Pharmacokinetic and Pharmacodynamic Effects of a γ-Secretase Modulator, PF-06648671, on CSF Amyloid-β Peptides in Randomized Phase I Studies

Jae Eun Ahn, Charles Carrieri, Fernando Dela Cruz, Terence Fullerton, Eva Hajos-Korcsok, Ping He, Constantino Kantaridis, Claire Leurent, Richann Liu, Jessica Mancuso, Laure Mendes da Costa, Ruolun Qiu, Jae Eun Ahn, Charles Carrieri, Fernando Dela Cruz, Terence Fullerton, Eva Hajos-Korcsok, Ping He, Constantino Kantaridis, Claire Leurent, Richann Liu, Jessica Mancuso, Laure Mendes da Costa, Ruolun Qiu

Abstract

γ-Secretase modulators (GSMs) represent a promising therapy for Alzheimer's disease by reducing pathogenic amyloid-β (Aβ) peptide production. Three phase I studies (NCT02316756, NCT02407353, and NCT02440100) investigated the safety/tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of the oral GSM, PF-06648671. A PK/PD indirect-response model was developed (using biomarker data) to simultaneously characterize differential effects of PF-06648671 on multiple Aβ species in cerebrospinal fluid (CSF). Healthy subjects (n = 120) received single doses or multiple-ascending doses of PF-06648671/placebo for 14 days. No serious adverse events occurred; severe adverse eventswere deemed not drug related. PF-06648671 decreased Aβ42 and Aβ40 concentrations in CSF, with greater effects on Aβ42, and increased Aβ37 and Aβ38 levels, particularly Aβ37. No significant change in total Aβ was observed. The PK/PD model well described the tendency of observed CSF Aβ data and the steady-state effects of PF-06648671, supporting its use for predicting central Aβ effects and optimal dose selection for GSMs in future trials.

Conflict of interest statement

J.E.A., C.C., F.D.C., T.F., C.K., J.M., L.MdC., and R.Q. are employees of, and hold stock options for, Pfizer Inc. P.H., E.H.‐K., C.L., and R.L. were employees of, and held stock options for, Pfizer Inc at the time of the research.

© 2019 Pfizer Inc. Clinical Pharmacology & Therapeutics published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
PK and PD plots from study B7991003 (serial CSF sampling biomarker study). (a) Plasma concentration of PF‐06648671 over time. (b–f) CSF concentration over time for (b) Aβ42, (c) Aβ40, (d) Aβ38, (e) Aβ37, and (f) total Aβ. Mean profiles are shown as bold lines, profiles from individual subjects are shown as thinner lines. Aβ, amyloid‐β; CSF, cerebrospinal fluid; PD, pharmacodynamic; PK, pharmacokinetic.
Figure 2
Figure 2
PK and PD plots from study B7991002 at days 0 and 14 (multiple‐ascending dose). (a) Plasma concentration of PF‐06648671 by dose. (b–f) CSF concentration by dose for (b) Aβ42, (c) Aβ40, (d) Aβ38, (e) Aβ37, and (f) total Aβ. Box plot shows median (bold horizontal line) as well as quartiles (25% and 75%), with whiskers to the last data point within 1.5 times the IQR. Black dots represent the outlier values (values outside 1.5 times the IQR above the 75% quartile and below the 25% quartile). Aβ, amyloid‐β; CSF, cerebrospinal fluid; IQR, interquartile range; PD, pharmacodynamic; PK, pharmacokinetic.
Figure 3
Figure 3
PK/PD model schematic. γ, sigmoidicity constant; Aβ, amyloid‐β; Conc, plasma concentration of drug; CSF, cerebrospinal fluid; EC 50, plasma concentration at half Emax; Emax, maximum response achievable; IC 50, plasma concentration at half Imax; Imax, maximum inhibition achievable; kin, zero‐order rate of Aβ production; kout, first‐order turnover constant of Aβ; PD, pharmacodynamic; PK, pharmacokinetic.
Figure 4
Figure 4
Visual predictive check for multiple‐dose data on day 14 of B7991002. (a) Aβ42. (b) Aβ40. (c) Aβ38. (d) Aβ37. Circles represent observed data; gray solid and dashed lines represent observed median and 90% percentiles; black lines represent the corresponding simulation percentiles. Baseline was not presented for better visualization of dose response. Aβ, amyloid‐β; CSF, cerebrospinal fluid.
Figure 5
Figure 5
Predicted average steady‐state effects of PF‐06648671 on CSF samples. (a) Aβ42 percent change from baseline. (b) Aβ40 percent change from baseline. (c) Aβ37 percent change from baseline. (d) Ratio of Aβ42:Aβ40. (e) Ratio of Aβ42:Aβ38.* (f) Ratio of Aβ42:Aβ37. Dotted line represents the median, with the shaded area representing the 95% CI. *95% CI for ratio of Aβ42:Aβ38 prediction was truncated due to missing values. Aβ, amyloid‐β; CI, confidence interval; CSF, cerebrospinal fluid.

References

    1. World Health Organization . Dementia: key facts <> (2017). Accessed May 4, 2018.
    1. Hort, J. et al EFNS guidelines for the diagnosis and management of Alzheimer's disease. Eur. J. Neurol. 17, 1236–1248 (2010).
    1. Glenner, G.G. & Wong, C.W. Alzheimer's disease and Down's syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem. Biophys. Res. Commun. 122, 1131–1135 (1984).
    1. Hardy, J. & Allsop, D. Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends Pharmacol. Sci. 12, 383–388 (1991).
    1. Lee, V.M. , Balin, B.J. , Otvos, L. Jr & Trojanowski, J.Q. A68: a major subunit of paired helical filaments and derivatized forms of normal Tau. Science 251, 675–678 (1991).
    1. Wiltfang, J. et al Highly conserved and disease‐specific patterns of carboxyterminally truncated Abeta peptides 1‐37/38/39 in addition to 1‐40/42 in Alzheimer's disease and in patients with chronic neuroinflammation. J. Neurochem. 81, 481–496 (2002).
    1. Bursavich, M.G. , Harrison, B.A. & Blain, J.F. Gamma secretase modulators: new Alzheimer's drugs on the horizon? J. Med. Chem. 59, 7389–7409 (2016).
    1. Burdick, D. et al Assembly and aggregation properties of synthetic Alzheimer's A4/beta amyloid peptide analogs. J. Biol. Chem. 267, 546–554 (1992).
    1. Roher, A.E. et al Structural alterations in the peptide backbone of beta‐amyloid core protein may account for its deposition and stability in Alzheimer's disease. J. Biol. Chem. 268, 3072–3083 (1993).
    1. Velasco, P.T. et al Synapse‐binding subpopulations of Abeta oligomers sensitive to peptide assembly blockers and scFv antibodies. ACS Chem. Neurosci. 3, 972–981 (2012).
    1. Lambert, M.P. et al Diffusible, nonfibrillar ligands derived from Abeta1‐42 are potent central nervous system neurotoxins. Proc. Natl. Acad. Sci. USA 95, 6448–6453 (1998).
    1. Crump, C.J. , Johnson, D.S. & Li, Y.M. Development and mechanism of gamma‐secretase modulators for Alzheimer's disease. Biochemistry 52, 3197–3216 (2013).
    1. Kumar‐Singh, S. et al Nonfibrillar diffuse amyloid deposition due to a gamma(42)‐secretase site mutation points to an essential role for N‐truncated A beta(42) in Alzheimer's disease. Hum. Mol. Genet. 9, 2589–2598 (2000).
    1. Janelidze, S. et al CSF Aβ42/Aβ40 and Aβ42/Aβ38 ratios: better diagnostic markers of Alzheimer disease. Ann. Clin. Transl. Neurol. 3, 154–165 (2016).
    1. Baldeiras, I. et al Addition of the Aβ42/40 ratio to the cerebrospinal fluid biomarker profile increases the predictive value for underlying Alzheimer's disease dementia in mild cognitive impairment. Alzheimers Res. Ther. 10, 33 (2018).
    1. Dovey, H.F. et al Functional gamma‐secretase inhibitors reduce beta‐amyloid peptide levels in brain. J. Neurochem. 76, 173–181 (2001).
    1. Bateman, R.J. et al A gamma‐secretase inhibitor decreases amyloid‐beta production in the central nervous system. Ann. Neurol. 66, 48–54 (2009).
    1. Doody, R.S. et al A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N. Engl. J. Med. 369, 341–350 (2013).
    1. Barten, D.M. , Meredith, J.E. Jr , Zaczek, R. , Houston, J.G. & Albright, C.F. γ‐Secretase inhibitors for Alzheimer's disease. Drugs R. D. 7, 87–97 (2006).
    1. Tamayev, R. & D'Adamio, L. Inhibition of γ‐secretase worsens memory deficits in a genetically congruous mouse model of Danish dementia. Mol. Neurodegener. 7, 19 (2012).
    1. Searfoss, G.H. et al Adipsin, a biomarker of gastrointestinal toxicity mediated by a functional gamma‐secretase inhibitor. J. Biol. Chem. 278, 46107–46116 (2003).
    1. Wong, G.T. et al Chronic treatment with the γ‐secretase inhibitor LY‐411,575 inhibits β‐amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J. Biol. Chem. 279, 12876–12882 (2004).
    1. Coric, V. et al Safety and tolerability of the gamma‐secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch. Neurol. 69, 1430–1440 (2012).
    1. Probst, G. et al Discovery of (R)‐4‐cyclopropyl‐7,8‐difluoro‐5‐(4‐(trifluoromethyl)phenylsulfonyl)‐4,5‐dihydro‐1H‐pyrazolo[4,3‐c]quinoline (ELND006) and (R)‐4‐cyclopropyl‐8‐fluoro‐5‐(6‐(trifluoromethyl)pyridin‐3‐ylsulfonyl)‐4,5‐dihydro‐2H‐pyrazolo[4,3‐c]quinoline (ELND007): metabolically stable γ‐secretase inhibitors that selectively inhibit the production of amyloid‐β over Notch. J. Med. Chem. 56, 5261–5274 (2013).
    1. Weissmiller, A.M. et al A gamma‐secretase inhibitor, but not a gamma‐secretase modulator, induced defects in BDNF axonal trafficking and signaling: evidence for a role for APP. PLoS One 10, e0118379 (2015).
    1. Kounnas, M.Z. , Lane‐Donovan, C. , Nowakowski, D.W. , Herz, J. & Comer, W.T. NGP 555, a γ‐secretase modulator, lowers the amyloid biomarker, Aβ42, in cerebrospinal fluid while preventing Alzheimer's disease cognitive decline in rodents. Alzheimers Dement. (N. Y.) 3, 65–73 (2017).
    1. Page, R.M. et al Generation of Aβ38 and Aβ42 is independently and differentially affected by familial Alzheimer disease‐associated presenilin mutations and gamma‐secretase modulation. J. Biol. Chem. 283, 677–683 (2008).
    1. Ohki, Y. et al Phenylpiperidine‐type γ‐secretase modulators target the transmembrane domain 1 of presenilin 1. EMBO J. 30, 4815–4824 (2011).
    1. Pozdnyakov, N. et al γ‐Secretase modulator (GSM) photoaffinity probes reveal distinct allosteric binding sites on presenilin. J. Biol. Chem. 288, 9710–9720 (2013).
    1. Blain, J.F. et al Characterization of FRM‐36143 as a new gamma‐secretase modulator for the potential treatment of familial Alzheimer's disease. Alzheimers Res. Ther. 8, 34 (2016).
    1. Wolfe, M.S. gamma‐Secretase inhibitors and modulators for Alzheimer's disease. J. Neurochem. 120(suppl. 1), 89–98 (2012).
    1. Soares, H.D. et al The γ‐secretase modulator, BMS‐932481, modulates Aβ peptides in the plasma and cerebrospinal fluid of healthy volunteers. J. Pharmacol. Exp. Ther. 358, 138–150 (2016).
    1. Toyn, J.H. et al Robust translation of gamma‐secretase modulator pharmacology across preclinical species and human subjects. J. Pharmacol. Exp. Ther. 358, 125–137 (2016).
    1. Wagner, S.L. et al Pharmacological and toxicological properties of the potent oral gamma‐secretase modulator BPN‐15606. J. Pharmacol. Exp. Ther. 362, 31–44 (2017).
    1. Yu, Y. et al Safety, tolerability, pharmacokinetics, and pharmacodynamics of the novel gamma‐secretase modulator, E2212, in healthy human subjects. J. Clin. Pharmacol. 54, 528–536 (2014).
    1. Hajos‐Korcsok, E. et al Preclinical pharmacology and toxicology of PF‐06648671, a novel gamma secretase modulator: comparison to gamma secretase inhibitors. Neurodegener. Dis. 17(suppl. 1), 86 (2017) (abstract ADPD7‐0759).
    1. Li, T. et al γ‐secretase modulators do not induce Aβ‐rebound and accumulation of beta‐C‐terminal fragment. J. Neurochem. 121, 277–286 (2012).
    1. Bateman, R.J. , Wen, G. , Morris, J.C. & Holtzman, D.M. Fluctuations of CSF amyloid‐beta levels: implications for a diagnostic and therapeutic biomarker. Neurology 68, 666–669 (2007).
    1. Li, J. et al Effect of human cerebrospinal fluid sampling frequency on amyloid‐β levels. Alzheimers Dement. 8, 295–303 (2012).
    1. Zhang, L. , Beal, S.L. & Sheiner, L.B. Simultaneous vs. sequential analysis for population PK/PD data I: best‐case performance. J. Pharmacokinet. Pharmacodyn. 30, 387–404 (2003).

Source: PubMed

3
Subscribe