13
カルボキシルエステラーゼ活性を利用したプロドラッグの開 誌名 誌名 Journal of pesticide science ISSN ISSN 1348589X 巻/号 巻/号 353 掲載ページ 掲載ページ p. 229-239 発行年月 発行年月 2010年8月 農林水産省 農林水産技術会議事務局筑波産学連携支援センター Tsukuba Business-Academia Cooperation Support Center, Agriculture, Forestry and Fisheries Research Council Secretariat

カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

カルボキシルエステラーゼ活性を利用したプロドラッグの開発

誌名誌名 Journal of pesticide science

ISSNISSN 1348589X

巻/号巻/号 353

掲載ページ掲載ページ p. 229-239

発行年月発行年月 2010年8月

農林水産省 農林水産技術会議事務局筑波産学連携支援センターTsukuba Business-Academia Cooperation Support Center, Agriculture, Forestry and Fisheries Research CouncilSecretariat

Page 2: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

11111111111111111111111111111

Review 11111111111111111111111111111

(Special Topic)

J. Pestic. Sci., 35(3), 229-239 (2010) DOI: 10.1 584/jpestics.R1 0-03

Prodrug approach using carboxylesterases activity:

catalytic properties and gene regulation of carboxylesterase in mammalian tissue

Teruko IMAI* and Masakiyo HOSOKAWA t

Graduate School 01 Pharmaceutical Sciences, Kumamoto Uniνersity, 5-1 Oe-honmachi Kumamoto, 862-0973, Japan t Faculty 01 Pharmaceutical Sciences, Chi加 Institute01 Science, 15-8 Shiomi-cho, Choshi, Chiba, 288-0025, Japan

(Received April1, 2010; Accepted June 1,2010)

A prodrug is a pharmacologically inactive d巴rivativeof an active parent drug, and is bioconverted to the active

drug in vivo. Through chemical modification of a drug to a prodrug, we are able to deliver drugs to the target site, to optimize therapy and minimize toxicity. A m司jorpathway for the bioconversion ofprodrugs to the active par-

ent drugs is via carboxylesterase (CES) activity. Among human CES isozymes, hCEl and hCE2 predominantly participate in the hydrolysis of prodrugs in the liver and small int巴stine,respectively, although the substrate specificity is quite different between two isozymes; therefore, we can rationally design prodrugs based on the en-

勾 mecharacteristics. However, since the expression levels of CES vary among individuals, there is a range of

pharmacological responses following prodrug administration. Species differences are caused by tissue-dependent

hydrolase activity mediated by CES, which makes it difficult to predict effectiveness in humans from a preclini-

cal study using animals. Accordingly, understanding the regulation of CES expression and species difference of CES catalytic properties will be h巴lpfulin the design of prodrugs with increased specificity and enhanced

physicochemical and biological properties. <C) Pesticide Science Society of Japan

Keywords: prodrug, carboxylesterase, subs仕atespecificity, species difference, gene regulation.

Introduction

In the current drug development paradigm, it is necess紅 Yto

design compounds with minimal or no side effects, and to specifically combat a target disease. The conversion of a drug

to a prodrug that is pharmacologically inactive, but can be-

come actlve Vla an enzymatlc reactlOn, IS an lmportant s仕at同

egy in targeting a drug to the site of action. By transformation

of the drug to its prodrug, we can minimize or eliminate pos・

sible drug toxiciザ tooptimize therapy. Chemotherapy using

prodrugs was developed in the 1970s, and several prodrugs are now in clinical use. This approach is still accepted as an

integral part of new drug design processes, because we can improve, delay, prolong, con仕01,and specifically express the

action of the p紅 entdrug using a prodrug.1,2)

Prodrugs are mostly ester derivatives which are constructed

* To whom correspondence should be addressed. E-mail: [email protected] Published online July 15, 2010 (T Pesticide Science Society of Japan

企omhydroxyl and carboxyl groups of the parent drug, be-

cause they can be enzymatically converted to parent drugs by

hydrolases that widely exist in vivo. Carboxylesterase (CES,

EC 3 .l.l.I) is responsible for the activation of ester and

amide prodrugs,3) belongs to a super-family called the α,s-hydrolase-fold family, and is a member of the serine esterases, which are found in various mammalian tissueρ) Mammalian

CESs comprise a multigene family, and their isozymes紅巳

classified into five fundamental groups based on the homol-

ogy of the amino acid sequence.6) CESl and CES2 families

play a major role in the bioconversion of prodrugs. The ex-

pression levels of CESs and their tissue dis仕ibutionprofiles

affect the fate of prodrugs in the body.7,8) Furthermore, CES

isozymes have inter-related subs仕atespecificity but may be

classified according to their hydrolase activities towards se・

lected subs回 tes;therefore, the prodrug can be rationally de-signed on the basis of the characteristics of human CES

isozymes. For example, oseltamivir and temocapril are metab問

。lizedin the liver by human CES 1 but not by human CES2 in

the small intestine.9,1O)

In the design of prodrugs specifically susceptible to CES, it

Page 3: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

specific tissue should be sufficiently converted at the fastest

rate of k4 at the target site, compared with other・prodrugcon-

version rates (kj to k3). When the bioavailable dose of a

poorly absorbed drug is increased by administration of a suit-

able prodrug, its kinetics should be optimized to increase drug delivery to the systemic circulation. 1n this case, prodrug con-

version takes place prior to arrival in the blood. The rate of

prodrug conversion should be adequately fast, relative to the metabolism of the prodrug and parent drug. Prodrug excretion

and metabolism cause a decrease in drug yield. Also, the

slower conversion rate of the prodrug than the metabolism of

the parent drug causes a decrease in drug yield due to sequen-

tial metabolism ofth巴parentdrug, and thus a reduction in the

potential bioavailable dose of the drug. Figure 2A shows the

typical blood concentration profile of a poorly converted proω

drug. 1n order to obtain a high blood concentration of the par胸

巴ntdrugラ asshown in Fig. 2B and C, orally administered pro胎

drug should be extensively converted to the parent drug in the

intestine and liver through which they first pass before enter-

ing the systemic circulation. If the prodrug is completely con-

verted to the parent drug during the first pass, then the phar-

macodynamics and toxicity all depend on the original drug,

provided that the disposable moiety is inert.

Journal of Pesticide Science

is important to evaluate the affinity of a prodrug to the corre-

sponding CES as well as individual variations in the CES ex-

pression level at the target tissue. CES activity may show

inter-individual variations due to both genetic polぅrmorphisms

as well as environmental factors, and these may influence tox-

icity induced by the parent ester.jI.l

2) Furthermore, the effec-

tiveness and safety of prodrugs should be confirmed in a pre-

c1inical study using animal and cell culture models. The tissue

distribution of CESl and CES2 isozymes differs among ani-

mals, inc1uding humans, and their variation leads to species

differences in tissue-sp巴cifichydrolase activity. It should also

be noted that substrate specificity differs among orthologous

CES isozymes in various species.

The present review discusses the development of prodrugs,

which is an important issue in the design of a new drug, fo-

cusing on the varying substrate specificity of human CES

lsozymesラ speciesdifferences in CES-sp巴cifictissue hydroly側

sis, and the genomic structure and regulation of CES genes.

T. lmai and M.日osokawa230

First reachcd organ

General description of prodrug pharmacokinetics

Figure 1 shows the bioconversion proc巴sseswhich may follow

the oral administration of a prodrug. As indicated by the conω

version rate constants, the prodrug is converted to the parent

drug at the absorption site (kj), in the liver (k2), in the blood

(k3), and other distribution sites in the body (k4). The concen-tration profile of th巴 prodrugand parent drug depends upon

the aim of the prodrug. For example, a prodrug delivered to a

1.

;liAJiと¥Poor h¥"drol刊 ;s

:"iot hydrolyzcd

囚mwzm匂匂'M匂剖

2a市mg匂mw

制諸国ロ'M向》』

cwg自@償問〈

i ExtcnsI¥'c hydrolysis

Time

:'¥ot hydrolyzcd

i Time

nomorc hydrolysis, bccauscor abscllceof

Int:lct prodrug

Time

-iJaa宮hSam-喜a

B一吉田三

2350E〈

き吉

て¥

Fig. 2. Drug amount at administ巴redsite and blood after adminis-

tration of prodr・ug.明rhenprodrug is oral1y administered, the intestine

is the administered site and the first organ reached is the liver. In cu-

taneous and intravenous administration, the administered site is the

skin and veins, respectively, and the first organ reached is the lung in

both cases 一一一一一, prodrug; 一一一, parent drug; 山内山,

metabolit巴ofprodrug (Metabolite A); -一一也勺 metaboliteof parent

drug (Metabolite B).

Time

E副高山計匹歪~

I Parenl Drug卜一一一一ヶ+げ官.t油 olite8ト一一ー参ーミ;:Laom--

Fig. 1. Distribution of prodrug after oral administration and bio-

conversion ofprodrug.

Page 4: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

Vol. 35, No.3, 229-239 (2010)

2. Prodrug Conversion Mediated by CES

in the Intestine and Liver

Most drug-metabolizing enzymes present in the liver are also

found in the small intestine; however, their levels are gener-ally much lower in the small intestine. The importance of

small intestinal metabolizing enzymes aris巴sfrom th巴 loca-

tion of this organ, which can result in reduced systemic up-take of drugs. The small intestine and liver play a significant

role in the metabolism of oral xenobiotics and drugs.

Hydrolase activity in the liver and small intestine in mam-

mals is attributable to several巴sterasemolecules.13,14) The

content of CES in rat liver is found to be about lmg per g of

fresh tissue while the microsomal fraction contains about 30

mg CES per g of microsomal protein.15) 1n the liver and intes剛

tine, CES 1 and CES2 isozymes are present, and play critical roles in prodrug bioconversion. 1n humans, CES2 isozyme,

hCE2, predominantly presents in the small intestine, and the hydrolysis pattern for several substrates in th巴 humansmall

intestine microsomes is nearly the sam巴 asthose of recombト

nant hCE2.16) Although human liver microsomes express both

CESl isozym巴(hCE1)and hCE2, the hepatic substrate speci-ficity c10sely resembles recombinant hCEl. Fur‘thermore, the

anti-hCEl antibody showed 80-95% inhibition ofhepatic hy-

drolysis, and the residual hydrolase activity is due to hCE2.6)

hCEl dominantly contributes to hepatic hydrolysis rather than

hCE2; thus, the first-pass hydrolysis of the prodr・ugdep巴nds

on the activity of hCEl and hCE2 in the liver and intestine,

respectively.

日owever,both hCE 1 and hCE2 in the liver are inter酬indi-

vidually variable to a great extent. Furthermore, an age-de輪

pendent expression was observed. 1n general, the adult human

liver expresses significantly higher hCEl and hCE2 than the

pediatric liver, which shows significantly higher expression

than the fetal group.12) Within the pediatric group (0-10

y巴ars),the hydrolysis of oseltamivir varies by 127-fold in agreement with the variation in the abundance of hCE 1 Y) 1n adults,日osokawaet al. reported a more thanふfoldrange of

variance in hCEl protein levels among 12 human liver micro-

somes,17) and Xu et al. reported a 3-fold range of variance for

hCE2 among 13 human liver microsomesI8); thereforeヲ the

prodrug should be design巴dby considering the variatior

3. Intestinal Hydrolysis during the Process of

Absorption

Most prodrugs aimed to improve oral bioavailability of their

parent drugs possess adequate membrane perm巴abilitydue to

increasing lipophilicity by ester formation.19) Thereforeラ the

prodrug is easily taken up into epithelial cells and hydrolyzed

to the parent drug, as shown in Fig. 3. The parent drug con-verted from the prodrug is present at the highest concentra-

tion in epithelial cells, and can be transported by passive and

active transport into not only blood vessels but also the intes-

Prodrug approach using carboxyl巴steraseactivity 231

Blood vessel

Fig. 3. Absorption and hydrolysis of prodrug in the intestinal ep-ithelial cell.

tinal lumen. Therefore, it is difficult to achieve complete drug

absorption. A prodrug hydrolyzed extensively in epithelial

cells results in lower blood concentration of the parent drug,

as shown in Fig. 2C. We studied the relation beれNeenpr・odrug

absorption and its mucosal hydrolysis using in SItll rat singl巴"

pass perfusion.20.21) 1sovaleryl propranolol (isovaleryl-PL) was

well absorbed by epithelial cells by passive diffusion, and then was completely hydr叫yzedto PL and isovaleric acid at

the rate limited by the uptake of isovaleryl側 PLinto mucosal

cells. The produced PL and isovaleric acid were passively

transported to the luminal side and blood vessels according to

the pH-partitioning th巴ory.PL, a basic drug, was transported into intestinal lumen (pH 6.5), and isovaleric acidラ anacidic

compound, was absorbed into blood vessels (pH 7.4).21) 1n

contrast, when intestinal CES was inhibited by a specific in-

hibitor, hydrolysis of isovaleryl-PL during absorption was in-

hibited by about 80%, resulting in increased absorption of the intact prodrug.21) Okudaira et a1.22

) reported that an ester舗 type

prひdrug,ME3229, is taken up into mucosal cells at a rate

compatible with its lipophilicity, and th巴ncompletely hy-

drolyzed. The p昌rentdrug produced in mucosal cells was pre柳

dominantly pumped out by an active effiux transporter. When

a prodrug is scarce1y hydrolyzed in the human small intestine, it is transported into blood vessels in an intact form. 1f an inω

tact prodrug is taken up into the liver and then rapidly hy附

drolyze札itshows an ideal blood concentration profile, as shown in Fig. 2B. Thus, intestinal hydrolysis is markedly im-portant in determining prodrug bioavailability. Extensive hy-

drolysis degrades a prodrug intended to improve intestinal

membrane permeability of the drug. It is therefore necessary

to consider the hydrolysis susceptibility of prodrugs and the

transport properties of the parent drug in a prodrug design.

4. Substrate Specificity of Human CESl and CES2

Them民jorhydrolase in the human liver and small intestine is

hCEl and hCE2, respectively. hCEl and hCE2 exhibit 48% homology, and their distinct substrate sp巴cificityhas been re-

ported.5.7,16) The m司jorintestinal CES, hCE2, mainly hy-

drolyzes prodrugs into which an alcohol group of a pharma-

cological active drug is modified with a small acyl group.5,7)

Prodrugs grouped in this category inc1ude CP下1123)and as-

Page 5: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

232 T. lmai and M. Hosokawa

pirin.24) In contrast, prodrugs into which the carboxyl group

ofthe phaロnacologicallyactive drug is modified with a small

a1cohol group are preferentially hydrolyzed by hCEl, and

have been numerously developed as pharmaceutical medi-

cines, for example, oseltamivir,9) meperidine,25) capecitabine,26) oxybutynin,27) camostat mesilate28) and angiotensin-convert-

ing 巴回yme(ACE) inhibitors29) such as temocapril and

enalapril. A number of successful prodrugs are stable in the

human intestine and rapidly hydrolyzed in the liver; therefore, prodrugs can be designed by taking advantage of the

markedly different subs仕atespecificity between hCEl and

hCE2.

Thus, the distinct subs仕atespecifici匂rof hCEl and hCE2

Good substrate for

hCE2

II-OH

-ー/'

.n.....CH2

~3

Hヘ A60F-oτH-Lh:2

JZA匂

Journal 01 Pesticide Science

might be related to the s仕uctureoftheir reaction sites. Cataly-

sis of ester c1eavage by CES is achieved via a triad of catalytic

amino acids (Ser203, His450 and Glu336). Carboxylesterases

c1eave the ester via a阿 o-stepreaction, as shown in Fig. 4. At neutral pH, the active site, Glu336, exists as the charged form, which facilitates the removal of a proton from His. This loss

subsequently results in transfer of a proton仕omthe adjacent

Ser203 to the opposing nitrogen of His, generating an oxygen nuc1eophile that can attack也ecarbonyl carbon of the sub-

strate. The hydrogen bonds between the negatively charged

oxygen of the tetrahedral intermediate and the NH group of

Gly123 and Gly124 then stabilize the negatively charged oxy-

gen. This configuration, in which the negativ巴lycharged car-

Hヘ A<f'C-O:"'H-~\wE03

/ι0

/ryo-ce

Fig. 4. Hydrolyzing mechanism ofCES.

Page 6: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

Vol. 35, No.3, 229-239 (2010)

boxyloxygen is hydrogen bonded to阿 10NH groups, is called an oxyanion hole. When the tetrahedral intermediate of an

acyl group is formed,自己 a1coholproduct is releas巴d企omthe

en可me.The acyl-enzyme intermediate is then attacked in an

identical fashion with water acting as the nuc1eophile, leading to release of the carboxylic acid and return of the catalytic

amino acids to their original state.

Steric hindrance in the vicinity of the reaction site of hCE2

may occur with substrates containing a bul匂1acyl moiety in

the process ofthe formation ofthe acyl-hCE2 intermediate in

the first step of hydrolysis.16) Interestingly, PL derivatives,

generally good subs仕atesfor hCE2, are irregularly hy-

drolyzed by hCE2 depending on the structure of the acyl

group. PL derivatives substituted by 3-methyl acyl group are

scarcely hydrolyzed by recombinant hCE2, while propranolol derivatives with 2-methyl acyl groups are巴asilyhydrolyzed at

almost the same rate as the corresponding s仕aightacyl deriv-

atives.16) In general, the chemical hydrolysis of ester bonds is

sterically hindered by the substituent methyl group at the 2-

position rather than the 3-position. Both findings, specific re-duction of the hydrolysis rate by substitution of a methyl

group at the 3・positionand the low hydrolysis rate for a sub-

strate with a large acyl group, suggest that the acyl-enzyme intermediate is difficult to form due to the presence of steric

interference in the active site region ofhCE2.

In con位astto hCE2, hCE1 preferentially recognizes a sub-

strate with a large acyl moiety, and also catalyses the hydroly-

sis of prodrugs modified with a small acyl group even with

limited activity. The substrate-binding site of hCE1 consists

of a “small, rigid" pocket and a “large, flexible" pocket,29,30)

and these pockets allow hCEl to act on structurally distinct

compounds containing either large or small a1cohol moieties.

The distinct active site between hCEl and hCE2 might be

caused by different amino acid sequences, especially the lack

of a loop structure consisting of 15 amino acids

Interestingly, hCE1 enantioselectively catalyzes the hydrol-ysis of a substrate. For example, S-PL derivatives,16) S-co-caine,31) d-methylphenidate32

) and cis-cypermethrin ana-

logues33) are poor substrates of hCE1, in contrast to the op-

posing enantiomer. The differences in the hydrolysis rate be-

tween these enantiomers have been explained b

Prodrug approach using carboxy1esterase activity 233

(AF036930) and dog CES Dl (AB023629) show 92.9, 81.1 and 79.7% homology with hCE1, respectively. The rat CESl family inc1udes four isozymes, Hydrolase A (ES10; X5 1 974), Hydrolase B (X81825), Hydrolase C (RL1; Ul0698) and rat

egasyn (X81395), and the mouse CES1 family inc1udes at

least three isozymes, Es-x (Y12887), mouse CES mMHl

(AB023631) and mouse egasyn (S80191). Thus, a number of

CES 1 isozymes have been identified as proteins. In contrast, few CES2 isozyme have been identified as proteins, such as

rabbit CES2 (P14943), mouse mCES2 (ML3; BC031170) and

two rat m司jorCES2 isozyme, rCES2 (RL4; AB010635) and

AY034877.

It is expected that orthologous isozymes wi11 show similar subs仕atespecificity, because of their 60-95% homology. For example, mouse mCES1, a mouse CES1 family isozyme, hy-drolyzes temocapril, similar to human hCEl,35) and rat CES1

isozyme (Hydrolase A and Hydrolase B) hydrolyzes

deltamethrin and esfenvalerate, such as human hCEl.36) F町・

thermore, rat rCES2, a rat CES2 family isozyme, hydrolyzes methylprednisolone hemisuccinate, as does human hCE237l; however, these examples are limited. In most cases, subs仕atesare hydrolyzed with markedly different affinity among the

same CES family isozymes. l-RS cis-permethrin is hy-

drolyzed by CES1 isozyme of rats and rabbits, but not by human hCE1, although l-RS trans-permethrin is hydrolyzed by rabbit, rat and human CES1 isozymes.38

) Rabbit CES1 is

100-to 1000-fold more efficient at converting CPT-ll to SN・

38 than human hCEl.39) Thus, the variation of hydrolase ac-

tivity among the same CES family depends on the substrate, and it is difficult to predict the affinity of the isozyme to the

substrate.40)

Furthermore, the expression level is an important factor in

tissue hydrolase activity. In order to c1arifシtheexpression of

the CES family in several tissues, CES1 and CES2 levels were measured by Northem blots, RlユPCRand real-time

PCR.8) The human hCE1 is highly expressed in the liver, lung

and other tissues, but human hCE2 is limitedly expressed in the small intestine and kidney at a high level. In most animals, CES 1 isozyme is highly expressed in the liver and lung, and CES2 is present in the small intestine; however, in the kidney, only CES 1 is expressed in the rat and hamster, and both CES 1

and CES2 are present in the mouse, although CES2 is prefer-entially expressed in humans, monkeys and dogs. Interest-ingly, no CES isozyme is present in the dog

Page 7: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

Journal 01 Pesticide Science

tration of intact prodrug and markedly high concentration of

PL were detected in dog plasma due ωfirst四passhydrolysis by

CES D 1 in the lung. In rats, higher plasma concen仕ationof

intact prodrug than PL was observed a丘巴rintravenous admin-

istration of isovaleryl♂L, because it was hydrolyzed in the

liver and blood, but not in the lung叩 Thus,first-pass hydroly-

sis in the lung and small intestine results in markedly different

pharmacokinetics between rats and dogs. CES is dis仕ibuted

in almost all organs of the body; therefore, hydrolysis of the

prodrug in the administered site and the first reached organ

significantly affects the fate of prodrugs.

T. lmai and M. Hosokawa 234

Gene Structure and Regulation of CES Isozymes

The CES genes comprise a multigene family and isozymes

are c1assified into at least five groups (CESI-CES5) and sev-

eral subgroups according to the homology of the amino acid

sequence.4,47) Genomic s仕ucturesof the genes encoding these

enzymes have been determined: CESl genes are located on

chromosome 16 containing 14 exons and span about

30Kb48,49) and CES2 genes are also located on chromosome

16 containing 12 (15) exons and span about 11 kb.50,51) Re-

centl弘同10CESl genes, CESIAl (AB119997) and CESIA2 (AB119998) have been identified in the human genome.47)

Both genes reside in chromosome 16q 13-q22.1戸53)in a tail-

to-tail manner, separated by about a 9 kb intergenic region.

The exon-intron structure is totally conserved between the

two genes and the homology of the exon and promoter re-

gions is 98% and 91 % at the nuc1eotide level, respectively Tables 1 and 2 shows the known or predicted CESl and

7.

Carboxylesterase (CES1) Genes and Enzymes Examineda) Table 1.

Strand Chromosome NO.of GenBank mRNA CES CES

Mammal location Amino acids (or *N-scan ID) family gene

Negative

Negative

16 567 L07765 CESl CESl Human

16 567 *16.56.002 CESl CESl Chimp

Oran伊 tan

Baboon

566 CR857194 CESl CESl

567 CESl CESl

Negative

Negative

20 566 *20.55.002 CESl CESl Rhesus

18 558 NP031980 CESl CESl Cow

566 X63323 CESl EST1 旧

M

EEDC

Positive 565 AB023629 CESl CESl

Negative

2

566 AB114676 CESl CESl

Negative

Negative

Negative

Negative

Negative

565 AF036930 CESl EST1 Rabbit

19 565 X51974 CESl CES3 Rat

8 565 NP067431 CESl CESl Mouse

8 565 NM053200 CESl CES3 Mouse

8

a) Ref. 54

activity will differ among several species. The different tissue

hydrolase activity causes diverse pharmacokinetics of the pro-

drug among species. Oseltamivir exhibits good oral bioavail-

ability compared to the parent acid form in most experimental

animals and humans. In humans, oseltamivir is rapidly ab-

sorbed and almost completely hydrolyzed to the active form

by hCEl in the liver after oral administration;42) however, os-巴ltamiviris hardly hydrolyzed by rat CES 1 isozymes in the

liver, but easily hydrolyzed in the blood. Although oseltamivir is extensively absorbed in rats after oral dosing, hydrolase ac-tivity in blood is not sufficient to achieve complete conversion

of oseltamivir to the active foロn;43)therefore, another metabo-lites mediated by cytochrome P450 were observed in the

blood and liver in rats.44) The blood concentration profile in

Fig. 2A is similar to that after administration of oseltamivir in

rats.

We reported species difference in the pharmacokinetics of

propranolol (PL) derivatives after oral and intravenous admin-

istration.45) When isovaleryl開PLwas orally administ巴redto

dogs, significantly higher plasma concentration of PL was ob-served than that following adminis仕ationof PL (parent drug).

Hydrophobic isovaleryl・PLis easily absorbed as an intact

prodrug企omdog intestine due to a lack of esterase and com-

pletely converted to PL in the liver, resulting in high plasma concen仕ationof PL, such as in Fig. 2B. However, rats showed nearly the same plasma PL concentration as oral dosing of

PL, because ofthe complete hydrolysis ofisovaleryl-PL in the rat intestine,21,45) such as in Fig. 2C. V.司lenisovaleryl-PL was

intravenously administered to dogs, a negligible low concen-

8

562

551

NP598421

NP031980

CESl

CESl

CES22

CESN

Mouse

Mouse

Page 8: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

Vol. 35, No.三229-239(2010) Prodrug approach using carboxylesterase activity 235

Table 2. Carboxylesterase (CES2) Genes and Enzymes Examined叫

CES CES GenBa対cmRNA NO.of Chromosome Mammal Strand

gene family (01・ホN-scanID) Amino acids location

Human CES2 CES2 BX538086 559 16 Positive

Chimp CES2 CES2 本20.66.008 559 16 Positive

Baboon CES2 CES2 561

Rhesus CES2 CES2 京20.66.008 561 20 Positive

Cow CES2 CES2 BCI02288 533 18 Positiv巴

Rabbit EST2 CES2 P14943 532

Hamster CES2 CES2 D28566 561

Hamster CES6 CES2 D50577 559

Rat CES2 CES2 ABOI0632 560 Positive

Rat CES2.1 CES2 ABOlO635 561 Positive

Rat CES6 CES2 AY034877 558 19 Negative

Mouse CES2 CES2 NP663558 561 8 Positive

Mouse CES5 CES2 BC055622 559 8 Positive

Mouse CES6 CES2 NP598721 558 8 Positive

α) Ref. 54

CES2 gene locations of three primate species and four non-

primate eutherian mammals based upon published reports,

and BLAT interrogation of human, chimp, rhesus, mouse, rat,

cow, dog and cat genom巴S.54)With the exc巴ptionof rat CES2

and CES3 genes, which are located on chromosomes 1 and

19, respectively, CESl and CES2 genes fr・omthe other exam-

ined five mammalian genomes were syntonic. 1n addition, 10

of 11 mammalian CESl genes were transcribed on the negか

tive strandラ and9 of 10 CES2 genes were transcribed on the

positive strand.

Recently, we indentified and characterized dexamethasone-induced methylprednisolone hemisuccinate (MPHS) hydro-

lase in rat liver microsomes. 1ntraperitoneal injection of

dexamethasone resulted in a significant increase in the level

of MPHS hydrolase activity accompanied by the induction

of a specific CES isozyme, AB0106353.5) To confirm

that AB010635 encodes the dexamethasone-induced CES

isozyme, cDNA cloning was performed and the obtained

cDNA was expressed in Sf9 ce11s using a baculovirus-medi-

ated expression system. The recombinant CES pr悦巴incould

hydrolyze MPHS and exhibited biochemical characteristics

similar to those of CES RL4. Co11ectively, the results indi-

cated that dexamethasone-induced MPHS hydrolase in liver・

mlcrosom巴sis a rat CES2 isozyme. Interestingly, the results

also showed that this rat CES2 isozyme exists in plasma and

that the amount ofthis protein is increased by dexamethasone.

The 5'-ftanking regions of CESl and CES2 genes were おか

lated 企ommouse, rat and human genomic DNA by PCR am-

plification. The mouse CES gene (mCESl), rat CES gene

(rCESJ) and two individual human CES genes (CESIAl and

lA2) w巴refound to belong to the CES 1 family.55) The mouse

mCES2,56) rat rCES257) and human CES2Al genes were found

to belong to the CES2 family.55) A TATA box does not pre-

cede the transcription start site of any of the CES promoters,

as shown in Figs. 4 and 5. CES promoters share s巴veralcom-

mon binding sites for transcription factors among the same

CES families, suggesting that orthologous CES genes have

evolutiona11y conserved transcriptional regulatory patterns.

Potential binding sites of CES promoters for transcriptional

factors include specificity protein (Sp) 1, Sp3, CCAAT box

binding protein (C/EBP), upstream stimulatory factor (USF)

1, nuclear factor (NF) Y, nuclear factor kappa light chain en-

hancer of activates B c巴l1s(NFkB), peroxisome proliferator

activat巴dreceptor (PPAR), glucocorticoid receptor (GR), and

GR GR議襲警護竺竺 I 亡ニトー GRE- - GC box -ICCMT boxトー ムー

Human CES 1A1 gene

NF-1 NF-Y GR GR

--GCb 一己Rat CES1 gene

GR GR NF-Y C v 謬欝明BP ,--.

一一一 GRE→CCMTbOxr GC box…糊 一GCbox←Mouse Ces 1 gene

Fig. 5. Structur巴 ofth巴 5'fianking region of mammal and human

CESI genes.

Page 9: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

236 T. lmai and M. Hosokawa

- GCbox.Lイ正否box GC box GC boxι

GR GR

開ouseCes2 gcne

GFミ自決

Rat CES2 gene

Fig. 6. Structure of the 5' flanking region of mammal and human CES2 genes

hepatocyte nuc1ear factor (HNF)-4αbinding sites.

We have found that mCES2 is expressed in various tissu巴s

with higher levels of expression in the liver, kidney and small

intestine. lt was shown that three transcription factors, Sp 1, Sp3 and USFl, could bind to the promoter region of th巴

mCES2 gene, leading to synergistic transactivation of the pr・0嗣

江lOter.57)

The mouse CES2 isozyme, mCES2, is thought to play il11-

portant roles in lipid metabolisl11 and is expressed in the liver,

kidney, and small intestine at high levels; therefore, we exam-

ined the molecular・mechanismscontrolling this tissu巴附specific

expression of mCES2. We found that HNF-4αcould enhanc巴

transcription of the mCES2 gene in vitro and in vivo, and its

effect on mCES2 prol110ter activity was repressed by small

heterodimer partner (SHP) and chenodeoxycholic acid

(CDCA) in luciferase assays (Fig. 7). Accordingly, mCES2 gene transcnptlOn was r・epressedby CDCA treatment in

l110use immortalized hepatocytes. The repression of mCES2

gene transcription might result frol11 the combined effects of

both inhibition of the HNF-4αtransactivation ability by SHP

and reduction ofthe HNF-4αexpression level. Thus, HNF-4α

plays an important role in the regulation of mCES2 gene tran-

scription.58)

We have also isolated and characterized two gen巴sencod-

ing human CES1Al (ABlI9997) and CES lA2 (ABlI9998),

CDCA

Fig. 7. Possible model for down-regulation of mCES2 gene tran-scription by CDCA treatment.

JOllrnal 0/ Pesticide Science

and also c10ned and sequenced the 5' ftanking region of each

gene in order to e1ucidate the structure of the promoter.47)

Only six nuc1eotide differences resulted in four amino acid

differences in the open reading frame, and all of the differ欄

ences existed in exon 1. Since exon 1 of the CESl gene en-

codes a signal peptide region, intracellular localization of the CESl gene product was prelil11inarily investigated using邑 sig-

nal peptide/EYFP-ER chil11era pr叫 ein-expressingsystem. It

was interesting that the CESIAl signal peptide/EYFP-ER

chil11era protein was localized to the endoplasmic reticulul11,

whereas the CESIA2 signal peptide/EYFP-ER chimera pro-

tein was distributed in the endoplasmic reticulul11 and cytosol.

These results suggested that CESIAl and CESIA2 have dif国

ferent intracellular localizations and different expression pro-

files in liver differentiation. We therefore investigated the

transcriptional regulation of these two CES genes. Reporter

gene assay and electrophoretic l110bility shift assay demon-

strated that Sp 1 and C/EBPαcould bind to each responsive

element of the CESIAl promoter but Spl and C/EBP could

not bind to the responsive element of the CESIA2 pro鮒

moter.47)

Fukami et al.59) reported that the sequ巴nc巴sof the CES1A2

gene downstr・eamand upstream of intron 1 are identical with

those of the CESIAl and CES1A3 genes, respectively. A

CES1Al variant in which exon 1 is converted with that ofthe

CES1A3 gene (transcript is CESIA2) has been identified. It

was found that the CES1A2 gene is a variant of the CESIA3

pseudogene. The expression level ofCESlAl mRNA is much

higher than that of CESIA2 l11RNA in the liver.47) Since

CESIAl is highly variable in the individual liver,60) it was

thought that these results provided inforl11ation on the inteト

individual variation ofhuman CES1.

For the first time, we reported that DNA methylation is in“

volved in CES1Al gene expression in the hUl11an liver and

kidney.61) The tissue-specific expression of the CES1Al gene

was exal11ined us幻ing5柵-azaか嶋之2'-deoxycyt託idine(β5-aza.駒-dC)and

biおSl叫Ilfit鉛巴 sequ巴ncing.Treatl11ent of HEK293 cells, hUl11an el11-

bryonic kidney cells not expressing the CES1Al gene, with 5-aza-dC caused l11arked expression ofthe CESIAl gene. Bisul-

fite sequencing revealed that the region around the transcri

8. Genetic Polymorphism

Recently, Geshi et al. 62) reported that CES1A2-816A/C poly-

Page 10: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

237

く CES1A1

4109?で132J

Prodrug approach using carboxy1esterase activity

CE剖 A1variant (producing CES1A2] ¥刊

目111111111111L3'361

一A一

521守

1

1

:-S一4

・E』唱E-

c-E一3

・'ιEE

問一

c一2固園町盟

-

。「

L4laz---a

:

'

釦円

Fhu

uH

一句。

2一

-

A内一

-EEE』「BEE-

-

-

-

au-

E

-

-

CE

・E・---;l

E

J

n;1

5

7

:1

d

-

-

同一

51TZ

Eel

il

t

-

-

n

-

-

-812411

29'S品目

E

1

0

1

A「J1HHH日開

HHH

ta--4EE

a

輔、4

0

.

時、一世

E

・TE

3

f

-

1

4圏・・圃圃圃..

1|

nonsynonymous SNPs, 100C>T (Arg34Trp: allele frequency 0.002) or lA-T (Met1Leu: allele frequency 0.002), showed low AUC ratios.66) Both haplo旬pesare important for hCE2 to

activate prodrugs, but hepatic hydrolase activity might be kept at a certain level due to the compensatory activity of hCE1.

Thus, prodrugs are hydrolyzed even in a subject with a variant

CES gene, and show a pharmacological effect. There is no re-

port that severe toxicity of a prodrug is caused by genetic

polymorphism of the CES gene; however, understanding ge-netic polymorphisms is important to confirm the safety and

effectiveness of noble prodrugs.

Conclusion

CESs are widely dis仕ibutedin all mammalian species, and play an important role in the bioconversion of prodrugs.

M勾orhuman CES isozymes, hCEl and hCE2, show different substrate specificity, resulting in tissue-specific hydrolysis. Therefore, successful prodrug design will be improved by fur-

ther detailed analysis of the substrate recognition and expres-

sion of human CES isozymes. Furthermore, detailed analysis of the species difference of tissue activity mediated by CES

might help in the prec1inical study of prodrug development.

Acknowledgements

The authors would like to thank all colleagues for their tremendous encouragement and helpful discussions.

1) B. Testa: Biochem. Pharmacol. 68,2097-2106 (2004). 2) T. Imai and M. Otagiri:“Chirality in Dmg Design and Develop-

ment," ed. by 1. K. Reddy and R. Mehvar: Marcel Dekker, Inc., NewYork,pp.l01-137,2004

3) B. M. Liederer, R. Borchardt: J. Pharm. Sci. 95, 1177-1195 (2006).

4) T. Satoh and M. Hosokawa: Annu. Rev. Pharmacol. Toxicol. 38,

CESl gene structure and haplotyp巴.

|1234cymk

Fig. 8.

References

morphism was significantly associated with the anti・hyperten-

sive efficacy of imidapril medication. Imidapril is a prodrug

ACE inhibitor, which requires hepatic activation by hCEl to form an active metabolite. It was shown that -816C allele

had higher transcriptional activity than the -816A allele.

Since no putative transcription factor recognition site was

found around the -816A/C region, it was speculated白atthis

polymorphism was a marker of other functional polymor-

phism(s). Recently, our investigation into the CESIA2 pro-moter region (ca. 1 kB) in 100 Japanese hypertensive patients

revealed ten SNPs at positions -816, -674, -427, -62, 47, -46, -41, -40, -37, and -32, and one IID at -34.63)

Pairwise D'姐 d~ showed that all of these polymorphisms were in high linkage disequilibrium (LD). From all eleven

polymorphisms spanning this region, fo町 haplotypeswere

obtained as infer haplotypes which had合'equenciesof more

than 1 %, and they accounted for 96% of the alleles. Three

consisted of the same SNPs between -62 to -32 and the

-34 IID as the most common haplotype (frequency of 54%), which accounted for 74% and residual 22% was the minor

haplotype. Interestingly, -816A/C was in high LD with the

m司jorand minor haplotypes (D'=0.92, ~=0.85).63) In con-仕astto the m司jorhaplotype, the minor haplotype had higher transcription and Sp 1 binding activity due to the presence of

two putative Sp 1 binding sites. The Sp 1 binding site variation

in the CESIA2 promoter affects the pharmacological effect

and the -816A/C might be a good candidate for pharmacoge-

netic study of CESl・activatedprodrugs.

More recently, Sai et al. reported a gene-dose effect of

functional CESIA genes on SN-38 formation in irinotecan-

treated Japanese cancer patients.64) lrinotecan is a prodrug of

SN-38 and is well hydrolyzed by hCE2 at high affinity in

comparison with hCEI23); however, hCEl also plays an im-portant role in the hydrolysis of irinotecan in the human

liver65) because of its major expression. In this study, CESIA diplotypes [combination ofhaplotypes A (lA3-1Al), B (lA2-

lA1), C (lA3-varlAl) and D (lA2-varlAl)] (Fig. 8) and the

m司jor SNPs (-75T>G and -30G>A in lAl, and 816A>C in lA2 and lA3) were determined in 177 Japanese

cancer patients. The associations of CESl geno匂rpes,the

number offunctional CESl genes (lAl, lA2 and varlAl) and

major SNPs, with an AUC ratio of (SN-38+SN・38G)/irinote-

can, a parameter of in vivo CES activity, we

Vol. 35, No. 3, 229-239 (2010)

Page 11: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

238 T. Imai and M. Hosokawa

257-288 (1998).

5) T. Satoh, P. Taylor, W. F. Bosron, P. Sanghani, M. Hosokawa and

B. N. Du: Drug Metab. Dispos. 30, 488-493 (2002).

6)工Satohand M. Hosokawa: Chem. Biol. Interact. 162, 195-211

(2006)

7) T. Imai: Drug Metab. Pharmacokinet. 21,173-185 (2006). 8) M. Hosokawa: Moleculω.13,412-431 (2008). 9) D. Shi, J. Yang, D. Yang, E. L. LeCluyse, C. Black, L. You, F.

Akhlaghi and B. Yan: J. Pharmacol. Exp. Ther. 319, 1477-1484 (2006).

10) T. Imai, M. Imoto, H. Sakamoto and M. Hashimoto: Drug Metab. Dispos. 33,1185-1190 (2005).

11) C. Jewell, P. Benett, E. Mutch, C. Ackermann and F.M.

Williams: Biochem. Pharmacol. 74, 932-939 (2007). 12) D. Yang, R. E. Pearce, X. Wang, R. Gaedigk, Y.-J. Y. Wan and

B. Yan: Biochem. Pharmacol. 77, 238-247 (2009). 13) R. Mentlein, S. Heiland and E. Heymann: Arch. Biochem. Bio-

phys. 200, 547-559 (1980). 14) R. Mentlein, H. Rix-Matz印 刷 E.Heymann,: Biochim. Bio-

phys. Acta 964, 319-328 (1988).

15) E. Heymann, R. Mentlein, R. Schmalz, C. Schwabe and F. Wa-

genmann: Eur. J. Biochem. 102,509-519 (1979). 16) T. Imai, M. Taketani, M. Shii, M. Hosokawa and K. Chiba:

DrugMetab. Dispos. 34,1734-1741 (2006). 17) M. Hosokawa, T. Endo, M. Fujisawa, S. Hara, N. Iwata, Y. Sato

and T. Satoh: Drug Metab. Dispos. 23,1022-1027 (1995). 18) G. Xu, W. Zhang, M. Ma, K. and H. L. McLeod: Clin. Cancer

Res. 8, 2605-2611 (2002). 19) L. Mizen and G. Burton: Pharm. Biotechnol. 11, 345-365

(1998).

20) K. Masaki, M. Taketani and T. Imai: Drug Metab. Dispos. 34,

398-404 (2006)

21) K. Masaki, M. Hashimoto and T. Imai: Drug Metab. Dispos. 35,

1089ー1095(2007).

22) N. Okudaira, T. Takebayashi, G. C. Speirs, 1. Komiya, Y.

Sugiyama: J. Pharmacol. Exp. Ther. 294, 580-587 (2000).

23) R. Humerickhouse, K. Lohrbach, L. Li, W. F. Bosron and M. E. Dolan: Cancer. Res. 60, 1189-1192 (2000).

24) M. Tang, M. Mukundan, J. Yang, N. Charpentier, E. L.

LeCluyse, C. Black, D. Yang, D. Shi and B. Yan: J. Pharmacol. Exp. Ther. 319, 1467-1476 (2006).

25) J. Zhang, J. C. Bumell, N. Dumaual and W. F. Bosron: J. Phar-

macol. Exp. Ther. 290, 314-ー318(1999).

26) S. K. Quinney, S. P. Sanghani, W. 1. Davis, T. D. Hurley, Z. Sun,

D. J. Murry加 dW. F. Bosron: J. Pharmacol. Exp. Ther. 313, 1011-1016 (2005).

27) S. Takahashi, M. Katoh, T. Saitoh, M. Nak司jimaand T. Yokoi: J.

Pharm. Sci. 97, 5434-5445 (2008). 28) S. Takai, A. Matsuda, Y. Usami, T. Adachi, T. Sugiyama, Y.

Katagiri, M. Tatematsu and K. Hirano: Biol. Pharm目 Bull.20, 869-873 (1997).

29) S. Bencharit, C. L. Morton, E. L. Howord-Williams, M. K.

Danks, P. M. Potter and M. R. Redin

Journal 01 Pesticide Science

Cashman and W. F. Bosron: Drug Metab. Dispos., 25,

1089-1096 (1997).

32) Z. Sun, D. J. Murry, S. P. Sanghani, W. 1. Davis, N. Y. Kedishvili,

Q. Zou, T. D. Hurley and W. F. Bosron: J. Pharmacol.

Exp. Ther. 310,469-476 (2004) 33) H. Huang, C. D. Fleming, K. Nishi, M. R. Redinbo and B. D.

Hammock: Chem. Rω. Toxicol. 18, 1371-1377 (2005). 34) S. Bencharit, C. L. Morton, Y. Xue, P. M. Potter and M. R. Red-

inbo・NatStruct Biol., 10: 349-56 (2003).

35) T. Furihata, M. Hosokawa, N. Koyano, T. Nakamura, T. Satoh

and K. Chiba: Drug. Metab. Dispos. 32, 1170-1177 (2004) 36) S. J. Godin, E. J. Scollon, M. F. Hughes, P. M. Potter, M. J. Dか

Vito and M. K. Ross: Drug. Metab. Dispos. 34, 1764ー1771(2006).

37) T. Furihata, M. Hosokawa, A. F可ii,M. Derbel,工 Satohand K.

Chiba: Biochem. Pharmacol. 69,1287-1297 (2005). 38) M. K. Ross, A. Bora宅jani,C. C. Edwards and P. M. PO杭er:

Biochem. Pharmacol. 71, 657-669 (2006) 39) M. K. Danks, C. L. Morton, E. J. Krull, P. J. Cheshire, L. B.

Richmond, C. W. Naeve, C. A. Pawlik, P. J. Houghton and P. M.

Potter: Clin Cancer Res. 5, 917-924 (1999). 40) M. Taketani, M. Shii, K. Ohura, S. Ninomiya and T. Imai: Life

Sci. 81, 924-932 (2007). 41) B. Lin, M. Sedlacek, 1. Manoharan, R. Boopathy, E. G. Duysen,

P. Masson and O. Lockridge: Biochem. Pharmacol. 70, 1673ー1684(2005).

42) J. J. Schentag, G. Hill, T. Chu and C. R. Rayner: J. Clin. Phar-macol. 47, 689-696 (2007)

43) W. Li, P. A. Escarpe, E. J. Eisenberg, K. C. Cundy, C. Sweet, K.

J. Jakeman, J. Merson, W. Lew, M. Williams, L. Zhang, C. U.

Kim, N. Bischofberger, M. S. Chen and D. B. Mendel: Antimi-

crob. Agents Chemother. 42,647-653 (1998).

44) D. J. Sweeny, G. Lynch, A. M. Bidgood, W. Lew, K. Y. Wang

and K. C. Cundy: Drug Metab. Dispos. 28, 737ー741(2000).

45) Y. Yoshigae, T. Imai,工 Asoand M. Otagiri: L扮 Sci.62,

1231-1241 (1998).

46)工Imai,Y. Yoshigae, M. Hosokawa, K. Chiba, M. Otagiri: J.

Pharmacol. Exp. Ther. 307, 1234-1242 (2003). 47) M. Hosokawa, T. Furihata, Y. Yaginuma, N. Yamamoto, N.

Watanabe, E. Tsukada, Y. Ohhata, K. Kobayashi, T. Satoh and

K. Chiba: Drug Metab. Pharmacokinet. 23, 73-84 (2008). 48) F. Shibata, Y. Takagi,M. Kitajima, T. Kuroda and T. Omura: Ge-

nomics. 17,76-82 (1993).

49) T. Langmann, A. Becker, C. Aslanidis, F. Notka, H. Ullrich, H.

Schwer and G. Schmitz: Biochim. Biophys. Acta 1350, 65-74

(1997).

50) H. Schewer, T. Langmann, R. Daig, A. Becker, C. Aslandis, G

Schmitz: Biochem. Biophys. Res. Commun. 233, 117-120

(1997).

51) S. Marsh, M. Xiao, J. Yu, R. Ahluwalia, M. Minton, R.R. Freimuth, P-Y. Kwok and H. L. McLeod: Genomics 8

Page 12: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

Vol. 35, No.3, 229-239 (2010)

55) M. Hosokawa, T. Furihata, Y. Yaginuma, N. Yamamoto, N. Ko-

yano, A. Fujii, Y. Nagahara, T. Satoh and K. Chiba: Drug

Metab. Rev. 39,1-15 (2007). 56) T. Furihata, M. Hosokawa, F. Nakata, T. Satoh and K. Chiba:

Arch. Biochem. Biophys. 416,101-109 (2003). 57)工 Furihata,M. Hosokawa, T. Satoh and K. Chiba: Biochem. J.

384,101-110(2004).

58)工Furihata,M. Hosokawa, M. Masuda, T. Satoh and K. Chiba:

Arch. Biochem. Biophys. 447, 107-117 (2006). 59) T. Fukami, M. Nak句ima,T. Maruichi, S. Takahashi, M.

Takamiya, Y. Aoki, H. L. McLeod and T. Yokoi: Pharmaco-

genet. Genomics 18, 911-920 (2008). 60) M. Hosokawa, Y. Endo, M. Fujisawa, S. Hara, N. Iwata, Y. Sato

and T. Satoh: Drug. Metab. Dispos. 23, 1022-1027 (1995) 61) T. Hori and M. Hosokawa: Xenobiotica. 40,119-128 (2010) 62) E. Geshi, T. Kimura, M. Yoshimura, H. Su四 ki,S. Koba, T.

Sakai,工 Saito,A. Koga, M. Muramatsu and T. Katagiri:時'Per-

Prodrug approach using carboxy1esterase activity 239

tens. Res. 28,719-725 (2005). 63) M 而 shimura,T. Kimura, M. Ishii, K. Ishii, T. Matsuura, E

Geshi, M. Hosokawa, M. Muramatsu: Biochem. Biophys. Res. Commun. 369, 939-942 (2008).

64) K. Sai, Y. Saito, N. Tatewaki, M. Hosokawa, N. Kaniwa, T.

Nishimaki-Mogami, M. Naito, J. Sawada, K. Shirao, T. Ham-

aguchi, N. Yamamoto, H. Kunitoh, T. Tamura, Y. Yamada, Y.

Ohe, T. Yoshida, H. Minami, A. Ohtsu, Y. Matsumura, N. Saijo,

H. Okuda: British J. Clin. Pharmacol. (2010). in press

65) J. G. Sla枕er,P. Su, J. P. Sams, L. J. Schaaf and L. C. Wienkers:

Drug Metab. Dispos. 25,1157-1164 (1997)

66) S.-R. Kim, K.Sai, T Kagawa, H. Jinno, S. Ozawa, N. Kaniwqa,

Y. Saito, A. Akasawa, K. Matsumoto, H. Saito, N. Kamatani, K

Shirao, N. Yamamoto, T. Yoshida, H. Minami, A. Ohtsu, N

Saiojo叩 dJ. Sawada: Drug Metab. Dispos. 35, 1865-1872 (2007)

Page 13: カルボキシルエステラーゼ活性を利用したプロドラッグの開 発hCEl dominantly contributes to hepatic hydrolysis rather than hCE2; thus, the first-pass hydrolysis

1111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111

英文編掲載報文・短報等の要旨1111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111

総説(スペシャルトピック:カルポキシルエステ

ラーゼ)

カルボキシルエステラーゼの分子構造と機能調節並びに新

分類法

佐藤哲男,細川正清

本総説では,カルボキシルエステラーゼの分子構造と機

能について概説した.カルボキシルエステラーゼには複数

の分子種があり,従来それらは基質特異性の違いに基づい

て分類されていたが,その方法では個々の分子種間で重複

が多いために明確に区別する事が困難であっfこ.それを解

消する目的で,著者らは各分子種の分子構造や機能の違い

に基づく新たな分類法を提唱した. 2010年に著者らが提唱

した分類法を基本とするカルボキシルエステラーゼ分子種

の新分類法が研究者間で国際的に承認された.また,動物

および人の各臓器に特有のカルボキシルエステラーゼ分子

種の遺伝子発現と,その調節機構についても明らかにした.

さらに,カルボキシルエステラーゼ分子種の一種であるエ

ガシンが,有機リン剤に極めて高い親和性を有する性質を

利用して,有機リン剤暴露時における新たなバイオマーカー

を開発した. この方法は,従来のコリンエステラーゼ阻害

法に比べてはるかに鋭敏であることが,動物および人にお

ける有機リン剤暴露時の結果から明らかになった.

カルボキシルエステラーゼ活性を利用したプロドラッグの

開発:哨乳動物組織におけるカルボキシルエステラーゼの

触媒特性と発現調節

今井輝子,細川正清

プロドラッグはそれ自体は薬理学的に不活性であり,生

体内で薬理効果の強い親薬物に変換される.プロドラッグ

化の長所は,標的臓器への薬物の到達,治療効果の最適化,

そして,副作用の回避を可能にすることである.プロドラッ

グ化のための化学修飾には,エステル結合やアミド結合が

利用されることが多く,そのため,プロドラッグの生体内

変換にはカルボキシルエステラーゼ (Carboxylesterase,CES)

が重要な役割を果たしている. CESには基質特異性の異な

る複数の IS0勾岨eが存在し,その発現組織は異なる.例え

ば, ヒト肝臓と小腸には,それぞれ hCElとhCE2が主に発

現し,臓器特異的な加水分解活性に寄与する. したがって,

これらの酵素の基質認識性の相違を利用することによって,

プロドラッグは理論的にデザインすることができるはずで

日本農薬学会誌 35(3),373-376 (2010)

ある. しかしながら, CESの発現レベルは個人差があり,

その結果,薬理効果の個人間差異を惹起することがある.

また,動物種によって CESの発現分子やその発現量は異な

るため,前臨床の動物実験で得られた結果からヒトにおけ

る効果を予測することは困難である. したがって,プロド

ラッグ創薬においては,代謝活性化の中心的役割を担って

いる CESの基質特異性,種差,臓器差,個人差を把握する

ことは非常に重要である.

カルボキシルエステラーゼ阻害剤のインシリコデザインと

評価

Shana V. Stoddard, Xiaozhen Yu, Philip M. Potter,

Randy M. Wadkins

カルボキシルエステラーゼ (CE)はさまざまな基質分解

や薬剤代謝にかかわる重要な酵素である. CE阻害剤の開発

は,薬剤の寿命延長や体内分布の調節,代謝された薬物の

毒性の軽減や除去,殺虫剤耐性害虫の低減などに役立つこ

とが期待される.ここでは,晴乳類 CEの既知の阻害剤の

主要なクラスについて概説するとともに,特異性のある新

規阻害剤を開発するための計算化学的なアプローチについ

ても述べる.インシリコにおける阻害剤開発において,構

造のドッキング,データベース検索,多次元 QSAR,そし

てdenovoドラッグデザインと結びついた QSARを使った

新規手法を含む一連の方案を議論する.本研究では, ヒト

小腸のカルボキシルエステラーゼ (hiCE)阻害剤をデザイ

ンしたが,その手法は他の組織や生体からの CEを含む多

くの酵素阻害剤の開発に広く応用できるであろう.

(文責:編集事務局)

ヒトカルボキシルエステラーゼ 1による農薬分解とその構

Andrew C. Hemmert, Ma抗hewR. Redinbo

ヒトカルボキシルエステラーーゼ 1(hCE1)は主な肝臓の

カルボキシルエステラーゼである.第 I相薬物代謝経路に

おいて,広範囲な生体内物質,生体外異物および農薬は解

毒される.これまで,有機リン剤を含むさまざまなリガン

ドと複合体を形成している hCElのX線結晶構造体が十数

種類あまり解析され, hCElによる農薬の代謝分解に有用情

報を与えてきた.例えば,触媒的トリアッドを構成してい

るさまざまな結合部位,およびこの部位を取り囲んでいる

長くてフレキシブルなループが基質特異性に関与している