Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-r6qrq Total loading time: 0 Render date: 2024-04-25T14:45:23.228Z Has data issue: false hasContentIssue false

2 - Drug Development for Alzheimer’s Disease: An Historical Perspective

from Section 1 - Advancing Alzheimer’s Disease Therapies in a Collaborative Science Ecosystem

Published online by Cambridge University Press:  03 March 2022

Jeffrey Cummings
Affiliation:
University of Nevada, Las Vegas
Jefferson Kinney
Affiliation:
University of Nevada, Las Vegas
Howard Fillit
Affiliation:
Alzheimer’s Drug Discovery Foundation
Get access

Summary

While Alzheimer’s disease (AD) remains one of the very few common chronic diseases of aging and old age without any effective treatments to slow or prevent the illness, an historical perspective can provide the context for why this is true. The same historical perspective demonstrates that this lag is primarily a frame shift in time, rather than due to excess difficulties in developing drugs for the disease. Indeed, the historical perspective suggests that today, we are “right on time” for an explosion in new drugs for the disease after 40 years of rapid progress in basic and clinical research. The first drugs (cholinergic agents) for AD were approved after only about 15-20 years of research in the field, including the time needed for basic discovery, clinical development and regulatory approval.  Today, after 40 years of AD research, many disease-modifying drugs are in clinical development. Considering that cancer, diabetes and hypertension research began more than 80-100 years ago, the rapid progress in AD research can actually be viewed as impressive progress.

Type
Chapter
Information
Alzheimer's Disease Drug Development
Research and Development Ecosystem
, pp. 25 - 33
Publisher: Cambridge University Press
Print publication year: 2022

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Newton, RD. The identity of Alzheimer’s disease and senile dementia and their relationship to senility. J Ment Sci 1948; 94: 225–49.CrossRefGoogle ScholarPubMed
Neumann, MA, Cohn, R. Incidence of Alzheimer’s disease in large mental hospital; relation to senile psychosis and psychosis with cerebral arteriosclerosis. AMA Arch Neurol Psychiatry 1953; 69: 615–36.Google Scholar
Roth, M, Tomlinson, BE, Blessed, G. Correlation between scores for dementia and counts of ‘senile plaques’ in cerebral grey matter of elderly subjects. Nature 1966; 209: 109–10.Google Scholar
Tomlinson, BE, Blessed, G, Roth, M. Observations on the brains of demented old people. J Neurol Sci 1970; 11: 205–42.CrossRefGoogle ScholarPubMed
Tomlinson, BE, Blessed, G, Roth, M. Observations on the brains of non-demented old people. J Neurol Sci 1968; 7: 331–56.Google Scholar
Terry, RD, Gonatas, NK, Weiss, M. The ultrastructure of the cerebral cortex in Alzheimer’s disease. Trans Am Neurol Assoc 1964; 89: 12.Google Scholar
Katzman, R. Editorial: The prevalence and malignancy of Alzheimer disease. A major killer. Arch Neurol 1976; 33: 217–18.CrossRefGoogle ScholarPubMed
Davies, P, Maloney, AJ. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976; 2: 1403.Google Scholar
Bowen, DM. Biochemistry of dementias. Proc R Soc Med 1977; 70: 351–3.Google Scholar
Whitehouse, PJ, Price, DL, Clark, AW, Coyle, JT, DeLong, MR. Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol 1981; 10: 122–6.CrossRefGoogle ScholarPubMed
Summers, WK, Majovski, LV, Marsh, GM, Tachiki, K, Kling, A. Oral tetrahydroaminoacridine in long-term treatment of senile dementia, Alzheimer type. N Engl J Med 1986; 315: 1241–5.Google Scholar
Atri, A. Current and future treatments in Alzheimer’s disease. Semin Neurol 2019; 39: 227–40.Google ScholarPubMed
Leber, P. Guidelines for the Clinical Evaluation of Antidementia Drugs. First draft. Technical Report. FDA Neuro-Pharm Group; 1990.Google Scholar
Folstein, MF, Folstein, SE, McHugh, PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975; 12: 189–98.Google Scholar
Guy, W. Clinical Global Impressions. ECDEU Assessment Manual for Psychopharmacology – Revised. Rockville, MD: US Department of Health, Education, and Welfare, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, National Institute of Mental Health, Psychopharmacology Research Branch, Division of Extramural Research Programs; 1976: 218–22.Google Scholar
Rosen, WG, Mohs, RC, Davis, KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64.Google Scholar
Farlow, M, Gracon, SI, Hershey, LA, et al. A controlled trial of tacrine in Alzheimer’s disease. The Tacrine Study Group. JAMA 1992; 268: 2523–9.Google Scholar
Davis, KL, Thal, LJ, Gamzu, ER, et al. A double-blind, placebo-controlled multicenter study of tacrine for Alzheimer’s disease. The Tacrine Collaborative Study Group. N Engl J Med 1992; 327: 1253–9.Google Scholar
Food and Drug Administration. Early Alzheimer’s Disease: Developing Drugs for Treatment. Guidance for Industry. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER); 2018.Google Scholar
Glenner, GG, Wong, CW. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 1984; 120: 885–90.Google Scholar
Glenner, GG. Alzheimer’s disease. The commonest form of amyloidosis. Arch Pathol Lab Med 1983; 107: 281–2.Google Scholar
Grundke-Iqbal, I, Iqbal, K, Quinlan, M, et al. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J Biol Chem 1986; 261: 6084–9.Google Scholar
Hardy, J, Selkoe, DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002; 297: 353–6.Google Scholar
Schenk, D, Barbour, R, Dunn, W, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999; 400: 173–7.Google Scholar
Gilman, S, Koller, M, Black, RS, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 2005; 64: 1553–62.Google Scholar
Holmes, C, Boche, D, Wilkinson, D, et al. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet 2008; 372: 216–23.CrossRefGoogle ScholarPubMed
Salloway, S, Sperling, R, Fox, NC, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med 2014; 370: 322–33.Google Scholar
Salloway, S, Sperling, R, Gilman, S, et al. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology 2009; 73: 2061–70.Google Scholar
Doody, RS, Farlow, M, Aisen, PS, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med 2014; 370: 311–21.Google Scholar
Bullain, S, Doody, R. What works and what does not work in Alzheimer’s disease? From interventions on risk factors to anti-amyloid trials. J Neurochem 2020; 155: https://doi.org/10.1111/jnc.15023.Google Scholar
van Dyck, CH. Anti-amyloid-β monoclonal antibodies for Alzheimer’s disease: pitfalls and promise. Psychiatry 2018; 83: 311–19.Google Scholar
Green, RC, Schneider, LS, Amato, DA, et al. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA 2009; 302: 2557–64.Google Scholar
Imbimbo, BP. Why did tarenflurbil fail in Alzheimer’s disease? J Alzheimers Dis 2009; 17: 757–60.Google Scholar
Doody, RS, Raman, R, Farlow, M, et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med 2013; 369: 341–50.Google Scholar
Egan, MF, Kost, J, Voss, T, et al. Randomized trial of verubecestat for prodromal Alzheimer’s disease. N Engl J Med 2019; 380: 1408–20.Google Scholar
Hara, Y, McKeehan, N, Fillit, HM. Translating the biology of aging into novel therapeutics for Alzheimer disease. Neurology 2019; 92: 8493.Google Scholar
Clark, CM, Pontecorvo, MJ, Beach, TG, et al. Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid-beta plaques: a prospective cohort study. Lancet Neurol 2012; 11: 669–78.Google Scholar
McNamee, LM, Walsh, MJ, Ledley, FD. Timelines of translational science: from technology initiation to FDA approval. PLoS One 2017; 12: e0177371.Google Scholar
Endo, A. A historical perspective on the discovery of statins. Proc Jpn Acad Ser B Phys Biol Sci 2010; 86: 484–93.Google Scholar
Qaseem, A, Snow, V, Cross, TJ Jr., et al. Current pharmacologic treatment of dementia: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med 2008; 148: 370–8.Google Scholar
Cummings, J, Ritter, A, Zhong, K. Clinical trials for disease-modifying therapies in Alzheimer’s disease: a primer, lessons learned, and a blueprint for the future. J Alzheimers Dis 2018; 64: S322.Google Scholar
Cummings, J. Lessons learned from Alzheimer disease: clinical trials with negative outcomes. Clin Transl Sci 2018; 11: 147–52.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement (N Y) 2020; 6: e12050.Google Scholar
Cummings, J, Blennow, K, Johnson, K, et al. Anti-tau trials for Alzheimer’s disease: a report from the EU/US/CTAD Task Force. J Prev Alzheimers Dis 2019; 6: 157–63.Google Scholar
Heneka, MT, Carson, MJ, El Khoury, J, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol 2015; 14: 388405.Google Scholar
Miguel-Alvarez, M, Santos-Lozano, A, Sanchis-Gomar, F, et al. Non-steroidal anti-inflammatory drugs as a treatment for Alzheimer’s disease: a systematic review and meta-analysis of treatment effect. Drugs Aging 2015; 32: 139–47.Google Scholar
Hampel, H, Caraci, F, Cuello, AC, et al. A path toward precision medicine for neuroinflammatory mechanisms in Alzheimer’s disease. Front Immunol 2020; 11: 456.Google Scholar
Wang, X, Sun, G, Geng, M. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids: shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res 2019; 29: 787803.Google Scholar
Xia, C, Dickerson, BC. Multimodal PET imaging of amyloid and tau pathology in Alzheimer disease and non-Alzheimer disease dementias. PET Clin 2017; 12: 351–9.Google Scholar
Risacher, SL, Saykin, AJ. Neuroimaging in aging and neurologic diseases. Handb Clin Neurol 2019; 167: 191227.Google Scholar
Sevigny, J, Suhy, J, Chiao, P, et al. Amyloid PET screening for enrichment of early-stage Alzheimer disease clinical trials: experience in a phase 1b clinical trial. Alzheimer Dis Assoc Disord 2016; 30: 17.Google Scholar
Sevigny, J, Chiao, P, Bussière, T, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature 2016; 537: 50–6.Google Scholar
Schindler, SE, Bollinger, JG, Ovod, V, et al. High-precision plasma beta-amyloid 42/40 predicts current and future brain amyloidosis. Neurology 2019; 93: e164759.Google Scholar
Barthelemy, NR, Bateman, RJ, Hirtz, C, et al. Cerebrospinal fluid phospho-tau T217 outperforms T181 as a biomarker for the differential diagnosis of Alzheimer’s disease and PET amyloid-positive patient identification. Alzheimers Res Ther 2020; 12: 26.Google Scholar
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: 535–62.Google Scholar
Dhana, K, Evans, DA, Rajan, KB, Bennett, DA, Morris, MC. Healthy lifestyle and the risk of Alzheimer dementia: findings from 2 longitudinal studies. Neurology 2020; 95: e374–83.Google Scholar
Kivipelto, M, Ngandu, T. Good for the heart and good for the brain? Lancet Neurol 2019; 18: 327–8.Google Scholar
Andrieu, S, Coley, N, Lovestone, S, Aisen, PS, Bruno Vellas, B. Prevention of sporadic Alzheimer’s disease: lessons learned from clinical trials and future directions. Lancet Neurol 2015; 14: 926–44.Google Scholar
Lopez Lopez, C, Tariot, PN, Caputo, A, et al. The Alzheimer’s Prevention Initiative Generation Program: study design of two randomized controlled trials for individuals at risk for clinical onset of Alzheimer’s disease. Alzheimers Dement (N Y) 2019; 5: 216–27.Google Scholar
Alzheimer’s Association. Changing the Trajectory of Alzheimer’s Disease: How a treatment by 2025 Saves Lives and Dollars. Chicago, IL: Alzheimer’s Association; 2015.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×