A Critical Evaluation of Disease-Modifying Strategies in Parkinson Disease

Publication
Article
NeurologyLiveApril 2021
Volume 4
Issue 2

A trio of clinicians from Georgetown University explore the current status of disease-modifying therapies for individuals with Parkinson disease.

Fernando L. Pagan, MD, Vice Chairman, Department of Neurology; Director, Movement Disorders Program; Medical Director, Georgetown University Hospital National Parkinsonism Foundation Center of Excellence; Medical Director, Translational Neurotherapeutics Program; Associate Professor, Department of Neurology, Georgetown University

Fernando L. Pagan, MD

A NUMBER OF DISEASE-MODIFYING THERAPIES have been investigated in Parkinson disease (PD), and they all encountered a common roadblock: overcoming the efficacy of levodopa. Major obstacles are evident in the inability to translate preclinical data into therapies and in the failure to identify reliable biomarkers. Novel approaches like immunotherapies, gene therapies, and tyrosine kinase inhibition provide alternate methods for the development of disease modification in PD.

The Valley of Death: From Preclinical Mechanisms to Clinical Effect

Previous PD research efforts focused on motor function and symptoms, although PD includes complexities beyond these. The Neuroprotective Exploratory Trials in PD (NET-PD) program intended to accelerate therapeutic evaluation in PD trials.1 Preclinical animal data provided strong justification for clinical investigation into more than 80 compounds.2,3 The incentives for initiation of NET-PD were the relatively good drug safety profiles. These drugs were studied in concurrent clinical trials, but ultimately, they led to collective failure. Several shortcomings have since been noted that strongly dispute the supposed rigor of NET-PD studies, including lack of biomarkers that could be reliably measured in animals and the noninclusion of patients with PD to prove target engagement and drug effects. Pharmacological diligence and drug dose selection were absent—most NET-PD studies tested a single-dose level—suggesting that potential clinical or biomarker effects were missed. Some studies progressed to phase 3, such as SURE-PD3 and STEADY-PD III,4,5 but these late-stage studies showed no effect on the primary motor outcome.

Collectively, these studies were predominantly early-stage, lacked a specific biomarker, did not employ satisfactory dose selection strategies, and, overall, focused on motor outcomes over a short period of time (<12 months) in levodopa-treated PD patients. The accelerated NET-PD studies were a “rushed effort” that omitted adequate pharmacological scrutiny and aimed to show the efficacy of neuroprotective agents on motor symptoms compared with levodopa.

The Conundrum: Biomarkers and Clinical Effects

Charbel Moussa, MBBS, PhD, Associate Professor of Neurology; Director, Laboratory for Dementia and Parkinsonism; Scientific and Clinical Research Director, Translational Neurotherapeutics Program, National Parkinsons’s Foundation Center; Director, Neurosciences Grand Rounds; Director, Lewy Body Dementia Research Center of Excellence, Department of Neurology, Georgetown University

Charbel Moussa, MBBS, PhD

PD involves motor and nonmotor symptoms, loss of dopaminergic neurons, and accumulation of misfolded alpha-synuclein (αS) oligomers.6-8 Research in PD is lagging that of other neurodegenerative diseases, like Alzheimer disease (AD), owing to the availability in those other diseases of dopaminergic and other symptomatic therapies. In idiopathic PD, a biomarker system to evaluate investigational disease-modifying drug effects is absent. Conversely, in AD, novel drug candidates rely on the amyloid/tau/ neurodegeneration system that may collectively represent pathological changes in the AD brain.9

PD is a progressive, multisymptomatic disease that requires increasingly more medications as the disease worsens. The primary limitation in phase 2 and 3 PD trials is emphasis on a short-term motor outcome in patients who are already medicated. Another common limitation of disease modification in PD trials is lack of treatment-specific target engagement and related biomarkers. PD treatment is unlikely to be similar to that of, say, hypertension (eg, a readout of the mean arterial pressure indicates immediate drug response), but an effective PD treatment would create a prolonged biological process that should halt or reverse neuropathology. Therefore, both earlier intervention in biomarker-confirmed PD and longer treatment in drug-naïve patients are needed to demonstrate motor and nonmotor benefit. Alternatively, medically optimized patients must be treated significantly longer compared with predicted placebo and/or standard of care (eg, levodopa, dopamine agents, etc) to observe the effects of long-term therapies that modify multiple pathologies integral to the “brain motor system,” including dopamine loss, oligomeric αS, and hyperphosphorylated tau (p-tau). As in AD, measurement of these biomarkers together as a “mix” may help to evaluate the efficacy of disease-modifying drugs.

The diagnostic error at the onset of motor symptoms is often significant; PD can be confused with other alpha-synucleinopathies such as Lewy body dementia (LBD) and primary tauopathies. Studying motor symptoms without biomarker confirmation is detrimental to clinical trials that exclusively focus on motor effects. The limitations of motor assessments and of scales like the Unified Parkinson’s Disease Rating Scale (UPDRS) must be recognized.10 For instance, the UPDRS should be performed together with developing technologies like wearable devices that could provide objective measures.

In addition to motor assessment, biomarkers should be used as measures of underlying biology for subtyping.11 For example, targeting leucine-rich repeat kinase 2–associated PD12 and glucocerebrosidase (GBA)-associated PD13 are efforts in the right direction within genetically defined conditions. αS gene duplication, multiplication, and mutations are causal genetic factors in familial PD.14 The cerebrospinal fluid (CSF) levels of αS oligomers increase in PD, while total αS decreases, compared with age-matched controls.15-17 These data provide an important distinction between normal, natively unfolded αS (referred to as total levels), which has an essential housekeeping function, and misfolded oligomers, which may form toxic fibrils and induce Lewy body pathology. The toxicity of αS is due to the transitional oligomers that are the building blocks of fibrils,18 and these transitional oligomers are measurable in the CSF of those with PD. Furthermore, p-tau leads to motor and nonmotor symptoms that may overlap with those of PD, and p-tau should be considered to be integral to a PD biomarker mix that represents the anatomical and molecular basis of motor and nonmotor symptoms in PD.

Studying Dopamine Metabolism as a Biomarker

In animal models, the accumulation of αS aggregates impairs dopamine transmission, but eliminating these aggregates enhances dopamine release and utilization in the nigrostriatum.19 Reducing αS oligomers results in improved dopaminergic neuron activity in PD patients20; therefore, simultaneous measurement of oligomeric αS and dopamine is an index of functional activity of dopamine neurons. Total and oligomeric αS as well as 2 dopamine metabolites—homovanillic acid (HVA) and 3,4-dihydroxyphenylacetic acid (DOPAC)—are robustly measured in the CSF of patients with PD.20 The brain contributes 10% to 15% of the plasma levels of HVA21; most of the circulating levels of HVA come from intestinal metabolism of dopamine, while DOPAC is a principal metabolite of brain dopamine and is transported from the brain to the plasma for excretion.22 Therefore, dopamine utilization and metabolism should be measured via concurrent CSF and plasma levels of HVA and DOPAC to indicate drug disease-modifying effects.

Tyrosine Kinase Inhibition

Yasar Torres-Yaghi, MD, Attending Physician, Movement Disorders; Assistant Professor, Department of Neurology, Georgetown University

Yasar Torres-Yaghi, MD

In 2014, a seminal report showed the effects of an oncologic agent called nilotinib (Tasigna; Novartis) as a potential treatment in PD.23 Nilotinib lowers αS, including oligomers; reduces p-tau, via autophagy; and improves dopamine metabolism. A phase 1 study in late-stage (Hoehn &Yahr stages 4-5) levodopa-unresponsive PD and LBD patients demonstrated the feasibility of nilotinib.24 Nilotinib potently targets discoidin domain receptor (DDR)-1 at IC50 of 1 nM and much less potently inhibits Abelson (c-Abl) at 20 nM. It is FDA approved for chronic myelogenous leukemia.25

Phase 2 studies investigated the effects of nilotinib in medically optimized patients with moderately severe PD for 6 months,26 12 months,20 and 27 months.27 These phase 2 studies were underpowered for clinical outcomes, but they all met their primary objectives, demonstrating that nilotinib is safe. However, significant differences existed among these studies. Pagan et al showed that CSF nilotinib concentration is adequate to inhibit DDR1, but not c-Abl, and nilotinib treatment stabilized total αS, partially reduced oligomeric αS and lowered p-tau in a dose-dependent manner.20 Simuni et al, however, did not explore nilotinib effects on these biomarkers.26 Methodologically, Simuni et al performed one-time collection of CSF and serum, after 2 hours, that led to underestimation of drug availability.26 In 2019, Pagan et al staggered CSF collection at 1-4 hours, 30 minutes after blood draw, to obtain population-based pharmacokinetics.20,28 Monoamine oxidase B (MAO-B) inhibitors were allowed (>30%) in the study by Simuni et al,26 thus altering the level of dopamine metabolism, whereas the studies by Pagan et al excluded MAO-B inhibitors.20 Simuni et al26 did not control for levodopa in medically optimized patients28 and did not detect changes in dopamine metabolites. In contrast, data from animal studies,23 human phase 1 studies,24 phase 2 studies in PD, and phase 2 studies in levodopa-naïve patients with AD consistently demonstrated dose-dependent increases in dopamine after nilotinib treatment.20,29 This discrepancy could also be due to concurrent measurement of 17 metabolites that reduce the optimization of specific dopamine metabolites in serum and CSF by Simuni et al,28 whereas Pagan et al20,28 and Turner et al29 measured only 2 dopamine metabolites in plasma and CSF. It is imperative to mention that the placebo and nilotinib groups did not decline using UPDRS at 6 months and 12 months, indicating that all patients were medically optimized. However, treatment of 27 months with nilotinib suggested that 300 mg is beneficial for motor, cognitive, and behavioral symptoms in medically optimized patients.27 These long-term effects are consistent with disease modification showing evidence of increased autophagy flux in animal models and CSF microRNA levels in PD and LBD patients.23,30 In addition to nilotinib, which will be investigated in phase 3 studies (KeifeRx), 2 c-Abl inhibitors are currently in phase 2 clinical trials, including K0706 in early PD (Sun Pharmaceuticals) and LBD (at Georgetown University) and bosutinib (Bosulif; Pfizer) in LBD (also at Georgetown University).

Immune and Gene Therapies and Other Strategies

TABLE. Select Therapies in Phase 3 Trials in Parkinson Disease31

TABLE. Select Therapies in Phase 3 Trials in Parkinson Disease31

Several other strategies are in the pipeline for PD (TABLE).31 Deferiprone (Ferriprox; Chiesi) is FDA approved to treat some thalassemia syndromes as an iron chelation agent and is currently in phase 2/3 assessment for PD (at University Hospital, in Lille, France).32 The effects of glucagon-like peptide analogue (exenatide) on mitochondrial function (at Karolinska Institut, in Solna, Sweden) and a muscarinic and sigma-1 receptor agonist (ANAVEX 2-73; Avanex Life Science) are being investigated in phase 2 studies.

Targeting αS with the monoclonal antibody (mAb) BIIB054 (Biogen) did not meet its primary objectives. The mAb PRX002/Ro7046015 (Prothena/ Roche) and the glucocerebrosidase inhibitor GZ402671/SAR02671 (Sanofi), which prevents αS accumulation, are currently in phase 2. A number of phase 1 studies using anti-αS mAbs, like MEDI1341 (AstraZeneca/Takeda), ABBV-0805 (AbbVie), LU AF82422 (Lundbeck), PD01 AFFITOPE (AFFiRiS), and NPT200-11/UCB0599 (Neuropore/UCB) are being studied to prevent αS aggregation. mAbs may have significant hurdles entering the cytoplasm and ligating αS en route to degradation. This strategy has yet to prove its success in human clinical trials.

Several gene therapies are also being explored in PD, including phase 2 studies using adeno-associated virus (AAVs) to increase GBA in the subthalamic nucleus (GAD-AAV; MeiraGTx), single implant into the putamen to increase aromatic L-amino acid decarboxylase (AADC) levels (AADC-AAV2/VY/AADC02; Voyager/ Neurocrine), and bilateral putamen injection of AADC using a lentivirus (ProSavin, OXB-101; Oxford BioMedica/Sio Gene Therapies). Another anti-GBA agent that employs AAV is PR001 (Prevail Therapeutics). These gene therapies are still in early development, and while scientifically plausible, they face many challenges regarding stability of viral vectors; gene-of-interest expression, distribution, and spread of AAV/lentivirus; long-term gene expression and reversibility; adverse effects; and single vs multiple injections or surgeries.

Many agents show promise in having disease-modifying effects in PD, but in order to prove their viability, a specific target engagement and related biomarkers must be studied with the correctly termed clinical outcomes. Each agent may require unique criteria.

REFERENCES
1. Ravina BM, Fagan SC, Hart RG, et al. Neuroprotective agents for clinical trials in Parkinson’s disease: a systematic assessment. Neurology. 2003;60(8):1234-1240. doi:10.1212/01.wnl.0000058760.13152.1a
2. Olanow CW, Wunderle KB, Kieburtz K. Milestones in movement disorders clinical trials: advances and landmark studies. Mov Disord. 2011;26(6):1003-1014. doi:10.1002/mds.23727
3. Writing Group for the NINDS Exploratory Trials in Parkinson Disease (NET-PD) Investigators; Kieburtz K, Tilley BC, Elm JJ, et al. Effect of creatine monohydrate on clinical progression in patients with Parkinson disease: a randomized clinical trial. JAMA. 2015;313(6):584-593. doi:10.1001/jama.2015.120
4. Parkinson Study Group STEADY-PD III Investigators. Isradipine versus placebo in early Parkinson disease: a randomized trial. Ann Intern Med. 2020;172(9):591-598. doi:10.7326/M19-2534
5. Clinical trials in the spotlight: study of urate elevation in Parkinson’s disease, phase 3. National Institute of Neurological Disorders and Stroke. Updated March 27, 2019. Accessed March 11, 2021. https://www.ninds.nih.gov/Disorders/Clinical-Trials/Study-Urate-Elevation-Parkinsons-Disease-Phase-3-SURE-PD3
6. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839-840. doi:10.1038/42166
7. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. Alpha-synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with Lewy bodies. Proc Natl Acad Sci. U S A. 1998;95(11):6469-6473. doi:10.1073/pnas.95.11.6469
8. Goedert M. Alpha-synuclein and neurodegenerative diseases. Nat Rev Neurosci. 2001;2(7):492-501. doi:10.1038/35081564
9. Jack CR Jr, Bennett DA, Blennow K, et al. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology. 2016;87(5):539-547. doi:10.1212/WNL.0000000000002923
10. Regnault A, Boroojerdi B, Meunier J, Bani M, Morel T, Cano S. Does the MDS-UPDRS provide the precision to assess progression in early Parkinson’s disease? learnings from the Parkinson’s progression marker initiative cohort. J Neurol. 2019;266(8):1927-1936. doi:10.1007/s00415-019-09348-3
11. Espay AJ, Schwarzschild MA, Tanner CM, et al. Biomarker-driven phenotyping in Parkinson’s disease: a translational missing link in disease-modifying clinical trials. Mov Disord. 2017;32(3):319-324. doi:10.1002/mds.26913
12. Korecka JA, Thomas R, Christensen DP, et al. Mitochondrial clearance and maturation of autophagosomes are compromised in LRRK2 G2019S familial Parkinson’s disease patient fibroblasts. Hum Mol Genet. 2019;28(19):3232-3243. doi:10.1093/hmg/ddz126
13. Do J, McKinney C, Sharma P, Sidransky E. Glucocerebrosidase and its relevance to Parkinson disease. Mol Neurodegener. 2019;14(1):36. doi:10.1186/s13024-019-0336-2
14. Nussbaum RL, Polymeropoulos MH. Genetics of Parkinson’s disease. Hum Mol Genet. 1997;6(10):1687-1691. doi:10.1093/hmg/6.10.1687
15. El-Agnaf OM, Salem SA, Paleologou KE, et al. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006;20(3):419-425. doi:10.1096/fj.03-1449com
16. Mollenhauer B, El-Agnaf OMA, Marcus K, Trenkwalder C, Schlossmacher MG. Quantification of alpha-synuclein in cerebrospinal fluid as a biomarker candidate: review of the literature and considerations for future studies. Biomark Med. 2010;4(5):683-699. doi:10.2217/bmm.10.90
17. Park MJ, Cheon SM, Bae HR, Kim SH, Kim JW. Elevated levels of alpha-synuclein oligomer in the cerebrospinal fluid of drug-naive patients with Parkinson’s disease. J Clin Neurol. 2011;7(4):215-222. doi:10.3988/jcn.2011.7.4.215
18. Uversky VN, Li J, Fink AL. Evidence for a partially folded intermediate in alpha-synuclein fibril formation. J Biol Chem. 2001;276(14):10737-10744. doi:10.1074/jbc.M010907200
19. Wegrzynowicz M, Bar-On D, Calo L, et al. Depopulation of dense alpha-synuclein aggregates is associated with rescue of dopamine neuron dysfunction and death in a new Parkinson’s disease model. Acta Neuropathol. 2019;138(4):575-595. doi:10.1007/s00401-019-02023-x
20. Pagan FL, Hebron ML, Wilmarth B, et al. Nilotinib effects on safety, tolerability, and potential biomarkers in Parkinson disease: a phase 2 randomized clinical trial. JAMA Neurol. 2020;77(3):309-317. doi:10.1001/jamaneurol.2019.4200
21. Eisenhofer G, Kopin IJ, Goldstein DS. Catecholamine metabolism: a contemporary view with implications for physiology and medicine. Pharmacol Rev. 2004;56(3):331-349. doi:10.1124/pr.56.3.1
22. Kopin IJ. Catecholamine metabolism: basic aspects and clinical significance. Pharmacol Rev. 1985;37(4):333-364.
23. Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha-synuclein in Parkinson’s disease models. Hum Mol Genet. 2013;22(16):3315-3328. doi:10.1093/hmg/ddt192
24. Pagan F, Hebron M, Valadez EH, et al. Nilotinib effects in Parkinson’s disease and dementia with Lewy bodies. J Parkinsons Dis. 2016;6(3):503-517. doi:10.3233/JPD-160867
25. Manley PW, Drueckes P, Fendrich G, et al. Extended kinase profile and properties of the protein kinase inhibitor nilotinib. Biochim Biophys Acta. 2010;1804(3):445-453. doi:10.1016/j.bbapap.2009.11.008
26. Simuni T, Fiske B, Merchant K, et al; Parkinson Study Group NILO-PD Investigators and Collaborators. Efficacy of nilotinib in patients with moderately advanced Parkinson disease: a randomized clinical trial. JAMA Neurol. Published online December 14, 2020. doi:10.1001/jamaneurol.2020.4725
27. Pagan FL, Wilmarth B, Torres-Yaghi Y, et al. Long-term safety and clinical effects of nilotinib in Parkinson’s disease. Mov Disord. Published online November 20, 2020. doi:10.1002/mds.28389
28. Pagan FL, Hebron ML, Wilmarth B, et al. Pharmacokinetics and pharmacodynamics of a single dose nilotinib in individuals with Parkinson’s disease. Pharmacol Res Perspect. 2019;7(2):e00470. doi:10.1002/prp2.470
29. Turner RS, Hebron ML, Lawler A, et al. Nilotinib effects on safety, tolerability, and biomarkers in Alzheimer’s disease. Ann Neurol. 2020;88(1):183-194. doi:10.1002/ana.25775
30. Fowler AJ, Hebron M, Balaraman K, et al. Discoidin domain receptor 1 is a therapeutic target for neurodegenerative diseases. Hum Mol Genet. 2020;29(17):2882-2898. doi:10.1093/hmg/ddaa177
31. McFarthing K, Buff S, Rafaloff G, Dominey T, Wyse RK, Stott SRW. Parkinson’s disease drug therapies in the clinical trial pipeline: 2020. J Parkinsons Dis. 2020;10(3):757-774. doi:10.3233/JPD-202128
32. Rohani M, Razmeh S, Ali Shahidi G, Alizadeh E, Orooji M. A pilot trial of deferiprone in pantothenate kinase-associated neurodegeneration patients. Neurol Int. 2017;9(4):7279. doi:10.4081/ni.2017.7279
Related Videos
Jessica Ailani, MD
Frederic Schaper, MD, PhD
Jaime Imitol, MD
Jason M. Davies, MD, PhD
Carolyn Bernstein, MD
Prashanth Rajarajan, MD, PhD
Mandy Alhajj, DO, James Dolbow, DO & Neel Fotedar, MD
Riley Bove, MD
© 2024 MJH Life Sciences

All rights reserved.