You are on page 1of 14

Original Manuscript

Clinical Pharmacology
Pharmacokinetics, Safety, Tolerability, in Drug Development
2018, 00(0) 1–14
and Pharmacodynamics of Alicapistat, C 2018, The American College of

Clinical Pharmacology
DOI: 10.1002/cpdd.598
a Selective Inhibitor of Human Calpains
1 and 2 for the Treatment of Alzheimer
Disease: An Overview of Phase 1
Studies

Hoi-Kei Lon, Nuno Mendonca, Sandra Goss, Ahmed A. Othman, Charles Locke,
Ziyi Jin, and Beatrice Rendenbach-Mueller

Abstract
Alicapistat is an orally active selective inhibitor of calpain 1 and 2 whose overactivation has been linked to Alzheimer
disease (AD). Three studies were conducted in healthy subjects (18-55 years), 1 in healthy elderly subjects (65 years),
and 1 in patients with mild to moderate AD. Four studies assessed pharmacokinetics, 1 study in healthy subjects assessed
pharmacodynamics (sleep parameters, particularly rapid eye movement [REM], as a measure of central nervous system
[CNS] penetration and activity), and all studies assessed safety. Participants received single doses or multiple twice-daily
doses of alicapistat for up to 14 days. Maximum alicapistat plasma concentrations were reached in 2 to 5 hours; half-life
was 7 to 12 hours postdose. Alicapistat exposure was dose proportional in the alicapistat 50- to 1000-mg dose range.
Exposure of the alicapistat R,S diastereomer was approximately 2-fold greater than exposure of the R,R diastereomer in
healthy young and elderly subjects and patients with AD. Alicapistat at 400- or 800-mg twice-daily doses had no effect on
REM sleep parameters, whereas the active control, donepezil at 10 mg twice daily, affected sleep parameters. Across all
trials, the incidence of treatment-emergent adverse events was similar in the placebo and alicapistat groups. There were
no clinically significant changes in vital signs and laboratory measurements. The lack of an effect of alicapistat on sleep
suggests that concentrations in the CNS were inadequate or that preclinical studies do not predict alicapistat effects in
humans.

Keywords
calpain inhibitor, pharmacokinetics, pharmacodynamics, biopharmaceutics, neuroscience, clinical trials

Alzheimer disease (AD) is the most common form of The calpains are a family of proteases constitu-
dementia1 ; with the aging of the population, AD will af- tively expressed in an inactive form, some of which
fect an estimated 14 million people in the United States are activated by Ca2+ .9 Calpain 1 and 2 are Ca2+ -
by 2050.2 The main pathologic features of AD include activated calpains and members of a subfamily of
the accumulation of amyloid plaques, neurofibrillary cysteine proteinases. Calpain 1 and 2 differ in Ca2+
tangles, and diffuse loss of neurons and synapses.3,4 The
pathogenesis of AD is unclear, but many hypotheses AbbVie Inc., North Chicago, IL, USA
have been proposed.1,4
Submitted for publication 18 January 2018; accepted 28 May 2018.
Currently approved treatment options include
acetylcholinesterase inhibitors (AChE-I’s) and the N- Corresponding Author:
Beatrice Rendenbach-Mueller, PhD, Senior Project Leader, Neuroscience
methyl-D-aspartate receptor antagonist memantine,5 Development, AbbVie Deutschland GmbH & Co. KG, Knollstrasse,
both of which have modest symptomatic effects.6–8 Building 34, Room 205, 67061 Ludwigshafen, Germany
Therefore, efficacious treatment of AD, in particular (e-mail: beatrice.rendenbach-mueller@abbvie.com)
drugs that can halt or delay progression, is an unmet Ahmed A. Othman is a fellow of the American College of Clinical
medical need.8 Pharmacology.
2 Clinical Pharmacology in Drug Development 2018, 00(0)

hippocampal acetylcholine (ACh) release, (age-related)


cognitive decline in rodents and monkeys, social recog-
O
nition in rodents, and rapid eye movement (REM) sleep
HN (R,S) N parameters, similar to effects observed with the AChE-
H
(R) O I donepezil. In short, alicapistat dose-dependently in-
O creased ACh levels in the medial prefrontal cortex and
N hippocampus, as well as glutamate levels in the hip-
O
pocampus of rats, as assessed by microdialysis. The
same doses resulted in significant enhancement of
REM sleep time and the number of REM episodes.
Figure 1. Chemical structure of alicapistat. The effects on both ACh release and REM sleep were
similar to those of donepezil. In addition to its effects
sensitivities required for in vitro activation, have highly on biochemical and sleep parameters, alicapistat sig-
conserved amino acid sequences, and are expressed nificantly improved cognitive deficits, as seen in rodent
ubiquitously, including in the central nervous system assays such as the social recognition task and the T-
(CNS).9,10 Overactivation of calpain 1 and 2 has been maze alternation task, as well as in the delayed-match-
linked to neurodegenerative diseases including AD.10,11 to-sample task in aged rhesus monkeys. In addition to
In animal models of AD, inhibition of calpain 1 the effects on cognition, several experiments showed a
and 2 restores normal synaptic function and cognitive blockade of N-methyl-D-aspartate (NMDA)–induced
behavior and decreases amyloid plaques.12,13 neurotoxicity in hippocampal slices and an attenuation
Alicapistat (ABT-957), an orally active selective of in vivo NMDA-induced cholinergic neurodegenera-
small-molecule inhibitor of human calpain 1 and tion after treatment with alicapistat. These findings sup-
porcine calpain 2 that has high selectivity for calpain port the hypothesis that selective calpain inhibition by
1 and 2 versus other cysteine proteases such as cathep- alicapistat may have the potential for both symptomatic
sins B, K, L, and S, was being investigated for the effects and attenuation of the neurodegeneration occur-
treatment of AD in a program that has since been ter- ring with AD. In the aforementioned multiple animal
minated. Alicapistat is a 1:1 mixture of 2 interconvert- models, efficacy was achieved at mean unbound alicapi-
ible diastereomers (alicapistat R,S and alicapistat R,R stat plasma concentrations of 9 to 48 ng/mL and a geo-
diastereomers) with a molecular weight of 434 g/mol metric mean of 13 ng/mL, corresponding to mean total
(Figure 1). In preclinical studies, alicapistat exhibited plasma concentrations of 290 to 690 ng/mL and a ge-
moderate permeability in human Caco-2 cells and was ometric mean of 418 ng/mL. Assuming that the most
a substrate for efflux via P-glycoprotein but not breast conservative preclinical value for the CSF/plasma con-
cancer resistance protein in Madin-Darby canine kid- centration (0.25) would apply to humans, it was esti-
ney II cells (unpublished observations). In vitro, the mated that an efficacious concentration of alicapistat in
fraction of alicapistat binding to human plasma pro- the CSF would be approximately 4 ng/mL (unpublished
tein was approximately 0.3; the fraction bound to observations).
α1-acid glycoprotein was approximately 0.4. Alicapis- This report summarizes the results of 5 phase 1 stud-
tat was metabolized in vitro primarily by cytochrome ies conducted to assess the pharmacokinetics, safety,
P450 (CYP) 3A4 (94%) and, to a far lesser extent, tolerability, and pharmacodynamics (ie, effect on sleep
by CYP2C198, CYP2D6, and CYP3A5; therefore, it parameters) of alicapistat in healthy subjects and in
was expected that strong inhibitors of CYP3A4 would patients with mild to moderate AD.
raise exposure to alicapistat. Alicapistat was stable
in liver microsomes and hepatocytes across species
(mouse, rat, monkey, dog, and human) and demon- Methods
strated low liability to cytosolic carbonyl reductase ac- Study Designs
tivities, with low liver cytosol intrinsic clearance (0.005 Studies 1, 2, and 3 were single-center, randomized,
and 0.07 L/h/kg in monkeys and humans, respectively). double-blind, placebo-controlled trials in healthy men
In preclinical animal models (rats, dogs, and monkeys), and women designed primarily to test the pharma-
alicapistat penetrated the CNS with cerebrospinal fluid cokinetics, safety, and tolerability of single-escalating,
(CSF)/plasma concentration ratios ranging from ap- multiple-escalating, or multiple fixed oral doses of al-
proximately 0.25 to 0.60 across species (unpublished icapistat (Table 1). Tested doses ranged from 50 to
observations). 1000 mg. Study 1 also included an open-label, 3-period,
Alicapistat, like other selective calpain inhibitors, randomized crossover substudy to test the effects of
has demonstrated efficacy in a broad range of an- food and ketoconazole (400 mg once daily) on the pro-
imal models relevant to AD, including cortical and file of single doses of alicapistat 150 mg. Study 2 also
Lon et al 3

Table 1. Phase 1 Studies of Alicapistat

Pharmacokinetics Blood and CSF Sampling;


Study Participants Study Design Sample Size Determination

1 Healthy men and women aged Single-center study conducted at CPPM Part 1: blood before dosing and 0.5, 1, 1.5, 2,
18-55 years Research Unit, Grayslake, Illinois, between 3, 4, 6, 9, 12, 16, 24, 36, 48, 60, and 72 hours
n = 59 March 2012 and August 2012 (IRB: Vista postdose.
Health System, Waukegan, Illinois). Part 2: blood before dosing and 0.5, 1, 1.5, 2,
Part 1: randomized, double-blind, 3, 4, 6, 9, 12, 16, 24, 36, 48, 60, 72, and 96
placebo-controlled, single-dose study of hours postdose.
alicapistat (50, 100, 200, 400, 700, and Determination of sample size for
1000 mg). Six serial groups with up to 8 tolerability assessment in part 1: the
subjects/group (6 subjects received a probability that a given AE would not be
single dose of alicapistat [5 subjects in the observed in a group of 6 subjects was
50-mg group], and 2 subjects received a 0.004 for an AE with a true population
single dose of placebo). incidence rate of 0.5.
Part 2: randomized, 3-period, open-label
crossover study of single doses of
alicapistat 150 mg (n = 12). In periods 1
and 2, subjects received alicapistat under
fasting or nonfasting conditions,
respectively. In period 3, subjects received
400 mg of ketoconazole once daily under
fasting conditions.
2 Groups 1-4: healthy men and Single-center study conducted at Groups 1 and 2: blood before the morning
women aged 18-55 years Northwestern Lake Forest Hospital, dose on day 1 and 0.5, 1, 1.5, 2, 3, 4, 6, 9,
Group 5: healthy men and Grayslake, Illinois, between June 2013 and and 12 hours (before the evening dose),
women aged  65 years January 2014 (IRB: Vista Health System, before the morning dose on days 3, 5, and
n = 55 Waukegan, Illinois). 6, and before the morning dose on day 7
Groups 1-4: randomized, double-blind, and after 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12 (before
placebo-controlled, multiple-dose study the evening dose), 12.5, 13, 13.5, 14, 15, 16,
(each group: alicapistat, n = 9; placebo, 18, 21, 24, 36, 48, 60, and 72 hours.
n = 3).
r Group 1: alicapistat 100 mg or Groups 3 and 4: blood before the morning
dose on day 1 and 0.5, 1, 1.5, 2, 3, 4, 6, 9,
placebo twice daily for 7 days
r Group 2: alicapistat 200 mg or and 12 hours (before the evening dose),
before the morning dose on days 3, 5, and
placebo twice daily for 7 days
r Group 3: alicapistat 400 mg or 6, before the morning dose on day 7, and
after 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12 (before the
placebo twice daily for 14 days
r Group 4: alicapistat 800 mg or evening dose), 12.5, 13, 13.5, 14, 15, 16, 18,
21, 24, 36, 48, 60, and 72 hours, before the
placebo twice daily for 14 days
r Group 5: open-label single-dose morning dose on days 10, 12, and 13, and
before the morning dose on day 14 and
study of alicapistat 400 mg (n = 7) after 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12 (before the
evening dose), 12.5, 13, 13.5, 14, 15, 16, 18,
21, 24, 36, 48, 60, and 72 hours.
Group 5: blood before the dose on day 1
and after 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12, 24, 36,
48, 60, and 72 hours.
Determination of sample size for
tolerability assessment: the probability
that a given AE would not be observed in
a group of 8 or 9 subjects was 0.004 and
0.002, respectively, for an AE with a true
population incidence rate of 0.5.

(Continued)
4 Clinical Pharmacology in Drug Development 2018, 00(0)

Table 1. Continued

Pharmacokinetics Blood and CSF Sampling;


Study Participants Study Design Sample Size Determination

Determination of sample size for the


comparison of single-dose AUC in the
elderly with that in the nonelderly in
study 1: it was anticipated that there
would be data for 8 elderly subjects in this
study and 6 nonelderly subjects at the
same dose in study 1. For these sample
sizes, the calculated power for a test at a
significance level of .05 on the difference
between the elderly and nonelderly with
respect to AUC is 0.895 if the ratio of
AUC central values is 2.00. The
corresponding power for Cmax is 0.74.
3 Healthy men and women aged Single-center study conducted at Orlando Blood before the morning dose on day 1
 65 years Clinical Research Center, Orlando, Florida, and at 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours
n = 24 between May 2014 and July 2014 (IRB: (before the evening dose), before the
Aspire IRB, Santee, California). morning dose on days 3, 5, and 6, before
Randomized, double-blind, the morning dose on day 7 and after 0.5,
r Group 1: alicapistat 200 mg (n =
placebo-controlled, multiple-dose study: 1, 1.5, 2, 3, 4, 6, 9, 12 (before the evening
dose), 12.5, 13, 13.5, 14, 15, 16, 18, 21, and
8) or placebo (n = 4) twice daily 24 hours, before the morning dose on
for 14 days
r Group 2: alicapistat 550 mg (n = days 10, 12, and 13, and before the
morning dose on day 14 and after 0.5, 1,
8) or placebo (n = 4) twice daily 1.5, 2, 3, 4, 6, 9, 12 (before the evening
for 14 days dose), 12.5, 13, 13.5, 14, 15, 16, 18, 21, 24,
36, 48, 60, and 72 hours.
CSF: approximately 12 hours after the last
dose of alicapistat on day 14.
Determination of sample size for
tolerability assessment: the probability
that a given AE would not be observed in
a group of 8 subjects was 0.004 for an AE
with a true population incidence rate of
0.5.
4 Men and women aged Single-center study conducted by Compass Blood before the morning dose on day 1
55-90 years meeting Research, LLC, Orlando, Florida, between and at 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 9, and 12
NINCDS/ADRDA criteria September 2014 and March 2016 (IRBs, hours (before the evening dose), before
for probable AD and on separate for each investigator: Schulman the morning dose on days 5 and 6, before
stable dose of AChE-I Associates IRB, Cincinnati, Ohio; MidLands the morning dose on day 7 and at 0.5,
n = 17 Independent Review Board, Overland 0.75, 1, 1.5, 2, 3, 4, 6, 9, 12 (before the
Park, Kansas; New England IRB, Newtown, evening dose), 12.5, 12.75, 13, 13.5, 14, 15,
Massachusetts; IntegReview IRB, Austin, 16, 18, 21, 24, 36, 48, 60, and 72 hours.
Texas). Determination of sample size for
Randomized, double-blind, tolerability assessment: the probability
r Group 1: alicapistat 200 mg (n =
placebo-controlled, multiple-dose study: that a given AE would not be observed in
a group of 7 patients was 0.008 for an AE
9) or placebo (n = 3) twice daily with a true population incidence rate of
for 7 days
r Group 2: alicapistat 550 mg (n = 0.5.

3) or placebo (n = 2) twice daily


for 7 days

(Continued)
Lon et al 5

Table 1. Continued

Pharmacokinetics Blood and CSF Sampling;


Study Participants Study Design Sample Size Determination

5 Healthy men aged 25-45 years Single-center study conducted by Clinilabs, Blood approximately 1.75 hours after the
with regular sleep habits Inc., New York, New York, between May evening dose and at the end of
n = 17 2015 and September 2015 (IRB: New polysomnography.
England IRB, Newtown, Massachusetts). Determination of sample size: it was
Randomized, single-blind, placebo- and estimated that 16 subjects would allow
active-controlled, 4-period crossover detection of a difference of 3%, 4%, and
study with 4 groups (n = 4 each, but 1 5% versus placebo (power of 0.816, 0.968,
r Regimen A: alicapistat 400 mg
subject was replaced by another): and 0.997, respectively, assuming an SD of
2.9%) in REM as a percentage of total
twice daily
r Regimen B: alicapistat 800 mg sleep time and a difference of 17, 23, and
31 minutes (power of 0.630, 0.873, and
twice daily
r Regimen C: placebo in the 0.987, respectively, assuming an SD of 20.5
minutes) in latency to REM sleep.
morning and 10 mg donepezil in
the evening
r Regimen D: placebo twice daily

AChE-I, acetylcholinesterase inhibitor; AD, Alzheimer disease; AE, adverse event; AUC, area under the plasma concentration-versus-time curve; CSF,
cerebrospinal fluid; Cmax , maximum observed plasma concentration; ECG, electrocardiogram; IRB, institutional review board; NINCDS/ADRDA, Na-
tional Institute of Neurological and Communicative Disorders and Stroke/Alzheimer’s Disease and Related Disorders Association; REM, rapid eye
movement.

included an open-label substudy to test a single dose of ratory profile, and 12-lead electrocardiogram (ECG).
alicapistat 400 mg in subjects  65 years (the antici- For study 4, patients had to meet the National In-
pated age range for most patients with AD), whereas stitute of Neurological and Communicative Disorders
study 3 included only subjects  65 years. Study 4 and Stroke/Alzheimer’s Disease and Related Disorders
was a multicenter, randomized, double-blind, placebo- Association criteria for probable AD.
controlled, multiple-dose trial conducted in patients For all studies, major exclusion criteria included
with mild to moderate AD who were treated with stable the use of any known inhibitors or inducers of
doses of AChE-I’s and assessed the pharmacokinetics, drug-metabolizing enzymes (eg, CYPs and uridine 5 -
safety, and tolerability of alicapistat. Study 5 was a ran- diphospho-glucuronosyltransferase) within 30 days be-
domized, single-blind, placebo- and active-controlled, fore the first dose of study drug and through the end
4-period crossover trial that assessed sleep parameters of the study. In older participants (group 5 of study 2
in healthy men to test the ability of alicapistat to pene- [all subjects were 65 years], study 3 [all subjects were
trate and influence the CNS, compared with a positive 65 years], and study 4 [all patients were 55-90 years]),
control that would improve sleep (donepezil). psychoactive medications were not allowed within the
The study protocols were approved by institutional 14-day period before receiving alicapistat; in study 5,
review boards (IRBs), and informed consent was ob- any sleep or CNS-active medications were not allowed
tained from all participants; the specific IRBs are listed within 30 days before study drug administration.
in Table 1 for each study. Study 4 in patients with AD
was registered at ClinicalTrials.gov (NCT02220738). Pharmacokinetics
Designs and the primary objectives of the phase 1 tri- Serial blood samples were taken at protocol-dependent
als are shown in Table 1. An additional phase 1 study times following administration of alicapistat for all
(study 6; NCT02573740) was designed to assess the studies except study 5 (Table 1), in which only 2
safety and tolerability of multiple doses of alicapistat blood samples were collected, 1.75 hours following
in patients with mild AD and patients with mild cogni- the evening dose and at the end of polysomnography.
tive impairment due to AD (Supplemental Appendix). Alicapistat diastereomers were measured at AbbVie
(North Chicago, Illinois) using a validated stereoselec-
Subjects tive liquid chromatography/mass spectrometry method.
For studies 1, 2, 3, and 5, healthy subjects were those Alicapistat diastereomers were extracted from plasma
judged to be in general good health based on med- samples, previously treated with 2% potassium phos-
ical history, physical examination, vital signs, labo- phoric acid, using 50%/50% v/v hexane/ethyl acetate
6 Clinical Pharmacology in Drug Development 2018, 00(0)

by a semiautomated 96-well plate method. Deuter- ods: 1 session each on day -2 (for screening), day -1, and
ated internal standards for each diastereomer were day 1. The polysomnography assessments, taken via a
included. The samples were injected onto a C18 col- multichannel montage, included electroencephalogram
umn (Acquity UPLC BEH, 1.7-μm bead size, 2.1 (EEG) activity, electrooculographic activity, and sub-
mm diameter × 50 mm length; Waters, Milford, Mas- mental electromyographic activity. Subjects received 1
sachusetts) and eluted at 25°C and 0.2 mL/min with dose of study drug in the morning (approximately 30
a mobile phase consisting of 55%/45%/0.1% v/v/v minutes after starting breakfast) and 1 dose in the
methanol/water/ammonium hydroxide. Eluted compo- evening (approximately 2 hours before the “lights-off”
nents were quantified with an API 5500 mass spec- period). Each of the 4 study periods was separated
trometer (Sciex, Framingham, Massachusetts) using from the others by at least 10 days. Subjects slept in
positive ion electrospray ionization in the multiple- the sleep laboratory for 3 nights during their first study
reaction monitoring mode and detecting the mass/ period and for 2 nights in subsequent periods. Treat-
charge 434 to 174 transition for the unlabeled di- ment assignment was concealed from the subjects, and
astereomers and the 439 to 174 transition for the they were blindfolded during administration of study
deuterated internal standards. The chromatography/ drug. Study staff who evaluated the polysomnographic
mass spectrometry system was controlled by Analyst assessments also were blinded to treatment assignment.
software, version 1.5 or 1.6 (Applied Biosystems, Fos-
ter City, California). The validated range for the assay Safety
was 0.0469 to 46.9 ng/mL for the alicapistat R,S di- Safety was evaluated throughout the studies based on
astereomer (r2  0.998) and 0.0551 to 55.1 ng/mL for assessments of adverse events (AEs), vital signs, physi-
the alicapistat R,R diastereomer (r2  0.997). The in- cal examination, brief neurological examination, ECG
trarun and interrun coefficients of variation (CVs) were measurements, laboratory tests, and Columbia-Suicide
3.9% and 4.0% for alicapistat R,S and R,R diastere- Severity Rating Scale (C-SSRS).
omers, respectively; the mean intrarun bias ranged be-
tween -4.1% and 5.1%, whereas the mean interrun bias Statistical Methods
was -0.6% to 2.6%. Across studies in this article, the fi-
nal lower limit of quantitation (LLOQ) for a 0.1-mL Computation for the statistical tests was performed
plasma sample was 0.0442 to 0.0477 ng/mL for the ali- with SAS software (Cary, North Carolina), version 8.2
capistat R,S diastereomer and 0.0519 to 0.0561 ng/mL or 9.2, on a Dell workstation using the Unix operat-
for the alicapistat R,R diastereomer. ing system. For the linear mixed-effects model anal-
In study 3, a CSF sample was also collected approx- ysis, SAS procedure PROC MIXED was used with
imately 12 hours after the last dose of alicapistat 200 or the Kenward-Roger option specified. SAS procedures
550 mg twice daily on day 14. The CSF samples were PROC UNIVARIATE and PROC MEANS were used
analyzed for the 2 diastereomers by a method similar to obtain summary statistics. Determinations of sample
to that for the plasma samples. The validated range for size from the perspective of the tolerability assessments
the assay was 0.0543 to 44.2 ng/mL for the alicapistat are reported in Table 1.
R,S diastereomer (r2  0.995) and 0.0637 to 51.8 ng/mL
for the alicapistat R,R diastereomer (r2  0.999). The Pharmacokinetic Analyses
intrarun and interrun CVs were 3.2% and 3.4% for Pharmacokinetic parameters, including the maximum
alicapistat R,S and R,R diastereomers, respectively; the observed plasma concentration (Cmax ), time to Cmax
mean intrarun bias ranged between -2.3% and 12.1%, (tmax ), terminal-phase elimination half-life (t½ ), and
whereas the mean interrun bias was -1.4% to 6.0%. area under the plasma concentration-versus-time curve
The LLOQ in study 3 for a 0.1-mL CSF sample was (AUC), were determined using noncompartmental
0.0547 ng/mL for the alicapistat R,S diastereomer and methods. Summary statistics of pharmacokinetic pa-
0.0642 ng/mL for the alicapistat R,R diastereomer. rameters were calculated by regimen for each study.

Pharmacodynamics Study 1 (Healthy Subjects 18-55 years)


Polysomnography was used in study 5 to assess the ef- To address the issue of linear kinetics and dose pro-
fects of alicapistat on various sleep parameters, partic- portionality, an analysis of covariance (ANCOVA) was
ularly those characterizing REM, including number of performed for each pharmacokinetic parameter (tmax ,
REM episodes, latency to REM sleep, REM density, dose-normalized Cmax , and dose-normalized AUC) in
duration of REM, and REM as a proportion of total turn. Logarithmic transformation was employed for
sleep time, as described previously,14–17 using donepezil Cmax and AUC. For the investigation of the effect of
as a positive control. Briefly, 3 nightly polysomnogra- food, an analysis was performed on data from periods
phy assessments were done in each of the 4 study peri- 1 and 2 (in one period, alicapistat was administered
Lon et al 7

under fasting conditions; in another period, alicapis- the possibility that the regimens had unequal variances
tat was administered with a high-fat/high-calorie break- was considered. Within the framework of the model,
fast). Analysis of variance was performed for tmax and the hypothesis of no difference between each active
the natural logarithms of Cmax and AUC from time 0 drug regimen and placebo was tested. Each of these
to infinity (AUC0- ) of alicapistat diastereomers. The tests was performed at a significance level of .05.
model included fixed effects for sequence, period, and
regimen. The subjects were viewed as a random sam- Safety Analyses
ple. For the assessment of the effect of ketoconazole For all studies, the number and percentage of sub-
on the pharmacokinetics of alicapistat, an analysis was jects with treatment-emergent AEs (TEAEs) were tab-
performed on the same variables analyzed for the as- ulated by Medical Dictionary for Regulatory Ac-
sessment of the effect of food. A repeated-measures tivities preferred term and system organ class with
analysis was performed on the data of the fasting regi- breakdowns by dose level, regimen, or treatment,
men in periods 1 and 2 (alicapistat administered alone) as useful. Also, potentially clinically significant val-
and the data from period 3 (alicapistat with ketocona- ues, according to predefined criteria, for labora-
zole). The model had effects for regimen (alicapistat tory parameters, blood pressure, pulse rate, and
alone or alicapistat with ketoconazole), the period in weight were identified. ECG QT and QTc inter-
which the fasting regimen was administered, and the in- val values > 450 milliseconds and changes from
teraction of regimen and the period factor. baseline > 30 milliseconds were noted.
Study 1 (Healthy Subjects 18-55 years). For the sched-
Study 2 (Healthy Subjects 18-55 and 65 years) uled measurements of vital signs and ECG inter-
To address the issue of linear kinetics and dose vals, repeated-measures analyses were performed. The
proportionality, ANCOVA was performed on dose- model had effects for baseline value, dose level (with
normalized Cmax and AUC of the morning-dose inter- placebo considered a dose level), time of measure-
val of study day 7 (groups 1-2) and day 14 (groups 3-4). ment, and the interaction of dose level and time of
Subjects were classified by dose level. Body weight was measurement.
included as a covariate in the model. A necessary con- Study 2 (Healthy Subjects 18-55 and 65 years). For
dition for including an explanatory variable in the final quantitative ECG variables, for each of study days 1,
model was that the regression coefficient be significant 7, and 14, a linear mixed-effects model analysis was
at a level of 0.10. performed on the scheduled postdose measurements.
The mixed-effects model for these analyses had ef-
fects for baseline value, dose level (with placebo con-
Study 3 (Healthy Subjects  65 years)
sidered a dose level), time of measurement, and the
To address the issue of linear kinetics and dose pro-
interaction of dose level with time of measurement.
portionality, analyses were performed on each pharma-
One-way ANCOVA was performed on the clinical lab-
cokinetic parameter of the morning-dose interval of
oratory variable data at the end of the regimen (study
days 7 and 14 in turn using a linear mixed-effects model.
day 7 for groups 1 and 2 and study day 14 for groups
Observations were classified by dose level and day, and
3 and 4). The baseline value was the covariate. Subjects
there was an effect in the model for the interaction of
were classified by regimen, with placebo considered a
dose level and day. For measures of drug exposure, body
regimen. For blood pressure and pulse rate and their
weight was a covariate.
orthostatic changes, ANCOVA was performed on the
measurements of the last day of the regimen (prior to
Study 4 (Patients With Alzheimer Disease) dose and 4 hours postdose on study day 7 for groups 1
Pharmacokinetic parameters in group 1 (alicapistat and 2 and on study day 14 for groups 3 and 4).
200-mg twice-daily dose) were determined using non- Study 3 (Healthy Subjects  65 years). For quanti-
compartmental methods. tative ECG variables, blood pressure, and pulse rate,
a repeated-measures analysis was performed on the
Pharmacodynamic Analyses scheduled postdose measurements on day 1. The
Study 5 (Healthy Men 25-45 years). For the sleep linear mixed-effects model for these analyses had
parameters, a linear mixed-effects model was used to effects for baseline value, dose level (with placebo
perform an analysis of the data obtained from the considered a dose level), time of measurement, and the
night of day 1. The model had fixed effects for period, interaction of dose level with time of measurement. A
regimen, and cohort. The subjects were viewed as second analysis for ECG variables, blood pressure, and
a random sample. An appropriate structure for the pulse rate was performed on the data from days 7 and
covariance matrix for the observations from a subject 14 jointly. This analysis corresponded with that for day
was selected for each sleep parameter, and in particular 1, but the observations from a subject were classified
8 Clinical Pharmacology in Drug Development 2018, 00(0)

Alicapistat R,R Diastereomer


Alicapistat R,S Diastereomer
1600 1600

Concentraon, ng/mL

Concentraon, ng/mL
1400 1400
1200 1200
1000 1000
800 800
600 600
400 400
200 200
0 0
0 8 16 24 32 40 48 56 64 72 0 8 16 24 32 40 48 56 64 72
Time, h Time, h
Figure 2. Alicapistat R,S and R,R diastereomer arithmetic mean plasma concentration-versus-time profiles after administration of
single oral doses of alicapistat to healthy subjects, in linear and log-linear (inset) scales. Concentration data of the first 6 groups
(50, 100, 200, 400, 700, and 1000 mg) are results from study 1. Concentration data of the 400-mg group in healthy elderly subjects are
results from study 2.

by day as well as by time relative to dosing. The model Overall, food did not significantly affect the alicapi-
had effects for baseline value, dose level, day, time stat exposure. Coadministration of alicapistat 150 mg
relative to dosing, and all the 2- and 3-way interactions with the CYP3A inhibitor ketoconazole resulted in
of dose level, day, and time relative to dosing. The an approximately 2.2-fold increase in alicapistat di-
correlation among the repeated measurements from a astereomer Cmax values and a 3.7-fold increase in
subject was accounted for by applying an appropriate AUC values relative to alicapistat administration alone
variance-covariance structure in the model. It was (Figure 4).
intended that the structure allow for the possibility that Multiple Twice-Daily Oral Doses of Alicapistat. In healthy
the correlation of a pair of measurements on the same subjects  65 years, plasma exposure (Cmax and AUC
day would be higher than a pair of measurements from from time 0 to 12 hours [AUC0-12 ]) of alicapistat
different days. ANCOVA was performed on the clinical R,S was approximately 1.9-fold greater than alicapis-
laboratory variable data from day 14. tat R,R for the 200-mg twice-daily dose and approxi-
mately 1.6-fold greater for the 550-mg twice-daily dose
(Figure 5; Table 3). In contrast, the concentration of
Results alicapistat diastereomers in CSF appeared to be simi-
Pharmacokinetics lar; the mean alicapistat R,S and R,R concentrations
Single Oral Doses of Alicapistat. After oral dosing in in CSF samples after 14 days of dosing were 3.76 and
healthy subjects, plasma concentrations of alicapistat 4.26 ng/mL, respectively, for the 200-mg twice-daily
diastereomers reached peak levels within 2 to 4 hours dose and 7.84 and 9.14 ng/mL, respectively, for the
on average (Figure 2; Table 2). The t½ of alicapistat 550-mg twice-daily dose. Thus, the total targeted ali-
diastereomers ranged from 7 to 10 hours. Exposures capistat CSF drug concentration as projected from pre-
(Cmax and AUC) of the alicapistat R,S diastereomer clinical studies (4 ng/mL) was achieved or exceeded at
were approximately 2-fold greater than those of the R,R these doses in healthy subjects  65 years.
diastereomer for all alicapistat doses. The increase in al- Comparison of alicapistat diastereomer values for
icapistat diastereomer Cmax values was less than dose Cmax and AUC0-12 between subjects  65 years and
proportional (P < .05 for trend; especially notable in young subjects (Table 3) indicated that Cmax values were
the alicapistat 50- to 400-mg dose range), and the in- 21% to 33% lower and AUC0-12 values were 17% to 26%
crease in AUC0- was dose proportional (Figure 3A) in higher in subjects  65 years than in young subjects at
the alicapistat 50- to 1000-mg dose range. Renal elimi- the 200-mg twice-daily dose.
nation of unchanged alicapistat was negligible (data not In patients 55 to 90 years with mild to moder-
shown). ate AD, plasma exposures (Cmax and AUC0-12 ) of
Alicapistat diastereomer exposures were approxi- alicapistat R,S were approximately 2-fold greater than
mately 13% to 29% higher in healthy elderly subjects those of alicapistat R,R (Table 3), consistent with
 65 years than in young adult subjects following a sin- results of the other presented studies. At the 200-
gle 400-mg dose, but this difference was not statistically mg twice-daily dose, exposures (Cmax , AUC0-12 ) of
significant (P  .1740, ANCOVA; Table 2). alicapistat R,S and R,R diastereomers in patients
Lon et al 9

Table 2. Pharmacokinetic Parameters of Alicapistat R,S and R,R Diastereomers in Plasma After a Single Oral Dose

Alicapistat Parameter, Mean ± SD

Healthy
Subjects
Healthy Subjects  65 years

Study 2
Study 1 (Group 5)

Dose Group 50 mg 100 mg 200 mg 400 mg 700 mg 1000 mg 400 mg


(Number of Subjects) (n = 5) (n = 6) (n = 6) (n = 6) (n = 6) (n = 6) (n = 7)

Alicapistat R,S diastereomer, mean ± SD


Cmax , ng/mL 210 ± 125 462 ± 221 633 ± 371 842 ± 275 1780 ± 937 2030 ± 679 1300 ± 519
tmax , h 1.8 ± 0.3 2.4 ± 1.0 2.6 ± 0.9 2.6 ± 1.7 3.6 ± 1.6 3.6 ± 2.0 2.6 ± 1.1
AUC0- , ng·h/mL 734 ± 411 1770 ± 538 3240 ± 1200 5530 ± 1700 10 100 ± 4000 15 200 ± 6020 8100 ± 4460
t½ , h
Harmonic mean ± 7.7 ± 2.0 9.5 ± 3.2 7.2 ± 1.4 7.8 ± 2.5 8.0 ± 1.6 7.4 ± 2.1 9.0 ± 2.9
pseudo SD
Mean ± SD 8.1 ± 2.0 10 ± 2.7 7.6 ± 2.2 8.3 ± 2.2 8.3 ± 1.9 7.9 ± 2.1 9.8 ± 3.2
Alicapistat R,R diastereomer, mean ± SD
Cmax , ng/mL 93 ± 49 206 ± 87 288 ± 145 452 ± 126 952 ± 484 1100 ± 295 636 ± 208
tmax , h 1.8 ± 0.3 2.4 ± 1.0 2.6 ± 0.9 2.7 ± 1.6 3.6 ± 1.6 3.8 ± 1.9 2.6 ± 1.1
AUC0– , ng·h/mL 385 ± 196 883 ± 235 1630 ± 473 3100 ± 732 5540 ± 1810 8370 ± 2200 4080 ± 1750
t½ , h
Harmonic mean ± 7.0 ± 1.4 9.1 ± 2.8 7.5 ± 1.5 8.3 ± 1.5 8.5 ± 1.8 7.6 ± 1.7 8.8 ± 2.2
pseudo SD
Mean ± SD 7.3 ± 1.7 9.6 ± 2.3 7.9 ± 2.2 8.6 ± 1.6 8.9 ± 2.0 8.0 ± 1.6 9.3 ± 2.4
AUC0– , area under the plasma concentration-versus-time curve from time 0 to infinity; Cmax , maximum observed plasma concentration; t½ , terminal-
phase elimination half-life; tmax , time to Cmax .

A B
(n g •h /m L /m g )
0 -1 2, ng•h/mL/mg
ng•h/mL/mg
g •h /m L /m g )

50 50

40 40
0 - , (n

UC 0–12
AU C0–∞

30 30 R,S
D o s e -No r m a liz e d AUC

AUC

R,R
R,S
R,S (Healthy Elderly)
o s e -No r m a liz e d A

20 R,R 20
Dose-Normalized

DDose-Normalized

R,R (Healthy Elderly)


R,S (Healthy Elderly)
R,R (Healthy Elderly) R,S (Mild AD)
R,R (Mild AD)
10 10

0 0
1 00

2 00

4 00

5 50

8 00
50
100
200

400

700

1000

Dose, mg
Dose (mg) Dose (mg)
Dose, mg
Figure 3. Mean ± SD alicapistat R,S and R,R diastereomer dose-normalized AUC0- after administration of single oral doses of
alicapistat (A) and mean ± SD alicapistat R,S and R,R diastereomer dose-normalized AUC0-12 after administration of multiple twice-
daily oral doses of alicapistat (B). AD, Alzheimer disease; AUC, area under the plasma concentration-versus-time curve; AUC0- , AUC
from time 0 to infinity; AUC0-12 , AUC from time 0 to 12 hours.

with mild to moderate AD were comparable with the In the sleep study (study 5), alicapistat R,S and R,R
exposures observed in healthy subjects  65 years diastereomer plasma concentrations following 400-
(Table 3). The increase in AUC0-12 was dose propor- and 800-mg twice-daily doses were comparable with
tional (Figure 3B) in the alicapistat 100- to 800-mg concentrations observed at corresponding doses from
twice-daily dose range. study 2.
10 Clinical Pharmacology in Drug Development 2018, 00(0)

1600 1600 R,R Fasting


R,S Fasting
R,S Non-fasting R,R Non-fasting

Alicapistat R,R Diastereomer


10000 10000
Alicapistat R,S Diastereomer
1400 1400
R,S Ketoconazole R,R Ketoconazole

Concentration, ng/mL
1000
Concentration, ng/mL
1000
1200 1200
100 100

1000 10
1000 10

800 1 800 1

0.1 0.1
600 600
0.01 0.01
400 0 8 16 24 32 40 48 56 64 72 80 88 96 400 0 8 16 24 32 40 48 56 64 72 80 88 96

200 200

0 0

0 8 16 24 32 40 48 56 64 72 80 88 96 0 8 16 24 32 40 48 56 64 72 80 88 96
Time, h Time, h

Figure 4. Arithmetic mean ± SD alicapistat R,S and R,R diastereomer plasma concentration-versus-time profiles after administration
of a single oral dose of alicapistat 150 mg to healthy subjects (n = 11) during part 2 of study 1, in linear and log-linear (inset) scales.
Subjects received alicapistat under fasting conditions (period 1), nonfasting conditions (period 2), and with 400 mg of ketoconazole
once daily (period 3).

Day 1 Day 7 Day 14


1800
Alicapistat R,S Diastereomer

1600
Concentraon, ng/mL

100 mg
1400 200 mg
1200 400 mg
800 mg
1000 200 mg (Healthy Elderly)
550 mg (Healthy Elderly)
800
200 mg (Mild AD)
600

400

200

0
2

4
0

12

8
31

31

32

33

33

34

34

35

36

36

37

37

38
14

15

15

16

16

Day 1 Day 7 Day 14


1000
Alicapistat R,R Diastereomer
Concentraon, ng/mL

800 100 mg
200 mg
400 mg
600 800 mg
200 mg (Healthy Elderly)
550 mg (Healthy Elderly)
400 200 mg (Mild AD)

200

0
2

4
0

12

8
31

31

32

33

33

34

34

35

36

36

37

37

38
14

15

15

16

16

Time, h
Figure 5. Alicapistat R,S and R,R diastereomer arithmetic mean plasma concentration-versus-time profiles after administration of
multiple twice-daily oral doses of alicapistat, in linear scale. Concentration data of the first 4 groups (100, 200, 400, and 800 mg)
are results from study 2. Concentration data of the 200- and 550-mg groups in healthy elderly subjects are results from study 3.
Concentration data of the 200-mg group in patients with mild to moderate AD are results from study 4. AD, Alzheimer disease.

Pharmacodynamics alicapistat in the morning and evening (Table 4). In sub-


Compared with placebo in study 5, there were no sig- jects receiving placebo in the morning and donepezil
nificant changes in any of the sleep-related EEG pa- 10 mg in the evening, expected changes in REM
rameters following administration of 400 or 800 mg sleep EEG parameters with donepezil were observed,
Lon et al 11

Table 3. Pharmacokinetic Parameters of Alicapistat R,S and R,R Diastereomers in Plasma on the Last Day of Twice-Daily Oral Dosing

Patients
With Mild to
Moderate
Healthy Subjects Healthy Subjects  65 years AD

Study 2 (Groups 1-4) Study 3 Study 4

100 mg 200 mg 400 mg 800 mg 200 mg 550 mg 200 mg


Dose Group Twice Daily Twice Daily Twice Daily Twice Daily Twice Daily Twice Daily Twice Daily
(Number of Subjects) (n = 9) (n = 9) (n = 9) (n = 9) (n = 7) (n = 7) (n = 8)
Last Day Day 7 Day 7 Day 14 Day 14 Day 14 Day 14 Day 7

Alicapistat R,S diastereomer, mean ± SD


Cmax , ng/mL 391 ± 134 1060 ± 725 1440 ± 796 2130 ± 841 711 ± 151 1880 ± 755 1120 ± 483
tmax , h 3.67 ± 1.58 2.83 ± 1.87 5.00 ± 3.32 3.44 ± 2.01 5.14 ± 1.07 3.64 ± 1.49 3.31 ± 1.83
AUC0-12 , ng·h/mL 1690 ± 344 3260 ± 1130 6510 ± 2280 10 600 ± 3160 3810 ± 602 8530 ± 2350 4400 ± 1390
t½ , h
Harmonic mean ± 9.38 ± 1.95 11.3 ± 1.97 9.60 ± 2.82 11.0 ± 4.32 9.48 ± 4.16 11.7 ± 2.94 Not
pseudo SD determined
Mean ± SD 9.78 ± 2.17 11.7 ± 2.42 10.7 ± 4.35 12.5 ± 4.69 11.2 ± 4.99 12.4 ± 3.36 Not
determined
Alicapistat R,R diastereomer, mean ± SD
Cmax , ng/mL 167 ± 55.8 458 ± 274 772 ± 335 1160 ± 472 360 ± 74.1 1210 ± 568 524 ± 197
tmax , h 3.67 ± 1.58 2.94 ± 1.91 5.11 ± 3.22 3.56 ± 1.94 5.43 ± 0.98 3.64 ± 1.49 3.25 ± 1.91
AUC0–12 , ng·h/mL 804 ± 125 1620 ± 543 3670 ± 1040 6110 ± 1940 2040 ± 420 5420 ± 1520 2300 ± 664
t½ , h
Harmonic mean ± 8.48 ± 1.54 9.37 ± 1.37 7.99 ± 2.16 9.63 ± 4.20 8.23 ± 2.80 10.8 ± 2.75 Not
pseudo SD determined
Mean ± SD 8.71 ± 1.46 9.58 ± 1.65 8.46 ± 2.10 11.2 ± 4.36 9.34 ± 3.93 11.4 ± 2.88 Not
determined
AD, Alzheimer disease; AUC0-12 , area under the plasma concentration-versus-time curve from time 0 to 12 hours; Cmax , maximum observed plasma
concentration; t½ , terminal-phase elimination half-life; tmax , time to Cmax .

Table 4. Study 5: Summary of Sleep-EEG Parameters at Baseline and After Single AM and PM Oral Doses of PBO, Donepezil, or
Alicapistat

Treatment AM / PM

PBO/PBO PBO/Donepezil 10 mg Alicapistat 400 mg/400 mg Alicapistat 800 mg/800 mg


(n = 17) (n = 16) (n = 17) (n = 14)
Parameter, mean
± SD Baseline Day 1 Baseline Day 1 Baseline Day 1 Baseline Day 1

REM episodes, n 4 ± 1 4 ± 1 4 ± 1 4 ± 1a 4 ± 1 4 ± 1 5 ± 2 4 ± 1
Latency to REM, 57.1 ± 38.2 64.0 ± 26.9 68.3 ± 19.8 52.2 ± 31.0 59.9 ± 21.2 69.6 ± 41.3 53.8 ± 25.9b 78.7 ± 24.6
min
REM density, 7.2 ± 4.5 7.6 ± 4.7 6.6 ± 4.1 10.7 ± 7.1a 8.5 ± 6.2 9.1 ± 5.9 6.4 ± 4.0b 8.4 ± 4.8
number/min
REM duration, min 82.1 ± 33.8 71.7 ± 23.2 68.3 ± 24.7 74.3 ± 24.0 79.7 ± 34.5 73.4 ± 29.7 78.0 ± 32.5 72.6 ± 21.9
REM as percent of 20.5 ± 6.5 18.0 ± 5.2 17.5 ± 5.3 21.7 ± 5.1a 19.6 ± 7.3 18.4 ± 6.5 18.9 ± 7.4 19.1 ± 5.3
total sleep
EEG, electroencephalogram; PBO, placebo; REM, rapid eye movement.
a P < .05 versus PBO/PBO.
b n = 13.
12 Clinical Pharmacology in Drug Development 2018, 00(0)

Table 5. Summary of TEAEs Study 3 (Healthy Subjects 65 years)


One subject receiving alicapistat 550 mg had a seri-
TEAE, n/N (%)
ous and severe TEAE of acute myocardial infarction
Study Placebo All Alicapistat on treatment day 13 and was discontinued from the
study. The TEAE was not considered to be related to
1 2/12 (16.7) 6/35 (17.1) alicapistat.
2 3/12 (25.0) 11/43 (25.6)
3 4/8 (50.0) 8/16 (50.0)
4 2/5 (40.0) 4/12 (33.3)
Study 4 (Patients With Alzheimer Disease)
5 3/17 (17.6)a 2/17 (11.8)
The proportions of patients who reported 1 TEAE
were similar in patients who received alicapistat 200 mg
TEAE, treatment-emergent adverse event. twice daily (44%) and those who received placebo twice
a Subjects received placebo in the morning and donepezil 10 mg in the

evening or placebo at both times; no subjects (0/4) who received placebo


daily (40%); no patients who received 550 mg alicapistat
alone experienced an AE. twice daily reported a TEAE. There were no deaths or
serious TEAEs reported in treated patients. One patient
who was screened had a serious AE (severe syncope)
and was discontinued from the study before beginning
including increases in the number of REM episodes
a study drug regimen.
during sleep, REM density, and REM time as a per-
centage of total sleep time.
Study 5 (Healthy Men 25-45 years)
Safety There was no pattern to the nature or frequency of
In general, the incidence of TEAEs was similar in the TEAEs following administration of alicapistat, and all
placebo and alicapistat treatment groups (Table 5). Ex- events were mild in severity. There were no deaths or
cept for minor and inconsistent effects on blood pres- serious TEAEs.
sure, pulse rate, PR interval, and QTcF interval at
higher doses, there were no clinically significant changes Discussion
in other vital signs and laboratory measurements across Alicapistat is a potent and selective inhibitor of calpain
doses of alicapistat. All subjects were negative for sui- and has demonstrated efficacy in a broad range of
cidal ideation or behavior as assessed by the C-SSRS.18 animal models relevant to AD. To our knowledge, this
is the first report of a comprehensive phase 1 program
Study 1 (Healthy Subjects 18-55 years) for an orally active, selective, small-molecule calpain in-
Most TEAEs were mild in severity. One subject who hibitor. The pharmacokinetic and safety characteristics
received alicapistat reported 2 TEAEs (hypotension of alicapistat were favorable for a clinical develop-
and presyncope) that were moderate in severity as as- ment candidate. Exposure was dose proportional
sessed by the investigator. TEAEs reported by subjects and without a food effect or dramatic perturbations
who received alicapistat were abdominal pain, diarrhea, related to age. There were no patterns that could have
nausea, pharyngitis, dizziness, headache, paresthesia, indicated safety concerns. However, the pharmaco-
confused state, and dysmenorrhea. One subject who re- dynamic results were not supportive of continued
ceived alicapistat reported 2 TEAEs (dizziness and con- development.
fused state) that were assessed by the investigator as The pharmacokinetic characteristics of alicapistat
possibly related to the study drug. One subject who re- were generally consistent across the 5 phase 1 stud-
ceived alicapistat discontinued from the study owing to ies discussed in this report. Cmax was reached in 2 to
a TEAE of neutropenia that was assessed by the inves- 5 hours, and t½ ranged from 7 to 12 hours, depend-
tigator as not related to alicapistat and mild in severity. ing on dose and study. AUC was dose proportional in
No deaths, serious AEs, or severe AEs occurred during the alicapistat 50- to 1000-mg dose range. Exposure
the study. to the alicapistat R,S diastereomer in plasma was ap-
proximately 2-fold greater than exposure to the R,R
Study 2 (Healthy Subjects 18-55 and 65 years) diastereomer in both healthy young subjects and sub-
The majority of TEAEs were mild in severity and jects 65 years, as well as in patients with mild to
considered unrelated to alicapistat. The most common moderate AD. The greater exposure to the R,S diastere-
TEAEs were discomfort after lumbar puncture (11.1%), omer was favorable, as it is a far more potent inhibitor
constipation (5.6%), nausea (5.6%), and headache of calpain (inhibitor constant [Ki ], 56 nM) than the
(5.6%). No subjects discontinued the study prematurely R,R diastereomer (Ki , 1330 nM) in vitro. Overall, food
because of a TEAE. did not significantly affect the alicapistat pharmacoki-
There were no serious TEAEs or deaths. netic profile, but coadministration of alicapistat with
Lon et al 13

ketoconazole resulted in an approximately 2.2-fold in- not increase further). Of note, alicapistat is a substrate
crease in alicapistat diastereomer Cmax values and a of P-glycoprotein, an efflux transporter, which might
3.7-fold increase in AUC values relative to alicapistat have contributed to its limited CNS levels.22 Therefore,
administration alone. Because alicapistat is a substrate the lack of a pharmacodynamic effect of alicapistat
of CYP3A4, the increases in alicapistat exposure when in the sleep study suggests that inadequate concentra-
CYP3A4 was inhibited by ketoconazole were consistent tions of alicapistat might have been achieved in the
with expectations. Pharmacokinetics was comparable CNS in that population, that higher concentrations are
in young and elderly subjects and patients with mild to needed in humans than in animals to observe pharma-
moderate AD. codynamic effects, or that animal studies are not at all
No safety signals of concern were identified across predictive of the effects of alicapistat in humans. This
the 5 alicapistat phase 1 trials, and the incidence of failure to confirm a central pharmacodynamic effect
TEAEs was similar in placebo and alicapistat groups. represents a major risk for further development of
There were no deaths or serious TEAEs related to alicapistat for AD.
alicapistat, and all subjects were negative for suicidal
ideation or behavior. Safety was similar regardless of
age. Acknowledgments
The pharmacodynamic potential of alicapistat was Medical writing support was provided by Richard M. Ed-
probed by measuring REM sleep parameters in healthy wards, PhD, and Michael J. Theisen, PhD, of Complete Pub-
men. To potentially exert an effect in patients with AD, lication Solutions, LLC (North Wales, Pennsylvania), which
a therapeutic agent must be able to reach the CNS. was funded by AbbVie. AbbVie and the authors thank the
Studies have shown that donepezil reaches therapeutic participants in the clinical trials and all study investigators
concentrations in CSF in humans19 and that it mod- for their contributions. The authors thank all colleagues in
ulates brain function as reflected by changes in REM AbbVie preclinical functions who provided preclinical data on
sleep parameters in patients with AD, probably via its ABT-957 and its diastereomers.
cholinergic actions. In animal models, both donepezil
and alicapistat had robust effects (eg, ACh release and
EEG REM sleep), indicating effects within the CNS, Declaration of Conflicting Interests
which will be described in detail in a separate publica- All authors are employees of AbbVie and may own AbbVie
tion. Therefore, we assessed the pharmacodynamic ef- stock or options.
fects of alicapistat 400 or 800 mg twice daily on sleep
behavior, including REM, in healthy subjects as an indi-
cation of CNS penetration and activity (study 5). Com- Funding
pared with placebo, alicapistat had no effect on REM AbbVie Inc. funded these studies. AbbVie participated in the
sleep parameters, whereas the active control, donepezil, study design, research, data collection, analysis and interpre-
had the expected effects on REM sleep,20 suggesting tation of data, writing, reviewing, and approving the publica-
that the study was appropriately designed, powered, tion.
and conducted. The lack of an effect on REM sleep This clinical trial data can be requested by any qualified
was unexpected because the CSF total alicapistat con- researchers who engage in rigorous, independent scientific re-
centration at the doses used was predicted to be ap- search, and will be provided following review and approval of
proximately 8 ng/mL (based on study 3). Although a research proposal and Statistical Analysis Plan (SAP) and
the estimated levels of alicapistat stereoisomers in the execution of a Data Sharing Agreement (DSA). Data requests
CSF (range, 3.76-9.14 ng/mL, corresponding to 8.66- can be submitted at any time and the data will be accessible
21.1 nM) did not reach the in vitro Ki values (R,S di- for 12 months, with possible extensions considered. For more
astereomer, 56 nM; R,R diastereomer, 1330 nM), this information on the process, or to submit a request, visit the
CSF concentration of alicapistat exceeded the concen- following link: https://www.abbvie.com/our-science/clinical-
tration of 4 ng/mL, suggested to be efficacious based trials/clinical-trials-data-and-information-sharing/data-and-
on animal studies. However, it is known that animal information-sharing-with-qualified-researchers.html.
models have limited predictive power for estimating the
achieved concentrations of drugs in human CSF.21 The
use of higher doses of alicapistat in the sleep study
References
was not considered because, although earlier preclin- 1. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl
ical toxicology studies had suggested that they could J Med. 2010;362(4):329–344.
be tolerated (up to 250-500 mg/kg in mice; up to 800- 2. Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer
1000 mg/kg in dogs), there was a ceiling effect on drug disease in the United States (2010-2050) estimated using
exposure with greater doses (ie, levels of alicapistat did the 2010 census. Neurology. 2013;80(19):1778–1783.
14 Clinical Pharmacology in Drug Development 2018, 00(0)

3. Getz GS. Calpain inhibition as a potential treatment of of normal subjects. Psychiatry Clin Neurosci. 2002;
Alzheimer’s disease. Am J Pathol. 2012;181(2):388–391. 56(3):307–308.
4. Reitz C, Brayne C, Mayeux R. Epidemiology of 16. Riemann D, Gann H, Dressing H, Muller WE, Alden-
Alzheimer disease. Nat Rev Neurol. 2011;7(3):137–152. hoff JB. Influence of the cholinesterase inhibitor galan-
5. Yiannopoulou KG, Papageorgiou SG. Current and fu- thamine hydrobromide on normal sleep. Psychiatry Res.
ture treatments for Alzheimer’s disease. Ther Adv Neurol 1994;51(3):253–267.
Disord. 2013;6(1):19–33. 17. Schredl M, Weber B, Leins ML, Heuser I. Donepezil-
6. Birks J. Cholinesterase inhibitors for Alzheimer’s disease. induced REM sleep augmentation enhances memory
Cochrane Database Syst Rev. 2006(1):CD005593. performance in elderly, healthy persons. Exp Gerontol.
7. McShane R, Areosa Sastre A, Minakaran N. Me- 2001;36(2):353–361.
mantine for dementia. Cochrane Database Syst Rev. 18. Posner K, Brown GK, Stanley B, et al. The Columbia-
2006(2):CD003154. Suicide Severity Rating Scale: initial validity and inter-
8. Cummings J, Aisen PS, DuBois B, et al. Drug develop- nal consistency findings from three multisite studies with
ment in Alzheimer’s disease: the path to 2025. Alzheimers adolescents and adults. Am J Psychiatry. 2011;168(12):
Res Ther. 2016;8:39. 1266–1277.
9. Goll DE, Thompson VF, Li H, Wei W, Cong J. The cal- 19. Litvan I, Phipps M, Pharr VL, et al. Randomized
pain system. Physiol Rev. 2003;83(3):731–801. placebo-controlled trial of donepezil in patients with
10. Vosler PS, Brennan CS, Chen J. Calpain-mediated sig- progressive supranuclear palsy. Neurology. 2001;57(3):
naling mechanisms in neuronal injury and neurodegen- 467–473.
eration. Mol Neurobiol. 2008;38(1):78–100. 20. Jackson S, Ham RJ, Wilkinson D. The safety and toler-
11. Ferreira A. Calpain dysregulation in Alzheimer’s disease. ability of donepezil in patients with Alzheimer’s disease.
ISRN Biochem. 2012;2012:728571. Br J Clin Pharmacol. 2004;58(suppl 1):1–8.
12. Trinchese F, Fa M, Liu S, et al. Inhibition of cal- 21. Nau R, Sorgel F, Eiffert H. Penetration of drugs
pains improves memory and synaptic transmission in through the blood-cerebrospinal fluid/blood-brain bar-
a mouse model of Alzheimer disease. J Clin Invest. rier for treatment of central nervous system infections.
2008;118(8):2796–2807. Clin Microbiol Rev. 2010;23(4):858–883.
13. Medeiros R, Kitazawa M, Chabrier MA, et al. Cal- 22. Miller DS, Bauer B, Hartz AM. Modulation of P-
pain inhibitor A-705253 mitigates Alzheimer’s disease- glycoprotein at the blood-brain barrier: opportunities
like pathology and cognitive decline in aged 3xTgAD to improve central nervous system pharmacotherapy.
mice. Am J Pathol. 2012;181(2):616–625. Pharmacol Rev. 2008;60(2):196–209.
14. Hornung OP, Regen F, Dorn H, et al. The effects of
donepezil on postlearning sleep EEG of healthy older Supporting Information
adults. Pharmacopsychiatry. 2009;42(1):9–13. Additional supporting information may be found on-
15. Kanbayashi T, Sugiyama T, Aizawa R, et al. Effects line in the Supporting Information section at the end
of donepezil (Aricept) on the rapid eye movement sleep of the article.

You might also like