INFORMATION TO USERS

This manuscript has been reproduced from the microfilm master. UMI films the text (firectfy from the original or copy submitted. Thus, some thesis and dissertation copies are in typewriter face, while others may be from any type of computer printer.

The quality of this reproduction is dependent upon the quality of the copy submitted. Broken or indistinct print, colored or poor quality illustrations and photographs, print bleedthrough. substandard margins, and improper alignment can adversely affect reproduction.

In the unlikely event that the author did not send UMI a complete manuscript and there are missing pages, these will be noted. Also, if unauthorized copyright material had to be removed, a note will indicate the deletion.

Oversize materials (e.g., maps, drawings, charts) are reproduced by sectioning the original, beginning at the upper left-hand comer and continuing from left to right in equal sections with smafl overlaps.

Photographs included in the original manuscript have been reproduced xerographically in this copy. Higher quality 6’ x 9* black and.white photographic prints are available for any photographs or illustrations appearing in this copy for an additional charge. Contact UMI directly to order.

ProQuest Information and Learning 300 North Zeeb Road. Ann Arbor. Ml 48106-1346 USA 800-521-0600

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. POWER LAWS AND DYNAMICS IN MACROSCOPIC & MICROSCOPIC

SYSTEMS: PHARMACEUTICAL PERSPECTIVES

DISSERTATION

Presented in Partial Fulfillm ent o f the Requirements for

the Degree Doctor o f Philosophy in the

Graduate School o f The Ohio State University

By

Teh-Min Hu, M.S.

*****

The Ohio State University 2002

Dissertation Committee:

Dr. W illiam L. Hayton, Adviser Approved by

D r. Kenneth K . Chan

Dr. Susan R. Maflery A dviser Dr. Mark A . Morse College o f Pharmacy

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. UMI Number 3049038

UMI Microform 3049038 Copyright 2002 by ProQuest Information and Learning Company. Ail rights reserved. This microform edition is protected against unauthorized copying under Title 17. United States Code.

ProQuest Information and Learning Company 300 North Zeeb Road P.O. Box 1346 Ann Arbor. Ml 48106-1346

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ABSTRACT

Power-Iaw relationships (y = ax1*) are ubiquitous and extensively studied. The first

part o f my dissertation investigated theoretical and empirical power-law relationships that

are drug-related. Statistical analysis and Monte Carlo simulation were used to

characterize uncertainty in the allometric exponent (b) o f drag clearance. W hile the

individual b values o f 91 drugs generally fe ll w ithin a broad range between 0.2 and 1.2,

the b value from the aggregated clearance values (adjusted to a common a value) was

0.74, with a 99% C l o f 0.71 to 0.76. However, the b value for the predominantly renally

excreted drugs tended towards 0.67. The simulation results suggested that the wide range

o f b values observed for individual drugs could have resulted from random variability in

clearance values determined in the lim ited number o f species used for each drug.

Chapter 3 characterized a novel power-law relationship. Using drug interaction

information reported in 1981,1991 and 1999, a drug-drug interaction network was

described in which the interacting drugs were treated as nodes and were connected with

undirected links that represented interactions. The connectivity o f the resulting network

followed a power-law distribution. The scaling exponent was close to -1 3 and

independent o f the network size. A dynamic model was proposed to account for the

observed scale-free structure o f the network.

i i

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Chapters 4 and 5 experimentally and theoretically explored complex interactions

among pathophysioIogicaQy relevant molecules. Chapter 4 investigated dynamic

interactions among the enzyme superoxide dismutase (SOD), the free radicals nitric oxide

(NO) and superoxide (O O , and the antioxidant glutathione (GSH) in an in v itro model

system. The results showed that SOD's effects on nitrosation were biphasic and dynamic

in nature; i.e., while low concentrations o f SOD were pro-nitrosative, high SOD

concentrations inhibited nitrosation. However, even the in itia lly inhibitory, high SOD

concentrations (> 500 U/ml) became pro-nitrosative over time. SOD predominantly

exhibited the pro-nitrosative effect when GSH was present. Theoretical results in chapter

5 suggested that GSH may modulate the nitrosation reaction in a switch-like manner, and

that concurrently high NO and O f generation may result in nonlinear dynamics o f the

nitrogen oxide species.

i i i i i i

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. To My Parents & T o th e M em ory o f M y G randm a

hr

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ACKNOWLEDGMENTS

I thank my adviser, Dr. W illiam Hayton, fo r inspiration, intellectual support,

encouragement and enthusiasm, which made this dissertation possible. His modest,

rigorous and open-minded attitude toward scientific study has had a great influence on

me. I am sincerely grateful for his patience and insights in editing my scientific writings,

which made me a better scientist I thank Dr. Susan M ailery for giving me the

opportunity to study chemistry and biology o f reactive nitrogen and oxygen species in her

lab, which fed to the findings in Chapters 4 and 5. I am greatly appreciative for the

tremendous time and energy that she spent on our co-authored paper. I am very thankful

for her encouragement, patience and advice, i also express my thanks to Ping Pei for

helping me in Dr. Mallery’s lab. I thank Dr. Morse for the insightful comments and

suggestions for my paper and dissertation. I am also grateful to Dr. Chan for his

invaluable suggestions for my dissertation. Full scholarship was granted by the M inistry

o f National Defense in Taiwan. I wish to thank my previous colleagues in National

Defense Medical Center, Drs. O liver Y-P. Hu, An-Rong Lee, Da-Pang Wang, Li-Chien

Chang and Ming-Kuan Hu, who helped and encouraged me during my study in the U.S.

I am deeply indebted to Sbih-Jiuan fo r tremendous help and support, stimulating

discussions and companionship. Last, but not least, I thank my parents for their

unconditional love, fu ll support and understanding.

v

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. VITA

April 28,1967 ------Bom - Taipei, Taiwan

1988 -1989 ------Undergraduate Research, National Defense

Medical Center (NDMC), Taipei, Taiwan

1989 ------B.S. Pharmacy, NDMC

1989-1991 ------Teaching Associate, NDMC

1991 -1 9 9 3 ------Graduate Research, NDMC

1993 ------M.S. Pharmaceutical Sciences, NDMC

1993 - 1997 ------Teaching and Research Associate, NDMC

1997-present ------Visiting Scholarship, The Ohio State University

PUBLICATIONS

Full Papers

1. Teh-Min Hu and W illiam L. Hayton, AUometric scaling o f xenobiotic clearance: uncertainty versus universality. AAPS PharmSci, 3(4) a rticle 2 9 ,20 01.

2. O liver Yoa-Pu Hu, Hung-Shang Tang, H-Y Lane, W-H Chang and Teh-Min Hu. Novel single-point plasma or saliva dextromethorphan method for determining CYP2D6 a c tiv ity . / . Pharm acol Exp.. Ther. 285(3): 955-960,1998

3. Oliver Yoa-PuHil Teh-Min Hri. B-L Chen and K-M Hm, Various o f n»*e and extent o f absorption in bioequivalent study o f norfloxacin tablet. Chin. Pharm. J. 47:363-376,1995

v i

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 4. Shung-Tai Ho, Jhi-Joung Wang, O liver Yoa-Pu Hu and Teh-Min Hw_ The effect o f aging on the pharmacokinetics o f nalbuphine in rabbits. Biopharm. Drug Dispos. 16: 695-701,1995

5. O liver Yao-Pu Hu, Teh-Min Hu and Hung-Shang Tang, Determination o f galactose in human blood by high performance liquid chromatography: comparison w ith enzymatic method and application to pharmacokinetic study o f galactose in patients w ith liver dysfunction. J. Pharm. ScL 84:231-235,1995

6. An-Rong Lee and Teh-Min H it Determination o f guaiphenesin in anti-tussive pharmaceutical preparations containing dextromethorphan by first- and second- derivative ultraviolet spectrophotometry. /. Pharm. Biomed. Anal. 12:747-752, 1994

7. O liver Yoa-Pu Hu, Teh-Min Hu. Shu-Fen Chiao, Kai-Min Chu, Shu-Fen Chan, Jin- Shing Lai and Ping-Hong Chung, Comparative bioavailability study o f digoxin tablets - an example o f using nonspecific analytical methodology. J. Clin. Pharm. Assoc . 1 :37-47,1992

8. Teh-Min Hn and An-Rong Lee, Simultaneous assay o f aminophylline and phenobarbital in compound tablets by UV spectrophotometry. Chin. Pharm. J. 44: 31-36,1992

A b stra cts

1. Teh-Min Hu Susan R. Mallery and W illiam L. Hayton, Kinetic modeling o f nitric oxide reactions in the celL AAPS Annual Meeting and Exposition, Oct. 21-25,2001, Denver, Colorado, USA.

2. Teh-Min Hu and W illiam L. Hayton, AHometric scaling o f drug clearance: variability versus universality. AAPS Annual Meeting and Exposition, Oct. 21-25,2001, Denver, Colorado, USA.

3. Teh-Min Hu. Oliver Yoa-Pu Hu and Hun-Shang Tang, The kinetics o f urinary excretion o f galactose in normal subjects and patients w ith liver diseases. AAPS Tenth Annual Meeting and Exposition, Nov. 5-9,1995, Miami beach, Florida, USA {Pharmaceutical Research, 12(suppl), 9 : S 103,1995).

F IE L D S O F STU D Y

M ajor Field: Pharmacy

vu

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. TABLE OF CONTENTS

Abstract _____ ....______...______i i

Dedication ------iv

Acknowledgments mw...^.«.»...*...... *m...... v

V it a ______v i

List o f Tables ------x i

List o f Figures ------x ii

Chapters:

1. Introduction ------I

2. Allometric Scaling o f Xenobiotic Clearance: Uncertainty versos Universality

2.1 Introduction ------I I 22. Materials and methods ------13 2.2.1 Data collection and statistical analysis ______13 2 2 2 Monte C arlo sim ulation ______13 2.3 Results ...... 14 2 A Discussion ______....______16 2.5 Summaiy 22 2.6 ...... 23

3. Power-Law Scaling o f the Drug-Drug Interaction Network

3.1 Introduction ______44

vrn

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 3 3 Materials and methods ______47 3.2.1 Data acquisition ______47 3 3 3 Data analysis __ 48 3 3 3 Theoretical considerations ______49 33 Results ------50 3.4 Discussion ______52 3.5 Conclusion ______60 3.6 Summary ______60 3.7 References ______61

4. Dynamic Biphasic Effect o f Superoxide Dismutases on Nitric-Oxide-Mediated

Nitrosation Reaction

4.1 Introduction ------....------71 43 Materials and methods ______73 43.1 Materials ______73 433 Kinetics of DAN nitrosation ------73 4 3 3 O xidation o f DHR b y SIN-1 ______74 43.4 Cytochrome c reduction assay ______74 43 3 HPLC determination o f SIN-1 degradation kinetics ------75 43.6 Method validation ______....------76 43 Results ------76 4.4 Discussion ------80 43 Conclusion ______88 4.6 Summary ------88 1.7 References ...... 90

5. Kinetic Modeling o f Nitric-Oxide-Associated Reaction Network: Biological

Implications

5.1 Introduction ______106 5 3 M odel ------—...______109 53.1 Reaction chemistry and rate constants ------109 533 Numerical simulations ______110 5 3 R e s u lts ______I I I 53.1 Kinetic profiles ------I I I 5 3 3 GSH as a dynamic sw itch ______111 533 Nonlinear dynamics at high NO and CV input rates ______112 5 3 .4 Phase portraits ..... ______113 5.4 Discussion ______114 5 3 S u m m a ry ______118 ix

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 5*6 **H *t**t**H ****H **H *H H tt*t*»H **<*tM ******H ************M «****H «*H ****(**t*(*»(4*M I(|* ^ (S

Appendices

Appendix A MATHEMATICA Program for the Monte Carlo Simulations ___ 133 Appendix B Release Mechanism o f Nitric Oxide and Superoxide from SIN-1 139

BibEograpliy ------...______.... 141

x

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. LIST OF TABLES

Table Page

2.1 Allometric scaling parameters obtained from linear regressions o f the Iog-!og-transfonned CL versus BW data o f 115 xenobiotics ______33

2.2 Simulated b values in different scenarios w ith varied body weight ranges __ 38

23 Summary ofthe statistical results in Fig. 2.3 ______39

3.1 Basic characteristics o f drug-drug interaction networks ______64

3.2 The top-10 most interacting drugs in the three databases ______65

5.1 The rate constants used for the simulation ------123

5 2 Parameter values for the simulation ______124

x i

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. LIST OF FIGURES

Figpre Page

2.1 The frequency distribution o f the b values for the 91 xenobiotics that showed statistically significant correlation between log CL and log BW in Table 2.1 ------... 40

2 2 The frequency distribution o f the simulated b values in the ten scenarios where the number o f animal species and the range o f body weight were v a rie d ______...__ ___ .... 41

2.3 The relationship between normalized clearances fCI r ^ l and body weights (BW) for the 91 xenobiotics (n = 460) that showed statistically significant correlation between log CL and log BW in Table 2.1 __ .... 42

2.4 The deviation between the fitte d and the observed human C L fo r 68 xi^itics ...... 43

3.1 A model for the drug-drug interaction network ______.... 66

3.2 A drug-drug interaction network consisted o f966 drugs (dots) and 33 interactions (lines) — _____ ..._ 67

3.3 The frequency distribution for the number o f interactions a given drug has. Data were acquired from databases published in 1981 (351 drugs, 3A ), 1991 (636 drugs, 3B) and 1999 (966 drugs, 3C) ______. . . . 68

3.4 The probability degree distributions for the drug-drug interaction networks .... 69

3.5 Cumulative preferential linking for the drug-drug interaction network. The line corresponds to no preferential linking ______.... 70

4.1 The chemical structures o f 3-morphohnosydnommine (SIM-1), 2,3- diaminonaphthalene (DAN) and dihydrorhodamine 123 (D H R ) ______.... 95

xn

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. 4.2 Nitrosation kinetics o f DAN in the absence and presence o f different levels o f Cn^n-S O D ______96

43 Time- and concentration-dependent, biphasic, dose-response relationship for the effect o f Cu,Zn-SOD on DAN nitrosation ------97

4.4 Effect o f DAN concentration on SOD modulation o f nitrosation kinetics ___ 98

4.5 Time- and concentration-dependent, biphasic, dose-response relationships for the effects o f Mn-SOD and Fe-SOD ______99

4.6 Effect o f intact Cu^n-SOD (•), heat-inactivated Cu^n-SOD (O), BSA (■ ), and metal ions (Cu** A,C uv V ,Z n "r O ) on DAN nitrosation ______100

4.7 Effect o f glutathione (GSH) on Cu^ii-SOD modulation o f DAN nitrosation .. 101

4.8 Effect o f Cu,Zn-SOD on SIN-1 mediated oxidation ofDHR ______102

4.9 Effect o f Cn,Zn-SOD on NO release kinetics o f SIN-1 measured by HPLC __ 103

4.10 Comparison o f the nitrosation kinetics among three SOD isozymes at 1000 U /m l ______104

4.11 Proposed mechanisms for SOD’s biphasic effect on SIN-1-mediated nitrosation reaction ...... 105

5.1 The model ...... i...... 125

5.2 Concentration-thne profiles ofNO, 0 2~, ONOO", N20 3, GSNO, and GSH. Scenarios: (a) kt - k2; (b ) k t - 0.5 k2; (c ) kt = 0.25 k2; (d) k t - 0.125 k2; (e) kt = 0.0625 k2. [GSHjbaai = 10 mM, k2 - 1*10'7 M/s for a ll scenarios ___ 126

53 Concentration-time profiles ofNO , 0>“ ONOO" N203, GSNO, and GSH. Scenarios: (a) kt = k2; (b ) kt = 0 3 k2; (c ) kt = 0.25 k2; (d) kt - 0.125 k2; (e) kt = 0.0625 k2. [GSH]b«sai = I m M , k2 = 1*10*7 M/s for a ll scenarios ____ 127

5.4 Concentration-time profiles ofN O , 0 2_, ONOO", N20 3, GSNO, and GSH. Scenarios: (a) k t = k2; (b ) kt = 0 3 (c ) kt = 0 3 5 k2; (d ) kt = 0.125 k2; (e) k ( = 0.0625 fcj. [GSH]bosai= 0 .1 m M , k2 — 1*10"7 M/s for a ll scenarios __ 128

53 Concentration-time profiles o f N^Oj and GSH at the initial GSH level equal to 1 mM. Arrow: start o f zero-order GSH input (kj). k2 = 6 jiM /m in ; kt = 0 3 k2; ks = (a) 20 tiM/m in; (b) 10 pM/min; (c) 5 pM/min; (d ) 2 3 p M /m in; (e) 135 p M /m in ______129

x iii

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. 5.6 Concentration-time profiles ofNO , 0 2”, ONOO- , N->03, GSNO, and GSH. Scenarios: (a) k t — k2; (b) kt = 0.5 k2; (c) kt = 0 2 5 k2; (d) kt = 0.125 k2; (e) k [ = 0.0625 k2^ [GSHJbasai = 10 m M , k2 = 1*IQS M/s for all scenarios _ 130

5.7 (A) Phase plane analysis o f [NO]s.s. (B) [NOlss versos k /k 2, k2= 1*10'5 M/s; kf/ky = (a) 4 ; (b) 2; (c) I ; (d) 0.5; (e ) 0 2 5 ; ( f) 0.125 ______131

5.8 (A ) Phase plane analysis o f [NO]s.s. (B) [NOlss versus k /k 2_ k2= 1*10*® M /s; k /k 2 = (a) 4 ; (b) 2; (c) I ; (d ) 0 .5 ; (e ) 0 2 5 ; (0 0.125 ______132

B .l Mechanism for the release ofNO and O f from SIN-1 ______138

B2 HPLC chromatograms for the decomposition kinetics o f SIN-1 ______139

x iv

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. C H APTER I

INTRODUCTION

Many seemingly unrelated scientific disciplines have recently devoted to study the

emerging properties o f system structures and dynamics.1'15 The system can be as small

and confined as a cell6 or as large and unbounded as the World-Wide Web (WWW).3*5

W hile the interactions among the system elements may be complex and the system

behavior is d ifficu lt to predict, some system properties can be described by a simple

power-law equation.

y=ax* (I)

The idea that one can find sim plicity out o f complexity has attracted scientists from

diverse fields who have pondered how such regularity could exist in nature. A recent

colloquium highlighted the intensity o f growing interests in finding a power-law

relationship in physical, biological, economical and social systems.16

In the pharmaceutical field, a well-known example o f a power-law relationship is

the allometric scaling o f pharmacokinetic (PK) parameters across species.1718 That PK

parameters (clearance, volume o f distribution, half life ) o f a wide variety o f drugs can be

I

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. scaled by equation I, tnw hichy is the PK parameter andx is the body weight, is itse lf an

astounding, yet useful concept The elimination o f a drag from the body involves

complex physiological processes and is dependent on drug characteristics. The existence

o f a simple power-law relationship in describing PK parameters for a variety o f drugs in

different animal species suggested that macroscopicaily drug elimination is governed by

sim ilar structural and functional attributes among species. Although the underlying

principle remained unknown, the concept o f allometric scaling has been widely applied in

predicting PK parameters in human from parameter values determined in animals.19,20

Accordingly, allometric scaling has been an alternate method used for the prediction o f

the first-time-in-man dose during drug development,21,22 in which case the clearance (CL)

values o f a given drug studied in different species were scaled and, in most cases, the CL

value in human can be extrapolated from the obtained scaling relationship from other

animal species.

One o f the problems in applying allometric scaling for dose estimation is the

uncertainty in determining the b value in equation 1. For example, to obtain the b value

in scaling CL, a common procedure is to use linear regression o f the log-log-transformed

CL versus body weight data. This process is subjected to statistical uncertainty when a

lim ited number o f data points are used. However, to my surprise, most studies reported a

b value without addressing the uncertainty issue. When the literature data were

examined, the b value obtained from the scaling o f CL fo r a variety o f xenobiotics tended

to be scattered. Was the scatter o f b values related to the variability in PK properties

among different drugs? Or was it due to the uncertainty in estimation o f this value? The

answers to these questions are important because they may determine how the allometric 2

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. scaling is implemented and applied. Specifically, if different drugs should have different

scaling properties, the current practice o f allometric scaling in the pharmaceutical

sciences should focus on improving the experimental design in order to obtain a more

reliable estimate o f the 6 value. Otherwise, one may start to consider the feasibility o f

using a fixed-exponent approach to a specific group o f drugs when allometric scaling is

used for estimation o f the first-time-in-man dose. The question o f whether a universal

exponent exists in scaling drug CL coincides with a century-long quest for a unifying

theory in the allometric scaling o f basal metabolic rate.23*26 Using statistical analyses and

Monte Carlo simulations, Chapter 2 addressed the uncertainty and universality issues in

allometric scaling o f drug clearance.27

Allometric scaling in pharmacokinetics illustrates that drug disposition in complex

physiological systems can give rise to a power-law relationship when the problem was

examined at a global level. It also signifies that one may find the same regularity across

different disciplines, which motivated the study in Chapter 3. The system described in

Chapter 3 was a growing network which consisted o f drugs that interact with each other.

The structure and the dynamics o f the drug-drug interaction network were characterized

and the connectivity o f a given drug (the number o f interaction a given drug has) was

found to follow a power-law distribution, which again can be represented by equation I.

The finding o f a power-law distribution in the drug-drug interaction network concurred

w ith recent m ultidisciplinary interests in analyzing complex networks,1'15 which are

introduced as follows.

Generally speaking, a network is composed o f nodes (vertices) that are connected

by links (edges). The degree o f a node specifies the number o f links that a node has.1 3

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. Traditionally the study o f complex network has been in the field o f graph theory in

mathematics, h i a random graph, the links are placed randomly among nodes, and the

degree distribution o f a random graph therefore follows a Poisson distribution, which is a

bell-shaped curve w ith a peak at the average degree.1 The question is: are complex

networks in the real world governed by random processes?

Complex networks exist in different forms in nature. For example, the

interconnection o f molecules by chemical reactions inside a cell forms a metabolic

network.6 In a society, networks are formed by direct or indirect interactions o f

individuals.1 Barabasi and Albert2 in 1999 found that for most large real networks the

degree distribution followed power-law degree distribution. In the past three years,

sim ilar findings have been extended to a number o f real networks, such as the WWW3*5

and cellular,6 ecological,7 and scientific collaboration networks8 etc. Moreover,

theoretical models that were able to capture the general feature o f the power-law

distribution have been proposed.9' 14 Two recent reviews1'15 highlighted the intensity o f

growing interest in understanding the topology and evolution o f complex networks.

Drug-drug interaction poses a potential threat to patients who receive treatment with

m ultiple medications. The incidence o f drug interactions w ill be increased when the

number o f co-medicated drugs is increased. The general mechanisms for an interaction

to occur include the pharmacokinetic interactions (or AD ME interactions) by which the

processes o f drug absorption, distribution, metabolism and excretion are affected and the

pharmacodynamic interactions by which the effects o f a drug at its site o f action are

modified.28 Although studies on a particular interaction were occasionally reported and

4

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. drug-drug interaction information has accumulated over time, there has been lack o f a

systematic approach fo r analyzing the aggregate drug-drug interaction information.

I f the interacting drugs were treated as nodes connected w ith undirected links that

represent the interactions, a drug-drug interaction network can be obtained. Questions

pertaining to network analysis then arise: What is the topology o f the drug-drug

interaction network? How does this network evolve as a dynamic system? The answers

to these questions may provide insights into the nature o f drug-drug interactions. In

Chapter 3, we constructed three drug-drug interaction networks from collective drug

interaction information spanning over two decades. The results indicated that the degree

distribution o f all three networks followed a power law w ith an invariant scaling

exponent close to - 1 3 .

The beauty o f a power taw probably rests on its indication that a complex system is

self-organizing.16 A t a microscopic leveL, the details o f the system may be chaodc so that

one may not have a clear idea about how the system w ill behave. When the system is

examined globally, some governing principles o f the system may be revealed in a simple

form .

However, life is not always as simple as the power-law relationship might have

suggested. This is especially true in biological and ecological systems. How does one

account for a sudden change in an ecological or biological state? Why does the same set

o f genes control different phenotypes? Why does the same molecule play a totally

opposite role in physiological and pathophysiological conditions? For example^ the free

radical nitric oxide (NO) is both a tumor promoter and suppressor.29 Most o f these

questions are d ifficu lt to explore experimentally and systematically, because too many 5

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. variables are involved. In the NO case, a subsystem can be isolated and examined both

experimentally and theoretically. Sometimes, one may be amazed by how a simple

model system can give rise to diverse outcomes, which lead to a totally opposite notion as

compared w ith that described for power-law relationships.

The complexity o f a system is often reflected by its dynamics, a subject that deals

w ith change, and w ith systems that evolve in time.30 By studying the dynamics o f a

sim plified model, the complex phenomena observed in the real system can be accounted

for. Some examples are: (i) a biochemical gene switch that can be realized by just two

coupled differential equations;30 (ii) complex dynamic transitions in epidemics can be

understood by a simple mathematical model;31 (iii) models o f coupled biochemical

reactions can predict the emergent properties o f biological signaling networks.32 The use

o f theoretical models provides the following advantages when biological complexity is

dealt w ith :33

• It is possible to ask questions that may be inaccessible to experiments or hard to

address experimentally.

• Through modeling, testable predictions can be formed. Counterintuitive

explanations or surprising predictions are also provided.

• Modeling approach allows a rapid exploration o f different mechanisms and o f a

large range o f conditions. It also allows researcher to identify key parameters

o f the system.

• Modeling approach provides a unified theoretical framework that accounts for

available experimental observations and supports or not experimental

conclusions. 6

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. • Conceptualization by theoretical models leads to clarification o f hypothesis.

• Modeling approach allows analysis o f complex situations in which multiple and

coupled variables are involved, for which sheer intuition provides no insight.

In Chapters 4 and 5, examples were given to demonstrate how systems can be

examined experimentally and theoretically in a dynamic sense. Because the studies were,

by their very nature, dynamic, we were able to identify some important features

associated w ith the enzyme superoxide dismutase and the reactions mediated by NO.

From pharmaceutical perspectives, it is important for pharmaceutical scientists to

have both global and dynamic views on biochemical, cellular and physiological function,

toward the objective o f improved understanding o f the disease state.

References

1. Albert R, Barabasi AL. Statistical mechanics o f complex networks. Mod. Phys. 2002;74:47-97.

2. Barabasi AL, Albert R. Emergence o f scaling in random networks. Science. 1999;286:509-512.

3. Albert R, Jeong H, Barabasi AL. The diameter ofthe world-wide web. Nature. I999;40I:I30.

4. Huberman BA, Adamic LA. Growth dynamics ofthe world-wide web. Nature. 1999;40I:l3l.

5. Adamic LA, Huberman BA. Power-law distribution o f the world wide web. Science. 2000^87:2115.

6. Jeong H, Tombor B, Albert R, Oltvai ZN, Barabasi AL. The large-scale organization o f metabolic networks. Nature. 2000;407:651-654.

7

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. 7. Montoya JM, Sole RV. Small world patterns in food webs. AxXiv.org e-Print archive. 2000. http://www.arxiv.org/abs/cond-mat/0011195

8. Newman MEJ. The structure o f scientific collaboration networks. Proc Natl Acad SciUSA. 2001;98:404-409.

9. Barabasi AL, Albert R, Jeong H. Mean-fieid theory for scale-free random networks. Physica A. 1999;272:173-187.

10. Krapivsky PL, Redner S, Leyvraz F. Connectivity o f growing random networks. Phys. Rev. Lett 2000;85:4629-4632.

11. Dorogovtsev SN, Mendes JFF, Samukhin AN. Structure o f growing networks with preferential linking. Phys. Rev. Lett 2000;85:4633-4636.

12. Albert R, Barabasi AL. Topology o f evolving networks: Local events and universality. Phys. Rev. Lett 2000;85:5234-5237.

13. Kirillova OV. Communication networks with an emergent dynamical structure. Phys. Rev. Lett 2001;87:06870I;l-4.

14. Dorogovtsev SN, Mendes JFF. Scaling properties o f scale-free evolving networks: Continuous approach. Phys. Rev. E. 200I;63:056I25;l-I9 .

15. Dorogovtsev SN, Mendes JFF. Evolution o f networks. ArXiv.org e-Print archive. 2001. http://www.arxiv.org/abs/ cond-mat/0106144

16. Turcotte DL, Rundle JB. Self-organized complexity in the physical, biological, and social sciences. Proc Natl Acad Sci USA. 20G2;99(suppL 1)^463-2465.

17. Boxenbaum H. Interspecies scaling allometry, physiological time, and the ground plan o f pharmacokinetics. J Pharmacokin Biopharm. 1982;10:201-227.

18. Mordenti J. Man versus beast Pharmacokinetic scaling in mammals. J Pharm Sci. 1986;75:1028-1040.

8

Reproduced with permission ofthe copyright owner. Further reproduction prohibited without permission. 19. Sawada Y, Hanano M, Sugiyama Y, Iga T. Prediction o f disposition o f beta-Iactam antibiotics in humans from pharmacokinetic parameters in animals. J Pharmacokin Biopbann. 1984;12:241-261.

20. Feng MR, Lou X, Brown RR, Hutchaleelaha A. Allometric pharmacokinetic scaling: Towards the prediction o f human oral pharmacokinetics. Pharm Res. 2000;17:410-418.

21. Bonate PL, Howard D. Prospective allometic scaling: Does the emperor have clothes? J C lin Pharmacol. 2000;40:665-670.

22. Mahmood I. Critique o f prospective allometric scaling: Does the emperor have clothes? J C lin Pharmacol. 2000;40:671-674.

23. West GB, Brown JH, Enquist BJ. A general model for the origin o f allometric scaling laws in biology. Science. 1997;276:122-126.

24. West GB, Brown JH, Enquist BJ. The fourth dimension o f life : Fractal geometry and allometric scaling o f organisms. Science. 1999;284:1677-1679.

25. Banavar JR, Mari tan A, Rinaldo A. Size and form in efficient transportation networks. Nature. 1999;399:130-132.

26. Dodds PS, Rothman DH, Weitz JS. Re-examination o f the "3/4-Iaw" o f metabolism. J Theor Biol. 2001;209:9-27.

27. Hu TM, Hayton WL. Allometric scaling o f xenobiodc clearance: Uncertainty versus universality. AAPS PharmSci. 20013(4), article 29. (http://www.phannsci.org/)

28. Stockley IH. Drug interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, London, UK: Pharmaceutical Press, 1999.

29. Wink DA, Vodovotx Y, Laval J, Laval F, Dewhirst MW, M itchell JB. The multifaceted roles o f nitric oxide in cancer. Carcinogenesis. I998;19(5):7l 1-721.

9

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 30. Strogatz SH. Nonlinear dynamics and chaos. Reading M A: Addison-Wesley Publishing Company, 1994.

31. Earn DJD, Rohani P, Bofker MB, Grenfell BT. A simple model for complex dynamical transitions in epidemics. Science. 2000;287:667-670.

3 2 . Bhalla U S , Iyengar R . Emergent properties o f networks o f biological signaling pathways. Science. 1999;283:381-387.

33. Leloup J-C, Goldbeter A. Modeling the molecular regulatory mechanism o f circadian rhythms in Drosophila. Bioessays. 2000;22:84-93.

10

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. C H AP TER 2

Allom etric Scaling o f Xenobfotic Clearance: Uncertainty versus Universality

2.1 Introduction

Biological structures and processes from cellular metabolism to population

dynamics are affected by the size o f the organism.1'2 Although the sizes o f m am m alian

species span 7 orders o f magnitude, interspecies sim ilarities in structural, physiological

and biochemical attributes result in an empirical power law (the allometric equation) that

characterizes the dependency o f biological variables on body mass:

Y = a BW b

where Y is the dependent biological variable o f interest, a is a normalization constant

known as the aQometric coefficient. BW is the body weight, and b is the allometric

exponent. The exponential form can be transformed into a linear function:

LogY = Log a + b(LogBW)

and a and b can be estimated from the intercept and slope o f a linear regression analysis.

The magnitude o f b characterizes the rate o f change o f a biological variable subjected to

a change o f body mass and reflects the geometric and dynamic constraints o f the body.3'4

Although allometric scaling o f physiological parameters has been a century-long

II

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. endeavor, no consensus has been reached as to whether a universal scaling exponent

exists. In particular, discussion has centered on whether the basal metabolic rate scales as

the 2/3 or 3/4 power o f the body mass.1'2,3*9

Allometric scaling has been applied in pharmacokinetics for approximately two

decades. The major interest has been prediction o f pharmacokinetic parameters in man

from parameter values determined in animals.10'15 Clearance has been the most studied

parameter, as it determines the drug-dosing rate. In most cases, the pharmacokinetics o f a

new drug was studied in several animal species and the allometric relationship between

pharmacokinetic parameters and the body weight was determined using linear regression

o f the log-transformed data. One or more o f the following observations frequently apply

to the resulting publications: (i) little attention was given to uncertainty in the a and b

values; while the correlation coefficient was frequently reported, the confidence intervals

o f the a and b values were infrequently addressed, (ii) The a and b values were used for

interspecies extrapolation o f pharmacokinetics without analysis o f the uncertainty in the

predicted parameter values, (iii) The b value o f clearance was compared w ith either the

value 2/3 from “surface law” or 3/4 from “Kleiber’s law” and the allometric scaling o f

basal metabolic rate.

This study investigated the possible impact o f the uncertainty in allometric scaling

parameters on predicted pharmacokinetic parameter values. A statistical analysis o f the

allometric exponent o f clearance from I IS xenobiotics and a Monte Carlo simulation was

combined to characterize the uncertainty in the allometric exponent for clearance and to

investigate whether a universal exponent may exist for the scaling o f xenobiotic

clearance. 12

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 12 Materials and Methods

2.2.1 D ata c o lle ctio n and s ta tis tic a l analysis

Clearance (CL) and BW data fo r 115 substances were collected from published

studies in which at least three animal species were used for the purpose o f interspecies

comparison o f pharmacokinetics. A total o f 18 species (16 mammals, 2 birds) with body

weight spanning I04 were involved (Table 2.1).16"90 The published studies generally did

not control or standardize across species the (i) dosage, (if) numbers o f individuals

studied per species, (iii) principal investigator, (iv) blood sampling regime, and (v)

gender.

Linear regression was performed on the log-transformed data according to the

equation. Log CL = log a + b * log BW. Values for a and b were obtained from the

intercept and the slope o f the regression, along with the coefficient o f determination (r).

Statistical inferences about b were performed in the following form:

Ho: b = pi

H i:b*pbi«0,I,2

Where po= 0, pi = 2/3 and P? =3/4, respectively. The 95% and 99% confidence intervals

(C l) were also calculated for each b value.

h i addition, the CL values for each individual xenobiotic were normalized so that

a ll compounds bad the same a value. Linear regression analysis was applied to the

pooled, normalized CL versus BW data fo r the 91 xenobiotics that showed sta tistica lly

significant correlation between log CL and log BW in Table 2.1.

2.2.2 M onte C a rlo sim ula tion

13

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The power function CL = a BW b was used to generate a set o f error-free CL

versus BW data (Appendix A). The values for BW were 0.02,0.25,2.5,5,14 and 70 kg,

which represented the body weights o f mouse, rat, rabbit, monkey, dog and human,

respectively. The values o f a and b used in the simulation werelOO and 0.75,

respectively. Random error was added to the calculated CL values, assuming a normal

distribution o f error with either a 20% or a 30% coefficient o f variation (CV), using the

function RANDOM in Mathematics 4.0 (Wolfram Research, Champaign, IL). The b and

r values were obtained by applying linear regression analyses on the log-log- transformed

error-containing CL versus BW data using the Mathematics function REGRESS. Ten

scenarios w ith a variety o f sampling regimens that covered different numbers o f animal

species (3 - 6) with various body weight ranges (5.6 - 3500 fold) were simulated (n =

100 per scenario). The simulations mimicked the sampling patterns commonly adopted

in the published interspecies pharmacokinetics studies.

2 3 Results

The aQometric scaling parameters and their statistics are listed in Table 2.1. O f 115

compounds, 24 (—21%) showed no correlation between clearance and body weight; i.e.,

there was a lack o f statistical significance fo r the regression (p > 0.05). This generally

occurred when only 3 species were used. Among the remaining 91 cases, the mean ±

SJ3. o f the b values was 0.74 ±0.16 with a wide range from 039 to 13, Fig. 2.1. The

frequency distribution o f the b values appeared to be Gaussian. The mean significantly

differed from 0.67 (p < 0.001), but not 0.75. When the b value o f each substance was

tested statistically against both 0.67 and 0.75, the majority o f the cases (81% and 98% at 14

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. the level o f significance equal to 0.05 and 0.01, respectively) failed to reject the null

hypotheses raised against both values (Table 2.1); i.e., individual b values did not differ

from 0.67 and 0.75. The wide range o f 95% and 99% C l o f b highlighted the uncertainty

associated with its determination o f b values in most studies.

The ten animal groups studied by Monte Carlo simulation had mean b values (n =

100 per simulation) close to the assigned true value, 0.75, Table 2 2 . However, the 95%

C l in the majority o f the scenarios failed to distinguish the expected value 0.75 from 0.67.

Only Scenario 4 at the level o f 20% CV secluded the possibility that b was 0.67 with

95% confidence. When the experimental error was set at 30% CV, none o f the

simulations distinguished between b values o f 0.67 and 0.75 w ith 95% confidence. The

mean r value ranged from 0.925 to 0.996, suggesting that the simulated experiments w ith

a 20% and a 30% CV in experimental bias were not particularly noisy. The frequency

distributions o f b values are shown in Fig. 2.2.

Fig. 2 3 shows the relationship between normalized clearances and body weights (n

=460) for the 91 xenobiotics that showed a statistically significant correlation in Table

2.1. The regression slope was 0.74 and the 99% C l was 0.71 - 0.76. The normalised

clearances were divided into four groups: 9 proteins (Group I, n =41), 21 compounds

eliminated mainly via renal secretion (Group 2, n = 105), 39 compounds eliminated

mainly via extensive metabolism (Group 3, n = 203), and 22 compounds eliminated by

both renal excretion and metabolism (Group 4, n = 111), Fig. 23. The summary o f the

regression results appears in Table 23. W hile Groups 1,3 and 4 had a b value close to

0.75 and significantly different from 0.67 (p < 0.001), Group 2 had a b value close to

0.67 and significantly different from 0.75 (p < 0.001). 15

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 2.4 Discussion

Successful prediction o f human clearance values using allometric scaling and

clearance values measured in animals depends heavily on the accuracy o f the b value.

Retrospective analysis o f published results for 115 substances indicated that the

commonly used experimental designs result in considerable uncertainty in this parameter,

Table 2.1.

CL values for twenty-four o f the substances listed in Table 2.1 failed to follow the

allometric equation at the 95% confidence level. The failures appeared to result from the

following: (i) only three species were studied in 16 cases, which severely lim ited the

robustness o f the statistics; in the remaining S failed cases one or more o f the following

occurred: (ii) the species were studied in different labs in 3 cases, (iii) small (n = 2) or

unequal (n = 2-10) numbers o f animals per species were studied in 4 cases, (iv ) different

dosages among species were used in 2 cases, and (v) high interspecies variability in

UDP-glucuronosyltransferase activity was proposed in one case.75 The failure o f these

24 cases to follow the allometric equation appeared for the most part, therefore, to result

from deficiencies in experimental design; i.e., failure o f detection rather than failure o f

the particular substance’s CL to follow the allometric relationship.

How well did allometry applied to animal CL values predict the human CL value?

One indication is how close the human CL value fe ll to the fitted line. O f the 91

substances that followed the allometric equation, 68 included human as one o f the

species. In 41 cases, the human CL value fe ll below the line and in 27 cases it fe ll above,

Fig. 2.4. The mean deviation was only 0.62% and the m ajority o f deviations were less

than 50%. It therefore appeared that fo r most o f the 68 substances studied in which 16

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. human was one o f the species, the human CL value did not deviate systematically or

extraordinarily from the fitted allometric equation. The tendency, noted by othersI0’12, o f

the CL value for human to be lower than that predicted from animal CL values was

therefore not apparent in this large data set

The b values fo r the 91 substances that followed the allometric equation appeared

to be normally distributed around a mean value o f 0.74 although the range o f values was

quite broad, Fig. 2.1. W hile impossible to answer definitively with these data, the

question o f whether there is a “universal” b value is o f interest Does the distribution

shown in Fig. 2.1 reflect a universal value w ith deviation about the mean due to

measurement errors, or are there different b values fo r the various clearance mechanisms

involved in clearance? The Monte Carlo simulations indicated that introduction o f

modest amounts o f random error in CL determinations. Fig. 2.2, resulted in a distribution

o f b values not unlike that shown in Fig. 2.1. This result supported the possibility that a

universal b value operates and that the range o f values seen in Table 2.1 resulted from

random error in CL determination coupled w ith the uncertainty that accrued from use o f a

lim ited number o f species. However, examination o f subsets o f the 91 substances

segregated by elimination pathway showed a b value around 0.75, except fo r substances

cleared prim arily by the kidneys; the b value fo r this subgroup was 0.65 (see below) and

the C l excluded a value larger than 0.70.

The central tendency o f the b values is o f interest, particularly given the recent

interest in the question o f whether basal metabolic rate scales with a b value o f 0.67 or

0.75.3’4’8’9 When examined individually, the 95% C l o f the b values for most o f the 91

substances included both values, although the mean for a ll the b values tended toward 17

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 0.75. So that a ll CL values could be viewed together, a normalization process was used

that assumed a common a value for a ll 91 substances and CL values were adjusted

accordingly, F ig . 2 3 . F it o f the allom etric equation to th is data set gave a b value o f 0.74

and its C l included 0.75 and excluded 0.67. Normalized CL values randomly scattered

about the line w ith one exception: in the body weight range 20 - 50 kg (dog, minipig,

sheep and goat) the normalized CL values generally fe ll above the line.

The 91 substances were segregated by molecular size (protein) and by major

elimination pathway (renal excretion, metabolism, combination o f both), Fig. 23. With

the exception o f the renal excretion subgroup, the normalized CL values for the

subgroups showed b values sim ilar to the combined group and their CIs included 0.75

and excluded 0.67, Table 23. The renal excretion subgroup (21 substances and 105 CL

values), however, showed a b value o f 0.65 w ith a C l that excluded 0.75. This result was

surprising as it appeared to contradict b values o f 0.77 reported fo r both mammalian

glomerular filtration rate and effective renal plasma flow,91"93 although it was consistent

w ith a b value o f 0.66 reported for intraspecies scaling o f inulin-based glomerular

filtration rate in humans,94 and with a b value o f 0.69 for scaling creatinine clearance.95

Whether the metabolic rate scales to the 2/3 or the 3/4 power o f body weight has

been the subject o f debate fo r many years. No consensus has been reached. The surface

law that suggested a proportional relationship between the metabolic rate and the body

surface area was first conceptualized in the 19th century. It has gained support from

empirical data6’ 96 as w ell as statistical6’ 9 and theoretical6’ 97 results. In 1932, Kleiber's

empirical analysis led to the 3/4-power law, which has recently been generalized as the

quarter-power law by West et al.3’4 Different theoretical analyses based on nutrient- 18

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. supply networks3,8 and four-dimensional biology4 a ll suggested that the quarter-power

law is the universal scaling law in biology.98 However, the claim o f universality was

challenged by Dodds et al.9 whose statistical and theoretical re-analyses cannot exclude

0.67 as the scaling exponent o f the basal metabolic rate.

The logic behind the pursuit o f a universal law for the scaling o f energy metabolism

across animal species is mainly based on the assumption that an optimal design o f

structure and function operates across animal species.3-4-8- 99,101 Given the fact that

mammals utilize the same energy source (oxygen) and energy transport systems

(cardiovascular, pulmonary) and the possibility that evolutionary force may result in a

design principle that optimizes energy metabolism systems across species, the existence

o f such a law might be possible. However, with available data and analyses a conclusion

has not been reached.

A large body o f literature data has indicated that the allometric scaling relationship

applies to the clearance o f a variety o f xenobiotics. It has been speculated that xenobiotic

clearance is related to metabolic rate and clearance b values have frequently been

compared w ith either 0.67 or 0.75. The b values obtained from the scaling o f clearance

for a variety o f xenobiotics tended to be scattered. M y analysis indicated that the b value

generally fe ll within the broad range between 0 and I or even higher. The scatter o f b

values may have resulted from the uncertainty that accrued from the regression analysis

o f a lim ited number o f data points as discussed above. In addition, the scatter may have

involved the variability in pharmacokinetic properties among different xenobiotics. The

latter rendered the prediction o f the b value extremely difficult. Moreover, the discussion

o f “universality” o f the b value was less possible in this regard. From the 19

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. pharmacokinetics point o f view, lack o f a unique b value for a il drugs may be considered

as a norm. In this regard, the uncertainty and variability became a universal

phenomenon. To determine whether a unique b value exists for the scaling o f CL, a more

rigorous experimental design has to be included to control the uncertainty that is possible

to obscure the conclusion, hi doing so, a study that includes the CL data fo r a variety o f

drags covering the animal species in the scope that is sim ilar to its counterpart in scaling

basal metabolic rate might be sufficient but extremely unrealistic. Therefore, from the

perspectives o f pharmacokinetics where drug is the center o f discussion, it is almost

impossible to address whether the b value o f CL tended to be dominated by one or two

values. However, from the perspectives o f physiology where the function o f a body is o f

interest, systematic analysis o f currently available data in interspecies scaling o f CL may

provide some insight into the interspecies scaling o f energy metabolism. The rationale

behind this line o f reasoning was that the elimination o f a xenobiotic from a body is a

manifestation o f physiological processes such as blood flow and oxygen consumption.

Interestingly, the two competitive exponent values, but not others, in theorizing the

scaling o f energy metabolism reappeared in the present analysis. The value 0.75

appeared to be the central tendency o f the b values for the CL o f a majority o f

compounds, except fo r that o f drugs whose elimination was mainly via kidney.

Whether allometric scaling could be used for the prediction o f the first-time-in-man

dose has been o f debate.102*103 Figure 2.4 showed that a reasonable error range could be

achieved when human CL was predicted by the animal data for some drags. However,

the success shown in the retrospective analysis does not necessarily warrant success in

prospective applications. As indicated by my analyses on the uncertainty o f b values and 20

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. as illustrated in Bonate and Howard’s commentary,102 caution is needed when allometric

scaling is applied in a prospective manner. Besides, the use o f a deterministic equation in

predicting individual CL data may be questionable because the inter-subject variability

cannot be accounted for. Nevertheless, allometric scaling could be an alternative tool, if

the population mean CL is to be estimated and the uncertainty is adequately addressed.

When the uncertainty in the determination o f a b-value is relatively large, a fixed-

exponent approach might be feasible, fin this regard, 0.75 might be used fo r substances

that are eliminated mainly by metabolism, and by metabolism and excretion combined,

whereas 0.67 might apply for drop that are eliminated mainly by renal excretion.

21

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 2.5 Summary

Statistical analysis and Monte Carlo simulation were used to characterize

uncertainty in the allometric exponent (b) o f xenobiotic clearance (CL). CL values for

115 xenobiotics wore from published studies in which at least three species were used for

the purpose o f interspecies comparison o f pharmacokinetics. The b value for each

xenobiotic was calculated along w ith its confidence interval (C l). For 24 xenobiotics

(21%) there was no correlation between log CL and log body weight. For the other 91

cases, the mean ±S D oftheb values was 0.74 ±0.16; range: 0.29- 1.2. M ost(8I% )of

these individual b values did not differ from either 0.67 or 0.75 at p = 0.05. When CL

values fo r the subset o f 91 substances were normalized to a common body weight

coefficient (a), the b value for the 460 adjusted CL values was 0.74; the 99% C l was 0.71

- 0.76, which excluded 0.67. Monte Carlo simulation indicated that the wide range o f

observed b values could have resulted from random variability in CL values determined

in a lim ited number o f species, even though the underlying b value was 0.75. From the

normalized CL values, four xenobiotic subgroups were examined: those that were (i)

protein, and those that were (if) eliminated mainly by renal excretion, (iii) metabolism or

(iv) by renal excretion and metabolism combined. A il subgroups except (ii) showed a b

value not different from 0.75. The b value for the renal excretion subgroup (21

xenobiotics, 105 CL values) was 0.65, which differed from 0.75 but not from 0.67.

22

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 2.6 References

1. Schmidt-Nielsen EC. Scaling: Why Is Animal Size So Important? Princeton, NJ: Cambridge U niversity Press, 1983.

2. Calder WA III. Size, Function and Life History. Cambridge, MA: Harvard U niversity Press, 1984.

3. West GB, Brown JH, Enquist BJ. A general model for the origin o f allometric scaling laws in biology. Science. 1997;276:122-126.

4. West GB, Brown JH, Enquist BJ. The fourth dimension o f life: Fractal geometry and allometric scaling o f organisms. Science. 1999;284:1677-1679.

5. ECIeiber M . Body size and metabolism. H ilgardia. I932;6:315-353.

6. Heusner AA. Energy metabolism and body size. I. Is the 0.75 mass exponent o f Kleiber’s equation a statistical artifact? Respir Physiol. 1982;48:1-12.

7. Feldman HA, McMahon TA. The 3/4 mass exponent for energy metabolism is not a statistical artifact. Respir Physiol. 1983;52:149-163.

8. Banavar JR, Maritan A, Rinaldo A. Size and form in efficient transportation networks. Nature. 1999;399:130-132.

9. Dodds PS, Rothman DH, Weitz JS. Re-examination o f the "3/4-law" o f metabolism. J Theor Biol. 2001;209:9-27.

10. Boxenbaum H. Interspecies scaling, allometry, physiological time, and the ground plan o f pharmacokinetics. J Pharmacokin Biopharm. 1982;10:201-227.

11. Sawada Y , Hanano M, Sugiyama Y, Iga T. Prediction o f disposition o f beta-Iactam antibiotics in humans from pharmacokinetic parameters in animals. J Pharmacokin Biopharm. 1984;12:241-261.

12. M ordent! J . Man versus beast: Pharmacokinetic scaling in mammals. J Pharm ScL 1986;75:1028-1040. 23

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 13. Mahmood I, Balian JD. Interspecies scaling: Prediction clearance o f drugs in humans. Three different approaches. Xenobiotica. 1996;26:887-895.

14. Feng MR, Lou X, Brown RR, Hutchaleelaha A. Allometric pharmacokinetic scaling: Towards the prediction o f human oral pharmacokinetics. Pharm Res. 2000;17:410-418.

15. Mahmood L Interspecies scaling o f renaily secreted drugs. Life Sci. I998;63:2365- 2371.

16. McGovren JP, Williams MG, Stewart JC. Interspecies comparison o f acivicin pharmacokinetics. Drug Metab Dispo. 1988;16:18-22.

17. Brazzell RK, Park YH, Wooldridge CB, et al. Interspecies comparison o f the pharmacokinetics o f aldose reductase inhibitors. Drug Metab Dispos. 1990; 18:435- 440.

18. Bjorkman S, Redke F. Clearance o f fentanyl, alfentanil, methohexitone, thiopentone and ketamine in relation to estimated hepatic blood flow in several animal species: Application to prediction o f clearance in man. J Pharm Pharmacol. 2000;52:1065-1074.

19. Cherkofsky SC. I-Aminocyciopropanecarfaoxylic acid: Mouse to m an interspecies pharmacokinetic comparisons and allometric relationships. J Pharm Sci. 1995;84:1231-1235.

20. Robbie G, Chiou WL. Elucidation o f human amphotericin b pharmacokinetics: Identification o f a new potential factor affecting interspecies pharmacokinetic scaling. Pharm Res. 1998;15:1630-1636.

21. Paxton JW, Kim SN, W hitfield LR. Pharmacokinetic and toxicity scaling o f the antitumor agents amsacrine and C I-92I, a new analogue, in mice, rats, rabbits, dogs, and humans. Cancer Res. 1990;50:2692-2697.

22. GreneLerouge NAM, Bazin-Redureau ML Debray M , Schermann JM. Interspecies scaling o f clearance and volume o f distribution fo r digoxin-specific Fab. Toxicol AppI Pharmacol. 1996;138:84-89.

24

i i Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 23. Lave T, Dupm S, Schmidt C, Chou RC, Jaeck D, Coassolo PH. Integration o f in vitro data into allometric scaling to predict hepatic metabolic clearance in man: Application to 10 extensively metabolized drags. J Pharm Sci. 1997;86:584-590.

24. Bazin-Redureau M, Pepin S, Hong G, Debray M, Scherrmann JM. Interspecies scaling o f clearance and volume o f distribution for horse antivenom FCab^. Toxicol Appl PharmacoL 1998; 150:295-300.

25. Lashev LD, Pashov DA, Marinkov TN. Interspecies differences in the pharmacokinetics o f kanamycin and apramycin. Vet Res Comm. 1992;16:293-300.

26. Patel BA, Boudinot FD, Schinazi RF, Gallo JM, Chu CK. Comparative pharmacokinetics and interspecies scaling o f 3’-azido-3’-deoxy-thymidine (AZT) in several mammalian species. J Pharmacobio-Dyn. 1990;13:206-211.

27. Kurihara A, Naganuma H, Hisaoka M, Tokiwa H, Kawahara Y. Prediction o f human pharmacokinetics o f panipenem-betamipron, a new carbapenem, from animal data. Antimicrob Ag Chemother. 1992;36:1810-1816.

28. Mehta SC, Lu DR. Interspecies pharmacokinetic scaling o f BSH in mice, rats, rabbits, and humans. Biopharm Drug Dispos. 1995;16:735-744.

29. Bonati M, Latini R, Tognoni G. Interspecies comparison o f in vivo caffeine pharmacokinetics in man, monkey, rabbit, rat, and mouse. Drug Metab Rev. 1984- 85; 15:1355-1383.

30. Kaye B, Brearley CJ, Cussans NJ, Herron M, Humphrey MJ, M ollatt AR. Formation and pharmacokinetics o f the active drug candoxatrilat in mouse, rat, rabbit, dog and man following administrationo f the produg candoxatril. Xenobiotica. 1997;27:1091-1102.

31. Mordend J, Chen SA, Moore JA, Ferraiolo BL, Green JD. Interspecies scaling o f clearance and volume o f distribudon data fo r five therapeudc proteins. Pharm Res. 1991;8:1351-1359.

32. Sawada Y, Hanano M, Sugiyama Y, Iga T. Predicdon o f the disposition ofb-Iactam antibiotics in humans from pharmacokinetic parameters in animals. J Pharmacokinet Biopharm. 1984;12:241-261.

25

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 33. Matsushita H, Suzuki H, Sugiyama Y, et al. Predictioa o f the phanuacokiuetics o f cefodizime and cefotetan in humans fiom pharmacokinetic parameters in animals. J Pharmacobio-Dyn. 1990;13:602-611.

34. Mordenti J. Pharmacokinetic scale-up: Accurate prediction o f human pharmacokinetic profiles fiom animal data. J Pharm Sci. I985;74:1097-1099.

35. Feng MR, Loo J, Wright J. Disposition o f the antipsychotic agent CI-I007 in rats, monkeys, dogs, and human cytochrome p450 2D6 extensive metaholizers: Species comparison and allometric scaling. Drug Metab Dispos. 1998;26:982-988.

36. Hildebrand M. Inter-species extrapolation o f pharmacokinetic data o f three prostacyclin-mimetics. Prostaglandins. 1994;48:297-312.

37. Ericsson H, Tholander B, Bjorkman JA, Nordlander M, Regardh CG. Pharmacokinetics o f new calcium channel antagonist clevidipine in the rat, rabbit, and dog and pharmacokinetic/pharmacodynamic relationship in anesthetized dogs. Drug Metab Dispo. 1999;27:558-564.

38. Sangalli L, Bortolotti A, Jiritano L, Bonati M. Cyclosporine pharmacokinetics in rats and interspecies comparison in dogs, rabbits, rats, and humans. Drug Metab Dispo. 1998;16:749-753.

39. Kim SH, Kim WB, Lee MG. Interspecies pharmacokinetic scaling o f a new carbapenem, D A -l 131, in mice, rats, rabbits and dogs, and prediction o f human pharmacokinetics. Biopharm Drug Dispos. 1998;19:231-235.

40. Klotz U, Antonin K-H, Bieck PR. Pharmacokinetics and plasma binding o f diazepam in man, dog, rabbit, guinea pig and ra t J Pharmacol Exp Ther. 1976;199:67-73.

41. Kaul S, Daudekar KA, Schilling BE, Barbhaiya RH. Toxicokinetics o f 2\3’- deoxythymidine, stavudine (D4T). Drug Metab Dispos. 1999;27:1-12.

42. Sanwald-Ducray P, Dow J. Prediction o f the pharmacokinetic parameters o f reduced-dolasetron in man using in vitro-in vivo and interspecies allometric scaling. Xenobiotica. 1997;27:189-201.

26

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 43. Kawakami J, Yamamoto K, Sawada Y, Iga T. Prediction o f brain delivery o f ofloxacin, a new quinolone, in the human from animal data. J Pharmacokinet Biopharm. 1994;22:207-227.

44. Tsunekawa Y, Hasegawa T, Nadai M , Takagi K, Nafaeshima T. Interspecies differences and scaling fo r the pharmacokinetics o f xanthine derivatives. J Pharm Pharmacol. 1992;44:594-599.

45. Bregante MA, Saez P, Aramayona JJ, et al. Comparative pharmacokinetics o f enrofloxacin in mice, rats, rabbits, sheep, and cows. Am J Vet Res. 1999;60:1111- I I 16.

46. Duthu GS. Interspecies correlation o f the pharmacokinetics o f erythromycin, oleandomycin, and tylosin. J Pharm Sci. 1985;74:943-946.

47. Efthymiopoulos C, Battaglia R, Strolin Benedetti M. Animal pharmacokinetics and interspecies scaling o f FCE 22101, a penem antibiotic. J Antimicrob Chemother. 1991;27:517-526.

48. Jezequel SG. Fluconazole: Interspecies scaling and allometric relationships o f pharmacokinetic properties. J Pharm Pharmacol. 1994;46:196-199.

49. Segre G, Bianchi E, Zanolo G. Pharmacokinetics o f flunoxaprofen in rats, dogs, and monkeys. J Pharm Sci. 1988;77:670-673.

50. Khor SP, Amyx H, Davis ST, Nelson D, Baccanari DP, Spector T. Dihydropyrimidine dehydrogenase inactivation and 5-fluorouraciI pharmacokinetics: Allom etric scaling o f animal data, pharmacokinetics and toxicodynamics o f 5-fIuorouracil in humans. Cancer Chemother Pharmacol. 1997;39:233-238.

51. Clark B, Smith DA. Metabolism and excretion o f a chromone carfaoxylic acid (FPL 52757) in various animal species. Xenobiotica. 1982;12:147-153.

52. Nakajima Y, Hattori K, Shmsei M, et al. Physiologically-based pharmacokinetic analysis o f grepafloxacin. B iol Pharm B ull. 2000;23:1077-1083.

27

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 53. Baggot JD. Application o f interspecies scaling to the bispyridinhim oxime HI-6. Am J V e t Res. 1994;55:689-691.

54. Lave T, Levet-Trafit B, Schmitt-Hoffinann AH, et aL Interspecies scaling o f interferon disposition and comparison o f allometric scaling w ith concentration-time transformations. J Pharm Sci. 1995;84:1285-1290.

55. Sakai T, Hamada T, Awata N, Watanabe J. Pharmacokinetics o f an antiallergic agent, l-(2-ethoxyethyI)-2-(hexahydro-4-methyl-l H-1,4-diazepin-I-yl)-l H- benzimidazole difumarate (KG-2413) after oral administration: Interspecies differences in rats, guinea pigs and dogs. J Pharmacohio-Dyn. 1989;12:530-536.

56. Lave T, Saner A, Coassoto P, Brandt R, Schmitt-Hoffinan AH, Chou RC. A n im a l pharmacokinetics and interspecies scaling fiom animals to man o f lamifiban, a new platelet aggregation inhibitor. J Pharm PharmacoL 1996;48:573-577.

57. Richter WF, Gallati H, Schiller CD. Animal pharmacokinetics o f the tumor necrosis factor receptor-immunoglobulin fusion protein lenercept and their extrapolation to humans. Drug Metab Dispos. 1999;27:21-25.

58. Lapka R, Rejholec V, Sechser T, Peterkova M, Smid M. Interspecies pharmacokinetic scaling o f metazosin, a novel alpha-adrenergic antagonist. Biopharm Drug Dispo. 1989;10:581-589.

59. Ahr HJ, Boberg M, Brendet E, Krause HP, Steinke W. Pharmacokinetics o f m iglitoi: Absorption, distribution, metabolism, and excretion following administration to rats, dogs, and man. Arzneim Forsch. 1997;47:734-745.

60. Siefert HM, Domdey-Bette A , Henninger K, Hucke F, Kohlsdorfer C, Stass HH. Pharmacokinetics o f the 8-methoxyquinoIone, moxifloxacin: A comparison in humans and other mammalian species. J Anthnicrob Chemother. 1999;43 (SuppL B):69-76.

61. Lave T, Portmann R, Schenker G, etaL Interspecies pharmacokinetic comparisons and allometric scaling o f napsagatran, a low molecular weight thrombin inhibitor. J Pharm PharmacoL 1999;51:85-91.

28

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 62. Higuchi S, Shiobara Y. Comparative pharmacokinetics o f nicardipine hydrochloride, a new vasodilator, in various species. Xenobiotica. 1980;10:447- 454.

63. Mitsuhashi Y, Sugiyama Y, Ozawa S, et aL Prediction o f ACNU plasma concentration-time profiles in humans by animal scale-up. Cancer Chemother Pharmacol. 1990;27:20-26.

64. Yoshimura M, Kojima J, Ito T, Suzuki J. Pharmacokinetics o f (K-351), a new antihypertensive agent L Studies on interspecies variation in laboratory animals. J Pharmacobio-Dyn. 1985;8:738-750.

65. Gombar CT, Harrington GW, Pylypiw HM Jr, et al. Interspecies scaling o f the pharmacokinetics ofN-nitrosodimethylamine. Cancer Res. 1990;50:4366-4370.

66. Mukai H, Watanabe S, Tsuchida K, Morino A. Pharmacokinetics o f NS-49, a phenethyfamine class a I A-adrenoceptor agonist, at therapeutic doses in several animal species and interspecies scaling o f its pharmacokinetic parameters. Int J Pharm. 1999;186:215-222.

67. Owens SM, Hardwick WC, Blackall D. Phencyclidine pharmacokinetic scaling among species. J Pharmacol Exp Ther. 1987;242:96-101.

68. Ishigami M, Saburomaru K, Niino K, et al. Pharmacokinetics o f procateroi in the rat, rabbit, and beagle dog. Arzneim Forsch. 1979;29:266-270.

69. Khor SP, McCarthy K, DuPont M, Murray K, Timony G. Pharmacokinetics, pharmacodynamics, allometry, and dose selection o f rPSGL-lg for phase I trial. J Pharmacol Exp Ther. 2000;293:618-624.

70. Mordenti J, Osaka G, Garcia K, Thomsen K, Licko V, Meng G. Pharmacokinetics and interspecies scaling o f recombinant human factor VOL Toxicol Appl PharmacoL 1996;136:75-78.

71. Coassolo P, Fischli W, Clozel J-P, Chou RC. Pharmacokinetics o f remikiren, a potent orally active inhibitor ofhuman renin, in rat, dog, and primates. Xenobiotica. 1996;26:333-345.

29

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 72. Widman M, Nilsson LB, Bryske B, Lundstrom J. Disposition o f remoxipride in different species. Arzneim Forsch. 1993;43:287-297.

73. Lashev L, Pashov D, Kanelov L Species specific pharmacokinetics o f rolitetracyclme. J Vet Med A. 1995;42:201-208.

74. Herault JP, Donat F, Barzu T, et al. Pharmacokinetic study o f three synthetic AT- binding pentasaccharides in various animal species-extrapolation to humans. Blood Coagul FibrinoL 1997;8:161-167.

75. Ward ECW, Azzarano LM, Bondineii WE, et al. Preclinical pharmacokinetics and interspecies scaling o f a novel vitronectin receptor antagonist. Drug Metab Dispos. 1999;27:1232-1241.

76. Lin C, Gupta S, Loebenberg D, Cayen MN. Pharmacokinetics o f an eveminomicin (SCH 27899) in mice, rats, rabbits, and cynomolgus monkeys following intravenous administration. Antimicrob Ag Chemother. 2000;44:916-919.

77. Chung M , Radwanski E, Loebenberg D, et al. Interspecies pharmacokinetic scaling o f Sch 34343. J Antimicrob Chemother. 1985;I5 (Suppl. C):227-233.

78. H inderling PH, D ilea C , ECoziol T , M illin g to n G. Comparative kinetics o f sematOide in four species. Drug Metab Dispo. 1993;21:662-669.

79. Walker DEC, Ackland MJ, James GC, et al. Pharmacokinetics and metabolism o f sildenafil in mouse, rat, rabbit, dog, and man. Xenobiotica. 1999;29:297-310.

80. Brocks DR, Freed ML Martin DE, et al. Interspecies pharmacokinetics o f a novel hematoregulatory peptide (SK&F 107647) in rats, dogs, and oncologic patients. Pharm Res. 1996;13:794-797.

81. Cosson VF, Fuseau E, Efthymiopoulos C, Bye A. Mixed effect modelingof sumatriptan pharmacokinetics during chug development I: Interspecies allometric scaling. J Pharmacokin Biopharm. 1997;25:149-167.

82. Leusch A , Troger W, GreischeL A, Roth W. Pharmacokinetics o f the MI-agonist talsaclidine in mouse, ra t rabbit and monkey, and extrapolation to man. Xenobiotica. 2000;30:797-813. 30

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 83. van Hoogdalem EJ, Soeishi Y , Matsushima H, Higuchi S. Disposition o f the selective a l A-adrenoceptor antagonist in humans: Comparison w ith data fiom interspecies scaling. J Pharm Sci. I997;86:l 156-1161.

84. Craze CA, Kelm GR, Meredith MP. Interspecies scaling o f tebufelone pharmacokinetic data and application to preclinical toxicology. Pharm Res. 1995;12:895-901.

85. Gaspari F, Bonati M. Interspecies metabolism and pharmacokinetic scaling o f theophylline disposition. Drug Metab Rev. 1990;22:179-207.

86. Davi H, Tronquet C, Calx J, et al. Disposition o f dludronate (Skelid) in animals. Xenobiotica. 1999;29:1017-1031.

87. Pahlman I, Kankaanranta S, Palmer L. Pharmacokinetics o f tolterodine, a muscarinic receptor antagonist, in mouse, rat and dog. Arzneim Forsch. 2001;51:134-144.

88. Tanaka E, Ishikawa A, Horie T. hi vivo and in vitro trimethadione oxidation activity o f the liver from various animal species including mouse, ham ster, rat, rabbit, dog, monkey and human. Human Exp Toxicol. 1999;18:12-16.

89. Izumi T, Enomoto S, Hosiyama K, et al. Prediction o f the human pharmacokinetics o f troglitazone, a new and extensively metabolized antidiabetic agent, after oral administration, with an animal scale-up approach. J Pharmacol Exp Ther. 1996;277:1630-1641.

90. Grmdel JM, O'Neil PG, Yorgey KA, et aL The metabolism o f zomephac sodium L Disposition in laboratory animals and man. Drug Metab Dispo. 1980;8:343-348.

91. Singer MA, Morton AR. Mouse to elephant: Biological scaling and Kt/V. Am J Kidney Dis. 2000;35:306-309.

92. Singer MA. O f mice and men and elephants: Metabolic rate sets glomerular filtration rate. Am J Kidney Dis. 2001;37:164-178.

93. Edwards NA. Scaling o f renal functions in mammals. Comp Biochem Physiol. 1975;52A:63-66- 31

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 94. Hayton WL. Maturation and growth o f renal function: Dosing renaiiy cleared drugs in children. AAPS PbarmScL 2000;2(l), article 3. Available from: http://www.pharmsci.Org/scientifIcjoumaIs/pharnisci/joumaI/3.htnil

95. Adolph EF. Quantitative relations in the physiological constituents o f mammals. Science. 1949;109:579-585.

96. Rubner M. Ober den enifluss der korpergrdsse auf stoff und kraftwechsel. Z Biol. 1883;19:535-562.

97. Heusner A. Energy metabolism and body size. II. Dimensional analysis and energetic non-similarity. Resp Physiol. 1982;48:13-25.

98. West GB. The origin o f universal scaling laws in biology. Physica A. 1999;263:104-113.

99. Murray CD. The physiological principle o f minimum work. L The vascular system and the cost o f blood volume. Proc Natl Acad Sd U S A. 1926;12:207-214.

100. Cohn DL. Optimal systems: L The vascular system. Bull Math Biophys. 1954;16:59-74.

101. Cohn DL. Optimal systems: IL The vascular system. Bull Math Biophys. 1955;17:219-227.

102. Bonate PL* Howard D. Prospective ailometic scaling: Does the emperor have clothes? J C lin Pharmacol. 2000;40:665-670.

103. Mahmood L Critique o f prospective allometric scaling: Does the emperor have clothes? J C lin Pharmacol. 2000;40:671-674.

32

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

16 17 17 17 18 19 20 25 21 22 23 24 26 27 23 28 29 30 31 32 32 32 32 33 Rer. BW data (con inued) versus ip , dg, mk, cz mk, dg, , 1 ms. it, mk, dg, hm dg, mk, it, hm ms. cz, dg, mk, it. n, mk, cz, hm cz, mk, n, ( sh dg, rt>, it, ms, it, mk, hm mk, it, hm ms, dg, it>, it, hm ms, dg, it>, it, ms, ib it, ms, hm dg, ib, it, it, ms, sh, ib, ck, pn ck, ib, hm sh, dg, mk, it, dg ms, mk, ib, n, gp, ms, ms, ml, it, ib, hm ib, it, ml, ms, ms, it, ib, hm ib, it, ms, hm mk, ib, it, ms, ms, it, ib, dg, hm dg, ib, it, ms, hm hm mk, ib, mk, it, dg, ib, n, hm mk, ms, dg, ib, it, ms, ms, it, ib, dg, mk, hm mk, dg, ib, it, hm mk, ms, dg, ib, it, ms, ms, it, ib, dg, mk dg, ib, it, ms, m n.d. n.d. n.d. n.d. n.d. 0.54-1.6 0.52-1.4 0.39-1.1 0.37 - 0.78 0.028-1.6 0,047- 1.9 0.016-1.0 0.74 -0.94 m m im m n.d. n.d. n.d. n.d. n.d.'*' n.d. 0.17-1.7 0.16-1.8 0.28-1.2 0.38-1.2 0.27-1.20.48-1.5 n.d. 0.18-1.2 n.d. 0.19-0.73 n.d. 0.53 • 0.840.006-1.1 0.43 - 0.94 0.52-0.81 0.39 - 0.93 0.55 - 0.93 0.52 - 0.830.35 - 0.84 0.43 - 0.93 0.18-1.0 0.20 - 0.94 0.22 0.22 - 0.84 0.72 -1.2 95% Cl orb 99% Cl ofb ow Species 0.77- 0.91 0.45- 0.70 • ♦ * t • • • e ee ee ee ee ee ee M e eee »M ♦ ♦♦ ♦ p««» 0.06 0.15 *** 0.06 r*m 0.976 0.9550.829 0.902 0 .7 5 -1 .4 0.716 0.982 0.924 0.975 0.663 0.945 0.959 0.975 0.917 0.926 0.823 0.849 b l.l 1.0 0.57 0.98 0.96 0.80 0.57 0.56 0.68 0.74 0.59 a 16 0.69 26 47 0.75 0.975 0.59 - 0.92 0.48-1.0 25 1.0 0.67 0.992 3.9 1.5 6.9 2.1 9.6 0.66 0.986 6.7 0.57 6.3 0.53 4.5 0.68 0.41 0.93 0.834 0.39 0.94 0.84 0.988 0 .1 0 0.033 0.53 0.990 G b 1 -Aminocyelopropanecarboxylnte 1 2.6 0.72 Acivin ALQI567 Anti-dinoxin Fab Anti-dinoxin Antipyrine AL0I576ALQI750 Alfentanil B Amphotericin 0.36 AZT Amsacrinc Fab2 Antivenom ApramycinBetamipron Bosentan 38 0.46 2.8 0.906 BSH CaffeineCandoxatrilat CD4*I Cefazolin Cefmetazole 6.3 0.74 12 0.981 Cefodizime Cefoperazone Cofotetan Tabic 2.1 Allometric scaling parameters obtained from linear regressions of the log-log-transformed CL log-log-transformed ofthe Tabic from 2.1 obtained linear parameters regressions Allometric scaling of 115 xenobiotics (a; b; exponent) coefficient; allometric allometric xenobiotics of 115 w w

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. without prohibited reproduction Further owner. copyright the of permission with Reproduced

Tabic 2.1. (continued) "L "a i o *3 9 m a o « 1 1 95% Cl of b 1 99% Cl of b a 1 R e f. 1 © o € 73 t ■S - r £ « © oo "d « o o - O d •*r i m s o •d cn d £ efi ff- s £ o d d o 4 ■o n U <3 © - r © s o OO SO a a © o © cn t^- © - p oo cn — TT d i ea o s X £ o » » f ■S Os © u o o cn OS s o 00 a cn - r © n* © o *d nr mi mi n* d d » » e • •3 ■s a O © •A N •A © oo cn o © © oo ■d © Os cn o m N d £ efi £ d 1 • ♦ a ■e i JS 5 © © IA SO a © 00 cn « •A © cn © © cn cn SO - r n* d t £ o a « § I i o o •e 73 c— a [ 00 U cn oo 00 © © OO •A © ■d cn ■d - e t£ ea o > e o d d •e •o JS 'C O © © cn ♦ © © © oq •d oo •A °o - p ©' cn d so £ > (A a o C d l e • *o | • • * © d 00 a cn o © IA © © o Co p © < d cn — - e cn d £ m efi E • 1 l •e •3 JS o w d cn d cn •d *d a oo © mi d £ efi £ i a as £ d d 1 40 1 4 73 JS *o IS : j o © * d sO Q cn cn - P a - r d mi cn o d cn cn cn n* d efi £ es s o o i i t ■3 e . o © IA o d cn a TT * - e d cn cn cn ■d - n cn - r d 3 efi £ o cn s es s d i j* •3 JS » : j • DU sO d oo c— * i » [ 46 1 • ■e ■X ■SP DC. u DU cn cn o d © o © d o cn d - r sO •d — - r «n n* c— d £ c l e s ■# d © © o *d d 00 00 SO © Es. OO •d d £ 5 . c ea o e c e € a a • d © © d © cn EE. cn d SO cn d - r - c - c o oo d 00 cn Ml d £ £ £ efi o s 3 O o 3 i i 48 1 73 t o r* <2 •d d © oo Cu d © cn m p -d d efi e O as 3 X a 2 c s d 49 I a 4 JS a o © p •Cl Du © d Ml cn o © d c— d e^ cn ^r cn d £ m £ o I 3 B k 1 i 50 I 4 i 4 a a o © cn d cn d © DU CU U Ml cn r*- Ml - r r— d cn d 00 sO d cn oo d © c— Ml d £ k i i ■e 1 o s o © SO d 3 Ml d oo 00 ■d *d Ml cn d o 2 o cs o X C3 d d ■e •o JS a a a so o © 00 M d © d - P sO cn d © © - r o SCI cn o Ml cn d © d £ uf £ efi uk £ m » i i T3 O O S 3 18 18 56 36 54 25 55 57 58 59 41 46 23 23 23 32 60 61 62 65 63 64 43 66 Refl (continued) m ms, rt. i v.d^. ps, rt.ms, ihm it, dg, mk sh, gt, it>, ck, pn gt, sh, it>, it. it*. PS ms, it, rt>, dg, mk ms, it, rt>, it, ds SP. it, mk, dg, hm it, mk, dg it>, ms, ms, it, mk, dg, hm it,ms, it), hm it, it>, dg it, it>, it, ml, it), dg, hm it, it), ps,dg, hm it, dg, hm ms, it,dg, hm ms, n, it>, dg, ms, mk,n, hm it>, ms, it,ms, mk, dg it, dg, mk it>, it.dg, mk, hm it,ms, lb, dg, hm it, it), mk, dg ms, hr, it, it), mk, pg dg, ms, it, mk, dg, hm dg it, it>, m n.d. n.d. n.d. 0.56-1.0 0.24-1.3 0.65-1.2 0.53 0.53 - 0.79 0.26 - 0.86 0.30 - 0,80 0 .7 1 - 1.4 m m m m m n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. 0.26-1.5 n.d. 0,64-1.1 0.51-1.2 0,15-1.2 n.d. 0,61 0,61 - 1.0 0 .48 - 1.1 0.51-1.2 n.d. 0.55-1.1 0.32-1.3 0.28-1.3 n.d 0.66 - 0.92 0.15-0.44 0.29-0.95 0.018-1.2 0.52 - 0.90 0.36-1.1 0.53-0.97 0.31-0.97 n.d. 0.047-1.3 n.d, 0.90 -1 .2 95% C l orb 99% C l orb Species 0 .7 5 - l.l 0.58- 0.74 0.38- 0.74 0 .4 0 -0.70 • • * * ♦ • •• •• •• ♦* *• *• 0.2 0.1 0 3 ••• ♦** ♦♦♦ ♦♦♦ pWI 0.08 0.05 r*(4 0.980 0.632 0.991 0.992 0.973 0.923 0.997 0.992 0.949 0.842 0.968 0.796 0.962 0.893 1.1 0.741 1.1 0.998 0,85 0,970 0.810.56 0.970 0,75 0.79 0.29 0.62 0.86 0.56 0.74 0.55 0.66 0.93 0.972 0,64 0,994 a b 10 14 0.84 0.983 48 73 62 67 0.68 0.850 50 59 59 69 81 0,77 3.7 0,71 6.1 0.88 0.887 2.9 610 0,0079 a Interferon Interferon Kanamycin Kctaminc 119 Iloprost Lamifiban Lamivudine 15 Lcncrcent KG-2413 Lomefloxacin Mctazocin 11 Methohcxitone Mibcfradil Midazolam Mi^litolMofarotene 7,4 0.64 0.998 Moxalaclam 5.0 0.66 Moxifloxacin 20 Norfloxacin Napsasalran Nicardipine NimustineNinradilol N-Nitrosodimethylamine 42 0.83 NS-49 14 Table 2,1. Table (continued) 2,1.

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 23 27 68 69 43 67 31 31 70 43 46 72 23 73 74 31 71 75 76 77 78 79 80 74 Ref. (continued) m Species Species ms, gp, it, rti, mk, dg mk, rti, it, gp, hm ms, dg, mk, rt, ms, rt, mk, dg, hm dg, mk, rt, hm dg, n, mt, it, rt>, dg rt>, it, hm dg, iti, ft, pg mk, rt, ms, ms, n, pn. mk, dg, hm dg, mk, pn. n, ms, hm mk, rt>, n, ms, rt, mk, hm mk, rt, ms, hm hm n, mk, ms, rt>, rt, ms, mk, ml, dg, rt, ms, rt, hs, dg, hm dg, hs, rt, hm ms, dg, rt>, ck rt, pit, pg, rt>, it, rfa, bb rfa, it, dg mk, rt, ms, mk rb, hm rt, dg, ms, rb, mk, it, ms, hm dg, rb, rt, hut dg, rt, ms, rt, dg, hm dg, rt, rt, rb, bb rb, rt, n.d, n.d, n.d. n.d. n.d. n.d. n.d. n.d, n.d. 0.58-1.2 0.37-1.2 0.51 0.51 -0.81 0,31-0.98 m n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. 0.17-1,1 0,51 0,51 -1,0 0.49-1.4 0.13-1.7 0.26-1.1 0.33-0.95 n.d. 0,55 0,55 - 0,83 0 .36 -1 .0 0,27 - 0.98 0,33-0,95 0,12-1.1 0.24-0,90 0.50-0.79 0.39-0.94 0.034-1.3 0.45 0.45 - 0.97 0.72 -1.1 0.59 - 0,73 ♦ • • • ♦ • ♦t ♦ ♦ ♦ * ♦ •• 0.1 ♦♦♦ 0.1 •t* n"" 95% Cl ofb 99% Cl orb 0.10 0.06 0.07 0.09 0.06 r>(f> 0.946 0.939 0.995 0.992 0.891 0.910 0.999 0.982 0.990 0.964 0.80 0.57 0.71 0.42 0.710 0.89 0.989 0.71 0,9950.67 0.898 0.55-0.87 0.34-1.1 0.64 0.976 0.53 0.62 0.966 0.63 a b 13 11 30 0.69 0.996 98 0.64 0.81 29 50 20 0.66 29 69 28 0.66 0.999 3.4 0.65 7.5 0.64 6.8 7.2 0.16 0.350.78 0.87 0.979 0.10 Phencyclidine 52 0.64 lisand-1 filyconrotein P-selcctin 0.0060 0.93 Procalerol PanipenemPefloxacin 12 0.61 0.977 0 .4 8 -0.74 0.39 - 0.82 Recombinant CD4 Recombinant Recombinant growth hormone growth Recombinant V human III factor Recombinant RelaxinRemikiren Remoxipride 6.0 0.80 0.992 0.66 - 0.93 0 .55-1.0 Ofloxacin Oleandomycin Ro 24-6173 Ro Rolitetracycline Sanorg 32701 Sanorg SB-263123Sch27899 34343Sch Sematilide Sildenafil 15 13 0.80 0.812 0.77 0.924 SR 80027 SR SK&F107647 Table (continued) 2,1.

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

18 74 81 82 83 84 41 87 31 86 85 27 88 89 43 48 41 41 90 , hm , ps, hs, 01 it, rb, bb, hm ms, ms, rt, mk, rb, hm rt, dg, rb, hm ms, ms, n, mk, hm rt, dg rb, rt, mk, ds, hm ms, hs, it, hs, ms, rb, mk, dg, hm rt, rb, dg, sh it,ms, rb, bb dg, rt, sp, rb, rt, rb, dg, hm ms, n,dg, hm ms, rt, mk, dg, hm it,ms, hs, ib, dg, mk, hm ms, rt, mk, dg rt, dg, cw ms, it, ct, mk, hm ms, rt,ms, mk, dg, hm ms, ms, rt, ink,rb, hs, hm n.d. n.d. 0.60-1.1 0.66-1.0 0.51 0.51 -1.4 0.28 - 2.0 0.27-0.84 m m m n.d. n.d. n.d. 0.42-1.3 n.d. 0,32-1.3 0.57-1.4 0.32-1.7 0.54-1,1 0,19-1.4 0.62-1.0 0.45-1.2 0,63-1,7 0.30 - 0.79 n.d. 0.30 - 0,97 n.d. 0.36-1.24 n.d. 0.095-1.20.34-0.90 n.d. n.d. 0.40-0.71 0.64 - 0.98 0.57-1.1 0.50 - 0.90 0.39-1.0 0.71 0.71 -1.2 0,71 0,71 *0,97 0.72- 0.95 • « ♦ • * • • •« •» •• ♦♦ •** ♦♦♦ 005 0.053 0.978 0.993 0,971 0,993 0,950 0.919 0.927 0.978 0.942 0.874 0,963 0.993 0.983 0.902 1.0 1.2 0,55 0.84 0.63 0,59 0.65 0.84 0.973 0,79 0.81 0.80 0.69 0.81 0.988 0.70 19 12 17 0.84 0.986 32 61 31 1.9 54 62 0.62 26 0.95 0.981 1.6 0.68 = 0,67 b b « < 0,05 p 0: (♦); < p 0,01 (♦•); p<0,001 (♦*♦) b b = 0,75 and = 0,75 and b b » 0,75 ■ 0,67 b b b pig; ct, cat; cw, cow; gt, goal; ntt, marmoset; hs, hamster hs, marmoset; ntt, goal; gt, cow; cw, cat; ct, pig; rt, rat; rb, rabbit; bb, baboon; mk, monkey; dg, dog; hm, human; ms, mouse; cz, chimpanzee; sh, sheep; ck, chicken; pn, pigeon; gp, guinea pig; pg, pig; guinea gp, pigeon; pn, chicken; ck, sheep; sh, chimpanzee; cz, mouse; ms, human; hm, dog; dg, monkey; mk, baboon; bb, rabbit; rb, rat; rt, Excluding 7) = 0,01 (column 6) and Excluding = 0.05 (column level significance BWthe at and CU between ofcorrelation a lack of because determined not n,d,: determination of Coefficient SR90I07A Stavudine Sumatriptan Talsaclidine 37 Thiopentone 3.5 Tamsulosin Tebufelone Theophylline Zidovudine Tosufioxacin 64 TolcaponeTolterodine TrimethadioneTroglitazone Zalcitabine 12 4.1 15 0.82 Tiludronale activaor Tissue-nlasminojien 1,5 0.56 0.977 Tylosin Zomepirac 10 Table (continued) 2,1, ,M ,ri both Excluding m

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. b r r n Scenarios* ms rt rb mk dg hm range** 20% 30% 20% 30% CV CV CV CV I 0 • • 125 0.75 0.74 0 3 9 6 0 3 8 6 (0.63-0.87) (0.53 - 0 3 5 ) 2 • • • • 250 0.74 0.74 0.994 0 3 8 8 (0.64-0.84) (0.58-0.91) 3 • • • #• 0 700 0.75 0.75 0.996 0 3 9 0 (0.67-0.83) (0.62 - 0.88) 4 3500 0.75 0.75 0.996 0 3 8 9 (0.69-0.81) (0 .6 2 - 0.88) 5 •• • 20 0.76 0.72 0.992 0 3 5 4 (0.57-0.94) (0 .2 9 -1 .2 ) 6 •• 0 0 56 0.75 0.73 0 3 9 0 0 3 6 8 (0 .6 0 - 0.8 8 ) (0 .5 0 - 0.95) 7 •• 0 0 0 280 0.75 0.76 0 3 9 2 0 3 8 0 (0.65-0.85) (0 .5 8 - 0.93) S • 0 0 5.6 0.80 0.74 0 3 7 4 0.925 (0 .5 0 -1 .1 ) (023 - 13) 9 • 0 0 0 28 0.74 0.75 0 3 8 7 0371 (0.58-0.90) (0 .4 7 -1 .0 ) 10 0 0 0 14 0 .74 0.73 0388 0 3 6 9 (0.50-0.98) (0 .4 4 -1 .0 )

T able 2 2 Simulated b values In different scenarios w ith varied body weight ranges

ms: mouse, 0.02 kg; rC rat, 0.25 kg; rb: rabbit. 2.5 leg; mk: monkey, 5 kg; dg; dog, 14 leg; hm: human, . . 7 0 k g . ** Range= maximum body weight/minimum body weight in each scenario f The mean b value with 95% confidence interval (boldface in the parenthesis) was obtained from 100 simulations where linear regression analyses were applied to the log-log-transformed CLversus BW data with either a 20% or a 30% coefficient of variation (C V ) in CL. n The mean correlation coefficient (r) o f linear regression from 100 simulated experiments per scenario

38

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Group* no. o f no. o f slope, b (95% C l) (99% CD xenobiotics data points I 9 41 0.78 0 .7 3 -0 .8 3 0 .7 2 -0 .8 4

2 21 105 0.65 0.62-0.69 0.61 -0.70

3 39 203 0.75 0.72-0.78 0.70-0.79

4 22 111 0.76 0.71 -0.81 0.70-0.82

O verall 91 460 0.74 0.72-0.76 0.71-0.76

Table 23. Summary o f the statistical results in Fig 23

* Group I = protein; group 2 = xenobiotics that were eliminated mainly by renal excretion; group 3 = xenobiotics that were eliminated mainly by extensive metabolism; group 4 = xenobiotics that were eliminated by both renal excretion and non-renal metabolism.

39

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 30 Mean = 0.74. SD = 0.16 Nonnal distribution 25 -

20 - o>» c © 3 Er ^

10

5 -

i i 0.0 0-1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1.0 1.1 1.2 1.3 1.4 1.5 Allometric exponent

Figure 2.1. The frequency distribution o f the b values fo r the 91 xenobiotics that showed statistically significant correlation between log CL and log BW in Table 2.1. The frequency o f the b values, at an interval o f 0.1, from 0.2 to 1.2 was plotted against the midpoint o f each interval o f b values. The dotted line represents a fitted Gaussian distribution curve.

40

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. m too so as

60

40 4 0

20 20 0 0 338S23S35g3

mouse. rwL mbttt. monkey, dog m o urn ra t. m b t*. mortmy. dog. hunan

6 0

4 0

20 0 3333S38SS8S1 SS3S888SS8

mtmooitmortmy rmt, nobt nonhtyjjoB »> O — 30% CV — 20% CV O’ & u. 33S88SS83 HUS3S8SU

1Q0 wtmted monte* dog, numm ^ ^ roote. monte* dog

i 60

6 0

4 0

20 0 - 3SS388S33i 33S3S388S83

too monte*, dofc. o m it

6 0

6 0

4 0

20

:LJI l a , h j J l f c . 888388S88 33S8S3S8S88

Exponent

F ig u re 2 2 . The frequency distribution o f the simulated b values in the ten scenarios where the number o f animal species and the range o f body weight were varied. The b values were obtained by applying linear regression analyses on the Iog-Iog- transfbrmed, error-containing CL versus BW data w ith either a 20% (gray) or a 30% (black) coefficient o f variation (CV) in CL. 41

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1000

100

o *N o E

0.01 r

0.001 0.001 0.01 1 10 100 1000 10000 Body Weight (kg)

Figure 23. The relationship between normalized clearances fCL and body weights (BW) fo r the 91 xenobiotics (n = 460) that showed statistically significant correlation between log CL and log BW in Table 2.1. The relationship follows the equation: log C[.normalized = 0.74 log BW +• 0.015. r = 0.917. The 99% confidence interval o f the regression slope was 0.71 - 0.76. The different colors represent different subgroups o f xenobiotics: red. protein: blue, xenobiotics that were eliminated mainly (>70%) by renal excretion: green, xenobiotics that were eliminated mainly (>70%) by metabolism: black, xenobiotics that were eliminated by both renal excretion and metabolism. The result o f each subgroup can be viewed in the web version by moving the cursor to each symbol legend.

42

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. -150 -100 -50 0 50 100 150 % Deviation

Figure 2.4. The deviation between the fitted and the observed human CL for 68 xenobiotics. The fitted human CL o f each xenobiotic was obtained by applying linear regression on the log-log-transformed CL versus BW data from d iffe re n t animatspecies including human. The deviation was calculated as I00*(C LObsen,ed - CLfioed)/CLgned- The mean deviation was 0.62%.

43

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. C H A P T E R 3

Power-Law Scaling of the Drng-Drug Interaction Network

3.1 Introduction

Many complex natural phenomena could be described by a simple power-Iaw

equation. The question as to how such regularity existed in Nature has intrigued many

scientists in various disciplines. For example, in a recent colloquium 1 on “ self-organized

complexity in the physical, biological, and social sciences” held by the National

Academy o f Sciences (NAS) o f the United States, power-Iaw scaling was ubiquitous in

many examples presented therein, ranging horn allometric scaling in biology to social

networks.

A power-Iaw relationship between a measured quantity y and an independent

variable x is o f the form:

y = a x b ( 1)

where a and b are constants. The scaling exponent b characterizes the rate o f change in y

w ith respect to the change o f x, since

(2) d \n x

The b value could be either positive or negative, depending on the variables o f interest.

44

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. For example, the ailometric scaling o f metabolic rate 2 and xenobiotic clearance 3 has a

positive scaling exponent, whereas the exponents for the ailom etric scaling o f the cardiac

frequency and the half-life o f drugs are negative .2 Another example that w ill give rise to

a negative b value is the power-Iaw frequency-size distribution feund in many fields,

such as the frequency distribution for the rupture area o f earthquakes 1 and for the size o f

companies .4 Besides the sign o f the b value, its absolute magnitude is o f particular

importance, as it could be a manifestation o f the topology o f a system or o f the

underlying mechanism o f a process.

Complex networks can be found in systems as small as a cell and in those as large

as the human society: In the former, the interconnection o f molecules by chemical

reactions was considered as an intricately-woven web; in the latter, complex social

networks can be formed by direct or indirect interactions o f human beings. Generally

speaking, a network is composed o f nodes (vertices) that are connected by links (edges ).5

The degree o f a node specifies the number o f links that a node has. Before 1999, the real

networks mentioned above would be realized by the random graph theory in which the

connections o f nodes are considered as a random process .5 Accordingly, the theory

would predict that the probability o f a randomly selected node w ith exactly k links

(degree &), P(k), fellows a Poisson distribution, which is a bell-shaped curve with a peak

a t the mean k. However, the seminal work by Barabasi and Albert in 1999 indicated that

real networks including the World-Wide Web (WWW) have their degree distribution

significantly deviated from a Poisson distribution .6 In their empirical results, the degree

distribution o f the real networks was extremely skewed w ith the ta il following the power

law : P {k ) °c fc~T (3) 45

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. In the past 3 years, the power-Iaw degree distribution has beat described fo r a

number o f real networks, such as the WWWM and cellular , 10 ecological , 11 and scientific

ty e collaboration networks etc. Moreover, theoretical models that were able to capture the

general feature o f the power-Iaw distribution have been proposed . 13*18 Two recent

reviews 5’19 highlighted the intensity o f growing interests in understanding the topology

and evolution o f complex networks.

Drug-drug interaction poses a potential threat to patients who receive treatment with

multiple medications. Whether two co-medicated drugs w in interact with each other

depends on many factors: the drug properties (physicochemical, pharmacokinetic and

pharmacodynamic), the basic characteristics o f patients (age, sac, race etc) and the

disease states. The general mechanisms for an interaction to occur include the

pharmacokinetic interactions (or ADME interactions) by which the processes o f drug

absorption, distribution, metabolism and excretion are affected and the pharmacodynamic

interactions by which the effects o f a drug at its site o f action are modified .20 Although

some drug interactions may produce a beneficial effect, most interactions resulted in

either unwanted adverse reactions or reduced efficacy. While adverse drug reactions may

be due in part to elevated drug concentrations, additive or synergistic interactions and

combined toxicities, factors such as reduced chug concentrations or antagonistic

interactions may cause a reduction in efficacy .20 Sometimes the interactions are severe or

even fetal so that a drug is withdrawn from the market. Therefore, the detection o f

potential drug interactions has become an important part o f drug safety evaluation in the

drug development process, during which a new drug entity (NDE) must be tested for its

ability to modulate the pharmacokinetics or pharmacodynamics o f those frequently co- 46

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. prescribed drags .21 Furthermore, the detection o f drug interactions extends over the post­

market period. Currently, a substantial amount o f information about drug-drug

interactions has accumulated. I f one treats the interacting drugs as nodes and connect

those nodes w ith undirected links that represent the interactions, a drug-drug interaction

network can be obtained. Interesting questions pertaining to network analysis then arise:

What is the topology o f the drug-drug interaction network? How does this network

evolve? The answers to these questions may provide a heuristic view towards the nature

o f drug-drug interactions.

3.2 Materials and Methods

3.2.1 Data acquisition

The drug-drug interaction information was retrieved from a comprehensive source

reference-Drz/g Interactions: a source book o fadverse interactions, their mechanisms,

clinical importance and management by Stockley,20' “ *23 which was first published in

1981 and was continuously updated since then. Besides its comprehensiveness, the

information covered was mainly based on the published primary references in which

clinical and experimental evidences were provided. To investigate the evolution o f the

drug-drug interaction networks and the effect o f network size on its topology, we

extensively surveyed the source reference’s firs t (1981),23 second (1991)22 and fifth

(1999)20 editions, which were separated by a time interval o f 8 —10 years and had about

600,1200 and 2400 monographs, respectively. The source references included not only

the interaction cases, but also those that showed a lack o f interaction. The quality o f

interaction information varied widely (controlled versus uncontrolled clinical trials, case 47

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. reports, in vitro studies, animal studies ere), and it was therefore imperative to establish

inclusion and exclusion criteria. I only considered the drug-drug interactions that have

been detected in patients or healthy subjects and excluded (I) those where only in v itro

data were reported; (if) those where only animal studies were conducted; (iii)

manufacturer’s statements and generalized claims; (iv) those where the interactions were

found when more than 2 drugs were co-medicated and there was no further evidence to

support interaction for any particular pair o f drugs. Furthermore, pharmacokinetic

interaction between a pair o f drugs was considered only when the change o f mean

pharmacokinetic parameters in either drugs exceeded 20%. Overall, I obtained three

databases that consisted o f351,636 and 966 drugs and 742,1858, and 3351 pairs o f

interaction, respectively

3.2.2 Data analysis

Based on the drug-drug interaction information from the databases, I prepared

histograms that describe the frequency distributions for the number o f interactions that a

given drug has. Dividing each element o f the histogram into the total number o f drugs in

the database, the p ro b a b ility that a given drug has k interactions, P (k), can be obtained.

A. logarithmic binning approach ,24 w hich can smooth the obtained distributions fo r

larger values o f k while retaining the nature o f the distribution, was used according to the

following rule:

P j k ) = (4) l(k r,*D

48

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. w h e re ^ Pn(k ) is the summed p ro b a b ility in the nth b in and the b in w id th /^ ^ "* ,k ™ ) is

the number o f integers in the range k™ to . For the logarithmic binning I set bin

w idths w ithk™ I k™ = constant. To obtain the power-Iaw exponent, the log-Iog-

transformed data points ( log£„, log Pn) were fitted to a straight line using linear

regression, where logfc„ = log iy f k ^ k ^ } .

PAJEfC, a program for large-network analysis and visualization, was used for

constructing the drug-drug interaction network. This program is available at

http://vIado-rmfuni-Ii.si/pub/networks/paiek/

3.23 Theoretical considerations

The drug-drug interaction network consisted o f two basic elements: a node (vertex)

representing a given drug that has one or more interaction(s) w ith other drugs and a lin k

(an edge) representing the interaction between two drugs. The degree (ki) o f a node i in

the network was defined as the number o f links that this node has. Thus, the degree (£,)

o f a given drug in the drug-drug interaction networks described the number o f

interactions that link to this drug. The evolution o f a network is a dynamical process,

w hich im plies that the degree o f a node changes over time.

Considering a model for drug-drug interactions (F ig 3.1), the degree (kt ) o f drug i

w ill increase every tune a new interaction that associates w ith this drug is detected. The

new interaction may come from a drug that had never been detected previously. The

probability o f this process, is proportional to the degree o f drug /, k*. Moreover, the

49

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. new interaction may derive from a drag, say/, which already existed in the network, hi

this case, the probability o f interaction, denoted as

the degrees o f drug i and j , ktk j. The probabilities that assign to the above tw o processes

are based on the assumption that a drug has a higher chance to interact w ith drugs that

have a higher number o f interactions detected previously. This is the so-called “ rich-get-

richer” or preferential linking - a common phenomenon in other real networks .5

The size o f the network is continuously growing as the numbers o f its nodes (drugs)

and links (interactions) increase over time. After a relatively long period o f evolution,

the network may self-organize into a structure that can be characterized by the probability

distribution o f the degree (&), P(k), which by definition is the probability that a given

node has k lin ks (k > I). The empirical results o f the present study indicated that P(k)

follows a power-Iaw distribution, P(k) <* k' r , where yean be predicted using the

continuum approach 13*18 that was successfully applied to modeling the scientific

collaboration network by Barabasi et ah 25 and the web o f English words by Dorogovtsev

and Mendes .6

3 3 Results

The drug-drug interaction network is graphically represented in Fig. 33. The

network, consisting o f966 drugs and 3351 pairs o f interactions, was generated from the

updated drug-drug interaction information up to 1999. The dots represent the interacting

drugs and the lines the interactions that have been detected for any given pairs o f drugs.

This is a highly inhomogeneous network, w ith a dense interior fille d with highly

50

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. interacting drugs and a sparse exterior comprised o f a large number o f drugs that sparsely

interact The heterogeneity o f the network could be further exemplified by the following

observation: The top 40 most interacting drugs (red dots), while represented only 4.1 %

(40/966) o f all the drugs in the entire network, have their number o f interactions > 30 per

drug and together contributed more than 36 % (2421/6702) o f the interactions. In

contrast, as many as 696 (hugs (72 %) have 5 interactions or less per drug, which

constituted only 21 % o f all the interactions (1438/6702). The data suggested that the

network behavior was dominated by a small fraction o f drugs that exhibited a higher

tendency to interact with other drugs - a “rich-get-richer” phenomenon in the drug-drug

interaction network.

The frequency distributions for the number o f interactions a given drug has is

shown in Fig. 33, where the data were retrieved from the three drug-drug interaction

databases w ith the number o f interacting drugs increasing from 351 (Fig. 33A) to 966

(Fig. 33Q and the total number o f interactions for all drugs increasing from 1484 to

6702. The bar charts illustrated nearly L-shaped histograms, which upon log-tog

transformation corresponded to the scatter plots shown in the insets. By visual

examination, the scatter plots could be described by straight lines, hi addition, the

distribution was “fat-tailed” , which is one o f the characteristics o f a power-Iaw

distribution.

To estimate the power-Iaw exponent and to compare the degree distribution P(k) o f

the three databases, the frequency data in Fig. 33 were transformed to probabilities and

the resulting probability distributions were smoothed fo r larger k(k> 4) using the

logarithmic binning approach described in Materials and Methods. It is remarkable that 51

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. the probability degree distributions fo r the three networks w ith different sizes were

superimposed and followed a straight line with a slope close to -1.5 on a Iog-Iog plot, Fig..

3.4. The basic characteristics o f the drug-drug interaction networks studied are

summarized in Table 3.1. Notably, the average number o f interactions per drag (the

density o f the network), <&>, increased as the network size was enlarged, suggesting that

the network was subject to a non-linear growth. Nevertheless, the network exhibited

scale-free characteristics as it followed a power-Iaw degree distribution w ith an almost

invariant scaling exponent.

The top-ten most interacting drags in the three databases are listed in Table 32..

The members o f the top-ten lists during the evolution o f the network only changed to a

slight extent: Four members (phenylbutazone, probenecid, tolbutamide, and Ievadopa) in

the 1981 database fe ll out and were replaced by 4 new members (cimetidine, cyclosporin,

propranolol and theophylline) in the 1991 database, whereas 2 members (phenobarbital,

digoxin) in the 1991 databases were replaced by the other tw o new members

(carbamazepine, rifam picin).

3.4 Discussion

Many seemingly unrelated scientific disciplines have recently arrived at an

intersecting point - the discovery o f power-Iaw scaling. Especially, numerous power-Iaw

frequency distributions have been described for various attributes in diverse systems.

Examples are the number o f pages w ithin a WWW site 8 or the number o f links to a web

page ,7 the number o f chemical reactions in which a metabolic substrate participates , 10 the

month-to-month variation o f a hospital waiting list ,27 and the size o f a company .4 A 52

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. distribution in a power-Iaw form is free o f scale and often implies non-randomness .5 To

pharmaceutical scientists, a fam iliar example o f power-Iaw scaling is the ailometric

scaling o f a pharmacokinetic parameter (e.g., clearance) in different species with body

sizes spanning over many orders o f magnitude .3

In this chapter, I described a drug-drug interaction network in which the interacting

drugs were treated as nodes and were connected w ith undirected links that represent

interactions. The degree (the number o f interactions that a drug has) o f the resulting

network followed a power-Iaw distribution. The scaling exponent was close to -1.5 and

independent o f the network size. The power-Iaw degree distribution o f the drug-drug

interaction network can be derived according to the model in Fig. 3.1. The network was

assumed to be governed by the mechanism o f preferential linking, which suggested that

highly interacting drugs have a higher tendency than less interacting drags to acquire a

new interaction. During the time evolution, the number o f interaction (£,) o f drug i

increased as a result o f newly detected interactions that associated with it. The

interaction occurred either with drugs that had never been identified for any interaction or

with drugs that had existing interactions. The probability for the two interaction sources

was denoted as & and respectively, which are o f the following forms 25

(5)

and

(6 )

53

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Accordingly, the evolution o f the number o f interactions (£;) that drag i has can be

described as:

(7 )

dN dM where is the rate o f acquired interactions from new incoming drugs and - j -

represents the rate o f newly identified interactions among existing drugs in the network.

Note that the factor 2 in equation (7) implies that inside a network any given link has 2

equal ends that are distributed preferentially among the existing nodes. We assumed that

new incoming (hugs jo in the drug-drug interaction network at a constant rate; therefore,

dN - - a . (8)

Furthermore, I considered that the flux o f new interactions inside the existing network

increased nonlinearly and was proportional to tb, where b> 0 . Thus,

(9)

where a and f i in ( 8) and (9) are positive constants. The cumulative probability

in (7) is approximately equal to tp-„ if we assumed that m ultiple links (between the same

pah o f drugs) were absent in the drug-drug interaction network and applied the

procedures taken by Barabasi et al .25 Using (5), ( 8), (9) and the approximation for

^ <1* ^ , equation (7 ) can be w ritte n as:

(10)

54

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. where the total degree o f the n e tw o r k , £ tfj , can be derived from ( 8) and (9 ):

(II)

Substituting (11) into (10), we obtained

dki _ a +2 fit* (12) d t

. I I

According to my empirical data, > 90% o f drug interactions in a new database resulted

from interactions among drugs that were members o f the old database. Therefore, one

ft can a s s u m e ^ j-t **1 » a t for large t and equation ( 12) can be sim plified as:

f= (7 h ‘>- (i3>

The solution o f (13) with an in itia l condition ytj(/j) = I for the degree o f drug / at t = tt is:

(14) \ /

It remained to know the probability distribution o f tiy which was assumed to be uniform ly

distributed in the [ 0, t] interval because the interacting drags joined the network randomly

at a constant rate. Therefore, the probability distribution o f 4 was given as:

1 g iO = 7 - (15)

Using (14) and (15) and the change o f variable technique ,28 the degree distribution, P (k),

can be obtained as follows:

55

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. P ( * ) = g ( r f> fAl |=_i—fc^1+ST) (16) L ^ J U w l 6+1

Accordingly,

(17)

Compared w ith (3), yin the power-Iaw exponent is o f the following form:

Obviously, l 0. Moreover, y = 1.5, if 6 = I, which reproduced our

empirical finding.

I used the three databases to examine the assumptions o f preferential linking and

nonlinear growth in the interaction network. The average degree, , increased as the

network size enlarged (Table 3.1), which implied that the total number o f interactions in

the drug-drug interaction network increased noniineariy over time. The data suggested a

nonlinear growth o f the network. To demonstrate that preferential linking operated in the

interaction network, 1 evaluated the probability distribution o f a given drug w ith kt

interactions to acquire a new interaction. In so doing, the number o f drug interactions

was compared fo r drugs that appeared in two databases published at different times (e.g.,

1981 versus 1991,1991 versus 1999). The cumulative probability distribution was

calculated as follows :5

(19)

56

I Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. where M , is the number o f interactions added in the later database for drugs that have k,

interactions in the earlier database, and Ak is the total number o f interactions added to

the previous database. By plotting r(k) versus k in a log-Iog plot, whether preferential

linking existed can be determined from the slope o f the plot. If the slope is equal to I, it

indicates no preferential linking because M , /A k would be a constant in this case.

However, if the slope is greater than I, which may imply Akt / Ak <* k s (s > 0), and that

preferential linking occurs .3 O ur analyses showed that the slope o f the cum ulative

probability distributions in the log-log plots was larger than I (Fig. 3.5), thereby

suggesting that preferential linking operated in the drug-drug interaction network.

Moreover, the result was robust, regardless o f how the networks were compared. The

consistency o f this finding in conjunction w ith the invariant power-Iaw scaling o f the

degree distribution suggested that the global properties o f drug-drug interaction have

remained unchanged over the past decades, even though, at the microscopic level, new

drugs and new interaction mechanisms have emerged.

The present study demonstrated that power-Iaw frequency distribution exists in the

pharmaceutical system, which coincided w ith recent multidisciplinary interests in

network analysis and power-Iaw scaling. It was interesting to find that the evolution o f

the drug-drug interaction network was sim ilar to many complex networks that exhibited

scale-free characteristics .5 However, to many pharmaceutical scientists, a more

intriguing question could be: how could the current analysis add to our knowledge about

drug-drug interaction? Specifically, what does preferential linking implicate in drug

interactions?

57

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. What i f the probability fo r a drug to interact w ith any other drugs follows a Poisson

distribution? This would im ply that the process is random and the majority o f drugs have

the number o f interaction close to a characteristic (mean) value, which is the central

tendency phenomenon. The Poisson distribution would further im ply that it is

exponentially rare fo r a drug to have an extremely large (or small) number o f interactions,

say S standard deviations away from the mean. Were this the case, drug-drug

interactions would be much more d ifficu lt than is currently the case for researchers,

health care practitioners, and regulatory agencies to predict, prevent and set guidance for.

Indeed, we know the existence o f some classes o f drugs that have higher chance than

others to cause an interaction. This is strikingly similar to social networks in which a

small number o f individuals have disproportionately high popularity. Once a person has

been recognized for his/her success, the chance for this individual to get connected

increases. Eventually, the connectivity in the social network becomes inhomogeneous

and the degree distribution may follow the power law. Similar situations apply for the

drug-drug interaction network.

The heterogeneity o f the drug-drug interaction network could be understood as the

consequence o f at least two driving forces: the feature o f a drug and the decision-making

o f individuals who participate in the field. Like the social networks, the characteristics o f

a given drug may determine its detectability in a potential drug-interaction context. By

examining the lists for the top-10 most interacting drugs (Table 3.2), one finds that 4

compounds (phenytoin, alcohol, warfarin and lithium ) have remained on the list since

1981. Among them, phenytoin, warfarin and lithium are drugs w ith extremely narrow

therapeutic windows and are frequently used fo r diseases that require polypharmacy 58

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. treatment. Alcohol, though not a chug by a strict definition, is pharmacologically sim ilar

to many drugs that depress the CNS. The prevalence o f alcohol consumption and the

potential o f an interaction among alcohol and CNS depressants may have resulted in

alcohol being on the top lists over decades.

Although drugs w ith a low therapeutic index, such as digoxin, theophylline and

those mentioned above, tended to have high incidences o f drug interaction, cim etidine, a

drug on the top-ten lists in the 1991 and 1999 databases, reflected that the research on

drug interactions has undergone a significant transformation in recent years. When taken

alone, cimetidine is a rather safe drug. However, it inhibits the metabolism o f certain

coadministered drags, thereby increasing the potential fo r adverse drag reactions .29 The

ability o f cimetidine to inhibit the cytochrome P450 (CYP) fam ily o f enzymes, especially

CYP3A4 and CYP2D6, may contribute to its high propensity to cause an interaction .29

Recently, metabolic drug-drug interactions associated w ith CYP have become the central

issue o f drag interaction studies, partly due to the advances in the molecular biology o f

CYP and the development o f in v itro methodologies that facilitate the detection o f a

potential interaction .21 The rapid expansion in detecting CYP-related interactions was

evident by the addition o f carbamazepine (a substrate and an inducer fo r CYP3A4 )30 and

rifampicin (an inducer for CYP3A4, CYP2C9 and CYP2C19)30 to the top-10 list the first

time in the 1999 database (Table 3.2). Moreover, 9 o f the top 10 drags in the 1999

databases (Table 3.2) have been identified as substrates, inducers o r inhibitors o f the CYP

isozymes. 30

Human factors might play an important role in orchestrating the structure o f the

drug-drug interaction network, because the decision-making process o f researchers was 59

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. involved. Specifically, the decision o f winch drug interactions are to be investigated, a

small number o f drugs were more frequently identified for consideration than were others.

For example, a recent survey o f new molecular entities approved from 1987 to 1997

indicated that the most commonly studied interacting drugs did not change over this

period, w ith cimetidine and digoxin being the top 2 drugs o f choice for drug interaction

studies .31 These drugs were popular because either their unique characteristics in causing

an interaction have been long recognized or the chance o f these drugs to be

coadministered w ith the new chemical entities is high. The results suggested that the

decision-making process o f researchers was based on drug interaction information that

was biased toward more recognizable drugs. It was possible that this process might

contribute to the “rich-get-richer" phenomenon in the drug-drug interaction network.

3.5 Conclusion

This study from a network standpoint provided a heuristic view on drug-drug

interactions. W hile thug interaction information has continuously accumulated and the

nature o f interactions has been modified over the decades, the structure o f the drug-drug

interaction network remained robust with the degree distribution o f the network

following a power law w ith an invariant scaling exponent. A dynamic model was

proposed to account for the observed scale-free structure o f the network.

3.6 Summary

Using drug interaction information that spanned decades, a drug-drug interaction

network was described in which the interacting drugs were treated as nodes and were 60

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. connected w ith undirected links that represented interactions. The degree (the number o f

interactions that a drag has) o f the resulting network followed a power-Iaw distribution.

The scaling exponent was close to -1.5 and independent o f the network size. A dynamic

model was proposed to account for the observed scale-free structure o f the network.

This study from a network standpoint provided a heuristic view on drug-drug interactions.

While drag interaction information has continuously accumulated and the nature o f

interactions has evolved over the decades, the structure o f the drug-drug interaction

network remained robust with the degree distribution o f the network following a power

law w ith an invariant scaling exponent.

3.7 References

1. Turcotte DL, Rundle JB. Self-organized complexity in the physical, biological, and social sciences. Proc Natl Acad Sci USA. 2002;99(suppL l):2463-2465.

2. West GB, Brown JH, Enquist BJ. A general model for the origin o f ailometric scaling laws in biology. Science. 1997;276:122-126.

3. Hu TM, Hayton WL. Ailometric scaling o f xenobiotic clearance: Uncertainty versus universality. AAPS PharmSci. 2001^(4), article 29. (http://www.pharmsci.org /1

4. Axtell RL. Zipf distribution o f U.S. firm sizes. Science. 2001;293:1818-1820.

5. Albert R, Barabasi AJL. Statistical mechanics o f complex networks. Mod. Phys. 2002;74:47-97.

6 . Barabasi AL, Albert R. Emergence o f scaling in random networks. Science. 1999;286:509-512.

7. Albert R. Jeong H, Barabasi AL. The diameter o f the world-wide web. Nature. 1999;40I:I30.

8. Huberman BA Adamic LA. Growth dynamics o f the world-wide web. Nature. 1999;401:I3I.

61

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 9. Adamic LA, Huberman BA. Power-Iaw distribution o f tbe world wide web. Science. 2000287:2115.

10. Jeong H, Tombor B, Albert R, Oltvai ZN, Barabasi AL. The large-scale organization o f metabolic networks. Nature. 2000;407:651-654.

11. Montoya JM, Sole RV. Small world patterns in food webs. ArXiv.org e-Print archive. 2000. http://www.arxiv.org/abs/eond-mat/ 0 0 l 1195

12. Newman MEJ. The structure o f scientific collaboration networks. Proc Natl Acad Sci USA. 2001;98:404-409.

13. Barabasi AL, Albert R, Jeong H. Mean-field theory for scale-free random networks. PhysicaA. 1999272:173-187.

14. Krapivsky PL, Redner S, Leyvraz F. Connectivity o f growing random networks. Phys. Rev. Lett. 2000;85:4629-4632.

15. Dorogovtsev SN, Mendes JFF, Samukhin AN. Structure o f growing networks with preferential linking Phys. Rev. Lett. 2000;85:4633-4636.

16. Albert R, Barabasi AL. Topology o f evolving networks: Local events and universality. Phys. Rev. Lett. 2000;85:5234-5237.

17. Kirillova OV. Communication networks with an emergent dynamical structure. Phys. Rev. Lett. 2001;87:068701-+.

18. Dorogovtsev SN, Mendes JFF. Scaling properties o f scale-free evolving networks: Continuous approach. Phys. Rev. E. 200l;6305:6l25-+.

19. Dorogovtsev SN, Mendes JFF. Evolution o f networks. ArXiv.org e-Print archive. 2001. http://www.arxiv.org/abs/cond-mat/Q 106144

20. StockleylH. Drug interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, London, UK: Pharmaceutical Press, 1999.

21. Thummel KE, Kunze KL, Shen DD. Metabolically-based drug-drug interactions: principles and mechanisms. In: Levy RH, Thummel KE, Trager WF, Hansten PD, Eichelbaum M , eds. Metabolic drug interactions, Philadelphia, PA: Lippincott Williams & W ilkins, 20002-19.

22. StockleylH. Drug interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, Oxford, UK: Blackwell Scientific Publications, 1991.

62

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 23. StockleylH. Drag interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, Oxford, UK: Blackwell Scientific Publications, 1981.

24. MeakmP. Fractals, scaling and growth far from equilibrium, Cambridge, UK: Cambridge University Press, 1998.

25. Barabasi AL, Jeong H, Neda Z, Ravasz E, Schubert A, VicsekT. Evolution o f the social network o f scientific collaborations. ArXiv.org e-Print archive. 2001. httpjVwww.arxiv.org/abs/cond-mat/0104162

26. Dorogovtsev SN, Mendes JFF. Language as an evolving word web. Proc R Soc LondB. 2001;268:2603-2606.

27. Smethurst DP, Williams HC. Power laws: are hopital waiting lists self-regulating? Nature. 2001;410:652-653.

28. Hoel PG. Introduction to mathematical statistics, New York, NY: John Wiley & Sons, Inc, 1962.

29. Paine MF. H2-receptor antagonists. In: Levy RH, Thummel KE, Trager WF, Hansten PD, Eichelbaum M, eds. Metabolic drag interactions, Philadelphia, PA: Lippincott W illiams & Wilkins, 2000;653-659.

30. Papp-Jambor C, Jaschinski U, Forst H. Cytochrome P450 enzymes and their role in drug interactions. Anaesthesist. 2002;51:2-15.

31. Marroum PJ, Uppoor RS, Parmelee T, Ajayi F, Burnett A , Yuan R, Svadjian R, Lesko LJ, Balian JD. hi vivo drug-drug interaction studies-a survey o f all new molecular entities approved from 1987 to 1997. Clin Pharmacol Ther. 2000;68:280-285.

63

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. DATABASE Y E A R tB> N ™ k W w y(e)

I 1981 351 1484 4 2 1.48

2 1991 636 3716 5.8 1.50

3 1999 966 6702 6.9 1.44

Table 3.1. Basic characteristics o f drug-drug interaction networks

(a> Year o f publication <6) Total number o f interacting drugs (c) Total number o f interactions for a ll drugs w Average interactions per drug, = k/N (<) Power-Iaw exponents

64

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1981 1991 1999

RANK DRUG k ,' DRUG fc1 DRUG

I A lcohol 53 Phenytoin 107 Phenytoin 153

2 Phenytoin 51 W arfarin 102 A lcoho l 135

3 W arfarin 44 A lcohol 101 C im etidine 133

4 Phenobarbitai 26 Cimetidine 80 W arfarin 133

5 Lithium 24 Cyclosporin 56 Cyclosporin 120

6 Phenylbutazone 24 Propranolol 54 Carbamazepine 98

7 Probenecid 24 Lith ium 52 R ifam picin 96

8 Tolbutamide 23 Phenobarbitai 52 Lith ium 91

9 Levodopa 22 Theophylline 52 Theophylline 86

10 D igoxin 20 D igoxin 51 Propranolol 75

Table 3.2. The Top-10 Most Interacting Drugs in the Three Databases

^Number o f interactions

i Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure 3.1. A model fo r the drug-drug interaction network. Each node represents a drug. The line between two nodes represents an interaction. A t each time step, a given drug i inside an existing interaction network, can interact with a new interacting drug (blue nodes) outside the network, with a probability ,. A t the same time, drug i can also interact with another drug/ in the existing network with a probability f. The new node w ill become part o f the existing network and. therefore, the size o f the existing network w ill continuously grow due to the addition o f new nodes during the evolution o f the network.

66

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure 3.2. A drug-drug interaction network consisted o f966 drugs (dots) and 3351 interactions (lines). Red dots represented the top 40 most interacting drugs. PAJEK. a program for large-network analysis and visualization, was used for constructing the drug- drug interaction network. This program is available at http://viado.finf.uni- ii.si/pub/networks/paiek/

67

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ISO 1000

100 D o $ too o c © 10 \ 9 t OS GD 50 1 at to too TOGO

20 40 60 SO 100 120 t40 160 180 Number of Interactions

B 250 1000

200 too >» c tso © 3 g too i t at 50 i to too taoo

20 40 60 80 100 120 140 160 180 Number of Interactions

350 1000 300 too 250 o>» c 200 © 3 V 150 9 100 at too tooo

a 20 40 60 80 100 120 140 160 180 Number of Interactions

Figure 33. The frequency distribution for the number o f interactions a given drug has. Data were acquired from databases published in 1981 (3S1 drugs, A ), 1991 (636 drugs, B) and 1999 (966 drugs, Q . 68

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1

0.1

0.01 CL o 1981 0.001 V 1991 □ 1999 Slope ~ -1.49 0.0001 1 10 100

Figure 3.4. The probability degree distributions for the drug-drug interaction networks.

69

I Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1 0 • 1981-1991 O 1991-1999 Slope = 1

0.1 £ U 0.01

0.001

0.0001 1 10 100 Number of pre-existing interactions (k)

Figure 35. Cumulative preferential linking for the drug-drug interaction network. The tine corresponds to no preferential linking.

70

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. C H A P T E R 4

Dynamic Biphasic Effect o f Snperoxide Dismutases on Nitric-Oxide-Mediated

Nitrosation Reaction

4.1 Introduction

The free radical nitric oxide (NO) contributes to diverse physiological and

pathophysiological processes. NO is synthesized from L-arginine by three types o f nitric

oxide synthase (NOS).1 W hile nNOS (NOS I) and eNOS (NOS HI) are constitutive

isoforms first found in neurons and endothelial cells, respectively, iNOS (NOS II) is an

inducible isofrom present in a wide range o f cells and tissues, especially during

proinflammatory conditions.2*3 Despite over a decade o f intense research, many aspects

o f NO physiological chemistry remain both paradoxical and controversial.4 A t low

concentrations (~ nM), NO modulates normal physiological functions such as regulation

o f vascular tone and intracellular signaling via a direct interaction with its targets/

However, high NO levels (~ftM ) can induce cytotoxicity, presumably attributable to

oxidative and nitrosative stresses.5 While generally slow in its reactions w ith many

molecules, NO reacts rapidly w ith reactive oxygen species.6

The concurrent production o f O f and NO during inflammatory and immunological

reactions has complicated an understanding o f the pathological mechanisms related to

71

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. these two free radicals. In addition, complexity is introduced by other cellular factors

that can modulate the abundance o f both reactive species. Accordingly, the

consequences o f oxidative and nitrosative chemistry may depend heavily on a delicate

balance among the processes that govern the formation and elimination o f both causative

agents. Elevated NO levels present at inflammatory sites provide the opportunity for NO

to successfully compete w ith superoxide dismutase (SOD) for reaction w ith O 2', thereby

generating the highly reactive and cytotoxic peroxynitrite (ONOO~) molecule.6 While

O f reduces NO-mediated oxidation,7 hydroxylation7 and nitrosation reactions,8-9 SOD

attenuates the inhibitory effect o f O 2'.8-9 SOD therefore plays a pivotal role in

modulating the consequences o f oxidative and nitrosative chemistry.

SOD sometimes demonstrates anomalous bell-shaped, dose-response relationship.10

W hile low concentrations o f SOD are cardioprotective, higher levels can exacerbate acute

cardiac injury.1 M3 Several mechanisms have been proposed to explain SOD's biphasic

effects including increased hydrogen peroxide production resulting from dismutation o f

Oj-m-w soD's peroxidase activity,19 and the dual roles o f superoxide in initiating and

terminating radical chain reactions.20 Recently, studies by O ffer et a I.21 showed that low

levels o f Cu,Zn-SOD inhibited Oj'-induced ferrocyanide oxidation, while its

antioxidative effect was lost at high [Cu,Zn-SODj. The authors proposed that in its

oxidized form Cu,Zn-SOD oxidizes the target molecule that it was supposed to protect.21

Related studies by Liochev and Fridovich22 implied that Cu^n-SOD functions both as a

superoxide reductase and a superoxide oxidase. Consequently, the findings o f Offer et al.

were attributed to the increase in SOR activity that accompanied increased [Cu,Zn-SOD].

72

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. SOD may affect NO bioavailability via a mechanism other than the dismutation o f

Q f- Indeed, an Of-dismutation-independent mechanism has been implicated in SOD’s

capability to (i) reversibly convert nitroxyl anion (NO") to NO,23 and (ii) enhance the

formation o f free NO from L-argmme by NOS.24 These findings suggest that SOD might

directly affect the reaction kinetics o f reactive nitrogen oxide species. Therefore, a

complex dose-response relationship may be anticipated fo r SOD in NO-mediated

nitrosative chemistry. This study used NO-donor-based kinetic analyses to investigate

the effect o f SOD on NO-mediated nitrosation reactions. The present findings depict for

SOD a novel dose-response relationship, which underwent dynamic transformations and

was highly sensitive to the substrate concentration.

4.2 Materials and Methods

4.2.1 Materials

SIN-1 was purchased from Calbiochem (La Jolla, CA). Cu,Zn-SOD, Mn-SOD, Fe-

SOD, catalase, diaminonaphthalene (DAN), dihydrorhodamine 123 (DHR), glutathione

(GSH), diethyltriaminepetaacetic acid (DTPA) and a ll other chemicals were purchased

from Sigma Chemical Company (St. Louis, MO). The chemical structures o f SIN-1,

DAN and DHR are shown in Fig. 4.1.

4.2.2 Kinetics of DAN nitrosation

The nitrosation kinetics o f DAN was studied using a fluorescence spectrometer

(Perkin Elmer LS 50B, Norwalk, CT). Reactions were performed in a 96-well microtiter

plate format at 25 ± I °C in the sample chamber. The fluorescence intensity o f nitrosated

DAN was measured at excitation and emission wavelengths o f375 and 430 nm, 73

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. respectively. A. stock solution o f DAN (30 mM) was freshly prepared in dimethyl

sulfoxide (DMSO), protected from light exposure and stored at 4 °C. The dynamic

range o f DAN in the nitrosation kinetics was studied by varying the concentration o f

DAN (0.78 — 300 pM ) in 200 p i o f phosphate-buffered saline (pH 7.4) that contained 0.1

mM DTPA and 120 U/ml catalase to eliminate hydrogen peroxide. Reactions were

initiated by adding an aliquot o f 4 p i SIN-1 stock solution (5 mM in DMSO) into 200 pi

DAN-containmg reaction buffers. The fluorescence intensity after the addition o f SIN-1

was measured at 10-min intervals. The nitrosation o f DAN in the presence o f SOD (0 -

1000 U/ml) and SIN-1 (100 pM ) was studied in the same pH 7.4 reaction buffer at 25 ± I

°C by measuring the increase in fluorescence every 10 min up to 8 hr.

4.23 Oxidation of DHR by SIN-l

The oxidation o f DHR was determined at 25 ± I °C in the above-mentioned

reaction buffer except that DAN was replaced by DHR (10 mM stock in

dimethyiformamide stored at -20 °C and protected from light). The fluorescence o f

rhodamine 123 generated from the oxidation reaction was measured at excitation and

emission wavelengths o f S00 and 530 run, respectively. The effect o f SOD (0 - 1000

U/ml) on SIN-I mediated DHR oxidation was studied in 200-CI DHR-containing

reaction buffer, where the DHR concentration (0.39 DM in the final reaction mixture)

was chosen based on a pilot experiment in which the dynamic range o f DHR was

determined. The oxidation kinetics o f DHR was typically monitored every 5-min up to 4

hr.

4 2 4 Cytochrome c reduction assay

74

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The activity o f Cu,Zn-SOD, Mn-SOD and Fe-SOD was determined by measuring

the ability o f SOD to inhibit the reduction o f acetylated ferricytochrome c by O f

generated from xanthine/xanthine oxidase.23 Reaction buffers (50 mM phosphate

buffer/0.1 mM EDTA, pH 7.8) containing xanthine (500 pM ), ferricytochrome c (4 pM)

and SOD at various concentrations (0,0.05,0.1,0.5,1 U/ml) were prepared. The

reaction was initiated by adding 17 p i xanthine oxidase solution (~L U/m l) into 830 pi

reaction buffer. The change o f absorbance at 550 nm was measured fo r 3 min at 25°C.

4.25 HPLC determination o f S IN -l degradation kinetics

An HPLC method26 was modified to study the effect o f SOD on the degradation

kinetics o f SIN -l (Appendix B). The method consisted o f a reversed phase column

(Waters Nova-Pak Ctg3.9 x 300 mm), a mobile phase (10 mM sodium acetate buffer

(pH3. lyacetonitrile/methanol = 92/6.4/1.6, flow rate = I ml/min), and UV detection at

254 nm. SIN -l was separated from its two degradation products, SIN-l A and SIN-IC,

with retention times o f 4.0,9.0 and 9.8 min for S IN -l, SIN -l A and SIN-IC, respectively.

The kinetics o f the SIN -l degradation at 25 °C was studied by serial sampling (0,30,60,

90,120,180,240,300,360,480 min), with an initial [S IN -l] o f 100 pM, with and

without 1000 U/ml Cu,Zn-SOD in the same reaction buffer as described in the nitrosation

kinetics section. The dose-response relationship o f Cn,Zn-SOD for the degradation o f

S IN -l was further investigated by determining the relative quantity (expressed as %

control) o f SIN-I and its degradation products after 4 hr incubation in reaction buffer

containing 100 pM SIN-I and various concentrations o f Cu^Zn-SOD (1000,250,62.5,

15.6,3.13,0.78 U/ml).

75

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 4.2.6 Method validation

The concentrations o f DAN and DHR used in the study were determined in pilot

studies to exclude the possibility o f fluorescence quenching. In addition, nitrogen oxide

species such as N O f, N Q r and ONOCF, which were likely generated during the reaction,

were evaluated and shown not to nitrosate DAN. SOD was also shown not to affect the

fluorescence o f DAN and DHR. Nevertheless, suitable controls (e.g., SOD/DAN for

SIN-1-mediated nitrosation kinetics, SOD/DHR for oxidation kinetics) were included in

each experimental run and the fluorescence reported was after subtracting out the

background fluorescence o f the controls.

4 3 Results

Kinetics o f DAN nitrosation. The kinetic profile o f SIN-1-mediated DAN

nitrosation, Fig. 42, was characterized by the formation o f fluorescence a t430 nm. The

nitrosation for the control (curve L) peaked a t- 180 min after the addition o f SIN-1 (100

pM). After the maximum, the fluorescence intensity tended to decline gradually. W ith

increasing concentrations o f Cu^n-SOD up to 15.6 U/ml, the rate and extent o f DAN

nitrosation increased disproportionately, curves 2-6 o f Fig 42A. hi constrast, [Cu,Zn-

SOD] above 15.6 U/ml increasingly attenuated DAN nitrosation, curves 7-11 o f Fig 42B.

The kinetics o f fluorescence formation was dramatically affected by the presence o f the

highest [Cu, Zn-SOD], at which the fluorescence intensity accumulated in a slow and

sustained fashion (curve 11, Fig 42B).

76

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. SOD modulated SIN-l-mediated DAN nitrosation in a biphasicfashion. To farther

characterize the interaction between SOD and DAN nitrosation, a dose-response

relationship was constructed in accordance w ith the relative degree o f nitrosation

compared w ith the control versus [CuyZn-SOD] at different measurement times.

Remarkably, m ultiple biphasic dose-response curves characterized by a dynamic

transition among those curves emerged (Fig. 43 A). Furthermore, these data enabled a 3-

D representation o f the dynamic dose-response relationship (Fig. 43B). Based on Fig.

43, the maximum stimulatory effect o f SOD occurred initially at [Cu^n-SOD] < 10

U/ml, which corresponded to a hump in this dose region at early incubation time.

Furthermore, the size o f the hump in this region diminished and then disappeared at -

180 min. A fter this mark, the dose-response curve in the low Cu,Zn-SOD concentration

region changed insignificantly along time and became the ascending part o f a new

emerging hump whose size grew continuously and the concentration o f CtiyZn-SOD

corresponding to the hump also tended to increase. In contrast w ith the low

concentration region, the dose-response relationship in the high concentration region (30

-1000 U/ml) appeared to be steady in the early period while changing substantially in

the later period. Notably, high [Cu,Zn-SOD] reduced N-nitrosation to the extent that

DAN nitrosation levels were generally below control levels.

DAN concentrations affected SOD modulation o f nitrosation kinetics . To

investigate whether the substrate concentration affected the observed dose-response

phenomena, the concentration o f DAN was halved or doubled while keeping other

experimental conditions unchanged. Fig. 4.4 depicts the sharp contrast between two

experiments that used 4-fold differences o f [DAN]. W hile the time-dependent, bell- 77

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. shaped, dose-response relationship remained in both cases, there was a fundamental

difference in the way in which N-nitrosation was affected by the presence o f Cu,Zn-SQD.

A t [DAN] = 1.56 pM, Fig. 4.4A, the hump in the higher SOD region dominated that in

the lower region (~ 600 % at [Cu,Zn-SOD] ~ 30 U/mi at 480 min versus ~ 450 % at

[Cu,Zn-SOD] ~ 3 U/ml at 45 mm). In addition, the dynamic transition between the two

regions in Fig. 4.4A occurred much earlier than that in Fig. 43. However, the result was

somewhat reversed when [DAN] was increased by 4-fold (Fig. 4.4B). The effect o f

Ct^Zn-SOD peaked at the in itia l period and then dropped more slowly until it achieved a

second phase in which the maximum effect was shifted to the right (Fig. 4.4B).

Moreover, the dose-response relationship at [CnZn-SOD] > 100 U/ml was less perturbed

during the reaction period (Fig. 4.4B). To account for this observation, the kinetic

profiles for the three controls w ith different levels o f DAN were compared (Fig. A AC).

The time to attain maximum nitrosation appeared to be increased from ~ 2 hr for the

control with the lowest [DAN] to ~ 3 hr and ~ 5 hr for the control with two- and four-fold

higher [DAN], respectively. The extent o f nitrosation increased almost linearly.

A ll SOD isoforms demonstrated biphasic reaction kinetics. A lso examined was

whether Mn- and Fe-SOD elicited a biphasic effect sim ilar to that o f Cu,Zn-SOD. A

sim ilar dose-response relationship occurred for these SOD isoforms as well (Fig. 4.5),

although a smaller inhibitory effect was observed for both high [Mn-SOD] and [Fe-

SOD]; the maximal effective concentration at t= 480 min was increased from 313 U/ml

for Cu,Zn-SOD (Fig. 43 A) to 62.5 U/ml for both Mn- and Fe-SOD (Figs. 4.5A and

4 3 B ).

78

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Functional SOD was required fo r the biphasic effect. Heat inactivated (> 90 °C , 2

hr) Cu,Zn-SOD lost both its stimulatory and inhibitory effects on N-nitrosation (Fig. 4.6).

Free metal ions such as C u\C u"” andZn^ also had no effect on N-nitrosation (Fig. 4.6).

Bovine serum albumin (BSA) was used to test whether a nonspecific protein effect might

have been involved in the reaction. W hile BSA had no effect on nitrosation at low

concentrations, it inhibited the reaction to some extent at concentrations > 1 pM (Fig.

4.6).

Glutathione modulated the biphasic effect o f Cu,Zn-SOD in a concentration-

dependentfashion. Glutathione (GSH), a reduced thiol and an antioxidant, is an “NO

sink” that modulates cellular redox reactions. It was therefore postulated that GSH would

modify the effect o f SOD on the NO-mediated N-nitrosation reaction. Experiments were

designed (i) to determine the effect o f GSH on SIN-1-mediated N-nitrosation in the

absence o f SOD and (if) to investigate the dose-response relationship o f Cu^n-SOD on

N-nitrosation in the presence o f three levels o f GSH. GSH at or above 0.5 mM inhibited

SIN-1-mediated N-nitrosation by > 90% (Fig. 4.7A). Accordingly, the effect o f Cti,7n-

SOD was examined in the presence o f 0.2 ,1 and 5 mM GSH, which represented low,

intermediate and high physiologically relevant levels. While the biphasic effect o f

CuZn-SOD was retained at 0.2 mM GSH, it disappeared with higher [GSH] (Fig. 4.7B).

When the data were expressed as % control and compared w ith the dose-response curve

obtained from the experiment in which GSH was absent, it was remarkable that the effect

o f Cu,Zn-SOD was enhanced by the presence o f increasing [GSH] (up to I mM), which

was reflected in the upward shift o f the entire dose-response curve (Fig. 4.7Q . However,

the stimulant effect o f GSH disappeared at 5 mM GSH. 79

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The effect o f Cu,Zn-SOD on SIN-I-mediated oxidation o f DHR was monophasic.

To investigate whether the biphasic effect o f SOD m ight be observed in other reactions

mediated by NO and 02“, SIN-1-mediated oxidation o f the fluorescence dye DHR 123

was studied in the presence o f various levels o f CiiZn-SOD. Cu^Zn-SOD inhibited the

SIN-1-mediated oxidation o f DHR and the IC 50 increased over time (Fig. 4.8). Unlike N-

nitrosanon, Cu^Zn-SOD did not stimulate SIN-I-mediated oxidation o f DHR.

Cu,Zn-SOD inhibited the release o f NO from SIN-L An HPLC method was used to

follow S IN -l as w ell as its degradation intermediate, SIN-1 A. and its stable end product,

SIN-IC, whose formation was indicative ofNO release from SIN -I. Inclusion o f 1000

U/m l CaZn-SOD significantly inhibited the formation o f SIN-IC and thereby the release

o f NO (Fig. 4.9A). This inhibitory effect required functional Cu,Zn-SOD, as indicated

from the heat-inactivation result (Fig. 4.9B). To construct a dose-response curve over the

same concentration range o f Cu^Zn-SOD studied in the previous experiments, the peak

intensities o f S IN -l, SIN-I A and SIN-IC were measured after 4 hr incubation o f SIN -I.

High [Cu^n-SOD] preserved SIN-L and inhibited the formation o f SIN-IC (Fig. 4.9Q.

However, Cu^n-SOD had no effect on the degradation o f SIN-I and the formation o f its

degradation products at the low concentration range, during which Cu^n-SOD

stimulated the N-nitrosation reaction.

4,4 Discussion

The biological importance o f SOD was attributed to its ability to catalyze the

dismutation reaction o f superoxide. SOD was implicated in many diseases, including

cardiac or cerebral ischemia and neurodegeneration.27 However, some studies11"13 80

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. indicated that the beneficial effects o f SOD at low concentration were reversed at high

concentration, which resulted in a bell-shaped dose-response curve. Moreover, SOD can

both potentiate28"30 and attenuate31'33 NO-mediated toxicity. SOD effectively removes

0?~, thereby preventing the O^'-mediated damage that could be sustained during normal

c e llu la r oxidative metabolism as w e ll as during inflam m ation, h i addition, the removal o f

O2' by SOD may diminish peroxynitrite (ONOO~), a deleterious pro-oxidative molecule

formed by the direct reaction ofNO and O 2". These findings suggested a far more

complex role for SOD in the cellular m ilieu than was first appreciated.

The present study used S IN -l as a donor o f both NO and Oz~, to mimic a scenario

in which both free radicals were coexistent and sustainable, as might occurred in

activated macrophages. The bell-shaped dose-response relationship that characterized the

effect o f SOD on N-nitrosation o f DAN was consistent among the three isoforms o f SOD

(CuZn, Mn, Fe), although there existed subtle differences. Low levels o f SOD enhanced

the N-nitrosation o f DAN, which seemed to agree with the role o f SOD; i.e., SOD

dismutated O f, thereby increasing the availability o f NO for the N-nitrosation reaction.

Nevertheless, increasing levels o f SOD did not further increase the N-nitrosation, but

rather significantly inhibited it This observation suggested that additional activities

operated at high levels o f SOD. Varying the DAN concentration resulted in qualitatively

and quantitatively distinct bell-shaped, dose-response curves, suggesting that second-

order or other complex kinetics might be involved in DAN nitrosation.

SOD affected both the rate and extent o f the N-nitrosation kinetics. As for C.n /n -

SOD, low levels (up to 7.8 U/ml) increased the build-up rate o f DAN nitrosation, which

was indicated by the increased steepness o f the in itia l slope along w ith a shortened time SI

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. interval to reach the steady-state (Fig. 4.2A). In addition, the extent o f DAN nitrosation

was enhanced by increasing [Cu,Zn-SOD] (Fig, 4.2A). Remarkably, [Cn^Zn-SOD] as

low as I U/m l was enough to cause more than a 2-fold enhancement, which correlated

w ith the O f scavenging activity o f Cu^Zn-SOD. A t I U/ml, Cu,Zn-SOD resulted in

—50% inhibition o f ferricytochrome c reduction in the xanthine/xanthine oxidase system

(data not shown). When [Cu,Zn-SOD] reached 15.6 U/m l (Fig. 4.2A, curve 6), the

kinetic profile o f DAN nitrosation started to deviate from those obtained at lower

concentrations. This was considered to be a transition point at which the effect o f SOD at

high levels was distinct from that at low levels. A t [CurZn-SOD] > 15.6 U/ml, the rate

and extent o f the DAN nitrosation was constantly reduced by increasing [Cu,Zn-SOD]

(Fig. 4.2B). Compared with the control (Fig. 4.2A, curve 1), the kinetic profiles in curves

7-11 featured a much slower build-up o f the fluorescence intensity, thereby resulting in a

non-steady-state, dose-response relationship in the high [Cu,Zn-SOD] region (Fig. 43).

The slow DAN nitrosation rate could be partly attributed to a slow NO release rate

imposed by high [Cu,Zn-SOD] (Fig. 4.9).

Can the impediment o f NO release by high [SOD] fu lly account for the descending

part o f the bell-shaped dose-response curves? Is [SOD] itse lf a target o f nitrosation or can

it act as an “ NO sink” via other reactions? These are important questions associated with

the findings. SOD is a class o f metalloprotein consisting o f three isoforms that vary in

their metal centers. NO interacts with copper ions in Cu-containing proteins, such as

Cu.Zn-SOD23 and cytochrome c oxidase;34 it also interacts w ith Fe-containing proteins,

such as guanylyl cyclase35 and hemoglobin.36 It is therefore likely that via a direct

interaction between NO and metal centers imbedded in SOD the availability ofNO for 82

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. DAN nitrosation is lim ited. Besides metal centers, the protein structure o f SOD may

provide other functional groups to react w ith NO, thus serving as an NO sink.36 High

[BSA] inhibited DAN nitrosation to some extent (Fig. 4.6). Since the concentration

range o f BSA to inhibit the nitrosation was sim ilar to that o f [Cu,Zn-SOD], I

investigated whether the protein nature o f SOD played a role. BSA is a target o f NO-

mediated S-nitrosation,37 which might allow BSA to compete w ith DAN for nitrosation.

Furthermore, the inhibitory effect o f BSA and Cu,Zn-SOD only occurred over the

concentration range (1- 10 pM), which was comparable with that o f DAN (3.13 pM ) and

supported the competition mechanism.

I f high levels o f SOD competed w ith DAN for nitrosation, it was possible that the

competition could be abrogated by another nitrosation target I chose GSH to test this

hypothesis, because (i) GSH is a target fo r the NO-mediated S-nitrosation reaction in

vivo;38 (ii) GSH is a crucial antioxidant that may function together w ith SOD in

diminishing or eliminating oxidative and nitrosative stresses;39 (iii) the biologically

relevant concentrations o f GSH range from 0.5 pM to 10 mM;40 and (iv) it is important to

know how the apparent biphasic effect o f SOD w ill be modulated by GSH in different

physiological conditions. GSH effectively abolished DAN nitrosation in the

physiologically relevant concentration range (Fig. 4.7). Moreover, the biphasic effect o f

Cu,Zn-SOD was modulated by GSH in a nonlinear fashion. A t low [GSH] (0.2 mM), the

bell-shaped dose-response relationship was retained; however, the maximum response

was increased from - 400% (without GSH) to ~ 800% (w ith GSH) and the concentration

o f Cu^nrSOD to reach the maximum response was also increased about 10-fold (Fig.

4.7Q . When [GSH] was elevated to I mM, the maximum response was boosted by— 83

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1500% and the bell-shaped character o f the profile disappeared (Fig. 4.7Q . These

findings suggested that GSH might attenuate the apparent inhibitory effect o f CuJZn-SOD

on DAN nitrosation, thus revealing the stimulatory effect o f Cu^Zn-SOD. hi other words,

excess GSH might mask the reaction between SOD and NO by acting as an NO sink.

Surprisingly, the whole dose-response curve o f Cu^Zn-SOD fe ll dramatically at 5 mM

GSH (Fig. 4.7Q . This result may be explained by the fact that the maximum inhibitory

effect o f GSH on DAN nitrosation was achieved at [GSH] ~ I mM (Fig. 4.7A). Above

this level, the detection lim it might be approached; therefore, no further inhibition was

observable for the baseline. In this case, normalization may have resulted in an

underestimation o f the actual effect o f GSH on the Cu^n-SOD-mediated response.

Nevertheless, the data were consistent w ith the hypothesis that excess GSH may attenuate

interaction between SOD and NO. This result also identified a potential mechanism by

which SOD at high levels competed with DAN for nitrosation, and thereby reduced DAN

nitrosation.

A ll three SOD isoforms elicited the biphasic effect on the nitrosation reaction. It

was conceivable that the bell-shaped dose-response curve was a manifestation o f certain

intrinsic characteristics o f these enzymes. For example, the stimulatory effect o f SOD on

the nitrosation could be attributed to the Oi'-scavenging effect o f SOD.8,9 W hile the

nitrosation o f DAN is suppressed by the presence o f O f,8 later results9 demonstrated that

increasing concentrations o f O f dramatically reduced the nitrosation o f both DAN and

GSH in a buffer system in which DEA/NO was used as an NO donor and O f was

generated by the xanthine/xanthine oxidase reaction. The inhibition o f nitrosation by O f

was reversed by SOD.9 In the present study, SIN -I was used as both an NO and an O f 84

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. donor, because S IN -l releases NO and 02” at a 1:1 ratio.26,41 fa the presence o f SOD, the

NO/Q?~ molar ratio may be increased due to the elimination o f 02” by SOD. Excess NO

may undergo autooxidation in the presence o f molecular oxygen to farm nitrogen oxides

that are potent nitrosatfag agents.42 Therefore, the result was in concert w ith the previous

findings fa the sense that SOD stimulated N-nitrosation via elimination o f O 2'.

The earlier study did not report the biphasic effect o f SOD on N-nitrosation,9 and it

is unclear whether different NO and O 2" donor systems led to this discrepancy. The

systems differ fa their rates o f generation o f these species, w ith generation ofN O from

DEA/NO and from xanthine/xanthine oxidase being much faster than their generation

from S IN -l. For example, SIN-l generates NO and O 2'w ith a rate constant o f 0.0017

m in1 (current study) whereas the rate constant for the formation o f NO from DEA/NO9

was 0.3 m in'1. Moreover, the stoichiometry o f SOD and DAN may have contributed to

the contradictory results. The concentration o f DAN used fa the previous study was 200

pM, which was much higher than that o f SOD. fa contrast, the concentrations o f DAN

used fa this study (1.56 - 6.25 pM) were comparable to the high concentration range o f

SOD (0.5 —10 pM). Accordingly, if SOD can act as an NO sink, excess DAN may

produce a masking effect sim ilar to that o f high [GSH] as discussed above. I did not test

this conjecture because from the method validation I found that SIN-1-mediated DAN

nitrosation may undergo self-quenching at DAN concentrations above 25 pM . fa fact,

the entire experiment was designed to avoid possible fluorescence quenching caused by

high dye concentrations.

Distinctions among the three SOD isoforms were o f interest, especially since Mn-

and Fe-SOD, as compared w ith Cu,Zn-SOD, exerted a smaller inhibitory effect on 85

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. nitrosation at high concentrations (Fig. 4.5). Based on the kinetic profiles o f DAN

nitrosation in the presence o f 1000 U/ml SOD (Fig. 4.10), Mn- and Fe-SOD did not

produce either the stimulatory or inhibitory effect during the early period up to 150 min.

However, unlike the control, nitrosation in the presence o f Mn- and Fe-SOD did not level

o ff after 150 min. This could be interpreted by a mechanism in which the stimulatory

effect o f SOD was counteracted by an inhibitory effect at high concentrations. It was

apparent that Cu,Zn-SOD was more potent in terms o f its inhibitory action. While the

present study did not definitively answer the question as to how the differences among

the three SOD isoforms emerged, possibility that the difference resulted from differences

in Os'-scavengmg activity (data not shown) was eliminated.

A schematic mechanism was proposed to delineate the biphasic effect o f SOD on

SIN-1-mediated nitrosation (Fig. 4.11). SIN-1 releases NO and O?- and the two free

radicals react to form ONOO- at a nearly diffusion-lim ited rate .5 Since the second-order

rate constant fo r SOD's dism utation o f Qz~ is about 1/3 o f that o fN O / O 2- reaction ,5 the

finding that low levels o f SOD (< 10 nM) were sufficient to stimulate the nitrosation

reaction is perplexing. However, it suggested that mechanisms in addition to O f

scavenging may promote the pro-nitrosative effect o f SOD. It has been shown that the

nitrosation reaction is enhanced w ithin protein hydrophobic moieties .37 Therefore, SOD

may entrap and concentrate nitrosating species (e.g. N 2O3) as well as DAN, consequently

accelerating nitrosation kinetics, hi addition. SOD itse lf may be nitrosated through

autocatalyzation .57 As SOD concentrations increased. I speculated that SOD acts as a

nitrosating species sink, which competes w ith DAN for reaction w ith the nitrosating

86

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. species, N 2O3- High levels o f GSH would modify these interactions, since GSH has high

affinity towards N 2Q1. My data, which showed that GSH effectively quenched DAN

baseline GSH nitrosation and intensified SOD's pro-nitrosative effects, supported this

premise. Furthermore, given that NO interacts with copper ions in Cu-containing

proteins, such as Cu^Zn-SOD (23) and cytochrome c oxidase (34), CuZnSOD may also

lim it the NO availability via a direct interaction between its metal center and NO. My

data also support this concept Finally, NO availability can also be reduced by SOD’s

inhibitory effect on the kinetics o f NO release. However, this pathway cannot fully

explain the inhibitory effect o f SOD on N-nitrosation as indicated in Fig. 4.9. Overall,

the net effect o f SOD on nitrosation is determined by the counterbalance o f SOD’s

abilities to both promote and quench reactive species chemistry.

SOD elicits multiple (^'-independent actions that are related to NO: (t) CtiyZn-

SOD can reversibly convert nitroxy anion (NO") to NO;23 (ii) SOD can enhance free NO

formation from L-arginine by NO synthase;24 (in ) SOD catalyzes the decomposition o f S-

nitrosoglutathione, resulting in the sustained production o f NO;43 and (iv) peroxynitrite-

mediated nitration is catalyzed by SOD.44 Moreover, recent in v itro cell culture

studies have implicated a potential role for SOD in many biological functions linked to

NO. For example, the Cu,Zn-SOD gene in keratinocytes can be regulated by NO.45

(XZn-SOD was associated w ith NO-mediated apoptotfc cell death.46"48 Altogether, these

findings suggested that NO-mediated activities may be highly regulated by SOD or vice

versa. It was therefore conceivable that temporal and spatial distribution o f SOD in the

cellular m ilieu may play a significant role m NO-mediated reactions. The presence o f

biomolecules that are capable o f interrupting direct or indirect NO-SOD interactions 87

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. would further increase the complexity o f this system. That GSH can modulate the

biphasic effect o f SOD on N-nitrosation supported this view. Recently, nitros(yl)ation

has been considered as a prototypic redox-based signaling mechanism;49 proteins

involved in the signal transduction pathway can be regulated by NO via the modification

o f cysteine thiols and transition metal centers.49 Hence, SOD may modulate the dynamic

regulation o f NO-related posttranslational modifications o f proteins.

4 i Conclusion

This study pinpointed some intriguing aspects o f the action o f SOD on the NO-

mediated nitrosation reaction. SOD showed a concentration-dependent biphasic effect on

nitrosation. Its effects were sustained and underwent dynamic transformations. Finally,

SOD itse lf may serve as a substrate for the nitrosation reactions, depending on the

relative abundance o f other biomolecules. Overall, the kinetics were made complex

because more than one function o f SOD was involved, which may account for the bell­

shaped, dose-response relationship. This study provided insights into the unsettled nature

o fN O and O 2' in the cellular m ilieu, where the distribution and proportions o f reactants

may affect NO-mediated nitrosation in a nonlinearfashion.

4.6 Summary

N itric oxide (NO) and superoxide (O f) are two important radicals involved in

many physiological and pathological functions. SOD’s removal o f superoxide may

reduce the prochiction o f the deleterious molecule, peroxynitrite (ONOOr). While low

levels o f SOD are anti-oxidative, high levels o f SOD are pro-oxidative. However, the 88

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. mechanism underlying this idiosyncratic bell-shaped dose-activity relationship o f SOD

remains elusive. 3-Morpholinosydnonimine (S IN -l) was used to generate NO and Oz~

simultaneously in PBS buffer at pH 7.4. Diaminonaphthalene (DAN) and

dihydrorhodamine 123 (DHR) were used as molecular probes fo r assessment o f the

nitrosation and oxidation reactions, respectively. The effect o f SOD on SIN-1-mediated

nitrosation o f DAN and oxidation o f DHR was determined by kinetic measurements o f

the formation o f the florescent products o f DAN and DHR. SOD's effect on NO release

was studied by HPLC. The results showed that: (i) the biphasic effect o f Cu,Zn-SOD in

the nitrosation o f DAN mediated by SIN -l is time- and substrate concentration-

dependent; (ii) Mn-SOD and Fe-SOD exhibited sim ilar biphasic dose-response

phenomena; (iii) the biphasic effect o f SOD required functional enzyme; (iv) notably, the

effect o f Cu,Zn-SOD on S IN -l- mediated oxidation o f DHR was monophasic; (v) high

[CuZnSOD] inhibited the NO release rate; (vi) reduced glutathione (GSH) modulated the

biphasic effect o f SOD. These findings suggest that SOD’s superoxide scavenging effect

prevents NO from reacting with superoxide and thus increases the availability ofN O for

the nitrosation reaction. Moreover, SOD may also serve as an NO sink, thereby reducing

the N-nitrosation w ithin the physiological concentration range. Overall, complex kinetics

involving more than one function o f SOD may account for the observed bell-shaped

dose-response relationship. This study provides insights into the unsettled nature o f NO

and Oz~ in the cellular m ilieu where, the distribution and proportions o f reactants may

affect the outcome NO-mediated reactions in a nonlinear fashion.

89

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Acknowledgments

This work was supported by NIH/NIDCR P01 DE 12704, P30CA16058. The

majority o f this work was conducted in Dr. Mallery’s lab.

4.7 References

1. Masters BS. Structural variations to accommodate functional themes o f the isoforms o f NO synthases, hi: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000:91-104.

2. Szabo C. Pathophysiological roles o f nitric oxide in inflammation. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000:841-872.

3. Ganster RW, Geller DA. Molecular regulation o f inducible nitric oxide synthase. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st EcL, San Diego: Academic Press, 2000:129-156.

4. Grisham MB, Jourd'heuil D, Wink DA. N itric oxide. I. Physiological chemistry o f nitric oxide and its metaboIites:impiications in inflammation. Am. J. Physiol. 1999;276:G315-321.

5. Miranda KM, Espey MG, Jourd’heuil DJ, et al. The chemical biology o f nitric oxide, ha: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st EcL, San Diego: Academic Press, San Diego, 2000:41-55.

6. Patel RP, McAndrew J, SeDak H, et al. Biological aspects o f reactive nitrogen species. Biochim. Biophys. Acta. 1999:1411:385-400.

7. Miles AM, Scott Bohle D, Glassbrenner PA, et al. Modulation o f superoxide- dependent oxidation and hydroxylation reactions by nitric oxide. J. Biol. Chem. 1996;271:40-47.

8. Miles AM, Gibson MF, Kirshna M, Cook JC, et al. Effects o f superoxide on nitric oxide-dependent N-nitrosation reactions. Free Rad. Res. 1995;23:379-390.

90

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 9. Wink DA, Cook JA, Kim SY, et al. Superoxide modulates the oxidation and nitrosation o f thiols by nitric oxide-derived reactive intermediates. J. Biol. Chem. 1997;272:11147-11151.

10. Weber GF, Bruch HP. Pharmacology o f snperoxide-dismutase. Pharmazie. 1992;47:159-167.

11. Bernier M, Maiming AS, Hearse DJ. Reperfusion arrhythmias: dose-related protection by anti-free radical interventions. Am. J. Physiol. 1989^56:1344-1352.

12. Omar BA, McCord JM. The cardioprotective effect o f Mn-superoxide disinutase is lost at high doses in the postxschemic isolated rabbit heart. Free Rad. B iol. Med. 1990;9:473-478.

13. Omar BA, Gad NM, Jordan MC, et al. Cardioprotection by CtCZn-superoxide dismutase is lost at high doses in the reoxygenated heart Free Rad. BioL Med. 1990;9:465-471.

14. Mao GD, Thomas PD, Lopaschuk GD, Poznansky MJ. Superoxide dismutase (SOD)-catalase conjugates. J. BioL Chem. 1993;268:416-420.

15. Norris BCH, Hornsby P. Cytotoxic effects o f expression o f human superoxide dismutase in bovine adrenocortical cells. M utat Res. 1990;237:95-106.

16. Ishii T, Iwahashi H, Sugata R, Kido R. Superoxide dismutase enhances the toxicity o f 3-hydroxyanthranilic acid to bacteria. Free Rad. Res. Commun. 1991; 14:187- 194.

17. Scott MD, Meshnick SR, Eaton JW. Superoxide dismutase-rich bacteria. Paradoxical increase in oxidant toxicity. J. BioL Chem. 1987;262:3640-3645.

18. Kedziora J, Bartosz G. Down’s syndrome: a pathology involving the lack o f balance o f reactive oxygen species. Free Rad. BioL Med. 1988;4:317-330.

19. Yim MB, Chock PB, Stadtman ER. Copper, zinc superoxide dismutase catalyzed hydroxy radical production from hydrogen peroxide. Proc. Natl. Acad. Sci. USA I990;87:5006-5010.

91

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 20. McCord JM. The importance o f oxidant-antioxidant balance, hr Montagnier L, O livier R. PasquierC. eds. Oxidative Stress in Cancer, AIDS, and Neurodegenerative Diseases, New York: Marcel Dekker, 1998; I-7.

21. Offer T, Rosso A, Samuni A. The pro-oxidative activity o f SOD and nitroxide SOD mimics. Faseb J. 2000;14:1215-1223.

22. Liochev SI, Fridovich L Copper- and zinc-containing superoxide dismutase can act as a superoxide reductase and a superoxide oxidase. J. BioL Chem. 2000;275:38482-38485.

23. Murphy ME, Sies H. Reversible conversion o f nitroxyl anion to nitric oxide by superoxide dismutase. Proc. Natl. Acad. Sci. USA 1991;88:10860-10864.

24. Hobbs AJ, Fukuto JM, Ignarro LJ. Formation o f free nitric oxide from L-arginine by nitric oxide synthase: Direct enhancement o f generation by superoxide dismutase. Proc. NatL Acad. ScL USA I994;9l: I0992-I0996.

25. Mallery SR, Landwehr DJ, Ness GM, Clark YM, Hohl CM. Thiol redox modulation o f tumor necrosis factor-a responsiveness in cultured AIDS-reiated Kaposi’s sarcoma cells. J. Cell. Biochem. 1998;68:339-354.

26. Feelisch M, Ostrowski J, Noack EJ. On the mechanism o f NO release from sydnonimines. Cardiovasc. Pharmacol. !989;14(SuppL 11):S13-S22.

27. Beckman JS, Chen J, Crow JP, Ye YZ. Reactions o f nitric oxide, superoxide and peroxynitrite w ith superoxide dismutase in neurodegenration. Prog. Brain. Res. 1994;103:371-380.

28. Oury TD, Ho Y-S, Piantadosi CA, Crapo JD. Extracellular superoxide dismutase, nitric oxide, and central nervous system 02 toxicity. Proc. NatL Acad. ScL USA 1992;89:9715-9719.

29. Assreuy T, Cunha FQ, Epperfein M, Noronha-Dutra A , O’Donnell CA, Liew FY, Moncada S. Production o f nitric oxide and superoxide by activated macrophages and killin g o f Leishmania major. Eur. J. Immunol. 1994;24:672-676.

92

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 30. Gergel D, M isik V, Ondrias BC, Cederbaum AL Increased cytotoxicity o f 3- Morpholinosydnonimine to HepG2 cells in the presence o f superoxide dismutase. J. BioL Chem. 1995;270:20922-20929.

31. Bruneili L, Crow JP, Beckman JS. The comparative toxicity o f nitric oxide and peroxynitrite to Escherichia coii. Arch. Biochem. Biophys. 1995;316:327-334.

32. Lipton SA., Choi Y-B, Pan Z-H, Lei SZ, Chen H-SV, Sucher NJ, Loscalso J, Singe! DJ, Stamler JS. A redox-based mechanism for the neuroprotective and neurodestructive effects o f nitric oxide and related nitroso-compounds. Nature 1993;364:626-632.

33. Siegfried MR, Erhardt J, Rider T, Ma X-L, Lefer AM. Cardioprotection and attenuation o f endothelial dysfunction by organic nitric oxide donors in myocardial ischemia-reperfusion. J. Pharmacol. Exp. Ther. 1992;260:668-675.

34. Cooper CE. N itric oxide and cytochrome oxidase: Substrate, inhibitor or effector? Trends Biochem. ScL 2002;27:33-39.

35. Koesling D, Friebe A. Structure-function relationships in NO-sensitive guanytyt cyclase. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000^69-379.

36. Lancaster Jr. JR. The physical properties o f nitric oxide: determinants o f the dynamics ofNO in tissue. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000^209-224.

37. Nedospasov A, Rafikov R, Beda N, Nudler E. An autocatalytic mechanism o f protein nitrosylation. Proc. Natl. Acad. ScL USA 2000;97:13543-13548.

38. Gaston B. N itric oxide and thiol groups. Biochhn. Biophys. Acta I999;I411:323- 333.

39. Calabrese V, Bates TE, Stella AMG. NO synthase and NO-dependent signal pathways in brain aging and neurodegenerative disorders: The role o f oxidant/antioxidant balance. Neurochem. Res. 2000;25:1315-1341.

93

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 40. G riffith OW. Biologic and pharmacologic regulation o f mammalian glutathione synthesis. Free Rad. B iol. Med. 1999;27:922-935.

41. Bohn H, Schonafinger BC. Oxygen and oxidation promote the release o f nitric oxide from sydnonimines. J. Cardiovasc. Pharmacol. !989;l4(SuppI. 11): S6-SI2.

42. Marietta MA, Mammalian synthesis o f nitrite, nitrate, nitric oxide, and N- nitrosating agents. Chem. Res. Toxicol. 1988;1:249-257.

43. Jourd’heuil D, Stephen Laroux F, Miles AM, Wink DA, Grisham MB. Effect o f superoxide dismutase on the stability o f S-nitrosothiols. Arch. Biochem. Biophys. I999;36l;323-330.

44. Ischiropoulos H, Zhu L, Chen J, Tsai M, Martin JC, Smith CD, Bechman JS. Peroxynitrite-mediated tyrosine nitration catalyzed by superoxide dismutase. Arch. Biochem. Biophys. 1992;298:431-437.

45. Frank S, Kampfer H, Podda M, Kaufinann R, Pfeilschifter J. Identification o f copper/zinc superoxide dismutase as a nitric oxide-regulated gene in human (HaCaT) keratinocytes: Implications for keratinocyte proliferation. Biochem. J. 2000;346:719-728.

46. Troy CM, Derossi D, Prochiantz A, Greene LA, Shelanski ML, Downregulation o f Cu/Zn superoxide dismutase leads to cell death via the nitric oxide-peroxynitrite pathway. J. Nerurosci. 1996;16:253-261.

47. Morrison BM, Morrison JH, Amyotrophic lateral sclerosis associated with mutations in superoxide dismutase: a putative mechanism o f degeneration. Brain Res. Rev. 1999;29:121-135.

48. Ciriolo MR, De Martino A, Lafavia E, Rossi L» Carri MT, Rotilio G. Cn,Zn- superoxide dismutase-dependent apoptosis induced by nitric oxide in neuronal cells. J. BioL Chem. 2000;275:5065-5072.

49. Stamler JS, Lamas S, Fang FC. Nitrosylation: The prototypic redox-based signaling mechanism. CeO, 2001;106:675-683.

94

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. -14— 0 3-Morpholinosydnonimine (SIN-l)

NH2

n h 2 2,3-Diaminonaphthalene (DAN)

NH

Dihydrorhodamine 123 (DHR)

Figure 4.1. The chemical structures of 3-morphoIinosydnonimine (S IN -l), 23- diaminonaphthalene (DAN) and dihydrorhodamine 123 (D H R ).

95

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A 5 0 0

400 6 5 300 4 3 7 200 E c e too 5? ✓ Q 0 0 too 200 300 400 500 >* •* c £c B 500 1 400 7 8 s 9 o 300

200 10

too tt

0 100 200 300 400 500

Time (min)

Figure 4.2. Nitrosation kinetics o f DAN in the absence and presence o f different levels o f CiijZn-SOD. The reactions were performed in a 96-well microtiter plate form at SIN -l (100 pM) was added to PBS-based reaction buffer containing 3.13 pM DAN, 0.1 mM DTP A, 120 U/ml catalase and CitZn-SOD (0 - 1000 U/ml>, pH 7.4, at 25 ± I°C . The fluorescence intensity after the addition o f SIN -l was measured at 10-min intervals by fluorescence spectroscopy. Data represent the mean o f three measurements. Each curve represents a different [Cu,Zn-SOD]: I, control; 2,0.98 U/ml; 3,1.95 U/ml; 4, 3.9 U/ml; 5,7.8 U/ml; 6,15.6 U/ml; 7 ,31J2 U/ml; 8,62.5 U/ml; 9,125 U/ml; 10,500 U/ml; 11,1000 U/mL

96

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A 5 0 0 o

1 10 100 1000 Cu.Zh-SOD (U/ml)

B

Figure 43. Time- and concentration-dependent, biphasic, dose-response relationship fo r the effect o f Cu,Zn-SOD on DAN nitrosation. A. The fluorescence intensities at the same time point in FIG. 42. were expressed as % control and plotted against [Cu,Zn-SOD], Each curve (n = 3} represents the dose-response relationship at 20-mm intervals. The arrows indicate the direction o ftone progression starting at 45 min and ending a t485 min. B. Saddle-like dose-response surface was generated by plotting the relative fluorescence intensities against [Ct^Zn-SOD] and the reaction time.

97

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A

C 400

10 100 100 0 Cu,Zh-SOD (U/ml) B

~ 500

C 1 00

10 100 100 0 Cu,Zh-SOD (U/ml)

f 250 • - [D A N ] - 1 a |lM 200 -■>- [O M f-X TlllM .*■ * 150 .iT

100 200 300 Tim* On In)

Figure 4.4. Effect o f DAN concentration on SOD modulation o f nitrosatiott kinetics. Procedures were as described in FIG. 4.2, except (A ) Vz x [DAN] or (B) 2 x [DAN] was used. Mean dose-response curves (n = 4 ) were obtained by plotting the relative fluorescence intensities (% control) against [Cu^n-SOD]. The time interval for each curve is 20 min. The arrows indicate the direction o f time progression starting at 45 min and ending at 485 min. (Q The kinetics profiles for the three controls with different levels o f DAN . 98

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A

500 s c o 400 3? c 300 o 200

c 100 z < Q 1 10 100 1000 Mn-SOD (U/ml) B

500 2 c o 400 S! 300

200 e z 100 < Q 1 10 100 1000 Fe-SOD (U/ml)

Figure 4.5. Time- and concentration-dependent, biphasic, dose-response relationships fo r the effects o f Mn-SOD and Fe-SOD. Procedures were described in FIG . 42. and FIG. 43, except (A ) Mn-SOD and (B) Fe-SOD were studied. Reactions were initiated by adding S IN -l (100 |iM ) to PBS-based reaction buffer containing 3.13 pM DAN, 0.1 mM DTPA, 120 U/ml catalase and various activities o f SOD (0 —1000 U/m l). Values represent the mean o f three measurements.

99

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 400 0 350 1 300 o ^ 250 c o 3 200 CO (A 2 150

z < o

0.01 0.1 1 10 Concentration ftiM)

Figure 4.6. Effect o f intact Cu^Zn-SOD (•), heat-inactivated Cu,Zn-SOD (O), BSA (■), and metal ions (C u^ A , Cu+ V , ZnM" O ) on DAN nitrosation. The experiment followed the typical nitrosation kinetics procedures for Cu,Zn-SOD, accept Cn,Zn-SOD was replaced by heat-inactivated enzyme, BSA, Q 1SO4, CuCI or ZnCh- Equal molar concentration range (0.01 -1 0 pM) for all species was used. Dose-response curves were compared at 4 hr.

100

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. i n 140 i 120 i n n

40

o 2 4 S S 12 GSH (mM) B

10 333 i n 333 io n Cu£h-SOD(Uftnl)

1300 _ te n >aii 1 u n I 1200 f 1000 I m 1 s n

2n

i 10 i n io n Cu.2n-SOO(U/ml)

Figure 4.7. Effect o f glutathione (GSH) on Ci^Zn-SOD modulation o f DAN nitrosation. (A ) SIN-l (100 pM ) was added to PBS-based reaction buffer pH 7.4 containing 3.13 pM DAN. 0.1 mM DTPA, 120 U/ml catalase and GSH (0 - 10 mM). at 25± i°C . The fluorescence intensity after the addition o f SIN-1 was measured at 4 hr and was plotted against [GSH]. (B) A t three fixed GSH levels (0.2 mM, I mM, 5 mM), DAN nitrosation was studied in the absence and the presence o f 10,333,100,333 and 1000 U/ml QivZiv-SOD. Fluorescence intensity at 4 hr fo r each group was plotted against [Cu^n-SOD]. Values represent the mean ± SD (n =4). (C) The data in B were expressed as % controL The dose-response curve at t = 4 hr for Cu^n-SOD in the absence o f GSH was included fo r the purpose o f comparison.

101

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. o 120

Cu, Zn-SOD (U/ml)

Figure 4.8. Effect o f Cu^Zn-SOD on SIN -l mediated oxidatioii o f DHR. SIN-I (100 p M ) was added to pH 7.4 PBS b u ffe r containing 0 3 9 pM DHR. 0.1 mM DTPA. 120 U/ml catalase andCu,Zn-SOD (0 -1000 U/mI)», at 25 ± I°C . The fluorescence intensity after the addition o f S IN -l was measured at a 5-min interval; fluorescence was expressed as % control (n = 4 ± SD) and plotted at a 20-min intervals against [CuyZn-SOD]. The arrows indicate the direction o f time progression starting at 25 min and ending at 245 m in.

102

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A 3 .0 C o n tro l 2.S -O- - Co. Zn-SOD 1000 U/ml

5 2 .0

1.0

0 .5

0 too 200 3 00 4 0 0 500 T lm o (m in ) B 2 .0

Control CuZivSOD H.L- C tO n-S O O C 200

150

2 too

1 10 too 1000 Cu2n4QO (U/ml)

Figure 4.9. Effect o f Cu,Zn-SOD on NO release kinetics o f S IN -l measured by HPLC. SIN -i (100 |lM ) was added to a pH 7.4 PBS buffer containing 0.1 mM DTPA and 120 U/ml catalase, at 25 ± I°C. SIN -l and its degradation products (SIN-1A and SIN-IC) were determined by HPLC. (A ) SIN-IC formation in the absence and the presence o f 1000 U/ml CuyZn-SOD. (B) SIN-IC formation at 4 hr in the absence or presence o f either intact Cn,Zn-SOD (1000 U/m l) or heat-inactivated (ELL) Ca1Zn-SOD. (Q CuZn-SOD (I —1000 U/ml) was included; the solutions were incubated for 4 brand the peak intensity o f SIN -l and its degradation products were determined. Mean data were presented as %control, n = 3 ± SD.

103

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Control O Cu, Zn-SOD (1000 U/ml) A Mn-SOD (1000 U/ml) □ Fe-SOD (1000 U/ml) R 250

200 300 500 Tim e (m in )

Figure 4.10. Comparison o f the nitrosation kinetics among three SOD isozymes at 1000 U/mL SIN-1 (100 pM ) was added to the PBS pH 7.4 bufier containing 3.13 pM DAN, 0.1 mM DTP A, 120 U/ml catalase and SOD (1000 U/ml), at 25 ± l°C . The fluorescence intensity was measured at 10-min intervals; values represent the mean ± SD, n = 3.

104

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. SCO GSH

DAK

SOD

SIN-l - GSNO GSH

SOD

Figure 4.11. Proposed mechanisms fo r SOD’s biphasic effect on SIN-l-mediated nitrosation reaction

105

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CHAPTERS

Kinetic Modeling of Nitric-Oxide-Associated Reaction Network:

Biological Implications

5.1 Introduction

The free radical nitric oxide (NO) mediates a large number o f physiological and

pathophysiological processes. Despite more than a decade o f intense research, many

aspects o f NO physiological chemistry remain both paradoxical and controversial. A t

low concentrations (~ nM), NO modulates normal physiological functions such as

regulation o f vascular tone and intracellular signalingvia a direct interaction w ith its

targets.1 However, high NO levels can induce cytotoxicity, presumably attributable to

oxidative and nitrosative stresses.1 Elevated NO levels present at inflammatorysites

provide the opportunity for NO to compete w ith superoxide dismutase (SOD) for

reactions with superoxide (O O , thereby generating the highly reactive and cytotoxic

peroxynitrite (ONOO~) molecule.2 The findings in the previous chapter highlighted the

complexity o f NO-mediated chemical reactions.

Because o f the complex nature ofNO and technical difficulties in studying its

biological features, computational modeling has been used as an alternative tool fo r

understanding diverse aspects ofN O , which included (i) the biotransport o f NO;3"7 (if)

106

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. kinetics related to the generation and reaction ofNO;®"15 (iii) functions ofN O in

physiological and pathophysiological processes, such as neural signaling16 and wound

healing17 (iv) mechanistic role ofNO on the activation o f soluble guanyiyl cyclase.18

For example, mathematical modeling ofN O diffusion predicted that NO is not a locally

acting mediator, due to its rapid and wide spread d iffiisib ility. Thus, one NO-producing

cell can affect many hundred o f its neighboring cells.19 Modeling approaches were also

applied to examine the processes in the interaction o f ONOO" with Iow-density

lipoproteins (LDLs) in the plasma.11*13 The kinetic model predicted that plasma

\ONOO~J should be in nM range and ONOO" is a potential candidate for initiating

peroxidation o f LDLs.11 Once ONOO~ is formed, antioxidants have little effect on the

ONOO" level.13 Furthermore, a reaction/diffusion model was used to explore the

movement o f ONOO' into the LDL particle.12

The coexistence o fN O and O 2" in the physiological m ilieu gives rise to intricate

oxidative and nitrosative reactions. As described in Chapter 4, low \SOD\ stimulated,

whereas high [SOD] attenuated, NO-mediated nitrosation reaction. Glutathione (GSH)

modulated the biphasic effect o f SOD. The results, therefore, suggested that the

consequences o f oxidative and nitrosative chemistry may depend heavily on a delicate

balance among the processes that govern the formation and elimination ofN O and O f.

The formation ofNO in the tissue is catalyzed by nitric oxide synthases (NOS),

which are heme-containing enzymes. The expression o f the different NOS isoforms is

regulated by diverse mechanisms.20 W hile NOS I [NOS 1, neuronal NOS (nNOS)] and

NOS3 [NOS in , endothelial NOS (eNOS)] are low-output, constitutive enzymes whose

107

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. activities are regulated by Ca2+ and calmodulin, NOS2 (NOS H, inducible NOS (iNOS))

is a high-output enzyme, which after induction can produce a large amount ofN O in a

Ca2+-independent fashion.20 Apparently, the amount ofN O produced in tissues is a

dynamic process that depends on which enzyme is involved, and where and when the

enzyme is expressed.

La tissues, NO has an extremely short half-life, on the order o f seconds.21 In

addition to the high mutual reactivity between NO and OT,22 NO also undergoes

autooxidation w ith a rate that is dependent on the concentration o f oxygen.21 Thus, both

Q» and O 2" play an important role in the rapid scavenging ofNO . The respective

reaction intermediates, N 2O3 and ONOO", fo r NO-O 2 and N O -O f reactions were

attributed to the indirect cytotoxic actions ofN O .1 While N 2O3 was believed to be a

strong nitrosating agent towards targets, such as DNA and proteins, ONOO- is a potent

oxidant and has been shown to react w ith a large number o f biomolecules in vitro. The

oxidative activity o f ONOO- contributes substantially to tissue damage in inflammatory

and infective diseases.1

hi this study, I integrated key reaction pathways associated with NO-mediated

nitrosative and oxidative chemistry into a reaction network. The dynamics o f this

network was investigated. Two specific questions were examined: (i) what is the role o f

GSH in modulating nitrosative and oxidative species, such as N 2O3 and ONOO"? and (ii)

how does the system behave when the generation rate ofN O and O 2- varies?

108

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 5.2 Model

Fig. 1 integrated the reaction pathways related to NO and its reaction products,

N2O3, ONOO" and nitroso glutathione (GSNO). The concentrations o f NO, N 2O3,

ONOO- and GSNO as w ell as the free radical 0z~ and antioxidant GSH were simulated.

The general procedure was firs t to write a set o f coupled differential equations based on

the law o f mass action. The differential equations were then solved simultaneously by

numerical methods. Since the reaction kinetics o f the integrative system was the main

interest o f this study, transport and diffusion processes were not included in the model.

5.2.1 Reaction chemistry and rate constants

The NO production rates were estimated to be in the range from I x I O'10 to 1.6

xiO** M/s fo r adherent cells expressing iNOS.ts Using mathematical modeling, Vaughn

et al.14 predicted the rate o f NO production by vascular endothelium o f ~ I0 's M/s. In

addition, the NO production rate by basal epidermal cell was estimated to be 1.7 x 10~7

M/s.23 Cultured keratinocyte cells24 and fibroblasts25 generated Q f at a rate o f ~ I O'6 M/s

and - I O'5 M/s, respectively. The rate constants were summarized in Table I.26"36 The

rate equations for NO, O 2-, ONOO- GSNO, N2O3, GSNO and GSH were:

^ Q = k l +kia[GSNO]1[o;]-/ct[NO][o;]-ict,[m }2[o,] (I) at

= kz -^[VO ][C>;]-Ar5[O;][5OO]-^I0[G SVO l2[O 2-l (2)

d[ONOCT] = kt[NO][O;}-kflON0Gr][GSH]-(kT[GPX] dt (3) +k*[COz]+ k9[cyt cWONOOr]

109

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ^^^=k,[0N 0(T\[G S H \+klS.NM[GSH\-kw[GSN0f[0-1[ (4) at

^ ^ = k aim nO t}-k„lN,0,\lGSH\-kJ_N,p,} (5)

^ p . = k , -(*l[0W00-][CSHl+t„[iV!0J][C O T ])+ -^^P - (6) at Km +[G5oC/|

[GSSG]= [G S H \ - [G S H ] - [GSNO] (7)

S J tl Numerical simulations

Numerical simulations o f the model (equations 1 -7 ) were carried out using

NDSolve in Mathematica 4.0 [W olfram Research, Champagne, EL]. NDSolve can solve

s tiff differential equations using Backward Differentiation Formulas (or Gear Formulas).

The approximation error in NDSolve is controlled by two built-in functions o f

Mathematica, AccuracyGoal and PrecisionGoal, which specify absolute and relative error,

respectively. NDSolve attempts to calculate a solution, y(x) , w ith error less than

10'“ +|y(x)[ I0~p, where a and p are positive integers that represent the settings for

AccuracyGoal and PrecisionGoal, respectively.37 For this study, the error was set to no

more than I O'15.

The model consisted o f 7 dependent variables, which were the chemical species to

be simulated. The rate constants k i to k j were subjected to variation, while kt to £ /j were

fixed as constant parameters. Other constants were Vm> Km, [O ?], [CO>]»[cyt c], [SOD],

[GPX\. The values for these constants were shown in Table 2 .22-36*38-39

110

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 5 3 Results

533 Kinetic profiles

Figs 2-4 show the concentration-time profiles for NO, O 2-, ONOO-, N2O3, GSNO

and GSH in 3 separate simulations where the in itia l GSH level was varied. For each

simulation, 5 scenarios were considered. Each scenario had a 2-fold difference in the

generation rate o f NO, £/. The results indicated that changing basal GSH levels did not

alter the kinetics o f NO, O?-, and ONOO-, w ith the steady-state concentrations being in

the (jM , pM and nM range, respectively (left panels. Figs 2-4). The kinetics o f the three

species reached steady state w ithin 10 mm. The steady-state concentrations decreased

(NO and ONOO- ) o r increased (O 2-) corresponding to the reduction o f kt.

The kinetic profiles ofN jO j, GSNO and GSH were sensitive to the variation o f

basal GSH levels (right panels, Figs 2-4). The profiles were consistent in a sense that

[GSNO] continuously increased at the expense o f GSH and N 2O3. As a result, [GSH] was

depleted and the time for a complete depletion depended on the basal level o f GSH (Figs

2-4) and the input rate o fNO (individual curves in each figure). It is interesting to note

that GSH and GSNO followed zero-order kinetics before GSH was exhausted, which

disobeyed second-order rate equations for these two species. This discrepancy w ill be

discussed later.

5 3 3 GSH as a dynamic switch

As indicated in the model (Fig. I), the continuous generation o f NO contributes to

the formation o f^O ? , a potential nitrosating agent. Strikingly, N 2O3 was kept at an

extremely low level due to the presence o f GSH (Fig. 2). An instantaneous elevation o f

[N 2O3 } was apparent when [GSH] approached a value o f zero (Fig. 2). The kinetic III

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. profiles were therefore step-like. Moreover, the slower the NO-generation rate was, the

longer fo r this phenomenon to be initiated, and the lower was the new steady-state [NzOj[.

When the basal [GSH] was reduced to I and 0.1 mM (Figs 3 and 4), GSH was depleted

much more quickly, followed by much earlier switching o f than it was at 10 mM.

To further explore the switch-like phenomenon, a zero-order input o f GSH was started at

a given time after GSH was depleted. The result showed that the system responded in a

switch-like fashion (Fig. 5). [NjOj] was sensitive to the perturbation and was reduced to

a new steady state immediately. Overall, the data suggested that GSH acted as a dynamic

switch in the reaction network.

53 3 Nonlinear dynamics at high NO and Ch~ input rates

To study the system behavior at high NO and Ch~ input rates, simulations were

conducted by setting k{ an dk> 100 times higher, w hile keeping kjkz ratios and other

parameters the same as in Fig. 2. The results were surprising because the dynamic

patterns in Fig. 6 significantly deviated from those in Fig. 2. The concentrations o f NO

were below S pM for all simulations with ki/kz ratios in the range o f0.0625 — 0.5, but at

equal NO and QT input rate, [NO] reached to 70 pM at steady state (Fig. 6). Compared

with Fig. 2, [NO] was relatively lower while [O f]was higher in a ll scenarios in Fig. 6.

The result suggested that O f outcompeted w ith NO when both NO and O 2- input rates

were high. It further implied that an increase in NO production couldn’t guarantee an

increase in [NO] as long as the input o f O f was not fixe d . Another interesting result, as

compared with Fig. 2, was the elevation o f [ONOO-] in Fig. 6, which suggested that the

112

1i i Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. system was more efficient in terms o f the generation o f ONOO . Finally, the kinetics o f

N2O3, GSNO and GSH in the right panel o f Fig. 6 coincided w ith the role and the kinetics

o fN O .

53.4 Phase portraits'*0

A phase-portrait approach was used to account fo r the nonlinear dynamics

illustra ted in Fig. 6 . To construct a phase portrait, the relationship between and dt

[NO\ was plotted according to equation 1, assuming that the contribution o f GSNO to the

kinetics o f NO was negligible under the simulation conditions. Hence, equation I

became

-MWOlton-MiTOfto,!. (8)

The steady-state O -f concentration with the same assumption was

[OT] = . (9) * k&NOl+kJiSODl

Substituting (9) into ( 8), equation I became

Kk£NO\ * (10) dt ^ kt[NO]+k5[SOD] u

Since k<, £ j, k i2 , [SOD] and [O2 ] were known parameters, by varying the ratio ki/kz and

p lo ttin g the data ( versus [M?D, a set o f curves was obtained (Fig. 7A). The x- dt

axis intersect implies = q , ^ corresponding value on x-axis being the d t

steady-state concentration o f NO, [#£)]». By plotting [MTJj, versus k//%>, a nonlinear

curve was obtained. This curve captured the essence o f nonlinearity in NO kinetics in 113

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Fig. 6 ; if £( < 0 .5 ^, [iVO^s was low and insensitive to the variations in £ 7 ; nevertheless,

EM?]** increased disproportionately as jfc> increased beyond 0.5 cjI. For the purpose o f

comparison, Fig. 8 showed a scenario where the dynamics was approximately linear.

5.4 Discussion

Using mathematical modeling, this study examined two questions related to the

reaction kinetics o f nitrogen oxide species (NO*) in an integrative dynamic system: (i)

what is the role o f GSH in modulating nitrosative and oxidative species, such as N 2O3

and ONOO"? (ii) how does the system behave when the generation rate o f NO and O 2"

varies? It is d iffic u lt to study these types o f questions using experimental approaches,

since it may require simultaneous measurements o f multiple short-lived species at

extremely low concentrations, in complex physiological matrices. Nevertheless, one can

gain a reasonable picture o f the behavior o f an in vivo system by using a modeling

approach to integrate the knowledge about individual elements (reactions) o f the system.

The underlying rationale was that the behavior o f a complex system usually is not

determined by an additive process.

Glutathione (GSH), the major low-molecular-weight thiol compound in the cell, is

best known fo r its role as a superoxide scavenger in mediating cellular redox reactions .41

Since cellular GSH levels are as high as 10 mM and GSH reacts w ith ONOO" and N 1O3

in aqueous media, it was suggested that GSH is a scavenger fo r reactive NO* as well .42

Moreover, the S-nitrosation product o f GSH, S-nitrosoglutathione (GSNO) was

considered as a possible carrier molecule fo r NO .43 In Chapter 4, GSH was shown to

diminish the NO-mediated nitiosation reaction. It also modulated the biphasic dose- 114

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. response relationship for the effect o f SOD on NO-mediated nitrosation. Since \GSH\

ranges from several hundred [iM (extracellular) to 10 mM (intracellular), the effect o f

GSH on the kinetics ofNOx was simulated w ith initial [GSH\ = 0.1,1 and 10 mM,

respectively. That GSH didn’t affect the kinetic profiles o fNO and ONOO- (Figs 2 -4 )

reflected the fact that in the model GSH does not react w ith NO directly, and the GSH

pathway is not the predominant route for the elimination o f ONOO-. Besides GSH,

ONOO- also reacts w ith a w ide variety o f biomolecules. In th is study, the reactions o f

ONOO- w ith CO2, cytochrome c (cyt c) and glutathione peroxidase (GPX) were only

included because o f the relative importance o f these reactions (Fig. I). ONOO- reacts

rapidly w ith CO 2 (k = 5.8 x 104 W V 1) to generate nitrosoperoxycarbonate adduct, which

decomposes to NO 3' and CO 2 in the absence o f other reactive molecules .30*44

ONOO-+CO, -* ONO,CO; ( 11)

ono,co; -> no;+co 2 ( 12)

Since COWbicarbonate/carbonate is an important buffering system in vivo and the

concentrations o f CO 2 (13 mM in plasma) and bicarbonate (12 mM in intracellular fluid

and 25 - 30 mM in plasma) are high, the reaction between ONOO- and CO 2 could be the

major route o f ONOO- disappearance in vivo .44*45 Although the rate constants for the

reactions between ONOO- and GPX (2 x 106 M 'V 1),29 and between ONOO- and cyt c (2

x 105 M 'V 1)31 are re la tive ly higher than that fo r the ONOO- - CQ» reaction, the

contribution o f GPX and cyt c in eliminating ONOO- could be less significant due to

their relatively lower concentrations in biological fluids (Table 2). The rate constant for

the reaction between ONOO- and GSH is 135 x IQ 3 M 'V 1,28 which is the smallest 115

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. compared w ith those o f the abovementioned reactions. Therefore, GSH w ill be far less

effective than COz in modulating ONOO- levels in the physiological environment, where

GSH is less abundant, e.g. blood plasma. In contrast, GSH may become important inside

the cells because o f its high intracellular concentration (10 mM). The reason that

[ONOO~[ in Fig.2 was comparable to that in Figs 2-3 was because the reaction rate o f

ONOO- with I mMCOz was s till at least 4-fold larger than that with GSH, even though

[GSH] was as high as 10 mM.

Zero-order kinetics was observed fo r GSH and GSNO, which disobeyed the rate

laws for both species. Since ONOO- and N 2O3 were the two molecules responsible for

the depletion o f GSH and the accumulation o f GSNO in the model, the discrepancy was

resolved when the kinetics o f ONOO- and N 2O3 was examined. While [ONOCT] reached

steady state w ith in 10 m in, [W2O3] was kept at nearly constant and extremely low levels

before GSH was completely depleted (Fig. 2). Therefore, d[iV,0 2 ] ^ dt dt

approximately zero in the early period. From (3) and (5), one immediately obtains:

k£ONOCr\[GSH\ = £4[tf0][< X I-(fc7[GPXl+fcg[C02]+ k jc y t cD[OWOO‘ ]

(13)

ktI[Af,0J][GSH]=^2[iV0f(02]-kl3[N20J] (H)

Since [M 7j and [O f] also reached steady states very quickly (Fig. 2), and [GPX], [CO?],

[ 0 ?] and [cyt c] are constants, it implies that the term

k,[ONOOr][GSH\^iNMiGSH\ in (4 ) and ( 6 ) is close to a constant and, therefore,

the elimination o f GSH and the formation o f GSNO foDowed zero-order kinetics.

116

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The current study proposed that GSH can act as a dynamic switch that controls the

concentration ofN iG s, which is an intriguing phenomenon from both kinetic and

biological standpoints. Kinetically, the switch suggested an almost discontinuous

solution to the differential equations, which is counterintuitive because the processes

were considered continuous. How could it happen? By closely examining equation 5

and the rate constants (Table 1), one finds that kn[GSH] »kn when [GSH] = 1 -1 0 mM.

Accordingly, the elimination o f N 2O3 was controlled by GSH at high levels. Under this

circumstance, the hydrolysis o f N 2O3 (ku) becomes a redundant e lim ination pathway that

contributes insignificantly to the whole process. The concentration o f N 2O3 remained

quite low due to the presence o f sufficiendy high levels o f GSH (Fig. 2 or Fig. 3).

However, the once redundant pathway became increasingly important as GSH

approached a depletion point at which the alternate elimination pathway took over and a

new steady state switched on. The nearly instantaneous occurrence o f the new steady-

state can be attributed to the first-ord er rate constant ku (Table I), which corresponds to a

h a lf-life as short as 0.4 ms.

From a biological standpoint, a sudden elevation o f a reactive NOx in a

physiological system could be catastrophic. N-nitrosation may result in the deamination

o f DNA bases .38 Furthermore, S-nitrosation o f proteins was attributed to the inhibition o f

some enzymes, such as glyceraldehydes-3-phosphate dehydrogenase 46 and the DNA

repair enzyme 0 5-methylguanine-DNA-methyitransferase .47 A brupt changes in S-

nitrosation status may also disrupt redox-based signaling transduction pathways .48 GSH,

therefore, may play an important role in modulating NOx-mediated cytotoxicity. For

example, the NO donor DEA/NO only caused a modest toxicity in Chinese hamsterV79 117

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. cells.42 However, the toxicity was dramatically increased when GSH was depleted in

these cells. Recently, cellular GSH status was linked to the differential iNOS regulation

in hepatocytes and inflammatory cells.49 W hile iNOS induction in hepatocytes in vivo

and in vitro was dependent on the intracellular GSH status and correlated w ith NF-kB

binding, GSH-depletion had no eflect on the expression o f iNOS in inflammatorycells.49

5.5 Summary

Kinetic modeling was used to explore the reaction network associated w ith the flee

radicals NO and O 2” Numerical simulations provided two testable predictions: (i) GSH

may modulate the nitrosation reaction in a switch-like fashion; (ii) Concurrently high NO

and Oz~ generation may result in nonlinear dynamics o f nitrogen oxide species.

5.6 References

1. Miranda KM, Espey MG, Jourd’heuil DJ, et al. The chemical biology o f nitric oxide. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, San Diego, 2000;4l-55.

2. Patel RP, McAndrew J, SeOak H, et al. Biological aspects o f reactive nitrogen species. Biochim. Biophys. Acta. 1999;1411:385-400.

3. Burek DG. Can we model nitric oxide biotransport? A survey o f mathematical models fora simple diatomic molecule w ith surprisingly complex biological activities. Annu. Rev. Biomed. Eng. 2001;3:109-143.

4. Lancaster Jr. JR. Simulation o f the diffusion and reaction o f endogenously produced nitric oxide. Proc. Natl. Acad. Sci. USA 1994;91:8137-8141.

5. Lancaster Jr. JR. A tutorial on the diffusibility and reactivity o f free nitric oxide. N itric Oxide: Biology and Chemistry. t997;l(l):l8-30.

6. Vaughn MW, Kuo L, Liao JC. Effective diffusion distance o f nitric oxide in the microcirculation. Am. J. Physiol. 1998;274(5):H1705-K1714.

118

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 7. Shin H-W, George SC. Microscopic modeling o f NO and S-nftrosoghrtathione kinetics and transport in human airways. J. AppL Physiol. 2001;90:777-788.

8. Chen B, Deen WM. Analysis o f the effects o f ceil spacing and liquid depth on nitric oxide and its oxidation producs in cell cultures. Chem. Res. Toxicol. 2001;14:135- 147.

9. Goss SPA, Hogg N, Kaiyanaraman B. The effect o f nitric oxide release rates on the oxidation o f human low density lipoprotein. J. BioL Chem. 1997;272:21647-21653.

10. Stanbro WD. A kinetic model o f the system: Tyrosy I radical-nitrogen oxide- superoxide ion. J. Theor. BioL 1999;197:557-567.

11. Stanbro WD. Modeling the interaction o f peroxynitrite with Iow-density lipoproteins. I. Plasma levels o f peroxynitrite. J. Theor. B iol. 2000;205:457-464.

12. Stanbro WD. Modeling the interaction o f peroxynitrite with Iow-density lipoproteins. IL Reaction/diffusion model o f peroxinitrite in Iow-density lipoprotein particles. J. Theor. B iol. 2000;205:465-471.

13. Stanbro WD. Modeling the interaction o f peroxynitrite with Iow-density lipoproteins. IIL The role o f antioxidants. J. Theor. Biol. 2000;205:473-482.

14. Vaughn MW, Liao JC. Estimation o f nitric oxide production and reaction rates in tissue by use o f a mathematical modeL Am. J. Physiol. 1998;274(6):H2163-H2176.

15. Laurent M, Lepoivre M, Tenu J-P. Kinetic modeling o f the nitric oxide gradient generated in vitro by adherent cells expressing inducible nitric oxide synthase. Biochem. J. 1996314:109-113.

16. Philippides A, Husbands P, O’Shea M . Four-dimensional neuronal signaling by nitric oxide: A computational analysis. J. Neurosci. 200030(3): 1199-1207.

17. Cobbold CA, Sherratt JA. Mathematical modeling o f nitric oxide activity in wound healing can explain keloid and hypertrophic scarring. J. Theor. BioL 2000304357- 288.

18. Bellamy TC, Wood J, Garthwaite J. On the activation o f soluble guanytyl cyclase by nitric oxide. Proc. NatL Acad. ScL USA 2002;99( I):507-510.

19. Lancaster Jr. JR. The physical properties o f nitric oxide: Determinants o f the dynamics o f NO in tissue. In: Ignarro LL eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000309-224.

20. Kleinert H, Boissel J-P, Schwarz PM, Fbrstermann U. Regulation o f the expression o f nitric oxide synthase isoforms. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000;105-128. 119

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 21. Thomas DD, Liu X, Kantrow SP, Lancaster Jr. JR. The biological lifetim e o f nitric oxide: Implications fo r the perivascular dynamics ofN O and O 2- Proc. Natl. Acad. ScL USA 2001^8(1)355-360.

22. Beckman JS, Koppenol WH. N itric oxide, superoxide, and peroxynitrite: the good, the bad, and the ugly. Am. J. PhysioL 1996;271:C1424-C1437.

23. Savill NJ, Weller R, Sherratt JA. Mathematical modeling o f nitric oxide regulation o f rete peg formation in psoriasis. J. Theor. B iol. 2002;214: 1- 16 .

24. Turner CP, Toye AM, Jones OTG. Keratinocyte superoxide generation. Free Rad. BioL Med. l998;24(3):40l-407.

25. O’Donnell VB, Azzi A. High rates o f extracellular superoxide generation by cultured human fibroblasts: Involvement o f a lipid-metabolizing enzyme. Biochem. J. 1996318:805-812.

26. Huie RE, Padmaja S. The reaction o f NO w ith superoxide. Free. Rad. Res. Common. 1993;18:195-199.

27. Fiefden EM, Roberts PB, Bray RC, Lowe DJ, Mautner GN, Rodlio G, Calabrese L. The mechanism o f action o f superoxide dismutase from pulse radiolysis and electron paramagnetic resonance. Biochem. J. 1974;139:49-60.

28. Koppenol WH, Moreno JJ, Pryor WA, Ischiropoulos H, Beckman JS. Peroxynitrite, a cloaked oxidant formed by nitric oxide and superoxide. Chem. Res. Toxicol. 1992;5:834-842.

29. Sies H, Sharov VS, Klotz L-O, Briviba K. Glutathione peroxidase protects against peroxynitrite-mediated oxidations. J. Biol. Chem. 199737237812-27817.

30. Denicola A, Freeman BA, Trujillo M, Radi R. Peroxynitrite reaction with carbon dioxide/bicarbonate: kinetics and influence on peroxynitrite-mediated oxidations. Arch. Biochem. Biophys. 1996333:49-58.

31. Thomson L, Trujillo M, Telleri R, Radi R. Kinetics o f sytochrome c2~ oxidation by peroxynitrite: Implications for superoxide measurements in nitric oxide-producing biological systems. Arch. Biochem. Biophys. 1995319:491-497.

32. Jourd’heuil D, Mai C, Laroux F, W ink DA, Grisham MB. The reaction o f S- nitrosoglutathione with superoxide. Biochem. Biophys. Res. Common. 1998;244:525-530.

33. Keshive M , Singh S, Wishnok JS, Tannenbaum SR, Deen WM. Kinetics o f S- nitrosation o f thiols in nitric oxide solutions. Chem. Res. ToxicoL 1996;9:988-993.

120

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 34. W ink DA, Darbyshire JF, Ninas RW, Saavedra IE , Ford PC. Reactions o f the bioregulatoty agent nitric oxide in oxygenated aqueous media: Determination o f the kinetics fo r oxidation and nitrosation by intermediates generated in the NO/CK reaction. Chem. Res. ToxicoL 1993;6:23-27.

35. Licht WR, Tannenbaum SR, Deen WM. Use o f ascorbic acid to inhibit nitrosation: kinetic and mass transfer considerations for an in vitro system. Carcinogenesis. 1988;9:365-372.

36. Antunes F, Salvador A, Marinho HS, Alves R, Pinto RE. Lipid peroxidation in metochondrial inner membranes. I. An integrative kinetic model. Free Rad. Biol. Med. l996;2I(7):9I7-943.

37. Coombes KR, Hunt BR, Lipsman RL, Osbom JE, Stuck GJ. Differential equations with Mathematica. 2nd ed. New York: John W iley & Sons, Inc.,

38. Radi R, Denicola A, Alvarez B, Ferrer-Sueta G, Rubbo H. The biological chemistry o f peroxynitrite. hi: Ignarro U . eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, San Diego, 2000;57-82.

39. G riffith OW. Biologic and pharmacologic regulation o f mammalian glutathione synthesis. Free Rad. Biol. Med. 1999;27:922-935.

40. Strogatz SH. Nonlinear dynamics and chaos. Reading MA: Addison-Wesley Publishing Company, 1994.

41. Sies H. Glutathione and its role in cellular functions. Free Rad. Biol. Med. 199927:916-921.

42. Wink DA, Nims RW, Darbysbire JF, et al. Reaction kinetics for nitrosation o f cysteine and glutathione in aerobic nitric oxide solutions at neutral pH. Insights into the fate and physiological effects o f intermediates generated in the NO/Oj reaction. Chem. Res. ToxicoL 1994;7:519-525.

43. Gaston B. N itric oxide and thiol groups. Biochimica et Biphysica Acta. 1999;1411:323-333.

44. Pfeiffer S, Mayer B, Hemmens B. N itric oxide: chemical puzzles posed by a biological messenger. Angew. Chem. Int. Ed. 1999;38:1714-1731.

45. Murphy MP, Packer MA, Scarlett JL, Martin SW. Peroxynitrite: a biologically significant oxidant. Gen. Pharmac. 199821:179-186.

46. Vedia VL, McDonald B, Reet B, et al. N itric oxide-induced S-nitrosylation o f glyceraldehydes-3-phosphate dehydrogenase inhibits enzymatic activity and increases endogenous ADP-riboxylation. J. Biol. Chem. 199226724929-24932.

121

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 47. Laval F, Wink DA. Inhibition by nitric oxide o f the repair protein, O6- methylguanine-DNA-methyltransferase. Carcinogenesis. 1994;15:443-447.

48. Stamler JS, Lamas S, Fang FC. Nitrosylation: The prototypic redox-based signaling mechanism. C ell, 2001;106:675-683.

49. Vos TA, van Goor H, Tuyt L, et al. Expression o f inducible nitric oxide synthase in endotoxemic rat hepatocytes is dependent on the cellular glutathione status. Hepatology. l999;29(2):42l-426.

122

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CONSTANT VALUEREFERENCES

ki »IQ'S M s 1 Vaughn et aL,1* Laurent et al.,0 Savill et aL"

k2 10"*—>I0‘sM s'1 Turner et a!..-* O’Donnell and A2 zf“

kj varied

k* 6 .7 x to’ v r V Huie and Padmaja2*

k* 2.4 x tO’tiT's1 FieldenetaL"

k& 1.35 x tO1 MTl s‘l Koppenol et aL2*

k7 2 x I0 6M-l s ‘ SiesetaL2*

k* 5.8 x to4 VT1 s l Denicolaetal."

k» 2.5 x 10* NT's'1 Thomson et aL“

kio 6 x I0 *V T V Jourd’heuil et aL“

k» 5.6 x I07W ' s ‘ Keshive et al.0

ku 6 xI06 O T V WinketaL**

k ij 1.6 x 1 0 V Licth et aL"

v» 3.2 x 10"1 M s 1 AntunesetaL*

Table 5.1. The rate constants used fo r the simulation.

123

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. PARAMETERVALUEREFERENCE m 3 5 *iM Antunes et a l.^ [CO,] 1-25 mM Radi et a l.34

C yt c1^ 400 p M Radi et a l.J8

[SOD] I - 10 |iM Beckman22

[GPX] 5.8 Antunes e t a l.36

Km 50 |lM Antunes e t a l/6 [GSH] I -10 mM G r iffith "

Table 5.2. Parameter values fo r the simulation.

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure 5.1. The Model GPX: Ghitathtone peroxidase Cyt c: Cytochrome c

125

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100

80

O w z 20

10 200 400 600 8QQ 1000 T im e (m in )

4 . 10 .

8 3 . . & S. 2. O 4 . «Z 1. O 2. 0 200 400 600 800 1000 T im e (m in )

10 . 0 .5 a. 6. 0 .3 o 0 .2 « (9 0.1 2. 0 200 400 600 800 1000 T im e (m in )

Figure 5.2. Concentration-time profiles o f NO, 0 2-, ONOO-, N20 3, GSNO, and G SH . Scenarios: (a) k( = k2; (b ) kt = 0.5 k2; (c> kt -0 2 5 k2; (cl) kt = 0.125 k2; (e) k, - 0.0625 k3, [GSHJbasai = 10 m M , k2 = 1*10'7 M/s for all scenarios.

126

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. IQQ 0 .5 80

r c a. ~0.3 o 40 &M 0.2 z z 20 0.1 0 0 200 400 600 800 1000 T im e (m in )

4 l .

3 ^0.8 S £ 0 .6 2 g 0 .4 o M « 1 0 0 200 400 600 800 1000 T im e (m in ) o.s l.

0 .5 0 .8 C 0 *4 f 0.6 , 0 .3 Cu£ 3** 0.2 O 0.1 0 .2 a 10 0 200 400 600 300 1000 T im e (m in )

F ig ure 53 . Concentration-time profiles o f NO, 0 2 , ONOO , N20 3, GSNO, and GSH. Scenarios: (a) k, = k2; (b ) k, = 0.5 k2; (c ) k, = 0.25 k2; (d ) kt = 0.125 k2; (e) k, = 0.0625 k2. [GSIfJbosai = I m M , k2 = I*10'7 M/s for all scenarios.

127

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. too 0 .5 80

SO

40

20 0 .1 0 200 400 SOO 800 1000 T im e (m in )

4 0.1

0.08 3 S. 0.06 2 s M i 0 .04 o « 1 ° 0.02

200 400 SOO 800 1000 T im e (m in )

0.1 0 .5

mX 0.04 (3 0.02

200 4QQ S0Q 800 1000 T im e (m in )

Figure 5.4. Concentration-time profiles o f NO, 0 2", ONOO", N20 3, GSNO, and GSH. Scenarios: (a) k, = k2; (b ) kt = 05 k2; (c) k, = 0.25 k2; (d ) kt = 0.125 k2; (e) kt = 0.0625 k2, [GSHjbasai = 0.1 m M , k2 = I*1Q7 M/s for all scenarios.

128

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 0 .2 5

0.2

C 0 .1 5

N

0 .0 5

0 200 400 600 800 1000 Time (min)

0.8

0.6

= 0 .4

0.2

0 200 400 600 800 1000 Time (min)

Figure 5.5. Concentration-time profiles of N20 3 and GSH at the initial GSH level equal to 1 mM. Arrow: start of zero-order GSH input (£3). k2 = 6 |jM /m in ; kt = 0.5 k2; k3 = (a) 20 JiM/min; (b) 10 |iM/min; (c) 5 {iM/min; (d) 2.5 |iM/m in; (e) 1.25 pM/min.

129

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. TO.

O 0.2

10 . 0 b .c .d .i 200 400 600 300 1QQQ T im e (m in )

400

300 * i — 200 o2 a t ° 100 (9

Q 2 4 6 8 10 200 400 600 300 1000 T im e (m in ) T im e (m in )

10 . 5 0 . 8. r 40. ~ 3 0 . b 4 . o 20. a t 19 2 1 0 . d ------. 0 0 200 400 600 800 1000 T im e (m in )

Figure 5.6. Concentration-time profiles of NO, 0 2~, ONOO', N20 3, GSNO, and G SH . Scenarios: (a) k, - k2; (b ) kt = 0.5 k2; (c) kt = 0.25 fc2; (d ) k, = 0.125 fc2; (e) kt = 0.0625 k2, [GSHJbasai= 10 m M , k2 = l*10‘s M/s for all scenarios.

130

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1400 2200 1000

o 800 5 600 u_t 400 200

100 200 300 400 500 MO (pM)

500

* 4 0 0

*3 0 0 « 2 200

100

0.5 t. 1.5 2. 2.5 3. 3.5 4. fcl/kz

Figure 5.7. (A) Phase portraits o f [NOl (B) [NOlss versus k /k 2. k2= L*10‘5 M/s; k /k 2 = (a ) 4; (h ) 2; (c> I ; (d ) 0.5; (e) 0 2 5 ; (f) 0.125

I Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1 .5

2 4 S 8 10 12 L4 16 18 20 NO (^ M )

4 Z

0.5 1. 1.5 2. 2.5 3. 3.5 4. k i / k 2

Figure 5.8. (A ) Phase portraits o f [NOl (B) [NOlss versus k /k 2. k2= 1*10^ M/s; k /k 2 = (a) 4; (b) 2; (c) 1; (d) 0.5; (e) 0.25; (f) 0.125

132

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. APP E N D IX A

MATHEMATICA Program for the Monte Carlo Simulations

Needs["Statistics’ContinuousDistributions'"]; Needs["Graphics'Graphics'"]; Needs["Statistics'LinearRegression'"]; Needs["Statistics'ConfidenceIntervaIs'"]; Needs ["Graphics' M uItipleListPlot' "]; Needs["Statistics'DataManipuIation'"]; Needs["Statistics'ContinuousDistributions'"];

f[x j := 100*XA0.75; MassLst= {0.02,0.25,2.5,5,14,70};

RunMonteCarlo [n_, file_, cv_] — ModuIe[{data, m =0}, W hile[m < n, m ++; data= {m ,TabIe[{xLst[[fl], RNDdataft,cv][U|]}, {j, I, k }]}; RecordData[data, file , m]; If[Mod[m, 10] = 0, ReportProgress[m, a]]]; stream = O penRead[fiIe]; answer = ReadList[stream, Expression]; CIose[fiJe]; answer ]

RNDdata[i_, cv_j := TabIe[Random[NonnalDistribution[yLst[[i]], cv*yLst[[i]]]], {i, I, k}]; RecordData[data_, file_, current_j — Module[ {stream}, stream = OpenAppendffile]; Write[stream, data]; Qose[fiIeQ ReportProgress[current_, n_J — Prmt["Monte Carlo is ", N[Round[I00 current/n]],"% complete."] ExponentList[n_] — ModuIe[ {exponentdata= {}, Ioglogregress, tempLst, m = 0}, While[m < n, m++; foglogregressfm] = Regress[Log[answer[[m]][[2]]], { I, x }, x j; tempLst[m] = {ParameterTable /. IogIogregress[m]}; exponentdata= AppendTo[exponentdata, te m p L st[m ][[I]][[I]][[2 ]][[I]]] I; exponentdata IB

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. I RsquareListfnJ := ModuIe[{Rsquaredata = {}, loglogregress, m = 0}, W hile[m < a, m++; loglogregress[m] = Regress[Log[answer[[m]][[2]]], {1, x }, x]; Rsquaredata = AppendTo[Rsquaredata, RSquared /. loglogregress [m ]] 1; Rsquaredata ]

1. mouse to rabbit scaling (M oujtat, Rab)

ClearAII[xLst, yLst, ErrorFreeData]; xLst = Take[MassLst, 3]; yLst = fix ] I. x -> xLst; k = LengthfxJLst]; ErrorFreeData = TabIe[{xLst[[t]],yLst[[i]]}, {iT I, k}] RunMonteCarIo[ 100, "MouRbDatal.dta", 0.2]; MouRbDatal = ExponentList[ 100] RunMonteCarIo[ 100, "MouRbData2.dta", 0 3 ]; MouRbData2 = ExponentList[ 100] midpoints = , 035,"", 0.55,"", 0.75, "*, 0.95, "", 1.15,"", 1 3 5 }; MouRbcountsI = BinCounts[MouRbDatal, {0 3,1 .4 ,0 .!}]; MouRbcounts2 = BinCounts[MouRbData2, {0 3 ,1 .4 ,0 .!}]; mourbplot = BarChartfMouRbcounts I , MouRbcounts2, Bar Labels -> midpoints, PlotLabel-> {"mouse,rat^abbit"}, PIotRange-> {A il, {0,80}}, BarStyle -> {RGBColortf),0,0], RGBCoIorfl, I, I]}]

2. mouse to Monkey scaling (Mou,Rat, Rab,Mon)

ClearAll[xLst, yLst, ErrorFreeData]; xLst= Take[MassLst, 4]; yLst = f[x ] /. x -> xLst; k = Length[xLst]; ErrorFreeData= Table[{xLst[[i]], yLst[[i]]}, {i, I,k }] RunMonteCarlo[ 100, "MouMnData I .dta”, 0 3 ]; MouMnDatal = ExponentList[ 100] RunMonteCarIo[100, "MouMnData2.dta", 0 3 ]; MouMnData2 = ExponentList[ 100] MouMncounts! = BinCountsfMouMnDatal, {0 3,1 .4,0 .!}]; MouMncounts2 = BinCounts[MouMnData2, {0 3 ,1 .4 ,0 .!}]; m oum nplot= BarChart[MouMncounts 1, MouMncounts2, BarLabels -> midpoints, 134

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. PIotRange -> {A ll, {0,80 }}, PIotLabel -> {"mouse.raLrabbiLmonkey"}, BarStyle -> {RGBCoIor£0,0,0], RGBCoIor[l, I, I}}]

3. mouse to Dog scaling (MouRat, Rab,MonJ>)

DeIeteFile[”MouDoDataI .dta"] DeIeteFiIe["MouDoData2.dta'’] ClearAl![xLst, yLst, ErrorFreeData]; xLst= Take[MassLst, 5]; yLst = f[x ] /. x -> x L st; k = Length[xLst]; ErrorFreeData=TabIe[{xLst[[i]],yLst[[i]]}, {i, I,k }] RunMonteCarIo[IOO, "MouDoDatal .dta", 0 2 ]; MouDoDatal = ExponentList[ 100] RunMonteCarIo[lOO, ,'MouDoData2.dta", 0.3]; MouDoData2 = ExponentList[ 100] MouDocountsI = BinCounts [MouDoDatal, {0.2,1.4,0.!}]; MouDocounts2 = BinCounts[MouDoData2, {02, 1.4,0.!}]; moudoplot = BarChart[MouDocounts I , MouDocounts2, Bar Labe Is -> midpoints, PIotRange-> {A ll, {0,100}}, PIotLabe! -> {"mouse,rat/abbiLmonkey,dog"}, BarStyle -> {RGBColor[0,0,0], RGBCoIor[I, I, I]}]

4. mouse to human scaling (MouRat, Rab, Mon, D, H)

DeIeteFiIe[”allsca!e I .dta”] DeIeteFiIe[,,alIsca!e2.dta"] CIearAIl[xLst, yLst, ErrorFreeData]; DeleteFiIe["aIIscaIe.dta"]; xL st = MassLst; yLst = f[x ] /. x -> xL st; k = Lengtb[xLst]; ErrorFreeData = TabIe[{xLst[[i]],yLst[[i]]}, {i, l,k }] RunMonteCarIo[ 100, "allscalel.dta”, 02 ]; allscaledatal = ExponentList[I00] RunMonteCarIo[ 100, ”allscale2.dta'\ 0 3 ]; allscaledata2 = ExponentList[ 100] allscalecountsl = BinCounts[al!scaIedataI, {0 2 ,1 .4 ,0 .!}]; a!Iscalecounts2 = BinCounts[alIscaIedata2, {02,1.4,0.1}]; allscaleplot= BarChart[aIIscaIecounts I, a!IscaIecounts2, BarLabels -> midpoints, PIotLabel -> {"mouse,rat^abbiLxnonkey,dogRuman"}, PIotRange •> {AIL (0,100}}, BarStyle -> {RGBCoIor[0,0,0], RGBCoIor{l, I, I]}] 135

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 5. cat to Monkey scaling (Rat, Rab,Mon)

ClearAll[xLst, yLst, ErrorFreeData}; xLst= Take[MassLst, {2 ,4 ,1}]; yLst = f [x ] /. x -> xLst; k=Length[xLst]; ErrorFreeData= Table[{xLst[[i]], yLst[[i]]}, {i, I, k}] RunMonteCarIo[iOO, "RatMnDatal .dta", 0.2]; RatMnDatal = ExponenlList[ 100] RunMonteCarlo [100, "RatMnData2.dta”, 0 3 ]; RatMnData2 = ExponentList[ 100] RatMncountsI = BmCounts[RatMnDatal, {03,1.4,0.1}]; RatMncounts2 = BinCounts[RatMnData2, {03,1.4,0.1}]; ratm nplot= BarChart[RatMncounts 1, RatMncounts2, BarLabels -> midpoints, PIotRange -> {AD, {0,60}}, PIotLabel -> {"rat^rabbiunonkey"}, BarStyle-> {RGBCoIor[0, 0,0], RGBCoIor[I, I, I]}}

6. ra t to Dog scaling (Rat, Rab,Mon,D)

CIearAlI[xLst, yLst, ErrorFreeData]; xLst= Take[MassLst, (2, 5 ,1}]; yLst = f[x ] /. x -> xLst; k = Length[xLst]; ErrorFreeData= TabIe[{xLst[[i]], yLst[[i]]}, {i, I, k}] DeleteFile["RatDoData.dta"]; RunMonteCarIo[I00, "RatDoDatal.dta”, 03 ]; RatDoDatal = ExponentListf 100] RunMonteCar!o[100, "RatDoData2.dta’\ 0 3 ]; RatDoData2 = ExponentList[ 100] RatDocountsI = BinCounts[RatDoDataI, {0 3 ,1 .4 ,0 .!}]; RatDocounts2 = BinCounts[RatDoData2, {0.2,1.4,0.1}]; ra tdop lot= BarChart[RatDocounts I, RatDocounts2, BarLabels -> midpoints, PIotRange -> {A ll, {0,60}}, PIotLabel -> {"ratjabbit^nonkey.dog"}, BarStyle -> {RGBCoIor[0,0,0], RGBCoIor[I, I, I]}]

7. rat to Human scaling (Rat, Rab>fonJDJH)

DeleteFiIe["RatHuDatal.dta”] DeIeteF9e[nRatHuData2.dtan] ClearAII [xLst, yLst, ErrorFreeData]; 136

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. xLst= TakefMassLst, {2,6,1}]; yLst= f[x] /. x -> xLst; k = Length[xLst]; ErrorFreeData= TabIe[{xLst[[i]], yLst[[i]]}, {i, l,k }] RunMonteCarlo[100, "RatHuDatal .dta1’, 0 3 ]; RatHuDatal = ExponentList[ 100] RunMonteCar lo [100, "RatHuData2.dta", 0.3]; RatHuData2 = ExponentList[ 100] RatHucountsI = BinCounts[RatHuDatal, {03, 1.4,0.1}]; RatHucounts2 = BinCounts[RatHuData2, {03, 1.4,0.1}]; rathuplot = BarChart[RatHucounts I, RatHucounts2, BarLabels -> midpoints, PIotRange -> {A ll, (0 ,8 0 }], PIotLabel -> {"raLrabbiMnonkey,dog,human”}, BarStyle -> {RGBCoIor[0,0,0], RGBCoIor[l, I, I]}]

8. rabbit to Dog scaling (Rab,Mon, D)

ClearAII[xLst, yLst, ErrorFreeData}; xLst= Take[MassLst, {3,5,1}]; yLst = fix ] /. x -> xLst; k = Length[xLst]; ErrorFreeData=TabIe[{xLst[[i]],yLst[[i]]}, (i, I, fc}] RonMonteCarIo[lOO, "RabDoDatal.dta”, 0.2}; RabDoDatal = ExponentList[ 100} RunMonteCarlo[lQ0, ”RabDoData2.dta", 0 3 ]; RabDoData2 = ExponentList[ 100] RabDocountsl = BinCounts[RabDoDatal, {0.2,1.4,0.!}]; RabDocounts2 = BinCounts[RabDoData2, {0 3 ,1 .4 ,0 .!}]; rabdoplot = BarChartfRabDocountsl, RabDocounts2, BarLabels -> midpoints, PIotRange -> {A ll, {0,80}}, PIotLabel -> {"rabbit^nonkey,dog”}, BarStyle-> {RGBColor[0,0,0], RGBCoIor[l, I, I]}]

9. Rabbit to human scaling (Rab, Mon, D, H)

ClearAU[xLst, yLst, ErrorFreeData]; xLst= TakefMassLst, -4]; yLst=f[x] /. x -> xLst; k = Length[xLst}; ErrorFreeData= TabIe[{xLst[[i]], yLst[[i]]}, {i, l,k }] RunMonteCarIo[100, "RabHuDatal .dta”, 0 3 ]; RabHuDatal = ExponentList[ 100] RunMonteCarIo[100, ”RabHuData2.dta”, 0 3 ]; RabHuData2 = ExponentList[ 100] RabHucountsl = BinCounts[RabHuDataI, {0 3 ,1 .4 ,0 .!}]; 137

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. RabHucounts2= BinCounts[RabHuData2, (02, 1.4,0.1}]; rabhuplot= BarChart[RabHucounts I, RabHucounts2, BarLabels -> midpoints, PIotRange -> {A ll, {0,80}}, PIotLabel -> {"rabbitjnonkey,dog,human"}, BarStyle-> (RGBCoIorfl), 0,0], RGBColor[l, I, I]}] 10. Monkey to Htonan scaling

CIearAH[xLst, yLst, ErrorFreeData]; xLst= TakefMassLst, -3]; yLst= fix ] /. x -> xLst; k = Length[xLst]; ErrorFreeData= TabIe[{xLst[[i]], yLst[[i]]}, {i, 1, k}] RunMonteCarlo [ 100, "MonHuDatal.dta”, 02]; MonHuDatal = ExponentList[ 100] RunMonteCarIo[ 100, ”MonHuData2.dta", 0.3]; MonHuData2 = ExponentList[ 100] MonHucountsI = BinCounts[MonHuDataI, {02, 1.4,0.1}]; MonHucounts2 = BinCounts[MonHuData2, {0.2,1.4,0.1}]; m onhnplot= BarChart[MonHucounts I, MonHucounts2, BarLabels -> midpoints, PIotRange -> {A ll, {0,80}}, PIotLabel -> {"monkey,dog^iuman"}, BarStyle -> {RGBCoIor[0,0,0], RGBCoIor[I, I, I]}] Show[GraphicsArray[{{mourbpIot, moumnplot}, {moudoplot, allscaleplot}, {ratmnplot, ratdoplot}, {rathuplot, rabdoplot}, {rabhuplot, monhuplotj}]]

138

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A PP E N D IX B

Release Mechanism o f N itric Oxide and Snperoxide from SIN-1

o tr ONOO-

,0 . SIN -1 SIN-ia n o x

NO* 0 N -N=C— CN r r * % cN

SIN-1 SIN -1C

Figure B .l. Mechanism fo r the release o f NO and O j' from SIN-1

139

i Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure B J. HPLC chromatograms for the decomposition kinetics o f SIN-I

140

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. BIBLIOGRAPHY

Adamic LA, Huberman BA. Power-law distribution o f the world wide web. Science. 2000^87:2115

Adolph EF. Quantitative relations in the physiological constituents o f mammals. Science. 1949;109:579-585.

Ahr HJ, Boberg M, Brendel E, Krause HP, Steinke W. Pharmacokinetics o f m iglitol: Absorption, distribution, metabolism, and excretion following administrationto rats, dogs, and man. Arzneim Forsch. 1997;47:734-745.

Albert R, Barabasi AL. Statistical mechanics o f complex networks. Mod. Phys. 2002;74:47-97.

Albert R, Barabasi AL. Topology o f evolving networks: Local events and universality. Phys. Rev. Lett. 2000;85:5234-5237.

Albert R, Jeong H, Barabasi AL. The diameter o f the world-wide web. Nature. 1999;40l:l30.

Antunes F, Salvador A, Marinho HS, Alves R, Pinto RE. Lipid peroxidation in metochondrial inner membranes. L An integrative kinetic model. Free Rad. BioL Med. 1996^1(7)^17-943.

Assreuy T, Cunha FQ, Epperlein M, Noronha-Dutra A, O’Donnell CA, Liew FY, Moncada S. Production o f nitric oxide and superoxide by activated macrophages and killin g o f Leishmania major. Eur. J. Immunol. 1994;24:672-676

A xtell RL. Z ip f distribution o f U.S. firm sizes. Science. 2001;293:1818-1820.

Baggot JD. Application o f inter species scaling to the bispyridinium oxime HI-6. Am i Vet Res. 1994;55:689-691.

141

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Banavar JR, Maritan A , Rinaldo A. Size and form in efficient transportation networks. Nature. 1999,399:130-132.

Barabasi AL, Albert R, Jeong H. Mean-field theory for scale-free random networks. PhysicaA. 1999;272:173-187.

Barabasi A L, Albert R. Emergence o f scaling in random networks. Science. 1999;286:509-512

Barabasi A L, Jeong H, Neda Z, Ravasz E, Schubert A , Vicsek T. Evolution o f the social network o f scientific collaborations. ArXiv.org e-Print archive. 2001. http://www.arxiv.Org/abs/cond-mat/0104162

Bazin-Redureau M, Pepin S, Hong G, Debray M , Schemnann JM. Interspecies scaling o f clearance and volume o f distribution for horse antivenom F(ab*)2. Toxicol AppI PharmacoL 1998;150:295-300.

Beckman JS, Chen J, Crow JP, Ye YZ. Reactions o f nitric oxide, superoxide and peroxynitrite with superoxide dismutase in neurodegenration. Prog. Brain. Res. 1994;103:371-380

Beckman JS, Koppenol WH. N itric oxide, superoxide, and peroxynitrite: the good, the bad, and the ugly. Am. J. Physiol. 1996;271:C1424-C1437.

Bellamy TC, Wood J, Garthwaite J. On the activation o f soluble guanylyt cyclase by nitric oxide. Proc. NatL Acad. Sci. USA 2002;99(I):507-510.

Bernier M , Manning AS, Hearse DJ. Reperfusion arrhythmias: dose-related protection by anti-free radical interventions. Am. J. Physiol. 1989;256:1344-1352

Bhalla US, Iyengar R. Emergent properties o f networks o f biological signaling pathways. Science. 1999;283:381-387.

Bjorkman S, Redke F. Clearance o f fentanyl, alfentanii, methohexitone, thiopentone and ketamine in relation to estimated hepatic blood flow in several animal species: Application to prediction o f clearance in man. J Pbarm PharmacoL 2000;52:1065-1074.

Bohn H, Schonafinger K. Oxygen and oxidation promote the release o f nitric oxide from sydnonimines. J. Cardiovasc. PharmacoL I989;I4(Suppt. 11): S6-S12

Bonate PL, Howard D. Prospective aOometic scaling: Does the emperor have clothes? J C lin PharmacoL 2000;40:665-670.

Bonati M , Latini R, Tognoni G. Interspecies comparison o f in vivo caffeine pharmacokinetics in man, monkey, rabbit, rat, and mouse. Drug Metab Rev. 1984- 85;I5:I355-I383.

142

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Boxenbaum H. Interspecies scaling, allometry, physiological time, and the ground plan o f pharmacokinetics. J Pharmacokm Biopharm. 1982;10:201-227.

Brazzell RK, Park YH, Wooldridge CB, et al. Interspecies comparison o f the pharmacokinetics o f aldose reductase inhibitors. Drug Metab EHspos. 1990;18:435-440.

Bregante M A, Saez P, Aramayona JJ, et aL Comparative pharmacokinetics o f enrofloxacin in mice, rats, rabbits, sheep, and cows. Am J Vet Res. 1999;60:1111-1116.

Brocks DR, Freed M I, Martin DE, et aL Interspecies pharmacokinetics o f a novel hematoregulatory peptide (SK&F 107647) in rats, dogs, and oncologic patients. Pharm Res. 1996;13:794-797.

Brunelli L, Crow JP, Beckman JS. The comparative toxicity o f nitric oxide and peroxynitrite to Escherichia coli. Arch. Biochem. Biophys. 1995316:327-334

Burek DG. Can we model nitric oxide biotransport? A survey o f mathematical models for a simple diatomic molecule with surprisingly complex biological activities. Annu. Rev. Biomed. Eng. 2001;3:109-143.

Calabrese V , Bates TE, Stella AMG. NO synthase and NO-dependent signal pathways in brain aging and neurodegenerative disorders: The role o f oxidant/antioxidant balance. Neurochem. Res. 2000;25:1315-1341

Calder WA III. Size, Function and Life History. Cambridge, M A: Harvard University Press, 1984.

Chen B, Deen WM. Analysis o f the effects o f cell spacing and liquid depth on nitric oxide and its oxidation producs in cell cultures. Chem. Res. Toxicol. 2001;14:135-147.

Cherkofsky SC. I-Aminocyclopropanecarboxylic acid: Mouse to man interspecies pharmacokinetic comparisons and aHometric relationships. J Pharm Sci. 1995;84:1231- 1235.

Chung M , Radwanski E, Loebenberg D, et aL Interspecies pharmacokinetic scaling o f Sch 34343. J Antmricrob Chemother. I985;I5 (SuppL Q:227-233.

C iriolo MR, De Martino A, Lafavia E, Rossi L, Cam MT, RotOio G. Cu^n-superoxide dismutase-dependent apoptosis induced by nitric oxide in neuronal cells. J. BioL Chem. 2000;275:5065-5072

Clark B, Smith DA. Metabolism and excretion o f a chromone carboxylic acid (FPL 52757) in various animal species. Xenobiotica. 1982;12:147-153.

Coassolo P, Fischli W, Clozel J-P, Chou RC. Pharmacokinetics o f remikiren, a potent orally active inhibitor o f human renin, in rat, dog, and primates. Xenobiotica. 199636333-345. 143

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Cobbold CA, Shenatt JA. Mathematical modeling o f nitric oxide activity in wound healing can explain keloid and hypertrophic scarring. J. Theor. B iol. 2000;204:257-288.

Cohn DL. Optimal systems: L The vascular system. Bull Math Biophys. 1954;16:59-74.

Cohn DL. Optimal systems: IL The vascular system. Bull Math Biophys. I955;17:219- 227.

Coombes KR, Hunt BR, Lipsman RL, Osborn JE, Stuck GJ. Differential equations with Mathematica. 2nd ed. New York: John W iley & Sons, Inc.,

Cooper CE. N itric oxide and cytochrome oxidase: Substrate, inhibitor or effector? Trends Biochem. Sci. 2002;27:33-39

Cosson VF, Fuseau E, Efthymiopoulos C, Bye A. Mixed effect modeling o f sumatriptan pharmacokinetics during drug development I: Interspecies allometric scaling. J Pharmacokin Biopharm. 1997;25:149-167.

Cruze CA, Kelm GR, Meredith MP. Interspecies scaling o f tebufelone pharmacokinetic data and application to preclinical toxicology. Pharm Res. 1995;12:895-901.

Davi H, Tronquet C, Calx J, et aL Disposition o f diudronate (Skelid) in animals. Xenobiotica. 1999;29:1017-1031.

Denicola A, Freeman BA, Trujillo M, Radi R. Peroxynitrite reaction w ith carbon dioxide/bicarbonate: kinetics and influence on peroxynitrite-mediated oxidations. Arch. Biochem. Biophys. 1996;333:49-58.

Dodds PS, Rothman DH, Weitz JS. Re-examination o f the "3/4-law " o f metabolism. J Theor Biol. 2001;209:9-27.

Dorogovtsev SN, Mendes JFF, Samukhin AN. Structure o f growing networks w ith preferential linking. Phys. Rev. Lett 2000;85:4633-4636.

Dorogovtsev SN, Mendes JFF. Evolution o f networks. ArXiv.org e-Print archive. 2001. http://www.arxiv.Qrg/abs/cond-mat/Ql06144

Dorogovtsev SN, Mendes JFF. Language as an evolving word web. Proc R Soc Lond B. 2001;268:2603-2606.

Dorogovtsev SN, Mendes JFF. Scaling properties o f scale-free evolving networks: Continuous approach. Phys. Rev. E. 200I;63:056125;I-I9.

Duthu GS. Interspecies correlation o f the pharmacokinetics o f erythromycin, oleandomycin, and tylosin. J Pharm Sci. 1985;74:943-946.

144

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Earn DJD, Rohani P, BoDcer MB, Grenfell BT. A simple model fo r complex dynamical transitions in epidemics. Science. 2000;287:667-670.

Edwards NA. Scaling o f renal functions in mammals. Comp Biochem Physiol. l975;52A:63-66.

Efthymiopoulos C, Battaglia R, Strolin Benedetti M . Animal pharmacokinetics and interspecies scaling o f FCE 22101, a penem antibiotic. J Antimicrob Chemother. 1991;27:517-526.

Ericsson H, Tholander B, Bjorkman JA, Nordlander M , Regardh CG. Pharmacokinetics o f new calcium channel antagonist clevidipine in the rat, rabbit, and dog and pharmacokinetic/pharmacodynamic relationship in anesthetized dogs. Drug Metab Dispo. 1999;27:558-564.

Feelisch M , Ostrowski J, Noack EJ. On the mechanism o f NO release from sydnonimines. Cardiovasc. PharmacoL l989;l4(Suppl. ll):S13-S22

Feldman HA, McMahon TA. The 3/4 mass exponent for energy metabolism is not a statistical artifact. Respir Physiol. 1983;52:149-163.

Feng MR, Loo J, W right J. Disposition o f the antipsychotic agent Cl-1007 in rats, monkeys, dogs, and human cytochrome p450 2D6 extensive metabolizers: Species comparison and aUometric scaling. Drug Metab Dispos. 1998;26:982-988.

Feng MR, Lou X , Brown RR, Hutchaleelaha A. AUometric pharmacokinetic scaling: Towards the prediction o f human oral pharmacokinetics. Pharm Res. 2000;17:410-418.

Fielden EM, Roberts PB, Bray RC, Lowe DJ, Mautner GN, R otilio G, Calabrese L. The mechanism o f action o f superoxide dismutase from pulse radiolysis and electron paramagnetic resonance. Biochem. J. 1974;139:49-60.

Frank S, Kampfer H, Podda M, Kaufmann R, Pfeilschifter J. Identification o f copper/zinc superoxide dismutase as a nitric oxide-regulated gene in human (HaCaT) keratinocytes: Implicadons for keratinocyte proliferation. Biochem. J. 2000;346:719-728

Ganster RW, Geller DA. Molecular regulation o f inducible nitric oxide synthase. In: Ignarro L f. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000;129-156

Gaspari F, Bonati M . Interspecies metabolism and pharmacokinetic scaling o f theophylline disposition. Drug Metab Rev. 1990;22:179-207.

Gaston B. N itric oxide and thiol groups. Biochim. Biophys. Acta I9 9 9 ;l4 l 1:323-333

145

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Gergel D, M isik V, Ondrias K, Cederbaum AL Increased cytotoxicity o f 3- Morpholinosydnonimine to HepG2 cells in the presence o f superoxide dismutase. J. Biol. Chem. 1995;270:20922-20929

Gombar CT, Harrington GW, Pylypiw HM Jr, et al. Interspecies scaling o f the pharmacokinetics o fN-nitrosodimethylamine. Cancer Res. 1990;50:4366-4370.

Goss SPA, Hogg N, Kalyanaraman B. The effect o f nitric oxide release rates on the oxidation o f human low density lipoprotein. J. Biol. Chem. 1997;272:21647-21653.

GreneLerouge NAM, Bazin-Redureau M I, Debray M , Schermann JM. Interspecies scaling o f clearance and volume o f distribution for digoxin-specific Fab. Toxicol Appl PharmacoL 1996;138:84-89.

G riffith OW. Biologic and pharmacologic regulation o f mammalian glutathione synthesis. Free Rad. BioL Med. 1999;27:922-935

Grindei JM, O'Neil PG, Yorgey KA, et aL The metabolism o f zomephac sodium L Disposition in laboratory animals and man. Drug Metab Dispo. 1980;8:343-348.

Grisham MB, Jounfheuil D, Wink DA. N itric oxide. L Physiological chemistry o f nitric oxide and its metabolites:implications in inflammation. Am. J. Physiol. 1999;276:G315- 321

Hayton WL. Maturation and growth o f renal function: Dosing renally cleared drugs in children. AAPS PharmSci. 2000;2(l), article 3. Available from: http://www.pharmsci.Org/scientificjournaIs/pharmsci/journal/3.html

Herault JP, Donat F, Barzu T, et al. Pharmacokinetic study o f three synthetic AT-binding pentasaccharides in various animal species-extrapolation to humans. Blood Coagul Fibrinol. 1997;8:161-167.

Heusner AA. Energy metabolism and body size. IL Dimensional analysis and energetic non-similarity. Resp Physiol. 1982;48:13-25.

Heusner AA. Energy metaholism and body size. I. Is the 0.75 mass exponent o f Kleiber's equation a statistical artifact? Respir Physiol. 1982;48:1-12.

Higuchi S, Shiobara Y . Comparative pharmacokinetics o f nicardipine hydrochloride, a new vasodilator, in various species. Xenobiotica. 1980;10:447-454.

Hildebrand M . Inter-species extrapolation o f pharmacokinetic data o f three prostacyclin- mimetics. Prostaglandins. 1994;48:297-312.

Hinderling PH, Dilea C, Koziol T, M illington G. Comparative kinetics o f sematilide in four species. Drug Metab Dispo. 1993;21:662-669.

146

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Hobbs AJ, Fukuto JM, Ignarro LJ. Formation o f free nitric oxide from L-arginine by nitric oxide synthase: Direct enhancement o f generation by superoxide dismutase. Proc. Natl. Acad. Sci. USA I9 9 4;9 l:10992-10996

Hoei PG. Introduction to mathematical statistics, New York, NY: John WQey & Sons, Inc, 1962.

Hu TM, Hayton WL. AUometric scaling o f xenobiotic clearance: Uncertainty versus universality. AAPS PhannSci. 20013(4). article 29. (http://www.pharmsci.oniA

Huberman BA, Adamic LA. Growth dynamics o f the world-wide web. Nature. I999;40I:131.

Huie RE, Padmaja S. The reaction o f NO w ith superoxide. Free. Rad. Res. Commun. 1993;18:195-199.

Ischiropoulos H, Zhu L, Chen J, Tsai M, Martin JC, Smith CD, Bechman JS. Peroxynitrite-mediated tyrosine nitration catalyzed by superoxide dismutase. Arch. Biochem. Biophys. 1992;298:431-437

fehigami M, Saburomaru K, Niino K, et al. Pharmacokinetics o f in the rat, rabbit, and beagle dog. Arzneim Forsch. 1979;29:266-270.

Ishii T, Iwahashi H, Sugata R, Kido R. Superoxide dismutase enhances the toxicity o f 3- hydroxyanthranilic acid to bacteria. Free Rad. Res. Commun. 1991;14:187-194

Izumi T, Enomoto S, Hosiyama K, et al. Prediction o f the human pharmacokinetics o f troglitazone, a new and extensively metabolized antidiabetic agent, after oral administration, w ith an animal scale-up approach. J Pharmacol Exp Ther. 1996;277:1630-1641.

Jeong H, Tombor B, Albert R, Oltvai ZN, Barabasi AL. The large-scale organization o f metabolic networks. Nature. 2000;407:651-654.

Jezequel SG. Fluconazole: Interspecies scaling and aUometric relationships o f pharmacokinetic properties. J Pharm PharmacoL 1994;46:196-199.

Jourd’heuil D, Mai C, Laroux F, W ink DA, Grisham MB. The reaction o f S- nitrosoghitathione with superoxide. Biochem. Biophys. Res. Commun. 1998344:525- 530.

Jourd’heuil D, Stephen Laroux F, Miles AM, W ink DA, Grisham MB. Effect o f superoxide dismutase on the stability o f S-nitrosothiols. Arch. Biochem. Biophys. 1999361323-330

Kaul S, Daudekar KA, Schilling BE, Barbhaiya RH. Toxicokinetics o f 2*3 - deoxythymidine, stavudine (D4T). Drug Metab Dispos. 199937:1-12. 147

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Kawakami J, Yamamoto EC, Sawada Y , Iga T. Prediction o f brain delivery o f ofloxacin, a new quinolone, in the human ftom animal data. J Pharmacokinet Biopharm. 1994^2:207- 227.

Kaye B, Brearley CJ, Cussans NJ, Herron M, Humphrey MJ, M ollatt AR. Formation and pharmacokinetics o f the active drag candoxatrilat in mouse, rat, rabbit, dog and man following administration o f the produg candoxatriL Xenobiotica. 1997;27:1091-1102.

Kedziora J, Bartosz G. Down’s syndrome: a pathology involving the lack o f balance o f reactive oxygen species. Free Rad. BioL M ed 1988;4:317-330

Keshive M, Singh S, Wishnok JS, Tannenbaum SR, Deen WM. Kinetics ofS-nitrosation o f thiols in nitric oxide solutions. Chem. Res. ToxicoL 1996;9:988-993.

Khor SP, Amyx H, Davis ST, Nelson D, Baccanari DP, Spector T. Dihydropyrimidine dehydrogenase inactivation and 5-fluorouraciI pharmacokinetics: AUometric scaling o f animal data, pharmacokinetics and toxicodynamics o f 5-fluorouraciI in humans. Cancer Chemother Pharmacol. 1997;39:233-238.

Khor SP, McCarthy EC, DuPont M , Murray EC, Timony G. Pharmacokinetics, pharmacodynamics, allometry, and dose selection o f rPSGL-Ig for phase I tria l. J Pharmacol Exp Ther. 2000;293:618-624.

Kim SH, Kim WB, Lee MG. Interspecies pharmacokinetic scaling o f a new carbapenem, DA-1131, in mice, rats, rabbits and dogs, and prediction o f human pharmacokinetics. Biopharm Drug Dispos. 1998;19:231-235.

K irillova OV. Communication networks with an emergent dynamical structure. Phys. Rev. Lett. 2001;87:06870l;l-4.

Kleiber M. Body size and metabolism. Hilgardia. 1932;6:315-353.

ECIemert H, Boissel J-P, Schwarz PM, Fdrstermann U. Regulation o f the expression o f nitric oxide synthase isoforms, hi: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st E d, San Diego: Academic Press, 2000;I05-128.

ECIotz U, Antonin K-H, Bieck PR. Pharmacokinetics and plasma binding o f diazepam in man, dog, rabbit, guinea pig and rat. J Pharmacol Exp Ther. 1976;199:67-73.

Koesling D, Friebe A . Structure-function relationships in NO-sensitive guanylyl cyclase. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed, San Diego: Academic Press, 2000;369-379

Koppenol WH, Moreno JJ, Pryor WA, Ischiropoulos H, Beckman JS. Peroxynitrite, a cloaked oxidant formed by nitric oxide and superoxide. Chem. Res. ToxicoL I992;5:834- 842.

148

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Krapivsky PL* Redner S, Leyvraz F. Connectivity o f growing random networks. Phys. Rev. Lett. 2000;85:4629-4632.

Kurihara A , Naganuma EL Hisaoka M , Tokiw a EL Kawahara Y . Prediction o f human pharmacokinetics o f panipenem-betamipron, a new carbapenem, from animal data. Antimicrob Ag Chemother. 1992^6:1810-1816.

Lancaster Jr. JR. A tutorial on the d iffiisib ility and reactivity o f free nitric oxide. N itric Oxide: Biology and Chemistry. I997;l(l):l8-30.

Lancaster Jr. JR. Simulation o f the diffusion and reaction o f endogenously produced nitric oxide. Proc. Natl. Acad. ScL USA 1994;91:8137-8141.

Lancaster Jr. JR. The physical properties o f nitric oxide: determinants o f the dynamics o f NO in tissue, hi: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000^09-224

Lancaster Jr. JR. The physical properties o f nitric oxide: Determinantso f the dynamics o f NO in tissue, hi: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000^09-224.

Lapka R, Rejholec V, Sechser T, Peterkova M, Smid M. Interspecies pharmacokinetic scaling o f metazosin, a novel alpha-adrenergic antagonist Biopharm Drug Dispo. 1989;10:581-589.

Lashev LD, Pashov DA, Kanelov I. Species specific pharmacokinetics o f rolitetracycline. J Vet Med A. 1995;42:201-208.

Lashev LD, Pashov DA, Marinkov TN. Interspecies differences in the pharmacokinetics o f kanamycin and apramycin. Vet Res Comm. 1992;16:293-300.

Laurent M , Lepoivre M, Tenu J-P. Kinetic modeling o f the nitric oxide gradient generated in vitro by adherent cells expressing inducible nitric oxide synthase. Biochem. J. 1996;314:109-113.

Laval F, Wink DA. Inhibition by nitric oxide o f the repair protein, 0 6-methyIguanine- DNA-methyltransferase. Carcinogenesis. 1994;15:443-447.

Lave T, Dupin S, Schmidt C, Chou RC, Jaeck D, Coassolo PH. hitegration o f in vitro data into aUometric scaling to predict hepatic metabolic clearance in man: Application to 10 extensively metabolized drugs. J Pharm Sci. 1997;86:584-590.

Lave T, Levet-Trafit B, Schmitt-Hoffinann AH, et aL Interspecies scaling o f interferon disposition and comparison o f aUometric scaling w ith concentration-time transformations. J Pharm Sci. 1995;84:1285-1290.

149

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Lave T, Portmann R, Schenker G, et al. Interspecies pharmacokinetic comparisons and aUometric scaling o f napsagatran, a low molecular weight thrombin inhibitor. J Pharm PharmacoL 1999;51:85-91.

Lave T, Saner A , Coassolo P, Brandt R, Scbmitt-Hoffman AH, Chou RC. Animal pharmacokinetics and interspecies scaling from animals to man o f lamifiban, a new platelet aggregation inhibitor. J Pharm PharmacoL 1996;48:573-577.

Leloup J-C, Goldbeter A. Modeling the molecular regulatory mechanism o f circadian rhythms in Drosophila. Bioessays. 2000;22:84-93.

Leusch A, Troger W, Greischel A, Roth W. Pharmacokinetics o f the Ml-agonist talsaclidine in mouse, rat, rabbit, and monkey, and extrapolation to man. Xenobiotica. 2000;30:797-813.

Licht WR, Tannenbaum SR, Deen WM. Use o f ascorbic acid to inhibit nitrosation: kinetic and mass transfer considerations for an in vitro system. Carcinogenesis. 1988;9:365-372.

Lin C, Gupta S, Loebenberg D, Cayen MN. Pharmacokinetics o f an eveminomicin (SCH 27899) in mice, rats, rabbits, and cynomolgus monkeys following intravenous administration. Antimicrob Ag Chemother. 2000;44:916-919.

Liochev SI, Fridovich L Copper- and zinc-containing superoxide dismutase can act as a superoxide reductase and a superoxide oxidase. J. Biol. Chem. 2000;275:38482-38485

Lipton SA., Choi Y-B, Pan Z-H, Lei SZ, Chen H-SV, Sucher NJ, Loscaiso J, Singel DJ, Stamler JS. A redox-based mechanism fo r the neuroprotecdve and neurodestructive effects o f nitric oxide and related nitroso-compounds. Nature 1993;364:626-632

Mahmood L Balian JD. Interspecies scaling: Predicdon clearance o f drugs in humans. Three different approaches. Xenobiotica. 1996;26:887-895.

Mahmood L Critique o f prospective aUometric scaling: Does the emperor have clothes? J C lin PharmacoL 2000;40:671-674.

Mahmood L Interspecies scaling o f renaily secreted drugs. Life Sci. 1998;63:2365-2371.

MaUery SR, Landwehr DJ, Ness GM, Clark YM, Hohl CM. Thiol redox modulation o f tumor necrosis factor-a responsiveness in cultured AIDS-related Kaposi’s sarcoma cells. J. CeU. Biochem. 1998;68:339-354

Mao GD, Thomas PD, Lopaschuk GD, Poznansky MJ. Superoxide dismutase (SOD)- catalase conjugates. J. BioL Chem. 1993;268:416-420

M arietta M A , Mammalian synthesis o f nitrite, nitrate, nitric oxide, and N-nitrosadng agents. Chem. Res. ToxicoL 1988;1:249-257. 150

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Marroum PJ, Uppoor RS, Parmelee T, Ajayi F, Bumett A, Yuan R, Svadjian R, Lesko LJ, Balian JD. la vivo drug-drug interaction stadies-a survey o f all new molecular entities approved from 1987 to 1997. C lin Pharmacol Ther. 2000;68:280-285.

Masters BS. Structural variations to accommodate functional themes o f the isoforms o f NO synthases. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 2000^1-104

Matsushita H, Suzuki H, Sugiyama Y , et al. Prediction o f the pharmacokinetics o f cefodizime and cefotetan in humans from pharmacokinetic parameters in animals. J Pharmacobio-Dyn. 1990;13:602-611.

McCord JM. The importance o f oxidant-antioxidant balance, hr Montagnier L, O livier R, Pasquier C. eds. Oxidative Stress in Cancer, AIDS, and Neurodegeneradve Diseases, New York: Marcel Dekker, l998;l-7

McGovren JP, W illiams MG, Stewart JC. Interspecies comparison o f acivicin pharmacokinetics. Drug Metab Dispo. 1988;16:18-22.

Meakm P. Fractals, scaling and growth far from equilibrium, Cambridge, UK: Cambridge University Press, 1998.

Mehta SC, Lu DR. Interspecies pharmacokinetic scaling o f BSH in mice, rats, rabbits, and humans. Biopharm Drug Dispos. 1995;16:735-744.

Miles AM , Gibson MF, Kirshna M, Cook JC, et al. Effects o f superoxide on nitric oxide- dependent N-nitrosation reactions. Free Rad. Res. 1995;23:379-390

Miles AM , Scott Bohle D, Glassbrenner PA, et al. Modulation o f superoxide-dependent oxidation and hydroxyiation reactions by nitric oxide. J. Biol. Chem. 1996;271:40-47

Miranda KM, Espey MG, Jourd’heuil DJ, et al. The chemical biology o f nitric oxide. In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed, San Diego: Academic Press, San Diego, 2000;41-55

Miranda KM , Espey MG, Jourd’heuil DJ, et aL The chemical biology o f nitric oxide, hi: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed, San Diego: Academic Press, San Diego, 2000;4I-55.

MitsuhasM Y , Sugiyama Y, Ozawa S, et al. Prediction o f ACNU plasma concentration­ time profiles in humans by animal scale-up. Cancer Chemother PharmacoL 1990;27:20- 26.

Montoya JM, Sole RV. Small world patterns in food webs. ArXiv.org e-Print archive. 2000. http://www.arxiv.org/abs/cond-mat/0011195

151

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Mordenti J, Chen SA, Moore JA, Ferraiolo BL, Green JD. Interspecies scaling o f clearance and volume o f distribution data for five therapeutic proteins. Pharm Res. 1991;8:1351-1359.

Mordenti J, Osaka G, Garcia K , Thomsen FC, Licko V, Meng G. Pharmacokinetics and interspecies scaling o f recombinant human factor V III. Toxicol Appl PharmacoL 1996;136:75-78.

Mordenti J. Man versus beast: Pharmacokinetic scaling in mammals. J Pharm Sci. 1986;75:1028-1040.

Mordenti J. Pharmacokinetic scale-up: Accurate prediction o f human pharmacokinetic profiles from animal data. J Pharm Sci. 1985;74:1097-1099.

Morrison BM, Morrison JH, Amyotrophic lateral sclerosis associated w ith mutations in superoxide dismutase: a putative mechanism o f degeneration. Brain Res. Rev. 1999;29:121-135

Mukai H, Watanabe S, Tsuchida K, Morino A. Pharmacokinetics o f NS-49, a phenethylamme class a l A-adrenoceptor agonist, at therapeutic doses in several animal species and interspecies scaling o f its pharmacokinetic parameters. Int J Pharm. 1999;186:215-222.

Murphy ME, Sies H. Reversible conversion o f nitroxyl anion to nitric oxide by superoxide dismutase. Proc. Natl. Acad. Sci. USA 1991;88:10860-10864

Murphy MP, Packer MA, Scarlett JL, Martin SW. Peroxynitrite: a biologically significant oxidant Gen. Pharmac. 1998;31:179-186.

Murray CD. The physiological principle o f minimum work. L The vascular system and the cost o f blood volume. Proc Natl Acad Sci U S A. 1926;12:207-214.

Nakajima Y, Hattori K, Shinsei M , et al. Physiologically-based pharmacokinetic analysis o f grepafloxacin. B iol Pharm Bull. 2000;23:1077-1083.

Nedospasov A , Rafikov R, Beda N, N udler E. An autocatalytic mechanismo f protein nitrosylation. Proc. Natl. Acad. Sci. USA 2000;97:13543-13548

Newman MEJ. The structure o f scientific collaboration networks. Proc Natl Acad Sci USA. 2001;98:404-409.

Norris KH, Homsby P. Cytotoxic effects o f expression o f human superoxide dismutase in bovine adrenocortical cells. Mutat. Res. 1990;237:95-106

O’Donnell VB, Azzi A. High rates o f extracellular superoxide generation by cultured human fibroblasts: Involvement o f a lipid-metabolizing enzyme. Biochem. J. 1996;318:805-812. 152

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. O ffer T, Rosso A , Samuni A. The pro-oxidative activity o f SOD and nitroxide SOD mimics. Faseb J. 2000;14:1215-1223

Omar BA, Gad NM, Jordan MC, et a t Cardioprotection by Co^Zn-superoxide dismutase is lost at high doses m the reoxygenated heart Free Rad. B iol. Med. 1990;9:465-471

Omar BA, McCord JM. The cardioprotective effect o f Mn-superoxide dismutase is lost at high doses in the posdschemic isolated rabbit heart Free Rad. B iol. Med. 1990;9:473-478

Oury TD, Ho Y-S, Piantadosi CA, Crapo JD. Extracellular superoxide dismutase, nitric oxide, and central nervous system 02 toxicity. Proc. Natl. Acad. Sci. USA 1992;89:9715- 9719

Owens SM, Hardwick WC, Blackall D. Phencyclidine pharmacokinetic scaling among species. J Pharmacol Exp Ther. 1987;242:96-101.

Pahlman I, Kankaanranta S, Palmer L. Pharmacokinetics o f tolterodine, a muscarinic receptor antagonist, in mouse, rat and dog. Arzneim Forsch. 2001;51:134-144.

Paine MF. Hrreceptor antagonists. In: Levy RH, Thummel BCE, Trager WF, Hansten PD, Eichelbaum M, eds. Metabolic drug interactions, Philadelphia, PA: Lippincott W illiams & WOkms, 2000;653-659.

Papp-Jambor C, Jaschinski U, Forst H. Cytochrome P450 enzymes and their role in chug interactions. Anaesthesist 2002;51:2-15.

Patel BA, Boudinot FD, Schinazi RF, Gallo JM, Chu CK. Comparative pharmacokinetics and interspecies scaling o f 3'-azido-3’-deoxy-thymidine (AZT) in several mammalian species. J Pharmacobio-Dyn. I990;13r206-2lI.

Patel RP, McAndrew J, Sellak H, et al. Biological aspects o f reactive nitrogen species. Biochim. Biophys. Acta. 1999;14t 1:385-400

Paxton JW, Kim SN, W hitfield LR. Pharmacokinetic and toxicity scaling o f the antitumor agents amsacrine and C I-92I, a new analogue; in mice, rats, rabbits, dogs, and humans. Cancer Res. 1990;50:2692-2697.

Pfeiffer S, Mayer B, Hemmens B. N itric oxide: chemical puzzles posed by a biological messenger. Angew. Chem. hit. Ed. 1999;38:1714-1731.

Philippides A , Husbands P, O’Shea M . Four-dimensional neuronal signaling by nitric oxide: A computational analysis. J. Neurosci. 2000;20(3):I199-1207.

Radi R, Denicola A , Alvarez B, Ferrer-Sueta G, Rubbo EL The biological chemistry o f peroxynitrite. h r Ignarro LJ. eds. Nhric Oxide: Biology and Pathobiology 1st Ed., San D iego: Academic Press, San Diego, 2QOO;57-82.

153

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Richter WF, Gallati H, Schiller CD. Animat pharmacokinetics o f the tumor necrosis factor receptor-immunoglobulin fusion protein Ienercept and their extrapolation to humans. Drug Metab Dispos. 1999;27:21-25.

Robbie G, Cfaiou WL. Elucidation o f human amphotericin b pharmacokinetics: Identification o f a new potential factor affecting interspecies pharmacokinetic scaling. Pharm Res. 1998;15:1630-1636.

Rubner M. Ober den enifluss der kdrpergrdsse auf stoff und kraftwechsel. Z Biol. 1883;19:535-562.

Sakai T, Hamada T, Awata N, Watanabe J. Pharmacokinetics o f an antiallergic agent, l- (2-ethoxyethyl)-2-(hexahydro-4-methy1-1H -1,4-diazepin-1 -y 1)-1 H-benzimidazoIe difumarate (KG-2413) after oral administration: Interspecies differences in rats, guinea pigs and dogs. J Pbarmacobio-Dyn. 1989;12:530-536.

Sangalti L, Bortolotti A , Jiritano L, Bonati M. Cyclosporine pharmacokinetics in rats and interspecies comparison in dogs, rabbits, rats, and humans. Drug Metab Dispo. 1998;16:749-753.

Sanwald-Ducray P, Dow J. Prediction o f the pharmacokinetic parameters o f reduced- dolasetron in man using in vitro-in vivo and interspecies aUometric scaling. Xenobiotica. 1997;27:189-201.

Savill NJ, W eller R, Sherratt JA. Mathematical modeling o f nitric oxide regulation o f rete peg formation m psoriasis. J. Theor. Biol. 2002;214:1-16.

Sawada Y , Hanano M , Sugiyama Y , Iga T. Prediction o f disposition o f beta-lactam antibiotics in humans from pharmacokinetic parameters in animals. J Pharmacokin Biopharm. 1984;12:241-261.

Schmidt-Nielsen K. Scaling: Why Is Animal Size So Important? Princeton, NJ: Cambridge University Press, 1983.

Scott MD, Meshnick SR, Eaton JW. Superoxide dismutase-rich bacteria. Paradoxical increase in oxidant toxicity. J. B ioi. Chem. 1987;262:3640-3645

Segre G, Bianchi E, Zanolo G. Pharmacokinetics o f flunoxaprofen in rats, dogs, and monkeys. J Pharm Sci. 1988;77:670-673.

Shin H-W, George SC. Microscopic modeling ofNO and S-nitrosoglutathione kinetics and transport in human airways. J. Appf. PfaysioL 2001;90:777-788.

Siefert HM, Domdey-Bette A , Henninger K, Hucke F, KohlsdorferC, Stass HH. Pharmacokinetics o f the 8-methoxyqumoIone, moxifloxacin: A comparison in humans and other mammalian species. J Antimicrob Chemother. I999;43 (SuppL B):69-76.

154

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Siegfried MR, Erfaardt J, Rider T, Ma X-L, Lefer AM . Cardioprotection and attenuation o f endothelial dysfimction by organic nitric oxide donors in myocardial ischemia- reperfusion. J. PharmacoL Exp. Ther. 1992;260:668-675

Sies H, Sharov VS, Klotz L-O, Briviba K. Glutathione peroxidase protects against peroxynitrite-mediated oxidations. J. BioL Chem. 1997;272:27812-27817.

Sies H. Glutathione and its role in cellular functions. Free Rad. BioL Med. 1999;27:916- 921.

Singer M A, Morton AR. Mouse to elephant: Biological scaling and Kt/V. Am J Kidney Dis. 2000;35:306-309.

Singer MA. O f mice and men and elephants: Metabolic rate sets glomerular filtration rate. Am J Kidney Dis. 2001;37:164-178.

Smethurst DP, W illiams HC. Power laws: are hopital waiting lists self-regulating? Nature. 2001;410:652-653.

Stamler JS, Lamas S, Fang FC. Nitrosylation: The prototypic redox-based signaling mechanism. CeU, 2001;106:675-683

Stanbro WD. A kinetic model o f the system: Tyrosyl radical-nitrogen oxide-superoxide ion. J. Theor. BioL 1999;197:557-567.

Stanbro WD. Modeling the interaction o f peroxynitrite w ith low-density lipoproteins. L Plasma levels o f peroxynitrite. J. Theor. BioL 2000;205:457-464.

Stanbro WD. Modeling the interaction o f peroxynitrite with low-density lipoproteins. IL Reaction/diffusion model o f peroxinitrite in low-density lipoprotein particles. J. Theor. BioL 2000;205:465-471.

Stanbro WD. Modeling the interaction o f peroxynitrite w ith low-density lipoproteins. QL The role o f antioxidants. J. Theor. BioL 2000;205:473-482.

Stockley IH. Drug interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, London, UK: Pharmaceutical Press, 1999.

Stockley IH . Drug interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, Oxford, UK: Blackwell Scientific Publications, 1991.

Stockley IH . Drug interactions: a source book o f adverse interactions, their mechanisms, clinical importance and management, Oxford, UK: Blackwell Scientific Publications, 1981.

155

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Strogatz SH. Nonlinear dynamics and chaos. Reading M A: Addison-Wesley Publishing Company, 1994.

Szabo C. Pathophysiological roles o f nitric oxide in inflammation.In: Ignarro LJ. eds. N itric Oxide: Biology and Pathobiology 1st Ed., San Diego: Academic Press, 20OO;84l- 872

Tanaka E, Ishikawa A, Horie T. hi vivo and in vitro trhnethadione oxidation activity o f the liver from various animal species including mouse, hamster, rat, rabbit, dog, monkey and human. Human Exp Toxicol. 1999;18:12-16.

Thomas DD, Liu X , Kantrow SP, Lancaster Jr. JR. The biological lifetim e o f nitric oxide: Implications for the perivascular dynamics o f NO and O 2. Proc. Natl. Acad. Sci. USA 200l;98(l)355-360.

Thomson L, T rujillo M, Telleri R, Radi EL Kinetics o f sytochrome c2+ oxidation by peroxynitrite: Implications for superoxide measurements in nitric oxide-producing biological systems. Arch. Biochem. Biophys. 1995;319:491-497.

Thummel KE, Kunze KL, Shen DD. Metabolically-based drug-drug interactions: principles and mechanisms. In: Levy RH, Thummel KE, Trager WF, Hansten PD, Eichelbaum M, eds. Metabolic drug interactions, Philadelphia, PA: Lippincott Williams & W ilkins, 20003-19.

Troy CM, Derossi D, Prochiantz A, Greene LA, Shelanski M L, Downregulation o f Cu/Zn superoxide dismutase leads to cell death via the nitric oxide-peroxynitrite pathway. J.Nerurosci. 1996;16:253-261

Tsunekawa Y, Hasegawa T, Nadai M , Takagi K, Nabeshima T. Interspecies differences and scaling for the pharmacokinetics o f xanthine derivatives. J Pharm Pharmacol. 1992;44:594-599.

Turcotte DL, Rundle JB. Self organized complexity in the physical, biological, and social sciences. Proc Natl Acad Sci USA. 2002;99(suppl. 1)3463-2465.

Turner CP, Toye AM, Jones OTG. Keratinocyte superoxide generation. Free Rad. Biol. Med. 199834(3):401-407.

van Hoogdalem EJ, Soeishi Y, Matsushima EL Higuchi S. Disposition o f the selective a I A-adrenoceptor antagonist tamsulosin in humans: Comparison w ith data from interspecies scaling. J Pharm Sci. 1997;86:1156-1161.

Vaughn MW, Kuo L, Liao JC. Effective diffusion distance o f nitric oxide in the microcirculation. Am. J. Physiol. 1998;274(5):H1705-H1714.

Vaughn MW, Liao JC. Estimation o f nitric oxide production and reaction rates in tissue by use o f a mathematical model. Am. J. PhysioL 1998;274{6):H2163-H2176. 156

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Vedia VL, McDonald B, Reet BT et aL N itric oxide-induced S-nitrosylation o f glyceraldehydes-3-phosphate dehydrogenase inhibits enzymatic activity and increases endogenous ADP-riboxylation. J. BioL Chem. 1992;267:24929-24932.

Vos TA, van Goor H, Tuyt L, et al. Expression o f inducible nitric oxide synthase in endotoxemic rat hepatocytes is dependent on the cellular glutathione status. Hepatofogy. 1999;29(2):421-426.

W alker D K , Ackland M I, James GC, et aL Pharmacokinetics and metabolism o f sildenafil in mouse, rat, rabbit, dog, and man. Xenobiotica. 1999;29:297-310.

Ward KW, Azzarano LM, Bondinell WE, et al. Preclinical pharmacokinetics and interspecies scaling o f a novel vitronectin receptor antagonist. Drug Metab Dispos. 1999;27:1232-1241.

Weber GF, Bruch HP. Pharmacology o f superoxide-dismutase. Pharmazie. 1992;47:159- 167

West GB, Brown JH, Enquist BJ. A general model for the origin o f aUometric scaling laws in biology. Science. 1997;276:122-126.

West GB, Brown JH, Enquist BJ. The fourth dimension o f life : Fractal geometry and aUometric scaling o f organisms. Science. 1999;284:1677-1679.

West GB. The origin o f universal scaling laws in biology. PhysicaA. 1999;263:104-113.

Widman M, Nilsson LB, Bryske B, Lundstrom J. Disposition o f remoxipride in different species. Arzneim Forsch. 1993;43:287-297.

W ink DA, Cook JA, Kim SY, et aL Superoxide modulates the oxidation and nitrosation o f thiols by nitric oxide-derived reactive intermediates. J. B iol. Chem. 1997;272:11147- I I 151

W ink DA, Darbyshire JF, Nims RW, Saavedra JE, Ford PC. Reactions o f the bioregulatory agent nitric oxide in oxygenated aqueous media: Determination o f the kinetics fo r oxidation and nitrosation by intermediates generated in the NO/CK reaction. Chem. Res. ToxicoL 1993;6:23-27.

W ink DA, Nims RW, Darbyshire JF, et al. Reaction kinetics fo r nitrosation o f cysteine and glutathione in aerobic nitric oxide solutions at neutral pH. Insights into the fate and physiological effects o f intermediates generated in the NO/Oz reaction. Chem. Res. ToxicoL 1994;7:519-525.

Wink DA, Vodovotx Y, Laval J, Laval F, Dewhirst MW, M itchell JB. The multifaceted roles o f nitric oxide in cancer. Carcinogenesis. I998;I9(5):711-721.

157

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Yim MB, Chock PB, Stadtman ER. Copper, zinc superoxide dismutase catalyzed hydroxy radical production to rn hydrogen peroxide. Proc. Nad. Acad. Sci. USA 1990;87:5006-5010

Yoshimura M , Kojima J, Ito T, Suzuki J. Pharmacokinetics o f nipradilol (K-351), a andbypertensive agent. L Studies on interspecies variation in laboratory animals. J Pharmacobio-Dyn. 1985;8:738-750.

158

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.