Emedicine Parkinsons Diseasae

Embed Size (px)

Citation preview

  • 8/2/2019 Emedicine Parkinsons Diseasae

    1/31

    Parkinson DiseaseRobert A Hauser, MD, MBA, Professor of Neurology, Molecular Pharmacology and Physiology, Director, Parkinson's Disease andMovement Disorders Center, University of South Florida; Clinical Chair, Signature Interdisciplinary Program in Neuroscience

    Rajesh Pahwa, MD, Professor of Neurology, Director, Parkinson Disease and Movement Disorder Center, Department of Neurology,University of Kansas Medical Center; Kelly E Lyons, PhD, Research Associate Professor of Neurology, Director of Research andEducation, Parkinson's Disease and Movement Disorder Center, University of Kansas Medical Center; Theresa McClain, MSN,ARNP, Parkinson's Disease and Movement Disorders Center, University of South Florida

    Updated: Apr 27, 2010

    Introduction

    Background

    Parkinson disease (Parkinson's disease, PD) is a progressive neurodegenerative disorder associated with a loss of

    dopaminergic nigrostriatal neurons. It is named after James Parkinson, the English physician who described the shaking

    palsy in 1817.

    Parkinson disease is recognized as one of the most common neurological disorders, affecting approximately 1% o

    individuals older than 60 years. Cardinal features include resting tremor, rigidity, bradykinesia, and postural instability.

    Pathophysiology

    The major neuropathologic findings in Parkinson disease are a loss of pigmented dopaminergic neurons in the substantia

    nigra and the presence of Lewy bodies. The loss of dopaminergic neurons occurs most prominently in the ventral lateralsubstantia nigra. Approximately 60-80% of dopaminergic neurons are lost before the motor signs of Parkinson disease

    emerge.

    Lewy bodies are concentric, eosinophilic, cytoplasmic inclusions with peripheral halos and dense cores. The presence o

    Lewy bodies within pigmented neurons of the substantia nigra is characteristic, but not pathognomonic, of idiopathic

    Parkinson disease. Lewy bodies also are found in the cortex, nucleus basalis, locus ceruleus, intermediolateral column of

    the spinal cord, and other areas. Lewy bodies are not specific to Parkinson disease, as they are found in some cases of

    atypical parkinsonism, Hallervorden-Spatz disease, and other disorders. Incidental Lewy bodies are found at postmortem

    in patients without clinical signs of parkinsonism. The prevalence of incidental Lewy bodies increases with age. Incidenta

    Lewy bodies have been hypothesized to represent the presymptomatic phase of Parkinson disease.

    No standard criteria exist for the neuropathologic diagnosis of Parkinson disease, as the specificity and sensitivity of thecharacteristic findings have not been established clearly. Individuals presenting with primary dementia may exhibi

    neuropathologic features indistinguishable from those of Parkinson disease.

    Alpha-synuclein is a major structural component of Lewy bodies. All Lewy bodies stain for alpha-synuclein and most also

    stain for ubiquitin.

    Stages in the development of Parkinson disease-related pathology. Adapted from Braak H, Ghebremedhin E, Rub

    U, Bratzke H, Del Tredici K. Cell Tissue Res. 2004 Oct;318(1):121-34.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    2/31

    Recent studies demonstrate that Lewy-body pathology in Parkinson disease actually begins in the olfactory bulb and

    lower brainstem (see image above or Media file 4). [1 ]These early stages are associated with premotor symptoms such as

    loss of sense of smell and rapid eye movement (REM) sleep behavior disorder (RBD). [2 ]The pathology ascends up the

    brainstem to later involve the midbrain and nigrostriatal dopaminergic neurons. This stage correlates with onset of the

    motor phase of the disease and patients may exhibit bradykinesia, rigidity, and tremor. The pathology continues to

    ascend late in the disease to affect the cortex and patients may then exhibit cognitive dysfunction and dementia.

    Motor circuit in Parkinson disease

    The basal ganglia motor circuit modulates cortical output necessary for normal movement (see following image or Mediafile 1).

    Schematic representation of the basal ganglia - thalamocortical motor circuit and its neurotransmitters in the

    normal state. From Vitek J. Stereotaxic surgery and deep brain stimulation for Parkinson's disease and

    movement disorders. In: Watts RL, Koller WC, eds. Movement Disorders: Neurologic Principles and Practice. New

    York: McGraw-Hill, 1997:240. Used with kind permission. Copyright, McGraw-Hill Companies, Inc.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    3/31

    Signals from the cerebral cortex are processed through the basal ganglia-thalamocortical motor circuit and return to the

    same area via a feedback pathway. Output from the motor circuit is directed through the internal segment of the globus

    pallidus (GPi) and the substantia nigra pars reticulata (SNr). This inhibitory output is directed to the thalamocortical

    pathway and suppresses movement.

    Two pathways exist within the basal ganglia circuit; they are referred to as the direct and indirect pathways. In the direct

    pathway, outflow from the striatum directly inhibits GPi and SNr. The indirect pathway comprises inhibitory connections

    between the striatum and the external segment of the globus pallidus (GPe) and the GPe and the subthalamic nucleus

    (STN). The subthalamic nucleus exerts an excitatory influence on the GPi and SNr. The GPi/SNr sends inhibitory output

    to the ventral lateral (VL) nucleus of the thalamus. Striatal neurons containing D1 receptors constitute the direct pathway

    and project to the GPi/SNr. Striatal neurons containing D2 receptors are part of the indirect pathway and project to theGPe.

    Dopamine is released from nigrostriatal (SNc) neurons to activate the direct pathway and inhibit the indirect pathway. In

    Parkinson disease, decreased striatal dopamine causes increased inhibitory output from the GPi/SNr (see following

    image or Media file 2).

    Schematic representation of the basal ganglia - thalamocortical motor circuit and the relative

    change in neuronal activity in Parkinson disease. From Vitek J. Stereotaxic surgery and deep brain

    stimulation for Parkinson's disease and movement disorders. In: Watts RL, Koller WC, eds

    Movement Disorders: Neurologic Principles and Practice. New York: McGraw-Hill, 1997:241. Used

    with kind permission. Copyright, McGraw-Hill Companies, Inc.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    4/31

    This increased inhibition of the thalamocortical pathway suppresses movement. Via the direct pathway, decreased striata

    dopamine stimulation causes decreased inhibition of the GPi/SNr. Via the indirect pathway, decreased dopamine

    inhibition causes increased inhibition of the GPe, resulting in disinhibition of the STN. Increased STN output increases

    GPi/SNr inhibitory output to the thalamus.

    Frequency

    International

    The incidence has been estimated to be 4.5-21 cases per 100,000 population per year. Estimates of Parkinson disease

    prevalence range from 18-328 per 100,000 population, with most studies yielding a prevalence of approximately 120 per

    100,000.

    Sex

    Parkinson disease is about 1.5 times more common in men than in women.

    Age

    The incidence and prevalence of Parkinson disease increase with age. The average age of onset is approximately 60

    years. Onset in persons younger than 40 years is relatively uncommon.

    Clinical

    History

    Parkinson disease may have a long premotor phase. Mid-life risk factors for the later development of Parkinson

    disease include constipation and daytime sleepiness. These may well be the first clinical manifestations of the

    disease but are nonspecific. Additional features that commonly precede onset of motor signs include decreased

    sense of smell and REM behavior disorder (RBD).

    o REM behavior disorder is a sleep disorder in which there is a loss of normal atonia during REM sleep.

    o Patients are observed by their bed partners to act out their dreams and the partners may note kicking

    hitting, talking, or crying out.

    o In one study, 38% of 50-year-old men with REM behavior disorder and no neurologic signs went on to

    develop Parkinsonism.[3 ]

    o REM behavior disorder is common throughout the course of Parkinson disease.

    o Postuma et al found that the subsequent development of Parkinson disease in patients with idiopathic

    REM sleep behavior disorder (RBD) could be predicted on the basis of polysomnography results. In a

    longitudinally studied cohort of patients with idiopathic RBD who were initially free of neurodegenerative

    disease, those who developed Parkinson disease had increased tonic chin electromyographic activity

    during REM sleep at baseline compared with those who remained disease-free (62.7 +/- 6% vs 41 +/

    6%, p =0.02). The interval between polysomnography and disease onset was 6.7 years.[4 ]

    Onset of motor signs in Parkinson disease is typically asymmetric, with the most common initial finding being an

    asymmetric resting tremor in an upper extremity. About 20% of patients first experience clumsiness in one hand

    Over time, patients notice symptoms related to progressive bradykinesia, rigidity, and gait difficulty.

    Tremor usually begins in one upper extremity and initially may be intermittent. As with most tremors, the

    amplitude increases with stress and resolves during sleep. After several months or years, the tremor may affect

    the extremities on the other side, but asymmetry is usually maintained. Parkinson disease tremor may alsoinvolve the lower extremities, tongue, lips, or chin.

    The initial symptoms of Parkinson disease may be nonspecific and include fatigue, depression, constipation, and

    sleep problems.

    Some patients experience a subtle decrease in dexterity and may notice a lack of coordination with activities

    such as playing golf or dressing.

    Some patients complain of aching or tightness in the calf or shoulder region.

    The first affected arm may not swing fully when walking, and the foot on the same side may scrape the floor.

    Over time, axial posture becomes progressively flexed and strides become shorter.

    Decreased swallowing may lead to excess saliva in the mouth and ultimately drooling.

    Symptoms of autonomic dysfunction are common and include constipation, sweating abnormalities, sexua

    dysfunction, and seborrheic dermatitis.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    5/31

    Sleep disturbances are common.

    The best clinical predictors of a pathology diagnosis of Parkinson disease are the following:

    o Asymmetry

    o Presence of resting tremor

    o Good response to dopamine replacement therapy

    Long-term disability in Parkinson disease is usually related to dementia and balance dysfunction.

    Physical

    The 3 cardinal signs of Parkinson disease are resting tremor, rigidity, and bradykinesia. Of these cardinal features, 2 of 3

    are required to make the clinical diagnosis. Postural instability (balance dysfunction) is the fourth cardinal sign, but i

    emerges late in the disease, usually after 8 years or more.

    The characteristic Parkinson disease tremor is present and most prominent with the limb at rest.

    o The usual frequency is 3-5 Hz.

    o The tremor may appear as a pill-rolling motion of the hand or a simple oscillation of the hand or arm.

    o The same tremor may be observed with the arms outstretched (position of postural maintenance) and a

    less prominent, higher frequency kinetic tremor is also common.

    Rigidity refers to an increase in resistance to passive movement about a joint.

    o The resistance can be either smooth (lead pipe) or oscillating (cogwheeling).

    o Cogwheeling is thought to reflect tremor rather than rigidity and may be present with tremors no

    associated with an increase in tone (ie, essential tremor).o Rigidity usually is tested by flexing and extending the patient's relaxed wrist.

    o Rigidity can be made more obvious with voluntary movement in the contralateral limb.

    Bradykinesia refers to slowness of movement but also includes a paucity of spontaneous movements and

    decreased amplitude of movement. Bradykinesia is also expressed as micrographia (small handwriting)

    hypomimia (decreased facial expression), decreased blink rate, and hypophonia (soft speech).

    Postural instability refers to imbalance and loss of righting reflexes. Its emergence is an important milestone

    because it is poorly amenable to treatment and a common source of disability in late disease.

    Patients may experience freezing when starting to walk (start-hesitation), during turning, or while crossing a

    threshold, such as going through a doorway.

    Dementia generally occurs late in Parkinson disease and affects 15-30% of patients. Short-term memory and

    visuospatial function may be impaired, but aphasia is not present. Cognitive dysfunction within a year of onset of

    motor features suggests a diagnosis of Lewy body disease, a disease closely related to Parkinson disease and

    marked by the presence of cortical Lewy bodies. See Parkinson Disease Dementia for more information.

    Causes

    Most cases of idiopathic Parkinson disease are believed to be due to a combination of genetic and environmental factors

    At both ends of the spectrum are rare cases that appear to be due solely to one or the other.

    Environmental risk factors associated with the development of Parkinson disease include use of pesticides, living

    in a rural environment, consumption of well water, exposure to herbicides, and proximity to industrial plants or

    quarries.

    Several individuals have been identified who developed parkinsonism after self-injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP).[5 ]

    o These patients developed bradykinesia, rigidity, and tremor, which progressed over several weeks and

    improved with dopamine replacement therapy.

    o MPTP crosses the blood-brain barrier and is oxidized to MPP+ by the enzyme monoamine oxidase

    (MAO) type B.

    o MPP+ accumulates in mitochondria and interferes with the function of complex I of the respiratory chain.

    o A chemical resemblance between MPTP and some herbicides and pesticides suggested that an MPTP-

    like environmental toxin might be a cause of Parkinson disease, but no specific agent has been

    identified. Nonetheless, mitochondrial complex I activity is reduced in Parkinson disease, suggesting a

    common pathway with MPTP-induced parkinsonism.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    6/31

    The oxidation hypothesis suggests that free radical damage, resulting from dopamine's oxidative metabolism,

    plays a role in the development or progression of Parkinson disease.

    o The oxidative metabolism of dopamine by MAO leads to the formation of hydrogen peroxide. Hydrogen

    peroxide normally is cleared rapidly by glutathione.

    o If hydrogen peroxide is not cleared adequately, it may lead to the formation of highly reactive hydroxy

    radicals that can react with cell membrane lipids to cause lipid peroxidation and cell damage. In

    Parkinson disease, levels of reduced glutathione are decreased, suggesting a loss of protection against

    formation of free radicals. Iron is increased in the substantia nigra and may serve as a source of dono

    electrons, thereby promoting the formation of free radicals.

    o Indices of lipid peroxidation are increased in Parkinson disease.

    o Thus, Parkinson disease is associated with increased dopamine turnover, decreased protective

    mechanisms (glutathione), increased iron (a pro-oxidation molecule), and evidence of increased lipid

    peroxidation. This hypothesis raised concern that increased dopamine turnover due to levodopa

    administration could increase oxidative damage and accelerate loss of dopamine neurons. However

    there is no clear evidence that levodopa accelerates disease progression.

    The role of genetic factors has been studied in twins.

    o If genetic factors are important, concordance in genetically identical monozygotic (MZ) twins will be

    greater than in dizygotic (DZ) twins, who share only about 50% of genes. Early twin studies generally

    found low and similar concordance rates for MZ and DZ pairs.

    o In a recent study of 193 twins, overall concordance for MZ and DZ pairs was similar. However, in 16

    pairs of twins in whom Parkinson disease was diagnosed at or before age 50 years, all 4 MZ pairs, but

    only 2 of 12 DZ pairs, were concordant. This suggests that while genetic factors may not be very

    important when the disease begins after age 50 years, genetic factors appear to be very important when

    the disease begins at or before age 50 years.

    The identification of a few large families with apparent familial Parkinson disease sparked further interest in the

    genetics of the disease.

    o One large family with highly penetrant, autosomal-dominant, autopsy-proven Parkinson disease

    originated in the town of Contursi in the Salerno province of southern Italy. Of 592 family members, 50

    were affected by Parkinson disease. These individuals were characterized by early age of disease onset

    (mean age 47.5 y), rapid progression (mean age at death 56.1 y), lack of tremor, and good response to

    levodopa therapy.

    o Linkage analysis incriminated a region in chromosome bands 4q21-23, and sequencing revealed an A

    for-G substitution at base 209 of the alpha-synuclein gene. Termed PD-1, this mutation codes for asubstitution of threonine for alanine at amino acid 53.

    o Five small Greek kindreds also were found to have the PD-1 mutation. In a German family, a differen

    point mutation in the alpha-synuclein gene (a substitution of C for G at base 88, producing a substitution

    of proline for alanine at amino acid 30) confirmed that mutations in the alpha-synuclein gene can cause

    Parkinson disease. A few additional familial mutations in the alpha-synuclein gene have been identified

    and are now collectively called PARK1. It is now clear that these mutations are an exceedingly rare

    cause of Parkinson disease.

    Alpha-synuclein is a major component of Lewy bodies in all Parkinson disease.

    o All Lewy bodies stain for alpha-synuclein, and most also stain for ubiquitin, which conjugates with

    proteins targeted for proteolysis. Abnormal aggregation of alpha-synuclein into filamentous structures

    may precede ubiquitization.o One hypothesis states that the PD-1 mutation alters the configuration of alpha-synuclein into a

    structure that could aggregate into sheets.

    o All Parkinson disease may be associated with abnormal folding of alpha-synuclein, leading to excessive

    aggregation and neuronal death.

    o Although sporadic Parkinson disease is not caused by a mutation in the alpha-synuclein gene, active

    investigation is underway into proteins that interact with alpha-synuclein, including those that guide

    promote, or prevent aggregation of the protein.

    o As Parkinson disease, dementia with Lewy bodies, and multiple system atrophy (MSA) all exhibit Lewy

    bodies that stain for alpha-synuclein, they have been designated "alpha-synucleinopathies."

    A recent hypothesis suggests that Parkinson disease is caused by abnormalities of the proteosome system

    which is responsible for clearing abnormal proteins.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    7/31

    Several homozygous deletions in a gene dubbed parkin (PARK2), which is located on chromosome 6, have been

    found to cause autosomal-recessive juvenile parkinsonism (AR-JP). This form of parkinsonism differs

    pathologically from Parkinson disease in that no Lewy bodies are found in the substantia nigra.

    Several other gene abnormalities have been identified in families with Parkinson disease and these are

    designated PARK3 -PARK12.

    It has been estimated that all currently known genetic causes of Parkinson disease account for less than 5% of

    Parkinson disease cases.

    Differential Diagnoses

    Alzheimer Disease Normal Pressure Hydrocephalus

    Cardioembolic Stroke Parkinson-Plus Syndromes

    Cortical Basal Ganglionic Degeneration Progressive Supranuclear Palsy

    Essential Tremor Striatonigral Degeneration

    Hallervorden-Spatz Disease

    Lacunar Syndromes

    Multiple System Atrophy

    Other Problems to Be Considered

    Jakob-Creutzfeldt and other prion diseases

    Parkinsonism can be caused by a variety of degenerative disorders, as well as toxins, infections, and vascular or

    structural lesions.

    Parkinsonism also can be induced by medications that block dopamine receptors (eg, neuroleptics, antiemetics) o

    deplete intraneuronal dopamine stores (eg, reserpine, tetrabenazine).

    Workup

    Laboratory Studies

    No laboratory biomarkers exist for Parkinson disease.

    Serum ceruloplasmin concentration is obtained as a screening test for Wilson disease. It should be obtained inpatients who present with parkinsonian symptoms when younger than 40 years. In cases in which Wilson

    disease is suspected, 24-hour urinary copper and slit lamp examination of the eyes also should be obtained.

    Imaging Studies

    Magnetic resonance imaging (MRI) and computed tomography (CT) scan are unremarkable in Parkinson

    disease.

    o No imaging study is required in patients with a typical presentation. Such patients are aged 55 years or

    older; have a slowly progressive, asymmetric parkinsonism with resting tremor and bradykinesia o

    rigidity; and demonstrate a good response to dopamine replacement therapy.

    o

    MRI is useful to exclude multi-infarct state, hydrocephalus, and the lesions of Wilson disease.o MRI should be obtained in patients whose clinical presentation does not allow a high degree o

    diagnostic certainty, including those who lack tremor, have an acute or stepwise progression, or are

    younger than 55 years.

    Positron emission tomography (PET) and single photon emission CT (SPECT) are useful diagnostic imaging

    studies. They are not widely available and may not be covered by insurance. Moreover, they are not needed for

    routine clinical diagnosis in patients with a typical presentation.

    o At the onset of symptoms, patients with Parkinson disease show approximately 30% decrease in 18F

    dopa uptake in the contralateral putamen.

    o 18F-dopa is taken up by the terminals of dopamine neurons and converted to 18F-dopamine. The rate

    of striatal 18F accumulation reflects transport of 18F-dopa into dopamine neurons and its

    decarboxylation to 18F-dopamine.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    8/31

    o 11C-Nomifensine and cocaine analogues such as 123I-Beta-CIT bind to dopamine reuptake sites on

    nigrostriatal terminals and provide an index of the integrity of nigrostriatal projections.

    o Deficits on fluorodopa PET or -CIT SPECT indicate a dopamine deficiency syndrome but do no

    necessarily differentiate Parkinson disease from atypical parkinsonisms including multiple system

    atrophy and progressive supranuclear palsy.

    Treatment

    Medical Care

    The goal of medical management of Parkinson disease is to provide control of signs and symptoms for as long aspossible while minimizing adverse effects. Medications usually provide good symptomatic control of motor signs for 4-6

    years. After this, disability progresses despite best medical management, and many patients develop long-term motor

    complications including fluctuations and dyskinesia. Additional causes of disability in late disease include postura

    instability (balance difficulty) and dementia.

    Putative neuroprotective therapy

    Neuroprotective therapies are defined as those that slow the underlying loss of dopamine neurons. Currently, no proven

    neuroprotective therapies exist for Parkinson disease.

    If a neuroprotective therapy were available for Parkinson disease, it would be administered from the time of diagnosis

    onward.

    Selegiline

    o Selegiline is the medication that first has garnered wide interest as a possible neuroprotective agent.

    o Laboratory investigations continue to provide evidence that selegiline affords a neuroprotective effect fo

    dopamine neurons independent of MAO-B inhibition.

    o Selegiline has been demonstrated to protect cultured dopamine neurons from MPP+ toxicity, an effec

    that cannot be attributed to MAO-B inhibition. Tatton and Greenwood demonstrated that selegiline

    protects dopamine cells in mice from MPTP toxicity even when administered after a delay sufficient to

    allow the oxidation of MPTP to MPP+.

    o In cell-culture systems, selegiline's neuroprotective effect is mediated by new protein synthesis

    Selegiline induces transcriptional events that result in increased synthesis of antioxidant and anti-apoptotic proteins. Recent evidence indicates that one of selegiline's metabolites, desmethylselegiline, is

    the active agent for neuroprotection.

    o Selegiline's amphetamine metabolites may interfere with its neuroprotective actions.

    o In the clinical study called DATATOP (deprenyl [selegiline] and tocopherol [vitamin E] antioxidative

    therapy of parkinsonism), the Parkinson Study Group evaluated the ability of these 2 medications to

    delay progression of clinical disability in early Parkinson disease. Eight hundred patients were

    randomized to receive selegiline (10 mg/d) or placebo and tocopherol (2000 IU/d) or placebo. Patients

    assigned to receive selegiline, with placebo or with tocopherol, experienced a significant delay in the

    need for levodopa therapy (hazard ratio = 0.50, P

  • 8/2/2019 Emedicine Parkinsons Diseasae

    9/31

    emerged between the third and fifth years of treatment, and no obvious explanation regarding its cause

    was identified.

    o Many questions have been raised regarding the results and methodology of this study. Mortality rates

    were not significantly different between groups when the analysis was based on what patients actually

    were taking and not on intention to treat. In addition, the mortality rate was unusually high in both groups

    (28% in patients receiving selegiline, 18% in those not receiving selegiline).

    o In a more recent study, patients with early Parkinson disease were randomized to selegiline or placebo

    and levodopa was added as needed. Over 7 years, patients receiving selegiline experienced less clinica

    progression and required less levodopa than patients receiving placebo.[7 ]

    Rasagilineo Rasagiline is a MAO-B inhibitor that exhibits neuroprotective effects in cell culture and animal models. In

    a clinical trial of patients with early Parkinson disease (TEMPO), treatment with rasagiline for 1 year

    provided significantly greater improvement than treatment with placebo for 6 months followed by

    rasagiline for 6 months. [8 ]When patients were followed long term (5.5-6 y), those who started rasagiline

    at the beginning of the study experienced 16% less progression of disability than those who started i

    after 6 months.[9 ]

    o ADAGIO was a large and rigorous delayed-start study of rasagiline. Patients with early disease were

    randomized to active drug (1 or 2 mg) or placebo for 9 months and then all patients went on active drug

    for 9 months. Results showed that patients who received rasagiline 1 mg/d from the start of the trial had

    less progression of clinical disability than subjects who received rasagiline 1 mg/d after a delay of 9

    months.[10 ]

    o A study by Olanow et al of 1176 patients with untreated Parkinson disease showed early treatment with

    rasagiline provided benefits potentially consistent with disease-modifying effects. The outcome was

    positive for rasagiline 1 mg/d but not 2 mg/d, although a subanalysis evaluating the most affected

    quartile at baseline was positive for both 1 mg/d and 2 mg/d. The reason behind the discrepancy in

    outcome between the 2 doses is not known.[11 ]

    Co-enzyme Q10 is another drug of interest. It is a scavenger of free radicals. In a preliminary study, co-enzyme

    Q10 1200 mg/d slowed progression of Parkinson disease disability.[12 ]

    Further studies of rasagiline and co-enzyme Q10 are required.

    Symptomatic therapy

    Levodopa, coupled with a peripheral decarboxylase inhibitor (PDI), remains the standard of symptomatictreatment for Parkinson disease. It provides the greatest antiparkinsonian benefit with the fewest adverse effects

    in the short term. However, its chronic use is associated with the development of fluctuations and dyskinesias.

    Dopamine agonists provide symptomatic benefit comparable to levodopa/PDI in early disease but lack sufficient

    efficacy to control signs and symptoms by themselves in more advanced disease.

    Dopamine agonists cause more sleepiness, hallucinations, edema, and impulse control disorders than levodopa.

    Prospective, double-blind studies have demonstrated that initial treatment with a dopamine agonist, to which

    levodopa can be added as necessary, causes less motor fluctuations and dyskinesias than levodopa alone.

    Subsequent analyses of these studies indicate that the benefit of dopamine agonists in delaying moto

    complications is due to their ability to delay the need for levodopa/PDI.[13,14 ]

    Dopamine agonists can be used as initial symptomatic therapy in early disease, rather than levodopa/PDI, to

    delay the onset of motor fluctuations and dyskinesia. This strategy is usually reserved for younger individuals(

  • 8/2/2019 Emedicine Parkinsons Diseasae

    10/31

    The Continuous Dopaminergic Stimulation (CDS) hypothesis posits that pulsatile dopamine receptor stimulation

    induces dyskinesia, whereas smoother more continuous dopamine receptor stimulation causes less dyskinesia.

    In contrast to levodopa, the long-acting dopamine agonists (ie, bromocriptine, pramipexole, ropinirole

    cabergoline) provide relatively smooth and sustained receptor stimulation. In marmosets with MPTP-induced

    parkinsonism, levodopa administration causes significantly more dyskinesia than bromocriptine or ropinirole.

    Dopamine agonists alone (without concurrent treatment with levodopa/PDI) rarely cause fluctuations o

    dyskinesia.

    Prospective clinical trials have demonstrated that initial treatment with a dopamine agonist to which levodopa can

    be added causes less motor fluctuations and dyskinesia than levodopa alone.

    A recent MPTP marmoset study found that the addition of entacapone (which increases the half-life of levodopa)was associated with less motor fluctuations and less dyskinesia than treatment with the same regimen o

    levodopa alone. This finding is consistent with the CDS hypothesis. A clinical trial (STRIDE-PD) is now underway

    to determine if levodopa plus entacapone (levodopa/carbidopa/entacapone) delays the occurrence of dyskinesia

    compared with levodopa/carbidopa when levodopa is first required.

    Early disease treatment strategies

    Studies demonstrate that a patient's quality of life deteriorates quickly if treatment is not instituted at or shortly

    after diagnosis.[15 ]

    The younger the patient, the more emphasis the authors place on long-term considerations to guide early

    treatment. Young patients have a longer life expectancy and are more likely to develop motor fluctuations and

    dyskinesias.

    For older patients and those with cognitive impairment, less emphasis is placed on long-term considerations; the

    focus is on providing adequate symptomatic benefit in the near term with as few adverse effects as possible.

    MAO-B inhibitors provide mild symptomatic benefit, have excellent side effect profiles, and may improve long-

    term outcomes. These characteristics make MAO-B inhibitors a good choice as initial treatment for many

    patients. When the MAO-B inhibitor alone is not sufficient to provide good control of motor symptoms, another

    medication (eg, dopamine agonist or levodopa) is added.

    Levodopa is the most efficacious symptomatic medication with few short-term side effects, but its chronic use is

    associated with the development of fluctuations and dyskinesias. Once fluctuations and dyskinesias become

    problematic, they are difficult to resolve.

    Dopamine agonists provide moderate symptomatic benefit and rarely cause fluctuations and dyskinesias by

    themselves, but they have more side effects than levodopa, including sleepiness and impulse control disordersHowever, these side effects resolve upon lowering the dose or discontinuing the medication.

    Dopamine agonists and levodopa are started at a low dose, escalated slowly, and titrated to control symptoms.

    For patients younger than 65 years, the authors often use a dopamine agonist and then add levodopa/PDI when

    the dopamine agonist (with or without an MAO-B inhibitor) no longer provides good control of motor symptoms.

    Dopamine agonists may provide good symptom control for several years.

    For patients who are demented or older than 70 years (those who may be prone to adverse effects, such as

    hallucinations, from dopamine agonists), and for those likely to require treatment for only a few years, the authors

    may elect not to use a dopamine agonist and depend on levodopa/PDI as primary symptomatic therapy.

    For patients aged 65-70 years, the authors make a judgment based on general health and cognitive status. The

    more robust and cognitively intact the patient, the more likely the authors are to treat with a dopamine agonis

    prior to levodopa and add levodopa/PDI when necessary. When introducing a dopamine agonist, starting at a low dose and escalating slowly is important. The dose should

    be titrated upward until symptoms are controlled, the maximum dose is reached, or adverse effects become

    intolerable. The most common adverse effects of dopamine agonists are nausea, orthostatic hypotension

    hallucinations, somnolence, and impulse control disorders. Nausea usually can be reduced by having the patien

    take the medication after meals. Domperidone, a peripheral dopamine agonist available outside the US, is very

    helpful in relieving refractory nausea. Patients on dopamine agonists should be routinely asked about sleepiness

    sudden onset of sleep, and impulse control disorders such as pathologic gambling, shopping, internet use, o

    sexual activity.

    Levodopa/PDI is introduced at a low dose and escalated slowly. Currently available levodopa preparations in the

    United States include levodopa/carbidopa, levodopa/carbidopa CR, levodopa/carbidopa orally disintegrating

  • 8/2/2019 Emedicine Parkinsons Diseasae

    11/31

    tablet, and levodopa/carbidopa/entacapone. The orally disintegrating tablet is bioequivalent to ora

    levodopa/carbidopa but dissolves on the tongue without the need to swallow it with water.

    The levodopa dose is titrated to control clinical symptoms; most patients experience a good response on a daily

    dosage of 400-600 mg/d for 3-5 years or more. Doses higher than those necessary to control symptoms

    adequately should be avoided.

    If nausea occurs, the levodopa dose may be taken following a meal. Additional measures to alleviate nausea

    include adding extra carbidopa or introducing domperidone.

    A recently completed study (FIRST STEP) demonstrated that treatment with levodopa/carbidopa/entacapone

    (Stalevo) provided greater symptomatic benefit than levodopa/carbidopa at the same levodopa dose, without an

    increase in motor complications.

    [16 ]

    Another study (STRIDE-PD) is ongoing and will evaluate whethelevodopa/carbidopa/entacapone causes less dyskinesia than levodopa/carbidopa.

    For patients who have disability due to tremor that is not adequately controlled with dopaminergic medication, an

    anticholinergic agent can be used. Anticholinergic medications provide good tremor relief in approximately 50%

    of patients but do not improve bradykinesia or rigidity. Because tremor may respond to one anticholinergic

    medication and not another, a second anticholinergic usually is tried if the first is not successful. These

    medications should be introduced at a low dose and escalated slowly to minimize adverse effects, which include

    memory difficulty, confusion, and hallucinations. Adverse cognitive effects are relatively common, especially in

    the elderly.

    Advanced disease treatment strategies

    Patients initially experience stable, sustained benefit through the day in response to levodopa. However, after

    several months to years, many patients notice that the benefit from immediate release levodopa/carbidopa wears

    off after 4-5 hours. Over time, this shortened duration of response becomes more fleeting, and clinical status

    fluctuates more and more closely in concert with peripheral levodopa concentration. Ultimately, benefit lasts only

    1-2 hours. The time when medication is providing benefit for bradykinesia, rigidity, and tremor is called "on" time

    and the time when medication is not providing benefit is called "off" time.

    By several months to years after the introduction of levodopa, many patients develop peak-dose dyskinesia

    consisting of choreiform, twisting/turning movements that occur when levodopa-derived dopamine levels are

    peaking. At this point, increasing dopamine stimulation is likely to worsen peak-dose dyskinesias and decreasing

    dopamine stimulation may worsen Parkinson disease motor signs and increase off time. The therapeutic

    window lies above the threshold required to improve symptoms (on threshold) and below the threshold for peak-

    dose dyskinesia (dyskinesia threshold). The therapeutic window narrows over time because of a progressivedecrease in the threshold for peak-dose dyskinesia.

    Although many patients prefer mild dyskinesia to off time, the clinician should recognize that dyskinesias can be

    sufficiently severe to be troublesome to the patient, either by interfering with activities or because of discomfort.

    Asking patients how they feel during both off time and time with dyskinesia is important in titrating medication

    optimally. Having patients fill out a diary may be helpful; the diary should be divided into half-hour time periods

    on which the patient denotes whether they are off, on without dyskinesia, on with nontroublesome dyskinesia, or

    on with troublesome dyskinesia (see image below or Media file 3). The goal of medical management is to

    minimize off time and time on with troublesome dyskinesia.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    12/31

    Parkinson disease diary. The patient or caregiver should place 1 check mark in each half-hour

    time slot to indicate the patient's predominant response during most of that period. The goa

    of therapeutic management is to minimize off time and on time with troublesome dyskinesia

    Copyright Robert Hauser, 1996. Used with permission.

    Treating motor fluctuations in the absence of peak-dose dyskinesia is relatively easy. Several different strategies

    either alone or in combination, can be used to provide more sustained dopaminergic therapy. Possible strategies

    include adding a dopamine agonist, catechol-O -methyltransferase (COMT) inhibitor, or MAO-B inhibitor; dosing

    levodopa more frequently; increasing the levodopa dose; or switching from immediate release to CR levodopa or

    levodopa/carbidopa/entacapone. Unless limited by the emergence of peak-dose symptoms such as dyskinesia

    or hallucinations, dopaminergic therapy should be increased until off time is eliminated.

    The treatment of patients with both motor fluctuations and troublesome peak-dose dyskinesia can be difficult

    The goal of treatment in this situation is to provide as much good functional time through the day as possible.

    This is accomplished by maximizing on time without troublesome dyskinesia. An attempt is made to reduce both

    off time and time with troublesome or disabling dyskinesia. Unfortunately, a decrease in dopaminergic therapy

    may increase off time and an increase in dopaminergic therapy may worsen peak-dose dyskinesia.

    For patients with severe fluctuations and dyskinesia, the best balance between off time and troublesome

    dyskinesia is sought. The patient's relative preference for off time versus dyskinesia needs to be taken into

    account.

    For patients with motor fluctuations and dyskinesia on levodopa/PDI, the addition of a dopamine agonist, COMT

    inhibitor, or MAO-B inhibitor may be helpful. Dyskinesia may increase when these medications are added

    necessitating the downward titration of levodopa.

    For patients on CR levodopa, switching to immediate release levodopa/carbidopa often provides a more

    consistent and predictable dosing cycle and allows finer titration. In general, smaller levodopa doses are

    administered more frequently. A dose should be sought that is sufficient to provide benefit without causing

    troublesome dyskinesia. The time to wearing-off then determines the appropriate interdose interval. The extreme

  • 8/2/2019 Emedicine Parkinsons Diseasae

    13/31

    of this strategy is using liquid levodopa, a solution with which the dose can be titrated finely and administered

    every hour. Amantadine may also be of benefit to reduce dyskinesia.

    COMT inhibitors inhibit the peripheral metabolism of levodopa to 3-O -methyldopa (3-OMD), thereby prolonging

    the levodopa half-life and making more levodopa available for transport across the blood-brain barrier over a

    longer time.

    Tolcapone was the first COMT inhibitor available for clinical use. Because of the potential risk of hepatotoxicity,

    liver function test monitoring is required, and it should be used only in patients who are experiencing motor

    fluctuations on levodopa that cannot be adequately controlled with other medications.

    If dyskinesia emerges, the levodopa dose should be reduced. In patients who already have dyskinesia, the

    levodopa dose often is reduced by 30-50% at the time tolcapone is introduced. Entacapone is a COMT inhibitor that does not cause hepatotoxicity; liver function tests are not required with this

    medication. A combination tablet of levodopa/carbidopa/entacapone is now available.

    Levodopa/PDI, dopamine agonists, and anticholinergics each provide good benefit for tremor in approximately

    50% of patients. If a patient is experiencing troublesome tremor and symptoms are not controlled adequately

    with one medication, another should be tried. If the tremor is not controlled adequately with medication

    thalamotomy or thalamic stimulation surgery may be considered at any time during the disease.

    For patients who have motor fluctuations and dyskinesia that cannot be adequately managed with medication

    manipulation, surgery is considered.

    Surgical Care

    Stereotactic surgery has made a resurgence in the treatment of Parkinson disease, largely due to long-termcomplications of levodopa therapy resulting in significant disability despite optimal medical management.

    A better understanding of basal ganglia physiology and circuitry and improvements in surgical techniques, neuroimaging

    and electrophysiologic recording have allowed surgical procedures to be performed more accurately and with lowe

    morbidity. Deep brain stimulation (DBS) has become the surgical procedure of choice for Parkinson disease because it

    does not involve destruction of brain tissue, it is reversible, it can be adjusted as the disease progresses or adverse

    events occur, and bilateral procedures can be performed without a significant increase in adverse events.

    Deep brain stimulation

    o DBS is an FDA-approved treatment for Parkinson disease.

    o The DBS system consists of a lead that is implanted into the targeted brain structure (thalamus, globus

    pallidus interna, subthalamic nucleus). The lead is connected to an implantable pulse generator (IPG)

    which is the power source of the system that is generally implanted in the subclavicular region of the

    chest cavity. The lead and the IPG are connected by an extension wire that is tunneled down the neck

    under the skin.

    o DBS provides monopolar or bipolar electrical stimulation to the targeted brain area. Stimulation

    amplitude, frequency, and pulse width can be adjusted to control symptoms and eliminate adverse

    events. The patient can turn the stimulator on or off using an Access Review Therapy Controller or a

    handheld magnet. The usual stimulation parameters are amplitude of 1-3 V, frequency of 135-185 Hz,

    and pulse width of 60-120 microseconds.

    o DBS has been proposed to work by resetting abnormal firing patterns in the brain leading to a reduction

    in parkinsonian symptoms.

    o The response from DBS is only as good as the patient's best "on" time with the exception of tremorwhich may have greater improvement than with medication; however, after DBS, the amount of daily

    "on" time is significantly extended.

    o DBS requires regular follow-up to adjust stimulation parameters to account for symptom changes due to

    disease progression and adverse effects.

    Thalamic stimulation

    o Thalamic stimulation involves implantation of a DBS lead in the ventral intermediate (VIM) nucleus of the

    thalamus.

    o Thalamic stimulation provides significant control of Parkinson disease tremor but does not affect the

    other symptoms of Parkinson disease such as rigidity, bradykinesia, dyskinesia, or motor fluctuations.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    14/31

    o Studies of thalamic DBS have demonstrated good initial and long-term tremor control up to 7 years after

    implantation; however, long-term studies have shown a significant worsening in other parkinsonian

    symptoms such as bradykinesia and rigidity and worsening of gait leading to major disability.

    o Candidates for thalamic DBS are patients with disabling medication-resistant tremor who have minima

    rigidity or bradykinesia. They should not have significant cognitive impairment, mood or behaviora

    disturbances, or other factors that may increase the risk of surgery.

    o The role of thalamic DBS is limited in Parkinson disease.

    Pallidal stimulation

    o Pallidal stimulation involves implantation of a DBS lead in the globus pallidus interna (GPi).

    o Pallidal stimulation significantly controls all the cardinal symptoms of Parkinson disease (tremor, rigidity

    bradykinesia) as well as dyskinesia.

    o Long-term studies up to 4 years after pallidal DBS have continued to show significant improvements in

    the cardinal features of Parkinson disease and dyskinesia compared with presurgery.

    o Candidates for pallidal DBS include levodopa-responsive patients with medication-resistant disabling

    motor fluctuations and/or levodopa-induced dyskinesia without significant cognitive impairment

    behavioral issues, or mood problems.

    Subthalamic stimulation

    o Subthalamic stimulation is currently the most common surgical procedure for Parkinson disease and

    involves implantation of a DBS lead in the subthalamic nucleus (STN).

    o STN DBS controls all of the cardinal symptoms of Parkinson disease as well as motor fluctuations and

    dyskinesia. STN DBS also often results in significant reductions in antiparkinsonian medications.

    o On average, dyskinesia and antiparkinsonian medications are reduced by 50-80%.

    o Multiple studies have examined the effects of STN DBS and have shown significant improvements in the

    motor symptoms of tremor, rigidity, and bradykinesia as well as activities of daily living.

    o Long-term follow-up reports have demonstrated that significant improvements in motor function and

    activities of daily living are maintained for up to 5 years after surgery.

    o Candidates for STN DBS include levodopa-responsive patients with medication-resistant disabling moto

    fluctuations and/or levodopa-induced dyskinesia without significant cognitive impairment, behaviora

    issues, or mood problems.

    Pallidal stimulation versus subthalamic stimulation

    o No large controlled trials have been completed comparing STN and GPi stimulation; however, a large

    well-designed study is currently underway.

    o Several small uncontrolled studies have compared STN and GPi stimulation. Most studies have showngreater improvement after STN DBS compared with GPi DBS, and antiparkinsonian medications were

    reduced only after STN DBS. Therefore, STN DBS is currently the surgical procedure of choice for

    Parkinson disease.

    Complications of DBS

    o Complications can be separated into surgical complications occurring within 30 days of the procedure;

    complications related to the components of the DBS system; and complications from the stimulation

    which generally can be resolved by adjustments of the stimulation parameters.

    o Surgical complications are comparable to those seen with other neurosurgical procedures. Serious

    adverse events such hemorrhage, ischemic lesions, seizures, or death occur in 1-2% of patients

    Infection occurs in approximately 3-5% of patients and may require explantation of the device until the

    infection is resolved.o Misplacement of the lead may also occur in approximately 10% of patients and require additiona

    surgery to correct lead placement.

    o Device-related complications include malfunction of the IPG, displacement of the lead, skin erosion, and

    device fractures. These complications can occur in up to 25% of patients and generally require

    additional surgery.

    o Stimulation side effects include paresthesia, muscle spasms, visual disturbances, mood changes, and

    pain. These side effects are generally easily resolved with adjustments to the stimulation parameters.

    o Although not considered a complication, the IPG (battery) is generally replaced every 3-5 years and

    requires additional outpatient surgery.

    Lesion surgeries involve the destruction of targeted areas of the brain to control the symptoms of Parkinson

    disease.

    o Lesion surgeries for Parkinson disease have largely been replaced by DBS.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    15/31

    o Thalamotomy involves destruction of a part of the thalamus, generally the ventral intermediate (VIM)

    nucleus, to relieve tremor. Thalamotomy has little effect on bradykinesia, rigidity, motor fluctuations, o

    dyskinesia.

    o Most patients with Parkinson disease who undergo thalamotomy have significant improvement in tremo

    of the limbs contralateral to the side of the lesion.

    o Bilateral thalamotomy is generally avoided because complications, especially speech and cognitive

    impairment, are common.

    o Pallidotomy involves destruction of a part of the GPi, which is overactive in Parkinson disease.

    o Pallidotomy studies have demonstrated significant improvements in each of the cardinal symptoms of

    Parkinson disease (tremor, rigidity, bradykinesia) as well as a significant reduction in dyskinesia.o Bilateral pallidotomy is not recommended because complications are relatively common and include

    speech difficulties, dysphagia, and cognitive impairment.

    o Subthalamotomy involves destruction of a part of the STN, which is also hyperactive in Parkinson

    disease.

    o Subthalamotomy studies have shown significant improvements in the cardinal features of Parkinson

    disease as well as the reduction of motor fluctuations and dyskinesia.

    Transplantation

    o Neural transplantation is a potential treatment for Parkinson disease because the neuronal degeneration

    is site and type specific (ie, dopaminergic), the target area is well defined (ie, striatum), postsynaptic

    receptors are relatively intact, and the neurons provide tonic stimulation of the receptors and appear to

    serve a modulatory function.

    o Multiple sources of dopamine-producing cells, including fetal nigral cells, sympathetic ganglia, carotid

    body glomus cells, PC-12 cells, and neuroblastoma cells, have been studied.

    o Transplantation of autologous adrenal medullary cells and fetal porcine cells were not found to be

    effective in double-blind studies and have been abandoned.

    o A double-blind study of GDNF demonstrated that it was not superior to placebo in controlling the

    symptoms of Parkinson disease and, therefore, due to the lack of benefit and concerns regarding

    adverse events, clinical trials have been discontinued.

    o Double-blind studies demonstrated that transplanted fetal mesencephalic cells can survive

    transplantation. However, these studies showed only minimal benefit in measures of Parkinson disease

    symptoms and often resulted in the development of dyskinesia even in the absence of antiparkinsonian

    medications.

    o In double-blink studies, transplanted cultured human retinal pigment epithelial cells (RPE) were found tobe no different than sham surgery after 12 months of follow-up.

    o Several studies have recently demonstrated the safety of gene therapy as a treatment for Parkinson

    disease and larger studies have been initiated to examine the efficacy of this procedure. The goal of

    these studies is to modify genes involved in the development of Parkinson disease.

    o In the laboratory, the use of stem cells is being investigated.

    For more information, see eMedicine article Surgical Treatment of Parkinson Disease.

    Consultations

    Consider physical therapy, occupational therapy, and speech therapy consultations.

    Consider neurosurgical consultation for patients with medically refractory tremor or troublesome dyskinesia

    and/or motor fluctuations that cannot be controlled with medication adjustments. Patients with dementia o

    significant psychiatric or behavioral problems are not candidates for the current neurosurgical treatments fo

    Parkinson disease.

    Medication

    The cornerstone of symptomatic treatment for Parkinson disease is dopamine replacement therapy.

    The criterion standard of symptomatic therapy is levodopa (L-dopa), the metabolic precursor of dopamine, in combination

    with a peripheral decarboxylase inhibitor (PDI). This combination provides the greatest symptomatic benefit with the

    fewest short-term adverse effects.

  • 8/2/2019 Emedicine Parkinsons Diseasae

    16/31

    Anticholinergic drugs can be used as an alternative to L-dopa for treating resting tremor. However, they are not highly

    effective against bradykinesia, gait disturbance, or other features of advanced parkinsonism.

    Dopamine agonists (bromocriptine, pergolide, pramipexole, ropinirole) can be used as monotherapy to improve

    symptoms in early disease or as adjuncts to levodopa in patients whose response to L-dopa is deteriorating and those

    who are experiencing fluctuations in their response to L-dopa.

    MAO-B inhibitors provide symptomatic benefit as monotherapy in early disease and as adjuncts to levodopa in patients

    experiencing motor fluctuations.

    Rasagiline is a second-generation MAO-B inhibitor. Unlike selegiline, rasagiline does not have amphetamine metabolites

    It is effective as monotherapy in early Parkinson disease and as an adjunct to levodopa in patients with moto

    fluctuations. Recommended dose is 1 mg/d.

    COMT inhibitors increase the peripheral half-life of levodopa, thereby delivering more levodopa to the brain over a longer

    time.

    Dopamine prodrugs

    Dopamine does not cross the blood-brain barrier, but levodopa does. L-dopa is decarboxylated to dopamine in the brain

    and in the periphery. The formation of dopamine in the blood causes many of L-dopa's adverse effects.

    When administered alone, levodopa induces a high incidence of nausea and vomiting. A PDI such as carbidopa iscombined with levodopa to reduce the incidence of nausea and vomiting by inhibiting the peripheral conversion o

    levodopa to dopamine.

    Levodopa/PDI is the criterion standard of symptomatic treatment for Parkinson disease; it provides the greates

    antiparkinsonian efficacy in moderate to advanced disease with the fewest acute adverse effects.

    Levodopa/Carbidopa (Sinemet, Sinemet CR, Parcopa)

    Large, neutral amino acid that is absorbed in proximal small intestine by saturable carrier-mediated transport system

    Absorption decreased by meals, which include other large neutral amino acids. Only patients with meaningful moto

    fluctuations must consider a low-protein or protein-redistributed diet. Greater consistency of absorption achieved when

    levodopa taken 30 min or more before or 1 h or more after meals. Nausea often reduced if L-dopa taken immediatelyfollowing meals. Some patients with nausea benefit from additional carbidopa in doses up to 200 mg/d

    No maximal dose per se. Patients should receive lowest dose that provides good control of parkinsonian symptoms. If

    parkinsonian disability present, dose should be escalated until adequate control achieved or adverse effects become

    intolerable. Some patients require 2000 mg or more per d

    Half-life of levodopa/carbidopa approximately 2.5 h

    CR formulation more slowly absorbed and provides more sustained levodopa levels than immediate release form. CR

    form as effective as immediate release form when levodopa first required and may be more convenient when fewer

    intakes are required. Patients with wearing-off motor fluctuations (and no dyskinesia) often benefit from prolongation of

    short duration response when switched from immediate release to CR form. However, patients with meaningfu

    fluctuations and dyskinesia often experience increase in dyskinesia when switched to CR form. To convert patient from

    immediate release to CR form, increase daily dosage by approximately 20% while number of intakes reduced by 30-50%

    Most patients controlled on levodopa dose of 300-600 mg for several years.

    Dosing

    Adult

    Immediate-release form: 25 mg carbidopa/100 mg levodopa one half tab PO qd; increase daily dose by one half tab per

    wk to initial maintenance dose of 25/100 mg tid; may increase by 1 tab qd each wk until optimal clinical response

    achieved

    Parcopa form: 25 mg carbidopa/100 mg levodopa or 25 mg carbidopa/250 mg levodopa; dissolves in mouth, may take

    prn or scheduled if swallowing tab is difficult or water not available

    CR form: 1 tab PO qd; increase daily dose by 1 tab each wk to achieve initial maintenance dose of 25/100 mg tid or

    50/200 mg bid

  • 8/2/2019 Emedicine Parkinsons Diseasae

    17/31

    Pediatric

    Not established

    Interactions

    Hydantoins, pyridoxine, phenothiazine, and hypotensive agents may decrease effects of levodopa; concurrent antacids or

    nonspecific MAOIs increase levodopa toxicity

    Contraindications

    Documented hypersensitivity, narrow-angle glaucoma' malignant melanoma relative contraindication; if meaningfu

    parkinsonian disability present, consider benefit/risk ratio

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Most common acute adverse effects are nausea, hypotension, or hallucinations; long-term adverse effects include motor

    fluctuations and dyskinesia (chorea); for patients experiencing motor fluctuations, dietary protein can be distributed evenly

    throughout day or redistributed to evening to minimize fluctuations in levodopa absorptionAbrupt withdrawal of treatment may result in neuroleptic malignant syndrome (NMS); use cautiously in patients with

    history of MI, arrhythmias, asthma, or peptic ulcer disease

    Dopamine agonists

    Dopamine agonists directly stimulate postsynaptic dopamine receptors to provide antiparkinsonian benefit. All available

    dopamine agonists stimulate D2 receptors--an action that is thought to be clinically beneficial. The role of other dopamine

    receptors is currently unclear.

    Dopamine agonists are effective as monotherapy in early Parkinson disease and as adjuncts to levodopa/PDI in

    moderate to advanced disease. They provide antiparkinsonian efficacy approximately equal to levodopa/PDI when

    symptomatic therapy is first required.

    After 6 months to a few years, they are not as effective as levodopa/PDI. For patients with motor fluctuations on

    levodopa/PDI, the addition of a dopamine agonist reduces off time, improves motor function, and allows lower levodopa

    doses.

    Dopamine agonists have been proven to reduce the development of motor fluctuations and dyskinesias when used as an

    initial therapy and continued once levodopa is added.

    Dopamine agonists may slow disease progression based on changes in PET scans, but the evidence is not ye

    conclusive.

    Apomorphine (Apokyn)

    Short-acting dopamine agonist approved in the United States for SC injection as a rescue agent to treat acute immobility

    episodes (hypomobility or "off-periods") in PD. These episodes consist of inability to rise from a chair, speak, or walk and

    may occur toward the end of the dose interval or may be spontaneous and unpredictable in onset.

    Dosing

    Adult

    Dosage is individualized

    Test dose: 2 mg (0.2 mL) SC for 1 dose initially during hypomobility, if tolerated (ie, blood pressure remains stable), may

    use for subsequent hypomobility episodes

    Establishing dose: If patient tolerates test dose and hypomobility responds, 2 mg is the dose to use for subsequent

  • 8/2/2019 Emedicine Parkinsons Diseasae

    18/31

    hypomobility episodes

    If patient tolerates test dose, but hypomobility does not respond to test dose, may increase dose by 1 mg (0.1 mL) q2-3 d

    until response is observed; not to exceed 6 mg (0.6 mL)/dose

    Note: Administer only 1 dose per hypomobility episode, do not repeat dose; administer with antiemetic drug

    Pediatric

    Not established

    Interactions

    Coadministration with 5HT3 antagonists used for emesis or irritable bowel syndrome (eg, ondansetron, dolasetrongranisetron, palonosetron, alosetron) may cause hypotension and loss of consciousness; coadministration with drugs tha

    increase QTC interval (eg, thioridazine, quinidine, sotalol, erythromycin, dofetilide) may increase arrhythmia potential

    metabolized by catechol-o-methyltransferase (COMT), coadministration with COMT inhibitors (eg, entacapone

    tolcapone) may decrease elimination

    Contraindications

    Documented hypersensitivity to apomorphine or metabisulfite

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Causes severe nausea and vomiting and must be administered with an antiemetic drug (but not with antiemetic agents

    that are 5HT3 antagonists); may cause orthostatic hypotension, faintness, hallucinations, fluid retention, chest pain

    increased sweating, flushing, pallor, dyskinesia, rhinorrhea, and extreme drowsiness (may fall asleep during waking hours

    without warning)

    Bromocriptine (Parlodel)

    Semisynthetic ergot alkaloid derivative that is strong D2 receptor agonist and weak D1 receptor antagonist. FDAapproved as adjunct to levodopa/carbidopa; less effective than other dopamine agonists. May relieve akinesia, rigidity

    and tremor in PD. Mechanism of therapeutic effect is direct stimulation of dopamine receptors in corpus striatum

    Approximately 28% absorbed from GI tract and metabolized in liver. Elimination half-life approximately 50 h with 85%

    excreted in feces and 3-6% eliminated in urine

    Initiate at low dosage and individualize. Increase daily dosage slowly until maximum therapeutic response achieved. If

    possible, maintain the dosage of levodopa during this introductory period. Assess dosage titrations q2wk to ensure tha

    lowest dosage producing optimal therapeutic response is not exceeded. If adverse reactions mandate, reduce dose

    gradually in 2.5-mg increments.

    Dosing

    Adult

    1.25 mg (one half of a 2.5 mg tab) PO qd; increase by 1.25 mg/d per wk to 1.25 mg tid with meals; increase q2-4wk by

    2.5 mg/d with meals; usual range 10-40 mg/d divided tid/qid; safety has not been demonstrated in dosages that exceed

    100 mg/d

    Pediatric

    Not established

    Interactions

    Ergot alkaloids increase toxicity; amitriptyline, butyrophenone, imipramine, methyldopa, phenothiazine, and reserpine

    may decrease effects

  • 8/2/2019 Emedicine Parkinsons Diseasae

    19/31

    Contraindications

    Documented hypersensitivity, ischemic heart disease, peripheral vascular disorders

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Adverse effects include nausea, hypotension, hallucinations, and somnolence; use cautiously in patients with renal o

    hepatic disease

    Pergolide (Permax)

    Pergolide was withdrawn from the US market March 29, 2007, because of heart valve damage resulting in cardiac

    valve regurgitation. It is important not to abruptly stop pergolide. Health care professionals should assess patients' need

    for dopamine agonist (DA) therapy and consider alternative treatment. If continued treatment with a DA is needed

    another DA should be substituted for pergolide. For more information, see FDA MedWatch Product Safety Alert and

    Medscape Alerts: Pergolide Withdrawn From US Market

    Potent dopamine receptor agonist at both D1 and D2 receptor sites, approximately 10 times more potent than

    bromocriptine on a mg per mg basis. In PD, pergolide believed to exert its therapeutic effect by directly stimulatingpostsynaptic dopamine receptors in striatum

    Usually administered in divided doses tid.

    Dosing

    Adult

    0.05 mg PO qd days 1 and 2; gradually increase by 0.1 or 0.15 mg/d q3d over next 12 d, followed by incrementa

    increases of 0.25 mg/d q3d until optimal therapeutic dose achieved; usual maximum dose 3-6 mg/d; usually administered

    in divided doses tid

    Pediatric

    Not established

    Interactions

    Concurrent use of pergolide and levodopa may cause or exacerbate preexisting states of confusion and hallucinations o

    dyskinesia

    Dopamine antagonists such as neuroleptics (eg, phenothiazine, butyrophenone, thioxanthenes, metoclopramide) may

    diminish effectiveness of pergolide; because pergolide mesylate is >90% bound to plasma proteins, exercise caution in

    coadministering with other drugs known to affect protein binding

    Contraindications

    Documented hypersensitivity

    Precautions

    Pregnancy

    B - Fetal risk not confirmed in studies in humans but has been shown in some studies in animals

    Precautions

    May cause valvular heart disease (yearly echocardiograms recommended for patients on chronic therapy); inhibits

    secretion of prolactin; causes transient rise in serum concentrations of growth hormone and decrease in serum

    concentrations of luteinizing hormone; adverse effects include nausea, hypotension, hallucinations, and somnolence; use

    caution in patients who have been treated for cardiac dysrhythmias; may cause or exacerbate preexisting states of

    confusion and hallucinations or dyskinesia

  • 8/2/2019 Emedicine Parkinsons Diseasae

    20/31

    Pramipexole (Mirapex)

    Nonergot dopamine agonist with specificity for D2 dopamine receptor. Also binds to D3 and D4 receptors. Readily

    absorbed from GI tract with >90% bioavailability, minimally metabolized in liver with half-life of approximately 8-12 h.

    Primarily excreted in urine; for patients with CrCl 35-60 mL/min, administer bid (max 1.5 mg bid); for CrCl 15-35 mL/min

    administer qd (not to exceed 1.5 mg/d)

    FDA approved as monotherapy in early disease and as adjunct to levodopa/PDI in more advanced stages.

    Dosing

    Adult

    Week 1: 0.125 mg PO tid; week 2: 0.25 mg tid; week 3: 0.5 mg tid; continue escalating by 0.25 mg tid each week as

    clinically appropriate; usual range 1.54.5 mg/d

    Pediatric

    Not established

    Interactions

    Cimetidine may increase toxicity; increases levels of levodopa if given concurrently

    Contraindications

    Documented hypersensitivity

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Adverse effects include nausea, hallucinations, and somnolence; somnolence may emerge even after administration a

    stable dose for many months; some patients experience relatively sudden waves of irresistible sleepiness; patients

    should be warned not to drive if experiencing drowsiness; somnolence usually resolves with dose reduction o

    discontinuation; use caution in patients with renal insufficiency and preexisting dyskinesias

    Ropinirole (Requip and Requip XL)

    Nonergot dopamine agonist that has high relative in vitro specificity and full intrinsic activity at D2 subfamily of dopamine

    receptors; binds with higher affinity to D3 than to D2 or D4 receptor subtypes. Has moderate affinity for opioid receptors,

    and its metabolites have negligible affinity for dopamine D1, 5HT 1, 5HT 2, benzodiazepine, GABA, muscarinic, alpha 1-

    alpha 2- and beta-adrenoreceptors. Mechanism of action is stimulation of dopamine receptors in striatum

    When administered as adjunct to levodopa, concurrent dose of levodopa may be decreased gradually as tolerated. FDA

    approved as monotherapy in early disease and as adjunct to levodopa/PDI in more advanced diseaseReadily absorbed from GI tract with 55% bioavailability and metabolized to inactive metabolites in liver by CYP1A2. Half-

    life approximately 6 h with inactive metabolites primarily excreted in urine.

    Dosing

    Adult

    Ropinirole Week 1: 0.25 mg PO tid; week 2: 0.5 mg tid; week 3: 0.75 mg tid; after week 4, if necessary, increase by 1.5

    mg/d on a weekly basis up to 9 mg/d, and then by 3 mg/d weekly to total dose as high as 24 mg/d; discontinue gradually

    over 7-d period; decrease frequency of administration from tid to bid for 4 d; for remaining 3 d, decrease frequency to qd

    prior to complete withdrawa

    Ropinirole XL: Start at 2 mg/d for 2 wk and then increase by 2 mg q2wk until desired response achieved or dose reaches

    24 mg/d; discontinue gradually over 7-d period

  • 8/2/2019 Emedicine Parkinsons Diseasae

    21/31

    Pediatric

    Interactions

    Estrogens may reduce clearance by 36% (adjust ropinirole dose if estrogen therapy stopped or started during treatment)

    substrates or inhibitors of CYP1A2 (eg, quinolone antibiotics, erythromycin, cimetidine, diltiazem, fluvoxamine, mexiletine

    tacrine) may alter clearance (adjust ropinirole dose if therapy with potent CYP1A2 inhibitor stopped or started during

    treatment); dopamine antagonists (eg, phenothiazines, butyrophenones, thioxanthenes, metoclopramide, neuroleptics)

    may diminish effectiveness; CNS depressants may have additive sedative effects

    Contraindications

    Documented hypersensitivity

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Adverse effects include nausea, hypotension, hallucinations, and somnolence; patients should be warned not to drive i

    experiencing drowsiness; somnolence usually resolves with dose reduction or discontinuation

    Dopamine receptor agonists may potentiate dopaminergic effects of levodopa and may cause or exacerbate preexistingdyskinesia; decreasing dose of levodopa may ameliorate this effec

    Cases of retroperitoneal fibrosis, pulmonary infiltrates, pleural effusion, and pleural thickening have been reported; these

    complications do not always resolve completely upon drug cessation

    Use caution in patients taking CNS depressants; monitor for signs and symptoms of orthostatic hypotension

    Cases of rhabdomyolysis have been reported

    Rotigotine (Neupro)

    April 2008: A recall was issued for Neupro patch in the United States because of crystal formation in the patch

    resulting in decreased dopamine absorption transdermally. As of August 1, 2008, the patch is still unavailable, although

    the manufacturer is working to correct the defect and hopefully return it to the market. For more information, see

    Medscape News

    Dopamine agonist stimulating D3, D2, and D1 receptors. Improvement in Parkinson-related symptoms thought to be its

    ability to stimulate D2 receptors within the caudate putamen in the brain. Available as a transdermal patch that provides

    continuous delivery for 24 h (2 mg/24 h [10 cm 2], 4 mg/24 h [20 cm2], or 6 mg/24 h [30 cm2]). Indicated for symptoms o

    early Parkinson disease.

    Dosing

    Adult

    2 mg/24 h (10 cm2) transdermal qd initially; may increase qwk by 2 mg/24 h, not to exceed 6 mg/24 h

    Remove previous day's patch before applying new patch; rotate application site each day between left and right sides of

    body and upper and lower parts of body

    Pediatric

    Indication not applicable to children

    Interactions

    Dopamine antagonists (eg, antipsychotics, metoclopramide) may decrease effect

    Contraindications

    Documented hypersensitivity

    Precautions

  • 8/2/2019 Emedicine Parkinsons Diseasae

    22/31

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Common adverse effects include dermal reactions at patch site, dizziness, nausea, vomiting, drowsiness, and insomnia;

    less common adverse effects that may be hazardous to patient include sudden sleep onset, hallucinations, and postura

    hypotension; weight gain secondary to fluid retention has been observed; rapid dose reduction or abrupt withdrawal may

    cause hyperpyrexia and confusion; apply to clean, dry, and intact skin on abdomen, thigh, hip, flank, shoulder, or upper

    arm

    Anticholinergics

    These agents provide benefit for tremor in approximately 50% of patients but do not improve bradykinesia or rigidity. If

    one anticholinergic does not work, try another.

    Trihexyphenidyl (Artane, Trihexy)

    Synthetic tertiary amine anticholinergic agent, reduces incidence and severity (by 20%) of akinesia, rigidity, and tremor

    and secondary symptoms such as drooling. In addition to suppressing central cholinergic activity, also may inhibi

    reuptake and storage of dopamine at central dopamine receptors, thereby prolonging action of dopamine.

    Dosing

    Adult

    1-2 mg/d PO; increase by 2 mg/d at intervals of 3-5 d; usual range 4-15 mg/d divided tid/qid; young adults may tolerate

    15-20 mg/d divided tid/qid; older individuals may tolerate no more than 4-8 mg/d

    Pediatric

    Interactions

    Decreases effects of levodopa; increases effects of narcotic analgesics, phenothiazines, tricyclic antidepressants

    quinidine, and anticholinergics

    Contraindications

    Documented hypersensitivity; glaucoma, particularly angle-closure glaucoma; pyloric or duodenal obstruction, stenosing

    peptic ulcers; prostatic hypertrophy or bladder neck obstructions; achalasia (megaesophagus); myasthenia gravis

    megacolon

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    PrecautionsAdverse effects include dry mouth and dry eyes, memory difficulty, confusion, and rarely urinary retention; use caution in

    patients with tachycardia, cardiac arrhythmias, hypertension, hypotension, prostatic hypertrophy (particularly in elderly),

    or any tendency toward urinary retention, liver or kidney disorders, or obstructive disease of GI or GU tract; when used to

    treat extrapyramidal reactions that result from phenothiazines in psychiatric patients, antiparkinson agents may

    exacerbate mental symptoms and precipitate toxic psychosis

    Benztropine mesylate (Cogentin)

    Partially blocks striatal cholinergic receptors to help balance cholinergic and dopaminergic activity.

    Dosing

  • 8/2/2019 Emedicine Parkinsons Diseasae

    23/31

    Adult

    0.5-6 mg/d PO qd or divided bid; start elderly patients at lower dose; titrate in 0.5-mg increments at 5- to 6-d intervals; not

    to exceed 6 mg/d

    Pediatric

    Interactions

    Decreases effects of levodopa; increases effects of narcotic analgesics, phenothiazines, tricyclic antidepressants

    quinidine, and anticholinergics

    Contraindications

    Documented hypersensitivity; glaucoma, particularly angle-closure glaucoma; pyloric or duodenal obstruction, stenosing

    peptic ulcers; prostatic hypertrophy or bladder neck obstructions; achalasia (megaesophagus); myasthenia gravis

    megacolon

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Adverse effects include dry mouth and dry eyes, memory difficulty, confusion, and rarely urinary retention; use cautiously

    in patients with tachycardia, cardiac arrhythmias, hypertension, hypotension, prostatic hypertrophy (particularly in elderly)

    or any tendency toward urinary retention, liver or kidney disorders, or obstructive disease of GI or GU tract; when used to

    treat extrapyramidal reactions that result from phenothiazines in psychiatric patients, antiparkinson agents may

    exacerbate mental symptoms and precipitate toxic psychosis

    MAO-B inhibitors

    These agents inhibit the activity of MAO-B oxidases that are responsible for inactivating dopamine and possibly the

    conversion of compounds into neurotoxic types.

    Selegiline (Eldepryl)

    An irreversible inhibitor of MAO, it acts as a "suicide" substrate for enzyme; MAO converts it to an active moiety which

    combines irreversibly with active site or enzyme's essential FAD cofactor. Blocks breakdown of dopamine and extends

    duration of action of each dose of L-dopa. Often allows L-dopa dose reduction that is needed for optimal effect. Because

    selegiline has greater affinity for type B than for type A active sites, it can serve as selective inhibitor of MAO type B a

    recommended dose. However, doses higher than 10 mg/d may inhibit MAO-A sites significantly. Its metabolites

    amphetamine and methamphetamine, may inhibit dopamine reuptake and enhance dopamine release

    FDA approved as adjunct to levodopa/carbidopa in patients who exhibit deterioration in response to that therapy. Fo

    patients who are experiencing motor fluctuations on levodopa/carbidopa, addition of selegiline reduces off time, improves

    motor function, and allows levodopa dose reductions. If patient experiences increase in troublesome dyskinesia, reduce

    levodopa dose

    Rapidly absorbed and has 73% bioavailability. Metabolized in liver to N -desmethylselegiline, L-amphetamine, and L

    methamphetamine. Half-life approximately 10 h; metabolites excreted in urine

    Because inhibition of MAO-B is irreversible, loss of activity is function of new protein synthesis and may last severa

    months. No evidence of additional benefit from doses >10 mg/d

    After 2-3 days of treatment, attempt to reduce dose of levodopa/carbidopa. Reduction of 10-30% appears typical. Further

    reductions of levodopa/carbidopa may be possible during continued selegiline therapy.

    Dosing

    Adult

    5 mg PO bid with breakfast and lunch; not to exceed 10 mg/d

    Pediatric

  • 8/2/2019 Emedicine Parkinsons Diseasae

    24/31

    Interactions

    Concurrent meperidine may cause stupor, muscular rigidity, severe agitation, and elevated temperature; concurren

    tricyclic or serotonin reuptake inhibitor antidepressant may cause severe toxicity; one case of hypertensive crisis in a

    patient taking selegiline and ephedrine has been reported

    Contraindications

    Documented hypersensitivity; concomitant meperidine or other opioids; concomitant tricyclic or serotonin reuptake

    inhibitor antidepressants (relative contraindication)

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    Risks associated with dose >10 mg/d are associated with nonselective inhibition of MAO; concurrent tyramine-containing

    foods and other indirect-acting sympathomimetics may cause hypertensive crisis

    Rasagiline (Azilect)

    Irreversible MAO-B inhibitor that blocks dopamine degradation. Not metabolized to amphetamine derivatives. Main

    metabolite, aminoindan, has some activity and has been shown to improve motor and cognitive functions in experimenta

    models. Indicated for Parkinson disease as initial monotherapy or as adjunctive therapy with levodopa.

    Dosing

    Adult

    Monotherapy: 1 mg PO qd

    Adjunctive therapy with levodopa: 0.5 mg PO qd; may increase to 1 mg PO qd

    Mild hepatic impairment or coadministration with CYP1A2 inhibitors: 0.5 mg PO qd

    Pediatric

    Interactions

    P450 CYP1A2 substrate; coadministration with drugs that inhibit CYP1A2 (eg, cimetidine, clarithromycin, erythromycin)

    may decrease elimination and increase toxicity; coadministration with TCAs, SSRIs, serotonin-norepinephrine reuptake

    inhibitors (SNRIs), nonselective MAOIs, or selective MAO-B inhibitors has caused severe CNS toxicity associated with

    hyperpyrexia and death; consuming tyramine-rich foods (eg, cheese, red wine, beer, sausage, avocado) may cause

    hypertensive crisis; also see Contraindications

    Contraindications

    Documented hypersensitivity; moderate-to-severe hepatic impairment (Child-Pugh score >6); concurrent use with

    meperidine, tramadol, methadone, propoxyphene; dextromethorphan, St. John's wort, mirtazapine, cyclobenzaprine

    sympathomimetic amines (eg, pseudoephedrine, cocaine, ephedrine), other MAOIs, or local anesthetics containingepinephrine; pheochromocytoma

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    Precautions

    May cause dyskinesias, hallucinations, or hypotension; if emergent surgery is necessary, benzodiazepines, mivacurium,

    rapacuronium, fentanyl, morphine, or codeine may be used cautiously; melanoma may develop more frequently in those

    taking rasagiline than in matched controls

  • 8/2/2019 Emedicine Parkinsons Diseasae

    25/31

    N-methyl-D-aspartic acid inhibitors

    Increases dopaminergic activity in peripheral and central nervous system by augmenting dopamine release and inhibiting

    cellular reuptake.

    Amantadine (Symmetrel)

    Inhibits the N -methyl-D-aspartic acid (NMDA) receptor-mediated stimulation of acetylcholine release in rat striatum. May

    also enhance dopamine release, inhibit dopamine reuptake, stimulate postsynaptic dopamine receptors, or enhance

    dopamine receptor sensitivity. Efficacy as monotherapy and as adjunct to levodopa/PDI in treating PD. Provides some

    benefit for tremor, rigidity, and bradykinesia

    Readily and almost completely absorbed from GI tract; is not metabolized. Half-life approximately 9-37 h and prolonged in

    renal insufficiency. Excreted 90% unchanged in urine.

    Dosing

    Adult

    100 mg PO in am; increase by 100 mg/d each wk prn; not to exceed 100 mg qid

    Pediatric

    Interactions

    Drugs with anticholinergic or CNS stimulant activity increase toxicity; concurrent hydrochlorothiazide plus triamterene may

    decrease urinary excretion of amantadine with subsequent increased plasma concentrations

    Contraindications

    Documented hypersensitivity

    Precautions

    Pregnancy

    C - Fetal risk revealed in studies in animals but not established or not studied in humans; may use if benefits outweigh

    risk to fetus

    PrecautionsCommon adverse effects are confusion and hallucinations; use caution in patients with liver disease, history of recurren

    and eczematoid dermatitis, uncontrolled psychosis, seizures, and in those receiving CNS stimulant drugs; reduce dose in

    renal disease when treating PD; do not discontinue medication abruptly

    Catechol-o-methyltransferase (COMT) inhibitors

    These agents inhibit the peripheral metabolism of levodopa, making more levodopa available for transport across the

    blood-brain barrier over a longer time. For patients with motor fluctuations on levodopa/carbidopa, the addition of a COMT

    inhibitor decreases off time, improves motor function, and allows lower levodopa doses. Patients who already have

    dyskinesia on levodopa/PDI are likely to experience a worsening of dyskinesia, thereby necessitating a levodopa dose

    reduction. In such patients, consider reducing levodopa dose at the time of introduction, especially with tolcapone.

    Tolcapone (Tasmar)

    Adjunct to levodopa/carbidopa therapy in PD. Mechanism related to its ability to inhibit COMT and alter plasma

    pharmacokinetics of levodopa. When tolcapone given in conjunction with levodopa and an aromatic amino acid

    decarboxylase inhibitor (eg, carbidopa), plasma levels of levodopa are more sustained than after administration o

    levodopa and an aromatic amino acid decarboxylase inhibitor alone. These sustained plasma levels of levodopa may

    result in more constant dopaminergic stimulation in brain, possibly leading to greater effects on signs and symptoms of

    PD as well as increased adverse effects of levodopa (which sometimes require levodopa dose decrease). Enters CNS to

    a minimal extent but has been shown to inhibit central COMT activity in animals. FDA approved as adjunct to

    levodopa/carbidopa for patients who are experiencing motor fluctuations

    Because of risk of hepatotoxicity, it is reserved for patients who have not responded adequately to or are not appropriate

    candidates for other adjunctive medications. Patients should sign informed consent; strict liver function test monitoring

  • 8/2/2019 Emedicine Parkinsons Diseasae

    26/31

    required. If impro