Opinion
Special Issue: Rise of Machines in Medicine
Insights into Computational Drug Repurposing for Neurodegenerative Disease

https://doi.org/10.1016/j.tips.2019.06.003Get rights and content

Highlights

  • Computational drug repurposing has the potential to drastically reduce the cost and development time for therapies for neurodegenerative disease.

  • Advent of artificial intelligence (AI) and machine learning (ML) algorithms will facilitate the integration of several modalities of data to advance computational drug repurposing for neurodegenerative disease.

  • The creation of large-scale transcriptomic and electronic medical record (EMR) databases provides a novel opportunity for computational drug repurposing.

  • Computational drug repurposing for neurodegenerative disease is uniquely challenging due to the lack of efficacious validation methods and long and heterogeneous disease course.

Computational drug repurposing has the ability to remarkably reduce drug development time and cost in an era where these factors are prohibitively high. Several examples of successful repurposed drugs exist in fields such as oncology, diabetes, leprosy, inflammatory bowel disease, among others, however computational drug repurposing in neurodegenerative disease has presented several unique challenges stemming from the lack of validation methods and difficulty in studying heterogenous diseases of aging. Here, we examine existing approaches to computational drug repurposing, including molecular, clinical, and biophysical methods, and propose data sources and methods to advance computational drug repurposing in neurodegenerative disease using Alzheimer’s disease as an example.

Section snippets

Computational Drug Repurposing Can Be a Fast and Cheap Tool for Drug Development

Neurodegenerative disease has posed significant and unique challenges to effective drug discovery over the past century. More than 5 million Americans are living with Alzheimer’s disease (AD) and more than 500 000 have been diagnosed with Parkinson’s disease (PD), the two most common neurodegenerative disorders 1., 2.. Millions more are suffering from rarer conditions, such as frontotemporal dementia (FTD), Huntington’s disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and

Existing Computational Approaches to Drug Repurposing

Previously, successful repurposed drugs came from serendipitous events in the laboratory and clinic. One relevant example of this is the use of zonisamide for the treatment of PD 18., 19.. Upon using zonisamide to treat a Japanese epilepsy patient with PD, Murata discovered that zonisamide also improved the patient’s PD symptoms [18]. Based on this serendipitous finding, zonisamide was approved as an anti-PD therapy in 2009 in Japan. Recently, the emergence of high-throughput molecular,

Lack of Validation Strategies and Economic Incentives Limits the Utility Computational Drug Repurposing for Neurodegenerative Disease

A major obstacle to computational drug repurposing for neurodegenerative disease is the lack of clear validation strategies. While all in silico strategies for computational drug repurposing require in vitro and in vivo experimental validation methods, this problem is more acute in the case of drug repurposing for complex and heterogeneous neurodegenerative disease. Unlike cancer, in which much progress has been made in modeling disease using highly-specific molecular subtype-related cell lines

Concluding Remarks and Future Perspectives

Computational drug repurposing strategies have the potential to dramatically reduce the time and cost of developing therapies for neurodegenerative disease. The advent of large-scale computing capacity combined with the creation of public transcriptomic databases such as CMap, large de-identified EMR databases, and large-scale structural data represent a unique opportunity to apply computational methods to discover novel drugs for neurodegenerative indications. In spite of these resources,

Acknowledgments

The authors thank members of the Sirota laboratory, especially Ishan Paranjpe, for their helpful discussions. M.S. and M.P. are supported by National Institute on Aging (NIA) 1R0AG057683 and R01AG060393. A.T. is supported by NIA F31AG058439.

Disclaimer Statement

Marina Sirota is a former employee of Pfizer and is a scientific advisor to twoXAR.

Glossary

Artificial intelligence (AI)
a field of computer science that develops computer systems which mimic human ability to problem solve and learn. Examples of AI relevant to computational drug repurposing include ML.
Deeply learned autoencoder
a type of AI model that seeks to determine efficient data encodings to represent data in a lower dimension and then reconstruct the original data. Denoising autoencoders introduce noise by randomly setting some of the input values to be missing and learn to

References (142)

  • J.M. Dean

    Probabilistic linkage of computerized ambulance and inpatient hospital discharge records: a potential tool for evaluation of emergency medical services

    Ann. Emerg. Med.

    (2001)
  • E. Tolosa

    The premotor phase of Parkinson’s disease

    Park. Relat. Disord.

    (2007)
  • K.B. Smith

    The effect of statins on rate of cognitive decline in mild cognitive impairment

    Alzheimer’s Dement. Transl. Res. Clin. Interv.

    (2017)
  • J. Shepherd

    Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial

    Lancet

    (2002)
  • M.A. Nance

    Genetics of Huntington disease

    Handb. Clin. Neurol.

    (2017)
  • H.S. Hoe

    The effects of amyloid precursor protein on postsynaptic composition and activity

    J. Biol. Chem.

    (2009)
  • C. Song

    Expression of polyglutamine-expanded huntingtin induces tyrosine phosphorylation of N-methyl-D-aspartate receptors

    J. Biol. Chem.

    (2003)
  • C.L. Gooch

    The burden of neurological disease in the United States: a summary report and call to action

    Ann. Neurol.

    (2017)
  • A. Katsnelson

    Neurodegeneration: from cellular concepts to clinical applications

    Sci. Transl. Med.

    (2016)
  • P. McColgan et al.

    Huntington’s disease: a clinical review

    Eur. J. Neurol.

    (2018)
  • J. Correale

    Progressive multiple sclerosis: from pathogenic mechanisms to treatment

    Brain

    (2017)
  • S.J. Kolb et al.

    Spinal muscular atrophy: a timely review

    Arch. Neurol.

    (2011)
  • M.L. Crismon

    Tacrine: first drug approved for Alzheimer’s disease

    Ann. Pharmacother.

    (1994)
  • J.S. Birks et al.

    Donepezil for dementia due to Alzheimer’s disease

    Cochrane Database Syst. Rev.

    (2018)
  • C. Loy et al.

    Galantamine for Alzheimer’s disease and mild cognitive impairment

    Cochrane Database Syst. Rev.

    (2006)
  • J. Birks et al.

    Rivastigmine for Alzheimer’s disease

    Cochrane Database Syst. Rev.

    (2015)
  • A. Corbett

    Drug repositioning for Alzheimer’s disease

    Nat. Rev. Drug Discov.

    (2012)
  • R.A. Hodos

    In silico methods for drug repurposing and pharmacology

    Wiley Interdiscip. Rev. Syst. Biol. Med.

    (2016)
  • K.I. Kaitin

    Deconstructing the drug development process: the new face of innovation

    Clin. Pharmacol. Ther.

    (2010)
  • L. Li

    Identification of type 2 diabetes subgroups through topological analysis of patient similarity

    Sci. Transl. Med.

    (2015)
  • H. Xue

    Review of drug repositioning approaches and resources

    Int. J. Biol. Sci.

    (2018)
  • Y. Nishimura

    Overcoming obstacles to drug repositioning in Japan

    Front. Pharmacol.

    (2017)
  • M. Sirota

    Discovery and preclinical validation of drug indications using compendia of public gene expression data

    Sci. Transl. Med.

    (2011)
  • J. Li

    A survey of current trends in computational drug repositioning

    Brief. Bioinform.

    (2016)
  • J. Lamb

    The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease

    Science

    (2006)
  • A.M. Brum

    Connectivity Map-based discovery of parbendazole reveals targetable human osteogenic pathway

    Proc. Natl. Acad. Sci. U. S. A.

    (2015)
  • V. Van Noort

    Novel drug candidates for the treatment of metastatic colorectal cancer through global inverse gene-expression profiling

    Cancer Res.

    (2014)
  • J.T. Dudley

    Computational repositioning of the anticonvulsant topiramate for inflammatory bowel disease

    Sci. Transl. Med.

    (2011)
  • E. Ferrero et al.

    Connecting genetics and gene expression data for target prioritisation and drug repositioning

    BioData Min.

    (2018)
  • L.M. Bekris

    Genetics of Alzheimer disease

    J. Geriatr. Psychiatry Neurol.

    (2010)
  • M.A. Beydoun

    Sex differences in the association of the apolipoprotein E epsilon 4 allele with incidence of dementia, cognitive impairment, and decline

    Neurobiol. Aging

    (2012)
  • S.C. Neu

    Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis

    JAMA Neurol.

    (2017)
  • A.F. Curtis

    Sex differences in the prevalence of genetic mutations in FTD and ALS

    Neurology

    (2017)
  • S. Moreno-Grau

    Exploring APOE genotype effects on Alzheimer’s disease risk and amyloid β burden in individuals with subjective cognitive decline: the FundacioACE Healthy Brain Initiative (FACEHBI) study baseline results

    Alzheimers Dement.

    (2017)
  • E.N. Manning

    APOE ε4 is associated with disproportionate progressive hippocampal atrophy in AD

    PLoS One

    (2014)
  • J.S. Damoiseaux

    Gender modulates the APOE 4 effect in healthy older adults: convergent evidence from functional brain connectivity and spinal fluid tau levels

    J. Neurosci.

    (2012)
  • O. Liraz

    ApoE4 induces Aβ42, tau, and neuronal pathology in the hippocampus of young targeted replacement apoE4 mice

    Mol. Neurodegener.

    (2013)
  • J.A. Luchsinger

    Aggregation of vascular risk factors and risk of incident Alzheimer disease

    Neurology

    (2005)
  • M. Kivipelto

    Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study

    BMJ

    (2001)
  • J. Li

    Vascular risk factors promote conversion from mild cognitive impairment to Alzheimer disease

    Neurology

    (2011)
  • Cited by (0)

    View full text