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

Harald Hampel, Coralie Williams, Adrien Etcheto, Federico Goodsaid, Frédéric Parmentier, Jean Sallantin, Walter E Kaufmann, Christopher U Missling, Mohammad Afshar, Harald Hampel, Coralie Williams, Adrien Etcheto, Federico Goodsaid, Frédéric Parmentier, Jean Sallantin, Walter E Kaufmann, Christopher U Missling, Mohammad Afshar

Abstract

Introduction: The search for drugs to treat Alzheimer's disease (AD) has failed to yield effective therapies. Here we report the first genome-wide search for biomarkers associated with therapeutic response in AD. Blarcamesine (ANAVEX2-73), a selective sigma-1 receptor (SIGMAR1) agonist, was studied in a 57-week Phase 2a trial (NCT02244541). The study was extended for a further 208 weeks (NCT02756858) after meeting its primary safety endpoint.

Methods: Safety, clinical features, pharmacokinetic, and efficacy, measured by changes in the Mini-Mental State Examination (MMSE) and the Alzheimer's Disease Cooperative Study-Activities of Daily Living scale (ADCS-ADL), were recorded. Whole exome and transcriptome sequences were obtained for 21 patients. The relationship between all available patient data and efficacy outcome measures was analyzed with unsupervised formal concept analysis (FCA), integrated in the Knowledge Extraction and Management (KEM) environment.

Results: Biomarkers with a significant impact on clinical outcomes were identified at week 57: mean plasma concentration of blarcamesine (slope MMSE:P < .041), genomic variants SIGMAR1 p.Gln2Pro (ΔMMSE:P < .039; ΔADCS-ADL:P < .063) and COMT p.Leu146fs (ΔMMSE:P < .039; ΔADCS-ADL:P < .063), and baseline MMSE score (slope MMSE:P < .015). Their combined impact on drug response was confirmed at week 148 with linear mixed effect models.

Discussion: Confirmatory Phase 2b/3 clinical studies of these patient selection markers are ongoing. This FCA/KEM analysis is a template for the identification of patient selection markers in early therapeutic development for neurologic disorders.

Keywords: Alzheimer's disease; association rules; biomarker; genomic analysis; knowledge extraction management; machine learning; mixed effect models; precision medicine; unsupervised analysis.

Conflict of interest statement

Harald Hampel is an employee of Eisai Inc. and serves as Senior Associate Editor for the Journal Alzheimer's & Dementia; during the past three years he had received lecture fees from Servier, Biogen and Roche, research grants from Pfizer, Avid, and MSD Avenir (paid to the institution), travel funding from Eisai, Functional Neuromodulation, Axovant, Eli Lilly and company, Takeda and Zinfandel, GE‐Healthcare and Oryzon Genomics, consultancy fees from Qynapse, Jung Diagnostics, Cytox Ltd., Axovant, Anavex, Takeda and Zinfandel, GE Healthcare, Oryzon Genomics, and Functional Neuromodulation, and participated in scientific advisory boards of Functional Neuromodulation, Axovant, Eisai, Eli Lilly and company, Cytox Ltd., GE Healthcare, Takeda and Zinfandel, Oryzon Genomics and Roche Diagnostics. He is co‐inventor in the following patents as a scientific expert and has received no royalties: In Vitro Multiparameter Determination Method for The Diagnosis and Early Diagnosis of Neurodegenerative Disorders Patent Number: 8916388. In Vitro Procedure for Diagnosis and Early Diagnosis of Neurodegenerative Diseases Patent Number: 8298784. Neurodegenerative Markers for Psychiatric Conditions Publication Number: 20120196300. In Vitro Multiparameter Determination Method for The Diagnosis and Early Diagnosis of Neurodegenerative Disorders Publication Number: 20100062463. In Vitro Method for The Diagnosis and Early Diagnosis of Neurodegenerative Disorders Publication Number: 20100035286. In Vitro Procedure for Diagnosis and Early Diagnosis of Neurodegenerative Diseases Publication Number: 20090263822. In Vitro Method for The Diagnosis of Neurodegenerative Diseases Patent Number: 7547553. CSF Diagnostic in Vitro Method for Diagnosis of Dementias and Neuroinflammatory Diseases Publication Number: 20080206797. In Vitro Method for The Diagnosis of Neurodegenerative Diseases Publication Number: 20080199966. Neurodegenerative Markers for Psychiatric Conditions Publication Number: 20080131921. Coralie Williams, Adrien Etcheto, and Frédéric Parmentier are employed by Ariana Pharmaceuticals. Mohammad Afshar is employed by and a shareholder of Ariana Pharmaceuticals. Federico Goodsaid is employed by and a shareholder of Regulatory Pathfinders. Walter E. Kaufmann is employed by Anavex Life Sciences Corp. Christopher U. Missling is employed by and a shareholder of Anavex Life Sciences Corp.

© 2020 The Authors. Alzheimer's & Dementia: Translational Research & Clinical Interventions published by Wiley Periodicals, Inc. on behalf of Alzheimer's Association.

Figures

FIGURE 1
FIGURE 1
Overview of study design, data availability, and analytical methods. (a) Summary of consecutive clinical trials ANAVEX2‐73‐002 involving two parts: Part A over 5 weeks and Part B over 52 weeks, and the ANAVEX2‐73‐003 extension study over a planned 208‐week period immediately after the initial trial, with a cumulative 265 weeks. (b) Summary of integrated data sources. A total of 2527 features, from 1152 descriptors, were used for each subject, including 837 genomic sequences with amino acid changes, from a total of 27,155 annotated genes and 185 RNA expression profiles. Patient descriptors are shown in gray and outcomes in pink. (c) Classification of number of patient descriptors incorporated in the two analytical steps applied in the study: (1) Unsupervised FCA rule‐based analysis of response at week 57 and (2) longitudinal confirmation using mixed effect modeling of response over 148 weeks with markers found at week 57 to model two groups of ANAVEX2‐73 concentrations
FIGURE 2
FIGURE 2
Linear mixed effect (LME) models of change in Mini‐Mental State Examination (MMSE) and Alzheimer's Disease Co‐operative Study‐Activities of Daily Living scale (ADCS‐ADL) since baseline over 148 weeks. To analyze the effect of high concentration on outcome over time, significant fixed effect terms linked to concentration were kept in the model. This has the effect of “correcting” for all other parameters except concentration. Because part of the response signal is not explained by the model (random error), this “residual” was added to the adjusted response values. For each time point, the model generates an adjusted predicted outcome for each patient. This adjusted outcome includes the residual mentioned above. For each time point, a mean and standard deviation of the adjusted outcome were calculated for the 21 patients and represented as solid points and error bars linked by dotted lines. A, LME‐adjusted slopes for the high concentration (green) cohort versus low and medium concentration patient cohort (magenta) with time (in weeks) against adjusted change in MMSE. Average adjusted values with residuals at the population level were plotted at each time point (dotted line). B, LME‐adjusted slopes for the high concentration (green) cohort versus low and medium concentration patient cohort (magenta) with time (in weeks) against adjusted change in ADCS‐ADL. Average adjusted values with residuals at the population level were plotted at each time point (dotted line)
FIGURE 3
FIGURE 3
Unadjusted values of change in Alzheimer's Disease Co‐operative Study‐Activities of Daily Living scale (ADCS‐ADL) since baseline over 148 weeks. A, The plot presents (unadjusted) mean trajectories of change in ADCS‐ADL scores over interim 148 weeks of subgroups of ANAVEX2‐73‐002/003 patients depending on biomarker status (absent or present) and of patients given standard of care. The subgroups of ANAVEX2‐73‐002/003 patients are represented (blue, green, orange, pink, purple, and turquoise) with plot depending on identified biomarker and baseline criteria characteristics. B, Summary of the mean and standard deviations of different patient groups unadjusted change in ADCS‐ADL scores at 14 time points (weeks 0, 5, 17, 31, 41, 53, 57, 83, 96, 109, 122, 135, and 148). All patients in the ANAVEX2‐73 study are presented along with each patient subgroup depending on inclusion criteria. The standard of care mean changes from baseline for ADCS‐ADL scores were obtained from the literature with −6.7 point change in 1 year 42 and −10.5 change in 18 months/70 wk 43

References

    1. Cummings J, Lee G, Ritter A, Zhong K. Alzheimer's disease drug development pipeline: 2018. Alzheimers Dement Transl Res Clin Interv. 2018;4:195‐214.
    1. Dong A, Toledo JB, Honnorat N, et al. Heterogeneity of neuroanatomical patterns in prodromal Alzheimer's disease: links to cognition, progression and biomarkers. Brain J Neurol. 2017;140(3):735‐747.
    1. Gamberger D, Lavrač N, Srivatsa S, Tanzi RE, Doraiswamy PM. Identification of clusters of rapid and slow decliners among subjects at risk for Alzheimer's disease. Sci Rep. 2017;7(1):6763.
    1. Evans S, McRae‐McKee K, Wong MM, Hadjichrysanthou C, De Wolf F, Anderson R. The importance of endpoint selection: how effective does a drug need to be for success in a clinical trial of a possible Alzheimer's disease treatment?. Eur J Epidemiol. 2018;33(7):635‐644.
    1. Su T‐P, Su T‐C, Nakamura Y, Tsai S‐Y. Sigma‐1 receptor as a pluripotent modulator in the living system. Trends Pharmacol Sci. 2016;37(4):262‐278.
    1. Nguyen L, Lucke‐Wolds BP, Mookerjee S, Kaushal N, Matsumoto RR. Sigma‐1 receptors and neurodegenerative diseases: towards a hypothesis of sigma‐1 receptors as amplifiers of neurodegeneration and neuroprotection. Adv Exp Med Biol. 2017;964:133‐152.
    1. Ruscher K, Wieloch T. The involvement of the sigma‐1 receptor in neurodegeneration and neurorestoration. J Pharmacol Sci. 2015;127(1):30‐35.
    1. Mavlyutov TA, Epstein ML, Verbny YI, et al. Lack of sigma‐1 receptor exacerbates ALS progression in mice. Neuroscience. 2013;240:129‐134.
    1. Christ MG, Huesmann H, Nagel H, Kern A, Behl C. Sigma‐1 receptor activation induces autophagy and increases proteostasis capacity in vitro and in vivo. Cells. 2019;8(3):211.
    1. Reyes ST, Guo SG, Castillo JB, et al. Sigma‐1 receptor target occupancy study with dynamic PET scan analysis of ANAVEX®2‐73, a clinical candidate for neurodegenerative and neurodevelopmental diseases. Alzheimers Dement J Alzheimers Assoc. 2018;14(7):P1547.
    1. Lahmy V, Meunier J, Malmström S, et al. Blockade of tau hyperphosphorylation and Aβ1‐42 generation by the aminotetrahydrofuran derivative ANAVEX2‐73, a mixed muscarinic and σ1 receptor agonist, in a nontransgenic mouse model of Alzheimer's disease. Neuropsychopharmacology. 2013;38(9):1706‐1723.
    1. Villard V, Espallergues J, Keller E, Vamvakides A, Maurice T. Anti‐amnesic and neuroprotective potentials of the mixed muscarinic receptor/sigma 1 (σ1) ligand ANAVEX2‐73, a novel aminotetrahydrofuran derivative. J Psychopharmacol Oxf Engl. 2011;25(8):1101‐1117.
    1. Voges O, Weigmann I, Bitterlich N, Missling C, Schindler C. A Phase I Dose Escalation Study to Investigate Safety, Tolerability, and Pharmacokinetics of ANAVEX 2‐73 in Healthy Male Subjects. CNS Summit 2014. Innovations in Clinical Neuroscience, 12(Suppl B), 1‐20.
    1. Macfarlane S, Maruff P, Cecchi M, Moore D, Zografidis T, Missling C. New exploratory Alzheimer's drug anavex 2‐73: dose dependent clinical cognitive improvement observed in Mini Mental State Examination (MMSE) and other cognitive markers in a Phase 2a study in mild‐to‐moderate alzheimer's patients. Alzheimers Dement J Alzheimers Assoc. 2016;12(7):P419.
    1. Hyman DM, Taylor BS, Baselga J. Implementing genome‐driven oncology. Cell. 2017;168(4):584‐599.
    1. Kelloff GJ, Sigman CC. Cancer biomarkers: selecting the right drug for the right patient. Nat Rev Drug Discov. 2012;11:201.
    1. Morris‐Rosendahl DJ, Fiebich BL. The future of genetic testing for drug response. Dialogues Clin Neurosci. 2004;6(1):27‐37.
    1. Roden DM, Wilke RA, Kroemer HK, Stein CM. Pharmacogenomics: the genetics of variable drug responses. Circulation. 2011;123(15):1661‐1670.
    1. Lee CH, Yoon H‐J. Medical big data: promise and challenges. Kidney Res Clin Pract. 2017;36(1):3.
    1. Salsburg D. Hundreds of patients, thousands of observations: the curse of dimensionality in clinical research. Drug Inf J. 1993;27(3):597‐609.
    1. Sinha A, Hripcsak G, Markatou M. Large datasets in biomedicine: a discussion of salient analytic issues. J Am Med Inform Assoc. 2009;16(6):759‐767.
    1. Jullian N, Jourdan N, Afshar M. Hypothesis generation for scientific discovery. Examples from the use of KEM®, a rule‐based method for multi‐objective analysis and optimization. Solvay Pharmaceuticals Conferences. 2008;9: Towards Drugs of the Future: Key Issues in Lead Finding and Lead Optimization:75‐80. 10.3233/978-1-58603-949-3-75
    1. Liquiere M, Sallantin J. Structural Machine Learning with Galois Lattice and Graphs. Proceedings of the Fifteenth International Conference on Machine Learning. 1998:305‐313.
    1. Guze SB. Diagnostic and Statistical Manual of Mental Disorders. 4th ed. (DSM‐IV). Am J Psychiatry. 1995;152(8):1228‐1228. 10.1176/ajp.152.8.1228
    1. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer's disease. Neurology. 1984;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 Psychiatr Res. 1975;12(3):189‐198.
    1. Galasko D, Bennett D, Sano M, et al. 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.
    1. Marsico M, Grant A, Chandler J. The reliability and validity of the Alzheimer's disease cooperative study–activities of daily living (ADCS‐ADL) in China. Alzheimers Dement J Alzheimers Assoc. 2015;11(7):P444.
    1. Illumina . Understanding Illumina Quality Scores 2012. Available at: .
    1. Ewing B, Hillier L, Wendl MC, Green P. Base‐calling of automated sequencer traces using phred. I. Accuracy assessment. Genome Res. 1998;8(3):175‐185.
    1. Cingolani P, Platts A, Wang LL, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff. Fly (Austin). 2012;6(2):80‐92.
    1. Li B, Ruotti V, Stewart RM, Thomson JA, Dewey CN. RNA‐Seq gene expression estimation with read mapping uncertainty. Bioinformatics. 2010;26(4):493‐500.
    1. Wagner GP, Kin K, Lynch VJ. Measurement of mRNA abundance using RNA‐Seq data: RPKM measure is inconsistent among samples. ResearchGate. 2012;131(4):281‐285.
    1. Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality‐controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(Database issue):D362‐D368.
    1. Ganter B, Wille R. Formal Concept Analysis: Mathematical Foundations. 1st ed Berlin, Heidelberg, Germany: Springer‐Verlag; 1997.
    1. Gasmi Gh, Yahia SB, Nguifo EM, Slimani Y. IGB: A new informative generic base of association rules In: Ho TB, Cheung D, Liu H, eds. Advances in Knowledge Discovery and Data Mining. Heidelberg, Germany: Springer Berlin Heidelberg; 2005:81‐90. 10.1007/11430919_11
    1. Nebot V, Berlanga R. Finding association rules in semantic web data. Knowl‐Based Syst. 2012;25(1):51‐62.
    1. Zhao Y, Zhang H, Figueiredo F, Cao L, Zhang C. Mining for Combined Association Rules on Multiple Datasets. In: Proceedings of the 2007 International Workshop on Domain Driven Data Mining DDDM ’07. New York, NY, USA: ACM; 2007:18‐23. 10.1145/1288552.1288555
    1. Srikant R, Agrawal R. Mining generalized association rules. Data Min. 1997;13(2):161‐180.
    1. Agrawal R, Imieliński T, Swami A. Mining Association Rules between Sets of Items in Large Databases. In: New York, NY, USA; 1993:207‐216. 10.1145/170036.170072. Accessed November 12, 2018.
    1. Afshar M, Lanoue A, Sallantin J. Multiobjective/Multicriteria Optimization and Decision Support in Drug Discovery. Comprehensive Medicinal Chemistry II. 2007:4:767‐774. 10.1016/B0-08-045044-X/00275-3
    1. Abtroun L, Bunouf P, Gendreau RM, Vitton O. Is the efficacy of milnacipran in fibromyalgia predictable? A data‐mining analysis of baseline and outcome variables. Clin J Pain. 2016;32(5):435‐440.
    1. Williams C, Polom K, Adamczyk B, et al. Machine learning methodology applied to characterize subgroups of gastric cancer patients using an integrated large biomarker dataset. Eur J Surg Oncol. 2019;45(2):e79.
    1. Breuer R, Mattheisen M, Frank J, et al. Detecting significant genotype–phenotype association rules in bipolar disorder: market research meets complex genetics. Int J Bipolar Disord. 2018;6(1):24.
    1. Blattberg RC, Kim B‐D, Neslin SA. Market basket analysis In: Blattberg RC, Kim B‐D, Neslin SA, eds. Database Marketing: Analyzing and Managing Customers. International Series in Quantitative Marketing. New York, NY: Springer New York; 2008:339‐351. 10.1007/978-0-387-72579-6_13
    1. Tan PN, Steinbach M, Kumar V. Introduction to Data Mining. 1st ed Boston, MA: Addison‐Wesley Longman Publishing Co.; 2005.
    1. Chen Y‐F, Ni X, Fleisher AS, Zhou W, Aisen P, Mohs R. A simulation study comparing slope model with mixed‐model repeated measure to assess cognitive data in clinical trials of Alzheimer's disease. Alzheimers Dement Transl Res Clin Interv. 2018;4:46‐53.
    1. Broadhouse KM, Suo C, Singh MAF, et al. What happens to the hippocampus 12‐months after training? Longitudinal linear mixed effects model analysis of mild cognitive impairment in the smart trial. Alzheimers Dement J Alzheimers Assoc. 2017;13(7):P260.
    1. R Core Team . R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2018. Available at:
    1. Bates D, Maechler M, Bolker B, et al. Lme4: Linear Mixed‐Effects Models Using “Eigen” and S4 2018. Available at:
    1. Cohen J. Statistical Power Analysis for the Behavioral Sciences. New York, NY: Routledge Academic; 1988.
    1. Roses AD, Lutz MW, Amrine‐Madsen H, et al. A TOMM40 variable‐length polymorphism predicts the age of late‐onset Alzheimer's disease. Pharmacogenomics J. 2010;10(5):375‐384.
    1. Alexander R, Burns DK, Welsh‐Bohmer KA, et al. DT‐02‐02: Tommorrow: Results from a phase 3 trial to delay the onset of MCI due to AD and qualify a genetic biomarker algorithm. Alzheimer's and Dementia. 2019;15(7):P1488‐P1489. 10.1016/j.jalz.2019.08.011
    1. Ou S‐HI. Crizotinib: a novel and first‐in‐class multitargeted tyrosine kinase inhibitor for the treatment of anaplastic lymphoma kinase rearranged non‐small cell lung cancer and beyond. Drug Des Devel Ther. 2011;5:471‐485.
    1. Bruder GE, Keilp JG, Xu H, et al. Catechol‐O‐methyltransferase (COMT) genotypes and working memory: associations with differing cognitive operations. Biol Psychiatry. 2005;58(11):901‐907.
    1. de Frias CM, Annerbrink K, Westberg L, Eriksson E, Adolfsson R, Nilsson L‐G. COMT gene polymorphism is associated with declarative memory in adulthood and old age. Behav Genet. 2004;34(5):533‐539.
    1. Hilderman RJ, Hamilton HJ. Evaluation of interestingness measures for ranking discovered knowledge In: Cheung D, Williams GJ, Li Q, eds. Advances in Knowledge Discovery and Data Mining. Lecture Notes in Computer Science. Heidelberg, Germany: Springer Berlin Heidelberg; 2001:247‐259.
    1. Muth C, Bales KL, Hinde K, Maninger N, Mendoza SP, Ferrer E. Alternative models for small samples in psychological research. Educ Psychol Meas. 2016;76(1):64‐87.
    1. Swanson C. Clinical and Biomarker Updates from BAN2401 Study 201 in Early AD: Pre‐specified Subgroup Analysis in BAN2401 Study 201. Presented at the: October 24, 2018; 11th Clinical Trials on Alzheimer's Disease (CTAD) Conference. Barcelona, Spain. . Accessed: February, 2020.
    1. Cummings J. Clinical and Biomarker Updates from BAN2401 Study 201 in Early AD: BAN2401 Study 201 Design and Topline Results. Presented at the: 11th Clinical Trials on Alzheimer's Disease (CTAD) Conference; October 24, 2018; Barcelona, Spain. . Accessed: February, 2020.
    1. Clark C, Sheppard L, Fillenbaum G, et al. Variability in annual Mini‐Mental State Examination score in patients with probable Alzheimer disease: a clinical perspective of data from the consortium to establish a registry for alzheimer's disease. Arch Neurol. 1999;56(7):857‐862.
    1. Han L, Cole M, Bellavance F, McCusker J, Primeau F. Tracking cognitive decline in Alzheimer's disease using the Mini‐Mental State Examination: a meta‐analysis. Int Psychogeriatr. 2000;12(2):231‐247.
    1. Maher‐Edwards G, Watson C, Ascher J, et al. Two randomized controlled trials of SB742457 in mild‐to‐moderate Alzheimer's disease. Alzheimers Dement Transl Res Clin Interv. 2015;1(1):23‐36.
    1. Schneider LS, Sano M. Current Alzheimer's disease clinical trials: methods and placebo outcomes. Alzheimers Dement J Alzheimers Assoc. 2009;5(5):388‐397.

Source: PubMed

3
Sottoscrivi