1. journal 1- levodopa-indvvvuced dyskinesias.pdf

10
J Neurol (2008) 255 [Suppl 4]:32–4 I 10.1007 s00415-008-4006-    J    O    N     4    0    0    6 Francesca Del Sorbo Alberto Albanese Levodopa-induced dyskinesias and the r management Why do PD patients develop dyskinesias? Following a period of stable response to dopaminer- gic medication (the “honeymoon” period), Parkinson’s disease (PD) patients gradually develop two progres- sive clinical phenomena requiring changes in the clin- ical management: motor fluctuations and dyskinesias. These two phenomena are intrinsically interconnected, but often require opposite treatment attitudes, because increases of dopaminergic treatment often improve fluctuations, but worsen dyskinesias. A possible solution to this dilemma resides in the administration of addi- tional anti-dyskinetic medication; unfortunately, how- ever, drugs with enough anti-dyskinetic potency have not yet been developed. Epidemiology Dyskinesias are a central side effect of dopaminergic therapy and represent a major clinical problem in the anagement of patients with PD. Early studies per- ormed before the levodopa era did not describe the resence of motor fluctuation or dyskinesias [32]. Dys- inesias were first observed following the introduction of levodopa monotherapy. In his seminal paper on le- vodopa treatment of PD, Cotzias [20] first observed “the F. Del Sorbo · A. Albanese ( ) Fondazione IRCCS Istituto Neurologico Carlo Besta Via G. Celoria, 11 20133 Milano, Italy Tel.: +39-02/2394-2552 Fax: +39-02/2394-2539 E-Mail: [email protected] A. Albanese Univ ersità Cattolica del Sacro Cuore Milano, Italy of disability. Their prec ise etiolog y is still poorly understood, althou gh it is recognized that dopaminer- gic pre-synaptic and post-synap- tic mechanisms are involved to- gether with extra-dopaminergic factors. The phenomenology of dyskinesias encompasses a vari- able mixture of two prevalent fea- tures: dystonia and chorea. We have studied their time course follow- ing a single acute levodopa chal- lenge and have found that dysto- nia occurs throughout the duration of the on period, whereas chorei- form movements occur only at the peak of therapeutic dopaminer- gic motor responses. This allows a schematic relationship to be drawn between a shor t duration motor re- sponse and the occurrence of dys- tonia and chorea. There is currently no satisfactory treatment for dys- kinesias. Managing the therapeutic window does not provide an ade- quate solution due to the appear- ance of a dyskinesia threshold dose that narrows the therapeutic mar- gin. High frequency stimulation of the subthalamic nucleus probab ly as some specific anti-dyskinetic action, but is limited by the small number of patients who are candi- dates for this treatment. Resear ch efforts are currently focused on the development of specific anti-dys- kinetic medications. Their avail- ability will certainly change the current clinical practice and will widen again the therapeutic win- dow of dopaminergic medications hat has now become too narrow. Key words  Parkinson’s disease · dyskinesias · levodopa · dopamine agonists Abstract  This paper reviews the epidemiology, pathophysiol- ogy, clinical features and ratio- nale for managing dyskinesias as- sociated with Parkinson’s disease. These are a common clinical prob- lem occurring in up to 90% of pa- tients and more frequently affect those with early-onset. Dyskinesias have a negative impact on quality of life and are an important cause

Upload: elisa-cynthia-ardaricka

Post on 04-Jun-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 1/10

J Neurol (2008) 255 [Suppl 4]:32–4I 10.1007 s00415-008-4006-

   J   O   N    4

   0   0   6

Francesca Del Sorbo

Alberto Albanese

Levodopa-induced dyskinesias

and the r management

Why do PD patients develop dyskinesias?

Following a period of stable response to dopaminer-

gic medication (the “honeymoon” period), Parkinson’sdisease (PD) patients gradually develop two progres-sive clinical phenomena requiring changes in the clin-ical management: motor fluctuations and dyskinesias.These two phenomena are intrinsically interconnected,but often require opposite treatment attitudes, becauseincreases of dopaminergic treatment often improvefluctuations, but worsen dyskinesias. A possible solutionto this dilemma resides in the administration of addi-tional anti-dyskinetic medication; unfortunately, how-

ever, drugs with enough anti-dyskinetic potency havenot yet been developed.

Epidemiology

Dyskinesias are a central side effect of dopaminergictherapy and represent a major clinical problem in the

anagement of patients with PD. Early studies per-ormed before the levodopa era did not describe theresence of motor fluctuation or dyskinesias [32]. Dys-inesias were first observed following the introduction

of levodopa monotherapy. In his seminal paper on le-vodopa treatment of PD, Cotzias [20] first observed “the

F. Del Sorbo · A. Albanese ()Fondazione IRCCS Istituto NeurologicoCarlo Besta

Via G. Celoria, 1120133 Milano, Italy Tel.: +39-02/2394-2552Fax: +39-02/2394-2539E-Mail: [email protected]

A. AlbaneseUniversità Cattolica del Sacro CuoreMilano, Italy 

of disability. Their precise etiologyis still poorly understood, althoughit is recognized that dopaminer-

gic pre-synaptic and post-synap-tic mechanisms are involved to-gether with extra-dopaminergicfactors. The phenomenology ofdyskinesias encompasses a vari-able mixture of two prevalent fea-tures: dystonia and chorea. We havestudied their time course follow-ing a single acute levodopa chal-lenge and have found that dysto-nia occurs throughout the durationof the on period, whereas chorei-form movements occur only at the

peak of therapeutic dopaminer-gic motor responses. This allows aschematic relationship to be drawnbetween a short duration motor re-sponse and the occurrence of dys-tonia and chorea. There is currentlyno satisfactory treatment for dys-

kinesias. Managing the therapeuticwindow does not provide an ade-quate solution due to the appear-

ance of a dyskinesia threshold dosethat narrows the therapeutic mar-gin. High frequency stimulation ofthe subthalamic nucleus probably

as some specific anti-dyskineticaction, but is limited by the smallnumber of patients who are candi-dates for this treatment. Researchefforts are currently focused on thedevelopment of specific anti-dys-kinetic medications. Their avail-ability will certainly change thecurrent clinical practice and will

widen again the therapeutic win-dow of dopaminergic medicationshat has now become too narrow.

Key words  Parkinson’s disease ·dyskinesias · levodopa · dopamineagonists

Abstract  This paper reviewsthe epidemiology, pathophysiol-ogy, clinical features and ratio-

nale for managing dyskinesias as-sociated with Parkinson’s disease.These are a common clinical prob-lem occurring in up to 90% of pa-tients and more frequently affectthose with early-onset. Dyskinesiashave a negative impact on qualityof life and are an important cause

Page 2: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 2/10

33

reversible induction of athetoid movements, which havebeen observed thus far only in patients with PD and onlywhen the therapeutic effect was significant”. Later ob-servations clearly identified dyskinesias as a side-effectof levodopa treatment [4, 5]. It was later recognized thatdyskinesias can also be caused by dopamine agonists,particularly if short-acting and of adequate potency [22,

35]. It became thus clear that any type of exogenous do-paminergic stimulation in a denervated striatum cancause dyskinesias [46], but pulsatile stimulation pro-duced by short-acting drugs (as typically occurs withlevodopa) particularly favors their occurrence [67]. Theexpression “levodopa-induced dyskinesias” is still cur-rently used, although levodopa is not the only drug caus-ing dyskinesias in PD patients [55].

Awareness of these levodopa complications has in-fluenced treatment strategies for PD. In modern series,dyskinesias occur later than in earlier series, with an ap-proximate risk of dyskinesias just short of 40 % follow-ing 4–6 years of levodopa monotherapy [1]. The intro-

duction of dopamine agonists since early disease stageshas further reduced and delayed the occurrence of dys-kinesias [80]. Based on published series, it has been esti-mated that PD patients treated for less than 5 years havean 11 % risk of dyskinesias, those treated for 6–9 yearshave a risk of 32 %, whereas patients treated for morethan 10 years have a risk of 89 % [23]. However, thesefigures are likely to decrease due to the widespread useof dopamine agonists, COMT and MAO inhibitors in theearly disease stages.

The three most important risk factors positively as-sociated with increased occurrence of dyskinesias areyounger age at disease onset [40, 79], longer disease du-

ration [70, 73] and longer duration of pulsatile dopami-nergic treatment (typically, levodopa) [23, 80]. The firsttwo factors are interrelated and almost all patients withearly onset disease [81] develop dyskinesias, whereasthey are less frequent in patients with late onset PD [43].PD patients with early disease onset have a high proba-bility to carry mutations for monogenic PD forms andtherefore early onset and genetic predisposition are twooverlapping and possibly interrelated risk factors. Otherrisk factors associated with increased risk of dyskinesiasare female gender [50, 86] and the occurrence of spe-

cific polymorphisms for dopamine receptors or dopa-ine transporters [29, 36, 68].Monogenic forms of PD offer a unique viewpoint to

appreciate the role played by genetic factors on the oc-currence of dyskinesias. Dyskinesias occur in all pa-ients with  parkin  disease (PARK2) [38], whereas only

84% of patients with PINK1 disease (PARK6) present

dyskinesias [2]. Parkin disease patients have an earlierean age at disease onset (24 years, compared to 31.6)

indicating that genetic make-up influences both vari-ables. The role played by genetic factors is better high-ighted in PINK1 patients by comparing those with one

utated allele to patients with two mutated alleles orwith wild-type PD [52]. Heterozygous carrier PINK1 pa-tients overlap to wild-type PD patients for age at onsetand disease duration, but have a higher incidence of dys-

inesias approaching that of PINK1 patients with bothalleles affected [52] (Table1).

Dyskinesias are an important cause of disability inPD and can reduce the patients’ quality of life. Mild, non-

disabling dyskinesias that occur in early disease stagesdo not impact significantly on quality of life [53]. How-ever, in patients with advanced disease, more severe dys-

inesias may cause poor quality of life, depression andincreased health care costs [71]. It has been observedthat peak-dose dyskinesias are better accepted by pa-tients than off-period morning dystonia and diphasicdyskinesias [15]. Hence, the patients tolerate on  statedyskinesias better than those occurring in the off or par-tial-off periods, because of the associated good motorstate and lack of pain or discomfort. The quality of lifeof patients with advanced PD is not only limited by theoccurrence of dyskinesias, but also by insufficient do-

aminergic medication secondary to a low dyskinesiathreshold [80] (see Fig.6).

Pathophys ology

Levodopa and dopamine agonists do not induce dyski-nesias in PD patients (or in experimental animals) that

ave never been previously treated with dopaminergicedications. The process by which the brain becomes

sensitized such that each administration of dopaminer-

Number o alleles a ected

um er o pat ents : ) : ) : ) : )

ge at onset years) . .

sease urat on years) . . .

um er o pat ents w t yston a at onset ) )

um er o pat ents w t motor uctuat ons ) ) ) )

Number o patients with dyskinesias (%) 151 (47 ) 1 (55 %) ** (9 %)

* Significantly different from wild-type patients (p 0.05) and from patients with one affected allele (p 0.05),** Significantly different from wild-type patients (p 0.01)

Table Comparison o dyskinesias in wild-type PD

patients (no allele a ected), PINK1 single heterozy-gote patients (1 allele a ected), and PINK 1 patientscarrying two mutate a e es. From [52

Page 3: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 3/10

34

gic therapy modifies the response to subsequent dopa-minergic treatments, is called priming. In this way, withrepeated treatment over time, the chance of dopaminer-gic stimulation eliciting dyskinesias is increased andonce dyskinesias have been established, their severityincreases. The priming process, which is responsible forthe insidious evolution of dyskinesias over time of treat-

ment, is associated with changes in receptors for dopa-mine or other neurotransmitters [27, 59].

Following priming, the development of dyskinesiaslargely depends on two additional factors, the pulsatileadministration of levodopa (or another short-acting do-paminergic agent) and the degree of dopaminergic de-nervation in the striatum. The latter plays an importantrole in setting the threshold required for dyskinesias todevelop [9]. A direct relationship between the degree ofstriatal denervation and the time required to developdyskinesias has been demonstrated in monkeys ren-dered parkinsonian with 1-methyl-4-phenyl-1,2,3,6-tet-rahydropyridine [78]. The same observation has also

been replicated in PD patients [42] and has been indi-rectly confirmed by the finding that patients with dopa-responsive dystonia, who have parkinsonism withoutnigrostriatal denervation, uncommonly develop dyski-nesias [21].

Dopaminergic denervation and dopaminergic prim-ing induce a series of anatomical and functional changesin the striatum leading to further and more persistentdysfunction. Dyskinesias are probably generated by apersistent enhancement of the responsiveness of stria-tal medium sized spiny neurons to dopaminergic treat-ment. This is an aftermath of dopamine depletion and isassociated with over-expression of specific components

of the signal transduction machinery. If protracted, thiscondition may ultimately lead to long-term changes ingene expression, which will permanently affect the func-tion of striatal medium spiny neurons [77].

Pre-synaptic dopaminergic changes provide a first-level explanation for the occurrence of both motor com-plications and dyskinesias. The occurrence of severedopaminergic denervation implies the loss of compen-satory mechanism in the nigrostriatal system, and al-ters the routes of levodopa uptake and metabolism inthe brain. When the dopaminergic system is not severelycompromised, the processing of exogenous levodopa oc-curs primarily in nigrostriatal neurons, where levodopa

is converted to dopamine, stored in synaptic vesicles,and released by physiological stimuli into the extracellu-lar space, where clearance of dopamine is ensured frompresynaptic reuptake mechanism. In the striatum of pa-tients with mild PD, levodopa levels may be buffered bythe compensatory effort of surviving nigrostriatal neu-rons providing more or less constant dopamine receptoractivation. This is clinically evidenced by the occurrenceof long-duration anti-parkinsonian responses whichoutlasts the half-life of levodopa, enabling a stable mo-

tor state. Studies have shown that in more advanced PD,with severe lesions of the nigrostriatal pathway, exoge-nous levodopa might be decarboxylated to dopamine atnon-dopaminergic sites, especially serotoninergic neu-rons, glia and endothelial cells, that are not equippedto store, release and reuptake dopamine in a controlledashion [12, 47, 55]. As a result, intrasynaptic dopamine

evels no longer remain constant, but mirror the broadluctuation in plasma levodopa levels that occur after

each dose, causing dopamine receptors to be exposedto alternating high and low concentrations of dopa-

ine [61]. The clinical counterpart of this condition isthe observation of short-duration motor responses to le-vodopa administration (Fig.1). The combination of do-

amine depletion and the administration of short-liveddopaminergic drugs, causing a pulsatile stimulation ofstriatal dopamine receptors in contrast to the tonic stim-ulation that occurs under physiologic circumstances, re-sults in the development of dyskinesias.

Post-synaptic dopaminergic changes are also crucial

to the development of dyskinesias in PD. Loss of dopa-inergic terminals in the striatum and non-physiologi-

cal dopaminergic stimulation can cause plastic changesaltering the normal functioning of the striatum, furthercontributing to pulsatile dopamine stimulation. The de-velopment of dyskinesias is not correlated with any con-sistent variations in the density of dopamine receptors[8, 55], but rather to downstream signal transduction

athways causing post-synaptic changes in proteins andgene expression [7, 26, 28]. Dopaminergic priming andbehavioral sensitization must cause molecular changesat the post-synaptic level in order to cause dyskinesias.

Non-dopaminergic neurons are also considered to

contribute to the pathophysiology of PD dyskinesias. Adysregulation of medium sized striatal spiny neuronscan contribute to the pathogenesis of dyskinesias [10,18]. Non-physiological stimulation of dopamine recep-tors on striatal spiny neurons leads to changes in sub-unit phosphorylation pattern of coexpressed ionotropic

MDA and AMPA glutamatergic receptors [17]. Resul-tant sensitization of these receptors augments corticalexcitatory input to the spiny efferent neurons, thus al-tering striatal output in ways that compromise motorunction [14]. Consistent with this possibility is the ob-

servation that drugs that block glutamate receptors tendto diminish motor complications in both parkinsonian

odels and patients [19]. In addition, enhancement ofetabotropic glutamatergic receptors may also contrib-ute to the pathogenesis of dyskinesias [76]. Other recep-tor types located in the basal ganglia also make an im-

ortant contribution to the functional state of striataloutput neurons and are targets for novel anti-dyskinetictreatments. They include: cannabinoid CB1, adenosine

2A, serotonergic 5HT A, 5HT1A, -2 adrenergic and opi-oid receptors [11].

Page 4: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 4/10

35

Clinical features

The perception of dyskinesias may differ between doc-tors and PD patients (Table2). It is not uncommon toobserve that patients stand dyskinesias in the on-pe-riod when they are mobile and try to avoid unsatisfac-

tory partial-on periods, particularly if dystonic dyski-nesias occur.The phenomenology of dyskinesias is variable; they

can occur at different times following intake of a dopa-minergic medication and variably combine dystonic andchoreic movements. The dystonic nature of dyskinesiasin PD was recognized early, when the expressions “im-provement-dystonia-improvement” (I-D-I) and “dysto-nia-improvement-dystonia” (D-I-D) were used [58] todescribe what was otherwise called “diphasic” [6] and“peak-dose” [54] dyskinesias (Fig. 2). At the same time it

was also noted that choreic features are characteristic ofdyskinesias occurring in the on-period [45]. On-periodand off-period dyskinesias differ in several ways: theirst are mainly choreic and dystonic whereas the latter

are purely dystonic. Tremor and pain (likely related torigidity) may be present in the off-period and decreasewith improving motor condition [49, 51] (Fig.3). This

gradient provides a clinical continuum between a hyper-dopaminergic and a hypo-dopaminergic state [51].

Dysk nes as follow ng acute levodopa challenge

The clinical phenomenology of dyskinesias (namely,their choreiform or dystonic appearance) has been cor-related to the motor response elicited by an acute le-vodopa challenge.

Ten PD patients (three men and seven women) witha diagnosis according to the Queen Square Brain Bankcriteria [33] were evaluated. They had disabling motor

luctuations with prolonged and, at least occasionally,unpredictable off-periods (patients spent 25% or moreof the waking day in the off-state) and on-state dyskine-sias. Their Hoehn-Yahr stage was ≥ III in the practicallydefined off-condition [44].

All patients were on dopaminergic therapy with le-vodopa and one dopamine agonist at the moment of en-rollment and gave their written informed consent. Theincluded patients were assessed in the morning, in the

ractically defined off-condition [44]. The clinical eval-

days

    P   o   s    t  -   s   y   n   a   p    t    i   c   n   e   u   r   o   n

lial cell

   L  e  v  o  d  o

  p  a

    P   o   s    t  -   s   y   n   a   p    t    i   c   n   e   u   r   o   n

   L  e  v  o  d  o

  p  a

Early stage PD Late stage PD

hours

Long-duration response

us stror  or atumus str

l l ell

Short-duration response

Fig.1 ong-duration responses depend on the availability of functionaldopaminergic nigrostriatal terminals,whereas short-duration responses aregenerate y evo opa pro uce outside nigrostriatal dopaminergic termnals, once these are degenerated. Inear y isease stages, ong- uration responses allow or a continuous clinical

benefit (“honey moon” period); lateron, short-duration responses appearand long-duration responses graduallywane. Super-sensitive post-synaptic opaminergic receptors are schematicallys own in ye ow

Table yskinesias: patients’ vs. doctors’ viewpoints

octors

ys nes as as a motor p enomenon

There are no tools to evaluate the impact of dyskinesias on the patients’ua ty o e

at ents

May not be aware of dyskinesias (particularly peak-dose dyskinesias)

  n erest mate t e r actua preva ence

Estimate the prevalence of fluctuations and dyskinesias that areclinically significant

Page 5: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 5/10

3

uation of each patient was carried out using the follow-ing outcome measures: parkinsonian motor disabilitywas assessed using the Unified Parkinson’s Disease Rat-ing Scale motor score (UPDRS) [24], upper limb speed

as an index of bradikinesia was assessed by the tappingtest (TT) [62]; the evaluation of topography, type andseverity of dyskinesias was performed while the patientwas sitting, walking and performing TT, by using items11 and 12 of the Unified Huntington’s Rating Scale [34],applied to face, mouth, trunk, upper and lower limbs.Following baseline assessment of all the measured vari-

ables, the patients received an acute levodopa/carbidopa250/25 mg) challenge followed by TT and dyskinesias

rating every 15 min and by UPDRS motor assessmentevery 7 min. The complete clinical evaluation was car-ried out until the TT score returned <15% better thanbaseline TT score.

The patients had a mean age of 61.2 (±10.4) yearsand a disease duration of 12.3 (±5) years. They had beentreated with levodopa for 10.6 (± 5.1) years, had motorluctuations for the last 4.1 (±2.3) years and dyskine-

sias for 3.9 (± 2.5) years. At the time of evaluation the to-tal levodopa equivalent dose was 1,086.5(±529) mg. Allthe patients had an excellent response to levodopa with

an average improvement of 53.4 % following the acutechallenge.

The drug challenge produced appreciable short-du-ration motor responses in every patient, as shown by thetime-dependent decrease of UPDRS motor score and in-crease of TT scores (Fig.4 ). Onset of motor improve-

ent occurred between 15 and 45 min following theevodopa challenge and averaged 28.5± 8.5min. The on-eriod lasted for a minimum of 130 and a maximum of

240 min (on average 179.5 ± 41.3 min). Dyskinesias oc-curred in all patients during the on-period, with dys-tonic, choreic or combined features (Fig.4 B).

At baseline evaluation, fixed dystonia was present in

ive patients. In two of them it exclusively involved thetrunk, whereas in the other three the distribution wasgeneralized. All patients had on-period dystonia withthe features of mobile or fixed dystonia. Two patients

resented chorea and dystonia in combination duringthe entire on-period. Dystonia was more commonly de-tected in the mouth (9 patients), in the upper limbs (9

atients), in the lower limbs (9 patients), in the trunk8 patients) and in the face (5 patients). Dystonia se-

verity was ranked in decreasing order from the trunkmean score 2.0± 1.6, legs 1.5± 1.5, arms 1.4± 1.3, mouth

0.8±1.0 and to the face 0.6±1.0).Chorea was not observed at baseline and only oc-

curred in the on-period. It was detected in nine patientsstarting 41.7± 26.8 min following levodopa challenge. Inthree patients, limb chorea preceded the observable  onstate by an average of 13.5± 9.5 min. On-period choreaoccurred in decreasing frequency from the upper limbs8 patients), to the lower limbs (7 patients), the trunk2 patients), and the mouth or face (1 patient). Cho-

rea severity was maximal in the trunk (0.6±1.1), andilder in the legs and arms (0.4 ± 0.7), or face and mouth

0.1±0.2).

Peak-dose

quare wave

Diphasic

b

F g. 2 ea - ose ys inesias (correspon ing to I-D-I) are t e typica on-periodyskinesias observed in PD ( ); diphasic dyskinesias (corresponding to D-I-D) occur when patients are in the transitional state from the off- to the on-period );square-wave dyskinesias encompass a combination o both types C)

Fig. The transition rom peak-dose to diphasic dyskinesias and o -period dys-tonia is associated with increasing pain and dystonia and the occurrence o tremor.

is p enomeno ogica gra ient appears as a c inica continuum etween a yper-opaminergic an a ypo- opaminergic state in patients wit comp icate P

Page 6: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 6/10

37

When patients returned to the of  state, off-perioddystonia recurred at the same degree and characteris-tics in all of five patients who had it at baseline, whereasit was not observed in the remaining patients. No cho-reic movements were detected in the off-period.

This observational study following acute levodopachallenge was designed to evaluate in a controlled settingthe onset and presentation of dyskinesias, from the  off 

otor condition preceding the acute challenge throughthe complete duration of the  on state, until the end ofshort-duration motor response. This allowed the com-

arative features of choreic and dystonic dyskinesias to

be observed through the off and on-periods, and to rec-ognize that chorea is only present during the peak of

otor effect. Dystonia, instead, was observed through-out the observation period and increased during the du-ration of motor response. Diphasic dyskinesias, whichoccurred at the beginning and end of motor response,were mainly dystonic, whereas peak-dose dyskinesiaswere mixed, choreic and dystonic (Fig.5). These dataextend and complete previous suggestions that dyski-nesias constitute a continuum with changing phenome-nology [51] and better specify how PD motor responseis associated to chorea and dystonia.

It has been already observed that dystonia can also

occur in the off motor state [49, 51, 85]. Off-period dys-tonia is mainly fixed and turns into a mobile form dur-ing the on-period. As the motor response induced byacute challenge vanishes, dystonia turns back again toa fixed form.

Rat onale for management

The observation that dyskinesias increase in the on stateas prompted attempts to identify perfectly titrated in-

dividual doses for continuous infusions of levodopa [72,74] or dopamine agonists [65, 66]. This was supposed

Fig. ean motor improvement emonstrate y TT an UPDRS motor score (A),compare to t e mean c orea an ystonia scores B) following an acute challengewith levodopa/carbidopa (250/25 mg). X-axes show time in minutes ollowing theacute a ministration; Y-axes s ow mean scores, as out ine in t e text

Fixed dystonia

Short-duration

motor response

Chorea

Mobile dystonia

dyskinesi

Time

(hours)1

    D    i   p    h   a   s    i   c    d   y   s    k    i   n   e   s    i   a

    D    i   p    h   a   s    i

    d   y   s    k    i   n   e   s    i   a

 state transition threshold

Fig.5 chematic representation of dyskinesias oc-curring in relation with a short-duration motor re-sponse (red line) elicited by levodopa. Dystonia isn icate in ue, c orea in green. Fixe ystonia

occurs throughout the duration o motor response,prevalently in the o   state. en t e pat ent turnsn (crossing t e otte re t res o ), mo i e ys-

tonia (hatched blue area) occurs during the on-pe-rio ; c orea ( atc e green area) can a itiona y eobserved at peak dose. This schematic drawing alsondicates that peak-dose dyskinesias are choreic andystonic, w ereas ip asic ys inesias are ystonic,ut not choreic

Page 7: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 7/10

3

to avoid the occurrence of severe on-state dyskinesias,

yet providing an adequate motor condition for the du-ration of infusion. This historical approach was basedon the observation that motor fluctuations and dys-kinesias coexisted in patients with complicated PD. Itseemed therefore reasonable to address both phenom-ena using a unique treatment strategy [16]. Unfortu-nately, attempts to ameliorate dyskinesias in advancedPD patients by giving smaller, more frequent doses of le-vodopa or a continuous infusion turned out to be coun-ter-productive [56]. Attempts to manage the therapeuticwindow by changing the pharmacokinetics of levodopaor of dopamine agonists were not efficacious in control-ling on-period dyskinesias. The reason why this option

does not provide an efficient solution is highlighted inFig.6 which schematically shows a typical sigmoid dose-effect curve for anti-parkinsonian response to levodopa(or dopamine agonists). In order to elicit maximal anti-parkinsonian effect an optimal dose   of levodopa(or dopamine agonist) must be given. There is no advan-tage of further increasing the medication, as this wouldprovide no additional anti-parkinsonian effect (dashedred area). This is a very efficacious treatment in earlydisease stages, when dyskinesias are not present. How-ever, after dyskinesias are established, a threshold med-ication dose for dyskinesias becomes apparent. Oncethe dyskinesia threshold moves to the left of the max-

imal antiparkinsonian dose line

, dyskinesias becomeinevitably associated with the maximal anti-parkinso-nian effect. In practical terms, it becomes necessary toprovide suboptimal treatment to keep the patient freefrom dyskinesias. As PD treatment and disease prog-ress, the dyskinesia threshold moves further to the leftand the therapeutic window   narrows, making thethreshold of maximal anti-parkinsonian effect withoutdyskinesias move further down.

Continuous levodopa infusion corrects the phar-

acokinetic causes leading to dyskinesias, as it allows

avoiding the peaks and troughs of repeated oral admin-istration [63]; however, the pharmacodynamic causesof dyskinesias illustrated by Fig.6 remain. This explainswhy dyskinesias are still observed when the patients aretreated enough to reach an adequate on state [3, 60, 64].Similarly, dyskinesias do not disappear when patientsare treated with continuous infusions of dopamine ag-onists [37, 82].

High frequency stimulation of the subthalamic nu-cleus or the globus pallidum provide an alternative ap-

roach to the treatment of dyskinesias in PD [83]. Thetwo targets are considered to reduce dyskinesias by

eans of different anatomofunctional mechanisms

[13]. Globus pallidum stimulation probably has a directanti-dyskinetic effect by interfering with pallidal out-low, whereas subthalamic nucleus stimulation reduces

edication requirements (thus changing the pharma-codynamic dose-effect curve) [57] and in addition mayalso affect pallidal outflow at the level of the ansa len-ticularis, situated just below the subthalamic nucleus.Deep brain stimulation of the subthalamic nucleus hasstriking ameliorative effects on off-period dystonia [41],yielding an average reduction in dyskinesias of approx-imately 70% [39]. It must be remember that the numberof candidate patients for deep brain stimulation repre-sent a minority of the PD population, because strict se-

ection criteria are usually implemented in most centers[75].A third line of treatment is based on the administra-

tion of specific antidyskinetic drugs capable to move tothe right the dyskinesia threshold depicted in Fig.6.

ovel potential therapeutic strategies attempt to act onnon-dopaminergic neurotransmitter systems and re-duce the expression of dyskinesias, once they are estab-ished, without interfering with the anti-parkinsonian

activity of dopaminergic drugs [7, 25]. The first (and

    A   n    t    i   p   a   r    k    i   n   s   o   n    i   a   n   r   e   s   p   o   n   s   e

Levodopa dose

1 Antiparkinsonian threshold

d se (25% motor

improvement)

2 Dyskinesia threshold dos

  Therapeutic window

 Optimal dos

  Maximal antiparkinsonian

effect

6 Slope

7 Maximal antiparkinsonian

effect without dyskinesias

75

6

1

3

2 4

Fig.6 igmoid dose-e ect curve oranti-par insonian response to t e aministration of increasing doses of levodopa (blue). Motor-related thresholdsare re , ys inesia-re a te t res o sare green. See text or an explanation

Page 8: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 8/10

3

still only) drug with proven anti-dyskinetic efficacy isamantadine, a glutamate antagonist active on NMDA re-ceptors, whose activity lends further support to the glu-tamatergic hypothesis on the pathogenesis of dyskine-sias [31, 69]. The improvement of dyskinesias is around50 % without concomitant worsening of parkinsoniansymptoms [48, 84].

Other potential anti-dyskinetic candidates have beenexcluded or are still under scrutiny. Sarizotan demon-strated potential efficacy in an open label trial, but sub-sequently proved to be ineffective in two double blind

lacebo-controlled studies [30]. Atypical neurolep-tics (particularly clozapine) have also been considered,but presently there is insufficient evidence to supportthe efficacy of clozapine in reducing dyskinesias [69].Other promising anti-dyskinetic drugs act on glutamateNMDA, AMPA or mGluR), serotonin (5HT1A, 5HT1B/D,

5HT2A–2C), adenosine (A2), opiate or other receptors.

Their potential efficacy is currently under scrutiny.

onflict of interest The authors have no conflict of interest to de-clare.

References

1. Ahlskog JE, Muenter MD (2001) Fre-quency of levodopa-related dyskine-sias and motor fluctuations as esti-mated from the cumulative literature.Mov Disord 16:448–458

. Albanese A, Valente EM, Romito LM,Bellacchio E, Elia AE, Dallapiccola B(2005) The PINK1 phenotype can beindistinguishable from idiopathic Par-kinson disease. Neurology 64:1958–1960

3. Antonini A, Isaias IU, Canesi M, ZibettiM, Mancini F, Manfredi L, Dal FM,Lopiano L, Pezzoli G (2007) Duodenallevodopa infusion for advanced Par-kinson’s disease: 12-month treatmentoutcome. Mov Disord 22:1145–1149

4. Barbeau A (1969) L-Dopa therapy inParkinson’s disease: a critical review ofnine years’ experience. Can Med AssocJ 101:59–68

5. Barbeau A (1971) Long-term side-ef-fects of levodopa. Lancet 1:395

. Barbeau A (1975) Diphasic dyskinesiaduring levodopa therapy. Lancet 1:756

7. Bezard E, Brotchie JM, Gross CE (2001)Pathophysiology of levodopa-induceddyskinesia: potential for new therapies.Nat Rev Neurosci 2:577–588

8. Blanchet PJ, Calon F, Morissette M,Hadj TA, Belanger N, Samadi P, Gron-din R, Gregoire L, Meltzer L, Di PT, Be-dard PJ (2004) Relevance of the MPTPprimate model in the study of dyskine-sia priming mechanisms. Parkinson-ism Relat Disord 10:297–304

9. Boyce S, Rupniak NM, Steventon MJ,Iversen SD (1990) Nigrostriatal dam-age is required for induction of dyski-

nesias by l-dopa in squirrel monkeys.Clin Neuropharmacol 13:448–458

10. Bravi D, Mouradian MM, Roberts JW,Davis TL, Sohn YH, Chase TN (1994)Wearing-off fluctuations in Parkin-son’s disease: contribution of postsyn-aptic mechanisms. Ann Neurol 36:27–31

11. Brotchie JM (2005) Nondopaminergicmechanisms in levodopa-induced dys-kinesia. Mov Disord 20:919–931

12. Brown WD, Taylor MD, Roberts AD,Oakes TR, Schueller MJ, Holden JE,Malischke LM, Dejesus OT, Nickles RJ(1999) FluoroDOPA PET shows thenondopaminergic as well as dopami-nergic destinations of levodopa. Neu-rology 53:1212–1218

13. Burchiel KJ, Anderson VC, Favre J,Hammerstad JP (1999) Comparison of

allidal and subthalamic nucleus deepbrain stimulation for advanced Par-kinson’s disease: results of a random-ized, blinded pilot study. Neurosurgery

5:1375–138214. Calon F, Rajput AH, Hornykiewicz

O, Bedard PJ, Di Paolo T (2003) Le-vodopa-induced motor complicationsare associated with alterations of glu-amate receptors in Parkinson’s dis-

ease. Neurobiol Dis 14:404–41615. Chapuis S, Ouchchane L, Metz O, Ger-

baud L, Durif F (2005) Impact of themotor complications of Parkinson’sdisease on the quality of life. Mov Dis-ord 20:224–230

16. Chase TN, Engber TM, Mouradian MM(1994) Palliative and prophylactic ben-efits of continuously administered do-

aminomimetics in Parkinson’s dis-ease. Neurology 44:S15–S18

17. Chase TN, Konitsiotis S, Oh JD (2001)Striatal molecular mechanisms andmotor dysfunction in Parkinson’s dis-ease. Adv Neurol 86:355–360

18. Chase TN, Oh JD, Blanchet PJ (1998)Neostriatal mechanisms in Parkinson’sdisease. Neurology 51:S30–S35

19. Chase TN, Oh JD, Konitsiotis S (2000)Antiparkinsonian and antidyskinetic

activity of drugs targeting central glu-amatergic mechanisms. J Neurol 247:

S36–S4220. Cotzias GC, van Woert MH, Schiffer

LM (1967) Aromatic amino acids andmodification of parkinsonism. N Engl Med 276:374–379

1. De la Fuente-Fernandez R (1999)Drug-induced motor complicationsin dopa-responsive dystonia: implica-tions for the pathogenesis of dyskine-sias and motor fluctuations. Clin Neu-ropharmacol 22:216–219

2. Durif F, Deffond D, Dordain G,Tournilhac M (1994) Apomorphineand diphasic dyskinesia. Clin Neuro-pharmacol 17:99–102

3. Fabbrini G, Brotchie JM, GrandasF, Nomoto M, Goetz CG (2007) Le-vodopa-induced dyskinesias. Mov Dis-ord 22:1379–1389

4. Fahn S, Elton RL, Members of the UP-DRS Development Committee (1987)Unified Parkinson’s disease ratingscale. In: Fahn S, Marsden CD, CalneD, Goldstein M (eds) Recent develop-ments in Parkinson’s disease. Macmil-lan Healthcare Information, FlorhamPark, pp 153–163

5. Fox SH, Lang AE, Brotchie JM (2006)Translation of nondopaminergic treat-ments for levodopa-induced dyskine-sia from MPTP-lesioned nonhumanprimates to phase IIa clinical studies:keys to success and roads to failure.Mov Disord 21:1578–1594

6. Gerfen CR (1995) Dopamine-recep-tor function in the basal ganglia. ClinNeuropharmacol 18:S162–S177

7. Gerfen CR, Engber TM, Mahan LC, Su-sel Z, Chase TN, Monsma FJ, Sibley DR(1990) D1 and D2 dopamine receptor-regulated gene expression of striato-nigral and striatopallidal neurons. Sci-ence 250:1429–1432

8. Gerfen CR, Miyachi S, Paletzki R,

Brown P (2002) D1 dopamine receptorsupersensitivity in the dopamine-de-pleted striatum results from a switchin the regulation of ERK1/2/MAP ki-nase. J Neurosci 22:5042–5054

9. Gilgun-Sherki Y, Djaldetti R, MelamedE, Offen D (2004) Polymorphism incandidate genes: implications for therisk and treatment of idiopathic Par-kinson’s disease. Pharmacogenomics J4:291–306

Page 9: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 9/10

4

30. Goetz CG, Damier P, Hicking C, LaskaE, Muller T, Olanow CW, Rascol O, RussH (2007) Sarizotan as a treatment fordyskinesias in Parkinson’s disease: adouble-blind placebo-controlled trial.Mov Disord 22:179–186

31. Goetz CG, Poewe W, Rascol O, SampaioC (2005) Evidence-based medical re-view update: pharmacological and sur-

gical treatments of Parkinson’s disease:2001 to 2004. Mov Disord 20:523–539

32. Hoehn MM, Yahr MD (1967) Parkin-sonism: onset, progression and mor-tality. Neurology 17:427–442

33. Hughes AJ, Ben-Shlomo Y, Daniel SE,Lees AJ (1992) What features improvethe accuracy of clinical diagnosis inParkinson’s disease? A clinicopatho-logic study. Neurology 42:1142–1146

34. Huntington Study Group (1996) Uni-fied Huntington’s Disease Rating Scale:reliability and consistency. Mov Disord11:136–142

35. Isacson O, Hantraye P, Maziere M, So-froniew MV, Riche D (1990) Apomor-

phine-induced dyskinesias after exci-totoxic caudate-putamen lesions andthe effects of neural transplantation innon-human primates. Prog Brain Res82:523–533

36. Kaiser R, Hofer A, Grapengiesser A,Gasser T, Kupsch A, Roots I, Brock-moller J (2003) L-dopa-induced ad-verse effects in PD and dopaminetransporter gene polymorphism. Neu-rology 60:1750–1755

37. Katzenschlager R, Hughes A, Evans A,Manson AJ, Hoffman M, Swinn L, WattH, Bhatia K, Quinn N, Lees AJ (2005)Continuous subcutaneous apomor-phine therapy improves dyskinesias

in Parkinson’s disease: a prospectivestudy using single-dose challenges.Mov Disord 20:151–157

38. Khan NL, Graham E, Critchley P,Schrag AE, Wood NW, Lees AJ, BhatiaKP, Quinn N (2003) Parkin disease: aphenotypic study of a large case series.Brain 126:1279–1292

39. Kleiner-Fisman G, Liang GS, MobergPJ, Ruocco AC, Hurtig HI, Baltuch GH,Jaggi JL, Stern MB (2007) Subthalamicnucleus deep brain stimulation for se-vere idiopathic dystonia: impact on se-verity, neuropsychological status, andquality of life. J Neurosurg 107:29–36

40. Kostic V, Przedborski S, Flaster E, Ster-

nic N (1991) Early development of le-vodopa-induced dyskinesias and re-sponse fluctuations in young-onsetParkinson’s disease. Neurology 41:202–205

41. Krack P, Pollak P, Limousin P, Benaz-zouz A, Deuschl G, Benabid AL (1999)From off-period dystonia to peak-dosechorea. The clinical spectrum of vary-ing subthalamic nucleus activity. Brain122:1133–1146

42. Kumar A, Mann S, Sossi V, Ruth TJ,Stoessl AJ, Schulzer M, Lee CS (2003)11C]DTBZ-PET correlates of le-

vodopa responses in asymmetric Par-kinson’s disease. Brain 126:2648–2655

43. Kumar N, Van Gerpen J , Bower JH,Ahlskog JE (2005) Levodopa-dyskine-sia incidence by age of Parkinson’s dis-ease onset. Mov Disord 20:342–344

44. Langston JW, Widner H, Goetz CG,Brooks D, Fahn S, Freeman T, WattsR (1992) Core assessment programor intracerebral transplantations

(CAPIT). Mov Disord 7:2–1345. Lees AJ, Shaw KM, Stern GM (1977)

“Off period” dystonia and “on period”choreoathetosis in levodopa-treatedpatients with Parkinson’s disease. Lan-cet 2:1034

4 . Linazasoro G (2007) Pathophysiologyof motor complications in Parkinsondisease: postsynaptic mechanisms arecrucial. Arch Neurol 64:137–140

47. Lopez A, Munoz A, Guerra MJ, Laban-deira-Garcia JL (2001) Mechanisms of

he effects of exogenous levodopa onhe dopamine-denervated striatum.

Neurosc ence 103:639–65148. Luginger E, Wenning GK, Bosch S,

Poewe W (2000) Beneficial effects ofamantadine on L-dopa-induced dyski-nesias in Parkinson’s disease. Mov Dis-ord 15:873–878

49. Luquin MR, Scipioni O, VaamondeJ, Gershanik O, Obeso JA (1992) Le-vodopa-induced dyskinesias in Par-kinson’s disease: clinical and pharma-cological classification. Mov Disord7:117–124

50. Lyons KE, Hubble JP, Troster AI, PahwaR, Koller WC (1998) Gender differ-

ences in Parkinson’s disease. Clin Neu-ropharmacol 21:118–12151. Marconi R, Lefebvre-Caparros D, Bon-

net AM, Vidailhet M, Dubois B, Agid Y(1994) Levodopa-induced dyskinesiasin Parkinson’s disease phenomenologyand pathophysiology. Mov Disord 9:2–12

52. Marongiu R, Ferraris A, Ialongo T,Michiorri S, Soleti F, Ferrari F, Elia AE,Ghezzi D, Albanese A, Altavista MC,Antonini A, Barone P, Brusa L, CortelliP, Martinelli P, Pellecchia MT, PezzoliG, Scaglione C, Stanzione P, Tinazzi M,Zecchinelli A, Zeviani M, Cassetta E,Garavaglia B, Dallapiccola B, Bentivo-

glio AR, Valente EM (2008) PINK1 het-erozygous rare variants: prevalence,significance and phenotypic spectrum.Hum Mutat 29:565–541

53. Marras C, Lang A, Krahn M, Tomlin-son G, Naglie G (2004) Quality of lifein early Parkinson’s disease: impact ofdyskinesias and motor fluctuations.Mov Disord 19:22–28

54. Marsden CD, Parkes JD (1976) “On-off” effects in patients with Parkin-son’s disease on chronic levodopa ther-apy. Lancet 1:292–296

55. Metman LV, Konitsiotis S, Chase TN(2000) Pathophysiology of motor re-sponse complications in Parkinson’sdisease: hypotheses on the why, where,and what. Mov Disord 15:3–8

56. Metman LV, van den Munckhof P,Klaassen AA, Blanchet P, MouradianMM, Chase TN (1997) Effects of supra-threshold levodopa doses on dyskine-sias in advanced Parkinson’s disease.Neurology 49:711–713

57. Moro E, Scerrati M, Romito LM, RoselliR, Tonali P, Albanese A (1999) Chronicsubthalamic nucleus stimulation re-duces medication requirements in Par-kinson’s disease. Neurology 53:85–90

58. Muenter MD, Sharpless NS, Tyce GM,Darley FL (1977) Patterns of dystonia(‘I-D-I’ and ‘D-I-D’) in response to L-dopa therapy for Parkinson’s disease.Mayo Clin Proc 52:163–174

59. Nash JE, Brotchie JM (2000) A com-mon signaling pathway for striatalNMDA and adenosine A2a receptors:implications for the treatment of Par-kinson’s disease. J Neurosci 20:7782–7789

0. Nilsson D, Nyholm D, Aquilonius SM(2001) Duodenal levodopa infusion inParkinson’s disease – long-term expe-rience. Acta Neurol Scand 104:343–348

1. Nutt JG, Holford NH (1996) The re-sponse to levodopa in Parkinson’s dis-ease: imposing pharmacological lawand order. Ann Neurol 39:561–573

2. Nutt JG, Woodward WR, Hammer-stad JP, Carter JH, Anderson JL (1984)

The “on-off” phenomenon in Parkin-son’s disease. Relation to levodopa ab-sorption and transport. N Engl J Med310:483–488

3. Nyholm D, Askmark H, Gomes-TrolinC, Knutson T, Lennernas H, Nystrom C,Aquilonius SM (2003) Optimizing le-vodopa pharmacokinetics: intestinalinfusion versus oral sustained-releasetablets. Clin Neuropharmacol 26:156–163

64. Nyholm D, Nilsson Remahl AI, Diz-dar N, Constantinescu R, Holmberg B,Jansson R, Aquilonius SM, AskmarkH (2005) Duodenal levodopa infusionmonotherapy vs oral polypharmacy in

advanced Parkinson disease. Neurol-ogy 4:21 –22365. Obeso JA, Luquin MR, Martinez Lage

JM (1986) Intravenous lisuride cor-rects oscillations of motor perfor-mance in Parkinson’s disease. AnnNeurol 19:31–35

. Obeso JA, Luquin MR, Vaamonde J,Grandas F, Martinez Lage JM (1987)Continuous dopaminergic stimulationin Parkinson’s disease. Can J Neurol Sci14:488–492

Page 10: 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

8/14/2019 1. journal 1- Levodopa-indvvvuced dyskinesias.pdf

http://slidepdf.com/reader/full/1-journal-1-levodopa-indvvvuced-dyskinesiaspdf 10/10

67. Olanow CW, Obeso JA (2000) Prevent-ing levodopa-induced dyskinesias.Ann Neurol 47:S167–S176

8. Oliveri RL, Annesi G, Zappia M, Civi-telli D, Montesanti R, Branca D, Nico-letti G, Spadafora P, Pasqua AA, Citt-adella R, Andreoli V, Gambardella A,Aguglia U, Quattrone A (1999) Dopa-mine D2 receptor gene polymorphism

and the risk of levodopa-induced dys-kinesias in PD. Neurology 53:1425–1430

69. Pahwa R, Factor SA, Lyons KE, OndoWG, Gronseth G, Bronte-Stewart H,Hallett M, Miyasaki J, Stevens J, WeinerWJ (2006) Practice parameter: treat-ment of Parkinson disease with mo-tor fluctuations and dyskinesia (an ev-idence-based review): report of theQuality Standards Subcommittee ofthe American Academy of Neurology.Neurology 66:983–995

70. Parkinson Study Group (1996) Impactof deprenyl and tocopherol treatmenton Parkinson’s disease in DATATOP

patients requiring levodopa. Ann Neu-rol 39:37–45

71. Pechevis M, Clarke CE, Vieregge P,Khoshnood B, Deschaseaux-Voinet C,Berdeaux G, Ziegler M (2005) Effectsof dyskinesias in Parkinson’s diseaseon quality of life and health-relatedcosts: a prospective European study.Eur J Neurol 12:956–963

72. Quinn N, Parkes JD, Marsden CD(1984) Control of on/off phenomenonby continuous intravenous infusion oflevodopa. Neurology 34:1131–1136

73. Rascol O, Brooks DJ, Korczyn AD, DeDeyn PP, Clarke CE, Lang AE, AbdallaM (2006) Development of dyskinesiasin a 5-year trial of ropinirole and L-dopa. Mov Disord 21:1844–1850

74. Sage JI, McHale DM, Sonsalla P, Vita-liano D, Heikkila RE (1989) Contin-

uous levodopa infusions to treat com-plex dystonia in Parkinson’s disease.

Neurology 39:888–89175. Saint-Cyr JA, Albanese A (2006) STN

DBS in PD: selection criteria for sur-ery should include cognitive and psy-

chiatric factors. Neurology 66:1799–1800

76. Samadi P, Gregoire L, Morissette M,Calon F, Hadj TA, Belanger N, DridiM, Bedard PJ, Di PT (2008) Basal gan-glia group II metabotropic glutamatereceptors specific binding in non-hu-man primate model of L-Dopa-in-duced dyskinesias. Neuropharmacol-ogy 54:258–268

77. Santini E, Valjent E, Fisone G (2008)Parkinson’s disease: levodopa-induced

dyskinesia and signal transduction.FEBS J 275:1392–1399

78. Schneider JS (1989) Levodopa-induceddyskinesias in parkinsonian monkeys:relationship to extent of nigrostria-al damage. Pharmacol Biochem Behav

34:193–1979. Schrag A, Hovris A, Morley D, Quinn

N, Jahanshahi M (2003) Young- versusolder-onset Parkinson’s disease: im-pact of disease and psychosocial con-sequences. Mov Disord 18:1250–1256

80. Schrag A, Quinn N (2000) Dyskinesiasand motor fluctuations in Parkinson’sdisease. A community-based study.Bra n 123:2297–2305

81. Schrag A, Schott JM (2006) Epidemi-ological, clinical, and genetic charac-teristics of early-onset parkinsonism.Lancet Neurol 5:355–363

82. Stocchi F, Ruggieri S, Vacca L, Olanow

CW (2002) Prospective randomizedtrial of lisuride infusion versus oral le-vodopa in patients with Parkinson’sdisease. Brain 125:2058–2066

83. The Deep-Brain Stimulation for Par-kinson’s Disease Study Group (2001)Deep-brain stimulation of the subtha-lamic nucleus or the pars interna ofthe globus pallidus in Parkinson’s dis-ease. N Engl J Med 345:956–963

84. Verhagen Metman L, Del Dotto P, vanden Munckhof P, Fang J, MouradianMM, Chase TN (1998) Amantadineas treatment for dyskinesias and mo-tor fluctuations in Parkinson’s disease.Neurology 50:1323–1326

85. Vidailhet M, Bonnet AM, MarconiR, Gouiderkhouja N, Agid Y (1994)Do parkinsonian symptoms and le-vodopa-induced dyskinesias start inthe foot? Neurology 44:1613–1616

86. Zappia M, Annesi G, Nicoletti G, Ara-bia G, Annesi F, Messina D, PuglieseP, Spadafora P, Tarantino P, CarrideoS, Civitelli D, De Marco EV, Ciro-Can-diano IC, Gambardella A, QuattroneA (2005) Sex differences in clinicaland genetic determinants of levodopapeak-dose dyskinesias in Parkinsondisease: an exploratory study. ArchNeurol 62:601–605