Clinical Proteomics - download.e-bookshelf.de · Proteomics in Drug Research. 2006 ISBN-13:...

30
Clinical Proteomics From Diagnosis to Therapy Edited by Jennifer E. Van Eyk and Michael J. Dunn

Transcript of Clinical Proteomics - download.e-bookshelf.de · Proteomics in Drug Research. 2006 ISBN-13:...

  • Clinical Proteomics

    From Diagnosis to Therapy

    Edited by

    Jennifer E. Van Eyk and Michael J. Dunn

    InnodataFile Attachment9783527622160.jpg

  • Clinical Proteomics

    Edited by

    Jennifer E. Van Eyk and

    Michael J. Dunn

  • Related Titles

    Spirin, A. S., Swartz, J. R. (Eds.)

    Cell-free Protein Synthesis

    Methods and Protocols

    2007

    ISBN-13: 978-3-527-31649-6

    Omenn, G. S. (Ed.)

    Exploring the Human Plasma Proteome

    2006

    ISBN-13: 978-3-527-31757-8

    Jungblut, P. R., Hecker, M. (Eds.)

    Proteomics of Microbial Pathogens

    2006

    ISBN-13: 978-3-527-31759-2

    Hamacher, M., Marcus, K., Stühler, K., van Hall, A., Warscheid, B., Meyer, H. E. (Eds.)

    Proteomics in Drug Research

    2006

    ISBN-13: 978-3-527-31226-9

    Tsai, C. S.

    Biomacromolecules - Introduction to Structure, Function and

    Informatics

    2006

    ISBN-13: 978-0-470-08011-5

    Wink, M. (Ed.)

    An Introduction to Molecular Biotechnology

    Molecular Fundamentals, Methods and Applications in Modern Biotechnology

    2006

    ISBN-13: 978-3-527-31412-6

  • Clinical Proteomics

    From Diagnosis to Therapy

    Edited by

    Jennifer E. Van Eyk and Michael J. Dunn

  • The Editors

    Dr. Jennifer van Eyk

    Johns Hopkins University

    Bayview Medical Campus

    5200 Eastern Ave.

    Baltimore, MD 21224

    USA

    Prof. Dr. Michael J. Dunn

    Proteome Research Centre

    UCD Conway Institute

    Belfield

    Dublin 4

    Ireland

    Cover

    The front cover picture is based on a 2-D-DIGE

    comparing a cell lysate preparation and secretome

    preparations, both derived from SW620 cells. It was

    published by Schwarte-Waldhoff et al. in Proteomics

    Clin. Appl. 2007, 1, 47-61. Lysate proteins are shown

    in red and secretome proteins in green.

    Reproduced with kindly permission of the Medical

    Proteome-Center, University of Bochum, Germany.

    The background protein image has been reproduced

    with the kind permission of Professor Alfredo Ricci,

    Dpto. di Chimica Organica, Universita di Bologna,

    Italy.

    All books published by Wiley-VCH are carefully

    produced. Nevertheless, authors, editors, and

    publisher do not warrant the information contained

    in these books, including this book, to be free of

    errors. Readers are advised to keep in mind that

    statements, data, illustrations, procedural details or

    other items may inadvertently be inaccurate.

    Library of Congress Card No.:

    applied for

    British Library Cataloguing-in-Publication Data

    A catalogue record for this book is available from the

    British Library.

    Bibliographic information published by

    the Deutsche Nationalbibliothek

    Die Deutsche Nationalbibliothek lists this

    publication in the Deutsche Nationalbibliografie;

    detailed bibliographic data are available on the

    Internet at .

    # 2008 WILEY-VCH Verlag GmbH & Co. KGaA,Weinheim

    All rights reserved (including those of translation into

    other languages). No part of this book may be repro-

    duced in any form - by photoprinting, microfilm, or

    any other means - nor transmitted or translated into a

    machine language without written permission from

    the publishers. Registered names, trademarks, etc.

    used in this book, even when not specifically marked

    as such, are not to be considered unprotected by law.

    Composition Thomson Digital, Noida, India

    Printing betz-druck GmbH, Darmstadt

    Bookbinding Litges & Dopf GmbH, Heppenheim

    Cover Design Anne Kessler, Karlsruhe

    Printed in the Federal Republic of Germany

    Printed on acid-free paper

    ISBN 978-3-527-31637-3

  • Contents

    Editor’s Overview XIX

    List of Contributors XXI

    I Technologies 1

    1 Preanalytical Issues in Clinical Proteomic Studies 3Roisean E. Ferguson, Rosamonde E. Banks

    1.1 Introduction 31.2 Preanalytical Factors 31.2.1 Biological Variation 41.2.1.1 Intrinic Influences/Factors 41.2.1.2 Extrinsic Influences/Factors 51.2.2 Technical Variables 51.2.2.1 Specimen/Sample Collection Mode 51.2.2.2 Type of Sample Container 61.2.2.3 Sample Processing and Handling Conditions 71.2.2.4 Sample Storage 71.3 Summary and Concluding Remarks 8

    2 Protein Separation by Two-Dimensional Electrophoresis 13Pamela M. Donoghue, Miroslava Stastna, Michael J. Dunn

    2.1 Introduction 132.2 2DE: Protein Solubilization and Sample Preparation 142.3 2DE: Protein Separation 152.3.1 Focusing in the First Dimension 152.3.2 Advances in IEF 162.4 Improving Proteomic Coverage by Subcellular Fractionation 212.5 Protein Detection and Image Analysis 242.6 The Future of 2DE 25

    V

  • 3 Protein Separation: Liquid Chromatography 31Miroslava Stastna, Jennifer Van Eyk

    3.1 Introduction 313.2 Liquid Chromatography 323.2.1 HPLC Separation Principles 323.2.2 Reversed-Phase LC (RPLC, 1DLC) 343.2.3 Affinity Chromatography 373.2.4 Size-Exclusion Chromatography 383.2.5 Ion-Exchange Chromatography 393.2.6 Two-Dimensional LC 403.2.6.1 Chromatofocusing to Reversed Phase 403.2.6.2 Ion-Exchange Reversed-Phase Liquid Chromatography 413.2.7 Three-Dimensional Liquid Chromatography 413.2.8 LC Image Analysis Requirement 423.2.9 Mass Spectrometry for LC 423.2.9.1 MALDI-TOF MS 433.2.9.2 ESI-MS/MS 433.3 Conclusions 46

    4 HPLC in Protein Discovery 53Timothy J. Barder

    4.1 Introduction 534.2 LC-Based Approaches in Peptide Mass Mapping 544.3 LC-Based Approaches in Protein Mapping 544.4 Orthogonal 2D HPLC Separations 564.5 Conclusion 57

    5 IEF Analysis of Peptides for Biomarkers Discovery 61Ali R. Vaezzadeh, Catherine G. Zimmermann-Ivol, Jacques Deshusses,

    Alexander Scherl, Denis F. Hochstrasser

    5.1 Introduction 615.2 Background 625.2.1 Isoelectric Focusing 625.2.2 Shotgun Proteomics 625.2.3 Shotgun IEF 635.3 Shotgun IEF Workflow 645.4 Applications 665.5 Discussion and Outlook 66

    6 Capillary Electrophoretic Separations for Clinical Proteomics 73Brian M. Balgley, Weijie Wang, Xueping Fang, Don L. DeVoe, Cheng S. Lee

    6.1 Introduction 736.2 (Single-Dimension) Capillary Electophoretic Separation 746.3 Capillary Electrophoresis-Based Multidimensional Separations 746.3.1 Capillary Liquid Chromatography–Capillary Electrophoresis 75

    VI Contents

  • 6.3.2 Capillary Electrophoresis–Capillary Electrophoresis 756.3.3 Capillary Electrophoresis–Liquid Chromatography 766.3.3.1 Characterization of Human Saliva Proteome 776.3.3.2 Targeted Tissue Proteomics 796.4 Conclusions 85

    7 Quantitative Proteomics Using Nano-LC with High Accuracy

    Mass Spectrometry 89Ljiljana Paša-Tolić, Jon M. Jacobs, Wei-Jun Qian, Richard D. Smith

    7.1 Introduction 897.2 Fundamentals of a High Mass Accuracy-Based LC–MS

    Approach 907.3 Nano-LC–MS for Enhanced Sensitivity and Dynamic

    Range Coverage 927.4 Further Developments for Increasing Proteomic Throughput 947.5 Obtaining More Robust Quantitative Proteomic Measurements 947.6 Summary and Perspective 96

    8 Antibody Microarrays for Protein and Glycan Detection 101Songming Chen, Brian B. Haab

    8.1 Introduction 1018.2 Antibody Preparation and Microarray Production 1028.3 Sandwich Assays with Fluorescence Detection 1048.4 Antibody Microarrays with Lectin Detection 1068.5 Conclusion 1068.6 Detailed Protocols 107

    9 Biomarker Identification: The Role of Experimental Design,

    Statistics, and Data Sharing 113Marc R.Wilkins

    9.1 Introduction 1139.2 Experimental Designs for Biomarker Discovery 1149.3 Identification of Biomarker Proteins 1169.4 Biomarker Validation and the Issue of Data Sharing 1189.5 Conclusions 119

    II Cancer 121

    10 Applications of Stable Isotope Tagging Based Quantitative

    Proteomics in Cancer Research 123Ru Chen, Teresa A. Brentnall, Ruedi Aebersold

    10.1 Introduction 12310.2 Stable Isotope Tagging Methods 12410.2.1 Chemical Labeling of Stable Isotope Tags 125

    Contents VII

  • 10.2.2 Biological Incorporation of Stable Isotope Tags 12710.3 Applications in Studies of Tissue Samples 12810.3.1 Whole Tumor Tissue Labeled with ICAT 12810.3.2 Whole Tumor Tissue Labeled with ICAT and iTRAQ 13010.3.3 Isolated Tumor Cells Labeled with ICAT 13110.3.4 Isolated Tumor Cells Labeled with 16O/18O 13110.4 Applications in Studies of Bodily Fluids 13210.4.1 Pancreatic Juice Labeled with ICAT 13310.4.2 Nipple Aspirate Fluid Labeled with ICAT 13310.4.3 CSF 13410.5 Applications in Studies of Cell Lines 13510.5.1 Ovarian Cancer Cell Lines Labeled with ICAT 13510.5.2 Breast Cancer Cell Lines Labeled with 18O Labeling 13510.5.3 Prostate Cancer Cell Lines Labeled with SILAC 13610.5.4 Secretome by Pancreatic Cancer Cell Line Labeled with SILAC 13610.6 Applications in Studies of Protein Interaction 13710.7 Applications in Studies of Posttranslational Modifications (PTM) 13810.8 Summary 139

    11 Two-Dimensional Liquid Separations, Protein Microarrays,

    and Mass Spectrometry in Comprehensive Analysis of

    Posttranslational Modifications and Biomarker

    Discovery in Cancers 145Tasneem H. Patwa, Jia Zhao, David E. Misek, David M. Lubman

    11.1 Challenges in Biomarker Discovery: The Emerging Role of

    Posttranslational Modifications 14511.2 Proteomics in Disease Research 14611.3 The Problem of Identifying and Characterizing Posttranslational

    Modifications: Current Efforts 14711.4 Microarrays in Proteomic Investigations 14811.5 A Comprehensive Method Combining Liquid Separations,

    Microarrays, and Mass Spectrometry 14811.6 2D Liquid-Based Separations and Mass Mapping 15011.7 Posttranslational Modification (PTM) Analysis 15211.8 Phosphorylation 15311.9 Glycosylation 15411.10 Autoimmune (Humoral) Response Studies 15611.11 Future of a 2DLC, Microarray Methodologies in Discovery and

    Diagnostics 158

    12 Development and Use of Reversed-Phase Protein Microarrays

    for Clinical Applications 165Virginia Espina, Julia Wulfkuhle, Valerie S. Calvert,

    Kirsten H. Edmiston, Lance A. Liotta, Emanuel F. Petricoin III

    12.1 Introduction 165

    VIII Contents

  • 12.2 The Growing Role of Protein Arrays in Molecular Diagnostics 16712.3 Reversed-Phase Arrays: Enabling Technology for Patient-Tailored

    Therapeutics 16712.4 Use of Reversed-Phase Arrays for Signal Pathway Profiling

    of Human Cancer 16912.5 Use of Reversed-Phase Arrays: A View to the Future 171

    13 Cyclin-Dependent Kinase Inhibitors and Cancer: Usefulness

    of Proteomic Approaches in Assessment of the Molecular

    Mechanisms and Efficacy of Novel Therapeutics 177Marian Hajduch, Helena Skalnikova, Petr Halada, David Vydra,

    Petr Dzubak, Marta Dziechciarkova, Miroslav Strnad,

    Danuta Radioch, Suresh Jivan Gadher, Hana Kovarova

    13.1 Introduction 17713.2 Proteomic Analysis of Cancer Cells Responding to the

    Synthetic CDKI 18213.3 Two-Dimensional Protein Maps of Cancer Cells Treated by CDKI 18413.3.1 Model of Hematological Malignancy: CEM T-Lymphoblastic

    Leukemia 18413.3.2 Solid Tumor Model: A549 Lung Adenocarcinoma Cells 18713.4 Evaluation of the Protein Maps: Possible Pathways Relevant to

    Anticancer Effects of CDK Inhibition 18913.4.1 Candidate Biomarkers Identified Using the Hematological

    Malignancy Model 18913.4.2 Candidate Biomarkers Identified Using the Solid Tumor Model 19213.5 Biomarker Validation Studies Focused on the crkl Protein 19413.6 Conclusions 195

    III Cardiac Disease 203

    14 Diagnostic Markers for Monitoring Heart Transplant Rejection 205Ciara A. McManus, Marlene L. Rose, Michael J. Dunn

    14.1 Introduction 20514.2 Acute Rejection 20614.3 Chronic Rejection 20814.4 Cardiopulmonary Bypass 21214.5 Conclusions 213

    15 The Study of Microheterogeneity in Human Plasma Proteins:

    Application to Acute Myocardial Infarction 217Randall W. Nelson, Urban A. Kiernan, Dobrin Nedelkov,

    Kemmons A. Tubbs, Eric E. Niederkofler

    15.1 Background 21715.2 Technical Approach 218

    Contents IX

  • 15.3 Programmatic Study of Disease: Population Proteomics

    Versus Myocardial Infarction 21915.3.1 Preliminary Screening, (Putative) Biomarker Discovery and

    Identification 22215.3.2 Verification 22315.3.3 Knowledge Assembly and Next-Generation Assay Design 22315.3.4 Data Generation 22515.3.5 Data Analysis 22515.3.6 Blind and Randomized Challenge of Final Assay 22715.4 Summary 228

    16 Discovery of Biomarkers for Cardiovascular Diseases 231Anthony O. Gramolini, Andrew Emili

    16.1 Current Proteomic Technologies Available for CVD Biomarker

    Searches 23216.2 Challenging Issues for Proteomic Profiling 23316.3 Screening Blood for Biomarkers 23416.4 Tissue Surveys 23516.5 The Value of Animal Models 23616.6 After Technology Platform and Sample Selection:

    What Makes a Good Biomarker? 23716.7 Ongoing Considerations 23816.8 Outlook 238

    17 Development of Biomarker Development Pipeline:

    Search for Myocardial Ischemia Biomarkers 241Qin Fu, Shijun Sheng, Jennifer E. Van Eyk

    17.1 Introduction 24117.2 Myocardial Ischemia and Infarction 24217.3 Lessons Learned from Cardiac Troponin 24517.4 Building a Biomarker Development Platform I-Discovery 24617.4.1 High-Abundant Protein Partitioning 24817.4.2 Utilizing Multiple Protein Separation Methods to Maximize

    Proteome Coverage: A Synergistic Approach 25017.5 Validation 25417.5.1 Technologies in Validation 25617.5.2 Cohorts for the Validation 25617.6 Summary 257

    18 The Albuminome as a Tool for Biomarker Discovery 263Rebekah L. Gundry, Robert J. Cotter

    18.1 Protein–Protein Interactions and Protein-Centric

    Approaches in Proteomics 263

    X Contents

  • 18.2 Defining the Albuminome 26518.3 The Albuminome as a Tool in Biomarker Discovery 27018.4 Role of the Albuminome in Cardiovascular Proteomics 27518.5 Other Plasma Subproteomes 27618.6 Conclusion 277

    19 Application of Metabolomics to Cardiovascular

    Biomarker and Pathway Discovery 279Gregory D. Lewis, Robert E. Gerszten

    19.1 Introduction 27919.2 The Birth of Metabolomics 27919.3 Technologies to Define the Human Metabolome 28119.4 The Diagnostic Utility of Metabolic Peak Patterns:

    A Call for Unambiguous Identification 28219.5 Pathway Analysis of Metabolomic Data 28319.6 Rationale for Metabolomic Approaches to Study Atherosclerosis

    and its Complications: The Role of Proinflammatory Lipid

    Metabolites 28519.7 Unanticipated Roles of ‘‘Intracellular’’ Metabolites 28619.8 Clinical Rationale for Applying Metabolomics to Coronary

    Heart Disease 28619.9 Impediments to Human Applications 28719.10 Application of Metabolomics to Unique Human Cardiovascular

    Disease Models 28719.11 Conclusion 290

    IV Vascular Disease: Pulmonary, Diabetes and Brain 295

    20 Urinary Biomarkers in Diabetic Nephropathy and Other

    Glomerular Diseases 297John M. Arthur, T. Brian Powell

    20.1 Urine as a Source of Protein Biomarkers 30020.2 Size Selectivity of Urine Proteins as a Marker 30120.3 Charge Specificity of Urine Proteins as a Marker 30220.4 Markers in Diabetic Nephropathy 30320.5 Markers for Other Glomerular Diseases 30420.6 Urine Proteomics 30420.7 Proteomics and Glomerular Disease Markers 30520.8 Diabetic Nephropathy 30820.9 Lupus Nephritis 31120.10 Other Glomerular Diseases 31220.11 Summary 315

    Contents XI

  • 21 Pulmonary Proteomics 323Jan Hirsch, Lorraine B. Ware, Michael A. Matthay

    21.1 Introduction 32321.2 The Proteome of Bronchoalveolar Lavage Fluid 32321.3 BAL Studies in Animals 32521.4 Plasma and Serum Measurements 32621.5 Induced Sputum 32721.6 Pulmonary Edema Fluid 32821.7 Nasal Lavage Fluid 32921.8 Exhaled Breath Condensates 33021.9 Cell Analysis 33021.10 Frozen Tissue Slices 33521.11 Pleural Effusions 33721.12 Other Samples 33821.13 Summary 338

    22 Proteomics Providing Insights into Major Psychiatric Disorders 349Melanie Föcking, Kyla Pennington, Jane English, Michael Dunn,

    David Cotter

    22.1 Introduction 34922.1.1 Schizophrenia and Affective Disorders: Definitions and

    Epidemiology 35022.1.2 Schizophrenia and Affective Disorders: Brain Changes 35122.1.3 Effects of Psychiatric Drug Treatments on the Brain 35222.1.4 Application of Microarrays to Psychiatric Disorders 35222.1.5 The Value of Proteomic Approaches in Investigating the

    Pathophysiology of Major Psychiatric Disorders 35322.1.6 Gel-Based Proteomic Methods 35422.1.7 Non-Gel-Based Methods 35522.1.8 Creating a Subproteome 35822.2 The Importance of Validation 35922.2.1 What Samples can be Used? 35922.3 Drug Discovery 36022.3.1 Pharmacoproteomic Investigations of the Brain 36022.4 Studies from Our Group 36122.4.1 Dorsolateral Prefrontal Cortex 36122.4.2 White Matter 36222.4.3 Anterior Cingulate Cortex 36322.4.4 Insular Cortex 36622.4.5 Hippocampus 36722.4.6 Studies of Effects of Psychotropic Medication 36822.4.7 Confounding Factors Influencing Human Postmortem Brain

    Proteomic Studies 36922.4.8 Final Conclusions 369

    XII Contents

  • V Toxicity, Bacterial and Viral Infections 379

    23 Redox Proteomics Analysis of Oxidative Modified Brain

    Proteins in Alzheimer’s Disease and Mild Cognitive Impairment:

    Insights into the Progression of This Dementing Disorder 381Rukhsana Sultana, D. Allan Butterfield

    23.1 Introduction 38123.1.1 Proteomics 38123.1.2 Mass Spectrometry and Database Searching 38423.1.3 Oxidative Stress in AD 38523.1.4 Oxidative Stress in MCI 38623.2 Redox Proteomics in AD and MCI 38723.2.1 Oxidized Proteins in AD and MCI Identified Using Redox

    Proteomics 38823.2.1.1 Energy Dysfunction 38823.2.1.2 Proteasome-Related Proteins 38923.2.1.3 Cholinergic System 39023.2.1.4 Structural Proteins 39023.2.1.5 Cell Cycle, Phosphorylated Tau, and Ab Production 39123.2.1.6 pH Regulation Protein 39123.2.1.7 Neurotransmitter-Related Proteins 39123.3 Conclusion 392

    24 Toxicoproteomics: Correlating Tissue and Serum Proteomics

    in Liver Injury 403B. Alex Merrick

    24.1 The Field of Toxicoproteomics 40324.2 Toxicoproteomics and Pharmaceutical Development 40424.3 Disciplines and Platforms for Toxicoproteomic Research 40624.4 Correlating Tissue and Serum Analysis 40824.5 Toxicoproteomic Studies in Liver Injury 41024.6 Acetaminophen 41124.7 Carbon Tetrachloride 41724.8 Bromobenzene 41824.9 Wyeth 14643 41824.10 Hydrazine 41924.11 Thioacetamide 42024.12 New Blood Biomarkers in Liver Injury 42124.13 Expectations and Reality 42324.14 Future Trends and Tools in Toxicoproteomics 42424.15 Summary 425

    25 Biomarkers for Renal Disease and Uremic Toxins 435Eric Schiffer, Harald Mischak, Raymond C. Vanholder

    25.1 Introduction 435

    Contents XIII

  • 25.2 Proteome Analysis 43625.3 Technical Aspect of CE–MS 43825.3.1 Ionization and Choice of Mass Spectrometers 43825.3.2 CE–MS Coupling 43925.3.3 Coating 44025.3.4 Sample Preparation 44025.3.5 Data Evaluation 44125.3.6 Identification of Biomarkers 44125.4 Application of Proteomic Techniques to Uremic Toxicity 44425.5 Conclusion 447

    26 HIV and Other Viral Screens 453David R. Graham

    26.1 Introduction 45326.2 Current State of Clinical Virology 45326.2.1 Diagnosis 45526.2.2 Treatment 45926.3 Predicting Pathogenicity: A Need for Clinical Viral

    Proteomics 45926.4 Concepts in Virology as They Relate to Proteomics: The Problem

    of Mutation 46026.5 Understanding the Biological Limitations of the Pathogen to

    Succeed in a Proteomic Approach 46126.5.1 Culture 46126.5.2 Purity – Enveloped/Nonenveloped 46326.6 The Requirements and Approaches to Proteomic Research 46426.6.1 Decision Tree 46426.6.2 Broad-Based Approaches 46526.6.2.1 Protein Identifications or Posttranslational Modifications 46526.6.3 Liquid Chromatography-Based Approaches 46626.6.3.1 Reverse-Phase HPLC (rp-HPLC) – Separation Based on

    Hydrophobicity 46726.6.3.2 Two-Dimensional HPLC 46826.7 Pushing the Envelope of Detection 46926.7.1 Protein Interactions 47026.7.1.1 Affinity Capture Based 47026.7.2 Protein Identification 47126.7.2.1 Antibody Based 47126.7.2.2 Mass Spectrometry Based 47126.8 Bioinformatics (Host Proteins Versus Viral Proteins) 47226.8.1 Are Molecular Approaches Sufficient? 47226.8.1.1 Laying the groundwork 47326.8.1.2 Virus sequencing 475

    XIV Contents

  • 26.8.2 Toward the Real World – Emerging Clinical Applications 47526.8.2.1 Protein Arrays 47526.8.2.2 The Evolution of Protein Arrays – Multiplex Assays 47626.8.3 Assessing Immune Responses Using Arrays and Multiplex

    Assays 47626.8.3.1 Proper Data Analysis 47826.8.3.2 The Development of Clinical Cohorts for Validation 478

    VI Autoimmune Disease and Autoantibodies 481

    27 Application of Fungal Cyclic Peptides and Metabolites 483Jan Nedvěd, Miroslav Sulc, Alexandr Jegorov,

    Anastassios Giannakopulos, Vladimir Havlicek

    27.1 Introduction 48327.2 Role of Mass Spectrometry in Fungal Diagnostics 48427.3 The Importance of Secondary Fungal Metabolites: Mycotoxins and

    Peptides 48627.4 Biological Activities of Cyclic Peptides 48927.5 Mass Spectrometry of Cyclic Peptides 49027.6 Concluding Remarks 496

    28 Microarray Approaches to Autoantibody Profiling 511John M. Astle, Thomas Kodadek

    28.1 Introduction 51128.2 Native Antigen Microarrays 51328.2.1 Protein Microarrays 51428.2.2 Peptide Microarrays 51628.2.3 Glycan Microarrays 51828.2.4 Lipid Microarrays 51828.2.5 Reverse-Phase Microarrays 51928.2.6 Antibody Microarrays 52128.3 Antigen Mimic Microarrays 52328.4 Antibody Microarrays in the Clinic 527

    29 Identification of Tumor Antigen Directed Autoantibodies 533Sandra Faca, Sam Hanash

    29.1 Introduction 53329.2 Proteomic Approaches for the Identification of Tumor Antigen

    Directed Autoantibodies 53429.2.1 Some of the Potential Diagnostic Tumor Antigens Directed

    Autoantibodies Identified by 2D western blot 53529.2.1.1 PGP 9.5 535

    Contents XV

  • 29.2.1.2 Annexins 1 and 2 53729.2.1.3 Calreticulin 53829.2.1.4 Other Potential Diagnostic Antigens Identified 53929.3 Development of a High-Throughput Microarray Approach

    to Screen for Diagnostic Cancer Antigens 54029.3.1 Identification of UCH-L3 as a Potential Diagnostic Marker

    for Colon Cancer Based on Screening Using Natural Protein

    Microarray-Based Approach 54429.4 Conclusion 546

    30 Antibody and Reverse Capture Protein Microarrays

    for Clinical Proteomics 549Harvey B. Pollard, Ofer Eidelman, Meera Srivastava, Catherine Joswik,

    Stephen Rothwell, Gregory P. Mueller, David M. Jacobowitz,

    William B. Guggino, Jerry Wright, Pamela L. Zeitlin,

    Cloud P. Paweletz

    30.1 Introduction 54930.2 Antibody Microarrays 55030.2.1 A Technique in Development for Many Years 55030.2.2 Analysis of Patient Sera 55030.2.3 A Million Different Proteins in the Human Proteome 55230.2.4 Novel Expression Patterns for Hundreds of Known

    Proteins 55230.3 Reverse Capture/Reversed-Phase Protein Microarrays 55330.3.1 A Massively Parallel Platform for Clinical Applications 55330.3.2 Discovery Platform for Signaling Pathway Analysis 55330.3.3 Peptide Arrays for Discovery of Autoantibodies 55530.4 Bioinformatics for Microarray Platforms 55630.4.1 Antibody Microarrays 55630.4.1.1 Overview 55630.4.1.2 Surfaces and Printers 55630.4.1.3 Detection Strategies 55730.4.1.4 Antibody Validation 55830.4.1.5 Normalization and Quantitation 55830.4.1.6 Interpretation 56330.4.2 Reverse Capture/Phase Protein Microarrays 56330.4.2.1 Overview 56330.4.2.2 Substratum Selection 56430.4.2.3 Detection Strategies 56430.4.2.4 Antibody Validation 56430.4.2.5 Normalization and Quantitation 56530.4.2.6 Interpretation 56530.5 Conclusions 566

    XVI Contents

  • 31 Use of Antibody Microarrays in the Analysis of Inflammation,

    Autoimmunity, Viral Infection, and Cancer Metastases 571Rodney Lui, Angus Brown, Bosco Wu, Ming-Wei Lin, John Thompson,

    Filip Braet, Wayne Dyer, JoDee Lattimore, Peter Macdonald,

    Stephen Adelstein, Cristobal G. dos Remedios

    31.1 Introduction 57131.1.1 Extracellular Proteins 57231.1.2 How the Antibody Array Works 57231.1.3 Numerical Analyses 57431.1.4 Comparison of Antibody Arrays with Flow Cytometry 57531.2 Inflammation 57531.2.1 Ischemic Heart Disease 57531.3 Autoimmune Disease 58231.3.1 Background to Systemic Lupus Eythematosus (SLE) 58231.3.2 Features of SLE 58231.4 Melanoma Cancer 58331.4.1 The Diagnostic Question 58331.4.2 Outlier Patients 58331.4.3 Hierarchical Clustering 58431.5 Vaccine Inflammation 58431.5.1 Smallpox Vaccination 58431.5.2 DotScan Data 58631.6 Concluding Remarks 588

    VII Translation: Discovery to the Clinic 593

    32 The Future: Translation from Discovery to the Clinic – Roles

    of HUPO and Industry in Biomarker Discovery 595Gilbert S. Omenn, Peipei Ping

    32.1 Introduction 59532.2 Brief Introduction of HUPO Initiatives 59632.3 General Lessons Illustrated with the Human Plasma Proteome

    Project (HPPP) 59732.4 Brief Descriptions of Other HUPO Initiatives 60232.4.1 The Human Liver Proteome Project (HLPP) 60232.4.2 The HUPO Brain Proteome Project (HBPP) 60332.4.3 The HUPO Cardiovascular Initiative (HCVI) 60332.4.4 The Human Disease Glycomics/Proteome Initiative (HGPI) 60432.4.5 The HUPO Proteomic Standards Initiative (PSI) 60432.4.6 The HUPO Antibody Initiative (HAI) 60532.4.7 The Umbrella HUPO Biomarker Initiative has Several

    Components 60532.4.7.1 The Human Kidney-Urine Proteome Project (HKUPP) 60532.4.7.2 The HUPO Cancer Biomarker Initiative 605

    Contents XVII

  • 32.4.7.3 HUPO Biomarkers Bioinformatics (BioBio) Initiative 60632.4.8 The HUPO/Invitrogen Test Sample Project 60632.5 General Strategies for the Use of Plasma or Serum for

    Protein Biomarkers 60732.6 Pitfalls in Biomarker Discovery and Validation Studies 60832.7 Concluding Remarks 609

    33 Requirements of a Good Biomarker: Translation

    into the Clinical Laboratory 615Mario Plebani, Martina Zaninotto, Monica Maria Mion

    33.1 Introduction 61533.2 Translational Research 61533.3 Biomarker: What Does It Mean? 61533.3.1 Biomarkers and Drug Development/Evaluation 61733.3.2 Biomarkers in Laboratory Medicine 61833.4 Development of Biomarkers: From Discovery to

    Clinical Application 62133.5 Discovery 62233.5.1 Goal (Targeted)-Dependent Options 62333.5.2 Technologies 62333.6 Validation 62433.7 Standardization 62533.8 Clinical Association and Clinical Benefit 62633.9 Conclusions 628

    34 Translation of Protein Biomarkers for Clinical Development 633Ian McCaffery, V. Dan Fitzpatrick, Shen Wu Wang, John M. Rossi,

    Haifeng Bao, Sid V. Suggs, John Ferbas, Scott D. Patterson

    34.1 Introduction 63334.2 Development of Biomarkers for Early Drug Development 63434.3 Marker of Biochemical Coverage – Development of an Assay of

    Phospho S6 Protein as a PD Biomarker for Rapamycin Inhibition

    of the mTOR Pathway 63834.3.1 PI3K/Akt/mTOR Pathway Background 63834.3.2 Therapeutics Directed at Inhibition of the mTOR Pathway 64034.3.3 Existing PBMC-Based PD Biomarker Assay of mTOR Inhibition 64034.3.4 Development of a Flow Cytometry Based PD Biomarker Assay and

    Comparison of PBMC and Whole Blood Results 64134.3.5 Summary of Comparison of PD Biomarker Assays 64634.4 Discussion 64734.5 Summary 650

    Index 653

    XVIII Contents

  • Editor’s Overview

    Clinical proteomic is an emerging discipline within proteomics that is exclusively

    focused on its application to biomedicine. This book focuses on biomarker discovery

    and validation and the pipeline required to increase the probability of success from the

    view point of proteomics. It is hoped that sophisticated proteomic techniques and a

    better understanding of basic pathophysiologies and protein chemistries will enhance

    early disease detection, prognosis, risk stratification and therapeutic monitoring.

    Clinical proteomics is a transdisciplinary field garnering aspects coming from

    epidemiology, clinical chemistry, clinical medicine and proteomics. It is being built

    on these disciplines, adapting experimental design, application, technologies and

    knowledge to produce acceptable workflows. With respect to proteomics, success will

    depend on the field developing and testing technologies and approaches for both

    discovery and/or validation of biomarkers specifically in body fluids. The first section

    of the book (Chapters 1–9) is composed of chapters dealing with the underlying

    technologies which underpin proteomic applications – exploring the potential and

    limitations of the various methods specifically for biomarker analysis. The subse-

    quent sections highlight examples where proteomics is being applied in various

    relevant clinical areas: cancer (Chapters 10–13); cardiovascular disease (Chapters 14–

    19); vascular disease including stroke (Chapters 20–23); toxicity, bacterial and viral

    assessment (Chapters 24–27); and autoantibody signatures (Chapters 28–31). In the

    final section, three chapters deal with the broader context of clinical proteomics from

    the perspective of clinical chemistry, academia and industry.

    The dream of clinical proteomics is that patients’ lives are extended and the quality

    of life can be improved by including proteomics in a clinicians’ diagnostic arsenal.

    This book provides insight into the collective knowledge researchers have gained

    over the last several years into a single resource book. It is aimed at addressing the

    current issues surrounding the application of proteomics to the development of

    biomarkers. There has been considerable hype around clinical proteomics and its

    potential to change medicine. That potential remains. Nevertheless, there are many

    challenges which need to be met.

    Sincerely

    The Editors

    Michael Dunn and Jennifer Van Eyk

    XIX

  • Short Summary

    This is the first book to overview the emerging field of clinical proteomics with

    sections focused on the latest technologies and summaries by leaders in the fields of

    cancer, cardiovascular disease, toxicology, bacterial, viral and autoantibody assess-

    ments. This book provides insight into the collective knowledge researchers have

    gained over the last several years into a single resource book. It is aimed at

    addressing the current issues surrounding the application of proteomics to the

    development of biomarkers. There has been considerable excitement around clinical

    proteomics and its potential to changemedicine. That potential remains but clinical,

    technical and translational issues still need to be overcome.

    Acknowledgements

    The reasons for creating a book are numerous. The people to thanks are also

    numerous. We thank the members of our labs who help to develop and create the

    vision for biomarker discovery and our families, who provide the support to allow

    those dream. Finally, we thank the authors who shared their insights and under-

    standing.

    XX Editor’s Overview

  • List of Contributors

    Stephen Adelstein

    Clinical Immunology

    Royal Prince Alfred Hospital

    Camperdown

    Australia

    Ruedi H. Aebersold

    Institute of Molecular Systems

    Biology,

    ETH Zurich

    and Faculty of Science

    University of Zurich

    Wolfgang-Pauli-Str. 16

    8093 Zurich

    Switzerland

    N. Leigh Anderson

    The Plasma Proteome Institute

    Washington DC 20009-3450

    USA

    John M. Arthur

    Medical University of

    South Carolina

    Division of Nephrology

    PO Box 250623

    Charleston, SC 29425-2220

    USA

    John M. Astle

    Departments of Internal Medicine

    and Molecular Biology

    University of Texas Southwestern

    Medical Center

    5323 Harry Hines Blvd

    Dallas, Texas 75390-9185

    USA

    Brian M. Bagley

    Calibrant Biosystems,

    910 Clopper Road,

    Suite 220N,

    Gaithersburg

    MD 20878

    USA

    Rosamonde E. Banks

    Cancer Research UK

    Clinical Centre

    St James’s University Hospital

    Beckett Street

    Leeds LS9 7TF

    UK

    Haifeng Bao

    Amgen, Inc.

    Molecular Sciences

    One Amgen Center Drive,

    Mail Stop 1-1A

    Thousand Oaks, CA 91320-1799

    USA

    XXI

  • Timothy J. Barder

    Eprogen, Inc.,

    8205 S. Cass Ave. Ste. 106,

    Darien, IL 60561

    USA

    Luca Bini

    Department of Molecular Biology

    University of Siena

    Via Fiorentina

    153100 Siena

    Italy

    Mary E. Bollard

    Department of Biochemistry

    University of Cambridge

    Tennis Court Road

    Cambridge

    UK

    Filip Braet

    Australian Key Centre for

    Microscopy and Microanalysis

    The University of Sydney

    Sydney 2006

    Australia

    Teresa A. Brentnall

    Department of Medicine

    University of Washington

    1959 NE Pacific St

    Seattle, WA 98195

    USA

    Angus Brown

    Bosch Institute (F13)

    The University of Sydney

    Sydney 2006

    Australia

    D. Allan Butterfield

    Center of Membrane Sciences

    University of Kentucky

    Lexington, KY 40506

    USA

    Valerie S. Calvert (12)

    Center for Applied Proteomics and

    Molecular Medicine

    George Mason University

    10900 University Blvd, MS 4E3

    Manassas, VA 20110

    USA

    Richard M. Caprioli

    Department of Biochemistry

    Vanderbilt-Ingram Comprehensive

    Cancer Center

    9160 MRB III

    Vanderbilt University

    School of Medicine

    Nashville, TN 37232-8575

    USA

    Julio E. Celis

    Inst. Cancer Bio/Human

    Genome Res

    Danish Cancer Society

    Strandboulevarden 49

    2100 Copenhagen

    Danmark

    Daniel W. Chan

    Biomarker Discovery Center

    Johns Hopkins University

    Baltimore, MD

    USA

    Ru Chen

    Department of Medicine

    University of Washington

    1959 NE Pacific St

    Seattle, WA 98195

    USA

    Songming Chen

    The Van Andel Research Institute

    333 Bostwick

    Grand Rapids, MI 49503

    USA

    XXII List of Contributors

  • David E Clemmer

    Department of Chemistry

    Indiana University

    800 E. Kirkwood Ave.

    Bloomington, IN 47405

    USA.

    Garry L. Corthals

    Biomedical Proteomics Research

    Group

    Geneva University Hospital/LCCC

    24 rue Micheli-du-Crest

    1211 Genève 14

    Switzerland

    David R. Cotter

    Johns Hopkins University

    School of Medicine

    725 N. Wolfe St.

    B7 Biophysics

    Baltimore, MD 21205

    USA

    Robert J. Cotter

    Johns Hopkins University

    School of Medicine

    725 N. Wolfe St.

    B7 Biophysics

    Baltimore, MD 21205

    USA

    Jacques Deshusses

    Biomedical Proteomics Research

    Group,

    Department of Structural Biology

    and Bioinformatics,

    Geneva University,

    1 Michel Servet,

    1211 Geneva,

    Switzerland

    Don L. DeVoe

    Department of Mechanical

    Engineering and Bioengineering

    Program

    University of Maryland,

    College Park, MD 20742

    USA

    Pamela M. Donoghue

    Immune Regulation Research

    Group

    Trinity College

    Dublin 2

    Ireland

    Cristobal G. dos Remedios

    Bosch Institute (F13)

    The University of Sydney

    Sydney 2006

    Australia

    Michael J. Dunn

    University College Dublin

    Proteome Research Centre

    UCD Conway Institute of

    Biomolecular and Biomedical

    Research

    Dublin 4

    Ireland

    Wayne Dyer

    Australian Red Cross Blood Service

    Clarence Street

    Sydney 2000

    Australia

    Marta Dziechciarkova

    Palacky University and

    University Hospital Olomouc

    Faculty of Medicine and Dentistry

    Puskinova 6

    77520 Olomouc

    Czech Republic

    List of Contributors XXIII

  • Petr Dzubak

    Faculty of Medicine and Dentistry

    Palacky University and University

    Hospital Olomouc

    Puskinova 6

    77520 Olomouc

    Czech Republic

    Kirsten H. Edmiston

    Center for Applied Proteomics and

    Molecular Medicine

    George Mason University

    10900 University Blvd, MS 4E3

    Manassas, VA 20110

    USA

    Ofer Eidelman

    Department of Anatomy,

    Physiology and Genetics

    USU School of Medicine (USUHS)

    4301 Jones Bridge Road

    Bethesda, MD 20814

    USA

    Andrew Emili

    Banting and Besst Department of

    Medical Research and Department

    of Medical Genetics and

    Microbiology

    Donnelly Centre for Cellular and

    Biomolecular Research

    University of Toronto

    Canada

    Jane English

    Royal College of Surgeons in

    Ireland

    Department of Psychiatry

    Education and Research Centre

    Beaumont Hospital

    Dublin 9

    Ireland

    Virginia Espina

    Center for Applied Proteomics and

    Molecular Medicine

    George Mason University

    10900 University Blvd, MS 4E3

    Manassas, VA 20110

    USA

    Sandra Faca

    Fred Hutchinson Cancer Research

    Center

    1100 Fairview Avenue N., M5-C800

    P.O. Box 19024

    Seattle, WA 98109

    USA

    Xueping Fang

    Department of Chemistry and

    Biochemistry

    University of Maryland,

    College Park, MD 20742

    USA

    John Ferbas

    Amgen, Inc.

    Clinical Immunology

    One Amgen Center Drive,Mail Stop

    30E-3-C

    Thousand Oaks, CA 91320-1799

    USA

    Roisean E. Ferguson

    Cancer Research UK Clinical

    Centre

    St James’s University Hospital

    Beckett Street

    Leeds LS9 7TF

    UK

    XXIV List of Contributors

  • V. Dan Fitzpatrick

    Amgen, Inc.

    Molecular Sciences

    One Amgen Center Drive, Mail

    Stop 1-1A

    Thousand Oaks, CA 91320-1799

    USA

    Melanie Föcking

    Royal College of Surgeons in

    Ireland

    Department of Psychiatry

    Education and Research Centre

    Beaumont Hospital

    Dublin 9

    Ireland

    Qin Fu

    Department of Medicine

    Johns Hopkins Bayview Proteomics

    Center

    Johns Hopkins University

    Baltimore, MD 21224

    USA

    Suresh Jivan Gadher

    Beckman Coulter International S.A.

    22, rue Juste Olivier, Casa Postale

    1059

    1260 Nyon 1

    Switzerland

    Robert E. Gerszten, M.D.

    Cardiology Division and Center for

    Immunology and Inflammatory

    Diseases

    Massachusetts General Hospital

    East-8307

    149 13th Street

    Charlestown, MA 02129

    USA

    Anastassios Giannakopulos

    Institute of Microbiology

    Academy of Sciences of the

    Czech Republic

    Videnska 1083

    142 20 Prague

    Czech Republic

    David R. Graham

    Bayview NHLBI Proteomics Center

    Johns Hopkins School of Medicine

    Bayview Campus

    5200 Eastern Avenue

    Baltimore, MD 21224

    USA

    Anthony Gramolini

    Department of Physiology and

    Heart and Stroke/Richard Lewar

    Center of Excellence

    University of Toronto

    112 College St, Rm 307

    Toronto, Ontario

    Canada

    William B. Guggino

    Department of Physiology,

    Johns Hopkins University

    School of Medicine,

    JHMI,

    Baltimore, MD 21205

    USA

    Rebekah L. Gundry (18)

    Johns Hopkins University

    School of Medicine

    725 N. Wolfe St.

    B7 Biophysics

    Baltimore, MD 21205

    USA

    Brian B. Haab

    The Van Andel Research Institute

    333 Bostwick

    Grand Rapids, MI 49503

    USA

    List of Contributors XXV

  • Marian Hajduch

    Faculty of Medicine and Dentistry

    Palacky University and University

    Hospital Olomouc

    Puskinova 6

    77520 Olomouc

    Czech Republic

    Petr Halada

    Institute of Microbiology

    Czech Academy of Sciences

    Videnska 1083

    14220 Praha

    Czech Republic

    Samir M. Hanash

    Fred Hutchinson Cancer Research

    Center

    1100 Fairview Avenue N., M5-C800

    P.O. Box 19024

    Seattle, WA 98109

    USA

    Gerald Hart

    725 N Wolfe Street

    Biochemistry Department

    Baltimore, MD 21205

    USA

    Vladimir Havlicek

    Institute of Microbiology

    Academy of Sciences of the Czech

    Republic

    Videnska 1083

    142 20 Prague

    Czech Republic

    Jan Hirsh

    Department of Anesthesia and

    Perioperative Medicine

    University of California

    505 Parnassus Ave. HSW 825

    San Francisco, CA 94143-0130

    USA

    Denis F. Hochstrasser

    Biomedical Proteomics Research

    Group

    Clinical Chemistry Laboratory

    Geneva University Hospital

    Geneva

    Switzerland

    Don Hunt

    Chemistry Department

    University of Virginia

    Charlottesville, VA 22908

    USA

    David M. Jacobowitz

    Laboratory of Clinical Science,

    National Institute for Mental

    Health,

    Bethesda, MD 20892

    USA

    Jon M. Jacobs

    Biological Sciences Division

    Pacific Northwest National

    Laboratory

    P. O. Box 999

    Richland, WA 99352

    USA

    Alexandr Jegorov

    Institute of Microbiology

    Academy of Sciences of the Czech

    Republic

    Videnska 1083

    142 20 Prague

    Czech Republic

    XXVI List of Contributors

  • Robert L. Jesse

    Department of Internal Medicine

    Cardiology Division

    Medical College of Virginia

    Virginia Commonwealth University

    Veterans Health Administration

    12th and Marshall Streets

    Richmond, VA 23298-0051

    USA

    Catherine Joswik

    Department of Anatomy

    Physiology and Genetics

    USU School of Medicine (USUHS)

    4301 Jones Bridge Road

    Bethesda, MD 20814

    USA

    Urban A. Kiernan

    Intrinsic Bioprobes, Inc.

    2155 E. Conference Drive, Suite 104

    Tempe, AZ 85284

    USA

    Thomas Kodadek

    University of Texas

    Southwestern Medical Center

    Departments of Internal Medicine

    and Molecular Biology

    5323 Harry Hines Blvd

    Dallas, TX 75390-9185

    USA

    Hana Kovarova

    Czech Academy of Sciences

    Institute of Animal Physiology and

    Genetics

    Rumburska 89

    27721 Libechov

    Czech Republic

    JoDee Lattimore

    Sydney Melanoma Unit

    Royal Prince Alfred Hospital

    Camperdown

    Australia

    Cheng S. Lee

    Department of Chemistry and

    Biochemistry

    University of Maryland

    College Park, MD 20742

    USA

    Gregory D. Lewis

    Center for Immunology and

    Inflammatory Diseases

    Massachusetts General Hospital

    Charlestown, MA and Harvard

    Medical School

    Boston, MA

    USA

    Daniel C. Liebler

    Southwest Environmental Health

    Sciences Center

    College of Pharmacy

    University of Arizona

    Tucson, AZ 85721-0207

    USA

    Ming-Wei Lin

    Clinical Immunology

    Royal Prince Alfred Hospital

    Camperdown

    Australia

    Lance A. Liotta

    Center for Applied Proteomics and

    Molecular Medicine

    George Mason University

    10900 University Blvd, MS 4E3

    Manassas, VA 20110

    USA

    David M. Lubman

    Department of Surgery

    1150 West Medical Center Drive

    Ann Arbor, MI 48109-1055

    USA

    List of Contributors XXVII

  • Rodney Lui

    Bosch Institute (F13)

    The University of Sydney

    Sydney 2006

    Australia

    Peter MacDonald

    Cardiopulmonary Transplant Unit

    St. Vincent’s Hospital

    Darlinghurst, 2010

    Australia

    Ian McCaffery

    Amgen, Inc.

    Molecular Sciences

    One Amgen Center Drive,

    Mail Stop 1-1A

    Thousand Oaks, CA 91320-1799

    USA

    Dr. Ciara A. McManus

    University College Dublin

    Proteome Research Centre

    UCD Conway Institute of

    Biomolecular and Biomedical

    Research

    Dublin 4

    Ireland

    Michael A. Matthay

    Department of Anesthesia and

    Perioperative Medicine

    University of California

    505 Parnassus Ave. HSW 825

    San Francisco, CA 94143-0130

    USA

    Bruce Alex Merrick

    Proteomics Group

    National Institute of Environmental

    Health Sciences

    P.O. Box 12233

    Research Triangle Park, NC 27709

    USA

    Monica Maria Mion

    Department of Laboratory Medicine

    University-Hospital

    Via Giustiniani 2

    35128 Padova

    Italy

    Harald Mischak

    Mosaiques-Diagnostics and

    Therapeutics AG

    Mellendorfer Str. 7-9

    30173 Hannover

    Germany

    David E. Misek

    University of Michigan

    Department of Surgery

    1150 West Medical Center Drive

    Ann Arbor, MI 48109-1055

    USA

    Gregory P. Mueller

    Department of Anatomy,

    Physiology and Genetics

    USU School of Medicine (USUHS)

    4301 Jones Bridge Road

    Bethesda, MD 20814

    USA

    Dobrin Nedelkov

    Intrinsic Bioprobes, Inc.

    2155 E. Conference Drive, Suite 104

    Tempe, AZ 85284

    USA

    Jan Nedvěd

    Institute of Microbiology

    Academy of Sciences of the Czech

    Republic

    Videnska 1083

    142 20 Prague

    Czech Republic

    XXVIII List of Contributors