<<

CNS Drug Reviews Vol. 11, No. 2, pp. 195–212 © 2005 Neva Press, Branford, Connecticut

Bremazocine: A ê- with Potent and Other Pharmacologic Properties

Juanita Dortch-Carnes1 and David E. Potter2

1Department of Pharmacology/Toxicology, Morehouse School of Medicine, Atlanta, GA, USA; 2Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, USA

Keywords: Analgesia — — Diuresis — — ê-Opioid — Ocu- lar hypotension — Respiration.

ABSTRACT

Bremazocine is a ê- agonist with potent analgesic and activities. As an analgesic it is three- to four-times more potent than , as determined in both hot plate and tail flick tests. Bremazocine and other analogs were synthe- sized in an effort to produce with greater ê-opioid receptor selectivity and with minimal morphine-like side effects. Unlike morphine bremazocine is devoid of physical and psychological dependence liability in animal models and produces little or no respi- ratory depression. While bremazocine does not produce the characteristic euphoria asso- ciated with morphine and its abuse, it has been shown to induce dysphoria, a property that limits its clinical usefulness. Similarly to morphine, repeated administration of bremazo- cine leads to tolerance to its analgesic effect. It has been demonstrated that the marked di- uretic effect of bremazocine is mediated primarily by the central nervous system. Because of its side effects (disturbance in the perception of space and time, abnormal visual experience, disturbance in body image perception, de-personaliza- tion, de-realization and loss of self control) bremazocine has limited potential as a clinical analgesic. However, its possible utility for the therapy of and drug addiction war- rants further consideration because of its ability to decrease ethanol and self-ad- ministration in non-human primates. In addition, the ability of bremazocine-like drugs to lower intraocular pressure and to minimize ischemic damage in animal models suggests their possible use in the therapy of glaucoma and cardiovascular disease.

Address correspondence and reprint requests to: Juanita Dortch-Carnes, Ph.D., Morehouse School of Med- icine, 720 Westview Dr. S.W., Atlanta, GA 30310-1495, USA. Tel.: +1 (404) 752-1755, Fax: +1 (404) 752-1164, E-mail: [email protected].

195 196 J. DORTCH-CARNES AND D. E. POTTER

Fig. 1. Absolute stereochemistry of bremazocine enantiomers.

OVERVIEW

Bremazocine is a benzomorphan analog that is commonly classified pharmacologically as a relatively selective ê-opioid receptor agonist. Although bremazocine functions as a potent analgesic when administered peripherally and centrally (87,88,99), it does not dem- onstrate cross-tolerance with morphine, produces minimal respiratory depression at anal- gesic doses, and substitutes for other ê-agonists in operant drug discriminatory procedures (87,106,117). As ê-opioid agonists appear to be more effective in females as compared to males. In some experimental settings, bremazocine has been shown to in- teract with other opioid receptors as well. For example, it possesses antagonist-like prop- erties at the ì- and ä-receptors (30,88,117). Because of other pharmacologic actions, bre- mazocine or similar drugs might have clinical utility in diseases such as glaucoma, arthritis and inflammatory bowel syndrome.

DISCOVERY

Bremazocine, (2-[1-hydroxy-cyclopropylmethyl]-5-ethyl-9,9-dimethyl-2¢-hydroxy-6, 7-benzomorphan) is a member of the class of 9,9-dimethyl-6,7-, and was first synthesized at A. C. F. Chemiefarma and N. V. Maarssen in the Netherlands (99). It was synthesized in an effort to develop drugs that had greater receptor selectivity for ê-opioid receptors, because available evidence suggested that such agents would be powerful analgesics without morphine-like side effects. Efforts to optimize morphine-like compounds produced benzomorphan derivatives, ethylketocyclazocine (41,112) and bremazocine (99), both with relative selectivity for the ê-opioid receptor. The enantiomers of bremazocine (Fig. 1) have been found to have very different affinities at opioid receptors with different pharmacological activities and pro- files (99). The opioid effects of racemic bremazocine have been found to be due largely to the agonist activity of its (–)-enantiomer at the ê-opioid receptor (99,21,14). The (–)-enan- tiomer of bremazocine is an important tool for opioid receptor studies; the (+)-enantiomer, which does not interact avidly with opioid receptors, has some of the non-opioid pharma- cological effects of bremazocine.

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 197

PHARMACOLOGY

Studies Characterizing the Analgesic Activity of Bremazocine Bremazocine, a potent, long-acting agonist that interacts with ê-opioid receptors, has been shown to be three- to four-times more potent than morphine in hot plate and tail flick tests in mice (99). Its analgesic activity in the tail flick test was antagonized more effec- tively by Mr-2266, a relatively selective ê-, than by , a non- selective opioid receptor antagonist. Unlike morphine, bremazocine does not produce mydriasis or Straub tail phenomenon in mice. Moreover, it is devoid of respiratory de- pressant activity in rats. Initially, chronic administration of bremazocine to monkeys did not lead to a morphine-like withdrawal syndrome, although tolerance to its analgesic ef- fects occurred; and in this paradigm, morphine remained an effective analgesic. Con- versely, treatment of morphine-tolerant animals with bremazocine did not cause analgesia. These data were interpreted as suggesting that ì-receptors are largely unoccupied by bre- mazocine and that ê-opioid receptors do not mediate analgesia. Subsequently, however, Spencer and coworkers demonstrated that bremazocine has analgesic activity in mice that is closer to that of than to ethylketocyclazocine (114), using four antinocicep- tive tests. These results indicated that bremazocine may be active at ì- as well as ê-opioid receptors. The fact that abrupt cessation of administration of bremazocine precipitated a withdrawal syndrome in morphine-dependent rats supported this conclusion (115). As alluded to previously, in initial tests for analgesic activity, bremazocine was shown to be three- to four-times more potent than morphine (99). The two most common behav- ioral assays to assess such changes are the hot plate and tail flick tests, where a heat source is applied to either the hind paw or tail of an animal. Studies carried out in the late 1980s demonstrated the ability of bremazocine to produce an increase in the withdrawal re- sponse latency to noxious hind paw thermal stimulation in rats as early as two days after birth (50). In addition, bremazocine selectively inhibited the late C-fiber reflex discharge in cats, recorded in sectioned lumbo-sacral ventral root filaments, after supramaximal electrical stimulation of the ipsilateral sural or common peroneal nerve (5). Both effects were demonstrated to be dose-dependent and reversible by opioid antagonists. Further- more, in a study using nonhuman primates, bremazocine and other ê-agonists produced dose-dependent increases in tail-withdrawal latencies from both 50 and 55°C water (31). Studies examining bremazocine’s analgesic potential continued into the 1990s and its antinociceptive effects in rodents (6,52) and nonhuman primates (62,63) were again dem- onstrated to be dose-dependent and opioid receptor-mediated. After evaluating data related to the analgesic activity of bremazocine (Table 1), Romer and co-workers found that there were differences in the potency of the ê-opioid agonist when it was administered parenterally versus orally (99). When injected subcutaneously, bremazocine was determined to be three to four-times more potent than morphine as an analgesic agent. However, after oral administration to mice, bremazocine was only half as potent as morphine in the hot plate test, but three-times more potent in the tail flick pro- cedure. In the mouse, when compared to ketocyclazocine (a short-acting ê-opioid ag- onist), bremazocine was between three- and four-times less potent after subcutaneous ad- ministration, but approximately twice as potent by the oral route. When using the shock titration method (24) in the rhesus monkey (Table 2), bremazocine was found to be 180- and 57-times more potent than morphine or ketocyclazocine, respectively, when adminis-

CNS Drug Reviews, Vol. 11, No. 2, 2005 198 J. DORTCH-CARNES AND D. E. POTTER tered intravenously. In addition, bremazocine had a three-fold longer duration of action than ketocyclazocine. In another study, Furst compared the analgesic and sedative efficacy of ì- and ê- as a function of time, in rats and mice (39). This study concluded that three opioid agonists with ê-receptor activity (bremazocine, ethylketocyclazocine, and pentazocine) were inactive against heat nociception, but like ì-agonists they produced potent, long-lasting analgesia in the acetic acid writhing test. In addition, when bremazo- cine was co-administered with morphine, there was a significant prolongation of the du- ration of analgesic action, without any influence on the potency. Furthermore, time-re- sponse curves and ED50 values of ê-opioid agonists were similar to those of ì-opioid agonists. The time-course of the analgesic effect of bremazocine in the mouse writhing test is compared to other ê-opioid agonists in Table 3. The adverse side effects of bremazocine include sedation, respiratory depression, diuresis and constipation. Hayes and Tyers examined the involvement of ì- and ê- receptors in several opiate-like effects in conscious mice, which may correspond to clinical side effects (48). In this study, the authors tested a variety of opiate drugs with dif- ferent selectivities for ì- and ê-opioid receptors for their effects on behavior, pupil di- ameter, body temperature, respiratory rate, and gastrointestinal propulsion. The investi- gators concluded that ì-receptor agonists produce their antinociceptive effects as well as opiate-like side effects by interacting with the ì-receptors. In regard to the ê-receptor agonists, such as bremazocine, the opiate-like side effects occurred at much higher doses than those needed for antinociception, suggesting that the ê-opioid agonist-induced side effects were due either to interaction with the ì-receptor or to some other non-specific action.

TABLE 1. Analgesic activity of bremazocine, ketocyclazocine and morphine, and their antagonism by naloxone, and Mr-2266 in mice

Test (n = 10, Parameter per dose) (mg/kg) Route Antagonist Bremazocine Ketocyclazocine Morphine d a b Hot plate ED50 s.c. None 0.7 0.17 2.1 p.o. 12d 20a 5.8c

Tail flick ED50 s.c. None 0.7 0.26 3.0 p.o. 7.5 18 20.5

Antagonism AD50 i.v. Naloxone >3.2 0.07 0.005 of analgesia (–44%) (tail flick) i.v. Mr-2266 0.46 0.043 0.008 a b c d Note. Optimal pretreatment times for the drugs were 15, 30, 60, 90 min; ED50, effective dose in 50% of the animals; AD50, antagonistic dose in 50% of the animals.

TABLE 2. Analgesic activity of bremazocine, ketocyclazocine, and morphine in rhesus monkeys Bremazocine Ketocyclazocine Morphine Test Parameter Route (n =5) (n =2) (n =4) Shock titration MED (mg/kg) i.v. 0.0056 0.32 1 Duration of action (min) 200 60 200 Note. MED, minimal effective dose. Data from ref. 99.

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 199

Opioid Receptor Selectivity/Signal Transduction

The existence of multiple opioid receptor subtypes has been long recognized. These re- ä ê ì ceptors were initially termed , , and ; according to new nomenclature they are OP1, OP2, and OP3 receptors, respectively (71,75). More recently, the existence of opioid re- ceptor subtypes has been confirmed by molecular cloning (17,35,122). ê-Opioid receptors are believed to arise first in evolution as indicated by nociception research in amphibians (110), followed by ä-opioid and ì-opioid receptors. Interestingly, the brain of amphibians expresses mostly ê-opioid receptor binding sites. Putative ê-opioid receptor subtypes are differentially labeled by arylacetamides (e.g., ) and benzomorphans (e.g., bre- ê ê mazocine): 1-receptors bind arylacetamides more avidly, whereas 2-receptors bind benzomorphans more selectively (100). The explanation for this differential binding, based upon structure-activity relationship, could be due to: 1) the putative existence of dif- ferent affinity states (i.e., high affinity versus low affinity) of the same ê-opioid receptor; or 2) the existence of homo/heterodimer formation between various opioid receptors. For example, bremazocine has very similar affinity for the ê- and ì-opioid receptors but much less affinity for the ä-opioid receptor. Therefore, the non-selectivity and/or relative selec- tivity of bremazocine in a particular tissue could be based on either the existence of heterodimers consisting of ê-opioid receptor/ì-opioid receptor, or the preference for the low affinity state of the ê-opioid receptor homodimer. It should be noted that (–)-opioid receptor agonists can produce biological effects that are independent of opioid receptors particularly at higher doses. At low concentrations (nM), ê-opioid receptor agonists me- diate spinal analgesia by actions on ê-opioid receptors, but at higher concentrations (ìM), cation (K/Ca) channel activity can be impaired independent of the activity at opioid re- ceptors. With respect to spinal analgesia, there is evidence for gender-linked differences in the antinociceptive activity (46). For example, female rats show fluctuations in ê-opioid receptor density and sensitivity across the estrous cycle. Likewise, human females are ê reported to be more sensitive than males to the analgesic action of 2-opioid receptor agonists.

TABLE 3. Time course of the analgesic effect of ê-agonists in the mouse writhing test Percentage analgesia at time (min) Mean time (min) Dose, after drug administration* to reach 50% s.c. of maximal Compound (mg/kg) 10 20 30 60 90 120 analgesic effect Pentazocine 5.0 20 18 22 20 13 10 58 10.0 40 48 54 49 38 22 15.0 48 62 76 70 68 55 EKC 0.050 38 45 40 39 32 18 79 0.100 49 60 58 48 41 29 0.250 67 80 78 79 68 40 Bremazocine 0.05 66 NT 56 51 25 NT 64 0.10 88 NT 85 75 51 2 0.25 100 NT 100 88 75 23 *n/dose = 10 mice; EKC, ethylketocyclazocine; NT, not tested. Data from ref. 39.

CNS Drug Reviews, Vol. 11, No. 2, 2005 200 J. DORTCH-CARNES AND D. E. POTTER

Studies conducted in the early to mid 1980s on isolated guinea pig ileum (77) and mouse vas deferens (46) indicated a preference of bremazocine for ê-opioid receptors. Binding studies with [3H](–)-bremazocine performed using rat brain membrane prepara- tions have shown that this compound differs in its binding properties from previously in- vestigated exogenous or endogenous opioids (99). In contrast to morphine, bremazocine did not produce mydriasis or the Straub tail phenomenon in mice. In addition, it lacks any significant effects on respiration in monkeys; likewise, programmed administration in the same species does not lead to a morphine-like withdrawal syndrome upon cessation of drug treatment or upon naloxone challenge. More recent studies utilizing the highly se- lective ê-opioid receptor antagonist, (nor-BNI), confirmed that in many biological systems the effects of bremazocine are mediated primarily by ê-receptors (14,26,53,93,94,101). James and Goldstein developed a method for measuring binding selectivity of ligands for multiple opioid receptors. By using radioligands, that were partially selective for each receptor type, in combination with brain membranes enriched in a particular receptor type, they found that the selectivity profile for bremazocine was 3.1, 19, and 10.1 for ì, ä, and ê receptors, respectively (59). Because the anatomical distribution of ê-opioid receptors in the central nervous system across species has shown widespread and significant differences, it is important to con- sider the ê-opioid receptor distribution in humans. As shown by RT-PCR, expression of ê-opioid receptor mRNA in the human brain is consistent with the involvement of ê-opioid receptors in pain perception, neuroendocrine physiology, and affective behavior and cognition (107). In general, the distribution of ê-opioid receptors follows the pattern of -derived peptides. The cloned ê-opioid receptor has high affinity for -A (1-17), an endogenous ê-opioid agonist (16), and for the synthetic ê-selective ligands U-50488H and U-69593, confirming that the cloned receptor is of the ê-opioid receptor type. Pharmacological studies with ê-selective ligands demonstrated the existence of two distinct subtypes of ê ê -opioid receptors, one of which is U-69593-sensitive ( 1) and the other bremazocine-sen- ê ê ê ê sitive ( 2). Both subtypes of the -opioid receptors ( 1 and 2) have been further divided into four types (19,86,127). The absence of subdivision-specific antagonists limits the studies of pharmacological properties of these putative subdivision-selective agonists. It ê has been shown that the application of a 1-selective, irreversible antagonist inhibited the ê antinociceptive effect of U-69593 (a 1-selective agonist) without inhibiting the agonist ê property of bremazocine (a 2-selective agonist), suggesting the existence of subdivisions ê ê within the 1- and 2-subtypes (113). However, it is possible that the subdivisions within the respective subtypes could represent either different affinity states of a (–)-opioid re- ceptor subtype, or homo/heterodimers. Although widely used as a ê-opioid agonist, bremazocine has been reported to have roughly equal affinities at the ì-, ä-, and ê-binding sites (72). Bremazocine, as well as other ê-opioid receptor agonists, have been demonstrated to have moderate affinity for the ó ó 1-receptor subtype. The -receptor was initially classified as an opioid receptor (45). However, since the time it was cloned in 1996 (75), it has become evident that the ó-re- ceptor is a single transmembrane-spanning protein targeted by other drugs of abuse and is no longer considered to be a member of the opioid receptor family. At the cellular level, it has been demonstrated that, ê-opioid receptors are predominant- / ly coupled to heterotrimeric Gi Go proteins that are sensitive to pertussis toxin (PTX). In

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 201

ê / some cells, the coupling of -opioid receptors to PTX-insensitive Gs Gz proteins has been described. In the native state, activation of the ê-opioid receptors usually evokes stimu- lation of phospholipase-Câ-induced inhibition of adenylyl cyclase, inhibition of vesicular release of neurotransmitters and interactions with a number of cation channels, including K+-channels (G-protein inwardly rectifying, delayed rectifying, and Big-K) and voltage- sensitive Ca2+ channels. In most excitable tissues, K-channel activities are stimulated whereas calcium channel activity is inhibited. The activities of the described effectors are modulated by both the GTP-bound form of the Gs-subunit as well as free Gâã-subunits. The Gi-mediated events are modulated by RGS proteins that reduce the lifetime of Gâã-GTP thereby terminating its effects. Liberated Gâã subunit is presumed to activate spe- cific isoforms of phosphatidyl inositol 3-kinase; subsequently, this event leads to acti- vation of Ras and the Raf/MEK/Erk kinase cascade promoting protective and/or anti- apoptotic effects (111). Bremazocine has been shown to be a pure antagonist at the ä-re- ceptor in the hamster vas deferens (76), and at the ì-receptor in the rat vas deferens (42). It has now been accepted that the cellular activities of ê-opioid receptors are modulated dy- namically by dimerization and by desensitization/downregulation. In contrast to ì-opioid receptors and ä-opioid receptors, ê-opioid receptors do not seem to be predisposed to endocytosis. Studies conducted in our laboratory using the isolated iris-ciliary body preparation from New Zealand White rabbits have demonstrated a biphasic or bimodal effect of bre- mazocine on cAMP accumulation (101). A a concentration of 0.0001 ìM bremazocine en- hanced isoproterenol (ISO)-stimulated cAMP accumulation, whereas at concentrations of 0.01 to 10 ìM, bremazocine was inhibitory. However, in the absence of stimulation by ISO, bremazocine at the same concentrations elevated cAMP levels above base levels. These effects were antagonized by pretreatment with nor-BNI suggesting that (–)-opioid receptors were involved. Inhibitory effects on cAMP accumulation could be antagonized by pretreatment with PTX indicating the involvement of a Gi protein. The biphasic nature of cAMP in this tissue could reflect differences in the activation (or inhibition) of isoforms of either adenylyl cyclase (formation) or phophodiesterase (degradation).

Effects on Motor Activity In rodents, ì- and ä-agonists usually increase locomotion, whereas ê-agonists decrease locomotion (73) in addition to inducing ataxia and sedation (57). However, in preweanling (29) and monoamine-depleted rats (54), ê-opioid agonists have been reported to markedly increase locomotor activity. In Syrian hamsters, ê-agonists elicit biphasic effects inducing hyperactivity at lower doses and hypoactivity at higher doses (105). In a study comparing the ability of the ê-opioid agonists U-50488H, BRL-52537, and bremazocine to modify spontaneous motor activity in mice (66), it was determined that higher analgesic doses of each of the ê-agonists reduced rearing, motility, and locomotion in non-habituated an- imals. These effects were blocked by the selective ê-opioid receptor antagonist, nor-BNI, and the irreversible, relatively selective ê-receptor antagonist, DIPPA. In contrast, lower sub-analgesic doses of bremazocine (0.075 mg/kg) time-dependently increased motor ac- tivity that were not blocked by nor-BNI or DIPPA, but were completely eliminated by naloxone (0.1 mg/kg), thus indicating that the stimulatory effects of the ê-opioids on loco- motor activity were mediated, in part, by opiate receptors other than the ê-subtypes. The stimulatory effects of U-50488H and bremazocine were not observed in habituated animals.

CNS Drug Reviews, Vol. 11, No. 2, 2005 202 J. DORTCH-CARNES AND D. E. POTTER

Effects on Ethanol and Cocaine Use

Endogenous opioid peptides have been suggested to be involved in ethanol and cocaine addiction (51,83). In regard to the effects of bremazocine on alcohol and drug use, Nestby and colleagues reported that bremazocine displayed a mixed-action at multiple types of opioid receptors in rat brain (83). In addition, bremazocine caused a prolonged inhibition of 24 h free-choice ethanol self-administration without affecting natural reinforcement and without rapid development of tolerance to its inhibitory effect. In a separate study conducted by Cosgrove and Carrol, pretreatment with bremazocine dose-dependently de- creased self-administration of cocaine, ethanol and phenylcyclohexylpiperidine (PCP) (22). This effect was generalized to non-drug reinforcers (saccharin and food) indicating that bremazocine did not selectively decrease drug-maintained behavior. Bremazocine treatment was also effective in reducing the demand and response strength of smoked co- caine (base), oral ethanol and PCP. Mello and Negus found that bremazocine and two other ê-agonists reduced cocaine self-administration in rhesus monkeys. This suppression of self-administration of cocaine was maintained over 10 days of drug treatment (78).

Discriminative Stimulus Effects

The discriminative stimulus property of bremazocine was first demonstrated in the rat (106). This characteristic was determined to be a result of an interaction of bremazocine with ê-opioid receptors and ó-receptors; the latter receptors were formerly considered part of the opioid receptor family (75), but are now considered to constitute a distinct group of receptors (97). Studies in pigeons (91), squirrel monkeys (82,94), and rats (92) produced further evidence of the discriminative stimulus properties of bremazocine and other ê-opioids (13). More recent studies have demonstrated that the discriminative stimulus effect of the ê-opioid receptor agonist, , could be reproduced by bremazocine and other ê-agonists (15).

Effects on Anterior Pituitary Hormones

ê-Opioid receptor agonists are known to cause demonstrable neuroendocrine effects in humans and nonhuman primates that include release of the anterior pituitary peptide hor- mones, such as prolactin (40,89,116). As early as 1976, it has been demonstrated that mor- phine and other opioid compounds such as the then new benzomorphan derivative, bremazocine, inhibit the secretion of luteinizing hormone (LH) in rats of both sexes (12,18,85). Marko and Romer found that acute administration of bremazocine (0.005–1 mg/kg, s.c.) or morphine (10–20 mg/kg, s.c.) diminished serum LH levels and sponta- neous ovulation in female rats in a dose-dependent manner (74). Chronic treatment with bremazocine significantly diminished LH and testosterone secretion in male rats, which in turn led to a reduced weight of the prostate gland, while prolactin and follicle stimulating hormone (FSH) secretions were not significantly affected. Pretreatment with the ì-re- ceptor antagonist, naloxone, which increases LH secretion in rats, significantly antago- nized the inhibiting effect of morphine, but not that of bremazocine on LH secretion. It was, therefore, concluded that morphine and bremazocine showed qualitative and quanti- tative differences in their endocrine activity profiles.

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 203

Diuretic Effect

ê-Opioid agonists are unique among opioid-receptor agonists in that they consistently produce diuresis over a relatively wide dose-range (68,69); other opioids, such as mor- phine, can produce both antidiuretic and diuretic effects depending on the dose and species used (4). Synthetic benzomorphans such as ethylketocyclazocine, , and bremazocine, have been found to increase urine output in normally hydrated as well as in water-loaded animals (55,68,69,98,103,109). Several studies in rodents have reported that the ê-opioid receptor-mediated diuretic effect is due mainly to inhibition of release from the neurohypophysis (69,84,121). Studies in primates also suggest the in- volvement of vasopressin (ADH) in the diuretic effects of ê-agonists (7,64) as well as the presence of ê-opioid receptors in relevant hypothalamic nuclei (90,108). Although one study has shown that the diuretic effect of bremazocine is mediated partially in the periphery (103), most studies indicate that the diuretic effect of ê-agonists is mediated primarily in the central nervous system (11,60,68).

Cardiovascular Effects

The action of opioids on the cardiovascular system varies according to dose, species examined, and route of administration (33,104). Some studies have demonstrated a mor- phine-mediated cardiovascular effects linked to the central nervous system (37,67). How- ever, in intact animals, bremazocine activates ê-opioid receptors centrally and peripherally to produce dose-dependent hypotension and bradycardia. These effects are attributable to inhibition of sympathoadrenal outflow as demonstrated by reductions in plasma levels of norepinephrine (34). El-Sharkawy and colleagues performed in vitro experiments that generated evidence of the direct effects of bremazocine (32) and other opioid agonists (9) on vascular smooth muscle. The effects of the agents varied depending on the agonist used, the concentration tested, and the pre-contracting agent used. In aortic rings pre-con- tracted with norepinephrine, U-50488H, ethylketocyclazocine (EKC), and morphine caused constriction at low concentrations, but relaxation at high concentrations; bremazo- cine caused only relaxation over a relatively wide dose range. Bremazocine did, however, produce contraction in tissues pre-contracted with prostaglandin F2á. Dependence on the pre-contracting agent may have its basis in the difference between the mechanisms by which various pre-contracting agents evoke increases in vascular smooth muscle tone. In studies on pithed rabbits with continuously stimulated sympathetic outflow, EKC (33) and bremazocine (34) were shown to persistently decrease the release of norepinephrine and, consequently, lower blood pressure. In separate studies, bremazocine (0.2 mg/kg, i.v.) produced a slight decrease in systolic blood pressure only at 45 min after drug adminis- tration (P < 0.02); no change in diastolic or mean blood pressure was observed at this time point. A marked decrease in blood pressure was observed with 0.4 mg/kg, i.v. bremazo- cine; the peak effect was observed at 60 min after drug administration (P < 0.0005). The change occurred in both systolic and diastolic blood pressures. Higher doses (0.6 mg/kg, i.v.) of bremazocine did not produce any additional change in systolic, diastolic or mean blood pressures. A significant dose-dependent decrease in heart rate was produced by bremazocine (0.2–0.6 mg/kg, i.v.). The bradycardia was most marked at 60 min after drug administration.

CNS Drug Reviews, Vol. 11, No. 2, 2005 204 J. DORTCH-CARNES AND D. E. POTTER

In the heart, ischemic preconditioning (IPC) is a phenomenon of increased resistance to cellular injury or prolonged ischemia that follows exposure to single or multiple periods of brief coronary occlusion. The phenomenon has two phases: an acute or early phase that lasts for about 1–3 h, and a delayed phase that appears approximately 24 h after the acute phase. The delayed phase may last for up to 72 h. Opioid receptor activation, specifically ä and ê, seems to be an important component of both the acute and delayed phases of IPC. Bremazocine has been reported to exacerbate infarct size in the isolated rat heart through an as yet unknown mechanism (1). In contrast, prior stimulation of ê-opioid re- ceptors with the selective agonist, U-50488H, reduced lactate dehydrogenase (LDH) ac- tivity (an index of cell injury) and increased the percentage of non-blue cells (an index of viability) in the culture medium of rat ventricular myocytes (126). The induction of heat shock protein (HSP-70) appears to mediate the delayed cardioprotection induced by pre- treatment with U-50448H. Both the “anti-preconditioned state” (bremazocine) and the de- layed cardioprotection (U-50448H) were antagonized by the relatively selective antag- onist, nor-BNI. These seemingly paradoxical findings could reflect the biphasic effect bremazocine has on signal transduction pathways such as adenylyl cyclase/cAMP. In other cases, ê-receptor agonists, particularly those in the arylacetamide class, blocked Na/K channels in the heart that are independent of action on “classical” opioid receptors (96). These receptor-independent effects could account for the activity of ê-receptor agonists in models of ischemia and/or arrhythmias.

Respiratory Effects Respiratory depression is a clinically significant effect of many opioid analgesics. The respiratory depressant effects of opioids are dose-related, stereospecific and reversed by opioid antagonists (59), indicating that the respiratory effects are mediated through opioid receptors. ì-Selective agonists such as morphine, induce pronounced effects on respi- ration at analgesic doses (61). In contrast, ê-selective opioids, like bremazocine (38,99), produce only modest suppressive effects on respiration at doses that produce analgesia in dogs (38) and rodents (48,99). Studies with primates generated slightly different results (14) in that bremazocine and other ê-agonists were found to produce significant respi- ratory depression at high doses. In the same study, the most selective ê-agonist studied, U-50488H, had only modest effects on respiration at all doses; thereby, suggesting that the respiratory effects of the opioids used were not mediated through a ê-receptor mechanism. Hayes and Tyers found that the analgesia produced by Mr-2034 and bremazocine was me- diated through ê-receptors. It was concluded that the opioid side effects that appeared at much higher doses, including respiratory depression induced by bremazocine, could be at- tributed to an interaction with the ì-receptor or to some other nonspecific action (48). In any event, the studies in non-human primates reinforce the suggestion that the respiratory depressant effects of bremazocine and other more non-selective ê-agonists occur at much higher doses than those required to induce analgesia and behavioral changes.

Effects on Neurotransmission Although a few reports have been generated concerning the effects of ê-opioid agonists on biogenic amine-derived neurotransmitters such as norepinephrine, acetylcholine and the amino acid neurotransmitters (aspartate, glutamate, and ã-aminobutyric acid), for the most part the effects of ê-agonists on neurotransmission in the central nervous system are

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 205 largely associated with dopamine. It is well documented that dopaminergic activity can be modulated by more than one type of opioid receptors. In general, it is agreed that ì-se- lective agonists increase, whereas ê-selective agonists decrease dopamine release in the brain (23). In keeping with this premise, bremazocine has been demonstrated to inhibit the release of dopamine in the nucleus accumbens and in the dorsal caudate of freely moving rats (23). This effect was shown to be mediated by activation of ê-opioid receptors. Bremazocine’s effect on norepinephrine and amino acid neurotransmitter release has been shown to be inhibitory and also linked to activation of ê-receptors. Heijna and colleagues have generated evidence that the inhibitory effect of bremazocine on dopamine activity is linked to activation of ä-opioid receptors coupled to DA-sensitive adenylate cyclase (49). The finding that ì- and ê-agonists produce opposite effects on dopamine release would predict that ì- and ê-agonists might have also opposite effects on the reinforcing prop- erties of cocaine. However, if ê-agonists like bremazocine, EKC, Mr-2033 and enadoline act as low efficacy ligands for ì-receptors, the combination of low efficacy ì- and high efficacy ê-activity could result in complementary effects on cocaine self-administration. Archer and colleagues have suggested that the combination of a short-acting ê-agonist and a long-acting ì-antagonist could be especially useful for cocaine abuse treatment, because both ê-agonists and ì-antagonists reduce dopamine release from the nucleus accumbens (3). Because ê-opioid agonists, such as bremazocine, have been shown to inhibit veratri- dine-induced calcium influx and glutamate release in mammalian cerebral cortical slices (10), one would predict that activation of ê-opioid receptors should offer some level of neuroprotection. Indeed, Chen and colleagues demonstrated that pretreatment with BRL-52537 provided robust neuroprotection in rats without altering ischemia-evoked re- lease of dopamine and its metabolites (17). Perhaps this property could be utilized to treat other neuropathies, including the optic neuropathy that characterizes glaucoma.

Ocular Effects

Evidence of the presence of opioid receptors in the iris of rabbits and humans was first generated in the 1980s (27,28,36). In addition, the intraocular pressure lowering effect of morphine has been demonstrated in both animal and human subjects (28). Similar to mor- phine, bremazocine has been demonstrated to lower the intraocular pressure in rabbits (102). Further studies with bremazocine have generated information regarding the mech- anism of this effect. Bremazocine-induced ocular hypotension has been linked to activa- tion of ê-opioid receptors, because the response was inhibited by pretreatment with nor- BNI, a relatively selective ê-receptor antagonist (102). Furthermore, the intraocular pressure lowering effect of bremazocine was demonstrated to be associated with inhi- bition of cAMP and norepinephrine release in the iris-ciliary body (ICB) of rabbits (101), as well as increases in atrial natriuretic peptide (93,101) and inositol phosphate levels in the rabbit ICB preparation (26). More recently, bremazocine has been shown to elevate CNP levels in the aqueous humor of rabbits, and this event was associated with an increase in total outflow facility (93). Because the increase in outflow facility could be antagonized by pretreatment with either isatin (a purported NPR antagonist) or nor-BNI (a relatively selective ê-opioid re- ceptor antagonist), an association was made between the ability of bremazocine to elevate CNP/ANP levels and increase outflow facility. Thus, it was concluded that the enhanced

CNS Drug Reviews, Vol. 11, No. 2, 2005 206 J. DORTCH-CARNES AND D. E. POTTER egress of aqueous humor caused by bremazocine is mediated, in part, by a paracrine effect of natriuretic peptides on outflow pathways.

Clinical Relevance

ê-Opioid agonists are of particular clinical interest as analgesics because of their ability to modulate antinociceptive functions without ì-opioid-related side-effects including con- stipation, pruritus, and respiratory depression. However, the centrally-mediated effects of ê-agonists, such as sedation and dysphoria, tend to limit their potential clinical usefulness (89,119). It is possible that ê-agonists that do not penetrate the pial-glial barrier would have greater utility. Although no clinical trials have been conducted with bremazocine in humans, a number of clinically relevant experiments using various animal models have been per- formed. Initially, the majority of these type studies were centered on the possible use of bremazocine as an opioid analgesic that would be devoid of the bothersome side effects associated with the use of morphine. Indeed, bremazocine has been demonstrated to be a potent long-lasting analgesic in rodents and nonhuman primates. Clinical studies have in- dicated that sensitivity to analgesia induced by ê-opioid agonists is influenced to a signif- icant degree by gender (females > males). More recent studies with bremazocine in sub- human primates indicate the possibility of treating substance abuse (e.g., cocaine).

Adverse Effects, Tolerance, and Dependence

A number of clinical studies have revealed that members of the benzomorphan class of ê-opioids elicit psychotomimetic and dysphoric effects similar to those of the anesthetics [e.g., (PCP) and ]. Initial pharmacologic studies sug- gested that these effects of the ê-agonists were mediated by ó-(PCP) receptors (75,128). Later, it was determined that they were mediated principally by activation of ê-opioid re- ceptors (65,89). Because humans are the only species in which psychotomimetic parameters can be measured, a pertinent question that remains to be answered is: What are the human equiv- alents of the ê-opioid receptor-induced aversive effects in rodents and monkeys? Detailed clinical studies of the effects of the benzomorphan derivatives (44), N-allyl- normetazocine (25), and Mr-2033 (56,79,80), determined that these compounds induced some or all of the psychotomimetic effects mentioned above. Unlike morphine, bremazocine (32 mg/kg, s.c.) has not produced mydriasis or the Straub tail phenomenon in mice, nor has it induced respiratory depression in rats (99). Bremazocine’s adverse effects and development of tolerance have been examined most extensively in pre-clinical studies using monkeys. However, as with morphine, repeated administration of bremazocine has led to the development of tolerance to its analgesic effect. Mello and Negus demonstrated evidence of transient sedation and occasional vom- iting produced by bremazocine in in a preclinical chronic study including monkeys (78). In a separate study of monkeys that were programmed to self-administer bremazocine, neither naloxone challenge nor cessation of drug treatment led to morphine-like with- drawal symptoms (99). Only yawning, head shaking, and some retching were observed. In a self-administration human study, despite continual access to the drug for a total of six weeks, bremazocine was not self-administered, thus suggesting that it may have consid-

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 207 erably less abuse potential than morphine in humans as well (99). However, bremazocine has very limited potential as a therapeutically useful analgesic due to its psychotomimetic side-effects.

CONCLUSIONS

ê-Opioid receptor agonists have become important pharmacological tools in studying a variety of functions modulated by opioid receptors (2,125). Preclinical studies suggest the possibility of their utility in specific therapeutic areas. For example, they are being eval- uated in the treatment of cocaine abuse (20,81) due to their ability to lower excessively el- evated dopamine levels in the nucleus accumbens. More recently, ê-agonists have been considered for their potential usefulness in the therapy of HIV-1 (70) and ischemia-related cardiovascular disorders (95). Future preclinical studies examining the potential usefulness of bremazocine and other ê-opioid agonists as potential therapy for the above mentioned chronic disease states will have to ultimately address the issues of tolerance, duration of action, and severity of unde- sirable side effects such as emesis, sedation and dysphoria. Alternatively, centrally-me- diated side effects can be minimized by site-directed delivery or new formulations. The use of combinations of low efficacy ì- and high efficacy ê-agonists, as was suggested for therapy of cocaine addiction, should also be considered. Another potential therapeutic in- dication for peripherally acting ê-opioid receptor agonists (e.g., asimadoline) is arthritis (8,118). ê-Opioid receptor agonists act at multiple sites in the inflammatory cascade to: 1) reduce adhesion molecule expression; 2) inhibit cell trafficking; 3) suppress mRNA ex- pression and protein levels of substance-P and calcitonin gene-related protein; and 4) reduce TNF-(expression and release). The ability of ê-opioid receptor agonists to sup- press TNF-(expression) could also have some potential benefit in treating glaucomatous neurodegeneration (120,123). Currently, further research with bremazocine is limited by the availability of the com- pound. However, one group has developed a novel method of synthesizing the ê-opioid enantiomers (43), thereby facilitating their availability for continued use as pharmaco- logical tools for the in vitro and in vivo exploration of the ê-opioid receptor system. Additional preclinical research with ê (–)-opioid agonists, such as bremazocine, is needed to determine their potential utility in the treatment of: 1) drug/alcohol dependence; 2) gastrointestinal disorders, such as irritable bowel syndrome; 3) cardiovascular diseases; 4) arthritis; and 5) glaucoma. Acknowledgments. The research on bremazocine’s ocular effects was supported by NIH/NEI grants EY-011977, EY-012807 and EY-014793. The authors express their sincere appreciation to Luanna Bartholomew for her assistance in preparing this review.

ADDENDUM. Chemical Names of Compounds ANP, atrial natriuretic peptide; BRL-52537,(+/–)-dichlorophenyl)acetyl-2-(1-pyrrolidinyl)methylpiperadine; CNP, C–type natriuretic peptide; DIPPA, 2-(3,4-dichlorophenyl)-N-methyl-N-[(1S)-1-(3-isothiocyanatophenyl)-2-(1-pyrrolidinyl)-

CNS Drug Reviews, Vol. 11, No. 2, 2005 208 J. DORTCH-CARNES AND D. E. POTTER ethyl]acetamide; EKC, ethylketocyclazocine; HSP-70, heat shock protein 70; Mr-2266, (–)-(1R,5R,9R)-5,9-diethyl-2-(3–furylmethyl)-2¢-hydroxy-6,7-benzomorphan; Mr-2033,(+/–)-(1-R/S,5-R/S,2=R/S)-5,9-dimethyl-2¢-hydroxy-2-tetrahydrofurfuryl-6,7-benzo- morphan; Mr-2034, (–)-N-(2-tetrahydrofurfuryl)-normetazocine; Nor-BNI, nor–; U-50488H, 3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzene-acetamide; U-69593, trans-3,4-dichloro-N-methyl-N-[7-(1-pyrrolidinyl)cyclohexyl]benzene-acetamide methanesulfonate.

REFERENCES

1. Aitchison KA, Baxter GF, Awan MM, Smith M, Yellon DM, Opie LH. Opposing effects on infarction of ä- and ê-opioid receptor activation in the isolated rat heart: Implications for ischemic preconditioning. Basic Res Cardiol 2000;95:1–10. 2. Amarante LH, Duarte ID. The ê-opioid agonist (+/–)-bremazocine elicits peripheral antinociception by acti- vation of the L-arginine/nitric oxide/cyclic GMP pathway. Eur J Pharmacol 2002;454(1):19–23. 3. Archer S, Glick SD, Bidlack J. Cyclazocine revisited. Neurochem Res 1996;21:1369–1373. 4. Aziz LA, Forsling ML, Woolf CJ. The effect of intracerebroventricular injections of morphine on vaso- pressin release in the rat. J Physiol 1981;311:401–409. 5. Banna NR, Al-Amen HA, Jabbur SJ. Selective inhibition of nociceptive flexion reflex discharge by the ê agonist bremazocine. Neuropharmacology 1987;26(2/3):271–274. 6. Bartok RE, Craft RM. Sex differences in opioid antinociception. J Pharmacol Exp Ther 1997;282(2): 769–778. 7. Bellissant E, Denolle T, Sinnassamy P, et al. Systemic and regional hemodynamic and biological effects of a new ê-opioid agonist, , in healthy volunteers. J Pharmacol Exp Ther 1996;278:232–242. 8. Binder W, Machelska H, Mousa S, et al. Analgesic and anti-inflammatory effects of two novel ê-opioid pep- tides. Anesthesiology 2001;94(6):1034–1044. 9. Bolme P, Fuxe K, Agnati LF, Bradley R, Smythies J. Cardiovascular effects of morphine and opioid peptides following intracisternal administration in chloralose-anesthetized rats. Eur J Pharmacol 1978;48(3): 319–324. 10. Bradford HF, Crowder JM, White EJ. Inhibitory actions of opioid compounds on calcium fluxes and neuro- transmitter release from mammalian cerebral cortical slices. Br J Pharmacol 1986;88(1):87–93. 11. Brooks DP, Giardina G, Gellai M, et al. Opiate receptors within the blood-brain barrier mediate ê-agonist-in- duced water diuresis. J Pharmacol Exp Ther 1993;266:164–171. 12. Bruni JF, Van Vugt D, Marshall S, Meites J. Effects of naloxone, morphine and methionine on serum prolactin, luteinizing hormone, follicle stimulating hormone, thyroid stimulating hormone and growth hormone. Life Sci 1977;21(3):461–466. 13. Butelman ER, Baron SP, Woods JH. Ethanol effects in pigeons trained to discriminate MK-801, PCP or CGS-19755. Behav Pharmacol 1993;4(1):57–60. 14. Butelman ER, Negus SS, Ai Y, de Costa BR, Woods JH. ê- effects of systemically admin- istered nor-binaltorphimine in a thermal antinociception assay in rhesus monkeys. J Pharmacol Exp Ther 1993;267(3):1269–1276. 15. Carey GJ, Bergman J. Enadoline discrimination in squirrel monkeys: Effects of opioid agonists and antago- nists. J Pharmacol Exp Ther 2001;297(1):215–223. 16. Chavkin C, James IF, Goldstein A. Dynorphin is a specific endogenous ligand of the ê-opioid receptor. Science 1982;215:413–415. 17. Chen TY, Goyagi T, Toung TJ, et al. Prolonged opportunity for ischemic neuroprotection with selective ê-opioid agonists in rats. Stroke 2004;35(5):1180–1185. 18. Cicero TJ, Meyer ER, Bell RD, Koch GA. Effects of morphine and on serum testosterone and lu- teinizing hormone levels and on the secondary sex organs of the male rat. Endocrinology 1976;98(2): 367–372. 19. Clark JA, Liu L, Price M, Hersh B, Edelson M, Pasternack GW. ê-Opiate receptor multiplicity: Evidence for ê ê two U50,488-sensitive 1 subtypes and a novel 3 subtype. J Pharmacol Exp Ther 1989;251:461–468.

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 209

20. Collins SL, Kunko PM, Ladenheim B, Cadet JL, Carroll FI, Izenwasser S. Chronic cocaine increases ê-opio- id receptor density: Lack of effect by selective dopamine uptake inhibitors. Synapse 2002;45(3):153–158. 21. Corbett AD, Kosterlitz HW. Bremazocine is an agonist at ê-opioid receptors and an antagonist at ì-opioid receptors in the guinea-pig myenteric plexus. Br J Pharmacol 1986;89(1):245–249. 22. Cosgrove KP, Carroll ME. Effects of bremazocine on self-administration of smoked cocaine base and orally delivered ethanol, phencyclidine, saccharin, and food in rhesus monkeys: A behavioral economic analysis. J Pharmacol Exp Ther 2002;301:993–1002. 23. Di Chiara G, Imperato A. Opposite effects of ì- and ê-opiate agonists on dopamine release in the nucleus accumbens and in the dorsal caudate of freely moving rats. J Pharmacol Exp Ther 1988;244(3):1067–1080. 24. Dixon AK, Hill RC, Roemer D, Scholtysik G. Pharmacological properties of 4(1-methyl-4-piperidylidine)- 9,10 dihydro-4H-benzo-[4,5]cyclohepta[1,2]-thiophene hydrogen maleate (pizotifen). Arzneimittelfor- schung 1977;27(10):1968–1979. 25. Domino EF, Chodoff P, Corssen G. Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin Pharmacol Ther 1965;40:279–291. 26. Dortch-Carnes J, Potter DE. Effect of bremazocine, a ê-opioid receptor agonist, on inositol phosphate for- mation in isolated iris-ciliary bodies. Pharmacology 2002;66:100–106. 27. Drago F, Gorgone G, Spina F, et al. Opiate receptors in the rabbit iris. Naunyn Schmiedeberg’s Arch Pharmacol 1980;315:1–4. 28. Drago F, Panissidi G, Bellomio F, Dal Bello A, Aguglia E, Gorgone G. Effects of opiates and opioids on intraocular pressure of rabbits and humans. Clin Exp Pharmacol Physiol 1985;12:107–113. 29. Duke MA, Meier TL, Bolanos CA, Crawford CA, McDougall SA. Paradoxical effects of ê-opioid stimu- lation on the locomotor activity and Fos immunoreactivity of the preweanling rat: role of dopamine re- ceptors. Behav Neurosci 1997;111(5):1114–1122. 30. Dunwiddie TV, Johnson KJ, Proctor WR. Bremazocine differentially antagonizes responses to selective ì- and ä-opioid receptor agonists in rat hippocampus. Br J Pharmacol 1987;91:523–530. 31. Dykstra LA, Gmerek DE, Winger G, Woods JH. ê-Opioids in rhesus monkeys. II. Analysis of the antago- nistic actions of and â-funaltrexamine. J Pharmacol Exp Ther 1987;242:421–427. 32. El-Sharkawy TY, Al-Shireida MF, Pilcher CW. Vascular effects of some opioid receptor agonists. Can J Physiol Pharmacol 1991;69:846–851. 33. Ensinger H, Hedler L, Schurr C, Starke K. Ethylketocyclazocine decreases noradrenaline release and blood pressure in the rabbit at a peripheral opioid receptor. Naunyn Schmiedeberg’s Arch Pharmacol 1984;328: 20–23. 34. Ensinger H, Hedler L, Szabo B, Starke K. Bremazocine causes sympatho-inhibition and hypotension in rabbits by activation peripheral ê-receptors. J Cardiovasc Pharmacol 1986;8:470–475. 35. Evans CJ, Keith DE, Morrison H, Magendzo K, Edwards RH. Cloning of a ä-opioid receptor by functional expression. Science 1992;258:1952–1955. 36. Fanciullacci M, Boccuni M, Pietrini U, Sicuteri F. Search for opiate receptors in human pupil. Int J Clin Pharmacol Res 1980;1:109–113. 37. Feldberg W, Wei E. The central origin and mechanism of cardiovascular effects of morphine as revealed by naloxone in cats. J Physiol 1977;272(1):99P–100P. 38. Freye E, Harting E, Schenk GK. An opiate that induces sedation and analgesia without respiratory de- pression. Anesth Analg 1983;62:483–488. 39. Furst S. Pharmacological interaction of opiates with various classes of centrally acting dopaminergic drugs. Drug Metab Drug Interact 1991;9(2):77–102. 40. Gilbeau PM, Hosobuchi Y, Lee NM. Dynorphin effects on plasma concentrations of anterior pituitary hor- mones in the nonhuman primate. J Pharmacol Exp Ther 1986;238:974–977. 41. Gilbert PE, Martin WR. The effects of morphine and -like drugs in the nondependent morphine- dependent and cyclazocine-dependent chronic spinal dog. J Pharmacol Exp Ther 1976;198:66–82. 42. Gillan MGC, Kosterlitz HW, Magnan J. Unexpected antagonism in the rat vas deferens by benzomorphans which are agonists in other pharmacological tests. Br J Pharmacol 1981;72:13–15. 43. Greiner E, Folk JE, Jacobson AE, Rice KC. A novel and facile preparation of bremazocine enantiomers through optically pure N-norbremazocines. Bioorg Med Chem 2004;12:233–238. 44. Haertzen CA. Subjective effects of antagonists cyclazocine and nalorphine on the addiction re- search center inventory (ARCI). Psychopharmacologia 1970;18(4):366–377. 45. Hanner M, Moebius FF, Flandorfer A, et al. Purification, molecular cloning, and expression of the mam- malian sigma1-binding site. Proc Natl Acad Sci USA 1996;93(15):8072–8077. 46. Harris JA, Chang PC, Drake CT. ê-Opioid receptors in rat spinal cord: Sex-linked distribution differences. Neuroscience 2004;124(4):879–890. 47. Hayes A, Kelly A. Profile of activity of ê-receptor agonists in the rabbit vas deferens. Eur J Pharmacol 1985;110(3):317–322.

CNS Drug Reviews, Vol. 11, No. 2, 2005 210 J. DORTCH-CARNES AND D. E. POTTER

48. Hayes AG, Tyers MB. Determination of receptors that mediate opiate side effects in the mouse. Br J Phar- macol 1983;79:731–736. 49. Heijna MH, Hogenboom F, Portochese PS, Mulder AH, Schoffelmeer AN. ì- and ä-opioid receptor-me- diated inhibition of adenylate cyclase activity stimulated by release of endogenous dopamine in rat neostria- tal slices: Demonstration of potent ä-agonist activity of bremazocine. J Pharmacol Exp Ther 1989;249(2): 864–868. 50. Helmstetter FJ, Calcagnetti DJ, Cramer CP, Fanselow MS. Ethylketocyclazocine and bremazocine analgesia in neonatal rats. Pharmacol Biochem Behav 1988;30(4):817–821. 51. Hertz. A. Endogenous opioid systems and alcohol addiction. Psychopharmacologia 1997;129:99–111. 52. Horan PJ, de Costa BR, Rice KC, Porreca F. Differential antagonism of U69,593- and bremazocine-induced antinociception by(–)-UPHIT: Evidence of ê-opioid receptor multiplicity in mice. J Pharmacol Exp Ther 1991;257:1154–1161. 53. Horan P, Taylor J, Yamamura HI, Porreca F. Extremely long-lasting antagonistic actions of nor-binaltorphi- mine (nor-BNI) in the mouse tail-flick test. J Pharmacol Exp Ther 1992;260(3):1237–1243. 54. Huges NR, McKnight AT, Woodruff GN, Hill MP, Crossman AR, Brotchie JM. ê-Opioid receptor agonists increase locomotor activity in the monoamine-depleted rat model of parkinsonism. Mov Disord 1998;13: 228–233. 55. Huidobro-Toro JP, Parada S. Pharmacological evidence for and endogenous role of ê-opiate receptor in fluid and electrolyte balance. Eur J Pharmacol 1984;107:1–9. 56. Hutchinson M, Kosterlitz HW, Leslie FM, Waterfield AA. Assessment in the guinea-pig ileum and mouse vas deferens of benzomorphans which have strong antinociceptive activity but do not substitute for mor- phine in the dependent monkey. Br J Pharmacol 1975;55(4):541–546. 57. Jackson A, Cooper SJ. Observational analysis of the effects of ê-opioid agonists on open field behavior in the rat. Psychopharmacologia 1988;94:248–253. 58. Jaffe JH, Martin WR. Opioid analgesics and antagonists. In: Gilman AG, Goodman LS, Gilman A, Eds. The pharmacological basis of therapeutics. New York: Macmillan, 1980;494–534. 59. James IF, Goldstein A. Site-directed alkylation of multiple opioid receptors. I. Binding selectivity. Mol Pharmacol 1984;25:337–342. 60. Kapusta DR, Obih JC. Central ê-opioid receptor-evoked changes in renal function in conscious rats: Partici- pation of renal nerves. J Pharmacol Exp Ther 1993;267:197–204. 61. Keats AS. The effects of drugs on respiration in man. Annu Rev Pharmacol Toxicol 1985;25:41–65. 62. Ko MC, Butelman ER, Woods JH. Activation of peripheral ê-opioid receptors inhibits -induced thermal nociception in rhesus monkeys. J Pharmacol Exp Ther 1999; 289(1):378–385. 63. Ko MC, Johnson MD, Butelman ER, Willmont KJ, Mosberg HI, Woods JH. Intracisternal nor-binaltorphi- mine distinguishes central and peripheral ê-opioid antinociception in rhesus monkeys. J Pharmacol Exp Ther 1999;291(3):1113–1120. 64. Kramer HJ, Uhl W, Ladstetter B, Becker A. Influence of asimadoline, a new ê-opioid receptor agonist, on tubular water absorption and vasopressin secretion in man. Br J Clin Pharmacol 2000;50:227–235. 65. Kumor KM, Haertzen CA, Johnson RE, Kocher T, Jasinski D. Human psychopharmacology of ketocyclazo- cine as compared with cyclazocine, morphine and placebo. J Pharmacol Exp Ther 1986;238(3):960–968. 66. Kuzman A, Sandin J, Terenius L, Ogren S. Dose- and time-dependent bimodal effects of ê-opioid agonists on locomotor activity in mice. J Pharmacol Exp Ther 2000;295:1031–1042. 67. Laubie M, Schmitt H, Vincent M, Remond G. Central cardiovascular effects of morphinomimetic peptides in dogs. Eur J Pharmacol 1977;46(1):67–71. 68. Leander JD. A ê-opioid effect: Increased urination in the rat. J Pharmacol Exp Ther 1983;224:89–94. 69. Leander JD. Further study of ê-opioids on increased urination. J Pharmacol Exp Ther 1983;227:35–41. 70. Lokensgard JR, Gekker G, Peterson PK. ê-Opioid receptor agonist inhibition of HIV-1 envelope glycoprote- in-mediated membrane fusion and CXCR4 expression on CD4(+) lymphocytes. Biochem Pharmacol 2002; 63(6):1037–1041. 71. Lord JAH, Waterfield AA, Hughes J, Kosterlitz HW. Endogenous opioid peptides, multiple agonists and re- ceptors. Nature (Lond) 1977;267:495–497. 72. Magnan J, Paterson SJ, Tavani A, Kosterlitz HW. The binding spectrum of narcotic analgesic drugs with dif- ferent agonist and antagonist properties. Naunyn Schmiedeberg’s Arch Pharmacol 1982;319:197–205. 73. Mansour A, Fox CA, Akil H, Watson SJ. Opioid-receptor mRNA expression in the rat CNS: Anatomical and functional implications. Trends Neurosci 1995;18:22–29. 74. Marko M, Romer D. Inhibitory effect of a new opioid agonist on reproductive endocrine activity in rats of both sexes. Life Sci 1983;33:233–240. 75. Martin WR, Eades CG, Thompson JA, Huppler RE, Gilbert PE. The effects of morphine-and nalorphine-like drugs in the nondependent and morphine-dependent chronic spinal dog. J Pharmacol Exp Ther 1976;197: 517–532.

CNS Drug Reviews, Vol. 11, No. 2, 2005 BREMAZOCINE 211

76. McKnight AT, Corbett AD, Marcolo M, Kosterlitz HW. The opioid receptors in the hamster vas deferens are of the ä-type. Neuropharmacology 1985;24:1011–1017. 77. McKnight AT, Paterson SJ, Corbett AD, Kosterlitz HW. Acute and persistent effects of beta-funaltrexamine on the binding and agonist potencies of opioids in the myenteric plexus of the guinea-pig ileum. Neuropep- tides 1984;5(1–3):169–172. 78. Mello NK, Negus SS. Effects of ê-opioid agonists on cocaine- and food-maintained responding by rhesus monkeys. J Pharmacol Exp Ther 1998;286:812–824. 79. Merz H, Stockhaus K, Wick H. Stereoisomeric 5,9-dimethyl-2¢-hydroxy-2-tetrahydrofurfuryl-6,7-benzo- morphans, strong analgesics with non-morphine-like action profiles. J Med Chem 1975;18(10):996–1000. 80. Merz H, Stockhaus K. N-[(Tetrahydrofuryl)alkyl] and N-(alkoxyalkyl) derivatives of (–)- normetazocine, compoumds with differentiated opioid action profiles. J Med Chem 1979;22(12):1475–1483. 81. Mori T, Nomura M, Nagase H, Narita M, Suzuki T. Effects of a newly synthesized ê-opioid receptor agonist, TRK-820, on the discriminative stimulus and rewarding effects of cocaine in rats. Psychopharma- cology (Berl) 2002;161(1):17–22. 82. Negus SS, Picker MJ, Dykstra LA. Interactions between the discriminative stimulus effects of ì- and ê-opioid agonists in the squirrel monkey. J Pharmacol Exp Ther 1991;256(1):149–158. 83. Nestby P, Schoffelmeer ANM, Homberg JR, et al. Bremazocine reduces unrestricted free-choice ethanol self-administration in rats without affecting sucrose preference. Psychopharmacologia 1999;142:309–317. 84. Oiso Y, Iwasaki Y, Kondo K, Takatsuki K, Tomita A. Effect of the opioid ê-receptor agonist U50488H on the secretion of arginine vasopressin: Study on the mechanism of U50488H-induced diuresis. Neuroendo- crinology 1988;48:658–662. 85. Pang CN, Zimmermann E, Sawyer CH. Morphine inhibition of the preovulatory surges of plasma luteiniz- ing hormone and follicle stimulating hormone in the rat. Endocrinology 1977;101(6):1726–1732. ê 86. Paul D, Pick CG, Tive LA, Pasternak GW. Pharmacological characterization of nalorphine a 3 analgesic. J Pharmacol Exp Ther 1991;257:1–7. 87. Pazos A, Tristan C, Florez J. A comparative study of the respiratory depressant and analgesic effects of bre- mazocine, a ê-agonist. Life Sci 1983;33:579–581. 88. Petrillo P, Gambino MC Tavani A. Bremazocine induces antinociception, but prevents opioid-induced con- stipation and catatonia in rats and precipitates withdrawal in morphine-dependent rats. Life Sci 1984;35: 917–927. 89. Pfeiffer A, Braun S Mann K, Meyer HD, Brantl V. Anterior pituitary responses to a ê-opioid agonist in man. J Clin Endocrinol Metab 1986;62:181–185. 90. Pfeiffer A, Pasi A, Mehraien P, Herz A. Opiate receptor binding sites in human brain. Brain Res 1982;248: 87–96. 91. Picker MJ, Dykstra LA. Discriminative stimulus effects of ì- and ê-opioids in the pigeon: Analysis of the effects of full and partial ì- and ê-agonists. J Pharmacol Exp Ther 1989;249(2):557–566. 92. Picker MJ, Doty P, Stevens S, Mattox SR, Dykstra LA. Discriminative stimulus properties of U50488 and morphine: Effects of training dose on stimulus substitution patterns produced by ì- and ê-opioid agonists. J Pharmacol Exp Ther 1990;254(1):13–22. 93. Potter DE, Russell KRM, Manhiani M. Bremazocine increases C-type natriuretic peptide levels in aqueous humor and enhances outflow facility. J Pharmacol Exp Ther 2004;309:548–553. 94. Powell KR, Holtzman SG. Differential antagonism of the rate-decreasing effects of ê-opioid receptor agonists by and norbinaltorphimine. Eur J Pharmacol 1999;377:21–28. 95. Pugsley MK. The diverse molecular mechanisms responsible for the actions of opioids on the cardiovas- cular system. Pharmacol Ther 2002;93(1):51–75. 96. Pugsley MK. The opioid receptor independent actions of ê-receptor agonists in the cardiovascular system. Curr Pharm Des 2004;10(20):2429–2444. 97. Quirion R, Bowen WD, Itzhak Y, et al. A proposal for the classification of ó-binding sites. Trends Pharma- col Sci 1992;13:85–86. 98. Richards ML, Sadee E. In vivo binding of benzomorphans to ì-, ä-, and ê-opioid receptors: Comparison with urine output in the rat. J Pharmacol Exp Ther 1985;233:425–432. 99. Romer D, Buscher H, Hill R, et al. Bremazocine: A potent, long-acting opiate ê-agonist. Life Sci 1980;27: 971–978. 100. Rusovici DE, Negus SS, Mello NK, Bidlack JM. ê-Opioid receptors are differentially labeled by arylacetamides and benzomorphans. Eur J Pharmacol 2004;485(1–3):119–1125. 101. Russell KRM, Potter DE. Biphasic alterations of cAMP levels and inhibition of norepinephrine release in iris-ciliary body by bremazocine. J Pharmacol Exp Ther 2001;298:941–946. 102. Russell KRM, Wang DR, Potter DE. Modulation of ocular hydrodynamics and iris function by bremazo- cine, a ê-opioid receptor agonist. Exp Eye Res 2000;70:675–682.

CNS Drug Reviews, Vol. 11, No. 2, 2005 212 J. DORTCH-CARNES AND D. E. POTTER

103. Salas SP, Roblero J, Ureta H, Huidobro-Toro JP. Diuretic effect of bremazocine, a ê-opioid with central and peripheral sites of action. J Pharmacol Exp Ther 1989;250(3):992–999. 104. Schaz K, Stock G, Simon W, et al. Enkephalin effects on blood pressure, heart rate, and baroreceptor reflex. Hypertension 1980;2(4):395–407. 105. Schnur P, Walker JM. Effects of U50488H on locomotor activity in the hamster. Pharmacol Biochem Behav 1990;36:813–816. 106. Shearman, GT, Herz A. Discriminative stimulus properties of bremazocine in the rat. Neuropharmacology 1981;20(12A):1209–1213. 107. Simonin F, Gaveriaux-Ruff C, Befort K, et al. ê-Opioid receptor in humans: cDNA and genomic cloning, chromosomal assignment, functional expression, pharmacology, and expression pattern in the central nervous system. Proc Natl Acad Sci USA 1995;92(15):7006–7010. ì ê 35 ¢ ã 108. Sim-Selley LJ, Daunais JB, Porrino LJ, Childers SR. and 1 opioid-stimulated, [ S]guanylyl-5 -O-( - thio)-triphosphate binding in cynomolgus monkey brain. Neuroscience 1999;94:651–662. 109. Slizgi GR, Ludens JH. Studies on the nature and mechanism of the diuretic activity of the opioid analgesic ethylketocyclazocine. J Pharmacol Exp Ther 1982;220:585–591. 110. Stevens CW. Opioid research in amphibians: An alternative pain model yielding insights on the evolution of opioid receptors. Brain Res Rev 2004;46(2):204–215. 111. Tegeder I, Geisslinger G. Opioids as modulators of cell death and survival-unraveling mechanisms and re- vealing new indications. Pharmacol Rev 2004;56(3):351–369. 112. Tepper P, Woods JH. Changes in locomotor activity and naloxone-induced jumping in mice produced by WIN 35 197-2 () and morphine. Psychopharmacology (Berl) 1978;58:125–129. 113. Tiberi M, Magnan J. Pharmacological characterization of the binding of, [3H]bremazocine in guinea-pig brain: Evidence for multiplicity of the ê-opioid receptors. Can J Physiol Pharmacol 1989;67:1336–1344. 114. Upton N, Sewell RDE, Spencer PSJ. Differentiation of potent ì- and ê-opiate agonists using heat and pressure antinociceptive profiles and combined potency analysis. Eur J Pharmacol 1982;78(4):421–429. 115. Upton N, Sewell RD, Spencer PS. Analgesic actions of ì- and ê-opiate agonists in rats. Arch Int Pharmaco- dyn Ther 1983;262(2):199–207. 116. Ur E, Wright DM, Bouloux MG, Grossman A. The effects of (U62066E), a ê-opioid receptor agonist, on neuronendocrine function in man. Br J Pharmacol 1997;120:781–784. 117. Von Voightlander PF, Lahti RA, Ludens JH. U-50,488H: A selective and structurally novel non-ì, ê-opioid agonist. J Pharmacol Exp Ther 1983;224(1):7–12. 118. Walker JS. Anti-inflammatory effects of opioids. Adv Exp Med Biol 2003;521:148–160. 119. Walsh SL, Strain EC, Abreu ME, Bigelow GE. Pharmacodynamic profile of enadoline, a selective ê-ag- onist, in humans. 60th Annual Meeting of College on Problems of Drug Dependence. June 13–18, 1998. Scottsdale, AZ. 120. Weinreb RN, Khaw PT. Primary open-angle glaucoma. Lancet 2004;363:1711–1720. 121. Yamada T, Nakao K, Itoh H, et al. Effects of naloxone on vasopressin secretion in conscious rats: Evidence for inhibitory role of endogenous opioid peptides in vasopressin secretion. Endocrinology 1989;125(2): 785–790. 122. Yasuda K, Raynor K, Kong H, et al. Cloning and functional comparison of ê and ä opioid receptors from mouse brain. Proc Natl Acad Sci USA 1993;90:6736–6740. 123. Yuan L, Neufeld AH. Tumor necrosis factor: A potentially neurodestructive cytokine produced by glia in the human glaucomatous optic nerve head. Glia 2000;32:42–50. 124. Yukimura T, Stock G, Stumpf H, Unger T, Ganten D. Effects of, [D-Ala2]-methionine-enkephalin on blood pressure, heart rate, and baroreceptor reflex sensitivity in conscious cats. Hypertension 1981;3(5):528–533. 125. Zernig G, Burke T, Lewis JW, Woods JH. Mechanism of blockade of opioid receptors: Evi- dence from in vitro and ex vivo binding and behavioral assay. J Pharmacol Exp Ther 1996;279(1):23–31. 126. Zhou J-J, Pei J-M, Wang G-Y, et al. Inducible HSP70 mediates delayed cardioprotection via U-50488H pretreatment in rat ventricular myocytes. Am J Physiol Heart Circ Physiol 2001;281:H40–H47. 127. Zukin RS, Eghbali M, Olive D, Unterwald EM, Tempel A. Characterization and visualization of rat and ê ê ê guinea pig brain -opioid receptors: Evidence for 1 and 2 opioid receptors. Proc Natl Acad Sci USA 1988; 85:4061–4065. 128. Zukin RS, Zukin SR. Demonstration of, [3H]cyclazocine binding to multiple opiate receptor sites. Mol Pharmacol 1981;20(2):246–254.

CNS Drug Reviews, Vol. 11, No. 2, 2005