Insights into globalization: comparison of patient characteristics and disease progression among geographic regions in a multinational Alzheimer's disease clinical program

Jeffrey L Cummings, Alireza Atri, Clive Ballard, Neli Boneva, Lutz Frölich, José Luis Molinuevo, Lars Lau Raket, Pierre N Tariot, Jeffrey L Cummings, Alireza Atri, Clive Ballard, Neli Boneva, Lutz Frölich, José Luis Molinuevo, Lars Lau Raket, Pierre N Tariot

Abstract

Background: Globalization of clinical trials has important consequences for trial planning and interpretation. This study investigated heterogeneity in patient characteristics and outcomes among world regions in the global idalopirdine Phase 3 clinical program.

Methods: Data were pooled from three 24-week randomized controlled trials in patients aged ≥ 50 years with mild-to-moderate Alzheimer's disease (AD) (n = 2506). Patients received idalopirdine (10, 30, or 60 mg/day) or placebo, added to cholinesterase inhibitor treatment. Patients were categorized into the following regions: Eastern Europe/Turkey (n = 759), Western Europe/Israel (n = 709), USA/Canada (n = 444), South America/Mexico (n = 361), Asia (n = 134), and Australia/South Africa (n = 99). For each region, operational characteristics, baseline demographic and clinical characteristics, adverse events, and mean change from baseline to week 24 in clinical rating scale scores (placebo group only) were summarized using descriptive statistics.

Results: Completion rates were 0.86-0.90 in all regions. Heterogeneity among global regions was evident. Protocol deviations were twice as common in South America/Mexico as in USA/Canada (2.64 vs 1.35 per patient screened). Educational level ranged from 9.2 years in South America/Mexico to 13.4 years in USA/Canada. APOE ε4 carriage was 80.6% in Australia/South Africa, 63.1% in Western Europe/Israel, and < 60% in other regions. Screening Mini-Mental State Examination scores were higher in Eastern Europe/Turkey (18.0) and USA/Canada (17.5) than in other regions (16.9-17.1). Baseline AD Assessment Scale-Cognitive subscale (ADAS-Cog) scores ranged from 24.3 in USA/Canada to 27.2 in South America/Mexico. Baseline AD Cooperative Study-Activities of Daily Living, 23-item version (ADCS-ADL23) scores ranged from 58.5 in USA/Canada to 53.5 in Eastern Europe/Turkey. In the placebo group, adverse events were 1.6-1.7 times more common in Western Europe/Israel, USA/Canada, and Australia/South Africa than in Eastern Europe/Turkey. On the ADAS-Cog, Australia/South Africa and Western Europe/Israel showed the most worsening among patients receiving placebo (1.56 and 1.40 points, respectively), whereas South America/Mexico showed an improvement (-0.71 points). All regions worsened on the ADCS-ADL23, from -3.21 points in Western Europe/Israel to -0.59 points in Eastern Europe/Turkey.

Conclusions: Regional heterogeneity-in terms of study conduct, patient characteristics, and outcomes-exists, and should be accounted for, when planning and conducting multinational AD clinical trials.

Trial registration: ClinicalTrials.gov, NCT01955161 . Registered on 27 September 2013. ClinicalTrials.gov, NCT02006641 . Registered on 5 December 2013. ClinicalTrials.gov, NCT02006654 . Registered on 5 December 2013.

Keywords: Activities of daily living; Alzheimer’s disease; Clinical trial; Cognitive decline; Cognitive dysfunction; Dementia; Disease progression; Globalization; Humans; Patient selection.

Conflict of interest statement

Ethics approval and consent to participate

All studies were conducted in accordance with the International Conference on Harmonisation Good Clinical Practice Guideline and the Declaration of Helsinki. Local ethics committees approved all aspects of study design. Eligible patients or their legal representatives provided written informed consent prior to the start of the studies.

Consent for publication

Not applicable.

Competing interests

JLC reported serving as a consultant to AbbVie, Acadia, Actinogen, Adamas, Alzheon, Anavex, Astellas, Avanir, Avid, Axovant, Boehringer Ingelheim, Bracket, Eisai, GE Healthcare, Genentech, Intra-Cellular Therapies, Lilly, H. Lundbeck A/S, MedAvante, Merck, Neurocog, Novartis, Orion, Otsuka, Pfizer, Piramal, QR Pharma, reMYND, Resverlogix, Roche, Suven, Takeda, Toyama, and Transition; receiving research support from Avid and Teva; owning stock options in Prana, Neurokos, Adamas, MedAvante, and QR Pharma; and owning the copyright of the Neuropsychiatric Inventory and all its derivatives. JLC is supported by Keep Memory Alive and a COBRE grant from the NIGMS (P20GM109025).

AA reported receiving honoraria for consulting, participating in independent data safety monitoring boards, providing educational lectures, programs, and materials, or serving on advisory boards for Allergan, the Alzheimer’s Association, Axovant, Biogen, Grifols, Harvard Medical School Graduate Continuing Education, Lundbeck, Merck, Roche/Genentech, Sunovion, and Suven; receiving book royalties from Oxford University Press; and having institutional contracts or receiving investigational clinical trial-related funding from the American College of Radiology, AbbVie, Avid, Biogen, Lilly, Lundbeck, Merck, and vTV.

CB reported receiving research grants from Acadia and H. Lundbeck A/S; and receiving honoraria from Acadia, Bristol-Myers Squibb, Heptares, Lilly, H. Lundbeck A/S, Novartis, Orion, Otsuka, and Roche.

NB and LLR are full-time employees of H. Lundbeck A/S.

LF reported receiving honoraria for consulting, providing educational lectures, materials, and programs, or serving on advisory boards for Avid–Eli Lilly & Co, Avraham Pharmaceuticals, Axon Neuroscience, Boehringer Ingelheim, GE Healthcare, H. Lundbeck A/S, Merck Sharpe & Dohme, Novartis, Nutricia, Pfizer, Piramal Imaging, Schwabe Pharma, TAD Pharma, and Takeda.

JLM reported receiving honoraria for consulting, providing educational lectures, or serving on advisory boards for ABL, Axovant, Boehringer Ingelheim, Eli Lilly & Co, Fujirebio, GE Healthcare, H. Lundbeck A/S, Merck Sharpe & Dohme, Novartis, Pfizer, Piramal Imaging, Roche, and Roche Diagnostics.

PNT reported receiving consulting fees from Abbott Laboratories, AbbVie, AC Immune, Auspex, Boehringer Ingelheim, Clintara, CME Inc., Corium, GliaCure, INSYS, and T3D; receiving consulting fees and research support from AstraZeneca, Avanir, Cognoptix, Lilly, H. Lundbeck A/S, Merck and Company, and Takeda; receiving research support from Elan, Functional Neuromodulation, Genentech, Novartis, Roche, Targacept, the National Institute on Aging, and the Arizona Department of Health Services; owning stock options in Adamas; and being listed as a contributor to a patent owned by the University of Rochester.

No other disclosures were reported.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
Normalized observed decline from baseline to week 24 across regions in placebo group. 100%, maximal decline on the scale observed across regions; 0%, minimum decline/maximum improvement observed on the scale across regions. ADAS-Cog Alzheimer’s Disease Assessment Scale—Cognitive subscale, ADCS-ADL23 Alzheimer’s Disease Cooperative Study—Activities of Daily Living, 23-item version, ADCS-CGIC Alzheimer’s Disease Cooperative Study—Clinical Global Impression of Change, MMSE Mini–Mental State Examination, NPI Neuropsychiatric Inventory

References

    1. Alzheimer’s Association 2016 Alzheimer’s disease facts and figures. Alzheimers Dement. 2016;12:459–509. doi: 10.1016/j.jalz.2016.03.001.
    1. Atri A. Effective pharmacological management of Alzheimer’s disease. Am J Manag Care. 2011;17(Suppl 13):S346–S355.
    1. Parsons CG, Danysz W, Dekundy A, Pulte I. Memantine and cholinesterase inhibitors: complementary mechanisms in the treatment of Alzheimer’s disease. Neurotox Res. 2013;24:358–369. doi: 10.1007/s12640-013-9398-z.
    1. Atri A, Rountree SD, Lopez OL, Doody RS. Validity, significance, strengths, limitations, and evidentiary value of real-world clinical data for combination therapy in Alzheimer’s disease: comparison of efficacy and effectiveness studies. Neurodegener Dis. 2012;10:170–174. doi: 10.1159/000335156.
    1. Cummings JL, Morstorf T, Zhong K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther. 2014;6:37. doi: 10.1186/alzrt269.
    1. Cummings J, Reynders R, Zhong K. Globalization of Alzheimer’s disease clinical trials. Alzheimers Res Ther. 2011;3:24. doi: 10.1186/alzrt86.
    1. Glickman SW, McHutchison JG, Peterson ED, Cairns CB, Harrington RA, Califf RM, Schulman KA. Ethical and scientific implications of the globalization of clinical research. N Engl J Med. 2009;360:816–823. doi: 10.1056/NEJMsb0803929.
    1. International Conference on Harmonisation (ICH). Ethnic factors in the acceptability of foreign clinical data. London: Guideline E5(R1); 1998.
    1. Atri A, Frölich L, Ballard C, Tariot PN, Molinuevo JL, Boneva N, et al. Effect of idalopirdine as adjunct to cholinesterase inhibitors on change in cognition in patients with Alzheimer disease: three randomized clinical trials. JAMA. 2018;319:130–142. doi: 10.1001/jama.2017.20373.
    1. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34:939–944. doi: 10.1212/WNL.34.7.939.
    1. Folstein MF, Folstein SE, McHugh PR. “Mini–mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiat Res. 1975;12:189–198. doi: 10.1016/0022-3956(75)90026-6.
    1. Rosen WG, Mohs RC. Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry. 1984;141:1356–1364. doi: 10.1176/ajp.141.11.1356.
    1. Galasko D, Bennett D, Sano M, Ernesto C, Thomas R, Grundman M, Ferris S. The Alzheimer’s Disease Cooperative Study. An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. Alzheimer Dis Assoc Disord. 1997;11(Suppl 2):S33–S39. doi: 10.1097/00002093-199700112-00005.
    1. Robert P, Ferris S, Gauthier S, Ihl R, Winblad B, Tennigkeit F. Review of Alzheimer’s disease scales: is there a need for a new multi-domain scale for therapy evaluation in medical practice? Alzheimers Res Ther. 2010;2:24. doi: 10.1186/alzrt48.
    1. Schneider LS, Olin JT, Doody RS, Clark CM, Morris JC, Reisberg B, et al. The Alzheimer’s Disease Cooperative Study. Validity and reliability of the Alzheimer’s Disease Cooperative Study—Clinical Global Impression of Change. Alzheimer Dis Assoc Disord. 1997;11(Suppl 2):S22–S32. doi: 10.1097/00002093-199700112-00004.
    1. Guy W. ECDEU assessment manual for psychopharmacology, revised. Rockville: National Institute of Mental Health; 1976.
    1. Cummings JL. The Neuropsychiatric Inventory: assessing psychopathology in dementia patients. Neurology. 1997;48(Suppl 6):S10–S16. doi: 10.1212/WNL.48.5_Suppl_6.10S.
    1. Grill JD, Raman R, Ernstrom K, Aisen P, Dowsett SA, Chen YF, et al. Comparing recruitment, retention, and safety reporting among geographic regions in multinational Alzheimer’s disease clinical trials. Alzheimers Res Ther. 2015;7:39. doi: 10.1186/s13195-015-0122-5.
    1. Henley DB, Dowsett SA, Chen YF, Liu-Seifert H, Grill JD, Doody RS, et al. Alzheimer’s disease progression by geographical region in a clinical trial setting. Alzheimers Res Ther. 2015;7:43. doi: 10.1186/s13195-015-0127-0.
    1. Hanley MJ, Abernethy DR, Greenblatt DJ. Effect of obesity on the pharmacokinetics of drugs in humans. Clin Pharmacokinet. 2010;49:71–87. doi: 10.2165/11318100-000000000-00000.
    1. Meng X, D’Arcy C. Education and dementia in the context of the cognitive reserve hypothesis: a systematic review with meta-analyses and qualitative analyses. PLoS One. 2012;7:e38268. doi: 10.1371/journal.pone.0038268.
    1. Thomas RG, Albert M, Petersen RC, Aisen PS. Longitudinal decline in mild-to-moderate Alzheimer’s disease: analyses of placebo data from clinical trials. Alzheimers Dement. 2016;12:598–603. doi: 10.1016/j.jalz.2016.01.002.
    1. Stern Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol. 2012;11:1006–1012. doi: 10.1016/S1474-4422(12)70191-6.
    1. Honig LS, Vellas B, Woodward M, Boada M, Bullock R, Borrie M, et al. Trial of solanezumab for mild dementia due to Alzheimer’s disease. N Engl J Med. 2018;378:321–330. doi: 10.1056/NEJMoa1705971.
    1. Kuller LH, Shemanski L, Manolio T, Haan M, Fried L, Bryan N, et al. Relationship between ApoE, MRI findings, and cognitive function in the Cardiovascular Health Study. Stroke. 1998;29:388–398. doi: 10.1161/01.STR.29.2.388.
    1. Teruel BM, Rodríguez JJL, McKeigue P, Mesa TC, Fuentes E, Cepero AV, et al. Interactions between genetic admixture, ethnic identity, APOE genotype and dementia prevalence in an admixed Cuban sample; a cross-sectional population survey and nested case-control study. BMC Med Genet. 2011;12:43. doi: 10.1186/1471-2350-12-43.
    1. Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, et al. for the APOE and Alzheimer Disease Meta-Analysis Consortium. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. JAMA. 1997;278:1349–1356. doi: 10.1001/jama.1997.03550160069041.
    1. Sadigh-Eteghad S, Talebi M, Farhoudi M. Association of apolipoprotein E epsilon 4 allele with sporadic late onset Alzheimer’s disease. A meta-analysis. Neurosciences (Riyadh) 2012;17:321–326.
    1. Koller D, Hua T, Bynum JPW. Treatment patterns with anti-dementia drugs in the United States: Medicare cohort study. J Am Geriatr Soc. 2016;64:1540–1548. doi: 10.1111/jgs.14226.
    1. Rojas G, Demey I, Arizaga RL. Medicamentos utilizados para trastornos cognitivos. Análisis de un millón y medio de prescripciones en la Argentina. Medicina (B Aires) 2013;73:213–223.
    1. Senanarong V, Harnphadungkit K, Prayoonwiwat N, Poungvarin N, Sivasariyanonds N, Printarakul T, et al. A new measurement of activities of daily living for Thai elderly with dementia. Int Psychogeriatr. 2003;15:135–148. doi: 10.1017/S1041610203008822.
    1. Chiu HFK, Lam LCW. Relevance of outcome measures in different cultural groups—does one size fit all? Int Psychogeriatr. 2007;19:457–466. doi: 10.1017/S1041610207004838.
    1. Binetti G, Mega MS, Magni E, Padovani A, Rozzini L, Bianchetti A, et al. Behavioral disorders in Alzheimer disease: a transcultural perspective. Arch Neurol. 1998;55:539–544. doi: 10.1001/archneur.55.4.539.
    1. Chow TW, Liu CK, Fuh JL, Leung VPY, Tai CT, Chen LW, et al. Neuropsychiatric symptoms of Alzheimer’s disease differ in Chinese and American patients. Int J Geriatr Psychiatry. 2002;17:22–28. doi: 10.1002/gps.509.
    1. Truzzi A, Ulstein I, Valente L, Engelhardt E, Coutinho ESF, Laks J, Engedal K. Patterns of neuropsychiatric sub-syndromes in Brazilian and Norwegian patients with dementia. Int Psychogeriatr. 2013;25:228–235. doi: 10.1017/S1041610212001640.
    1. Salazar Ricardo, Dwivedi Alok Kumar, Royall Donald R. Cross-Ethnic Differences in the Severity of Neuropsychiatric Symptoms in Persons With Mild Cognitive Impairment and Alzheimer's Disease. The Journal of Neuropsychiatry and Clinical Neurosciences. 2017;29(1):13–21. doi: 10.1176/appi.neuropsych.15120423.

Source: PubMed

3
Abonnieren