<<

JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Title: Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey i

Authors: Nicolas Veyres, Adjia Hamadjida, Philippe Huot

Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada NV Montreal Neurological Institute, Montreal, QC, Canada HA,PH Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada PH

Downloaded from jpet.aspetjournals.org at ASPET Journals on October 1, 2021

1 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Running title: Predictive value of parkinsonian primates in drug research

Text pages: 11 Word count abstract: 253 Word count introduction: 290 Word count discussion: 534 Word count manuscript: 2808 Number of figures: 9 Number of tables: 4 Number of references: 364 Number of supplementary tables: 3 Number of supplementary references: 231

Corresponding Author: Philippe Huot: Montreal Neurological Institute, 3801 University St, BT Downloaded from 209, Montreal, QC, Canada, H3A 2B4, Tel: +1-514-398-5957; Fax: +1-514-398-6644; Email: [email protected]

List of non-standard abbreviations jpet.aspetjournals.org AADC: L-aromatic amino acid decarboxylase L-DOPA: L-3,4-dihydroxyphenylalanine MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

Financial disclosure: There are no conflicts of interest.

at ASPET Journals on October 1, 2021

2 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Abstract

The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate is the gold- standard animal model of Parkinson’s disease (PD) and has been used to assess the effectiveness of experimental drugs on dyskinesia, and psychosis. Three species have been used in the majority of studies, the macaque, marmoset and squirrel monkey, the latter much less so than the first 2 species. However, the predictive value of each species at forecasting clinical efficacy, or lack thereof, is poorly documented. Here, we have reviewed all the published literature detailing Downloaded from pharmacological studies that assessed the effects of experimental drugs on dyskinesia, parkinsonism and psychosis in each of these species and have calculated their predictive value of jpet.aspetjournals.org success and failure at the clinical level. We found that, for dyskinesia, the macaque has a positive predictive value of 87.5% and a false positive rate of 38.1%, while the marmoset has a positive predictive value of 76.9% and a false positive rate of 15.6%. For parkinsonism, the macaque has a at ASPET Journals on October 1, 2021 positive predictive value of 68.2% and a false positive rate of 44.4%, while the marmoset has a positive predictive value of 86.9% and a false positive rate of 41.7%. No drug that alleviates psychosis in the clinic has shown efficacy at doing so in the macaque, while the marmoset has

100% positive predictive value. The small number of studies conducted in the squirrel monkey precluded us from calculating its predictive efficacy. We hope that our results will help in the design of pharmacological experiments and will facilitate the drug discovery and development process in PD.

3 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Introduction

Since its accidental discovery (Davis et al., 1979; Langston et al., 1983), 1-methyl-4- phenyl-1,2,3,6-tetrahydropyridine (MPTP) has been used extensively to model Parkinson’s disease

(PD) in a breadth of non-human primates, enabling the investigation of anatomical (Jan et al., 2000;

Zeng et al., 2008), behavioural (Pessiglione et al., 2004a; Pessiglione et al., 2004b), neuro-chemical

(Soghomonian et al., 1994; Huot et al., 2008), etc. aspects of the disease. The MPTP-lesioned primate has also been invaluable in the search of effective anti-dyskinetic and anti-parkinsonian Downloaded from agents. A review article previously examined the translational predictive value of the MPTP- lesioned primate for the development of anti-dyskinetic drugs, but it was published over a decade jpet.aspetjournals.org ago (Fox et al., 2006a), and many drug have since then been tested both in the primate and in clinical settings. This previous review did not examine the translational predictive value of the

MPTP-lesioned primate when it relates to the effect of drugs on parkinsonian disability or at ASPET Journals on October 1, 2021 psychosis, nor did it look at differences between different primate species when it comes to predict the clinical effectiveness of an experimental molecule.

We have therefore conducted a thorough and unbiased review of the literature in order to compare the predictive effectiveness of different MPTP-lesioned primate species on the clinical effectiveness of potential anti-dyskinetic, anti-parkinsonian and anti-psychotic agents.

We believe that this Review comes at a critical time, given that it has recently been suggested that some primate species might be more suited than other to conduct behavioural pharmacological research (Porras et al., 2012), and that there have been several failures of high- profile drugs in clinical trials (Cook et al., 2014; Bespalov et al., 2016), emphasising the need to use the best animal model possible in pre-clinical settings, to maximise chances of success when translating to the clinic.

4 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Methods

Three non-human primate species have been used in the majority of experiments to determine the anti-dyskinetic, anti-parkinsonian and anti-psychotic effect of experimental drugs: the macaque (both cynomolgus (Di Paolo et al., 1986) and rhesus (Burns et al., 1983), Macaca fascicularis and Macaca mulatta, respectively), common marmoset (Jenner et al., 1984) (Callithrix jacchus) and common squirrel monkey (Langston et al., 1984) (Saimiri sciureus). For each of these

3 primate species, we have reviewed all the studies that reported their effect, or lack thereof, on Downloaded from each of dyskinesia, parkinsonism and psychosis. We then sought which of these drugs tested in the non-human primate underwent clinical testing and compared their pre-clinical and clinical effects jpet.aspetjournals.org and calculated different predictive values (see “Endpoints” below).

Inclusion criteria at ASPET Journals on October 1, 2021 The literature search spans from 1983, time at which the first MPTP-lesioned non-human primate was engineered (Burns et al., 1983), until 10th December 2017. Studies published after this date, either online or in print format, are not included.

Only drugs whose pre-clinical and clinical efficacy, or lack thereof, was reported in peer- reviewed articles are included in this article. Results disseminated solely through abstracts or conference proceedings are therefore not reported in this article. We are aware that not all studies conducted on parkinsonian primates have been published, and that results of some clinical trials have not been disseminated through peer-reviewed journals; by choosing to include only studies and reports that were published in peer-reviewed journals, we may be introducing a selection bias in our analysis. For instance, whereas it has been studied in human (NCT00034814,

NCT00108667), the clinical effectiveness of will not be discussed here, as the results of the clinical trials where it was assessed were not published in peer-reviewed scientific journals.

5 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

In addition, we have excluded L-3,4-dihydroxyphenylalanine (L-DOPA) and inhibitors of L- aromatic amino acid decarboxylase (AADC, e.g. (Rinne et al., 1975) and

(Marsden et al., 1973)) from our analysis, as these were used in almost all studies cited here, to reverse parkinsonism and induce dyskinesia.

Research methodology

Literature review was conducted primarily through the United States National Library of Downloaded from

Medicine (NLM) database, accessed via PubMed. The search engine Google was used to complete the literature review and to access articles not indexed in the NLM database. The following terms jpet.aspetjournals.org were used to perform the literature search: abnormal involuntary movements, chorea, cynomolgus, dyskinesia, dyskinetic, dystonia, hallucinations, hyperactivity, hyperkinesia, L-DOPA, levodopa, macaque, marmoset, monkey, at ASPET Journals on October 1, 2021 MPTP, non-human primate, Parkinson’s disease, parkinsonian, parkinsonism, primate, psychosis, rhesus, squirrel monkey, visual hallucinations.

Definition of “efficacy”

Throughout our literature search, a reduction of dyskinesia/psychosis was considered such only if statistical significance was reached; trends were not considered. A drug was deemed to have anti-parkinsonian benefit if it alleviated parkinsonism as monotherapy or as adjunct to L-DOPA.

Impulse-control disorders (Weintraub et al., 2010) were not considered as a psychotic manifestation.

Several studies, especially in the macaque, were performed that administered drugs, e.g. , to reduce the development of dyskinesia and then conducted post-mortem experiments; we have included these studies in our analysis, even if their primary end-point was

6 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171 not determination of pharmacological efficacy. Studies reporting the effects of experimental drugs on rotational behaviour in the hemi-MPTP-lesioned primate, e.g. (Vermeulen et al., 1995), were not included.

In some instances, conflicting results were obtained with some drugs, e.g. studies encountered a therapeutic benefit, while others did not reach similar conclusions; whenever this happened, we calculated the predictive values using “best result”, i.e. the one that showed a therapeutic effect, weighing in the methodology of the studies (2009), if applicable. For instance, Downloaded from in the case of preladenant, Phase II studies have found anti-parkinsonian efficacy (Hauser et al.,

2011; Factor et al., 2013), while Phase III studies did not (Hauser et al., 2015; Stocchi et al., 2017), jpet.aspetjournals.org but in one instance, the active comparator, (Hauser et al., 2015), was also ineffective, despite proven anti-parkinsonian benefit in randomised-controlled trials (ParkinsonStudyGroup,

2005; Rascol et al., 2005), somewhat casting doubt on study conclusions; in this case, we at ASPET Journals on October 1, 2021 considered that preladenant exerted anti-parkinsonian benefit.

Endpoints

By comparing the outcomes of the primate studies on each of dyskinesia, parkinsonism and psychosis, we have calculated, for each species, the positive predictive value, the negative predictive value and the false positive rate.

Here, we define the positive predictive value of a species as the percentage of cases for which a primate species has correctly predicted that an anti-dyskinetic or anti-parkinsonian benefit would be achieved in the clinic. For the positive predictive value, the denominator was the number of drugs for which a therapeutic effect was achieved in the clinic, while the numerator was the number of these drugs that showed efficacy in the primate, i.e.

7 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

������ �� ��������� �ℎ�� �ℎ���� ������ �� �ℎ� �������

������ �� ���������� − ��������� ���������

Here, we define the negative predictive value of a species as the percentage of cases for which a primate species has correctly predicted that a lack of anti-dyskinetic or anti-parkinsonian benefit would be achieved in the clinic. For the negative predictive value, the denominator was the number of drugs for which a therapeutic effect was not achieved in the clinic, while the numerator was the number of these drugs that did not show efficacy in the primate, i.e. Downloaded from ������ �� ��������� �ℎ�� ��� ��� �ℎ�� ������ �� �ℎ� �������

������ �� ���������� − ����������� ���������

Here, we define the false positive rate of a species as the percentage of cases for which a jpet.aspetjournals.org primate species has incorrectly predicted that a lack of anti-dyskinetic or anti-parkinsonian benefit would be achieved in the clinic. For the false positive rate, the denominator was the number of drugs for which a therapeutic effect was not achieved in the clinic, while the nuemrator was the at ASPET Journals on October 1, 2021 number of these clinically-ineffective drugs that were deemed to be effective in the primate, i.e.

������ �� ���������� − ����������� ��������� �ℎ�� �ℎ���� �������� ������ �� �ℎ� �������

������ �� ���������� − ����������� ���������

Results

Pharmacological targets

Several pharmacological targets have been modulated in studies performed in the MPTP- lesioned macaque and the MPTP-lesioned marmoset. In contrast, only dopaminergic, opioidergic and targets have been assessed in studies conducted in the MPTP-lesioned squirrel monkey (see Supplementary Table 1 for the pharmacological profile of all molecules that have been tested in the MPTP-lesioned primate).

8 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

A total of 98 different molecules were assessed in the MPTP-lesioned macaque, 97 in the

MPTP-lesioned marmoset and 9 in the MPTP-lesioned squirrel monkey (Figure 1). Of all the molecules tested in the MPTP-lesioned primate, 64 have been assessed at the clinical level or are clinically-approved. Of these, 44 were tested in the macaque, 38 in the marmoset and 1 in the squirrel monkey (Figure 2). Because very few drugs have been tested in the MPTP-squirrel monkey compared to the macaque and the marmoset, we will not discuss it further in the text, but we have nevertheless included it in the Tables. Downloaded from

We have summarised our research results in Tables:

• Table 1: drugs tested in the MPTP-lesioned macaque; jpet.aspetjournals.org • Table 2: drugs tested in the MPTP-lesioned marmoset;

• Table 3: drugs tested in the MPTP-lesioned squirrel monkey;

• Table 4: drugs tested in the MPTP-lesioned primate that were tested in the clinic or are at ASPET Journals on October 1, 2021 clinically-available;

In each Table, the drugs are listed in numerical/alphabetical order.

We have also summarised our results in Figures:

• Figure 1: number of drugs tested in the MPTP-lesioned non-human primate;

• Figure 2: number of drugs assessed in the clinic or clinically-approved that were tested in the

MPTP-lesioned non-human primate;

• Figure 3: anti-dyskinetic positive predictive value of the MPTP-lesioned non-human primate;

• Figure 4: anti-dyskinetic negative predictive value of the MPTP-lesioned non-human primate;

• Figure 5: anti-dyskinetic false positive rate of the MPTP-lesioned non-human primate;

• Figure 6: anti-parkinsonian positive predictive value of the MPTP-lesioned non-human

primate;

9 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

• Figure 7: anti-parkinsonian negative predictive value of the MPTP-lesioned non-human

primate;

• Figure 8: anti-parkinsonian false positive rate of the MPTP-lesioned non-human primate;

• Figure 9: worsening of parkinsonism positive predictive value of the MPTP-lesioned non-

human primate.

Prediction of anti-dyskinetic effect Downloaded from

Of the 64 drugs that were tested in the clinic and in the MPTP-lesioned primate, 22 showed anti-dyskinetic effect in clinical trials, no anti-dyskinetic effect was found or reported for 36 drugs, jpet.aspetjournals.org and 4 drugs were found to have a deleterious effect on dyskinesia severity (see Supplementary

Table 2 for details).

MPTP-lesioned macaque at ASPET Journals on October 1, 2021 Of the 22 drugs that demonstrated an anti-dyskinetic effect in clinical settings, 16 were tested in the macaque, and an anti-dyskinetic effect was obtained with 14 (87.5% positive predictive value, Figure 3).

Of the 36 drugs for which no anti-dyskinetic effect was found or reported in the clinic, 21 were tested in the macaque. No anti-dyskinetic effect was encountered or reported with 13 (61.9% negative predictive efficacy, Figure 4), while an anti-dyskinetic action was found with 8 (38.1% false positive rate, Figure 5).

Of the 4 drugs that were found to have a deleterious effect on dyskinesia severity in the clinic, 2 were tested in the macaque and an exacerbation of dyskinesia could not be demonstrated in either case.

MPTP-lesioned marmoset

10 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Of the 22 drugs that demonstrated an anti-dyskinetic effect in clinical settings, 13 were tested in the marmoset and anti-dyskinetic effect was obtained with 10 (76.9% positive predictive value, Figure 3).

Of the 36 drugs for which no anti-dyskinetic effect was found or reported, 22 were tested in the marmoset and the absence of anti-dyskinetic effect was identified in 19 (86.4% negative predictive value, Figure 4), while anti-dyskinetic an action was found with 3 (15.6% false positive rate, Figure 5). Downloaded from

Of the 4 drugs that were found to have a deleterious effect on dyskinesia severity, 3 were tested in the marmoset and an exacerbation of dyskinesia could not be demonstrated in any case. jpet.aspetjournals.org

Prediction of anti-parkinsonian action

Of the 64 drugs that were tested in the clinic and in the MPTP-lesioned primate, 34 showed at ASPET Journals on October 1, 2021 anti-parkinsonian effect in clinical trials, no anti-parkinsonian effect was found or reported for 24 drugs, and 5 drugs were found to have a deleterious effect on parkinsonian disability (see

Supplementary Table 3 for details).

MPTP-lesioned macaque

Of the 34 drugs that showed anti-parkinsonian effect in clinical trials, 22 were tested in the macaque and anti-parkinsonian benefit was obtained with 15 (68.2% positive predictive value,

Figure 6).

Of the 24 drugs for which no anti-parkinsonian effect was found or reported, 18 were tested in the macaque and no anti-parkinsonian effect was encountered or reported with 6 (33.3% negative predictive value, Figure 7), while anti-parkinsonian action was found with 8 (44.4% false positive rate, Figure 8).

11 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Of the 5 drugs that were found to have a deleterious effect on parkinsonian disability, 4 were tested in the macaque and this deleterious effect on parkinsonism was correctly identified in

3 (75% predictive value, Figure 9).

MPTP-lesioned marmoset

Of the 34 drugs that showed anti-parkinsonian effect in clinical trials, 23 were tested in the marmoset and anti-parkinsonian effect was obtained with 20 (86.9% positive predictive value,

Figure 6). Downloaded from

Of these, 24 drugs did not find or did not report an anti-parkinsonian effect, 12 were tested in the marmoset and no anti-parkinsonian effect was encountered with 6 (50.0% negative predictive jpet.aspetjournals.org value, Figure 7), while anti-parkinsonian benefit was found with 5 (41.7% false positive rate,

Figure 8).

Of the 5 drugs that were found to have a deleterious effect on parkinsonian disability, 3 at ASPET Journals on October 1, 2021 were tested in the marmoset, all of which hindered parkinsonism (100% predictive value, Figure

9).

Prediction of anti-psychotic action

Of the 64 drugs that were tested in the clinic and in the MPTP-lesioned primate, 5 showed anti-psychotic effect in clinical trials/reports (, , , pimavanserin, ).

In comparison to dyskinesia and parkinsonism, the effect of experimental drugs on dopaminergic psychosis has been far less studied in the MPTP-lesioned primate. No drug that underwent clinical testing or that is clinically-available has demonstrated anti-psychotic effect in the MPTP-lesioned macaque or the MPTP-lesioned squirrel monkey. Pimavanserin was not tested

12 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171 in the MPTP-lesioned marmoset, but an anti-psychotic benefit was achieved with each of clozapine, mianserin, mirtazapine and quetiapine (100% positive predictive value).

Discussion

Here, we have reviewed all the literature published in peer-reviewed scientific journals that reported the results of pharmacological studies conducted in the MPTP-lesioned macaque, Downloaded from marmoset and squirrel monkey where the effects of experimental drugs on dyskinesia, parkinsonism and psychosis was assessed. By comparing the results obtained at the pre-clinical jpet.aspetjournals.org level with those obtained in clinical settings, we have calculated the predictive value of each primate species for these disease manifestations/treatment-related complications.

There are limitations to our analysis that have to be mentioned. Firstly, as we mentioned in at ASPET Journals on October 1, 2021 the Introduction, the results of several studies, both pre-clinical and clinical, have not been published and, although we aimed for exhaustiveness, our Review is necessarily incomplete, which may have affected the various rates presented. Secondly, the methodology of the clinical trials cited is highly-variable, ranging from observational reports to randomised controlled trials; they were weighed equally here. Thirdly, the methodology used in pre-clinical studies is, at times, different to the one used in clinical settings; one example is when a low dose of L-DOPA is administered to primates, in combination with an agent with potential anti-parkinsonian effect as adjunct therapy.

Lowering the L-DOPA dose administered may be poorly tolerated by patients, which is why this approach is seldom used in clinical trials. Fourthly, some clinical trials were performed in early- stage PD patients, while the degree of parkinsonism following MPTP administration is severe and would correspond to advanced-stage PD. Lastly, as several types of trials are part of our Review, in some of them, PD patients were taking anti-parkinsonian in addition to L-DOPA,

13 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171 which is generally not the case in primate studies, and the extent to which these molecules affected the results is undetermined.

Keeping these limitations in mind, the following general conclusions can be drawn:

• Relatively to the anti-dyskinetic effect of drugs, the macaque has higher positive predictive

value than the marmoset, but the marmoset has fewer false positive results than the macaque.

Both the macaque and the marmoset appear limited when it comes to predicting a detrimental

effect of experimental drugs on dyskinesia; Downloaded from

• Relatively to the anti-parkinsonian action of drugs, the marmoset has higher positive predictive

value and fewer false positive results than the macaque than the macaque, both species have jpet.aspetjournals.org high predictive values when it comes to forecasting a potentially deleterious effect of drugs on

parkinsonism;

• Relatively to the anti-psychotic effect of drugs, comments can be made only for the marmoset, at ASPET Journals on October 1, 2021 which has high positive predictive value;

• In comparison to the macaque and the marmoset, the squirrel monkey has been used in a small

number of studies and very few pharmacological targets have been assessed in this primate

species, which makes it impossible to calculate its predictive value.

At a time when the discovery and development process for drugs acting at the central nervous system level is facing challenges and has been marred by failures of high-profile candidates, it is our hope that this Review will help in the planning and design of pre-clinical experiments aimed at testing the effects of drugs on L-DOPA-induced dyskinesia, parkinsonian disability and dopaminergic psychosis, by helping experimenters and sponsors to plan their experiments in the animal model of PD with the highest translational potential for their specific end-point.

14 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Authorship contributions

Participated in research design: Veyres, Huot

Performed data analysis: Veyres, Huot

Wrote or contributed to the writing of the manuscript: Veyres, Hamadjida, Huot

Downloaded from

jpet.aspetjournals.org at ASPET Journals on October 1, 2021

15 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

References

(2009) Clinical Trials Handbook [Internet]. Wiley, Hoboken.

Agid Y, Pollak P, Bonnet AM, Signoret JL and Lhermitte F (1979) associated with

a peripheral blocking agent in treatment of Parkinson's disease. Lancet 1:570-

572.

Ahlskog JE, Wright KF, Muenter MD and Adler CH (1996) Adjunctive cabergoline therapy of

Parkinson's disease: comparison with placebo and assessment of dose responses and Downloaded from

duration of effect. Clin Neuropharmacol 19:202-212.

Akai T, Ozawa M, Yamaguchi M, Mizuta E and Kuno S (1995) Behavioral involvement of central jpet.aspetjournals.org dopamine D1 and D2 receptors in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-

lesioned parkinsonian cynomolgus monkeys. Jpn J Pharmacol 67:117-124.

Antonini A, Tesei S, Zecchinelli A, Barone P, De Gaspari D, Canesi M, Sacilotto G, Meucci N, at ASPET Journals on October 1, 2021 Mariani C and Pezzoli G (2006) Randomized study of and low-dose

in patients with Parkinson's disease and depression: effect on quality of life. Mov Disord

21:1119-1122.

Baron MS and Dalton WB (2003) Quetiapine as treatment for dopaminergic-induced dyskinesias

in Parkinson's disease. Mov Disord 18:1208-1209.

Barone P, Lamb J, Ellis A and Clarke Z (2007) versus placebo or for the

adjunctive treatment of patients with advanced Parkinson's disease. Mov Disord 22:483-

489.

Bedard P, Parkes JD and Marsden CD (1977) in Parkinson's disease. Br J Clin

Pharmacol 4Suppl 2:187S-190S.

Bedard PJ and Boucher R (1989) Effect of D1 receptor stimulation in normal and MPTP monkeys.

Neurosci Lett 104:223-228.

16 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Bedard PJ, Di Paolo T, Falardeau P and Boucher R (1986) Chronic treatment with L-DOPA, but

not bromocriptine induces dyskinesia in MPTP-parkinsonian monkeys. Correlation with

[3H]spiperone binding. Brain Res 379:294-299.

Belanger N, Gregoire L, Bedard P and Di Paolo T (2003a) Estradiol and

potentiate levodopa-induced locomotor activity in 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine monkeys. Endocrine 21:97-101.

Belanger N, Gregoire L, Bedard PJ and Di Paolo T (2006) DHEA improves symptomatic treatment Downloaded from

of moderately and severely impaired MPTP monkeys. Neurobiol Aging 27:1684-1693.

Belanger N, Gregoire L, Hadj Tahar A and Bedard PJ (2003b) Chronic treatment with small doses jpet.aspetjournals.org of cabergoline prevents dopa-induced dyskinesias in parkinsonian monkeys. Mov Disord

18:1436-1441.

Belluzzi JD, Domino EF, May JM, Bankiewicz KS and McAfee DA (1994) N-0923, a selective at ASPET Journals on October 1, 2021 dopamine D2 receptor agonist, is efficacious in rat and monkey models of Parkinson's

disease. Mov Disord 9:147-154.

Berg D, Becker G and Reiners K (1999) Reduction of dyskinesia and induction of akinesia induced

by morphine in two parkinsonian patients with severe sciatica. J Neural Transm (Vienna)

106:725-728.

Bespalov A, Steckler T, Altevogt B, Koustova E, Skolnick P, Deaver D, Millan MJ, Bastlund JF,

Doller D, Witkin J, Moser P, O'Donnell P, Ebert U, Geyer MA, Prinssen E, Ballard T and

Macleod M (2016) Failed trials for central nervous system disorders do not necessarily

invalidate preclinical models and drug targets. Nat Rev Drug Discov 15:516.

Bezard E, Brefel C, Tison F, Peyro-Saint-Paul H, Ladure P, Rascol O and Gross CE (1999) Effect

of the alpha 2 adrenoreceptor antagonist, idazoxan, on motor disabilities in MPTP-treated

monkey. Prog Neuropsychopharmacol Biol Psychiatry 23:1237-1246.

17 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Bezard E, Ferry S, Mach U, Stark H, Leriche L, Boraud T, Gross C and Sokoloff P (2003)

Attenuation of levodopa-induced dyskinesia by normalizing dopamine D3 receptor

function. Nat Med 9:762-767.

Bezard E, Hill MP, Crossman AR, Brotchie JM, Michel A, Grimee R and Klitgaard H (2004)

Levetiracetam improves choreic levodopa-induced dyskinesia in the MPTP-treated

macaque. Eur J Pharmacol 485:159-164.

Bezard E, Munoz A, Tronci E, Pioli EY, Li Q, Porras G, Bjorklund A and Carta M (2013a) Anti- Downloaded from

dyskinetic effect of anpirtoline in animal models of L-DOPA-induced dyskinesia. Neurosci

Res 77:242-246. jpet.aspetjournals.org Bezard E, Pioli EY, Li Q, Girard F, Mutel V, Keywood C, Tison F, Rascol O and Poli SM (2014)

The mGluR5 negative allosteric modulator dipraglurant reduces dyskinesia in the MPTP

macaque model. Mov Disord 29:1074-1079. at ASPET Journals on October 1, 2021 Bezard E, Tronci E, Pioli EY, Li Q, Porras G, Bjorklund A and Carta M (2013b) Study of the

antidyskinetic effect of eltoprazine in animal models of levodopa-induced dyskinesia. Mov

Disord 28:1088-1096.

Bibbiani F, Costantini LC, Patel R and Chase TN (2005a) Continuous dopaminergic stimulation

reduces risk of motor complications in parkinsonian primates. Exp Neurol 192:73-78.

Bibbiani F, Oh JD and Chase TN (2001) Serotonin 5-HT1A agonist improves motor complications

in rodent and primate parkinsonian models. Neurology 57:1829-1834.

Bibbiani F, Oh JD, Kielaite A, Collins MA, Smith C and Chase TN (2005b) Combined blockade

of AMPA and NMDA glutamate receptors reduces levodopa-induced motor complications

in animal models of PD. Exp Neurol 196:422-429.

18 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Bibbiani F, Oh JD, Petzer JP, Castagnoli N, Jr., Chen JF, Schwarzschild MA and Chase TN (2003)

A2A antagonist prevents -induced motor complications in animal models

of Parkinson's disease. Exp Neurol 184:285-294.

Blanchet P, Bedard PJ, Britton DR and Kebabian JW (1993) Differential effect of selective D-1

and D-2 agonists on levodopa-induced dyskinesia in 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine- exposed monkeys. J Pharmacol Exp Ther 267:275-279.

Blanchet PJ, Boucher R and Bedard PJ (1994) Excitotoxic lateral pallidotomy does not relieve L- Downloaded from

dopa-induced dyskinesia in MPTP parkinsonian monkeys. Brain Res 650:32-39.

Blanchet PJ, Calon F, Martel JC, Bedard PJ, Di Paolo T, Walters RR and Piercey MF (1995) jpet.aspetjournals.org Continuous administration decreases and pulsatile administration increases behavioral

sensitivity to a novel dopamine D2 agonist (U-91356A) in MPTP-exposed monkeys. J

Pharmacol Exp Ther 272:854-859. at ASPET Journals on October 1, 2021 Blanchet PJ, Grondin R and Bedard PJ (1996a) Dyskinesia and wearing-off following dopamine

D1 agonist treatment in drug-naive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned

primates. Mov Disord 11:91-94.

Blanchet PJ, Grondin R, Bedard PJ, Shiosaki K and Britton DR (1996b) Dopamine D1 receptor

desensitization profile in MPTP-lesioned primates. Eur J Pharmacol 309:13-20.

Blanchet PJ, Konitsiotis S and Chase TN (1997) Motor response to a dopamine D3 receptor

preferring agonist compared to in levodopa-primed 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine monkeys. J Pharmacol Exp Ther 283:794-799.

Blanchet PJ, Konitsiotis S and Chase TN (1998) reduces levodopa-induced

dyskinesias in parkinsonian monkeys. Mov Disord 13:798-802.

Blanchet PJ, Konitsiotis S, Whittemore ER, Zhou ZL, Woodward RM and Chase TN (1999)

Differing effects of N-methyl-D-aspartate receptor subtype selective antagonists on

19 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

dyskinesias in levodopa-treated 1-methyl-4-phenyl-tetrahydropyridine monkeys. J

Pharmacol Exp Ther 290:1034-1040.

Boman K and Meurman T (1970) Investigations on the effect of some drugs on the Parkinsonian

rigidity. Acta Neurol Scand 46:71-84.

Borgohain R, Szasz J, Stanzione P, Meshram C, Bhatt M, Chirilineau D, Stocchi F, Lucini V,

Giuliani R, Forrest E, Rice P, Anand R and Study I (2014a) Randomized trial of

add-on to levodopa in Parkinson's disease with motor fluctuations. Mov Disord 29:229-237. Downloaded from

Borgohain R, Szasz J, Stanzione P, Meshram C, Bhatt MH, Chirilineau D, Stocchi F, Lucini V,

Giuliani R, Forrest E, Rice P, Anand R and Study I (2014b) Two-year, randomized, jpet.aspetjournals.org controlled study of safinamide as add-on to levodopa in mid to late Parkinson's disease.

Mov Disord 29:1273-1280.

Braun A, Fabbrini G, Mouradian MM, Serrati C, Barone P and Chase TN (1987) Selective D-1 at ASPET Journals on October 1, 2021 dopamine receptor agonist treatment of Parkinson's disease. J Neural Transm 68:41-50.

Bronzova J, Sampaio C, Hauser RA, Lang AE, Rascol O, Theeuwes A, van de Witte SV, van

Scharrenburg G and Bruegel Study G (2010) Double-blind study of , a new

partial dopamine agonist, in early Parkinson's disease. Mov Disord 25:738-746.

Burns RS, Chiueh CC, Markey SP, Ebert MH, Jacobowitz DM and Kopin IJ (1983) A primate

model of parkinsonism: selective destruction of dopaminergic neurons in the pars compacta

of the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc Natl Acad

Sci U S A 80:4546-4550.

Butzer JF, Silver DE and Sahs AL (1975) Amantadine in Parkinson's disease. A double-blind,

placebo-controlled, crossover study with long-term follow-up. Neurology 25:603-606.

20 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Calon F, Goulet M, Blanchet PJ, Martel JC, Piercey MF, Bedard PJ and Di Paolo T (1995)

Levodopa or D2 agonist induced dyskinesia in MPTP monkeys: correlation with changes

in dopamine and GABAA receptors in the striatopallidal complex. Brain Res 680:43-52.

Calon F, Morissette M, Ghribi O, Goulet M, Grondin R, Blanchet PJ, Bedard PJ and Di Paolo T

(2002) Alteration of glutamate receptors in the striatum of dyskinetic 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine-treated monkeys following dopamine agonist treatment. Prog

Neuropsychopharmacol Biol Psychiatry 26:127-138. Downloaded from

Calon F, Morissette M, Goulet M, Grondin R, Blanchet PJ, Bedard PJ and Di Paolo T (1999)

Chronic D1 and D2 dopaminomimetic treatment of MPTP-denervated monkeys: effects on jpet.aspetjournals.org basal ganglia GABA(A)/benzodiazepine receptor complex and GABA content. Neurochem

Int 35:81-91.

Calon F, Morissette M, Goulet M, Grondin R, Blanchet PJ, Bedard PJ and Di Paolo T (2000) 125I- at ASPET Journals on October 1, 2021 CGP 64213 binding to GABA(B) receptors in the brain of monkeys: effect of MPTP and

dopaminomimetic treatments. Exp Neurol 163:191-199.

Cao X, Liang L, Hadcock JR, Iredale PA, Griffith DA, Menniti FS, Factor S, Greenamyre JT and

Papa SM (2007) Blockade of cannabinoid type 1 receptors augments the antiparkinsonian

action of levodopa without affecting dyskinesias in 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine-treated rhesus monkeys. J Pharmacol Exp Ther 323:318-326.

Carpentier AF, Bonnet AM, Vidailhet M and Agid Y (1996) Improvement of levodopa-induced

dyskinesia by propranolol in Parkinson's disease. Neurology 46:1548-1551.

Castro-Caldas A, Delwaide P, Jost W, Merello M, Williams A, Lamberti P, Aguilar M, Del Signore

S, Cesaro P and Parkinson-Control Study G (2006) The Parkinson-Control study: a 1-year

randomized, double-blind trial comparing (150 mg/day) with bromocriptine (25

21 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

mg/day) in early combination with levodopa in Parkinson's disease. Mov Disord 21:500-

509.

Cattaneo C, Ferla RL, Bonizzoni E and Sardina M (2015) Long-Term Effects of Safinamide on

Dyskinesia in Mid- to Late-Stage Parkinson's Disease: A Post-Hoc Analysis. J Parkinsons

Dis 5:475-481.

Close SP, Elliott PJ, Hayes AG and Marriott AS (1990) Effects of classical and novel agents in a

MPTP-induced reversible model of Parkinson's disease. Psychopharmacology (Berl) Downloaded from

102:295-300.

Clough CG, Bergmann KJ and Yahr MD (1984) Cholinergic and dopaminergic mechanisms in jpet.aspetjournals.org Parkinson's disease after long-term L-DOPA administration. Adv Neurol 40:131-140.

Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G and Pangalos MN (2014)

Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional at ASPET Journals on October 1, 2021 framework. Nat Rev Drug Discov 13:419-431.

Coppen A, Metcalfe M, Carroll JD and Morris JG (1972) Levodopa and L-tryptophan therapy in

Parkinsonism. Lancet 1:654-658.

Corsini GU, Del Zompo M, Gessa GL and Mangoni A (1979) Therapeutic efficacy of apomorphine

combined with an extracerebral inhibitor of dopamine receptors in Parkinson's disease.

Lancet 1:954-956.

Cox H, Togasaki DM, Chen L, Langston JW, Di Monte DA and Quik M (2007) The selective

kappa- receptor agonist U50,488 reduces L-dopa-induced dyskinesias but worsens

parkinsonism in MPTP-treated primates. Exp Neurol 205:101-107.

Cummings J, Isaacson S, Mills R, Williams H, Chi-Burris K, Corbett A, Dhall R and Ballard C

(2014) Pimavanserin for patients with Parkinson's disease psychosis: a randomised,

placebo-controlled phase 3 trial. Lancet 383:533-540.

22 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Davis GC, Williams AC, Markey SP, Ebert MH, Caine ED, Reichert CM and Kopin IJ (1979)

Chronic Parkinsonism secondary to intravenous injection of meperidine analogues.

Psychiatry Res 1:249-254.

Del Dotto P, Pavese N, Gambaccini G, Bernardini S, Metman LV, Chase TN and Bonuccelli U

(2001) Intravenous amantadine improves levadopa-induced dyskinesias: an acute double-

blind placebo-controlled study. Mov Disord 16:515-520.

Deuschl G, Vaitkus A, Fox GC, Roscher T, Schremmer D, Gordin A and Group CS (2007) Efficacy Downloaded from

and tolerability of versus Cabergoline in parkinsonian patients suffering from

wearing-off. Mov Disord 22:1550-1555. jpet.aspetjournals.org Di Paolo T, Bedard P, Daigle M and Boucher R (1986) Long-term effects of MPTP on central and

peripheral catecholamine and indoleamine concentrations in monkeys. Brain Res 379:286-

293. at ASPET Journals on October 1, 2021 Di Paolo T, Gregoire L, Feuerbach D, Elbast W, Weiss M and Gomez-Mancilla B (2014)

AQW051, a novel and selective nicotinic receptor alpha7 partial agonist,

reduces l-Dopa-induced dyskinesias and extends the duration of l-Dopa effects in

parkinsonian monkeys. Parkinsonism Relat Disord 20:1119-1123.

Doan VD, Grondin R, Hadj Tahar A, Gregoire L and Bedard PJ (1999) Effect of the selective D1

antagonists SCH 23390 and NNC 01-0112 on the delay, duration, and improvement of

behavioral responses to dopaminergic agents in MPTP-treated monkeys. Clin

Neuropharmacol 22:281-287.

Domino EF and Ni L (1998a) interactions with the dopamine D1-selective

receptor agonist SKF-82958 and the D2-selective receptor agonist N-0923 in 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine-induced hemiparkinsonian monkeys. J Pharmacol Exp

Ther 284:307-311.

23 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Domino EF and Ni L (1998b) Trihexyphenidyl potentiation of L-DOPA: reduced effectiveness

three years later in MPTP-induced chronic hemiparkinsonian monkeys. Exp Neurol

152:238-242.

Doshay LJ (1956) Five-year study of benztropine (cogentin) methanesulfonate; outcome in three

hundred two cases of paralysis agitans. J Am Med Assoc 162:1031-1034.

Durif F, Debilly B, Galitzky M, Morand D, Viallet F, Borg M, Thobois S, Broussolle E and Rascol

O (2004) Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo- Downloaded from

controlled study. Neurology 62:381-388.

Durif F, Vidailhet M, Assal F, Roche C, Bonnet AM and Agid Y (1997) Low-dose clozapine jpet.aspetjournals.org improves dyskinesias in Parkinson's disease. Neurology 48:658-662.

Duvoisin RC (1967) Cholinergic- antagonism in parkinsonism. Arch Neurol

17:124-136. at ASPET Journals on October 1, 2021 Ekesbo A, Andren PE, Gunne LM and Tedroff J (1997) (-)-OSU 6162 inhibits levodopa-induced

dyskinesias in a monkey model of Parkinson's disease. Neuroreport 8:2567-2570.

Emre M, Rinne UK, Rascol A, Lees A, Agid Y and Lataste X (1992) Effects of a selective partial

D1 agonist, CY 208-243, in de novo patients with Parkinson disease. Mov Disord 7:239-

243.

Factor SA, Wolski K, Togasaki DM, Huyck S, Cantillon M, Ho TW, Hauser RA and Pourcher E

(2013) Long-term safety and efficacy of preladenant in subjects with fluctuating

Parkinson's disease. Mov Disord 28:817-820.

Falardeau P, Bouchard S, Bedard PJ, Boucher R and Di Paolo T (1988) Behavioral and biochemical

effect of chronic treatment with D-1 and/or D-2 dopamine agonists in MPTP monkeys. Eur

J Pharmacol 150:59-66.

24 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Fenelon G, Gimenez-Roldan S, Montastruc JL, Bermejo F, Durif F, Bourdeix I, Pere JJ, Galiano

L and Schadrack J (2003) Efficacy and tolerability of entacapone in patients with

Parkinson's disease treated with levodopa plus a dopamine agonist and experiencing

wearing-off motor fluctuations. A randomized, double-blind, multicentre study. J Neural

Transm 110:239-251.

Fernandez HH, Friedman JH, Jacques C and Rosenfeld M (1999) Quetiapine for the treatment of

drug-induced psychosis in Parkinson's disease. Mov Disord 14:484-487. Downloaded from

Fernandez HH, Trieschmann ME, Burke MA, Jacques C and Friedman JH (2003) Long-term

outcome of quetiapine use for psychosis among Parkinsonian patients. Mov Disord 18:510- jpet.aspetjournals.org 514.

Fidalgo C, Ko WK, Tronci E, Li Q, Stancampiano R, Chuan Q, Bezard E and Carta M (2015)

Effect of blockade on L-DOPA-induced dyskinesia in animal models at ASPET Journals on October 1, 2021 of Parkinson's disease. Neuroscience 298:389-396.

Filion M, Tremblay L and Bedard PJ (1991) Effects of dopamine agonists on the spontaneous

activity of globus pallidus neurons in monkeys with MPTP-induced parkinsonism. Brain

Res 547:152-161.

Filipova M, Filip V, Macek Z, Mullerova S, Markova J, Kas S, Zizkova B, Krivka J, Votavova M

and Krejcova H (1988) Terguride in parkinsonism. A multicenter trial. Eur Arch Psychiatry

Neurol Sci 237:298-303.

Fox S, Silverdale M, Kellett M, Davies R, Steiger M, Fletcher N, Crossman A and Brotchie J

(2004) Non-subtype-selective opioid receptor antagonism in treatment of levodopa-induced

motor complications in Parkinson's disease. Mov Disord 19:554-560.

25 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Fox SH, Henry B, Hill M, Crossman A and Brotchie J (2002) Stimulation of cannabinoid receptors

reduces levodopa-induced dyskinesia in the MPTP-lesioned nonhuman primate model of

Parkinson's disease. Mov Disord 17:1180-1187.

Fox SH, Henry B, Hill MP, Peggs D, Crossman AR and Brotchie JM (2001) Neural mechanisms

underlying peak-dose dyskinesia induced by levodopa and apomorphine are distinct:

evidence from the effects of the alpha(2) adrenoceptor antagonist idazoxan. Mov Disord

16:642-650. Downloaded from

Fox SH, Lang AE and Brotchie JM (2006a) Translation of nondopaminergic treatments for

levodopa-induced dyskinesia from MPTP-lesioned nonhuman primates to phase IIa clinical jpet.aspetjournals.org studies: keys to success and roads to failure. Mov Disord 21:1578-1594.

Fox SH, Visanji NP, Johnston TH, Gomez-Ramirez J, Voon V and Brotchie JM (2006b) Dopamine

receptor agonists and levodopa and inducing psychosis-like behavior in the MPTP primate at ASPET Journals on October 1, 2021 model of Parkinson disease. Arch Neurol 63:1343-1344.

Frackiewicz EJ, Jhee SS, Shiovitz TM, Webster J, Topham C, Dockens RC, Whigan D, Salazar

DE and Cutler NR (2002) treatment for Parkinson's disease in combination

with levodopa/carbidopa. Ann Pharmacother 36:225-230.

FrenchClozapineParkinsonStudyGroup (1999) Clozapine in drug-induced psychosis in Parkinson's

disease. The French Clozapine Parkinson Study Group. Lancet 353:2041-2042.

Gagnon C, Bedard PJ and Di Paolo T (1990) Effect of chronic treatment of MPTP monkeys with

dopamine D-1 and/or D-2 receptor agonists. Eur J Pharmacol 178:115-120.

Gagnon C, Gomez-Mancilla B, Bedard PJ and Di Paolo T (1993) Chronic CY 208-243 treatment

of MPTP-monkeys causes regional changes of dopamine and GABAA receptors. Neurosci

Lett 163:31-35.

26 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Gagnon C, Gomez-Mancilla B, Markstein R, Bedard PJ and Di Paolo T (1995) Effect of adding

the D-1 agonist CY 208-243 to chronic bromocriptine treatment of MPTP-monkeys:

regional changes of brain dopamine receptors. Prog Neuropsychopharmacol Biol

Psychiatry 19:667-676.

Galvan A, Devergnas A, Pittard D, Masilamoni G, Vuong J, Daniels JS, Morrison RD, Lindsley

CW and Wichmann T (2016) Lack of Antiparkinsonian Effects of Systemic Injections of

the Specific T-Type Calcium Channel Blocker ML218 in MPTP-Treated Monkeys. ACS Downloaded from

Chem Neurosci.

Gerstenbrand F, Binder H, Kozma C, Pusch ST and Reisner T (1975) [Infusion therapy with mif jpet.aspetjournals.org (melanocyte inhibiting factor) in Parkinson's disease (author's transl)]. Wien Klin

Wochenschr 87:822-823.

Giladi N, Boroojerdi B, Korczyn AD, Burn DJ, Clarke CE and Schapira AH (2007) at ASPET Journals on October 1, 2021 transdermal patch in early Parkinson's disease: a randomized, double-blind, controlled

study versus placebo and ropinirole. Mov Disord 22:2398-2404.

Gnanalingham KK, Erol DD, Hunter AJ, Smith LA, Jenner P and Marsden CD (1995a) Differential

anti-parkinsonian effects of benzazepine D1 dopamine agonists with varying efficacies in

the MPTP-treated common marmoset. Psychopharmacology (Berl) 117:275-286.

Gnanalingham KK, Hunter AJ, Jenner P and Marsden CD (1995b) The differential behavioural

effects of benzazepine D1 dopamine agonists with varying efficacies, co-administered with

quinpirole in primate and rodent models of Parkinson's disease. Psychopharmacology

(Berl) 117:287-297.

Gnanalingham KK, Hunter AJ, Jenner P and Marsden CD (1995c) Selective dopamine antagonist

pretreatment on the antiparkinsonian effects of benzazepine D1 dopamine agonists in

27 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

rodent and primate models of Parkinson's disease--the differential effects of D1 dopamine

antagonists in the primate. Psychopharmacology (Berl) 117:403-412.

Goetz CG, Damier P, Hicking C, Laska E, Muller T, Olanow CW, Rascol O and Russ H (2007)

Sarizotan as a treatment for dyskinesias in Parkinson's disease: a double-blind placebo-

controlled trial. Mov Disord 22:179-186.

Goetz CG, Laska E, Hicking C, Damier P, Muller T, Nutt J, Warren Olanow C, Rascol O and Russ

H (2008) Placebo influences on dyskinesia in Parkinson's disease. Mov Disord 23:700-707. Downloaded from

Goetz CG, Stebbins GT, Chung KA, Nicholas AP, Hauser RA, Merkitch D and Stacy MA (2017)

Topiramate as an adjunct to amantadine in the treatment of dyskinesia in parkinson's jpet.aspetjournals.org disease: A randomized, double-blind, placebo-controlled multicenter study. Mov Disord

32:1335-1336.

Goetz CG, Tanner CM and Klawans HL (1984) in Parkinson's disease. Neurology at ASPET Journals on October 1, 2021 34:1092-1094.

Gomez-Mancilla B and Bedard PJ (1991) Effect of D1 and D2 agonists and antagonists on

dyskinesia produced by L-dopa in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated

monkeys. J Pharmacol Exp Ther 259:409-413.

Gomez-Mancilla B and Bedard PJ (1992a) Effect of chronic treatment with (+)-PHNO, a D2

agonist in MPTP-treated monkeys. Exp Neurol 117:185-188.

Gomez-Mancilla B and Bedard PJ (1992b) Effect of estrogen and progesterone on L-dopa induced

dyskinesia in MPTP-treated monkeys. Neurosci Lett 135:129-132.

Gomez-Mancilla B and Bedard PJ (1993) Effect of nondopaminergic drugs on L-dopa-induced

dyskinesias in MPTP-treated monkeys. Clin Neuropharmacol 16:418-427.

Gomez-Mancilla B, Boucher R and Bedard PJ (1991) Effect of clonidine and on rest

tremor in the MPTP monkey model of parkinsonism. Clin Neuropharmacol 14:359-366.

28 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Gomez-Mancilla B, Boucher R and Bedard PJ (1992a) Effect of LY 171555 and CY 208-243 on

tremor suppression in the MPTP monkey model of parkinsonism. Mov Disord 7:43-47.

Gomez-Mancilla B, Boucher R, Gagnon C, Di Paolo T, Markstein R and Bedard PJ (1993) Effect

of adding the D1 agonist CY 208-243 to chronic bromocriptine treatment. I: Evaluation of

motor parameters in relation to striatal catecholamine content and dopamine receptors. Mov

Disord 8:144-150.

Gomez-Mancilla B, Latulippe JF, Boucher R and Bedard PJ (1992b) Effect of ethosuximide on Downloaded from

rest tremor in the MPTP monkey model. Mov Disord 7:137-141.

Gomez-Ramirez J, Johnston TH, Visanji NP, Fox SH and Brotchie JM (2006) H3 jpet.aspetjournals.org receptor agonists reduce L-dopa-induced chorea, but not dystonia, in the MPTP-lesioned

nonhuman primate model of Parkinson's disease. Mov Disord 21:839-846.

Gordon PH, Pullman SL, Louis ED, Frucht SJ and Fahn S (2002) Mirtazapine in Parkinsonian at ASPET Journals on October 1, 2021 tremor. Parkinsonism Relat Disord 9:125-126.

Goulet M, Grondin R, Blanchet PJ, Bedard PJ and Di Paolo T (1996) Dyskinesias and tolerance

induced by chronic treatment with a D1 agonist administered in pulsatile or continuous

mode do not correlate with changes of putaminal D1 receptors in drug-naive MPTP

monkeys. Brain Res 719:129-137.

Goulet M, Grondin R, Morissette M, Maltais S, Falardeau P, Bedard PJ and Di Paolo T (2000)

Regulation by chronic treatment with cabergoline of dopamine D1 and D2 receptor levels

and their expression in the striatum of Parkinsonian-monkeys. Prog

Neuropsychopharmacol Biol Psychiatry 24:607-617.

Goulet M, Morissette M, Calon F, Blanchet PJ, Falardeau P, Bedard PJ and Di Paolo T (1997)

Continuous or pulsatile chronic D2 dopamine receptor agonist (U91356A) treatment of

drug-naive 4-phenyl-1,2,3,6-tetrahydropyridine monkeys differentially regulates brain D1

29 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

and D2 receptor expression: in situ hybridization histochemical analysis. Neuroscience

79:497-507.

Goulet M, Morissette M, Grondin R, Falardeau P, Bedard PJ, Rostene W and Di Paolo T (1999)

Neurotensin receptors and dopamine transporters: effects of MPTP lesioning and chronic

dopaminergic treatments in monkeys. Synapse 32:153-164.

Grandas F, Quinn N, Critchley P, Rohan A, Marsden CD and Stahl SM (1987) Antiparkinsonian

activity of a single oral dose of PHNO. Mov Disord 2:47-51. Downloaded from

Greenamyre JT, Eller RV, Zhang Z, Ovadia A, Kurlan R and Gash DM (1994) Antiparkinsonian

effects of hydrochloride, a glutamate antagonist, in rodent and primate models jpet.aspetjournals.org of Parkinson's disease. Ann Neurol 35:655-661.

Gregoire L, Jourdain VA, Townsend M, Roach A and Di Paolo T (2013) Safinamide reduces

dyskinesias and prolongs L-DOPA antiparkinsonian effect in parkinsonian monkeys. at ASPET Journals on October 1, 2021 Parkinsonism Relat Disord 19:508-514.

Gregoire L, Morin N, Ouattara B, Gasparini F, Bilbe G, Johns D, Vranesic I, Sahasranaman S,

Gomez-Mancilla B and Di Paolo T (2011) The acute antiparkinsonian and antidyskinetic

effect of AFQ056, a novel metabotropic type 5 antagonist, in l-Dopa-

treated parkinsonian monkeys. Parkinsonism Relat Disord.

Gregoire L, Rassoulpour A, Guidetti P, Samadi P, Bedard PJ, Izzo E, Schwarcz R and Di Paolo T

(2008) Prolonged 3-hydroxylase inhibition reduces development of levodopa-

induced dyskinesias in parkinsonian monkeys. Behav Brain Res 186:161-167.

Gregoire L, Samadi P, Graham J, Bedard PJ, Bartoszyk GD and Di Paolo T (2009) Low doses of

reduce dyskinesias and maintain antiparkinsonian efficacy of L-Dopa in

parkinsonian monkeys. Parkinsonism Relat Disord 15:445-452.

30 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Gregoire L, Smith T, Senanayake V, Mochizuki A, Miville-Godbout E, Goodenowe D and Di

Paolo T (2015) Plasmalogen precursor analog treatment reduces levodopa-induced

dyskinesias in parkinsonian monkeys. Behav Brain Res 286:328-337.

Grondin R, Bedard PJ, Britton DR and Shiosaki K (1997) Potential therapeutic use of the selective

dopamine D1 receptor agonist, A-86929: an acute study in parkinsonian levodopa-primed

monkeys. Neurology 49:421-426.

Grondin R, Bedard PJ, Hadj Tahar A, Gregoire L, Mori A and Kase H (1999a) Antiparkinsonian Downloaded from

effect of a new selective adenosine A2A receptor antagonist in MPTP-treated monkeys.

Neurology 52:1673-1677. jpet.aspetjournals.org Grondin R, Doan VD, Gregoire L and Bedard PJ (1999b) D1 receptor blockade improves L-dopa-

induced dyskinesia but worsens parkinsonism in MPTP monkeys. Neurology 52:771-776.

Grondin R, Goulet M, Di Paolo T and Bedard PJ (1996) Cabergoline, a long-acting dopamine D2- at ASPET Journals on October 1, 2021 like receptor agonist, produces a sustained antiparkinsonian effect with transient

dyskinesias in parkinsonian drug-naive primates. Brain Res 735:298-306.

Grondin R, Goulet M, Morissette M, Bedard PJ and Di Paolo T (1999c) Dopamine D1 receptor

mRNA and receptor levels in the striatum of MPTP monkeys chronically treated with SKF-

82958. Eur J Pharmacol 378:259-263.

Grondin R, Hadj Tahar A, Doan VD, Ladure P and Bedard PJ (2000) Noradrenoceptor antagonism

with idazoxan improves L-dopa-induced dyskinesias in MPTP monkeys. Naunyn

Schmiedebergs Arch Pharmacol 361:181-186.

Gruchet R (1952) [ in parkinsonism]. Therapie 7:114-121.

Hadj Tahar A, Belanger N, Bangassoro E, Gregoire L and Bedard PJ (2000a) Antidyskinetic effect

of JL-18, a clozapine analog, in parkinsonian monkeys. Eur J Pharmacol 399:183-186.

31 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Hadj Tahar A, Ekesbo A, Gregoire L, Bangassoro E, Svensson KA, Tedroff J and Bedard PJ (2001)

Effects of acute and repeated treatment with a novel dopamine D2 receptor ligand on L-

DOPA-induced dyskinesias in MPTP monkeys. Eur J Pharmacol 412:247-254.

Hadj Tahar A, Gregoire L, Bangassoro E and Bedard PJ (2000b) Sustained cabergoline treatment

reverses levodopa-induced dyskinesias in parkinsonian monkeys. Clin Neuropharmacol

23:195-202.

Hadj Tahar A, Gregoire L, Darre A, Belanger N, Meltzer L and Bedard PJ (2004) Effect of a Downloaded from

selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian

monkeys. Neurobiol Dis 15:171-176. jpet.aspetjournals.org Hamadjida A, Nuara SG, Gourdon JC and Huot P (2018) The effect of mianserin on the severity

of psychosis and dyskinesia in the parkinsonian marmoset. Prog Neuropsychopharmacol

Biol Psychiatry 81:367-371. at ASPET Journals on October 1, 2021 Hamadjida A, Nuara SG, Veyres N, Frouni I, Kwan C, Sid-Otmane L, Harraka MJ, Gourdon JC

and Huot P (2017) The effect of mirtazapine on dopaminergic psychosis and dyskinesia in

the parkinsonian marmoset. Psychopharmacology (Berl) 234:905-911.

Hansard MJ, Jackson MJ, Smith LA, Rose S and Jenner P (2011) A major metabolite of bupropion

reverses motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated common

marmosets. Behav Pharmacol 22:269-274.

Hansard MJ, Smith LA, Jackson MJ, Cheetham SC and Jenner P (2002a) Dopamine reuptake

inhibition and failure to evoke dyskinesia in MPTP-treated primates. Eur J Pharmacol

451:157-160.

Hansard MJ, Smith LA, Jackson MJ, Cheetham SC and Jenner P (2002b) Dopamine, but not

norepinephrine or serotonin, reuptake inhibition reverses motor deficits in 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine-treated primates. J Pharmacol Exp Ther 303:952-958.

32 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Hansard MJ, Smith LA, Jackson MJ, Cheetham SC and Jenner P (2004) The monoamine reuptake

inhibitor BTS 74 398 fails to evoke established dyskinesia but does not synergise with

levodopa in MPTP-treated primates. Mov Disord 19:15-21.

Hauser RA, Cantillon M, Pourcher E, Micheli F, Mok V, Onofrj M, Huyck S and Wolski K (2011)

Preladenant in patients with Parkinson's disease and motor fluctuations: a phase 2, double-

blind, randomised trial. Lancet Neurol 10:221-229.

Hauser RA, Hubble JP and Truong DD (2003) Randomized trial of the adenosine A(2A) receptor Downloaded from

antagonist in advanced PD. Neurology 61:297-303.

Hauser RA, Shulman LM, Trugman JM, Roberts JW, Mori A, Ballerini R and Sussman NM (2008) jpet.aspetjournals.org Study of istradefylline in patients with Parkinson's disease on levodopa with motor

fluctuations. Mov Disord 23:2177-2185.

Hauser RA, Stocchi F, Rascol O, Huyck SB, Capece R, Ho TW, Sklar P, Lines C, Michelson D at ASPET Journals on October 1, 2021 and Hewitt D (2015) Preladenant as an Adjunctive Therapy With Levodopa in Parkinson

Disease: Two Randomized Clinical Trials and Lessons Learned. JAMA Neurol 72:1491-

1500.

Hauser RA and Zesiewicz TA (1997) Sertraline for the treatment of depression in Parkinson's

disease. Mov Disord 12:756-759.

Hely MA, Morris JG, Reid WG, O'Sullivan DJ, Williamson PM, Rail D, Broe GA and Margrie S

(1994) The Sydney Multicentre Study of Parkinson's disease: a randomised, prospective

five year study comparing low dose bromocriptine with low dose levodopa-carbidopa. J

Neurol Neurosurg Psychiatry 57:903-910.

Henry B, Fox SH, Crossman AR and Brotchie JM (2001) Mu- and delta-opioid receptor antagonists

reduce levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson's

disease. Exp Neurol 171:139-146.

33 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Henry B, Fox SH, Peggs D, Crossman AR and Brotchie JM (1999) The alpha2-adrenergic receptor

antagonist idazoxan reduces dyskinesia and enhances anti-parkinsonian actions of L-dopa

in the MPTP-lesioned primate model of Parkinson's disease. Mov Disord 14:744-753.

Hill MP, Bezard E, McGuire SG, Crossman AR, Brotchie JM, Michel A, Grimee R and Klitgaard

H (2003) Novel antiepileptic drug levetiracetam decreases dyskinesia elicited by L-dopa

and ropinirole in the MPTP-lesioned marmoset. Mov Disord 18:1301-1305.

Hill MP, Brotchie JM, Crossman AR, Bezard E, Michel A, Grimee R and Klitgaard H (2004a) Downloaded from

Levetiracetam Interferes With the L-dopa priming process in MPTP-lesioned drug-naive

marmosets. Clin Neuropharmacol 27:171-177. jpet.aspetjournals.org Hill MP, Ravenscroft P, Bezard E, Crossman AR, Brotchie JM, Michel A, Grimee R and Klitgaard

H (2004b) Levetiracetam potentiates the antidyskinetic action of amantadine in the 1-

methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate model of at ASPET Journals on October 1, 2021 Parkinson's disease. J Pharmacol Exp Ther 310:386-394.

Hill MP, Ravenscroft P, McGuire SG, Brotchie JM, Crossman AR, Rochat C and Millan MJ (2006)

Antiparkinsonian effects of the novel D3/D2 dopamine receptor agonist, S32504, in MPTP-

lesioned marmosets: Mediation by D2, not D3, dopamine receptors. Mov Disord 21:2090-

2095.

Hille CJ, Fox SH, Maneuf YP, Crossman AR and Brotchie JM (2001) Antiparkinsonian action of

a delta opioid agonist in rodent and primate models of Parkinson's disease. Exp Neurol

172:189-198.

Hodgson RA, Bedard PJ, Varty GB, Kazdoba TM, Di Paolo T, Grzelak ME, Pond AJ, Hadjtahar

A, Belanger N, Gregoire L, Dare A, Neustadt BR, Stamford AW and Hunter JC (2010)

Preladenant, a selective A(2A) receptor antagonist, is active in primate models of

movement disorders. Exp Neurol 225:384-390.

34 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Hsu A, Togasaki DM, Bezard E, Sokoloff P, Langston JW, Di Monte DA and Quik M (2004)

Effect of the D3 dopamine receptor partial agonist BP897 [N-[4-(4-(2-

methoxyphenyl)piperazinyl)butyl]-2-naphthamide] on L-3,4-dihydroxyphenylalanine-

induced dyskinesias and parkinsonism in squirrel monkeys. J Pharmacol Exp Ther

311:770-777.

Huot P, Johnston TH, Fox SH and Brotchie JM (2015a) Pioglitazone may impair L-DOPA anti-

parkinsonian efficacy in the MPTP-lesioned macaque: results of a pilot study. Synapse Downloaded from

69:99-102.

Huot P, Johnston TH, Fox SH, Newman-Tancredi A and Brotchie JM (2015b) The highly-selective jpet.aspetjournals.org 5-HT(1A) agonist F15599 reduces L-DOPA-induced dyskinesia without compromising

anti-parkinsonian benefits in the MPTP-lesioned macaque. Neuropharmacology 97:306-

311. at ASPET Journals on October 1, 2021 Huot P, Johnston TH, Gandy MN, Reyes MG, Fox SH, Piggott MJ and Brotchie JM (2012a) The

Monoamine Re-Uptake Inhibitor UWA-101 Improves Motor Fluctuations in the MPTP-

Lesioned Common Marmoset. PLOS ONE 7:e45587.

Huot P, Johnston TH, Koprich JB, Aman A, Fox SH and Brotchie JM (2012b) L-745,870 reduces

L-DOPA-induced dyskinesia in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned

macaque model of Parkinson's disease. J Pharmacol Exp Ther 342:576-585.

Huot P, Johnston TH, Lewis KD, Koprich JB, Reyes MG, Fox SH, Piggott MJ and Brotchie JM

(2011) Characterization of 3,4-Methylenedioxymethamphetamine (MDMA) Enantiomers

In Vitro and in the MPTP-Lesioned Primate: R-MDMA Reduces Severity of Dyskinesia,

Whereas S-MDMA Extends Duration of ON-Time. J Neurosci 31:7190-7198.

Huot P, Johnston TH, Lewis KD, Koprich JB, Reyes MG, Fox SH, Piggott MJ and Brotchie JM

(2014) UWA-121, a mixed dopamine and serotonin re-uptake inhibitor, enhances L-DOPA

35 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

anti-parkinsonian action without worsening dyskinesia or psychosis-like behaviours in the

MPTP-lesioned common marmoset. Neuropharmacology 82:76-87.

Huot P, Johnston TH, Snoeren T, Koprich JB, Hill MP, Fox SH and Brotchie JM (2013) Use of

catechol-O-methyltransferase inhibition to minimize L-3,4-dihydroxyphenylalanine-

induced dyskinesia in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque.

Eur J Neurosci 37:831-838.

Huot P, Levesque M, Morissette M, Calon F, Dridi M, Di Paolo T and Parent A (2008) L-Dopa Downloaded from

treatment abolishes the numerical increase in striatal dopaminergic neurons in parkinsonian

monkeys. J Chem Neuroanat 35:77-84. jpet.aspetjournals.org Hyacinthe C, Barraud Q, Tison F, Bezard E and Ghorayeb I (2014) D1 receptor agonist improves

sleep-wake parameters in experimental parkinsonism. Neurobiol Dis 63:20-24.

Ikeguchi K and Kuroda A (1995) Mianserin treatment of patients with psychosis induced by at ASPET Journals on October 1, 2021 antiparkinsonian drugs. Eur Arch Psychiatry Clin Neurosci 244:320-324.

Investigators NETiPDF-Z (2015) Pioglitazone in early Parkinson's disease: a phase 2, multicentre,

double-blind, randomised trial. Lancet Neurol 14:795-803.

Iravani MM, Jackson MJ, Kuoppamaki M, Smith LA and Jenner P (2003) 3,4-

methylenedioxymethamphetamine (ecstasy) inhibits dyskinesia expression and normalizes

motor activity in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated primates. J

Neurosci 23:9107-9115.

Iravani MM, Tayarani-Binazir K, Chu WB, Jackson MJ and Jenner P (2006) In 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine-treated primates, the selective 5-hydroxytryptamine 1a agonist

(R)-(+)-8-OHDPAT inhibits levodopa-induced dyskinesia but only with\ increased motor

disability. J Pharmacol Exp Ther 319:1225-1234.

36 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Jackson MJ, Al-Barghouthy G, Pearce RK, Smith L, Hagan JJ and Jenner P (2004) Effect of 5-

HT1B/D receptor agonist and antagonist administration on motor function in

and MPTP-treated common marmosets. Pharmacol Biochem Behav 79:391-400.

Jackson MJ, Andree TH, Hansard M, Hoffman DC, Hurtt MR, Kehne JH, Pitler TA, Smith LA,

Stack G and Jenner P (2010) The dopamine D(2) receptor partial agonist

improves motor deficits in MPTP-treated common marmosets alone and combined with L-

dopa. J Neural Transm (Vienna) 117:55-67. Downloaded from

Jackson MJ, Smith LA, Al-Barghouthy G, Rose S and Jenner P (2007) Decreased expression of l-

dopa-induced dyskinesia by switching to ropinirole in MPTP-treated common marmosets. jpet.aspetjournals.org Exp Neurol 204:162-170.

Jackson MJ, Swart T, Pearce RK and Jenner P (2014) Cholinergic manipulation of motor disability

and L-DOPA-induced dyskinesia in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine at ASPET Journals on October 1, 2021 (MPTP)-treated common marmosets. J Neural Transm (Vienna) 121:163-169.

Jan C, Francois C, Tande D, Yelnik J, Tremblay L, Agid Y and Hirsch E (2000) Dopaminergic

innervation of the pallidum in the normal state, in MPTP-treated monkeys and in

parkinsonian patients. Eur J Neurosci 12:4525-4535.

Jellinger K (1982) Adjuvant treatment of Parkinson's disease with dopamine agonists: open trial

with bromocriptine and CU 32-085. J Neurol 227:75-88.

Jenner P, Rupniak NM, Rose S, Kelly E, Kilpatrick G, Lees A and Marsden CD (1984) 1-Methyl-

4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism in the common marmoset.

Neurosci Lett 50:85-90.

Jenner P, Zeng BY, Smith LA, Pearce RK, Tel B, Chancharme L and Moachon G (2000)

Antiparkinsonian and neuroprotective effects of in the mptp-treated common

marmoset. Exp Brain Res 133:178-188.

37 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Johnston LC, Jackson MJ, Rose S, McCreary AC and Jenner P (2010a) Pardoprunox reverses

motor deficits but induces only mild dyskinesia in MPTP-treated common marmosets. Mov

Disord.

Johnston TH, Fox SH, McIldowie MJ, Piggott MJ and Brotchie JM (2010b) Reduction of L-

DOPA-induced dyskinesia by the selective metabotropic glutamate receptor 5 antagonist

3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine in the 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine-lesioned macaque model of Parkinson's disease. J Pharmacol Exp Ther Downloaded from

333:865-873.

Johnston TH, Fox SH, Piggott MJ, Savola JM and Brotchie JM (2010c) The alpha adrenergic jpet.aspetjournals.org antagonist fipamezole improves quality of levodopa action in Parkinsonian primates. Mov

Disord 25:2084-2093.

Johnston TH, Huot P, Damude S, Fox SH, Jones SW, Rusche JR and Brotchie JM (2013a) at ASPET Journals on October 1, 2021 RGFP109, a histone deacetylase inhibitor attenuates L-DOPA-induced dyskinesia in the

MPTP-lesioned marmoset: a proof-of-concept study. Parkinsonism Relat Disord 19:260-

264.

Johnston TH, Huot P, Fox SH, Koprich JB, Szeliga KT, James JW, Graef JD, Letchworth SR,

Jordan KG, Hill MP and Brotchie JM (2013b) TC-8831, a nicotinic

agonist, reduces l-DOPA-induced dyskinesia in the MPTP macaque. Neuropharmacology

73C:337-347.

Johnston TH, Huot P, Fox SH, Wakefield JD, Sykes KA, Bartolini WP, Milne GT, Pearson JP and

Brotchie JM (2011) Fatty Acid Amide Hydrolase (FAAH) Inhibition Reduces L-3,4-

Dihydroxyphenylalanine-Induced Hyperactivity in the 1-Methyl-4-phenyl-1,2,3,6-

tetrahydropyridine-Lesioned Non-Human Primate Model of Parkinson's Disease. J

Pharmacol Exp Ther 336:423-430.

38 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Johnston TH, Millar Z, Huot P, Wagg K, Thiele S, Salomonczyk D, Yong-Kee CJ, Gandy MN,

McIldowie M, Lewis KD, Gomez-Ramirez J, Lee J, Fox SH, Martin-Iverson M, Nash JE,

Piggott MJ and Brotchie JM (2012) A novel MDMA analogue, UWA-101, that lacks

psychoactivity and cytotoxicity, enhances L-DOPA benefit in parkinsonian primates.

FASEB J 26:2154-2163.

Johnston TH, van der Meij A, Brotchie JM and Fox SH (2010d) Effect of

antagonism on levodopa-induced dyskinesia in the MPTP-macaque model of Parkinson's Downloaded from

disease. Mov Disord 25:1379-1390.

Jones CA, Johnston LC, Jackson MJ, Smith LA, van Scharrenburg G, Rose S, Jenner PG and jpet.aspetjournals.org McCreary AC (2010) An in vivo pharmacological evaluation of pardoprunox (SLV308)--a

novel combined dopamine D(2)/D(3) receptor partial agonist and 5-HT(1A) receptor

agonist with efficacy in experimental models of Parkinson's disease. Eur at ASPET Journals on October 1, 2021 Neuropsychopharmacol 20:582-593.

Kaakkola S, Teravainen H, Ahtila S, Rita H and Gordin A (1994) Effect of entacapone, a COMT

inhibitor, on clinical disability and levodopa metabolism in parkinsonian patients.

Neurology 44:77-80.

Kanda T, Jackson MJ, Smith LA, Pearce RK, Nakamura J, Kase H, Kuwana Y and Jenner P (1998a)

Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke

dyskinesia in parkinsonian monkeys. Ann Neurol 43:507-513.

Kanda T, Jackson MJ, Smith LA, Pearce RK, Nakamura J, Kase H, Kuwana Y and Jenner P (2000)

Combined use of the adenosine A(2A) antagonist KW-6002 with L-DOPA or with selective

D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in

MPTP-treated monkeys. Exp Neurol 162:321-327.

39 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Kanda T, Tashiro T, Kuwana Y and Jenner P (1998b) Adenosine A2A receptors modify motor

function in MPTP-treated common marmosets. Neuroreport 9:2857-2860.

Katzenschlager R, Jackson MJ, Rose S, Stockwell K, Tayarani-Binazir KA, Zubair M, Smith LA,

Jenner P and Lees AJ (2007) Antiparkinsonian activity of L-propyl-L-leucyl-glycinamide

or melanocyte-inhibiting factor in MPTP-treated common marmosets. Mov Disord 22:715-

719.

Katzenschlager R, Manson AJ, Evans A, Watt H and Lees AJ (2004) Low dose quetiapine for drug Downloaded from

induced dyskinesias in Parkinson's disease: a double blind cross over study. J Neurol

Neurosurg Psychiatry 75:295-297. jpet.aspetjournals.org Kebabian JW, Britton DR, DeNinno MP, Perner R, Smith L, Jenner P, Schoenleber R and Williams

M (1992) A-77636: a potent and selective dopamine D1 receptor agonist with

antiparkinsonian activity in marmosets. Eur J Pharmacol 229:203-209. at ASPET Journals on October 1, 2021 Klawans HL, Jr. and Weiner WJ (1974) Attempted use of haloperidol in the treatment of L-dopa

induced dyskinesias. J Neurol Neurosurg Psychiatry 37:427-430.

Klawans HL and Ringel SP (1973) A clinical study of methysergide in Parkinsonism: evidence

against a serotonergic mechanism. J Neurol Sci 19:399-405.

Klockgether T, Turski L, Honore T, Zhang ZM, Gash DM, Kurlan R and Greenamyre JT (1991)

The AMPA receptor antagonist NBQX has antiparkinsonian effects in monoamine-

depleted rats and MPTP-treated monkeys. Ann Neurol 30:717-723.

Ko WK, Camus SM, Li Q, Yang J, McGuire S, Pioli EY and Bezard E (2016) An evaluation of

istradefylline treatment on Parkinsonian motor and cognitive deficits in 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine (MPTP)-treated macaque models. Neuropharmacology 110:48-

58.

40 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Ko WK, Li Q and Bezard E (2014a) Effects of L-tryptophan on L-DOPA-induced dyskinesia in

the L-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaque model of

Parkinson's disease. Neurosci Lett 566:72-76.

Ko WK, Pioli E, Li Q, McGuire S, Dufour A, Sherer TB, Bezard E and Facheris MF (2014b)

Combined fenobam and amantadine treatment promotes robust antidyskinetic effects in the

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate model of

Parkinson's disease. Mov Disord 29:772-779. Downloaded from

Ko WKD, Li Q, Cheng LY, Morelli M, Carta M and Bezard E (2017) A preclinical study on the

combined effects of repeated eltoprazine and preladenant treatment for alleviating L- jpet.aspetjournals.org DOPA-induced dyskinesia in Parkinson's disease. Eur J Pharmacol 813:10-16.

Kobylecki C, Burn DJ, Kass-Iliyya L, Kellett MW, Crossman AR and Silverdale MA (2014)

Randomized clinical trial of for levodopa-induced dyskinesia in Parkinson's at ASPET Journals on October 1, 2021 disease. Parkinsonism Relat Disord 20:452-455.

Kobylecki C, Cenci MA, Crossman AR and Ravenscroft P (2010) Calcium-permeable AMPA

receptors are involved in the induction and expression of l-DOPA-induced dyskinesia in

Parkinson's disease. J Neurochem 114:499-511.

Kobylecki C, Hill MP, Crossman AR and Ravenscroft P (2011) Synergistic antidyskinetic effects

of topiramate and amantadine in animal models of Parkinson's disease. Mov Disord

26:2354-2363.

Koller WC and Herbster G (1987) Adjuvant therapy of parkinsonian tremor. Arch Neurol 44:921-

923.

Konitsiotis S, Blanchet PJ, Verhagen L, Lamers E and Chase TN (2000) AMPA receptor blockade

improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 54:1589-1595.

41 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Koprich JB, Fox SH, Johnston TH, Goodman A, Le Bourdonnec B, Dolle RE, Dehaven RN,

Dehaven-Hudkins DL, Little PJ and Brotchie JM (2011) The selective mu-opioid receptor

antagonist adl5510 reduces levodopa-induced dyskinesia without affecting

antiparkinsonian action in mptp-lesioned macaque model of Parkinson's disease. Mov

Disord 26:1225-1233.

Koprich JB, Huot P, Fox SH, Jarvie K, Lang AE, Seeman P and Brotchie JM (2013) The effects

of fast-off-D2 receptor antagonism on L-DOPA-induced dyskinesia and psychosis in Downloaded from

parkinsonian macaques. Prog Neuropsychopharmacol Biol Psychiatry 43:151-156.

Korczyn AD, Brunt ER, Larsen JP, Nagy Z, Poewe WH and Ruggieri S (1999) A 3-year jpet.aspetjournals.org randomized trial of ropinirole and bromocriptine in early Parkinson's disease. The 053

Study Group. Neurology 53:364-370.

Kulisevsky J, Pagonabarraga J, Pascual-Sedano B, Gironell A, Garcia-Sanchez C and Martinez- at ASPET Journals on October 1, 2021 Corral M (2008) Motor changes during sertraline treatment in depressed patients with

Parkinson's disease*. Eur J Neurol 15:953-959.

Lamid S and Jenkins RB (1975) Crossover clinical trial of benapryzine and trihexyphenidyl in

Parkinsonian patients. J Clin Pharmacol 15:622-626.

Lange KW, Loschmann PA, Wachtel H, Horowski R, Jahnig P, Jenner P and Marsden CD (1992)

Terguride stimulates locomotor activity at 2 months but not 10 months after 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine treatment of common marmosets. Eur J Pharmacol

212:247-252.

Langston JW, Ballard P, Tetrud JW and Irwin I (1983) Chronic Parkinsonism in humans due to a

product of meperidine-analog synthesis. Science 219:979-980.

42 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Langston JW, Forno LS, Rebert CS and Irwin I (1984) Selective nigral toxicity after systemic

administration of 1-methyl-4-phenyl-1,2,5,6-tetrahydropyrine (MPTP) in the squirrel

monkey. Brain Res 292:390-394.

Lees AJ, Lander CM and Stern GM (1978) Tiapride and sulpiride in Parkinson's disease. Lancet

2:1205.

Lemay S, Chouinard S, Blanchet P, Masson H, Soland V, Beuter A and Bedard MA (2004) Lack

of efficacy of a nicotine transdermal treatment on motor and cognitive deficits in Downloaded from

Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 28:31-39.

LeWitt PA, Guttman M, Tetrud JW, Tuite PJ, Mori A, Chaikin P and Sussman NM (2008) jpet.aspetjournals.org Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces "off" time in

Parkinson's disease: a double-blind, randomized, multicenter clinical trial (6002-US-005).

Ann Neurol 63:295-302. at ASPET Journals on October 1, 2021 Lewitt PA, Hauser RA, Lu M, Nicholas AP, Weiner W, Coppard N, Leinonen M and Savola JM

(2012) Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD

study). Neurology 79:163-169.

LeWitt PA, Lyons KE and Pahwa R (2007) Advanced Parkinson disease treated with rotigotine

transdermal system: PREFER Study. Neurology 68:1262-1267.

Lieberman A, Ranhosky A and Korts D (1997) Clinical evaluation of in advanced

Parkinson's disease: results of a double-blind, placebo-controlled, parallel-group study.

Neurology 49:162-168.

Lieberman AN, Gopinathan G and Neophytides A (1986) Efficacy of and mesulergine.

Eur Neurol 25:86-90.

43 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Lincoln L, Fisher R, Jackson MJ, Jenner P, Neumeyer J, Sromek AW, Lees AJ and Rose S (2016)

Oral r-(-)-11-o-valeryl-n-n-propylnoraporphine reverses motor deficits in mptp-treated

marmosets. Mov Disord 31:1381-1388.

Lindeboom SF and Lakke JP (1978) Deanol and physostigmine in the treatment of L-dopa-induced

dyskinesias. Acta Neurol Scand 58:134-138.

Loschmann PA, Chong PN, Nomoto M, Tepper PG, Horn AS, Jenner P and Marsden CD (1989)

Stereoselective reversal of MPTP-induced parkinsonism in the marmoset after dermal Downloaded from

application of N-0437. Eur J Pharmacol 166:373-380.

Loschmann PA, De Groote C, Smith L, Wullner U, Fischer G, Kemp JA, Jenner P and Klockgether jpet.aspetjournals.org T (2004) Antiparkinsonian activity of Ro 25-6981, a NR2B subunit specific NMDA

receptor antagonist, in animal models of Parkinson's disease. Exp Neurol 187:86-93.

Loschmann PA, Lange KW, Kunow M, Rettig KJ, Jahnig P, Honore T, Turski L, Wachtel H, Jenner at ASPET Journals on October 1, 2021 P and Marsden CD (1991) Synergism of the AMPA-antagonist NBQX and the NMDA-

antagonist CPP with L-dopa in models of Parkinson's disease. J Neural Transm Park Dis

Dement Sect 3:203-213.

Loschmann PA, Smith LA, Lange KW, Jahnig P, Jenner P and Marsden CD (1992) Motor activity

following the administration of selective D-1 and D-2 dopaminergic drugs to MPTP-treated

common marmosets. Psychopharmacology (Berl) 109:49-56.

Luginger E, Wenning GK, Bosch S and Poewe W (2000) Beneficial effects of amantadine on L-

dopa-induced dyskinesias in Parkinson's disease. Mov Disord 15:873-878.

Luquin MR, Laguna J, Herrero MT and Obeso JA (1993a) Behavioral tolerance to repeated

apomorphine administration in parkinsonian monkeys. J Neurol Sci 114:40-44.

Luquin MR, Laguna J and Obeso JA (1992) Selective D2 receptor stimulation induces dyskinesia

in parkinsonian monkeys. Ann Neurol 31:551-554.

44 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Luquin MR, Obeso JA, Laguna J, Guillen J and Martinez-Lage JM (1993b) The AMPA receptor

antagonist NBQX does not alter the motor response induced by selective dopamine agonists

in MPTP-treated monkeys. Eur J Pharmacol 235:297-300.

Mahmoudi S, Samadi P, Gilbert F, Ouattara B, Morissette M, Gregoire L, Rouillard C, Di Paolo T

and Levesque D (2009) Nur77 mRNA levels and L-Dopa-induced dyskinesias in MPTP

monkeys treated with docosahexaenoic acid. Neurobiol Dis 36:213-222.

Manson AJ, Iakovidou E and Lees AJ (2000) Idazoxan is ineffective for levodopa-induced Downloaded from

dyskinesias in Parkinson's disease. Mov Disord 15:336-337.

Manson AJ, Katzenschlager R, Hobart J and Lees AJ (2001) High dose naltrexone for dyskinesias jpet.aspetjournals.org induced by levodopa. J Neurol Neurosurg Psychiatry 70:554-556.

Maratos EC, Jackson MJ, Pearce RK, Cannizzaro C and Jenner P (2003) Both short- and long-

acting D-1/D-2 dopamine agonists induce less dyskinesia than L-DOPA in the MPTP- at ASPET Journals on October 1, 2021 lesioned common marmoset (Callithrix jacchus). Exp Neurol 179:90-102.

Maratos EC, Jackson MJ, Pearce RK and Jenner P (2001) Antiparkinsonian activity and dyskinesia

risk of ropinirole and L-DOPA combination therapy in drug naive MPTP-lesioned common

marmosets (Callithrix jacchus). Mov Disord 16:631-641.

Marino S, Sessa E, Di Lorenzo G, Digangi G, Alagna A, Bramanti P and Di Bella P (2008)

Sertraline in the treatment of depressive disorders in patients with Parkinson's disease.

Neurol Sci 29:391-395.

Marsden CD, Parkes JD and Rees JE (1973) A year's comparison of treatment of patients with

parkinson's disease with levodopa combined with carbidopa versus treatment with levodopa

alone. Lancet 2:1459-1462.

45 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Marti M, Rodi D, Li Q, Guerrini R, Fasano S, Morella I, Tozzi A, Brambilla R, Calabresi P,

Simonato M, Bezard E and Morari M (2012) Nociceptin/orphanin FQ receptor agonists

attenuate L-DOPA-induced dyskinesias. J Neurosci 32:16106-16119.

Martignoni E, Pacchetti C, Aufdembrinke B, Godi L, Albani G, Mancini F and Nappi G (1995)

Terguride in stable Parkinson's disease. Funct Neurol 10:143-146.

Martin WE, Loewenson RB, Resch JA and Baker AB (1974) A controlled study comparing

trihexyphenidyl hydrochloride plus levodopa with placebo plus levodopa in patients with Downloaded from

Parkinson's disease. Neurology 24:912-919.

McCall RB, Lookingland KJ, Bedard PJ and Huff RM (2005) Sumanirole, a highly dopamine D2- jpet.aspetjournals.org selective receptor agonist: in vitro and in vivo pharmacological characterization and

efficacy in animal models of Parkinson's disease. J Pharmacol Exp Ther 314:1248-1256.

Meco G, Fabrizio E, Di Rezze S, Alessandri A and Pratesi L (2003) Mirtazapine in L-dopa-induced at ASPET Journals on October 1, 2021 dyskinesias. Clin Neuropharmacol 26:179-181.

Meltzer HY, Mills R, Revell S, Williams H, Johnson A, Bahr D and Friedman JH (2010)

Pimavanserin, a Serotonin(2A) Receptor Inverse Agonist, for the Treatment of Parkinson's

Disease Psychosis. Neuropsychopharmacology 35:881-892.

Merello M, Lees AJ, Webster R, Bovingdon M and Gordin A (1994) Effect of entacapone, a

peripherally acting catechol-O-methyltransferase inhibitor, on the motor response to acute

treatment with levodopa in patients with Parkinson's disease. J Neurol Neurosurg

Psychiatry 57:186-189.

Mestre TA, Shah BB, Connolly BS, de Aquino C, Al Dhakeel A, Walsh R, Ghate T, Lui JP and

Fox SH (2014) , a histamine H2 receptor antagonist, does not reduce levodopa-

induced dyskinesia in Parkinson’s disease: a proof-of-concept study. Mov Disord Clin

Pract 1:219-224.

46 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Millan MJ, Di Cara B, Hill M, Jackson M, Joyce JN, Brotchie J, McGuire S, Crossman A, Smith

L, Jenner P, Gobert A, Peglion JL and Brocco M (2004) S32504, a novel naphtoxazine

agonist at dopamine D3/D2 receptors: II. Actions in rodent, primate, and cellular models of

antiparkinsonian activity in comparison to ropinirole. J Pharmacol Exp Ther 309:921-935.

Mizuno Y, Kondo T and Narabayashi H (1995) Pergolide in the treatment of Parkinson's disease.

Neurology 45:S13-21.

Mizuno Y, Nomoto M, Kondo T, Hasegawa K, Murata M, Takeuchi M, Ikeda J, Tomida T, Hattori Downloaded from

N and Rotigotine Trial G (2013) Transdermal rotigotine in early stage Parkinson's disease:

a randomized, double-blind, placebo-controlled trial. Mov Disord 28:1447-1450. jpet.aspetjournals.org Molinari SP, Kaminski R, Di Rocco A and Yahr MD (1995) The use of famotidine in the treatment

of Parkinson's disease: a pilot study. J Neural Transm Park Dis Dement Sect 9:243-247.

Montastruc JL, Puech AJ, Clanet M, Guiraud-Chaumeil B and Rascol A (1981) [Yohimbine in at ASPET Journals on October 1, 2021 treatment of Parkinson's disease. Preliminary results (author's transl)]. Nouv Presse Med

10:1331-1332.

Montastruc JL, Rascol O, Senard JM and Rascol A (1994) A randomised controlled study

comparing bromocriptine to which levodopa was later added, with levodopa alone in

previously untreated patients with Parkinson's disease: a five year follow up. J Neurol

Neurosurg Psychiatry 57:1034-1038.

Morin N, Gregoire L, Gomez-Mancilla B, Gasparini F and Di Paolo T (2010) Effect of the

metabotropic glutamate receptor type 5 antagonists MPEP and MTEP in parkinsonian

monkeys. Neuropharmacology 58:981-986.

Morin N, Gregoire L, Morissette M, Desrayaud S, Gomez-Mancilla B, Gasparini F and Di Paolo

T (2013a) MPEP, an mGlu5 receptor antagonist, reduces the development of L-DOPA-

47 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

induced motor complications in de novo parkinsonian monkeys: biochemical correlates.

Neuropharmacology 66:355-364.

Morin N, Morissette M, Gregoire L and Di Paolo T (2015a) Effect of a chronic treatment with an

mGlu5 receptor antagonist on brain serotonin markers in parkinsonian monkeys. Prog

Neuropsychopharmacol Biol Psychiatry 56:27-38.

Morin N, Morissette M, Gregoire L, Gomez-Mancilla B, Gasparini F and Di Paolo T (2013b)

Chronic treatment with MPEP, an mGlu5 receptor antagonist, normalizes basal ganglia Downloaded from

glutamate neurotransmission in L-DOPA-treated parkinsonian monkeys.

Neuropharmacology 73:216-231. jpet.aspetjournals.org Morin N, Morissette M, Gregoire L, Rajput A, Rajput AH and Di Paolo T (2015b) Contribution of

brain serotonin subtype 1B receptors in levodopa-induced motor complications.

Neuropharmacology 99:356-368. at ASPET Journals on October 1, 2021 Morissette M, Dridi M, Calon F, Hadj Tahar A, Meltzer LT, Bedard PJ and Di Paolo T (2006a)

Prevention of dyskinesia by an NMDA receptor antagonist in MPTP monkeys: effect on

adenosine A2A receptors. Synapse 60:239-250.

Morissette M, Dridi M, Calon F, Hadj Tahar A, Meltzer LT, Bedard PJ and Di Paolo T (2006b)

Prevention of levodopa-induced dyskinesias by a selective NR1A/2B N-methyl-D-

aspartate receptor antagonist in parkinsonian monkeys: implication of preproenkephalin.

Mov Disord 21:9-17.

Morissette M, Goulet M, Grondin R, Blanchet P, Bedard PJ, Di Paolo T and Levesque D (1998)

Associative and limbic regions of monkey striatum express high levels of dopamine D3

receptors: effects of MPTP and dopamine agonist replacement therapies. Eur J Neurosci

10:2565-2573.

48 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Morissette M, Goulet M, Soghomonian JJ, Blanchet PJ, Calon F, Bedard PJ and Di Paolo T (1997)

Preproenkephalin mRNA expression in the caudate-putamen of MPTP monkeys after

chronic treatment with the D2 agonist U91356A in continuous or intermittent mode of

administration: comparison with L-DOPA therapy. Brain Res Mol Brain Res 49:55-62.

Morissette M, Grondin R, Goulet M, Bedard PJ and Di Paolo T (1999) Differential regulation of

striatal preproenkephalin and preprotachykinin mRNA levels in MPTP-lesioned monkeys

chronically treated with dopamine D1 or D2 receptor agonists. J Neurochem 72:682-692. Downloaded from

Morissette M, Morin N, Gregoire L, Rajput A, Rajput AH and Di Paolo T (2016) Brain alpha7

nicotinic acetylcholine receptors in MPTP-lesioned monkeys and parkinsonian patients. jpet.aspetjournals.org Biochem Pharmacol 109:62-69.

Morissette M, Samadi P, Hadj Tahar A, Belanger N and Di Paolo T (2010) Striatal Akt/GSK3

signaling pathway in the development of L-Dopa-induced dyskinesias in MPTP monkeys. at ASPET Journals on October 1, 2021 Prog Neuropsychopharmacol Biol Psychiatry 34:446-454.

Muenter MD, Ahlskog JE, Bell G and McManis P (1988) PHNO [(+)-4-propyl-9-

hydroxynaphthoxazine]: a new and effective anti-Parkinson's disease agent. Neurology

38:1541-1545.

Munoz A, Li Q, Gardoni F, Marcello E, Qin C, Carlsson T, Kirik D, Di Luca M, Bjorklund A,

Bezard E and Carta M (2008) Combined 5-HT1A and 5-HT1B receptor agonists for the

treatment of L-DOPA-induced dyskinesia. Brain 131:3380-3394.

Nagata T, Shinagawa S, Tagai K and Nakayama K (2012) A case in which mirtazapine reduced

auditory hallucinations in a patient with Parkinson disease. Int Psychogeriatr:1-3.

Nash JE, Fox SH, Henry B, Hill MP, Peggs D, McGuire S, Maneuf Y, Hille C, Brotchie JM and

Crossman AR (2000) Antiparkinsonian actions of in the MPTP-lesioned

marmoset model of Parkinson's disease. Exp Neurol 165:136-142.

49 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Nash JE, Ravenscroft P, McGuire S, Crossman AR, Menniti FS and Brotchie JM (2004) The

NR2B-selective NMDA receptor antagonist CP-101,606 exacerbates L-DOPA-induced

dyskinesia and provides mild potentiation of anti-parkinsonian effects of L-DOPA in the

MPTP-lesioned marmoset model of Parkinson's disease. Exp Neurol 188:471-479.

Nomoto M, Jenner P and Marsden CD (1985) The dopamine D2 agonist LY 141865, but not the

D1 agonist SKF 38393, reverses parkinsonism induced by 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine (MPTP) in the common marmoset. Neurosci Lett 57:37-41. Downloaded from

Nomoto M, Jenner P and Marsden CD (1988) The D1 agonist SKF 38393 inhibits the

antiparkinsonian activity of the D2 agonist LY 171555 in the MPTP-treated marmoset. jpet.aspetjournals.org Neurosci Lett 93:275-280.

Nomoto M, Stahl S, Jenner P and Marsden CD (1987) Antiparkinsonian activity of (+)-PHNO in

the MPTP-treated common marmoset. Mov Disord 2:37-45. at ASPET Journals on October 1, 2021 Nutt JG, Gunzler SA, Kirchhoff T, Hogarth P, Weaver JL, Krams M, Jamerson B, Menniti FS and

Landen JW (2008) Effects of a NR2B selective NMDA glutamate antagonist, CP-101,606,

on dyskinesia and Parkinsonism. Mov Disord 23:1860-1866.

Oertel W, Eggert K, Pahwa R, Tanner CM, Hauser RA, Trenkwalder C, Ehret R, Azulay JP,

Isaacson S, Felt L and Stempien MJ (2017) Randomized, placebo-controlled trial of ADS-

5102 (amantadine) extended-release capsules for levodopa-induced dyskinesia in

Parkinson's disease (EASE LID 3). Mov Disord.

Oertel WH, Wolters E, Sampaio C, Gimenez-Roldan S, Bergamasco B, Dujardin M, Grosset DG,

Arnold G, Leenders KL, Hundemer HP, Lledo A, Wood A, Frewer P and Schwarz J (2006)

Pergolide versus levodopa monotherapy in early Parkinson's disease patients: The

PELMOPET study. Mov Disord 21:343-353.

50 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Oh JD, Bibbiani F and Chase TN (2002) Quetiapine attenuates levodopa-induced motor

complications in rodent and primate parkinsonian models. Exp Neurol 177:557-564.

Olanow CW, Damier P, Goetz CG, Mueller T, Nutt J, Rascol O, Serbanescu A, Deckers F and

Russ H (2004) Multicenter, open-label, trial of sarizotan in Parkinson disease patients with

levodopa-induced dyskinesias (the SPLENDID Study). Clin Neuropharmacol 27:58-62.

Olanow CW, Fahn S, Muenter M, Klawans H, Hurtig H, Stern M, Shoulson I, Kurlan R, Grimes

JD, Jankovic J and et al. (1994) A multicenter double-blind placebo-controlled trial of Downloaded from

pergolide as an adjunct to Sinemet in Parkinson's disease. Mov Disord 9:40-47.

Ondo WG, Tintner R, Voung KD, Lai D and Ringholz G (2005) Double-blind, placebo-controlled, jpet.aspetjournals.org unforced titration parallel trial of quetiapine for dopaminergic-induced hallucinations in

Parkinson's disease. Mov Disord 20:958-963.

Ouattara B, Belkhir S, Morissette M, Dridi M, Samadi P, Gregoire L, Meltzer LT and Di Paolo T at ASPET Journals on October 1, 2021 (2009) Implication of NMDA receptors in the antidyskinetic activity of cabergoline, CI-

1041, and Ro 61-8048 in MPTP monkeys with levodopa-induced dyskinesias. J Mol

Neurosci 38:128-142.

Ouattara B, Gregoire L, Morissette M, Gasparini F, Vranesic I, Bilbe G, Johns DR, Rajput A,

Hornykiewicz O, Rajput AH, Gomez-Mancilla B and Di Paolo T (2011) Metabotropic

glutamate receptor type 5 in levodopa-induced motor complications. Neurobiol Aging

32:1286-1295.

Ouattara B, Hoyer D, Gregoire L, Morissette M, Gasparini F, Gomez-Mancilla B and Di Paolo T

(2010) Changes of AMPA receptors in MPTP monkeys with levodopa-induced dyskinesias.

Neuroscience 167:1160-1167.

Pahwa R, Tanner CM, Hauser RA, Isaacson SH, Nausieda PA, Truong DD, Agarwal P, Hull KL,

Lyons KE, Johnson R and Stempien MJ (2017) ADS-5102 (Amantadine) Extended-Release

51 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Capsules for Levodopa-Induced Dyskinesia in Parkinson Disease (EASE LID Study): A

Randomized Clinical Trial. JAMA Neurol 74:941-949.

Palhagen SE, Carlsson M, Curman E, Walinder J and Granerus AK (2008) Depressive illness in

Parkinson's disease--indication of a more advanced and widespread neurodegenerative

process? Acta Neurol Scand 117:295-304.

Palhagen SE, Ekberg S, Walinder J, Granerus AK and Granerus G (2009) HMPAO SPECT in

Parkinson's disease (PD) with major depression (MD) before and after Downloaded from

treatment. J Neurol 256:1510-1518.

Papa SM, Auberson YP and Greenamyre JT (2004) Prolongation of levodopa responses by jpet.aspetjournals.org glycineB antagonists in parkinsonian primates. Ann Neurol 56:723-727.

Papa SM and Chase TN (1996) Levodopa-induced dyskinesias improved by a glutamate antagonist

in Parkinsonian monkeys. Ann Neurol 39:574-578. at ASPET Journals on October 1, 2021 Park DM, Findley LJ, Hanks G and Sandler M (1981) Nomifensine: effect in Parkinsonian patients

not receiving levodopa. J Neurol Neurosurg Psychiatry 44:352-354.

Park DM, Findley LJ and Teychenne PF (1977) Nomifensine in parkinsonism. Br J Clin Pharmacol

4Suppl 2:185S-186S.

Parkes JD, Baxter RC, Curzon G, Knill-Jones RP, Knott PJ, Marsden CD, Tattersall R and Vollum

D (1971) Treatment of Parkinson's disease with amantadine and levodopa. A one-year

study. Lancet 1:1083-1086.

ParkinsonStudyGroup (1999) Low-dose clozapine for the treatment of drug-induced psychosis in

Parkinson's disease. The Parkinson Study Group. N Engl J Med 340:757-763.

ParkinsonStudyGroup (2000a) A multicenter randomized controlled trial of remacemide

hydrochloride as monotherapy for PD. Parkinson Study Group. Neurology 54:1583-1588.

52 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

ParkinsonStudyGroup (2000b) Pramipexole vs levodopa as initial treatment for Parkinson disease:

A randomized controlled trial. Parkinson Study Group. JAMA 284:1931-1938.

ParkinsonStudyGroup (2001) Evaluation of dyskinesias in a pilot, randomized, placebo-controlled

trial of remacemide in advanced Parkinson disease. Arch Neurol 58:1660-1668.

ParkinsonStudyGroup (2003) A controlled trial of rotigotine monotherapy in early Parkinson's

disease. Arch Neurol 60:1721-1728.

ParkinsonStudyGroup (2005) A randomized placebo-controlled trial of rasagiline in levodopa- Downloaded from

treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch

Neurol 62:241-248. jpet.aspetjournals.org Pearce RK, Banerji T, Jenner P and Marsden CD (1998) De novo administration of ropinirole and

bromocriptine induces less dyskinesia than L-dopa in the MPTP-treated marmoset. Mov

Disord 13:234-241. at ASPET Journals on October 1, 2021 Pearce RK, Jackson M, Britton DR, Shiosaki K, Jenner P and Marsden CD (1999) Actions of the

D1 agonists A-77636 and A-86929 on locomotion and dyskinesia in MPTP-treated L-dopa-

primed common marmosets. Psychopharmacology (Berl) 142:51-60.

Pearce RK, Jackson M, Smith L, Jenner P and Marsden CD (1995) Chronic L-DOPA

administration induces dyskinesias in the 1-methyl-4- phenyl-1,2,3,6-tetrahydropyridine-

treated common marmoset (Callithrix Jacchus). Mov Disord 10:731-740.

Pearce RK, Smith LA, Jackson MJ, Banerji T, Scheel-Kruger J and Jenner P (2002) The

monoamine reuptake blocker brasofensine reverses akinesia without dyskinesia in MPTP-

treated and levodopa-primed common marmosets. Mov Disord 17:877-886.

Pessiglione M, Guehl D, Hirsch EC, Feger J and Tremblay L (2004a) Disruption of self-organized

actions in monkeys with progressive MPTP-induced parkinsonism. I. Effects of task

complexity. Eur J Neurosci 19:426-436.

53 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Pessiglione M, Guehl D, Jan C, Francois C, Hirsch EC, Feger J and Tremblay L (2004b) Disruption

of self-organized actions in monkeys with progressive MPTP-induced parkinsonism: II.

Effects of reward preference. Eur J Neurosci 19:437-446.

Philippens IH, Joosen MJ, Ahnaou A, Andres I and Drinkenburg WP (2014) Anti-Parkinson effects

of a selective alpha2C-adrenoceptor antagonist in the MPTP marmoset model. Behav Brain

Res 269:81-86.

Pinna A, Ko WK, Costa G, Tronci E, Fidalgo C, Simola N, Li Q, Tabrizi MA, Bezard E, Carta M Downloaded from

and Morelli M (2016) Antidyskinetic effect of A2A and 5HT1A/1B receptor ligands in two

animal models of Parkinson's disease. Mov Disord 31:501-511. jpet.aspetjournals.org Poewe WH, Rascol O, Quinn N, Tolosa E, Oertel WH, Martignoni E, Rupp M and Boroojerdi B

(2007) Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson's disease:

a double-blind, double-dummy, randomised controlled trial. Lancet Neurol 6:513-520. at ASPET Journals on October 1, 2021 Porras G, Li Q and Bezard E (2012) Modeling Parkinson's disease in primates: The MPTP model.

Cold Spring Harb Perspect Med 2:a009308.

Potts LF, Park ES, Woo JM, Dyavar Shetty BL, Singh A, Braithwaite SP, Voronkov M, Papa SM

and Mouradian MM (2015) Dual kappa-agonist/mu-antagonist opioid receptor modulation

reduces levodopa-induced dyskinesia and corrects dysregulated striatal changes in the

nonhuman primate model of Parkinson disease. Ann Neurol 77:930-941.

Pourcher E, Bonnet AM, Kefalos J, Dubois B and Agid Y (1989) Effects of etybenzatropine and

diazepam on levodopa-induced diphasic dyskinesias in Parkinson's disease. Mov Disord

4:195-201.

Pourcher E, Fernandez HH, Stacy M, Mori A, Ballerini R and Chaikin P (2012) Istradefylline for

Parkinson's disease patients experiencing motor fluctuations: results of the KW-6002-US-

018 study. Parkinsonism Relat Disord 18:178-184.

54 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Pourcher E, Gomez-Mancilla B and Bedard PJ (1992) Ethosuximide and tremor in Parkinson's

disease: a pilot study. Mov Disord 7:132-136.

Quik M, Cox H, Parameswaran N, O'Leary K, Langston JW and Di Monte D (2007) Nicotine

reduces levodopa-induced dyskinesias in lesioned monkeys. Ann Neurol 62:588-596.

Quik M, Mallela A, Chin M, McIntosh JM, Perez XA and Bordia T (2013a) Nicotine-mediated

improvement in L-dopa-induced dyskinesias in MPTP-lesioned monkeys is dependent on

dopamine nerve terminal function. Neurobiol Dis 50:30-41. Downloaded from

Quik M, Mallela A, Ly J and Zhang D (2013b) Nicotine reduces established levodopa-induced

dyskinesias in a monkey model of Parkinson's disease. Mov Disord 28:1398-1406. jpet.aspetjournals.org Quinn N, Illas A, Lhermitte F and Agid Y (1981) Bromocriptine and domperidone in the treatment

of Parkinson disease. Neurology 31:662-667.

Rampello L, Chiechio S, Raffaele R, Vecchio I and Nicoletti F (2002) The SSRI, , at ASPET Journals on October 1, 2021 improves bradykinesia in patients with Parkinson's disease treated with L-dopa. Clin

Neuropharmacol 25:21-24.

Rascol O, Arnulf I, Peyro-Saint Paul H, Brefel-Courbon C, Vidailhet M, Thalamas C, Bonnet AM,

Descombes S, Bejjani B, Fabre N, Montastruc JL and Agid Y (2001a) Idazoxan, an alpha-

2 antagonist, and L-DOPA-induced dyskinesias in patients with Parkinson's disease. Mov

Disord 16:708-713.

Rascol O, Blin O, Thalamas C, Descombes S, Soubrouillard C, Azulay P, Fabre N, Viallet F,

Lafnitzegger K, Wright S, Carter JH and Nutt JG (1999) ABT-431, a D1 receptor agonist

, has efficacy in Parkinson's disease. Ann Neurol 45:736-741.

Rascol O, Bronzova J, Hauser RA, Lang AE, Sampaio C, Theeuwes A and van de Witte SV (2012)

Pardoprunox as adjunct therapy to levodopa in patients with Parkinson's disease

55 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

experiencing motor fluctuations: results of a double-blind, randomized, placebo-controlled,

trial. Parkinsonism Relat Disord 18:370-376.

Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE and Lang AE (2000) A five-year study

of the incidence of dyskinesia in patients with early Parkinson's disease who were treated

with ropinirole or levodopa. 056 Study Group. N Engl J Med 342:1484-1491.

Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE and Abdalla M (2006a)

Development of dyskinesias in a 5-year trial of ropinirole and L-dopa. Mov Disord 21:1844- Downloaded from

1850.

Rascol O, Brooks DJ, Melamed E, Oertel W, Poewe W, Stocchi F and Tolosa E (2005) Rasagiline jpet.aspetjournals.org as an adjunct to levodopa in patients with Parkinson's disease and motor fluctuations

(LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a

randomised, double-blind, parallel-group trial. Lancet 365:947-954. at ASPET Journals on October 1, 2021 Rascol O, Dubois B, Caldas AC, Senn S, Del Signore S, Lees A and Parkinson RSG (2006b) Early

piribedil monotherapy of Parkinson's disease: A planned seven-month report of the

REGAIN study. Mov Disord 21:2110-2115.

Rascol O, Fabre N, Blin O, Poulik J, Sabatini U, Senard JM, Ane M, Montastruc JL and Rascol A

(1994) Naltrexone, an opiate antagonist, fails to modify motor symptoms in patients with

Parkinson's disease. Mov Disord 9:437-440.

Rascol O, Lees AJ, Senard JM, Pirtosek Z, Montastruc JL and Fuell D (1996) Ropinirole in the

treatment of levodopa-induced motor fluctuations in patients with Parkinson's disease. Clin

Neuropharmacol 19:234-245.

Rascol O, Nutt JG, Blin O, Goetz CG, Trugman JM, Soubrouillard C, Carter JH, Currie LJ, Fabre

N, Thalamas C, Giardina WW and Wright S (2001b) Induction by dopamine D1 receptor

56 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

agonist ABT-431 of dyskinesia similar to levodopa in patients with Parkinson disease. Arch

Neurol 58:249-254.

Riahi G, Morissette M, Levesque D, Rouillard C, Samadi P, Parent M and Di Paolo T (2012) Effect

of chronic l-DOPA treatment on 5-HT(1A) receptors in parkinsonian monkey brain.

Neurochem Int 61:1160-1171.

Riahi G, Morissette M, Parent M and Di Paolo T (2011) Brain 5-HT(2A) receptors in MPTP

monkeys and levodopa-induced dyskinesias. Eur J Neurosci 33:1823-1831. Downloaded from

Riahi G, Morissette M, Samadi P, Parent M and Di Paolo T (2013) Basal ganglia serotonin 1B

receptors in parkinsonian monkeys with L-DOPA-induced dyskinesia. Biochem Pharmacol jpet.aspetjournals.org 86:970-978.

Rinne UK, Birket-Smith E, Dupont E, Hansen E, Hyyppa M, Marttila R, Mikkelsen B, Pakkenberg

H and Presthus J (1975) Levodopa alone and in combination with a peripheral at ASPET Journals on October 1, 2021 decarboxylase inhibitor benserazide (Madopar) in the treatment of Parkinson's disease: A

controlled clinical trial. J Neurol 211:1-9.

Rinne UK, Bracco F, Chouza C, Dupont E, Gershanik O, Marti Masso JF, Montastruc JL, Marsden

CD, Dubini A, Orlando N and Grimaldi R (1997) Cabergoline in the treatment of early

Parkinson's disease: results of the first year of treatment in a double-blind comparison of

cabergoline and levodopa. The PKDS009 Collaborative Study Group. Neurology 48:363-

368.

Rose S, Jackson MJ, Smith LA, Stockwell K, Johnson L, Carminati P and Jenner P (2006) The

novel adenosine A2a receptor antagonist ST1535 potentiates the effects of a threshold dose

of L-DOPA in MPTP treated common marmosets. Eur J Pharmacol 546:82-87.

57 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Rose S, Scheller DK, Breidenbach A, Smith L, Jackson M, Stockwell K and Jenner P (2007)

Plasma levels of rotigotine and the reversal of motor deficits in MPTP-treated primates.

Behav Pharmacol 18:155-160.

Rouillard C, Bedard PJ and Di Paolo T (1990) Effects of chronic treatment of MPTP monkeys with

bromocriptine alone or in combination with SKF 38393. Eur J Pharmacol 185:209-215.

Rylander D, Iderberg H, Li Q, Dekundy A, Zhang J, Li H, Baishen R, Danysz W, Bezard E and

Cenci MA (2010) A mGluR5 antagonist under clinical development improves L-DOPA- Downloaded from

induced dyskinesia in parkinsonian rats and monkeys. Neurobiol Dis 39:352-361.

Samadi P, Gregoire L and Bedard PJ (2003) Opioid antagonists increase the dyskinetic response jpet.aspetjournals.org to dopaminergic agents in parkinsonian monkeys: interaction between dopamine and opioid

systems. Neuropharmacology 45:954-963.

Samadi P, Gregoire L and Bedard PJ (2004) The opioid agonist morphine decreases the dyskinetic at ASPET Journals on October 1, 2021 response to dopaminergic agents in parkinsonian monkeys. Neurobiol Dis 16:246-253.

Samadi P, Gregoire L, Hadj Tahar A, Di Paolo T, Rouillard C and Bedard PJ (2005a) Naltrexone

in the short-term decreases antiparkinsonian response to l-Dopa and in the long-term

increases dyskinesias in drug-naive parkinsonian monkeys. Neuropharmacology 49:165-

173.

Samadi P, Gregoire L, Morissette M, Calon F, Hadj Tahar A, Belanger N, Dridi M, Bedard PJ and

Di Paolo T (2008a) Basal ganglia group II metabotropic glutamate receptors specific

binding in non-human primate model of L-Dopa-induced dyskinesias. Neuropharmacology

54:258-268.

Samadi P, Gregoire L, Morissette M, Calon F, Hadj Tahar A, Dridi M, Belanger N, Meltzer LT,

Bedard PJ and Di Paolo T (2008b) mGluR5 metabotropic glutamate receptors and

dyskinesias in MPTP monkeys. Neurobiol Aging 29:1040-1051.

58 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Samadi P, Gregoire L, Rassoulpour A, Guidetti P, Izzo E, Schwarcz R and Bedard PJ (2005b)

Effect of kynurenine 3-hydroxylase inhibition on the dyskinetic and antiparkinsonian

responses to levodopa in Parkinsonian monkeys. Mov Disord 20:792-802.

Samadi P, Gregoire L, Rouillard C and Bedard PJ (2005c) Dyskinesias occur in response to saline

and naltrexone alone after priming with combination of dopaminergic agents and

naltrexone in the MPTP parkinsonian monkeys. Neurobiol Dis 19:266-272.

Samadi P, Gregoire L, Rouillard C, Bedard PJ, Di Paolo T and Levesque D (2006) Downloaded from

Docosahexaenoic acid reduces levodopa-induced dyskinesias in 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine monkeys. Ann Neurol 59:282-288. jpet.aspetjournals.org Samadi P, Morissette M, Calon F, Hadj Tahar A, Dridi M, Belanger N, Meltzer LT, Bedard PJ and

Di Paolo T (2008c) Normalization of GABAA receptor specific binding in the substantia

nigra reticulata and the prevention of L-dopa-induced dyskinesias in MPTP parkinsonian at ASPET Journals on October 1, 2021 monkeys. Synapse 62:101-109.

Samadi P, Morissette M, Levesque D and Di Paolo T (2010) BDNF levels are not related with

levodopa-induced dyskinesias in MPTP monkeys. Mov Disord 25:116-121.

Sampaio C, Bronzova J, Hauser RA, Lang AE, Rascol O, van de Witte SV, Theeuwes AA and

Rembrandt/Vermeer Study G (2011) Pardoprunox in early Parkinson's disease: results from

2 large, randomized double-blind trials. Mov Disord 26:1464-1476.

Savola JM, Hill M, Engstrom M, Merivuori H, Wurster S, McGuire SG, Fox SH, Crossman AR

and Brotchie JM (2003) Fipamezole (JP-1730) is a potent alpha2 adrenergic receptor

antagonist that reduces levodopa-induced dyskinesia in the MPTP-lesioned primate model

of Parkinson's disease. Mov Disord 18:872-883.

Schapira AH, Fox SH, Hauser RA, Jankovic J, Jost WH, Kenney C, Kulisevsky J, Pahwa R, Poewe

W and Anand R (2017) Assessment of Safety and Efficacy of Safinamide as a Levodopa

59 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Adjunct in Patients With Parkinson Disease and Motor Fluctuations: A Randomized

Clinical Trial. JAMA Neurol 74:216-224.

Schwab RS, Amador LV and Lettvin JY (1951) Apomorphine in Parkinson's disease. Trans Am

Neurol Assoc 56:251-253.

Serrano-Duenas M (2000) [Acute pharmacological test with clonidine for the tremor in patients

with Parkinson disease]. Rev Neurol 30:910-913.

Shen W, Plotkin JL, Francardo V, Ko WK, Xie Z, Li Q, Fieblinger T, Wess J, Neubig RR, Lindsley Downloaded from

CW, Conn PJ, Greengard P, Bezard E, Cenci MA and Surmeier DJ (2015) M4 Muscarinic

Receptor Signaling Ameliorates Striatal Plasticity Deficits in Models of L-DOPA-Induced jpet.aspetjournals.org Dyskinesia. Neuron 88:762-773.

Shiosaki K, Jenner P, Asin KE, Britton DR, Lin CW, Michaelides M, Smith L, Bianchi B,

Didomenico S, Hodges L, Hong Y, Mahan L, Mikusa J, Miller T, Nikkel A, Stashko M, at ASPET Journals on October 1, 2021 Witte D and Williams M (1996) ABT-431: the diacetyl prodrug of A-86929, a potent and

selective dopamine D1 receptor agonist: in vitro characterization and effects in animal

models of Parkinson's disease. J Pharmacol Exp Ther 276:150-160.

Shoulson I and Chase TN (1976) Clonidine and the anti-parkinsonian response to L-DOPA or

piribedil. Neuropharmacology 15:25-27.

Shoulson I, Penney J, McDermott M, Schwid S, Kayson E, Chase T, Fahn S, Greenamyre JT, Lang

A, Siderowf A, Pearson N, Harrison M, Rost E, Colcher A, Lloyd M, Matthews M, Pahwa

R, McGuire D, Lew MF, Schuman S, Marek K, Broshjeit S, Factor S, Brown D, Feigin A,

Mazurkiewicz J, Ford B, Jennings D, Dilllon S, Comella C, Blasucci L, Janko K, Shulman

L, Wiener W, Bateman-Rodriguez D, Carrion A, Suchowersky O, Lafontaine AL, Pantella

C, Siemers E, Belden J, Davies R, Lannon M, Grimes D, Gray P, Martin W, Kennedy L,

Adler C, Newman S, Hammerstad J, Stone C, Lewitt P, Bardram K, Mistura K, Miyasaki

60 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

J, Johnston L, Cha JH, Tennis M, Panniset M, Hall J, Tetrud J, Friedlander J, Hauser R,

Gauger L, Rodnitzky R, Deleo A, Dobson J, Seeberger L, Dingmann C, Tarsy D, Ryan P,

Elmer L, Ruzicka D, Stacy M, Brewer M, Locke B, Baker D, Casaceli C, Day D, Florack

M, Hodgeman K, Laroia N, Nobel R, Orme C, Rexo L, Rothenburgh K, Sulimowicz K,

Watts A, Wratni E, Tariot P, Cox C, Leventhal C, Alderfer V, Craun AM, Frey J, McCree

L, McDermott J, Cooper J, Holdich T and Read B (2001) A randomized, controlled trial of

remacemide for motor fluctuations in Parkinson's disease. Neurology 56:455-462. Downloaded from

Sieradzan KA, Fox SH, Hill M, Dick JP, Crossman AR and Brotchie JM (2001) Cannabinoids

reduce levodopa-induced dyskinesia in Parkinson's disease: a pilot study. Neurology jpet.aspetjournals.org 57:2108-2111.

Silverdale MA, Crossman AR and Brotchie JM (2002) Striatal AMPA receptor binding is unaltered

in the MPTP-lesioned macaque model of Parkinson's disease and dyskinesia. Exp Neurol at ASPET Journals on October 1, 2021 174:21-28.

Silverdale MA, Nicholson SL, Crossman AR and Brotchie JM (2005) Topiramate reduces

levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson's

disease. Mov Disord 20:403-409.

Silverdale MA, Nicholson SL, Ravenscroft P, Crossman AR, Millan MJ and Brotchie JM (2004)

Selective blockade of D(3) dopamine receptors enhances the anti-parkinsonian properties

of ropinirole and levodopa in the MPTP-lesioned primate. Exp Neurol 188:128-138.

Singer C, Lamb J, Ellis A, Layton G and Sumanirole for Early Parkinson's Disease Study G (2007)

A comparison of sumanirole versus placebo or ropinirole for the treatment of patients with

early Parkinson's disease. Mov Disord 22:476-482.

61 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Smith CP, Oh JD, Bibbiani F, Collins MA, Avila I and Chase TN (2007) Tamoxifen effect on L-

DOPA induced response complications in parkinsonian rats and primates.

Neuropharmacology 52:515-526.

Smith L, De Salvia M, Jenner P and Marsden CD (1996) An appraisal of the antiparkinsonian

activity of piribedil in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated common

marmosets. Mov Disord 11:125-135.

Smith LA, Gordin A, Jenner P and Marsden CD (1997) Entacapone enhances levodopa-induced Downloaded from

reversal of motor disability in MPTP-treated common marmosets. Mov Disord 12:935-945.

Smith LA, Jackson MG, Bonhomme C, Chezaubernard C, Pearce RK and Jenner P (2000) jpet.aspetjournals.org Transdermal administration of piribedil reverses MPTP-induced motor deficits in the

common marmoset. Clin Neuropharmacol 23:133-142.

Smith LA, Jackson MJ, Al-Barghouthy G and Jenner P (2002a) The actions of a D-1 agonist in at ASPET Journals on October 1, 2021 MPTP treated primates show dependence on both D-1 and D-2 receptor function and

tolerance on repeated administration. J Neural Transm (Vienna) 109:123-140.

Smith LA, Jackson MJ, Al-Barghouthy G, Rose S, Kuoppamaki M, Olanow W and Jenner P (2005)

Multiple small doses of levodopa plus entacapone produce continuous dopaminergic

stimulation and reduce dyskinesia induction in MPTP-treated drug-naive primates. Mov

Disord 20:306-314.

Smith LA, Jackson MJ, Hansard MJ, Maratos E and Jenner P (2003) Effect of pulsatile

administration of levodopa on dyskinesia induction in drug-naive MPTP-treated common

marmosets: effect of dose, frequency of administration, and brain exposure. Mov Disord

18:487-495.

Smith LA, Jackson MJ, Johnston L, Kuoppamaki M, Rose S, Al-Barghouthy G, Del Signore S and

Jenner P (2006) Switching from levodopa to the long-acting dopamine D2/D3 agonist

62 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

piribedil reduces the expression of dyskinesia while maintaining effective motor activity in

MPTP-treated primates. Clin Neuropharmacol 29:112-125.

Smith LA, Tel BC, Jackson MJ, Hansard MJ, Braceras R, Bonhomme C, Chezaubernard C, Del

Signore S, Rose S and Jenner P (2002b) Repeated administration of piribedil induces less

dyskinesia than L-dopa in MPTP-treated common marmosets: a behavioural and

biochemical investigation. Mov Disord 17:887-901.

Soghomonian JJ, Pedneault S, Audet G and Parent A (1994) Increased glutamate decarboxylase Downloaded from

mRNA levels in the striatum and pallidum of MPTP-treated primates. J Neurosci 14:6256-

6265. jpet.aspetjournals.org Stathis P, Konitsiotis S, Tagaris G and Peterson D (2011) Levetiracetam for the management of

levodopa-induced dyskinesias in Parkinson's disease. Mov Disord 26:264-270.

Steece-Collier K, Chambers LK, Jaw-Tsai SS, Menniti FS and Greenamyre JT (2000) at ASPET Journals on October 1, 2021 Antiparkinsonian actions of CP-101,606, an antagonist of NR2B subunit-containing N-

methyl-d-aspartate receptors. Exp Neurol 163:239-243.

Stibe C, Lees A and Stern G (1987) Subcutaneous infusion of apomorphine and in the

treatment of parkinsonian on-off fluctuations. Lancet 1:871.

Stocchi F, Rascol O, Destee A, Hattori N, Hauser RA, Lang AE, Poewe W, Stacy M, Tolosa E,

Gao H, Nagel J, Merschhemke M, Graf A, Kenney C and Trenkwalder C (2013) AFQ056

in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-

finding study. Mov Disord 28:1838-1846.

Stocchi F, Rascol O, Hauser RA, Huyck S, Tzontcheva A, Capece R, Ho TW, Sklar P, Lines C,

Michelson D, Hewitt DJ and Preladenant Early Parkinson Disease Study G (2017)

Randomized trial of preladenant, given as monotherapy, in patients with early Parkinson

disease. Neurology 88:2198-2206.

63 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Stockwell KA, Scheller D, Rose S, Jackson MJ, Tayarani-Binazir K, Iravani MM, Smith LA,

Olanow CW and Jenner P (2009) Continuous administration of rotigotine to MPTP-treated

common marmosets enhances anti-parkinsonian activity and reduces dyskinesia induction.

Exp Neurol 219:533-542.

Stockwell KA, Scheller DK, Smith LA, Rose S, Iravani MM, Jackson MJ and Jenner P (2010)

Continuous rotigotine administration reduces dyskinesia resulting from pulsatile treatment

with rotigotine or L-DOPA in MPTP-treated common marmosets. Exp Neurol 221:79-85. Downloaded from

Stockwell KA, Virley DJ, Perren M, Iravani MM, Jackson MJ, Rose S and Jenner P (2008)

Continuous delivery of ropinirole reverses motor deficits without dyskinesia induction in jpet.aspetjournals.org MPTP-treated common marmosets. Exp Neurol 211:172-179.

Stoessl AJ, Mak E and Calne DB (1985) (+)-4-Propyl-9-hydroxynaphthoxazine (PHNO), a new

dopaminomimetic, in treatment of parkinsonism. Lancet 2:1330-1331. at ASPET Journals on October 1, 2021 Svenningsson P, Rosenblad C, Af Edholm Arvidsson K, Wictorin K, Keywood C, Shankar B,

Lowe DA, Bjorklund A and Widner H (2015) Eltoprazine counteracts l-DOPA-induced

dyskinesias in Parkinson's disease: a dose-finding study. Brain 138:963-973.

Tagai K, Nagata T, Shinagawa S, Tsuno N, Ozone M and Nakayama K (2013) Mirtazapine

improves visual hallucinations in Parkinson's disease: a case report. Psychogeriatrics

13:103-107.

Tamim MK, Samadi P, Morissette M, Gregoire L, Ouattara B, Levesque D, Rouillard C and Di

Paolo T (2010) Effect of non-dopaminergic drug treatment on Levodopa induced

dyskinesias in MPTP monkeys: common implication of striatal neuropeptides.

Neuropharmacology 58:286-296.

Tarsy D, Leopold N and Sax DS (1974) Physostigmine in choreiform movement disorders.

Neurology 24:28-33.

64 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Tayarani-Binazir K, Jackson MJ, Rose S, McCreary AC and Jenner P (2010a) The partial dopamine

agonist pardoprunox (SLV308) administered in combination with l-dopa improves efficacy

and decreases dyskinesia in MPTP treated common marmosets. Exp Neurol.

Tayarani-Binazir KA, Jackson MJ, Rose S, Olanow CW and Jenner P (2010b) Pramipexole

combined with levodopa improves motor function but reduces dyskinesia in MPTP-treated

common marmosets. Mov Disord 25:377-384.

Temlett JA, Chong PN, Oertel WH, Jenner P and Marsden CD (1988) The D-1 dopamine receptor Downloaded from

partial agonist, CY 208-243, exhibits antiparkinsonian activity in the MPTP-treated

marmoset. Eur J Pharmacol 156:197-206. jpet.aspetjournals.org Temlett JA, Quinn NP, Jenner PG, Marsden CD, Pourcher E, Bonnet AM, Agid Y, Markstein R

and Lataste X (1989) Antiparkinsonian activity of CY 208-243, a partial D-1 dopamine

receptor agonist, in MPTP-treated marmosets and patients with Parkinson's disease. Mov at ASPET Journals on October 1, 2021 Disord 4:261-265.

Teychenne PF, Park DM, Findley LJ, Rose FC and Calne DB (1976) Nomifensine in parkinsonism.

J Neurol Neurosurg Psychiatry 39:1219-1221.

Tison F, Keywood C, Wakefield M, Durif F, Corvol JC, Eggert K, Lew M, Isaacson S, Bezard E,

Poli SM, Goetz CG, Trenkwalder C and Rascol O (2016) A Phase 2A Trial of the Novel

mGluR5-Negative Allosteric Modulator Dipraglurant for Levodopa-Induced Dyskinesia in

Parkinson's Disease. Mov Disord 31:1373-1380.

Tison F, Negre-Pages L, Meissner WG, Dupouy S, Li Q, Thiolat ML, Thiollier T, Galitzky M,

Ory-Magne F, Milhet A, Marquine L, Spampinato U, Rascol O and Bezard E (2013)

Simvastatin decreases levodopa-induced dyskinesia in monkeys, but not in a randomized,

placebo-controlled, multiple cross-over ("n-of-1") exploratory trial of simvastatin against

65 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

levodopa-induced dyskinesia in Parkinson's disease patients. Parkinsonism Relat Disord

19:416-421.

Trabucchi M, Bassi S and Frattola L (1982) Effect of naloxone on the "on-off' syndrome in patients

receiving long-term levodopa therapy. Arch Neurol 39:120-121.

Trenkwalder C, Berg D, Rascol O, Eggert K, Ceballos-Baumann A, Corvol JC, Storch A, Zhang

L, Azulay JP, Broussolle E, Defebvre L, Geny C, Gostkowski M, Stocchi F, Tranchant C,

Derkinderen P, Durif F, Espay AJ, Feigin A, Houeto JL, Schwarz J, Di Paolo T, Feuerbach Downloaded from

D, Hockey HU, Jaeger J, Jakab A, Johns D, Linazasoro G, Maruff P, Rozenberg I, Sovago

J, Weiss M and Gomez-Mancilla B (2016a) A Placebo-Controlled Trial of AQW051 in jpet.aspetjournals.org Patients With Moderate to Severe Levodopa-Induced Dyskinesia. Mov Disord 31:1049-

1054.

Trenkwalder C, Stocchi F, Poewe W, Dronamraju N, Kenney C, Shah A, von Raison F and Graf at ASPET Journals on October 1, 2021 A (2016b) Mavoglurant in Parkinson's patients with l-Dopa-induced dyskinesias: Two

randomized phase 2 studies. Mov Disord 31:1054-1058.

Treseder SA, Jackson M and Jenner P (2000) The effects of central aromatic amino acid DOPA

decarboxylase inhibition on the motor actions of L-DOPA and dopamine agonists in

MPTP-treated primates. Br J Pharmacol 129:1355-1364.

Tsui JK, Wolters EC, Peppard RF and Calne DB (1989) A double-blind, placebo-controlled, dose-

ranging study to investigate the safety and efficacy of CY 208-243 in patients with

Parkinson's disease. Neurology 39:856-858.

Tyne H, Brooks J, Taylor J, Vinjamuri S, Baker G and Steiger M (2007) A double blind placebo

controlled study of modafinil for Parkinson's disease related fatigue. Parkinsonism Relat

Disord 13 Suppl 2:S113-S114.

66 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Tyne HL, Taylor J, Baker GA and Steiger MJ (2010) Modafinil for Parkinson's disease fatigue. J

Neurol 257:452-456.

Uchida S, Soshiroda K, Okita E, Kawai-Uchida M, Mori A, Jenner P and Kanda T (2015a) The

adenosine A2A receptor antagonist, istradefylline enhances anti-parkinsonian activity

induced by combined treatment with low doses of L-DOPA and dopamine agonists in

MPTP-treated common marmosets. Eur J Pharmacol 766:25-30.

Uchida S, Soshiroda K, Okita E, Kawai-Uchida M, Mori A, Jenner P and Kanda T (2015b) The Downloaded from

adenosine A2A receptor antagonist, istradefylline enhances the anti-parkinsonian activity

of low doses of dopamine agonists in MPTP-treated common marmosets. Eur J Pharmacol jpet.aspetjournals.org 747:160-165.

Uchida S, Tashiro T, Kawai-Uchida M, Mori A, Jenner P and Kanda T (2014) Adenosine A(2)A-

receptor antagonist istradefylline enhances the motor response of L-DOPA without at ASPET Journals on October 1, 2021 worsening dyskinesia in MPTP-treated common marmosets. J Pharmacol Sci 124:480-485. van der Stelt M, Fox SH, Hill M, Crossman AR, Petrosino S, Di Marzo V and Brotchie JM (2005)

A role for endocannabinoids in the generation of parkinsonism and levodopa-induced

dyskinesia in MPTP-lesioned non-human primate models of Parkinson's disease. FASEB J

19:1140-1142. van Vliet SA, Vanwersch RA, Jongsma MJ, Olivier B and Philippens IH (2008) Therapeutic effects

of Delta9-THC and modafinil in a marmoset Parkinson model. Eur Neuropsychopharmacol

18:383-389.

Vanover KE, Betz AJ, Weber SM, Bibbiani F, Kielaite A, Weiner DM, Davis RE, Chase TN and

Salamone JD (2008) A 5-HT2A receptor inverse agonist, ACP-103, reduces tremor in a rat

model and levodopa-induced dyskinesias in a monkey model. Pharmacol Biochem Behav

90:540-544.

67 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Verhagen Metman L, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM and Chase TN

(1998) Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's

disease. Neurology 50:1323-1326.

Vermeulen RJ, Drukarch B, Sahadat MC, Goosen C, Schoffelmeer AN, Wolters EC and Stoof JC

(1995) Morphine and naltrexone modulate D2 but not D1 receptor induced motor behavior

in MPTP-lesioned monkeys. Psychopharmacology (Berl) 118:451-459.

Vieregge A, Sieberer M, Jacobs H, Hagenah JM and Vieregge P (2001) Transdermal nicotine in Downloaded from

PD: a randomized, double-blind, placebo-controlled study. Neurology 57:1032-1035.

Villafane G, Cesaro P, Rialland A, Baloul S, Azimi S, Bourdet C, Le Houezec J, Macquin-Mavier jpet.aspetjournals.org I and Maison P (2007) Chronic high dose transdermal nicotine in Parkinson's disease: an

open trial. Eur J Neurol 14:1313-1316.

Visanji NP, de Bie RM, Johnston TH, McCreary AC, Brotchie JM and Fox SH (2008) The at ASPET Journals on October 1, 2021 nociceptin/orphanin FQ (NOP) receptor antagonist J-113397 enhances the effects of

levodopa in the MPTP-lesioned nonhuman primate model of Parkinson's disease. Mov

Disord 23:1922-1925.

Visanji NP, Fox SH, Johnston T, Reyes G, Millan MJ and Brotchie JM (2009a) Dopamine D3

receptor stimulation underlies the development of L-DOPA-induced dyskinesia in animal

models of Parkinson's disease. Neurobiol Dis 35:184-192.

Visanji NP, Fox SH, Johnston TH, Millan MJ and Brotchie JM (2009b) Alpha1-adrenoceptors

mediate dihydroxyphenylalanine-induced activity in 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine-lesioned macaques. J Pharmacol Exp Ther 328:276-283.

Visanji NP, Gomez-Ramirez J, Johnston TH, Pires D, Voon V, Brotchie JM and Fox SH (2006)

Pharmacological characterization of psychosis-like behavior in the MPTP-lesioned

nonhuman primate model of Parkinson's disease. Mov Disord 21:1879-1891.

68 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Weintraub D, Koester J, Potenza MN, Siderowf AD, Stacy M, Voon V, Whetteckey J, Wunderlich

GR and Lang AE (2010) Impulse control disorders in Parkinson disease: a cross-sectional

study of 3090 patients. Arch Neurol 67:589-595.

Wolz M, Lohle M, Strecker K, Schwanebeck U, Schneider C, Reichmann H, Grahlert X, Schwarz

J and Storch A (2010) Levetiracetam for levodopa-induced dyskinesia in Parkinson's

disease: a randomized, double-blind, placebo-controlled trial. J Neural Transm 117:1279-

1286. Downloaded from

Wong KK, Alty JE, Goy AG, Raghav S, Reutens DC and Kempster PA (2011) A randomized,

double-blind, placebo-controlled trial of levetiracetam for dyskinesia in Parkinson's jpet.aspetjournals.org disease. Mov Disord 26:1552-1555.

Wright A, Lees A and Stern GM (1986) Mesulergine and pergolide in previously untreated

Parkinson's disease. J Neurol Neurosurg Psychiatry 49:482-484. at ASPET Journals on October 1, 2021 Yan T, Rizak JD, Yang S, Li H, Huang B, Ma Y and Hu X (2014) Acute morphine treatments

alleviate tremor in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated monkeys. PLoS

One 9:e88404.

Zeng BY, Iravani MM, Jackson MJ, Rose S, Parent A and Jenner P (2008) Morphological changes

in serotonergic neurites in the striatum of levodopa primed MPTP-treated common

marmosets [Abstract]. Soc Neurosci.

Zhang D, Bordia T, McGregor M, McIntosh JM, Decker MW and Quik M (2014) ABT-089 and

ABT-894 reduce levodopa-induced dyskinesias in a monkey model of Parkinson's disease.

Mov Disord 29:508-517.

Zhang D, Mallela A, Sohn D, Carroll FI, Bencherif M, Letchworth S and Quik M (2013) Nicotinic

receptor agonists reduce L-DOPA-induced dyskinesias in a monkey model of Parkinson's

disease. J Pharmacol Exp Ther 347:225-234.

69 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Zhang D, McGregor M, Bordia T, Perez XA, McIntosh JM, Decker MW and Quik M (2015) alpha7

nicotinic receptor agonists reduce levodopa-induced dyskinesias with severe nigrostriatal

damage. Mov Disord 30:1901-1911.

Zubair M, Jackson MJ, Tayarani-Binazir K, Stockwell KA, Smith LA, Rose S, Olanow W and

Jenner P (2007) The administration of entacapone prevents L-dopa-induced dyskinesia

when added to dopamine agonist therapy in MPTP-treated primates. Exp Neurol 208:177-

184. Downloaded from

jpet.aspetjournals.org at ASPET Journals on October 1, 2021

70 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Footnotes

i iFunding: PH holds research support from Parkinson Canada, Fonds de Recherche Québec –

Santé, Natural Sciences and Engineering Research Council of Canada and the Weston Brain

Institute.

Downloaded from jpet.aspetjournals.org at ASPET Journals on October 1, 2021

71 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure legends

Figure 1 A total of 98 experimental drugs were tested in the MPTP-lesioned macaque, 97 in the MPTP- lesioned marmoset and 9 in the MPTP-lesioned squirrel monkey.

Figure 2 A total of 44 drugs that are either clinically-approved or that underwent clinical testing in PD were tested in the MPTP-lesioned macaque, 38 in the MPTP-lesioned marmoset and 1 in the MPTP-lesioned squirrel monkey.

Figure 3 The anti-dyskinetic positive predictive value of the MPTP-lesioned macaque is 87.5%, while it is 76.9% for the MPTP-lesioned marmoset. Downloaded from

Figure 4 The anti-dyskinetic negative predictive value of the MPTP-lesioned macaque is 61.9%, while it is 86.4% for the MPTP-lesioned marmoset. jpet.aspetjournals.org

Figure 5 The anti-dyskinetic false positive rate of the MPTP-lesioned macaque is 38.1%, while it is 15.6% for the MPTP-lesioned marmoset.

Figure 6 at ASPET Journals on October 1, 2021 The anti-parkinsonian positive predictive value of the MPTP-lesioned macaque is 68.2%, while it is 86.9% for the MPTP-lesioned marmoset.

Figure 7 The anti-parkinsonian negative predictive value of the MPTP-lesioned macaque is 33.3%, while it is 50.0% for the MPTP-lesioned marmoset.

Figure 8 The anti-parkinsonian false positive rate of the MPTP-lesioned macaque is 44.4%, while it is 41.7% for the MPTP-lesioned marmoset.

Figure 9 The worsening of parkinsonism positive predictive value of the MPTP-lesioned macaque is 75.0%, while it is 100.0% for the MPTP-lesioned marmoset.

72 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 1

number of drugs tested in the MPTP-lesioned non-human primate

100

90

80

70

60 Downloaded from 50

num be r of dr ugs 40

30 jpet.aspetjournals.org 20

10

0 macaque marmoset squirrel monkey

at ASPET Journals on October 1, 2021

73 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 2

number of drugs assessed in the clinic or clinically-approved that were tested in the MPTP-lesioned non-human primate

45

40

35

30 Downloaded from 25

20 num be r of dr ugs

15 jpet.aspetjournals.org 10

5

0 macaque marmoset squirrel monkey

at ASPET Journals on October 1, 2021

74 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 3

anti-dyskinetic positive predictive value of the MPTP- lesioned non-human primate

100

90

80

70

60 Downloaded from

50

40 pr e di c t i ve va l ue ( % )

30 jpet.aspetjournals.org 20

10

0 macaque marmoset

at ASPET Journals on October 1, 2021

75 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 4

anti-dyskinetic negative predictive value of the MPTP- lesioned non-human primate

100

90

80

70

60 Downloaded from

50

40 pr e di c t i ve va l ue ( % )

30 jpet.aspetjournals.org 20

10

0 macaque marmoset

at ASPET Journals on October 1, 2021

76 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 5

anti-dyskinetic false positive rate of the MPTP-lesioned non-human primate

50

40

30 Downloaded from r a t e ( % ) 20 jpet.aspetjournals.org 10

0 macaque marmoset

at ASPET Journals on October 1, 2021

77 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 6

anti-parkinsonian positive predictive value of the MPTP- lesioned non-human primate

100

90

80

70

60 Downloaded from

50

40 pr e di c t i ve va l ue ( % )

30 jpet.aspetjournals.org 20

10

0 macaque marmoset

at ASPET Journals on October 1, 2021

78 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 7

anti-parkinsonian negative predictive value of the MPTP- lesioned non-human primate

50

40

30 Downloaded from

20 pr e di c t i ve va l ue ( % ) jpet.aspetjournals.org 10

0 macaque marmoset

at ASPET Journals on October 1, 2021

79 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 8

anti-parkinsonian false positive rate of the MPTP-lesioned non-human primate

50

45

40

35

30 Downloaded from

25 r a t e ( % ) 20

15 jpet.aspetjournals.org 10

5

0 macaque marmoset

at ASPET Journals on October 1, 2021

80 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Figure 9

worsening of parkinsonism positive predictive value of the MPTP-lesioned non-human primate

100

90

80

70

60 Downloaded from

50

40 pr e di c t i ve va l ue ( % )

30 jpet.aspetjournals.org 20

10

0 macaque marmoset

at ASPET Journals on October 1, 2021

81 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Table 1: Drugs tested in the MPTP-lesioned macaque

drug dyskinesia parkinsonism psychosis references

17-alpha-oestradiol (Gomez-Mancilla ne ne na [N=1] and Bedard, 1992b)

(Gomez-Mancilla

17-beta-oestradiol and Bedard, 1992b; Ö Ö na [N=2] Belanger et al., Downloaded from

2003a)

(-)-3-(3-

(Gomez-Mancilla jpet.aspetjournals.org hydroxyphenyl)-N-n- ne Ö na and Bedard, 1991) propylpiperone [N=1]

(Gomez-Mancilla at ASPET Journals on October 1, 2021 and Bedard, 1991;

Gomez-Mancilla

and Bedard, 1992a; (+)-4-propyl-9- Luquin et al., 1992; hydroxynaphthoxazine ne Ö na Blanchet et al., [N=7] 1993; Luquin et al.,

1993b; Belluzzi et

al., 1994; Blanchet

et al., 1994)

(Hadj Tahar et al., (-)-OSU-6162 [N=1] Ö ne na 2001)

82 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Gomez-Mancilla 5-MDOT [N=1] Ö de na and Bedard, 1993)

(Munoz et al., 8-OH-DPAT [N=1] Ö ne na 2008)

(Blanchet et al.,

A-77,636 [N=2] Ö na 1993; Blanchet et

al., 1996b) Downloaded from

(Grondin et al., A-86,929 [N=1] Ö na 1997) jpet.aspetjournals.org (Koprich et al., ADL-5510 [N=1] Ö na 2011)

(Gregoire et al., AFQ-056 [N=1] Ö Ö na at ASPET Journals on October 1, 2021 2011)

(Blanchet et al.,

1998; Bibbiani et

al., 2005b; Rylander

et al., 2010; Bezard

amantadine [N=7] Ö de na et al., 2013b;

Gregoire et al.,

2013; Ko et al.,

2014b; Shen et al.,

2015)

83 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Bezard et al., anpirtoline [N=1] Ö de na 2013a)

(Filion et al., 1991;

Luquin et al.,

1993a; Akai et al.,

1995; Blanchet et

al., 1997; Doan et Downloaded from apomorphine [N=8] ne Ö na al., 1999; Silverdale

et al., 2002; jpet.aspetjournals.org Bibbiani et al.,

2003; Bibbiani et

al., 2005a) at ASPET Journals on October 1, 2021 (Di Paolo et al., AQW-051 [N=1] Ö Ö na 2014)

(Gomez-Mancilla et atropine [N=1] ne Ö na al., 1991)

(Gomez-Mancilla baclofen [N=1] ne de na and Bedard, 1993)

(Bezard et al., BP-897 [N=1] Ö ne na 2003)

(Bedard et al.,

bromocriptine [N=9] ne Ö na 1986; Falardeau et

al., 1988; Gagnon et

84 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

al., 1990; Rouillard

et al., 1990;

Gomez-Mancilla

and Bedard, 1991;

Blanchet et al.,

1993; Gomez-

Mancilla et al., Downloaded from

1993; Belluzzi et

al., 1994; Gagnon et jpet.aspetjournals.org al., 1995)

(Grondin et al.,

1996; Morissette et at ASPET Journals on October 1, 2021 al., 1998; Calon et

al., 1999; Goulet et

al., 1999;

Morissette et al.,

cabergoline [N=20] ne Ö na 1999; Calon et al.,

2000; Goulet et al.,

2000; Hadj Tahar et

al., 2000b; Calon et

al., 2002; Belanger

et al., 2003b;

Samadi et al.,

85 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

2008a; Samadi et

al., 2008b; Samadi

et al., 2008c;

Ouattara et al.,

2009; Morissette et

al., 2010; Ouattara

et al., 2010; Samadi Downloaded from

et al., 2010;

Ouattara et al., jpet.aspetjournals.org 2011; Riahi et al.,

2011)

CE [N=1] ne Ö na (Cao et al., 2007) at ASPET Journals on October 1, 2021 (Fidalgo et al., citalopram [N=1] Ö de na 2015)

(Hadj Tahar et al.,

2004; Morissette et

al., 2006a;

Morissette et al.,

CI-1041 [N=14] Ö ne na 2006b; Samadi et

al., 2008a; Samadi

et al., 2008b;

Samadi et al.,

2008c; Ouattara et

86 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

al., 2009;

Morissette et al.,

2010; Ouattara et

al., 2010; Samadi et

al., 2010; Tamim et

al., 2010; Ouattara

et al., 2011; Riahi et Downloaded from

al., 2011)

(Gomez-Mancilla et jpet.aspetjournals.org al., 1991; Gomez- clonidine [N=2] Ö Ö na Mancilla and

Bedard, 1993) at ASPET Journals on October 1, 2021 (Grondin et al., clozapine [N=1] Ö ne na 1999b)

(Koprich et al., CLR-151 [N=1] Ö de Ö 2013)

Co-101,244/PD-174,494 (Blanchet et al., Ö ne na [N=1] 1999)

(Munoz et al., CP-94,253 [N=1] ne ne na 2008)

(Steece-Collier et CP-101,606 [N=1] ne Ö na al., 2000)

87 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Konitsiotis et al., CX-516 [N=1] de ne na 2000)

(Gomez-Mancilla

and Bedard, 1991;

Gomez-Mancilla

and Bedard, 1992a;

Gomez-Mancilla et Downloaded from

al., 1992a; Blanchet CY-208,243 [N=8] ne Ö na et al., 1993; Gagnon jpet.aspetjournals.org et al., 1993;

Gomez-Mancilla et

al., 1993; Luquin et at ASPET Journals on October 1, 2021 al., 1993b; Gagnon

et al., 1995)

(Belanger et al.,

DHEA [N=2] ne Ö na 2003a; Belanger et

al., 2006)

(Gomez-Mancilla diazepam [N=1] Ö ne na and Bedard, 1993)

(Bezard et al., dipraglurant [N=1] Ö ne na 2014)

docosahexaenoic acid (Samadi et al., Ö ne na [N=5] 2006; Mahmoudi et

88 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

al., 2009; Tamim et

al., 2010; Riahi et

al., 2012; Gregoire

et al., 2015)

(Bezard et al.,

2013b; Pinna et al., eltoprazine [N=3] Ö de na 2016; Ko et al., Downloaded from

2017)

entacapone [N=1] ne Ö na (Huot et al., 2013) jpet.aspetjournals.org (Gomez-Mancilla et ethosuximide [N=1] ne Ö na al., 1992b)

F-15,599 [N=1] Ö ne na (Huot et al., 2015b) at ASPET Journals on October 1, 2021

(Johnston et al., famotidine [N=1] Ö Ö na 2010d)

(Rylander et al.,

fenobam [N=2] Ö Ö na 2010; Ko et al.,

2014b)

(Johnston et al., fipamezole [N=1] Ö Ö na 2010c)

(Bibbiani et al., GYKI-47,261 [N=1] Ö ne na 2005b)

89 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Bezard et al., 1999;

idazoxan [N=2] Ö Ö na Grondin et al.,

2000)

(Grondin et al.,

1999a; Bibbiani et istradefylline [N=3] ne/de Ö na al., 2003; Ko et al.,

2016) Downloaded from

(Hadj Tahar et al., JL-18 [N=1] Ö de na 2000a) jpet.aspetjournals.org L-745,870 [N=1] Ö ne na (Huot et al., 2012b)

L-tryptophan [N=1] Ö de na (Ko et al., 2014a)

(Bezard et al., levetiracetam [N=1] Ö ne na at ASPET Journals on October 1, 2021 2004)

(Papa and Chase, LY-235,959 [N=1] Ö ne na 1996)

(Blanchet et al., MDL-100,453 [N=1] Ö ne na 1999)

(Gomez-Mancilla meperidine [N=1] Ö ne na and Bedard, 1993)

(Gomez-Mancilla methysergide [N=1] Ö de na and Bedard, 1993)

(Gomez-Mancilla MK-801 [N=1] Ö de na and Bedard, 1993)

90 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Galvan et al., ML-218 [N=1] ne ne na 2016)

(Samadi et al.,

morphine [N=2] Ö Ö na 2004; Yan et al.,

2014)

(Morin et al., 2010;

Morin et al., 2013a; Downloaded from

Morin et al., 2013b;

MPEP [N=7] Ö de na Morin et al., 2015a; jpet.aspetjournals.org Morin et al., 2015b;

Morissette et al.,

2016) at ASPET Journals on October 1, 2021 (Johnston et al.,

MTEP [N=2] Ö de na 2010b; Morin et al.,

2010)

(Bezard et al., nafadotride [N=1] Ö de na 2003)

nalbuphine [N=1] Ö ne na (Potts et al., 2015)

(Gomez-Mancilla

naloxone [N=2] ne ne na and Bedard, 1993;

Samadi et al., 2003)

(Samadi et al., naltrexone [N=5] ne/de ne na 2003; Samadi et al.,

91 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

2005a; Samadi et

al., 2005c; Tamim

et al., 2010;

Koprich et al.,

2011)

(Klockgether et al.,

NBQX [N=2] ne Ö na 1991; Luquin et al., Downloaded from

1993b)

(Grondin et al.,

NNC-01-112 [N=1] Ö de na jpet.aspetjournals.org 1999b)

PAMQX [N=1] na Ö na (Papa et al., 2004)

(Blanchet et al., PD-128,907 [N=1] ne Ö na at ASPET Journals on October 1, 2021 1997)

(Gomez-Mancilla physostigmine [N=1] Ö de na and Bedard, 1993)

(Vanover et al., pimavanserin [N=1] Ö ne na 2008)

pioglitazone [N=1] Ö de na (Huot et al., 2015a)

(Gregoire et al., PPI-1011 [N=1] Ö ne na 2015)

(Visanji et al., prazosin [N=1] ne ne na 2009b)

92 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Hodgson et al.,

2010; Pinna et al., preladenant [N=3] ne Ö na 2016; Ko et al.,

2017)

(Gomez-Mancilla progesterone [N=1] ne ne na and Bedard, 1992b)

(Gomez-Mancilla Downloaded from propranolol [N=1] Ö de na and Bedard, 1993)

quetiapine [N=1] Ö ne na (Oh et al., 2002) jpet.aspetjournals.org (Bedard and

Boucher, 1989;

Gomez-Mancilla at ASPET Journals on October 1, 2021 and Bedard, 1991;

Gomez-Mancilla et

al., 1992a; Blanchet

et al., 1993; quinpirole [N=12] ne Ö na Blanchet et al.,

1994; Akai et al.,

1995; Grondin et

al., 1997; Samadi et

al., 2003; Samadi et

al., 2004; Bibbiani

et al., 2005b;

93 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Samadi et al.,

2005c; Hyacinthe et

al., 2014)

(Greenamyre et al., remacemide [N=1] ne Ö na 1994)

(Samadi et al.,

2005b; Gregoire et Downloaded from

al., 2008; Ouattara

Ro-61,8048 [N=6] Ö ne na et al., 2009; Tamim jpet.aspetjournals.org et al., 2010; Riahi et

al., 2012; Riahi et

al., 2013) at ASPET Journals on October 1, 2021 Ro 65-6570 [N=1] Ö ne na (Marti et al., 2012)

(Belluzzi et al.,

rotigotine [N=2] ne Ö na 1994; Domino and

Ni, 1998a)

(Gregoire et al., safinamide [N=1] Ö Ö na 2013)

(Bibbiani et al.,

sarizotan [N=2] Ö de na 2001; Gregoire et

al., 2009)

(Gomez-Mancilla SCH-23,390 [N=3] Ö de na and Bedard, 1991;

94 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Akai et al., 1995;

Grondin et al.,

1999b)

simvastatin [N=1] Ö ne na (Tison et al., 2013)

(Falardeau et al.,

1988; Bedard and

Boucher, 1989; Downloaded from

Gagnon et al., 1990; SKF-38,393 [N=6] ne Ö/ne na Rouillard et al., jpet.aspetjournals.org 1990; Blanchet et

al., 1993; Hyacinthe

et al., 2014) at ASPET Journals on October 1, 2021 (Blanchet et al.,

1993; Blanchet et

al., 1994; Akai et

al., 1995; Blanchet

et al., 1996a;

SKF-82,958 [N=18] ne Ö na Blanchet et al.,

1996b; Goulet et

al., 1996; Domino

and Ni, 1998a;

Morissette et al.,

1998; Calon et al.,

95 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

1999; Goulet et al.,

1999; Grondin et

al., 1999c;

Morissette et al.,

1999; Calon et al.,

2000; Calon et al.,

2002; Samadi et al., Downloaded from

2003; Samadi et al.,

2004; Bibbiani et jpet.aspetjournals.org al., 2005b; Samadi

et al., 2005c)

(Bezard et al.,

ST-198 [N=1] Ö de na at ASPET Journals on October 1, 2021 2003)

(Gomez-Mancilla

and Bedard, 1991;

sulpiride [N=3] Ö de na Akai et al., 1995;

Grondin et al.,

1999b)

(McCall et al., sumanirole [N=1] ne Ö na 2005)

(Konitsiotis et al., talampanel [N=1] Ö Ö na 2000)

tamoxifen [N=1] Ö ne na (Smith et al., 2007)

96 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Johnston et al., TC-8831 [N=1] Ö ne na 2013b)

(Gomez-Mancilla terguride [N=1] ne Ö na and Bedard, 1991)

(Domino and Ni,

trihexyphenidyl [N=2] ne Ö na 1998b; Domino and

Ni, 1998a) Downloaded from

(Blanchet et al.,

1995; Calon et al., jpet.aspetjournals.org U-91,356-A [N=4] ne Ö na 1995; Goulet et al.,

1997; Morissette et

al., 1997) at ASPET Journals on October 1, 2021 (Blanchet et al., U-99,194-A [N=1] Ö ne na 1997)

VU-0,476,406 [N=1] Ö ne na (Shen et al., 2015)

(Gomez-Mancilla yohimbine [N=1] Ö ne na and Bedard, 1993)

Ö: effective; ne: not effective; de: deleterious; na: not assessed

The number of studies reporting the effect of each drug is in brackets.

97 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Table 2: drugs tested in the MPTP-lesioned marmoset

drug dyskinesia parkinsonism psychosis references

D-(9)-THC (van Vliet et al., ne Ö na [N=1] 2008)

(Nomoto et al., (+)-PHNO ne Ö na 1987; Close et [N=2]

al., 1990) Downloaded from

(+)-N-n-propyl-

3-(3-

(Close et al., jpet.aspetjournals.org hydroxyphenyl) ne Ö na 1990) -piperidine [(+)-

3PPP] [N=1] at ASPET Journals on October 1, 2021 (-)-N-n-propyl-

3-(3- (Close et al., hydroxyphenyl) ne ne na 1990) -piperidine (-)-

3PPP [N=1]

(-)-N-0437 (Loschmann et ne Ö na [N=1] al., 1989)

(-)-OSU-6162 (Ekesbo et al., Ö ne na [N=1] 1997)

2-amino-5,6- (Close et al., ne Ö na dihydroxytetral 1990)

98 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

in (N,N-

dipropyl A-5,6-

DTN) [N=1]

R-(-)-11-OH- (Lincoln et al., ne Ö na NPa [N=1] 2016)

(Gnanalingham A-66,359 [N=1] ne de na et al., 1995c) Downloaded from

(Kebabian et al.,

1992; Pearce et jpet.aspetjournals.org al., 1995; Pearce A-77,636 [N=4] ne Ö na et al., 1999;

Smith et al., at ASPET Journals on October 1, 2021 2002a)

(Shiosaki et al.,

1996; Pearce et

A-86,929 [N=3] ne Ö na al., 1999;

Treseder et al.,

2000)

(Shiosaki et al., ABT-431 [N=1] ne Ö na 1996)

(Hill et al., amantadine Ö ne de 2004b; Visanji et [N=3] al., 2006;

99 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Kobylecki et al.,

2011)

(Jackson et al., aplindore [N=1] ne Ö na 2010)

(Loschmann et

al., 1992; Pearce

et al., 1995; Fox Downloaded from

et al., 2001; apomorphine ne Ö ne Maratos et al.,

[N=6] jpet.aspetjournals.org 2003; Visanji et

al., 2006;

Lincoln et al., at ASPET Journals on October 1, 2021 2016)

(Close et al.,

atropine [N=2] ne Ö ne 1990; Jackson et

al., 2014)

benztropine (Close et al., ne Ö na [N=1] 1990)

brasofensine (Pearce et al., ne Ö na [N=1] 2002)

bromocriptine (Close et al., ne Ö na [N=3] 1990; Pearce et

100 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

al., 1995; Pearce

et al., 1998)

(Hansard et al., BTS-74,398 ne Ö/ne na 2002b; Hansard [N=2] et al., 2004)

(Hansard et al., bupropion ne Ö/ne na 2002b; Hansard Downloaded from [N=2] et al., 2011)

(Visanji et al.,

clozapine [N=1] Ö ne Ö jpet.aspetjournals.org 2006)

CP-101,606 (Nash et al., ne Ö na [N=1] 2004) at ASPET Journals on October 1, 2021 (Loschmann et CPP [N=1] ne Ö na al., 1991)

(Temlett et al., CY-208,243 ne Ö na 1988; Temlett et [N=2] al., 1989)

(Henry et al., Ö ne ne [N=1] 2001)

(Smith et al.,

entacapone 1997; Smith et ne Ö na [N=4] al., 2003; Smith

et al., 2005;

101 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Zubair et al.,

2007)

fipamezole (Savola et al., Ö Ö na [N=1] 2003)

(Hansard et al., GBR-12,909 ne Ö na 2002b; Hansard [N=2] et al., 2002a) Downloaded from

haloperidol (Visanji et al., Ö de Ö [N=1] 2006) jpet.aspetjournals.org (Henry et al., idazoxan [N=1] Ö Ö na 1999)

IEM-1460 (Kobylecki et Ö ne na at ASPET Journals on October 1, 2021 [N=1] al., 2010)

ifenprodil (Nash et al., ne Ö na [N=1] 2000)

(Gomez-

[N=1] Ö ne na Ramirez et al.,

2006)

(Gomez-

[N=1] Ö ne na Ramirez et al.,

2006)

istradefylline (Kanda et al., ne Ö na [N=6] 1998a; Kanda et

102 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

al., 1998b;

Kanda et al.,

2000; Uchida et

al., 2014; Uchida

et al., 2015a;

Uchida et al.,

2015b) Downloaded from

(Visanji et al., J-113,397 [N=1] Ö ne na 2008) jpet.aspetjournals.org JNJ-27,063,699 (Philippens et ne Ö na [N=1] al., 2014)

(Hill et al., 2003; at ASPET Journals on October 1, 2021 levetiracetam Hill et al., Ö ne na [N=3] 2004a; Hill et

al., 2004b)

LY-141,865 (Nomoto et al., ne Ö na [N=1] 1985)

(Iravani et al., MDMA [N=1] Ö Ö na 2003)

melanocyte- (Katzenschlager inhibiting ne ne na et al., 2007) factor [N=1]

103 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

mianserin (Hamadjida et Ö de Ö [N=1] al., 2018)

mirtazapine (Hamadjida et Ö ne Ö [N=1] al., 2017)

(Jenner et al.,

modafinil [N=2] ne Ö na 2000; van Vliet

et al., 2008) Downloaded from

(Loschmann et N-0437 [N=1] ne Ö na al., 1989) jpet.aspetjournals.org N-methyl (Jackson et al., scopolamine ne Ö na 2014) [N=1] at ASPET Journals on October 1, 2021 nabilone [N=1] Ö ne na (Fox et al., 2002)

naltrexone (Henry et al., Ö ne na [N=1] 2001)

naltrindole (Henry et al., Ö ne na [N=1] 2001)

neostigmine (Jackson et al., ne Ö na [N=1] 2014)

(Hansard et al., ne Ö/de na [N=1] 2002b)

(Loschmann et NBQX [N=1] ne Ö na al., 1991)

104 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Close et al., nomifensine ne Ö na 1990; Hansard et [N=2] al., 2002b)

(Jackson et al., ne de na [N=1] 2014)

(Johnston et al.,

2010a; Jones et Downloaded from pardoprunox Ö Ö na al., 2010; [N=3] Tayarani-Binazir jpet.aspetjournals.org et al., 2010a)

(Pearce et al.,

1995; Maratos et at ASPET Journals on October 1, 2021 al., 2003; Fox et

pergolide [N=5] ne Ö na al., 2006b;

Uchida et al.,

2015a; Uchida et

al., 2015b)

physostigmine (Jackson et al., ne de na [N=1] 2014)

(Smith et al.,

1996; Smith et piribedil [N=4] ne Ö na al., 2000; Smith

et al., 2002b;

105 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Smith et al.,

2006)

(Iravani et al.,

2003; Fox et al.,

pramipexole 2006b; Iravani et ne Ö na [N=4] al., 2006;

Tayarani-Binazir Downloaded from

et al., 2010b)

quetiapine (Visanji et al.,

Ö ne Ö jpet.aspetjournals.org [N=1] 2006)

(Nomoto et al.,

1988; Close et at ASPET Journals on October 1, 2021 al., 1990;

Loschmann et

al., 1992;

quinpirole Gnanalingham et ne Ö na [N=8] al., 1995a;

Gnanalingham et

al., 1995b;

Pearce et al.,

1995; Kanda et

al., 2000;

106 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Treseder et al.,

2000)

(R)-(+)- (Iravani et al., OHDPAT Ö de na 2006) [N=1]

R-MDMA (Huot et al., Ö ne Ö [N=1] 2011) Downloaded from

(Loschmann et

al., 1992; jpet.aspetjournals.org Gnanalingham et raclopride Ö de na al., 1995c; [N=4] Ekesbo et al., at ASPET Journals on October 1, 2021 1997; Smith et

al., 2002a)

rauwolscine (Henry et al., Ö ne na [N=1] 1999)

RGFP-109 (Johnston et al., Ö ne na [N=1] 2013a)

rimonabant (van der Stelt et Ö Ö na [N=1] al., 2005)

Ro-25,6981 (Loschmann et ne Ö na [N=1] al., 2004)

107 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Pearce et al.,

1998; Maratos et

al., 2001; Hill et

al., 2003; Millan

et al., 2004;

Silverdale et al.,

2004; Fox et al., Downloaded from

2006b; Jackson

ropinirole et al., 2007; ne Ö na jpet.aspetjournals.org [N=13] Zubair et al.,

2007; Stockwell

et al., 2008; at ASPET Journals on October 1, 2021 Jackson et al.,

2010; Johnston

et al., 2010a;

Uchida et al.,

2015a; Uchida et

al., 2015b)

(Rose et al.,

2007; Stockwell

rotigotine [N=3] ne Ö na et al., 2009;

Stockwell et al.,

2010)

108 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Millan et al.,

S-32,504 [N=2] ne Ö na 2004; Hill et al.,

2006)

(Silverdale et al.,

2004; Hill et al., S-33,084 [N=3] ne Ö na 2006; Visanji et

al., 2009a) Downloaded from

S-MDMA (Huot et al., de Ö ne [N=1] 2011) jpet.aspetjournals.org S,S- (Hansard et al., hydroxybuprop ne Ö na 2011) ion [N=1] at ASPET Journals on October 1, 2021 SB-224,289-A (Jackson et al., ne ne na [N=1] 2004)

(Temlett et al.,

1988;

Loschmann et SCH-23,390 ne de na al., 1992; [N=4] Gnanalingham et

al., 1995c; Smith

et al., 2002a)

109 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Close et al., scopolamine ne Ö na 1990; Jackson et [N=2] al., 2014)

(Hansard et al., sertraline [N=1] ne de na 2002b)

(Nomoto et al.,

1985; Nomoto et Downloaded from

al., 1988; Close

et al., 1990; jpet.aspetjournals.org SKF-38,393 Loschmann et ne ne/de na [N=6] al., 1992;

Gnanalingham et at ASPET Journals on October 1, 2021 al., 1995a;

Gnanalingham et

al., 1995b)

(Gnanalingham

SKF-75,670 et al., 1995a; ne de na [N=2] Gnanalingham et

al., 1995b)

(Gnanalingham

SKF-80,723 et al., 1995a; ne Ö/de na [N=4] Gnanalingham et

al., 1995b;

110 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Gnanalingham et

al., 1995c;

Kanda et al.,

2000)

(Gnanalingham

SKF-82,958 et al., 1995a; ne Ö/de na [N=2] Gnanalingham et Downloaded from

al., 1995b)

SKF-83,565 (Gnanalingham

ne Ö na jpet.aspetjournals.org [N=1] et al., 1995a)

(Gnanalingham

et al., 1995a; at ASPET Journals on October 1, 2021 SKF-83,959 Gnanalingham et ne Ö na [N=3] al., 1995b;

Gnanalingham et

al., 1995c)

SKF-99,101-H (Jackson et al., Ö ne/de na [N=1] 2004)

(Hille et al., SNC-80 [N=1] ne Ö na 2001)

(Rose et al., ST-1535 [N=1] ne Ö na 2006)

111 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Temlett et al.,

sulpiride [N=2] ne de na 1988; Jones et

al., 2010)

(Lange et al., terguride [N=1] ne Ö na 1992)

(Silverdale et al., topiramate Ö ne na 2005; Kobylecki Downloaded from [N=2] et al., 2011)

trihexyphenidyl (Jackson et al.,

ne Ö na jpet.aspetjournals.org [N=1] 2014)

(Johnston et al., URB-597 [N=1] ne ne ne 2011) at ASPET Journals on October 1, 2021 (Huot et al., UWA-101 ne Ö ne 2012a; Johnston [N=2] et al., 2012)

UWA-121 (Huot et al., ne Ö ne [N=1] 2014)

UWA-122 (Huot et al., Ö Ö ne [N=1] 2014)

yohimbine (Henry et al., Ö ne na [N=1] 1999)

Ö: effective; ne: not effective; de: deleterious; na: not assessed

The number of studies reporting the effect of each drug is in brackets.

112 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Table 3: Drugs tested in the MPTP-lesioned squirrel monkey

drug dyskinesia parkinsonism psychosis references

(Zhang et al., ABT-089 [N=1] Ö ne na 2014)

(Zhang et al., ABT-107 [N=1] Ö ne na 2013)

(Zhang et al., Downloaded from ABT-126 [N=1] Ö ne na 2015)

(Zhang et al., jpet.aspetjournals.org 2013; Zhang et ABT-894 [N=3] Ö ne na al., 2014; Zhang

et al., 2015) at ASPET Journals on October 1, 2021 (Hsu et al., BP-897 [N=1] Ö de na 2004)

(Quik et al.,

2007; Quik et

al., 2013a; Quik nicotine [N=4] Ö ne na et al., 2013b;

Zhang et al.,

2015)

(Zhang et al., TC-8831 [N=1] Ö ne na 2013)

113 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Cox et al., U50-488 [N=1] Ö de na 2007)

varenicline (Zhang et al., Ö ne na [N=1] 2013)

Ö: effective; ne: not effective; de: deleterious; na: not assessed

The number of studies reporting the effect of each drug is in brackets.

Downloaded from jpet.aspetjournals.org at ASPET Journals on October 1, 2021

114 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

Table 4: drugs tested in the MPTP-lesioned primate that were tested in the clinic or are

clinically-available

drug dyskinesia parkinsonism psychosis references

(Stoessl et al., (+)-4-propyl-9- 1985; Grandas et hydroxynaphth ne Ö na al., 1987; Muenter oxazine Downloaded from et al., 1988)

(Rascol et al.,

ABT-431 ne Ö na 1999; Rascol et al., jpet.aspetjournals.org

2001b)

(Stocchi et al., at ASPET Journals on October 1, 2021 AFQ-056 Ö/ne ne na 2013; Trenkwalder

et al., 2016b)

(Parkes et al.,

1971; Butzer et al.,

1975; Verhagen

Metman et al.,

amantadine Ö Ö na 1998; Luginger et

al., 2000; Del

Dotto et al., 2001;

Oertel et al., 2017;

Pahwa et al., 2017)

115 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Schwab et al.,

1951; Corsini et apomorphine ne Ö na al., 1979; Stibe et

al., 1987)

(Trenkwalder et al., AQW-051 ne ne na 2016a)

(Boman and Downloaded from atropine ne Ö na Meurman, 1970)

benztropine ne Ö na (Doshay, 1956) jpet.aspetjournals.org (Frackiewicz et al., brasofensine ne ne na 2002)

(Agid et al., 1979; at ASPET Journals on October 1, 2021 Quinn et al., 1981;

Jellinger, 1982;

bromocriptine ne Ö na Hely et al., 1994;

Montastruc et al.,

1994; Castro-

Caldas et al., 2006)

bupropion ne Ö na (Goetz et al., 1984)

(Ahlskog et al.,

1996; Rinne et al., cabergoline ne Ö na 1997; Deuschl et

al., 2007)

116 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Rampello et al.,

2002; Palhagen et citalopram Ö Ö na al., 2008; Palhagen

et al., 2009)

(Shoulson and

Chase, 1976; clonidine na Ö/de na Serrano-Duenas, Downloaded from

2000)

(Durif et al., 1997; jpet.aspetjournals.org FrenchClozapinePa

rkinsonStudyGroup

clozapine Ö Ö Ö , 1999; at ASPET Journals on October 1, 2021 ParkinsonStudyGro

up, 1999; Durif et

al., 2004)

CP-101,606 Ö ne na (Nutt et al., 2008)

(Tsui et al., 1989; CY-208,243 ne Ö na Emre et al., 1992)

(Pourcher et al., diazepam Ö Ö na 1989)

dipraglurant Ö ne na (Tison et al., 2016)

(Svenningsson et eltoprazine Ö ne na al., 2015)

117 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Kaakkola et al.,

1994; Merello et entacapone de Ö na al., 1994; Fenelon

et al., 2003)

(Pourcher et al., ethosuximide ne ne/de na 1992)

(Molinari et al., Downloaded from

famotidine ne ne na 1995; Mestre et al.,

2014) jpet.aspetjournals.org (Lewitt et al., fipamezole Ö ne na 2012)

(Klawans and haloperidol Ö de na at ASPET Journals on October 1, 2021 Weiner, 1974)

(Manson et al.,

idazoxan Ö/ne ne na 2000; Rascol et al.,

2001a)

(Hauser et al.,

2003; Hauser et al.,

istradefylline ne Ö na 2008; LeWitt et al.,

2008; Pourcher et

al., 2012)

(Coppen et al., L-tryptophan ne ne na 1972)

118 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Wolz et al., 2010;

levetiracetam Ö/ne ne ne Stathis et al., 2011;

Wong et al., 2011)

melanocyte- (Gerstenbrand et ne ne na inhibiting factor al., 1975)

(Klawans and methysergide ne ne na Ringel, 1973) Downloaded from

(Ikeguchi and mianserin na na Ö Kuroda, 1995) jpet.aspetjournals.org (Gordon et al.,

2002; Meco et al.,

mirtazapine Ö Ö Ö 2003; Nagata et al., at ASPET Journals on October 1, 2021 2012; Tagai et al.,

2013)

(Tyne et al., 2007; modafinil ne ne na Tyne et al., 2010)

morphine Ö de na (Berg et al., 1999)

(Sieradzan et al., nabilone Ö ne na 2001)

(Trabucchi et al.,

naloxone Ö/ne Ö/ne na 1982; Fox et al.,

2004)

119 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Rascol et al.,

naltrexone Ö/ne ne na 1994; Manson et

al., 2001)

(Vieregge et al.,

2001; Lemay et al., nicotine na Ö/ne na 2004; Villafane et

al., 2007) Downloaded from

(Teychenne et al.,

1976; Bedard et al., jpet.aspetjournals.org nomifensine de Ö na 1977; Park et al.,

1977; Park et al.,

1981) at ASPET Journals on October 1, 2021 (Bronzova et al.,

2010; Sampaio et pardoprunox ne Ö na al., 2011; Rascol et

al., 2012)

(Lieberman et al.,

1986; Wright et al.,

1986; Olanow et pergolide ne Ö na al., 1994; Mizuno

et al., 1995; Oertel

et al., 2006)

120 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Tarsy et al., 1974;

Lindeboom and

physostigmine Ö/ne de na Lakke, 1978;

Clough et al.,

1984)

(Meltzer et al.,

pimavanserin ne ne Ö 2010; Cummings et Downloaded from

al., 2014)

(Investigators,

pioglitazone ne ne na jpet.aspetjournals.org 2015)

(Castro-Caldas et

piribedil ne Ö na al., 2006; Rascol et at ASPET Journals on October 1, 2021 al., 2006b)

(Lieberman et al.,

1997;

pramipexole ne Ö na ParkinsonStudyGro

up, 2000b; Poewe

et al., 2007)

(Hauser et al.,

2011; Factor et al.,

preladenant de Ö/ne na 2013; Hauser et al.,

2015; Stocchi et

al., 2017)

121 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(Koller and

Herbster, 1987; propranolol Ö Ö/ne na Carpentier et al.,

1996)

(Fernandez et al.,

1999; Baron and

Dalton, 2003; Downloaded from

Fernandez et al., quetiapine Ö/ne ne/de Ö/ne 2003; jpet.aspetjournals.org Katzenschlager et

al., 2004; Ondo et

al., 2005) at ASPET Journals on October 1, 2021 (ParkinsonStudyGr

oup, 2000a;

remacemide ne ne na ParkinsonStudyGro

up, 2001; Shoulson

et al., 2001)

(Rascol et al.,

1996; Korczyn et

ropinirole ne Ö na al., 1999; Rascol et

al., 2000; Rascol et

al., 2006a)

122 JPET Fast Forward. Published on March 9, 2018 as DOI: 10.1124/jpet.117.247171 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #247171

(ParkinsonStudyGr

oup, 2003; Giladi

et al., 2007; LeWitt rotigotine ne Ö na et al., 2007;

Mizuno et al.,

2013)

(Borgohain et al., Downloaded from

2014a; Borgohain

et al., 2014b;

safinamide Ö Ö na jpet.aspetjournals.org Cattaneo et al.,

2015; Schapira et

al., 2017) at ASPET Journals on October 1, 2021 (Olanow et al.,

2004; Goetz et al., sarizotan ne de na 2007; Goetz et al.,

2008)

(Gruchet, 1952; scopolamine ne Ö na Duvoisin, 1967)

(Hauser and

Zesiewicz, 1997;

sertraline ne ne na Antonini et al.,

2006; Kulisevsky

et al., 2008;

123 JPET #247171

Marino et al.,

2008)

simvastatin ne ne na (Tison et al., 2013)

SKF-38,393 ne ne na (Braun et al., 1987)

sulpiride Ö de na (Lees et al., 1978)

(Barone et al.,

sumanirole ne Ö/ne na 2007; Singer et al.,

2007)

(Filipova et al.,

terguride ne Ö na 1988; Martignoni

et al., 1995)

(Kobylecki et al.,

topiramate ne/de ne na 2014; Goetz et al.,

2017)

(Martin et al.,

trihexyphenidyl ne Ö na 1974; Lamid and

Jenkins, 1975)

(Montastruc et al., yohimbine ne Ö na 1981)

Ö: effective; ne: not effective; de: deleterious; na: not assessed

124 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Table S1: pharmacological profile of the drugs assessed in the MPTP-lesioned primate

drug mechanism of action category references

D-(9)-THC cannabinoid (Felder et al., CB1,2 agonist (dronabinol) agonist 1995)

(-)-3-(3- (Arnt et al.,

hydroxyphenyl)-N-n- D2 agonist, s ligand D2 agonist 1983; Iyengar et

propylpiperone al., 1991)

(+)-4-propyl-9- (Martin et al., hydroxynaphthoxazine D2 agonist, a2 ligand D2 agonist 1985) (PHNO)

(+)-N-n-propyl-3-(3- (Close et al., hydroxyphenyl)- D2 agonist D2 agonist 1990) piperidine [(+)-3PPP]

(-)-N-n-propyl-3-(3- (Close et al., hydroxyphenyl)- D2 partial agonist D2 partial agonist 1990) piperidine (-)-3PPP

(Loschmann et (-)-N-0437 D2-like agonist D2 agonist al., 1989)

(Burstein et al., (-)-OSU-6162 (PNU- D2 partial agonist, 5-HT2A D2 partial agonist 2011; Carlsson 96,391) partial agonist et al., 2011)

2-amino-5,6- (Mulder et al., D2 agonist D2 agonist dihydroxytetralin 1980; Gower

1 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (N,N-dipropyl A-5,6- and Marriott,

DTN) 1982)

(Tricklebank et

5-MDOT (5-MeO- non-selective 5-HT al., 1985; 5-HT agonist DMT) receptor agonist Spencer et al.,

1987)

(Lejeune et al., 8-OH-DPAT 5-HT1A agonist 5-HT1A agonist 1997)

17-alpha-oestradiol oestrogen receptor agonist, (Kuiper et al., oestrogen agonist (alfatradiol) 5a-reductase inhibitor 1997)

(Kuiper et al., 17-beta-oestradiol oestrogen receptor agonist oestrogen agonist 1997)

(Vanover et al., A-66,359 D1 antagonist D1 antagonist 1991)

(Kebabian et al., A-77,636 D1 agonist D1 agonist 1992)

(Michaelides et

al., 1995; A-86,929 D1 agonist D1 agonist Ehrlich et al.,

1997)

(Marks et al., ABT-089 (pozanicline) b2 nACh agonist 2009)

2 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Malysz et al., ABT-107 a7 nACh agonist nicotinic agonist 2010)

(Zhang et al.,

ABT-126 (nelonicline) a7 nACh agonist nicotinic agonist 2015; Haig et

al., 2016)

(Shiosaki et al., ABT-431 (adrogolide) D1 agonist D1 agonist 1996)

ABT-894 (sofinicline) a4b2 nACh agonist nicotinic agonist (Ji et al., 2007)

(Koprich et al., ADL-5510 µ antagonist µ antagonist 2011)

AFQ-056 (Vranesic et al., mGlu5 NAM mGlu5 NAM (mavoglurant) 2014)

(Matsubayashi

NMDA antagonist, s1 et al., 1997;

amantadine agonist, a7 nACh NMDA antagonist Peeters et al.,

antagonist 2004; Blanpied

et al., 2005)

(Schlicker et al., anpirtoline 5-HT1B > 5-HT1A agonist 5-HT1B agonist 1992)

dopamine partial (Heinrich et al., aplindore (DAB-452) D2,3 partial agonist agonist 2006)

D1 agonist, D2,3,4 partial (Millan et al., apomorphine dopamine agonist agonist, 5-HT2A,2B,2C 2002; Newman-

3 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther

antagonist, 5-HT1A partial Tancredi et al.,

agonist, a2A partial agonist, 2002a;

a2B,2C antagonist Newman-

Tancredi et al.,

2002b)

nicotinic partial (Feuerbach et AQW-051 a7 nACh partial agonist agonist al., 2015)

muscarinic (Hirose et al., atropine M1,2,3,4,5 antagonist antagonist 2001)

(Hirst et al., baclofen GABAB agonist GABAB agonist 2003)

benztropine M1,2 antagonist, DAT muscarinic (Newman et al.,

() inhibitor antagonist 1995)

(Bezard et al.,

BP-897 D3 partial agonist D3 partial agonist 2003; Pilleri and

Antonini, 2015)

non-selective brasofensine (NS-2214, DAT, NET, SERT monoamine re- (Singh, 2000) BMS-204,756) inhibitor uptake inhibitor

D2,3-preferential dopamine

bromocriptine (CB- agonist, 5-HT1A,1D partial (Millan et al., dopamine agonist 154) agonist, a2A,2B,2C 2002)

antagonist

4 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther non-selective (Cheetham et DAT, NET, SERT BTS-74,398 monoamine re- al., 1998; Singh, inhibitor uptake inhibitor 2000)

(Carroll et al., dual monoamine bupropion DAT, NET inhibitor 2009; Lapinsky re-uptake inhibitor et al., 2009)

D2,3 partial agonist, 5- (Millan et al., cabergoline HT1A,1D,2A,2B agonist, dopamine agonist 2002) a2A,2C antagonist

cannabinoid (Cao et al., CE CB1 antagonist antagonist 2007)

(Ben-Daniel et citalopram SERT inhibitor SERT inhibitor al., 2008)

(Barton and CI-1041 (besonprodil, White, 2004; PD-196,860, Co- NR2B antagonist NMDA antagonist Barton et al., 200,461) 2004)

(Ernsberger et

clonidine a2A,2B,2C agonist, I1 agonist a2 agonist al., 1987; Jasper

et al., 1998)

H1 antagonist, 5- (Lahti et al., atypical anti- clozapine HT2A,2B,2C,6,7 antagonist, 1993; Schotte et psychotic a1A,1B,1D antagonist, D4 al., 1996;

5 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther

antagonist, 5-HT1A,1F Yoshio et al.,

agonist 2001; Booth et

al., 2002; Knight

et al., 2004;

Purohit et al.,

2005)

D2 fast-off (Koprich et al., CLR-151 D2 antagonist antagonist 2013)

Co-101,244/PD- (Gill et al., NR2B antagonist NMDA antagonist 174,494 (Ro-631,908) 2002)

(Koe et al., CP-94,253 5-HT1B-preferential agonist 5-HT1B agonist 1992)

(Chenard et al.,

CP-101,606 1995; NR2B antagonist NMDA antagonist () Brimecombe et

al., 1997)

(Feng et al., CPP NR2A,2B,2C,2D antagonist NMDA antagonist 2004)

(Krintel et al., CX-516 (BDP-12) GluA2 PAM 2013)

(Andersen and CY-208,243 D1,2 agonist dopamine agonist Jansen, 1990)

6 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther

5-HT2A,2C,6,7 antagonist, H1 (Leysen et al.,

cyproheptadine antagonist, a1B,1D 5-HT antagonist 1981; Hoyer et

antagonist al., 1994)

(Bergeron et al.,

androgen receptor agonist, 1996; Maurice

a- and b-oestrogen et al., 1997;

DHEA receptor agonist, GABAA androgen agonist Sousa and

NAM, NMDA PAM, s1 Ticku, 1997;

agonist Chen et al.,

2005)

(Pritchett et al., diazepam GABAA PAM GABAA PAM 1989)

dipraglurant (ADX- (Chae et al., mGlu5 NAM mGlu5 NAM 48,621) 2013)

(Lampen et al.,

2001; Itoh et al., FFA1 agonist, RXR-a docosahexaenoic acid RXR agonist 2003; Motter agonist, TRPA1 agonist and Ahern,

2012)

eltoprazine (DU- 5-HT1A,1B agonist, 5-HT2C (Schipper et al., 5-HT1A,1B agonist 28,853) antagonist 1990)

(Lotta et al., entacapone (OR-611) COMT inhibitor COMT inhibitor 1995)

7 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther T-type calcium channel T-type calcium (Gomora et al., ethosuximide antagonist channel antagonist 2001)

(Newman- preferential post-synaptic F-15,599 5-HT1A agonist Tancredi et al., 5-HT1A agonist 2009)

famotidine H2 antagonist (Chremos, 1987)

(Porter et al., fenobam mGlu5 NAM mGlu5 NAM 2005)

(Johnston et al., fipamezole (JP-1730) a2A,2B,2C antagonist a2 antagonist 2010)

GBR-12,909 (Andersen, DAT inhibitor DAT inhibitor () 1989)

(Abraham et al., GYKI-47,261 AMPA antagonist AMPA antagonist 2000)

(Sunahara et al.,

D2-like antagonist, D1 1991; Tice et al.,

haloperidol antagonist, a1 antagonist, D2 antagonist 1994; Kroeze et

s1 antagonist al., 2003; Cobos

et al., 2007)

(Doxey et al.,

1984; idazoxan a2 antagonist, I2 agonist a2 antagonist Regunathan and

Reis, 1996)

8 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Schlesinger et IEM-1460 AMPA antagonist AMPA antagonist al., 2005)

(Reynolds and

Miller, 1989; ifenprodil NR1,2 antagonist NMDA antagonist Korinek et al.,

2011)

(Garbarg et al.,

imetit H3,4 agonist H3,4 agonist 1992; Liu et al.,

2001)

(Liu et al., 2001;

immepip H3,4 agonist H3,4 agonist Kitbunnadaj et

al., 2003)

istradefylline (KW- (Fredholm et al., A2A antagonist A2A antagonist 6002) 2011)

(Kawamoto et

J-113,397 ORL-1 antagonist ORL-1 antagonist al., 1999; Ozaki

et al., 2000)

dopamine (Liegeois et al., JL-18 D2,4 antagonist antagonist 1995)

(Philippens et JNJ-27,063,699 a2C antagonist a2 antagonist al., 2014)

(Patel et al., L-745,870 D4 antagonist D4 antagonist 1997)

9 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Fernstrom, L-tryptophan 5-HT precursor 5-HT agonist 1983)

(Lynch et al., levetiracetam SV2A antagonist SV2A antagonist 2004)

(Tsuruta et al., LY-141,865 D2 agonist D2 agonist 1981)

(Schoepp et al., LY-235,959 (active NMDA antagonist NMDA antagonist 1991; Herman et isomer of LY-274,614) al., 1995)

NR2A-preferential (Blanchet et al., MDL-100,453 NMDA antagonist antagonist 1999)

(Battaglia et al.,

DAT inhibitor, SERT 1988; Nash et dual monoamine MDMA inhibitor, 5-HT2A partial al., 1994; re-uptake inhibitor agonist Verrico et al.,

2007)

melanocyte-inhibiting

factor (Pro-Leu-Gly- (Verma et al., NH2, melanostatin, D2,4 PAM, opiate D2 PAM 2005; Pan and MSH release- antagonist Kastin, 2007) inhibiting hormone,

MIF-1)

10 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Poulain et al., meperidine () µ > k > d agonist opioid agonist 2001)

(Leysen et al.,

1981; Hoyer et

5-HT1D,1F,2A,2C antagonist, al., 1994; methysergide 5-HT antagonist 5-HT2B agonist Bonhaus et al.,

1997; Rothman

et al., 2000)

(Hyttel, 1982;

de Boer et al.,

5-HT2A,2C,3 antagonist, 1988; Hyttel, atypical anti- mianserin a1,2A,2C antagonist, H1 1994; Kooyman depressant antagonist et al., 1994;

Fernandez et al.,

2005)

(de Boer, 1996;

Tatsumi et al., a2A,2C antagonist, 5- mirtazapine (ORG- atypical anti- 1997; Anttila HT2A,2C,3 antagonist, H1 3770) depressant and Leinonen, antagonist 2001; Fernandez

et al., 2005)

NMDA antagonist, a7 (Amador and MK-801 () NMDA antagonist nACh antagonist Dani, 1991)

11 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther T-type calcium channel T-type calcium (Xiang et al., ML-218 antagonist channel antagonist 2011)

(Seeman et al., D2 partial agonist, DAT non-selective DAT 2009; modafinil inhibitor – not fully inhibitor Zolkowska et characterised al., 2009)

(Poulain et al., morphine µ > k > d agonist opioid agonist 2001)

(Gasparini et al.,

MPEP mGlu5 NAM, mGlu4 PAM mGlu5 NAM 1999; Mathiesen

et al., 2003)

(Cosford et al., MTEP mGlu5 NAM mGlu5 NAM 2003)

(Van der Weide

et al., 1986; Van N-0437 D2-like agonist D2 agonist der Weide et al.,

1988)

(Dowling and

Charlton, 2006; muscarinic N-methyl scopolamine M1,3 antagonist Fruchart- antagonist Gaillard et al.,

2006)

12 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther cannabinoid (Blaazer et al., nabilone CB1,2 agonist agonist 2011)

(Sautel et al.,

D2,3 antagonist, 5-HT1A dopamine 1995; Newman- nafadotride agonist antagonist Tancredi et al.,

1998)

(Poulain et al., nalbuphine µ > k > d agonist opioid agonist 2001)

(Poulain et al., naloxone µ = k > d antagonist opioid antagonist 2001)

(Emmerson et

al., 1994; naltrexone µ > k > d antagonist opioid antagonist Koprich et al.,

2011)

(Portoghese et naltrindole d > µ = k antagonist opioid antagonist al., 1988)

AMPA > kainate (Sheardown et NBQX AMPA antagonist antagonist al., 1990)

(Kavitha et al., neostigmine AChE inhibitor ChE inhibitor 2007)

non-selective nACh (Benowitz, nicotine nicotinic agonist agonist 2009)

13 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Wong et al., nisoxetine (LY-94,939) NET inhibitor NET inhibitor 1982)

(Andersen et al.,

1992; Nielsen NNC-01-112 D1 antagonist D1 antagonist and Andersen,

1992)

(Richelson and

Nelson, 1984; nomifensine (HOE- dual monoamine DAT = NET inhibitor, Close et al., 984) re-uptake inhibitor 1990; Tatsumi et

al., 1997)

(Jakubik et al., oxotremorine M1,2,3,4 agonist 1997)

PAMQX (CGP- NMDA antagonist (at (Ametamey et NMDA antagonist 78,608) site) al., 2000)

(Glennon et al., pardoprunox (SLV- D2,3 partial agonist, 5-HT1A dopamine partial 2006; Jones et 308) agonist agonist al., 2010)

(DeWald et al.,

1990; Akunne et

PD-128,907 D2,3 agonist dopamine agonist al., 1995;

Pugsley et al.,

1995)

14 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther

5-HT1A,1D partial agonist,

5-HT2A,2B agonist, D2 pergolide (LY- (Millan et al., agonist, D3,4 partial dopamine agonist 127,809) 2002) agonist, a2A,2B,2C partial

agonist

AChE inhibitor, BuChE (Luo et al., physostigmine ChE inhibitor inhibitor 2006)

pimavanserin (ACP- 5-HT2A inverse (Vanover et al., 5-HT2A,2C inverse agonist 103) agonist 2006)

(Smith, 2001; PPARg > a agonist, pioglitazone PPARg agonist Paddock et al., mitoNEET ligand 2007)

a1A,2A,2C antagonist, D2,3

partial agonist, D4 (Millan et al., piribedil dopamine agonist antagonist, 5-HT1A partial 2002)

agonist

ether lipid plasmalogen plasmalogen (Wood et al., PPI-1011 precursor precursor 2011)

(Newman- D2S agonist, D2L,3,4 partial pramipexole dopamine agonist Tancredi et al., agonist 2002a)

(Ford et al., prazosin a1A,1B,1C antagonist a1 antagonist 1997)

15 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther preladenant (SCH- (Hodgson et al., A2A antagonist A2A antagonist 420,814) 2009)

(Rupprecht et

al., 1993;

Falkenstein et

al., 1998; Meyer

et al., 1998a;

Meyer et al., nPR agonist, mPR agonist, 1998b; Baulieu PGRMC1 agonist, s1 progesterone PR agonist and agonist, nACh NAM, MR Schumacher, antagonist 2000; Maurice

et al., 2001;

Attardi et al.,

2007;

Johannessen et

al., 2011)

propranolol b1,2 antagonist b antagonist (Baker, 2005)

H1 antagonist, D2 (Schotte et al., antagonist, 5-HT2A atypical anti- 1996; Arnt and quetiapine antagonist, 5-HT1A,1B psychotic Skarsfeldt, agonist, a1A,1B,2C 1998; Richelson antagonist

16 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther and Souder,

2000)

quinpirole (LY- (Millan et al., D2-like agonist D2 agonist 171,555) 2002)

R-(-)-11-OH-NPa (Neumeyer et (apomorphine D1,2 agonist D1,2 agonist al., 1988) derivative)

(Lejeune et al., (R)-(+)-OHDPAT 5-HT1A agonist 5-HT1A agonist 1997)

(Lyon et al.,

1986; Battaglia

et al., 1988; 5-HT2A partial R-MDMA 5-HT2A partial agonist Nash et al., agonist 1994; Verrico et

al., 2007; Huot

et al., 2011)

(Wichmann et raclopride D2,3 antagonist D2,3 antagonist al., 1999)

(Weinshank et rauwolscine (iso- al., 1992; Uhlen yohimbine, a- a2A,2B,2C antagonist, 5- a2 antagonist et al., 1994; yohimbine, HT1D,2D antagonist Wainscott et al., corynanthidine) 1998)

17 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Subramaniam

NMDA antagonist, Na+ et al., 1996; remacemide NMDA antagonist channel blocker Santangeli et al.,

2002)

RGFP-109 HDAC1,3 inhibitor HDAC inhibitor (Rai et al., 2010)

rimonabant (SR- cannabinoid (Seely et al., CB1 antagonist 141,716) antagonist 2012)

(Fischer et al., Ro-25,6981 NR2B antagonist NMDA antagonist 1997)

kynurenine 3- kynurenine 3-hydroxylase (Rover et al., Ro-61,8048 hydroxylase inhibitor 1997) inhibitor

(Wichmann et NOP receptor agonist, µ > Ro 65-6570 opioid ligand al., 1999; Toll et k > d ligand al., 2016)

(Eden et al.,

ropinirole (SKF- 1991; Millan et D2,3,4 agonist dopamine agonist 101,468-A) al., 2002; Millan

et al., 2004b)

D1,2,3,4,5 agonist, a1A,B (Beaulieu et al., rotigotine (N-0427, N- ligand, a2A,2B,2C ligand, 5- dopamine agonist 1984; Belluzzi 0923) HT1A partial agonist, 5- et al., 1994; HT7 ligand

18 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Scheller et al.,

2009)

(Millan et al.,

S-32,504 D2,3 agonist dopamine agonist 2004a; Millan et

al., 2004b)

(Cussac et al.,

S-33,084 D3 antagonist D3 antagonist 2000; Millan et

al., 2000a)

(Verrico et al., SERT > DAT inhibitor, dual monoamine S-MDMA 2007; Huot et monoamine releaser re-uptake inhibitor al., 2011)

NET > DAT inhibitor, dual monoamine (Damaj et al., S,S- a4b2 nACh antagonist re-uptake inhibitor 2004)

MAO-B inhibitor, (Caccia et al.,

safinamide glutamate release inhibitor, MAO-B inhibitor 2006; Fariello,

Na+ channel blocker 2007)

sarizotan (EMD- 5-HT1A agonist, D2,3,4 (Bartoszyk et 5-HT1A agonist 128,130) antagonist al., 2004)

5-HT1B inverse (Selkirk et al., SB-224,289-A 5-HT1B inverse agonist agonist 1998)

D1 antagonist, 5-HT2C (Millan et al., SCH-23,390 D1 antagonist agonist 2001)

19 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther scopolamine muscarinic (Huang et al., M1,2,3,4 antagonist (hyoscine) antagonist 2001)

(Owens et al., sertraline SERT inhibitor SERT inhibitor 2001)

HMG-CoA reductase HMG-CoA (Hartman et al., simvastatin inhibitor reductase inhibitor 1992)

(Andersen and

Jansen, 1990; SKF-38,393 D1,5 partial agonist D1 partial agonist Sunahara et al.,

1991)

(Andersen and

Jansen, 1990; SKF-75,670 D1 partial agonist D1 partial agonist Gnanalingham

et al., 1995)

(Gnanalingham

et al., 1995; SKF-80,723 D1 agonist D1 agonist Kanda et al.,

2000)

(Gnanalingham

et al., 1995; D1 agonist, oestrogen SKF-82,958 D1 agonist Domino and Ni, receptor-a agonist 1998; Walters et

al., 2002)

20 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther (Gnanalingham SKF-83,565 D1 partial agonist D1 partial agonist et al., 1995)

(Gnanalingham D1 agonist, D2 partial et al., 1995; agonist, s1 allosteric Rashid et al., SKF-83,959 ligand, DAT inhibitor, D1 agonist 2007; Fang et NET inhibitor, SERT al., 2013; Guo et inhibitor al., 2013)

5-HT1B,1D > 5-HT1A (Hatcher et al., SKF-99,101-H 5-HT1B,1D agonist agonist 1995)

(Bilsky et al.,

SNC-80 d agonist d agonist 1995; Metcalf et

al., 2012)

(Bezard et al., ST-198 D3 antagonist D3 antagonist 2003)

(Stasi et al., ST-1535 A2A > A1 antagonist A2A antagonist 2006)

(Maitre et al., D2,3 antagonist, carbonic 1994; Burris et sulpiride anhydrase inhibitor, GHB D2,3 antagonist al., 1995; Sethi ligand et al., 2013)

sumanirole (PNU- (McCall et al., D2 agonist D2 agonist 95,666) 2005)

21 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther talampanel (GYKI- (Wang and Niu, GluA2 antagonist AMPA antagonist 537,773, LY-300,164) 2013)

oestrogen receptor a and b oestrogen receptor (Kuiper et al., tamoxifen modulator modulator 1997)

(Johnston et al.,

TC-8831 b2 nACh agonist nicotinic agonist 2013; Quik et

al., 2013)

a1A,2A,2B,2C antagonist, D2,3

partial agonist, D4 terguride (trans- (Millan et al., antagonist, 5-HT1A,1D,2A D2 partial agonist dihydro-lisuride) 2002) partial agonist, 5-HT2B

antagonist

Na+ channel blocker,

GABAA enhancer, AMPA topiramate AMPA antagonist (Perucca, 1997) antagonist, carbonic

anhydrase inhibitor

muscarinic (Giachetti et al., trihexyphenidyl M1 antagonist antagonist 1986)

(Von

U50-488 k agonist opioid agonist Voigtlander and

Lewis, 1982)

(Black et al., U-91,356-A D2 agonist D2 agonist 1997)

22 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther U-99,194-A (PNU- (Audinot et al., D3 antagonist D3 antagonist 99,194-A) 1998)

(Mor et al., URB-597 (KDS-4103) FAAH inhibitor FAAH inhibitor 2004)

UWA-101 (a- dual monoamine (Johnston et al., DAT = SERT inhibitor cyclopropyl-MDMA) re-uptake inhibitor 2012)

dual monoamine (Huot et al., UWA-121 DAT > SERT inhibitor re-uptake inhibitor 2014)

(Huot et al., UWA-122 SERT inhibitor SERT inhibitor 2014)

non-selective nACh (Mihalak et al., varenicline nicotinic agonist agonist/partial agonist 2006)

(Shen et al., VU-0,476,406 M4 PAM muscarinic PAM 2015)

(Bylund et al.,

1992; Adham et

al., 1993;

a2A,2B,2C antagonist, 5- Devedjian et al., yohimbine a2 antagonist HT1A,1B,1D,1F,2B antagonist 1994; Bonhaus

et al., 1999;

Millan et al.,

2000b)

23 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther 5-HT: 5-hydroxytryptamine (serotonin); a: alpha adrenoceptor (except when followed by nACh, where they correspond to a sub-unit); b: beta adrenoceptor (except when followed by nACh, where they correspond to a sub-unit); d: delta opioid receptor; k opioid receptor; µ opiod receptor; s: sigma receptor; A: adenosine receptor; AChE: acetylcholinesterase; AMPA: α- amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; BuChE: butyrylcholinesterase;

CB: cannabinoid receptor; ChE: ; COMT: catechol-O-methyltransferase; D: dopamine receptor; DAT: ; FAAH: fatty acid amide hydrolase; FFA: free fatty acid receptor; GABA: gamma-aminobutyric acid; GHB: gamma-hydroxybutyric acid;

GluA: AMPA receptor sub-unit; H: ; HDAC: histone deacetylase; HMG-CoA:

3-hydroxy-3-methylglutaryl-coenzyme A; I: imidazoline receptor; M: muscarinic receptor;

MAO-B: ; MDMA: 3,4-methylenedioxymethamphetamine; mGlu: metabotropic glutamate receptor; MPR: membrane progesterone receptor; MR: mineralocorticoid receptor; Na+: sodium; nACh: nicotinic acetylcholine receptor; NAM: negative allosteric modulator; NET: noradrenaline transporter; NMDA: N-methyl-D-aspartate; NOP: nociceptin receptor; NPR: natriuretic peptide receptor; NR: NMDA receptor sub-unit; ORL: opioid receptor- like; PAM: positive allosteric modulator; PGRMC1: progesterone receptor membrane component

1; PPAR: peroxisome proliferator-activated receptor; PR: progesterone receptor; RXR: retinoid X receptor; SERT: serotonin transporter; SV: synaptic vesicle glycoprotein; TRPA: transient receptor potential channel ankyrin.

24 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Table S2: comparison of the anti-dyskinetic action of drugs across species

marmose drug human macaque squirrel monkey t

(+)-4-propyl-9-

hydroxynaphth ne na ne na

oxazine

ABT-431 ne ne ne na

AFQ-056 Ö/ne Ö na na

amantadine Ö Ö Ö na

apomorphine ne ne ne na

AQW051 ne Ö na na

atropine ne ne ne na

benztropine ne na ne na

brasofensine ne na ne na

bromocriptine ne ne ne na

bupropion ne na ne na

cabergoline ne ne na na

citalopram Ö Ö na na

clonidine na Ö na na

clozapine Ö Ö Ö na

CP-101,606 Ö ne ne na

25 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther CY-208,243 ne ne ne na

diazepam Ö Ö na na

dipraglurant Ö Ö na na

eltoprazine Ö Ö na na

entacapone de ne ne na

ethosuximide ne ne na na

famotidine ne Ö na na

fipamezole Ö Ö Ö na

haloperidol Ö na Ö na

idazoxan Ö/ne Ö Ö na

istradefylline ne ne ne na

L-tryptophan na Ö na ne

levetiracetam Ö/ne Ö Ö na

melanocyte- ne na ne na inhibiting factor

methysergide ne Ö na na

mianserin na na Ö na

mirtazapine Ö na Ö na

modafinil ne na na na

morphine Ö Ö na na

nabilone Ö na Ö na

naloxone Ö/ne Ö na na

26 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther naltrexone Ö/ne ne/de Ö na

nicotine na na na Ö

nomifensine de na ne na

pardoprunox ne na Ö na

pergolide ne na ne na

physostigmine Ö/ne Ö ne na

pimavanserin ne Ö na na

pioglitazone ne Ö na na

piribedil ne na ne na

pramipexole ne na ne na

preladenant de ne na na

propranolol Ö Ö na na

quetiapine Ö/ne Ö Ö na

remacemide ne ne na na

ropinirole ne na ne na

rotigotine ne ne ne na

safinamide Ö Ö na na

sarizotan ne Ö na na

scopolamine ne na ne na

sertraline ne na na na

simvastatin ne Ö na na

sulpiride Ö Ö ne na

27 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther sumanirole ne ne na na

terguride ne ne ne na

topiramate de/ne na Ö na

trihexyphenidyl ne ne ne na

yohimbine na Ö Ö na

Ö: effective; ne: not effective; de: deleterious; na: not assessed

28 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Table S3: comparison of the anti-parkinsonian action of drugs across species

marmose drug human macaque squirrel monkey t

(+)-4-propyl-9-

hydroxynaphth Ö na Ö na

oxazine

ABT-431 Ö Ö Ö na

AFQ-056 ne Ö na na

amantadine Ö/ne ne/de ne na

apomorphine Ö Ö Ö na

AQW-051 ne Ö na na

atropine Ö Ö Ö na

benztropine Ö na Ö na

brasofensine ne na Ö na

bromocriptine Ö Ö Ö na

bupropion Ö na Ö na

cabergoline Ö Ö na na

citalopram Ö/ne de na na

clonidine Ö Ö/de na na

clozapine Ö ne ne na

CP-101,606 ne Ö Ö na

29 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther CY-208,243 Ö Ö Ö na

diazepam Ö ne na na

dipraglurant ne ne na na

eltoprazine ne de na na

entacapone Ö Ö Ö na

ethosuximide ne/de Ö na na

famotidine ne Ö na na

fipamezole ne Ö Ö na

haloperidol de na de na

idazoxan ne Ö Ö na

istradefylline Ö Ö Ö na

L-tryptophan ne de na na

levetiracetam ne ne ne na

melanocyte- ne na ne na inhibiting factor

methysergide ne de na na

mianserin na na de na

mirtazapine Ö na ne na

modafinil ne na Ö na

morphine de Ö na na

nabilone ne na ne na

naloxone Ö/ne ne na na

30 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther naltrexone ne ne ne na

nicotine Ö/ne na na ne

nomifensine Ö na Ö na

pardoprunox Ö na Ö na

pergolide Ö na Ö na

physostigmine de de de na

pimavanserin ne ne na na

pioglitazone ne de na na

piribedil Ö na Ö na

pramipexole Ö na Ö na

preladenant Ö Ö na na

propranolol Ö/ne de na na

quetiapine ne/de ne ne na

remacemide ne Ö na na

ropinirole Ö na Ö na

rotigotine Ö Ö Ö na

safinamide Ö Ö na na

sarizotan de de na na

scopolamine Ö na Ö na

sertraline ne na de na

simvastatin ne ne na na

sulpiride de de de na

31 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther sumanirole Ö Ö na na

terguride Ö Ö Ö na

topiramate ne na ne na

trihexyphenidyl Ö Ö Ö na

yohimbine Ö ne ne na

Ö: effective; ne: not effective; de: deleterious; na: not assessed

32 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Supplementary references

Abraham G, Solyom S, Csuzdi E, Berzsenyi P, Ling I, Tarnawa I, Hamori T, Pallagi I, Horvath K,

Andrasi F, Kapus G, Harsing LG, Jr., Kiraly I, Patthy M and Horvath G (2000) New non

competitive AMPA antagonists. Bioorg Med Chem 8:2127-2143.

Adham N, Kao HT, Schecter LE, Bard J, Olsen M, Urquhart D, Durkin M, Hartig PR, Weinshank

RL and Branchek TA (1993) Cloning of another human serotonin receptor (5-HT1F): a

fifth 5-HT1 receptor subtype coupled to the inhibition of adenylate cyclase. Proc Natl Acad

Sci U S A 90:408-412.

Akunne HC, Towers P, Ellis GJ, Dijkstra D, Wikstrom H, Heffner TG, Wise LD and Pugsley TA

(1995) Characterization of binding of [3H]PD 128907, a selective dopamine D3 receptor

agonist ligand, to CHO-K1 cells. Life Sci 57:1401-1410.

Amador M and Dani JA (1991) MK-801 inhibition of nicotinic acetylcholine receptor channels.

Synapse 7:207-215.

Ametamey SM, Kokic M, Carrey-Remy N, Blauenstein P, Willmann M, Bischoff S, Schmutz M,

Schubiger PA and Auberson YP (2000) Synthesis, radiolabelling and biological

characterization of (D)-7-iodo-N-(1-phosphonoethyl)-5-aminomethylquinoxaline-2,3-

dione, a glycine-binding site antagonist of NMDA receptors. Bioorg Med Chem Lett 10:75-

78.

Andersen PH (1989) The dopamine inhibitor GBR 12909: selectivity and molecular mechanism of

action. Eur J Pharmacol 166:493-504.

Andersen PH, Gronvald FC, Hohlweg R, Hansen LB, Guddal E, Braestrup C and Nielsen EB

(1992) NNC-112, NNC-687 and NNC-756, new selective and highly potent dopamine D1

receptor antagonists. Eur J Pharmacol 219:45-52.

33 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Andersen PH and Jansen JA (1990) Dopamine receptor agonists: selectivity and dopamine D1

receptor efficacy. Eur J Pharmacol 188:335-347.

Anttila SA and Leinonen EV (2001) A review of the pharmacological and clinical profile of

mirtazapine. CNS Drug Rev 7:249-264.

Arnt J, Bogeso KP, Christensen AV, Hyttel J, Larsen JJ and Svendsen O (1983) Dopamine receptor

agonistic and antagonistic effects of 3-PPP enantiomers. Psychopharmacology (Berl)

81:199-207.

Arnt J and Skarsfeldt T (1998) Do novel have similar pharmacological

characteristics? A review of the evidence. Neuropsychopharmacology 18:63-101.

Attardi BJ, Zeleznik A, Simhan H, Chiao JP, Mattison DR, Caritis SN and Obstetric-Fetal

Pharmacology Research Unit N (2007) Comparison of progesterone and glucocorticoid

receptor binding and stimulation of gene expression by progesterone, 17-alpha

hydroxyprogesterone caproate, and related progestins. Am J Obstet Gynecol 197:599 e591-

597.

Audinot V, Newman-Tancredi A, Gobert A, Rivet JM, Brocco M, Lejeune F, Gluck L, Desposte

I, Bervoets K, Dekeyne A and Millan MJ (1998) A comparative in vitro and in vivo

pharmacological characterization of the novel dopamine D3 receptor antagonists (+)-S

14297, nafadotride, GR 103,691 and U 99194. J Pharmacol Exp Ther 287:187-197.

Baker JG (2005) The selectivity of beta-adrenoceptor antagonists at the human beta1, beta2 and

beta3 adrenoceptors. Br J Pharmacol 144:317-322.

Barton ME and White HS (2004) The effect of CGX-1007 and CI-1041, novel NMDA receptor

antagonists, on kindling acquisition and expression. Epilepsy Res 59:1-12.

Barton ME, White HS and Wilcox KS (2004) The effect of CGX-1007 and CI-1041, novel NMDA

receptor antagonists, on NMDA receptor-mediated EPSCs. Epilepsy Res 59:13-24.

34 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Bartoszyk GD, Van Amsterdam C, Greiner HE, Rautenberg W, Russ H and Seyfried CA (2004)

Sarizotan, a serotonin 5-HT1A receptor agonist and dopamine receptor ligand. 1.

Neurochemical profile. J Neural Transm 111:113-126.

Battaglia G, Brooks BP, Kulsakdinun C and De Souza EB (1988) Pharmacologic profile of MDMA

(3,4-methylenedioxymethamphetamine) at various brain recognition sites. Eur J

Pharmacol 149:159-163.

Baulieu E and Schumacher M (2000) Progesterone as a neuroactive neurosteroid, with special

reference to the effect of progesterone on myelination. Steroids 65:605-612.

Beaulieu M, Itoh Y, Tepper P, Horn AS and Kebabian JW (1984) N,N-disubstituted 2-

aminotetralins are potent D-2 dopamine receptor agonists. Eur J Pharmacol 105:15-21.

Belluzzi JD, Domino EF, May JM, Bankiewicz KS and McAfee DA (1994) N-0923, a selective

dopamine D2 receptor agonist, is efficacious in rat and monkey models of Parkinson's

disease. Mov Disord 9:147-154.

Ben-Daniel R, Deuther-Conrad W, Scheunemann M, Steinbach J, Brust P and Mishani E (2008)

Carbon-11 labeled indolylpropylamine analog as a new potential PET agent for imaging of

the serotonin transporter. Bioorg Med Chem 16:6364-6370.

Benowitz NL (2009) Pharmacology of nicotine: addiction, smoking-induced disease, and

therapeutics. Annu Rev Pharmacol Toxicol 49:57-71.

Bergeron R, de Montigny C and Debonnel G (1996) Potentiation of neuronal NMDA response

induced by dehydroepiandrosterone and its suppression by progesterone: effects mediated

via sigma receptors. J Neurosci 16:1193-1202.

Bezard E, Ferry S, Mach U, Stark H, Leriche L, Boraud T, Gross C and Sokoloff P (2003)

Attenuation of levodopa-induced dyskinesia by normalizing dopamine D3 receptor

function. Nat Med 9:762-767.

35 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Bilsky EJ, Calderon SN, Wang T, Bernstein RN, Davis P, Hruby VJ, McNutt RW, Rothman RB,

Rice KC and Porreca F (1995) SNC 80, a selective, nonpeptidic and systemically active

opioid delta agonist. J Pharmacol Exp Ther 273:359-366.

Blaazer AR, Lange JH, van der Neut MA, Mulder A, den Boon FS, Werkman TR, Kruse CG and

Wadman WJ (2011) Novel indole and azaindole (pyrrolopyridine) cannabinoid (CB)

receptor agonists: design, synthesis, structure-activity relationships, physicochemical

properties and biological activity. Eur J Med Chem 46:5086-5098.

Black KJ, Gado MH and Perlmutter JS (1997) PET measurement of dopamine D2 receptor-

mediated changes in striatopallidal function. J Neurosci 17:3168-3177.

Blanchet PJ, Konitsiotis S, Whittemore ER, Zhou ZL, Woodward RM and Chase TN (1999)

Differing effects of N-methyl-D-aspartate receptor subtype selective antagonists on

dyskinesias in levodopa-treated 1-methyl-4-phenyl-tetrahydropyridine monkeys. J

Pharmacol Exp Ther 290:1034-1040.

Blanpied TA, Clarke RJ and Johnson JW (2005) Amantadine inhibits NMDA receptors by

accelerating channel closure during channel block. J Neurosci 25:3312-3322.

Bonhaus DW, Flippin LA, Greenhouse RJ, Jaime S, Rocha C, Dawson M, Van Natta K, Chang

LK, Pulido-Rios T, Webber A, Leung E, Eglen RM and Martin GR (1999) RS-127445: a

selective, high affinity, orally bioavailable 5-HT2B receptor antagonist. Br J Pharmacol

127:1075-1082.

Bonhaus DW, Weinhardt KK, Taylor M, DeSouza A, McNeeley PM, Szczepanski K, Fontana DJ,

Trinh J, Rocha CL, Dawson MW, Flippin LA and Eglen RM (1997) RS-102221: a novel

high affinity and selective, 5-HT2C receptor antagonist. Neuropharmacology 36:621-629.

36 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Booth RG, Moniri NH, Bakker RA, Choksi NY, Nix WB, Timmerman H and Leurs R (2002) A

novel phenylaminotetralin radioligand reveals a subpopulation of histamine H(1) receptors.

J Pharmacol Exp Ther 302:328-336.

Brimecombe JC, Boeckman FA and Aizenman E (1997) Functional consequences of NR2 subunit

composition in single recombinant N-methyl-D-aspartate receptors. Proc Natl Acad Sci U

S A 94:11019-11024.

Burris KD, Pacheco MA, Filtz TM, Kung MP, Kung HF and Molinoff PB (1995) Lack of

discrimination by agonists for D2 and D3 dopamine receptors. Neuropsychopharmacology

12:335-345.

Burstein ES, Carlsson ML, Owens M, Ma JN, Schiffer HH, Carlsson A and Hacksell U (2011) II.

In vitro evidence that (-)-OSU6162 and (+)-OSU6162 produce their behavioral effects

through 5-HT2A serotonin and D2 dopamine receptors. J Neural Transm (Vienna)

118:1523-1533.

Bylund DB, Blaxall HS, Iversen LJ, Caron MG, Lefkowitz RJ and Lomasney JW (1992)

Pharmacological characteristics of alpha 2-adrenergic receptors: comparison of

pharmacologically defined subtypes with subtypes identified by molecular cloning. Mol

Pharmacol 42:1-5.

Caccia C, Maj R, Calabresi M, Maestroni S, Faravelli L, Curatolo L, Salvati P and Fariello RG

(2006) Safinamide: from molecular targets to a new anti-Parkinson drug. Neurology

67:S18-23.

Cao X, Liang L, Hadcock JR, Iredale PA, Griffith DA, Menniti FS, Factor S, Greenamyre JT and

Papa SM (2007) Blockade of cannabinoid type 1 receptors augments the antiparkinsonian

action of levodopa without affecting dyskinesias in 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine-treated rhesus monkeys. J Pharmacol Exp Ther 323:318-326.

37 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Carlsson ML, Burstein ES, Kloberg A, Hansson S, Schedwin A, Nilsson M, Rung JP and Carlsson

A (2011) I. In vivo evidence for partial agonist effects of (-)-OSU6162 and (+)-OSU6162

on 5-HT2A serotonin receptors. J Neural Transm (Vienna) 118:1511-1522.

Carroll FI, Blough BE, Abraham P, Mills AC, Holleman JA, Wolckenhauer SA, Decker AM,

Landavazo A, McElroy KT, Navarro HA, Gatch MB and Forster MJ (2009) Synthesis and

biological evaluation of bupropion analogues as potential pharmacotherapies for

addiction. J Med Chem 52:6768-6781.

Chae E, Shin YJ, Ryu EJ, Ji MK, Ryune Cho N, Lee KH, Jeong HJ, Kim SJ, Choi Y, Seok Oh K,

Park CE and Soo Yoon Y (2013) Discovery of biological evaluation of pyrazole/imidazole

amides as mGlu5 receptor negative allosteric modulators. Bioorg Med Chem Lett 23:2134-

2139.

Cheetham S, Butler S, Hearson M, Kerrigan F, Martin K, Needham P, Prow M, Sargent B, Slater

N and Skill M (1998) BTS 74 398: a novel monoamine reuptake inhibitor for the treatment

of Parkinson's disease. Br J Pharmacol 123 Suppl 1:224P.

Chen F, Knecht K, Birzin E, Fisher J, Wilkinson H, Mojena M, Moreno CT, Schmidt A, Harada

S, Freedman LP and Reszka AA (2005) Direct agonist/antagonist functions of

dehydroepiandrosterone. Endocrinology 146:4568-4576.

Chenard BL, Bordner J, Butler TW, Chambers LK, Collins MA, De Costa DL, Ducat MF, Dumont

ML, Fox CB, Mena EE and et al. (1995) (1S,2S)-1-(4-hydroxyphenyl)-2-(4-hydroxy-4-

phenylpiperidino)-1-propanol: a potent new neuroprotectant which blocks N-methyl-D-

aspartate responses. J Med Chem 38:3138-3145.

Chremos AN (1987) Clinical pharmacology of famotidine: a summary. J Clin Gastroenterol 9

Suppl 2:7-12.

38 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Close SP, Elliott PJ, Hayes AG and Marriott AS (1990) Effects of classical and novel agents in a

MPTP-induced reversible model of Parkinson's disease. Psychopharmacology (Berl)

102:295-300.

Cobos EJ, del Pozo E and Baeyens JM (2007) Irreversible blockade of sigma-1 receptors by

haloperidol and its metabolites in guinea pig brain and SH-SY5Y human neuroblastoma

cells. J Neurochem 102:812-825.

Cosford ND, Tehrani L, Roppe J, Schweiger E, Smith ND, Anderson J, Bristow L, Brodkin J, Jiang

X, McDonald I, Rao S, Washburn M and Varney MA (2003) 3-[(2-Methyl-1,3-thiazol-4-

yl)ethynyl]-pyridine: a potent and highly selective metabotropic glutamate subtype 5

receptor antagonist with anxiolytic activity. J Med Chem 46:204-206.

Cussac D, Newman-Tancredi A, Sezgin L and Millan MJ (2000) The novel antagonist, S33084,

and GR218,231 interact selectively with cloned and native, rat dopamine D(3) receptors as

compared with native, rat dopamine D(2) receptors. Eur J Pharmacol 394:47-50.

Damaj MI, Carroll FI, Eaton JB, Navarro HA, Blough BE, Mirza S, Lukas RJ and Martin BR

(2004) Enantioselective effects of hydroxy metabolites of bupropion on behavior and on

function of monoamine transporters and nicotinic receptors. Mol Pharmacol 66:675-682. de Boer T (1996) The pharmacologic profile of mirtazapine. J Clin Psychiatry 57 Suppl 4:19-25. de Boer TH, Maura G, Raiteri M, de Vos CJ, Wieringa J and Pinder RM (1988) Neurochemical

and autonomic pharmacological profiles of the 6-aza-analogue of mianserin, Org 3770 and

its enantiomers. Neuropharmacology 27:399-408.

Devedjian JC, Esclapez F, Denis-Pouxviel C and Paris H (1994) Further characterization of human

alpha 2-adrenoceptor subtypes: [3H]RX821002 binding and definition of additional

selective drugs. Eur J Pharmacol 252:43-49.

39 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther DeWald HA, Heffner TG, Jaen JC, Lustgarten DM, McPhail AT, Meltzer LT, Pugsley TA and

Wise LD (1990) Synthesis and dopamine agonist properties of (+-)-trans-3,4,4a,10b-

tetrahydro-4-propyl-2H,5H-[1]benzopyrano [4,3-b]-1,4-oxazin-9-ol and its enantiomers. J

Med Chem 33:445-450.

Domino EF and Ni L (1998) Trihexyphenidyl interactions with the dopamine D1-selective receptor

agonist SKF-82958 and the D2-selective receptor agonist N-0923 in 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine-induced hemiparkinsonian monkeys. J Pharmacol Exp Ther

284:307-311.

Dowling MR and Charlton SJ (2006) Quantifying the association and dissociation rates of

unlabelled antagonists at the muscarinic M3 receptor. Br J Pharmacol 148:927-937.

Doxey JC, Lane AC, Roach AG and Virdee NK (1984) Comparison of the alpha-adrenoceptor

antagonist profiles of idazoxan (RX 781094), yohimbine, rauwolscine and corynanthine.

Naunyn Schmiedebergs Arch Pharmacol 325:136-144.

Eden RJ, Costall B, Domeney AM, Gerrard PA, Harvey CA, Kelly ME, Naylor RJ, Owen DA and

Wright A (1991) Preclinical pharmacology of ropinirole (SK&F 101468-A) a novel

dopamine D2 agonist. Pharmacol Biochem Behav 38:147-154.

Ehrlich PP, Ralston JW and Michaelides MR (1997) An Efficient Enantioselective Synthesis of

the D1 Agonist (5aR,11bS)-4,5,5a,6,7,11b-Hexahydro-2-propyl-3-thia- 5-

azacyclopenta[c]phenanthrene-9,10-diol (A-86929). J Org Chem 62:2782-2785.

Emmerson PJ, Liu MR, Woods JH and Medzihradsky F (1994) Binding affinity and selectivity of

at mu, delta and kappa receptors in monkey brain membranes. J Pharmacol Exp

Ther 271:1630-1637.

Ernsberger P, Meeley MP, Mann JJ and Reis DJ (1987) Clonidine binds to imidazole binding sites

as well as alpha 2-adrenoceptors in the ventrolateral medulla. Eur J Pharmacol 134:1-13.

40 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Falkenstein E, Schmieding K, Lange A, Meyer C, Gerdes D, Welsch U and Wehling M (1998)

Localization of a putative progesterone membrane binding protein in porcine hepatocytes.

Cell Mol Biol (Noisy-le-grand) 44:571-578.

Fang X, Guo L, Jia J, Jin GZ, Zhao B, Zheng YY, Li JQ, Zhang A and Zhen XC (2013) SKF83959

is a novel triple reuptake inhibitor that elicits anti-depressant activity. Acta Pharmacol Sin

34:1149-1155.

Fariello RG (2007) Safinamide. Neurotherapeutics 4:110-116.

Felder CC, Joyce KE, Briley EM, Mansouri J, Mackie K, Blond O, Lai Y, Ma AL and Mitchell RL

(1995) Comparison of the pharmacology and signal transduction of the human cannabinoid

CB1 and CB2 receptors. Mol Pharmacol 48:443-450.

Feng B, Tse HW, Skifter DA, Morley R, Jane DE and Monaghan DT (2004) Structure-activity

analysis of a novel NR2C/NR2D-preferring NMDA receptor antagonist: 1-(phenanthrene-

2-carbonyl) piperazine-2,3-dicarboxylic acid. Br J Pharmacol 141:508-516.

Fernandez J, Alonso JM, Andres JI, Cid JM, Diaz A, Iturrino L, Gil P, Megens A, Sipido VK and

Trabanco AA (2005) Discovery of new tetracyclic tetrahydrofuran derivatives as potential

broad-spectrum psychotropic agents. J Med Chem 48:1709-1712.

Fernstrom JD (1983) Role of precursor availability in control of monoamine biosynthesis in brain.

Physiol Rev 63:484-546.

Feuerbach D, Pezous N, Weiss M, Shakeri-Nejad K, Lingenhoehl K, Hoyer D, Hurth K, Bilbe G,

Pryce CR, McAllister K, Chaperon F, Kucher K, Johns D, Blaettler T and Lopez Lopez C

(2015) AQW051, a novel, potent and selective alpha7 nicotinic ACh receptor partial

agonist: pharmacological characterization and phase I evaluation. Br J Pharmacol

172:1292-1304.

41 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Fischer G, Mutel V, Trube G, Malherbe P, Kew JN, Mohacsi E, Heitz MP and Kemp JA (1997)

Ro 25-6981, a highly potent and selective blocker of N-methyl-D-aspartate receptors

containing the NR2B subunit. Characterization in vitro. J Pharmacol Exp Ther 283:1285-

1292.

Ford AP, Daniels DV, Chang DJ, Gever JR, Jasper JR, Lesnick JD and Clarke DE (1997)

Pharmacological pleiotropism of the human recombinant alpha1A-adrenoceptor:

implications for alpha1-adrenoceptor classification. Br J Pharmacol 121:1127-1135.

Fredholm BB, AP IJ, Jacobson KA, Linden J and Muller CE (2011) International Union of Basic

and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine

receptors--an update. Pharmacol Rev 63:1-34.

Fruchart-Gaillard C, Mourier G, Marquer C, Menez A and Servent D (2006) Identification of

various allosteric interaction sites on M1 muscarinic receptor using 125I-Met35-oxidized

. Mol Pharmacol 69:1641-1651.

Garbarg M, Arrang JM, Rouleau A, Ligneau X, Tuong MD, Schwartz JC and Ganellin CR (1992)

S-[2-(4-imidazolyl)ethyl]isothiourea, a highly specific and potent

agonist. J Pharmacol Exp Ther 263:304-310.

Gasparini F, Lingenhohl K, Stoehr N, Flor PJ, Heinrich M, Vranesic I, Biollaz M, Allgeier H,

Heckendorn R, Urwyler S, Varney MA, Johnson EC, Hess SD, Rao SP, Sacaan AI, Santori

EM, Velicelebi G and Kuhn R (1999) 2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a

potent, selective and systemically active mGlu5 receptor antagonist. Neuropharmacology

38:1493-1503.

Giachetti A, Giraldo E, Ladinsky H and Montagna E (1986) Binding and functional profiles of the

selective M1 muscarinic receptor antagonists trihexyphenidyl and dicyclomine. Br J

Pharmacol 89:83-90.

42 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Gill R, A, Bourson A, Buttelmann B, Fischer G, Heitz MP, Kew JN, Levet-Trafit B, Lorez

HP, Malherbe P, Miss MT, Mutel V, Pinard E, Roever S, Schmitt M, Trube G, Wybrecht

R, Wyler R and Kemp JA (2002) Pharmacological characterization of Ro 63-1908 (1-[2-

(4-hydroxy-phenoxy)-ethyl]-4-(4-methyl-benzyl)-piperidin-4-ol), a novel subtype-

selective N-methyl-D-aspartate antagonist. J Pharmacol Exp Ther 302:940-948.

Glennon JC, Van Scharrenburg G, Ronken E, Hesselink MB, Reinders JH, Van Der Neut M, Long

SK, Feenstra RW and McCreary AC (2006) In vitro characterization of SLV308 (7-[4-

methyl-1-piperazinyl]-2(3H)-benzoxazolone, monohydrochloride): a novel partial

dopamine D2 and D3 receptor agonist and serotonin 5-HT1A receptor agonist. Synapse

60:599-608.

Gnanalingham KK, Erol DD, Hunter AJ, Smith LA, Jenner P and Marsden CD (1995) Differential

anti-parkinsonian effects of benzazepine D1 dopamine agonists with varying efficacies in

the MPTP-treated common marmoset. Psychopharmacology (Berl) 117:275-286.

Gomora JC, Daud AN, Weiergraber M and Perez-Reyes E (2001) Block of cloned human T-type

calcium channels by succinimide antiepileptic drugs. Mol Pharmacol 60:1121-1132.

Gower AJ and Marriott AS (1982) Pharmacological evidence for the subclassification of central

dopamine receptors in the rat. Br J Pharmacol 77:185-193.

Guo L, Zhao J, Jin G, Zhao B, Wang G, Zhang A and Zhen X (2013) SKF83959 is a potent

allosteric modulator of sigma-1 receptor. Mol Pharmacol 83:577-586.

Haig G, Wang D, Othman AA and Zhao J (2016) The alpha7 Nicotinic Agonist ABT-126 in the

Treatment of Cognitive Impairment Associated with Schizophrenia in Nonsmokers: Results

from a Randomized Controlled Phase 2b Study. Neuropsychopharmacology 41:2893-2902.

Hartman GD, Halczenko W, Duggan ME, Imagire JS, Smith RL, Pitzenberger SM, Fitzpatrick SL,

Alberts AW, Bostedor R, Chao YS and et al. (1992) 3-Hydroxy-3-methylglutaryl-

43 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther coenzyme A reductase inhibitors. 9. The synthesis and biological evaluation of novel

simvastatin analogs. J Med Chem 35:3813-3821.

Hatcher JP, Slade PD, Roberts C and Hagan JJ (1995) 5-HT 1D receptors mediate SKF 99101H-

induced hypothermia in the guinea pig. J Psychopharmacol 9:234-241.

Heinrich JN, Brennan J, Lai MH, Sullivan K, Hornby G, Popiolek M, Jiang LX, Pausch MH, Stack

G, Marquis KL and Andree TH (2006) Aplindore (DAB-452), a high affinity selective

dopamine D2 receptor partial agonist. Eur J Pharmacol 552:36-45.

Herman BH, Vocci F and Bridge P (1995) The effects of NMDA receptor antagonists and nitric

oxide synthase inhibitors on opioid tolerance and withdrawal. Medication development

issues for opiate addiction. Neuropsychopharmacology 13:269-293.

Hirose H, Aoki I, Kimura T, Fujikawa T, Numazawa T, Sasaki K, Sato A, Hasegawa T, Nishikibe

M, Mitsuya M, Ohtake N, Mase T and Noguchi K (2001) Pharmacological properties of

(2R)-N-[1-(6-aminopyridin-2-ylmethyl)piperidin-4-yl]-2-[(1R)-3,3-difluorocyclopen tyl]-

2-hydroxy-2-phenylacetamide: a novel mucarinic antagonist with M(2)-sparing

antagonistic activity. J Pharmacol Exp Ther 297:790-797.

Hirst WD, Babbs AJ, Green A, Minton JA, Shaw TE, Wise A, Rice SQ, Pangalos MN and Price

GW (2003) Pharmacological characterisation of a cell line expressing GABA B1b and

GABA B2 receptor subunits. Biochem Pharmacol 65:1103-1113.

Hodgson RA, Bertorelli R, Varty GB, Lachowicz JE, Forlani A, Fredduzzi S, Cohen-Williams ME,

Higgins GA, Impagnatiello F, Nicolussi E, Parra LE, Foster C, Zhai Y, Neustadt BR,

Stamford AW, Parker EM, Reggiani A and Hunter JC (2009) Characterization of the potent

and highly selective A2A receptor antagonists preladenant and SCH 412348 [7-[2-[4-2,4-

difluorophenyl]-1-piperazinyl]ethyl]-2-(2-furanyl)-7H-pyrazolo[4,3-e ][1,2,4]triazolo[1,5-

44 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther c]pyrimidin-5-amine] in rodent models of movement disorders and depression. J

Pharmacol Exp Ther 330:294-303.

Hoyer D, Clarke DE, Fozard JR, Hartig PR, Martin GR, Mylecharane EJ, Saxena PR and

Humphrey PP (1994) International Union of Pharmacology classification of receptors for

5-hydroxytryptamine (Serotonin). Pharmacol Rev 46:157-203.

Huang F, Buchwald P, Browne CE, Farag HH, Wu WM, Ji F, Hochhaus G and Bodor N (2001)

Receptor binding studies of soft anticholinergic agents. AAPS PharmSci 3:E30.

Huot P, Johnston TH, Lewis KD, Koprich JB, Reyes MG, Fox SH, Piggott MJ and Brotchie JM

(2011) Characterization of 3,4-Methylenedioxymethamphetamine (MDMA) Enantiomers

In Vitro and in the MPTP-Lesioned Primate: R-MDMA Reduces Severity of Dyskinesia,

Whereas S-MDMA Extends Duration of ON-Time. J Neurosci 31:7190-7198.

Huot P, Johnston TH, Lewis KD, Koprich JB, Reyes MG, Fox SH, Piggott MJ and Brotchie JM

(2014) UWA-121, a mixed dopamine and serotonin re-uptake inhibitor, enhances L-DOPA

anti-parkinsonian action without worsening dyskinesia or psychosis-like behaviours in the

MPTP-lesioned common marmoset. Neuropharmacology 82:76-87.

Hyttel J (1982) Citalopram--pharmacological profile of a specific serotonin uptake inhibitor with

antidepressant activity. Prog Neuropsychopharmacol Biol Psychiatry 6:277-295.

Hyttel J (1994) Pharmacological characterization of selective serotonin reuptake inhibitors

(SSRIs). Int Clin Psychopharmacol 9 Suppl 1:19-26.

Itoh Y, Kawamata Y, Harada M, Kobayashi M, Fujii R, Fukusumi S, Ogi K, Hosoya M, Tanaka

Y, Uejima H, Tanaka H, Maruyama M, Satoh R, Okubo S, Kizawa H, Komatsu H,

Matsumura F, Noguchi Y, Shinohara T, Hinuma S, Fujisawa Y and Fujino M (2003) Free

fatty acids regulate insulin secretion from pancreatic beta cells through GPR40. Nature

422:173-176.

45 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Iyengar S, Wood PL, Mick SJ, Dilworth VM, Gray NM, Farah JM, Rao TS and Contreras PC

(1991) (+) 3-[3-hydroxyphenyl-N-(1-propyl) piperidine] selectively differentiates effects

of sigma ligands on neurochemical pathways modulated by sigma receptors: evidence for

subtypes, in vivo. Neuropharmacology 30:915-922.

Jakubik J, Bacakova L, El-Fakahany EE and Tucek S (1997) Positive cooperativity of acetylcholine

and other agonists with allosteric ligands on muscarinic acetylcholine receptors. Mol

Pharmacol 52:172-179.

Jasper JR, Lesnick JD, Chang LK, Yamanishi SS, Chang TK, Hsu SA, Daunt DA, Bonhaus DW

and Eglen RM (1998) Ligand efficacy and potency at recombinant alpha2 adrenergic

receptors: agonist-mediated [35S]GTPgammaS binding. Biochem Pharmacol 55:1035-

1043.

Ji J, Schrimpf MR, Sippy KB, Bunnelle WH, Li T, Anderson DJ, Faltynek C, Surowy CS, Dyhring

T, Ahring PK and Meyer MD (2007) Synthesis and structure-activity relationship studies

of 3,6-diazabicyclo[3.2.0]heptanes as novel alpha4beta2 nicotinic acetylcholine receptor

selective agonists. J Med Chem 50:5493-5508.

Johannessen M, Fontanilla D, Mavlyutov T, Ruoho AE and Jackson MB (2011) Antagonist action

of progesterone at sigma-receptors in the modulation of voltage-gated sodium channels. Am

J Physiol Cell Physiol 300:C328-337.

Johnston TH, Fox SH, Piggott MJ, Savola JM and Brotchie JM (2010) The alpha adrenergic

antagonist fipamezole improves quality of levodopa action in Parkinsonian primates. Mov

Disord 25:2084-2093.

Johnston TH, Huot P, Fox SH, Koprich JB, Szeliga KT, James JW, Graef JD, Letchworth SR,

Jordan KG, Hill MP and Brotchie JM (2013) TC-8831, a nicotinic acetylcholine receptor

46 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther agonist, reduces l-DOPA-induced dyskinesia in the MPTP macaque. Neuropharmacology

73C:337-347.

Johnston TH, Millar Z, Huot P, Wagg K, Thiele S, Salomonczyk D, Yong-Kee CJ, Gandy MN,

McIldowie M, Lewis KD, Gomez-Ramirez J, Lee J, Fox SH, Martin-Iverson M, Nash JE,

Piggott MJ and Brotchie JM (2012) A novel MDMA analogue, UWA-101, that lacks

psychoactivity and cytotoxicity, enhances L-DOPA benefit in parkinsonian primates.

FASEB J 26:2154-2163.

Jones CA, Johnston LC, Jackson MJ, Smith LA, van Scharrenburg G, Rose S, Jenner PG and

McCreary AC (2010) An in vivo pharmacological evaluation of pardoprunox (SLV308)--a

novel combined dopamine D(2)/D(3) receptor partial agonist and 5-HT(1A) receptor

agonist with efficacy in experimental models of Parkinson's disease. Eur

Neuropsychopharmacol 20:582-593.

Kanda T, Jackson MJ, Smith LA, Pearce RK, Nakamura J, Kase H, Kuwana Y and Jenner P (2000)

Combined use of the adenosine A(2A) antagonist KW-6002 with L-DOPA or with selective

D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in

MPTP-treated monkeys. Exp Neurol 162:321-327.

Kavitha CV, Gaonkar SL, Narendra Sharath Chandra JN, Sadashiva CT and Rangappa KS (2007)

Synthesis and screening for acetylcholinesterase inhibitor activity of some novel 2-butyl-

1,3-diaza-spiro[4,4]non-1-en-4-ones: derivatives of irbesartan key intermediate. Bioorg

Med Chem 15:7391-7398.

Kawamoto H, Ozaki S, Itoh Y, Miyaji M, Arai S, Nakashima H, Kato T, Ohta H and Iwasawa Y

(1999) Discovery of the first potent and selective small molecule opioid receptor-like

(ORL1) antagonist: 1-[(3R,4R)-1-cyclooctylmethyl-3- hydroxymethyl-4-piperidyl]-3-

ethyl-1, 3-dihydro-2H-benzimidazol-2-one (J-113397). J Med Chem 42:5061-5063.

47 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Kebabian JW, Britton DR, DeNinno MP, Perner R, Smith L, Jenner P, Schoenleber R and Williams

M (1992) A-77636: a potent and selective dopamine D1 receptor agonist with

antiparkinsonian activity in marmosets. Eur J Pharmacol 229:203-209.

Kitbunnadaj R, Zuiderveld OP, De Esch IJ, Vollinga RC, Bakker R, Lutz M, Spek AL, Cavoy E,

Deltent MF, Menge WM, Timmerman H and Leurs R (2003) Synthesis and structure-

activity relationships of conformationally constrained histamine H(3) receptor agonists. J

Med Chem 46:5445-5457.

Knight AR, Misra A, Quirk K, Benwell K, Revell D, Kennett G and Bickerdike M (2004)

Pharmacological characterisation of the agonist radioligand binding site of 5-HT(2A), 5-

HT(2B) and 5-HT(2C) receptors. Naunyn Schmiedebergs Arch Pharmacol 370:114-123.

Koe BK, Nielsen JA, Macor JE and Heym J (1992) Biochemical and behavioral studies of the 5-

HT1B receptor agonist, CP-94,253. Drug Dev Res 26:241-250.

Kooyman AR, Zwart R, Vanderheijden PM, Van Hooft JA and Vijverberg HP (1994) Interaction

between enantiomers of mianserin and ORG3770 at 5-HT3 receptors in cultured mouse

neuroblastoma cells. Neuropharmacology 33:501-507.

Koprich JB, Fox SH, Johnston TH, Goodman A, Le Bourdonnec B, Dolle RE, Dehaven RN,

Dehaven-Hudkins DL, Little PJ and Brotchie JM (2011) The selective mu-opioid receptor

antagonist adl5510 reduces levodopa-induced dyskinesia without affecting

antiparkinsonian action in mptp-lesioned macaque model of Parkinson's disease. Mov

Disord 26:1225-1233.

Koprich JB, Huot P, Fox SH, Jarvie K, Lang AE, Seeman P and Brotchie JM (2013) The effects

of fast-off-D2 receptor antagonism on L-DOPA-induced dyskinesia and psychosis in

parkinsonian macaques. Prog Neuropsychopharmacol Biol Psychiatry 43:151-156.

48 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Korinek M, Kapras V, Vyklicky V, Adamusova E, Borovska J, Vales K, Stuchlik A, Horak M,

Chodounska H and Vyklicky L, Jr. (2011) Neurosteroid modulation of N-methyl-D-

aspartate receptors: molecular mechanism and behavioral effects. Steroids 76:1409-1418.

Krintel C, Harpsoe K, Zachariassen LG, Peters D, Frydenvang K, Pickering DS, Gajhede M and

Kastrup JS (2013) Structural analysis of the positive AMPA receptor modulators CX516

and Me-CX516 in complex with the GluA2 ligand-binding domain. Acta Crystallogr D

Biol Crystallogr 69:1645-1652.

Kroeze WK, Hufeisen SJ, Popadak BA, Renock SM, Steinberg S, Ernsberger P, Jayathilake K,

Meltzer HY and Roth BL (2003) H1-histamine receptor affinity predicts short-term weight

gain for typical and atypical drugs. Neuropsychopharmacology 28:519-526.

Kuiper GG, Carlsson B, Grandien K, Enmark E, Ha Ggblad J, Nilsson S and Gustafsson JK (1997)

Comparison of the Ligand Binding Specificity and Transcript Tissue Distribution of

Estrogen Receptors alpha and beta. Endocrinology 138:863-870.

Lahti RA, Evans DL, Stratman NC and Figur LM (1993) Dopamine D4 versus D2 receptor

selectivity of dopamine receptor antagonists: possible therapeutic implications. Eur J

Pharmacol 236:483-486.

Lampen A, Meyer S and Nau H (2001) Phytanic acid and docosahexaenoic acid increase the

metabolism of all-trans-retinoic acid and CYP26 gene expression in intestinal cells.

Biochim Biophys Acta 1521:97-106.

Lapinsky DJ, Aggarwal S, Huang Y, Surratt CK, Lever JR, Foster JD and Vaughan RA (2009) A

novel photoaffinity ligand for the dopamine transporter based on . Bioorg Med

Chem 17:3770-3774.

Lejeune F, Newman-Tancredi A, Audinot V and Millan MJ (1997) Interactions of (+)- and (-)-8-

and 7-hydroxy-2-(di-n-propylamino)tetralin at human (h)D3, hD2 and h serotonin1A

49 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther receptors and their modulation of the activity of serotoninergic and dopaminergic neurones

in rats. J Pharmacol Exp Ther 280:1241-1249.

Leysen JE, Awouters F, Kennis L, Laduron PM, Vandenberk J and Janssen PA (1981) Receptor

binding profile of R 41 468, a novel antagonist at 5-HT2 receptors. Life Sci 28:1015-1022.

Liegeois JF, Bruhwyler J, Damas J, Rogister F, Masereel B, Geczy J and Delarge J (1995)

Modulation of the clozapine structure increases its selectivity for the dopamine D4 receptor.

Eur J Pharmacol 273:R1-3.

Liu C, Ma X, Jiang X, Wilson SJ, Hofstra CL, Blevitt J, Pyati J, Li X, Chai W, Carruthers N and

Lovenberg TW (2001) Cloning and pharmacological characterization of a fourth histamine

receptor (H(4)) expressed in bone marrow. Mol Pharmacol 59:420-426.

Loschmann PA, Chong PN, Nomoto M, Tepper PG, Horn AS, Jenner P and Marsden CD (1989)

Stereoselective reversal of MPTP-induced parkinsonism in the marmoset after dermal

application of N-0437. Eur J Pharmacol 166:373-380.

Lotta T, Vidgren J, Tilgmann C, Ulmanen I, Melen K, Julkunen I and Taskinen J (1995) Kinetics

of human soluble and membrane-bound catechol O-methyltransferase: a revised

mechanism and description of the thermolabile variant of the enzyme. Biochemistry

34:4202-4210.

Luo W, Yu QS, Kulkarni SS, Parrish DA, Holloway HW, Tweedie D, Shafferman A, Lahiri DK,

Brossi A and Greig NH (2006) Inhibition of human acetyl- and butyrylcholinesterase by

novel carbamates of (-)- and (+)-tetrahydrofurobenzofuran and methanobenzodioxepine. J

Med Chem 49:2174-2185.

Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh SM, Matagne A and Fuks B (2004)

The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug

levetiracetam. Proc Natl Acad Sci U S A 101:9861-9866.

50 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Lyon RA, Glennon RA and Titeler M (1986) 3,4-Methylenedioxymethamphetamine (MDMA):

stereoselective interactions at brain 5-HT1 and 5-HT2 receptors. Psychopharmacology

(Berl) 88:525-526.

Maitre M, Ratomponirina C, Gobaille S, Hode Y and Hechler V (1994) Displacement of [3H]

gamma-hydroxybutyrate binding by benzamide neuroleptics and but not

by other antipsychotics. Eur J Pharmacol 256:211-214.

Malysz J, Anderson DJ, Gronlien JH, Ji J, Bunnelle WH, Hakerud M, Thorin-Hagene K, Ween H,

Helfrich R, Hu M, Gubbins E, Gopalakrishnan S, Puttfarcken PS, Briggs CA, Li J, Meyer

MD, Dyhring T, Ahring PK, Nielsen EO, Peters D, Timmermann DB and Gopalakrishnan

M (2010) In vitro pharmacological characterization of a novel selective alpha7 neuronal

nicotinic acetylcholine receptor agonist ABT-107. J Pharmacol Exp Ther 334:863-874.

Marks MJ, Wageman CR, Grady SR, Gopalakrishnan M and Briggs CA (2009) Selectivity of ABT-

089 for alpha4beta2* and alpha6beta2* nicotinic acetylcholine receptors in brain. Biochem

Pharmacol 78:795-802.

Martin GE, Williams M, Pettibone DJ, Zrada MM, Lotti VJ, Taylor DA and Jones JH (1985)

Selectivity of (+)-4-propyl-9-hydroxynaphthoxazine [(+)-PHNO] for dopamine receptors

in vitro and in vivo. J Pharmacol Exp Ther 233:395-401.

Mathiesen JM, Svendsen N, Brauner-Osborne H, Thomsen C and Ramirez MT (2003) Positive

allosteric modulation of the human metabotropic glutamate receptor 4 (hmGluR4) by SIB-

1893 and MPEP. Br J Pharmacol 138:1026-1030.

Matsubayashi H, Swanson KL and Albuquerque EX (1997) Amantadine inhibits nicotinic

acetylcholine receptor function in hippocampal neurons. J Pharmacol Exp Ther 281:834-

844.

51 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Maurice T, Junien JL and Privat A (1997) Dehydroepiandrosterone sulfate attenuates dizocilpine-

induced learning impairment in mice via sigma 1-receptors. Behav Brain Res 83:159-164.

Maurice T, Urani A, Phan VL and Romieu P (2001) The interaction between neuroactive steroids

and the sigma1 receptor function: behavioral consequences and therapeutic opportunities.

Brain Res Brain Res Rev 37:116-132.

McCall RB, Lookingland KJ, Bedard PJ and Huff RM (2005) Sumanirole, a highly dopamine D2-

selective receptor agonist: in vitro and in vivo pharmacological characterization and

efficacy in animal models of Parkinson's disease. J Pharmacol Exp Ther 314:1248-1256.

Metcalf MD, Yekkirala AS, Powers MD, Kitto KF, Fairbanks CA, Wilcox GL and Portoghese PS

(2012) The delta opioid receptor agonist SNC80 selectively activates heteromeric mu-delta

opioid receptors. ACS Chem Neurosci 3:505-509.

Meyer C, Schmid R, Schmieding K, Falkenstein E and Wehling M (1998a) Characterization of

high affinity progesterone-binding membrane proteins by anti-peptide antiserum. Steroids

63:111-116.

Meyer C, Schmieding K, Falkenstein E and Wehling M (1998b) Are high-affinity progesterone

binding site(s) from porcine liver microsomes members of the sigma receptor family? Eur

J Pharmacol 347:293-299.

Michaelides MR, Hong Y, DiDomenico S, Jr., Asin KE, Britton DR, Lin CW, Williams M and

Shiosaki K (1995) (5aR,11bS)-4,5,5a,6,7,11b-hexahydro-2-propyl-3-thia-5-azacyclopent-

1- ena[c]-phenanthrene-9,10-diol (A-86929): a potent and selective dopamine D1 agonist

that maintains behavioral efficacy following repeated administration and characterization

of its diacetyl prodrug (ABT-431). J Med Chem 38:3445-3447.

Mihalak KB, Carroll FI and Luetje CW (2006) Varenicline is a partial agonist at alpha4beta2 and

a full agonist at alpha7 neuronal nicotinic receptors. Mol Pharmacol 70:801-805.

52 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Millan MJ, Cussac D, Gobert A, Lejeune F, Rivet JM, Mannoury La Cour C, Newman-Tancredi

A and Peglion JL (2004a) S32504, a novel naphtoxazine agonist at dopamine D3/D2

receptors: I. Cellular, electrophysiological, and neurochemical profile in comparison with

ropinirole. J Pharmacol Exp Ther 309:903-920.

Millan MJ, Di Cara B, Hill M, Jackson M, Joyce JN, Brotchie J, McGuire S, Crossman A, Smith

L, Jenner P, Gobert A, Peglion JL and Brocco M (2004b) S32504, a novel naphtoxazine

agonist at dopamine D3/D2 receptors: II. Actions in rodent, primate, and cellular models of

antiparkinsonian activity in comparison to ropinirole. J Pharmacol Exp Ther 309:921-935.

Millan MJ, Gobert A, Newman-Tancredi A, Lejeune F, Cussac D, Rivet JM, Audinot V, Dubuffet

T and Lavielle G (2000a) S33084, a novel, potent, selective, and competitive antagonist at

dopamine D(3)-receptors: I. Receptorial, electrophysiological and neurochemical profile

compared with GR218,231 and L741,626. J Pharmacol Exp Ther 293:1048-1062.

Millan MJ, Maiofiss L, Cussac D, Audinot V, Boutin JA and Newman-Tancredi A (2002)

Differential actions of antiparkinson agents at multiple classes of receptor.

I. A multivariate analysis of the binding profiles of 14 drugs at 21 native and cloned human

receptor subtypes. J Pharmacol Exp Ther 303:791-804.

Millan MJ, Newman-Tancredi A, Audinot V, Cussac D, Lejeune F, Nicolas JP, Coge F, Galizzi

JP, Boutin JA, Rivet JM, Dekeyne A and Gobert A (2000b) Agonist and antagonist actions

of yohimbine as compared to fluparoxan at alpha(2)-adrenergic receptors (AR)s, serotonin

(5-HT)(1A), 5-HT(1B), 5-HT(1D) and dopamine D(2) and D(3) receptors. Significance for

the modulation of frontocortical monoaminergic transmission and depressive states.

Synapse 35:79-95.

53 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Millan MJ, Newman-Tancredi A, Quentric Y and Cussac D (2001) The "selective" dopamine D1

receptor antagonist, SCH23390, is a potent and high efficacy agonist at cloned human

serotonin2C receptors. Psychopharmacology (Berl) 156:58-62.

Mor M, Rivara S, Lodola A, Plazzi PV, Tarzia G, Duranti A, Tontini A, Piersanti G, Kathuria S

and Piomelli D (2004) Cyclohexylcarbamic acid 3'- or 4'-substituted biphenyl-3-yl esters

as fatty acid amide hydrolase inhibitors: synthesis, quantitative structure-activity

relationships, and molecular modeling studies. J Med Chem 47:4998-5008.

Motter AL and Ahern GP (2012) TRPA1 is a polyunsaturated fatty acid sensor in mammals. PLoS

One 7:e38439.

Mulder AH, Stoof JC and Horn AS (1980) Activation of presynaptic alpha-noradrenaline receptors

in rat brain by the potent dopamine-mimetic N,N-dipropyl-5,6-ADTN. Eur J Pharmacol

67:147-150.

Nash JF, Roth BL, Brodkin JD, Nichols DE and Gudelsky GA (1994) Effect of the R(-) and S(+)

isomers of MDA and MDMA on phosphatidyl inositol turnover in cultured cells expressing

5-HT2A or 5-HT2C receptors. Neurosci Lett 177:111-115.

Neumeyer JL, Froimowitz M, Baldessarini RJ, Campbell A and Gao YG (1988)

Neuropharmacology and stereochemistry of dopamine receptor agonist and antagonist

enantiomeric pairs. J Recept Res 8:83-96.

Newman AH, Kline RH, Allen AC, Izenwasser S, George C and Katz JL (1995) Novel 4'-

substituted and 4',4"-disubstituted 3 alpha-(diphenylmethoxy) analogs as potent and

selective dopamine uptake inhibitors. J Med Chem 38:3933-3940.

Newman-Tancredi A, Cussac D, Audinot V, Nicolas JP, De Ceuninck F, Boutin JA and Millan MJ

(2002a) Differential actions of antiparkinson agents at multiple classes of monoaminergic

54 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther receptor. II. Agonist and antagonist properties at subtypes of dopamine D(2)-like receptor

and alpha(1)/alpha(2)-adrenoceptor. J Pharmacol Exp Ther 303:805-814.

Newman-Tancredi A, Cussac D, Quentric Y, Touzard M, Verriele L, Carpentier N and Millan MJ

(2002b) Differential actions of antiparkinson agents at multiple classes of monoaminergic

receptor. III. Agonist and antagonist properties at serotonin, 5-HT(1) and 5-HT(2), receptor

subtypes. J Pharmacol Exp Ther 303:815-822.

Newman-Tancredi A, Gavaudan S, Conte C, Chaput C, Touzard M, Verriele L, Audinot V and

Millan MJ (1998) Agonist and antagonist actions of antipsychotic agents at 5-HT1A

receptors: a [35S]GTPgammaS binding study. Eur J Pharmacol 355:245-256.

Newman-Tancredi A, Martel JC, Assie MB, Buritova J, Lauressergues E, Cosi C, Heusler P, Bruins

Slot L, Colpaert FC, Vacher B and Cussac D (2009) Signal transduction and functional

selectivity of F15599, a preferential post-synaptic 5-HT1A receptor agonist. Br J

Pharmacol 156:338-353.

Nielsen EB and Andersen PH (1992) Dopamine receptor occupancy in vivo: behavioral correlates

using NNC-112, NNC-687 and NNC-756, new selective dopamine D1 receptor antagonists.

Eur J Pharmacol 219:35-44.

Owens MJ, Knight DL and Nemeroff CB (2001) Second-generation SSRIs: human monoamine

transporter binding profile of and R-. Biol Psychiatry 50:345-350.

Ozaki S, Kawamoto H, Itoh Y, Miyaji M, Azuma T, Ichikawa D, Nambu H, Iguchi T, Iwasawa Y

and Ohta H (2000) In vitro and in vivo pharmacological characterization of J-113397, a

potent and selective non-peptidyl ORL1 receptor antagonist. Eur J Pharmacol 402:45-53.

Paddock ML, Wiley SE, Axelrod HL, Cohen AE, Roy M, Abresch EC, Capraro D, Murphy AN,

Nechushtai R, Dixon JE and Jennings PA (2007) MitoNEET is a uniquely folded 2Fe 2S

55 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther outer mitochondrial membrane protein stabilized by pioglitazone. Proc Natl Acad Sci U S

A 104:14342-14347.

Pan W and Kastin AJ (2007) From MIF-1 to endomorphin: the Tyr-MIF-1 family of peptides.

Peptides 28:2411-2434.

Patel S, Freedman S, Chapman KL, Emms F, Fletcher AE, Knowles M, Marwood R, McAllister

G, Myers J, Curtis N, Kulagowski JJ, Leeson PD, Ridgill M, Graham M, Matheson S,

Rathbone D, Watt AP, Bristow LJ, Rupniak NM, Baskin E, Lynch JJ and Ragan CI (1997)

Biological profile of L-745,870, a selective antagonist with high affinity for the dopamine

D4 receptor. J Pharmacol Exp Ther 283:636-647.

Peeters M, Romieu P, Maurice T, Su TP, Maloteaux JM and Hermans E (2004) Involvement of the

sigma 1 receptor in the modulation of dopaminergic transmission by amantadine. Eur J

Neurosci 19:2212-2220.

Perucca E (1997) A pharmacological and clinical review on topiramate, a new antiepileptic drug.

Pharmacol Res 35:241-256.

Philippens IH, Joosen MJ, Ahnaou A, Andres I and Drinkenburg WP (2014) Anti-Parkinson effects

of a selective alpha2C-adrenoceptor antagonist in the MPTP marmoset model. Behav Brain

Res 269:81-86.

Pilleri M and Antonini A (2015) Therapeutic strategies to prevent and manage dyskinesias in

Parkinson's disease. Expert Opin Drug Saf 14:281-294.

Porter RH, Jaeschke G, Spooren W, Ballard TM, Buttelmann B, Kolczewski S, Peters JU, Prinssen

E, Wichmann J, Vieira E, Muhlemann A, Gatti S, Mutel V and Malherbe P (2005)

Fenobam: a clinically validated nonbenzodiazepine anxiolytic is a potent, selective, and

noncompetitive mGlu5 receptor antagonist with inverse agonist activity. J Pharmacol Exp

Ther 315:711-721.

56 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Portoghese PS, Sultana M and Takemori AE (1988) Naltrindole, a highly selective and potent non-

peptide delta opioid receptor antagonist. Eur J Pharmacol 146:185-186.

Poulain R, Horvath D, Bonnet B, Eckhoff C, Chapelain B, Bodinier MC and Deprez B (2001) From

hit to lead. Combining two complementary methods for focused library design. Application

to mu opiate ligands. J Med Chem 44:3378-3390.

Pritchett DB, Luddens H and Seeburg PH (1989) Type I and type II GABAA-benzodiazepine

receptors produced in transfected cells. Science 245:1389-1392.

Pugsley TA, Davis MD, Akunne HC, MacKenzie RG, Shih YH, Damsma G, Wikstrom H, Whetzel

SZ, Georgic LM, Cooke LW and et al. (1995) Neurochemical and functional

characterization of the preferentially selective dopamine D3 agonist PD 128907. J

Pharmacol Exp Ther 275:1355-1366.

Purohit A, Smith C, Herrick-Davis K and Teitler M (2005) Stable expression of constitutively

activated mutant h5HT6 and h5HT7 serotonin receptors: inverse agonist activity of

antipsychotic drugs. Psychopharmacology (Berl) 179:461-469.

Quik M, Campos C, Bordia T, Strachan JP, Zhang J, McIntosh JM, Letchworth S and Jordan K

(2013) alpha4beta2 Nicotinic receptors play a role in the nAChR-mediated decline in L-

dopa-induced dyskinesias in parkinsonian rats. Neuropharmacology 71:191-203.

Rai M, Soragni E, Chou CJ, Barnes G, Jones S, Rusche JR, Gottesfeld JM and Pandolfo M (2010)

Two new pimelic diphenylamide HDAC inhibitors induce sustained frataxin upregulation

in cells from Friedreich's ataxia patients and in a mouse model. PLoS One 5:e8825.

Rashid AJ, So CH, Kong MM, Furtak T, El-Ghundi M, Cheng R, O'Dowd BF and George SR

(2007) D1-D2 dopamine receptor heterooligomers with unique pharmacology are coupled

to rapid activation of Gq/11 in the striatum. Proc Natl Acad Sci U S A 104:654-659.

57 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Regunathan S and Reis DJ (1996) Imidazoline receptors and their endogenous ligands. Annu Rev

Pharmacol Toxicol 36:511-544.

Reynolds IJ and Miller RJ (1989) Ifenprodil is a novel type of N-methyl-D-aspartate receptor

antagonist: interaction with polyamines. Mol Pharmacol 36:758-765.

Richelson E and Nelson A (1984) Antagonism by of neurotransmitter receptors of

normal human brain in vitro. J Pharmacol Exp Ther 230:94-102.

Richelson E and Souder T (2000) Binding of antipsychotic drugs to human brain receptors focus

on newer generation compounds. Life Sci 68:29-39.

Rothman RB, Baumann MH, Savage JE, Rauser L, McBride A, Hufeisen SJ and Roth BL (2000)

Evidence for possible involvement of 5-HT(2B) receptors in the cardiac valvulopathy

associated with fenfluramine and other serotonergic . Circulation 102:2836-

2841.

Rover S, Cesura AM, Huguenin P, Kettler R and Szente A (1997) Synthesis and biochemical

evaluation of N-(4-phenylthiazol-2-yl)benzenesulfonamides as high-affinity inhibitors of

kynurenine 3-hydroxylase. J Med Chem 40:4378-4385.

Rupprecht R, Reul JM, van Steensel B, Spengler D, Soder M, Berning B, Holsboer F and Damm

K (1993) Pharmacological and functional characterization of human mineralocorticoid and

glucocorticoid receptor ligands. Eur J Pharmacol 247:145-154.

Santangeli S, Sills GJ, Thompson GG and Brodie MJ (2002) Na(+) channel effects of remacemide

and desglycinyl-remacemide in rat cortical synaptosomes. Eur J Pharmacol 438:63-68.

Sautel F, Griffon N, Sokoloff P, Schwartz JC, Launay C, Simon P, Costentin J, Schoenfelder A,

Garrido F, Mann A and et al. (1995) Nafadotride, a potent preferential dopamine D3

receptor antagonist, activates locomotion in rodents. J Pharmacol Exp Ther 275:1239-

1246.

58 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Scheller D, Ullmer C, Berkels R, Gwarek M and Lubbert H (2009) The in vitro receptor profile of

rotigotine: a new agent for the treatment of Parkinson's disease. Naunyn Schmiedebergs

Arch Pharmacol 379:73-86.

Schipper J, Tulp MT and Sijbesma H (1990) Neurochemical profile of eltoprazine. Drug Metabol

Drug Interact 8:85-114.

Schlesinger F, Tammena D, Krampfl K and Bufler J (2005) Two mechanisms of action of the

adamantane derivative IEM-1460 at human AMPA-type glutamate receptors. Br J

Pharmacol 145:656-663.

Schlicker E, Werner U, Hamon M, Gozlan H, Nickel B, Szelenyi I and Gothert M (1992)

Anpirtoline, a novel, highly potent 5-HT1B receptor agonist with

antinociceptive/antidepressant-like actions in rodents. Br J Pharmacol 105:732-738.

Schoepp DD, Ornstein PL, Salhoff CR and Leander JD (1991) Neuroprotectant effects of

LY274614, a structurally novel systemically active competitive NMDA receptor

antagonist. J Neural Transm Gen Sect 85:131-143.

Schotte A, Janssen PF, Gommeren W, Luyten WH, Van Gompel P, Lesage AS, De Loore K and

Leysen JE (1996) compared with new and reference antipsychotic drugs: in

vitro and in vivo receptor binding. Psychopharmacology (Berl) 124:57-73.

Seely KA, Brents LK, Franks LN, Rajasekaran M, Zimmerman SM, Fantegrossi WE and Prather

PL (2012) AM-251 and rimonabant act as direct antagonists at mu-opioid receptors:

implications for opioid/cannabinoid interaction studies. Neuropharmacology 63:905-915.

Seeman P, Guan HC and Hirbec H (2009) Dopamine D2High receptors stimulated by

, lysergic acid diethylamide, salvinorin A, and modafinil. Synapse 63:698-

704.

59 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Selkirk JV, Scott C, Ho M, Burton MJ, Watson J, Gaster LM, Collin L, Jones BJ, Middlemiss DN

and Price GW (1998) SB-224289--a novel selective (human) 5-HT1B receptor antagonist

with negative intrinsic activity. Br J Pharmacol 125:202-208.

Sethi KK, Vullo D, Verma SM, Tanc M, Carta F and Supuran CT (2013) Carbonic anhydrase

inhibitors: synthesis and inhibition of the human carbonic anhydrase isoforms I, II, VII, IX

and XII with sulfonamides incorporating 4,5,6,7-tetrabromophthalimide moiety.

Bioorg Med Chem 21:5973-5982.

Sheardown MJ, Nielsen EO, Hansen AJ, Jacobsen P and Honore T (1990) 2,3-Dihydroxy-6-nitro-

7-sulfamoyl-benzo(F)quinoxaline: a neuroprotectant for cerebral ischemia. Science

247:571-574.

Shen W, Plotkin JL, Francardo V, Ko WK, Xie Z, Li Q, Fieblinger T, Wess J, Neubig RR, Lindsley

CW, Conn PJ, Greengard P, Bezard E, Cenci MA and Surmeier DJ (2015) M4 Muscarinic

Receptor Signaling Ameliorates Striatal Plasticity Deficits in Models of L-DOPA-Induced

Dyskinesia. Neuron 88:762-773.

Shiosaki K, Jenner P, Asin KE, Britton DR, Lin CW, Michaelides M, Smith L, Bianchi B,

Didomenico S, Hodges L, Hong Y, Mahan L, Mikusa J, Miller T, Nikkel A, Stashko M,

Witte D and Williams M (1996) ABT-431: the diacetyl prodrug of A-86929, a potent and

selective dopamine D1 receptor agonist: in vitro characterization and effects in animal

models of Parkinson's disease. J Pharmacol Exp Ther 276:150-160.

Singh S (2000) Chemistry, design, and structure-activity relationship of cocaine antagonists. Chem

Rev 100:925-1024.

Smith U (2001) Pioglitazone: mechanism of action. Int J Clin Pract Suppl:13-18.

60 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Sousa A and Ticku MK (1997) Interactions of the neurosteroid dehydroepiandrosterone sulfate

with the GABA(A) receptor complex reveals that it may act via the picrotoxin site. J

Pharmacol Exp Ther 282:827-833.

Spencer DG, Jr., Glaser T and Traber J (1987) Serotonin receptor subtype mediation of the

interoceptive discriminative stimuli induced by 5-methoxy-N,N-dimethyltryptamine.

Psychopharmacology (Berl) 93:158-166.

Stasi MA, Borsini F, Varani K, Vincenzi F, Di Cesare MA, Minetti P, Ghirardi O and Carminati P

(2006) ST 1535: a preferential A2A adenosine receptor antagonist. Int J

Neuropsychopharmacol 9:575-584.

Subramaniam S, Donevan SD and Rogawski MA (1996) Block of the N-methyl-D-aspartate

receptor by remacemide and its des-glycine metabolite. J Pharmacol Exp Ther 276:161-

168.

Sunahara RK, Guan HC, O'Dowd BF, Seeman P, Laurier LG, Ng G, George SR, Torchia J, Van

Tol HH and Niznik HB (1991) Cloning of the gene for a human dopamine D5 receptor with

higher affinity for dopamine than D1. Nature 350:614-619.

Tatsumi M, Groshan K, Blakely RD and Richelson E (1997) Pharmacological profile of

antidepressants and related compounds at human monoamine transporters. Eur J

Pharmacol 340:249-258.

Tice MA, Hashemi T, Taylor LA, Duffy RA and McQuade RD (1994) Characterization of the

binding of SCH 39166 to the five cloned dopamine receptor subtypes. Pharmacol Biochem

Behav 49:567-571.

Toll L, Bruchas MR, Calo G, Cox BM and Zaveri NT (2016) Nociceptin/Orphanin FQ Receptor

Structure, Signaling, Ligands, Functions, and Interactions with Opioid Systems. Pharmacol

Rev 68:419-457.

61 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Tricklebank MD, Forler C, Middlemiss DN and Fozard JR (1985) Subtypes of the 5-HT receptor

mediating the behavioural responses to 5-methoxy-N,N-dimethyltryptamine in the rat. Eur

J Pharmacol 117:15-24.

Tsuruta K, Frey EA, Grewe CW, Cote TE, Eskay RL and Kebabian JW (1981) Evidence that LY-

141865 specifically stimulates the D-2 dopamine receptor. Nature 292:463-465.

Uhlen S, Porter AC and Neubig RR (1994) The novel alpha-2 adrenergic radioligand [3H]-MK912

is alpha-2C selective among human alpha-2A, alpha-2B and alpha-2C adrenoceptors. J

Pharmacol Exp Ther 271:1558-1565.

Van der Weide J, De Vries JB, Tepper PG and Horn AS (1986) Pharmacological profiles of three

new, potent and selective dopamine receptor agonists: N-0434, N-0437 and N-0734. Eur J

Pharmacol 125:273-282.

Van der Weide J, De Vries JB, Tepper PG, Krause DN, Dubocovich ML and Horn AS (1988) N-

0437: a selective D-2 dopamine receptor agonist in in vitro and in vivo models. Eur J

Pharmacol 147:249-258.

Vanover KE, Kleven MS and Woolverton WL (1991) Blockade of the discriminative stimulus

effects of cocaine in rhesus monkeys with the D(1) dopamine antagonists SCH-39166 and

A-66359. Behav Pharmacol 2:151-159.

Vanover KE, Weiner DM, Makhay M, Veinbergs I, Gardell LR, Lameh J, Del Tredici AL, Piu F,

Schiffer HH, Ott TR, Burstein ES, Uldam AK, Thygesen MB, Schlienger N, Andersson

CM, Son TY, Harvey SC, Powell SB, Geyer MA, Tolf BR, Brann MR and Davis RE (2006)

Pharmacological and behavioral profile of N-(4-fluorophenylmethyl)-N-(1-

methylpiperidin-4-yl)-N'-(4-(2-methylpropylo xy)phenylmethyl) carbamide (2R,3R)-

dihydroxybutanedioate (2:1) (ACP-103), a novel 5-hydroxytryptamine(2A) receptor

inverse agonist. J Pharmacol Exp Ther 317:910-918.

62 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Verma V, Mann A, Costain W, Pontoriero G, Castellano JM, Skoblenick K, Gupta SK, Pristupa Z,

Niznik HB, Johnson RL, Nair VD and Mishra RK (2005) Modulation of agonist binding to

human dopamine receptor subtypes by L-prolyl-L-leucyl-glycinamide and a

peptidomimetic analog. J Pharmacol Exp Ther 315:1228-1236.

Verrico CD, Miller GM and Madras BK (2007) MDMA (Ecstasy) and human dopamine,

norepinephrine, and serotonin transporters: implications for MDMA-induced neurotoxicity

and treatment. Psychopharmacology (Berl) 189:489-503.

Von Voigtlander PF and Lewis RA (1982) U-50,488, a selective kappa opioid agonist: comparison

to other reputed kappa agonists. Prog Neuropsychopharmacol Biol Psychiatry 6:467-470.

Vranesic I, Ofner S, Flor PJ, Bilbe G, Bouhelal R, Enz A, Desrayaud S, McAllister K, Kuhn R and

Gasparini F (2014) AFQ056/mavoglurant, a novel clinically effective mGluR5 antagonist:

identification, SAR and pharmacological characterization. Bioorg Med Chem 22:5790-

5803.

Wainscott DB, Sasso DA, Kursar JD, Baez M, Lucaites VL and Nelson DL (1998)

[3H]Rauwolscine: an antagonist radioligand for the cloned human 5-hydroxytryptamine2b

(5-HT2B) receptor. Naunyn Schmiedebergs Arch Pharmacol 357:17-24.

Walters MR, Dutertre M and Smith CL (2002) SKF-82958 is a subtype-selective estrogen receptor-

alpha (ERalpha ) agonist that induces functional interactions between ERalpha and AP-1.

J Biol Chem 277:1669-1679.

Wang C and Niu L (2013) Mechanism of inhibition of the GluA2 AMPA receptor channel opening

by talampanel and its enantiomer: the stereochemistry of the 4-methyl group on the

diazepine ring of 2,3-benzodiazepine derivatives. ACS Chem Neurosci 4:635-644.

63 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Weinshank RL, Zgombick JM, Macchi MJ, Branchek TA and Hartig PR (1992) Human serotonin

1D receptor is encoded by a subfamily of two distinct genes: 5-HT1D alpha and 5-HT1D

beta. Proc Natl Acad Sci U S A 89:3630-3634.

Wichmann J, Adam G, Rover S, Cesura AM, Dautzenberg FM and Jenck F (1999) 8-acenaphthen-

1-yl-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one derivatives as orphanin FQ receptor

agonists. Bioorg Med Chem Lett 9:2343-2348.

Wong DT, Threlkeld PG, Best KL and Bymaster FP (1982) A new inhibitor of norepinephrine

uptake devoid of affinity for receptors in rat brain. J Pharmacol Exp Ther 222:61-65.

Wood PL, Smith T, Lane N, Khan MA, Ehrmantraut G and Goodenowe DB (2011) Oral

bioavailability of the ether lipid plasmalogen precursor, PPI-1011, in the rabbit: a new

therapeutic strategy for Alzheimer's disease. Lipids Health Dis 10:227.

Xiang Z, Thompson AD, Brogan JT, Schulte ML, Melancon BJ, Mi D, Lewis LM, Zou B, Yang

L, Morrison R, Santomango T, Byers F, Brewer K, Aldrich JS, Yu H, Dawson ES, Li M,

McManus O, Jones CK, Daniels JS, Hopkins CR, Xie XS, Conn PJ, Weaver CD and

Lindsley CW (2011) The Discovery and Characterization of ML218: A Novel, Centrally

Active T-Type Calcium Channel Inhibitor with Robust Effects in STN Neurons and in a

Rodent Model of Parkinson's Disease. ACS Chem Neurosci 2:730-742.

Yoshio R, Taniguchi T, Itoh H and Muramatsu I (2001) Affinity of serotonin receptor antagonists

and agonists to recombinant and native alpha1-adrenoceptor subtypes. Jpn J Pharmacol

86:189-195.

Zhang D, McGregor M, Bordia T, Perez XA, McIntosh JM, Decker MW and Quik M (2015) alpha7

nicotinic receptor agonists reduce levodopa-induced dyskinesias with severe nigrostriatal

damage. Mov Disord 30:1901-1911.

64 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther Zolkowska D, Jain R, Rothman RB, Partilla JS, Roth BL, Setola V, Prisinzano TE and Baumann

MH (2009) Evidence for the involvement of dopamine transporters in behavioral stimulant

effects of modafinil. J Pharmacol Exp Ther 329:738-746.

65 Veyres N, Hamadjida A, Huot P. Predictive value of parkinsonian primates in pharmacological studies, a comparison between the macaque, marmoset and squirrel monkey. J Pharmacol Exp Ther

66