Elsevier

Progress in Neurobiology

Volume 92, Issue 3, November 2010, Pages 330-344
Progress in Neurobiology

Rasagiline: A novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity

https://doi.org/10.1016/j.pneurobio.2010.06.008Get rights and content

Abstract

Rasagiline (N-propargyl-1-(R)-aminoindan) is a novel, highly potent irreversible monoamine oxidase (MAO)-B inhibitor, anti-Parkinsonian drug. Rasagiline is effective as monotherapy or adjunct to l-Dopa for patients with early and late Parkinson's disease (PD). Its S-isomer, TVP1022 is thousand times less potent as an MAO-B inhibitor. However, both compounds have similar molecular mechanisms of neuroprotection in neuronal cell cultures and animal neurodegenerative models, indicating that the neuroprotective effect of rasagiline does not depend on inhibition of MAO-B, but rather is associated with the N-propargyl moiety, which promotes mitochondrial viability and stabilizes permeability transition by regulating Bcl-2 family proteins. Novel findings demonstrated that the major metabolite of rasagiline, 1-(R)-aminoindan has antioxidant and neuroprotective capabilities and thus, may contribute to the overt activity of its parent compound, rasagiline. This paper will review the earlier and present studies in the development of rasagiline for treatment of PD and discuss its pharmacology and applicable mechanism of action.

Research highlights

▶ A potent selective irreversible monoamine oxidase -B inhibitor/anti-Parkinson's drug. ▶ Neuroprotective and neurorestorative activities in vitro and in vivo. ▶ Maintenance of the mitochondria integrity and related proteins and enzymes. ▶ Induction of neurotrophic factors. ▶ Its major metabolite has neuroprotective effects by itself in vitro and in vivo.

Introduction

The knowledge that dopamine (DA) is oxidatively deaminated equally by monoamine oxidase (MAO) types A and B (Collins et al., 1970), the dominance of MAO-B (80%), as compared to MAO-A in the extrapyramidal regions of human brain (Collins et al., 1970, O’Carroll et al., 1983, Squires, 1972) and the absence of “cheese reaction” in whole animal and isolated tissue preparations by the selective irreversible MAO-B inhibitor, l-deprenyl (selegiline) (Sandler et al., 1978), led to introduce the irreversible selective MAO-B inhibitor, selegiline as an adjunct to l-Dopa therapy of Parkinson's disease (PD) (Birkmayer et al., 1977, Birkmayer et al., 1975, Lees et al., 1977). The drug has been useful as an anti-Parkinson drug, both in monotherapy and as adjunct to l-Dopa therapy [the DATATOP (deprenyl and tocopherol antioxidative therapy of Parkinsonism)] (Parkinson Study Group, 1993, Parkinson Study Group, 1996, Parkinson Study Group, 1998, Parkinson Study Group, 1989), and has a l-Dopa sparing action (Birkmayer et al., 1977, Riederer et al., 1978). Selegiline irreversibly inhibits MAO-B by binding, mole per mole, covalently to the N-5 position of the isoalloxazine moiety of flavin adenine dinucleotide (FAD), the cofactor of MAO-B (Maycock et al., 1976, Riederer et al., 1982, Youdim, 1978). Selegiline is a propargylamine-derivative of l-amphetamine, and is metabolized in vivo to its major toxic metabolites l-amphetamine and l-methamphetamine (Reynolds et al., 1978) (Fig. 1). Thus, in pharmacological preparations and in vivo it possesses amphetamine-like sympathomimetic actions, which may result in increased blood pressure and heart rate, although in clinical practice it may not have a significant role compared to all other anti-Parkinsonian drugs (Finberg et al., 1981a, Simpson, 1978).

Based on these studies with selegiline, a new highly potent irreversible selective inhibitor of MAO-B, rasagiline [N-propargyl-1-(R)-aminoindan; TVP1012; Azilect] (Fig. 1), was identified and developed as an anti-Parkinsonian drug (Finberg et al., 1996, Finberg et al., 1999, Kalir et al., 1981, Sabbagh and Youdim, 1978, Sterling et al., 1998, Youdim and Bakhle, 2006, Youdim et al., 2001a). Rasagiline is the R-isomer of the selective MAO-B inhibitor, AGN 1135 (Kalir et al., 1981, Sabbagh and Youdim, 1978). In preclinical studies, rasagiline was shown to possess neuroprotective activity (Heikkila et al., 1985, Maruyama et al., 2001a, Tatton et al., 2002, Weinreb et al., 2004). More prominently, rasagiline induced an in vivo neurorestorative activity in the substantia nigra pars compacta (SNpC) neurons, when given post-N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or lactacystin (proteasome inhibitor) (Sagi et al., 2007, Zhu et al., 2008). This property was shown to be associated with activation of tyrosine kinase (TK) receptor and stimulation of various cell signaling transduction pathways (Sagi et al., 2007). An additional neuroprotective effect, shared by rasagiline and other propargyl derivative compounds is related to their ability to regulate the non-amyloidogenic processing of the Alzheimer's disease (AD) amyloid precursor protein (APP) (Bar-Am et al., 2004b, Yogev-Falach et al., 2002, Yogev-Falach et al., 2003). Thus, rasagiline may induce neuroprotective, neurorescue and long-term potentiating effects. Indeed, recent Phase III delayed-start clinical study in Parkinsonian subjects, ADAGIO (Attenuation of Disease Progression with Azilect Given Once-daily) indicated that rasagiline might be the first drug with the benefits of slowing the disease progression (Olanow et al., 2008, Olanow et al., 2009).

At its selective MAO-B inhibitory dosage, rasagiline does not produce the “cheese reaction”, as was shown in isolated tissue preparations or in vivo in rats and cats (Finberg et al., 1981a, Finberg and Youdim, 1985). Unlike selegiline, rasagiline is primarily metabolized by hepatic cytochrome P-450 isoenzyme 1A2 (CYP1A2)-mediated N-dealkylation to form the non-toxic compound 1-(R)-aminoindan (Fig. 1) (Chen and Swope, 2005, Chen et al., 2007). A kinetic and crystallographic analysis revealed that 1-(R)-aminoindan is not a substrate for MAO oxidation, but conversely, a weak reversible inhibitor (Sterling et al., 1998, Binda et al., 2005). Recent studies indicated the potential neuroprotective effect of 1-(R)-aminoindan, suggesting that it may contribute to the overall neuroprotective and anti-apoptotic effects of rasagiline (Bar-Am et al., 2004a, Bar-Am et al., 2007, Bar-Am et al., 2010). The current review presents findings-based studies of the pharmacology of rasagiline in PD and discusses its applicable functional mechanisms of action.

Section snippets

AGN 1135 as a selective MAO-B inhibitor

The successful use of the MAO-B inhibitor, selegiline [discovered in Hungary in the 1960s (Knoll and Magyar, 1972)], as an adjunct to l-Dopa in the treatment of PD (Birkmayer et al., 1977, Birkmayer et al., 1975, Riederer et al., 1982) or depressive illness (Quitkin et al., 1984), without the occurrence of toxic side-effects, encountered with other MAO inhibitors, led Kalir et al. (1981) to perform structure–activity search for other selective MAO-B inhibitors, using 5-hydroxytryptamine (5-HT)

MAO-B inhibitory activity

MAO-A and -B inhibitory activities of rasagiline and the racemic compound, AGN 1135 have been compared to those of selegiline in vitro and in vivo, in various tissues (e.g. brain, intestine and liver) of mice, rats, cats and monkeys (Youdim and Bakhle, 2006, Youdim et al., 2001a, Youdim et al., 2001b, Kupsch et al., 2001, Lamensdorf et al., 1996, Riederer and Youdim, 1986, Youdim, 2006). Rasagiline and its racemic form were shown to possess highly potent selective irreversible inhibitory

Cell culture and animal models of neurodegeneration

Rasagiline exerts its primary effect in PD presumably by MAO-B inhibition, resulting in a slower metabolism of endogenous and exogenous DA, thus providing symptomatic benefits (Finberg et al., 1996, Finberg et al., 1998). In addition, rasagiline has been shown in preclinical studies, to have broad neuroprotective/neurorestorative activities against a variety of neurotoxins in in vivo and neuronal cell cultures models of neurodegeneration, which may contribute to its possible disease-modifying

Summary and conclusions

Human studies with selegiline in PD patients, reported on improved symptoms and a delay in the need to start l-Dopa, but did not definitively show that selegiline altered the underlying pathophysiology of PD (Parkinson Study Group, 1993, Parkinson Study Group, 1998). However, recent clinical studies in PD patients demonstrated that in addition to its symptomatic benefits, rasagiline slows the underlying progression of PD disability (Olanow et al., 2008, Olanow et al., 2009). The ADAGIO trial

Acknowledgements

The support of Technion Research and Development (Haifa, Israel), Teva Pharmaceutical Co. (Netanya, Israel); Stein Foundation and Friedman Foundation (Philadelphia, USA) are gratefully acknowledged. The permission to use Figures from articles in Annals New York Academy of Science, British Journal of Pharmacology, Nature Review Neuroscience are acknowledged.

References (137)

  • M.K. Hu et al.

    New 1,2,3,4-tetrahydroisoquinoline derivatives as modulators of proteolytic cleavage of amyloid precursor proteins

    Bioorganic & Medicinal Chemistry

    (2008)
  • W. Huang et al.

    Neuroprotective effect of rasagiline, a selective monoamine oxidase-B inhibitor, against closed head injury in the mouse

    European Journal of Pharmacology

    (1999)
  • J. Knoll et al.

    Novel (−)deprenyl-derived selective inhibitors of B-type monoamine oxidase. The relation of structure to their action

    Biochemical Pharmacology

    (1978)
  • A.J. Lees et al.

    Deprenyl in Parkinson's disease

    Lancet

    (1977)
  • W. Linert et al.

    Dopamine, 6-hydroxydopamine, iron, and dioxygen—their mutual interactions and possible implication in the development of Parkinson's disease

    Biochimica et biophysica acta

    (1996)
  • W. Maruyama et al.

    Neuroprotection by propargylamines in Parkinson's disease: suppression of apoptosis and induction of prosurvival genes

    Neurotoxicology and Teratology

    (2002)
  • W. Maruyama et al.

    N-Propargyl-1 (R)-aminoindan, rasagiline, increases glial cell line-derived neurotrophic factor (GDNF) in neuroblastoma SH-SY5Y cells through activation of NF-kappaB transcription factor

    Neurochemistry International

    (2004)
  • W. Maruyama et al.

    Mechanism underlying anti-apoptotic activity of a (−) deprenyl-related propargylamine, rasagiline

    Mechanisms of Ageing and Development

    (2000)
  • J. Mills et al.

    Mitogen-activated protein kinase is involved in N-methyl-d-aspartate receptor regulation of amyloid precursor protein cleavage

    Neuroscience

    (1999)
  • Y. Mizuno et al.

    Role of mitochondria in the etiology and pathogenesis of Parkinson's disease

    Biochimica et biophysica acta

    (1995)
  • M.G. Murer et al.

    Brain-derived neurotrophic factor in the control human brain, and in Alzheimer's disease and Parkinson's disease

    Progress in Neurobiology

    (2001)
  • X.M. Ou et al.

    A novel role for glyceraldehyde-3-phosphate dehydrogenase and monoamine oxidase B cascade in ethanol-induced cellular damage

    Biological Psychiatry

    (2010)
  • S. Abu-Raya et al.

    Rasagiline, a monoamine oxidase-B inhibitor, protects NGF-different PC12 cells against oxygen–glucose deprivation

    Journal of Neuroscience Research

    (1999)
  • Y. Akao et al.

    Mitochondrial 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)-aminoindan

    Journal of Neurochemistry

    (2002)
  • C.D. Arnett et al.

    Turnover of brain monoamine oxidase measured in vivo by positron emission tomography using l-[11C]deprenyl

    Journal of Neurochemistry

    (1987)
  • A.W. Bach et al.

    cDNA cloning of human liver monoamine oxidase A and B: molecular basis of differences in enzymatic properties

    Proceedings of the National Academy of Sciences of the United States of America

    (1988)
  • G.B. Baker et al.

    Effects of p-chloro-beta-phenylethylamine on the uptake and release of putative amine neurotransmitters in rat brain [proceedings]

    British Journal of Pharmacology

    (1976)
  • O. Bar-Am et al.

    Contrasting neuroprotective and neurotoxic actions of respective metabolites of anti-Parkinson drugs rasagiline and selegiline

    Neuroscience Letters

    (2004)
  • O. Bar-Am et al.

    Aminoindan and hydroxyaminoindan, metabolites of rasagiline and ladostigil, respectively, exert neuroprotective properties in vitro

    Journal of Neurochemistry

    (2007)
  • O. Bar-Am et al.

    Regulation of Bcl-2 family proteins, neurotrophic factors, and APP processing in the neurorescue activity of propargylamine

    FASEB Journal

    (2005)
  • O. Bar-Am et al.

    The neuroprotective mechanism of 1-(R)-aminoindan, the major metabolite of the anti-parkinsonian drug rasagiline

    Journal of Neurochemistry

    (2010)
  • O. Bar-Am et al.

    Regulation of protein kinase C by the anti-Parkinson drug, MAO-B inhibitor, rasagiline and its derivatives, in vivo

    Journal of Neurochemistry

    (2004)
  • C. Binda et al.

    Crystal structures of monoamine oxidase B in complex with four inhibitors of the N-propargylaminoindan class

    Journal of Medicinal Chemistry

    (2004)
  • C. Binda et al.

    Binding of rasagiline-related inhibitors to human monoamine oxidases: a kinetic and crystallographic analysis

    Journal of Medicinal Chemistry

    (2005)
  • W. Birkmayer et al.

    The potentiation of the anti akinetic effect after l-dopa treatment by an inhibitor of MAO-B, deprenil

    Journal of Neural Transmission

    (1975)
  • J.J. Chen et al.

    Rasagiline: a second-generation monoamine oxidase type-B inhibitor for the treatment of Parkinson's disease

    American Journal of Health System Pharmacy

    (2006)
  • J.J. Chen et al.

    Clinical pharmacology of rasagiline: a novel, second-generation propargylamine for the treatment of Parkinson disease

    Journal of Clinical Pharmacology

    (2005)
  • V.W. Chow et al.

    An overview of APP processing enzymes and products

    Neuromolecular Medicine

    (2010)
  • G.G. Collins et al.

    Multiple forms of human brain mitochondrial monoamine oxidase

    Nature

    (1970)
  • J.A. deMarcaida et al.

    Effects of tyramine administration in Parkinson's disease patients treated with selective MAO-B inhibitor rasagiline

    Movement Disorders

    (2006)
  • M. Ebadi et al.

    Neuroprotective actions of selegiline

    Journal of Neuroscience Research

    (2002)
  • S. Eliash et al.

    Rasagiline and its (S) enantiomer increase survival and prevent stroke in salt-loaded stroke-prone spontaneously hypertensive rats

    Journal of Neural Transmission

    (2001)
  • J.D. Elsworth et al.

    Deprenyl administration in man: a selective monoamine oxidase B inhibitor without the ‘cheese effect’

    Psychopharmacology

    (1978)
  • J.P. Finberg et al.

    Pharmacology and neuroprotective properties of rasagiline

    Journal of Neural Transmission

    (1996)
  • J.P. Finberg et al.

    Pharmacology of rasagiline (N-propargyl-1R-aminoindan)

    Advances in Neurology

    (1999)
  • J.P. Finberg et al.

    Tyramine antagonistic properties of AGN 1135, an irreversible inhibitor of monoamine oxidase type B

    British Journal of Pharmacology

    (1981)
  • J.P. Finberg et al.

    Increased striatal dopamine production from l-DOPA following selective inhibition of monoamine oxidase B by R(+)-N-propargyl-1-aminoindan (rasagiline) in the monkey

    Journal of Neural Transmission

    (1998)
  • J.P. Finberg et al.

    Modification of blood pressure and nictitating membrane response to sympathetic amines by selective monoamine oxidase inhibitors, types A and B, in the cat

    British Journal of Pharmacology

    (1985)
  • J.P.M. Finberg et al.

    Pharmacology of selective propargyl “suicide” inhibitors of monoamine oxidase

  • J.P.M. Finberg et al.

    Selective irreversible propargyl derivative inhibitors of monoamine oxidase (MAO) without the cheese effect

  • Cited by (154)

    • Nanotechnology-empowered therapeutics targeting neurodegenerative diseases

      2023, Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology
    View all citing articles on Scopus
    View full text