0.97 0.69h) 0.02
JWH-018 0.01- 0.56 0.11
(0.05- 0.24)
3.47
(1.76- 6.81)
4.5 c) 4.5 2.9 h) 0.12
AM8936 0.001- 0.1 0.012
(0.006- 0.025)
0.41
(0.2- 0.81)
0.55 d) 0.55 0.55 # 0.022
Δ9-THC 0.1- 5.6 0.95
(0.5- 1.8)
25.58
(12.46- 50.47)
39.5 e) 39.5 36.4 h) 1.45
AM411 * 0.032- 1.0 0.15
(0.04- 0.51)
10.52
(7.07- 13.98)
6.8 f) 6.8 52 f) 2.08
AM4089 * 0.0032- 0.1 0.016
(0.004- 0.06)
1.04
(0.74- 1.34)
2.1 g) 2.1 46.7 g) 1.7

3. Results

3.1.1. AM2201 discrimination.

All the subjects learned to discriminate between i.p. injections of 0.1 mg/kg AM2201 from i.p. injections of saline within approximately 80 (SD=18.67) training sessions. Subjects started to acquire AM2201 discrimination at week five and control sessions after week seven show that injections of the training dose of 0.1 mg/kg AM2201 maintained an average of >80% responding on the drug-associated lever, whereas saline injections produced <20% responding on the drug-associated lever (Fig. 1, top panel). Administration of the training dose of AM2201 did not substantively alter response rates, with <10% difference in responding compared to saline values between sessions (Fig. 1, bottom panel). The mean group averages for rates of food-maintained responding during AM2201 and saline injections did not substantially differ during discrimination training sessions, i.e., 1.15 (± 0.14 S.E.M.) vs 1.27 (± 0.11 S.E.M.) responses per second after week 8, respectively (Fig. 1, bottom panel). Discriminative-stimulus control by the training dose of 0.1 mg/kg AM2201 was maintained throughout the study.

Figure 1.

Figure 1

Acquisition of discriminative-stimulus control in a group of male Sprague-Dawley rats trained to discriminate 0.1 mg/kg (i.p.) AM2201 from saline. Abscissae: Average of 5 training sessions from the first drug or saline injection. Ordinates: percentage of AM2201-associated responding (top) and response rates (bottom). Each data point represents the average (± S.E.M.) of effects in all subjects tested (at least six rats) at each dose. The horizontal dashed line at 80% and 20% drug lever responding (top) indicates the criterion for evidence of discriminative-stimulus control.

Injections of AM2201 (0.0032–0.1mg/kg) at the start and end of the study produced similar dose-dependent and full substitution for the training dose of 0.1 mg/kg AM2201, with a maximum of 86% (±11.9 S.E.M.) and 89% (±7.60 S.E.M.) responding on the drug associated-lever after administration of 0.1 mg/kg of AM2201 (Fig. 2, top left panel). Overall response rates were not significantly altered relative to control values following administration of any of the doses of AM2201 at the start and end of the study (Start: F(4, 35) = 2.29, P > 0.05; End: F(4, 35) = 1.97, P > 0.05; Fig. 2, bottom left panel). Table 1 shows that the calculated ED50 the AM2201-associated lever responding at the start and end of the study did not substantially differ indicating no change in substitution potency of AM2201 over the course of the study (Start: 0.031 mg/kg; 95% C.I.: 0.017–0.057; End: 0.035 mg/kg; 95% C.I.: 0.02–0.06).

Figure 2.

Figure 2.

Effects of the full cannabinoid agonist AM2201 determined at the start and end of the study in a group of male Sprague-Dawley rats trained to discriminate 0.1 mg/kg AM2201 from saline. Ordinates: percentage of AM2201-associated responding (top-right) and response rates (bottom-right) following i.p. injections of AM2201 or vehicle. The time course of action (top-left) and the response rates (bottom-left) of AM2201 training dose. Abscissae: S = saline; AM = AM2201 training dose in mg/kg (log scale). Data reported under S and AM are the average of 10 saline or 0.1 mg/kg AM2201 training session that were conducted before, during, and after testing of one of drugs in each panel. Each data point represents the average (± S.E.M.) in all subjects tested (at least six rats) at each dose or time point. The horizontal dashed line at 80% and 20% drug lever responding (top) indicates the criterion for evidence of discriminative-stimulus control.

The time course data shows that the discriminative-stimulus effects of the training dose of 0.1 mg/kg AM2201 was fully captured within 24-hrs with a maximum response of 89.04% on the AM2201-associated lever 10-min after treatment, ~45% from 30-min to 6-hrs, and <10% at 24-hrs post-treatment (F(4, 32) = 5.089, P < 0.05; Fig. 2, top right panel). The estimated functional in vivo half-life of 0.1 mg/kg AM2201 was 111.4 mins (95% C.I.: 61.9 −205.8; Fig. 2, top right panel). The time course of the response rates produced by 0.1 mg/kg AM2201 did not significantly decrease or increase over the course of the 24-hr assessment (F(4, 30) = 2.04, P > 0.05; Fig. 2, bottom right panel).

3.1.2. Pretreatment with CB antagonists.

Pretreatment with rimonabant (1.0 or 3.2 mg/kg) significantly blocked the discriminative-stimulus effects of 0.1 mg/kg AM2201 in a dose-dependent manner with a maximal reduction in AM2201-associated responding from ~90% to ~56 (t(14)= 2.26; P < 0.05) and 33% (t(14)= 3.78; P < 0.05), respectively (Fig. 3, top right panel). Both doses of rimonabant (1.0 or 3.2 mg/kg) did not significantly alter response rates (t(14)= 0.57 and t(14)= 0.701; P > 0.05, respectively; Fig. 3 bottom right panel). In contrast to rimonabant, pretreatment with AM6545 (1.0 or 3.2 mg/kg) did not block the discriminative-stimulus effects of training dose of 0.1 mg/kg AM2201 (t(12)= 0.60 and t(12)= 0.67; P > 0.05, respectively; Fig. 3, top left panel) or alter rates of food-maintained responding (t(12)= 0.80 and t(12)= 1.25, respectively; P > 0.05; Fig. 3, bottom left).

Figure 3.

Figure 3

Effects of pretreatment with CB1 antagonists in a group of male Sprague-Dawley rats trained to discriminate 0.1 mg/kg AM2201 from saline. Ordinates: the percentage of AM2201-associated responding (top) and response rates (bottom) 30 mins following injections of rimonabant (left) or AM6545 (right) and the training dose of AM2201. Abscissae: Each bar graph represents the average (± S.E.M.) in all subjects tested (at least six rats) at each dose. The horizontal dashed line at 80% drug lever responding (top) indicates the criterion for evidence of discriminative-stimulus control. See Figure 2 caption for other details.

3.1.3. Effect of CB1 full and partial agonists.

The CB1 agonists AM2201 (0.0032–0.1 mg/kg; Fig. 2), CP-55,940 (0.0032–0.32 mg/kg), JWH-018 (0.01–0.56 mg/kg), and AM8936 (0.001–0.1mg/kg) (Fig. 4, top left panel) produced dose-dependent and full substitution for the training doses of 0.1 mg/kg AM2201, with maxima of approximately 86, 81, 84 and 86% AM2201-associated lever responding, respectively. There was no significant change in the overall response rates after administering JWH-018 (F(6,41)= 1.44, P > 0.05), CP-55,940 (F(6,41)= 1.22, P > 0.05) and AM8936 (F(6,41)= 2.49, P > 0.05) compared to saline control values (Fig. 4, bottom left panel). The phytocannabinoid Δ9-THC (0.1–5.6 mg/kg) also fully substituted for the training dose of 0.1 mg/kg AM2201, with maxima of 97% responding on AM2201 associated-lever after administration of 5.6 mg/kg Δ9-THC (Fig. 4, top middle panel). Δ9-THC did not substantially alter overall rates of food-maintained responding (F(6,48)= 1.76, P > 0.05; Fig. 4, bottom middle panel). In contrast, the partial CB1 agonists AM411 (0.032–3.2 mg/kg) and AM4089 (0.0032–1.0 mg/kg) partially substituted for 0.1 mg/kg AM2201’s discriminative-stimulus effects as indicated by a plateau in the dose-response function and a maximal effect of approximately 55 and 50% on AM2201-associated lever after administration of 1.0–3.2 mg/kg AM411 and 0.1–1.0 mg/kg AM4089, respectively (Fig. 4, top middle panel). None of these partial agonists, AM411 or AM4089 showed a significant change in the overall response rates (F(6,48)= 1.76, F(5,40)= 1.62 and (F(6,43)= 0.62, P > 0.05; Fig. 4, bottom middle panel). The rank order of potency for CB1 ligands based on the calculated ED50 values was: AM8936 > AM2201 ≈ AM4089 ≈ CP-55,940 > JWH-018 > AM411 > Δ9-THC (Table 1; Fig. 5).

Figure 4.

Figure 4

Effects of full CB agonists: CP-55,940, JWH-018 and AM8936 (left), partial CB agonists: Δ9-THC, AM411, and AM4089 (middle) and non-CB ligands: fentanyl and atropine (right) determined in a group of male Sprague-Dawley rats trained to discriminate 0.1 mg/kg AM2201 from saline. Ordinates: the percentage of AM2201-associated responding (top) and response rates (bottom) 30-min following injections of CB1 agonists or 10-min following non-CB ligands. Abscissae: S = saline; AM = AM2201 training dose in mg/kg (log scale). Each data point represents the average (± S.E.M.) in all subjects tested (at least four rats) at each dose. The horizontal dashed line at 80% and 20% drug lever responding (top) indicates the criterion for evidence of discriminative-stimulus control. See Figure 2 caption for other details.

Figure 5.

Figure 5

The calculated ED50 values of the cannabinoid ligands: AM2201, JWH-018, CP-55,940, AM8936, AM4089, AM411 and Δ9-THC and their 95% confidence limits (C.I). Ordinates: ED50 values (mg/kg). Abscissae: Each bar graph represents the averaged ED50 from each ligand with their upper and lower limits (95% C.I) calculated from all the tested subjects. Each point within the bar graph represents the ED50 from a single subject (at least six rats). See Figure 2 caption for other details.

3.1.4. Effects of Non-cannabinoid Drugs.

The non-cannabinoid drugs fentanyl (0.0032–0.032 mg/kg) and atropine (0.032-1.0 mg/kg) did not generalize for the discriminative-stimulus effects of 0.1 mg/kg AM2201 at any of the tested doses (Fig. 4, top right panel). Doses of fentanyl tested in this study did not alter rates of food-maintained responding (F(4, 25)= 0.57, P > 0.05), whereas atropine produced dose-dependent decreases in response rates (F(5, 33)= 2.21, P < 0.05; Fig. 4, bottom right panel).

3.1.5. Pearson’s product-moment correlation coefficient.

The comparison between relative behavioral potencies of CB ligands that substituted for AM2201 with their previously published competitive receptor binding (in vitro Ki values) affinities at CB receptors are shown in Figure 6. Results show a strong association between the behavioral potencies and relative binding affinities (Ki values) at the CB1 receptors in rat brain tissue (r = 0.96, P < 0.005; Fig. 6, right panel; Table 1). In contrast, no strong association between the behavioral potencies and relative binding affinities (Ki values) from human CB2 receptors was found (r = 0.55, P > 0.05; Fig. 6, left panel; Table 1).

Figure 6.

Figure 6

Correlation between relative potencies of cannabinoid ligands (AM2201, AM8936, CP-55,940, JWH-018, Δ9-THC, AM411, and AM4089) in the present studies and their previously published relative affinities at CB1 and CB2 receptors in radioligand binding studies. Abscissae: affinity relative to AM2201 for inhibiting the binding of radioligand at rats CB1 (left panels) and human CB2 (right panels). Ordinates: behavioral potency relative to AM2201 based on ED50 values for producing discriminative-stimulus effects (Fig. 4; Table. 1).

4. Discussion

The present studies were conducted to further characterize the discriminative-stimulus effects of various CB ligands in rats trained to discriminate the highly potent full CB1 agonist AM2201. Results demonstrate that AM2201 serves as a robust discriminative-stimulus in male rats with no change in potency over the course of the study. The CB ligands (AM2201, CP-55,940, JWH-018, AM8936) and the phytocannabinoid Δ9-THC reliably and fully substituted for AM2201 in a dose-dependent manner without significantly altering the response rates. These findings are consistent with previous studies that have used other related CBs like JWH-018, AM5983, AM4054, and Δ9-THC as a discriminative-stimulus in rodents or nonhuman primates (Hruba and McMahon 2017; Järbe et al. 2016a; Järbe et al. 2016b; Järbe et al. 2012; Kangas et al. 2013; Wiley et al. 2014). Additionally, the time course data showed that intraperitoneal injections of the training dose of 0.1 mg/kg AM2201 displays a fast onset (>80 drug-associated responding at 10 min) and a short duration of in vivo action (<70 drug-associated responding after 60 min) with estimated functional in vivo half-life ~2.0 hrs. These findings are somewhat consistent with previous reports showing that AM2201 and other CBs like JWH-018 have a faster onset and shorter duration of action compared to Δ9-THC in Δ9-THC-trained rats (Gatch and Forster 2014; Järbe et al. 2011a; Järbe et al. 2016a) and produce effects on hypothermia, locomotor, and catalepsy that peak at ~2.0 hrs and lasts between 4.0-6.0 hrs after subcutaneous administration in rodents or monkeys (Carlier et al. 2018; Rodriguez and McMahon 2014). Moreover, the time course of AM2201’s effects are comparable with pharmacokinetic studies showing that administration of AM2201 produced dose-dependent effects with maximum plasma drug concentrations that last from 30 to120 mins in rats (Carlier et al. 2018).

Consistent with previous work in Δ9-THC or CB1 agonists trained animals, pretreatment with the selective CB1 inverse agonist/antagonists rimonabant blocked AM2201 discrimination (Ginsburg et al. 2012; Hruba and McMahon 2017; Järbe et al. 2016a; McMahon 2006; Wiley et al. 2014), whereas the peripherally restricted CB1 antagonist AM6545 had no effect. These data confirm that AM2201’s discriminative-stimulus effects are centrally mediated via the CB1 receptors. The lack of blockade by AM6545 is in line with the reported in vitro data showing this drug to function as a neutral CB1 antagonist with limited brain permeability relative to the neutral CB1 antagonist AM4113 and has rapid exertion from CNS via P-glycoprotein (Cluny et al. 2010; Tam et al. 2010). Likewise, other studies showed that unlike intracerebroventricular administration of CB1 antagonists, systemic administration of AM251, AM4113, or AM6545 reduced feeding behavior in a dose-dependent manner, suggesting that the pharmacological effects of these drugs are probably mediated by actions at peripheral CB1 receptors (Gómez et al. 2002; Randall et al. 2010; Sink et al. 2009). Also, intraperitoneal administration of AM6545 failed to generalize for rimonabants effects in rats further indicating that AM6545 is a peripherally restricted CB1 antagonist (Järbe et al. 2011b, but see Kangas et al., 2020).

The full generalization produced by the CB1 partial agonist Δ9-THC in AM2201-trained rats is consistent with previous studies in rats or non-human primates that have reported Δ9-THC to fully substitute for other higher efficacy CB1 agonists, including JWH-018, AM5983, and AM4054 (Järbe et al. 2014; Järbe et al. 2012; Kangas et al. 2013; Wiley et al. 2014). Other in vivo studies have also generally reported Δ9-THC to produce hypothermic, antinociceptive, and cataleptic effects that are comparable to the full efficacy CB1 agonists (Compton et al. 1992; Fan et al. 1994; Ginsburg et al. 2012; McMahon and Koek 2007; Wang et al. 2020). However, such observations are inconsistent with the in vitro characterization of Δ9-THC as a partial CB1 agonist (e.g., Burkey et al. 1997; Dutta et al. 2022; Sim et al. 1996). It is noteworthy that except for some studies showing Δ9-THC only partially reduces body temperature (McMahon and Koek 2007; Tai et al. 2015) and antagonizing the hypothermic effects of the high efficacy full CB1 agonist AM2389 in mice (Paronis et al. 2012), Δ9-THC’s CB1 partial agonist-like actions in animal studies has not been reliably demonstrated. Analysis of pharmacological tolerance was used to characterize the in vivo effects of high vs. low efficacy CB1 agonists by reporting a greater rightward shift on the dose-response curves after chronic administration of partial agonists (Desai et al. 2013; Paronis and Bergman 2011; Walker et al. 1995). Like these studies, Δ9-THC showed a greater tolerance compared to the higher efficacy CB1 agonists in nonhuman primates (Hruba et al. 2012). The disconnect between the in vivo and in vitro actions of Δ9-THC remains unclear but might be related to the lower amount of receptor occupancy required for Δ9-THC to produce full CB1-mediated discriminative-stimulus effects (Gifford et al. 1999). Also, the high lipophilic physicochemical properties of Δ9-THC likely lead to higher distribution and retention in the adipose tissue like the brain (e.g., Thomas et al. 1990), which might contribute to the observed full efficacy in the centrally mediated effects of Δ9-THC in drug discrimination studies.

The Δ8-THC analogue, AM411 partially substituted for AM2201. These data agree with previous reports suggesting that AM411 has CB1 partial agonist-like effects on antinociception and hypothermia (Thakur et al. 2013). Also, a greater degree of tolerance was observed in monkeys after chronic AM411 administration compared to the CB1 full agonists AM4054 and WIN 55212.2, suggesting that AM411 has a lower efficacy or partial effects at CB1 receptors (Desai et al. 2013). However, the above is somewhat inconsistent with the in vitro efficacy profile of AM411 indicating that it is a full CB1 agonist (Luk et al. 2004) and other studies showing that AM411 behaves as a full CB1 agonist in rats discriminating Δ9-THC (Lu et al. 2005) or in open field behaviors (Järbe et al. 2004). Differences in AM411’s effects in AM2201- versus Δ9-THC-trained subjects could be related to the difference in effectiveness of the training dose of Δ9-THC and AM2201 to serve as a discriminative-stimulus (Järbe et al. 2014), i.e., partial agonists like AM411 fully substitute in rats trained to discriminate a lower dose of AM2201. Also, it might be due to AM411’s effects on G protein-coupled inwardly rectifying potassium channels (GIRK). For example, Luk et al. (2004) suggests the role of desensitization rates in the overall efficacy of full CB1 agonists and demonstrates that AM411 has slower desensitization rates of GIRK channels like the partially effective agonist meth anandamide. Correspondingly, the 1-hydroxymethyl cannabinol analog AM4089 also partially substituted for AM2201’s discriminative-stimulus effects. This is consistent with its in vitro effects in the functional cAMP assay as well as hypothermia and antinociception studies in rodents (Thakur et al. 2013). To our knowledge, few studies have differentiated the in vivo behavioral pharmacology of CB1 agonists like AM411 and AM4089 to produce partial generalization for a highly potent and full efficacy CB1 agonist in discrimination studies. Evaluation of the ability of these drugs to antagonize the discriminative-stimulus effects of AM2201 or other full-efficacy CB1 agonists are needed to further confirm the in vivo partial CB1 agonist-like actions of these two drugs.

Previous work has suggested that the potency difference between CBs and the phytocannabinoid Δ9-THC might be a major contributing factor to the pronounced adverse effects of the psychoactive CBs. Here, the rank order of in vivo potencies of CB ligands versus Δ9-THC is consistent with the previously established in vitro CB1 Ki values (Hoffman et al. 2017; Khajehali et al. 2015; Thakur et al. 2005; Vigolo et al. 2015) showing that all the CBs (AM8936, AM4089, AM2201, CP-55,940, JWH-018, AM411) were ~ 80, 60, 30, 23, 9, and 6-fold more potent than Δ9-THC, respectively (Table 1; Fig. 5). Also, in keeping with previous reports (Järbe et al. 2011a), it is notable that the 95% confidence limits of the calculated ED50 value for Δ9-THC has a wider spread across individual subjects than the other CB1 ligands reflecting individual variability across subjects (Fig. 6). Studies have generally reported that the discriminative-stimulus effects of Δ9-THC and CB agonists are primarily mediated through actions at the CB1 receptors (e.g., Wiley et al. 2012); however, recent reports indicate that CB2 receptors are also expressed in the CNS and therefore might contribute to the behavioral effects of CBs and Δ9-THC, including their antinociceptive, locomotor, cataleptic, and abuse-related effects (e.g., Jordan and Xi 2019; Kibret et al. 2022; Wang et al., 2020). Consequently, we examined the association between the behavioral potencies and relative binding affinities of CB ligands at both CB1 and CB2 receptors by correlating the previously published CB receptors binding affinities (Ki values) from radioligand binding assays and the relative behavioral potencies from the present AM2201 discrimination studies. The comparison suggests a strong association between the behavioral potencies and relative binding affinities at the CB1 but not CB2 receptors further confirming that the full and partial generalization by CB drugs for AM2201 discrimination is due to activation of CB1 receptors.

5. Conclusion

Our results demonstrate that AM411 and AM4089 are more potent and display in vivo partial CB agonist-like actions that are centrally mediated via CB1 receptors and as such can be reliably used as pharmacological tools to distinguish between full and partial CB1 agonists.

Highlights.

  • CB1 agonists differentially substitute for AM2201’s discriminative stimulus effects

  • Behavioral potency and affinities at CB1 receptors strongly correlate

  • Unlike Δ9-THC, AM411/AM4089 display in vivo partial CB1 agonist-like actions

  • AM411/AM4089 can be used as tools to distinguish full vs partial CB1 agonists

ACKNOWLEDGMENTS

We would like to thank the National Institute of Drug Abuse drug supply program (Division of Therapeutics and Medical Consequences, NIDA) for providing Δ9-THC, and CP-55,940.

FUNDING

This research was partly supported by the National Institute on Drug Abuse grants R21DA045882 (AM-PI), P01DA009158 (AM-PI), and R37DA003801 (AM-PI). The authors have no other financial disclosures or potential conflicts of interest to declare.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

CONFLICTS OF INTEREST. The authors have no other financial disclosures or potential conflicts of interest to declare.

REFERENCES

  1. Burkey TH, Quock RM, Consroe P, Roeske WR, & Yamamura HI (1997). delta 9-Tetrahydrocannabinol is a partial agonist of cannabinoid receptors in mouse brain. European journal of pharmacology, 323(2-3), R3–R4. 10.1016/s0014-2999(97)00146-5 [DOI] [PubMed] [Google Scholar]
  2. Carlier J, Wohlfarth A, Salmeron BD, Scheidweiler KB, Huestis MA, & Baumann MH (2018). Pharmacodynamic Effects, Pharmacokinetics, and Metabolism of the Synthetic Cannabinoid AM-2201 in Male Rats. The Journal of pharmacology and experimental therapeutics, 367(3), 543–550. 10.1124/jpet.118.250530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Cluny NL, Vemuri VK, Chambers AP, Limebeer CL, Bedard EL, Wood JT, Lutz B, Zimmer A, Parker LA, Makriyannis A, & Sharkey KA (2010). A novel peripherally restricted cannabinoid receptor antagonist, AM6545, reduces food intake and body weight, but does not cause malaise, in rodents. British journal of pharmacology, 161(3), 629–642. 10.1111/j.1476-5381.2010.00908.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Compton DR, Gold LH, Ward SJ, Balster RL, & Martin BR (1992). Aminoalkylindole analogs: cannabimimetic activity of a class of compounds structurally distinct from delta 9-tetrahydrocannabinol. The Journal of pharmacology and experimental therapeutics, 263(3), 1118–1126. [PubMed] [Google Scholar]
  5. Coronado-Álvarez A, Romero-Cordero K, Macías-Triana L, Tatum-Kuri A, Vera-Barrón A, Budde EL, Machado S, Yamamoto T, Imperatori C, & Murillo-Rodríguez E (2021). The synthetic CB1 cannabinoid receptor selective agonists: Putative medical uses and their legalization. Progress in neuro-psychopharmacology & biological psychiatry, 110, 110301. 10.1016/j.pnpbp.2021.110301 [DOI] [PubMed] [Google Scholar]
  6. Grotenhermen F. (2003). Pharmacokinetics and pharmacodynamics of cannabinoids. Clinical pharmacokinetics, 42(4), 327–360. 10.2165/00003088-200342040-00003 [DOI] [PubMed] [Google Scholar]
  7. Desai RL, & Bergman J (2010). Drug discrimination in methamphetamine-trained rats: effects of cholinergic nicotinic compounds. Journal of Pharmacology and Experimental Therapeutics, 335(3), 807–816. 10.1124/jpet.110.173773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Desai RI, Barber DJ, & Terry P (2003). Dopaminergic and cholinergic involvement in the discriminative stimulus effects of nicotine and cocaine in rats. Psychopharmacology (Berl), 167(4):335–43. [DOI] [PubMed] [Google Scholar]
  9. Desai RT, Thakur GA, Vemuri VK, Bajaj S, Makriyannis A, & Bergman J (2013). Analysis of tolerance and behavioral/physical dependence during chronic CB1 agonist treatment: effects of CB1 agonists, antagonists, and noncannabinoid drugs. The Journal of pharmacology and experimental therapeutics, 344(2), 319–328. 10.1124/jpet.112.198374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Dutta S, Selvam B, Das A, & Shukla D (2022). Mechanistic origin of partial agonism of tetrahydrocannabinol for cannabinoid receptors. The Journal of biological chemistry, 298(4), 101764. 10.1016/j.jbc.2022.101764 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Fan F, Compton DR, Ward S, Melvin L, Martin BR (1994) Development of cross-tolerance between delta 9-tetrahydrocannabinol, CP 55,940 and WIN 55,212. Journal of Pharmacology and Experimental Therapeutics 271: 1383–1390. [PubMed] [Google Scholar]
  12. Finn DP, Haroutounian S, Hohmann AG, Krane E, Soliman N, & Rice ASC (2021). Cannabinoids, the endocannabinoid system, and pain: a review of preclinical studies. Pain, 162(Suppl 1), S5–S25. 10.1097/j.pain.0000000000002268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Gatch MB, & Forster MJ (2014). Δ9-Tetrahydrocannabinol-like discriminative stimulus effects of compounds commonly found in K2/Spice. Behavioural pharmacology, 25(8), 750–757. 10.1097/FBP.0000000000000093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Gifford AN, Bruneus M, Gatley SJ, Lan R, Makriyannis A, Volkow ND (1999) Large receptor reserve for cannabinoid actions in the central nervous system. Journal of Pharmacology and Experimental Therapeutics 288: 478–483. [PubMed] [Google Scholar]
  15. Ginsburg BC, Schulze DR, Hruba L, & McMahon LR (2012). JWH-018 and JWH-073: Δ9-tetrahydrocannabinol-like discriminative stimulus effects in monkeys. The Journal of pharmacology and experimental therapeutics, 340(1), 37–45. 10.1124/jpet.111.187757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Gómez R, Navarro M, Ferrer B, Trigo JM, Bilbao A, Del Arco T, Cippitelli A, Nava F, Piomelli D, & Rodríguez de Fonseca F (2002). A peripheral mechanism for CB1 cannabinoid receptor-dependent modulation of feeding. The Journal of neuroscience : the official journal of the Society for Neuroscience, 22(21), 9612–9617. 10.1523/JNEUROSCI.22-21-09612.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Han S, Thatte J, Buzard DJ, & Jones RM (2013). Therapeutic utility of cannabinoid receptor type 2 (CB(2)) selective agonists. Journal of medicinal chemistry, 56(21), 8224–8256. 10.1021/jm4005626 [DOI] [PubMed] [Google Scholar]
  18. Hart CL, Ward AS, Haney M, Comer SD, Foltin RW, & Fischman MW (2002). Comparison of smoked marijuana and oral Delta(9)-tetrahydrocannabinol in humans. Psychopharmacology, 164(4), 407–415. 10.1007/s00213-002-1231-y [DOI] [PubMed] [Google Scholar]
  19. Haney M; Hart CL; Vosburg SK; Nasser J; Bennett A; Zubaran C; Foltin RW (2004). Marijuana withdrawal in humans: effects of oral THC or divalproex. Neuropsychopharmacology 29, 158–70. [DOI] [PubMed] [Google Scholar]
  20. Hoffman AF, Lycas MD, Kaczmarzyk JR, Spivak CE, Baumann MH, Lupica CR (2017) Disruption of hippocampal synaptic transmission and long-term potentiation by psychoactive synthetic cannabinoid ‘Spice’ compounds: comparison with Δ(9) - tetrahydrocannabinol. Addict Biol 22: 390–399. 10.1111/adb.12334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Hruba L, Ginsburg BC, & McMahon LR (2012). Apparent inverse relationship between cannabinoid agonist efficacy and tolerance/cross-tolerance produced by Δ9-tetrahydrocannabinol treatment in rhesus monkeys. The Journal of pharmacology and experimental therapeutics, 342(3), 843–849. 10.1124/jpet.112.196444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Hruba L, & McMahon LR (2017). Apparent Affinity Estimates and Reversal of the Effects of Synthetic Cannabinoids AM-2201, CP-47,497, JWH-122, and JWH-250 by Rimonabant in Rhesus Monkeys. The Journal of pharmacology and experimental therapeutics, 362(2), 278–286. 10.1124/jpet.117.240572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Huestis MA (2007). Human cannabinoid pharmacokinetics. Chemistry & biodiversity, 4(8), 1770–1804. 10.1002/cbdv.200790152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Järbe TU, DiPatrizio NV, Lu D, & Makriyannis A (2004). (−)-Adamantyl-delta8-tetrahydrocannabinol (AM-411), a selective cannabinoid CB1 receptor agonist: effects on open-field behaviors and antagonism by SR-141716 in rats. Behavioural pharmacology, 15(7), 517–521. 10.1097/00008877-200411000-00008 [DOI] [PubMed] [Google Scholar]
  25. Järbe TU, Deng H, Vadivel SK, & Makriyannis A (2011a). Cannabinergic aminoalkylindoles, including AM678=JWH018 found in ‘Spice’, examined using drug (Δ(9)-tetrahydrocannabinol) discrimination for rats. Behavioural pharmacology, 22(5-6), 498–507. 10.1097/FBP.0b013e328349fbd5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Järbe TU, Gifford RS, Zvonok A, & Makriyannis A (2016a). Δ9-Tetrahydrocannabinol discriminative stimulus effects of AM2201 and related aminoalkylindole analogs in rats. Behavioural pharmacology, 27(2-3 Spec Issue), 211–214. 10.1097/FBP.0000000000000196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Järbe TU, LeMay BJ, Halikhedkar A, Wood J, Vadivel SK, Zvonok A, & Makriyannis A (2014). Differentiation between low- and high-efficacy CB1 receptor agonists using a drug discrimination protocol for rats. Psychopharmacology, 231(3), 489–500. 10.1007/s00213-013-3257-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Järbe TU, LeMay BJ, Thakur GA, Makriyannis A (2016b) A high efficacy cannabinergic ligand (AM4054) used as a discriminative stimulus: Generalization to other adamantyl analogs and Δ(9)-THC in rats. Pharmacol Biochem Behav 148: 46–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Järbe TU, LeMay BJ, Vemuri VK, Vadivel SK, Zvonok A, & Makriyannis A (2011b). Central mediation and differential blockade by cannabinergics of the discriminative stimulus effects of the cannabinoid CB1 receptor antagonist rimonabant in rats. Psychopharmacology, 216(3), 355–365. 10.1007/s00213-011-2226-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Järbe TU, Tai S, LeMay BJ, Nikas SP, Shukla VG, Zvonok A, & Makriyannis A (2012). AM2389, a high-affinity, in vivo potent CB1-receptor-selective cannabinergic ligand as evidenced by drug discrimination in rats and hypothermia testing in mice. Psychopharmacology, 220(2), 417–426. 10.1007/s00213-011-2491-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Jordan CJ, & Xi ZX (2019). Progress in brain cannabinoid CB2 receptor research: From genes to behavior. Neuroscience and biobehavioral reviews, 98, 208–220. 10.1016/j.neubiorev.2018.12.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Kangas BD, Delatte MS, Vemuri VK, Thakur GA, Nikas SP, Subramanian KV, Shukla VG, Makriyannis A, & Bergman J (2013). Cannabinoid discrimination and antagonism by CB(1) neutral and inverse agonist antagonists. The Journal of pharmacology and experimental therapeutics, 344(3), 561–567. 10.1124/jpet.112.201962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Kangas BD, Zakarian AS, Vemuri K, Alapafuja SO, Jiang S, Nikas SP, Makriyannis A, & Bergman J (2020). Cannabinoid Antagonist Drug Discrimination in Nonhuman Primates. The Journal of pharmacology and experimental therapeutics, 372(1), 119–127. 10.1124/jpet.119.261818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Katz JL, Kopajtic TA, Agoston GE, Newman AH (2004). Effects of N-substituted analogs of benztropine: diminished cocaine-like effects in dopamine transporter ligands. J Pharmacol Exp Ther. 309(2):650–60. [DOI] [PubMed] [Google Scholar]
  35. Khajehali E, Malone DT, Glass M, Sexton PM, Christopoulos A, Leach K (2015) Biased agonism and biased allosteric modulation at the CB1 cannabinoid receptor. Molecular pharmacology 88: 368–379. [DOI] [PubMed] [Google Scholar]
  36. Kibret BG, Ishiguro H, Horiuchi Y, & Onaivi ES (2022). New Insights and Potential Therapeutic Targeting of CB2 Cannabinoid Receptors in CNS Disorders. International journal of molecular sciences, 23(2), 975. 10.3390/ijms23020975 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Klumpers LE, Beumer TL, van Hasselt JGC, Lipplaa A, Karger LB, Kleinloog HD, Freijer JI, de Kam ML and van Gerven JMA (2012). Novel Δ9-tetrahydrocannabinol formulation Namisol® has beneficial pharmacokinetics and promising pharmacodynamic effects. British Journal of Clinical Pharmacology, 74: 42–53. 10.1111/j.1365-2125.2012.04164.X [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Lambert DM, & Fowler CJ (2005). The endocannabinoid system: drug targets, lead compounds, and potential therapeutic applications. Journal of medicinal chemistry, 48(16), 5059–5087. 10.1021/jm058183t [DOI] [PubMed] [Google Scholar]
  39. Li SM, Kopajtic TA, O’Callaghan MJ, Agoston GE, Cao J, Newman AH, Katz JL (2011). N-substituted benztropine analogs: selective dopamine transporter ligands with a fast onset of action and minimal cocaine-like behavioral effects. J Pharmacol Exp Ther. 336(2):575–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Lowe H, Toyang N, Steele B, Bryant J, & Ngwa W (2021). The Endocannabinoid System A Potential Target for the Treatment of Various Diseases. International journal of molecular sciences, 22(17), 9472. 10.3390/ijms22179472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Lu D, Meng Z, Thakur GA, Fan P, Steed J, Tartal CL, Hurst DP, Reggio PH, Deschamps JR, Parrish DA, George C, Järbe TU, Lamb RJ, & Makriyannis A (2005). Adamantyl cannabinoids: a novel class of cannabinergic ligands. Journal of medicinal chemistry, 48(14), 4576–4585. 10.1021/jm058175c [DOI] [PubMed] [Google Scholar]
  42. Luk T, Jin W, Zvonok A, Lu D, Lin XZ, Chavkin C, Makriyannis A, & Mackie K (2004). Identification of a potent and highly efficacious, yet slowly desensitizing CB1 cannabinoid receptor agonist. British journal of pharmacology, 142(3), 495–500. 10.1038/sj.bjp.0705792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. McMahon LR (2006). Characterization of cannabinoid agonists and apparent pA2 analysis of cannabinoid antagonists in rhesus monkeys discriminating Delta9-tetrahydrocannabinol. The Journal of pharmacology and experimental therapeutics, 319(3), 1211–1218. 10.1124/jpet.106.107110 [DOI] [PubMed] [Google Scholar]
  44. McMahon LR, & Koek W (2007). Differences in the relative potency of SR 141716A and AM 251 as antagonists of various in vivo effects of cannabinoid agonists in C57BL/6J mice. European journal of pharmacology, 569(1-2), 70–76. 10.1016/j.ejphar.2007.04.054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. National Academy of Sciences (2011) Guide for the Care and Use of Laboratory Animals, 8th edn. The national academies press: Washington, D.C. https://nap.nationalacademies.org/ [Google Scholar]
  46. Papanastasiou IP, Georgiadis MO, Iliopoulos-Tsoutsouvas C, Paronis CA, Brust CA, Tran NK, Ji L, Ma X, Wood JT, Zvonok N, Tong F, Bohn LM, Nikas SP, & Makriyannis A (2022). Improved cyclobutyl nabilone analogs as potent CB1 receptor agonists. European journal of medicinal chemistry, 230, 114027. 10.1016/j.ejmech.2021.114027 [DOI] [PubMed] [Google Scholar]
  47. Paronis CA, & Bergman J (2011). Buprenorphine and opioid antagonism, tolerance, and naltrexone-precipitated withdrawal. The Journal of pharmacology and experimental therapeutics, 336(2), 488–495. 10.1124/jpet.110.173823 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Paronis CA, Nikas SP, Shukla VG, & Makriyannis A (2012). Δ(9)-Tetrahydrocannabinol acts as a partial agonist/antagonist in mice. Behavioural pharmacology, 23(8), 802–805. 10.1097/FBP.0b013e32835a7c4d [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Randall PA, Vemuri VK, Segovia KN, Torres EF, Hosmer S, Nunes EJ, Santerre JL, Makriyannis A, Salamone JD. The novel cannabinoid CB1 antagonist AM6545 suppresses food intake and food-reinforced behavior. Pharmacology, Biochemistry, and Behavior. 97: 179–84. DOI: 10.1016/J.Pbb.2010.07.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Rodriguez JS, & McMahon LR (2014). JWH-018 in rhesus monkeys: differential antagonism of discriminative stimulus, rate-decreasing, and hypothermic effects. European journal of pharmacology, 740, 151–159. 10.1016/j.ejphar.2014.06.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Sachs J, McGlade E, & Yurgelun-Todd D (2015). Safety and Toxicology of Cannabinoids. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics, 12(4), 735–746. 10.1007/s13311-015-0380-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Schurman LD, Lu D, Kendall DA, Howlett AC, & Lichtman AH (2020). Molecular Mechanism and Cannabinoid Pharmacology. Handbook of experimental pharmacology, 258, 323–353. 10.1007/164_2019_298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Sim LJ, Hampson RE, Deadwyler SA, & Childers SR (1996). Effects of chronic treatment with delta9-tetrahydrocannabinol on cannabinoid-stimulated [35S]GTPgammaS autoradiography in rat brain. The Journal of neuroscience : the official journal of the Society for Neuroscience, 16(24), 8057–8066. 10.1523/JNEUROSCI.16-24-08057.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Singh H, Schulze DR, & McMahon LR (2011). Tolerance and cross-tolerance to cannabinoids in mice: schedule-controlled responding and hypothermia. Psychopharmacology, 215(4), 665–675. 10.1007/s00213-010-2162-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Sink KS, Segovia KN, Nunes EJ, Collins LE, Vemuri VK, Thakur G, Makriyannis A, & Salamone JD (2009). Intracerebroventricular administration of cannabinoid CB1 receptor antagonists AM251 and AM4113 fails to alter food-reinforced behavior in rats. Psychopharmacology, 206(2), 223–232. 10.1007/s00213-009-1602-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Tai S, Hyatt WS, Gu C, Franks LN, Vasiljevik T, Brents LK, Prather PL, & Fantegrossi WE (2015). Repeated administration of phytocannabinoid Δ(9)-THC or synthetic cannabinoids JWH-018 and JWH-073 induces tolerance to hypothermia but not locomotor suppression in mice, and reduces CB1 receptor expression and function in a brain region-specific manner. Pharmacological research, 102, 22–32. 10.1016/j.phrs.2015.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Tam J, Vemuri VK, Liu J, Batkai S, Mukhopadhyay B, Godlewski G, Osei-Hyiaman D, Ohnuma S, Ambudkar SV, Pickel J, Makriyannis A, & Kunos G (2010). Peripheral CB1 cannabinoid receptor blockade improves cardiometabolic risk in mouse models of obesity. The Journal of clinical investigation, 120(8), 2953–2966. 10.1172/JCI42551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Thakur GA, Bajaj S, Paronis C, Peng Y, Bowman AL, Barak LS, Caron MG, Parrish D, Deschamps JR, & Makriyannis A (2013). Novel adamantyl cannabinoids as CB1 receptor probes. Journal of medicinal chemistry, 56(10), 3904–3921. 10.1021/jm4000775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Thakur GA, Nikas SP, & Makriyannis A (2005). CB1 cannabinoid receptor ligands. Mini reviews in medicinal chemistry, 5(7), 631–640. 10.2174/1389557054368772 [DOI] [PubMed] [Google Scholar]
  60. Thomas BF, Compton DR, & Martin BR (1990). Characterization of the lipophilicity of natural and synthetic analogs of delta 9-tetrahydrocannabinol and its relationship to pharmacological potency. The Journal of pharmacology and experimental therapeutics, 255(2), 624–630. [PubMed] [Google Scholar]
  61. Thomas BF, Gilliam AF, Burch DF, Roche MI, & Seltzman HH (1998). Comparative receptor binding analyses of cannabinoid agonists and antagonists. The Journal of pharmacology and experimental therapeutics, 285(1), 285–292. [PubMed] [Google Scholar]
  62. Walker EA, Zernig G, Woods JH (1995) Buprenorphine antagonism of mu opioids in the rhesus monkey tail-withdrawal procedure. Journal of Pharmacology and Experimental Therapeutics 273: 1345–1352. [PubMed] [Google Scholar]
  63. Wang XF, Galaj E, Bi GH, Zhang C, He Y, Zhan J, Bauman MH, Gardner EL, & Xi ZX (2020). Different receptor mechanisms underlying phytocannabinoid- versus synthetic cannabinoid-induced tetrad effects: Opposite roles of CB1 /CB2 versus GPR55 receptors. British journal of pharmacology, 177(8), 1865–1880. 10.1111/bph.14958 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Wiley JL, Compton DR, Dai D, Lainton JA, Phillips M, Huffman JW, Martin BR (1998) Structure-activity relationships of indole-and pyrrole-derived cannabinoids. Journal of Pharmacology and Experimental Therapeutics 285: 995–1004. [PubMed] [Google Scholar]
  65. Wiley JL, Lefever TW, Cortes RA, & Marusich JA (2014). Cross-substitution of Δ9-tetrahydrocannabinol and JWH-018 in drug discrimination in rats. Pharmacology, biochemistry, and behavior, 124, 123–128. 10.1016/j.pbb.2014.05.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Wiley JL, Lefever TW, Marusich JA, Grabenauer M, Moore KN, Huffman JW, & Thomas BF (2016). Evaluation of first generation synthetic cannabinoids on binding at non-cannabinoid receptors and in a battery of in vivo assays in mice. Neuropharmacology, 110(Pt A), 143–153. 10.1016/j.neuropharm.2016.07.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Wiley JL, Marusich JA, Martin BR, & Huffman JW (2012). 1-Pentyl-3-phenylacetylindoles and JWH-018 share in vivo cannabinoid profiles in mice. Drug and alcohol dependence, 123(1-3), 148–153. 10.1016/j.drugalcdep.2011.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES