+ All Categories
Home > Documents > [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent...

[Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent...

Date post: 08-Dec-2016
Category:
Upload: julia
View: 225 times
Download: 8 times
Share this document with a friend
123
Progress in Medicinal Chemistry – Vol. 44, Edited by F.D. King and G. Lawton r 2006 Elsevier B.V. All rights reserved. 6 Recent Progress in Cannabinoid Research JULIA ADAM, PHILLIP M. COWLEY, TAKAO KIYOI, ANGUS J. MORRISON and CHRISTOPHER J.W. MORT Organon Research, Newhouse, Lanarkshire, ML1 5SH, Scotland, UK INTRODUCTION 208 CANNABINOID RECEPTORS AND ENDOCANNABINOIDS 208 ANANDAMIDE TRANSPORT INHIBITORS 210 The Hydrophobic Chain 211 The Carboxamide/Carboxylate Group 211 The Polar Head Group 211 FATTY AMIDE ACID HYDROLASE INHIBITORS 212 Organosulfonate and Organophosphonate Analogues 213 Trifluoromethylketones 215 a-Ketoheterocycles 216 Carbamates 217 Other Structures 219 CANNABINOID RECEPTOR AGONISTS 220 Classical Cannabinoids 220 Cannabidiol Derivatives (Resorcinols) 233 Non-classical Cannabinoids 235 Endocannabinoid Derivatives 237 Indole and its Derivatives 247 CB 2 AGONISTS 259 CB 2 Selective Classical Cannabinoids 260 Indoles and Indazoles 262 Resorcinol Derivatives 266 Benzo[c]Chromen-6-one Derivatives 268 Other Heterocyclic CB 2 Agonists 269 DOI: 10.1016/S0079-6468(05)44406-9 207
Transcript
Page 1: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Progress in Medicinal Chemistry –Vol. 44,Edited by F.D. King and G. Lawtonr 2006 Elsevier B.V. All rights reserved.

6 Recent Progress in Cannabinoid

Research

JULIA ADAM, PHILLIP M. COWLEY, TAKAO KIYOI, ANGUSJ. MORRISON and CHRISTOPHER J.W. MORT

Organon Research, Newhouse, Lanarkshire, ML1 5SH, Scotland, UK

INTRODUCTION

208

CANNABINOID RECEPTORS AND ENDOCANNABINOIDS

208

ANANDAMIDE TRANSPORT INHIBITORS

210

The Hydrophobic Chain

211

The Carboxamide/Carboxylate Group

211

The Polar Head Group

211

FATTY AMIDE ACID HYDROLASE INHIBITORS

212

Organosulfonate and Organophosphonate Analogues

213

Trifluoromethylketones

215

a-Ketoheterocycles

216

Carbamates

217

Other Structures

219

CANNABINOID RECEPTOR AGONISTS

220

Classical Cannabinoids

220

Cannabidiol Derivatives (Resorcinols)

233

Non-classical Cannabinoids

235

Endocannabinoid Derivatives

237

Indole and its Derivatives

247

CB2 AGONISTS

259

CB2 Selective Classical Cannabinoids

260

Indoles and Indazoles

262

Resorcinol Derivatives

266

Benzo[c]Chromen-6-one Derivatives

268

Other Heterocyclic CB2 Agonists

269

DOI: 10.1016/S0079-6468(05)44406-9 207

Page 2: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH208

THERAPEUTIC APPLICATIONS OF CANNABINOID AGONISTS

270

CB1 RECEPTOR ANTAGONISTS

272

1,5-Diaryl-pyrazoles

273

4,5-Dihydro-1H-pyrazole Derivatives

282

Imidazole-, Thiazole-, Pyrrole- and Triazole-based CB1 Receptor Antagonists

285

Pyridine-, Phenyl-, Pyrimidine- and Pyrazole-based CB1 Receptor Antagonists

295

Azetidine-based CB1 Receptor Antagonists

301

Substituted Amide-based CB1 Receptor Antagonists

303

Hydantoin-based CB1 Receptor Analogues

304

Recent CB1 Receptor Antagonists

307

THERAPEUTIC APPLICATIONS OF CB1 RECEPTOR ANTAGONISTS

308

CB2 RECEPTOR ANTAGONISTS

310

SUMMARY AND FUTURE PROSPECTS

313

REFERENCES

313

INTRODUCTION

This article aims to update the previous review of the cannabinoids in thisseries, which was written in 1998 by Mechoulam et al. [1]. The intervening7 year period has seen a great deal of activity in the area that has bothexpanded our knowledge of cannabinoid pharmacology and delivered novelligands and drug candidates. Cannabinoid ligands and their pharmacologyhave also been the topic of a number of recent review articles [2–7].

CANNABINOID RECEPTORS AND ENDOCANNABINOIDS

The previous review article in this series described the discovery andcloning of cannabinoid CB1 and CB2 receptors and a classification of thesereceptors was provided in 2002 [1, 6]. Cannabinoid CB1 receptors areexpressed primarily within the central nervous system (CNS), where theyare widely distributed. The CB1 receptor is also present in some peripheraltissues. The CB2 receptor is found mainly in cells of the immune systemand exhibits 48% homology with the CB1 receptor. Both receptors arecoupled to Gi/o proteins. Evidence exists for the presence of additionalcannabinoid receptor subtypes, as covered by a recent review article [8].Furthermore, a recent patent application described high-affinity binding ofa number of cannabinoid ligands to GPR55, suggesting that this might beone of the additional cannabinoid receptors responsible for the pharma-cological observations [9].

Page 3: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 209

Since the cloning of the cannabinoid receptors, their endogenous ligands,the endocannabinoids, have received a great deal of research interest.A number of recent review articles have extensively covered the end-ocannabinoid system [10–12], so the coverage in this article will be brief.The best studied of the endocannabinoids are anandamide (N-arachidonyl-

ethanolamine, AEA)(1) and 2-arachidonylglycerol (2-AG)(2). Anandamidewas first identified from porcine brain extracts by Devane and co-workers in1992 [13], while 2-AG was first reported in 1995 to have been isolated fromcanine gut [14] and rat brain [15]. More recently, noladin ether (2-arachidonyl-glyceryl ether, 2-AGE)(3) [16], virodhamine (O-arachidonyl-ethanolamine)(4)[17] and N-arachidonyl-dopamine (NADA)(5) [18] were proposed as endog-enous ligands for the cannabinoid receptors. In a subsequent publication, theauthors failed to detect noladin ether in mammalian brains and questioned therelevance of this compound as an endocannabinoid [19]. Anandamide, noladinether and NADA have functional selectivity for CB1 receptors, virodhamine isCB2 selective and 2-AG is essentially non-selective.

(CH2)3Rn-C4H9 4

(1) Anandamide R = CONH(CH2)2OH

(2) 2-Arachidonylglycerol R = CO2CH(CH2OH)2

(3) Noladin ether R = CH2OCH(CH2OH)2

(4) Virodhamine R = CO2(CH2)2NH2

(5) N-Arachidonyl-dopamine R = CONH(CH2)2-3,4-(OH)2-Ph

The biosyntheses of anandamide and 2-AG have been studied in depth[10]. These compounds appear to be synthesised on demand in response tocertain stimuli, rather than being stored in cells. Little is known regardingthe biosynthesis of noladin ether, virodhamine or NADA.The mechanism of release of endocannabinoids from cells and their subse-

quent re-uptake remain as points of discussion. Several groups have proposedthe existence of an anandamide transporter that can selectively release andremove endocannabinoids from their site of action [20–22]. Indeed, a numberof compounds have been proposed as inhibitors of the putative anandamidetransporter as will be outlined in the following section. An alternative sugges-tion is that the release and re-uptake of endocannabinoids is a simple diffusionprocess, with concentrations and hence diffusion being driven by the enzymefatty acid amide hydrolase (FAAH) [23]. This suggestion has recently beenrefuted by Fegley and co-workers [24], who investigated anandamide internal-isation and activity in wild-type and FAAH knock-out mice treated withanandamide transport (ANT) inhibitors, concluding that anandamide uptakewas independent of FAAH activity. The discussions will only be concludedonce the putative anandamide transporter is identified and cloned.

Page 4: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH210

FAAH was originally purified and cloned from rat liver microsomes andis able to catalyse the hydrolysis of anandamide and 2-AG, in addition toother long-chain fatty acid amides [25]. Studies into the structure and role ofthis enzyme have generated interest in the potential therapeutic applicationsof FAAH inhibitors [26–28]. FAAH knock-out mouse brains contained 15-fold higher levels of anandamide than their wild-type counterparts and theseanimals have also been shown to be more responsive to exogenously ad-ministered anandamide [29]. These animals also showed a reduced responseto painful stimuli, supporting the hypothesis that FAAH inhibition mayprovide novel analgesics. Levels of 2-AG were not elevated in the FAAHknock-out animals, apparently due to the existence of alternative metabolicfates for this compound [30].

ANANDAMIDE TRANSPORT INHIBITORS

To date the ANT inhibitors reported in the literature are all derivatives of long-chain fatty acids. These long-chain fatty acids can be split into three regions: thehydrophobic chain, the carboxamide/carboxylate group and the polar headgroup. These regions are highlighted for anandamide (1) (Figure 6.1). Com-prehensive reviews of ANT inhibitors and their potential as drugs for CNS-related disorders have been recently reported [31, 32], therefore the followingdiscussion will only highlight some of the structure–affinity relationships (SAR)established. The interested reader is directed to the review of Lopez-Rodrıguezet al. [31] for a more detailed discussion.

NH

O

n-C5H11

OH

(1) Anandamide

hydrophobic chaincarboxamide/carboxylategroup

polar head group

Fig. 6.1 Structure of anandamide.

Page 5: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.1 SAR OF THE HYDROPHOBIC CHAIN [33] R-CONH(CH2)2OH

Cpd. R IC50 (mM)

(1)a

(CH2)3-4

n-C4H9

15.1

(6)

(CH2)7-2

n-C6H13

10.6

(7)

(CH2)7-n-C8H17

10.5

(8)n-C8H17

(CH2)7- >100

(9) n-C17H35 >100

aAnandamide.

J. ADAM ET AL. 211

THE HYDROPHOBIC CHAIN

A variety of long-chain hydrocarbons have been found to be tolerated inthis position as well as the parent hydrocarbon. In general, a minimum ofone double bond located near or at the mid-point of the carbon chain isrequired for activity. In addition, the double bond must be cis configured(Table 6.1) [33].

THE CARBOXAMIDE/CARBOXYLATE GROUP

A wide range of groups are tolerated in this position including esters, am-ides, thioesters, ureas, acyl hydrazines and carbamates exhibiting a widerange of potencies and microsomal stabilities (Table 6.2) [24, 33–36].

THE POLAR HEAD GROUP

The head group has been extensively studied and a diverse range of groupsare tolerated including aliphatic, cyclic, aromatic and heteroaromaticgroups [33, 34, 37]. In general, the presence of a polar head group is fa-voured but not essential. The presence of aromatic/heteroaromatic moietiesis well tolerated; in addition, bulky groups can also be accommodated(Table 6.3).The furan derivative (23) has been tested in vivo in tests of motor activity

(open-field test) and antinociception (hot-plate test) as well as its capacity toenhance the hypokinetic and/or analgesic actions of subeffective doses of

Page 6: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.2 SAR OF THE CARBOXAMIDE/CARBOXYLATE GROUP

(CH2)3R4

n-C4H9

Cpd. R IC50

(mM)Ref. Cpd. R IC50

(mM)Ref.

(1) –CONH(CH2)2OH 15.1 [33] (14)

NH

O

NH

OH

Me 1.78 [35]

(10) –CO2(CH2)2OH 6.7 [33] (15)O

O

NH

OH

Me

7.1 [35]

(11) –CONH2 9 [33] (16)

NH

O

OOH

Me 3.29 [35]

(12)

-CON O25 [33] (17)

NH

O

NH

OH

OMe

17.7 [36]

(13) O-C(O)SCH2

9 [34] (18)

NH

O

OH 2.5 [24]

RECENT PROGRESS IN CANNABINOID RESEARCH212

anandamide [38]. It was found that (23) was mostly inactive when admin-istered alone, but was able to potentiate the action of an exogenous dose ofanandamide that produced no effect by itself.

FATTY AMIDE ACID HYDROLASE INHIBITORS

The enzyme FAAHwas first identified in the 1980s [39], cloned in the 1990s [25]and more recently, the crystal structure [40] of FAAH bound to methylarachidonyl phosphonate has been published. The FAAH enzyme has widesubstrate specificity capable of catalysing the hydrolysis of a wide variety of

Page 7: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.3 SAR OF THE POLAR HEAD GROUP

(CH2)3CONHR4

n-C4H9

Cpd. R IC50

(mM)Ref. Cpd. R IC50

(mM)Ref.

(1) –(CH2)2OH 15.1 [33] (22)

OH2.2 [33]

(19) –Et 48.5 [33] (23)

O

0.8 [34]

(20)

OH

Me 10.4 [33] (24) -( CH2)2O O 16.5 [37]

(21)

OH

Me 37.7 [33]

J. ADAM ET AL. 213

bioactive fatty acid amides and esters including the endogenous cannabinoidligands anandamide and 2-AG. Several other long-chain fatty acids and amidesalso act as substrates including palmitoylethanolamide, stearoylethanolamide,oleoylethanolamide and linoleoylethanolamide [41–44]. It has been postulatedthat selective FAAH inhibitors may elevate the levels of endogenouscannabinoids while avoiding the unwanted behavioural effects associated withthe CB1 agonists. A number of comprehensive reviews on the pharmacology,possible therapeutic uses and SAR of FAAH inhibitors have been publishedrecently [26, 45, 46]. Therefore, the following discussion will focus on high-lighting the types of structures that act as FAAH inhibitors.

ORGANOSULFONATE AND ORGANOPHOSPHONATE ANALOGUES

The first compound to find widespread use as an inhibitor of AEA break-down was phenylmethylsulfone (PMSF) (25). This non-selective serine pro-tease inhibitor has been found to act as an irreversible inhibitor of FAAHpreventing AEA breakdown [41, 47], and is commonly added to binding

Page 8: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH214

assays to improve the metabolic stability of AEA [48, 49]. PMSF has beenfound to increase the potency of AEA in inhibiting contractions in theguinea pig myenteric plexus [50], and to potentiate the effects of anandamidein vivo [51, 52]. However, the low potency (IC50 ¼ 290 nM) [53] and poorselectivity of this ligand has led to the development of more potent andselective sulfonyl fluoride inhibitors of FAAH. A number of fatty acidsulfonyl fluorides with improved potencies have been reported [53, 54], inparticular, palmitylsulfonyl fluoride (26) and arachidonylsulfonyl fluoride(27, AM374). Both these derivatives can inhibit FAAH with IC50s in the lownanomolar range (IC50 values of 7 and 0.11 nM, respectively). In addition,both these compounds have good selectivity over the CB1 receptor (IC50

values of 520 and 304 nM, respectively). Furthermore, AM374 (27) has beenfound to produce a significant inhibition of [3H]-acetylcholine release fromhippocampal brain slices [55] and to enhance the behavioural effects of lowdoses of anandamide in vivo [56].

(CH2)4R4

n-C4H9

(25) (26) (27) R = SO2F(28) R = PO(OMe)F

(29)

PhCH2SO2F n-C16H33SO2F n-C12H25PO(OMe)F

(CH2)3COCF34

n-C3H7(CH2)10PO(OMe)F2

n-C4H9

(31)(30) (32)

n-C8H17PO(OMe)F

A number of organophosphate compounds have been found to irreversiblyinhibit FAAH, the most studied being methylarachidonyl fluorophosphonate,MAFP (28) [57]. MAFP was initially designed and developed as an inacti-vator of cytosolic phospholipase A2 (cPLA2) [58, 59]. It was found to be ahighly potent and selective inhibitor of FAAH (relative to other amide hy-drolytic enzymes). However, it also bound irreversibly to the CB1 receptorand prevented the subsequent binding of CP 55,940 [60]. A detailed study onmodifications to the alkyl chain of (28) has recently been carried out [61].Both saturated (29) and unsaturated (30) analogues inhibit FAAH. In par-ticular, the fully saturated C-12 derivative (29), gave a highly potent inhibitorof FAAH (IC50 ¼ 3 nM) and the CB1 receptor (KD ¼ 2:5 nM). Shortening ofthe alkyl chain to the C-8 derivative (31),O-1887, gave a slight drop in FAAHaffinity (IC50 ¼ 15 nM) but importantly, (31) failed to interact with the CB1

receptor (KD>10,000 nM).

Page 9: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 215

TRIFLUOROMETHYLKETONES

The first substrate analogue inhibitors of FAAH were reported in 1994.The anandamide analogues prepared represented three classes of putativetransition-state inhibitors: a-trifluoromethyl ketones, a-ketoesters anda-ketoamides [62]. In the initial screening studies, it was found that thetrifluoromethyl ketone compounds tested were effective inhibitors of AEAhydrolysis. A selected set of a-keto esters also inhibited hydrolysis, whilea-keto amides were ineffective. In particular, arachidonyl trifluoromethylketone (32), gave almost 100% inhibition of anandamide hydrolysis. Adetailed investigation of the structural requirements for FAAH inhi-bition with a-trifluoromethyl ketones has been carried out by Boger andco-workers [63].The electrophilic carbonyl is essential for activity; replacement with a

methyl ketone results in complete loss of activity (Table 6.4, (33) and (34)).Saturated alkyl chains are tolerated but must be at least seven carbons long,shortening the chain length further results in dramatic losses of potency(Table 6.4, (36)–(39)). In contrast, extending the chain from seven throughseventeen carbons has little effect on potency (Table 6.4, (36) and (38)).Incorporation of the oleamide D9,10 cis double bond is favoured with analmost 2-fold increase in potency compared to its saturated congener (cf.(33) and (36)), the trans isomer is also tolerated (Table 6.4, (35)). Replace-ment of the oleamide D9,10 cis double bond and alkyl tail with a phenylgroup (Table 6.4, (40)) gives a further enhancement in affinity.

Table 6.4 FAAH INHIBITORS – TRIFLUOROMETHYL KETONES [63]

CF3R

O

Cpd. R IC50 (mM)a Cpd. R IC50 (mM)a

(33)n-C8H17 (CH2)7-

0.46 (37) n-C9H19- 0.13

(34)n-C8H17 (CH2)7COMe

>100 (38) n-C7H15- 1.2

(35)n-C8H17

(CH2)7- 0.78 (39) n-C5H11- 52.8

(36) n-C17H35- 0.24 (40) Ph(CH2)7- 0.12

aInhibition of [14C]-oleamide hydrolysis in rat liver plasma membranes.

Page 10: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH216

a-KETOHETEROCYCLES

The Boger group [64–66] has extensively studied the use of a-ketoheterocyclesas FAAH inhibitors. In their initial studies, a range of a-ketoheterocyclesbased on oleic acid was synthesised. A range of five- and six-memberedmonocyclic heterocycles and three bicyclic heterocycles (benzothiazole, ben-zimidazole and benzoxazole) was examined. Although many of the com-pounds tested were found to inhibit FAAH activity with micromolaraffinities, the best results were obtained with heterocycles that incorporateda weakly basic nitrogen a – to the heterocycle (Table 6.5) [64].Following on from this, the benzoxazole scaffold was examined in detail

and it was found that incorporation of a second weakly basic nitrogen togive the a-keto oxalopyridone (47) led to a >150-fold enhancement inbinding affinity (Table 6.5). Using this head group, modifications to theoleyl side chain were investigated and found to parallel the SAR found withthe trifluoromethyl ketones previously studied by Boger [63] (see above), thebest results being obtained with saturated chain lengths of C12–C18. Afurther enhancement in potency was seen when a phenyl ring was introducedinto an optimally long side chain. The best compound identified from thisseries was the oxalopyridone (48) with Ki values of 94 and 200 pM in humanand rat, respectively. However, when tested in vivo, this compound failed topotentiate the effects of anandamide [67].

Table 6.5 FAAH INHIBITORS – a-KETOHETEROCYCLES [64]

(CH2)7CORn-C8H17

Cpd. R Ki (mM) Cpd. R Ki (mM)

(33) –CF3 0.082 (44) N

O

0.37

(41)

S

N >100 (45) N >100

(42)

O

N 0.017 (46)

N

N 0.11

(43)

N N

NN

Me

0.065 (47) NN

O

0.0023

Page 11: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 217

Following on from these studies, Boger and co-workers [65, 66] discloseda series of substituted oleyl a�ketooxazoles exploring the introductionof substituents in the 4 and 5 positions. Analogous to the observations madein the benzoxazole series, incorporation of a second heterocyclic ringcontaining a weakly basic nitrogen proximal to the oxazole substantiallyincreased FAAH inhibition. Replacement of the oleyl side chain with theoptimal side chain found in the trifluoromethyl ketone and benzoxazoleseries gave a further enhancement in FAAH inhibitiory activity. In partic-ular, the pyridyl oxazole derivative (49) had a Ki of 7 nM and was found tohave good selectivity over a panel of serine hydrolases. In addition, thecompound failed to bind to the CB1 or CB2 receptor [67]. Compound (49)has been tested in vivo in mice and found to augment not only exogenouslyadministered AEA but also the activity of endogenously produced fatty acidamide in models of antinociception. These effects could be inhibited bypretreatment with a CB1 receptor antagonist. Importantly, the anti-nociceptive properties of (49) occurred in the absence of any significanteffects on motility or motor coordination that typically accompany theglobal activation of CB1 receptors by direct agonists [67].Boger and co-workers [68] have recently extended their studies to encom-

pass heterocyclic sulfoxides and sulfones. In general, these compounds wereless potent (up to 1,000 fold) than their a-keto heterocycle congeners. The bestresults were obtained with the benzoxazole sulfoxide (50) with a Ki of 2.6mM.

Ph(CH2)5

O O

NN

Ph(CH2)5

O

O

N N

Ph(CH2)5S O

N

O

(48) (49) (50)

CARBAMATES

Tarzia et al. [69, 70] have recently reported the FAAH inhibitory activity ofa series of alkylcarbamic acid aryl esters. The starting point for their studieswas the known serine hydrolase inhibitor carbamyl (51) that had no activityat FAAH. Replacement of the small methyl group of carbamyl (51) withmore lipophilic groups and, in particular, bulky lipophilic groups resulted inincreased affinity at FAAH (Table 6.6). Exploration of replacements of thenaphthyl moiety revealed that replacement with a biphenyl group resulted inimproved affinity and in particular, the 3-biphenylyl group proved effective

Page 12: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.6 SAR OF CARBAMYL DERIVATIVES [69, 70]R1NHCOOR2

Compound R1 R2 IC50 (nM)

(51) Me 1-Naphthyl >30,000(52) Et p-Tolyl >30,000(53) Cyclohexyl 2-Naphthyl 324(54) Cyclohexyl 4-Biphenylyl 2,297(55) Cyclohexyl 3-Biphenylyl 63

RECENT PROGRESS IN CANNABINOID RESEARCH218

(Table 6.6). The most potent compound identified in this series was thecyclohexyl 3-biphenylyl carbamate (55) with an IC50 of 63 nM.Following on from these initial studies, a systematic exploration of phenyl

ring substitution on the distal phenyl ring was carried out [70]. A range ofgroups were tolerated giving good inhibitory activity in the nanomolar range.The compound giving best potency in this series was the amido biphenylderivative (56) with an IC50 value of 4.6 nM. In addition, this compound wasselective against three other serine hydrolases, had no effect on the AEAtransporter and failed to bind to the CB1 and CB2 receptors [71]. When testedin vivo, (56) augmented the levels of endogenous AEA and produced CB1-dependent anxiolytic and antinociceptive properties. Importantly, as with thea-keto heterocycle derivative (49), the carbamate inhibitor (56) did not induceany of the common side effects associated with direct CB agonists.

O

OMeNH

O

ONH

CONH2

(51) (56)

Bristol-Myers Squibb has recently disclosed two different series of carba-mate-based FAAH inhibitors. The first of these is a series of 4,5-diaryl-imidazoles in which 30 compounds are specifically claimed, an example beingcompound (57). This compound is reported to have an IC50 value ofo10nM. In addition, (57) was also active in vivo in rodent models of chemo-induced, thermal and neuropathic pain [72]. The second series of compoundsis based on oxime carbamoyl FAAH inhibitors such as (58). Compound (58)is reported to have an IC50 value of o10nM and activity in rodent models ofinflammatory pain, thermal pain and inflammatory oedema [73].

Page 13: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 219

A series of dioxane carbamate inhibitors has recently been disclosed bySanofi-Aventis. No biological data are provided for specific compounds,although most compounds are described as having IC50 values for FAAHinhibition ranging from 0.005–1 mM. Compound (59) is one of over 25compounds specifically claimed [74].Sanofi-Aventis has disclosed a series of piperidine- and piperazine-alkyl

carbamates as cannabinoid and/or FAAH modulators. No compounds arespecifically claimed in the patent. Compound (60) is reported to have anIC50 value of 85 mM and is active in a mouse pain model [75].

NN

PhPh

(CH2)6

Me

NH

O

O

F NH

n-BuOO

ON

F(57) (58)

PhO

O NHCO2CH2CONH2

N N

NHCO2CH2CONHMe

Me

(59) (60)

OTHER STRUCTURES

A number of arachidonic acid substrate analogues have been found to in-hibit FAAH. Hillard and co-workers [37] found during their studies into theSAR of ANT inhibitors (see above) that these ligands also inhibit FAAH.From these studies, the most potent FAAH inhibitor identified waschloromethylarachidonyl amide (61) with an IC50 value of 14 nM. This lig-and also bound to the CB1 receptor in the presence and absence of PMSFbut with different binding affinities (84 nM and 2.6 mM, respectively) sug-gesting that it may also be acting as a substrate at FAAH. In addition, thearchetypal AEA transport inhibitor AM 404 (22) was also an excellentinhibitor of FAAH (IC50 ¼ 0:5mM) more than 5-fold lower than its bindingaffinity value for AEA uptake (IC50 ¼ 3:4mM) [37].Another arachidonyl-based inhibitor of FAAH is arachidonyl serotonin

(62), which was reported to inhibit FAAH from rat basophilic leukaemiacells with an IC50 value of 5.6 mM, and with very little affinity at CB1

receptors [76].

Page 14: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH220

A series of palmitoylethanolamine-derived inhibitors has been describedin the literature as FAAH inhibitors [77, 78]. This study explored the effectof shortening the chain length and replacement of the ethanolamine headgroup with primary, secondary and tertiary amide alternatives. Of the com-pounds synthesised and tested, two compounds gave reasonable affinitiesfor FAAH inhibition, palmitoyl-isopropylamide (63) (IC50 ¼ 13mM) andpalmitoyl-allylamide (64) (IC50 ¼ 3:4mM). Both these compounds had littleaffinity for either CB1 or CB2 receptors.Finally, derivatives of the endogenous compound 2-octyl-g-bromoacetate

(65) have been reported as FAAH inhibitors [79]. In a limited SAR study, itwas found that replacement of the bromine with a chlorine atom had littleeffect on affinity. The replacement of the alkyl chain with oleyl-chain mimicsresulted in an increase of affinity for FAAH (approximately 5-fold). Theremoval of the halogen and replacement with either a proton or methylresulted in inactive compounds. The most potent compound identified inthis series was compound (66) with an IC50 value of 0.6 mM [79].

(CH2)3CONH(CH2)2R4

n-C4H9

NH

OH

(61) R = Cl

(62) R =

(63) (64)

n-C15H31CONHs-Bu n-C15H31CONHallyl

O

O

O

Brn-C6H13

Me O

O

O

Br(CH2)4n-C8H17

(65) (66)

CANNABINOID RECEPTOR AGONISTS

CLASSICAL CANNABINOIDS

Before the discovery of specific cannabinoid receptors, the term ‘cannabinoid’was used to describe the biologically active constituents of the Cannabis sativaplant, including D9-THC (67), cannabidiol (68) and their analogues and de-rivatives, many of which have characteristic pharmacological effects.

Page 15: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.7 D9-THC AND CANNABIDIOL

Mouse ED50 (mmol/kg)a

Cpd. CB1 Ki (nM) CB2 Ki (nM) SA TF RT Ref.

(67) D9-THC 41 3.2 4.5 4.5 [80]21 36 [81]

(68) Cannabidiol 4,350 108 483 58 [80]1,265 230 [82]

2,860 [81]

aGeneral procedures for mouse behavioural experiments are described in Reference [141].SA ¼ spontaneous activity; TF ¼ tail flick; RT ¼ rectal temperature.

J. ADAM ET AL. 221

D9-THC, the main psychoactive component of cannabis, is a moderatelypotent partial agonist of the CB1 and CB2 receptors, while cannabidiol haslittle affinity for either receptor (Table 6.7). The term ‘classical cannabinoids’is used to describe cannabinoid receptor modulators structurally related to(67), which have a tricyclic dibenzopyran core. While several other structuraltypes of cannabinoid receptor modulators have been discovered in recentyears, the classical cannabinoids are still by far the most extensively studiedgroup in terms of SAR and pharmacology.

O

Me

MeMe

OH

OH

Me

OH

Me

(67) ∆9-THC (68) Cannabidiol

1'

2'

3'

4'

5'3

456

6a

10a7

8

910

11

12

7

12

3

45

6

8

10

9

1'

2' 3'

4'

5'1''

2''

3''

4''

5''

O

Me

MeMe

OH

(73) ∆8-THC

1'

2'

3'

4'

5'3

456

6a

10a7

8

910

11

12

Page 16: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH222

The naturally occurring isomers (67) and (73) have very similar pharma-cological profiles. Owing to the greater chemical stability of (73) and itsanalogues, SAR around this template has been much more widely studiedthan those around (67). While caution must be exercised in comparing dataobtained by different laboratories and using different assay formats, thegeneral trends that emerge provide a qualitative picture of the structuralrequirements for activity. Three pharmacophoric elements around theclassical cannabinoid template have been shown to be important for CB1

receptor binding and/or activation: the hydrocarbon side chain at C3, thesubstituent at C9 and the phenolic hydroxyl group at C1 [83–85]. The effectsof alterations to the tricyclic core have also been explored. QuantitativeSAR (QSAR) pharmacophore models have been used to rationalise theobserved SAR and to predict the activity of novel compounds [86–89].

C3 Side-chain modifications

During the 1940s, more than 15 years before the structure of (67) waselucidated as the major psychoactive constituent of marijuana [90] and 40years before the identification of specific cannabinoid receptors, Adams andco-workers [91, 92] demonstrated that a 10,20-dimethylheptyl (10,20-DMH)side chain was the optimal 3-alkyl substituent for cannabinoid activity invivo in the D6a,10a-THC series. In this early research, the ataxia test in dogswas used as a standard measure of psychoactive cannabinoid activity. Theisomeric 10,10-DMH side chain was found to be less potent than the 10,20-DMH one in the D6a,10a-THC series, but the two side chains have similarbinding affinity and potency in the D8-THC series. The 10,10-DMH sidechain has the advantage of not containing any chiral centres and has there-fore found its place as a widely used replacement for the pentyl side chain of(67) and (73) in synthetic analogues.Tables 6.8–6.11 illustrate the wide range of C3 side-chain modified D8-

THC analogues that have been reported in the literature, together withassociated in vitro and in vivo data. The affinity of classical cannabinoidanalogues for the CB1 receptor has been shown to correlate with depressionof spontaneous activity and the production of antinociception, hypothermiaand catalepsy in mice, and with psychomimetic activity in humans [93].However, in some cases, there were unexplained differences between theobserved trends in binding affinity and the trends in activity in mousebehavioural models. This may point to differences in efficacy among fullagonists, partial agonists and antagonists/inverse agonists, or may reflectdifferences in in vivo metabolism or blood–brain barrier penetration or acombination of these factors.

Page 17: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.8 D8-THC ALKYL SIDE CHAINS

O

Me

MeMe R

OH

Mouse ED50 (mmol/kg)

Cpd. R CB1 Ki (nM) CB2 Ki (nM) SA TF RT Ref.

(69) 1,1-Dimethylethyl 14 a >100 a [96](70) 1,1-Dimethylpropyl 14 a 25 a [96](71) n-Butyl 65 9.0 10 6.3 [98](72) 1,1-Dimethylbutyl 11 a 1.9 4.2 [96](73) n-Pentyl (D8-THC) 44 2.9 4.8 4.5 [80]

29 25 [97](74) 1R-Me-pentyl 7.6 6.1 1.5 2.4 [94](75) 1S-Me-pentyl 20 0.3 4.8 4.8 [94](76) 2R-Me-pentyl 19 0.6 2.1 2.1 [94](77) 2S-Me-pentyl 11 0.6 6.1 1.5 [94](78) 3R-Me-pentyl 38 10 5.2 12 [94](79) 3S-Me-pentyl 53 3 1.2 4.8 [94](80) 4-Me-pentyl 141 3 1.0 30 [94](81) 1,1-Dimethylpentyl 3.9 1.1 0.4 1.5 [96](82) n-Hexyl 41 1.2 1.8 0.10 [98](83) 1,1-Dimethylhexyl 2.7 0.17 0.21 0.11 [96](84) n-Heptyl 22 0.14 0.61 0.16 [98]

0.43 0.39 [99](85) 1,1-Dimethylheptyl 0.77 0.27 0.14 0.15 [80]

0.83 0.49 [100](86) 1S,2R-DMH 0.46 0.03 0.03 0.07 [101](87) 1R,2S-DMH 0.6 0.04 0.10 0.07 [101](88) 1R,2R-DMH 0.84 0.45 0.42 0.63 [101](89) 1S,2S-DMH 0.81 0.12 0.18 0.19 [101](90) n-Octyl 8.5 0.39 0.34 0.24 [98](91) 1,1-Dimethyloctyl 0.09 0.24 0.30 1.2 [96](92) 1,1-Dimethylnonyl 1.6 2.1 5.1 3.2 [96](93) 1,1-Dimethyldecyl 6.1 b 38 9.4 [96](94) 1,1-Dimethylundecyl 26 b b b [96](95) 1,1-Dimethyldodecyl 126 Inactive [96]

aEffects not dose dependent.bPartial effect reported (SA/TF max o80%; RT max decrease o4 1C).

J. ADAM ET AL. 223

Page 18: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.9 D8-THC CONSTRAINED, UNSATURATED AND AROMATIC HYDRO-CARBON SIDE CHAINS

O

Me

MeMe R

OH

Mouse ED50 (mmol/kg)

Cpd. R CB1 Ki

(nM)CB2 Ki

(nM)SA TF RT Ref.

(96) (1-Hexyl)c-propyl 0.44 0.86 [82](97) (1-Hexyl)c-pentyl 0.45 1.92 [102](98) Adamantyl 7 [103](99) (1-c-Pentyl-1-methyl)ethyl 0.34 0.39 [97](100) (1-c-Hexyl-1-methyl)ethyl 0.57 0.65 [97](101) (1-c-Heptyl-1-methyl)ethyl 0.94 0.22 [97](102) But-3-ynyl 367 a a a [98](103) Hex-2-ynyl 11 a a a [98]

11 148 9.6 28.7 [104](104) Hept-1-ynyl 36 a 3.3 3.0 [98]

0.65 3.1 [105]172 [107]

(105) 1,6-Heptadiynyl 460 a a a [98](106) cis-Hept-1-enyl 0.86 0.09 0.09 0.13 [98](107) 1-Methyleneheptyl 2.2 3.3 [105](108) Oct-2-ynyl 4.9 0.51 0.03 2.1 [98]

5 1.4 0.088 2.8 [104](109) 2,7-Octadiynyl 4.7 a a 1.1 [98](110) cis-Oct-2-enyl 3.2 0.05 0.11 0.07 [98]

4.6 [107](111) Oct-3-ynyl 9.0 a 1.5 a [98](112) cis-Oct-3-enyl 3.4 0.09 0.16 0.19 [98]

6.9 [107](113) Oct-4-ynyl 19 a 3.3 4.2 [98]

21 [107](114) cis-Oct-4-enyl 11 0.22 0.12 0.57 [98](115) Non-2-ynyl 3.7 2.2 0.31 1.9 [98]

4 6.6 0.41 3.8 [104](116) Benzyl 68 86 [109](117) 1,1-Dimethylbenzyl 12 0.91 [109]

aPartial effect reported (SA/TF max o80%; RT max decrease o4 1C).

RECENT PROGRESS IN CANNABINOID RESEARCH224

Page 19: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.10 D8-THC SIDE CHAINS FUNCTIONALISED IN THE 10-POSITION

O

Me

MeMe R

OH

Cpd. R CB1 Ki (nM) CB2 Ki (nM) Ref.

(118) 1-Oxoheptyl 22 84 [105](119) 1-Benzoyl 297 24 [109](120) 1-Hydroxyheptyl 86 66 [105](121) (2-Hexyl)-1,3-dioxolane-2-yl 0.52 0.22 [102](122) 1,3-Dithiolane-2-yl 168 103 [102](123) (2-Pentyl)-1,3-dithiolane-2-yl 0.85 0.58 [97](124) (2-Hexyl)-1,3-dithiolane-2-yl 0.32 0.52 [105](125) (2-c-Pentyl)-1,3-dithiolane-2-yl 9.5 2.7 [97](126) (2-c-Hexyl)-1,3-dithiolane-2-yl 1.9 1.1 [97](127) (2-c-Heptyl)-1,3-dithiolane-2-yl 1.8 6.6 [97](128) (2-Phenyl)-1,3-dithiolane-2-yl 17 18 [109](129) (2-Hexyl)-4,5-dimethyl-1,3-dithiolane-2-yl 32 20 [102](130) (2-Hexyl)-1,3-benzodithiolane-2-yl 57 257 [102](131) 2-Hexyl-1,3-dithiane-2-yl 1.8 3.6 [102](132) 1-Iodoheptyl 328 [110]

J. ADAM ET AL. 225

Table 6.8 shows the simple alkyl side-chain modifications of (67). Thebinding affinity of these analogues increases with increasing side-chainlength, from butyl up to octyl. Methyl substituents in the 10- and 20-positionsof the side chain have a beneficial effect on CB1 receptor affinity and in vivopotency, as previously shown by Adams. Huffman et al. [94] further dem-onstrated that methyl substitution in the 30-position resulted in compoundswith comparable binding affinity to the parent compound, while methylsubstitution in the 40-position decreased binding affinity. A trend similar tothat seen for the pentyl side chain was observed for monomethyl substi-tution on the heptyl side chain (data not shown) [95]. Reduced CB1 bindingaffinity was seen for compounds with 10,10-dimethyl-substituted side chainslonger than octyl (92–95). These compounds also showed reduced, or no invivo activity [96].Compounds with conformationally restrained side chains, unsaturated

side chains and aromatic side chains have been synthesised in order to betterdefine the steric requirements of the binding site in the side-chain region.These are shown in Table 6.9. On average, the conformationally restrained

Page 20: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.11 OTHER FUNCTIONALISED D8-THC SIDE CHAINS

O

Me

MeMe R

OH

Mouse ED50 (mmol/kg)

Cpd. R CB1 Ki

(nM)CB2 Ki

(nM)SA TF RT Ref.

(73) Pentyl (D8-THC) 44 2.9 4.8 4.5 [80]a 4.8 49 [112]

(133) 5-Fluoropentyl 12 6.3 31 [112]57 [110]

9 [81](134) 5,5,5-Trifluoropentyl 0.82 0.82 a [112]

25 [110]30 [81]

(135) 5-Bromopentyl a 0.51 1.0 [112]7.6 [110]

(136) 5-Iodopentyl 0.23 0.90 1.8 [112]7.8 [110]

(137) 5-Bromo-1,1-dimethylpentyl 0.08 0.05 0.09 [80]0.43 [110]

(138) 7-Bromo-1,1-dimethylheptyl 0.13 0.13 0.19 [80]1.3 [110]

RECENTPROGRESSIN

CANNABIN

OID

RESEARCH

226

Page 21: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

(139) (1-Hexyl)-2,2-dichlorocyclopropyl 1.3 0.29 [82](140) (1-Hexyl)-2,2-dibromocyclohexyl 0.71 1.0 [82](141) 5-Cyano-1,1-dimethylpentyl 0.43 0.02 0.01 0.52 [113]

1.8 1.1(142) 6-Cyano-1,1-dimethylhexyl 0.6 0.02 0.03 0.01 [113]

0.19 2.9 [107](143) 5-Cyano-5-hydroxy-1,1-dimethylpentyl 13 1.4 0.29 3.9 [113]

21 3.2 [107](144) 6-Cyano-6-phenyl-1,1-dimethylhexyl 9.2 14 3.1 9.2 [113]

18 5.8 [107](145) 5-(p-Cyanophenoxy)-1,1-dimethylpentyl 3 0.22 2.5 9.2 [113]

1.5 1.1 [107](146) 5-Carboxy-1,1-dimethylpentyl 222 4.0 [107](147) 5-(N,N-dimethyl carboxamido)-1,1-dimethylpentyl 0.86 0.004 0.075 0.29 [113]

2.5 2.0 [107](148) 5-(N,N-diethyl carboxamido)-1,1-dimethylpentyl 13 0.20 0.23 0.91 [113]

24 2.5 [107](149) 5-[N-(piperidin-1-yl)carboxamido]-1,1-dimethylpentyl 1.2 0.09 0.34 0.36 [113]

4.5 3.2 [107](150) 5-[N-(p-chlorophenyl)carboxamido]-1,1-dimethylpentyl 187 a a a [113]

395 11 [107](151) 5-[N-(o,p-dichlorophenyl)carbox amido]-1,1-dimethylpentyl 18 a 26 7.0 [113]

41 37 [107](152) 5-[N-(p-aminosulfonylphenyl) carboxamido]-1,1-dimethylpentyl 19 3.8 5.9 3.1 [113]

42 10 [107](153) 5-{N-[(p-aminosulfonylphenyl) methyl]carboxamido}-1,1-dimethylpentyl 60 56 a 67 [113]

180 2.3 [107](154) 5-{N-[(p-aminosulfonylphenyl)ethyl] carboxamido}-1,1-dimethylpentyl 29 a >176 >176 [113]

246 [107]

aPartial effect reported (SA/TF max o80%; RT max decrease o4 1C).

J.ADAM

ETAL.

227

Page 22: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH228

10,10-dimethyl cycloalkyl side-chain analogues of (99–101) were found tohave in vitro activity similar to their straight-chain analogues (83), (85) and(91), respectively. However, there was no trend of increasing CB1 receptor-binding affinity with increasing ring size to mirror the effect of increasingchain length in the open-chain series [97].Compounds with cis double bonds in the side chain were in general found

to be more potent and efficacious than their triple-bond congeners, both inin vivo and in in vitro functional assays [98, 106, 107]. QSAR models havebeen generated for the compounds with unsaturated [108] and 10,10-dimethyl[96] side chains to determine more precisely the pharmacophoric require-ments of the receptor. It is postulated that for optimum potency, the sidechain must be of a suitable length and flexibility to have the ability to loopback so that its terminus is in proximity to the phenolic ring. The widelyused, potency enhancing 10- and 20-methyl substituents would be expected toincrease the tendency of the side chain to adopt a looped back, rather thanan extended conformation.Aromatic rings are also tolerated in the side chain, as demonstrated by

Krishnamurthy et al. [109]. The 10,10-dimethylbenzyl compound (117)showed higher binding affinity than the simple benzyl compound (116).Compound (117) also showed around 13-fold selectivity for CB2 over CB1

binding.A range of different functional groups has been introduced into the 10-

position of the C3 side chain, as shown in Table 6.10. A number of differentfunctionalities were shown to be tolerated in this position, with lipophilicgroups such as methyl and dithiolane being preferred over polar groups suchas ketones and alcohols. Ketone (118) had similar CB1 receptor affinity tothe unfunctionalised n-heptyl compound (84), while alcohol (120) had loweraffinity. The phenyl ketone (119) had lower CB1 receptor affinity than thesimple benzyl-substituted compound (116), but higher CB2 affinity, withabout 12-fold selectivity for CB2 over CB1.A dithiolane group in the 10-position has been shown to be at least as

effective as the 10,10-dimethyl group in enhancing the binding affinity of theclassical cannabinoids, as can be seen by comparing compounds (123) and(124) with compounds (83) and (85). However, the constrained dithiolanecompounds (125–127) showed decreased activity compared to their 10,10-dimethyl analogues (99–101). In contrast to its 10,10-dimethyl and ketoneanalogues, (117) and (119), the phenyl dithiolane compound (128) does notexhibit any CB2 selectivity.Surprisingly, an iodo substituent in the 10-position was not well tolerated,

suggesting that this substituent had an unfavourable interaction withthe receptor or a detrimental effect on the conformation of the side chain[110].

Page 23: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 229

A range of 10-substituted D8-THC, D9-THC and D6a,10a-THC derivatives,including many of those in Tables 6.9 and 6.10 has been disclosed in a patentapplication by Moore et al. [111]. The compounds are described as eitheragonists or antagonists of the CB1 and/or CB2 receptors. The in vivo activityof (1-cyclohexyl-1-methyl) ethyl compound (100) in a rat haemorrhagicshock model and the in vitro cytotoxic effects of the 10,10-dimethylbenzylcompound (117) against glioma cells are described.Halogen, cyano, carboxylic acid and amide functional groups have been

introduced at the terminus of the C3 side chain, as shown in Table 6.11. Thisarea of the molecule seems to be tolerant to both lipophilic and more polargroups. Halogen substitution at the terminal carbon of the side chain ledto enhancement of affinity, with the bulkier halogens, bromine and iodine,producing the largest effects [110, 112]. The less lipophilic 5-cyano-10,10-dimethylpentyl and 5-(N,N-dimethyl-carboxamido)-10,10-dimethylpentylcompounds (141) and (147) showed high affinity and in vivo potency similarto the 5-bromo-10,10-dimethylpentyl compound (137). However, thecyanohydrin compound (143) showed decreased affinity and potency. Thismay indicate less tolerance for a hydrogen bond donor in this position. Thecorresponding carboxylic acid (146) showed very low binding affinity atCB1, but retained affinity at CB2. The piperidine hydrazide compound (149)exhibited high binding affinity despite the presence of a hydrogen bonddonor. In this case, the hydrogen bond donor was separated by a longerlinker group from the tricyclic core.Chloro- and sulfonamide-substituted aromatic amides showed decreased

binding affinity and in vivo potency compared to the simple aliphatic am-ides. Side chains with an additional (CH2)1-2 linker between the amide andthe phenylsulfonamide group resulted in partial or absent in vivo effects[113]. The (CH2)-linked compound, (153), showed around 80-fold selectivityfor CB2 over CB1 binding [107].A number of compounds have been prepared that contain both a double

or triple bond and a terminal functional group in the side chain [98, 107,114]. In general, the combined modifications reinforced the SAR trends seenfor the individual modifications.

C9 Substituent modifications

A major route of metabolism for (67) and (73) is oxidation at the C9 po-sition to form hydroxymethyl and carboxyl metabolites. The hydroxymethylmetabolites are potent CB1 agonists with pharmacological profiles similar tothe parent compounds, while the carboxy metabolites have reduced activity[115]; the 9-carboxy analogue of (73) does not bind to the CB1 receptor [93].

Page 24: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.12 D8-THC C9 SUBSTITUENTS

O

R

MeMe

OH

n-C5H11

Mouse ED50 (mmol/kg)

Cpd. R CB1 Ki (nM) CB2 Ki (nM) SA TF RT Ref.

(73) Methyl (D8-THC) 44 2.9 4.8 4.5 [80]126 [93]

a 4.8 49 [112]23 a [116]

(155) Hydroxymethyl 55 [93]26 7.4 [117]

(156) Carboxyl >10,000 [93]b [115]

(157) Fluoromethyl a 15 21 [112]107 [93]

(158) Isopropyl 76 a [116](159) n-Butyl 90 c [116](160) 1-Hydroxyethyl 78 a [116]

a [118](161) 1-Hydroxypropyl 109 a [116](162) Phenyl 178 a [116](163) Benzyl 82 a [116](164) 2-Phenylethyl 106 a [116]

a [118]

aPartial effect reported (SA/TF max o80%; RT max decrease o4 1C).bInactive.cSee text.

RECENT PROGRESS IN CANNABINOID RESEARCH230

Some alternative substituents have also been introduced in the C9 position,as shown in Table 6.12.Of the direct methyl group replacements reported, small groups such as

hydroxymethyl and fluoromethyl are tolerated, while larger groups in thisposition result in compounds with reduced in vivo potency and efficacy. Then-butyl derivative (159) was the only C9 methyl replacement analogue thatshowed a marked effect on hypothermia (5 1C decrease at 168 mmol/kg;ED50 not calculated) [116].

Page 25: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.13 D8-THC-DMH C9 SUBSTITUENTS

Compound CB1 Ki (nM) CB2 Ki (nM) Ref.

(85) Methyl (D8-THC-DMH) 0.77 [80]0.83 0.49 [100]

(165) Hydroxymethyl (HU-210) 0.10 0.17 [117]0.23 [120]

(166) Carboxyl (CT-3) 32 171 [117]

J. ADAM ET AL. 231

The CB1 binding affinity of the hydroxymethyl and carboxyl analoguescan be increased by substituting the C3 pentyl side chain for a dimethylheptylside chain (Table 6.13). 11-Hydroxy-10,10-DMH D8-THC, HU-210 (165), is anextremely potent cannabinoid agonist that has been widely used as a phar-macological tool [119]. Its (+) enantiomer, HU-211 (dexanabinol), which isin clinical development for the treatment of cognitive disorders, does not havehigh affinity for CB1 receptors [120].The dimethylheptyl side-chain analogue of 9-carboxy-D8-THC, ajulemic

acid, CT-3, (166) is currently in clinical development for treatment of painand inflammation [121, 122]. Compound (166) does show some affinity forCB1 and CB2 receptors, but may also exert anti-inflammatory and analgesiceffects through other mechanisms. It does not appear to be psychoactive inhumans [123].

O

R

MeMe

n-C6H13

OH

Me Me

(165) HU-210 R = CH2OH

(166) CT-3 R = COOH

C1 modifications

In general, modification or deletion of the C1 phenolic hydroxyl groupresults in significantly reduced CB1 receptor affinity [93]. A number of1-deoxy and 1-alkoxy D8-THC analogues have been shown to be selectiveligands for the CB2 receptor (see below).

Page 26: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH232

Core modifications

There are limited examples in the literature where substituents have beenintroduced in other positions on the tricyclic core (Table 6.14). Substitu-tion at the C2 position of the aryl ring with an iodo substituent resulted inonly a slight drop in binding affinity. However, a nitro substituent in the2-position was not tolerated. Molecular modelling studies suggested thatthis may be attributed to dramatic reductions in the electron densityaround the phenolic hydroxyl group and the oxygen in the pyran ring.Substitution at the C4 position with either a bromo or a nitro groupresulted in loss of affinity at CB1, perhaps as the result of an unfavourablesteric interaction in the binding site. The 2,4-diiodo and 2,4-dinitro com-pounds were also inactive [80].Tetracyclic compounds in which the C3 side chain is conformationally

restricted by linking to either the C2 or the C4 position have been describedby Khanolkar et al. [99]. All the tetracyclic compounds had lower CB1 andCB2 affinity than the analogous non-constrained compounds (84). The best-tolerated constraint was the ‘southbound’ constraint in compound (173).

Table 6.14 D8-THC SUBSTITUTIONS AT C2 AND C4

OMe

Me

OH

n-C5H11

Me

R2

4

Mouse ED50 (mmol/kg)

Cpd. R CB1 Ki (nM) CB2 Ki (nM) SA TF RT Ref.

(167) 4-Br 5,250 >250 >250 >250 [80](168) 2-I a 0.68 20 [112]

89 [80](169) 2,4-di-I >10,000 >175 >175 >175 [80](170) 2-NO2 >10,000 29 a 101 [80](171) 4-NO2 1,630 >275 >275 40 [80](172) 2,4-di-NO2 >10,000 >75 >75 >75 [80](173) (Tetracyclic) 22 59 [99](174) (Tetracyclic) 402 162 [99](175) (Tetracyclic) 542 456 [99]

aPartial effect reported (SA/TF max o80%; RT max decrease o4 1C).

Page 27: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 233

O

Me

MeMe

OH

R2

R1 O

Me

MeMe

OH

n-C6H13

(173) R1 = H, R2 = n-C6H13

(174) R1 = n-C5H11, R2 = H(175)

CANNABIDIOL DERIVATIVES (RESORCINOLS)

Cannabidiol has low affinity for CB1 and CB2 receptors and is not psycho-active, but has nevertheless shown a number of pharmacological activities ofits own including anti-inflammatory and neuroprotective effects. Some sidechain-modified cannabidiol derivatives have also been evaluated for can-nabinoid receptor affinity and these are shown in Table 6.15.Bisogno et al. [124] have shown that, in contrast to classical cannabinoid

derivatives, the unnatural (+) enantiomers of cannabidiol derivatives havehigher affinity for CB1 and CB2 receptors than the natural (�) enantiomers,as shown in Table 6.16. Within both the (�) series and the (+) series, theSAR showed some similarity to those in the classical cannabinoid series, withextended and halogenated side chains tending to increase binding affinity. The100,100-dimethylheptyl, 7-hydroxy (+)-cannabidiol analogue (187) exhibitedparticularly high CB1 binding affinity. In the same study, cannabidiol andsome of its analogues were also found to be weak agonists of VR1 receptorsand weak inhibitors of anandamide uptake and degradation [124].Further (+)-cannabidiol analogues were prepared and evaluated by Fride

et al. [125], who showed that carboxylic acid derivatives (189) and (191) alsohave high CB1 receptor affinity [126]. In vivo activity was assessed in lo-comotion, rearing, catalepsy, analgesia (hot plate), hypothermia and inhi-bition of intestinal motility in mice as a percentage of the maximum possibleeffect at a single dose. The (+)-cannabidiol analogues were shown to inhibitdefecation and reduce arachidonic acid-induced ear swelling in mice. Fewcentral cannabinoid effects were observed [125]. The carboxylic acid andalcohol derivatives have been described in a patent application by the samegroup, as therapeutically useful modulators/regulators of the immune sys-tem and gastrointestinal tract [127].

Page 28: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.15 CANNABIDIOL SIDE-CHAIN MODIFICATIONS

OH

Me

R

OH

Me

Mouse ED50 (mmol/kg)

Cpd. R CB1

Ki (nM)CB2

Ki (nM)SA TF RT Ref.

(68) Pentyl (cannabidiol, CBD) 4,350 108 483 58 [80]1,265 230 [82]

>10,000 >10,000 [124](176) 1,1-Dimethylheptyl (CBD-DMH) >10,000 1,800 [124](177) (1-Hexyl)cyclopropyl 59 99 [82](178) 5,5,5-Trifluoropentyl 1,480 >75 27 >75 [80](179) (2-Hexyl)-1,3-dithiolan-2-yl 136 50 [105](180) (1-Hexyl)-2,2-dichlorocyclopropyl 665 33 [82](181) (1-Hexyl)-2,2-dibromocyclohexyl 189 63 [82]

Table 6.16 ENANTIOMERS OF CANNABIDIOL DERIVATIVES

Compound CB1 Ki (nM) CB2 Ki (nM) Ref.

(68) (�)-Cannabidiol (CBD) >10,000 >10,000 [124](182) (+)-CBD 842 203 [124](176) (�)-CBD-DMH >10,000 1,800 [124](183) (+)-CBD-DMH 17 211 [124](184) (�)-7-OH-CBD >10,000 >10,000 [124](185) (+)-7-OH-CBD 5.3 101 [125](186) (�)-7-OH-CBD-DMH 4,400 671 [124](187) (+)-7-OH-CBD-DMH 2.5 44 [124](188) (�)-1-CO2H-CBD >10,000 >10,000 [124](189) (+)-1-CO2H-CBD 13 322 [125](190) (�)-1-CO2H-CBD-DMH 1,900 5,000 [124](191) (+)-1-CO2H-CBD-DMH 5.8 156 [125]

RECENT PROGRESS IN CANNABINOID RESEARCH234

Page 29: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 235

OH

R1

R2

OH

Me

(185) R1 = CH2OH, R2 = n-C5H11

(187) R1 = CH2OH, R2 = C(Me)2n-C6H13

(189) R1 = CO2H, R2 = n-C5H11

(191) R1 = CO2H, R2 = C(Me)2n-C6H13

Oxidation of the phenol ring of cannabidiol (73) or cannabinol to a qui-none ring has been shown to afford compounds with anti-tumour activity.However, these compounds do not bind to the CB1 receptor and theirmechanism of action is unclear [128].

NON-CLASSICAL CANNABINOIDS

The term ‘non-classical cannabinoids’ is applied to a group of bicycliccompounds identified by researchers at Pfizer in the 1980s [129]. Thesecompounds lack the pyran ring of the classical cannabinoids and the secondphenolic hydroxyl group of the cannabidiols, resulting in a simplified sub-structure represented by CP 47,497 (192) [130, 131]. The non-classical can-nabinoids still retain the three main pharmacophoric elements describedabove for the classical cannabinoids and the SAR in these regions parallelsthat of the classical cannabinoids [132].A fourth important pharmacophoric element was established for the non-

classical cannabinoid series in the form of a southern aliphatic hydroxylgroup. Addition of this group to (192) resulted in the high-affinity CB1 andCB2 receptor full agonist CP 55,940 (193) [129, 133], the tritiated form ofwhich was used to first demonstrate specific cannabinoid binding sites inbrain tissue [134]. Its enantiomer, CP 56,667 (194) has lower affinity for theCB1 receptor (Table 6.17).

n-C6H13

OH

OH

Me Me

R1 n-C6H13

OH

OH

OHMe Me

(192) CP 47,497 R1 = H(193) CP 55,940 R1 = (CH2)3OH (194) CP 56,667

Page 30: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.17 NON-CLASSICAL CANNABINOIDS

Ki (nM) Mouse ED50 (mmol/kg)

Compound CB1 CB2 SA TF RT Ref.

(192) CP 47,497 9.5 [93]a 0.94 4.1 [135]

(193) CP 55,940 0.92 [93]0.69 [81]

0.11 0.24 1.1 [135](194) CP 56,667 62 [93]

24 [81]8.0 a a [135]

(195) (-)CP 55,244 0.11 [136](196) (+)CP 55,244 >1,000 [136](197) (Hybrid) 71 [137](198) (Hybrid) 1,353 [137]

aPartial effect reported (SA/TF max o80%; RT max decrease o4 1C).

RECENT PROGRESS IN CANNABINOID RESEARCH236

Melvin et al. [136] have shown that the positioning of the southern al-iphatic hydroxyl group is critical for optimal binding. The constrained an-alogue of (193), (-)-CP 55,244 (195) showed very high CB1 binding affinityand complete enantioselectivity. Conformational studies with diastereoiso-mers of (195) have suggested that the ability to form an intramolecularhydrogen bond between the phenolic hydroxyl group and the southern al-iphatic hydroxyl group may be important for receptor binding [138]. How-ever, molecular docking of these compounds in a homology model based onthe bovine rhodopsin X-ray crystal structure has suggested that these twohydroxyl groups could form intermolecular hydrogen bonds with differentresidues in the receptor-binding site [139].

n-C6H13

OH

OH

OHMe Me

n-C6H13

OH

OH

OHMe Me

(195) (-)-CP 55,244 (196) (+)-CP 55,244

Classical/non-classical hybrid cannabinoids, such as (197) and (198), havebeen described by Tius et al. [137, 140]. In these compounds, a southern

Page 31: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 237

aliphatic hydroxyl group was added to the classical cannabinoid template.Addition of a C-6a hydroxyethyl substituent resulted in a loss of bindingaffinity, while the introduction of C-6b hydroxyethyl or hydroxypropylgroups resulted in increased affinity compared to the corresponding ethyl orpropyl analogues.

n-C5H11

OH

OH

OH

O

Me

n-C5H11

OH

OH

Me OOH

(197) (198)

ENDOCANNABINOID DERIVATIVES

The development of SAR for endocannabinoid-derived structures has pri-marily focused on the anandamide skeleton (1) with a large number ofpublications addressing the requirements for activity and stability of thisscaffold. More recently, some SAR has begun to emerge for the other end-ocannabinoids, in particular 2-AG (2). The following discussion will focuson highlighting some of the main features that contribute to affinity and/orstability; each endocannabinoid will be treated separately. A number ofdetailed reviews on this subject have been published [142–146].A number of protocols are available for measuring cannabinoid-binding

affinity and as such there is a variation in reported Ki values for end-ocannabinoids across labs. For this reason, wherever possible, the relativeaffinity compared to AEA (measured in that protocol) will be given in anattempt to provide a benchmark for comparison.

Anandamide derivatives

As outlined earlier, anandamide was the first among the endogenous can-nabinoid receptor agonists to be identified. It exhibits higher binding affinityfor the CB1 receptor (K i ¼ 89 nM) than for the CB2 receptor (Ki ¼ 371 nM)[81]. Anandamide has typical cannabinoid activities including decreasedspontaneous motor activity, immobility and production of hypothermia andanalgesia [147, 148]. However, this action in vivo is of shorter duration than

Page 32: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH238

that of some of the classical cannabinoids. The low potency and short-lasting activity in vivo was recognised to be due to AEA susceptibility toenzymatic hydrolysis by FAAH (see above). Anandamide SAR studies canbe split into three categories; modifications to the fatty acid chain, mod-ifications to the carboxamide group and modifications to the polar headgroup.

The fatty acid chain

In general for the C20 series, maximal activity is achieved with amides ofarachidonic acid (1) [81], mead acid (199) [149], and dihomo-g-linoleic acid(200) [150] (see Table 6.18). Decreasing the unsaturation (201), (202), orabolishment of the n-pentyl chain (203) [150] led to less active or inactivecompounds. Variable results were seen with longer chains. The C22:4 n-6analogue (204) is as active as AEA (1) whereas the C22:6 n-3 analogue (205)is less active than the C20:5 n-3 analogue (203) [150]. Replacement of thedouble bonds with triple bonds (206) resulted in loss of activity [150] (seeTable 6.18). Forcing the fatty acid chain into a hairpin conformation bycyclisation (207) also resulted in inactive compounds [151].In a study looking at oxygenated metabolites of AEA and their inter-

action with the cannabinoid system, a series of hydroxylated alkyl chainswas prepared using different lipoxygenases as biocatalysts [152–154]. Of theseven AEA derivatives prepared, only the 5R-hydroxy (208), 12S-hydroxy(209) and 15S-hydroxy (210) derivatives had any affinity for the CB1 re-ceptor. Interestingly, the 13S-hydroxy compound (211) that was inactive atthe CB1 receptor displayed some affinity for the CB2 receptor [152] (seeTable 6.18).To improve the metabolic stability of AEA analogues, a number of mod-

ifications to the C2 site of AEA have been explored [150, 155, 156]. Intro-duction of a methyl (212) or dimethyl group (213) in this position gave amodest increase in binding affinity and enhanced metabolic stability (cf.with and without PMSF). In contrast, use of an ethyl (214) or isopropylgroup (215) resulted in a drop in binding affinity. Introduction of a chiralmethyl group into the C-2 position had little effect on binding affinity butthe R-isomer (216) had improved metabolic stability compared to itsenantiomer (217) [157]. In addition, both (216) and (217) retained theirselectivity over the CB2 receptor (see Table 6.18).Several groups have suggested that the C16–C20 portion of AEA and the

C-3 pentyl side chain of (67) may play a similar role in the binding site of theCB receptor [158–161]. To explore this, a number of groups have substitutedthe C16–C20 pentyl side chain with a 10,10-dimethylheptyl chain, a widely

Page 33: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.18 AEA DERIVATIVES – THE FATTY ACID CHAIN SARRCONH(CH2)2OH

Cpd. R CB1 (nM)+PMSF

CB1 (nM)�PMSF

CB2 (nM) Relative CB1 Ki

(Ki/KiAEA)Ref.

(1)

(CH2)3-4

n-C4H9

89 5,400 371 1 [81]

(199)

(CH2)3-3

n-C7H15753 1,810 0.96 [149]

(200)

(CH2)6-3

n-C4H9

53 1.4 [150]

(201)

(CH2)9-2

n-C4H9

1,500 38 [150]

(202)(CH2)9-n-C8H17

>1,000 >26 [150]

(203)

(CH2)3-5

Me162 4 [150]

(204)

(CH2)5-4

n-C4H9

34 0.9 [150]

(205)

(CH2)2-6

Me324 8.3 [150]

(206)(CH2)3-

4n-C4H9

No activity [150]

(Continued )

J.ADAM

ETAL.

239

Page 34: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

(207) O

n-Bu

(CH2)3-

OH

No activity [151]

(208)

3

OH

(CH2)3-n-C4H9

680 710 7.6 [152]

(209) n-C5H11

2

OH

(CH2)3-150 500 1.7 [152]

(210)

3

n-C4H9

OH

(CH2)3-600 >1,000 6.7 [152]

Table 6.18 CONTINUED

Cpd. R CB1 (nM)+PMSF

CB1 (nM)�PMSF

CB2 (nM) Relative CB1 Ki

(Ki/KiAEA)Ref.

RECENTPROGRESSIN

CANNABIN

OID

RESEARCH

240

Page 35: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

(211) (CH2)7-

2

n-C5H11

OH

>1,000 600 >11 [152]

(212)

4 Me

n-C4H9

53 137 0.6 [155]

(213)

4 Me

Men-C4H9

47 41 0.5 [155]

(214)

4 Et

n-C4H9

461 285 5.2 [155]

(215)

4 i-Pr

n-C4H9

4,030 2,920 45 [155]

(216)

4 Me

n-C4H9

54 457 4,905 0.7 [157]

(217)

4 Me

n-C4H9

35 3,941 4,259 0.45 [157]

(218)

4

n-C6H13

Me

Me(CH2)3-

7 0.08 [158]

J.ADAM

ETAL.

241

Page 36: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH242

used replacement in classical cannabinoids (see above). While the replace-ment of this portion of the molecule did not result in as great an enhance-ment as with the classical cannabinoids, the dimethylheptyl derivative (218)(Table 6.18) did produce a 13-fold enhancement in CB1 binding affinitycompared with AEA [158].In summary, the receptor recognises unsaturated and partially saturated

fatty acyl chains 20–22 carbons long. In addition to the receptor-bindingstudies detailed here, a large body of work examining the conformat-ional requirements and potential folding arrangements of the fatty acidchain via a number of different computational methods have been published[162].

The carboxamide group

A limited series of amide replacements have been examined includingthioamides (219) [163], reverse amides (220) [164], heterocycles (221) [164],ethers (222) [165], carbamates (223), (224) [165], ureas (225) [165] andketones (226) [166] (see Table 6.19). In general, replacement of the amideresults in a loss of binding affinity compared to AEA. In some cases, re-placement of the amide results in increased stability with regard to hydro-lysis by FAAH [164, 165].

Table 6.19 AEA DERIVATIVES – THE CARBOXAMIDE GROUP SAR

(CH2)3R4

n-C4H9

Cpd. R CB1 (nM)+PMSF

CB1 (nM)– PMSF

CB2

(nM)Relative CB1 Ki

(Ki/Ki AEA)Ref.

(1) –CONH(CH2)2OH 89 5,400 371 1 [81](219) –CSNH(CH2)2OH 1,380 6,300 17.7 [163](220) –CH2NHCO(CH2)2OH 115 134 3,540 1.9 [164](221)

N

O

Me

679 6.8� 106 0.11 [164]

(222) –CH2O(CH2)2OH 797 8.95 [165](223) –CH2OCONHn-Pr 471 5.3 [165](224) –NHCO2(CH2)2F 331 3.7 [165](225) –NHCONH(CH2)2F 55 52 0.6 [165](226) –CO(CH2)2OH 360 4 [166]

Page 37: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 243

The polar head group

The most extensive investigation of SAR has been carried out in this regionof AEA. The following SAR has been established. In general, a secondaryamide is optimal. The primary amide is inactive (227) and tertiary amidesare less active than AEA or inactive (228), (229) [37, 150, 163]. The hydroxylgroup is not required for activity and a wide range of replacements aretolerated such as hydrogen (230), small alkyl (231), halogen (232), (233),ether (234), phosphate (235) or aromatic (236) (see Table 6.20). In partic-ular, the chloroethyl derivative (232) was shown to have 11-fold improvedaffinity compared to AEA.The introduction of a methyl into the head group at the C10-position and

in particular the R-isomer (237) resulted in 4-fold higher affinity than AEA,while the S- isomer (238) had 2-fold lower affinity than AEA [48, 157], theR-isomer (237) was also resistant to enzymatic hydrolysis [157]. However,introduction of larger alkyl groups into the C10 position such as the i-butylanalogue (239) leads to a large drop in activity [155]. Methylation of the C20

position also leads to improved affinity compared to AEA. In particular, theR-isomer (240) has 4-fold enhanced affinity; this derivative is also stable toenzymatic hydrolysis by FAAH [48] (see Table 6.20).Extension of the head group by insertion of a methylene group is tol-

erated, the N-propanol derivative (241) having a slight increase in bindingaffinity compared to AEA. Further extension of the carbon chain, such asthe butyl derivative (242), led to a decrease in activity [150, 151].Replacement of the ethanolamine head group is also well tolerated. Sub-

stitution with a cyclopropyl (243) [37], allyl (244) or propargyl group (245)[164] all led to an increase in binding affinity compared to AEA. Replace-ment of the head group with aromatics is also allowed. The phenyl deriv-ative (246) retains affinity at the CB1 receptor [37], whereas the 2-substitutedN-methyl pyrrole (247) has a 2-fold improved affinity compared to AEA[167]. Interestingly, the 3-substituted furan derivative (23) that has micro-molar affinity for the AEA transporter (see above) does not bind to the CB1

receptor, but has good affinity for the CB2 receptor [167]. These results aresummarised in Table 6.20.Overall, these results suggest that the hydroxyl group is not vital for

binding and that both hydrophobic and hydrophilic head groups canbe accommodated. The cavity in which the head group binds is relativelysmall as only modest variations in this position lead to high-affinity lig-ands. With regard to CB2 selectivity, very few reports have looked in detailat the requirements for CB2 binding in AEA derivatives and most ligandssynthesised to date have tended to be relatively selective for the CB1

receptor.

Page 38: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.20 AEA DERIVATIVES – THE POLAR HEAD GROUP SAR

(CH2)3COR4

n-C4H9

Cpd. R CB1 (nM) +PMSF CB1 (nM) - PMSF CB2 (nM) Relative CB1 Ki (Ki/Ki AEA) Ref.

(1) –NH(CH2)2OH 89 5,400 371 1 [81](227) –NH2 >1,000 >25 [150](228) –NEt2 >1,000 >25 [150](229) –N[(CH2)2OH]2 174 2.2 [163](230) –NHEt 34 0.87 [150](231) –NHn-Pr 7.1 0.05 [37](232) –NH(CH2)2Cl 5.29 3,400 195 0.09 [164](233) –NH(CH2)2F 26.7 4,640 908 0.43 [164](234) –NH(CH2)2OMe 85.2 2 [150](235) –NH(CH2)2OP(O)(OH)2 190.8 4.9 [150](236)

-NH(CH2)2SO2NH2

163 250 1.8 [156]

(237)

-NH CH2OH

CH320.6 28.3 868 0.26 [157]

RECENTPROGRESSIN

CANNABIN

OID

RESEARCH

244

Page 39: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

(238)

-NH CH2OH

CH3173 268 8,216 2.2 [157]

(239)

-NH CH2OH

i-Bu 5,420 >10,000 61 [155]

(240)

-NHCH2 OH

CH320 28 0.26 [48]

(241) �NH(CH2)3OH 29.9 0.76 [150](242) �NH(CH2)4OH 497.4 12.7 [150](243) �NHc–Pr 2.2 193 0.015 [37](244) �NHCH2CHQCH2 9.91 2,980 226 0.16 [164](245) �NHCH2CRCH 10.8 4,900 290 0.18 [164](246) �NHPh 109 0.76 [37](247)

-NHCH2 N

Me

124 70 0.43 [167]

(23) -NHCH2

O

>1,000 67 >3.5 [167]

J.ADAM

ETAL.

245

Page 40: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH246

2-Arachidonylglycerol

2-AG (2) was first isolated in 1995 from canine gut [14] and rat brains [15] andshown to be an endogenous CB ligand [14]. (2) is present in the rat brain inamounts 170–1,000 times greater than AEA [15, 168] and has been shown to bea potent full agonist at both the CB1 and CB2 receptors (CB1: K i ¼ 472 nMand CB2: K i ¼ 1; 400 nM) [14, 169]. In addition, this endocannabinoid hastypical cannabinoid-like activities including decreased spontaneous motor ac-tivity, immobility and production of hypothermia and analgesia [14]. TheFAAH enzyme has been reported to hydrolyse (2) four times faster than itshydrolysis of AEA [170]. In addition, it has been proposed that the majorenzymatic hydrolysis pathway of (2) occurs via the monoacylglycerol lipaserather than FAAH [171, 172].Compound (2) has been reported to induce a rapid transient increase of

intracellular free Ca2+ concentration in NG108-15 and HL-60 cells througha cannabinoid receptor-dependent mechanism [173, 174]. Furthermore, theCa2+ increase in HL-60 cells appeared to be CB2 mediated as the effectcould be blocked with a CB2 antagonist but not a CB1 antagonist [174]. Verylittle work has been carried out on the SAR of this endocannabinoid. Ingeneral, for the CB1 receptor, arachidonic acid was found to be the preferredfatty acid moiety, although some partially saturated structures had almostcomparable activities [173]. It appears that the presence of a double bond atthe D5-position is crucial for activity. With respect to the head group, the2-glycerol isomer is preferable over both the 1- and 3-analogues. The estermoiety can be replaced by a ketone but agonistic activity in NG108-15 cellsdrops approximately 100-fold [175]. With regard to the CB2 receptor, muchof the SAR overlaps with the results obtained for the CB1 receptor with theexception that glycerol esters of C22 fatty acids do not show appreciableactivity in the CB1 assay, but do show appreciable activity in the CB2 assay[173].

Noladin ether

Noladin ether (3) was recently isolated from porcine brain [16] and found tobind to the CB1 receptor (K i ¼ 21:2 nM), to bind weakly to the CB2 receptor(Ki>3 mM) and it causes typical cannabinoid-like effects such as sedation,hypothermia, intestinal immobility and mild antinociception in mice [16].This endocannabinoid had previously been synthesised independently byboth Mechoulam and co-workers [176] and Sugiura et al. [173]. SAR studiesof this endocannabinoid are lacking in the literature, however, a recentpublication highlighted the importance of the tetra-unsaturated C20 chain

Page 41: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 247

for high affinity at the CB1 receptor, increased saturation or replacement ofthe double bonds (with, for example, a cyclopropyl) had a detrimental effecton binding affinity [177].

INDOLE AND ITS DERIVATIVES

Indoles

The discovery and cloning of the CB1 and CB2 receptors has opened the wayfor the pharmaceutical industry to identify cannabinoid agonists with com-pletely new templates. However, the first novel class of cannabinoid agonists(i.e. those not derived from THC) was discovered accidentally by theSterling-Winthrop Research Group while studying conformationally re-strained analogues of pravadoline (248) [178]. These pravadoline analoguesdisplayed reduced ability to behave as non-steroidal anti-inflammatoryagents that inhibit cyclooxygenase but increased ability to bind to the CB1

receptor.

O N

OH

Me

Me

Me

C5H11

N

O N

O

O

Me

N

N

O

O

Me

OMe

(270) (254a) (R)-(+) WIN 55,212-2(248) Pravadoline

In this initial study they found that the 4-methoxyphenyl ring could bereplaced by a number of other aromatic rings, in particular, the 1-naphthylring was found to impart good potency. In addition, small substituents inthe indole 2-position were favoured and importantly that potency resided inonly one enantiomer of the aminoalkyl indole (AAI) (see Table 6.21). Onecompound of particular importance synthesised at this time is WIN 55,212-2(254a), a potent agonist at both the CB1 and CB2 receptors. Although thiscompound was not developed as a drug, it has been widely used as a phar-macological tool in cannabinoid research. The Sterling-Winthrop researchgroup [179] went on to carry out an extensive SAR study on unconstrainedaminoalkyl indoles with over 100 analogues synthesised and tested, and

Page 42: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.21 AMINOALKYL INDOLES – SAR OF THE BENZOXAZINE CORE [178]

N

O N

O

O

R

Ar

Cpd. Ar R MVD activity IC50 (nM)a CB1 IC50 (nM)

(249) 4-MeO-C6H4 H 44.5 249(250) 4-MeO-C6H4 Me 123 152(251) 4-MeO-C6H4 Et 28% @ 10 mM 27% @ 1mM(252) 2-F-C6H4 Me 76 1,426(253) 1-Naphthyl H 2 7.37(254) 1-Naphthyl Me 6 5.56(255) 5-Quinolinyl Me 21 132.5(R)-(+)-(254a) 0.43 2.77(S)-(�)-(254b) >10,000 8,002

aMouse vas deferens inhibitory activity.

RECENT PROGRESS IN CANNABINOID RESEARCH248

extended their previously reported SAR to include variations in the natureof the aminoalkyl substituent, the substituents on the naphthyl group andsubstituents on the indole nucleus. In general, similar trends were seen withregard to the key structural features required for cannabinoid activity i.e.an aromatic (bi)aroyl ring in the 3-position, a small substituent in the2-position and a cyclic aminoethyl substituent in the 1-position.Following on from this they demonstrated that the aminoethyl side chain

could also be replaced (see Table 6.22) [180], the most potent ligands bearinga naphthoyl moiety. In particular, the piperidinyl analogue (258) was foundto have a high affinity in the binding assay and potency in the mouse vasdeferens assay.In an attempt to rationalise these results with a pharmacophore model

that would fit both the classical cannabinoids and AAIs, Huffmann and co-workers [181] suggested that the ketonic oxygen of WIN 55,212-2 wasaligned with the phenolic hydroxyl of (67). In this arrangement, the naph-thyl moiety of the AAIs would overlay the cyclohexyl and pyran rings of theclassical cannabinoid structure. The indole nitrogen and substituent at-tached to it would then be placed in a corresponding position to the alkylchain on C3. To test this hypothesis, Huffman designed a number of AAIsin which the aminoethyl chain was replaced with other substituents and in

Page 43: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.22 AMINOALKYL INDOLES – SAR OF THE N-1 SUBSTITUENT [180]

NR'

O

R

Cpd. R R0 MVD activity IC50 (nM) CB1 IC50 (nM)

(256)

N

Me

-CH2

Me 2.4 6.5

(257)

N-CH2

Me

Me 1.2 5.4

(258)

N-CH2

Me

H 0.47 1.2

J. ADAM ET AL. 249

particular, carbon chains. Short side chains resulted in inactive compounds,whereas chains with four to six carbons produced best results [181] (seeTable 6.23). In particular, the n-pentyl substituent (263) was optimal ex-hibiting 2-fold greater affinity than WIN 55,212-2 and 4-fold greater affinitythan (67) in the in vitro binding assay. In addition, (263) also displayedpotent in vivo activity.Following on from this, and to further exemplify this pharmacophore

model, Huffman [182] described a novel hybrid structure that combined thehydroxydibenzopyran ring of THC and the indole moiety of the AAIs intoone molecule. It was found that the hybrid molecule (270) had a similaraffinity (19 nM) for the CB1 receptor in vitro as (67) (41 nM). The compoundwas also active in vivo in the mouse tetrad model of cannabimimetic activityand had comparable potency to (67) [182].An alternative binding model has been proposed by Xie et al. [183] using

2D-NMR spectroscopy and molecular modelling with (254a) and 9-nor-9b-OH-hexahydrocannabinol (HHC). In this case it is suggested that the ke-tonic group of (254a) corresponds to the phenolic hydroxy group, thenaphthyl moiety to the C3 alkyl side chain and the nitrogen of the morph-olinyl group to the C9 hydroxyl of HHC. A similar structural alignment has

Page 44: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.23 AMINOALKYL INDOLES – N-1 ALKYL CHAIN SUBSTITUENTS [181]

NMe

O

R

Mouse ED50 (mmol/kg)

Cpd. R CB1 Ki (nM) SA TF RT

(259) Me >10,000(260) Et 1,180(261) n-Pr 164 18.7 84.7 99.1(262) n-Bu 22 2.6 0.23 4.1(263) n-Pentyl 9.5 0.7 0.25 4.3(264) n-Hexyl 48 o2.7 9.5 17.1(265) n-Heptyl >10,000 117 >261 >261(266) 4-Pentenyl 38 3.5 0.34 5.6(267) 2-Phenylethyl 1,250 No maxa No maxa No maxa

(268) c-Hexylethyl 46 55.4 58.7 69.7(269) c-Propylmethyl 140 6.5 7.1 36.9(254a)b 24 0.19 1.4 1.5(67)c 41 0.92 2.7 2.5

a‘‘No max’’ indicates that the compound produced only slightly greater than 50% of thepresumed maximal effect.bWIN 55,212-2.cD9-THC.

RECENT PROGRESS IN CANNABINOID RESEARCH250

been proposed by Razdan and co-workers [184]. To test the hypothesis, aseries of 4-substituted indoles was prepared containing the key structuralfeatures of the AAIs except that the naphthyl substituent was transposedfrom position 3 to position 4 via an oxygen linker. A total of nine com-pounds were synthesised and tested. Alkyl chain-substituted compoundsfailed to show any affinity either in vitro or in vivo (see Table 6.24(271)–(273)). On the other hand, introduction of a naphthyl moiety eitherdirectly or via a number of different linkers (274)–(279), in particular, aketomethylene group (277), led to compounds with moderate potency invitro and in vivo [184].The validity of pharmacophore models that overlay the classical can-

nabinoids and AAIs has been brought into question by recent mutation workon the CB1 receptor. It was found that mutation of Lys-192 in the third

Page 45: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.24 AMINOALKYL INDOLES – SAR 4-SUBSTITUTED INDOLES [184]

N

OR

N

O

Mouse ED50 (mg/kg)

Cpd. R CB1 Ki (nM) SA TF RT

(271) CH(CH3)C5H11 >10,000 >100 >100 >100(272) (CH2)8CH3 >10,000 >100 >100 >100(273) CH(CH3)(CH2)3Ph >10,000 >100 >100 >100(274) CH2-1-Naphthyl 221 3.5 3.3 12.3(275) CH2-2-Naphthyl 1,300 >30 >30 >30(276) CH2CO-1-Naphthyl 287 4.6 8.4 >30(277) CH2COCH2-1-Naphthyl 127 6.1 1.7 18.7(278) (CH2)3-1-Naphthyl 2,220 >30 >30 >30(279) (CH2)4-1-Naphthyl 3,390 55 78 79(254a)a 8.7 0.1 0.4 12(67)b 41 1.0 1.4 1.4

aWIN 55,212-2.bD9-THC.

J. ADAM ET AL. 251

transmembrane domain to an Ala resulted in a complete loss of binding forthe classical cannabinoid (165), in contrast to this; binding of (254a) was onlyslightly affected [185]. These results would suggest that the AAIs interact withthe CB1 receptor in a somewhat different fashion from the dibenzopyrancannabinoids.One alternative that has been proposed is that the main binding arrange-

ment of the AAIs to the receptor is through aromatic stacking. To explorethis proposal a number of naphthyl methanes have been prepared [186].These compounds lack the ketone functionality that is thought to play thesame role as the phenoxy group in the classical cannabinoids. Hence there isno viable possibility for hydrogen-bonding interactions between the receptorand the ligand at this position. Substitutions in both the 4-position of thenaphthyl and the 2-position of the indole were explored (see Table 6.25). Inthe case of the unsubstituted indole, binding affinity was maintained withonly a small drop in binding affinity being observed compared to theirnaphthoyl congeners (280)–(282). In contrast to this, substitution in the

Page 46: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.25 AMINOALKYL INDOLES – SAR OF SUBSTITUTED NAPHTHYLMETH-ANES [186]

R'

N

C5H11

R

Cpd. R R0 CB1 Ki (nM) Cpd. R R0 CB1 Ki (nM)

(280) H H 22 (284) Me Me 127(281) H Me 23 (285) Me OMe 323(282) H OMe 17 (254a)a 9.9(283) Me H 151 (67)b 41

aWIN 55,212-2.bD9-THC.

RECENT PROGRESS IN CANNABINOID RESEARCH252

2-position of the indole had a marked effect on binding, with affinity drop-ping up to 19-fold (cf. (282) and (285)).The high binding affinities obtained for the unsubstituted indole com-

pounds thus support the hypothesis that H-bonding may not be the mainbinding feature of the AAIs and potentially aromatic stacking may be thedominant interaction. To explain the sudden drop in potency for the2-methyl substituted indoles, the authors initiated a molecular modellingand receptor-docking study. From this study, they conclude that the lossin binding affinity is due to the loss of an aromatic stacking interaction[186].More recently, the utility of the indole group as a scaffold for cannabinoid

agonists has been demonstrated by a number of new patent applicationsappearing in the literature (286)–(290) [187–190]. Of particular note is com-pound (286) that is reported to have 18-fold selectivity for the CB1 receptor(CB1: K i ¼ 0:08 nM; CB2: K i ¼ 1:44 nM). In addition to the indole scaffold,a number of patent applications by AstraZeneca claim indole-like scaffoldssuch as benzimidazoles (289) [191–193] and azaindoles (290) [194]. Althoughthese compounds bind to both CB1 and CB2 receptors, the inventors claimthat they may be useful in treating diseases without the associated CNS sideeffects.

Page 47: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 253

NN

O

NN

Ph

N

O

(CH2)5F

I

N

NH

O

OMe

O

Ph

N

O

MeO(288)(286) (287)

N N

Ph

t-BuCONH

OEt

N

Nt-Bu

O

(F3CCH2)2N

(290)(289)

Pyrrole and its derivatives

In Huffman’s [181] overlay model of the classical cannabinoids and AAIs,the benzenoid portion of the indole ring did not participate in the overlapand only the pyrrole ring was involved. In order to investigate this, a ho-mologous series of pyrroles was prepared and their pharmacology examined(see Table 6.26). In analogy to the indoles, the best results were obtainedwith the n-pentyl analogue (295), although this compound was approxi-mately 10-fold less potent than its indole analogue.Following on from this Tarzia et al. [195] designed and synthesised a

range of pyrrole compounds based on the Huffman overlay. A range ofsubstituents was explored around the pyrrole ring, the selections are detailedin Table 6.27. As seen with the AAIs, replacement of the naphthyl moietywith a monocyclic aromatic ring was detrimental to affinity. In particular,replacement with a phenyl ring resulted in complete loss of activity (299).A similar result was seen when the naphthyl moiety was replaced with analiphatic chain (301). Replacement of the ketone group with an amide istolerated, albeit with a loss in binding affinity. In addition, depending on the

Page 48: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.26 PYRROLE DERIVATIVES – N-1 ALKYL CHAIN SUBSTITUENTS [181]

N

R

O

Mouse ED50 (mg/kg)

Cpd. R CB1 Ki (nM) SA TF RT

(291) Me >10,000 106 No maxa 53.3(292) Et >10,000 84 No maxa 77.2(293) n-Pr >10,000 86 81.8 90.1(294) n-Bu 666 No maxa No maxa >108(295) n-Pentyl 87 3.6 1.2 78.8(296) n-Hexyl 399 8.8 9.6 62.8(297) n-Heptyl 309 11 9.7 52.1(254a)b 24 0.19 1.4 1.5(67)c 41 0.92 2.7 2.5

a‘‘No max’’ indicates that the compound produced only slightly greater than 50% of thepresumed maximal effect.bWIN 55,212-2.cD9-THC.

Table 6.27 PYRROLE DERIVATIVES – SAR OF SUBSTITUTED PYRROLES [195]

N

n-C5H11

MeMe

COR1

R2

Cpd. R1 R2 CB1 Ki (nM) CB2 EC50 (nM)

(298) 1-Naphthyl H 45 10(299) Ph H >1,000 >1,000(300) 1-Naphthyl Br 13 6.8(301) HO(CH2)3 H >3,000 >10,000(302) o-(Ac)C6H4NH H 367 >1,000(303) c-HexylNH H 415 483

RECENT PROGRESS IN CANNABINOID RESEARCH254

Page 49: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 255

amide substitution an interesting selectivity profile was seen, for example,the substituted aniline compound (302) gave a moderate binding affinity atthe CB1 receptor (367 nM) but lost all affinity at the CB2 receptor(>1,000 nM). In contrast, the aminocyclohexyl amide (303) retained affin-ity at both receptors albeit reduced compared to the naphthyl analogues.The best results were obtained with the 4-bromo substituted compound(300) giving good affinity at both the CB1 (13 nM) and CB2 (6.8 nM) re-ceptor. The substituted pyrrole (300) was also tested for intrinsic activity atthe CB1 receptor using [35S]-GTPgS binding in rat cerebellar membranesand found to behave as a full agonist with an EC50 value of 140 nM.The pyrazole scaffold is usually associated with cannabinoid antagonists/

inverse agonists and in particular, rimonabant (382) (see below). However,recent work examining a series of (382) analogues for antagonist activitygave rise to a number of compounds that also displayed cannabimimeticeffects in vivo [196]. In particular, replacement of the N-piperazine in (382)with carbon chains that more directly correspond to the lipophilic side chainof (67) gave compounds exhibiting agonistic properties in the mouse triadmodel of cannabimimetic activity (see Table 6.28). The best results wereobtained with 2-fluoroethyl amide (306) that had a relatively low bindingaffinity (852 nM), but had a moderate potency in vivo [approximately 5-foldless active than (67)].

Indenes

In the early clinical trials of Pravadoline (248), the Sterling-Winthrop Re-search group observed CNS side effects. In an effort to circumvent theseissues, indenes were explored as an alternative core [197]. The rationalebehind this approach came from the observation that sulindac (308), anindene analogue of indomethacin (309), had anti-inflammatory propertiescomparable to (309) but without the CNS-associated side effects.

SOMe

Me

CO2H

F

NMe

CO2H

MeO

O Cl

(308) Sulindac (309) Indomethacin

Page 50: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.28 PYRROLE DERIVATIVES – SAR OF SUBSTITUTED PYRAZOLES [196]

NN

Cl

Cl

Cl

Me CONHR

Mouse ED50 (mg/kg)

Cpd. R CB1 Ki (nM) SA TF RT

(304) n-Pentyl 32 11 21 11(305) n-Heptyl 48 27 20 12(306) (CH2)2F 852 7a 13 8(307) n-Pr 167 9 24 10(382)b 6.2 >30 >30 >30(67)c 41 0.92 2.7 2.5

aEstimated ED50 – non-linear dose–effect curve.bRimonabant.cD9-THC.

RECENT PROGRESS IN CANNABINOID RESEARCH256

A series of 11 compounds was prepared with and without a 2-Me subs-tituent. Owing to the method of preparation, the 2-Me substituted com-pounds were prepared as a mixture of stereoisomers (4:1; E:Z), whereas theunsubstituted compounds had predominantly E configuration (>95%). Itwas found that, as with the AAIs, naphthyl-substituted derivatives werepotent agonists in vitro and in vivo (see Table 6.29). Following on from thisReggio et al. [198] extended this work by preparing the stereo-defined in-denes in a bid to identify the bioactive conformation of the AAIs. Throughconformational analysis it had been established that the indene E-isomermimics the s-trans conformation of (254a) while the Z-isomer mimics thes-cis conformation (Figure 6.2). Measurement in CB1/CB2 binding assaysrevealed that the E-naphthylindene isomer had significantly higher bindingaffinity than the Z-isomer (see Table 6.29). This would then suggest that thes-trans conformer of (254a) and related AAIs is the bioactive conformer atthe cannabinoid receptors.

Page 51: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.29 INDENES – SAR OF SUBSTITUTED INDENES

N

O

R

R'

Cpd. R R0 CB1 IC50 (nM) CB2 Ki (nM) Ref.

(310) Ph H, >95% E 33%@1mM [197](311) 1-Naphthyl H, >95% E 1 [197](312) 1-Naphthyl Me, 4:1 E:Z 10 [197](313) 1-Naphthyl H, 100% E 2.72 2.72 [198](314) 1-Naphthyl H, 100% Z 148 132 [198](315) 1-Naphthyl Me, 100% E 2.89 2.05 [198](316) 1-Naphthyl Me, 100% Z 1945 658 [198](254a)a 5.56 [197]

2.48 0.28 [198]

aWIN 55,212-2.

N

O

N

O

O

N

O

N

O

O

N

O

N

O

s-cis s-trans Z-isomer E-isomer

Fig. 6.2 Conformational analysis of indene isomers.

J. ADAM ET AL. 257

Page 52: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH258

Other structures

A number of new scaffolds unrelated to the classical/non-classical cannabinoidsand AAIs have been reported in the literature to deliver cannabinoid agonists.Bayer [199–203] has claimed in a series of patents a number of aryl

sulfonyl esters as cannabinoid agonists for the treatment and prophylaxis ofneurodegenerative diseases. Following on from this a number of publica-tions detailing the in vitro and in vivo profiles on two of these compounds,BAY 38-7271 (317) and BAY 59-3074 (318), have been published.Compound (317) was found to have a high affinity for rat and human CB1

receptors (K i ¼ 0:46 and 1.09 nM, respectively), as well as for recombinanthuman CB1 and CB2 receptors (K i ¼ 1:85 and 5.96 nM, respectively) andalso exhibited full agonist activity using the [35S]-GTPgS technique with anEC50 value at human CB1 receptors of 15.8 nM (no data reported for theCB2 receptor). (317) also displayed typical cannabinoid-like behaviour inthe rat hypothermia model and drug discrimination assays which could bereversed by the selective CB1 antagonist, rimonabant (382). In addition,(317) also proved effective in a rat traumatic brain injury model reducinginfarct volume by 70% at 100 ng/kg/h infusion starting immediately aftersubdural haematoma. It also reduced infarct volume (27% at 1,000 ng/kg/h)in a rat model of focal cerebral ischaemia. Phase I studies concluded that(317) was safe and well tolerated when administered intravenously for either1 or 24 h in humans and (317) is currently being developed for the treatmentof traumatic brain injury and cerebral ischaemia [204, 205].The structurally related compound BAY 59-3074 (318) also has high

affinity at human CB1/CB2 receptors (K i ¼ 48:3 and 45.5 nM, respectively).However, and in contrast to (317), (318) exhibited partial agonist activity athCB1 using the [35S]-GTPgS technique with an EC50 of 142.7 nM [206, 207].The compound also behaved as a partial agonist at hCB2 receptors with anEC50 of 15.8 nM. BAY 59-3074 (318) also displayed typical cannabinoidactivity in vivo in rat hypothermia and drug discrimination assays that couldbe reversed by the selective CB1 antagonist rimonabant (382). At doses of0.3–3mg/kg p.o., (318) induced antihyperalgesic and antiallodynic effectsagainst thermal or mechanical stimuli in rat models of chronic neuropathyand inflammatory pain. The antiallodynic efficacy was maintained for 2weeks of daily administration, and no withdrawal symptoms were seenduring the 2 days after the last dosing regimen [206, 207].Novartis AG has filed a patent application on novel naphthalene deriv-

atives as potent cannabinoid agonists, especially at the CB1 receptor [208].One compound was specifically claimed, the naphthalene derivative (319),which exhibited CB1 binding with a Ki value of 15 nM. This compound wasalso active in an in vivo model of neuropathic pain, reversing hyperalgesia

Page 53: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 259

caused by partial ligation of the sciatic nerve in Wistar rats after oral dosingwith an ED50 of 0.18mg/kg.In addition, Novartis filed a patent application on a series of quinazolines

as cannabinoid agonists [209]. Compound (320) is one of the two com-pounds specifically claimed and exhibited CB1 and CB2 binding with Ki

values of 34 and 11 nM, respectively. It was shown to be a full agonist at theCB1 receptor with an EC50 of 132 nM (no functional data for the CB2

receptor). Compound (320) was also active in the neuropathic pain modeldescribed above with an ED50 of 0.5mg/kg after oral dosing.AstraZeneca has filed a patent application on novel bis-aryl compounds

as CB1/CB2 agonists that lack CNS penetration and thus avoid the un-wanted side effects associated with activation of central CB1 receptors [210].Over 100 compounds are specifically claimed e.g. (321). Compounds weretested in receptor binding assays using human CB1 and CB2 receptor prep-arations. Respective Ki values were in the ranges 50–5,000 and 15–2,800 nM,although no specific data were presented.Shionogi [211] has filed a patent application on a series of thiazine de-

rivatives; no compounds are specifically claimed. The thiazine derivative(322) was active in vivo in the formalin-induced licking-and-biting model inICR mice with an ED50 of 1.5mg/kg after oral dosing.

O OSO2(CH2)3CF3

HO

OCN

F3C OSO2(CH2)3CF3 O

On-C5H11

(317) BAY 38-7271 (318) BAY 59-3074 (319)

O

HO(CH2)2NHN

N

O

OSO2NHMe

Me

Me

EtO2C

Me

N

Me OH

Ph

N

S N

S

SMe

(320) (321) (322)

CB2 AGONISTS

Receptor subtype selective CB2 agonists are seen as potential candidates forthe treatment of a variety of diseases, including pain-related indications. Thepromise of useful therapeutic effects without unwanted CNS side effectsmakes the development of CB2 selective compounds a particularly attractive

Page 54: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH260

proposition. Several classes of CB2 selective agonists, including classicalcannabinoids, resorcinols, indoles, indazoles and other heterocyclic deriv-atives and their SAR studies have been reported.

CB2 SELECTIVE CLASSICAL CANNABINOIDS

Of the three main pharmacophoric elements in the classical cannabinoidtemplate discussed previously, the phenolic hydroxyl group at C1 has beenshown to be the most important for CB2 selectivity. In 1996, a group atMerck Frosst [212] and the Huffman group [213] independently demon-strated that removal of this hydroxyl group in the 10,10-dimethylheptyl seriesleads to compounds with high CB2 receptor affinity and between 8- and 40-fold selectivity for CB2 over CB1 binding affinity (Table 6.30). The MerckFrosst paper further describes modification of the phenol to a methoxygroup, resulting in higher CB2 selectivity [212].

O

R1

MeMe

n-C6H13

Me Me

R2

(323) R1 = Me, R2 = H(324) R1 = CH2OH, R2 = H(325) R1 = Me, R2 = OMe

The CB2 receptor has been shown to tolerate shorter C3 side chains thanthe CB1 receptor and the Huffman group has exploited this in combinationwith C1 modifications to develop highly selective CB2 receptor ligands (Ta-bles 6.31 and 6.32) [214, 215].The 11-hydroxy analogues of the 1010-dimethyl compounds in Tables 6.31

and 6.32 were also prepared by the Huffman group. In general, theseshowed higher affinity for both CB1 and CB2 receptors than the simplemethyl analogues, but reduced CB2 selectivity (data not shown) [215].

Table 6.30 1-DEOXY AND 1-METHOXY D8-THC DERIVATIVES

Cpd. CB1 Ki (nM) CB2 Ki (nM) CB1/CB2 Ref.

(85) D8-THC-DMH 0.83 0.49 1.7 [212](323) 1-Deoxy-D8-THC-DMH 250 21 12 [212]

23 2.9 8 [213](324) 1-Deoxy-11-hydroxy-D8-THC-DMH 1.2 0.032 38 [213](325) 1-Methoxy-D8-THC-DMH 15,850 20 793 [212]

924 65 14 [214]

Page 55: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.31 1-DEOXY D8-THC C3 SIDE-CHAIN ANALOGUES

O

Me

MeMe R

Cpd. R CB1 Ki (nM) CB2 Ki (nM) CB1/CB2 Ref.

(326) 1,1-Dimethylethyl 2,150 58 37 [214](327) 1,1-Dimethylpropyl 2,290 14 164 [214](328) n-Bu 2,790 54 52 [214](329) 1,1-Dimethylbutyl 677 3.4 199 [214](330) n-Pentyl (1-Deoxy-D8-THC) >10,000 32 >312 [214](331) 1,1-Dimethylpentyl 399 10 40 [214](332) n-Hexyl 1,610 273 6 [214](333) 1,1-Dimethylhexyl 295 19 16 [214](323) 1,1-Dimethylheptyl (DMH) 23 2.9 8 [213](334) 1,1-Dimethyloctyl 51 76 0.7 [214](335) 1,1-Dimethylnonyl 178 449 0.4 [214]

Table 6.32 1-METHOXY D8-THC C3 SIDE-CHAIN ANALOGUES [215]

O

Me

MeMe R

OMe

Cpd. R CB1 Ki (nM) CB2 Ki (nM) CB1/CB2

(336) 1,1-Dimethylethyl >10,000 1,867 >5.4(337) 1,1-Dimethylpropyl >10,000 1,404 >7.1(338) 1,1-Dimethylbutyl >10,000 325 >31(339) 1,1-Dimethylpentyl 4,001 43 93(340) 1,1-Dimethylhexyl 3,134 18 174(325) 1,1-Dimethylheptyl (DMH) 713 57 12

J. ADAM ET AL. 261

Page 56: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH262

Compounds where the C1 hydroxyl group is linked to the C2 position bya cyclic ether link, with a range of different groups in the C9 position, wereprepared by Reggio et al. [216]. These compounds showed only modest CB2

selectivity. Compound (341) was the most potent and selective with a bind-ing affinity (Ki) of 5.8 nM at CB2 and 26 nM at CB1.A patent application from Merck Frosst, published in 1997, discloses CB2

selective compounds with modified core structures where the pyran oxygenis replaced by nitrogen, such as (342), which has a Ki of around 64 nM atCB2 and around 25,000 nM at CB1 [217].

O

OH

MeMe n-C5H11

O

N

Me

MeMe OMe

Me(341)

(342)

INDOLES AND INDAZOLES

Indomethacin morpholinyl amide (343) was found as a weak but CB2 se-lective agonist through a topological similarity search using WIN 55,212-2(254a) as the template [218]. The binding affinity (Ki) of (343) was 435 and>20,000 nM for hCB2 and hCB1, respectively. A subsequent SAR studyrevealed that N-(2,3-dichlorobenzoyl) and N-naphthoyl indole derivativesshowed improved activity. Further optimisation of the substituents on theindole C3-position was also performed, and the more selective and potentCB2 agonists, L-768,242 (344) and L-759,787 (345) were discovered. Thesecompounds showed >100-fold CB2 selectivity with significant potency (Ki

of 12 and 8.5 nM, respectively).

N

X N

R2 O

Me

O

R1

(343) X = -CH2CO-, R1 = OMe, R2 = 4-Cl-phenyl

(344) L-768,242 X = -(CH2)2-, R1 = OMe, R2 = 2,3-diCl-phenylKi(hCB1) = 1917nM, Ki(hCB2) = 12nM

(345) L-759,787 X = -CH2-, R1 = H, R2 = 1-naphthylKi(hCB1) = 877nM, Ki(hCB2) = 8.5nM

Page 57: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 263

During the study of N-alkylindole derivatives as cannabinoid ligands,JWH-015 (346) was found as a potent CB2 selective agonist, Ki (CB2)13.8 nM with modest selectivity, ðCB1Þ : ðCB2Þ ¼ 28 [81]. For the optimi-sation study of (346), more than 40 indoles were prepared and their CB1 andCB2 receptor affinities were determined [219]. In most cases, N-pentyl in-doles showed significant activity but low receptor subtype selectivity,whereas N-propyl derivatives had better selectivity for CB2. The SAR of thesubstituents on the naphthyl ring was also studied, and it is noteworthy thatintroduction of a methoxy group in the 2-position of the naphthyl ringreduced the binding affinity only for the CB1 receptor, even though thecompound bears a pentyl group on the indole ring. As a result, three CB2

selective compounds, JWH-120 (347), JWH-151 (348) and JWH-267 (349)were found; JWH-151 was a full agonist whereas JWH-120 and JWH-267exhibited partial activity in GTPgS binding assay.

N

O

R1

MeR

2

R3

R4

(346) JWH-015 R1 = n-Pr, R2 = R3 = R4 = H Ki(CB1) = 383nM, Ki(CB2) = 13.8nM

(347) JWH-120 R1 = n-Pr, R2 = R4 = H, R3 = Me Ki(CB1) = 1054nM, Ki(CB2) = 6.1nM

(348) JWH-151 R1 = n-Pr, R2 = R3 = H, R4 = OMe Ki(CB1) >10,000nM, Ki(CB2) = 30nM

(349) JWH-267 R1 = n-C5H11, R2 = OMe, R3 = R4 = H Ki(CB1) = 381nM, Ki(CB2) = 7.2nM

The group of Bristol-Myers Squibb found a C3 amido-indole (350) as alead compound for their CB2 agonist program [220]. Compound (350)demonstrated moderate binding affinity for CB2 (Ki 250 nM), which wasimproved dramatically to a Ki of 8 nM by the introduction of methoxygroup on the C7 of the indole core. The 7-methoxy indole (351) showedgood receptor subtype selectivity, CB1 : CB2 ¼ 500, however, the methylester on the phenylalanine moiety was susceptible to microsomal hydrolysis.In an effort to discover non-ester CB2 agonists, compound (352), having a(1S)-fenchyl group instead of the phenylalanine methyl ester, was prepared.The introduction of a larger alkyl group onto the C2 position of the indolering decreased the CB2 binding affinity (H>Me>Et>n-Pr; Table 6.33).The non-substituted compound (353) showed dose-dependent inhibitoryactivity in an in vivo anti-inflammatory model [lipopolysaccharide (LPS)-induced TNF-a production in mice] after i.v. administration with an ED50

of 5mg/kg. Unfortunately, (353) was inactive after oral administration, and

Page 58: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.33 1-AMINOALKYLINDOLE ANALOGUES

N

NH

O

NO

R

Me

Me

Me

OMe

Cpd. R CB1 Ki (nM) CB2 Ki (nM) CB1/CB2 Ref.

(352) Me 10,000 30 333 [220](353) H 245 11 22 [221](354) Et 103 [220](355) n-Pr 6% @ 100nM [220]

RECENT PROGRESS IN CANNABINOID RESEARCH264

the poor oral efficacy was thought to be caused by metabolical instability,e.g. de-alkylation at the N1 position.

N

NH

O

N

O

Ph

CO2Me

Me

R

(350) R = H, Ki(CB2) = 250 nM

(351) R = OMe, Ki(CB2) = 8nM, CB1/CB2 = 500

It was hypothesised that the increased activity of indoles bearing smallergroups at the C2 position described above would be derived from the con-formational preference of conformer A over conformer B as a result of steric

Page 59: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

N

NH

O

N

O

R

Me

Me

Me

OMe

N

ONH

N

O

R

OMe

MeMe

Me

Amide conformer A Amide conformer B

Fig. 6.3 Structures of amide conformers A and B.

J. ADAM ET AL. 265

effects (Figure 6.3) [221]. Based on this hypothesis, indazoles and indo-lopyridones, such as (356) and (357) were designed. For (356), it was pre-dicted that conformer A would be preferred over conformer B, as the resultof an intramolecular stereoelectronic interaction. Actually, (356) was morepotent than all of the indole derivatives mentioned above. The conforma-tionally constrained indolopyridone (357) had the most potent activity in theseries [K iðCB2Þ ¼ 1:0 nM]. In the in vivo model, (357) exhibited inhibitionactivity against LPS-induced TNF-a release both after i.v. and oral admin-istration. CB2-selective indazole derivatives, such as (358), were also re-ported from the research group of the University of Connecticut [222]. TheKi values of (358) for CB2 and CB1 were 0.15 and 6.84 nM, respectively.Another type of C3 amido-indole, exemplified by (359), was disclosed bySanofi Synthelabo [223].

NN

NO

N

O

Me

Me

Me

OMe

H

N

NO

N

O

Me

Me

Me

OMe

(356) Ki(CB2) = 2 nM (357) Ki(CB2) = 1 nM

Page 60: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH266

NN

NH

O

N

ON

(CH2)3NHSO2MeCl

NHO

Me

MeMe

Me

(358) (359)

N

O

I

NMe

NO2

N

SO

O

F

S

OO

NHSO2Me

Me

(360) AM1241 (361)

AM1241 (360) exhibited high affinity and selectivity for CB2 [K iðCB1Þ ¼

280 nM, Ki ðCB2Þ ¼ 3:4 nM]. (360) Dose dependently inhibited experimentalneuropathic pain in a spinal nerve ligation-induced tactile and thermal hyper-sensitivity model [224]. Other indole derivatives bearing sulfonamide moietieson the side chain, such as compound (361), were disclosed [225]. Though 67derivatives including pyridyl and other heteroaromatics instead of the indolecore were listed, no specific biological data were shown.

RESORCINOL DERIVATIVES

As previously discussed, phenol derivatives bearing a six-membered carbo-cyclic ring next to the hydroxy group of the phenol represent a major classof cannabinoid ligands. These compounds correspond to pyran ring-openedanalogues of (67). Several efforts have been made to find CB2 selectiveagonists among the series.

Page 61: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 267

MeO

CH2OH

OMe

n-C6H13

Me Me

Me

Me

HU-308 (362)

One of the conspicuous resorcinols is HU-308 (362), which is a CB2-specific agonist; this compound does not bind to CB1 (Ki>10 mM), but has asignificant affinity for CB2 (K i ¼ 22:7 nM) [226]. HU-308 elicited analgesicactivity in a formalin-induced peripheral pain model and an anti-inflam-matory effect on arachidonic acid-induced ear inflammation, though itshowed no activity in a tetrad of behavioural tests, which are linked to CNSactivity (Table 6.34).In order to study the SAR of the resorcinol derivatives, about 40 com-

pounds were synthesised and tested in biological assays [227]. The 2-cyclohexylresorcinol derivative, O-1422 (363), showed binding affinity forboth CB1 and CB2 receptors, with a moderate CB2 selectivity [K iðCB1Þ ¼

11 nM and K iðCB2Þ ¼ 1:5 nM]. Reducing the ring size of the carbon ring tocyclopentyl (364) decreased CB1 affinity by 9-fold and CB2 affinity by5-fold, though increasing the ring size to cycloheptyl (365) did not affect thebinding affinity. Introduction of a methyl group at the 2- or 3-position of

Table 6.34 RESORCINOL DERIVATIVES [227]

R

OH

OH

n-C6H13

Me Me

Cpd. R CB1 Ki (nM) CB2 Ki (nM) CB1/CB2

(363) O-1422 c-Hexyl 11 1.5 7.3(364) O-1424 c-Pentyl 95 7 14(365) O-1656 c-Heptyl 18 2 9.0(366) O-1658 2-Methyl c-hexyl 16 1 16(367) O-1657 3-Methyl c-hexyl 14 0.8 18(368) O-1659 4-Methyl c-hexyl 45 5 12

Page 62: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.35 CYCLOHEXYL RESORCINOL DERIVATIVES [227]

MeO

OMe

n-C6H13

Me Me

MeR

Cpd. R CB1 Ki (nM) CB2 Ki (nM) CB1/CB2

(369) O-1999 H >10,000 466 >21(370) O-1966A OH 5,055 23 220

RECENT PROGRESS IN CANNABINOID RESEARCH268

the cyclohexyl ring did not dramatically alter the affinity. However, movingthe methyl group to the 4-position (368) decreased affinity for both can-nabinoid receptors. Conversion of the two phenolic hydroxy groups of (367)to methoxy groups (369) reduced both CB1 and CB2 binding activity, CB1

affinity was decreased more than 700-fold. Further efforts to find more CB2

selective agonists in the dimethoxyphenyl series resulted in discovery of O-1966A (370), which has good affinity and selectivity for CB2 [K iðCB1Þ ¼

5; 055 nM and K iðCB2Þ ¼ 23 nM, respectively] (Table 6.35).

OH

O

OH

(CH2)4

Me Me

MeMe

AM1703 (371)

Another resorcinol derivative, AM 1703 (371), which has a carbon–car-bon triple bond at the end of the alkyl chain, was reported to be a highlypotent and selective CB2 agonist (K iðCB2Þ ¼ 0:59 pM, with 500-fold selec-tivity against CB1) [228].

BENZO[C]CHROMEN-6-ONE DERIVATIVES

Benzo[c]chromen-6-one derivatives, such as AM 1710 (372), were found tobe CB2 selective agonists [229]. Although these compounds are structurallysimilar to the THC derivatives, the dimethyl group on 6-position of the

Page 63: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 269

THCs is converted to a carbonyl group and the C-ring is aromatised. Speciesdifferences in CB2 binding activities of these compounds were observed:(372) and AM 1714 (373) showed 10- to 15-fold higher affinities at rat CB2

than human CB2 [230].

OOn-C6H13

Me Me

OH

OR

(372) AM1710 R = Me Ki = 28nM (humanCB2) Ki = 2.0nM (ratCB2)

(373) AM1714 R = H Ki = 29nM (humanCB2) Ki = 1.9nM (ratCB2)

OTHER HETEROCYCLIC CB2 AGONISTS

Benzimidazole-5-carboxamide-based agonists have been reported [231], forexample, compound (374) showed good activity and about 100-fold selec-tivity for CB2 [K iðCB1Þ ¼ 84:8 nM, K iðCB2Þ ¼ 0:8 nM]. Thirty-five relatedcompounds were also disclosed along with (374). Tetrahydroquinolone(375a) also exhibited CB2 agonist activity [232] and the CB2 binding affinitywas 17-fold enhanced by the conversion of the six-membered carbon ring tothe corresponding eight-membered ring (375b). Tetrahydroisoquinoline(376) and its derivatives were reported as CB2 agonists [233]. To estimate theCB2 agonist activity of (376), the inhibition activity on forskolin-stimulatedcyclic AMP production was tested and the IC50 was 0.4 nM.

NH

c-Hep

O

MeMe

N

N

c-Hex

OEt

O

(CF3CH2)2NN

NH(CH2)2Ph

X

O

n-Bu

O

(376) IC50(CB2) = 0.4nM

(374)

Ki(CB1) = 908nM Ki(CB2) = 1.6nM

(375a) X = CH2

(375b) X = (CH2)3

Ki(CB1) = 2851nM Ki(CB2) = 28nM

Page 64: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH270

Several types of CB2 agonists with monocyclic core structures have beenreported, these compounds being structurally distinct from other series ofCB2 agonists. Tetrazine derivative (377) exhibited 90-fold selectivity for CB2

and 2-imino-1,3-thiazolidine (378) showed more than 500-fold selectivity[234, 235]. A pyridone analogue (379) of a bicyclic CB1 agonist, CP 47,497(192) showed CB2 selective activity [K iðCB1Þ ¼ 973 nM, K iðCB2Þ ¼ 56 nM)][236], though the origin of the selectivity was unclear. The pyridine deriv-atives such as (380) were disclosed by Glaxo; (380) was more than 100 timesselective for CB2 over CB1 receptors [237].

N

S

N

OSEt

i-Pr

N

N N

N

CO2Et

CO2Et

Ph

Ph

N

O

OH

n-Pr

n-C5H11

Ki(CB1) = >5000nM

Ki(CB2) = 9nM

(378)

Ki(CB1) = 1672nM

Ki(CB2) = 19nM

(377) (379)

N

NH

O

NNH

Cl

CF3

(380)

THERAPEUTIC APPLICATIONS OF CANNABINOID AGONISTS

Standardised preparations of cannabinoid agonists are available for ther-apeutic use in some countries [238]. Dronabinol (MarinolTM), an oral prep-aration of D9-THC (67), is used clinically as an appetite stimulant in AIDSpatients and an antiemetic in cancer chemotherapy. A synthetic analogue of(67), nabilone (CesametTM), (381), is also used to suppress nausea andvomiting in cancer chemotherapy.There have been a number of studies to evaluate the therapeutic effect of

cannabinoids against spastic disorders, including multiple sclerosis and spinalcord injury. For example, a randomised placebo-controlled trial in more than

Page 65: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 271

600 multiple sclerosis patients with oral cannabis extract or (67) was performedand an objective improvement in mobility was observed in the study [239].

O

O

OH

n-C6H13

Me MeMe

MeO

CH2OH

OH

n-C6H13

Me MeMe

Me

(165) HU-210 (381) Nabilone

Though cannabinoids exhibit a wide range of antinociceptive activities in avariety of animal models, e.g. neuropathic pain [240], hyperalgesia [241] andinflammatory pain [242], there are a limited number of reports in human trialson the use of cannabinoids against pain disorders. One of the early clinicaltrials was a preliminary double-blind study of oral (67) in patients experi-encing cancer pain, which was published in 1975 [243]. While pain relief wasdemonstrated to be significantly superior to placebo after administration of15 and 20mg of (67), some CNS side effects including substantial sedationand mental clouding were observed. A more recent clinical study revealed thatthe maximally tolerated dose of cannabis extract in 40 cancer patients was0.15mg/kg [238]. There have been very few clinical trials of cannabinoids forthe treatment of neuropathic pain and it was reported that an overall benefitof oral dronabinol was not obtained on refractory neuropathic pain becauseof the CNS side effects [244]. One of the potential uses of cannabinoids is toreduce the dosage of morphine administered to treat chronic pain disorders.Holdcroft et al. [245] reported that cannabis extract decreased the require-ment of morphine in a familial Mediterranean fever patient. The CNS sideeffects of cannabinoid agonists are thought to be caused by CB1 receptoractivation in brain. Recently, it was reported that CB2 selective agonistsshowed antinociceptive effects in various pre-clinical pain models includingmodels of hyperalgesia and neuropathic pain without any CNS cannabinoideffects [246]. To date, the mechanism of inhibition of pain responses by CB2

agonists has not been resolved completely, however, an indirect action oninflammatory processes and stimulation of the release of endogenous opioidshave been proposed [247]. An alternative approach is topical administrationof a cannabinoid agonist. Analgesic and anti-hyperalgesic effects of a top-ically applied CB1/CB2 agonist HU-210 (165) to human skin were reported[248] and no psychotomimetic side effects were observed.

Page 66: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH272

It has been revealed that cannabinoids exhibit neuroprotectant activitiesin both in vitro and in vivo models [249]. The neuroprotective effects aremainly based on regulation of transmitter release, modulation of calciumhomeostasis, anti-oxidant properties and modulation of immune responses.A number of neurological disorders, including brain trauma, cerebral is-chaemia, Parkinson’s disease and Alzheimer’s disease represent possibletherapeutic areas for cannabinoids with neuroprotective properties. Can-nabinoids are also suggested to have potential against glaucoma due to theirneuroprotective nature and lowering of intraocular pressure [250].Other therapeutic uses of cannabinoid agonists have been reported. The

potential of cannabinoids as a treatment for asthma is supported experi-mentally. A CB1 agonist, (R)-methanandamide (21), inhibited nerve growthfactor (NGF)-induced airway hyperresponsiveness in vivo [251]. The anti-pruritic effect of cannabinoids has been reported, the action being mediatedby both CB1 and CB2 pathways [252]. Treatment with cannabis extractimproved urinary tract symptoms of multiple sclerosis patients significantlyin an open-label pilot study [253].CB2 selective agonists are thought to be promising agents against a va-

riety of disorders related to the immune system, because of their immuno-modulatory properties and lack of psychotropic effects. Myocardialischaemia-reperfusion injury [254], atherosclerosis [255], glioma [256], leu-kaemia/lymphoma [257] and osteoporosis [258] are suggested as target dis-eases for CB2 agonists. Cough reflex was also inhibited by a CB2 agonistJWH-133 (329) in a guinea pig model of cough [259].

NH

OH

MeO

(CH2)34

n-C4H9

O

Me

n-Pr

Me MeMe

Me

(21) R-Methanandamide

(329) JWH-133

CB1 RECEPTOR ANTAGONISTS

In recent years pre-clinical testing and, in particular, clinical data onSR141716A (Rimonabant, Acomplia) (382) from Sanofi-Aventis (previously

Page 67: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 273

Sanofi-Synthelabo) has stimulated a great deal of interest in the discovery ofcannabinoid CB1 receptor antagonists. This section aims to cover the struc-tures and therapeutic applications of CB1 antagonists published to date.Reviews of CB1 receptor antagonists and their therapeutic applications haverecently appeared in the literature [260–262].

1,5-DIARYL-PYRAZOLES

The most important class of CB1 receptor antagonists identified to date arethe 1,5-diaryl-pyrazoles. This class includes rimonabant (382), first describedin patent applications from Sanofi just over a decade ago [263, 264]. Rimo-nabant has proved invaluable in the elucidation of cannabinoid receptorpharmacology, as described in the section on therapeutic applications below.Several groups have published on structural analogues of (382), one of the

earliest being the disclosure of CP 272871 (383) from Pfizer, which displayslower affinity for the CB1 receptor than (382), in addition to reduced se-lectivity over the CB2 receptor subtype. Both (382) and (383) have beenshown to act as inverse agonists rather than neutral antagonists in vitro[265]. A recently published patent application from Sanofi-Aventis claims aseries of 4-cyanopyrazole analogues of (382), with 42 specific examples [266].Makriyannis and co-workers [267] have published on SAR within the 1,5-

diaryl-pyrazole class of compounds. These studies indicated a requirementfor a para-substituted phenyl ring at the 5-position of the pyrazole, iodo-phenyl proving optimal in terms of potency. Retention of the 3-carboxamidemoiety was important, as was retention of the 2,4-dichlorophenyl substitu-ent at the 1-position. The most potent compound in the series, AM 251(384), showed slightly higher affinity for the CB1 receptor than rimonabantcoupled with excellent selectivity over the CB2 receptor. Furthermore, thepresence of the iodophenyl moiety allows for use of this compound as asingle photon emission computed tomography (SPECT) ligand for imagingstudies [268].SR147778 (385) is a close structural analogue of (382), first described in a

patent application from Sanofi-Synthelabo [269]. In this application, (385) isclaimed to have an improved duration of action over (382) following oraldosing in mice. This was demonstrated through receptor occupancy studiesand the ability of the compounds to reverse cannabinoid agonist-inducedhypothermia. Compound (385) also displayed slightly higher affinity forCB1 receptors than (382) (Ki values of 5.4 and 34 nM, respectively) whileretaining excellent selectivity over CB2 receptors. A subsequent publicationfrom Sanofi-Synthelabo further described the in vitro and in vivo profilesof (385) [270]. The compound reversed WIN 55,212-2-mediated effects

Page 68: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH274

(hypothermia, analgesia and gastrointestinal transit) in mice following bothi.p. and p.o. administration. (385) also reduced ethanol or sucrose con-sumption in mice and rats and food intake in fasted and non-deprived rats.The compound is currently in Phase II clinical trials.

NN

Me

Cl

Cl

R1

NH

O N

NN

NC

Cl

NHPh

O

MeO

NN

Et

Cl

Cl

Br

NH

O N

R

(382) SR141716A (Rimonabant) R1 = Cl

(384) AM 251 R1 = I

(383) CP-272871

(385) R = H, SR147778

(386) R = OH

A close analogue of (385), the 4-hydroxypiperidine analogue (386) wasrecently described in a patent application from Sanofi-Synthelabo [271] andperhaps formed as a metabolite of the parent compound.Martin and co-workers [196] have also published on the discovery and

SAR of pyrazole cannabinoids as described in the CB1 agonist section. Theanalogues were tested for CB1 receptor binding affinity and in a battery of in

Page 69: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 275

vivo tests, including hypomobility, antinociception and hypothermia. Var-iations of the pyrazole substitution pattern in positions 1, 4 and 5 led tocompounds with varying affinity that consistently behaved as antagonists invivo. Introduction of ether moieties into the 3-position of the pyrazole coreled to the identification of both partial agonists and antagonists. For ex-ample, compound O-1043 (387) retained a Ki of 53 nM and demonstratedantagonism in vivo. An analogue, in which the amide was replaced with aketone (O-1271) (388), retained a Ki of 82 nM and again demonstrated an-tagonism in vivo.Makriyannis and Liu claimed a series of pyrazole analogues in a patent

application published in 2003 [272]. Of the 29 compounds specifically ex-emplified in the patent application, compound (389) was demonstrated toreduce lever presses when administered to rats that were trained to expectdelivery of a food pellet as the outcome. It was proposed that the reductionin lever pressing was the result of decreased appetite brought about by CB1

receptor antagonism.

NN

Me

Cl

Cl

Cl

R

NN

R1

Cl

Cl

R2

NH

O N

(387) O-1043 R = OCH2-2,4-diF-phenyl(388) O-1271 R = CO(CH2)4Me

(389) R1 = CH2OH, R2 = 3-pyridyl (390) NIDA-41020 R1 = Me, R2 = OMe

Horti and co-workers [273] published on their studies to produce ana-logues of rimonabant with reduced lipophilicity. The aim of the work was toproduce ligands for positron emission tomography (PET) studies in hu-mans. Highly lipophilic ligands were noted to be a problem due to resultinghigh levels of non-specific binding. An additional limiting factor in the workwas the need to be able to introduce an appropriate atom for a PET la-belling study (18F, 76Br or 11C). NIDA-41020 (390) retained high CB1 re-ceptor affinity (K i ¼ 4:1 nM, (382) K i ¼ 1:8 nM in the same assay format)and a reduced lipophilicity in comparison with (382) (e logDoct values of4.78 and 5.36, respectively).

Page 70: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH276

Several studies have been reported on the application of conformationalrestraint to the 1,5-diaryl-pyrazole series in an attempt to provide com-pounds with modified properties. In one approach, a Sanofi-Synthelabopatent application claimed a series of conformationally restrained com-pounds, exemplified by compound (391). Compounds of the invention werestated to be CB1 receptor antagonists with Ki values below 5� 10�7M andselectivity over CB2 receptors of at least 10-fold [274].Lange and co-workers [275] published on conformationally restrained

analogues of rimonabant (382), again constraining the pyrazole 4-positionand the 5-aryl substituent into a ring system. The highest affinity compoundwithin the series was compound (392), which displayed a pKi value of 7.2((382) pKi 7.6 in the same assay). This compound (392) was found to beslightly more potent than (382) in a cell-based functional assay for CB1

antagonism (pA2 values of 8.8 and 8.6, respectively). In a subsequent pub-lication, Ruiu et al. [276] synthesised and characterised NESS-0327 (393).This compound had also been prepared and tested in the publication fromLange et al. (393) was reported to show higher affinity for the CB1 receptorthan (382) (Ki values of 350 fM and 1.8 nM, respectively), in addition tohigher affinity for the CB2 receptor (Ki values of 21 and 514 nM, respec-tively). Selectivity for the CB1 receptor over the CB2 receptor was calculatedat more than 60,000-fold for (393). Potent antagonism was demonstrated ina variety of in vitro and in vivo paradigms for (393).

NN

Cl

Cl

Cl

NH

O NS

NN

Cl

Cl

NH

O N

R

X

N

NH

O NMe

N

Cl

Cl

(391) (392) X = (CH2)2, R = NO2

(393) NESS-0327 X = CH2, R = Cl

(394)

Thomas and co-workers [277] reported that irradiation of (382) with a450W high-pressure mercury lamp brought about photocyclisation to aconstrained analogue (394). The structure of the product was elucidatedthrough NMR and X-ray diffraction analysis. The compound retained highaffinity for the CB1 receptor (K i ¼ 48 nM) and good selectivity over the CB2

receptor (K i ¼ 3; 340 nM).

Page 71: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 277

A series of 21 compounds, differing from (382) in the amide region, hasbeen synthesised by Franciso et al. [278] and their biological propertiesstudied.The biological activity of the compounds was assessed in two binding as-

says using [3H]-CP 55,940 (see Table 6.36) and [3H]-rimonabant (data notshown), and their functional activity determined using a GTPgS assay. Upon

Table 6.36 MODIFIED AMIDE SUBSTITUENT ANALOGUES OF RIMONABANT(382) – IN VITRO DATA

NN

Cl

Cl

Cl

Me CONHR

Cpd R CB1 Ki (nM) CB2 Ki (nM) [ 35S] GTP–gS EC50 (nM)

(382) Piperidine 6.18 313a 56,300(395) Et 46.3 3,110 30,300(396) n-Pr 29.9 2,960 11,100(397) i-Pr 29.4 1,740 16,000(398) n-Bu 13.4 1,600 8,450(399) i-Bu 11.5 704 7,540(400) n-Pentyl 11.4 1,110 5,270(401) n-Hexyl 18.1 6,870a 29,400(402) c-Hexyl 2.46 228 26,000a

(403) Morpholinyl 22.9 2,400 38,900(404) OH 1,690 7,820a

(405) (CH2)2OH 385 4,270 1.67� 106a

(406) (CH2)3OH 160 1,250 241,000(407) (CH2)4OH 154 5,720a 304,000(408) (S)-(+) CH(CH3)CH2OH 117 5,900 294,000(409) (R)-(�) CH(CH3)CH2OH 117 1,770 292,000(410) NH2 374 121,200 4.19� 105a

(411) NHMe 555 6,660 1.2� 107a

(412) NHEt 143 6,061 4.95� 105a

(413) NHn-Pr 74.8 2,620 105,000(414) NHn-Bu 50.9 2,850 128,000(415) NHi-Bu 41.8 2,190 73,600

aValue greater than the highest point on the displacement curve.

Page 72: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH278

increasing the length of the alkyl chain, the CB1 binding affinity increases withi-butyl and n-pentyl analogues (399) (400) showing the highest affinity. In-creasing the chain length beyond this (e.g. n-hexyl (401)) resulted in a decreasein affinity compared with (399) and (400). In the hydroxyalkylamide series(404)–(409) and the alkylhydrazide series, this trend was also evident. Theintroduction of a hydroxyl group led to a marked decrease in affinity (cf.(395) and (405), (396) and (406)). There was little difference in binding affinityobserved between the two hydroxymethylethyl enantiomers (408) and (409)with the incorporation of the hydroxyl group again proving to have a del-eterious effect on CB1 binding. The morpholinyl analogue of rimonabant(403) again had comparatively low affinity (K i ¼ 22:9 nM), confirming thatthe presence of a polar oxygen was not particularly well tolerated. Within theseries, the most potent side chain was found to be cyclohexyl (402). Theaffinity of (402) was only slightly improved compared with (382), implyingthat the piperidine nitrogen is not likely to be involved in any crucial elec-trostatic or hydrogen-bond formation with the receptor. Throughout the se-ries, the CB2 receptor affinity was modest with CB1/CB2 selectivity rangingfrom 5-fold (404) to 378-fold (401), and all the compounds were found to beinverse agonists. Five compounds with reasonably high CB1 receptor affinities(399)–(403) were investigated for their ability to antagonise the CB1 agonist-mediated inhibition of electrically evoked contraction of mouse vas deferenstissue. All the compounds tested were found to produce antagonism. In linewith the earlier obtained binding data, (402) was found to be the most potentin the vas deferens experiment. Compounds (400), (402) and (403) also dis-played a noticeable inverse effect, enhancing the amplitude of the twitch priorto addition of the cannabinoid agonist WIN 55,212-2, an effect not shown by(399) and (401).A QSAR study was performed to understand the pharmacophoric re-

quirements of the aminopiperidine region. A number of conformations ofeach molecule were generated using a quenched molecular dynamics ap-proach and the resultant conformers overlaid by superimposing theirpyrazole rings using a root mean square minimisation procedure. A com-parative molecular field analysis (CoMFA) approach was then used to de-rive a QSAR and allow visualisation of the regions where changes in stericor electrostatic properties resulted in changes in measured biological prop-erty (CB1, CB2 and GTPgS binding). From this, it was observed that a sidechain no longer than 3 A in length could be tolerated in the aminopiperidineregion. Furthermore, increased affinity and potency were predicted forcompounds having a substituent bearing a positive charge density in thisregion.CoMFA has also been used by Shim and co-workers [279] to help un-

derstand the interaction between rimonabant (382) and the CB1 receptor.

Page 73: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 279

Conformational analysis of (382) was first performed using the AM1 cal-culation method. Four distinct low-energy conformers (Tg, Ts, Cg, Cs) wereidentified corresponding to either trans (T) or cis (C) geometry around theamide bond and either skew (s) or gauche (g) depending on their torsionangle around the {(CQO)–N–N–C} bond in the aminopiperidine region. Inthe unprotonated form of (382), the Tg conformer was found to be the mostenergetically favourable followed by the Cg, Ts and Cs forms, which was inagreement with the AM1 calculated values. In the protonated state, the moststable conformation was Ts, followed by Tg, which was more stable than Cs

and Cg. Both protomeric states were examined in the subsequent QSARmodel since the extensive conjugation of the molecule made it uncertain asto which form would predominate under physiological conditions. ADISCO-derived pharmacophore model was developed using a previouslypublished superposition model [280] of cannabinoid agonists CP 55,244(196) and WIN 55,212-2 (254a). Construction of a 3D-QSAR CoMFAmodel for (382) and a series of 31 closely related structural analogues wasperformed using binding affinities determined from displacement of [3H]-CP55,940 in rat brain membranes. The Tg, Cg and Ts conformers only wereconsidered for the neutral species, while the Ts, Tg and Cs conformers in theprotonated species were used in the model. The values obtained for the r2

and cross-validated r2 were indicative of statistical robustness and predict-ivity in all cases except for the protonated Tg conformer where the r2cv valuefell to 0.36, below the accepted threshold for internal predictive ability. Thedata obtained from the studies suggested a crucial role for the N1 aromaticring in forming a hydrophobic interaction with the CB1 receptor. It was alsosuggested that the second aryl ring may prevent the change in receptorconformation necessary for agonist activity. In addition, it was postulatedthat the C3 amide substituent might interact with the Lys-192 residue, be-lieved to be participating in a specific ligand-binding interaction, therebyprohibiting agonist activity and also inducing or stabilising the receptorconformation required for inverse agonism.This latter theme was subsequently explored by Hurst et al. [281]. Receptor

models of the R and R* forms of CB1 were constructed using the 2.8 A crystalstructure of bovine rhodopsin. Rimonabant (382) and a conformationallyconstrained analogue (VCHSR), in which the (N-piperidinyl)-3-carboxamidemoiety of rimonabant was replaced by 3-[(E)-2-cyclohexylethenyl], wereevaluated in these models, and in vitro, in both the wild-type CB1 receptor andin a mutant in which the Lys residue at position 192 had been mutated to Ala.The two ligands were docked into the aromatic transmembrane helix (TMH)3-4-5-6 regions of the active and inactive TMH bundle, and the energy mi-minised using the AMBER force field. In the inactive R complex, (382)was observed to form a hydrogen bond with Lys-192 (TM3.28) which is part

Page 74: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH280

of a salt bridge with Asp-366 (TM6.58). In addition, aromatic stackinginteractions were formed with Trp-279 (TM5.43) with the dichlorophenyl ringinteracting with Phe-200 (TM3.36) and the monochlorophenyl ring interact-ing with Tyr-275 (TM5.39). In the active state, the relative positions of keyTMH3 and TMH6 residues Lys-192 and Asp-366 were observed to havechanged, breaking their salt bridges and preventing the interaction betweenLys-192 and the C3-substituent of (382). In the VCHSR/CB1 R complex, themodelled interactions were very similar to those of (382), but lacked thehydrogen-bonding interaction between Lys-192 and the C3 substituent.Analysis of the VCHSR/CB1 R* complex suggested that VCHSR shouldhave approximately equal affinity for both the R and R* states, conferring theproperty of neutral antagonism. Consistent with this hypothesis, it was foundthat VCHSR behaved as a neutral antagonist at the wild-type CB1 receptor,as witnessed by the attenuation of the inhibition of the calcium current by(254a) in superior cervical ganglion neurons. It was therefore proposed thatthe interaction between the Lys-192 residue and the C3 substituent of (382)was the principal determinant in the high affinity for the inactive R state andhence the experimentally observed inverse agonism.Molecular modelling and automated docking studies by Salo et al. [282]

also confirmed that Lys-192 played a key role in the binding mode ofHU210 (165), CP 55,940 (193), rimonabant (382), WIN 55,212-2 (254a) and2-AG. Further mutagenesis and molecular modelling work [283] comparedthe interaction of (382), (254a) and anandamide (1) with CB1 wild type andF3.25A, F3.36A, W5.43 and W6.48 mutants. The binding of (382) and(254a) was found to be affected by the F3.36A, W5.43 and W6.48 muta-tions, implying their presence in the binding site of these two ligands. Incontrast, only the F3.25A mutation was found to influence the binding of(1), suggesting the existence of a distinct binding site. It was therefore sug-gested that an aromatic micro domain in the TMH3-4-5-6 region comprisingF3.36A, W4.64, Y5.39, W5.43 and W6.48 was responsible for binding both(382) and (254a), but that the endocannabinoid-binding pocket may onlypartially overlap with the former lipophilic domain.Patent applications from Pfizer disclosed 1,5-diaryl-pyrazoles bearing

bioisosteric replacements for the 3-carboxamide moiety. One applicationshowed that the amide could be replaced by a-aminoketones as exemplifiedby compound (416) [284]. The corresponding alcohols and their ethers werealso described, including compounds that allowed the amine substituent andether to form a ring system, such as a morpholine unit. This application alsoallowed for the replacement of the 1,5-diaryl-pyrazole by a 1,2-diaryl-imidazole bearing a 3-carbonyl substituent, as exemplified by compound(417). A further patent application from Pfizer claims compounds in whichimidazoles replace the 3-carboxamide moiety in the 1,5-diaryl-pyrazole

Page 75: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 281

series [285]. Compound (418), one of the compounds specifically claimed inthe patent, showed a CB1 receptor affinity of 371 nM.

NN

Me

ClCl

NN

Ph

Me

N N

ClCl

O N

Cl

NN

ClCl

O N

(418)(417)(416)

In addition, Pfizer has identified a number of related, fused bicyclicpyrazole analogues [286–292]. These compounds, of which structures(419–425) are specified examples, are claimed to be of use in the treatmentof a number of diseases including alcoholism, psychosis, tobacco abuse,Parkinson’s disease and obesity.

N

N

NN

OCl

Cl

PhCl

(419)

N

N

NN

NCl

Cl

Cl

N

Me

(420)

NN

Cl

FF

F

N

O

i-Pr NN

Cl

Cl

N i-Pr

O

(421) (422)

Page 76: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH282

N

NN

N

NCl

F

H2NCO NHi-Pr

(425)

X

N

NN

NCl

Cl

N

Me

(423) X = CH(424) X = N

4,5-DIHYDRO-1H-PYRAZOLE DERIVATIVES

Solvay Pharmaceuticals have claimed 4,5-dihydro-1H-pyrazole derivativesin a series of patent applications [293–296], compounds (426–429) representexamples of the structures involved.

NN

Ph

Cl N

SMe

SNEt2

O O

NN

Ph

Cl N

NH

N

SO O

Cl(426) R = Cl (SLV-319)(427) R = CF3 (SLV-326)

(428)

(429)

N

N

Ph

Cl N

NHMe

S

O O

R

Solvay subsequently published in vitro, in vivo, computational and X-raydiffraction data relating to this series of 3,4-diarylpyrazolines [297]. In this

Page 77: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 283

publication, a number of compounds were evaluated, in the first instance invitro at the human CB1 and CB2 receptors, expressed into Chinese hamsterovary (CHO) cells by radioligand binding. In addition, the CB1 functionalactivity was measured via an arachidonic acid release functional assay. Re-sults for a number of the compounds are shown in Table 6.37.For compounds (431–434), a significant improvement in binding affinity

was observed when the 4-Me group in (430) was substituted with either Cl(431), 2,4,6-trimethyl (433) or F (434). Replacement of 4-Me with 4-OMe(432) failed to improve the affinity or functional activity. In the amidine-substituted series (435)–(447), dimethyl compounds (435) and (436) both

Table 6.37 PYRAZOLINE DERIVATIVES AND RIMONABANT (382) – IN VITRODATA [297]

N N

Cl

N

N

R2R3

S OO

R1

Cpd. R1 R2 R3 CB1 Ki (nM) CB1 pA2 CB2 Ki (nM)

(382) 25 8.6 1,580(430) 4-Me H H 197 8.4 >1,000(431) 4-Cl H H 16.1 9.5 >1,000(432) 4-OMe H H 196 8.3 >1,000(433) 2,4,6-Me3 H H 24.2 9.4 >1,000(434) 4-F H H 52.6 9.0 >1,000(435) 4-Cl Me Me 280 8.5 >1,000(436) 4-F Me Me >1,000 o7.5 >1,000(437) 2-Cl H Me 75.4 8.3 >1,000(438) 3-Cl H Me 13.9 8.6 >1,000(439) 4-CF3 H Me 221 9.3 >1,000(440) 4-Cl H Me 25.2 8.7 >1,000(441) H H Me 170 7.5 >1,000(442) 4-F H Me 338 8.5(443) 4-Me H Me 119 8.6 >1,000(444) 3-CF3 H Me 36.5 9.1(445) 2,4,6-Me3 H Me 54.2 9.4(446) 4-OMe H Me 22.9 8.0(447) 3,4-Benzo H Me 21.8 8.5

Page 78: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH284

show reduced affinity compared with their unsubstituted counterparts (431)and (434), respectively. The affinity of the amidine-NH2 analogues wasbroadly similar to that of the amidine-NHMe compounds. 3-Chloro (438)and 4-chloro (440) were approximately equipotent, however, there was someloss of activity witnessed when the substituent was moved to the 2-position(437). In contrast to (432), the 4-methoxy analogue (446) in the amidine-NHMe series was found to have excellent CB1 affinity (22.9 nM) while tri-fluoromethyl substitution at the 3-position (444) also yielded a more potentCB1 ligand than the 4-CF3 congener (439). A bulky 2-naphthyl-substitutedanalogue also proved to have excellent affinity. The amidine functionalitywas shown to be crucial for good CB1 affinity, since replacement by anamide drastically reduced affinity, whereas the substitution of the amidine-NH2 group for SMe, was more tolerated. Racemic (439) and (440) wereresolved and each of the enantiomers tested for their CB1 affinity. In eachcase, the levorotatory (4S-) eutomers were found to be more potent thantheir dextrorotatory equivalents; 4S-(439) (427) showed an affinity of35.9 nM and 4S-(440) (426) had an affinity of 7.8 nM. Throughout the se-ries, good CB1/CB2 selectivity was evident.Table 6.38 describes some of the key in vivo data for the significant

pyrazoline compounds in a CB1 agonist-induced hypotension rat model andhypothermia mouse model, compared with the Sanofi-Aventis CB1 antag-onist, rimonabant (382). From the initial disappointing discovery that pro-genitors (430) and (431) were inactive when administered orally in bothassays, it was found that (435), the amidine-NMe2 analogue of (431) didexhibit in vivo effects. Subsequently, (437)–(439) were also found to have invivo efficacy in both assays when administered orally. In common with thein vitro data, the 4S-analogues of (439) and (440) were found to be active invivo, while the 4R-enantiomers failed to display CB1 activity.

Table 6.38 PYRAZOLINE DERIVATIVES AND RIMONABANT(382) – IN VIVO DATA

Cpd. Rat hypotensionED50

aMouse hypothermia

LEDbCpd. Rat hypotension

ED50a

Mouse hypothermiaLEDb

(382) 3.2 3 (439) 8.9 3(430) >30 >30 (440) 15 3(431) >30 >30 (427) 2.0 1(435) >30 10 (426) 5.5 3

aAntagonism of CB agonist (CP 55,940)-induced hypotension in rat, expressed as ED50 (mg/kg,p.o.).bAntagonism of CB agonist (WIN 55,212-2)-induced hypothermia in mouse, expressed as leasteffective dose (LED) (mg/kg, p.o.).

Page 79: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 285

An X-ray diffraction study of (426) revealed the existence of a hydrogenbond between the N-atom at the 2-position of the dihydropyrazole ring andthe hydrogen atom of the amidine. A series of Confort-generated conform-ers of (426) were generated and minimised (MOPAC, PM3) and then com-pared with the favoured trans-gauche Tg conformer [279] of rimonabant(382). The conformer of (426) which overlapped best with the Tg conformerof (382) was then docked into a reconstructed model of the CB1 receptor[281]. It was found that hydrogen bond was formed between one of theoxygen atoms of SO2 and the Asp-366:Lys-192 salt bridge. An additionalH-bond was also observed between Ser-383 and the remaining SO2 oxygenatom. A stacking interaction could be envisaged between the 4-Cl-phenylring of (426) and the Phe-170 residue. Two hydrophobic pockets, defined byTrp-279, Phe-200, Trp-356 and Tyr-275, Trp-255, Phe-278 bind the two4-Cl-phenyl rings.Investigation of the differences in crystal packing between (431) and (426)

from comparison of their respective X-ray structures, revealed that (431)was more tightly packed than (442), reflected in their respective meltingpoints of 235 and 170 1C. It was postulated that the absence of in vivoactivity for (431) may be explained by the resultant reduction in watersolubility and dissolution rate compared with (426). The comparatively highcalculated polar surface area of (431) (122.5 A2) compared with (426)(89.3 A2) was also proposed as a factor influencing the marked difference inbioavailability between the two related compounds. Compound (426) (SLV-319) is currently being developed with Bristol-Myers Squibb for the poten-tial treatment of obesity and other metabolic disorders. Phase I trials forobesity were started in April 2004. Earlier Phase I clinical trials for thetreatment of schizophrenia and psychosis, which commenced in April 2002,appear to have been abandoned.

IMIDAZOLE-, THIAZOLE-, PYRROLE- AND TRIAZOLE-BASEDCB1 RECEPTOR ANTAGONISTS

4,5-Diarylimidazole derivatives, exemplified by (448) have been claimed byMerck as either antagonists or inverse agonists of the CB1 receptor [298]. In thisseries of compounds, the central imidazole motif can be considered a bioisost-ere of the pyrazole core of rimonabant. A similar approach was utilised bySolvay Pharmaceuticals [299] in a patent application published shortly afterthat of Merck. The isomeric 1,2-diaryl-4-carboxamide nucleus (449) can againbe considered as a structural isostere, serving to orient the two substituted arylgroups and carboxamide group in geometry analogous to that of (382). Analternative methylimidazole core, 1,2-diaryl-5-methylimidazole (450), was also

Page 80: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH286

utilised by Merck [300]. More recently, Pfizer has disclosed a series of novelimidazole derivatives useful in the treatment of obesity, alcoholism and de-pression [301]. The specified compound (451) was stated to have CB1 bindingaffinity in the range of 1–10nM.

N

N

Me

NH

O

Cl

Cl

Cl N

NNH

O

Cl

Cl

Cl

N

(448) (449)

N

N

O

NH

Cl

Cl

Cl

Me

N

NN

i-PrNH

O

ClCl

(450) (451)

Novel 6-aminopurine compounds, for example, (452) have been claimed byPfizer as particularly useful in the treatment of obesity and alcohol abuse [302].In conjunction with an opioid receptor antagonist, the related compounds suchas (453) are claimed as a combination therapy for treating alcohol, cocaine ortobacco addiction or dependence, reducing alcohol withdrawal symptoms andaiding in the cessation or lessening of alcohol abuse, substance abuse or be-havioural dependency, including gambling [303]. Compound (453) has alsobeen claimed to have utility in combination with a nicotinic acetylcholine ago-nist in the treatment of obesity or compulsive overeating [304] and compulsivegambling, nicotine and drug dependence [305].

Page 81: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 287

N

N N

N

N Cl

Cl

N

Cl

N

N N

N

N

CONH2EtNH

Cl

Cl

S

N

O

NH

X

Cl

Cl

Cl

(452) (453)(454) X = CH(455) X = N

Thiazole-based cannabinoid CB1 modulators were claimed in two patentapplications by AstraZeneca [306, 307] where the strategy of bioisostericpyrazole-replacement is evidenced by the specified compounds (454) and(455), respectively. No specific biological data were presented but the spec-ified compounds were stated to have IC50 values o200 nM. A centralthiazole motif was also utilised by Solvay Pharmaceuticals [308]. In vivoantagonism was evaluated using CP 55,940-induced hypotension in rats,however no specific results were given. Compound (455) was shown to havebroadly similar affinity at both CB1 and CB2 receptor subtypes with pKi

values of 7.8 and 8.1, respectively.A thiazole core has also been utilised by Solvay Pharmaceuticals [309] in

the search for a novel bioisosteric replacement of the rimonabant (382)pyrazole core. The affinity of several compounds for the human CB1 re-ceptor was determined in transfected CHO cells using tritium-labelled CP55940. Antagonism was determined in the same cell line by WIN 55212-2-induced release of arachidonic acid. The pKi of (456) was found to be 6.9,while the pA2 value was measured as 8.7. A series of six 1,2,4-triazole an-alogues has been prepared by Jagerovic et al. [310], via their correspondingN-acylbenzamides (Table 6.39).

NH

H

H

H N

S Ph

n-C5H11

O N

N

N

O

NH

Cl

Cl

Cl

(456)(457)

Page 82: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.39 STRUCTURES OF 1,2,4-TRIAZOLE ANALOGUES OFRIMONABANT (382)

N

NN (CH2) MeR

2

R1

n

Cpd. R1 R2 n Cpd. R1 R2 n

(458) 4-Cl H 1 (461) 4-Cl 2,4-di-Cl 5(459) 4-Cl 4-Cl 1 (462) 2,4-di-Cl 2,4-di-Cl 5(460) 4-Cl 4-Cl 5 (463) H H 6

RECENT PROGRESS IN CANNABINOID RESEARCH288

The biological activity of the compounds was examined by assessing theirability to antagonise WIN 55,212-2 induced inhibition of electrically in-duced contractions in mouse vas deferens and guinea pig ileum. Compounds(459)–(463) were not seen to induce modification of the contractile responsein either the vas deferens or ileum tissues when administered alone, effec-tively ruling out the possibility that these compounds behaved as agonists.Compound (458) was found to inhibit contractile activity in the guinea pigileum, an effect that was not antagonised by (382), thus precluding can-nabinoid agonist activity, however the mechanism underlying the observedinhibition was not investigated further. Upon incubation of (458–463) in thepresence of WIN 55,212-2, analogue (461), bearing the familiar (4-chloro-phenyl)-(2,4-dichlorophenyl) substitution, induced a significant and dose-dependent decrease of the inhibition produced by the aminoalkylindoleagonist in both tissues. The binding affinity of (461) in rat cerebellularmembranes using [3H]-rimonabant and [3H]-WIN 55,212-2 was found to bemoderate (K i ¼ 855:6 and 748 nM, respectively) compared with rimonabant(382) (K i ¼ 4 nM). The effects of (461) in vivo were also assessed in thecannabinoid tetrad behavioural tests (nociception, temperature, spontane-ous motility and catalepsy). Compound (461), administered intraperitone-ally (1mg/kg) was found to behave as a cannabinoid antagonist, reversingthe effects of WIN 55,212-2 (1.5mg/kg, i.p.).In addition to triazole-based biosteres of (382), thiazoles and imidazoles

have also been investigated by Lange et al. [311]. The three series of com-pounds were assessed on their CB1 binding affinity and functional activity

Page 83: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 289

and their selectivity over the CB2 receptor in CHO cells stably transfectedwith human CB1 and CB2 receptor.Comparison of thiazole-based regioisomers (464) and (465) has revealed a

marked preference for the phenyl substitution of the former, a trend alsoobserved in the subsequent imidazole series (data not shown), but not in thetriazole series as evident from the data presented for (466) and (467) (Table6.40). The thiazole (464) was approximately 8-fold less potent than thecorresponding imidazole (470) (Table 6.41) suggesting that the seeminglysubtle change of the central scaffold imparts a marked change in CB1 re-ceptor recognition.Ethyl substitution at the imidazole 5-position (469) was found to increase

potency over the unsubstituted analogue (468), while methyl substitution(470) had a slightly deleterious effect on binding (Table 6.41). Chloro (491),bromo (492), cyano (493) and fluoromethyl (494) substitution at this positionwere all well tolerated (Table 6.43). Introduction of a chloro-substitutedpyridine (475) in place of the more usual p-chlorophenyl group (470) resultedin a slight loss of affinity for the CB1 receptor, as did replacement of the p-chloro group of (470) with bromo (471), fluoro (472) and in particular, met-hoxy (473). Trifluoromethyl substitution (474) however, was well tolerated.The effect of replacing the carboxamide N-piperidinyl group of (470) was

also investigated. Reducing the ring of the piperidinyl group by one carbon

Table 6.40 THIAZOLE- AND TRIAZOLE-BASED BIOISOSTERES OF RIMONABANT(382) – IN VITRO DATA [311]

S

N

N

O

N

R1

Cl

R2

Cl

N N

N

N

O

N

R4

Cl

R3

Cl

Cpd. R1 R2 R3 R4 CB1 Ki (nM) CB1 pA2 CB2 Ki (nM)

(382) 25 8.6 1,580(464) Cl H 227 8.1 5,841(465) H Cl >1,000 7.2 4,668(466) Cl H 356 8.3 3,562(467) H Cl 382 7.6 5,444

Page 84: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.41 IMIDAZOLE-BASED BIOISOSTERES OF RIMONABANT (382) – IN VITRODATA [311]

N

N

N

O

N

X

R3

Cl

R1

R2

Cpd. R1 R2 R3 X CB1 Ki (nM) CB1 pA2 CB2 Ki (nM)

(468) Cl H Cl C 23 8.2 542(469) Cl Et Cl C 14 9.0 430(470) Cl Me Cl C 30 8.6 608(471) Br Me Cl C 60 8.5 489(472) F Me Cl C 52 7.7 765(473) OMe Me Cl C 106 8.7 326(474) CF3 Me Cl C 29 8.6 634(475) Cl Me Cl N 55 8.4 758

RECENT PROGRESS IN CANNABINOID RESEARCH290

(476) yielded a compound approximately equipotent with (470) whileincreasing the ring (477) or introducing a morpholine group (478) reducedthe binding affinity (Table 6.42). Larger bicyclic substituents were well tol-erated, as evidenced by (479), (485) and (486). Cyclopentylamine (480),cyclohexylamine (481) and cycloheptylamine (482) substitutents were alsoaccommodated in the lipophilic binding pocket. However, the presence of ahydroxyl group resulted in a marked decrease in affinity, for example, (483)and (484).The CB1 functional activity of a number of the imidazole-based com-

pounds reported exceeds that measured for rimonabant (382) (Table 6.43).While marked CB1/CB2 selectivity is witnessed throughout the series, theselectivity of direct (382) analogue (470) was found to be approximately 3-fold lower than for (382) itself (Table 6.44).The in vivo properties of compounds (464), (466) and (468)–(470) relative

to (382) were investigated in two animal models: a CB1 agonist-inducedhypotension rat model and a CB1 agonist-induced hypothermia mousemodel. Shown to be only moderately active in the in vitro screens, thiazole(464) and triazole (466) analogues failed to demonstrate convincing oralactivity. Compound (468) was active in the hypotension model but showed

Page 85: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.42 IMIDAZOLE-BASED BIOISOSTERES OF RIMONABANT (382) – IN VITRODATA [311]

N

N

R

OCl

Cl

Cl

Me

Cpd. R CB1 Ki (nM) CB1 pA2 CB2 Ki (nM)

(476)NN

H 27 8.2 774

(477)

NNH

64 8.7 505

(478)NN

HO

197 7.5 3,297

(479)NN

H 40 9.8 1,412

(480) -NHc-Pentyl 33 8.9 357(481) -NHc-Hexyl 35 9.1 160(482) -NHc-Heptyl 35 9.1 349(483)

NH

OH399 7.3 3,469

(484)N OH

172 o7.5 3,959

(485)

N

34 8.4 696

(486)NH 19 9.1 54

(487) -NHOt-Bu 333 8.2 242(488)

NH

CF3

171 8.9 1,984

(489) -NEt2 828 7.4 2,520(490) -Ot-Bu 94 8.6 815

J. ADAM ET AL. 291

Page 86: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.43 IMIDAZOLE-BASED BIOISOSTERES OF RIMONABANT (382) – IN VITRODATA [311]

N

N

N

O

N

X

R3

Cl

R1

R2

Cpd. R1 R2 R3 X CB1 Ki (nM) CB1 pA2 CB2 Ki (nM)

(491) Cl Cl Cl C 27 8.5 823(492) Cl Br Cl C 23 8.4 746(493) Cl CN Cl C 30 8.6 1,590(494) Cl CH2F Cl C 36 8.9 906

Table 6.44 IMIDAZOLE DERIVATIVES AND RIMONABANT(382) – IN VIVO DATA [311]

Cpd. Rat hypotensionED50

aMouse hypothermia

LEDbCpd. Rat hypotension

ED50a

Mouse hypothermiaLEDb

(382) 3.2 3 (468) 11.7 >30(464) >30 Not determined (469) 15.8 30(466) 23.6 >30 (470) 2.4 10

aAntagonism of CB agonist (CP 55,940)-induced hypotension in rat, expressed as ED50 (mg/kg,p.o.).bAntagonism of CB agonist (WIN 55,212-2)-induced hypothermia in mouse, expressed as leasteffective dose (LED) (mg/kg, p.o.).

RECENT PROGRESS IN CANNABINOID RESEARCH292

no activity in the mouse hypothermia model. The 5-ethyl (469) and in par-ticular, the 5-methyl (470) analogues showed in vivo efficacy in both models,strongly suggesting that the 5-methylimidazole scaffold behaves as a truebioisostere of the pyrazole ring of (382).Reconstruction [297] of the CB1 homology model reported by Hurst et al.

[281] was carried out, and ligands (464), (466) and (470) docked into thereceptor along with rimonabant (382). A perfect 3D overlap of (382) and(470) was observed. In the case of the thiazole (464) and triazole (466), the

Page 87: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 293

carboxamide and aryl rings were found to overlap well with (382), althoughthe central heterocycle deviated slightly from the orientation adopted by (382)and (470). The importance of the substitution patterns on the phenyl ringswas also investigated. It was been reported that (382) binds into the active siteof the CB1 protein in its most stable so-called transdichlorophenyl (TDC) con-formation, in which the dichlorophenyl ring is oriented trans with respect tothe carbonyl of the carboxamide group. It has been proposed that the saltbridge formed between residues Lys-192 and Asp-366 is stabilised by a hy-drogen-bonding interaction with the carboxamide carbonyl of (382) and iscrucial for conferring inverse agonism [281]. The 4-chlorophenyl ring wasobserved to participate in direct stacking interactions with Trp-255, Tyr-275,Phe-278, and Trp-279 forms a stacking interaction with both aromatic ringsof the ligand. In addition, the 2,4-dichloro ring is located in the proximity offurther two aromatic residues, Trp-356 and Phe-200, where a small lipophilicpocket enhances the binding interaction with the 2-chloro substituent. It wasproposed that the TDC conformation of (382) is stabilised by a weak hydro-gen-bond interaction between the carboxamide NH and the proximal nitro-gen atom in the pyrazole core. It was postulated that the TDC conformationof (472) is also stabilised by such an interaction, favouring this conformationover the alternative cisdichlorophenyl (CDC) conformer, which would potentiallygive rise to steric occlusion between the ortho-chloro substituent on the 2,4-dichlorophenyl ring and the residues Phe-278 or Val-364.A series of 4,5-diarylimidazole-2-carboxamides has been reported as hu-

man CB1 inverse agonists [312]. The initial HTS hit, (495), based on ananalogous scaffold to (382), was found to have moderate affinity for the CB1

receptor (7,000 nM).

N

NH

Ph

Ph SAc

Ph

(495)

The relatively low affinity was ascribed to the absence of substitution onthe phenyl rings and the absence of an amide group to form an interactionwith the receptor. Among the compounds synthesised during the hit opt-imisation program were (496–507) (Table 6.45).The trichloro derivative corresponding most closely to rimonabant (382),

(496) was found to have excellent CB1 affinity (IC50 ¼ 6:1 nM) (cf. (382):

Page 88: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.45 4,5-DIARYLIMIDAZOLE DERIVATIVES – IN VITRO DATA [312]

N

N

Cl

ClR2

R3

NHR1

OR4

Cpd. R1 R2 R3 R4 CB1 IC50 (nM)

(496) N-Piperidinyl H Cl Me 6.1(497) c-Hexyl H Cl Me 4.0(498) N-Morpholinyl H Cl Me 170(499) c-Pentyl H Cl Me 17(500) c-Heptyl H Cl Me 8.0(501) c-Hexyl H Cl Me 70(502) Ph H Cl Me 60(503) c-Hexyl H Cl Et 4.1(504) N-Piperidinyl Cl H Me 190(505) c-Hexyl Cl H Me 46(506) N-Piperidinyl Cl Cl Me 16(507) c-Hexyl Cl Cl Me 5.2

RECENT PROGRESS IN CANNABINOID RESEARCH294

K i ¼ 6 nM). The closely related cyclohexyl analogue (497) was found to beslightly more potent than both. However, as witnessed in the analogous seriesby Lange et al. [311], replacement of the N-piperidinyl group by N-morph-olinyl was detrimental to the CB1 binding affinity, suggesting that polarfunctionality was not well tolerated into the amidoalkyl region. A cycloheptylgroup at this position was relatively well tolerated (500), with a more sub-stantial decrease in affinity reported upon reduction of the cycloalkyl ring tocyclopentyl (499). The isomer of (496) in which the phenyl substitution wasswapped between the two aryl rings (504) was found to be considerably lesspotent than (496) and indeed less potent than either of the tetrachloro an-alogues (506) and (507). Replacement of the imidazole N-Me group by N-Et(503) resulted in a broadly equipotent analogue.A number of oxazole derivatives were also synthesised, of which (508) was

among the most potent (IC50 ¼ 80 nM). In general however, the bindingaffinities reported for the oxazole compounds were at best modest, andcomparable with the related imidazole NH series (data not shown).

Page 89: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 295

N

O

Cl

Cl

Cl

NHc-Hex

O

(508)

In vivo studies on the leading compounds (496) and (497) were carriedout. Preliminary rat PK studies (1mg/kg i.v., 2mg/kg p.o.) for (496) indi-cated a good profile for i.v. administration (AUCnom ¼ 1:32 mMhkg=mg;Clp ¼ 27:5ml=min=kg; Vdss ¼ 3:7L=kg; t1=2 ¼ 2:4 h) and oral absorption(F ¼ 50%), in addition to a high brain:plasma ratio (B:P ¼ 2.99–3.40 be-tween 0.25 and 4 h). An immediate, dose-dependent and prolonged reduc-tion in food intake was subsequently observed in a body weight loss studycarried out using diet-induced obese rats. At doses of 1, 3 and 10mg/kg p.o.,the reduction was reported to be �13.4%, �30.6% and �67.5% relative tothe vehicle-treated animals. At 18 h after dosing of (496) at 10mg/kg, acumulative, statistically significant dose-dependent weight loss of 13 g wasobserved, compared with a 6 g gain for the vehicle-treated animals. A poorerresult was obtained with (497), with a net weight loss of 2 g versus a 10 gweight gain for the vehicle-treated animals. This result was, however, in linewith a poorer PK profile, resulting in lower bioavailability (F ¼ 30%) andslower brain penetration (B : P ¼ 0:8422:61 between 0.25 and 4 h).Stated to be particularly useful in the treatment of obesity, a series of 1,5-

diaryl-pyrrole-based compounds from AstraZeneca was claimed in a recentpatent application [313], including compound (509). Although no specificbiological data were presented, the CB1 receptor affinities are claimed to beless than 1 mM and preferably less than 200 nM.

PYRIDINE-, PHENYL-, PYRIMIDINE- AND PYRAZOLE-BASEDCB1 RECEPTOR ANTAGONISTS

Six-membered heterocycles have also been extensively used to mimic the centralpyrazole scaffold of rimonabant (382). Merck and Co. has utilised pyridines(510) [314] , and pyrimidines (511) [315] in this capacity. Sanofi-Aventis has alsoclaimed a series of pyridine-based analogues [316], as exemplified by (512) andadditionally the non-heteroatom containing terphenyl (513) [317].Pfizer too, has filed a patent application to protect a series of closely related

pyrimidine-2-carboxamides [318]. Although no compounds are specificallyclaimed, the examples include compound (514) in which the chloro-substituted

Page 90: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH296

diaryl motif is once again present. The affinity of this analogue in the CB1

GTPgS binding assay was claimed to be less than 20nM.

N MePh

Ph

O

NHPh

N

Ph

OCH2Ph

Cl

OMe

O

N

N

Cl

Cl

Cl

O

F

(509)

(511)

(510)

O

NH

N

X

Cl

ClCl

N

N

N

O

Ph

Ac

Cl

ClCl

(512) X = N(513) X = CH

(514)

In a recent communication from Merck [319], the synthesis and biologicalactivities of several 5,6-diarylpyridine derivatives as CB1 inverse agonistswere described. From the initial HTS screening on the Merck sample col-lection was identified (515) which was found to have moderate activity(IC50 ¼ 530 nM) at the human CB1 receptor. It was proposed that thepyridine ring behaved as an alternative scaffold to the pyrazole ring of (382),with the pyridine nitrogen overlapping with the N2 atom of the pyrazole. Inaddition, it was speculated that the 3-cyano group might act as a bioisostere

Page 91: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 297

for the carboxamide of (382). Investigation of the phenyl substitution of thisseries yielded significant improvement in CB1 potency (Table 6.46).

(515)

N

O

NMe2

CN

MeO

The initial 4-Cl analogue (516) was found to exhibit only modest affinity,however derivative (517) showed CB1 inverse agonist activity comparablewith (382) while also demonstrating good selectivity over the CB2 receptor.

Table 6.46 5,6-DIARYLPYRIDINE DERIVATIVES – IN VITRO BINDING DATA [319]

N

R1

CN

O

R2

R3

R4

Cpd. R1 R2 R3 R4 CB1 IC50 (nM) Cpd. R1 R2 R3 R4 CB1 IC50 (nM)

(382) 6.2 (522) Cl Cl Cl 3-Cl 5.8(516) H H Cl H 2,800 (523) Cl Cl Cl 4-CF3 3.4(517) Cl Cl Cl H 11 (524) Cl H Cl 3,4-di-F 18(518) Cl Cl Cl 4-F 3.1 (525) Cl Cl H 3,4-di-F 8(519) Cl Cl Cl 3,5-di-F 1.8 (526) F Cl Cl 3,4-di-F 7.2(520) Cl Cl Cl 2,4-di-F 2.7 (527) Me Cl Cl 3,4-di-F 6.3(521) Cl Cl Cl 3,4-di-F 1.3 (528) F F F 3,4-di-F 66

Page 92: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH298

The 4-fluoro (518), 4-trifluoromethyl (523) and 3-chloro derivatives (522)also showed improved binding. 3,5-Disubstitution was also well tolerated(519), however the 3,4-difluoro compound (521) had the most potent inverseagonism of the series with an EC50 of 8 nM, with an average 110% maximalresponse and 400-fold selectivity over CB2. In addition to the crucial role ofthe 4-chloro substituent on the 5-phenyl ring, the 2,4-dichloro substitutionon the 6-phenyl ring, which mimics that of (382), was also found to beoptimal.Replacement of the 3-cyano functionality was also investigated with a

series of 3-amido derivatives (Table 6.47). Of these, the 3,4-difluorobenzylanalogues were the most potent. Modification, including lengthening andbranching the R2 and R3 alkyl groups have comparatively little effect onpotency (cf. (529)–(531)). Replacement of the 3,4-difluorobenzyl motif withn-butyl did however considerably reduce the CB1 binding affinity.Cyclohexylmethyl did appear to be tolerated in this position with com-pounds approximately equipotent with their benzyl counterparts (cf. (529)and (537)) suggesting that this group binds in a lipophilic pocket rather thanmaking a direct aromatic interaction.

Table 6.47 5,6-DIARYL-3-AMIDOPYRIDINE DERIVATIVES – IN VITRO BINDINGDATA [319]

N

Cl

OR1

Cl

Cl

O NHR2

Cpd. R1 R2 CB1 IC50 (nM) Cpd. R1 R2 CB1 IC50 (nM)

(529) 3,4-di-F-Bn Me 3.1 (534) n-Bu H 32(530) 3,4-di-F-Bn Et 1.9 (535) n-Bu Me 17(531) 3,4-di-F-Bn n-Pr 1.7 (536) n-Bu n-Pr 21(532) 3,4-di-F-Bn (CH2)2F 1.5 (537) CH2(c-Hexyl) Me 3.4(533) 3,4-di-F-Bn i-Pr 1.8 (538) CH2(c-Hexyl) n-Pr 5.2

Page 93: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.48 5,6-DIARYL 2-AMINOPYRIDINE DERIVATIVES – IN VITRO BINDINGDATA [319]

N

ClCl

Cl

NR1R2

O

Cpd. R1 R2 CB1 IC50 (nM) Cpd. R1 R2 CB1 C50 (nM)

(539) –(CH2)5– 340 (544) H c-Heptyl 35(540) H n-Hexyl 43 (545) H Piperidin-1-yl 56(541) H n-Hept-4-yl 32 (546) H Ph 210(542) H c-Pentyl 53 (547) H Bn 31(543) H c-Hexyl 95

J. ADAM ET AL. 299

Investigation of a 2-amido series was also carried out by Meurer et al.(Table 6.48). These compounds were found to be generally less potent thanthe 2-alkoxy-3-amido series with (545), the closest analogue of (382), amongthe most potent. In addition, the n-hexyl (540) and 4-heptyl (541) analogueswere found to have comparable affinity, as was the benzyl congener (547).The relatively poor binding of the 2- and 3-amido pyridine series comparedwith (382) was ascribed to either the difference in ring size between pyridineand pyrazole resulting in a slight change in relative orientation of substitu-ents, and/or the basicity of the nitrogen and finally the absence of a neigh-bouring methyl group to force the amide group of rimonabant into aposition perpendicular to the pyrazole ring.In vivo studies were also performed on those compounds deemed to have

the most favourable binding and functional assay data, (521) and (531).Pharmacokinetic studies carried out on Sprague-Dawley rats (1mg/kg i.v.,2mg/kg p.o.) using compound (521) suggested favourable i.v. PK properties(AUCnom ¼ 9:3mMhkg=mg; Clp ¼ 3:6ml=min=kg; Vdss ¼ 0:8L=kg; t1=2 ¼3:6 h), moderate oral absorption (F ¼ 27%) but slow brain penetration anda low brain:plasma ratio (B : P ¼ 0:0320:26 at 0.25–4 h). Compound (521)was subsequently investigated in a food intake and body weight loss

Page 94: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH300

study using diet-induced obese rats. In contrast to (382), oral administrationof (521) and 1, 3 and 10mg/kg did not lead to an immediate reduction infood intake at any of the doses, however after 18 h after dosing at 10mg/kg,a cumulative but non-significant (p >0.05) 22% reduction in food intakeand a dose-dependent weight loss (1 g compared with an 8 g gain for thevehicle-treated animals) was observed. It was postulated that the slow CNSpenetration witnessed in the earlier PK study was preventing an immediateeffect on the food intake.Pyrazines have been utilised first by AstraZeneca [320–325] and subse-

quently by Bristol-Myers Squibb [326]. Although no specific biological datawere presented in the initial two AstraZeneca patent applications, com-pounds including the specified (548) are claimed to have IC50 values lessthan 1 mM with the most preferred compounds having an IC50 value of lessthan 200 nM. Nine compounds are specifically claimed in the later As-traZeneca application [322] including (549) in which the N-piperidin-1-ylcarboxamide of (382) is replaced with the bioisosteric piperidin-1-yl-oxy-carbonyl. Subsequently, a tetrazole-containing 5,6-bis(4-methylphenyl)-pyrazine derivative (550) was reported to have an IC50 of 1.4 nM [323].A related tetrasubstituted compound, (551), which had been disclosed ge-nerically in an earlier AstraZeneca application [321], has been specificallyclaimed [324]. The affinity of this compound for the CB1 receptor was de-scribed only as less than 1 mM. Finally, from AstraZeneca, a third series oftetrasubstituted pyrazines was revealed [327], of which compound (552) wasstated to have excellent CB1 affinity (1.8 nM). The now-familiar diaryl-pyrazine carboxamides have more recently been the subject of a patentapplication from Bristol-Myers Squibb [326]. Of the several compoundsclaimed, (553) shows that a hydroxymethyl functionality is tolerated adja-cent to the carboxamide nitrogen but no specific data were presented in theapplication.

N

N

X

O

N

R1

R1

N

N

NH

O

N

R1

R1

R2

NN N

N

N

O

(548) R1 = H, X = NH

(549) R1 = Cl, X = O

(550) R1 = Me, R2 =

(551) R1 = Cl, R2 =

Page 95: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 301

N

N

Me

Me

NH

O

Me

MeOH

N

N

Ot-Bu

O

Cl

Cl

O

N

(553)(552)

Merck has recently utilised a furo[2,3-b]pyridine core (554) as abioisosteric replacement for the pyrazole scaffold of rimonabant (382)[328]. The same basic pharmacophore, that of two halo-substituted arylgroups and a third hydrophobic motif proximal to a hydrogen-bond ac-ceptor, can be witnessed in the benzodioxole-based compounds, such as(555), disclosed by Roche [329].

N

O

NH2Ph

O

Cl

Cl

Cl

O

O

SN

O O

F

Cl

Cl

(554) (555)

AZETIDINE-BASED CB1 RECEPTOR ANTAGONISTS

A number of azetidine-based compounds have been disclosed in patent ap-plications from Aventis Pharma for CB1-modulated treatment of diseasessuch as obesity, Parkinson’s disease, schizophrenia, respiratory and neuro-logical diseases [330–334]. Compound (556) was specifically claimed for usein two formulation patent applications [330, 331] for a stable semi-solidcomposition and oral emulsion composition, respectively. The optional co-administration of an agent that activates norepinephrinergic and se-rotoninergic neurotransmission (for example, sibutramine) or dopaminergicneurotransmission was also claimed for the treatment of obesity. The op-tional use of a dopamine agonist (for example, levodopa) was claimed

Page 96: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH302

additionally for the reduction of dyskinesia in such neurological diseases asParkinson’s disease. Earlier Aventis patent applications disclose relatedazetidine derivatives such as (557) [334], (558) [333] and (559) [332]. Thestructural similarity to (382), of the three exemplified compounds shown, isless overt than the scaffold-hopping approaches discussed above, howeverthe p-chloro-substituted diaryl motif is preserved. The sulfonamide oxygenatoms may be performing the same hydrogen-bond acceptor role as thecarbonyl in rimonabant.

N

Cl Cl

NMeO2S R

N

Cl Cl

NHS

O O

S

N

ClCl

MeO2S

F

F

(557) R = 3,5-diF-phenyl(559) R = 5-Cl-2-pyridyl (558)(556)

O

ClCl

N NH

O

O

Cl

N N

O

N

Cl

BOC

(560) (561)

An azetidine motif was also present in two series of CB1 antagonist com-pounds disclosed by Vernalis Research [335, 336]. In the former, compound(560) was claimed to have an affinity of 285 nM in transfected HEK293 cellsusing tritium-labelled (382). Among the preferred indications were psycho-sis, schizophrenia, smoking cessation and eating disorders associated withexcessive food intake. Compound (561) was claimed to have an affinity of0.8 nM in the same binding assay [336].

Page 97: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 303

In two related applications from Roche, a pyrrole/imidazole [337] or 2-methylpyrrole [338] perform the role of central scaffold. Among the com-pounds specifically claimed are (562) and (563), stated to be antagonists orinverse agonists. Interestingly, a cyclohexylmethyl group is present whereone might expect the more usual halo-substituted phenyl, and in this respect,may share a related pharmacophore to the pyrrole-based CB1 agonists de-scribed by Wiley et al. [181] in which the most potent analogues bear anextended pyrrole N-alkyl substituent.

N

O

NO

O

N

S N

O

O

N

(562) (563)

SUBSTITUTED AMIDE-BASED CB1 RECEPTOR ANTAGONISTS

A number of distinct CB1 receptor modulators have been claimed by Merckfor indications including eating disorder, obesity, psychosis and drug de-pendence. In the earliest [339], the familiar biaryl pharmacophore is pre-served, although a substituted acyclic propyl chain, rather than aheterocyclic one, serves to orient the phenyl rings and carboxamide groupin the required configuration (564). Two closely related series were alsodescribed in applications from Merck [340, 341]. Again, no specific biolog-ical data were presented, however the benzofuran (565) and benzodioxananalogues (566) typify the series. More recently, Merck has claimed a fur-ther two series of CB1 antagonists [342, 343] of which (567) and (568) werespecifically claimed in each of the series, respectively.

NH

NH

Cl

NC

Me O

Et NH

O

Me

Cl

Cl R O

O

O

(564)

(565) R =

(566) R =

Page 98: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH304

NH

O

Ph

Me O

Me

Me

ClCl

ONH

N

CF3

Me

MeOMe

OH

CN

Cl

(567) (568)

HYDANTOIN-BASED CB1 RECEPTOR ANALOGUES

A series of 29 3-alkyl 5-arylimidazolidinones, or hydantoins, active at theCB1 receptor has been published by Kanyonyo et al. [344] with a subsequentpublication describing the relationship between the experimentally derivedlipophilicity and proposed modes of binding for non-polar and polar hyd-antoins derivatives [345] (Table 6.49).The binding data for compounds (569)–(592) were obtained by screening

at a concentration of 10 mM in a competitive displacement experiment usingtritium-labelled (382) in CHO membranes transfected with the human CB1

receptor. Within the series, affinity was seen to improve as the alkyl chainlength, and hence lipophilicity was increased. Compounds (588) and (589)were, however, surprisingly active for their calculated lipophilicity. The lip-ophilicity of the hydantoin analogues was measured by reverse-phase HPLCand found to correlate most closely with the values calculated by the CLIP[346] rather than CLOGP method [347]. In addition, para-substitution ofthe two phenyl rings improved potency, with 5,5’-bis(4-bromophenyl) sub-stitution, compounds (588)–(592), being optimal. For the non-polar hyd-antoins, the potency at the CB1 receptor decreased Br>OMe>F>Me>H,whereas no such relationship was evident for the non-polar compounds.Although certain polar groups, for example, morpholine (588) and hydroxyl(589) were well tolerated, the more basic amine group in compounds (570)and (571) had a strongly negative influence on binding. Molecular modellingstudies comparing the hydantoins with the non-classical cannabinoid HHCutilised molecular lipophilic potential and hydogen bond potential to gen-erate a superposition model. Two distinct binding modes, for polar and non-polar hydantoins, were identified. The three most potent compounds (588),(589) and (591) were further characterised using a [35S]-GTPgS binding as-say [348]. This assay allowed the test ligands to be defined as agonists (pos-itive intrinsic activity), partial agonists (partial positive intrinsic activity),antagonist (no intrinsic activity) or inverse agonists (negative intrinsicactivity). All three hydantoin ligands were found to behave as neutral

Page 99: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

Table 6.49 3-ALKYL-5,50-DIPHENYLIMIDAZOLIDINEDIONES – IN VITRO DATA[344,345]

N

NH

O

O

R1

R1(CH2) R

2n

Cpd. R1 R2 n c logPa % Displacement at 10 mMb

(569) H N-morpholine 2 2.16 o5(570) H N-piperidine 2 3.83 o15(571) H NMe2 2 2.83 o5(572) H Me 2 3.84 o20(573) H Me 3 4.26 25.1(574) H Me 4 4.8 35.4(575) H Me 5 5.37 35.6(576) H Me 7 6.50 61.2(577) H Ph 1 4.11 40.6(578) H i-Pr 0 2.89 o5(579) Me N-morpholine 2 3.49 23.9(580) Me Me 5 6.48 46.8(581) Me Me 6 7.04 51.3(582) OMe N-morpholine 2 2.73 21.7(583) OMe Me 5 5.81 66.6(584) F N-morpholine 2 2.81 30.3(585) F Me 5 5.81 40.6(586) F Me 6 6.42 51.4(587) F Me 7 6.94 62.5(588) Br N-morpholine 2 3.86 91.2(589) Br OH 3 3.76 88.4(590) Br Me 5 6.87 72.1(591) Br Me 6 7.45 89.2(592) Br Me 7 7.99 80.0

aLipophilicity calculated using the CLIP method.bResults expressed as the percentages of the displaced specific binding of [ 3H]-SR-141716A(mean7SEM, n ¼ 325).

J. ADAM ET AL. 305

antagonists with no intrinsic activity, but competitively inhibiting HU210-induced [35S]-GTPgS binding in rat cerebellum homogenates.A series of closely related thiohydantoins was subsequently investigated

by Muccioli et al. [349]. The key data are summarised in Table 6.50.

Page 100: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH306

Replacement of the oxygen in the 2-position of the hydantoin ring withsulfur was found to increase CB1 receptor potency between 2- and 4-fold. Asin the preceding series, para-substitution of the phenyl rings was found toincrease potency, with iodo-susbstitution, e.g. (602), showing greater affinitythan Br-substitution, e.g. (598), which in turn was more potent than thecorresponding chloro analogue (593) (R2 ¼ iBu). Substitution on the N3-position was also found to have a pronounced influence on CB1 bindingaffinity. Increasing the chain length to over six carbon atoms resulted in amarked loss of activity. Benzyl substitution (594) was better tolerated thanthe corresponding phenyl ethyl congener (595), while the truncated allylgroup, typified by analogues (600) and (603), was found to be optimal in thisseries. The most potent thiohydantoin analogue identified was found to beconsiderably less potent than rimonabant (382) (K i ¼ 589 and 5.4 nM, re-spectively), however, the compounds were selective over the CB2 receptor.Compounds (593), (594) and (600)–(602) were examined in the [35S]-GTPgSbinding assay and, like the related hydantoin series, found to behave asinverse agonists at the CB1 receptor.

Table 6.50 3-ALKYL-5,50-DIPHENYLTHIOXOIMIDAZOLIDIN-4-ONES – IN VITRODATA [349]

N

NH

S

O

R1

R1

R2

Cpd. R1 R2 CB1 Ki (nM)a Cpd. R1 R2 CB1 Ki (nM)a

(593) Cl i-Bu 2,089 (600) Br Allyl 871(594) Cl CH2Ph 2,188 (601) Br CH2Ph 993(595) Cl (CH2)2Ph 3,801 (602) I n-Bu 724(596) Br Et 2,193 (603) I Allyl 589(597) Br i-Pr 3,630 (382)a 5.4(598) Br n-Bu 1,412 (254a)b 3,802(599) Br i-Bu 1,778 (165)c 18.6

aRimonabant.bWIN55,212-2.cHU-210.

Page 101: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 307

An analogue of (-)-cannabidiol, O-2654 (604) has recently been disclosed asa high-affinity CB1 antagonist ligand [350]. Compound (604) was found to besignificantly more potent (K i ¼ 114 nM) than cannabidiol (K i ¼ 4:9mM) inthe displacement of tritium-labelled CP 55,940 in mouse brain tissues. Thevalue obtained for (604), while less potent than that reported for (382)(1.98–12.3 nM), is comparable with the early Lilly antagonist LY320135(K i ¼ 141 nM) [351]. In addition, (604) was found to antagonise WIN 55,212-2 by competing with the agonist for the CB1 receptors. Unlike (382), thecannabidiol analogue is thought to behave as a neutral antagonist.

RECENT CB1 RECEPTOR ANTAGONISTS

A recent patent application from Roche [352] described a 2-amino-benzothiazole series. Roche claimed that compound (605) exhibited anIC50 value of 0.73 mM at CB1, and showed in excess of 10-fold selectivityover the CB2 receptor. The compounds were described as being of potentialuse in the treatment of a range of diseases, including CNS and psychiatricdisorders, type-2 diabetes, gastrointestinal diseases, cardiovascular disor-ders, infertility disorders, inflammation, cancer, atherosclerosis, cerebralvascular incidents and cranial trauma.A series of 12 tetrahydroquinoline compounds, functioning as antagonists

or inverse agonists of the CB1 receptor, has been disclosed [353]. The req-uisite three hydrophobic groups, in the example (606), three unsubstitutedphenyl groups are present, as is an adjacent hydrogen-bond acceptor (eitherthe 2-oxo group of the central heterocycle, or the sulfonamide oxygen). Asulfonamide moiety was also present in a patent application, which at thetime of writing is the most recent disclosure from Merck and Co. [354].Again, a branched butyl chain forms the backbone from which two arylgroups are appended (607). CB1 functional activity for the compounds wasclaimed to be lower than 1 mM while the CB2 functional activity was re-ported to be higher than 1 mM.

Me

OH

OHMe

(CH2)3N3

N

S

O

MeCl

ClCl(604) (605)

Page 102: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH308

N

Ph

Ph

NHSO2Ph

OMe

NHSO2Et

Cl

Cl

(606) (607)

THERAPEUTIC APPLICATIONS OFCB1 RECEPTOR ANTAGONISTS

Our current understanding of the potential therapeutic applications of CB1

antagonists owes a great deal to the discovery of rimonabant (382). Indeed,clinical data demonstrating the efficacy of (382) in the treatment of obesityand nicotine addiction has provided a substantial driving force for the ex-panding research effort into this approach.The increase in appetite resulting from cannabis use has long been rec-

ognised and has also been observed in animals that have been administeredcannabinoid agonists [355]. The endocannabinoid system also appears toplay a key role in the regulation of appetite [356]. Endocannabinoid levels inthe limbic forebrain have been shown to increase with food deprivation,while 2-AG levels in the hypothalamus reduce during feeding [357]. Anand-amide (1) administration into the ventromedial hypothalamus has also beenshown to stimulate appetite in rats [358]. The hypothalamus uses leptin asthe primary signal to modulate food intake and energy balance and there isevidence linking leptin and endocannabinoids in the regulation of food in-take [359]. Di Marzo and co-workers associated defective leptin signallingwith elevated hypothalamic levels of endocannabinoids in obese db/db andob/ob mice and Zucker rats, cerebellar endocannabinoid levels were notaffected. Acute leptin treatment of normal rats and ob/ob mice reduced (1)and 2-AG levels in the hypothalamus.In additon to the central role of endocannabinoids in the regulation of

feeding behaviour, a peripheral role has also been described. Gomez and co-workers [360] reported that food deprivation produced a 7-fold reduction in(1) levels in the small intestine of rats, but not in the brain or stomach.Intestinal (1) levels returned to normal when feeding resumed. The authorsalso showed that peripheral, but not central administration of (382) reducedfood intake. The endocannabinoid system has also been reported to regulateperipheral lipogenesis [361].

Page 103: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 309

The role of the endocannabinoids and exogenously administered agonistsoutlined above led to an interest in testing the effects of selective CB1 receptorantagonists on feeding behaviour [261, 362, 363]. Rimonabant (382) andSR147778 (385) have been shown to reduce food intake and weight gain inordinary rats without altering water intake [270, 364–366]. Rimonabant (382)has also been shown to cause a transient decrease in food intake accompaniedby a sustained decrease in bodyweight and adiposity in dietary-induced obesemice [367]. Leptin, insulin and glucose levels were all normalised in these micefollowing (382) treatment, together with an increase in serum adiponectinlevels [367, 368]. While (382) did not modify HDL-cholesterol and had mod-est effects on total cholesterol, it significantly reduced triglycerides and LDL-cholesterol and increased the HDL/LDL-cholesterol ratio.CB1 knock-out mice exhibit resistance to dietary-induced obesity in terms

of food intake, weight gain, body fat composition and the development ofdietary-induced insulin resistance [369]. The effects of (382) on feeding be-haviour and body weight are not observed in these animals, confirming thatthese are mediated through the CB1 receptor [359, 367]. CB1 knock-out micedo not demonstrate a decrease in food intake under free feeding conditions,but food intake is reduced following food deprivation.Clinical trials with (382) have provided the most exciting evidence for the

utility of CB1 antagonists in the treatment of obesity, results of the Rimo-nabant in Obesity (RIO)-Europe trial having been published very recently[370, 371]. In this trial, 1,507 patients with body-mass index of 30 kg/m2 orgreater, or body-mass index greater than 27 kg/m2 with treated or untreateddyslipidaemia, hypertension or both, were randomised to receive double-blind treatment with placebo, 5mg (382) or 20mg (382) once daily in ad-dition to a mild hypocaloric diet (600 kcal/day deficit). The primary efficacyendpoint was weight change from baseline after 1-year treatment in theintention-to-treat population. Weight loss at 1 year was significantly greaterin patients treated with (382) 5mg (mean �3.4 kg) and 20mg (�6.6 kg)compared to placebo (�1.8 kg). Significantly more patients treated with(382) 20mg than placebo achieved weight loss of 5% or greater (50.9%versus 19.2%) and 10% or greater (27.4% versus 7.3%). Rimonabant (382),20mg, produced significantly greater improvements than placebo in waistcircumference, HDL-cholesterol, triglycerides, insulin resistance and prev-alence of metabolic syndrome. The effects of (382) 5mg were of less sig-nificance. (382) was generally well tolerated with mild and transient sideeffects including nausea, dizziness, diarrhoea and vomiting.Rimonabant (382) has also shown promise in pre-clinical and clinical

studies as an aid to smoking cessation. (382) decreases nicotine self-admin-istration in rats and nicotine-induced dopamine release in nucleus acumbens[372], and also reversed nicotine-seeking behaviour in rats several weeks

Page 104: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH310

after nicotine withdrawal [373]. In a 10-week, placebo-controlled trial, (382)was shown to prolong abstinence rates from tobacco during the final 4weeks of the treatment period [370]. Furthermore, while patients in placebogroups in several studies gained weight, (382) patients lost weight or expe-rienced less weight gain than those on placebo. Weight gain following nic-otine withdrawal is a major factor in reducing abstinence rates. In additionto their utility as aids to smoking cessation, CB1 receptor antagonists haveshown potential pre-clinically in the treatment of addiction to other sub-stances including morphine, heroin and alcohol [374–377].Rimonabant (382) was also included in a clinical study to assess the safety

and efficacy of four novel compounds for the treatment of schizophreniaand psychoaffective disorder [378]. The other compounds included in thetrial were a neurokinin NK3 antagonist, a serotonin 2A/2C antagonist and aneurotensin NTS1 antagonist. Haloperidol and placebo groups were used ascontrols in the study. Sixty-nine patients received (382) (20mg once perday), which failed to demonstrate efficacy in this trial. The reasons for thelack of efficacy may be due to inadequate dosing or an indication that CB1

antagonism is not appropriate in the treatment of this condition.Pre-clinical data support the potential application of CB1 antagonists in

the treatment of various other conditions. These include memory disorders[379], sexual dysfunction [380], neuro-inflammation [381] and asthma [382].

CB2 RECEPTOR ANTAGONISTS

CB2 receptor antagonists have received much less attention than their CB1

counterparts, with only a relatively small number of compounds availableand less clarity on their potential therapeutic role. A selection of the avail-able compounds that have been shown to act as antagonists of the CB2

receptor and their suggested utilities will be covered in this section.As observed in the CB1 antagonist section, pyrazole derivatives again

form an important class of antagonists for the CB2 receptor. A structuralanalogue of rimonabant (382), SR144528 (608), is a potent CB2 antagonist/inverse agonist identified by Sanofi [383]. This compound has proved to be auseful tool in determining the function of CB2 receptors. The compound hassubnanomolar affinity for CB2 receptors with 700-fold selectivity over CB1

receptors. Following oral administration, (608) totally displaced the ex vivo[3H]-CP 55,940 binding to mouse spleen membranes without interactingwith CB1 receptors in the brain.Conformationally restricted pyrazole-derived CB2 selective antagonists

were described in a patent application from Sanofi-Synthelabo [384]. Com-pounds included in the application, exemplified by compound (609) are

Page 105: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 311

claimed to act as antagonists at CB2 receptors (Kio5� 10�7M) with se-lectivity of at least 10-fold over CB1 receptors.Moving away from pyrazole-derived compounds, an aminoalkyl-indole,

AM630 (610), acts as a CB2 receptor antagonist/inverse agonist with a CB2 Ki

value of 31.2nM and 165-fold selectivity over the CB1 receptor subtype [385].It is interesting to note that this compound has been shown to act as a low-affinity partial agonist, antagonist or inverse agonist at CB1 receptors [6].Iwamura and Ueda [386] described compound (611) as a CB2 selective

inverse agonist in a patent application. The potential therapeutic roles ofCB2 antagonists are not clearly defined at the moment, although roles inregulation of the immune system and inflammation have been widely pro-posed. This patent application describes that activity of compound (611) ina mouse model of asthma, in which the compound suppressed immediateand late-phase asthmatic response and airway hyper-responsiveness.

NN

Cl

NH

O

NN

Cl

NH

O N

Cl

Cl

S

(608) SR144528 (609)

NI

N

O

O

MeO

NH

NH

O

O

O

O

O

O

(610) AM630 (611)

Page 106: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH312

A series of tricyclic pyrazoles has been synthesised and their CB1 and CB2

affinity determined in mouse cerebellum membranes and mouse spleen ho-mogenate, respectively [387]. Table 6.51 describes some of the pertinent data.Despite the close structural similarity between rimonabant (382) and the

fused pyrazole analogues described by Mussinu and co-workers [276], themajority of the latter show a moderate-to-strong preference for the CB2

receptor. In particular, (612), the closest homologue to (382) within theseries, shows very low nanomolar affinity (K i ¼ 0:34 nM) at CB2, while onlycomparatively modest CB1 affinity (2,050 nM). 6-Fluoro (613) and 6-bromo(614) analogues were also potent and selective CB2 ligands, while substi-tution of the larger 6-iodo group (615) resulted in a decrease in both affinityand selectivity. The unsubstituted analogue (618) exhibited a profile similar

Table 6.51 TRICYCLIC PYRAZOLE DERIVATIVES – CB1 AND CB2 BINDINGDATA [387]

N N

NHR4

O

R1

R2

R3

Cpd. R1 R2/R3 R4 CB1 Ki (nM)ax CB2 Ki (nM)ay CB1 Ki/CB2 Ki

(382)a 1.8 514 0.0035(612) 6-Cl 2,4-di-Cl N-piperidine 2,050 0.34 6,029(613) 6-F 2,4-di-Cl N-piperidine 1,268 0.225 5,635(614) 6-Br 2,4-di-Cl N-piperidine 1,570 0.27 5,814(615) 6-I 2,4-di-Cl N-piperidine 333 5.5 60(616) 5-Cl 2,4-di-Cl N-piperidine 8.25 0.23 3,587(617) 7-Cl 2,4-di-Cl N-piperidine 723 6.79 105(618) H 2,4-di-Cl N-piperidine 1152 0.385 2,992(619) 6-Me 2,4-di-Cl N-piperidine 363 0.04 9,810(620) 6-OMe 2,4-di-Cl N-piperidine 399 12.3 32(621) 6-Cl 4-Cl N-piperidine 1,787 0.9 1,985(622) 6-Cl H N-piperidine >5,000 48 104(623) 6-Cl 4-OMe N-piperidine 3,035 120 25(624) 6-Cl 2,4-di-Cl N-pyrrolidine 798 9.9 81(625) 6-Cl 2,4-di-Cl N(Me)2 1,881 144 13(626) 6-Cl 2,4-di-Cl NH2 2,183 455 5

aRimonabant.

Page 107: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 313

to that of the 6-Cl congener (612). Movement of the 6-Cl substituent to C5slightly increased CB2 affinity, while the 7-Cl analogue (617) witnessed aslight decrease in CB2 binding. The 6-Me analogue (619) was found to be themost potent and selective ligand, eclipsing the 250-fold CB2-selective Sanofi-Aventis ligand, SR144528 (608).

SUMMARY AND FUTURE PROSPECTS

It is hoped that the scope of this review article has given the reader a goodimpression of the rate at which cannabinoid research is progressing. The lastdecade has seen a rapid growth in cannabinoid research, which looks set tocontinue. The therapeutic potential of cannabinoid agonists has long beenacknowledged through the use of cannabis for medicinal purposes. How-ever, the accompanying psychotropic effects have restricted the usefulness ofthis approach. We are now starting to observe that at least some of thebenefits of cannabinoids can be realised without CNS side effects throughstrategies such as peripheral restriction of compounds, CB2 subtype selec-tivity and subtle modulation of the endocannabinoid system. Improved drugdelivery systems are also proving beneficial in this regard, although beyondthe scope of this review.Antagonism of cannabinoid receptors is now attracting a huge amount of

attention, following on the successful rimonabant (382) clinical trials. At thetime of writing, a launch date in 2006 is anticipated for (382) and it will beintriguing to observe the future of this mechanistic approach in the treat-ment of various conditions, particularly obesity. With an ever-growingknowledge of the endocannabinoid system, and the possible identification offurther receptor subtypes, the future of cannabinoid research seems verybright indeed.

REFERENCES

[1] Mechoulam, R., Hanus, L. and Fride, E. (1998) Prog. Med. Chem. 35, 199–243.[2] Barth, F. (1998) Expert. Opin. Ther. Patents 8, 301–313.[3] Goya, P. and Jagerovic, N. (2000) Expert. Opin. Ther. Patents 10, 1529–1538.[4] Adam, J. and Cowley, P. (2002) Expert. Opin. Ther. Patents 12, 1475–1489.[5] Herzog, D.L. (2004) Expert. Opin. Ther. Patents 14, 1435–1452.[6] Howlett, A.C., Barth, F., Bonner, T.I., Cabral, G., Casellas, P., Devane, W.A.,

Felder, C.C., Herkenham, M., Mackie, K., Martin, B.R., Mechoulam, R. andPertwee, R.G. (2002) Pharm. Rev. 54, 161–202.

[7] Palmer, S.L., Thakur, G.A. and Makriyannis, A. (2002) Chem. Phys. Lipids 121, 3–19.[8] Begg, M., Pacher, P., Batkai, S., Osei-Hyiaman, D., Offertaler, L., Mo, F.M., Liu, J. and

Kunos, G. (2005) Pharm. Ther. 106, 133–145.

Page 108: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH314

[9] Drmota, T., Greasley, P., Groblewski, T. (2004) PCT Int. Appl., WO 04, 74844; (2004)Chem. Abstr. 141, 238–643.

[10] De Petrocellis, L., Cascio, M.G. and Di Marzo, V. (2004) Br. J. Pharmacol. 141,765–774.

[11] Di Marzo, V., Bifulco, M. and De Petrocellis, L. (2004) Nature Rev. Drug Disc. 3,771–784.

[12] Piomelli, D. (2003) Nature Rev. Neurosci. 4, 873–884.[13] Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G.,

Gibson, D., Mandelbaum, A., Etinger, A. and Mechoulam, R. (1992) Science

(Washington DC) 258, 1946–1949.[14] Mechoulam, R., Ben-Shabat, S., Hanus, L., Ligumsky, M., Kaminski, N.E., Schatz, A.R.,

Gopher, A., Almog, S., Martin, B.R. and Compton, D.R. (1995) Biochem. Pharmacol. 50,83–90.

[15] Sugiura, T., Kondo, S., Sukagawa, A., Nakane, S., Shinoda, A., Itoh, K., Yamashita, A.and Waku, K. (1995) Biochem. Biophys. Res. Commun. 215, 89–97.

[16] Hanus, L., Abu-Lafi, S., Fride, S., Breuer, A., Vogel, Z., Shalev, D.E., Kustanovich, I.and Mechoulam, R. (2001) Proc. Natl. Acad. Sci. USA 98, 3662–3665.

[17] Porter, A.C., Sauer, J.-M., Knierman, M.D., Becker, G.W., Berna, M.J., Bao, J.,Nomikos, G.G., Carter, P., Bymaster, F.P., Leese, A.B. and Felder, C.C. (2002)J. Pharmacol. Exp. Ther. 301, 1020–1024.

[18] Huang, S.M., Bisogno, T., Trevisani, M., Al-Hayani, A., De Petrocellis, L., Fezza, F.,Tognetto, M., Petros, T.J., Krey, J.F., Chu, C.J., Miller, J.D., Davies, S.N., Geppetti, P.,Walker, J.M. and Di Marzo, V. (2002) Proc. Natl. Acad. Sci. USA 99, 8400–8405.

[19] Oka, S., Tsuchie, A., Tokumura, A., Muramatsu, M., Suhara, Y., Takayama, H.,Waku, K. and Sugiura, T. (2003) J. Neurochem. 85, 1374–1381.

[20] Battista, N., Gasperi, V., Fezza, F. and Maccarone, M. (2005) Therapy 2, 141–150.[21] Giuffrida, A., Beltramo, M. and Piomelli, D. (2001) J. Pharmacol. Exp. Ther. 298, 7–14.[22] Fowler, C.J. and Jacobbsen, S.O. (2002) Prostaglandins Leukot. Essent. Fatty Acids 66,

193–200.[23] Glaser, S.T., Abumrad, N.A., Fatade, F., Kaczocha, M., Studholme, K.M. and

Deutsch, D.G. (2003) Proc. Natl. Acad. Sci. USA 100, 4269–4274.[24] Fegley, D., Kathuria, S., Mercier, R., Li, C., Goutopoulos, A., Makriyannis, A. and

Piomelli, D. (2004) Proc. Natl. Acad. Sci. USA 101, 8756–8761.[25] Cravatt, B.F., Giang, G.K., Mayfield, S.P., Boger, D.L., Lerner, R.A. and Gilula, N.B.

(1996) Nature (London) 384, 83–87.[26] Cravatt, B.F. and Lichtman, A.H. (2003) Curr. Opin. Chem. Biol. 7, 469–475.[27] Deutsch, D.G., Ueda, N. and Yamamoto, S. (2002) Prostaglandins Leukot. Essent. Fatty

Acids 66, 201–210.[28] Fowler, C.J. (2004) Curr. Med. Chem.: Central Nervous System Agents 4, 161–174.[29] Cravatt, B.F., Demarest, K., Patricelli, M.P., Bracey, M.H., Giang, D.K., Martin, B.R.

and Lichtman, A.H. (2001) Proc. Natl. Acad. Sci. USA 98, 9371–9376.[30] Lichtman, A.H., Hawkins, E.G., Griffin, G. and Cravatt, B.F. (2002) J. Pharmacol. Exp.

Ther. 302, 73–79.[31] Lopez-Rodrıguez, M.L., Viso, A., Ortega-Gutierrez, S., Fernandez-Ruiz, J. and

Ramos, J.A. (2002) Curr. Med. Chem: Central Nervous System Agents 2, 129–141.[32] McFarland, M.J. and Barker, E.L. (2004) Pharmacol. Ther. 104, 117–135.[33] Piomelli, D., Beltramo, M., Glasnapp, S., Lin, S.Y., Xie Goutopoulos, X.-Q. and

Makriyannis, A. (1999) Proc. Natl. Acad. Sci. USA 96, 5802–5807.[34] Lopez-Rodrıguez, M.L., Viso, A., Ortega-Gutierrez, S., Fowler, C.J., Tiger, G., Lago, E.

and Fernandez-Ruiz, J. (2003) J.A. Ramos J. Med. Chem. 46, 1512–1522.

Page 109: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 315

[35] Makriyannis, A., Goutopoulos, A., Li, C., US Appl., US 05 20679; (2005) Chem. Abstr.142 (2005) 176603.

[36] Ortar, G., Ligresti, A., De Petrocellis, L., Morera, E. and Di Marzo, V. (2003) Biochem.

Pharmacol. 65, 1473–1481.[37] Jarrahian, A., Manna, S., Edgemond, W.S., Campbell, W.B. and Hilliard, C.J. (2000)

J. Neurochem. 74, 2597–2606.[38] de Lago, E., Fernandez-Ruiz, J.J., Ortega-Gutierrez, S., Viso, A., Lopez-Rodrıguez, M.L.

and Ramos, J.A. (2002) Eur. J. Pharmacol. 449, 99–103.[39] Schmid, P.C., Zuzarte-Augustin, M.L. and Schmid, H.H.O. (1985) J. Biol. Chem. 260,

14145–14149.[40] Bracey, M.H., Hanson, M.A., Masuda, K.R., Stevens, R.C. and Cravatt, B.F. (2002)

Science (Washington DC) 298, 1793–1796.[41] Desarnaud, F., Cadas, H. and Piomelli, D. (1995) J. Biol. Chem. 270, 6030–6035.[42] Ueda, N., Kurahashi, Y., Yamamoto, S. and Tokunaga, T. (1995) J. Biol. Chem. 270,

23823–23827.[43] Bisogno, T., Maurelli, S., Melck, D., De Petrocellis, L. and Di Marzo, V. (1997) J. Biol.

Chem. 272, 3315–3323.[44] Maccarrone, M., van der Stelt, M., Rossi, A., Veldink, G.A., Vliegenthart, J.F.G. and

Agro, A.F. (1998) J. Biol. Chem. 273, 32332–32339.[45] Bisogno, T., De Petrocellis, L. and Di Marzo, V. (2002) Curr. Pharm. Design 8, 125–133.[46] Lichtman, A.H. and Cravatt, B.F. (2002) Chem. Phys. Lipids 121, 135–148.[47] Deutsch, D.G. and Chin, S.A. (1993) Biochem. Pharmacol. 46, 791–796.[48] Abadji, V., Lin, S., Taha, G., Griffin, G., Stevenson, L.A., Pertwee, R.G. and

Makriyannis, A. (1994) J. Med. Chem. 37, 1889–1893.[49] Childers, S.R., Sexton, T. and Roy, M.B. (1994) Biochem. Pharmacol. 47, 711–715.[50] Pertwee, R.G., Fernando, S.R., Griffin, G., Abadji, V. and Makriyannis, A. (1995) Eur.

J. Pharmacol. 272, 73–78.[51] Compton, D.R. and Martin, B.R. (1997) J. Pharmacol. Exp. Ther. 283, 1138–1143.[52] Wiley, J.L., Dewey, M.A., Jefferson, R.G., Winckler, R.L., Bridgen, D.T., Willoughby,

K.A. and Martin, B.R. (2000) Life Sci. 67, 1573–1583.[53] Deutsch, D.G., Lin, S., Hill, W.A.G., Morse, K.L., Salehani, D., Arreaza, G., Omeir, R.L.

and Makriyannis, A. (1997) Biochem. Biophys. Res. Commun. 231, 217–221.[54] Segall, Y., Quistad, G.B., Nomura, D.K. and Casida, J.E. (2003) Bioorg. Med. Chem.

Lett. 13, 3301–3303.[55] Gifford, A.N., Bruneus, M., Lin, S., Goutopoulos, A., Makriyannis, A., Volkow, N.D.

and Gatley, S.J. (1999) Eur. J. Pharmacol. 383, 9–14.[56] Arizzi, M.N., Cervone, K.M., Aberman, J.E., Betz, A., Liu, Q., Lin, S., Makriyannis, A.

and Salamone, J.D. (2004) Life Sci. 74, 1001–1011.[57] Deutsch, D.G., Omeir, R., Arreaza, G., Salehani, D., Prestwich, G.D., Huang, Z. and

Howlett, A. (1997) Biochem. Pharmacol. 53, 255–260.[58] Street, I.P., Lin, H.-K., Laliberte, F., Ghomashchi, F., Wang, Z., Perrier, H., Tremblay,

N.M., Huang, Z., Weech, P.K. and Gelb, M.H. (1993) Biochemistry 32, 5935–5940.[59] Huang, Z., Payette, P., Abdullah, K., Cromlish, W.A. and Kennedy, B.P. (1996)

Biochemistry 35, 3712–3721.[60] Fernando, S.R. and Pertwee, R.G. (1997) Br. J. Pharmacol. 121, 1716–1720.[61] Martin, B.R., Beletskaya, I., Patrick, G., Jefferson, R., Winckler, R., Deutsch, D.G.,

Di Marzo, V., Dasse, O., Mahadevan, A. and Razdan, R.J. (2000) J. Pharmacol. Exp.

Ther. 294, 1209–1218.[62] Koutek, B., Prestwich, G.D., Howlett, A.C., Chin, S.A., Salehani, D., Akhavan, N. and

Deutsch, D.G. (1994) J. Biol. Chem. 269, 22937–22940.

Page 110: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH316

[63] Boger, D.L., Sato, H., Lerner, A.E., Austin, B.J., Patterson, J.E., Patricelli, M.P. andCravatt, B.F. (1999) Bioorg. Med. Chem. Lett. 9, 265–270.

[64] Boger, D.L., Sato, H., Lerner, A.E., Hedrick, M.P., Fecik, R.A., Miyauchi, H., Wilkie, G.D.,Austin, B.J., Patricelli, M.P. and Cravatt, B.F. (2000) Proc. Natl. Acad. Sci. USA 97,5044–5049.

[65] Boger, D.L., Miyauchi, H., Du, W., Hardouin, C., Fecik, R.A., Cheng, H., Hwang, I.,Hedrick, M.P., Leung, D., Acevedo, O., Guimaraes, C.R.W., Jorgensen, W.L. andCravatt, B.F. (2005) J. Med. Chem. 48, 1849–1856.

[66] Leung, D., Du, W., Hardouin, C., Cheng, H., Hwang, I., Cravatt, B.F. and Boger, D.L.(2005) Bioorg. Med. Chem. Lett. 15, 1423–1428.

[67] Lichtman, A.H., Leung, D., Shelton, C.C., Saghatelian, A., Hardouin, C., Boger, D.L.and Cravatt, B.F. (2004) J. Pharmacol. Exp. Ther. 311, 441–448.

[68] Du, W., Heng, H., Cheng, H., Hwang, I. and Boger, D.L. (2005) Bioorg. Med. Chem.

Lett. 15, 103–106.[69] Tarzia, G., Duranti, A., Tontini, A., Piersanti, G., Mor, M., Rivara, S., Plazzi, P.V.,

Park, C., Kathuria, S. and Piomelli, D. (2003) J. Med. Chem. 46, 2352–2360.[70] Mor, M., Rivara, S., Lodola, A., Plazzi, P.V., Tarzia, G., Duranti, A., Tontini, A.,

Piersanti, G., Kathuria, S. and Piomelli, D. (2004) J. Med. Chem. 47, 4998–5008.[71] Kathuria, S., Gaetani, S., Fegley, D., Valino, F., Duranti, A., Tontini, A., Mor, M.,

Tarzia, G., La Rana, G., Calignano, A., Giustino, A., Tattoli, M., Palmery, M., Cuomo,V. and Piomelli, D. (2003) Nature Medicine 9, 76–81.

[72] Sit, S.-Y., Xie, K. (2002) PCT Int. Appl., WO 02 87569; (2002) Chem. Abstr. 137,

353021.[73] Sit, S.-Y., Xie, K., Deng, H. (2003) PCT Int. Appl., WO 03 65989; (2003) Chem. Abstr.

139, 179893.[74] Abouabdellah, A., Bas, M., Dargazanli, G. (2004) PCT Int. Appl., WO 04 20430; (2004)

Chem. Abstr. 140, 235723.[75] Abouabdellah, A., Burnier, P., Hoornaert, C., Jeunesse, J., Puech, F. (2004) PCT Int.

Appl., WO 04 99176; (2005) Chem. Abstr. 142, 198102.[76] Bisogno, T., Melck, D., De Petrocellis, L., Bobrov, M.Y., Gretskaya, N.M., Bezuglov, V.V.,

Sitachitta, N., Gerwick, W.H. and Di Marzo, V. (1998) Biochem. Biophys. Res. Commun.

248, 515–522.[77] Vandervoorde, S., Jonsson, K.-O., Fowler, C.J. and Lambert, D.M. (2003) J. Med.

Chem. 46, 1440–1448.[78] Jonsson, K.-O., Vandervoorde, S., Lambert, D.M., Tiger, G. and Fowler, C.J. (2001)

Br. J. Pharmacol. 133, 1263–1275.[79] Patricelli, M.P., Patterson, J.E., Boger, D.L. and Cravatt, B.F. (1998) Bioorg. Med.

Chem. Lett. 8, 613–618.[80] Martin, B.R., Compton, D.R., Semus, S.F., Lin, S., Marciniak, G., Grzybowska, J.,

Charalambous, A. and Makriyannis, A. (1993) Pharmacol. Biochem. Behav. 46, 295–301.[81] Showalter, V.M., Compton, D.R., Martin, B.R. and Abood, M.E. (1996) J. Pharmacol.

Exp. Ther. 278, 989–999.[82] Papahatjis, D.P., Nikas, S.P., Andreou, T. and Makriyannis, A. (2002) Bioorg. Med.

Chem. Lett. 12, 3583–3586.[83] Razdan, R.K. (1986) Pharmacol. Rev. 38, 75–149.[84] Makriyannis, A. and Rapaka, R.S. (1990) Life Sci. 47, 2173–2184.[85] Seltzman, H.H. (1999) Curr. Med. Chem. 6, 685–704.[86] Reggio, P.H. (2003) Curr. Pharm. Des. 9, 1607–1633.[87] Fichera, M., Cruciani, G., Bianchi, A. and Musumarra, G. (2000) J. Med. Chem. 43,

2300–2309.

Page 111: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 317

[88] Schmetzer, S., Greenidge, P., Kovar, K.-A., Schulze-Alexandru, M. and Folkers, G.(1997) J. Comput. Aided Mol. Des. 11, 278–292.

[89] Thomas, B.F., Compton, D.R., Martin, B.R. and Semus, S.F. (1991) Mol. Pharmacol.

40, 656–665.[90] Gaoni, Y. and Mechoulam, R. (1964) J. Am. Chem. Soc. 86, 1646–1647.[91] Adams, R., MacKenzie, S. and Loewe, S. (1948) J. Am. Chem. Soc. 70, 664–668.[92] Adams, R., Harfenist, M. and Loewe, S. (1949) J. Am. Chem. Soc. 71, 1624–1628.[93] Compton, D.R., Rice, K.C., De Costa, B.R., Razdan, R.K., Melvin, L.S., Johnson, M.R.

and Martin, B.R. (1993) J. Pharmacol. Exp. Ther. 265, 218–226.[94] Huffman, J.W., Lainton, J.A.H., Banner, W.K., Duncan, S.G., Jordan, R.D., Yu, S., Dai, D.,

Martin, B.R., Wiley, J.L. and Compton, D.R. (1997) Tetrahedron 53, 1557–1576.[95] Huffman, J.W., Liddle, J., Duncan, S.G., Yu, S., Martin, B.R. and Wiley, J.L. (1998)

Bioorg. Med. Chem. 6, 2383–2396.[96] Huffman, J.W. (2003) Bioorg. Med. Chem. 11, 1397–1410.[97] Nadipuram, A.K., Krishnamurthy, M., Ferreira, A.M., Li, W. and Moore, B.M. (2003)

Bioorg. Med. Chem. 11, 3121–3132.[98] Martin, B.R., Jefferson, R., Winckler, R., Wiley, J.L., Huffman, J.W., Crocker, P.J.,

Saha, B. and Razdan, R.K. (1999) J. Pharmacol. Exp. Ther. 290, 1065–1079.[99] Khanolkar, A.D., Lu, D., Fan, P., Tian, X. and Makriyannis, A. (1999) Bioorg. Med.

Chem. Lett. 9, 2119–2124.[100] Khanolkar, A.D., Palmer, S.L. and Makriyannis, A. (2000) Chem. Phys. Lipids 108,

37–52.[101] Huffman, J.W., Duncan, S.G., Wiley, J.L. and Martin, B.R. (1997) Bioorg. Med. Chem.

Lett. 7, 2799–2804.[102] Papahatjis, D.P., Nikas, S.P., Kourouli, T., Chari, R., Xu, W., Pertwee, R.G. and

Makriyannis, A. (2003) J. Med. Chem. 46, 3221–3229.[103] Luk, T., Jin, W., Zvonok, A., Lu, D., Lin, X.-Z., Chavkin, C., Makriyannis, A. and

Mackie, K. (2004) Br. J. Pharmacol. 142, 495–500.[104] Ryan, W., Singer, M., Razdan, R.K., Compton, D.R. and Martin, B.R. (1995) Life Sci.

56, 2013–2020.[105] Papahatjis, D.P., Kourouli, T., Abadji, V., Goutopoulos, A. and Makriyannis, A. (1998)

J. Med. Chem. 41, 1195–1200.[106] Griffin, G., Wray, E.J., Rorrer, W.K., Crocker, P.J., Ryan, W.J., Saha, B., Razdan, R.K.,

Martin, B.R. and Abood, M.E. (1999) Br. J. Pharmacol. 126, 1575–1584.[107] Griffin, G., Williams, S., Aung, M., Razdan, R.K., Martin, B.R. and Abood, M.E.

(2001) Br. J. Pharmacol. 132, 525–535.[108] Keimowitz, A.R., Martin, B.R., Razdan, R.K., Crocker, P.J., Mascarella, S.W. and

Thomas, B.F. (2000) J. Med. Chem. 43, 59–70.[109] Krishnamurthy, M., Ferreira, A.M., Li, W. and Moore, B.M. (2003) Bioorg. Med.

Chem. Lett. 13, 3487–3490.[110] Nikas, S.P., Grzybowska, J., Papahatjis, D.P., Charalambous, A., Banijamali, A.R.,

Chari, R., Fan, P., Kourouli, T., Lin, S., Nitowski, A.J., Marciniak, G., Guo, Y., Li, X.,Wang, C.-L.J., Makriyannis, A. (2004) The AAPS J. 6, Article 30.

[111] Moore, B.M., Ferreira, A.M., Krishnamurthy, M. (2004) US Appl., US 04 242593;(2005) Chem. Abstr. 142, 23402.

[112] Charalambous, A., Lin, S., Marciniak, G., Banijamali, A., Friend, F.L., Compton,D.R., Martin, B.R. and Makriyannis, A. (1991) Pharmacol. Biochem. Behav. 40,509–512.

[113] Singer, M., Ryan, W.J., Saha, B., Martin, B.R. and Razdan, R.K. (1998) J. Med. Chem.

41, 4400–4407.

Page 112: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH318

[114] Ross, R.A., Gibson, T.M., Stevenson, L.A., Saha, B., Crocker, P., Razdan, R.K. andPertwee, R.G. (1999) Br. J. Pharmacol. 128, 735–743.

[115] Watanabe, K., Yamamoto, I., Oguri, K. and Yoshimura, H. (1980) Eur. J. Pharmacol.

63, 1–6.[116] Robertson, L.R., Duffley, R.P., Razdan, R.K., Martin, B.R., Harris, L.S. and Dewey, W.L.

(1984) J. Med. Chem. 27, 550–552.[117] Rhee, M.-H., Vogel, Z., Barg, J., Bayewitch, M., Levy, R., Hanus, L., Breuer, A. and

Mechoulam, R. (1997) J. Med. Chem. 40, 3228–3233.[118] Martin, B.R. (1985) Life Sci. 36, 1523–1530.[119] Ottani, A. and Giuliani, D. (2001) CNS Drug Rev. 7, 131–145.[120] Howlett, A.C., Champion, T.M., Wilken, G.H. and Mechoulam, R. (1990) Neurophar-

macology 29, 161–165.[121] Burstein, S.H. (2000) Curr. Pharm. Des. 6, 1339–1345.[122] Burstein, S.H. (1999) Pharmacol. Ther. 82, 87–96.[123] Burstein, S.H., Karst, M., Schneider, U. and Zurier, R.B. (2004) Life Sci. 75,

1513–1522.[124] Bisogno, T., Hanus, L., De Petrocellis, L., Tchilibon, S., Ponde, D.E., Brandi, I.,

Moriello, A.S., Davis, J.B., Mechoulam, R. and Di Marzo, V. (2001) Br. J. Pharmacol.

134, 845–852.[125] Fride, E., Feigin, C., Ponde, D.E., Breuer, A., Hanus, L., Arshavsky, N. and Mechoulam, R.

(2004) Eur. J. Pharmacol. 506, 179–188.[126] Hanus, L., Tchilibon, S., Ponde, D.E., Breuer, A., Fride, E. and Mechoulam, R. (2005)

Org. Biomol. Chem. 3, 1116–1123.[127] Mechoulam, R., Fride, E. (2005) PCT Int. Appl., WO 05 23741; (2005) Chem. Abstr. 142,

316978.[128] Kogan, N.M., Rabinowitz, R., Levi, P., Gibson, D., Sandor, P., Schlesinger, M. and

Mechoulam, R. (2004) J. Med. Chem. 47, 3800–3806.[129] Johnson, M.R., Melvin, L.S. (1986) In ‘‘Cannabinoids as Therapeutic Agents’’.

pp. 121–146, CRC Press, Boca Raton.[130] Melvin, L.S., Johnson, M.R., Harbert, C.A., Milne, G.M. and Weissman, A. (1984)

J. Med. Chem. 27, 67–71.[131] Xie, X.-Q., Yang, D.-P., Melvin, L.S. and Makriyannis, A. (1994) J. Med. Chem. 37,

1418–1426.[132] Melvin, L.S., Milne, G.M., Johnson, M.R., Subramaniam, B., Wilken, G.H. and

Howlett, A.C. (1993) Mol. Pharmacol. 44, 1008–1015.[133] Xie, X.-Q., Melvin, L.S. and Makriyannis, A. (1996) J. Biol. Chem. 271, 10640–10647.[134] Devane, W.A., Dysarz, F.A., Johnson, M.R., Melvin, L.S. and Howlett, A.C. (1988)

Mol. Pharmacol. 34, 605–613.[135] Compton, D.R., Johnson, M.R., Melvin, L.S. and Martin, B.R. (1992) J. Pharmacol.

Exp. Ther. 260, 201–209.[136] Melvin, L.S., Milne, G.M., Johnson, M.R., Wilken, G.H. and Howlett, A.C. (1995)

Drug Des. Discov. 13, 155–166.[137] Tius, M.A., Hill, W.A.G., Zou, X.L., Busch-Petersen, J., Kawakami, J.K., Fernandez-

Garcia, M.C., Drake, D.J., Abadji, V. and Makriyannins, A. (1995) Life Sci. 56,2007–2012.

[138] Xie, X.-Q., Pavlopoulos, S., DiMeglio, C.M. and Makriyannins, A. (1998) J. Med.

Chem. 41, 167–174.[139] Shim, J.-Y., Welsh, W.J. and Howlett, A.C. (2003) Biopolymers 71, 169–189.[140] Drake, D.J., Jensen, R.S., Busch-Petersen, J., Kawakami, J.K., Fernandez-Garcia, M.C.,

Fan, P., Makriyannins, A. and Tius, M.A. (1998) J. Med. Chem. 41, 3596–3608.

Page 113: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 319

[141] Little, P.J., Compton, D.R., Johnson, M.R., Melvin, L.S. and Martin, B.R. (1988)J. Pharmacol. Exp. Ther. 247, 1046–1051.

[142] Reggio, P.H. (2002) Prostaglandins Leukot. Essent. Fatty Acids 66, 143–160.[143] Di Marzo, V., Bisogno, T., De Petrocellis, L., Melck, D. and Martin, B.R. (1999) Curr.

Med. Chem. 6, 721–744.[144] Hillard, C.J. and Campbell, W.B. (1997) J. Lipid Res. 38, 2383–2398.[145] Palmer, S.L., Khanolkar, A.D. and Makriyannis, A. (2000) Curr. Pharm. Des. 6,

1381–1397.[146] Martin, B.R., Mechoulam, R. and Razdan, R.K. (1999) Life Sci. 65, 573–595.[147] Fride, E. and Mechoulam, R. (1993) Eur. J. Pharmacol. 231, 313–314.[148] Martin, B.R., Compton, D.R., Prescott, W.R., Little, P.J., Razdan, R.K., Johnson, M.R.,

Melvin, L.S., Mechoulam, R. and Ward, S.J. (1991) Pharmacol. Biochem. Behav. 40,471–478.

[149] Priller, J., Briley, E.M., Mansouri, J., Devane, W.A., Mackie, K. and Felder, C.C. (1995)Mol. Pharmacol. 48, 288–292.

[150] Sheskin, T., Hanus, L., Slager, J., Vogel, Z. and Mechoulam, R. (1997) J. Med. Chem.

40, 659–667.[151] Pinto, J., Potie, F., Rice, K.C., Boring, D., Johnson, M.R., Evans, D.M., Wilken, G.H.,

Cantrell, C.H. and Howlett, A.C. (1994) Mol. Pharmacol. 46, 516–522.[152] van der Stelt, M., van Kuik, J.A., Bari, M., van Zadelhoff, G., Leeflang, B.R., Veldink, G.A.,

Finazzi-Aggro, A., Vliegenthart, J.F.G. and Maccarrone, M. (2002) J. Med. Chem. 45,3709–3720.

[153] Edgemond, W.S., Hillard, C.J., Falck, J.R., Kearn, C.S. and Campbell, W.B. (1998)Mol. Pharmacol. 54, 180–188.

[154] Hampson, A.J., Hill, W.A.G., Zan-Phillips, M., Makriyannis, A., Leung, E., Eglen, R.M.and Bornheim, L.M. (1995) Biochim. Biophys. Acta 1259, 173–179.

[155] Adams, I.B., Ryan, W., Singer, M., Razdan, R.K., Compton, D.R. and Martin, B.R.(1995) Life Sci. 56, 2041–2048.

[156] Adams, I.B., Ryan, W., Singer, M., Thomas, B.F., Compton, D.R., Razdan, R.K. andMartin, B.R. (1995) J. Pharmacol. Exp. Ther. 273, 1172–1181.

[157] Goutopoulos, A., Fan, P., Khanolkar, A.D., Xie, X.-Q., Lin, S. and Makriyannis, A.(2001) Bioorg. Med. Chem. 9, 1673–1684.

[158] Ryan, W.J., Banner, W.K., Wiley, J.L., Martin, B.R. and Razdan, R.J. (1997) J. Med.

Chem. 40, 3617–3625.[159] Seltzman, H.H., Fleming, D.N., Thomas, B.F., Gilliam, A.F., McCallion, D.S., Pertwee, R.G.,

Compton, D.R. and Martin, B.R. (1997) J. Med. Chem. 40, 3626–3634.[160] Tong, W., Collantes, E.R. and Welsh, W.J. (1998) J. Med. Chem. 41, 4207–4215.[161] Thomas, B.F., Adams, I.B., Mascarella, S.W., Martin, B.R. and Razdan, R.K. (1996)

J. Med. Chem. 39, 471–479.[162] Chen, J.-Z., Han, X.-W. and Xie, X.-Q. (2005) Life Sci 76, 2053–2069 and references

cited therein.[163] Khanolkar, A.D., Abadji, V., Lin, S., Hill, W.A.G., Taha, G., Abouzid, K., Meng, Z.,

Fan, P. and Makriyannis, A. (1996) J. Med. Chem. 39, 4515–4519.[164] Lin, S., Khanolkar, A.D., Fan, P., Goutopoulos, A., Qin, C., Papahadjis, D. and

Makriyannis, A. (1998) J. Med. Chem. 41, 5353–5361.[165] Ng, E.W., Aung, M.M., Abood, M.E., Martin, B.R. and Razdan, R.K. (1999) J. Med.

Chem. 42, 1975–1981.[166] Fangour, S.E., Balas, L., Rossi, J.-C., Fedenyuk, A., Gretskaya, N., Bobrov, M.,

Bezulgov, V., Hillard, C.J. and Durrand, T. (2003) Bioorg. Med. Chem. Lett. 13,1977–1980.

Page 114: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH320

[167] Lopez-Rodrıguez, M.L., Viso, A., Ortega-Gutierrez, S., Lastres-Becker, I., Gonzalez, S.,Fernandez-Ruiz, J. and Ramos, J.A. (2001) J. Med. Chem. 44, 4505–4508.

[168] Stella, N., Schweitzer, P. and Piomelli, D. (1997) Nature (London) 388, 773–778.[169] Savinainen, J.R., Jarvinen, T., Laine, K. and Laitinen, J. (2001) Br. J. Pharmacol. 134,

664–672.[170] Goparaju, S.K., Ueda, N., Yamaguchi, H. and Yamamoto, S. (1998) FEBS Lett. 422,

69–73.[171] Saario, S.M., Savinainen, J.R., Laitinen, J.T., Jarvinen, T. and Niemi, R. (2004) Bioc-

hem. Pharmacol. 67, 1381–1387.[172] Dinh, T.P., Carpenter, D., Leslie, F.M., Freund, T.F., Katona, I., Sensi, S.L., Kathuria, S.

and Piomelli, D. (2002) Proc. Natl. Acad. Sci. USA 99, 10819–10824.[173] Sugiura, T., Kodaka, T., Nakane, S., Miyashita, T., Kondo, S., Suhara, Y., Takayama, H.,

Waku, K., Seki, C., Baba, N. and Ishima, Y. (1999) J. Biol. Chem. 274, 2794–2801.[174] Sugiura, T., Kondo, S., Kishimoto, S., Miyashita, T., Nakane, S., Kodaka, T., Suhara, Y.,

Takayama, H. and Waku, K. (2000) J. Biol. Chem. 275, 605–612.[175] Suhara, Y., Nakane, S., Arai, S., Takayama, H., Waku, K., Ishima, Y. and Sugiura, T.

(2001) Bioorg. Med. Chem. Lett. 11, 1985–1988.[176] Mechoulam, R., Fride, E., Ben-Shabat, S., Meiri, U. and Howowitz, M. (1998) Eur.

J. Pharmacol. 362, R1–R3.[177] Appendino, G., Ligresti, A., Minassi, A., Daddario, N., Bisogno, T. and Di Marzo, V.

(2003) Bioorg. Med. Chem. Lett. 13, 43–46.[178] D’Ambra, T.E., Estep, K.G., Bell, M.R., Eissenstat, M.A., Josef, K.A., Ward, S.J.,

Haydock, D.A., Baizman, E.G., Casiano, F.M., Beglin, N.C., Chippari, S.M., Grego, J.D.,Kullnig, R.K. and Daley, G.T. (1992) J. Med. Chem. 35, 124–135.

[179] Eissenstat, M.A., Bell, M.R., D’Ambra, T.E., Alexander, E.J., Daum, S.J., Ackerman, J.H.,Gruett, M.D., Kumar, V., Estep, K.G., Olefirowicz, E.M., Wetzel, J.R., Alexander, M.D.,Weaver III, J.D., Haycock, D.A., Luttinger, D.A., Casiano, F.M., Chippari, S.M.,Kuster, J.E., Stevenson, J.I. and Ward, S.J. (1995) J. Med. Chem. 38, 3094–3105.

[180] D’Ambra, T.E., Eissenstat, M.A., Abt, J., Ackerman, J.H., Bacon, E.R., Bell, M.R.,Carabateas, P.M., Josef, K.A., Kumar, V., Weaver III, J.D., Arnold, R., Casiano, F.M.,Chippari, S.M., Haycock, D.A., Kuster, J.E., Luttinger, D.A., Stevenson, J.I., Ward, S.J.,Hill, A., Khanolkar, A. and Makriyannis, A. (1996) Bioorg. Med. Chem. Lett. 6,17–22.

[181] Wiley, J.L., Compton, D.R., Dai, D., Lainton, J.A.H., Phillips, M., Huffman, J.W. andMartin, B.R. (1998) J. Pharmacol. Exp. Ther. 285, 995–1004.

[182] Huffman, J.W., Lu, J., Dai, D., Kitaygorodskiy, A., Wiley, J.L. and Martin, B.R. (2000)Bioorg. Med. Chem. 8, 439–447.

[183] Xie, X.-Q., Eissenstat, M. and Makriyannis, A. (1995) Life Sci. 56, 1963–1970.[184] Dutta, A.K., Ryan, W., Thomas, B.F., Singer, M., Compton, D.R., Martin, B.R. and

Razdan, R.K. (1997) Bioorg. Med. Chem. 5, 1591–1600.[185] Song, Z.H. and Bonner, T.I. (1996) Mol. Pharmacol. 49, 891–896.[186] Huffman, J.W., Mabon, R., Wu, M.-J., Lu, J., Hart, R., Hurst, D.P., Reggio, P.H.,

Wiley, J.L. and Martin, B.R. (2003) Bioorg. Med. Chem. 11, 539–549.[187] Makriyannis, A. and Deng, H. (2001) PCT Int. Appl., WO 01 28557; (2001) Chem. Abstr.

134, 311344.[188] Leftheris, K., Zhao, R., Bang-Chi, C., Kiener, P., Hong, W., Pandit, C.R., Wrobleski, S.,

Chen, P., Hynes, J., Jr., Longphre, M., Norris, D.J., Spergel, S. and Tokarski, J. (2001)PCT Int. Appl., WO 01 58869; (2001) Chem. Abstr. 135, 166827.

[189] Moloney, P.G. and Robertson, A.D., (2002) PCT Int. Appl., WO 02 36590; (2002)Chem. Abstr. 136, 369720.

Page 115: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 321

[190] Cowley, P.M., Caulfield, W., Tierney, J., Cairns, J., Adam-Worrall, J. and York, M.(2004) PCT Int. Appl., WO 04 00832; (2004) Chem. Abstr. 140, 59667.

[191] Page, D., Walpole, C. and Yang, H. (2004) PCT Int. Appl., WO 04 108689; (2005) Chem.

Abstr. 142, 56311.[192] Liu, Z., Page, D., Walpole, C. and Yang, H. (2004) PCT Int., Appl. WO 04 108688;

(2005) Chem. Abstr. 142, 56310.[193] Liu, Z., Page, D., Walpole, C. and Yang, H. (2004) PCT Int. Appl., WO 04 108712;

(2005) Chem. Abstr. 142, 56312.[194] Wei, Z., Dolaine, R., Walpole, C. and Yang, H. (2004) PCT Int. Appl., WO 04 087704;

(2004) Chem. Abstr. 141, 332183.[195] Tarzia, G., Duranti, A., Tontini, A., Spadoni, G., Mor, M., Rivara, S., Plazzi, P.V.,

Kathuria, S. and Piomelli, D. (2003) Bioorg. Med. Chem. 11, 3965–3973.[196] Wiley, J.L., Jefferson, R.G., Grier, M.C., Mahadevan, A., Razdan, R.K. andMartin, B.R.

(2001) J. Pharmacol. Exp. Ther. 296, 1013–1022.[197] Kumar, V., Alexander, M.D., Bell, M.R., Eissenstat, M.A., Casiano, F.M., Chippari, S.M.,

Haycock, D.A., Luttinger, D.A., Kuster, J.E., Miller, M.S., Stevenson, J.I. and Ward, S.J.(1995) Bioorg. Med. Chem. Lett. 5, 381–386.

[198] Reggio, P.H., Basu-Dutt, S., Barnett-Norris, J., Castro, M.T., Hurst, D.P., Seltzman, H.H.,Roche, M.J., Gilliam, A.F., Thomas, B.F., Stevenson, L.A., Pertwee, R.G. andAbood, M.E. (1998) J. Med. Chem. 41, 5177–5187.

[199] Heil, M., Meier, H., Naab, P., Voerste, A., De Vry, J.-M.-V., Denzer, D., Mauler, F.,Lustig, K., Lenfers, J.-B., (2002) PCT Int. Appl., WO 02 26702; (2002) Chem. Abstr. 136,279205.

[200] Heil, M., Meier, H., Naab, P., Voerste, A., De Vry, J.-M.-K., Denzer, D., Mauler, F.,Lustig, K., Hinz, V., Allwerhiligen, S., (2001) PCT Int. Appl., WO 01 74763; (2001)Chem. Abstr. 135, 288578.

[201] Mittendorf, J., Dressel, J., Matzke, M., Keldenich, J., Mauler, F., De Vry, J.-M.-K.,Franz, J., Spreyer, P., V +ohringer, V., Schuhmacher, J. S., Rock, M.-H., Horvath, E.,Friedel , A., (2000) PCT Int. Appl., WO 00 10968; (2000) Chem. Abstr. 132, 166512.

[202] Mittendorf, J., Dressel, J., Matzke, M., Keldenich, J., Mauler, F., De Vry, J.-M.-V.,Franz, J., Spreyer, P., V +ohringer, V., Schuhmacher, J. S., Rock, M.-H., Horvath, E.,Friedel, A., Mohrs, K.-H., Raddatz, S., Jork, R., (2000) PCT Int. Appl., WO 00 10967;(2000) Chem. Abstr. 132, 166012.

[203] Mittendorf, J., Dressel, J., Matzke, M., Keldenich, J., Mohrs, K.-H., Raddatz, S., Franz, J.,Spreyer, P., V+ohringer, V., Schuhmacher, J., Rock, M.-H., Horvath, E., Friedel, A.,Mauler, F., De Vry, J.-M.-V., Jork, R., (1998) PCT Int. Appl., WO 98 37061; (1998) Chem.Abstr. 129, 202764.

[204] Mauler, F., Horvath, E., De Vry, J., Jager, R., Schwarz, T., Sandmann, S., Corinna, W.,Heinig, R. and Bottcher, M. (2003) CNS Drug Rev. 9, 343–358.

[205] Mauler, F., Mittendorf, J., Horvath, E. and De Vry, J. (2002) J. Pharmacol. Exp. Ther.

302, 359–368.[206] De Vry, J. and Jentzsch, K.R. (2004) Eur. J. Pharmacol. 505, 127–133.[207] De Vry, J., Denzer, D., Reissmueller, E., Eijckenboom, M., Heil, M., Meier, H. and

Mauler, F. (2004) J. Pharmacol. Exp. Ther. 310, 620–632.[208] Brain, C.T., Culshaw, A.J., Dziadulewicz, E.K. and Schopfer, U. (2002) PCT Int. Appl.,

WO 02 42248; (2002) Chem. Abstr. 137, 6003.[209] Brain, C.T., Dziadulewicz, E.K. and Hart, T.W. (2003) PCT Int. Appl., WO 03 66603;

(2003) Chem. Abstr. 139, 180079.[210] Leung, C., Tomaszewski, M. and Woo, S. (2004) PCT Int. Appl., WO 04 60882; (2004)

Chem. Abstr. 141, 140172.

Page 116: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH322

[211] Arimura, A. (2005) PCT Int. Appl., WO 05 016351; (2005) Chem. Abstr. 142, 212367.[212] Gareau, Y., Dufresne, C., Gallant, M., Rochette, C., Sawyer, N., Slipetz, D.M.,

Tremblay, N., Weech, P.K., Metters, K.M. and Labelle, M. (1996) Bioorg. Med. Chem.

Lett. 6, 189–194.[213] Huffman, J.W., Yu, S., Showalter, V., Abood, M.E., Wiley, J.L., Compton, D.R.,

Martin, B.R., Bramblett, R.D. and Reggio, P.H. (1996) J. Med. Chem. 39,3875–3877.

[214] Huffman, J.W., Liddle, J., Yu, S., Aung, M.M., Abood, M.E., Wiley, J.L. andMartin, B.R.(1999) Bioorg. Med. Chem. 7, 2905–2914.

[215] Huffman, J.W., Bushell, S.M., Miller, J.R.A., Wiley, J.L. and Martin, B.R. (2002)Bioorg. Med. Chem. 10, 4119–4129.

[216] Reggio, P.H., Wang, T., Brown, A.E., Fleming, D.N., Seltzman, H.H., Griffin, G.,Pertwee, R.G., Compton, D.R., Abood, M.E. and Martin, B.R. (1997) J. Med. Chem.

40, 3312–3318.[217] Lau, C.K. (1996) Can. Pat. Appl., CA 2170850; (1997) Chem. Abstr. 126, 31275.[218] Gallant, M., Dufresne, C., Gareau, Y., Guay, D., Leblanc, Y., Prasit, P., Rochette, C.,

Sawyer, N., Slipetz, D.M., Tremblay, N., Metters, K.M. and Labelle, M. (1996) Bioorg.Med. Chem. Lett. 6, 2263–2268.

[219] Huffman, J.W., Zengin, G., Wu, M.-J., Lu, J., Hynd, G., Bushell, K., Thompson,A.L.S., Bushell, S., Tartal, C., Hurst, D.P., Reggio, P.H., Selley, D.E., Cassidy, M.P.,Wiley, J.L. and Martin, B.R. (2005) Bioorg. Med. Chem. 13, 89–112.

[220] Hynes, J., Jr., Leftheris, K., Wu, H., Pandit, C., Chen, P., Norris, D.J., Chen, B.-C.,Zhao, R., Kiener, P.A., Chen, X., Turk, L.A., Patil-Koota, V., Gillooly, K.M.,Shuster, D.J. and McIntyre, K.W. (2002) Bioorg. Med. Chem. Lett. 12, 2399–2402.

[221] Wrobleski, S.T., Chen, P., Hynes, J., Jr., Lin, S., Norris, D.J., Pandit, C.R., Spergel, S.,Wu, H., Tokarski, J.S., Chen, X., Gillooly, K.M., Kiener, P.A., McIntyre, K.W.,Patil-koota, V., Shuster, D.J., Turk, L.A., Yang, G. and Leftheris, K. (2003) J. Med.

Chem. 46, 2110–2116.[222] Makriyannis, A. and Liu, Q. (2003) PCT Int. Appl., WO 03 035005; (2003) Chem. Abstr.

138, 331733.[223] Barth, F., Rinaldi-Carmona, M. and Vernhet, C. (2004) PCT Int. Appl., WO 2004

050086; (2004) Chem. Abstr. 141, 23418.[224] Ibrahim, M.M., Deng, H., Zvonok, A., Cockayne, D.A., Kwan, J., Mata, H.P.,

Vanderah, T.W., Lai, J., Porreca, F., Makriyannis, A. and Malan, T.P., Jr. (2003) Proc.Natl. Acad. Sci. USA 100, 10529–10533.

[225] Kozlowski, J.A., Shankar, B.B., Shih, N.-Y. and Tong, L. (2004) PCT Int. Appl., WO2004 000807; (2004) Chem. Abstr. 140, 77029.

[226] Hanus, L., Breuer, A., Tchilibon, S., Shiloah, S., Goldenberg, D., Horowitz, M.,Pertwee, R.G., Ross, R.A., Mechoulam, R. and Fride, E. (1999) Proc. Natl. Acad. Sci.

USA 96, 14228–14233.[227] Wiley, J.L., Beletskaya, I.D., Ng, E.W., Dai, Z., Crocker, P.J., Mahadevan, A.,

Razdan, R.K. and Martin, B.R. (2002) J. Pharmacol. Exp. Ther. 301, 679–689.[228] Makriyannis, A. and Khanolkar, A. (2001) PCT Int. Appl., WO 01 28497; (2001) Chem.

Abstr. 134, 320880.[229] Makriyannis, A. and Khanolkar, A. (2001) PCT Int. Appl., WO 01 2839; (2001) Chem.

Abstr. 134, 305318.[230] Mukherjee, S., Adams, M., Whiteaker, K., Daza, A., Kage, K., Cassar, S., Meyer, M.

and Yao, B.B. (2004) Eur. J. Pharmacol. 505, 1–9.[231] Page, D., Walpole, C. and Yang, H. (2004) PCT Int. Appl., WO 04 035548; (2004)

Chem. Abstr. 140, 375170.

Page 117: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 323

[232] Tada, Y., Iso, Y. and Hanasaki, K. (2002) PCT Int. Appl., WO 02 053543; (2002) Chem.Abstr. 137, 93689.

[233] Ogawa, M., Takaoka, Y. and Ohhata, A. (2002) PCT Int. Appl., WO 02 10135; (2002)Chem. Abstr. 136, 151084.

[234] Makriyannis, A. and Deng, H. (2002) PCT Int. Appl., WO 02 58636; (2002) Chem.Abstr. 137, 140538.

[235] Hanasaki, K., Murashi, T. and Kai, H. (2001) PCT Int. Appl., WO 01 19807; (2001)Chem. Abstr. 134, 237483.

[236] Huffman, J.W., Lu, J., Hynd, G., Wiley, J.L. and Martin, B.R. (2001) Bioorg. Med.

Chem. 9, 2863–2870.[237] Green, R.H., Eatherton, A.J., Giblin, G.M.P., Jandu, K.S., Mitchell, W.L., Naylor, A.,

Palombi, G., Rawlings, D.A., Slingsby, B.P. and Whittington, A.R. (2004) PCT Int.Appl., WO 2004029027; (2004) Chem. Abstr. 140, 303544.

[238] Grotenhermen, F. and Muller-Vahl, K. (2003) Exp. Opin. Pharmacother. 4, 2367–2371.[239] Zajicek, J., Fox, P., Sanders, H., Wright, D., Vickery, J., Nunn, A. and Thompson, A.

(2003) Lancet 362, 1517–1526.[240] Scott, D.A., Wright, C.E. and Angus, J.A. (2004) Pain 109, 124–131.[241] Johanek, L.M. and Simone, D.A. (2004) Pain 109, 432–442.[242] Clayton, N., Marshall, F.H., Bountra, C. and O’Shaughnessy, C.T. (2002) Pain 96,

253–260.[243] Noyes, R., Jr., Brunk, S.F., Baram, D.A. and Canter, A. (1975) J. Clin. Pharmacol. 15,

139–143.[244] Attal, N., Brasseur, L., Guirimand, D., Clermond-Gnamien, S., Atlami, S. and

Bouhassira, D. (2004) Eur. J. Pain 8, 173–177.[245] Holdcroft, A., Smith, M., Jacklin, A., Hodgson, H., Smith, B., Newton, M. and

Evans, F. (1997) Anaesthesia 52, 483–486.[246] Malan, T.P., Ibrahim, M.M., Lai, J., Vanderah, T.W., Makriyannis, A. and Porreca, F.

(2003) Curr. Opin. Pharmacol. 3, 62–67.[247] Ibrahim, M.M., Porreca, F., Lai, J., Albrecht, P.J., Rice, F.L., Khodorova, A.,

Davar, G., Makriyannis, A., Vanderah, T.W., Mata, H.P. and Malan, T.P. (2005) Proc.Natl. Acad. Sci. USA 102, 3093–3098.

[248] Rukwied, R., Watkinson, A., McGlone, F. and Dvorak, M. (2003) Pain 102, 283–288.[249] Drysdale, A.J. and Platt, B. (2003) Curr. Med. Chem. 10, 2719–2732.[250] Tomida, I., Pertwee, R.G. and Azuara-Blanco, A. (2004) Br. J. Ophthalmol. 88, 708–713.[251] de Vries, A., van Rijnsoever, C., Engels, F., Henricks, P.A. and Nijkamp, F.P. (2001)

Br. J. Pharmacol. 134, 771–776.[252] Yasui, K., Morioka, Y. and Hanasaki, K. (2003) PCT Int. Appl., WO 03 70277; (2003)

Chem. Abstr. 139, 207822.[253] Brady, C.M., DasGupta, R., Dalton, C., Wiseman, O.J., Berkley, K.J. and Fowler, C.J.

(2004) Mult. Scler. 10, 425–433.[254] Di Filippo, C., Rossi, F., Rossi, S. and D’Amico, M. (2004) J. Leukoc. Biol. 75, 453–459.[255] Steffens, S., Veillard, N.R., Arnaud, C., Pelli, G., Burger, F., Staub, C., Zimmer, A.,

Frossard, J.L. and Mach, F. (2005) Nature (London) 434, 782–786.[256] Sanchez, C., de Ceballos, M.L., del Pulgar, T.G., Rueda, D., Corbacho, C., Velasco, G.,

Galve-Roperh, I., Huffman, J.W., Ramon y Cajal, S. and Guzman, M. (2001) CancerRes 61, 5784–5789.

[257] McKallip, R.J., Lombard, C., Fisher, M., Martin, B.R., Ryu, S., Grant, S., Nagarkatti,P.S. and Nagarkatti, M. (2002) Blood 100, 627–634.

[258] Bab, I., Mechoulam, R. and Shohami, E. (2004) PCT Int. Appl., WO 2004 103410;(2005) Chem. Abstr. 142, 28165.

Page 118: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH324

[259] Patel, H.J., Birrell, M.A., Crispino, N., Hele, D.J., Venkatesan, P., Barnes, P.J., Yacoub,M.H. and Belvisi, M.G. (2003) Br. J. Pharmacol. 140, 261–268.

[260] Lange, J.H.M. and Kruse, C.G. (2004) Curr. Opin. Drug Discovery Dev. 7, 498–506.[261] Smith, R.A. and Fathi, Z. (2005) Idrugs 8, 53–66.[262] Lange, J.H.M. and Kruse, C.G. (2005) Drug Disc. Today 10, 693–702.[263] Barth, F., Casellas, P., Congy, C., Martinez, S. and Carmona, M. (1993) Euro. Appl.,

EP-00576357; (1994) Chem. Abstr. 121, 280631.[264] Barth, F., Casellas, P., Congy, C., Martinez, S., Rinaldi, M. and Anne-Archard, G.

(1995) Euro. Appl., EP-00656354; (1995) Chem. Abstr. 123, 286006.[265] Meschler, J.P., Kraichely, D.M., Wilken, G.H. and Howlett, A.C. (2000) Biochem.

Pharmacol. 60, 1315–1323.[266] Barth, F., Congy, C., Martinez, S. and Rinaldi-Carmona, M. (2005) PCT Int. Appl., WO

05 00820; (2005) Chem. Abstr. 142, 93815[267] Lan, R., Liu, Q., Fan, P., Lin, S., Fernando, S.R., McCallion, D., Pertwee, R. and

Makriyannis, A. (1999) J. Med. Chem. 42, 769–776.[268] Gatley, S.J., Lan, R., Volkow, N.D., Pappas, N., King, P., Wong, C.T., Gifford, A.N.,

Pyatt, B., Dewey, S.L. and Makriyannis, A. (1998) J. Neurochem. 70, 417–423.[269] Camus, P., Martinez, S. and Rinaldi, M. (2000) PCT Int. Appl., WO 00 46209; (2000)

Chem. Abstr. 133, 164051.[270] Rinaldi-Carmona, M., Barth, F., Congy, C., Martinez, S., Oustric, D., Perio, A.,

Poncelet, M., Maruani, J., Arnone, M., Finance, O., Soubrie, P. and Le Fur, G. (2004)J. Pharmacol. Exp. Ther. 310, 905–914.

[271] Miscoria, G., Rinaldi, M. and Schofield, J. (2004) PCT Int. Appl., WO 04 58744; (2004)Chem. Abstr. 141, 71539.

[272] Makriyannis, A. and Liu, Q. (2003) PCT Int. Appl., WO 03 20217; (2003) Chem. Abstr.138, 238177.

[273] Katoch-Rouse, R., Pavlova, O.A., Caulder, T., Hoffman, A.F., Mukhin, A.G. andHorti, A.G. (2003) J. Med. Chem. 46, 642–645.

[274] Barth, F., Congy, G., Martinez, S. and Rinaldi, M. (2001) PCT Int. Appl., WO 01 32663;(2001) Chem. Abstr. 134, 340504

[275] Stoit, A.R., Lange, J.H.M., den Hartog, A.P., Ronken, E., Tipker, K., vanStuivenberg, H.H., Dijksman, J.A.R., Wals, H.C. and Kruse, C.G. (2002) Chem.

Pharm. Bull. 50, 1109–1113.[276] Ruiu, S., Pinna, G.A., Marchese, G., Mussinu, J.-M., Saba, P., Tambaro, S., Casti, P.,

Vargiu, R. and Pani, L. (2003) J. Pharmacol. Exp. Ther. 306, 363–370.[277] Francisco, M.E.Y., Burgess, J.P., George, C., Baley, G.S., Gilliam, A.F., Seltzmann, H.H.

and Thomas, B.F. (2003) Magn. Reson. Chem. 41, 265–268.[278] Francisco, M.E.Y., Seltzman, H.H., Gilliam, A.F., Mitchell, R.A., Rider, S.L.,

Pertwee, R.G., Stevenson, L.A. and Thomas, B.F. (2002) J. Med. Chem. 45, 2708–2719.[279] Shim, J.-Y., Welsh, W.J., Cartier, E., Edwards, J.L. and Howlett, A.C. (2002) J. Med.

Chem. 45, 1447–1459.[280] Shim, J.-Y., Collantes, E.R., Welsh, W.J. and Howlett, A.C. (1999) In ‘‘Rational Drug

Design Symposium Series’’. Parrill, A.L. and Reddy, M.R. (eds), pp. 165–184. AmericanChemical Society, Washington DC.

[281] Hurst, D.P., Lynch, D.L., Barnett-Norris, J., Hyatt, S.M., Seltzman, H.H., Zhong, M.,Song, Z.-H., Nie, J., Lewis, D. and Reggio, P.H. (2002) Mol. Pharmacol. 62, 1274–1287.

[282] Salo, O.M.H., Lahtela-Kakkonen, M., Gynther, J., Jarvinen, T. and Poso, A. (2004)J. Med. Chem. 47, 3048–3057.

[283] McAllister, S.D., Rizvi, G., Anavi-Goffer, S., Hurst, D.P., Barnett-Norris, J.,Lynch, D.L., Reggio, P.H. and Abood, M.E. (2003) J. Med. Chem. 46, 5139–5152.

Page 119: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 325

[284] Dow, R.L. and Hammond, M. (2004) PCT Intl. Appl., WO 04 52864; (2004) Chem.Abstr. 141, 54345.

[285] Dow, R.L. (2004) PCT Intl. Appl., WO 04 35566; (2004) Chem. Abstr. 140, 357343.[286] Carpino, P.A. and Griffith, D.A. (2004) US Appl., US 20040214838; (2004) Chem. Abstr.

141, 379939.[287] Griffith, D.A. and Hammond, M. (2004) US Appl., US 20040214837; (2004) Chem.

Abstr. 141, 379938.[288] Carpino, P.A. and Dow, R. L. (2004) US Appl., US 20040214855; (2004) Chem. Abstr.

141, 379924.[289] Carpino, P.A. and Dow, R. L. (2004) US Appl., US 20040214856; (2004) Chem. Abstr.

141, 395553.[290] Griffith, D.A. (2004) US Appl., US 20040157838; (2004) Chem. Abstr. 141, 190798.[291] Griffith, D.A. (2004) US Appl., US 20040157839; (2004) Chem. Abstr. 141, 190806.[292] Carpino, P.A. and Sakya, S.M. (2004) US Appl., US 2004024881; (2005) Chem. Abstr.

142, 38266.[293] Lange, J.H.M., Kruse, C.G., Tipker, J., Tulp, M.T.M. and van Vliet, B.J. (2001) PCT

Intl. Appl., WO 01 70700; (2001) Chem. Abstr. 135, 257239.[294] Lange, J.H.M., Kruse, C.G., Tipker, J. and Hoogendorn, J. (2002) PCT Intl. Appl., WO

02 76949; (2002) Chem. Abstr. 137, 279183.[295] Kruse, C.G., Lange, J.H.M., Tipker, J., Herremans, A.H.J. and van Stuivenberg, H.H.

(2003) PCT Intl. Appl., WO 03 26647; (2003) Chem. Abstr. 138, 287664.[296] Kruse, C.G., Lange, J.H.M., Tipker, J., Herremans, A.H.J. and van Stuivenberg, H.H.

(2003) PCT Intl. Appl., WO 03 26648; (2003) Chem. Abstr. 138, 287665.[297] Lange, J.H.M., Coolen, H.K.A.C., van Stuivenberg, H.H., Dijksman, J.A.R.,

Herremans, A.H.J., Ronken, E., Keizer, H.G., Tipker, K., McCreary, A.C., Veerman, W.,Wals, H.C., Stork, B., Verveer, P.C., den Hartog, A.P., de Jong, N.M.J., Adolfs, T.J.P.,Hoogendorn, J. and Kruse, C.G. (2004) J. Med. Chem. 47, 627–643.

[298] Finke, P.E., Mills, S.G., Plummer, C.W., Shah, S.K. and Truong, Q.T. (2003) PCT Intl.

Appl., WO 03 07887; (2003) Chem. Abstr. 138, 122647.[299] Kruse, C.G., Lange, J.H.M., Herremans, A.H.J. and Van Stuivenberg, H.H. (2003) PCT

Intl. Appl., WO 03 27076; (2003) Chem. Abstr. 138, 287674.[300] Hagmann, W.K., Qi, H. and Shah, S.K. (2003) PCT Intl. Appl., WO 03 63781; (2003)

Chem. Abstr. 139, 144003.[301] Carpino, P.A. (2005) PCT Intl. Appl., WO 05 09974; (2005) Chem. Abstr. 142, 198075.[302] Griffith, D.A. (2004) PCT Intl. Appl., WO 04 37823; (2004) Chem. Abstr. 140,

391293.[303] Coe, J.W., Iredale, P.A., McHardy, S.F. and McLean, S. (2005) US Appl., US-

20050043327; (2005) Chem. Abstr. 142, 233372.[304] Coe, J.W., Iredale, P.A. and Sands, S.B. (2004) US Appl., US-20040224962; (2004)

Chem. Abstr. 141, 400942.[305] Coe, J.W., Iredale, P.A. and Sands, S.B. (2004) US Appl., US-20040224963; (2004)

Chem. Abstr. 141, 400943.[306] Berggren, A.I.K., Bostrom, S.J., Elebring, S.T., Fallefors, L., Wilstermann, J.M. and

Greasley, P. (2004) PCT Intl. Appl., WO 04 58255; (2004) Chem. Abstr. 141,106460.

[307] Lindfors, L. (2004) PCT Intl. Appl., WO 04 69226; (2004) Chem. Abstr. 141, 195287.[308] Lange, J.H.M., Kruse, C.G., van Stuivenberg, H.H. and Sliedregt, L.A.J.M. (2005) US

Appl., US-20050065189; (2005) Chem. Abstr. 142, 336350.[309] Lange, J.H.M., Kruse, C.G., McCreary, A.C. and van Stuivenberg, H.H. (2004) PCT

Intl. Appl., WO 04 26301; (2004) Chem. Abstr. 140, 303680.

Page 120: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH326

[310] Jagerovic, N., Hernandez-Folgado, L., Alkorta, I., Goya, P., Navarro, M., Serrano, A.,Rodriguez de Fonseca, F., Dannert, M.T., Alsasua, A., Suardiaz, M., Pascual, D. andMartın, M.I. (2004) J. Med. Chem. 47, 2939–2942.

[311] Lange, J.H., van Stuivenberg, H.H., Coolen, H.K., Adolfs, T.J., McCreary, A.C.,Keizer, H.G., Wals, H.C., Veerman, W., Borst, A.J., de Looff, W., Verveer, P.C. andKruse, C.G. (2005) J. Med. Chem. 48, 1823–1838.

[312] Plummer, C.W., Finke, P.E., Mills, S.G., Wang, J., Tong, X., Doss, G.A., Fong, T.M.,Laoc, J.Z., Schaeffer, M.-T., Chen, J., Shen, C.-P., Stribling, D.S., Shearman, L.P.,Strack, A.M. and van der Ploe, L.H.T. (2005) Bioorg. Med. Chem. Lett. 15, 1441–1446.

[313] Berggren, A.I.K., Bostrom, S.J., Cheng, L., Elebring, S.T., Greasley, P., Nagard, M.,Wilstermann, J.M. and Terricabras, E. (2004) PCT Intl., Appl., WO 04 58249; (2004)Chem. Abstr. 141, 123553.

[314] Finke, P.E., Meurer, L.C., Debenham, J.S., Toupence, R.B. and Walsh, T.F. (2003)PCT Intl. Appl., WO 03 82191; (2003) Chem. Abstr. 139, 307686.

[315] Kopka, I.E., Li, B. and Hagmann, W.K. (2004) PCT Intl. Appl., WO 04 29204; (2004)Chem. Abstr. 140, 303694.

[316] Barth, F., Martinez, S. and Rinaldi-Carmona, M. (2003) PCT Intl. Appl., WO 03 84930;(2003) Chem. Abstr. 139, 307689.

[317] Barth, F., Martinez, S. and Rinaldi-Carmona, M. (2003) PCT Intl. Appl., WO 03 84943;(2003) Chem. Abstr. 139, 323435.

[318] Dow, R.L. (2004) PCT Intl. Appl., WO 04 110453; (2005) Chem. Abstr. 142, 56347.[319] Meurer, L.C., Finke, P.E., Mills, S.G., Walsh, T.F., Toupence, R.B., Goulet, M.T.,

Wang, J., Tong, X., Fong, T.M., Lao, J., Schaeffer, M.-T., Chen, J., Shen, C.-P.,Stribling, D.S., Shearman, L.P., Strack, A.M. and van der Ploeg, L.H.T. (2005) Bioorg.Med. Chem. Lett. 15, 645–651.

[320] Wilsterman, J.M. and Berggren, A.I.K. (2003) PCT Intl. Appl., WO 03 51850; (2003)Chem. Abstr. 139, 69284.

[321] Berggren, A.I.K., Bostrom, S.J., Elebring, S.T., Greasley, P., Terricabra, E. andWilstermann, J.M. (2003). PCT Intl. Appl., WO 03 51851; (2003) Chem. Abstr. 139,69285.

[322] Cheng, L., Berggren, K., Elebring, T. and Sorensen, H. (2004) PCT Intl. Appl., WO 04111033; (2004) Chem. Abstr. 142, 56361.

[323] Cheng, L. (2004) PCT Intl. Appl., WO 04 111034; (2005) Chem. Abstr. 142, 56362.[324] Cheng, L. (2004) PCT Intl. Appl., WO 04 111038; (2005) Chem. Abstr. 142, 56363.[325] Lindfors, L. (2004) PCT Intl. Appl., WO 04 69277; (2004) Chem. Abstr. 141, 212754.[326] Ellsworth, B.A., Sun, C. and Pendri, A. (2005) PCT Intl. Appl., WO 05 16286; (2005)

Chem. Abstr. 142, 261555.[327] Cheng, L. and Wilstermann, M. (2004) PCT Intl. Appl., WO 04 111039; (2005) Chem.

Abstr. 142, 56364.[328] Toupence, R.B., Debenham, J.S., Goulet, M.T., Madsen-Duggan, C.B., Walsh, T.F.

and Shah, S.K. (2004) PCT Intl. Appl., WO 04 12671; (2004) Chem. Abstr. 140, 163855.[329] Alanine, A., Beleicher, K.G.W., Haap, W., Kuber, D., Luebbers, T., Plancher, J.-M.,

Rogers-Evans, M., Schneider, G., Zuegge, J. and Roche, O. (2004) PCT Intl. Appl., WO04 13120; (2004) Chem. Abstr. 140, 163856.

[330] Peracchia, M.-T., Cote, S. and Bobineau, V. (2005) Euro. Appl., EP-01498122; (2005)Chem. Abstr. 142, 225814.

[331] Peracchia, M.-T., Cote S. and Bobineau, V. (2005) Euro. Appl., EP-01498123; (2005)Chem. Abstr. 142, 225814.

[332] Achard, D., Bouchard, H., Bouquerel, J., Filoche, B., Grisoni, S., Hittinger, A. andMyers, M. (2001) PCT Intl. Appl., WO 01 64634; (2001) Chem. Abstr. 135, 226875.

Page 121: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 327

[333] Achard, D., Bouchard, H., Bouquerel, J., Filoche, B., Grisoni, S., Hittinger, A. andMyers, M. (2001) PCT Intl. Appl., WO 01 64633; (2001) Chem. Abstr. 135, 226874.

[334] Achard, D., Bouchard, H., Bouquerel, J., Filoche, B., Grisoni, S., Hittinger, A. andMyers, M. (2001) PCT Intl. Appl., WO 01 64632; (2001) Chem. Abstr. 135, 226873.

[335] Davidson, J.E.P., Adams, D.R., Bickerdike, M.J. and Fletcher, A. (2004) PCT Intl.

Appl., WO 04 96209; (2004) Chem. Abstr. 141, 410804.[336] Davidson, J.E.P., Bentley, J.M., Dawson, C.E., Harrison, K., Mansell, H.L.,

Pither, A.L., Pratt, R.M., Roffey, J.M.A. and Ruston, V.J. (2004) PCT Intl. Appl.,WO 04 96794; (2004) Chem. Abstr. 141, 410806.

[337] Mayweg, A., Marty, H.P., Mueller, W., Narquizian, R., Neidhart, W., Pflieger, P. andRoever, S. (2004) PCT Intl. Appl., WO 04 60870; (2004) Chem. Abstr. 141, 140442.

[338] Guba, W., Haap, W., Marty, H.P. and Narquizian, R. (2004) PCT Intl., Appl., WO 0460888; (2004) Chem. Abstr. 141, 140427.

[339] Hagmann, W.K., Lin, L.S., Shah, S.K., Goulet, M.T. and Jewell, J.P. (2003) PCT Intl.

Appl., WO 03 82190; (2003) Chem. Abstr. 139, 307605.[340] Hagmann, W.K., Lin, L.S. and Shah, S.K. (2003) PCT Intl. Appl., WO 03 87037; (2003)

Chem. Abstr. 139, 337785.[341] Castonguay, L.A., Hagmann, W.K., Lin, L.S. and Shah, S.K. (2003) PCT Intl. Appl.,

WO 03 86288; (2003) Chem. Abstr. 139, 337784.[342] Hagmann, W.K., Lin, L.S., Shah, S.K., Guthikonda, R.N., Qi, H., Chang, L.L., Liu, P.,

Armstrong, H.M., Jewell, P.J. and Lanza, T.J. (2004) PCT Intl. Appl., WO 04 48317;(2004) Chem. Abstr. 141, 38363.

[343] Lin, L.S., Hagmann, W.K., Kumar, S., Yin, W. and Doss, G. (2004) PCT Intl. Appl.,

WO 04 58145; (2004) Chem. Abstr. 141, 106274.[344] Kanyonyo, M., Govaerts, S.J., Hermans, E., Poupaert, J.H. and Lambert, D.M. (1999)

Bioorg. Med. Chem. Lett. 9, 2233–2236.[345] Ooms, F., Wouters, J., Oscari, O., Happaerts, T., Bouchard, G., Carrupt, P.A., Testa, B.

and Lambert, D. (2002) J. Med. Chem. 45, 1748–1756.[346] Gaillard, P., Carrupt, P.A., Testa, B. and Boudon, A.J. (1994) Comput.-Aided Mol. Des.

8, 83–96.[347] DAYLIGHT Software 4.41 (1995) ‘‘Daylight Chemical Information System’’. Irvine, CA.[348] Pertwee, R.G. (1999) Curr. Med. Chem. 6, 635–664.[349] Muccioli, G.G., Martin, D., Scriba, G.K.E., Poppitz, W., Poupaert, J.H., Wouters, J.

and Lambert, D.M. (2005) J. Med. Chem. 48, 2509–2517.[350] Thomas, A., Ross, R.A., Saha, B., Mahadevan, A., Razdan, R.K. and Pertwee, R.G.

(2004) Eur. J. Pharmacol. 487, 213–221.[351] Felder, C.C., Joyce, K.E., Briley, E.M., Glass, M., Mackie, K.P., Fahey, K.J., Cullinan, G.J.,

Hunden, D.C., Johnson, D.W., Chaney, M.O., Koppel, G.A. and Brownstein, M. (1998)J. Pharmacol. Exp. Ther. 284, 291–297.

[352] Nettekoven, M.H. and Roever, S. (2004) PCT Intl. Appl., WO 05 00301; (2005) Chem.

Abstr. 142, 74558.[353] Sher, P.M., Sun, C., Sulsky, R.B., Wu, G. and Ewing, W.R. (2005) US Appl., US

20050009870; (2005) Chem. Abstr. 142, 134479.[354] Armstrong, H.M., Chang, L.L., Guthikonda, R.N., Hagmann, W.K., Lin, L.S. and

Shah, S.K. (2005) PCT Intl. Appl., WO 05 27837; (2005) Chem. Abstr. 142, 355050.[355] Kirkham, T.C. and Williams, C.M. (2001) Nutr. Res. Rev. 14, 65–86.[356] Cota, D., Marsicano, G., Lutz, B., Vicennati, V., Stalla, G.K., Pasquali, R. and

Pagotto, U. (2003) Int. J. Obesity Relat. Metab. Disord. 27, 289–301.[357] Kirkham, T.C., Williams, C.M., Fezza, F. and Di Marzo, V. (2001) Br. J. Pharmacol.

136, 550–557.

Page 122: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

RECENT PROGRESS IN CANNABINOID RESEARCH328

[358] Jamshidi, N. and Taylor, D.A. (2001) Br. J. Pharmacol. 134, 1151–1154.[359] Di Marzo, V., Goparaju, S.K., Wang, L., Liu, J., Batkai, S., Jarai, Z., Fezza, F.,

Miura, G.I., Palmiter, R.D., Sugiura, T. and Kunos, G. (2001) Nature (London) 410,822–825.

[360] Gomez, R., Navarro, M., Ferrer, B., Trigo, J.M., Bilbao, A., del Arco, I., Cippitelli, A.,Nava, F., Piomelli, D. and de Foncesca, F.R. (2002) J. Neuroscience 22, 9612–9617.

[361] Cota, D., Marsicano, G., Tschop, M., Grubler, Y., Flachskamm, C., Schubert, M.,Auer, D., Yassouridis, A., Thone-Reineke, C., Ortmann, S., Tomassoni, F., Cervino, C.,Nisoli, E., Linthorst, A.C.E., Pasquali, R., Lutz, B., Stalla, G.K. and Pagotto, U. (2003)J. Clin. Invest. 112, 423–431.

[362] Black, S.C. (2004) Curr. Opin. Invest. Drugs 5, 389–394.[363] Vickers, S.P. and Kennett, G.A. (2005) Curr. Drug Targets 6, 215–223.[364] Colombo, G., Agabio, R., Diaz, G., Lobina, C., Reali, R. and Gessa, G.L. (1998) Life

Sci. 63, PL113–PL117.[365] Rowland, N.E., Mukherjee, M. and Robertson, K. (2001) Psychopharmacology 159,

111–116.[366] Higgs, S., Williams, C.M. and Kirkham, T.C. (2003) Psychopharmacology 165, 370–377.[367] Ravinet Trillou, C., Arnone, M., Delgorge, C., Gonalons, N., Keane, P., Maffrand, J.-P.

and Soubrie, P. (2003) Am. J. Physiol. Regul. Integr. Comp. Physiol. 284, R345–R353.[368] Poirier, B., Bidouard, J.-P., Cadrouvele, C., Marniquet, X., Staels, B., O’Connor, S.E.,

Janiak, P. and Herbert, J.M. (2005) Diabetes Obesity Metab. 7, 65–72.[369] Ravinet Trillou, C., Delgorge, C., Menet, C., Arnone, M. and Soubrie, P. (2004) Int. J.

Obesity 28, 640–648.[370] Fernandez, J.R. and Allison, D.B. (2004) Curr. Opin. Invest. Drugs 5, 430–435.[371] Van Gaal, L.F., Rissanen, A.M., Scheen, A.J., Ziegler, O. and Rossner, S. (2005) Lancet

365, 1389–1397.[372] Cohen, C., Perrault, G., Voltz, C., Steinberg, R. and Soubrie, P. (2002) Behav. Pharma-

col. 13, 451–463.[373] Cohen, C., Perrault, G., Griebel, G. and Soubrie, P. (2005) Neuropsychopharmacology

30, 145–155.[374] Mascia, M.S., Obinu, M.C., Ledent, C., Parmentier, M., Bohme, G.A., Imperato, A.

and Fratta, M. (1999) Eur. J. Pharmacol. 383, R1–R2.[375] Solinas, M., Panlilio, L.V., Antoniu, K., Pappas, L.A. and Goldberg, S.R. (2003) J.

Pharmacol. Exp. Ther. 306, 93–102.[376] Colombo, G., Vacca, G., Serra, S., Salvatore, S., Carai, M.A.M. and Gessa, G.L. (2004)

Eur. J. Pharmacol. 498, 119–123.[377] Gessa, G.L., Serra, S., Vacca, G. and Carai, M. (2005) Alcohol Alcohol 40, 46–53.[378] Melzer, H.Y., Arvanitis, L., Bauer, D. and Rein, W. (2004) Am. J. Psychiatry 161, 975–984.[379] Molff, M.C. and Leander, J.D. (2003) Eur. J. Pharmacol. 477, 213–217.[380] Arnone, M. (2003) PCT Intl. Appl., WO 03 82256; (2003) Chem. Abstr. 139, 281284.[381] Bourrie, B. and Casellas, P. (2001) PCT Intl. Appl., WO 01 24798; (2001) Chem. Abstr.

134, 285593.[382] Hagmann, W.K., Lin, L.S., Shah, S.K., Guthikonda, R.N., Qi, H., Chang, L.L., Liu, P.,

Armstrong, H.M., Jewell, J.P. and Lanza, T.J., Jr. (2003) PCT Intl. Appl., WO 03 77847;(2003) Chem. Abstr. 139, 276471.

[383] Rinaldi-Carmona, M., Barth, F., Millan, J., Derocq, J.-M., Casellas, P., Congy, C.,Oustric, D., Sarran, M., Bouaboula, M., Calandra, B., Portier, M., Shire, D., Breliere, J.-C.and Le Fur, G. (1998) J. Pharmacol. Exp. Ther. 284, 644–650.

[384] Barth, F., Millan, J., Oustric, D., Rinaldi, M. and Vernhet, M. (2001) PCT Intl. Appl.,WO 01 32629; (2001) Chem. Abstr. 134, 340501.

Page 123: [Progress in Medicinal Chemistry] Progress in Medicinal Chemistry Volume 44 Volume 44 || 6 Recent Progress in Cannabinoid Research

J. ADAM ET AL. 329

[385] Ross, R.A., Brockie, H.C., Stevenson, L.A., Murphy, V.L., Templeton, F.,Makriyannis, A. and Pertwee, R.G. (1999) Br. J. Pharmacol. 126, 665–672.

[386] Iwamura, H. and Ueda, Y. (2004) US Appl., US-20040171613; (2004) Chem. Abstr. 141,218961.

[387] Mussinu, J.-M., Ruiu, S., Mule, A.C., Pau, A., Carai, M.A.M., Loriga, G.,Murineddu, G. and Pinn, G.A. (2003) Bioorg. Med. Chem. 11, 251–263.


Recommended