Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Drug Therapy for Parkinson’s Disease

Neal Hermanowicz, M.D.1

ABSTRACT

The fundamental concepts of the medical treatment of Parkinson’s disease are


simple, and remain based on the enhancement of dopaminergic transmission by means of
levodopa and dopamine agonists. Recently published practice parameters from the
American Academy of Neurology and an evidence-based review under the auspices of
the Movement Disorder Society provide guidance on motor complications and also
cognitive and psychiatric issues associated with Parkinson’s disease. The choices of
medications are increasing as are the routes of administration, with the arrival of injectable
and transdermal dopamine agonists and a monoamine inhibitor absorbed via the buccal
mucosa. Although simple conceptually, the actual care of patients with Parkinson’s disease
is often complex, requiring consideration of potential future complications and individu-
alized medication regimens, and minimizing the adverse effects of medications that range
from unpleasant to seriously disturbing.

KEYWORDS: Parkinson’s disease, levodopa, dopamine agonists, COMT inhibition,


MAO inhibition, dyskinesias, impulse control disorder

T he fundamental concepts of the pharmacolog- although the method of achieving optimal quality of
ical treatment of Parkinson’s disease (PD) are simple and life, our primary goal for all our patients, has not been
are almost entirely based on the deficiency of dopamine addressed by most clinical trials to date. We know
identified in 1959. Although the number of drugs has unequivocally, for example, that use of dopamine
increased and our sophistication in using them has agonists in conjunction with levodopa will reduce the
improved, levodopa and dopamine agonists, first ap- appearance of some motor complications compared
proved by the U.S. Food and Drug Administration with levodopa monotherapy. We do not know, how-
(FDA) in 1973 and 1974, respectively, remain the ever, which approach best serves our patients’ quality of
principal choices. life. Optimizing outcome from the patient’s perspec-
Although the concepts are simple and some tive guides therapy in the clinic, and in recent trials
clarity has been provided by the recent publication of more extensive quality-of-life measures have been
Practice Parameters by the American Academy of Neu- incorporated. In this article, the medical treatments
rology,1–3 the treatment of Parkinson’s disease continues for Parkinson’s disease are reviewed.
to be a daunting challenge even for clinicians with
considerable experience. Individual patient variability
of symptoms and responses to medications preclude a ACETYLCHOLINE RECEPTOR
formulaic approach to therapy. ANTAGONISTS
Some issues of therapy have been addressed by Before the arrival of levodopa, medications that block
careful analyses of evidence-based medical practice, postsynaptic acetylcholine receptors served as the

1
Department of Neurology, Movement Disorders Program, University Movement Disorders; Guest Editor, Robert L. Rodnitzky, M.D.
of California–Irvine; Phillip and Carol Traub Center for Parkinson’s Semin Neurol 2007;27:97–105. Copyright # 2007 by Thieme
Disease, Eisenhower Medical Center, Rancho Mirage, California. Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001,
Address for correspondence and reprint requests: Neal Hermano- USA. Tel: +1(212) 584-4662.
wicz, M.D., Department of Neurology, 105 Irvine Hall, University of DOI 10.1055/s-2007-971177. ISSN 0271-8235.
California–Irvine, Irvine, CA 92697-4275.
97
98 SEMINARS IN NEUROLOGY/VOLUME 27, NUMBER 2 2007

mainstay for the treatment of PD. Whether catego- to other therapies of the day, and also improvement in
rized as belladonna alkaloids (e.g., atropine) or as symptoms such as dysphagia and mental confusion now
synthetic drugs (trihexyphenidyl, benztropine, biper- regarded as poorly responsive to dopaminergic medica-
iden), the putative pharmacology was the same; central tion.8,9 Additionally, both short and longer responses to
cholinergic antagonism and the side effects mediated levodopa were reported long ago, the former after a
by central or peripheral blockade of acetylcholine are single dose and with a duration of a few hours, and the
similar among them.4 latter occurring after accumulated doses and lasting for
Although long used for treatment of PD, and days.9
probably because of their long use, the anticholinergic Of interest also was the observation of the emer-
drugs have not been subjected to the same scrutiny in gence of dyskinesias with relatively brief DOPA treat-
terms of efficacy and adverse events as more recent ment in some patients. Although the time course for
arrivals. Among them, benztropine was described as development of these movements was not specified in
particularly beneficial for tremor, a quality later ascribed detail, duration of treatment before their emergence
by some clinicians to this category in general. This ranged from 16 to 347 days. These movements were
notion of tremor specificity, however, seems to be largely noted to be evident only when the clinical improvement
propagated as folk medicine rather than based on rig- with DOPA was marked.9
orous assessment. A recent, careful literature review Modern data on the effectiveness of levodopa was
failed to locate data from clinical trials to substantiate obtained in the ELLDOPA (Early versus Late Levodopa
this belief.5 The same review was not able to identify by Therapy of Parkinson’s) trial.10 This was a multicenter
comparison study the also widely held belief that these study organized by the Parkinson Study Group and
medications are more poorly tolerated than later, ex- funded by the National Institutes of Health, with the
pressly dopaminergic drugs. One study examining anti- intent of determining whether levodopa treatment con-
cholinergic medications described an alarming increase tributed to a more rapid decline in PD. Subjects recently
in neuritic plaques and tangles correlating with chronic diagnosed and untreated were enrolled and randomly
use.6 Although to date this has been an isolated report, it assigned to placebo or to 150 mg, 300 mg, or 600 mg
does cause concern and warrants investigation by others. of levodopa divided into a regimen of three times per day
Side effects of anticholinergic medications may for 9 months. The primary outcome measure was the
include mental confusion, mydriasis with decreased score on the Unified Parkinson Disease Rating Scale
visual acuity, dry eyes, dry mouth, constipation, and (UPDRS). A secondary outcome was employed using
urinary retention. They are contraindicated in patients imaging by means of single-photon emission computed
with acute angle glaucoma and should be used cautiously tomography (SPECT) using 2-beta-carboxymethoxy-3-
in patients with peptic ulcer disease. Anticholinergic beta-(4-iodophenyl)tropane (b-CIT), a ligand for the
medications do remain in use for PD despite the lack presynaptic dopamine transporter and a presumed
of modern methods to assess their efficacy and adverse method for quantitative assessment of dopaminergic
effects. neurons. Subjects were clinically assessed during the
treatment period and again at the conclusion after a 2-
week medication washout. The ELLDOPA study re-
LEVODOPA ported for the first time in the long use of this drug a
The identification of dopamine deficiency in the brains dose-response relationship between dosage and clinical
of people with PD and the reduction of symptoms by effect. The impact of levodopa on individual symptoms,
oral supplementation of its chemical precursor, levodopa, such as tremor, rigidity, and bradykinesia, was not re-
not only remains one of the triumphs of clinical neuro- ported in the data analysis.
science, but also the cornerstone of PD medical treat- The ELLDOPA study showed that compared
ment and the basis of comparison for other medications. with placebo, levodopa improved scores of activities of
Because dopamine itself does not efficiently pass through daily living and motor assessment but not in the rela-
the blood-brain barrier, levodopa instead is provided. tively superficial analysis of mental function. Moreover,
Although in PD the numbers of nigral neurons produc- after the washout period, a significant and positive
ing dopamine are diminished by half or more, those that difference between those who received treatment and
remain have an increased rate of dopamine synthesis. the placebo group remained. One interpretation of this
Levodopa supplementation is used by these overdriven result proposed by the authors of the study was that
neurons to increase dopamine production and thereby levodopa may in fact have a protective effect and actually
reduce symptoms.7 delay disease progression. Alternatively, others have
Early reports of the treatment of parkinsonism pointed out that the washout period of 2 weeks may
with levodopa and DOPA (a racemic mixture of D and not have been sufficient. Predictably, subjects receiving
L types was used for cost reasons) described marked levodopa had more side effects than those on placebo,
improvement in symptoms, in some cases unresponsive notably dyskinesias and nausea.
DRUG THERAPY FOR PARKINSON’S DISEASE/HERMANOWICZ 99

A subgroup analysis of the subjects undergoing Currently levodopa is available in three prepara-
SPECT imaging with b-CIT showed that the group tions: (1) combined with carbidopa, (2) combined with
receiving the highest dosage of levodopa showed a carbidopa in an extended-release tablet, and (3) com-
significant reduction in binding compared with the bined with carbidopa and entacapone. The concept of an
placebo group, suggesting a greater decline in neurons. extended-release formulation of levodopa remains at-
The authors of ELLDOPA noted that this clinical/ tractive, but the present formulation does not achieve its
imaging mismatch has been observed in other studies intended result of reducing end-of-dose wearing off, nor
involving dopamine agonist drugs and posited another has it been found to reduce the appearance of other
possible explanation, that the treatment medication motor complications such as dyskinesias.18
might alter ligand binding. This is currently under
investigation, but to date the issue has not been
resolved. DOPAMINE AGONISTS
The motor complications of levodopa therapy Dopamine agonists alleviate symptoms of Parkinson’s
have been well documented, starting with early trials disease by simulating the action of dopamine at post-
and more recently in large, multicenter therapeutic synaptic receptors. Dopamine agonists have been avail-
studies. These studies report the appearance of choreic able since 1974 when bromocriptine, the first such
dyskinesias, end-of-dose wearing off in the range of 17 drug, was approved.19 Since then the number of such
to 45% and 37 to 57% of patients, respectively, after drugs available has increased to a total of four with
2 to 5 years of levodopa therapy.11–13 The pharma- pergolide, ropinirole, and pramipexole in the United
cokinetic profile of levodopa is often blamed as the States with a fifth, rotigotine, anticipated shortly.
source of these complications, and studies of animal Cabergoline, a long-acting, once-daily medication
models of PD using dopamine agonists support this widely used in Europe, is unavailable in the United
notion.14,15 States for the treatment of PD.
It is likely that the half life of levodopa is not the Dopamine agonists have some prominent advan-
sole contributing factor to the development of dyskine- tages when compared with levodopa. First, they are not
sias. Although a substantial fraction of patients will dependent on conversion to an active form by the
develop dyskinesias after treatment with levodopa for presynaptic neurons that are being depleted by PD.
2 years, the majority do not. The percentage apparently Second, as a group these medications have plasma half
increases with duration of therapy, but it remains that lives of several hours and are thus less likely to create the
some people are more likely to develop dyskinesias than motor complications of medical therapy attributed to the
others.16,17 Patients under age 50 years, for example, are shorter half life of levodopa, namely, end-of-dose wear-
recognized as being at increased risk. Why it is that some ing off and dyskinesias.
people are more likely to develop dyskinesias has not Dopamine agonists are useful agents for both
been identified. monotherapy and as adjuncts combined with levodopa.
Despite its inherent problems, it is unlikely that Both ropinirole and pramipexole, when used as mono-
levodopa will be supplanted by another medical therapy therapy for mild PD, have been reported to be equivalent
soon. Efforts to enhance the delivery of this drug and to levodopa as assessed by limited quality-of-life meas-
possibly reduce the complications attributed to its rela- ures and patient and examiner impressions in mild PD,
tively short half life are underway. but they are inferior in terms of motor improvement.13,20
The effective half life and duration of clinical Patients treated with levodopa who experience
benefit of levodopa are augmented when used in end-of-dose wearing off will have reduced off time and
combination with an inhibitor of catechol-O-methyl- may be able to reduce the levodopa dosage with the
transferase (COMT), the secondary pathway, after addition of a dopamine agonist. The reduction of off
decarboxylase, of levodopa degradation in the periph- time by adding a dopamine agonist has been reported to
ery. Currently underway is an international, multicen- be from 8 to 32%.1 It is important to emphasize that the
ter study to examine whether introduction of levodopa percentages of change in off and on time were derived
with a COMT inhibitor, specifically entacapone, will largely from different studies and, with the exception of
have an impact on the appearance of levodopa-associ- ropinirole and bromocriptine,21 not head-to-head com-
ated dyskinesia. parisons among the dopamine agonists.
A patch with transdermal delivery of levodopa has Two large, industry-sponsored clinical trials
long been discussed but so far has not been deemed reached the same conclusion that initial therapy with a
realistic because of the instability of the levodopa mol- dopamine agonist with levodopa added later forestalls
ecule with sunlight exposure and also difficulty with its the appearance of dyskinesias and also diminishes end-
permeability through the dermis. This is being reex- of-dose wearing off.13,20 An independent, rigorous ex-
amined in conjunction with stabilizers and other meth- amination of the literature established unequivocally that
ods to enhance absorption through the skin. initial treatment with a dopamine agonist in conjunction
100 SEMINARS IN NEUROLOGY/VOLUME 27, NUMBER 2 2007

with levodopa added later compared with levodopa and pramipexole in sudden, irresistible drowsiness, later
monotherapy does indeed reduce the appearance of these and more extensive examination of sleepiness in PD
problems.22 However, this analysis drew no conclusions indicates that this problem is more complex than a simple
as to which line of therapy is best as assessed by patient medication side effect.30,31 Sleep disorders are pervasive
quality-of-life measures. Preventing or forestalling the and diverse in PD, and, consequently, separating an effect
appearance of dyskinesias has been advocated as a goal from a drug from the disease itself can be challenging or
of therapy, sometimes leading to extreme avoidance of impossible.
levodopa, coined ‘‘levodopa phobia.’’23 One analysis of Another unpleasant phenomenon associated with
this problem of dyskinesias indicates that severe dyski- agonist medications that has recently received increased
nesias are an uncommon problem, more typically are not attention is the emergence of behaviors associated with
disruptive, and can be successfully addressed by medi- abnormal reward regulation, described as compulsions,
cation adjustment.24 impulse control disorders, or hedonistic homeostatic
Prescription of dopamine agonists as well as dysregulation.32–34 This has been manifested as compul-
sophistication in their use has increased in the past sive gambling; hypersexuality; and excessive eating, shop-
10 years. Now, methods of agonist administration are ping, and hobbyism. The reports to date indicate these
also increasing. The potent but short-acting agonist problems arise in association with dopamine agonists
apomorphine has long been used by episodic intramus- more so when they are used in conjunction with levo-
cular injection and subcutaneous infusion in Europe, and dopa, and affect somewhere in the range of a few to 14%
is also available as an intramuscular injection rescue of patients who use these medications.35 Although pra-
medication in the United States. Transdermal delivery mipexole has in some reports most often been associated
of a dopamine agonist has been tested for several years.25 with this phenomenon, this likely simply reflects the fact
At the time of this writing, rotigotine, a nonergot that it is more widely used in the United States than other
dopamine agonist, is available in some European coun- agonist medications. The scope of this problem is not yet
tries under the trade name Neupro as a once-daily patch clear, but it does warrant discussion with patients receiv-
used as monotherapy in early PD, and has been sub- ing dopaminergic medications. Patients may be unwilling
mitted for consideration to the U.S. FDA. Ropinirole to discuss these behaviors or fail to recognize them as
has been formulated into a prolonged release tablet possibly associated with their diagnosis and treatment.
administered once per 24 hours, and has been reported More conventional side effects of the agonist
to be beneficial as both monotherapy and as an adjunct medications as a class include nausea, orthostatic hypo-
for patients not optimally treated with levodopa alone.26 tension, dizziness, somnolence, hallucinations, and pedal
Pramipexole is under investigation in a twice-daily edema.
rather than three-times-a-day dosing regimen and at a
lower total daily dosage.
The modification of disease progression, or neu- MONOAMINE OXIDASE INHIBITORS
roprotection, has long been a goal in developing ther- Dopamine is degraded centrally to dihydroxyphenyl-
apeutics for PD. In vitro experiments point to several acetic acid (DOPAC), a process facilitated by mono-
areas where dopamine agonists may have a favorable amine oxidase (MAO). Substances that inhibit MAO
effect on reducing neuronal injury incurred by the and thus prolong the action of dopamine have been
processes of PD.27,28 Functional imaging using markers proposed for the treatment of PD since shortly after the
of dopamine metabolism both by positron emission appearance of levodopa.36
tomography (PET) and SPECT, using fluorodopa and Two isoenzymes of MAO have been identified,
b-CIT, respectively, have stimulated discussion of mod- type A and type B, according to their inhibition by
ification of neuronal survival in subjects treated with a clorgyline and selegiline, respectively.37 Both types A
dopamine agonist in comparison to those treated with and B are present in the brain and are associated with
levodopa.12,13 Both of these studies concluded that dopaminergic and serotonergic neurons. The predom-
agonist treatment had a favorable effect on altering inant type of MAO in the gut is A, which metabolizes
disease progression as measured by imaging, although tyramine. Inhibition of type A allows entry of tyramine
both studies also have been criticized on methodology to the circulation, where it may produce a pressor effect
and interpretation of results. To date no medication, mediated through the generation of sympathomimetic
including a dopamine agonist, has been definitively compounds. Selective inhibition of type B, however,
identified to slow the progression of PD. allows tyramine metabolism in the gut and thus avoids
A collection of case reports of sudden sleepiness in the risk of a hypertensive crisis, the so-called ‘‘cheese
a small number of patients treated with either ropinirole effect’’ of tyramine-containing foods.
or pramipexole invigorated examination of sleep disorders The first such medication to come into clinical use
in PD.29 Although this first report of ‘‘sleep attacks’’ and was selegiline, also known initially as deprenyl, which
associated car crashes suggested a role for ropinirole was approved in the United States in 1989 by the FDA
DRUG THERAPY FOR PARKINSON’S DISEASE/HERMANOWICZ 101

as an adjunct for patients who have experienced a decline levodopa to the CNS and thus prolong its clinical effect.
in their response to levodopa. Recently, selegiline has Of the two, tolcapone is the more potent inhibitor of
become available in a freeze-dried tablet preparation COMT. At 1 hour, tolcapone achieves red blood cell
(trade name Zelapar) that dissolves rapidly when placed COMT inhibition of 80% compared with 70% with
on the tongue. Intended initially for patients with entacapone; at 4 hours, the red blood cell COMT
dysphagia, it was determined that when administered inhibition was 60% and 10% for tolcapone and entaca-
in this fashion, selegiline is rapidly absorbed through the pone, respectively.43
buccal mucosa. By avoiding hepatic metabolism on first The administration regimens reflect these proper-
entering the circulation, the dosage in this form is ties of COMT inhibition. Entacapone is given at a fixed
reduced and may also be administered only once daily dose of 200 mg with each dose of levodopa, whereas
rather than twice a day as with the swallowed tablets. tolcapone is administered at 100 mg or 200 mg three
This administration method also reduces the production times a day at 6-hour intervals.
of amphetamine metabolites, suspected by some to Both tolcapone and entacapone reduce off time
contribute to adverse effects such as sleep disruption and increase on time in patients with motor fluctuations.
and agitation. This selegiline preparation at a dose of 2.5 In clinical trials of fluctuating patients, off time has been
mg once daily has been reported to reduce off time by 2 decreased by approximately an hour per day with en-
hours each day in patients with motor fluctuations.38 tacapone and 2 to 3 hours with tolcapone in relation to
Selegiline has not been demonstrated to be of value as a placebo.44,45 However, these numbers are not derived
monotherapy. from studies involving head to head comparison of
Rasagiline (trade name Azilect) has recently be- entacapone with tolcapone.
come available and received approval by the FDA as both Dyskinesia may be enhanced by the addition of a
monotherapy and an adjunct medication for PD. In a 26- COMT inhibitor, although this can be addressed by
week, randomized, double-blind, placebo-controlled trial reducing the levodopa dosage. Urine and other bodily
in subjects with mild PD receiving no other antiparkinson fluids may be discolored to an orange or amber hue,
medication (anticholinergics were allowed), rasagiline which is distressing to patients if they are not notified
showed modest efficacy.39 A second study of rasagiline beforehand but otherwise has no significance. Diarrhea
as an adjunct medication for subjects treated with levo- is induced in 5 to 10% of patients using either
dopa and experiencing motor fluctuations showed a entacapone or tolcapone. This sometimes sounds attrac-
reduction of off time of approximately 1 hour compared tive to people who are chronically constipated, but when
with placebo with a similar increase in on time.40 Rasagi- it does occur it too is distressing. People who are
line does not produce amphetamine metabolites. unaware of this potential side effect may be subjected
Both selegiline and rasagiline continue to be of unnecessarily to tests, such as colonoscopy, to identify
interest as agents that may modify the progression of the cause of the diarrhea.
PD. Laboratory investigations of both drugs indicate a The prescription of tolcapone dropped drastically
protective effect in models of PD.41,42 Selegiline was when three cases of fulminate liver toxicity
examined as a possible disease-slowing agent in the were associated with this drug.46 Tolcapone was re-
large, multicenter DATATOP (Deprenyl And Toco- moved from some European countries, and in the
pherol Antioxidative Therapy Of Parkinsonism) study, United States its prescription now requires use first of
which generated several publications and discussions but other reasonable alternatives, a signed patient consent,
was inconclusive regarding neuroprotection.11 Rasagi- and discontinuation of the drug if there is no benefit
line is currently being examined for its potential benefit after 3 weeks of use. Monitoring of serum transaminase
in slowing the progression of PD in a multicenter, levels monthly for 6 months is required and continued
multinational study. thereafter according to the clinician’s judgment.
The suspicion that the short half life of levodopa
contributes to the appearance of motor complications
CATECHOL-O-METHYLTRANSFERASE has lead to efforts to modify this by use of COMT
INHIBITORS inhibition, and studies are currently underway to ascer-
COMT metabolizes levodopa to the clinically useless tain whether this will be successful.
3-O-methyldopa both in the periphery and central
nervous system (CNS). This pathway assumes greater
significance in the periphery when the primary route for AMANTADINE
levodopa degradation, decarboxylation, is inhibited by Amantadine has long been used to treat the primary
carbidopa or benserazide. symptoms of PD,47 and it continues to be used despite
Tolcapone first became available in the United the lack of clarity of its pharmacological mechanism and
States in 1997 and was soon followed by entacapone. the arrival of levodopa and dopamine agonists. It re-
Both provide inhibition of COMT to enhance delivery of mains the choice of some clinicians for treating mild
102 SEMINARS IN NEUROLOGY/VOLUME 27, NUMBER 2 2007

symptoms when a dopaminergic medication is judged to subjects compared with placebo. Nausea and vomiting
be not yet necessary. Early double-blind trials indicated associated with rivastigmine were problematic in some
its efficacy in alleviating some of the primary symptoms subjects. Rivastigmine has been approved by the U.S.
of PD.48 FDA for the treatment of mild to moderate PD
In recent years, amantadine has risen in prom- dementia. At this writing, no controlled studies have
inence as the only medication that both alleviates symp- been published on the use of galantamine for PD
toms of PD and also reduces medication-induced dementia.
dyskinesia. Reported long ago as having a beneficial effect Memantine as a putative glutamate antagonist
on chorea associated with Huntington’s disease,49 this would be more attractive as an agent to improve both
antichorea effect was rediscovered in the choreic dyski- motor and cognitive symptoms in PD, but no such
nesias of treated PD.50 This was an important finding for supportive data have been published. Moreover, given
an old drug that eases management of an otherwise tricky its antiglutamate activity, it might be thought to reduce
problem, reducing dyskinesias without reducing mobility. dyskinesias, along the lines of amantadine. Again, no
Regarded by some as a relatively benign medica- such data have been published.
tion for mild PD, the side effects of amantadine can be
troubling or even harrowing. Livedo reticularis, a mot-
tled skin rash, may be induced by the drug, although this MEDICATIONS FOR AUTONOMIC
is often of no clinical significance other than the cos- DYSFUNCTION ASSOCIATED WITH
metic effect. More problematic is the leg edema that may PARKINSON’S DISEASE
be caused by amantadine, and the hallucinations some- Another very troubling symptom group is attributed to
times created by this drug are as troubling as any other autonomic dysfunction in PD. This includes sialorrhea,
PD therapy. Withdrawal of amantadine should be ac- orthostatic hypotension, urinary urgency and frequency,
complished slowly and with caution because its abrupt delayed gastric emptying and gastrointestinal transit,
cessation has been associated with acute delirium.51 constipation, and erectile and other sexual dysfunction.
All of these are common in PD, and all may and often do
occur in a single patient.
MEDICATIONS FOR COGNITIVE Large, placebo-controlled studies have not been
IMPAIRMENT ASSOCIATED WITH accomplished for many of these problems, but some data
PARKINSON’S DISEASE are available and pharmacological methods for address-
Dopaminergic medications often provide substantial ing these symptoms are often employed by practitioners.
improvement in the motor symptoms of PD but do Sialorrhea may be reduced by cautious use of
not necessarily address many of the nonmotor problems, small quantities of sublingual atropine 1% solution57
such as autonomic dysfunction, sleep disturbance, and or, failing this, by injection of the parotid glands with
cognitive impairment. These can be disabling and may botulinum toxin.58 Orthostatic hypotension can be
be the patient’s most prominent complaint. Some cog- improved by use of increased salt intake and volume-
nitive functions are influenced by levodopa or other expanding drinks. A second step is to use medications
dopaminergic medications,52–54 although the clinical that promote volume retention, fludrocortisone and
benefit, if any, may not be of significance to patients or midodrine.59 The former is a steroid and as such
their family members. should be used knowledgably in this osteoporosis-
No drugs have been developed to specifically prone group. A more recent method to stabilize pos-
address cognitive dysfunction in PD, although in prac- tural fall in blood pressure has been with pyridostig-
tice medications developed for Alzheimer’s disease have mine, ostensibly to increase sympathetic output by
often been prescribed in an off-label fashion. With the acting at the cholinergic, autonomic ganglion. This
exception of memantine, all of these medications are has been reported to be successful with and without the
cholinesterase inhibitors and, following the logic that use of midodrine.60,61
anticholinergic medications improve motor symptoms of Urinary urgency and frequency generally require
PD, it has been intuitively suspected these drugs may investigation by a urologist to characterize the mecha-
produce motor worsening. nism of the symptoms before initiating treatment, par-
Rivastigmine and donepezil have both been ticularly in men in whom bladder outflow obstruction by
examined in well-organized studies as agents to treat prostate enlargement may be a contributing factor.
dementia in PD, although the number of subjects in the Peripherally acting anticholinergic agents may be of
donepezil study was relatively small.55,56 Both drugs benefit in alleviating overactive bladder symptoms,62
show modest benefit in some measures of cognitive but an undesired consequence mediated by the same
function. Donepezil was not found to worsen scores on pharmacology may be exacerbation of problems with gut
the UPDRS, although rivastigmine was associated with motility and constipation and, as suggested in the
increase in tremor in a relatively small percentage of preceding paragraph, blood pressure volatility.
DRUG THERAPY FOR PARKINSON’S DISEASE/HERMANOWICZ 103

Erectile dysfunction occurs commonly among men 4. Onuaguluchi G. Drug treatment of parkinsonism and its
with PD and may respond to medications developed for assessment. In: Vinken PJ, Bruyn GW, eds. Handbook of
this problem such as sidenafil.63 Clinical Neurology. New York: John Wiley & Sons Inc;
1968:218–226
Constipation is another common PD problem; it
5. Katzenschlager R, Sampaio C, Costa J, Lees A. Anticholi-
was described in detail by James Parkinson in his 1817 nergics for symptomatic management of Parkinson’s disease.
monograph, and it still has relevance today. It is dis- Cochrane Database Syst Rev 2003;(2):CD003735
tressing to patients and difficult to satisfactorily treat. 6. Perry EK, Kilford L, Lees AJ, Burn DJ, Perry RH. Increased
Although the package inserts for dopaminergic medi- Alzheimer pathology in Parkinson’s disease related to
cations cite constipation as a potential side effect, the antimuscarinic drugs. Ann Neurol 2003;54:235–238
source of this in many patients may be primarily from 7. Lloyd KG, Davidson L, Hornykiewicz O. The neuro-
chemistry of Parkinson’s disease: effect of L-dopa therapy.
PD itself rather than from its treatment. Exercise, fiber,
J Pharmacol Exp Ther 1975;195:453–464
and fluids, the usual recommendations to alleviate con- 8. Cotzias GC, Papavasiliou PS, Gellene R. Experimental
stipation, usually do not work and more creative meth- treatment of parkinsonism with L-Dopa. Neurology 1968;
ods are necessary. Lactulose works but too often results 18:276–277
in unpleasant cramping, flatulence, and loose stools. 9. Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino
Polyethylene glycol, like lactulose, is an osmotic agent acids and modification of parkinsonism. N Engl J Med 1967;
effective for constipation but seems less likely to create 276:374–379
10. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the
the unpleasantness of the latter.
progression of Parkinson’s disease. Parkinson Study Group.
N Engl J Med 2004;351:2498–2508
11. Parkinson Study Group. Impact of deprenyl and tocopherol
CONCLUSION treatment on Parkinson’s disease in DATATOP patients
The heterogeneity of the clinical manifestations, the requiring levodopa. Parkinson Study Group. Ann Neurol
diversity of response to medications, and the potential 1996;39:37–45
for adverse effects all contribute to the challenge of the 12. Parkinson Study Group. Pramipexole vs levodopa as initial
treatment for Parkinson disease: A randomized controlled
medical management of Parkinson’s disease. Successful
trial. Parkinson Study Group. JAMA 2000;284:1931–
symptom management often is not a simple task that can 1938
be taught or learned in a strictly formulaic manner; it 13. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study
requires time, attention, and communication with our of the incidence of dyskinesia in patients with early
patients. When done poorly, the outcome can be more Parkinson’s disease who were treated with ropinirole or
distressing to patients than the symptoms of the disease levodopa. 056 Study Group. N Engl J Med 2000;342:1484–
the drugs are intended to alleviate. When done well, it 1491
14. Blanchet PJ, Calon F, Martel JC, et al. Continuous
results in sustained enhancement of quality of life and
administration decreases and pulsatile administration increases
reassurance to the patients and their families that this behavioral sensitivity to a novel dopamine D2 agonist
disease can be successfully treated and managed. (U-91356A) in MPTP-exposed monkeys. J Pharmacol Exp
Ther 1995;272:854–859
15. Bibbiani F, Costantini LC, Patel R, Chase TN. Continuous
REFERENCES dopaminergic stimulation reduces risk of motor complica-
tions in parkinsonian primates. Exp Neurol 2005;192:73–
1. Pahwa R, Factor SA, Lyons KE, et al. Quality Standards 78
Subcommittee of the American Academy of Neurology. 16. Schrag A, Ben-Shlomo Y, Brown R, et al. Young-onset
Practice Parameter: treatment of Parkinson disease with motor Parkinson’s disease revisited: clinical features, natural history,
fluctuations and dyskinesia (an evidence-based review): report and mortality. Mov Disord 1998;13:885–894
of the Quality Standards Subcommittee of the American 17. Sato K, Hatano T, Yamashiro K, et al. Prognosis of
Academy of Neurology. Neurology 2006;66:983–995 Parkinson’s disease: time to stage III, IV, V, and to motor
2. Miyasaki JM, Shannon K, Voon V, et al. Quality Standards fluctuations. Juntendo Parkinson Study Group. Mov Disord
Subcommittee of the American Academy of Neurology. 2006;21:1384–1395
Practice Parameter: evaluation and treatment of depression, 18. Koller WC, Hutton JT, Tolosa E, Capilldeo R. Immediate-
psychosis, and dementia in Parkinson disease (an evidence- release and controlled-release carbidopa/levodopa in PD:
based review): report of the Quality Standards Subcommittee a 5-year randomized multicenter study. Carbidopa/Levodopa
of the American Academy of Neurology. Neurology 2006; Study Group. Neurology 1999;53:1012–1019
66:996–1002 19. Calne DB, Teychenne PF, Claveria LE, et al. Bromocriptine
3. Suchowersky O, Gronseth G, Perlmutter J, et al. Quality in parkinsonism. BMJ 1974;4:442–444
Standards Subcommittee of the American Academy of 20. Holloway RG, Shoulson I, Fahn S, et al. Pramipexole vs
Neurology. Practice Parameter: neuroprotective strategies levodopa as initial treatment for Parkinson disease: a 4-year
and alternative therapies for Parkinson disease (an evidence- randomized controlled trial. Arch Neurol 2004;61:1044–
based review): report of the Quality Standards Subcommittee 1053
of the American Academy of Neurology. Neurology 2006; 21. Brunt ER, Brooks DJ, Korczyn AD, Montastruc JL,
66:976–982 Stocchi F. A six-month multicentre, double-blind,
104 SEMINARS IN NEUROLOGY/VOLUME 27, NUMBER 2 2007

bromocriptine-controlled study of the safety and efficacy of 39. Parkinson Study Group. A controlled trial of rasagiline in
ropinirole in the treatment of patients with Parkinson’s early Parkinson disease: the TEMPO Study. Arch Neurol
disease not optimally controlled by L-dopa. 043 Study 2002;59:1937–1943
Group. J Neural Transm 2002;109:489–502 40. Parkinson Study Group. A randomized placebo-controlled
22. Goetz CG, Poewe W, Rascol O, Sampaio C. Evidence-based trial of rasagiline in levodopa-treated patients with Parkinson
medical review update: pharmacological and surgical treat- disease and motor fluctuations: the PRESTO study. Arch
ments of Parkinson’s disease: 2001 to 2004. Mov Disord Neurol 2005;62:241–248
2005;20:523–539 41. Wu RM, Murphy DL, Chiueh CC. Neuronal protective and
23. Kurlan R. ‘‘Levodopa phobia:’’ a new iatrogenic cause of rescue effects of deprenyl against MPP þ dopaminergic
disability in Parkinson disease. Neurology 2005;64:923–924 toxicity. J Neural Transm Gen Sect 1995;100:53–61
24. Van Gerpen JA, Kumar N, Bower JH, Weigand S, Ahlskog 42. Youdim MB, Wadia A, Tatton W, Weinstock M. The anti-
JE. Levodopa-associated dyskinesia risk among Parkinson Parkinson drug rasagiline and its cholinesterase inhibitor
disease patients in Olmsted County, Minnesota, 1976–1990. derivatives exert neuroprotection unrelated to MAO inhib-
Arch Neurol 2006;63:205–209 ition in cell culture and in vivo. Ann NY Acad Sci 2001;
25. The Parkinson Study Group. A controlled trial of rotigotine 939:450–458
monotherapy in early Parkinson’s disease. Arch Neurol 43. Nutt JG. Effect of COMT inhibition on the pharmacoki-
2003;60:1721–1728 netics and pharmacodynamics of levodopa in parkinsonian
26. Stocchi F, Hersh BP, Earl NL, Scott BL. Safety and patients. Neurology 2000;55(suppl 4):S33–S37
tolerability of ropinirole 24-hour prolonged release in 44. Adler CH, Singer C, O’Brien C, et al. Randomized, placebo-
patients with early and advanced Parkinson’s disease. Poster controlled study of tolcapone in patients with fluctuating
presentation P894, Movement Disorder Society 10th Inter- Parkinson disease treated with levodopa-carbidopa. Tolca-
national Congress of Parkinson’s Disease and Movement pone Fluctuator Study Group III. Arch Neurol 1998;55:
Disorders; Kyoto, Japan: 2006 1089–1095
27. Yoshioka M, Tanaka K, Miyazaki I, et al. The dopamine 45. Parkinson Study Group. Entacapone improves motor
agonist cabergoline provides neuroprotection by activation of fluctuations in levodopa-treated Parkinson’s disease patients.
the glutathione system and scavenging free radicals. Neurosci Ann Neurol 1997;42:747–755
Res 2002;43:259–267 46. Assal F, Spahr L, Hadengue A, Rubbia-Brandt L, Burkhard
28. Le WD, Jankovic J, Xie W, Appel SH. Antioxidant property PR. Tolcapone and fulminant hepatitis. Lancet 1998;352:
of pramipexole independent of dopamine receptor activation 958
in neuroprotection. J Neural Transm 2000;107:1165–1173 47. Schwab RS, England AC Jr, Poskanzer DC, Young RR.
29. Frucht S, Rogers JD, Greene PE, Gordon MF, Fahn S. Amantadine in the treatment of Parkinson’s disease. JAMA
Falling asleep at the wheel: motor vehicle mishaps in persons 1969;208:1168–1170
taking pramipexole and ropinirole. Neurology 1999;52:1908– 48. Dallos V, Heathfield K, Stone P, Allen FA. Use of
1910 amantadine in Parkinson’s disease: results of a double-blind
30. Arnulf I, Konofal E, Merino-Andreu M, et al. Parkinson’s trial. BMJ 1970;4:24–26
disease and sleepiness: an integral part of PD. Neurology 49. Scotti G, Spinnler H. Amantadine and Huntington’s chorea.
2002;58:1019–1024 N Engl J Med 1971;285:1325–1326
31. Gjerstad MD, Alves G, Wentzel-Larsen T, Aarsland D, 50. Adler CH, Stern MB, Vernon G, Hurtig HI. Amantadine in
Larsen JP. Excessive daytime sleepiness in Parkinson disease: advanced Parkinson’s disease: good use of an old drug.
is it the drugs or the disease? Neurology 2006;67:853–858 J Neurol 1997;244:336–337
32. Giovannoni G, O’Sullivan JD, Turner K, Manson AJ, Lees 51. Factor SA, Molho ES, Brown DL. Acute delirium after
AJ. Hedonistic homeostatic dysregulation in patients with withdrawal of amantadine in Parkinson’s disease. Neurology
Parkinson’s disease on dopamine replacement therapies. 1998;50:1456–1458
J Neurol Neurosurg Psychiatry 2000;68:423–428 52. Costa A, Peppe A, Dell’Agnello G, et al. Dopaminergic
33. Driver-Dunckley E, Samanta J, Stacy M. Pathological modulation of visual-spatial working memory in Parkinson’s
gambling associated with dopamine agonist therapy in disease. Dement Geriatr Cogn Disord 2003;15:55–66
Parkinson’s disease. Neurology 2003;61:422–423 53. Molloy SA, Rowan EN, O’Brien JT, et al. Effect of levodopa
34. Pontone G, Williams JR, Bassett SS, Marsh L. Clinical on cognitive function in Parkinson’s disease with and without
features associated with impulse control disorders in dementia and dementia with Lewy bodies. J Neurol Neuro-
Parkinson disease. Neurology 2006;67:1258–1261 surg Psychiatry 2006;77:1323–1328
35. Voon V, Hassan K, Zurowski M, et al. Prevalence of 54. Cools R. Dopaminergic modulation of cognitive function-
repetitive and reward-seeking behaviors in Parkinson disease. implications for L-DOPA treatment in Parkinson’s disease.
Neurology 2006;67:1254–1257 Neurosci Biobehav Rev 2006;30:1–23
36. Hunter KR, Boakes AJ, Laurence DR, Stern GM. Mono- 55. Ravina B, Putt M, Siderowf A, et al. Donepezil for dementia
amine oxidase inhibitors and L-dopa. Br Med J 1970;3:388 in Parkinson’s disease: a randomised, double blind, placebo
37. Youdim MB, Edmondson D, Tipton KF. The therapeutic controlled, crossover study. J Neurol Neurosurg Psychiatry
potential of monoamine oxidase inhibitors. Nat Rev Neurosci 2005;76:934–939
2006;7:295–309 56. Emre M, Aarsland D, Albanese A, et al. Rivastigmine for
38. Waters CH, Sethi KD, Hauser RA, Molho E, Bertoni JM. dementia associated with Parkinson’s disease. N Engl J Med
Zydis selegiline reduces off time in Parkinson’s disease 2004;351:2509–2518
patients with motor fluctuations: a 3-month, randomized, 57. Hyson HC, Johnson AM, Jog MS. Sublingual atropine for
placebo-controlled study. Zydis Selegiline Study Group. Mov sialorrhea secondary to parkinsonism: a pilot study. Mov
Disord 2004;19:426–432 Disord 2002;17:1318–1320
DRUG THERAPY FOR PARKINSON’S DISEASE/HERMANOWICZ 105

58. Lagalla G, Millevolte M, Capecci M, Provinciali L, Ceravolo 61. Singer W, Opfer-Gehrking TL, Nickander KK, Hines SM,
MG. Botulinum toxin type A for drooling in Parkinson’s Low PA. Acetylcholinesterase inhibition in patients with
disease: a double-blind, randomized, placebo-controlled study. orthostatic intolerance. J Clin Neurophysiol 2006;23:476–
Mov Disord 2006;21:704–707 481
59. Senard JM, Brefel-Courbon C, Rascol O, Montastruc JL. 62. Winge K, Fowler CJ. Bladder dysfunction in Parkinsonism:
Orthostatic hypotension in patients with Parkinson’s disease: mechanisms, prevalence, symptoms, and management. Mov
pathophysiology and management. Drugs Aging 2001;18: Disord 2006;21:737–745
495–505 63. Zesiewicz TA, Helal M, Hauser RA. Sildenafil citrate
60. Singer W, Sandroni P, Opfer-Gehrking TL, et al. Pyridos- (Viagra) for the treatment of erectile dysfunction in
tigmine treatment trial in neurogenic orthostatic hypoten- men with Parkinson’s disease. Mov Disord 2000;15:305–
sion. Arch Neurol 2006;63:513–518 308

You might also like