Schweitzer Fachinformationen
Wenn es um professionelles Wissen geht, ist Schweitzer Fachinformationen wegweisend. Kunden aus Recht und Beratung sowie Unternehmen, öffentliche Verwaltungen und Bibliotheken erhalten komplette Lösungen zum Beschaffen, Verwalten und Nutzen von digitalen und gedruckten Medien.
Preface xi
1. Introduction 1Stig Pedersen-Bjergaard
1.1 What Is Bioanalysis? 1
1.2 What Is the Purpose of Bioanalysis, and Where Is It Conducted? 2
1.3 Bioanalysis Is Challenging 6
1.4 The Different Sections of This Textbook 7
2. Physicochemical Properties of Drug Substances 9Steen Honoré Hansen and Leon Reubsaet
2.1 Bioanalysis in General 9
2.2 Protolytic Properties of Analytes 10
2.3 Partitioning of Substances 12
2.4 Stereochemistry 15
2.5 Peptides and Proteins 18
3. Biological Samples: Their Composition and Properties, and Their Collection and Storage 23Steen Honoré Hansen
3.1 Introduction 23
3.2 Blood, or Whole Blood 24
3.3 Plasma and Serum 26
3.4 Urine 26
3.5 Feces 27
3.6 Saliva 28
3.7 Cerebrospinal Fluid 28
3.8 Synovial Fluid 28
3.9 Hair and Nails 29
3.10 Tissue (Biopsies) 29
4. General Chromatographic Theory and Principles 31Steen Honoré Hansen
4.1 General Introduction 31
4.2 General Chromatographic Theory 32
4.3 Theory of Partition 33
4.4 Retention 34
4.5 Separation Efficiency 35
4.6 Resolution 36
4.7 Selectivity 37
4.8 The Separation Process 38
4.9 Chromatographic Principles 41
4.10 Reversed Phase Chromatography 45
4.11 Size Exclusion Chromatography (SEC) 54
4.12 Ion Exchange Chromatography 56
4.13 Chiral Separations 57
5. Quantitative and Qualitative Chromatographic Analysis 61Steen Honoré Hansen
5.1 Collection of Chromatographic Data 61
5.2 Quantitative Measurements 62
5.3 Calibration Methods 63
5.4 Validation 67
5.5 Qualitative Analysis 69
6. Sample Preparation 73Stig Pedersen-Bjergaard, Astrid Gjelstad, and Trine Grønhaug Halvorsen
6.1 Why Is Sample Preparation Required? 73
6.2 What Are the Main Strategies? 75
6.3 Protein Precipitation 75
6.4 Liquid-Liquid Extraction 80
6.5 Solid-Phase Extraction 94
6.6 Dilute and Shoot 110
6.7 What Are the Alternative Strategies? 113
7. High-Performance Liquid Chromatography (HPLC) and High-Performance Liquid Chromatography-Mass Spectrometry (LC-MS) 123Steen Honoré Hansen and Leon Reubsaet
7.1 Introduction 123
7.2 The Solvent Delivery System 125
7.3 Degassing and Filtering of Mobile Phases 127
7.4 Injection of Samples 128
7.5 Temperature Control 128
7.6 Mobile Phases 129
7.7 Stationary Phases and Columns 130
7.8 Detectors 135
7.9 Mass Spectrometric Detection 143
8. Gas Chromatography (GC) 173Stig Pedersen-Bjergaard
8.1 Basic Principles of GC 173
8.2 GC Instrumentation 174
8.3 Carrier Gas 177
8.4 Stationary Phases 178
8.5 Separation Selectivity in GC 180
8.6 Columns 182
8.7 Injection Systems 183
8.8 Detectors 185
8.9 Derivatization 187
8.10 Gas Chromatography-Mass Spectrometry (GC-MS) 188
9. Analysis of Small-Molecule Drugs in Biological Fluids 207Steen Honoré Hansen and Stig Pedersen-Bjergaard
9.1 Plasma and Serum Samples 207
9.2 Whole Blood Samples 234
9.3 Dried Blood Spots 241
9.4 Urine Samples 245
9.5 Saliva 253
References 259
10. Analysis of Peptide and Protein Drugs in Biological Fluids 261Leon Reubsaet and Trine Grønhaug Halvorsen
References 282
11. Regulated Bioanalysis and Guidelines 283Martin Jørgensen and Morten A. Kall
11.1 Introduction 283
11.2 The Evolution of Regulated Bioanalysis 284
11.3 Bioanalytical Method Validation 286
11.4 Pre-study Validation 287
11.5 In-Study Validation 299
11.6 Documentation 300
11.7 Regulatory Requirements to Bioanalysis 300
11.8 Quality Systems in Regulated Bioanalysis 301
Index 305
Stig Pedersen-Bjergaard
School of Pharmacy, University of Oslo, Norway School of Pharmaceutical Sciences, University of Copenhagen, Denmark
Welcome to the field of bioanalysis! Through reading of this textbook, we hope you get fascinated by the world of bioanalysis, and also we hope that you learn to understand that bioanalysis is a highly important scientific discipline. In this chapter, five fundamental questions are raised and briefly discussed as an introduction to the textbook: (i) What is bioanalysis? (ii) What is the purpose of bioanalysis? (iii) Where is bioanalysis conducted? (iv) Why do you need theoretical understanding and skills in bioanalysis? And (v) how do you gain the understanding and the skills from reading this textbook?
In this textbook, we define bioanalysis as the chemical analysis of pharmaceutical substances in biological samples. The purpose of the chemical analysis is normally both to identify (identification) and to quantify (quantification) the pharmaceutical substance of interest in a given biological sample. This is performed by a bioanalytical chemist (scientist) using a bioanalytical method. The pharmaceutical substance of interest is often termed the analyte, and this term will be used throughout the textbook. Identification of the analyte implies that the exact chemical identity of the analyte is established unequivocally. Quantification of the analyte implies that the concentration of the analyte in the biological sample is measured. It is important to emphasize that quantification is associated with small inaccuracies, and the result is prone to errors. Thus, the quantitative data should be considered as an estimate of the true concentration. Based on theoretical and practical skills, and based on careful optimization and testing of the bioanalytical methods, the bioanalytical chemist tries to reduce the error level, providing concentration estimates that are very close to the true values.
Bioanalytical data are highly important in many aspects. As an example, a patient serum sample is analyzed for the antibiotic drug substance gentamicin, and gentamicin is measured in the sample at a concentration of 5 µg/ml. First, the identification of gentamicin in the blood serum sample confirms that the patient has taken the drug. This is important information because not all patients actually comply with the prescribed medication. Second, the exact concentration of gentamicin measured in the blood serum sample confirms that the amount of gentamicin taken is appropriate, as the recommended concentration level should be in the range of 4-10 µg/ml. For aminoglycoside antibiotics such as gentamicin, it is recommended to monitor the concentration in blood if the treatment is expected to continue for more than 72 hours as these antibiotics have the potential to cause severe adverse reactions, such as nephrotoxicity and ototoxicity.
As will be discussed in much more detail in this book, not only blood serum samples are used for bioanalysis. Bioanalysis can be performed on raw blood samples (whole blood) or on blood samples from which the blood cells have been removed (serum or plasma). Alternatively, bioanalysis can be performed from urine or saliva as examples, depending on the purpose of the bioanalysis. Bioanalysis is performed both on human samples and on samples from animal experiments.
Bioanalysis is conducted in the pharmaceutical industry, in contract laboratories associated with the pharmaceutical industry, in hospital laboratories, in forensic toxicology laboratories, and in doping control laboratories. In the pharmaceutical industry and in the associated contract laboratories, bioanalysis is basically conducted to support the development of new drugs and new drug formulations. In hospital laboratories, bioanalysis is used to monitor existing drugs in patient samples, to check that individual patients take their drugs correctly. In forensic toxicology laboratories and doping laboratories, bioanalysis is used to check for abuse of drugs and drug-related substances.
Bioanalytical laboratories are highly important in the development of new drugs and new drug formulations in the pharmaceutical industry. Thus, identification and quantification of drug substances and metabolites in biological samples like blood plasma, urine, and tissue play a very important role during drug development. Drug development begins with the identification of a medical need and hypotheses on how therapy can be improved. Drug discovery is the identification of new drug candidates based on combinatorial chemistry, high-throughput screening, genomics, and ADME (absorption, distribution, metabolism, and elimination). By combinatorial chemistry, a great number of new drug candidates are synthesized, and these are tested for pharmacological activity and potency in high-throughput screening (HTS) systems. The HTS systems simulate the interaction of the drug candidates with a specific biological receptor or target. Once a lead compound is found, a narrow range of similar drug candidates is synthesized and screened to improve the activity toward the specific target. Other studies investigate the ADME profile of drug candidates by analyzing samples collected at different time points from dosed laboratory animals (in vivo testing) and tissue cultures (in vitro testing).
Drug candidates passing the discovery phase are subjected to toxicity testing and further metabolism and pharmacological studies in the preclinical development phase. Both in vivo and in vitro tests are conducted, and various animal species are used to prove the pharmacokinetic profile of the candidate. The detailed information about the candidate forms the basis for further pharmaceutical research on the synthesis of raw materials, the development of dosage forms, quality control, and stability testing.
The clinical development phase can begin when a regulatory body has judged a drug candidate to be effective and to appear safe in healthy volunteers. In phase I, the goal is to establish a safe and efficient dosage regimen and to assess pharmacokinetics. Blood samples are collected and analyzed from a small group of healthy volunteers (20-80 persons). The data obtained form the basis for developing controlled phase II studies. The goal of phase II studies is to demonstrate a positive benefit-risk balance in a larger group of patients (200-800) and to further study pharmacokinetics. Monitoring of efficacy and monitoring of possible side effects are essential. Phase II studies can take up to two years to fulfill. At the end of phase II, a report is submitted to the regulatory body, and conditions for phase III studies are discussed. Additional information supporting the claims for a new drug is provided. Phase III begins when evidence for the efficacy of the drug candidate and supporting data have demonstrated a favorable outcome to the regulatory body. The phase III studies are large-scale efficacy studies with focus on the effectiveness and safety of the drug candidate in a large group of patients. In most cases, the drug candidate is compared with another drug already in use for treatment of the same condition. Phase III studies can last two to three years or more, and 3000-5000 patients can be involved. Carcinogenetic tests, toxicology tests, and metabolic studies in laboratory animals are conducted in parallel. The cumulative data form the basis for filing a new drug application to the regulatory body and for future plans for manufacturing and marketing. The regulatory body thoroughly evaluates the documentation that is provided before a market approval can be authorized and the drug product can be legally marketed. The time required from drug discovery to product launch is up to 12 years. Phase IV studies are studies that are conducted after product launch to demonstrate long-term effects and new claims, expand on approved claims, examine possible drug-drug interactions, and further assess pharmacokinetics. Several thousand patients participate in phase IV studies.
Bioanalytical measurements are conducted during drug discovery, preclinical development, and clinical development, and they are intended to (among other things) generate the experimental data to establish the pharmacokinetics, the toxicokinetics, and the exposure-response relationships for a new drug. The pharmacokinetics of a certain drug substance describes how the body affects the drug after administration (ADME): how the drug is absorbed (A) and distributed (D) in the body, and how the drug is metabolized (M) by metabolic enzymes and chemically changed to different types of metabolites, which in turn are excreted (E) from the body. Bioanalysis is used extensively in pharmacokinetic studies, among others, to establish blood concentration-time profiles, and to measure the rate of drug metabolism and excretion. This involves a large number of both animal and human samples.
Toxicokinetics studies, in contrast, are intended to investigate the relationship between the exposure of a new drug candidate in experimental animals and its toxicity. This type of information is used to establish a relationship between the possible toxic properties of a drug in animals and those in humans. Toxicokinetic studies involve bioanalysis in both animal and human...
Dateiformat: ePUBKopierschutz: Adobe-DRM (Digital Rights Management)
Systemvoraussetzungen:
Das Dateiformat ePUB ist sehr gut für Romane und Sachbücher geeignet – also für „fließenden” Text ohne komplexes Layout. Bei E-Readern oder Smartphones passt sich der Zeilen- und Seitenumbruch automatisch den kleinen Displays an. Mit Adobe-DRM wird hier ein „harter” Kopierschutz verwendet. Wenn die notwendigen Voraussetzungen nicht vorliegen, können Sie das E-Book leider nicht öffnen. Daher müssen Sie bereits vor dem Download Ihre Lese-Hardware vorbereiten.Bitte beachten Sie: Wir empfehlen Ihnen unbedingt nach Installation der Lese-Software diese mit Ihrer persönlichen Adobe-ID zu autorisieren!
Weitere Informationen finden Sie in unserer E-Book Hilfe.