Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

LRRK2 in Parkinson disease: challenges of clinical trials

Abstract

One of the most common monogenic forms of Parkinson disease (PD) is caused by mutations in the LRRK2 gene that encodes leucine-rich repeat kinase 2 (LRRK2). LRRK2 mutations, and particularly the most common mutation Gly2019Ser, are observed in patients with autosomal dominant PD and in those with apparent sporadic PD, who are clinically indistinguishable from those with idiopathic PD. The discoveries that pathogenic mutations in the LRRK2 gene increase LRRK2 kinase activity and that small-molecule LRRK2 kinase inhibitors can be neuroprotective in preclinical models of PD have placed LRRK2 at the centre of disease modification efforts in PD. Recent investigations also suggest that LRRK2 has a role in the pathogenesis of idiopathic PD and that LRRK2 therapies might, therefore, be beneficial in this common subtype of PD. In this Review, we describe the characteristics of LRRK2-associated PD that are most relevant to the development of LRRK2-targeted therapies and the design and implementation of clinical trials. We highlight strategies for correcting the effects of mutations in the LRRK2 gene, focusing on how to identify which patients are the optimal candidates and how to decide on the timing of such trials. In addition, we discuss challenges in implementing trials of disease-modifying treatment in people who carry LRRK2 mutations.

Key points

  • Leucine-rich repeat kinase 2 (LRRK2) has emerged as a promising target for disease-modifying Parkinson disease (PD) treatment because patients with LRRK2-associated PD (LRRK2-PD) constitute a homogeneous subgroup with shared underlying pathophysiology.

  • Preclinical studies indicate that reduction of LRRK2 activity or expression is neuroprotective; small-molecule LRRK2 inhibitors and antisense oligonucleotides have been developed and are now considered suitable for clinical exposure.

  • The low penetrance of LRRK2 mutations, a lack of markers that predict conversion to PD and ethical issues preclude implementation of clinical trials in asymptomatic LRRK2 mutation carriers.

  • Challenges in implementation of disease-modification trials in manifest LRRK2-PD include the low frequency of the condition, a lack of validated disease progression markers and insufficient measures of target engagement.

  • Clinical trials of specific LRRK2-targeted therapies with a focus on safety have already started, highlighting the rapid progress made over the past decade in LRRK2 research.

  • LRRK2-PD closely resembles idiopathic PD (iPD), suggesting that unravelling the mechanisms of LRRK2-PD will provide insight into iPD and that LRRK2-targeted therapies could be useful for the treatment of iPD.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Distribution of LRRK2 mutations worldwide.
Fig. 2: Structure and function of LRRK2.
Fig. 3: Potential benefits of disease modification with LRRK2-targeted therapies.
Fig. 4: Timing and challenges for disease-modifying trials in PD.

Similar content being viewed by others

References

  1. Gasser, T., Hardy, J. & Mizuno, Y. Milestones in PD genetics. Mov. Disord. 26, 1042–1048 (2011).

    Article  PubMed  Google Scholar 

  2. Hardy, J. Genetic analysis of pathways to Parkinson disease. Neuron 68, 201–206 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chang, D. et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat. Genet. 49, 1511–1516 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lunati, A., Lesage, S. & Brice, A. The genetic landscape of Parkinson’s disease. Rev. Neurol. 174, 628–643 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Funayama, M. et al. A new locus for Parkinson’s disease (PARK8) maps to chromosome 12p11.2-q13.1. Ann. Neurol. 51, 296–301 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Paisan-Ruiz, C. et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 44, 595–600 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Zimprich, A. et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44, 601–607 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Kett, L. R. & Dauer, W. T. Leucine-rich repeat kinase 2 for beginners: six key questions. Cold Spring Harb. Perspect. Med. 2, a009407 (2012). This article addresses six basic questions about LRRK2 biology as it relates to PD.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. West, A. B. Achieving neuroprotection with LRRK2 kinase inhibitors in Parkinson disease. Exp. Neurol. 298, 236–245 (2017). A detailed outline of the steps needed to achieve neuroprotection with LRRK2 inhibitors.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cookson, M. R. Mechanisms of mutant LRRK2 neurodegeneration. Adv. Neurobiol. 14, 227–239 (2017). A review of the mechanisms by which LRRK2 mutations can be linked to neurodegeneration.

    Article  PubMed  Google Scholar 

  11. Chen, J., Chen, Y. & Pu, J. Leucine-rich repeat kinase 2 in Parkinson’s disease: updated from pathogenesis to potential therapeutic target. Eur. Neurol. 79, 256–265 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Chan, S. L. & Tan, E. K. Targeting LRRK2 in Parkinson’s disease: an update on recent developments. Expert Opin. Ther. Targets 21, 601–610 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Cresto, N. et al. The unlikely partnership between LRRK2 and alpha-synuclein in Parkinson’s disease. Eur. J. Neurosci. 49, 339–363 (2019). This review discusses emergent data that suggest that synuclein and LRRK2 interact in important ways in PD.

    Article  PubMed  Google Scholar 

  14. Shihabuddin, L. S., Brundin, P., Greenamyre, J. T., Stephenson, D. & Sardi, S. P. New frontiers in Parkinson’s disease: from genetics to the clinic. J. Neurosci. 38, 9375–9382 (2018). This article highlights the utility of genetics to understand the pathogenic mechanisms and to develop novel therapeutic approaches for PD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Healy, D. G. et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case–control study. Lancet Neurol. 7, 583–590 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ozelius, L. J. et al. LRRK2 G2019S as a cause of Parkinson’s disease in Ashkenazi Jews. N. Engl. J. Med. 354, 424–425 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Lesage, S. et al. LRRK2 G2019S as a cause of Parkinson’s disease in North African Arabs. N. Engl. J. Med. 354, 422–423 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Bouhouche, A. et al. LRRK2 G2019S mutation: prevalence and clinical features in Moroccans with Parkinson’s disease. Parkinsons Dis. 2017, 2412486 (2017).

    PubMed  PubMed Central  Google Scholar 

  19. Tan, E. K. et al. The G2019S LRRK2 mutation is uncommon in an Asian cohort of Parkinson’s disease patients. Neurosci. Lett. 384, 327–329 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Simon-Sanchez, J. et al. Parkinson’s disease due to the R1441G mutation in Dardarin: a founder effect in the Basques. Mov. Disord. 21, 1954–1959 (2006).

    Article  PubMed  Google Scholar 

  21. Gorostidi, A., Ruiz-Martinez, J., Lopez de Munain, A., Alzualde, A. & Marti Masso, J. F. LRRK2 G2019S and R1441G mutations associated with Parkinson’s disease are common in the Basque country, but relative prevalence is determined by ethnicity. Neurogenetics 10, 157–159 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Satake, W. et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson’s disease. Nat. Genet. 41, 1303–1307 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Simon-Sanchez, J. et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat. Genet. 41, 1308–1312 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nalls, M. A. et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat. Genet. 46, 989–993 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hentati, F. et al. LRRK2 parkinsonism in Tunisia and Norway: a comparative analysis of disease penetrance. Neurology 83, 568–569 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lee, A. J. et al. Penetrance estimate of LRRK2 p.G2019S mutation in individuals of non-Ashkenazi Jewish ancestry. Mov. Disord. 32, 1432–1438 (2017). This study confirms that the penetrance of the Gly2019Ser mutation is 25–42.5% at the age of 80 years in all populations analysed.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ruiz-Martinez, J. et al. Penetrance in Parkinson’s disease related to the LRRK2 R1441G mutation in the Basque country (Spain). Mov. Disord. 25, 2340–2345 (2010).

    Article  PubMed  Google Scholar 

  28. Aasly, J. O. et al. Clinical features of LRRK2-associated Parkinson’s disease in central Norway. Ann. Neurol. 57, 762–765 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Gaig, C. et al. LRRK2 mutations in Spanish patients with Parkinson disease: frequency, clinical features, and incomplete penetrance. Arch. Neurol. 63, 377–382 (2006).

    Article  PubMed  Google Scholar 

  30. Marras, C. et al. Phenotype in parkinsonian and nonparkinsonian LRRK2 G2019S mutation carriers. Neurology 77, 325–333 (2011). This study shows that tremor is a more common presenting feature of LRRK2-PD than of iPD and that some nonmotor features are less prominent, but that the phenotypes of these two conditions largely overlap.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, C. et al. Clinical profiles of Parkinson’s disease associated with common leucine-rich repeat kinase 2 and glucocerebrosidase genetic variants in Chinese individuals. Neurobiol. Aging 35, 725.e1–6 (2014).

    Article  CAS  Google Scholar 

  32. Somme, J. H. et al. Cognitive and behavioral symptoms in Parkinson’s disease patients with the G2019S and R1441G mutations of the LRRK2 gene. Parkinsonism Relat. Disord. 21, 494–499 (2015).

    Article  PubMed  Google Scholar 

  33. Marras, C. et al. Motor and nonmotor heterogeneity of LRRK2-related and idiopathic Parkinson’s disease. Mov. Disord. 31, 1192–1202 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Nishioka, K. et al. A comparative study of LRRK2, PINK1 and genetically undefined familial Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 81, 391–395 (2010).

    Article  PubMed  Google Scholar 

  35. Gan-Or, Z. et al. LRRK2 mutations in Parkinson disease; a sex effect or lack thereof? A meta-analysis. Parkinsonism Relat. Disord. 21, 778–782 (2015).

    Article  PubMed  Google Scholar 

  36. Kestenbaum, M. & Alcalay, R. N. Clinical features of LRRK2 carriers with Parkinson’s disease. Adv. Neurobiol. 14, 31–48 (2017).

    Article  PubMed  Google Scholar 

  37. Trinh, J. et al. Genotype-phenotype relations for the Parkinson’s disease genes SNCA, LRRK2, VPS35: MDSGene systematic review. Mov. Disord. 33, 1857–1870 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Alcalay, R. N. et al. Parkinson disease phenotype in Ashkenazi Jews with and without LRRK2 G2019S mutations. Mov. Disord. 28, 1966–1971 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Saunders-Pullman, R. et al. Progression in the LRRK2-asssociated Parkinson disease population. JAMA Neurol. 75, 312–319 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Nabli, F. et al. Motor phenotype of LRRK2-associated Parkinson’s disease: a Tunisian longitudinal study. Mov. Disord. 30, 253–258 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Rizzone, M. G., Martone, T., Balestrino, R. & Lopiano, L. Genetic background and outcome of deep brain stimulation in Parkinson’s disease. Parkinsonism Relat. Disord. 64, 8–19 (2018).

    Article  PubMed  Google Scholar 

  42. Gaig, C. et al. Nonmotor symptoms in LRRK2 G2019S associated Parkinson’s disease. PLOS ONE 9, e108982 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Bergareche, A. et al. DAT imaging and clinical biomarkers in relatives at genetic risk for LRRK2 R1441G Parkinson’s disease. Mov. Disord. 31, 335–343 (2016).

    Article  PubMed  Google Scholar 

  44. Ruiz-Martinez, J. et al. Olfactory deficits and cardiac 123I-MIBG in Parkinson’s disease related to the LRRK2 R1441G and G2019S mutations. Mov. Disord. 26, 2026–2031 (2011).

    Article  PubMed  Google Scholar 

  45. Pont-Sunyer, C. et al. Sleep disorders in parkinsonian and nonparkinsonian LRRK2 mutation carriers. PLOS ONE 10, e0132368 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Ehrminger, M. et al. Sleep aspects on video-polysomnography in LRRK2 mutation carriers. Mov. Disord. 30, 1839–1843 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Alcalay, R. N. et al. Neuropsychological performance in LRRK2 G2019S carriers with Parkinson’s disease. Parkinsonism Relat. Disord. 21, 106–110 (2015).

    Article  PubMed  Google Scholar 

  48. Tolosa, E., Gaig, C., Santamaria, J. & Compta, Y. Diagnosis and the premotor phase of Parkinson disease. Neurology 72, S12–S20 (2009).

    Article  PubMed  Google Scholar 

  49. Noyce, A. J. et al. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann. Neurol. 72, 893–901 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Salat, D., Noyce, A. J., Schrag, A. & Tolosa, E. Challenges of modifying disease progression in prediagnostic Parkinson’s disease. Lancet Neurol. 15, 637–648 (2016). A discussion of the main issues of diagnosis and disease modification in prediagnostic PD.

    Article  PubMed  Google Scholar 

  51. Brockmann, K. et al. Clinical and brain imaging characteristics in leucine-rich repeat kinase 2-associated PD and asymptomatic mutation carriers. Mov. Disord. 26, 2335–2342 (2011).

    Article  PubMed  Google Scholar 

  52. Mirelman, A. et al. Nonmotor symptoms in healthy Ashkenazi Jewish carriers of the G2019S mutation in the LRRK2 gene. Mov. Disord. 30, 981–986 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pont-Sunyer, C. et al. The prodromal phase of leucine-rich repeat kinase 2-associated Parkinson disease: clinical and imaging studies. Mov. Disord. 32, 726–738 (2017). In this study of the LRRK2 Cohort Consortium (Michael J. Fox Foundation), Gly2019Ser LRRK2 mutation carriers score higher on motor tests than non-carriers, and dopamine transporter radioligand uptake in carriers is lower, but no differences in nonmotor symptoms are observed.

    Article  CAS  PubMed  Google Scholar 

  54. Sierra, M. et al. Olfaction and imaging biomarkers in premotor LRRK2 G2019S-associated Parkinson disease. Neurology 80, 621–626 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Postuma, R. B., Lang, A. E., Gagnon, J. F., Pelletier, A. & Montplaisir, J. Y. How does parkinsonism start? Prodromal parkinsonism motor changes in idiopathic REM sleep behaviour disorder. Brain 135, 1860–1870 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Mirelman, A. et al. Arm swing as a potential new prodromal marker of Parkinson’s disease. Mov. Disord. 31, 1527–1534 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Arora, S. et al. Investigating voice as a biomarker for leucine-rich repeat kinase 2-associated Parkinson’s disease. J. Parkinsons Dis. 8, 503–510 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Berg, D. et al. MDS research criteria for prodromal Parkinson’s disease. Mov. Disord. 30, 1600–1611 (2015).

    Article  PubMed  Google Scholar 

  59. Mirelman, A. et al. Application of the movement disorder society prodromal criteria in healthy G2019S-LRRK2 carriers. Mov. Disord. 33, 966–973 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kalia, L. V. et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol. 72, 100–105 (2015). This study of clinicopathological correlations demonstrates that Lewy body pathology in LRRK2-related PD could be a marker for nonmotor symptoms.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Vilas, D. et al. Clinical and neuropathological features of progressive supranuclear palsy in leucine rich repeat kinase (LRRK2) G2019S mutation carriers. Mov. Disord. 33, 335–338 (2018).

    Article  PubMed  Google Scholar 

  62. Poulopoulos, M., Levy, O. A. & Alcalay, R. N. The neuropathology of genetic Parkinson’s disease. Mov. Disord. 27, 831–842 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Rudi, K. et al. Conformational heterogeneity of the Roc domains in C. tepidum Roc-COR and implications for human LRRK2 Parkinson mutations. Biosci. Rep. 35, e00254 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Cook, D. A. et al. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis. 3, 11 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Fuji, R. N. et al. Effect of selective LRRK2 kinase inhibition on nonhuman primate lung. Sci. Transl Med. 7, 273ra215 (2015). The results of preclinical toxicity studies of LRRK2 kinase inhibitors in rodents and nonhuman primates.

    Article  CAS  Google Scholar 

  66. Denali Therapeutics. Our pipeline. Denali https://denalitherapeutics.com/pipeline (2019).

  67. Rudenko, I. N., Chia, R. & Cookson, M. R. Is inhibition of kinase activity the only therapeutic strategy for LRRK2-associated Parkinson’s disease? BMC Med. 10, 20 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhao, H. T. et al. LRRK2 antisense oligonucleotides ameliorate alpha-synuclein inclusion formation in a Parkinson’s disease mouse model. Mol. Ther. Nucleic Acids 8, 508–519 (2017). This study shows that administration of LRRK2 ASOs to the brain reduces LRRK2 protein levels and fibril-induced α-synuclein inclusions, and suggests LRRK2 ASOs as a therapeutic strategy for prevention of LRRK2-PD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Herzig, M. C. et al. LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice. Hum. Mol. Genet. 20, 4209–4223 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. US National Library of Medicine. ClinicalTrials.gov http://www.clinicaltrials.gov/ct2/show/NCT03976349 (2019).

  71. Alarcon-Aris, D. et al. Selective alpha-synuclein knockdown in monoamine neurons by intranasal oligonucleotide delivery: potential therapy for Parkinson’s disease. Mol. Ther. 26, 550–567 (2018).

    Article  CAS  PubMed  Google Scholar 

  72. US Food and Drug Administration. Developing products for rare diseases and conditions (FDA, 2018).

  73. Hartlova, A. et al. LRRK2 is a negative regulator of Mycobacterium tuberculosis phagosome maturation in macrophages. EMBO J. 37, e98694 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Bonet-Ponce, L. & Cookson, M. R. The role of Rab GTPases in the pathobiology of Parkinson’ disease. Curr. Opin. Cell Biol. 59, 73–80 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Alessi, D. R. & Sammler, E. LRRK2 kinase in Parkinson’s disease. Science 360, 36–37 (2018).

    Article  CAS  PubMed  Google Scholar 

  76. Steger, M. et al. Systematic proteomic analysis of LRRK2-mediated Rab GTPase phosphorylation establishes a connection to ciliogenesis. eLife 6, e31012 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Steger, M. et al. Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases. eLife 5, e12813 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Sheng, Z. et al. Ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations. Sci. Transl Med. 4, 164ra161 (2012).

    Article  PubMed  CAS  Google Scholar 

  79. Garrido, A. et al. Alpha-synuclein RT-QuIC in cerebrospinal fluid of LRRK2 linked Parkinson’s disease. Ann. Clin. Transl Neurol. 6, 1024–1032 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Di Maio, R. et al. LRRK2 activation in idiopathic Parkinson’s disease. Sci. Transl Med. 10, eaar5429 (2018). This study shows that wild-type LRRK2 kinase activity is selectively increased in substantia nigra dopaminergic neurons in postmortem brain tissue from patients with iPD, suggesting that LRRK2 has a role in iPD.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Cho, H. J. et al. MicroRNA-205 regulates the expression of Parkinson’s disease-related leucine-rich repeat kinase 2 protein. Hum. Mol. Genet. 22, 608–620 (2013).

    Article  CAS  PubMed  Google Scholar 

  82. Henderson, M. X. et al. LRRK2 inhibition does not impart protection from alpha-synuclein pathology and neuron death in non-transgenic mice. Acta Neuropathol. Commun. 7, 28 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Athauda, D. & Foltynie, T. Challenges in detecting disease modification in Parkinson’s disease clinical trials. Parkinsonism Relat. Disord. 32, 1–11 (2016). A review of ‘structural issues’ that are often not addressed after failed trials of disease-modification therapies, and a discussion of directions to overcome these challenges.

    Article  PubMed  Google Scholar 

  84. Lang, A. E. & Espay, A. J. Disease modification in parkinson’s disease: current approaches, challenges, and future considerations. Mov. Disord. 33, 660–677 (2018).

    Article  PubMed  Google Scholar 

  85. Ns-Park. Organization of Ns-Park network. Ns-Park https://parkinson.network/fr/fonctionnement/organisation-fonctionnement-du-reseau-ns-park (2019).

  86. NINDS Exploratory Trial in Parkinson Disease (NET-PD) FS-ZONE Investigators. Pioglitazone in early Parkinson’s disease: a phase 2, multicentre, double-blind, randomised trial. Lancet Neurol. 14, 795–803 (2015).

    Article  CAS  Google Scholar 

  87. Martin-Bastida, A. et al. Brain iron chelation by deferiprone in a phase 2 randomised double-blinded placebo controlled clinical trial in Parkinson’s disease. Sci. Rep. 7, 1398 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Carroll, C. B. & Wyse, R. K. H. Simvastatin as a potential disease-modifying therapy for patients with Parkinson’s disease: rationale for clinical trial, and current progress. J. Parkinsons Dis. 7, 545–568 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Athauda, D. et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet 390, 1664–1675 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Nukada, H., Kowa, H., Saitoh, T., Tazaki, Y. & Miura, S. A big family of paralysis agitans (author’s transl) [Japanese]. Rinsho Shinkeigaku 18, 627–634 (1978).

    CAS  PubMed  Google Scholar 

  91. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03866603 (2019).

  92. Denali Therapeutics. CENTOGENE and Denali Therapeutics announce strategic collaboration to recruit LRRK2 patients for clinical trials. GlobeNewswire https://www.globenewswire.com/news-release/2018/10/03/1600791/0/en/CENTOGENE-and-Denali-Therapeutics-Announce-Strategic-Collaboration-to-Recruit-LRRK2-Patients-for-Clinical-Trials.html (2019).

  93. Fraser, K. B. et al. Ser(P)-1292 LRRK2 in urinary exosomes is elevated in idiopathic Parkinson’s disease. Mov. Disord. 31, 1543–1550 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang, S. & West, A. B. Caught in the act: LRRK2 in exosomes. Biochem. Soc. Trans. 47, 663–670 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Chen, Z. et al. Synthesis and preliminary evaluation of [11C]GNE-1023 as a potent PET probe for imaging leucine-rich repeat kinase 2 (LRRK2) in Parkinson’s disease. ChemMedChem 14, 1580–1585 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wang, M., Gao, M., Xu, Z. & Zheng, Q. H. Synthesis of [11C]HG-10-102-01 as a new potential PET agent for imaging of LRRK2 enzyme in Parkinson’s disease. Bioorg. Med. Chem. Lett. 27, 1351–1355 (2017).

    Article  CAS  PubMed  Google Scholar 

  97. Malik, N., Gifford, A. N., Sandell, J., Tuchman, D. & Ding, Y. S. Synthesis and in vitro and in vivo evaluation of [3H]LRRK2-IN-1 as a novel radioligand for LRRK2. Mol. Imaging Biol. 19, 837–845 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Sardi, S. P., Cedarbaum, J. M. & Brundin, P. Targeted therapies for Parkinson’s disease: from genetics to the clinic. Mov. Disord. 33, 684–696 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Berg, D. et al. Echogenicity of the substantia nigra: association with increased iron content and marker for susceptibility to nigrostriatal injury. Arch. Neurol. 59, 999–1005 (2002).

    Article  PubMed  Google Scholar 

  100. Jennings, D. et al. Imaging prodromal Parkinson disease: the Parkinson Associated Risk Syndrome Study. Neurology 83, 1739–1746 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Iranzo, A. et al. Serial dopamine transporter imaging of nigrostriatal function in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 10, 797–805 (2011).

    Article  CAS  PubMed  Google Scholar 

  102. Sierra, M. et al. Prospective clinical and DaT-SPECT imaging in premotor LRRK2 G2019S-associated Parkinson disease. Neurology 89, 439–444 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Reetz, K. et al. Structural imaging in the presymptomatic stage of genetically determined parkinsonism. Neurobiol. Dis. 39, 402–408 (2010).

    Article  CAS  PubMed  Google Scholar 

  104. Thaler, A. et al. Neural correlates of executive functions in healthy G2019S LRRK2 mutation carriers. Cortex 49, 2501–2511 (2013).

    Article  PubMed  Google Scholar 

  105. Vilas, D. et al. Assessment of alpha-synuclein in submandibular glands of patients with idiopathic rapid-eye-movement sleep behaviour disorder: a case–control study. Lancet Neurol. 15, 708–718 (2016).

    Article  CAS  PubMed  Google Scholar 

  106. van Nuenen, B. F. et al. Cerebral pathological and compensatory mechanisms in the premotor phase of leucine-rich repeat kinase 2 parkinsonism. Brain 135, 3687–3698 (2012).

    Article  PubMed  Google Scholar 

  107. Helmich, R. C. et al. Reorganization of corticostriatal circuits in healthy G2019S LRRK2 carriers. Neurology 84, 399–406 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Ceravolo, R. et al. Nigral anatomy and striatal denervation in genetic parkinsonism: a family report. Mov. Disord. 30, 1148–1149 (2015).

    Article  PubMed  Google Scholar 

  109. Du, G., Lewis, M. M., Sica, C., Kong, L. & Huang, X. Magnetic resonance T1w/T2w ratio: a parsimonious marker for Parkinson disease. Ann. Neurol. 85, 96–104 (2019).

    Article  PubMed  Google Scholar 

  110. Vilas, D. et al. Nigral and striatal connectivity alterations in asymptomatic LRRK2 mutation carriers: a magnetic resonance imaging study. Mov. Disord. 31, 1820–1828 (2016).

    Article  CAS  PubMed  Google Scholar 

  111. Adler, C. H. et al. Submandibular gland needle biopsy for the diagnosis of Parkinson disease. Neurology 82, 858–864 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Iranzo, A. et al. Alpha-synuclein aggregates in labial salivary glands of idiopathic rapid eye movement sleep behavior disorder. Sleep 41, zsy101 (2018).

    Google Scholar 

  113. Doppler, K. et al. Dermal phospho-alpha-synuclein deposition in patients with Parkinson’s disease and mutation of the glucocerebrosidase gene. Front. Neurol. 9, 1094 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank H. Madoev for assistance with MDSGene programming and the design of Fig. 1 and C. Perier for the design of Figs 2–4.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to all aspects of manuscript preparation.

Corresponding author

Correspondence to Eduardo Tolosa.

Ethics declarations

Competing interests

E.T. has received research grants from the Spanish Network on Neurodegenerative Disorders (CIBERNED, Instituto de Salud Carlos III) and the Michael J. Fox Foundation for Parkinson’s Research (USA), and has received consultation fees from Denali Therapeutics. M.V. has received research grants from the Ministry of Economy and Competitiveness (MINECO, Spain), the Michael J. Fox Foundation for Parkinson’s Research (USA) and La Caixa Banking Foundation (Spain). C.K. is a medical adviser to CENTOGENE for genetic testing reports in the fields of movement disorders and dementia, excluding Parkinson disease. O.R. has received honorarium for scientific advice to companies developing neuroprotective therapies for Parkinson disease, including Biogen, Cerespir, Denali Therapeutics, Roche and Sanofi. O.R. has received grants for research programmes in neuroprotection for Parkinson disease from the French Ministry of Health (Programme Hospitalier de Recherche Clinique), The European Commission (FP7, H2020 programmes) and the Cure Parkinson UK Foundation.

Additional information

Peer review information

Nature Reviews Neurology thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related link

MDSGene database: www.mdsgene.org

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tolosa, E., Vila, M., Klein, C. et al. LRRK2 in Parkinson disease: challenges of clinical trials. Nat Rev Neurol 16, 97–107 (2020). https://doi.org/10.1038/s41582-019-0301-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41582-019-0301-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing