Bendtsen L et al. EFNS guideline on the treatment of tension-type headache—report of an EFNS task force.

Eur J Neurol. 2010;17:1318–1325.

Lyngberg AC et al. Has the prevalence of migraine and tension-type headache changed over a 12-year period?

A Danish population survey. Eur J Epidemiol. 2005;20:243–249.

Bendtsen L, Jensen R. Tension-type headache. Neurol Clin. 2009;27:525.

Fumal A, Schoenen J. Tension-type headache: current research and clinical management. Lancet Neurol.

2008;7:70–83.

Diener HC et al. Use of a fixed combination of acetylsalicylic acid, acetaminophen and caffeine compared with

acetaminophen alone in episodic tension-type headache: meta-analysis of four randomized, double-blind,

placebo-controlled, crossover studies. J Headache Pain. 2014;15:76.

Holroyd KA et al. Management of chronic tension-type headache with tricyclic antidepressant medication, stress

management therapy, and their combination: a randomized controlled trial. JAMA. 2001;285:2208–2215.

Bendtsen L, Jensen R. Mirtazapine in effective in the prophylactic treatment of chronic tension-type headache.

Neurology. 2004;62(10):1706–1711.

Zissis NP et al. A randomized, double-blind, placebo-controlled study of venlafaxine XR in out-patients with

tension-type headache. Cephalalgia. 2007;27(4):315–324.

Wachholtz AB et al. Effect of different meditation types in migraine headache medication use. Behav Med.

2015;11:1–8.

Wells RE et al. Meditation for migraines: a pilot randomized controlled trial. Headache. 2014;54(9):1484–1495.

Day MA et al. Mindfulness-based cognitive therapy for the treatment of headache pain: a pilot study. Clin J

Pain. 2014;30(2):152–161.

Ahn CB et al. A clinical pilot study comparing traditional acupuncture to combined acupuncture for treating

headache, trigeminal neuralgia and retro-auricular pain in facial palsy. J Acupunct Meridian Stud. 2011;4(1):29–

43.

Imitrex tablets (sumatriptan) [prescribing information]. Research Triangle Park, NC: GlaxoSmithKline; March

2012.

Imitrex injection (sumatriptan) [prescribing information]. Research Triangle Park, NC: GlaxoSmithKline;

October 2012.

Imitrex nasal spray (sumatriptan) [prescribing information]. Research Triangle Park, NC: GlaxoSmithKline;

March 2012.

Amerge (naratriptan) [prescribing information]. Research Triangle Park, NC: GlaxoSmithKline; October 2013.

Maxalt, Maxalt-MLT (rizatriptan) [prescribing information]. Whitehouse Station, NJ: Merck and Co; January

2013.

Zomig, Zomig-ZMT (zolmitriptan) [prescribing information]. Wilmington, DE: AstraZeneca Pharmaceuticals;

September 2012.

220.

221.

222.

223.

224.

225.

226.

227.

Axert (almotriptan) [prescribing information]. Titusville, NJ:Janssen Pharmaceuticals; August 2014.

Relpax (eletriptan) [prescribing information]. New York, NY: Pfizer; September 2013.

Frova (frovatriptan) [prescribing information]. Malvern, PA: Endo Pharmaceuticals; October 2013.

Zomig nasal spray (zolmitriptan) [prescribing information]. Wilmington, DE: AstraZeneca Pharmaceuticals;

September 2013.

Alsuma (sumatriptan) [prescribing information]. Columbia, MD: Meridian Medical Technologies; April 2014.

Sumavel DosePro (sumatriptan) [prescribing information]. San Diego, CA: Zogenix; February 2014.

Zecuity (sumatriptan) [prescribing information]. Conshohocken, PA: NuPathe; August 2013.

Tfelt-Hansen P et al. Triptans in migraine: a comparative review of pharmacology, pharmacokinetics and

efficacy. Drugs. 2000;60(6):1267.

p. 1245

Parkinson’s disease (PD) is a chronic, progressive movement disorder

resulting from loss of dopamine from the nigrostriatal tracts in the brain,

and is characterized by rigidity, bradykinesia, postural disturbances, and

tremor.

Case 59-1 (Questions 1, 2)

Treatment for PD is aimed at restoring dopamine supply through one, or

a combination, of the following methods: exogenous dopamine in the

form of a precursor, levodopa; direct stimulation of dopamine receptors

via dopamine agonists; and inhibition of metabolic pathways responsible

for degradation of levodopa.

Case 59-1 (Questions 3–17)

Therapy for PD is usually delayed until there is a significant effect on

quality of life; generally younger patients start with dopamine agonists or

monoamine oxidase type B (MAO-B) inhibitors, whereas older patients

may start with levodopa.

Case 59-1 (Questions 3,4),

Figure 59-2

Initial therapy with levodopa-sparing agents is associated with a lower

risk of developing motor complications than with levodopa, but all

patients will eventually require levodopa.

Case 59-1 (Questions 4–9),

Figure 59-2

Advanced PD is characterized by motor fluctuations including a gradual

decline in on-time, and by the development of troubling dopaminergicinduced dyskinesias. Modified carbidopa/levodopa formulations,

dopamine agonists, MAO-B inhibitors, and catechol-Omethyltransferase (COMT) inhibitors can reduce motor fluctuations;

amantadine can improve dyskinesias. Deep brain stimulation of the

globus pallidus interna or subthalamic nucleus may benefit patients with

advanced PD.

Case 59-1 (Questions

13–19), Figure 59-4

Antioxidants, dietary supplements, and other investigational therapies

have been studied for the management of Parkinson’s disease.

Case 59-2 (Question 1)

Comprehensive therapy for patients should include attention to many

progressive complications of PD, including neuropsychiatric

disturbances and autonomic dysfunction.

Case 59-3 (Questions 1, 2)

RESTLESS LEGS SYNDROME

RLS is a disabling sensorimotor disorder typically marked by irresistible

urge to move the legs (akathisia). It is often associated with

Case 59-4 (Question 1)

uncomfortable paresthesias or dysesthesias and often occurs in the

evening or night.

Several conditions are associated with or can aggravate RLS. Periodic

limb movements of sleep (PLMS) is not the same as RLS but often

coexists.

Case 59-4 (Question 2)

Dopamine agonists are first-line treatments for RLS. They are preferred

because they are longer acting than levodopa, and reduce symptoms

throughout the entire night. Other effective therapies include

carbidopa/levodopa, gabapentin, benzodiazepines, and opiates.

Case 59-4 (Question 3)

A common problem with long-term use of dopaminergic agents in RLS,

particularly levodopa, is an augmentation effect. This refers to a gradual

dosage intensification that occurs in response to a progressive

worsening of symptoms after an initial period of improvement. Gradual

withdrawal of therapy and substitution with other agents should be

performed, rather than continued dopaminergic dose escalation.

Case 59-4 (Question 4)

p. 1246

p. 1247

ESSENTIAL TREMOR

Essential tremor should be distinguished clinically from tremor associated

with PD or other causes.

Case 59-5 (Question 1)

Treatments of choice for essential tremor include propranolol or

primidone. In refractory cases, targeted botulinum toxin A injections can

be useful.

Case 59-5 (Question 2)

PARKINSON’S DISEASE

Incidence, Prevalence, and Epidemiology

PD is a chronic, progressive movement disorder first described in 1817 by Dr.

James Parkinson. Since that time, the term parkinsonism has come to refer to any

disorder associated with two or more features of tremor, rigidity, bradykinesia, or

postural instability.

1 Most cases of PD are of unknown cause and referred to as

idiopathic parkinsonism; however, viral encephalitis, cerebrovascular disease, and

hydrocephalus have symptoms similar to PD as part of their clinical presentation.

1

Unless otherwise stated, all references to PD in this chapter refer to the idiopathic

type.

The age at onset of PD is variable, usually between 50 and 80 years, with a mean

onset of 55 years.

2 Both the incidence and prevalence of PD are age-dependent, with

annual incidence estimates ranging from 10 cases/100,000 (age 50–59 years) to 100

cases/100,000 (age 80–89 years) and an estimated prevalence of 1% of the

population older than 65 years of age.

3

,

4 Men are affected slightly more frequently

than women.

3 Despite the availability of effective symptomatic treatments to improve

both quality of life and life expectancy, no cure exists. The symptoms of PD are

progressive, and within 10 to 20 years, significant immobility results for most

patients.

5 More rapid rates of symptom progression and motor disability have been

observed in patients of older age at disease onset.

6 PD itself does not cause death;

however, patients often succumb to complications related to impaired mobility and

function (e.g., aspiration pneumonia, thromboembolism) and overall frailty.

5

Etiology

The etiology of PD is poorly understood. Most evidence suggests it is multifactorial

and attributable to a complex interplay between age-related changes in the

nigrostriatal tract, underlying genetic risks, and/or environmental triggers. Support

for this hypothesis can be found in several historic observations. Notably, the

postviral parkinsonian symptoms occurring after epidemics of encephalitis in the

early 1900s, and the discovery that ingestion of a meperidine analog, 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine (MPTP), by heroin addicts in northern California

during the early 1980s caused a rapid and irreversible parkinsonism via the

oxidation of free radicals during metabolism by the MAO-B enzyme.

7

For an excellent in-depth discussion of the importance of the MPTP discovery and its influence on PD

research, please view the episode “My Father, My Brother, and Me” from the Public Broadcasting System

program Frontline at http://www.pbs.org/wgbh/pages/frontline/parkinsons/.

The relative contributions of environment and genetics to the occurrence of PD

remain controversial; rural living, pesticide exposure, and consumption of well

water have consistently been associated with increased lifetime risk of PD, whereas

cigarette smoking and caffeine ingestion appear to be protective.

8 Mutations in

several genes, including α-synuclein (SNCA), leucine-rich repeat kinase-2 (LRRK2),

parkin, PTEN- induced kinase-1 (PINK1), and DJ-1, have been observed in rare

familial inherited cases of PD, but these genes lack typical Mendelian patterns of

inheritance, and do not account for the threefold increased risk of developing PD for

individuals who have a first-degree relative affected with sporadic PD.

9 Recent

advances in molecular genetics and genome-wide association studies have revealed

other novel risk genes; however, the exact linkage between genetics, environment,

and clinical expression of disease remains uncertain.

10

Pathophysiology

The salient features of PD result from a loss of dopaminergic neurons in the

nigrostriatal tracts of the brain and development of abnormal intraneuronal protein

aggregates called Lewy bodies that interfere with neuronal function. The nigrostriatal

tracts are neuronal tracts between the substantia nigra and the striatum. They are part

of the extrapyramidal system of the brain involving the basal ganglia. This area is

involved with maintaining posture and muscle tone by regulating voluntary smooth

muscle activity. The pigmented neurons within the basal ganglia have dopaminergic f

ibers, and in PD, these dopamine-producing neurons are progressively depigmented.

The remaining dopaminergic-producing cells have been shown (in postmortem

examination) to contain Lewy bodies.

11 Lewy body pathology appears to ascend the

brain in a predictable manner in PD, beginning in the medulla oblongata in

preclinical stages (which may explain observations of anxiety, depression, and

olfactory disturbance), ascending to the midbrain (motor dysfunction), and spreading

eventually to the cortex (cognitive and behavioral changes).

12 The loss of dopamine

neurons, either from death or from dysfunction, results in loss of dopamine-mediated

inhibition of acetylcholine neurons. In PD, the typical balance of dopamine and

acetylcholine is lost, resulting in a relative increase in cholinergic activity.

The exact pathologic sequence leading to neurodegeneration is unclear, but free

radicals formed as by-products of dopamine auto-oxidation have been implicated.

The finding that a critical threshold of neuronal loss (at least 70%–80%) occurs

before PD becomes clinically apparent suggests that adaptive mechanisms (e.g.,

upregulation of dopamine synthesis or downregulation of synaptic dopamine

reuptake) may somehow influence disease progression during the preclinical stages.

p. 1247

p. 1248

Clinical Presentation of Parkinson’s Disease

CASE 59-1

QUESTION 1: L.M., a 55-year-old, right-handed male artist, presents to the neurology clinic complaining of

difficulty painting because of unsteadiness in his right hand. On questioning, he notes that it is becoming

increasingly difficult to get out of chairs after sitting for a long period because of tightness in his arms and legs.

He also reports having a loss in sense of smell. His medical history is significant for gout (currently requiring no

treatment), constipation, benign prostatic hypertrophy, and aortic stenosis. He does not smoke, but usually drinks

one alcoholic beverage in the evenings. His only prescription medication is citalopram 10 mg by mouth once

daily. On physical examination, L.M. is noted to be a well-developed, well-nourished man who displays a

notable lack of normal changes in facial expression and speaks in a soft, monotone voice. Examination of his

extremities reveals a slight ratchetlike rigidity in both arms and legs, and a mild resting tremor is present in his

right hand. His gait is slow but otherwise normal, with a slightly bent posture. His balance is determined to be

normal, with no retropulsion or loss of righting reflexes after physical threat. His genitourinary examination is

remarkable only for prostatic enlargement. The remainder of his physical examination is within normal limits.

Laboratory values and vitalsigns obtained at this visit include the following:

Blood pressure, 119/66 mm Hg

Heart rate, 71 beats/minute

Sodium, 132 mEq/L

Potassium, 4.4 mEq/L

Blood urea nitrogen, 19 mg/dL

Creatinine, 1.1 mg/dL

Thyroid stimulating hormone, 3.65 microunits/L

Vitamin B12, 612 pg/mL

Folate, 5.2 ng/mL

White blood cells, 4,400 cells/μL

Red blood cells, 5.9 × 10

6

/μL

Hemoglobin, 13.8 g/dL

Hematocrit, 41%

Uric acid, 6.3 mg/dL

How is PD diagnosed? Is neuroimaging or any other testing helpful in establishing the diagnosis of PD? What

signs and symptoms suggestive of PD are present in L.M., and which of these symptoms are among the classic

symptoms for diagnosing PD?

The foundation for establishing the diagnosis of PD remains firmly grounded in

obtaining a careful history and physical examination.

11 The neurologic examination to

assess motor function, along with a positive response to levodopa, is highly

diagnostic. The search for biomarkers of premotor PD in blood, cerebrospinal fluid,

and urine has not uncovered single, practical candidates that are sensitive and

specific.

9 Likewise, although imaging techniques can visualize nigrostriatal nerve

terminals of dopamine synthesis and identify presymptomatic pathology, their use

remains investigational, and routine use in asymptomatic, at-risk individuals is not

yet justified. Other associated premotor symptoms, such as hyposmia (a reduced

ability to smell and detect odors), rapid eye movement (REM) sleep disorder, and

softening and tonal changes of the voice are among the earliest symptoms to appear;

screening for these findings may prove more economically practical in identifying a

high risk population worthy of further diagnostic evaluation.

13 With further scientific

advancements, the diagnosis of PD may rely on clinical, imaging, genetic, and a panel

of laboratory biomarker data. However, by the time patients such as L.M. present

with symptoms, a substantial burden of neuropathologic evidence has accumulated,

and therefore, a diagnosis can be made clinically, without the need for further

laboratory or radiologic testing.

The classic features of PD—tremor, limb rigidity, and bradykinesia—are easily

recognized, particularly in advanced stages of disease. Limb rigidity and

bradykinesia are direct consequences of dopamine loss; by disinhibiting cholinergic

transmission, dopamine loss indirectly causes tremor. It is important to note that not

all classic features are required to make the diagnosis of PD. The presence of two or

more features indicates clinically probable PD.

14 Tremor, which is most often the

first symptom observed in younger patients, is usually unilateral on initial

presentation. Frequently, the low-amplitude tremor is of a pill-rolling type involving

the thumb and index finger (3–6 Hz); it is present at rest, worsens under fatigue or

stress, and is absent with purposeful movement or when asleep.

1 These features help

distinguish it from essential tremor, which usually manifests as a symmetric tremor in

the hands, often accompanied by head and voice tremor.

11 Approximately 20% of

patients with PD do not present with tremor.

11 Muscular rigidity resulting from

increased muscle tone often manifests as a cogwheel or ratchet (catch-release) type

of motion when an extremity is moved passively.

1 Rigidity may also be experienced

as stiffness or vague aching or limb discomfort.

11 Bradykinesia refers to an overall

slowness in initiating movement. Early in the disease, patients may describe this as

weakness or clumsiness of a hand or leg.

11 Because the disease progresses, difficulty

initiating and terminating steps results in a hurried or festinating gait; the posture

becomes stooped (simian posture), and postural reflexes are impaired.

1 Symptoms

that were unilateral on initial presentation progress asymmetrically and often become

bilateral and more severe with disease progression.

1 Patients with PD may develop

masked facies, or a blank stare with reduced eye blinking (Figure 59-1).

Figure 59-1 Clinical features of Parkinson’s disease. (Reprinted with permission from Rosdahl CB. Book of

Basic Nursing. 7th ed. Philadelphia, PA: Lippincott-Raven; 1999:1063.)

p. 1248

p. 1249

Because the diagnosis of PD is clinical, misdiagnosis can occur, leading to

inappropriate, ineffective, or delayed treatment. Several conditions can be mistaken

for PD and are important to distinguish from PD because they respond poorly to

dopaminergic medications and are associated with worse prognosis.

11 Falls or

dementia early in the disease, symmetric parkinsonism, wide-based gait, abnormal

eye movements, marked orthostatic hypotension, urinary retention, or marked

disability within 5 years after the onset of symptoms suggests alternative diagnoses to

PD.

11 Drugs may also mimic idiopathic PD. Drugs that act as antagonists at

dopaminergic D2

receptors (e.g., neuroleptics, prochlorperazine, and

metoclopramide), and others such as valproate, amiodarone, phenytoin, and lithium

may cause a state of drug-induced parkinsonism. This should be excluded before the

diagnosis of PD is established. Although generally reversible, symptoms may persist

for weeks or months after discontinuation of the offending agent.

11

L.M. initially presents with classic premotor features such as his soft, monotone

voice and reduced sense of smell, in addition to classic symptoms of PD. A

noticeable unilateral resting tremor is present along with decreased manual dexterity,

as evidenced by his difficulty handling a paintbrush. Handwriting abnormalities

occur frequently, particularly micrographia, a symptom of bradykinesia. Because

L.M. is an artist, this abnormality would be particularly troublesome. Rigidity

(ratcheting of the arms) and a mask-like facial expression also are present. Although

he has a partially stooped posture, it is difficult to attribute this entirely to the disease

because postural changes commonly occur with advancing age and, on physical

examination, his balance was normal. To confirm the diagnosis of PD, a therapeutic

trial of medication (levodopa) may be considered. A positive response to levodopa,

as evidenced by an improvement in motor function, suggests the diagnosis of PD.

However, patients with the tremor-predominant form of the disease may not respond

to levodopa, especially in the early stages of the disease.

11

Staging of Parkinson’s Disease

CASE 59-1, QUESTION 2: What are the stages of PD? In what stage of the disease is L.M.?

To assess the degree of disability and determine the rate of disease progression

relative to treatment, various scales have been developed. The most common of these

is the Hoehn and Yahr scale ( Table 59-1).

2

In general, patients in Hoehn and Yahr

2

stage 1 or 2 of PD have mild disease that does not interfere with activities of daily

living (ADLs) or work and usually requires minimal or no treatment. In stage 3

disease, daily activities are restricted and employment may be significantly affected

unless effective treatment is initiated. According to the scale, L.M. appears to be in

late stage 2, early stage 3 of the disease.

With advanced-stage disease (3, 4), most patients require a double or triple drug

therapy strategy. Patients with end-stage disease (stage 5) are severely incapacitated

and, because of advanced disease progression, often do not respond well to drug

therapy.

Treatment of Parkinson’s Disease

OVERVIEW OF THERAPY

Numerous national and international guidelines exist regarding the management of

motor and nonmotor symptoms of PD. These guidelines include the American

Academy of Neurology (AAN) Practice Parameters developed in 2002 and 2006,

15

,

16

the United Kingdom National Institute for Health and Care Excellence (NICE)

National Guideline developed in 2006 and revised in July 2017,

17 and the European

Federation of Neurological Societies Movement Disorder Society-European Section

(EFNS MDS-ES) Guidelines developed in 2013

18

; all are cited throughout the text.

Table 59-1

Staging of Disability in Parkinson’s Disease (PD)

Stage 1 Unilateral involvement only; minimal or no functional impairment

Stage 2 Bilateral involvement, without impairment of balance

Stage 3 Evidence of postural imbalance; some restriction in activities; capable of leading independent life;

mild-to-moderate disability

Stage 4 Severely disabled, cannot walk and stand unassisted; significantly incapacitated

Stage 5 Restricted to bed or wheelchair unless aided

Although most of this chapter is devoted to the drug therapy of PD, the importance

of nonpharmacologic, supportive care cannot be overemphasized. Exercise, physical

and occupational therapy, and good nutritional support can be beneficial at the earlier

stages to improve mobility, increase strength, and enhance well-being and mood.

17

Psychological support is often necessary in dealing with depression and other related

problems. Newly diagnosed patients and their family members need to be educated

about what to expect from the disease and the various forms of treatment available.

The support of family members is vital in establishing an overall effective

therapeutic plan.

Nonpharmacologic therapy should continue throughout the course of care;

however, due to the progressive nature of PD, pharmacologic therapy is often

necessary. Because the salient pathophysiologic feature of PD is the progressive loss

of dopamine from the nigrostriatal tracts in the brain, drug therapy for the disease is

aimed primarily at replenishing the supply of dopamine. This is accomplished

through one, or a combination, of the following methods: (a) administering exogenous

dopamine in the form of a precursor, levodopa; (b) stimulating dopamine receptors

within the striatum through the use of dopamine agonists (e.g., pramipexole,

ropinirole); or (c) inhibiting the major metabolic pathways within the brain that are

responsible for the degradation of levodopa and its metabolites. This latter effect is

achieved through the use of aromatic L-amino acid decarboxylase (AAD) inhibitors

(e.g., carbidopa), COMT inhibitors (e.g., entacapone), or MAO-B inhibitors (e.g.,

selegiline, rasagiline). Anticholinergics are also used; however, they are solely

efficacious for the cholinergic-mediated tremor, and their routine use is limited by

central nervous system (CNS) adverse effects, particularly in older patients. A

unique agent, amantadine, is also used occasionally and may provide modest benefits

via both dopaminergic and nondopaminergic (inhibition of glutamate) mechanisms

(Table 59-2). Because PD progresses, additional options, such as surgery, may be

appropriate for a subset of patients.

Despite optimization of both pharmacologic and nonpharmacologic therapies in

PD, physical disability is progressive and unavoidable. In many instances, adverse

effects of the medications themselves can lead to additional problems. These include

neuropsychiatric problems (e.g., cognitive impairment and dementia, hallucinations

and delirium, depression, agitation, anxiety), autonomic dysfunction (e.g.,

constipation, urinary problems, sexual problems, orthostasis, thermoregulatory

imbalances), falls, sleep disorders (e.g., insomnia or sleep fragmentation,

nightmares, RLS), and motor complications (e.g., disabling periods of either too

much [dyskinetic] or too little [akinetic] dopaminergic activity). In general, the more

efficacious medications are also those with the greatest risk of serious side effects

and motor complications.

p. 1249

p. 1250

Table 59-2

Medications Used for the Treatment of Parkinson’s Disease (PD)

Generic (Trade)

Name Dosage Unit Titration Schedule Usual Daily Dose Adverse Effects

Amantadine

(Symmetrel)

100-mg capsule and

tablet

Liquid: 50 mg/5 mL

100 mg every day;

increased by 100 mg

every 1–2 weeks

100–300 mg Orthostatic

hypotension,

insomnia,

depression,

hallucinations, livedo

reticularis,

xerostomia

Anticholinergic Agents

Benztropine

(Cogentin)

0.5-, 1-, and 2-mg

tablets

Injection: 1 mg/mL

0.5 mg every day

increased by 0.5 mg

every 5–6 days

1–3 mg given every

day to BID

Constipation,

xerostomia, dry skin,

dysphagia,

confusion, memory

impairment

Trihexyphenidyl

(Artane)

2- and 5-mg tablets

Liquid: 2 mg/5 mL

1–2 mg/day

increased by 1–2

mg every 3–5 days

6–15 mg divided

TID to QID

Constipation,

xerostomia, dry skin,

dysphagia,

confusion, memory

impairment

Combination Agents

Carbidopa–levodopa

(immediate

release)/entacapone

(Stalevo)

12.5-/50-/200-,

18.75-/75-/200-, 25-

/100-/200-, 31.25-

/125-/200-, 37.5-

/150-/200-, 50-/200-

/200-mg tablets

Titrate with

individual dosage

forms

(carbidopa/levodopa

and entacapone)

first, then switch to

combination tablet

Varies (see

individual drugs)

See individual drugs

Dopamine Replacement

Carbidopa–levodopa

(regular) (Sinemet)

10-/100-, 25-/100-,

and 25-/250-mg

tablets

25/100 mg BID,

increased by 25/100

mg weekly to effect

and as tolerated

30/300 to 150/1,500

mg divided TID to

QID

Nausea, orthostatic

hypotension,

confusion, dizziness,

hallucinations,

dyskinesias,

blepharospasm

Carbidopa–levodopa

(CR) (Sinemet CR)

25-/100- and 50-

/200-mg tablets

25/100 mg BID

(spaced at least 6

hours apart),

increased every 3–7

days

50/200 to 500/2,000

divided QID

Same as regular

Sinemet

Carbidopa–levodopa

(ER) (Rytary)

23.75-/95-, 36.25-

/145-, 48.75-/195-,

61.25-/245-mg

capsules

23.75/95 mg TID;

may increase to

36.25/145 mg TID

on day 4 and titrate

to response

Variable Same as regular

Sinemet

Carbidopa–levodopa

(enteralsuspension)

(Duopa)

4.63-/20-mg/mL in

100-mL cassette

Total daily dose

administered over

16 hours

Variable Same as regular

Sinemet

Carbidopa–levodopa

ODT (Parcopa)

10-/100-, 25-/100-,

and 25-/250-mg

tablets

25/100 TID,

increased every 1–2

days; if transferring

from regular

levodopa <1,500

mg/day, start 25/100

mg TID to QID

(start 25/250 mg

TID to QID if

already on >1,500

mg/day of regular

levodopa)

25/100 to 200/2,000

divided TID to QID

Same as regular

Sinemet; may occur

more rapidly than

with regular

Sinemet

Dopamine Agonists

Bromocriptine

(Parlodel)

2.5-mg tablet, 5-mg

capsule

1.25 mg BID, titrate

slowly as tolerated

(2.5 mg/day every

2-4 weeks)

10–40 mg divided

TID; Max 100

mg/day

Orthostatic

hypotension,

confusion, dizziness,

hallucinations,

nausea, muscle

cramps;

retroperitoneal,

pleural, pericardial

fibrosis; cardiac

valve thickening

Pramipexole

(Mirapex, Mirapex

Immediate release:

0.125-, 0.25-, 0.50-,

Immediate release:

0.375 mg divided

Immediate release:

1.5–4.5 mg divided

Orthostatic

hypotension,

ER) 0.75-, 1-, 1.5-mg

tablets

ER: 0.375-, 0.75-,

1.5-, 2.25-, 3-, 3.75-,

4.5-mg tablets

TID; titrate weekly

by 0.125–0.25

mg/dose

ER: 0.375 mg once

daily; titrate weekly

by 0.75 mg/dose

TID

ER: 1.5–4.5 mg

once daily

confusion, dizziness,

hallucinations,

nausea, somnolence

Ropinirole (Requip,

Requip XL)

0.25-, 0.5-, 1-, 2-, 3-,

4-, 5-mg tablet

XL: 2-, 4-, 6-, 8-, 12-

mg tablets

0.25 mg TID; titrate

weekly by 0.25

mg/dose

XL: 2 mg once

daily; titrate weekly

by 2 mg/day

3–12 mg divided

TID

XL: 3–12 mg once

daily

Orthostatic

hypotension,

confusion, dizziness,

hallucinations,

nausea, somnolence

p. 1250

p. 1251

Apomorphine

(Apokyn)

10 mg/mL injection Initial 2-mg

subcutaneous test

dose; begin with 2

mg; increase by 1

mg every few days

2–6 mg TID Orthostatic

hypotension,

drowsiness,

yawning, injection

site reactions,

nausea, vomiting

(administer with

trimethobenzamide,

not 5-

hydroxytryptamine3 [5-HT3

]

antagonists)

Rotigotine (Neupro) 1,-, 2-, 3-, 4-, 6-, 8-

mg/24 hour

transdermal delivery

system

Early stage: 2 mg/24

hour; Advanced

stage: 4 mg/24 hour;

titrate weekly by 2

mg/24 hour;

Application site

should be rotated

daily between

abdomen, thigh, hip,

flank, shoulder, or

upper arm and do

not use same site

within 14 days

4–6 mg/24 hour Hallucinations,

nausea, vomiting,

anorexia,

somnolence,

insomnia, dizziness,

hyperhidrosis, visual

disturbance,

peripheral edema,

and application site

reactions; avoid in

patients with known

sulfite sensitivity

COMT Inhibitors

Entacapone

(Comtan)

200-mg tablet 200 mg with each

administration of

carbidopa/levodopa,

up to 8 tablets daily

3–8 tablets daily Diarrhea,

dyskinesias,

abdominal pain,

urine discoloration

Tolcapone (Tasmar) 100-mg tablet 100 mg TID 300–600 mg divided

TID

Diarrhea,

dyskinesias,

abdominal pain,

urine discoloration,

hepatotoxicity

Monoamine Oxidase Type B (MAO-B) Inhibitors

Selegiline

(Eldepryl)

a

5-mg tablet, capsule 5 mg every morning;

may increase to 5

mg BID (5 mg with

breakfast and 5 mg

with lunch)

5–10 mg/day Insomnia, dizziness,

nausea, vomiting,

xerostomia,

dyskinesias, mood

changes; use

caution when

coadministered with

sympathomimetics

or serotoninergic

agents (increased

risk of serotonin

syndrome); avoid

tyramine-containing

foods

Selegiline ODT

(Zelapar)

1.25-mg tablet 1.25 mg every day;

avoid food or liquids

for 5 minutes before

and after

administration; may

increase to 2.5 mg

every day after 6

weeks

1.25–2.5 mg every

day

Insomnia, dizziness,

nausea, vomiting,

xerostomia,

dyskinesias, mood

changes; use

caution when

coadministered with

sympathomimetics

or serotoninergic

agents (increased

risk of serotonin

syndrome); avoid

tyramine-containing

foods

Rasagiline (Azilect) 0.5-, 1-mg tablets 0.5–1 mg once daily 0.5–1 mg/day Similar to selegiline

Safinamide

(Xadago)

b

50-, 100 mg tablets 50 mg once daily;

dose may be

increased after two

weeks to 100 mg

once daily

50-100 mg/day Similar to selegiline

aA transdermal formulation is also available, but not approved for use in PD.

bApproved only for use as adjunctive treatment to levodopa/carbidopa in patients experiencing “off” episodes.

BID, twice daily; COMT, catechol-O-methyltransferase; MAO-B, monoamine oxidase type B; ODT, orally

disintegrating tablet; QID, four times daily; TID, three times daily.

Treatment of Early Parkinson’s Disease

CASE 59-1, QUESTION 3: When should L.M. begin treatment for his PD?

p. 1251

p. 1252

In choosing when to treat the symptoms of PD and which therapy to use, care must

be taken to approach each patient individually. Although no consensus has been

reached about when to initiate symptomatic treatment, most healthcare professionals

agree that treatment should begin when the patient begins to experience functional

impairment as defined by (a) threat to employment status, (b) symptoms affecting the

dominant side of the body, or (c) bradykinesia or rigidity.

19

,

20

Individual patient

preferences also should be considered. Judging by the symptoms L.M. is displaying,

he would likely benefit from immediate treatment. His symptoms are unilateral but

are occurring on his dominant side and are interfering with his ability to paint, thus

affecting his livelihood. He is also showing signs of rigidity and bradykinesia but can

otherwise live independently. An algorithm for the management of patients with early

PD is presented in Figure 59-2. The long-term, individualized treatment plan is

usually characterized by frequent dosage adjustments because of the chronic and

progressive nature of the disease.

CASE 59-1, QUESTION 4: The decision is made to begin drug therapy for L.M. Should therapy be initiated

with a levodopa or levodopa-sparing therapy?

Despite advances in pharmacologic treatment options for PD, no therapy has been

proven to be disease-modifying or neuroprotective. Therapy continues to be

symptomatic, and levodopa remains the most effective antiparkinsonian agent.

11

However, the question of when to begin levodopa in the treatment of PD has been

historically debated. With long-term use, the efficacy of levodopa decreases and risk

for the development of motor fluctuations and dyskinesias increases. Because

escalating doses of levodopa are accompanied by a high frequency of undesirable

side effects, other methods of enriching dopamine supply have been developed. Two

such methods include the use of dopamine agonists and MAO-B inhibitors. Both of

these modalities have proven efficacy early in the disease.

17

Dopamine agonists, which bind directly to dopamine receptors, do not require

metabolic conversion to an active product and therefore act independently of

degenerating dopaminergic neurons. In clinical trials comparing dopamine agonists

with levodopa, ADLs and motor features are improved to a greater degree with

levodopa compared with dopamine agonists.

18 Although they are not as effective as

levodopa, the dopamine agonists have potential advantages. Unlike levodopa,

circulating plasma amino acids do not compete with dopamine agonists for

absorption and transport into the brain, eliminating administration constraints.

Dopamine agonists have a longer half-life than levodopa formulations, reducing the

need for multiple daily dosing. As a class, dopamine agonists provide adequate

control of symptoms when given as monotherapy in up to 80% of patients with earlystage disease.

21

Short- and long-term comparisons of patients with early disease started on either

levodopa or dopamine agonists have varying results. In trials extending 4 to 5 years,

therapy with dopamine agonists is associated with fewer motor complications such

as dyskinesias, delayed time to dyskinesias, and delayed need for initiation of

dopaminergic therapy.

22–24 Against levodopa as initial therapy, early trials of the

dopamine agonists appear to delay the onset of dyskinesias. In a randomized,

controlled trial evaluating the development of motor complications with levodopa or

the dopamine agonist pramipexole, 301 untreated patients with early PD were

randomly assigned to receive either pramipexole 0.5 mg 3 times daily or

carbidopa/levodopa 25/100 mg 3 times daily.

24 Patients in both arms could be

prescribed open-label levodopa as needed for disability during the maintenance

phase of the study. After a mean follow-up of 24 months, fewer pramipexole-treated

patients reached the primary endpoint of time to the first occurrence of wearing-off,

dyskinesias, or on–off motor fluctuations (28% vs. 51%; P<0.001) than the patients

initially randomly assigned to levodopa therapy. Additionally, patients in the

pramipexole group were receiving lower daily doses of levodopa, theoretically

conferring a reduced risk of developing motor complications. Long-term follow-up

of this cohort (mean = 6 years) revealed a persistently lower rate of dopaminergic

motor complications in the pramipexole-treated patients compared with those

receiving levodopa (50% vs. 68.4%, respectively; P = 0.002).

25

Figure 59-2 Suggested treatment algorithm for the management of early PD.

p. 1252

p. 1253

Despite the continued increased risk of motor complications in patients started on

levodopa, trials extending 10 to 15 years found no significant difference in disease

severity rating or rates of disabling dyskinesias.

26

,

27 Patients treated initially with

ropinirole were less likely to experience dyskinesias compared with those treated

initially with levodopa at the end of a 5-year evaluation.

22 Patients in the ropinirole

group used lower mean daily doses of levodopa (427 mg vs. 753 mg) but were

almost twice as likely to need open-label levodopa supplementation (66% vs. 36%).

Dyskinesias developed in 20% of the ropinirole-treated patients compared with 45%

of the levodopa-treated patients. For ropinirole-treated patients who were able to

remain on monotherapy without open-label levodopa supplementation, only 5%

experienced dyskinesia, compared with 36% of those receiving levodopa

monotherapy. Although the lower incidence of dyskinesias was shown to persist in a

long-term open-label follow-up of this study cohort, there was no difference in

disease severity between the ropinirole-treated group and the levodopa-treated group

at 10 years.

26 There may be additional trends toward better cognitive and healthrelated quality of life outcomes in the patients started on levodopa.

27 This may be due

in part to the fact that although dopamine agonists do delay motor complications, they

are not without side effects. Trials have consistently shown higher rates of adverse

drug reactions in patients initially started on dopamine agonists.

22

,

25 One open-label,

randomized trial found that 28% of patients discontinued initial therapy with

dopamine agonists due to side effects versus only 2% of patients initially given

levodopa.

28 Regardless of initial therapy chosen, due to disease progression,

levodopa therapy will eventually be required, motor complications will develop, and

disability will be present.

MAO-B inhibitors are an alternative method of enriching dopamine supply in early

disease. These agents increase nigrostriatal dopamine supply via the irreversible

inhibition of MAO-B, a major enzymatic pathway responsible for the metabolism of

dopamine in the brain, and require the presence of dopamine for clinical effect.

29

Though MAO-B inhibitors have been shown to delay the need for dopaminergic

therapy with levodopa, definitive efficacy comparisons are lacking.

29

Early guidance from the AAN supported either dopamine agonists or levodopa as

initial therapy for PD; more recent NICE and EFNS MDS-ES guidelines additionally

support the use of MAO-B inhibitors in initial therapy.

15

,

17

,

18 The long-term

effectiveness of dopamine agonists and MAO-B inhibitors compared with levodopa

as initial treatment for PD (PD MED) trial set out to compare and contrast levodopa

versus levodopa-sparing therapies.

28 Patients assigned to monotherapy with MAO-B

inhibitors or dopamine agonists were generally younger and healthier than those

assigned levodopa as initial therapy. At 7-year follow-up, 72% of patients initially

assigned to MAO-B inhibitors withdrew from initial therapy and changed treatment

compared to 50% in the dopamine agonist group and 7% in the levodopa group

(P<0.0001). Among those patients initiated on MOA-B inhibitors, 48% withdrew

due to side effects and 36% withdrew due to lack of efficacy, and for patients

initiated on dopamine agonists, 82% withdrew due to side effects and 16% withdrew

due to lack of efficacy.

28 As shown in prior studies, levodopa improved function and

quality of life to a greater degree; however, the primary mobility outcome did not

reach the prespecified minimally important difference. Mobility scores were not

significantly different between MAO-B inhibitors and dopamine agonists indicating

that MAO-B inhibitors are at least as effective as dopamine agonists in early therapy

of PD.

28 Additionally, at 7 years, patients on dopamine agonists were on the highest

overall amount of medication. The study confirms that initial treatment with a

dopamine agonist, MAO-B inhibitor, or levodopa may all be reasonable approaches.

Disease severity, degree of functional impairment, life expectancy, and age guide

therapeutic drug selection in early PD. In younger patients (e.g., age <65 years) with

milder disease, such as L.M., the initiation of levodopa-sparing therapy with either a

dopamine agonist or MAO-B inhibitor would be appropriate. Initiating levodopa

therapy later in the course of PD delays the development of motor complications,

particularly the troubling peak-dose, levodopa-induced dyskinesias, which

eventually develop with advancing PD. Patients with younger age at disease onset,

such as L.M., may be at an increased lifetime risk of developing dyskinesias.

19

Although the PD MED trial did conclude that MAO-B inhibitors are at least as

effective as dopamine agonists for initial therapy, other meta-analyses show that the

degree of symptomatic benefit over placebo is small in comparison with those of

dopamine agonists over placebo.

30 Overall, there is a lack of comparative data

between MAO-B inhibitors and dopamine agonists. Therefore, use of MAO-B

inhibitors in initial therapy is often limited to young patients with more mild

functional impairments. In older patients (e.g., age >65 years), those with more

significant functional impairments, or those with limited life expectancy, use of

levodopa may be warranted, as a result of the large number of symptomatic benefits

associated with its use despite the relatively high risk of motor complications and

side effects. Additionally, levodopa may be more appropriate initial treatment in

older patients, because they may be more likely to experience intolerable CNS side

effects from dopamine agonists and less likely to develop motor complications over

their lifetime.

19

In the case of L.M., his relatively young age, mild disease, and moderate functional

impairments make him a good candidate for initial therapy with a dopamine agonist.

L.M. will require levodopa therapy at a later time, when he reaches more advanced

stages of the disease. By initiating therapy first with a dopamine agonist, rescue

levodopa therapy can likely be started at smaller doses, and the onset of motor

complications that often occur with escalating doses and extended therapy with

levodopa may be delayed.

DOPAMINE AGONISTS

CASE 59-1, QUESTION 5: L.M. is to be started on a dopamine agonist. Which agent should be selected?

p. 1253

p. 1254

Table 59-3

Pharmacologic and Pharmacokinetic Properties of Dopamine Agonists

Bromocriptine Pramipexole Ropinirole Apomorphine Rotigotine

Type of

compound

Ergot derivative Nonergoline Nonergoline Nonergoline Nonergoline

Receptor

specificity

D2

, D1

,

a α1

, α2

,

5-HT

D2

, D3

, D4

, α2 D2

, D3

, D4 D1

, D2

, D3

, D4

,

D5

, α1

, α2

, 5-

HT1

, 5-HT2

D1

, D2

, D3

, 5-

HT1

Bioavailability 7% (first-pass

metabolism)

90% 55% (first-pass

metabolism)

<5% orally;

100%

subcutaneous

NA

Tmax

(minutes) 70–100 60–180 90 10–60 15–18 (hours);

no

characteristic

peak observed

Protein binding 90%–96% 15% 10%–40% >99.9% 89.5%

Elimination route Hepatic Renal, 90%

unchanged

Hepatic Hepatic and

extrahepatic

Hepatic

Half-life (hours) 2–8 8–12 6 0.5–1 3–7

aAntagonist.

5-HT, serotonin; NA, not applicable.

Two generations of dopamine agonists have been used for the treatment of

idiopathic PD, in early-stage PD as monotherapy, or as an adjunct to levodopa in

patients with advanced disease. The comparative pharmacologic and

pharmacokinetic properties of these agents are shown in Table 59-3. The firstgeneration dopamine agonists, which are derived from ergot alkaloids, include

bromocriptine, pergolide, and cabergoline. These older agents are now rarely used

because of increased risk of retroperitoneal, pleural, and pericardial f ibrosis, as

well as a two- to fourfold increased risk for cardiac valve f ibrosis when compared

with nonergoline dopamine agonists and controls.

31

,

32 Pergolide was voluntarily

withdrawn in the United States (US) in 2007 for this reason, and although

cabergoline continues to be used in Europe, it is only indicated in the United States

for the treatment of hyperprolactinemia. Pramipexole, ropinirole, apomorphine, and

rotigotine are second-generation nonergoline dopamine agonists. Of these agents,

pramipexole and ropinirole are commonly prescribed. Apomorphine is available

only in injectable form, for use as a rescue agent in the treatment of hypomobility

“off” episodes in patients with PD. Rotigotine, a once-daily transdermal formulation,

was recently re-introduced to the market.

Dopamine agonists work by directly stimulating postsynaptic dopamine receptors

within the striatum. The two families of dopamine receptors are D1 and D2

. The D1

-

like receptor family includes the D1 and D5 dopamine subtype receptors and the D2

-

like receptor family includes D2

, D3

, and D4 dopamine subtype receptors. Stimulation

of D2

receptors is largely responsible for reducing rigidity and bradykinesia,

whereas the precise role of the D1

receptors remains uncertain.

33 Pramipexole and

ropinirole exhibit selectivity for D2

receptors without any significant affinity for D1

receptors whereas rotigotine has activity at all dopamine receptors.

34–36 Although the

dopamine agonists differ slightly from each other in terms of their affinities for

dopamine receptor subtypes, these agents produce similar clinical effects when used

to treat PD, and no compelling evidence favors one agent over another strictly on

efficacy measures. Instead, experience with the nonergoline dopamine agonists,

specifically pramipexole and ropinirole, makes them currently preferred as initial

dopamine agonists. Thus, either agent would be acceptable as initial therapy in L.M.

CASE 59-1, QUESTION 6: The decision is made to begin pramipexole in L.M. How effective is pramipexole

in the initial treatment of PD? How does ropinirole compare?

Pramipexole

Pramipexole has been well studied as monotherapy in patients with early-stage PD

and as an adjunct to levodopa therapy in advanced-stage disease.

34–36 These trials

were multicenter, placebo-controlled, parallel-group studies, and parts of the Unified

Parkinson’s Disease Rating Scale (UPDRS) were used as the primary outcome

measures, specifically improvement in ADLs (Part II) and motor function scores

(Part III). Each evaluation on the UPDRS is rated on a scale of 0 (normal) to 4 (can

barely perform). Lower scores on the UPDRS after treatment indicate an

improvement in overall performance.

To view the UPDRS and other assessment tools for the symptoms of Parkinson’s disease, please visit

http://www.movementdisorders.org/MDS/Education/Rating-Scales.htm.

The evidence for the efficacy of pramipexole in early PD comes from two largescale, double-blind, placebo-controlled studies that included a total of 599 patients

with early-stage PD (mean disease duration of 2 years).

35

,

36

In the first study, 264

patients were randomly assigned to receive one of four fixed doses (1.5, 3, 4.5, or 6

mg/day) or placebo.

35 The pramipexole-treated patients had a 20% reduction in their

total UPDRS scores compared with baseline values, whereas no significant

improvement was observed in the placebo-treated patients. A trend toward

decreased tolerability was noted because the pramipexole dosage was escalated,

especially in the 6 mg/day group. A second study of 335 patients titrated doses up to

the maximal tolerated dose (not to exceed 4.5 mg/day) and then followed patients for

a 6-month maintenance phase.

36 The mean pramipexole maintenance dosage was 3.8

mg/day. Those treated with pramipexole experienced significant improvements in

both the ADL scores (22%–29%; P<0.0001) and motor scores (25%–31%;

P<0.0001), whereas there were no significant changes in the placebo group.

Ropinirole

Similar to pramipexole, ropinirole is a synthetic nonergoline dopamine agonist.

Although the drug is pharmacologically similar to pramipexole, it has some distinct

pharmacokinetic properties (Table 59-3). Unlike pramipexole, which is primarily

eliminated by renal excretion, ropinirole is hepatically metabolized and undergoes

significant first-pass hepatic metabolism.

37 Similar to

p. 1254

p. 1255

pramipexole, ropinirole is approved for use as monotherapy in early-stage idiopathic

PD and as an adjunct to levodopa therapy in patients with advanced-stage disease.

Ropinirole has not been directly compared with pramipexole in randomized,

double-blind comparisons, but it appears to have comparable efficacy in indirect

comparison. In several randomized, double-blind, multicenter, parallel-group studies

comparing ropinirole with placebo, bromocriptine, or levodopa, 6 months of

monotherapy with ropinirole in patients with early PD significantly improves

UPDRS motor scores by approximately 20% to 30% compared with baseline

values.

38–40

Rotigotine

Rotigotine is a nonergoline dopamine receptor agonist that is formulated in a

transdermal patch delivery system designed for once-a-day application. It was

voluntarily withdrawn from the U.S. market in 2008 because of problems with crystal

formation in the patches; however, the patch was reformulated and re-approved by

the Food and Drug Administration (FDA) in April 2012. The original and

reformulated patches have demonstrated efficacy as monotherapy in early-stage

PD,

41–44 whereas only the original formulation has demonstrated efficacy as

adjunctive therapy to levodopa in patients with advanced stages of PD.

45

,

46

It is,

however, used in both situations.

A randomized, controlled trial conducted in Japanese patients with early-stage PD

found that the mean improvement in UPDRS Part II and III sum scores was 8.4 in the

rotigotine-treated group versus 4.1 in the placebo group (P = 0.002; 95% CI, –7.0 to

–1.7).

47 The average dose of rotigotine was 12.8 mg/24 hours in this study which

exceeds the highest recommended dose for early PD (6 mg/24 hours). Despite clear

benefits in improving ADLs and motor function, there is insufficient evidence at this

time to support that rotigotine prevents or delays motor fluctuations or dyskinesias.

48

The transdermal delivery may have several advantages over traditional oral

preparations including possible increase in adherence, ease of use in patients with

swallowing difficulty, and a more continuous stimulation of dopamine receptors. In

theory, these benefits may translate into improved efficacy. In randomized, controlled

trials comparing rotigotine to ropinirole as monotherapy or pramipexole as adjunct

therapy, rotigotine has been found to be noninferior when typical clinical doses are

used.

44

,

46

Dosing

CASE 59-1, QUESTION 7: How are pramipexole and ropinirole dosed? What is the appropriate dose of

pramipexole for L.M.?

Dopamine agonists should always be initiated at a low dosage and gradually

titrated to the maximal effective dose, as tolerated. This approach minimizes adverse

effects that may result in nonadherence or discontinuation of the drug. In clinical

trials, the maximal effective doses are variable and correlate with disease severity

and tolerability. Studies have found no difference in efficacy or safety measures

between immediate and long-acting formulations of dopamine agonists.

49 One fixeddose study of pramipexole in early PD showed that most patients responded

maximally at a dosage of 0.5 mg 3 times daily, although it has been shown to be

effective and well tolerated in doses as high as 4.5 mg/day (divided).

34

L.M. has normal renal function; therefore, immediate-release pramipexole should

be started at an initial dosage of 0.125 mg 3 times daily for 5 to 7 days. His dosage

may be increased weekly by 0.125 to 0.25 mg/dose as tolerated and up to the

maximal effective dose, not to exceed the maximum dose of 4.5 mg/day.

34 Patients

with a creatinine clearance of less than 50 mL/minute should be dosed less frequently

than those with normal renal function.

34 Patients with a creatinine clearance of 30 to

50 mL/minute should receive a starting dose of 0.125 mg twice daily up to a maximal

dose of 0.75 mg 3 times daily; patients with a creatinine clearance of 15 to 30

mL/minute should receive a starting dose of 0.125 mg daily up to a maximal dose of

1.5 mg daily. Pramipexole has not been studied in patients with a creatinine

clearance of less than 15 mL/minute or those receiving hemodialysis. A once-daily

extended-release formulation of pramipexole is also available; patients can be

switched overnight from immediate-release pramipexole at the same daily dose. The

use of the extended-release formulation is not recommended for patients with a

creatinine clearance less than 30 mL/minute or for those receiving hemodialysis.

Ropinirole should be initiated at a dosage of 0.25 mg 3 times daily with gradual

titration to clinical response in weekly increments of 0.25 mg/dose over the course of

4 to 6 weeks, not to exceed 24 mg/day.

37 Patients wishing to take the drug less

frequently can be switched directly to an extended-release once-daily formulation,

selecting the dose that most closely matches the total daily dose of the immediaterelease formulation. No dose adjustments for ropinirole are necessary in patients

with renal dysfunction and can be used in doses up to 18 mg/day in patients on

hemodialysis

Transdermal rotigotine is available in several patch strengths (Table 59-2). For

patients with early-stage PD, it is recommended to start rotigotine at 2 mg/24 hours

with dose increases of 2 mg/24 hours no sooner than weekly based on clinical

response and tolerability.

50 The maximum recommended dose in early PD is 6 mg/24

hours whereas 8 mg/24 hours is recommended in advanced PD. Clinical trials have

often used doses up to 16 mg/24 hours.

46

,

51 The time to reach steady state plasma

concentrations of rotigotine is approximately 2 to 3 days after initial patch

application.

50 A multinational, open-label study demonstrated that patients with

advanced PD taking pramipexole <2 mg/day or ropinirole <9 mg/day can be safely

switched directly to rotigotine with no cross-titration.

52 Rotigotine is metabolized via

conjugation and N-dealkylation, and the inactive conjugates are excreted in the urine.

No adjustments are required for patients with hepatic or renal insufficiency.

Adverse Effects

CASE 59-1, QUESTION 8: What are the adverse effects of pramipexole and ropinirole? How can these be

managed?

Because pramipexole, ropinirole, and rotigotine are all approved for use as

monotherapy in early-stage disease and as adjunctive therapy in advanced-stage

disease, the adverse events of these agents have been evaluated as a function of

disease stage. In studies of patients with early-stage disease, the most common side

effects were nausea (28%–47%), application site reactions (39%, rotigotine only),

dizziness (25%–40%), somnolence (22%–40%), insomnia (17%–19%), constipation

(14%), asthenia (14%), hallucinations (9%), and leg edema (5%).

21

,

34–37,38–40,51

Administration with food may help to relieve nausea and/or vomiting. With continued

use, many patients exhibit tolerance to the gastrointestinal side effects. CNS side

effects were the most common reason for discontinuation of these agents with older

patients particularly susceptible to hallucinations and other CNS side effects. The

incidence of orthostatic hypotension was relatively low (1%–9%) and may in part

reflect the exclusion of patients with underlying cardiovascular disease in several of

the studies.

In advanced-stage disease, the most common adverse events of dopamine agonists

were orthostatic hypotension (10%–54%), dyskinesias (26%–47%), application site

reactions (46%,

p. 1255

p. 1256

rotigotine only, dose related), nausea (25%), insomnia (27%), hallucinations

(11%–17%), somnolence (11%), and confusion (10%).

34

,

37

,

50

,

53 The most common

reasons for discontinuing these agents are mental disturbances (e.g., nightmares,

confusion, hallucinations, insomnia) and orthostatic hypotension. Dyskinesias that are

experienced when dopamine agonists are used in combination with levodopa in

advanced-stage disease may require lowering the dose of levodopa or, in some

cases, the dopamine agonist.

Sudden, excessive daytime somnolence, including while driving, has been

reported with dopamine agonists and has resulted in accidents.

34

,

37

,

50

,

54 Affected

patients did not always report warning signs before falling asleep and believed they

were alert immediately before the event. Labeling for these drugs includes a warning

that patients should be alerted to the possibility of falling asleep while engaged in

daily activities. Patients should be advised to refrain from driving or other

potentially dangerous activities until they have gained sufficient experience with the

dopamine agonist to determine whether it will hinder their mental and motor

performance. Caution should be advised when patients are taking other sedating

medications or alcohol in combination with dopamine agonists. If excessive daytime

somnolence does occur, patients should be advised to contact their healthcare

practitioner.

Dopamine agonist therapy in patients with PD is associated with 2- to 3.5-fold

increased odds of developing an impulse control disorder.

55 The frequency appears

similar for both pramipexole and ropinirole whereas it is less clear with rotigotine.

In one study, a prevalence of 6.1% was noted for pathologic gambling in patients

with PD compared with 0.25% for age- and sex-matched controls.

56 These cases may

represent variations of a behavioral syndrome termed dopamine dysregulation

syndrome.

57 Other features of the syndrome have been reported, including punding

(carrying out repetitive, purposeless motor acts), hypersexuality, walkabout (having

the urge to walk great distances during on-times, often with no purpose or destination

and abnormalities in time perception), compulsive buying, binge eating, drug

hoarding, and social independence or isolation.

57 The syndrome appears to be more

common among younger, male patients with early-onset PD, as well as those having

novelty-seeking personality traits, depressive symptoms, and current use of alcohol

or tobacco.

55

,

58 Management of impulse control disorders can be challenging,

because it often requires modification of dopaminergic therapies, which must be

carefully balanced with the accompanying risk of worsening motor function.

Underlying depression, if present, should be treated and may improve impulse

control. Nonpharmacologic measures (such as limiting access to money or the

Internet) may be helpful; in some cases, antipsychotic drugs may be considered, but

must also be used carefully to avoid precipitating motor disability.

59

Although L.M. is younger than 65 years of age, he may be at increased risk for

visual hallucinations and cognitive problems from dopamine agonist therapy. He

should be monitored closely for occurrence or exacerbation of these side effects. He

should also be evaluated for lightheadedness before initiation of pramipexole and

counseled to report dizziness or unsteadiness, because this may lead to falls. He

should be reassured that if these effects are caused by pramipexole, they should

subside with time and that he should not drive or operate complex machinery until he

can assess the effect of the drug on his mental status. L.M. should be counseled about

the possibility of excessive, and potentially unpredictable, daytime somnolence

because pramipexole is introduced. L.M. does not appear to have a problem with

excessive alcohol use; however, he and his family should be educated about his

increased risk for impulse control disorders and advised to report any new, unusual,

or uncharacteristic behaviors or increased use of alcohol.

MONOAMINE OXIDASE-B INHIBITORS

The monoamine oxidase-B inhibitors are irreversible inhibitors of MAO-B, a major

enzymatic pathway responsible for the metabolism of dopamine in the brain, and

inhibition increases the amount of striatal dopamine.

60 The discovery that ingestion of

MPTP causes rapid and irreversible parkinsonism led to the finding that the

neurotoxicity associated with MPTP is not directly caused by MPTP itself, but rather

the oxidized product, L-methyl-4-phenylpyridinium (MPP).

7 The conversion to MPP

is a two-step process mediated in part by MAO-B. Inhibition of MAO-B can inhibit

the oxidative conversion of dopamine to potentially reactive peroxides. This finding

has led investigators to study whether inhibition of this enzyme is neuroprotective.

Selegiline

In animals, pretreatment with selegiline (also referred to as deprenyl) protects

against neuronal damage after the administration of MPTP.

29 The Deprenyl and

Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) study was

designed to test the hypothesis that the combined use of selegiline and an antioxidant

(α-tocopherol) early in the course of the disease may slow disease progression.

61 The

primary outcome was the length of time that patients could be sustained without

levodopa therapy (an indication of disease progression). Early treatment with

selegiline 10 mg/day delayed the need to start levodopa therapy by approximately 9

months compared with patients given placebo; however, long-term observation

showed that the benefits of selegiline were not sustained and diminished with time. In

an extension of the study, patients originally assigned to selegiline tended to reach the

endpoint of disability after 1 year of observation even more quickly than did those

not assigned to receive selegiline.

62

Initial selegiline treatment did not alter the

development of levodopa adverse effects such as dyskinesias and wearing-off and

on–off phenomena.

Although selegiline did not prove to be neuroprotective, the finding that MAO-B

inhibitors can delay the need for dopaminergic therapy by at least several months is

clinically relevant and is supported by several randomized, controlled trials.

63

,

64

Aside from the DATATOP trial, studies of selegiline are limited by relatively small

sample sizes. As a result, a large meta-analysis of MAO-B inhibitors in early PD

was conducted.

30 When analyzed for selegiline, the levodopa dose necessary to

control symptoms was 67 mg (14–119 mg; P = 0.01) lower in the selegiline arm.

This analysis found that the total, motor, and ADL scores of the UPDRS were

significantly improved in the selegiline arm at 3 months. A subsequent Cochrane

Review found that the selegiline-treated arms scored 3.79 points better (95% CI:

2.21, 5.3) when looking at the weighted mean difference in motor UPDRS score.

29

Although oral selegiline continues to be FDA-approved only as adjunct treatment to

carbidopa/levodopa, the evidence suggests efficacy in monotherapy, and it is used as

monotherapy in practice.

Rasagiline

Rasagiline is a second-generation, irreversible selective inhibitor of MAO-B that is

FDA-approved as both monotherapy and adjunct therapy in PD. Rasagiline is

otherwise differentiated from selegiline primarily in that it is a more potent inhibitor

of MAO-B and does not carry the same potential for adverse effects resulting from

metabolites.

65 Like selegiline, rasagiline has also been found to protect from MPTPinduced parkinsonism in animal models.

29

Rasagiline was studied as monotherapy in early PD in a randomized, double-blind,

placebo-controlled trial comparing rasagiline 1 or 2 mg daily with placebo (n =

404).

66 After 6 months of therapy, the mean adjusted change in UPDRS scores

compared with placebo was improved in both the 1- and 2-mg groups (P<0.001).

66

These changes are quantitatively similar to those

p. 1256

p. 1257

observed with levodopa therapy. This study used a delayed-start design, wherein

at the end of the initial 6 months of treatment, patients who received placebo were

then switched over to receive active treatment with rasagiline, and the rasagilinetreated patients continued on therapy. After an additional 6 months of study, it was

found that patients receiving rasagiline for all 12 months had less functional decline

than patients in whom rasagiline was delayed.

67 The mean adjusted difference in total

UPDRS scores at 12 months for patients receiving rasagiline 2 mg/day for all 12

months was –2.29 compared with the delayed-start rasagiline 2-mg group (P = 0.01).

As a result, rasagiline was FDA-approved for use as monotherapy in early PD.

These encouraging findings also suggested that neuroprotection might be afforded by

rasagiline and prompted a larger, more definitive study.

The Attenuation of Disease Progression with Azilect Given Once-daily

(ADIAGO) study was also a randomized, placebo-controlled trial using the delayedstart methodology, but with a much larger sample size (n = 1,176).

68 Patients were

randomly assigned to receive rasagiline or placebo for 36 weeks, at which time

rasagiline-treated subjects continued therapy, and the placebo group was switched to

rasagiline; all patients were then followed for an additional 36 weeks. To prove

disease modification attributable to rasagiline with either dose, the early-start

treatment group had to meet each of three hierarchic endpoints, based on magnitude

and rate of change of UPDRS scores during different periods of the study. At the end

of the study, rasagiline failed to meet all of the prespecified endpoints and the authors

were unable to confirm any disease-modifying effects.

68

No direct comparisons of rasagiline and selegiline are available. Existing data

were compared in an industry-sponsored, indirect meta-analysis, which found a

significant advantage for rasagiline monotherapy on UPDRS scores.

69 Additionally,

risk for discontinuation and adverse events favored rasagiline. Rasagiline is

primarily metabolized via cytochrome P450 (CYP) 1A2 and N-dealkylation,

whereas selegiline is metabolized via CYP2B6, CYP2C9, and CYP3A4/5 to

amphetamine-like metabolites (L-methamphetamine and L-amphetamine). As a result

of their MAO-B inhibition, similar precautions regarding drug interactions exist;

caution should be taken when using MAO-B inhibitors with proserotonergic agents

such as antidepressants, triptans, and linezolid due to the risk of serotonin syndrome.

Although tyramine-challenge studies have not demonstrated any clinically significant

reactions with these MAO-B inhibitors, the product labeling still contains a warning

that patients should be advised to restrict tyramine intake.

70

,

71

Dosing

Selegiline is available in a 5-mg capsule or tablet and as a 1.25-mg orally

disintegrating tablet. It is also available in a transdermal patch, but this formulation is

not approved for use in PD (approved for treatment of major depressive disorder).

The bioavailability of conventional selegiline is low, and it undergoes extensive

hepatic first-pass metabolism into amphetamine-based metabolites, which may

contribute to neurologic side effects.

60 The usual dosage of conventional selegiline is

10 mg/day administered as 5 mg in the morning and early afternoon. It is not given in

the evening because excess stimulation from metabolites (L-methamphetamine and Lamphetamine) can cause insomnia and other psychiatric side effects.

72 The orally

disintegrating tablet formulation dissolves in the mouth in contact with saliva and

undergoes pregastric absorption. This formulation minimizes the effect of first-pass

metabolism in comparison with conventional selegiline, resulting in higher plasma

concentrations and reductions in the amphetamine-based metabolites.

60 Selegiline

should be used with caution in patients with severe hepatic impairment or with

creatinine clearance less than 30 mL/minute.

Rasagiline is available in 0.5- and 1-mg tablets. When used as monotherapy, it is

initiated at 1 mg daily. When combined with levodopa, the initial dose is lowered to

0.5 mg daily and can be increased to 1 mg daily based on response. In patients with

mild hepatic impairment, the dose should be reduced to 0.5 mg daily, but use should

be avoided in those with moderate-to-severe hepatic impairment. Additionally,

because rasagiline lacks the amphetamine-like metabolites of selegiline, it also lacks

the time-specific dosing constraints.

Adverse Effects

The most common adverse effects of both agents include nausea (6%–20%),

dizziness (11%–14%), headache (4%–14%), and dry mouth (4%–6%).

60

,

70

,

72 CNS

effects such as hallucinations, vivid dreams, and confusion also occur but at a lower

rate as compared with dopamine agonists. Although the oral disintegrating tablet

reduces the amphetamine-based metabolites, there are higher rates of reported side

effects, likely as a result of the higher plasma concentration of selegiline.

60 A large

meta-analysis of MAO-B inhibitors in early PD found a higher incidence of reported

side effects among the MAO-B inhibitor (all but one study used selegiline) versus

non-MAO-B inhibitor groups (OR: 1.36, 1.02–1.8, P = 0.04).

30 However, these

agents are generally considered well tolerated.

70

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

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

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

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more