The Influence of Baseline Alzheimer's Disease Severity on Cognitive Decline and CSF Biomarkers in the NILVAD Trial

Laila Abdullah, Fiona Crawford, Magda Tsolaki, Anne Börjesson-Hanson, Marcel Olde Rikkert, Florence Pasquier, Anders Wallin, Sean Kennelly, Ghania Ait-Ghezala, Daniel Paris, Suzanne Hendrix, Kaj Blennow, Brian Lawlor, Michael Mullan, Laila Abdullah, Fiona Crawford, Magda Tsolaki, Anne Börjesson-Hanson, Marcel Olde Rikkert, Florence Pasquier, Anders Wallin, Sean Kennelly, Ghania Ait-Ghezala, Daniel Paris, Suzanne Hendrix, Kaj Blennow, Brian Lawlor, Michael Mullan

Abstract

We examined the effects of a dihydropyridine calcium channel blocker nilvadipine with anti-inflammatory properties on cognition and cerebrospinal fluid (CSF) biomarkers by baseline Alzheimer's disease (AD) severity. Exploratory analyses were performed on the dataset (n = 497) of a phase III randomized placebo-controlled trial to examine the response to nilvadipine in AD subjects stratified by baseline AD severity into very mild (MMSE ≥ 25), mild (MMSE 20-24) and moderate AD (MMSE < 20). The outcome measures included total and subscale scores of the Alzheimer's Disease Assessment Scale Cognitive 12 (ADAS-Cog 12), the Clinical Dementia Rating Scale sum of boxes (CDR-sb) and the AD composite score (ADCOMS). Cerebrospinal fluid biomarkers Aβ38, Aβ40, Aβ42, neurofilament light chain (NFL), neurogranin, YKL-40, total tau and P181 tau (ptau) were measured in a subset of samples (n = 55). Regression analyses were adjusted for confounders to specifically examine the influence of nilvadipine and baseline AD severity on cognitive outcomes over 78-weeks. Compared to their respective placebo-controls, nilvadipine-treated, very mild AD subjects showed less decline, whereas moderate AD subjects showed a greater cognitive decline on the ADAS-Cog 12 test and the ADCOMS. A lower decline was observed after nilvadipine treatment for a composite memory trait in very mild AD subjects and a composite language trait in mild AD subjects. Cerebrospinal fluid Aβ42/Aβ40 ratios were increased in mild AD and decreased in moderate AD patients treated with nilvadipine, compared to their respective controls. Among moderate AD subjects, levels of ptau, total tau, neurogranin and YKL-40 increased in subjects treated with nilvadipine compared to placebo. These studies suggest that baseline AD severity influenced the treatment outcome in the NILVAD trial and that future clinical trials of nilvadipine should be restricted to mild and very mild AD patients. Trial Registration: NCT02017340 Registered 20 December 2013, https://ichgcp.net/clinical-trials-registry/NCT02017340 EUDRACT Reference Number 2012-002764-27 Registered 04 February 2013, https://www.clinicaltrialsregister.eu/ctr-search/search?query=2012-002764-27.

Keywords: cerebrospinal fluid Aβ42/Aβ40 ratios; cognitive decline; exploratory analysis; mild Alzheimer's disease; nilvadipine.

Copyright © 2020 Abdullah, Crawford, Tsolaki, Börjesson-Hanson, Olde Rikkert, Pasquier, Wallin, Kennelly, Ait-Ghezala, Paris, Hendrix, Blennow, Lawlor and Mullan.

Figures

Figure 1
Figure 1
Further stratification of the mild AD group by increasing the increment of MMSE scores by 1 starting from ≥20 up to ≥25. Mean ± SE (for MMSE ≥ 20 n = 154 for nilvadipine and n = 157 for placebo; ≥21 n = 125 for nilvadipine and 136 for placebo; ≥22 n = 103 for nilvadipine and n = 117 for placebo; ≥23 n = 70 for nilvadipine and n = 99 for placebo; ≥ 24 n = 57 for nilvadipine and n = 68 for placebo; ≥25 n = 36 for nilvadipine and n = 44 for placebo). Mean change from baseline for the total ADAS-Cog 12 scores show the least decline among nilvadipine-treated subjects compared to placebo-treated subjects with MMSE score of ≥25.
Figure 2
Figure 2
Grouping of ADAS-cog and CDR-sb sub-scales to examine specific cognitive traits (A) Sub-scales of the ADAS-cog 12 test were grouped based on traits for memory, language and praxis to account for the topography of tissue loss in AD depending on the stage of disease. (B) Sub-scales of the ADAS-cog 12 and the CDR-sb were analyzed by PCA, which resulted in grouping of sub-scales into four factors that explained most of the variance in the dataset. Composite variables were then generated, which included sub-scales identified in each factor by PCA, and were named factors 1 through 4. Note, factor 1 also contains the orientation sub-scale from the ADAS-Cog in addition to the ones from the CDR-sb.
Figure 3
Figure 3
Data on ADCOMS and ADAS-Cog 12 test. Nilvadipine-treated very mild AD subjects show less cognitive decline compared to controls on the ADCOMS and the ADAS-Cog 12 tests. Mean ± SE [n = 82 for moderate AD (MMSE ≤ 19) on nilvadipine, n = 94 for moderate AD on placebo, n = 118 for mild AD (MMSE 20-24) on nilvadipine, n = 113 for mild AD on placebo, n = 36 for very mild AD (MMSE ≥ 35) on nilvadipine and n = 44] for very mild AD on placebo for the change in ADAS-Cog 12 scores. There was a significant effect for the interaction between treatment, time and baseline AD severity as assessed by MMSE scores after correcting for the confounding effects of APOE, gender and education, p < 0.05. (A) Stratifications show that very mild AD subjects treated with nilvadipine have lower scores on the ADCOMS and the ADAS-Cog 12 compared to placebo after 78 weeks. post-hoc analysis stratified by time show a significant treatment effect at 78 weeks for the ADCOMS. (B) Mild AD subjects treated with nilvadipine scored similarly to their placebo controls on both the ADCOMS and the ADAS-Cog 12 (C). However, moderate AD subjects treated with nilvadipine scored higher on both the ADCOMS and the ADAS-Cog 12 at 78 weeks compared to those on placebo. *p < 0.05.
Figure 4
Figure 4
Nilvadipine treatment effects on cognitive traits. Very mild AD subjects show less decline on the memory trait, whereas mild AD subjects show less decline on the language trait, after nilvadipine treatment compared to placebo. Mean ± SE (n = 82 for moderate AD on nilvadipine, n = 94 for moderate AD on placebo, n = 118 for mild AD on nilvadipine, n = 113 for mild AD on placebo, n = 36 for very mild AD on nilvadipine, n = 44 for very mild AD on placebo) for the change in memory, language and praxis traits of grouped ADAS-cog 12 sub-scales. (A) There was a significant effect for the interaction between treatment, time and baseline AD severity on the memory trait. post-hoc stratifications by time show that very mild AD subjects treated with nilvadipine had significantly less decline on the memory trait compared to their controls. (B) There was also a significant interaction between treatment, time and baseline AD severity for the language trait. post-hoc stratifications by time show that mild AD subjects treated with nilvadipine had less decline on the language trait compared to the placebo-treated mild AD subjects. (C) There was no effect seen for the praxis trait. *p < 0.05.
Figure 5
Figure 5
Cerebrospinal fluid Aβ biomarkers. Ratios of CSF Aβ42/Aβ40 increase in nilvadipine-treated mild AD but decrease in moderate AD patients compared to their respective placebo groups. (Mean ± SE n = 9 for moderate AD on nilvadipine, n = 12 for moderate AD on placebo, n = 14 for mild AD on nilvadipine, n = 20 for mild AD on placebo). (A) Ratios of Aβ42/Aβ40 were higher in mild AD treated with nilvadipine compared to those treated with placebo (p = 0.067). There was a significant decrease in Aβ42/Aβ40 in moderate AD treated with nilvadipine compared to placebo. Also in moderate AD subjects, levels of (B) Aβ38 and (C) Aβ40 were elevated and (D) Aβ42 levels were unchanged in moderate AD subjects treated with nilvadipine. Levels of Aβ42 were non-significantly higher in mild AD treated with nilvadipine. *p < 0.05.
Figure 6
Figure 6
Cerebrospinal fluid tau biomarkers. Total tau and P181Tau levels were increased in nilvadipine-treated moderate AD patients compared to their respective placebo groups. (Mean ± SE n = 9 for moderate AD on nilvadipine, n = 12 for moderate AD on placebo, n = 14 for mild AD on nilvadipine, n = 20 for mild AD on placebo). Levels of (A) total tau and (B) P181 tau were increased in moderate AD subjects treated with nilvadipine. In mild AD subjects, total tau or P181 tau did not differ between nilvadipine-treated and placebo control groups.*p < 0.05.

References

    1. Davies P. Neurochemical studies: an update on Alzheimer's disease. J Clin Psychiatry. (1988) 49(Suppl.):23–8.
    1. Tandon A, Rogaeva E, Mullan M, St George-Hyslop PH. Molecular genetics of Alzheimer's disease: the role of beta-amyloid and the presenilins. Curr Opin Neurol. (2000) 13:377–84. 10.1097/00019052-200008000-00003
    1. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, et al. . Alzheimer's disease. Lancet. (2016) 388:505–17. 10.1016/S0140-6736(15)01124-1
    1. Kehoe PG, Passmore PA. The renin-angiotensin system and antihypertensive drugs in Alzheimer's disease: current standing of the angiotensin hypothesis? J Alzheimers Dis. (2012) 30(Suppl. 2):S251–S268. 10.3233/JAD-2012-111376
    1. Kovacs GG, Milenkovic I, Wohrer A, Hoftberger R, Gelpi E, Haberler C, et al. . Non-alzheimer neurodegenerative pathologies and their combinations are more frequent than commonly believed in the elderly brain: a community-based autopsy series. Acta Neuropathol. (2013) 126:365–84. 10.1007/s00401-013-1157-y
    1. James BD, Wilson RS, Boyle PA, Trojanowski JQ, Bennett DA, Schneider JA. Tdp-43 stage, mixed pathologies, and clinical Alzheimer's-type dementia. Brain. (2016) 139:2983–93. 10.1093/brain/aww224
    1. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. . Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. N Engl J Med. (2014) 370:311–21. 10.1056/NEJMoa1312889
    1. Sevigny J, Suhy J, Chiao P, Chen T, Klein G, Purcell D, 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) 30:1–7. 10.1097/WAD.0000000000000144
    1. Soininen H, Solomon A, Visser PJ, Hendrix SB, Blennow K, Kivipelto M, et al. . (2017) 24-month intervention with a specific multinutrient in people with prodromal Alzheimer's disease (lipididiet): a randomised, double-blind, controlled trial. Lancet Neurol. 16:965–75. 10.1016/S1474-4422(17)30332-0
    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) 378:321–30. 10.1056/NEJMoa1705971
    1. Liu-Seifert H, Case MG, Andersen SW, Holdridge KC, Aisen PS, Kollack-Walker S, et al. . Delayed-start analyses in the phase 3 solanezumab expedition3 study in mild Alzheimer's disease. J Prev Alzheimers Dis. (2018) 5:8–14. 10.14283/jpad.2018.1
    1. Paris D, Quadros A, Humphrey J, Patel N, Crescentini R, Crawford F, et al. . Nilvadipine antagonizes both abeta vasoactivity in isolated arteries, and the reduced cerebral blood flow in appsw transgenic mice. Brain Res. (2004) 999:53–61. 10.1016/j.brainres.2003.11.061
    1. Paris D, Bachmeier C, Patel N, Quadros A, Volmar CH, Laporte V, et al. . Selective antihypertensive dihydropyridines lower abeta accumulation by targeting both the production and the clearance of abeta across the blood-brain barrier. Mol Med. (2011) 17:149–62. 10.2119/molmed.2010.00180
    1. Paris D, Ait-Ghezala G, Bachmeier C, Laco G, Beaulieu-Abdelahad D, Lin Y, et al. . The spleen tyrosine kinase (syk) regulates alzheimer amyloid-beta production and tau hyperphosphorylation. J Biol Chem. (2014) 289:33927–44. 10.1074/jbc.M114.608091
    1. Bachmeier C, Beaulieu-Abdelahad D, Mullan M, Paris D. Selective dihydropyiridine compounds facilitate the clearance of beta-amyloid across the blood-brain barrier. Eur J Pharmacol. (2011) 659:124–9. 10.1016/j.ejphar.2011.03.048
    1. Morin A, Mouzon B, Ferguson S, Paris D, Saltiel N, Lungmus C, et al. . Treatment with nilvadipine mitigates inflammatory pathology and improves spatial memory in aged htau mice after repetitive mild TBI. Front Aging Neurosci. (2018) 10:292. 10.3389/fnagi.2018.00292
    1. Hanyu H, Hirao K, Shimizu S, Sato T, Kiuchi A, Iwamoto T. Nilvadipine prevents cognitive decline of patients with mild cognitive impairment. Int J Geriatr Psychiatry. (2007) 22:1264–6. 10.1002/gps.1851
    1. Lawlor B, Segurado R, Kennelly S, Olde Rikkert M.G. M., Howard R, et al. . Nilvadipine in mild to moderate alzheimer disease: a randomised controlled trial. PLoS Med. (2018) 15:e1002660. 10.1371/journal.pmed.1002660
    1. Verma N, Beretvas SN, Pascual B, Masdeu JC, Markey MK, Alzheimer's Disease Neuroimaging I. New scoring methodology improves the sensitivity of the Alzheimer's disease assessment scale-cognitive subscale (ADAS-COG) in clinical trials. Alzheimers Res Ther. (2015) 7:64. 10.1186/s13195-015-0151-0
    1. Wang J, Logovinsky V, Hendrix SB, Stanworth SH, Perdomo C, Xu L, et al. . Adcoms: a composite clinical outcome for prodromal Alzheimer's disease trials. J Neurol Neurosurg Psychiatr. (2016) 87:993–9. 10.1136/jnnp-2015-312383
    1. Lawlor B, Kennelly S, O'Dwyer S, Cregg F, Walsh C, Coen R, et al. . Nilvad protocol: a European multicentre double-blind placebo-controlled trial of nilvadipine in mild-to-moderate Alzheimer's disease. BMJ Open. (2014) 4:e006364. 10.1136/bmjopen-2014-006364
    1. Meulenbroek O, O'Dwyer S, de Jong D, van Spijker G, Kennelly S, Cregg F, et al. . European multicentre double-blind placebo-controlled trial of nilvadipine in mild-to-moderate Alzheimer's disease-the substudy protocols: nilvad frailty; nilvad blood and genetic biomarkers; nilvad cerebrospinal fluid biomarkers; nilvad cerebral blood flow. BMJ Open. (2016) 6:e011584. 10.1136/bmjopen-2016-011584
    1. Mattsson N, Andreasson U, Persson S, Carrillo MC, Collins S, Chalbot S, et al. . Csf biomarker variability in the Alzheimer's association quality control program. Alzheimers Dement. (2013) 9:251–61. 10.1016/j.jalz.2013.01.010
    1. Hansson O, Zetterberg H, Buchhave P, Londos E, Blennow K, Minthon L. Association between csf biomarkers and incipient Alzheimer's disease in patients with mild cognitive impairment: a follow-up study. Lancet Neurol. (2006) 5:228–34. 10.1016/S1474-4422(06)70355-6
    1. Mattsson N, Insel PS, Palmqvist S, Portelius E, Zetterberg H, Weiner M, et al. . Cerebrospinal fluid tau, neurogranin, and neurofilament light in Alzheimer's disease. EMBO Mol. Med. (2016) 8:1184–1196. 10.15252/emmm.201606540
    1. Hansson O, Janelidze S, Hall S, Magdalinou N, Lees A, Andreasson U, et al. . Blood-based NFL: a biomarker for differential diagnosis of Parkinsonian disorder. Neurology. (2017) 88:930–7. 10.1212/WNL.0000000000003680
    1. Palmqvist S, Insel PS, Stomrud E, Janelidze S, Zetterberg H, Brix B, et al. . Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer's disease. EMBO Mol Med. (2019) 11:e11170. 10.15252/emmm.201911170
    1. Huntley J, Bor D, Hampshire A, Owen A, Howard R. Working memory task performance and chunking in early Alzheimer's disease. Br J Psychiatry. (2011) 198:398–403. 10.1192/bjp.bp.110.083857
    1. Hendrix S, Logovinsky V, Perdomo C, Wang J, Satlin A. Introducing a new tool for optimizing responsiveness to decline in early ad. Alzheimer's Dement. (2012) 8:S764–5. 10.1016/j.jalz.2013.08.086
    1. Liu-Seifert H, Andersen S, Case M, Sparks J, Holdridge KC, Wessels AM, et al. . Statistical properties of continuous composite scales and implications for drug development. J Biopharm Stat. (2017) 27:1104–14. 10.1080/10543406.2017.1315819
    1. Serrano-Pozo A, Qian J, Muzikansky A, Monsell SE, Montine TJ, Frosch MP, et al. Thal amyloid stages do not significantly impact the correlation between neuropathological change and cognition in the Alzheimer disease continuum. J Neuropathol Exp Neurol. (2016) 75:516–26. 10.1093/jnen/nlw026
    1. Furman JL, Vaquer-Alicea J, White C, III, Cairns NJ, Nelson PT, Diamond MI. Widespread tau seeding activity at early braak stages. Acta Neuropathol. (2017) 133:91–100. 10.1007/s00401-016-1644-z
    1. Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, et al. . Recent publications from the Alzheimer's disease neuroimaging initiative: reviewing progress toward improved ad clinical trials. Alzheimers. Dement. (2017) 13:e1–e85. 10.1016/j.jalz.2016.10.006
    1. Knopman DS. Clinical trial design issues in mild to moderate Alzheimer disease. Cogn Behav Neurol. (2008) 21:197–201. 10.1097/WNN.0b013e318190cf75
    1. Posner H, Curiel R, Edgar C, Hendrix S, Liu E, Loewenstein DA, et al. . Outcomes assessment in clinical trials of Alzheimer's disease and its precursors: readying for short-term and long-term clinical trial needs. Innov Clin Neurosci. (2017) 14:22–29.
    1. Hendrix S, Visser PJ, Solomon A, Kivipelto M, Hartmann T. Adcoms: A Post-Hoc Analysis Using Data From the Lipididiet Trial in Prodromal Alzheimer's Disease. Barcelona: CTAD; (2018).
    1. Swanson CJ, Zhang Y, Dhadda S, Wang J, Kaplow J, Lai YK, et al. Treatment of early ad subjects with ban2401, an ANTI-Aβ protofibril monoclonal antibody, significantly clears amyloid plaque and reduces clinical decline. Alzheimer's Dement J Alzheimer's Assoc. (2018) 14:P1668 10.1016/j.jalz.2018.07.009
    1. Niemantsverdriet E, Ottoy J, Somers C, De Roeck E, Struyfs H, Soetewey F, et al. . The cerebrospinal fluid abeta1-42/abeta1-40 ratio improves concordance with amyloid-pet for diagnosing Alzheimer's disease in a clinical setting. J Alzheimers Dis. (2017) 60:561–76. 10.3233/JAD-170327
    1. Lewczuk P, Riederer P, O'Bryant SE, Verbeek MM, Dubois B, Visser PJ, et al. . Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: an update of the consensus of the task force on biological markers in psychiatry of the world federation of societies of biological psychiatry. World J Biol Psychiatry. (2018) 19:244–328. 10.1080/15622975.2017.1375556
    1. de Jong DLK, de Heus RA, Rijpma A, Donders R, Rikkert O, Günther M, et al. . Effects of nilvadipine on cerebral blood flow in patients with Alzheimer disease. Hypertension. (2019) 74:413–20. 10.1161/HYPERTENSIONAHA.119.12892
    1. Sagare AP, Bell RD, Zlokovic BV. Neurovascular dysfunction and faulty amyloid beta-peptide clearance in alzheimer disease. Cold Spring Harb Perspect Med. (2012) 2:a011452. 10.1101/cshperspect.a011452
    1. Schindler SE, Sutphen CL, Teunissen C, McCue LM, Morris JC, Holtzman DM, et al. . Upward drift in cerebrospinal fluid amyloid beta 42 assay values for more than 10 years. Alzheimers Dement. (2018) 14:62–70. 10.1016/j.jalz.2017.06.2264
    1. Barrera-Ocampo A, Lopera F. Amyloid-beta immunotherapy: the hope for Alzheimer disease? Colomb. Med. (Cali). (2016) 47:203–12. 10.25100/cm.v47i4.2640
    1. Abdullah L, Crawford F, Langlois H, Tsolaki M, Börjesson-Hanson A, Olde Rikkert M, et al. Exploratory analyses suggest less cognitive decline with nilvadipine treatment in very mild Alzheimer's disease subjects. BioRxiv. (2018) 345652 10.1101/345652

Source: PubMed

3
Předplatit