Next Article in Journal
Metronidazole Encephalopathy EEG Features: A Case Report with Systematic Review of the Literature
Next Article in Special Issue
Acute DOB and PMA Administration Impairs Motor and Sensorimotor Responses in Mice and Causes Hallucinogenic Effects in Adult Zebrafish
Previous Article in Journal
Does Insomnia Exist without Hyperarousal? What Else Can There Be?
Previous Article in Special Issue
Analyzing the Open/Deep Web to Better Understand the New/Novel Psychoactive Substances (NPS) Scenarios: Suggestions from CASSANDRA and NPS.Finder Research Projects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting the Orexin System for Prescription Opioid Use Disorder

by
Alessandra Matzeu
* and
Rémi Martin-Fardon
Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2020, 10(4), 226; https://doi.org/10.3390/brainsci10040226
Submission received: 10 March 2020 / Revised: 7 April 2020 / Accepted: 8 April 2020 / Published: 10 April 2020

Abstract

:
Prescription opioids are potent analgesics that are used for clinical pain management. However, the nonmedical use of these medications has emerged as a major concern because of dramatic increases in abuse and overdose. Therefore, effective strategies to prevent prescription opioid use disorder are urgently needed. The orexin system has been implicated in the regulation of motivation, arousal, and stress, making this system a promising target for the treatment of substance use disorder. This review discusses recent preclinical studies that suggest that orexin receptor blockade could be beneficial for the treatment of prescription opioid use disorder.

1. Introduction

The abuse of opioids and opioid overdoses have escalated dramatically in recent years. In 2017, around 1.7 million people in the United States suffered from substance use disorders that are related to prescription opioid pain relievers [1]. Major obstacles to the treatment of opioid use disorder (OUD) are successful detoxification and the prevention of overdose and relapse [2]. Pain is clinically managed using various potent analgesics, such as oxycodone, hydrocodone, fentanyl, and remifentanil. The nonmedical use of these prescription opioids has become a major concern because of dramatic increases in abuse and overdose [3,4,5]. Only a few medications are available for the treatment of OUD. Methadone and buprenorphine, for example, are opioid receptor agonists and are commonly used as replacement therapy for the treatment of OUD. These medications can be helpful to prevent opioid craving and relapse. However, they are poorly utilized because of their restricted access, the need to be administered in a maintenance regimen, the risk of overdose, and the social stigma associated with OUD [1]. This indicates that OUD still requires significant medical attention, thus underscoring the need to discover novel pharmacotherapies.
The accumulating evidence indicates that the orexin (Orx) system may be a treatment target for substance use disorder. Orexin projections are found throughout the brain [6,7] and participate in the regulation of several physiological functions (e.g., the regulation of feeding, sleep/wake states, and energy homeostasis; for a review, see [8]). Orexin neurons play a role in modulating reward function, particularly drug-directed behavior [9]. It has been shown that Orx neurons are activated by food-, morphine-, cocaine-, and alcohol-related stimuli [9,10,11,12,13]. Behavior that is motivated by drugs under high-demand conditions, and the motivation for drug-seeking that is potentiated by drug-conditioned stimuli, have been shown to involve the orexin system [14,15]. Nearly half of Orx neurons in rats express μ-opioid receptors, the main target of opioids [16,17]. Pharmacologically targeting the Orx system may therefore reduce opioid intake, craving, and relapse.
The present review discusses preclinical studies that demonstrated the involvement of the Orx system in substance use disorder in general and prescription OUD in particular.

2. The Orexin System and Substance Use Disorder

Orexin A (OrxA) and orexin B (OrxB), also called hypocretin 1 (Hcrt1) and hypocretin 2 (Hcrt2), respectively, are neuropeptides that were independently discovered in 1998 by two different research groups [18]. OrxA and OrxB derive from a common precursor (prepro-orexin) and are post-translational products of the Orx neuropeptide precursor gene. Orexin cell bodies are found in the lateral hypothalamus (LH) [19], the perifornical hypothalamus, the dorsomedial hypothalamus, and some scattered cells in the paraventricular nucleus [20]. Evidence indicates that reward-seeking functions are primarily associated with the recruitment of Orx cells in the LH [21,22], whereas arousal- and stress-related processes are associated with the recruitment of Orx neurons in the dorsomedial hypothalamus/perifornical hypothalamus [21,22]. Orexin receptor 1 (OrxR1; Hcrt-r1) and orexin receptor 2 (OrxR2; Hcrt-r2) [18,23,24] are involved in the regulation of highly motivated behavior, stress, and arousal [8,22,25,26,27,28,29,30,31], suggesting the potential of targeting them for the treatment of addiction. OrxR1 and OrxR2 mRNA expression is found throughout the brain [32,33], and each receptor subtype plays a different physiological role. The prefrontal cortex exhibits high OrxR1 mRNA expression, and the nucleus accumbens expresses high OrxR2 mRNA expression [32]. OrxR1 signaling has mainly been implicated in reward-seeking behavior, whereas OrxR2 signaling has been implicated in arousal and sleep/wake cycle regulation [22].
The orexin system has been shown to contribute to the regulation of motivation, especially highly motivated behavior, arousal, and stress, making this system an ideal target for addiction treatment [8,22,25,26,27,28,29,30,31]. The Orx system plays a prominent role in the motivation for reward (including drugs of abuse), stress, and anxiety [8,22,25,26,27,28,29,30,31], all of which contribute to problematic drug abuse [34,35]. Neural systems that are involved in processing natural rewards and drugs of abuse overlap; therefore, neuroplasticity that is caused by drug exposure may be responsible for maladaptive, compulsive, and addictive behavior [36,37,38]. Pharmacological manipulation of the Orx system is particularly effective in modifying the conditioned effects of drug cues in conditioned place preference and reinstatement studies [9,39,40]. We recently found that the Orx system is engaged to a greater extent by drugs of abuse than by natural non-drug reinforcers [1]. For example, although stimuli that were conditioned to cocaine, alcohol, and a conventional reinforcer (i.e., palatable sweet solution) were equally effective in eliciting reinstatement, the OrxR1 antagonist SB334867 selectively reversed the conditioned reinstatement that was induced by a cocaine- or alcohol-related stimulus, but had no effects on the same stimulus that was conditioned to a conventional reinforcer ([11,41,42] this laboratory). One possible explanation for the preferential reversal of conditioned reinstatement for drugs vs. non-drugs by SB334867 could be that the drug-induced neuroadaptation to neural systems that controls motivation under normal conditions (i.e., toward conventional reinforcers) is only revealed when blocking OrxR1 with SB334867. There is evidence of drug-induced dysregulation of Orx transmission. Earlier findings revealed that Orx mRNA expression is upregulated in the LH in inbred alcohol-preferring (iP) rats following chronic alcohol consumption [39]. Moreover, Orx production has been reported to be affected by opioids. Nine days after 1 week of 3 h/day heroin self-administration, a significant increase in Orx mRNA levels was observed in the LH [43]. Other studies have reported greater Orx expression during acute withdrawal from chronic morphine or heroin exposure [44,45]. Furthermore, it was recently reported that a higher number of Orx cells are found in the postmortem brains of heroin addicts [46], and in rodents exposed to long term morphine [46] or cocaine [47,48], suggesting that increased numbers of Orx-expressing neurons may have a crucial role in defining propensity and severity in the addiction phenotype. Because OrxA has been found to decrease the excitability of brain reward systems in the LH [49], it is possible that alterations of Orx expression may also be involved in negative affective states that are associated with opioid withdrawal and abstinence, which may be important in the modulation of opioid-seeking behavior [9,50].
One class of OrxR antagonists has been synthesized to target OrxR1. These single OrxR antagonists (SORAs), have 50-fold greater selectivity for OrxR1 over OrxR2 [24,51]. For example, N-(2-methyl-6-benzoxazolyl)-N’-1,5-naphthyridin-4-yl urea (SB334867) [52,53] has been shown to have very high selectivity for OrxR1, bioavailability, and potency [24,51]. SORAs have also been synthesized to specifically target OrxR2, with ≥ 250-fold selectivity for OrxR2 over OrxR1 [24,51], and the capacity to reduce numerous addiction-like behaviors. Findings that have described the use of SORAs that target OrxR2 on drug-motivated behavior are inconsistent vs. OrxR1-specific SORAs, such as SB334867. A third class of OrxR antagonists, referred to as dual OrxR antagonists (DORAs), target both OrxR1 and OrxR2, and have been developed to treat insomnia. Examples of DORAs include almorexant ([2R]-2-[(1S)-6,7-dimethoxy-1-(2-[4-(trifluoromethyl)phenyl]ethyl]-3,4-dihydro-1H-isoquinolin-2-yl)-N-methyl-2-phenylacetamide [ACT078573]) [54,55] and suvorexant ([(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl]-[5-methyl-2-(triazol-2-yl)phenyl]methanone [MK4305]) [56]. To date, although most of the research in the field that has studied the effects of DORAs on addiction-like behaviors has tested almorexant [57,58,59], interest in the use of suvorexant [60,61] has recently arisen. Suvorexant was approved for the treatment of insomnia in the United States in 2014 (for review, see [62]), and could be used to treat drug-induced sleep disruptions (e.g., [63]) that are a significant contributor to relapse. Most preclinical studies that have examined the efficacy of blocking OrxRs tested only acute antagonist administration. Repeated OrxR1 inhibition, under some conditions, has been shown to exert longer-lasting preventive effects against drug-related, cue-induced cocaine-seeking [64]. The orexin neurons interface with several neural systems and are involved in various physiological processes. Repeated OrxR inhibition may cause nonspecific side effects by influencing numerous neural pathways. Dysfunctions of the Orx system can cause narcolepsy [65,66,67], while pharmacological manipulations of the Orx system can have nonspecific side effects on food intake [68,69,70], inhibitory responses [71,72,73], and learning and memory [74,75,76]. Such untoward effects need to be considered when investigating the clinical potential of OrxR antagonist treatment.

3. Manipulation of the Orexin System to Treat Prescription Opioid Use Disorder

The interest in testing OrxR antagonists for the treatment of prescription OUD was spawned by earlier studies that suggested that the Orx system has a function in regulating the high motivation to take and seek drugs of abuse [77,78,79,80,81,82]. Some recent studies have investigated the potential of targeting the orexin system for the treatment of prescription opioid misuse. OrxR1 antagonism was shown to effectively reduce the motivation for oxycodone, fentanyl, and remifentanil in preclinical rodent studies [83,84,85,86,87].
We recently reported that rats self-administered oxycodone under a long-access schedule. During these 12 h daily sessions, the rats exhibited a significant increase in oxycodone intake and signs of oxycodone dependence [83]. Discriminative stimuli (SD) were conditioned to oxycodone self-administration and eventually gained the ability to reinstate oxycodone-seeking, and this conditioned effect was resistant to extinction over five presentations of the SD (i.e., 30 days after the last oxycodone self-administration session). Importantly, this study also found that OrxR1 blockade with SB334867, but not OrxR2 blockade with TCSOX229, decreased oxycodone intake (Table 1), which was consistent with studies of heroin, fentanyl, and remifentanil self-administration [84,85,87,88,89]. However, these oxycodone data partially contrasted with findings on cocaine self-administration, in which OrxR1 blockade with SB334867 did not affect cocaine self-administration [77,90], unless the contingency to take cocaine was kept under a high-effort schedule (e.g., high fixed-ratio or progressive-ratio schedule of reinforcement; for a review, see [91]). Notably, however, Orx silencing reduced cocaine self-administration under a low-effort schedule (i.e., FR1 timeout 20 s schedule of reinforcement) [92] under extended-access conditions (6 h/day). These findings suggest the feasibility of normalizing compulsive drug intake in addicted individuals by inhibiting Orx signaling. We also found that the OrxR1 antagonist SB334867, but not the OrxR2 antagonist TCSOX229, prevented the SD-induced conditioned reinstatement of oxycodone-seeking behavior ([83], Table 1), thus supporting the involvement of Orx, particularly in reward-seeking that was elicited by external stimuli when the motivation for drug-seeking was augmented by giving stimuli that were conditioned to the drug [15]. Requiring further exploration are the mechanisms by which SB334867 produced alterations of oxycodone-seeking and -taking behavior in our study [83]. SB334867 may lower the general motivation for oxycodone, reflected by decreases in self-administration and reinstatement, suggesting the involvement of OrxR1 in oxycodone’s reinforcing effects and oxycodone-related, SD-driven, appetitive behavior. Numerous studies reported that cue- and context-induced reward-seeking behavior for cocaine, heroin, alcohol, fentanyl, and sucrose involved OrxR1-mediated Orx transmission [12,28,39,40,42,87,88,93,94]. Overall, these results demonstrate that OrxR1 is implicated in the mediation of the reinforcing effects of oxycodone and appetitive behavior that is controlled by oxycodone-related stimuli, and may be a promising therapeutic target for the treatment and prevention of prescription OUD.
Rats will also voluntarily self-administer remifentanil, a µ-opioid agonist with a short duration of action, and achieve similar breakpoints to heroin on a progressive-ratio schedule of reinforcement [95]. In 2015, Porter-Stransky et al. [84] employed a behavioral economics approach in rats and observed extensive individual variability in the intake of the ultra-short-acting opioid remifentanil [84]. In a behavioral economics procedure, the Q0 value is a hypothetical measure of the amount of drug an individual would use while the drug is entirely free (i.e., free consumption, likewise a measure of the hedonic set point). The Q0 value is measured by a demand curve intercept of the ordinate and the slope of the demand curve. The demand elasticity, α, denotes how consumption fluctuates relative to cost independently of Q0 [96,97]. These behavioral economics demand curve approaches are useful for measuring the motivation for drugs, because they capture individual differences in the motivation for drugs, enable comparisons between different drugs, and offer unique parameters of motivation. Briefly, rats with a low baseline Q0 (i.e., low takers) exhibit high demand elasticity, α (i.e., lower responding as the drug price increases, signifying low motivation for the drug), while rats with a high Q0 (i.e., high takers) show low demand elasticity, α, by continuing to self-administer the drug in spite of the higher cost, thus reflecting higher motivation for the drug. Using this model, the authors found that the OrxR1 antagonist SB334867 reduced the motivation to self-administer remifentanil only in low-taker rats (Table 2). Similarly, SB334867 attenuated the cue-induced, but not drug-induced, reinstatement of remifentanil-seeking only in the low-taker group (Table 2). These results appear to be in contrast with previous observations of the effects of OrxR1 blockade on the motivation for different drugs of abuse (e.g., cocaine and alcohol). Indeed, SB334867 was shown to more effectively reduce alcohol drinking in high-alcohol-preferring rats [98] as well as intake- and cue-induced reinstatement in rats with a high demand for cocaine [99,100]. These differences support the hypothesis that different neurobiological mechanisms mediate the motivation for different drugs of abuse. Two follow-up studies from the same group used the behavioral economics approach, and confirmed the ability of SB334867 to reduce the motivation for remifentanil and decrease cue-induced reinstatement ([85,86], Table 2). Moreover, they identified the ventral pallidum as an important brain region for the regulation of the motivation for remifentanil and cue-induced reinstatement [85,86]. The ventral pallidum receives Orx inputs, has high OrxR1 mRNA expression [20,32], and has been critically linked to the drive for reward-seeking behavior [101,102,103,104], suggesting that this region may be a site where Orx modulates reward behavior. Interestingly, the authors showed that the inhibition of remifentanil-seeking by SB334867 persisted beyond the pharmacological availability of SB334867 [86].
Another recent study investigated the efficacy of SB334867 in reducing the motivation for fentanyl ([87], Table 3). Using the behavioral economics procedure, the study found that rats readily self-administered fentanyl and exhibited somatic signs of withdrawal. The level of motivation for fentanyl predicted the magnitude of the reinstatement of fentanyl-seeking behavior that was induced by the presentation of stimuli that were previously concomitant with the drug. Further corroborating the potential use of SB334867 for the treatment of prescription OUD, the study also showed that systemic pretreatment with SB334867 decreased the motivation for fentanyl without altering sucrose self-administration. The blockade of OrxR1 with SB334867 reduced the cue-induced reinstatement of fentanyl. Orexin neurons are activated in response to drug-associated cues and contexts [13,89,105], and OrxR1 functionality is crucial for the cue-induced reinstatement of oxycodone-, heroin-, remifentanil-, cocaine-, alcohol-, nicotine-, and sucrose-seeking [83,84,88,93,105,106]. Importantly, the authors of this study [87] found a positive association between the motivation for fentanyl and the efficacy of SB334867 in reducing fentanyl intake and seeking, underscoring the relevance of OrxR1 signaling to the maintenance of motivated behavior, especially in individuals that exhibit high motivation for the drug. Although Orx neurons have been shown to be activated by food-related cues [9,107], SB334867 can selectively decrease drug-seeking without interfering with regular food intake in ad libitum-fed animals [77,108,109], or reward-seeking that is produced by stimuli that are conditioned to a palatable sweet solution [42,94]. Such findings demonstrate that OrxR1 may be a viable treatment target to selectively reduce the craving and motivation for prescription opioids. One common limitation of these studies, however, is that the efficacy of an OrxR2 antagonist was not tested [84,85,86,87], in contrast to our study with oxycodone [83]. Thus, we cannot completely exclude the possibility that OrxR2 signaling may also regulate the motivation for remifentanil and fentanyl. OrxR2 signaling mediates stressful states and arousal, aspects that characterize substance use disorders in general. In fact, a recent study showed that antagonizing OrxR2 with NBI-80713 produced a significant decrease in heroin self-administration only in rats that were made heroin-dependent [110], strongly suggesting that OrxR2 might play an important role in prescription OUD. Future studies are warranted to systematically explore the role of OrxR2 signaling, both alone and combined with OrxR1 signaling, to better understand the potential to target the orexin system to treat prescription OUD.

4. Potential Use of DORAs to Maintain Abstinence and Prevent Relapse

Acute treatment with opioids for pain management appears to improve sleep quality [111]. However, as use continues, sleep deficits can occur [112]. Sleep architecture is altered as opioid dependence and withdrawal develop [113,114]. Alterations of sleep architecture that occur during withdrawal from short-term opioid administration may be different from changes that occur during withdrawal from chronic use. Significant insomnia commonly occurs among opioid users long after withdrawal, characterized by increases in sleep latency and time awake after sleep onset, and accompanying frequent arousals [115]. Withdrawal-associated sleep problems, including sleep loss and worse sleep quality, have been suggested to contribute to opioid use and relapse. Sleep disturbances are prominent symptoms of substance-related disorders and could account for a substantial proportion of sleep problems [63]. Opioid exposure is known to directly suppress the hypothalamic Orx system [116], perhaps explaining the lethargy and cognitive dysfunction that are induced by opioid use [117,118]. Shortly after its discovery, the key role of the Orx system in wakefulness became clear, raising the possibility that OrxR antagonism may be a new therapeutic approach for the treatment of insomnia by blocking the Orx-mediated drive of wakefulness [54,119]. Under physiological conditions, Orx neuron activity and peptide levels show diurnal variations. High Orx activity is correlated with wakefulness, and low Orx activity is correlated with sleep [120,121,122]. The wake-promoting functions of Orx are supported by the finding that arousal is increased by the administration of exogenous Orx peptides and the optogenetic activation of Orx neurons [119,123,124,125]. Nevertheless, the neurobiology of sleep/wake cycling is complex and regulated by interactions between sleep-promoting systems (e.g., the inhibitory γ-aminobutyric acid system) and wake-promoting systems (e.g., Orx, acetylcholine, and monoamines) [126,127].
Consistent with the sleep-promoting role of Orx, SORAs that are selective for OrxR2 were shown to have sleep-promoting properties, whereas SORAs that are selective for OrxR1 were largely ineffective with regard to promoting sleep [128,129]. In addition to SORAs, some DORAs have been synthesized as potential therapeutics for the management of sleep/wake disorders [62]. Interestingly, OrxR2 SORAs appear to require a higher receptor occupancy to achieve their effect on sleep maintenance compared with DORAs, suggesting that OrxR1 also plays a role in sleep maintenance [128]. DORAs block the activity of OrxR1 and OrxR2 to reduce the threshold of the transition to sleep and attenuate Orx-mediated arousal [62]. Among DORAs, suvorexant has been approved by the United States Food and Drug Administration for the treatment of insomnia (for a review, see [62]). Because of the efficacy of suvorexant in treating sleep disorders and the efficacy of OrxR antagonists in reducing “drug motivation,” some clinical trials are now studying the efficacy of suvorexant in normalizing sleep, craving, and stress states in patients who are affected by substance use disorders, including OUD [130,131]. However, even though clinical trials are presently ongoing to examine the efficacy of suvorexant on OUD (see [130,131]), to our knowledge no data are available yet. Moreover, other potential OrxR antagonists have been synthesized for the management of sleep disorders [62,132]. One major obstacle to the treatment of OUD is the recurrence of relapse to opioid misuse even after a long time of imposed or voluntary abstinence [2]. Chronic opioid use induces the development of physical dependence and withdrawal symptoms when drug use is interrupted. Withdrawal symptoms gradually fade within a few days but can sometimes last for weeks after use is discontinued [2]. In patients who suffer from prescription OUD, insomnia is a major withdrawal symptom that might ultimately represent one of the major factors that triggers relapse. Thus, alleviating sleep disturbances during opioid withdrawal might attenuate relapse vulnerability. We hypothesize that the sleep-promoting Orx system is compromised as opioid dependence and withdrawal develop. It has been shown that suvorexant exhibits a long half-life (up to 13 h), and depending on the dose administered, sufficient occupancy of OrxRs occurs [132,133]. Therefore, daily treatment with a DORA compound, such as suvorexant, in the evening within 30 minutes of bedtime, should not only be sufficient to relieve sleep disturbances during opioid withdrawal but could also reduce craving and attenuate relapse vulnerability the following day [130,132].

5. Conclusions

To date, only a few studies have investigated the efficacy of SORAs in reducing the intake and reinstatement of synthetic prescription opioids, and most of them were focused on OrxR1, neglecting the potential importance of OrxR2. Moreover, no preclinical study has reported the use of DORAs to treat prescription OUD, indicating that more preclinical work would be needed to elucidate the ways these compounds can be beneficial in the clinical conditions of prescription OUD. To achieve and maintain abstinence, treatments for prescription OUD should alleviate not only symptoms of craving and the motivation for the drug, but they should also reduce anxiety-related states provoked by opioid abuse and sleep disturbances that occur as dependence and withdrawal develop. With regard to the anxiety-related states induced by opioids, clinical studies have demonstrated the participation of the Orx system in the emotive perturbation that occurs during alcohol withdrawal [134,135]. This could also be the case for prescription opioids. Knowing the extensive projections of the Orx system throughout the brain, its role is not likely to be limited to sleep regulation, but encompasses other neuropsychiatric disorders, such as anxiety, depression, and prescription OUD. As of today, only a limited amount of preclinical studies suggest a role of OrxRs in prescription OUD, and even though more investigations are needed to elucidate the ways in which prescription opioids perturb the Orx system as dependence and withdrawal develop, the existing literature strongly supports the hypothesis that the inhibition of Orx signaling could be an original approach for the treatment of prescription OUD.

Acknowledgments

This is publication number 29950 from The Scripps Research Institute. Supported by the National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism (grant no. AA006420 and AA026999 to RM-F) and by the Merck Investigator Studies Program (grant no. MISP59371 to AM). The authors thank Michael Arends for assistance with manuscript preparation. The authors declare no conflict of interest.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Orxorexin
OrxR1orexin receptor 1
OrxR2orexin receptor 2
SORAssingle orexin receptor antagonists
DORAsdual orexin receptor antagonists
SB334867N-(2-methyl-6-benzoxazolyl)-N’-1,5-naphthyridin-4-yl urea

References

  1. NIH-NIDA. Medications to Treat Opioid Use Disorder. National Institute on Drug Abuse, Advancing Addiction Science. 2018. Available online: https://www.drugabuse.gov/publications/research-reports/medications-to-treat-opioid-addiction/overview (accessed on 22 February 2020).
  2. Volkow, N.D.; Jones, E.B.; Einstein, E.B.; Wargo, E.M. Prevention and Treatment of Opioid Misuse and Addiction: A Review. JAMA Psychiatry 2019, 76, 208–216. [Google Scholar] [CrossRef] [PubMed]
  3. Jones, C.M.; Muhuri, P.K.; Lurie, P.G. Trends in the Nonmedical Use of OxyContin, United States, 2006 to 2013. Clin. J. Pain 2017, 33, 452–461. [Google Scholar] [CrossRef] [PubMed]
  4. Muhuri, P.K.; Gfroerer, J.C.; Davies, C. Associations of Nonmedical Pain Reliever Use and Initiation of Heroin Use in the United States; SAMHSA; CBHSQ (Center for Behavioral Health Statistics and Quality) Data Rev. August 2013. Available online: https://www.samhsa.gov/data/sites/default/files/DR006/DR006/nonmedical-pain-reliever-use-2013.htm (accessed on 22 February 2020).
  5. Kolodny, A.; Courtwright, D.T.; Hwang, C.S.; Kreiner, P.; Eadie, J.L.; Clark, T.W.; Alexander, G.C. The prescription opioid and heroin crisis: A public health approach to an epidemic of addiction. Annu. Rev. Public Health 2015, 36, 559–574. [Google Scholar] [CrossRef] [PubMed]
  6. Date, Y.; Ueta, Y.; Yamashita, H.; Yamaguchi, H.; Matsukura, S.; Kangawa, K.; Nakazato, M. Orexins, orexigenic hypothalamic peptides, interact with autonomic, neuroendocrine and neuroregulatory systems. Proc. Natl. Acad. Sci. USA 1999, 96, 748–753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Peyron, C.; Tighe, D.K.; van den Pol, A.N.; de Lecea, L.; Heller, H.C.; Sutcliffe, J.G.; Kilduff, T.S. Neurons containing hypocretin (orexin) project to multiple neuronal systems. J. Neurosci. 1998, 18, 9996–10015. [Google Scholar] [CrossRef] [Green Version]
  8. Tsujino, N.; Sakurai, T. Role of orexin in modulating arousal, feeding, and motivation. Front. Behav. Neurosci. 2013, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  9. Harris, G.C.; Wimmer, M.; Aston-Jones, G. A role for lateral hypothalamic orexin neurons in reward seeking. Nature 2005, 437, 556–559. [Google Scholar] [CrossRef]
  10. Dayas, C.V.; McGranahan, T.M.; Martin-Fardon, R.; Weiss, F. Stimuli linked to ethanol availability activate hypothalamic CART and orexin neurons in a reinstatement model of relapse. Biol. Psychiatry 2008, 63, 152–157. [Google Scholar] [CrossRef]
  11. Martin-Fardon, R.; Zorrilla, E.P.; Ciccocioppo, R.; Weiss, F. Role of innate and drug-induced dysregulation of brain stress and arousal systems in addiction: Focus on corticotropin-releasing factor, nociceptin/orphanin FQ, and orexin/hypocretin. Brain Res. 2010, 1314, 145–161. [Google Scholar] [CrossRef] [Green Version]
  12. Jupp, B.; Krstew, E.; Dezsi, G.; Lawrence, A.J. Discrete cue-conditioned alcohol-seeking after protracted abstinence: Pattern of neural activation and involvement of orexin receptors. Br. J. Pharmacol. 2011, 162, 880–889. [Google Scholar] [CrossRef] [Green Version]
  13. Martin-Fardon, R.; Cauvi, G.; Kerr, T.M.; Weiss, F. Differential role of hypothalamic orexin/hypocretin neurons in reward seeking motivated by cocaine versus palatable food. Addict. Biol. 2018, 23, 6–15. [Google Scholar] [CrossRef] [PubMed]
  14. Hollander, J.A.; Pham, D.; Fowler, C.D.; Kenny, P.J. Hypocretin-1 receptors regulate the reinforcing and reward-enhancing effects of cocaine: Pharmacological and behavioral genetics evidence. Front. Behav. Neurosci. 2012, 6, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mahler, S.V.; Moorman, D.E.; Smith, R.J.; James, M.H.; Aston-Jones, G. Motivational activation: A unifying hypothesis of orexin/hypocretin function. Nat. Neurosci. 2014, 17, 1298–1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Lalovic, B.; Kharasch, E.; Hoffer, C.; Risler, L.; Liu-Chen, L.Y.; Shen, D.D. Pharmacokinetics and pharmacodynamics of oral oxycodone in healthy human subjects: Role of circulating active metabolites. Clin. Pharmacol. Ther. 2006, 79, 461–479. [Google Scholar] [CrossRef]
  17. Peckham, E.M.; Traynor, J.R. Comparison of the antinociceptive response to morphine and morphine-like compounds in male and female Sprague-Dawley rats. J. Pharmacol. Exp. Ther. 2006, 316, 1195–1201. [Google Scholar] [CrossRef] [Green Version]
  18. Sakurai, T.; Amemiya, A.; Ishii, M.; Matsuzaki, I.; Chemelli, R.M.; Tanaka, H.; Williams, S.C.; Richardson, J.A.; Kozlowski, G.P.; Wilsonet, S.; et al. Orexins and orexin receptors: A family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 1998, 92, 573–585. [Google Scholar] [CrossRef] [Green Version]
  19. DiLeone, R.J.; Georgescu, D.; Nestler, E.J. Lateral hypothalamic neuropeptides in reward and drug addiction. Life Sci. 2003, 73, 759–768. [Google Scholar] [CrossRef]
  20. Baldo, B.A.; Daniel, R.A.; Berridge, C.W.; Kelley, A.E. Overlapping distributions of orexin/hypocretin- and dopamine-beta-hydroxylase immunoreactive fibers in rat brain regions mediating arousal, motivation, and stress. J. Comp. Neurol. 2003, 464, 220–237. [Google Scholar] [CrossRef]
  21. Harris, G.C.; Aston-Jones, G. Arousal and reward: A dichotomy in orexin function. Trends Neurosci. 2006, 29, 571–577. [Google Scholar] [CrossRef]
  22. Aston-Jones, G.; Smith, R.J.; Sartor, G.C.; Moorman, D.E.; Massi, L.; Tahsili-Fahadan, P.; Richardson, K. Lateral hypothalamic orexin/hypocretin neurons: A role in reward-seeking and addiction. Brain Res. 2010, 1314, 74–90. [Google Scholar] [CrossRef] [Green Version]
  23. Ammoun, S.; Holmqvist, T.; Shariatmadari, R.; Oonk, H.B.; Detheux, M.; Parmentier, M.; Kukkonen, J.P. Distinct recognition of OX1 and OX2 receptors by orexin peptides. J. Pharmacol. Exp. Ther. 2003, 305, 507–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Scammell, T.E.; Winrow, C.J. Orexin receptors: Pharmacology and therapeutic opportunities. Annu. Rev. Pharmacol. Toxicol. 2011, 51, 243–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Aston-Jones, G.; Smith, R.J.; Moorman, D.E.; Richardson, K.A. Role of lateral hypothalamic orexin neurons in reward processing and addiction. Neuropharmacology 2009, 56 (Suppl. 1), 112–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Koob, G.F. Brain stress systems in the amygdala and addiction. Brain Res. 2009, 1293, 61–75. [Google Scholar] [CrossRef] [Green Version]
  27. Berridge, C.W.; Espana, R.A.; Vittoz, N.M. Hypocretin/orexin in arousal and stress. Brain Res. 2010, 1314, 91–102. [Google Scholar] [CrossRef] [Green Version]
  28. Cason, A.M.; Smith, R.J.; Tahsili-Fahadan, P.; Moorman, D.E.; Sartor, G.C.; Aston-Jones, G. Role of orexin/hypocretin in reward-seeking and addiction: Implications for obesity. Physiol. Behav. 2010, 100, 419–428. [Google Scholar] [CrossRef] [Green Version]
  29. Giardino, W.J.; de Lecea, L. Hypocretin (orexin) neuromodulation of stress and reward pathways. Curr. Opin. Neurobiol. 2014, 29, 103–108. [Google Scholar] [CrossRef] [Green Version]
  30. James, M.H.; Campbell, E.J.; Dayas, C.V. Role of the Orexin/Hypocretin System in Stress-Related Psychiatric Disorders. Curr. Top Behav. Neurosci. 2017, 33, 197–219. [Google Scholar]
  31. Kuwaki, T.; Zhang, W. Orexin neurons and emotional stress. Vitam. Horm. 2012, 89, 135–158. [Google Scholar]
  32. Marcus, J.N.; Aschkenasi, C.J.; Lee, C.E.; Chemelli, R.M.; Saper, C.B.; Yanagisawa, M.; Elmquist, J.K. Differential expression of orexin receptors 1 and 2 in the rat brain. J. Comp. Neurol. 2001, 435, 6–25. [Google Scholar] [CrossRef]
  33. Trivedi, P.; Yu, H.; MacNeil, D.J.; Van der Ploeg, L.H.; Guan, X.M. Distribution of orexin receptor mRNA in the rat brain. FEBS Lett. 1998, 438, 71–75. [Google Scholar] [CrossRef] [Green Version]
  34. Wise, R.A.; Koob, G.F. The development and maintenance of drug addiction. Neuropsychopharmacology 2014, 39, 254–262. [Google Scholar] [CrossRef] [PubMed]
  35. Koob, G.F.; Volkow, N.D. Neurobiology of addiction: A neurocircuitry analysis. Lancet Psychiatry 2016, 3, 760–773. [Google Scholar] [CrossRef]
  36. Kelley, A.E.; Berridge, K.C. The neuroscience of natural rewards: Relevance to addictive drugs. J. Neurosci. 2002, 22, 3306–3311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Kalivas, P.W.; O’Brien, C. Drug addiction as a pathology of staged neuroplasticity. Neuropsychopharmacology 2008, 33, 166–180. [Google Scholar] [CrossRef] [Green Version]
  38. Wanat, M.J.; Willuhn, I.; Clark, J.J.; Phillips, P.E. Phasic dopamine release in appetitive behaviors and drug addiction. Curr. Drug Abuse Rev. 2009, 2, 195–213. [Google Scholar] [CrossRef]
  39. Lawrence, A.J.; Cowen, M.S.; Yang, H.J.; Chen, F.; Oldfield, B. The orexin system regulates alcohol-seeking in rats. Br. J. Pharmacol. 2006, 148, 752–759. [Google Scholar] [CrossRef]
  40. Smith, R.J.; Tahsili-Fahadan, P.; Aston-Jones, G. Orexin/hypocretin is necessary for context-driven cocaine-seeking. Neuropharmacology 2009, 58, 179–184. [Google Scholar] [CrossRef] [Green Version]
  41. Martin-Fardon, R.; Weiss, F. Differential Effects of an Orx/Hcrt Antagonist on Reinstatement Induced by a Cue Conditioned to Cocaine vs. Palatable Natural Reward. In 2009 Neuroscience Meeting Planner; Program No. 65.21; Society for Neuroscience: Chicago, IL, USA, 2009. [Google Scholar]
  42. Martin-Fardon, R.; Weiss, F. N-(2-methyl-6-benzoxazolyl)-N’-1,5-naphthyridin-4-yl urea (SB334867), a hypocretin receptor-1 antagonist, preferentially prevents ethanol seeking: Comparison with natural reward seeking. Addict. Biol. 2014, 19, 233–236. [Google Scholar] [CrossRef] [Green Version]
  43. Zhou, Y.; Leri, F.; Cummins, E.; Kreek, M.J. Individual differences in gene expression of vasopressin, D2 receptor, POMC and orexin: Vulnerability to relapse to heroin-seeking in rats. Physiol. Behav. 2015, 139, 127–135. [Google Scholar] [CrossRef] [Green Version]
  44. Georgescu, D.; Zachariou, V.; Barrot, M.; Mieda, M.; Willie, J.T.; Eisch, A.J.; Yanagisawa, M.; Nestler, E.J.; Dileone, R. Involvement of the lateral hypothalamic peptide orexin in morphine dependence and withdrawal. J. Neurosci. 2003, 23, 3106–3111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhou, Y.; Bendor, J.; Hofmann, L.; Randesi, M.; Ho, A.; Kreek, M.J. Mu opioid receptor and orexin/hypocretin mRNA levels in the lateral hypothalamus and striatum are enhanced by morphine withdrawal. J. Endocrinol. 2006, 191, 137–145. [Google Scholar] [CrossRef] [Green Version]
  46. Thannickal, T.C.; John, J.; Shan, L.; Swaab, D.F.; Wu, M.-F.; Ramanathan, L.; McGregor, R.; Chew, K.-T.; Cornford, M.; Yamanaka, A.; et al. Opiates increase the number of hypocretin-producing cells in human and mouse brain and reverse cataplexy in a mouse model of narcolepsy. Sci. Transl. Med. 2018, 10, eaao4953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. James, M.H.; Stopper, C.M.; Zimmer, B.A.; Koll, N.E.; Bowrey, H.E.; Aston-Jones, G. Increased Number and Activity of a Lateral Subpopulation of Hypothalamic Orexin/Hypocretin Neurons Underlies the Expression of an Addicted State in Rats. Biol. Psychiatry 2019, 85, 925–935. [Google Scholar] [CrossRef] [PubMed]
  48. Pantazis, C.B.; James, M.H.; Bentzley, B.S.; Aston-Jones, G. The number of lateral hypothalamus orexin/hypocretin neurons contributes to individual differences in cocaine demand. Addict. Biol. 2019, e12795, [Epub ahead of print]. [Google Scholar] [CrossRef] [PubMed]
  49. Boutrel, B.; Kenny, P.J.; Specio, S.E.; Martin-Fardon, R.; Markou, A.; Koob, G.F.; de Lecea, L. Role for hypocretin in mediating stress-induced reinstatement of cocaine-seeking behavior. Proc. Natl. Acad. Sci. USA 2005, 102, 19168–19173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Leri, F.; Zhou, Y.; Goddard, B.; Levy, A.; Jacklin, D.; Kreek, M.J. Steady-state methadone blocks cocaine seeking and cocaine-induced gene expression alterations in the rat brain. Eur. Neuropsychopharmacol. 2009, 19, 238–249. [Google Scholar] [CrossRef] [Green Version]
  51. Zhou, L.; Sun, W.L.; See, R.E. Orexin Receptor Targets for Anti-Relapse Medication Development in Drug Addiction. Pharmaceuticals (Basel) 2011, 4, 804–821. [Google Scholar] [CrossRef]
  52. Porter, R.A.; Chan, W.N.; Coulton, S.; Johns, A.; Hadley, M.S.; Widdowson, K.; Jerman, J.C.; Brough, S.J.; Coldwell, M.; Smart, D.; et al. 1,3-Biarylureas as selective non-peptide antagonists of the orexin-1 receptor. Bioorg. Med. Chem. Lett. 2001, 11, 1907–1910. [Google Scholar] [CrossRef]
  53. Smart, D.; Sabido-David, C.; Brough, S.J.; Jewitt, F.; Johns, A.; Porter, R.A.; Jerman, J.C. SB-334867-A: The first selective orexin-1 receptor antagonist. Br. J. Pharmacol. 2001, 132, 1179–1182. [Google Scholar] [CrossRef] [Green Version]
  54. Brisbare-Roch, C.; Dingemanse, J.; Koberstein, R.; Hoever, P.; Aissaoui, H.; Flores, S.; Mueller, C.; Nayler, O.; Van Gerven, J.; De Haas, S.L.; et al. Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat. Med. 2007, 13, 150–155. [Google Scholar] [CrossRef] [PubMed]
  55. Malherbe, P.; Borroni, E.; Pinard, E.; Wettstein, J.G.; Knoflach, F. Biochemical and electrophysiological characterization of almorexant, a dual orexin 1 receptor (OX1)/orexin 2 receptor (OX2) antagonist: Comparison with selective OX1 and OX2 antagonists. Mol. Pharmacol. 2009, 76, 618–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Cox, C.D.; Garbaccio, R.M. Discovery of allosteric inhibitors of kinesin spindle protein (KSP) for the treatment of taxane-refractory cancer: MK-0731 and analogs. Anticancer Agents Med. Chem. 2010, 10, 697–712. [Google Scholar] [CrossRef] [PubMed]
  57. Steiner, M.A.; Lecourt, H.; Jenck, F. The dual orexin receptor antagonist almorexant, alone and in combination with morphine, cocaine and amphetamine, on conditioned place preference and locomotor sensitization in the rat. Int. J. Neuropsychopharmacol. 2013, 16, 417–432. [Google Scholar] [CrossRef] [Green Version]
  58. LeSage, M.G.; Perry, J.L.; Kotz, C.M.; Shelley, D.; Corrigall, W.A. Nicotine self-administration in the rat: Effects of hypocretin antagonists and changes in hypocretin mRNA. Psychopharmacology 2010, 209, 203–212. [Google Scholar] [CrossRef] [Green Version]
  59. Hoch, M.; Hay, J.L.; Hoever, P.; de Kam, M.L.; te Beek, E.T.; van Gerven, J.M.; Dingemanse, J. Dual orexin receptor antagonism by almorexant does not potentiate impairing effects of alcohol in humans. Eur. Neuropsychopharmacol. 2013, 23, 107–117. [Google Scholar] [CrossRef]
  60. Simmons, S.J.; Gentile, T.A. Cocaine abuse and midbrain circuits: Functional anatomy of hypocretin/orexin transmission and therapeutic prospect. Brain Res. 2019, 1731, 146164. [Google Scholar] [CrossRef]
  61. Gentile, T.A.; Simmons, S.J.; Barker, D.J.; Shaw, J.K.; Espana, R.A.; Muschamp, J.W. Suvorexant, an orexin/hypocretin receptor antagonist, attenuates motivational and hedonic properties of cocaine. Addict. Biol. 2018, 23, 247–255. [Google Scholar] [CrossRef] [Green Version]
  62. Coleman, P.J.; Gotter, A.L.; Herring, W.J.; Winrow, C.J.; Renger, J.J. The Discovery of Suvorexant, the First Orexin Receptor Drug for Insomnia. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 509–533. [Google Scholar] [CrossRef]
  63. Garcia, A.N.; Salloum, I.M. Polysomnographic sleep disturbances in nicotine, caffeine, alcohol, cocaine, opioid, and cannabis use: A focused review. Am. J. Addict. 2015, 24, 590–598. [Google Scholar] [CrossRef]
  64. Zhou, L.; Smith, R.J.; Do, P.H.; Aston-Jones, G.; See, R.E. Repeated orexin 1 receptor antagonism effects on cocaine seeking in rats. Neuropharmacology 2012, 63, 1201–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bassetti, C.L.; Adamantidis, A.; Burdakov, D.; Han, F.; Gay, S.; Kallweit, U.; Khatami, R.; Koning, F.; Kornum, B.R.; Lammers, G.J.; et al. Narcolepsy—clinical spectrum, aetiopathophysiology, diagnosis and treatment. Nat. Rev. Neurol. 2019, 15, 519–539. [Google Scholar] [CrossRef] [PubMed]
  66. Barateau, L.; Lopez, R.; Dauvilliers, Y. Clinical neurophysiology of CNS hypersomnias. Handb. Clin. Neurol. 2019, 161, 353–367. [Google Scholar] [PubMed]
  67. Mahoney, C.E.; Cogswell, A.; Koralnik, I.J.; Scammell, T.E. The neurobiological basis of narcolepsy. Nat. Rev. Neurosci. 2019, 20, 83–93. [Google Scholar] [CrossRef]
  68. Inutsuka, A.; Inui, A.; Tabuchi, S.; Tsunematsu, T.; Lazarus, M.; Yamanaka, A. Concurrent and robust regulation of feeding behaviors and metabolism by orexin neurons. Neuropharmacology 2014, 85, 451–460. [Google Scholar] [CrossRef] [Green Version]
  69. Barson, J.R.; Leibowitz, S.F. Orexin/Hypocretin System: Role in Food and Drug Overconsumption. Int. Rev. Neurobiol. 2017, 136, 199–237. [Google Scholar]
  70. Sakurai, T. Orexins and orexin receptors: Implication in feeding behavior. Regul. Pept. 1999, 85, 25–30. [Google Scholar] [CrossRef]
  71. Muschamp, J.W.; Hollander, J.A.; Thompson, J.L.; Voren, G.; Hassinger, L.C.; Onvani, S.; Kamenecka, T.M.; Borgland, S.L.; Kenny, P.J.; Carlezon, W.A. Hypocretin (orexin) facilitates reward by attenuating the antireward effects of its cotransmitter dynorphin in ventral tegmental area. Proc. Natl. Acad. Sci. USA 2014, 111, E1648–E1655. [Google Scholar] [CrossRef] [Green Version]
  72. Gentile, T.A.; Simmons, S.J.; Watson, M.N.; Connelly, K.L.; Brailoiu, E.; Zhang, Y.; Muschamp, J.W. Effects of Suvorexant, a Dual Orexin/Hypocretin Receptor Antagonist, on Impulsive Behavior Associated with Cocaine. Neuropsychopharmacology 2018, 43, 1001–1009. [Google Scholar] [CrossRef] [Green Version]
  73. Wiskerke, J.; James, M.H.; Aston-Jones, G. The orexin-1 receptor antagonist SB-334867 reduces motivation, but not inhibitory control, in a rat stop signal task. Brain Res. 2020, 1731, 146222. [Google Scholar] [CrossRef]
  74. Wayner, M.J.; Armstrong, D.L.; Phelix, C.F.; Oomura, Y. Orexin-A (Hypocretin-1) and leptin enhance LTP in the dentate gyrus of rats in vivo. Peptides 2004, 25, 991–996. [Google Scholar] [CrossRef] [PubMed]
  75. Zhao, X.; Zhang, R.; Tang, S.; Ren, Y.; Yang, W.; Liu, X.; Tang, J. Orexin-A-induced ERK1/2 activation reverses impaired spatial learning and memory in pentylenetetrazol-kindled rats via OX1R-mediated hippocampal neurogenesis. Peptides 2014, 54, 140–147. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, L.; Zou, B.; Xiong, X.; Pascual, C.; Xie, J.; Malik, A.; Xie, J.; Sakurai, T.; Xie, X. Hypocretin/orexin neurons contribute to hippocampus-dependent social memory and synaptic plasticity in mice. J. Neurosci. 2013, 33, 5275–5284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Borgland, S.L.; Chang, S.-J.; Bowers, M.S.; Thompson, J.L.; Vittoz, N.; Floresco, S.B.; Chou, J.; Chen, B.T.; Bonci, A. Orexin A/hypocretin-1 selectively promotes motivation for positive reinforcers. J. Neurosci. 2009, 29, 11215–11225. [Google Scholar] [CrossRef]
  78. Mahler, S.V.; Smith, R.J.; Moorman, D.E.; Sartor, G.C.; Aston-Jones, G. Multiple roles for orexin/hypocretin in addiction. Prog. Brain Res. 2012, 198, 79–121. [Google Scholar]
  79. Calipari, E.S.; Espana, R.A. Hypocretin/orexin regulation of dopamine signaling: Implications for reward and reinforcement mechanisms. Front. Behav. Neurosci. 2012, 6, 54. [Google Scholar] [CrossRef] [Green Version]
  80. Yeoh, J.W.; Campbell, E.J.; James, M.H.; Graham, B.A.; Dayas, C.V. Orexin antagonists for neuropsychiatric disease: Progress and potential pitfalls. Front. Neurosci. 2014, 8, 36. [Google Scholar] [CrossRef] [Green Version]
  81. Boutrel, B.; Steiner, N.; Halfon, O. The hypocretins and the reward function: What have we learned so far? Front. Behav. Neurosci. 2013, 7, 59. [Google Scholar] [CrossRef] [Green Version]
  82. Marchant, N.J.; Millan, E.Z.; McNally, G.P. The hypothalamus and the neurobiology of drug seeking. Cell Mol. Life Sci. 2012, 69, 581–597. [Google Scholar] [CrossRef]
  83. Matzeu, A.; Martin-Fardon, R. Targeting the orexin system for prescription opioid use disorder: Orexin-1 receptor blockade prevents oxycodone taking and seeking in rats. Neuropharmacology 2020, 164, 107906. [Google Scholar] [CrossRef]
  84. Porter-Stransky, K.A.; Bentzley, B.S.; Aston-Jones, G. Individual differences in orexin-I receptor modulation of motivation for the opioid remifentanil. Addict. Biol. 2017, 22, 303–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Mohammadkhani, A.; Fragale, J.E.; Pantazis, C.B.; Bowrey, H.E.; James, M.H.; Aston-Jones, G. Orexin-1 Receptor Signaling in Ventral Pallidum Regulates Motivation for the Opioid Remifentanil. J. Neurosci. 2019, 39, 9831–9840. [Google Scholar] [CrossRef] [PubMed]
  86. Mohammadkhani, A.; James, M.H.; Pantazis, C.B.; Aston-Jones, G. Persistent effects of the orexin-1 receptor antagonist SB-334867 on motivation for the fast acting opioid remifentanil. Brain Res. 2019, 1731, 146461. [Google Scholar] [CrossRef] [PubMed]
  87. Fragale, J.E.; Pantazis, C.B.; James, M.H.; Aston-Jones, G. The role of orexin-1 receptor signaling in demand for the opioid fentanyl. Neuropsychopharmacology 2019, 44, 1690–1697. [Google Scholar] [CrossRef]
  88. Smith, R.J.; Aston-Jones, G. Orexin/hypocretin 1 receptor antagonist reduces heroin self-administration and cue-induced heroin seeking. Eur. J. Neurosci. 2012, 35, 798–804. [Google Scholar] [CrossRef]
  89. Moorman, D.E.; James, M.H.; Kilroy, E.A.; Aston-Jones, G. Orexin/hypocretin neuron activation is correlated with alcohol seeking and preference in a topographically specific manner. Eur. J. Neurosci. 2016, 43, 710–720. [Google Scholar] [CrossRef] [Green Version]
  90. Espana, R.A.; Oleson, E.B.; Locke, J.L.; Brookshire, B.R.; Roberts, D.C.; Jones, S.R. The hypocretin-orexin system regulates cocaine self-administration via actions on the mesolimbic dopamine system. Eur. J. Neurosci. 2010, 31, 336–348. [Google Scholar] [CrossRef] [Green Version]
  91. James, M.H.; Mahler, S.V.; Moorman, D.E.; Aston-Jones, G. A Decade of Orexin/Hypocretin and Addiction: Where Are We Now? Curr. Top. Behav. Neurosci. 2017, 33, 247–281. [Google Scholar]
  92. Schmeichel, B.E.; Matzeu, A.; Koebel, P.; Vendruscolo, L.F.; Sidhu, H.; Shahryari, R.; Kieffer, B.L.; Koob, G.F.; Martin-Fardon, R.; Contet, C. Knockdown of hypocretin attenuates extended access of cocaine self-administration in rats. Neuropsychopharmacology 2018, 43, 2373–2382. [Google Scholar] [CrossRef] [Green Version]
  93. Smith, R.J.; See, R.E.; Aston-Jones, G. Orexin/hypocretin signaling at the orexin 1 receptor regulates cue-elicited cocaine-seeking. Eur. J. Neurosci. 2009, 30, 493–503. [Google Scholar] [CrossRef] [Green Version]
  94. Martin-Fardon, R.; Weiss, F. Blockade of hypocretin receptor-1 preferentially prevents cocaine seeking: Comparison with natural reward seeking. Neuroreport 2014, 25, 485–488. [Google Scholar] [CrossRef] [PubMed]
  95. Panlilio, L.V.; Schindler, C.W. Self-administration of remifentanil, an ultra-short acting opioid, under continuous and progressive-ratio schedules of reinforcement in rats. Psychopharmacology 2000, 150, 61–66. [Google Scholar] [CrossRef] [PubMed]
  96. Bentzley, B.S.; Fender, K.M.; Aston-Jones, G. The behavioral economics of drug self-administration: A review and new analytical approach for within-session procedures. Psychopharmacology 2013, 226, 113–125. [Google Scholar] [CrossRef] [PubMed]
  97. Bentzley, B.S.; Jhou, T.C.; Aston-Jones, G. Economic demand predicts addiction-like behavior and therapeutic efficacy of oxytocin in the rat. Proc. Natl. Acad. Sci. USA 2014, 111, 11822–11827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Moorman, D.E.; Aston-Jones, G. Orexin-1 receptor antagonism decreases ethanol consumption and preference selectively in high-ethanol--preferring Sprague--Dawley rats. Alcohol 2009, 43, 379–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Bentzley, B.S.; Aston-Jones, G. Orexin-1 receptor signaling increases motivation for cocaine-associated cues. Eur. J. Neurosci. 2015, 41, 1149–1156. [Google Scholar] [CrossRef]
  100. James, M.H.; Bowrey, H.E.; Stopper, C.M.; Aston-Jones, G. Demand elasticity predicts addiction endophenotypes and the therapeutic efficacy of an orexin/hypocretin-1 receptor antagonist in rats. Eur. J. Neurosci. 2019, 50, 2602–2612. [Google Scholar] [CrossRef]
  101. Humphries, M.D.; Prescott, T.J. The ventral basal ganglia, a selection mechanism at the crossroads of space, strategy, and reward. Prog. Neurobiol. 2010, 90, 385–417. [Google Scholar] [CrossRef]
  102. Root, D.H.; Melendez, R.I.; Zaborszky, L.; Napier, T.C. The ventral pallidum: Subregion-specific functional anatomy and roles in motivated behaviors. Prog. Neurobiol. 2015, 130, 29–70. [Google Scholar] [CrossRef] [Green Version]
  103. Smith, K.S.; Tindell, A.J.; Aldridge, J.W.; Berridge, K.C. Ventral pallidum roles in reward and motivation. Behav. Brain Res. 2009, 196, 155–167. [Google Scholar] [CrossRef] [Green Version]
  104. Stephenson-Jones, M. Pallidal circuits for aversive motivation and learning. Curr. Opin. Behav. Sci. 2019, 26, 82–89. [Google Scholar] [CrossRef]
  105. Plaza-Zabala, A.; Flores, A.; Maldonado, R.; Berrendero, F. Hypocretin/orexin signaling in the hypothalamic paraventricular nucleus is essential for the expression of nicotine withdrawal. Biol. Psychiatry 2012, 71, 214–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Moorman, D.E.; James, M.H.; Kilroy, E.A.; Aston-Jones, G. Orexin/hypocretin-1 receptor antagonism reduces ethanol self-administration and reinstatement selectively in highly-motivated rats. Brain Res. 2017, 1654 Pt A, 34–42. [Google Scholar] [CrossRef] [Green Version]
  107. Harris, G.C.; Aston-Jones, G. Activation in extended amygdala corresponds to altered hedonic processing during protracted morphine withdrawal. Behav. Brain Res. 2007, 176, 251–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Choi, D.L.; Davis, J.F.; Fitzgerald, M.E.; Benoit, S.C. The role of orexin-A in food motivation, reward-based feeding behavior and food-induced neuronal activation in rats. Neuroscience 2010, 167, 11–20. [Google Scholar] [CrossRef] [PubMed]
  109. Alcaraz-Iborra, M.; Carvajal, F.; Lerma-Cabrera, J.M.; Valor, L.M.; Cubero, I. Binge-like consumption of caloric and non-caloric palatable substances in ad libitum-fed C57BL/6J mice: Pharmacological and molecular evidence of orexin involvement. Behav. Brain Res. 2014, 272, 93–99. [Google Scholar] [CrossRef]
  110. Schmeichel, B.E.; Barbier, E.; Misra, K.K.; Contet, C.; E Schlosburg, J.; Grigoriadis, D.; Williams, J.P.; Karlsson, C.; Pitcairn, C.; Heilig, M.; et al. Hypocretin receptor 2 antagonism dose-dependently reduces escalated heroin self-administration in rats. Neuropsychopharmacology 2015, 40, 1123–1129. [Google Scholar] [CrossRef] [Green Version]
  111. Dimsdale, J.E.; Norman, D.; DeJardin, D.; Wallace, M.S. The effect of opioids on sleep architecture. J. Clin. Sleep Med. 2007, 3, 33–36. [Google Scholar]
  112. Peles, E.; Schreiber, S.; Adelson, M. Variables associated with perceived sleep disorders in methadone maintenance treatment (MMT) patients. Drug Alcohol Depend. 2006, 82, 103–110. [Google Scholar] [CrossRef]
  113. Wang, D.; Teichtahl, H. Opioids, sleep architecture and sleep-disordered breathing. Sleep Med. Rev. 2007, 11, 35–46. [Google Scholar] [CrossRef]
  114. Lewis, S.A.; Oswald, I.; Evans, J.I.; Akindele, M.O. Heroin and human sleep. Electroencephalogr. Clin. Neurophysiol. 1970, 28, 429. [Google Scholar] [CrossRef]
  115. Chakravorty, S.; Vandrey, R.G.; He, S.; Stein, M.D. Sleep Management among Patients with Substance Use Disorders. Med. Clin. N. Am. 2018, 102, 733–743. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Y.; van den Pol, A.N. Mu-opioid receptor-mediated depression of the hypothalamic hypocretin/orexin arousal system. J. Neurosci. 2008, 28, 2814–2819. [Google Scholar] [CrossRef] [PubMed]
  117. Cherrier, M.M.; Amory, J.K.; Ersek, M.; Risler, L.; Shen, D.D. Comparative cognitive and subjective side effects of immediate-release oxycodone in healthy middle-aged and older adults. J. Pain 2009, 10, 1038–1050. [Google Scholar] [CrossRef] [Green Version]
  118. Rosow, C.E. The clinical usefulness of agonist-antagonist analgesics in acute pain. Drug Alcohol Depend. 1987, 20, 329–337. [Google Scholar] [CrossRef]
  119. Adamantidis, A.R.; Zhang, F.; Aravanis, A.M.; Deisseroth, K.; de Lecea, L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature 2007, 450, 420–424. [Google Scholar] [CrossRef]
  120. Gotter, A.L.; Roecker, A.J.; Hargreaves, R.; Coleman, P.J.; Winrow, C.J.; Renger, J.J. Orexin receptors as therapeutic drug targets. Prog. Brain Res. 2012, 198, 163–188. [Google Scholar]
  121. Taheri, S.; Sunter, D.; Dakin, C.; Moyes, S.; Seal, L.; Gardiner, J.; Rossi, M.; Ghatei, M.; Bloom, S. Diurnal variation in orexin A immunoreactivity and prepro-orexin mRNA in the rat central nervous system. Neurosci. Lett. 2000, 279, 109–112. [Google Scholar] [CrossRef]
  122. Zeitzer, J.M.; Buckmaster, C.L.; Parker, K.J.; Hauck, C.M.; Lyons, D.M.; Mignot, E. Circadian and homeostatic regulation of hypocretin in a primate model: Implications for the consolidation of wakefulness. J. Neurosci. 2003, 23, 3555–3560. [Google Scholar] [CrossRef] [Green Version]
  123. Hagan, J.J.; Leslie, R.A.; Patel, S.; Evans, M.L.; Wattam, T.A.; Holmes, S.; Benham, C.D.; Taylor, S.G.; Routledge, C.; Hemmati, P.; et al. Orexin A activates locus coeruleus cell firing and increases arousal in the rat. Proc. Natl. Acad. Sci. USA 1999, 96, 10911–10916. [Google Scholar] [CrossRef] [Green Version]
  124. Mieda, M.; Willie, J.T.; Hara, J.; Sinton, C.M.; Sakurai, T.; Yanagisawa, M. Orexin peptides prevent cataplexy and improve wakefulness in an orexin neuron-ablated model of narcolepsy in mice. Proc. Natl. Acad. Sci. USA 2004, 101, 4649–4654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Sasaki, K.; Suzuki, M.; Mieda, M.; Tsujino, N.; Roth, B.; Sakurai, T. Pharmacogenetic modulation of orexin neurons alters sleep/wakefulness states in mice. PLoS ONE 2011, 6, e20360. [Google Scholar] [CrossRef] [PubMed]
  126. Saper, C.B. The neurobiology of sleep. Continuum 2013, 19, 19–31. [Google Scholar] [CrossRef] [PubMed]
  127. Scammell, T.E.; Arrigoni, E.; Lipton, J.O. Neural Circuitry of Wakefulness and Sleep. Neuron 2017, 93, 747–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Gotter, A.L.; Forman, M.S.; Harrell, C.M.; Stevens, J.; Svetnik, V.; Yee, K.L.; Li, X.; Roecker, A.J.; Fox, S.V.; Tannenbaum, P.L.; et al. Orexin 2 Receptor Antagonism is Sufficient to Promote NREM and REM Sleep from Mouse to Man. Sci. Rep. 2016, 6, 27147. [Google Scholar] [CrossRef] [Green Version]
  129. Jacobson, L.H.; Chen, S.; Mir, S.; Hoyer, D. Orexin OX2 Receptor Antagonists as Sleep Aids. Curr. Top. Behav. Neurosci. 2017, 33, 105–136. [Google Scholar]
  130. James, M.H.; Fragale, J.E.; Aurora, R.N.; Cooperman, N.A.; Langleben, D.D.; Aston-Jones, G. Repurposing the dual orexin receptor antagonist suvorexant for the treatment of opioid use disorder: Why sleep on this any longer? Neuropsychopharmacology 2020, 45, 717–719. [Google Scholar] [CrossRef]
  131. Campbell, E.J.; Norman, A.; Bonomo, Y.; Lawrence, A.J. Suvorexant to treat alcohol use disorder and comorbid insomnia: Plan for a phase II trial. Brain Res. 2020, 1728, 146597. [Google Scholar] [CrossRef]
  132. Gotter, A.L.; Winrow, C.J.; Brunner, J.; Garson, S.L.; Fox, S.V.; Binns, J.; Harrell, C.M.; Cui, D.; Yee, K.L.; Stiteler, M.; et al. The duration of sleep promoting efficacy by dual orexin receptor antagonists is dependent upon receptor occupancy threshold. BMC Neurosci. 2013, 14, 90. [Google Scholar] [CrossRef] [Green Version]
  133. Sun, H.; Kennedy, W.P.; Wilbraham, D.; Lewis, N.; Calder, N.; Li, X.; Ma, J.; Yee, K.L.; Ermlich, S.; Mangin, E.; et al. Effects of suvorexant, an orexin receptor antagonist, on sleep parameters as measured by polysomnography in healthy men. Sleep 2013, 36, 259–267. [Google Scholar] [CrossRef] [Green Version]
  134. Bayerlein, K.; Kraus, T.; Leinonen, I.; Pilniok, D.; Rotter, A.; Hofner, B.; Schwitulla, J.; Sperling, W.; Kornhuber, J.; Biermann, T. Orexin A expression and promoter methylation in patients with alcohol dependence comparing acute and protracted withdrawal. Alcohol 2011, 45, 541–547. [Google Scholar] [CrossRef] [PubMed]
  135. Von Der Goltz, C.; Koopmann, A.; Dinter, C.; Richter, A.; Grosshans, M.; Fink, T.; Wiedemann, K.; Kiefer, F. Involvement of orexin in the regulation of stress, depression and reward in alcohol dependence. Horm. Behav. 2011, 60, 644–650. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of the effect of OrxR blockade on oxycodone-related behaviors.
Table 1. Summary of the effect of OrxR blockade on oxycodone-related behaviors.
OXYCODONEProcedureSpeciesSexSORAs
OrxR1
SORAs
OrxR2
DORAs
Intakeself-administrationratmalent
Reinstatementcue-inducedratmalent
Data from [83]; ↓ = decrease; = no effect; nt = not tested.
Table 2. Summary of the effect of OrxR blockade on remifentanil-related behaviors.
Table 2. Summary of the effect of OrxR blockade on remifentanil-related behaviors.
REMIFENTANILProcedureSpeciesSexSORAs
OrxR1
SORAs
OrxR2
DORAs
Intakeself-administrationratmalentnt
Reinstatementcue-inducedratmalentnt
drug-inducedratmalentnt
Data from [84,85,86]; ↓ = decrease; = no effect; nt = not tested.
Table 3. Summary of the effect of OrxR blockade on fentanyl-related behaviors.
Table 3. Summary of the effect of OrxR blockade on fentanyl-related behaviors.
FENTANYLProcedureSpeciesSexSORAs
OrxR1
SORAs
OrxR2
DORAs
Intakeself-administrationratmalentnt
Reinstatementcue-inducedratmalentnt
Data from [87]; ↓ = decrease; nt = not tested.

Share and Cite

MDPI and ACS Style

Matzeu, A.; Martin-Fardon, R. Targeting the Orexin System for Prescription Opioid Use Disorder. Brain Sci. 2020, 10, 226. https://doi.org/10.3390/brainsci10040226

AMA Style

Matzeu A, Martin-Fardon R. Targeting the Orexin System for Prescription Opioid Use Disorder. Brain Sciences. 2020; 10(4):226. https://doi.org/10.3390/brainsci10040226

Chicago/Turabian Style

Matzeu, Alessandra, and Rémi Martin-Fardon. 2020. "Targeting the Orexin System for Prescription Opioid Use Disorder" Brain Sciences 10, no. 4: 226. https://doi.org/10.3390/brainsci10040226

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop