Shirasaki 2006

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

JOURNAL OF OCULAR PHARMACOLOGY AND THERAPEUTICS

Volume 22, Number 6, 2006


© Mary Ann Liebert, Inc.

Retinal Penetration of Calpain Inhibitors in Rats


After Oral Administration

YOSHIHISA SHIRASAKI,1 MASAZUMI YAMAGUCHI,1 and HIROYUKI MIYASHITA2

ABSTRACT

Calpain-mediated proteolysis has been involved in neuronal cell death of retinal neuro-
logical degeneration. An aldehyde-based calpain inhibitor, SJA6017 (1), was effective fol-
lowing oral administration in a rat retinal ischemia model but had low oral bioavailability.
The aim of this study was to identify calpain inhibitors with good retinal penetration after
oral dosing. The orally bioavailable inhibitors, hemiacetal 3 (SNJ-1715), amphipathic ke-
toamide 5 (SNJ-1945), and pyridine ketoamide 6 (SNJ-2008), were evaluated for their retinal
pharmacokinetic (PK) profiles. The retinal drug exposure of these inhibitors was more than
tenfold higher than 1. Among these compounds, 5 exhibited the most favorable retinal PK
properties, such as good penetration and long half-life. Comparisons of 5 and the structurally
related ketoamide 6 suggested that the presence of a methoxy diethylene glycol moiety re-
sulted in the inhibitor with high penetration into the retina and the sustained high retinal
levels. Ketoamide 5 was selected as the development candidate for the treatment of retinal
diseases.

INTRODUCTION mice, which are an accepted animal model for


photoreceptor degeneration in retinitis pigmen-
tosa.10,11 These results suggest that calpains are a
C ALPAINS ARE CALCIUM-ACTIVATED intracellular
cysteine endoproteases, and 15 genes of
isozymes have, so far, been identified.1–3 Two ma-
potential target for retinal neurological disorders,
such as glaucoma and retinitis pigmentosa.
jor isoforms, -calpain (calpain I) and m-calpain A number of calpain inhibitors have been
(calpain II), are ubiquitously present in mam- synthesized and some of them have demon-
malian cells. These enzymes have been impli- strated neuroprotective efficacy in several animal
cated in numerous neurological disorders, in- pharmacological models through injections.12–19
cluding stroke, Alzheimer’s disease, central Because most of retinal diseases are chronic dis-
nervous system diseases, multiple sclerosis, eases, intravenous (i.v.) administration is incon-
spinal cord injury, and traumatic brain injury venient. Topical instillation to the eyes would be
(TBI).4–7 Previously, our researchers have re- desirable, but it cannot readily deliver drugs into
ported that an activation of calpains occurred in the retina because of several pathological and
retinal neuronal cell death during retinal is- anatomical barriers, such as nasolacrimal drain-
chemia-reperfusion8 and acute ocular hyperten- age, metabolism, protein binding, and lens bar-
sion in rats.9 It has also been shown that calpains rier.20 Therefore, oral drugs are preferable for the
are involved in photoreceptor cell death in rd1 treatment of the retinal diseases. Our group has

1Senju Pharmaceutical Co., Ltd., Hyogo, Japan.


2Faculty of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.

417
418 SHIRASAKI ET AL.

reported that oral administration of a highly po- We have improved the aqueous solubility of 4 by
tent inhibitor, dipeptidyl aldehyde 1 (SJA6017),21,22 incorporating a nonionic amphiphile and identi-
showed neuroprotective efficacy in the rat retinal fied a dipeptidyl -ketoamide 5 (SNJ-1945) that
ischemia model.3 However, a relatively high dose showed good plasma exposure in monkeys (Fig.
(500 mg/kg) was required, presumably owing to 1).25 In addition, introduction of a pyridine moi-
low oral bioavailability (BA), which was ascribed ety as a basic water-soluble moiety also provided
to a metabolically labile aldehyde moiety and low a potent and orally bioavailable ketoamide 6
water solubility.23 Consequently, we investigated (SNJ-2008) (Fig. 1).26
whether replacing the aldehyde warhead with Our aim was to discover orally available cal-
more chemically stable warheads, such as -ke- pain inhibitors with good retinal penetration.
toamide and cyclic hemiacetal moiety, could im- Drug penetration into the retina is restricted by
prove the oral BA (Fig. 1). The cyclic hemiacetal the inner and outer blood-retinal barriers (BRB).
derivative 2 showed a high metabolic stability Therefore, the plasma drug levels are not always
with adequate aqueous solubility. Further opti- parallel to the retinal levels.27,28 Although the in-
mization of 2 for inhibitory activity provided an hibitors 3, 5, and 6 showed good plasma expo-
oral available inhibitor 3 (SNJ-1715) with an ac- sure following oral administration, it is uncertain
ceptable potency.23,24 On the other hand, -ke- whether the inhibitors penetrate into the retina.
toamide analog 425 (Fig. 1) was equipotent to the In this study, we evaluated the inhibitory activi-
original aldehyde 1, and more membrane-per- ties, oral BA, and retinal penetration in rats of cal-
meable and metabolically stable than 1, but it pain inhibitors, original aldehyde 1, hemiacetal 3,
showed poor oral absorption in monkeys, likely amphipathic ketoamide 5, and pyridine ke-
owing to its extremely low aqueous solubility. toamide 6.

FIG. 1. Drug design concept of novel calpain inhibitors.


RETINAL PENETRATION OF CALPAIN INHIBITORS 419

METHODS dling, housing, and experimentation conformed to


the ARVO Resolution for the Use of Animals in
Materials Ophthalmic and Vision Research. Animals were
fasted overnight before dosing in the case of oral
The calpain inhibitors, SJA6017 (1),21,22
dosing. Animals were allowed unrestricted access
SNJ-1715 (3),23,24 SNJ-1945 (5),25 and SNJ-2008
to water. Rats (n  4–5 per time point) were dosed
(6),26 were synthesized, as reported previously.
with compounds 1, 3, 5, and 6 orally by gavage at
Sodium carboxymethyl cellulose (CMC-Na) and
a dose of 10 or 500 mg/kg. They were also dosed
polyethylene glycol 300 were purchased from
with 1, 3, 5, and 6 at 3 mg/kg by bolus injection
Wako Pure Chemical Industries, Ltd. (Osaka,
through a tail vein. The animals were anesthetized
Japan).
with isoflurane at a predetermined time (0.1 [i.v.
only], 0.25, 0.5, 1, 2, 4, and 8 h after dosing), and
Calpain inhibition assay blood samples (5–6 mL) were collected from the
Calpain inhibition assays were performed, as abdominal aorta into heparinized syringes. Then,
described in the previous paper,29 using com- these animals, including the eyes, were perfused
mercial -calpain (human erythrocyte; Cal- with saline (12 mL). The eye globes were enucle-
biochem, San Diego, CA) and m-calpain (porcine ated, and the retinas were excised. Plasma was pre-
kidney; Calbiochem). Assay solution, including pared by centrifugation of the blood. All plasma
80 mM Tris-HCl (pH 7.4), 15 mM -mercap- and retina samples were frozen and stored at
toethanol, and 15 nmol of enzyme was used. The 30°C until analysis.
assay solution (150 L), 0.5 mM Boc-Val-Leu-Lys-
AMC (50 L; Bachem, Budendorf, Switzerland), Preparation of plasma samples
and dimethyl sulfoxide (2.5 L) including in- Plasma samples (0.2 mL) were mixed with
hibitor were placed in each well of 96-well plates. phosphate-buffered saline (1 mL) and the solu-
Reaction was started by the addition of 25 mM tion was transferred to OASIS® HLB (60 mg) car-
CaCl2 (50 L) to a test well and 1 mM of ethyl- tridges (Waters; Tokyo, Japan). The solid phase
enediaminetetraacetic acid (50 L) in a blank was washed with 5% aqueous MeOH (1 mL) and
well. Enzyme activity was determined by the in- eluted MeOH (1 mL). The eluent was evaporated
crease of fluorescence (ex  360 nm, em  440 to dryness in vacuo at 40°C, reconstituted with 1
nm) monitored at 37°C, using a CYTO FLUOR mL of mobile phase (MeOH/H2O/HCO2H 
multiwell plate reader (Perspective Biosystems, 40:60:0.1 for 1, 5, and 6 or MeCN/H2O  70:30
Foster City, CA). Percent enzyme inhibition was for 3) and 10-L aliquots of these solutions were
determined by a comparison of this activity to analyzed by liquid chromatography/mass spec-
that of a solution without inhibitor. trometry/mass spectrometry (LC-MS/MS; de-
scribed below).
Dose preparation
For pharmacokinetic studies, the i.v. dose of 1, Preparation of retinal samples
3, 5, or 6 (3 mg/kg) was administered in a vehi- The retinas were weighted and homogenized
cle consisting of polyethylene glycol 300/saline in MeOH (5 mL). The homogenates were cen-
(3:7) at a concentration of 0.6 mg/mL (5 mL/kg). trifuged at 3000g for 10 min. The supernatant (4
The oral dose (10 mg/kg for 3, 5, and 6) was ad- mL) was transferred to glass test tubes and evap-
ministered in a suspension in 0.5% CMC-Na so- orated to dryness in vacuo at 40°C. The residue
lution at a concentration of 2 mg/mL (5 mL/kg). was reconstituted with mobile phase (1 mL),
Compound 1 for oral dose (500 mg/kg) was for- transferred to 1.5-mL tubes, and centrifuged at
mulated as a suspension in 0.5% CMC-Na solu- 10,000g for 10 min. Then, 10-L aliquots of the
tion at a concentration of 100 mg/mL (5 mL/kg). supernatants were analyzed by LC-MS/MS.

Pharmacokinetic and retinal penetration studies LC-MS/MS analysis of plasma and


retina samples
Male Sprague-Dawley rats (weight 200–250 g)
were purchased from Charles River Laboratories The plasma and retinal concentration of each
Japan, Inc. (Yokohama, Japan). All aspects of han- test compound were determined by turbo ion
420 SHIRASAKI ET AL.

spray on an API 4000 triple-quadrupole mass Determination of pharmacokinetic parameters


spectrometer (Applied Biosystems/MDS-Sciex;
The pharmacokinetic parameters were deter-
Ontario, Canada) equipped with a turbo ion
mined from the resulting mean plasma or retinal
spray source using multiple reaction monitoring
concentration versus time profiles by using the
(MRM). Chromatography was performed on a
noncompartmental model analysis (WinNonlin
NANOSPACE SI-2 HPLC system (Shiseido;
version 2.1; Pharsight, Mountain View, CA). The
Tokyo, Japan) with Shiseido Capcell pak® C18
calculated parameters are maximal concentration
MG-II column (75  1.5 mm, 5 m) at 40°C, us-
of compounds (Cmax), time required to reach Cmax
ing a mobile phase (MeOH/H2O/HCO2H 
(Tmax), plasma and retina terminal half-lives (T1/2
40:60:0.1) for compounds 1, 5, and 6 or Shiseido
and t1/2, respectively), the area under the curves
Capcell pak C18 ACR (150  1.5 mm, 3 m) at
from time 0 to infinity (AUC) in plasma or retina,
40°C, using a mobile phase (MeCN/H2O  70:30)
the volume of distribution at steady state (Vss),
for 3 at a flow rate of 0.20 mL/min. The dwell time
and plasma total clearance (Cl). Oral bioavail-
for the MRM was 150 ms. A peak was defined by
ability (F) was calculated from the plasma AUC
manual integration. The negative ion mode was
values after oral and i.v. dosing, corrected by the
used for 1 and 3. The compounds 5 and 6 were
ratio of the doses administrated.
measured by positive ion mode. Multiple reaction
monitoring using the product ions (m/z 371.1 
158.5 for 1, m/z 350.2  117.0 for 3, m/z 492.2 
102.8 for 5, and m/z 495.1  106.1 for 6) was used
RESULTS
for quantification. The levels of these compounds
in plasma or retina samples were determined by
Inhibitory activities
using standard curves in which control plasma or
control retina was spiked with increasing concen- Previously, we evaluated the calpain in-
trations of these compounds (5–50 ng for plasma, hibitory activities of some inhibitors in a dye
2.5–50 ng for retina). A regression fit, 1/X, was used assay method.22,24,25 In this study, the activi-
to quantify the unknowns. The limits of quantifi- ties of aldehyde 1, hemiacetal 3, amphipathic
cation for these compounds were 5 ng/mL in ketoamide 5, and pyridine ketoamide 6 were
plasma and 2.5 ng/g of tissue in retina, respec- determined under the same conditions, using
tively. Data reduction were performed using Sciex a fluorogenic substrate to compare the poten-
Analyst versus 1.3 software (Applied Biosystems/ tial retinal efficacy. The data are summarized
MDS-Sciex, Ontario, Canada), and the mean and in Table 1. The inhibitory activities of 3, 5, and
standard deviation was calculated by using Micro- 6 are acceptable and comparable to original
soft Excel® software (Microsoft, Tokyo, Japan). aldehyde 1.

TABLE 1. ENZYME INHIBITORY ACTIVITY, PHYSICOCHEMICAL PROPERTIES OF CALPAIN INHIBITORS

IC50 (M)a
Metab.b
Compound -calpain m-calpain (%) Mw c cLogD7d

1 0.028 0.023 3e 372 3.21


3 0.087 0.071 100e 351 0.51
5 0.062 0.045 41f 491 2.27
6 0.029 0.017 23g 494 3.31
aTheIC50 values were determined by the fluorescence assay method (n  2).
bMetabolic stability represented in residual percent after incubation with human
S9 for 0.5 h at 37°C.
cMolecular
weight.
dThe dissociative partition coefficient ACD cLogD was calculated using Advanced Chemistry Development Soft-
7
ware V4.76 for Solaris (ACD/Labs).
eReference 23.
fReference 25.
gReference 26.
RETINAL PENETRATION OF CALPAIN INHIBITORS 421

TABLE 2. PLASMA PK PARAMETERS OF CALPAIN INHIBITORS IN RATS

Parameters 1 3 5 6

Route i.v. p.o. i.v. p.o. i.v. p.o. i.v. p.o.


Dose (mg/kg) 3.0 500/(10)g 30.0 100.0 30.0 100.0 30.0 100.0
The number of animals 4.0 4 50.0 50.0 50.0 50.0 50.0 50.0
(n/time point)
AUCa (M  h) 1.50 45/(0.90)g 2.60 5.70 2.30 7.40 4.50 6.80
g  h/mL 0.56 16/(0.34)g 7.40 2.00 1.10 3.60 2.20 3.40
Cmax (M) — 9.4/(0.19)g — 5.60 — 3.50 — 8.60
(g/mL) 3.5/(0.70)g 2.00 1.70 4.30
Tmax (h) — 1.0 — 0.25 — 0.25 — 0.25
T1/2b (h) 0.70 — 1.90 — 3.80 — 2.10 —
zc (h1) 0.99 — 0.39 0.18 0.33
Vssd (L/kg) 6.10 — 2.30 — 4.30 — 0.98 —
Cle (mL/min/kg) 890.0 — 550.0 — 440.0 — 220.0 —
Ff (%) 18 66 96 45

PK, pharmacokinetic; AUC, area under the curves; i.v., intravenous; p.o., per os.
aThe area under the curves for 0 h to infinity.
bThe terminal half-life.
cThe terminal phase elimination rate constant.
dThe steady-state volume of distribution.
eThe total clearance.
fOral bioavailability.
gThe values dose-normalized to 10 mg/kg.

Oral plasma bioavailability P3 moiety, ketoamide 6 demonstrated the signif-


icantly lower Vss value (0.98 L/kg) than 5 (Vss 
The oral plasma bioavailability (BA) of 1, 3, 5,
4.3 L/kg).
and 6 were investigated in Sprague-Dawley rats
dosed i.v. at 3 mg/kg8 and orally at 10 mg/kg
for 3, 5, and 6 or 500 mg/kg23,26 for 1 (Table 2,
Retinal penetration
Fig. 2). An oral dose of 500 mg/kg compound 1 The retinal drug levels were determined after
showed neuroprotective efficacy in the rat retinal oral administration of 1, 3, 5, and 6. The results
ischemia model. The oral dose was selected to
compare the potential efficacy of these com-
pounds. Aldehyde 1 had a very high rate of total
body clearance (Cl  89 mL/min/kg), which ex-
ceeds the hepatic blood flow for a rat (approxi-
mately 60 mL/min/kg), resulting in a short half-
life, in spite of its large volume of distribution at
steady state (Vss) of 6.1 L/kg. Compounds 3, 5,
and 6 are well absorbed and showed significantly
higher oral BA than original aldehyde 1. The Tmax
of 3, 5, and 6 were shorter (Tmax  0.25 h) than
that of 1. After i.v. dosing, 3, 5, and 6 showed
longer plasma terminal half-life (T1/2) and lower
total clearance (Cl), compared to 1. Hemiacetal 3
demonstrated relatively higher clearance (Cl  55
mL/min/kg), compared to the value estimated
by its excellent in vitro metabolic stability (Table
1). Ketoamide 5 exhibited excellent BA (F  96%)
FIG. 2. Plasma levels of aldehyde 1 (500 mg/kg), hemi-
and the longest half-life (T1/2  4.3 h) among acetal 3, amphipathic ketoamide 5, and pyridine ke-
these compounds. Although the structural dif- toamide 6 after oral administration to rats (10 mg/kg).
ference between ketoamides 5 and 6 was the only Results represent mean  standard deviation (n  4–5).
422 SHIRASAKI ET AL.

ratio in these compounds, whereas it had the


shortest retinal half-life (t1/2  2.8 h). Ketoamide
5 exhibited moderate Cmax (0.40 M), high AUC
(2.4 M  h), C8h (0.11 M), and R/P ratio (0.32).
In addition, 5 displayed the longest t1/2 (4.3 h) as
well as plasma half-life (T1/2). Pyridine ke-
toamide 6 showed relatively low AUC and R/P
ratio, whereas the t1/2 was moderate (2.9 h). The
elimination rate constant (z) in retina of 6 was
larger than that of 5, in spite of the larger
lipophilicity of 6. Among these compounds, ke-
toamide 5 exhibited more favorable retinal PK
properties, such as good penetration, long half-
life, and high C8h.

FIG. 3. Retinal levels of aldehyde 1 (500 mg/kg), hemi-


acetal 3, amphipathic ketoamide 5, and pyridine ke- DISCUSSION
toamide 6 after oral administration to rats (10 mg/kg).
Results represent mean  standard deviation. (n  4–5).
Original aldehyde 1 showed very poor retinal
AUC and R/P ratio (0.062), although it exhib-
were summarized in Figure 3 and Table 3. After ited low molecular weight (Mw  372) and a
oral administration of these compounds, the Cmax high Vss value (Vss  6.1 L/kg). It was suggested
in plasma and retinas occurred simultaneously that the aldehyde moiety of 1 formed a covalent
and the retinal half-lives (t1/2) roughly reflected adduct with nucleophiles contained in the cir-
their plasma T1/2 values. The retina-to-plasma culatory system. Furthermore, relatively high
AUC ratios (R/P ratio) of 3, 5, and 6 were signif- lipophilicity (cLogD7 3.21) may lead to an active
icantly higher than that of original aldehyde 1. efflux by transporters such as P-glycoprotein (P-
The retinal Cmax, AUC, and concentration at 8 h gp) of retinal pigmented epithelium.27 Both the
after oral administration (C8h) of 3, 5, and 6 were reactive aldehyde moiety and high lipophilic na-
slightly less or comparable to those of 1, although ture may be the limiting factors of retinal pene-
the oral doses of 3, 5, and 6 were fiftyfold less tration.
than that of 1. Hemiacetal 3 showed the highest Hemiacetal 3 and ketoamide 5 and 6 exhibited
Cmax (1.8 M), AUC, (2.6 Mh), and R/P (0.46) both higher oral BA and retinal penetration com-

TABLE 3. RETINAL PK PARAMETERS AFTER SINGLE ORAL ADMINISTRATION


(1500 MG/KG; 3, 5 AND 6: 10 MG/KG) OF CALPAIN INHIBITORS IN RATS

Parameters 1 3 5 6

R/P ratioa 0.062 0.46 0.32 0.10


AUCb (M  h) 2.8/(0.056)f 2.6 2.4 0.71
(g  h/mL) 1.0/(0.021)f 0.91 1.1 0.35
Cmax (M) 0.60/(0.012)f 1.8 0.40 0.51
(g/mL) 0.22/(0.045)f 0.63 0.20 0.25
Tmax (h) 1.0 0.25 0.25 0.25
T1/2c (h) 2.8 2.3 4.3 2.9
zd (h1) 0.25 0.30 0.16 0.24
C8he (M) 0.10/(0.0021)f 0.050 0.11 0.021
(g/mL) 0.037/(0.00078)f 0.018 0.054 0.010

PK, pharmacokinetic; R/P, retina to plasma; AUC, areas under the curve.
aRetina-to-plasma AUC ratio.
bThe area under the curves for 0 h to infinity.
cThe terminal half-life.
dThe terminal phase elimination rate constant.
eThe retinal concentration at 8 h after administration.
fThe value dose-normalized to 10 mg/kg.
RETINAL PENETRATION OF CALPAIN INHIBITORS 423

pared to 1, owing to the presence of a more chem- nal levels exceeded 0.1 M at an oral dose of 10
ically stable warhead instead of aldehyde. Hemi- mg/kg, the effective oral dose in the ischemia
acetal 3 showed the highest retinal AUC, R/P ra- model could be an order of magnitude smaller
tio, and Cmax values, although the oral BA and than that of 1. On the other hand, the mean reti-
Vss were lower than those of 5. This is most likely nal levels of 3 and 5 were 0.06–1.8 and 0.11–0.40
the result of a smaller molecular weight and M, respectively, after oral dosing of 10 mg/kg.
lower lipophilicity, which leads to weaker plasma These indicated that the compounds would pro-
protein binding and an increase in unbound frac- vide retinal efficacy at a significantly lower oral
tions in plasma. The relatively high plasma clear- dosage, compared to 1. In fact, 3 and 5 demon-
ance of 3 may be ascribed to low plasma protein strated neuroprotective efficacy at a fivefold
bindings and metabolism by plasma esterases. lower dose (100 mg/kg) in the rat retinal ischemia
Hemiacetal 3 demonstrated a shorter retinal half- model,23,31 although these were not effective at a
life (t1/2), presumably owing to its high mem- dose of 30 mg/kg. The ketoamide 5 exhibited the
brane permeability and low lipophilic nature. longest t1/2 value and the highest concentration
Among these compounds, ketoamide 5 showed at 8 h after oral administration (C8h  0.11 M)
the most favorable PK profile. The retinal AUC among these compounds. Therefore, 5 is the most
value of 5 was approximately equal to that of favorable compound as a candidate for the treat-
hemiacetal 3. Furthermore, ketoamide 5 demon- ment of chronic retinal diseases.
strated the longest retinal half-life (t1/2) and the
highest concentration at 8 h after oral adminis-
tration (C8h). Ketoamide 5 incorporates methoxy
diethylene glycol (mDEG) moiety as a nonionic
CONCLUSIONS
amphiphile at the P3 site and exhibits adequate
In conclusion, we evaluated pharmacokinetic
lipophilicity. On the other hand, structurally re-
properties in the retina after oral administration
lated ketoamide 6 has a pyridineethanol as a ba-
of the several classes of calpain inhibitors, hemi-
sic water-soluble moiety at the P3 site and pos-
acetal 3, ketoamide 5, and ketoamide 6, to obtain
sesses a similar lipophilicity to 1. It has been
an oral drug for the treatment of retinal disorders.
reported that the introduction of a basic moiety
This study confirmed that these compounds have
led to an increase in plasma half-life and volume
remarkably higher penetration into the retina af-
of distribution through a charge-charge interac-
ter oral administration than original aldehyde 1.
tion between the protonated base and a region of
Ketoamide 5, containing an amphipathic moiety,
negative charge in the membrane phospho-
had excellent oral BA, high retinal penetration,
lipids.30 However, incorporation of basic pyri-
and a long retinal half-life. Comparisons of 5 with
dine moiety resulted in lower Vss, retinal pene-
pyridine ketoamide 6 revealed that the mDEG
tration, and larger, shorter half-life. These
moiety at the P3 provided the good PK proper-
unfavorable results may be caused by strong
ties in plasma and retinas. Ketoamide 5 would
plasma protein binding owing to its high
show the retinal efficacy in various retinal disor-
lipophilic nature, leading to active elimination
ders in lower oral dosage, compared to 1 and a
from the retina by efflux transporters in BRB,
long duration of action. In future work, we will
such as P-gp.27 Comparisons of ketoamides 5 and
assess ketoamide 5 as the candidate for further
6 revealed that mDEG moiety at the P3 site of 5
development in adequate pharmacological mod-
played a crucial role for well penetrating into the
els, which reflect retinal diseases.
retina and sustaining the high retinal drug levels
through the addition of the adequate lipophilic-
ity to the dipeptidyl ketoamide backbone.
This study disclosed that orally bioavailable in- ACKNOWLEDGMENTS
hibitors 3, 5, and 6 showed higher retinal pene-
tration than original aldehyde 1. Compound 1 The authors would like to thank Drs. Yukuo
was detected at levels 0.1–0.6 M in the retina fol- Yoshida and Mitsuyoshi Azuma for reading the
lowing an oral dose of 500 mg/kg, which is the manuscript. In addition, we thank Noriko Nada
minimum effective dose in the rat retinal is- for supporting laboratory work. Also, we would
chemia model. The potencies of these inhibitors like to thank Drs. Yutaka Kawamatsu, Jun Inoue,
were comparable to that of 1 (Table 1). If the reti- and Masayuki Nakamura for useful discussions.
424 SHIRASAKI ET AL.

REFERENCES 20. Raghava, S., Hammond, M., and Kompella, U.B. Pe-
riocular routes for retinal drug delivery. Exp. Opin.
1. Branca, D. Calpain-related diseases. Biochem. Biophys. Drug Deliv. 1:99–114, 2004.
Res. Commun. 322:1098–1104, 2004. 21. Fukiage, C., Azuma, M., Nakamura, Y., et al. SJA6017,
2. Ono, Y., Sorimachi, H., and Suzuki, K. In Wang, A newly synthesized peptide aldehyde inhibitor of
K.K.W., and Yuen, P.-W., eds. Calpain: Pharmacology calpain: Amelioration of cataract in cultured rat
and Toxicology of Calcium-Dependent Protease, Philadel- lenses. Biochim. Biophys. Acta. 1361:304–312, 1997.
phia, PA: Taylor & Francis, 1999; 1–3. 22. Inoue, J., Nakamura, M., Cui, Y., et al. Structure-ac-
3. Huang, Y., and Wang, K.K. The calpain family and tivity relationship study and drug profile of N-(4-flu-
human disease. Trends Mol. Med. 7:355–362, 2001. orophenylsulfonyl)-L-valyl-L-leucinal (SJA6017) as a
4. Wang, K.K.W., and Yuen, P.-W. Calpain inhibition: potent calpain inhibitor. J. Med. Chem. 46:868–871,
An overview of its therapeutic potential. Trends Phar- 2003.
macol. Sci. 15:412–419, 1994. 23. Shirasaki, Y., Nakamura, M., Yamaguchi, M., et al. Ex-
5. Stracher, A. Calpain inhibitors as therapeutic agents ploration of orally available calpain inhibitors 2:
in nerve and muscle degeneration. Ann. N. Y. Acad. Peptidyl hemiacetal derivatives. J. Med. Chem.
Sci. 884:52–59, 1999. 49:3926–3932, 2006.
6. Yuen, P.-W., and Wang, K.K.W. Therapeutic poten- 24. Nakamura, M., Yamaguchi, M., Sakai, O., et al. Explo-
tial of calpain inhibitors in neurodegenerative disor- ration of cornea permeable calpain inhibitors as anti-
ders. Exp. Opin. Invest. Drugs 5:1291–1304, 1996. cataract agents. Bioorg. Med. Chem. 11:1371–1379, 2003.
7. Zatz, M., and Starling, A. Calpains and disease. N. 25. Shirasaki, Y., Miyashita, H., Yamaguchi, M., et al. Ex-
Engl. J. Med. 352:2413–2423, 2005. ploration of orally available calpain inhibitors: Pep-
8. Sakamoto, Y., Nakajima, T., Fukiage, C., et al. In- tidyl -ketoamides containing an amphiphile at P3
volvement of calpain isoforms in ischemia-reperfusion site. Bioorg. Med. Chem. 13:4473–4484, 2005.
injury in rat retina. Curr. Eye Res. 21:571–580, 2000. 26. Shirasaki, Y., Miyashita, H., and Yamaguchi, M.
9. Oka, T., Tamada, Y., Nakajima, M., et al. Presence of Exploration of orally available calpain inhibitors
calpain-induced proteolysis in retinal degeneration 3.:Dipeptidyl -ketoamide derivatives containing pyri-
and dysfunction in a rat model of acute ocular hy- dine moiety. Bioorg. Med. Chem. 14:5691–5698, 2006.
pertension. J. Neurosci. Res. 83:1342–1351, 2006. 27. Hughes, P.M., Olejnik, O., Chang-Lin, J.E., et al. Top-
10. Sharma, A.K., and Rohrer, B. Calcium-induced calpain ical and systemic drug delivery to the posterior seg-
mediates apoptosis via caspase-3 in a mouse photore- ments. Adv. Drug Deliv. Rev. 57:2010–2032, 2005.
ceptor cell line. J. Biol. Chem. 279:35564–35572, 2004. 28. Steuer, H., Jaworski, A., Elger, B., et al. Functional
11. Paquet-Durand, F., Azadi, S., Hauck, S.M., et al. Cal- characterization and comparison of the outer blood-
pain is activated degenerating photoreceptors in the retina barrier and the blood-brain barrier. Invest. Oph-
rd1 mouse. J Neurochem. 96:802–814, 2006. thalmol. Vis. Sci. 46:1047–1053, 2005.
12. Donkor, I.O. A survey of calpain inhibitors. Curr. Med. 29. Meyer, S.L., Bozyczko-Coyne, D., Mallya, S.K., et al.
Chem. 7:1171–1188, 2000. Biologically active monomeric and heterodimeric re-
13. Wells G.J., and Bihovsky, R. Calpain inhibitors as po- combinant human calpain I produced using the bac-
tential treatment for stroke and other neurodegener- ulovirus expression system. Biochem. J. 314:511–519,
ative disease: Recent trends and developments. Exp. 1996.
Opin. Ther. Patents 8:1707–1727, 1998. 30. Alker, D., Campbell, S.F., and Cross, P.E. Long-acting
14. Lee, K.S., Franks, S., Vanderklish, P., et al. Inhibition dihydropyridine calcium antagonists. 6. Structure-ac-
of proteolysis protects hippocampal neurons from is- tivity relationships around 4-(2,3-dichlorophenyl)-3-
chemia. Proc. Natl. Acad. Sci. USA 57:7233–7237, 1991. (ethoxycarbonyl)-2-[(2-hydroxyethoxy)methyl]-5-
15. Rami, A., and Krieglstein, J. Protective effects of cal- (methoxycarbonyl)-6-methyl-1,4-dihydropyridine. J.
pain inhibitors against neuronal damage caused by Med. Chem. 34:19–24, 1991.
cytotoxic hypoxia in vitro and ischemia in vivo. Brain 31. Shirasaki, Y., Miyashita, H., Nakamura, M., et al. PCT
Res. 609:67–70, 1993. Int. Appl. WO2005056519, 2005.
16. Hong, S.-C., Goto, Y., Lanzino, G., et al. Neuropro-
tection with a calpain inhibitor in a model of focal Received: April 20, 2006
cerebral ischemia. Stroke 25:663–669, 1994. Accepted: July 19, 2006
17. Markgraf, C.G., Velayo, N.L., Johnson, M.P., et al. Six-
hour window of opportunity for calpain inhibition in Reprint Requests: Yoshihisa Shirasaki
focal cerebral ischemia in rats. Stroke 29:152–158, 1998.
Pharmacokinetic Group
18. Bartus, R.T., Hayward, N.J., Elliott, P.J., et al Calpain
inhibitor AK-295 protects neurons from focal brain
Laboratory for Preclinical Research
damage. Stroke 25:2265–2270, 1994. Senju Pharmaceutical Co., Ltd.
19. Neuhof, W., Götte, O., Trumbeckaite, S., et al. A novel 1-5-4 Murotani, Nishiku
water-soluble and cell-permeable calpain inhibitor Kobe, Hyogo 651-2241
protects myocardial and mitochondrial function in Japan
postischemic reperfusion. Biol. Chem. 384:1597–1603,
2003. E-mail: shirasaki@senju.co.jp

You might also like