Parkinson’s disease – a review of pathogenesis, recent advances in management, and challenges of care in sub-Saharan Africa

Authors

  • Akintomiwa I. Makanjuola Department of Neurology, University College Hospital, Ibadan, Nigeria
  • Funmilola T. Taiwo Department of Neurology, University College Hospital, Ibadan, Nigeria
  • Joseph O. Yaria Department of Neurology, University College Hospital, Ibadan, Nigeria
  • Rufus O. Akinyemi Department of Neurology, University College Hospital, Ibadan, Nigeria; and Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
  • Adesola Ogunniyi Department of Neurology, University College Hospital, Ibadan, Nigeria; and Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria

DOI:

https://doi.org/10.51496/jogm.v1.35

Keywords:

Parkinson's disease, Management, Dopamine replacement, Neurodegeneration, low-resource setting

Abstract

Parkinson’s disease (PD) remains a common neurodegenerative movement disorder with significant morbidity, which is expected to increase worldwide in the coming decades. Since its initial description, much has been elucidated about its etiology, pathogenesis, and the role of genetic and environmental risk factors. Effective treatments, including surgical therapies, have been discovered. Despite these strides, many questions remain unanswered; PD remains an active research area with ongoing efforts to discover newer treatment modalities and identify neuroprotective strategies. As with many neurological conditions, there is an unequal distribution of health resources, resulting in some management challenges in low resource settings, especially sub-Saharan Africa (SSA). In this communication, we provide an overview of PD etiopathogenesis, including genetics and management strategies, including some recent advances with respect to treatment options and disease modification approaches. Finally, we discuss some challenges of PD management in low-resource settings and highlight efforts to turn the tide.

References

Parkinson J. An essay on the shaking palsy. 1817. J Neuropsychiatry Clin Neurosci 2002 Mar; 14(2): 223–36. doi: 10.1176/jnp.14.2.223 DOI: https://doi.org/10.1176/jnp.14.2.223

Dorsey ER, Elbaz A, Nichols E, Abd-Allah F, Abdelalim A, Adsuar JC, et al. Global, regional, and national burden of Parkinson’s disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 2018 Nov; 17(11): 939–53.

Tekle-Haimanot R, Abebe M, Gebre-Mariam A, Forsgren L, Heijbel J, Holmgren G, et al. Community-based study of neurological disorders in rural central Ethiopia. Neuroepidemiology 1990; 9(5): 263. doi: 10.1159/000110783 DOI: https://doi.org/10.1159/000110783

Dotchin C, Msuya O, Kissima J, Massawe J, Mhina A, Moshy A, et al. The prevalence of Parkinson’s disease in rural Tanzania. Mov Disord 2008 Aug 15; 23(11): 1567–672. doi: 10.1002/mds.21898 DOI: https://doi.org/10.1002/mds.21898

Schoenberg B, Osuntokun B, Adeuja A, Bademosi O, Nottidge V, Anderson D, et al. Comparison of the prevalence of Parkinson’s disease in black populations in the rural United States and in rural Nigeria: door-to-door community studies. Neurology 1988; 38(4): 645–6. doi: 10.1212/WNL.38.4.645 DOI: https://doi.org/10.1212/WNL.38.4.645

Akinyemi RO. Epidemiology of Parkinsonism and Parkinson’s disease in Sub-Saharan Africa: Nigerian profile. J Neurosci Rural Pract 2012 Sep; 3(3): 234. doi: 10.4103/0976-3147.102586 DOI: https://doi.org/10.4103/0976-3147.102586

Marras C, Beck JC, Bower JH, Roberts E, Ritz B, Ross GW, et al. Prevalence of Parkinson’s disease across North America. NPJ Park Dis 2018 41. 2018 Jul 10; 4(1): 1–7. doi: 10.1038/s41531-018-0058-0 DOI: https://doi.org/10.1038/s41531-018-0058-0

Okubadejo NU, Bower JH, Rocca WA, Maraganore DM. Parkinson’s disease in Africa: a systematic review of epidemiologic and genetic studies. Mov Disord 2006 Dec 1; 21(12): 2150–6. DOI: https://doi.org/10.1002/mds.21153

Williams U, Bandmann O, Walker R. Parkinson’s disease in Sub-Saharan Africa: a review of epidemiology, genetics and access to care. J Mov Disord 2018 May 25; 11(2): 53–64. doi: 10.14802/jmd.17028 DOI: https://doi.org/10.14802/jmd.17028

Dotchin C, Walker R. The management of Parkinson’s disease in sub-Saharan Africa. Expert Rev Neurother 2012 Jun; 12(6): 661–6. doi: 10.1586/ern.12.52 DOI: https://doi.org/10.1586/ern.12.52

Ascherio A, Schwarzschild M. The epidemiology of Parkinson’s disease: risk factors and prevention. Lancet Neurol 2016 Nov 1; 15(12): 1257–72. doi: 10.1016/S1474-4422(16)30230-7 DOI: https://doi.org/10.1016/S1474-4422(16)30230-7

Simon DK, Tanner CM, Brundin P. Parkinson disease epidemiology, pathology, genetics and pathophysiology. Clin Geriatr Med 2020 Feb 1; 36(1): 1. doi: 10.1016/j.cger.2019.08.002 DOI: https://doi.org/10.1016/j.cger.2019.08.002

Gasser T, Hardy J, Mizuno Y. Milestones in PD genetics. Mov Disord 2011 May 1; 26(6): 1042–8. doi: 10.1002/mds.23637 DOI: https://doi.org/10.1002/mds.23637

Kouli A, Torsney KM, Kuan W-L. Parkinson’s disease: etiology, neuropathology, and pathogenesis. In: Stoker TB, Greenland JC, eds. Parkinson’s disease: pathogenesis and clinical aspects. Brisbane: Codon Publications; 2018, pp. 3–26. DOI: https://doi.org/10.15586/codonpublications.parkinsonsdisease.2018.ch1

Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-genome wide association study. Lancet Neurol 2019 Dec 1; 18(12): 1091.

Ferreira M, Massano J. An updated review of Parkinson’s disease genetics and clinicopathological correlations. Acta Neurol Scand 2017 Mar 1; 135(3): 273–84. doi: 10.1111/ane.12616 DOI: https://doi.org/10.1111/ane.12616

Bandres-Ciga S, Diez-Fairen M, Kim JJ, Singleton AB. Genetics of Parkinson’s disease: an introspection of its journey towards precision medicine. Neurobiol Dis 2020 Apr 1; 137: 104782. doi: 10.1016/j.nbd.2020.104782 DOI: https://doi.org/10.1016/j.nbd.2020.104782

O’Regan G, DeSouza R, Balestrino R, Schapira A. Glucocerebrosidase mutations in Parkinson disease. J Parkinsons Dis 2017; 7(3): 411–22. doi: 10.3233/JPD-171092 DOI: https://doi.org/10.3233/JPD-171092

Schapira AHV. Glucocerebrosidase and Parkinson disease: recent advances. Mol Cell Neurosci 2015 May 1; 66: 37. doi: 10.1016/j.mcn.2015.03.013 DOI: https://doi.org/10.1016/j.mcn.2015.03.013

Ince PG, Clark B, Holton J, Revesz T, Wharton SB. Diseases of movement and system degenerations. In: Love S, Louis DN, Ellison DW, eds. Greenfield’s neuropathology. 8th ed. London: Hodder Arnold; 2008, pp. 889–1030. DOI: https://doi.org/10.1201/b13319-14

Braak H, Tredici K Del, Rüb U, Vos R de, Steur EJ, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 2003; 24: 197–211. doi: 10.1016/S0197-4580(02)00065-9 DOI: https://doi.org/10.1016/S0197-4580(02)00065-9

Jankovic J, Tan EK. Parkinson’s disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry 2020 Aug 1; 91(8): 795–808. doi: 10.1136/jnnp-2019-322338 DOI: https://doi.org/10.1136/jnnp-2019-322338

Karpinar D, Balija M, Kügler S, Opazo F, Rezaei-Ghaleh N, Wender N, et al. Pre-fibrillar alpha-synuclein variants with impaired beta-structure increase neurotoxicity in Parkinson’s disease models. EMBO J 2009 Oct; 28(20): 3256–68. doi: 10.1038/emboj.2009.257 DOI: https://doi.org/10.1038/emboj.2009.257

Winner B, Jappelli R, Maji S, Desplats P, Boyer L, Aigner S, et al. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc Natl Acad Sci U S A 2011 Mar 8; 108(10): 4194–9. doi: 10.1073/pnas.1100976108 DOI: https://doi.org/10.1073/pnas.1100976108

Polymeropoulos M, Lavedan C, Leroy E, SIde S, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997 Jun 27; 276(5321): 2045–7. doi: 10.1126/science.276.5321.2045 DOI: https://doi.org/10.1126/science.276.5321.2045

Douglas PM, Dillin A. Protein homeostasis and aging in neurodegeneration. J Cell Biol 2010 Sep 6; 190(5): 719. doi: 10.1083/jcb.201005144 DOI: https://doi.org/10.1083/jcb.201005144

Kaushik S, Cuervo A. Proteostasis and aging. Nat Med 2015 Dec 8; 21(12): 1406–15. DOI: https://doi.org/10.1038/nm.4001

Xilouri M, Brekk O, Stefanis L. α-Synuclein and protein degradation systems: a reciprocal relationship. Mol Neurobiol 2013; 47(2): 537–51. doi: 10.1007/s12035-012-8341-2 DOI: https://doi.org/10.1007/s12035-012-8341-2

Heikkila R, Nicklas W, Vyas I, Duvoisin R. Dopaminergic toxicity of rotenone and the 1-methyl-4-phenylpyridinium ion after their stereotaxic administration to rats: implication for the mechanism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity. Neurosci Lett 1985 Dec 18; 62(3): 389–94. doi: 10.1016/0304-3940(85)90580-4 DOI: https://doi.org/10.1016/0304-3940(85)90580-4

Bradbury A, Costall B, Domeney A, Jenner P, Kelly M, Marsden C, et al. 1-methyl-4-phenylpyridine is neurotoxic to the nigrostriatal dopamine pathway. Nature 1986 Jan 2; 319(6048): 56–7. doi: 10.1038/319056a0 DOI: https://doi.org/10.1038/319056a0

Betarbet R, Sherer T, MacKenzie G, Garcia-Osuna M, Panov A, Greenamyre J. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 2000 Dec; 3(12): 1301–6. doi: 10.1038/81834 DOI: https://doi.org/10.1038/81834

Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, Korell M, et al. Rotenone, Paraquat, and Parkinson’s disease. Environ Health Perspect 2011 Jun; 119(6): 866. DOI: https://doi.org/10.1289/ehp.1002839

Schapira A, Cooper J, Dexter D, Clark J, Jenner P, Marsden C. Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem 1990 Mar 1; 54(3): 823–7. doi: 10.1111/j.1471-4159.1990.tb02325.x DOI: https://doi.org/10.1111/j.1471-4159.1990.tb02325.x

Ashrafi G, Schlehe JS, LaVoie MJ, Schwarz TL. Mitophagy of damaged mitochondria occurs locally in distal neuronal axons and requires PINK1 and Parkin. J Cell Biol 2014 Sep 1; 206(5): 655–70. doi: 10.1083/jcb.201401070 DOI: https://doi.org/10.1083/jcb.201401070

Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998 Apr 9; 392(6676): 605–8. doi: 10.1038/33416 DOI: https://doi.org/10.1038/33416

Valente EM, Abou-Sleiman PM, Caputo V, Muqit MMK, Harvey K, Gispert S, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science 2004 May 21; 304(5674): 1158–60. doi: 10.1126/science.1096284 DOI: https://doi.org/10.1126/science.1096284

Blesa J, Trigo-Damas I, Quiroga-Varela A, Jackson-Lewis VR. Oxidative stress and Parkinson’s disease. Front Neuroanat 2015 Jul 8; 9(91): 1–9. doi: 10.3389/fnana.2015.00091 DOI: https://doi.org/10.3389/fnana.2015.00091

Kim S, Kwon S, Kam T, Panicker N, Karuppagounder S, Lee S, et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 2019 Aug 21; 103(4): 627–641.e7. doi: 10.1016/j.neuron.​2019.05.035

Wood H. New models show gut-brain transmission of Parkinson disease pathology. Nat Rev Neurol 2019 Sep 1; 15(9): 491. doi: 10.1038/s41582-019-0241-x DOI: https://doi.org/10.1038/s41582-019-0241-x

Svensson E, Horváth-Puhó E, Thomsen RW, Djurhuus JC, Pedersen L, Borghammer P, et al. Vagotomy and subsequent risk of Parkinson’s disease. Ann Neurol 2015 Oct 1; 78(4): 522–9. doi: 10.1002/ana.24448 DOI: https://doi.org/10.1002/ana.24448

Breen D, Halliday G, Lang A. Gut-brain axis and the spread of α-synuclein pathology: vagal highway or dead end? Mov Disord 2019 Mar 1; 34(3): 307–16. doi: 10.1002/mds.27556 DOI: https://doi.org/10.1002/mds.27556

Gibb WR, Lees AJ. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry 1988; 51(6): 745. doi: 10.1136/jnnp.51.6.745 DOI: https://doi.org/10.1136/jnnp.51.6.745

Postuma R, Berg D, Stern M, Poewe W, Olanow C, Oertel W, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord 2015 Oct 1; 30(12): 1591–601. DOI: https://doi.org/10.1002/mds.26424

Hoehn M, Yahr M. Parkinsonism: onset, progression and mortality. Neurology 1967 May; 17(5): 427–42. doi: 10.1212/WNL.17.5.427 DOI: https://doi.org/10.1212/WNL.17.5.427

Goetz C, Poewe W, Rascol O, Sampaio C, Stebbins G, Counsell C, et al. Movement disorder society task force report on the Hoehn and Yahr staging scale: status and recommendations. Mov Disord 2004 Sep; 19(9): 1020–8. doi: 10.1002/mds.20213 DOI: https://doi.org/10.1002/mds.20213

Goetz CG, Tilley BC, Shaftman SR, Stebbins GT, Fahn S, Martinez-Martin P, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord 2008 Nov 15; 23(15): 2129–70. DOI: https://doi.org/10.1002/mds.22340

Postuma R, Aarsland D, Barone P, Burn D, Hawkes C, Oertel W, et al. Identifying prodromal Parkinson’s disease: pre-motor disorders in Parkinson’s disease. Mov Disord 2012 Apr 15; 27(5): 617–26. doi: 10.1002/mds.24996 DOI: https://doi.org/10.1002/mds.24996

DeLong MR, Wichmann T. Circuits and circuit disorders of the Basal Ganglia. Arch Neurol 2007 Jan 1; 64(1): 20–4. DOI: https://doi.org/10.1001/archneur.64.1.20

Brown P, Williams D. Basal ganglia local field potential activity: character and functional significance in the human. Clin Neurophysiol 2005 Nov; 116(11): 2510–9. doi: 10.1016/j.clinph.2005.05.009 DOI: https://doi.org/10.1016/j.clinph.2005.05.009

McIntyre C, Savasta M, Walter B, Vitek J. How does deep brain stimulation work? Present understanding and future questions. J Clin Neurophysiol 2004; 21(1): 40–50. doi: 10.1097/​00004691-200401000-00006

Brocks D. Anticholinergic drugs used in Parkinson’s disease: an overlooked class of drugs from a pharmacokinetic perspective. J Pharm Pharm Sci 2004; 2(2): 39–46.

Fahn S. The medical treatment of Parkinson disease from James Parkinson to George Cotzias. Mov Disord 2015 Jan 1; 30(1): 4–18. doi: 10.1002/mds.26102 DOI: https://doi.org/10.1002/mds.26102

Doshay L, Constable K, Zier A. Five year follow-up of treatment with trihexyphenidyl (artane); outcome in four hundred eleven cases of paralysis agitans. J Am Med Assoc 1954 Apr 17; 154(16): 1334–6. doi: 10.1001/jama.1954.02940500014005 DOI: https://doi.org/10.1001/jama.1954.02940500014005

Halliday A, Nathan P. Methyl phenidate in Parkinsonism. Br Med J 1961 Jun 10; 1(5240): 1652. doi: 10.1136/bmj.1.5240.1652 DOI: https://doi.org/10.1136/bmj.1.5240.1652

Schwab R, Tillmann W. Artane in the treatment of Parkinson’s disease; a report of its effectiveness alone and in combination with benadryl and parpanit. N Engl J Med 1949; 241(13): 483–5. doi: 10.1056/NEJM194909292411303 DOI: https://doi.org/10.1056/NEJM194909292411303

Hornykiewicz O. Basic research on dopamine in Parkinson’s disease and the discovery of the Nigrostriatal Dopamine Pathway: the view of an eyewitness. Neurodegener Dis 2008 Mar; 5(3–4): 114–7. doi: 10.1159/000113678 DOI: https://doi.org/10.1159/000113678

Fahn S. The 200-year journey of Parkinson disease: reflecting on the past and looking towards the future. Parkinsonism Relat Disord 2018 Jan 1; 46 Suppl 1: S1–5. DOI: https://doi.org/10.1016/j.parkreldis.2017.07.020

Bertler A, Rosengren E. Occurrence and distribution of catechol amines in brain. Acta Physiol Scand 1959 Dec 12; 47(6): 350–61.

Sano I, Gamo T, Kakimoto Y, Taniguchi K, Takesada M, Nishinuma K. Distribution of catechol compounds in human brain. Biochim Biophys Acta 1959 Apr; 32(C): 586–7. doi: 10.1016/0006-3002(59)90652-3 DOI: https://doi.org/10.1016/0006-3002(59)90652-3

Ehringer H, Hornykiewicz O. Distribution of noradrenaline and dopamine (3-hydroxytyramine) in the human brain and their behavior in diseases of the extrapyramidal system. Parkinsonism Relat Disord 1998 Aug; 4(2): 53–7. doi: 10.1016/S1353-8020(98)​00012-1

Hornykiewicz O. [The tropical localization and content of noradrenalin and dopamine (3-hydroxytyramine) in the substantia nigra of normal persons and patients with Parkinson’s disease]. Wien Klin Wochenschr 1963 May 3; 75(1): 309–12.

Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature 1957 Nov 30; 180(4596): 1200. doi: 10.1038/1801200a0 DOI: https://doi.org/10.1038/1801200a0

Carlsson A, Lindqvist M, Magnusson T, Waldeck B. On the presence of 3-hydroxytyramine in brain. Science 1958 Feb 28; 127(3296): 471. doi: 10.1126/science.127.3296.471-a DOI: https://doi.org/10.1126/science.127.3296.471

Anden N, Carlsson A, Dahlstroem A, Fuxe K, Hillarp N, Larsson K. Demonstration and mapping out of nigro-neostriatal dopamine neurons. Life Sci 1964 Jun 1; 3(6): 523–30. doi: 10.1016/0024-3205(64)90161-4 DOI: https://doi.org/10.1016/0024-3205(64)90161-4

Poirier L, Sourkes T. Influence of the substantia nigra on the catecholamine content of the striatum. Brain 1965 Mar; 88(1): 181–92. doi: 10.1093/brain/88.1.181 DOI: https://doi.org/10.1093/brain/88.1.181

Sourkes T, Poirier L. Influence of the substantia nigra on the concentration of 5-hydroxytryptamine and dopamine of the striatum. Nature 1965 Jul 10; 207(993): 202–3. DOI: https://doi.org/10.1038/207202a0

Cotzias GC, Van Woert MH, Schiffer LM. aromatic amino acids and modification of Parkinsonism. N Engl J Med 1967 Feb 16; 276(7): 374–9. doi: 10.1056/NEJM196702162760703 DOI: https://doi.org/10.1056/NEJM196702162760703

Cotzias GC, Papavasiliou PS, Gellene R. Modification of Parkinsonism – chronic treatment with L-Dopa. New Engl Med 1969 Jan 14; 280(7): 337–45. DOI: https://doi.org/10.1056/NEJM196902132800701

Yahr MD, Duvoisin RC, Schear MJ, Barrett RE, Hoehn MM. Treatment of Parkinsonism with levodopa. Arch Neurol 1969 Oct 1; 21(4): 343–54. doi: 10.1001/archneur.1969.0048016​0015001

Parkinson’s disease in adults. United Kingdom National Institute for Health and Care Excellence (NICE) guideline; 2017.

Armstrong M, Okun M. Diagnosis and treatment of Parkinson disease: a review. JAMA 2020; 323: 548–60. doi: 10.1001/jama.2019.22360 DOI: https://doi.org/10.1001/jama.2019.22360

Sawada H, Oeda T, Kuno S, Nomoto M, Yamamoto K, Yamamoto M, et al. Amantadine for dyskinesias in Parkinson’s disease: a randomized controlled trial. PLoS One 2010; 5(12): 1–7. doi: 10.1371/journal.pone.0015298 DOI: https://doi.org/10.1371/journal.pone.0015298

van Kessel S, Frye A, El-Gendy A, Castejon M, Keshavarzian A, van Dijk G, et al. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat Commun 2019 Dec 1; 10(1): 1–11. DOI: https://doi.org/10.1038/s41467-019-08294-y

Friedman JH. ‘Drug Holidays’ in the treatment of Parkinson’s disease: a brief review. Arch Intern Med 1985 May 1; 145(5): 913–5. doi: 10.1001/archinte.145.5.913 DOI: https://doi.org/10.1001/archinte.145.5.913

Koziorowski D, Friedman A. Levodopa ‘drug holiday’ with amantadine infusions as a treatment of complications in Parkinson’s disease. Mov Disord 2007 May 15; 22(7): 1033–6. DOI: https://doi.org/10.1002/mds.21448

Fahn S. Is levodopa toxic? Neurology 1996; 47(6 Suppl 3): 184S–195S. doi: 10.1212/WNL.47.6_Suppl_3.184S DOI: https://doi.org/10.1212/WNL.47.6_Suppl_3.184S

Verschuur CVM, Suwijn SR, Boel JA, Post B, Bloem BR, van Hilten JJ, et al. Randomized delayed-start trial of levodopa in Parkinson’s disease. N Engl J Med 2019 Jan 23; 380(4): 315–24. doi: 10.1056/NEJMoa1809983 DOI: https://doi.org/10.1056/NEJMoa1809983

Cilia R, Akpalu A, Sarfo FS, Cham M, Amboni M, Cereda E, et al. The modern pre-levodopa era of Parkinson’s disease: insights into motor complications from sub-Saharan Africa. Brain 2014 Oct 1; 137(10): 2731. DOI: https://doi.org/10.1093/brain/awu195

Rascol O, Brooks D, Korczyn A, De Deyn P, Clarke C, Lang A. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med 2000 May 18; 342(20): 1484–91. doi: 10.1056/NEJM200005183422004 DOI: https://doi.org/10.1056/NEJM200005183422004

Holloway R, Shoulson I, Fahn S, Kieburtz K, Lang A, Marek K, et al. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch Neurol 2004 Jul; 61(7): 1044–53. DOI: https://doi.org/10.1001/archneur.61.7.1044

Gray R, Ives N, Rick C, Patel S, Gray A, Jenkinson C, et al. Long-term effectiveness of dopamine agonists and monoamine oxidase B inhibitors compared with levodopa as initial treatment for Parkinson’s disease (PD MED): a large, open-label, pragmatic randomised trial. Lancet 2014 Sep 27; 384(9949): 1196–205. DOI: https://doi.org/10.1016/S0140-6736(14)60683-8

Fox SH, Lang AE. ‘Don’t delay, start today’: delaying levodopa does not delay motor complications. Brain 2014 Oct 1; 137(10): 2628–30. doi: 10.1093/brain/awu212 DOI: https://doi.org/10.1093/brain/awu212

Ogunniyi A. Treatment of parkinsonian syndromes in developing countries. Afr J Med Med Sci 1997 Aug; 26(1–2): 101–3.

Weaver FM, Follett K, Stern M, Hur K, Harris C, Marks WJ, et al. Bilateral deep brain stimulation vs best medical therapy for patients with advanced Parkinson disease: a randomized controlled trial. JAMA 2009 Jan 7; 301(1): 63. doi: 10.1001/jama.2008.929 DOI: https://doi.org/10.1001/jama.2008.929

Bond AE, Shah BB, Huss DS, Dallapiazza RF, Warren A, Harrison MB, et al. Safety and efficacy of focused ultrasound thalamotomy for patients with medication-refractory, tremor-dominant Parkinson disease: a randomized clinical trial. JAMA Neurol 2017 Dec 1; 74(12): 1412. doi: 10.1001/jamaneurol.2017.3098 DOI: https://doi.org/10.1001/jamaneurol.2017.3098

Martínez-Fernández R, Máñez-Miró J, Rodríguez-Rojas R, Del Álamo M, Shah B, Hernández-Fernández F, et al. Randomized trial of focused ultrasound subthalamotomy for Parkinson’s disease. N Engl J Med 2020 Dec 24; 383(26): 2501–13. doi: 10.1056/NEJMoa2016311 DOI: https://doi.org/10.1056/NEJMoa2016311

Seppi K, Chaudhuri KR, Coelho M, Fox SH, Katzenschlager R, Lloret SP, et al. Update on treatments for nonmotor symptoms of Parkinson’s disease – an evidence-based medicine review. Mov Disord 2019 Feb 1; 34(2): 180. doi: 10.1002/mds.27602 DOI: https://doi.org/10.1002/mds.27602

Cummings J, Isaacson S, Mills R, Williams H, Chi-Burris K, Corbett A, et al. Pimavanserin for patients with Parkinson’s disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet 2014 Feb 8; 383(9916): 533–40. DOI: https://doi.org/10.1016/S0140-6736(13)62106-6

Hacksell U, Burstein ES, McFarland K, Mills RG, Williams H. On the discovery and development of pimavanserIn: a novel drug candidate for Parkinson’s psychosis. Neurochem Res 2014 Mar 30; 39(10): 2008–17. doi: 10.1007/s11064-014-1293-3

van der Kolk N, de Vries N, Kessels R, Joosten H, Zwinderman A, Post B, et al. Effectiveness of home-based and remotely supervised aerobic exercise in Parkinson’s disease: a double-blind, randomised controlled trial. Lancet Neurol 2019 Nov 1; 18(11): 998–1008. doi: 10.1016/S1474-4422(19)30285-6 DOI: https://doi.org/10.1016/S1474-4422(19)30285-6

Bloem BR, Okun MS, Klein C. Parkinson’s disease. Lancet 2021 Jun 12; 397(10291): 2284–303. doi: 10.1016/S0140-6736(21)​00218-X

Lang A, Rodriguez R, Boyd J, Chouinard S, Zadikoff C, Espay A, et al. Integrated safety of levodopa-carbidopa intestinal gel from prospective clinical trials. Mov Disord 2016 Apr 1; 31(4): 538–46. doi: 10.1002/mds.26485 DOI: https://doi.org/10.1002/mds.26485

Liu XD, Bao Y, Liu G jian. Comparison between levodopa-carbidopa intestinal gel infusion and subthalamic nucleus deep-brain stimulation for advanced Parkinson’s disease: a systematic review and meta-analysis. Front Neurol 2019 Aug 27; 10(934): 1–10. doi: 10.3389/fneur.2019.00934 DOI: https://doi.org/10.3389/fneur.2019.00934

LeWitt P, Hauser R, Pahwa R, Isaacson S, Fernandez H, Lew M, et al. Safety and efficacy of CVT-301 (levodopa inhalation powder) on motor function during off periods in patients with Parkinson’s disease: a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Neurol 2019 Feb 1; 18(2): 145–54. doi: 10.1016/S1474-4422(18)30405-8 DOI: https://doi.org/10.1016/S1474-4422(18)30405-8

Rosebraugh M, Voight E, Moussa E, Jameel F, Lou X, Zhang G, et al. Foslevodopa/foscarbidopa: a new subcutaneous treatment for Parkinson’s disease. Ann Neurol 2021 Jul 1; 90(1): 52–61. doi: 10.1002/ana.26073 DOI: https://doi.org/10.1002/ana.26073

Torti M, Vacca L, Stocchi F. Istradefylline for the treatment of Parkinson’s disease: is it a promising strategy? Expert Opin Pharmacother 2018 Nov 2; 19(16): 1821–8. doi: 10.1080/14656566.2018.1524876

Lees A, Ferreira J, Rascol O, Poewe W, Rocha J, McCrory M, et al. Opicapone as adjunct to levodopa therapy in patients with parkinson disease and motor fluctuations: a randomized clinical trial. JAMA Neurol 2017 Feb 1; 74(2): 197–206. doi: 10.1001/jamaneurol.2016.4703 DOI: https://doi.org/10.1001/jamaneurol.2016.4703

Ferreira J, Lees A, Poewe W, Rascol O, Rocha J, Keller B, et al. Effectiveness of opicapone and switching from entacapone in fluctuating Parkinson disease. Neurology 2018 May 22; 90(21): E1849–57. DOI: https://doi.org/10.1212/WNL.0000000000005557

Greenwood J, Pham H, Rey J. Opicapone: a third generation COMT inhibitor. Clin Park Relat Disord 2021; 4: 1–5. doi: 10.1016/j.prdoa.2020.100083 DOI: https://doi.org/10.1016/j.prdoa.2020.100083

The Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. N Engl J Med 1993 Jan 21; 328(3): 176–83. doi: 10.1056/NEJM199301213280305 DOI: https://doi.org/10.1056/NEJM199301213280305

Olanow C, Rascol O, Hauser R, Feigin P, Jankovic J, Lang A, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009 Sep 24; 361(13): 1268–78. doi: 10.1056/NEJMoa0809335 DOI: https://doi.org/10.1056/NEJMoa0809335

Ward C. Does selegiline delay progression of Parkinson’s disease? A critical re-evaluation of the DATATOP study. J Neurol Neurosurg Psychiatry 1994; 57(2): 217–20. DOI: https://doi.org/10.1136/jnnp.57.2.217

Brys M, Fanning L, Hung S, Ellenbogen A, Penner N, Yang M, et al. Randomized phase I clinical trial of anti-α-synuclein antibody BIIB054. Mov Disord 2019 Aug 1; 34(8): 1154–63. doi: 10.1002/mds.27738 DOI: https://doi.org/10.1002/mds.27738

Pagano G, Taylor K, Cabrera J, Marchesi M, Zago W, Tripuraneni R, et al. PASADENA: a Phase 2 study to evaluate the safety and efficacy of prasinezumab in early Parkinson’s disease; Part 1 Week-52 results [Internet]. MDS Virtual Congress. 2020. Available from: https://www.mdsabstracts.org/abstract/pasadena-a-phase-2-study-to-evaluate-the-safety-and-efficacy-of-prasinezumab-in-early-parkinsons-disease-part-1-week-52-results/ [cited 31 August 2021].

Mullin S, Smith L, Lee K, D’Souza G, Woodgate P, Elflein J, et al. Ambroxol for the treatment of patients with Parkinson disease with and without glucocerebrosidase gene mutations: a nonrandomized, noncontrolled trial. JAMA Neurol 2020 Apr 1; 77(4): 427–34. doi: 10.1001/jamaneurol.2019.4611 DOI: https://doi.org/10.1001/jamaneurol.2019.4611

Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet 2017 Oct 7; 390(10103): 1664–75.

Simmering JE, Welsh MJ, Liu L, Narayanan NS, Pottegård A. Association of glycolysis-enhancing α-1 blockers with risk of developing Parkinson disease. JAMA Neurol 2021 Apr 1; 78(4): 407–13. doi: 10.1001/jamaneurol.2020.5157 DOI: https://doi.org/10.1001/jamaneurol.2020.5157

Barker RA, Barrett J, Mason SL, Björklund A. Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson’s disease. Lancet Neurol 2013 Jan 1; 12(1): 84–91. DOI: https://doi.org/10.1016/S1474-4422(12)70295-8

Freed CR, Greene PE, Breeze RE, Tsai W-Y, DuMouchel W, Kao R, et al. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Engl J Med 2001 Mar 8; 344(10): 710–9. doi: 10.1056/NEJM200103083441002 DOI: https://doi.org/10.1056/NEJM200103083441002

Olanow CW, Goetz CG, Kordower JH, Stoessl AJ, Sossi V, Brin MF, et al. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson’s disease. Ann Neurol 2003 Sep 1; 54(3): 403–14. DOI: https://doi.org/10.1002/ana.10720

Kordower JH, Goetz CG, Chu Y, Halliday GM, Nicholson DA, Musial TF, et al. Robust graft survival and normalized dopaminergic innervation do not obligate recovery in a Parkinson disease patient. Ann Neurol 2017 Jan 1; 81(1): 46. doi: 10.1002/ana.24820 DOI: https://doi.org/10.1002/ana.24820

Barker R. Designing stem-cell-based dopamine cell replacement trials for Parkinson’s disease. Nat Med 2019 Jul 1; 25(7): 1045–53. doi: 10.1038/s41591-019-0507-2 DOI: https://doi.org/10.1038/s41591-019-0507-2

Parmar M, Grealish S, Henchcliffe C. The future of stem cell therapies for Parkinson disease. Nat Rev Neurosci 2020 Jan 6; 21(2): 103–15. DOI: https://doi.org/10.1038/s41583-019-0257-7

Palfi S, Gurruchaga JM, Ralph GS, Lepetit H, Lavisse S, Buttery PC, et al. Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson’s disease: a dose escalation, open-label, phase 1/2 trial. Lancet 2014 Mar 29; 383(9923): 1138–46. DOI: https://doi.org/10.1016/S0140-6736(13)61939-X

Palfi S, Gurruchaga JM, Lepetit H, Howard K, Ralph GS, Mason S, et al. Long-term follow-up of a phase I/II study of ProSavin, a lentiviral vector gene therapy for Parkinson’s disease. Hum Gene Ther Clin Dev 2018 Sep 1; 29(3): 148–55. doi: 10.1089/humc.2018.081 DOI: https://doi.org/10.1089/humc.2018.081

LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty AW, Eskandar EN, et al. AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol 2011 Apr 1; 10(4): 309–19. DOI: https://doi.org/10.1016/S1474-4422(11)70039-4

Christine CW, Bankiewicz KS, Van Laar AD, Richardson RM, Ravina B, Kells AP, et al. Magnetic resonance imaging–guided phase 1 trial of putaminal AADC gene therapy for Parkinson’s disease. Ann Neurol 2019 May 1; 85(5): 704–14. doi: 10.1002/ana.25450 DOI: https://doi.org/10.1002/ana.25450

Nutt JG, Curtze C, Hiller A, Anderson S, Larson PS, Van Laar AD, et al. Aromatic L-Amino Acid Decarboxylase Gene Therapy Enhances Levodopa Response in Parkinson’s Disease. Mov Disord 2020 May 1; 35(5): 851–8. DOI: https://doi.org/10.1002/mds.27993

Whone A, Luz M, Boca M, Woolley M, Mooney L, Dharia S, et al. Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain 2019 Mar 1; 142(3): 512–25. doi: 10.1093/brain/awz023 DOI: https://doi.org/10.1093/brain/awz023

Axelsen TM, Woldbye DPD. Gene therapy for Parkinson’s disease: an update. J Parkinsons Dis 2018; 8(2): 195–215. doi: 10.3233/JPD-181331 DOI: https://doi.org/10.3233/JPD-181331

Marques T, van Rumund A, Oeckl P, Kuiperij H, Esselink R, Bloem B, et al. Serum NfL discriminates Parkinson disease from atypical parkinsonisms. Neurology 2019 Mar 26; 92(13): e1479–86. DOI: https://doi.org/10.1212/WNL.0000000000007179

Lin C, Li C, Yang K, Lin F, Wu C, Chieh J, et al. Blood NfL: a biomarker for disease severity and progression in Parkinson disease. Neurology 2019 Sep 10; 93(11): e1104–11. doi: 10.1212/WNL.0000000000008088 DOI: https://doi.org/10.1212/WNL.0000000000008088

Oosterveld LP, Verberk IMW, Majbour NK, El-Agnaf OM, Weinstein HC, Berendse HW, et al. CSF or serum neurofilament light added to α-Synuclein panel discriminates Parkinson’s from controls. Mov Disord 2020 Feb 1; 35(2): 288–95. doi: 10.1002/mds.27897 DOI: https://doi.org/10.1002/mds.27897

Dotchin CL, Msuya O, Walker RW. The challenge of Parkinson’s disease management in Africa. Age Ageing 2007 Mar 1; 36(2): 122–7. doi: 10.1093/ageing/afl172 DOI: https://doi.org/10.1093/ageing/afl172

Dekker MCJ, Coulibaly T, Bardien S, Ross OA, Carr J, Komolafe M. Parkinson’s disease research on the African Continent: obstacles and opportunities. Front Neurol 2020 Jun 19; 11(512): 1–9. doi: 10.3389/fneur.2020.00512 DOI: https://doi.org/10.3389/fneur.2020.00512

Mokaya J, Gray W, Carr J. Beliefs, knowledge and attitudes towards Parkinson’s disease among a Xhosa speaking black population in South Africa: a cross-sectional study. Parkinsonism Relat Disord 2017 Aug 1; 41: 51–7. DOI: https://doi.org/10.1016/j.parkreldis.2017.05.009

Mokaya J, Dotchin CL, Gray WK, Hooker J, Walker RW. The accessibility of Parkinson’s disease medication in Kenya: results of a national survey. Mov Disord Clin Pract 2016 Jul 1; 3(4): 376–81. doi: 10.1002/mdc3.12294 DOI: https://doi.org/10.1002/mdc3.12294

Okubadejo N, Ojo O, Wahab K, Abubakar S, Obiabo O, Salawu F, et al. A nationwide survey of Parkinson’s disease medicines availability and affordability in Nigeria. Mov Disord Clin Pract 2018 Nov 16; 6(1): 27–33. DOI: https://doi.org/10.1002/mdc3.12682

WHO. ATLAS country resources for neurological disorders. World Health Organization (WHO), Geneva; 2017.

Smith M, Francis P. Parkinson’s disease in Africa: a South African perspective. World Neurol 2020; 35(4): 11.

Mansour A, Fayed Z. DBS in Parkinson’s disease: experience from Egypt (Conference Abstract) [Internet]. MDS International Congress. 2019. Available from: https://www.mdsabstracts.org/abstract/dbs-in-parkinsons-disease-experience-from-egypt/ [cited 7 September 2021].

Rahmani M, Benabdeljlil M, Bellakhdar F, Faris MEA, Jiddane M, El Bayad K, et al. Deep brain stimulation in Moroccan patients with Parkinson’s disease: the experience of Neurology Department of Rabat. Front Neurol 2018 Jul 31; 9(532): 1–10. doi: 10.3389/fneur.2018.00532 DOI: https://doi.org/10.3389/fneur.2018.00532

Okubadejo N. African Section [Internet]. Available from: https://www.movementdisorders.org/MDS-Africa [cited 7 September 2021].

Ojo O, Abubakar S, Iwuozo E, Nwazor E, Ekenze O, Farombi T, et al. The Nigeria Parkinson disease registry: process, profile, and prospects of a collaborative project. Mov Disord 2020 Aug 1; 35(8): 1315–22. DOI: https://doi.org/10.1002/mds.28123

Blanckenberg J, Bardien S, Glanzmann B, Okubadejo NU, Carr JA. The prevalence and genetics of Parkinson’s disease in sub-Saharan Africans. J Neurol Sci 2013 Dec 15; 335(1–2): 22–5. doi: 10.1016/j.jns.2013.09.010 DOI: https://doi.org/10.1016/j.jns.2013.09.010

Rizig M, Okubadejo N, Salama M, Thomas O, Akpalu A, Gouider R. The International Parkinson Disease Genomics Consortium Africa. Lancet Neurol 2021 May 1; 20(5): 335. doi: 10.1016/S1474-4422(21)00100-9 DOI: https://doi.org/10.1016/S1474-4422(21)00100-9

Oluwole OG, Kuivaniemi H, Abrahams S, Haylett WL, Vorster AA, van Heerden CJ, et al. Targeted next-generation sequencing identifies novel variants in candidate genes for Parkinson’s disease in Black South African and Nigerian patients. BMC Med Genet 2020 Feb 4; 21(1): 1–11. doi: 10.1186/s12881-020-0953-1 DOI: https://doi.org/10.1186/s12881-020-0953-1

Fothergill-Misbah N, Maroo H, Cham M, Pezzoli G, Walker R, Cilia R. Could Mucuna pruriens be the answer to Parkinson’s disease management in sub-Saharan Africa and other low-income countries worldwide? Parkinsonism Relat Disord 2020 Apr 1; 73: 3–7. doi: 10.1016/j.parkreldis.2020.​03.002

Katzenschlager R, Evans A, Manson A, Patsalos PN, Ratnaraj N, Watt H, et al. Mucuna pruriens in Parkinson’s disease: a double blind clinical and pharmacological study. J Neurol Neurosurg Psychiatry 2004 Dec 1; 75(12): 1672–7. doi: 10.1136/jnnp.2003.028761

Cilia R, Laguna J, Cassani E, Cereda E, Pozzi N, Isaias I, et al. Mucuna pruriens in Parkinson disease: a double-blind, randomized, controlled, crossover study. Neurology 2017 Aug 1; 89(5): 432–8. doi: 10.1212/WNL.0000000000004175 DOI: https://doi.org/10.1212/WNL.0000000000004175

Cilia R, Laguna J, Cassani E, Cereda E, Raspini B, Barichella M, et al. Daily intake of Mucuna pruriens in advanced Parkinson’s disease: a 16-week, noninferiority, randomized, crossover, pilot study. Parkinsonism Relat Disord 2018 Apr 1; 49: 60–6. doi: 10.1016/j.parkreldis.2018.01.014 DOI: https://doi.org/10.1016/j.parkreldis.2018.01.014

Published

02-11-2021

How to Cite

Makanjuola, A. I. ., Taiwo, F. T. ., Yaria, J. O. ., Akinyemi, R. O. ., & Ogunniyi, A. . (2021). Parkinson’s disease – a review of pathogenesis, recent advances in management, and challenges of care in sub-Saharan Africa. Journal of Global Medicine, 1(2), e35. https://doi.org/10.51496/jogm.v1.35

Issue

Section

Review Articles

Categories