LEVODOPA

CASE 59-1, QUESTION 9: L.M. has responded well to pramipexole 1 mg 3 times daily for the past 18

months, with an increased ability to paint and carry out ADLs. During the past few weeks, however, he has

noticed a gradual worsening in his symptoms and once again is having difficulty holding a paintbrush. He

currently complains of feeling more “tied up,” he has more difficulty getting out of a chair, and his posture is

slightly more stooped. He also notes that he feels tired throughout much of the day. He remains able to carry

out most of his ADLs without assistance. Should levodopa be considered for the treatment of his PD symptoms

at this time?

Dopamine itself does not cross the blood–brain barrier. Levodopa, a dopamine

precursor with no known pharmacologic action of its own, crosses the blood–brain

barrier, where it is converted by the enzyme aromatic amino acid (dopa)

decarboxylase to dopamine. Levodopa has been a mainstay of PD treatment since the

1960s. Nearly all patients will eventually require treatment with the drug, regardless

of their initial therapy.

Because significant amounts of levodopa are peripherally (extracerebrally)

metabolized to dopamine by dopa decarboxylase, extremely high doses are necessary

if administered alone. For this reason, levodopa is always coadministered with a

dopa decarboxylase inhibitor. This allows the dose of levodopa to be decreased by

75%.

73 By combining levodopa with a dopa decarboxylase inhibitor that does not

penetrate the blood–brain barrier, a decrease in the peripheral conversion of

levodopa to dopamine can be achieved, whereas the desired conversion within the

striatum remains unaffected. This combination also shortens the time needed to

achieve optimal effects by several weeks, because the dopa decarboxylase inhibitor

substantially decreases dose-limiting levodopa-induced nausea and vomiting. The

two peripheral decarboxylase inhibitors in clinical use are benserazide (unavailable

in the US) and carbidopa. A fixed combination of carbidopa and levodopa is

available in ratios of 1:4 (carbidopa/levodopa 25/100) and 1:10

(carbidopa/levodopa 10/100 and 25/250). Carbidopa/levodopa is also available as

an immediate-release, orally disintegrating tablet and extended-release capsule, both

of which may be used in early PD. In addition, controlled-release, extended-release,

and intestinal gel

p. 1257

p. 1258

formulations are available and are discussed for management of motor

complications.

The optimal time to initiate levodopa therapy must be individualized because

chronic levodopa use is associated with the development of motor complications and

dyskinesias. These observations led to the understanding that chronic levodopa

therapy may actually accelerate the neurodegenerative process through formation of

free radicals generated by dopamine metabolism.

74 The Earlier versus Later

Levodopa Therapy in Parkinson’s Disease (ELLDOPA) study was designed to

determine whether long-term use of levodopa accelerates neurodegeneration and

paradoxically worsens PD.

74 After 42 weeks, the severity of symptoms as measured

by changes in the total UPDRS score increased more in the placebo group than in all

of the groups receiving levodopa. This validated that levodopa use does not result in

accelerated progression of the disease based on clinical evaluations. In untreated

individuals, there is little reason to start levodopa until the patient reports worsening

of function (socially, vocationally, or otherwise). The need for levodopa therapy may

be delayed by initiating therapy first with a dopamine agonist or MAO-B inhibitor.

This approach is particularly advantageous in younger patients who will likely live

many years with PD and have a high lifetime risk for developing motor

complications. In the case of L.M., he is now experiencing bothersome symptoms

despite near-maximal dopamine agonist therapy, and it has progressed sufficiently to

threaten his job performance. Although the dose of pramipexole could be increased,

he may experience more daytime somnolence; thus, levodopa should be added to his

regimen.

Dosing

CASE 59-1, QUESTION 10: The decision is made to begin L.M. on carbidopa/levodopa. How should it be

dosed?

About 70 to 100 mg/day of carbidopa is necessary to saturate peripheral dopa

decarboxylase.

73

It is usually unnecessary and more costly to administer carbidopa in

higher amounts. Therapy should be initiated with immediate-release

carbidopa/levodopa 25 mg/100 mg 3 times a day. The immediate-release formulation

is preferred because it allows for much easier dose adjustment. In the case of L.M.,

the dose can then be increased by 100 mg of levodopa every day or every other day

up to eight tablets (800 mg) or to the maximal effective dose, to individual

requirements, or as tolerated.

Troublesome peak-dose dyskinesias may occur as a result of high levels of

dopaminergic drug activity. If these occur, the dose or frequency may be adjusted,

while giving consideration to balancing the most useful dose (e.g., maximizing the

patient’s on-time) with that which does not produce unacceptable side effects (e.g.,

troublesome dyskinesias). Because L.M. is currently being treated with a dopamine

agonist, he must be monitored closely for the development of motor complications

with the addition of levodopa.

Most patients with early PD will begin therapy on levodopa 300 mg/day and will

demonstrate response before reaching 1,000 mg/day.

11

,

73 When levodopa doses

exceed 750 mg/day, patients such as L.M. can be switched from the 1:4 ratio of

carbidopa/levodopa to the 1:10 ratio to prevent providing excessive amounts of

decarboxylase inhibitor. For example, if L.M. needed 800 mg/day of levodopa, two

carbidopa/levodopa 10/100 tablets 4 times daily could be administered. If L.M. had

not been initially treated with a dopamine agonist, some clinicians would consider

adding a dopamine agonist after the daily levodopa dose has been increased to more

than 600 mg because dopamine agonists directly stimulate dopamine receptors, have

longer half-lives, and result in a lower incidence of dyskinesias, thus providing a

smoother dopaminergic response.

Clinical response to levodopa therapy may be altered by modifying dietary amino

acid ingestion.

73 Levodopa is actively transported across the blood–brain barrier by

a large neutral amino acid transport system. This transport system also facilitates the

blood-to-brain transport of amino acids such as L-leucine, L-isoleucine, L-valine, and

L-phenylalanine. Levodopa and these neutral amino acids compete for transport

mechanisms, and high plasma concentrations of these amino acids can decrease brain

concentrations of levodopa.

75 Patients should be instructed to take immediate-release

carbidopa/levodopa 30 minutes before or 60 minutes after meals for optimal

efficacy. If nausea develops and administration with food is considered, a lowprotein meal should be encouraged.

Adverse Effects

CASE 59-1, QUESTION 11: Since initiating carbidopa/levodopa, L.M. reports feeling restless and agitated at

times. To what extent is levodopa contributing to these problems? How should they be managed?

Although levodopa is the most effective agent for PD, it is associated with many

undesirable side effects, such as nausea, vomiting, and anorexia (50% of patients);

postural hypotension (30% of patients); and cardiac arrhythmias (10% of patients).

11

In addition, mental disturbances are encountered in 15% of patients, and abnormal

involuntary movements (dyskinesias) can be seen in up to 55% of patients during the

first 6 months of levodopa treatment.

76

Although more commonly encountered with dopamine agonists, psychiatric side

effects are also associated with levodopa therapy and include confusion, depression,

psychosis, hypomania, and vivid dreams.

73 Those with underlying or preexisting

psychiatric disorders and those receiving high doses of levodopa for prolonged

periods are at greatest risk.

77 Concurrent anticholinergics or amantadine therapy can

exacerbate these symptoms. Advancement of the PD itself correlates with cognitive

decline and greater frequency of CNS findings, possibly mediated through an

underlying Lewy body pathology.

17

In some situations, it may be difficult to separate

the respective drug from disease effects.

Some patients receiving levodopa experience psychomotor excitation (e.g.,

overactivity, restlessness, agitation). Similarly, hypomania has been reported in up to

8% of patients and is characterized by grandiose thinking, flight of ideas, tangential

thinking, and poor social judgment. Normal sexual activity often is restored with

improved motor function; however, hypersexuality and libido are increased in about

1% of levodopa-treated patients.

77

In general, most of the mental disturbances are dose-related and can be lessened

by reducing the dose of the dopaminergic agent. In patients such as L.M. who are

concurrently receiving levodopa and a dopamine agonist, the dose reduction should

be attempted first with the dopamine agonist. If symptoms do not improve, a

reduction in the dose of levodopa may also be warranted. These dose reductions

may, however, be impractical for L.M. because a return of parkinsonian symptoms is

likely, and the benefits of levodopa therapy may outweigh the risk for mental

disturbances.

ANTICHOLINERGICS

CASE 59-1, QUESTION 12: What role do anticholinergic drugs play in the treatment of PD? Should L.M.

receive an anticholinergic agent?

Anticholinergic drugs have been used to treat PD since the mid-1800s, when it

was discovered that symptoms were reduced by the belladonna derivative hyoscine

(scopolamine).

17 Currently,

p. 1258

p. 1259

they are used to specifically target tremor. Although tremor may improve with

dopaminergic drugs via the restoration of balance between dopamine and

acetylcholine, resolution of tremor may be incomplete. Anticholinergic drugs such as

benztropine and trihexyphenidyl work by blocking the excitatory neurotransmitter

acetylcholine in the striatum, which minimizes the effect of the relative increase in

cholinergic sensitivity. Until the late 1960s, when amantadine and levodopa were

introduced, anticholinergics were a mainstay of treatment; however, because of their

undesirable side effect profile and poor efficacy relative to levodopa in treating

bradykinesia and rigidity, anticholinergic agents are no longer used as first-line

agents. These drugs produce both peripherally and centrally mediated adverse

effects. Peripheral effects, such as dry mouth, blurred vision, constipation, and

urinary retention, are common and bothersome.

20 Anticholinergic agents can increase

intraocular pressure and should be avoided in patients with angle-closure glaucoma.

CNS effects can include confusion, impairment of recent memory, hallucinations, and

delusions.

17

,

20 Patients with PD are often more susceptible to these central effects

because of advanced age, intercurrent illnesses, and impaired cognition.

20

Anticholinergics are usually reserved for the treatment of resting tremor early in the

disease, particularly in younger patients with preserved cognitive function. Given his

history and clinical presentation and based on the absence of tremor, L.M. would not

be an appropriate candidate for an anticholinergic drug.

Treatment through the Progression of Parkinson’s

Disease

LEVODOPA AND ADJUNCTIVE THERAPY

The chronic, progressive nature of PD predicts that patients will develop worsening

of motor control through the later stages of the disease; thus, enhanced motor control

through the use of levodopa, in conjunction with symptomatic adjunctive agents, will

be necessary. Agent selection is guided by efficacy for motor symptom control,

adverse effects, and motor complication risk. Agents indicated by the AAN,

16

EFNS/MDS-ES,

18 and NICE

17 guidelines for symptomatic adjunct therapy and

management of motor complications in late PD are outlined in Figure 59-3.

MOTOR COMPLICATIONS

CASE 59-1, QUESTION 13: L.M. had a dramatic improvement in all of his parkinsonian symptoms with the

initiation of levodopa therapy after being maintained on carbidopa/levodopa 25 mg/250 mg 4 times a day.

After 6 months of treatment, he began to experience dyskinesias occurring 1 to 2

hours after a dose. They manifested as facial grimacing, lip smacking, tongue

protrusion, and rocking of the trunk. Decreasing his pramipexole dose to 0.5 mg 3

times daily and gradually decreasing his dosage of carbidopa/levodopa to 25 mg/250

mg 3 times daily lessened the symptoms. After 3 years of levodopa therapy, more

serious problems have begun to emerge. In the mornings, L.M. often experiences

immobility. Nearly every day, he has periods (lasting for a few minutes) in which he

cannot move, followed by a sudden switch to a fluid-like state, often associated with

dyskinetic activity. He continues to take carbidopa/levodopa 3 times daily, but gains

symptomatic relief for only 3 to 4 hours after a dose. Also, the response to a given

dose varies and is often less in the afternoon. At times, he becomes “frozen,”

particularly when he needs to board an elevator or is required to move quickly. What

are possible explanations for these alterations in clinical response?

Though variable, the initial levodopa response period may last up to 5 years. After

this initial period of stability, 50% to 90% of patients receiving levodopa for 5 or

more years will eventually experience motor complications.

78 Motor complications

may present as periods of either too much (dyskinetic) dopaminergic activity, too

little (akinetic) dopaminergic activity, or in combination.

In evaluating these motor fluctuations, it is important to ascertain which effects are

attributable to the disease and which are attributable to the drug. For example,

levodopa-induced peak-dose dyskinesias often appear concurrently with the

development of motor fluctuations.

78

,

79 Peak-dose choreiform dyskinesias, brief,

irregular and jerky movements, are the most common form of dyskinesias that occur

with chronic levodopa (and sometimes dopamine agonist) therapy. These symptoms

frequently subside at the end of the dosing interval because levodopa levels fall.

Their severity is related to levodopa dose, disease duration and stage, and younger

age at onset.

79

If peak-dose dyskinesias occur, the following strategies should be

considered: The levodopa dose can be lowered but given more frequently;

consideration can be given to switching patients to immediate-release tablets if

taking modified-release formulations of carbidopa/levodopa (for ease in refining

dose adjustments); agents that prolong the half-life of levodopa but do not provide

stable levodopa plasma concentrations (e.g., COMT inhibitor, MAO-B inhibitor) can

be added; or an antidyskinetic agent such as amantadine can be used. Variations in

the rate and extent of levodopa absorption, dietary substrates (e.g., large neutral

amino acids) that compete with cerebral transport mechanisms, levodopa drug–drug

interactions (Table 59-4), and competition for receptor binding by levodopa

metabolites can further explain the variable responses observed to levodopa.

Figure 59-3 Levodopa metabolism in the human body. AADC, aromatic amino acid decarboxylase; COMT,

catechol-O-methyltransferase; DOPAC, 3,4-dihydroxyphenylacetic acid; MAO, monoamine oxidase; 3-MT, 3-

methoxytyramine.

p. 1259

p. 1260

Two of the more common motor complications are the wearing-off effect and the

on–off effect. The wearing-off effect is most predictable, occurring in the latter part

of the dosing interval after a period of relief. Because levodopa is a short-acting

agent with an elimination half-life of about 1.5 hours, much of the effect from the

evening dose will dissipate by morning.

73

,

78 For this reason, it is not surprising that

L.M. is experiencing a period of immobility on arising. This is alleviated in most

patients shortly after taking the morning dose. Wearing off can be improved by

various means such as shortening the dosing interval or with the addition of

adjunctive therapy (if not already present) with a dopamine agonist, MAO-B

inhibitor or levodopa extender such as a COMT inhibitor. The on–off effect is

described as random fluctuations from mobility (often associated with dyskinesias)

to the parkinsonian state, which appear suddenly as if a switch has been turned on or

off. These fluctuations can last from minutes to hours and increase in frequency and

intensity with time. Despite accompanying dyskinesias, most patients prefer to be in

an on rather than an off (or akinetic) state; however, for some patients, dyskinesias

can be more disabling than parkinsonism.

Early in the course of disease, it is usually possible to control parkinsonian

symptoms without inducing dyskinesias via medication adjustments; however,

because the disease advances and the therapeutic window narrows, cycling between

on periods complicated by dyskinesia and off periods with resulting immobility is

common.

78

,

79 Eventually, despite adjustments in levodopa dose, many patients with

advanced PD experience either mobility with severe dyskinesias or complete

immobility. In most patients, this effect has no clear relationship to the timing of the

dose or levodopa serum levels.

78

The pathophysiologic basis for motor complications and dyskinesias is not entirely

clear, but incomplete delivery of dopamine to central receptors is likely

responsible.

78 Because the disease progresses and dopamine terminals are lost, the

capacity to store dopamine presynaptically is diminished, impairing the ability to

maintain relatively constant striatal dopamine concentrations.

78 As a consequence,

dopamine receptors are subject to intermittent, pulsatile stimulation, rather than a

more physiologic tonic stimulation. Overactivity of excitatory pathways mediated by

neurotransmitters such as glutamate may also be involved.

78

CASE 59-1, QUESTION 14: What options are available to reduce the motor fluctuations experienced by

L.M.?

MODIFIED-RELEASE CARBIDOPA/LEVODOPA FORMULATIONS

A more sustained delivery of levodopa than that achieved with routine oral dosing

may theoretically reduce motor complications, by more effectively replicating normal

physiology. Three products designed to prolong carbidopa/levodopa delivery

include a controlled-release tablet, an extended-release capsule, and an enteral

suspension administered as a gastrointestinal infusion. The use of the enteral

suspension for gastrointestinal infusion has shown reductions in off-time of 1.91

hours (P = 0.0018) and an increase in on-time of 1.86 hours (P = 0.0059) in a 12-

week study of patients with severe motor fluctuations and hyper/dyskinesias.

80 As a

result of its pump administration via a permanent duodenal/upper jejunal tube, this

method is typically reserved as last-line therapy for use in specialized centers.

Controlled-Release Carbidopa/Levodopa

A controlled-release (CR) tablet formulation of carbidopa/levodopa is available,

containing 25-mg carbidopa and 100-mg levodopa or 50-mg carbidopa and 200 mg

levodopa in an erodible polymer matrix that retards gastric dissolution. Although offtime should theoretically be reduced by the slower rate of plasma levodopa decline,

clinical study has generally not found a sustained difference in off-time, or a

reduction in dyskinesias, with the CR preparation compared with the immediaterelease preparation.

17

,

18 As a result, switching to CR carbidopa/levodopa as a

primary strategy to reduce off-time or lessen dyskinesias is not recommended.

17

A likely reason for the lack of superior effect with CR carbidopa/levodopa as

compared with the immediate-release formulation is its variable absorption.

Controlled-release carbidopa/levodopa is about 30% less bioavailable than the

immediate-release formulation. Patients converted from standard

carbidopa/levodopa to the CR formulation should receive a dose that will provide

10% more levodopa, and then the dose should be titrated upward to clinical

reponse.

81

Interestingly, administration with food enhances levodopa peak by 25%.

Given that there is no obvious advantage to CR carbidopa/levodopa, L.M. should not

be switched to this formulation.

Extended-Release Carbidopa/Levodopa

The extended-release (ER) capsule formulation of carbidopa/levodopa contains a

combination of both immediate- and extended-release beads designed to circumvent

the pharmacokinetic concerns seen with the CR tablet formulation. Although both

formulations persist for up to 6 hours, the ER capsule provides a faster time to

symptom relief, with a peak that is comparable to the immediate-release formulation

(1 hour vs. up to 2 hours for CR).

82 Similar to CR, the ER formulation reduces

levodopa bioavailability by approximately 50%.

83 As such, the manufacturer

provides specific dose conversions from immediate-release preparations. Converse

to the CR preparation, high-fat and high-calorie food reduces levodopa peak and may

delay absorption by 2 to 3 hours, and it is recommended that the first dose of the day

be given 1 to 2 hours prior to eating. In patients with difficulty swallowing, these

capsules may be opened and sprinkled on applesauce, which is an additional

advantage to the CR formulation.

The ability of the ER formulation to reduce off-time was evaluated in a placebocontrolled study randomizing 393 patients to immediate or ER formulations over 22

weeks.

84 After being titrated to a stable dose during weeks 1 to 9, patients were

followed for an additional 13 weeks. Compared to baseline evaluations, extendedrelease patients had a reduction in off-time by approximately 1 hour/day (P<0.0001)

when compared to the IR formulation. This effect was accompanied by an increase in

on-time without troubling dyskinesia of approximately 1 hour (P = 0.0002). Notably,

of patients converted per manufacturer direction, 60% of patients required a further

dosage increase and 13% required a dosage decrease.

84

p. 1260

p. 1261

Table 59-4

Levodopa Drug Interactions

Drug Interaction Mechanism Comments

Anticholinergics ↓ Levodopa effect ↓ Gastric emptying,

thus ↑ degradation of

levodopa in gut, and

Watch for ↓ levodopa effect when

anticholinergics cause ↓ GI motility. When

anticholinergic therapy is discontinued in a

↓ amount absorbed patient on levodopa, watch for signs of

levodopa toxicity. Theoretic interaction with

minor clinicalsignificance

Ferrous sulfate ↓ Levodopa oral

absorption by 50%

Formation of

chelation complex

Avoid concomitant administration or

separate administration by at least 2 hours

Food ↓ Levodopa effect Large, neutral amino

acids compete with

levodopa for

intestinal absorption

Although levodopa is usually taken with

meals to slow absorption and ↓ central

emetic effect, high-protein diets should be

avoided

MAOI (e.g.,

phenelzine,

tranylcypromine)

Hypertensive crisis Peripheral dopamine

and norepinephrine

Avoid combination use; monoamine oxidase

type B (MAO-B) inhibitors such as

selegiline can be used, but the dose of

levodopa should be reduced after 2–3 days

of therapy; carbidopa might minimize

hypertensive reaction to levodopa in

patients receiving an MAOI.

Methyldopa ↑ or ↓ levodopa

effect

Acts as central and

peripheral

decarboxylase

inhibitor

Observe for response; may need to switch

to another antihypertensive.

Metoclopramide ↓ Levodopa effect Central dopamine

blockade

Avoid combination use

Neuroleptics (e.g.,

butyrophenones,

phenothiazines)

↓ Levodopa effect Central blockade of

dopamine

neurotransmission

Avoid combination use; important

interaction

Phenytoin ↓ Levodopa effect Mechanism

unknown

Avoid combination use

Pyridoxine (vitamin

B6)

↓ Levodopa effect Peripheral

decarboxylation of

levodopa

Not observed when levodopa given with

carbidopa; avoid levodopa monotherapy

with vitamin B6 supplementation

Tricyclic

antidepressants

↓ Levodopa effect Levodopa

degradation in gut

because of delayed

emptying

Use with caution

GI, gastrointestinal; MAOI, monoamine oxidase inhibitor.

Based upon this evidence, the ER formulation may improve off-time for L.M., but

is unlikely to impact his dyskinesias. Therefore, it may not be appropriate to switch

therapy. Taking his immediate-release carbidopa/levodopa more frequently while

avoiding substantial increases in the total daily dose, which could worsen his

dyskinesias, may improve his condition. Taking his morning dose before arising from

bed may help with his early-morning problems. If the symptoms that L.M. is

experiencing are not improved with these changes, a number of adjunctive agents

such as dopamine agonists, apomorphine rescue, amantadine, COMT inhibitors, and

MAO-B inhibitors can be considered.

DOPAMINE AGONISTS

The effectiveness of pramipexole added to levodopa therapy in advanced PD was

evaluated in a multicenter, placebo-controlled study of 360 patients with a mean

disease duration of 9 years.

85 Pramipexole was titrated gradually to the maximal

effective dose as tolerated. At the end of a 6-month maintenance period, patients

treated with pramipexole had a 22% improvement in their ADLs (P<0.0001) and a

25% improvement in their motor scores (P<0.01) compared with baseline values.

Patients treated with pramipexole also had a 31% improvement in the mean off-time,

compared with a 7% improvement in the placebo-treated group (P<0.0006).

Dyskinesias and hallucinations were more common in pramipexole-treated patients

and necessitated levodopa dose reduction in 76% of patients compared with 54% in

the placebo group. The total daily levodopa dose was decreased by 27% in those

treated with pramipexole compared with 5% in the placebo group.

Ropinirole has been found to improve motor scores when added to levodopa

therapy in patients with advanced-stage disease.

53

In a multicenter, double-blind,

randomized parallel-group study, patients treated with ropinirole achieved a greater

reduction in percent of time spent off during waking hours when compared to placebo

(11.7% vs. 5.1%), which was a difference of 0.4 hours. A greater amount of

ropinirole-treated patients achieved a significant 20% reduction in time spent off

(35% vs. 13%, P = 0.003). In those treated with levodopa, the levodopa dose was

decreased by an average of 19%.

Long-acting dopamine agonist formulations, including ER ropinirole and

rotigotine, have also shown reductions in off-time. In a study of 208 PD patients not

optimally controlled with levodopa after up to 3 years of therapy with less than 600

mg/day of levodopa, a prolonged-release, once-daily formulation of ropinirole was

found to improve motor scores in a similar fashion to increase the levodopa dose;

however, only 3% of ropinirole-treated subjects experienced dyskinesias compared

with 17% of levodopa-treated patients (P<0.001).

86

In moderate-to-advanced PD,

treatment benefits were observed within 2 weeks of initiation.

87 Extended-release

ropinirole has been found to reduce off-time more than the immediate-release

preparation in advanced PD. A randomized, double-blind trial of 343 patients over

24 weeks found that compared to the immediate-release formulation, significantly

more patients given the ER formulation maintained at least a 20% reduction in offtime (adjusted OR: 1.82, 1.16–2.86, P = 0.009).

88 These results are confounded by

the relatively higher doses of ER ropinirole and greater reductions in levodopa

achieved by the ER formulation. Rotigotine has also been evaluated for reduction in

off-time in advanced PD patients and

p. 1261

p. 1262

has been shown to significantly reduce off-time by approximately 2.5 hours, with

benefits seen as early as the first week of therapy.

50

Because L.M. has advanced disease and is experiencing motor fluctuations despite

a treatment regimen that includes a dopamine agonist, further dose adjustments of the

dopamine agonist may provide little additional benefit. Any adjustments must be

made with consideration of worsening his dyskinesias and the possibility of

exacerbating CNS adverse effects.

Apomorphine

Apomorphine is an injectable dopamine agonist that is approved as rescue therapy

for treatment of hypomobility or off episodes in patients with PD. In a randomized,

double-blind, parallel-group study of 29 patients, rescue treatment with apomorphine

resulted in a 34% reduction in off-time, approximately 2 hours, compared with 0% in

the placebo group (P = 0.02).

89 Mean UPDRS motor scores were reduced by 23.9

points (62%) in those treated with apomorphine, compared with 0.1 (1%) in those

receiving placebo (P<0.001). Adverse events in the apomorphine group included

yawning (40%), dyskinesias (35%), drowsiness or somnolence (35%), nausea or

vomiting (30%), and dizziness (20%), although only yawning was statistically

different from placebo (40% vs. 0%; P = 0.03).

Because nausea and vomiting frequently occur with apomorphine treatment, it

should be administered with an antiemetic such as trimethobenzamide. The antiemetic

should be started 3 days before initiating apomorphine and continued for the first 2

months of treatment.

89 Apomorphine should not be used with ondansetron and other

serotonin antagonists used to treat nausea because the combination may cause severe

hypotension. In addition, other antiemetics, such as prochlorperazine and

metoclopramide, should not be given concurrently because these dopamine

antagonists may decrease the effectiveness of apomorphine.

Doses of apomorphine range from 2 to 6 mg per subcutaneous injection. A 2-mg

test dose is recommended while monitoring blood pressure. If tolerated, the

recommendation is to start with a dose of 2 mg and increase the dose by 1 mg every

few days if needed. Peak plasma levels are observed within 10 to 60 minutes after

dosing, so the onset of therapeutic effect is rapid. The test dose and titration are timeconsuming and must be done under physician supervision, and patients may require

someone else to inject the drug once hypomobility has occurred. In severe cases,

subcutaneous infusions have been used, but are limited to research settings and highlevel clinical centers. For these reasons, apomorphine is not widely used. Given that

L.M. is experiencing motor fluctuations almost daily, long-term frequent

apomorphine use would not be a viable solution in his situation.

AMANTADINE

The antiviral agent, amantadine, was serendipitously found to improve PD symptoms

when a patient given the drug for influenza experienced a remission in her

parkinsonism.

90 Amantadine reduces all the symptoms of parkinsonian disability in

about 50% of patients, usually within days after starting therapy; however, early

trials indicated that amantadine efficacy might be limited by the development of

tachyphylaxis within 1 to 3 months.

91 Although the trials suggested reductions in

dyskinesias by up to 40%, previous evaluations of amantadine for the reduction of

dyskinesias have been limited by methodological concerns.

The pathophysiologic basis for the efficacy of amantadine in PD is not entirely

understood; it may augment dopamine release and possibly inhibit its reuptake.

92

Anticholinergic action has also been suggested. Excess glutamatergic activity has

been implicated in the pathophysiology of dopaminergic dyskinesias; amantadine has

been found to be an antagonist at N-methyl-D-aspartate (NMDA) receptors and to

block glutamate transmission.

93 Amantadine has consistently shown approximately

50% reductions in dyskinesia severity and duration, without adversely impacting

motor performance.

94–97 Two trials have evaluated long-term efficacy of amantadine

in patients with levodopa-induced dyskinesia after treatment for 6 months to 1 year.

These trials assessed for change in UPDRS motor examination and motor

complication subscores with amantadine washout over 3 weeks to 3 months. Those

discontinuing amantadine had deterioration in levodopa-induced dyskinesia

compared to the continuing group within a median time of 7 days suggesting the

possibility of continued benefit despite the tachyphylaxis suggested in earlier

trials.

98

,

99 Guidance suggests reserving consideration for amantadine use to those in

whom dyskinesia is not adequately managed on other therapies.

17

The decision to use amantadine in L.M. should be based on whether his

dyskinesias are deemed more problematic than his duration of off-time. If so,

amantadine should be started at 100 mg/day taken at breakfast; an additional 100-mg

dose can be taken with lunch 5 to 7 days after initiation. The dose can be increased to

a maximum of 300 mg/day; however, doses in excess of 200 mg/day are associated

with increased adverse effects and should be used cautiously. Amantadine is renally

excreted, and the dose should be reduced in patients with renal impairment.

100

If the

dyskinesias that L.M. is experiencing are tolerable but the duration of off-time is

more problematic, then selection of another agent such as a COMT inhibitor or

MAO-B inhibitor may be more appropriate than the initiation of amantadine at this

time.

Side effects of amantadine mainly involve the gastrointestinal (e.g., nausea,

vomiting) and CNS (e.g., dizziness, confusion, insomnia, nightmares, and

hallucinations). Patients receiving concomitant anticholinergic therapy may

experience more prominent CNS side effects.

100 Livedo reticularis, a rose-colored

mottling of the skin usually involving the lower extremities, can occur with

amantadine as early as 2 weeks after initiating therapy. The consequences of livedo

reticularis are entirely cosmetic, and discontinuation of therapy is unnecessary. Ankle

edema may be seen in association with livedo reticularis. Elevation of the legs,

diuretic therapy, and dosage reduction often alleviate the edema.

CATECHOL-O-METHYLTRANSFERASE INHIBITORS

COMT is an enzyme widely distributed throughout the body that is responsible for

the biotransformation of many catechols and hydroxylated metabolites, including

levodopa. When carbidopa, an inhibitor of aromatic AAD, is coadministered with

levodopa, the peripheral conversion of levodopa to dopamine via this pathway is

inhibited; as a consequence, the conversion of levodopa to 3-O-methyldopa (3-

OMD) by COMT is amplified and becomes the major metabolic pathway for

levodopa degradation. The metabolite 3-OMD lacks antiparkinsonian activity and

may compete with levodopa for transport into the circulation and brain. By

preventing its peripheral degradation, the therapeutic effect of levodopa can be

extended via inhibition of COMT.

Entacapone and tolcapone are selective, reversible, and potent COMT inhibitors

that increase the amount of levodopa available for transport across the blood–brain

barrier (Figure 59-4) to prolong its therapeutic effect. Use of these agents is

associated with increased on-time and a decrease in the daily levodopa dose.

101

,

102

The pharmacologic and pharmacokinetic effects of entacapone and tolcapone are

compared in Table 59-5.

101–103 Two uncontrolled trials and one controlled evaluation

of patients with motor fluctuations newly placed on entacapone compared switching

patients to tolcapone versus continuing the newly started entacapone. These trials

alluded to the therapeutic benefits of tolcapone compared to entacapone; however,

tolcapone is associated with cases of fatal, acute fulminant liver failure. This has led

to stringent liver function monitoring requirements and limited clinical use. If

initiated, liver function monitoring should be performed at baseline and every 2 to 4

weeks for the first 6 months, followed periodically thereafter as clinically

necessary.

101 Because of the risks for hepatotoxicity associated with tolcapone,

entacapone is the preferred COMT inhibitor and would be a good choice for L.M. if

he desires an increase in his on-time.

p. 1262

p. 1263

Figure 59-4 Suggested treatment algorithm for the management motor complications in late PD. DBS, deep brain

stimulation.

Entacapone

CASE 59-1, QUESTION 15: Six months after adjusting the frequency of his carbidopa/levodopa dose and

adding amantadine, L.M. reports that his dyskinesias are not too bothersome, but he is now having increased

periods (lasting a few minutes) in which he cannot move. He is currently taking amantadine 100 mg twice daily,

pramipexole 0.5 mg 3 times daily, and immediate-release carbidopa/levodopa 25 mg/250 mg 5 times a day, but

“even on a good day” gains symptomatic relief for only about 2 to 3 hours after a dose. The decision is made to

initiate entacapone therapy and gradually discontinue pramipexole as guided by symptoms or side effects. How

effective is entacapone for reducing the symptoms of PD?

Early initiation of a COMT inhibitor at the same time that levodopa is first

introduced has been proposed as a way of reducing the development of levodopainduced motor complications

104

; theoretically, this strategy should provide more

stable plasma levels of levodopa and lessen pulsatile stimulation of striatal

dopamine receptors normally observed with intermittent levodopa dosing. This

strategy was tested in the Stalevo Reduction in Dyskinesia Evaluation in Parkinson

Disease (STRIDE-PD) study, a multicenter, double-blind study that randomly

assigned 747 patients to initiate either carbidopa/levodopa or

carbidopa/levodopa/entacapone 4 times daily.

104 Surprisingly, patients randomly

assigned to receive carbidopa/levodopa/entacapone actually had a shorter time to

onset of dyskinesia (hazard ratio: 1.29; P = 0.04) and increased dyskinesia frequency

at week 134 (42% vs. 32%; P = 0.02). These findings may have been confounded by

an increased use of dopaminergic therapy in the entacapone group. The findings of

the STRIDE-PD study do not support the early administration of entacapone in

combination with levodopa to reduce the occurrence of motor complications.

Table 59-5

Pharmacologic and Pharmacokinetic Properties of Catechol-OMethyltransferase Inhibitors

Tolcapone Entacapone

Bioavailability 65%–68% 35%

Tmax

(hours) 1.7 1

Protein binding 99.9% 98%

Metabolism Glucuronidation; CYP3A4,

CYP2A6

methylated by COMT

Isomerization, glucuronidation

Half-life (hours) 2–3 Biphasic; 0.4–0.7, 2.4

Time to reverse COMT inhibition

(hours)

16–24 8

Maximal COMT inhibition at 200-mg

dose

>80% 65%

Increase in levodopa AUC 100% 35%

Increase in levodopa half-life 75% 85%

Dosing method TID, spaced 6 hours apart With every administration of

levodopa

AUC, area under the curve; COMT, catechol-O-methyltransferase; CYP, cytochrome P-450; TID, three times

daily.

p. 1263

p. 1264

The efficacy and safety of entacapone as an adjunct to levodopa therapy in the

presence of motor complications was established in two pivotal multicenter,

randomized, double-blind, placebo-controlled trials.

105

,

106 Subjects for both studies

had idiopathic PD with motor fluctuations, including wearing-off phenomena, despite

maximal tolerated doses of levodopa. In both trials, patients were randomly assigned

to receive either entacapone 200 mg or placebo (up to 10 doses/day) with each dose

of carbidopa/levodopa.

105

,

106

In both trials, significant improvements in off-time

(approximately 1 hour), UPDRS scores (10% improvement), and levodopa doses

(reductions of approximately 80–100 mg/day) were consistent.

Dosing

CASE 59-1, QUESTION 16: When should entacapone be initiated in L.M., and how should it be dosed?

Entacapone is approved for use as adjunctive therapy to levodopa for the treatment

of PD in patients experiencing wearing-off or end-of-dose deterioration. It is given

as one 200-mg tablet with each carbidopa/levodopa administration, up to eight

tablets/day. It is available in a combination tablet with a 1:4 ratio of immediaterelease carbidopa/levodopa that patients can be switched to once they are stabilized

on individual formulations of carbidopa/levodopa and entacapone. If dyskinesias

occur, it may be necessary to lower the levodopa dose by approximately 10% to

25%, particularly if the patient is receiving more than 800 mg/day of levodopa.

Although pramipexole is being discontinued in L.M., he should still be monitored for

dyskinesias, especially during the first few weeks of therapy, as it may also be

necessary to lower his carbidopa/levodopa dose.

Adverse Effects

CASE 59-1, QUESTION 17: What are the adverse effects of entacapone, and how should they be

managed?

Most entacapone-induced adverse effects are consistent with increased levodopa

exposure. They include dyskinesias (50%–60%), dizziness (10%–25%), nausea

(15%–20%), and hallucinations (1%–14%).

105

,

106 Reducing the levodopa dosage by

10% to 15% as a strategy for circumventing these effects is successful in about onethird of patients experiencing dyskinesias. Other adverse effects related to

entacapone include urine discoloration (11%–40%), diarrhea (10%), and abdominal

pain (6%).

105

,

106 Urine discoloration (brownish orange) is attributed to entacapone

and its metabolites and is considered benign, but patients should be counseled

regarding this effect. The most common reason for withdrawal from clinical studies

and discontinuation of therapy was severe diarrhea (2.5%).

107 Although the results of

the STRIDE-PD study indicated that patients taking the combination of

carbidopa/levodopa/entacapone may be at an increased risk for cardiovascular

events (e.g., heart attack, stroke, and cardiovascular death) compared with those

taking carbidopa/levodopa, subsequent FDA analysis has found no increased risk.

108

MONOAMINE OXIDASE-B INHIBITORS

Selegiline

Selegiline may have a role as a symptomatic adjunct to levodopa in more advanced

disease. Studies have found improvement in the wearing-off effect in 50% to 70% of

patients and a reduction in as much as 30% in the total daily dose of levodopa

without improvements in on-time.

109

,

110 This improvement has also been shown to

arrive with an initial worsening of dyskinesis in 60% of treated patients which may

be counteracted with levodopa dose reductions. Selegiline has been shown to reduce

off-time by 32% (2.2 hours) compared with 9% (0.6 hours; P<0.001) for placebo in

a 12-week, randomized, multicenter, parallel-group, double-blind study, but was not

reproducible in an identical trial.

111

,

112 Authors hypothesize that this conflict may be

due in part to large and potentially variable placebo effects seen in PD trials. The

on–off effect is less responsive to the addition of selegiline.

Rasagiline

Rasagiline has also been studied as an adjunct to levodopa in advanced disease.

When added to levodopa therapy, rasagiline can improve motor fluctuations,

reducing off-time by 1.4 hours and 1.8 hours (for the 0.5- and 1-mg/day groups,

respectively) compared with 0.9 hours for placebo.

113 Patients treated with 0.5

mg/day and 1 mg/day of rasagiline had 0.49 hour (0.08–0.91; P = 0.02) and 0.94 hour

(1.36–0.51; P<0.001) less off-time compared with placebo. Significant

improvements were reported in the UPDRS subscores for ADLs in the off state and

motor performance in the on state, as well as clinician global assessments.

Dyskinesias were slightly worsened in the 1-mg/day group. As adjunctive therapy to

levodopa, rasagiline appears to provide similar benefit to entacapone.

16

In the

Lasting effect in Adjunct therapy with Rasagiline Given Once daily (LARGO) study,

when compared with entacapone 200 mg administered with each levodopa dose,

rasagiline 1 mg/day reduced total daily off-time in a similar manner (decrease of

21% or 1.18 hours for rasagiline and 21% or 1.2 hours for entacapone).

114 Reduction

of levodopa dose may be necessary if dyskinesias occur when rasagiline is added in

combination with levodopa.

Safinamide

Safinamide (Xadago) was approved by the FDA in May 2017 as an adjunct to

carbidopa/levodopa for patients experiencing “off” episodes. It is not approved as

monotherapy and thus has not been previously discussed. The side effect profile is

similar to rasagiline (see Table 59-2). Patients should additionally be monitored for

visual changes as retinal detachment and loss of photoreceptor cells were noted in

animal studies. The list of contraindicated medications is extensive and should be

reviewed prior to starting therapy. Safinamide has a half–life of 20-26 hours, reaches

steady state in 5-6 days, and can be taken without regard to meals. It requires hepatic

dose adjustment but no renal dose adjustments. The extent of use in clinical practice

and eventual place in therapy are unknown at this time.

Surgical Therapies for PD

CASE 59-1, QUESTION 18: L.M. is now classified as Hoehn and Yahr

2

late stage 3. He is now on

amantadine 100 mg twice daily, immediate-release carbidopa/levodopa 25 mg/250 mg 5 times daily, and

entacapone 200 mg 5 times daily with carbidopa/levodopa administrations. His overall control of his PD has

diminished greatly in the last couple of months. His on-time averages around 6 hours/day, with the majority of it

accompanied by troublesome dyskinesias. Most days he needs some assistance with ADLs. His cognitive

function remains well preserved, and he is not depressed. He has heard about surgical procedures that might

benefit patients with PD. Is surgical therapy superior to medical therapy in patients with advanced PD?

Two types of surgical therapies have been used in patients with advanced PD who

cannot be adequately controlled with medications. The first involves making an

irreversible surgical lesion in a specific location in the brain (e.g., posteroventral

pallidotomy or stereotaxic thalamotomy); the second involves surgical implantation

of a device that sends electrical impulses to specific parts of the brain (e.g., deep

brain stimulation [DBS]) (Figure 59-5). Posteroventral pallidotomy has been shown

to reduce dyskinesias on the contralateral side and may permit the use of higher

dosages of levodopa for managing rigidity and bradykinesia.

115 However, a

significant disadvantage is the need to make a lesion near the optic tract, which may

risk visual loss. Other possible risks of pallidotomy include weakness, paralysis, and

hemorrhage that can cause stroke and speech difficulty. Stereotaxic thalamotomy has

been shown to reduce symptoms of debilitating tremor and improve rigidity in

patients with PD.

116 This intervention has eliminated contralateral tremor in 80% of

patients, and improvement has been sustained for up to 10 years. However, as with

pallidotomy, a disadvantage of thalamotomy is the need to make an irreversible

lesion in the basal ganglia that may limit the effectiveness of newer procedures

because they become available. Thus, DBS is now the preferred surgical method for

treating advanced PD that cannot be adequately controlled with medications. DBS

uses an implanted electrode in the brain, in either the subthalamic nucleus (STN) or

the globus pallidus interna (GPi), which is connected to a subcutaneously implanted

pacemaker. This permits delivery of high-frequency stimulation to the desired target.

Advantages of DBS include no need for an irreversible brain lesion, and it provides

flexibility for altering the target site and program stimulation parameters.

p. 1264

p. 1265

Figure 59-5 Deep brain stimulation. A pulse generator, surgically implanted in a pouch beneath the clavicle, sends

high-frequency electrical impulses to the thalamus, thereby blocking the nerve pathways associated with tremors in

PD. (Adapted with permission from Smeltzer SC, Bare BG. Textbook of Medical-Surgical Nursing. 9th ed.

Philadelphia, PA: Lippincott Williams & Wilkins; 2000.)

The efficacy of DBS in advanced PD was shown in a two-part study of 255

patients with idiopathic PD responsive to levodopa but with persistent and disabling

motor symptoms.

117 Patients were randomly assigned to DBS or best medical therapy

and followed for 6 months. Patients receiving DBS gained a mean of 4.6 hours/day of

on-time without troubling dyskinesias compared with 0 hours/day for best medical

therapy (P<0.001). Additionally, 71% of patients receiving DBS experienced

clinically meaningful motor function improvements (≥5-point change in UPDRS

motor score) compared with only 32% of patients receiving best medical therapy

(P<0.001).

117

CASE 59-1, QUESTION 19: What surgical therapy would be most appropriate for L.M.?

L.M. appears to be an ideal candidate for DBS. Candidates for DBS should have

idiopathic PD and be levodopa-responsive, but continue to experience motor

complications or tremor despite optimal pharmacotherapeutic regimens. Ideally,

DBS should be avoided in patients with preexisting cognitive or psychiatric

problems owing to a slight risk of decline in cognition. No strict age limitation for

DBS exists, but patients younger than 70 years of age, such as L.M., appear to

recover from surgery more quickly and show greater motor improvements. Deep

brain stimulation of the STN consistently demonstrates marked reduction in the need

for escalating levodopa dosages compared with DBS of the GPi,

118–121 but data

suggest other nonmotor symptoms such as visual processing speed and depression

may be affected more favorably when targeting the GPi.

118

Investigational Pharmacotherapy

CASE 59-2

QUESTION 1: K.B. is a 61-year-old woman who presents to the movement disorders clinic after referral

from her family doctor for a presumptive diagnosis of PD. She is Hoehn and Yahr

2

stage 1, with slightly

decreased arm swing on the left side and unilateral resting hand tremor. Her past medical history is significant

for hypertension and mild renal insufficiency (serum creatinine of 1.4 mg/dL). Since her initial visit with her

family physician, she has been researching information about different PD treatments from several PD-related

websites. Are there any antioxidants, dietary supplements, or other investigational therapies that may benefit

K.B.?

ANTIOXIDANTS

Antioxidants have been hypothesized to benefit patients with PD through their ability

to act as free radical scavengers. The most comprehensive evaluation of antioxidant

therapy for PD comes from the DATATOP study.

109

,

122

,

123

In this study, patients with

early PD were assigned to one of four treatment regimens: α-tocopherol (2,000

international units[IU]/day) and selegiline placebo; selegiline 10 mg/day and αtocopherol placebo; selegiline and α-tocopherol active treatments; or dual placebos.

The primary endpoint was time to requirement of levodopa therapy. After

approximately 14 months of follow-up, no difference was seen between the αtocopherol group and placebo group in time to require levodopa.

123 Thus, despite the

theoretic benefit, clinical data are lacking to support the routine use of α-tocopherol,

and it would not be recommended in K.B.

124

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

TRIPASS XR تري باس

Kana Brax Laberax

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

CELEPHI 200 MG, Gélule

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more