178
New insight into Acyclovir Renal Handling and Nephrotoxicity by Patrina Francia Gunness A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Pharmaceutical Sciences University of Toronto © Copyright by Patrina Francia Gunness 2011

New insight into Acyclovir Renal Handling and Nephrotoxicity · proximal tubular cells and the toxicity may be caused by the parent drug’s noxious acyclovir aldehyde ... containing

  • Upload
    ngodan

  • View
    214

  • Download
    0

Embed Size (px)

Citation preview

New insight into Acyclovir Renal Handling

and Nephrotoxicity

by

Patrina Francia Gunness

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Graduate Department of Pharmaceutical Sciences

University of Toronto

© Copyright by Patrina Francia Gunness 2011

ii

New insight into Acyclovir Renal Handling and Nephrotoxicity

Patrina Francia Gunness

Doctor of Philosophy

Graduate Department of Pharmaceutical Sciences

University of Toronto

2011

Abstract

Drug – induced nephrotoxicity is a serious adverse reaction that can have deleterious effects on

a patient’s health and well-being. Acyclovir is an example of such an agent that causes the

aforesaid effects. The drug induces severe nephrotoxicity in patients. The etiology of acyclovir

– induced nephrotoxicity has not been fully elucidated. The overall objective of this thesis is to

gain new insight into the pathogenesis of acyclovir – induced nephrotoxicity.

Cytotoxicity studies showed that acyclovir induced human renal proximal tubular (HK-2) cell

death, in vitro, and that the degree of this toxicity was significantly reduced by co-exposure to 4-

methylpyrazole. The results suggest that acyclovir induces direct insult to human renal

proximal tubular cells and the toxicity may be caused by the parent drug’s noxious acyclovir

aldehyde metabolite.

Transepithelial transport studies illustrated that acyclovir does not inhibit the transport of

creatinine across porcine renal proximal tubular (LLC-PK1) or HK-2 cell monolayers. The

results suggest that acyclovir does not inhibit the tubular secretion of creatinine in vitro, and

possibly, in vivo, as well. Therefore, the abrupt, pronounced and transient elevations in the

levels of plasma creatinine observed in patients may be solely and genuinely due to reduced

iii

GFR as a result of acyclovir – induced nephrotoxicity, and not to a tubular interaction between

creatinine and acyclovir.

Employing human embryonic kidney cells (HEK293) containing the full-length human ABCG2

gene encoding the wildtype ABCG2 amino acid sequence; cell accumulation studies showed

that in the presence of the human breast cancer resistance protein (BCRP) inhibitor,

fumitremorgin C (FTC), there was significant intracellular accumulation of acyclovir. The

results suggest that acyclovir is a substrate for the efflux transporter and bears several potential

implications with respect to the renal transport mechanisms and pathogenesis of the direct

tubular damage induced by the drug.

Synthesizing all the data, the results contribute to a better understanding of the pathogenesis of

acyclovir – induced nephrotoxicity. Moreover, the research highlights the need for future

studies that will aid in further elucidation of the underlying cell and molecular mechanism(s) of

this toxicity and potential therapies for prevention of the direct renal tubular injury induced by

the drug.

iv

Acknowledgements

I would like to express my sincerest gratitude to several individuals who have provided their

endless support and guidance to me during my Ph.D. studies.

First, I would like to express my deepest gratitude to my supervisor, Dr. Gideon Koren. I would

like to thank you always taking the time to share your knowledge with me and for providing me

with support, advice, guidance and patience during my graduate studies and the preparation of

this thesis. I consider myself very fortunate to have had you as my Ph.D. mentor. You have

instilled in me, invaluable lessons in academia and scientific research, which I will continue to

use as I move forward in my career. Thank you.

I would also like to thank Dr. Katarina Aleksa for her continuous support, guidance and

encouragement. Thank you for always taking the time to offer your knowledge and advice. It

was tremendously appreciated.

Finally, I would like to thank my advisory committee members, Drs. Shinya Ito, Cecil Pace-

Asciak and Anna Taddio for their advice and guidance throughout my studies. Thank you.

v

Table of Contents

Table of contents.................................................................................................................v

List of Publications..............................................................................................................xiii

List of Abbreviations...........................................................................................................xiv

List of Tables.......................................................................................................................xix

List of Figures......................................................................................................................xx

Chapter 1: General Introduction......................................................................................1

1.1 Acyclovir – induced nephrotoxicity in children: new insight into

its mechanism of toxicity, interaction with creatinine and tubular

transport........................................................................................................1

1.2 Acyclovir......................................................................................................1

1.2.1 Acyclovir use...................................................................................1

1.2.2 Acyclovir: mechanism of action......................................................2

1.2.3 Acyclovir metabolism and excretion................................................3

1.2.4 Acyclovir – induced nephrotoxicity.................................................5

1.2.5 Mechanism(s) of acyclovir – induced nephrotoxicity......................5

1.2.5a Acyclovir – induced crystalluria...........................................5

1.2.5b Acyclovir – induced direct renal tubular cell injury.............6

1.2.6 Acyclovir aldehyde: its potential role in direct renal tubular

Injury.................................................................................................6

1.3 Acyclovir and creatinine: interaction during tubular secretion?...................7

1.3.1 Creatinine..........................................................................................7

1.3.2 Renal tubular secretion of creatinine: opportunity for

interaction with other drugs and subsequent consequences............7

1.4 Role of the human breast cancer resistance protein (BCRP) in

vi

the transport of acyclovir: potential implications in tubular

transport and nephrotoxicity...................................................................9

1.4.1 BCRP...........................................................................................9

1.4.2 Acyclovir as a potential substrate of human BCRP:

renal tubular transport and toxicological significance.................10

1.5 References................................................................................................11

Chapter 2: Hypotheses and Objective..........................................................................16

2.1 Hypotheses...............................................................................................16

2.2 Objectives.................................................................................................17

Chapter 3: Comparison of the novel HK-2 human renal proximal tubular

cell line with the standard LLC-PK1 cell line in studying

drug-induced nephrotoxicity.......................................................................18

3.1 Abstract......................................................................................................19

3.2 Introduction...............................................................................................19

3.3 Materials and methods...............................................................................23

3.3.1 Chemicals.......................................................................................23

3.3.2 HK-2 cells......................................................................................24

3.3.2a Culturing conditions of HK-2 cells for ifosfamide

Experiments........................................................................24

3.3.2b Culturing conditions of HK-2 cells for acyclovir

Experiments........................................................................25

3.3.3 LLC-PK1 cells................................................................................25

3.3.3a Culturing conditions of LLC-PK1 cells for

ifosfamide experiments.......................................................25

vii

3.3.3b Culturing conditions of LLC-PK1 cells for

acyclovir experiments......................................................25

3.3.4 Experimental methods used to determine whether HK-2

cells are an appropriate model to study ifosfamide

– induced nephrotoxicity..............................................................26

3.3.4a Determination of CYP enzyme mRNA expression

in HK-2 cells by RT-PCR................................................26

3.3.4b Determination of CYP enzyme protein expression

in HK-2 cells by Western blotting....................................27

3.3.4c Determination of renal proximal tubule metabolism

of ifosfamide in HK-2 cells by LC-MS.............................27

3.3.4d Determination of GSH levels in HK-2 and LLC-PK

Cells..................................................................................29

3.3.5 Experimental method used to determine whether HK-2

cells are an appropriate model to study acyclovir –

induced nephrotoxicity..................................................................31

3.3.5a Determination of cytotoxicity in HK-2 and

LLC-PK1 cells...................................................................31

3.3.6 Statistical analyses.........................................................................31

3.4 Results.......................................................................................................32

3.4.1 CYP mRNA and protein expression in HK-2 cells........................32

3.4.2 Renal proximal tubular metabolism of ifsofamide by HK-2

and LLC-PK1 cells.........................................................................34

viii

3.4.3 Depletion of GSH and GSSG in HK-2 and LLC-PK1

cells...........................................................................................34

3.4.4 Acyclovir – induced cytotoxicity in LLC-PK1 and

HK-2 cells.................................................................................36

3.5 Discussion.............................................................................................37

3.6 Acknowledgements..............................................................................40

3.7 Statement of significance......................................................................40

3.8 References.............................................................................................41

Chapter 4: Acyclovir – induced nephrotoxicity; the role of the acyclovir

aldehyde metabolite.................................................................................44

4.1 Abstract................................................................................................45

4.2 Introduction..........................................................................................45

4.3 Materials and methods..........................................................................48

4.3.1 Cell culture................................................................................48

4.3.2 Protein expression and enzymes activities of class I ADH

and ALDH2 isozymes in HK-2 cells.........................................49

4.3.2a Cytosol and mitochondria protein fraction for

western blot assays.........................................................50

4.3.3 Western blot assays....................................................................51

4.3.3a ADH protein expression.................................................51

4.3.3b ALDH2 protein expression.............................................52

4.3.4 Enzymes activities assays............................................................52

4.3.4a Whole cell lysate for enzymes activities assays..............52

4.3.4b ADH and ALDH enzymes activities assays....................53

4.3.5 Cell viability................................................................................54

ix

4.3.5a Co-exposure to 4-methylpyrazole...................................55

4.3.6 Determination of aldehyde production.......................................55

4.3.7 Comparison of the ADH protein expression between

HK-2 cells and human kidney tissue..........................................56

4.3.8 Statistical analyses......................................................................57

4.4 Results.....................................................................................................57

4.4.1 Class I ADH and ALDH2 protein expression.............................57

4.4.2 ADH and ALDH enzyme activity...............................................60

4.4.3 The effect of 4-methylpyrazole on HK-2 cell viability...............63

4.4.4 Aldehyde production in HK-2 cells exposed to acyclovir..........65

4.4.5 Comparison of the ADH protein expression level between

HK-2 cells and human kidney....................................................66

4.5 Discussion................................................................................................67

4.6 Statement of significance.........................................................................73

4.7 Acknowledgements.................................................................................73

4.8 References...............................................................................................74

4.9 Additional experiments not published.....................................................79

4.9.1 The effect of CMMG on cell viability.........................................79

4.9.2 Materials and methods.................................................................79

4.9.2a Exposure to CMMG.........................................................79

4.9.2b Statistical analyses............................................................79

4.9.3 Results......................................................................................... 79

Chapter 5: The effect of acyclovir on the tubular secretion of creatinine

in vitro.........................................................................................................81

5.0 Abstract...................................................................................................82

5.1 Introduction.............................................................................................82

x

5.2 Materials and methods...............................................................................87

5.2.1 Cell culture.....................................................................................87

5.2.2 Transepithelial transport studies....................................................87

5.2.2a Tetraethylammonium (TEA) transport across cell

monolayers.........................................................................89

5.2.2b Acyclovir transport across cell monolayers.......................89

5.2.2c The effect of acyclovir on creatinine transport

across cell monolayers........................................................89

5.2.3 Statistical analyses..........................................................................90

5.3 Results........................................................................................................90

5.3.1 TEA transport across LLC-PK1 and HK-2 cell

monolayers......................................................................................90

5.3.2 Acyclovir transport across LLC-PK1 and HK-2 cell

monolayers.......................................................................................93

5.3.3 The effect of acyclovir on creatinine transport across

LLC-PK1and HK-2 cell monolayers..............................................96

5.4 Discussion....................................................................................................99

5.5 Acknowledgements....................................................................................103

5.6 Statement of significance............................................................................103

5.7 References..................................................................................................104

5.8 Additional experiments not published........................................................108

5.8.1 The paracellular flux (basolateral-to-apical) of

D-[1-3H(N)] mannitol......................................................................108

5.8.2 Materials and methods.....................................................................108

5.8.3 Results.............................................................................................108

Chapter 6: Acyclovir is a substrate for the human breast cancer resistance

protein (BCRP/ABCG2): implications for renal tubular transport

xi

and acyclovir – induced nephrotoxicity..................................................115

6.1 Abstract..................................................................................................116

6.2 Introduction............................................................................................116

6.3 Materials and methods...........................................................................118

6.3.1 Cell culture.................................................................................118

6.3.2 Determination of protein expression of human BCRP

in overexpressing HEK293 cells................................................119

6.3.3 Whole cell lysate for western blot assays...................................119

6.3.3a Mock or overexpressing HEK293 cells..........................119

6.3.3b Human placenta tissue....................................................120

6.3.4 Western blot assay......................................................................120

6.3.5 Hoescht 33342 dye efflux assay.................................................121

6.3.6 Cell accumulation assay.............................................................121

6.3.7 Statistical analyses......................................................................122

6.4 Results....................................................................................................123

6.4.1 The protein expression of human BCRP in

overexpressing HEK293 cells.....................................................123

6.4.2 The functionality of BCRP in overexpressing HEK293

cells..............................................................................................124

6.4.3 Intracellular accumulation of [8-14

C] acyclovir...........................125

6.5 Discussion................................................................................................126

6.6 Statement of significance.........................................................................128

6.7 References...............................................................................................129

xii

6.8 Additional experiments not published......................................................133

6.8.1 The effect of acyclovir on HEK293 cell viability.........................133

6.8.2 Materials and methods..................................................................133

6.8.2a Cytotoxicity assay.............................................................133

6.8.2a Statistical analyses.............................................................133

6.8.3 Results...........................................................................................134

Chapter 7: Summary of research findings....................................................................135

7.1 Summary of research findings and their significance................................135

7.1.1 To investigate whether acyclovir – induced nephrotoxicity

is due to, in part, direct insult to renal tubular cells........................135

7.1.2 To determine whether acyclovir aldehyde plays a role in

the direct renal tubular injury induced by acyclovir.......................136

7.1.3 To determine whether acyclovir inhibits the renal tubular

secretion of creatinine.....................................................................136

7.1.4 To determine whether acyclovir is a substrate for human

BCRP..............................................................................................138

7.2 References...................................................................................................139

Chapter 8: General Discussion and Conclusions............................................................141

8.1 Acyclovir and direct renal tubular injury.....................................................141

8.2 Acyclovir-creatinine tubular interaction......................................................145

8.3 Renal tubular transport of acyclovir.............................................................146

8.4 Limitations and future directions.................................................................147

8.5 Conclusions..................................................................................................150

8.6 References....................................................................................................152

xiii

List of Publications

Gunness, P., Aleksa, K., Kosuge, K., Ito, S., and Koren, G. 2010. Comparison of the novel HK-

2 human renal proximal tubular cell line with the standard LLC-PK1 cell line in studying drug-

induced nephrotoxicity. Can J Physiol Pharmacol 88: 448-455. This article was originally

published by NRC Research Press.

Gunness, P., Aleksa, K., and Koren, G. 2010. The effect of acyclovir on the tubular secretion of

creatinine in vitro. J Transl Med 8: 139-149. This article was originally published by BioMed

Central.

Gunness, P., Aleksa, K., and Koren, G. 2011. Acyclovir is a substrate for the human breast

cancer resistance protein (BCRP/ABCG2): implications for renal tubular transport and acyclovir

– induced nephrotoxicity. Can J Physiol Pharmacol. [In press]. This article will be originally

published by NRC Research Press.

Gunness, P., Aleksa, K., Bend, J., and Koren, G. 2011. Acyclovir – induced nephrotoxicity: the

role of the acyclovir aldehyde metabolite. Transl Res. [In press]. This article will be originally

published by Elsevier.

xiv

List of Abbreviations

x g - times gravitational force

α-MEM - alpha modified minimum essential medium

◦C - degrees celsius

% - percent

µg – microgram

µL - microlitre

µm - micron

µg/mL - microgram per millilitre

ABCG - adenosine triphosphate (ATP) binding cassette transporter

ADH - alcohol dehydrogenase

ALDH - aldehyde dehydrogenase

ALDH2 - aldehyde dehydrogenase 2

ANOVA - analysis of variance

ATCC - American type culture collection

BCRP - human breast cancer resistance protein

Bcrp1 - murine breast cancer resistance protein

BSO - L-buthionine sulfoximine

CaCl2 - calcium chloride

cDNA - copy DNA

CE - collision energy

CIHR - Canadian Institutes of Health Research

cm2 - square centimetre

CMMG - 9-carboxymethoxymethylguanine

xv

CO2 - carbon dioxide

CYP - cytochrome P450 enzyme

DCEIF - dechloroethylifosfamide

DMEM/F12 - Dulbecco's modified Eagle's minimum essential medium/Ham's F-12

DMEM - Dulbecco's modified Eagle's minimum essential medium

DNA - deoxyribonucleic acid

DNTB - dithiobis-2-nitrobenzoic acid

dNTP - deoxyribonucleotide triphosphate

DP - declustering potential

DPM - disintegrations per minutes

ECL - enhanced chemiluminescence reagent

EDTA - ethylenediaminetetraacetic acid

EMEM - Eagles’s minimum essential medium

FBS - fetal bovine serum

FTC - fumitremorgin C

GFR - glomerular filtration rate

GR - glutathione reductase

GSH - glutathione

GSSG - glutathione disulfide

HEK293 - human embryonic kidney cell line

HEPES - 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

HK-2 - human renal proximal tubular cells

hOCT1 - human organic cation transporter 1

hOCT2 - human organic cation transporter 2

HPLC - high performance liquid chromatography

xvi

hr - hour

hrs - hours

HSD - honestly significant difference

IgG - HRP - immunoglobulin G – horseradish peroxidase

KCl - potassium chloride

kD – kilo dalton

kV - kilo volts

L - litre

LC-MS - Liquid chromatography – mass spectrometry

LC-MS/MS - liquid chromatography tandem mass spectrometry

LLC-PK1 - porcine renal proximal tubular cells

M2VP - 1-methyl-2-vinylpyridinium trifluoromethanesulfonate

MgCl2 - magnesium chloride

mg/kg - milligram per kilogram

mg/mL - milligram per millilitre

mg/L – milligram per litre

min - minute

mins - minutes

mL - millilitre

mL/min - millilitre per minute

mm - millimetre

mmol/L – millimole per litre

MRM - multiple reactions monitored

mRNA - messenger RNA

M.W. - molecular weight

xvii

m/z - mass-to-charge ratio

NAD+

- nicotinamide adenine dinucleotide

NADH - reduced form of nicotinamide adenine dinucleotide

NADPH - nicotinamide adenine dinucleotide phosphate-oxidase

NCBI - National centre for Biotechnology information

NIH - National Institutes of Health

nmol/L – nanomole per litre

ng - nanogram

ng/mL - nanogram per millilitre

nm - nanometre

NP- 40 - nonyl phenoxypolyethoxylethanol

NaCl - sodium chloride

O2 - oxygen

OAT - organic anion transporter

OCT - organic cation transporter

PBS - phosphate buffer saline

PBST - PBS with Tween® 20 detergent

PMSF - phenylmethylsulfonyl fluoride

pOCT1 - porcine organic cation transporter 1

pOCT2 - porcine organic cation transporter 2

PVDF - polyvinylidene difluoride

RNA - ribonucleic acid

RT-PCR - reverse transcription polymerase chain reaction

SDS-PAGE - sodium dodecyl sulfate – polyacrylamide gel electrophoresis

SD – standard deviation

xviii

SE - standard error

secs - seconds

SFM - serum free medium

TEA - tetraethylammonium

Tris-HCL - tris(hydroxymethyl)aminomethane hydrochloride

USA - United States of America

v/v - volume per volume

w/v - weight per volume

xix

List of Tables

Table 1. Summary of some important pharmacokinetic parameters

of acyclovir in children [1 – 17 years]..........................................................2

Table 2. HK-2 versus LLC-PK1 as a model for ifosfamide – induced

nephrotoxicity................................................................................................36

Table 3. Cases of elevated plasma creatinine levels in children who

received intravenous acyclovir......................................................................86

xx

List of Figures

Figure 1. Structural formula of acyclovir..............................................................1

Figure 2. Schematic diagram illustrating the mechanism of action

of acyclovir..............................................................................................3

Figure 3. Metabolism of acyclovir in humans........................................................4

Figure 4. Total Ribonucleic acid (RNA) was isolated from human

renal proximal tubular (HK-2) cells

and reverse transcribed...........................................................................32

Figure 5. Western blot of human renal proximal tubular (HK-2)

cells for cytochrome P450 (CYP) enzymes............................................33

Figure 6. Metabolism of ifosfamide by human renal proximal tubular

(HK-2) cells............................................................................................34

Figure 7. Glutathione (GSH) depletion in human (HK-2) and porcine

(LLC-PK1) renal proximal tubular cells.................................................35

Figure 8. Acyclovir – induced cytotoxicity in human (HK-2) and

porcine (LLC-PK1) renal proximal tubular cells...................................37

Figure 9A. The alcohol dehydrogenase (ADH) protein expression in

Human renal proximal tubular (HK-2) cell.............................................58

xxi

Figure 9B. The aldehyde dehydrogenase (ALDH) protein expression

in human renal proximal tubular (HK-2) cells.....................................58

Figure 10A. The alcohol dehydrogenase (ADH) protein expression in

human kidney........................................................................................59

Figure 10B. The aldehyde dehydrogenase (ALDH) protein expression in

human kidney........................................................................................59

Figure 11A. The alcohol dehydrogenase (ADH) enzyme activity in

human renal proximal tubular (HK-2) cells...........................................61

Figure 11B. The aldehyde dehydrogenase enzyme activity in human

renal proximal tubular (HK-2) cells.......................................................61

Figure 12A. The alcohol dehydrogenase (ADH) enzyme activity in

human kidney.........................................................................................62

Figure 12B. The aldehyde dehydrogenase enzyme activity in human

kidney.....................................................................................................62

Figure 13. The effect of 4-methylpyrazole on human renal proximal

tubular (HK-2) cell viability...................................................................64

Figure 14. Aldehyde production in human renal proximal tubular

(HK-2) cells exposed to acyclovir...........................................................65

xxii

Figure 15. Comparison of the alcohol dehydrogenase (ADH) protein

expression level between the immortalized human renal

proximal tubular (HK-2) cell line and human kidney...........................66

Figure 16. The effect of 9-carboxymethoxymethylguanine (CMMG) on

human renal proximal tubular (HK-2 cell) viability..............................80

Figure 17. Tetraethylammonium (TEA) transport across porcine renal

proximal tubular cell (LLC-PK1) monolayers.......................................91

Figure 18. Tetraethylammonium (TEA) transport across human renal

proximal tubular cell (HK-2) monolayers..............................................92

Figure 19. Acyclovir transport across porcine renal proximal tubular

cell (LLC-PK1) monolayers...................................................................94

Figure 20. Acyclovir transport across human renal proximal tubular

cell (HK-2)monolayers...........................................................................95

Figure 21. The effect of acyclovir on creatinine transport across

porcine renal proximal tubular cell (LLC-PK1) monolayers.................97

Figure 22. The effect of acyclovir on creatinine transport across

human renal proximal tubular cell (HK-2) monolayers.........................98

Figure 23. The paracellular flux of mannitol across porcine renal

xxiii

proximal tubular cell (LLC-PK1) monolayers that were used

for determining the transepithelial transport of

tetraethylammonium (TEA) across the cell

monolayers............................................................................................109

Figure 24. The paracellular flux of mannitol across human renal proximal

tubular cell (HK-2) monolayers that were used for determining

the transepithelial transport of tetraethylammonium (TEA) across

the cell monolayers................................................................................110

Figure 25. The paracellular flux of mannitol across porcine renal

proximal tubular cell (LLC-PK1) monolayers that were

used for determining the transepithelial transport of

acyclovir across the cell monolayers.......................................................111

Figure 26. The paracellular flux of mannitol across human renal proximal

tubular cell (HK-2) monolayers that were used for

determining the transepithelial transport of acyclovir

across the cell monolayers.......................................................................112

Figure 27. The paracellular flux of mannitol across porcine renal

proximal tubular cell (LLC-PK1) monolayers that were

used to determine acyclovir inhibits the tubular

transport of creatinine..............................................................................113

Figure 28. The paracellular flux of mannitol across human renal

proximal tubular cell (HK-2) monolayers that were used to

xxiv

determine acyclovir inhibits the tubular

transport of creatinine.........................................................................114

Figure 29. The protein expression of human breast cancer resistance

protein (BCRP) in the overexpressing human embryonic

kidney (HEK293) cells.........................................................................123

Figure 30. The functionality of the human BCRP in overexpressing

human embryonic kidney (HEK293) cells............................................124

Figure 31. Intracellular accumulation of [8-14

C] acyclovir.....................................125

Figure 32. The effect of acyclovir on human embryonic kidney

(HEK293) cell viability..........................................................................134

1

Chapter 1

General introduction

1.1 Acyclovir – induced nephrotoxicity in children: new insight into its mechanism of

toxicity, interaction with creatinine and tubular transport.

Drug – induced nephrotoxicity is a serious adverse reaction observed in clinical practice that can

limit the use of effective therapeutic agents (Izzedine et al. 2005; Patzer 2008) and can have

detrimental effects on a patient’s overall health and well-being. Acyclovir is an example of such

an agent. The overall objective of this thesis is to gain new insight into the nephrotoxicity of an

old, yet widely used antiviral agent. The following chapters will reveal findings from novel

studies that examined the mechanism of its direct tubular injury, its interaction with creatinine

and its tubular transport.

1.2 Acyclovir

1.2.1 Acyclovir use

Figure 1. Structural formula of acyclovir (M.W. 225.2)

For over 25 years, acyclovir [9-(2-hydroxyethoxymethyl)guanine] has been routinely used to

treat several types of viral infections in children (Elion 1983; Richards et al. 1983; Wagstaff et

al. 1994). Acyclovir is most effective against herpes mediated viruses, including the herpes

2

simplex virus (types I and II) and the varicella zoster virus (Elion 1983). Acyclovir can be

administered topically, orally or intravenously (Richards et al. 1983). This thesis examines the

adverse renal effects of acyclovir that has been administered via the intravenous route to

children. Acyclovir is typically administered [via slow infusion] over a period of 1 hour, every

8 hours for 5 to 10 days (Bryson 1984).

The pharmacokinetic profile of acyclovir is zero – order (Whitley et al. 1982). The

pharmacokinetics of acyclovir is similar between adults and children over 1 year of age (Blum et

al. 1982). Compared to patients over 1 year of age, the total clearance is approximately one

third less and the half – life of acyclovir is increased by 1 hour in children less than 1 year of age

(Blum et al. 1982).

Table 1. Summary of some important pharmacokinetic parameters of acyclovir in children [1 –

17 years] (Hintz et al. 1982).

Acyclovir Dose

(mg/kg)

Steady State Peak

Plasma Levels

(µg/mL)

Half – Life

(hr)

Steady State

Volume of

Distribution

(L/1.73 m2)

Total Clearance

(mL/min/1.73 m2)

5 – 15 10 – 20 3 45 335

1.2.2 Acyclovir: mechanism of action

The mechanism of action of acyclovir has been completely elucidated. The antiviral activity of

acyclovir is a result of its inhibition of viral deoxyribonucleic acid (DNA) replication (Richards

et al. 1983). Acyclovir is known to act at three points along the viral DNA replication pathway

(Elion 1983; Richards et al. 1983). First, acyclovir competes with deoxynucleosides for

phosphorylation by viral or cellular thymindine kinase. The phosphorylated acyclovir, acyclovir

triphosphate then competes with deoxynucleoside triphosphates for viral DNA polymerase, and

is subsequently incorporated into the growing viral DNA strand. Acyclovir does not have a 3’

3

hydroxyl group that is required for DNA elongation, and therefore, incorporation of acyclovir

triphosphate into the DNA strand results in DNA chain termination (Elion 1983; Richards et al.

1983).

Figure 2. Schematic diagram illustrating the mechanism of action of acyclovir (Richards et al.

1983).

1.2.3 Acyclovir metabolism and excretion

Acyclovir does not require biotransformation to an active metabolite for its antiviral activity or

for its excretion (de Miranda et al. 1982). Intravenously administered acyclovir undergoes

minimal metabolism in humans (de Miranda et al. 1982). For example, for a given dose of

acyclovir, approximately 62 – 91 % is eliminated unchanged in the urine (de Miranda et al.

1982). The major metabolite of acyclovir is 9-carboxymethoxymethylguanine (CMMG), while

4

its minor metabolite is 8-hydroxy-9-(2-hydroxyethoxymethyl)guanine (de Miranda et al. 1982).

An estimated 8 – 14 % and less than 0.2 % of a given dose of acyclovir is eliminated as the

CMMG and 8-hydroxy-9-(2-hydroxyethoxymethyl)guanine metabolites, respectively, in the

kidney (de Miranda et al. 1982). Acyclovir is excreted to a minor extent in feces (< 2 %) and

expired air (< 0.1 %) (de Miranda et al. 1982).

Figure 3. Metabolism of acyclovir in humans (de Miranda et al. 1982; Helldén et al. 2006).

5

1.2.4 Acyclovir – induced nephrotoxicity

Acyclovir is widely regarded as a safe antiviral agent (Bryson 1984; Keeney et al. 1982).

Generally, the drug is well tolerated, with only minor irritation at the site of injection (Bryson

1984; Keeney et al. 1982). However, severe nephrotoxicity which often leads to acute renal

failure has been observed in patients (Ahmad et al. 1994; Bianchetti et al. 1991; Brigden et al.

1982; Chou et al. 2008; Genc et al. 2010; Keeney et al. 1982; Vachvanichsanong et al. 1995;

Vomiero et al. 2002). Acyclovir – induced renal failure occurs in approximately 12 to 48 % of

cases (Bean and Aeppli 1985; Keeney et al. 1982). Acyclovir – induced nephrotoxicity is

typically evidenced by acute renal failure, elevated plasma creatinine levels or the occurrence of

abnormal urine sediments (Ahmad et al. 1994; Bianchetti et al. 1991; Brigden et al. 1982; Chou

et al. 2008; Keeney et al. 1982; Vachvanichsanong et al. 1995; Vomiero et al. 2002).

1.2.5 Mechanism(s) of acyclovir – induced nephrotoxicity

1.2.5a Acyclovir – induced crystalluria

Acyclovir – induced nephrotoxicity is believed to be secondary to crystalluria which leads to

obstructive nephropathy (Bianchetti et al. 1991; Lyon et al. 2002; Mason 2008; Peterslund et al.

1998; Sawyer et al. 1988). Typically, crystalluria develops within 24 – 48 hours of the initiation

of acyclovir therapy (Izzedine et al. 2005). Polarizing microscopy shows that acyclovir forms

birefringent needle-shaped crystals in the urine (Genc et al. 2010; Lyon et al. 2002; Mason

2008; Sawyer et al. 1988). Strategies including avoidance of rapid bolus intravenous injection,

sufficient hydration and dose adjustments are often recommended for the prevention of

acyclovir – induced crystalluria (Brigden et al. 1982; Sawyer et al. 1988).

6

1.2.5b Acyclovir – induced direct renal tubular cell injury

Clinical evidence of nephrotoxicity in the absence of crystalluria (Ahmad et al. 1994; Vomiero

et al. 2002) suggests that acyclovir may also induce direct insult to renal tubular cells. For

example, renal biopsies show that acyclovir administration is associated with the occurrence of

various degenerative changes in tubular epithelial cells including bulging (Vomiero et al. 2002),

flattened and vacuolated epithelial cells (Ahmad et al. 1994; Vomiero et al. 2002).

Additionally, dilated tubular lumens, the presence of casts in the tubular lumen (Vomiero et al.

2002), loss of proximal-distal tubular differentiation and epithelial cell mitoses, which have

been suggested to be the result of acute tubular necrosis, have been reported in patients (Ahmad

et al. 1994). Studies have not investigated whether acyclovir induces direct insult to renal

tubular epithelial cells.

1.2.6 Acyclovir aldehyde: its potential role in direct renal tubular injury

Acyclovir is metabolized by alcohol dehydrogenase to produce an aldehyde metabolite, acyclovir

aldehyde, which is subsequently metabolized by aldehyde dehydrogenase to form the CMMG

metabolite (Figure 3). Aldehydes are reactive chemicals that are frequently produced

endogenously as intermediate drug metabolites (O’Brien et al. 2005). The findings from

numerous studies suggest that aldehyde metabolites mediate the toxicities [i.e. hepatotoxicity,

neurotoxicity, bladder toxicity, nephrotoxicity] that are associated with their parent drugs

(O’Brien et al. 2005). For example, the bladder toxicity that is associated with the

chemotherapeutic drug, cyclophosphamide is believed to be caused by its aldehyde metabolite,

acrolein (Ramu et al. 1995). Similarly, the chloroacetaldehyde metabolite of ifosfamide

(Walker et al. 1994) has been shown to cause the nephrotoxicity that occurs during

7

administration of the chemotherapeutic agent (Dubourg et al. 2001). While, studies suggest that

the atropaldehyde metabolite may be responsible for the hepatotoxicity and aplastic anemia that

is associated with its parent antiepileptic drug, felbamate (Kapetanovic et al. 2002). Therefore, it

is possible that the acyclovir aldehyde metabolite may cause the direct renal tubular injury that is

associated with the parent antiviral agent; this hypothesis has never been tested.

1.3 Acyclovir and creatinine: interaction during tubular secretion?

1.3.1 Creatinine

Creatinine is an endogenous compound that is produced non-enzymatically from creatine in

skeletal muscle (Toto 1995). Once produced in the skeletal muscle, creatinine is transported into

the blood and then excreted in the kidney (Toto 1995). Creatinine is freely filtered by the

glomerulus and it is not re-absorbed to a significant extent (Toto 1995). Thus, plasma creatinine

is a widely used measure of renal function in clinical practice (Levey et al. 1988; Narayanan and

Appleton 1980; Perrone et al. 1992; Toto 1995). Plasma creatinine levels are used to calculate

the Glomerular Filtration Rate (GFR) (Levey et al. 1988; Narayanan and Appleton 1980; Perrone

et al. 1992; Toto 1995). The GFR is a measure of the amount of fluid that filters into the

Bowman’s capsule per unit time (Silverthorn 1988). Plasma creatinine concentration is inversely

related to GFR (Levey et al. 1988; Narayanan and Appleton 1980; Perrone et al. 1992; Toto

1995). Therefore, increased plasma creatinine concentrations indicate impaired renal function

(Levey et al. 1988; Narayanan and Appleton 1980; Perrone et al. 1992; Toto 1995).

8

1.3.2 Renal tubular secretion of creatinine: opportunity for interaction with other drugs

and subsequent consequences

In addition to filtration, approximately 10 – 20 % of the body load of creatinine is secreted into

the kidney (Toto 1995). Creatinine is secreted into the tubule lumen via active transporter

systems (Arendshorst and Selkurt 1970; Berglund et al. 1975; Burgess et al. 1982; Burry and

Dieppe 1976; Dubb et al. 1978; Dutt et al. 1981; Eisner et al. 2010; Kastrup et al. 1985; Myre et

al. 1987; Okuda et al. 2006; Opravil et al. 1993; Tschuppert et al. 2007; Urakami et al. 2004).

The renal tubular transport mechanisms of creatinine have not been fully elucidated; however,

both acid and base active secreting mechanisms appear to play a role in its transport.

The active renal secretion of creatinine creates the opportunity for other drugs that may share

similar transport mechanisms with the compound; to compete with it for tubular secretion. The

competition between creatinine and other agents for renal tubular transport results in the

inhibition of the secretion of creatinine and a subsequent elevation in plasma creatinine levels

that are not due to decreased GFR or renal function. Examples of some non-nephrotoxic drugs

that inhibit the renal tubular secretion of creatinine and subsequently induce transient,

pronounced elevations in plasma creatinine levels that are unreflective of impaired renal function

include cimetidine (Blackwood et al. 1976; Burgess et al. 1982; Dubb et al. 1978; Dutt et al.

1981; Haggie et al. 1976), dronedarone (Tschuppert et al. 2007), pyrimethamine (Opravil et al.

1993), salicylates (Burry and Dieppe 1976) and trimethoprim (Berglund et al. 1975; Kastrup et

al. 1985; Myre et al. 1987).

A review of the literature shows that similar to the aforementioned non-nephrotoxic drugs;

marked, transient elevations (up to 9 fold above baseline levels in some cases) in plasma

9

creatinine levels have been observed within 24 – 48 hrs of initiation of acyclovir therapy in

patients (Bianchetti et al. 1991; Brigden et al. 1982; Chou et al. 2008; Keeney et al. 1982;

Vachvanichsanong et al. 1995; Vomiero et al. 2002). The pronounced increases in plasma

creatinine levels are often unaccompanied by signs of overt nephrotoxicity (please refer to Table

3, Chapter 5, for a summary of the acyclovir cases).

Studies reveal that like the non-nephrotoxic drugs, acyclovir may share similar renal organic

cation and anion transporter systems with creatinine (Takeda et al. 2002). Therefore it is

plausible the acyclovir inhibits the tubular secretion of creatinine. It is imperative to determine

whether acyclovir inhibits the secretion of creatinine because if this is the case, then in addition

to creatinine, other biological markers of renal function, such as inulin, should always be used to

assess renal function in patients during the course of acyclovir therapy. Research has not

elucidated whether acyclovir inhibits the renal tubular secretion of creatinine. In this thesis, the

inhibition of creatinine secretion by acyclovir via the organic cation transporter (OCT) system

was examined.

1.4 Role of the human breast cancer resistance protein (BCRP) in the transport of

acyclovir: potential implications in tubular transport and nephrotoxicity

1.4.1 BCRP

The BCRP is the second member of the subfamily G of the human adenosnine triphosphate

(ATP) – binding cassette (ABC) transporter superfamily (Dean et al. 2001; Mau and Unadkat

2005; Robey et al. 2009). The efflux transporter (Doyle et al. 1998; Rocchi et al. 2000) is

responsible for the transport of both endogenous (i.e. 17β-estradiol) (Chen et al. 2003) and

exogenous substrates (i.e. mitoxantrone, daunorubicin) (Doyle et al. 1998; Ozvegy et al. 2001).

10

The protein is widely expressed in human tissues (Allikmets et al. 1998; Doyle et al. 1998;

Maliepaard et al. 2001) including the placenta, gastrointestinal tract, breast, liver and kidney.

1.4.2 Acyclovir as a potential substrate of human BCRP: renal tubular transport and

toxicological significance

Jonker and colleagues have shown that in mice with the wildtype Abcg2 gene, which codes for

Bcrp1 protein (murine ortholog of human ABCG2 gene, which codes for the BCRP protein),

there was a significantly higher accumulation (approximately 5 fold) of acyclovir in breast milk,

compared to mice with the non-functional Abcg2-/-

gene. The results suggest that acyclovir is a

substrate for murine Bcrp1 and hence, the antiviral agent may also be a substrate for human

BCRP (Jonker et al. 2005), however, this hypothesis have never been directly tested.

It is important to determine whether acyclovir is a substrate for human BCRP because this may

aid in the better understanding of the pathogenesis of the direct renal tubular injury induced by

the drug. The efflux transporter is localized in the apical membrane of renal tubular cells (Huls

et al. 2008), and hence, may play a significant role in the efflux of acyclovir from tubular cells.

Therefore, factors, such as genetic polymorphisms (Sparreboom et al. 2004; Cusatis et al. 2006;

Zhang et al. 2006; Yu et al. 2006; Yamasaki et al. 2008; Pollex et al. 2010) that affect functional

expression of BCRP may result in the reduced or abolished renal tubular expression of the efflux

transporter. Reduced or abolished expression of the transporter can result in the reduced cellular

efflux and increased intracellular concentration of acyclovir and subsequent detrimental

nephrotoxic consequences, such as direct tubular injury.

11

1.5 References

Ahmad, T., Simmonds, M., McIver, A.G., and McGraw, M.E. 1994. Reversible renal failure in

renal transplant patients receiving oral acyclovir prophylaxis. Pediatr Nephrol 8: 489-491.

Allikmets, R., Schriml, L.M., Hutchinson, A., Romano-Spica, V., and Dean, M. 1998. A human

placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in

multidrug resistance. Cancer Res 58: 5337-5339.

Arendshorst, W.J., and Selkurt, E.E. 1970. Renal tubular mechanisms for creatinine secretion in

the guinea pig. Am J Physiol 218: 1661-1670.

Bean, B., and Aeppli, D. 1985. Adverse effects of high-dose intravenous acyclovir in ambulatory

patients with acute herpes zoster. J Infect Dis 151: 362-365.

Berglund, F., Killander, J., and Pompeius, R. 1975. Effect of trimethoprim-sulfamethoxazole on

the renal excretion of creatinine in man. J Urol 114: 802-808.

Bianchetti, M.G., Roduit, C., and Oetliker, O.H. 1991. Acyclovir-induced renal failure: course

and risk factors. Pediatr Nephrol 5: 238-239.

Blackwood, W.S., Maudgal, D.P., Pickard, R.G., Lawrence, D., and Northfield, T.C. 1976.

Cimetidine in duodenal ulcer. Controlled trial. Lancet 2: 174-176.

Blum, M.R., Liao, S.H., and de Miranda, P. 1982. Overview of acyclovir pharmacokinetic

disposition in adults and children. Am J Med 73: 186-192.

Brigden, D., Rosling, A.E., and Woods, N.C. 1982. Renal function after acyclovir intravenous

injection. Am J Med 73: 182-185.

Bryson, Y.J. 1984. The use of acyclovir in children. Pediatr Infect Dis 3: 345-348.

Burgess, E., Blair, A., Krichman, K., and Cutler, R.E. 1982. Inhibition of renal creatinine

secretion by cimetidine in humans. Ren Physiol 5: 27-30.

Burry, H.C., and Dieppe, P.A. 1976. Apparent reduction of endogenous creatinine clearance by

salicylate treatment. Br Med J 2: 16-17.

Chen, Z.S., Robey, R.W., Belinsky, M.G., Shchaveleva, I., Ren, X.Q., Sugimoto, Y., Ross, D.D.,

Bates, S.E., and Kruh, G.D. 2003. Transport of methotrexate, methotrexate polyglutamates, and

17beta-estradiol 17-(beta-D-glucuronide) by ABCG2: effects of acquired mutations at R482 on

methotrexate transport. Cancer Res 63: 4048-4054.

Chou, J.W., Yong, C., and Wootton, S.H. 2008. Case 2: Rash, fever and headache....first, do no

harm. Paediatr Child Health 13: 49-52.

12

de Miranda, P., Good, S.S., Krasny, H.C., Connor, J.D., Laskin, O.L., and Lietman, P.S. 1982.

Metabolic fate of radioactive acyclovir in humans. Am J Med 73: 215-220.

Doyle, L.A., Yang, W., Abruzzo, L.V., Krogmann, T., Gao, Y., Rishi, A.K., and Ross, D.D.

1998. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad

Sci U S A 95: 15665-15670.

Dubb, J.W., Stote, R.M., Familiar, R.G., Lee, K., and Alexander, F. 1978. Effect of cimetidine

on renal function in normal man. Clin Pharmacol Ther 24: 76-83.

Dubourg, L., Michoudet, C., Cochat, P., and Baverel, G. 2001. Human kidney tubules detoxify

chloroacetaldehyde, a presumed nephrotoxic metabolite of ifosfamide. J Am Soc Nephrol 12:

1615-1623.

Dutt, M.K., Moody, P., and Northfield, T.C. 1981. Effect of cimetidine on renal function in man.

Br J Clin Pharmacol 12: 47-50.

Eisner, C., Faulhaber-Walter, R., Wang, Y., Leelahavanichkul, A., Yuen, P.S., Mizel, D., Star,

R.A., Briggs, J.P., Levine, M., and Schnermann, J. 2010. Major contribution of tubular secretion

to creatinine clearance in mice. Kidney Int 77: 519-526.

Elion, G.B. 1983. The biochemistry and mechanism of action of acyclovir. J Antimicrob

Chemother 12: 9-17.

Genc, G., Ozkaya, O., Acikgoz, Y., Yapici, O., Bek, K., Gulnar Sensoy, S., and Ozyurek, E.

2010. Acute renal failure with acyclovir treatment in a child with leukemia. Drug Chem Toxicol

33: 217-219.

Haggie, S.J., Fermont, D.C., and Wyllie, J.H. 1976. Treatment of duodenal ulcer with cimetidine.

Lancet 1: 983-984.

Hellden, A., Lycke, J., Vander, T., Svensson, J.O., Odar-Cederlof, I., and Stahle, L. 2006. The

aciclovir metabolite CMMG is detectable in the CSF of subjects with neuropsychiatric symptoms

during aciclovir and valaciclovir treatment. J Antimicrob Chemother 57: 945-949.

Hintz, M., Connor, J.D., Spector, S.A., Blum, M.R., Keeney, R.E., and Yeager, A.S. 1982.

Neonatal acyclovir pharmacokinetics in patients with herpes virus infections. Am J Med 73: 210-

214.

Huls, M., Brown, C.D., Windass, A.S., Sayer, R., van den Heuvel, J.J., Heemskerk, S., Russel,

F.G., and Masereeuw, R. 2008. The breast cancer resistance protein transporter ABCG2 is

expressed in the human kidney proximal tubule apical membrane. Kidney Int 73: 220-225.

Izzedine, H., Launay-Vacher, V., and Deray, G. 2005. Antiviral drug-induced nephrotoxicity.

Am J Kidney Dis 45: 804-817.

13

Jonker, J.W., Merino, G., Musters, S., van Herwaarden, A.E., Bolscher, E., Wagenaar, E.,

Mesman, E., Dale, T.C., and Schinkel, A.H. 2005. The breast cancer resistance protein BCRP

(ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat Med 11: 127-129.

Kapetanovic, I.M., Torchin, C.D., Strong, J.M., Yonekawa, W.D., Lu, C., Li, A.P., Dieckhaus,

C.M., Santos, W.L., Macdonald, T.L., Sofia, R.D., and Kupferberg, H.J. 2002. Reactivity of

atropaldehyde, a felbamate metabolite in human liver tissue in vitro. Chem Biol Interact 142:

119-134.

Kastrup, J., Petersen, P., Bartram, R., and Hansen, J.M. 1985. The effect of trimethoprim on

serum creatinine. Br J Urol 57: 265-268.

Keeney, R.E., Kirk, L.E., and Bridgen, D. 1982. Acyclovir tolerance in humans. Am J Med 73:

176-181.

Levey, A.S., Perrone, R.D., and Madias, N.E. 1988. Serum creatinine and renal function. Annu

Rev Med 39: 465-490.

Lyon, A.W., Mansoor, A., and Trotter, M.J. 2002. Urinary gems: acyclovir crystalluria. Arch

Pathol Lab Med 126: 753-754.

Maliepaard, M., Scheffer, G.L., Faneyte, I.F., van Gastelen, M.A., Pijnenborg, A.C., Schinkel,

A.H., van De Vijver, M.J., Scheper, R.J., and Schellens, J.H. 2001. Subcellular localization and

distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer

Res 61: 3458-3464.

Mason, W.J., and Nickols, H.H. 2008. Crystalluria from acyclovir use. N Engl J Med 358: e14.

Myre, S.A., McCann, J., First, M.R., and Cluxton, R.J., Jr. 1987. Effect of trimethoprim on

serum creatinine in healthy and chronic renal failure volunteers. Ther Drug Monit 9: 161-165.

Narayanan, S., and Appleton, H.D. 1980. Creatinine: a review. Clin Chem 26: 1119-1126.

O'Brien, P.J., Siraki, A.G., and Shangari, N. 2005. Aldehyde sources, metabolism, molecular

toxicity mechanisms, and possible effects on human health. Crit Rev Toxicol 35: 609-662.

Okuda, M., Kimura, N., and Inui, K. 2006. Interactions of fluoroquinolone antibacterials, DX-

619 and levofloxacin, with creatinine transport by renal organic cation transporter hOCT2. Drug

Metab Pharmacokinet 21: 432-436.

Opravil, M., Keusch, G., and Luthy, R. 1993. Pyrimethamine inhibits renal secretion of

creatinine. Antimicrob Agents Chemother 37: 1056-1060.

Patzer, L. 2008. Nephrotoxicity as a cause of acute kidney injury in children. Pediatric

Nephrology 23: 2159-2173.

14

Perrone, R.D., Madias, N.E., and Levey, A.S. 1992. Serum creatinine as an index of renal

function: new insights into old concepts. Clin Chem 38: 1933-1953.

Peterslund, N.A., Larsen, M.L., and Mygind, H. 1988. Acyclovir crystalluria. Scand J Infect Dis

20: 225-228.

Ramu, K., Fraiser, L.H., Mamiya, B., Ahmed, T., and Kehrer, J.P. 1995. Acrolein mercapturates:

synthesis, characterization, and assessment of their role in the bladder toxicity of

cyclophosphamide. Chem Res Toxicol 8: 515-524.

Richards, D.M., Carmine, A.A., Brogden, R.N., Heel, R.C., Speight, T.M., and Avery, G.S.

1983. Acyclovir. A review of its pharmacodynamic properties and therapeutic efficacy. Drugs

26: 378-438.

Sawyer, M.H., Webb, D.E., Balow, J.E., and Straus, S.E. 1988. Acyclovir-induced renal failure.

Clinical course and histology. Am J Med 84: 1067-1071.

Silverthorn, D.U. 1998. The Kidneys. In Human Physiology An Integrated approach. Edited by

Brake, D.R.. Prentice Hall., Upper Saddle River, New Jersey. pp. 518-542.

Takeda, M., Khamdang, S., Narikawa, S., Kimura, H., Kobayashi, Y., Yamamoto, T., Cha, S.H.,

Sekine, T., and Endou, H. 2002. Human organic anion transporters and human organic cation

transporters mediate renal antiviral transport. J Pharmacol Exp Ther 300: 918-924.

Toto, R.D. 1995. Conventional measurement of renal function utilizing serum creatinine,

creatinine clearance, inulin and para-aminohippuric acid clearance. Curr Opin Nephrol

Hypertens 4: 505-509.

Tschuppert, Y., Buclin, T., Rothuizen, L.E., Decosterd, L.A., Galleyrand, J., Gaud, C., and

Biollaz, J. 2007. Effect of dronedarone on renal function in healthy subjects. Br J Clin Pharmacol

64: 785-791.

Urakami, Y., Kimura, N., Okuda, M., and Inui, K. 2004. Creatinine transport by basolateral

organic cation transporter hOCT2 in the human kidney. Pharm Res 21: 976-981.

Vachvanichsanong, P., Patamasucon, P., Malagon, M., and Moore, E.S. 1995. Acute renal failure

in a child associated with acyclovir. Pediatr Nephrol 9: 346-347.

Vomiero, G., Carpenter, B., Robb, I., and Filler, G. 2002. Combination of ceftriaxone and

acyclovir - an underestimated nephrotoxic potential? Pediatr Nephrol 17: 633-637.

Wagstaff, A.J., Faulds, D., and Goa, K.L. 1994. Aciclovir. A reappraisal of its antiviral activity,

pharmacokinetic properties and therapeutic efficacy. Drugs 47: 153-205.

Walker, D., Flinois, J.P., Monkman, S.C., Beloc, C., Boddy, A.V., Cholerton, S., Daly, A.K.,

Lind, M.J., Pearson, A.D., Beaune, P.H., and Jeffrey, R.I. 1994. Identification of the major

15

human hepatic cytochrome P450 involved in activation and N-dechloroethylation of ifosfamide.

Biochem Pharmacol 47: 1157-1163.

Whitley, R.J., Blum, M.R., Barton, N., and de Miranda, P. 1982. Pharmacokinetics of acyclovir

in humans following intravenous administration. A model for the development of parenteral

antivirals. Am J Med 73: 165-171.

16

Chapter 2

Hypotheses and Objective

2.1 Hypotheses

The preceding chapter highlights that there are several important knowledge gaps in the study of

the nephrotoxicity that is induced by the widely used antiviral agent, acyclovir. To summarize;

to date, research has not investigated whether: (1) acyclovir induces direct insult to renal tubular

cells, (2) the acyclovir aldehyde metabolite plays a role in the pathogenesis of this

nephrotoxicity, (3) whether the antiviral agent inhibits the tubular secretion of the biological

marker of renal function, creatinine and (4) whether acyclovir is a substrate for the human BCRP

efflux transporter which is expressed in the human kidney and therefore, may have important

toxicological consequences in the pathogenesis of its direct renal tubular injury.

Therefore, the following hypotheses were derived for this thesis:

(I) Acyclovir – induced nephrotoxicity is due to, in part, direct insult to renal tubular

cells.

(II) Acyclovir aldehyde plays a role in the direct renal tubular injury induced by

acyclovir.

(III) Acyclovir inhibits the renal tubular secretion of creatinine.

(IV) Acyclovir is a substrate for human BCRP.

17

2.2 Objectives

The objectives of this thesis were:

(I) To determine whether acyclovir – induced nephrotoxicity is due to, in part, direct

insult to renal tubular cells.

(II) To determine whether acyclovir aldehyde plays a role in the direct renal tubular

injury induced by acyclovir.

(III) To determine whether acyclovir inhibits the renal tubular secretion of creatinine.

(IV) To determine whether acyclovir is a substrate for human BCRP.

18

Chapter 3

Comparison of the novel HK-2 human renal proximal tubular cell

line with the standard LLC-PK1 cell line in studying drug-induced

nephrotoxicity

Patrina Gunness,a,b

Katarina Aleksa,a Kazuhiro Kosuge,

a Shinya Ito,

a,b Gideon Koren

a,b

aDivision of clinical Pharmacology and Toxicology, The Hospital for Sick Children, 555

University Avenue, Toronto, ON, M5G 1X8, Canada

bGraduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University

of Toronto, ON, M5S 3M2, Canada

This article has been published: Gunness, P., Aleksa, K., Kosuge, K., Ito, S., and Koren, G.

2010. Comparison of the novel HK-2 human renal proximal tubular cell line with the standard

LLC-PK1 cell line in studying drug-induced nephrotoxicity. Can J Physiol Pharmacol 88: 448-

455. This article was originally published by NRC Research Press.

[PG performed the acyclovir experiments and prepared the manuscript for submission; KA and

KK performed the ifosfamide experiments]

19

3.1 Abstract

Established cell lines are widely used as in vitro models in toxicology studies. The choice of an

appropriate cell line is critical when performing studies to elucidate drug-induced toxicity in

humans. The porcine renal proximal tubular cell line (LLC-PK1) is routinely used to study the

nephrotoxic effects of drugs in humans. However, there are significant interspecies differences in

drug pharmacokinetics and pharmacodynamics. The objective of this study was to determine

whether the human renal proximal tubular cell line (HK-2) is an acceptable model to use when

performing in vitro toxicity studies to predict effects in humans. We examined 2 nephrotoxic

agents, ifosfamide and acyclovir that exhibit different clinical nephrotoxic patterns. HK-2 cells

metabolized IFO to its nephrotoxic metabolite, chloroacetaldehyde. Acyclovir induced a

concentration-dependent decrease in HK-2 cell viability, suggesting that acyclovir may induce

direct insult to renal proximal tubular cells. The results support clinical pathology data in humans

and suggest that HK-2 cells are a suitable model to use in in vitro toxicity studies to determine

drug-induced nephrotoxicity in humans.

3.2 Introduction

Over the past 25 yrs, in vitro models have become widely used in toxicology studies, with

established cell lines being the most common models for in vitro toxicity studies (Zucco et al.

2004). The choice of the appropriate cell line may be critical when performing in vitro studies to

elucidate the mechanisms of drug-induced toxicity in humans. The porcine renal proximal

tubular cell line LLC-PK1 is routinely used to study drug-induced nephrotoxicity.

LLC-PK1 cells, derived from the kidney of a juvenile male Hampshire pig, retain morphological

and biochemical characteristics similar to those of human renal proximal tubular cells (Perantoni

20

and Berman 1979). However, there are significant interspecies differences in drug disposition

(Riddick 1998; Eaton and Klaassen 2001). Therefore, caution must be taken when extrapolating

results from LLC-PK1 cells to humans. Conceptually, the use of a cell line derived from the

human kidney would be a more appropriate model to use for in vitro toxicity studies.

The human renal proximal tubular cell line HK-2 was derived from the healthy kidney of an

adult male (Ryan et al. 1994). Similar to LLC-PK1 cells, the HK-2 cell line has retained

morphological and biochemical characteristics consistent with those of human renal proximal

tubular cells (Ryan et al. 1994). To validate HK-2 cells as a nephrotoxic model and compare it

with LLC-PK1 cells, we used 2 nephrotoxic drugs with substantial serious effects in children.

Ifosfamide is an alkylating agent that is used in the treatment of various pediatric tumors,

including Wilms' tumor, rhabdomyosarcoma, neuroblastoma, bone sarcomas, and soft tissue

sarcomas (Sladek 1988; Carli et al. 2003). However, ifosfamide induces nephrotoxicity in

approximately 30 % of treated children (Skinner et al. 1996). Ifosfamide – induced

nephrotoxicity is characterized by renal glomerular and tubular damage (Skinner et al. 1996;

Loebstein and Koren 1998; Skinner 2003). Severe cases of ifosfamide – induced nephrotoxicity

are characterized by the Fanconi syndrome (Aleksa et al. 2005a; Rossi et al. 1999), in which

phosphate, glucose, amino acids, and low molecular weight proteins are lost from the renal

tubules (Rossi et al. 1999; Skinner 2003).

Ifosfamide is a prodrug that is metabolized in vivo to the alkylating ifosfamide mustard (Brade et

al. 1985) by various cytochrome P450 (CYP) enzymes (CYP 3A4, CYP 3A5, CYP 3A7, and

CYP 2B6) (Chang et al. 1993; Walker et al. 1994; Chen et al. 2005; McCune et al. 2005). The

metabolism of ifosfamide results in production of the nephrotoxic metabolite chloroacetaldehyde

21

(Aleksa et al. 2005a), and therefore cell lines that are used to study ifosfamide nephrotoxicity

must possess the enzymes necessary for metabolizing the drug to chloroacetaldehyde.

Additionally, ifosfamide is administered clinically as a racemic mixture of its R and S

enantiomers (Roy et al. 1999). Therefore, cell lines employed in in vitro ifosfamide

nephrotoxicity studies should ideally be able to metabolize both enantiomers of ifosfamide.

Aleksa et al. (2005a) reported that LLC-PK1 cells are a suitable model for studying ifosfamide

nephrotoxicity. However, interspecies differences in drug disposition exist, and hence the results

obtained from ifosfamide nephrotoxicity studies that employ LLC-PK1 cells may not provide a

suitable prediction of events that could occur in humans.

Acyclovir (9-(2-hydroxyethoxymethyl) guanine) is an antiviral drug that is used in the treatment

of several types of viral infections in children, including herpes simplex virus types 1 and 2 and

varicella-zoster virus (Elion 1983; Richards et al. 1983; Wagstaff et al. 1994). There is minimal

toxicity observed with use of acyclovir, and local irritation at the site of injection is observed in

some patients (Keeney et al. 1982). However, nephrotoxicity and, in some cases, serious acute

renal failure have been reported in children and adults (Brigden et al. 1982; Peterslund et al.

1988; Sawyer et al. 1988; Becker et al. 1993; Ahmad et al. 1994; Vachvanichsanong et al. 1995).

Acyclovir is generally thought to induce nephrotoxicity via crystalluria, which leads to

obstructive nephropathy (Brigden et al. 1982; Sawyer et al. 1988; Lyon et al. 2002; Mason and

Nickols 2008). Adequate hydration, avoidance of rapid intravenous doses, and dose adjustments

are recommended preventative strategies for acyclovir-induced nephrotoxicity (Brigden et al.

1982; Sawyer et al. 1988). However, a recent study by Schreiber et al. (2008) found that

adequate hydration did not prevent nephrotoxicity in some children who received acyclovir

22

therapy. Additionally, there have been several reports of acyclovir-induced nephrotoxicity with

biopsy evidence of tubular damage in the absence of crystal formation (Becker et al. 1993;

Ahmad et al. 1994; Vomiero et al. 2002). These reports suggest that acyclovir induces direct

insult to renal tubular cells. Results from in vitro toxicity studies using an appropriate cell line

might enable us to determine whether acyclovir induces direct renal tubular damage and the

mechanisms by which this occurs in humans. Ideally, the in vitro studies should use a cell line,

such as HK-2, that has been derived from human kidney. Given that interspecies variation would

be absent, HK-2 should be the more appropriate model, compared with nonhuman-derived cell

lines, with which to predict acyclovir nephrotoxicity in humans.

In children, ifosfamide and acyclovir are first-line treatments for cancer and viral infections,

respectively. However, in some children, the use of ifosfamide or acyclovir results in severe

nephrotoxicity, which adversely affects the overall health of children. Therefore, elucidating the

mechanisms of this drug – induced nephrotoxicity will aid in the design of safer drug therapy for

children.

Currently, owing to interspecies differences in drug pharmacology and toxicology, the use of

LLC-PK1 cells in in vitro nephrotoxicity studies limits the extrapolation of results to humans.

The results from studies using a cell line derived from human kidney, such as the HK-2 cell line,

would be more applicable to humans. Therefore, we hypothesized that the HK-2 cell line is an

acceptable in vitro model to use in nephrotoxicity studies. The objective of this study was to

determine whether the HK-2 cell line is an acceptable cell culture model to use in in vitro studies

that are aimed at elucidating the etiology of drug-induced nephrotoxicity in humans.

23

3.3 Materials and methods

To determine the appropriateness of HK-2 cells as a model to study ifosfamide – induced

nephrotoxicity, the following experiments were conducted: (i) Reverse Transcription-Polymerase

Chain Reaction (RT-PCR) and western blots were performed to determine messenger RNA

(mRNA) and protein expression of CYP3A and CYP2B in HK-2 cells, (ii) high performance

liquid chromatography-mass spectrometry (LC-MS) was performed to determine the renal

proximal tubular metabolism of ifosfamide in HK-2 cells, and (iii) a standard glutathione (GSH)

colorimetric assay was used to determine depletion of GSH levels in HK-2 cells after exposure to

the GSH-depleting agent L-buthionine sulfoximine (BSO).

To determine whether HK-2 cells are an appropriate model to study acyclovir-induced

nephrotoxicity, HK-2 cells were exposed to a range of acyclovir concentrations [0 – 2000 µg/mL

(0 – 8.89 mmol/L)] for 24 hrs, and cytotoxicity was measured by alamarBlue™

assay.

3.3.1 Chemicals

Racemic ifosfamide (50/50 (R/S)-ifosfamide) and individual enantiomers of 2-

dechloroethylifosfamide [(R)-2-DCEIF, (S)-2-DCEIF] and 3-dechloroethylifosfamide [(R)-3-

DCEIF, (S)-3-DCEIF] were purchased from Niomech, Germany. Deuterated racemic 2-DCEIF

(d2-2-DCEIF) (50/50 R/S) and 3-DCEIF (d4-3-DCEIF) (50/50 R/S) were kindly provided by Dr.

Susan Ludeman of Duke University (USA). Individual ifosfamide enantiomers [(S)-ifosfamide

and (R)-ifosfamide] were kindly provided by Mr. Ben Skeed (Chiroscience, England) Acyclovir

sodium solution (Zovirax®) was purchased from the Hospital for Sick Children pharmacy

(Toronto, Ontario, Canada).

24

For western blotting, rabbit anti-human polyclonal peptide antibody to CYP 2B6, rabbit anti-

human polyclonal antibody to CYP 3A5, and rabbit anti-human polyclonal antibody to CYP

3A4 were all purchased from BD Biosciences (Mississauga, Ontario, Canada). Donkey anti-

rabbit immunoglobulin G – horseradish peroxidise (IgG-HRP) was purchased from Amersham

Bioscience (Baie d’Urfe, Québec, Canada). Fluorometric alamarBlue reagent was purchased

from Invitrogen Canada Inc. (Burlington, Canada. Glutathione colorimetric assay kits, GT-10,

were purchased from Oxford Biomedical Research (Rochester Hills, Michigan, USA).

3.3.2 HK-2 cells

HK-2 cells were purchased from the American Type Culture Collection (ATCC) (Manassas,

Virginia, USA). The ifosfamide and acyclovir experiments were conducted by different

laboratory personnel and at different times. Therefore, the HK-2 cells were cultured differently

for ifofamide and acyclovir experiments as outlined in the following sections. All experiments

were conducted on cells that were grown to 80 – 85 % confluence.

3.3.2a Culturing conditions of HK-2 cells for ifosfamide experiments

The cells were maintained according to the recommendations provided by Detrisac et al. 1984.

The cells were cultured in Dulbecco's modified Eagle's minimum essential medium/Ham's F-12

(DMEM/F12) supplemented with 10 % (v/v) fetal bovine serum (FBS) (Gibco, Burlington,

Ontario, Canada) with 2 µmol/L L-glutamine, 20 mmol/L Hepes buffer (Gibco), 10 mg/L inulin,

5.5 mg/L transferrin, 6.7 µg/L sodium selenite, 100 Units/mL penicillin, and 100 µg of

streptomycin. Cells were maintained at 37◦C in a sterile, humidified atmosphere of 5 % CO2 and

95% O2.

25

3.3.2b Culturing conditions of HK-2 cells for acyclovir experiments

The cells were maintained according to ATCC guidelines. Briefly, cells were cultured in

keratinocyte serum-free medium (SFM) supplemented with 5 ng/mL human recombinant

epidermal growth factor and 0.05 mg/mL bovine pituitary extract (Invitrogen Canada Inc.). Cells

were maintained at 37◦C in a sterile, humidified atmosphere of 5 % CO2 and 95 % O2.

3.3.3 LLC-PK1 cells

The LLC-PK1 cell line was purchased from ATCC. The cells were maintained according to

ATCC guidelines. All experiments were conducted on cells that were grown to 80 – 85 %

confluence.

3.3.3a Culturing conditions of LLC-PK1 cells for ifosfamide experiments

The cells were cultured in DMEM supplemented with 2 mmol/L L-glutamine, 100 Units/mL

penicillin, 100 µg streptomycin (Invitrogen Canada Inc.), and 10 % (v/v) FBS (Gibco). Cells

were maintained at 37◦C in a sterile, humidified atmosphere of 5 % CO2 and 95 % O2.

3.3.3b Culturing conditions of LLC-PK1 cells for acyclovir experiments

The cells were cultured in HyClone MEM alpha modified (α-MEM, Fisher Scientific, Ottawa,

Ontario, Canada), supplemented with 2 mmol/L L-glutamine, 100 Units/mL penicillin, 100 µg of

streptomycin (Invitrogen Canada Inc.), and 10 % (v/v) FBS (Invitrogen Canada Inc.). Cells were

maintained at 37◦C in a sterile, humidified atmosphere of 5 % CO2 and 95 % O2.

26

3.3.4 Experimental methods used to determine whether HK-2 cells are an appropriate

model to study ifosfamide – induced nephrotoxicity

3.3.4a Determination of CYP enzyme mRNA expression in HK-2 cells by RT-PCR

Total RNA was extracted from HK-2 cells by using an RNeasy kit (Qiagen, Toronto, Ontario,

Canada) and subsequently reverse transcribed after previous digestion of possible contaminating

genomic DNA. The reverse transcription reaction was performed with 10 µg of total RNA.

Previously published primers were chosen for PCR amplifications of CYP 3A4 [forward primer

5'-GCAAAGAGCAACACAGAGCTG-3'; reverse primer 5'-GTGATAGCCAGCACAGGCTG-

3'] (Xu et al. 2005), CYP 3A5 [forward primer 5'-GAAGAAAAGTCGCCTCAAC-3'; reverse

primer 5'-AAGAAGTCCTTGCGTGTGTCTA-3'] (Jover et al. 2001), and β-actin [forward

primer 5'-CTACAATGAGCTGCGTGTGG-3'; reverse primer 5'-

TAGCTCTTCTCCAGGGAGGA-3'] (Giannone et al. 1998). Specific primers were designed for

CYP3A7 [foward primer 5’-CCTCTGCCTTTTTTGGGAAATGC-3’; reverse primer 5'-

GAGCTTTGTGGGTCTCAGAG-3']. A 2 µmol/L reverse transcriptase product was used for

PCR amplification in a 20 µmol/L reaction. The annealing temperature was 61◦C for CYP 3A4,

CYP 3A5, CYP 3A7, and β-actin. The PCR reactions were optimized to ensure that PCR

products for each gene were obtained in the linear range of the reaction. Results were quantified

on a 1.5 % agarose gel and digitalized by Fluorchem (Innotech USA Inc., Tarrytown, New York,

USA).

Each PCR reaction consisted of 5 µmol/L of Expand High Fidelity buffer without magnesium

chloride (MgCl2) (Roche, Mississauga, Ontario, Canada), 0.2 mmol/L each deoxyribonucleotide

triphosphate (dNTP), 2.5 mmol/L MgCl2, 3 µmol/L of copy DNA (cDNA), 300 ng of each

27

primer, 3.5 Units of Expand High Fidelity enzyme (Roche), and water to a final volume of 50

mL. The amplification conditions were as follows: an initial denaturation at 95◦C for 5 mins

followed by 35 cycles at 95◦C for 30 secs, 54

◦C for 30 secs, and 72

◦C for 40 secs, with a final

elongation step of 72◦C for 10 mins. The experiment was not replicated.

3.3.4b Determination of CYP enzyme protein expression in HK-2 cells by Western blotting

The protein expression of CYP3A and CYP2B was detected in HK-2 cells by subjecting samples

(30 µg total protein was loaded in each lane) to one-dimensional sodium dodecyl sulfate –

polyacrylamide gel electrophoresis (SDS PAGE) on 10 % gels. CYP 3A4 and CYP 2B6

supersomes (BD Biosciences) were used to confirm the presence of these proteins. Proteins were

transferred to a hydrophobic polyvinylidene difluoride (PVDF) membrane (Amersham).

Nonspecific sites were blocked with 5 % (w/v) low-fat milk in phosphate-buffered saline (PBS)

solution overnight at 4◦C, and proteins were incubated with polyclonal antibodies (anti-CYP 3A5

or anti-CYP 3A4 (1:500) or anti-CYP 2B6 (1:500)). Membranes were then incubated with

donkey anti-rabbit IgG horseradish peroxidase-linked secondary antibody at a dilution of 1:8000

and developed with the Amersham enhanced chemiluminescence (ECL) detection cocktail. The

ECL-detected blots were exposed to radiographic film (Hyperfilm ECL, Amersham) and

developed with a Kodak developer. The experiment was performed in replicates of 3.

3.3.4c Determination of renal proximal tubule metabolism of ifosfamide in HK-2 cells by

LC-MS

The HK-2 or LLC-PK1 cells were seeded and allowed to rest for 24 hrs after which they were

incubated with media containing (R)-ifosfamide or (S)-IF (0, 1, 10, 100, and 1000 µmol/L) for

28

96 hrs. Cell culture media were pooled, and on the final day of the experiment the cells were

solubilized with 100 µmol/L of 0.1 % (w/v) SDS containing 1 mmol/L EDTA to assess

metabolite production. The samples were flash-frozen in liquid nitrogen and stored at -80◦C so

that they could be analyzed in batches. On the day of the experiment, 50 ng of d6-2-DCEIF and

50 ng of d4-3-DCEIF (internal standards) were added, and the samples were vortexed for 30 secs.

Five mL of methylene chloride was added, and the samples were vortexed for 1 min and then

centrifuged at 2095 x g for 10 mins at 4◦C. Once the aqueous layer was removed, the organic

layer was dried under nitrogen. Samples were reconstituted with 30 µmol/L of ethyl acetate. A 1

µmol/L sample was analyzed for (R)-IF, (S)-IF, (R)-2-DCEIF, (S)-2-DCEIF, (R)-3-DCEIF, and

(S)-3-DCEIF by using LC-MS. The experiment was replicated 3 times. Results are presented as

means ± SD of 3 independent experiments.

LC-MS conditions

An 1100 series high performance liquid chromatography (HPLC) (Agilent Technologies,

Mississauga, Ontario, Canada) equipped with an 1100 series binary pump was used for analysis.

Analytes were separated with a Chiral-AGP column (150 mm x 4.0 mm; 5 µm) (Chrom Tech,

Madison, Wisconsin, USA) by using (i) 10 mmol/L ammonium acetate in water (pH 7) and (ii)

30 mmol/L ammonium acetate in water (pH 4). Samples were introduced into the

chromatographic system with an Agilent 1100 series autosampler. Analysis was carried out

under ambient temperatures.

Mass spectrometric detection was performed on an LC-MS/MS API4000 triple-quadrupole

(Applied Biosystems, MDS Sciex, Foster City, California, USA) equipped with an API turbo ion

spray ionization source. The mass spectrometer was operated in positive ion mode, and the

29

analytes were quantified by using multiple reactions monitored (MRM). The transitions

monitored were m/z 199.1 to 92.1 (approximately 50 – 60 % separation) for (R)- and (S)-2-

DCEIF, m/z 199.1 to 78.1 for (R)- and (S)-3-DCEIF, m/z 205.2 to 96.2 (approximately 50 – 60

% separation) for (R)- and (S)-d6-2-DCEIF, and m/z 203.2 to 77.9 for (R)- and (S)-d4-3-DCEIF.

Although (R) and (S)-isofamide were not routinely monitored, their transitions were m/z 261.1 to

154. The declustering potential (DP), collision energy (CE), and other parameters for all the

analytes as well as the internal standard were optimized individually. The ion spray voltage was

set at 5.5 kV, and the source temperature was maintained at 500◦C. Data acquisition was

performed with MDS Sciex Analyst software (version 1.4). Calibration and analytical standard

curves were constructed by using the peak area ratios of analyte to internal standard. Test

samples and quality control samples were then interpolated from the calibration curve to obtain

the concentrations of the respective analytes.

3.3.4d Determination of GSH levels in HK-2 and LLC-PK cells

To determine GSH levels in HK-2 and LLC-PK1 cells before and after treatment with ifosfamide

with or without BSO, 1E+06 cells were plated per well in a 12-well plate. After 24 hrs, the cells

were treated with various concentrations of ifosfamide (1 – 1000 µmol/L) and (or) 250 µmol/L

BSO. Twenty-four hours after the last treatment, cells were washed with 500 µL of ice-cold PBS

(pH 7.4), after which 200 µL of 0.05 % trypsin was added. After 1 min of incubation, the cells

were washed with 500 µL of ice-cold PBS, collected, and then centrifuged at 3300 x g for 10

mins at 4◦C. A 100 µL aliquot of PBS was added to the pellet, and the sample was vortexed to

form a suspension. Fifty µL aliquots of the suspension were used for the glutathione disulfide

30

(GSSG) and GSH assay. The GSSG and GSH levels were determined with a modification of the

GT-30 GSSG/GSH colorimetric assay kit.

A 10 µL aliquot of 1-methyl-2-vinylpyridinium trifluoromethanesulfonate (M2VP) scavenger

from the GT-30 kit was added to the 50 µL cellular suspension. The sample was mixed and then

incubated for 10 mins at room temperature, after which 50 µL of ice-cold 5 % metaphosphoric

acid was added and the sample vortexed for 20 secs and then centrifuged at 1000 x g for 10 mins

at 4◦C. A 50 µL sample was transferred to a 96-well plate. A 50 µL aliquot of chromogen (5,5'-

dithiobis-2-nitrobenzoic acid (DNTB) in sodium phosphate (NaPO4) with

ethylenediaminetetraacetic acid (EDTA) and ethanol) from the GT-30 kit were added to each

sample. Absorbance was measured at 412 nm. The GSSG was quantified by using a standard

curve with a concentration range of 0.05 – 1.5 µmol/L.

For the GSH assay, 50 µL of ice-cold 5 % metaphosphoric acid was added to a 50 µL cellular

suspension. The sample was vortexed for 20 secs and then centrifuged at 1000 x g for 10 mins at

4◦C. A 50 µL sample was transferred to a 96-well plate. A 50 µL aliquot of chromogen (DNTB

in NaPO4 with EDTA and ethanol), 50 µL of enzyme (GR in NaPO4 with EDTA), and 50 µL of

NADPH from the GT-30 kit were added to each sample. Absorbance was measured at 412 nm.

GSH was quantified by using a standard curve with a concentration range of 0.1 – 3 µmol/L. The

experiment was performed in replicates of 3. Results are presented as means ± SD of 3

independent experiments.

31

3.3.5 Experimental method used to determine whether HK-2 cells are an appropriate

model to study acyclovir – induced nephrotoxicity

3.3.5a Determination of cytotoxicity in HK-2 and LLC-PK1 cells

Cells were seeded 2.5E+05 cells per well in 12-well plates (VWR International, Mississauga,

Ontario, Canada). Cells were exposed to a range of acyclovir concentrations (0 – 2000 µg/mL) in

complete growth media for 24 hrs. The acyclovir concentrations correspond to concentrations of

the drug that can be encountered by renal proximal tubular cells (Hintz et al. 1982).

Cytotoxicity was measured at the end of the 24 hrs exposure to acyclovir. The alamarBlue™

(10% (v/v) final concentration) was added to cell cultures 2.5 hrs before the end of the

incubation period with acyclovir. After incubation, cytotoxicity was assessed by measuring

fluorescence on a BioTek® Synergy HT microplate fluorometer (Fisher Scientific) at excitation

and emission wavelengths of 540 and 590 nm, respectively. The experiment was performed in

replicates of 3. Results are presented as means ± standard error (SE) of 3 independent

experiments.

3.3.6 Statistical analyses

Statistical analyses of the results obtained from the ifosfamide experiments were performed with

SigmaStat 3.1 software. Statistically significant differences between the control and treatment

groups were determined by unpaired Student's t tests. Differences were considered significant if

p<0.05.

32

Statistical analyses of the results obtained from the acyclovir experiments were performed with

SPSS 14.0 for Windows. Statistically significant differences between the control and treatment

groups were determined by one way Analysis of Variance (ANOVA) followed by 2-sided

Dunnett's post hoc tests. Differences were considered significant if p<0.05.

3.4 Results

3.4.1 CYP mRNA and protein expression in HK-2 cells

Figure 4 illustrates that HK-2 cells possess CYP 3A4 and CYP 3A7 mRNA. The RNA identity

was confirmed by sequencing and comparison with the BLAST database. Immunoblot analyses

identified a single band of CYP 3A4 at 50 kDa (Figure 5). CYP 3A5 and CYP 2B6 were not

detected. The CYP 3A4, CYP 3A5, and CYP 2B6 supersomes were used as positive controls.

Figure 4. Total ribonucleic acid (RNA) was isolated from human renal proximal tubular (HK-2)

cells and reverse transcribed. The copy DNA (cDNA) was probed for the presence/absence of

cytochrome P450 (CYP) enzymes with the primers described in the Materials and methods.

Lane 1 – CYP 3A4; Lane 2 – CYP 3A7; Lane 3 – CYP 3A5; Lane 4 – β-actin; Lane 5 – CYP

3A4 primer; Lane 6 – CYP 3A7 primer; Lane 7 – CYP 3A5 primer. The CYP mRNA

expression presented is from 1 experiment.

33

Figure 5. Western blot of human renal proximal tubular (HK-2) cells for cytochrome P450

(CYP) enzymes. Total protein from HK-2 cells and pure CYP 2B6 and CYP 3A4 and 3A5

supersomes were separated on a 10 % sodium dodecyl sulfate – polyacrylamide gel

electrophoresis (SDS-PAGE) and subsequently immunoblotted with CYP 3A4, 3A5 and 2B6

antibodies. The CYP 3A4 protein was present in the cells and had the expected molecular

weight of 49 kDa. The CYP 3A4 antibody did not cross react with the CYP 3A5 supersomes,

CYP 2B6, CYP 3A4 and CYP 3A5 supersomes (positive controls) are shown in the first lane for

each CYP (lanes 1, 4 and 7). The HK-2 total protein is shown in the other two lanes (lanes 2 and

3 for CYP3A4, lanes 5 and 6 for CYP 3A5 and lanes 8 and 9 for CYP 2B6). The western blot

presented is representative of 3 independent experiments.

34

3.4.2 Renal proximal tubular metabolism of ifsofamide by HK-2 and LLC-PK1 cells

Figure 6 shows that HK-2 cells were capable of metabolizing both ifosfamide enantiomers,

suggesting that the CYP enzymes are active. Similar levels of 2-DCEIF and 3-DCEIF

metabolites were produced with both the 100 and 1000 µmol/L treatments.

Figure 6. Metabolism of ifosfamide by human renal proximal tubular (HK-2) cells. The graph

illustrates total N-dechloroethylifosfamide (N-DCEIF) production by HK-2 cells incubated with

ifosfamide (100 or 1000 µmol/L) over a 96 hr period. Media along with the solubilized cells

were collected and 1 µL of sample was analyzed. Results are presented as the mean ±

standard deviation (SD) from 3 independent experiments.

3.4.3 Depletion of GSH and GSSG in HK-2 and LLC-PK1 cells

Direct measurement of GSH revealed that GSH levels decreased significantly from 1.25 µmol/L

in the untreated HK-2 cells (p<0.05) to 0.14 µmol/L in the BSO-depleted HK-2 cells. The GSSG

levels also decreased significantly from 0.11 to 0 µmol/L (p<0.05) in the HK-2 cells (Figure 7).

The GSH levels in untreated LLC-PK1 cells were 5-fold higher than levels in untreated HK-2

35

cells. The GSH levels decreased significantly in LLC-PK1 cells (from 5.68 to 0.76 µmol/L, p<

0.05), as did the GSSG levels (1.14 to 0 µmol/L, p<0.05).

Figure 7. Glutathione (GSH) depletion in human (HK-2) and porcine (LLC-PK1) renal proximal

tubular cells. The GSH and glutathione disulfide (GSSG) levels were determined using a

colorimetric method described in the Materials and methods section. The GSH and GSSG levels

were significantly decreased in both HK-2 and LLC-PK1 cells treated with 250 µmol/L L-

buthionine sulfoximine (BSO) for 24 hrs. Results are means ± standard deviation (SD) of 3

independent experiments. Statistically significant (p<0.05) differences from untreated HK-2

control are denoted by the symbol *. Statistically significant (p<0.05) differences from untreated

LLC-PK1 control are denoted by the symbol †.

-1

0

1

2

3

4

5

6

7

HK-2 control HK-2 BSO depletion LLC-PK1 control LLC-PK1 BSO

depletion

con

cen

tra

tio

n (

µm

ol/

L)

GSH

GSSG

36

Table 2. HK-2 versus LLC-PK1 as a model for ifosfamide – induced nephrotoxicity

HK-2 LLC-PK1

CYP 3A4 and 3A7 are present at the mRNA

level

CYP 3A and 2B are present at both the mRNA

and protein level

CYP 3A4 is detectable at the protein level

Although CYPs are homologous to human

there are some differences

Glutathione levels are significantly reduced

following treatment with BSO

Glutathione levels are significantly reduced

following treatment with BSO

IF can be metabolized to its 2-DCEIF and 3-

DCEIF metabolites

IF can be metabolized to its 2-DCEIF and 3-

DCEIF metabolites

3.4.4 Acyclovir – induced cytotoxicity in LLC-PK1 and HK-2 cells

Acyclovir induced LLC-PK1 and HK-2 cytotoxicity (as reflected by decreased cell viability) in a

concentration-dependent manner (Figure 8). Acyclovir concentrations of 500, 1000, 1500, and

2000 µg/mL induced 43 %, 62 %, 75 %, and 87 % (LLC-PK1) and 17 %, 32 %, 44 %, and 55 %

(HK-2) decreases in cell viability, respectively, compared with viability in the untreated control

cells. The decreases in cell viability were statistically significant (p<0.05).

37

Figure 8. Acyclovir – induced cytotoxicity in human (HK-2) and porcine (LLC-PK1) renal

proximal tubular cells. Cells were exposed to acyclovir in complete growth media for 24 hrs.

Cytotoxicity (measured as a function of cell viability) was measured using the fluorometric

alamarBlue™

assay. Cell viability is expressed as a percent (%) of the fluorescence of untreated

cell cultures. Results are presented as the mean ± standard error (SE) of 3 independent

experiments. Statistically significant (p<0.05) differences between untreated control and treated

groups are denoted by the symbol *.

3.5 Discussion

Ifosfamide and acyclovir are 2 essential therapeutic agents used to treat children with various

types of cancers and viral infections, respectively. Unfortunately, the use of ifosfamide or

acyclovir induces severe nephrotoxicity in some children (Brigden et al. 1982; Peterslund et al.

1988; Sawyer et al. 1988; Becker et al. 1993; Vachvanichsanong et al. 1995; Skinner 2003;

Ahmad et al. 1994; Skinner et al. 1996; Loebstein and Koren 1998; Rossi et al. 1999).

Nephrotoxicity can adversely affect a child's overall health and well-being, and it is therefore

*

*

*

*

*

*

*

*

0

20

40

60

80

100

120

0 500 1000 1500 2000

cell

via

bil

ity

(%

co

ntr

ol)

[acyclovir] (µg/mL)

HK-2 cells

LLC-PK1 cells

38

imperative to investigate the mechanisms of drug-induced nephrotoxicity to aid in the design of

safer drug therapies for children.

A major issue in elucidating the toxicological mechanisms of drugs on organ function is in

identifying an experimental model that approximates the human situation. Over the past few

decades, the porcine renal proximal tubular cell line LLC-PK1 has been extensively used for this

purpose. While the similarities between human and porcine physiology, pharmacology, and renal

function have been widely acknowledged, it would be preferable to attempt to identify a human

cell line that can more accurately reflect conditions in humans. The HK-2 cell line has emerged

as such an alternative. Yet, to date, little effort has been made to compare it with standard

experimental cell lines.

The results of this study employing 2 nephrotoxic drugs, ifosfamide and acyclovir, show that the

human-derived renal proximal tubular cell line HK-2 may be a suitable model for studying drug-

induced nephrotoxicity in humans. The RT-PCR, Western blot, and LC-MS analyses revealed

that similar to the human kidney, HK-2 cells express CYP 3A4 mRNA and protein (Figures 4

and 5). Additionally, RT-PCR analyses show that HK-2 cells express CYP 3A7 mRNA (Figure

4). Compared with mRNA expression of CYP 3A4, which of CYP 3A7 is lower in HK-2 cells.

The lower expression of CYP 3A7 in HK-2 cells may be due to the fact that in humans the

expression of CYP 3A7 is reduced shortly after birth (de Wildt et al. 1999). Although CYP 3A5

and CYP 2B6 enzymes are expressed in the kidney (Aleksa et al. 2005b), their expression was

not detected in the HK-2 cells. The lack of detection of CYP 3A5 (mRNA and protein) and CYP

2B6 (protein) expression in HK-2 cells may be due to reduced or absent expression of the

enzymes in the cell line. Aleksa et al. (2005a) found that LLC-PK1 cells were capable of the

39

stereoselective metabolism of ifosfamide. In this present study, we observed that HK-2 cells

were also able to metabolize both ifosfamide enantiomers (Figure 6). Furthermore, results from

the study revealed that, similar to the effects of BSO in LLC-PK1 cells, BSO was able to

efficiently reduce GSH and GSSG levels in HK-2 cells (Figure 7).

Similarly, the results of our cytotoxicity studies revealed that HK-2 cells are an appropriate

model for studying acyclovir – induced nephrotoxicity. Acyclovir induced a concentration-

dependent decrease in LLC-PK1 and HK-2 cell viability (Figure 8). However, in contrast to HK-

2 cells, greater decreases in viability were observed in LLC-PK1 cells. The differences observed

in acyclovir-induced toxicity between LLC-PK1 and HK-2 cells could be attributed to

interspecies differences in drug handling, and they highlight the caution that must be exercised

when nonhuman-derived cell lines are used to predict human responses to drugs and chemicals.

Our finding of acyclovir-induced toxicity in HK-2 and LLC-PK1 cells is the first experimental

evidence to suggest that acyclovir may cause direct insult to renal proximal tubular cells, and

hence it supports clinical pathology data of severe renal damage without evidence of crystalluria

in patients on acyclovir therapy (Becker et al. 1993; Ahmad et al. 1994; Vomiero et al. 2002).

Taken together, the findings from this study indicate that although both LLC-PK1 and HK-2

cells may be reasonable models for in vitro nephrotoxicity studies, the existence of interspecies

differences in drug pharmacology and toxicology may make HK-2 cells the more acceptable cell

culture model to use in in vitro studies aimed at elucidating the nephrotoxicity of drugs in

humans. However, this current working hypothesis remains to be further tested with other

nephrotoxic drugs.

40

3.6 Acknowledgements

The research was funded by a grant from the Canadian Institutes of Health Research (CIHR).

3.7 Statement of significance

To the best of our knowledge, this study is the first to provide in vitro experimental evidence

which supports clinical evidence that acyclovir induces direct insult to renal tubular cells.

Furthermore, the results illustrate that the human renal proximal tubular cell line, HK-2 cells are

an appropriate in vitro model to study drug – induced nephrotoxicity without the need for

concern regarding the interspecies differences that exists in the pharmacological and

toxicological disposition of drugs.

41

3.8 References

Ahmad, T., Simmonds, M., McIver, A.G., and McGraw, M.E. 1994. Reversible renal failure in

renal transplant patients receiving oral acyclovir prophylaxis. Pediatr. Nephrol. 8: 489-491.

Aleksa, K., Halachmi, N., Ito, S., and Koren, G. 2005a. A tubule cell model for ifosfamide

nephrotoxicity. Can. J. Physiol. Pharmacol. 83: 499-508.

Aleksa, K., Matsell, D., Krausz, K., Gelboin, H., Ito, S., and Koren, G. 2005b. Cytochrome P450

3A and 2B6 in the developing kidney: implications for ifosfamide nephrotoxicity. Pediatr.

Nephrol. 20: 872-885.

Becker, B.N., Fall, P., Hall, C., Milam, D., Leonard, J., Glick, A., and Schulman, G. 1993.

Rapidly progressive acute renal failure due to acyclovir: case report and review of the literature.

Am. J. Kidney Dis. 22: 611-615.

Brade, W.P., Herdrich, K., and Varini, M. 1985. Ifosfamide: pharmacology, safety and

therapeutic potential. Cancer Treat. Rev. 12: 1-47.

Brigden, D., Rosling, A.E., and Woods, N.C. 1982. Renal function after acyclovir intravenous

injection. Am. J. Med. 73: 182-185.

Carli, M., Passone, E., Perilongo, G., and Bisogno, G. 2003. Ifosfamide in pediatric solid tumors.

Oncology, 65: 99-104.

Chang, T.K.H., Weber, G.F., Crespi, C.L., and Waxman, D.J. 1993. Differential activation of

cyclophosphamide and ifosphamide by cytochromes P-450 2B and 3A in human liver

microsomes. Cancer Res. 53: 5629-5637.

Chen, C.S., Jounaidi, Y., and Waxman, D.J. 2005. Enantioselective metabolism and cytotoxicity

of R-ifosfamide and S-ifosfamide by tumor cell-expressed cytochromes P450. Drug Metab.

Dispos. 33: 1261-1267.

de Wildt, S.N., Kearns, G.L., Leeder, J.S., and van den Anker, J.N. 1999. Cytochrome P450 3A:

ontogeny and drug disposition. Clin. Pharmacokinet. 37: 485-505.

Detrisac, C.J., Sens, M.A., Garvin, A.J., Spicer, S.S., and Sens, D.A. 1984. Tissue culture of

human kidney epithelial cells of proximal tubule origin. Kidney Int. 25: 383-390.

Eaton, D.L., and Klaassen, C.D. 2001. Principles of toxicology. In Casarett and Doull's

toxicology: the basic science of poisons. Edited by Klaassen, C.D. McGraw-Hill Companies Inc.,

New York. pp. 11-34.

Elion, G.B. 1983. The biochemistry and mechanism of action of acyclovir. J. Antimicrob.

Chemother. 12: 9-17.

42

Giannone, J.V., Li, W., Probst, M., and Okey, A.B. 1998. Prolonged depletion of AH receptor

without alteration of receptor mRNA levels after treatment of cells in culture with 2,3,7,8-

tetrachlorodibenzo-p-dioxin. Biochem. Pharmacol. 55: 489-497.

Hintz, M., Connor, J.D., Spector, S.A., Blum, M.R., Keeney, R.E., and Yeager, A.S. 1982.

Neonatal acyclovir pharmacokinetics in patients with herpes virus infections. Am. J. Med. 73:

210-214.

Jover, R., Bort, R., Gomez-Lechon, M.J., and Castell, J.V. 2001. Cytochrome P450 regulation by

hepatocyte nuclear factor 4 in human hepatocytes: a study using adenovirus-mediated antisense

targeting. Hepatology, 33: 668-675.

Keeney, R.E., Kirk, L.E., and Bridgen, D. 1982. Acyclovir tolerance in humans. Am. J. Med. 73:

176-181.

Loebstein, R., and Koren, G. 1998. Ifosfamide-induced nephrotoxicity in children: critical

review of predictive risk factors. Pediatrics, 101: E8.

Lyon, A.W., Mansoor, A., and Trotter, M.J. 2002. Urinary gems: acyclovir crystalluria. Arch.

Pathol. Lab. Med. 126: 753-754.

Mason, W.J., and Nickols, H.H. 2008. Images in clinical medicine. Crystalluria from acyclovir

use. N. Engl. J. Med. 358: e14.

McCune, J.S., Risler, L.J., Phillips, B.R., Thummel, K.E., Blough, D., and Shen, D.D. 2005.

Contribution of CYP3A5 to hepatic and renal ifosfamide N-dechloroethylation. Drug Metab.

Dispos. 33: 1074-1081.

Perantoni, A., and Berman, J.J. 1979. Properties of Wilms' tumor line (TuWi) and pig kidney

line (LLC-PK1) typical of normal kidney tubular epithelium. In Vitro. 15: 446-454.

Peterslund, N.A., Larsen, M.L., and Mygind, H. 1988. Acyclovir crystalluria. Scand. J. Infect.

Dis. 20: 225-228.

Richards, D.M., Carmine, A.A., Brogden, R.N., Heel, R.C., Speight, T.M., and Avery, G.S.

1983. Acyclovir: a review of its pharmacodynamic properties and therapeutic efficacy. Drugs.

26: 378-438.

Riddick, D.S. 1998. Drug biotransformation. In Principles of Medical Pharmacology. Edited by

Kalant, H., and Roschlau, W.H.E. Oxford University Press, New York. pp. 38-54.

Rossi, R., Pleyer, J., Schafers, P., Kuhn, N., Kleta, R., Deufel, T., and Jurgens, H. 1999.

Development of ifosfamide-induced nephrotoxicity: prospective follow-up in 75 patients. Med.

Pediatr. Oncol. 32: 177-182.

43

Roy, P., Tretyakov, O., Wright, J., and Waxman, D.J. 1999. Stereoselective metabolism of

ifosfamide by human P-450s 3A4 and 2B6. Favorable metabolic properties of R-enantiomer.

Drug Me tab. Dispos. 27: 1309-1318.

Ryan, M.J., Johnson, G., Kirk, J., Fuerstenberg, S.M., Zager, R.A., and Torok-Storb, B. 1994.

HK-2: an immortalized proximal tubule epithelial cell line from normal adult human kidney.

Kidney Int. 45: 48-57.

Sawyer, M.H., Webb, D.E., Balow, J.E., and Straus, S.E. 1988. Acyclovir-induced renal failure.

Clinical course and histology. Am. J. Med. 84: 1067-1071.

Schreiber, R., Wolpin, J., and Koren, G. 2008. Determinants of aciclovir-induced nephrotoxicity

in children. Paediatr. Drugs. 10: 135-139.

Skinner, R. 2003. Chronic ifosfamide nephrotoxicity in children. Med. Pediatr. Oncol. 41: 190-

197.

Skinner, R., Pearson, A.D., English, M.W., Price, L., Wyllie, R.A., Coulthard, M.G., and Craft,

A.W. 1996. Risk factors for ifosfamide nephrotoxicity in children. Lancet. 348: 578-580.

Sladek, N.E. 1988. Metabolism of oxazaphosphorines. Pharmacol. Ther. 37: 301-355.

Vachvanichsanong, P., Patamasucon, P., Malagon, M., and Moore, E.S. 1995. Acute renal failure

in a child associated with acyclovir. Pediatr. Nephrol. 9(3): 346-347.

Vomiero, G., Carpenter, B., Robb, I., and Filler, G. 2002. Combination of ceftriaxone and

acyclovir--an underestimated nephrotoxic potential? Pediatr. Nephrol. 17: 633-637.

Wagstaff, A.J., Faulds, D., and Goa, K.L. 1994. Aciclovir: a reappraisal of its antiviral activity,

pharmacokinetic properties and therapeutic efficacy. Drugs, 47: 153-205.

Walker, D., Flinois, J.P., Monkman, S.C., Beloc, C., Boddy, A.V., Cholerton, S., et al. 1994.

Identification of the major human hepatic cytochrome P450 involved in activation and N-

dechloroethylation of ifosfamide. Biochem. Pharmacol. 47: 1157-1163.

Xu, H., Rajesan, R., Harper, P., Kim, R.B., Lonnerdal, B., Yang, M., et al. 2005. Induction of

cytochrome P450 1A by cow milk-based formula: a comparative study between human milk and

formula. Br. J. Pharmacol. 146: 296-305.

Zucco, F., De Angelis, I., Testai, E., and Stammati, A. 2004. Toxicology investigations with cell

culture systems: 20 years after. Toxicol. In Vitro, 18: 153-163.

44

Chapter 4

Acyclovir – induced nephrotoxicity: the role of the acyclovir

aldehyde metabolite

Patrina Gunness,a,b

Katarina Aleksa,a,c

John Bend,d Gideon Koren

a,b

aDivision of clinical Pharmacology and Toxicology, The Hospital for Sick Children, 555

University Avenue, Toronto, ON, M5G 1X8, Canada

bGraduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University

of Toronto, ON, M5S 3M2, Canada

cSchool of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, Ontario,

N2L 3G1, Canada

dDepartment of Pathology, Schulich School of Medicine and Dentistry, DSB4044, The

University of Western Ontario, London, Ontario, N6A 3K7, Canada

This article has been accepted for publication: Gunness, P., Aleksa, K., Bend, J., and Koren,

G. 2011. Acyclovir – induced nephrotoxicity; the role of the acyclovir aldehyde metabolite.

Transl Res. [In press]. This article will be originally published by Elsevier.

[PG performed all experiments and prepared the manuscript for submission]

45

4.1 Abstract

For decades, acyclovir – induced nephrotoxicity, was believed to be secondary to crystalluria.

Clinical evidence of nephrotoxicity in the absence of crystalluria suggests that acyclovir induces

direct insult to renal tubular cells. We postulated that acyclovir is metabolized by ADH enzyme

to acyclovir aldehyde, which is further metabolized by the ALDH2 enzyme to CMMG. We

hypothesized that acyclovir aldehyde plays a role in acyclovir – induced nephrotoxicity. The

HK-2 cells were used as our in vitro model. Western blot and enzymes activities assays were

performed to determine whether the HK-2 cells express ADH and ALDH2 isozymes,

respectively. Cytotoxicity (measured as a function of cell viability) assays were conducted to

determine; (1) whether the acyclovir aldehyde plays a role in acyclovir – induced nephrotoxicity

and (2) whether CMMG induces cell death. A colorimetric assay was performed to determine

whether acyclovir was metabolized to an aldehyde, in vitro. Our results illustrated that: (A) HK-

2 cells express ADH and ALDH2 isozymes, (B) 4-methylpyrazole rendered significant

protection against cell death, (C) CMMG does not induce cell death and (D) acyclovir was

metabolized to an aldehyde in tubular cells. These data indicate that acyclovir aldehyde is

produced in HK-2 cells and that inhibition of its production by 4-methylpyrazole offers

significant protection from cell death, in vitro; suggesting that acyclovir aldehyde may cause the

direct renal tubular insult associated with acyclovir.

4.2 Introduction

Acyclovir, an acyclic nucleoside (Brigden and Whiteman 1985; Elion 1983) is commonly used

for the treatment of viral infections (Bianchetti et al. 1991; Brigden et al. 1982; Fletcher et al.

1989; Hintz et al. 1982; Keeney et al. 1982; Vachvanisanong et al. 1995). The herpes simplex

and varicella zoster viruses are among the viruses which acyclovir is used clinically (Biachetti et

46

al. 1991; Brigden et al. 1982; Hintz et al. 1982). Acyclovir is generally well tolerated (Keeney et

al. 1982), however, severe nephrotoxicity has been shown to occur in some children (Ahmad et

al. 1994; Genc et al. 2010; Schreiber et al. 2008).

It has been widely believed that acyclovir – induced nephrotoxicity is caused by crystalluria

(Genc et al. 2010; Lyon et al. 2002; Mason et al. 2008; Peterslund et al. 1998; Potter and Krill

1986; Sawyer et al. 1988). However, clinical evidence of nephrotoxicity in the absence of

crystal formation (Ahmad et al. 1994; Vomiero et al. 2002) has suggested that acyclovir may

induce direct insult to renal tubular cells. For example, renal biopsies have demonstrated that

acyclovir exposure was associated with flattened, vacuolated (Ahmad et al. 1994; Vomiero et al.

2002), bulging epithelial cells (Vomiero et al. 2002) and no evidence of crystals. Recently, we

provided the first experimental evidence that acyclovir can induce direct damage to renal tubular

cells (Gunness et al. 2010).

Biotransformation of acyclovir to an active metabolite is not required for its anti-viral activity or

for its excretion (Elion 1983). In humans, acyclovir undergoes minimal metabolism, such that a

given intravenous dose of acyclovir is eliminated mainly unchanged (62 – 91%) in the urine (de

Miranda et al. 1982a). The predominant and pharmacologically inactive (de Miranda et al.

1982a) metabolite of acyclovir is CMMG, while 8-hydroxy-9-(2-hydroxyethoxymethyl)guanine

is considered the minor metabolite (de Miranda et al. 1982a). High performance liquid

chromatography analyses of urine show that in humans, the CMMG metabolite accounts for

approximately 9 to 14 %, and the 8-hydroxy-9-(2-hydroxyethoxymethyl)guanine metabolite

accounts for less than 0.2 % of a given dose of acyclovir, respectively.

47

The present study focuses on the metabolic pathway of acyclovir to CMMG. It has been

postulated that acyclovir is metabolized by the ADH enzyme to acyclovir aldehyde, and

acyclovir aldehyde is subsequently metabolized by the ALDH enzyme to CMMG (de Miranda

and Burnette 1994; de Miranda and Good 1982; de Miranda and Good 1992).

Aldehydes are reactive, toxic chemicals that are often produced endogenously as intermediate

drug metabolites and have been suggested to mediate several drugs – induced toxicities,

including hepatotoxicity and nephrotoxicity (O’Brien et al. 2005). For example, the

chloroacetaldehyde metabolite of ifosfamide, produced from the oxidation of the

chemotherapeutic agent (Walker et al. 1994) has been shown to cause nephrotoxicity (Dubourg

et al. 2001). We set up to investigate whether the acyclovir aldehyde metabolite may be the

source of the direct renal tubular damage associated with the use of acyclovir. This hypothesis

has not been previously investigated.

In addition to the kidney’s well-defined physiological functions, including the regulation of

osmolarity, maintenance of electrolyte balance, synthesis of hormones and removal of waste

substances (Silverthorn 1998), the kidney is also actively involved in drug metabolism (Anders

1980; Lohr et al. 1998). For instance, Aleksa and colleagues illustrated that ifosfamide is

metabolized in human and porcine kidneys, in vitro (Aleksa et al. 2006). While, Diamond and

Quebbemann showed that in vivo, in chickens, p-nitrophenol is renally metabolized to its sulfate

and glucuronide metabolites (Diamond and Quebbemann 1981). It was previously elaborated

that some of the body load of acyclovir is metabolized by the ADH and ALDH enzymes (de

Miranda and Burnette 1994; de Miranda and Good 1982; de Miranda and Good 1992). The

human kidney expresses both ADH and ALDH enzymes (Engeland and Maret 1993; Harada et

48

al. 1980; Nishimura and Naito 2006). Therefore, the renal proximal tubular cells may have the

machinery to locally metabolize acyclovir to its aldehyde metabolite.

Several ADH (Engeland and Maret 1993) and ALDH (Harada et al. 1980) isozymes exists in

humans. Currently, it is unknown which ADH and ALDH isozyme(s) may be involved in the

metabolism of acyclovir in humans. In our studies, we focused on the human class I ADH

enzymes because they are referred to as prototypical ADHs (Estonius et al. 1996) and they are

highly expressed in the human kidney (Engeland and Maret 1993). With respect to the ALDH

enzyme, we postulated that the ALDH2 enzyme may be specifically involved in the metabolism

of acyclovir. The ALDH2 enzyme is highly expressed in the human kidney (Harada et al. 1980)

and moreover, studies have illustrated that polymorphism of the ALDH2 enzyme prolonged the

elimination half life of acyclovir in humans (Hara et al. 2008). The results from the study by

Hara and colleagues (Hara et al. 2008) are the first to provide evidence for the potential role of

ALDH2 in the metabolism of acyclovir.

The objectives of this study were to examine whether acyclovir – induced nephrotoxicity is

secondary to tubular production of acyclovir aldehyde.

4.3 Materials and methods

4.3.1 Cell culture

The HK-2 cell line was employed as our in vitro model. The cells were maintained according to

ATCC guidelines. Briefly, cells were cultured in Keratinocyte-SFM supplemented with 5 ng/mL

human recombinant epidermal growth factor 1-53 and 0.05 mg/mL bovine pituitary extract.

Cells were maintained at 37°C in a sterile, humidified atmosphere of 5 % CO2 and 95 % O2. All

experiments were conducted on cell monolayers that were approximately 80 – 85 % confluent.

49

4.3.2 Protein expression and enzymes activities of class I ADH and ALDH2 isozymes in

HK-2 cells

The protein expression and activities of class I ADH and ALDH2 isozymes in HK-2 cells have

not been previously investigated. The determination of the presence of class I ADH and ALDH2

functional isozymes in HK-2 cells was imperative to our study because the cells were used as our

model to determine whether acyclovir aldehyde; which is produced via the oxidation of acyclovir

by the ADH enzyme, and further metabolized by the ALDH enzyme to CMMG, is the source of

the direct renal tubular insult associated with acyclovir.

Qualitative western blot analyses were conducted to determine whether the HK-2 cells expressed

the class I ADH and ALDH2 isozymes. The enzymes activities assays were conducted to

determine whether HK-2 cells expressed functional class I ADH and ALDH2 isozymes.

Western blots analyses were conducted in duplicates. Enzymes activities assays were conducted

in triplicates. Total protein for all assays was quantified using the Bradford reagent (Sigma-

Aldrich Canada Ltd., Oakville, Ontario, Canada). The absorbance was measured at 595 nm

using a BioTek® Synergy HT microplate reader (Fisher Scientific). The lysate from human liver

tissue obtained from a deceased adult male was used as a positive control for western blot assays.

Human liver is known to express high levels of class I ADH (Engeland and Maret 1993) and

ALDH2 isozymes (Harada et al. 1980).

In order to extrapolate the biological significance of the results obtained from HK-2 cells to the

human kidney, the lysate from the human kidney of a deceased adult male was used to determine

functional protein expression of the class I ADH and ALDH2 enzymes. The human liver and

kidney tissues were obtained from the Co-operative Human Tissue network, University of

50

Pennsylvania Medical Center, USA. The ADH protein expression in the human kidney was

compared to the level of expression in the HK-2 cells.

4.3.2a Cytosol and mitochondria protein fraction for western blot assays

HK-2 cells

The media from HK-2 cell monolayers was removed and the cells were washed (2X) with ice-

cold PBS solution. Cell monolayers were scraped in a modified lysis buffer ([50 mmol/L

tris(hydroxymethyl)aminomethane hydrochloride (Tris-HCL, pH7.4), 1 % (v/v) nonyl

phenoxypolyethoxylethanol (NP-40), 0.25 % (w/v) sodium deoxycholate, 150 mmol/L sodium

chloride (NaCl), 1 mmol/L EDTA, 1 mmol/L phenylmethylsulfonyl fluoride (PMSF), 1 µg/mL

aprotonin, 1 µg/mL leupeptin, 1 µg/mL pepstatin] (Millipore 2007). The cell homogenate was

centrifuged at 600 x g for 20 mins at 4◦C. The pellet was discarded and the supernatant was

centrifuged at 100 000 x g for 2 hrs at 4◦C. The supernatant (cytosol fraction) was stored at -

80◦C until analyses.

To obtain the mitochondria fraction from HK-2 cells, cell monolayers were scraped in the

modified lysis buffer as described above. The cell homogenate was centrifuged at 600 x g for 20

mins at 4◦C. The pellet was discarded and the supernatant was centrifuged at 15 000 x g for 10

mins at 4◦C. The supernatant was discarded and the pellet was re-suspended in lysis buffer. The

homogenate was briefly sonicated and subsequently centrifuged at 15 000 x g for 5 mins at 4◦C.

The supernatant (mitochondria fraction) was stored at -80◦C until analyses.

51

Human liver and kidney tissue

The isolation of the mitochondrial and cytosolic fractions from human liver and kidney tissue

was performed using identical isolation procedures. Human liver or kidney tissues (2 grams)

were washed twice with ice-cold PBS. The tissues were homogenized using a polytron

homogenizer (Brinkmann Instruments Canada, Mississauga, Ontario, Canada). The tissues were

homogenized in the modified lysis buffer described above for HK-2 cells. The homogenates

were centrifuged at 600 x g for 20 mins at 4◦C. The pellets were discarded and the supernatants

were centrifuged at 15 000 x g for 10 mins at 4◦C. The pellets (Fraction A; containing

mitochondria protein) and supernatants (Fraction B; containing cytosol protein) were processed

as follows. Fraction A was re-suspended in lysis buffer and fraction B was transferred to a clean

centrifuge tube. Fraction A was briefly sonicated and subsequently centrifuged at 15 000 x g for

5 mins at 4◦C. The pellets were discarded and the resultant supernatants (mitochondria fraction)

were stored at -80◦C until analyses. Fraction B was centrifuged at 100 000 x g for 2 hrs at 4

◦C.

The pellets were discarded and the resultant supernatants (cytosol fraction) were stored at -80◦C

until analyses.

4.3.3 Western blot assays

4.3.3a ADH protein expression

For electrophoresis samples, total cytosol protein was mixed with 2X Laemmli buffer (Laemmli

1970). Total human liver (1 µg), kidney (10 µg) or HK-2 (30 µg) cytosol protein were resolved

on a 12 % SDS-PAGE. Resolved proteins were transferred unto Hybond™

- P PVDF

membranes (GE Healthcare Canada Inc., Mississauga, Ontario, Canada) at 100 V for 1 hr in

transfer buffer ([25 mmol/L Tris, 192 mmol/L glycine, 20 % (v/v) methanol, pH 8.3] (Towbin et

52

al. 1979). Blots were blocked in 5 % (w/v) skim milk overnight at 4◦C. Blots were then washed

in 5 % milk and subsequently incubated with rabbit polyclonal ADH antibody (sc-22750, Santa

Cruz Biotechnology, Inc., Santa Cruz, California, USA) in 5 % skim milk overnight at 4◦C. The

primary antibody was diluted 1:500 for use. Following the incubation, the primary antibody was

removed and blots were washed in 5 % skim milk. Blots were subsequently incubated with

donkey anti-rabbit IgG-HRP antibody (sc-2313, Santa Cruz Biotechnology, Inc.) in 5 % skim

milk for 2 hrs at room temperature. The secondary antibody was diluted 1:5000 for use. The

secondary antibody was removed and the blots were washed sequentially in 5 % skim milk, PBS

with Tween® 20 detergent (PBST) and PBS solutions. Blots were developed using Western

Lightning® Plus – ECL (PerkinElmer, Woodbridge, Ontario, Canada). Blots were exposed to

Kodak™

BioMax Light Film (PerkinElmer).

4.3.3b ALDH2 protein expression

For ALDH2 protein expression, western blot analyses were performed as described above.

Total human liver (30 µg), kidney (40 µg) or HK-2 (100 µg) mitochondria protein were resolved

on a 12 % SDS-PAGE. The primary antibody used was goat polyclonal ALDH2 antibody (sc-

48838, Santa Cruz Biotechnology, Inc.). The primary ALDH2 antibody was diluted 1:200 for

use. The secondary antibody used was donkey anti-goat IgG-HRP (sc-2354, Santa Cruz

Biotechnology, Inc.). The secondary antibody was diluted 1:5000 for use.

4.3.4 Enzymes activities assays

4.3.4a Whole cell lysate for enzymes activities assays

The HK-2, as well as human kidney whole cell lysates were used for enzymes activities assays.

53

HK-2 cells

Briefly, the media from HK-2 cell monolayers was removed and the cells were washed twice

with ice-cold PBS. Cell monolayers were scraped in a modified lysis buffer ([50 mmol/L Tris-

HCL (pH7.4), 1 % (v/v) NP-40, 150 mmol/L NaCl] (Millipore 2007). The cell homogenate was

centrifuged at 600 x g for 20 mins at 4◦C. The pellet was discarded and the resultant supernatant

(whole cell lysate) was stored at -80◦C until analyses.

Human kidney

Human kidney tissue (2 grams) was washed twice with ice-cold PBS. The tissue was

homogenized using a polytron homogenizer (Brinkmann Instruments Canada). The tissue was

homogenized in the modified lysis buffer and subsequently processed as outlined above for the

HK-2 cells.

4.3.4b ADH and ALDH enzymes activities assays

The enzymes activities assays were performed as previously described (Clemens et al. 1995),

with modification. The ADH and ALDH enzymes use nicotinamide adenine dinucleotide

(NAD+) as a co-factor, such that during enzymatic activity, NAD

+ is reduced to NADH (Pawan

1972). The NADH molecule absorbs lights at 340 nm (Walker 1992). The optical density at 340

nm can be measured using a spectrophotometer (Walker 1992).

ADH enzyme activity

Briefly, 200 µg of total protein was added to 0.5 mol/L Tris-HCL (pH 7.4), 3 mmol/L NAD+ in

the presence or absence of 10 mmol/L ethanol. The reaction components were incubated at 37◦C

for 10 mins prior to addition of ethanol to the mixture. The final reaction volume was 2 mL.

54

The reaction mixture was incubated for 4 hrs, and then the optical density at 340 nm was

measured using BioTek®

Synergy HT microplate reader (Fisher Scientific).

ALDH enzyme activity

The ALDH enzyme activity assays were as described above for the determination of ADH

enzyme activity. One mM propionaldehyde was used as the substrate for ALDH2.

4.3.5 Cell viability

Cytotoxicity (measured as a function of cell viability) assays were performed to determine

whether the acyclovir aldehyde may be the source of the direct renal tubular injury associated

with the use of the parent drug. As previously described, acyclovir is metabolized to acyclovir

aldehyde by the ADH enzyme and acyclovir aldehyde is subsequently metabolized by the ALDH

enzyme to CMMG (de Miranda and Burnette 1994; de Miranda and Good 1982; de Miranda and

Good 1992). Therefore, if acyclovir aldehyde is the source of tubular damage; inhibition of the

ADH enzyme by 4-methylpyrazole should alleviate toxicity. We used 4-methylpyrazole to

inhibit the class I ADH isozymes. The 4-methylpyrazole inhibitor has been used in in vitro

studies to inhibit the ADH enzymes (Gyamfi and Wan 2006). Furthermore, and as reviewed by

Crabb et al., compared to other classes of ADH enzymes, class I ADH isozymes are highly

sensitive to pyrazoles inhibition (Crabb et al. 2004). Importantly, 4-methyplyrazole has been

shown to inhibit the metabolism of acyclovir to CMMG (de Miranda and Good 1982).

Cell viability assays were performed in 12-well plates. Cell viability was assessed using the

fluorescent alamarBlue® assay. Briefly, 2.5 hrs prior to the end of the 24 hour incubation period,

alamarBlue® reagent was added to each well. The final concentration of alamarBlue

® reagent in

each well was 10 % (v/v). Cell viability was measured using BioTek®

Synergy HT microplate

55

reader at excitation and emission wavelengths of 540 and 590 nm, respectively. Cell viability

assays were performed in replicates of 9.

4.3.5a Co-exposure to 4-methylpyrazole

Cells were seeded (1.25E+05 cells/well) in 12-well plates. Exposure to 4-methylpyrazole

(Sigma-Aldrich Canada Ltd.) was performed similar to the regimen outlined by Gyamfi and Wan

(Gyamfi and Wan 2006). Initially, cell monolayers were incubated with 4-methylpyrazole (500

µmol/L) for 1 hr. Following the incubation period, the media was removed, and cell monolayers

were incubated with acyclovir (0 – 2000 µg/mL) in the presence or absence of 4-methylpyrazole

for 24 hrs. The concentrations of acyclovir used in our study are compatible to the

concentrations of the anti-viral drug encountered by the human kidney (Hintz et al. 1982). Cell

viability was assessed at the end of the 24 hr incubation period as described above.

4.3.6 Determination of aldehyde production

In order to determine if an aldehyde was produced in tubular cells exposed to acyclovir, a

colorimetric aldehyde detection assay was performed using the Purpald® reagent (Sigma-Aldrich

Canada Ltd.). The Purpald® reagent reacts with aldehydes to form colorless adducts, which

must be oxidized to form chromogens (Quesenberry and Lee 1996). The chromogens absorb

light at 550 nm (Quesenberry and Lee 1996). The absorbance at 550 nm can be measured using

a spectrophotometer (Quesenberry and Lee 1996).

The assay was performed similar to the methods described by Quesenberry and Lee

(Quesenberry and Lee 1996). Briefly, cells were seeded (2.5E+05 cells/dish) in petri dishes

(VWR International). At the desired confluence, cell monolayers were incubated with acyclovir

(0 or 2000 µg/mL) for 3 hrs. At the end of the incubation period, cell monolayers were washed

56

(2X) with ice-cold PBS. The cell monolayers were then scraped in ice-cold PBS and briefly

sonicated. The homogenate was centrifuged at 15 000 x g for 5 mins at 4◦C. The pellet was

discarded and the supernatant was processed as follows. One mL of 33 mmol/L Purpald®

reagent (Sigma-Aldrich Canada Ltd.) was added to 1 mL of the supernatant. The mixture was

incubated at room temperature for 2 hrs. One mL of 34 mmol/L sodium periodate (Sigma-

Aldrich Canada Ltd.) was subsequently added and the mixture was incubated at room

temperature for an additional 2 hrs. The optical density was subsequently measured at 550 nm

using a Shimadzu UV160U spectrophotometer (Mandel Scientific Co. Ltd., Guelph, Ontario,

Canada). The assay was not conducted in replicates.

4.3.7 Comparison of the ADH protein expression between HK-2 cells and human kidney

tissue

In order to compare the ADH protein expression between HK-2 cells and the human kidney

tissue, it was necessary to quantify ADH protein expression obtained from the western blots

analyses. Protein expression was quantified using the Image Processing and Analysis in Java

(ImageJ) software. Since, different amounts of total cytosol protein from HK-2 (30 µg) and

human kidney (10 µg) were resolved on separate SDS-PAGE analyses, it was assumed that total

amounts of protein that was resolved was linear to the intensity of the protein band.

Additionally, the expression was not normalized to a housekeeping gene because protein

expression between two different biological matrices, an immortalized cell line and human

tissue, were compared. The integrated densities of the ADH protein bands obtained from the

ImageJ analyses of western blots were used to compare the protein expression between HK-2

cells and human kidney. Since, 3X more total protein from the HK-2 cell cytosol was used in

57

SDS-PAGE analyses, compared to human kidney; the integrated density obtained from human

kidney was multiplied by 3 before being compared to that of HK-2. The quantification was

performed using the 2 western blots that were each obtained from analyses of ADH protein

expression in HK-2 and human kidney.

4.3.8 Statistical analyses

Statistical analyses of the data were performed using IBM® SPSS

® Statistics version 19 software.

The data obtained from the ADH and ALDH enzymes activities assays were analyzed using

paired t-tests. One way ANOVA followed by Tukey’s honestly significant difference (HSD)

post hoc tests were conducted to test the statistical significance of the data obtained from cell

viability assays that were performed to determine whether acyclovir aldehyde plays a role in

acyclovir – induced nephrotoxicity. The ANOVA followed by Dunnett’s post hoc tests were

conducted to assess the statistical significance of the data from cell viability assays that were

conducted to determine whether CMMG induced HK-2 cell death.

4.4 Results

4.4.1 Class I ADH and ALDH2 protein expression

Figures 9A and 9B illustrate class I ADH and ALDH2 protein expression in HK-2 cells,

respectively. The ADH (Figure 9A) and ALDH2 (Figure 9B) proteins are expressed at low

levels in HK-2 cells. Figures 10A and 10B confirm the protein expression of the class I ADH

and ALDH2 enzymes in human kidney, respectively. The proteins are expressed at moderate

levels in human kidney.

58

A.

B.

Figure 9. The (A) alcohol dehydrogenase (ADH) and (B) aldehyde dehydrogenase (ALDH)

protein expression in human renal proximal tubular (HK-2) cells. Western blot analyses were

performed to determine whether HK-2 cells express class I ADH and ALDH2 isozymes. Total

cytosol [liver:1 µg, HK-2 cells: 30 µg] or mitochondrial [liver:30 µg, HK-2 cells: 100 µg]

protein were resolved on a 12 % sodium dodecyl sulfate – polyacrylamide gel electrophoresis

(SDS-PAGE) for analyses if ADH or ALDH protein expression, respectively. The ADH blots

were incubated with rabbit polyclonal ADH antibody (sc-22750) diluted 1:500 in 5 % (w/v) skim

milk. The secondary antibody used was donkey anti-rabbit immunoglobulin G – horseradish

peroxidase (IgG-HRP) antibody (sc-2313) diluted 1:5000 in 5 % skim milk. The ALDH blots

were incubated with goat polyclonal ALDH2 antibody (sc-48838) diluted 1:200 in 5 % skim

milk. The secondary antibody used was donkey anti-goat immunoglobulin G – horseradish

peroxidase (IgG-HRP) antibody (sc-2354) diluted 1:5000 in 5 % skim milk. Western blot

analyses were performed in duplicates. The blots illustrated are representative blots.

59

A.

B.

Figure 10. The (A) alcohol dehydrogenase (ADH) and (B) aldehyde dehydrogenase (ALDH)

protein expression in human kidney. Western blot analyses were performed to determine

whether human kidney expresses class I ADH and ALDH2 isozymes. Total cytosol [liver:1 µg,

kidney: 10 µg] or mitochondrial [liver:30 µg, kidney: 40 µg] protein were resolved on a 12 %

sodium dodecyl sulfate – polyacrylamide gel electrophoresis (SDS-PAGE) for analyses if ADH

or ALDH protein expression, respectively. The ADH blots were incubated with rabbit

polyclonal ADH antibody (sc-22750) diluted 1:500 in 5 % (w/v) skim milk. The secondary

antibody used was donkey anti-rabbit immunoglobulin G – horseradish peroxidase (IgG-HRP)

antibody (sc-2313) diluted 1:5000 in 5 % skim milk. The ALDH blots were incubated with goat

polyclonal ALDH2 antibody (sc-48838) diluted 1:200 in 5 % skim milk. The secondary

antibody used was donkey anti-goat immunoglobulin G – horseradish peroxidase (IgG-HRP)

antibody (sc-2354) diluted 1:5000 in 5 % skim milk. Western blot analyses were performed in

duplicates. The blots illustrated are representative blots.

60

4.4.2 ADH and ALDH enzyme activity

Figures 11A and 11B illustrate ADH and ALDH enzyme activities in HK-2 cells, respectively.

Figure 11A shows that in the presence of the ADH substrate, ethanol, there was a significant

(p<0.05) increase in the optical density at 340 nm. Similarly, in the presence of the ALDH

substrate, propionaldehyde, there was a significant (p<0.05) increase in the optical density at 340

nm (Figure 11B).

The ADH and ALDH enzymes activities in human kidney are illustrated in Figures 12A and

12B, respectively. In the presence of the ADH substrate, ethanol, there appeared to be an

increase in the optical density at 340 nm (Figure 12A). Similarly, Figure 12B shows that in the

presence of the ALDH substrate, propionaldehyde, there appeared to be an increase in the optical

density at 340 nm.

61

A.

B.

Figure 11. The (A) alcohol dehydrogenase (ADH) and (B) aldehyde dehydrogenase enzyme

activity in human renal proximal tubular (HK-2) cells. Total (200 µg) protein from HK-2 cells

was added to 0.5 mol/L tris(hydroxymethyl)aminomethane hydrochloride (Tris-HCL, pH 7.4), 3

mmol/L nicotinamide adenine dinucleotide (NAD+) in the presence or absence of the substrates,

10 mmol/L ethanol (determination of ADH activity) or 1 mmol/L propioaldehyde (determination

of ALDH activity). The reaction mixture was incubated for 4 hrs, and then the optical density at

340 nm was measured using BioTek® Synergy HT microplate reader. The enzyme activity

assays were performed in triplicates. The results are presented as the mean ± standard error

(SE). The statistically significant (p<0.05) difference between the optical density measured at

340 nm in the presence or absence of ethanol is denoted by the symbol*.

*

0.16

0.165

0.17

0.175

0.18

0 10

A340

(arb

itra

ry a

bso

rba

nce

un

its)

[ethanol] (mmol/L)

*

0.16

0.165

0.17

0.175

0.18

0 1

A340

(arb

itra

ry a

bso

rba

nce

un

its)

[propionaldehyde] (mmol/L)

62

A.

B.

Figure 12. The (A) alcohol dehydrogenase (ADH) and (B) aldehyde dehydrogenase enzyme

activity in human kidney. Total (200 µg) protein from human kidney tissue was added to 0.5

mol/L tris(hydroxymethyl)aminomethane hydrochloride (Tris-HCL, pH 7.4), 3 mmol/L

nicotinamide adenine dinucleotide (NAD+) in the presence or absence of the substrates, 10

mmol/L ethanol (determination of ADH activity) or 1 mmol/L propioaldehyde (determination of

ALDH activity). The reaction mixture was incubated for 4 hrs, and then the optical density at

340 nm was measured using BioTek® Synergy HT microplate reader. The enzyme activity

assays were performed in triplicates. The results are presented as the mean ± standard error

(SE).

*

0.16

0.18

0.2

0.22

0.24

0.26

0.28

0.3

0 10

A340

(arb

itra

ry a

bso

rba

nce

un

its)

[ethanol] (mmol/L)

*

0.16

0.17

0.18

0.19

0.2

0.21

0.22

0.23

0 1

A340

(arb

itra

ry a

bso

rba

nce

un

its)

[propionaldehyde] (mmol/L)

63

4.4.3 The effect of 4-methylpyrazole on HK-2 cell viability

Figure 13 illustrates the effect of 4-methylpyrazole on HK-2 cell viability. Compared to

untreated control, acyclovir (500 – 2000 µg/mL) induced significant (p<0.05) concentration –

dependent decreases (17, 30, 40 and 49 %) in HK- 2 cell viability, respectively. Compared to

untreated control, significant (p<0.05) concentration – dependent decreases (15, 26, 38 and 47

%) in the viability of HK-2 cells were also observed following co-exposure to acyclovir and 4-

methylpyrazole, respectively.

The magnitude of the decreases in the viability of HK-2 cells co-exposed to acyclovir and 4-

methylpyrazole was less, compared to cells that were exposed to acyclovir. The differences

between the magnitude of the decrease in HK-2 cell viability exposed to acyclovir and HK-2

cells co-exposed to acyclovir and 4-methylpyrazole was significant (p<0.05) for all respective

acyclovir concentrations.

64

Figure 13. The effect of 4-methylpyrazole on human renal proximal tubular (HK-2) cell

viability. Cells were seeded (1.25E+05 cells/well) in 12-well plates. Initially, cell monolayers

were incubated with 4-methylpyrazole (500 µmol/L) for 1 hr. Following the incubation period,

the media was removed, and cell monolayers were incubated with acyclovir (0 – 2000 µg/mL) in

the presence or absence of 4-methylpyrazole for 24 hrs. Cell viability assays was assessed at the

end of the 24 hr incubation period. Cell viability was assessed using the fluorescent alamarBlue®

assay. The fluorescence was measured using a BioTek® Synergy HT microplate reader at

excitation and emission wavelengths of 540 and 590 nm, respectively. Cell viability assays were

performed in replicates of 9. The cell viability results are expressed as a percentage of the

fluorescence of untreated control cell monolayers and presented as the mean ± standard error

(SE). Statistically significant (p<0.05) differences between the fluorescence from untreated

control cell monolayers and cell monolayers exposed to acyclovir are denoted by the symbol*.

Statistically significant (p<0.05) differences between the fluorescence from cell monolayers

exposed to acyclovir and cell monolayers co-exposed to acyclovir and 4-methylpyrazole are

denoted by the symbol#.

65

4.4.4 Aldehyde production in HK-2 cells exposed to acyclovir

Figure 14 illustrates aldehyde production in HK-2 cells exposed to acyclovir (0, 2000 µg/mL).

Compared to untreated control, there appeared to be an increase in absorbance at 550 nm, in the

lysate of HK-2 cells that were exposed to acyclovir (2000 µg/mL).

Figure 14. Aldehyde production in human renal proximal tubular (HK-2) cells exposed to

acyclovir. Briefly, cell were seeded (2.50E+05 cells/dish) in petri dishes. Cell monolayers

were incubated with acyclovir (0 or 2000 µg/mL) for 3 hrs. Aldehyde production in cells was

determined using the Purpald® reagent. The absorbance was measured at 550 nm using a

Shimadzu UV160U spectrophotometer. The assay was not conducted in replicates.

0.000

0.001

0.002

0.003

0.004

0.005

0.006

0.007

0.008

0.009

0 2000

intr

ace

llu

lar

ald

ehy

de

pro

du

ctio

n

(arb

itra

ry a

bso

rba

nce

un

its,

A550)

[acyclovir] (µg/mL)

66

4.4.5 Comparison of the ADH protein expression level between HK-2 cells and human

kidney

Figure 15 compares the ADH protein expression level between HK-2 cells and human kidney.

Compared to HK-2 cells, the ADH protein expression in human kidney is approximately 30 fold

higher.

Figure 15. Comparison of the alcohol dehydrogenase (ADH) protein expression level

between the immortalized human renal proximal tubular (HK-2) cell line and human

kidney. The ADH protein expression in HK-2 cells and the human kidney tissue was quantified

using the Image Processing and Analysis in Java (ImageJ) software. The results are expressed as

the ADH protein expression level (relative to HK-2 cells) and are presented as the mean ± SE

from the quantification performed using the 2 western blots that were each obtained from

analyses of ADH protein expression in HK-2 and human kidney.

0

5

10

15

20

25

30

35

40

HK-2 Kidney

AD

H p

rote

in e

xp

ress

ion

lev

el

(co

mp

are

d t

o H

K-2

cell

s)

67

4.5 Discussion

The HK-2 cells were used as the in vitro model in our studies. These cells express functional

class I ADH (Figures 9A and 11A) and ALDH2 (Figures 9B and 11B) isozymes. Therefore, the

HK-2 cells possess the enzymatic machinery to locally produce acyclovir aldehyde and

subsequently metabolize it to CMMG.

The HK-2 cells are derived from the normal kidney of an adult male and have functional and

biochemical characteristics of well-differentiated renal proximal tubular cells (Ryan et al. 1994).

Furthermore, the use of HK-2 cells in our study eliminated the concern of inter-species

differences that exist in the pharmacological and toxicological disposition of acyclovir (de

Miranda et al. 1982a; 1982b). While, the predominant location of acyclovir – induced tubular

injury, along the nephron, is presently unknown; it is highly probable that the direct renal tubular

injury induced by acyclovir occurs mainly in the proximal tubule segment of the nephron. The

proximal tubule segment of the nephron is considered to be the predominant site of toxicant –

induced renal damage due to its leaky epithelium and active transport systems that results in the

accumulation of xenobiotics and local toxicity to the epithelial cells (Schnellmann 2001).

Additionally, compared to other segments of the nephron, the renal proximal tubular cells are

known to express increased levels of drug biotransformation enzymes that may result in the

production of increased concentrations of reactive metabolites (Schnellmann 2001). The HK-2

cells are of proximal tubular origin and therefore the use of the cells to study acyclovir – induced

nephrotoxicity allows for a better elucidation of the direct tubular damage induced by acyclovir,

in vitro, and possibly in vivo, as well. However, marked differences exist between in vitro and in

vivo systems (Davila et al. 1998), and thus, caution should be exercised during the extrapolation

of in vitro results to in vivo systems.

68

A critical element in our results is that 4-methylpyrazole rendered significant protection from

acyclovir – induced cell death (Figure 13). These results suggest that the acyclovir aldehyde

metabolite may play a role in the direct renal tubular injury associated with the parent drug, as

prevention of its modulation decreases toxicity. These results are the first to provide

experimental evidence which suggest that the acyclovir aldehyde may directly be involved in the

pathogenesis of the direct toxicity. Furthermore, our results suggest that acyclovir was oxidized

by the ADH enzyme to produce an aldehyde metabolite in HK-2 cells, in vitro, and in turn

provide further support for the appropriate use of the HK-2 cells in our study.

Based on the results presented in Figure 13, it can be argued that CMMG, and not the acyclovir

aldehyde may induce cell death. The figure illustrated that the magnitude of cell death was

significantly greater in cells exposed to acyclovir, compared to cells co-exposed to acyclovir and

4-methylprazole. In the absence of 4-methylpyrazole, acyclovir was likely oxidized to acyclovir

aldehyde, which was further metabolized to CMMG. It has been postulated that CMMG is

responsible for acyclovir – induced neurotoxicity (Hellden et al. 2003; 2006). However,

presently there is no biologically plausible evidence to support the hypothesis (Hellden et al.

2003; 2006). Therefore, we tested the effect of CMMG on HK-2 cell viability to determine if the

metabolite induces cell death. Cell monolayers were exposed to increasing concentrations (0 –

23.9 µg/mL) of CMMG for the same duration of time that they were exposed to acyclovir. Our

results illustrate that CMMG does not induce HK-2 cell death (data not shown).

Figure 15 illustrates that compared to untreated control; there was increased absorbance at 550

nm in the lysate obtained from the cell monolayer that was exposed to acyclovir (2000 µg/mL).

The results suggest that an aldehyde may have been produced in the cells exposed to acyclovir,

69

and hence, provide additional proof for the oxidation of acyclovir to acyclovir aldehyde via the

action of the ADH enzyme in human renal proximal tubular HK-2 cells.

Synthesizing all these data, the results of our study suggest that acyclovir aldehyde may be the

source of the direct renal tubular injury associated with the use of acyclovir. Intermediate

aldehyde metabolites have been suggested to play a leading role in several drugs – induced

toxicities, including aplastic anemia, urotoxicity, hepatoxicity, neurotoxicity and nephrotoxicity

(O’Brien et al. 2005). For example, acrolein, the aldehyde metabolite of the chemotherapeutic

agent, cyclophosphamide, is believed to be responsible for cyclophosphamide – induced

urotoxicity (Ramu et al. 1995). Similarly, it has been suggested that atropaldehyde, the aldehyde

metabolite of the antiepileptic agent, felbamate, mediates the parent drug – induced aplastic

anemia and hepatotoxicity (Kapetanovic et al. 2002). Biologically plausible mechanisms of

acyclovir aldehyde – induced cytotoxicity may include lipid peroxidation (Shaw and Jayatilleke

1987), depletion or inhibition of detoxifying compounds or enzymes (i.e. glutathione, ALDH)

(Kapetanovic et al. 2002; Shangari and O’Brien 2004), formation of reactive oxygen species

(O’Brien et al. 2005; Shaw and Jayatilleke 1987), mitochondrial damage (Shangari and O’Brien

2004), the formation of protein (Maggs and Park 1988) and deoxyribonucleic acid (DNA)

adducts (Wang et al. 2002).

The site(s) of acyclovir metabolism to CMMG has not been studied. To the best of our

knowledge, the acyclovir aldehyde metabolite has not been measured in any human biological

specimens, including plasma and urine. In this study, we hypothesized that the acyclovir

aldehyde may play a role in the direct renal tubular injury induced by acyclovir. Aldehydes are

unstable, highly reactive electrophilic compounds (O’Brien et al. 2005) and therefore in order for

acyclovir aldehyde to induce damage to renal proximal tubular cells, the metabolite must be

70

produced locally in the cells at a concentration that is sufficient to cause toxicity. Our results

suggest that human renal proximal tubular cells have the capacity to locally metabolize acyclovir

to its aldehyde metabolite. However, the concentrations of acyclovir aldehyde encountered by

human renal proximal tubular cell in vitro or in vivo are not known. Future studies using the

immortalized HK-2 cell line or primary human renal proximal tubular cell cultures could be

performed to determine those data. However, regardless of the analytical method used to

quantify the amount of acyclovir aldehyde that is produced in tubular cells, it will be necessary

to produce a standard curve using the pure acyclovir aldehyde compound. Presently, to the best

of our knowledge, the acyclovir aldehyde compound is not commercially available, and the

process of synthesizing it is close to impossible because the process is time consuming, tedious,

costly and the stability of the aldehyde is unknown and/or not guaranteed. However, in our

study, if acyclovir aldehyde was the cause of HK-2 cell death, then sufficient amounts were

formed to induce substantial cell death, which was partially, yet significantly alleviated

following co-exposure to 4-methylpyrazole.

In the HK-2 cells, the effect of 4-methylpyrazole on HK-2 cell viability was small. The small

magnitude of the effect was likely due to the inherent low protein expression of the ADH

enzyme in HK-2 cells, leading to limited metabolism of acyclovir to acyclovir aldehyde and

hence, inhibition by 4-methylpyrazole. Future studies can be conducted using cells that

overexpress human ADH in order to determine whether co-exposure to 4-methylpyrazole renders

a greater magnitude of protection from acyclovir – induced cytotoxicity. The results may

provide further evidence to support the hypothesis that the acyclovir aldehyde may play a role in

direct cytotoxicity.

71

In order to extrapolate the biological significance of our cell results to humans, we performed

western blot analyses and enzyme activity assays on a sample of human kidney tissue. The

human kidney is known to express both class I ADH (Engeland and Maret 1993; Nishimura and

Naito 2006) and ALDH2 (Harada et al. 1980) enzymes. Our results (Figures 10 and 12) confirm

the functional protein expression of the enzymes in the human kidney. Comparison of the ADH

protein expression between HK-2 cells and human kidney revealed that the protein expression of

the enzyme may be at least 30 fold higher in human kidney (Figure 15). Therefore, it is very

likely that inhibition of the ADH enzyme by 4-methylpyrazole would have a substantially more

pronounced effect in human renal proximal tubular cells, in vivo.

There may be other potential explanations for the small measured effect of 4-methylpyrazole on

HK-2 cell viability. For instance, it is plausible that the small effect may have been due to the

noxious effect(s) of the acyclovir aldehyde metabolite on ADH expression and/or activity.

Aldehydes are known to reduce the activity of enzymes (O’Brien et al. 2005), specifically its

detoxifying enzyme, aldehyde dehydrogenase (Doorn et al. 2006; Kapetanovic et al. 2002). The

effect(s) of aldehydes on the expression or activity of alcohol dehydrogenase enzymes is not

known. However, it is probable that formation of aldehyde can affect ADH functional

expression either through direct or indirect mechanisms of toxicity (Gregus and Klaassen 1998).

Second, it is likely that the enzyme was only partially inhibited by 4-methylpyrazole. Finally, it

can be speculated that the small effect may have been also due to the possibility that the parent

drug; acyclovir may also play a role in the direct toxicity. As previously elaborated, acyclovir

undergoes minimal metabolism via the ADH and ALDH pathway,5 therefore, it is possible that

the ADH enzyme has a weak affinity for the parent drug, and if this is the case then in addition,

to its aldehyde metabolite, the parent drug may also play an active role and may even be the

72

more predominant offending nephrotoxic. Biologically plausible mechanism(s) of the parent

drug’s toxicity is not known. Acyclovir’s primary mechanism of pharmacological action

involves inhibition of viral DNA replication (Elion 1983).

In addition to illustrating that locally produced acyclovir aldehyde may be the source of

acyclovir – induced nephrotoxicity, the results of our studies may also provide a potential

mechanism for the variation in the incidence of acyclovir – induced nephrotoxicity. Several

factors, including age (Meier and Seitz 2008), gender (Chrostek et al. 2003), tissue type (Brown

et al. 1996; Engeland and Maret 1993; Nishimura and Naito 2006) and more importantly, genetic

polymorphisms (Bosron and Li 1986; Mizoi et al. 1994; Mulligan et al. 2003; Stickel and

Osterreicher 2006) are well known to affect the functional expression of enzymes, including the

ADH and ALDH enzymes. Altered functional expression of ADH or ALDH enzymes affects the

disposition of substrates that are metabolized by the enzymes and may subsequently contribute to

the inter-individual variation in the occurrence of certain drug – induced toxicities.

Unlike their effect on nephrotoxicity, the effect of genetic polymorphisms of the ADH or ALDH

enzymes on hepatotoxicity has been well documented. For instance, ethanol – induced

hepatotoxicity is largely attributed to locally produced, acetaldehyde (Matsuzaki and Seiber

1977), from the oxidation of ethanol by ADH, which is subsequently metabolized by the ALDH

enzyme to acetate (Zakhari 2006). Genetic polymorphisms of the ADH or ALDH enzymes

resulting in increased production or reduced catabolism of acetaldehyde may increase the risk of

occurrence of hepatotoxicity in patients, respectively (Crabb et al. 2004). It is conceivable that

genetic polymorphisms of ADH and ALDH enzymes may alter the kidney disposition of

acyclovir and increase the risk of nephrotoxicity in some individuals.

73

In conclusion, the novel evidence presented in this study suggests that the acyclovir aldehyde

may cause direct renal tubular injury. There is need for several future studies including the

determination of the cellular and molecular mechanism(s) of acyclovir aldehyde – induced

toxicity and the specific ADH and ALDH isozymes that are responsible for the metabolism of

acyclovir and its aldehyde metabolite, respectively, and the affinity of the enzymes for the

respective substrates. Such studies will aid in a better understanding of the pathogenesis of the

direct renal tubular injury induced by acyclovir and may lead to potential therapy for these

serious adverse effects.

4.6 Statement of significance

The results from the study suggest that locally produced acyclovir aldehyde may play a role in

the direct renal tubular injury associated with the use of its parent drug, and hence offers the first

insight into the potential underlying mechanism(s) of this drug – induced nephrotoxicity.

4.7 Acknowledgements

The authors declare that they have no conflicts of interest.

All authors have read the journal’s policy on disclosure of potential conflicts of interest.

We would like to thank GlaxoSmithKline (Raleigh, NC) for providing us with the CCMG

chemical compound for this research study.

The study was supported by the grant from CIHR.

74

4.8 References

Ahmad, T., Simmonds, M., McIver, A.G., and McGraw, M.E. 1994. Reversible renal failure in

renal transplant patients receiving oral acyclovir prophylaxis. Pediatr Nephrol 8: 489-491.

Aleksa, K., Ito, S., and Koren, G. 2006. Enantioselective metabolism of ifosfamide by the

kidney. Chirality 18: 398-405.

Anders, M.W. 1980. Metabolism of drugs by the kidney. Kidney Int 18: 636-647.

Bianchetti, M.G., Roduit, C., and Oetliker, O.H. 1991. Acyclovir-induced renal failure: course

and risk factors. Pediatr Nephrol 5: 238-239.

Bosron, W.F., and Li, T.K. 1986. Genetic polymorphism of human liver alcohol and aldehyde

dehydrogenases, and their relationship to alcohol metabolism and alcoholism. Hepatology 6:

502-510.

Brigden, D., Rosling, A.E., and Woods, N.C. 1982. Renal function after acyclovir intravenous

injection. Am J Med 73: 182-185.

Brigden, D., and Whiteman, P. 1985. The clinical pharmacology of acyclovir and its prodrugs.

Scand J Infect Dis Suppl 47: 33-39.

Brown, C.J., Zhang, L., and Edenberg, H.J. 1996. Gene expression in a young multigene family:

tissue-specific differences in the expression of the human alcohol dehydrogenase genes ADH1,

ADH2, and ADH3. DNA Cell Biol 15: 187-196.

Chrostek, L., Jelski, W., Szmitkowski, M., and Puchalski, Z. 2003. Gender-related differences in

hepatic activity of alcohol dehydrogenase isoenzymes and aldehyde dehydrogenase in humans. J

Clin Lab Anal 17: 93-96.

Clemens, D.L., Halgard, C.M., Miles, R.R., Sorrell, M.F., and Tuma, D.J. 1995. Establishment

of a recombinant hepatic cell line stably expressing alcohol dehydrogenase. Arch Biochem

Biophys 321: 311-318.

Crabb, D.W., Matsumoto, M., Chang, D., and You, M. 2004. Overview of the role of alcohol

dehydrogenase and aldehyde dehydrogenase and their variants in the genesis of alcohol-related

pathology. Proc Nutr Soc 63: 49-63.

Davila, J.C., Rodriguez, R.J., Melchert, R.B., and Acosta, D., Jr. 1998. Predictive value of in

vitro model systems in toxicology. Annu Rev Pharmacol Toxicol 38: 63-96.

de Miranda, P., and Burnette, T.C. 1994. Metabolic fate and pharmacokinetics of the acyclovir

prodrug valaciclovir in cynomolgus monkeys. Drug Metab Dispos 22; 55-59.

75

de Miranda, P., and Good, S.S. 1982. Biotransformation of acyclovir to 9-

carboxymethoxymethylguanine. Federation Proceedings 41: 1733.

de Miranda, P., and Good, S.S. 1992. Species differences in the metabolism and disposition of

antiviral nucleoside analogues. 1. Acyclovir. Antivir Chem Chemother 3: 1-8.

de Miranda, P., Good, S.S., Krasny, H.C., Connor, J.D., Laskin, O.L., and Lietman, P.S. 1982a.

Metabolic fate of radioactive acyclovir in humans. Am J Med 73: 215-220.

de Miranda, P., Krasny, H.C., Page, D.A., and Elion, G.B. 1982b. Species differences in the

disposition of acyclovir. Am J Med 73: 31-35.

Diamond, G.L., and Quebbemann, A.J. 1981. In vivo quantification of renal sulfate and

glucuronide conjugation in the chicken. Drug Metab Dispos 9: 402-409.

Doorn, J.A., Hurley, T.D., and Petersen, D.R. 2006. Inhibition of human mitochondrial aldehyde

dehydrogenase by 4-hydroxynon-2-enal and 4-oxonon-2-enal. Chem Res Toxicol 19: 102-110.

Dubourg, L., Michoudet, C., Cochat, P., and Baverel, G. 2001. Human kidney tubules detoxify

chloroacetaldehyde, a presumed nephrotoxic metabolite of ifosfamide. J Am Soc Nephrol 12:

1615-1623.

Elion, G.B. 1983. The biochemistry and mechanism of action of acyclovir. J Antimicrob

Chemother 12 Suppl B: 9-17.

Engeland, K., and Maret, W. 1993. Extrahepatic, differential expression of four classes of human

alcohol dehydrogenase. Biochem Biophys Res Commun 193: 47-53.

Estonius, M., Svensson, S., and Hoog, J.O. 1996. Alcohol dehydrogenase in human tissues:

localisation of transcripts coding for five classes of the enzyme. FEBS Lett 397: 338-342.

Fletcher, C.V., Englund, J.A., Bean, B., Chinnock, B., Brundage, D.M., and Balfour, H.H., Jr.

1989. Continuous infusion of high-dose acyclovir for serious herpesvirus infections. Antimicrob

Agents Chemother 33: 1375-1378.

Genc, G., Ozkaya, O., Acikgoz, Y., Yapici, O., Bek, K., Gulnar Sensoy, S., and Ozyurek, E.

2010. Acute renal failure with acyclovir treatment in a child with leukemia. Drug Chem Toxicol

33; 217-219.

Gregus Z. and Klaassen, C.D. 2001. Mechanisms of Toxicity. In Casarett and Doull's Toxicology

The Basic Science of Poisons. Edited by Klaassen, C.D. The Mc-Graw Hill Companies Inc.,

New York. pp 35-81.

Gunness, P., Aleksa, K., Kosuge, K., Ito, S., and Koren, G. 2010. Comparison of the novel HK-2

human renal proximal tubular cell line with the standard LLC-PK1 cell line in studying drug-

induced nephrotoxicity. Can J Physiol Pharmacol 88: 448-455.

76

Gyamfi, M.A., and Wan, Y.J. 2006. The effect of ethanol, ethanol metabolizing enzyme

inhibitors, and Vitamin E on regulating glutathione, glutathione S-transferase, and S-

adenosylmethionine in mouse primary hepatocyte. Hepatol Res 35: 53-61.

Hara, K., Suyama, K., Itoh, H., and Nagashima, S. 2008. Influence of ALDH2 genetic

polymorphisms on aciclovir pharmacokinetics following oral administration of valaciclovir in

Japanese end-stage renal disease patients. Drug Metab Pharmacokinet 23: 306-312.

Harada, S., Agarwal, D.P., and Goedde, H.W. 1980. Electrophoretic and biochemical studies of

human aldehyde dehydrogenase isozymes in various tissues. Life Sci 26: 1773-1780.

Hellden, A., Lycke, J., Vander, T., Svensson, J.O., Odar-Cederlof, I., and Stahle, L. 2006. The

aciclovir metabolite CMMG is detectable in the CSF of subjects with neuropsychiatric symptoms

during aciclovir and valaciclovir treatment. J Antimicrob Chemother 57; 945-949.

Hellden, A., Odar-Cederlof, I., Diener, P., Barkholt, L., Medin, C., Svensson, J.O., Sawe, J., and

Stahle, L. 2003. High serum concentrations of the acyclovir main metabolite 9-

carboxymethoxymethylguanine in renal failure patients with acyclovir-related neuropsychiatric

side effects: an observational study. Nephrol Dial Transplant 18: 1135-1141.

Hintz, M., Connor, J.D., Spector, S.A., Blum, M.R., Keeney, R.E., and Yeager, A.S. 1982.

Neonatal acyclovir pharmacokinetics in patients with herpes virus infections. Am J Med 73: 210-

214.

Kapetanovic, I.M., Torchin, C.D., Strong, J.M., Yonekawa, W.D., Lu, C., Li, A.P., Dieckhaus,

C.M., Santos, W.L., Macdonald, T.L., Sofia, R.D., and Kupferberg, H.J. 2002. Reactivity of

atropaldehyde, a felbamate metabolite in human liver tissue in vitro. Chem Biol Interact 142:

119-134.

Keeney, R.E., Kirk, L.E., and Bridgen, D. 1982. Acyclovir tolerance in humans. Am J Med 73:

176-181.

Laemmli, U.K. 1970. Cleavage of structural proteins during the assembly of the head of

bacteriophage T4. Nature 227; 680-685.

Lohr, J.W., Willsky, G.R., and Acara, M.A. 1998. Renal drug metabolism. Pharmacol Rev 50:

107-141.

Lyon, A.W., Mansoor, A., and Trotter, M.J. 2002. Urinary gems: acyclovir crystalluria. Arch

Pathol Lab Med 126: 753-754.

Maggs, J.L., and Park, B.K. 1988. Drug-protein conjugates--XVI. Studies of sorbinil

metabolism: formation of 2-hydroxysorbinil and unstable protein conjugates. Biochem

Pharmacol 37: 743-748.

77

Mason, W.J., and Nickols, H.H. 2008. Crystalluria from acyclovir use. N Engl J Med 358; e14.

Matsuzaki, S., and Sieber, C.S. 1977. Increased susceptibility of hepatic mitochondria to the

toxicity of acetaldehyde after chronic ethanol consumption. Biochem Biophys Res Commun 75:

1059-1065.

Meier, P., and Seitz, H.K. 2008. Age, alcohol metabolism and liver disease. Curr Opin Clin Nutr

Metab Care 11: 21-26.

Millipore. 2007. Millipore Technical Publications. RIPA Buffer - Preparation of modified

radioimmunoprecipitation (RIPA) buffer.

[http://www.millipore.com/userguides/tech1/mcproto402].

Mizoi, Y., Yamamoto, K., Ueno, Y., Fukunaga, T., and Harada, S. 1994. Involvement of genetic

polymorphism of alcohol and aldehyde dehydrogenases in individual variation of alcohol

metabolism. Alcohol Alcohol 29: 707-710.

Mulligan, C.J., Robin, R.W., Osier, M.V., Sambuughin, N., Goldfarb, L.G., Kittles, R.A.,

Hesselbrock, D., Goldman, D., and Long, J.C. 2003. Allelic variation at alcohol metabolism

genes ( ADH1B, ADH1C, ALDH2) and alcohol dependence in an American Indian population.

Hum Genet 113: 325-336.

Nishimura, M., and Naito, S. 2006. Tissue-specific mRNA expression profiles of human phase I

metabolizing enzymes except for cytochrome P450 and phase II metabolizing enzymes. Drug

Metab Pharmacokinet 21: 357-374.

O'Brien, P.J., Siraki, A.G., and Shangari, N. 2005. Aldehyde sources, metabolism, molecular

toxicity mechanisms, and possible effects on human health. Crit Rev Toxicol 35: 609-662.

Pawan, G.L. 1972. Metabolism of alcohol (ethanol) in man. Proc Nutr Soc 31; 83-89.

Peterslund, N.A., Larsen, M.L., and Mygind, H. 1988. Acyclovir crystalluria. Scand J Infect Dis

20: 225-228.

Potter, J.L., and Krill, C.E., Jr. 1986. Acyclovir crystalluria. Pediatr Infect Dis 5: 710-712.

Quesenberry, M.S., and Lee, Y.C. 1996. A rapid formaldehyde assay using purpald reagent:

application under periodation conditions. Anal Biochem 234: 50-55.

Ramu, K., Fraiser, L.H., Mamiya, B., Ahmed, T., and Kehrer, J.P. 1995. Acrolein mercapturates:

synthesis, characterization, and assessment of their role in the bladder toxicity of

cyclophosphamide. Chem Res Toxicol 8: 515-524.

Ryan, M.J., Johnson, G., Kirk, J., Fuerstenberg, S.M., Zager, R.A., and Torok-Storb, B. 1994.

HK-2: an immortalized proximal tubule epithelial cell line from normal adult human kidney.

Kidney Int 45: 48-57.

78

Sawyer, M.H., Webb, D.E., Balow, J.E., and Straus, S.E. 1988. Acyclovir-induced renal failure.

Clinical course and histology. Am J Med 84: 1067-1071.

Schnellmann, R.G. 2001. Toxic responses of the kidney. In Casarett and Doull's Toxicology The

Basic Science of Poisons. Edited by Klaassen, C.D. The Mc-Graw Hill Companies Inc., New

York. pp 491-503.

Schreiber, R., Wolpin, J., and Koren, G. 2008. Determinants of aciclovir-induced nephrotoxicity

in children. Paediatr Drugs 10: 135-139.

Shangari, N., and O'Brien, P.J. 2004. The cytotoxic mechanism of glyoxal involves oxidative

stress. Biochem Pharmacol 68: 1433-1442.

Shaw, S., and Jayatilleke, E. 1987. Acetaldehyde-mediated hepatic lipid peroxidation: role of

superoxide and ferritin. Biochem Biophys Res Commun 143: 984-990.

Silverthorn, D.U. 1998. The Kidneys. In Human Physiology An Integrated approach. Edited by

Brake, D.R.. Prentice Hall., Upper Saddle River, New Jersey. pp. 518-542.

Stickel, F., and Osterreicher, C.H. 2006. The role of genetic polymorphisms in alcoholic liver

disease. Alcohol Alcohol 41: 209-224.

Towbin, H., Staehelin, T., and Gordon, J. 1979. Electrophoretic transfer of proteins from

polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad

Sci U S A 76: 4350-4354.

Vachvanichsanong, P., Patamasucon, P., Malagon, M., and Moore, E.S. 1995. Acute renal failure

in a child associated with acyclovir. Pediatr Nephrol 9: 346-347.

Vomiero, G., Carpenter, B., Robb, I., and Filler, G. 2002. Combination of ceftriaxone and

acyclovir - an underestimated nephrotoxic potential? Pediatr Nephrol 17: 633-637.

Walker, D., Flinois, J.P., Monkman, S.C., Beloc, C., Boddy, A.V., Cholerton, S., Daly, A.K.,

Lind, M.J., Pearson, A.D., Beaune, P.H., and Jeffrey, R.I. 1994. Identification of the major

human hepatic cytochrome P450 involved in activation and N-dechloroethylation of ifosfamide.

Biochem Pharmacol 47: 1157-1163.

Walker, J.R.L. 1992. Spectrophotometric determination of enzyme activity: alcohol

dehydrogenase (ADH). Biochemical Education 20: 42-43.

Wang, M., McIntee, E.J., Cheng, G., Shi, Y., Villalta, P.W., and Hecht, S.S. 2000. Identification

of DNA adducts of acetaldehyde. Chem Res Toxicol 13; 1149-1157.

Zakhari, S. 2006. Overview: how is alcohol metabolized by the body? Alcohol Res Health 29:

245-254.

79

4.9 Additional experiments not published

4.9.1 The effect of CMMG on cell viability

Based on the results presented in Figure 13, it may be postulated that CMMG and not the

acyclovir aldehyde metabolite is responsible for HK-2 cell death. Therefore, it was necessary to

determine whether CMMG induces HK-2 cell death.

4.9.2 Materials and methods

4.9.2a Exposure to CMMG

A cytotoxicity (alamarBlue® assay) was performed to determine whether CMMG

(GlaxoSmithKline, Raleigh, North Carolina, USA) induces HK-2 cell death. The assay and cell

culture regimen was performed as previously described (section 4.3.5). Cell monolayers were

incubated with CMMG [0 – 23.9 µg/mL (0 – 100 µmol/L)] for 24 hours. Cell viability was

assessed at the end of the incubation period. The assay was performed in replicates of 9.

4.9.2b Statistical analyses

The statistical analyses of the data were performed using the IBM®

SPSS® Statistics version 19

software. The ANOVA followed by Dunnett’s post hoc tests were conducted to assess the

statistical significance of the data from cell viability assays that were conducted to determine

whether CMMG induced HK-2 cell death.

4.9.3 Results

Figure 16 illustrates the effect of CMMG on HK-2 cell viability. Compared to untreated control,

CMMG (0.0239 and 0.239 µg/mL) did not induce HK-2 cell death. Exposure to CMMG (2.39

and 23.9 µg/mL) induced significant (p<0.05) increases in HK-2 cell viability.

80

Figure 16. The effect of 9-carboxymethoxymethylguanine (CMMG) on human renal

proximal tubular (HK-2 cell) viability. Cells were seeded (1.25E+05 cells/well) in 12-well

plates. Cell monolayers were incubated with CMMG [0 – 23.9 µg/mL (0 – 100 µmol/L)] for 24

hrs. Cell viability assays was assessed at the end of the 24 hr incubation period. Cell viability

was assessed using the fluorescent alamarBlue®

assay. The fluorescence was measured using a

BioTek® Synergy HT microplate reader at excitation and emission wavelengths of 540 and 590

nm, respectively. Cell viability assays were performed in replicates of 9. The cell viability

results are expressed as a percentage of the fluorescence of untreated control cell monolayers and

presented as the mean ± standard error (SE). Statistically significant (p<0.05) differences

between the fluorescence from untreated control cell monolayers and cell monolayers exposed to

acyclovir are denoted by the symbol*.

* *

0

20

40

60

80

100

120

0 0.02 0.24 2.39 23.90

cell

via

bil

ity

(% u

ntr

ea

ted

co

ntr

ol)

[CMMG] (µg/mL)

81

Chapter 5

The effect of acyclovir on the tubular secretion of creatinine in vitro

Patrina Gunness,a,b

Katarina Aleksa,a Gideon Koren

a,b

aDivision of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, 555

University Avenue, Toronto, ON, M5G 1X8, Canada

bGraduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University

of Toronto, ON, M5S 3M2, Canada

This article has been published: Gunness, P., Aleksa, K., and Koren, G. 2010. The effect of

acyclovir on the tubular secretion of creatinine in vitro. J Transl Med 8: 139-149. This article

was originally published by BioMed Central.

[PG performed all experiments and prepared the manuscript for submission]

82

5.0 Abstract

While generally well tolerated, severe nephrotoxicity has been observed in some children

receiving acyclovir. A pronounced elevation in plasma creatinine in the absence of other clinical

manifestations of overt nephrotoxicity has been frequently documented. Several drugs have

been shown to increase plasma creatinine by inhibiting its renal tubular secretion rather than by

decreasing glomerular filtration rate. Creatinine and acyclovir may be transported by similar

tubular transport mechanisms, thus, it is plausible that in some cases, the observed increase in

plasma creatinine may be partially due to inhibition of tubular secretion of creatinine, and not

solely due to decreased GFR. Our objective was to determine whether acyclovir inhibits the

tubular secretion of creatinine. The LLC-PK1 and HK-2 renal proximal tubular cell monolayers

cultured on microporous membrane filters were exposed to [2-14

C] creatinine (5 µmol/L) in the

absence or presence of quinidine (1000 µmol/L), cimetidine (1000 µmol/L) or acyclovir (22 – 89

µmol/L) in incubation medium. Results illustrated that in evident contrast to quinidine, acyclovir

did not inhibit creatinine transport in LLC-PK1 and HK-2 cell monolayers. The results suggest

that acyclovir does not affect the renal tubular handling of creatinine, and hence, the pronounced,

transient increase in plasma creatinine is due to decreased GFR, and not to a spurious increase in

plasma creatinine.

5.1 Introduction

Acyclovir is an antiviral agent that is commonly used to treat severe viral infections including

herpes simplex and varicella zoster, in children (Bryson 1984). Acyclovir is generally well

tolerated (Keeney et al. 1982), however, in some cases; severe nephrotoxicity has been reported

(Bianchetti et al. 1991; Brigden et al. 1982; Chou et al. 2008; Keeney et al. 1982; Potter and Krill

1986; Vachvanichsanong et al. 1995; Vomiero et al. 2002). Acyclovir – induced nephrotoxicity

83

is typically evidenced by elevated plasma creatinine and urea levels, the occurrence of abnormal

urine sediments or acute renal failure (Bianchetti et al. 1991; Brigden et al. 1982; Chou et al.

2008; Keeney et al. 1982; Vachvanichsanong et al. 1995; Vomiero et al. 2002).

Crystalluria leading to obstructive nephropathy is widely believed to be the mechanism of

acyclovir – induced nephrotoxicity (Sawyer et al. 1988). However, there are several documented

cases of acyclovir – induced nephrotoxicity in the absence of crystalluria (Ahmad et al. 1994;

Vachvanichsanong et al. 1995; Vomiero et al. 2002); suggesting that acyclovir induces direct

insult to tubular cells. Recently, we provided the first in vitro experimental evidence which

supports existing clinical evidence of direct renal tubular damage induced by acyclovir (Gunness

et al. 2010).

A systematic review of the literature reveals a pronounced, transient elevation (up to 9 fold in

some cases) of plasma creatinine levels in children, often without any other clinical evidence of

overt nephrotoxicity (Table 3). Similar to the cases described in Table 3; a marked, transient

increase in plasma creatinine levels has been observed in some patients who received the non-

nephrotoxic drugs, cimetidine (Blackwood et al. 1976; Burgess et al. 1982; Dubb et al. 1978;

Dutt et al. 1981; Haggie et al. 1976), trimethoprim (Berglund et al. 1975; Kastrup et al. 1985;

Myre et al. 1978), pyrimethamine (Opravil et al. 1993), dronedarone (Tschuppert et al. 2007) and

salicylates (Burry and Dieppe 1976).

Creatinine, a commonly used biomarker that is used to assess renal function, is eliminated by the

kidney via both glomerular filtration and tubular secretion (Toto 1995). The mechanisms

underlying the renal tubular transport of creatinine has not been fully elucidated. As explained

by Urakami and colleagues (Urakami et al. 2004), both acid and base secreting mechanisms may

84

play a role in the renal tubular transport of creatinine (Arenshorst and Selkurt 1976; Berglund et

al. 1975; Burry and Dieppe 1976; Dubb et al. 1978; Dutt et al. 1981; Eisner et al. 2010; Kastrup

et al. 1985; Myre et al. 1987; Okuda et al. 2006; Opravil et al. 1993; Tschuppert et al. 2007).

Hence, some drugs may share similar renal tubular transport mechanisms with creatinine. Drugs

that share transport mechanisms with creatinine may compete with it for tubular transport, and

subsequently inhibit creatinine secretion to result in a ungenuine elevation of plasma creatinine

that may not be due to decreased GFR. Cimetidine (Blackwood et al. 1976; Burgess et al. 1982;

Dubb et al. 1978; Dutt et al. 1981; Haggie et al. 1976), trimethoprim (Berglund et al. 1975;

Kastrup et al. 1985; Myre et al. 1978), pyrimethamine (Opravil et al. 1993), dronedarone

(Tschuppert et al. 2007) and salicylates (Burry and Dieppe 1976) are examples of drugs that

share similar renal tubular transport mechanisms with creatinine and induce spurious increases in

plasma creatinine by competing with and subsequently inhibiting its secretion.

Similar to creatinine, both acid and base secreting pathways may be involved in the renal tubular

transport of acyclovir (Takeda et al. 2002). Additionally, it is likely that creatinine (Eisner et al.

2010; Okuda et al. 2006; Urakami et al. 2004) and acyclovir (Takeda et al. 2002) may be

transported by similar organic anion transporters (OAT) and OCTs. Therefore, it is plausible

that acyclovir may compete with and successively inhibit renal secretion of creatinine, resulting

in elevations in plasma creatinine that may be disproportional to the degree of renal dysfunction.

Employing plasma creatinine levels to estimate GFR, results from previous studies (Genc et al.

2010; Schreiber et al. 2008) have illustrated that acyclovir – induced nephrotoxicity induces a

significant reduction in GFR in children. However, based on: (1) the cases presented in Table 3,

(2) the awareness that several non-nephrotoxic drugs are known to induce transient increases in

plasma creatinine (Berglund et al. 1975; Blackwood et al. 1976; Burgess et al. 1982; Burry and

85

Dieppe 1976; Dubb et al. 1978; Dutt et al. 1981; Haggie et al. 1976; Kastrup et al. 1985; Myre et

al. 1987; Opravil et al. 1993; Tschuppert et al. 2007) and (3) the knowledge that acyclovir and

creatinine may share similar renal tubular transport mechanisms; we hypothesized that the

pronounced, transient increase in plasma creatinine levels observed in some patients may be

partially due to the inhibition of renal tubular secretion of creatinine by acyclovir, and not

entirely the result of decreased GFR. To the best of our knowledge, the effect of acyclovir on the

renal tubular secretion of creatinine in vitro has not been previously evaluated. Thus, the

objective of the study was to determine whether acyclovir inhibits the renal tubular secretion of

creatinine. It is important to determine whether acyclovir inhibits the tubular transport of

creatinine, because if this is the case, then in addition to creatinine, other biomarkers should

always be employed to assess renal function in patients receiving acyclovir treatment.

In the present study we were specifically interested in determining the possible interaction

between creatinine and acyclovir during renal tubular transport by the OCT pathway. The

porcine renal tubular cell line, LLC-PK1, has been used as an in vitro renal tubular model in a

vast array of transepithelial transport studies. Furthermore, the LLC-PK1 cells are an

appropriate in vitro model for specifically studying renal tubular transport of organic cations

because they are known to possess functional OCTs (Fauth et al. 1988; Saito et al. 1992;

Urakami et al. 2005). However, although the LLC-PK1 cells retain similar physiological and

biochemical properties compared to human renal proximal tubular cells (Perantoni and Berman

1979), interspecies differences in drug disposition exists (Davila et al. 1998; Eaton and Klaassen

2001; Riddick 1998). Hence, the use of a human renal proximal tubular cell line, such as the

HK-2 cell line, would be a more suitable in vitro model to study the mechanisms of renal tubular

86

drug transport in humans. Porcine LLC-PK1 and human HK-2 cells were employed in our

transepithelial transport studies.

Table 3. Cases of elevated plasma creatinine levels in children who received intravenous

acyclovir

Patient Magnitude of increase

in plasma creatinine

(from baseline)

Relevant clinical details References

1 child 5 fold increase within 2

days

Creatinine returned to normal in 4 days

Elevated urea

No other pathology reported

[Brigden et al. 1982]

10 children

transient elevation

No further impairment reported

[Keeney et al. 1982]

3 children

4 fold increase within 4

days

Mild reduction in urine output

Creatinine returned to normal 1 week

following acyclovir discontinuation

[Biachetti et al. 1991]

1 child

2 fold increase within 6

days

Creatinine continued to increase

following acyclovir discontinuation.

Creatinine returned to normal within 1

week

Elevated urea

Mild proteinuria

[Vacvanichsanong et

al. 1995]

3 children

9 fold increase within 2

to 3 days

High urea

Urinary α1-microglobulin and albumin

Creatinine returned to normal in 3 – 9

days

[Vomiero et al. 2002]

1 child

3 fold increase within 4

days

No other information provided

[Chou et al. 2008]

87

5.2 Materials and methods

5.2.1 Cell culture

The LLC-PK1 cells were cultured in growth medium which consisted of MEM alpha modified,

supplemented with 2 mmol/L L-glutamine, 100 Units/mL penicillin, 100 µg streptomycin and 10

% (v/v) FBS. The HK-2 cells were cultured in growth medium which consisted of Keratinocyte-

SFM, supplemented with human recombinant epidermal growth factor 1-53 (5 ng/mL) and

bovine pituitary extract (0.05 mg/mL). The LLC-PK1 and HK-2 cells were maintained at 37°C

in a sterile, humidified atmosphere of 5 % CO2 and 95 % O2.

5.2.2 Transepithelial transport studies

The transepithelial transport studies were conducted as outlined by Urakami and colleagues

(Urakami et al. 2005) with modifications. The LLC-PK1 and HK-2 cells were seeded at

densities of 4.5E+05 cells/0.9 cm2 and 5.0E+05 cells/0.9 cm

2, respectively, on microporous

membrane filter inserts (3 µm pore size, 0.9 cm2 growth area) that were placed inside cell culture

chambers (VWR International). A consistent (1 mL) volume of growth or incubation medium

(containing no substrates, radiolabeled or non-radiolabeled substrates) was placed in the apical

and basolateral compartments of the cell culture chambers during culturing of the cells or during

all transport experiments. The LLC-PK1 and HK-2 cell monolayers used for transport studies

were cultured in growth medium for 6 and 3 days, respectively, after seeding. All transepithelial

transport studies were conducted on confluent cell monolayers.

At the time of commencement of the transport experiments, the growth medium from the cell

culture chamber was removed and both sides of the cell monolayers were washed twice with

incubation medium (145 mmol/L NaCl, 3 mmol/L KCl, 1 mmol/L CaCl2, 0.5 mmol/L MgCl2, 5

88

mmol/L D-glucose and 5 mmol/L HEPES (pH 7.4)). Incubation medium was used for all

transport experiments. Cell monolayers were incubated with medium for 10 mins. Following

the 10 mins incubation period, the medium was removed and the cell monolayers were incubated

with medium as follows: the medium added to the basolateral compartment of the cell culture

chamber contained respective radiolabeled and non-radiolabeled substrates and the medium

added to the apical compartment of the cell culture chamber contained neither radiolabeled nor

non-radiolabeled substrates. The radiolabeled and non-radiolabeled substrates used in the

transport studies are outlined below.

The transepithelial transport (basolateral-to-apical) of radiolabeled substrates across the cell

monolayers was assessed at specific intervals (LLC-PK1: 0, 15, 30, 45 and 60 mins; HK-2: 0,

7.5, 15, 22.5 and 30 mins) over 60 and 30 mins, respectively. Studies were conducted over

different duration of times in LLC-PK1 and HK-2 cells due to differences in the integrity of the

cell monolayers. The paracellular flux (basolateral-to-apical) of D-[1-3H(N)] mannitol

(PerkinElmer) across the cell monolayers was used to assess the integrity of cell monolayers. A

priori decision was made to eliminate the results from any cell monolayers where the

paracellular flux of D-[1-3H(N)] mannitol across LLC-PK1 or HK-2 cell monolayers was greater

than 5 % over the respective experimental period.

The transport of radiolabeled substrates was assessed by measuring the radioactivity of 50 µL

aliquots of medium that were sampled from the apical and basolateral compartments of the cell

culture chamber, at the aforementioned specified time intervals for the respective cell line.

Radioactivity was measured as disintegrations per minutes (DPM) using a LS 6500 liquid

scintillation (Beckman Coulter Canada Inc., Mississauga, Ontario, Canada).

89

5.2.2a Tetraethylammonium (TEA) transport across cell monolayers

In order to determine whether the LLC-PK1 and HK-2 cells used in the present studies possessed

functional organic cation transporters; TEA transport across cell monolayers was assessed. The

TEA is a classical organic cation substrate for OCTs (Fauth et al. 1988; Grundemann et al. 1994;

Saito et al. 1992). The transport of TEA across LLC-PK1 and HK-2 cell monolayers was

assessed in the presence and absence of the known inhibitor of organic cation transport (Fauth et

al. 1988; Saito et al. 1992; Urakami et al. 2004; 2005), quinidine (Sigma-Aldrich Canada Ltd.).

Cell monolayers were incubated with medium (containing [ethyl-1-14

C] TEA (5 µmol/L)

(American Radiolabeled Chemicals Inc., St. Loius, Missouri, USA) in the presence or absence of

quinidine (1000 µmol/L). The transport of TEA was assessed as described above.

5.2.2b Acyclovir transport across cell monolayers

The transport of acyclovir across LLC-PK1 or HK-2 cell monolayers was assessed in the

presence or absence of quinidine. Cell monolayers were incubated with medium (containing [8-

14C] acyclovir (500 nmol/L) (American Radiolabeled Chemicals Inc.)) in the presence or absence

of quinidine (1000 µmol/L). The transport of acyclovir was assessed as described above.

5.2.2c The effect of acyclovir on creatinine transport across cell monolayers

The transport of creatinine was assessed across LLC-PK1 or HK-2 cell monolayers in the

presence or absence of acyclovir. Cell monolayers were incubated with medium (containing [2-

14C] creatinine (5 µmol/L) (American Radiolabeled Chemicals Inc.)) in the presence or absence

of quinidine (1000 µmol/L), cimetidine (1000 µmol/L) (Sigma-Aldrich Canada Ltd.) or

acyclovir (22 to 89 µmol/L) (Pharmacy at the Hospital for Sick Children). The acyclovir

90

concentrations used in the experiments are representative of concentrations of acyclovir that are

found in the plasma and hence, are the concentrations which creatinine may encounter in plasma.

5.2.3 Statistical analyses

Statistical analyses of the data were performed using IBM® SPSS

® Statistics version 19 software.

Statistical analyses were performed using ANOVA followed by Tukey’s HSD post hoc tests.

Statistical analyses were performed on substrate radioactivity (DPM) data. Data are presented as

the mean ± SE from 3 cell monolayer experiments. Data were considered statistically significant

if p< 0.05.

5.3 Results

5.3.1 TEA transport across LLC-PK1 and HK-2 cell monolayers

The TEA was transported across LLC-PK1 cell monolayers in a time – dependent manner over

the experimental study period (Figure 17). The results illustrate that there was a significant

(p<0.05) decrease in the concentration of [ethyl-14

C] TEA in the apical compartment in the

presence of quinidine at 30, 45 and 60 mins.

Our results illustrate that TEA was transported across HK-2 cell monolayers in a time –

dependent manner over the experimental period (Figure 18). The concentration of [ethyl-14

C]

TEA in the apical compartment was significantly (p < 0.05) decreased in the presence of

quinidine at 22.5 and 30 minutes.

91

Figure 17. Tetraethylammonium (TEA) transport across porcine renal proximal tubular cell

(LLC-PK1) monolayers. The transport (basolateral-to-apical) of TEA was assessed in LLC-PK1

cells monolayers. Cell monolayers were exposed to [ethyl-1-14

C] TEA (5 µmol/L) in the

presence or absence of quinidine (1000 µmol/L) for 60 mins. The transport of TEA was assessed

by measuring the appearance of [ethyl-1-14

C] TEA radioactivity in the apical compartment at

specific time intervals (0, 15, 30, 45 and 60 mins) for 60 mins. Radioactivity was measured as

disintegrations per minute (DPM). The TEA transport is expressed as the concentration of

[ethyl-1-14

C] TEA in the apical compartment. Results are presented as the mean (± standard

error (SE)) from 3 cell monolayer experiments. * p<0.05, compared to [ethyl-1-14

C] TEA

radioactivity in the apical compartment in the absence of quinidine.

*

**

0

50

100

150

200

250

300

0 15 30 45 60

[eth

yl-

1-1

4C

] T

EA

tra

nsp

ort

(nm

ol/

L)

time (mins)

TEA

TEA + quinidine

92

Figure 18. Tetraethylammonium (TEA) transport across human renal proximal tubular cell

(HK-2) monolayers. The transport (basolateral-to-apical) of TEA was assessed in HK-2 cells

monolayers. Cell monolayers were exposed to [ethyl-1-14

C] TEA (5 µmol/L) in the presence or

absence of quinidine (1000 µmol/L) for 30 mins. The transport of TEA was assessed by

measuring the appearance of [ethyl-1-14

C] TEA radioactivity in the apical compartment at

specific time intervals (0, 7.5, 15, 22.5 and 30 mins) for 30 mins. Radioactivity was measured as

disintegrations per minute (DPM). The TEA transport is expressed as the concentration of [ethyl-

1-14

C] TEA in the apical compartment. Results are presented as the mean (± standard error (SE))

from 3 cell monolayer experiments. * p<0.05, compared to [ethyl-1-14

C] TEA radioactivity in

the apical compartment in the absence of quinidine.

*

*

0

50

100

150

200

250

300

350

0 7.5 15 22.5 30

[eth

yl-

1-1

4C

] T

EA

tra

nsp

ort

(nm

ol/

L)

time (mins)

TEA

TEA + quinidine

93

5.3.2 Acyclovir transport across LLC-PK1 and HK-2 cell monolayers

Acyclovir appeared to be transported across LLC-PK1 cell monolayers in a time – dependent

manner from 30 to 60 mins (Figure 19). There was a trend of decreased concentration of [8-14

C]

acyclovir in the apical compartment in the presence of quinidine over the experimental study

period. Acyclovir transport was not significantly (p>0.05) inhibited in the presence of quinidine.

Acyclovir was transported across HK-2 cell monolayers in a time – dependent manner over the

experimental study period (Figure 20). Results illustrate that the concentration of [8-14

C]

acyclovir in the apical compartment was significantly (p<0.05) decreased in the presence of

quinidine at 15, 22.5 and 30 mins.

94

Figure 19. Acyclovir transport across porcine renal proximal tubular cell (LLC-PK1)

monolayers. The transport (basolateral-to-apical) of acyclovir was assessed in LLC-PK1 cells

monolayers. Cell monolayers were exposed to [8-14

C] acyclovir (5E-02 µmol/L) in the presence

or absence of quinidine (1000 µmol/L) for 60 mins. The transport of acyclovir was assessed by

measuring the appearance of [8-14

C] acyclovir radioactivity in the apical compartment at specific

time intervals (0, 15, 30, 45 and 60 mins) for 60 mins. Radioactivity was measured as

disintegrations per minute (DPM). Acyclovir transport is expressed as the concentration of [8-14

C] acyclovir in the apical compartment. Results are presented as the mean (± standard error

(SE)) from 3 cell monolayer experiments.

0

1

0 15 30 45 60

[8-1

4C

] a

cycl

ov

ir t

ran

spo

rt

(nm

ol/

L)

time (mins)

acyclovir

acyclovir + quinidine

95

Figure 20. Acyclovir transport across human renal proximal tubular cell (HK-2) monolayers.

The transport (basolateral-to-apical) of acyclovir was assessed in HK-2 cells monolayers. Cell

monolayers were exposed to [8-14

C] acyclovir (5E-02 µmol/L) in the presence or absence of

quinidine (1000 µmol/L) for 30 mins. The transport of acyclovir was assessed by measuring the

appearance of [8-14

C] acyclovir radioactivity in the apical compartment at specific time intervals

(0, 7.5, 15, 22.5 and 30 mins) for 30 mins. Radioactivity was measured as disintegrations per

minute (DPM). Acyclovir transport is expressed as the concentration of [8-14

C] acyclovir in the

apical compartment. Results are presented as the mean (± standard error (SE)) from 3 cell

monolayer experiments. * p<0.05, compared to [8-14

C] acyclovir radioactivity in the apical

compartment in the absence of quinidine.

** *

0

1

2

3

4

5

6

7

8

0 7.5 15 22.5 30

[8-1

4C

] a

cycl

ov

ir t

ran

spo

rt

(nm

ol/

L)

time (mins)

acyclovir

acyclovir + quinidine

96

5.3.3 The effect of acyclovir on creatinine transport across LLC-PK1 and HK-2 cell

monolayers

Figure 21 illustrates that in contrast to quinidine and cimetidine, acyclovir (22 to 89 µmol/L) did

not inhibit creatinine transport across LLC-PK1 cell monolayers. The concentration of [2-14

C]

creatinine in the apical compartment over the experimental study period was similar between cell

monolayers exposed to creatinine in the presence or absence of acyclovir (22 to 89 µmol/L). In

contrast, there was a decrease in the concentration of [2-14

C] creatinine in the apical

compartment in the presence of quinidine or cimetidine, compared to the concentration of [2-14

C]

creatinine in the apical compartment in the absence of quinidine or cimetidine. Creatinine

transport was significantly (p<0.05) inhibited in the presence of quinidine or cimetidine at 30 and

45 mins.

Figure 22 illustrates that in contrast to quinidine, acyclovir (22 to 89 µmol/L) did not inhibit

creatinine transport across HK-2 cell monolayers. The concentration of [2-14

C] creatinine in the

apical compartment over the experimental study period was similar between cell monolayers

exposed to creatinine in the presence or absence of acyclovir (22 to 89 µmol/L). In contrast, the

concentration of [2-14

C] creatinine was decreased in the apical compartment in the presence of

quinidine, compared to the concentration of [2-14

C] creatinine in the apical compartment in the

absence of quinidine. Creatinine transport was significantly (p<0.05) inhibited in the presence of

quinidine at 30 mins. The concentration of [2-14

C] creatinine appeared to be decreased in the

apical compartment in presence of cimetidine, compared to the concentration of [2-14

C]

creatinine in the apical compartment in the absence of cimetidine.

97

Figure 21. The effect of acyclovir on creatinine transport across porcine renal proximal tubular

cell (LLC-PK1) monolayers. The transport (basolateral-to-apical direction) of creatinine was

assessed in LLC-PK1 cells monolayers. Cell monolayers were exposed to [2-14

C] creatinine (5

µmol/L) in the presence or absence of quinidine (1000 µmol/L), cimetidine (1000 µmol/L) or

acyclovir (22 to 89 µmol/L) for 60 mins. The transport of creatinine was assessed by measuring

the appearance of [2-14

C] creatinine radioactivity in the apical compartment at specific time

intervals (0, 15, 30, 45 and 60 mins) for 60 mins. Radioactivity was measured as disintegrations

per minute (DPM). Creatinine transport is expressed as the concentration of [2-14

C] creatinine in

the apical compartment. Results are presented as the mean (± standard error (SE)) from 3 cell

monolayer experiments. * p<0.05, compared to [2-14

C] creatinine radioactivity in the apical

compartment in the absence of quinidine, cimetidine or acyclovir.

*

*

**

0

20

40

60

80

100

120

140

160

180

0 15 30 45 60

[2-1

4C

] cr

eati

nin

etr

an

spo

rt

(nm

ol/

L)

time (mins)

creatinine

creatinine + quinidine

creatinine + cimetidine

creatinine + acyclovir (22 µmol/L)

creatinine + acyclovir (44 µmol/L)

creatinine + acyclovir (67 µmol/L)

creatinine + acyclovir (89 µmol/L)

98

Figure 22. The effect of acyclovir on creatinine transport across human renal proximal tubular

cell (HK-2) monolayers. The transport (basolateral-to-apical) of creatinine was assessed in HK-

2 cells monolayers. Cell monolayers were exposed to [2-14

C] creatinine (5 µmol/L) in the

presence or absence of quinidine (1000 µmol/L), cimetidine (1000 µmol/L) or acyclovir (22 to

89 µmol/L) for 30 mins. The transport of creatinine was assessed by measuring the appearance

of [2-14

C] creatinine radioactivity in the apical compartment at specific time intervals (0, 7.5, 15,

22.5 and 30 mins) for 30 mins. Radioactivity was measured as disintegrations per minute

(DPM). Creatinine transport is expressed as the concentration of [2-14

C] creatinine in the apical

compartment. Results are presented as the mean (± standard error (SE)) from 3 cell monolayer

experiments. * p<0.05, compared to [2-14

C] creatinine radioactivity in the apical compartment in

the absence of quinidine, cimetidine or acyclovir.

*

0

50

100

150

200

250

300

350

400

450

500

0 7.5 15 22.5 30

[2-1

4C

] cr

eati

nin

etr

an

spo

rt

(nm

ol/

L)

time (mins)

creatinine

creatinine + quinidine

creatinine + cimetidine

creatinine + acyclovir (22 µmol/L)

creatinine + acyclovir (44 µmol/L

creatinine + acyclovir (67 µmol/L

creatinine + acyclovir (89 µmol/L)

99

5.4 Discussion

The objective of our study was to determine whether acyclovir inhibits creatinine transport. The

LLC-PK1 and HK-2 cell lines were employed as our in vitro models. The results suggest that

LLC-PK1 (Figure 17) and HK-2 (Figure 18) cells possess functional OCTs, thereby making

them appropriate models to study the renal tubular transport of organic cations such as creatinine

and acyclovir. In contrast to LLC-PK1 cells, the presence of functional OCTs in HK-2 cells has

not been previously reported. Hence, our study is the first to report that HK-2 cells possess

functional OCTs, thereby making them an invaluable in vitro model to study the renal tubular

transport of organic cations in humans.

Importantly, in contrast to quinidine (LLC-PK1 and HK-2) (Figures 19 and 20) or cimetidine

(LLC-PK1) (Figure 19), acyclovir did not inhibit creatinine transport across both types of cell

monolayers; suggesting that acyclovir does not affect the renal tubular handling of creatinine.

As previously explained; (1) the marked, transient increase in plasma creatinine observed in

some patients who received acyclovir (Table 3) is similar to that observed in some patients who

received non-nephrotoxic drugs that share similar renal tubular transport with creatinine and

hence compete with and subsequently inhibit creatinine secretion (Berglund et al. 1975;

Blackwood et al. 1976; Burgess et al. 1982; Burry and Dieppe 1976; Dubb et al. 1978; Dutt et al.

1981; Haggie et al. 1976; Kastrup et al. 1985; Myre et al. 1987; Opravil et al. 1993; Tschuppert

et al. 2007) and (2) acyclovir may share similar renal tubular transport mechanisms with

creatinine (Eisner et al. 2010; Okuda et al. 2006; Takeda et al. 2002; Urakami et al. 2004).

Hence, if this is the case, it is possible that our results illustrate that acyclovir did not inhibit the

tubular transport of creatinine for the following reasons:

100

First, as reviewed by Andreev et al. (Andreev et al. 1999), some drugs, such as phenacemide and

vitamin D derivatives induce a marked, transient increase in plasma creatinine in the absence of

other significant signs of renal impairment by other less well understood mechanisms, including

interference with the Jaffé-based assay for creatinine measurement and modification of the

production rate and release of creatinine, respectively. Thus, acyclovir may affect plasma

creatinine levels by a yet unknown mechanism(s).

Second, based on our results, it can be argued that acyclovir did not inhibit creatinine transport

across LLC-PK1 cell monolayers because in contrast to creatinine (Figure 21), the OCT pathway

in the LLC-PK1 cells did not appear to play a significant role in acyclovir transport (Figure 19),

and hence acyclovir was unlikely to compete with and subsequently inhibit creatinine transport

via the OCT pathway present in the cells. Furthermore interspecies differences in drug

disposition (Eaton and Klaassen 2001; Riddick 1998) and protein expression (Mersch-

Sundermann et al. 2004) for instance, may provide an explanation for the lack of inhibition of

creatinine transport by acyclovir in LLC-PK1 cells. For example, the degree of amino acid

sequence similarity between porcine OCT1 (pOCT1) and hOCT1 is approximately 78 % (NCBI

Unigene 2009a), while porcine OCT2 (pOCT2) and hOCT2 share approximately 86 % amino

acid sequence homology (NCBI Unigene 2009b).

However, in contrast to the results obtained in LLC-PK1 cells, the OCT pathway in human HK-2

cells played a significant role in both acyclovir (Figure 20) and creatinine transport (Figure 22),

yet similar to the results obtained in LLC-PK1 cells, acyclovir did not inhibit creatinine transport

in human HK-2 cells. The results from previous studies suggest that the OCTs may mediate the

renal tubular transport of both creatinine (Okuda et al. 2006; Urakami et al. 2004) and acyclovir

(Takeda et al. 2002). However, while OCT2 appears to be primarily responsible for creatinine

101

transport (Okuda et al. 2006; Urakami et al. 2004), it appears that OCT1 may be predominantly

accountable for acyclovir transport (Takeda et al. 2002). Reviewed by Dresser et al. (Dresser et

al. 2001), OCT1 and OCT2 are both located in the human kidney, therefore it is possible that

renal secretion of creatinine and acyclovir may be mediated by different OCTs; OCT2 and

OCT1, respectively. Thus, acyclovir may not impede creatinine tubular transport in vitro and

possibly in vivo, in humans as well.

The knowledge that OCT1, rather than OCT2, mediate acyclovir transport may also provide an

explanation for the insignificant transport of acyclovir across LLC-PK1 cells (Figure 19). In

contrast to OCT2 (Grundemann et al. 1997), OCT1 has not been specifically identified in LLC-

PK1 cells. The LLC-PK1 cells may lack or have reduced expression of OCT1. Therefore, LLC-

PK1 cells may be unable to transport acyclovir via their existing OCT system, and hence may be

an inappropriate model to examine acyclovir transport via the same. Furthermore, if the

plausible lack of or reduced OCT1 expression in LLC-PK1 cells resulted in the absence of

significant acyclovir transport across the cell monolayers (Figure 19), then the results provide

additional support for the postulation that acyclovir and creatinine may be transported via

different OCTs.

Third, we employed in vitro models in our studies. Although in vitro models are widely used in

pharmacology and toxicology studies to address questions at both the cellular and molecular

level, there are several major disadvantages of in vitro models that limit their ability to accurately

predict responses in vivo (Davila et al. 1998; Zucco et al. 2004). Major disadvantages include

disruption of cellular structural integrity and intercellular relationships, the production of

artifactual drug binding sites that does not normally exist in vivo, differences between in vitro

and in vivo drug pharmacokinetics and altered protein expression (Davila et al. 1998).

102

Therefore, the transport of creatinine and/or acyclovir in vitro may be altered from its transport

in vivo, in humans.

In our study, we investigated the possible interaction between creatinine and acyclovir at the

OCT pathway. However, it is also possible that the interaction between creatinine and acyclovir

may be occurring at the OAT pathway, rather than at the OCT pathway. Results from studies

suggest that the OAT system may play a fundamental role in both creatinine (Arendshorst and

Selkurt 1970; Burry and Dieppe 1976; Eisner et al. 2010) and acyclovir (Takeda et al. 2002)

transport. The LLC-PK1 cells do not possess OATs (Hori et al. 1993; Mertens et al. 1988), and

therefore are an inappropriate in vitro model to study the possible interaction between creatinine

and acyclovir at the OAT pathway. The expression of functional OATs in HK-2 cells is

currently unknown and we did not determine the same in our study. However, if functional

OATs are expressed in HK-2 cells, and both creatinine and acyclovir were significantly

transported by the same OAT(s), then, in the presence of acyclovir, decreased creatinine

transport across the cell monolayers would have likely been observed. Alternatively, as

suggested for OCTs, creatinine and acyclovir may have been transported by different OATs

expressed in the HK-2 cells, such that acyclovir did not hinder creatinine transport via the OAT

pathway.

Engaging both animal (LLC-PK1) and human (HK-2) cell models, we illustrated that acyclovir

did not inhibit creatinine transport. Taken together, the results suggest that acyclovir does not

affect the renal tubular transport of creatinine, in vitro and possibly, in vivo, in humans as well.

Therefore, the pronounced, transient elevation in plasma creatinine observed in some children

may be solely due to decreased GFR as a result of renal dysfunction induced by acyclovir, and

not due to a spurious acyclovir-creatinine interaction on the tubular level.

103

5.5 Acknowledgements

The study was supported by a grant from CIHR.

5.6 Statement of significance

These are the first experimental results which show that acyclovir does not inhibit the tubular

transport of creatinine in vitro, and possibly, in vivo. Thus, the marked, transient elevation in

plasma creatinine levels observed in some patients may be solely due to decreased GFR as a

result of acyclovir – induced nephrotoxicity, and not due the inhibition of creatinine secretion by

acyclovir.

104

5.7 References

NCBI Unigene. 2009a. Organic cation transporter 1 (OCT1).

[http:www.ncbi.nlm.gov/UniGene/clus.cgi?ORG=Ssc&CID=23507&itool=HomolGeneMainRep

ort].

NCBI Unigene. 2009b. Solute carrier family 22 (organic cation transporter), member 2

(SLC22A2).

[http://www.ncbi.nlm.nih.gov/UniGene/clus.cgi?UGID=454108&TAXID=9823&SEARCH=org

anic cation transporter 2].

Ahmad, T., Simmonds, M., McIver, A.G., and McGraw, M.E. 1994. Reversible renal failure in

renal transplant patients receiving oral acyclovir prophylaxis. Pediatr Nephrol 8: 489-491.

Andreev, E., Koopman, M., and Arisz, L. 1999. A rise in plasma creatinine that is not a sign of

renal failure: which drugs can be responsible? J Intern Med 246: 247-252.

Arendshorst, W.J., and Selkurt, E.E. 1970. Renal tubular mechanisms for creatinine secretion in

the guinea pig. Am J Physiol 218: 1661-1670.

Berglund, F., Killander, J., and Pompeius, R. 1975. Effect of trimethoprim-sulfamethoxazole on

the renal excretion of creatinine in man. J Urol 114: 802-808.

Bianchetti, M.G., Roduit, C., and Oetliker, O.H. 1991. Acyclovir-induced renal failure: course

and risk factors. Pediatr Nephrol 5: 238-239.

Blackwood, W.S., Maudgal, D.P., Pickard, R.G., Lawrence, D., and Northfield, T.C. (1976).

Cimetidine in duodenal ulcer. Controlled trial. Lancet 2: 174-176.

Brigden, D., Rosling, A.E., and Woods, N.C. 1982. Renal function after acyclovir intravenous

injection. Am J Med 73: 182-185.

Bryson, Y.J. 1984. The use of acyclovir in children. Pediatr Infect Dis 3: 345-348.

Burgess, E., Blair, A., Krichman, K., and Cutler, R.E. 1982. Inhibition of renal creatinine

secretion by cimetidine in humans. Ren Physiol 5: 27-30.

Burry, H.C., and Dieppe, P.A. 1976. Apparent reduction of endogenous creatinine clearance by

salicylate treatment. Br Med J 2: 16-17.

Chou, J.W., Yong, C., and Wootton, S.H. 2008. Case 2: Rash, fever and headache....first, do no

harm. Paediatr Child Health 13: 49-52.

Davila, J.C., Rodriguez, R.J., Melchert, R.B., and Acosta, D., Jr. 1998. Predictive value of in

vitro model systems in toxicology. Annu Rev Pharmacol Toxicol 38: 63-96.

105

Dresser, M.J., Leabman, M.K., and Giacomini, K.M. 2001. Transporters involved in the

elimination of drugs in the kidney: organic anion transporters and organic cation transporters. J

Pharm Sci 90: 397-421.

Dubb, J.W., Stote, R.M., Familiar, R.G., Lee, K., and Alexander, F. 1978. Effect of cimetidine

on renal function in normal man. Clin Pharmacol Ther 24: 76-83.

Dutt, M.K., Moody, P., and Northfield, T.C. 1981. Effect of cimetidine on renal function in man.

Br J Clin Pharmacol 12: 47-50.

Eaton, D.L., and Klaassen, C.D. 2001. Principles of Toxicology. In Casarett & Doull's

Toxicology, the basic science of poisons. Edited by Klassen, C.D. The Mc-Graw Hill

Companies, New York. pp 3-34.

Eisner, C., Faulhaber-Walter, R., Wang, Y., Leelahavanichkul, A., Yuen, P.S., Mizel, D., Star,

R.A., Briggs, J.P., Levine, M., and Schnermann, J. 2010. Major contribution of tubular secretion

to creatinine clearance in mice. Kidney Int 77: 519-526.

Fauth, C., Rossier, B., and Roch-Ramel, F. 1988. Transport of tetraethylammonium by a kidney

epithelial cell line (LLC-PK1). Am J Physiol 254: F351-357.

Genc, G., Ozkaya, O., Acikgoz, Y., Yapici, O., Bek, K., Gulnar Sensoy, S., and Ozyurek, E.

2010. Acute renal failure with acyclovir treatment in a child with leukemia. Drug Chem Toxicol

33: 217-219.

Grundemann, D., Babin-Ebell, J., Martel, F., Ording, N., Schmidt, A., and Schomig, E. 1997.

Primary structure and functional expression of the apical organic cation transporter from kidney

epithelial LLC-PK1 cells. J Biol Chem 272: 10408-10413.

Grundemann, D., Gorboulev, V., Gambaryan, S., Veyhl, M., and Koepsell, H. 1994. Drug

excretion mediated by a new prototype of polyspecific transporter. Nature 372: 549-552.

Gunness, P., Aleksa, K., Kousage, K., Ito, S. and Koren, G. 2010. Comparison of the novel HK-2

human renal proximal tubular cell line to the standard LLC-PK1 cell line in studying drug-

induced nephrotoxicity. Can J Physiol Pharmacol 88: 448-455.

Haggie, S.J., Fermont, D.C., and Wyllie, J.H. 1976. Treatment of duodenal ulcer with cimetidine.

Lancet 1: 983-984.

Hori, R., Okamura, M., Takayama, A., Hirozane, K., and Takano, M. 1993. Transport of organic

anion in the OK kidney epithelial cell line. Am J Physiol 264: F975-980.

Kastrup, J., Petersen, P., Bartram, R., and Hansen, J.M. 1985. The effect of trimethoprim on

serum creatinine. Br J Urol 57: 265-268.

106

Keeney, R.E., Kirk, L.E., and Bridgen, D. 1982. Acyclovir tolerance in humans. Am J Med 73:

176-181.

Mersch-Sundermann, V., Knasmuller, S., Wu, X.J., Darroudi, F., and Kassie, F. 2004. Use of a

human-derived liver cell line for the detection of cytoprotective, antigenotoxic and cogenotoxic

agents. Toxicology 198: 329-340.

Mertens, J.J., Weijnen, J.G., van Doorn, W.J., Spenkelink, B., Temmink, J.H., and van Bladeren,

P.J. 1988. Differential toxicity as a result of apical and basolateral treatment of LLC-PK1

monolayers with S-(1,2,3,4,4-pentachlorobutadienyl)glutathione and N-acetyl-S-(1,2,3,4,4-

pentachlorobutadienyl)-L-cysteine. Chem Biol Interact 65: 283-293.

Myre, S.A., McCann, J., First, M.R., and Cluxton, R.J., Jr. 1987. Effect of trimethoprim on

serum creatinine in healthy and chronic renal failure volunteers. Ther Drug Monit 9: 161-165.

Okuda, M., Kimura, N., and Inui, K. 2006. Interactions of fluoroquinolone antibacterials, DX-

619 and levofloxacin, with creatinine transport by renal organic cation transporter hOCT2. Drug

Metab Pharmacokinet 21: 432-436.

Opravil, M., Keusch, G., and Luthy, R. 1993. Pyrimethamine inhibits renal secretion of

creatinine. Antimicrob Agents Chemother 37: 1056-1060.

Perantoni, A., and Berman, J.J. 1979. Properties of Wilms' tumor line (TuWi) and pig kidney

line (LLC-PK1) typical of normal kidney tubular epithelium. In Vitro 15: 446-454.

Potter, J.L., and Krill, C.E., Jr. 1986. Acyclovir crystalluria. Pediatr Infect Dis 5, 710-712.

Riddick, D.S. 1998. Drug Biotransformation. In Principles of Medical Pharmacology. Edited by

Kalant, H., and Roschlau, W.H.E. Oxford University Press, New York. pp 38-54.

Saito, H., Yamamoto, M., Inui, K., and Hori, R. 1992. Transcellular transport of organic cation

across monolayers of kidney epithelial cell line LLC-PK1. Am J Physiol 262: C59-66.

Sawyer, M.H., Webb, D.E., Balow, J.E., and Straus, S.E. 1988. Acyclovir-induced renal failure.

Clinical course and histology. Am J Med 84; 1067-1071.

Schreiber, R., Wolpin, J., and Koren, G. 2008. Determinants of aciclovir-induced nephrotoxicity

in children. Paediatr Drugs 10: 135-139.

Takeda, M., Khamdang, S., Narikawa, S., Kimura, H., Kobayashi, Y., Yamamoto, T., Cha, S.H.,

Sekine, T., and Endou, H. 2002. Human organic anion transporters and human organic cation

transporters mediate renal antiviral transport. J Pharmacol Exp Ther 300: 918-924.

Toto, R.D. 1995. Conventional measurement of renal function utilizing serum creatinine,

creatinine clearance, inulin and para-aminohippuric acid clearance. Curr Opin Nephrol

Hypertens 4: 505-509

107

Tschuppert, Y., Buclin, T., Rothuizen, L.E., Decosterd, L.A., Galleyrand, J., Gaud, C., and

Biollaz, J. 2007. Effect of dronedarone on renal function in healthy subjects. Br J Clin Pharmacol

64: 785-791.

Urakami, Y., Kimura, N., Okuda, M., and Inui, K. 2004. Creatinine transport by basolateral

organic cation transporter hOCT2 in the human kidney. Pharm Res 21: 976-981.

Urakami, Y., Kimura, N., Okuda, M., Masuda, S., Katsura, T., and Inui, K. 2005. Transcellular

transport of creatinine in renal tubular epithelial cell line LLC-PK1. Drug Metab Pharmacokinet

20: 200-205.

Vachvanichsanong, P., Patamasucon, P., Malagon, M., and Moore, E.S. 1995. Acute renal failure

in a child associated with acyclovir. Pediatr Nephrol 9: 346-347.

Vomiero, G., Carpenter, B., Robb, I., and Filler, G. 2002. Combination of ceftriaxone and

acyclovir - an underestimated nephrotoxic potential? Pediatr Nephrol 17: 633-637.

Zucco, F., De Angelis, I., Testai, E., and Stammati, A. 2004. Toxicology investigations with cell

culture systems: 20 years after. Toxicol In Vitro 18: 153-163.

108

5.8 Additional experiments not published

5.8.1 The paracellular flux (basolateral-to-apical) of D-[1-3H(N)] mannitol

As mentioned earlier in the chapter (section 5.2.2), the paracellular flux (basolateral-to-apical) of

D-[1-3H(N)] mannitol across the LLC-PK1 or HK-2 cell monolayers to assess the integrity of

cell monolayers. A priori decision was made to eliminate the results from any cell monolayers

where the paracellular flux of D-[1-3H(N)] mannitol across LLC-PK1 or HK-2 cell monolayers

was greater than 5 % over the respective experimental period.

5.8.2 Materials and methods

The radioactivity of D-[1-3H(N)] mannitol was assessed as previously described (section 5.2.2).

5.8.3 Results

The paracellular flux (basolateral-to-apical) of D-[1-3H(N)] mannitol across LLC-PK1 or HK-2

cell monolayers was not greater than 5 % over the respective experimental time periods [LLC-

PK1:60 mins; HK-2: 30 mins].

109

Figure 23. The paracellular flux of mannitol across procine renal proximal tubular cell (LLC-

PK1) monolayers that were used for determining the transepithelial transport of

tetraethylammonium (TEA) across the cell monolayers. Cell monolayers were exposed to D-[1-3H(N)] mannitol (10 nmol/L) for 60 mins. The paracellular flux of mannitol was assessed by

measuring the appearance of D-[1-3H(N)] mannitol radioactivity in the apical compartment at

specific time intervals (0, 15, 30, 45 and 60 mins) for 60 mins. Radioactivity was measured as

disintegrations per minute (DPM). The paracellular flux of mannitol is expressed as the percent

(%) of radioactivity on the apical side at the respective time interval, compared to the basolateral

side at time zero. Results are presented as the mean ± standard error (SE) from 3 cell monolayer

experiments.

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0 15 30 45 60

D-[

1-3

H(N

)] m

an

nit

ol

pa

race

llu

lar

flu

x

(% o

f ra

dio

act

ivit

y o

n t

he

ap

ica

l si

de

at

the

resp

ecti

ve

tim

e in

terv

al

co

mp

are

d t

o t

he

ba

sola

tera

lsi

de

at

tim

e

zero

)

time (mins)

TEA

TEA + quinidine

110

Figure 24. The paracellular flux of mannitol across human renal proximal tubular cell (HK-2)

monolayers that were used for determining the transepithelial transport of tetraethylammonium

(TEA) across the cell monolayers. Cell monolayers were exposed to D-[1-3H(N)] mannitol (10

nmol/L) for 30 mins. The paracellular flux of mannitol was assessed by measuring the

appearance of D-[1-3H(N)] mannitol radioactivity in the apical compartment at specific time

intervals (0, 7.5, 15, 22.5 and 30 mins) for 30 mins. Radioactivity was measured as

disintegrations per minute (DPM). The paracellular flux of mannitol is expressed as the percent

(%) of radioactivity on the apical side at the respective time interval, compared to the basolateral

side at time zero. Results are presented as the mean ± standard error (SE) from 3 cell monolayer

experiments.

-0.50

0.00

0.50

1.00

1.50

2.00

2.50

3.00

3.50

4.00

4.50

0 7.5 15 22.5 30

D-[

1-3

H(N

)] m

an

nit

ol

pa

race

llu

lar

flu

x

(% o

f ra

dio

act

ivit

y o

n t

he

ap

ica

l si

de

at

the

resp

ecti

ve

tim

e in

terv

al

co

mp

are

d t

o t

he

ba

sola

tera

l si

de

at

tim

e

zero

)

time (mins)

TEA

TEA + quinidine

111

Figure 25. The paracellular flux of mannitol across procine renal proximal tubular cell (LLC-

PK1) monolayers that were used for determining the transepithelial transport of acyclovir across

the cell monolayers. Cell monolayers were exposed to D-[1-3H(N)] mannitol (10 nmol/L) for 60

mins. The paracellular flux of mannitol was assessed by measuring the appearance of D-[1-3H(N)] mannitol radioactivity in the apical compartment at specific time intervals (0, 15, 30, 45

and 60 mins) for 60 mins. Radioactivity was measured as disintegrations per minute (DPM).

The paracellular flux of mannitol is expressed as the percent (%) of radioactivity on the apical

side at the respective time interval, compared to the basolateral side at time zero. Results are

presented as the mean ± standard error (SE) from 3 cell monolayer experiments.

-0.20

0.00

0.20

0.40

0.60

0.80

1.00

1.20

0 15 30 45 60

D-[

1-3

H(N

)] m

an

nit

ol

pa

race

llu

lar

flu

x

(% o

f ra

dio

act

ivit

y o

n t

he

ap

ica

l si

de

at

the

resp

ecti

ve

tim

e in

terv

al

co

mp

are

d t

o t

he

ba

sola

tera

l si

de

at

tim

e

zero

)

time (mins)

acyclovir

acyclovir + quinidine

112

Figure 26. The paracellular flux of mannitol across human renal proximal tubular cell (HK-2)

monolayers that were used for determining the transepithelial transport of acyclovir across the

cell monolayers. Cell monolayers were exposed to D-[1-3H(N)] mannitol (10 nmol/L) for 30

mins. The paracellular flux of mannitol was assessed by measuring the appearance of D-[1-3H(N)] mannitol radioactivity in the apical compartment at specific time intervals (0, 7.5, 15,

22.5 and 30 mins) for 30 mins. Radioactivity was measured as disintegrations per minute

(DPM). The paracellular flux of mannitol is expressed as the percent (%) of radioactivity on the

apical side at the respective time interval, compared to the basolateral side at time zero. Results

are presented as the mean ± standard error (SE) from 3 cell monolayer experiments.

-0.50

0.00

0.50

1.00

1.50

2.00

2.50

3.00

3.50

4.00

4.50

5.00

0 7.5 15 22.5 30

D-[

1-3

H(N

)] m

an

nit

ol

pa

race

llu

lar

flu

x

(% o

f ra

dio

act

ivit

y o

n a

pic

al

sid

e a

t th

e re

spec

tiv

e ti

me

inte

rva

l co

mp

are

d t

o t

he

ba

sola

tera

lsi

de

at

tim

e ze

ro)

time (mins)

acyclovir

acyclovir + quinidine

113

Figure 27. The paracellular flux of mannitol across procine renal proximal tubular cell (LLC-

PK1) monolayers that were used to determine acyclovir inhibits the tubular transport of

creatinine. Cell monolayers were exposed to D-[1-3H(N)] mannitol (10 nmol/L) for 60 mins.

The paracellular flux of mannitol was assessed by measuring the appearance of D-[1-3H(N)]

mannitol radioactivity in the apical compartment at specific time intervals (0, 15, 30, 45 and 60

mins) for 60 mins. Radioactivity was measured as disintegrations per minute (DPM). The

paracellular flux of mannitol is expressed as the percent (%) of radioactivity on the apical side at

the respective time interval, compared to the basolateral side at time zero. Results are presented

as the mean ± standard error (SE) from 3 cell monolayer experiments.

-0.30

-0.20

-0.10

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

0 15 30 45 60

D-[

1-3

H(N

)] m

an

nit

ol

pa

race

llu

lar

flu

x

(% o

f ra

dio

act

ivit

y o

n t

he

ap

ica

l si

de

at

the

resp

ecti

ve

tim

e in

terv

al

co

mp

are

d t

o t

he

ba

sola

tera

l si

de

at

tim

e

zero

)

time (mins)

creatinine

creatinine + quinidine

creatinine + cimetidine

creatinine + acyclovir (22 µmol/L)

creatinine + acyclovir (44 µmol/L)

creatinine + acyclovir (67 µmol/L)

creatinine + acyclovir (89 µmol/L)

114

Figure 28. The paracellular flux of mannitol across human renal proximal tubular cell (HK-2)

monolayers that were used to determine acyclovir inhibits the tubular transport of creatinine.

Cell monolayers were exposed to D-[1-3H(N)] mannitol (10 nmol/L) for 30 mins. The

paracellular flux of mannitol was assessed by measuring the appearance of D-[1-3H(N)] mannitol

radioactivity in the apical compartment at specific time intervals (0, 7.5, 15, 22.5 and 30 mins)

for 30 mins. Radioactivity was measured as disintegrations per minute (DPM). The paracellular

flux of mannitol is expressed as the percent (%) of radioactivity on the apical side at the

respective time interval, compared to the basolateral side at time zero. Results are presented as

the mean ± standard error (SE) from 3 cell monolayer experiments.

0.00

0.50

1.00

1.50

2.00

2.50

3.00

3.50

4.00

4.50

5.00

0 7.5 15 22.5 30

D-[

1-3

H(N

)] m

an

nit

ol

pa

race

llu

lar

flu

x

(% o

f ra

dio

act

ivit

y o

n t

he

ap

ica

l si

de

at

the

resp

ecti

ve

tim

e in

terv

al

co

mp

are

d t

o t

he

ba

sola

tera

l si

de

at

tim

e

zero

)

time (mins)

creatinine

creatinine + quinidine

creatinine + cimitidine

creatinine + acyclovir (22 µmol/L)

creatinine + acyclovir (44 µmol/L)

creatinine + acyclovir (67 µmol/L)

creatinine + acyclovir (89 µmol/L)

115

Chapter 6

Acyclovir is a substrate for the human breast cancer resistance

protein (BCRP/ABCG2): implications for renal tubular transport

and acyclovir – induced nephrotoxicity

Patrina Gunness,a,b

Katarina Aleksa,a,c

Gideon Korena,b

aDivision of clinical Pharmacology and Toxicology, The Hospital for Sick Children, 555

University Avenue, Toronto, ON, M5G 1X8, Canada

bGraduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University

of Toronto, ON, M5S 3M2, Canada

cSchool of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, Ontario,

N2L 3G1, Canada

This article has been accepted for publication: Gunness, P., Aleksa, K., and Koren, G. 2011.

Acyclovir is a substrate for the human breast cancer resistance protein (BCRP/ABCG2):

implications for renal tubular transport and acyclovir – induced nephrotoxicity. Can J Physiol

Pharmacol. [In press]. This article will be originally published by NRC Research Press.

[PG performed all experiments and prepared the manuscript for submission]

116

6.1 Abstract

The human BCRP is widely expressed in human tissues, including the kidney. In mice, Bcrp1

(murine BCRP ortholog) mediates the transport of acyclovir into breast milk. It is plausible that

acyclovir is also a substrate for human BCRP. The objective of the study was to determine

whether acyclovir is a substrate for human BCRP. Transfected human embryonic kidney

(HEK293) cells [containing the wildtype ABCG2 gene] were exposed to [8-14

C] acyclovir (1

µM) in the presence or absence of the BCRP inhibitor, fumitremorgin C (FTC). Intracellular

acyclovir accumulation was assessed using a liquid scintillation counter. Co-exposure to FTC

resulted in a significant (5-fold) increase in the intracellular accumulation of acyclovir. The

results suggest that acyclovir is a substrate for the human BCRP. The study is the first to provide

direct evidence for the role of human BCRP in acyclovir transport and its potential significance

with respect to renal tubular transport of acyclovir and the direct renal tubular insult induced by

the drug.

6.2 Introduction

The human BCRP is the second member of the subfamily G of the human ABC transporter

superfamily (Dean et al. 2001; Mao and Unadkat 2005; Robey et al. 2009). The 72-kDa protein

(Mao and Unadkat 2005) is an efflux transporter (Doyle et al. 1998; Rocchi et al. 2000)

responsible for the transport of both endogenous and exogenous substrates (Doyle et al. 1998;

Ozvegy et al. 2001). The protein is widely expressed in human tissues, including the placenta,

liver (Allikmets et al. 1988; Doyle et al. 1998; Maliepaard et al. 2001) and kidney (Huls et al.

2008).

117

In mice, it was illustrated that Bcrp1 (murine BCRP ortholog) is responsible for the transport of

acyclovir into breast milk (Jonker et al. 2005). Compared to female mice with functional Bcrp1,

the accumulation of acyclovir in breast milk was significantly less in mice with non-functional

Bcrp1. The results suggested that acyclovir is a substrate for mice Bcrp1 and hence, it may also

be a substrate for human BCRP. However, interspecies differences exist between murine Bcrp1

and human BCRP with respect to amino acid sequences, tissue expression (Allen et al. 1999) and

function (Gonzalez-Lobato et al. 2010). Therefore, the results presented by Jonker and

colleagues cannot be used to definitively conclude that acyclovir is a substrate for human BCRP.

The role of human BCRP in the transport of acyclovir has not been previously investigated. We

hypothesized that acyclovir is a substrate for human BCRP. Our group has been interested in the

study of the pathogenesis of acyclovir – induced nephrotoxicity in children, particularly the

direct renal tubular insult that is induced by the drug. Therefore, in this present study, the

relevance of our results to the renal tubular transport of acyclovir and the direct renal tubular

injury induced by the drug will be discussed.

The antiviral agent, acyclovir may cause severe nephrotoxicity, which can often lead to acute

renal failure in patients (Ahmad et al. 1994; Bianchetti et al. 1991; Brigden et al. 1982; Chou et

al. 2008; Genc et al. 2010; Keeney et al. 1982; Vachvanichsanong et al. 1995; Vomiero et al.

2002). It has long been believed that acyclovir – induced nephrotoxicity is secondary to

crystalluria (Bianchetti et al. 1991; Lyon et al. 2002; Mason and Nickols 2008; Peterslund et al.

1988; Sawyer et al. 1988). However, clinical evidence of nephrotoxicity in the absence of

crystalluria suggests that the drug induces direct insult to renal tubular cells (Ahmad et al. 1994;

Vomiero et al. 2002). Employing both porcine and human renal proximal tubular cells exposed

to concentrations of acyclovir that may be encountered in clinical practice (Hintz et al. 1982), we

118

recently provided the first experimental evidence to support the above clinical evidence

(Gunness et al. 2010). Elucidation of the renal tubular transport mechanisms of acyclovir is

critical for understanding the etiology of the direct renal tubular insult induced by the antiviral

agent. Research has shown that the human BCRP transporter is localized to the apical membrane

of renal tubular cells (Huls et al. 2008). Therefore, if acyclovir is a substrate for human BCRP

and the transporter plays a significant role in renal transport of acyclovir, then reduced or

abolished function of the efflux transporter may result in the increased intracellular accumulation

of acyclovir and subsequent detrimental nephrotoxic consequences, such as direct injury to renal

tubular cells.

6.3 Materials and methods

6.3.1 Cell culture

Stably transfected human embryonic kidney (HEK293) cells containing the full-length human

ABCG2 gene encoding the wildtype ABCG2 amino acid sequence were used as the in vitro

model (Dr. Robert W. Robey, National Institutes of Health (NIH), Bethesda, Maryland, USA).

The cells were maintained as described by Morisaki and colleagues (Morisaki et al. 2005). The

expression of the ABCG2 protein was enforced by selection in G418 (Invitrogen Canada Inc.).

The cells were cultured in Eagles’s Minimum Essential Medium (EMEM) (ATCC)

supplemented with FBS (10 %), penicillin (100 Units/mL), streptomycin (100 µg/mL) and G418

(2 mg/mL). Cells were maintained at 37°C in a sterile, humidified atmosphere of 5 % CO2 and

95 % O2. All experiments were conducted on cell monolayers that were grown to 85 – 90 %

confluence. Hereinafter, these cells will be referred to as ‘overexpressing HEK293 cells’.

119

6.3.2 Determination of protein expression of human BCRP in overexpressing HEK293

cells

Qualitative western blots assays were conducted to confirm the protein expression of human

BCRP in the overexpressing HEK293 cells. Whole cell lysate was used for western blot assays.

Human placenta and mock HEK293 cells [transfected with the empty PC DNA 3.1 vector] were

used as the positive and negative control, respectively, for western blot assays. The human

placenta is known to express high levels of BCRP (Allikmets et al. 1998; Doyle et al. 1998;

Maliepaard et al. 2001). The human placenta tissue was obtained from Mount Sinai Hospital

(Toronto, Ontario, Canada). Mock HEK293 cells were also obtained from Dr. Robert W. Robey.

Total protein for western blot assays was quantified using the Bradford reagent (Sigma-Aldrich

Canada Ltd.).

6.3.3 Whole cell lysate for western blot assays

6.3.3a Mock or overexpressing HEK293 cells

The media from cell monolayers was removed and the cells were washed (2X) with ice-cold

PBS. Cell monolayers were scraped in a modified lysis buffer ([50 mmol/L Tris-HCL (pH7.4), 1

% (v/v) NP-40, 0.25 % (w/v) sodium deoxycholate, 150 mmol/L NaCl, 1 mmol/L EDTA, 1

mmol/L PMSF, 1 µg/mL aprotonin, 1 µg/mL leupeptin, 1 µg/mL pepstatin] (Millipore 2007).

The cell homogenate was centrifuged at 600 x g for 20 mins at 4◦C. The pellet was discarded

and the supernatant (whole cell lysate) was stored at -80◦C until analyses.

120

6.3.3b Human placenta tissue

Human placenta tissue (1 gram) was washed (2X) with ice-cold PBS. The tissue was

homogenized using a polytron homogenizer. The tissue was homogenized in the modified lysis

buffer described above for the HEK293 cells. The homogenate was centrifuged at 600 x g for 20

mins at 4◦C. The pellet was discarded and the supernatant (whole cell lysate) was stored at -80

◦C

until analyses.

6.3.4 Western blot assay

For electrophoresis samples, total protein was mixed with 2X Laemmli buffer (Laemmli 1970).

Total protein from human placenta (20 µg), HEK293 [transfected with the empty PC DNA 3.1

vector] (50 µg) or overexpressing HEK293 (50 µg) cells were resolved on a 10 % SDS-PAGE.

Resolved proteins were transferred unto Hybond™

- P PVDF membranes at 100 V for 1 hour in

transfer buffer [25 mmol/L Tris, 192 mmol/L glycine, 20 % (v/v) methanol, pH 8.3] (Towbin et

al. 1979). Blots were blocked in 5 % (w/v) skim milk overnight at 4◦C. Blots were then washed

in 5 % milk and subsequently incubated with mouse BCRP antibody (BXP-21, Kamiya

Biomedical Company, Seattle, Washington, USA) in 5 % skim milk overnight at 4◦C. The

primary antibody was diluted 1:120 for use. Following the incubation, the primary antibody was

removed and blots were washed in 5 % skim milk. Blots were subsequently incubated with goat

anti-mouse IgG-HRP antibody (sc-2005, Santa Cruz Biotechnology, Inc.) in 5 % skim milk for 2

hours at room temperature. The secondary antibody was diluted 1:5000 for use. The secondary

antibody was removed and the blots were washed sequentially in 5 % skim milk, PBST and PBS

solutions. Blots were developed using Western Lightning® Plus-ECL. Blots were exposed to

Kodak™

BioMax Light Film. The western blot assay was conducted in duplicates.

121

6.3.5 Hoescht 33342 dye efflux assay

The functional activity of the human BCRP in overexpressing HEK293 cells was assessed using

the Hoechst 33342 efflux assay. The assay was performed as described by Brown and

colleagues (Brown et al. 2008) with modification. The media from cell monolayers was removed

and cells were washed (2X) with warm Kreb’s buffer. Cell monolayers were subsequently

incubated in Kreb’s solution containing Hoechst 33342 (1 µmol/L) in the presence or absence of

the BCRP inhibitor, FTC (10 µmol/L) for 90 mins at 37◦C. Following the incubation period, the

cell monolayers were washed (3X) with ice-cold Kreb’s solution. The cell monolayers were then

further incubated in Kreb’s solution in the presence or absence of FTC for an additional 30 mins

at 37◦C. At the end of the incubation period, cell monolayers were washed (2X) with ice-cold

Kreb’s buffer and subsequently lysed using 1 % (v/v) Triton-X (Sigma-Aldrich Canada Ltd.).

The homogenate was centrifuged at 15 000 x g for 5 mins. Hoechst 33342 fluorescence was

measured using a BioTek® Synergy HT microplate reader at excitation and emission

wavelengths of 350 nm and 480 nm, respectively. The Hoechst 33342 efflux assay was

conducted in replicates of 9.

6.3.6 Cell accumulation assay

A cell accumulation assay was conducted to determine whether acyclovir is a substrate for

human BCRP. The assay was conducted as previously described (Bachmeier et al. 2006; Pollex

et al. 2010), with modification. Briefly, cell monolayers (overexpressing HEK293 cells) were

pre-treated with FTC (10 µmol/L) for 30 mins. Following the incubation period, the monolayers

were incubated with [8-14

C] acyclovir (1 µmol/L) in the presence or absence of FTC for an

additional 2 hrs. The media was then removed and the monolayers were washed (2X) with ice-

122

cold PBS. Cells were solubilized with NaOH (1N) on ice for 5 mins. The solubilized cells were

subsequently neutralized with HCL (1N). The cell lysates were centrifuged at 15 000 x g for 5

mins at 4◦C. Intracellular [8-

14C] acyclovir accumulation was determined by measuring the

radioactivity of the supernatant (600 µL) using a Beckman Coulter LS 6500 liquid scintillation

counter. Radioactivity was measured as disintegrations per minute (DPM). Intracellular [8-14

C]

acyclovir accumulation was normalized to total cell protein concentrations (mg/mL). Total

protein was quantified using the Bradford reagent (Sigma-Aldrich Canada Ltd.). The cell

accumulation assay was conducted in replicates of 3.

6.3.7 Statistical analyses

Statistical analyses of the data were performed using IBM® SPSS

® Statistics version 19 software.

Independent student t-tests were conducted to compare the data obtained from different

conditions of Hoechst 33342 or [8-14

C] acyclovir cell accumulation assays.

123

6.4 Results

6.4.1 The protein expression of human BCRP in overexpressing HEK293 cells

Figure 29 illustrates the protein expression of human BCRP in the overexpressing HEK293 cells,

showing that human BCRP was expressed in the overexpressing HEK293 cells.

Figure 29. The protein expression of human breast cancer resistance protein (BCRP) in the

overexpressing human embryonic kidney (HEK293) cells. Western blot assays were performed

to confirm the protein expression of human BCRP in the overexpressing HEK293 cells. Total

protein from human placenta (20 µg), mock (50 µg) or overexpressing HEK293 cells (50 µg)

were resolved on a 10 % sodium dodecyl sulfate – polyacrylamide gel electrophoresis (SDS-

PAGE). Blots were incubated with mouse BCRP antibody (BXP-21) diluted 1:120 in 5 % (w/v)

skim milk. The secondary antibody used was goat anti-mouse IgG-HRP antibody (sc-2005)

diluted 1:5000 in 5 % skim milk. Blots were developed using Western Lightning®

Plus-ECL

reagent. Blots were exposed to Kodak™

BioMax Light Film. The western blot assay was

performed in duplicates. The blot illustrated is a representative blot.

124

6.4.2 The functionality of BCRP in overexpressing HEK293 cells

The human BCRP was functional in the overexpressing HEK293 cells. In the presence of FTC,

the intracellular accumulation of Hoechst 33342 dye was 11-fold greater (p<0.05), compared to

its accumulation in cells that were not co-exposed to FTC (Figure 30).

Figure 30. The functionality of the human BCRP in overexpressing human embryonic kidney

(HEK293) cells. The Hoechst 33342 efflux assay was performed to determine whether the

human BCRP was functional in overexpressing HEK293 cells. Cell monolayers were incubated

in Kreb’s solution containing Hoechst 33342 (1 µmol/L) in the presence or absence of the BCRP

inhibitor, fumitremorgin C (FTC) (10 µmol/L) for 90 mins at 37◦C. The monolayers were

subsequently washed and incubated in Kreb’s solution in the presence or absence of FTC for an

additional 30 mins at 37◦C. Intracellular Hoechst 33342 fluorescence was measured in the

lysates using a BioTek® Synergy HT microplate reader at excitation and emission wavelengths

of 350 nm and 480 nm, respectively. The assay was conducted in replicates of 9. Intracellular

Hoechst 33342 accumulation is expressed as the fold change compared to control. The data is

presented as the mean ± standard error (SE). The statistical significance (p<0.05) of the

difference in the arbitrary fluorescent values between control and treated cell monolayers is

denoted by the symbol*.

*

0

2

4

6

8

10

12

14

control FTC

intr

ace

llu

lar

Ho

ech

st 3

33

42

acc

um

ula

tio

n)

(fo

ld c

ha

ng

e co

mp

are

d t

o c

on

tro

l)

125

6.4.3 Intracellular accumulation of [8-14

C] acyclovir

Figure 31 illustrates the intracellular accumulation of [8-14

C] acyclovir. In the presence of FTC,

the intracellular accumulation of [8-14

C] acyclovir was 5-fold greater (p<0.05), compared to its

accumulation in cells that were not co-exposed to FTC.

Figure 31. Intracellular accumulation of [8-14

C] acyclovir. Cellular accumulation assays were

conducted to determine whether acyclovir is a substrate for human BCRP. Cell monolayers

(overexpressing HEK293 cells) were pre-treated with FTC (10 µmol/L) for 30 mins. Following

the incubation period, the monolayers were incubated with [8-14

C] acyclovir (1 µmol/L) in the

presence or absence of FTC for an additional 2 hrs. Intracellular [8-14

C] acyclovir accumulation

was determined by measuring the radioactivity of the lysate (600 µL) using Beckman Coulter LS

6500 liquid scintillation counter. Radioactivity was measured as disintegrations per minute

(DPM). Intracellular [8-14

C] acyclovir accumulation was normalized to total cell protein

concentrations (mg/mL). The cell accumulation assays were conducted in replicates of 3.

Intracellular [8-14

C] acyclovir accumulation is expressed as the fold change compared to control.

The data is presented as the mean ± standard error (SE). The statistical significance (p<0.05) of

the difference in the intracellular [8-14

C] acyclovir accumulation (dpm/mg/mL) between control

and treated cell monolayers is denoted by the symbol*.

*

0

1

2

3

4

5

6

7

control FTC

intr

ace

llu

lar

[8-1

4C

] a

cycl

ov

ir a

ccu

mu

lati

on

(fo

ld c

ha

ng

e co

mp

are

d t

o c

on

tro

l)

126

6.5 Discussion

Results from the western blot (Figure 29) and Hoechst 33342 efflux (Figure 30) assays

confirmed the functional expression of human BCRP in the overexpressing HEK293 cells. The

results provide evidence illustrating that the cells were an appropriate in vitro model to use in our

study. Results from the cell accumulation assay illustrated that inhibition of human BCRP

caused significant accumulation of [8-14

C] acyclovir (Figure 31). Inhibition of the transporter

activity impeded the efflux of acyclovir from the cells resulting in the increased intracellular

accumulation of the antiviral agent. The study is the first to provide direct evidence illustrating

that acyclovir is a substrate for human BCRP and has the potential to contribute to a better

understanding of; (1) the renal tubular transport mechanisms of acyclovir and (2) the etiology of

the direct renal tubular injury induced by the drug.

Studies that have examined the renal tubular transport mechanisms of acyclovir are limited.

Huls and colleagues showed that functional BCRP is expressed in the apical membrane of human

renal proximal tubular cells (Huls et al. 2008). Therefore, the results of our study suggest that

since acyclovir is a substrate for the human BCRP; the transporter may play an active role in the

efflux of acyclovir from tubular cells.

The tissue expression profile of BCRP suggests that the efflux transporter may play a critical role

in tissue defense (Leslie et al. 2005; Mao and Unadkat 2005). For instance, BCRP expression in

human syncytiotrophoblast is believed to protect the fetus from exposure to circulating harmful

xenobiotics in the maternal blood (Leslie et al. 2005). Similarly, BCRP expression in the apical

membrane of epithelial cells lining the gastrointestinal tract suggests that the transporter provides

defense against oral exposure to harmful exogenous compounds (Leslie et al. 2005). Likewise,

127

the expression of BCRP in the apical membrane of human renal proximal tubular suggest that the

transporter may protect the tubular cells against accumulation of high intracellular concentrations

of xenobiotics, such as acyclovir, that may induce cytotoxicity.

Polymorphisms of drug transporters may decrease or abolish their functionality which may in

turn, result in reduced affinity of transporters for substrates, increased intracellular accumulation

of the substrates and resultant toxicity (Maeda and Sugiyama 2008). Studies have specifically

shown that mutations in the ABCG2 gene result in the reduced affinity of the transporter for

substrates and subsequent decreased efflux of the substrate by the dysfunctional transporter

(Cusatis et al. 2006; Pollex et al. 2010; Sparreboom et al. 2004; Yamasaki et al. 2008; Zhang et

al. 2006). Moreover, a study has shown that polymorphism in the ABCG2 gene may be directly

associated with drug – induced toxicity in humans (Cusatis et al. 2006), most likely due to the

reduced efflux of the substrate by the ABCG2 transporter. Hence, if human BCRP plays a

predominant role in the renal transport of acyclovir, then, reduced or abolished functionality of

human BCRP may impede the cellular efflux of acyclovir, which will result in the accumulation

of high intracellular concentrations of the antiviral agent and adverse cellular effects.

Further studies are required to determine the affinity of the transporter for acyclovir. Moreover,

in vivo studies can be performed to determine whether the transporter plays a significant role in

the renal tubular efflux of the drug and in the pathogenesis of the direct renal tubular injury that

is induced by acyclovir. For instance, studies can be conducted using mice with the wildtype or

knockout Abcg to determine whether the transporter plays a significant role in the efflux of

acyclovir. The studies can also be used to determine whether acyclovir induces a greater degree

of adverse renal tubular effects in Abcg knockout mice, compared to Abcg wildtype. Future

studies could also be employed to determine the potential effects of polymorphisms on the

128

transport of acyclovir by the human BCRP efflux transporter. Nevertheless, the results of our

study provides novel evidence which illustrate that acyclovir is a substrate for human BCRP and

provides a rationale for aforementioned future studies that can be employed in order to obtain a

better understanding of the etiology of the direct renal tubular injury that is induced by the

widely used antiviral agent.

6.6 Statement of significance

The results from this study are the first to show that acyclovir is a substrate for the human BCRP

transporter. These novel findings aid in the further elucidation of the renal transport mechanisms

of acyclovir and hence, may in turn, contribute to a better understanding of the overall etiology

of the direct renal tubular injury that is induced by the drug.

129

6.7 References

Ahmad, T., Simmonds, M., McIver, A.G., and McGraw, M.E. 1994. Reversible renal failure in

renal transplant patients receiving oral acyclovir prophylaxis. Pediatr Nephrol 8: 489-491.

Allen, J.D., Brinkhuis, R.F., Wijnholds, J., and Schinkel, A.H. 1999. The mouse

Bcrp1/Mxr/Abcp gene: amplification and overexpression in cell lines selected for resistance to

topotecan, mitoxantrone, or doxorubicin. Cancer Res 59; 4237-4241.

Allikmets, R., Schriml, L.M., Hutchinson, A., Romano-Spica, V., and Dean, M. 1998. A human

placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in

multidrug resistance. Cancer Res 58: 5337-5339.

Bachmeier, C.J., Trickler, W.J., and Miller, D.W. 2006. Comparison of drug efflux transport

kinetics in various blood-brain barrier models. Drug Metab Dispos 34: 998-1003.

Bianchetti, M.G., Roduit, C., and Oetliker, O.H. 1991. Acyclovir-induced renal failure: course

and risk factors. Pediatr Nephrol 5: 238-239.

Brigden, D., Rosling, A.E., and Woods, N.C. 1982. Renal function after acyclovir intravenous

injection. Am J Med 73: 182-185.

Brown, C.D., Sayer, R., Windass, A.S., Haslam, I.S., De Broe, M.E., D'Haese, P.C., and

Verhulst, A. 2008. Characterisation of human tubular cell monolayers as a model of proximal

tubular xenobiotic handling. Toxicol Appl Pharmacol 233: 428-438.

Chou, J.W., Yong, C., and Wootton, S.H. 2008. Case 2: Rash, fever and headache....first, do no

harm. Paediatr Child Health 13: 49-52.

Cusatis, G., Gregorc, V., Li, J., Spreafico, A., Ingersoll, R.G., Verweij, J., Ludovini, V., Villa,

E., Hidalgo, M., Sparreboom, A. and Baker, S.D. 2006. Pharmacogenetics of ABCG2 and

adverse reactions to gefitinib. J Natl Cancer Inst 98: 1739-1742.

Dean, M., Hamon, Y., and Chimini, G. 2001. The human ATP-binding cassette (ABC)

transporter superfamily. J Lipid Res 42: 1007-1017.

Doyle, L.A., Yang, W., Abruzzo, L.V., Krogmann, T., Gao, Y., Rishi, A.K., and Ross, D.D.

1998. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad

Sci U S A 95: 15665-15670.

Genc, G., Ozkaya, O., Acikgoz, Y., Yapici, O., Bek, K., Gulnar Sensoy, S., and Ozyurek, E.

2010. Acute renal failure with acyclovir treatment in a child with leukemia. Drug Chem Toxicol

33: 217-219.

130

Gonzalez-Lobato, L., Real, R., Prieto, J.G., Alvarez, A.I., and Merino, G. 2010. Differential

inhibition of murine Bcrp1/Abcg2 and human BCRP/ABCG2 by the mycotoxin fumitremorgin

C. Eur J Pharmacol 644: 41-48.

Gunness, P., Aleksa, K., Kosuge, K., Ito, S., and Koren, G. 2010. Comparison of the novel HK-2

human renal proximal tubular cell line with the standard LLC-PK1 cell line in studying drug-

induced nephrotoxicity. Can J Physiol Pharmacol 88: 448-455.

Hintz, M., Connor, J.D., Spector, S.A., Blum, M.R., Keeney, R.E., and Yeager, A.S. 1982.

Neonatal acyclovir pharmacokinetics in patients with herpes virus infections. Am J Med 73: 210-

214.

Huls, M., Brown, C.D., Windass, A.S., Sayer, R., van den Heuvel, J.J., Heemskerk, S., Russel,

F.G., and Masereeuw, R. 2008. The breast cancer resistance protein transporter ABCG2 is

expressed in the human kidney proximal tubule apical membrane. Kidney Int 73: 220-225.

Jonker, J.W., Merino, G., Musters, S., van Herwaarden, A.E., Bolscher, E., Wagenaar, E.,

Mesman, E., Dale, T.C., and Schinkel, A.H. 2005. The breast cancer resistance protein BCRP

(ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat Med 11: 127-129.

Keeney, R.E., Kirk, L.E., and Bridgen, D. 1982. Acyclovir tolerance in humans. Am J Med 73:

176-181.

Laemmli, U.K. 1970. Cleavage of structural proteins during the assembly of the head of

bacteriophage T4. Nature 227: 680-685.

Leslie, E.M., Deeley, R.G., and Cole, S.P. 2005. Multidrug resistance proteins: role of P-

glycoprotein, MRP1, MRP2, and BCRP (ABCG2) in tissue defense. Toxicol Appl Pharmacol

204: 216-237.

Lyon, A.W., Mansoor, A., and Trotter, M.J. 2002. Urinary gems: acyclovir crystalluria. Arch

Pathol Lab Med 126: 753-754.

Maeda, K., and Sugiyama, Y. 2008. Impact of genetic polymorphisms of transporters on the

pharmacokinetic, pharmacodynamic and toxicological properties of anionic drugs. Drug Metab

Pharmacokinet 23: 223-235.

Maliepaard, M., Scheffer, G.L., Faneyte, I.F., van Gastelen, M.A., Pijnenborg, A.C., Schinkel,

A.H., van De Vijver, M.J., Scheper, R.J., and Schellens, J.H. 2001. Subcellular localization and

distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer

Res 61: 3458-3464.

Mao, Q., and Unadkat, J.D. 2005. Role of the breast cancer resistance protein (ABCG2) in drug

transport. AAPS J 7: E118-133.

Mason, W.J., and Nickols, H.H. 2008. Crystalluria from acyclovir use. N Engl J Med 358: e14.

131

Millipore. 2007. Millipore Technical Publications. RIPA Buffer - Preparation of modified

radioimmunoprecipitation (RIPA) buffer.

[http://www.millipore.com/userguides/tech1/mcproto402].

Morisaki, K., Robey, R.W., Ozvegy-Laczka, C., Honjo, Y., Polgar, O., Steadman, K., Sarkadi,

B., and Bates, S.E. 2005. Single nucleotide polymorphisms modify the transporter activity of

ABCG2. Cancer Chemother Pharmacol 56: 161-172.

Ozvegy, C., Litman, T., Szakacs, G., Nagy, Z., Bates, S., Varadi, A., and Sarkadi, B. 2001.

Functional characterization of the human multidrug transporter, ABCG2, expressed in insect

cells. Biochem Biophys Res Commun 285: 111-117.

Peterslund, N.A., Larsen, M.L., and Mygind, H. 1988. Acyclovir crystalluria. Scand J Infect Dis

20: 225-228.

Pollex, E.K., Anger, G., Hutson, J., Koren, G., and Piquette-Miller, M. 2010. Breast cancer

resistance protein (BCRP)-mediated glyburide transport: effect of the C421A/Q141K BCRP

single-nucleotide polymorphism. Drug Metab Dispos 38: 740-744.

Robey, R.W., To, K.K., Polgar, O., Dohse, M., Fetsch, P., Dean, M., and Bates, S.E. 2009.

ABCG2: a perspective. Adv Drug Deliv Rev 61; 3-13.

Rocchi, E., Khodjakov, A., Volk, E.L., Yang, C.H., Litman, T., Bates, S.E., and Schneider, E.

2000. The product of the ABC half-transporter gene ABCG2 (BCRP/MXR/ABCP) is expressed

in the plasma membrane. Biochem Biophys Res Commun 271: 42-46.

Sawyer, M.H., Webb, D.E., Balow, J.E., and Straus, S.E. 1988. Acyclovir-induced renal failure.

Clinical course and histology. Am J Med 84: 1067-1071.

Sparreboom, A., Gelderblom, H., Marsh, S., Ahluwalia, R., Obach, R., Principe, P., Twelves, C.,

Verweij, J., and McLeod, H.L. 2004. Diflomotecan pharmacokinetics in relation to ABCG2

421C>A genotype. Clin Pharmacol Ther 76: 38-44.

Towbin, H., Staehelin, T., and Gordon, J. 1979. Electrophoretic transfer of proteins from

polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad

Sci U S A 76: 4350-4354.

Vachvanichsanong, P., Patamasucon, P., Malagon, M., and Moore, E.S. 1995. Acute renal failure

in a child associated with acyclovir. Pediatr Nephrol 9: 346-347.

Vomiero, G., Carpenter, B., Robb, I., and Filler, G. 2002. Combination of ceftriaxone and

acyclovir - an underestimated nephrotoxic potential? Pediatr Nephrol 17: 633-637.

Yamasaki, Y., Ieiri, I., Kusuhara, H., Sasaki, T., Kimura, M., Tabuchi, H., Ando, Y., Irie, S.,

Ware, J., Nakai, Y., Higuchi, S., and Sugiyama, Y. 2008. Pharmacogenetic characterization of

132

sulfasalazine disposition based on NAT2 and ABCG2 (BCRP) gene polymorphisms in humans.

Clin Pharmacol Ther 84: 95-103.

Zhang, W., Yu, B.N., He, Y.J., Fan, L., Li, Q., Liu, Z.Q., Wang, A., Liu, Y.L., Tan, Z.R., Fen, J.,

Huang, Y.F., and Zhou, H.H. 2006. Role of BCRP 421C>A polymorphism on rosuvastatin

pharmacokinetics in healthy Chinese males. Clin Chim Acta 373: 99-103.

133

6.8 Additional experiments not published

6.8.1 The effect of acyclovir on HEK293 cell viability

Since, we previously showed that acyclovir induces cytotoxicity (Chapter 3), it was critical to

determine whether the concentration of acyclovir used in the cell accumulation assay caused

toxicity to HEK293 cells.

6.8.2 Materials and methods

6.8.2a Cytotoxicity assay

A cytotoxicity (measured as a function of cell viability) assay was performed to determine

whether the concentration of acyclovir (1 µmol/L) were toxic to the overexpressing HEK293

cells. The assay was performed in 12-well plates. Cell viability was assessed using the

fluorescent alamarBlue® assay. Cell monolayers were exposed to acyclovir (1 µmol/L) for 2 hrs.

Following the incubation period, the media was removed and the monolayers were washed (2X)

with warm PBS. Fresh media was added to each well and the cell monolayers were subsequently

incubated with the alamarBlue®

reagent for 2.5 hrs. The final concentration of alamarBlue®

reagent in each well was 10 % (v/v). Cell viability was measured using BioTek® Synergy HT

microplate reader at excitation and emission wavelengths of 540 and 590 nm, respectively. The

assay was performed in replicates of 3.

6.8.2b Statistical analyses

Independent student t-tests were conducted to compare the data (arbitrary fluorescence units)

between untreated control and treated cell monolayers.

134

6.8.3 Results

Acyclovir was not toxic to the overexpressing HEK293 cells. Exposure to acyclovir (1 µmol/L)

had no effect on HEK293 cell viability (Figure 32).

Figure 32. The effect of acyclovir on human embryonic kidney (HEK293) cell viability. Cell

(overexpressing HEK293 cells) monolayers were incubated with acyclovir (0 or 1 µmol/L) for 2

hrs. Cell monolayers were subsequently washed with warm PBS and incubated with fresh media

and alamarBlue® reagent for 2.5 hrs. The final concentration of alamarBlue

® reagent in each

well was 10 % (v/v). Cell viability was measured using BioTek® Synergy HT microplate reader

at excitation and emission wavelengths of 540 and 590 nm, respectively. The cytotoxicity assay

was performed in replicates of 3. The results are expressed as a percentage of the fluorescence

of untreated control cell monolayers and presented as the mean ± standard error (SE). The

statistical significance (p<0.05) of the difference between the fluorescence from untreated

control cell monolayers and cell monolayers exposed to acyclovir is denoted by the symbol*.

0

20

40

60

80

100

120

control acyclovir

cell

via

bil

ity

(% u

ntr

ea

ted

co

ntr

ol)

135

Chapter 7

Summary of research findings

7.1 Summary of research findings and their significance

The results from this thesis have revealed several novel and important findings, which, when

taken together contribute to a better understanding of acyclovir – induced nephrotoxicity. The

results particularly aid in an improved insight into the cause of the drug’s direct renal tubular

injury, its tubular interaction with creatinine and renal transport mechanisms. Below is a

summary of the main research findings presented in this thesis and their relevant significance.

7.1.1 To investigate whether acyclovir – induced nephrotoxicity is due to, in part, direct

insult to renal tubular cells

Clinical evidence of tubular toxicity in the absence of crystal formation (Ahmad et al. 1994;

Vomiero et al. 2002) suggests that in addition to crystalluria, acyclovir may also induce direct

injury to renal epithelial cells. Studies have not investigated whether acyclovir induces direct

insult to renal tubular epithelial cells.

Employing both porcine LLC-PK1 and human HK-2 cells, results illustrated that acyclovir

induced a significant concentration – dependent decrease in cell viability. The results are the

first experimental evidence to support existing clinical data which suggest that the antiviral

agent may induce direct insult to renal tubular cells and thus, aid in a more comprehensive

understanding of the pathogenesis of the nephrotoxicity that is induced by the widely used

antiviral agent.

136

7.1.2 To determine whether acyclovir aldehyde plays a role in the direct renal tubular

injury induced by acyclovir

As previously elaborated, in humans, acyclovir is metabolized to an acyclovir aldehyde

intermediate metabolite (de Miranda et al. 1982). Reactive aldehyde metabolites are often

produced endogenously via drug metabolism (O’Brien et al. 2005). Numerous studies have

shown that the aldehyde metabolites cause the toxicities that are associated with the parent drug

(Dubourg et al. 2001; Kapetanovic et al. 2002; Ramu et al. 1995). Therefore, it is plausible that

the acyclovir aldehyde metabolite may cause the direct renal tubular injury that is associated with

acyclovir. Previous studies have not tested this viable hypothesis.

Results from this thesis illustrated that co-exposure to the ADH enzyme inhibitor, 4-

methylpyrazole, induced significant protection from acyclovir – induced HK-2 cell death. The

results suggest that acyclovir aldehyde may cause the direct renal tubular injury associated with

its parent drug. Further studies are required to determine the mechanisms of acyclovir aldehyde

– induced cytotoxicity. Nonetheless, the results from this research are the first to highlight that

the locally produced acyclovir aldehyde may play a role in the direct tubular injury and aids in

the further elucidation of the development of this renal toxicity.

7.1.3 To determine whether acyclovir inhibits the renal tubular secretion of creatinine

Creatinine shares renal tubular transport mechanisms with other drugs, which provides a

favourable opportunity for these drugs to compete with and subsequently inhibit the tubular

transport of creatinine. Inhibition of tubular secretion of creatinine results in an elevation of

plasma creatinine levels that are unreflective of reduced GFR or renal dysfunction. Several non-

nephrotoxic drugs are known to hinder the renal tubular secretion of creatinine to result in

137

transient, marked elevations in plasma creatinine levels that are not due to impaired renal

function (Berglund et al. 1975; Blackwood et al. 1976; Burgess et al. 1982; Burry and Dieppe

1976; Dubb et al. 1978; Dutt et al. 1981; Haggie et al. 1976; Kastrup et al. 1985; Myre et al.

1987; Opravil et al. 1993; Tschuppert et al. 2007).

Case reports (please refer to Table 4, Chapter 5, for a summary of the acyclovir cases) show that

acyclovir induces similar pronounced, transient elevations in plasma creatinine levels in patients

(Bianchetti et al. 1991; Brigden et al. 1982; Chou et al. 2008; Keeney et al. 1982;

Vachvanisanong et al. 1995; Vomiero et al. 2002). Studies reveal that similar to the non-

nephrotoxic drugs; acyclovir may share similar renal organic cation transporter systems with

creatinine (Takeda et al. 2002). Therefore it is plausible the acyclovir may inhibit the tubular

secretion of creatinine; a hypothesis that has not been previously investigated. It is critical to

determine whether acyclovir inhibits the secretion of creatinine because if this is the case, then

creatinine may not be the most appropriate biological marker to employ in order to assess the

renal function of patients administered acyclovir, and thus, other biological markers of renal

function, such as inulin, should always be used to assess renal function in these patients.

The inhibition of creatinine secretion by acyclovir via the OCT system was investigated in this

thesis. Transepithelial transport studies revealed that in contrast to quinidine, acyclovir did not

inhibit the transport of creatinine across LLC-PK1 or HK-2 cell monolayers. The results suggest

that acyclovir does not inhibit the tubular secretion of acyclovir in vitro, and possibly in humans

as well. Therefore, the abrupt, transient and pronounced elevations in plasma levels of creatinine

that are observed in patients may be solely and genuinely due to decreased GFR as a result of

acyclovir – induced renal dysfunction.

138

7.1.4 To determine whether acyclovir is a substrate for human BCRP

Results from a previous study suggest that acyclovir may be a substrate for the human BCRP

(Jonker et al. 2005). Studies have not yet directly determined whether acyclovir is a substrate for

the human BCRP transporter. It is crucial to determine whether acyclovir is a substrate for

human BCRP because this may aid in the further elucidation of the pathogenesis of the direct

tubular injury that is induced by the drug. Human BCRP is located in the apical membrane of

renal tubular cells (Huls et al. 2008), and thus, may play an important role in the renal transport

of acyclovir, and provide protection against the high intracellular accumulation of cytotoxic

acyclovir. Therefore, factors such as genetic polymorphisms may result in reduced or abolished

function of the efflux transporter which can subsequently result in increased intracellular

accumulation of acyclovir and cytotoxicity.

Results from this research showed that acyclovir is a substrate for the human BCRP. Further

studies are required to determine: (1) the affinity of the transporter for the drug, (2) whether the

transporter plays a significant role in the renal transport of acyclovir and (3) the effect of BCRP

polymorphisms on the transport of the drug. Nonetheless, the findings from this thesis are novel

and the first experimental evidence to illustrate that acyclovir is a substrate for human BCRP and

hence, provides a viable rationale for the aforesaid studies to be employed which will contribute

to an overall better understanding of the etiology of acyclovir – induced direct renal tubular

injury.

139

7.2 References

Ahmad, T., Simmonds, M., McIver, A.G., and McGraw, M.E. 1994. Reversible renal failure in

renal transplant patients receiving oral acyclovir prophylaxis. Pediatr Nephrol 8: 489-491.

Berglund, F., Killander, J., and Pompeius, R. 1975. Effect of trimethoprim-sulfamethoxazole on

the renal excretion of creatinine in man. J Urol 114: 802-808.

Bianchetti, M.G., Roduit, C., and Oetliker, O.H. 1991. Acyclovir-induced renal failure: course

and risk factors. Pediatr Nephrol 5: 238-239.

Blackwood, W.S., Maudgal, D.P., Pickard, R.G., Lawrence, D., and Northfield, T.C. 1976.

Cimetidine in duodenal ulcer. Controlled trial. Lancet 2: 174-176.

Brigden, D., Rosling, A.E., and Woods, N.C. 1982. Renal function after acyclovir intravenous

injection. Am J Med 73: 182-185.

Burgess, E., Blair, A., Krichman, K., and Cutler, R.E. 1982. Inhibition of renal creatinine

secretion by cimetidine in humans. Ren Physiol 5: 27-30.

Burry, H.C., and Dieppe, P.A. 1976. Apparent reduction of endogenous creatinine clearance by

salicylate treatment. Br Med J 2: 16-17.

Chou, J.W., Yong, C., and Wootton, S.H. 2008. Case 2: Rash, fever and headache....first, do no

harm. Paediatr Child Health 13: 49-52.

de Miranda, P., Good, S.S., Krasny, H.C., Connor, J.D., Laskin, O.L., and Lietman, P.S. 1982.

Metabolic fate of radioactive acyclovir in humans. Am J Med 73: 215-220.

Dubb, J.W., Stote, R.M., Familiar, R.G., Lee, K., and Alexander, F. 1978. Effect of cimetidine

on renal function in normal man. Clin Pharmacol Ther 24: 76-83.

Dubourg, L., Michoudet, C., Cochat, P., and Baverel, G. 2001. Human kidney tubules detoxify

chloroacetaldehyde, a presumed nephrotoxic metabolite of ifosfamide. J Am Soc Nephrol 12:

1615-1623.

Dutt, M.K., Moody, P., and Northfield, T.C. 1981. Effect of cimetidine on renal function in man.

Br J Clin Pharmacol 12: 47-50.

Haggie, S.J., Fermont, D.C., and Wyllie, J.H. 1976. Treatment of duodenal ulcer with cimetidine.

Lancet 1: 983-984.

Huls, M., Brown, C.D., Windass, A.S., Sayer, R., van den Heuvel, J.J., Heemskerk, S., Russel,

F.G., and Masereeuw, R. 2008. The breast cancer resistance protein transporter ABCG2 is

expressed in the human kidney proximal tubule apical membrane. Kidney Int 73: 220-225.

140

Jonker, J.W., Merino, G., Musters, S., van Herwaarden, A.E., Bolscher, E., Wagenaar, E.,

Mesman, E., Dale, T.C., and Schinkel, A.H. 2005. The breast cancer resistance protein BCRP

(ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat Med 11: 127-129.

Kapetanovic, I.M., Torchin, C.D., Strong, J.M., Yonekawa, W.D., Lu, C., Li, A.P., Dieckhaus,

C.M., Santos, W.L., Macdonald, T.L., Sofia, R.D., and Kupferberg, H.J. 2002. Reactivity of

atropaldehyde, a felbamate metabolite in human liver tissue in vitro. Chem Biol Interact 142:

119-134.

Kastrup, J., Petersen, P., Bartram, R., and Hansen, J.M. 1985. The effect of trimethoprim on

serum creatinine. Br J Urol 57: 265-268.

Keeney, R.E., Kirk, L.E., and Bridgen, D. 1982. Acyclovir tolerance in humans. Am J Med 73:

176-181.

Myre, S.A., McCann, J., First, M.R., and Cluxton, R.J., Jr. 1987. Effect of trimethoprim on

serum creatinine in healthy and chronic renal failure volunteers. Ther Drug Monit 9: 161-165.

O'Brien, P.J., Siraki, A.G., and Shangari, N. 2005. Aldehyde sources, metabolism, molecular

toxicity mechanisms, and possible effects on human health. Crit Rev Toxicol 35: 609-662.

Opravil, M., Keusch, G., and Luthy, R. 1993. Pyrimethamine inhibits renal secretion of

creatinine. Antimicrob Agents Chemother 37: 1056-1060.

Ramu, K., Fraiser, L.H., Mamiya, B., Ahmed, T., and Kehrer, J.P. 1995. Acrolein mercapturates:

synthesis, characterization, and assessment of their role in the bladder toxicity of

cyclophosphamide. Chem Res Toxicol 8: 515-524.

Takeda, M., Khamdang, S., Narikawa, S., Kimura, H., Kobayashi, Y., Yamamoto, T., Cha, S.H.,

Sekine, T., and Endou, H. 2002. Human organic anion transporters and human organic cation

transporters mediate renal antiviral transport. J Pharmacol Exp Ther 300: 918-924.

Tschuppert, Y., Buclin, T., Rothuizen, L.E., Decosterd, L.A., Galleyrand, J., Gaud, C., and

Biollaz, J. 2007. Effect of dronedarone on renal function in healthy subjects. Br J Clin Pharmacol

64: 785-791.

Vachvanichsanong, P., Patamasucon, P., Malagon, M., and Moore, E.S. 1995. Acute renal failure

in a child associated with acyclovir. Pediatr Nephrol 9: 346-347.

Vomiero, G., Carpenter, B., Robb, I., and Filler, G. 2002. Combination of ceftriaxone and

acyclovir - an underestimated nephrotoxic potential? Pediatr Nephrol 17: 633-637.

141

Chapter 8

General Discussion and Conclusions

8.1 Acyclovir and direct renal tubular injury

Employing both porcine (LLC-PK1) and human (HK-2) renal proximal tubular epithelial cells,

the thesis provides the first experimental evidence to support existing clinical evidence which

suggest that acyclovir induces direct insult to renal tubular cells (Figure 8). The findings shed

new insight into the pathogenesis of the nephrotoxicity induced by the antiviral agent.

The next step in the research was to determine whether the locally produced acyclovir aldehyde

metabolite may play a role in the direct tubular insult that is induced by the drug. While, both

porcine (LLC-PK1) and human (HK-2) renal tubular epithelial cells were employed to determine

whether acyclovir induces direct insult to renal tubular cells, inter-species differences exist in

drug disposition (Riddick 1998). The in vitro results obtained from exposure of LLC-PK1 or

HK-2 cells to acyclovir are an excellent example of the inter-species differences that exist in the

disposition of acyclovir. Figure 8 shows that compared to HK-2 cells, the LLC-PK1 cells were

more susceptible to the cytotoxic effects of acyclovir. Therefore, the HK-2 cells were used in all

subsequent studies conducted to study acyclovir – induced nephrotoxicity. The HK-2 cells are

derived from the normal kidney of an adult male and have biochemical and functional

characteristics of well-differentiated renal proximal tubular epithelial cells (Ryan et al. 1994).

Moreover, while, the principle site of acyclovir – induced tubular injury, along the nephron,

remains to be established; it is highly possible that the injury occurs predominantly in the

proximal tubule segment of the nephron. The leaky epithelium, active transporter systems and

142

increased levels of drug biotransformation enzymes in the tubular cells (Schnellmann 2001) are

examples of some morphological and biochemical characteristics that enhances the susceptibility

of the proximal tubule segment of the nephron to toxicant – induced renal damage. The HK-2

cells are epithelial cells of proximal tubular origin and therefore the use of the cells to study

acyclovir – induced nephrotoxicity allows for a more relevant elucidation of the tubular cellular

insult that acyclovir may induce in humans.

Western blot and enzyme activity assays confirmed that the HK-2 cells express functional ADH

(Figures 9A and 11A) and ALDH (Figures 9B and 11B) enzymes. Thus, the cells contained the

enzymatic machinery to locally metabolize acyclovir to its acyclovir aldehyde metabolite,

thereby, making them an appropriate in vitro model to employ in order to study the direct tubular

insult induced by acyclovir.

Importantly, the results from this thesis illustrate that co-exposure to 4-methylpyrazole render

significant protection from acyclovir – induced cell death (Figure 13). The results are the first to

suggest that the acyclovir aldehyde may cause the direct renal tubular injury associated with its

parent drug, because inhibition of its formation resulted in significant alleviation of HK-2

cytotoxicity. Furthermore, these results suggest that local metabolism of acyclovir to an

aldehyde metabolite occurs in human renal proximal tubular cells, in vitro. Additional support

for the local metabolism of acyclovir in human renal proximal tubular cells in vitro can be

derived from the results presented in Figure 14, where the results showed that an aldehyde was

produced in HK-2 cells that were exposed to acyclovir. Taken together, the in vitro results

suggest that human renal proximal tubular cells may be capable of locally metabolizing acyclovir

to its acyclovir aldehyde, in vivo and the aldehyde metabolite may be the ultimate source of the

direct renal tubular toxicity associated with the use of the antiviral.

143

The significant role of acyclovir aldehyde in the direct renal toxicity may be criticized due to the

small alleviation in HK-2 cell death caused by co-exposure to 4-methylpyrazole. A possible

explanation for this observation is that the innate low expression of the ADH enzyme in the cells

resulted in limited local metabolism of acyclovir to its aldehyde metabolite, and inhibition by 4-

methylpyrazole.

In order to extrapolate the biological significance of our in vitro cell results to humans, ADH

protein expression in HK-2 cells and human kidney was compared. Analyses (Figure 15)

suggest that compared to HK-2 cells, the ADH protein expression may be approximately 30 fold

higher in human kidney. Thus, it is highly likely that the local metabolism of acyclovir to

acyclovir aldehyde and the subsequent inhibition of this pathway by 4-methylpyrazole may be

greatly exacerbated in human renal proximal tubular cells, in vivo.

Another possible reason for the small effect that 4-methylpyrazole had on HK-2 cell viability

may be due to the noxioius effect(s) of the acyclovir aldehyde metabolite on the protein

expression and/or function of ADH enzyme. Aldehydes are known to reduce the activity of

enzymes (O’Brien et al. 2005), specifically its detoxifying enzyme, aldehyde dehydrogenase

(Doorn et al. 2006; Kapetanovic et al. 2002). Presently, the effect(s) of aldehydes on the

expression or activity of alcohol dehydrogenase enzymes is not known. However, it is probable

that aldehydes can affect ADH protein expression and/or function either through direct or

indirect mechanisms of toxicity (Gregus and Klaassen 2001).

The results from this study also provide some explanation of the lack of occurrence of

nephrotoxicity in all patients. The occurrence of acyclovir – induced nephrotoxicity is

independent of age and gender (Schreiber et al. 2008). However, genetic polymorphisms

144

(Borson and Li 1986; Mizoi et al. 1994; Mulligan et al. 2003; Stickel and Osterreicher 2006) are

well known to affect the functional expression of enzymes, including the ADH and ALDH

enzymes. Thus, it is likely that such polymorphisms that affect the functional expression of the

ADH or ALDH enzymes may subsequently influence the occurrence or severity of acyclovir –

induced nephrotoxicity in patients. Altered functional expression of ADH or ALDH enzymes

may perturb the local renal tubular disposition of acyclovir such that increased ADH or reduced

ALDH functional expression may result in increased intracellular accumulation of the pernicious

acyclovir aldehyde metabolite and cytotoxicity. A previous study has shown that genetic

polymorphisms in the ALDH2 gene influence of pharmacokinetics of acyclovir in patients, such

that polymorphisms that resulted in the reduced functionality of ALDH2 protein resulted in

significant increases in the elimination half-life of acyclovir (Hara et al. 2008).

Overall, the above findings highlight an important toxicological phenomenon which is

commonly ignored: the fact that the kidney produces its own poison. The kidney, being exposed

to large amounts of parent drugs and their active and/or inactive metabolites, is not commonly

regarded as a drug metabolizing organ, and from a whole body perspective, the liver and

intestine are responsible for most of the burden of drug metabolism.

It is evident that the small amounts of acyclovir metabolism in the kidney is not likely to change

the fact that most of the body load of acyclovir is eliminated unchanged. Yet, as our results

suggest, the kidney is able to produce sufficient amounts of acyclovir aldehyde to cause damage

to tubular cells. A similar phenomenon was reported by our laboratory with the

chemotherapeutic agent, ifosfamide (Aleksa et al. 2005), where the parent drug itself is not

nephrotoxic, but its aldehyde metabolite, chloroacetaldehyde, produced by the renal tubular

cells, is highly toxic.

145

These findings have several basic and important implications: first, one has to seriously consider

the drug metabolizing capacity of organs and cells in the context of producing high local

concentrations of toxic metabolites, rather than amounts that would endanger the whole body.

Second, the fact that only some patients are affected, sparing many others, suggests that

polymorphisms in drug metabolizing enzymes, (in our case, the alcohol and aldehyde

dehydrogenase) may predict patients who might be adversely affected. Finally, the metabolic

patterns of local aldehyde products should direct us toward effective therapies, based on the

mechanisms of injury. In the case of ifosfamide, this has lead us to successfully treat tubular

cells, in vitro, animals and children with the antioxidant, N-Aceytlcysteine (Chen et al. 2007;

2008; Hanly et al. 2011). This thesis suggests similar future work towards prevention of

acyclovir – induced tubular toxicity.

8.2 Acyclovir-creatinine tubular interaction

Utilizing both LLC-PK1 and HK-2 cells, the results (Figure 21 and 22) show that acyclovir does

not inhibit the transepithelial transport of creatinine across the cell monolayers. The reasons for

the lack of inhibition of creatinine secretion by acyclovir in our experiments are detailed in

Chapter 5 (section 5.4) of the thesis and include; (1) the possibility that the increase in the levels

of plasma creatinine may be due to the interference of acyclovir with the Jaffé-based assay for

creatinine measurement, and not due to a tubular inetraction between creatinine and acyclovir

and (2) various limitations of the in vitro models employed in the experiments. Nonetheless, the

results clearly suggest that acyclovir does not inhibit the tubular secretion of creatinine in vitro,

and possibly in vivo, as well. Therefore, the abrupt, marked and transient elevation in the levels

of plasma creatinine observed in some patients may be solely due to decreased GFR as a result of

acyclovir – induced renal dysfunction.

146

8.3 Renal tubular transport of acyclovir

Elucidation of the renal transport mechanisms of drugs, such as acyclovir, that induce direct

insult to tubular cells, is imperative in order to understand the complete etiology of its toxicity.

The human BCRP efflux transporter is localized to the apical membrane of renal tubular cells

(Huls et al. 2008), suggesting that the transporter may play a protective role against the high

intracellular accumulation of xenobiotics in renal tubular cells and the resultant cytotoxicity.

Therefore, if acyclovir is a substrate for human BCRP and the transporter plays a significant role

in the renal transport of acyclovir, then reduced or abolished function of the efflux transporter

may result in the increased intracellular accumulation of acyclovir and subsequent detrimental

nephrotoxic consequences, such as direct injury to renal tubular cells.

The results (Figure 31) clearly illustrate that acyclovir is a substrate for human BCRP. The

findings are novel and bear several important implications with respect to renal transport and the

etiology of the direct renal tubular injury that is induced by the drug. First, the studies suggest

that the efflux transporter may play an active role in the tubular transport of acyclovir. Second,

factors such as genetic polymorphisms that reduce or abolish the functionality of BCRP may

result in the increased intracellular accumulation of acyclovir and subsequent toxicity. Previous

studies have demonstrated that mutations in the ABCG2 gene result in the reduced affinity of the

transporter for substrates and subsequent decreased efflux of the substrate by the dysfunctional

transporter (Cusatis et al. 2006; Pollex et al. 2010; Sparreboom et al. 2004; Yamasaki et al. 2008;

Zhang et al. 2006). More critical are the results from a study that has shown that polymorphism

in the ABCG2 gene may be directly associated with drug – induced toxicity in humans (Cusatis

et al. 2006), most likely due to the reduced efflux of the substrate by the transporter.

147

Hence, these novel findings from this research aid in the further elucidation of the renal transport

mechanisms of acyclovir and in turn, contribute to a more thorough understanding of the

etiology of the direct renal tubular injury that is induced by the antiviral agent.

8.4 Limitations and future directions

The series of studies in this thesis present several novel findings that aid in a more complete

understanding of the etiology of acyclovir – induced nephrotoxicity, however, there are some

limitations of the employed experimental models and there is a need for several future studies to

be conducted. First and foremost, the study employed in vitro cell culture models. Over the past

25 years, in vitro models have gained increased use in pharmacology and toxicology studies,

with established animal and human cell lines being the most common models used for such said

studies (Zucco et al. 2004). Compared to in vivo models, in vitro human cell culture models

provide several main advantages, including; (1) cost-effectiveness, (2) absence of inter-species

differences, (3) increased control over experimental conditions and (4) better elucidation of

underlying cell and molecular mechanisms of action or toxicity (David et al. 1998; Plant 2004).

On the other hand, several disadvantages exists with the use of in vitro human cell culture

models including; (1) the disruption of the normal integrity of the cell structure and inter-cellular

relationships, (2) the production of artifactual drug binding sites that does not normally exist in

vivo, (3) differences between in vitro and in vivo drug pharmaco- or toxicokinetics and (Davila et

al. 1998) and (4) altered protein expression (Plant 2004). Therefore, extreme caution should be

employed when extrapolating the in vitro results obtained from this thesis to humans.

Second, while HK-2 serves as a good model to study acyclovir – induced nephrotoxicity, the

cells expressed inherent low expression of the ADH enzyme in the HK-2 cells, which may have

148

limited the degree of local renal intracellular metabolism of acyclovir to acyclovir aldehyde and

inhibition of the pathway by 4-methylpyrazole. Therefore, co-exposure studies with 4-

methylpyrazole could be performed using transformed cell lines that have been transfected with

the wildtype human ADH enzyme, such that the enzyme would be functionally overexpressed in

the cell lines. Furthermore, the ADH or ALDH isozyme(s) that are responsible for the

metabolism of acyclovir has not been established. In vitro metabolism studies utilizing pure

human ADH or ALDH isozymes and respective enzyme inhibitors could be conducted to

decipher the specific isozyme(s) that are responsible for the metabolism of acyclovir. Acyclovir

and CMMG concentrations could then be subsequently measured using HPLC coupled with UV,

fluorescence or mass spectrometry detectors. The HPLC coupled with UV (Darville et al. 2007),

fluorescence (Svensson et al. 1997) or mass spectrometry (Helldén et al. 2003; 2006) detectors

have been used in previous studies to measure the concentrations of acyclovir and CMMG in

biological matrices.

We postulated that the small effect of 4-methylpyrazole may have been due to the partial

inhibition of the ADH enzyme. Therefore, future studies can be performed employing a series of

concentrations of 4-methylpyrazole to determine the degree of ADH inhibition in the HK-2 cells

and the effect on the viability of cells co-exposed to 4-methylpyrazole and acyclovir.

In this thesis, it was hypothesized that renal tubular cells have the enzymatic machinery required

to locally metabolize acyclovir to its aldehyde metabolite. The results of the present research

suggest that human renal proximal tubular cells were able to locally metabolize acyclovir to

acyclovir aldehyde and that the metabolite was produced in sufficient quantity to cause toxicity;

because prevention of its generation provided significant protection against cell death. Future

studies could be performed to provide further evidence to support the above results. For

149

example, intracellular levels of acyclovir and CMMG could be measured in HK-2 cell

monolayers exposed to acyclovir (0 – 2000 µg/mL) for 24 hours. Samples could be

subsequently analyzed using the aforementioned HPLC techniques. The measurement of

CMMG in these samples would provide indirect evidence for the local production of the

acyclovir aldehyde metabolite. Subsequent studies could also be attempted to measure the levels

of acyclovir aldehyde in HK-2 cells in order to provide direct evidence for the local renal tubular

cellular metabolism of acyclovir to its noxious aldehyde metabolite. Studies can also be

executed to further investigate the underlying cell and molecular mechanism(s) of renal tubular

cell death.

Human kidney tissue, which has been shown in this thesis to express innately higher levels of the

ADH and ALDH enzymes, compared to the HK-2 cell line, could also be used to conduct

metabolism studies in order to determine whether human renal tubular cells are able to locally

metabolize acyclovir. The studies should be conducted using plasma (5 – 20 µg/mL)

concentrations of acyclovir in order to prevent cytotoxicity.

Fourth, the results from the transepithelial transport studies show that acyclovir does not inhibit

the secretion of creatinine across LLC-PK1 or HK-2 cell monolayers. However, one significant

and potential reason for this observation may have been due the possibility that acyclovir and

creatinine were transported by different hOCTs. Therefore, studies could be conducted using

cells overexpressing specific hOCTs in order to determine the interaction of acyclovir and

creatinine at individual hOCTs.

Finally, while the cell accumulation studies clearly show that acyclovir is a substrate for the

human BCRP and suggest that efflux transporter may play a role in the tubular transport of

150

acyclovir; further studies are required to determine whether the transporter does play a

significant role in renal transport of acyclovir. In vivo studies can be conducted using mice with

the wildtype or knockout Abcg gene to determine whether the transporter plays a significant role

in the efflux of acyclovir. The studies can also be used to determine whether acyclovir induces a

greater degree of adverse renal tubular effects in Abcg knockout mice, compared to Abcg

wildtype. The results from the studies will address the importance of the efflux transporter in the

pathogenesis of the direct renal tubular injury induced by the drug. However, due to the inter-

species differences that exists between murine Bcrp1 and human BCRP (see section 6.1), caution

should be employed during extrapolation of the results to humans. Future studies could also be

employed to determine the affinity of the transporter for the drug and the potential effects of

genetic polymorphisms of the ABCG2 gene on the transport of acyclovir.

8.5 Conclusions

Several new pieces of evidence are presented in this thesis that aids in a better understanding of

the overall etiology of acyclovir – induced nephrotoxicity. First, results suggest that in addition

to crystalluria, acyclovir induces direct insult to human renal proximal tubular cells and that the

insult may be caused by the drug’s aldehyde intermediate metabolite which is locally produced

in human renal proximal tubular cells. Second, the results suggest that the abrupt, marked and

transient elevations in the levels of plasma creatinine observed in patients is solely and

genuinely due to reduced GFR as a result of acyclovir – induced renal impairment and not due

to a tubular interaction between the drug and creatinine. Third, the research suggests that

acyclovir is a substrate for human BCRP; results which in turn, bear critical implications for

renal transport and acyclovir – induced nephrotoxicity. Finally, due to the genetic

polymorphisms in human ADH, ALDH and BCRP genes; the results that suggest that acyclovir

151

aldehyde may cause the direct toxicity associated with the parent drug and that the drug is a

substrate for human BCRP, respectively, present a possible explanation for the occurrence of

acyclovir – induced nephrotoxicity in some patients, while sparing other individuals.

In summary, the results presented in this thesis, fills several knowledge gaps that existed in the

study of the pathogenesis of the direct renal damage caused by acyclovir. Critically, the study

highlights the need for future studies that will contribute to the further understanding of the

fundamental cell and molecular mechanism(s) of this toxicity and potential therapies for

deterrence of this renal damage.

152

8.6 References

Aleksa, K., Halachmi, N., Ito, S., and Koren, G. 2005. A tubule cell model for ifosfamide

nephrotoxicity. Can J Physiol Pharmacol 83: 499-508.

Bosron, W.F., and Li, T.K. 1986. Genetic polymorphism of human liver alcohol and aldehyde

dehydrogenases, and their relationship to alcohol metabolism and alcoholism. Hepatology 6:

502-510.

Chen, N., Aleksa, K., Woodland, C., Rieder, M., and Koren, G. 2007. The effect of N-

acetylcysteine on ifosfamide-induced nephrotoxicity: in vitro studies in renal tubular cells. Transl

Res 150: 51-57.

Chen, N., Aleksa, K., Woodland, C., Rieder, M., and Koren, G. 2008. N-Acetylcysteine prevents

ifosfamide-induced nephrotoxicity in rats. Br J Pharmacol 153: 1364-1372.

Cusatis, G., Gregorc, V., Li, J., Spreafico, A., Ingersoll, R.G., Verweij, J., Ludovini, V., Villa,

E., Hidalgo, M., Sparreboom, A., and Baker, S.D. 2006. Pharmacogenetics of ABCG2 and

adverse reactions to gefitinib. J Natl Cancer Inst 98: 1739-1742.

Darville, J.M., Lovering, A.M., and MacGowan, A.P. 2007. Development, evaluation and

application of an isocratic high-performance liquid chromatography (HPLC) assay for the

simultaneous determination of aciclovir and its metabolite 9-carboxymethoxymethylguanine in

human serum and cerebrospinal fluid. Int J Antimicrob Agents 30: 30-33.

Davila, J.C., Rodriguez, R.J., Melchert, R.B., and Acosta, D., Jr. 1998. Predictive value of in

vitro model systems in toxicology. Annu Rev Pharmacol Toxicol 38: 63-96.

Doorn, J.A., Hurley, T.D., and Petersen, D.R. 2006. Inhibition of human mitochondrial aldehyde

dehydrogenase by 4-hydroxynon-2-enal and 4-oxonon-2-enal. Chem Res Toxicol 19: 102-110.

Gregus Z. and Klaassen, C.D. 2001. Mechanisms of Toxicity. In Casarett and Doull's Toxicology

The Basic Science of Poisons. Edited by Klaassen, C.D. The McGraw-Hill Companies Inc., New

York. pp 35-78.

Hanly, L.N., Chen, N., Aleksa, K., Cutler, M., Bajcetic, M., Palassery, R., Regueira, O., Turner,

C., Baw, B., Malkin, B., Freeman, D., Rieder, M.J., Vasylyeya, T.L. and Koren. G. 2011. N-

acetylcysteine as a Novel Prophylactic Treatment for Ifosfamide-Induced Nephrotoxicity in

Children: Translational Pharmacokinetics. J Clin Pharmacol. [Epub ahead of print].

Hara, K., Suyama, K., Itoh, H., and Nagashima, S. 2008. Influence of ALDH2 genetic

polymorphisms on aciclovir pharmacokinetics following oral administration of valaciclovir in

Japanese end-stage renal disease patients. Drug Metab Pharmacokinet 23: 306-312.

153

Hellden, A., Lycke, J., Vander, T., Svensson, J.O., Odar-Cederlof, I., and Stahle, L. 2006. The

aciclovir metabolite CMMG is detectable in the CSF of subjects with neuropsychiatric symptoms

during aciclovir and valaciclovir treatment. J Antimicrob Chemother 57: 945-949.

Hellden, A., Odar-Cederlof, I., Diener, P., Barkholt, L., Medin, C., Svensson, J.O., Sawe, J., and

Stahle, L. 2003. High serum concentrations of the acyclovir main metabolite 9-

carboxymethoxymethylguanine in renal failure patients with acyclovir-related neuropsychiatric

side effects: an observational study. Nephrol Dial Transplant 18: 1135-1141.

Huls, M., Brown, C.D., Windass, A.S., Sayer, R., van den Heuvel, J.J., Heemskerk, S., Russel,

F.G., and Masereeuw, R. 2008. The breast cancer resistance protein transporter ABCG2 is

expressed in the human kidney proximal tubule apical membrane. Kidney Int 73: 220-225.

Kapetanovic, I.M., Torchin, C.D., Strong, J.M., Yonekawa, W.D., Lu, C., Li, A.P., Dieckhaus,

C.M., Santos, W.L., Macdonald, T.L., Sofia, R.D.,and Kupferberg, H.J. 2002. Reactivity of

atropaldehyde, a felbamate metabolite in human liver tissue in vitro. Chem Biol Interact 142:

119-134.

Mizoi, Y., Yamamoto, K., Ueno, Y., Fukunaga, T., and Harada, S. 1994. Involvement of genetic

polymorphism of alcohol and aldehyde dehydrogenases in individual variation of alcohol

metabolism. Alcohol Alcohol 29: 707-710.

Mulligan, C.J., Robin, R.W., Osier, M.V., Sambuughin, N., Goldfarb, L.G., Kittles, R.A.,

Hesselbrock, D., Goldman, D., and Long, J.C. 2003. Allelic variation at alcohol metabolism

genes ( ADH1B, ADH1C, ALDH2) and alcohol dependence in an American Indian population.

Hum Genet 113: 325-336.

O'Brien, P.J., Siraki, A.G., and Shangari, N. 2005. Aldehyde sources, metabolism, molecular

toxicity mechanisms, and possible effects on human health. Crit Rev Toxicol 35: 609-662.

Plant, N. 2004. Strategies for using in vitro screens in drug metabolism. Drug Discov Today 9:

328-336.

Pollex, E.K., Anger, G., Hutson, J., Koren, G., and Piquette-Miller, M. 2010. Breast cancer

resistance protein (BCRP)-mediated glyburide transport: effect of the C421A/Q141K BCRP

single-nucleotide polymorphism. Drug Metab Dispos 38: 740-744.

Riddick, D.S. 1998. Drug Biotransformation. In Principles of Medical Pharmacology. Edited by

Kalant, H., and Roschlau, W.H.E. Oxford University Press, New York. pp 38-54.

Ryan, M.J., Johnson, G., Kirk, J., Fuerstenberg, S.M., Zager, R.A., and Torok-Storb, B. 1994.

HK-2: an immortalized proximal tubule epithelial cell line from normal adult human kidney.

Kidney Int 45: 48-57.

154

Schnellmann, R.G. 2001. Toxic responses of the kidney. In Casarett and Doull's Toxicology The

Basic Science of Poisons. Edited by Klaassen, C.D. The McGraw-Hill Companies Inc., New

York. pp 491-514.

Schreiber, R., Wolpin, J., and Koren, G. 2008. Determinants of aciclovir-induced nephrotoxicity

in children. Paediatr Drugs 10: 135-139.

Sparreboom, A., Gelderblom, H., Marsh, S., Ahluwalia, R., Obach, R., Principe, P., Twelves, C.,

Verweij, J., and McLeod, H.L. 2004. Diflomotecan pharmacokinetics in relation to ABCG2

421C>A genotype. Clin Pharmacol Ther 76: 38-44.

Stickel, F., and Osterreicher, C.H. 2006. The role of genetic polymorphisms in alcoholic liver

disease. Alcohol Alcohol 41: 209-224.

Svensson, J.O., Barkholt, L., and Sawe, J. 1997. Determination of acyclovir and its metabolite 9-

carboxymethoxymethylguanine in serum and urine using solid-phase extraction and high-

performance liquid chromatography. J Chromatogr B Biomed Sci Appl 690: 363-366.

Yamasaki, Y., Ieiri, I., Kusuhara, H., Sasaki, T., Kimura, M., Tabuchi, H., Ando, Y., Irie, S.,

Ware, J., Nakai, Y., Higuchi, S., and Sugiyama, Y. 2008. Pharmacogenetic characterization of

sulfasalazine disposition based on NAT2 and ABCG2 (BCRP) gene polymorphisms in humans.

Clin Pharmacol Ther 84: 95-103.

Zhang, W., Yu, B.N., He, Y.J., Fan, L., Li, Q., Liu, Z.Q., Wang, A., Liu, Y.L., Tan, Z.R., Fen, J.,

Huang, Y.F., and Zhou, H.H. 2006. Role of BCRP 421C>A polymorphism on rosuvastatin

pharmacokinetics in healthy Chinese males. Clin Chim Acta 373: 99-103.