Menu Expand
Metal Chelation in Medicine

Metal Chelation in Medicine

Robert R Crichton | Roberta J Ward | Robert C Hider

(2016)

Additional Information

Book Details

Abstract

Metal chelators are emerging as versatile tool with many medical applications. Their versatility allows them to be used in chelation therapy for treating diseases caused by toxic and heavy metal poisoning, chelating agents are capable of binding to toxic metal ions to form complex structures which are easily excreted from the body removing them from intracellular or extracellular spaces. In addition, metal chelators can also be applied as contrast agents in MRI scanning.
Metal Chelation in Medicine provides a clear and timely perspective on the role of chelating agents in the management of metal intoxications and storage diseases. Written by leaders in the field of chelators, this publication is at the cutting-edge of the subject. It covers a broad range of topics such as the use of metal chelators in non-invasive assessment of brain iron overload, and the treatment of systemic iron overload and neurodegenerative diseases. As such it is particularly valuable to clinicians treating metal poisonings and metal storage diseases. However, it is also a useful text for researchers, industry professionals and university students with a specific interest in medicinal chemistry, chelation, metal ions, imaging and non-invasive techniques.

Table of Contents

Section Title Page Action Price
Cover Cover
Metal Chelation in Medicine i
Preface v
Contents vii
Chapter 1 - Metal Toxicity – An Introduction 1
1.1 Introduction 1
1.2 Essential Metals 2
1.2.1 Sodium and Potassium 2
1.2.2 Magnesium and Calcium 2
1.2.3 Zinc 3
1.2.4 Other Essential Metals 3
1.3 Toxicity Due to Essential Metals 4
1.3.1 Sodium and Potassium 4
1.3.2 Calcium and Magnesium 7
1.3.3 Zinc 8
1.3.4 Other Essential Metals 9
1.3.4.1 Cobalt Toxicity 9
1.3.4.2 Manganese Toxicity 9
1.3.4.3 Iron and Copper Toxicity 10
1.3.4.4 Molybdeum Toxicity 12
1.4 Non-Essential Metals 12
1.5 Toxicity Due to Non-Essential Metals 13
1.5.1 Lead 13
1.5.2 Cadmium 14
1.5.3 Mercury 15
1.5.4 Aluminium 15
1.6 Sources and Routes of Exposure with Particular Reference to Non-Essential Metals 18
1.7 Conclusions 19
Abbreviations 20
References 20
Chapter 2 - Basic Principles of Metal Chelation and Chelator Design 24
2.1 Introduction 24
2.2 Ligand Chemistry 26
2.3 Ligand Selectivity 29
2.4 Comparison of Bidentate Ligands 31
2.4.1 Catechols 31
2.4.2 Hydroxamates 33
2.4.3 Hydroxypyridinones 33
2.4.4 Hydroxypyranones 35
2.4.5 Aliphatic Diamines 35
2.4.6 Heterocyclic Amines 35
2.4.7 Hydroxyquinolines 36
2.4.8 Aminocarboxylates (Including Oligodentate Ligands) 36
2.4.9 Hydroxycarboxylates (Including Tri- and Hexadentate Ligands) 37
2.5 Ligand Denticity – The Chelate Effect 37
2.6 Complex Lability 39
2.7 Redox Activity 41
2.8 Biological Properties 43
2.9 Lipophilicity and Molecular Weight 43
2.10 Ligand Metabolism 44
2.11 Ligand Binding to Serum Albumin 45
2.12 Chelator Pharmacokinetics 47
2.13 Toxicity of Chelators 49
2.13.1 Inhibition of Metal-Dependent Enzymes 49
2.13.2 Nephrotoxicity 51
2.13.3 Neurotoxicity 51
2.13.4 Immunotoxicity 51
2.13.5 Genotoxicity 52
2.14 Chelators and Nutrition 52
2.15 Conclusions 53
Abbreviations 53
References 53
Chapter 3 - Chelation Therapy For Heavy Metals 56
3.1 Introduction 56
3.2 General Strategies Against Heavy Metal Intoxication 58
3.2.1 Toxicology of Heavy Metals 58
3.2.2 Treatment for Heavy Metal Intoxication 59
3.2.2.1 Conventional Chelators 61
3.2.2.2 New Chelators 61
3.2.3 Drawbacks and Misuse of Chelators 62
3.3 Arsenic 63
3.3.1 Absorption and Metabolism of Arsenicals 63
3.3.2 Toxicity of Arsenicals 64
3.3.2.1 Mechanism of Arsenic Toxicity 64
3.3.2.2 Clinical Toxicity of Arsenicals 65
3.3.3 Diagnostic Measures and Interventional Levels 66
3.3.4 Chelation Therapy for Arsenic Intoxication 66
3.3.4.1 Chronic Arsenic Poisoning 68
3.4 Mercury 68
3.4.1 Absorption and Metabolism of Mercurials 70
3.4.1.1 Hg0 70
3.4.1.2 Inorganic Mercury 71
3.4.1.3 Organic Mercury 72
3.4.2 Toxicology of Mercury 73
3.4.2.1 Mechanism of Mercury Toxicity 73
3.4.2.2 Clinical Toxicity of Mercury 74
3.4.3 Diagnostics and Intervention Levels 75
3.4.4 Chelation Treatment for Mercury Poisoning 76
3.5 Lead 78
3.5.1 Lead Absorption and Metabolism 78
3.5.2 Toxicity of Lead 80
3.5.2.1 Mechanism of Lead Toxicity 80
3.5.3 Diagnostic Measures and Intervention Levels 82
3.5.4 Treatment of Lead Poisoning 82
3.6 Cadmium 85
3.6.1 Absorption and Metabolism of Cadmium 86
3.6.2 Toxicity of Cadmium 87
3.6.2.1 Mechanism of Cadmium Toxicity 88
3.6.2.2 Clinical Toxicity of Cd 88
3.6.3 Diagnostic Markers and Intervention Level for Cd Intoxication 90
3.6.4 Chelation Treatment for Cadmium Poisoning 90
3.7 Noble Metals 92
3.7.1 Silver Poisoning 92
3.7.2 Gold Poisoning 93
3.8 Other Metals 94
3.8.1 Acute Iron Poisoning 94
3.8.2 Acute Copper Poisoning 95
3.8.3 Thallium Poisoning 95
3.9 Conclusion 96
Abbreviations 97
References 98
Chapter 4 - Treatment of Systemic Iron Overload 106
4.1 Consequences of Transfusional Iron Overload 106
4.1.1 Iron Homeostasis in the Absence of Blood Transfusion 106
4.1.2 Effects of Inherited Anaemias and Blood Transfusion on Iron Homeostasis 107
4.2 Impact of Transfusion on Iron Distribution and its Consequences 109
4.3 Desirable Features of Clinically Useful Iron Chelators 113
4.3.1 High Iron Binding Constant and Selectivity for Iron 113
4.3.2 Chelation of Iron Pools for Balance Without Inhibition of Key Metabolic Pools 114
4.4 Monitoring Iron Overload and Its Treatment 117
4.4.1 Monitoring Trajectory of Iron Overload and Distribution 117
4.4.1.1 Serum Ferritin (SF) 117
4.4.1.2 Liver Iron Concentration (LIC) Monitoring 120
4.4.1.3 Methods for Measuring LIC 121
4.4.1.4 Myocardial Iron Estimation (T2* or Other Measures) 122
4.4.1.5 Monitoring of Other Organ Functions and Iron-Mediated Damage 122
4.4.1.6 Urinary 24 h Iron Estimation 123
4.4.1.7 Plasma Non-Transferrin Iron (NTBI) and Labile Plasma Iron (LPI) 123
4.5 Properties and Clinical Beneficial Effects of Available Iron Chelators 124
4.5.1 Desferrioxamine (Desferal) DFO 124
4.5.1.1 Physicochemical Properties and Iron Binding 124
4.5.1.2 Pharmacokinetics of Free Ligand and Iron Complexes 125
4.5.1.3 Effects on Iron Balance and Ferritin 125
4.5.1.4 Long-Term Effects on Survival 126
4.5.1.5 Effects on the Heart 126
4.5.1.6 Other Long-Term Effects on Morbidity 127
4.5.2 Deferiprone (DFP) 127
4.5.2.1 Physicochemical Properties and Iron Binding 127
4.5.2.2 Pharmacokinetics 128
4.5.2.3 Effects on Iron Balance 129
4.5.2.4 Effects on the Heart 129
4.5.3 Deferasirox (Exjade®) DFX 130
4.5.3.1 Pharmacokinetics and Metabolism 130
4.5.3.2 Iron Excretion and Iron Balance 131
4.5.3.3 Effects on the Heart 132
4.6 Unwanted Effects of Iron Chelators 132
4.6.1 Role of Iron Deprivation in Chelator Toxicity 132
4.6.2 Desferrioxamine Tolerability and Unwanted Effects 133
4.6.3 Deferiprone Tolerability and Unwanted Effects 134
4.6.4 Deferasirox Tolerability and Unwanted Effects 134
4.7 Practical Use of Chelators: A Personal Approach 135
4.7.1 Patients with Acceptable Levels of Body Iron but Continuing Transfusion 135
4.7.2 Patients with Unacceptably High Levels of Body Iron 136
4.7.3 Patients with Unacceptably High Levels of Myocardial Iron 136
4.7.4 Patients in Heart Failure 137
4.7.5 Patients with Rapidly Falling Serum Ferritin or Low Values <1000 µ L−1 138
4.7.6 Patients not Responding to Monotherapy Regimes: Combined Chelators 139
4.7.7 Combinations of DFP with DFO 139
4.7.8 Combinations of DFO Plus DFX 140
4.7.9 Combination of DFP Plus DFX 140
4.8 Conclusions 140
Abbreviations 141
References 141
Chapter 5 - Treatment of Neurodegenerative Diseases by Chelators 153
5.1 Introduction 153
5.2 The Aging Brain 154
5.2.1 Brain Iron Homeostasis and Aging 154
5.2.2 Brain Copper Homeostasis and Aging 156
5.2.3 Brain Zinc Homeostasis and Aging 156
5.3 Inflammation and Aging 157
5.4 Neurodegenerative Diseases 157
5.5 Blood Brain Barrier 158
5.6 Metal Ions and Neurodegenerative Diseases 159
5.6.1 Parkinson’s Disease (PD) 160
5.6.2 Alzheimer’s Disease (AD) 163
5.6.3 Intracerebral Haemorrhage 166
5.6.4 Multiple Sclerosis 168
5.6.5 Friederich’s Ataxia 168
5.7 Neurodegeneration with Brain Iron Accumulation (NBIA) Diseases 169
5.7.1 Mutation in Genes Associated with Iron Metabolism 169
5.7.1.1 Aceruloplasminaemia 169
5.7.1.2 Neuroferritinopathy 172
5.7.1.3 Pantothenate Kinase-Associated Neurodegeneration 172
5.7.1.4 Huntington’s Disease 174
5.7.1.5 Wilson’s Disease 175
5.8 Macular Degeneration 176
5.9 Conclusion and Perspectives 177
Abbreviations 177
References 177
Chapter 6 - Chelation of Actinides 183
6.1 The Medical and Public Health Relevance of Actinide Chelation 183
6.1.1 Actinide Metabolism and Clinical Course 184
6.1.2 Current Treatment Recommendations for Actinide Contamination 186
6.1.3 Limitations of Current Therapies 187
6.2 Designing Chemical Structures for Actinide Chelation 187
6.2.1 Coordination Chemistry Criteria 188
6.2.2 Synthetic Approaches to New Actinide-Selective Agents 188
6.2.2.1 Polyamino-Carboxylic Acid Derivatives 189
6.2.2.2 Siderophore Mimics 190
6.2.2.3 Poly-Phosphonic Acid Chelators and Macrocyclic Structures 192
6.3 Evaluating Actinide Chelation Efficacy 193
6.3.1 In vitro Evaluation Techniques 193
6.3.1.1 Solution Thermodynamics 193
6.3.1.2 High-Throughput Screening Methods 194
6.3.1.3 In vitro and Ex vivo Binding 195
6.3.2 In vivo Efficacy Determination 196
6.3.2.1 Animal Model Selection 196
6.3.2.2 In vivo Decorporation Studies 197
6.4 Development of Viable Actinide Chelation Treatments 199
6.4.1 Formulation Development 199
6.4.1.1 Structural Modifications of DTPA 200
6.4.1.2 Pharmaceutical Approaches 201
6.4.2 Safety Determination and Regulatory Approval 202
6.4.2.1 The Animal Rule 203
6.4.2.2 Current Status of Existing Actinide Chelation Products 205
Abbreviations 206
References 207
Chapter 7 - Evaluation of Iron Overload by Non-Invasive Measurement Techniques 213
7.1 Introduction 213
7.2 Principles of Iron Measurement Techniques 215
7.2.1 Reference Standards for Iron Quantification Methods 215
7.2.1.1 Liver Biopsy 215
7.2.1.2 Quantitative Heart Biopsy and Autopsy 216
7.2.1.3 Bone Marrow Iron 216
7.2.1.4 Post-Mortem Brain Iron 216
7.2.2 Biochemical Properties of Iron Storage Molecules 217
7.2.3 Magnetic Properties of Iron Storage Molecules 218
7.2.3.1 Specific Magnetic Volume Susceptibility (Electron Magnetism) 220
7.2.3.2 Iron Concentration and Wet-to-Dry Weight Ratio 223
7.2.3.3 Other Magnetic Properties 224
7.3 Iron Measurements: Technique, Analysis, and Calibration 224
7.3.1 Spin-Echo Relaxometry (R2/T2) 226
7.3.2 Spin-Lattice Relaxometry (T1/R1) 229
7.3.3 Gradient-Echo Relaxometry (T2*/R2*) 229
7.3.3.1 Hepatic R2* 230
7.3.3.2 Cardiac R2* (T2*) 230
7.3.3.3 Chemical Shift Relaxometry (CSR) 232
7.3.4 Biomagnetic Susceptometry 233
7.3.4.1 Biomagnetic Liver Susceptometry (Electronic Susceptometry) 233
7.3.4.2 MR-Biosusceptometry and MR-QSM (Nuclear Susceptometry) 234
7.3.5 Other MRI Iron Measurement Techniques 235
7.3.6 Quantitative X-ray Iron Measurement Techniques 237
7.4 Applications of In vivo Iron Assessment 237
7.4.1 Liver 238
7.4.2 Spleen 239
7.4.3 Heart 240
7.4.4 Pancreas 240
7.4.5 Pituitary 241
7.4.6 Brain 242
7.4.7 Iron in Other Organs, Glands, and Tissue 244
7.4.7.1 Kidney 244
7.4.7.2 Adrenal Glands 245
7.4.7.3 Thyroid 245
7.4.7.4 Testes and Ovaries 245
7.4.7.5 Bone Marrow 246
7.5 Conclusion and Prospects 247
Abbreviations 248
Acknowledgement 248
References 248
Chapter 8 - Chelators for Diagnostic Molecular Imaging with Radioisotopes of Copper, Gallium and Zirconium 260
8.1 Introduction 260
8.1.1 Positron Emission Tomography and Molecular Imaging with Peptides and Proteins 261
8.1.2 Reactive Functional Groups for Attachment to Biomolecules 262
8.1.3 Requirements for Chelators that Complex Positron-Emitting Metal Radioisotopes 263
8.2 Macrocyclic Chelators for Copper-64: Lessons in Kinetic Stability 265
8.2.1 Cu Complexes of Macrocycles: Radiolabelling and In vivo Stability 266
8.2.1.1 Cyclen, Cyclam, Teta and Dota – Not All Macrocycles are Equal to the Task! 266
8.2.1.2 Alternatives to Teta and Dota – Towards Greater In vivo Stability 269
8.2.1.3 Nota and Sarcophagine Derivatives – The State of the Art in Bifunctional Chelators for 64Cu 271
8.2.2 Kinetic Stability is Critical to In vivo Stability – You Can’t Have One Without the Other 275
8.2.3 Targeting Based on Chelate Reactivity: Bioreduction and Dissociation 281
8.2.4 Outlook for Clinical Applications of Copper-64 282
8.3 Acyclic and Macrocyclic Chelators for Gallium-68: A Short Half-Life Necessitates Efficient Radiolabelling 283
8.3.1 Gallium-68 in the Clinic: Its Current Utility and Future Potential 283
8.3.2 Macrocycles for Gallium-68 286
8.3.3 Halfway Between Macrocycles and Open-Chain Chelators: Data Derivatives 289
8.3.4 Acyclic Chelators for Gallium-68 Provide Rapid Radiolabelling at Room Temperature 290
8.3.5 Siderophores for Coordination of Gallium-68 294
8.3.6 Chelator Design and Clinical Impact 295
8.4 Development of Chelators for Zirconium-89: Work in Progress 295
8.4.1 Hexadentate Chelators for Zirconium-89: Sufficient, But Suboptimal 296
8.4.2 Octadentate Chelators for 89Zr4+: Will Saturation of the Coordination Sphere Provide Enhanced Stability In vivo 299
8.4.3 Metastable Lipophilic Zirconium Chelates 302
8.5 Conclusions 303
Abbreviations 304
Acknowledgements 305
References 305
Subject Index 313