the stereoselective pharmacodynamics of the enantiomers of

189
1 THE STEREOSELECTIVE PHARMACODYNAMICS OF THE ENANTIOMERS OF PERHEXILINE Giovanni Licari B. Health Sc. (Hons) Supervisors: Associate Professor Benedetta C. Sallustio Professor Andrew A. Somogyi University of Adelaide School of Medical Sciences Faculty of Health Science Discipline of Pharmacology April 2013 A thesis submitted for the degree of Doctor of Philosophy

Upload: others

Post on 11-Jan-2022

12 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The stereoselective pharmacodynamics of the enantiomers of

1

THE STEREOSELECTIVE PHARMACODYNAMICS OF

THE ENANTIOMERS OF PERHEXILINE

Giovanni Licari B. Health Sc. (Hons)

Supervisors:

Associate Professor Benedetta C. Sallustio

Professor Andrew A. Somogyi

University of Adelaide

School of Medical Sciences

Faculty of Health Science

Discipline of Pharmacology

April 2013

A thesis submitted for the degree of Doctor of Philosophy

Page 2: The stereoselective pharmacodynamics of the enantiomers of

2

Table  of  Contents  ABSTRACT  ...........................................................................................................................................  5  

ABSTRACTS  AND  PRESENTATIONS  ......................................................................................................  17  

CHAPTER  ONE:  INTRODUCTION  ..........................................................................................................  24  

1.1   CLINICAL  HISTORY  OF  PERHEXILINE  .................................................................................................................  25  

1.2   CORONARY  HEART  DISEASE  ............................................................................................................................  27  

1.2.1   ENDOTHELIAL  DYSFUNCTION  ......................................................................................................................  28  

1.2.1.1   NADPH  OXIDASE  .................................................................................................................................  28  

1.2.2   ANGINA  ..................................................................................................................................................  29  

1.2.3   INCIDENCE  OF  CAD  ...................................................................................................................................  30  

1.2.4   ISCHEMIC  HEART  DISEASE  ..........................................................................................................................  31  

1.3   CELLULAR  ENERGETICS  ..................................................................................................................................  32  

1.3.1   FATTY  ACID  METABOLISM  ..........................................................................................................................  33  

1.3.1.1    THE  CARNITINE  PALMITOYLTRANSFERASE  (CPT)  SYSTEM  ..............................................................................  35  

1.3.1.2    BETA  OXIDATION  ..................................................................................................................................  38  

1.3.2   CARBOHYDRATE  METABOLISM  ....................................................................................................................  40  

1.3.2.1    ATP  DERIVED  FROM  PYRUVATE  ..............................................................................................................  41  

1.3.2.2    CROSS  TALK  BETWEEN  PATHWAYS  ...........................................................................................................  41  

1.3.3      METABOLIC  DYSFUNCTION  DURING  ISCHEMIA  ..............................................................................................  43  

1.3.3.1    MYOCARDIAL  METABOLIC  AGENTS  ...........................................................................................................  44  

1.4   PHARMACODYNAMICS  OF  PERHEXILINE  ............................................................................................................  46  

1.4.1   RACEMIC  PERHEXILINE  ...............................................................................................................................  46  

1.4.1.1   ANTI-­‐INFLAMMATORY  MECHANISM  OF  PERHEXILINE  ....................................................................................  46  

1.4.1.2   PERHEXILINE  IN  HEART  FAILURE  ...............................................................................................................  47  

1.4.1.3   PERHEXILINE  IN  AORTIC  STENOSIS  .............................................................................................................  47  

1.4.1.4   ANTI-­‐ANGINAL  ACTION  OF  PERHEXILINE  .....................................................................................................  48  

1.5   TOXICITY  OF  PERHEXILINE  ..............................................................................................................................  49  

1.5.1   CLINICAL  TOXICITY  ....................................................................................................................................  49  

Page 3: The stereoselective pharmacodynamics of the enantiomers of

3

1.5.2   IN  VITRO  TOXICITY  STUDIES  ........................................................................................................................  50  

1.5.3   IN  VIVO  PERHEXILINE  TOXICITY  STUDIES  ........................................................................................................  51  

1.6        PHARMACOKINETICS  OF  PERHEXILINE  ............................................................................................................  52  

1.6.1   STUDIES  WITH  RACEMIC  PERHEXILINE  ...........................................................................................................  52  

1.6.2   CYTOCHROME  P450S  ...............................................................................................................................  54  

1.6.2.1   CYP2D6  .............................................................................................................................................  55  

1.6.2.2   ALLELES  PRODUCING  NO  FUNCTIONAL  CYP2D6  PROTEIN  .............................................................................  55  

1.6.2.3   ALLELES  PRODUCING  FUNCTIONING  PROTEIN  WITH  DECREASED  ACTIVITY  .........................................................  56  

1.6.2.4   MULTIPLE  CYP2D6  COPIES  .....................................................................................................................  58  

1.6.2.5  CYP2D6  AND  PERHEXILINE  METABOLISM  ......................................................................................................  58  

1.6.3   ANIMAL  MODEL  CYP2D6  POOR  METABOLISM  ..............................................................................................  59  

1.7   CHIRALITY  ..................................................................................................................................................  62  

1.7.1   PHARMACOKINETICS  OF  PERHEXILINE  ENANTIOMERS  .......................................................................................  63  

1.8   AIMS  AND  HYPOTHESES  ................................................................................................................................  66  

CHAPTER  TWO:  METHODS  .................................................................................................................  68  

2.1   (+)-­‐  AND  (-­‐)-­‐PERHEXILINE  PREPARATION  ..........................................................................................................  69  

2.2   ETHICS  ......................................................................................................................................................  71  

2.3   ANIMALS  ...................................................................................................................................................  71  

2.4   PERHEXILINE  DOSAGE  DETERMINATION  AND  ADMINISTRATION  ............................................................................  71  

2.5   TISSUE  PREPARATION  FOR  HPLC  ....................................................................................................................  73  

2.6   MEASUREMENT  OF  (+)-­‐  AND  (-­‐)-­‐PERHEXILINE  IN  PLASMA  AND  TISSUE  ...................................................................  73  

2.6.1   VALIDATION  OF  HPLC  PERHEXILINE  ASSAY  IN  LIVER  HOMOGENATE  AND  PLASMA  .................................................  75  

2.7   MEASUREMENT  OF  OH-­‐PERHEXILINE  IN  PLASMA  AND  LIVER  ................................................................................  78  

2.7.1   VALIDATION  OF  HPLC  OH-­‐PERHEXILINE  ASSAY  IN  LIVER  HOMOGENATE  .............................................................  81  

2.8   NEUTROPHIL  PREPARATION  ...........................................................................................................................  83  

CHAPTER   THREE:   LACK   OF   CLINICALLY   SIGNIFICANT   STEREOSELECTIVITY   IN   THE   INHIBITION   OF   HUMAN  

NEUTROPHIL  SUPEROXIDE  FORMATION  BY  (+)-­‐  AND  (-­‐)-­‐PERHEXILINE  ................................................  85  

INTRODUCTION  .....................................................................................................................................................  88  

Page 4: The stereoselective pharmacodynamics of the enantiomers of

4

MATERIALS  AND  METHODS  .....................................................................................................................................  90  

CHEMICALS  ..........................................................................................................................................................  90  

RESULTS  ..............................................................................................................................................................  93  

DISCUSSION  .........................................................................................................................................................  94  

CHAPTER  FOUR:  METABOLISM  AND  HEPATOTOXICITY  OF  THE  MYOCARDIAL  METABOLIC  AGENT  PERHEXILINE  

IN  SPRAGUE  DAWLEY  AND  DARK  AGOUTI  RATS  ...............................................................................  104  

INTRODUCTION  ...................................................................................................................................................  109  

MATERIALS  AND  METHODS  ...................................................................................................................................  111  

RESULTS  ............................................................................................................................................................  115  

DISCUSSION  .......................................................................................................................................................  117  

CHAPTER   FIVE:   THE   ENANTIOMERS   OF   THE   MYOCARDIAL   METABOLIC   AGENT   PERHEXILINE   DISPLAY  

DIVERGENT  EFFECTS  ON  HEPATIC  ENERGY  METABOLISM  AND  PERIPHERAL  NEURAL  FUNCTION  IN  RATS   130  

INTRODUCTION  ...................................................................................................................................................  134  

METHODS  ..........................................................................................................................................................  136  

RESULTS  ............................................................................................................................................................  140  

DISCUSSION  .......................................................................................................................................................  144  

CHAPTER  SIX:  GENERAL  DISCUSSION  ................................................................................................  163  

BIBLIOGRAPHY  .................................................................................................................................  173  

 

Page 5: The stereoselective pharmacodynamics of the enantiomers of

5

Abstract  

Page 6: The stereoselective pharmacodynamics of the enantiomers of

6

In the past decade there has been growing interest and evidence that altered myocardial

energy metabolism plays a major role in the onset and/or progression of ischaemic heart

disease and heart failure. The anti-anginal agent perhexiline, marketed in Australia as 50:50

racemic mixture of (+)- and (-)-enantiomers, was one of the original metabolic agents

introduced for clinical use in the 1970’s. Its mechanism of action is thought to involve

manipulation of cellular energetics by inhibiting fatty acid metabolism through its interaction

with the carnitine palmitoyltransferase system. This allows for a ‘switch’ in energy towards

utilisation of carbohydrates, resulting in a greater net production of ATP per unit of oxygen

used. However, perhexiline has a narrow therapeutic index. Hence clinical usage requires

careful dosage individualisation using therapeutic drug monitoring in order to minimise

adverse effects such as peripheral neuropathy and hepatotoxicity associated with elevated

plasma perhexiline concentrations. There are currently no studies investigating the toxicity

and efficacy of the individual enantiomers of perhexiline in vivo and my aim was to address

this considerable gap in knowledge.

Study 1: This study investigated the effects of (+) and (-)-perhexiline on superoxide formation

by NADPH oxidase in intact neutrophils from healthy subjects and patients with symptomatic

stable angina pectoris.

(-)-Perhexiline showed a greater inhibition of superoxide formation than (+)-perhexiline

(P<0.05, Wilcoxon matched paired test) in neutrophils from 11 healthy volunteers, with

median IC50 values of 1.19 and 1.64µM, respectively. In 6 patients with angina, neutrophil

superoxide formation was also significantly inhibited by both (+)- and (-)-perhexiline with

IC50 values of 2.07µM (1.04-2.95µM) and 1.35µM (1.08-1.86µM) respectively; however

there was no significant difference between the two enantiomers.

Page 7: The stereoselective pharmacodynamics of the enantiomers of

7

This study demonstrates that both enantiomers dose dependently inhibit superoxide formation

by neutrophil NADPH oxidase and are both likely to contribute to the beneficial anti-oxidant

properties of perhexiline.

Study 2: This study compared female Dark Agouti (DA) and Sprague Dawley (SD) rats, as

models of CYP2D6 metabolism and hepatotoxicity after administration of perhexiline.

Median (range) perhexiline concentrations in liver and plasma of the SD strain (n=4) were

5.42 (0.92 – 8.22) µg/g and 0.09 (0.04 – 0.13) mg/L, respectively. The DA strain (n=4)

showed higher plasma (0.50 (0.16 – 1.13) mg/L, P<0.05) and liver (24.47 (9.40 – 54.70) µg/g,

P<0.05) perhexiline concentrations, and cis-OH-perhexiline concentrations in plasma and

liver of DA rats were 2.5 and 3.7 fold higher compared to SD rats. In both strains the plasma

concentration ratio of (+):(-) enantiomers was approximately 2:1.When compared to their

controls, DA rats treated with perhexiline had significantly higher plasma LDH

concentrations (2-fold, P<0.05), whilst there were no changes in biochemical liver function

tests in the SD group.

Study 3: The aim of this study was to administer the enantiomers of perhexiline to the DA

animal model. The hypothesis was that there is enantioselectivity in the toxic effects

associated with perhexiline in a clinical setting.

Racemic perhexiline did not affect hepatic lipid or glycogen content. However, (-)-

perhexiline induced a concentration-dependent increase in glycogen content, while (+)-

perhexiline induced concentration-dependent increases in lipid and decreases in glycogen

content. Racemic (p<0.001) and (+)-perhexiline (p<0.05) induced peripheral nerve

dysfunction, while (-)-perhexiline induced no change. Thus, (+)-perhexiline, but not (-)-

Page 8: The stereoselective pharmacodynamics of the enantiomers of

8

perhexiline displayed the toxicity (hepatic steatosis and neuropathy) associated with clinical

use. Mean (s.e.m.) plasma perhexiline concentrations in rats treated with racemate, (+)- or (-)-

enantiomer were 0.84 (0.40), 0.67 (0.07) and 0.29 (0.04) mg/L, respectively, corresponding to

the mid-upper clinical therapeutic range.

To my knowledge the studies performed throughout this thesis are the first to test the anti-

inflammatory as well as toxic effects of the individual enantiomers of perhexiline. The results

suggest that the administration of racemic perhexiline is indeed the administration of two

drugs with very different pharmacokinetic as well as pharmacodynamics properties. This not

only helps us breach the gap in the current knowledge on how racemic perhexiline may exert

its toxic effects, but also introduces the realistic possibility that the use of an enantiomeric

formulation of perhexiline may increase efficacy or decrease toxicity providing a safer

alternative to racemic perhexiline use.

Page 9: The stereoselective pharmacodynamics of the enantiomers of

9

Declaration

Name: ........................................................ Date: ...............................

I certify that this work contains no material which has been accepted for the award of any

other degree or diploma in any university or other tertiary institution to John Licari and, to the

best of my knowledge and belief, contains no material previously published or written by

another person, except where due reference has been made in the text. I certify that no part of

this work will, in the future, be used in a submission for any other degree or diploma in any

university or other tertiary institution without the prior approval of the University of Adelaide

and where applicable, any partner institution responsible for the joint-award of this degree

I give consent to this copy of my thesis when deposited in the University Library, being made

available for loan and photocopying, subject to the provisions of the Copyright Act 1968.

I also give permission for the digital version of my thesis to be made available on the web, via

the University’s digital research repository, the Library catalogue, the Australasian Digital

Theses Program (ADTP) and also through web search engines, unless permission has been

granted by the University to restrict access for a period of time.

Signature: ................................................... Date: ...........................

 

Page 10: The stereoselective pharmacodynamics of the enantiomers of

10

Acknowledgements

I would like to deeply express my gratitude and appreciation to my principal supervisor

Associate Professor Benedetta Sallustio for her support and guidance through out my

candidature. Her dedication and commitment to her students is above and beyond her call of

duty. Her hard work and many publications is a testament to her professionalism and wisdom.

Thank you Betty, without your patience and guidance throughout the years, this thesis would

still be a long way from completion.

Thank you to Professor Andrew Somogyi for support throughout my candidature. His

knowledge and consideration to detail has made it possible to complete this work.

I would like to thank other staff of the Discipline of Pharmacology at The University of

Adelaide, their support and advice that has been instrumental in preparing me for the many

experiments undertaken throughout my candidature.

I would like to give a special thank you to Professor Ray Morris, head of the Clinical

Pharmacology Department at The Queen Elizabeth Hospital for his support throughout my

candidature. Professor Morris is not only an exceptional pharmacologist but also a fantastic

human being whose diplomacy and advice has made my stay at The Queen Elizabeth

Pharmacology Department a most enjoyable and memorable experience.

To my fellow PhD candidates, it has been an honour and pleasure knowing and working with

all of you. I will never forget the friendships and laughs we had during my candidature.

Page 11: The stereoselective pharmacodynamics of the enantiomers of

11

I would like to thank all the staff of the Cardiology Department at The Queen Elizabeth

Hospital. In particular Professor John Horowitz for the many evenings we shared talking

about perhexiline over a cup of coffee. Professor John Beltrame for his support in helping me

acquire a position as a Research Assistant. A thank you to Dr Doan Ngo whose bubbly and

yet ‘take charge’ attitude has helped me stay on track towards completing my PhD, with a

smile.

To all the staff at The Queen Elizabeth animal house, thank you for your technical expertise

and advice that has been instrumental in helping complete all my studies.

I would also thank all the staff of the Clinical Pharmacology Department at The Queen

Elizabeth Hospital for their constant support and infinite patience through my candidature, in

particular Stace Kalaitsidis, Fiona Wicks and Dr Ian Westley for all their advice and technical

support.

I wish to acknowledge and thank all the patients and healthy volunteers who gave up their

time to participate in my studies.  

To all my friends, thank you for your constant emotional support.

Last but not least I would like to thank my family. To my parents, Natale and Connie, for

their love, emotional and financial support and for always believing in me. I will never

underestimate my father’s self-sacrifice, leaving his home country and family behind so I

could receive an education. I also sincerely thank my brother Pat and sister Antonella for

always being there. Last but certainly not least, I sincerely thank my wonderful wife Simone,

whose love, dedication, support, encouragement, patience and understanding has been saintly

Page 12: The stereoselective pharmacodynamics of the enantiomers of

12

throughout the years and without whom I don’t think I would have been able to finish this

thesis. Late in the thesis preparation, we welcomed our son, Natale, whose birth has made me

the happiest person in the world and inspired the completion of this thesis.

Page 13: The stereoselective pharmacodynamics of the enantiomers of

13

Manuscript 1

Lack of Clinically significant Stereoselectivity in the Inhibition of Human Neutrophil

Superoxide Formation by (+)- and (-)-Perhexiline

Licari G __________________________________

Study conception and design, management and interpretation of the data, manuscript

preparation, critical revision of the manuscript.

Kennedy JA __________________________________

Study design

Horowitz JD __________________________________

Study design, interpretation of data, critical revision of manuscript.

Sallustio BC __________________________________

Study conception and design, management and interpretation of the data, manuscript

preparation, critical revision of the manuscript.

Page 14: The stereoselective pharmacodynamics of the enantiomers of

14

Manuscript 2

Metabolism and Hepatotoxicity of the Myocardial Metabolic Agent Perhexiline in Sprague

Dawley and Dark Agouti Rats

Licari G __________________________________

Study conception and design, management and interpretation of the data, manuscript

preparation, critical revision of the manuscript.

Milne RW __________________________________

Study design

Somogyi AA __________________________________

Interpretation of data, critical revision of manuscript.

Sallustio BC __________________________________

Study conception and design, management and interpretation of the data, manuscript

preparation, critical revision of the manuscript.

Page 15: The stereoselective pharmacodynamics of the enantiomers of

15

Manuscript 3

The enantiomers of the myocardial metabolic agent perhexiline display divergent effects on

hepatic energy metabolism and peripheral neural function in rats

Licari G __________________________________

Study conception and design, management and interpretation of the data, manuscript

preparation, critical revision of the manuscript.

Milne RW __________________________________

Study design, critical revision of manuscript

Somogyi AA __________________________________

Interpretation of data, critical revision of manuscript.

Sallustio BC __________________________________

Page 16: The stereoselective pharmacodynamics of the enantiomers of

16

Study conception and design, management and interpretation of the data, manuscript

preparation, critical revision of the manuscript.

 

Page 17: The stereoselective pharmacodynamics of the enantiomers of

17

Abstracts  and  Presentations    

J Licari, AA Somogyi, J Pierides and BC Sallustio. The effects of the enantiomers of

perhexiline on neutrophil superoxide formation. Presented at: 42nd Australasian Society of

Clinical and Experimental Pharmacologists and Toxicologists. Brisbane. 2008

J Licari, AA Somogyi, J Pierides and BC Sallustio. Comparison of Sprague Dawley and Dark

Agouti rats as animal models of perhexiline-induced hepatotoxicity. Presented at: 16th North

American Regional International Society for the Study of Xenobiotics. October 18-22

Maryland Baltimore, USA. 2010.

This work has been subject to intellectual property and as such two provisional patent

applications were lodged in September 2012. Australian patent No. 2012903850 and US

patent No. 61697214: Use of enantiomers of perhexiline. As a result of the pending

intellectual property over this thesis I was not able to submit any papers for publication at this

time.

Page 18: The stereoselective pharmacodynamics of the enantiomers of

18

List of abbreviations

ACC Acetyl-CoA carboxylase

AGPAT Acylglycerol-phosphate acyltransferase

ANOVA Analysis of variance

AUC Area under the curve

ATP Adenosine triphosphate

ADP Adenosine diphosphate

CAT Carnitine acyltransferase

CHD Coronary Heart Disease

CI Confidence interval

CO2 Carbon dioxide

CPT Carnitine palmitoyl-transferase

CV Coefficient of variation

CYP Cytochrome P450 enzyme

D Dose

DA Dark Agouti

DGAT Diacylglycerol acyltransferase

EM Extensive Metaboliser

eNOS Endothelial nitric oxide synthase

ETC Electron transport chain

FA Fatty acid

FACoA Fatty acyl CoA

FACS Fatty AcylCoA synthase

FAD Flavin adenine dinucleotide

FADH2 Reduced form of FAD

FFA Free fatty acids

Page 19: The stereoselective pharmacodynamics of the enantiomers of

19

GLUT-1 Glucose transporter -1

GLUT-4 Glucose transporter 4

G-6-P Glucose -6-phosphate

GPAT Glycerol-phosphate acyltransferase

HepG2 Hepatoma cell line

HK Hexokinase

HLM Human liver microsomes

HPG Hydroxyphenylglyoxylate

HPLC High performance liquid chromatography

IC Inhibitory constant

IM Intermediate metaboliser

IS Internal standard

Km Michaelis-Menten constant

LCFA Long chain fatty acids

LDH Lactate dehydrogenase

n/a Not available

NAD nicotinamide adenine dinucleotide

NADH reduced form of NAD

NO Nitric oxide

NOS Nitric oxide synthase

NYHA New York Heart Association

O2- Super oxide

ONOO - Peroxynitrate

OH Hydroxy

PAP Phosphatidic acid phosphohydrolase

PDH Pyruvate dehydrogenase

Page 20: The stereoselective pharmacodynamics of the enantiomers of

20

PHR Peak height ratio

PM Poor metaboliser

PMA Phorbol myristate acetate

PX Perhexiline

ROS Reactive oxygen species

SD Sprague Dawley

TCA Tri-carboxylic acid

TDM Therapeutic drug monitoring

TQEH The Queen Elizabeth Hospital

UM Ultra-rapid metaboliser

Page 21: The stereoselective pharmacodynamics of the enantiomers of

21

List of Figures

Chapter 1

Figure 1. Chemical structure of perhexiline. The star refers to the chiral centre of the

molecule.

Figure 2. The energy metabolics of the myocardial cell. Red arrows demonstrate the pathway

FA undertake to eventually lead to the production of ATP. Blue arrows indicate the pathway

taken by carbohydrates eventually leading to the production of ATP.

Figure 3. The fate of FA as they enter the myocyte. Free FA are converted to FACoA by

FACS. At this point it can either undergo β-oxidation, eventually leading to the production of

ATP via the ETC, or it can divert to the storage pathway leading to the eventual formation of

triglycerides via enzymatic processes. FA-Fatty Acid, FACS-Fatty AcylCoA synthase,

FACoA- Fatty AcylCoA, GPAT- Glycerol-phosphate acyltransferase, AGPAT- Acylglycerol-

phosphate acyltransferase, PAP- phosphatidic acid phosphohydrolase, DGAT- Diacylglycerol

acyltransferase, ETC- Electro Transport Chain, ATP- Adenosine Tri-Phosphate

Figure 4. Illustration on transport of FACoA to the mitochondrial membrane. The CPT

system consisting of CPT-1, located on the outer mitochondrial membrane and CPT-2 located

on inner mitochondrial membrane, aids in the passage of fatty acyl CoA into the mitochondria

via the addition of a carnitine group.

Figure 5. Metabolism of FACoA. In the mitochondrial matrix FACoA, undergoes a series of

enzymatic processes to produce acyl-CoA, the entry substrate of the citric acid cycle. During

Page 22: The stereoselective pharmacodynamics of the enantiomers of

22

this process FAD and NAD are oxidised to FADH2 and NADH. The final step involves the

addition of a CoA molecule to form acyl-CoA.

Figure 6. This is a diagrammatic representation of the cross talk between the two major

pathways for energy metabolism. The products of β-oxidation (NADH and Acetyl-CoA) have

a negative feedback on the activation of PDH, thus inhibiting the conversion of glucose to

energy and favouring FA utilisation as the preferred energy source. Similarly PDH regulates

its own activation in a negative manner as a result of excessive pyruvate conversion to

Acetyl-CoA.

Figure 7. The major metabolic pathway for perhexiline and formation of OH- perhexiline

metabolites.

Figure 8. CYP2D6 phenotype definition and distribution in a random European population

(N=316). The numbered arrows indicate the range of the sparteine metabolic ratio associated

with number of functional genes. Taken from Zanger et al.

Figure 9. The mono- and di- hydroxyl metabolites resulting from perhexiline metabolism.

Page 23: The stereoselective pharmacodynamics of the enantiomers of

23

Chapter 2

Figure 10. The preparation and resolution of the (+) and (-) enantiomers of perhexiline from

racemic perhexiline maleate

Figure 11. Sample chromatogram from the method described. As shown on the

chromatogram, the retention time for the internal standard was 8.5 min, the (+) enantiomer

was 21.6 min, and the (-) enantiomer was 22.7 min.

Figure 12. Sample chromatogram for the method described. The retention times of the

internal standard (fendiline) a) are 18.8 min, and the OH perhexiline metabolites cis- b) 10.8

min trans-1- c) 12.4 and trans-2- d) 13.6 min respectively.

Figure 13. Various phases following centrifugation of the cellular fraction of blood through

the Lymphoprep® medium.

 

Page 24: The stereoselective pharmacodynamics of the enantiomers of

24

Chapter  One:  Introduction    

Page 25: The stereoselective pharmacodynamics of the enantiomers of

25

1.1 Clinical History of Perhexiline

Perhexiline [(±)-2-(2, 2-dicyclohexylethyl) piperidine] (Figure 1) was developed by

Richardson-Merrell Pharmaceuticals in the late 1960s [1] and was first introduced for clinical

use in the treatment of exertional angina in the 1970s [2]. Perhexiline was recognised as one

of the more effective anti-anginal treatments at the time [3], with additional desirable effects

which facilitated its use including no negative inotropic effects on blood pressure or heart

rate. Despite all the positive outcomes from taking the drug, there were severe adverse events

reported in the late 1970s to mid 1980s, particularly hepatotoxicity and neurotoxicity, that

eventually resulted in its withdrawal from the market [4]. However, it was still prescribed in

other countries, especially Australia and New Zealand, for the treatment of refractory angina

but only when therapy was accompanied by therapeutic drug monitoring, initially to identify

the phenotype of the patient, but also to maintain plasma perhexiline concentrations within a

narrow therapeutic range associated with efficacy and lack of significant toxicity [3].

A double-blinded cross-over trial, conducted in the 1970s showed that perhexiline reduced the

effects associated with exercise-induced tachycardia in healthy volunteers [5] and was also

responsible for reducing the occurrence of anginal symptoms in patients with a history of

coronary heart disease. In so doing, the frequency of nitroglycerine tablet intake was noted to

reduce. Clinically, perhexiline displays improved oxygen utilization, no increase in heart rate

or blood pressure as well as improvement in myocardial oxygen consumption [5, 6].

In the late 1980s after the discovery that perhexiline toxicity was associated with elevated

concentrations in plasma [7], a double blind placebo-controlled cross-over clinical study

revealed the efficacy of perhexiline in patients who were already receiving maximal tolerated

pharmacological therapy [8]. To participate in the study, all patients needed to have

Page 26: The stereoselective pharmacodynamics of the enantiomers of

26

diagnostic evidence of coronary artery disease or a history of myocardial infarction as well as

being unsuitable for revascularisation procedures. All patients included were on maximal

tolerated doses of combinations of β-blockers, nitrates, calcium antagonists or all three. The

results showed, as in in the earlier trials, perhexiline was associated with little change in

hemodynamic effects, but did produce an increase in myocardial efficiency and a decrease in

the frequency of angina attacks [5, 6, 8]. This trial confirmed that by titrating doses to keep

plasma concentrations below 0.6mg/L, perhexiline not only shows efficacy in the absence of

any adverse effects but could be used safely in combination with conventional anti-anginal

agents without having to drastically alter the current dosing regimen, providing there is strict

therapeutic drug monitoring.

The treatment of stable and refractory stable angina was and still is the primary indication for

perhexiline; however, more recently it has been shown to be efficacious in the treatment of

other forms of cardiovascular disease such as atherosclerosis [9], endothelial dysfunction [9,

10], unstable angina [11], aortic valve disease [12] and heart failure [13], the mechanisms of

which will be presented further on in the chapter.

Page 27: The stereoselective pharmacodynamics of the enantiomers of

27

Figure 1. The chemical structure of perhexiline. The star refers to the chiral centre of the

molecule.

1.2 Coronary heart disease

Within the past century, according to the Australian Institute of Health and Welfare (AIHW)

cardiovascular disease has grown to become the most prevalent disease in the world today

[14, 15]. Coronary heart disease (CHD) is the most prevalent form of cardiovascular disease.

It is a culmination of a series of events that present as a result of diseased coronary

vasculature, which is also commonly referred to as coronary artery disease (CAD). In most

cases, CAD clinically manifests in the form of angina and/or a myocardial infarction (MI),

which are primarily due to impedance in coronary blood flow to the myocardium as a result of

the formation of atherosclerotic plaques within the vascular arterial wall itself.

78 H. ASHRAFIAN ET AL.

FIG. 1. The chemical structure of perhexiline.

carboxyl groups, it had been proposed by some investigators, based purely on chemical andstructural considerations, that reminiscent of this maleate interaction, perhexiline’s putativebiological targets might also have a group equivalent to two oxygen atoms separated by4.49 A

!

. However, attempts to predict perhexiline’s function based on structural similaritieswith other molecules (e.g., the histamine H1 receptor antagonist—carbinoxamine) have notproved insightful due the dissimilarity of even these seemingly similar molecules (Dawsonet al. 1986).

Pharmaceutical preparations of perhexiline contain a racemic mixture of (+) and (–)enantiomers. As will be discussed, perhexiline is metabolized to one of its six monohy-droxy metabolites or its dihydroxy metabolites. These include a cis, trans-1, and trans-2monohydroxy variant for each enantiomer. The predominant metabolites are the cis-4-axialand to a lesser extent the trans-4 equatorial monohydroxyperhexiline diastereoisomers.These are depicted in Figure 2 (Davies et al. 2006a, 2006b).

PHARMACOLOGY

Mechanism of Action (Table 1)

Interest in the development of perhexiline was aroused by the physiological success ofthe related compound hexadylamine. Although principally investigated in isolated perfusedheart preparations, hexadylamine was noted to exhibit vasodilation effects. Specifically,hexadylamine’s efficacy as a coronary vasodilation was the major impetus for the eval-uation of its derivatives as antianginal agents. Although the mechanism of vasodilationwas never fully delineated, it was noted that despite the increased cardiac output, cardiacwork remained constant in the context of a reduced cardiac oxygen extraction. In effect,hexadylamine increased cardiac oxygen efficiency (Rowe et al. 1963). Perhexiline wassimilarly extensively studied using pharmacological techniques in animals (Hudak et al.1973).

Cardiovascular Drug Reviews, Vol. 25, No. 1, 2007

*

Page 28: The stereoselective pharmacodynamics of the enantiomers of

28

1.2.1 Endothelial Dysfunction

Endothelial dysfunction has been increasingly regarded as the underlying cause of

atherosclerosis, diabetes, hypertension and heart failure. It is characterised by an imbalance of

nitric oxide (NO) availability to the endothelial wall, which results in endothelium-dependent

vasodilation impairment. NO is produced by NO synthase (NOS) which is specific to the

environment where it is located, for example NO produced in the endothelium is synthesised

by endothelial NOS (eNOS) [16]. Free radicals, such as the reactive oxygen species (ROS) in

particular the superoxide anion (O2-) have been shown to react with NO reducing its

bioavailability and therefore reducing its vascular effects. It undergoes an extremely rapid

reaction with nitric oxide to form peroxynitrite (ONOO-), which is a potent oxidising agent in

its protonated form, peroxynitrous acid.

Chemical reaction for the formation of ONOO-

O2- · + · NO → ONOO –

1.2.1.1 NADPH Oxidase

NADPH oxidase is a multicomplex enzyme which when active is the major source of O2- in

the cardiovascular system. The complex itself is made up of a flavocytochrome b b558 unit

comprising of two membrane bound subunits p22phox (the oxidase) and gp91phox (also known

as Nox 2), responsible for the stability and the activity of the enzyme, and at least four

cytosolic subunits p47phox, p67phox, p40phox and Rac 1 or 2 which when activated translocate

the membrane and interact with the flavocytochrome b b558 unit [17, 18]. Upon activation by

Page 29: The stereoselective pharmacodynamics of the enantiomers of

29

various stimuli such as angiotensin II, inflammatory cytokines or hypoxia, the oxidase uses

NADPH as an electron donor to generate O2- from molecular oxygen [19] .

There have been 4 isoforms of Nox identified (Nox 1-5), which are encoded by separate genes

and have differential expression throughout the cardiovascular system. The predominant

isoforms in cardiomyocytes [20], endothelial cells [21] and fibroblasts [22] are Nox 2 and 4,

whereas in vascular smooth muscle cells Nox 1 and 4 are preferentially expressed [23].

1.2.2 Angina

Angina pectoris is a result of the classical situation of supply and demand. It occurs when the

demand for oxygen exceeds the rate at which blood is able to deliver it to the myocardium.

The supply is greatly affected in patients suffering from CAD as they are at greater risk from

developing atherosclerosis, the major contributor of ischaemic heart diseases.

Heavy feeling, tightness, sharp stabbing pain or squeezing of the chest are characteristic

symptoms associated with angina presentation. The pain experienced in angina is one of

gradual increase that can last from a few minutes to thirty minutes. The pain experienced is

not localized to the heart, but may be referred pain that can radiate to the shoulder, arms,

neck, throat, jaw and teeth [24].

Patients suffering from angina are classified as having stable, unstable or variant forms

according to the degree of impairment the disease has on physical activity.

The clinical presentation of stable angina is commonly associated with chest pain that is short

in duration (typically less than 15 minutes). The onset of chest pain does not occur while at

Page 30: The stereoselective pharmacodynamics of the enantiomers of

30

rest, but becomes progressively worse upon exertion. Rest or nitroglycerine therapy usually

relieves this pain [25].

In contrast to stable angina, unstable angina manifests itself when at rest, as unpredictable

chest pain often of lengthy duration, Upon examination, patients suffering from unstable

angina often present with transient ischemic, ST-segment and T-wave abnormalities upon

ECG analysis. Patients suffering from unstable angina are at high risk of the disease

progressing to MI. The mechanism underlying the development of unstable angina is a

atherosclerotic plaque rupture accompanied by thrombus formation [25].

As the name suggest, the onset of variant angina is unpredictable. The pain associated with

this angina syndrome can occur either at rest or during normal physical activity, but is not

brought about by exercise. The frequency of variant angina attacks seems to also be time

dependent, with ischemic episodes manifesting with mild exercise in the early morning,

whilst other episodes occur at rest between midnight and early morning. In a patient suffering

from variant angina, ECG analysis commonly reveals ST-segment elevation with reciprocal

depression. The mechanism underlying variant angina is believed to be due to coronary artery

spasm that is an effect of endothelial dysfunction [26, 27].

1.2.3 Incidence of CAD

CHD is the most common form of heart disease in Australia. The 2001 national health survey

revealed that approximately 1.9% (approx. 355,600) of Australians suffered some form of

CHD, and of these, 266,700 suffered from angina and the remainder reported having a MI

[15]. CHD is the largest single cause of death in Australia, accounting for 26,063 deaths in

2002. However, data collected over a 12-year period between 1991 until 2002 have indicated

Page 31: The stereoselective pharmacodynamics of the enantiomers of

31

that death rates have declined by approximately 41% in males and 40% in females [15]. The

survey data also revealed that hospitalisation as a result of CHD was greatest in Australians

who were 60 years of age and over, and hospitalisation rates had increased by 11.7% between

1993-94 and 2001-02 [15].

These findings were further explained by the international Monitoring Trends and

Determinants in Cardiovascular Disease (MONICA) project. This was a 10-year study that

monitored cardiovascular disease in 37 populations over 21 countries (including Australia)

and its aim was to establish factors affecting mortality and morbidity rates. The main findings

were that a decrease in mortality was largely determined by interventions that affected and

changed coronary event rates [28].

It has been suggested, based on trends in the early 2000s, that by 2020 CHD will be the single

leading health problem of the world [15].

1.2.4 Ischemic Heart Disease

Coronary Artery Disease (CAD) is one of the major contributors of myocardial ischemia.

Myocardial ischemia is defined as a reduction in coronary blood flow such that there is an

imbalance between the body’s oxygen supply and demand [29]. As a result of this imbalance,

myocardial function and in particular energy metabolism, is greatly reduced. In order to

restore balance, myocardial function and energy metabolism have been the target of many

therapeutic interventions.

Page 32: The stereoselective pharmacodynamics of the enantiomers of

32

1.3 Cellular Energetics

Reduction of coronary blood flow will affect oxygen supply and demand and as such will be

the determining factor for whether the cell will utilise carbohydrates or fatty acids (FA) as the

primary source of energy (Figure 2).

Figure 2. The energy metabolics of the myocardial cell. Red arrows demonstrate the pathway

FA undertakes to eventually lead to the production of ATP. Blue arrows indicate the pathway

taken by carbohydrates eventually leading to the production of ATP. CPT= carnitine

palmitoyl-transferase, PDH= pyruvate dehydrogenase, GLUT= glucose transporter, TCA=

tricarboxylic acid cycle, ETC= electron transport chain, HK= hexokinase

Page 33: The stereoselective pharmacodynamics of the enantiomers of

33

1.3.1 Fatty Acid Metabolism

FA metabolism is a complex process involving many enzymes. It begins with the transport of

FA to the heart as albumin-bound FA via the blood stream. Once in the myocyte, free FA and

acyl-CoA synthetase together with ATP, catalyse the formation of long-chain fatty acyl-CoA

(LCFA). The LCFA can undertake one of two pathways, they can be converted to

triglycerides by lipoprotein lipase as an energy storage option, or they can undergo β-

oxidation in the mitochondria leading to the production of ATP (see Figure 3) [30, 31].

LCFA is impermeable to the mitochondrial membrane. In order to facilitate its uptake, the

mitochondria have evolved a carnitine palmitoyl-transferase system, involving the enzymes

carnitine palmitoyl-transferase (CPT)-1 and -2 and carnitine acyltransferase (CAT) [32, 33].

CPT-1 esterifies acyl-CoA to carnitine to form long-chain acyl-carnitine in the outer

membrane. It is transported from the outer mitochondrial membrane by carnitine acyl-

transferase (CAT), to the inner mitochondrial membrane where it is converted back to LCFA

and carnitine by CPT-2.

Page 34: The stereoselective pharmacodynamics of the enantiomers of

34

Figure 3. The fate of the fatty acid as it enters the myocyte. Free FA is converted to FACoA

by FACS. At this point it can either undergo β-oxidation, eventually leading to the production

of ATP via the ETC, or it can divert to the storage pathway leading to the eventual formation

of triglycerides via enzymatic processes. FA-Fatty Acid, FACS-Fatty AcylCoA synthase,

FACoA- Fatty AcylCoA, GPAT- Glycerol-phosphate acyltransferase, AGPAT- Acylglycerol-

phosphate acyltransferase, PAP- phosphatidic acid phosphohydrolase, DGAT- Diacylglycerol

acyltransferase, ETC- Electro Transport Chain, ATP- Adenosine Tri-Phosphate

Page 35: The stereoselective pharmacodynamics of the enantiomers of

35

1.3.1.1 The Carnitine Palmitoyltransferase (CPT) System

This system is one that has evolved to facilitate the entry of free FA into the mitochondrial

matrix, via a series of enzymes. The first enzyme encountered by free FA is CPT-1, the key

and rate limiting enzyme involved in the transfer of fatty acyl-CoA into the mitochondria,

located on the outer mitochondrial membrane [32, 34]. Here the fatty acyl groups undergo

esterification with carnitine to form fatty acyl-carnitine, which is then able to cross the outer

membrane where it reaches the inner mitochondrial membrane. Acylcarnitine-carnitine

translocase facilitates the entry of fatty acyl-carnitine into the matrix where it is converted

back to fatty acyl-CoA and carnitine by CPT-2 [31, 33, 35] (Figure 4).

CPT-1 is a single polypeptide, 88 kDa in size and has both inhibitor and catalytic domains.

Studies have found that CPT-1 consists of two hydrophobic domains H1 and H2. The

orientation of positively and negatively charged residues in the H1 domain, suggests that the

NH2 terminus faces the cytosol [36]. Expression of a truncated NH2 terminus of CPT-1

involving yeast studies, showed a decrease in the activity of the CPT-1 regulator malonyl-

CoA [37], suggesting a pivotal role of the cytosolic NH2 terminus in the regulation of CPT-1

[38].

CPT-2 is a 71kDa protein containing a 25 amino acid NH2 terminal. It is located inside the

inner mitochondrial membrane and its function is to reform the fatty acyl-CoA and carnitine

from fatty acyl-carnitine [38, 39] (Figure 4).

There are two isoforms of CPT-1 found in both human liver and muscle cells and therefore

have been named L-CPT 1 and M-CPT 1 respectively. They have different affinities to

carnitine with the L-CPT 1 having a Km of 30 µM and the M-CPT 1 a Km of 500 µM [40].

Page 36: The stereoselective pharmacodynamics of the enantiomers of

36

Malonyl-CoA is a physiological inhibitor of CPT-1 synthesised in the sarcolemma by acetyl-

CoA carboxylase (ACC), which requires acetyl-CoA as a substrate [41]. Studies have shown

that malonyl-CoA binding to CPT-1 is non-competitive but also has allosteric properties. Rat

liver mitochondria treated with proteases, which specifically digest the region of the protein

responsible for allosteric inhibition without damaging the catalytic centre, have shown a

reduced response to malonyl-CoA while the catalytic site was still active [38, 42, 43]. In

contrast, the administration of trypsin caused inactivation of the catalytic site but little or no

reduction of malonyl-CoA binding.

Page 37: The stereoselective pharmacodynamics of the enantiomers of

37

Figure 4. Illustration of the Carnitine Palmitoyltransferase System consisting of CPT-1,

located on the outer mitochondrial membrane and CPT-2 located on inner mitochondrial

membrane, which aids in the passage of fatty acyl CoA into the mitochondria via the addition

of a carnitine group.

Page 38: The stereoselective pharmacodynamics of the enantiomers of

38

1.3.1.2 Beta Oxidation

In the mitochondria, LCFA undergo the process of β-oxidation. The rate of fatty acid β-

oxidation is dependent upon the availability of the exogenous FA and the energy requirement

of the tissue [32, 44]. β-Oxidation involves the repeated cleavage of two carbon acetyl-CoA

units, as a result of the enzymatic activity of acyl-CoA dehydrogenase, enoyl-CoA hydratase,

L-3-hydroxyacyl-CoA dehydrogenase and 3-ketoacyl-CoA thiolase, leading to the formation

of NADH and FADH2. [31, 44] (Figure 5).

The final step in this process is the entry of acetyl-CoA into the tricarboxylic acid cycle

(TCA), where it is eventually oxidised to form NADH, FADH2 and CO2. NADH and FADH2

are reoxidised by the mitochondrial electron transport chain, thereby generating ATP. The

flux through the TCA is highly regulated by the concentrations of NAD+/NADH and

ADP/ATP as they have potential inhibitory effects on the key enzymes in this cycle [35].

Page 39: The stereoselective pharmacodynamics of the enantiomers of

39

Figure 5. Metabolism of FACoA. In the mitochondrial matrix FACoA, undergoes a series of

enzymatic processes to produce acyl-CoA, the entry substrate of the citric acid cycle. During

this process FAD and NAD are oxidised to FADH2 and NADH. The final step involves the

addition of a CoA molecule to form acyl-CoA.

Page 40: The stereoselective pharmacodynamics of the enantiomers of

40

1.3.2 Carbohydrate Metabolism

Carbohydrates are the cell’s alternate energy source. This source accounts for about 10-40%

of the energy produced in the cell. Entry of glucose in the cell is tightly regulated by enzymes

and is dependent upon factors such as the concentration of transporters and by the

transmembrane glucose gradient [45]. The two main transporters involved in the uptake of

glucose under normal physiological conditions are GLUT 1 and GLUT 4, which are located

on the sarcolemmal membrane and the intracellular microsomal vesicles [46, 47]. These

transporters facilitate the entry of glucose into the cell where it is phosphorylated to glucose-

6-phosphate (G-6-P) by the enzyme hexokinase (HK). G-6-P can either undergo

glycogenolysis and stored as glycogen to be called upon by the cell whenever it is needed, or

it can undergo glycolysis and be broken down to form ATP. Glycogenolysis is regulated by

the amount of G-6-P and indirectly by insulin, which causes the activation of the glycogen

synthetase enzyme [31, 47].

The pathway contributing to the energy production in the metabolism of carbohydrates is that

of glycolysis. G-6-P undergoes a series of enzyme-catalysed reactions. The key enzymes

phosphofructokinase-1, phosphofructokinase-2, and glyceraldehyde 3-phosphate

dehydrogenase all act to convert G-6-P to pyruvate while at the same time producing ATP

molecules [48]. Lactate can also be used as a source of pyruvate in extreme conditions due to

its conversion by lactate dehydrogenase (LDH).

Page 41: The stereoselective pharmacodynamics of the enantiomers of

41

1.3.2.1 ATP Derived From Pyruvate

Pyruvate is transported into the mitochondria where it is converted to acetyl-CoA via

decarboxylation by pyruvate dehydrogenase (PDH) (Figure 6). This is the key to an

irreversible reaction in the metabolism of carbohydrates. The activity of PDH is highly

dependent on its phosphorylation state and amount of substrate available. PDH is in its

inactive form when phosphorylated by a PDH-kinase, whose activity depends upon the

concentration of pyruvate, ADP, acetyl-CoA/CoA and NAD+/NADH. In its active form, PDH

is dephosphorylated by a PDH-phosphatase and its activity is dependent upon Ca2+ and Mg2+

concentrations. Acetyl-CoA then enters the TCA cycle, as previously described in fatty acid

oxidative metabolism [49-51].

1.3.2.2 Cross Talk Between Pathways

FA and glucose metabolism are not entirely independent processes. They require a degree of

inter-relation in order to achieve optimum myocardial performance. In a healthy human heart,

FA utilisation is favoured and therefore is primarily metabolised as a source of energy. β-

Oxidation of FA produces compounds that have an inhibitory effect on carbohydrate

metabolism. The high mitochondrial concentrations of acetyl-CoA/CoA and NAD+/NADH

produced by FA oxidation activate PDH-kinases which then act to phosphorylate and thereby

inhibit PDH, thus putting a halt on the key irreversible reaction in carbohydrate metabolism

(Figure 6) [51, 52].

Page 42: The stereoselective pharmacodynamics of the enantiomers of

42

Figure 6. This is a diagrammatic representation of the cross talk between the two major

pathways for energy metabolism. The products of β-oxidation (NADH and Acetyl-CoA) have

a negative feedback on the activation of PDH, thus inhibiting the conversion of glucose to

energy and favouring FA utilisation as the preferred energy source. Similarly PDH regulates

its own activation in a negative manner as a result of excessive pyruvate conversion to

Acetyl-CoA.

Page 43: The stereoselective pharmacodynamics of the enantiomers of

43

1.3.3 Metabolic Dysfunction During Ischemia

During periods of ischemia, the primary effect on the myocardium is mitochondrial metabolic

dysfunction. As the myocyte prefers to utilise FA as its primary energy source (see section

1.6.1 for more detail), impaired blood flow reduces not only the concentration of free FA

available, but also reduces the supply of oxygen to the myocardium, thereby diminishing the

cells’ ability to undergo aerobic respiration [31, 53, 54]. Under ischemic conditions, the free

FA continue to undergo β-oxidation producing NADH and acetyl-CoA, activators of PDH

kinase, which in turn phosphorylates PDH resulting in its inhibition. The lack of oxygen leads

to a decrease in FA flux through the electron transport chain and TCA cycle, resulting in

decreased ATP formation and an increase in cytosolic acetyl-CoA concentrations resulting in

further inhibition of PDH. As the cell detects a decrease in ATP formation, it switches energy

source to carbohydrates. The lack of blood flow stimulates an increase in the concentration of

the glucose uptake transporters GLUT-1 and -4 on the cell surface in order to facilitate the

transport of more glucose into the cell. Glucose is then converted to pyruvate, which moves

into the mitochondrial matrix to be decarboxylated by PDH [31, 50, 55] (see section 1.6.3 for

further detail). However, PDH is inactive as a result of the NADH and acetyl-CoA produced

by β-oxidation, causing pyruvate concentrations in the cell to rise. Under normal conditions

lactate consumption would serve as an alternate source of pyruvate, however under ischaemic

conditions, the increase of pyruvate causes the cell to go from a state of lactate consumption,

to a state of lactate production via the enzyme LDH. An increase in lactate concentrations

causes a decrease in cellular pH [50, 56].

Page 44: The stereoselective pharmacodynamics of the enantiomers of

44

As ATP concentrations are depleted, lactate concentrations increase and cellular pH decreases

(↑H+), cardiac efficiency also begins to decrease. At low pH the cell is unable to maintain

Ca2+ homeostasis and requires more ATP to do so, leading to a decrease in contractile work,

which also requires ATP [29, 57]. Therefore a reduction in ATP and a decrease in pH will not

only cause the impairment of the Ca2+ pump but also a reduction in the contractile function of

myocardial segments.

1.3.3.1 Myocardial metabolic Agents

In the last two decades, there have been significant advances towards the knowledge and

understanding on the workings of the myocardium. However, despite advances in

haemodynamic pharmacological therapies and surgical interventions, there are still significant

proportions of refractory or relapsing IHD and HF patients. As mentioned earlier in this

chapter, these disease states are characterised by an imbalance in energy production in

myocytes. The myocardium’s ability to metabolise fatty acids is substantially reduced forcing

the switch to its alternate energy source, carbohydrates. However, in severe disease, flux

through this pathway is also severely impaired, potentially resulting in myocyte cell death. It

is not well understood why this switch in substrate utilisation from FFA to carbohydrates

takes place and whether this is an adaptive mechanism for cardio-protection or whether it

contributes to the progression of the disease.

Recently, targeting myocardial energy metabolism has gained significant momentum as a

novel approach in treating HF and IHD, and several studies have shown that the use of

myocardial metabolic agents to modify myocardial substrate utilisation improves left

ventricular ejection fraction (LVEF) [11], while reducing FFA metabolism and increasing

Page 45: The stereoselective pharmacodynamics of the enantiomers of

45

glucose metabolism [58], resulting in a more mechanically efficient, oxygen-sparing

myocardium, characteristics that are deficient in IHD and HF.

Current agents used clinically to treat ischaemic conditions have shown to be metabolic

modulators:

Trimetazidine

Trimetazidine (1-[2,3,4 – trimethoxybenzyl] piperazine dihydrochloride) is believed to exert

its metabolic modulation via inhibition of the enzyme long-chain 3-ketoacyl coenzyme-A

thiolase (LC 3-KAT), one that is involved in the β-oxidation pathway [59]. The advantageous

characteristics of trimetazidine therapy are that it displays no negative inotropic effect either

at rest or upon exertion and there is no need for therapeutic drug monitoring [59].

Etomoxir

Etomoxir is a potent CPT-1 inhibitor that was initially introduced as an antidiabetic agent due

to its hypoglycaemic effects [60]. In a double blind randomized clinical trial, the therapeutic

effects of etomoxir were studied. Although etomoxir displayed a trend for increased exercise

tolerance at various doses, the trial had to be prematurely terminated as there were reports of

elevated liver transaminase levels indicating the potential for hepatotoxicity [61].

Ranolazine

Ranolazine (±-N-(2,6-dimethyphenyl)-4-[2-hydroxy-3-(2-methoxyphenoxy)-propyl]-1-

piperazineacetamide), is currently used for the treatment of chronic stable angina. MARISA

(Monotherapy Assessment of Ranolazine in Stable Angina) was a placebo-controlled

Page 46: The stereoselective pharmacodynamics of the enantiomers of

46

randomized study to assess the efficacy of ranolazine in stable angina. The results showed

that ranolazine increased exercise duration, while increasing the time between angina

episodes without having any negative haemodynamic effects [62]. Ranolazine’s mechanism

of action is not entirely understood, however studies to date have shown that may work via

the inhibition of 3-KAT [63], a mechanism that is similar to that of trimetazidine.

1.4 Pharmacodynamics of Perhexiline

1.4.1 Racemic Perhexiline

Perhexiline is arguably one of the first myocardial metabolic agents used clinically and unlike

other myocardial metabolic agents is clinically available in Australia and New Zealand. It has

been recently reintroduced in the United Kingdom and also gained orphan status in the United

States of America making it an appropriate platform for the development of new myocardial

metabolic agents.

1.4.1.1 Anti-Inflammatory Mechanism of Perhexiline

Perhexiline has been shown to inhibit super oxide (O2-) production via NADPH oxidase [9].

O2- derived from NADPH oxidase in phagocytic and endothelial cells is the major source of

superoxide contributing to endothelial dysfunction. The NADPH oxidase complex is made up

of four major units: two membrane bound subunits p22phox and gp91phox and two cytosolic

components p47phox and p67phox [64]. This preassembled enzyme cannot produce O2- ·, as it

first needs to be activated. This is accomplished by phosphorylation of the p47phox subunit,

which then interacts with the membrane subunits. [64, 65]. In a study on isolated human

neutrophils, perhexiline significantly inhibited superoxide formation via NADPH oxidase by

Page 47: The stereoselective pharmacodynamics of the enantiomers of

47

up to 50% at clinically relevant concentrations. Furthermore, the same group was able to

demonstrate that perhexiline had no effect on the assembly of the enzyme and it was only the

pre-assembled complex that was affected [9].

1.4.1.2 Perhexiline In Heart Failure

There is a growing body of evidence to suggest that heart failure is associated with altered

myocardial energetics. Studies have confirmed that the ratio of the high-energy phosphate

storage molecule, phosphocreatine (PCr) and adenosine triphosphate (ATP), are significantly

diminished in patients with HF, including non-occlusive hypertrophic cardiomyopathy

(HCM) [66].

In a double blind, placebo-controlled study in HF patients, perhexiline significantly improved

myocardial function and increased the PCr:ATP ratio from 1.27± 0.02 to 1.73± 0.02,

significantly improving myocardium energetics [67]. Perhexiline was also recently shown to

improve myocardial energetics and function in patients with HCM, a subtype of HF, for

which there is currently no cure [13]. Thus, perhexiline may be a clinically useful

pharmacotherapy in the management of heart failure.

1.4.1.3 Perhexiline In Aortic Stenosis

Perhexiline has been shown to provide symptomatic relief in elderly patients suffering from

aortic stenosis [12]. Although this study was uncontrolled, Unger et al showed that

administration of perhexiline to elderly patients (mean age 80 years, refractory to treatment

and/or unable to undergo aortic valve replacement, as well as having baseline New York

Heart Association (NYHA) classification scores of 3 or 4) relieved the symptomatic

Page 48: The stereoselective pharmacodynamics of the enantiomers of

48

characteristics of aortic stenosis in 13 of 15 patients over the first 3 months of treatment [12].

This finding has some major clinical implications, as there is currently no alternative to aortic

valve replacement surgery. Perhexiline could provide another option for those patients who

do not want to undertake the risk of open-heart surgery, who are of advanced age and/or

frailty where the risk of surgery outweighs potential benefits, as well as patients whose

disease state does not make them suitable candidates for aortic valve replacement.

1.4.1.4 Anti-anginal Action of Perhexiline

Early on in its history, the mechanism of action associated with perhexiline use was elusive

and it was first described as a calcium, sodium and potassium channel inhibitor [1, 68]. It was

discovered more recently that perhexiline's primary mechanism of action was not the

inhibition of these ion channels, but rather appeared to be associated with effects on cellular

metabolism [69].

It is now considered that perhexiline’s main mechanism of action is the inhibition of β

oxidation via the carnitine palmitoyltransferase -1 (CPT-1) enzyme [1]. A study [69] using

isolated cardiac and hepatic mitochondria (from Sprague Dawley rats) incubated with

perhexiline found IC50 (inhibitory constant) values for perhexiline that were lower for cardiac

(77 µmol/L) than hepatic (148 µmol/L) CPT-1. The only compound that had a lower IC50 was

the naturally occurring malonyl-CoA. Perhexiline inhibition was of a non-competitive nature

with respect to malonyl-CoA and another CPT-1 inhibitor hydroxyphenylglyoxylate (HPG).

When the protease, nagarase, was added to the incubation mixture both malonyl-CoA and

HPG lost their affinity for CPT-1 (nagarase destroyed the binding site on CPT-1) whereas

perhexiline was not significantly affected, suggesting that although perhexiline is a

Page 49: The stereoselective pharmacodynamics of the enantiomers of

49

competitive inhibitor of palmitoyl-CoA, it may bind to an alternate site to that of the

endogenous inhibitor malonyl-CoA.

Although the exact mechanism by which perhexiline inhibits CPT-1 is not conclusively

known, its IC50 values show a wider picture as to how this drug selectively inhibits cardiac

CPT-1, thereby reducing fatty acid metabolism and as a result redirecting myocyte cellular

metabolism towards carbohydrate utilization [10, 70].

1.5 Toxicity of Perhexiline

1.5.1 Clinical Toxicity

The chronic administration of perhexiline where supra-therapeutic concentrations are

maintained in plasma, leads to severe toxic effects, principally peripheral neuropathy and/or

hepatotoxicity in the form of micro- and macro-vesicular steatosis.

Case reports throughout the 1970s and 1980s all associated administration of perhexiline

maleate with peripheral neuropathy in a sub-group of patients treated. In all instances patients

suffered from paraesthesia, muscular weakness and, in severe cases, muscular atrophy, most

of which subsided after cessation of perhexiline [71, 72].

An early study conducted by Morgan et al investigated four patients treated chronically with

perhexiline who had developed liver disease. Three out of the four patients developed micro

nodular cirrhosis with severe hepatitis and steatosis, two of which also showed peripheral

neuropathy. The other patient developed moderate hepatitis with peripheral neuropathy [73].

Although the report states the dosage of perhexiline used in each case, there is no indication

of plasma perhexiline concentrations or those of its primary hydroxy metabolites (see

Page 50: The stereoselective pharmacodynamics of the enantiomers of

50

metabolism below on pg. 51), making it difficult to ascertain where these patients lie in the

therapeutic range.

Other side-effects associated with the use of perhexiline include weight loss and

hypoglycaemia (in diabetics) [73]. The theory as to why only a small sub-population of

perhexiline administered patients suffered from the severe adverse effects was speculative. In

these early studies, the general opinion was that these adverse effects were associated with

elevated plasma perhexiline concentrations due to either: hepatic disease (possibly drug-

induced) leading to impairment in the metabolism of perhexiline, or some genetic variable

that impeded perhexiline metabolism [73, 74]. It was later realised this resulted from impaired

metabolism of perhexiline due to a genetic polymorphism of the CYP2D6 enzyme present in

approximately 7% of the Caucasian population (discussed late in this Chapter).

1.5.2 In Vitro Toxicity Studies

The administration of perhexiline to primary rat hepatocytes demonstrated a significant effect

on β-oxidation. Perhexiline at low concentrations (5 µmol/L) seems to have no significant

effect on ATP production, β-oxidation or triglyceride formation after being incubated for a

24-hour period. However after a 72-hour incubation period, perhexiline significantly

decreased β-oxidation and increased ATP and triglyceride formation with no significant effect

on oxygen consumption or cell death when compared to control [75].

At higher concentrations (25 µmol/L), perhexiline showed a substantial decrease in ATP

production, β-oxidation and triglyceride formation after a 24-hour incubation period. This was

accompanied by a diminished cell count, which suggests that at these concentrations,

perhexiline exhibits toxic effects [75].

Page 51: The stereoselective pharmacodynamics of the enantiomers of

51

Concentrations of perhexiline in the mitochondria were found to be 20-fold higher than those

of the initial incubate concentration [75] and may provide some further explanation to its

toxic effects. The increase in mitochondrial perhexiline concentration is reliant on the

mitochondrial membrane potential. As it enters the intramembranous space of the

mitochondria, the uncharged perhexiline molecule becomes partially protonated due to the

acidic environment, allowing it to cross the mitochondrial membrane down the

electrochemical potential. Once inside the mitochondria, the alkaline conditions disassociate

the proton from perhexiline and thus trapping the perhexiline molecule inside. [75].

At high concentrations (200 µmol/L) perhexiline seems to decrease mitochondrial respiration

by completely inhibiting the respiratory chain. This eventually results in cell death mainly via

oxidative dephosphorylation, which is a result of proton re-entry into the cell without

producing any ATP via ATP-synthase and uncoupling of the respiratory chain, which in turn

prevents the cell pumping protons back into the intramembranous space [75].

1.5.3 In Vivo Perhexiline Toxicity Studies

A study involving two different rat strains, the Dark Agouti (DA) and the Sprague Dawley

(SD), demonstrated significant neuronal toxicity especially in the DA strain. In this study, the

DA strain was proposed to represent of a model of human CYP2D6 poor metabolism,

whereas the SD was a model of extensive metabolism (see section 1.5.2 for more details).

Perhexiline induced neuronal lipidosis in histological sections in the DA rat after 4 weeks

treatment with 80 mg/kg/day given orally. At this dosage, the dorsal root ganglion taken from

these rats revealed a considerable number of inclusion bodies, which were lamellar in

appearance. A reduction in neuronal motor latency was also noted in perhexiline-treated DA

rats when compared to control (P<0.05) or SD (P<0.025) rats. Hepatic biochemical analyses

Page 52: The stereoselective pharmacodynamics of the enantiomers of

52

revealed no significant differences when compared to controls. Furthermore, this lipidosis

became more severe with prolonged exposure to perhexiline, whereas the SD rats showed no

such inclusion bodies [76].

1.6 Pharmacokinetics of Perhexiline

1.6.1 Studies with Racemic Perhexiline

Studies in the 1970s with 14C - perhexiline revealed that 6-12 hr. after oral administration in

humans absorption was greater than 88% [77], although absolute bioavailability is unknown

as perhexiline is only available as an oral formulation. Perhexiline has a volume of

distribution of approximately 1470 L [78] and is highly tissue (mitochondrial) [79] and

plasma protein bound (>90%) [80].

Perhexiline is almost exclusively hepatically cleared by metabolism via CYP2D6 (with minor

contributions of CYP3A4 and CYP2B6) to its major metabolites cis-OH-(±)-perhexiline,

trans1-OH-(±)-perhexiline, trans2-OH-(±)-perhexiline [81]. The conversion of perhexiline to

its cis hydroxyl metabolite is the major pathway in its metabolism (Figure 7) [81]. PMs (see

below) are not able to produce the cis metabolite and as a result have substantially increased

plasma perhexiline concentrations. [79, 82].

Previous studies have reported that patients who developed peripheral neuropathy had a

(mean ± s.d) metabolic ratio (the ratio of unchanged parent drug to metabolite produced) of

3.78 ± 0.43, significantly higher (p<0.01) than patients that hadn’t developed the neuropathy

with a metabolic ratio of 1.07 ± 0.18. The pharmacokinetic profile showed a higher

Page 53: The stereoselective pharmacodynamics of the enantiomers of

53

concentration of unchanged parent drug compared to metabolite in those patients suffering

from peripheral neuropathy when compared to those that didn’t [74, 83].

Figure 7. The major metabolic pathway for perhexiline and formation of OH- perhexiline

metabolites.

Page 54: The stereoselective pharmacodynamics of the enantiomers of

54

1.6.2 Cytochrome P450s

Cytochrome P450s (CYP’s) are a group of enzymes, which are involved in the degradation,

and elimination of exogenous compounds from the body. They play a key role in the

metabolism of substances or chemicals derived from either the environment or dietary intake

(including pharmaceuticals/xenobiotics), converting these to more excretable compounds as a

body defence mechanism [84, 85].

CYP enzymes are classified by a number (indicating the family they belong to), a subfamily

letter and another number to identify the individual enzyme within the subfamily, and in some

cases an asterix followed by a number at the end, to identify any allelic variants that may be

associated with that gene, e.g. CYP2D6*4 [85].

Humans have 57 cytochrome P450 genes but only the encoded proteins from the CYP1,

CYP2 and CYP3 families seem to be of great importance in terms of drug metabolism. Each

CYP enzyme displays a unique characteristic, whether it is by demonstrating a higher affinity

of a drug to a specific protein region or greater selectivity towards one particular enantiomer

when drugs are administered as a racemic mixture [85].

Cytochrome P450 metabolism mainly occurs in the liver, however the epithelium in the small

intestine also contains cytochrome P450 enzymes and may play a role in the bioavailability of

a drug whose primary mechanism of elimination is via cytochrome P450 metabolism. In

particular, CYP3A enzymes are concentrated in the tip of villi in the upper small intestine (as

well as the liver) and can metabolise a wide array of compounds. As perhexiline is primarily

metabolised by CYP2D6, this CYP3A family has only a minor effect on its clearance.

Page 55: The stereoselective pharmacodynamics of the enantiomers of

55

1.6.2.1 CYP2D6

CYP2D6 has been one of the most widely studied cytochrome P450 enzymes with respect to

polymorphisms. Although its expression in the liver is relatively low (2-4%), it is responsible

for the metabolism of about 25% of currently available drugs [86], which include:

antidepressants, antipsychotics, antiarrhythmics, antiemetics, β-blockers and opioids [85, 86].

The CYP2D6 enzyme is a result of the expression of the CYP2D6 gene located on

chromosome 22q13.1 [87]. The gene is made up of 9 exons with an open reading frame of

1491 base pairs, which code for 497 amino acids. Due to its highly polymorphic nature, there

is a large inter-individual variability in the activity of CYP2D6 within a population [88].

There are over 70 reported CYP2D6 allele variants [86] which result in altered enzymatic

function by having no enzymatic activity, reduced enzymatic activity or by having increased

enzymatic activity. This has resulted in the classification of subpopulations into poor

metabolisers (PM), intermediate metabolisers (IM), extensive metabolisers (EM) and ultra-

rapid metabolisers (UM) respectively (Figure 8) [86, 89]. In a Caucasian population UMs,

EMs, IMs, and PMs account for approximately 3-5%, 70-80%, 10-17% and 5-10%,

respectively [86].

1.6.2.2 Alleles Producing No Functional CYP2D6 Protein

Null alleles result in a non-functional protein, which in turn leads to no detectable CYP2D6

enzymatic activity. The *3, *4, *5, *6, *7, *8, *11, *12, *13, *14, *15, *16, *18, *19, *20,

*21, *38, *40, *42, *44, *56 and *62 are all variant alleles which encode non functional

proteins as a result of either a point mutation, deletion or addition of a single nucleotide or the

deletion/rearrangement of genes [86, 90]. CYP2D6s highly polymorphic nature leads to a

Page 56: The stereoselective pharmacodynamics of the enantiomers of

56

non-functional protein due to the presence of these allelic variants, giving rise to the PM

phenotype. This has a major consequence in clinical settings especially when administered a

pharmacological agent that is highly reliant on CYP2D6 for its metabolism. Furthermore, this

impaired ability to metabolise certain drugs will inevitably lead to drug-induced toxicity as a

result of its accumulation, especially for those agents which have a narrow therapeutic index

such as the antiarrhythmic drugs flecainide (therapeutic range 200-1000 ng/mL) [91] and

mexiletine (therapeutic range of 0.5-2.0 µg/mL) [92].

1.6.2.3 Alleles Producing Functioning Protein with Decreased Activity

In some cases there are allelic variants, which produce a functional protein; however such

proteins have significantly impaired catalytic activity. Alleles *10, *14, *17, *18, *36, *41,

*47, *49, *50, *51, *54, *55 and *57 are responsible for the residual function of the resulting

protein [86, 93]. A decrease in function is a result of decreased substrate affinity to the

enzyme as well as the production of an unstable protein.

The presence of these allelic variants give rise to IMs, however, the highly polymorphic

nature of CYP2D6 results in an overlap in the population distribution with EMs [93].

Page 57: The stereoselective pharmacodynamics of the enantiomers of

57

Figure 8. CYP2D6 phenotype definition and distribution in a random European population

(N=316). The numbered arrows indicate the range of the sparteine metabolic ratio associated

with number of functional genes. Taken from Zanger et al [89].

Page 58: The stereoselective pharmacodynamics of the enantiomers of

58

1.6.2.4 Multiple CYP2D6 Copies

Ultra rapid metabolisers (UM) are those that are able to metabolise probe substrates for

CYP2D6 at a rate which surpasses that of the wild type CYP2D6*1 allele. The ability of this

particular sub population to metabolise CYP2D6 substrates at an elevated rate is mainly due

to the CYP2D6 gene being subject to copy number variations. As an extreme example, it has

been reported that in a Swiss family, the father and two of his children had up to 13 copies of

a functional CYP2D6 allele [94].

1.6.2.5 CYP2D6 and Perhexiline Metabolism

Perhexiline is a substrate for CYP2D6 and as such is subject to polymorphic metabolism.

PM’s are at risk of toxicity due to excess plasma and tissue perhexiline concentrations

following doses associated with efficacy in EMs. In contrast, UM’s may be too efficient at

eliminating the drug and therefore struggle to attain target concentrations in plasma and risk

having reduced or no clinical benefit; although this sub-population has been little studied.

Horowitz et al., demonstrated almost no side effects with optimum efficacy in patients where

plasma perhexiline concentrations were kept between 0.15 - 0.60 mg/L [7]. In clinical practice

in this laboratory, the individualised dosage schedule to achieve this target concentration can

range from 50 mg/week in PMs up to 1,000 mg/day in UM’s (i.e., a 140-fold range).

In a pharmacokinetic study by Horowitz et al. [95], five patients were administered a single

oral dose of either 150 or 300 mg of perhexiline. The results of the study revealed that after

the dose was doubled, the mean peak plasma perhexiline concentration, as well as area under

the concentration versus time curve (AUC), was 4.3 and 5.3 times greater, respectively. They

also showed that the mean elimination half-life was concentration-dependent with a half-life

Page 59: The stereoselective pharmacodynamics of the enantiomers of

59

of 11.2 ± 2.1 hours after the 150-mg dose and 19.1 ± 2.8 hours after the 300-mg dose

respectively. As perhexiline has a high hepatic clearance almost exclusively via CYP2D6,

these results suggest that perhexiline metabolism was saturable and its pharmacokinetics non-

linear in nature. The narrow therapeutic range and such widely varying dosage requirements,

calls for strict therapeutic drug monitoring (TDM) upon clinical administration of perhexiline

maleate to individualise dosages in order to optimise efficacy without toxicity.

1.6.3 Animal Model CYP2D6 Poor Metabolism

The rat is commonly used as an animal model for the identification of toxicity and drug

disposition in humans. They can be genetically modified or specifically bred between species

to develop a unique characteristic that mimics a human condition or disease state. One such

breed is the Dark Agouti rat, which is an established model of ‘poor’ metabolism involving

the CYP2D enzyme family [96]. Debrisoquine is a commonly used substrate for the human

CYP2D6 enzyme and a deficiency in the metabolism of this drug may be due to a defective

enzyme as a result of mutations in its synthesis or a decrease in its expression, leading to

lower enzyme activity and therefore a phenotypic poor metaboliser. Al-Dabbagh et al [96]

used inbred rat strains in order to determine an appropriate model of human polymorphic drug

metabolism, using the metabolic ratio; that is the ratio of parent drug to metabolite (4-hydroxy

debrisoquine) recovered in urine over a set period of time. In this study they observed the

metabolism of 5 mg/kg of debrisoquine over 24 hr. in Wistar, Lewis, Fischer A/GUS, PVG,

BN and Dark Agouti rats with three rats of each strain. Of all the strains, the Dark Agouti and

the Lewis showed drug and metabolite recoveries which suggested that the metabolic ratio of

the Dark Agouti rat was consistent with that of human poor CYP2D metaboliser (PM),

whereas the Lewis strain showed a metabolic ratio which was very similar to a human

extensive metaboliser [96].

Page 60: The stereoselective pharmacodynamics of the enantiomers of

60

Not only does the Dark Agouti strain seem to be more deficient in the metabolism of

debrisoquine than any other, but the female Dark Agouti shows greater impairment than its

male counterpart suggesting a sex-dependent difference [97]. When debrisoquine 4-

hydroxylase activity of the Dark Agouti rat was compared with that of the Fischer and Lewis

strains (models of extensive metabolisers), it was significantly reduced by 3.5- to 5-fold,

which was consistent with previous studies [97]. Interestingly Kahn et al. also showed that all

female rats from each strain had significantly impaired 4-hydroxylase activity when compared

to the male in the same strain, with the Dark Agouti female exhibiting the lowest activity of

all [97].

In a later study by Gonzalez et al. it was discovered that the Dark Agouti rat possessed two

CYP2D proteins, which were termed 2db1 and 2db2 and are now known as CYP2D1 and

CYP2D2. In that study they put forward the notion that it was only the CYP2D1 enzyme that

possesses the catalytic activity to metabolise debrisoquine and that CYP2D2 had no activity

towards debrisoquine [98]. Furthermore CYP2D2 concentrations were 30 times less abundant

in male Dark Agouti rats when compared to Sprague Dawley rats and almost absent in female

Dark Agouti rats.

In 1989 Matsunaga et al., discovered three more CYP2D proteins termed CYP2D3, 4 and 5.

The study also suggested that the inability of the Dark Agouti rat to metabolise debrisoquine

to its metabolites was a result of dramatically reduced CYP2D1 concentrations and that

CYP2D2, CYP2D3, CYP2D4 and CYP2D5 did not exhibit any metabolic activity towards the

drug [99]. The theory that CYP2D1 was the only CYP2D enzyme with the capacity to

metabolise debrisoquine in the rat was resolved by Yamamoto et al (1998) and Schulz et al

(1999). Using immunochemical measurements with anti-CYP2D peptides it was revealed that

there was no significant difference in CYP2D1 protein expression between the proposed poor

Page 61: The stereoselective pharmacodynamics of the enantiomers of

61

metaboliser model, the Dark Agouti and the proposed extensive metaboliser, the Sprague

Dawley rat. However the expression of CYP2D2 was decreased between the two species with

the Dark Agouti strain showing significantly lower protein content. In order to eliminate the

possibility that the Dark Agouti rats’ inability to metabolise debrisoquine was the result of a

deficient CYP2D1 protein, recombinant CYP2D1 and CYP2D2 was expressed in yeast. The

result clearly showed that CYP2D2 was 20-fold more effective at metabolising debrisoquine

than CYP2D1. The introduction of quinine (inhibitor of the CYP2D mediated oxidation of

debrisoquine) reduced CYP2D2 mediated oxidation of debrisoquine by 53-67%, whereas

CYP2D1 activity was inhibited by 0-6%. They concluded that the metabolism of debrisoquine

in the Dark Agouti female rat was primarily mediated by the CYP2D2 enzyme [100-102].

However, this DA/SD rat model does not hold up for all human CYP2D6 metabolised drugs.

In humans, propranolol is metabolised by CYPs 2D6, 1A2, 2C19 [102] and UDP-

glucuronyltransferase [103], whereas in rats the main metabolic pathway is via the CYP2D

isoform. In a report by Komura et al. they studied the effects of metabolism on propranolol

and metoprolol in DA and Wistar rats. Administration of metoprolol to DA and Wistar rats

coincided with the pharmacokinetic profiles of human PM and EM respectively, whereas the

Km of propranolol for its high affinity enzyme in human liver microsomes was 3-4 µM, 20-50

fold higher than in the corresponding values in rats [104]. The low Km value together with the

involvement of only the CYP2D isoform, may explain why the DA rat is not always an

appropriate model of CYP2D6 human metabolism [104, 105].

Page 62: The stereoselective pharmacodynamics of the enantiomers of

62

1.7 Chirality

In 1811 Aarago [106] and in 1812 Biot [106], showed the effects of crystals on the plane of

polarized light. In 1848 Louis Pasteur was the first to recrystallise racemic sodium ammonium

salt of tartaric acid and discovered that 2 different crystalline structures were produced [107].

Pasteur went on to demonstrate that of these 2 different crystalline structures, only one was

fermented by bacteria. It was not until 1847, when van’t Hoff showed that there was a

relationship between three-dimensional structures, optical activity and an asymmetrical

carbon and thus the concept of chirality began.

Chiral compounds have an asymmetric atom and thus form isomers, which are compounds

that share the same molecular formula but differ in their spatial and structural arrangements.

Enantiomers are stereoisomers that have identical chemical features, however they differ in

their spatial arrangement and thus have a handedness property; that is they may exist as a

right or left form and are not superimposable [108]. Enantiomer is the nomenclature given to

each chiral compound form and these enantiomers may exist as dextro (+) and or levo (-),

which can have different properties with respect to protein, enzyme or transporter

interactions. R (Rectus) and S (Sinister) nomenclature often seen associated with enantiomers,

refers to the configurations of the atoms attached to the chiral centre. Based on the sequence

rule systems devised by Cahn, Ingold and Prelog in 1956, the atoms attached to the chiral

centre are ranked based on their atomic number. Highest priority is given to the atom with the

highest atomic number. If the priority sequence is to the right the nomenclature given is R and

if the priority sequence is to the left then it is designated as S. The terms (+) and (-) or d and l

can also be used, however these do not relate to structure but rather refer to the effects on

polarised light.

Page 63: The stereoselective pharmacodynamics of the enantiomers of

63

Many commonly prescribed medications are currently in a racemic formulation and one could

argue that these compounds contain a 50% impurity as one enantiomer may be responsible for

all the therapeutic effects and the other may attribute to the toxic effects. An example is

ketamine, S-ketamine is an anaesthetic agent, while its enantiomeric counterpart (R-ketamine)

causes hallucinations [109]. It is clear that enantiomers have variable effects in different

biological environments whether it may be due to an increase in affinity for a certain receptor

or a different pharmacokinetic profile. As a result of this variability in activity it may actually

be safer or more efficacious to market single enantiomers as the therapeutic compounds rather

than the racemate mixture [110].

1.7.1 Pharmacokinetics of Perhexiline Enantiomers

Perhexiline is a chiral compound, which has a chiral carbon forming four different bonds and

it exists as (+)- and (-)- enantiomers (Figure 1). The enantiomers are metabolised to cis-OH-

(+)-perhexiline, cis-OH-(-)-perhexiline, trans1-OH-(+)-perhexiline, trans1-OH-(-)-

perhexiline, trans2-OH-(+)-perhexiline and trans2-OH-(-)-perhexiline (Figure 9) [79, 82].

Although several studies have demonstrated enantioselectivity in the pharmacokinetics of

perhexiline, to date there are no published papers which have investigated the

pharmacodynamic effects of either the (+)- or (-)- enantiomers of perhexiline or consider that

enantiomers may differ in their pharmacodynamic and toxicological properties.

The first report on the pharmacokinetics of perhexiline enantiomers was in 1986 by Gould et

al [111] who conducted a study investigating the pharmacokinetic properties of the racemic

and enantiomeric formulations of perhexiline. Eight healthy volunteers were given an oral

dose of 300mg of racemic, (+)- or (-)- perhexiline on separate occasions. Blood samples were

taken at an interval of 2 hr. for the next 24 hr. and urine was collected daily. Their results

Page 64: The stereoselective pharmacodynamics of the enantiomers of

64

showed that the peak plasma perhexiline concentration for the (+)- enantiomer was 2.5-fold

higher than that of the (-)- enantiomer, which showed no significant difference to 300 mg of

racemic formulation. The (-)- enantiomer seemed to be cleared much more rapidly than the

(+)- enantiomer by not only showing a lower plasma perhexiline concentration, but also a 28-

fold increase in metabolite production when compared to the racemic mix, which was also

significantly greater than the (+)- enantiomer. These observations were confirmed by Davies

et al [79] and Inglis et al [80], who also showed that both enantiomers displayed non-linear

kinetics.

Studies with human liver microsomes suggested that CYP2D6 is the main enzyme involved in

the metabolism of both the (+)- or (-)- enantiomers to their respective hydroxyl metabolites in

EMs [79]; although elimination seems to be enantioselective for the (-)- perhexiline rather

than (+)- perhexiline [79]. However CYP3A4 and CYP2B6 appear to be the main enzymes

involved in metabolising the enantiomers to their metabolites in PMs [79].

Page 65: The stereoselective pharmacodynamics of the enantiomers of

65

Figure 9. The chiral structures of the mono- and di- hydroxyl metabolites resulting from

perhexiline metabolism, taken from Davies et al [82].

PERHEXILINE 79

FIG. 2. The six different perhexiline metabolites. Perhexiline is metabolized to one of its six monohydroxymetabolites or its dihydroxymetabolites. These include a cis, trans-1 and trans-2 monohydroxy variant for eachenantiomer (from Davies et al. Drug Metab Dispos 2006).

These investigations revealed that perhexiline was both a coronary and a systemic va-sodilator. This effect was independent of the adrenergic system. By systemic administrationperhexiline, 0.0055 mg/kg i.v, increased coronary and femoral blood flow despite a fall insystemic blood pressure (–13% to –49% with 0.3 to 3 mg/kg i.v.). This finding contrastedwith nitrates, which, when given systemically, reduced blood pressure at the expense offemoral blood flow (Hudak et al. 1970). These coronary vasodilator effects may reflect theweak L-type calcium channel-blocking effect of perhexiline; however, as the experiments

TABLE 1. The different biochemical properties of perhexiline and their putative clinical effects.

Biochemical effect Clinical relevance

Inhibition of transmitochondrial carnitine“shuttle” (CPT1/CPT2 inhibition)

Critical to metabolic antiischemic andincreased efficiency effects

L-Type Ca2+ channel antagonism Potential negative inotropic effects (probablyonly pertinent in overdose)

K+-channel inhibition (HERG and Kv1.5channel)

Occasional Q-T prolongation (perhaps inpatients with underlying mutations in K+

channels, e.g., KCNQ1 or on other Q-Tprolonging agents)

Potentiation of insulin release and reduction ofblood sugar levels

May be of benefit to diabetic andinsulin-resistant individuals but requiredalertness to hypoglycemia and potential dosereduction in insulin or insulinotropic agents

Potentiation of platelet responsiveness to nitricoxide, increased cGMP responsiveness andinhibition of NADPH oxidase

Pertinent to antiischemic effects, especially inacute coronary syndromes and aorticstenosis

Cardiovascular Drug Reviews, Vol. 25, No. 1, 2007

Page 66: The stereoselective pharmacodynamics of the enantiomers of

66

The evidence suggests that the enantiomers of perhexiline are metabolised at different rates

by different enzymes. This reveals certain gaps in our knowledge about the pharmacodynamic

properties of perhexiline in the human body, as the correlation between the different

pharmacokinetic and pharmacodynamic effects of the individual perhexiline enantiomers is

yet to be described. For example, it is not known whether it is the (+)- or (-)- enantiomer that

may be responsible for the peripheral neuropathy and/or hepatic steatosis seen in clinical

toxicity of perhexiline, or whether just one of the enantiomers is responsible for the inhibition

of CPT-1, or potentially whether the therapeutic effects of one enantiomer may be masked by

the adverse effects of the other?

Hence, one of the aims of this study was to investigate the pharmacodynamic effects of the

(+)- and (-)- perhexiline enantiomers.

1.8 Aims and Hypotheses

Perhexiline’s clinical use is complicated by its complex pharmacokinetics, which without

therapeutic drug monitoring to individualise therapy, has the potential to cause severe hepatic

and/or neurotoxicity in some patients. This thesis describes the investigation of the

pharmacodynamic properties of (+)- and (-)- perhexiline in vitro and in vivo using a rat model

with the intent of determining if (+)- perhexiline or its optical antipode is responsible for the

steatosis and peripheral neuropathy seen in clinical practice. If either enantiomer demonstrates

a superior pharmacodynamic profile, whether it is improved toxicity or efficacy, then it may

be possible to develop an enantiomerically based formulation that is less toxic and/or more

efficacious than the racemic compound currently prescribed.

Page 67: The stereoselective pharmacodynamics of the enantiomers of

67

The overall aim of this thesis was addressed by developing a rat model that would mimic the

pharmacokinetics and metabolism of perhexiline in humans. By administering clinically

relevant doses of either (+)- or (-)- perhexiline, it should be possible to determine whether

either one, or both are responsible for toxicity. In addition, enantioselectivity with respect to

one of perhexiline’s potential beneficial effects, inhibition of NADPH oxidase, was also

tested in vitro.

The following hypotheses were tested:

1. That the enantiomers of perhexiline demonstrate different potencies for the inhibition

of neutrophil NADPH oxidase induced superoxide formation.

2. That the enantiomers of perhexiline demonstrate enantioselectivity in hepato- and

neuronal toxicity in the DA agouti animal model.

Page 68: The stereoselective pharmacodynamics of the enantiomers of

68

Chapter  Two:  Methods  

Page 69: The stereoselective pharmacodynamics of the enantiomers of

69

2.1 (+)- and (-)-Perhexiline Preparation

The Chemistry Department at The University of Adelaide prepared the perhexiline

enantiomers for this study. The (+)- and (-)- perhexiline were prepared by using a previously

reported method from our laboratory [112]. Racemic perhexiline base was obtained from

racemic perhexiline maleate source. Briefly, the maleate salt was removed by dissolving it in

a sodium hydroxide solution, which was then extracted with ether. These extracts were then

washed with water and dried with magnesium sulphate. The solvent was discarded to obtain

pure racemic perhexiline.

Individual perhexiline enantiomers were obtained by adding, (S)-1,1′-Binaphthalene-2,2′-diyl

hydrogen phosphate (S-BNPA) in acetone which resulted in the precipitation of a

diastereomeric salt of (-)-perhexiline + (S)-BNPA. The isolation of the (-)-perhexiline

enantiomer from the BNPA salt was achieved by the addition of a chloroform and ammonia

solution. Using (+)- enriched perhexiline solution, the procedure was repeated with (R)-

BNPA (Figure 10).

Page 70: The stereoselective pharmacodynamics of the enantiomers of

70

Figure 10. Preparation and resolution of the (+) and (-) enantiomers of perhexiline from

racemic perhexiline maleate [112]

Page 71: The stereoselective pharmacodynamics of the enantiomers of

71

2.2 Ethics

The liver toxicity study was approved by the Animal Ethics Committee of The University of

Adelaide (approval number M79/07) and the SA Pathology/CHN Animal Ethics Committee

(approval number 120/07). The neutrophil studies were approved by The Queen Elizabeth

Hospital Human Research Ethics Committee (approval number 2007056).

2.3 Animals

Female Sprague Dawley and DA rats (weight 180-200 g) were purchased from The

University of Adelaide animal house (Adelaide, South Australia, Australia). The rats were

individually housed at The Queen Elizabeth Hospital animal house. For the first week, the rats

were allowed to acclimatise to their environment and were fed standard rat chow. During this

period, behaviour and feeding patterns were observed and noted. The food pellets were

weighed every evening and morning in order to determine the food consumption of these rats

(being nocturnal feeders). The larger Sprague Dawley rats consumed around 21 g of food

(approx. 7 large pellets), whereas the smaller DA strain consumed less food by comparison (4

pellets, approximately 13 g of food). All rats had ad libitum access to food and water at all

times. The animals were kept in controlled conditions, in facilities which provided a 12 hr.

day/night cycle at a temperature of approximately 22 °C.

2.4 Perhexiline Dosage Determination and Administration

The dosage regime for the toxicity study was modified from a previous report by Meier et al

[76]. Meier et al orally dosed female DA animals for a period of 8 weeks at 80 mg/kg of

perhexiline maleate and observed a mean plasma perhexiline concentration of 0.6 mg/L, the

Page 72: The stereoselective pharmacodynamics of the enantiomers of

72

upper limit of the perhexiline therapeutic range in humans. At this dosage, the dorsal root

ganglion taken from these rats revealed a considerable number of inclusion bodies, which

were lamellar in appearance.

My dosing regimen was based on the toxicity observed from the study of Meier et al. At 80

mg/kg, Mallory bodies accumulated in dorsal root ganglia giving rise to peripheral

neuropathy associated with clinical toxicity following chronic perhexiline dosing. However

the lack of biochemical changes in the liver, as well as plasma perhexiline concentrations that

were near or within the human therapeutic range, indicated that an increase in dosage could

achieve hepatic toxicity in the present study. The dose selected in the pilot study was 200

mg/kg, hence 2.5-fold greater than Meier et al [75].

Administration of the drug proved to be challenging. Initial attempts were made to

incorporate the drug directly into the food pellets in order to mask its taste. Perhexiline

maleate was dissolved in ethanol and the solution was injected into food pellets that were then

left to allow the ethanol to evaporate overnight, leaving just the perhexiline maleate.

Administration of these ‘perhexiline pellets’ was ineffective, as the rats appeared to only eat

parts of the pellet, probably avoiding those areas that contained the drug. After seeking

veterinarian advice, a mixture of peanut paste and perhexiline maleate was spread on the food

pellet. This proved to be an effective way to administer perhexiline to these rats. During

active treatment, each animal was housed individually and the required daily dose of

perhexiline maleate was coated onto 7 rat chow pellets for SD rats, or 4 pellets for DA rats,

placed in the cages at 4-5 pm each day, and left until the next dose. Food and perhexiline

consumption was monitored daily and extra drug-free pellets provided if necessary.

Page 73: The stereoselective pharmacodynamics of the enantiomers of

73

2.5 Tissue Preparation for HPLC

Liver and heart tissue was dissected from the sacrificed rats, snap frozen in liquid nitrogen

and stored at -80°C. The tissue was partially thawed in order to remove a slice, which was

then completely thawed in saline solution. The smaller piece was dried and then weighed. A

volume of cold phosphate buffer (0.15 M, pH 6) twice that of the tissue weight was added and

the piece of tissue was further diced into smaller sections. A tissue homogenizer was used to

reduce the solid tissue pieces resulting in a homogenates of heart and liver. The aliquots were

taken and stored at -80°C.

2.6 Measurement of (+)- and (-)-Perhexiline in Plasma and Tissue

Perhexiline was extracted and quantified using the previously published method of Davies et

al. [112], which had been validated in human plasma and liver microsomes. Briefly, 50 µl of a

10 mg/L solution of prenylamine (internal standard) as well as 50 µl of 2 M NaOH and 4 ml

of a 10% ethyl acetate in n-hexane were added to 500 µl of liver or heart homogenate. This

homogenate preparation was shaken for 15 min at 100 oscillations per min and then

centrifuged at 3000 rpm for 10 min at 10°C. It was then placed in a dry ice/ethanol bath

whereby the aqueous layer was snap frozen and subsequently discarded and the organic phase

collected. The organic layer was transferred to 5 ml glass tubes and placed in a vacuum

centrifuge to dry at room temperature. Once dried, 200 µL of the derivatising agent, naphthyl

ethyl isocyanate (NEIC) in acetone (0.05%), was added, vortex mixed and incubated at room

temperature for 5 min. Following this incubation, the addition of 200 µL of a 0.5M NaOH

solution was followed by that of 3 mL of 10% ethyl acetate in n-hexane. The 5 mL tube was

caped and vortex mixed for 5 min, then centrifuged at 2500 rpm for a further 3 min at 10°C.

The aqueous phase of the mixture was snap frozen in a dry ice/ethanol bath and the organic

Page 74: The stereoselective pharmacodynamics of the enantiomers of

74

layer transferred to another 5 mL tube and the aqueous layer discarded. The tube was placed

in a vacuum centrifuge at room temperature until it was completely dry. The resulting residue

was reconstituted with 150 µL of the 80% methanol and 20% water mobile phase.

The HPLC conditions were as described in Davies et al. [112]. Briefly, analyses were

conducted on an Agilent 1100 series HPLC apparatus (Agilent Technologies, Forest Hill,

Victoria, Australia) operated by Chemstation for LC 3D software. The hardware consisted of

a model G1322A degasser, a model G1311A pump operating at 1 mL/min, a model G1313A

autosampler and a model G1321A fluorescence detector with excitation and emission

wavelengths of 218 and 334 nm, respectively. Resolution of the diastereomers was achieved

using a Merck Purospher RP-18E column (5 µm, 125 mm × 4 mm) at ambient temperature.

The mobile phase was composed of 80% methanol and 20% water for the first 12 min, at

which time the methanol was increased to 86% in a linear gradient over 1 min and maintained

at that concentration for the remaining 16 min of each sample run time. An example of the

chromatogram obtained under these conditions is shown in Figure 11.

The same method and conditions were used to determine concentrations of the perhexiline

enantiomers in plasma.

Page 75: The stereoselective pharmacodynamics of the enantiomers of

75

2.6.1 Validation of HPLC Perhexiline Assay in Liver Homogenate and Plasma

Calibration and quality control (QC) samples were prepared using control liver and heart

homogenates spiked with (±)-perhexiline maleate dissolved in 10% methanol with 0.1M HCL

As the method was a minor modification of a previously published assay established in our

laboratory [112], an abbreviated revalidation was carried out by determining intra- and inter-

assay accuracy and precision which were required to be within 15% except at the lower limit

of quantitation (LLOQ) where 20% was acceptable. Calibration curves were only accepted if

the r2 was greater than 0.98.

The intra assay validation in liver homogenate for (+)-perhexiline demonstrated a coefficient

of variation (CV) and a bias of 5 and 7% for the highest calibrator (2 mg/L, n=6) and 17 and -

7% for the lowest calibrator (0.05 mg/L, n=6), respectively. (-)- Perhexiline had a CV and

bias of 6% at the highest calibrator (2 mg/L, n=6), whereas the lower calibrator (0.05 mg/L,

n=6) had a CV and bias of 18 and 6%, respectively. The inter assay validation for (+)-

perhexiline had a CV of 10 and 8% and a bias of -5 and 15% for the highest and lowest

calibrators respectively, whereas (-)-perhexiline had a CV of 13 and 16% and a bias of -7.0

and <0.1% for the highest and lowest calibrators respectively.

Two QC samples were run with each assay, comprising liver homogenate spiked to a

concentration of 0.075 and 0.75 mg/L of (±)-perhexiline in QC1 and QC2, respectively. The

inter assay CV and bias for QC1 was 14 and 13% for (+)-perhexiline, and 9 and 13% for (-)-

perhexiline. For QC2, the CV and bias was 8 and 14% for (+)-perhexiline, 8 and 13% for (-)-

perhexiline respectively.

Page 76: The stereoselective pharmacodynamics of the enantiomers of

76

As there was not enough heart homogenate to perform a validation assay in this tissue, we

used calibration curves and QC’s that were made up in liver homogenate.

The plasma concentrations of perhexiline and its enantiomers were measured by a published

method that was established and validated in our laboratory [112]. As the method was a minor

modification of a previously published assay established in our laboratory, an abbreviated

revalidation was carried out by determining intra- and inter-assay accuracy and precision. The

intra assay validation in plasma for (+)-perhexiline demonstrated a coefficient of variation

(CV) and a bias of 3 and 5% for the highest calibrator (2 mg/L, n=6) and 10 and 10% for the

lowest calibrator (0.05 mg/L, n=6), respectively. (-)- Perhexiline had a CV and bias of 3 and

5% at the highest calibrator (2 mg/L, n=6), whereas the lower calibrator (0.05 mg/L, n=6) had

a CV and bias of 10 and 10%, respectively. The inter assay validation for (+)-perhexiline

(n=3) had a CV of 1 and 9% and a bias -1 and -10% of for the highest and lowest calibrators

respectively, whereas (-)-perhexiline (n=3) had a CV of 1.0 and 13% and a bias of -1 and -

13% for the highest and lowest calibrators respectively.

Page 77: The stereoselective pharmacodynamics of the enantiomers of

77

Figure 11. A liver homogenate chromatogram from the method described. A) Represents a

chromatogram from a blank sample without internal standard. B) Represents a chromatogram

from a rat liver dosed with 200 mg/kg of racemic perhexiline. The chromatography conditions

allows for separation of (+)- and (-)- perhexiline and the prenylamine internal standard, the

retention time for the internal standard was 10.7 min, the (+) perhexiline enantiomer was 25.1

min, and the (-) perhexiline enantiomer was 27.1 min.

Page 78: The stereoselective pharmacodynamics of the enantiomers of

78

2.7 Measurement of OH-perhexiline in Plasma and Liver

Measurement of cis-, trans1- and trans2-OH-perhexiline was also based on a previously

published method by Davies et al [82], which had been validated for plasma and liver

microsomes. The assay was therefore revalidated to measure OH-perhexiline in heart, liver

and plasma homogenate. Briefly, 100 µL of 10 mg/L of the internal standard (fendiline) was

added to the liver homogenate. This was followed by the addition of 50 µL of 2M NaOH and

4 mL of a 30% dichloromethane in n-hexane. This homogenate preparation was shaken for 15

min at 100 oscillations per minute and then centrifuged at 3000 rpm for 10 min at 10°C. It

was then snap frozen in a dry-ice/ethanol bath whereby the aqueous layer was discarded and

the organic phase was decanted into 5 ml glass tube. It was then placed in a vacuum

centrifuge to dry at room temperature. Once dried, 200 µL of the derivatising agent, dansyl

chloride in acetone (0.015M), was added, vortex mixed and incubated at 40°C for 1 hr.

Following this incubation, 200 µL of a 0.3M NaHCO3 solution was added, followed by 3 mL

of n-hexane. The 5 mL tube was caped and vortex mixed for 5 min, then centrifuged at 2500

rpm for a further 3 min at 10°C resulting in an organic and aqueous layer. The mixture was

snap frozen in a dry-ice/ethanol bath and the organic layer was decanted into a 5 mL tube and

the aqueous layer discarded. The tube was placed in a vacuum centrifuge at room temperature

until it was completely dry. The resulting residue was reconstituted with 150 µL of the 80%

methanol and 20% water mobile phase.

The HPLC instrument and analytical column were the same as for the enantiomer assay

mentioned previously in section 2.6 with minor modifications. This assay was performed at a

temperature of 30 oC and the mobile phase was 80% methanol and 20% distilled water for the

first 14 min, at which time the methanol was increased to 100% for the remaining 8 min of

each sample run time [82]. An example of the chromatogram obtained under these conditions

Page 79: The stereoselective pharmacodynamics of the enantiomers of

79

is shown in Figure 12.

A)

B)

Page 80: The stereoselective pharmacodynamics of the enantiomers of

80

Figure 12. Chromatogram of liver homogenate for the method described. A) Represents a

blank sample without internal standard. B) Represents a chromatogram from a rat liver

following administration of 200 mg/kg of (+)-, (-)- or racemic perhexiline. The retention

times for the OH-perhexiline (Px) metabolites were: cis- 10.8 min, trans-1- 12.4 and trans-2-

13.6 min, with the internal standard (fendiline) at 18.8 min, respectively.

Page 81: The stereoselective pharmacodynamics of the enantiomers of

81

2.7.1 Validation of HPLC OH-Perhexiline Assay in Liver Homogenate

Pure cis- trans1- and trans2-OH-perhexiline were obtained from Sigma Pharmaceuticals

(South Croydon, Victoria, Australia). Calibration and QC samples were prepared by spiking

control liver homogenates with each metabolite prepared in 10% methanol with 0.1M HCL.

The intra-assay validation for cis-, trans-1- and trans-2- OH-perhexiline had CVs of 2.39,

3.23 and 3.20 and biases of -3.33, -5.33 and -4.67% for the highest calibrator (5 mg/L),

respectively. At the lowest calibrator cis-, trans-1- and trans-2- OH-perhexiline had CV’s

11.91, 12.45 and 10.65% and biases of -8, -12 and -16% respectively. The inter-assay

validation for cis-, trans-1- and trans-2- OH-perhexiline had CVs of 0.25, 0.17 and 0.18%

and biases of -1.80, -1.96 and -1.88% for the highest calibrator (5 mg/L), respectively. At the

lowest calibrator (0.05 mg/L) the CVs were 6.25, 8.2 and 5.39% and biases were 5.2, 5.2 and

-3.2% for cis-, trans-1- and trans-2- OH-perhexiline, respectively. Inter- and intra-assay

validations were performed with 6 replicates.

Two QC samples were run with each assay. Control liver homogenate was spiked with cis-,

trans-1- and trans-2- OH-perhexiline to final concentrations of 0.075 or 0.75 mg/L for QC1

and QC2, respectively. The inter-assay CV and bias at the lowest QC was 17 and 7% for cis-,

13 and 4% for trans-1- and 19 and 2% for trans-2- OH-perhexiline respectively. At the

highest QC the CV and bias was 13 and 4% for cis-, 10 and 1% for trans-1- and 13 and -2%

for trans-2- OH-perhexiline respectively

Plasma OH-perhexiline concentrations were measured by a published method that’s

established and validated in our laboratory [82]. As the method was a minor modification of a

previously published assay established in our laboratory, an abbreviated revalidation was

carried out by determining intra- and inter-assay accuracy and precision. The intra-assay

Page 82: The stereoselective pharmacodynamics of the enantiomers of

82

validation (n=6) for cis-, trans-1- and trans-2- OH-perhexiline had CVs of 7, 6 and 6% and

biases of 1.6, 0.4 and 0.4% for the highest calibrator (5 mg/L), respectively. At the lowest

calibrator cis-, trans-1- and trans-2- OH-perhexiline had CVs of 17, 19 and 19% and biases

of 4, 16 and 8% respectively. The inter-assay validation (n=3) for cis-, trans-1- and trans-2-

OH-perhexiline had CVs of 8, 5 and 6% and biases of -2, -5 and -5% for the highest calibrator

(5 mg/L), respectively. At the lowest calibrator (0.05 mg/L) the CVs were 12, 13 and 13%

and biases were -7, -13 and -13% for cis-, trans-1- and trans-2- OH-perhexiline, respectively.

Page 83: The stereoselective pharmacodynamics of the enantiomers of

83

2.8 Neutrophil Preparation

Neutrophils were prepared using a previously established method in our laboratory [113].

Briefly, 20 mL of blood was collected in 4 mL of 4.5% EDTA. The blood was centrifuged at

150 x g for 10 min, resulting in the separation of plasma from blood cells. The plasma was

discarded and a volume of Hanks Balanced Salt Solution (HBSS) equal to that of the

discarded plasma was added. The blood was then underlayed with Lymphoprep® (Axis-

Shield, Oslo, Norway) and spun at 500 x g for 30 min.

Figure 13. This figure shows the various phases following centrifugation of the cellular

fraction of blood through the Lymphoprep® medium.

The Lymphoprep separates the constitutive blood cells into different layers. As shown in

Figure 13, at the top is the media layer, followed by the lymphocytes and monocytes layer,

followed by the Lymphoprep and the red blood cell layer at the base. At the top of the red

blood cell layer and just below the Lyphoprep, there is a much smaller layer of neutrophils.

Layers above the neutrophil and red blood cell layers were aspirated and discarded. The red

blood cells were lysed with lysis buffer, spun down at 500 x g for 10 min and lysed blood

cells discarded, leaving only a neutrophil pellet. This was washed with 15 mL of HBSS

Media

Lymphocytes and Monocytes

Lymphoprep

RBC Neutrophils

Page 84: The stereoselective pharmacodynamics of the enantiomers of

84

resuspended and counted using trypan blue exclusion. The cells were then plated at 1.6 x 106

and incubated with (+)-, (-)-, or racemic perhexiline for 1 hr. at 37°C (as described in chapter

3). The cells were dark adapted and incubated for a further 15 min following the addition of

10 µM of the chemiluminescent agent lucigenin. A picolite luminometer (Packard

Instruments) was used to measure superoxide formation following the stimulation by 4 µM

fMLP (as described in Chapter 3). Superoxide formation was measured over a 5 min period

with the area under the chemiluminescence versus time curve (AUC) determined by the linear

trapezoidal method after subtracting blank values.

Concentration response curves were fitted by comparing sigmoidal dose response models

with a slope =1 or variable slope (hill equation). The sigmoidal dose response curve with a

variable slope was determined to be the more appropriate model by demonstrating more

stringent 95% confidence intervals as well as a greater coefficient of determination (R2).

 

Page 85: The stereoselective pharmacodynamics of the enantiomers of

85

Chapter   Three:   Lack   of   Clinically   significant  

Stereoselectivity   in   the   Inhibition   of   Human  

Neutrophil   Superoxide   Formation   by   (+)-­‐   and   (-­‐)-­‐

Perhexiline  

Text in manuscript. This manuscript is prepared for submission to and in accordance with the guideline of the journal Biochemical Pharmacology.  

Page 86: The stereoselective pharmacodynamics of the enantiomers of

86

There is a growing body of evidence suggesting that inflammation may play a crucial role in

the progression of cardiac disorders such as atherosclerosis, endothelial dysfunction and heart

failure. With an ageing population it is becoming increasingly difficult to treat such patients

without the use of invasive procedures that carry a higher morbidity and mortality rate thus

impacting quality of life. Therefore there is substantial emphasis for the development of

pharmacological agents to treat such patients.

The anti-anginal agent perhexiline has been reported to demonstrate anti-inflammatory

properties at a clinically relevant concentration. Due to its chiral properties perhexiline is

marketed as a racemate containing a 50:50 mixture of (+)- and (-)- perhexiline. However the

use of this drug has been limited due to its hepatic and neuronal toxicity and as such it is only

available in Australia and New Zealand under strict therapeutic drug monitoring.

There are currently no studies in the reported literature that investigate the toxicity of the

individual perhexiline enantiomers (Chapter 5) or their anti-inflammatory effects.

In this study we investigated the effects of (+)- and (-)-perhexiline on neutrophils, (a major

component in the inflammatory process responsible for the production of O2-, which is critical

in the eventual damage of cellular membranes and DNA) from healthy volunteers and patients

with stable angina. This study demonstrates that both enantiomers dose-dependently inhibit

O2- formation by neutrophil NADPH oxidase and that this inhibition is enantioselective

favouring (-)-perhexiline, providing further impetus to study the effects of the individual

perhexiline enantiomers in other disorders where inflammation may progress or even

contribute to the disease.

Page 87: The stereoselective pharmacodynamics of the enantiomers of

87

Lack of Clinically Significant Stereoselectivity in the Inhibition of Human Neutrophil

Superoxide Formation by (+)- and (-)-Perhexiline

Giovanni Licari 1, 2, Andrew A. Somogyi 1, John D Horowitz, 2,3, Robert W Milne, 4, Jennifer A

Kennedy, 2,3, Benedetta C Sallustio 1, 2

1 Discipline of Pharmacology, School of Medical Sciences, The University of Adelaide,

Adelaide, South Australia

2 Department of Cardiology and Clinical Pharmacology, The Queen Elizabeth Hospital,

Woodville, South Australia

3 Discipline of Medicine, School of Medicine, The University of Adelaide, Adelaide, South

Australia

4 School of Pharmacy and Medical Sciences, University of South Australia, Adelaide South

Australia

Corresponding Author: Associate Professor Benedetta Sallustio Department of Clinical Pharmacology The Queen Elizabeth Hospital The Basil Hetzel Institute for Medical Research 28 Woodville Road Woodville SA 5011 Phone 61 8 8222 6510 Fax 61 8 8222 6033 Email [email protected]

Reprint Request: To corresponding author, as above

Short Title: (+)- and (-)-Perhexiline Inhibit Superoxide Formation

Funding: Mr John Licari was the recipient of a Queen Elizabeth Hospital Research Foundation/ Adelaide University scholarship

Page 88: The stereoselective pharmacodynamics of the enantiomers of

88

Introduction

Atherosclerosis, diabetes, hypertension and heart failure are all disorders that have been

linked to endothelial dysfunction as a result of reactive oxygen species (ROS) [1]. NADPH

oxidase is one of the major sources of superoxide production in cardiovascular disease [1, 2].

Phagocytes such as neutrophils use the reactive superoxide anion as a defence against

invading pathogens via an inflammatory response. In addition, superoxide is also a crucial

component in the regulation of key physiological processes and enzymes such as apoptosis

and cellular signalling [3, 4]. The various isoforms of NADPH oxidase comprise differing

subunits that make up the enzyme. Phagocytic NADPH oxidase is made up of two membrane

bound subunits, p22phox and NOX-2, and three cytosolic subunits, p40phox, p47phox and p67phox.

This preassembled enzyme requires activation, which is accomplished by phosphorylation of

the p47phox subunit allowing interactions with the membrane subunits [5, 6].

Perhexiline, 2-(2,2-dicyclohexylethyl)piperidine, is an efficacious antianginal drug approved

for clinical use in Australia and New Zealand in patients who are refractory or contraindicated

to conventional drugs and/or surgical therapy. Its antianginal actions are believed to be a

result of the inhibition of carnitine palmitoyltransferase-1 (CPT-1), the rate limiting enzyme

in fatty acid oxidation [7]. This inhibition facilitates a shift in myocardial metabolism from

lipid to carbohydrate utilization, resulting in an oxygen sparing effect. This effect is highly

advantageous especially in periods of myocardial ischemia, since carbohydrate metabolism

utilises less oxygen to produce the same amount of adenosine tri-phosphate (ATP) when

compared to the oxidation of fatty acids [8-11]. However recent studies have shown that the

therapeutic effects of perhexiline extend beyond management of angina and the drug is also

beneficial in patients with acute coronary syndromes [12], inoperable aortic valvular stenosis

[13] and heart failure [14]. It has been shown that perhexiline increases platelet

responsiveness to nitric oxide and also decreases superoxide release by neutrophils [15]. The

Page 89: The stereoselective pharmacodynamics of the enantiomers of

89

latter is of interest as there is increasing evidence that nitric oxide availability is considerably

reduced as a result of its interaction with superoxide, leading to the formation of bioactive

compounds such as peroxynitrite which has been linked to hypercholesterolemia,

hypertension, endothelial dysfunction and heart failure [2, 16]. In a study by Kennedy et al.,

perhexiline was found to inhibit superoxide formation by NADPH oxidase present in human

neutrophils and human endothelial cells [17]. Considering that these two cell types are the

major source of superoxide in the cardiovascular system, this effect of perhexiline may

contribute to its beneficial effects in treating cardiovascular disease [17]. However, despite its

significant clinical efficacy, at high concentrations perhexiline can cause severe neurotoxicity

[18, 19], and hepatotoxicity [20] and its clinical use is guided by therapeutic drug monitoring.

Perhexiline is a chiral compound and is formulated and marketed as a racemic (50:50)

mixture of (+)- and (-)-enantiomers. Most studies involving perhexiline have been carried out

using the racemic formulation so that the pharmacological actions of the individual

enantiomers are relatively unknown. Enantiomers of the same drug can have differing effects

in the same biological environment. A classic example of this is the anticoagulant drug

warfarin, which was introduced for clinical use over 40 years ago. Warfarin exists as R- and

S-warfarin enantiomers with the pharmacological activity residing predominately with the S-

enantiomer [21]. In this study, we aimed to investigate the contribution of the individual

enantiomers to the pharmacological effects of perhexiline on superoxide formation by human

neutrophils from both healthy subjects and patients with stable angina pectoris.

Page 90: The stereoselective pharmacodynamics of the enantiomers of

90

Materials and Methods

Chemicals

Bis-N-methylacridinium nitrate (lucigenin), fMLP (N-formyl Met Leu Phe) and perhexiline

maleate were purchased from Sigma Chemical Company (Castle Hill, NSW, Australia). (+)-

and (-)-perhexiline (HCL salts) were synthesised and characterised as previously described

[22].

Methods

The study was approved by the Ethics of Human Research Committee at the Queen Elizabeth

Hospital. Eleven healthy subjects and 6 patients were recruited and gave written informed

consent to participate. Inclusion criteria for healthy subjects required them to be free from

illness and not taking any medication, including anti-inflammatory agents that might affect

neutrophil function. Patients were included if they were aged 18 years or over, were

scheduled for an angiogram or angioplasty at the Queen Elizabeth Hospital’s cardiac

catheterisation laboratory, and had symptomatic stable angina. They were excluded from the

study if they were receiving calcineurin inhibitors, corticosteroids, cytotoxic

immunosuppressants, immunosuppressant antibodies, sirolimus derivatives, azathioprine,

mycophenolate and any long acting nitrates or perhexiline.

Neutrophil Extraction

Blood (20 ml) was collected in a 50 mL tube containing 4% EDTA at a pH of 7.35 and

neutrophils extracted as per Kennedy et al [17]. Briefly, the blood was centrifuged at 150 x g

for 10 minutes and the subsequent plasma was discarded and replaced with an equal volume

of Hanks Balanced Salt Solution (HBSS) pH 7.4. Using a Ficoll–Hypaque gradient (Axis-

Page 91: The stereoselective pharmacodynamics of the enantiomers of

91

Shield, Oslo, Norway), blood components were separated into three layers after centrifugation

at 550 x g for 30 minutes. The upper and middle layers consisted of media, monocytes and

lymphocytes and were discarded. The lower layer, consisting of red blood cells and

neutrophils was resuspended, to the exact volume taken up by the upper and middle layers,

with lysis buffer (ammonium chloride 155 mM, EDTA 100 µM, NaHCO3 10 mM), in order to

lyse the red blood cells. The cells were then centrifuged at 500 x g for 10 minutes and the

supernatant discarded, leaving the neutrophil pellet. Neutrophils were resuspended with lysis

buffer re-centrifuged and then washed and resuspended in HBSS.

Chemiluminescence assay of superoxide

Extracted neutrophils were resuspended at 1.6x106 cells in a final volume of 300 µL of HBSS.

They were incubated for 45 min at 37°C in the presence of (+)-, or (-)-perhexiline at a final

concentration of 0.2, 0.5, 1, 2 and 10 µM or vehicle, and then dark adapted for a further 15

min. Superoxide release was stimulated by the addition of fMLP (4 µM final concentration)

and the response was measured in arbitrary chemiluminescent units by a luminometer

(Berthold Technologies, AutoLumat Plus LB953, Germany) over a 5 min period with the area

under the chemiluminescence versus time curve (AUC) determined by the linear trapezoidal

method after subtracting blank values.

Cell Viability

Neutrophil viability was measured by trypan blue exclusion and was not affected by the

incubation with (+)- or (-)-perhexiline, with up to 99% viability achieved (data not shown)

when compared to controls.

Page 92: The stereoselective pharmacodynamics of the enantiomers of

92

Data analysis

For each subject, superoxide AUC in samples incubated with vehicle was used as the value

for maximal superoxide formation and AUCs measured in the presence of test drugs were

expressed as a percentage of the maximal AUC. Concentration-response curves were

constructed describing percentage superoxide inhibition versus the log of the drug

concentration and the Hill equation used to derive individual IC50 values. Data were not

normally distributed and are presented as a median (range) IC50 values and paired

comparisons analysed non-parametrically by the Wilcoxon matched-pairs signed rank test.

Page 93: The stereoselective pharmacodynamics of the enantiomers of

93

Results

Neutrophils were extracted from 11 healthy subjects (6 males, 5 females ranging in age from

25 – 50 years). Both (+)- and (-)-perhexiline inhibited superoxide formation in a

concentration-dependent manner after stimulation by fMLP (Fig.1). The median (range) IC50

of (+) perhexiline was 1.64 µM (0.54-3.58µM), which was significantly higher (P<0.05)

when compared to that of (-)-perhexiline, 1.19 µM (0.55-2.01µM) (table 1).

Six patients diagnosed with angina participated in this study; their clinical status and

medications are shown in Table 2. Both enantiomers of perhexiline also inhibited superoxide

formation by neutrophils isolated from patients (Fig. 2). The IC50 values for (+)- and (-)-

perhexiline were not significantly different, with median values of 2.07 µM (1.04-2.95 µM)

and 1.35 µM (1.08-1.86 µM) respectively, and were similar to those observed with

neutrophils from healthy subjects (Table 1).

Median baseline superoxide AUC’s in healthy volunteers, 1.92 x 108 (0.56-4.12), were not

significantly different (P=1.00) to those in patient volunteers, 1.65 x 108 (0.64-4.58).

Page 94: The stereoselective pharmacodynamics of the enantiomers of

94

Discussion

Recent studies have suggested that the release of superoxide from activated neutrophils may

play a major role in the progression of acute coronary syndromes and in ischemic reperfusion

injury. Buffon et al. showed that in patients with unstable angina, neutrophils were activated

as they crossed the coronary vascular bed [23]. Activated neutrophils release superoxide

which not only damages the vascular endothelium contributing to its dysfunction, but also

mediates the recruitment of more neutrophils via the increased expression of adhesion

molecules [23]. Availability of nitric oxide, an important physiological vasodilatory factor in

the myocardial endothelium, is reduced by superoxide. Its reaction not only decreases nitric

oxide synthase expression but it also binds to free nitric oxide to produce peroxynitrite, a

powerful oxidising agent capable of inflicting damage to the myocardial cells’ DNA and

proteins [6, 16, 24].

The main finding of this study is that both (+)-perhexiline and (-)-perhexiline showed

significant inhibition of superoxide formation by neutrophils isolated from both healthy

subjects and patients diagnosed with myocardial ischemia. In both subject groups, the IC50

values for inhibition of superoxide formation were similar to those previously reported with

the racemic formulation [17] and were within the range of total plasma perhexiline

concentrations attained clinically. However, perhexiline is highly protein bound in plasma

[25] and unbound concentrations are unlikely to exceed 0.1 µM. Therefore the clinical

relevance of this effect may be small unless there is significant concentrations of perhexiline

within the neutrophil membrane following longer term exposures. Perhexiline is highly

concentrated within tissues [29] and it is possible that with chronic dosing, at steady state the

intracellular concentrations achieved clinically may be comparable to those attained following

the short-term (1hr) incubations utilised in this study.

Page 95: The stereoselective pharmacodynamics of the enantiomers of

95

In healthy subjects, (-)-perhexiline was a slightly more potent inhibitor of neutrophil

superoxide formation, with a median IC50 approximately 30% lower than that of (+)-

perhexiline (eudismic ratio = 1.37). However, this small difference is unlikely to be of clinical

significance, since, due to enantioselectivity in their metabolism [26], systemic exposure to

(+)-perhexiline is on average 1.5 to 2.5-fold greater than that of (-)-perhexiline. The higher

concentration of circulating (+)-enantiomer may therefore compensate for its slightly higher

IC50 values for superoxide inhibition. Thus, both enantiomers are likely to contribute to any

clinical anti-inflammatory effects of perhexiline via inhibition of superoxide formation.

The lack of clinically significant enantioselectivity in the inhibition of neutrophil superoxide

formation by (+)- and (-)-perhexiline is also supported by the observations in the patient

group, where both enantiomers inhibited superoxide formation in a concentration-dependent

manner with IC50 values comparable to those observed for healthy subjects. However, in the

patient group, we were unable to detect the small difference in potency probably due to the

smaller sample size. All of the patients participating in this study had hypertension and/or

angina. Two patients had other metabolic disorders such as diabetes and

hypercholesterolemia. All were treated with statins, 2 patients were treated with angiotensin

converting enzyme inhibitors and 2 were given medication to manage their diabetes (Table 1).

Despite the array of drugs these patients received, (+)-and (-)-perhexiline showed a similar

pattern in the inhibition of superoxide formation when compared to healthy volunteers.

Racemic perhexiline has previously been shown to directly inhibit neutrophil NADPH

oxidase activity [17]. Using phorbol myristate acetate (PMA) (a chemical which promotes the

assembly of the NADPH oxidase subunits), Kennedy et al. demonstrated that racemic-

perhexiline does not affect the assembly of the NADPH oxidase subunits in neutrophils [17],

and that it does not react directly with superoxide [17]. In this study, the slight

Page 96: The stereoselective pharmacodynamics of the enantiomers of

96

enantioselectivity with respect to inhibition of superoxide formation in healthy subjects

further supports direct inhibition of NADPH oxidase activity, since although enantiomers

share the same physical and chemical properties, their structural differences can affect their

orientation and interactions with proteins.

The inhibitory activity of perhexiline with respect to NADPH oxidase may extend to other

cells and tissues that express the enzyme. Vascular endothelial cells have been shown to

express the membrane bound NOX-2 and its subunits p40phox, p47phox, and p67phox,

which are the same units that make up the NADPH oxidase present in neutrophils [27, 28],

suggesting that NADPH oxidase may play an important role in controlling the tone of

vascular smooth muscle and therefore consolidating its contribution to cardiovascular

diseases. The current study has shown that both enantiomers of perhexiline inhibit the

formation of superoxide by neutrophils isolated from both healthy volunteers and patients

with angina. Furthermore, superoxide inhibition by (+)- or (-)-perhexiline did not appear to be

affected by the co-administration of drugs used to manage angina or other common

underlying diseases.

Page 97: The stereoselective pharmacodynamics of the enantiomers of

97

Table 1 Individual and median (range) IC50s and Hill slope coefficients for neutrophil

NADPH superoxide inhibition from A) 11 healthy volunteers and B) 6 patients (* indicates

P<0.05 versus (+)-perhexiline).

A)

(-)-Perhexiline (+)-Perhexiline

ID IC50 (µM) Hill Coefficient IC50 (µM) Hill Coefficient

1 0.88 1.24 0.90 2.15

2 1.31 0.92 2.46 1.29

3 1.19 1.26 0.68 0.62

4 1.94 1.78 3.58 1.71

5 1.65 1.54 1.64 1.95

6 1.12 1.29 2.36 2.49

7 0.55 0.77 0.54 1.53

8 2.02 2.29 2.80 1.40

9 0.59 2.42 0.61 3.65

10 1.92 1.45 2.49 0.95

11 0.59 1.21 1.23 0.87

Median

Range

1.19*

(0.55 – 2.02)

1.29

(0.77 – 2.42)

1.64

(0.54 – 3.58)

1.53

(0.62 – 3.65)

Page 98: The stereoselective pharmacodynamics of the enantiomers of

98

B)

(-)-Perhexiline (+)-Perhexiline

ID IC50 (µM) Hill Coefficient IC50 (µM) Hill Coefficient

1 1.86 4.77 2.95 0.87

2 1.23 1.45 1.04 0.96

3 1.47 0.96 2.21 1.58

4 1.08 1.85 1.92 3.52

5 1.18 2.06 1.35 1.71

6 1.45 1.46 2.21 0.96

Median

Range

1.34

(1.08 – 1.86)

1.65

(0.96 – 4.77)

2.06

(1.04 – 2.95)

1.27

(0.87 – 3.52)

Page 99: The stereoselective pharmacodynamics of the enantiomers of

99

Table 2 Patient’s clinical status and medications. Y.o= years old

Patient Age (years)/Sex Medication

1 Female

73

Simvastatin, irbesartan, esomeprazole

2 Male

59

Aspirin, atorvastatin, thyroxine, irbesartan, metformin,

ketoprofen and diltiazem

3 Male

47

Perindopril, clopidogrel, aspirin, diltiazem and

atorvastatin

4 Female

60

Aspirin, atenolol

5 Male

63

Aspirin, rosuvastatin, metoprolol

6 Male

86

Warfarin, esomeprazole, frusemide, clopidogrel,

atorvastatin, candesartan, salbutamol, levodopa,

meloxicam, tramadol and amitriptyline

Page 100: The stereoselective pharmacodynamics of the enantiomers of

100

Figure 1 Concentration-response curve for inhibition of superoxide formation in neutrophils

from a) healthy subjects and b) patients after incubation with (+)● or (-)▲ perhexiline and

stimulation by 4 µM fMLP. Data show median and range for 11 subjects. The median IC50

and Hill slope as in Table 2 were used to construct the line of best fit.

 

10-0.5 100.0 100.5 101.00

20

40

60

80

100 - Perhexiline+ Perhexiline

Concentration (µM)

% O

2- Inhi

bitio

n

10-0.75 10-0.25 100.25 100.75 101.25

0

20

40

60

80

100(-) Perhexiline(+) Perhexiline

Concentration (µM)

% O

2- Inhi

bitio

n

a)

b)

Page 101: The stereoselective pharmacodynamics of the enantiomers of

101

References

1. Fenster, B.E., P.S. Tsao, and S.G. Rockson, Endothelial dysfunction: clinical

strategies for treating oxidant stress. Am Heart J, 2003. 146(2): p. 218-26.

2. Kalinowski, L. and T. Malinski, Endothelial NADH/NADPH-dependent enzymatic

sources of superoxide production: relationship to endothelial dysfunction. Acta

Biochim Pol, 2004. 51(2): p. 459-69.

3. Li, P.F., R. Dietz, and R. von Harsdorf, Superoxide induces apoptosis in

cardiomyocytes, but proliferation and expression of transforming growth factor-beta1

in cardiac fibroblasts. FEBS Lett, 1999. 448(2-3): p. 206-10.

4. Stocker, R. and J.F. Keaney, Jr., Role of oxidative modifications in atherosclerosis.

Physiol Rev, 2004. 84(4): p. 1381-478.

5. Babior, B.M., J.D. Lambeth, and W. Nauseef, The neutrophil NADPH oxidase. Arch

Biochem Biophys, 2002. 397(2): p. 342-4.

6. Cave, A.C., et al., NADPH oxidases in cardiovascular health and disease. Antioxid

Redox Signal, 2006. 8(5-6): p. 691-728.

7. Kennedy, J.A., S.A. Unger, and J.D. Horowitz, Inhibition of carnitine

palmitoyltransferase-1 in rat heart and liver by perhexiline and amiodarone. Biochem

Pharmacol, 1996. 52(2): p. 273-80.

8. Stanley, W.C., Partial fatty acid oxidation inhibitors for stable angina. Expert Opin

Investig Drugs, 2002. 11(5): p. 615-29.

9. Rupp, H., A. Zarain-Herzberg, and B. Maisch, The use of partial fatty acid oxidation

inhibitors for metabolic therapy of angina pectoris and heart failure. Herz, 2002.

27(7): p. 621-36.

10. Lee, L., J. Horowitz, and M. Frenneaux, Metabolic manipulation in ischaemic heart

disease, a novel approach to treatment. Eur Heart J, 2004. 25(8): p. 634-41.

Page 102: The stereoselective pharmacodynamics of the enantiomers of

102

11. van der Vusse, G.J., M. van Bilsen, and J.F. Glatz, Cardiac fatty acid uptake and

transport in health and disease. Cardiovasc Res, 2000. 45(2): p. 279-93.

12. Stewart, S., et al., Relationship between plasma perhexiline concentration and

symptomatic status during short-term perhexiline therapy. Ther Drug Monit, 1996.

18(6): p. 635-9.

13. Unger, S.A., M.A. Robinson, and J.D. Horowitz, Perhexiline improves symptomatic

status in elderly patients with severe aortic stenosis. Aust N Z J Med, 1997. 27(1): p.

24-8.

14. Ashrafian, H., L. Williams, and M.P. Frenneaux, The pathophysiology of heart

failure: a tale of two old paradigms revisited. Clin Med, 2008. 8(2): p. 192-7.

15. Willoughby, S.R., et al., Beneficial clinical effects of perhexiline in patients with

stable angina pectoris and acute coronary syndromes are associated with potentiation

of platelet responsiveness to nitric oxide. Eur Heart J, 2002. 23(24): p. 1946-54.

16. Chirkov, Y.Y. and J.D. Horowitz, Impaired tissue responsiveness to organic nitrates

and nitric oxide: a new therapeutic frontier? Pharmacol Ther, 2007. 116(2): p. 287-

305.

17. Kennedy, J.A., et al., Effect of the anti-anginal agent, perhexiline, on neutrophil,

valvular and vascular superoxide formation. Eur J Pharmacol, 2006. 531(1-3): p. 13-

9.

18. Meier, C., et al., Perhexiline-induced lipidosis in the dark Agouti (DA) rat. An animal

model of genetically determined neurotoxicity. Brain, 1986. 109 ( Pt 4): p. 649-60.

19. Shah, R.R., et al., Impaired oxidation of debrisoquine in patients with perhexiline

neuropathy. Br Med J (Clin Res Ed), 1982. 284(6312): p. 295-9.

20. Le Gall, J.Y., et al., Perhexiline maleate toxicity on human liver cell lines. Gut, 1980.

21(11): p. 977-84.

Page 103: The stereoselective pharmacodynamics of the enantiomers of

103

21. Kaminsky, L.S. and Z.Y. Zhang, Human P450 metabolism of warfarin. Pharmacology

& therapeutics, 1997. 73(1): p. 67-74.

22. Davies, B.J., et al., Enantioselective assay for the determination of perhexiline

enantiomers in human plasma by liquid chromatography. J Chromatogr B Analyt

Technol Biomed Life Sci, 2006. 832(1): p. 114-20.

23. Buffon, A., et al., Widespread coronary inflammation in unstable angina. N Engl J

Med, 2002. 347(1): p. 5-12.

24. White, C.R., et al., Superoxide and peroxynitrite in atherosclerosis. Proc Natl Acad

Sci U S A, 1994. 91(3): p. 1044-8.

25. Inglis, S.C., et al., Effect of CYP2D6 metabolizer status on the disposition of the (+)

and (-) enantiomers of perhexiline in patients with myocardial ischaemia.

Pharmacogenetics and genomics, 2007. 17(5): p. 305-12.

26. Gould, B.J., A.G. Amoah, and D.V. Parke, Stereoselective pharmacokinetics of

perhexiline. Xenobiotica; the fate of foreign compounds in biological systems, 1986.

16(5): p. 491-502.

27. Jones, S.A., et al., Expression of phagocyte NADPH oxidase components in human

endothelial cells. Am J Physiol, 1996. 271(4 Pt 2): p. H1626-34.

28. Bayraktutan, U., et al., Expression of functional neutrophil-type NADPH oxidase in

cultured rat coronary microvascular endothelial cells. Cardiovasc Res, 1998. 38(1): p.

256-62.

29. Deschamps, D., et al., Inhibition by perhexiline of oxidative phosphorylation and the

beta-oxidation of fatty acids: possible role in pseudoalcoholic liver lesions.

Hepatology, 1994. 19(4): p. 948-61.

 

Page 104: The stereoselective pharmacodynamics of the enantiomers of

104

Chapter   Four:   Metabolism   and   Hepatotoxicity   of   the  

Myocardial   Metabolic   Agent   Perhexiline   in   Sprague  

Dawley  and  Dark  Agouti  Rats  

Text in manuscript. This manuscript is prepared for submission to and in accordance with the

guideline of the journal Drug Metabolism and Disposition.

Page 105: The stereoselective pharmacodynamics of the enantiomers of

105

As we have previously shown enantioselectivity for (-)-perhexiline in its anti-oxidant effects,

we now needed to test if (+)- or (-)- perhexiline demonstrated enantioselectivity towards

toxicity in an in vivo scenario. In order to do this an animal model that mimicked human

perhexiline metabolism and toxicity needed to be established.

The two candidates I decided for this study were the DA and the SD rat strains. The DA strain

has been extensively used as a poor metaboliser model for substrates for CYP2D6, whilst the

SD strain has been used as a model of extensive metabolism for substrates for CYP2D6.

Although a previous report involving dosing the DA rat with racemic perhexiline has been

published, there was no investigation of the metabolites of perhexiline or the validity as a

model of poor metabolism of perhexiline.

This publication was the first to report the metaboliser status of both the DA and SD strain

with regards to perhexiline metabolism. Although not a model of CYP2D6 poor metabolism,

the female DA rats attained plasma perhexiline concentrations in the mid-high end of the

clinical therapeutic range, with enantioselective disposition similar to humans and with a

biochemical marker of perhexiline-induced toxicity. Making the DA rat the preferable model

to conduct future toxicity and efficacy studies on (+)- and (-)- perhexiline

 

Page 106: The stereoselective pharmacodynamics of the enantiomers of

106

Metabolism and Hepatotoxicity of the Myocardial Metabolic Agent Perhexiline in

Sprague Dawley and Dark Agouti Rats

Giovanni Licari 1, 2, Andrew A. Somogyi 1, Robert W Milne, 3, Benedetta C Sallustio 1, 2

1 Discipline of Pharmacology, School of Medical Sciences, The University of Adelaide,

Adelaide, South Australia

2 Department of Clinical Pharmacology, Basil Hetzel Institute, The Queen Elizabeth

Hospital, Woodville, South Australia

3 School of Pharmacy and Medical Sciences, University of South Australia, Adelaide South

Australia

Page 107: The stereoselective pharmacodynamics of the enantiomers of

107

2. Running Title Page

Metabolism and Hepatotoxicity of Perhexiline

Corresponding Author: Associate Professor Benedetta Sallustio

Department of Clinical Pharmacology

The Queen Elizabeth Hospital

The Basil Hetzel Institute for Medical Research

37a Woodville Road

Woodville SA 5011

Phone: +61 8 8222 6510

Fax: +61 8 8222 6033

Email: [email protected]

Short Title: Pharmacodynamics of Perhexiline Enantiomers

Funding: Mr John Licari was the recipient of a Queen Elizabeth Hospital

Research Foundation/ Adelaide University scholarship

Text pages: 20

Tables: 1

Figures: 2

References: 36

Abstract: 279 words

Introduction: 847 words

Discussion: 964 words

Page 108: The stereoselective pharmacodynamics of the enantiomers of

108

Abbreviations:

ATP, adenosine triphosphate; CPT, carnitine palmitoyl transferase; CYP, cytochrome P450;

DA, Dark Agouti; EM, extensive metaboliser; HPLC, high performance liquid

chromatography; IM, intermediate metaboliser; PM, poor metaboliser; SD, Sprague Dawley;

UM, ultra rapid metaboliser.

Page 109: The stereoselective pharmacodynamics of the enantiomers of

109

Introduction

The anti-anginal agent, perhexiline is thought to improve myocardial function via inhibition

of the carnitine palmitoyltransferase (CPT) shuttle (CPT-1 and -2), causing a switch in

myocardial energy metabolism from the β-oxidation of fatty acids to the utilization of

carbohydrates (Kennedy et al., 2000), which require approximately 10-15 % less oxygen in

order to generate an equivalent amount of ATP (Kodde et al., 2007). More recently it has

become clear that perhexiline also exerts beneficial effects in acute coronary syndrome, heart

failure, hypertrophic cardiomyopathy and inoperable aortic stenosis, which may involve

additional mechanisms of action, including anti-oxidant effects (Horowitz et al., 2010; Lee et

al., 2005; Unger et al., 1997; Willoughby et al., 2002). Despite these beneficial effects,

perhexiline’s clinical use is limited, and requires therapeutic drug monitoring to individualise

dose, due to its potential to cause severe, concentration-dependent (Horowitz et al., 1986),

peripheral neuropathy (Singlas et al., 1978) and hepatotoxicity (Morgan et al., 1984). The

latter initially presenting as raised plasma liver enzymes including aspartate transaminase and

alkaline phosphatase and then developing into steatosis.

Perhexiline is a chiral molecule formulated as a racemic mixture of equal amounts of (+)- and

(-)-enantiomers. In humans, it is almost entirely metabolised by CYP2D6 to its primary

mono-hydroxy metabolites cis-OH-perhexiline, trans-1-OH-perhexiline and trans-2-OH-

perhexiline (Davies et al., 2007; Wright et al., 1973). We, and others, have previously

demonstrated significant enantioselectivity in its clinical pharmacokinetics, although both

enantiomers were substrates for CYP2D6 (Davies et al., 2006a; Gould et al., 1986; Inglis et

al., 2007). CYP2D6 is highly polymorphic (Ingelman-Sundberg et al., 2007; Sachse et al.,

1997; Zanger et al., 2004; Zhou et al., 2009) and, as a result, there is an approximately 10- to

20-fold inter-individual variability in the apparent oral clearance of perhexiline (Sallustio et

Page 110: The stereoselective pharmacodynamics of the enantiomers of

110

al., 2002), with clinical doses for CYP2D6 poor (PM), extensive (EM) and ultrarapid (UM)

metabolisers ranging from 100 mg/week, 100 - 200 mg/day and up to 500 mg/day,

respectively (Sallustio et al., 2002). The plasma concentration ratio of cis-OH-

perhexiline:perhexiline has been used to phenotype patients with respect to CYP2D6

metaboliser status, with a ratio < 0.3 indicating a CYP2D6 PM (Sallustio et al., 2002).

Significantly PM subjects do not form cis-OH-perhexiline and the trace amounts of

metabolites formed derive from CYP3A4 and CYP2B6 catalysed formation of the trans-OH

metabolites (Davies et al., 2007).

Altered myocardial energy metabolism is now recognised as a major feature of many forms of

heart disease and is being investigated as a novel therapeutic target (Fragasso et al., 2008;

Horowitz et al., 2010). Perhexiline may be a clinically useful prototypic drug to further

understand the mechanisms of action of myocardial metabolic agents. However, the

pharmacodynamics of its individual enantiomers have not previously been investigated. An

animal model with similar metabolism of perhexiline to humans is therefore needed to

investigate their mechanisms of action and relationships with pharmacokinetics. The female

Dark Agouti (DA) rat has been used extensively as a model of the human CYP2D6 PM

phenotype, whilst Sprague Dawley (SD) and Wistar rats have been traditionally used as

models of extensive metabolism (Schulz-Utermoehl et al., 1999). The aim of this study was to

compare female SD and female DA rats as in vivo models of perhexiline metabolism and

hepatotoxicity.

Page 111: The stereoselective pharmacodynamics of the enantiomers of

111

Materials and Methods

Materials

Perhexiline maleate, dansyl chloride, (R)-(−)-1-(1-napthyl)ethyl isocyanate (NEIC) and the

internal standard prenylamine lactate were purchased from Sigma Chemical Company (Castle

Hill, NSW, Australia), HPLC grade solvents (acetone, ethyl acetate, methanol and n-hexane)

and sodium hydroxide were purchased from Merck (Kilsyth, Victoria, Australia). All other

reagents were of analytical grade.

Animals- Dosing and Sampling

Animal ethics approval was granted from both the University of Adelaide and the Institute of

Medical and Veterinary Science animal ethics committees. Female SD and DA rats were

purchased from the University of Adelaide animal services. Animals were housed in a

temperature-controlled room with free access to water. For the first week, all animals were

fed standard rat chow and monitored for their daily consumption of food. The control group

(n=4) was administered pellets coated with vehicle (peanut paste, used to mask the bitter taste

of the perhexiline compound). During active treatment, 200 mg/kg of perhexiline maleate was

mixed with peanut paste and coated onto 7 rat chow pellets for SD rats (n=4), or 4 pellets for

DA rats (n=4), placed in the cages at 4-5 pm each day, and left until the next dose. Food and

perhexiline consumption was monitored daily and extra drug-free pellets provided if

necessary.

At the 8 week time point, the animals were euthanized under isoflurane (2-3%), blood was

taken via a cardiac puncture and livers were collected, snap frozen in liquid nitrogen and

stored at -80°C.

Page 112: The stereoselective pharmacodynamics of the enantiomers of

112

Biochemistry Assessments

Each blood sample was placed in a lithium heparin tube and plasma was obtained after

centrifugation at 3000 rpm at 10°C. Liver function was assessed by measuring plasma

concentrations of albumin, total bilirubin, alanine transaminase (ALT), aspartate transaminase

(AST), alkaline phosphatase (ALP) and gamma glutamyl transpeptidase (GGT). The analyses

were performed by the Clinical Chemistry Unit (Institute of Medical and Veterinary Science),

on a Beckman Coulter instrument (Gladesville, NSW, Australia).

Preparation of Liver Homogenates

Livers were thawed at room temperature in saline, then dried and weighed. Ice cold 0.15 M

phosphate buffer, pH 6.0 was added (2 ml per g liver), the liver was cut to smaller pieces,

transferred to a 50 mL glass tube and homogenised on ice (IKA® T10 basic Ultra-Turrax).

The homogenate was then poured into a glass tissue grinder on ice and ground with 5 passes

into a fine homogenate that was then assayed for perhexiline and metabolites.

Quantitation of perhexiline and its cis- and trans-OH-metabolites.

Concentrations of (+)- and (-)-perhexiline in plasma and liver homogenate were determined

using the method of Davies et al (Davies et al., 2006c). The concentrations of cis-, trans-1-

and trans-2-OH-(±)-perhexiline in plasma and liver homogenate were determined separately

by the method of Davies et al (Davies et al., 2006b). Although this assay resolves the cis-,

trans-1- and trans-2-OH metabolites of perhexiline, each is the sum of the metabolites formed

by both (+)- and (-)-perhexiline. Thus, plasma and liver perhexiline concentrations will also

be presented as total (±)-perhexiline concentrations, and the ratio of (+)/(-) enantiomers will

be shown separately. The assay for (+)-, (-)- perhexiline and OH-perhexiline was validated

Page 113: The stereoselective pharmacodynamics of the enantiomers of

113

with plasma and liver homogenate and demonstrated inter- and intra-assay coefficient of

variation (CV) and bias, within ±15% for the highest limit of quantitation (HLOQ), 2 mg/L

and within ±20% for the lowest limit of quantitation (LLOQ), 0.02mg/L. Samples above

HLOQ were diluted accordingly in the same matrix, to fit within the calibration range.

Electron Microscopy

Liver tissue was dissected into cubes of approximately 0.5 mm3, and fixed for one hour in

fixative (4% formaldehyde and 1.5% glutaraldehyde in sodium cacodylate buffer, pH 7.2).

The fixed tissue was post-fixed in 2% osmium tetroxide in sodium cacodylate buffer, en bloc

stained with 2% uranyl acetate and dehydrated through 70%, 90% and 100% ethanol. The

tissue was then processed through 1,2-epoxypropane, a 50/50 mixture of 1,2-epoxypropane

and Procure 812 resin (Electron Microscopy Sciences, Fort Washington, USA) and two

changes of 100% resin. Tissue and resin were transferred to Beem capsules and placed in an

oven overnight at 90 oC.

Survey sections of tissue blocks were cut with glass knives and stained with Toluidine Blue.

Thin sections were cut at approximately 100 nm thickness on a Porter-Blum ultra microtome

(Sorvall, Newtown, USA) using a diamond knife (Micro Star Technologies, Huntsville,

USA). Thin sections were stained with Reynolds’ lead citrate and examined in a Hitachi H-

600 transmission electron microscope (Tokyo, Japan 1983). A minimum of 2-3 sections were

examined for each tissue. Hepatic lipid vesicles and cytosolic glycogen were identified as

described in previous papers, and content was measured as a percentage of the field of view.

Page 114: The stereoselective pharmacodynamics of the enantiomers of

114

Statistical analysis

Between-group comparison of plasma and tissue concentrations of perhexiline, OH-

perhexiline, biomarkers for liver injury or glycogen and lipid contents was carried out using a

Mann-Whitney U test. Statistical analysis was performed with GraphPad Prism version 4.02

for Windows (San Diego, California USA), significance level adopted was P<0.05, all data

are presented as median (range).

Page 115: The stereoselective pharmacodynamics of the enantiomers of

115

Results

Plasma Perhexiline and OH-Perhexiline Concentrations

Plasma perhexiline concentrations were significantly different in DA rats compared to SD rats

(Table 1). DA rats treated with racemic perhexiline had 5.6-fold higher median plasma

concentrations compared to the SD strain (p<0.05). In both SD and DA rats treated with

racemate, plasma (+)-perhexiline was always present at a higher concentration than (-)-

perhexiline, with median ratios of (+)/(-) of 2.1 and 1.8, respectively (Table 1). Plasma cis-

OH-perhexiline concentrations were 2.6-fold greater for the DA compared to the SD strain

(p<0.05). Plasma concentrations of trans-1-OH- and trans-2-OH-perhexiline were below the

limit of quantification in both the SD and DA rats, and in both strains were lower than the

concentrations of cis-OH-perhexiline. The median ratio of plasma cis-OH-

perhexiline:perhexiline concentrations in DA (0.89, 0.50-1.56) was not significantly different

to that in SD (1.55, 0.44-5.50) (P=0.30).

Liver Homogenate Perhexiline and OH-Perhexiline Concentrations

Median tissue homogenate perhexiline concentrations were also significantly different

between the two rat strains (Table 1). The DA rat showed perhexiline concentrations 4.5-fold

higher than those of the SD strain (p<0.05). Similar to plasma, the ratio of (+)/(-) perhexiline

concentrations in liver was also approximately 2:1 in both strains. Cis-OH-perhexiline was

the major metabolite in both SD and DA livers (Table 1).

Relative accumulation of total (±)-perhexiline in liver versus plasma was 60- and 48-fold in

the SD and DA rats, respectively. In the DA rat livers, median cis-, trans-1- and trans-2-

metabolite concentrations were 28-, 31- and 33-fold higher than in plasma. In the SD strain

Page 116: The stereoselective pharmacodynamics of the enantiomers of

116

cis-OH-perhexiline concentrations were 23-fold higher in liver compared to plasma, however

it was not possible to determine the relative tissue accumulation of the trans-OH metabolites

as their plasma concentrations were below the limit of quantification. The median ratio of

hepatic cis-OH-perhexiline:perhexiline concentrations in DA (0.66, 0.31-0.91) was not

significantly different to that in SD (1.19, 0.13-5.26) (P=0.34).

Biochemistry

At 8 weeks, liver function tests on DA rats showed significantly elevated LDH (P<0.05) in

the perhexiline-treated group (250 (221-352) U/L) compared to the control group (146 (105-

159) U/L), but no significant differences in any of the other tests. The SD rats showed no

significant differences in liver function tests.

Histology

Representative electron micrographs of livers from SD and DA rats are shown in figure 1.

Cytoplasmic glycogen pools were clearly evident in the control SD rats (Figure 1A), and

control DA livers, although not as intense or as large as those from SD control livers (Figure

1C). All other cellular structures and organelles appeared morphologically similar with 1 or 2

lipid droplets present in the field of view. Electron micrographs from perhexiline treated SD

and DA rats are shown in Figures 2A and B, respectively. In each strain, glycogen and lipid

content were assessed by measuring their area as a percentage of the field view. Figure 2,

shows significantly lower (p<0.05) glycogen content in SD rats treated with perhexiline

compared to SD controls.

Page 117: The stereoselective pharmacodynamics of the enantiomers of

117

Discussion

The DA rat has been extensively used in studies as a model of human CYP2D6 PM (Barham

et al., 1994; Komura et al., 2005; Zysset et al., 1988). CYP2D6 is a major enzyme involved in

the metabolism of many drugs and its highly polymorphic nature makes it difficult to predict

the pharmacokinetics and pharmacodynamics of compounds that are extensively metabolised

by CYP2D6, often resulting in adverse effects or lack of efficacy (Ingelman-Sundberg, 2005).

The anti-anginal drug perhexiline is one such compound, with high plasma perhexiline

concentrations increasing the risk of severe neurotoxicity and hepatotoxicity (Singlas et al.,

1978), particularly in CYP2D6 PM.

In this study both the DA and SD strains had many similarities to humans with respect to

perhexiline metabolism. For example, the ratio of (+)- to (-)-perhexiline in plasma and liver

was approximately 2:1, consistent with previous clinical studies (Gould et al., 1986; Inglis et

al., 2007). Furthermore, similar to studies in humans (Amoah et al., 1986; Davies et al.,

2006b), cis-OH perhexiline was also the major metabolite formed in both the SD and DA

strains. The 5-fold higher plasma perhexiline concentrations in DA versus SD rats suggest

that the DA rat has a reduced capacity to clear perhexiline. However, unlike CYP2D6 PMs

that do not produce any cis-OH-perhexiline (Davies et al., 2007), the cis-metabolite was still

the major metabolite formed in the DA, with a plasma metabolic ratio of 0.89 similar to the

SD strain (1.55) but higher than the ratio of 0.3 used to identify PM in humans. Thus, the DA

rat is not as suitable a CYP2D6 PM model for perhexiline, consistent with previous

observations that this strain’s suitability as a model for CYP2D6 PM is substrate specific

(Komura et al., 2005). Despite the DA rat not being a true PM model we were able to attain

clinically relevant plasma concentrations of perhexiline, whereas in the SD rat they were all

below the lower limit of the clinical therapeutic range (0.15 mg/L).

Page 118: The stereoselective pharmacodynamics of the enantiomers of

118

In the rat, CYP2D1 and CYP2D2 enzymes perform the same function as human CYP2D6

(Hiroi et al., 2002; Matsunaga et al., 1989), catalysing the 4-hydroxylation of debrisoquine

(Ohishi et al., 1993; Schulz-Utermoehl et al., 1999). Yamato et al., have shown by

transfecting yeast with recombinant plasmids which encode for CYP2D1 and 2D2, that

although both these enzymes are involved in the hydroxylation of debrisoquine (Yamamoto et

al., 1998), CYP2D2 has a higher affinity for debrisoquine and therefore predominately

catalyses its 4-hydroxylation. CYP2D1 has a lower affinity and may contribute to 4-

hydroxylation at high substrate concentrations (Hiroi et al., 2002). Furthermore,

immunoblotting studies revealed that hepatic CYP2D2 levels in the female DA rat were 30- to

40- fold lower than those reported in female SD and Wistar rats (Schulz-Utermoehl et al.,

1999). Our observation of significant but lower metabolism of perhexiline in the DA rat

suggests that its hepatic concentrations were sufficiently high to enable CYP2D1-catalysed

metabolism of the OH-metabolites, accounting for the formation of cis-OH-perhexiline in the

female DA rats. Indeed, there was significant hepatic accumulation of perhexiline in both

strains, with similar liver:plasma concentration ratios in DA and SD rats of approximately

50:1 (Table 1). Alternatively, in the rat cis-OH-perhexiline may also be formed by CYPs

other than the 2D family.

Elevated plasma perhexiline concentrations have been linked to adverse effects such as

peripheral neuropathy and hepatotoxicity (Horowitz et al., 1986; Morgan et al., 1984; Singlas

et al., 1978), with elevated biochemical markers of hepatotoxicity such as alkaline

phosphatase and aspartate transaminase (Morgan et al., 1984). In the DA rats, LDH was the

only biochemical marker to be significantly elevated (2-fold compared to control, p<0.05),

whereas the SD strain showed no significant changes in any of the tests. Although LDH is a

general marker of tissue damage and not a specific marker of liver dysfunction it indicates

that the DA rat may be more sensitive to perhexiline toxicity, most likely due to the higher

Page 119: The stereoselective pharmacodynamics of the enantiomers of

119

systemic and liver exposure to perhexiline. The lack of clear biochemical evidence of

hepatotoxicity probably reflects the fact that only one DA rat had plasma perhexiline

concentrations above the clinical therapeutic range. However, Meir et al., also reported that

there were no changes in biochemical liver function tests in the DA rat after chronic

perhexiline administration (Meier et al., 1986). Alternatively the rat may be less sensitive to

perhexiline-induced hepatotoxicity.

Upon histological analysis, livers from SD rats treated with perhexiline had lower glycogen

contents (P<0.05) compared to vehicle treated animals (Figures 1 and 2), consistent with the

inhibition of CPT-1 by perhexiline, leading to increased carbohydrate utilisation and depletion

of glycogen stores. However, a similar effect was not observed in the DA rats. Liver biopsies

from patients with perhexiline-induced hepatotoxicity typically show steatosis and

phospholipidosis (Kopelman et al., 1977; McDonald, 1977). In this study, there was no

statistically significant lipid accumulation in either rat strain.

In summary, our results suggest that the pharmacokinetic properties of the female DA rat

make it a model of intermediate / extensive CYP2D6 metabolism of perhexiline. By utilising

the female DA rat we were able to achieve perhexiline concentrations within the

recommended human therapeutic range as well as demonstrate similar enantioselective

pharmacokinetics of perhexiline as observed in humans, whereas the concentrations achieved

with the SD rat were sub-therapeutic. In addition, the higher perhexiline concentrations in DA

rats were also associated with biochemical toxicity suggesting that for future investigations of

the toxicity or efficacy of perhexiline in vivo, the female DA rat may be the more suitable

model.

Page 120: The stereoselective pharmacodynamics of the enantiomers of

120

Table 1: Plasma and liver concentrations of parent perhexiline and OH-metabolites in DA

and SD rats dosed orally at 200 mg/kg/day for 2 months. (*P < 0.05, #P=0.057 compared to

DA)

DA strain SD strain

Plasma

(±)-perhexiline (mg/L)

Ratio (+):(-) perhexiline

Cis-OH-perhexiline (mg/L)

Trans-1-OH-perhexiline (mg/L)

Trans-2-OH-perhexiline (mg/L)

0.50 (0.16-1.13)

1.83 (1.67-2.32)

0.44 (0.25-0.57)

<LLOQ

<LLOQ

0.09 (0.04-0.13)*

2.13 (2.0-3.0)

0.17 (0.04-0.23)*

<LLOQ

<LLOQ

Liver

(±)-perhexiline (mg/Kg)

Ratio (+):(-) perhexiline

Cis-OH-perhexiline (mg/Kg)

Trans-1-OH-perhexiline (mg/Kg)

Trans-2-OH-perhexiline (mg/Kg)

24.47 (9.4-54.70)

2.25 (1.84-2.48)

12.66 (7.50-14.48)

1.22 (0.58-1.36)

1.14 (0.50-1.80)

5.42 (0.92-8.22)*

1.87 (1.24-2.14)

3.44 (0.71-8.50)#

0.46 (0.29-1.00)

0.15 (0.04-0.36)

Liver:Plasma Concentrations

(±)-perhexiline

Cis-OH-perhexiline

Trans-1-OH-perhexiline

Trans-2-OH-perhexiline

53.6 (37.2-60.0)

27.7 (23.7-35.7)

31.0 (28.1-45.3)

33.3 (21.2-60.0)a

55.6 (10.2-89.3)

23.3 (3.1-212.5)

N.D.

N.D.

Page 121: The stereoselective pharmacodynamics of the enantiomers of

121

Figure 1: Electron micrographs of liver tissue taken from A) the SD control group dosed with

vehicle for a period of 8 weeks (n=4), B) the SD group treated with perhexiline maleate

(200mg/kg/day), C) the DA control group treated with vehicle for 8 weeks (n=4), and D) the

DA group treated with perhexiline maleate (200mg/kg/day) for a period of 8 weeks (n=4).

Arrows represent glycogen stores within the hepatocytes. Magnification 30,000x.

A)

→ B)

C)

D)

Page 122: The stereoselective pharmacodynamics of the enantiomers of

122

Figure 2: Mean (s.e.m) of hepatic glycogen and lipid contents as a percentage of the field of

view in both the DA (A) and SD rat (B) strains, following chronic dosing of 200 mg/kg of

racemic perhexiline maleate (Rac) or vehicle control (Cont) over 8 weeks. *= P<0.05 versus

control group (Mann Whitney) N=4 for each group).

Page 123: The stereoselective pharmacodynamics of the enantiomers of

123

Acknowledgments John K Brealey, from the Electron Microscope Unit of The Queen Elizabeth Hospital,

conducted the electron microscopy work in this project. John Pierides from the department of

Surgical Pathology and Cytopathology carried out histological analysis for this project.

Page 124: The stereoselective pharmacodynamics of the enantiomers of

124

References

Amoah AG, Gould BJ, Parke DV, Lockhart JD (1986). Further studies on the

pharmacokinetics of perhexiline maleate in humans. Xenobiotica 16(1): 63-68.

Barham HM, Lennard MS, Tucker GT (1994). An evaluation of cytochrome P450 isoform

activities in the female dark agouti (DA) rat: relevance to its use as a model of the CYP2D6

poor metaboliser phenotype. Biochem Pharmacol 47(8): 1295-1307.

Davies BJ, Coller JK, James HM, Somogyi AA, Horowitz JD, Sallustio BC (2006a). The

influence of CYP2D6 genotype on trough plasma perhexiline and cis-OH-perhexiline

concentrations following a standard loading regimen in patients with myocardial ischaemia.

Br J Clin Pharmacol 61(3): 321-325.

Davies BJ, Coller JK, Somogyi AA, Milne RW, Sallustio BC (2007). CYP2B6, CYP2D6, and

CYP3A4 catalyze the primary oxidative metabolism of perhexiline enantiomers by human

liver microsomes. Drug Metab Dispos 35(1): 128-138.

Davies BJ, Herbert MK, Coller JK, Somogyi AA, Milne RW, Sallustio BC (2006b).

Determination of the 4-monohydroxy metabolites of perhexiline in human plasma, urine and

liver microsomes by liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci

843(2): 302-309.

Davies BJ, Herbert MK, Culbert JA, Pyke SM, Coller JK, Somogyi AA, et al. (2006c).

Enantioselective assay for the determination of perhexiline enantiomers in human plasma by

liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 832(1): 114-120.

Page 125: The stereoselective pharmacodynamics of the enantiomers of

125

Fragasso G, Salerno A, Spoladore R, Bassanelli G, Arioli F, Margonato A (2008). Metabolic

therapy of heart failure. Curr Pharm Des 14(25): 2582-2591.

Gould BJ, Amoah AG, Parke DV (1986). Stereoselective pharmacokinetics of perhexiline.

Xenobiotica 16(5): 491-502.

Hiroi T, Chow T, Imaoka S, Funae Y (2002). Catalytic specificity of CYP2D isoforms in rat

and human. Drug Metab Dispos 30(9): 970-976.

Horowitz JD, Chirkov YY (2010). Perhexiline and hypertrophic cardiomyopathy: a new

horizon for metabolic modulation. Circulation 122(16): 1547-1549.

Horowitz JD, Sia ST, Macdonald PS, Goble AJ, Louis WJ (1986). Perhexiline maleate

treatment for severe angina pectoris--correlations with pharmacokinetics. International

journal of cardiology 13(2): 219-229.

Ingelman-Sundberg M (2005). Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6):

clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J

5(1): 6-13.

Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C (2007). Influence of

cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic

and clinical aspects. Pharmacol Ther 116(3): 496-526.

Page 126: The stereoselective pharmacodynamics of the enantiomers of

126

Inglis SC, Herbert MK, Davies BJ, Coller JK, James HM, Horowitz JD, et al. (2007). Effect

of CYP2D6 metabolizer status on the disposition of the (+) and (-) enantiomers of perhexiline

in patients with myocardial ischaemia. Pharmacogenet Genomics 17(5): 305-312.

Kennedy JA, Kiosoglous AJ, Murphy GA, Pelle MA, Horowitz JD (2000). Effect of

perhexiline and oxfenicine on myocardial function and metabolism during low-flow

ischemia/reperfusion in the isolated rat heart. J Cardiovasc Pharmacol 36(6): 794-801.

Kodde IF, van der Stok J, Smolenski RT, de Jong JW (2007). Metabolic and genetic

regulation of cardiac energy substrate preference. Comp Biochem Physiol A Mol Integr

Physiol 146(1): 26-39.

Komura H, Iwaki M (2005). Pharmacokinetics and metabolism of metoprolol and propranolol

in the female DA and female Wistar rat: the female DA rat is not always an animal model for

poor metabolizers of CYP2D6. J Pharm Sci 94(2): 397-408.

Kopelman P, Morgan PG (1977). Liver damage after perhexiline maleate. Lancet 1(8013):

705.

Lee L, Campbell R, Scheuermann-Freestone M, Taylor R, Gunaruwan P, Williams L, et al.

(2005). Metabolic modulation with perhexiline in chronic heart failure: a randomized,

controlled trial of short-term use of a novel treatment. Circulation 112(21): 3280-3288.

Matsunaga E, Zanger UM, Hardwick JP, Gelboin HV, Meyer UA, Gonzalez FJ (1989). The

CYP2D gene subfamily: analysis of the molecular basis of the debrisoquine 4-hydroxylase

deficiency in DA rats. Biochemistry 28(18): 7349-7355.

Page 127: The stereoselective pharmacodynamics of the enantiomers of

127

McDonald GS (1977). Liver damage after perhexiline maleate. Lancet 1(8020): 1056.

Meier C, Wahllaender A, Hess CW, Preisig R (1986). Perhexiline-induced lipidosis in the

dark Agouti (DA) rat. An animal model of genetically determined neurotoxicity. Brain 109 (

Pt 4): 649-660.

Morgan MY, Reshef R, Shah RR, Oates NS, Smith RL, Sherlock S (1984). Impaired

oxidation of debrisoquine in patients with perhexiline liver injury. Gut 25(10): 1057-1064.

Ohishi N, Imaoka S, Suzuki T, Funae Y (1993). Characterization of two P-450 isozymes

placed in the rat CYP2D subfamily. Biochim Biophys Acta 1158(3): 227-236.

Sachse C, Brockmoller J, Bauer S, Roots I (1997). Cytochrome P450 2D6 variants in a

Caucasian population: allele frequencies and phenotypic consequences. Am J Hum Genet

60(2): 284-295.

Sallustio BC, Westley IS, Morris RG (2002). Pharmacokinetics of the antianginal agent

perhexiline: relationship between metabolic ratio and steady-state dose. British journal of

clinical pharmacology 54(2): 107-114.

Schulz-Utermoehl T, Bennett AJ, Ellis SW, Tucker GT, Boobis AR, Edwards RJ (1999).

Polymorphic debrisoquine 4-hydroxylase activity in the rat is due to differences in CYP2D2

expression. Pharmacogenetics 9(3): 357-366.

Page 128: The stereoselective pharmacodynamics of the enantiomers of

128

Singlas E, Goujet MA, Simon P (1978). Pharmacokinetics of perhexiline maleate in anginal

patients with and without peripheral neuropathy. Eur J Clin Pharmacol 14(3): 195-201.

Unger SA, Robinson MA, Horowitz JD (1997). Perhexiline improves symptomatic status in

elderly patients with severe aortic stenosis. Aust N Z J Med 27(1): 24-28.

Willoughby SR, Stewart S, Chirkov YY, Kennedy JA, Holmes AS, Horowitz JD (2002).

Beneficial clinical effects of perhexiline in patients with stable angina pectoris and acute

coronary syndromes are associated with potentiation of platelet responsiveness to nitric oxide.

Eur Heart J 23(24): 1946-1954.

Wright GJ, Leeson GA, Zeiger AV, Lang JF (1973). Proceedings: The absorption, excretion

and metabolism of perhexiline maleate by the human. Postgrad Med J 49: Suppl 3:8-15.

Yamamoto Y, Tasaki T, Nakamura A, Iwata H, Kazusaka A, Gonzalez FJ, et al. (1998).

Molecular basis of the Dark Agouti rat drug oxidation polymorphism: importance of CYP2D1

and CYP2D2. Pharmacogenetics 8(1): 73-82.

Zanger UM, Raimundo S, Eichelbaum M (2004). Cytochrome P450 2D6: overview and

update on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs Arch Pharmacol

369(1): 23-37.

Zhou SF, Liu JP, Chowbay B (2009). Polymorphism of human cytochrome P450 enzymes

and its clinical impact. Drug Metab Rev 41(2): 89-295.

Page 129: The stereoselective pharmacodynamics of the enantiomers of

129

Zysset T, Zeugin T, Kupfer A (1988). In-vivo and in-vitro dextromethorphan metabolism in

SD and DA rat. An animal model of the debrisoquine-type polymorphic oxidation in man.

Biochem Pharmacol 37(16): 3155-3160.

Page 130: The stereoselective pharmacodynamics of the enantiomers of

130

Chapter   Five:   The   enantiomers   of   the   myocardial  

metabolic   agent   perhexiline   display   divergent   effects  

on   hepatic   energy  metabolism   and   peripheral   neural  

function  in  Rats  

Text in manuscript. This manuscript is prepared for submission to and in accordance with the

guideline of the journal Drug Metabolism and Disposition

Page 131: The stereoselective pharmacodynamics of the enantiomers of

131

As we have now established our animal model of perhexiline metabolism and toxicity, we

now needed to examine if there is enantioselectivity in perhexiline induced toxicity. At a

biochemical level, the clinical toxicity of perhexiline manifests as an increase in alkaline

phosphatase and alanine transaminase suggesting a degree of liver injury. Histologically,

perhexiline has been shown to increase hepatic lipid content in the form of macro- and micro-

scopic steatosis, with appearance of lipid filled vesicles within the cytosolic compartment of

the hepatocyte. Peripheral neuropathy is the other major clinical toxicity associated with

perhexiline with patients experiencing loss of sensation in fingers as well as muscular

weakness.

This publication was the first to report the toxic effects of (+)- or (-)- perhexiline in an animal

model.

Perhexiline’s enantiomers exert substantially different effects on hepatic lipid and glycogen

accumulation, and on neural function. Our data suggest that the toxicity of racemic

perhexiline resides predominately in the (+)-enantiomer, consistent with inhibition of

carnitine palmitoyltransferase-1, whilst (-)-perhexiline may offer greater potential long-term

safety. Based on the finding of this study future development, with (-)-perhexiline as a

prototype may lead to the development of a safer formulation of perhexiline one which would

not require therapeutic drug monitoring.

Page 132: The stereoselective pharmacodynamics of the enantiomers of

132

The enantiomers of the myocardial metabolic agent perhexiline display divergent effects

on hepatic energy metabolism and peripheral neural function in rats

G. Licari1,2, R. W. Milne3, A. A. Somogyi2 and B. C. Sallustio1,2.

1Department of Clinical Pharmacology, The Queen Elizabeth Hospital, Woodville, South

Australia, Australia; 2Discipline of Pharmacology, University of Adelaide, Adelaide, South

Australia, Australia; 3School of Pharmacy and Medical Science, University of South

Australia, Adelaide, South Australia, Australia.

Corresponding Author: Associate Professor Benedetta Sallustio

Department of Clinical Pharmacology

The Queen Elizabeth Hospital

Basil Hetzel Institute for Translational Medical Research

28 Woodville Road

Woodville SA 5011

Phone: +61 8 8222 6510

Fax: +61 8 8222 6033

Email: [email protected]

Page 133: The stereoselective pharmacodynamics of the enantiomers of

133

Short Title: Pharmacodynamics of Perhexiline Enantiomers

Funding: Mr John Licari was the recipient of a Queen Elizabeth Hospital

Research Foundation/ Adelaide University scholarship

Text pages: 16

Tables: 2

Figures: 5

References: 30

Introduction: 622 words

Discussion: 2,534 words

A list of non-standard abbreviations used in the paper

ATP, adenosine triphosphate; CPT, carnitine palmitoyl transferase; CYP, cytochrome P450;

DA, Dark Agouti; HPLC, high performance liquid chromatography.

Key Words: Perhexiline, hepatotoxicity, neurotoxicity, enantioselectivity

Page 134: The stereoselective pharmacodynamics of the enantiomers of

134

Introduction

The anti-anginal agent perhexiline is approved in Australia for the treatment of angina

pectoris refractory to other therapies (Horowitz et al., 1979), however it also has been shown

to have clinically beneficial effects, including significant improvements in myocardial

energetics, in heart failure (Lee et al., 2005) and hypertrophic cardiomyopathy (Abozguia et

al., 2010), and it may also be useful in inoperable aortic stenosis (Unger et al., 1997).

Perhexiline inhibits carnitine palmitoyltransferase 1 (CPT-1) (Kennedy et al., 1996), which

forms part of the carnitine shuttle that translocates free fatty acids from the cytoplasm to the

mitochondrion in order to undergo β-oxidation. CPT-1 is the rate-limiting enzyme for this

pathway catalyzing the conversion of fatty acyl CoA to fatty acyl carnitine. Inhibition of

CPT-1 causes a “switching” to carbohydrate utilization, the so-called Randle phenomenon

(Hue et al., 2009): – it has been postulated that this switch is associated with an improvement

of approximately 13% in the efficiency of myocardial oxygen utilization.

Although modulation of myocardial energy metabolism is an emerging therapeutic target for

the treatment of cardiovascular disease (Lee et al., 2004) the clinical use of perhexiline has

been limited to otherwise refractory cases because of the potential risk of hepato- and neuro-

toxicity during long-term administration (Morgan et al., 1984; Singlas et al., 1978). This has

been attributed, in part, to lipid storage disorders in hepatocytes and Schwann cells, including

steatosis and lysosomal phospholipidosis, which have also been described with other CPT-1

inhibitors (Guigui et al., 1988). Although this problem generally remains subclinical provided

plasma perhexiline concentrations are kept within a defined therapeutic range (Horowitz et

al., 1986), it remains a major impediment to the routine use of the drug.

Perhexiline is a chiral compound formulated as a racemate, with prior investigations of its

enantiomers restricted to pharmacokinetic studies (Gould et al., 1986). Using an animal

Page 135: The stereoselective pharmacodynamics of the enantiomers of

135

model of potential perhexiline-induced hepato and neuro-toxicity, the aim of this study was to

test the hypothesis that the two enantiomers differ in their toxicological potential during long-

term therapy. Studies were performed in female Dark Agouti (DA) rats, which we have

previously shown are similar to humans with respect to perhexiline metabolism and

disposition, and the previously demonstrated facility for attaining clinically relevant plasma

perhexiline concentrations in this model (Licari et.al. manuscript in preparation).

Page 136: The stereoselective pharmacodynamics of the enantiomers of

136

Methods

Chemicals

(±)-Perhexiline maleate (racemate) was kindly provided by Sigma Pharmaceuticals (Rowville,

Victoria, Australia). Pure (+)- and (-)-perhexiline were prepared as either the maleate or

hydrochloride salts using a previously published method (Davies et al., 2006b). The

derivatising reagent (R)-(−)-1-(1-napthyl) ethyl isocyanate (NEIC) and the internal standard

prenylamine lactate were purchased from Sigma Aldrich (Castle Hill, NSW, Australia).

Methanol, acetone, ethyl acetate, n-hexane and sodium hydroxide were purchased from

Thermo Fisher (Scoresby, Australia); all solvents were of HPLC grade. All other reagents

were of analytical grade. DA rats were purchased from the University of Adelaide animal

facilities.

In vivo effects of (+)- and (-)-perhexiline in DA rats

Animals

The Adelaide University and the Institute of Medical and Veterinary Services animal ethics

committees granted approval for this study. Pilot experiments were conducted with racemic

perhexiline in order to develop dosing schedules associated with clinically relevant plasma

concentrations and the development of steatosis (Licari et.al., manuscript in preparation).

Thereafter, DA rats (n=4 in each group) were administered vehicle (peanut paste- control

group), or 200 mg/kg daily of (±)-, (+)- or (-)- perhexiline maleate for a period of 8 weeks.

After determining food intake (4 pellets for DA rats), perhexiline maleate was administered

mixed with peanut paste and coated onto standard rat chow and left until consumption

(overnight). On week 4 of dosing, blood was collected from the tail vein in order to assess

liver function as well as to determine plasma (+)-, (-)- and OH-perhexiline concentrations. On

Page 137: The stereoselective pharmacodynamics of the enantiomers of

137

week 8 of dosing, animals were anesthetised and blood was collected via a cardiac puncture in

order to assess liver function and determine the concentrations of perhexiline enantiomers and

the OH-metabolites, as well as lactate, glucose and triglyceride concentrations. Animals were

then euthanized and hepatic, cardiac and neuronal tissues were harvested in order to

determine perhexiline enantiomer and metabolite tissue concentrations and morphological

changes. Tissues were cut in half and either immediately snap frozen in liquid nitrogen or

placed into fixative solution for electron microscopy analysis.

Hepatic and Neuronal Morphology

Hepatic and neuronal tissue was dissected into cubes of approximately 0.5 mm in each

dimension and was fixed for one hour in electron microscopy fixative (4% formaldehyde and

1.5% glutaraldehyde in sodium cacodylate buffer, pH 7.2). The fixed tissue was post-fixed in

2% osmium tetroxide in sodium cacodylate buffer, en bloc stained with 2% uranyl acetate and

dehydrated through 70%, 90% and 100% ethanol. The tissue was then processed through 1,2-

epoxypropane, a 50/50 mixture of 1,2-epoxypropane and Procure 812 resin (Electron

Microscopy Sciences, Fort Washington, USA) and two changes of 100% resin. Tissue and

resin were transferred to Beem capsules and placed in an oven overnight at 90oC.

Survey sections of these hepatic and neuronal tissue blocks were cut with glass knives and

stained with Toluidine Blue. Thin sections were cut at approximately 100 nm thickness on a

Porter-Blum ultramicrotome (Sorvall, Newtown, USA) using a diamond knife (Micro Star

Technologies, Huntsville, USA). Thin sections were stained with Reynolds’ lead citrate and

examined in a Hitachi H-600 transmission electron microscope (Tokyo, Japan 1983).

Glycogen and lipids were identified (as described by Singh et al., 1997) and total content was

Page 138: The stereoselective pharmacodynamics of the enantiomers of

138

measured as a percentage of the field of view. Two to three sections were examined for each

tissue and the average value used.

Neuronal Function

At week 4 and 8, neuronal function was measured using von Frey filament testing of treated

and non-treated groups (Chaplan et al., 1994). The thickness of each filament corresponds to

a specific amount of pressure (in grams). Each animal was placed in a specially designed

plastic container, with the bottom replaced by mesh flooring. The animal was left for 10-15

minutes to settle into its new environment. In ascending order, von Frey filaments were

applied from beneath the mesh flooring to the plantar surface of the paw until the filament

buckled and was held there for a total of 10 seconds. A positive response was noted if the paw

was withdrawn within the 10-second period. If a filament induced 6 positive responses, the

grams corresponding to that filament were recorded as the paw withdrawal threshold.

Measurement of perhexiline and OH-perhexiline in plasma, liver and heart

Heart and liver homogenate was prepared on ice in 0.15 M phosphate buffer at pH 7.4, using

2 ml of buffer per gram of tissue. The homogenate was then stored at -80°C until analysis.

The concentration of (+)- and (-)- perhexiline in plasma and tissues (heart and liver

homogenate) was measured according to the method of Davies et al (Davies et al., 2006b).

The concentrations of cis-, trans1- and trans2-OH-perhexiline in plasma and liver

homogenate were measured according to the method of Davies et al (Davies et al., 2006b)

Page 139: The stereoselective pharmacodynamics of the enantiomers of

139

Statistical Analysis

Between-group comparison of plasma and tissue concentration of (+)- and (-)-perhexiline

were performed using Residual Maximum Likelihood (REML), treating treatments as fixed

and rats as random to take into account the paired nature of (+)- and (-)- perhexiline

concentrations following the administration of (±)- perhexiline, and the unpaired nature of

between group comparisons. Analyses were carried out using GenStat 13th edition (VSN

International Ltd, UK). Comparisons of cis-, trans1- and trans2-OH-metabolite formation

following administration of (+)- and (-)- perhexiline was performed using a two-way

ANOVA. Between-group comparisons of neuronal function, plasma liver function tests, and

tissue glycogen and lipid contents were carried out using a one-way ANOVA, with a

Bonferroni’s multiple comparisons post test. The relationship between (+)- and (-)-perhexiline

concentrations, and hepatic lipid or glycogen contents were also assessed using multiple

linear regressions. Data are expressed as mean ± S.E.M. All statistical analyses were

performed with GraphPad Prism version 4.02 for Windows, GraphPad software (San Diego

California USA), with a significance level of P<0.05 adopted.

Page 140: The stereoselective pharmacodynamics of the enantiomers of

140

Results

Plasma Biochemistry

Plasma liver function tests were carried out using samples collected at both the 4 and 8 week

time points (Table 1). There were no significant (P>0.12) changes in biochemistry at 4 weeks.

At 8 weeks, only plasma lactate concentrations were significantly higher in animals receiving

(-)-perhexiline when compared to controls (P<0.05), (+)-perhexiline (P<0.01) and (±)-

perhexiline (P<0.01).

Hepatic Histology

Representative electron micrographs for each treatment group are shown in Figure 1. Large

numbers of lipid-laden vacuoles were associated with (+)- perhexiline treatment (Figure 1B)

and large glycogen “pools” with (-)- perhexiline treatment (Figure 1C), whilst (±)-perhexiline

treatment (Figure 1D) resembled controls (Figure 1A).

When measured as a percentage of the area of the field of view, hepatic lipid and glycogen

content did not vary between control rats and those treated with racemic perhexiline.

However, in (+)- perhexiline-treated rats only, there was 6-, 4- and 8-fold higher hepatic lipid

content (p<0.05), compared to control, (-)-perhexiline and (±)-perhexiline, respectively

(Bonferroni post hoc test) (Figure 2A). Conversely, rats treated with (–)-perhexiline produced

2- and 14-fold higher hepatic glycogen content compared to controls (p < 0.01) and (+)-

perhexiline (p < 0.001, figure 2b), respectively.

In order to test the hypothesis that the enantiomers exert contrasting effects on lipid and

glycogen accumulation, correlations between hepatic enantiomer concentrations and effects

Page 141: The stereoselective pharmacodynamics of the enantiomers of

141

were sought (including baseline data from controls and racemate-treated animals). As shown

in Figure 3A, there was a direct correlation (r = 0.79, p = 0.004) between hepatic

concentration of (+)-enantiomer and lipid content, while a direct correlation was also seen (r =

0.78, p= 0.003) between (-)-enantiomer concentration and glycogen content (Figure 3B).

Furthermore, (+)-enantiomer concentrations were inversely correlated (r = -0.66, p = 0.03)

with glycogen content (Figure 3C).

Stepwise multiple regression confirmed diverging effects of both (+)- and (-)-perhexiline

concentrations on hepatic glycogen contents (p = 0.02 for (+)-perhexiline, p = 0.0004 for (-)-

perhexiline, adjusted R2 = 0.76), but only a significant effect of (+)-perhexiline on hepatic

lipid contents (p = 0.0004, adjusted R2 = 0.62).

Neural function and histology

Compared to controls, (-)-perhexiline treatment for 8 weeks (figure 2c) had no effect on

peripheral neural function. In contrast, rats treated with racemic perhexiline or (+)-perhexiline

revealed attenuation of response to the von Frey test compared to controls (p < 0.001 and p <

0.01, respectively) and to (-)-perhexiline (p < 0.001 and p < 0,05, respectively), consistent

with the induction of peripheral nerve dysfunction. Nevertheless, electron microscopic

examination of dorsal root ganglia revealed only occasional Mallory bodies, in approximately

1% of nerve cells from rats treated with (+)-perhexiline, with no Mallory bodies identified in

other treatment groups.

Page 142: The stereoselective pharmacodynamics of the enantiomers of

142

Perhexiline Concentrations

DA rats administered pure enantiomers attained plasma (+)-perhexiline concentrations of 0.67

(± 0.07, range 0.52-0.80) mg/L, whereas the concentrations in animals administered (-)-

perhexiline were 0.29 (± 0.04, range 0.22-0.39) mg/L. In rats treated with racemate, (±)-

perhexiline plasma concentrations averaged 0.84 ± 0.40 mg/L. The ratio of plasma (+)- to (-)-

perhexiline concentrations was approximately 2:1 irrespective of whether drug was

administered as racemate or individual enantiomers.

In order to make comparisons between animals treated with racemate versus those treated

with individual enantiomers, the plasma and tissue concentrations of (+)- or (-)-perhexiline

were corrected for the 2-fold difference in enantiomer dose, and are shown in Figure 4A

adjusted to the equivalent of a 100 mg/kg/day enantiomeric dose. When administered as

individual enantiomers, the concentrations of (+)-perhexiline were higher (p <0.05) than those

of (-)-perhexiline in liver and heart, but not plasma (Figure 4A). There was extensive (>20-

fold) tissue accumulation of both enantiomers, with higher tissue:plasma concentration ratios

for (+)- compared to (-)-perhexiline (p <0.05) in both heart and liver (Figure 4B).

Administration of (±)-perhexiline revealed significantly higher (p<0.05) concentrations of

(+)- compared to (-)-perhexiline in liver and plasma, but not heart (Figure 4A), with extensive

accumulation of both (+)- and (-)-perhexiline in tissues, although the ratio of tissue:plasma

concentrations was significantly higher (P<0.05) for (+)- compared to (-)-perhexiline only in

the heart. In addition, although the tissue:plasma concentration of (-)-perhexiline in heart and

liver following (±)-perhexiline was similar to that following the pure enantiomer, (+)-

perhexiline accumulation in heart and liver was significantly lower (p <0.05) following the

racemate compared to the pure enantiomer.

Page 143: The stereoselective pharmacodynamics of the enantiomers of

143

OH-Perhexiline Concentrations

In the DA rat perhexiline formed cis- and trans-mono-hydroxy metabolites (Figure 5). When

administered as the individual enantiomers, the cis-OH metabolite was the major metabolite

detected for both (+)- and (-)-perhexiline. Plasma cis-OH-(-)-perhexiline concentrations were

significantly higher (p<0.0001) than cis-OH-(+)-perhexiline concentrations, whereas the

concentrations of trans-1- and trans-2-OH metabolites showed no significant difference

between enantiomers (Figure 5A). In liver there was no significant difference in metabolite

concentrations between (+)- and (-)-perhexiline (Figure 5B). The mean (± s.e.m.) total

metabolite:parent ratio for (-)-perhexiline was significantly higher than that for (+)-

perhexiline, in both plasma (2.34 ± 0.45 vs. 0.39 ± 0.07, p<0.05) and liver (1.11 ± 0.30 vs.

0.18 ± 0.03, p<0.05). When administered as the racemate, total cis-OH-(±)-perhexiline was

similarly the major metabolite detected in plasma and liver. Unfortunately, our cis/trans-OH-

perhexiline assay did not resolve the metabolites of (+)- and (-)-perhexiline and therefore it

was not possible to compare the metabolism of the enantiomers following administration of

the racemate. However, it appears that following administration of racemic perhexiline, the

majority of cis-, trans-1- and trans-2-OH-metabolites derive from (-)-, (+)- and (-)-

perhexiline, respectively.

Page 144: The stereoselective pharmacodynamics of the enantiomers of

144

Discussion

Since its introduction in the 1970s, perhexiline has undergone meticulous scrutiny to

determine its mechanism(s) of toxicity and efficacy. Horowitz et al., postulated that

perhexiline was a potent CPT inhibitor and exerted its therapeutic effects via the inhibition of

CPT1 (Kennedy et al., 1996). However, more recent studies have shown that perhexiline

produced increased function in perfused rat hearts in the absence of any change in palmitate

oxidation, suggesting that the inhibition of CPT alone may not account for improved

myocardial function (Unger et al., 2005) and that other mechanisms, including its nitric oxide

sensitizing and anti-inflammatory properties may also be important (Kennedy et al., 2006;

Unger et al., 1997; Willoughby et al., 2002). Part of the difficulty in clearly determining the

mechanisms of action of perhexiline is the lack of investigation of the pharmacodynamic

properties of the individual enantiomers. This is the first study to investigate the effects of the

individual enantiomers on in vivo neuronal and hepatic function, the main targets of

perhexiline toxicity. We clearly show that, at clinically relevant concentrations, the two

enantiomers have significantly different effects on both hepatic and neuronal function.

(+)-Perhexiline, clearly produced concentration-dependent hepatic lipid accumulation (figures

2 and 3), consistent with the proposed mechanism of action of perhexiline, inhibition of CPT-

1, and previous clinical reports of hepatic steatosis (Lewis et al., 1979). In addition, (+)-

perhexiline also resulted in concentration-dependent decreases in hepatic glycogen contents

(Figures 2 and 3), also consistent with the inhibitory effects of this enantiomer on CPT-1

triggering a metabolic switch of energy sources from free fatty acid to carbohydrate

utilization (Kennedy et al., 1996) resulting in decreased glycogen formation. Kennedy et al.

have shown that racemic perhexiline inhibits CPT-1 with an IC50 of 77 and 148 µmol/L in

isolated cardiac and hepatic mitochondria, respectively (Kennedy et al., 1996). We

demonstrate that, following administration of the individual enantiomers, mean (s.e.m)

Page 145: The stereoselective pharmacodynamics of the enantiomers of

145

hepatic concentrations of (+)- and (-)-perhexiline were 104.9 (± 18.39) and 17.05 (± 1.4)

µg/g tissue, respectively, or, assuming a tissue density of 1 g/ml, 375 µM and 61.6 µM,

respectively. Thus, the hepatic concentrations of (+)-perhexiline were 2.5-fold higher than the

reported IC50, again consistent with inhibition of hepatic CPT-1.

Surprisingly, histology from the animals treated with the (-)-perhexiline revealed a completely

different metabolic effect, with (-)-perhexiline causing a concentration-dependent

accumulation of glycogen (figures 2 and 3), but no effect on hepatic lipid contents (Figure

2A). Thus, it can be hypothesised that, unlike (+)-perhexiline, (-)-perhexiline does not inhibit

hepatic CPT1, but may directly affect the pathways of carbohydrate utilization, a metabolic

effect unique to (-)-perhexiline. However, as the hepatic concentrations of (-)-perhexiline

were 2.5-fold lower than the reported IC50 for CPT-1 inhibition, we cannot rule out the

possibility that at higher concentrations (-)-perhexiline may also inhibit hepatic CPT-1.

The mechanism by which (-)-perhexiline causes glycogen accumulation is unclear. Insulin

stimulates glycogen synthesis and perhexiline may have some insulin sensitizing effects

(Willoughby et al., 2002), by a mechanism which is yet to be clearly defined. An insulin like

effect would be beneficial and similarly lead to enhanced myocardial glucose utilization and

function (Fath-Ordoubadi et al., 1997). The observation in isolated rat hearts that perhexiline

increased cardiac efficiency without any change in fatty acid oxidation, suggests a mechanism

of action that may not be CPT-1 dependent, but may include an effect of (-)-perhexiline on

carbohydrate/glycogen utilisation. Studies in primary hepatocyte cultures have also shown

glycogen accumulation following exposure to perhexiline (Lageron et al., 1981), and there

have been clinical reports linking the use of perhexiline to hepatic glycogen accumulation. In

one report, liver biopsies from two patients receiving perhexiline maleate revealed

characteristics similar to those seen in type-1 glycogen storage disease (Lageron et al., 1977).

Page 146: The stereoselective pharmacodynamics of the enantiomers of

146

Nonetheless, all of the metabolic changes in our study occurred without any significant

changes in traditional liver function test, except for an increase in plasma lactate in animals

treated with (-)-perhexiline.

Interestingly, (+)-perhexiline was also associated with neurotoxicity, since only animals

treated with (+)- or (±)-perhexiline showed increased paw withdrawal thresholds in the Von

Frey testing (Figure 5). This may suggest that inhibition of CPT-1 in neurons contributes to

perhexiline-induced peripheral neuropathy. Mier et al., also reported that racemic perhexiline

caused decreased peripheral neuronal function in DA rats, as well as inducing the

accumulation of Mallory bodies within dorsal root ganglia (Meier et al., 1986). Electron

microscopy of the dorsal root ganglion in our study revealed that only animals treated with

(+)-perhexiline had similar inclusions and lamellar bodies, however these were only present

in fewer than 1% of cells.

Formation of drug-induced Mallory bodies is a common characteristic seen in hepatic and

neuronal tissues after administration of cationic amphiphilic drugs, which can diffuse through

the lysosomal lipid membrane and become protonated in the intra-lysosomal acidic

environment (Kornhuber et al., 2010). Protonated perhexiline accumulates in lysosomes

forming complexes with phospholipids, preventing their degradation by lysosomal enzymes,

eventually resulting in phospholipidosis and Mallory body formation (Deschamps et al.,

1994). The development of hepatic steatosis has also been reported for other substances that

inhibit mitochondrial oxidation of fatty acids such as ethanol (Grunnet et al., 1986),

amiodarone (Fromenty et al., 1990) and ibuprofen (Freneaux et al., 1990). Deschamps et al.,

reported that, like other cationic amphiphilic drugs, perhexiline can also diffuse across the

outer mitochondrial membrane along the mitochondrial membrane potential, becoming

protonated in the intramembranous space where it accumulates and uncouples oxidative

Page 147: The stereoselective pharmacodynamics of the enantiomers of

147

phosphorylation causing a marked decrease in ATP levels as well as cell viability

(Deschamps et al., 1994). However, this direct mitochondrial toxicity occurs at higher

concentrations than those causing inhibition of β-oxidation (Deschamps et al., 1994).

Although both lysosomal and mitochondrial accumulation are unlikely to be enantioselective

as they depend on the amphiphilic nature of perhexiline, which should be the same for both

enantiomers, the higher concentrations of (+)-enantiomer may theoretically contribute to a

higher risk of lysosomal and mitochondrial dysfunction compared to (-)-perhexiline at the

same dose.

All of the observed metabolic changes in the DA rats were attained with plasma perhexiline

concentrations within the mid-high end of the clinical therapeutic range. We, and others, have

previously demonstrated that (-)-perhexiline is cleared more rapidly than (+)-perhexiline in

humans (Davies et al., 2007; Gould et al., 1986; Inglis et al., 2007), due to greater

metabolism of the (-)-enantiomer by CYP2D6 (Davies et al., 2007). Similar to humans, we

report significant enantioselectivity in the pharmacokinetics of perhexiline in DA rats. When

administered as the racemate, plasma concentrations of (+)-perhexiline were significantly

higher than those of (-)-perhexiline, consistent with greater clearance of the (-)-enantiomer,

When administered as the pure enantiomers, although not statistically significant, plasma

concentrations of (+)-perhexiline were again greater than those of (-)-perhexiline. However,

the lack of statistical significance may have been due to the small sample size, since the total

metabolite to parent ratio in both plasma and liver was statistically significantly greater for (-

)-compared to (+)-perhexiline, again consistent with greater metabolism of the (-)-enantiomer.

There was extensive tissue distribution of both perhexiline enantiomers and the metabolites,

with tissue:plasma concentration ratios >20, suggesting the involvement of uptake

transporters and/or significant tissue binding. Other studies have also reported significantly

Page 148: The stereoselective pharmacodynamics of the enantiomers of

148

higher concentrations of perhexiline in tissues compared to plasma (Davies et al., 2006a;

Meier et al., 1986). However, we demonstrate significantly different tissue distribution for

(+)- and (-)-perhexiline when administrations as the pure enantiomer but not when

administered as the racemate. In addition, administration of (+)-perhexiline as the pure

enantiomer resulted in tissue:plasma concentration ratios 2- to 3-fold higher than

administration of the racemic formulation. This suggests enantioselectivity in the net-uptake

of perhexiline and possible inhibition of the net-uptake of (+)-perhexiline by (-)-perhexiline or

the metabolites of (-)-perhexiline. Alternatively, (+)- perhexiline may have a higher affinity

for tissue binding than (-)- perhexiline, and may be displaced by (-)-perhexiline and/or the

metabolites of (-)-perhexiline. During incubation with human liver microsomes both

enantiomers are highly bound to microsomes, although the binding did not appear to be

enantioselective (Davies et al., 2007). Distribution of the individual enantiomers into

intracellular organelles may also be responsible for the difference in accumulation. Other

cationic amphiphilic drugs have been shown to accumulate up to 100- fold in organelles

within the cell (Kornhuber et al., 2010), and similar accumulation has been reported for

perhexiline in mitochondria and lysosomes (Deschamps et al., 1994), however, as previously

discussed this process is unlikely to be enantioselective.

Finally, the mechanism of CPT inhibition suggests that (+)- perhexiline may potentiate its

own accumulation in tissues, particularly liver where it causes significant lipid accumulation

(Figure 3B). The octanol-water partition coefficient (LogP) of perhexiline is approximately 7,

making it very lipophilic, allowing it to bind and diffuse into these lipid deposits and thereby

increasing its intra-hepatic volume of distribution as a result of its inhibition on CPT-1. This

would also explain the lower tissue:plasma concentration ratio of (-)- perhexiline as it does

not induce an accumulation of lipids (Figure 3D), and the lack of enantioselectivity following

Page 149: The stereoselective pharmacodynamics of the enantiomers of

149

administration of the racemic formulation, as both enantiomers could distribute equally into

lipid deposits.

In conclusion, the current study demonstrates that although chemically identical, the (+)- and

(-)- enantiomers of perhexiline have different effects on hepatic energy metabolism and

neuronal function. In DA rats, only (+)-perhexiline appeared to inhibit hepatic CPT1, causing

hepatic steatosis and peripheral neuronal dysfunction, as observed in clinical cases of

perhexiline toxicity. In contrast, (-)-perhexiline affected hepatic carbohydrate utilisation,

causing accumulation of glycogen. All of the observed metabolic changes occurred at

clinically relevant concentrations, and without significant changes in LFTs. Further studies

are clearly required to investigate the effects of (+)- and (-)-perhexiline on myocardial energy

metabolism and function, to determine whether an enantiomeric formulation of perhexiline

may have improved efficacy and/or safety.

Page 150: The stereoselective pharmacodynamics of the enantiomers of

150

Figure Legends

Figure 1: Transmission electron micrographs of livers from DA rats treated for 8 weeks with

A) vehicle, B) (+)-perhexiline, C) racemic perhexiline and D) (-)-perhexiline. Solid hatched

arrows indicate glycogen stores and solid arrows indicate lipid vesicles.

Figure 2: A) hepatic lipid content, B) hepatic glycogen content and C) Von Frey paw

withdrawal thresh holds in animals treated with vehicle, (+)- (n=3), (-)- (n=4) or (±)- (n=4)

perhexiline (*P<0.05 compared to controls, (-)- and (±)-perhexiline; ##P<0.01 compared to

controls; $P<0.001 compared to (-)-perhexiline).

Figure 3: Linear regression analyses of hepatic (+)-perhexiline concentration on A) lipid or

B) glycogen content, and (-)-perhexiline concentration on C) glycogen content and D) lipid

content.

Figure 4: (A) Dose-corrected plasma (n), hepatic (n) and myocardial (n) perhexiline

concentrations and (B) hepatic (n) and myocardial (n) tissue:plasma concentration ratios of

perhexiline in DA rats administered (+)-, (-)- or (±)-perhexiline over a 2 month period.

#P<0.05 c.f. (+)-PX in liver; *p<0.05 c.f. (+)-PX in heart; %p<0.05 c.f. R-(+)-PX in liver;

$p<0.05 c.f. R-(+)-PX in plasma; @p<0.05 c.f. R (+)-Px in heart.

Figure 5: Cis- (n), Trans-1- (n) and Trans-2- (n) OH-perhexiline concentrations in A) liver

and B) plasma, and C) liver:plasma concentration ratio in animals treated with vehicle, (+)-, (-

)- or (±)-perhexiline. Comparison between enantiomers and metabolites were performed using

a 2 way ANOVA (*P<0.05, ***P<0.01 compared to (+)- perhexiline). Racemic perhexiline

data is included for visual reference only and not for statistical analysis.

Page 151: The stereoselective pharmacodynamics of the enantiomers of

151

Table 1: Plasma biochemistry results following 4 and 8 weeks of dosing with vehicle

(control) or 200 mg/kg/day of racemic, (-)- or (+)- perhexiline in DA rats. (**P<0.01 c.f.

control). Data are expressed as mean ±SEM.

Control Racemic (-) (+) 4 weeks

Albumin (g/l)

21.75 (±1.50)

20.25 (±0.50)

22.5 (±1.29)

21.75 (±1.89)

Total Bilirubin (µmol/L)

1.75 (±0.50)

2.50 (±0.58)

2.25 (±0.50)

2.25 (±0.96)

GGT (U/L)

2.50 (±0.58)

1.75 (±0.96)

1.75 (±0.96)

2.25 (±0.96)

ALP (U/L)

97.00 (±4.08)

75.75 (±14.73)

83.25 (±6.29)

92.00 (±17.87)

ALT (U/L)

59.25 (±8.10)

53.50 (±3.70)

61.75 (±5.50)

71.50 (±10.34)

AST (U/L)

83.75 (±9.54)

78 (±3.00)

83 (±2.94)

175 (±134.37)

LDH (U/L)

189.75 (±71.29)

177.50 (±136.76)

140.00 (±44.40)

267.75 (±291.81)

8 weeks Albumin

(g/l) 19.00

(±1.41) 19.00

(±2.16) 19.75

(±0.96) 21.00

(±1.63) Total Bilirubin

(µmol/L) 2.00

(±0.58) 1.50

(±0.00) 0.75

(±0.50) 1.75

(±0.50) GGT (U/L)

1.25 (±0.50)

0.50 (±0.58)

1.75 (±1.50)

2.75 (±0.50)

ALP (U/L)

70.00 (±18.22)

80.75 (±8.42)

80.00 (±12.03)

61.25 (±10.24)

ALT (U/L)

66.50 (±14.82)

69.25 (±11.67)

62.00 (±16.43)

73.00 (±12.96)

AST (U/L)

108.75 (±43.80)

113.75 (±23.46)

166.75 (±81.63)

190.50 (±125.46)

LDH (U/L)

202.00 (±132.05)

268.25 (±58.45)

586.25 (±371.05)

375.25 (±234.63)

Glucose (mmol/L)

14.38 (±2.22)

15.28 (±0.75)

13.68 (±1.96)

13.13 (±5.77)

Lactate (mmol/L)

2.93 (±0.88)

2.23 (±0.24)

3.78 ** (±0.35)

2.03 (±0.57)

Triglycerides (mmol/L)

0.60 (±0.20)

0.78 (±0.22)

0.55 (±0.10)

0.55 (±0.07)

Page 152: The stereoselective pharmacodynamics of the enantiomers of

152

Figure 1.

Page 153: The stereoselective pharmacodynamics of the enantiomers of

153

Figure 2.

Page 154: The stereoselective pharmacodynamics of the enantiomers of

154

Figure 3.

Page 155: The stereoselective pharmacodynamics of the enantiomers of

155

Figure 4.

B)

A)

Page 156: The stereoselective pharmacodynamics of the enantiomers of

156

Figure 5.

Page 157: The stereoselective pharmacodynamics of the enantiomers of

157

Acknowledgments John K Brealey, from the Electron Microscope Unit of The Queen Elizabeth Hospital,

conducted the electron microscopy work in this project. John Pierides from the department of

Surgical Pathology and Cytopathology carried out histological analysis for this project.

Page 158: The stereoselective pharmacodynamics of the enantiomers of

158

References

Abozguia K, Elliott P, McKenna W, Phan TT, Nallur-Shivu G, Ahmed I, et al. (2010).

Metabolic modulator perhexiline corrects energy deficiency and improves exercise capacity in

symptomatic hypertrophic cardiomyopathy. Circulation 122(16): 1562-1569.

Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL (1994). Quantitative assessment of

tactile allodynia in the rat paw. J Neurosci Methods 53(1): 55-63.

Davies BJ, Coller JK, Somogyi AA, Milne RW, Sallustio BC (2007). CYP2B6, CYP2D6, and

CYP3A4 catalyze the primary oxidative metabolism of perhexiline enantiomers by human

liver microsomes. Drug Metab Dispos 35(1): 128-138.

Davies BJ, Herbert MK, Coller JK, Somogyi AA, Milne RW, Sallustio BC (2006a).

Determination of the 4-monohydroxy metabolites of perhexiline in human plasma, urine and

liver microsomes by liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci

843(2): 302-309.

Davies BJ, Herbert MK, Culbert JA, Pyke SM, Coller JK, Somogyi AA, et al. (2006b).

Enantioselective assay for the determination of perhexiline enantiomers in human plasma by

liquid chromatography. Journal of chromatography. B, Analytical technologies in the

biomedical and life sciences 832(1): 114-120.

Deschamps D, DeBeco V, Fisch C, Fromenty B, Guillouzo A, Pessayre D (1994). Inhibition

by perhexiline of oxidative phosphorylation and the beta-oxidation of fatty acids: possible

role in pseudoalcoholic liver lesions. Hepatology 19(4): 948-961.

Page 159: The stereoselective pharmacodynamics of the enantiomers of

159

Fath-Ordoubadi F, Beatt KJ (1997). Glucose-insulin-potassium therapy for treatment of acute

myocardial infarction: an overview of randomized placebo-controlled trials. Circulation

96(4): 1152-1156.

Freneaux E, Fromenty B, Berson A, Labbe G, Degott C, Letteron P, et al. (1990).

Stereoselective and nonstereoselective effects of ibuprofen enantiomers on mitochondrial

beta-oxidation of fatty acids. J Pharmacol Exp Ther 255(2): 529-535.

Fromenty B, Fisch C, Labbe G, Degott C, Deschamps D, Berson A, et al. (1990).

Amiodarone inhibits the mitochondrial beta-oxidation of fatty acids and produces

microvesicular steatosis of the liver in mice. J Pharmacol Exp Ther 255(3): 1371-1376.

Gould BJ, Amoah AG, Parke DV (1986). Stereoselective pharmacokinetics of perhexiline.

Xenobiotica 16(5): 491-502.

Grunnet N, Kondrup J (1986). The effect of ethanol on the beta-oxidation of fatty acids.

Alcohol Clin Exp Res 10(6 Suppl): 64S-68S.

Guigui B, Perrot S, Berry JP, Fleury-Feith J, Martin N, Metreau JM, et al. (1988).

Amiodarone-induced hepatic phospholipidosis: a morphological alteration independent of

pseudoalcoholic liver disease. Hepatology 8(5): 1063-1068.

Horowitz JD, Mashford ML (1979). Perhexiline maleate in the treatment of severe angina

pectoris. Med J Aust 1(11): 485-488.

Page 160: The stereoselective pharmacodynamics of the enantiomers of

160

Horowitz JD, Sia ST, Macdonald PS, Goble AJ, Louis WJ (1986). Perhexiline maleate

treatment for severe angina pectoris--correlations with pharmacokinetics. International

journal of cardiology 13(2): 219-229.

Hue L, Taegtmeyer H (2009). The Randle cycle revisited: a new head for an old hat. Am J

Physiol Endocrinol Metab 297(3): E578-591.

Inglis SC, Herbert MK, Davies BJ, Coller JK, James HM, Horowitz JD, et al. (2007). Effect

of CYP2D6 metabolizer status on the disposition of the (+) and (-) enantiomers of perhexiline

in patients with myocardial ischaemia. Pharmacogenet Genomics 17(5): 305-312.

Kennedy JA, Beck-Oldach K, McFadden-Lewis K, Murphy GA, Wong YW, Zhang Y, et al.

(2006). Effect of the anti-anginal agent, perhexiline, on neutrophil, valvular and vascular

superoxide formation. Eur J Pharmacol 531(1-3): 13-19.

Kennedy JA, Unger SA, Horowitz JD (1996). Inhibition of carnitine palmitoyltransferase-1 in

rat heart and liver by perhexiline and amiodarone. Biochem Pharmacol 52(2): 273-280.

Kornhuber J, Henkel AW, Groemer TW, Stadtler S, Welzel O, Tripal P, et al. (2010).

Lipophilic cationic drugs increase the permeability of lysosomal membranes in a cell culture

system. J Cell Physiol 224(1): 152-164.

Lageron A, Poupon R, de Saint-Maur PP, Levy VG (1977). Liver ganglioside storage after

perhexiline maleate. Lancet 1(8009): 483.

Page 161: The stereoselective pharmacodynamics of the enantiomers of

161

Lageron A, Scotto J, Gautier M (1981). Effects of Pexid on liver cell cultures. Ultrastructural

and histoenzymological studies. European journal of clinical pharmacology 19(6): 417-421.

Lee L, Campbell R, Scheuermann-Freestone M, Taylor R, Gunaruwan P, Williams L, et al.

(2005). Metabolic modulation with perhexiline in chronic heart failure: a randomized,

controlled trial of short-term use of a novel treatment. Circulation 112(21): 3280-3288.

Lee L, Horowitz J, Frenneaux M (2004). Metabolic manipulation in ischaemic heart disease, a

novel approach to treatment. Eur Heart J 25(8): 634-641.

Lewis D, Wainwright HC, Kew MC, Zwi S, Isaacson C (1979). Liver damage associated with

perhexiline maleate. Gut 20(3): 186-189.

Meier C, Wahllaender A, Hess CW, Preisig R (1986). Perhexiline-induced lipidosis in the

dark Agouti (DA) rat. An animal model of genetically determined neurotoxicity. Brain 109 (

Pt 4): 649-660.

Morgan MY, Reshef R, Shah RR, Oates NS, Smith RL, Sherlock S (1984). Impaired

oxidation of debrisoquine in patients with perhexiline liver injury. Gut 25(10): 1057-1064.

Singh A, Gilroy C, Chu I, Villeneuve DC (1997). Toxicity of PCB 105 in the rat liver: an

ultrastructural and biochemical study. Ultrastruct Pathol 21(2): 143-151.

Singlas E, Goujet MA, Simon P (1978). Pharmacokinetics of perhexiline maleate in anginal

patients with and without peripheral neuropathy. Eur J Clin Pharmacol 14(3): 195-201.

Page 162: The stereoselective pharmacodynamics of the enantiomers of

162

Unger SA, Kennedy JA, McFadden-Lewis K, Minerds K, Murphy GA, Horowitz JD (2005).

Dissociation between metabolic and efficiency effects of perhexiline in normoxic rat

myocardium. J Cardiovasc Pharmacol 46(6): 849-855.

Unger SA, Robinson MA, Horowitz JD (1997). Perhexiline improves symptomatic status in

elderly patients with severe aortic stenosis. Aust N Z J Med 27(1): 24-28.

Willoughby SR, Stewart S, Chirkov YY, Kennedy JA, Holmes AS, Horowitz JD (2002).

Beneficial clinical effects of perhexiline in patients with stable angina pectoris and acute

coronary syndromes are associated with potentiation of platelet responsiveness to nitric oxide.

Eur Heart J 23(24): 1946-1954.

Page 163: The stereoselective pharmacodynamics of the enantiomers of

163

Chapter  Six:  General  Discussion  

Page 164: The stereoselective pharmacodynamics of the enantiomers of

164

The progressive advancement in medical care has led to an increase in the average life

expectancy and ageing of the general population, leading to a spectrum and severity of

diseases and outcomes that were much less common in the past. Acute coronary disease,

angina and other cardiovascular diseases are therefore becoming more prominent in society.

These cardiovascular disorders could be considered "rich man diseases", along with diabetes

and metabolic syndrome X. In a world where life-threatening infectious diseases are now

more associated with developing and third world countries, the rich man diseases are major

causes of morbidity and mortality in developed countries.

As a result there is ever increasing pressure to develop new treatment modalities. Perhexiline

is an old drug with a remarkable spectrum of clinical efficacy, which is limited by its toxicity,

particularly in ‘at risk’ poor and intermediate metabolisers, and the need for ongoing

therapeutic drug monitoring to individualise dosage. Perhexiline is marketed in Australia as a

50:50 racemic mixture of the (+)- and (-)- enantiomers, raising the potential for a future,

enantiomerically-pure preparation that may avert some or all of the toxicity issues. However,

the safety and efficacy of the individual enantiomers, to our knowledge, have never been

described.

The main aim of my thesis was to investigate the pharmacodynamic properties of (+)- and (-)-

perhexiline in vitro and in vivo to determine whether (+)- perhexiline or its optical antipode is

responsible for the beneficial effects and toxicity seen in clinical practice.

I hypothesised that: the enantiomers of perhexiline may demonstrate enantioselectivity in the

reduction of neutrophil NADPH oxidase induced superoxide formation; and that the

enantiomers of perhexiline demonstrate enantioselectivity in hepato- and neuronal toxicity in

the DA animal model.

Page 165: The stereoselective pharmacodynamics of the enantiomers of

165

The first experiments described in Chapter 3, were intended to investigate one of the potential

beneficial effect of perhexiline and investigate the antioxidant potency of the enantiomers.

There is evidence suggesting that oxidative stress plays a significant role in the development

of acute myocardial diseases [114]. Reactive oxygen species (ROS) are major protagonists

involved in the oxidative stress mechanism and O2- undergoes a chemical reaction with nitric

oxide resulting in the formation of the peroxynitrite radical, causing DNA and/or membrane

damage, but it also reduces the bioavailability of nitric oxide resulting in endothelial

dysfunction, and other diseases.

Kennedy et al [9] reported that, at a therapeutic concentration, perhexiline was able to inhibit

O2- generation via neutrophil NADPH oxidase by approximately 60% and thus demonstrated

the versatility of this potent CPT-1 inhibitor. By inhibiting the production of O2-, perhexiline

could theoretically increase nitric oxide availability and thus reduce the development of

myocardial diseases such as endothelial dysfunction. These results strongly suggested that

perhexiline may have more than one mechanism of action and its therapeutic benefits do not

solely rely on inhibition of CPT-1.

My aim was to determine which of the perhexiline enantiomers inhibited neutrophil O2-

generation (as measured by superoxide lucigenin chemiluminescence) via NADPH oxidase. I

hypothesised that inhibition of O2- generation via neutrophil NADPH oxidase may be

enantioselective and hence only one enantiomer may be responsible for the activity of the

racemic mixture.

I have shown in Chapter 3 (manuscript 1), that the individual enantiomers of perhexiline are

able to inhibit O2- generation via neutrophil NADPH oxidase in a dose dependant manner and

although statistically there was a difference in their potency with IC50 values of 1.19 and 1.64

Page 166: The stereoselective pharmacodynamics of the enantiomers of

166

µM for (-)- and (+)- perhexiline, respectively. Clinically this is unlikely to be significant as (-

)- perhexiline is eliminated much more rapidly than (+)- perhexiline. IC50 values were similar

in healthy volunteer and patients. Furthermore, at clinically relevant concentrations, both (+)-

and (-)- perhexiline inhibited O2- generation via neutrophil NADPH oxidase by 60%, similar

to that previously reported by Kennedy et al [9]. This current study is the first to describe any

potential therapeutic characteristics of the individual enantiomers of perhexiline. It therefore

concurs that (-)- perhexiline may be a suitable replacement for racemic perhexiline as it does

exhibit the classical toxic effects associated with perhexiline (see chapter 5) as well as having

an equal potency towards its antioxidant effects.

In Chapter 4 (manuscript 2), I aimed to develop an animal model that would mimic the

CYP2D6 catalysed metabolism of perhexiline in humans.

My hypothesis was that if the individual enantiomers displayed differing pharmacokinetic

properties then these characteristics should also be present in my in vivo animal model to

properly investigate the enantiomers’ pharmacodynamic properties. This hypothesis was

tested by investigating the female SD animal as a model of CYP2D6 extensive metabolism

and the DA rat as a model of CYP2D6 poor metabolism, as previously described [76].

Following the administration of racemic perhexiline to the DA rat, the higher systemic

exposure to (+)- versus (-)- perhexiline was similar to that seen previously in humans [80]. In

this animal model, I was able to achieve plasma concentrations within the therapeutic range

and furthermore display the pattern of metabolite formation similar to that of humans. In the

SD, it was difficult to attain perhexiline concentrations within the clinical therapeutic range.

Based on the results from this study the female SD animal was not the most appropriate

model to test for perhexiline enantiomer toxicity. Plasma perhexiline levels in this model were

Page 167: The stereoselective pharmacodynamics of the enantiomers of

167

well below the therapeutic perhexiline range and hydroxy metabolites were also much lower

in those seen clinically in EMs; suggesting that this strain is very efficient in eliminating

perhexiline from its system, which may protect the animal from perhexiline associated

toxicity. On the other hand, our “poor metaboliser” model, the female DA rat strain, is not a

true model of perhexiline poor metabolism due to the presence of cis-OH- perhexiline in both

plasma and tissue, and a metabolic ratio that was similar to that of a human

intermediate/extensive metabolism of CYP2D6. However, in this model not only was I able to

achieve plasma perhexiline concentrations, which were within the plasma perhexiline

therapeutic range, but I also demonstrated that this model does indeed demonstrate a pattern

of metabolite formation which is similar to what we observe in humans. The results from this

project suggest that the DA animal model would be an appropriate one to determine future

toxicity and efficacy studies involving perhexiline and its enantiomers. Indeed, the female DA

does develop perhexiline-induced hepatic steatosis and neuronal dysfunction, as can be seen

in the electron micrograph images presented in Chapter 5, which resemble the perhexiline

toxicity described in a clinical setting. These findings suggest that one must be careful when

using the terms “Poor and Extensive CYP2D6 metaboliser” models, as the functional validity

of these terms depends on the drug(s) being tested, rather than the animal model adopted.

Therefore the female DA rat is an appropriate model to assess perhexiline toxicity in future

studies, however there is a need to be cautious when associating perhexiline toxicity to

metaboliser status when using this model.

In Chapter 5 (manuscript 3) my aim was to use the animal model developed in Chapter 4 to

test the perhexiline enantiomers for toxicity, particularly the toxicity which is associated with

perhexiline in a clinical setting. I hypothesised that one enantiomers may display the steatosis

and neurotoxicity associated with perhexiline while the other may not.

Page 168: The stereoselective pharmacodynamics of the enantiomers of

168

This study, to the best of my knowledge, is the first to give insight into the pharmacokinetics

and pharmacodynamics of the enantiomers of perhexiline, administered as an enantiomeric

formulation, in an animal model with metabolism similar to humans. The results confirmed as

per a previous human study by Davies et al [79], that the CYP2D enzymes involved in the

metabolism of perhexiline are enantioselective for (-)- perhexiline. The data indicate that cis-

hydroxy-(-)-perhexiline is present at much higher concentrations in blood, hepatic and

myocardial tissue than parent drug. In contrast, in the (+)- perhexiline-treated group, cis-

hydroxy-(+)-perhexiline was present at much lower concentrations than the parent compound

in all tissues. Furthermore it was observed that (+)-perhexiline was present at higher

concentrations in all tissues analysed, when compared to its antipode. Similar results could be

seen in the group administered racemic perhexiline, both in the enantioselectivity in

metabolism as well as the distribution into tissues. There were a few surprising results in this

study. The first when comparing the tissue and plasma concentrations of (+)-perhexiline

administered as a racemic or enantiomeric formulation. When administered as an

enantiomeric formulation, (+)-perhexiline showed a hepatic to plasma ratio of approximately

150:1, whereas administered as a racemic formulation, the ratio was 50:1. However, there was

no significant difference in plasma concentrations whether (+)-perhexiline was administered

in its enantiomeric or racemic formulation. These results may have unmasked an

enantioselective transporter effect that seems to favour the uptake of (+)-perhexiline or, as

reported in Chapter 5, (-)-perhexiline may play an inhibitory role in the uptake or efflux of

(+)-perhexiline in or out of the cell, when administered as racemic formulation. These studies

confirmed that the two enantiomers are pharmacokinetically different and there may be

enantioselectivity, not only in metabolism, but also in transport in and out of tissues.

The other unexpected finding in this series of studies was that the pharmacodynamics of the

two enantiomers differed significantly. It is known that perhexiline is a potent CPT-1 inhibitor

Page 169: The stereoselective pharmacodynamics of the enantiomers of

169

[69], and this is believed to be the mechanism of action that gives it its therapeutic benefits.

However it is evident from the numerous clinical case reports, that once above the therapeutic

plasma concentration range (>0.6 mg/L) for an extended period, perhexiline can cause hepato-

and neuro- toxicities. The micro- and macro-scopic features associated with this toxicity are

attributed to the increased lipid and phospholipid deposits within the hepatocyte as well as the

neuron, suggesting that CPT-1 inhibition may also contribute to its toxic effects. The electron

microscopy data from this hepatotoxicity study clearly demonstrated that, following oral

administration of (+)-perhexiline, this group of DA rats displayed a significant accumulation

of lipid within the hepatocyte compared to either the control or the (-)-perhexiline-treated

groups. I also noted that the increase in lipid deposit occurred in a dose-dependent manner as

explained in Chapter 5. Furthermore, neuro-toxicity, as assessed via Von frey filament

analysis, was greatest for (+)-perhexiline. This was further established in electron

micrographs of the dorsal root ganglia where Mallory bodies were only found in those rats

treated with (+)-perhexiline, although this was only in 1% of cells. These results indicate that

the steatosis seen in a clinical setting of perhexiline-induced toxicity may be attributed to the

presence of (+)-perhexiline. Surprisingly, upon examination of the electron microscope

images of hepatocytes from the (-)-perhexiline-treated group, I observed a different metabolic

effect. Instead of an increase in lipid, I observed an increase in glycogen. There is evidence

from various case reports that glycogen accumulates within the cell after administration of

racemic perhexiline [115].

At the beginning of this project, the overall aim was to consider whether one could develop an

enantiomerically pure perhexiline formulation that would be more efficacious and/or less

toxic than the current racemic mixture. The main hurdle in determining this was (as

mentioned in section 1.4 of this thesis) that perhexiline seems to have more than one

Page 170: The stereoselective pharmacodynamics of the enantiomers of

170

mechanism of therapeutic action and furthermore it is still not clear whether inhibition of

CPT-1 is its primary therapeutic mechanism of action.

The experiments described in Chapter 3 were a replication and extension of the study of

Kennedy et al [9] that showed both the enantiomers inhibited the production of superoxide via

neutrophil NADPH oxidase with the same potency as the racemic counterpart, providing

evidence that the perhexiline enantiomers both have therapeutic potential. In Chapter 4, I

tested and refined a model to assess the toxicity of racemic, (+)- or (-)- perhexiline, and in

Chapter 5, I tested these compounds and obtained good evidence that the enantiomers are

different both in their pharmacokinetic and pharmacodynamic properties.

The major limitation throughout this project was the availability of the pure form of each

enantiomer. The enantiomers were synthesised in house at the University of Adelaide

chemistry department. This was an expensive process costing approximately $1000 per mg of

enantiomers produced. As a result of this expensive process as well as the available funding at

the time, I was restricted to treating only 4 rats with (+)- or (-)-perhexiline, although each

enantiomer demonstrated different and significant effects in cellular energetics. If pure forms

of (+)- and (-)- perhexiline had been readily available, there would have been an opportunity

to establish dose-response relationships to determine if these effects were concentration

dependent.

Although in Chapter 3 I determined that the enantiomers of perhexiline had effective anti-

inflammatory properties as measured by superoxide inhibition, I was unable to investigate the

major site of action and conclusively determine what effects perhexiline enantiomers had on

the myocardium. I did not know whether myocardium accumulated glycogen or lipid after

administration of (-)- or (+)- perhexiline, respectively, as in liver. I was unable to determine if

Page 171: The stereoselective pharmacodynamics of the enantiomers of

171

these effects were beneficial or detrimental to the myocardium. Hence future studies might

consider these experiments. Furthermore, one might consider performing Langendorff heart

studies where (±), (+)- or (-)-perhexiline could be perfused and compare any changes in

myocardial contractile function and heart rate, while at the same time comparing effects on

energetics, resulting in a more conclusive therapeutic profiling of the individual enantiomers.

In summary, in consideration of hypotheses set out in Chapter 1, I have concluded the

following: hypothesis 1) is partially accepted as both (+)- and (-)-perhexiline have an equally

potent effect as assessed by IC50 values on superoxide inhibition via neutrophil NADPH

oxidase. Hypothesis 2 can be accepted, as I was able to investigate the pharmacodynamic

properties of both perhexiline enantiomers in a rat model of perhexiline toxicity, with plasma

concentrations that are observed clinically.

In conclusion, with respect to the findings within this thesis, clinical administration of

perhexiline is essentially the administration of two drugs with very different properties. It is

evident from the data presented in this thesis that each enantiomer affects cellular metabolism

in a different way. (+)-Perhexiline appears to impede lipid metabolism resulting in steatosis, a

common effect seen in a clinical setting of perhexiline toxicity. In contrast, (-)-perhexiline

causes an accumulation of glycogen, the implications of which are yet to be elucidated. Each

enantiomer of perhexiline has an effect on the two cellular pathways that are responsible for

the production of cellular energy required for cell function.

Further studies need to be conducted which would help address the gaps in our current

knowledge. As we now have a toxic and partial therapeutic profile of the perhexiline

enantiomers, future studies could consider: (a) the dose-response relationships for each

enantiomer on the female DA rat, (b) the potency of the individual enantiomers for inhibition

Page 172: The stereoselective pharmacodynamics of the enantiomers of

172

of CPT-1, (c) what transporters are involved in both the uptake and efflux of perhexiline, (d)

whether these transporters are enantioselective, and (e) the efficacy of the individual

enantiomers in an animal model of angina or heart failure. Do both enantiomers display

therapeutic effects? What is the effect of glycogen accumulation in the liver, heart and whole

organism? Is glycogen accumulation therapeutic? Can perhexiline enantiomers be used to

decrease risk of myocardial damage pre or post surgery? These are the kind of questions that

need investigating and may one day lead to the development of a marketable

enantiomerically-pure drug with decreased toxicity and increased efficacy for a variety of

myocardial disorders.

Page 173: The stereoselective pharmacodynamics of the enantiomers of

173

BIBLIOGRAPHY  

 1. Ashrafian, H., J.D. Horowitz, and M.P. Frenneaux, Perhexiline. Cardiovasc Drug Rev,

2007. 25(1): p. 76-97.

2. Burns-Cox, C.J., et al., Clinical evaluation of perhexiline maleate in patients with

angina pectoris. British medical journal, 1971. 4(5787): p. 586-8.

3. Inglis, S. and S. Stewart, Metabolic therapeutics in angina pectoris: history revisited

with perhexiline. Eur J Cardiovasc Nurs, 2006. 5(2): p. 175-84.

4. Bakke, O.M., et al., Drug safety discontinuations in the United Kingdom, the United

States, and Spain from 1974 through 1993: a regulatory perspective. Clinical

pharmacology and therapeutics, 1995. 58(1): p. 108-17.

5. Grupp, I.L., C.A. Bunde, and G. Grupp, Effects of perhexiline maleate on exercise-

induced tachycardia. J Clin Pharmacol J New Drugs, 1970. 10(5): p. 312-5.

6. Roberts, L.N. and G.P. Mason, Clinical trial of a new antianginal drug: perhexiline

maleate. J Clin Pharmacol New Drugs, 1972. 12(8): p. 342-8.

7. Horowitz, J.D., et al., Perhexiline maleate treatment for severe angina pectoris--

correlations with pharmacokinetics. Int J Cardiol, 1986. 13(2): p. 219-29.

Page 174: The stereoselective pharmacodynamics of the enantiomers of

174

8. Cole, P.L., et al., Efficacy and safety of perhexiline maleate in refractory angina. A

double-blind placebo-controlled clinical trial of a novel antianginal agent.

Circulation, 1990. 81(4): p. 1260-70.

9. Kennedy, J.A., et al., Effect of the anti-anginal agent, perhexiline, on neutrophil,

valvular and vascular superoxide formation. Eur J Pharmacol, 2006. 531(1-3): p. 13-

9.

10. Kennedy, J.A., et al., The effects of perhexiline on the rat coronary vasculature. Eur J

Pharmacol, 1999. 370(3): p. 263-70.

11. Horowitz, J.D. and M.L. Mashford, Perhexiline maleate in the treatment of severe

angina pectoris. The Medical journal of Australia, 1979. 1(11): p. 485-8.

12. Unger, S.A., M.A. Robinson, and J.D. Horowitz, Perhexiline improves symptomatic

status in elderly patients with severe aortic stenosis. Aust N Z J Med, 1997. 27(1): p.

24-8.

13. Abozguia, K., et al., Metabolic modulator perhexiline corrects energy deficiency and

improves exercise capacity in symptomatic hypertrophic cardiomyopathy. Circulation,

2010. 122(16): p. 1562-9.

14. Reports from the Australian Institute of Health and Welfare (AIHW). International

journal of technology assessment in health care, 1995. 11(3): p. 624-6.

Page 175: The stereoselective pharmacodynamics of the enantiomers of

175

15. Hacker, S., Australian Institute of Health and Welfare (AIHW) 2004. Heart, stroke

and vascular diseases—Australian facts 2004. AIHW and National Heart Foundation

of Australia (Cardiovascular Disease Series No. 22), 2004.

16. Zhang, Y., et al., Modulating endothelial nitric oxide synthase: a new cardiovascular

therapeutic strategy. American journal of physiology. Heart and circulatory

physiology, 2011. 301(3): p. H634-46.

17. Cave, A.C., et al., NADPH oxidases in cardiovascular health and disease.

Antioxidants & redox signaling, 2006. 8(5-6): p. 691-728.

18. Cave, A., Selective targeting of NADPH oxidase for cardiovascular protection.

Current opinion in pharmacology, 2009. 9(2): p. 208-13.

19. Lambeth, J.D., NOX enzymes and the biology of reactive oxygen. Nature reviews.

Immunology, 2004. 4(3): p. 181-9.

20. Li, J.M., et al., Activation of NADPH oxidase during progression of cardiac

hypertrophy to failure. Hypertension, 2002. 40(4): p. 477-84.

21. Bayraktutan, U., L. Blayney, and A.M. Shah, Molecular characterization and

localization of the NAD(P)H oxidase components gp91-phox and p22-phox in

endothelial cells. Arterioscler Thromb Vasc Biol, 2000. 20(8): p. 1903-11.

22. Chamseddine, A.H. and F.J. Miller, Jr., Gp91phox contributes to NADPH oxidase

activity in aortic fibroblasts but not smooth muscle cells. American journal of

physiology. Heart and circulatory physiology, 2003. 285(6): p. H2284-9.

Page 176: The stereoselective pharmacodynamics of the enantiomers of

176

23. Ushio-Fukai, M., et al., p22phox is a critical component of the superoxide-generating

NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in

vascular smooth muscle cells. J Biol Chem, 1996. 271(38): p. 23317-21.

24. Jawad, E. and R. Arora, Chronic stable angina pectoris. Disease-a-month : DM, 2008.

54(9): p. 671-89.

25. Murabito, J.M., et al., Prognosis after the onset of coronary heart disease. An

investigation of differences in outcome between the sexes according to initial coronary

disease presentation. Circulation, 1993. 88(6): p. 2548-55.

26. Kusama, Y., et al., Variant angina and coronary artery spasm: the clinical spectrum,

pathophysiology, and management. Journal of Nihon Medical School = Nihon Ika

Daigaku zasshi, 2011. 78(1): p. 4-12.

27. Prinzmetal, M., et al., Angina pectoris. I. A variant form of angina pectoris;

preliminary report. Am J Med, 1959. 27: p. 375-88.

28. Tunstall-Pedoe, H., et al., Contribution of trends in survival and coronary-event rates

to changes in coronary heart disease mortality: 10-year results from 37 WHO

MONICA project populations. Monitoring trends and determinants in cardiovascular

disease. Lancet, 1999. 353(9164): p. 1547-57.

29. Opie, L.H., The heart: physiology, from cell to circulation1998, Philadelphia, USA:

Lippincott-Raven.

Page 177: The stereoselective pharmacodynamics of the enantiomers of

177

30. Lee, L., J. Horowitz, and M. Frenneaux, Metabolic manipulation in ischaemic heart

disease, a novel approach to treatment. Eur Heart J, 2004. 25(8): p. 634-41.

31. Liedtke, A.J., Alterations of carbohydrate and lipid metabolism in the acutely

ischemic heart. Prog Cardiovasc Dis, 1981. 23(5): p. 321-36.

32. Calvani, M., E. Reda, and E. Arrigoni-Martelli, Regulation by carnitine of myocardial

fatty acid and carbohydrate metabolism under normal and pathological conditions.

Basic Res Cardiol, 2000. 95(2): p. 75-83.

33. van der Vusse, G.J., M. van Bilsen, and J.F. Glatz, Cardiac fatty acid uptake and

transport in health and disease. Cardiovasc Res, 2000. 45(2): p. 279-93.

34. McGarry, J.D., et al., Observations on the affinity for carnitine, and malonyl-CoA

sensitivity, of carnitine palmitoyltransferase I in animal and human tissues.

Demonstration of the presence of malonyl-CoA in non-hepatic tissues of the rat.

Biochem J, 1983. 214(1): p. 21-8.

35. Stanley, W.C., Myocardial energy metabolism during ischemia and the mechanisms of

metabolic therapies. Journal of cardiovascular pharmacology and therapeutics, 2004. 9

Suppl 1: p. S31-45.

36. Kolodziej, M.P. and V.A. Zammit, Mature carnitine palmitoyltransferase I retains the

N-terminus of the nascent protein in rat liver. FEBS letters, 1993. 327(3): p. 294-6.

Page 178: The stereoselective pharmacodynamics of the enantiomers of

178

37. Brown, N.F., et al., Expression of a cDNA for rat liver carnitine palmitoyltransferase I

in yeast establishes that catalytic activity and malonyl-CoA sensitivity reside in a

single polypeptide. J Biol Chem, 1994. 269(42): p. 26438-42.

38. McGarry, J.D. and N.F. Brown, The mitochondrial carnitine palmitoyltransferase

system. From concept to molecular analysis. European journal of biochemistry /

FEBS, 1997. 244(1): p. 1-14.

39. Rufer, A.C., R. Thoma, and M. Hennig, Structural insight into function and regulation

of carnitine palmitoyltransferase. Cell Mol Life Sci, 2009. 66(15): p. 2489-501.

40. Brown, N.F., et al., Mitochondrial carnitine palmitoyltransferase I isoform switching

in the developing rat heart. J Biol Chem, 1995. 270(15): p. 8952-7.

41. Saddik, M., et al., Acetyl-CoA carboxylase regulation of fatty acid oxidation in the

heart. J Biol Chem, 1993. 268(34): p. 25836-45.

42. Fraser, F., C.G. Corstorphine, and V.A. Zammit, Evidence that both the acyl-CoA-

and malonyl-CoA binding sites of mitochondrial overt carnitine palmitoyltransferase

(CPT I) are exposed on the cytosolic face of the outer membrane. Biochem Soc Trans,

1996. 24(2): p. 184S.

43. Kashfi, K. and G.A. Cook, Proteinase treatment of intact hepatic mitochondria has

differential effects on inhibition of carnitine palmitoyltransferase by different

inhibitors. The Biochemical journal, 1992. 282 ( Pt 3): p. 909-14.

44. Schulz, H., Regulation of fatty acid oxidation in heart. J Nutr, 1994. 124(2): p. 165-71.

Page 179: The stereoselective pharmacodynamics of the enantiomers of

179

45. Bell, G.I., et al., Molecular biology of mammalian glucose transporters. Diabetes

Care, 1990. 13(3): p. 198-208.

46. Luiken, J.J., et al., Regulation of cardiac long-chain fatty acid and glucose uptake by

translocation of substrate transporters. Pflugers Arch, 2004. 448(1): p. 1-15.

47. Kraegen, E.W., et al., Glucose transporters and in vivo glucose uptake in skeletal and

cardiac muscle: fasting, insulin stimulation and immunoisolation studies of GLUT1

and GLUT4. Biochem J, 1993. 295 ( Pt 1): p. 287-93.

48. Stanley, W.C., F.A. Recchia, and G.D. Lopaschuk, Myocardial substrate metabolism

in the normal and failing heart. Physiol Rev, 2005. 85(3): p. 1093-129.

49. Depre, C., J.L. Vanoverschelde, and H. Taegtmeyer, Glucose for the heart.

Circulation, 1999. 99(4): p. 578-88.

50. Stanley, W.C., et al., Regulation of myocardial carbohydrate metabolism under

normal and ischaemic conditions. Potential for pharmacological interventions.

Cardiovasc Res, 1997. 33(2): p. 243-57.

51. Patel, M.S. and L.G. Korotchkina, Regulation of the pyruvate dehydrogenase complex.

Biochem Soc Trans, 2006. 34(Pt 2): p. 217-22.

52. McCormack, J.G. and R.M. Denton, Influence of calcium ions on mammalian

intramitochondrial dehydrogenases. Methods Enzymol, 1989. 174: p. 95-118.

Page 180: The stereoselective pharmacodynamics of the enantiomers of

180

53. Stanley, W.C., Cardiac energetics during ischaemia and the rationale for metabolic

interventions. Coron Artery Dis, 2001. 12 Suppl 1: p. S3-7.

54. Turer, A.T., et al., Metabolomic profiling reveals distinct patterns of myocardial

substrate use in humans with coronary artery disease or left ventricular dysfunction

during surgical ischemia/reperfusion. Circulation, 2009. 119(13): p. 1736-46.

55. Hansford, R.G. and L. Cohen, Relative importance of pyruvate dehydrogenase

interconversion and feed-back inhibition in the effect of fatty acids on pyruvate

oxidation by rat heart mitochondria. Arch Biochem Biophys, 1978. 191(1): p. 65-81.

56. Kerbey, A.L., et al., Regulation of pyruvate dehydrogenase in rat heart. Mechanism of

regulation of proportions of dephosphorylated and phosphorylated enzyme by

oxidation of fatty acids and ketone bodies and of effects of diabetes: role of coenzyme

A, acetyl-coenzyme A and reduced and oxidized nicotinamide-adenine dinucleotide.

Biochem J, 1976. 154(2): p. 327-48.

57. Chandler, M.P., et al., Increased nonoxidative glycolysis despite continued fatty acid

uptake during demand-induced myocardial ischemia. Am J Physiol Heart Circ

Physiol, 2002. 282(5): p. H1871-8.

58. Oliver, M.F. and L.H. Opie, Effects of glucose and fatty acids on myocardial

ischaemia and arrhythmias. Lancet, 1994. 343(8890): p. 155-8.

59. Kantor, P.F., et al., The antianginal drug trimetazidine shifts cardiac energy

metabolism from fatty acid oxidation to glucose oxidation by inhibiting mitochondrial

Page 181: The stereoselective pharmacodynamics of the enantiomers of

181

long-chain 3-ketoacyl coenzyme A thiolase. Circulation research, 2000. 86(5): p. 580-

8.

60. Ratheiser, K., et al., Inhibition by etomoxir of carnitine palmitoyltransferase I reduces

hepatic glucose production and plasma lipids in non-insulin-dependent diabetes

mellitus. Metabolism: clinical and experimental, 1991. 40(11): p. 1185-90.

61. Holubarsch, C.J., et al., A double-blind randomized multicentre clinical trial to

evaluate the efficacy and safety of two doses of etomoxir in comparison with placebo

in patients with moderate congestive heart failure: the ERGO (etomoxir for the

recovery of glucose oxidation) study. Clinical science, 2007. 113(4): p. 205-12.

62. Chaitman, B.R., Efficacy and safety of a metabolic modulator drug in chronic stable

angina: review of evidence from clinical trials. Journal of cardiovascular

pharmacology and therapeutics, 2004. 9 Suppl 1: p. S47-64.

63. Chaitman, B.R., et al., Anti-ischemic effects and long-term survival during ranolazine

monotherapy in patients with chronic severe angina. Journal of the American College

of Cardiology, 2004. 43(8): p. 1375-82.

64. Babior, B.M., J.D. Lambeth, and W. Nauseef, The neutrophil NADPH oxidase. Arch

Biochem Biophys, 2002. 397(2): p. 342-4.

65. Cave, A.C., et al., NADPH oxidases in cardiovascular health and disease. Antioxid

Redox Signal, 2006. 8(5-6): p. 691-728.

Page 182: The stereoselective pharmacodynamics of the enantiomers of

182

66. Hudsmith, L.E. and S. Neubauer, Magnetic resonance spectroscopy in myocardial

disease. JACC. Cardiovascular imaging, 2009. 2(1): p. 87-96.

67. Lee, L., et al., Metabolic modulation with perhexiline in chronic heart failure: a

randomized, controlled trial of short-term use of a novel treatment. Circulation, 2005.

112(21): p. 3280-8.

68. Barry, W.H., J.D. Horowitz, and T.W. Smith, Comparison of negative inotropic

potency, reversibility, and effects on calcium influx of six calcium channel antagonists

in cultured myocardial cells. Br J Pharmacol, 1985. 85(1): p. 51-9.

69. Kennedy, J.A., S.A. Unger, and J.D. Horowitz, Inhibition of carnitine

palmitoyltransferase-1 in rat heart and liver by perhexiline and amiodarone. Biochem

Pharmacol, 1996. 52(2): p. 273-80.

70. Kennedy, J.A., et al., Effect of perhexiline and oxfenicine on myocardial function and

metabolism during low-flow ischemia/reperfusion in the isolated rat heart. J

Cardiovasc Pharmacol, 2000. 36(6): p. 794-801.

71. Lorentz, I.T. and M. Shortall, Perhexilene neuropathy: a report of two cases.

Australian and New Zealand journal of medicine, 1983. 13(5): p. 517-8.

72. Wijesekera, J.C., et al., Peripheral neuropathy due to perhexilene maleate. J Neurol

Sci, 1980. 46(3): p. 303-9.

73. Morgan, M.Y., et al., Impaired oxidation of debrisoquine in patients with perhexiline

liver injury. Gut, 1984. 25(10): p. 1057-64.

Page 183: The stereoselective pharmacodynamics of the enantiomers of

183

74. Singlas, E., M.A. Goujet, and P. Simon, Pharmacokinetics of perhexiline maleate in

anginal patients with and without peripheral neuropathy. Eur J Clin Pharmacol, 1978.

14(3): p. 195-201.

75. Deschamps, D., et al., Inhibition by perhexiline of oxidative phosphorylation and the

beta-oxidation of fatty acids: possible role in pseudoalcoholic liver lesions.

Hepatology, 1994. 19(4): p. 948-61.

76. Meier, C., et al., Perhexiline-induced lipidosis in the dark Agouti (DA) rat. An animal

model of genetically determined neurotoxicity. Brain, 1986. 109 ( Pt 4): p. 649-60.

77. Wright, G.J., et al., Proceedings: The absorption, excretion and metabolism of

perhexiline maleate by the human. Postgraduate medical journal, 1973. 49: p. Suppl

3:8-15.

78. Hussein, R., et al., Population pharmacokinetics of perhexiline from very sparse,

routine monitoring data. Therapeutic drug monitoring, 2001. 23(6): p. 636-43.

79. Davies, B.J., et al., CYP2B6, CYP2D6, and CYP3A4 catalyze the primary oxidative

metabolism of perhexiline enantiomers by human liver microsomes. Drug Metab

Dispos, 2007. 35(1): p. 128-38.

80. Inglis, S.C., et al., Effect of CYP2D6 metabolizer status on the disposition of the (+)

and (-) enantiomers of perhexiline in patients with myocardial ischaemia.

Pharmacogenetics and genomics, 2007. 17(5): p. 305-12.

Page 184: The stereoselective pharmacodynamics of the enantiomers of

184

81. Cooper, R.G., D.A. Evans, and E.J. Whibley, Polymorphic hydroxylation of

perhexiline maleate in man. J Med Genet, 1984. 21(1): p. 27-33.

82. Davies, B.J., et al., Determination of the 4-monohydroxy metabolites of perhexiline in

human plasma, urine and liver microsomes by liquid chromatography. J Chromatogr

B Analyt Technol Biomed Life Sci, 2006. 843(2): p. 302-9.

83. Shah, R.R., et al., Impaired oxidation of debrisoquine in patients with perhexiline

neuropathy. Br Med J (Clin Res Ed), 1982. 284(6312): p. 295-9.

84. Nebert, D.W. and D.W. Russell, Clinical importance of the cytochromes P450.

Lancet, 2002. 360(9340): p. 1155-62.

85. Wilkinson, G.R., Drug metabolism and variability among patients in drug response. N

Engl J Med, 2005. 352(21): p. 2211-21.

86. Zhou, S.F., Polymorphism of human cytochrome P450 2D6 and its clinical

significance: Part I. Clinical pharmacokinetics, 2009. 48(11): p. 689-723.

87. Gough, A.C., et al., Localization of the CYP2D gene locus to human chromosome

22q13.1 by polymerase chain reaction, in situ hybridization, and linkage analysis.

Genomics, 1993. 15(2): p. 430-2.

88. Ingelman-Sundberg, M., Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6):

clinical consequences, evolutionary aspects and functional diversity. The

pharmacogenomics journal, 2005. 5(1): p. 6-13.

Page 185: The stereoselective pharmacodynamics of the enantiomers of

185

89. Zanger, U.M., S. Raimundo, and M. Eichelbaum, Cytochrome P450 2D6: overview

and update on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs Arch

Pharmacol, 2004. 369(1): p. 23-37.

90. Kagimoto, M., et al., Multiple mutations of the human cytochrome P450IID6 gene

(CYP2D6) in poor metabolizers of debrisoquine. Study of the functional significance

of individual mutations by expression of chimeric genes. J Biol Chem, 1990. 265(28):

p. 17209-14.

91. Haefeli, W.E., et al., Potent inhibition of cytochrome P450IID6 (debrisoquin 4-

hydroxylase) by flecainide in vitro and in vivo. Journal of cardiovascular

pharmacology, 1990. 15(5): p. 776-9.

92. Vandamme, N., et al., Stereoselective hydroxylation of mexiletine in human liver

microsomes: implication of P450IID6--a preliminary report. Journal of cardiovascular

pharmacology, 1993. 21(1): p. 77-83.

93. Sakuyama, K., et al., Functional characterization of 17 CYP2D6 allelic variants

(CYP2D6.2, 10, 14A-B, 18, 27, 36, 39, 47-51, 53-55, and 57). Drug metabolism and

disposition: the biological fate of chemicals, 2008. 36(12): p. 2460-7.

94. Johansson, I., et al., Inherited amplification of an active gene in the cytochrome P450

CYP2D locus as a cause of ultrarapid metabolism of debrisoquine. Proceedings of the

National Academy of Sciences of the United States of America, 1993. 90(24): p.

11825-9.

Page 186: The stereoselective pharmacodynamics of the enantiomers of

186

95. Horowitz, J.D., et al., High-performance liquid chromatographic assay of perhexiline

maleate in plasma. Journal of pharmaceutical sciences, 1981. 70(3): p. 320-2.

96. Al-Dabbagh, S.G., J.R. Idle, and R.L. Smith, Animal modelling of human polymorphic

drug oxidation--the metabolism of debrisoquine and phenacetin in rat inbred strains.

The Journal of pharmacy and pharmacology, 1981. 33(3): p. 161-4.

97. Kahn, G.C., et al., Sex and strain differences in hepatic debrisoquine 4-hydroxylase

activity of the rat. Drug metabolism and disposition: the biological fate of chemicals,

1985. 13(4): p. 510-6.

98. Gonzalez, F.J., et al., Debrisoquine 4-hydroxylase: characterization of a new P450

gene subfamily, regulation, chromosomal mapping, and molecular analysis of the DA

rat polymorphism. DNA, 1987. 6(2): p. 149-61.

99. Matsunaga, E., et al., The CYP2D gene subfamily: analysis of the molecular basis of

the debrisoquine 4-hydroxylase deficiency in DA rats. Biochemistry, 1989. 28(18): p.

7349-55.

100. Yamamoto, Y., et al., Molecular basis of the Dark Agouti rat drug oxidation

polymorphism: importance of CYP2D1 and CYP2D2. Pharmacogenetics, 1998. 8(1):

p. 73-82.

101. Schulz-Utermoehl, T., et al., Polymorphic debrisoquine 4-hydroxylase activity in the

rat is due to differences in CYP2D2 expression. Pharmacogenetics, 1999. 9(3): p. 357-

66.

Page 187: The stereoselective pharmacodynamics of the enantiomers of

187

102. McGinnity, D.F., et al., Automated definition of the enzymology of drug oxidation by

the major human drug metabolizing cytochrome P450s. Drug metabolism and

disposition: the biological fate of chemicals, 2000. 28(11): p. 1327-34.

103. Midha, K.K., et al., Pharmacokinetics of glucuronidation of propranolol following

oral administration in humans. Biopharmaceutics & drug disposition, 1983. 4(4): p.

331-8.

104. Komura, H. and M. Iwaki, Pharmacokinetics and metabolism of metoprolol and

propranolol in the female DA and female Wistar rat: the female DA rat is not always

an animal model for poor metabolizers of CYP2D6. J Pharm Sci, 2005. 94(2): p. 397-

408.

105. Cleary, J., et al., The influence of pharmacogenetics on opioid analgesia: studies with

codeine and oxycodone in the Sprague-Dawley/Dark Agouti rat model. The Journal of

pharmacology and experimental therapeutics, 1994. 271(3): p. 1528-34.

106. Smith, S.W., Chiral toxicology: it's the same thing...only different. Toxicological

sciences : an official journal of the Society of Toxicology, 2009. 110(1): p. 4-30.

107. Gal, J., The discovery of biological enantioselectivity: Louis Pasteur and the

fermentation of tartaric acid, 1857--a review and analysis 150 yr later. Chirality,

2008. 20(1): p. 5-19.

108. Brocks, D.R., Drug disposition in three dimensions: an update on stereoselectivity in

pharmacokinetics. Biopharm Drug Dispos, 2006. 27(8): p. 387-406.

Page 188: The stereoselective pharmacodynamics of the enantiomers of

188

109. Himmelseher, S. and E. Pfenninger, [The clinical use of S-(+)-ketamine--a

determination of its place]. Anasthesiol Intensivmed Notfallmed Schmerzther, 1998.

33(12): p. 764-70.

110. Patil, P.A. and M.A. Kothekar, Development of safer molecules through chirality.

Indian J Med Sci, 2006. 60(10): p. 427-37.

111. Gould, B.J., A.G. Amoah, and D.V. Parke, Stereoselective pharmacokinetics of

perhexiline. Xenobiotica; the fate of foreign compounds in biological systems, 1986.

16(5): p. 491-502.

112. Davies, B.J., et al., Enantioselective assay for the determination of perhexiline

enantiomers in human plasma by liquid chromatography. Journal of chromatography.

B, Analytical technologies in the biomedical and life sciences, 2006. 832(1): p. 114-

20.

113. Unger, S.A., et al., Dissociation between metabolic and efficiency effects of

perhexiline in normoxic rat myocardium. J Cardiovasc Pharmacol, 2005. 46(6): p.

849-55.

114. Fenster, B.E., P.S. Tsao, and S.G. Rockson, Endothelial dysfunction: clinical

strategies for treating oxidant stress. Am Heart J, 2003. 146(2): p. 218-26.

115. Lageron, A., J. Scotto, and M. Gautier, Effects of Pexid on liver cell cultures.

Ultrastructural and histoenzymological studies. European journal of clinical

pharmacology, 1981. 19(6): p. 417-21.

Page 189: The stereoselective pharmacodynamics of the enantiomers of

189