Accepted Manuscript Naltrexone and Nalmefene Attenuate Cocaine Place Preference in Male Mice
Kyle A. Windisch, Brian Reed, Mary Jeanne Kreek PII:
S0028-3908(18)30403-9
DOI:
10.1016/j.neuropharm.2018.07.025
Reference:
NP 7275
To appear in:
Neuropharmacology
Received Date:
22 March 2018
Accepted Date:
22 July 2018
Please cite this article as: Kyle A. Windisch, Brian Reed, Mary Jeanne Kreek, Naltrexone and Nalmefene Attenuate Cocaine Place Preference in Male Mice, Neuropharmacology (2018), doi: 10.1016/j.neuropharm.2018.07.025
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
TITLE: Naltrexone and Nalmefene Attenuate Cocaine Place Preference in Male Mice
AUTHORS: Kyle A Windischa, Brian Reeda, Mary Jeanne Kreeka
AFFILIATIONS: aLaboratory
of the Biology of Addictive Diseases, The Rockefeller University, 1230 York
Avenue, New York, NY 10065, USA
CORRESPONDING AUTHOR: Kyle A Windisch, PhD The Laboratory of the Biology of Addictive Diseases The Rockefeller University 1230 York Ave, Box #171 New York, NY 10065 Email:
[email protected]
Declarations of interest: none
ACCEPTED MANUSCRIPT
ABSTRACT: Cocaine addiction treatment is difficult due to the current lack of approved pharmacotherapuetics. Several preclinical and clinical studies have demonstrated that the mu opioid receptor (MOPr) antagonist/kappa opioid receptor (KOPr) partial agonist naltrexone (NTX) reduces the subjective effects and self-administration of cocaine. However, very limited research has examined the ability of the structurally similar MOPr antagonist/KOPr partial agonist nalmefene (NMF) to reduce cocaine reward. Here we examine the effect of low (1 mg/kg) and high (10 mg/kg) doses of NTX or NMF on cocaine place preference. In vivo characterization of these NTX and NMF doses were performed to examine their effectiveness at MOPr and KOPr. RESULTS: Both NTX doses and high dose NMF significantly reduced cocaine place preference. Conversely, a significant place avoidance was observed for high dose NTX and both NMF doses. Interestingly, neither NTX nor NMF blocked cocaine-induced hyperlocomotion. High dose NTX and both NMF doses fully blocked MOPr agonist morphine-induced thermal analgesia as well as KOPr agonist U50,488H-induced locomotor discoordination. However, low dose NTX fully blocked morphine analgesia but not U50,488H locomotor discoordination suggesting that low dose NTX is effective at MOPr but not KOPr. CONCLUSION: Both NTX and NMF block the place preference, but not locomotor activating, effects of cocaine. These results suggest that both NTX and NMF may be viable pharmacotheraputics for some aspects of cocaine addiction. This is an important step to understanding the potential mechanism(s) of action of NTX and NMF for the development of more efficacious pharmacological treatments for substance use disorders.
ACCEPTED MANUSCRIPT
1 2
1. INTRODUCTION Abuse and dependence of cocaine remains a major public health issue with
3
significant social and economic consequences. However, no targeted
4
pharmacotherapeutic option currently exists to combat cocaine addiction. Cocaine acts
5
primarily by increasing extracellular concentrations of dopamine as well as other
6
monoamines (serotonin and norepinephrine) in the brain mesocorticolimbic system (Ritz
7
et al., 1987). As the endogenous opioid system is highly expressed in key regions of the
8
mesocorticolimbic system (Mansour et al., 1987, 1988) and has been shown to regulate
9
monoamine signaling (Di Chiara and Imperato, 1988; Zhang et al., 2004a), the opioid
10
system may be involved in cocaine reinforcement and reward. Prior exposure of mu
11
opioid receptor (MOPr) agonists, such as morphine, have been shown to enhance the
12
reinforcing aspects of cocaine and vice versa, a phenomenon known as cross-
13
sensitization (Lett, 1989; Shippenberg and Heidbreder, 1995; Shippenberg et al., 1998).
14
Opioid receptor ligands, such as naloxone, block cocaine reward (Houdi et al., 1989;
15
Simmons and Self, 2009). The cross-sensitization between opioids and cocaine as well
16
as ability of opioid antagonists to block cocaine reward suggest common
17
neurobiological pathways for opiates and cocaine and, therefore, pharmacotherapies
18
currently approved for the treatment of opiate addiction may be efficacious in treating
19
cocaine addiction.
20
Naltrexone (NTX) is an FDA approved medication for the treatment of opioid use
21
disorder and alcoholism that has been shown in vitro to act as an antagonist at MOPr
22
and partial agonist at kappa opioid receptors (KOPr), albeit with a low maximal effect for
23
KOPr (Ghirmai et al., 2008; Wentland et al., 2009). Several clinical studies have
1
ACCEPTED MANUSCRIPT
24
demonstrated the potential viability of NTX for the treatment of cocaine addiction in both
25
reducing cocaine use as measured by cocaine-negative urine samples (Kosten et al.,
26
1989; Oslin et al., 1999; Pettinati et al., 2008; Schmitz et al., 2014; Schmitz et al., 2001)
27
and attenuating the subjective effects of cocaine (Comer et al., 2013; Kosten et al.,
28
1992; Sofuoglu et al., 2003) (however see also Hersh et al., 1998; Modesto-Lowe et al.,
29
1997; Schmitz et al., 2004; Schmitz et al., 2009; Walsh et al., 1996). Overall, the prior
30
clinical studies suggest that higher doses of NTX (150 mg/day) than what is typically
31
prescribed for the treatment of alcoholism (50 mg/day) may be necessary to regulate
32
the reinforcing aspects of cocaine in clinical populations, particularly populations with
33
polysubstance dependence (e.g., cocaine and alcohol co-dependence).
34
Several preclinical studies have examined the potential viability of NTX in
35
reducing the rewarding effects of cocaine. Bilsky et al. (1992) found that 56 mg/kg NTX
36
administered 4 hours prior to cocaine conditioning reduced but did not fully block the
37
rewarding effects of cocaine as measured by conditioned place preference (CPP).
38
Lower doses of NTX (2-3 mg/kg, sc) administered 10-15 minutes prior to cocaine fully
39
block cocaine place conditioning (Sala et al., 1995; Suzuki et al., 1992). As well, high
40
dose chronic NTX (120 and 240 mg but not 10 - 60 mg implant) administered via
41
osmotic pump fully blocks cocaine CPP (Mitchem et al., 1999). In preclinical studies of
42
alcohol exposure, doses of NTX as low as 0.1 - 0.3 mg/kg have been shown to reduce
43
seeking and operant self-administration of alcohol (Henderson-Redmond and
44
Czachowski, 2014; Williams and Broadbridge, 2009). Together with the clinical NTX
45
findings, these studies suggest that higher doses of NTX, potentially doses that act at
2
ACCEPTED MANUSCRIPT
46
both the KOP and MOP receptors, are necessary to block the rewarding effects of
47
cocaine.
48
Nalmefene (NMF) is a MOPr antagonist/KOPr partial agonist (Ghirmai et al.,
49
2008) structurally similar to that of NTX (methylene substitution at 6 position of NTX;
50
see figure 1) that is equipotent to NTX at MOPr and roughly two-fold more potent than
51
NTX at KOPr and delta opioid receptors (deHaven-Hudkins et al., 1990; Michel et al.,
52
1985). Though originally considered an antagonist for KOPr, NMF was recently shown
53
to act as a partial KOPr agonist both in vitro and in vivo (Bart et al., 2005; Stahl et al.,
54
2015). No published preclinical studies to date have examined the effect of NMF on
55
cocaine reward. Clinically, Grosshans et al. (2015) reported a reduction in cocaine
56
craving with “as needed” nalmefene in a female patient. This case report, along with the
57
structural and pharmacological similarity of NMF and NTX, suggests that NMF may
58
attenuate the rewarding aspects of cocaine; however, more experiments are needed to
59
clarify the ability of NMF to block cocaine reward.
60
Here we attempted to clarify the ability of the MOPr antagonists/KOPr partial
61
agonists naltrexone and nalmefene to regulate the rewarding aspects of cocaine. For
62
this we examined the ability of low (1 mg/kg) and high (10 mg/kg) doses of NTX or NMF
63
to block the rewarding and locomotor stimulating effects of cocaine using unbiased
64
place conditioning and open field assays. Further, we characterized the NTX and NMF
65
doses for in vivo effectiveness at the mu and kappa opioid receptors.
66 67
2. Materials and Methods
68
2.1 Subjects
3
ACCEPTED MANUSCRIPT
69
Adult male C57Bl/6J mice (10 weeks old on arrival; Jackson Laboratory, Bar
70
Harbor, ME) were housed in groups of four with free access to food and water in light
71
(12:12 hour light/dark cycle; lights on at 1900 for CPP, thermal analgesia, and rotarod
72
assays; lights on at 0700 locomotor activity assay) and temperature (22oC) controlled
73
rooms. For all experiments upon arrival animals were allowed 1 week to habituate to the
74
environment prior to the start of the testing. Animals were weighed and handled a
75
minimum of three times as well as received two separate mock injections to habituate to
76
experimental procedures. Animal care and experimental procedures were conducted
77
according to the National Institutes of Health guide for the care and use of Laboratory
78
animals (NIH Publications No. 8023, revised 1978). The experimental protocols used
79
were approved by the Institutional Animal Care and Use Committee of The Rockefeller
80
University.
81 82 83
2.2 Drugs The drugs used were cocaine hydrochloride (Sigma-Aldrich, St. Louis, MO),
84
morphine hydrochloride (Sigma-Aldrich, St. Louis, MO), U50,488H (Sigma-Aldrich, St.
85
Louis, MO), naltrexone hydrochloride (Tocris Bioscience, Minneapolis, MN), and
86
nalmefene hydrochloride (Baker Norton Pharmaceuticals, Miami, FL). All drugs were
87
dissolved in sterile saline and injected at a volume of 5 mL/kg body weight. Sterile
88
saline was used as vehicle treatment for all experiments.
89 90
2.3 Place Conditioning Apparatus
4
ACCEPTED MANUSCRIPT
91
The mouse place preference chambers have three distinct compartments
92
separated by removable doors (ENV-3013; Med Associate, VT). Movement within each
93
compartment is tracked by individual infrared photobeams on a photobeam strip (six
94
beams in the white and black compartments and two beams in the smaller central gray
95
compartment). The center compartment had a solid neutral gray floor and gray walls.
96
The black and white compartments (16.8 x 12.7 x 12.7 cm) had stainless steel rod and
97
mesh floors, respectively.
98 99 100
2.4 Place Conditioning Experiments were performed in a dimly-lit, sound attenuated chamber described
101
above. A 15 mg/kg dose of cocaine was selected based both on previous studies
102
(Bailey et al., 2012; Velazquez-Sanchez et al., 2010) and pilot work demonstrating that
103
this dose is sufficient to produce robust cocaine-induced CPP (data not shown).
104
During the pre-conditioning session (D1), adult mice (12 weeks at start;
105
n=8/group) were placed in the center compartment and allowed free access to all
106
compartments (see Figure 2a: timeline place conditioning experiments). The time spent
107
in each compartment was recorded for 30 min. During the conditioning sessions, mice
108
received two injections. Mice were first injected with either saline, NTX, or NMF 30
109
minutes prior to the conditioning session and returned to their home cage. Animals were
110
then injected with either saline or cocaine (15 mg/kg) and immediately placed into the
111
appropriate conditioning compartment for 30 min. Conditioning sessions occurred at the
112
same time each day (1000-1600 hours) with animals injected with drug (NTX or NMF +
113
cocaine) or saline (saline + saline) treatment on alternate days, for a total of eight
5
ACCEPTED MANUSCRIPT
114
conditioning sessions (four drug and four saline). Animals did not receive NTX or NMF
115
on saline conditioning days. The study used an unbiased design in which drug paired
116
compartment (white or black) was counterbalanced across animals. The post-
117
conditioning test session was performed on the day after the last conditioning session
118
(24 hours following final drug conditioning session) and was identical to the pre-
119
conditioning session. Each mouse had free access to all compartments for 30 minutes.
120
Place preference was calculated as percent preference for the drug paired chamber
121
during the post-test session. NTX and NMF were tested in separate experiments.
122 123 124
2.5 Open Field Mouse locomotor activity was assessed using an open field arena that consisted
125
of a standard rat shoebox cage within a locomotor activity monitoring frame (Kinder
126
Scientific, San Diego, CA). Distance traveled consisted of horizontal movement within
127
the arena and data were collected in 5-minute bins. Separate cohorts of drug naïve
128
adult male C57Bl/6J mice were used to examine the effect of low (1 mg/kg) dose and
129
high (10 mg/kg) NTX or NMF pretreatment on cocaine induced-hyperlocomotion and
130
locomotor sensitization (n=8/group). Animals were habituated to the locomotor boxes
131
during a single 30-minute session to reduce a potential novelty confound (see Figure
132
2b: timeline open field experiments). For locomotor sensitization the injection
133
pretreatment time, doses, and days (saline versus drug) were identical to those used for
134
the place conditioning experiments; however, separate groups were run for dose [i.e.,
135
one group with subgroups pretreated with low dose (1 mg/kg) NTX or NMF and another
136
group with subgroups pretreated with high dose (10 mg/kg) NTX or NMF]. Mice were
6
ACCEPTED MANUSCRIPT
137
first injected with either saline, NTX, or NMF 30 minutes prior to the locomotor session
138
and returned to their home cage. Animals were then injected with either saline or
139
cocaine (15 mg/kg) and immediately placed into the open field arena and allowed to
140
freely explore the arena for 30 minutes. Locomotor sessions occurred at the same time
141
each day (1200-1700 hours) with animals injected with drug (NTX or NMF + cocaine)
142
and saline (saline + saline) treatment on alternate days, for a total of eight locomotor
143
sessions (four drug and four saline).
144 145 146
2.6 Rotarod NMF was shown to act as a KOPr partial agonist both in vitro, in [35S]GTPγS
147
assays, and in vivo, in humans, (Bart et al., 2005), but was shown to antagonize KOPr
148
-arrestin-2 recruitment (i.e., extremely biased partial agonist; Stahl et al., 2015). KOPr
149
-arrestin-2 recruitment by differentially G-protein biased KOPr agonists in vivo has
150
been associated with motor incoordination in the rotarod assay, as a measure of
151
sedation (Dunn et al., 2018; White et al., 2015). Therefore, blockade of KOPr agonist
152
U50,488H-induced locomotor discoordination using rotarod was used to characterize
153
the effect of NTX and NMF (0, 1, or 10 mg/kg) at the KOPr. Balance and motor
154
coordination was measured on an accelerating rotarod (rod diameter 1.25”; IITC Life
155
Science Inc., Woodland Hills, CA). Drug naïve mice were trained on the apparatus twice
156
daily for up to 5 days. During the first training session, mice were on the rotarod for at
157
least 120 seconds with a constant 3 rpm rotation. For the second session, the rod
158
initially rotated at 3 rpm and gradually increased to a maximum of 15 rpm over a 5 min
159
period. Mice walked on the accelerating rod for a minimum of 200 seconds. For the
7
ACCEPTED MANUSCRIPT
160
remaining training sessions, the top rpm was set to 30 and mice were to again remain
161
on the rod for a minimum of 200 seconds. During training, if a mouse fell from the rod
162
prior to the end of session, they were gently placed back on to the rod. Mice continued
163
training until they were able to remain on the rotarod for at least 150 seconds at the last
164
training session.
165
For testing, the rod initially rotated at 3 rpm and gradually increased to a
166
maximum of 30 rpm over a 5 minute period (the maximum length of the trial). The
167
latency to fall off the rod was measured by the rotarod timer. Between testing, animals
168
were returned to their home cage after falling from the rod. On the testing day, each
169
mouse first completed two drug-free baseline trials to determine performance prior to
170
drug administration. Animals then received a pretreatment injection of either NTX or
171
NMF (0, 1.0, or 10 mg/kg) 30 min prior to an injection of the kappa opioid receptor
172
agonist U50,488H (0 or 10 mg/kg). Rotarod performance was assessed at -10 (+20 min
173
following pretreatment), +30, and +60 minutes following U50,488H administration.
174
Effects of drug treatment were calculated as percent of the average baseline time on
175
the rod before falling.
176 177 178
2.7 Morphine Thermal Analgesia One week following rotarod assay, mice were tested for blockade of morphine-
179
induced analgesia by NTX or NMF (0, 1.0, or 10 mg/kg) using the standard hot-plate
180
method. The apparatus consisted of a clear, vertical, plexiglas cylinder placed on a
181
hotplate (IITC Model 39 D; IITC Inc., Woodland Hills; CA). Mice were habituated to the
182
apparatus by placement onto the room temperature (22 ºC) plate for 30 seconds twice
8
ACCEPTED MANUSCRIPT
183
on each of the two days preceding the testing day. On the day of testing, mice were
184
allowed to walk on the hotplate (54ºC ± 0.1°C) for up to 45 seconds (maximum allowed
185
latency; to avoid tissue damage). Latency to jump, lick a hind paw, or flutter/flick a hind
186
paw (defined as vigorous shaking of paw while standing or stepping such that the paw
187
is extended out of vertical body plane away from body) was recorded. Two baseline
188
measurements separated by 15 min were recorded for each animal to determine
189
performance prior to drug administration. Animals then received a pretreatment injection
190
of either NTX or NMF (0, 1.0, or 10 mg/kg) 30 min prior an injection of the mu opioid
191
receptor agonist morphine (0 or 15 mg/kg; i.p.). Thermal analgesia was assessed at -10
192
(+20 min following pretreatment), +20, +40, and +60 minutes following morphine
193
administration.
194 195 196
2.8 Statistics Conditioned place preference was calculated as percent preference for the drug
197
paired chamber during the post-test session using the following formula: (time drug-
198
paired side post-test)/(time drug-paired side post-test + time saline paired side post-
199
test). The CPP experiments were designed to examine dose related differences with
200
NTX and NMF run in separate cohorts. The locomotor experiments were designed to
201
examine drug related differences with low and high doses run in separate cohorts. A
202
conservative approach to the statistical analyses was used to reduce the potential of
203
error attributable to environmental differences between cohorts. Differences in CPP
204
were analyzed separately for NTX and NMF using two-way analysis of variances
205
(ANOVAs) with pretreatment (saline, 1.0, and 10 mg/kg NTX or NMF) and drug (saline
9
ACCEPTED MANUSCRIPT
206
or 15 mg/kg cocaine) as factors followed by Tukey post hocs for multiple comparisons.
207
Difference in open field locomotor activity across conditioning sessions were analyzed
208
separately for low (1 mg/kg) and high (10 mg/kg) dose during drug paired (days 2, 4, 6,
209
and 8) sessions using two-way repeated measures ANOVAs with treatment and day as
210
factors followed by Tukey post hocs for multiple comparisons for treatment and session.
211
U50488H-induced locomotor discoordination were analyzed as percent of baseline time
212
(in seconds), with the following standard transformation: [(test latency – average
213
baseline latency)/(average baseline latency)] X 100 and analyzed separately for NTX
214
and NMF using two-way repeated measures ANOVAs with treatment and time as
215
factors followed by Dunnett post hocs for multiple comparisons. Antinociceptive effects
216
of morphine were analyzed as percent maximum possible effect (%MPE), with the
217
following standard transformation: %MPE = [(test latency – baseline latency)/(maximum
218
cutoff latency – baseline latency)] X 100 and were analyzed separately for NTX and
219
NMF using two-way repeated measures ANOVAs with treatment and time as factors
220
followed by Dunnett post hocs for multiple comparisons. All analyses were conducted
221
using GraphPad Prism 7.0c (GraphPad Software, La Jolla, CA).
222 223
3. RESULTS
224
3.1 Conditioned Place Preference/Aversion
225
The mean (SEM) percent preference for the drug paired side during the post-
226
test session for NTX is shown in Figure 3a. For NTX, significant effects of both
227
pretreatment [F(2,42)=31.56, p <0.0001] and drug [F(1,42)=30.05, p <0.0001] were
228
observed. No significant pretreatment x drug interaction was found [F(2,42)=1.451, p =
10
ACCEPTED MANUSCRIPT
229
0.45]. Post hoc tests revealed that a significant place preference was observed for
230
saline + cocaine treated mice compared to saline + saline control mice (p<0.01). Both
231
low (p<0.05) and high (p<0.001) dose NTX significantly attenuated the place preference
232
for the cocaine-paired side. High, but not low, dose NTX alone was observed to induce
233
a significant place aversion compared to saline + saline control (p<0.05), although there
234
was a trend towards aversion in the low dose (Figure 3a).
235
The mean (SEM) percent preference for drug paired side during post-test
236
session for NMF is shown in Figure 3b. For NMF, a significant effect of pretreatment
237
[F(2,42)=21.05, p <0.001] and drug [F(1,42)=37.04, p <0.0001] were observed. No
238
significant pretreatment x drug interaction was found [F(2,42)=1.183, p = 0.55]. Post hoc
239
tests revealed that a significant place preference was observed for saline + cocaine
240
treated mice compared to saline + saline control (p<0.05). In contrast with NTX, high
241
(p<0.05), but not low (p=0.09), dose NMF significant attenuated the place preference for
242
the cocaine-paired side, although there was a trend towards attenuation in the low dose.
243
Both low (p<0.05) and high (p<0.01) dose NMF alone were observed to induce a
244
significant place aversion compared to saline + saline control (Figure 3b).
245 246
3.2 Locomotor Sensitization
247
3.2.1 Low dose NTX or NMF Pretreatment
248
The mean (SEM) distance traveled (in cm) for low (1 mg/kg) dose NTX or NMF
249
plus cocaine across drug exposure days (days 2, 4, 6, and 8) are shown in Figure 4a.
250
For drug exposure sessions, a significant effect of treatment [F(5,126)=61.07, p
251
<0.0001], day [F(3,126)=5.12, p<0.0001], and treatment x day interaction
11
ACCEPTED MANUSCRIPT
252
[F(15,126)=8.59, p <0.0001] were observed. Post hoc tests revealed a significant
253
increase in locomotion in the saline + cocaine (p<0.01) and low NTX + cocaine (p<0.01)
254
groups compared to saline + saline control on day 2. Additionally, a significant increase
255
in locomotion was observed for the saline + cocaine, low NTX + cocaine, and low NMF
256
+ cocaine groups compared to saline + saline control on day 4 (p<0.01 each group
257
versus control), day 6 (p<0.0001 each group versus control), and day 8 (p<0.0001 each
258
group versus control). A significant increase in locomotion (i.e., locomotor sensitization)
259
was observed on day 6 and day 8 for the saline + cocaine (p<0.01), low NTX + cocaine
260
(p<0.0001), and low NMF + cocaine (p<0.001) groups compared to day 2. No effect of
261
pretreatment by NTX or NMF was observed (Figure 4a).
262 263 264
3.2.2 High dose NTX or NMF Pretreatment The mean (SEM) distance traveled (in cm) for high (10 mg/kg) NTX or NMF
265
plus cocaine across drug exposure days (days 2, 4, 6, and 8) are shown in Figure 4b.
266
For drug exposure days, a significant effect of treatment [F(5,126)=59.91, p <0.0001],
267
day [F(3,126)=4.73, p <0.0001], and treatment x day interaction [F(15,126)=4.68, p
268
<0.0001] were observed. Post hoc tests revealed a significant increase in locomotion in
269
the saline + cocaine (p<0.05), high NTX + cocaine (p<0.01), and high NMF + cocaine
270
(p<0.01) groups compared to saline + saline control on day 2. Additionally, a significant
271
increase in locomotion was observed for the saline + cocaine, high NTX + cocaine, and
272
high NMF + cocaine groups compared to saline + saline control on day 4 (p<0.001 each
273
group versus control), day 6 (p<0.0001 each group versus control), and day 8
274
(p<0.0001 each group versus control). Post hoc tests revealed a significant increase in
12
ACCEPTED MANUSCRIPT
275
locomotion (i.e., locomotor sensitization) on day 6 (p=0.02) and day 8 (p<0.01) for the
276
saline + cocaine group compared to day 2. A significant increase in locomotion was
277
observed for high NTX + cocaine (p<0.001) and high NMF + cocaine (p<0.001) groups
278
on day 8 compared to day 2. No effect of pretreatment by NTX or NMF was observed
279
(Figure 4b).
280 281
3.3 Effectiveness of NTX and NMF at the Kappa Opioid Receptor (U50,488H
282
locomotor discoordination assay)
283
The average baseline latency before falling, across groups was 224 6 seconds.
284
No group differences were observed for baseline latency [F(5,42)=0.28, p =0.92].
285
Following U50,488H, latency decreased to 123 20 seconds at +30 min maximal time
286
point for the saline + U50 group. The mean (SEM) percent baseline time before falling
287
from the rotarod for low (1 mg/kg) and high (10 mg/kg) NTX (a) and NMF (b) are shown
288
in Figure 5. A significant effect of treatment [F(5,84)=16.43, p <0.05] and time
289
[F(2,84)=9.13, p <0.0001] were observed. No significant treatment x time interaction
290
was observed [F(10,84)=5.96, p = 0.07]. Post hoc tests revealed no significant effect of
291
either dose of NTX or NMF on locomotor discoordination 20 minutes following injection
292
compared to vehicle (p>0.99). A significant locomotor discoordination was observed for
293
the saline + U50 group both 30 minutes (p<0.01) and 60 minutes (p<0.05) following U50
294
administration compared to saline + saline control. As well, a significant locomotor
295
discoordination was observed for the low NTX + U50 group 30 minutes following U50
296
administration compared to saline + saline control (p<0.05). The reduction in percent
297
baseline time before falling for the low NTX + U50 group was not significantly different
13
ACCEPTED MANUSCRIPT
298
from the saline + U50 group (p=0.76). High dose NTX + U50 and well as both doses of
299
NMF + U50 were not significantly different from saline + saline control at both the 30
300
and 60 minute time point following U50 administration (p> 0.9). As well, these doses
301
(high NTX, low and high NMF) were significantly different from the saline + U50 group at
302
the 30 minute time point (p 0.01).
303 304
3.4 Effectiveness of NTX and NMF at Mu Opioid Receptor (morphine thermal
305
analgesia assay)
306
The average baseline latency, across groups was 15.9 0.5 seconds. No group
307
differences were observed for baseline latency [F(5,42)=0.45, p =0.81]. Following
308
morphine, latency increased to 44.1 0.9 seconds at +20 min maximal time point for
309
the saline + morphine group. The mean (SEM) percent maximal effect for low (1
310
mg/kg) and high (10 mg/kg) NTX (a) and NMF (b) are shown in Figure 6. A significant
311
effect of treatment [F(5,126)=48.58, p <0.0001], time [F(3,126)=8.31, p <0.0001], and
312
treatment x time interaction [F(15,126)=22.85, p <0.0001] were observed. Post hoc
313
tests revealed no significant effect of either dose of NTX or NMF on thermal analgesia
314
20 minutes following injection compared to vehicle (p>0.67). A significant analgesic
315
effect was observed for the saline + morphine group at 20, 40, and 60 minutes
316
(p<0.0001) following morphine administration compared to saline + saline control. No
317
significant difference in thermal analgesia was observed for either dose of NTX or NMF
318
at 20, 40, and 60 minutes following morphine administration compared to saline + saline
319
control (p>0.75).
320
14
ACCEPTED MANUSCRIPT
321 322
4. DISCUSSION There are conflicting reports on the effectiveness of the MOPr antagonist/KOPr
323
partial agonist NTX for treating cocaine use disorder depending on the patient
324
population examined. In multiple studies in which the subject population was restricted
325
to individuals with only cocaine use disorder, the standard NTX dose (i.e., 50 mg/day)
326
was sufficient to moderately reduce the positive subjective effects of cocaine (Kosten et
327
al., 1992; Sofuoglu et al., 2003), cocaine craving (Comer et al., 2013), and cocaine use
328
(Kosten et al., 1989; Schmitz et al., 2014; Schmitz et al., 2001). In studies with a subject
329
population with polysubstance dependence, in particular cocaine and alcohol co-
330
dependence, 50-100 mg/day NTX treatment did not significantly reduce cocaine use or
331
craving (Hersh et al., 1998; Modesto-Lowe et al.; Schmitz et al., 2009; Schmitz et al.,
332
2004). However, 150 mg/day NTX significantly reduced cocaine use for patients with
333
cocaine and alcohol co-dependence (Oslin et al., 1999; Pettinati et al., 2008). It should
334
be noted that medication compliance was relatively low for these studies (e.g., Pettinati,
335
et al., 2008; Schmitz, et al., 2001). Therefore, to interpret the positive treatment effects
336
of NTX for reducing cocaine use one should also consider the low study and medication
337
compliance as well as the known side effects of NTX. As well, these potential clinical
338
“complications” may limit the therapeutic benefit of NTX, and by extension NMF, in a
339
cocaine dependence treatment context. Use of extended release NTX (i.e., Vivitrol, a
340
once month intramuscular injection) may reduce the compliance related complications
341
associated with daily oral NTX administration. Unfortunately, however, no clinical
342
studies to date have examined the effectiveness of Vivitrol in reducing cocaine use and
15
ACCEPTED MANUSCRIPT
343
craving in cocaine dependent patients without other substance dependence (i.e.,
344
cocaine but not alcohol dependent population).
345
Although there is growing preclinical and clinical evidence for the effectiveness of
346
NTX for the treatment of cocaine addiction very limited research has examined the
347
structurally similar MOPr antagonist/KOPr partial agonist NMF for treating cocaine use
348
disorder. No preclinical studies to date have examined the effect of NMF on cocaine
349
reward. Clinically, a single case report has described “as needed” NMF (18 mg)
350
reducing cocaine craving in a female cocaine-dependent patient for 5 months
351
(Grosshans et al., 2015). Here we report, to our knowledge for the first time, direct
352
comparison of NTX and NMF in mouse models. We examined the effectiveness of low
353
(1 mg/kg) and high (10 mg/kg) doses of NTX and NMF on attenuating the rewarding
354
effects of cocaine.
355
As has been reported previously (e.g. Bailey et al., 2012; Velazquez-Sanchez et
356
al., 2010), a significant conditioned place preference was observed for 15 mg/kg
357
cocaine. Pretreatment with NTX or NMF 30 minutes prior to cocaine injection was
358
observed to attenuate the place preference for cocaine for both the low and high doses
359
of NTX and high dose NMF. This is in line with prior preclinical NTX studies which have
360
demonstrated that either acute or chronic NTX pretreatment significantly reduces
361
cocaine place preference (Bilsky et al., 1992; Mitchem et al., 1999; Sala et al., 1995;
362
Suzuki et al., 1992). Importantly, this is the first observation of NMF attenuating cocaine
363
place preference. The interaction between NTX or NMF with cocaine cannot be directly
364
ruled out as being strictly behavioral rather than pharmacological in nature from these
365
experiments. However, prior studies have shown that NTX, but not lithium chloride,
16
ACCEPTED MANUSCRIPT
366
blocks cocaine place preference despite both NTX and lithium chloride having similar
367
conditioned place aversions when administered alone (Sala et al., 1995; Suzuki et al.,
368
1992). This suggests that the blockade of cocaine place preference by NTX, and by
369
extension NMF, observed here may be pharmacological in nature. One potential
370
mechanism may be that NTX and NMF attenuate cocaine-induced striatal dopaminergic
371
efflux via their KOPr partial agonist effects as full KOPr agonists have been observed to
372
fully block cocaine-induced striatal dopamine efflux (Zhang et al., 2004a, b). Further
373
studies are needed to clarify the precise mechanism(s) by which NTX and NMF block
374
the rewarding effects of cocaine.
375
In our in vivo characterization in C57Bl/6 mice, both the low and high doses of
376
NTX fully block MOPr agonist induced analgesia; however, the high, but not low, dose
377
of NTX fully blocked KOPr agonist-induced locomotor discoordination. This suggests
378
that the low (1 mg/kg) dose of NTX is fully active at MOPr, but not KOPr. Similar to the
379
findings with NTX, both the low and high doses of NMF fully block MOPr agonist
380
induced analgesia; however, both NMF doses were observed to fully block KOPr
381
agonist-induced locomotor discoordination. Prior in vitro studies (deHaven-Hudkins et
382
al., 1990; Ghirmai et al., 2008; Michel et al., 1985) suggest that NMF is more potent
383
than NTX at the KOPr. This is supported by our finding that systemic administration of 1
384
mg/kg NMF, but not 1 mg/kg NTX, is able to fully block the KOPr-agonist induced
385
locomotor discoordination.
386
Together with the CPP data, our findings suggest that the rewarding effects of
387
cocaine are reduced by a MOPr selective dose of NTX (1 mg/kg) and fully attenuated by
388
a NTX or NMF dose (i.e., 10 mg/kg) that is active at both mu and kappa opioid
17
ACCEPTED MANUSCRIPT
389
receptors. This indicates that for the MOPr antagonists/KOPr partial agonists NTX and
390
NMF, reduction of the rewarding effects of cocaine can be achieved by MOPr selective
391
doses; however, further reductions are observed at doses active at both the MOP and
392
KOP receptors. Systemic and striatal administration of full KOPr agonists (e.g., Dyn A1-
393
17,
394
cocaine place preference in both rats and mice (Crawford et al., 1995; Shippenberg et
395
al., 1996; Zhang et al., 2004a, b). This provides some support for the attenuation of
396
cocaine place preference by high dose NTX/NMF being in part due to their partial
397
agonist effects at the KOPr. However, due to a lack of selective KOPr partial agonists
398
the precise role of KOPr partial agonism on attenuating cocaine reward cannot be
399
determined. As well, the effect of high dose NTX/NMF may be due to other factors
400
beyond KOPr partial agonism (e.g., signaling events downstream of MOPr or action at
401
delta opioid receptors).
402
U50488H, U69593, and R-84760) has been observed to significantly attenuate
In addition to increased potency at KOPr and DOPr, NMF has been shown to
403
have a longer duration of action than NTX across several species with a mean terminal
404
half-life of 11-13 hrs versus 9 hrs in humans, respectively (Dixon et al., 1987; Gal et al.,
405
1986; Ingman et al., 2005; Kyhl et al., 2016; Misra et al., 1976; Teklezgi et al., 2018;
406
Wall et al., 1981). In rats, similar plasma half-lives were observed for NTX (1.4 hr) and
407
NMF (50 min) (Misra et al., 1976; Murthy et al., 1996); it should be noted, however, that
408
different routes of drug administration (ip versus iv, respectively) were utilized for these
409
studies. Similar reports of naltrexone and/or nalmefene in mice have not been reported;
410
but as no significant species difference was observed for plasma NTX binding across
411
species, in particular between rats and mice (20% and 22%, respectively), plasma half-
18
ACCEPTED MANUSCRIPT
412
lives for NTX and NMF may be similar (Ludden et al., 1976). Both compounds are
413
extensively metabolized by the liver with species specific differences in metabolites
414
(Dayton and Inturrisi, 1976; Dixon et al., 1987; Ingman et al., 2005; Misra et al., 1976;
415
Murthy et al., 1996; Wall et al., 1981). In particular, for humans the major metabolites of
416
NTX are the biologically inert glucuronide conjugated NTX and peripherally active opioid
417
receptor antagonist 6--naltrexol (Wall et al., 1981) while rats have major metabolites of
418
glucuronide conjugated NTX, 7,8-dihydro-14-hydroxy-normorphinone, and 7,8-dihydro-
419
14-hydroxy-normorphine with negligible 6--naltrexol observed in plasma (Misra et al.,
420
1976; Rodgers et al., 1980). Conversely, for humans the major metabolite of NMF is
421
glucuronide conjugated NMF (Dixon et al., 1987; Ingman et al., 2005) while in rats NMF
422
is metabolized to the potentially biologically active nornalmefene (Murthy et al., 1996).
423
The biological relevance of these species-specific divergences in NTX and NMF
424
metabolites are predominately unknown.
425
In humans NTX and NMF have been shown to fully block the physiological and
426
behavioral effects of fentanyl up to 48 hrs following acute dosing (Gal et al., 1986).
427
Despite rather rapid plasma clearance (1.4 hrs in humans and 50 min in rats), NMF has
428
been observed to slowly dissociate from central MOPr with a clearance half-life of 28.6
429
hrs (Kim et al., 1997) and ~50-70% occupancy 50 hrs post administration (Ingman et al.,
430
2005). Similar protracted effects of opioid ligands have been observed. For example, in
431
vivo the plant-derived KOPr agonist Salvinorin A has a short duration of action on
432
thermal analgesia in mice with maximal effect at 10-15 min and no effect 30-45 min
433
following administration (Ansonoff et al., 2006; John et al., 2006). However, Salvinorin A
434
has been shown to significantly reduce striatal extracellular dopamine concentration in
19
ACCEPTED MANUSCRIPT
435
mice for 10 hours following administration (Zhang et al., 2005). Therefore, although the
436
efficacies of NTX and NMF at MOPr and KOPr were examined here for double the
437
duration of place conditioning and locomotor activity assays (i.e., 60 minutes) it is likely
438
that both compounds are still centrally active far longer than the period measured.
439
Further research is needed to examine the duration of action of NTX and NMF at MOPr
440
and KOPr centrally, in particular regarding striatal dopamine levels.
441
Mixed findings have been reported as to the place conditioned effect of NTX
442
alone. Some studies have shown that 5 – 15 minute pretreatment with low dose NTX
443
(0.01-1 mg/kg) prior to the conditioning session was neutral in the conditioned place
444
preference paradigm, producing neither preference nor aversion for the drug paired
445
compartment (Bespalov et al., 1999; Parker and Rennie, 1992). Others, however, have
446
reported that either low dose NTX (0.1 – 1 mg/kg) administered immediately prior to the
447
conditioning session or higher dose NTX (3-10 mg/kg) produced a conditioned place
448
aversion (Bechara and van der Kooy, 1985; Daniels et al., 2016; Suzuki et al., 1992).
449
The inconsistencies across studies in the literature are not clearly explained by
450
procedural differences alone. Here, a significant place avoidance was found for high
451
(10mg/kg) dose NTX and both doses of NMF (1.0 and 10 mg/kg) with a 30-minute
452
pretreatment using an unbiased conditioning paradigm. Previous studies have shown
453
that blockade of central endogenous opioid activity by MOPr antagonists can induce a
454
CPA (Mucha et al., 1985). As both low and high dose NTX and NMF were found to fully
455
block MOPr-induced thermal analgesia, the observed place aversion for NTX and NMF
456
is consistent with their antagonist action at MOPr.
20
ACCEPTED MANUSCRIPT
457
Systemic administration of NTX has been reported to block cocaine-induced
458
hyperlocomotion (Sala et al., 1995); however, chronic NTX exposure via osmotic pump
459
has been shown to only slightly reduce (Kunko et al., 1998) or even increase cocaine-
460
induced hyperlocomotion (Lesscher et al., 2005). Here, no effect of either NTX or NMF
461
pretreatment on cocaine-induced locomotor stimulation or locomotor sensitization was
462
observed. This was an unexpected, though not unprecedented, finding. Prior studies
463
have demonstrated a dissociation between the rewarding and locomotor activating
464
aspects of various drugs including cocaine (Jocham et al., 2006; Pum et al., 2008;
465
Rademacher et al., 2000; Rademacher and Steinpreis, 2002; Ukai and Holtzman,
466
1988). This suggests that the rewarding effects of cocaine, as measured by conditioned
467
place preference, and the cocaine-induced hyperlocomotion and locomotor sensitization
468
may involve divergent neural pathways. The former pathway may be modulated by the
469
opioid system, with the latter not necessarily directly interacting with the opioid system.
470
It should be noted that only a single dose of cocaine was used for the place conditioning
471
and cocaine-induced hyperlocomotion assays. This dose (15 mg/kg) was selected
472
based both on previous studies (Bailey et al., 2012; Velazquez-Sanchez et al., 2010)
473
and pilot work demonstrating that this dose is sufficient to produce a robust cocaine
474
place preference. The inconsistency between these findings and the literature for NTX
475
blockade of cocaine-induced hyperlocomotion may be due to the dose of cocaine used
476
(15 mg/kg versus 10 mg/kg used by Sala et al. 1995). Further research is needed to
477
clarify the role of NTX/NMF in attenuating cocaine-induced hyperlocomotion and
478
cocaine reward.
21
ACCEPTED MANUSCRIPT
479
Overall, the structurally and pharmacologically similar compounds NTX and NMF
480
were both demonstrated to attenuate the rewarding effects of cocaine in C57Bl/6 mice,
481
as reflected in dose-dependent reduced conditioned place preference to cocaine. Both
482
of these compounds also produced aversive effects in the place conditioning paradigm
483
when administered alone. The cocaine CPP findings suggest that antagonism/partial
484
agonism at the KOPr may further attenuates the rewarding effects of cocaine in non-
485
dependent male C57Bl/6 mice. Further research is necessary to determine the potential
486
mechanism(s) by which high dose NTX or NMF treatment reduces cocaine craving in
487
cocaine and alcohol co-dependent patient populations.
488 489
Acknowledgements: The authors gratefully thank Dr. Eduardo Butelman for his
490
constructive discussion of the data and manuscript. We also thank Aurora Grutman for
491
her assistance with the open field assay.
492
Funding: This work was supported by the Gary R. Helman Postdoctoral Research
493
Fellowship (KAW), the Robertson Therapeutic Development Fund (BR, MJK), and the
494
Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (MJK). Sponsors had
495
no role in study design; collection, analysis, and interpretation of data; writing
496
manuscript; or journal selection for publication.
497
22
ACCEPTED MANUSCRIPT
498
References
499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541
Ansonoff, M. A., Zhang, J., Czyzyk, T., Rothman, R. B., Stewart, J., Xu, H., Zjwiony, J., Siebert, D. J., Yang, F., Roth, B. L., Pintar, J. E., 2006. Antinociceptive and hypothermic effects of Salvinorin A are abolished in a novel strain of kappaopioid receptor-1 knockout mice. J Pharmacol Exp Ther 318, 641-648. 10.1124/jpet.106.101998 Bailey, J., Ma, D., Szumlinski, K. K., 2012. Rapamycin attenuates the expression of cocaine-induced place preference and behavioral sensitization. Addict Biol 17, 248-258. 10.1111/j.1369-1600.2010.00311.x Bart, G., Schluger, J. H., Borg, L., Ho, A., Bidlack, J. M., Kreek, M. J., 2005. Nalmefene induced elevation in serum prolactin in normal human volunteers: partial kappa opioid agonist activity? Neuropsychopharmacology 30, 22542262. 10.1038/sj.npp.1300811 Bechara, A., van der Kooy, D., 1985. Opposite motivational effects of endogenous opioids in brain and periphery. Nature 314, 533-534. Bespalov, A. Y., Tokarz, M. E., Bowen, S. E., Balster, R. L., Beardsley, P. M., 1999. Effects of test conditions on the outcome of place conditioning with morphine and naltrexone in mice. Psychopharmacology (Berl) 141, 118-122. Bilsky, E. J., Montegut, M. J., Delong, C. L., Reid, L. D., 1992. Opioidergic modulation of cocaine conditioned place preferences. Life Sci 50, PL85-90. Comer, S. D., Mogali, S., Saccone, P. A., Askalsky, P., Martinez, D., Walker, E. A., Jones, J. D., Vosburg, S. K., Cooper, Z. D., Roux, P., Sullivan, M. A., Manubay, J. M., Rubin, E., Pines, A., Berkower, E. L., Haney, M., Foltin, R. W., 2013. Effects of acute oral naltrexone on the subjective and physiological effects of oral D-amphetamine and smoked cocaine in cocaine abusers. Neuropsychopharmacology 38, 2427-2438. 10.1038/npp.2013.143 Crawford, C. A., McDougall, S. A., Bolanos, C. A., Hall, S., Berger, S. P., 1995. The effects of the kappa agonist U-50,488 on cocaine-induced conditioned and unconditioned behaviors and Fos immunoreactivity. Psychopharmacology (Berl) 120, 392-399. Daniels, S., Marshall, P., Leri, F., 2016. Alterations of naltrexone-induced conditioned place avoidance by pre-exposure to high fructose corn syrup or heroin in Sprague-Dawley rats. Psychopharmacology (Berl) 233, 425-433. 10.1007/s00213-015-4121-9 Dayton, H. E., Inturrisi, C. E., 1976. The urinary excretion profiles of naltrexone in man, monkey, rabbit, and rat. Drug Metab Dispos 4, 474-478. deHaven-Hudkins, D. L., Brostrom, P. A., Allen, J. T., Lesko, L. J., Ferkany, J. W., Kaplita, P. V., Mavunkel, B. J., Rzeszotarski, W. J., Steranka, L. R., 1990. Pharmacologic profile of NPC 168 (naltrexone phenyl oxime), a novel compound with activity at opioid receptors. Pharmacology Biochemistry and Behavior 37, 497-504. 10.1016/0091-3057(90)90019-e Di Chiara, G., Imperato, A., 1988. Opposite effects of mu and kappa opiate agonists on dopamine release in the nucleus accumbens and in the dorsal caudate of freely moving rats. J Pharmacol Exp Ther 244, 1067-1080.
23
ACCEPTED MANUSCRIPT
542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587
Dixon, R., Gentile, J., Hsu, H. B., Hsiao, J., Howes, J., Garg, D., Weidler, D., 1987. Nalmefene: safety and kinetics after single and multiple oral doses of a new opioid antagonist. J clin pharmacol 27, 233-239. Dunn, A. D., Reed, B., Guariglia, C., Dunn, A. M., Hillman, J. M., Kreek, M. J., 2018. Structurally-related kappa opioid receptor agonists with substantial differential signaling bias: neuroendocrine and behavioral effects in C57BL6 mice. Intl J Neuropsychopharm, In Press. Gal, T. J., DiFazio, C. A., Dixon, R., 1986. Prolonged blockade of opioid effect with oral nalmefene. Clinical Pharmacology and Therapeutics 40, 537-542. 10.1038/clpt.1986.220 Ghirmai, S., Azar, M. R., Polgar, W. E., Berzetei-Gurske, I., Cashman, J. R., 2008. Synthesis and biological evaluation of alpha- and beta-6-amido derivatives of 17-cyclopropylmethyl-3, 14beta-dihydroxy-4, 5alpha-epoxymorphinan: potential alcohol-cessation agents. J Med Chem 51, 1913-1924. 10.1021/jm701060e Grosshans, M., Mutschler, J., Kiefer, F., 2015. Treatment of cocaine craving with asneeded nalmefene, a partial kappa opioid receptor agonist: first clinical experience. Int Clin Psychopharmacol 30, 237-238. 10.1097/YIC.0000000000000069 Henderson-Redmond, A., Czachowski, C., 2014. Effects of systemic opioid receptor ligands on ethanol- and sucrose seeking and drinking in alcohol-preferring (P) and Long Evans rats. Psychopharmacology (Berl) 231, 4309-4321. 10.1007/s00213-014-3571-9 Hersh, D., Van Kirk, J. R., Kranzler, H. R., 1998. Naltrexone treatment of comorbid alcohol and cocaine use disorders. Psychopharmacology 139, 44-52. DOI 10.1007/s002130050688 Houdi, A. A., Bardo, M. T., Van Loon, G. R., 1989. Opioid mediation of cocaineinduced hyperactivity and reinforcement. Brain Res 497, 195-198. Ingman, K., Hagelberg, N., Aalto, S., Nagren, K., Juhakoski, A., Karhuvaara, S., Kallio, A., Oikonen, V., Hietala, J., Scheinin, H., 2005. Prolonged central mu-opioid receptor occupancy after single and repeated nalmefene dosing. Neuropsychopharmacology 30, 2245-2253. 10.1038/sj.npp.1300790 Jocham, G., Lezoch, K., Muller, C. P., Kart-Teke, E., Huston, J. P., de Souza Silva, M. A., 2006. Neurokinin receptor antagonism attenuates cocaine's behavioural activating effects yet potentiates its dopamine-enhancing action in the nucleus accumbens core. Eur J Neurosci 24, 1721-1732. 10.1111/j.14609568.2006.05041.x John, T. F., French, L. G., Erlichman, J. S., 2006. The antinociceptive effect of salvinorin A in mice. Eur J Pharmacol 545, 129-133. 10.1016/j.ejphar.2006.06.077 Kim, S., Wagner, K. N., Villemagne, V. L., Kao, P. F., Dannals, R. F., Ravert, H. T., Job, T., Dixon, R. B., Civelek, A. C., 1997. Longer occupancy of opioid receptors by nalmefene compared to naloxone as measured in vivo by a dualdetector system. Journal of Nuclear Medicine 38, 1726-1731. Kosten, T., Silverman, D. G., Fleming, J., Kosten, T. A., Gawin, F. H., Compton, M., Jatlow, P., Byck, R., 1992. Intravenous cocaine challenges during naltrexone maintenance: a preliminary study. Biol Psychiatry 32, 543-548.
24
ACCEPTED MANUSCRIPT
588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632
Kosten, T. R., Kleber, H. D., Morgan, C., 1989. Role of opioid antagonists in treating intravenous cocaine abuse. Life Sciences 44, 887-892. 10.1016/00243205(89)90589-4 Kunko, P. M., French, D., Izenwasser, S., 1998. Alterations in locomotor activity during chronic cocaine administration: effect on dopamine receptors and interaction with opioids. J Pharmacol Exp Ther 285, 277-284. Kyhl, L. E., Li, S., Faerch, K. U., Soegaard, B., Larsen, F., Areberg, J., 2016. Population pharmacokinetics of nalmefene in healthy subjects and its relation to mu-opioid receptor occupancy. Br J Clin Pharmacol 81, 290-300. 10.1111/bcp.12805 Lesscher, H. M., Hordijk, M., Bondar, N. P., Alekseyenko, O. V., Burbach, J. P., van Ree, J. M., Gerrits, M. A., 2005. Mu-opioid receptors are not involved in acute cocaine-induced locomotor activity nor in development of cocaine-induced behavioral sensitization in mice. Neuropsychopharmacology 30, 278-285. 10.1038/sj.npp.1300529 Lett, B. T., 1989. Repeated Exposures Intensify Rather Than Diminish the Rewarding Effects of Amphetamine, Morphine, and Cocaine. Psychopharmacology 98, 357-362. Doi 10.1007/Bf00451687 Ludden, T. M., Malspeis, L., Baggot, J. D., Sokoloski, T. D., Frank, S. G., Reuning, R. H., 1976. Tritiated Naltrexone Binding in Plasma from Several Species and Tissue Distribution in Mice. journal of pharmaceutical sciences 65, 712-716. DOI 10.1002/jps.2600650521 Mansour, A., Khachaturian, H., Lewis, M. E., Akil, H., Watson, S. J., 1987. Autoradiographic differentiation of mu, delta, and kappa opioid receptors in the rat forebrain and midbrain. J Neurosci 7, 2445-2464. Mansour, A., Khachaturian, H., Lewis, M. E., Akil, H., Watson, S. J., 1988. Anatomy of CNS opioid receptors. Trends Neurosci 11, 308-314. Michel, M. E., Bolger, G., Weissman, B. A., 1985. Binding of a new opiate antagonist, nalmefene, to rat brain membranes. Methods Find Exp Clin Pharmacol 7, 175177. Misra, A. L., Bloch, R., Vardy, J., Mule, S. J., Verebely, K., 1976. Disposition of (15,163H)naltrexone in the central nervous system of the rat. Drug Metab Dispos 4, 276-280. Mitchem, L. D., Kruschel, C. K., Dallman, E., Anders, K. A., Czapiga, M., Panos, J. J., Steinpreis, R. E., 1999. The effects of the naltrexone implant on rodent social interactions and cocaine-induced conditioned place preference. Pharmacol Biochem Behav 62, 97-102. Modesto-Lowe, V., Burleson, J. A., Hersh, D., Bauer, L. O., Kranzler, H. R., 1997. Effects of naltrexone on cue-elicited craving for alcohol and cocaine. Drug Alcohol Depend 49, 9-16. Mucha, R. F., Millan, M. J., Herz, A., 1985. Aversive Properties of Naloxone in NonDependent (Naive) Rats May Involve Blockade of Central Beta-Endorphin. Psychopharmacology 86, 281-285. Doi 10.1007/Bf00432214 Murthy, S. S., Mathur, C., Kvalo, L. T., Lessor, R. A., Wilhelm, J. A., 1996. Disposition of the opioid antagonist, nalmefene, in rat and dog. xenobiotica 26, 779-792.
25
ACCEPTED MANUSCRIPT
633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676
Oslin, D. W., Pettinati, H. M., Volpicelli, J. R., Wolf, A. L., Kampman, K. M., O’Brien, C. P., 1999. The Effects of Naltrexone on Alcohol and Cocaine Use in Dually Addicted Patients. Journal of Substance Abuse Treatment 16, 163-167. 10.1016/s0740-5472(98)00039-7 Parker, L. A., Rennie, M., 1992. Naltrexone-induced aversions: assessment by place conditioning, taste reactivity, and taste avoidance paradigms. Pharmacol Biochem Behav 41, 559-565. Pettinati, H. M., Kampman, K. M., Lynch, K. G., Suh, J. J., Dackis, C. A., Oslin, D. W., O'Brien, C. P., 2008. Gender differences with high-dose naltrexone in patients with co-occurring cocaine and alcohol dependence. J Subst Abuse Treat 34, 378-390. 10.1016/j.jsat.2007.05.011 Pum, M. E., Carey, R. J., Huston, J. P., Muller, C. P., 2008. Role of medial prefrontal, entorhinal, and occipital 5-HT in cocaine-induced place preference and hyperlocomotion: evidence for multiple dissociations. Psychopharmacology (Berl) 201, 391-403. 10.1007/s00213-008-1296-3 Rademacher, D. J., Anders, K. A., Riley, M. G., Nesbitt, J. T., Steinpreis, R. E., 2000. Determination of the place conditioning and locomotor effects of amperozide in rats: a comparison with cocaine. Exp Clin Psychopharmacol 8, 434-443. Rademacher, D. J., Steinpreis, R. E., 2002. Effects of the selective mu(1)-opioid receptor antagonist, naloxonazine, on cocaine-induced conditioned place preference and locomotor behavior in rats. Neurosci Lett 332, 159-162. Ritz, M., Lamb, R., Goldberg, Kuhar, M., 1987. Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science 237, 12191223. 10.1126/science.2820058 Rodgers, R. M., Taylor, S. M., Dickinson, R. G., Ilias, A. M., Lynn, R. K., Gerber, N., 1980. Metabolism and choleretic effect of naltrexone in the isolated perfused rat liver. Drug Metab Dispos 8, 390-396. Sala, M., Braida, D., Colombo, M., Groppetti, A., Sacco, S., Gori, E., Parenti, M., 1995. Behavioral and biochemical evidence of opioidergic involvement in cocaine sensitization. J Pharmacol Exp Ther 274, 450-457. Schmitz, J. M., Green, C. E., Stotts, A. L., Lindsay, J. A., Rathnayaka, N. S., Grabowski, J., Moeller, F. G., 2014. A two-phased screening paradigm for evaluating candidate medications for cocaine cessation or relapse prevention: modafinil, levodopa-carbidopa, naltrexone. Drug Alcohol Depend 136, 100-107. 10.1016/j.drugalcdep.2013.12.015 Schmitz, J. M., Lindsay, J. A., Green, C. E., Herin, D. V., Stotts, A. L., Moeller, F. G., 2009. High-dose naltrexone therapy for cocaine-alcohol dependence. Am J Addict 18, 356-362. 10.3109/10550490903077929 Schmitz, J. M., Stotts, A. L., Rhoades, H. M., Grabowski, J., 2001. Naltrexone and relapse prevention treatment for cocaine-dependent patients. Addictive Behaviors 26, 167-180. 10.1016/s0306-4603(00)00098-8 Schmitz, J. M., Stotts, A. L., Sayre, S. L., DeLaune, K. A., Grabowski, J., 2004. Treatment of cocaine-alcohol dependence with naltrexone and relapse prevention therapy. Am J Addict 13, 333-341. 10.1080/10550490490480982
26
ACCEPTED MANUSCRIPT
677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722
Shippenberg, T. S., Heidbreder, C., 1995. Sensitization to the conditioned rewarding effects of cocaine: pharmacological and temporal characteristics. J Pharmacol Exp Ther 273, 808-815. Shippenberg, T. S., LeFevour, A., Heidbreder, C., 1996. kappa-Opioid receptor agonists prevent sensitization to the conditioned rewarding effects of cocaine. J Pharmacol Exp Ther 276, 545-554. Shippenberg, T. S., LeFevour, A., Thompson, A. C., 1998. Sensitization to the conditioned rewarding effects of morphine and cocaine: differential effects of the κ-opioid receptor agonist U69593. European Journal of Pharmacology 345, 27-34. 10.1016/s0014-2999(97)01614-2 Simmons, D., Self, D. W., 2009. Role of mu- and delta-opioid receptors in the nucleus accumbens in cocaine-seeking behavior. Neuropsychopharmacology 34, 19461957. 10.1038/npp.2009.28 Sofuoglu, M., Singha, A., Kosten, T. R., McCance-Katz, F. E., Petrakis, I., Oliveto, A., 2003. Effects of naltrexone and isradipine, alone or in combination, on cocaine responses in humans. Pharmacology Biochemistry and Behavior 75, 801-808. 10.1016/s0091-3057(03)00157-6 Stahl, E. L., Zhou, L., Ehlert, F. J., Bohn, L. M., 2015. A novel method for analyzing extremely biased agonism at G protein-coupled receptors. Mol Pharmacol 87, 866-877. 10.1124/mol.114.096503 Suzuki, T., Shiozaki, Y., Masukawa, Y., Misawa, M., Nagase, H., 1992. The role of mu- and kappa-opioid receptors in cocaine-induced conditioned place preference. Jpn J Pharmacol 58, 435-442. Teklezgi, B. G., Pamreddy, A., Baijnath, S., Kruger, H. G., Naicker, T., Gopal, N. D., Govender, T., 2018. Time-dependent regional brain distribution of methadone and naltrexone in the treatment of opioid addiction. Addiction Biology. 10.1111/adb.12609 Ukai, M., Holtzman, S. G., 1988. Effects of beta-funaltrexamine on ingestive behaviors in the rat. Eur J Pharmacol 153, 161-165. Velazquez-Sanchez, C., Ferragud, A., Murga, J., Carda, M., Canales, J. J., 2010. The high affinity dopamine uptake inhibitor, JHW 007, blocks cocaine-induced reward, locomotor stimulation and sensitization. Eur Neuropsychopharmacol 20, 501-508. 10.1016/j.euroneuro.2010.03.005 Wall, M. E., Brine, D. R., Perez-Reyes, M., 1981. Metabolism and disposition of naltrexone in man after oral and intravenous administration. Drug Metab Dispos 9, 369-375. Walsh, S. L., Sullivan, J. T., Preston, K. L., Garner, J. E., Bigelow, G. E., 1996. Effects of naltrexone on response to intravenous cocaine, hydromorphone and their combination in humans. journal of pharmacology and experimental therapeutics 279, 524-538. Wentland, M. P., Lou, R., Lu, Q., Bu, Y., Denhardt, C., Jin, J., Ganorkar, R., VanAlstine, M. A., Guo, C., Cohen, D. J., Bidlack, J. M., 2009. Syntheses of novel high affinity ligands for opioid receptors. Bioorg Med Chem Lett 19, 22892294. 10.1016/j.bmcl.2009.02.078 White, K. L., Robinson, J. E., Zhu, H., DiBerto, J. F., Polepally, P. R., Zjawiony, J. K., Nichols, D. E., Malanga, C. J., Roth, B. L., 2015. The G protein-biased kappa-
27
ACCEPTED MANUSCRIPT
723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743
opioid receptor agonist RB-64 is analgesic with a unique spectrum of activities in vivo. J Pharmacol Exp Ther 352, 98-109. 10.1124/jpet.114.216820 Williams, K. L., Broadbridge, C. L., 2009. Potency of naltrexone to reduce ethanol selfadministration in rats is greater for subcutaneous versus intraperitoneal injection. Alcohol 43, 119-126. 10.1016/j.alcohol.2008.11.003 Zhang, Y., Butelman, E. R., Schlussman, S. D., Ho, A., Kreek, M. J., 2004a. Effect of the endogenous kappa opioid agonist dynorphin A(1-17) on cocaine-evoked increases in striatal dopamine levels and cocaine-induced place preference in C57BL/6J mice. Psychopharmacology (Berl) 172, 422-429. 10.1007/s00213003-1688-3 Zhang, Y., Butelman, E. R., Schlussman, S. D., Ho, A., Kreek, M. J., 2004b. Effect of the kappa opioid agonist R-84760 on cocaine-induced increases in striatal dopamine levels and cocaine-induced place preference in C57BL/6J mice. Psychopharmacology (Berl) 173, 146-152. 10.1007/s00213-003-1716-3 Zhang, Y., Butelman, E. R., Schlussman, S. D., Ho, A., Kreek, M. J., 2005. Effects of the plant-derived hallucinogen salvinorin A on basal dopamine levels in the caudate putamen and in a conditioned place aversion assay in mice: agonist actions at kappa opioid receptors. Psychopharmacology (Berl) 179, 551-558. 10.1007/s00213-004-2087-0
28
ACCEPTED MANUSCRIPT
744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785
Figure Captions. Fig 1. Structures of naltrexone and nalmefene. Fig 2. a) Timeline for place conditioning experiments. Following pretest session, mice received four saline (saline + saline) and four drug (NTX or NMF + cocaine) conditioning sessions (8 days total; saline first conditioning session). Mice received pretreatment (saline, NTX, or NMF) 30 minutes prior to saline or cocaine injection after which they were immediately placed into and confined to appropriate conditioning compartment for 30 minutes. Following conditioning, on day 10 mice had a 30-minute non-drug post-test session. Place preference or aversion was calculated as percent preference for the drug paired side during post-conditioning test (time drug pair side)/ (time drug pair side + saline pair side)x100. b) Timeline for cocaine-induced locomotor sensitization assay. Injections and drug exposure were identical to that for the place conditioning experiment. Fig 3. Effect of naltrexone (a) or nalmefene (b) on cocaine condition place preference (n=8/group). Percent preference for drug paired side during 30-minute drug-free postconditioning test session. Cocaine significantly increased preference for the drug paired side (a and b). Both doses of naltrexone (a) and high dose nalmefene (b) significantly decreased percent preference for the cocaine paired compartment. High dose naltrexone and both doses of nalmefene caused significant aversion for the drug paired compartment. (*p<0.05) Fig 4. Effect of naltrexone and nalmefene on locomotor activity across drug exposure days (n=8/group). Locomotor activity in open field is expressed as mean distance traveled in cm (SEM) during 30 min session on drug exposure days. Cocaine alone (a and b), low dose naltrexone or nalmefene + cocaine (a), and high dose naltrexone or nalmefene + cocaine (b) significantly increased locomotor activity compared to saline control (*p<0.05) as well as were significantly increased on final activity session (day 8) compared to first activity session (day 1; # p<0.05). Fig 5. Effect of naltrexone or nalmefene on kappa opioid receptor agonist U50,488H-induced locomotor discoordination (n=8/group). Significant locomotor discoordination was observed for U50,488H (10 mg/kg) alone compared to vehicle control (* p<0.05). High, but not low, dose naltrexone (a) and both doses of nalmefene (b) fully blocked U50,488H-induced locomotor discoordination (# p<0.05; compared to U50,488H group). Fig 6. Effect of naltrexone or nalmefene on mu opioid receptor agonist morphine-induced thermal analgesia (n=8/group). A significant antinociceptive response was observed for morphine (15 mg/kg) alone compared to vehicle control (* p<0.05). Both doses of naltrexone (a) and nalmefene (b) fully blocked morphine induced antinociception.
29
Figure 1.
Figure 2.
ACCEPTED MANUSCRIPT
Figure 3.
ACCEPTED MANUSCRIPT
Figure 4.
ACCEPTED MANUSCRIPT
Figure 5.
ACCEPTED MANUSCRIPT
Figure 6.
ACCEPTED MANUSCRIPT
Direct comparison structurally similar ligands naltrexone (NTX) and nalmefene (NMF)
NTX and NMF attenuate cocaine-induced conditioned place preference
NTX and NMF alone induce conditioned place avoidance
NTX and NMF have no effect on cocaine-induced hyperlocomotion
NMF in vivo is more potent at kappa opioid receptors than NTX