You are on page 1of 15

CNS Drugs 2003; 17 (7): 475-489

THERAPY IN PRACTICE 1172-7047/03/0007-0475/$30.00/0

© Adis Data Information BV 2003. All rights reserved.

Levodopa-Induced Response
Fluctuations in Patients with
Parkinson’s Disease
Strategies for Management
Teus van Laar
Department of Neurology, Groningen University Hospital, Groningen, The Netherlands

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 475
1. Clinical Symptomatology of Motor Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477
2. Pathophysiology of Motor Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 478
2.1 Peripheral Pharmacokinetics of Levodopa . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 478
2.2 Central Pharmacokinetics and Pharmacodynamics of Levodopa . . . . . . . . . . . . . . . . . . . . . . . . 478
3. Influence of Age and Duration/Dose of Levodopa on Motor Complications . . . . . . . . . . . . . . . . . . . 479
4. Preclinical Data on the Treatment of Response Fluctuations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 480
4.1 Role for Dopamine D1 Receptor Stimulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 480
4.2 Possible Influence of Priming . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 481
5. Clinical Treatment Strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 481
5.1 Wearing-Off Phenomena . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 481
5.1.1 Optimisation of Dopamine Agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 481
5.1.2 Increasing the Duration of Effect of Levodopa Doses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 483
5.2 At-Random Response Fluctuations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 484
5.2.1 Rescue Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 484
5.2.2 Continuous Stimulation of Dopamine Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 485
5.3 Dyskinesias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 486
6. Stereotactic Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 486
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487

Abstract Fluctuations in response to levodopa in patients in the advanced stages of


idiopathic Parkinson’s disease occur frequently and are a difficult problem to
treat. Patients who are treated with levodopa have an additional 10% risk of
experiencing response fluctuations with each year of treatment: 50% of patients
have this problem after 5 years of receiving levodopa therapy and almost 100% of
patients after 10 years.
The mechanisms by which response fluctuations occur are only partially
understood and can be divided into three main types: (i) presynaptic neuronal
degeneration leading to a lack of buffering of released levodopa, which is mainly
related to wearing-off phenomena; (ii) postsynaptic changes in dopamine receptor
sensitivity and number, partially caused by the presynaptic changes, which are
clinically related to at-random response fluctuations; and (iii) pharmacokinetic
476 van Laar

and pharmacodynamic influences of exogenously administered dopaminergic


agents.
Several oral and parenteral treatment strategies are recommended to manage
response fluctuations, such as optimisation of dopamine receptor agonist therapy
in combination with a reduction of the levodopa load; use of slow-release
levodopa formulations; use of catechol-O-methyltransferase inhibitors; an
increase of levodopa dose frequency; use of high-dose amantadine; and intermit-
tent or continuous use of apomorphine and/or levodopa. Continuous stimulation
of dopamine receptors with dopaminergic agents is one of the crucial basic steps
in the treatment of patients at an advanced stage of Parkinson’s disease, and the
preferential use of dopamine receptor agonists has proven to be successful in the
prevention and treatment of response fluctuations.

Idiopathic Parkinson’s disease (IPD) is a progres- initial period of stable response, mostly lasting 2–5
sive neurodegenerative disorder for which no restor- years, about 70% of patients develop a variety of
ative or neuroprotective therapy is available. As a incapacitating motor adverse events in response to
result of a continuous cell loss in the nigrostriatal levodopa treatment, mainly consisting of at-random
system of about 10% per year, as measured by response fluctuations and dyskinesias.[4] Data on the
positron emission tomography (PET),[1,2] the disease occurrence of motor fluctuations show a huge vari-
progresses despite the use of currently available ance. The Sinemet CR First study described motor
medications. fluctuations in only 20% of the patients after 5 years
The administration of dopamine precursors, of levodopa use.[5] This is in contrast to other data,
levodopa in particular, has been the gold standard in which suggest that, in young-onset IPD patients, up
the treatment for IPD since the discovery of a defi- to 30% of the patients show dyskinesia after only 1
ciency of dopamine in the basal ganglia of patients year of levodopa therapy.[6] About 30% of all
with Parkinson’s disease. Because levodopa is de- patients with Parkinson’s disease remain very stable
graded by peripheral decarboxylase to dopamine, throughout the course of the disease and seem not to
which does not pass the blood-brain barrier (BBB), be affected by these adverse motor reactions.[4] At
it has to be coadministered with a decarboxylase
the other end of the spectrum, almost 100% of
inhibitor (carbidopa or benserazide).
patients with young-onset disease develop motor
The majority of patients respond well to
complications. These complications are less likely
levodopa treatment, at least during the initial years
to occur in patients whose symptoms start after the
of therapy. Individual differences in the degree of
age of 70 years.[6,7]
symptom improvement have been reported, and ap-
proximately 15–20% of patients do not respond to It is clear that both levodopa and the progressive
levodopa,[3] very probably because they do not have degeneration of the nigrostriatal system in IPD are
Parkinson’s disease but rather one of the parkinsoni- responsible for the occurrence of these motor com-
an syndromes such as multiple system atrophy or plications. In recent years, a variety of possible
progressive supranuclear palsy (PSP). The UK approaches to managing these fluctuating motor
Brain Bank mentions that a positive response to problems have been suggested. This review pro-
levodopa is one of the supportive criteria for the vides an overview of the different possible strate-
clinical diagnosis of IPD.[3] gies, including some theoretical background. The
Even in patients who respond well to levodopa challenge for every neurologist involved in extra-
initially, control of motor symptoms gradually di- pyramidal disorders will be to individualise these
minishes during the course of treatment.[4] After an basic rules for their own patients with IPD.

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 477

1. Clinical Symptomatology of A variant of the wearing-off phenomenon is the


Motor Complications delayed-on or ‘no-on’ response, in which the latency
of effect increases.[10] Patients with this complica-
Motor complications may have various phenom- tion may have to wait 45–90 minutes before a dose
enologies. The most important drug-related motor of levodopa results in a clinical benefit. In the no-on
complications are summarised in table I. phenomenon, a single dose predictably or randomly
The first sign of the loss of symptom control, does not give any benefit at all.[11]
usually occurring after a few years of therapy, is
called end-of-dose deterioration (or ‘wearing-off’). With continuing levodopa treatment, the loss of
This term refers to the observation that the duration symptom control progresses to a situation character-
of response to levodopa administration appears to ised by severe, disabling motor oscillations (‘on-off’
have become shorter. The patient loses the smooth phenomena) and dyskinesias, the latter occurring
response during the day and starts to feel the effect predominantly during ‘on’ periods. Patients seem to
of single oral levodopa doses. The duration of the go through alternating periods of overtreatment and
beneficial effect of each levodopa dose becomes undertreatment in a totally unpredictable way. Peri-
progressively shorter. On average, the effects of a ods of immobility (‘off’) and episodes of normal or
dose of levodopa last about 2–3 hours in patients involuntary movements (on) occur at random, show-
with wearing off phenomena.[8] If the interval be- ing no apparent relationship to levodopa dosing.
tween doses in these patients happens to be longer, Dyskinesias are a common problem, occurring in
nocturnal or early-morning akinesia may be the ulti- 30–80% of patients treated over the long term.[12,13]
mate wearing-off phenomenon. The dyskinesias can be categorised according to
Besides this ‘short-duration’ response, a ‘long- their time-relationship with the levodopa dose. Most
duration’ response also exists. This is the time from patients begin to experience dyskinesias at times of
complete withdrawal of levodopa until the parkin- peak effect of levodopa (‘peak-dose dyskine-
sonian deterioration is maximal. This process may sia’).[12,13] Although they may actually be linked to
last up to 2 weeks.[9] This long-duration response is plasma concentration peaks, most affected patients
important, because it determines the baseline func- have dyskinesias during the entire period of the
tion on which the other motor fluctuations are super- effect of a single levodopa dose, making the term
imposed. The long-duration response is a very slow ‘interdose dyskinesia’ more appropriate. Most com-
fluctuation, but nevertheless it is important in ana- monly choreic movements (hyperkinesia) are seen
lysis of the whole picture. in the trunk and extremities, whereas dystonic
movement patterns prevail in the craniocervical re-
Table I. Subtypes of drug-related motor complications that can gion. In a subgroup of patients, the dyskinesias are
occur in patients with Parkinson’s disease
associated with the onset of clinical benefit. They
Response fluctuations appear to be linked to a critical plasma levodopa
Predictable concentration and may occur during this critical
Wearing-off
level at the beginning and the end of the dose effect
Nocturnal akinesia
and are therefore called ‘diphasic dyskinesias’.[14] A
Early-morning akinesia
Delayed on/no-on phenomena
particularly common form of diphasic dyskinesia is
Long-duration response
represented by stereotypic movements such as kick-
Unpredictable ing with one or both legs.
At-random on-off fluctuations Off-period dystonia is related to low plasma
Dyskinesia levodopa concentrations and occurs in 20–30% of
Peak/interdose dyskinesia patients treated long term.[15-17] It is a dystonic pos-
Diphasic dyskinesia turing, generally of the feet or legs, and is almost
Off-period dystonia
always painful. It most commonly occurs early in

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
478 van Laar

the morning (without levodopa overnight) or in be- peripheral concentration of levodopa is to inhibit
tween levodopa doses during the day. It is worse on catechol-O-methyltransferase (COMT), using drugs
the side most affected by parkinsonism.[18] such as entacapone.
Other factors causing changes in the pharmaco-
2. Pathophysiology of kinetics of levodopa are variability in gastric
Motor Complications motility (mostly too slow) and competition for trans-
port across the BBB with large neutral amino acids
The short-lived and unpredictable response to
(LNAAs). The LNAAs mainly originate from pro-
oral levodopa is caused by either the peripheral
tein-rich meals, including meat. The influx of
pharmacokinetic properties of the drug or the central
levodopa into the brain directly correlates negative-
pharmacokinetic and pharmacodynamic factors.
ly with the sum of all circulating LNAAs.[19] Periph-
2.1 Peripheral Pharmacokinetics
erally acting dopamine receptor antagonists such as
of Levodopa
domperidone and cisapride are able to improve slow
gastric emptying and may therefore improve the
The plasma elimination half-life of levodopa is effect of the administered levodopa. Strategies to
short (about 2 hours), which explains the short dura- improve gastric emptying and/or absorption may be
tion of action (2–3 hours) in patients with advanced effective, especially if ‘no-on’ or ‘delayed-on’ phe-
disease.[19] Strategies to compensate for this are to nomena occur; these strategies may include taking
increase the amount of levodopa passing the BBB or levodopa at least half an hour before meals, crushing
to delay the breakdown of dopamine in the brain. the levodopa and suspending it in sparkling water or
Figure 1 shows the routes by which levodopa is taking dispersible levodopa formulations, and com-
metabolised. bining these approaches with protein reduction dur-
A decarboxylase inhibitor is given to prevent the ing day-time hours.[20-22]
peripheral metabolism of levodopa. To prevent the
breakdown of dopamine in the brain, a monoamine 2.2 Central Pharmacokinetics and
oxidase (MAO) inhibitor, such as selegiline (depre- Pharmacodynamics of Levodopa
nyl), can be given. A third method to increase the
Current evidence suggests that wearing-off phe-
Periphery Brain nomena are closely related to the extent of degenera-
tion of dopaminergic neurons. As neurodegenera-
tion progresses, the neuronal dopamine stores be-
3-OMD 3-OMD
come increasingly dependent on the precursor
COMT COMT levodopa. Levodopa turnover in the remaining neu-
Blood-brain barrier

Levodopa Levodopa
rons is possibly increased, and the newly synthesis-
ed dopamine is released more rapidly. Moreover,
AADC AADC
extraneuronal dopamine synthesis, by glia cells and
Dopamine Dopamine other levodopa decarboxylase-containing cells, may
MAO-B COMT become more important for intrasynaptic dopamine
DOPAC 3-MT
levels. Thus, the functional status of the dopaminer-
gic neurons begins to reflect the availability of exog-
COMT MAO-B enous levodopa, and the neurons lose their ability to
HVA
maintain an adequate dopaminergic tone by the end
Fig. 1. Metabolism of levodopa. 3-MT = 3-methoxytyramine; of each levodopa dosing interval.[23]
3-OMD = 3-O-methyldopa; AADC = aromatic amino acid decarbox-
ylase; COMT = catechol-O-methyltransferase; DOPAC =
The view that wearing-off reflects the loss of
3,4-dihydroxyphenylacetic acid; HVA = homovanillic acid; MAO-B = functional dopaminergic neurons is supported by a
monoamine oxidase type B. study comparing the kinetic profiles of the reappear-

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 479

ance of parkinsonian symptoms and circulating the antiparkinsonian effect of levodopa remains
levodopa concentrations in patients with Parkin- largely unchanged with disease progression, but the
son’s disease after abrupt cessation of optimal intra- threshold for the induction of dyskinesia is gradually
venous levodopa administration.[24] From studies in reduced as patients move to a more advanced dis-
a range of patients, varying from patients who had ease stage.[27] Thus, the therapeutic window for
never been treated to those experiencing on-off fluc- levodopa progressively decreases with disease se-
tuations, it was concluded that the progressive verity and becomes almost nonexistent in patients
neurodegeneration in Parkinson’s disease reduces with on-off fluctuations (see figure 2).
the ability of the brain to buffer the fluctuations in
synaptical concentrations of levodopa. This idea 3. Influence of Age and Duration/Dose
was also supported by the fluorodopa PET study, of Levodopa on Motor Complications
which showed less striatal accumulation of [18F]-
dopa in patients experiencing motor fluctuations It has been suggested that the rate at which re-
than in those with a stable response.[25] sponse fluctuations after levodopa treatment devel-
However, wearing-off is not only caused by pre- op is associated with the age of the patient at the
synaptic neuronal degeneration; it is also influenced onset of IPD. A study by Kostic et al.[28] showed that
by postsynaptic alterations.[24] Patients with Parkin- response fluctuations developed more rapidly in
son’s disease who received a continuous apomor- patients with young-onset disease (onset of Parkin-
phine infusion showed clear differences in the rate son’s disease between 21 and 40 years) than in a
of decline of their motor responses, with a faster matched population of older patients (>40 years at
decline occurring in the more severely affected onset of Parkinson’s disease). About 30% of the
patients.[26] These findings suggest a postsynaptic patients with young-onset disease had developed
involvement of wearing-off phenomena, because response fluctuations after 1 year of levodopa treat-
apomorphine mainly acts via postsynaptic dopamine ment.
receptors. Other determinants of response fluctuations have
been suggested to include the severity of disease at
Response variations of the on-off type and dys- the onset of treatment, the duration of disease and
kinesias are at least partially caused by hypersensi- the duration and the dose of levodopa therapy.[29,30]
tivity of postsynaptic receptors, resulting from expo- Response fluctuations were reported to be more
sure to fluctuating dopamine levels. Fluctuations in
dopaminergic tone are considered to be the result of Dyskinesia threshold
intermittent levodopa administration and the gradual
loss of the buffering capacity of the dopamine sys- Stable
Levodopa concentration

tem as described above. This also explains the tem-


Wearing-off
poral relationship between the occurrence of wear-
ing-off and on-off motor fluctuations. With disease Therapeutic window At random
progression, the shape of the dose-response curve of
levodopa changes dramatically.[27] Patients with ear-
Minimal effective concentration
ly Parkinson’s disease who have never been treated
with levodopa show a smooth, slowly rising dose-
response curve. In patients at more advanced disease
stages, the dose-response curves become increasing-
Stages of IPD
ly steeper to become an all-or-nothing response. In
Fig. 2. Change in the therapeutic window of levodopa related to the
these patients, a small change in the plasma concen- stage of idiopathic Parkinson’s disease (IPD), showing the narrow-
tration of levodopa has a profound effect on the ing of the window (difference between the concentration that has
control of motor symptoms. The threshold dose for therapeutic versus adverse effects) as the disease progresses.

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
480 van Laar

likely to occur in patients who started with levodopa drome in primates. MPTP is known to stimulate the
in Hoehn and Yahr stage I or II. However, another production of neurotoxic substances in the brain that
study could not show any relationship between re- are able to damage dopaminergic neurons in the
sponse fluctuations and the age at onset, presenting substantia nigra.
symptom, duration of illness, stage of the disease at
the time of initiation of levodopa or the mean or last 4.1 Role for Dopamine D1
daily dose of levodopa. Nevertheless, response fluc- Receptor Stimulation
tuations were less likely in patients who were at least
60 years old at the onset of Parkinson’s disease and Shortly after the discovery of dopamine receptor
had started levodopa >2 years after the first symp- heterogeneity, it was recognised that dopamine re-
toms of disease.[31] In keeping with this pattern, it ceptors of the D2 type primarily mediate the motor
was also shown that response fluctuations are a effects of central dopamine stimulation. However,
likely event in those patients who had required a various lines of evidence now support the view that
recent increase in their levodopa dose. dopamine D1 receptors are also able to mediate
A randomised controlled study in previously un- central motor effects. Experimental data, obtained in
treated patients with Parkinson’s disease assessed MPTP-treated primates, support a role for D1 recep-
the effect of delaying the onset of treatment with tors in the motor effects of dopaminomimetic drugs.
levodopa. The study compared the effect of initiat- For instance, studies with selective D1 and D2 ago-
ing treatment with bromocriptine and later adding nists have clearly demonstrated that selective D1
levodopa with initiating treatment with levodopa. A agonists, such as the benzazepine SKF 81297, have
significant decrease in motor complications in fa- profound antiparkinsonian effects in MPTP-lesion-
vour of the patients who used bromocriptine mono- ed animals.[33] Also, the coadministration of a selec-
therapy for as long as possible was reported. The tive D2 agonist, such as bromocriptine or quinpirole
patients receiving bromocriptine alone initially (LY171555), with a selective D1 agonist produced
showed a mean delay in the onset of response fluctu- synergistic antiparkinsonian effects compared with
ations of 4.9 years (n = 25), whereas the group that a D2 agonist alone.[34] Evaluation of the potent selec-
started therapy on levodopa showed a mean delay of tive D1 agonist A-86929 as a new therapeutic mo-
just 2.7 years (n = 29) before onset of response dality in the treatment of Parkinson’s disease has
fluctuations. The percentage of patients with fluctu- recently been proposed, particularly in patients with
ations increased directly after the start of levodopa dyskinesias.[35]
therapy.[32] This study clearly showed the benefit of It is generally recognised that levodopa and apo-
delaying levodopa as long as possible with respect morphine are currently the most potent drugs avail-
to the reduced risk of developing response fluctua- able for the treatment of Parkinson’s disease.[36,37]
tions. The striking fact is that these drugs are both D1 and
D2 receptor agonists. This seems to support the
4. Preclinical Data on the Treatment of synergistic effect of D1 receptor agonists on the
Response Fluctuations action of D2 receptor agonists. Unfortunately, no
clinical comparisons have been done between selec-
Part of the evidence for the pathophysiology of tive D2 and combined D1/D2 agonists. Looking at
motor fluctuations (see section 2) is derived from the monotherapy trial data of dopamine receptor
experimental studies in primates treated with agonists that have different affinities for D1 and D2
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine receptors versus placebo, no clinically significant
(MPTP). MPTP is an impurity in certain illicit drugs differences in effect could be determined.[38] Unfor-
and has been identified as the cause of parkinsonian tunately, these trials had different designs and so a
syndromes in addicts who had been using them. real comparison is not possible, and it may be a
MPTP is able to induce a stable parkinsonian syn- result of methodological differences that no clear

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 481

differences could be detected. Nevertheless, the terioration. This can be achieved via several routes,
preclinical data with respect to the affinities of cur- which are summarised in figure 3. The initial ap-
rently available dopamine agonists for dopamine proach is to optimise dopamine agonist therapy as
receptor subtypes do not provide enough guidance far as possible (see section 5.1.1) by titrating up to
to select a particular agent. maximally tolerated dosages of the individual ago-
nists. If this is ineffective, an attempt should be
4.2 Possible Influence of Priming made to increase the duration of effect of levodopa
Several MPTP experiments with primates have (see section 5.1.2).
provided the insight that the potential of levodopa to
induce dyskinesia is much higher than that of the 5.1.1 Optimisation of Dopamine Agonists
different dopamine agonists.[39,40] Once the dyskine- All dopamine agonists share a potent activity at
sias were established by levodopa, treatment with D2 receptors, but they differ with respect to their
any of the dopaminergics could easily induce subse- effects on other types of CNS receptors (including
quent dyskinesia. Selective D1 receptor compounds D1 receptors), their pharmacokinetic profile and the
produced less dyskinesia than selective D2 receptor optimal daily doses (table II). Differences in the
compounds at lower doses, and both produced less elimination half-life are the main determinants of
dyskinesia compared with levodopa. Once primed, the duration of action of orally administered dop-
the message for the production of dyskinesia seems amine agonists, which ranges from 4–8 hours for
to be permanently stored. The same phenomenon bromocriptine to 70–105 hours for cabergoline.[47]
can be seen in patients with Parkinson’s disease. Most dopamine agonists have to be administered
Primate research has shown that levodopa is able to 3–4 times per day to achieve stable responses over
change the balance in the direct and indirect path- 24 hours. Agonists circumvent some of the limita-
ways in the striatum. Levodopa increases direct
tions inherent in the use of levodopa. Directly acting
pathway firing and decreases indirect pathway fir-
dopamine agonists do not require metabolic activa-
ing, leading to dyskinesia.[41]
tion and are therefore not dependent on the function-
These data should encourage great caution with
al status of presynaptic dopaminergic neurons. In
the practice of administering test doses of levodopa
addition, they do not compete with amino acids for
in order to see if a patient is levodopa responsive.
absorption sites and are not subject to peripheral
This can be the start of very difficult-to-handle
metabolism, which may interfere with the effects of
dyskinesia and should therefore be avoided.
levodopa. Finally, dopamine agonists with a long
5. Clinical Treatment Strategies First step
Try to optimise dopamine agonist doses
In the following sections, the rationale for the use (combined with reduction in levodopa dose)
of different drugs or treatment strategies and their
efficacy in the management of response fluctuations
If not effective enough
in patients with Parkinson’s disease will be re-
viewed. Readers should also refer to several review Secondary steps
articles on the treatment of response fluctua- Try to increase duration of effect of levodopa:
tions.[42-46] • increase frequency of levodopa doses
• increase levodopa dose per administration
• use slow-release levodopa formulations
5.1 Wearing-Off Phenomena • use MAO-B and/or COMT inhibitors

Fig. 3. Treatment schedule for wearing-off phenomena in patients


In general, wearing-off can be treated adequately with Parkinson’s disease who are receiving levodopa. COMT =
by striving for a longer duration of action of the catechol-O-methyltransferase; MAO-B = monoamine oxidase type
dopaminomimetics to overcome the end-of-dose de- B.

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
482 van Laar

Table II. Overview of the pharmacokinetic, pharmacodynamic and therapeutic properties of currently available dopamine receptor
agonists[48]
Agonist Receptor profile Pharmacokinetics Optimal daily dose
D1 D2 serotonin bioavailability (%) tmax (min) t1/2 (h) (mg)
Apomorphine + +++ + 100 5–15 0.5 3–8 mg/h (infusion)
(subcutaneous)
Bromocriptine – ++ + 8 70–100 4–8 15–40
Cabergoline + +++ + 60–80 120–240 70–105 0.5–5
Pergolide + +++ + 20 60–120 27 3–5
Pramipexole 0 +++ 0 >90 60–180 8–12 3–5
Ropinirole 0 +++ 0 55 90 6 12–24
0 = no binding affinity; t1/2 = half-life; tmax = time to peak concentration; – indicates antagonist activity; + indicates binding affinity from
minimal (+) to strong (+++).

half-life theoretically provide a more natural tonic antiparkinsonian effect. Published data from the
stimulation of dopamine receptors. dopamine agonist monotherapy trials versus
Dopamine agonists have proven to be useful levodopa confirm this idea only partially. Based on
agents in the treatment of levodopa-induced re- large, randomised, controlled trials, bromocriptine
sponse fluctuations in patients with advanced proved to be less efficacious than levodopa as
Parkinson’s disease. All currently available oral monotherapy.[54,55] The statistical differences were
dopamine agonists have been studied primarily as already clear within 1 year.[56-59] Ropinirole mono-
adjunct therapy to levodopa. All agonists are able to therapy effectively treated 50% of the patients for 3
provide significant improvement in the Activities of years and 30% of the patients for 5 years, which is
Daily Living scores and/or motor scores after add- still a lower efficacy than is seen with levodopa, but
on therapy (see review by Clarke[49]). Reductions in not statistically significantly lower.[56] All studies
levodopa dosage between 175 and 265 mg/day can that have compared dopamine agonist monotherapy
be achieved with concomitant administration of with levodopa monotherapy have shown that
dopamine agonists. The main adverse effects are levodopa seems to be slightly more effective,
gastrointestinal complaints, postural hypotension,
though this was not statistically significant using the
confusion and hallucinations; these do not differ
Unified Parkinson’s Disease Rating Scale (UPDRS)
greatly between the different dopamine agonists. If
III scores except in the study with bromo-
gastrointestinal or hypotensive adverse effects arise,
criptine.[47,56-59] The same studies showed a signif-
domperidone up to 90 mg/day can be added to
reduce the peripheral dopamine agonistic-mediated icant reduction in the occurrence of response fluctu-
aspect of these effects. Pergolide is an extensively ations and dyskinesia and a significant increase in
studied add-on dopamine agonist, mainly in com- the latency of onset of fluctuations and dyskinesias
parison with bromocriptine, and has been shown to in patients receiving dopamine agonists (i.e. 5–10%
have a statistical benefit over bromocriptine.[50-53] of patients receiving dopamine agonist monotherapy
When using adjunct therapy with dopamine ago- developed response fluctuations/dyskinesia vs
nists, physicians have to titrate to maximal tolerated 31–36% in the levodopa groups). These important
doses of the individual agonists in combination with data have shown that dopamine agonists as mono-
a maximal reduction of the levodopa dose. If these therapy are effective and that there are no significant
two strategies are not combined optimally, patients differences among the agonists. The effects are
will very probably experience an increase of dys- comparable to those achieved with levodopa during
kinesia and no significant reduction in off time. the first years of treatment, and their preventive
As a class, dopamine agonists are generally con- effect on the occurrence of response fluctuations is
sidered less effective than levodopa in terms of proven. On this basis, dopamine agonist should be

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 483

prescribed as first-line therapy if possible and if the duration of action of levodopa. The two pos-
tolerated. sibilities are MAO-B or COMT inhibition.
Selegiline, a selective MAO-B inhibitor, blocks
5.1.2 Increasing the Duration of Effect of the degradation of synaptic dopamine in the basal
Levodopa Doses ganglia, which theoretically increases the duration
The easiest way to increase the duration of a drug of action of dopamine. Five monotherapy trials have
effect per dose is to increase the dose. This is only proven that the theoretical advantage of selegeline
acceptable for levodopa if peak dose dyskinesias are also can be seen in clinical practice, because patients
not already present or result from the dose increase. showed improved UPDRS scores.[62] However, the
If dyskinesias are present or result from the dose effects are small, and the duration of effects lasted
increase, the other option is to prescribe the original from 3 months up to 1 year. Add-on therapy to
dose more frequently to avoid peak dose concentra- levodopa dosages made a reduction of the levodopa
tions and to overcome the end-of-dose deterioration. dose possible; however, this was not associated with
Different technologies have been used to produce enhanced symptomatic control.
controlled-release preparations of levodopa (e.g. The other possibility is COMT inhibition. In
Sinemet®1 CR [carbidopa-levodopa] and Madopar® many European countries, the only drug currently
Hydrodynamically Balanced System [HBS]).[60] available is entacapone, a peripherally acting
These formulations provide a mean increase of 1–2 COMT inhibitor. The other COMT inhibitor, tol-
hours in the duration of single-dose effects. How- capone, was withdrawn from the market in the EU
ever, there are some drawbacks. The plasma concen- because of severe hepatotoxicity, leading to death in
trations of levodopa build up slowly, leading to a a few patients. In Europe, tolcapone is only avail-
delayed on time, and patients do not experience the able in Switzerland, and use must be accompanied
‘kick on’ effect of a normal-release levodopa prepa- by intensive monitoring of liver function. The drug
ration. The total daily dose of slow-release formula- is also available in the US, where the labelling was
tions has to be increased by 30–50% of the original changed. Frequent monitoring of liver function is
dose in order to achieve the same clinical response, mandatory in the US; serum levels of alanine amino-
because the absorption of these formulations is less transferase (ALT) and aspartate aminotransferase
than with the normal-release levodopa prepara- (AST) should be determined at baseline and then
tions.[61] The effect during daytime is very modest, every 2 weeks for the first year of therapy, every 4
and the best way to use slow-release formulations is weeks for the next 6 months and every 8 weeks
to add them as evening doses, just before falling thereafter. Another adverse event responsible for
asleep, in order to treat nocturnal akinesia and there- many withdrawals from tolcapone was diarrhoea
by promote sleep. Sometimes a single evening dose (3–10%). Tolcapone and entacapone prevent the
of a slow-release formulation is even effective in (peripheral) conversion of levodopa to 3-O-
preventing early-morning akinesia or dystonia. The methyldopa, which delivers more levodopa at the
reason for this is unclear and does not seem to be BBB. The percentage of levodopa from each tablet
explained by the small increase of the elimination that enters the central compartment is therefore in-
half-life. creased, creating a longer duration of action.[63,64]
Dose fractioning does reduce dyskinesia. How- The official indication to prescribe entacapone is
ever, it worsens response fluctuations because most the presence of wearing-off symptoms. COMT in-
of the reduced doses no longer elicit a clinical re- hibitors can be prescribed only as adjunct therapy,
sponse or elicit a very short response. because monotherapy with COMT inhibitors in
Finally, using enzymatic inhibition of conversion Parkinson’s disease is not effective (unpublished
steps from levodopa to its metabolites can increase observations). The benefit of entacapone is a quick-

1 Use of tradenames is for product identification only and does not imply endorsement.

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
484 van Laar

er onset of effect of levodopa without increasing the might be considered a candidate for stereotactic
peak concentrations and a longer duration of effect. surgery (see section 6).
The mean increase in on time is about 5% per day.
Other studies showed an extra duration per levodopa 5.2.1 Rescue Therapy
dose of about 30 minutes and a total increase of Apomorphine has been known since 1884 as an
mean on time per day of about 2 hours.[65-67] Enta- emetic with antiparkinsonian properties. A very po-
capone has to be coupled to each levodopa dose tent dopamine agonist, it has to be administered
because of its very short half-life (1–2 hours). parenterally because of a strong first-pass effect.
One study has been published in which selegiline Since the introduction of domperidone, a peripheral
and entacapone were combined as adjunct therapy to dopamine antagonist that prevents the emetic action
levodopa. The combination was more effective than of apomorphine on the chemoreceptor trigger zone
each individual adjunct therapy.[68] However, outside the BBB, apomorphine has been used in
selegiline plus entacapone may induce dyskinesia, clinical practice for the treatment of at-random re-
and selegiline also carries the risk of provoking sponse fluctuations. It can be used in two ways:
hallucinations.[68] If dyskinesias occur with enta- intermittent treatment for off periods (as rescue ther-
capone therapy, the original levodopa dose should apy) and continuous infusion by pump via the sub-
be reduced by 15–30%. cutaneous or intravenous route.
Intermittent injections are helpful, because the
absorption half-life of apomorphine is short (min-
5.2 At-Random Response Fluctuations utes), leading to an onset of effect within 5–10
minutes. This gives patients a form of ‘kick on’
Although wearing-off phenomena can be treated effect. The mean duration of effect is about 45–60
successfully, this is certainly not the case with at- minutes. The optimal dose is determined by increas-
random response fluctuations. If the fluctuations ing the dose in 1mg increments, starting with 1mg
occur unpredictably, it is difficult to create a more per injection up to a maximum of 10mg per injec-
predictive and stable motor functioning. Figure 4 tion. Patients should not need more than five injec-
summarises the possible approaches before a patient tions per day to overcome their sudden off periods.
If more than five injections are required, continuous
Rescue therapy: infusion of apomorphine is a better option in order to
• subcutaneous apomorphine injections
• soluble levodopa formulations (drink)
create stable plasma concentrations, which are im-
portant in the treatment of response fluctua-
tions.[69,70]
Optimisation of continuous dopamine receptor stimulation: Apomorphine injection therapy has recently be-
• subcutaneous apomorphine by pump/portacath
• intraduodenal levodopa
come available in some countries as a disposable
penject system, which is very convenient. In situa-
tions where this is not available, physicians can
Reduce dyskinesias: prescribe syringes – the same as those used by
• high-dose amantadine
• D2 antagonists (tiapride/clozapine)
diabetic patients – to inject the apomorphine subcu-
taneously. Patients generally can inject themselves
without difficulty. Apomorphine injections do not
Stereotactic surgery: disturb the daily medication regimen but rather sup-
• bilateral STN stimulation
• unilateral pallidotomy or pallidal stimulation
plement it to prevent gaps in motor functioning. If
patients do not like to inject themselves, they can
Fig. 4. Approaches to the management of at-random response
fluctuations and dyskinesias in patients with Parkinson’s disease
use apomorphine intranasal sprays. The only disad-
who are receiving levodopa. STN = subthalamic nucleus stimula- vantage is that black crustae can appear in the nasal
tion. cavity; this is reversible by stopping the intranasal

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 485

application.[71,72] These intranasal sprays can be pro- A good alternative to intravenous infusion of
duced easily by local pharmacists. levodopa is the intraduodenal route, which is also
Another alternative to apomorphine injection suitable for patients not experiencing fluctuations
therapy is a soluble levodopa solution, which can be but with dysphagia.[74,75] The intraduodenal route is
made by dissolving crushed levodopa tablets in car- tolerated quite well if a percutaneous endoscopic
bonated water and adding vitamin C to prevent auto- gastrostomy is created.
oxidation. However, the disadvantage of soluble Alternatively, parenteral administration of pro-
levodopa is the overall increase of the levodopa drugs of levodopa, the methyl and ethyl esters, can
load, with a possible negative impact on motor be considered. These prodrugs can be given intra-
complications. Patients can also use Madopar® Dis- venously or subcutaneously. Good results have been
pers, which has about the same effect. However, described with this strategy.[76]
Madopar® Dispers does not provide the ‘kick on’ Another approach is to stimulate dopamine re-
effect of apomorphine, because the latency of onset ceptors continuously by administering apomorphine
of effect is about 30 minutes. The advantage of the subcutaneously via a pump. The most important
Madopar® Dispers on the other hand is the longer indication for this treatment is response fluctuations
duration of action, which may last up to 2 hours.[73] otherwise unresponsive to optimal oral treatment,
leading to more than five or six sudden, unpredict-
5.2.2 Continuous Stimulation of able off periods per day. Many of these patients
Dopamine Receptors currently undergo stereotactic surgery (see section
Clinical trials have shown that motor response 6). However, those patients who do not fulfil the
fluctuations in patients with Parkinson’s disease can rather strict criteria for stereotactic surgery, with
be improved by avoiding fluctuations in the supply mood or cognitive problems, or who want to post-
of levodopa to the remaining nigrostriatal dopamin- pone surgery can be treated very well with a subcu-
ergic neurons. The validity of this approach has been taneous apomorphine pump.
demonstrated in studies using continuous intrave- The starting dose for subcutaneous infusion is 1
nous infusion of levodopa to achieve stable plasma mg/hour, which is the same as for intravenous deliv-
concentrations and a constant supply to the brain ery, and can be gradually increased by 1 mg/hour up
(see review by Coleman[61]). Patients with on-off to maximal 10 mg/hour every 2 hours. The delivery
fluctuations generally remain on for the duration of should be as continuous as possible, which means
the infusion. that the infusion rate should be kept the same as
Obviously, continuous intravenous administra- much as possible. The infusion rate can only be
tion is a highly impractical way of treating patients adapted, for a few hours, in the case of severe
with a chronic disease. Also, continuous infusion afternoon motor worsening. Patients may develop
has its limitations. It was shown that constant-rate tolerance to apomorphine if the infusion is given 24
levodopa infusions, able to turn the patients on hours/day.[77] In these cases, the infusion should be
within 1–3 hours of starting the infusion, were una- stopped at night or continued at a very low rate to
ble to keep patients on around-the-clock.[74] The off provide some night-time relief and to prevent the
periods still occurred, but later in the day, indicating catheter from becoming sludged. Using the pump
a diurnal pattern not related to stable plasma concen- this way, patients can also give themselves boluses
trations. Another influence was the increasing of apomorphine during the night.
LNAA load over the day, which seemed to be at The main adverse event associated with this ap-
least partly related to the off periods. The severity of proach is the development of subcutaneous nodules,
the response fluctuations did not correlate with the caused by a toxic effect of apomorphine on the
severity of the disease but rather with the size of the subcutaneous tissue. These can be treated and pre-
oral levodopa doses taken before. vented by dilution of the apomorphine solution

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
486 van Laar

(from 1% to, for example, 0.5%), daily change of the of amantadine may be a possible neuroprotective
injection site and treatment of the injection sites effect. A recently published study has claimed a
with hydrocortisone cream (0.1%) after the needle survival advantage in patients treated with amanta-
has been removed.[78] Dilution of the apomorphine dine.[84]
is the most powerful way to reduce subcutaneous Dopamine antagonists can also be used to reduce
nodules and should be prescribed therefore as a fluctuations. Clozapine is the best-studied com-
standard. pound and has been studied in dosages between 6.25
The alternative to the subcutaneous infusion of and 50 mg/day. It may be an alternative if amanta-
apomorphine is intravenous administration, which dine is not tolerated or effective.[85-87]
can be achieved by a portacath in the subclavian
artery. This may be a good alternative for patients 6. Stereotactic Surgery
who have developed severe skin nodules, which
make inserting new needles painful and diminish the Patients whose response fluctuations cannot be
subcutaneous absorption of apomorphine. Good re- controlled with the strategies mentioned in section 5
sults have been reported with these portacath may be candidates for stereotactic surgery, which
systems and apomorphine.[79] has shown a revival during the past decade.
Two techniques are available: deep brain stimu-
5.3 Dyskinesias lation and lesioning. The major advantage of stimu-
lation over lesioning is the reversibility of the tech-
If dyskinesias occur, dose fractionating should be nique. The disadvantage of stimulation is the exten-
avoided, as although it reduces dyskinesia, it wors- sive postoperative management of the electrode
ens response fluctuations (see section 5.2.1). (Sub)- settings required to achieve an optimal result. There
threshold doses have a very short duration of action, are three main targets to which one of these two
if any effect at all, especially in the advanced stages techniques can be applied: the ventral intermediate
of the disease.[80] nucleus of the thalamus, the subthalamic nucleus
Two main approaches can be helpful in improv- and the internal part of the globus pallidus. The
ing dyskinesias: dopamine antagonists and amanta- correct patient selection is crucial to achieve an
dine. Amantadine is a glutamate NMDA receptor optimal effect, as is selection of the appropriate
antagonist. The usefulness of NMDA antagonists in target. Bilateral subthalamic nucleus stimulation is
the suppression of motor complications due to long- currently the most popular approach, in part because
term levodopa therapy is supported by a recent of the positive results and in part because of the
experimental study in MPTP-induced parkinsonism possibility of reducing dopaminergic medication
in monkeys using the selective NMDA antagonist postoperatively, which significantly reduces drug-
LY235959.[81] Recently, several publications have related adverse events.
supported this concept. Dyskinesia severity was re- In addition to experiencing uncontrollable re-
duced by 60% in a group of 14 patients with a mean sponse fluctuations or tremor, patients must meet
amantadine dose of 350mg.[82] The authors also some additional criteria. There must be a good re-
showed a direct relationship between the plasma sponse to levodopa. If the patient does not show a
concentration of amantadine and the dyskinesia re- good response to levodopa, no improvement with
duction, which could be described by a logarithmic surgery can be expected. No behavioural, mood or
function. A very important finding was that the cognitive impairments should be present, because
positive effects lasted for at least 1 year,[83] which they have a negative impact on the final results.
provides enough clinical evidence to use amanta- Lesioning can only be done unilaterally, because
dine in higher doses for dyskinesia. However, in otherwise too many adverse effects, such as speech
elderly patients, these higher doses of amantadine and swallowing difficulties as well as pyramidal
increase the risk for hallucinations. An extra benefit disorders, occur.

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 487

Table III summarises the indications and magni- Acknowledgements


tudes of effect of the different techniques. Adverse The author has not received any funding for the prepara-
effects more specific to the stereotactic procedure tion of this manuscript. There are no conflicts of interest that
are infections, intracranial haematoma and lead could be relevant to the contents of this manuscript.
breakage or disconnection, which induce permanent
neurological impairment in about 3% of all patients. References
1. Morrish PK, Sawle GV, Brooks DJ. The rate of progression of
All these data are extensively discussed in a special Parkinson’s disease: a longitudinal [18F]DOPA PET study. In:
Battistin L, Scarlato G, Caraceni T, et al., editors. Advances in
supplement of Movement Disorders published re- neurology. Vol. 69. Philadelphia (PA): Lippincott-Raven Pub-
cently.[88] lishers, 1996: 427-31
2. Leenders KL. Pathophysiology of movement disorders studied
using PET. J Neural Transm Suppl 1997; 50: 39-46
3. Hughes AJ, Daniel SE, Kilford L, et al. Accuracy of the clinical
7. Conclusions diagnosis of idiopathic Parkinson’s disease: a clinico-patho-
logical study of 100 cases. J Neurol Neurosurg Psychiatry
1992; 55: 181-4
Currently, the most important goal in the treat- 4. Marsden CD, Parkes JD. Success and problems of long-term
ment of motor complications associated with levodopa therapy in Parkinson’s disease. Lancet 1977; I: 345-9
5. Block G, Liss C, Reines S, et al. Comparison of immediate-
levodopa is achieving stable pharmacokinetic release and controlled-release carbidopa/levodopa in PD: a
profiles as much as possible.[89] Therefore, regular multicenter 5-year study. Eur Neurol 1997; 37: 23-7
6. Golbe LI. Young-onset Parkinson’s disease: a clinical review.
administration of levodopa in combination with Neurology 1991; 41: 168-73
dopaminomimetics, which all have a relatively long 7. Schrag A, Quinn N. Dyskinesias and motor fluctuations in
Parkinson’s disease: a community based study. Brain 2000;
half-life, seems to be the best strategy. It is impor- 123: 2297-305
8. Fahn S. Fluctuations of disability in Parkinson’s disease: patho-
tant to delay the use of short-acting formulations of physiological aspects. In: Marsden CD, Fahn S, editors. Move-
levodopa for as long as possible, especially in ment disorders. London: Butterworth Scientific, 1982: 123-45
9. Nutt JG, Holford NH. The response to levodopa in Parkinson’s
patients with young-onset IPD, in order to delay the disease: imposing pharmacological law and order. Ann Neurol
onset of response fluctuations. However, it is also 1996; 39: 561-73
10. Melamed E, Bitton V, Zelig O. Delayed onset of response to
clear that stable pharmacokinetics do not solve all single doses of L-dopa therapy. Clin Neuropharmacol 1986; 9:
problems. The major challenge at this time is to gain 182-8
11. Melamed E, Bitton V, Zelig O. Episodic unresponsiveness to
a better insight into the pharmacodynamic changes single doses of L-dopa in parkinsonian fluctuators. Neurology
accompanying the response fluctuations. Only if we 1986; 36: 100-3
12. Marsden CD, Parkes JD, Quinn N. Fluctuations of disability in
get a better understanding of these mechanisms will Parkinson’s disease: clinical aspects. In: Marsden CD, Fahn S,
we be able to make real progress in the treatment of editors. Movement disorders. London: Butterworth Scientific,
1982: 96-122
response fluctuations. 13. Lees AJ, Shaw KM, Stern GM. Off-period dystonia and on-
period choreoathetosis in levodopa-treated Parkinson’s disease
Table III. Indications and results of the different stereotactic targets
[letter]. Lancet 1977; II: 1034
in patients with motor response fluctuations to levodopa
14. Muenter MD, Sharpless NS, Tyce GM, et al. Patterns of
dystonia in response to L-dopa therapy for Parkinson’s dis-
Decrease in Subthalamic Globus Ventral ease. Mayo Clin Proc 1977; 52: 163-74
nucleus pallidus intermediate 15. Melamed E. Early-morning dystonia: a late side effect of long-
nucleus of the term levodopa therapy in Parkinson’s disease. Arch Neurol
thalamus 1979; 36: 308-10
Tremor +++ ++ +++ 16. Nausieda PA, Weiner WJ, Klawans HL. Dystonic foot response
of parkinsonism. Arch Neurol 1980; 37: 132-6
Akinesia +++ ++ 0
17. Curtis L, Lees AJ, Stern GM, et al. Effect of L-dopa on course
Rigidity +++ ++ + of Parkinson’s disease. Lancet 1984; II: 211-2
Dyskinesia + +++ + 18. Poewe WH, Lees AJ, Stern GM. Dystonia in Parkinson’s dis-
Levodopa dose ++ 0 0 ease: clinical and pharmacological features. Ann Neurol 1988;
23: 73-8
0 = no effect; + indicates minimal effect; ++ indicates moderate
19. Nutt JG, Woodward WR, Carter JH, et al. Influence of fluctua-
effect; +++ indicates marked effect.
tions of plasma large neutral amino acids with normal diets on

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
488 van Laar

the clinical response to levodopa. J Neurol Neurosurg Psychia- 38. Lledo A. Dopamine agonists: the treatment for Parkinson’s
try 1989; 52: 481-7 disease in the XXI century? Parkinsonism Relat Disord 2001;
20. Baruzzi A, Cantin M, Riva R. Influence of meal ingestion time 7: 51-8
on pharmacokinetics of orally-administered levodopa in 39. Bedard PJ, Di Paolo T, Falardeau P, et al. Chronic treatment
parkinsonian patients. Clin Neuropharmacol 1987; 10: 527-37 with L-dopa, but not bromocriptine, induces dyskinesia in
21. Carter JH, Nutt JH, Woodward WR. Amount and distribution of MPTP-parkinsonian monkeys. Brain Res 1986; 379: 294-9
dietary protein affects clinical response to levodopa in Parkin- 40. Pearce RK, Banerji T, Jenner P, et al. De novo administration of
son’s disease. Neurology 1990; 39: 401-4 ropinirole and bromocriptine induces less dyskinesia than L-
22. Kurth MC, Tetrud LW, Irwin I, et al. Oral levodopa/carbidopa dopa in the MPTP-treated marmoset. Mov Disord 1998; 13:
solution versus tablets in Parkinson’s patients with severe 234-41
fluctuations: a pilot study. Neurology 1993; 43: 1036-9 41. Obeso JA, Rodriguez-Oroz MC, Rodriguez M, et al. Patho-
23. Fabbrini G, Juncos J, Mouradian MM, et al. Levodopa pharma- physiology of the basal ganglia in Parkinson’s disease. Trends
cokinetic mechanisms and motor fluctuations in Parkinson’s Neurosci 2000; 23 Suppl. 10: S8-S19
disease. Ann Neurol 1987; 21: 370-6 42. Marsden CD. Parkinson’s disease. J Neurol Neurosurg Psychia-
24. Fabbrini G, Mouradian MM, Juncos JL, et al. Motor fluctua- try 1994; 57: 672-81
tions in Parkinson’s disease: central pathophysiological mech- 43. Wolters EC, Tissingh G, Bergmans PLM, et al. Dopamine
anisms. Pt I. Ann Neurol 1988; 24: 366-71 agonists in Parkinson’s disease. Neurology 1995; 45 Suppl. 3:
25. Mouradian MM, Juncos JL, Fabbrini G, et al. Motor fluctua- S28-34
tions in Parkinson’s disease: central and pathophysiological 44. Uitti RJ, Ahlskog JE. Comparative review of dopamine receptor
mechanisms. Pt II. Ann Neurol 1988; 24: 372-8 agonists in Parkinson’s disease. CNS Drugs 1996; 5 (5):
26. Leenders KL, Palmer AJ, Quinn N, et al. Brain dopamine 369-88
metabolism in patients with Parkinson’s disease measured 45. Stocchi F, Nordera G, Marsden CD. Strategies for treating
with PET. J Neurol Neurosurg Psychiatry 1986; 49: 853-60 patients with Parkinson’s disease with disastrous fluctuations
27. Bravi D, Mouradian MM, Roberts JW, et al. Wearing-off and dyskinesias. Clin Neuropharmacol 1997; 20 (2): 95-115
fluctuations in Parkinson’s disease: contribution of postsynap- 46. Olanow CW, Watts RL, Koller WC. An algorithm for the
tic mechanisms. Ann Neurol 1994; 36: 27-31 management of Parkinson’s disease (2001): treatment guide-
28. Kostic V, Przedborski S, Flaster E, et al. Early development of lines. Neurology 2001; 56 Suppl. 5: S1-S88
levodopa-induced dyskinesias and response fluctuations in 47. Kulisevsky J, Lopez-Villegas D, Garcia-Sanchez C, et al. A six-
young-onset Parkinson’s disease. Neurology 1991; 41: 202-5 month study of pergolide and L-dopa in de novo Parkinson’s
29. Horstink MWIM, Zijlmans JCM, Pasman JW, et al. Which risk disease patients. Clin Neuropharmacol 1998; 21: 358-62
factors predict the levodopa response in fluctuating Parkin- 48. Watts RL. The role of dopamine agonists in early Parkinson’s
son’s disease. Ann Neurol 1990; 27: 537-43 disease. Neurology 1997; 49 (1 Suppl. 1): S34-48
30. De Jong GJ, Meerwaldt JD, Schmitz PIM. Factors that influence 49. Clarke CE. Dopamine agonist therapy in advanced Parkinson’s
the occurrence of response variations in Parkinson’s disease. disease. Neurology Rev Int 1998; 2: 1-5
Ann Neurol 1987; 22: 4-7 50. Boas J, Worm-Petersen J, Dupont J, et al. The levodopa dose-
31. Roos RAC, Vredevoogd CB, Van der Velde EA. Response sparing capacity of pergolide compared to that of bromo-
fluctuations in Parkinson’s disease. Neurology 1990; 40: criptine in an open-label, cross-over study. Eur J Neurol 1996;
1344-6 3: 44-9
32. Montastruc JL, Rascol O, Senard JM, et al. A randomised 51. Olanow CW, Fahn S, Muenter M, et al. A multicenter double-
controlled study comparing bromocriptine to which levodopa blind placebo-controlled trial of pergolide as an adjunct to
was later added, with levodopa alone in previously untreated Sinemet in Parkinson’s disease. Mov Disord 1994; 9 (1): 40-7
patients with Parkinson’s disease: a five year follow-up. J 52. LeWitt PA, Ward CD, Larsen TA, et al. Comparison of
Neurol Neurosurg Psychiatry 1994; 57: 1034-8 pergolide and bromocriptine therapy in parkinsonism. Neurol-
33. Vermeulen RJ, Drukarch B, Sahadat MCR, et al. The selective ogy 1983; 33 (8): 1009-14
dopamine D1 receptor agonist SKF 81297, stimulates motor 53. Pezzoli G, Martignoni E, Pacchetti C, et al. Pergolide compared
behavior of MPTP-lesioned monkeys. Eur J Pharmacol 1993; with bromocriptine in Parkinson’s disease: a multicenter,
235: 143-7 crossover, controlled study. Mov Disord 1994; 9 (4): 431-6
34. Vermeulen RJ, Drukarch B, Sahadat MCR, et al. The dopamine 54. Comparisons of therapeutic effects of levodopa, levodopa and
D1 agonist SKF 81297 and the dopamine D2 agonist LY selegiline, and bromocriptine in patients with early, mild
171555 act synergistically to stimulate motor behavior of Parkinson’s disease: three year interim report: Parkinson’s
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned Disease Research Group in the United Kingdom. BMJ 1993;
parkinsonian rhesus monkeys. Mov Disorder 1994; 9: 664-72 307 (6902): 469-72
35. Grondin R, Bédard PJ, Britton DR, et al. Potential therapeutic 55. Hely MA, Morris JG, Reid WG, et al. The Sydney Multicentre
use of the selective dopamine D1 receptor agonist, A-86929: Study of Parkinson’s disease: a randomised, prospective five
an acute study in parkinsonian levodopa-primed monkeys. year study comparing low dose bromocriptine with low dose
Neurology 1997; 49: 412-26 levodopa-carbidopa. J Neurol Neurosurg Psychiatry 1994; 57
36. Goetz CG, Koller WC, Poewe W, et al. Management of (8): 903-10
Parkinson’s disease: an evidence-based review. Levodopa. 56. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of
Mov Disord 2002; 17 Suppl. 4: S23-37 the incidence of dyskinesia in patients with early Parkinson’s
37. Goetz CG, Koller WC, Poewe W, et al. Management of disease who were treated with ropinirole or levodopa: 056
Parkinson’s disease: an evidence-based review. DA agonists – Study Group. N Engl J Med 2000; 342: 1484-91
non-ergot derivatives: apomorphine. Mov Disord 2002; 17 57. Barone P, Bravi D, Bermejo-Pareja F, et al. Pergolide mono-
Suppl. 4: S83-9 therapy in the treatment of early PD: a randomized, controlled

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)
Levodopa-Induced Response Fluctuations 489

study: Pergolide Monotherapy Study Group. Neurology 1999; 74. Nutt JG, Carter JH, Lea ES, et al. Motor fluctuations during
53: 573-9 continuous levodopa infusions in patients with Parkinson’s
disease. Mov Disord 1997; 12: 285-92
58. Parkinson Study Group G. Pramipexole vs levodopa as initial
treatment for Parkinson disease: a randomized controlled trial. 75. Melamed E, Zoldan J, Galili-Mosberg R, et al. Current manage-
JAMA 2000; 284: 1931-8 ment of motor fluctuations in patients with advanced Parkin-
son’s disease treated chronically with levodopa. J Neural
59. Rinne UK, Bracco F, Chouza C, et al. Early treatment of
Transm Suppl 1999; 56: 173-83
Parkinson’s disease with cabergoline delays the onset of motor
complications: results of a double-blind levodopa controlled 76. Djaldetti R, Melamed E. Levodopa ethylester: a novel rescue
trial. Drugs 1998; 55 Suppl. 1: S23-30 therapy for response fluctuations in Parkinson’s disease. Ann
Neurol 1996; 39: 401-4
60. Poewe WH, Lees AJ, Stern GM. Treatment of motor fluctua-
tions in Parkinson’s disease with an oral sustained-release 77. Gancher GT, Nutt JG, Woodward WR. Time course of tolerance
preparation of L-dopa: clinical and pharmacokinetic observa- to apomorphine in Parkinsonism. Clin Pharmacol Ther 1992;
tions. Clin Neuropharmacol 1986; 9: 430-9 52: 504-10
61. Coleman RJ. Current drug therapy for Parkinson’s disease. 78. Neef C, van Laar T. Pharmacokinetic-pharmacodynamic rela-
Drugs Aging 1992; 2 (2): 112-24 tionships of apomorphine in patients with Parkinson’s disease.
62. Goetz CG, Koller WC, Poewe W, et al. Management of Clin Pharmacokinet 1999; 37: 257-71
Parkinson’s disease: an evidence-based review. MAO-B inhib- 79. Stocchi F, Farina C, Nordera G, et al. Implantable venous
itors for the treatment of Parkinson’s disease. Mov Disord access system for apomorphine infusion in complicated
2002; 17 Suppl. 4: S38-44 Parkinson’s disease [letter]. Mov Disord 1999; 14: 358
63. Kurth MC, Adler CH, Hilaire MS, et al. Tolcapone improves 80. Verhagen Metman L, Van den Munckhof P, Klaassen AAG, et
motor function and reduces levodopa requirement in patients al. Effects of supra-threshold levodopa doses on dyskinesias in
with Parkinson’s disease experiencing motor fluctuations. advanced Parkinson’s disease. Neurology 1997; 49: 711-3
Neurology 1997; 48: 81-7
81. Papa SM, Chase TN. Levodopa-induced dyskinesias improved
64. Parkinson Study Group. Entacapone improves motor fluctua- by a glutamate antagonist in Parkinsonian monkeys. Ann
tions in levodopa-treated Parkinson’s disease patients. Ann Neurol 1996; 39: 574-57
Neurol 1997; 42: 747-55
82. Verhagen Metman L, Del Dotto P, Van den Munckhof P, et al.
65. Rinne UK, Larsen JP, Siden A, et al. Entacapone enhances the Amantadine as treatment for dyskinesias and motor fluctua-
response to levodopa in parkinsonian patients with motor tions in Parkinson’s disease. Neurology 1998; 50: 1323-6
fluctuations. Neurology 1998; 51: 1309-14
83. Metman LV, Del Dotto P, LePoole K, et al. Amantadine for
66. Ruottinen HM, Rinne UK. Entacapone prolongs levodopa re- levodopa-induced dyskinesias: a 1-year follow-up study. Arch
sponse in a one month double blind study in parkinsonian Neurol 1999; 56: 1383-6
patients with levodopa related fluctuations. J Neurol
Neurosurg Psychiatry 1996; 60: 36-40 84. Uitti Rj, Rajput AH, Ahlskog JE, et al. Amantadine treatment is
an independent predictor of survival in Parkinson’s disease.
67. Nutt JG. Effects of COMT inhibition on the pharmacokinetics Neurology 1996; 46: 1551-6
of L-dopa. Adv Neurol 1996; 69: 493-6
85. Bennett JP, Elke R, Landow RN, et al. Suppression of dyskine-
68. Lyytinen J, Kaakkola S, Ahtila S, et al. Simultaneous MAO-B
sias in advanced Parkinson’s disease. Neurology 1993; 43:
and COMT inhibition in L-dopa treated patients with Parkin-
1551-5
son’s disease. Mov Disord 1997; 12: 497-505
86. Trosch RM, Friedman JH, Lannon MC, et al. Clozapine use in
69. Lees AJ. Dopamine agonists in Parkinson’s disease: a look at
Parkinson’s disease: a retrospective analysis of a large mul-
apomorphine. Fundam Clin Pharmacol 1993; 7: 121-8
ticentered clinical experience. Mov Disord 1998; 13: 377-82
70. Colosimo C, Merello M, Albanese A. Clinical usefulness of
87. Durif F, Vidailhet M, Assal F, et al. Low-dose clozapine
apomorphine in movement disorders. Clin Neuropharmacol
improves dyskinesias in Parkinson’s disease. Neurology 1997;
1994; 17: 243-59
48: 658-62
71. van Laar T, Jansen ENH, Essink AWG, et al. Intranasal
88. Deuschl G, Krack P, Volkmann J. Deep brain stimulation for
apomorphine in parkinsonian on-off fluctuations. Arch Neurol
movement disorders. Mov Disord 2002; 7 Suppl. 3: S1-211
1992; 49: 482-4
89. Chase TN. The significance of continuous dopaminergic stimu-
72. Dewey RB, Maraganore DM, Ahlskog JE. A double-blind pla-
cebo-controlled study of intranasal apomorphine spray as a lation in the treatment of Parkinson’s disease. Drugs 1998; 55
rescue agent for off-states in Parkinson’s disease. Mov Disord Suppl. 1: 1-9
1998; 13: 782-7
73. Merello M, Pikielny R, Cammarota A, et al. Comparison of Correspondence and offprints: Dr Teus van Laar, Depart-
subcutaneous apomorphine versus dispersible Madopar laten-
cy and effect duration in Parkinson’s disease patients: a doub-
ment of Neurology, Groningen University Hospital,
le-blind single-dose study. Clin Neuropharmacol 1997; 20: Hanzeplein 1, Groningen, 9700 RB, The Netherlands.
165-7 E-mail: t.van.laar@neuro.azg.nl

© Adis Data Information BV 2003. All rights reserved. CNS Drugs 2003; 17 (7)

You might also like