Skip to main content

Advertisement

Log in

Population PKPD Modeling of BACE1 Inhibitor-Induced Reduction in Aβ Levels In Vivo and Correlation to In Vitro Potency in Primary Cortical Neurons from Mouse and Guinea Pig

  • Research Paper
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

ABSTRACT

Purpose

The aims were to quantify the in vivo time-course between the oral dose, the plasma and brain exposure and the inhibitory effect on Amyloid β (Aβ) in brain and cerebrospinal fluid, and to establish the correlation between in vitro and in vivo potency of novel β-secretase (BACE1) inhibitors.

Methods

BACE1-mediated inhibition of Aβ was quantified in in vivo dose- and/or time-response studies and in vitro in SH-SY5Y cells, N2A cells, and primary cortical neurons (PCN). An indirect response model with inhibition on Aβ production rate was used to estimate unbound in vivo IC 50 in a population pharmacokinetic-pharmacodynamic modeling approach.

Results

Estimated in vivo inhibitory potencies varied between 1 and 1,000 nM. The turnover half-life of Aβ40 in brain was predicted to be 0.5 h in mouse and 1 h in guinea pig. An excellent correlation between PCN and in vivo potency was observed. Moreover, a strong correlation in potency was found between human SH-SY5Y cells and mouse PCN, being 4.5-fold larger in SH-SY5Y cells.

Conclusion

The strong in vivo-in vitro correlation increased the confidence in using human cell lines for screening and optimization of BACE1 inhibitors. This can optimize the design and reduce the number of preclinical in vivo effect studies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

AD:

Alzheimer’s disease

APP:

Amyloid precursor protein

Aβ:

Amyloid β peptide

BACE1:

β-site APP-cleaving enzyme 1

CSF:

Cerebrospinal fluid

CV:

Coefficient of variation

PCN:

Primary cortical neurons

sAPPβ:

Soluble N terminal fragment of APP

REFERENCES

  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(7):939–44.

    Article  CAS  PubMed  Google Scholar 

  2. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362(4):329–44.

    Article  CAS  PubMed  Google Scholar 

  3. Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimers Dement. 2012;8(2):131–68.

    Article  Google Scholar 

  4. Hardy J. The amyloid hypothesis for Alzheimer’s disease: a critical reappraisal. J Neurochem. 2009;110(4):1129–34.

    Article  CAS  PubMed  Google Scholar 

  5. Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011;10(9):698–712.

    Article  CAS  PubMed  Google Scholar 

  6. Wolfe MS. gamma-Secretase inhibitors and modulators for Alzheimer’s disease. J Neurochem. 2012;120 Suppl 1:89–98.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Walsh DM, Teplow DB. Alzheimer’s disease and the amyloid β-protein. Prog Mol Biol Transl Sci. 2012;107:101–24.

    Article  CAS  PubMed  Google Scholar 

  8. Lichtenthaler SF, Haass C, Steiner H. Regulated intramembrane proteolysis–lessons from amyloid precursor protein processing. J Neurochem. 2011;117(5):779–96.

    Article  CAS  PubMed  Google Scholar 

  9. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–6.

    Article  CAS  PubMed  Google Scholar 

  10. Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of Aβ 42(43) and Aβ 40 in senile plaques with end-specific Aβ monoclonals: evidence that an initially deposited species is Aβ 42(43). Neuron. 1994;13(1):45–53.

    Article  CAS  PubMed  Google Scholar 

  11. Younkin SG. Evidence that Aβ 42 is the real culprit in Alzheimer’s disease. Ann Neurol. 1995;37(3):287–8.

    Article  CAS  PubMed  Google Scholar 

  12. Steiner H, Capell A, Leimer U, Haass C. Genes and mechanisms involved in β-amyloid generation and Alzheimer’s disease. Eur Arch Psychiatr Clin Neurosci. 1999;249(6):266–70.

    Article  CAS  Google Scholar 

  13. Citron M. Alzheimer’s disease: strategies for disease modification. Nat Rev Drug Discov. 2010;9(5):387–98.

    Article  CAS  PubMed  Google Scholar 

  14. Panza F, Frisardi V, Imbimbo BP, Capurso C, Logroscino G, Sancarlo D, et al. REVIEW: gamma-Secretase inhibitors for the treatment of Alzheimer’s disease: the current state. CNS Neurosci Ther. 2010;16(5):272–84.

    Article  CAS  PubMed  Google Scholar 

  15. Kreft AF, Martone R, Porte A. Recent advances in the identification of gamma-secretase inhibitors to clinically test the Aβ oligomer hypothesis of Alzheimer’s disease. J Med Chem. 2009;52(20):6169–88.

    Article  CAS  PubMed  Google Scholar 

  16. Niva C, Parkinson J, Olsson F, van Schaick EA, Lundkvist J, Visser SAG. Has inhibition of Aβ production adequately been tested as therapeutic approach in mild AD? A model-based meta-analysis of γ-secretase inhibitor data. Eur J Clin Pharmacol. 2013. doi:10.1007/s00228-012-1459-3.

    PubMed  Google Scholar 

  17. Hussain I, Powell D, Howlett DR, Tew DG, Meek TD, Chapman C, et al. Identification of a novel aspartic protease (Asp 2) as β-secretase. Mol Cell Neurosci. 1999;14(6):419–27.

    Article  CAS  PubMed  Google Scholar 

  18. Sinha S, Anderson JP, Barbour R, Basi GS, Caccavello R, Davis D, et al. Purification and cloning of amyloid precursor protein β-secretase from human brain. Nature. 1999;402(6761):537–40.

    Article  CAS  PubMed  Google Scholar 

  19. Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, et al. β-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286(5440):735–41.

    Article  CAS  PubMed  Google Scholar 

  20. Yan R, Bienkowski MJ, Shuck ME, Miao H, Tory MC, Pauley AM, et al. Membrane-anchored aspartyl protease with Alzheimer’s disease β-secretase activity. Nature. 1999;402(6761):533–7.

    Article  CAS  PubMed  Google Scholar 

  21. Lin X, Koelsch G, Wu S, Downs D, Dashti A, Tang J. Human aspartic protease memapsin 2 cleaves the β-secretase site of β-amyloid precursor protein. Proc Natl Acad Sci U S A. 2000;97(4):1456–60.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Cole SL, Vassar R. BACE1 structure and function in health and Alzheimer’s disease. Curr Alzheimer Res. 2008;5(2):100–20.

    Article  CAS  PubMed  Google Scholar 

  23. Stockley JH, O'Neill C. The proteins BACE1 and BACE2 and β-secretase activity in normal and Alzheimer’s disease brain. Biochem Soc Trans. 2007;35(Pt 3):574–6.

    CAS  PubMed  Google Scholar 

  24. Mullan M, Houlden H, Windelspecht M, Fidani L, Lombardi C, Diaz P, et al. Nat Genet. 1992;2(4):340–2.

    Article  CAS  PubMed  Google Scholar 

  25. Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96–9.

    Article  CAS  PubMed  Google Scholar 

  26. McConlogue L, Buttini M, Anderson JP, Brigham EF, Chen KS, Freedman SB, et al. Partial reduction of BACE1 has dramatic effects on Alzheimer plaque and synaptic pathology in APP Transgenic Mice. J Biol Chem. 2007;282(36):26326–34.

    Article  CAS  PubMed  Google Scholar 

  27. Nishitomi K, Sakaguchi G, Horikoshi Y, Gray AJ, Maeda M, Hirata-Fukae C, et al. BACE1 inhibition reduces endogenous Aβ and alters APP processing in wild-type mice. J Neurochem. 2006;99(6):1555–63.

    Article  CAS  PubMed  Google Scholar 

  28. Swahn BM, Kolmodin K, Karlstrom S, von Berg S, Soderman P, Holenz J, et al. Design and synthesis of β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE1) inhibitors with in vivo brain reduction of β-Amyloid peptides. J Med Chem. 2012;55(21):9346–61.

    Article  CAS  PubMed  Google Scholar 

  29. Jeppsson F, Eketjall S, Janson J, Karlstrom S, Gustavsson S, Olsson LL, et al. Discovery of AZD3839, a potent and selective BACE1 clinical candidate for the treatment of Alzheimers Disease. J Biol Chem. 2012;287(49):41245–57.

    Article  CAS  PubMed  Google Scholar 

  30. Lu Y, Zhang L, Nolan CE, Becker SL, Atchison K, Robshaw AE, et al. Quantitative pharmacokinetic/pharmacodynamic analyses suggest that the 129/SVE mouse is a suitable preclinical pharmacology model for identifying small-molecule gamma-secretase inhibitors. J Pharmacol Exp Ther. 2011;339(3):922–34.

    Article  CAS  PubMed  Google Scholar 

  31. Stachel SJ, Coburn CA, Sankaranarayanan S, Price EA, Wu G, Crouthamel M, et al. Macrocyclic inhibitors of β-secretase: functional activity in an animal model. J Med Chem. 2006;49(21):6147–50.

    Article  CAS  PubMed  Google Scholar 

  32. Stanton MG, Stauffer SR, Gregro AR, Steinbeiser M, Nantermet P, Sankaranarayanan S, et al. Discovery of isonicotinamide derived β-secretase inhibitors: in vivo reduction of β-amyloid. J Med Chem. 2007;50(15):3431–3.

    Article  CAS  PubMed  Google Scholar 

  33. Fukumoto H, Takahashi H, Tarui N, Matsui J, Tomita T, Hirode M, et al. A noncompetitive BACE1 inhibitor TAK-070 ameliorates Aβ pathology and behavioral deficits in a mouse model of Alzheimer’s disease. J Neurosci. 2010;30(33):11157–66.

    Article  CAS  PubMed  Google Scholar 

  34. Chang WP, Huang X, Downs D, Cirrito JR, Koelsch G, Holtzman DM, et al. β-secretase inhibitor GRL-8234 rescues age-related cognitive decline in APP transgenic mice. FASEB J. 2011;25(2):775–84.

    Article  CAS  PubMed  Google Scholar 

  35. Lu Y, Riddell D, Hajos-Korcsok E, Bales K, Wood KM, Nolan CE, et al. CSF Aβ As An Effect Biomarker For Brain Aβ Lowering Verified by Quantitative Preclinical Analyses. J Pharmacol Exp Ther. 2012;342(2):366–75.

    Google Scholar 

  36. Rueeger H, Lueoend R, Rogel O, Rondeau JM, Mobitz H, Machauer R, et al. Discovery of cyclic sulfone hydroxyethylamines as potent and selective β-site APP-cleaving enzyme 1 (BACE1) inhibitors: structure-based design and in vivo reduction of amyloid β-peptides. J Med Chem. 2012;55(7):3364–86.

    Article  CAS  PubMed  Google Scholar 

  37. Wood S, Wen PH, Zhang J, Zhu L, Luo Y, Babu-Khan S, et al. Establishing the relationship between in vitro potency, pharmacokinetic, and pharmacodynamic parameters in a series of orally available, hydroxyethylamine-derived β-secretase inhibitors. J Pharmacol Exp Ther. 2012;343(2):460–7.

    Article  CAS  PubMed  Google Scholar 

  38. Weiss MM, Williamson T, Babu-Khan S, Bartberger MD, Brown J, Chen K, et al. Design and preparation of a potent series of hydroxyethylamine containing β-secretase inhibitors that demonstrate robust reduction of central β-amyloid. J Med Chem. 2012;55(21):9009–24.

    Article  CAS  PubMed  Google Scholar 

  39. Dineen TA, Weiss MM, Williamson T, Acton P, Babu-Khan S, Bartberger MD, et al. Design and synthesis of potent, orally efficacious hydroxyethylamine derived β-site amyloid precursor protein cleaving enzyme (BACE1) inhibitors. J Med Chem. 2012;55(21):9025–44.

    Article  CAS  PubMed  Google Scholar 

  40. Gravenfors Y, Viklund J, Blid J, Ginman T, Karlstrom S, Kihlstrom J, et al. New Aminoimidazoles as β-Secretase (BACE-1) inhibitors showing amyloid-β (Aβ) lowering in Brain. J Med Chem. 2012;55(21):9297–311.

    Article  CAS  PubMed  Google Scholar 

  41. Sankaranarayanan S, Holahan MA, Colussi D, Crouthamel MC, Devanarayan V, Ellis J, et al. First demonstration of cerebrospinal fluid and plasma Aβ lowering with oral administration of a β-site amyloid precursor protein-cleaving enzyme 1 inhibitor in nonhuman primates. J Pharmacol Exp Ther. 2009;328(1):131–40.

    Article  CAS  PubMed  Google Scholar 

  42. May PC, Dean RA, Lowe SL, Martenyi F, Sheehan SM, Boggs LN, et al. Robust central reduction of amyloid-β in humans with an orally available, non-peptidic β-secretase inhibitor. J Neurosci. 2011;31(46):16507–16.

    Article  CAS  PubMed  Google Scholar 

  43. Swahn BM, Holenz J, Kihlstrom J, Kolmodin K, Lindstrom J, Plobeck N, et al. Aminoimidazoles as BACE-1 inhibitors: the challenge to achieve in vivo brain efficacy. Bioorg Med Chem Lett. 2012;22(5):1854–9.

    Article  CAS  PubMed  Google Scholar 

  44. Bueters T, Dahlstrom J, Kvalvagnaes K, Betner I, Briem S. High-throughput analysis of standardized pharmacokinetic studies in the rat using sample pooling and UPLC-MS/MS. J Pharm Biomed Anal. 2011;55(5):1120–6.

    Article  CAS  PubMed  Google Scholar 

  45. Heisey SR. Brain and choroid plexus blood volumes in vertebrates. Comp Biochem Physiol. 1968;26(2):489–98.

    Article  CAS  PubMed  Google Scholar 

  46. Park S, Sinko PJ. P-glycoprotein and mutlidrug resistance-associated proteins limit the brain uptake of saquinavir in mice. J Pharmacol Exp Ther. 2005;312(3):1249–56.

    Article  CAS  PubMed  Google Scholar 

  47. Borgegård T, Minidis A, Jureus A, Malmborg J, Rosqvist S, Gruber, S., Almqvist, H., et al. In vivo analysis using a presenilin-1-specific inhibitor: Presenilin 1- containing g-secretase complexes mediate the majority of CNS Ab production in the mouse. 3 2011;1:30–45.

  48. Friden M, Ducrozet F, Middleton B, Antonsson M, Bredberg U, Hammarlund-Udenaes M. Development of a high-throughput brain slice method for studying drug distribution in the central nervous system. Drug Metab Dispos. 2009;37(6):1226–33.

    Article  CAS  PubMed  Google Scholar 

  49. Dayneka NL, Garg V, Jusko WJ. Comparison of four basic models of indirect pharmacodynamic responses. J Pharmacokinet Biopharm. 1993;21(4):457–78.

    Article  CAS  PubMed  Google Scholar 

  50. Asai M, Hattori C, Iwata N, Saido TC, Sasagawa N, Szabo B, et al. The novel β-secretase inhibitor KMI-429 reduces amyloid β peptide production in amyloid precursor protein transgenic and wild-type mice. J Neurochem. 2006;96(2):533–40.

    Article  CAS  PubMed  Google Scholar 

  51. Liu L, Duff K. A technique for serial collection of cerebrospinal fluid from the cisterna magna in mouse. J Vis Exp 2008 Nov 10;(21).

  52. Truong AP, Toth G, Probst GD, Sealy JM, Bowers S, Wone DW, et al. Design of an orally efficacious hydroxyethylamine (HEA) BACE-1 inhibitor in a preclinical animal model. Bioorg Med Chem Lett. 2010;20(21):6231–6.

    Article  CAS  PubMed  Google Scholar 

  53. Lu Y. Integrating experimentation and quantitative modeling to enhance discovery of Beta amyloid lowering therapeutics for Alzheimer’s disease. Front Pharmacol. 2012;3(177):1–6.

    Google Scholar 

  54. Danhof M, Van der Graaf PH, Jonker DM, Visser SAG, Zuideveld KP. Mechanism-based pharmacokinetic-pharmacodynamic modeling for the prediction of in vivo drug concentration-effect relationships -application in drug candidate selection and lead optimization. 2007. Comprehensive Medicinal Chemistry (2nd Ed.) Volume 5: “ADME-Tox: The Fate of Drugs in the Body”. Editors Bernard Testa and Han van de Waterbeemd.

  55. Bueters T, Ploeger BA, Visser SA. The virtue of translational PKPD modeling in drug discovery: selecting the right clinical candidate while sparing animal lives. Drug Discov Today. 2013. doi:10.1016/j.drudis.2013.05.001.

    PubMed  Google Scholar 

  56. Shimmyo Y, Kihara T, Akaike A, Niidome T, Sugimoto H. Flavonols and flavones as BACE-1 inhibitors: structure-activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim Biophys Acta. 2008;1780(5):819–25.

    Article  CAS  PubMed  Google Scholar 

  57. Beck M, Bruckner MK, Holzer M, Kaap S, Pannicke T, Arendt T, et al. Guinea-pig primary cell cultures provide a model to study expression and amyloidogenic processing of endogenous amyloid precursor protein. Neuroscience. 2000;95(1):243–54.

    Article  CAS  PubMed  Google Scholar 

  58. Yang HC, Chai X, Mosior M, Kohn W, Boggs LN, Erickson JA, et al. Biochemical and kinetic characterization of BACE1: investigation into the putative species-specificity for β- and β’-cleavage sites by human and murine BACE1. J Neurochem. 2004;91(6):1249–59.

    Article  CAS  PubMed  Google Scholar 

  59. Vandermeeren M, Geraerts M, Pype S, Dillen L, Van Hove C, Mercken M. The functional gamma-secretase inhibitor prevents production of amyloid β 1–34 in human and murine cell lines. Neurosci Lett. 2001;315(3):145–8.

    Article  CAS  PubMed  Google Scholar 

  60. Gabrielsson J, Fjellstrom O, Ulander J, Rowley M, Van Der Graaf PH. Pharmacodynamic-pharmacokinetic integration as a guide to medicinal chemistry. Curr Top Med Chem. 2011;11(4):404–18.

    Article  CAS  PubMed  Google Scholar 

Download references

ACKNOWLEDGMENTS AND DISCLOSURES

The authors would like to thank Eva Spennare for determining the fraction unbound in brain; Jenny Johansson for measuring the plasma protein binding; Hongmei Yan for collecting the data into the data-analysis sheet; Sveinn Briem and his team for performing the bioanalysis; Elin Lundkvist and Fredrik Olsson for supporting the in vitro experiments; and Kristina Eliason for supporting the in vivo experiments.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juliette Janson.

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

Supplement 1

(PDF 71 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Janson, J., Eketjäll, S., Tunblad, K. et al. Population PKPD Modeling of BACE1 Inhibitor-Induced Reduction in Aβ Levels In Vivo and Correlation to In Vitro Potency in Primary Cortical Neurons from Mouse and Guinea Pig. Pharm Res 31, 670–683 (2014). https://doi.org/10.1007/s11095-013-1189-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11095-013-1189-y

KEY WORDS

Navigation