422
Views
0
CrossRef citations to date
0
Altmetric
Review

Pharmacotherapy for Alzheimer’s disease: what’s new on the horizon?

ORCID Icon, , , ORCID Icon, ORCID Icon & ORCID Icon
Pages 1305-1323 | Received 11 Mar 2022, Accepted 30 Jun 2022, Published online: 11 Jul 2022

References

  • Alzheimer’s Association Facts and Figures. Alzheimer’s association; 2021.
  • Khoury R, Grossberg GT. Deciphering Alzheimer’s disease: predicting new therapeutic strategies via improved understanding of biology and pathogenesis. Expert Opin Ther Targets. 2020 Sep;24(9):859–868.
  • Jack CR Jr., Bennett DA, Blennow K, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535–562.
  • Khoury R, Ghossoub E, Diagnostic biomarkers of Alzheimer’s disease: a state-of-the-art review. Biomarkers in Neuropsychiatry. 2019 Dec 01;1:100005.
  • Nikolac Perkovic M, Pivac N. Genetic markers of Alzheimer’s disease. Adv Exp Med Biol. 2019;1192:27–52.
  • Khoury R, Patel K, Gold J, et al. Recent progress in the pharmacotherapy of Alzheimer’s disease. Drugs Aging. 2017 Nov;34(11):811–820.
  • Cummings J, Fox N. Defining disease modifying therapy for Alzheimer’s disease. J Prev Alzheimer’s Dis. 2017;4(2):109–115.
  • Cummings J, Aisen P, Apostolova LG, et al., Aducanumab: appropriate use recommendations. J Prev Alzheimer’s Dis. 8(4): 398–410. 2021.
  • Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004 Aug 01;3(8):673–683.
  • Bartus RT, Dean RL 3rd, Beer B, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science (New York, NY). 1982 Jul 30; 217(4558)408–414.
  • Yang. Inhibition of Cholinesterase by Octohydroaminoacri- dine Succinate [Master’s Thesis]. Changchun, Jilin, China: Jilin University; 2007.
  • Xiao S, Wang T, Ma X, et al. Efficacy and safety of a novel acetylcholinesterase inhibitor octohydroaminoacridine in mild-to-moderate Alzheimer’s disease: a Phase II multicenter randomised controlled trial. Age Ageing. 2017 Sep 1;46(5):767–773.
  • NCT03283059. Octohydroaminoacridine succinate tablet for mild-to-moderate Alzheimer’s disease. Available from: https://clinicaltrials.gov/ct2/show/NCT03283059?cond=NCT03283059&draw=2&rank=1
  • Wang X, Sun G, Feng T, et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019 Oct 01;29(10):787–803.
  • Kong LN, Geng MY, Mu L, et al. [Effects of acidic oligose on differentially expressed genes in the mice model of Alzheimer’s disease by microarray]. Yao xue xue bao = Acta pharmaceutica Sinica. 2005Dec; 40: 1105–1109
  • Wang T, Kuang W, Chen W, et al. A phase II randomized trial of sodium oligomannate in Alzheimer’s dementia. Alzheimer’s Res Ther. 2020 Sep 14;12(1):110.
  • Syed YY. Sodium Oligomannate: first approval. Drugs. 2020 Mar;80(4):441–444.
  • Xiao S, Chan P, Wang T, et al. A 36-week multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase 3 clinical trial of sodium oligomannate for mild-to-moderate Alzheimer’s dementia. Alzheimer’s Res Ther. 2021 Mar 17;13(1):62.
  • NCT05114499. Efficacy and safety of donepezil and sodium oligomannate in patients with mild to moderate Alzheimer’s disease. Available from: https://clinicaltrials.gov/ct2/show/NCT05114499?term=NCT05114499&draw=2&rank=1
  • NCT04520412. A study of sodium oligomannate (GV-971) in participants with mild to moderate Alzheimer’s disease (GREEN MEMORY). Available from: https://clinicaltrials.gov/ct2/show/NCT04520412?term=NCT04520412&draw=2&rank=1
  • NCT04229927. BPDO-1603 intervention trial in patients with moderate-to-severe Alzheimer’s disease. Available from: https://clinicaltrials.gov/ct2/show/NCT04229927?term=NCT04229927&draw=2&rank=1
  • Cruz MP. Guanfacine extended-release tablets (Intuniv), a nonstimulant selective alpha(2A)-adrenergic receptor agonist for attention-deficit/hyperactivity disorder. Pharm Ther. 2010;35(8):448–451.
  • Alamo C, López-Muñoz F, Sánchez-García J. Mechanism of action of guanfacine: a postsynaptic differential approach to the treatment of attention deficit hyperactivity disorder (ADHD). Actas espanolas de psiquiatria. 2016May;44(3):107–112.
  • Avery RA, Franowicz JS, Studholme C, et al. The alpha-2A-adrenoceptor agonist, guanfacine, increases regional cerebral blood flow in dorsolateral prefrontal cortex of monkeys performing a spatial working memory task. Neuropsychopharmacol. 2000 Sep;23(3):240–249.
  • Arnsten AF, Steere JC, Hunt RD. The contribution of alpha 2-noradrenergic mechanisms of prefrontal cortical cognitive functionPotential significance for attention-deficit hyperactivity disorder. Arch Gen Psychiatry. 1996 May;53(5):448–455.
  • Iwanami A, Saito K, Fujiwara M, et al. Safety and efficacy of guanfacine extended-release in adults with attention-deficit/hyperactivity disorder: an open-label, long-term, phase 3 extension study. BMC Psychiatry. 2020 Oct 02;20(1):485.
  • Barcelos NM, Van Ness PH, Wagner AF, et al. Guanfacine treatment for prefrontal cognitive dysfunction in older participants: a randomized clinical trial. Neurobiol Aging. 2018 Oct;70:117–124.
  • Kalaria RN, Andorn AC. Adrenergic receptors in aging and Alzheimer’s disease: decreased alpha 2-receptors demonstrated by [3H]p-aminoclonidine binding in prefrontal cortex. Neurobiol Aging. 1991 Mar-Apr;12(2):131–136.
  • NCT03116126. Noradrenergic add-on therapy with guanfacine (NorAD). Available from: https://clinicaltrials.gov/ct2/show/NCT03116126?term=NCT03116126&draw=2&rank=1
  • Sykiotis GP, Bohmann D. Stress-activated cap’n’collar transcription factors in aging and human disease. Sci Signal. 2010 Mar 9;3(112):re3.
  • Dehghan E, Goodarzi M, Saremi B, et al. Hydralazine targets cAMP-dependent protein kinase leading to sirtuin1/5 activation and lifespan extension in C. elegans. Nat Commun. 2019 Oct 28;10(1):4905.
  • Scarpa S, Cavallaro RA, D’Anselmi F, et al. Gene silencing through methylation: an epigenetic intervention on Alzheimer disease. J Alzheimers dis. 2006 Aug;9(4):407–414.
  • NCT04842552. Effect of Hydralazine on Alzheimer’s Disease (EHSAN).
  • Tomova T, Doncheva N, Mihaylova A, et al. An experimental study on phytochemical composition and memory enhancing effect of Ginkgo biloba seed extract. Folia Med (Plovdiv). 2021 Apr 30;63(2):203–212.
  • Yang X, Zheng T, Hong H, et al. Neuroprotective effects of Ginkgo biloba extract and Ginkgolide B against oxygen–glucose deprivation/reoxygenation and glucose injury in a new in vitro multicellular network model. Front Med. 2018 June 01;12(3):307–318.
  • Liu H, Ye M, Guo H. An updated review of randomized clinical trials testing the improvement of cognitive function of ginkgo biloba extract in healthy people and Alzheimer’s patients. Front Pharmacol. 2020;10:1688.
  • Zheng Y, Xie Y, Qi M, et al. Ginkgo biloba extract is comparable with donepezil in improving functional recovery in Alzheimer’s disease: results from a multilevel characterized study based on clinical features and resting-state functional magnetic resonance imaging. Front Pharmacol. 2021;12:721216.
  • Nowak A, Kojder K, Zielonka-Brzezicka J, et al. The use of Ginkgo Biloba L. as a Neuroprotective Agent in the Alzheimer’s Disease. Front Pharmacol. 2021;12. 775034.
  • Mitchell DC, Knight CA, Hockenberry J, et al. Beverage caffeine intakes in the U.S. Food Chem Toxicol. 2014 Jan;63:136–142.
  • Londzin P, Zamora M, Kąkol B, et al. Potential of caffeine in Alzheimer’s disease-a review of experimental studies. Nutrients. 2021;13(2):537.
  • NCT04570085. Effect of CAFfeine on cognition in Alzheimer’s disease (CAFCA). Available from: https://clinicaltrials.gov/ct2/show/NCT04570085?term=NCT04570085&draw=2&rank=1
  • Salat D, Helfgott J, Helmer K, et al. Neuroimaging methods in the randomized pivotal study of renew NCP‐5 for the treatment of mild cognitive impairment (MCI) due to Alzheimer’s disease or mild dementia of the Alzheimer’s type: neuroimaging/evaluating treatments. Alzheimers Dement. 2020 December 01;16(S4). DOI:https://doi.org/10.1002/alz.046189.
  • NCT03721705. Renew NCP-5 for the treatment of mild cognitive impairment due to Alzheimer’s Disease (AD) or mild dementia of the Alzheimer’s type. Available from: https://clinicaltrials.gov/ct2/show/NCT03721705
  • Ricciarelli R, Fedele E. The amyloid cascade hypothesis in Alzheimer’s disease: it’s time to change our mind. Curr Neuropharmacol. 2017;15(6):926–935.
  • Davies P, Koppel J. Mechanism-based treatments for Alzheimer’s disease. Dialogues Clin Neurosci. 2009;11(2):159–169.
  • O’Nuallain B, Freir DB, Nicoll AJ, et al. Amyloid beta-protein dimers rapidly form stable synaptotoxic protofibrils. J Neurosci. 2010;30(43):14411–14419.
  • Schmidt AM, Sahagan B, Nelson RB, et al. The role of RAGE in amyloid-beta peptide-mediated pathology in Alzheimer’s disease. Curr Opin Invest Drugs. 2009 Jul;10(7):672–680.
  • Tan MS, Tan L, Jiang T, et al. Amyloid-β induces NLRP1-dependent neuronal pyroptosis in models of Alzheimer’s disease. Cell Death Dis. 2014;5(8):e1382–e1382.
  • Tucker S, Möller C, Tegerstedt K, et al. The murine version of BAN2401 (mAb158) selectively reduces amyloid-β protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. J Alzheimers dis. 2015;43(2):575–588.
  • Logovinsky V, Satlin A, Lai R, et al. Safety and tolerability of BAN2401–a clinical study in Alzheimer’s disease with a protofibril selective Aβ antibody. Alzheimer’s Res Ther. 2016 Apr 6;8(1):14.
  • Swanson CJ, Zhang Y, Dhadda S, et al. A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer’s disease with lecanemab, an anti-Aβ protofibril antibody. Alzheimer’s Res Ther. 2021 Apr 17;13(1):80.
  • Salloway S, Chalkias S, Barkhof F, et al. Amyloid-related imaging abnormalities in 2 phase 3 studies evaluating aducanumab in patients with early Alzheimer disease. JAMA Neurol. 2022;79(1):13–21.
  • NCT03887455. A study to confirm safety and efficacy of lecanemab in participants with early alzheimer’s disease (clarity AD). Available from: https://clinicaltrials.gov/ct2/show/NCT03887455
  • NCT04468659. AHEAD 3-45 study: a study to evaluate efficacy and safety of treatment with lecanemab in participants with preclinical Alzheimer’s disease and elevated amyloid and also in participants with early preclinical Alzheimer’s disease and intermediate amyloid. Available from: https://clinicaltrials.gov/ct2/show/NCT04468659?term=NCT04468659&draw=2&rank=1
  • Bohrmann B, Baumann K, Benz J, et al. Gantenerumab: a novel human anti-Aβ antibody demonstrates sustained cerebral amyloid-β binding and elicits cell-mediated removal of human amyloid-β. J Alzheimers dis. 2012;28(1):49–69.
  • Ostrowitzki S, Lasser RA, Dorflinger E, et al. A phase III randomized trial of gantenerumab in prodromal Alzheimer’s disease. Alzheimer’s Res Ther. 2017 Dec 8;9(1):95.
  • Klein G, Delmar P, Voyle N, et al. Gantenerumab reduces amyloid-β plaques in patients with prodromal to moderate Alzheimer’s disease: a PET substudy interim analysis. Alzheimer’s Res Ther. 2019 Dec 12;11(1):101.
  • Klein G, Delmar P, Kerchner GA, et al. Thirty-six-month amyloid positron emission tomography results show continued reduction in amyloid burden with subcutaneous gantenerumab. J Prev Alzheimer’s Dis. 2021;8(1):3–6.
  • Salloway S, Farlow M, McDade E, et al. A trial of gantenerumab or solanezumab in dominantly inherited Alzheimer’s disease. Nat Med. 2021 Jul;27(7):1187–1196.
  • Taylor NP. UPDATE: roche dismisses early filing chatter for its Alzheimer’s drug as speculation, remains focused on phase 3 trial; 2021. Available from: https://www.fiercebiotech.com/biotech/roche-to-seek-early-fda-approval-for-alzheimer-s-drug-following-lilly-through-door-blown
  • Buntz B. Roche’s amyloid-targeting gantenerumab wins breakthrough therapy designation for Azheimer’s. Available from: https://www.drugdiscoverytrends.com/roches-amyloid-targeting-gantenerumab-wins-breakthrough-therapy-designation-for-azheimers
  • NCT03444870. Efficacy and safety study of gantenerumab in participants with early alzheimer’s disease (AD). Available from: https://clinicaltrials.gov/ct2/show/NCT03444870
  • NCT03443973. Safety and efficacy study of gantenerumab in participants with early Alzheimer’s Disease (AD). Available from: https://clinicaltrials.gov/ct2/show/NCT03443973
  • Tian Hui Kwan A, Arfaie S, Therriault J, et al., Lessons learnt from the second generation of anti-amyloid monoclonal antibodies clinical trials. Dement Geriatr Cogn Disord. 49(4): 334–348. 2020.
  • Siemers ER, Friedrich S, Dean RA, et al. Safety and changes in plasma and cerebrospinal fluid amyloid beta after a single administration of an amyloid beta monoclonal antibody in subjects with Alzheimer disease. Clin Neuropharmacol. 2010 Mar-Apr;33(2):67–73.
  • Farlow M, Arnold SE, van Dyck CH, et al. Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease. Alzheimers Dement. 2012 Jul;8(4):261–271.
  • Doody RS, Thomas RG, Farlow M, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370(4):311–321.
  • Honig LS, Vellas B, Woodward M, et al. Trial of solanezumab for mild dementia due to Alzheimer’s disease. N Engl J Med. 2018 Jan 25;378(4):321–330.
  • NCT02760602. A study of Solanezumab (LY2062430) in participants with prodromal Alzheimer’s disease (ExpeditionPRO). Available from: https://www.clinicaltrials.gov/ct2/show/NCT02760602
  • NCT02008357. Clinical trial of solanezumab for older individuals who may be at risk for memory loss (A4). Available from: https://clinicaltrials.gov/ct2/show/NCT02008357
  • Lowe SL, Willis BA, Hawdon A, et al. Donanemab (LY3002813) dose-escalation study in Alzheimer’s disease. Alzheimers Dementia. 2021;7(1):e12112.
  • Mintun MA, Lo AC, Duggan Evans C, et al. Donanemab in early Alzheimer’s disease. N Engl J Med. 2021;384(18):1691–1704.
  • NCT04437511. A study of donanemab (LY3002813) in participants with early Alzheimer’s disease (TRAILBLAZER-ALZ 2). Available from: https://www.clinicaltrials.gov/ct2/show/NCT04437511
  • Adolfsson O, Pihlgren M, Toni N, et al. An effector-reduced anti-β-amyloid (Aβ) antibody with unique aβ binding properties promotes neuroprotection and glial engulfment of Aβ. J Neurosci. 2012;32(28):9677–9689.
  • Guthrie H, Honig LS, Lin H, et al. Safety, tolerability, and pharmacokinetics of crenezumab in patients with mild-to-moderate Alzheimer’s disease treated with escalating doses for up to 133 weeks. J Alzheimers dis. 2020;76(3):967–979.
  • Cummings JL, Cohen S, van Dyck CH, et al. ABBY: a phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology. 2018 May 22;90(21):e1889–e1897.
  • Salloway S, Honigberg LA, Cho W, et al. Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer’s disease (BLAZE). Alzheimer’s Res Ther. 2018 Sep 19;10(1):96.
  • Hoffman M. Crenezumab phase 3 CREAD trials in Alzheimer disease discontinued by Roche; 2019.
  • Rios-Romenets S, Lopera F, Sink KM, et al. Baseline demographic, clinical, and cognitive characteristics of the Alzheimer’s prevention initiative (API) autosomal-dominant Alzheimer’s disease Colombia trial. Alzheimers Dement. 2020 Jul;16(7):1023–1030.
  • Dolton MJ, Chesterman A, Moein A, et al. Safety, tolerability, and pharmacokinetics of high-volume subcutaneous crenezumab, with and without recombinant human hyaluronidase in healthy volunteers. Clin Pharmacol Ther. 2021 Nov;110(5):1337–1348.
  • Yang L, Liu Y, Wang Y, et al. Azeliragon ameliorates Alzheimer’s disease via the janus tyrosine kinase and signal transducer and activator of transcription signaling pathway. Clinics (Sao Paulo). 2021;76:e2348–e2348.
  • Galasko D, Bell J, Mancuso JY, et al. Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease. Neurology. 2014 Apr 29;82(17):1536–1542.
  • NCT02080364. Evaluation of the efficacy and safety of azeliragon (TTP488) in patients with mild alzheimer’s disease (STEADFAST). Available from: https://www.clinicaltrials.gov/ct2/show/NCT02080364
  • Chris Min K, Dockendorf MF, Palcza J, et al. Pharmacokinetics and Pharmacodynamics of the BACE1 Inhibitor verubecestat (MK-8931) in healthy Japanese adults: a randomized, placebo-controlled study. Clin Pharmacol Ther. 2019 May;105(5):1234–1243.
  • Forman M, Palcza J, Tseng J, et al. Safety, tolerability, and pharmacokinetics of the β-site amyloid precursor protein-cleaving enzyme 1 inhibitor verubecestat (mk-8931) in healthy elderly male and female subjects. Clin Transl Sci. 2019 Sep;12(5):545–555.
  • Egan MF, Kost J, Tariot PN, et al. Randomized trial of verubecestat for mild-to-moderate Alzheimer’s disease. N Engl J Med. 2018;378(18):1691–1703.
  • Egan MF, Kost J, Voss T, et al. Randomized trial of verubecestat for prodromal Alzheimer’s disease. N Engl J Med. 2019 Apr 11;380(15):1408–1420.
  • Cárdenas-Aguayo Mdel C, Gómez-Virgilio L, DeRosa S, et al. The role of tau oligomers in the onset of Alzheimer’s disease neuropathology. ACS Chem Neurosci. 2014 Dec 17;5(12):1178–1191.
  • Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–259.
  • Kametani F, Hasegawa M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front Neurosci. 2018;12:25.
  • Congdon EE, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease. Nat Rev Neurol. 2018 Jul;14(7):399–415.
  • Panza F, Solfrizzi V, Seripa D, et al. Tau-centric targets and drugs in clinical development for the treatment of Alzheimer’s disease. Biomed Res Int. 2016;2016:3245935.
  • Wilcock GK, Gauthier S, Frisoni GB, et al. Potential of low dose leuco-methylthioninium bis(hydromethanesulphonate) (LMTM) monotherapy for treatment of mild Alzheimer’s disease: cohort analysis as modified primary outcome in a phase iii clinical trial. J Alzheimers dis. 2018;61(1):435–457.
  • Hashweh NN, Bartochowski Z, Khoury R, et al. An evaluation of hydromethylthionine as a treatment option for Alzheimer’s disease. Expert Opin Pharmacother. 2020 April 12;21(6):619–627.
  • NCT03446001. Safety and efficacy of trx0237 in subjects with Alzheimer’s disease followed by open-label treatment. Available from: https://clinicaltrials.gov/ct2/show/NCT03446001
  • Rosenzweig-Lipson S, Barton R, Gallagher M, et al. HOPE4MCI trial: first trial targeting reduction of hippocampal overactivity to treat mild cognitive impairment due to Alzheimer’s disease with AGB101. Alzheimers Dement. 2020;16(S9):e045331.
  • Sendrowski K, Sobaniec W, Stasiak-Barmuta A, et al. Study of the protective effects of nootropic agents against neuronal damage induced by amyloid-beta (fragment 25-35) in cultured hippocampal neurons. Pharmacol Rep. 2015 Apr;67(2):326–331.
  • Sugata S, Hanaya R, Kumafuji K, et al. Neuroprotective effect of levetiracetam on hippocampal sclerosis-like change in spontaneously epileptic rats. Brain Res Bull. 2011 Aug 10;86(1–2):36–41.
  • Amatniek JC, Hauser WA, DelCastillo-Castaneda C, et al. Incidence and predictors of seizures in patients with Alzheimer’s disease. Epilepsia. 2006 May;47(5):867–872.
  • Vossel K, Ranasinghe KG, Beagle AJ, et al. Effect of levetiracetam on cognition in patients with Alzheimer disease with and without epileptiform activity: a randomized clinical trial. JAMA Neurol. 2021 Nov 1;78(11):1345–1354.
  • NCT03486938. Study of AGB101 in mild cognitive impairment due to Alzheimer’s disease (HOPE4MCI). Available from: https://clinicaltrials.gov/ct2/show/NCT03486938?term=NCT03486938&draw=2&rank=1
  • Ruscher K, Wieloch T. The involvement of the sigma-1 receptor in neurodegeneration and neurorestoration. J Pharmacol Sci. 2015 Jan;127(1):30–35.
  • Lahmy V, Long R, Morin D, et al. Mitochondrial protection by the mixed muscarinic/σ1 ligand ANAVEX2-73, a tetrahydrofuran derivative, in Aβ25-35 peptide-injected mice, a nontransgenic Alzheimer’s disease model. Front Cell Neurosci. 2014;8:463.
  • Voges, O , Weigmann, I , Bitterlich, N , Missling, C , and Schindler, C. Abstracts of Poster Presentations: CNS Summit 2014: November 13-16, 2014 Boca Raton, Florida. Innov Clin Neurosci. 2015 May-Jun;12(SupplB):1–20.
  • Hampel H, Williams C, Etcheto A, et al. A precision medicine framework using artificial intelligence for the identification and confirmation of genomic biomarkers of response to an Alzheimer’s disease therapy: analysis of the blarcamesine (ANAVEX2-73) phase 2a clinical study. Alzheimers Dement. 2020;6(1):e12013–e12013. (N Y)
  • NCT03790709. ANAVEX2-73 for treatment of early Alzheimer’s disease.
  • Kanagasingam S, Chukkapalli SS, Welbury R, et al. Porphyromonas gingivalis is a strong risk factor for Alzheimer’s disease. J Alzheimers Dis Rep. 2020;4(1):501–511.
  • Dominy SS, Lynch C, Ermini F, et al. Porphyromonas gingivalis in Alzheimer’s disease brains: evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv. 2019 Jan;5(1):eaau3333.
  • Cicinelli C. Cortexyme reports GAIN trial data demonstrated relationship between reduction of P. gingivalis Infection and Slowing of Alzheimer’s Disease Progression. Available from: https://www.cortexyme.com/cortexyme-reports-gain-trial-data-demonstrated-relationship-between-reduction-of-p-gingivalis-infection-and-slowing-of-alzheimers-disease-progression/
  • Cortexyme. cortexyme announces clinical hold on atuzaginstat’s investigational new drug application. Available from: https://www.cortexyme.com/cortexyme-announces-clinical-hold-on-atuzaginstats-investigational-new-drug-application
  • Kinney JW, Bemiller SM, Murtishaw AS, et al. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 2018;4(1):575–590. (N Y)
  • CL R, CN A, MF M. NM101 phase III study of NE3107 in Alzheimer’s disease: rationale, design and therapeutic modulation of neuroinflammation and insulin resistance. Neurodegener Dis Manag. 2021 Aug;11(4):289–298.
  • NCT04669028. A phase 3 study of NE3107 in Alzheimer’s disease. Available from: https://clinicaltrials.gov/ct2/show/NCT04669028?term=NE3107&cond=alzheimer&draw=2&rank=1
  • Castellano CA, Nugent S, Paquet N, et al. Lower brain 18F-fluorodeoxyglucose uptake but normal 11C-acetoacetate metabolism in mild Alzheimer’s disease dementia. J Alzheimers dis. 2015;43(4):1343–1353.
  • Lin T-Y, Liu H-W, Hung T-M. The ketogenic effect of medium-chain triacylglycerides [mini review]. Front Nutr. 2021 Nov 18;8. DOI:https://doi.org/10.3389/fnut.2021.747284.
  • Walker JA, Nelleman L, Henderson ST, et al. Clinical development of tricaprilin, a ketogenic drug for Alzheimer’s disease. Alzheimers Dement. 2020;16(S9):e038787.
  • Henderson S, Walker J, Gold M, et al. Investigational new Alzheimer’S drug tricaprilin: possible effects of APOEε4 non-carrier selection and site effects on study outcomes in a phase 2/3 study in mild-to-moderate Alzheimer’S disease patients. Alzheimers Dement. 2018;07/01(14): P1521–P1522.
  • NCT04187547. A phase III multi regional clinical trial (mrct) of tricaprilin in mild to moderately severe probable alzheimer’s disease with optional open label extension. Available from: https://clinicaltrials.gov/ct2/show/NCT04187547
  • Cummings J. New approaches to symptomatic treatments for Alzheimer’s disease. Mol Neurodegener. 2021;16(1):2.
  • Rahimi J, Kovacs GG. Prevalence of mixed pathologies in the aging brain. Alzheimer’s Res Ther. 2014 Nov 21;6(9):82.
  • program AA. Available from: https://www.fda.gov/drugs/information-health-care-professionals-drugs/accelerated-approval-program.
  • Planche V, Villain N, Food US. Drug administration approval of aducanumab-is amyloid load a valid surrogate end point for Alzheimer disease clinical trials? JAMA Neurol. 2021Nov1;78(11):1307–1308.
  • Knopman DS, Jones DT, Greicius MD. Failure to demonstrate efficacy of aducanumab: an analysis of the EMERGE and ENGAGE trials as reported by Biogen, December 2019. Alzheimers Dement. 2021;17(4):696–701.
  • Liu KY, Howard R. Can we learn lessons from the FDA’s approval of aducanumab? Nat Rev Neurol. 2021 Nov 01;17(11):715–722.
  • Grossberg GT, Tong G, Burke AD, et al. Present algorithms and future treatments for Alzheimer’s disease. J Alzheimers dis. 2019;67(4):1157–1171.
  • Doggrell SA. Still grasping at straws: donanemab in Alzheimer’s disease. Expert Opin Investig Drugs. 2021 Aug;30(8):797–801.
  • Cummings JL, Tong G, Ballard C. Treatment combinations for Alzheimer’s Disease: current and future pharmacotherapy options. J Alzheimers dis. 2019;67(3):779–794.
  • Darbyshire J, Foulkes M, Peto R, et al. Immediate versus deferred zidovudine (AZT) in asymptomatic or mildly symptomatic HIV infected adults. Cochrane Database Syst Rev. 3;2000:Cd002039
  • Park A. The story behind the first AIDS drug; 2017. Available from: https://time.com/4705809/first-aids-drug-azt
  • Cummings J, Feldman HH, Scheltens P. The “rights” of precision drug development for Alzheimer’s disease. Alzheimer’s Res Ther. 2019 Aug 31;11(1):76.
  • Cole MA, Seabrook GR. On the horizon-the value and promise of the global pipeline of Alzheimer’s disease therapeutics. Alzheimers Dement. 2020;6(1):e12009. (N Y)
  • Sengupta U, Nilson AN, Kayed R. The role of amyloid-ß oligomers in toxicity, propagation, and immunotherapy. EBioMedicine. 2016;6:42–49.
  • Cummings J. The role of biomarkers in Alzheimer’s disease drug development. Adv Exp Med Biol. 2019;1118:29–61.
  • Selkoe DJ. Treatments for Alzheimer’s disease emerge. Science (New York, NY). 2021 Aug 6;373(6555):624–626.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.