73
Views
1
CrossRef citations to date
0
Altmetric
Review

Adjunctive therapy in Parkinson’s disease: the role of rasagiline

&
Pages 285-294 | Published online: 02 Jul 2012

Abstract

Parkinson’s disease is the second most common neurodegenerative disorder, currently affecting 1.5 million people in the US. In this review, we describe the diagnostic and pathological features of Parkinson’s disease, as well as its clinical course. We then review pharmacologic treatments for the disease, with a particular focus on therapies adjunctive to levodopa and specifically the role of rasagiline. We review the four pivotal rasagiline trials, and discuss rasagiline and its use as adjunctive therapy for Parkinson’s disease. Finally, we discuss potential side effects, drug interactions, and other practical aspects concerning the use of rasagiline in Parkinson’s disease.

Introduction

Parkinson’s disease (PD) is the second most common neurodegenerative disorder, currently affecting 1.5 million people in the US.Citation1 The clinical picture of PD was astutely described by an apothecary surgeon, James Parkinson, in an historic paper entitled “An essay on the shaking palsy”, published in 1817, yet it was not until years later that the malady of “paralysis agitans”, also known as the shaking palsy, was given the name “Parkinson’s disease” by Dr Jean Martin Charcot.Citation2,Citation3 The pathologic hallmark of PD is loss of dopaminergic neurons within the substantia nigra, as well as intracellular accumulation of Lewy bodies.Citation4

Parkinson’s disease

Cardinal clinical features

The four cardinal clinical features of PD are tremor, bradykinesia, rigidity, and postural instability. The UK Parkinson’s Disease Society Brain Bank criteria are the most commonly used guidelines for the diagnosis of idiopathic PD.Citation5 Required is the presence of bradykinesia along with at least one of the following: muscular rigidity, 4–6 Hz rest tremor, and postural instability not caused by primary visual, vestibular, cerebellar, or proprioceptive dysfunction, along with careful consideration of exclusion and supportive criteria ().Citation6 As such, unilateral or asymmetric presentation is often the norm, and a progressive advancement of symptoms, along with a favorable response to levodopa therapy, is commonly required to make the clinical diagnosis. In addition to these typical motor symptoms, PD patients frequently experience a number of nonmotor symptoms throughout the course of the disease.Citation7 Common nonmotor symptoms are cognitive problems, ranging from mild cognitive impairment with attention and set- shifting deficits early on, to frank dementia in some patients in the later stages of the disorder, as well as depression and anxiety.Citation7 In addition, many PD patients suffer from pain syndromes and gastrointestinal problems, such as constipation, dysphagia, sialorrhea, and delayed gastric emptying.Citation7,Citation8 Other autonomic symptoms, such as hypotension, urinary dysfunction, and sexual dysfunction, frequently accompany the typical motor symptoms of PD at some point as well.Citation7,Citation8 In fact, as a synucleinopathy with progressive burden of intraneuronal Lewy bodies, the progression of nonmotor symptoms of PD can be correlated pathologically with accrual of Lewy bodies. Accumulation of Lewy bodies at Braak stage 1 starts at the level of the lower medulla oblongata, so symptoms such as impaired olfaction because of involvement of the olfactory nucleus are experienced.Citation9 According to Braak staging, the neuro-pathologic changes of PD progress in an ascending fashion to next involve areas such as the great raphe nucleus, locus ceruleus, and reticular formation in stage 2, before the basal portions of the midbrain, forebrain, and substantia nigra pars compacta are affected in stage 3 when the PD motor symptoms become noticeable. Stages 4–6 involve spread to the amygdala, mesocortex, and neocortex, and correlate with clinical progression and specifically development of dementia.Citation9

Table 1 UK Parkinson’s Disease Society Brain Bank diagnostic criteria for Parkinson’s diseaseCitation6

Pathogenesis

A summary of pathologic findings in PD entails consideration of the decline and loss of dopaminergic cells within the substantia nigra pars compacta and Lewy body accumulation.Citation10Citation13 At a deeper cellular level, creation of free oxygen radicals by disrupted antioxidative mechanisms creates damage to important proteins, as well as lipids and DNA, and thus damage to dopaminergic cells which are especially susceptible to oxidative stressors.Citation14,Citation15

The in vivo PD model created utilizing the compound 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) establishes the basic mechanism of PD by inducing loss of dopaminergic cells in animal models and has also been theorized to induce acquired parkinsonism in some humans after recreational consumption of an illicit drug containing the premetabolite. The MPTP compound is known to inhibit mitochondrial complex I.Citation16,Citation17

Given that contributing pathogenic factors combine to create the cellular changes inducing PD, microglial activation and astrocytic reactions in the substantia nigra have been implicated in post mortem studies as possible associated links.Citation18Citation20 With activation of microglia and the associated cytokine cascade, a neuroinflammatory response has been documented in the brains of PD patients as well as via in vivo imaging studies demonstrating nigrostriatal dopaminergic terminal loss.Citation21Citation29

With the knowledge that Lewy bodies are found in patients with PD and established as part of the pathologic process contributing to the mechanism of PD, it is felt that the alpha synuclein found in Lewy bodies promotes accumulation of other Lewy bodies.Citation30 In fact, alpha synuclein in the physiologic state exists in an alpha-helical conformation; however, in the pathologic state, it refolds into a beta sheet conformation and then promotes other alpha-helical synuclein proteins to reconfigure as well.Citation31

Calcium homeostasis within the striatal dopamine structures is a critical process for maintaining adequate dopamine levels. This is achieved through a balance of intracellular calcium via ion channels.Citation32,Citation33 In PD patients, who already have a decrease in dopaminergic striatal cells, glutamatergic input in an attempt to activate and compensate for the dopaminergic loss ironically leads to glutamate excitotoxicity.Citation34 Glutamate causes activation of NMDA and AMPA receptors which, in turn, activate voltage-gated ion channels, leading to an influx of extracellular calcium.Citation35,Citation36 This leads to a cascade of processes, which further promotes energy expenditure and subsequent mitochondrial damage and apoptosis.Citation37Citation39

The mechanism of apoptosis or programmed cell death is felt to be another contributor in the pathogenesis of PD.Citation13,Citation40 Inflammatory processes that promote microglia may activate cell surface receptors for tumor necrosis factor as part of the extrinsic apoptotic process leading to dopaminergic cell death.Citation41 Also, other cell surface receptors linked to the extrinsic apoptotic process are felt to be overexpressed in PD patients.Citation7,Citation42 Additionally, enhanced mitochondrial cell permeability leads to intrinsic apoptotic cell death and, as mentioned previously, can promote a neurotoxic cascade in an effort to balance dopamine concentrations.Citation13

Monotherapy

Patients with early PD are usually managed with a single drug in monotherapy to address the motor symptoms of PD. Therapeutic choices for early PD are levodopa, dopamine agonists, monoamine oxidase type B inhibitors, amantadine, and trihexylphenidyl. Levodopa is considered the most effective treatment for the motor symptoms of PD.Citation7 Levodopa requires active transportation across the blood–brain barrier and is then converted into dopamine by dopaminergic neurons via aromatic-L-amino acid decarboxylase.Citation13 Nondopaminergic and glial cells also convert levodopa and the product is packed into vesicular monoamine transporter 2 vesicles at synaptic terminals. Citation43Citation45 A substudy to the ELLDOPA (Earlier versus Later Levodopa) study, addressing the concern that levodopa could hasten the course of the disease, has rejected this hypothesis.Citation46 Levodopa therapy has been shown to increase life expectancy in PD, is available at a low cost, and is usually well tolerated, even in the elderly patient.Citation7,Citation47 The major downside of levodopa therapy is its short half-life and the resultant pulsatile stimulation of dopamine receptors, leading to the development of motor fluctuations after several years of treatment.Citation48 Motor fluctuations consist of periods of excessive movements or dyskinesias (most commonly at peak dose of levodopa), and periods of “off time”, with paucity of movement, or akinesia. In order to avoid or delay the development of motor fluctuations, neurologists will often start younger patients on longer-acting dopamine agonists or monoamine oxidase B inhibitors first, or use levodopa as an adjunct to a longer-acting agent. The caveat here is that dopamine agonists are often poorly tolerated by the elderly and by cognitively impaired patients, so levodopa monotherapy, despite its limitations, is preferred in this setting.

Need for adjunctive therapy

Adjunctive therapy is most commonly used when motor fluctuations develop in a patient treated with levodopa (). Some clinicians will also use adjunctive treatment to keep the levodopa dosage low, in order to delay the onset of motor fluctuations, but there are no evidenced-based guidelines to support this practice. A common strategy to extend the half-life of levodopa is the addition of a catechol-O-methyltransferase inhibitor such as entacapone or tolcapone, which can help the “wearing off ” effect in between levodopa doses. Entacapone and tolcapone work to inhibit catechol-Omethyltransferase which otherwise causes the breakdown and degradation of levodopa. Entacapone acts peripherally and tolcapone acts both centrally and peripherally.Citation4 While both are effective adjuncts, entacapone is usually the preferred agent due to concerns about hepatotoxicity with tolcapone and the resulting requirements for liver enzyme monitoring.Citation7

Table 2 Adjuncts to levodopa therapyCitation5,Citation7

Dopamine agonists, such as pramipexole and ropinirole, are utilized in early PD as monotherapy and for advanced PD in combination with levodopa, monoamine oxidase B inhibitors, or other adjuncts.Citation7 Dopamine agonists work directly on dopamine receptors and do not depend on surviving dopaminergic cells or dopamine metabolism, as does levodopa.Citation13 Dopamine agonists have been shown to delay the onset of dyskinesias when used in monotherapy and to treat “wearing off ” effectively when added to levodopa.Citation7

Monoamine oxidase inhibition is utilized to prevent degradation of dopamine and thus extends the length of usefulness of levodopa therapy or endogenous dopamine. Monoamine oxidase is normally found within the outer membrane of mitochondria, and its presence in the central nervous system accounts for a portion of oxidative degradation of dopamine as well as subsequent oxidative stress and further degradation of striatal neurons.Citation49 In fact, within the central nervous system, it is the monoamine oxidase B enzyme that is predominant. By blocking the monoamine oxidase B enzyme, drugs such as selegiline and rasagiline have found usefulness as treatments for early PD and as adjunctive therapy.

Selegiline

Utilizing the notion that, of the two types of monoamine oxidase contained within the human body, the brain contains mostly the monoamine oxidase type B enzyme, selegiline became noteworthy for its selective and irreversible binding to monoamine oxidase type B and thereby showed promise in exerting “antiakinetic” properties in PD patients treated with levodopa.Citation50 Previously known as (−) deprenyl, selegiline has been identified as a methamphetamine derivative with a propargyl group attached to a nitrogen molecule. With the discovery that positioning of the N-propargyl group chemically in relation to the “parent” aromatic ring helps to confer irreversibility and enzyme inhibition activity, other compounds were identified and tested for monoamine oxidase B inhibition properties as well.Citation51

Selegiline has been demonstrated to lack the “cheese effect” despite high amounts of exogenous tyramine given in animal and human models, although theoretical questions have been posed regarding whether the lack of a tyramine effect remains despite chronicity of exposure to selegiline.Citation52,Citation53 In combination with levodopa, selegiline has been proven to be effective for mild “on-off ” motor symptoms as well as motor akinesias. However, in some subjects, the combination of levodopa and selegiline promotes dyskinesias.Citation54 DATATOP (deprenyl and tocopherol antioxidative therapy of parkinsonism) was a large, double-blind, placebo-controlled study comparing treatment of early PD patients with selegiline, tocopherol, or a combination of selegiline and tocopherol versus placebo.Citation55 Selegiline, but not tocopherol, was found to delay time until disability requiring levodopa significantly, but because of its known symptomatic benefits, a neuroprotective effect could not be claimed.Citation56,Citation57

Rasagiline

Rasagiline is a novel second-generation monoamine oxidase B inhibitor that is selective and irreversible. Although selegiline is metabolized to L-methamphetamine and L-amphetamine via first-pass effects, rasagiline does not metabolize into an amphetamine derivative.Citation58,Citation59 The amphetamine metabolite of selegiline is felt to be neurotoxic, so any potential neuroprotective effect of selegiline is thus negated.Citation60 On the other hand, rasagiline is an isomer of N-propargyl-1-aminoindan, which is a nonamphetamine metabolite. It is the propargyl chain that confers rasagiline with the irreversible and selective ability to inhibit monoamine oxidase B and is also thought to provide some degree of neuroprotection.Citation51,Citation61,Citation62 Neurorescue effects have also been postulated based on the results of in vivo studies showing reduction of proapoptotic proteins and reduction of proteins signaling programmed cell death.Citation63 Studies have also shown that treatment with propargylamine enhances expression of the cell survival genes GDNF and BDNF, which are both components felt to be important in neuroprotective and neurorescue activity toward dopaminergic and cholinergic neurons.Citation63Citation65 Further proposed mechanisms by which rasagiline is felt to be neuroprotective include those in which there is upregulation of cellular antioxidant activity as well as the promotion of factors against apoptosis.Citation66 The antioxidant activity is conferred as an increase in superoxide dismutase and catalase and the antiapoptotic potential is conferred due to stabilization of the mitochondrial membrane via binding of rasagiline to a portion of the membrane, which instigates the apoptotic process.Citation67Citation70 Rasagiline also hinders the apoptotic process by downregulating proapoptotic proteins.Citation71,Citation72 A dose of 1 mg in a PD patient reaches a maximal serum concentration within 0.5–0.7 hours of ingestion.Citation73,Citation74 Although the terminal half-life is 1.34 hours, this does not correlate with biologic activity because restoration of monoamine oxidase B activity depends on enzyme synthesis. Imaging studies measuring brain monoamine oxidase B occupancy after rasagiline dosing have indicated enzyme recovery may take up to 40 days in healthy populations.Citation75 Studies in animal models have also supported the idea that rasagiline offers some amount of neuroprotection. For instance, pretreatment with rasagiline has attenuated the process by which MPTP utilizes the monoamine oxidase B enzyme and is transformed into a further potent neurotoxin in animal models.Citation76,Citation77

A possible disease-modifying effect has also been postulated after observing results of a human clinical trial using a novel delayed-start study design for rasagiline monotherapy in early PD, ie, the ADAGIO (Attenuation of Disease Progression with Azilect Given Once-daily) trial.Citation78 The US Food and Drug Administration recently reviewed the ADAGIO trial and did not grant rasagiline an approved indication for neuroprotection.Citation79 However, rasagiline is approved by the Food and Drug Administration as early monotherapy and also adjunctive therapy in PD, based on the results of four pivotal clinical trials.

Pivotal clinical trials

The TEMPO (Rasagiline in Early Monotherapy for Parkinson’s disease Outpatients) study utilized a delayed-start model, which was randomized and double-blinded. The purpose was to evaluate the safety and efficacy of rasagiline in PD patients who were not yet advanced enough in their disease to require levodopa therapy.Citation80,Citation81 The primary means of measuring drug efficacy was the change in total Unified Parkinson’s Disease Rating Scale (UPDRS) score. The first half of the study period was 26 weeks long in total, included 404 patients with early PD, and randomized patients to placebo or rasagiline 1 mg or 2 mg per day. After 6 months, the adjusted effect size (drug effect minus placebo effect) according to UPDRS score was −4.20 in the 1 mg group and −3.56 in the 2 mg group compared with placebo. After 6 months, the placebo group was switched to 2 mg to become the delayed-start group. The groups assigned rasagiline 1 mg and 2 mg continued on for a further 6 months. A total of 371 patients completed this half of the study. Mean adjusted total UPDRS scores in the rasagiline 2 mg group versus the delayed-start group showed a 2.29 unit smaller increase in score whereas the rasagiline 1 mg group showed a 1.82 smaller increase in score versus the delayed-start group. Overall, the patients treated earlier than later had less functional decline, implying a potential neuroprotective effect since the delayed-start patients did not catch up to the early-start patients.Citation82 A subsequent review of a subgroup of patients followed over a period of 4.5, 5.0, and 5.5 years demonstrated continued benefit and less progression compared with the delayed-start group and even continued a similar benefit beyond 6 years.Citation83Citation85 The ADAGIO delayed-start trial was subsequently designed to help address the question of neuroprotection for PD patients.

In the ADAGIO trial, 1176 untreated PD subjects were enrolled and randomized to rasagiline at a dose of either 1 mg or 2 mg for 72 weeks or to placebo for the first 36 weeks and then rasagiline 1 mg or 2 mg for the remaining 36 weeks. Positive endpoint goals included: proving superiority to placebo in the rate of change in UPDRS scores between weeks 12 and 36; proving superiority compared with delayed-start treatment, as indicated by less change in UPDRS scores between baseline and week 72; and proving noninferiority to delayed-start treatment in the rate of change in UPDRS scores between weeks 48 and 72. In the early-start 1 mg group, all three end points were achieved. The group that started 2 mg early did not meet all three criteria based on the finding that there was no significant difference in change of UPDRS score between baseline and week 72 in the early-start versus delayed-start groups.

Two additional landmark trials specifically addressed the utility of adding rasagiline therapy onto the regimen for PD patients. The LARGO (Lasting Effect in Adjunct Therapy with Rasagiline Given Once Daily) study evaluated the safety and efficacy of adjunctive rasagiline therapy in patients treated with levodopa and compared this group with subjects taking either placebo and levodopa or entacapone and levodopa. Results of this 18-week study revealed a similar reduction in “off ” time for the rasagiline and entacapone adjunctive groups compared with placebo. Interestingly, a subanalysis found that the rasagiline group had significantly less freezing of gait, postural instability, and gait disturbance, as well as improved motor scores when the patient was “off ” compared with those on placebo.Citation86

In the PRESTO (Parkinson’s Rasagiline: Efficacy and Safety in the Treatment of “Off ”) study, subjects with at least 2.5 hours of daily “off ” time despite maximal levodopa treatment were randomized to receive rasagiline 0.5 mg/day or 1 mg/day or placebo along with their maximized levodopa regimen for a period of 26 weeks. If needed, the levodopa dose could be reduced within the first 6 weeks of the trial. Results of this study indicated that both the 0.5 mg and 1 mg rasagiline groups experienced reduction of “off ” hours by 1.41 and 1.85 hours, respectively, compared with placebo, at 0.91 hours. Rasagiline was also found to improve clinical global impression scores significantly; UPDRS scores for activities of daily living during “off ” time and UPDRS motor scores improved during “on” time in both the 0.5 mg and 1 mg groups when compared with placebo. In the rasagiline 0.5 mg group that experienced increased “on” time and reduced “off ” time, subjects experienced all of their increased “on” time without dyskinesias. The rasagiline 1 mg group experienced some dyskinesias, with added improvement in “on” time. The total additional “on” time with dyskinesias was found to be a mean of 0.37 hours. Additionally, the rasagiline 0.5 mg group experienced significant improvement in postural instability and gait and tremor during “on” times, and the rasagiline 1 mg group experienced significant improvement in UPDRS subscores for rigidity, bradykinesia, and tremor.Citation87

In all, the TEMPO, LARGO, PRESTO, and ADAGIO trials have demonstrated sustained improvement of motor function in patients with early PD, and also improvement in motor function and reduction of “off ” hours, as well as improvement in difficult to treat postural instability, gait disturbance, and freezing of gait symptoms in patients with moderate to advanced PD and motor fluctuations.

Side effects and interactions

Rasagiline is generally well tolerated and, because of the lack of amphetamine metabolites, which in preclinical trials have shown adverse effects upon cardiovascular and psychiatric systems, has a more favorable side effect profile compared with selegiline, which often causes insomnia, confusion, and psychosis.Citation88,Citation89 Rates of dizziness, hypotension, confusion, and hallucinations were found to be no different from those on placebo in clinical trials of rasagiline.Citation80,Citation81,Citation86,Citation87 Goetz et al performed a post hoc analysis of the TEMPO and PRESTO trials in order to evaluate the safety of rasagiline specifically in the elderly, given that this population is especially prone to side effects from drugs used in Parkinson’s disease.Citation90 Analysis of the data from these trials for patients aged 70 years or older did not show any statistically significant association between side effects, age, and use of rasagiline. Although adverse events occurred more frequently in the elderly, this was irrespective of treatment with rasagiline or placebo.Citation90 summarizes the adverse events encountered in one or more of the four pivotal trials. The tyramine or “cheese” reaction whereby a heightened vasopressor response is experienced when dietary tyramine is combined with monoamine oxidase inhibitors, was not associated with administration of rasagiline, even when the amount of tyramine exceeded typical dietary intake.Citation91,Citation92 Serotonin syndrome, on the other hand, is a risk in patients taking either selegiline or rasagiline in combination with meperidine.Citation93,Citation94 Furthermore, concomitant use of tramadol, methadone, propoxyphene, dextromethorphan, cyclobenzaprine, and St John’s wort are contraindicated in patients taking rasagiline, according to the prescribing information.Citation95 Although there were a limited number of patients taking selective serotonin reuptake inhibitors in the TEMPO, LARGO, PRESTO, and ADAGIO trials, no patients were reported to suffer from serotonin syndrome while participating in these studies.Citation67,Citation80,Citation81,Citation86,Citation87,Citation94 Fluoxetine and fluvoxamine were not allowed to be used during the trials due to their long half-lives.Citation94 However, in the postmarketing period, nonfatal cases of serotonin syndrome have been reported in patients treated concomitantly with antidepressants and rasagiline.Citation95

Table 3 Adverse events from pivotal clinical trials

Drug interactions involving the cytochrome P450 (CYP isoenzymes) and rasagiline are of concern. CYP inhibitors such as ciprofloxacin, cimetidine, and fluvoxamine can significantly increase the area under the concentration–time curve for rasagiline.Citation94,Citation96 Prescribing recommendations state that rasagiline dosing should not exceed 0.5 mg daily in patients taking CYP inhibitors and that the dose should be reduced in patients with mild hepatic impairment.Citation97 On the other hand, CYP inducers such as omeprazole can decrease the area under the curve for rasagiline.Citation94 A summary of contraindications to rasagiline therapy is outlined in .

Table 4 Contraindications and other considerations for rasagiline dosing

Potential benefits on cognition and quality of life

In addition to treating the motor symptoms of PD, rasagiline has shown promise in the realm of enhancing cognition. A multicenter, double-blind, placebo-controlled trial has recently been conducted involving 55 nondemented but cognitively impaired PD patients.Citation98 The study was conducted over 12 weeks and patients on dopaminergic treatment were randomized to receive either placebo or rasagiline 1 mg. Neuropsychologic assessment was performed at baseline and at the end of the 12-week period. A significant improvement was found in a test measure of attention as well as a suggestion of benefit in the area of executive function, with significant improvements seen in a test of verbal fluency. There was a trend toward improvement in semantic verbal fluency as well. However, the trial did not show improvement in memory or visual–spatial domains.Citation98

A cost-utility study by Hudry et al, performed in Finland, compared levodopa therapy alone (standard care) versus rasagiline with levodopa and entacapone with levodopa, measuring quality-adjusted life-years and number of months spent with less than 25% of “off ” time. Parity pricing for rasagiline and entacapone was assumed, based on the World Health Organization daily drug dose system. The study showed adjunctive rasagiline therapy to have greater cost-effectiveness and cost reductions compared with standard care, per treated patient. After 2 years, rasagiline showed 0.13 (95% confidence interval 0.08–0.17) additional quality-adjusted life-years and 5.2 (3.6–6.7) additional months of reduced “off ” time compared with levodopa alone.Citation99

Practical applications

When faced with the clinical problem of treating motor fluctuations in PD, adjunctive therapy with longer-acting agents in addition to short-acting levodopa is often needed. Rasagiline is an effective and well tolerated adjunctive medication in this setting, and is proven to reduce “off ” time to an extent similar to that of entacapone. Adjunctive rasagiline is usually initiated at a dose of 0.5 mg in the morning, and can be increased if needed to 1 mg daily given in a single dose. Levodopa therapy or other concomitant PD drugs might have to be reduced after addition of rasagiline if dyskinesias occur. Rasagiline is an attractive choice in this setting because of the potential for neuroprotection based on preclinical and clinical data (even though the drug is not approved by the Food and Drug Administration for this indication), as well as easy once-daily dosing, favorable tolerability compared with selegiline (amphetamine metabolite) or dopamine agonists (often cause sedation, orthostasis, psychosis and dopamine dysregulation syndrome), and the suggestion from clinical trials of a potential effect on cognition, freezing of gait, and postural instability. The disadvantages of rasagiline therapy are related to cost (currently no generic formulation is available), and a number of potential drug–drug interactions that have to be carefully considered prior to the decision to initiate rasagiline therapy.

Disclosure

KDG has received speaking honoraria from Medtronic and Teva Pharmaceuticals. VKH has received consulting and speaking honoraria from Teva Pharmaceuticals.

References

  • Parkinson.org [homepage on the Internet]OberdorfJSchmidtPImproving care for people with Parkinson’s diseaseNational Parkinson Foundation22010 Available from: http://www.parkinson.org/NationalParkinsonFoundation/files/c2/c2abb799-4e2b-41c5-8c06-47-f3424a58d0.pdfAccessed February 6, 2012
  • ParkinsonJAn essay on the shaking palsy. 1817J Neuropsychiatry Clin Neurosci200214222323611983801
  • HaasLFJean Martin Charcot 1825–1893 and Jean Baptiste Charcot 1867–1936J Neurol Neurosurg Psychiatry200171452411561039
  • SiderowfASternMUpdate on Parkinson diseaseAnn Intern Med2003138865165812693888
  • SuchowerskyOReichSPerlmutterJPractice parameter: diagnosis and prognosis of new onset Parkinson disease (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of NeurologyNeurology200666796897516606907
  • HughesAJDanielSEKilfordLLeesAJAccuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinicopathological study of 100 casesJ Neurol Neurosurg Psychiatry19925531811841564476
  • ZesiewiczTHauserRMedical treatment of motor and nonmotor features of Parkinson’s diseaseContinuum20071311238
  • MiyasakiJMShannonKVoonVPractice parameter: Evaluation and treatment of depression, psychosis, and dementia in Parkinson disease (an evidence-based review) Report of the Quality Standards of Subcommittee of the American Academy of NeurologyContinuum2007131193201
  • BraakHBohlJMullerCStanley Fahn Lecture 2005: The staging procedure for the inclusion body pathology associated with sporadic Parkinson’s disease reconsideredMov Disord200621122042205117078043
  • FornoLSConcentric hyaline intraneuronal inclusions of Lewy type in the brains of elderly persons (50 incidental cases): relationship to parkinsonismJ Am Geriatr Soc19691765575754182529
  • IshiyamaMYagishitaSHasegawaKYokoyamaTUltrastructural study (transmission and scanning EM) of cortical Lewy bodiesNeuropathology2006262A58
  • BraakHDel TrediciKRubUde VosRAJansen SteurENBraakEStaging of brain pathology related to sporadic Parkinson’s diseaseNeurobiol Aging200324219721112498954
  • KincsesZTVecseiLPharmacological therapy in Parkinson’s disease: focus on neuroprotectionCNS Neurosci Ther201117534536720438581
  • BogaertsVTheunsJvan BroeckhovenCGenetic f indings in Parkinson’s disease and translation into treatment: a leading role for mitochondria?Genes Brain Behav20087212915117680806
  • MakerHSWeissCSilidesDJCohenGCoupling of dopamine oxidation (monoamine oxidase activity) to glutathione oxidation via the generation of hydrogen peroxide in rat brain homogenatesJ Neurochem19813625895937463078
  • GerlachMRiedererPPrzuntekHYoudimMBMPTP mechanisms of neurotoxicity and their implications for Parkinson’s diseaseEur J Pharmacol199120842732861815982
  • LangstonJWBallardPTetrudJWIrwinIChronic parkinsonism in humans due to a product of meperidine-analog synthesisScience198321945879799806823561
  • McGeerPLItagakiSBoyesBEMcGeerEGReactive microglias are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brainsNeurology1988388128512913399080
  • ImamuraKHishikawaNSawadaMNagatsuTYoshidaMHashizumeYDistribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson’s disease brainsActa Neuropathol (Berl)2003106651852614513261
  • MirzaBHadbergHThomsenPMoosTThe absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson’s diseaseNeuroscience200095242543210658622
  • MogiMHaradaMNarabayashiHInagakiHMinamiMNagatsuTInterleukin (IL)-1 beta, IL-2, IL-4, IL-6 and transforming growth factor-alpha levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson’s diseaseNeurosci Lett1996211113168809836
  • MogiMHaradaMKondoTRiedererPNagatsuTInterleukin-2 but not basic fibroblast growth factor is elevated in parkinsonian brain. Short communicationJ Neural Transm19961038–9107710819013395
  • MogiMHaradaMKondoTNarabayashiHRiedererPNagatsuTTransforming growth factor-beta 1 levels are elevated in the striatum and in ventricular cerebrospinal fluid in Parkinson’s diseaseNeurosci Lett199519321291327478158
  • MogiMHaradaMKondoTRiedererPNagatsuTBrain beta 2-microglobulin levels are elevated in the striatum in Parkinson’s diseaseJ Neural Transm Park Dis Dement Sect19959187927605592
  • MogiMHaradaMKondoTInterleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patientsNeurosci Lett199418021471507700568
  • MogiMHaradaMRiedererPNarabayashiHFujitaKNagatsuTTumor necrosis factor-alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patientsNeurosci Lett19941651–22082108015728
  • MogiMHaradaMKojimaKBeta 2-microglobulin decrease in cerebrospinal fluid from parkinsonian patientsNeurosci Lett19891041–22412462682394
  • GerhardAPaveseNHottonGIn vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s diseaseNeurobiol Dis200621240441216182554
  • OuchiYYoshikawaESekineYMicroglial activation and dopamine terminal loss in early Parkinson’s diseaseAnn Neurol200557216817515668962
  • ChauKYChingHLSchapiraAHCooperJMRelationship between alpha synuclein phosphorylation, proteasomal inhibition and cell death: relevance to Parkinson’s disease pathogenesisJ Neurochem200911031005101319493164
  • OlanowCWPrusinerSBIs Parkinson’s disease a prion disorder?Proc Natl Acad Sci U S A200910631125711257219666621
  • BonciAGrillnerPMercuriNBBernardiGL-Type calcium channels mediate a slow excitatory synaptic transmission in rat midbrain dopaminergic neuronsJ Neurosci19981817669367039712641
  • PingHXShepardPDApamin-sensitive Ca(2+)-activated K+ channels regulate pacemaker activity in nigral dopamine neuronsNeuroreport1996738098148733751
  • RodriguezMCObesoJAOlanowCWSubthalamic nucleus-mediated excitotoxicity in Parkinson’s disease: a target for neuroprotectionAnn Neurol1998443 Suppl 1S175S1889749591
  • DingledineRBorgesKBowieDTraynelisSFThe glutamate receptor ion channelsPharmacol Rev199951176110049997
  • GreenamyreJTPorterRHAnatomy and physiology of glutamate in the CNSNeurology19944411 Suppl 88S7S137526272
  • HetzCAER stress signaling and the BCL-2 family of proteins: from adaptation to irreversible cellular damageAntioxid Redox Signal20079122345235517854276
  • LindholmDWootzHKorhonenLER stress and neurodegenerative diseasesCell Death Differ200613338539216397584
  • GheorghiadeMKonstamMABurnettJCJrShort-term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status TrialsJAMA2007297121332134317384438
  • KerrJFWyllieAHCurrieARApoptosis: a basic biological phenomenon with wide-ranging implications in tissue kineticsBr J Cancer19722642392574561027
  • HirschECHunotSNeuroinflammation in Parkinson’s disease: a target for neuroprotection?Lancet Neurol20098438239719296921
  • FerrerIBlancoRCutillasBAmbrosioSFas and Fas-L expression in Huntington’s disease and Parkinson’s diseaseNeuropathol Appl Neurobiol200026542443311054182
  • HuotPParentADopaminergic neurons intrinsic to the striatumJ Neurochem200710161441144717286588
  • KitahamaKGeffardMAranedaSLocalization of L-DOPA uptake and decarboxylating neuronal structures in the cat brain using dopamine immunohistochemistryBrain Res20071167567017692830
  • YamadaHAimiYNagatsuITakiKKudoMAraiRImmunohistochemical detection of L-DOPA-derived dopamine within serotonergic fibers in the striatum and the substantia nigra pars reticulata in Parkinsonian model ratsNeurosci Res20075911717586078
  • FahnSthe Parkinson Study GroupDoes levodopa slow or hasten the rate of progression of Parkinson’s disease?J Neurol2005252Suppl 4IV37IV4216222436
  • RajputAHUittiRRajputATimely levodopa (LD) administration prolongs survival in Parkinson’s diseaseParkinsonism Relat Disord19973315916518591070
  • ZesiewiczTSullivanKHauserRLevodopa-induced dyskinesia in Parkinson’s disease: epidemiology, etiology and treatmentCurr Neurol Neurosci Rep20077430231017618536
  • DostertPLStrolin BenedettiMTiptonKFInteractions of monoamine oxidase with substrates and inhibitorsMed Res Rev19899145892644497
  • BirkmayerWRiedererPYoudimMBHLinauerWThe potentiation of the anti akinetic effect after L-dopa treatment by an inhibitor of MAO-B, DeprenylJ Neural Trans1975363–4303326
  • KalirASabbaghAYoudimMBHSelective acetylenic ‘suicide’ and reversible inhibitors of monoamine oxidase types A and BBr J Pharmacol198173155647284698
  • SandlerMGloverVAshfordASternGMAbsence of “cheese effect” during deprenyl therapy: some recent studiesJ Neural Trans1978433–4209215
  • ElsworthJDGloverVReynoldsGPDeprenyl administration in man: a selective monoamine oxidase B inhibitor without the ‘cheese effect’Psychopharmacology1978571333896466
  • LeesAJKohoutLJShawKMDeprenyl in Parkinson’s diseaseLancet19973108042791795
  • Parkinson Study GroupDATATOP: a multicenter controlled clinical trial in early Parkinson’s diseaseArch Neurol19894610105210602508608
  • LeWittPANeuroprotection by anti-oxidant strategies in Parkinson’s diseaseEur Neurol199333Suppl 124308375428
  • ShoulsonIDATATOP: a decade of neuroprotective inquiry. Parkinson Study Group. Deprenyl and tocopherol antioxidative therapy of parkinsonism1998Ann Neurol443 Suppl 1S160S1669749589
  • MahmoodIClinical pharmacokinetics and pharmacodynamics of selegiline. An updateClin Pharmacokinet1997332911029260033
  • ShinHSMetabolism of selegiline in humans. Identification, excretion, and stereochemistry of urine metabolitesDrug Metab Dispos19972566576629193866
  • Bar AmOAmitTYoudimMBContrasting neuroprotective and neurotoxic actions of respective metabolites of anti-Parkinson drugs rasagiline and selegilineNeurosci Lett2004355316917214732458
  • BindaCHubalekFLiMCrystal structures of monoamine oxidase B in complex with four inhibitors of the N-propargylaminoindan classJ Med Chem20044771767177415027868
  • MaruyamaWNaoiMNeuroprotection by (−)-deprenyl and related compoundsMech Ageing Dev19991112–318920010656536
  • Bar-AmOWeinrebOAmitTYoudimMBRegulation of Bcl-2 family proteins, neuroprotective factors, and APP processing in the neurorescue activity of propargylamineFASEB J200519131899190116148027
  • MurerMGYanQRaisman-VozariRBrain-derived neurotrophic factor in the control human brain and in Alzheimer’s disease and Parkinson’s diseaseProg Neurobiol20016317112411040419
  • WangLMuramatsuSLuYDelayed delivery of AAV-GDNF prevents nigral neurodegeneration and promotes functional recovery in a rat model of Parkinson’s diseaseGene Ther20029638138911960314
  • MandelSWeinrebOAmitTYoudimMBMechanism of neuroprotective action of the anti-Parkinson drug rasagiline and its derivativesBrain Res Brain Res Rev200548237938715850677
  • MaruyamaWAkaoYCarrilloMCKitaniKYoudimMBNaoiMNeuroprotection by propargylamines in Parkinson’s disease: suppression of apoptosis and induction of prosurvival genesNeurotoxicol Teratol200224567568212200198
  • CarrilloMCMinamiCKitaniKEnhancing effect of rasagiline on superoxide dismutase and catalase activities in the dopaminergic system in the ratLife Sci200067557758510993123
  • AkaoYMaruyamaWShimizuSMitochondrial permeability transition mediates apoptosis induced by N-methyl(R) salsolinol, an endogenous neurotoxin, and is inhibited by Bcl-2 and rasagiline, N-propargyl-1(R)-aminoindanJ Neurochem200282491392312358797
  • YoudimMBAmitTFalach-YogevMBar AmOMaruyamaWNaoiMThe essentiality of Bcl-2, PKC and proteasome-ubiquitin complex activations in the neuroprotective-antiapoptotic action of the anti-Parkinson drug, rasagilineBiochem Pharmacol20036681635164114555244
  • MaruyamaWAkaoYYoudimMBDavisBANaoiMTransfection-enforced Bcl-2 overexpression and an anti-Parkinson drug, rasagiline, prevent nuclear accumulation of glyceraldehyde-3-phosphate dehydrogenase induced by an endogenous dopaminergic neurotoxin, N-methyl(R)salsolinolJ Neurochem200178472773511520893
  • WeinrebOBar-AmOAmitTChillag-TalmorOYoudimMBNeuroprotection via pro-survival protein kinase C isoforms associated with Bcl-2 family membersFASEB J200418121471147315247150
  • RabeyJMSagiIHubermanMRasagiline mesylate, a new MAO-B inhibitor for the treatment of Parkinson’s disease: a doubleblind study as adjunctive therapy to levodopaClin Neuropharmacol200023632433011575866
  • SternMBMarekKLFriedmanJDouble-blind, randomized, controlled trial of rasagiline as monotherapy in early Parkinson’s disease patientsMov Disord200419891692315300656
  • FreedmanNMMishaniEKrauszYIn vivo measurement of brain monoamine oxidase B occupancy by rasagiline, using (11)C-l-deprenyl and PETJ Nucl Med200546101618162416204711
  • KupschASautterJGotzMEMonoamine oxidase-inhibition and MPTP-induced neurotoxicity in the non-human primate: comparison of rasagiline (TVP 1012) with selegilineJ Neural Transm20011088–9985100911716151
  • HeikkilaREDuvoisinRCFinbergJPYoudimMBPrevention of MPTP-induced neurotoxicity by AGN-1133 and AGN-1135, selective inhibitors of monoamine oxidase-BEur J Pharmacol198511633133173935467
  • OlanowCWRascolOHauserRA double-blind, delayedstart trial of rasagiline in Parkinson’s diseaseN Engl J Med2009361131268127819776408
  • HurleyDFDA committee: Rasagiline rejected as ‘disease modifying’ for Parkinson Disease20111121167
  • Parkinson Study GroupA controlled trial of rasagiline in early Parkinson disease: the TEMPO StudyArch Neurol200259121937194312470183
  • Parkinson Study GroupA controlled, randomized, delayedstart study of rasagiline in early Parkinson diseaseArch Neurol200461456156615096406
  • FahnSOakesDShoulsonILevodopa and the progression of Parkinson’s diseaseN Engl J Med2004351242498250815590952
  • HauserRLewMHurtigHOndoWWojcieszekJEarly treatment with rasagiline is more beneficial than delayed treatment start in the long-term management of Parkinson’s disease: P251Mov Disord200520S75
  • Parkinson Study GroupEffect of lazabemide on the progression of disability in early Parkinson’s diseaseAnn Neurol1996401991078687199
  • ShultsCWOakesDKieburtzKEffects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional declineArch Neurol200259101541155012374491
  • RascolOBrooksDJMelamedERasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomized, double-blind, parallel-group trialLancet2005365946394795415766996
  • Parkinson Study GroupA randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO studyArch Neurol200562224124815710852
  • MontastrucJLChaumerliacCDesboeufKAdverse drug reactions to selegiline: a review of the French pharmacovigilance databaseClin Neuropharmacol200023527127511154095
  • AbassiZABinahOYoudimMBCardiovascular activity of rasagiline, a selective and potent inhibitor of mitochondrial monoamine oxidase B: comparison with selegilineBr J Pharmacol2004143337137815339864
  • GoetzCGSchwidSEberlySSafety of rasagiline in elderly patients with Parkinson diseaseNeurology2006661427142916682679
  • Da PradaMZurcherGWuthrichIHaefelyWEOn tyramine, food, beverages and the reversible MAO inhibitor moclobemideJ Neural Transm Suppl19882631563283290
  • Parkinson Study GroupTyramine challenge to assess the safety of rasagiline monotherapy in a placebo-controlled multicenter trial for early Parkinson’s disease (the TEMPO Study)Neurology200156Suppl 3A345
  • ZornbergGLBodkinJACohenBMSevere adverse interaction between pethidine and selegilineLancet199133787352461670882
  • ChenJJLyAVRasagiline: A second-generation monoamine oxidase type-B inhibitor for the treatment of Parkinson’s diseaseAm J Health Syst Pharm2006631091592816675649
  • Azilect.com [homepage on the Internet]Anti-depressant chart reviewTeva Neuroscience, Inc2010 Available from: http://www.azilect.com/HCP/Tools-Resources/Anti-Depressant-Fact-Card.aspxAccessed February 13, 2012
  • ChenJJSwopeDMClinical pharmacology of rasagiline: a novel, second-generation propargylamine for the treatment of Parkinson diseaseJ Clin Pharmacol200545887889416027398
  • Azilect.com [homepage on the Internet]Azilect dosage: Parkinson’s disease medication dosingTeva Neuroscience2010 Available from: http://www.azilect.com/HCP/Azilect-Therapy/Dosing.aspxAccessed February 13, 2012
  • HanagasiHAGurvitHUnsalanPThe effects of rasagiline on cognitive deficits in Parkinson’s disease patients without dementia: a randomized, double-blind, placebo-controlled, multicenter studyMov Disord201126101851185821500280
  • HudryJRinneJOKeranenTEckertLCochranJMCost-utility model of rasagiline in the treatment of advanced Parkinson’s disease in FinlandAnn Pharmacother200640465165716569799