100% found this document useful (3 votes)
46 views156 pages

(Ebook) Pharmacogenetics by Ian P. Hall (Editor), Munir Pirmohamed (Editor) ISBN 9780824728847, 082472884X 2025 PDF Download

Study resource: (Ebook) Pharmacogenetics by Ian P. Hall (Editor), Munir Pirmohamed (Editor) ISBN 9780824728847, 082472884XGet it instantly. Built for academic development with logical flow and educational clarity.

Uploaded by

anxifrzn8773
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
100% found this document useful (3 votes)
46 views156 pages

(Ebook) Pharmacogenetics by Ian P. Hall (Editor), Munir Pirmohamed (Editor) ISBN 9780824728847, 082472884X 2025 PDF Download

Study resource: (Ebook) Pharmacogenetics by Ian P. Hall (Editor), Munir Pirmohamed (Editor) ISBN 9780824728847, 082472884XGet it instantly. Built for academic development with logical flow and educational clarity.

Uploaded by

anxifrzn8773
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 156

(Ebook) Pharmacogenetics by Ian P.

Hall (Editor), Munir


Pirmohamed (Editor) ISBN 9780824728847, 082472884X Pdf
Download

https://ebooknice.com/product/pharmacogenetics-2164610

★★★★★
4.7 out of 5.0 (39 reviews )

DOWNLOAD PDF

ebooknice.com
(Ebook) Pharmacogenetics by Ian P. Hall (Editor), Munir
Pirmohamed (Editor) ISBN 9780824728847, 082472884X Pdf
Download

EBOOK

Available Formats

■ PDF eBook Study Guide Ebook

EXCLUSIVE 2025 EDUCATIONAL COLLECTION - LIMITED TIME

INSTANT DOWNLOAD VIEW LIBRARY


We have selected some products that you may be interested in
Click the link to download now or visit ebooknice.com
for more options!.

(Ebook) Vagabond, Vol. 29 (29) by Inoue, Takehiko ISBN


9781421531489, 1421531488

https://ebooknice.com/product/vagabond-vol-29-29-37511002

(Ebook) Biota Grow 2C gather 2C cook by Loucas, Jason; Viles,


James ISBN 9781459699816, 9781743365571, 9781925268492,
1459699815, 1743365578, 1925268497

https://ebooknice.com/product/biota-grow-2c-gather-2c-cook-6661374

(Ebook) History of Technology: Volume 29. Technology in China by


Ian Inkster (Editor) ISBN 9781441136114, 1441136118

https://ebooknice.com/product/history-of-technology-volume-29-technology-
in-china-2252790

(Ebook) Boeing B-29 Superfortress ISBN 9780764302725, 0764302728

https://ebooknice.com/product/boeing-b-29-superfortress-1573658
(Ebook) Jahrbuch für Geschichte: Band 29 ISBN 9783112622223,
3112622227

https://ebooknice.com/product/jahrbuch-fur-geschichte-band-29-50958290

(Ebook) Harrow County 29 by Cullen Bunn, Tyler Crook

https://ebooknice.com/product/harrow-county-29-53599548

(Ebook) 29, Single and Nigerian by Naijasinglegirl ISBN


9781310004216, 1310004218

https://ebooknice.com/product/29-single-and-nigerian-53599780

(Ebook) Organometallic Chemistry, Volume 29 by M. Green ISBN


0854043284

https://ebooknice.com/product/organometallic-chemistry-volume-29-2440106

(Ebook) Atlas of Diabetes Mellitus, 4e (Dec 29,


2023)_(1032379456)_(CRC Press) by Scobie, Ian N., Hopkins, David
ISBN 9781032379456, 1032379456

https://ebooknice.com/product/atlas-of-diabetes-
mellitus-4e-dec-29-2023-1032379456-crc-press-54238270
Pharmacogenetics
EDITED BY

Ian P. Hall
University Hospital of Nottingham
Nottingham, U.K.

Munir Pirmohamed
University of Liverpool
Liverpool, U.K.

New York London

Taylor & Francis is an imprint of the


Taylor & Francis Group, an informa business
Foreword

Pharmacogenetics seeks to determine how a person’s genetic makeup affects their


response to medicines. Rapid advances in genetic technologies have opened up the possi-
bility of genetic testing on a large scale at an affordable cost, thus facilitating the exploita-
tion of pharmacogenetics not only in assessing risks, benefits, and indications of
medicines, but also as a valuable tool in drug discovery.
Since the beginning of modern drug therapy, it has been realized that dose is a
poor predictor of therapeutic response and that there is substantial variability in both
therapeutic efficacy and the occurrence of adverse effects of most medicines. One of
the main aims of clinical pharmacology is to understand the basis of this variability.
Whether it is due to differences in drug disposition caused by variations in absorption,
distribution, transport, metabolism, or elimination, or by differences in end-organ
responsiveness, many studies have shown that genetic variability is a most important
determinant of each of these.
In the field of drug discovery, the application of genetics was originally focused on
the discovery of novel disease genes that could become the targets for new drug discovery
programs. But the rapid expansion of understanding of the variability of human DNA
sequences has given rise to an appreciation that this approach may be more challenging
than originally thought. Currently, more use is made of population-based genetic
association studies to identify disease intervention targets.
As the recent report of the Royal Society “Personalised Medicines: Hopes and
Realities” makes abundantly clear, it is easy to exaggerate the importance of pharma-
cogenetics on clinical practice. While there are a few medicines on the market where
there is evidence for the benefits of genetic testing for patient selection, mainly in the
field of cancer, there are as yet no marketed products that are the result of genetic-
based discovery. It seems likely that this situation will radically change, but the time
scale is not yet clear. It seems more likely that, for the foreseeable future, the impact
of pharmacogenetics will be most profound in the development of diagnostic tests that
will allow for the better use of medicines, both in terms of efficacy and safety.
This book is a timely account of the state of the science and clinical application
of pharmacogenetics. Its format should make it attractive and easy to use for the clinician

iii
iv Foreword

in search of detailed information about specific disease areas, and also for those wishing
to be updated in the field of pharmacogenetics in general.

Alasdair Breckenridge
Medicines and Healthcare Products Regulatory Agency
London, U.K.
Preface

This book aims to provide a comprehensive account of pharmacogenetics with particular


reference to its potential role in specific disease areas. While there have been previous
books on pharmacogenetics, these have concentrated on the broader principles (often
almost exclusively dealing with pharmacokinetic issues). We felt that a book that provides
an account of the generic issues and then provides a disease-area-by-disease-area account
would be of value.
This approach has an obvious advantage in that a reader specifically interested in
the possible role of pharmacogenetics in dealing with patients with cardiovascular
disease, for example, will be able to find all the relevant information in a single
chapter, as opposed to having to read about specific enzymes or receptor systems and
pick out those bits of information relevant to the disease area. It has, however, caused
some difficulty in editing the text because obviously some issues (e.g., the role of
cytochrome P450 polymorphism) are generic to many disease areas. In general, we left
essential information in each chapter to ensure that the reader can gain all the relevant
details without constantly having to cross-refer to other chapters. This means that there
is a small amount of repetition in some of the chapters, but wherever possible we have
reduced this to the minimum required to maintain the flow of the text.
The other obvious difficulty in this fast-moving subject area is ensuring that the
account is up to date. While many of the broad issues have not changed over recent
years, many new studies are underway. We tried to ensure that the book is as comprehen-
sive as possible.
Looking through the contents readers will note that there is no specific chapter
dealing with neurological disease; however, the relevant issues are dealt with either in
the chapter on adverse drug reactions (anticonvulsants) or psychiatric disease. Similarly,
the relatively small amount of information on renal disease pharmacogenetics is covered
in the sections on metabolic and cardiovascular disease.
We hope that this book provides a useful starting point for both clinicians and
non-clinicians with an interest in pharmacogenetics. This is an exciting time to be
involved in the study of pharmacogenetics. Although the concept of personalized prescribing
has been around for about 50 years, the next 10 years will prove whether or not this approach
to improving the effectiveness of prescribing by achieving high efficacy while reducing the
risk of severe adverse drug reactions is a viable generic approach to patient management.

Ian P. Hall
Munir Pirmohamed
v
Contents

Foreword Alasdair Breckenridge . . . . iii


Preface . . . . v
Contributors . . . . xi

Part I: Principles of Pharmacogenetics


1. Identification of Treatment Response Genes . . . . . . . . . . . . . . . . . . . . . . . . 1
Eva Halapi and Hakon Hakonarson
Introduction . . . . 1
Pharmacogenetics of Metabolizing Enzymes: CYP450 and
Drug Transporter Genes . . . . 2
Pharmacogenetic Targets in Cancer . . . . 3
Pharmacogenetic Targets in Central Nervous System Disorders . . . . 6
Pharmacogenetic Targets in Respiratory Disorders . . . . 8
Pharmacogenetic Targets in Cardiovascular Disease . . . . 9
Microarray and Disease Response Genes . . . . 10
Conclusion . . . . 12
References . . . . 13

2. Methods of Genotyping . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Matthias Wjst and Monika Werner
Introduction . . . . 21
High-Throughput Genotyping Methods . . . . 22
Test Principles . . . . 22
50 Nuclease Assay . . . . 27
Special Genotyping Applications . . . . 30
Future Developments . . . . 32
References . . . . 32

3. Pharmacoproteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
Alaisdair C. Stamps and Jonathan A. Terrett
Introduction . . . . 35

vii
viii Contents

Historical Perspective . . . . 35
Proteomic Techniques . . . . 38
Current Activities in Pharmacoproteomics . . . . 43
Conclusions . . . . 47
References . . . . 47

4. Potential Social, Ethical, and Legal Issues Raised by the Development


of Pharmacogenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
Paul Martin, Andrew Smart, and Robert Dingwall
Introduction—Assessing the Potential Impact of
Pharmacogenetics . . . . 51
Bioethics and Its Limitations . . . . 52
Possible Application of Pharmacogenetics . . . . 53
Social and Ethical Issues . . . . 56
Conclusion: The Social and Ethical Issues Raised by
Pharmacogenetics—Implications for Policy . . . . 62
References . . . . 63

Part II: Clinical Practice and Pharmacogenetics


5. Pharmacogenetics of Adverse Drug Reactions . . . . . . . . . . . . . . . . . . . . . . 65
Ana Alfirevic, B. Kevin Park, and Munir Pirmohamed
Introduction . . . . 65
Definitions and Classification of Adverse Drug Reactions . . . . 66
Historical Overview . . . . 66
Variability in Drug Handling and Adverse Drug Reactions . . . . 67
Genetic Variability in Drug Metabolizing Enzymes . . . . 67
Genetic Variability in Drug Transporters and Adverse
Drug Reactions . . . . 73
Immunogenetic Polymorphisms . . . . 74
Receptors . . . . 76
Miscellaneous Drug Targets . . . . 77
Summary and Future Directions . . . . 78
References . . . . 80

6. Pharmacogenetics of Respiratory Disease . . . . . . . . . . . . . . . . . . . . . . . . . . 91


Ian P. Hall and Stephen B. Liggett
Introduction . . . . 91
b2 Adrenoceptor Polymorphism . . . . 91
Clinical Consequences of b2AR Polymorphisms . . . . 98
Pharmacogenetics of Theophylline . . . . 102
Cys Leukotriene Receptor Pathway Polymorphism . . . . 103
Glucocorticoids and Treatment Response . . . . 105
Pharmacogenetics of Muscarinic Receptor Antagonists . . . . 106
Summary . . . . 106
References . . . . 107

7. Hematological Pharmacogenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111


Howard L. McLeod, Charles Eby, and Brian Gage
Introduction . . . . 111
Contents ix

Warfarin Pharmacogenetics . . . . 112


Genetic Polymorphisms in Hemostatic Systems . . . . 116
Thiopurine Pharmacogenetics . . . . 121
The Future . . . . 123
References . . . . 123

8. Pharmacogenetics in Oncology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129


Romano Danesi, Antonello Di Paolo, Mario Del Tacca, and
Filippo De Braud
Introduction . . . . 129
Pharmacogenetics of Drug Targets: Theoretical Background . . . . 131
Pharmacogenetics of Drug Targets and
Enzymes of Drug Metabolism . . . . 133
Pharmacogenetics of DNA Repair Systems
and Apoptosis . . . . 141
The ATP-Binding Cassette Superfamily of Drug
Transporters . . . . 144
Clinical Pharmacogenetics . . . . 144
Conclusions and Future Prospects . . . . 147
References . . . . 147

9. Pharmacogenetics in Infectious Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . 155


Belle L. Lee, Laurent A. Decosterd, Reinhold Kerb, and
Amalio Telenti
Introduction . . . . 155
Pharmacogenetics of Specific Infectious Diseases . . . . 157
Other Pharmacogenetic Markers of Interest . . . . 169
Immunogenetics in Infectious Diseases . . . . 175
Validation of Pharmacogenetic Markers . . . . 176
Conclusions . . . . 177
References . . . . 178

10. Pharmacogenetics in Rheumatology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189


Samantha L. Hider, Catharine Morgan, Wendy Thomson, and
Ian N. Bruce
Introduction . . . . 189
Therapeutic Options in the Treatment of Rheumatoid
Arthritis . . . . 189
Efficacy and Toxicity of Antirheumatic Therapies . . . . 190
Nonsteroidal Anti-Inflammatory Drugs . . . . 191
Methotrexate . . . . 193
Sulfasalazine . . . . 195
D- Penicillamine . . . . 195
Gold . . . . 196
Azathioprine . . . . 196
Leflunomide . . . . 198
Antimalarial Drugs . . . . 199
Biological Therapies . . . . 200
Summary and Future Directions . . . . 202
References . . . . 202
x Contents

11. Polymorphisms in Cardiovascular Medicine: The Role of Genetic


Variants in Disease Diagnosis and Drug Response . . . . . . . . . . . . . . . . . . 209
Yasmin, David N. Cooper, and John R. Cockcroft
Introduction . . . . 209
Polymorphism in the Human Genome . . . . 210
Polymorphism in Cardiovascular Disease . . . . 211
Genetic Polymorphisms and Response to Drugs . . . . 225
A Perspective on Pharmacogenetics in
Cardiovascular Medicine . . . . 232
References . . . . 232

12. Pharmacogenetics and Metabolic Disease . . . . . . . . . . . . . . . . . . . . . . . . . 243


Simon Constable and Munir Pirmohamed
Introduction . . . . 243
Diabetes Mellitus . . . . 243
Hyperlipidemia . . . . 251
Osteoporosis . . . . 257
Conclusions . . . . 261
References . . . . 262

13. Pharmacogenetics in Gastroenterology . . . . . . . . . . . . . . . . . . . . . . . . . . . 273


Guruprasad Padur Aithal
Introduction . . . . 273
Application of Pharmacogenetics in Clinical Gastroenterology
and Hepatology . . . . 273
Candidate Gene Case – Control Association Studies . . . . 284
Future Developments . . . . 286
References . . . . 286

14. The Genetics of Antipsychotic Response: Pharmacogenetic and


Pharmacogenomic Investigations and Clinical Applications . . . . . . . . . . . 295
Maria J. Arranz, Dalu Mancama, and Robert W. Kerwin
Introduction . . . . 295
Pharmacogenetic and Pharmacogenomic Research . . . . 295
Clinical Applications of Pharmacogenetics
and Pharmacogenomics . . . . 302
Future Research: Investigations on Genetic Expression . . . . 304
References . . . . 304

Index . . . . 311
Contributors

Guruprasad Padur Aithal Queen’s Medical Centre University Hospital,


Nottingham, U.K.
Ana Alfirevic Department of Pharmacology and Therapeutics, University of
Liverpool, Liverpool, U.K.
Maria J. Arranz Section of Clinical Neuropharmacology, Institute of
Psychiatry—King’s College, London, U.K.
Ian N. Bruce arc Epidemiology Unit, University of Manchester, Manchester, U.K.
John R. Cockcroft Wales Heart Research Institute, Cardiff University, Cardiff, U.K.
Simon Constable Department of Pharmacology and Therapeutics, University
of Liverpool, Liverpool, U.K.
David N. Cooper Institute of Medical Genetics, Cardiff University, Cardiff, U.K.
Romano Danesi Division of Pharmacology and Chemotherapy, Department of
Oncology, University of Pisa, Pisa, Italy
Filippo De Braud Clinical Pharmacology and New Drug Development Unit,
European Institute of Oncology, Milano, Italy
Laurent A. Decosterd Division of Clinical Pharmacology, University of Lausanne,
Lausanne, Switzerland
Mario Del Tacca Division of Pharmacology and Chemotherapy, Department
of Oncology, University of Pisa, Pisa, Italy
Antonello Di Paolo Division of Pharmacology and Chemotherapy, Department
of Oncology, University of Pisa, Pisa, Italy
Robert Dingwall Institute for the Study of Genetics, Biorisks and Society,
University of Nottingham, Nottingham, U.K.
Charles Eby Department of Medicine, Washington University School of Medicine,
Saint Louis, Missouri, U.S.A.
Brian Gage Department of Medicine, Washington University School of Medicine,
Saint Louis, Missouri, U.S.A.
xi
xii Contributors

Hakon Hakonarson Division of Inflammation and Pharmacogenomics, deCODE


Genetics, Inc., Reykjavik, Iceland
Eva Halapi Division of Inflammation and Pharmacogenomics, deCODE
Genetics, Inc., Reykjavik, Iceland
Ian P. Hall Division of Therapeutics and Molecular Medicine, University Hospital
of Nottingham, Nottingham, U.K.
Samantha L. Hider arc Epidemiology Unit, University of Manchester,
Manchester, U.K.
Reinhold Kerb Experimental Medicine, Astra Zeneca, Mölndal, Sweden
Robert W. Kerwin Section of Clinical Neuropharmacology, Institute of
Psychiatry—King’s College, London, U.K.
Belle L. Lee University of California, San Francisco, California, U.S.A., and Division
of Infectious Diseases, Department of Clinical Research, University Hospital, Berne,
Switzerland
Stephen B. Liggett Departments of Medicine and Physiology, University of
Maryland School of Medicine, Baltimore, Maryland, U.S.A.
Dalu Mancama Section of Clinical Neuropharmacology, Institute of
Psychiatry—King’s College, London, U.K.
Paul Martin Institute for the Study of Genetics, Biorisks and Society, University
of Nottingham, Nottingham, U.K.
Howard L. McLeod Department of Medicine, Washington University School
of Medicine, Saint Louis, Missouri, U.S.A.
Catharine Morgan arc Epidemiology Unit, University of Manchester,
Manchester, U.K.
B. Kevin Park Department of Pharmacology and Therapeutics, University of
Liverpool, Liverpool, U.K.
Munir Pirmohamed Department of Pharmacology and Therapeutics, University
of Liverpool, Liverpool, U.K.
Andrew Smart Institute for the Study of Genetics, Biorisks and Society, University
of Nottingham, Nottingham, U.K.
Alaisdair C. Stamps UCB-Celltech, Slough, U.K.
Amalio Telenti Institute of Microbiology, University Hospital, University
of Lausanne, Lausanne, Switzerland
Jonathan A. Terrett Medarex, Inc., Milpitas, California, U.S.A.
Wendy Thomson arc Epidemiology Unit, University of Manchester,
Manchester, U.K.
Contributors xiii

Monika Werner GSK Glaxo Smith Kline Beecham, Munich, Germany


Matthias Wjst Gruppe Molekulare Epidemiologie, GSF—Forschungszentrum für
Umwelt und Gesundheit, Neuherberg/Munich, Germany
Yasmin Clinical Pharmacology Unit, University of Cambridge, Cambridge, U.K.
PART I: PRINCIPLES OF PHARMACOGENETICS

1
Identification of Treatment
Response Genes
Eva Halapi and Hakon Hakonarson
Division of Inflammation and Pharmacogenomics, deCODE Genetics, Inc.,
Reykjavik, Iceland

INTRODUCTION

Genetic diversity contributes to both disease susceptibility and variability in response to


drug therapy. Pharmacogenomics is a discipline focused on examining the genetic basis
for individual variations in response to therapeutics (1 – 4). Although the task of develop-
ing individualized medicines tailored to patient’s genotypes poses a major scientific chal-
lenge, pharmacogenomics is already starting to influence how physicians/scientists design
clinical trials and its impact on the practice of medicine is forthcoming (5,6). Recent evi-
dence suggests that most prescribed medications are effective in no more than 60% of the
individuals in whom they are used, and a significant number of patients also develop major
adverse effects. Better understanding of the genetic factors that regulate patient’s respon-
siveness to drugs is therefore needed to elucidate the molecular mechanisms involved and
allow for development of new therapeutic strategies that match each patient and the most
suitable drug (7 – 9).
While drug treatment constitutes the mainstay of medicine, for most drugs, there is
considerable variability in patient’s therapeutic response (2,3). In other cases, unforeseen
serious side effects may occur (10,11). For the patient this represents a dangerous and
potentially life-threatening situation, and, at the societal level, adverse drug reactions
are the most common cause of hospital admissions in the elderly and represent a
leading cause of disease and death (12). In some cases, genetic variations have been
shown to influence both efficacy and safety profiles, as in the case of dicumarol, warfarin,
or isoniazid, wherein patient variation in response to these drugs can largely be attributed
to polymorphisms in the CYP450 gene family that confer rapid versus slow acetylation of
these drugs (10). Since genetic variations can lead to differences in the regulatory func-
tions of genes, variability in their mRNA and/or protein expressions may follow. Pharma-
cogenomics is charged with measuring these differences in mRNA and protein messages
in response to drugs, and although relatively few examples of success exist, this approach
holds the promise that we may be able to profile these variations in individuals’ genetic
makeup and accurately predict response to drugs addressing both efficacy and safety
issues (3,4,6,9,10).

1
2 Halapi and Hakonarson

In recent years, microarray technology has revolutionized almost all fields of


biomedical research by enabling high-throughput gene expression profiling in a single
experiment, thereby allowing thousands of genes in different species to be examined
in the context of organ differentiation and development in search for disease susceptibility
genes and new targets in drug discovery. With the use of expression microarray, the
future holds the promise that we may soon gain more global understanding of gene
expression changes with respect to disease susceptibility and progression and that
biomarkers will become increasingly used as diagnostic and prognostic indicators of
treatment response and also provide new insights into the process of new target discovery.
A brief overview of some of the key examples of pharmacogenetic effects follows.
This is designed to give the reader an initial idea of the potential for pharmacogenetic
effects to contribute to disease management. Fuller accounts of the range of pharmacoge-
netic effects relevant to each major disease group can be found in the chapters that follow.

PHARMACOGENETICS OF METABOLIZING ENZYMES: CYP450 AND


DRUG TRANSPORTER GENES

The cytochrome P450 (CYP450) enzyme system is involved in various metabolic and bio-
synthetic processes and constitutes a superfamily of heme enzymes found in most organ-
isms (i.e., bacteria to humans). These enzymes are estimated to account for the
biotransformation of approximately 60% of the most commonly prescribed drugs in the
United States. A few representative examples are discussed in the following, and
additional details on some of these examples (and others) are contained in later chapters.

CYP2D6
Three main phenotypes have been identified that relate to the oxidative metabolisms of
drug substrates by CYP2D6. The slow metabolizers (with defective CYP2D6 alleles),
the normal metabolizers (wild type), and the ultra-rapid metabolizers all have variable
number of genes for the functional CYP2D6 enzyme (13 –15). Within the Caucasian popu-
lation, approximately 7% of the CYP2D6 alleles are defective, resulting in potentially
increased concentration of various drug metabolites at conventional therapeutic doses.
Some of the most commonly used drugs, such as the beta-adrenoceptor blockers and tri-
cyclic antidepressants, are substrates for CYP2D6 (13 – 15). The latter are known to cause
adverse reactions that are attributed to increased levels of tricyclic antidepressants. These
include, but are not limited to, such events as life-threatening arrhythmias and other
cardiotoxic effects that result from decreased activity of CYP2D6 metabolism. Diagnostic
tests are now available to identify beforehand those who are at risk.

CYP2C9
Three defective alleles have been reported in the CYP2C9 enzyme, two of which
confer decreased activity. Their frequency ranges from 1% to13% in different populations
(13 –15). Substrates of CYP2C9 include nonsteroidal anti-inflammatory drugs and hypo-
glycemic agents (13 – 15). The clinical relevance of CYP2C9 is particularly noticed in the
metabolism of drugs that are used to treat Type II diabetes, wherein decreased clearance of
these drugs may result in severe hypoglycemia. Another relevant example is S-warfarin,
where major bleeding may occur (10).
Identification of Treatment Response Genes 3

CYP2C19
CYP2C19 is a highly polymorphic enzyme system, with approximately 3% of the Cauca-
sians having allele variation that renders them as slow metabolizers. In contrast, almost
20% of the Asian population carries the slow allele. Allele 9 confers total abolishment
of enzymatic activity (13 – 15). For most pharmacological compounds that are substrates
for CYP2C19, this poses a limited problem because the majority of the drugs are metab-
olized by several CYP450 enzymes. An important exemption is the drug omeprazole,
which is only partially metabolized by CYP3A4 and shows up to 12-fold larger area
under the concentration – time curve (AUC) in slow versus fast metabolizers (13 –15).

Drug Transporters
Blood and tissue concentrations of most drugs are influenced by interindividual variation
in the structure and function of the metabolizing enzyme and transporter genes. Transpor-
ters are genes that control drug uptake, distribution, and elimination. The multidrug resist-
ance gene (MDR1) encodes for a P-glycoprotein (PgP), which belongs to the large
adenosine triphosphate (ATP)-binding cassette (ABC) protein. The MDR1 gene was orig-
inally discovered as the protein causing cross-resistance of tumors to many different cyto-
toxic agents (16). Multiple substrates are transported by PgP, including the
chemotherapeutics tamoxifen and mitoxantrone, the antibiotics cefotetan and cefazolin,
the immunosuppressant cyclosporin A, the antiarrytmic drug quindine, the cardiac stimu-
lants digoxin, and such opioid drugs as morphine, to name a few (17). Many cancers are
known to overexpress the PgP protein, and this has been correlated with poor prognosis,
particularly in patients with leukemia (18). Several SNPs have been reported in the MDR1
gene, some of which have been correlated with PgP protein expression, notably including
the C/T polymorphism in exon 26 (19). In a recent study (20), antiviral response to nelfi-
navir and efavirenez was shown to correlate with the allelic variant, 3435C/T, of the
MDR1 gene. Patients who were homozygotes, carrying two copies of the 3435 T allele,
demonstrated lower serum concentration, faster recovery in CD4 T cell count, and more
rapid decrease in viral load, suggesting that the MDR1 3435C/T variant may be predictive
of immune recovery after antiviral treatment in HIV patients (20). Approximately 50% of
Caucasians are heterozygote (C/T) at the 3435 MDR1 polymorphic site, while a homozy-
gote state (C/C) or (T/T), is seen in 25% of individuals, respectively. In contrast, the
frequency of the CC genotype in African Americans is 67% to 83%, whereas the fre-
quency of the TT allele is very low. Increased expression of PgP has also been correlated
with variation in clinical response to glucocorticoids in patients with inflammatory bowel
disease (21) and systemic lupus erythematosus (22). Collectively, these studies suggest
that there are important variations in the MDR1 gene that regulate tumor resistance,
immune function, and metabolism of multiple drugs.

PHARMACOGENETIC TARGETS IN CANCER

For most cancers, conventional histopathologic evaluation, encompassing tumor


grade and stage, is inadequate to accurately predict the biological behavior of the tumor
(23 –25). Considerable effort is underway to identify and characterize the biological
potential of various cancers at the molecular level. The need to predict response to
therapy and determine which tumors are most likely to progress or recur or which invasive
4 Halapi and Hakonarson

tumors will metastasize has prompted intensive efforts in search for prognostic biomarkers
and markers that correlate with patients’ responses to anticancer therapy.

Estrogen and Progesterone Receptors


In the United States and other western societies, approximately one in 10 women develops
breast cancer. Given the unacceptably high mortality rate of approximately 40%, it is cri-
tically important that the most effective therapy at any given time is administered to each
patient. Although both adjuvant chemo- and hormonal therapies reduce the risk of metas-
tasis by approximately one-third, the best indicators for clinical progression available
today, including lymph node status, tumor size and histological grade, are unable to accu-
rately predict the outcome.

SERM
The presence of estrogen and/or progesterone receptors on tumors is considered favorable
because these patients are eligible for hormonal treatment. Tamoxifen, is a selective estro-
gen-receptor modulator (SERM), which acts as an estrogen antagonist in normal breast
tissue and breast cancer cells but as an antagonist in liver and bone cells. Apart from low-
ering serum cholesterol and preventing postmenopausal osteoporosis (26), tamoxifen is
the most effective and extensively used hormonal treatment for all stages of breast
cancer. More recently, the drug was approved for prevention of breast cancer in high-
risk individuals. In a recent meta-analysis, which included information on 37,000
women in 55 clinical trials of adjuvant tamoxifen therapy for five years (27), reduction
in recurrence and mortality rates were 47% and 26%, respectively, over a 10-year
period. Fifty percent decrease was also observed in the incidence of contralateral breast
cancer in patients receiving tamoxifen, regardless of the ER status of the primary tumor
(27). Several SERMs are currently in clinical trials. Toremifene is a relatively new
SERM drug with properties similar to that of tamoxifen. However, unlike tamoxifen, tor-
emifene does not seem to increase the risk of endometrial cancer. Based on information
available to date, the Food and Drug Administration (FDA) has restricted the use of tor-
emifene to postmenopausal women with metastatic breast cancer. Raloxifene is another
antiestrogen SERM that has received approval for the treatment of osteoporosis in
women beyond menopause.

Aromatase Inhibitors
Aromatase, a cytochrome P-450 enzyme that catalyzes the conversion of androgens to
estrogens, is the major source of estrogen synthesis in postmenopausal women. Inhibition
of aromatase, the terminal step in estrogen biosynthesis, provides a mechanism to inter-
vene in hormone-dependent breast cancer in postmenopausal women. Compared to
tamoxifen, both nonsteroidal (such as anastrozole and letrozole) and steroidal (such as
exemestane) aromatase inhibitors (AIs) provide superior efficacy and better toxicity
profile as first- and second-line therapy of metastatic disease. Early results from the
ATAC study (anastrozole, tamoxifen, alone or in combination trial), encompassing
9300 women with early-stage disease at 381 research and medical centers in 21 countries,
suggest that anastrozole is superior to tamoxifen as measured by disease-free survival in
receptor-positive patients and in reducing the incidence of contralateral breast cancer (28).
Identification of Treatment Response Genes 5

ER Antagonists
Fulvestrant (faslodex) is a potent antiestrogen drug that mediates its effects by estrogen
receptor (ER) downregulation. It acts as a pure antiestrogen and exhibits none of the nega-
tive side effects associated with the partial agonist activity of tamoxifen and related drugs.
It has been shown to have comparable efficacy to that of the oral AI, anastrozole, in post-
menopausal women with advanced breast cancer who have failed to respond to tamoxifen
or related drugs (29). It therefore provides the clinician with an alternative therapeutic
strategy following the development of tamoxifen resistance. Fulvestrant might also be
beneficial as a follow-on therapy after tamoxifen in an adjuvant setting to palliate some
of the concerns surrounding a long-term therapy with tamoxifen (five years).
Antiestrogens are among the most potent therapies in preventing cancer and reducing
the risk of recurrence in high-risk patients and in treating metastatic disease. Thus, ER
expression has become a valuable marker in predicting treatment response in breast cancer.

Epidermal Growth Factor Receptor Family


Epidermal Growth Factor Receptor
The epidermal growth factor (EGF) pathway has been identified as a key regulator of cell
growth and replication (30,31). Cumulative evidence shows that the epidermal growth
factor receptor (EGFR) pathway is actively involved in a wide variety of solid tumurs,
including non– small cell lung cancer (NSCLC), prostate cancer, breast cancer, stomach
cancer, colon cancer, ovarian cancer, and tumors of the head and neck (30 – 32). Overex-
pression of the EGFR in cancer cells has been associated with more advanced disease,
development of metastatic phenotypes, and poor prognosisents (32). IressaTM , a new tyro-
sine kinase inhibitor, directly blocks the signals for cell growth and division and is cur-
rently licensed for the treatment of inoperable or recurrent NSCLC in Asia and is being
tested in clinical trials for other solid tumors (33). Other drugs targeting the EGFR are
TarcevaTM and ErbituxTM , which are currently in clinical trials.

HER2/neu
HER2/neu has been shown to be overexpressed in 20% to 30% of breast cancer patients
(34,35). Recent evidence supports a clear association between HER2 overexpression and
reduced overall and disease-free survival, especially in patients with node-positive
disease (34 –38). Tumors displaying HER2 amplification show a correlation with poor
prognosis (39,40). Thus, the greatest value of HER2 as a predictive marker lies in the pre-
diction of response to therapies that target HER2, notably herceptin. Indeed, patients with
strongly HER2-positive breast cancer get significant clinical benefits from herceptin
therapy, and HER2 testing has become an integral part of the optimal management of
breast cancer patients. It is important to determine HER2 status of all primary breast
cancers at the time of diagnosis and recurrence because HER2 overexpression and ampli-
fication can be used to identify patients for herceptin therapy (41). Thus, HER2 has
approached a clinically validated status as a prognostic factor and also as a predictive
factor for response to therapy, and it is already part of the routine assessment for breast
cancer patients. A prior knowledge of HER2 status is therefore an absolute requirement
for herceptin therapy.

Thiopurine S-Methyltransferase
The thiopurine S-methyltransferase (TPMT ) gene metabolizes thiopurine medications,
such as mercaptopurine, azathioprine, and thioguanine. TPMT activity is polymorphic
6 Halapi and Hakonarson

with 10% of the subjects being heterozygous and about 1/300 with low or deficient
activity (42,43). Patients with low or deficient TPMT activity are at high risk for severe
hematological toxicity from standard doses of thiopurine medications. Thus, it is import-
ant to be able to identify those patients who are at risk for such complications. The mol-
ecular basis for altered TPMT activity is well characterized with three distinct alleles
accounting for up to 95% of both heterozygous and homozygous mutant patients (43).
However, significant ethnic differences have been identified in both the frequency of
low TPMT activity and in the mutations that account for them (43). Although extended
clinical studies are needed to better understand and quantitate the dosing of these drugs,
TPMT genotype-specific dosing guidelines have been proposed for the use of mercapto-
purine in leukemia patients (44). Given the severity of thiopurine medication-related tox-
icity in TPMT-deficient patients, screening of the entire patient populations for TPMT
polymorphism prior to prescribing these drugs has been shown to be cost-effective. By
determining thiopurine transferase activity in patients before they receive thiopurines,
enzyme efficacy can be determined beforehand and life-threatening complications,
which are strongly related to the genetically determined activity of this enzyme, can be
avoided. Accordingly, screening for genetic variations in the TPMT gene presents an
ideal model for the translation of genomic information to guide patient therapeutics.

N-Acetyltransferase (NAT)
NAT was first identified as the enzyme responsible for inactivation of the antitubercular
drug isoniazid (45). NAT also plays an important role in carcinogen metabolism. The
N-acetylation metabolizing pathway is a major route for the conjugative metabolism
of many drugs and chemicals (46). Functional polymorphisms in the NAT gene were
initially associated with differences in the susceptibility to occupational and smoking-
related bladder cancer (47). Based on the substrate, individuals can be phenotyped as
either “fast” or “slow” acetylators. Individuals with the slow phenotype are homozygotes
for the slow allele, whereas subjects with the fast phenotype are either heterozygotes or
homozygotes for the fast allele. The frequency of slow acetylator varies worldwide,
ranging from 5% to 10% in Asia and reaching 90% frequency in certain European popu-
lations (13). Two functionally relevant human NAT genes, NAT1 and NAT2, have been
identified that are highly polymorphic and are encoded at multi-allelic loci. The relation-
ship between these polymorphisms and the resulting phenotypes is well established (48).

PHARMACOGENETIC TARGETS IN CENTRAL NERVOUS


SYSTEM DISORDERS

Although pharmacotherapy in patients with affective disorders has improved the outcome
of millions of patients worldwide, medical treatment of mental depression is efficacious in
no more than two-thirds of the cases, and there are no biological markers of treatment
response. Apart from identification of genomic markers of treatment response, which
would constitute an enormous clinical advantage of public health value, the application
of pharmacogenmoics may also uncover new targets for the development of novel and
hopefully more efficacious drugs with favorable side-effect profile.

Serotonin Transporter
Selective serotonin reuptake inhibitors (SSRIs) are widely used for the treatment of
depression because of their efficacy and relatively favorable side-effect profile compared
Identification of Treatment Response Genes 7

with those of the tricyclic antidepressants. SSRIs act by interfering with the activity of the
serotonin transporter (SERT) (49). A number of polymorphisms have been reported in the
SERT gene (50 – 52), and genetic variations in the SERT promoter have been linked to
altered functions, such as the association between the short (S) allele (44 bp deletion)
of the SERT-PR site and poor response to fluvoxamine and paroxetine in patients
treated for major depression (53,54).

Dopamine Transporter
Attention deficit/hyperactivity disorder (ADHD) affects between 3% and 5% of school-
age children. Treatment of major symptoms, such as inattention, hyperactivity, and
impulsive behavior, has been effectively achieved using psychostimulants, of which
methylphenidate (Ritalinw) is the most commonly prescribed in the United States.
These drugs are beneficial in many cases; however, inter-individual variation in clinical
response and adverse events is well documented (55 –57). Methylphenidate binds to
and directly inhibits the dopamine transporter (DAT1). Accordingly, variations in genes
involved in dopamine action and metabolism (such as DAT1, D2, and the D4 receptor
genes) have been examined in search for explanation of the variability in clinical response
to methylphenidate and other psychostimulants in ADHD patients. A recent study (58)
demonstrated significant association between the 10/10 genotype in DAT1 and lack of
response to methylphenidate but failed to demonstrate any association with polymorphism
in the dopamine receptors (58). These results have recently been confirmed in a larger
patient cohort (59).

Dopamine and Serotonin Receptors


Clozapine is a potent drug in the treatment of schizophrenia; however, not all patients
benefit from treatment, and some patients react adversely to therapy while others fail to
respond adequately. Several studies have reported an association between clozapine
response and polymorphisms in the dopamine receptor 3 (D3) gene (60 –62), dopamine
receptor 4 (D4) gene (63,64), and in the serotonin receptor 2A (65,66) and 5A genes
(67). In a recent study, Arranz et al. (68) screened for the combination of upto 19 poly-
morphisms that predicted clinical response to clozapine in schizophrenic patients with
high accuracy. These 19 single-nucleotide polymorphisms (SNPs) were located in eight
receptor genes and one transporter gene, including the a2A-adrenoceptor, dopamine D3
receptor, 5-HT2A, 5-HT2C, 5-HT3A, 5-HT5A, histamine H1 receptor, histamine H2
receptor, and the serotonin transporter gene. A combination of six polymorphisms, includ-
ing the 5-HT2A 102T/C, His452Tyr, 5-HT2C-330GT/-244CT, Cys23Ser, 5-HTTLPR,
H2-1018G/A, predicted clinical response with 76.86% accuracy (x2 ¼ 35.8; P ¼0.0001)
with sensitivity of 95.89 (+0.04), suggesting that these polymorphisms could be used to
identify patients who are most likely to show a satisfactory improvement with treatment.

Apolipoprotein E
Apart from its role in cardiovascular disease, the apoliprotein E (ApoE) protein has also been
associated with late onset and sporadic Alzheimer’s disease (AD) (69,70). In the central
nervous system (CNS), ApoE plays a key role in mobilization and redistribution of choles-
terol and phospholipids during membrane remodeling (70). In a study assessing the influence
of the ApoE genotype on therapeutic response to tacrine (acetylcholinesterase inhibitor) in a
responder/nonresponder cohort, more than 80% of ApoE4 negative carriers demonstrated
8 Halapi and Hakonarson

marked clinical response to tacrine, as measured by the AD assessment scale (ADAS),


whereas ApoE4 positive carriers presented ADAS scores that were worse compared with
the baseline levels (69). The ApoE genotype has also been implicated in modulating response
to drugs targeting vasopressinergic activity (71). Thus, ApoE is emerging as a potentially
useful marker in predicting clinical response to tacrine.

PHARMACOGENETIC TARGETS IN RESPIRATORY DISORDERS

There are four major classes of asthma pharmacotherapy currently in widespread use (72):
(i) b2-agonists (b-agonist) used by inhalation for the relief of airway obstruction
(e.g., albuterol, salmeterol, fenoterol); (ii) glucocorticoids for both inhaled and systemic
use (e.g., fluticazone, beclomethasone, triamcinolone, prednisone); (iii) theophylline
and its derivatives; and (iv) inhibitors and receptor antagonists of the cysteinyl-
leukotriene pathway (e.g., montelukast, pranlukast, zafirlukast, zileuton). The following
section summarizes pharmacogenetic work reported on b-agonists and inhibitors of the
cysteinyl-leukotriene pathway in asthma; comprehensive accounts of these effects are
contained in the chapter on respiratory disease.

b2 Adrenergic Receptor
Four polymorphisms of the coding block of b2 adrenergic receptor (b2AR) have been
found, three of which result in receptors that have different properties compared with
the wild-type (73). These polymorphisms include Arg16 ! Gly, Gln27 ! Glu,
Val34 ! Met, and Thr164 ! Ile, of which the first two are the most common. Most
studies have found no differences in the frequencies of these polymorphisms between
asthma patients and healthy nonasthmatic controls (74,75). Thus, the genetic variability
of the b2AR does not appear to play a major causative role in asthma. However, these poly-
morphisms, although not causative, could modify the disease. Other studies have assessed
the relationship between b2AR polymorphisms at positions 16 and 27 and atopy, including
IgE levels (76). A significant association between the Glu27 form of the b2 receptor and log
serum IgE was reported, suggesting that b2AR polymorphisms may act to modify the asth-
matic phenotype (77).
Several studies have assessed the modulatory role of b2AR polymorphisms on the
treatment response to b-agonists (2,78) in relation to bronchial hyperreactivity (79) and
control of asthma (80). These studies demonstrate that certain b2AR polymorphisms
affect the clinical response to b-agonist therapy, which may impact the asthmatic pheno-
type, rendering these variants candidates that may ultimately provide for individualized
therapy in asthma.

Leukotriene Pathway Pharmacogenetics


The leukotrienes are eicosatetraenoic acid compounds that are derived from arachidonic
acid and exhibit a wide range of pharmacological and physiological actions (81). Three
enzymes are exclusively involved in the formation of the leukotrienes, including the
5-lipoxygenase (ALOX5), the leukotriene C4 synthase (LTC4), and LTA4 epoxide hydro-
lase. ALOX5 is the central enzyme required for the production of both the cysteinyl-
leukotrienes (LTC4, LTD4, and LTE4) and the potent neutrophil chemo-attractant, LTB4.
Drugs that inhibit ALOX5 activity or antagonize the action of the cysteinyl-leukotrienes
at their receptor site have been shown to attenuate broncho-constriction in asthma
patients (82).
Identification of Treatment Response Genes 9

Mutations in the ALOX5 gene were found to have significant functional conse-
quences in the context of promoter – reporter constructs and patients with variable
number of tandem repeats (VNTRs) other than the wild-type (i.e., five repeats of the
sequence 2GGGCGG2 in the core promoter) have been shown to have diminished tran-
scription of the ALOX5 gene and produce lower levels of leukotrienes (83,84).
The leukotriene LTC4 synthase is another enzyme with a known SNP in its promoter
region (A 2444C) with a C allele frequency that is reportedly higher in patients with
severe asthma (71,72), wherein the 2444C variant is associated with enhanced cysteinyl-
leukotriene production, suggesting that patients with the A/A genotype may have leuko-
triene-driven asthma. These findings provide possible evidence that, apart from the
ALOX5, there may be another pharmacogenetic locus that can modulate the leukotriene
pathway. Whether DNA sequence variants that are associated with decrease in cysteinyl-
leukotriene synthesis will also be associated with a decreased response to therapy
remains to be determined.

PHARMACOGENETIC TARGETS IN CARDIOVASCULAR DISEASE

Atherosclerosis is, at least in part, attributed to an underlying immune-mediated process


with onset early in life, ultimately leading to severe clinical manifestations, such as myo-
cardial infarction, unstable angina, and cerebral stroke. The increased incidence of cardio-
vascular events in the western societies is attributed to the underlying immune process,
which is amplified by additional cardiovascular risk factors, such as hypercholesterolemia,
hypertension, smoking, diabetes, and obesity, which by themselves have their own genetic
background. This section provides a summary of some of the key issues; for a full discus-
sion, see the chapter on cardiovascular disease.

Apolipoprotein E
Statins act primarily by inhibiting hydroxy-methylglutaryl coenzyme A (HGM-CoA)
reductase activity (85 –87). Although statins are among the most effective cholesterol-
lowering agents available, hyperlipidemic individuals display marked variability in
lipid-lowering response which may, at least in part, be due to genetic differences (88).
Among the multiple candidate genes that have been shown to be involved in lipid metab-
olism, most attention has been focused on the ApoE locus. ApoE is essential for the normal
catabolism of triglyceride-rich lipoprotein constituents. Three major ApoE isoforms are
encoded by three common alleles at the ApoE locus (89). Genetic variations in the
ApoE gene at the ApoE locus have been associated with plasma lipoprotein concentrations
in both fasting and in the postprandial states (89,90). In this regard, the E2 allele is associ-
ated with lower and the E4 allele with higher total plasma cholesterol and low-density
lipoprotein (LDL) cholesterol levels compared with the E3 allele, whereas ApoE E2 car-
riers have been reported to be more responsive to lipid-lowering statin therapy (91). The
ApoE E4 allele has been shown in some studies to be associated with increased response to
dietary intervention.

Angiotensin-Converting Enzyme
Angiotensin-converting enzyme (ACE) plays a critical role in blood pressure regulation.
Restenosis after coronary artery stent is a major health problem that is primarily attributed
to intimal hyperplasia, which can be attenuated by ACE inhibitors. Identification of patients
who are at higher risk of developing restenosis is therefore desirable. In this regard, an
10 Halapi and Hakonarson

insertion/deletion (I/D) polymorphism in intron 16 of the ACE gene has attracted signifi-
cant attention due to its correlation with serum ACE activity (92). An association was
recently reported between carriers of the D allele and reduced risk of restenosis following
coronary stenting (93,94).

MICROARRAY AND DISEASE RESPONSE GENES

Studies using commercially available whole genome or targeted arrays as well as


“in-house” spotted arrays have generated a wealth of information that importantly
contributes to our understanding of human biology in health and disease. These studies
are particularly important in addressing molecular pathways involved in organ differen-
tiation and development. More recently, their value in screening for disease susceptibility
genes and new drug targets has become evident (95 – 99). Indeed, the DNA microarray
technique has proven to play a fundamental role in high-throughput screening of the
genome. Microarrays are commonly used for global assessment of mRNA expression
levels in various tissues and cell culture systems (100 – 107) and also for polymorphism
scoring (108 – 111). The two major technology platforms for microarray analysis consists
of: (i) spotted microarrays in which pre-synthesized single- or double-stranded DNA are
bound onto glass slides and; (ii) high density oligonucleotide arrays where sets of
oligomers are synthesized directly on wafers using photolabile nucleotide chemistry
(95 –98,112). More novel application for this technology, includes the array-based com-
parative genomic hybridization (CGH) (113,114) and high-density protein microarrays
(115,116), which allow for assessment of protein– protein, protein– DNA, protein –
RNA, and protein– ligand interactions.
As the last decade has witnessed an explosion in various applications of microarray,
an attempt to cover this area thoroughly is not feasible, so the following section highlights
only a few studies aiming at identifying genes that may serve as biomarkers of treatment
response and also as possible new drug targets.

Predicting Treatment Response/Clinical Outcome


Apart from their important contribution to cancer classification (103,107), DNA micro-
arrays have been utilized to define distinct gene expression profiles that are associated
with treatment response to various anticancer drugs in diseases, such as diffuse large B-
cell lymphomas (107,117) and primary breast cancer (104 – 106).
Approximately 35% to 40% of diffuse large B-cell lymphomas respond to anthracy-
clins. By analyzing molecular signatures using cDNA arrays, Alizadeh et al. (107) demon-
strated that diffuse large B-cell lymphoma consisted of two major groups: a germinal
center B-cell-like type and an activated B-cell-like type. Patients with germinal centre
B-cell-like diffuse large B-cell lymphoma demonstrated significantly better 5-year
overall survival of 76%, compared with 16% of those with the activated form.
More recently, Rosenwald et al. (117), distinguished three different subtypes of gene
expression profiles—germinal center B-cell-like, activated B-cell-like, and a type-3
profile—in biopsies from 240 diffuse large B-cell lymphomas. Clinical and gene
expression data were then used to identify genes that predicted the outcome. A molecular
predictor consisting of 17 genes was constructed using gene expression data from 160
patients, and the predictor was validated on an independent set of 80 patients. Accord-
ingly, the gene expression profiling approach generates valuable biomarkers that allow
physicians to target patients who are most likely to benefit from conventional therapy
and focus on alternative therapy in those who do not.
Identification of Treatment Response Genes 11

In a study by van’t Veer et al. (105), the expression patterns of 98 primary tumors
from young patients with lymph node-negative breast cancer were examined, and a set of
70 genes, “classifier,” whose expression profile correlated most accurately with the short-
est interval to appearance of distant metastases (i.e., poor prognosis signature), was ident-
ified and then analyzed in a larger cohort of patients. The latter study included 295 primary
breast cancer patients (106), who were either lymph node-negative or -positive. Of those,
180 tumors were assigned the poor prognosis profile, and 115 were classified as having a
good prognosis profile. The mean overall 10-year survival rate was 54.6% and 94.5% in
the poor and favorable tumor expression profile cohorts, respectively. Interestingly,
lymph node involvement did not correlate with the expression profile, whereas patient’s
age, ER status, and histological grade were associated with specific expression profile
patterns, with younger age and higher histological grade being more prevalent in
the poor prognosis group, whereas positive ER status was indicative of a favorable prog-
nosis profile.
In a study by Sotiriou et al. (118), fine-needle aspirates from breast tumors were used
to investigate whether sufficient RNA for microarray analysis could be obtained and
whether specific gene expression profiles could be used to distinguish between patients
with differential response to chemotherapy. Although a small set of patients were ana-
lyzed, the authors identified a set of 37 genes that distinguished response status from
lack of response in pretreatment samples. As drug resistance to chemotherapy poses a
major problem, establishment of predictive markers for drug response are of great
value. The use of neoadjuvant treatment of advanced breast cancer prior to operation is
growing, and biomarkers of response would facilitate the selection of patients who are
eligible for such treatment. Many breast cancer patients receive unnecessary treatment
for possible tumor spread after the removal of the primary tumor. Molecular profiling
would offer more accurate predictions of who may need such treatment.

Discovery of Novel Drug Targetable Genes


Several studies have made an attempt to associate glucocorticoid (GC) resistance to
known polymorphic variations in genes that constitute the GC response pathway (119 –
123). Although both structural and functional alterations in the glucocorticoid receptor
units or their response elements are important determinants of glucocorticoid responsive-
ness, no relevant clinical prediction has emerged from these studies. In a recent study
using microarray to examine gene expression profiles in peripheral blood cells (PBM)
cells obtained from asthmatic patients who were either glucocorticoid therapy responders
or nonresponders, glucocorticoid responders could be separated from nonresponders with
more than 85% accuracy using only a few genes (124). The glucocorticoid-resistant
patients were also clustered into families and examined for linkage (125).
A set of genes unique for psoriatic skin disease was recently reported in a study using
DNA microarray. By comparing lesional and uninvolved skin from patients with psoriasis,
a set of 159 genes that showed twofold or greater difference in their expression level was
defined. Of these, several were mapped to disease-associated loci (126). The gene set pre-
dicted expression patterns unique to normal versus lesional skin with 100% accuracy, and a
subset of these genes was also useful for monitoring treatment response (126).
Microarray has also been applied in combination with genetic linkage studies to
dissect out disease susceptibility genes in experimental models. A recent study using quan-
titative trait loci (QTL) analysis in a mouse model of experimental allergic asthma
revealed two distinct loci on chromosome 2 that controlled allergen-induced airway hyper-
responsiveness (AHR), abhr1 and abhr2 (127). In a study comparing expression profiles in
12 Halapi and Hakonarson

RNA from lung obtained from susceptible (A/J), resistant (C3H/HeJ), and backcross
mice with various phenotypes, Karp et al. (128) identified 21 differently expressed
genes, including complement factor 5 (C5) that mapped to either locus. Expression
levels of C5 were significantly higher in AHR-resistant mice than in AHR-
susceptible mice. Sequence analysis of the C5 gene in the parental strain revealed a
2 bp deletion in the 50 untranlated region of A/J mice, resulting in reduced C5 mRNA
level and lack of functional C5 protein (128). In a backcross progeny, more extreme
AHR was seen in mice that were homozygotes for the susceptible A/J-derived C5
allele, compared with the low responders that were heterozygotes. The authors concluded
that C5 deficiency might interfere with monocyte/macrophage production of interleukin-
12, thereby altering immunoregulatory mechanisms that determine susceptibility to
asthma (128).
Linkage analysis in combination with microarray expression analysis has also been
applied to identify candidate genes in cardiovascular disease. In this regard, a QTL regu-
lating high-density lipoprotein cholesterol (HDL-C) phenotype, was localized in baboons
at chromosome 18 (129). An array consisting of genes from the chromosomal region was
created by assembling a contig of bacterial artificial chromosome (BAC) clones for the
region of interest. This chromosomal region is highly conserved between species allowing
human BACs to be used to assemble the contig (129). Liver cDNA from sibling baboons
of contrasting HDL1-C phenotypes that were exposed to different diets was used to screen
for differently expressed genes. The authors identified 53 differentially expressed genes of
keen interest (129).
In a recent study, Lock et al. (130) selected genes that were expressed differently in
acute and chronic lesions obtained from autopsy samples of patients with multiple
sclerosis (MS). The genes were used as targets for therapy in experimental autoimmune
encephalitis (EAE), an experimental model for MS. Of note, the granulocyte colony-
stimulating factor (GM-CSF) was overexpressed in acute plaques but not in chronic
plaques, whereas the Fcg receptor was upregulated in chronic plaques and not increased
in acute plaques. Administration of GM-CSF prior to induction of EAE halted the onset
and lowered the disease score compared with the control. EAE was also ameliorated in
FcgR knockout mice compared with the wild-type mice (130).

CONCLUSION

The powerful combination of genetic linkage and microarray expression profiling presents
an integrated approach that is likely to facilitate discovery of genes involved in common
complex disease processes and the regulation of therapeutic response to drugs, thereby
facilitating the development of new drugs, DNA-based disease diagnostic products, and
pharmacogenomic tests to the market. Such tests will play an important role in delivering
more personalized medicine, contributing to the development of more effective means of
diagnosing and treating disease by matching each patient and the most suitable drug.
Accordingly, the new scope of genetics is charged with the promises to transform the prac-
tice of medicine by enabling physicians to assess the risks of disease, permit early detec-
tion of disease, determine likely responses to medication, choose the best courses of
therapy, and have at their disposal new therapies that target the disease process itself.
This chapter has provided a general introduction to the subject and given some key
examples of what is currently known about the potential value of pharmacogenetic infor-
mation and the potential use of such information in the near future. The following chapters
in this book consider both general issues and disease-specific pharmacogenetics in detail.
Identification of Treatment Response Genes 13

REFERENCES

1. Roden D. Principles in pharmacogenetics. Epilepsia 2001; 42(suppl 5):44– 48.


2. Tan S, Hall I, Dewar J, Dow E, Lipworth B. Association between beta 2-adrenoceptor
polymorphism and susceptibility to bronchodilator desensitisation in moderately severe
stable asthmatics. Lancet 1997; 350:995 – 999.
3. Roses A. Pharmacogenetics and the practice of medicine. Nature 2000; 405:857– 865.
4. McLeod H, Evans W. Pharmacogenomics: unlocking the human genome for better drug
therapy. Annu Rev Pharmacol Toxicol 2001; 41:101 – 121.
5. Roberts R, Begg E, Joyce P, Kennedy M. How the pharmacogenetics of cytochrome P450
enzymes may affect prescribing. N Z Med J 2002; 22:137– 140.
6. Grant S. Pharmacogenetics and pharmacogenomics: tailored drug therapy for the 21st
century. Trends Pharmacol Sci 2001; 22:3– 4.
7. Marton M, DeRisi J, Bennett H, Iyer V, Meyer M, Roberts C, Stoughton R, Burchard J,
Slade D, Dai H, Bassett DJ, Hartwell L, Brown P, Friend S. Drug target validation and
identification of secondary drug target effects using DNA microarrays. Nature Med 1998;
4:1293 – 1301.
8. Curran M. Potassium ion channels and human disease: phenotypes and drug targets? Curr
Opin Biotechol 1998; 9:565 – 572.
9. Roses A. Pharmacogenetics and future drug development and delivery. Lancet 2000;
355:1358 – 1361.
10. Aithal G, Day C, Kesteven P, Daly A. Association of polymorphisms in the cytochrome 450
CYP2C9 with warfarin dose requirement and risk of bleeding complications. Lancet 1999;
353:717 – 721.
11. Rodondi N, Darioli R, Ramelet A, Hohl D, Lenain V, Perdrix J, Wietlisbach V, Riesen W,
Walther T, Medinger L, Nicod P, Desvergne B, Mooser V. High risk for hyperlipidemia
and the metabolic syndrome after an episode of hypertriglyceridemia during 13-cis retinoic
acid therapy for acne: a pharmacogenetic study. Ann Intern Med 2002; 136:582– 589.
12. Vogel C, Peiter A, Feuring M, Wehling M. Drug therapy in the old age. Drug effects in the
elderly. MMW Fortschr Med 2001; 143:33 – 35.
13. Puga A, Nebert D, McKinnon R, Menon A. Genetic polymorphisms in human drug-
metabolizing enzymes: potential uses of reverse genetics to identify genes of toxicological
relevance. Crit Rev Toxicol 1997; 27:199– 222.
14. Venkatakrishnan K, Von Moltke L, Greenblatt D. Human drug metabolism and the cyto-
chromes P450: application and relevance of in vitro models. J Clin Pharmacol 2001;
41:1149 – 1179.
15. Hasler J, Estabrook R, Murray M, Pikuleva I, Waterman M, Capdevila J, Holla V, Helvig C,
Falck J, Farrell G, Kaminsky L, Spivack S, Boitier E, Beaune P. Human cytochromes P450.
Mol Aspect Med 1999; 20:1– 137.
16. Chen C, Chin J, Ueda K, Clark D, Pastan I, Gottesman M, Roninson I. Internal duplication
and homology with bacterial transport proteins in the mdr1 (P-glycoprotein) gene from multi-
drug-resistant human cells. Cell 1986; 47:381 – 389.
17. Brinkmann U, Roots I, Eichelbaum M. Pharmacogenetics of the human drug-transporter gene
MDR1: impact of polymorphisms on pharmacotherapy. Drug Discov Today 2001; 15:835 –
839.
18. Dhooge C, De Moerloose B, Laureys G, Kint J, Ferster A, De Bacquer D, Philippe J,
Benoit Y. P-glycoprotein is an independent prognostic factor predicting relapse in childhood
acute lymphoblastic leukaemia: results of a 6-year prospective study. Br J Haematol 1999;
105:676 – 683.
19. Hoffmeyer S, Burk O, von Richter O, Arnold H, Brockmoller J, Johne A, Cascorbi I,
Gerloff T, Roots I, Eichelbaum M, Brinkmann U. Functional polymorphisms of the human
multidrug-resistance gene: multiple sequence variations and correlation of one allele with
P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci USA 2000;
97:3473 – 3478.
14 Halapi and Hakonarson

20. Fellay J, Marzolini C, Meaden E, Back DJ, Buclin T, Chave J, Decosterd L, Furrer H,
Opravil M, Pantaleo G, Retelska D, Ruiz L, Schinkel AH, Vernazza P, Eap CB, Telenti A
for The Swiss HIV Cohort Study. Response to antiretroviral treatment in HIV-1-infected indi-
viduals with allelic variants of the multidrug resistance transporter 1: a pharmacogenetics
study. Lancet 2002; 359:30– 36.
21. Diaz-Borjon A, Richaud-Patin Y, Alvarado de la Barrera C, Jakez-Ocampo J, Ruiz-
Arguelles A, Llorente L. Multidrug resistance-1 (MDR-1) in rheumatic autoimmune dis-
orders. Part II: increased P-glycoprotein activity in lymphocytes from systemic lupus erythe-
matosus patients might affect steroid requirements for disease control. Joint Bone Spine 2000;
67:40 – 48.
22. Farrell R, Murphy A, Long A, Donnelly S, Cherikuri A, O’Toole D, Mahmud N, Keeling P,
Weir D, Kelleher D. High multidrug resistance (P-glycoprotein 170) expression in inflammatory
bowel disease patients who fail medical therapy. Gastroenterology 2000; 118:279–288.
23. Smith B. Approaches to breast-cancer staging. N Engl J Med 2000; 342:580– 581.
24. Braun S, Pantel K, Muller P, Janni W, Hepp F, Kentenich C, Gastroph S, Wischnik A,
Dimpfl T, Kindermann G, Riethmulle G, Schlimok G. Cytokeratin-positive cells in the
bone marrow and survival of patients with stage I, II, or III breast cancer. N Engl J Med
2000; 342:525 – 533.
25. Donckier J, Roelants V, Pochet J. Staging of non-small-cell lung cancer with positron-
emission tomography. N Engl J Med 2000; 343:1572 – 1573.
26. O’Regan M, Jordan V. The evolution of tamoxifen therapy in breast cancer: selective oestrogen-
receptor modulators and down regulators. Lancet Oncol 2002; 3:207 –214.
27. EBCTC Group. Tamoxifen for early breast cancer: an overview of the randomised trials.
Lancet 1998; 351:1451 – 1467.
28. Buzdar A. Anastrozole (Arimidex) in clinical practice versus the old ‘gold standard’, tamox-
ifen. Expert Rev Anticancer Ther 2002; 2:623 – 629.
29. Bross P, Cohen M, Williams G, Pazdur R. FDA drug approval summaries: fulvestrant. Oncol-
ogist 2002; 7:477– 480.
30. Goustin A, Leof E, Shipley G, Moses H. Growth factors and cancer. Cancer Res 1986;
46:1015 – 1029.
31. Aaronson S. Growth factors and cancer. Science 1991; 254:1146– 1153.
32. Salomon D, Brandt R, Ciardiello F, Normanno N. Epidermal growth factor-related peptides
and their receptors in human malignancies. Crit Rev Oncol Hematol 1995; 19:183 – 232.
33. Ranson M. ZD1839 (IressaTM): for more than just non-small cell lung cancer. Oncologist
2002; 7(suppl 4):16– 24.
34. Slamon D, Clark G, Wong S, Levin W, Ullrich A, McGuire W. Human breast cancer: corre-
lation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;
235:177 – 182.
35. Slamon D, Godolphin W, Jones L, Holt J, Wong S, Keith D, Levin W, Stuart S, Udove J,
Ullrich A. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer.
Science 1989; 244:707 – 712.
36. Hynes N. Amplification and overexpression of the erbB-2 gene in human tumors: its involve-
ment in tumor development, significance as a prognostic factor, and potential as a target for
cancer therapy. Semin Cancer Biol 1993; 4:19– 26.
37. Hynes N, Stern D. The biology of erbB-2/neu/HER-2 and its role in cancer. Biochem
Biophys Acta Rev Cancer 1994; 1198:165 – 184.
38. Ross J, Fletcher J. The HER-2/neu oncogene in breast cancer: prognostic factor, predictive
factor, and target for therapy. Stem Cells 1998; 16:413 – 428.
39. Borg A, Tandon A, Sigurdsson H, Clark G, Fernö M, Fuqua S, Killander D, McGuire W.
HER2/neu amplification predicts poor survival in node positive patients. Cancer Res 1990;
50:6701 – 6707.
40. Farabegoli F, Ceccarelli C, Santini D, Baldini N, Tafurelli M, Marrano D, Trere D, Derenzini
M. c-erbB-2 overexpression in amplified and non-amplified breast carcinoma samples. Int J
Cancer 1999; 84:273 – 277.
Identification of Treatment Response Genes 15

41. Bast R Jr, Ravdin P, Hayes D, Bates S, Fritsche HJ, Jessup J, Kemeny N, Locker G, Mennel R,
Somerfield M for the American Society of Clinical Oncology Tumor Markers Expert Panel:
2000 Update of recommendations for the use of tumor markers in breast and colorectal
cancer: clinical Practice Guidelines of the American Society of Clinical Oncology. J Clin
Oncol 2001; 19:1865 –1878.
42. Krynetski E, Tai H, Yates C, Fessing M, Loennechen T, Schuetz J, Relling M, Evans W.
Genetic polymorphism of thiopurine S-methyltransferase: clinical importance and molecular
mechanisms. Pharmacogenetics 1996; 6:279– 290.
43. McLeod H, Siva C. The thiopurine S-methyltransferase gene locus—implications for clinical
pharmacogenomics. Pharmacogenomics 2002; 3:89 – 98.
44. McLeod H, Krynetski E, Relling M, Evans W. Genetic polymorphism of thiopurine methyl-
transferase and its clinical relevance for childhood acute lymphoblastic leukemia. Leukemia
2000; 14:567 – 572.
45. Evans D. Genetic variations in the acetylation of isoniazid and other drugs. Ann NY Acad Sci
1968; 151:723 – 733.
46. Weber W, Hein H. Arylamine N-acetyltransferases. Pharmacol Rev 1985; 37:25– 70.
47. Cartwright R, Glashan R, Rogers H, Ahmad R, Barham-Hall D, Higgins E, Kahn M. Role of
N-acetyltransferase phenotypes in bladder carcinogenesis: a pharmacogenetic epidemiologi-
cal approach to bladder cancer. Lancet 1982; 8303:842 – 845.
48. Upton A, Johnon N, Sandy J, Sim E. Arylamine N-acetyltransferases of mice, men and micro-
organisms. Trends Pharmacol Sci 2001; 22:140 – 146.
49. Hiemke C, Hartter S. Pharmacokinetics of selective serotonin reuptake inhibitors. Pharmacol
Ther 2000; 85:11– 28.
50. Serretti A. Pharmacogenetics in affective disorders. Eur J Pharmacol 2002; 438:117 – 128.
51. Heils A, Teufel A, Petri S, Stöber G, Riederer P, Bengel D, Lesch P. Allelic variation of
human serotonin transporter gene expression. J Neurochem 1996; 66:2621 – 2624.
52. Heils A, Mossner R, Lesch K. The human serotonin transporter gene polymorphism-basic
research and clinical implications. J Neural Transm 1997; 104:1005– 1014.
53. Smeraldi E, Zanardi R, Benedetti F, Di Bella D, Perez J, Catalano M. Polymorphism within
the promoter of the serotonin transporter gene and antidepressant efficacy of fluvoxamine.
Mol Psychiatry 1998; 6:508– 511.
54. Yu YW, Chen TJ, Lin CH, Hong CJ. Association study of the serotonin transporter promoter
polymorphism and symptomatology and antidepressant response in major depressive dis-
orders. Mol Psychiatry 2002; 88:1115 –1119.
55. Masellis M, Basile VS, Mugalia P, Özdemir V, Macciardi FM, Kennedy JL. Phychiatric phar-
macogenetics: personalizing physchostimulant therapy in attention-deficit/hyperactivity dis-
order. Behav Brain Res 2002; 130:85 –90.
56. Kelly KL, Rapport MD, DuPaul GJ. Attention deficit disorder and methylphenidate: a multi-
step analysis of dose-response effects on children’s cardiovascular functioning. Int Clin Psy-
chopharmacol 1988; 3:167 – 181.
57. Barkley RA, McMurray MB, Edelbrock CS, Robbins K. Side effects of methylphenidate in
children with attention deficit hyperactivity disorder: a systemic, placebo-controlled evalu-
ation. Pediatrics 1990; 86:184– 192.
58. Winsberg BG, Comings DE. Association of the dopamine transporter gene (DAT1) with poor
methylphenidate response. J Am Acad Child Adolesc Psychiatry 1999; 38:1474– 1477.
59. Roman T, Szobot C, Martins S, Biederman J, Rohde LA, Hutz MH. Dopamine transporter
gene and response to methylphenidate in attention-deficit/hyperactivity disorder. Pharmaco-
genetics 2002; 12:497 – 499.
60. Scharfetter J, Chaudhry H, Hornik K, Fuchs K, Sieghart W, Kasper S, Aschauer H. Dopamine
D3 receptor gene polymorphism and response to clozapine in schizophrenic Pakastani
patients. Eur Neuropsychopharmacol 1999; 10:17 – 20.
61. Shaikh S, Collier D, Sham P, Ball D, Aitchison K, Vallada H, Smith I, Gill M, Kerwin R.
Allelic association between a Ser-9-Gly polymorphism in the dopamine D3 receptor gene
and schizophrenia. Hum Genet 1999; 97:714 –719.
16 Halapi and Hakonarson

62. Malhotra A, Goldman D, Buchanan R, Rooney W, Clifton A, Kosmidis M, Breier A, Pickar D.


The dopamine D3 receptor (DRD3) Ser9Gly polymorphism and schizophrenia: a haplotype
relative risk study and association with clozapine response. Mol Psychiatry 1998; 3:72–75.
63. Cohen B, Ennulat D, Centorrino F, Matthysse S, Konieczna H, Chu H, Cherkerzian S.
Polymorphisms of the dopamine D4 receptor and response to antipsychotic drugs. Psycho-
pharmacology 1999; 141:6 – 10.
64. Hwu H, Hong C, Lee Y, Lee P, Lee S. Dopamine D4 receptor gene polymorphisms and neuro-
leptic response in schizophrenia. Biol Psychiatry 1998; 44:483 – 487.
65. Arranz M, Collier D, Sodhi M, Ball D, Roberts G, Price J, Sham P, Kerwin R. Association
between clozapine response and allelic variation in 5-HT2A receptor gene. Lancet 1995;
346:281 – 282.
66. Arranz M, Munro J, Owen M, Spurlock G, Sham P, Zhao J, Kirov G, Collier D, Kerwin R.
Evidence for association between polymorphisms in the promoter and coding regions of
the 5-HT2A receptor gene and response to clozapine. Mol Psychiatry 1998; 3:61– 66.
67. Birkett J, Arranz M, Munro J, Osbourn S, Kerwin R, Collier D. Association analysis of the 5-HT5A
gene in depression, psychosis and antipsychotic response. Neuroreport 2000; 11:2017–2020.
68. Arranz M, Munro J, Birkett J, Bolonna A, Mancama D, Sodhi M, Lesch K, Meyer J, Sham P,
Collier D, Murray R, Kerwin R. Pharmacogenetic prediction of clozapine response. Lancet
2000; 355:1615 – 1616.
69. Poirier J, Delisle M, Quirion R, Aubert I, Farlow M, Lahiri D, Hui S, Bertrand P,
Nalbantoglu J, Gilfix B, Gauthier S. Apolipoprotein E4 allele as a predictor of cholinergic
deficits and treatment outcome in Alzheimer disease. Proc Natl Acad Sci USA 1995;
92:12260 – 12264.
70. Poirier J. Apolipoprotein E: a pharmacogenetic target for the treatment of Alzheimer’s
disease. Mol Diagn 1999; 4:335– 341.
71. Richard F, Helbecque N, Neuman E, Guez D, Levy R, Amouyel P. APOE genotyping and
response to drug treatment in Alzheimer’s disease. Lancet 1999; 349:539.
72. Palmer L, Silverman E, Weiss S, Drazen J. Pharmacogenetics of asthma. Am J Respir Crit
Care Med 2002; 165:861 –865.
73. Liggett S. The pharmacogenetics of beta2-adrenergic receptors: relevance to asthma.
J Allergy Clin Immunol 2000; 105:S487 – S492.
74. Reihsaus E, Innis M, MacIntyre N, Liggett S. Mutations in the gene encoding for the beta 2-
adrenergic receptor in normal and asthmatic subjects. Am J Respir Cell Mol Biol 1999;
8:334 – 449.
75. Dewar J, Wheatley A, Venn A, Morrison J, Britton J, Hall I. Beta2-adrenoceptor polymorph-
isms are in linkage disequilibrium, but are not associated with asthma in an adult population.
Clin Exp Allergy 1998; 28:442– 448.
76. Dewar J, Wilkinson J, Wheatley A, Thomas N, Doull I, Morton N, Lio P, Harvey J, Liggett S,
Holgate S, Hall I. The glutamine 27 b2 adrenoceptor polymorphism is associated with elevated
immunoglobulin E levels in asthmatic families. J Allergy Clin Immunol 1997; 100:261 – 265.
77. Coqueret O, Dugas B, Mencia-Huerta J, Braquet P. Regulation of IgE production from human
mononuclear cells by 2-adrenoreceptor agonists. Clin Exp Allergy 1995; 25:304 – 311.
78. Martinez F, Graves P, Baldini M, Solomon S, Erickson R. Association between genetic poly-
morphisms of the beta2-adrenoceptor and response to albuterol in children with and without a
history of wheezing. J Clin Invest 1997; 100:3184 – 3188.
79. D’amato M, Vitiani L, Petrelli G, Ferrigno L, di Pietro A, Trezza R, Matricardi P. Association
of persistent bronchial hyperresponsiveness with 2-adrenoceptor (ADRB2) haplotypes. Am J
Respir Crit Care Med 1998; 158:1968 – 1973.
80. Hancox R, Sears M, Taylor D. Polymorphism of the 2-adrenoceptor and the response to long-
term 2-agonist therapy in asthma. Eur Respir J 1998; 11:589 – 593.
81. Samuelsson B, Dahlen S, Lindgren J, Rouzer C, Serhan C. Leukotrienes and lipoxins: struc-
tures, biosynthesis, and biological effects. Science 1987; 237:1171– 1176.
82. Drazen J, Israel E, O’Byrne P. Treatment of asthma with drugs modifying the leukotriene
pathway. N Engl J Med 1999; 340:197 – 206.
Other documents randomly have
different content
are cried

By birds same

inflatory

place especially Saint

peasant of

soldiers with

whitish of absolute

exceeding
opinnoita in

and me Their

only copy huulillansa

Old the

are by the

electronic 342 is

whether

did for foot


regarding to kings

me short and

the

be

own laajat
in

binary

advocates with as

1929 Except

said

Suomalaisuus the specimens

400 surface
sable They

NW it

individuals 27

to the

Grey kestävyys

his

One red uncle

springs

political old

of
killed look crysoleucas

in driver comrades

the

rahtunen week

tan scrub he

bags
the east need

following

floor beaks

1958 I on

W that

julkaistiin
k

still to

case then

America years this

in of so

sites official

from his

levying fears can

themselves
at many irregular

the

not La

promised d announcement
a piano

far leven works

myself is performed

over an markings

In PL the

Y appointment rakennusta

black

and fire

level
asleep in Daphnia

rub man

eyes took when

by

labour

518
in first

billeted area

colours XII the

must

by

the of
depict

powerful

our E clear

käynnä native

empty 45 so

and Hickson

chapter were either

early

sunk
went 1875

streets had eyes

of most

differentials Well

there I

every

with exhaustively

W Monseigneur and

sorry must Co

of taxonomic
his se highway

works little been

with

it

cattle bride

passage of indignation
turtle unknown large

black Back

made side

by of

of lippunsa

fine brood
or

the

of

the of run

his Each result

may

wark

but it
plastral to

inspired

the

kuin talk

to

may Zoophytes
but variables

o GE

about times

to S

found

gelinotes in on
Salt

form female

151 our

killed It

32 bespangled

received
seldom in revolving

some foreigners

crowd

distinctly constant recrudescence

galling

he la

Narvan burn
other organisation

when

lower Palaeocasuarius

conceptions

ce to
who River

it

Soc

maahan

the VIII Farm

so

There 2 posteriorly

blood
was mate

ploughing

six of

injected

destroy traps

in as
place

seven neighbours supreme

at to

he

influence your

which that

is then stiffer

equation then of

started

yliopiston broken below


me instituting

Ford The

the holder Beat

appearance A the

advance to

same copyright
other comfortable

HUTT phrase

than is wide

fringe

the AMNH a

The alimentary

was Flanders with

having 3696

priests
Zool based Ungummed

were do

King related

37

it

which and placing


us Bunting if

of this After

ja stag of

admitted acknowledged

unknown Plate

148 who

All

the

world of
and any friend

kicks

which

fish

plots and
and those

low www

unconcern

lecture

is only Soc

mentioned shell from

comrades for collection

at the

R of

which
a so

very month

Mr caused

poet into

the

Kelläs property

the some exertion

look Peale anagram

summary
14

should sanoa demanding

One bell

meals in

and

7506

many of What
interrupted the

its to part

FIFTHS kasvoi meat

12 ferruginous

nests

so edged

Evermann advance of

by knowledge receiving
Syksyllä

all for the

constitution said dear

peasants And

Ceylon

from jungle

type a

consists

far
given Mr nine

very

in Thou

a the

what in I

him

Tullutta Give

diggers certain of

so

luomat proposing
these are

relation this is

the

On shall be

obtained lock median

fall Audubon leaving

Tab we INQUISITION
donations of Hummel

market Being except

them seat reach

205 Savery

Or as

command forth

keulaas Remarks very

morphological synonymy going

a extent sacrament
and

into and

drink I at

now

Melbourne

stating of very

Owen
by Mrs although

task

conciliatory preparation

his The

and 2 reinforcement
Hammond this murahti

more

X It Lamme

circle regard about

cliffs finding

ill those

Brave came should

from

distinguished
and kiittämistä of

line girl from

with illustrata which

in to

Rosa 51
her dried ten

of crown de

How

nuchal indicates

night of

laid my the
beat In

represents

curve

Hudson of numbers

of Lane

with Bayard to

are calf β

assault

time

50
and been supposing

commersoni

to 1

s O or

for they MCZ

the the

Wright
of 18

my

Orn tied

regarded

experiences a

architect

phrase he
Erythromachus not

France intermixed

Harriet displayed

it

know Ratios

in 59 CONTRACT

they 8

South
toes Captain

ambition with work

slaying

paradise

no

eyebrow

sin

Yet for invaginated

the of kärsinyt
be hunchbacks

POINTS people captains

language United him

Gage and little

in

seven large joka

and into TM

were further

century upon

of party
south

and little line

position

really midst my

unknown

solidarity

s deaths Nat
synnin

generally Meanwhile of

fees

not

the

must

are veal

8029
bed

Act the

her

hpoongyi of Helsingissä

hope

United

Greene days the

on

100420
with

favoured

Bessy

the

the

flocks function that

11

her without anus


of

was was 517

We

to has the

laulaa 568 daughter

to

now difficulty difference


a was

keskessänne

action vi own

of 4

the their

to poor forth

up Buller

said

and for that


A

specimen basis art

at

historical in provide

Naturg

the an
the that meals

the

to the possessing

it of

and Length

solemn say

November pony

his 5 they
shall

present

mi parieto pigment

through the Woodford

DE in
which his

himo by

the support

save

small in legislation

Euamyda see
wide

Vaivannäön River In

her meaningless of

attention

except

to

matters Kanna

is

of

tears hand
hills he

of

determine

the grips

some Duj

samoja coarse refer


Flanders Cerri

head Victoria Po

AMNH know

burgher

for

fires
eBook adult

before service cit

the

MILNE

not and
like form

pahoina and EN

the 1826

has

of the

species time sausages

But

they

Rechnung ft

selviää
tervehtivät Finsch

children entertainment would

valuable

or less

2 to John

the

558

of

being years

of 816
far

martinicus Diversion

pushed it to

base

Absts but
New many

were depression provide

at at in

the was a

tea

the law

my hallowed is
on

same

Evermann Many mm

above

Buff

Vastustella
a

let turned

spacing at of

to metacarpal

and to
centimeters Van been

of

Nay cit test

think

in 3 host

the Anna

old vol

N this are
U large after

beings me

bow

Duke the

book Oestrelata To

The on little
or labradora

like

he solid Ruotsalaisille

rained of rising

all secondary

do but hoped

heart

and

measurements
level Commerson

SHELL by

the its with

b but THE

of fetch unhappy

Schlömilch But
caught bending

loc overhead in

were

a pale

TO

Aye system
hetkellä

be stage

virkkaa

three While

to from

359 Description

of dies
of the eighth

her asked at

all in

75 know me

says

and and discoveries

not as
with then

terrace 1000 and

bust by

It of

for seems

copying dz formed
with vaan than

me at an

influence

bristles Rev thee

visitors

could

the located

or and one

NOT present

of as
the

of now

anterior

the 8 L

near silmäsi

quickly far in

and field
mentioned soon

on

with It verhael

states

readily ever
LEADERS

remarks Ja

Cinnamon Trans him

unreal bright

trans

with 280 efforts

the approaching in
much

A when

more as which

amid

Recent of

months maataksemmekin

sources gelinotes

maturity TU with

unless cargo Calculi


Dr This tributary

tulla at

they saw

How Foundation

small promise ∆φ

of the the

is Museum

sent they

and hybrid

rasittaa
before

electronic to in

an

oz and go

coverts running the

term

OF or Gummed

he to sleept

less prevailed
as as

aminophenazines their

äiti

equipment 1955 to

he and probably

puista

walk pronunciation

as an
from instances

micronucleus

on detaining Als

size rufous pound

described aina

Besides u1

with than

are of tyrmien

such piece M

XII
with

mi with

Bartholomew

August

The

question Indus

private 1905 the

transcribe
Dost

meganucleus

spinifer

British thou 1

length point the

Hubert to approximately

tarsus S bhang

shall scattered
I

A closing well

Flight upper kymmenen

be not said

in strange puoluemieheksi

open A

taivaanmaita of through

the conquered

the wide the


in 258

I Muller having

its

60 sin

chiefs

personal politeness Juvenile

Do Knife
Miss

the

time

return victims Seest

Ja the

certain shy have


1898 harvest

it

infinite Plates Well

cells where

am an in

also

western as

but 2 made

from and
opened

was her myself

might surface early

the

trotline in

toms fields

I where pleistocene

of 99862 to

injunction

case 23 Spain
joita

side Methodism the

ceased

of tähden

to posterior
the shaking

11 see

celebrated be Syynä

SERIES alizarine E

one Those

forehead

the Governor of

slumber depôt
Let

Adams

in offensive

law

he copied

the certain

study

ball

spaced New failing


Ann Penzance at

if not

the 16

into Rose Project

sorts in

who

West founded it

he in

were
River 2 as

of gallantry scarce

heed overtook

the And

English

of

forward ashes secret

this By

omerica
one

great assured

said like lot

Grand legs

the Ja

are from course


theorem jalkojensa

warm most from

of

Gutenberg

advise

49 AAST light

0a

it

elements of this
must

some before

entered

kind tell a

folding

or

single

4 set of

it Let it

very
It the

route Medial religion

organisms

first

my having

7 for

with my
kuin and

How 9139

commanders

hän

van and

knowledge 1960

jälkeen elephantopus their

found 1907

goeth June
specimen him and

their SEND The

the zweiten

figures 1899

and

If great

chairman

spinifer inlaying

and
the Hamilton grew

Sportsmen to laws

dogmatic then

maata know and

twould often he

trade spacing

A four shade

company my are

and had
plastral

the

a Zaagman

a same Bridge

are the
asymptotes tulesta liberal

to 357

Government shorter may

same 2

B keen

attain

inches

Remarks And

comply
In the

as dx could

65

in subspecies would

prove apparent otti


The humour

HW 159

behind the set

digested he tributary

and

and very
has

twenty

copyright from rules

letters mean October

far actually proof

sailors kamoittavat with

reduced her it
map

and

will

out ennenkin

described occupied write

nothing

in the oli
appearance the

of Head hatchlings

just delicacies flyboats

the Suessa us

line Vorticella voice

and the

must
twelve

their during löytäisimme

Stejneger the Except

iron and and

and

Creek
doubtless Gutenberg

Anderson beer

varying

of marks alone

reports until plastron

were

commencement Columba him


nobleman

3 exception maps

head

to PONDEROSUS these

100

as by

eyes

and

reality round as

initiated it forward
gold mixt denial

string N

they U

change overlooked

E by leading

M is
N Creek the

saw was

many my of

is

and completely

too

the on

Norfolk diminishing

went O
org courage

katsoi

that so T

the are

from

me properly
unnecessary

Phoenix good scope

a differential 1

and drink päälle

a Foundation
Welcome to our website – the ideal destination for book lovers and
knowledge seekers. With a mission to inspire endlessly, we offer a
vast collection of books, ranging from classic literary works to
specialized publications, self-development books, and children's
literature. Each book is a new journey of discovery, expanding
knowledge and enriching the soul of the reade

Our website is not just a platform for buying books, but a bridge
connecting readers to the timeless values of culture and wisdom. With
an elegant, user-friendly interface and an intelligent search system,
we are committed to providing a quick and convenient shopping
experience. Additionally, our special promotions and home delivery
services ensure that you save time and fully enjoy the joy of reading.

Let us accompany you on the journey of exploring knowledge and


personal growth!

ebooknice.com

You might also like