Parkinson's Disease

Home ] Up ]


Pages Below:

Parkinson's Disease 


Definition

  • degenerative condition
  • primarily affects extrapyramidal pathways where dopamine is the neurotransmitter
  • characterised by
  • bradykinesia/akinesia
  • rigidity
  • tremor at rest
  • postural instability

Epidemiology

  • incidence 20 /100,000 /year
  • prevalence 200/100,000
  • affects approx 1:200 of the population >60
  • no significant gender bias
  • worldwide
    • appears to be more common in Europe and North America.

History

  • stage 1
    • pill-rolling tremor
    • micrographia
    • difficulty with fine movements
  • stage 2
    • poverty of blinking
    • impassive face
    • dribbling and swallowing difficulties
    • monotony of speech and ↓ volume of voice
  • stage 3
    • disorders of posture
    • flexion of neck
    • lack of arm swing
  • stage 4
    • loss of balance
    • lack of righting reflexes
    • retropulsion (??)
    • short steps
    • shuffling gait
    • festination
  • stage 5
    • pain
    • depression
    • taste disturbance
    • autonomic disturbance
      • constipation
      • postural hypotension
      • urinary frequency

Examination

  • observation
    • lack of facial expression
    • drooling of saliva
    • poor posture
      • stooping
      • slumped in chair
    • quiet, monotonous speech
  • Gait
  • Neuro
    • Bradykinesia
    • tone
      • rigidity
    • tremor
      • at rest
      • often asymmetrical
      • pill rolling
    • power
      • usually normal 
    • sensation
      • normal
    • reflexes
      • normal

Pathophysiology

  • results from the loss of inhibitory dopaminergic neurones 
  • these project from the substantia nigra of the mid-brain to the striatum of the basal ganglia (caudate nucleus and putamen).
  • Macroscopically 
    • loss of melanin pigmentation
  • microscopically 
    • severe neuronal loss demonstrated
    • remaining neurones often contain Lewy bodies
  • symptoms manifest when 60-80% of nigrostriatal dopaminergic neurones have been lost
  • Other fibres terminating in the corpus striatum include 
    • excitatory cholinergic nerves
    • adrenergic
    • serotonergic
  • overall effect in favour of excitatory cholinergic mechanisms

Causes of Parkinsonian Syndrome  

  • idiopathic nigrostriatal degeneration (Parkinson’s disease)  
  • post-encephalitic
  • drug induced
    • dopamine antagonists 
      • phenothiazines and other antipsychotic drugs 
    • depleting striatal dopamine
      • methyldopa
      • reserpine 
    • rare causes
      • manganese poisoning
      • carbon monoxide poisoning
      • cerebrovascular disease
      • Wilson’s disease
      • MPTP poisoning
        • MPTP is converted in neuronal mitochondria to a toxic free radical
        • specifically toxic to dopamine producing cells 

Differential Diagnosis

  • Idiopathic
    • Brain stem Lewy body Parkinson's disease
  • Secondary
    • Drugs
    • Head Trauma

Principles of treatment

  • levodopa
    • + dopa-decarboxylase inhibitors
    • levodopa / bensarizide
    • levodopa / carbidopa
  • MAO / COMT inhibitors
    • selegiline
    • entacapone
  • dopamine agonists
    • bromocriptine
    • pergolide
    • ropinirole
    • carbegoline
    • pramiprexole
    • apomorphine
  • anti-cholinergics
  • amantadine
restoring the dopaminergic-cholinergic balance in the striatum by
  • increasing dopamine effects with levodopa/dopamine agonists
  • blocking acetylcholine actions with anticholinergics
Although those with mild disease may start on anti-cholinergic drugs, the major disability of parkinsonian patients is hypokinesia with difficulty walking, dressing and eating. Where this occurs, levodopa is given, often in addition to anti-cholinergics.

Patients with Parkinson’s disease are more likely to be levodopa responsive.  Other causes are less likely to respond.

Physiotherapy, occupational therapy and social support are important adjuncts to treatment.

 

Drugs affecting the dopaminergic system.

 

Dopaminergic activity can be enhanced by the following methods:

 

1)      Administration of Levodopa with a peripheral dopa decarboxylase inhibitor.

2)      Increased release of endogenous dopamine.

3)      Stimulation of dopamine receptors.

4)      Inhibition of catechol-O-methyl transferase (COMT).

5)      Inhibition of monoamine oxidase type B (MAO-B).

 

Levodopa

As the immediate precursor of dopamine in the catecholamine synthesis, levodopa is decarboxylated in the brain to replenish striatal dopamine.  Dopamine itself cannot be given as it is not absorbed orally and does not cross the blood-brain barrier.  The nigrostriatal dopamine deficiency is therefore partially corrected and an improvement in symptoms of rigidity and bradykinesia are noted.

 

Levodopa is used in combination with a peripheral dopa-decarboxylase inhibitor, which reduces the adverse peripheral effects of dopamine.  The peripheral inhibitors are drugs which block peripheral levadopa decarboxylation.  These polar drugs cannot cross the blood-brain barrier and thus do not influence decarboxylation of levodopa in the brain.  This combined therapy allows a four to five fold decrease in the levodopa dose and the incidence of vomiting and arrhythmias is also reduced.  Central side effects such as hallucinations are not reduced by the combination therapy. 

 

Combined preparations (co-careldopa or co-beneldopa) are used in idiopathic Parkinson’s Disease.  However, levodopa is contraindicated in schizophrenia and must not be used for parkinsonism caused by antipsychotic drugs, as it can exacerbate the psychotic symptoms. 

Pharmacokinetics

Levodopa is absorbed from the jejunum and may compete with dietary aromatic amino acids.  Oral absorption is variable and following administration of 15mg/kg levodopa, peak plasma concentrations occur at 1 to 2 hours.  An increased half life is achieved by combining the levodopa with a COMT inhibitor, such as tolcapone.  Further, in the presence of dopa decarboxylase inhibitors, the plasma levodopa concentrations rise, excretion of dopamine and its metabolites falls and the availability of levodopa within the brain for conversion to dopamine increases.

 

Clinical Use

The combined preparations of levodopa and carbidopa (Sinemet) or levodopa and benserazide (Moldopar) are given three times daily starting at a low dose, which is increased after 2-3 weeks and then reviewed at intervals of 6-8 weeks.  The dose starts at 200-300mg and can increase up to 1g daily of levodopa.  After 2-5 years, the efficacy of levodopa becomes limited by the complications of motor fluctuations and dyskinesias.  Motor fluctuations are ‘wearing-off’ where individual doses produce only short lived effects, or ‘on-off’ where the patient may switch from symptomatic benefit from medication (on) to an akinetic-rigid state (off). These states are often without any predictable relationship to the time of the drug doses.

 

Dyskinesias are involuntary movements occurring in association with drug treatment.  Twisting and turning movements are seen when the dopamine levels are high (peak-dose dyskinesias) and painful sustained muscle contractions, typically of the feet are seen when dopamine levels are low (wearing-off dystonias). These fluctuations and dyskinesias can be partially alleviated in some patients by

1)      Frequent small doses of levodopa drugs

2)      Controlled release preparations

3)      Combined use of levodopa preparations with selegiline (MAO-B inhibitor) or a direct dopamine agonist (pergolide).

 

Adverse effects

These include nausea and vomiting, postural hypotension, involuntary movements (dystonia) including akathisia, chorea and myoclonus, psychological disturbances, cardiac arrhythmias and endocrine effects of levodopa including stimulation of growth hormone and suppression of prolactin.  Dopamine antagonists such as Domperidone can be used to reduce nausea and Clozapine is used to treat the hallucinations.

 

Drug interactions

MAO I’s can produce hypertensive reactions if given concurrently with levodopa.  The hypotensive actions of other drugs are potentiated by levodopa.

 

As the efficacy of levodopa falls off with long term treatment, this indicates that the underlying neurological disease progresses and is not reversed by levodopa treatment.  Many neurologists are now advising that dopamine agonists alone should be used initially to treat Parkinson’s Disease, paticularly in younger patients who are more at risk of developing the dopa-induced dyskinesias.

Increased release of endogenous dopamine

 

Since Parkinson’s disease results from the loss of inhibitory dopaminergic neurones in the nigrostriatal system, increasing endogenous release of dopamine from the remaining neurones can help to restore the normal neural circuits.

 

Amantidine

Mechanism:  Inhibits reuptake of dopamine into nerve terminals.

Clinical use:  Improves mild bradykinesia, tremor and rigidity in approximately 60% of       patients.  Less   effective than levodopa and action declines with time.  Oral dose is       initially 100mg daily, increased after one week to 100mg twice daily.

Adverse effects:  Relatively free from side effects compared with levodopa although

those experienced are qualitatively similar.  GI upsets, anorexia, nausea, livedo      reticularis, peripheral oedema, peripheral oedema, manifestations of CNS toxicity           (nervousness, inability to concentrate, insomnia, dizziness, convulsions,             hallucinations, blurred vision etc), rarely leucopenia.

Pharmacokinetics:  Half-life varies between 10 and 30 h thus it takes 4-7 days to reach     steady-state concentrations.  95% is eliminated by the kidney, thus

contraindicated in severe renal impairment.

 

Dopamine receptor agonists

These attempt to bypass the need for endogenous dopamine in the nigrostriatal system by directly stimulating dopamine receptors.  They are currently used as an adjunctive therapy in patients for whom use of levodopa alone is no longer adequately controlling their symptoms; however, it is currently being studied as to whether early use of these drugs in order to delay commencement of levodopa therapy could slow disease progression.  These drugs share many of the side effects experienced with levodopa, and when used in combination with levodopa, abnormal involuntary movements and confusion are common.  Peripheral side-effects such as nausea (due to stimulation of dopamine receptors in the chemoreceptor trigger zone) can be reduced by use of a peripheral dopamine receptor antagonist such as domperidone.  This is unable to cross the blood-brain barrier, hence its selective peripheral action.

 

This group of drugs comprises the ergot derivatives (bromocryptine, lysuride and pergolide) and the non-ergot derivatives (ropinirole, apomorphine and pramipexole).

 

Bromocryptine

Mechanism:  Potent agonist at D2 receptors (which underlies its efficacy in treating          PD) and a weak partial agonist at D1 receptors.

Clinical use:  Useful in reducing ‘off’ periods in levodopa therapy and stabilising    fluctuations in late-stage disease.  Dose is 1-2.5 mg at night, increased to a    maximum total of 10-40 mg per day, given in 3 divided doses.  Taken with food.

Adverse effects:  Nausea and vomiting, headache, dizziness, postural hypotension,            vasospasm of fingers and toes, constipation; with high doses, confusion, dyskinesia, hallucination, leg cramps, pleural effusions.

Pharmacokinetics:  Half-life is 6-8 h, longer than levodopa hence usefulness in reducing    fluctuations.

 

Lysuride

Mechanism:  Agonist at D2 receptors and 5-HT1 and 5-HT2 receptors.

Clinical use: Similar to bromocryptine.  Dose is initially 200 mg with food at bedtime,          increasing to max. 5mg daily in 4 divided doses.  Taken with food.

Adverse effects: CNS (headache, lethargy, confusion, dizziness, hallucinations), GI           (N+V, constipation, diarrhoea), rashes, cardiac arrhythmias, rarely severe           hypotension.

Pharmacokinetics:  Complete absorption but extensive first-pass metabolism.  Half-life of 8h.

 

Pergolide

Mechanism:  Agonist at D1, D2, 5-HT1 and 5-HT2 receptors.

Clinical use:  Used in combination with levodopa.  Oral dose is initially 50mg at bedtime,     increasing to a maximum of 2-5 mg daily, in 3 divided doses.  May allow cautious      reduction of dose of levodopa.

Adverse effects:  Similar CNS and GI symptoms to lysuride; also, abdominal pain             dyspepsia, pleural effusion, rhinitis, dyspnoea, pericarditis, hypotension, rash.

Pharmacokinetics:  Hepatic metabolism.  Half-life 15-42h.

 

Ropinirole

Mechanism:  Agonist at D2, D3 and D4 receptors.

Clinical use:  Either alone or as an adjunct to levodopa (may allow a reduction in dose of   levodopa of up to 20%).  Dose is usually 3-9 mg per day.  Oral.

Adverse effects:  CNS (hallucinations, drowsiness, confusion), GI (nausea, abdominal       pain, vomiting), somnolence, occasionally severe hypotension.

Pharmacokinetics:  Metabolised in the liver.  Half-life 6h.

Drug interactions:  Ciprofloxacin reduces clearance of ropinirole due to inhibition of           cytochrome P4501A2.

 

Apomorphine

Mechanism:  Agonist at D1 and D2 receptors.

Clinical use:  Used to stabilise refractory fluctuations in on-off activity with levodopa         therapy.  Use requires specialist supervision.  Patient must be established on         domperidone for 3 days before apomorphine is commenced.  Dose is initially 0.5        mg given s.c., increasing to 3-30mg daily in divided doses.  Dose regime may        progress to s.c. infusion.

Adverse effects:  severe N+V, dyskinesias, postural instability, cognitive impairment,         confusion, hallucinations, sedation, postural hypotension.

Pharmacokinetics:  Half-life is only 30 mins.

 

 

 

 

Inhibition of Catechol-O-Methyl Transferase (COMT)

 

COMT is an enzyme which metabolises L-dopa.  Thus inhibition of COMT reduces metabolism of L-dopa, increasing the amount of dopamine in the nigrostriatal neurones.

 

Tolcapone

Mechanism:  Tolcapone is a reversible competitive inhibitor of COMT. It is relatively        specific for central COMT, thus increasing the concentration of L-dopa in the       brain, without significantly increasing the concentration in the plasma.

Clinical use:  Recently use has been suspended following reports of serious

hepatotoxicity.  Previously used as an adjunct to levodopa, reducing end-of-dose

deterioration and allowing a reduced dose of levodopa.

Adverse effects:  Hepatitis, nausea, hallucinations, dyskinesia, occasional severe   hypotension.

Pharmacokinetics:  Hepatic metabolism.  

 

Inhibition of monoamine oxidase type B (MAO B)

Inhibition of MAO B prevents intraneuronal degradation of dopamine, increasing the availability of dopamine in the nigrostriatal neurones.  MAO B predominates in the dopaminergic regions of the CNS whereas MAO A acts more peripherally.  Selective inhibition of MAO B thus protects from unwanted peripheral effects of non-selective MAO inhibitors (‘cheese reaction’, drug interactions etc).

 

Selegiline

Mechanism:  Selective MAO B inhibitor so metabolism of dopamine in the striatum is

   decreased without affecting the other major transmitter amines. Increased levels of

   dopamine enhances the dopaminergic function in the nigrostratal tract.

   Selegiline does not produce the hypertensive reactions with ‘cheese and wine’ when  

   using non-selective MAO inhibitors e.g. phenelzine, because MAO-A remains active

   and metabolises noradrenaline and tyramine (which triggers the hypertensive reaction). 

   It may slow the neurodegenerative process of Parkinson’s Disease by preventing the

   breakdown of substances but this is unconfirmed.  E.g. MPTP is taken up by and kills

  dopaminergic neurones, is metabolised by MAO-B to the toxic product MPP+.

 

Clinical use:  Used in severe parkinsonism as an adjunct to levodopa to reduce end-of-  

  dose deterioration.  As with dopamine receptor agonists, it has been suggested that early

  treatment with selegiline, delaying onset of levodopa treatment, may delay disease

  progression, but there is no convincing evidence to support this claim. Unconfirmed

  studies have suggested that L-dopa and selegiline together may be associated with 

  increased mortality in long term. Can be given to patients with Parkinson’s Disease or

  symptomatic parkinsonism but not for drug-induced extrapyramidal disorders.

 

Dose: 10 mg daily as 2 divided doses of 5mg at breakfast and 5 mg at midday.

            Sudden withdrawal may exacerbate symptoms.

 

Adverse effects:  CNS (depression, confusion, psychosis, agitation, headache, tremor,       dizziness, vertigo), GI (constipation, diarrhoea, N+V), hypotension, back and

joint pain, muscle cramps.

 

Pharmacokinetics:   Oral selegiline is 100% absorbed but is extensively metabolised by the liver to active metabolite desmethylselegiline, which is also a MAO-B inhibitor. It is then metabolised to amphetamine and methamphetamine.  Plasma T1/2 is long with a mean value of 39 hours.

 

Drug interactions:

Tyramine:  Hypertension occurs at very high doses as MAO-B selectivity is lost and the

     pressor response to tyramine is potentiated. Cheese & wine reactions are rare. 

Amantadine and centrally acting anti-muscarinic agents e.g. orphenadrine, potentiate the

     anti-parkinsonian effects of selegiline.

Hyperpyrexia and CNS toxicity with pethidine

Antidepressants – hypertension and CNS excitation with fluoxetine, paroxetine.  CNS

     toxicity with TCA’s.  

 

 

Drugs affecting the cholinergic system

 

Muscarinic receptor antagonists

E.g. benzhexol, orphenadrine, procyclidine

 

Use

·         Their main use is parkinsonian effects caused by anti-psychotic agents (drug-induced parkinsonism) that can’t be withdrawn from psychiatric patients

·         For patients with mild parkinsonian symptoms especially tremor can use anti-muscarinic drugs alone initially or with selegiline.  L-dopa may be added to the treatment or substituted for as the symptoms progress.

·         Also used in post-encephalitic parkinsonism.

·         Benhexol, orphenadrine (antihistamine) and procyclidine are effective at decreasing parkinsonian tremor and to some extent rigidity and sialorrhoea.  Bradykinesia is least reduced. 

·         In an acute parkinsonian crisis i.v. procyclidine (5mg) or benztropine (2mg) are effective.

·         If patient is on a long term dopamine receptor antagonist, anticholinergics should be reserved for those with severe Parkinson’s disease.

 

Dose

·         Given in divided doses, increased every 2-5 days until their optimum benefit is reached or their toxic effects appear.

·         All are given orally either before eating if dry mouth or after meal if GI symptoms predominate. (Benzatropine and orphenadrine can be given intravenously or intramuscularly when treating phenothiazine-induced dystonia.)

·         Benzhexol 1-4 mg tds (up to maximum15mg /day)

·         Orphenadrine 150-400mg bd

·         Benztropine 0.5-4g daily in single or divided doses (maximum 6mg daily)

·         Procycilidine 2.5. mg tds (maximum of 60 mg /day)

 

Mechanism of action

·         Non-selective muscarinic receptor antagonists restore balance to the dopaminergic/ cholinergic pathways in the striatum by correcting the central cholinergic excess in parkinsonism thought to be due to the dopamine deficiency.

·         By acting on the postsynaptic muscarinic receptor in the striatal nuclei of the basal ganglia they antagonise the excitatory action of acetylcholine and thus reduce the extrapyramidal effects seen in Parkinson’s Disease.

 

Adverse effects (antimuscarinic effects)

1)      CNS – confusion, disorientation, excitation, sedation, psychosis

2)      CVS – tachycardia, arrhythmia

3)      GI – dry mouth, constipation

4)      GUS – Urinary urgency and frequency (contraindicated in prostatic hypertrophy)

5)      Paralysis of visual accommodation and glaucoma (contraindicated in glaucoma)

 

Pharmacokinetics

·         Benzhexol, Orphenadrine and Procyclidine can all be administered orally and are metabolised and excreted by the liver.  Orphenadrine has the longest half-life of 13.7-16.1 hours and is metabolised to an active metabolite. 

·         Benzatropine is excreted very slowly so changes in dose should be carried out gradually.

·         Procyclidine has a t1/2 of 13.7-16.1 hours and benzhexol t1/2 is 3-7 hours.

 

Drug interactions

·         Antimuscarinic effects e.g. dry mouth, urinary retention and constipation are enhanced with concominant use of nefopam, disopyramide, TCA’s, MAOI’s, antihistamines, phenothiazines and amantadine. 

·         Antagonise the GI effect of cisapride, metoclopramide and domperidone.

·         Parasympathomimetics: effects are antagonised.

 

 

Drugs used in other extrapyramidal diseases

 

DRUG-INDUCED EXTRAPYRAMIDAL SYNDROMES

·         Mostly seen after phenothiazine treatment  for schizophrenia and other psychoses and after anti-emetics with dopamine receptor blocking properties e.g. metaclopramide.

·         Symptoms include:

·         Parkinsonian symptoms in elderly patients

·         Acute dystonic reactions of rigidity, muscle spasm and opisthotonus

                        progressing to tardive dyskinesia in younger patients.

·         Tardive dyskinesia

·         Treatment is decrease in dosage of drugs or long term anticholinergic drug therapy. 

CHOREA

·         Commonly Huntingdon’s Chorea or secondary to cerebrovascular damage in basal ganglia. 

·         There is repetitive semi-purposeful movement resembling L-dopa induced dyskinesia.

·         It is due to a deficiency of inhibitory GABA, which results in the overactivity of the dopamine systems in basal ganglia.

·         Treatment:  Tetrabenazine which depletes dopamine from the  nerve endings

                          Haloperidol or phenothiazine which are dopamine receptor antagonists

                          Sodium Valproate which increases GABA availability in brain

 

DYSTONIA

·         Slowly sustained abnormal movement is dystonia.

·         Depending on where affected can get blephrospasm, torticollis, segmental dystonia and others.  I.m. Botulinum toxin may be helpful.

 

 

 

 

 

SUMMARY

 

·         The aims of the pharmacological treatments for Parkinson’s Disease are to replace or mimic the action of DOPAMINE (e.g. L-dopa) and to use muscarinic  antagonists to inhibit the excitatory effect of ACETYLCHOLINE.

·         Combination of L-dopa and a dopa-decarboxylase inhibitor are first line therapy for Parkinson’s Disease.  The benefit of treating with MAO-B inhibitor, selgeline, to retard disease progression is unconfirmed.  Dopamine agonists and COMT inhibitors are helpful as adjuvent drugs for patients with end of dose deterioration and reducing on-off motor fluctuations.

·         Polypharmacy is inevitable and as the doses are increased with the progression of the disease there is increased incidence of unwanted effects from the drugs used.

·         Anticholinergic drugs are the first-line treatment for drug-induced parkinsonism due to anti-psychotic drugs. 

·         Besides medication, Parkinson’s Disease may be treated surgically e.g. thalamotomy or pallidotomy, physical and speech therapy are used to keep the patients as active as possible.  There may be benefit from transplantation of foetal dopaminergic neurones although this is still under investigation.

 

Home ] Up ]