Ultra-Low-Dose Opioid Antagonists Enhance Opioid Analgesia and Reduce Tolerance

Share Embed


Descripción

Chapter 1

Ultra-Low-Dose Opioid Antagonists Enhance Opioid Analgesia and Reduce Tolerance Lindsay H. Burns, Todd W. Vanderah, and Hoau-Yan Wang

Abstract Ultra-low-dose opioid antagonists have been shown to enhance opioid analgesia and attenuate the tolerance to analgesic effects normally seen with chronic opioid administration. This chapter reviews the early work with ultralow-dose opioid antagonists starting with electrophysiological recordings of dorsal root ganglion neurons and continuing to antinociception in rodents. These pharmacological findings have not adhered to typical dose response curves and have instead been reported to occur at wide ranges of extremely low doses of several opioid antagonists as well as with the rare opioid agonist. Optimal dose ranges have also been reported to vary with sex and strain of rat. Translation into small clinical studies has been met with varied results, related to variations in dose, route of administration, and antagonist selected. Nevertheless, the clinical studies that have demonstrated enhanced analgesia or opioid sparing effects have utilized opioid antagonist doses in lower dose ranges than the studies that failed to demonstrate efficacy. Furthermore, a large double-blind, placebo- and active-controlled clinical trial demonstrated enhanced opioid analgesia with the extremely low dose of 2 μg naltrexone/patient/day. Preclinical data also extend the effects of ultra-low-dose opioid antagonists to neuropathic pain, which is comparatively resistant to opioid treatment and, interestingly, to cannabinoid analgesia. The mechanism of action has been shown to be the prevention of a chronic opioid-induced mu opioid receptor–G protein coupling switch that is associated with analgesic tolerance and dependence. Finally, recent data shows that this G protein coupling switch is controlled by filamin A and that a high-affinity interaction of naloxone or naltrexone with this scaffolding protein mediates their prevention of the altered coupling.

Keywords: Naloxone; Naltrexone; Neuropathic pain; G protein coupling; Filamin A

L.H. Burns (), T.W. Vanderah, and H.-Y. Wang Pain Therapeutics, Inc., Preclinical Development, 2211 Bridgepointe Parkway, Suite 500, San Mateo, CA 94404 e-mail: [email protected] R. Dean et al. (eds.), Opiate Receptors and Antagonists, © Humana Press, a part of Springer Science + Business Media, LLC 2009

3

4

1.1

L.H. Burns et al.

Introduction

Opioid analgesics are widely used to manage moderate or severe pain, yet their use is hampered by unwanted side effects, tolerance to their analgesic effects, and physical dependence. The fact that patients often balance analgesia with side effects illustrates the narrow therapeutic window of opioids. Side effects range from nausea, constipation, somnolence, and pruritis to the more serious respiratory depression. Chronic pain patients need analgesia over prolonged or indefinite time periods, yet these patients very often experience a loss of analgesic potency with continued use. Finally, physicians and patients alike are wary of initiating opioid therapy due to the fears or stigma around opioid dependence and even the potential for addiction. As a consequence, the field of pain management is in great need of improved analgesic therapies that provide adequate analgesia for moderate-to-severe pain with fewer adverse effects and minimal potential for tolerance, dependence, and addiction. One such therapy involves the combination of opioid agonists with “ultra-lowdose” opioid antagonists. In contrast to the typical blockade of opioid receptor functions by higher concentrations of opioid antagonists, ultra-low-dose opioid antagonist cotreatment enhances and prolongs opioid analgesia and also prevents or reverses opioid analgesic tolerance. These phenomena have been demonstrated by preclinical data and have now randomized double-blind, controlled clinical trial data. This chapter will review the effects of ultra-low-dose opioid antagonists on analgesia, tolerance, and the hyperalgesia and allodynia in a model of neuropathic pain, while the attenuation of physical dependence and the addictive properties of opioids will be reviewed in Chapater 13. This chapter will also provide a historical overview of studies probing the mechanism of action for these effects from the initial electrophysiology findings to more recent molecular pharmacology data and identification of the high-affinity binding site. These studies indicate that chronic opioid administration causes alterations in the G protein system associated with mu opioid receptors (MORs) and that ultra-low-dose naloxone (and perhaps other opioid antagonists) suppresses these aberrant signaling changes via a picomolar interaction with filamin A.

1.2

Preclinical Evidence of Enhanced Opioid Analgesia and Reduced Tolerance

The history of ultra-low-dose opioid antagonists starts with electrophysiology studies on dorsal root ganglion (DRG) cell cultures. Crain and Shen first observed that while opioid agonists normally inhibit, or shorten, the action potential duration of DRG cells, lower doses of opioid agonists induce an opposite, excitatory effect – a prolongation of the action potential (39, 9). These researchers noted that prolonged exposure to an opioid agonist would also produce this prolongation of action potentials, and they theorized that these excitatory effects of opioids contributed to opioid tolerance (10). This same publication proposed a regula-

1

Low-Dose Naltrexone Improves Opioid Analgesia

5

tion by GM1 ganglioside, since exogenous administration of GM1 ganglioside mimicked the prolonged opioid exposure. The hypothesis that excitatory effects of opioids are mediated by opioid receptors coupling to the excitatory G protein Gs rather than the usual inhibitory G proteins, Gi or Go, was suggested by a blockade of the action potential prolongation by administration of cholera toxin-A, an agent that blocks Gs from being activated by receptor stimulation (40). Using their in vitro model of action potential prolongation by low concentrations of morphine, Crain and Shen (11) showed that ultra-low-dose naloxone blocked the action potential prolongation when added at low picomolar concentrations. In the same 1995 publication, Crain and Shen showed that ultra-low-dose naltrexone (10 ng/kg, s.c.) also enhanced and prolonged morphine’s antinociceptive potency in mice. This seminal report also included data demonstrating that cotreatment with ultra-low-dose naltrexone dramatically attenuated morphine withdrawal after an acute high dose of morphine or after a 4-day, twice daily, progressively increasing morphine dosing schedule. Finally, an assessment of tolerance was made in the mice receiving the escalating 4-day morphine treatment, by comparing subsequent antinociception to that of naïve mice. Tolerance was partially prevented by ultra-low-dose naltrexone. A subsequent study by a different lab extended the work of Crain and Shen, demonstrating enhanced morphine antinociception by ultra-low-dose naltrexone administered either systemically or intrathecally to rats (36). This study also demonstrated that coadministration of ultra-low-dose naltrexone (0.05 ng) prevents analgesic tolerance over 7 days of daily intrathecal administration of morphine (15 μg). An even lower intrathecal dose of naltrexone (0.005 ng) as well as i.p. administration of both drugs was less effective. Using rats instead of mice, these data demonstrated enhanced morphine analgesia, a complete prevention of tolerance over a 7-day treatment period, a lower end of an effective ultra-low-dose range by intrathecal administration, and a spinal site of action. Data also showed that established morphine tolerance could be reversed, at least partially. Crain and Shen also followed up their earlier work by demonstrating that both analgesic tolerance and precipitated withdrawal-associated hyperalgesia in mice could be prevented by ultra-low-dose naltrexone combined with either morphine (45) or oxycodone (44). In a further extension of their in vitro data, Crain and Shen demonstrated that acute hyperalgesia elicited by a low dose of an opioid agonist, referred to clinically as “paradoxical hyperalgesia” (27,29), could be reversed by ultra-low-dose naltrexone coadministration, yielding a strong analgesic effect (15). While the early in vitro work primarily utilized naloxone, ultra-low doses of the kappa opioid antagonist nor-binaltorphamine (nor-BNI) and the mu opioid antagonist beta-funaltrexamine (β-FNA) (Crain and Shen, personal communication), as well as the nonselective opioid antagonist diprenorphine (41), have also enhanced the inhibitory effects of opioid agonists in vitro. While most published in vivo work was initially limited to naltrexone, ultra-low-dose naloxone was also recently used in vivo to prevent analgesic tolerance (53; Fig. 1.1). In addition, ultra-low doses of naloxone and nalmefene have demonstrated beneficial effects in clinical studies discussed below. Perhaps more surprisingly, ultra-low doses of the opioid agonists

6

L.H. Burns et al.

Morphine + NLX Morphine (10 mg/kg) NLX (10 ng/kg)

Tail-Flick Latency (s)

10

*

*

*

Day 3

Day 5

Day 7

8 6 4 2 0 BLINE

Day 1

Fig. 1.1 Co-treatment with ultra-low-dose NLX (10 ng/kg) prevented the antinociceptive tolerance caused by chronic morphine (10 mg/kg, s.c., twice daily for 7 days). Rats treated with morphine + NLX showed stable tail-flick latencies over the week of treatment, while tail-flick latencies of rats receiving morphine alone declined to a level not significantly different from NLX alone. *p < 0.05 and **p < 0.01 for morphine + NLX versus morphine. BLINE predrug baseline, NLX naloxone. Reprinted from Wang et al. (53), with permission from Elsevier

etorphine and dihydroetorphine (41) and of [des-Tyr] fragments of the endogenous opioids dynorphin A-(1–13) and β-endorphin (1–27) (42) also enhance the inhibitory effects of opioid agonists in vitro. Further, ultra-low-dose etorphine enhances morphine’s potency in vivo (43). Together, these findings hinted that these phenomena are not unique to naloxone or naltrexone and are also not dependent on opioid antagonist activity.

1.3

Dose Effects and Dependency on Strain and Sex

Ultra-low-dose opioid antagonist effects do not follow a typical dose-response pattern, but typically span several log units without conforming to bell-shaped curves. A wide range of doses has been used successfully. Although some initial studies tested antagonist:agonist ratios rather that discrete antagonist doses, the much narrower effective dose range for agonists than for ultra-low-dose antagonists in enhancing agonist effects suggests that the antagonist dose is independent of agonist dose. This method was likely initiated for ease of calculating antagonist dose. An examination of the antinociceptive effects of various ratios of naltrexone:oxycodone (1:109, 1:107, and 1:105) combined with a range of oxycodone doses (0.03–3 mg/kg) in male Swiss Webster mice illustrates the wide range of effective doses of naltrexone (Fig. 1.2). Whereas all naltrexone:oxycodone dose ratios examined enhanced the areas under the curve (AUCs) of tailflick latencies

1

Low-Dose Naltrexone Improves Opioid Analgesia

7

Oxycodone

0.03 0.10 0.30 1.00 3.00

0.03 0.10 0.30 1.00 3.00

0.03 0.10 0.30 1.00 3.00

65 60 55 50 45 40 35 30 25 20 15 10 5 0

Nalrexone:Oxycodone 1:10E7 Naltrexone:Oxycodone 1:10E5

0.03 0.10 0.30 1.00 3.00

AUC Tail Flick Latency (hr*sec)

Naltrexone:Oxycodone 1:10E9

Oxycodone Doses (mg/kg) Fig. 1.2 Dose-response of ultra-low-dose naltrexone:oxycodone ratios in enhancing oxycodone antinociception in the 52°C hot water immersion tail-flick tests in male Swiss Webster mice. Three different naltrexone:oxycodone ratios (1:109, 1:107, and 1:105) were superimposed onto a dose-response of oxycodone (0.03, 0.1, 0.3, 1, and 3 mg/kg, s.c.). These naltrexone:oxycodone ratios enhanced the effects of all doses of oxycodone, although the 1:109 and 1:107 ratios more potently enhanced the antinociceptive effect of the 3 mg/kg oxycodone dose than did the 1:105 dose ratio. Data are means ± SEM., n = 3. Reprinted from (2) in Recent Developments in Pain Research, with permission from Research Signpost

of the oxycodone doses, the strongest antinociceptive effects were obtained with the 3 and 300 pg/kg naltrexone doses, which appear to enhance the antinociceptive effects of 3 mg/kg oxycodone to a greater extent than a 30 ng/kg dose. While wide antagonist dose ranges have been used effectively, the optimal doses of antagonist vary with both the opioid agonist it is combined with as well as the sex of the animal. The prevention of tolerance and withdrawal-associated hyperalgesia by ultra-low-dose naltrexone was demonstrated at 0.3, 3, 300, and 3,000 ng/kg naltrexone when combined with 3 mg/kg morphine (45) and at 1 pg/kg or 1 ng/kg naltrexone when combined with oxycodone (44). The higher naltrexone doses used in these studies were used in males, suggesting males are less sensitive to ultra-low-dose antagonist effects. Interestingly, although these studies used the same morphine dose for males and females, a tenfold lower dose of oxycodone was administered to males, confirming the previously described increased sensitivity to opioid analgesic effects in male rodents (28). Hence, while male rodents may be more sensitive to opioid analgesia, female rodents appear to be more sensitive to ultra-low-dose naltrexone, suggesting some degree of dissociation between the two. In addition, low doses of oxycodone elicited a greater degree of hyperalgesia in male than in female rats (23), while the reverse was reported with morphineinduced hyperalgesia (22). These findings likely contribute to the variations in

8

L.H. Burns et al.

optimal ultra-low-dose opioid antagonist dose ranges noted both with sex of the animal and the opioid used in combination. In addition to sex differences, the effects of ultra-low-dose naltrexone on morphine antinociception and tolerance can also vary with rat strain (46). While the enhancement of morphine antinociception and attenuation of morphine tolerance was observed in both Sprague–Dawley and Long–Evans rats, naltrexone at the dose range tested (0.1–100 ng/kg) did not enhance morphine antinociception over a range of doses (2.5–7.5 mg/kg) in Fisher 344 or Lewis rats, nor did 10 or 100 ng/kg naltrexone significantly attenuate tolerance in these two strains. Given that pg/kg doses of naltrexone most effectively enhanced oxycodone antinociception in females in the previously mentioned Shen study (44), the question remains whether this even lower range might enhance antinociception in these two rat strains. Nevertheless, clear strain differences exist expanding on the initial report that ultra-low-dose naltrexone enhances morphine antinociception in Swiss Webster mice but antagonizes it in 129/SvEv mice (14). The authors attribute this opposite effect of ultra-low-dose naltrexone in 129/SvEv mice, along with the enhanced potency of morphine and general lack of tolerance in this strain, to its deficiency in GM1 ganglioside. The Terner study (46) also noted strain-specific sex differences. In Sprague– Dawley rats, males and females differed in the dose that most effectively enhanced antinociception, with the best effect in males observed at 10 ng/kg and the highest efficacy in females occurring at the lowest dose tested at 0.1 ng/kg. This greater sensitivity to ultra-low-dose naltrexone in female Sprague–Dawley rats, despite the females being less sensitive to the opioid itself, concurs with the findings by Shen and colleagues in Swiss Webster mice (44, 45). An additional strain-dependent sex difference was revealed in the Terner study; 10 ng/kg naltrexone significantly reversed established morphine tolerance only in female Long–Evans rats. In a separate study examining age and sex effects, morphine antinociception was enhanced by low-dose naltrexone in mature female but not in mature male rats (18–22 weeks) and was negligible in younger rats (21). However, the naltrexone dose range used in that study was again comparatively high (0.002–2 μg/kg), and the effect in mature females was “inversely related to dose.”

1.4

Ultra-Low-Dose Naltrexone Effects on Neuropathic Pain and Cannabinoid Analgesia

Although opioids are not typically very effective in neuropathic pain, recent preclinical data has shown that ultra-low-dose naltrexone enhances the antihyperalgesic and antiallodynic effects of morphine (49) or oxycodone (30) in the L5/L6 spinal nerve ligated rats. The antihyperalgesic effects of intrathecal delivery of these opioids combined with naltrexone at 0.33 ng were complete, though some tolerance developed after 10 days of twice daily administration. Further, while intrathecal morphine did not elicit any antiallodynic effect in this model, its combination with ultra-low-dose naltrexone produced a moderate though transient

1

Low-Dose Naltrexone Improves Opioid Analgesia

9

antiallodynic effect. With twice daily oral delivery in this rat model of neuropathic pain, ultra-low-dose naltrexone (0.3–3 μg/kg) combined with oxycodone produced strong antihyperalgesic and antiallodynic effects. Tolerance to these effects was dramatically reduced compared to the more rapid tolerance that developed with oral oxycodone alone. The oral 0.3 μg/kg naltrexone dose combination produced an antihyperalgesic effect that showed no tolerance over this time period. Notably, the enhanced antihypersensitivity effects of the opioids in these studies occurred at slightly higher doses of naltrexone than those shown to enhance antinociception in intact rats. Moreover, these preclinical data suggest that ultra-low-dose naltrexone may augment opioid effects in neuropathic pain and minimize the development of tolerance to them. Further extending the area of research with ultra-low-dose opioid antagonists, a recent study showed ultra-low-dose naltrexone to enhance cannabinoid-induced analgesia (35). This finding may reflect the well-established interaction of the opioid and cannabinoid systems (32), rather than a specific interaction of naltrexone’s binding site on filamin A with cannabinoid receptors. The complete abolition of the analgesic effect of the cannabinoid agonist/naltrexone combination by a cannabinoid antagonist suggests that this analgesia is dependent on the cannabinoid system with any opioid effects upstream. The fact that a high dose of naltrexone only slightly (and nonsignificantly) enhanced here the cannabinoid-induced analgesia suggests that that the potentiation of cannabinoid analgesia by ultra-low-dose naltrexone has some opioid component but does not exclusively rely on opioid receptors.

1.5

Clinical Studies of Ultra-Low-Dose Opioid Antagonist Effects in Analgesia

Clinical experience with ultra-low-dose opioid antagonists has grown from case reports and small clinical studies to blinded, randomized, controlled clinical trials. In contrast to preclinical reports that mostly use naltrexone, clinical studies have reported benefits with ultra-low-doses of naloxone, naltrexone, and also nalmefene. In addition, “ultra-low” doses, dosing schedules, and settings have varied along with results. Most small clinical studies assessed postoperative analgesia and used i.v. (intravenous) administration, while current clinical trials have examined analgesia and dependence and have used oral delivery over 3 weeks or 3 months. While no published clinical studies or clinical trials have been able to assess ultra-low-dose opioid antagonist effects on analgesic tolerance, a case report noted strong analgesia in a severely opioid tolerant diabetic neuropathy patient from ultra-low-dose naltrexone (1 μg orally, b.i.d.) combined with methadone (16). This patient reported a pain intensity reduction from 9 to 3 within 24 h that persisted through a 1-month follow-up. In a postoperative setting, the first clinical study showed that ultra-low-dose naloxone in a continuous i.v. infusion of 0.25 μg/kg/h, but not 1 μg/kg/h, produced an opioid-sparing effect of morphine delivered by patient-controlled analgesia (PCA) following hysterectomy (18). In another study in women undergoing lower

10

L.H. Burns et al.

abdominal surgery, a single dose of 15 or 25 μg nalmefene administered before morphine PCA decreased severity of pain 24 h later and also decreased the need for antiemetics and antipruritics (26). A subsequent study using morphine PCA with or without naloxone at an estimated dose of 0.57 followed by 0.19 μg/kg/h failed to demonstrate enhanced analgesia or opioid-sparing effects (4). Reducing the i.v. naloxone dose tenfold in a later study to an estimated 0.05 followed by 0.006 μg/kg/h, Cepeda again did not demonstrate opioid-sparing effects or enhanced morphine analgesia but reported decreased nausea and pruritis (5). It should be noted that in both studies, PCA doses were increased by 20% when pain intensity exceeded 4 and again at 6 on a 0–10 scale, and a rescue dose 2.5 times the PCA solution was administered when pain intensity reached 6, so accurate assessment of doses is difficult. However, the PCA solution contained 6 μg/cc in the first study and 0.6 μg/cc in the second. A small randomized, double-blind clinical trial of 156 patients presenting to the emergency department with acute, severe pain did not demonstrate any greater analgesic efficacy of a single i.v. bolus of morphine (0.1 mg/kg) combined with one of three different doses of naloxone (0.1, 0.01, or 0.001 ng/kg) compared to the morphine alone (1). In contrast to the only other study that used a single i.v. bolus of an ultra-low-dose opioid antagonist, that is nalmefene administered prior to morphine PCA postoperatively and assessed pain scores 24 h later (26), the present study gave one combined bolus and assessed pain intensity over the next 4 h. Possibly refers to confounding the results of the study were additional doses of morphine or other opioids administered within the 4 h period. While 13 patients in the morphine-only group received additional morphine, 8, 10, and 14 patients in the 0.1, 0.01, and 0.001 ng/ kg naloxone groups required additional morphine, respectively. Although, this data may suggest an opioid-sparing effect at the 0.1 ng/kg naltrexone level these differences were not significant due to the small group sizes. Besides the small group sizes in this study, the diversity of pain conditions and changing rates of pain in patients presenting to the emergency department may have hindered this trial. Nevertheless, this study was unable to demonstrate an effect of ultra-low-dose naloxone in the dose range of 0.001–0.1 ng/kg in augmenting opioid analgesia in acute, severe pain. The first large, randomized, double-blind clinical trial assessed oxycodone and ultra-low-dose naltrexone (Oxytrex™) versus oxycodone alone delivered orally in 350 osteoarthritis patients with pain ≥5 on a 0–10 scale for 3 weeks (7). Naltrexone was formulated with oxycodone at 1 μg per tablet, so that the Oyxtrex b.i.d. dose group received 2 μg/day while the Oxytrex q.i.d. group received 4 μg/day. Both were compared to placebo and oxycodone alone administered q.i.d. in an immediate-release formulation. All active treatment groups received the same daily dose of oxycodone escalating from a starting dose of 10–40 mg/day in the final week. The Oxytrex b.i.d. treatment, with 2 μg/day naltrexone, provided significantly greater analgesia than both placebo and oxycodone by the end of week 2, with the strongest differences seen at the end of week 3. Neither oxycodone itself nor Oxytrex q.i.d., with 4 μg/day naltrexone, separated from placebo in reduction of pain intensity, a result not inconsistent with prior clinical trials of oxycodone at these doses (37, 33). Although this trial included mostly female patients, a sub-analysis by gender illustrated a stronger treatment effect in males, and a stronger separation

1

Low-Dose Naltrexone Improves Opioid Analgesia

11

Fig. 1.3 Reduction in pain intensity in males and females. Oxytrex b.i.d. provided the greatest reduction in pain intensity scores in both males and females. At week 3, Oxytrex b.i.d. was significantly better than placebo in males and significantly better than oxycodone in females. Reprinted from Chindalore et al. (7) with permission from American Pain Society

of Oxytrex b.i.d. from placebo in males and a statistical separation of Oxytrex b.i.d. only from oxycodone in females, possibly due to a greater placebo effect in females (Fig. 1.3). Moreover, this first clinical trial demonstrated both enhanced analgesia and the ability to dose b.i.d. without any extended release mechanism, illustrating the prolonged duration of effect seen preclinically. In addition, the enhanced analgesic efficacy of the treatment regimen containing 2 but not 4 μg/patient/day of naltrexone, combined with its ∼20% bioavailability by oral delivery, may shed light on earlier studies using i.v. naloxone at higher doses. A subsequent clinical trial in 750 low-back pain patients showed that Oxytrex b.i.d. provided equivalent analgesia from a significantly lower total average daily dose compared to oxycodone q.i.d. (54). In this trial, patients titrated their dose to a pain score ≤2 or to tolerable side effects up to a maximum of 80 mg/day before being fixed for 12 weeks on their individual doses. Oxytrex b.i.d. also significantly reduced the number of moderate-to-severe events of constipation, somnolence, and pruritis seen with oxycodone alone. To summarize clinical experience with ultra-low-dose opioid antagonists, some studies may have failed due to “ultra-low” doses that are too high, possibly impeding the effect of the opioid by classical receptor antagonism. In addition, differences in efficacy were observed between studies in postoperative or acute pain and chronic pain populations, possibly suggesting greater efficacy in chronic pain, though one demonstrated enhanced efficacy of morphine intra-operatively. Gender differences in sensitivity to ultra-low-dose opioid antagonists have not emerged clinically, though two positive studies were performed in females. Finally, while some positive studies noted enhanced analgesia, others noted opioid-sparing effects or decreased side effects.

12

1.6

L.H. Burns et al.

Mechanism of Ultra-Low-Dose Opioid Antagonists

The mechanism of action of ultra-low-dose opioid antagonists in enhancing and prolonging analgesia and preventing tolerance has long been hypothesized as a blockade of excitatory signaling opioid receptors. Mu opioid receptors preferentially couple to pertussis toxin-sensitive G proteins, Gi and Go, to inhibit the adenylyl cyclase/cyclic AMP (cAMP) pathway (31, 8). Since the in vitro excitatory effects of low doses of opioids (the shortening instead of prolongation of the action potential duration) could be pharmacologically blocked by cholera toxin-A, a compound that blocks activation of the “excitatory” G protein Gs by their associated receptors (40), Crain and Shen hypothesized that the excitatory response to opioids, whether from low doses or prolonged exposure, was mediated by MORs coupling to Gs (12). While Gi and Go proteins are known to inhibit adenylyl cyclase and subsequent production of cAMP from ATP, Gs stimulates this enzyme causing the opposite effects on the cell. Based on their electrophysiology work, Crain and Shen also hypothesized that opioid antagonists selectively antagonized such “excitatory opioid receptors” that signal via Gs proteins (13). Much of the controversy over this initial hypothesis arose from earlier research suggesting that observable excitatory effects of opioids were not due to a G protein coupling partner novel to MOR, that is, a switch in G protein coupling, but were simply due to altered signaling of the Gβγ dimer contained in the original Gi or Go proteins (20). The Gβγ dimer of the heterotrimeric G proteins coupling to the MOR are thought to contribute to the analgesic effects of opioids by signaling to ion channels and inhibiting cellular activities by hyperpolarization (24, 38). A Gβγ activation of adenylyl cyclase instead would counteract analgesia as would stimulation of adenylyl cyclase by a Gs protein. Molecular pharmacology work has demonstrated a decrease in MOR Gi and Go coupling concurrent with a striking appearance of Gs coupling in central nervous system (CNS) tissues of rats treated with morphine (10 mg/kg, s.c.) twice daily for 7 days, and an attenuation of these changes by cotreatment with ultra-low-dose naloxone (10 μg/kg, s.c.) (53). A morphine-induced association of MOR with Gs was also demonstrated in Chinese hamster ovary (CHO) cells and in spinal cord (6) by the group who initially demonstrated morphine-induced adenylyl cyclase activation by Gβγ. The development of Gs coupling by MORs may also contribute to the hypersensitivities to thermal and tactile stimuli noted to occur after chronic morphine administration (47, 48). Wang et al. (2005) used a previously established coimmunoprecipitation procedure (50, 25, 55, 56, 3) with antibodies to various Gα proteins to separately isolate each Gα protein along with its associated receptor proteins under basal and morphine-stimulated conditions. Subsequently, the relative amounts of MOR protein associated with each Gα protein were detected via Western blots with a selective antibody directed against an N-terminal epitope of MOR. These relative quantities demonstrated treatment-associated changes in levels of MOR coupling to Gi, Go, and Gs without discernible effects on the expression of either MORs or G proteins (Fig. 1.4). The specificity of antibodies was rigorously demonstrated, and the instatement of Gs coupling with chronic morphine treatment

Low-Dose Naltrexone Improves Opioid Analgesia

MOR immunreactivity Optical Density (Arbitrary Unit)

1

13

15000

#

*

#

*

10000

* * 5000

**

*

#

*

##

*

** *

0

G protein:

sioqsioq sioqsioq sioqsioq sioqsioq Vehicle

Morphine

Morphine

NLX

+ NLX Fig. 1.4 Densitometric quantifications of Western blots showing that chronic morphine-induced Gs–Mu opioid receptor (MOR) coupling is attenuated by cotreatment with ultra-low-dose naloxone (NLX). MOR protein is detected in immunoprecipitates of Gαi, Gαo and Gαs of spinal cord from rats treated twice daily for 7 days with saline, morphine (10 mg/kg, s.c.), morphine + NLX (10 ng/kg, s.c.) or NLX (10 ng/kg, s.c.). Data are means ± s.e.m. derived from four individual rats in each of the treatment groups that were processed individually (n = 4). Solid bars indicate basal coupling, and hatched bars indicate coupling after receptor stimulation by in vitro morphine.*p < 0.05 versus same Gα protein in vehicle and NLX-treated groups.**p < 0.05 versus same Gα protein in morphine group. Morphine-stimulated coupling was significantly greater (p < 0.01) than basal coupling of each Gα protein within each treatment group. Reprinted from Wang et al. (53), with permission from Elsevier

and its attenuation by cotreatment with ultra-low-dose naloxone were confirmed using tritiated DAMGO instead of the MOR antibody. Wang et al. (53) also examined the interaction of Gβγ with adenylyl cyclase using a similar coimmunoprecipitation procedure using CNS tissues from the same morphine, morphine and naloxone or vehicle-treated rats. Chronic morphine instated an interaction of the MOR-associated Gβγ with adenylyl cyclase types II and IV, and these Gβγ–adenylyl cyclase interactions were similarly attenuated by ultra-low-dose naloxone cotreatment. These treatment-mediated changes in association between Gβγ and adenylyl cyclase type II and IV also occurred without alterations in expression levels of these signaling molecules. We have also demonstrated that the Gβγ that interacts with adenylyl cyclase after chronic morphine treatment originates from a Gs protein that couples to the MOR as opposed to its native G protein (51). Ultra-low-dose opioid antagonists were initially thought to preferentially bind a subset of MORs (13), and a Gs-coupling MOR subpopulation was again recently proposed (6). However, since naloxone prevents MOR–Gs coupling at concentrations well below its affinity for MOR and since our coimmunoprecipitation data showed it to alter the coupling behavior of MOR, we considered proteins that

14

L.H. Burns et al.

interact with MOR and MOR-associated G proteins as the most likely targets, particularly those able to interact with multiple MORs. We recently identified a pentapeptide segment of C-terminal filamin A, a scaffolding protein known to interact with MOR (34), as the high-affinity binding site of ultra-low-dose naloxone in preventing the chronic opioid-induced MOR–Gs coupling (52). Using organotypic striatal slice cultures, we showed that peptide fragments containing the binding site on filamin A abolished the prevention by 10 pM naloxone of both the chronic morphine-induced MOR–Gs coupling and the downstream cAMP excitatory signal. A competition curve showed that naltrexone or naloxone binds filamin A with an affinity of ∼ 4pM, that is ∼200-fold higher than their affinity for MOR (17, 19). This data establishes filamin A as the target for ultra-low-dose naloxone, naltrexone in their prevention of Gs coupling by MOR, and presumably also their prevention of analgesic tolerance and dependence.

1.7

Conclusions

This chapter has provided an overview of ultra-low-dose opioid antagonist research from the initial discovery of preventing opioid excitatory effects observed by electrophysiology in vitro to preclinical findings of enhanced and prolonged analgesia and reduced analgesic tolerance in vivo. The dose–response relationship spans several log units and does not conform to a bell-shaped curve, although dramatic sex and strain effects have been noted within various dose ranges. This chapter reviewed preclinical and clinical studies conducted with a variety of doses and opioid agonist/antagonist combinations in a variety of settings. The recent identification of the high-affinity interaction binding by naloxone or naltrexone to filamin A may partially explain the varied results seen clinically with various opioid antagonists, doses, and routes of administration, on top of the different patient populations, conditions, and endpoints between studies. Further, the observed sex and strain differences could potentially reflect differences in filamin A levels. Although, a few studies have failed to demonstrate benefits of ultra-low-dose opioid antagonists, most of the available evidence, including results of the larger clinical trials, clearly indicates that certain combinations can substantially improve opioid therapy available today. The effects of ultra-low-dose naltrexone on physical dependence and addictive properties of opioids are reviewed in the Chapter 13.

References 1. Bijur PE, Schechter C, Esses D, Chang AK, Gallagher EJ (2006) Intravenous bolus of ultralow-dose naloxone added to morphine does not enhance analgesia in emergency department patients. J Pain 7:75–81. 2. Burns, LH (2005) Ultra-low-dose opioid antagonists enhance opioid analgesia while decreasing tolerance, dependence and addictive properties. In: Recent Developments in Pain Research, Anna Capasso (Ed.), pp 115–136.

1

Low-Dose Naltrexone Improves Opioid Analgesia

15

3. Cai G, Wang H-Y, Friedman E (2002) Increased dopamine receptor signaling and dopamine receptor–protein coupling in denervated striatum. J Pharmacol Exp Ther 302:1105–1112. 4. Cepeda MS, Africano JM, Manrique AM, Fragoso W, Carr DB (2002) The combination of low dose naloxone and morphine in PCA does not decrease opioid requirements in the postoperative period. Pain 96:73–79. 5. Cepeda MS, Alvarez H, Morales O, Carr DB (2004) Addition of ultra low dose naloxone to postoperative morphine PCA: unchanged analgesia and opioid requirement but decreased incidence of opioid side effects. Pain 107:41–46. 6. Chakrabarti S, Regec A, Gintzler AR (2005) Biochemical demonstration of mu-opioid receptor association with Gsα: enhancement following morphine exposure. Mol Brain Res 135:217–224. 7. Chindalore VL, Craven RA, Butera PG, Yu KP, Burns LH, Friedmann N (2005) Adding ultralow-dose naltrexone to oxycodone enhances and prolongs analgesia. J Pain 6:392–399. 8. Connor M, Christie MD (1999) Opioid receptor signalling mechanisms. Clin Exp Pharmacol Physiol 26:493–499. 9. Crain SM, Shen K-F (1990) Opioids can evoke direct receptor-mediated excitatory effects on sensory neurons. Trends Pharmacol Sci 11:77–81. 10. Crain SM, Shen K-F (1992) After chronic opioid exposure sensory neurons become supersensitive to the excitatory effects of opioid agonists and antagonists as occurs after acute elevation of GM1 ganglioside. Brain Res 575:13–24. 11. Crain SM, Shen K-F (1995) Ultra-low concentrations of naloxone selectively antagonize excitatory effects of morphine on sensory neurons, thereby increasing its antinociceptive potency and attenuating tolerance/dependence during chronic cotreatment. Proc Natl Acad Sci USA 92:10540–10544. 12. Crain SM, Shen K-F (1998) Modulation of opioid analgesia, tolerance and dependence by Gs-coupled, GM1 ganglioside-regulated opioid receptor functions. Trends Pharmacol Sci 19:358–365. 13. Crain SM, Shen K-F (2000a) Antagonists of excitatory opioid receptor functions enhance morphine’s analgesic potency and attenuate opioid tolerance/dependence liability. Pain 84: 121–131. 14. Crain SM, Shen K-F (2000b) Enhanced analgesic potency and reduced tolerance of morphine in 129/SvEv mice: evidence for a deficiency in GM1 ganglioside-regulated excitatory opioid receptor functions. Brain Res 856:227–235. 15. Crain SM, Shen K-F (2001) Acute thermal hyperalgesia elicited by low-dose morphine in normal mice is blocked by ultra-low-dose naltrexone, unmasking potent opioid analgesia. Brain Res 888:75–82. 16. Cruciani RA, Lussier D, Miller-Saultz D, Arbuck DM (2003) Ultra-low dose oral naltrexone decreases side effects and potentiates the effect of methadone. J Pain Symptom Management 25:491–494. 17. Emmerson P, Liu M, Woods J, Medzihradsky F (1994) Binding affinity and selectivity of opioids at mu, delta and kappa receptors in monkey brain membranes. J Pharmacol Exp Ther 271:1630–1637. 18. Gan TJ, Ginsberg B, Glass PSA, Fortney J, Jhaveri R, Perno R (1997) Opioid-sparing effects of a low-dose infusion of naloxone in patient-administered morphine sulfate. Anesthesiology 87:1075–1081. 19. Gharagozlou P, Demirci H, Clark J, Lameh J (2003) Activity of opioid ligands in cells expressing cloned μ opioid receptors. BMC Pharmacology 3:1471–2210. 20. Gintzler AR, Chakrabarti S (2001) Opioid tolerance and the emergence of new opioid receptorcoupled signaling. Mol Neurobiol 21:21–33. 21. Hamman SR, Malik H, Sloan JW, Wala EP (2004) Interactions of “ultra-low” doses of naltrexone and morphine in mature and young male and female rats. Recept Chan 10:73–81. 22. Holtman JR and Wala EP (2005) Characterization of morphine-induced hyperalgesia in male and female rats. Pain 114:62–70. 23. Holtman JR and Wala EP (2006) Characterization of the antinociceptive effect of oxycodone in male and female rats. Pharmacol Biochem Behav 83:100–108.

16

L.H. Burns et al.

24. Ikeda K, Kobayashi T, Kumanishi T, Niki H, Yano R (2000) Involvement of G-proteinactivated inwardly rectifying K + (GIRK) channels in opioid-induced analgesia. Neurosci Res 38:113–116. 25. Jin LQ, Wang H-Y, Friedman E (2001) Stimulated D(1) dopamine receptors couple to multiple G-alpha proteins in different brain regions. J Neurochem 78:981–990. 26. Joshi GP, Duffy L, Chehade J, Wesevich J, Gajraj N, Johnson ER (1999) Effects of prophylactic nalmefene on the incidence of morphine-related side effects in patients receiving intravenous patient-controlled analgesia. Anesthesiology 90:1007–1011. 27. Kayser V, Besson JM, Guilbaud G (1987) Paradoxical hyperalgisic effect of exceedingly low doses of systemic morphine in an animal model of persistent pain (Freund’s adjuvant-induced arthritis rats). Brain Res 414:155–157. 28. Kest B, Sarton E, Dahan A (2000) Gender differences in opioid-mediated analgesia. Anesthesiology 93:539–547. 29. Kiyatkin EA (1989) Morphine: some puzzles of a well-known substance. Int J Neurosci 45:231–246. 30. Largent-Milnes TM, Guo W, Wang H-Y, Burns LH, Vanderah T (2008) Oxycodone + ultralow-dose naltrexone attenuates neuropathic pain and associated mu opioid receptor–Gs coupling. J Pain 9:700–713. 31. Laugwitz KL, Offermanns S, Spicher K, Schultz G (1993) Mu and delta opioid receptors differentially couple to G protein subtypes in membranes of human neuroblastoma SH-SY5Y cells. Neuron 10:233–242. 32. Maldonado R, Valverde O (2003) Participation of the opioid system in cannabinoid-induced antinociception and emotional like responses. Eur Neuropsychopharmacol 13:401–410. 33. Matsumoto A, Ma T, Babul N, Ahdieh H, Lee D (2002) Oxymorphine ER (20 mg and 40 mg) provides superior efficacy compared with placebo and oxycontin (20 mg) in pain associated with osteoarthritis: results of a randomized, controlled trial. In: 10th World Congress of Pain. San Diego, CA. 34. Onoprishvili I, Andria M, Kramer H, Ancevska-Taneva N, Hiller J, Simon E (2003) Interaction between the μ opioid receptor and fliamin A is involved in receptor regulation and trafficking. Mol Pharmacol 64:1092–1100. 35. Paquette J, Olmstead M (2005) Ultra-low dose naltrexone enhances cannabinoid-induced antinociception. Behav Pharmacol 16:597–603. 36. Powell KJ, Abul-Husn NS, Jhamandas A, Olmstead MC, Beninger RJ, Jhamandas K (2002) Paradoxical effects of the opioid antagonist naltrexone on morphine analgesia, tolerance, and reward in rats. J Pharmacol Exp Ther 300:588–596. 37. Roth SH, Fleischmann RM, Burch FX, Dietz R, Bockow B, Rapoport RJ, Rutstein J, Lacouture PG (2000) Around-the-clock, controlled-release oxycodone therapy for osteoarthritis-related pain. Arch Intern Med 160:853–860. 38. Saegusa H, Kurihara T, Zong S, Minowa O, Kazuno A, Han W, Matsuda Y, Yamanaka H, Osanai M, Noda T, Tanabe T (2000) Altered pain responses in mice lacking α1E subunit of the voltage-dependent Ca2 + channel. Proc Natl Acad Sci USA 97:6132–6137. 39. Shen KF, Crain SM (1989) Dual opioid modulation of the action potential duration of mouse dorsal root ganglion neurons in culture. Brain Research 491:227–242. 40. Shen K-F, Crain SM (1990) Cholera toxin-A subunit blocks opioid excitatory effects on sensory neuron action potentials indicating mediation by Gs-linked opioid receptors. Brain Research 525:225–231. 41. Shen KF, Crain SM (1994) Antagonists at excitatory opioid receptors on sensory neurons in culture increase potency and specificity of opiate analgesics and attenuate development of tolerance/dependence. Brain Res 636:286–297. 42. Shen KF, Crain SM (1995) Specific N- or C-terminus modified dynorphin and beta-endorphin peptides can selectively block excitatory opioid receptor functions in sensory neurons and unmask potent inhibitory effects of opioid agonists. Brain Res 673:30–38.

1

Low-Dose Naltrexone Improves Opioid Analgesia

17

43. Shen KF, Crain SM (1997) Ultra-low doses of naltrexone or etorphine increase morphine’s antinociceptive potency and attenuate tolerance/dependence in mice. Brain Res 757: 176–190. 44. Shen K-F, Crain SM, Moate P, Boston R, de Kater AW, Schoenhard GL (2002a) PTI-801, a novel formulation of oxycodone, shows absence of tolerance, physical dependence and naloxone-precipitated withdrawal effects in mice. J Pain 3:49. 45. Shen K-F, Crain SM, Moate P, Boston R, de Kater AW, Schoenhard GL (2002b) PTI-555, reverses and prevents morphine-induced tolerance and naloxone-precipitated withdrawal in mice chronically treated with morphine. J Pain 3:50. 46. Terner JM, Barrett AC, Lomas LM, Negus SS, Picker MJ (2006) Influence of low doses of naltrexone on morphine antinociception and morphine tolerance in male and female rats of four strains. Pain 122:90–101. 47. Vanderah TW, Gardell LR, Burgess SE, Ibrahim M, Zhong C-M, Ossipov MH, Lai J, Malan Jr. TP, Porreca F. (2000) Repeated spinal opioid administration produces abnormal pain and antinociceptive tolerance which is reversed by dynorphin antiserum. J Neurosci., 20: 7074–7079. 48. Vanderah TW, Suenaga NMH, Ossipov MH, Malan Jr. TP, Lai J, Porreca F. (2001) Descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci 21:279–286. 49. Vanderah TW, Burns LH (2004) Ultra-low-dose naltrexone plus morphine blocks thermal hyperalgesia and attenuates mechanical hypersensitivity in a neuropathic pain model. In: 2nd Joint Meeting of the American and Canadian Pain Societies, Vancouver, BC. 50. Wang H-Y, Friedman E (1999) Effects of lithium on receptor-mediated activation of G proteins in rat brain cortical membranes. Neuropharmacology 38:403–414. 51. Wang H-Y, Burns LH (2006) Gβγ that interacts with adenylyl cyclase in opioid tolerance originates from a Gs protein. J Neurbiol 66:1302–1310. 52. Wang H-Y, Frankfurt M, Burns LH (2008) High-affinity naloxone binding to filamin A prevents mu opioid receptor - Gs coupling underlying opioid tolerance and dependence. PLoS One 3:e1554. 53. Wang H-Y, Friedman E, Olmstead MC, Burns LH (2005) Ultra-low-dose naloxone suppresses opioid tolerance, dependence and associated changes in Mu opioid receptor–G protein coupling and Gβγ signaling. Neuroscience 135:247–261. 54. Webster LR, Butera PG, Moran LV, Wu N, Burns LH, Friedmann N (2006) Oxytrex minimizes physical dependence while providing effective analgesia: a randomized controlled trial in low-back pain. J Pain 7:937–946. 55. Zhang S-P, Wang H-Y, Lovenberg T, Codd E (2001) Functional studies of bradykinin receptors in Chinese hamster ovary cells stably expressing the human B2 bradykinin receptor. Internat Immunopharmacol 1:955–965. 56. Zhen X, Torres C, Wang H-Y, Friedman E (2001) Protein phosphatase 1 regulates brain D1A dopamine receptor phosphorylation: role in dopaminergic dysfunction after in utero cocaine exposure. J Neurosci 21:9160–9167.

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.