novel mechanism-based inhibitors of serine proteases

68
NOVEL MECHANISM-BASED INHIBITIORS OF SERINE PROTEASES A Thesis by Xiangdong Gan M.S., Sichuan University, P. R. China, 1994 Submitted to the College of Liberal Arts and Sciences and the faculty of the Graduate School of Wichita State University in partial fulfillment of the requirements for the degree of Master of Science December 2005

Upload: others

Post on 03-Feb-2022

8 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Novel mechanism-based inhibitors of serine proteases

NOVEL MECHANISM-BASED INHIBITIORS OF SERINE PROTEASES

A Thesis by

Xiangdong Gan

M.S., Sichuan University, P. R. China, 1994

Submitted to the College of Liberal Arts and Sciencesand the faculty of the Graduate School of

Wichita State University in partial fulfillment ofthe requirements for the degree of

Master of Science

December 2005

Page 2: Novel mechanism-based inhibitors of serine proteases

ii

NOVEL MECHANISM-BASED INHIBITIORS OF SERINE PROTEASES

I have examined the copy of this thesis for form and content and recommend it be accepted inpartial fulfillment of the requirement for the degree of Master of Science, with a major inChemistry.

___________________________________William C. Groutas, Committee Chair

We have read this thesis

and recommend its acceptance

__________________________________Erach R. Talaty, committee Member

__________________________________Ram P. Singhal, Committee Member

__________________________________Lop-Hing Ho, Committee Member

Page 3: Novel mechanism-based inhibitors of serine proteases

iii

ACKNOWLEDGEMENTS

I would like to express my sincere heart felt gratitude to my advisor, Dr. William C. Groutas

for his excellent guidance, support and encouragement throughout my studies at Wichita State

University. It is my great pleasure to be one of his students. He is a great teacher, a great

researcher and a great advisor.

I also would like to extend my gratitude to Dr. Erach R. Talaty, Dr. Ram P. Singhal, and Dr.

Lop-Hing Ho for their guidance and for being a member of my thesis committee. Furthermore, I d

like thank Dr. Kevin Alliston for his assistance with the biochemical studies. I am also thankful

to the members of my group and friends for their help and kindness. I sincerely thank all the

faculty and staff of the department of chemistry at Wichita State University.

Finally, I would like to thank my family for their love and support.

Page 4: Novel mechanism-based inhibitors of serine proteases

iv

ABSTRACT

The design and in vitro biochemical evaluation of two novel classes of mechanism-based

inhibitors of human leukocyte elastase (HLE) that inactivate the enzyme via an unprecedented

enzyme-induced sulfonamide fragmentation cascade is described. The inhibitors incorporate in

their structure either an appropriately-functionalized saccharin scaffold, or a 1,2,

5-thiadiazolidin-3-one-1,1-dioxide scaffold. The inactivation of the enzyme by these inhibitors

was found to be efficient, time-dependent and to involve the active site. Biochemical, HPLC,

and mass spectrometric studies show that the interaction of these inhibitors with HLE results in

the initial formation of a Michaelis-Menten complex and subsequent formation of a tetrahedral

intermediate with the active site serine (Ser-195). Collapse of the tetrahedral intermediate with

tandem fragmentation results in the formation of a highly reactive conjugated sulfonyl imine

which can either react with water to form a relatively stable acyl enzyme and/or undergo a

Michael addition reaction with an active site nucleophilic residue (His-57). The results also

demonstrate convincingly the superiority of the 1, 2, 5-thiadiazolidin-3-one-1,1-dioxide scaffold

over the saccharin scaffold in the design of inhibitors of (chymo)trypsin-like serine proteases.

Page 5: Novel mechanism-based inhibitors of serine proteases

v

TABLE OF CONTENTS

CHAPTER PAGE

1 INTRODUCTION.................................................................................................................1

1.1 Proteases ............................................................................................................................1

1.2 Terminology and Classification ..........................................................................................2

1.3 Serine Proteases .................................................................................................................4

1.3.1 Catalytic Mechanism .................................................................................................4

1.3.2 Human Leukocyte Elastase ........................................................................................6

1.3.3 Substrate Specificity ..................................................................................................8

1.4 Enzyme Inhibition ..............................................................................................................9

1.4.1 Reversible Inhibition ............................................................................................... 10

1.4.2 Irreversible Inhibition .............................................................................................. 13

1.4.3 Inhibitors of Human Leukocyte Elastase..................................................................17

2 DESIGN RATIONALE AND RESEARCH GOALS .......................................................... 23

2.1 Design Rationale for Inhibitors Derived from Saccharin Scaffold ............................... 24

2.2 Design Rationale for Inhibitors Derived from 1, 2, 5-Thiadiazolidin-3-one 1, 1 Dioxide

Scaffold ...................................................................................................................... 25

2.3 Research Goals ........................................................................................................... 26

3 EXPIREMENTAL .............................................................................................................. 27

3.1 Enzyme Assays and Inhibition Studies: Incubation Method ........................................ 27

3.2 Enzyme Assays and Inhibition Studies: Progress Curve Method ................................. 28

Page 6: Novel mechanism-based inhibitors of serine proteases

vi

3.3 Hydroxylation Reactivation of Inactivated HLE ......................................................... 29

3.4 Substrate Protection.................................................................................................... 30

3.5 Efficiency of Inactivation (Determination of Partition Ratio) ...................................... 30

3.6 Reactivation of HLE-Inhibitor Complex ..................................................................... 31

3.7 HPLC Studies. Product Analysis and Identification of Inhibitor with HLE..................32

4 RESULTS AND DISCUSSION..........................................................................................33

4.1 Inactivation of HLE by Derivatives of (I) (Compound 8-13)....................................... 33

4.2 Inactivation of HLE by Derivatives of (II) (Compound 4-7)........................................45

5 CONCLUSIONS.................................................................................................................52

REFFERENCES .................................................................................................................... 53

Page 7: Novel mechanism-based inhibitors of serine proteases

vii

LIST OF TABLES

Table Page

1.1 NC-IUBMB Definition for Subclassifications of Peptidases ........................................2

1.2 Classes of Proteinases According to Catalytic Mechanism (Dunn, 1992) .....................4

1.3 Human Elastases..........................................................................................................7

1.4 Substrate Specificity of Neutropil-Derived Serine Proteases ........................................8

1.5 Templates Employed in Serine Protease Inhibitor Design (Mechanism-based Inhibitors)

................................................................................................................................. 18

4.1 Inhibitory Activity of Compounds 8-13 Towards Human Leukocyte Elastase and Bovine

Trypsin ..................................................................................................................... 33

4.2 Inhibitory Activity of Compounds 4-7 Towards Human Leukocyte Elastase and Bovine

Trypsin ..................................................................................................................... 38

Page 8: Novel mechanism-based inhibitors of serine proteases

viii

LIST OF FIGURES

Figure Page

1.1 Hydrolysis of Amide Bonds by Proteases. ...................................................................1

1.2 Terminology of Specific Subsites of Proteases and the Complementary Features of the

Substrate.....................................................................................................................3

1.3 Mechanism of Amide Hydrolysis Catalyzed by a Serine Protease ................................6

1.4 Mechanism-based Inactivation of a Serine Protease via an Enzyme-induced Lossen

Rearrangement.......................................................................................................... 19

1.5 Mechanism of the Gabriel-Colman Rearrangement ....................................................20

1.6 Mechanism-based Inactivation of a Serine Protease via an Enzyme-induced

Gabriel-Colman Rearrangement................................................................................ 21

1.7 Mechanism-based Inactivation of a Serine Protease via an Enzyme-induced Formation

of a Michael Acceptor............................................................................................. 17

2.1 General Structures of Inhibitors (I-II)......................................................................... 24

2.2 (a) Spontaneous Fragmentaion of -Amido Sulfonopeptides to Yield a Michael

Acceptor; (b) -Amido Sulfonamide Motif in Phthalimide and Saccharin Derivatives.

................................................................................................................................. 25

4.1 Structures of Compounds 8-13................................................................................... 33

4.2 Time-dependent Loss of Enzymatic Activity. ............................................................ 35

4.3 Kinetics of Inactivation of Human Leukocyte Elastase by Compound 9..................... 36

4.4 Kinetics of Inactivation of Bovine Trypsin by Compound 9....................................... 37

Page 9: Novel mechanism-based inhibitors of serine proteases

ix

4.5 Postulated Mechanism of Action of Inhibitor (I) ........................................................ 38

4.6 Subtrate Protection. ...................................................................................................39

4.7 Effect of Hydroxylamine on Enzyme Reactivation..................................................... 40

4.8 Inactivation of Human Leukocyte Elastase as a function of the Molar Ratio of Inhibitor

9 to Enzyme.............................................................................................................. 41

4.9 HPLC Analysis of Products Formed by Incubating Human Leukocyte Elastase with

Inhibitor 9................................................................................................................. 43

4.10 Structures of Compounds 4-7................................................................................... 45

4.11 Time-dependent Loss of Enzymatic Activity ........................................................... 46

4.12 Time-dependent Loss of Enzymatic Activity ........................................................... 48

4.13 Postulated Mechanism of Inhibition of Human Leukocyte Elastase by Inhibitor (II) 50

Page 10: Novel mechanism-based inhibitors of serine proteases

1

CHAPTER 1

INTRODUCTION

1.1 Proteases

Living systems are shaped by an enormous variety of biochemical reactions, nearly all of

which are mediated by a series of remarkable biological catalysts (enzymes). Biochemical

research since Pasteur s era has shown that although enzymes are subject to the same laws of

nature that govern the behavior of other substances, enzymes differ from ordinary chemical

catalysts in several important respects: a) higher reaction rates; b) milder reaction conditions;

c) greater reaction specificity; and d) capacity for regulation.1

Proteases (proteinases) are enzymes that can selectively catalyze the hydrolysis of peptide

bonds (Figure 1.1). Proteolytic enzymes are involved in a great variety of physiological

processes, including blood coagulation, digestion, complement activation, phagocytosis,

protein turnover (tissue remodeling), fibrinolysis, ovulation and fertilization, and hormone

generation and degradation. Poor regulation of the activity of proteases leads to a range of

diseases (cancer metastasis, inflammation, rheumatoid arthritis, muscle degradation, etc.)

HN

ONH

O

OP1

P1' HN

OOH

O

P1

+ H2NO

P1'Protease

Figure 1.1 Hydrolysis of Amide Bonds by Proteases

Page 11: Novel mechanism-based inhibitors of serine proteases

2

1.2 Terminology and Classification

In chemical terms, the enzymatic cleavage of peptide bonds is considered as hydrolysis,

usually called proteolysis. The enzymes responsible for the catalysis of proteolysis have been

named proteases , a term that originated in the nineteenth century German literature on

physiological chemistry. The International Union of Biochemistry and Molecular Biology

(IUBMB) (1984) has recommended to use the term peptidase for the subset of peptide bond

hydrolases (Subclass E.C.3.4). The widely used term protease is synonymous with peptidase.

Peptidases are further divided into exopeptidases , acting only near a terminus of a

polypeptide chain, and endopeptidases , acting internally in polypeptide chains. The term

proteinase used previously has been replaced by endopeptidase for consistency. In

addition, the EC list specifies different subtypes of exopeptidases and endopeptidases (Table

1.1).

Table 1.1 NC-IUBMB Definition for Subclassifications of Peptidases

Subclasses Activity

Exopeptidases Cleave near a terminus of peptides or proteins Aminopeptidases Remove a single amino acid from the free N-terminus Dipeptidyl peptidases Remove a dipeptide from the free N-terminus Tripeptidyl peptidases Remove a tripeptide from the free N-terminus Carboxypeptidases Remove a single amino acid from the free C-terminus Peptidyl dipeptidases Remove a dipeptide from the free C-terminus Dipeptidases Cleave dipeptides Omega peptidases Remove terminal residues that are substituted, cyclized or Linked by isopeptide bondsEndopeptidases Cleave internally in peptides or proteins Oligopeptidases Cleave preferentially on substrates smaller than proteins

Page 12: Novel mechanism-based inhibitors of serine proteases

3

To define a common nomenclature on the interaction of a substrate with a peptidase, the

system of Berger and Schechter (1976) has become generally accepted and used (Figure 1. 2).

This system is based on a schematic interaction of amino acid residues of the substrate with

specific binding subsites located on the enzyme. By convention, the subsites on the protease

are called S (for subsites e.g. S3, S2, S1, S1 , S2 , S3 ) and the substrate amino acid residues are

called P (for peptide e.g. P3, P2, P1, P1 , P2 , P3 ). The numbering of the residues is given from

the scissile bond2.

Figure 1.2 Terminology of Specific Subsites of Proteasesand the Complementary Features of the Substrate

Based on the nature of a key catalytic residue located at the active site of the proteases,

proteases are classified as serine, cysteine, aspartic and metallo proteases.

The catalytic nucleophile in serine and cysteine poteinases is the hydroxyl group of the

active site serine and the sulphydryl group of the active site cysteine, respectively. In aspartic

proteinases, two aspartic acid residues directly bind the nucleophilic water molecule. Metallo

proteinases contain a metal ion (typically zinc) that is usually bound by three amino acids.

The nucleophile is a water molecule, as in aspartic proteinases, positioned and possibly

activated by the active site metal ion (Table 1.2) 3.

HNNH

HN

NH

HN

NHP3 P1 P2'

P2O

O

O

O

OP1' P3'

S2 S1' S3'

S3 S1 S2'scissile bond

O

Page 13: Novel mechanism-based inhibitors of serine proteases

4

Table 1.2 Classes of Proteinases According to Catalytic Mechanism (Dunn, 1992)

Class Catalytic Residue Examples

Serine Ser195 (His57, Asp102)chymotrypsin, trypsin, elastase, thrombin, proteinase 3,

factor Xa, plasmin, cathepsin G

Cysteine Cys25 (His159) Cathepsins B, L, S

Aspartic Asp32, Asp215 pepsin, HIV protease, rennin, cathepsin D

Metallo- Zn2+ Angiotensin converting enzyme (ACE), MMP-2, MMP-9

1.3 Serine Proteases

Serine proteases are known as a diverse and widespread group of proteolytic enzymes.

They are so named because they have a common catalytic mechanism involving a reactive

Ser residue. The serine proteases include digestive enzymes from prokaryotes and eukaryotes,

as well as more specialized proteins that participate in development, blood coagulation

(clotting), inflammation, and numerous other processes. Elastase, trypsin and chymotrypsin

are some of the best studied serine proteases.

1.3.1 Catalytic Mechanism

The classical mechanism of serine peptidases is based on the catalytic triad of Ser195,

His57 and Asp102. After formation of a Michaelis complex, the carbonyl carbon atom of the

scissile bond is attacked by the active site Ser (Figure 1.3). The nucleophilicity of the serine

is enhanced by an adjacent histidine (His57) functioning as a general base catalyst. The

formation of an acyl enzyme complex is achieved through a tetrahedral intermediate which

collapses to yield the acyl enzyme and the cleavage of the peptide bond. Deacylation occurs

Page 14: Novel mechanism-based inhibitors of serine proteases

5

via the same mechanistic steps. In this case, the nucleophilic attack is performed by a bound

water molecule resulting in the release of the peptide and restoration of the Ser-hydroxyl of

the enzyme. The mechanism requires a binding site for the oxyanion of the tetrahedral

intermediate. This site is formed by the backbone amides of Ser195 and Gly193. The

catalytic contribution of the third member in the catalytic triad, the conserved Asp, has been

controversial. The early suggestion that the Asp accepts a proton to become uncharged in the

transition state has been opposed by newer experimental and theoretical data (Kossiakoff and

Spencer 1981; Warshel et al 1989). Further suggested roles of the Asp include stabilization of

the imidazole orientation (Rogers and Bruice 1974) as well as stabilization of the local

structure around the active site (Lau and Bruice 1999). In addition, it has been proposed that

the hydrogen bond between Asp and His is a low-barrier hydrogen bond which accounts for

the transition state stabilization (Frey et al 1994). 3

The nucleophilicity of the serine is enhanced by an adjacent histidine functioning as a

general base catalyst. The general base catalysis is involved in the formation of the

tetrahedral transition state intermediate, where the imidazole group of His-57 functions as a

general base to abstract the proton from the O of Ser-195 (during the acylation) or from a

water molecule (during the deacylation), facilitating the nucleophilic attack. This newly

protonated imidazole then provides general acid catalysis to assist the collapse of the

tetrahedral transition state intermediate by transferring its proton to the leaving group (the

scissile amide nitrogen during the acylation, and O of Ser-195 during the deacylation). As a

proton shuttle , the imidazole of His-57 is involved in a direct interaction with the substrate,

and a slight movement of the imidazole group may also be required for such function.

Page 15: Novel mechanism-based inhibitors of serine proteases

6

Therefore, the position and orientation of the imidazole ring is critical for catalysis.

R1 NH

HN

O

OR1'

OP1

P1'

N N

Ser195 Gly193

H H

NN

His57

H O Asp102

O

HO

Ser195

NN

His57

H O Asp102

O

HO

H

H2NR1

O

P1'

R1 NH

OHO

O

P1

NN

His57

H O Asp102

O

HO

HN

Ser195

N

Gly193

H

R1 NH

O

O

P1

HHN N

Ser195 Gly193

O

R1 NH

NHO

OR1'

OP1

P1'

HHN N

Ser195 Gly193

NN

His57

H O Asp102

O

OH

Tetrahedralintermediate

Primed aminoterminal fragment

Acyl-enzyme

Non-primed carboxyterminal fragment

Michaelis complex

E Ser195 E

Ser195 ESer195 E

Figure 1.3 Mechanism of Amide Hydrolysis Catalyzed by a Serine Protease.

1.3.2 Human Leukocyte Elastase

The fibrous protein elastin, which comprises an appreciable percentage of all protein

content in some tissues, such as arteries, some ligaments and the lungs, can be hydrolysed or

otherwise destroyed by a select group of enzymes classified as elastases. Elastases are

derived from many tissues in man, including the pancreas, neutrophils, macrophages,

monocytes, platelets, smooth muscle cells and fibroblasts. Elastases are defined by their

ability to generate soluble peptides from insoluble elastin fibres by a proteolytic process

Page 16: Novel mechanism-based inhibitors of serine proteases

7

called elastinolysis. Examples of human elastases include human leukocyte elastase (HLE)

(E.C. 3.421.37, Pancreatic elastase II (PE-II) (E.C. 3.4.21.71), and Macrophage

metalloelastase (MMP-12) (E.C. 3.4.24.65) (Table 1.3). HLE and PE are serine proteinases

whereas MMP-12 is a metallo-protease. The catalytic triads of HLE and PE-II are composed

of the residues of His57, Asp102 and Ser195. The catalytic site of active MMP-12 consists of a

catalytic zinc ion that is co-ordinated to three stabilizing histidine residues: His218, His222 and

His228 according to human fibroblast collagenase-1 (MMP-1) numbering system.

Table 1.3 Human Elastases

Enzyme Class Molecular weight(kDa) Catalytic residues

Human leukocyte elastase Serine protease 30 His57, Asp102, Ser195

Pancreatic elastase II Serine protease 26 His57, Asp102 , Ser195

Macrophage metalloelastase Metalloprotease 54 Zn, His218, His222 , His228

Human leukocyte elastase (HLE) is a glycosylated, strongly basic serine protease with a

molecular weight of approximately 30 kDa and is found in the azurophilic granules of human

polymorphonuclear leukocytes (PMN).4 This enzyme is released from PMN upon

inflammatory stimuli and has been implicated as a pathogenic agent in a number of disease

states such as pulmonary emphysema5, 6, rheumatoid arthritis7, adult respiratory distress

syndrome (ARDS)8, glomerulonephiritis9, cystic fibrosis10, 11 and cancer12. Increased

proteolysis, especially elastolysis, may occur in the lung parenchyma as a result of an

imbalance between HLE and its major endogenous inhibitor -PI, because of either an

Page 17: Novel mechanism-based inhibitors of serine proteases

8

acquired or an inherited deficiency of the protease inhibitor. As replacements to -PI,

synthetic, low molecular weight HLE inhibitors delivered to the site of unregulated PMN

elastase activity can be potentially useful in the treatment of pulmonary emphysema and

related diseases.

1.3.3 Substrate Specificity

The ability of proteases to selectively act upon a small number of targets in a myriad of

potential physiological substrates is necessary for maintaining the fidelity of most biological

functions. While substrate selection by proteases is governed by many factors, a principal

determinant is the substrate specificity of the enzyme s active site. Knowledge of a protease s

substrate specificity can greatly facilitate the elucidation of the physiological substrates of the

protease, which is essential for defining its role in complex biological pathways.

Determination of substrate specificity also can provide the basis for potent and selective

substrates and inhibitors. Table 1.4 summarizes the specificity of the three neutrophil-derived

proteases.

Table 1.4 Substrate Specificity of Neutrophil-Derived Serine Proteases

Enzyme S5 S4 S3 S2 S1 S1' S2' S3'

HLE Hydrophobic AlaAlaVal

ProValLeuNva

Hydrophobic

Cat G Polar AlaValThr

ProMet

Phe (not mapped)

PR 3 Pro Ala ProAlaAbu

Hydrophobic

Peptide P5 P4 P3 P2 P1 P1' P2' P3'

Page 18: Novel mechanism-based inhibitors of serine proteases

9

The substrate sequential specificity of serine proteases has often been mapped with

chromogenic substrates such as peptidyl p-nitroanilides, which identify favorable Pn-Sn

interactions. The most reliable information about the Pn -Sn interactions is from X-ray crystal

structures of protease-protein inhibitor complexes, which are not available for most serine

proteases.

Over the last few years a number of elegant combinatorial approaches have been

developed to determine the substrate specificity of proteases. All of these combinatorial

methods involve the generation of libraries of potential substrates, proteolysis of favorable

substrates, and identification of the cleaved substrate sequences. Combinatorial substrate

libraries can be divided into two categories depending on the method of generation:

biological and synthetic. Biological library methods include the use of substrate phage

display13, 14, the randomization of amino acids at physiological cleavage sites15, and the use of

in vitro expression cloning of complementary DNA (cDNA) libraries to determine

physiological substrates16. While biological combinatorial methods are very powerful, they

are complicated by the lack of homogeneity in their presentation of potential substrates and

the fact these methods are often time consuming and technically demanding17.

1.4 Enzyme Inhibition

Many substances alter the activity of an enzyme by reversibly combining with it in a way

that influences the binding of substrate and/or its turnover number. Substances that reduce an

enzyme s activity in this way are known as inhibitors. A large part of the modern

Page 19: Novel mechanism-based inhibitors of serine proteases

10

pharmaceutical arsenal consists of enzyme inhibitors. For example, AIDS is treated almost

exclusively with drugs that inhibit the activity of certain viral enzymes.

Inhibitors act through a variety of mechanisms.

1.4.1 Reversible Inhibition

A. Competitive Inhibition

A substance that competes directly with a normal substrate for an enzyme s

substrate-binding site is known as a competitive inhibitor. Such an inhibitor usually

resembles the substrate so that it specifically binds to the active site but differs from the

substrate in that it cannot react as the substrate does.

The general model for competitive inhibition is given by the following reaction scheme:

E + S ES P + E

+

I

EI + S NO REACTION

k1

k-1

k2

KI

Here it is assumed that I, the inhibitor, binds reversibly to the enzyme and is in rapid

equilibrium with it so that

KI =[ E ][ I ]

[ EI ]

and EI, the enzyme-inhibitor complex, is catalytically inactive. A competitive inhibitor

Page 20: Novel mechanism-based inhibitors of serine proteases

11

therefore reduces the concentration of free enzyme available for substrate binding.

Competitive inhibition studies are also used to determine the affinity of transition state

analogs for an enzyme s active site. If an enzyme preferentially binds its transition state, then

it can be expected that transition state analogs, stable molecules that geometrically and

electronically resemble the transition state, are potent inhibitors of the enzyme. Hundreds of

transition state analogs for various enzymes have been reported. Some are naturally occurring

antibiotics. Others were designed to investigate the mechanism of particular enzymes or to

act as specific enzyme inhibitors for therapeutic or agriculture use. Indeed, the theory that

enzymes bind transition states with higher affinity than substrates has led to a rational basis

for drug design based on the understanding of specific enzyme reaction mechanism.

B. Uncompetitive Inhibition

In uncompetitive inhibition, the inhibitor binds directly to the enzyme-substrate complex

but not to the free enzyme:

E + S ES P + E

+

I

ESI NO REACTION

k1

k-1

k2

K'I

In this case, the inhibitor binding step has the dissociation constant

Page 21: Novel mechanism-based inhibitors of serine proteases

12

K'I =[ ES ][ I ]

[ ESI ]

The uncompetitive inhibitor, which need not resemble the substrate, presumably distorts

the active site, thereby rendering the enzyme catalytically inactive. Uncompetitive inhibition

requires that the inhibitor affect the catalytic function of the enzyme but not its substrate

binding. This is difficult to envision for single-substrate enzymes. In actuality, uncompetitive

inhibition is significant only in multisubstrate enzymes.

C. Noncompetitive Inhibition

If both the enzyme and the enzyme-substrate complex bind inhibitor, the following

mode results:

E + S ES P + E

+

I

ESI NO REACTION

k1

k-1

k2

K'I

+

I

EI

KI

This phenomenon is known as noncompetitive inhibition. Presumably, a noncompetitive

inhibitor binds to enzyme sites that participate in both substrate binding and catalysis. The

two dissociation constants for inhibitor binding are not necessarily equivalent.

KI =[ E ][ I ]

[ EI ]and K'I =

[ ES ][ I ]

[ ESI ]

Page 22: Novel mechanism-based inhibitors of serine proteases

13

1.4.2 Irreversible Inhibition

If an inhibitor binds irreversibly to an enzyme, the inhibitor is classified as an

irreversible inhibitor or an inactivator. Reagents that chemically modify specific amino acid

residues can act as irreversible inhibitors. For example, the compounds used to identify the

catalytic Ser and His residues of serine proteases are irreversible inhibitors of these enzymes.

Irreversible in this context, however, does not necessarily mean that the enzyme activity

never returns, only that the enzyme becomes dysfunctional for an extended (but unspecified)

period of time. A compound that forms a covalent bond with an enzyme may lead to the

formation of an E-I complex that regains activity slowly. There are other cases of irreversible

inhibition in which no covalent bond forms at all, but the compound binds so tightly to the

active site of the enzyme that the rate constant for release of the compound from the enzyme

is exceedingly small. This, in effect, produces irreversible inhibition. Irreversible inhibitors

include affinity labeling agents and mechanism-based inhibitors.

A. Affinity Labeling Agents

Affinity labeling agents are generally reactive (electrophilic) compounds that alkylate or

acylate enzyme nucleophiles. Often more than one enzyme nucleophile reacts with these

compounds, and if a crude system containing more than one enzyme is being used, reactions

with multiple enzymes can occur. The closer that the structure of the affinity labeling agent

resembles that of the substrate for the target enzyme, the greater the specificity that will be

Page 23: Novel mechanism-based inhibitors of serine proteases

14

attained. A less reactive variation of affinity labeling agents, termed quiescent affinity

labeling agents, has been described. These inactivators contain an electrophilic carbon that is

stable in solution in the presence of external nucleophiles but which can react with an enzyme

active site nucleophile involved in covalent catalysis.

B. Mechanism-based Inhibitor

The name mechanism-based enzyme inactivator conjures up the idea of an enzyme

inactivator that depends on the mechanism of the targeted enzyme. In the broadest sense of

the term any of the inactivators that utilize the enzyme mechanism could be classified as

mechanism-based enzyme inactivators. In fact, Krantz and Pratt have suggested this general

classification for mechanism-based enzyme inhibitors. However, there is a more narrow

definition of mechanism-based enzyme inhibitor that Silverman has invoked. A

mechanism-based enzyme inactivator is an unreactive compound whose structure resembles

that of either the substrate or product of the target enzyme, and which undergoes a catalytic

transformation by the enzyme to a species that, prior to release from the active site,

inactivates the enzyme. A mechanism-based inhibitor requires a step to convert the compound

to the inactivating species by the catalytic machinery of the enzyme. Most often these

inhibitors result in covalent bond formation with the enzyme (Scheme 1.1), and, therefore,

dialysis or gel filtration does not restore enzyme activity. In other words, the inhibition is

irreversible.

Page 24: Novel mechanism-based inhibitors of serine proteases

15

E + I EI EI* E - I*kon

koff

k2 k3

k4

E + I*

Scheme 1.1

The ratio of product release to inactivation is termed the partition ratio and represents

the efficiency of the mechanism-based enzyme inactivator. When inactivation is the result of

the formation of an E I* adduct, the partition ratio is described by k4/k3. The partition ratio

depends on the rate of diffusion of I* from the active site, its reactivity, and the proximity of

an appropriate nucleophile, radical, or electrophile on the enzyme for covalent bond

formation. It does not depend on the initial inactivator concentration. There are some cases

where the partition ratio has been shown to be 0, that is, every turnover of inactivator

produces inactivated enzyme.

The two principal areas where mechanism-based enzyme inactivators have been most

useful are in the study of enzyme mechanisms and in the design of new potential drugs.

Mechanism-based inhibition is time dependent. Following a rapid equilibrium between

the enzyme and the mechanism-based inhibitor to give the EI complex, there is a slower

reaction that converts the inhibitor to the form that actually inactivates the enzyme. This

produces a time-dependent loss of enzyme activity. Formation of the EI complex occurs

rapidly, and the rate of inactivation is proportional to added inhibitor until sufficient inhibitor

is added to saturate all of the enzyme molecules. Then there is no further increase in rate with

additional inhibitor, that is, saturation kinetics is observed. Mechanism-based enzyme

Page 25: Novel mechanism-based inhibitors of serine proteases

16

inhibitors act as modified substrates for the target enzymes and bind to the active site.

Therefore, addition of a substrate or competitive reversible inhibitor slows down the rate of

enzyme inactivation. This is referred to as substrate protection of the enzyme.

Most enzyme inhibitor drugs (EID) are noncovalent, reversible inhibitors. The basis for

their effectiveness is the tightness of their binding to the enzyme, so that they compete with

the binding of substrates (S) for the enzyme. The efficacy of the drug continues as long as the

enzyme is complexed with the drug (E EID). Because the enzyme concentration is low and

fixed, the equilibrium between E + EID and E EID will depend on the concentrations of

EID and S. When the concentration of EID diminishes because of metabolism, the

concentration of E EID diminishes and the concentration of E S increases. To maintain the

pharmacological effect of the drug, administration of the drug several times a day, then,

becomes necessary. An effective mechanism-based enzyme inhibitor, however, could form a

covalent bond to the enzyme. This would mean that frequent administration of the drug

would not be necessary. Inactivation of an enzyme, however, induces gene-encoded synthesis

of that enzyme, but this could take hours to days before sufficient newly synthesized enzyme

is present. Although affinity labeling agents also could form a covalent bond to the enzyme,

the reactivity of these compounds renders them generally unappealing for drug use because

of the possibility that they could react with multiple enzymes or other biomolecules, thereby

leading to toxicity and side effects. Mechanism-based enzyme inhibitors, however, are

unreactive compounds, so nonspecific reactions with other biomolecules would not be a

problem. Only enzymes that are capable of catalyzing the conversion of these compounds to

the form that inactivates the enzyme, and also have an appropriately positioned active site

Page 26: Novel mechanism-based inhibitors of serine proteases

17

group to form a covalent bond, would be susceptible to inactivation. With a sufficiently

clever design it should be possible to minimize the number of enzymes that would be affected.

Provided the partition ratio is zero or a small number, in which case potentially toxic product

release would not be important, then a mechanism-based enzyme inhibitor could have the

desirable drug properties of specificity and low toxicity18.

1.4.3 Inhibitors of Human Leukocyte Elastase

The design of potent, synthetic inhibitors of HLE requires a thorough understanding of

the catalytic mechanism by which serine proteinases degrade proteins. This general

mechanism is illustrated in Figure 1.3. The catalytic triad of serine proteinases must generate

the tetrahedral intermediate in order for the enzyme to complete its task of cleaving

proteins. Synthetic, small molecule inhibitors of HLE may be realized if a proper design

element is built into the inhibitor which may cause an interference with the generation of the

tetrahedral intermediate. Numerous reports have been generated which describe in detail the

chemical strategy in designing potent and selective HLE inhibitors. All of these compounds,

however, can be divided into three subclasses: electrophilic ketones, acylating agents, and

miscellaneous agents.

Mechanism-based inhibitors are included in the miscellaneous agent subclass. They are

very important because mechanism-based inhibitors have some advantages against others. In

general, the design of a mechanism-based inhibitor involves the use of a suitable template to

which appropriate recognition and reactivity elements are appended. A wide range of

templates have been used in the design of mechanism-based inhibitors of serine proteases

Page 27: Novel mechanism-based inhibitors of serine proteases

18

such as HLE (Table 1.5).

NR1

O

O

OSO2R2 NN

R1

O

O

OSO2R3

R2N

NR1

R2

O

OOSO2R3

N

O

O

R1OSO2R2 X

N

O

Z

L

SN

OR1

LO O

N SN

R1O

R2L

O OS

NR

O

LO O

A B C

D E F

G H

Table 1.5 Templates Employed in Serine Protease Inhibitor Design (Mechanism-based Inhibitors)

The succinimide A19-22, hydantoin B23, dihydrouracil C24, and phthalimide D25, 26

templates, and variants thereof27, were first used in comjunction with the design of

mechanism-based inhibitors of (chymo)trypsin-like proteases that inactive the target enzyme

via an enzyme-induced Lossen rearrangement [Fig. 1.4].

Page 28: Novel mechanism-based inhibitors of serine proteases

19

N

O

O

OSO2R2

OH N

NH

Ser195

His57

E

N

O

OSO2R2

OHN

NH

Ser195

His57

EO

HN

NH

Ser195

His57

E

N

O

O

O

OSO2R2

N

NH

Ser195

His57

E

N

OO

CO

N

NH

Ser195

His57

EHN

OO

O

Figure 1.4 Mechanism-based Inactivation of a Serine Proteasevia an Enzyme-induced Lossen Rearrangement

As shown in Fig.1.4, these compounds react with the catalytic serine residue to give a

ring-opened species which undergoes a Lossen rearrangement to generate a latent

enzyme-bounded isocyanate. This isocyanate is subsequently attacked by the imidazole ring

of the catalytic histidine residue (His57) to give an enzyme-bound imidazole-N-carboxamide.

The double hit process leading to the formation of the enzyme inhibitor complex is

supported by 13C NMR studies28. The aforementioned approach has been extended to the

phthalimide template (Table 1.5, D). Kerrigan and coworkers29, 30 have demonstrated that the

introduction of hydrophobic and/or chiral substituents through an amide linkage at the

6-position of the phthalimide template enhances both the potency and selectivity of

(sulfonyloxy) phthalimide inhibitors. The mechanism of action of inhibitor D is similar to

that of A [Fig. 1.4]. The succinimide and phthalimide templates, as well as the saccharin

template, have also been used in the synthesis of mechanism-based inhibitors designed to

inactivate a serine protease via an enzyme-induced Gabriel-Colman rearrangement31. This

Page 29: Novel mechanism-based inhibitors of serine proteases

20

rearrangement involves the reaction of a phthalimido- or saccharino- acetic ester or ketone

with an alkoxide to yield a ring expansion product. The reaction is believed to involve

alkoxide-induced ring opening, followed by a prototropic shift of the resulting imide anion to

the carbanion and subsequent ring closure [Fig. 1.5].

XN

O

Z

RO

XN

Z

O OR

X

OR

O

N

Z

X

OR

O

NH

Z

XNH

ZO OR

XNH

ZO

XNH

ZOH

( X=CO, SO2; Z=COOR )

Figure 1.5 Mechanism of the Gabriel-Colman Rearrangement

It was reasoned that an appropriate saccharin or phthalimide derivative E might undergoes

enzyme-induced ring opening followed by a prototropic shift and loss of leaving group L to

yield a reactive electrophilic species (a Michael acceptor) which, upon further reaction with a

nearby nucleophilic residue, would lead to inactivation of the enzyme, as illustrated in Fig.

1.6.

Page 30: Novel mechanism-based inhibitors of serine proteases

21

XN

O

ZL

OH N

NH

Ser195

His57

E

XN

Z

O O

HN

NH

Ser195

His57

E

L

X

O

O

N

LZ

N

NH

Ser195

His57

E

X

O

O

HN

N

NH

Ser195

His57

E

X

O

O

HN

Ser195

His57

E

Z

L

ZN NH

X

O

O

HN

Ser195

His57

E

Z

NNH

Figure 1.6 Mechanism-based Inactivation of a Serine Proteasevia an Enzyme-induced Gabriel-Colman Rearrangement

The enzyme induced generation of a reactive Michael acceptor has also been

accomplished by employing various heterocyclic templates, including phthalimide D,

saccharin F32-36, 1,2,5-thiadiazolidin-3-one 1,1 dioxide H (Table 1.5)37, 38. In each instance,

enzyme-induced ring opening is followed by elimination to yield a reactive N-sulfonyl imine

(a Michael acceptor) which, upon further reaction with an active site neucleophilic residue

(His57) leads to irreversible inactivation of the enzyme [Fig. 1.7]39.

Page 31: Novel mechanism-based inhibitors of serine proteases

22

SN

O OH N

NH

Ser195

His57

E

SN

OHN

NH

Ser195

His57

EO

N

NH

Ser195

His57

E

S

O

O

N

NH

Ser195

His57

E

S

OO

O OL

O OL

NO O

HN

O O

Figure 1.7 Mechanism-based Inactivation of a Serine Proteasevia Enzyme-induced Formation of a Michael Acceptor

Page 32: Novel mechanism-based inhibitors of serine proteases

23

CHAPTER 2

DESIGN RATIONALE AND RESEARCH GOALS

Chronic obstructive pulmonary disease (COPD) (pulmonary emphysema and chronic

bronchitis) affects more than 16 million Americans and is the fourth most common cause of

death40. The pathogenesis of COPD is currently poorly understood41, 42. COPD is a

multifactorial disorder that is characterized by airway inflammation and the influx of

neutrophils, macrophages and natural killer lymphocytes to the lungs. This is accompanied by

the extracellular release of an array of proteases (serine, cysteine and metallo- proteases),

including the serine endopeptidases elastase, proteinase 3 and cathepsin G43, leading to a

protease/antiprotease imbalance44, 45. The presence of elevated levels of proteases in the lungs

leads to the degradation of lung elastin and other components of the extracellular matrix46, 47;

however, the identity, origin and precise role of the proteases involved in the pathogenesis of

COPD have not been rigorously defined as yet41, 42. The design and utility of novel

mechanism-based (suicide) inhibitors in mechanistic enzymology and drug discovery are

well-documented18. A mechanism-based inhibitor is an inherently unreactive compound that

acts as a substrate and is processed by the catalytic machinery of an enzyme, generating a

highly reactive electrophilic species which, upon further reaction with an active site

nucleophilic residue, leads to irreversible inactivation of the enzyme48. Inhibitors of this type

offer many potential advantages, including high enzyme specificity, since the latent reactivity

in the inhibitor is unmasked following catalytic processing of the inhibitor by the target

enzyme only.

Page 33: Novel mechanism-based inhibitors of serine proteases

24

SN

O

O OSO2NHR

N SN

O

O OSO2NHR

P1

R2

S1

S2 Sn'

(I) (II)

Figure 2.1 General Structures of Inhibitors (I-II)

2.1 Design Rationale for Inhibitors Derived from Saccharin Scaffold (Fig. 2.1 (I))

The biochemical rationale underlying the design of inhibitor (I) was based on the

following observations: (a) replacement of the carbonyl group in peptides with SO2 yields

-amido sulfonamides which are known to undergo a spontaneous fragmentation reaction, as

illustrated in Figure 2.2 (a)49, 50; (b) in contrast to acyclic -amido sulfonamides,

N-(phthalimidosulfonyl)-L-phenylalanine methyl ester 13 (Figure 2.2 (b), X = CO) and

related compounds have been shown to be stable (8); (c) inhibitors based on the saccharin

scaffold are known to dock to the active site of (chymo)trypsin-like proteases, an event that is

followed by acylation of the active site serine (Ser195)51-55; and, (d) the imidazole ring of

histidine residues located at the active site of enzymes is known to undergo facile Michael

addition reactions with conjugated systems56, 57. Based on these considerations, we

reasoned that an entity such as (I) might inactivate a target serine protease via a sequence of

steps involving the initial formation of a Michaelis-Menten complex, followed by

enzyme-induced ring opening and tandem fragmentation, leading to the release of an amine

or aminoacid ester, sulfur dioxide, and the formation of a Michael acceptor (in this instance,

an N-sulfonyl imine)58 capable of reacting with an active site nearby nucleophilic residue

(His57), ultimately leading to inactivation of the enzyme.

Page 34: Novel mechanism-based inhibitors of serine proteases

25

NH

NN

O

O

O

H

H

R1 R2

R

NH

N SN

O O

H

H

R1 R2

R

O ONH

N

O

R1

R

+ SO2 + H2NO

R2

a)

b)

XN

O

SO

NH

RO

X=CO,SO2

Figure 2.2 (a) Spontaneous Fragmentation of -Amido Sulfonopeptides to Yield a MichaelAcceptor; (b) -Amido Sulfonamide Motif in Phthalimide and Saccharin Derivatives.

2.2 Design Rationale for Inhibitors Derived from 1, 2, 5-Thiadiazolidin-3-one 1, 1

Dioxide Scaffold (Fig. 2.1 (II))

While the saccharin scaffold has been used in the design of protease inhibitors59, 60, it has

some serious limitations. For instance, structural constraints, such as the lack of a tetrahedral

carbon, preclude its binding to the active site in a substrate-like fashion. Secondly, synthetic

constraints severely limit the ready availability of ring-substituted derivatives. These

constraints make the systematic optimization of potency and enzyme selectivity of potential

inhibitors problematic. In contrast, a scaffold that binds to the active site of a target protease

like a substrate and is capable of orienting recognition elements toward the S and S´ subsites

Page 35: Novel mechanism-based inhibitors of serine proteases

26

would be expected to exhibit superior characteristics, since such a template would make

possible the exploitation of (a) binding interactions with multiple S and S´ subsites and, (b)

the subtle differences that exist in the various subsites of closely-related proteases. Based on

the above considerations, a versatile heterocyclic template, 1, 2, 5-thiadiazolidin-3-one-1, 1-

dioxide scaffold, (Figure 2.1, structure (II)) has been used in the design of a general class of

mechanism-based inhibitors of HLE61-69.

2.3 Research Goals

With respect to the above two types of compounds (I II), the following objectives were

explored:

1) Do inhibitors (I II) inactivate serine proteases, in particular HLE?

2) Do derivatives of I and II function as mechanism-based inhibitors?

3) What is the mechanism of inactivation of inhibitors (I II)?

4) Are inhibitors based on scaffold (II) more effective than those based on scaffold (I)?

Page 36: Novel mechanism-based inhibitors of serine proteases

27

CHAPTER 3

EXPERIMENTAL

3.1 Enzyme Assays and Inhibition Studies: Incubation Method

(A) Human Leukocyte Elastase (HLE)

Human leukocyte elastase ([E]f = 0.7 M) was assayed by mixing 10 L of a 70 M

enzyme solution in 0.05 M sodium acetate buffer, pH 5.5, 100 L dimethyl sulfoxide, and

890 L of 0.1 M HEPES buffer, pH 7.25, in a thermostatted test tube. A 100 L aliquot was

transferred to a thermostatted cuvette containing 880 L of 0.1 M HEPES, pH 7.25, and 20

L of a 7.0 mM solution of MeOSuc-Ala-Ala-Pro-Val-p-NA ([S]f = 0.14 M), and the change

in absorbance was monitored at 410 nm for 1 minute. In a typical inhibition run, 10 L of a

21 mM solution of inhibitor 9 ([I]f = 0.21 mM) in dimethyl sulfoxide and 90 L dimethyl

sulfoxide were mixed with 10 L of a 70 M enzyme solution ([E]f = 0.7 M) and 890 L of

M HEPES buffer, pH 7.25, and placed in a constant temperature bath, Aliquots (100 L)

were withdrawn at different time intervals and transferred to a cuvette containing 20 L of a

7.0 mM substrate solution ([S]f = 0.14 mM) and 880 L of 0.1 M HEPES buffer, pH 7.25.

The absorbance was monitored at 410 nm for 1 minute. The kinetics data obtained by using

the incubation method was analyzed by determining the slopes of the semilogarithmic plots

of enzymatic activity remaining vs time using eq 3.1 below, where [E]t/[E]o is the amount of

active enzyme remaining at time t.70

ln([E]t/[E]o) = kobs t (3.1)

Page 37: Novel mechanism-based inhibitors of serine proteases

28

(B) Trypsin

Bovine trypsin was assayed spectrophotometrically by mixing 10 L of a 0.50 mM

enzyme solution (0.1 M Tris-HCl buffer containing 0.01 M CaCl2, pH 7.2), 20 L dimethyl

sulfoxide, and 970 L 0.025 M sodium phosphate buffer containing 0.1 M NaCl, pH 7.51, in

a thermostated test tube. A 100 L aliquot was transferred to a thermostated cuvette

containing 10 L of a 60 mM solution of N -benzoyl-L-Arg p-nitroanilide and 890 L 0.025

M sodium phosphate buffer, and the change in absorbance was monitored at 410 nm for one

minute. In a typical inhibition run, 20 L of a 5.00 mM inhibitor solution in dimethyl

sulfoxide was mixed with 10 L of a 0.50 mM enzyme solution and 970 L 0.025 M sodium

phosphate buffer containing 0.1 M NaCl, pH 7.51, and placed in a constant temperature bath.

Aliquots (100 L) were withdrawn at different time intervals and transferred to a cuvette

containing 10 L of a 60 mM solution of N -benzoyl-L-Arg p-nitroanilide and 890 L 0.025

M sodium phosphate buffer, and the change in absorbance was monitored at 410 nm for one

minute. The pseudo first-order rate constants (kobs) were obtained by determining the slopes

of the semilogarithmic plots of enzymatic activity remaining vs time using eq 3.1 , where

[Et]/[Eo] is the amount of active enzyme remaining at time t. These are the average of two or

three determinations. The potency of the inhibitors was expressed in terms of the bimolecular

rate constant kobs/[I] M-1 s-1.

3.2 Enzyme Assays and Inhibition Studies: Progress Curve Method

The apparent second-order rate constants kinact/KI of compounds were determined by the

Page 38: Novel mechanism-based inhibitors of serine proteases

29

progress curve method (23). Thus, in a typical run 5 L of a 2.0 M HLE solution was added

to a solution containing 10 L of inhibitor (0.5 mM solution in dimethyl sulfoxide), 15 L

substrate (7.0 mM MeOSuc-Ala-Ala-Pro-Val p-NA), and 970 L 0.1 M HEPES buffer, pH

7.25, and the absorbance was continuously monitored at 410 nm for 600 s. Control curves in

the absence of inhibitor were linear. The pseudo-first order rate constants, kobs, for the

inhibition of HLE as a function of time were determined according to eq 2 below, where A is

the absorbance at 410 nm, vo is the reaction velocity at t = 0, vs is the final steady-state

velocity, kobs is the observed first-order rate constant, and Ao is the absorbance at t = 0. This

involved fitting by non-linear regression analysis the A ~ t data into eq 3.2 (SigmaPlot, Jander

Scientific) to determine kobs. The second order rate constants (kinact/KI) were determined in

duplicate or triplicate by calculating kobs/[I] and then correcting for the substrate

concentration and Michaelis constant using eq 3.3.

A = vst + {(vo vs) (1 e-kobs t)/kobs} + Ao (3.2)

kobs/[I] = kinact/KI {1 + [S]/Km} (3.3)

3.3 Hydroxylamine Reactivation of Inactivated HLE.

A solution containing 980 L 0.1 M HEPES buffer, pH 7.25, 10 L 21.9 mM inhibitor

([I]f = 219 M) in DMSO, and 10 L 21.9 M HLE ([E]f = 219 nM) was incubated for 30

minutes. The enzyme was totally inactivated, as shown by withdrawing an aliquot and

assaying for remaining enzyme activity. Excess hydroxylamine (100 L of a 0.50 M solution

in water) was then added to the fully-inactivated enzyme. Aliquots (100 L) were removed at

various time intervals (from one minute to 24 h) and assayed for remaining enzyme activity

Page 39: Novel mechanism-based inhibitors of serine proteases

30

by mixing with 20 L of a 3.86 mM solution of MeOSuc-Ala-Ala-Pro-Val p-nitroanilide ([S]f

= 77.2 M), and 880 L 0.1 M HEPES buffer, pH 7.25), and monitoring the absorbance at

410 nm. Enzyme activity was determined by comparing the activity of an enzyme solution

containing no inhibitor (control) with the activity of an enzyme solution containing inhibitor

at the same time point.

3.4 Substrate Protection

In separate experiments, the kobs/[I] values were determined by incubating HLE with

inhibitor 9 in the absence and presence of substrate. In the former case, 10 L of HLE ([E]f

= 219 nM) was incubated with 10 L of inhibitor ([I]f = 21.9 M) dissolved in DMSO and

0.1 M HEPES buffer (980 L), pH 7.25, in a thermostatted cuvette. Aliquots (100 L) were

withdrawn at different time intervals and added to a thermostatted cuvette containing 0.M

HEPES buffer (880 L), pH 7.25, and MeOSuc-Ala-Ala-Pro-Val pNA ([S]f = 77.2 M). The

absorbance was monitored at 410 nm and the kobs/[I] M-1s-1 value was then determined. The

kobs/[I] value was also determined by repeating the experiment in the presence of substrate:

HLE ([E]f = 219 nM) was incubated with inhibitor ([I]f = 21.9 M), and

MeOSuc-Ala-Ala-Pro-Val pNA ([S]f =231.6 M) in 0.1 M HEPES buffer (974 L, pH 7.25)

in a thermostatted cuvette. Aliquots (100 L) were withdrawn at different time intervals and

added to a thermostatted cuvette containing MeOSuc-Ala-Ala-Pro-Val pNA ([S]f = 77.2 M)

and 0.1 M HEPES buffer (880 L), pH 7.25.

3.5 Efficiency of Inactivation (Determination of Partition Ratio)

Page 40: Novel mechanism-based inhibitors of serine proteases

31

Ten microliters of inhibitor ([I]f = 3.5-42 M) in DMSO was incubated with 10 L HLE

([E]f = 0.70 M) and 980 L of 0.1 M HEPES buffer, pH 7.25, for 15 minutes. At the end of

the 15-minute incubation period enzyme activity was assayed by transferring an aliquot (100

L) to a cuvette containing 20 L MeOSuc-Ala-Ala-Pro-Val p-NA ([S]f = 0.14 M) and 880

L of 0.1 M HEPES buffer, pH 7.25, and monitoring the absorbance at 405 nm. The partition

ratio was calculated as described by Knight and Waley by plotting the fraction of remaining

enzyme activity ([E]t/[E]o) versus the initial ratio of inhibitor to enzyme ([I]/[E]o).

3.6 Reactivation of the HLE-Inhibitor Complex

Forty L of a 70.0 M solution of human leukocyte elastase was incubated with excess

inhibitor (10 L of a 25.2 mM solution in dimethyl sulfoxide), 40 L DMSO and 410 L 0.1

M HEPES buffer, pH 7.25 at 25.0 oC. After the solution was incubated for 30 minutes, a 25

L was removed and assayed for enzymatic activity (the enzyme was found to be completely

inhibited). Excess inhibitor was removed via Centricon-10 filtration by centrifuging at

14,000g for 45 minutes at 25.0 oC. Buffer (500 L 0.1 M HEPES buffer, pH 7.25) was added

to the HLE-inhibitor complex and the centrifugation was repeated at 14,000g for 1 h at 25.0

oC. The HLE-inhibitor complex was dissolved in 2.0 mL buffer and aliquots (100 L) were

withdrawn at different time intervals and added to a cuvette containing 20 L of 7.0 mM

MeOSuc-Ala-Ala-Pro-Val p-nitroanilide, 880 L of 0.1 M HEPES buffer, pH 7.25, and

monitoring the absorbance at 410 nm. The amount of active enzyme was determined by

comparing the activity of an enzyme solution containing no inhibitor (control) with the

activity of an enzyme solution containing inhibitor at the same time point.

Page 41: Novel mechanism-based inhibitors of serine proteases

32

3.7 HPLC Studies. Product Analysis and Identification from the Incubation of Inhibitor

with HLE.

A solution of HLE (50 L) containing 1 mg/mL of enzyme was added to a test tube

containing 300 L 0.1 M HEPES buffer, pH 7.25, containing 0.5 M NaCl and 0.2 mM

inhibitor. A second sample containing 0.1 N HEPES buffer, pH 7.25 with 0.5 M NaCl, and

0.2 mM inhibitor but no enzyme was used as a control. Both samples were incubated in a

water bath at 23 oC for 24 h. Fifty microliters of a saturated dansyl chloride solution (18

mg/mL) in methanol was added to each test tube and after 1 h the samples were analyzed by

HPLC. Both samples were stored at 4 oC for 72 h after which the clear solutions were

re-injected sequentially along with compound 14 (N-dansyl-L-Phe-OCH3) (used as a

standard).

N

O2S NHCOOCH3(L)

14

Each sample (20 L) and dansyl standard 14 (5 L) were sequentially injected into a 250 x

4.6 mm C18 Luna(2) column (Phenomenex) with a flow rate of 1.5 mL/min. Mobile phase: A

10% acetonitrile, 25% methanol, 65% water; B 10% acetonitrile, 90% methanol using a

gradient of 55% B to 100% B over 30 min using two 510 pumps, a 717+ refrigerated

autosampler, and a 474 fluorescence detector (excitation 380 emission 470), all controlled

Page 42: Novel mechanism-based inhibitors of serine proteases

33

by Millenium software (Waters). The fraction eluting from 10 to 11.5 min was collected and

analyzed by electrospray ionization (ESI) and collision-induced dissociation (CID) mass

spectrometry using a Finnigan LCQ-DeccaTM ion-trap mass spectrometer (Thermoquest).

Page 43: Novel mechanism-based inhibitors of serine proteases

34

CHAPTER 4

RESULTS AND DISCUSSION

4.1 Inactivation of HLE by Derivatives of (I) (Compounds 8-13)

SN

O

O OSO2 NHR

( I )

R= CH2CHPh2 (8) CH (CH2Ph ) COOCH3 (9)

( L - isomer ) CH (CH2Ph ) COOCH3 (10)

( D - isomer ) benzyl (11) phenethyl (12)

N

O

SO2 NHO

Ph

COOCH3

(13)

Figure 4.1 Structures of Compounds 8-13

(A) Relationship of Structure to Inhibitory Potency and Specificity

The inhibitory activity of compounds 8-13 toward HLE was evaluated using the

incubation method. The potency of the inhibitors was expressed in terms of the bimolecular

rate constants (kobs/[I] M-1 s-1), and these are listed in Table 4.1.

Table 4.1 Inhibitory Activity of Compounds 8-13 Towards Human Leukocyte Elastase and Bovine Trypsin

kobs/[I] (M-1s-1)Compound

Elastase Trypsin

8 270 80

9 870 290

10 870 320

11 90 Inactive

12 60 Inactive

13 Inactive

Page 44: Novel mechanism-based inhibitors of serine proteases

35

As shown in Table 4.1, with the exception of phthalimide derivative 13, the rest of the

compounds were also found to inhibit HLE. It is evident that inhibitory activity is

dependent on the nature of R. Assuming that R is oriented toward the S subsites, the spatial

requirements observed for R may simply reflect the inability of the saccharin template to bind

to the active site of the enzyme in a strictly substrate-like fashion. Furthermore, in the case of

compound 12, for example, the short alkyl chain (one methylene group) may not be sufficient

to place the phenyl ring close enough to Phe-41 of HLE for an optimal hydrophobic

-stacking interaction. The fact that phthalimide derivative 13 is devoid of any inhibitory

activity suggests that the interaction of the saccharin and phthalimide templates with the

active site involves a poorly-understood delicate interplay of binding and electronic

interactions that affect potency.

Among compounds 8-13, inhibitor 9 is the most potent one. The incubation of HLE with

inhibitor 9 led to rapid time-dependent loss of enzymatic activity (Figure 4.2). The enzyme

slowly regained virtually all of its activity after 24 h. Kitz and Wilson analysis70 of the data

(Figure 4.3) yielded a bimolecular rate constant kobs/[I] of 870 M-1 s-1. Interestingly, the

potency of the corresponding D isomer 10 was comparable to that of the L-isomer (kobs/[I]

870 M-1 s-1).

Page 45: Novel mechanism-based inhibitors of serine proteases

36

Time (hours)

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8 2.0 2.2 24.0

Perc

ent R

emai

ning

Act

ivity

(%)

0

20

40

60

80

100

120

Figure 4.2 Time-dependent Loss of Enzymatic Activity. a) Excess inhibitor 9 ([I]f = 0.21mM) wasincubated with human leukocyte elastase ([E]f = 0.70 M) in 0.1 M HEPES buffer, pH 7.25, 25 oC, aliquotswere withdrawn at different time intervals, and assayed for enzymatic activity using MeOSuc-Ala-Ala-Pro-Valp-nitroanilide ([S]f = 0.14 mM) (solid circles); b) A 300-fold excess of inhibitor 9 was incubated with bovinetrypsin ([E]f = 0.84 M) in 0.1 M Tris buffer, pH 7.51, containing M CaCl2, aliquots were withdrawn at differenttime intervals, and assayed for enzymatic activity using N-p-Tosyl-Gly-Pro-Lys p-nitroanilide ([S]f = 0.1 mM)(open circles).

Page 46: Novel mechanism-based inhibitors of serine proteases

37

Time (min)

0 2 4 6 8

Log

(%R

emai

ning

Act

ivity

)

0.0

0.5

1.0

1.5

2.0

I/E=50

I/E=200

I/E=400

I/E=600

Figure 4.3 Kinetics of Inactivation of Human Leukocyte Elastase ([E]f = 0.70 M) byCompound 9. Inhibitor 9 was incubated with human leukocyte elastase (the [I]/[E] ratio varied between 50 to600) in 0.1 M HEPES buffer, pH 7.25, at 0 oC. Aliquots were withdrawn periodically and assayed for remainingenzyme activity using MeOSuc-Ala-Ala-Pro-Val p-nitroanilide.

The specificity of compounds 8-12 was briefly investigated using bovine trypsin. Thus,

incubation of compound 9 with the enzyme led to rapid inactivation of the enzyme (Table

4.1), followed by gradual and full regain of enzyme activity after 24 h. The regain in

enzymatic activity was somewhat faster when compared to the analogous interaction with

HLE. Replotting of the data yielded a kobs/[I] value of 290 M-1 s-1 (Figure 4.4).

Page 47: Novel mechanism-based inhibitors of serine proteases

38

Time (min)

0 2 4 6 8

Log

(%R

emai

ning

Aci

tivity

)

0.0

0.5

1.0

1.5

2.0

I/E=300

I/E=500

I/E=800

I/E=1000

Figure 4.4 Kinetics of Inactivation of Bovine Trypsin by Compound 9. Excess inhibitor(the [I]/[E] ratio varied between 300 to 1000) was incubated with bovine trypsin ([E]f = 0.84 M) in 0.1 M Trisbuffer, pH 7.51, containing 0.021 M CaCl2. Aliquots were withdrawn at different time intervals and assayed forremaining enzyme activity using N-p-Tosyl-Gly-Pro-Lys p-nitroanilide ([S]f = 0.1 mM).

The results of the studies (Table 4.1) clearly indicate that (a) inhibitor recognition by

these enzymes is critically dependent on interactions with the S subsites of the enzyme, and

(b) potent inhibitors of trypsin-like enzymes can, in principle, be designed that primarily

exploit S interactions only and do not incorporate in their structure a Lys or Arg side chain.

Since the presence of the latter is associated with poor pharmacokinetics and low selectivity,

optimized derivatives of (I) may offer several distinct advantages.

Page 48: Novel mechanism-based inhibitors of serine proteases

39

(B) Mechanism of Inactivation

SN

O

O OSO2

HO Ser195 EHis57

NHR

( I )

SN

O OSO2 NHR

Ser195 EHis57-O O

SN

O

O O

OSer195 E

His57

SNH

O

O O

OSer195

His57E

H2O

SHN

O

O O

OSer195 E

His57

OH SNH2

O

O O

OSer195 E

His57

HO Ser195 EHis57

+ Low molecularweight product

( III ) ( IV )

( V )

Figure 4.5 Postulated Mechanism of Action of Inhibitor (I)

The proposed tentative mechanism of action of (I) (Figure 4.5) was probed as follows: a

substrate protection experiment was carried out in order to demonstrate that the interaction of

(I) with HLE involves the active site. This is clearly evident in Figure 4.6

Page 49: Novel mechanism-based inhibitors of serine proteases

40

Time (min)

0 2 4 6 8 10

Log

(% R

emai

ning

Act

ivity

)

1.0

1.2

1.4

1.6

1.8

2.0

2.2

Figure 4.6 Substrate Protection. HLE (219 nM) was incubated with inhibitor 9 (21.9 M)in 0.1 M HEPES buffer, pH 7.25, aliquots were taken at different time intervals, and assayed for remainingenzyme activity using MeOSuc-Ala-Ala-Pro-Val p-nitroanilide (77.2 M). The experiment was repeated byincubating HLE (219 nM), inhibitor 9 (21.9 M) and MeOSuc-Ala-Ala-Pro-Val p-nitroanilide (0.232 mM) in 0.1 M HEPES buffer, pH 7.25. Aliquots were taken at different time intervals andassayed for remaining enzyme activity using MeOSuc-Ala-Ala-Pro-Val p-nitroanilide (77.2 M). The data wasthen analyzed using the method of Kitz and Wilson .

where the presence of substrate in the incubation mix led to a decrease in kobs/[I] from 320 to

220 M-1 s-1. Next, the possible formation of an HLE-inhibitor acyl enzyme complex (or

complexes) was investigated by adding excess hydroxylamine (0.50 M) in 0.1 M HEPES

buffer, pH 7.25) to HLE that had been fully inactivated with a 100-fold excess of inhibitor 9,

and the regain in enzymatic activity was monitored over a 24 h period (Figure 4.7).

Page 50: Novel mechanism-based inhibitors of serine proteases

41

Time (hour)

0.0 0.5 1.0 1.5 2.0 25.0

Per

cent

Rem

aini

ng A

ctiv

ity

0

10

20

30

40

50

60

70

80

90

100

Figure 4.7 Effect of Hydroxylamine on Enzyme Reactivation. Human leukocyte elastase(219 nM) was totally inactivated by incubating with a 100-fold excess of inhibitor 9 for thirty minutes in 0.1 MHEPES buffer, pH 7.25. Excess hydroxylamine was added (0.045 mM final concentration, closed circles), andaliquots were removed at different time intervals, and assayed for remaining enzyme activity usingMeOSuc-Ala-Ala-Pro-Val p-nitroanilide (77.2 M).

The data suggest that the regain in enzymatic activity arises from the possible

presence of a labile acyl linkage that leads to active enzyme upon treatment with

hydroxylamine. Thus, it can be reasonably assumed that the interaction of the inhibitor with

HLE leads to acylation of the active site serine, however, further structural studies are needed

to arrive at a definitive conclusion regarding the precise structure of the acyl enzyme complex.

The partition ratio, a parameter that corresponds to the number of molecules of inhibitor

necessary to inactivate a single molecule of enzyme, and thus describes how efficiently a

mechanism-based inhibitor inactivates an enzyme, was determined by plotting the fraction of

Page 51: Novel mechanism-based inhibitors of serine proteases

42

remaining enzyme activity after a 15-minute incubation period versus the initial ratio of

inhibitor to enzyme (Figure 4.8).

0.00

20.00

40.00

60.00

80.00

100.00

0 10 20 30 40 50 60 70 80 90 100

[I] / [E]

Perc

ent R

emai

ning

Act

ivity

Figure 4.8 Inactivation of Human Leukocyte Elastase as a Function of the Molar Ratio ofInhibitor 9 to Enzyme. HLE (70 M) and various amounts of inhibitor 9 (0.35-4.20 M in 0.1 M HEPESbuffer, pH 7.25, were incubated for fifteen minutes, aliquots were withdrawn at the end of the incubation period,and assayed for remaining enzyme activity using MeOSuc-Ala-Ala-Pro-Val p-nitroanilide. Thefractional activity remaining is proportional to the molar ratio of inhibitor to enzyme.

The extent of inactivation was found to be linearly dependent on the inhibitor to enzyme

molar ratio. Extrapolation of the linear part of the curve to the line of complete inactivation

yielded a partition ratio of 40 for inhibitor 9, attesting to a moderately efficient inhibitor.

The reactivation of the complex formed between HLE and inhibitor 9 was also

investigated. Thus, HLE was totally inactivated using excess inhibitor 9. The excess inhibitor

was then removed be Centricon-10 filtration, and the regain in enzymatic activity was

monitored. The experiment was repeated using inhibitor 10. In both cases total regain of

Page 52: Novel mechanism-based inhibitors of serine proteases

43

enzymatic activity was observed, suggesting the likely formation of one enzyme-inhibitor

complex. The deacylation rate constants (kdeacyl) for the HLE-inhibitor complexes derived

from inhibitors 9 and 10 were determined by replotting the data according to eq 3.1. These

were found to be 0.0028 and 0.0041 s-1, respectively.

To establish that the interaction of 9 with HLE produces an N-sulfonyl imine that arises

from an enzyme-induced fragmentation process, the products formed by incubating inhibitor

9 with HLE were identified using HPLC and mass spectrometry. Initial HPLC analysis of the

assay solution showed that a trace of (L) Phe-OCH3 was observed in this assay, possibly

arising from the slow hydrolysis of inhibitor 9, which was labeled by dansyl chloride

resulting in a small background. Therefore, a control given identical treatment in each step of

the experiment, except for the addition of HLE, was used in order to subtract out this

background. HPLC analysis of the two samples, after performing the product identification

procedure described under EXPERIMENT, showed that a peak with an identical retention

time to compound 14 standard s peak (10.3 min) was observed in both samples. The sample

including HLE had an area 2.7 times greater than the control, demonstrating that this

compound is a product arising from the HLE-inhibitor 9 reaction (Figure 4.9). The fractions

eluting from 10 to 11.5 min of both the standard 14 (Figure 4.9A) and the HLE-inhibitor

reaction (Figure 4.9C) were collected and analyzed by mass spectrometry. ESI revealed

identical protonated molecule peaks of 413 amu, and CID of the 413 peak of each sample

gave identical gave identical fragmentation patterns, with a principal peak at 301 amu.

Page 53: Novel mechanism-based inhibitors of serine proteases

44

A

B

C

D

Figure 4.9 HPLC Analysis of Products Formed by Incubating Human Leukocyte Elastasewith Inhibitor 9. (a) Standard: compound 14 (N-Dansyl-L-Phe-OCH3); (b) Control: 20 Linjection of assay composed of 300 L of 0.1 M HEPES buffer, pH 7.25, containing 0.5 M NaCl, and 0.2 mMcompound 9 after incubation for 24 h at 23 C, addition of 50 L saturated dansyl chloride in methanol, and 72h incubation at 4 oC.; (c) Compound 9 / HLE reaction: 20 L injection of assay composed of 300 L of 0.1 MHEPES buffer, pH 7.25, with 0.5 M NaCl, 0.2 mM compound 9, and 50 L HLE (1 mg/mL) after incubation for24 h at 23 C, addition of 50 L saturated dansyl chloride in methanol and 72 h incubation at 4 oC; (d) Overlayof panels B and C: the peaks with 10.3 min retention times were one compound and had identical molecularion peaks and fragmentation patterns, corresponding to standard 14.

0.00

20.00

40.00

60.00

80.00

100.00

120.00

140.00

160.00

180.00

200.00

220.00

240.00

260.00

280.00

300.00

Minutes0.00 1.00 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10.00 11.00 12.00 13.00 14.00 15.00 16.00

10.3

01

0.00

20.00

40.00

60.00

80.00

100.00

120.00

140.00

160.00

180.00

200.00

220.00

240.00

260.00

280.00

300.00

Minutes0.00 1.00 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10.00 11.00 12.00 13.00 14.00 15.00 16.00

10.3

43

0.00

20.00

40.00

60.00

80.00

100.00

120.00

140.00

160.00

180.00

200.00

220.00

240.00

260.00

280.00

300.00

Minutes0.00 1.00 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10.00 11.00 12.00 13.00 14.00 15.00 16.00

6.95

0 10.2

95

0.00

20.00

40.00

60.00

80.00

100.00

120.00

140.00

160.00

180.00

200.00

220.00

240.00

260.00

280.00

300.00

Minutes0.00 1.00 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10.00 11.00 12.00 13.00 14.00 15.00 16.00

Page 54: Novel mechanism-based inhibitors of serine proteases

45

Therefore, the product arising from the inhibition of HLE by inhibitor 9 is confirmed to be (L)

Phe-OCH3. The chemical competence of the cascade steps outlined in Figure 4.5 was readily

established by stirring inhibitor 9 with excess sodium methoxide in methanol at room

temperature which led to rapid disappearance of the inhibitor. NMR product analysis revealed

the presence of a roughly 1:1 mixture of (o-carboxymethyl)benzene-sulfonamide and

Phe-OCH3. Taken together, these results suggest that the binding of (I) to the active site of

HLE leads to acylation of the enzyme with concomitant sulfonamide fragmentation resulting

in the release of L-Phe-OCH3, SO2 and the formation of N-sulfonyl imine (III) (Figure 4.5).

The available evidence suggests that (III) ultimately leads to the formation of a stable acyl

enzyme whose structure can be tentatively represented by structure (V). The slow deacylation

rate observed with (V) may be the result of a conformational change in the enzyme that

perturbs the positions of the catalytic residues and/or H-bonding between the SONH2 group

of the tethered inhibitor and the imidazole ring of His-57, thereby impairing the ability of

His-57 to function in general base catalysis. Lastly, while the initial design of inhibitor (I) and

postulated mechanism of action invoked the likely involvement of a double hit mechanism

leading to the formation of species (IV), the available data can be adequately explained by the

formation of a stable acyl enzyme species, particularly in light of earlier high-field NMR

studies62 which demonstrated the formation of formaldehyde from species (IV). In previous

studies using saccharin derivatives of the type Saccharin-CH2X, where X is a good leaving

group (halide, carboxylate, etc.), a similar mechanism involving the formation an N-sulfonyl

imine (structure (III), Figure 4.5) was proposed for the inactivation of HLE51, 36. It should be

noted, however, that compounds represented by (I) are intrinsically more stable chemically

Page 55: Novel mechanism-based inhibitors of serine proteases

46

and employ a sulfonamide fragmentation reaction as the driving force for the formation of the

N-sulfonyl imine.

4.2 Inactivation of HLE by Derivatives of (II) (Compounds 4-7)

R2N

P1

SN

O

O O

S NHCOOR

O O

( II )

P1= isobutylR2= benzylR= CH3 (L, L) (4)

(L, D) (5)benzyl (L, L) (6)H (L, L) (7)

Figure 4.10 Structures of Compounds 4-7

(A) Relationship of Structure to Inhibitory Potency and Specificity

All four derivatives of (II) were found to inactivate HLE rapidly and in a time-dependent

fashion (Table 4.2).

Table 4.2 Inhibitory Activity of Compounds 4-7 Toward Human Leukocyte Elastase and Bovine Trypsin.

Compound HLEa Trypsinb

4 (L,L) 6,700 (870)c 480

5 (L,D) 38,800 (870)c 500

6 (L,L) 5,900 350

7(L,L) 9,600 1200akinact/KI M-1 s-1 values (progress curve method) are the average of several runs fit to equation 2 with R2 > 0.999;bkobs/[I] M-1 s-1 values (incubation method) and are the average of two or three determinations;cvalues in parentheses are for the corresponding saccharin derivatives (ref 19).

Page 56: Novel mechanism-based inhibitors of serine proteases

47

The incubation of inhibitor 4 with HLE led to rapid and time-dependent inactivation of

the enzyme (Figure 4.11).

Figure 4.11 Time-dependent Loss of Enzymatic Activity. Excess inhibitor 4 ([I]f = 14 M)was incubated with human leukocyte elastase ([E]f = 0.7 M) in 0.1 M HEPES buffer, pH 7.25, 25 oC, aliquotswere withdrawn at different time intervals, and assayed for enzymatic activity using MeOSuc-Ala-Ala-Pro-Val p-nitroanilide.

The enzyme regained its activity slowly but completely within 24 h (half-life for enzyme

reactivation ~ 3 h). The kinact/KI M-1 s-1 values for inhibitors 4-7 are listed in Table 4.2, along

with the values for the corresponding saccharin derivatives for compounds 4 and 5 (in

parentheses). It is clearly evident from these results that the 1, 2, 5-thiadiazolidin-3-one 1,1

dioxide scaffold is superior to the saccharin scaffold, yielding highly potent inhibitors of HLE.

This is in agreement with the design that derivatives of (II) can bind to the enzyme in a more

Time (h)

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8 2.0 2.2 2.4 2.6 2.8 3.0 3.2 24.0

Per

cent

Rem

aini

ng A

ctiv

ity

0

20

40

60

80

100

120

Page 57: Novel mechanism-based inhibitors of serine proteases

48

substrate-like fashion by varying the P1 to satisfy the enzyme specificity.

Surprisingly, the most potent inhibitor was found to be diastereomer 5, and this was

significantly more potent than diastereomer 4, suggesting that the binding of 5 to the active

site leads to a more effective - stacking interaction between Phe-41 and the phenyl group in

the inhibitor. Furthermore, because the S and S´ subsites of HLE are fairly hydrophobic, the

enzyme shows a strong preference for hydrophobic substrates and inhibitors. Thus, the

relatively high potency of inhibitor 7, despite the presence of a polar group, is also surprising.

Clearly, further structural studies are needed in order to gain a better understanding of these

observations. Taken together, these findings suggest that inhibitors having optimized potency

and pharmacokinetics can be realized using this type of mechanism-based inhibitor.

Previous studies have shown that the attachment of various leaving groups (halogen,

carboxylate, heterocyclic sulfide, sulfone, etc.) to the 1, 2, 5-thiadiazolidine-3-one 1,1 dioxide

scaffold yields highly potent inhibitors of HLE62-69. Compared to those inhibitors,

compounds 4-7 are less potent by one to two orders of magnitude, however, sulfonamide

derivatives 4-7 are intrinsically more stable chemically and appear to strike a better balance

between potency and chemical stability.

An intriguing observation, previously made with the corresponding saccharin derivatives

(19), was their observed inhibitory activity toward bovine trypsin. Thus, despite the fact that

those compounds lacked a basic side chain (the primary substrate specificity residue P1 for

trypsin is Lys or Arg), the compounds did exhibit inhibition toward trypsin, suggesting that

favorable binding interactions with the S´ subsites are sufficient for bestowing inhibitory

activity, and that the presence of a basic side chain is not necessary for inhibitory activity.

Page 58: Novel mechanism-based inhibitors of serine proteases

49

Based on those considerations, the inhibitory activity of compounds 4-7 toward trypsin was

investigated. All four compounds were found to be time-dependent inhibitors of trypsin. For

example, incubation of compound 7 with trypsin led to rapid and efficient time-dependent

loss of enzyme activity (Figure 4.12).

Figure 4.12 Time-dependent Loss of Enzymatic Activity. Excess inhibitor 7 (at the indicated inhibitor toenzyme molar ratios) was incubated with 10 L bovine trypsin ([E]f = 5.0 M) in 0.025 phosphate buffercontaining M NaCl, pH 7.51, 25 oC, aliquots were withdrawn at different time intervals, and assayed forenzymatic activity using N -benzoyl-L-Arg p-nitroanilide.

Only partial regain of enzymatic activity was observed after 24 h, and the extent of

reactivation was found to be dependent on the inhibitor/enzyme ratio used. Replotting of the

data according to equation 1 yielded the observed rate constant of enzyme inactivation (kobs)

and the potency of the inhibitors was then expressed as the bimolecular rate constant, kobs/[I]

M-1 s-1 (values are listed in Table 4.2). Assuming that these compounds bind to the active

site of trypsin, then the results suggest that favorable binding interactions with the S´ subsites

Time (h)

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8 2.0 2.2 24.0

Perc

ent R

emai

ning

Act

ivity

(%)

0

20

40

60

80

100

120

I/E=5

I/E=20

Page 59: Novel mechanism-based inhibitors of serine proteases

50

can be exploited and used to guide the design of potent inhibitors of trypsin and trypsin-like

serine proteases that lack a basic side chain. The presence of a basic side chain in thrombin

inhibitors has been shown to affect adversely oral bioavailability. Compounds 4-7 were

inactive toward papain, a prototypical cysteine protease, when incubated with the enzyme at

an [I]/[E] ratio of 250 for 0.5 h.

(B) Mechanism of Inactivation

Derivatives of scaffold (II) were envisioned to inactivate HLE by the mechanism shown

in Figure 4.13, where binding to the active site of the enzyme is followed by nucleophilic

attack by Ser195 to form a tetrahedral adduct. Collapse of the adduct via a sulfonamide

fragmentation-driven process as anticipated to generate a highly reactive electrophilic species

(an N-sulfonyl imine) which could be trapped by the active site histidine (His57) to form

enzyme-inhibitor adduct (A) (Figure 4.13, path (a)), or react with water to form a stable acyl

enzyme (B) (Figure 4.13, path (b)). It should also be noted that, since the P1 group2 in

inhibitor (II) serves as the primary specificity residue that is accommodated at the primary

specificity subsite (S1) of a target enzyme, enzyme selectivity can be attained by simply

varying the nature of P1. In the present case, since HLE shows a strong preference for

medium size hydrophobic side chains (such as those of Val and Leu), (L) Leu was choosen as

P1. Furthermore, in order to enhance the binding interactions of (II) with the S´ subsites of the

enzyme, an aromatic residue (Phe) was linked to the heterocyclic scaffold for a favorable

-stacking interaction with Phe-41 (located close to the S2´ subsite). Interactions with the S´

subsites were also probed by investigation of the corresponding inhibitor derived from D-Phe

Page 60: Novel mechanism-based inhibitors of serine proteases

51

methyl ester. Lastly, in order to optimize aqueous solubility without compromising potency,

the presence of a polar group was also investigated.

P1

SN

O

O OSO2

HO Ser195 EHis57

NH

( II )

SN

O OSO2 N

H

Ser195 EHis57-O O

SN

O

O O

OSer195 E

His57

SNH

O

O O

OSer195

His57E

H2O

R2 SNH2

O

O O

OSer195 E

His57

R2

COOR

Ph

P1

R2

COOR

Ph

P1

R2

path (b)

path (a)P1

R2

P1

( A )

( B )

Figure 4.13 Postulated Mechanism of Inhibition ofHuman Leukocyte Elastase by Inhibitor (II).

The complete recovery of enzymatic activity (Figure 4.11) suggests that the interaction

of (II) with HLE leads to the eventual formation of a relatively stable acyl enzyme (Figure

4.13, structure B). It s not intuitively obvious what the determinants of acyl enzyme (B)

stability are, although it s tempting to speculate that the SO2NH2 may be involved in a

Page 61: Novel mechanism-based inhibitors of serine proteases

52

H-bonding interaction with His-57, impeding its ability to function in general base catalysis.

Subtle differences in the make up of the active sites of HLE and trypsin may account for the

differences in the observed deacylation rates for the two enzymes.

Page 62: Novel mechanism-based inhibitors of serine proteases

53

CHAPTER 5

CONCLUSIONS

In conclusion, two novel classes of mechanism-based inhibitors of serine proteases that

incorporate in their structure either an appropriately-functionalized saccharin scaffold, or a 1,

2, 5-thiadiazolidin-3-one 1, 1 dioxide scaffold capable of generating a highly reactive

Michael acceptor by an enzyme-induced sulfonamide fragmentation process. The inactivation

of the enzyme by these inhibitors was found to be efficient, time-dependent and to involve

the active site. Biochemical studies show that the interaction of these inhibitors with HLE

results in the initial formation of a Michaelis-Menten complex and subsequent formation of a

tetrahedral intermediate with the active site serine (Ser-195). Collapse of the tetrahedral

intermediate with tandem fragmentation results in the formation of a highly reactive

conjugated sulfonyl imine which can either react with water to form a relatively stable acyl

enzyme and/or undergo a Michael addition reaction with an active site nucleophilic residue

(His-57). The results also demonstrate convincingly the superiority of the 1, 2,

5-thiadiazolidin-3-one-1, 1-dioxide scaffold over the saccharin scaffold in the design of

inhibitors of (chymo)trypsin-like serine proteases.

Page 63: Novel mechanism-based inhibitors of serine proteases

54

LIST OF REFERENCES

Page 64: Novel mechanism-based inhibitors of serine proteases

55

1. Donald Voet; Judith G. Voet; Charlotte W. Pratt Fundamentals of Biochemistry John

Wiley & Sons, Inc. 1998, 282-283

2. Schechter, I.; Berger, A. Biochem. Biophys. Chem. Commun. 1968, 27, 157-162

3. H. John. Smith; Claire. Simons. Proteinase and Peptidase Inhibition; Taylor & Francis:

London and New York, 1985, 2-5

4. Jerry W Skiles & Arco Y Jeng Leukocyte elastase and macrophage metalloelastase

inhibitors Ashley Publications Ltd. 1999, 870

5. Janoff A, Neutral Proteases of Human Polymorphonuclear Leukocytes. Urban and

Schwartzenberg, Baltimore, USA 1978, 390-417

6. Janoff A, Am. Respir. Dis. 1985, 132, 417-433

7. Ekerot L.; Ohlsson K.; Adv. Exp. Med. Biol. 1984, 167, 335-344

8. Merritt TA; Cochrane CG; Holcomb K; Bohl B et al. J. Clin. Invest. 1983, 72, 656-666

9. Sanders E; Davies M; Coles A. Renal Physiol. 1980, 3, 335-359

10. Jackson AH; Hill SL; Afford SC; Stockley RA. J. Respir. Dis. 1984, 65, 114-124

11. Suter S; Schaad UB; Roux L; Nydegger UE; Waldvogel FA. J. Infect. Dis. 1984, 149,

523-531

12. Inada M.; Yamashita J-I.; Nakano S.; Ogawa M. Anti-Cancer Res. 1998, 18, 885-890

13. D. J. Matthews; J. A. Well. Science 1993, 260, 1113-1117

14. M. M. Smith; L. Shi; M. Navre. J. Biol. Chem. 1995, 270, 6440-6449

15. A. Bevan; C. Brenner; R. S. Fuller. Proc. Natl. Acad. Sci. USA 1998, 95, 10384-10389

16. S. Kothakota; T. Azuma; C. Reinhard; A. Klippel; J. Tang; K. Chu; T. J. McGarry; M, W.

Kirschner; K. Koths; D. J. Kwiatkowski. Science 1997, 278, 294-298

17. Dustin J. Maly; Lily Huang; Jonathan A. Ellman. Chembiochem 2002, 3, 16-37

18. Silverman, R. B. The Organic Chemistry of Enzyme-Catalyzed Reactions (revised

edition); Academic Press; San Diego, 2002;

19. Groutas, W. C.; Giri, P. K.; Crowley, J. P.; Castrisos, J. C.; Brubaker, M. J. Biochem.

Biophys. Re.s Comm. 1986, 141, 741

Page 65: Novel mechanism-based inhibitors of serine proteases

56

20. Groutas, W. C.; Brubaker, M. J.; Stanga, M. A.; Castrisos, J. C.; Crowley, J. P.; Schatz, E.

J. J. Med. Chem. 1989, 32, 1607

21. Groutas, W. C.; Venkataraman, R.; Brubaker, M. J.; Stanga, M. A. Biochemistry 1991,

30, 4132-4136

22. Groutas, W. C.; Venkataraman, Radhika; Brubaker, Michael J.; Epp. Jeffrey;

Chong, Lee S.; Stanga, Michael A.; McClenahan, Jerald J.; Tagusagawa, Fusao.

Biochim. Biophys. Acta 1993, 1164(3), 283-8

23. Groutas, W. C.; Stanga, M. A.; Castrisos, J. C.; Schatz, E. J. J. Enz. Inh. 1990, 3, 237-243

24. Groutas, William C.; Huang, He; Epp, Jeffrey B.; Venkataraman, Radhika; McClenahan,

Jerald J.; Tagusagawa, Fusao. Biochim. Biophys. Acta 1994, 1227(3), 130-6

25. Neumann, U; Gutschow, M. J. Biol. Chem. 1994, 269, 21561-21567

26. Kerrigan, J. E.; Shirley, J. J. Bioorg. Med. Chem. Lett. 1996, 6, 451-456

27. Groutas, William C.; Castrisos, James C.; Stanga, Michael A.; Kuang, Rong Ze;

Venkataramam, Radhika; Epp, Jeffrey B.; Brubaker, Michael J.; Chong, Lee S. Bioorg.

Med. Chem. Lett. 1993, 3(6), 1163-8

28. Groutas, William C.; Stanga, Michael A.; Brubaker, Michael J. J. Am. Chem. Soc. 1989,

111(5), 1931-2

29. Vagnoni, L. M.; Gronostaj, M.; Kerrigan, J. E. Bioorg. Med. Chem. 2001, 9, 637-645

30. Kerrigan, J. E.; Walters, M. C.; Forrester, K. J.; Crowder, J. B.; Christopher, L. K. Biorg.

Med. Chem. 2000, 10, 27-30

31. Hill, J. H. M. J. Org. Chem. 1965, 30, 620-622

32. Groutas, William C.; Houser-Archield, Nadene; Chong, Lee S.; Venkataraman, Radhika;

Epp, Jeffrey B.; Huang, He; McClenahan, Jerald J. J. Med. Chem. 1993, 36(21), 3178-81

33. Groutas, William C.; Huang, He; Venkataraman, Radhika; Houser-Archield, Nadene;

Epp, Jeffrey B. Bioorganic & Medicinal Chemistry 1993, 1(4), 273-7

34. Groutas, William C.; Epp, Jeffrey B.; Venkataraman, Radhika; Kuang, Rongze; Truong,

Tien My; McClenahan, Jerald J.; Prakash, Om. Bioorg. Med. Chem. 1996, 4(9),

1393-1400.

Page 66: Novel mechanism-based inhibitors of serine proteases

57

35. Groutas, William C.; Chong, Lee S.; Venkataraman, Radhika; Kuang, Rongze; Epp, Jeff

B.; Houser-Archield, Nadene; Huang, He; Hoidal, John R Arch. Biochem. Biophys. 1996,

332(2), 335-340

36. Hlasta, D. J.; Subrmanyam, C.; Bell, M. R.; Carbateas, P. M.; Court, J. J.; Desai, R. C.;

Drozd, M. L.; Eickhoff, W. M.; Ferguson, E. W.; Gordon, R. J.; Johnson, J. A.; Kumar,

V.; Maycock, A. L.; Mueller, K. R.; Pagani, E. D.; Robinson, D. T.; Saindane, M. T.;

Silver, P. J.; Subramanian, S.; Dunlap, R. P.; Franke, C. A; Mura, A. J.; Rowlands, A. G.

J. Med. Chem. 1995, 38, 739-744

37. Groutas, W. C.; Chong, L. S.; Venkataraman, R. Biochem. Biophys. Res. Comm. 1993,

197, 730-739

38. Kuang, Rongze; Venkataraman, Radhika; Ruan, Sumei; Groutas, William C.

Bioorg. Med. Chem. Lett. 1998, 8(5), 539-544

39. Zhong, Jiaying; Groutas, William C. Current Topics in Medicinal Chemistry (Sharjah,

United Arab Emirates) 2004, 4(12), 1203-1216

40. Stang, P; Lydick, E.; Silberman, C.; Kempel, A.; Keating, E. T. Chest 2000, 117,

354S-359S

41. Croxton, T. L.; Weinmann, G. G.; Senior, R. M.; Hoidal, J. R. Am. J. Respir.

Crit. Care Med. 2002, 165, 838-844

42. Calverley, P. M.; Walker, P. ; Lancet 2003, 362, 1053-1061

43. Warren, L. L.; Downey, G. P. ; Am. J. Respir. Crit. Care Med. 2001,164, 896-904.

44. Shapiro, S. D. Eur. Respir. J. Suppl. 2003, 44, 30S-32S

45. Stockley, R. A. Am. J. Respir. Crit. Care Med. 1999, 160, S49-S52

46. Kawabata, K.; Hagio, T.; Matsuoka, S. Eur. J. Pharmacol. 2002, 451, 1-10

47. Moraes, J.; Chow, C. W.; Downey, G. P. Crit. Care Med. 2003, 31, S189-S194

48. Silverman, R. B. Meth. Enzymol. 1995, 249, 240-283

49. Sommerfeld, T.; Seebach, D. ; Angew. Chem. Int. Ed. 1995, 34, 553-554

50. Paik, S.; White, E. H. ; Tetrahedron 1996, 52, 5303-5318

51. Groutas, W. C., Houser-Archield, N., Chong, L. S., Venkataraman, R., Epp, J. B.,

Huang, H., McClenahan, J. J. J. Med. Chem. 1993, 36, 3178-3181

Page 67: Novel mechanism-based inhibitors of serine proteases

58

52. Hlasta, D. J.; Subramanyam, C.; Bell, M. R.; Carabateas, P. M.; Court, J. J.; Desai,

R. C.; Drozd, M. L.; Eickhoff, W. M.; Ferguson, E. W.; Gordon, R. J.; Johnson, J.

A.; Kumar, V.; Maycock, A. L.; Mueller, K. R.; Pagani, E. D.; Robinson, D. T.;

Saindane, M. T.; Silver, P. J.; Subramanian, S. ; J. Med. Chem. 1995, 38, 739-744

53. Groutas, W. C.; Kuang, R.; Venkataraman, R.; Epp, J. B.; Ruan, S.; Prakash, O.

Biochemistry 1997, 36, 4739-4750

54. Kuang, R.; Epp, J. B.; Ruan, S.; Yu, H.; Huang, P.; He, S.; Tu, J.; Schechter, N.

M.; Turbov, J.; Froelich, C. J.; Groutas, W. C. J. Am. Chem. Soc. 1999, 121,

8128-8129

55. Groutas, W. C.; Epp, J. B.; Kuang, R.; Ruan, S.; Chong, L. S.; Venkataraman, R.;

Tu, J.; He, S.; Yu, H.; Fu, Q.; Li, Y.; Truong, T. M.; Vu, N. T. Arch. Biochem. Biophys.

2001, 385, 162-169

56. Banzon, J. A.; Kuo, J. M.; Miles, B. W.; Fischer, D. R.; Stang, P. J.; Raushel, F.

M. Biochemistry 1995, 34, 743

57. Schwab, J. M.; Li, W.; Ho, C. J. Am. Chem. Soc. 1984, 106, 7293-7294

58. Georg, G. I.; Harriman, G. C. B.; Peterson, S. A. J. Org. Chem. 1995, 60, 7366-7368

59. Groutas, W. C.; Houser-Archield, N.; Chong, L. S.; Venkataraman, R.; Epp, J. B.;

Huang, H.; McClenahan, J. J. ; J. Med. Chem. 1993, 36, 3178-3181

60. Groutas, W. C.; Epp, J. B.; Venkataraman, R.; Kuang, R.; Truong, T. M.;

McClenahan, J. J. ; Bioorg. Med. Chem. 1996, 4, 1393-1400.

61. Groutas, W. C.; Kuang, R.; Venkataraman, R. Biochem. Biophys. Res. Comm. 1994, 198,

341-349

62. Groutas, W. C.; Kuang, R.; Venkataraman, R., Epp, J. B.; Ruan, S.; Prakash, O.

Biochemistry 1997, 36, 4739-4750

63. Groutas, W. C.; Kuang, R.; Epp, J. B.; Venkataraman, R.; Truong, T. M.; Ruan, S.

Bioorg. Med. Chem. 1998, 6, 661-667.

64. Kuang, R.; Venkataraman, R.; Ruan, S.; Groutas, W. C. Bioorg. Med. Chem. Lett.

1998, 8, 539-544.

65. Kuang, R.; Epp, J. B.; Ruan, S.; Yu, H.; Huang, P.; He, S.; Tu, J.; Schechter, N.

M.; Turbov, J.; Froelich, C. J.; Groutas, W. C. J. Am. Chem. Soc. 1999, 35,

Page 68: Novel mechanism-based inhibitors of serine proteases

59

8128-8129

66. Kuang, R.; Epp, J. B.; Ruan, S.; Chong, L. S.; Venkataraman, R.; Tu, J.; He, S.;

Truong, T. M.; Groutas, W. C. Bioorg. Med. Chem. 2000, 8, 1005-1016

67. Groutas, W. C.; Kuang, R.; Venkataraman, R.; Truong, T. M. Bioorg. Med. Chem.

2000, 8, 1713-1717

68. Groutas, W. C.; He, S.; Kuang, R.; Tu, J., Chan, H-K. Bioorg. Med. Chem. 2001,

9, 1543-1548

69. Groutas, W. C.; Epp, J. B.; Kuang, R.; Ruan, S.; Chong, L. S.; Venkataraman, R.;

Tu, J.; He, S.; Fu, Q.; Li, Y-H.; Truong, T. M.; Vu, N. Arch. Biochem. Biophys. 2001,

385, 162-169

70. Kitz, R.; Wilson, I. B. J. Biol. Chem. 1962, 12, 2940-2945