INTRODUCTION The array of pharmacologic and
surgical treatments available for the treatment of
idiopathic (or Lewy body) Parkinson's disease (PD)
is broader than for any other degenerative disease
of the central nervous system. Management of
individual patients requires careful consideration
of a number of factors including the patient's
symptoms and signs, age, stage of disease, degree of
functional disability, and level of physical
activity and productivity. Treatment can be divided
into nonpharmacologic, pharmacologic, and surgical
therapy.
Nonpharmacologic management of Parkinson's
disease:
Parkinson's disease (PD) is a chronic disorder that
requires broad-based management including patient
and family education, support group services,
general wellness maintenance, exercise, and
nutrition.
EDUCATION The prospect of having a chronic and
progressive neurologic disease is frightening. Many
individuals are familiar with Parkinson's Disease
(PD) and may even have had first-hand acquaintance
with its disabling effects in an affected family
member or friend. Education is essential in order to
provide the patient and family with some
understanding and control over the disorder.
However, caution should be exercised in newly
diagnosed patients with mild symptoms and an
uncertain future with regard to progression; early
overexposure to potentially disturbing material may
be counterproductive. Focused education surrounding
particular symptoms may be more effective and is
available through books written for the lay
audience; national and regional Parkinson's disease
organizations, which publish educational pamphlets
and organize symposia for patients and families; and
the Internet. A useful central information resource
is the "We Move" Foundation at
www.wemove.org .
SUPPORT The emotional and psychologic needs
of the patient and family should be addressed.
Normal reactions of anger, depression, anxiety, and
social and economic concerns often begin with the
onset of the disease and evolve as it progresses.
Support for the caregiver is particularly important
as he or she learns to cope with the increasing
needs of the spouse or parent, or more rarely, a son
or daughter. Support groups are especially valuable
for allowing interactions with other patients or
families with similar experiences and for providing
access to useful educational information.
For patients with early Parkinson's disease (PD),
referral to another affected patient or family may
be less overwhelming than a large support group
composed of patients with advanced disease.
Young-onset PD groups also have been formed in some
locales, and a young-onset PD handbook is available.
There are also some national support organizations.
Referral of the patient and/or family to a
psychologist or psychiatric social worker
experienced in dealing with chronic illness may be
appropriate in some cases. In other instances,
referral for legal, financial, or occupational
counseling is indicated.
EXERCISE AND PHYSICAL THERAPY Regular
exercise promotes a feeling of physical and mental
well-being; it is especially valuable due to the
chronic nature of Parkinson's disease (PD) and its
associated progressive motor limitations. Exercise
will not slow the progression of akinesia, rigidity,
or gait disturbance, but it can prevent or alleviate
some secondary orthopedic effects of rigidity and
flexed posture such as shoulder, hip, and back pain,
and it may also improve function in some motor
tasks.
Stretching and strengthening exercises to improve
flexibility and strength should be emphasized.
Strengthening of extensor muscles can help
counteract the flexed posture of PD. Brisk walks,
swimming, and water aerobic exercises are
particularly useful. Referral to a physical
therapist or exercise group may be a good way to get
patients started in such activities.
A practice parameter from the American Academy of
Neurology (AAN) issued in 2006 concluded that
various physical therapy modalities are probably
effective in improving functional outcome for
patients with PD. These modalities include:
Multidisciplinary rehabilitation with standard
physical and occupational therapy components.
Treadmill training with body weight support. Balance
training and high-intensity resistance training.
Cued exercises with visual (mirror), auditory
(metronome), and tactile feedback. Active music
therapy.
The AAN review found that the magnitude of the
observed functional improvement with these
interventions is small and not sustained after
therapy is discontinued. Nevertheless, many patients
gain lasting confidence and a sense of control over
one aspect of the disease, especially if they have
never engaged in physical activity in the past.
SPEECH THERAPY Dysarthria and hypophonia
are common manifestations of Parkinson's disease
(PD). The practice parameter from the American
Academy of Neurology (AAN) issued in 2006 concluded
that speech therapy for patients with PD may be
helpful in improving speech volume.
This conclusion was based on two small studies that
compared speech therapy with no therapy. One
employed speech therapy emphasizing prosodic
features of pitch and volume reinforced with visual
feedback, while the other employed therapy aimed
solely at maximizing phonatory effort. Both of these
methods were found to be possibly effective by the
AAN.
NUTRITION Elderly patients with chronic
illness are at risk for poor nutrition and weight
loss. Prompt recognition and management of this
problem is important to avoid loss of bone and
muscle mass. No specific diet influences the course
of Parkinson's disease (PD), although certain
recommendations can be made. A high fiber diet and
adequate hydration help manage the constipation of
PD. Large, high-fat meals that slow gastric emptying
and interfere with medication absorption should be
avoided. Dietary protein restriction is not
necessary except in some patients with advanced
disease and motor fluctuations in whom competition
with other amino acids interferes with L-dopa
absorption. There is no evidence at this time that
large doses of vitamin E or other antioxidants are
useful in PD.
DIAGNOSIS OF PD Correct diagnosis is
fundamental to the appropriate therapy of
Parkinson's disease (PD), although the same menu of
antiparkinson drugs is used to treat all of the
various parkinsonian syndromes. The four cardinal
signs of parkinsonism are rest tremor, rigidity,
akinesia, and gait disturbance. Usual criteria for a
clinical diagnosis of PD require the presence of at
least two of these four features; diagnostic
certainty increases in proportion to the
predominance of rest tremor as a finding, especially
if it is unilateral.
Postmortem and magnetic resonance imaging (MRI)
studies indicate a substantial diagnostic error rate
based upon the use of these criteria. In some
studies, up to 25 percent of patients with a
diagnosis of PD during life were found to have other
causes of parkinsonian symptoms at autopsy, such as
cerebrovascular disease involving the basal ganglia
or other neurodegenerative disorders, such as
multiple system atrophy and progressive supranuclear
palsy. On the other hand, the clinical diagnostic
accuracy of PD and the other parkinsonian syndromes,
confirmed by neuropathology, increases substantially
if patients are evaluated and observed throughout
the long course of their illness by specialists at
movement disorders centers.
The clinical features most suggestive of idiopathic
PD include asymmetric or unilateral onset, the
presence of rest tremor, and a clear cut response to
treatment with levodopa. Clues to the diagnosis of
the other parkinsonian syndromes include a history
of exposure to dopamine receptor blocking drugs such
as antipsychotic agents or metoclopramide,
hyperactive tendon reflexes, Babinski signs,
cerebellar signs, prominent abnormalities of ocular
motility, the early development of dementia,
significant postural instability, and major
autonomic manifestations (orthostatic hypotension,
atonic bladder, sexual impotence, and
gastrointestinal dysmotility).
A practice parameter from the American Academy of
Neurology (AAN) concluded that features probably
useful for distinguishing other parkinsonian
syndromes from PD include early falls, poor response
to levodopa, symmetry of motor manifestations, lack
of tremor, and early autonomic dysfunction.
NEUROPROTECTIVE THERAPY The pharmacologic
treatment of Parkinson's disease (PD) can be divided
into neuroprotective and symptomatic therapy. In
practice, however, nearly all of the available
treatments are symptomatic in nature and do not
appear to slow or reverse the natural course of the
disease.
Neuroprotective therapy of PD is still theoretical,
but it is based on the concept that the three to
four hundred thousand at-risk dopaminergic neurons
in the human substantia nigra can somehow be
protected from the complex degenerative process that
causes premature cell death and depletion of
dopamine. Once identified and shown to be effective,
neuroprotective drugs could be used in patients with
early clinical signs of disease or potentially even
prior to the appearance of disease in those shown to
be at genetic risk.
Several potential neuroprotective agents for PD have
shown some promise in animals and/or humans and are
undergoing further investigation. Selegiline and
rasagiline (both monoamine oxidase inhibitors),
dopamine agonists, and the complex I mitochondrial
fortifier coenzyme Q10 have been evaluated in
clinical trials and are receiving the most attention
as possible neuroprotective agents.
MAO B INHIBITORS Monoamine oxidase (MAO B)
inhibitors such as selegiline and rasagiline have
been studied as neuroprotective agents due to their
ability to block free radical formation from the
oxidative metabolism of dopamine; these agents may
also inhibit apoptosis (programmed cell death). In
addition to a possible neuroprotective effect,
selegiline has a mild symptomatic benefit.
Selegiline The possibility of long-term
neuroprotection with selegiline has neither been
confirmed nor disproven: A large prospective,
double-blind, placebo-controlled multicenter study
(the DATATOP study) found that selegiline (deprenyl)
10 mg daily delayed the progression of parkinsonian
signs in previously untreated patients by nine
months. However, a small but measurable reduction of
parkinsonian symptoms attributable to selegiline
confounded the findings of this study, thereby
casting doubt on the likelihood that the delayed
progression of symptoms was due to a true
neuroprotective effect. A subsequent trial that took
this symptomatic effect into account noted a mild
neuroprotective effect. Selegiline treatment of
patients with early Parkinson's disease (PD) in the
absence of levodopa was associated with a decreased
risk for developing later freezing of gait,
suggesting a possible neuroprotective effect.
Prior treatment with selegiline in the DATATOP
cohort did not reduce the occurrence of subsequent
levodopa-associated motor fluctuations in this
population; no persistent, long-term benefit in
slowing the progression of PD was demonstrated with
selegiline.
This lack of long-term benefit in part accounted for
the conclusion issued in a 2002 report of the
Quality Standards Subcommittee of the American
Academy of Neurology (AAN) that there is
insufficient evidence to recommend the use of
selegiline for a neuroprotective effect. A practice
parameter from the AAN issued in 2006 found no
interim studies that would alter this conclusion.
Rasagiline The selective MAO B inhibitor
rasagiline has neuroprotective properties in animal
models. Results from a short term randomized
controlled trial of rasagiline monotherapy versus
placebo, using a delayed start design, suggested
that it might slow progression of parkinsonian
disability; patients treated with rasagiline had a
smaller increase in mean adjusted total Unified
Parkinson's Disease Rating Scale (UPDRS) score
compared with those who were treated with placebo.
However, the symptomatic benefit of rasagiline,
rather than a neuroprotective effect, may have been
responsible for this result. Further trials with a
greater number of patients are planned to test the
hypothesis that rasagiline slows the progression of
PD. The AAN practice parameter concluded that there
is insufficient evidence to support or refute the
use rasagiline for neuroprotection in patients with
PD.
DOPAMINE AGONISTS Dopamine agonists are
neuroprotective in the laboratory because they are
antioxidants and free radical scavengers and because
of feedback reduction of endogenous dopamine
turnover. These findings have led to the hypothesis
that the use of agonists early in the clinical
course of PD may slow progression of the underlying
neurodegeneration. Later studies employing
radiographic markers of basal ganglia function,
although highly controversial, have provided
further, tentative support for a possible
neuroprotective effect of dopamine agonists.
One potential biologic marker uses single photon
emission computed tomography (SPECT) with the
dopamine transporter ligand [123I]beta-CIT (B-CIT)
as an anatomic measure of nigrostriatal integrity
and as a surrogate marker of PD progression. Earlier
investigations of the natural history of PD had
shown a 5 to 10 percent reduction of B-CIT uptake
per year in a heterogeneous mix of patients with PD.
Pramipexole The CALM-PD study evaluated 82
patients with early PD, using SPECT B-CIT scans as a
surrogate marker of neuroprotection, and found that
the patients who were randomly assigned to receive
pramipexole (0.5 mg three times per day)
demonstrated less of a decline in striatal B-CIT
uptake over four years compared with those treated
with carbidopa/levodopa (25/100 mg three times per
day). There was no difference between the two
treatment groups for the change in the UPDRS scores
from baseline.
Ropinirole A randomized trial studied 162
patients eligible for analysis who were assigned to
ropinirole or levodopa treatment, using positron
emission tomography (PET) scanning and the dopa
decarboxylase ligand 18F-fluorodopa ((18)F-dopa) as
a measure of nigrostriatal integrity. There was
significantly less decline in (18)F-dopa uptake in
patients assigned to ropinirole compared with those
assigned to levodopa. Another randomized trial
employing (18)F-dopa PET as a surrogate marker of
neuroprotection studied 45 patients who were
assigned to ropinirole or levodopa. At two years,
the ropinirole treatment group showed a smaller
reduction in the primary endpoint of putaminal
(18)F-dopa uptake compared with placebo, but the
difference was not statistically significant (13
versus 18 percent, respectively).
Interpretation It is uncertain whether these
imaging studies reflect changes in the underlying
pathology of PD or differential pharmacologic
"regulatory" changes directly attributable to the
drugs themselves. Therefore, these findings raise
the possibility that dopamine agonists may be
neuroprotective, but confirmation is required in
additional clinical studies, including prospective
data in untreated patients.
The AAN practice parameter noted that significance
of the studies evaluating pramipexole and ropinirole
is uncertain, given the lack of validation for the
surrogate measures of neuroprotection employed (i.e,
SPECT B-CIT and (18)F-dopa PET scans) and the
absence of placebo control groups. The AAN statement
concluded that there is insufficient evidence to
support or refute the use of pramipexole or
ropinirole for neuroprotection in patients with PD.
LEVODOPA Accumulating clinical trial data
suggest that levodopa either slows the progression
of PD or has a prolonged benefit even after the drug
has been stopped. These data are presented
separately. The AAN practice parameter concluded
that levodopa is possibly neuroprotective for at
least nine months and does not accelerate disease
progression.
OTHER AGENTS
Coenzyme Q10 Interest in coenzyme Q10 has
been stimulated by evidence that mitochondrial
dysfunction may play a role in the pathogenesis of
PD. In a small clinical trial, 80 subjects with
early untreated PD were randomly assigned to three
dosage groups of coenzyme Q10 or to placebo, and
were followed for progression of disease as measured
by the Unified Parkinson's Disease Rating Scale
(UPDRS). Treatment with coenzyme Q10 at the highest
dosage (1200 mg daily) was associated with a lower
rate of disability progression over 16 months
compared with placebo. Although the results did not
achieve statistical significance, they did meet the
prespecified criteria for a positive trend for the
trial. However, the study was underpowered to detect
a neuroprotective effect. As with selegiline, it is
not entirely clear whether the benefit of coenzyme
Q10 was due to neuroprotection or to symptomatic
improvement; the study investigators considered the
symptomatic effect of coenzyme Q10 to be negligible.
These promising results regarding coenzyme Q10
require confirmation in clinical trials with larger
samples of patients. A practice parameter from the
AAN concluded that there is insufficient evidence to
support or refute the use of coenzyme Q10 for
neuroprotection in patients with PD.
Vitamin E In the randomized controlled
DATATOP trial of patients with early PD, Vitamin E
(tocopherol) was included as a treatment arm. There
was no beneficial effect of vitamin E compared with
placebo for the primary end point of average time to
onset of disability requiring levodopa use. Given
these data, the AAN practice parameter concluded
that vitamin E should not be considered for
neuroprotection [10].
Riluzole A randomized controlled trial of
patients with early PD found no beneficial effect of
riluzole compared with placebo as measured by change
in the UPDRS. However, this study was not
sufficiently powered to exclude a modest
neuroprotective effect of riluzole.
SUMMARY AND CONCLUSIONS Neuroprotective
therapy of Parkinson's disease (PD) is still
theoretical, but it is based on the concept that
dopaminergic neurons in the substantia nigra can be
protected from the degenerative process that causes
premature cell death and depletion of dopamine,
leading to the development of PD. Monoamine oxidase
(MAO B) inhibitors such as selegiline and rasagiline
have been studied as neuroprotective agents due to
their ability to block free radical formation from
the oxidative metabolism of dopamine; these agents
may also inhibit apoptosis. Dopamine agonists are
neuroprotective in the laboratory because they are
antioxidants and free radical scavengers and because
of feedback reduction of endogenous dopamine
turnover. Interest in coenzyme Q10 has been
stimulated by evidence that mitochondrial
dysfunction may play a role in the pathogenesis of
PD. No treatment for PD has been proven to be
neuroprotective. However, levodopa is possibly
neuroprotective and does not accelerate disease
progression. Existing clinical trial evidence in
patients with PD is insufficient to support or
refute the possibility of neuroprotection for
selegiline, rasagiline, pramipexole, ropinirole,
coenzyme Q10, and riluzole. There is no evidence
that Vitamin E is neuroprotective.
SYMPTOMATIC THERAPY The decision to
initiate symptomatic medical therapy in patients
with Parkinson's disease (PD) is determined by the
degree to which the patient is functionally
impaired. The timing of this decision varies greatly
among patients but is influenced by a number of
factors, including [1]: The effect of disease on the
dominant hand The degree to which the disease
interferes with work, activities of daily living, or
social and leisure function The presence of
significant bradykinesia or gait disturbance
Personal philosophy regarding the use of drugs
The major drugs available for symptomatic therapy
include: Levodopa MAO B inhibitors Dopamine agonists
COMT inhibitors Anticholinergic agents Amantadine.
In addition to these agents, low-dose estrogen may
be helpful as adjunctive therapy in postmenopausal
women.
Levodopa
Levodopa (L-dopa) is well established as the most
effective drug for the symptomatic treatment of
idiopathic or Lewy body PD. It is particularly
effective for the management of akinetic symptoms
and should be introduced when these become disabling
and are uncontrolled by other antiparkinsonian
drugs. Tremor and rigidity can also respond to
levodopa therapy, but postural instability is less
likely to do so.
Levodopa is combined with a peripheral decarboxylase
inhibitor to block its conversion to dopamine in the
systemic circulation and liver (before it crosses
the blood-brain barrier) in order to prevent nausea,
vomiting, and orthostatic hypotension. The
decarboxylase inhibitor is carbidopa. The
combination drug carbidopa/levodopa
(immediate-release Sinemet) is available in tablets
of 10/100, 25/100, and 25/250 mg, with the numerator
referring to carbidopa and the denominator referring
to the levodopa dose. An immediate-release
formulation of carbidopa/levodopa (Parcopa) is
available that dissolves on the tongue and can be
taken without water, but there are no published
studies of this formulation, and its onset of action
is no different from Sinemet.
In Europe and Canada, benserazide is the peripheral
decarboxylase inhibitor. The combination drug
benserazide/levodopa (Madopar or Prolopa) is
available in 25/100 and 50/200 mg tablets.
Controlled-release formulations of
carbidopa/levodopa and benserazide/levodopa are
available as Sinemet CR and Madopar HBS,
respectively.
Dosing Treatment should be initiated with
small doses such as one-half tablet of
carbidopa/levodopa (Sinemet) 25/100 mg three times
daily with meals, titrated upward over several weeks
to 25/100 mg three times daily as tolerated and
according to the response. See table 1. Elderly
patients or those with dementia should begin with
smaller doses and titrate more slowly because of
their increased susceptibility to psychiatric side
effects. The usual practice is to use the lowest
dose that produces a useful clinical response. This
varies from patient to patient, but at the start
typically is in the vicinity of 300 to 600 mg of
levodopa daily.
Table-1. Drug
treatment of Parkinson's disease |
Generic name |
Trade name |
Usual
starting dose |
Usual
maintenance dose |
Mechanism |
Trihexyphenidyl |
Artane |
1 mg BID |
2 mg BID-TID |
Anticholinergic |
Benztropine |
Cogentin |
0.5 mg BID |
1 to 2 mg
BID-TID |
Anticholinergic |
Amantadine |
Symmetrel |
100 mg BID |
100 mg BID-TID |
? |
Selegiline |
Eldepryl |
5 mg |
5 mg qam |
MAO B inhibitor |
Carbidopa/levodopa |
Sinemet |
25/100 mg TID |
25/250 mg
TID-QID |
Dopamine
precursor |
Carbidopa/levodopa |
Sinemet CR |
25/100 mg TID |
50/200 mg TID |
Dopamine
precursor |
Apomorphine |
Apokyn |
2 mg SC test
dose |
2 to 10 mg SC
TID |
Dopamine
agonist |
Bromocriptine |
Parlodel |
2.5 mg daily |
5 to 10 mg QID |
Dopamine
agonist |
Pergolide |
Permax |
0.05 mg daily |
0.5 to 1.0 mg
TID |
Dopamine
agonist |
Pramipexole |
Mirapex |
0.125 mg TID |
1.5 mg TID |
Dopamine
agonist |
Ropinirole |
Requip |
0.25 mg TID |
1.0 mg TID |
Dopamine
agonist |
Entacapone |
Comtan |
200 mg with
L-dopa |
600 to 800 mg a
day |
COMT inhibitor |
Tolcapone |
Tasmar |
100 mg TID |
100 to 200 mg
TID |
COMT inhibitor |
The vast majority of patients
with idiopathic PD will enjoy a significant
therapeutic response to moderate doses of levodopa
(400 to 600 mg daily); complete absence of response
to a dose of 1000 to 1500 mg/day strongly suggests
that the original diagnosis of PD was incorrect and
that the diagnosis should be revised to one of the
other parkinsonian syndromes, such as multiple
system atrophy or progressive supranuclear palsy.
Controlled release levodopa preparations are less
completely absorbed and require a dose up to 30
percent higher to achieve an equivalent clinical
effect. The clinical effect of each tablet is
typically less dramatic than for immediate release
preparations, since controlled release formulations
reach the brain more slowly. This presents a
disadvantage in assessing the response of patients
just beginning therapy. As a result, it is
recommended that therapy be initiated with an
immediate release preparation with a subsequent
switch to controlled release if desired. Both the
immediate and the controlled release formulations
appear to maintain a similar level of symptom
control after several years of use.
Patients taking levodopa for the first time should
take each dose with a meal or snack to avoid nausea,
a common early side effect. Patients with more
advanced disease, especially those with motor
fluctuations, often notice that a dose of levodopa
is more effective if taken on an empty stomach one
hour before or after meals due to reduced
competition with other amino acids for
gastrointestinal absorption.
Small starting doses of levodopa combined with a
decarboxylase inhibitor (eg, Sinemet, Madopar, or
Prolopa) are more likely to cause nausea because of
inadequate amounts of carbidopa; this can be managed
by administering supplemental doses of carbidopa
(Lodosyn) or by use of antiemetics such as
trimethobenzamide (Tigan) or domperidone taken prior
to Sinemet. Phenothiazine antiemetics and
metoclopramide should be avoided because they are
dopamine receptor blockers that can aggravate the
parkinsonian symptoms.
Adverse effects Nausea, somnolence,
dizziness, and headache are among the more common
side effects that may accompany treatment with
levodopa, but they are not likely to be serious in
most patients. More serious adverse reactions to
levodopa (mainly in older patients) may include
confusion, hallucinations, delusions, agitation, and
psychosis. Levodopa may also induce a mild to
moderate elevation in serum
homocysteine levels, which in turn may be
associated with an increased
risk of hip fractures in elderly patients.
Compulsive dopaminergic drug use has been reported
in patients taking dopamine agonists, typically in
conjunction with levodopa therapy. However, it is
unclear if these behavioral issues arise with
levodopa monotherapy. The "higher doses of
dopaminergic medications than necessary to
effectively control parkinsonian symptoms should be
avoided," but noted that some patients with worse
(ie, more rapidly progressive) disease may require
high doses of dopaminergic medications to manage
symptoms and therefore may be at greater risk for
the development of motor complications. However,
motor complications do not necessarily cause
clinically significant functional impairment and can
potentially be managed.
Given these data, practitioners should always try to
find the lowest but still effective dose of
dopaminergic medication, either singly or in
combination, for patients with PD, each of whom must
be evaluated and managed according to his or her
individual needs. The increase in motor fluctuations
over time is most likely due to the progressive
degeneration of nigrostriatal dopamine terminals,
which increasingly limits the normal physiologic
uptake and release of dopamine, thereby leading to
reduced buffering of the natural fluctuations in
plasma levodopa levels that occur due to levodopa's
90-minute pharmacologic half-life. Controlled
release preparations are useful for management of
these fluctuations, although, in one report, the use
of Sinemet CR from the start of therapy, in an
effort to provide more continuous stimulation of
dopamine receptors, was not associated with fewer
motor complications than immediate release Sinemet.
There has been longstanding concern among some
clinicians that levodopa causes motor fluctuations
and dyskinesia by its potential to promote oxidative
stress and accelerated neurodegeneration, rather
than by the change in levodopa pharmacodynamics that
occurs with natural progression of the underlying
disease. Therefore, it is commonly proposed that the
initiation of levodopa be delayed until symptoms
significantly interfere with function. Others
contend, however, that there is no strong evidence
that levodopa is responsible for late motor
complications, and that delay of treatment
unnecessarily deprives patients of therapeutic
benefit early in the disease, when the potential for
sustained improvement is greatest. A sudden
exacerbation of Parkinson's disease (PD)
characterized by an akinetic state that lasts for
several days and does not respond to treatment with
antiparkinson medication is called acute akinesia.
This phenomenon is very different from the more
common wearing "off" effects.
Motor fluctuations A substantial number of
patients develop levodopa-induced complications
within several years of starting levodopa. These
include motor fluctuations (the wearing-off
phenomenon), involuntary movements known as
dyskinesia, abnormal postures of the extremities and
trunk known as dystonia, and a variety of complex
fluctuations in motor function. As many as 50
percent of patients on levodopa for five years
experience motor fluctuations (MF) and dyskinesia.
These symptoms are especially common in patients
with young-onset (eg, under the age of 50)
Parkinson's disease (PD); they are unique to
levodopa and are not produced by the other
antiparkinson drugs. Such motor complications occur
in at least 50 percent of patients after 5 to 10
years of treatment. In the large group of patients
with early PD studied in the DATATOP study, motor
complications occurred in 30 percent after only two
years of treatment with levodopa. However, in a
study of early PD, the prevalence of motor
complications was only 20 percent after five years
of treatment with levodopa.
Retrospective data from a study of the effect of
pramipexole and levodopa on early PD (the CALM-PD
study) suggest that the earlier occurrence of motor
fluctuations in the course of PD is associated with
higher cumulative levodopa doses and higher
cumulative levodopa-equivalent doses (ie, levodopa
plus the dopamine agonist pramipexole). In contrast,
prospective data from the same study suggest that a
later onset of motor fluctuations in PD is
associated with initial treatment with pramipexole
rather than levodopa.
Patients typically experience a smooth and even
response to the early stages of levodopa treatment.
As the disease advances, however, the effect of
levodopa begins to wear off approximately four hours
after each dose, leaving patients anticipating the
need for their next dose. This phenomenon may be
explained by the observation that dopamine nerve
terminals are able to store and release dopamine
early in the course of disease but, with more
advanced disease and increasing degeneration of
dopamine terminals, the concentration of dopamine in
the basal ganglia is much more dependent upon plasma
levodopa levels. Plasma levels may fluctuate
erratically because of the 90 minute half-life of
levodopa and the frequently unpredictable intestinal
absorption of this medication.
Motor fluctuations (MF) are alterations between
periods of being "on," during which the patient
enjoys a good response to medication, and being
"off" during which the patient experiences symptoms
of their underlying parkinsonism.
Dyskinesia consists of abnormal involuntary
movements that are usually choreic or dystonic but,
when more severe, may be ballistic or myoclonic.
Dyskinesia usually appears when the patient is "on."
It may occasionally occur in the form of painful
dystonia when the patient is "off," especially in
the morning on awakening, when dystonic intorsion of
a foot (usually on the side of greater parkinsonian
involvement) occurs as a withdrawal reaction because
of the long interval without medication overnight.
Surgery for advanced PD is
another therapeutic option, as bilateral deep brain
stimulation of the subthalamic nucleus or globus
pallidus appears to improve motor function in
selected patients with advanced typical PD and MF,
whose condition cannot be further improved by
medical therapy.
WEARING-OFF PHENOMENON Patients with
advanced Parkinson's disease (PD) begin to be aware
of a wearing "off" or end-of-dose effect less than
four hours following a dose of levodopa.
Alteration of levodopa dosing Wearing "off" can
initially be managed by increasing the dose of
levodopa, if the patient is not having side effects
and is taking a relatively low dose. However,
increasing the dose often increases side effects
without effectively increasing the dose duration.
Shortening the interdose interval while
administering lower doses is usually a more
effective approach. However, it is often difficult
to titrate the dose precisely, and some patients
begin to exhibit an "all or none" response whereby
individual lower doses produce no evident clinical
response. This occurs because the pharmacologic
response threshold is higher in advanced disease
than it is in earlier disease.
Liquid Sinemet (carbidopa/levodopa) is occasionally
used for patients when titration of the dose and
dose interval using tablets is difficult. However,
this approach is not typically practical since
Sinemet is insoluble in water and no commercial
preparation of liquid Sinemet is available.
Instructions for preparation of a daily supply of
liquid Sinemet are available, but use of this
approach is best left to the specialist.
The sustained-release forms of levodopa preparations
(eg, Sinemet CR) may be useful in the early stages
of the wearing "off" phenomenon and may add up to 90
additional minutes throughout the day to levodopa's
duration of effect. However, Sinemet CR is less well
absorbed than immediate release Sinemet; thus, an
individual dose increase of approximately 30 percent
may be required to achieve the same clinical
response. Sustained-release carbidopa/levodopa does
not decrease "off" time compared with immediate
release formulations.
Addition of a second drug Addition of a second
drug is indicated if the adjustments cited above are
not successful (table 1).
Dopamine agonists Dopamine agonists are commonly
used to reduce the amount of "off" time in patients
with advanced PD and may also allow for the dose of
levodopa to be reduced. The drugs currently approved
by the United States Food and Drug Administration
(FDA) include bromocriptine (Parlodel), pergolide
(Permax), pramipexole (Mirapex), ropinirole
(Requip), and apomorphine (Apokyn). Cabergoline is
approved by the FDA only for the treatment of
hyperprolactinemic disorders, and its use for
advanced PD is off label. Studies comparing the
efficacy of various dopamine agonists have found
either no significant difference or only mild
superiority of one agent over another.
The dopamine agonist apomorphine administered
subcutaneously can be used for rapid onset (usually
within 10 minutes) rescue therapy when patients
suddenly turn "off". In a randomized, double-blind,
placebo-controlled study of 29 patients with
advanced PD and two hours or more of "off" time
despite aggressive oral therapy, administration of
subcutaneous apomorphine (2 to 10 mg) resulted in
successful amelioration of "off" state events
following 95 percent of injections compared with 23
percent receiving placebo injection.
One review concluded that the magnitude and pattern
of the motor response to a single subcutaneous dose
of apomorphine is qualitatively comparable to that
of oral levodopa; a 4 mg dose achieved a clinically
significant improvement in 75 percent of patients.
Cabergoline may have
some utility for reduction of "off" time in patients
with advanced PD, but data are limited. In a single
center, 24-week study of 37 patients (19 active, 18
placebo), treatment with cabergoline (mean dose 5.4
mg/day) was associated with a significant decrease
in daily "off" time compared with placebo (2 versus
0.7 hours [40 versus 18 percent]). However, these
results are limited by a potentially confounding
baseline difference in "off" time duration between
the treatment groups. In another single center,
24-week study of 27 patients (17 active, 10
placebo), patients treated with cabergoline (mean
dose 4.9 mg/day) had an increase in "on" time (2.7
hours [30 percent]) and a decrease in "off" time
(3.3 hours [59 percent]), but no information was
provided for the placebo group about these
parameters.
Cabergoline treatment was not associated with
increased dyskinesia in these trials. However, a
retrospective case control study of 210 patients
with PD found that high cumulative dose and
long-term treatment with cabergoline was associated
with an increased risk of cardiac valvulopathy
detected on transthoracic echocardiography.
In a randomized controlled trial,
bromocriptine decreased
"off" time compared with placebo (8 versus 3
percent, respectively), but the difference was not
statistically significant.
COMT inhibitors Catechol-O-methyl transferase
(COMT) inhibitors such as tolcapone (Tasmar)
and entacapone (Comtan)
may prolong and potentiate the levodopa effect and
reduce the "off" time when given with a dose of
levodopa. The net result is an increased levodopa
effect in fluctuating patients. These medications
may allow a reduction in the total daily levodopa
dose by as much as 30 percent. The starting dose of
tolcapone is 100 mg three times daily; the clinical
effect is evident immediately. The dose of
entacapone is one 200 mg tablet with each dose of
levodopa, up to a maximum of eight doses per day.
The most common side effects of these drugs are due
to increased dopaminergic stimulation and include
dyskinesia, psychiatric effects (mainly visual
hallucinations), nausea, diarrhea, and orthostatic
hypotension. The adverse effects are managed by
lowering the dose of levodopa either before or after
the addition of tolcapone or entacapone. Both drugs
may also cause a brown-orange urine discoloration.
In clinical trials, tolcapone was associated with
transient, asymptomatic elevations of transaminases
(AST and ALT) in 1 to 3 percent of subjects exposed
to the drug. Three reported deaths from
hepatotoxicity in patients using tolcapone prompted
its removal from the market in Canada and Europe,
although it is still available in the United States
with the recommendation that it be used for
treatment of motor fluctuations only after other
methods have been exhausted and with monitoring of
ALT and AST levels for the first six months of
therapy. Entacapone has thus far not been associated
with hepatotoxicity. Monitoring of liver enzymes
with liver function tests (LFTs) must be done at
baseline and then every two weeks for the first year
of tolcapone therapy, then every four weeks for the
next six months, then every eight weeks thereafter.
Monitoring of LFTs should be resumed at the previous
frequency if the tolcapone dose is increased to 200
mg three times a day. Tolcapone should be
discontinued if the ALT or AST exceeds the upper
limit of normal or if the clinical signs and
symptoms suggest the onset of liver failure.
MAO B inhibitors Rasagiline is a selective
monoamine oxidase (MAO) B inhibitor. It has
potential long-term effects on dopamine transmission
because it acts irreversibly on MAO B receptors.
Rasagiline appears to
be effective for motor complications in PD as
demonstrated in randomized clinical trials. One of
these, the 18-week multicenter LARGO trial,
evaluated 687 patients with PD who had motor
fluctuations (MF) for at least one hour every day
despite optimum levodopa/dopa decarboxylase therapy.
Patients were randomly assigned to adjunct therapy
with either rasagiline 1 mg daily, entacapone 200 mg
with every levodopa dose, or placebo. Both
rasagiline and entacapone reduced mean daily "off"
time (the primary outcome measure) by about one hour
compared with placebo, and both increased daily "on"
time without troublesome dyskinesia compared with
placebo. The beneficial effect of rasagiline was
independent of age (<70 versus 70 years) and of
adjunct use of dopamine agonists.
Rasagiline was well tolerated in these studies. The
frequency of dopaminergic adverse events in the
LARGO trial was similar to that seen in the
entacapone and placebo groups.
Rasagiline is approved by the European Commission as
initial monotherapy in patients with early PD and as
adjunct treatment in moderate to advanced PD. It
received similar approval by the United States Food
and Drug Administration in May 2006.
Selegiline is another
selective MAO B inhibitor. Unlike rasagiline,
selegiline is metabolized to amphetamine
derivatives. Although selegiline may extend the
levodopa effect, the clinical benefit this produces
is usually relatively mild. Results from a small
randomized controlled trial suggest that orally
disintegrating selegiline may also be beneficial,
although the study did not report change in levodopa
dose.
Other strategies Anticholinergic drugs and
amantadine are not very
effective in managing the wearing "off" effect and
are rarely indicated for this purpose, given the
other more effective options. Early studies of
adenosine A2A antagonists as adjunctive therapy in
PD have yielded promising results, and clinical
trials are ongoing.
Preliminary studies suggest that eradication of
Helicobacter colonization, which is present in about
half of the population, may be a useful method for
improving levodopa absorption and reducing motor
fluctuations in patients with PD. These require
confirmation in larger clinical trials before
routine testing for H. pylori and antibiotic
eradication can be recommended.
Guideline recommendations for treating "off" time
An evidenced-based practice parameter from the AAN
issued in 2006 made the following recommendations
for the treatment of "off" time in patients with PD
and motor fluctuations: Entacapone and rasagiline
are established as effective and should be offered
to reduce "off" time Pergolide, pramipexole,
ropinirole, and tolcapone are probably effective and
should be considered to reduce "off" time, with the
stipulation that the adverse effects of tolcapone
(hepatotoxicity) and pergolide (valvular fibrosis)
require that they be used with caution and
monitoring Apomorphine, cabergoline, and selegiline
are possibly effective and may be considered to
reduce "off" time Sustained release
carbidopa/levodopa does not decrease "off" time
compared with immediate release carbidopa/levodopa;
bromocriptine does not reduce "off" time compared
with placebo; both may be disregarded to reduce
"off" time.
UNPREDICTABLE OFF PERIODS Transitions from
being "on" to being "off" can be sudden and
unpredictable in some patients. Unlike the wearing
"off" phenomenon at the end of a levodopa dose
cycle, there is sometimes no obvious relationship
between the time of levodopa administration and the
appearance of "off" episodes in patients with
unpredictable "off" periods. These periods typically
occur in patients with advanced Parkinson's disease
(PD) who are also experiencing motor fluctuations
(MF) and severe dyskinesia.
Management of these individuals is similar to that
for patients who are having problems with wearing
"off," although it typically is much more difficult.
Direct observation of the patient during a prolonged
outpatient visit as he or she cycles through such
episodes is useful to determine the relationship of
levodopa doses to "off" episodes. In some cases,
these episodes occur at times of peak levodopa
effect due to excessive rather than insufficient
dopaminergic stimulation; such patients are best
treated by reducing rather than raising the levodopa
dose.
Addition of a COMT inhibitor or a dopamine agonist
can be helpful; marked reduction of the levodopa
dose together with the addition of high doses of a
dopamine agonist may be required. Controlled release
levodopa preparations (eg, Sinemet CR) are usually
not helpful and occasionally exacerbate the
situation.
Competition with neutral amino acids for transport
across the gut and into the brain may be responsible
for "offs" that appear following meals. A protein
redistribution diet in which most protein intake is
reserved for the evening was useful in approximately
two-thirds of such patients in small studies,
although this type of diet tends to be impractical
for long-term use.
Episodic freezing is a special form of unpredictable
"off" in which patients suddenly become immobilized
for seconds to minutes at a time. This complication
usually occurs while walking when it may cause
falls; it is often not medication related and is
very resistant to treatment. When freezing is more
prolonged and accompanied by the emergence of other
parkinsonian signs, treatment is similar to patients
with other forms of the wearing "off" effect.
ACUTE AKINESIA Acute akinesia is a sudden
exacerbation of Parkinson's disease (PD)
characterized by an akinetic state that lasts for
several days and does not respond to treatment with
antiparkinson medication. This phenomenon is
different from wearing "off" effects and may occur
in patients not previously treated with levodopa.
Acute akinesia should prompt a search for systemic
infection or other intercurrent medical problems
that are capable of causing a sudden worsening of
parkinsonism. In a review of this problem in 26
patients, acute akinesia appeared after a flu-like
syndrome in six patients, hip joint surgery or bone
fractures in eight patients, gastrointestinal
disturbances in three patients, and various
medication manipulations in the remaining patients.
Four patients died in spite of treatment. Episodes
of acute akinesia may therefore have serious
consequences and usually warrant acute
hospitalization in order to identify and correct the
underlying cause.
FAILURE OF ON-RESPONSE Patients with motor
fluctuations (MF) sometimes fail to turn "on"
following a dose of levodopa. This has been called
the "no-on" response. In some cases, this is due to
delayed gastric motility, which results in
inadequate plasma concentrations in advanced
patients who have a narrow therapeutic window. A
common reason for the "no-on" phenomenon is an
excessively prolonged or severe "off" period
occurring before the "no-on." This is best managed
by avoiding "offs."
The prokinetic agent cisapride increases
gastrointestinal motility and may be helpful in such
patients, but the drug has been associated with a
number of drug interactions and fatal cardiac
arrhythmias, prompting the manufacturer to severely
restrict its availability in the United States. The
prokinetic drug metoclopramide is a dopamine
receptor blocker that should be avoided. Patients
should be encouraged to take levodopa on an empty
stomach and avoid protein at the time of drug
administration.
Domperidone is a
D2-blocker with selective peripheral activity in the
upper gastrointestinal tract; it does not cross the
blood-brain barrier and therefore lacks the
neurologic side effects of metoclopramide. It is
currently not available in the United States but is
available in Canada and other countries. Although
data are limited, domperidone (starting at 20 mg
four times daily) may be useful as a prokinetic
agent to treat delayed gastric emptying in patients
with PD. However, animal studies suggest that, like
cisapride, domperidone may increase the risk of
cardiac arrhythmias.
DYSKINESIA Dyskinesia refers to a variety
of involuntary movements, which occur as a direct
effect of levodopa. Other antiparkinson drugs are
much less likely to produce these motor
abnormalities but may exacerbate them once they have
appeared following treatment with levodopa.
Dyskinesia is sometimes mistaken for manifestations
of progressive Parkinson's disease (PD) or confused
with tremor by patients and their families, rather
than recognized as reversible consequences of
treatment. Dyskinesia occurs in 30 to 40 percent of
patients treated with levodopa by five years and
nearly 60 percent by ten years, but not all
dyskinesia requires treatment. A retrospective study
suggests that the rate of dyskinesia requiring
medication adjustment at five and ten years after
levodopa treatment is 17 and 43 percent,
respectively. Dyskinesia is usually choreiform in
type, manifested by continuous, restless appearing
movements of the extremities, head, face, trunk, and
respiratory muscles. These dyskinetic movements are
remarkably well tolerated by most patients since
patients feel entirely relieved of their
parkinsonism while dyskinesia is present. However,
severe dyskinesia may take the form of large
amplitude, ballistic movements that interfere with
function and become very disturbing to patients and
their families.
Levodopa was given in relatively high doses when it
was first used as therapy for PD. As a result,
dyskinesia was often seen early in treatment,
especially in those with advanced disease who were
being treated for the first time. The subsequent use
of more modest doses resulted in its later
appearance, months to years after initiating
levodopa. Dyskinesia is especially common in
patients with young-onset PD.
Peak-dose dyskinesia is most common. It occurs 60 to
90 minutes following a dose of levodopa. Early in
the disease, this complication can be managed by
lowering the medication dose, switching to a
controlled release preparation, or reducing
adjunctive drugs such as dopamine agonists,
selegiline, or anticholinergic drugs. However, in
more advanced patients with brittle responses,
reducing the dose of levodopa may result in complete
failure to generate an "on" response. In this
situation, the dose of dopamine agonist should be
greatly increased and the levodopa dose reduced,
since dopamine agonists are much less likely to
induce dyskinesia than levodopa.
An unusual pattern sometimes evolves in which
dyskinesia peaks twice after each dose (diphasic
dyskinesia) - when patients turn "on" and again as
they begin to turn "off". In the second phase,
dyskinesia in one body part may coexist with the
emergence elsewhere of parkinsonian signs such as
tremor and dyskinesia. This pattern is often
unrecognized and may only be appreciated if the
patient is observed during a prolonged outpatient
visit.
The diphasic pattern is notoriously difficult to
manage and usually requires more frequent levodopa
dosing to prevent wearing "off" prior to each dose.
However, this strategy often leads to progressively
increasing dyskinesia as the day goes on. Addition
of a dopamine agonist and a marked reduction in the
levodopa dose should be tried in such patients.
Sustained release levodopa is best avoided in
patients with severe or complex patterns of
dyskinesia since absorption may be delayed and
dyskinesia tends to progressively increase into the
afternoon and evening.
Amantadine Amantadine
may be useful for treating dyskinesia in advanced
PD. Several studies have shown short-term benefit.
As an example, a single-center randomized controlled
trial found that amantadine administration compared
with placebo was associated with a 24 percent
reduction in the total dyskinesia score. In
addition, a placebo-controlled study involving 17
patients showed that the beneficial effects of
amantadine on motor response fluctuations were
maintained for at least one year; initial and
one-year reductions in dyskinesia scores were 60 and
56 percent, respectively.
In another placebo-controlled study in 40 patients
with dyskinesia, amantadine treatment for 15 days
resulted in a 45 percent reduction in dyskinesia
scores compared with placebo. However, the benefit
in this study lasted less than eight months, and
amantadine withdrawal resulted in a rebound with
increase of dyskinesia in 11 patients. Amantadine
was not associated with worsening of parkinsonism
symptoms in these studies. The dose of amantadine
for dyskinesia is one tablet (100 mg) one to three
times a day. Side effects may include peripheral
edema, psychosis, and livedo reticularis.
Clozapine Low doses
of the antipsychotic clozapine (30 to 50 mg/day)
reduced dyskinesia in several open-label studies,
and low dose clozapine (12.5 to 75 mg/day) was
significantly more effective than placebo in
treating levodopa-induced dyskinesia in a
double-blind, randomized controlled trial of 50
patients. The usefulness of clozapine is limited by
its potential for inducing
bone marrow suppression, but this risk may be
acceptably low with monitoring. Clozapine treatment
requires obtaining the white blood cell count (WBC)
and absolute neutrophil count (ANC) at baseline and
weekly for the first six months of continuous
treatment, followed by biweekly monitoring
thereafter.
The dibenzodiazepine derivative
olanzapine has similar
properties to clozapine. In a randomized controlled
trial of 10 patients with PD, low-dose olanzapine
was effective in reducing dyskinesia, but was
associated with unacceptable increases in
parkinsonism and "off" time.
Guideline recommendations for reducing dyskinesia
An evidenced-based practice parameter from the AAN
issued in 2006 made the following recommendations
for the treatment of dyskinesia in patients with PD
and motor fluctuations: Amantadine is possibly
effective and may be considered for reducing
dyskinesia. There is insufficient evidence to
support or refute the effectiveness of clozapine in
reducing dyskinesia.
DYSTONIA Dystonia is a more sustained
abnormal posture than dyskinesia. Dystonic postures
usually involve the limbs but can affect the face,
neck, or trunk. Dystonia can be a manifestation of
early untreated Parkinson's disease (PD) or may
appear as a complication of levodopa treatment. A
careful history is required since, when due to
levodopa, dystonia can occur either as a peak
levodopa effect or during "off" periods due to
levodopa withdrawal. Withdrawal dystonia most
commonly occurs in the early morning when it
produces painful flexion and inversion postures of
the feet and toes.
Peak dystonia is managed similarly to peak
dyskinesia. "Off" period dystonia that occurs early
in the morning is managed either by taking sustained
release levodopa before retiring or by taking
levodopa or a dopamine agonist during the night or
first thing in the morning before arising. "Off"
period dystonia during the day is managed similarly
to other forms of the wearing "off" effect.
Another form of levodopa withdrawal is
akathisia (motor restlessness)
or restless legs, which usually occurs at
night, several hours after the last dose of
levodopa. This is managed by providing levodopa or a
dopamine agonist before retiring.
RECOMMENDATIONS The following treatment
suggestions represent my approach to some difficult
management issues that occur in advanced Parkinson's
disease (PD). Wearing "off" phenomenon Document the
pattern of motor fluctuations (MF). Obtain a careful
and accurate history, and observe the patient
directly in an outpatient setting. Examine the
effect of diet, and avoid taking levodopa with high
protein meals. A sustained-release levodopa
formulation may be beneficial, but only in the early
stages of wearing "off" in patients with less
advanced PD. In patients with more advanced PD,
reduce the levodopa dose interval by 30 to 60
minutes. This may require the addition of an extra
levodopa dose at the end of the day. In most cases,
individual levodopa doses should be left unchanged.
Consider adding the COMT inhibitors entacapone
(Comtan) or tolcapone (Tasmar). Entacapone should be
given first because of the small risk that tolcapone
can cause an elevation of liver enzymes. Be prepared
to lower the levodopa dose by up to 30 percent
because of the increased peak levodopa effect if
tolcapone is used.Consider adding an oral dopamine
agonist such as pramipexole or ropinirole. Watch for
dopaminergic toxicity such as visual hallucinations
and confusion, and be prepared to lower the levodopa
dose. Consider parenteral apomorphine in patients
with sudden and severe wearing "off" effects. This
rescue therapy is very effective, but it has the
disadvantage of requiring a prophylactic antiemetic
such as trimethobenzamide. In addition, the
effective dose of parenteral apomorphine must be
established for each patient by administration
during a prolonged outpatient evaluation prior to
initiating therapy. Consider the MAO B inhibitors
rasagiline and selegiline. Be aware that selegiline
exerts only a mild effect on the wearing "off"
phenomenon, while rasagiline has an effect
comparable to entacapone. Rasagiline is now approved
in the United States and in the European Union.
Unpredictable "off" periods Document that "off"
periods are unpredictable and long lasting. In many
cases, they are sudden wearing "off" effects or
transient freezing episodes.
Avoid taking levodopa with high protein meals
Evaluate and treat the possible effects of anxiety,
which may precipitate sudden "off" episodes Consider
raising the levodopa dose. Plasma levodopa levels
may be falling below the therapeutic threshold
Alternatively, consider lowering the levodopa dose.
In rare cases, sudden "off" episodes may be due to
excessive levodopa effects. Failure or delay of the
"on" response- Avoid taking levodopa with high
protein meals- Examine gastrointestinal absorption
-Avoid wearing "off" effects. Failure or delay of
the "on" responses often occur after prolonged
wearing "off" episodes
Dyskinesia and dystonia: Lower the levodopa dose
when possible. Replace a portion of the levodopa
dose with a dopamine agonist, if necessary Replace
sustained-release levodopa with regular levodopa, if
dyskinesia is occurring in the late afternoon and
evening. Add amantadine to counteract dyskinesia.
Manage diphasic dyskinesia with more frequent
levodopa dosing. Use middle-of-the-night levodopa or
a dopamine agonist to treat early morning "off"
period dystonia. Reduce the levodopa dose intervals
or add a dopamine agonist to treat "off" period
dystonia during the day.
Neurotoxic versus neuroprotective effects The
concern that prolonged use of levodopa may directly
hasten the degeneration of dopamine neurons in the
substantia nigra by promoting the generation of free
radicals and oxidative stress is the basis for
delaying the use of levodopa in the treatment of PD.
However, the evidence is not strong enough to
justify a definitive conclusion regarding levodopa
toxicity to dopamine neurons. In vitro, levodopa is
toxic to cultured dopamine neurons, although it does
not damage dopamine neurons in normal humans (who do
not have PD) or intact animals. Nevertheless, it
remains possible that levodopa is toxic in patients
with PD. In one study in rodents, for example,
levodopa increased neuronal damage in animals with
partial injury to dopaminergic neurons. However,
this was not confirmed in subsequent reports.
A consensus conference convened
to discuss the issue of levodopa toxicity reached
the following conclusions: There is no evidence that
levodopa causes neuronal death in animal models of
parkinsonism The relevance of in vitro studies of
levodopa toxicity to clinical use of levodopa is
highly uncertain There is no evidence that chronic
administration of levodopa exacerbates the
degenerative process in PD Late motor complications
arise due to the combination of progressive
degeneration of dopamine neurons and the reversible
effects of levodopa administration
Accumulating clinical trial data suggest that
levodopa, rather than being neurotoxic, either slows
the progression of PD or has a prolonged benefit
even after the drug has been stopped. To address the
continuing uncertainty surrounding the long term
effect of levodopa, the Earlier versus Later
Levodopa Therapy in Parkinson Disease (ELLDOPA)
study examined 361 patients with newly diagnosed PD
and randomly assigned them to one of three
carbidopa/levodopa doses (37.5 mg/150 mg; 75 mg/300
mg; 150 mg /600 mg) three times daily or placebo for
40 weeks, followed by withdrawal of treatment for
two weeks. At 42 weeks, when the underlying native
disease would be theoretically revealed as a result
of the two week washout, all groups assigned to
levodopa showed significantly less worsening in the
symptoms of parkinsonism (as measured by the UPDRS)
than did the placebo group. Patients receiving the
highest levodopa dose schedule (600 mg/day) had the
lowest (better) UPDRS score but also had
significantly more dyskinesia. Hypertonia,
infection, headache, and nausea were also more
common than in the placebo group. Therefore, the
clinical data suggested, surprisingly, that the use
of levodopa for 40 weeks was neuroprotective.
On the other hand, imaging data from a substudy of
116 patients supported the observations from two
previous studies that levodopa treatment is
associated with a greater decline in basal ganglia
uptake of dopamine. The substudy used single photon
emission computed tomography (SPECT) to assess
striatal dopamine by measuring [123I]beta-CIT
uptake, and showed greater reduction in
nigrostriatal dopamine transport in patients taking
levodopa compared with those on placebo. Once again,
the question of levodopa toxicity versus
levodopa-related down regulation of the dopamine
transporter receptors could not be resolved.
Therefore, the question of potential neuroprotective
versus neurotoxic effects of levodopa can not yet be
answered.
Further clinical trials are underway to study the
effects of levodopa on the progression of PD. In the
meantime, levodopa remains the most effective
therapy for PD, and should be introduced if there is
sufficient compromise of quality of life or
functional ability to warrant treatment.
MAO B inhibitors
Selegiline (Eldepryl), a selective
monoamine oxidase (MAO) type B inhibitor, is
modestly effective as symptomatic treatment for PD
and may have neuroprotective properties. In many
individuals, however, selegiline monotherapy does
not produce a functionally significant benefit,
thereby leaving patients disappointed. However, the
use of selegiline in early PD is a reasonable option
as long as the patient understands its limitations.
The selective MAO B inhibitor rasagiline has
neuroprotective properties in animal models and
appears modestly effective as symptomatic treatment
for PD in human clinical trials.
Rasagiline is
approved by the European Commission as initial
monotherapy in patients with early PD and as adjunct
treatment in moderate to advanced PD. It received
similar approval by the United States Food and Drug
Administration in May 2006.
Effectiveness Evidence supporting the
symptomatic effect of MAO B inhibitors for PD has
been bolstered by the findings of a meta-analysis
that examined data from 17 randomized trials
involving 3525 patients. These individual trials
compared MAO B inhibitors (predominately selegiline)
with either levodopa or placebo (predominately
placebo) in the treatment of early PD. Many of these
trials were limited by short-term follow-up, poor
reporting of results, and absence of quality of life
data. With these limitations in mind, the following
observations were made: Data for clinical rating
scales were available from six trials of selegiline;
treatment with MAO B inhibitors was associated with
significantly better total scores, motor scores, and
activities of daily living scores on the Unified
Parkinson's Disease Rating Scale (UPDRS) at three
months compared with controls. Data on the need for
levodopa were available from eight studies with a
median follow-up of 13 months; treatment with MAO B
inhibitors was associated with a reduction in the
need for additional levodopa compared with controls.
Data on motor complications were available from five
trials; treatment with MAO B inhibitors was
associated with a modest reduction in the
development of motor fluctuations compared with
controls. However, MAO B treatment was not
associated with a significant difference in the
incidence of dyskinesia. Data on mortality were
available from 10 trials, nine of which involved
selegiline. MAO B inhibitor treatment was not
associated with increased mortality compared with
controls, in contrast to one observational study
from the United Kingdom that showed increased
mortality in patients using selegiline. The results
of the UK study have not been confirmed by
subsequent reports, including an earlier
meta-analysis.
Additional evidence supporting the long-term
symptomatic benefit of selegiline for PD comes from
the continuation phase of a randomized controlled
trial involving 157 patients with PD, in which
patients who were initially assigned to selegiline
in the earlier phase of the study were treated with
combined selegiline and levodopa, while those
initially assigned to placebo were treated with
combined placebo and levodopa. At seven years,
treatment with the combination of selegiline and
levodopa was associated with significantly better
symptom control than treatment with placebo and
levodopa. Uncertainty remains about the relative
risks and benefits of MAO B inhibitors, as few
trials compared them with other antiparkinson
medications. Comparative data are particularly
lacking for the dopamine agonists.
Dosing The dose of selegiline used in
DATATOP was 5 mg twice daily, with the second dose
given at noon to avoid
insomnia. However, lower doses are sufficient
to induce MAO B inhibition, and 5 mg once a day in
the morning is currently recommended. Doses higher
than 10 mg daily are of no additional benefit and
may result in nonselective MAO inhibition, thereby
placing the patient at risk of hypertensive crisis
in the absence of dietary restrictions.
Adverse effects Nausea and headache are
associated with the use of MAO B inhibitors, and the
amphetamine metabolites of selegiline can cause
insomnia. Selegiline often causes confusion in the
elderly, thereby limiting its use in patients with
late-onset of disease. As previously mentioned,
selegiline enhances the effect of levodopa by
slowing its oxidative metabolism. Thus, it may
increase levodopa-induced side effects such as
dyskinesia and psychiatric toxicity. However, the
need for continued selegiline is debatable once
patients have reached the point of requiring
levodopa.
Serious adverse reactions have rarely
occurred following the concomitant use of selegiline
with tricyclic antidepressants or selective
serotonin reuptake inhibitors (SSRIs). In practice,
the vast majority of patients on these combinations
are able to tolerate them for years without
problems. However, the Physicians' Desk Reference
(PDR) warns not to use selegiline with either
tricyclics or SSRIs. The possible interaction of
SSRI and MAO B inhibitor treatment in patients with
PD is discussed in greater detail separately. Unlike
nonselective MAO inhibitors, selegiline does not
precipitate a hypertensive crisis in patients who
concomitantly ingest tyramine-containing foods.
Dopamine agonists The dopamine agonists
(DAs) are a group of synthetic agents that directly
stimulate dopamine receptors. The drugs currently
approved by the United States Food and Drug
Administration (FDA) include
bromocriptine (Parlodel),
pergolide (Permax),
pramipexole (Mirapex),
ropinirole (Requip),
and injectable apomorphine.
Apomorphine and lisuride are additional DAs
that can be administered parenterally for "rescue
therapy" in patients experiencing sudden akinetic
episodes. Lisuride is not currently approved in the
United States, but it is available in Europe.
Injectable apomorphine has been approved by the
United States FDA for treatment of motor
fluctuations in PD.
Unlike carbidopa/levodopa
(Sinemet), these drugs are direct agonists that do
not require metabolic conversion, do not compete
with amino acids for transport across the gut or
into the brain, and do not depend upon neuronal
uptake and release. An additional advantage over
immediate-release forms of levodopa is the longer
duration of action of most of these agents.
Monotherapy Dopamine agonists were
initially introduced as adjunctive treatment for
advanced PD complicated by reduced levodopa
response, motor fluctuations, dyskinesia, and other
adverse effects of levodopa. However, the
hypothetical concern that free radicals generated by
the oxidative metabolism of dopamine contribute
further to the degeneration of dopaminergic neurons
has prompted some investigators, despite lack of
conclusive evidence, to advocate the early use of
DAs as an levodopa sparing strategy.
With this approach, treatment with levodopa can be
postponed and saved for a later time in the course
of the disease, when disability worsens and the less
effective agonists no longer provide adequate
benefit. This strategy is based upon the unproven
concept that the long-term duration of a given
patient's responsiveness to levodopa is finite and
that the drug, like money in a savings or retirement
account, should be rationed. However, whether
reduced responsiveness to levodopa over time is due
to a decline in drug response or progression of
underlying PD is currently uncertain.
Controlled trials have shown that bromocriptine,
pergolide, pramipexole, and ropinirole are all
effective in patients with advanced PD complicated
by motor fluctuations and dyskinesia. However, these
drugs are ineffective in patients who have shown no
therapeutic response to levodopa.
Several studies have examined the use of DAs in
patients with early PD; pramipexole, ropinirole, and
pergolide were effective as monotherapy in patients
with early disease. In relatively long-term studies,
patients with early PD treated with DA monotherapy
have a lower incidence of dyskinesia and motor
fluctuations compared with those treated with
levodopa. As an example, one study found that
the cumulative incidence of dyskinesia over five
years was 20 percent in patients treated with
ropinirole (plus or minus supplementation with
levodopa) and 45 percent in patients treated with
levodopa [55].
A second four-year trial found a similar 20 percent
absolute reduction in the development of dyskinesia
and a 15 percent reduction in wearing "off" with
pramipexole compared with levodopa. On the other
hand, initial treatment with levodopa resulted in
lower incidences of freezing, somnolence, and leg
edema (the latter two attributable to side effects
of pramipexole) and provided for better symptomatic
control. Both treatments resulted in similar quality
of life.
In practice, while symptoms can be controlled
initially with DAs, few patients with progressive
disease can be satisfactorily maintained on DA
monotherapy for more than a few years before
levodopa is needed. Studies comparing the long-term
effects of levodopa monotherapy versus early
bromocriptine or ropinirole followed by the delayed
addition of levodopa have produced mixed results,
showing either fewer motor complications with
combined therapy or no significant difference.
Thus, the hypothesis that early DA monotherapy
reduces the future incidence of motor complications
is supported by several clinical trials, but this
benefit occurs at the expense of reduced efficacy
when compared with levodopa. Furthermore, one
comparative study found that while early treatment
with bromocriptine was associated with a slightly
lower incidence of motor complications compared with
levodopa therapy, overall disability scores were
worse in the bromocriptine group throughout the
first years of therapy.
Effectiveness The few studies that have
compared the efficacy of various DAs with each other
have found either no significant difference or only
mild superiority of one agent over another.
The two classes of DAs, ergot (bromocriptine and
pergolide) and nonergot (pramipexole and
ropinirole), stimulate dopamine D2 receptors
preferentially. Some stimulate D1 receptors. D1 and
D2 are the most important of the five known dopamine
receptors that relate to levodopa therapy.
Differences in receptor selectivity are as follows:
Pramipexole and ropinirole are nonergot compounds
that also selectively stimulate D3 dopamine
receptors in the ventral striatum, a non motor
region of the basal ganglia. Since D3 receptors are
not present in the dorsal or motor striatum, their
role in drug responsiveness in PD is unclear.
Ropinirole and pramipexole also differ from
bromocriptine and pergolide in their ability to
nonselectively stimulate serotonin or adrenergic
receptors. Bromocriptine is a mixed D1 agonist and
antagonist, and a D3 agonist. Pergolide stimulates
D1 receptors. Apomorphine is a short-acting D1 and
D2 receptor agonist.
Whether these differences will translate into
greater efficacy and less toxicity of the newer DAs
(pramipexole and ropinirole) over the older ergot
compounds (bromocriptine and pergolide) remains to
be seen. At the present time, however, there is
probably no indication to switch from one agonist to
another in a patient experiencing a satisfactory
therapeutic benefit from one agent.
Dosing The DAs generally require
administration at least three times a day at
maintenance doses: Bromocriptine (Parlodel) is
usually started at 1.25 mg twice a day; the dose is
increased at two to four week intervals by 2.5 mg a
day. Most patients can be managed on 20 to 40 mg
daily in three to four divided doses, although total
daily doses as high as 90 mg can be used.
Pramipexole (Mirapex) is usually started at 0.125 mg
three times a day. The dose should be increased
gradually by 0.125 mg per dose every five to seven
days. Most patients can be managed on total daily
doses of 1.5 to 4.5 mg. Ropinirole (Requip) is
usually started at 0.25 mg three times a day. The
dose should be increased gradually by 0.25 mg per
dose each week for four weeks to a total daily dose
of 3 mg. Most patients can be managed on this dose.
After week four, the ropinirole dose may be
increased weekly by 1.5 mg a day up to a maximum
total daily dose of 24 mg. Benefit most commonly
occurs in the dosage range of 12 to 16 mg per day.
Pergolide (Permax) is best avoided because of the
potential for cardiac valve problems (see "Valvular
heart disease" below). If used, it is usually
started at 0.05 mg a day for two days, then
increased by 0.1 or 0.15 mg a day every three days
for 12 days. After that, the dose may be increased
by 0.25 mg a day every three days until the optimal
therapeutic response is achieved. The usual total
daily dose range is 1.5 to 3 mg in three divided
doses. The maximum total daily dose is 5 mg.
Apomorphine (Apokyn) may be administered either as
intermittent rescue injections or as continuous
infusions to treat "off" episodes or
levodopa-induced motor fluctuations. A challenge
test dose must precede routine use. This is usually
done with a 2 mg subcutaneous injection under
medical supervision and monitoring of standing and
supine blood pressure before the injection, and
repeated at 20, 40, and 60 minutes after.
Antiemetic therapy (eg, with trimethobenzamide) is
initiated three days prior to starting apomorphine
and is usually continued for two months before
reassessing need. However, the use of apomorphine is
contraindicated with ondansetron and other serotonin
receptor agonists commonly used to treat nausea and
vomiting, as the combination may cause severe
hypotension and loss of consciousness. In addition,
dopamine antagonists used to treat nausea and
vomiting such as prochlorperazine and metoclopramide
should be avoided, as they may reduce the
effectiveness of apomorphine.
The usual starting dose for intermittent apomorphine
use is 2 mg, if the patient tolerates and responds
to the test dose. The dose may be increased by 1 mg
per dose every two to four days to a maximum of 6 mg
per dose. The average dosing frequency is three
times daily and should not exceed five times a day
dosing or a total daily dose of 20 mg.
Adverse effects of dopamine agonists Adverse
effects caused by dopamine agonists (DAs) are
similar to those of levodopa, including nausea,
vomiting, sleepiness, orthostatic hypotension,
confusion, and hallucinations. Peripheral edema is
common with the chronic use of DAs but is rare in
patients using levodopa alone.
These adverse effects can usually be avoided by
initiating treatment with very small doses and
titrating to therapeutic levels slowly over several
weeks. Patients intolerant of one agonist may
tolerate another. As with all of the
antiparkinsonian drugs, elderly and demented
patients are much more susceptible to psychiatric
side effects.
Accumulating evidence suggests that the use of DAs
as a class may lead to compulsive use of
dopaminergic drugs and/or impulse control disorders
in a small percentage of patients taking these
drugs. The use of ergotamine DAs is also associated
with a risk of valvular heart disease.
Adverse events with apomorphine
are usually mild and consist, predominantly of
cutaneous reactions and neuropsychiatric problems.
The incidence of these problems is higher in
patients receiving continuous infusion than in those
receiving intermittent subcutaneous injections.
Chest pain, angina, and orthostatic hypotension are
more serious problems; orthostasis peaks 20 minutes
after dosing and lasts at least 90 minutes. A test
dose of apomorphine to establish tolerance and
responsiveness is essential prior to routine
administration.
Ergot-related side effects such as Raynaud
phenomenon, erythromelalgia, and retroperitoneal or
pulmonary fibrosis are uncommon with bromocriptine
and pergolide, and they do not occur at all with the
nonergot agonists ropinirole and pramipexole.
Dopamine receptor agonists decrease prolactin
concentration. Thus, there is a potential for
decreased milk production in postpartum women taking
these agents, which are contraindicated in women who
are breast feeding. The manufacturer of pramipexole
has issued a warning regarding somnolence that can
occur abruptly and without premonition, particularly
at a dose above 1.5 mg/day. Patients with PD who
drive are at particular risk of developing these
"sleep attacks". They advise that patients be warned
of this potential side effect and asked about
factors that may increase the risk of drowsiness,
such as concomitant sedating medications, sleep
disorders, and medications that increase pramipexole
levels (eg, cimetidine).
Valvular heart disease Mounting evidence
suggests that treatment with pergolide or
cabergoline is associated with a clinically and
statistically significant risk of valvular heart
disease, with lesions similar to those associated
with carcinoid, ergot, and fenfluramine-induced
valvular disease. The risk of valvular heart disease
appears to increase relative to the cumulative dose
of pergolide or cabergoline, and the mechanism is
probably related to pergolide and cabergoline
activation of serotonin (5-HT 2B type) receptors
expressed on heart valves, which in turn leads to
valvular overgrowth. In a nested case-control study
from the United Kingdom involving a cohort of 11,417
patients taking antiparkinsonian drugs, 31 patients
with newly diagnosed cardiac valve regurgitation
were compared with 664 controls. The risk of cardiac
valve regurgitation was significantly increased with
current use of pergolide (incidence-rate ratio [IRR]
7.1, 95% CI 2.3-22.3) and cabergoline (IRR 4.9, 95%
CI 1.5-15.6) but not with other DAs. The risk of
cardiac valve regurgitation was increased only for
patients taking pergolide or cabergoline for six
months or longer, and was particularly increased for
both medications at doses higher than 3 mg daily. In
a case-control study from Italy involving 155
patients taking DAs for PD and 90 control subjects,
the frequency of clinically important valve
regurgitation determined by echocardiography was
significantly higher in patients taking pergolide or
cabergoline compared with controls (23.4 and 28.6
versus 5.6 percent, respectively) but not in those
taking nonergot DAs (0 percent).
Thus, pergolide and cabergoline should not be used
as treatment for PD. For patients already taking
these agents and doing well, we recommend discussing
the risk of valvular heart disease with the patient
and family, and switching to a non-ergot DA such as
ropinirole or pramipexole whenever possible.
Patients who choose to continue therapy with
pergolide or cabergoline despite the risk should
have periodic echocardiograms to evaluate the
cardiac valves for deformity or regurgitation.
Dopaminergic dysregulation syndrome
Compulsive use of dopaminergic drugs develops in a
small number of patients with PD and has been termed
the dopaminergic dysregulation syndrome (DDS). DDS
typically involves male patients with early onset PD
who take increasing quantities of dopaminergic drugs
despite increasingly severe drug-induced dyskinesia.
DDS can be associated with a cyclical mood disorder
characterized by hypomania or manic psychosis.
Tolerance (or frank dysphoria) to the mood elevating
effects of dopaminergic therapy develops, and a
withdrawal state occurs with dose reduction or
withdrawal. Impulse control disorders including
hypersexuality and pathologic gambling may accompany
DDS. A form of complex, prolonged, purposeless, and
stereotyped behavior called punding may be also be
associated with DDS.
DDS appears to be uncommon but not rare. In a series
of 202 patients with PD, criteria for DDS were
fulfilled in seven (3.4 percent). DDS may occur more
frequently with dopaminergic agonists than with
levodopa, but data are scarce. A small case-control
study found that susceptibility factors for DDS
included younger age at disease onset, higher
novelty seeking personality traits, depressive
symptoms, and alcohol intake.
Management of DDS is not well studied. Practitioners
should limit dopaminergic dose increases when
possible, particularly in patients who may have
increased susceptibility to DDS. Continuous
subcutaneous apomorphine infusions may be useful to
suppress off-period dysphoria, and low doses of
clozapine or quetiapine may be useful for some
patients.
Impulse control disorders Dopamine agonist
therapy may be associated with an increased risk of
impulse control disorders including pathologic
gambling, compulsive sexual behavior, or
compulsive buying, as illustrated by the following
reports: In a Canadian case series of 297 patients
with PD, the lifetime prevalence of pathologic
gambling, hypersexuality, or compulsive shopping was
13.7 percent in patients on DAs. The lifetime
prevalence of pathologic gambling for all patients
and for those receiving any DA was 3.4 and 7.2
percent, respectively, compared with a lifetime
prevalence of 1.0 percent in the Ontario population. There was a statistically significant
association of pathologic gambling and
hypersexuality with earlier PD onset and DA therapy. In line with earlier retrospective studies, pathologic gambling did not develop in
patients receiving levodopa monotherapy. Another series of 272
patients with PD found that criteria for impulse
control disorders, either anytime during the course
of PD or currently active, were met by 6.6 and 4.0
percent of patients, respectively. The
frequency of pathologic gambling, compulsive sexual
behavior, and compulsive buying anytime during PD
were 2.6, 2.6, and 1.5 percent, respectively. On
multivariate analysis, significant predictors of an
active impulse control disorder were use of a DA and
a history of impulse control disorder symptoms
before the onset of PD.
In a retrospective case series of 11 patients with
PD who developed pathologic gambling linked to DA
therapy, the pathologic gambling resolved with
tapering or discontinuation of DA therapy in all
patients available for follow-up (8 of 11). Only 1
of 11 patients in the series met criteria for the
dopaminergic dysregulation syndrome described above,
although six patients simultaneously developed other
inappropriate behaviors such as hypersexuality.
COMT inhibitors The catechol-O-methyl
transferase (COMT) inhibitors tolcapone (Tasmar) and
entacapone (Comtan) are useful as levodopa extenders. They are ineffective when given alone, but
they may prolong and potentiate the levodopa effect
when given with a dose of levodopa. These
medications are mainly used to treat patients with
motor fluctuations who are experiencing end-of-dose
wearing "off" periods. When given to patients
without motor fluctuations, entacapone did not
improve UPDRS motor scores but was associated with
several improved quality of life measures.
Inhibition of catechol-O-methyl transferase reduces
the peripheral (entacapone) and central (tolcapone)
methylation of levodopa and dopamine, which in turn
increases the plasma half-life of levodopa, produces
more stable plasma levodopa concentrations, and
prolongs the therapeutic effect of each dose. Use of
COMT inhibitors may allow a reduction in the total
daily levodopa dose by as much as 30 percent. The
net result is an increased levodopa effect.
Dosing The starting dose of tolcapone is 100 mg
three times daily; the clinical effect is evident
immediately. The dose of entacapone is one 200 mg
tablet with each dose of levodopa, up to a maximum
of eight doses per day.
Adverse effects The most common side effects of
tolcapone are due to increased dopaminergic
stimulation and include dyskinesia, hallucinations,
confusion, nausea, and orthostatic hypotension. The
adverse effects are managed by lowering the dose of
levodopa either before or after the addition of
tolcapone. Diarrhea poorly responsive to antidiarrheal medications appears in approximately 5
percent of patients. An orange discoloration of the
urine is a common but benign adverse event.
Elevations in liver enzymes may rarely occur.
Three reported deaths from hepatotoxicity in
patients using tolcapone prompted its removal from
the market in Canada and Europe, although it is
still available in the United States with the
recommendation that it be used for treatment of
motor fluctuations only after other methods have
been exhausted and with regular monitoring of ALT
and AST levels.
Side effects of entacapone are similar to tolcapone,
although entacapone has thus far not been associated
with hepatotoxicity.
Anticholinergics Dopamine and acetylcholine are
normally in a state of electrochemical balance in
the basal ganglia. In PD, dopamine depletion
produces a state of cholinergic sensitivity so that
cholinergic drugs exacerbate and anticholinergic
drugs improve parkinsonian symptoms.
Centrally acting anticholinergic drugs such as
trihexyphenidyl (Artane) and benztropine (Cogentin)
have been used for many years in PD and continue to
have a useful role. Other
anticholinergic agents such as biperiden (Akineton),
orphenadrine (Disipal), and procyclidine (Kemadrin)
produce similar effects and are more commonly used
in Europe than the United States. Benztropine also
may increase the effect of dopamine by inhibiting
its presynaptic reuptake, but it is not known
whether this contributes to its mechanism of action.
Anticholinergic drugs are most useful as monotherapy
in patients under age 70 with disturbing tremor who
do not have significant akinesia or gait
disturbance. They also may be useful in patients
with more advanced disease who have persistent
tremor despite treatment with levodopa or DAs.
Dosing Trihexyphenidyl is the most widely
prescribed anticholinergic agent, although there is
little evidence to suggest that one drug in this
class is superior to another. The starting dose of
trihexyphenidyl is 0.5 to 1.0 mg twice daily, with a
gradual increase to 2 mg three times daily.
Benztropine traditionally is more commonly used by
psychiatrists for the management of antipsychotic
drug-induced parkinsonism; the usual dose is 0.5 to
2.0 mg twice daily.
Adverse effects Adverse effects of
anticholinergic drugs are common and often limit
their use. Elderly and cognitively impaired patients
are particularly susceptible to memory impairment,
confusion, and hallucinations and should not receive
these drugs. When an anticholinergic drug is used to
treat sialorrhea or urinary frequency, peripherally
acting agents such as propantheline (probanthine)
should be used, although confusion and
hallucinations are not infrequent adverse effects
with these drugs as well. Younger patients usually
tolerate these agents better than the elderly,
although some experience dysphoric symptoms,
sedation, or memory impairment.
Peripheral antimuscarinic side effects include dry
mouth, blurred vision, constipation, nausea, urinary
retention, impaired sweating, and tachycardia.
Caution is advised in patients with known prostatic
hypertrophy or closed-angle glaucoma.
Discontinuation of anticholinergic drugs should be
performed gradually to avoid withdrawal symptoms
that may manifest as an acute exacerbation of
parkinsonism, even in those in whom the clinical
response has not seemed significant.
Amantadine Amantadine is an antiviral agent that
has mild antiparkinsonian activity. Its
mechanism of action is uncertain; it is known to
increase dopamine release, inhibit dopamine
reuptake, stimulate dopamine receptors, and it may
possibly exert central anticholinergic effects. Amantadine has N-methyl-D-aspartate
(NMDA) receptor antagonist properties that may
account for its therapeutic effect by interfering
with excessive glutamate neurotransmission in the
basal ganglia.
In early uncontrolled clinical trials, two-thirds of
patients receiving amantadine monotherapy showed an
improvement in akinesia, rigidity, and tremor.
Subsequent controlled studies demonstrated that it
was more effective than anticholinergic drugs for
akinesia and rigidity. The benefit induced by
amantadine appears to be transient in some patients;
it is best used as short-term monotherapy in those
with mild disease. Amantadine is of little benefit
when added to levodopa, although the addition of
levodopa to amantadine causes significant additive
improvement.
Amantadine in divided doses of 200 to 400 mg a day
may reduce the intensity of levodopa-induced
dyskinesia and motor fluctuations in patients with
PD. Although the published randomized trials on
amantadine in advanced PD are limited by serious
methodological flaws and small numbers of patients, experience has shown that individual patients
with advanced PD who have motor fluctuations and
dyskinesia can benefit dramatically, at least for a
while, from the addition of amantadine to a regimen
of levodopa.
Dosing The dose of amantadine in early PD is 200
to 300 mg daily; there is no evidence that larger
doses are of additional benefit. The main advantage
of this agent is a low incidence of side effects. It
is excreted unchanged in the urine and should be
used with caution in the presence of renal failure.
Adverse effects Peripheral side effects include
livedo reticularis and ankle edema, which are rarely
severe enough to limit treatment. Confusion,
hallucinations, and nightmares occur infrequently,
but unpredictably, even after long periods of use
without side effects. These effects are more likely
when amantadine is used together with other
antiparkinsonian drugs in older patients.
Estrogen Low-dose estrogen may be helpful as
adjunctive therapy in postmenopausal women with
motor fluctuations on antiparkinsonian medication.
In one study, administration of Premarin 0.625 mg
daily for eight weeks significantly improved "on"
time and motor control in such women, although it
did not result in global improvement on an
activities of daily living rating scale. There
is no evidence that estrogen has a specific effect
on dopamine receptors; the benefit attributable to
estrogen use may be related to an overall sense of
well being. It is not clear if these results would
be similar in women taking combined
estrogen/progestin therapy (necessary in women with
an intact uterus). Furthermore, concerns about
adverse effects associated with long-term
estrogen/progestin therapy may limit its use in PD.
SUMMARY AND RECOMMENDATIONS Either levodopa or a
dopamine agonist (DA) can be used initially for
patients who require symptomatic therapy.
Practitioners should always try to find the lowest
but still effective dose of dopaminergic medication,
either singly or in combination, for patients with
PD, each of whom must be evaluated and managed in a
highly individual way. Levodopa (combined with a
peripheral decarboxylase inhibitor, ie, Sinemet,
Madopar, or Prolopa) is the most effective
symptomatic therapy for Parkinson's disease (PD) and
should be introduced when the patient and physician
jointly decide that quality of life, particularly
related to job performance or self care, is
substantially compromised. However, levodopa is
associated with a higher risk of dyskinesia than the
DAs. There does not appear to be a benefit of
initiating treatment with controlled release
levodopa compared with the immediate release
preparation, and the former may limit the ability to
follow the initial response to therapy. As a result,
it is recommended that therapy be initiated with an
immediate release preparation with a subsequent
switch to controlled release if indicated. With the exception of pergolide
and cabergoline, the dopamine agonists are a useful
group of drugs that may be used either as
monotherapy in early PD or in combination with other
antiparkinsonian drugs for treatment of more
advanced disease. They are ineffective in patients
who show no response to levodopa. They possibly
delay initiation of levodopa therapy and the
subsequent appearance of levodopa dyskinesia and
motor fluctuations, but at the risk of slightly less
efficacy and increased adverse effects. Pergolide and cabergoline
should not be used for PD because of the risk of
valvular heart disease. It is reasonable to initiate
therapy with a DA in younger patients (age <65) with
PD, and with levodopa in elderly patients (age >65).
However, there are exceptions to these general
rules, and all treatments should be individualized.
Levodopa is the drug of choice if symptoms seriously
threaten the patient's lifestyle. Selegiline has
mild symptomatic benefit, and it may be used in
patients with early PD. Its use should be limited to
patients with early disease since the symptomatic
benefits are unlikely to be significant in those
with more advanced PD. Nevertheless, patients should
understand that there may not be much symptomatic
improvement if selegiline is the initial treatment
for early PD, and early follow-up and consideration
of additional symptomatic therapy should be
arranged. The value of selegiline for
neuroprotection is unclear. Rasagiline is a newer
MAO B inhibitor that produces significant benefit as
monotherapy in PD, as demonstrated in randomized
clinical trials. It therefore has a better defined
role than selegiline in the symptomatic treatment of
patients with early PD. Anticholinergic drugs should
be reserved for younger patients in whom tremor is
the predominant problem. Their use in older or
demented individuals and those without tremor is
strongly discouraged. Amantadine is a relatively
weak antiparkinsonian drug with low toxicity that is
most useful in treating patients with early or mild
PD and perhaps later when dyskinesia becomes
problematic. |