Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Endophenotype-based in silico network medicine discovery combined with insurance record data mining identifies sildenafil as a candidate drug for Alzheimer’s disease

Matters Arising to this article was published on 22 May 2023

An Author Correction to this article was published on 20 March 2023

This article has been updated

Abstract

We developed an endophenotype disease module-based methodology for Alzheimer’s disease (AD) drug repurposing and identified sildenafil as a potential disease risk modifier. Based on retrospective case–control pharmacoepidemiologic analyses of insurance claims data for 7.23 million individuals, we found that sildenafil usage was significantly associated with a 69% reduced risk of AD (hazard ratio 0.31, 95% confidence interval 0.25–0.39, P < 1.0 × 10–8). Propensity score-stratified analyses confirmed that sildenafil is significantly associated with a decreased risk of AD across all four drug cohorts tested (diltiazem, glimepiride, losartan and metformin) after adjusting for age, sex, race and disease comorbidities. We also found that sildenafil increases neurite growth and decreases phospho-tau expression in neuron models derived from induced pluripotent stem cells from patients with AD, supporting mechanistically its potential beneficial effect in AD. The association between sildenafil use and decreased incidence of AD does not establish causality, which will require a randomized controlled trial.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: A diagram illustrating an endophenotype network-based drug repurposing framework for AD.
Fig. 2: Network-based in silico drug repurposing for AD.
Fig. 3: Longitudinal analyses reveal that sildenafil usage is significantly associated with reduced likelihood of AD in a longitudinal patient database with 7.23 million individuals.
Fig. 4: Subgroup analyses of five drug cohort studies to evaluate confounding by disease comorbidities.
Fig. 5: Sex-specific and age-specific subgroup analyses across five drug cohort studies.
Fig. 6: Experimental validation of sildenafil’s likely mechanism of action in AD.

Similar content being viewed by others

Data availability

Data supporting the findings of this study are available within Supplementary Tables. The human protein–protein interactome and drug-target network can be downloaded from https://github.com/ChengF-Lab/endoAD. Tissue/brain-specific expression data were downloaded from GTEx database (https://www.gtexportal.org/home/). The expression datasets used in this study were downloaded from the Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/geo/) with accession codes GSE65067, GSE74437, GSE74438, GSE64398, GSE53480, GSE56772 and GSE57583. Health insurance claims data are available from the MarketScan Medicare Claims database (2012 to 2017) based on the Medicare Advantage and Fee for services from IBM MarketScan Research Databases.

Code availability

Source codes for network proximity analysis and disease module identification are available at https://github.com/ChengF-Lab/GPSnet.

Change history

References

  1. Alzheimer’s, A. 2016 Alzheimer’s disease facts and figures. Alzheimers Dement. 12, 459–509 (2016).

    Article  Google Scholar 

  2. Espay, A. J. et al. Revisiting protein aggregation as pathogenic in sporadic Parkinson and Alzheimer diseases. Neurology 92, 329–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Voorhees, J. R. et al. (-)-P7C3-S243 protects a rat model of Alzheimer’s disease from neuropsychiatric deficits and neurodegeneration without altering amyloid deposition or reactive glia. Biol. Psychiatry 84, 488–498 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Long, J. M. & Holtzman, D. M. Alzheimer disease: an update on pathobiology and treatment strategies. Cell 179, 312–339 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Makin, S. The amyloid hypothesis on trial. Nature 559, S4–S7 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Selkoe, D. J. & Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cummings, J. L., Morstorf, T. & Zhong, K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res. Ther. 6, 37 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Tasaki, S., Gaiteri, C., Mostafavi, S., De Jager, P. L. & Bennett, D. A. The molecular and neuropathological consequences of genetic risk for Alzheimer’s dementia. Front. Neurosci. 12, 699 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Nelson, M. R. et al. The support of human genetic evidence for approved drug indications. Nat. Genet. 47, 856–860 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Wang, Q. et al. A Bayesian framework that integrates multi-omics data and gene networks predicts risk genes from schizophrenia GWAS data. Nat. Neurosci. 22, 691–699 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fang, H., De Wolf, H., Knezevic, B. & Burnham, K. L. A genetics-led approach defines the drug target landscape of 30 immune-related traits. Nat. Genet. 51, 1082–1091 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fang, J. et al. Harnessing endophenotypes and network medicine for Alzheimer’s drug repurposing. Med. Res. Rev. 40, 2386–2426 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ghiassian, D. et al. Endophenotype network models: common core of complex diseases. Sci. Rep. 6, 27414 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Swarup, V. et al. Identification of evolutionarily conserved gene networks mediating neurodegenerative dementia. Nat. Med. 25, 152–164 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Chung, J. et al. Genome-wide association study of Alzheimer’s disease endophenotypes at prediagnosis stages. Alzheimers Dement. 14, 623–633 (2018).

    Article  PubMed  Google Scholar 

  16. Pascoal, T. A. et al. Synergistic interaction between amyloid and tau predicts the progression to dementia. Alzheimers Dement. 13, 644–653 (2017).

    Article  PubMed  Google Scholar 

  17. Benbow, S. J. et al. Synergistic toxicity between tau and amyloid drives neuronal dysfunction and neurodegeneration in transgenic C. elegans. Hum. Mol. Genet. 29, 495–505 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pickett, E. K. et al. Amyloid β and tau cooperate to cause reversible behavioral and transcriptional deficits in a model of Alzheimer’s disease. Cell Rep. 29, 3592–3604.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Greene, J. A. & Loscalzo, J. Putting the patient back together: social medicine, network medicine, and the limits of reductionism. N. Engl. J. Med. 377, 2493–2499 (2017).

    Article  PubMed  Google Scholar 

  20. Goh, K. I. et al. The human disease network. Proc. Natl Acad. Sci. USA 104, 8685–8690 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cheng, F. et al. Network-based approach to prediction and population-based validation of in silico drug repurposing. Nat. Commun. 9, 2691 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Huang, Y. et al. A systems pharmacology approach uncovers wogonoside as an angiogenesis inhibitor of triple-negative breast cancer by targeting Hedgehog signaling. Cell Chem. Biol. 26, 1143–1158 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cheng, F., Kovacs, I. A. & Barabasi, A. L. Network-based prediction of drug combinations. Nat. Commun. 10, 1197 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Cheng, F. et al. A genome-wide positioning systems network algorithm for in silico drug repurposing. Nat. Commun. 10, 3476 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Guney, E. et al. Network-based in silico drug efficacy screening. Nat. Commun. 7, 10331 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mittal, S. et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 357, 891–898 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bayik, D. et al. Myeloid-derived suppressor cell subsets drive glioblastoma growth in a sex-specific manner. Cancer Discov. 10, 1210–1225 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Claassen, J. A. New cardiovascular targets to prevent late-onset Alzheimer disease. Eur. J. Pharmacol. 763, 131–134 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Aguirre-Plans, J. et al. Proximal pathway enrichment analysis for targeting comorbid diseases via network endopharmacology. Pharmaceuticals 11, 61 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhou, Y. et al. AlzGPS: a genome-wide positioning systems platform to catalyze multi-omics for Alzheimer’s therapeutic discovery. Alzheimer’s Res. Ther. 13, 24 (2021).

    Article  Google Scholar 

  31. Cheng, F. et al. admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties. J. Chem. Inf. Model. 52, 3099–3105 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Gomm, W. et al. Association of proton pump inhibitors with risk of dementia: a pharmacoepidemiological claims data analysis. JAMA Neurol. 73, 410–416 (2016).

    Article  PubMed  Google Scholar 

  33. Cuadrado-Tejedor, M. et al. Sildenafil restores cognitive function without affecting β-amyloid burden in a mouse model of Alzheimer’s disease. Br. J. Pharmacol. 164, 2029–2041 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ding, J. et al. Antihypertensive medications and risk for incident dementia and Alzheimer’s disease: a meta-analysis of individual participant data from prospective cohort studies. Lancet Neurol. 19, 61–70 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Xue, M. et al. Diabetes mellitus and risks of cognitive impairment and dementia: a systematic review and meta-analysis of 144 prospective studies. Ageing Res. Rev. 55, 100944 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

  37. Huang, Y. & Mucke, L. Alzheimer mechanisms and therapeutic strategies. Cell 148, 1204–1222 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yuskaitis, C. J. & Jope, R. S. Glycogen synthase kinase-3 regulates microglial migration, inflammation, and inflammation-induced neurotoxicity. Cell Signal. 21, 264–273 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Na, Y. R. et al. The early synthesis of p35 and activation of CDK5 in LPS-stimulated macrophages suppresses interleukin-10 production. Sci. Signal. 8, ra121 (2015).

    Article  PubMed  Google Scholar 

  40. Karikari, T. K. et al. Diagnostic performance and prediction of clinical progression of plasma phospho-tau181 in the Alzheimer’s Disease Neuroimaging Initiative. Mol. Psychiatry 26, 429–442 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Su, M. et al. Mechanisms associated with type 2 diabetes as a risk factor for Alzheimer-related pathology. Mol. Neurobiol. 56, 5815–5834 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Gabin, J. M., Tambs, K., Saltvedt, I., Sund, E. & Holmen, J. Association between blood pressure and Alzheimer disease measured up to 27 years prior to diagnosis: the HUNT study. Alzheimers Res. Ther. 9, 37 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Wang, J. et al. Carvedilol as a potential novel agent for the treatment of Alzheimer’s disease. Neurobiol. Aging 32, 2321 (2011).

    Article  Google Scholar 

  44. Heckman, P. R. A., Blokland, A. & Prickaerts, J. From age-related cognitive decline to Alzheimer’s disease: a translational overview of the potential role for phosphodiesterases. Adv. Neurobiol. 17, 135–168 (2017).

    Article  PubMed  Google Scholar 

  45. Li, C. & Gotz, J. Tau-based therapies in neurodegeneration: opportunities and challenges. Nat. Rev. Drug Discov. 16, 863–883 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Plattner, F., Angelo, M. & Giese, K. P. The roles of cyclin-dependent kinase 5 and glycogen synthase kinase 3 in tau hyperphosphorylation. J. Biol. Chem. 281, 25457–25465 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Katsumoto, A., Takeuchi, H., Takahashi, K. & Tanaka, F. Microglia in Alzheimer’s disease: risk factors and inflammation. Front. Neurol. 9, 978 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Du, J. et al. Inhibition of CDKS by roscovitine suppressed LPS-induced *NO production through inhibiting NFκB activation and BH4 biosynthesis in macrophages. Am. J. Physiol. Cell Physiol. 297, C742–C749 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhang, J. et al. Phosphodiesterase-5 inhibitor sildenafil prevents neuroinflammation, lowers β-amyloid levels and improves cognitive performance in APP/PS1 transgenic mice. Behav. Brain Res. 250, 230–237 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Puzzo, D. et al. Phosphodiesterase 5 inhibition improves synaptic function, memory, and amyloid-β load in an Alzheimer’s disease mouse model. J. Neurosci. 29, 8075–8086 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. García-Barroso, C. et al. Tadalafil crosses the blood–brain barrier and reverses cognitive dysfunction in a mouse model of AD. Neuropharmacology. 64, 114–123 (2013).

    Article  PubMed  Google Scholar 

  52. Samudra, N. et al. A pilot study of changes in medial temporal lobe fractional amplitude of low frequency fluctuations after sildenafil administration in patients with Alzheimer’s disease. J. Alzheimers Dis. 70, 163–170 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sheng, M. et al. Sildenafil improves vascular and metabolic function in patients with Alzheimer’s disease. J. Alzheimers Dis. 60, 1351–1364 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Menche, J. et al. Uncovering disease-disease relationships through the incomplete interactome. Science 347, 1257601 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Cummings, J. et al. Aducanumab: appropriate use ecommendations. J. Prev. Alzheimers Dis. 8, 398–410 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Sienski, G. et al. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci. Transl. Med. 13, eaaz4564 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cadar, D. et al. Individual and area-based socioeconomic factors associated with dementia incidence in England: evidence from a 12-year follow-up in the English Longitudinal Study of Ageing. JAMA Psychiatry 75, 723–732 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Ghofrani, H. A. et al. Sildenafil: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nat. Rev. Drug Discov. 5, 689–702 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Andrews, S. J. et al. Mendelian randomization indicates that TNF is not causally associated with Alzheimer’s disease. Neurobiol. Aging 84, 241.e1–241.e3 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Williams, D. M. et al. Lipid lowering and Alzheimer disease risk: a mendelian randomization study. Ann. Neurol. 87, 30–39 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Rolland, T. et al. A proteome-scale map of the human interactome network. Cell 159, 1212–1226 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Rual, J. F. et al. Towards a proteome-scale map of the human protein–protein interaction network. Nature 437, 1173–1178 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Csabai, L., Olbei, M., Budd, A., Korcsmaros, T. & Fazekas, D. SignaLink: multilayered regulatory networks. Methods Mol. Biol. 1819, 53–73 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Stenson, P. D. et al. The Human Gene Mutation Database: towards a comprehensive repository of inherited mutation data for medical research, genetic diagnosis and next-generation sequencing studies. Hum. Genet. 136, 665–677 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Pinero, J. et al. DisGeNET: a comprehensive platform integrating information on human disease-associated genes and variants. Nucleic Acids Res. 45, D833–D839 (2017).

    Article  CAS  PubMed  Google Scholar 

  66. Rappaport, N. et al. MalaCards: an amalgamated human disease compendium with diverse clinical and genetic annotation and structured search. Nucleic Acids Res. 45, D877–D887 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. Koscielny, G. et al. Open Targets: a platform for therapeutic target identification and validation. Nucleic Acids Res. 45, D985–D994 (2017).

    Article  CAS  PubMed  Google Scholar 

  68. Edgar, R., Domrachev, M. & Lash, A. E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Bouter, Y. et al. Deciphering the molecular profile of plaques, memory decline and neuron loss in two mouse models for Alzheimer’s disease by deep sequencing. Front. Aging Neurosci. 6, 75 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Grubman, A., Choo, X. Y. & Chew, G. et al. Transcriptional signature in microglia associated with Aβ plaque phagocytosis. Nat. Commun. 12, 3015 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Rothman, S. M. et al. Human Alzheimer’s disease gene expression signatures and immune profile in APP mouse models: a discrete transcriptomic view of Aβ plaque pathology. J. Neuroinflammation 15, 256 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wang, H. et al. Genome-wide RNA-seq study of the molecular mechanisms underlying microglia activation in response to pathological tau perturbation in the rTg4510 tau transgenic animal model. Mol. Neurodegener. 13, 65 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Benjamini, Y. & Yekutieli, D. The control of the false discovery rate in multiple testing under dependency. Ann. Stat. 29, 1165–1188 (2001).

    Article  Google Scholar 

  75. Eppig, J. T. et al. Mouse Genome Informatics (MGI): resources for mining mouse genetic, genomic, and biological data in support of primary and translational research. Methods Mol. Biol. 1488, 47–73 (2017).

    Article  CAS  PubMed  Google Scholar 

  76. Savas, J. N. et al. Amyloid accumulation drives proteome-wide alterations in mouse models of Alzheimer’s disease-like pathology. Cell Rep. 21, 2614–2627 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kim, D. K. et al. Molecular and functional signatures in a novel Alzheimer’s disease mouse model assessed by quantitative proteomics. Mol. Neurodegener. 13, 2 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2018).

    Article  CAS  PubMed  Google Scholar 

  79. Yunxia, W. et al. Therapeutic target database 2020: enriched resource for facilitating research and early development of targeted therapeutics. Nucleic Acids Res. 48, D1031–D1041 (2019).

    Google Scholar 

  80. Barbarino, J. M., Whirl-Carrillo, M., Altman, R. B. & Klein, T. E. PharmGKB: a worldwide resource for pharmacogenomic information. Wiley Interdiscip. Rev. Syst. Biol. Med. 10, e1417 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Gaulton, A. et al. The ChEMBL database in 2017. Nucleic Acids Res. 45, D945–D954 (2017).

    Article  CAS  PubMed  Google Scholar 

  82. Gilson, M. K. et al. BindingDB in 2015: a public database for medicinal chemistry, computational chemistry and systems pharmacology. Nucleic Acids Res. 44, D1045–D1053 (2016).

    Article  CAS  PubMed  Google Scholar 

  83. Harding, S. D. et al. The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new Guide to IMMUNOPHARMACOLOGY. Nucleic Acids Res. 46, D1091–D1106 (2018).

    Article  CAS  PubMed  Google Scholar 

  84. Desai, R. J. et al. Targeting abnormal metabolism in Alzheimer’s disease: the Drug Repurposing for Effective Alzheimer’s Medicines (DREAM) study. Alzheimers Dement. 6, e12095 (2020).

    Google Scholar 

  85. Lee, E. et al. Evaluation of Medicare claims data as a tool to identify dementia. J. Alzheimers Dis. 67, 769–778 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Charlson, M. E. et al. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J. Chronic Dis. 40, 373–383 (1987).

    Article  CAS  PubMed  Google Scholar 

  87. Lash, T. L. et al. Methodology, design, and analytic techniques to address measurement of comorbid disease. J. Gerontol. A Biol. Sci. Med. Sci. 62, 281–285 (2007).

    Article  PubMed  Google Scholar 

  88. Ye, J. et al. Three data-driven phenotypes of multiple organ dysfunction syndrome preserved from early childhood to middle adulthood. AMIA Annu. Symp. Proc. 2020, 1345–1353 (2021).

    PubMed  PubMed Central  Google Scholar 

  89. Kitsak, M. et al. Tissue specificity of human disease module. Sci. Rep. 6, 35241 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Son, Y., Kim, K. & Cho, H.-R. Sildenafil protects neuronal cells from mitochondrial toxicity induced by β-amyloid peptide via ATP-sensitive K channels. Biochem. Biophys. Res. Commun. 500, 504–510 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was primarily supported by the National Institute of Aging (NIA) of the National Institutes of Health (NIH) under Award Number R01AG066707 to F.C. This work was supported in part by U01AG073323, 3R01AG066707-01S1, 3R01AG066707-02S1, and 1R56AG074001-01 to F.C. This work was supported in part by the Translational Therapeutics Core of the Cleveland Alzheimer’s Disease Research Center (NIH/NIA: P30AG072959) to F.C., A.A.P. and J.C. This work was supported in part by the Brockman Foundation, Project 19PABH134580006-AHA/Allen Initiative in Brain Health and Cognitive Impairment, the Elizabeth Ring Mather & William Gwinn Mather Fund, S. Livingston Samuel Mather Trust, and the Louis Stokes VA Medical Center resources and facilities to A.A.P. This work was supported in part by the NIA grant R35AG071476 and the Alzheimer’s Disease Drug Discovery Foundation (ADDF) to J.C.

Author information

Authors and Affiliations

Authors

Contributions

F.C. conceived the study. J.F. and Y.Z. performed all multi-omics and network proximity experiments and analysis. P.Z. and C.W.C. performed all patient data analysis. J.T. performed human microglia and iPSC experiments and data analysis. Y.H., S.S., A.A.P., L.L. and J.C. interpreted the data analysis. F.C., J.F., P.Z., A.A.P. and J.C. drafted the manuscript. All authors critically revised and approved the manuscript.

Corresponding author

Correspondence to Feixiong Cheng.

Ethics declarations

Competing interests

J.C. has provided consultation to Acadia, Actinogen, Alkahest, Alzheon, Annovis, Avanir, Axsome, Biogen, BioXcel, Cassava, Cerecin, Cerevel, Cortexyme, Cytox, EIP Pharma, Eisai, Foresight, GemVax, Genentech, Green Valley, Grifols, Karuna, Merck, Novo Nordisk, Otsuka, Resverlogix, Roche, Samumed, Samus, Signant Health, Suven, Third Rock and United Neuroscience pharmaceutical and assessment companies. J.C. has stock options in ADAMAS, AnnovisBio, MedAvante and BiOasis. The other authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks Heather Allore, Emre Guney and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Proof-of-concept of network module of Alzheimer’s disease (AD).

A subnetwork highlighting disease module (AD seed module) characterized by both amyloidosis and tauopathy under the human protein-protein interactome model. The AD seed module includes 227 protein–protein interactions (PPIs) (edges or links) connecting 102 unique proteins (nodes).

Extended Data Fig. 2 Heatmap illustrates the network proximity between molecular targets of 21 ongoing repurposable AD drugs and 37 AD disease modules.

These drugs could target amyloid, tau, or both amyloid and tau pathologies or their related pathways in in vitro or in vivo mouse models or in patients with AD. We built three AD network modules by assembling experimentally validated (seed) genes in amyloidosis (amyloid), tauopathy (tau) and AD (characterized by both amyloid and tau). In addition, we also built disease modules from 34 differentially expressed gene (DEG) sets derived from transcriptomics data (including microarray and bulk RNA-sequencing) from AD genetic mouse.

Extended Data Fig. 3 The efficacy of endophenotype-based repurposable drugs in Alzheimer’s disease (AD).

Molecular targets of 21 ongoing repurposable AD drugs that target both Amyloid and Tau have significantly closer network distance with AD network modules built from transcriptomics (a) and proteomics (b) data from AD genetic mouse, in comparison to drugs targeting amyloid or tau alone. In a, we built AD modules from 34 differentially expressed gene sets derived from transcriptomics data (including microarray, and bulk RNA-sequencing) from AD genetic mouse. In b, we built AD modules for 10 differentially expressed protein sets from proteomics data in AD genetic mouse. P values were calculated by Wilcoxon test (one-side) in a and b.

Extended Data Fig. 4 Network-based in silico drug repurposing for Alzheimer’s disease (AD).

13 AD disease modules, including 3 AD seed genes and 10 differentially expressed protein (DEP) sets from AD mouse proteomics data, were used to screen FDA-approved drugs for AD. A sankey plot illustrates a global view of 66 drug candidates identified by network proximity. Drugs are grouped by their first-level Anatomical Therapeutic Chemical Classification (ATC) codes.

Extended Data Fig. 5 Longitudinal analyses reveal that sildenafil usage is significantly associated with reduced likelihood of AD in individuals with coronary artery disease (CAD).

Five comparator analyses were conducted for coronary artery disease patients including: (a) sildenafil vs. matched non- sildenafil, (b) sildenafil vs. diltiazem (an anti-hypertensive drug), (c) sildenafil vs. losartan (an anti-hypertensive drug candidate in an AD clinical trial [ClinicalTrials.gov Identifier: NCT02913664]), (d) sildenafil vs. glimepiride (an anti-diabetic drug), and (e) sildenafil vs. metformin (an anti-diabetic drug in an AD clinical trial [ClinicalTrials.gov Identifier: NCT00620191]). For each comparator, we estimated the propensity score by using the variables described in Table 1. Then, we estimated the un-stratified Kaplan-Meier curves, conducted propensity score-stratified (n strata = 10) log-rank test and Cox model. All p-values in the Kaplan Meier plots are based on two-sided log-rank test. Sildenafil n = 19,093; diltiazem n = 51,771; losartan n = 111,592; glimepiride n = 30,083; metformin n = 91,705.

Extended Data Fig. 6 Longitudinal analyses reveal that sildenafil usage is significantly associated with reduced likelihood of AD in individuals with hypertension (HT).

Five comparator analyses were conducted for hypertension patients including: (a) sildenafil vs. matched non- sildenafil, (b) sildenafil vs. diltiazem (an anti-hypertensive drug), (c) sildenafil vs. losartan (an anti-hypertensive drug candidate in an AD clinical trial [ClinicalTrials.gov Identifier: NCT02913664]), (d) sildenafil vs. glimepiride (an anti-diabetic drug), and (e) sildenafil vs. metformin (an anti-diabetic drug in an AD clinical trial [ClinicalTrials.gov Identifier: NCT00620191]). For each comparator, we estimated the propensity score by using the variables described in Table 1. Then, we estimated the un-stratified Kaplan-Meier curves, conducted propensity score-stratified (n strata = 10) log-rank test and Cox model. All p-values in the Kaplan Meier plots are based on two-sided log-rank test. Sildenafil n = 49,541; diltiazem n = 119,097; losartan n = 339,940; glimepiride n = 74,018; metformin n = 275,328.

Extended Data Fig. 7 Longitudinal analyses reveal that sildenafil usage is significantly associated with reduced likelihood of AD in individuals with type-2 diabetes (T2D).

Five comparator analyses were conducted for type-2 diabetes patients including: (a) sildenafil vs. matched non- sildenafil, (b) sildenafil vs. diltiazem (an anti-hypertensive drug), (c) sildenafil vs. losartan (an anti-hypertensive drug candidate in an AD clinical trial [ClinicalTrials.gov Identifier: NCT02913664]), (d) sildenafil vs. glimepiride (an anti-diabetic drug), and (e) sildenafil vs. metformin (an anti-diabetic drug in an AD clinical trial [ClinicalTrials.gov Identifier: NCT00620191]). For each comparator, we estimated the propensity score by using the variables described in Table 1. Then, we estimated the un-stratified Kaplan-Meier curves, conducted propensity score stratified (n strata = 10) log-rank test and Cox model. All p-values in the Kaplan Meier plots are based on two-sided log-rank test. Sildenafil n = 21,978; diltiazem n = 51,300; losartan n = 156,308; glimepiride n = 100,298; metformin n = 367,754.

Supplementary information

Supplementary Information

Supplementary Materials and Methods, Results and Discussion, Figs. 1–9 and Tables 1–15.

Reporting Summary.

Supplementary Tables

Supplementary Table 1. Functional enrichment analysis result for the AD seed gene set (n = 144). Supplementary Table 2. Detailed information of 21 existing drugs in AD clinical trials to test the clinical efficacy of endophenotype hypothesis. Supplementary Table 3. Anti-AD clinical, in vivo and BBB properties from publicly available databases and in silico predictions for the 66 potential AD drugs. Supplementary Table 4. Prioritizing 100 candidate drugs with the lowest z score by AD seed module using the PxEA approach and obtaining 56 candidate drugs. Supplementary Table 5. 120 FDA-approved drugs with reported clinical or preclinical (in vivo) evidence in AD from the AlzGPS database. Supplementary Table 6. Network proximity scores between molecular targets of 12 ongoing repurposable AD drugs (dually targeting amyloid and tau) and AD seed module. Supplementary Table 8. Age-stratified analysis for all comparisons of sildenafil. Supplementary Table 9. Sex-based subgroup analyses for sildenafil. Supplementary Table 13. List of experimentally validated (seed) genes in amyloidosis (amyloid), tauopathy (tau) and AD. Supplementary Table 14. Transcriptomic and proteomic datasets from transgenic AD mouse models.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fang, J., Zhang, P., Zhou, Y. et al. Endophenotype-based in silico network medicine discovery combined with insurance record data mining identifies sildenafil as a candidate drug for Alzheimer’s disease. Nat Aging 1, 1175–1188 (2021). https://doi.org/10.1038/s43587-021-00138-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00138-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing