2004 Linazasoro Farmacos No Utiles PD

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Movement Disorders

Vol. 19, No. 7, 2004, pp. 743–754


© 2004 Movement Disorder Society

Research Review

Recent Failures of New Potential Symptomatic Treatments for


Parkinson’s Disease: Causes and Solutions

Gurutz Linazasoro, MD*

Centro de Neurologı́a y Neurocirugı́a funcional, Clı́nica Quirón, San Sebastián, Guipúzcoa, Spain

Abstract: One major goal of current research in Parkinson’s of the working hypotheses and design of clinical trials, the
disease (PD) is the discovery of novel agents to improve validity of tools in current use to evaluate a specific effect, and
symptomatic management. The object of these new treatments the selectivity of the drugs used. All these factors may explain
should be to provide effective symptom control throughout the failure. This review focuses on pharmacological and surgical
course of the disease without the development of side effects treatments tested to improve the management of patients with
such as motor and psychiatric complications. Results of several motor fluctuations and dyskinesias. Some of the recent trials
clinical trials of new treatment options reported in the past 2 and possible reasons for their lack of success are critically
years have shown negative or unsatisfactory results. Most of analysed. Finally, some suggestions to avoid further failures
the drugs and surgical procedures used in these studies had and improve results are proposed. © 2004 Movement Disorder
been tested previously in 1-methyl-4-phenyl-1,2,3,6-tetrahy-
Society
dropyridine (MPTP) monkeys as well as in the classic 6-hy-
Key words: Parkinson’s disease; motor complications; ani-
droxydopamine–lesioned rat model. They raise several ques-
tions about the true reliability of animal studies, the adequacy mal models; treatment alternatives

A major goal of current research in Parkinson’s dis- degenerative process is quite limited to the dopaminergic
ease (PD) is the discovery of novel agents to improve neurons localised in the pars compacta of the substantia
symptomatic management in the hope that new treat- nigra (SNpc), and damaging the nigrostriatal pathway at
ments will provide effective and sustained symptom con- different levels in animals reproduces the striatal dopa-
trol throughout the course of the disease, without induc- mine (L-dopa) deficiency characteristic of PD.2 Rodent
ing side effects such as motor and psychiatric models were the first available and proved very useful as
complications.1 To reach this objective, it is important to early screens in the development of antiparkinsonian
have relevant models of the disease in which new phar- therapy. The unilateral 6-hydroxydopamine (6-OHDA)-
macological agents and surgical procedures could be lesioned rodent, through the ability to induce rotational
tested before clinical assessment. In some ways, PD has behaviour with apomorphine or amphetamine, has a high
been considered paradigmatic for other pathological con- predictive value to detect potential anti-parkinsonian
ditions, particularly neurodegenerative illness, and has agents.3 Because rotational behaviour does not resemble
the advantage of some unique features. For instance, the the motor abnormalities seen in PD, new methods have
been developed in recent years to better assess these and
also levodopa-induced dyskinesias.4,5
*Correspondence to: Dr. Gurutz Linazasoro, Centro de Neurologı́a y
Neurocirugı́a funcional, Clı́nica Quirón, Parque Alcolea s/n, 20012 San
The possibility of inducing parkinsonism in subhuman
Sebastián, Guipúzcoa, Spain. E-mail: glinazasoro@terra.es primates with the administration of 1-methyl-4-phenyl-
Received 14 September 2003; Revised 23 December 2003; Accepted 1,2,5,6-tetrahydropiridine (MPTP)6 led to the belief that
2 February 2004
Published online 21 April 2004 in Wiley InterScience (www. the predictability would be greatly improved because the
interscience.wiley.com). DOI: 10.1002/mds.20120 evaluation of motor, and to some extent nonmotor, be-

743
744 G. LINAZASORO

haviour would be closer to that of humans.7,8 The use- beneficial effects against parkinsonism and L-dopa–
fulness of these models has been reviewed recently.3,6 induced dyskinesias.
With the help of these models, further insights into the
pathophysiology of parkinsonism and dyskinesia have Dopaminergic Agents
significantly contributed to the formulation of more ac- Several new dopaminergic drugs are in advanced clin-
curate hypotheses as starting points to develop new treat- ical development and will soon be released to the market:
ment approaches.3,6 Despite high hopes of discovering sumanirole, rotigotine, L-dopa methylester and ethyl-
more effective pharmacological and surgical approaches, ester, and the triple combination of L-dopa/carbidopa/
the results of several clinical trials reported in the past 2 entacapone. Piribedil, which has been in use in France
years do not uphold such expectations. Indeed, most of for more than 2 decades, will become available in other
them have shown negative or at best unsatisfactory re- countries, and new drugs designed to improve the dopa-
sults when tried in PD patients (Table 1). Most of the minergic transmission to produce a better clinical re-
drugs and surgical procedures used in these studies had sponse are in the pipeline.
been tested previously in MPTP monkeys as well as the
classic 6-OHDA–lesioned rat model. The results of all Drugs Acting at the Dopamine Transporter Level.
these studies raise several questions about the true reli- These drugs increase the synaptic levels of dopamine
ability and validity of animal data, the adequacy of the by blocking the action of the synaptic dopamine trans-
current working hypotheses, and the presently used tools porter.11 The higher the levels of dopamine in the syn-
to evaluate a specific effect. apse, the longer and stronger the antiparkinsonian effect.
Brasofensine is one of these drugs, producing a potenti-
NEW PHARMACOLOGICAL TREATMENTS ation of the effects of L-dopa in MPTP-lesioned monkeys
FOR MOTOR FLUCTUATIONS AND without inducing dyskinesia.12 In 8 patients receiving the
LEVODOPA-INDUCED DYSKINESIAS: drug, an initial improvement over a 4-week period was
RECENT CLINICAL TRIALS observed, but this improvement has been lost by the end
This article focuses on pharmacological and surgi- of the study, and no significant differences were shown
cal treatments to improve the management of patients against placebo. A possible explanation of this may be
already suffering from motor fluctuations and dyski- that the metabolism of brasofensine in human beings is
nesia. New concepts on the organization of the basal different from that in monkeys, invalidating all preclin-
ganglia and the influence of different neurotransmit- ical toxicological investigations.12 Other drugs with the
ters and neuromodulators on motor function9,10 have same mechanism of action, such as NS2330, are now
led to the development of new drugs with potentially being developed.

TABLE 1. Treatment strategies with symptomatic potential in complicated PD:


Comparison between animal and human data
Animal data Human data
Park LID Park LID
Dopamine uptake blockers (brasofensine) ⫹⫹⫹ ⫹⫹⫹ - ??
D1 receptor agonists ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ -
Partial D2 agonists ⫹⫹⫹ ⫹⫹⫹ ⫹/⫺ ⫹/⫺
Adenosine A2a antagonists ⫹⫹⫹ ⫹⫹⫹ ⫹⫹ ⫹/⫺
Alfa 2 antagonists - ⫹⫹⫹ - ⫹/⫺
GABAergic ??? ??? ⫹/⫺ ⫹/⫺
Antiserotoninergic drugs (5HT2c) ⫹⫹⫹ ⫹⫹⫹ ?? ??
Serotoninergic drugs (5-HT1a) - ⫹⫹⫹ - ⫹⫹⫹
Glutamate antagonists ⫹⫹⫹ ⫹⫹⫹ ⫹/⫺ ⫹/⫺
Cannabinoid agonists ⫹⫹⫹ ⫹⫹⫹ - ⫹⫹
Cannabinoid antagonists - ⫹⫹⫹ ?? ??
Opioid antagonists - ⫹⫹⫹ - -
Opioid agonists ⫹⫹ - - -
Functional surgery ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹
Fetal transplantation ⫹⫹⫹ ⫹⫹⫹ ⫹/⫺ -

Park, parkinsonism; LID, levodopa-induced dyskinesias; ⫹⫹⫹, good effect; ⫹⫹, moder-
ate effect; ⫹/⫺, slight effect; -, no effect; PD, Parkinson’s disease.

Movement Disorders, Vol. 19, No. 7, 2004


FAILURES OF SYMPTOMATIC TREATMENTS FOR PD 745

Selective Dopamine Receptor Agonists. higher than full agonists mainly in patients with L-dopa–
D1 receptors have been repeatedly involved in the induced dyskinesia or psychiatric disturbances. Ter-
origin of L-dopa–induced dyskinesia,13,14 and selective guride is an example of a partial D2 agonist exhibiting
D1 receptors agonists have been shown to be highly both antiparkinsonian and antidyskinetic effects in
effective in MPTP monkeys, producing a natural motor MPTP-treated monkeys,23 but when administered to pa-
response without dyskinesia. Even more, D1 agonist did tients with motor complications the results were incon-
not produce dyskinesias in parkinsonian monkeys al- sistent. In some cases, terguride reduced dyskinesia with-
ready primed to develop dyskinesias,15 and this change out worsening parkinsonism, but in others, it worsened
occurring after a short period of administration might be both dyskinesia and PD.24
interpreted as a reversal of the underlying mechanism.
Some confusion still exists about the real motor effect of Miscellaneous.
D1 agonists due to the previous use of drugs without a Other drugs acting at the dopaminergic system
full D1 profile, such as CY 20824316 and SKF 38393. through varied mechanisms of action are being explored,
Dihydrexidine was the first high-affinity full D1 agonist although none of them have as yet been tested in humans.
tested in both animals17 and humans,18 and it produced a Among these drugs are dopamine stabilisers (such as
dramatic reduction of parkinsonian signs in severely OSU-6162), which are drugs with a weak D2 receptor
afflicted MPTP-treated monkeys17; in the only clinical antagonistic capacity, depending on the dopaminergic
trial reported, 4 patients received the drug intravenous- tone,25 D3, D4, and D5 agonists, D3 antagonists, and so
ly18: three of them suffered severe postural hypotension on. Recent data indicate the existence of a linear increase
and were withdrawn from the study; the remaining pa- of the expression of D3 receptors in relation to the
tient showed a 70% improvement in UPDRS score. The severity of dyskinesia in MPTP monkeys receiving L-do-
significant drop in blood pressure precluded human stud- pa.26 Dyskinesia was alleviated with BP 897, a partial
ies. agonist at the D3 receptor, when, importantly, the ben-
ABT-431 (along with isochromans A77636 and eficial therapeutic response to L-dopa was not affected.26
A86930), similar in structure and pharmacological prop- In contrast, full D3 antagonists, either nonselective or
erties to dihydrexidine and from which a comparable highly selective, also improved dyskinesia but at the
effect might be expected, were the next D1 agonists expense of a worsening of parkinsonism.26 While these
investigated. These agents had been used in MPTP mon- results support the use of D3 partial agonists in combi-
keys with good effect against parkinsonism and dyski- nation with L-dopa in patients with L-dopa–induced dys-
nesia.19 However, ABT-431, as with dihydrexidine, had kinesia, other studies have shown that D3 antagonists
poor oral bioavailability, although it was tolerated better may exert antiparkinsonian effects by their own.27 Such
than the latter. When tested in humans with PD and contradictions in animal models add confusion to the
motor complications, ABT-431 had a similar action to expected effects when these drugs are eventually tested
L-dopa, but without significant modification of dyskine- on patients.
sia.20,21 These results were somewhat disappointing, be-
cause expectation had been high, particularly concerning NONDOPAMINERGIC STRATEGIES
its potential antidyskinetic effect. Isochromans have not
Adenosine A2a Receptor Antagonists
been tested in PD patients; they do not have the problems
of bioavailability reported with dihydrexidine and ABT- Adenosine A2a receptors are localised to striatal cho-
431 but can cause severe side effects and behavioural linergic interneurons and also to the cell bodies of striatal
tolerance. Dinapsoline (DAR-201) has good oral avail- ␥-aminobutyric acid (GABA)ergic neurons, which orig-
ability, does not induce tolerance, and has been shown to inate the indirect pathway28; they are, therefore, colo-
be efficacious in rats and marmosets with parkinson- calised with D2 receptors.28 Through this selective lo-
ism.22 calisation, A2a receptors can influence both striatal
GABA and acetylcholine release.
Partial Agonists. Adenosine A2a agonists stimulate the indirect path-
This class of compounds that may act either as ago- way, increasing GABA release in the external globus
nists or antagonists, depending on the sensitivity of do- pallidus (GPe), whereas antagonists lead to a reduction in
pamine receptors: they exert agonist activity at dener- the levels of GABA in the GPe. Overactivity of the
vated supersensitive sites and competitively inhibit indirect pathway is one of the hallmarks of parkinson-
complete agonists at fully innervated normosensitive re- ism,9,10 so A2a antagonists could be beneficial in motor
ceptors. Thus, their therapeutic window appears to be symptoms. As these receptors are apparently absent in

Movement Disorders, Vol. 19, No. 7, 2004


746 G. LINAZASORO

the striatal neurons, which initiate the direct pathway and dyskinesias was observed. Thus, results are contradictory
in other dopaminergic pathways, these drugs could be and several explanations can be offered: different actions
free of the major side effects of other antiparkinsonian of L-dopa and apomorphine, and distinct neural mecha-
drugs.28 This hypothesis has been proved in 6-OHDA nisms underlying dyskinesia induced by L-dopa or apo-
rats and MPTP monkeys when the animals experienced a morphine.38 A prospective multicentric study including a
more natural motor response than with L-dopa.28 KW- large sample of fluctuating PD patients has been stopped
6002, VER-11135, SCH-58261, and SCH-63390 are because of a lack of efficacy in an interim analysis. What
some A2a antagonists tested that did not induce dyski- is clear from these small studies is that tolerability,
nesia even in primed animals. particularly regarding cardiovascular side effects, should
The expectations raised by these preclinical studies be improved, and animal studies heightened this expec-
have been only partially confirmed in two small clinical tation.
trials with KW-6002 in humans.29,30 Eighty-three PD Mirtazapine is a special case because it acts on the
patients with motor fluctuations and dyskinesia were noradrenergic and serotoninergic transmission. A few
included in a double-blind randomized study with three observations in patients have suggested that it might
arms (placebo, 20 mg/d KW6002, and 40 mg/d alleviate tremor and dyskinesia by a still unknown mech-
KW6002).29 The number of hours per day off were anism,39 but this benefit may be due to the antidepressant
slightly reduced (0.5–1.5 hours); dyskinesia remained and sedative effects of this drug.
unchanged throughout the study. Nausea was the most
common side effect, and 7% of those participating ex- GABAergic Drugs
perienced an increase in lipase level for no known rea- The pathophysiological basis of motor complications
son.29 In another study of 16 parkinsonian patients with is still a matter for debate. It has been suggested that
motor complications, a 40% potentiation of the L-dopa chronic and pulsatile administration of L-dopa might
effect was observed, with no increase in dyskinesia30; in result in an imbalance between the direct and indirect
this study, L-dopa was administered as an infusion. pathways connecting the striatum to the internal segment
The case of theophylline is confusing: like caffeine, it of the globus pallidus (GPi), leading to the appearance of
is not a specific adenosine antagonist. Open-label studies motor fluctuations and dyskinesia.1 GABA is the main
have shown that it could ameliorate PD symptoms with- neurotransmitter of both the indirect and direct striato-
out worsening dyskinesia.31 However, these results have pallidal pathways: numerous GABAergic neurons are
not been confirmed in a double blind study.32 found in the human basal ganglia as well as GABAa and
GABAb receptors. GABAa receptors upregulation in the
Alfa2 Noradrenergic Receptor Antagonists GPi correlates with the development of dyskinesias in-
The role of noradrenaline and adrenaline in motor duced by L-dopa or dopamine agonists in monkeys40,41
function is far from clear: even the distribution and role and PD patients.42,43 Also, direct injection of muscimol
of adrenergic receptors within the basal ganglia nuclei is (a GABA agonist) on the thalamus, GPi, and subthalamic
not totally known. Despite this background of uncer- nucleus (STN) in patients mimics the therapeutic effects
tainty, it was found that idazoxan, a selective alfa2 re- of lesions or deep brain stimulation (DBS) of these
ceptor antagonist, extended the motor action of a dose of nuclei.44 However, the influence of orally administered
L-dopa in MPTP-lesioned monkeys and reduced L-dopa– GABAergic drugs on parkinsonian motor disability is
induced dyskinesia.33 Similar effects have been reported contradictory. While early morning dystonia can be im-
recently with JP-173034 and yohimbine,35 and it has been proved by baclofen,45 other GABAergic compounds
suggested that these effects are mediated through an have shown no efficacy or have worsened PD symp-
action on the indirect pathway. Two clinical trials have toms.46 – 48 Gabapentin increases the synthesis and re-
been conducted to date36,37: in the first one, the effect of lease of GABA and may induce motor side effects, such
idazoxan on an apomorphine challenge was assessed in 8 as chorea and dystonia,49 suggesting that it may act on
patients with PD and motor complications.36 Four of the circuitry involved in motor control. We have shown
them experienced severe side effects, such as headache, that gabapentin improves the basal off motor score,
nausea, vomiting, and hypotension and did not complete thereby reducing the strength of the motor response after
the study. In the remaining 4 patients, a nonsignificant an L-dopa dose50 but other clinical and pharmacological
effect was found in both, duration of the motor benefit parameters are not improved and the overall clinical
and dyskinesias. In the other study, 18 patients with PD situation of patients remains unchanged. Previous studies
were assessed by using a L-dopa challenge.37 In this also suggested a moderate beneficial effect of gabapentin
study, the on time did not change but a benefit against in PD.51,52 To further complicate this scenario, it has

Movement Disorders, Vol. 19, No. 7, 2004


FAILURES OF SYMPTOMATIC TREATMENTS FOR PD 747

been shown that flumazenil, a GABA antagonist, may exogenous L-dopa, may affect the response alterations
improve features of PD.53 This was an open study in complicating L-dopa treatment of PD. Sarizotan is a
which 8 patients received an acute, intravenous dose of selective 5-HT1a agonist, which showed a potent anti-
flumazenil. These confusing results illustrate the diffi- dyskinetic effect without affecting parkinsonism in ani-
culty in predicting the consequences of the manipulation mal studies.62 Peak dose choreiform dyskinesias were
of the GABA system. It is the main inhibitory neuro- reduced by more than 90%.62 These results have been
transmitter in the central nervous system and GABA replicated in 17 patients,63 but long-term studies are
receptors are found in virtually all the basal ganglia needed.
nuclei. Therefore, a presumed positive effect derived
from the action on GABA receptors located in a given Glutamate Antagonists
nucleus may be hampered by the action on the same Glutamate is the main excitatory neurotransmitter in
receptor located in a different nucleus. Consequently the brain.64 – 66 Two major types of glutamate receptors
until more specific GABA drugs are synthesised, no firm have been identified: ionotropic and metabotropic. Iono-
conclusion about efficacy and mechanism of action can tropic are coupled to ion channels and can be divided in
be made. N-methyl-D-aspartate (NMDA), AMPA, and kainate re-
ceptors. Metabotropic receptors are coupled to G pro-
Drugs Acting on Serotoninergic Transmission teins. All these types of receptors are composed of mul-
The basal ganglia, mainly the striatum and the GPi/ tiple subtypes (NR1 and NR2 belong to the NMDA
substantia nigra pars reticulata (SNpr) complex, receive a family, mGluR1 to mGluR8 belong to the metabotropic
dense serotoninergic innervation mainly from the brain- family, etc.). The precise location and function of many
stem raphe nucleus. Among the different subtypes of of these subtypes are not totally known, most of them are
these receptors, 5HT2c is the most interesting for PD.54 distributed in various nuclei of the basal ganglia and in
This receptor is located at its highest concentration in the other brain structures, and there is a lack of drugs with an
GPi and SNpr, and it has been shown that its binding is adequate selectivity profile for a given receptor. Thus,
increased in postmortem brain tissue of parkinsonian the resultant effects of any glutamate antagonist may be
patients.54 Thus, targeting this receptor may be of benefit absolutely different if administered locally in a nucleus
in PD as can be inferred from the 5HT2c knockout rat, or systemically.64 – 66 In addition, this lack of precision
which displays spontaneous hyperlocomotion55 and has may produce severe side effects. This finding emphasises
been also shown in lesioned rats.56 At present, no selec- the difficulties in attributing an effect to a glutamate
tive 5HT2c antagonists are marketed and no human data antagonist and explains the possibility of obtaining con-
are available, although some atypical neuroleptics with tradictory results. Finally, glutamate has two faces: it is
this action, such as clozapine, are less prone to induce essential to the normal development and function of the
parkinsonism. In line with this, actions on this same brain, participating in key functions such as memory
receptor but located in the STN have been claimed to be formation and many others, but in certain circumstances,
responsible for oral dyskinesias, which were reverted it may be deleterious. But too little is as harmful as too
with a selective antagonist drug.57 Ritanserin, a selective much, because it participates actively in the natural re-
5HT2 antagonist, significantly ameliorated L-dopa–in- covery process after a lesion as has been shown in
duced dyskinesias in a single-blind, placebo-controlled, stroke.67 Caution is needed before using these drugs in
study performed in 10 patients,58 but this result has not clinical trials.
been replicated. There is a large amount of evidence supporting the
After Durif and colleagues59 noted that fluoxetine active role played by glutamate in the origin of parkin-
could improve L-dopa–induced dyskinesia, other sub- sonism and L-dopa–induced dyskinesia.68 For instance,
types of serotonin receptors (striatal 5HT1a receptors) the changes in the phosphorylation state of glutamate
have been implicated in the appearance of dyskinesia. receptors as a result of the denervation and the pulsatile
However, this single study was short and open, and the treatment with L-dopa seem to be critical in the origin of
effect of fluoxetine could be related to its tendency of L-dopa–induced dyskinesia.68 As with other classes of
worsening parkinsonism.60 In a recent open label study, drugs, the antiparkinsonian and antidyskinetic profile of
more than 400 parkinsonian patients with depression this type of compounds has been demonstrated elegantly
were given sertraline and neither parkinsonism nor L- in MPTP-treated monkeys,64 – 66 where these effects have
dopa–induced dyskinesia were modified.61 Conceivably, been shown with a large number of agents acting quite
serotoninergic 5-HT1a autoreceptors, by regulating the selectively at NR2B,69 NR2A, AMPA, and mGluR570
release of serotonin as well as dopamine formed from receptor site. But many of these compounds cannot be

Movement Disorders, Vol. 19, No. 7, 2004


748 G. LINAZASORO

used in humans, because of side effects, or when tested, which produces different compensatory modifications in
have produced poor results. For instance, ifenprodil and distinct nuclei.
Ro 25-6981 behaved remarkably in MPTP monkeys, but We have shown that the systemic administration of the
ifenprodil was not efficacious in PD patients.71 Remace- CB1 agonist WIN55512 to parkinsonian rats produced a
mide was able to potentiate the effects of L-dopa in decrease in the firing rate of STN neurons, which was
animal models,72 but when used in PD patients already abolished by injection of the antagonist SR141716A.85
taking L-dopa, no significant effect was seen in parkin- An opposite effect was observed in intact rats, a finding
sonism or dyskinesia.73 Riluzole is another NMDA an- also obtained with dopamine agonists.85,86 Moreover,
tagonist that has been given for motor complications WIN55512 did not alter the neurophysiological charac-
after showing good effect in animals,74 and a 30% re- teristics of nigral dopaminergic neurons. These data sug-
duction in dyskinesia in a small trial of 6 parkinsonian gest that CB1 agonists may exert antiparkinsonian ac-
patients,75 but a large, prospective multicenter trial has tions, whereas antagonists may ameliorate L-dopa–
been discontinued because lack of efficacy was detected induced dyskinesia. By contrast, results of other
in an interim analysis. Dextromethorphan has produced experiments81 and of the only clinical trial to date, failed
confusing results in humans, in part due to its narrow to support our data.87 In this study, nabilone, a cannabi-
therapeutic window,76 lamotrigine was ineffective in a noid agonist was given to 9 patients with L-dopa–induced
double-blind trial77 but amantadine has been shown to dyskinesia in a double-blind, randomized, placebo-con-
ameliorate L-dopa–induced dyskinesia in acute and long- trolled, crossover study.87 Nabilone produced a mean
term studies,78,79 an effect that has been attributed to its 20% reduction in the dyskinesia score without modifying
antiglutamatergic properties, although amantadine is a parkinsonian symptoms and signs; it was particularly
“dirty” drug with many other actions. effective in reducing diphasic dyskinesia and, to a lesser
New antiglutamatergic agents are at a very early extent, off period dyskinesia. A possible explanation of
stages of development, but they have to overcome all the these contradictory results is that the antidyskinetic ef-
problems already mentioned before coming into clinical fect of nabilone might be related to an action on the GPe
use. To solve these problems, we have to increase our CB1 receptors.81,87 Again, the apparently consistent re-
knowledge at the molecular level about the involvement sults obtained in animals could not be replicated in
of glutamate in the pathophysiology of parkinsonism and humans; clearly further hypotheses need to be formu-
dyskinesia, which would led to the development of se- lated and ideally tested on animals before continuing
lective drugs with a positive risk/benefit profile. with clinical studies.

Opioid Receptor Agonists and Antagonists


Cannabinoids Agonists and Antagonists
Opioid peptides are involved in many aspects of basal
The levels of cannabinoid receptors 1 (CB1) in the ganglia function, as can be inferred from the wide dis-
basal ganglia are the highest in the brain, comparable to tribution of opioid receptors within these structures.
dopamine receptors.80,81 This distribution of receptors is Changes in the expression of striatal opioids (enkephalin
consistent with their profound effects on motor function, and dynorphin) colocalised with dopamine receptors on
besides their well known involvement in other functions, the GABAergic projection neurons occur in very early
such as cognition and pain. Their main effect is to inhibit phases of PD and further changes are induced by the
the release of the neurotransmitter, by blocking the up- treatment with L-dopa.88 But problems mentioned for
take of GABA.80,81 The motor effects of cannabinoids other systems are applicable here: there are different
are complex.80 – 83 Cannabinoids activate movement opioid receptor types located on different nuclei, of
when microinjected into the striatum or into the SNpr, which stimulation or blockade result in variable effects
and movement is inhibited when they are injected di- on motor behaviour. Opioids act as neuromodulators
rectly into the STN or the GPe. Recent studies in animal influencing the activity of dopamine and GABA neurons
models suggest that CB1 receptor antagonists could in different directions, depending on the nucleus and
prove useful in the treatment of parkinsonian symptoms receptor involved (reviewed in Henry and Brotchie89).
and L-dopa–induced dyskinesia and cannabinoid agonists Immunocytochemical and PET studies have demon-
may be of great value as antidyskinetic agents.80,81,84 strated increased opioid neuropeptide transmission in
This suggestion has to be accepted with great caution animals and patients with PD and L-dopa–induced dys-
because the final effect of a systemically administered kinesia.88 –90 Thus, opioid receptor antagonists have been
CB1 agonist or antagonist will depend on the clinical studied as potential new treatment for dyskinesia.89 Mu
situation (not treated vs chronically treated patients), (cyprodime) and delta (naltrindole) selective receptor

Movement Disorders, Vol. 19, No. 7, 2004


FAILURES OF SYMPTOMATIC TREATMENTS FOR PD 749

antagonists, have been shown to be efficacious in im- preferable to live without basal ganglia than with abnor-
proving dyskinesia in primed MPTP-lesioned primates.89 mally functioning basal ganglia (“No news is better than
Very little effect has been observed in the few patients bad news”). This is a clear example of adaptation to new
who have been treated with naloxone91 and naltrexone.92 experimental data; further adaptations will be needed to
Enadoline, a kappa opioid agonist, has been demon- explain new findings. (2) Surgical techniques do not
strated to exert an anti-akinetic action and to potentiate produce cognitive adverse effects. This idea relies on the
the motoric effects of L-dopa in reserpinized rats.93 To well-demonstrated segregation of functions across the
date, the only data on human beings were obtained with basal ganglia-thalamo-cortical circuits.9,10,95 Usually, as-
spiradoline, a kappa agonist with lower specificity and sociative and limbic functions of nuclei conforming the
potency than enadoline (see Hughes and colleagues93). basal ganglia are related to the activity of neurons lo-
No antiparkinsonian effects were found in this study, but cated on their ventral part, which are connected with
the interpretation of the results were difficult because cortical areas functionally related to cognitive and emo-
dose-limiting behavioural abnormalities appeared. Thus, tional functions. Because lesions and electrodes are
as is the case with many of the drugs reviewed, a better placed on motor portions of these nuclei, only motor
understanding of the function of the opioid system and effects are expected. And, in fact, the motor effects of
the synthesis of more selective compounds is needed. these procedures are dramatic.94 However, their impact
on cognitive and emotional functions are controversial:
SURGICAL TREATMENTS FOR PD PATIENTS whereas some authors have failed to find major effects,
WITH MOTOR COMPLICATIONS others have found concerning adverse effects.96 Some
factors seem to influence the appearance of these adverse
Pallidotomy and Deep Brain Stimulation effects, such as the age of patients, the existence of prior
Current surgical procedures are based on the existence subclinical cognitive disturbances, or abnormalities on
of hyperactivity at the level of the subthalamopallidal magnetic resonance imaging.97 Cognitive and emotional
pathway,9,10 a feature that is considered to be the phys- functions have not been adequately addressed in the
iological hallmark of the parkinsonian state. Abolition of larger studies conducted to date and the true frequency of
this hyperactivity by lesioning or inhibiting neural activ- cognitive and emotional disturbances may be underesti-
ity by implanting electrodes in the STN or the GPi results mated.98
in a dramatic improvement of parkinsonism in animal
models of the disease.9,10 Lesioning procedures, such as Transplantation
pallidotomy and, to a lesser extent subthalamotomy, and The success of cell replacement for the treatment of
pallidal and subthalamic DBS have been used in parkin- PD is based on two hypotheses: first, the predominant
sonian patients,94 and as predicted, major improvements symptoms develop because of the dysfunction or loss of
in motor function have been obtained. However, several the dopaminergic neurons in the nigrostriatal pathway;
aspects are not totally understood: (1) pallidal surgery second, dopaminergic neurons grafted into the dopam-
abolishes L-dopa–induced dyskinesias. This finding is ine-deficient striatum can replace the neurons lost as a
probably the most striking effect of any type of surgical result of the disease process and can reverse, at least in
procedure into the GPi and is hard to reconcile with part, the major symptoms of the disease.99 Extensive
previous models of the organization of the basal ganglia- animal studies have demonstrated that immature dopa-
thalamo-cortical circuits in which only firing rate data mine neurons survive, re-establish dopaminergic inner-
were considered.9 According to these models, any type vation when grafted into the denervated striatum, and
of dyskinesia is associated with a reduction in the activ- restore baseline dopamine synthesis and release.99 Re-
ity of the subthalamopallidal pathway, and a further sults of a series of small open-label trials with carefully
reduction of this activity will lead to the appearance or selected patients indicated that human fetal dopaminer-
worsening of such dyskinesias, a finding that is not gic neurons can survive and function in the striatum of
compatible with surgical experience. New models have patients with PD and most of these patients showed a
incorporated changes in the pattern of neuronal activity remarkable and long-lasting clinical improvement. Nev-
and neuronal synchronization as crucial features of the ertheless, two double-blind sham-surgery controlled tri-
organization of these circuits.95 Thus, abolition of abnor- als have failed to reproduce predicted results; there has
mal neuronal patterns and “normalisation” of existent been only modest efficacy and a significant number of
abnormal rhythms in these nuclei in PD patients with patients have developed severe off-state dyskinesias after
dyskinesias can be the underlying basis of the improve- transplantation.100,101 Paradoxically, experiments in
ment in L-dopa–induced dyskinesia. In one sense, it is 6-OHDA–lesioned rats with L-dopa–induced dyskinesia

Movement Disorders, Vol. 19, No. 7, 2004


750 G. LINAZASORO

have shown that grafting dopaminergic fetal cells into apy. For instance, the demonstration of the existence of
the striatum produced an improvement in motor behav- a neuronal hyperactivity in the STN and GPi/SNpr in
iour, including dyskinesia, which was accompanied by a experimental models of PD led to the hypothesis that
reversal of the molecular mechanisms theoretically re- reducing or abolishing this activity would result in an
sponsible for dyskinesia.4 Once again, a substantial dis- improvement of parkinsonian cardinal symptoms and
cordance between animal data and human reality is ev- signs. And this strategy was the origin of current DBS
ident. For this reason, it would be interesting to perform techniques, which have changed the quality of life of
transplantation experiments in animals with dyskinesias many patients with advanced PD. However, attempts to
before clinical studies. Curiously, transplantation of test other hypotheses have failed. Hence, limitations of
other type of cells such as human pigmentary epithelial the models, problems of current understanding of func-
retinal cells102 or the direct striatal infusion of trophic tional organization of basal ganglia, and questions re-
factors, such as glial-derived neurotrophic factor lated to the pharmacological properties of the drugs
(GDNF),103 have led to remarkable improvements in a tested can explain recent pitfalls (see Table 2). For in-
few PD patients. As was the case with fetal neural stance, current animal models are usually models of an
transplantation, clinical benefit is accompanied by incre- acute dopaminergic deficit, although more chronic intox-
ments in the uptake of fluorodopa in PET studies. No off ication protocols have been developed.2,6,104 Of interest,
period dyskinesias have been reported in these patients,
the effects of certain drugs are quite different in monkeys
but many questions still remain about the exact mecha-
with acute or chronic MPTP parkinsonism.105 However,
nism of action and long-term effects of these procedures,
they are not totally comparable with PD, which is a
and double-blind, sham-controlled studies with these
chronic, slowly progressive, and “more than dopaminer-
cells and GDNF are now planned.
gic” disease. At present, there is no evidence to show that
SOME CLUES TO UNDERSTANDING chronic administration of low dose of any toxic sub-
THERAPEUTIC FAILURES stance mimics the progressive degenerative nature of PD.
What are the main reasons for these pitfalls and con- Therefore, animal models offer a static rather than a
tradictory results? Likely, this is a multifactorial prob- dynamic perspective of the disease. Moreover, the to-
lem. Both the 6-OHDA rat and the MPTP primate are pography of neuronal loss within the SNpc is different in
quite adequate models of the disease.3,6 In fact, all do- animals acutely lesioned or intoxicated from that in
paminomimetic drugs exert very stereotyped effects in PD,106,107 a difference less marked in chronic animal
these models that can be easily reproduced and that models of PD.2,104 Finally, the clinical picture of PD
predict their later effectiveness. Nonetheless, it should be varies greatly from patient to patient, whereas the behav-
recognized that these models are not perfect, and these iour of parkinsonian rats or monkeys is very homoge-
imperfections could explain some recent failures. More- neous.
over, currently, we have an incomplete understanding of The effects of L-dopa and other antiparkinsonian drugs
basal ganglia functional organization on which to base may be totally different, depending on the severity of
preclinical investigation of new treatments.9,10,94,95 Nev- nigrostriatal denervation. Rodent and primate models of
ertheless, it has produced some major advances in ther- moderate striatal dopaminergic deficiency are avail-

TABLE 2. Some problems with current methods


A) Related to animal models and assessment methods.
Animals are models of an acute, stable, severe, and pure dopaminergic deficiency.
Animals display a homogeneous behavioural disturbance.
Genetic factors, aging-related factors, etc., cannot be properly addressed in animals.
Cognitive, emotional and other nondopaminergic signs are difficult to evaluate in animals.
B) Related to limited knowledge of basal ganglia function.
Incomplete knowledge of the distribution and function of many receptors (and, perhaps, neurotransmitter systems) within the basal ganglia.
C) Related to treatment strategies.
Incomplete knowledge of the mechanism of action of certain drugs (i.e., levodopa).
Lack of drugs with complete selectivity for a given receptor in a given localization.
Action of drugs on receptors located outside the basal ganglia and involved in motor control or other functions.
Interactions between different neurotransmitters at the molecular level after the action of a drug.
Partial or dual effects of a drug are very difficult to evaluate.
Pharmacokinetic and pharmacodynamic characteristics of tested drugs.
Incomplete understanding of the mechanism of action of surgical therapies.

Movement Disorders, Vol. 19, No. 7, 2004


FAILURES OF SYMPTOMATIC TREATMENTS FOR PD 751

able.2,6,86,104 Partial lesion protocols provide ideal mod- To improve current symptomatic treatments with new
els to evaluate and predict the effects of some therapies drugs, cell lines in which test the effects of drugs on the
on the prevention of the development of motor compli- molecular mechanisms underlying parkinsonism and
cations and dyskinesia. For instance, working with the dyskinesia can be developed so that drugs can be tested
triple combination of L-dopa/carbidopa/entacapone, it before conducting the “motor” experiments in ani-
has been shown that q.i.d administration to marmosets mals.115 Finally, new technologies such as DNA mi-
with severe dopamine deficiency produces a maximal croarrays,116 RNA silencing or RNA interference and
antiparkinsonian benefit with minimal dyskinesia com- high-throughput screening of drugs may provide new
pared to L-dopa/carbidopa alone,108 but the difference is tools to develop more selective antiparkinsonian drugs
not so evident when the same drug regimens are admin- and to refine current models.
istered to marmosets with a moderate dopaminergic de-
nervation. We have also seen that the effects of L-dopa REFERENCES
and apomorphine on the neuronal activity of the STN is 1. Obeso JA, Linazasoro G, Gorospe A, et al. Complications asso-
more homogeneous in moderately 6-OHDA–lesioned ciated with chronic levodopa therapy in Parkinson’s disease. In:
rats.86 There is also controversy about the earlier use of Olanow CW, Obeso JA, editors. Beyond the decade of the brain.
Vol. 2. Kent: Wells Medical Ltd; 1997. p 11–36.
DBS techniques or cell therapy approaches. So it would 2. Tolwani RJ, Jakowec MW, Petzinger GM, et al. Experimental
be important to have data on animal with less-extensive models of Parkinson’s disease: insights from many models. Lab
nigral lesions. The influence of other factors such as the Anim Sci 1999;49:363–371.
3. Cenci MA, Whishaw IQ, Schallert T. Animal models of neuro-
genetic background, the effects of aging, and so on, logical deficits: how relevant is the rat? Nat Rev Neurosci 2002;
cannot be adequately addressed in these models. 3:574 –579.
PD is a multiple system disorder and the degenerative 4. Lee CS, Cenci MA, Sculzer M, Bjorklund A. Embryonic ventral
mesencephalic grafts improve levodopa-induced dyskinesia in a
process is not limited to the dopaminergic nigrostriatal rat model of Parkinson’s disease. Brain 2000;123:1365–1379.
pathway.109 Other cell populations containing different 5. Andersson M, Hilbertson A, Cenci MA. Striatal fosB expression
neurotransmitters, such as the noradrenergic locus cer- is causally linked with L-dopa-induced abnormal involuntary
movements and the associated upregulation of striatal prodynor-
uleus,110 the serotoninergic raphe nucleus, the cholin- phin mRNA in a rat model of Parkinson’s disease. Neurobiol Dis
ergic and glutamatergic pedunculopontine nuclei, and the 1999;6:461– 474.
cholinergic nucleus of Meynert are also affected. This 6. Jenner P. The contribution of the MPTP-treated primate model to
the development of new treatment strategies for Parkinson’s
pattern of involvement may account for the appearance disease. Parkinsonism and related disorders 2003;9:131–137.
of some motor and nonmotor symptoms and signs. The 7. Burns RS, Chiueh CC, Markey SP, et al. A primate model of
involvement of some of these nuclei may be very early, parkinsonism: selective destruction of dopaminergic neurons in
the pars compacta of the substantia nigra by N-methyl-4-phenyl-
earlier even than the SNpc damage.110 These degenera- 1,2,3,6-tetrahydropyridine. Proc Natl Acad Sci U S A 1983;80:
tive changes are not reproduced in current experimental 4546 – 4550.
models. 8. Burns RS, Markey SP, Phillips JM, Chiueh CC. The neurotoxic-
Most of the drugs tested act at the level of neurochem- ity of MPTP in the monkey and man. Can J Neurol Sci 1984;11:
166 –168.
ical systems, of which the relative contribution to the 9. DeLong MR. Primate models of movement disorders of basal
pathophysiology of motor abnormalities has not been ganglia origin. Trends Neurosci 1990;13:281–285.
totally elucidated. It is also unknown if there exists 10. Brotchie JM. The neural mechanisms underlying levodopa-in-
duced dyskinesia. Ann Neurol 2000;47(Suppl. 1):105–114.
always a human counterpart of a given receptor in a 11. Jaber M, Jones S, Giros B, Caron MG. The dopamine transporter:
given nucleus. For instance, the existence of dopamine a crucial component regulating dopamine transmission. Mov Dis-
receptors into the human STN has not been conclusively ord 1997;12:629 – 633.
12. Pearce RKB, Smith LA, Jackson MJ, et al. The monoamine
proven,111 the colocalization of D1 and D2 receptors in a reuptake blocker brasofensine reverses akinesia without dyskine-
proportion of striatal neurons112 is ignored, etc. sia in MPTP-treated and levodopa-primed common marmosets.
All these arguments clearly limit the predictive value Mov Disord 2002;5:877– 886.
13. Mailman R, Huang X, Nichols DE. Parkinson’s disease and D1
of animal studies. With the increased complexity of dopamine receptors. Curr Opin Invest Drugs 2001;2:1582–1591.
drugs acting on other neurotransmission systems, there is 14. Boyce S, Rupniak NM, Steventon MJ, Iversen SD. Differential
a still lower predictive value as illustrated by the case of effects of D1 and D2 agonists in MPTP-treated primates: func-
fetal transplantation.100,101 Furthermore, it is important to tional implications for Parkinson’s disease. Neurology 1990;40:
927–933.
make further efforts to better define the function of the 15. Pearce RK, Jackson M, Britton DR, et al. Actions of the D1
basal ganglia in health and disease, along with the de- agonists A-77636 and A-89929 on locomotion and dyskinesia in
velopment of alternative models of the disease.113,114 All MPTP-treated L-dopa primed common marmosets. Psychophar-
macology (Berl) 1999;142:51– 60.
these efforts should be completed with the implementa- 16. Tsui JK, Wolters EC, Peppard RF, Calne DB. A double-blind,
tion of thoroughly conducted development programs. placebo-controlled, dose-ranging study to investigate the safety

Movement Disorders, Vol. 19, No. 7, 2004


752 G. LINAZASORO

and efficacy of CY 208243 in patients with Parkinson’s disease. 36. Manson AJ. Iakovidou E, Lees AJ. Idazoxan is ineffective for
Neurology 1989;39:856 – 858. levodopa-induced dyskinesias in Parkinson’s disease. Mov Dis-
17. Taylor JR, Lawrence MS, Redmond DE, et al. Dihydrexidine, a ord 2000;15:336 –337.
full dopamine D1 agonist, reduces MPTP-induced parkinsonism 37. Rascol O, Arnulf I, Peyro-Saint PH, et al. Idazoxan, an alpha-2
in monkeys. Eur J Pharmacol 1991;199:389 –391. antagonist, and L-DOPA-induced dyskinesias in patients with
18. Blanchet PJ, Fang J, Gillespie M, et al. Effects of the full Parkinson’s disease. Mov Disord 2001;16:708 –713.
dopamine D1 receptor agonist dihydrexidine in Parkinson’s dis- 38. Fox SH, Henry B, Hill MP, et al. Neural mechanisms underlying
ease. Clin Neuropharmacol 1998;21:339 –343. peak-dose dyskinesia induced by levodopa ans apomorphine are
19. Shiosaki K, Jenner P, Asin KE, et al. ABT-431: the diacetyl distinct: evidence from the effects of the alpha-2 adrenoceptor
prodrug of A-86929, a potent and selective dopamine D1 receptor antagonist idazoxan. Mov Disord 2001;16:642– 650.
agonist: in vitro characterization and effects in animal models of 39. Pact V, Giduz T. Mirtazapine treats resting tremor, essential
Parkinson’s disease. J Pharmacol Exp Ther 1996;276:150 –160. tremor and levodopa-induced dyskinesias. Neurology 1999;53:
20. Rascol O, Blin O, Thalamas C, et al. ABT-431, a D1 receptor 1154.
agonista prodrug, has efficacy in Parkinson’s disease. Ann Neurol 40. Calon F, Goulet M, Blanchet PJ, et al. Levodopa or D2 agonist
1999;45:736 –741. induced dyskinesia in MPTP monkeys: correlation with changes
21. Rascol O, Nutt J, Blin O, et al. Induction by D1 receptor agonista in dopamine and GABAa receptors in the striatopallidal complex.
ABT-431 of dyskinesia similar to levodopa in patients with Brain Res 1995;680:43–52.
Parkinson’s disease. Arch Neurol 2001;58:249 –254.
41. Calon F, Morissette M, Goulet M, et al. Chronic D1 and D2
22. Gulwadi AG, Korpinen CD, Mailman RB, et al. Dinapsoline:
dopaminomimetic treatment of MPTP-denervated monkeys: ef-
characterization of a D1 dopamine receptor agonist in a rat model
fects on basal ganglia GABA (A)/benzodiazepine receptor com-
of Parkinson’s disease. J Pharmacol Exp Ther 2001;296:338 –
plex and GABA content. Neurochem Int 1999;35:81–91.
344.
23. Brücke T, Bankeiwicz K, Harvey-White J, Kopin I. The partial 42. Calon F, Di Paolo T. Levodopa response motor complications:
dopamine agonist terguride in the MPTP-induced hemiparkinso- GABA receptors and preproenkephalin expression in human
nian monkey model. Eur J Pharmacol 1988;148:445– 448. brain. Parkinsonism Relat Disord 2002;8:449 – 454.
24. Baronti F, Mouradian MM, Conant KE, et al. Partial dopamine 43. Calon F, Morisette M, Rajput AH, et al. Changes in GABA
agonist therapy of levodopa-induced dyskinesias. Neurology receptors and dopamine turnover in the postmortem brains of
1992;42:1241–1243. parkinsonians with levodopa-induced motor complications. Mov
25. Nichols NF, Cimini MG, Haas JV, et al. PNU-96391A Disord 2003;18:241–253.
(OSU6162) antagonizes the development of behavioural sensiti- 44. Lozano AM, Carella F. Physiologic studies in the human brain in
zation induced by dopamine agonists in a rat model for Parkin- movement disorders. Parkinsonism Relat Disord 2002;8:455–
son’s disease. Neuropharmacology 2002;43:817– 824. 458.
26. Bézard E, Ferry S, Mach U, et al. Attenuation of levodopa- 45. Lees AJ, Shaw KM, Stern GM. Baclofen in Parkinson’s disease.
induced dyskinesia by normalizing dopamine D3 receptor func- J Neurol Neurosurg Psychiatry 1978;41:707–708.
tion. Nat Med 2003;9:762–777. 46. Turjanski N, Lees AJ. Gamma vinyl GABA in the treatment of
27. Silverdale MA, Millan MJ, Crossman AR, Brotchie JM. Dopa- levodopa-induced dyskinesias. J Neurol Neurosurg Psychiatry
mine D3 receptor blockade, and not stimulation, is associated 1992;55:413.
with anti-parkinsonian activity in the MPTP-lesioned, non-human 47. Ziegler M, Fournier V, Bathien N, et al. Therapeutic response to
primate model of Parkinson’s disease. Soc Neurosci Abstr 2002: progabide in neuroleptic- and L-dopa-induced dyskinesias. Clin
S63.001. Neuropharmacol 1987;10:238 –246.
28. Richardson PJ, Kase H, Jenner PG. Adenosine A2a receptor 48. Price PA, Parkes JD, Marsden CD. Sodium valproate in the
antagonists as new agents for the treatment of Parkinson’s dis- treatment of levodopa-induced dyskinesias. J Neurol Neurosurg
ease. Trends Pharmacol 1997;18:338 –344. Psychiatry 1978;41:702–706.
29. Hauser R, Hubble JP, Truong DD and the Istradephylline US-001 49. Reeves AL, So EL, Sharbrough FW, Krahn LE. Movement dis-
Study Group. Randomized trial of the Adenosine A2A receptor orders associated with the use of gabapentin. Epilepsia 1996;37:
antagonist istradephylline in advanced PD. Neurology 2003;61: 988 –990.
297–303. 50. Van Blercom N, Lasa A, Verger K, et al. Effects of gabapentin on
30. Sherzai A, Bara-Jiménez W, Sherzai A, Dimitrova T, et al. the motor response to levodopa: a double-blind, placebo-con-
Adenosine A2a antagonist treatment of Parkinson’s disease. Neu- trolled, crossover study in patients with complicated Parkinson’s
rology 2003;61:293–296. disease. Clin Neuropharmacol 2004 (in press).
31. Mally J, Stone TW. Theophylline, a not selective adenosine
51. Olson WL, Gruenthal M, Mueller ME, Olson WH. Gabapentin
antagonist in Parkinson’s disease. J Pharm Pharmacol 1994;46:
for parkinsonism: a double-blind, placebo-controlled, crossover
515–517.
trial. Am J Med 1997;102:60 – 66.
32. Kulisevsky J, Barbanoj M, Gironell A, et al. A double-blind
crossover, placebo-controlled study of the adenosine A2A antag- 52. Chaná P, de Marinis A, Barrientos N. Gabapentin and motor
onist theophylline in Parkinson’s disease. Clin Neuropharmacol fluctuations in Parkinson’s disease. Mov Disord 1997;12:608.
2002;25:25–31. 53. Ondo W, Hunter C. Flumazenil, a GABA antagonist may im-
33. Henry B, Fox SH, Peggs D, et al. The alpha 2-adrenergic receptor prove features of Parkinson’s disease. Mov Disord 2003;18:683–
antagonist idazoxan reduces dyskinesia and enhances anti-parkin- 684.
sonian actions of levodopa in the MPTP-lesioned primate model 54. Fox SH, Brotchie JM. 5-HT2C receptor binding is increased in
of Parkinson’s disease. Mov Disord 1999;14:744 –753. the substantia nigra pars reticulata in Parkinson’s disease. Mov
34. Hill MP, Savola JM, Merivouri H, et al. Potential applications of Disord 2000;15:1064 –1069.
the alpha2 adrenergic antagonist, JP-1730, in reducing the prob- 55. Tecott LH, Sun LM, Akana SF, et al. Eating disorder and epilepsy
lem of L-dopa-induced motor fluctuations in Parkinson’s disease: in mice lacking 5-HT2c serotonin receptor. Nature 1995;374:
studies in the rodent and primate models of Parkinson’s disease. 542–546.
Mov Disord 2002;17(Suppl. 5):S71. 56. Fox SH, Moser B, Brotchie JM. Behavioural effects of 5-HT2C
35. Gómez Mancilla B, Bédard PJ. Effects of nondopaminergic drugs receptor antagonism in the substantia nigra zona reticulata of the
on L-dopa-induced dyskinesias in MPTP monkeys. Clin Neurop- 6-hydroxydopamine-lesioned rat model of Parkinson’s disease.
harmacol 1993;16:418 – 427. Exp Neurol 1998;151:35– 49.

Movement Disorders, Vol. 19, No. 7, 2004


FAILURES OF SYMPTOMATIC TREATMENTS FOR PD 753

57. Eberle-Wang K, Lucki I, Chesselet MF. A role for the subtha- 79. Verhagen Metman L, Del Dotto P, LePoole K, et al. Amantadine
lamic nucleus in 5-HT2c-induced oral dyskinesia. Neuroscience for levodopa-induced dyskinesias: a 1-year follow-up study. Arch
1996;72:117–128. Neurol 1999;56:1383–1386.
58. Meco G, Marini S, Lestingi L, et al. Controlled single-blind 80. Sañudo-Peña MC, Tsou K, Walker JM. Motor actions of canna-
cross-over study of ritanserin and placebo in L-dopa-induced binoids in the basal ganglia output nuclei. Life Sci 1999;65:703–
dyskinesias in Parkinson’s disease. Curr Ther Res 1988;43:262– 713.
270. 81. Brotchie JM. CB1 cannabinoid receptor signalling in Parkinson’s
59. Durif F, Vidailhet M, Bonnet AM, et al. Levodopa-induced disease. Curr Opin Pharmacol 2003;3:54 – 61.
dyskinesias are improved by fluoxetine. Neurology 1995;45: 82. Sañudo-Peña MC, Walker JM. Effects of intrapallidal cannabi-
1855–1858. noids on rotational behaviour in rats: interactions with the dopa-
60. Richard IH, Maughn A, Kurlan R. Do serotonin reuptake inhib- minergic system. Synapse 1998;28:27–32.
itor antidepressants worsen Parkinson’s disease? A retrospective 83. Sañudo-Peña MC, Walker JM. Role of the subthalamic nucleus
case series. Mov Disord 1999;14:155–157. on cannabinoid actions in the substantia nigra of the rat. J Neu-
61. Kulisewski J, Campos V, Martı́nez P, Sancho J. Treatment of rophysiol 1997;77:1635–1638.
depression with sertraline does not worsen motor symptoms in 84. Sañudo-Peña MC, Patrick SL, Khen S, et al. Cannabinoids effect
Parkinson’s disease. Neurology 2003;60:A291. in basal ganglia in a rat model of Parkinson’s disease. Neurosci
62. Bibbiani F, Oh JD, Chase TN. Serotonin 5-HT1A agonist im- Lett 1998;248:171–174.
proves motor complications in rodent and primate parkinsonian 85. Ruiz Ortega JA, Linazasoro G, Bilbao G, et al. Effect of WIN
models. Neurology 2002;58:P05.092. 55,512 on subthalamic nucleus and substantia nigra zona com-
63. Bara W, Sherzai A, Bibbiani F, et al. 5-HT1a agonist effects in pacta neurons in anaesthetized rats (in preparation).
advanced Parkinson’s disease. Neurology 2003;60:A507. 86. Ruiz Ortega JA, Linazasoro G, Ugedo L. Effects of systemic and
64. Linazasoro G. Mecanismos glutamatérgicos en la enfermedad de intrasubthalamic dopaminergic drugs administration on the neu-
Parkinson. Neurologia 2000(Suppl. 6):21–30. rophysiological activity of the subthalamic nucleus of rats with
65. Klockgether T, Turski L. Toward an understanding of the role of moderate and severe lesions of the nigrostriatal pathway. Eur
glutamate in experimental parkinsonism: agonist-sensitive sites in J Neurosci (submitted).
the basal ganglia. Ann Neurol 1993;34:585–593. 87. Sieradzan KA, Fox SH, Hill MP, et al. Cannabinoids reduce
66. Kaur S, Starr MS. Differential effects of intrastriatal and intrani- levodopa-induced dyskinesias in Parkinson’s disease: a pilot
gral injections of glutamate antagonists on motor behaviour in the study. Neurology 2001;57:2108 –2111.
reserpine-treated rat. Neuroscience 1997;76:345–354. 88. Jenner P. Factors influencing the onset and persistence of dyski-
67. Ikonomidou C, Turski L. Why did NMDA receptor antagonists nesia in MPTP-treated primates. Ann Neurol 2000;47(Suppl.
fail clinical trials for stroke and traumatic brain injury? Lancet 1):90 –104.
Neurol 2002;1:383–386.
89. Henry B, Brotchie JM. Potential of opioid antagonists in the
68. Chase TN, Oh JD. Striatal mechanisms and pathogenesis of
treatment of levodopa-induced dyskinesias in Parkinson’s dis-
parkinsonian signs and motor complications. Ann Neurol 2000;
ease. Drugs Ageing 1996;9:149 –158.
47(Suppl. 1):S122–S130.
90. Piccini P, Weeks RA, Brooks DJ. Alterations in opioid receptor
69. Nash JE, Brotchie JM. Characterization of striatal NMDA recep-
binding in Parkinson’s disease patients with levodopa-induced
tors involved in the generation of parkinsonian symptoms: intra-
dyskinesias. Ann Neurol 1997;42:720 –726.
striatal microinjection studies in the 6-OHDA-lesioned rat. Mov
Disord 2002;17:455– 466. 91. Sandyk R, Snider SR. Naloxone treatment of L-dopa-induced
70. Hill MP, McGuire SG, Crossman AR, et al. The mGluR5 receptor dyskinesias in Parkinson’s disease. Am J Psychiatry 1986;143:
antagonist SIB-1830 reduces levodopa-induced dyskinesia in the 118.
MPTP-lesioned primate model of Parkinson’s disease. Soc Neu- 92. Manson AJ, Katzenschlager R, Hobart J, et al. High dose nal-
rosci Abstr 2001;25:200. trexone for dyskinesias induced by levodopa. J Neurol Neurosurg
71. Montastruc JL, Rascol O, Senard JM, et al. A pilot study of Psychiatry 2001;70:554 –556.
N-methyl-D-aspartate (NMDA) antagonist in Parkinson’s dis- 93. Hughes NR, McKnight AT, Woodruff GN, et al. Kappa-opioid
ease. J Neurol Neurosurg Psychiatry 1992;55:630 – 631. receptor agonists increase locomotor activity in the monoamine-
72. Greenamyre JT, Eller RV, Zhang Z, et al. Antiparkinsonian depleted rat model of parkinsonism. Mov Disord 1998;13:228 –
effects of remacemide hydrochloride, a glutamate antagonist, in 233.
rodent and primate models of Parkinson’s disease. Ann Neurol 94. Krack P, Hamel W, Mehdorm HM, Deuschl G. Surgical treatment
1994;35:655– 661. of Parkinson’s disease. Curr Opin Neurol 1999;12:417– 425.
73. Parkinson Study Group. A randomized, controlled trial of rema- 95. Vitek JL. Physiology of hypokinetic and hyperkinetic movement
cemide for motor fluctuations in Parkinson’s disease. Neurology disorders: model for dyskinesia. Ann Neurol 2000;47(Suppl. 1):
2001;56:455– 462. S131–S140.
74. Starr MS, Starr BS, Kaur S. Stimulation of basal and L-dopa- 96. Van Horn G, Schiess MC, Soukup VM. Subthalamic deep brain
induced motor activity by glutamate antagonists in animal models stimulation: neurobehavioural concerns. Arch Neurol 2001;58:
of Parkinson’s disease. Neurosci Biobehav Rev 1997;21:437– 1205–1206.
446. 97. Grupo de estudio de estimulación cerebral profunda en la enfer-
75. Merims D, Ziv I, Djaldetti R, et al. Riluzole for levodopa-induced medad de Parkinson. Estimulación palidal y subtalámica en la
dyskinesias in advanced Parkinson’s disease. Lancet 1999;353: enfermedad de Parkinson: Enseñanzas de los malos resultados.
1764 –1765. Neurologia 2001;16:298 –302.
76. Vergahen Metman L, Blanchet PJ, van den Munckhof P, et al. A 98. Linazasoro G. Subthalamic deep brain stimulation for advanced
trial of dextromethorphan in parkinsonian patients with motor Parkinson’s disease: all that glitters is not gold. J Neurol Neuro-
response complications. Mov Disord 1998;13:414 – 417. surg Psychiatry 2003;74:827.
77. Shinotoh H, Vingerhoets FJG, Lee CS, et al. Lamotrigine trial in 99. Dunnett SB, Björklund A. Prospects for new restorative and
idiopathic parkinsonism: a double-blind, placebo-controlled, neuroprotective treatments in Parkinson’s disease. Nature 1999;
crossover study. Neurology 1997;48:1282–1285. 399(Suppl.):A32–A39.
78. Vergahen Metman L, Del Dotto P, van den Munckhof P, et al. 100. Freed CR, Greene PE, Breeze RE, et al. Transplantation of
Amantadine as treatment for dyskinesias and motor fluctuations embryonic dopamine neurons for severe Parkinson’s disease.
in Parkinson’s disease. Neurology 1998;50:1323–1326. N Eng J Med 2001;344:710 –719.

Movement Disorders, Vol. 19, No. 7, 2004


754 G. LINAZASORO

101. Olanow CW, Goetz CG, Kordower JH, et al. A double-blind and avoids dyskinesia in the MPTP model of Parkinson’s disease.
controlled trial of bilateral fetal nigral transplantation in Parkin- Neurology 2002;58:P05.096.
son’s disease. Ann Neurol 2003;54:403– 414. 109. Dickson DW. Neuropathology of parkinsonian disorders. In: To-
102. Watts RL, Raiser CD, Stover NP, et al. Stereotaxic intrastriatal losa E, Jankovic J, editors. Parkinson’s disease and movement
implantation of retinal pigment epithelial cells attached to micro- disorders. 4th ed. New York: Lippincott Williams and Wilkins;
carriers in advanced Parkinson’s disease patients: a pilot study in 2002. p 256 –270.
six patients. Parkinsonism Relat Disord 2001;7:S87. 110. Rye D, DeLong MR. Time to focus on the locus. Arch Neurol
103. Gill SS, Patel NK, Hotton GR, et al. Direct brain infusion of 2003;60:320.
glial-derived neurotrophic factor in Parkinson disease. Nat Med 111. Augood SJ, Hollingsworth ZR, Standaert DG, et al. Localization
2003;9:589 –595. of dopamine markers in the human subthalamic nucleus. J Comp
104. Bezard E, Imbert C, Gross CE. Experimental models of Parkin- Neurol 2000;421:247–255.
son’s disease: from the static to the dynamic. Rev Neurosci
112. Aizman O, Brismar H, Uhlén P, et al. Anatomical and physio-
1998;9:71–90.
logical evidence for D1 and D2 dopamine receptor colocalization
105. Bezard E. Neuroprotection for Parkinson’s disease: a call for
in neostriatal neurons. Nat Neurosci 2000;3:226 –230.
clinically driven experimental deign. Lancet Neurol 2003;2:393.
106. Pifl C, Schingnitz G, Hornykiewicz O. The neurotoxin MPTP 113. Beal MF. Experimental models of Parkinson’s disease. Nat Rev
does not reproduce in the rhesus monkey the interregional pattern Neurosci 2001;2:325–332.
of striatal dopamine loss typical of human idiopathic Parkinson’s 114. Dawson TM. New animal models for Parkinson’s disease. Cell
disease. Neurosci Lett 1988;92:228 –233. 2000;101:115–118.
107. Kish SH, Shannak K, Hornykiewicz O. Uneven pattern of dopa- 115. Gorba T, Allsopp TE. Pharmacological potential of embryonic
mine loss in the striatum of patients with idiopathic Parkinson’s stem cells. Pharmacol Res 2003;47:269 –278.
disease. N Eng J Med 1988;318:876 – 880. 116. Sturla LM, Fernández-Teijeiro A, Pomeroy SL. Application of
108. Jenner P, Al-Barghoulthy G, Smith L, et al. Initiation of entaca- microarrays to neurological disease. Arch Neurol 2003;60:676 –
pone with levodopa further improves antiparkinsonian activity 683.

Movement Disorders, Vol. 19, No. 7, 2004

You might also like