You are on page 1of 6

Neuroscience Letters 735 (2020) 135177

Contents lists available at ScienceDirect

Neuroscience Letters
journal homepage: www.elsevier.com/locate/neulet

Research article

The antagonistic activity profile of naloxone in μ-opioid receptor agonist- T


induced psychological dependence
Atsushi Nakamuraa, Kana Yasufukub, Shinji Shimadaa, Hiroyuki Aritomia, Youko Furuea,
Hiroki Chibaa, Mami Muramotoa, Kenji Takaseb, Katsumi Koikeb, Tomoko Matsumotoc,
Tomoka Shimadac, Ryosuke Watarid, Takanobu Matsuzakid, Toshiyuki Asakib,
Toshiyuki Kanemasab, Masahide Fujitab,*
a
Research Area for Pharmacological Evaluation, Shionogi TechnoAdvance Research Co., Ltd, 1-1, 3-chome, Futaba-cho, Toyonaka, 561-0825, Osaka, Japan
b
Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, 561-0825, Osaka, Japan
c
Research Area for Candidate Selection, Shionogi TechnoAdvance Research Co., Ltd, 1-1, 3-chome, Futaba-cho, Toyonaka, 561-0825, Osaka, Japan
d
Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, 561-0825, Osaka, Japan

A R T I C LE I N FO A B S T R A C T

Keywords: Naloxone is a μ-opioid receptor antagonist that has been used to prevent overdose-related respiratory depression
Competitive antagonist and deaths by the illicit use of opioids. Naloxone can also deter the abuse potential of opioids, but little has been
Conditioned place preference reported regarding its antagonistic activity profile against opioid-induced psychological dependence. This study
Dopamine release aimed to confirm the antagonistic activity profile of naloxone against several μ-opioid receptor agonists and
μ-Opioid receptor agonist
investigate whether naloxone could affect the psychological dependence induced by widely used μ-opioid re-
Naloxone
ceptor agonist, oxycodone. In the Guanosine-5’-o-(3-thio) triphosphate (GTPγS) binding assay, naloxone
(30−30,000 nM) inhibited the GTPγS binding induced by oxycodone, hydrocodone, morphine, and fentanyl. It
elicited parallel rightward shifts in the concentration-response curves, indicating that naloxone possessed a
competitive antagonistic activity profile against these μ-opioid receptor agonists. In the conditioned place
preference test, oxycodone (0.01−1 mg/kg, i.v.) produced dose-dependent increases in place preference. The
increased place preference induced by oxycodone (1 mg/kg) was significantly attenuated by co-administration of
naloxone at a dose of 0.5 mg/kg but not 0.01 mg/kg. Naloxone (0.5 mg/kg, i.v.) also blocked oxycodone (1 mg/
kg)-induced dopamine release in nucleus accumbens; however, at a lower dose (0.01 mg/kg), it did not affect the
intrinsic dopamine release by oxycodone. These results indicate that the psychological dependence of oxycodone
could be antagonized by naloxone, depending on the dose. This characterization might lead to a better under-
standing of the competitive antagonistic activity profile of naloxone for μ-opioid receptor in the brain.

1. Introduction intravenous (i.v.) administration is most effective in many fatal cases


because of rapid onset of action that can rescue overdose-related re-
Clinically, several μ-opioid receptor agonists such as oxycodone, spiratory depression [4].
hydrocodone, morphine, and fentanyl have been used to manage Some environmental and drug-related factors contribute to over-
moderate to severe pain. The clinical use of opioids is often accom- dose-related respiratory depression and deaths, but the potential for
panied by many side effects including emesis, constipation, and seda- abuse associated with opioid misuse and addiction is considered to be
tion [1]; the increased rate of overdose-related respiratory depression the leading cause [5]. Opioid abusers often use i.v. administration route
and deaths caused by the illicit use of μ-opioid receptor agonists is one to attain a larger magnitude of euphoria and many abuse-deterrent
of the most serious issues that not only impacts the public health ne- approaches including formulating agonist/ antagonist combinations
gatively but also increases economic burden [2]. Naloxone is an an- [6], adding aversive agents [7], modifying the delivery system [8], and
tagonist with a high binding affinity for the μ-opioid receptor [3] and developing prodrugs [9] have been challenged. Of these approaches, a
has been used to prevent such fatal incidents. Several routes adminis- recent study on the human potential for abuse raised the possibility that
tration of naloxone have been developed for clinical use, but its i.v. co-administration of oxycodone and naloxone (1:0.5 ratio) could


Corresponding author at: Neuroscience Laboratory for Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan.
E-mail address: masahide.fujita@shionogi.co.jp (M. Fujita).

https://doi.org/10.1016/j.neulet.2020.135177
Received 18 May 2020; Received in revised form 11 June 2020; Accepted 15 June 2020
Available online 20 June 2020
0304-3940/ © 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
A. Nakamura, et al. Neuroscience Letters 735 (2020) 135177

produce lesser euphoria than that of oxycodone [10]. This is a rational Seisakusyo Co., Ltd., Tokyo, Japan) that was made of acrylic resin
pharmacological challenge, but little has been reported about the an- board and divided into two equal-sized compartments [14]. One com-
tagonist activity profile of naloxone and its appropriate ratio to be used partment was white with a textured floor and the other was black with
in preventing opioid abuse. a smooth floor to create equally preference compartments. The place
For many decades, the conditioned place preference paradigm has conditioning paradigm consisted of three phases (the pre-conditioning
been used to evaluate the psychological dependence related to opioid test, drug conditioning and the post-conditioning test). In the pre-
abuse in rodents [11]. A dose of μ-opioid receptor agonists causes place conditioning test, the rats that had not been treated with either drugs or
preference by activating the mesolimbic dopamine system, which is saline were then placed on the platform. The time spent in each com-
projected from the ventral tegmental area to the nucleus accumbens partment during a 900 s session was recorded. Conditioning sessions (4
(NAC) [12]. Thus, the concentration of dopamine released in NAC is days for drugs, 4 days for saline) were conducted once daily for 8 days.
considered to be one of the critical parameters in the brain to estimate Immediately after i.v. administration of oxycodone and naloxone, these
the psychological dependence induced by μ-opioid receptor agonists. rats were placed in the compartment opposite that in which they had
This study aimed to investigate the ability of naloxone to prevent μ- spent the most time in the preconditioning test for 60 min. On alter-
opioid receptor agonist- induced psychological dependence. Of these native days, these animals received saline and were placed in the other
agonists, oxycodone was consumed the most globally from 2000 to compartment for 60 min. On the day after the final conditioning ses-
2014 [13], and was thus selected for this study. Here, we have revealed sion, a postconditioning test that was identical to the preconditioning
that naloxone can inhibit oxycodone-induced increases in place pre- test was performed.
ference and dopamine release, indicating the antagonism of naloxone
against oxycodone-induced psychological dependence. 2.5. In vivo microdialysis study and quantification of dopamine

2. Materials and methods The rat in vivo microdialysis study and quantification of dopamine
in NAC were performed. Before implantation of a cannula, the rats were
2.1. Drugs anesthetized with inhaled isoflurane and/or subcutaneous administra-
tion of three types of anesthesia mixture (15 mg/kg alphaxanm,
Naloxone hydrochloride (naloxone) was obtained from Tocris 0.15 mg/kg medetomidine, and 2.5 mg/kg butorphanol) for surgery. A
Bioscience (Bristol, UK). Oxycodone hydrochloride (oxycodone) and guide cannula (AG-8; Eicom, Kyoto, Japan) was implanted into the NAC
morphine hydrochloride (morphine) were obtained from Shionogi & in the anesthetized rats (from the bregma: anterior, +4.0 mm; lateral,
Co., Ltd. (Osaka, Japan). Hydrocodone bitartrate (hydrocodone) and −0.8 mm; ventral, −6.8 mm; angle of 16 degrees) according to the
fentanyl citrate (fentanyl) were obtained from Mallinckrodt Inc. atlas of Paxinos and Watson and fixed to the skull with cranioplastic
(Hobart, NY, USA). All drugs were dissolved in dimethyl sulfoxide cement. Two to five days after surgery, microdialysis probes (A-I-8-02,
(Nacalai Tesque, Inc., Kyoto, Japan) and 0.9 % physiological saline 2-mm membrane length; Eicom) were slowly inserted into the NAC
(Otsuka Pharmaceutical Factory, Inc., Tokushima, Japan) for in vitro through guide cannulas under anesthesia, and the rats were placed in
and in vivo experiments, respectively. For in vivo experiments, drugs experimental cages. The probes were perfused continuously (2 mL/min)
were administered intravenously at a volume of 2 mL/kg. In the an- with artificial cerebrospinal fluid: 0.9 mM MgCl2, 147.0 mM NaCl,
tagonist study, 1–50 % of naloxone to oxycodone was used as a dose 4.0 mM KCl, and 1.2 mM CaCl2. Outflow fractions were collected every
ratio (1:0.01, 1:0.1, and 1:0.5 of oxycodone and naloxone). In addition, 20 min. After 3 baseline fractions were collected from the rat NAC, rats
the molecular weight of oxycodone hydrochloride (351.82 g/mol) and were given drugs or saline. Dialysis samples were collected for 60 min
naloxone hydrochloride (363.84 g/mol) was used to calculate a molar after treatment and analyzed by high-performance liquid chromato-
ratio of active pharmaceutical ingredients (1:0.0097, 1:0.097, and graphy (Eicom) with electrochemical detection (Eicom). Dopamine was
1:0.48 of oxycodone and naloxone). separated by column chromatography, and identified and quantified by
the use of standards, as described previously [15].
2.2. Animals
2.6. Measurement of plasma and brain concentrations
This study used 160 male Sprague Dawley rats weighing 180−240 g
(Charles River Laboratories Japan, Inc., Kanagawa, Japan). The rats The concentrations of oxycodone and naloxone in the plasma and
were housed in a room maintained at 23 °C ± 1 °C under a 12-h light/ brain were measured by liquid chromatography with tandem mass
dark cycle with ad libitum access to food and water. Each rat was used spectrometry method. The rats were given with i.v. administration of
only once. All procedures for the animal experiments were approved by oxycodone or naloxone. Blood samples and whole brain without cere-
the Animal Care and Use Committee of Shionogi Research Laboratories bellum samples were collected from an indwelling arterial cannula at 5,
(Osaka, Japan) and were performed according to the guidelines of the 15, and 60 min after drug administration, and transferred into the ice-
Association for Assessment and Accreditation of Laboratory Animal cold polypropylene tubes. The blood samples were centrifuged at 3000
Care International (AAALAC) guidelines. × g for 10 min at 4 °C. The brain samples were homogenized in saline
solution. The obtained plasma samples and brain samples were stored
2.3. [35S]-GTPγS binding assay frozen at −80 °C until the assay. The concentrations in the plasma and
brain samples were determined according to the similar method de-
The antagonism of naloxone against recombinant human μ-opioid scribed previously [16].
receptor (PerkinElmer Life and Analytical Sciences, Inc., Kanagawa,
Japan) was measured by a functional [35S]-GTPγS assay, as previously 2.7. Statistical analysis
described [3]. In this assay, [35S]-GTPγS bindings of μ-opioid receptor
agonists, oxycodone, hydrocodone, morphine and fentanyl, were de- The data are expressed as mean ± standard error of the mean
termined in absence and presence of naloxone. (SEM). GraphPad Prism 6.0 software (San Diego, CA, USA) was used to
perform the statistical analyses. The statistical significance of differ-
2.4. Conditioned place preference test ences among groups was assessed by a one- or two-way analysis of
variance (ANOVA), followed by Dunnett’s multiple comparison test. A
A conditioned place preference study was conducted using a shuttle probability value (p) of < 0.05 was considered to be statistically sig-
box (1460 mm × 450 mm × 600 mm, w × l × h; KN-80; Natsume nificant. The pharmacokinetic parameters of oxycodone and naloxone

2
A. Nakamura, et al. Neuroscience Letters 735 (2020) 135177

Fig. 1. Naloxone antagonism against oxycodone, hydrocodone, morphine, and fentanyl at human μ-opioid receptors as indicated by the [35S]-GTPγS binding assay.
Concentration-response curves of oxycodone (A), hydrocodone (B), morphine (C), and fentanyl (D) with different concentrations of naloxone in the binding of [35S]-
GTPγS to membranes containing the human μ-opioid receptor. Data are presented as the mean and SEM for 3 independent experiments.

were calculated using noncompartmental analysis module in Phoenix 0.01 mg/kg naloxone) did not alter place preferences compared with
WinNonlin software (version 8.1; Certara L.P., Princeton, NJ) based on that of oxycodone alone, but the 1:0.5 combination (1 mg/kg oxyco-
a non-compartment model analysis with uniform weighting. done + 0.5 mg/kg naloxone) significantly reduced the place preference
(Fig. 2B; p = 0.0178 oxycodone 1 mg/kg vs oxycodone 1 mg/kg +
3. Results naloxone 0.5 mg/kg). To understand the underlying mechanism in-
volved in changes in psychological dependence, we also evaluated the
3.1. The antagonistic activity profile of naloxone against several μ-opioid effect of naloxone on dopamine release in NAC. Oxycodone (1 mg/kg)
receptor agonists significantly increased the dopamine release as compared with that of
the vehicle, which was dose-dependently reduced by the co-adminis-
At first, the antagonistic activity profile of naloxone against several tration of naloxone (Fig. 2C; two-way ANOVA, p < 0.0001, vehicle vs
μ-opioid receptor agonists was examined by a functional GTPγS assay. oxycodone 1 mg/kg; Dunnett's post-hoc test, p < 0.0001, vehicle vs
Naloxone (30−30,000 nM) inhibited the opioid-induced GTPγS oxycodone, 1 mg/kg at 6–60 min post-administration). The 1:0.01
binding and elicited parallel rightward shifts in the concentration-re- combination (1 mg/kg oxycodone + 0.01 mg/kg naloxone) did not
sponse curves of oxycodone (Fig. 1A), hydrocodone (Fig. 1B), morphine produce any changes as compared with that in oxycodone alone,
(Fig. 1C), and fentanyl (Fig. 1D). whereas the 1:0.1 mixture (1 mg/kg oxycodone + 0.1 mg/kg naloxone)
and 1:0.5 combination (1 mg/kg oxycodone + 0.5 mg/kg naloxone)
3.2. Effect of naloxone on oxycodone-induced place preference and reduced the dopamine release significantly (two-way ANOVA,
dopamine release in NAC p = 0.0029, oxycodone 1 mg/kg vs oxycodone 1 mg/kg + naloxone
0.1 mg/kg; Dunnett's post-hoc test, p < 0.0001, vehicle vs oxycodone 1
Next, we used oxycodone as the most commonly consumed μ-opioid mg/kg, at 6–48 min post-administration; two-way ANOVA, p < 0.0001,
receptor agonist and investigated naloxone antagonism on psycholo- oxycodone 1 mg/kg vs oxycodone 1 mg/kg + naloxone 0.5 mg/kg;
gical dependence in rats. Oxycodone (0.01−1 mg/kg) produced a dose- Dunnett's post-hoc test, p < 0.0001, vehicle vs oxycodone 1 mg/kg at
dependent increase in the preference for drug-associated place times; 6–60 min post-administration).
oxycodone at 0.3 mg/kg and 1 mg/kg increased the place preference
significantly as compared with that of the vehicle (Fig. 2A; p = 0.0001 3.3. The plasma and brain concentrations of co-administered oxycodone
vehicle vs oxycodone 0.3 mg/kg, p = 0.0028 vehicle vs oxycodone 1 and naloxone
mg/kg). Based on these results, we used 1 mg/kg of oxycodone as an
adequate evaluation dose for further experiments. In the conditioned When measuring the plasma concentration after co-administration
place preference study, the 1:0.01 combination (1 mg/kg oxycodone + with oxycodone (1 mg/kg) and naloxone (0.01−0.5 mg/kg), the

3
A. Nakamura, et al. Neuroscience Letters 735 (2020) 135177

Fig. 2. Effects of naloxone on oxycodone-in-


duced place preference and dopamine release
in the NAC.
Rats were placed and conditioned in either
compartment for 60 min after intravenous ad-
ministration of oxycodone (0.03–1 mg/kg) (A),
or co-administration of oxycodone (1 mg/kg)
and naloxone (0.01 or 0.5 mg/kg) (B). The
preference for drug paired place was calculated
as the mean difference in times spent in the
compartment between the post-conditioning
test and the pre-conditioning test in each group
[14]. Data are presented as the mean and SEM
for 10–21 rats (A, B). **p < 0.01 or ***p <
0.001 vs. the vehicle-treated group, and *p <
0.05 vs. the vehicle-treated group with oxyco-
done (one-way ANOVA, Dunnett's post hoc
test). For in vivo microdialysis experiments, the
rats were intravenously administered vehicle,
oxycodone, or naloxone at time 0 (C). Dopa-
mine release (%) was calculated as the per-
centages of the corresponding baseline levels,
and data are represented as the mean and SEM
for 7 or 8 rats. F(4, 60) = 14.85, ###p < 0.001
vs oxycodone (1 mg/kg) (two-way ANOVA,
Dunnett's post-hoc test).

highest plasma concentrations of oxycodone and naloxone were ob- reported that a dopamine D3 antagonist suppresses oxycodone (3 mg/
served at 5 min post-administration as compared with those at 15 min kg, i.p.)-induced place preference [20]. The relationship between oxy-
and 60 min, respectively. Afterward, the concentrations gradually de- codone-induced dopamine release and place preference might be par-
creased (Fig. 3A and B). The ratio of the plasma and brain concentra- tially predictable, but there is little evidence regarding the effect of
tions of oxycodone to naloxone did not change among the cotreatment naloxone in the oxycodone-induced place preference, and comparable
groups (Fig. 3C). The rat serum protein binding rates of oxycodone and experimental protocols including administration route and dose have
naloxone were 18.9 % and 61.0 %, respectively. not been used to fully clarify this. The present study shows that oxy-
codone-induced place preference could be dose-dependently inhibited
4. Discussion by naloxone. Administering a combination of oxycodone and naloxone
at a dose ratio of 1:0.5 (molar ratio of 1:0.48), but not 1:0.01 (molar
There have been many non-clinical reports regarding the pharma- ratio of 1:0.0097), did not produce any preference for drug-associated
cological effects of naloxone against the μ-opioid receptor agonists such place times. This finding is partly consistent with the results on drug
as DAMGO and morphine [17,18]. However, the antagonistic activity discrimination paradigms in mice [21]. In the microdialysis study, we
profile of naloxone for clinically used μ-opioid receptor agonists has not revealed that co-administration of naloxone (0.01−0.5 mg/kg) reduced
been revealed. In this study, naloxone dose-dependently ameliorated the oxycodone-induced dopamine release in NAC dose-dependently,
the increased GTPγS activation by several μ-opioid receptor agonists and a combination dose ratio of 1:0.5, but not 1:0.1 and 1:0.01, of
including oxycodone, fentanyl, and hydrocodone. A previous study oxycodone and naloxone did not cause the dopamine release. These
reported that the Schild slope of naloxone is 1.0 based on Schild re- results show that 50 % administration dose ratio of naloxone to oxy-
gression analysis [3], indicating that naloxone is a competitive an- codone sufficiently reduces the intrinsic oxycodone-induced psycholo-
tagonist against these μ-opioid receptor agonists. gical dependence, and a ratio of lower than 10 % may not be effective.
A previous report indicates that oxycodone (0.5 mg/kg, i.v.)-in- A previously reported clinical study revealed a similar antagonism by
duced dopamine release is abolished by naloxone (3 mg/kg, i.v.) in the 50 % dose ratio of naloxone to oxycodone in opioid-experienced drug
nucleus accumbens [19], suggesting that oxycodone increases the do- users [10]. In another study with partial μ-opioid receptor agonist bu-
pamine release via μ-opioid receptor activation. Another research group prenorphine, 25 % dose ratio of naloxone also reduced the abuse

4
A. Nakamura, et al. Neuroscience Letters 735 (2020) 135177

Fig. 3. Plasma and brain concentrations after


co-administration with naloxone and oxyco-
done in rats.
Rats were intravenously administered oxyco-
done (1 mg/kg) and naloxone (0.01, 0.1, or
0.5 mg/kg). Blood samples were collected at 5,
15, and 60 min, and brain samples were col-
lected at 60 min after drug administration. The
time-course of the plasma concentration of
oxycodone (A) and naloxone (B), and the brain
concentrations (C) were measured according to
a previously described method (9). The dis-
tribution ratio between the brain and plasma is
expressed as a Kp brain value. Data are re-
presented as the mean ± SEM for three rats.

liability of buprenorphine [22], suggesting that an appropriate ratio of the concentration-response curve of oxycodone approximately 100-fold
naloxone could antagonize μ-opioid receptor agonist-induced psycho- to the right compared with that of oxycodone alone; however, 10 nM
logical dependence. This is the first report showing a mechanism-based naloxone did not greatly affect the curve. These findings are in line with
antagonistic activity profile of naloxone in psychological dependence. the observed antagonistic response of naloxone following administra-
Some groups have reported that oxycodone binds selectively to μ- tion of a 1:0.5, but not a 1:0.01 combination of oxycodone and na-
opioid receptor and oxycodone-induced antinociception is mediated by loxone.
the μ-opioid receptor [23–25]. Our previous study also revealed that In conclusion, we have found that naloxone exhibits a competitive
the increased GTPγS binding by oxycodone was completely antagonized antagonistic activity profile for widely prescribed μ-opioid receptor
by a μ-opioid receptor antagonist β-funaltrexamine [26], indicating that agonists and inhibits the opioid-induced pharmacological effects in the
oxycodone produces pharmacological effects by mainly acting on the μ- brain. Although the respective pharmacological effects observed in this
opioid receptor. Another group reported the possible involvement of κ- study do not directly represent the clinical pharmacological effects, our
opioid receptor in its antinociception [27], but an inadequate dose of results provide substantial evidence for the clinical use of naloxone to
nor-binaltorphimine, a κ-opioid receptor antagonist, might be used for manage adverse effects of opioids.
the pharmacological study. In the present study, oxycodone-induced
place preference and dopamine release were inhibited by naloxone Funding
(0.5 mg/kg) that could antagonize μ-opioid receptor selectively in rats
[28], suggesting that not only the antinociceptive effect but also the This research did not receive any specific grant from funding
psychological dependence of oxycodone are mediated via the μ-opioid agencies in the public, commercial, or not-for-profit sectors.
receptor. In addition, oxycodone exhibits various pharmacological ef-
fects depending on the plasma concentrations [16], but plasma con-
CRediT authorship contribution statement
centration of oxycodone (1 mg/kg) that associated with the psycholo-
gical dependence was 28.5 ng/ml in rats, which is almost similar to that
Atsushi Nakamura: Conceptualization, Formal analysis,
required for the antinociceptive effect (47.6 ng/mL). This also indicates
Validation, Writing - original draft. Kana Yasufuku:
that naloxone (0.5 mg/kg) might ameliorate the oxycodone (1 mg/kg)-
Conceptualization. Shinji Shimada: Investigation, Methodology.
induced antinociceptive effect.
Hiroyuki Aritomi: Investigation, Methodology. Youko Furue:
Pharmacokinetic studies revealed that both oxycodone and na-
Investigation, Methodology, Validation. Hiroki Chiba: Investigation,
loxone were not observed for their pharmacokinetic interaction. When
Methodology, Validation. Mami Muramoto: Investigation,
assuming free concentrations of oxycodone at 5 min (plasma
Methodology. Kenji Takase: Investigation, Methodology, Validation.
concentration × Kp brain × unbound fraction ratio), the estimated
Katsumi Koike: Investigation, Methodology. Tomoko Matsumoto:
concentration (≥ 2 μM) shows about 400 % binding in the GTPγS
Investigation, Methodology, Validation. Tomoka Shimada:
functional assay; this suggests that oxycodone at 1 mg/kg could pro-
Investigation, Methodology. Ryosuke Watari: Investigation,
duce sufficient brain-related pharmacological effects in rats. However,
Methodology. Takanobu Matsuzaki: Investigation, Methodology,
the estimated free concentrations of naloxone at 0.01 mg/kg and
Validation. Toshiyuki Asaki: Project administration. Toshiyuki
0.5 mg/kg were calculated to be approximately 10 nM and 300 nM,
Kanemasa: Project administration. Masahide Fujita: Project admin-
respectively. In the GTPγS functional assay, naloxone at 300 nM shifted
istration, Writing - review & editing.

5
A. Nakamura, et al. Neuroscience Letters 735 (2020) 135177

Declaration of Competing Interest S. Datta, T.R. Deer, B. Fellows, V. Galan, V. Grami, H. Hansen, S. Helm Ii, R. Justiz,
D. Koyyalagunta, Y. Malla, A. Navani, K.H. Nouri, R. Pasupuleti, N. Sehgal,
S.M. Silverman, T.T. Simopoulos, V. Singh, D.R. Solanki, P.S. Staats, R. Vallejo,
The authors declare no conflicts of interest. B.W. Wargo, A. Watanabe, J.A. Hirsch, Responsible, safe, and effective prescription
of opioids for chronic non-cancer pain: american society of interventional pain
physicians (ASIPP) guidelines, Pain Physician 20 (2017) S3–S92.
Acknowledgments [14] A. Nakamura, H. Ono, A. Ando, M. Hinata, K. Niidome, S. Omachi, G. Sakaguchi,
S. Shinohara, Suppression of the acute upregulation of phosphorylated-extracellular
All authors are employees of Shionogi & Co., Ltd, and Shionogi regulated kinase in ventral tegmental area by a μ-opioid receptor agonist is related
to resistance to rewarding effects in a mouse model of bone cancer, J. Pharmacol.
TechnoAdvance Research Co., Ltd, the manufacturer of oxycodone and Sci. 133 (2017) 9–17, https://doi.org/10.1016/j.jphs.2016.11.004.
morphine, and have complete access to the data that supports this [15] T. Mori, T. Kanbara, M. Harumiya, Y. Iwase, A. Masumoto, S. Komiya,
publication. We thank Trent Rogers, PhD, from Edanz Group (https:// A. Nakamura, M. Shibasaki, T. Kanemasa, G. Sakaguchi, T. Suzuki, Establishment of
opioid-induced rewarding effects under oxaliplatin- and Paclitaxel-induced neuro-
en-author-services.edanzgroup.com/) for editing a draft of this manu-
pathy in rats, J. Pharmacol. Sci. 126 (2014) 47–55, https://doi.org/10.1254/jphs.
script. 14134fp.
[16] A. Nakamura, M. Hasegawa, H. Ito, K. Minami, K. Koike, N. Habu-Tomita, K. Nanba,
Appendix A. Supplementary data K. Hamaguchi, E. Noshi, H. Hashimoto, I. Nishino, Y. Okabayashi, K. Koyabu,
T. Kihara, Y. Iwamoto, Y. Inoue, M. Narita, T. Suzuki, A. Kato, Distinct relations
among plasma concentrations required for different pharmacological effects in
Supplementary material related to this article can be found, in the oxycodone, morphine, and fentanyl, J. Pain Palliat. Care Pharmacother. 25 (2011)
online version, at doi:https://doi.org/10.1016/j.neulet.2020.135177. 318–334, https://doi.org/10.3109/15360288.2011.620689.
[17] A.E. Takemori, P.S. Portoghese, Comparative antagonism by naltrexone and na-
loxone of mu, kappa, and delta agonists, Eur. J. Pharmacol. 104 (1984) 101–104,
References https://doi.org/10.1016/0014-2999(84)90374-1.
[18] E. Floettmann, K. Bui, M. Sostek, K. Payza, M. Eldon, Pharmacologic profile of
naloxegol, a peripherally acting μ-opioid receptor antagonist, for the treatment of
[1] W.L. Hasler, L.A. Wilson, L.A. Nguyen, W.J. Snape, T.L. Abell, K.L. Koch,
opioid-induced constipation, J. Pharmacol. Exp. Ther. 361 (2017) 280–291,
R.W. McCallum, P.J. Pasricha, I. Sarosiek, G. Farrugia, M. Grover, L.A. Lee,
https://doi.org/10.1124/jpet.116.239061.
L. Miriel, J. Tonascia, F.A. Hamilton, H.P. Parkman, Gastroparesis Clinical Research
[19] C.M. Van der Weele, K.A. Porter-Stransky, O.S. Mabrouk, V. Lovic, B.F. Singer,
Consortium. Opioid use and potency are associated with clinical features, quality of
R.T. Kennedy, B.J. Aragona, Rapid dopamine transmission within the nucleus ac-
life, and use of resources in patients with gastroparesis, Clin. Gastroenterol.
cumbens: dramatic difference between morphine and oxycodone delivery, Eur. J.
Hepatol. 17 (2019) 1285–1294, https://doi.org/10.1016/j.cgh.2018.10.013.
Neurosci. 40 (2014) 3041–3054, https://doi.org/10.1111/ejn.12709.
[2] R.A. Rudd, P. Seth, F. David, L. Scholl, Increases in drug and opioid-involved
[20] V. Kumar, A. Bonifazi, M.P. Ellenberger, T.M. Keck, E. Pommier, R. Rais,
overdose deaths - United States, 2010–2015, MMWR Morb. Mortal. Wkly. Rep. 65
B.S. Slusher, E. Gardner, Z.B. You, Z.X. Xi, A.H. Newman, Highly selective dopa-
(2016) 1445–1452, https://doi.org/10.15585/mmwr.mm655051e1.
mine D3 receptor (D3R) antagonists and partial agonists based on eticlopride and
[3] T. Kanemasa, T. Koike, K. Takase, T. Arai, A. Nakamura, Y. Morioka, M. Hasegawa,
the D3R crystal structure: New leads for opioid dependence treatment, J. Med.
Pharmacological profile of naldemedine, a peripherally acting μ-opioid receptor
Chem. 25 (2016) 7634–7650, https://doi.org/10.1021/acs.jmedchem.6b00860.
antagonist: Comparison with naloxone and naloxegol, J. Pharmacol. Exp. Ther.
[21] D.M. Walentiny, J.M. Wiebelhaus, P.M. Beardsley, Nociceptin/orphanin FQ re-
(2020), https://doi.org/10.1124/jpet.119.264515 (in press).
ceptors modulate the discriminative stimulus effects of oxycodone in C57BL/6
[4] S.R. Mueller, A.Y. Walley, S.L. Calcaterra, J.M. Glanz, I.A. Binswanger, A review of
mice, Drug Alcohol Depend. 187 (2018) 335–342, https://doi.org/10.1016/j.
opioid overdose prevention and naloxone prescribing: implications for translating
drugalcdep.2018.02.035.
community programming into clinical practice, Subst. Abus. 36 (2015) 240–253,
[22] J.D. Jones, J.M. Manubay, S. Mogali, V.E. Metz, G. Madera, S. Martinez,
https://doi.org/10.1080/08897077.2015.1010032.
M. Mumtaz, S.D. Comer, Abuse liability of intravenous buprenorphine vs. bupre-
[5] L.R. Webster, Risk factors for opioid-use disorder and overdose, Anesth. Analg. 125
norphine/naloxone: importance of absolute naloxone amount, Drug Alcohol
(2017) 1741–1748, https://doi.org/10.1213/ANE.0000000000002496.
Depend. 179 (2017) 362–369, https://doi.org/10.1016/j.drugalcdep.2017.06.033.
[6] T.H. Tran, B.L. Griffin, R.H. Stone, K.M. Vest, T.J. Todd, Methadone, buprenor-
[23] K.K. Lemberg, V.K. Kontinen, A.O. Siiskonen, K.M. Viljakka, J.T. Yli-Kauhaluoma,
phine, and naltrexone for the treatment of opioid use disorder in pregnant women,
E.R. Korpi, E.A. Kalso, Antinociception by spinal and systemic oxycodone: why does
Pharmacotherapy 37 (2017) 824–839, https://doi.org/10.1002/phar.1958.
the route make a difference? In vitro and in vivo studies in rats, Anesthesiology 105
[7] C. Walter, C. Knothe, J. Lötsch, Abuse-deterrent opioid formulations: pharmacoki-
(2006) 801–812, https://doi.org/10.1097/00000542-200610000-00027.
netic and pharmacodynamic considerations, Clin. Pharmacokinet. 55 (2016)
[24] E.M. Peckham, J.R. Traynor, Comparison of the antinociceptive response to mor-
751–767, https://doi.org/10.1007/s40262-015-0362-3.
phine and morphine-like compounds in male and female Sprague-Dawley rats, J.
[8] M.E. Abreu, G.E. Bigelow, L. Fleisher, S.L. Walsh, Effect of intravenous injection
Pharmacol. Exp. Ther. 316 (2006) 1195–1201, https://doi.org/10.1124/jpet.105.
speed on responses to cocaine and hydromorphone in humans,
094276.
Psychopharmacology (Berl.) 154 (2001) 76–84, https://doi.org/10.1007/
[25] M. Narita, A. Nakamura, M. Ozaki, S. Imai, K. Miyoshi, M. Suzuki, T. Suzuki,
s002130000624.
Comparative pharmacological profiles of morphine and oxycodone under a neu-
[9] Y.H. Lee, D.L. Brown, H.Y. Chen, Current impact and application of abuse-deterrent
ropathic pain-like state in mice: evidence for less sensitivity to morphine,
opioid formulations in clinical practice, Pain Physician 20 (2017) E1003–E1023.
Neuropsychopharmacology 33 (2008) 1097–1112, https://doi.org/10.1038/sj.npp.
[10] S.V. Colucci, P.J. Perrino, M. Shram, C. Bartlett, Y. Wang, S.C. Harris, Abuse po-
1301471.
tential of intravenous oxycodone/naloxone solution in nondependent recreational
[26] A. Nakamura, M. Hasegawa, K. Minami, T. Kanbara, T. Tomii, A. Nishiyori,
drug users, Clin. Drug Investig. (2014) 421–429, https://doi.org/10.1007/s40261-
M. Narita, T. Suzuki, A. Kato, Differential activation of the μ-opioid receptor by
014-0192-3.
oxycodone and morphine in pain-related brain regions in a bone cancer pain model,
[11] J.M. van Ree, D. de Wied, Prolyl-leucyl-glycinamide (PLG) facilitates morphine
Br. J. Pharmacol. 168 (2013) 375–388, https://doi.org/10.1111/j.1476-5381.2012.
dependence, Life Sci. 19 (1976) 1331–1339, https://doi.org/10.1016/0024-
02139.x.
3205(76)90430-6.
[27] F.B. Ross, M.T. Smith, The intrinsic antinociceptive effects of oxycodone appear to
[12] M. Narita, M. Funada, T. Suzuki, Regulations of opioid dependence by opioid re-
be kappa-opioid receptor mediated, Pain 73 (1997) 151–157, https://doi.org/10.
ceptor types, Pharmacol. Ther. 89 (2001) 1–15, https://doi.org/10.1016/s0163-
1016/s0304-3959(97)00093-6.
7258(00)00099-1.
[28] K. Rutten, W. Schröder, T. Christoph, T. Koch, T.M. Tzschentke, Selectivity profiling
[13] L. Manchikanti, A.M. Kaye, N.N. Knezevic, H. McAnally, K. Slavin, A.M. Trescot,
of NOP, MOP, DOP and KOP receptor antagonists in the rat spinal nerve ligation
S. Blank, V. Pampati, S. Abdi, J.S. Grider, A.D. Kaye, K.N. Manchikanti, H. Cordner,
model of mononeuropathic pain, Eur. J. Pharmacol. 15 (2018) 41–48, https://doi.
C.G. Gharibo, M.E. Harned, S.L. Albers, S. Atluri, S.M. Aydin, S. Bakshi, R.L. Barkin,
org/10.1016/j.ejphar.2018.03.008.
R.M. Benyamin, M.V. Boswell, R.M. Buenaventura, A.K. Calodney, D.L. Cedeno,

You might also like