Vous êtes sur la page 1sur 14

MSc ANALYTICAL TOXICOLOGY (2010 -11)

The below gives a detailed description of each of the four taught


modules and the project:
Semester 1 (September to December)

Module 1 - Principles of Analytical Toxicology


Module 2 - Biochemical Toxicology

Semester 2 (January to April)


Module 3 - Clinical and Forensic Toxicology
Module 4 - Advanced Analytical Toxicology
The project (module 5) takes place following semester 2, from May to the beginning of
September.

Module 1

PRINCIPLES OF ANALYTICAL TOXICOLOGY

Module Organisers: Dr Andrew Kicman


Analytical Toxicology
The aim of this module is to introduce the student to the major chromatographic and spectroscopic
techniques used in analytical toxicology, the instrumentation used and the scientific principles
underlying the instrumentation, and its use in sample analysis.
At the end of the module the student should be able to:

Describe the chromatographic and spectroscopic instrumentation used and explain how it
operates to enable analysis
Explain the scientific principles underlying each chromatographic and spectroscopic
technique
Compare and contrast the various techniques giving advantages and disadvantages of each
Decide which technique(s) would be most appropriate for the analysis of a given
sample and explain the reasons for your decision
Give examples of the uses for each technique in analytical science, with particular emphasis
on analytical toxicology.

Introduction to analytical toxicology


Introduction to course structure and content
Integration of analytical toxicology as covered by the four taught modules and the research
project
Commonly applied techniques, past, present and future
Clinical and forensic perspectives
Immunochemical techniques
Immunoassay
Immunoglobulins

Competitive and non-competitive immunoassays

Chromatography
Introduction to separation science
The role of analytical chemistry in analytical science
Analytical techniques used in analytical science
The elements of analysis
The pros and cons of the various techniques
Maximising information and evidential value
What is separation science? Basis of separation
Terminology
Definition of chromatography
Chromatographic theory
The mechanisms of separation Introducing the major separation techniques used in analytical
chemistry
Exploring the basis of separation - intermolecular interactions (hydrogen bonding, van der
Waals forces, ionic interactions)
High Performance Liquid Chromatography (HPLC) Introducing instrumentation
Normal phase HPLC
Reverse phase HPLC
Mechanisms of separation operating in HPLC
Stationary and mobile phases

Detecting the sample

Uses of HPLC
Gas Chromatography (GC) Instrumentation
Carrier gases
Injectors
Chromatographic columns and stationary phases
Use of temperature
Detectors
How to manipulate separation
Uses of GC (including headspace analysis)
Chirality
Separation of enantiomers
Direct and indirect methods
Sample Handling Solvent and solid state extraction
Manipulation of pH to aid separation
Analysis of conjugates

Spectroscopy and Spectrometry


Introduction to spectroscopy
The five properties of light
Spectral regions
Spectroscopic techniques
Energy levels

Electronic Spectroscopy: Absorption


UV/Visible absorption spectra
Chromophores
Absorbance
Beers Law and concentration
The measurement of spectra and application of UV/Visible spectroscopy
Comparison of instrument types: single beam, double beam, Diode array, Fourier-Transform
Mass Spectrometry
The technique
Instrumentation
Applications
Hyphenated Mass Spectrometric techniques and applications (LC-MS, GC-MS)
Application of techniques
Substance identification
Quantification
Additional Spectrophotometry/spectrometry
Atomic Absorption and Emission Spectroscopy
Inductively Coupled Plasma Mass Spectrometry
Optical Emission Spectroscopy
Mathematics for Analysts

Mathematical tools for analysts (chemometrics)


Use of concentration units
Chemical calculations for practical analytical work
Reporting of results, use of concentration units and reference ranges

Quality Assurance

Traceability
Reference materials and drug standards.
Quality Control procedures
Quality Assurance
Method validation
Uncertainty budget fit for purpose
International Committee on Harmonisation (ICH), uncertainty and guidelines
(e.g. ISO 17025)
Laboratory accreditation (e.g. UKAS )

Specimen Preparation and Extraction

Specimen collection, preservation and storage, chain of custody and documentation


of specimens
Specimen preparation and extraction techniques including how to develop
methods, importance of pKa to liquid and solid phase extraction efficiency

Practicals, Workshops and Tutorials


Chemistry catch-up Polarity, lipophilicity, pKa

Solubility
Intermolecular interactions
Functional groups and chemical nomenclature

Calculations for analysts (Chemometrics)


Worked examples of chemical calculations
Common errors in calculations and their consequences
Laboratory skills
Health and safety in the laboratory
Laboratory notebook keeping
Weighing top pan and analytical balances
Correct use and care of pipettes.
Pipette skills exercise simple quantification (mass and volume)
Volumetric work
Calibrating volumetric apparatus
Preparing standard solutions and determining the concentration of an unknown
Buffer preparation weighing, use of pH meters, concentrations and dilutions
Spot tests / Colorimetric
Analysis of a common drug
Determination of concentration using UV fixed wavelength
Gas Chromatography (Direct injection of alcohol)
Analysis of a chemical mixture
Identification of components using retention time
Evaluation of data and preparation of reports
Gas Chromatography-Mass Spectrometry:
Analysis of a chemical mixture
Identification of components using MS library searching
Evaluation of data and preparation of reports
GC-MS data handling workshop - Matching MS spectra from samples against library spectra
Using MS spectra to select ions for selected ion monitoring (SIM)
Quantification using SIM data including QA
High Performance Liquid Chromatography model analytes, e.g. anticonvulsants
The elements of the instrument
Setting up methods
Sample injection
Determining plate number and resolution
Data analysis and interpretation
High Performance Liquid Chromatography-MS
o Analysis of a simple drug mixtures, e.g. drugs of abuse
Spectroscopy
UV-Vis:

Instrument calibration QA & QC


Photometric calibration
Resolution
Determining concentration using UV-vis

Specimen Preparation And Extraction

Specimen collection, preservation and storage, chain of custody and documentation


of specimens
Specimen preparation and extraction techniques including how to develop
methods, importance of pKa to liquid and solid phase extraction efficiency

Introduction to the Principles of Qualitative and Quantitative Analysis


o Substance identification
o Quantification with and without use of internal standard
Tutorials to support practicals

MODULE 2

BIOCHEMICAL TOXICOLOGY

Module Organisers: Dr Andrew Kicman


Biochemical Toxicology
The aims of this module are to provide an understanding of the basic principles of drug absorption,
distribution, metabolism and excretion and elementary pharmacokinetics. The basic concepts,
mechanisms and principles of toxicology and pharmacogenetics in relation to drug development, safety
evaluation, requirements for toxicity testing and risk assessment, adverse drug reactions and toxicity.
At the end of the module the student should be able to:

Understand the absorption, distribution and excretion of xenobiotics


Identify the potential sites, reactions and factors which influence drug metabolism
Appreciate the significance of drug metabolism on biological activity
Understand the principles of pharmacokinetics and be able to define and calculate the
commonly used pharmacokinetic parameters
Have an understanding of the basic concepts and principles of toxicology
Have an appreciation of the mechanisms underlying the toxicity of drugs and other
xenobiotics
Have a theoretical basis for understanding the role of drug metabolism in relation to toxicity
Have an appreciation of risk assessment and the role of toxicological information
Be able to interpret and integrate toxicological information
Have gained problem solving abilities in relation to toxicology
Be able to apply these skills in the assessment of actual and potential drug toxicity

Chemistry for biologists / Biology for chemists

Anatomy and physiology


Basic anatomy and physiology including major organ systems (eg brain, kidney, liver, lungs
and GI tract)
Basic Introduction to clinical biochemistry
Including blood, plasma, urine, electrolytes, fluid balance, proteins, enzymes, hormones
Medical terminology and acronyms
Introduction to pharmacological classification and drug action

Introduction to clinical pharmacology


Drug classification and control
Mechanisms of drug action
Acute and chronic drug toxicity and potential for drug abuse

Introduction to drug distribution


Basic definitions of absorption, distribution and elimination.
Drug transport in the blood, protein binding.
Transfer within body fluid compartments, drug tissue localization.
Blood-brain barrier, placental drug transfer
Renal and biliary excretion.
Biological fate of xenobiotics
Introduction to basic concepts in xenobiotic biotransformation
Functionalisation reactions
Oxidations
Reductions
Hydrolysis as Phase I or functionalisation reactions
Conjugation reactions
Glucuronidation
Sulphation
N-acetylation
Glutathione conjugation
Methylation
Amino acid conjugation
Factors affecting drug metabolism
Physiological and environmental factors affecting biotransformation
Pharmacological significance of xenobiotic biotransformation
Effect of disease and genetic polymorphism on human xenobiotic biotransformation
Introduction to Pharmacokinetics
Simplifying assumptions
The concept of compartments
Intravenous drug administration
Definitions of pharmacokinetic parameters
Elimination rate constant half-life
Volume of distribution
Clearance
Intravenous drug administration
Compartmental models
Oral drug administration
Determination of absorption rate
Bioavailability
Use of AUC to determine bioavailability
Urinary drug excretion
Multiple dosing
Drug accumulation

Steady-state
Dose regimens

Basic principles of toxicology


Introduction to the science of toxicology and its importance
Dose response relationship and what may be derived from it
Types of toxic response
The adverse responses to chemical and drug exposure which may be manifested by mammalian
organisms such as humans
Reactive intermediates
The types of chemically reactive intermediates that may be produced by metabolism of a
xenobiotic and the consequences of this
Genotoxicity
Mechanisms of DNA modification by xenobiotics
Chemical carcinogenesis
N-Oxidation and drug toxicity
Mechanisms of arylamine mediated hepatic and bladder carcinogenesis
Induction of ferrihaemoglobinemia by arylhydroxylamines
Sulphur toxins
Toxic sulphur conjugates derived from glutathione: MDMA and dopamine
Target organ toxicity
Liver
Kidney
Lung
CNS - The reasons that specific organs are targeted are discussed
Toxicokinetics
Discussion of the relationship between pharmacokinetics and the toxicity of drugs and
chemicals
Specific examples of drug toxicity
Particular drugs will be studied in detail in relation to their toxicities and mechanisms
underlying them

MODULE 3 CLINICAL AND FORENSIC TOXICOLOGY

Module Organisers: Professor Robin Braithwaite


The aim of this module is to provide an in depth understanding of common drugs and chemical
poisons relevant to both clinical and forensic toxicology. A strong emphasis will be placed on the
application of modern analytical methods to the measurement of drugs, chemical poisons and their
metabolites in biological fluids in both a clinical and medicolegal context. Although this is a standalone module it is designed to be integrated with Module 4 (Advanced Analytical Toxicology), which
has a high laboratory practical element. The emphasis of this module is to integrate aspects of drug
pharmacology and metabolism with extensive use of both clinical and forensic case reports. The

module will include assessed coursework, data evaluation, report preparation and presentation,
tutorials and workshops.
At the end of the module the student should be able to:

Understand the clinical and forensic issues associated with the laboratory investigation of
acute and chronic poisoning, including drug and substance misuse
Understand the main clinical and biochemical features associated with acute and chronic
poisoning due to the ingestion of important drug and chemical poisons
Understand the pharmacokinetics and metabolism of important drug and chemical poisons
and relationship to acute and chronic toxicity, including sudden or unexpected deaths
Critically review and evaluate the significance of analytical data from human poisoning cases
Present the significance of analytical findings accurately and concisely, including court
reporting
Demonstrate an understanding of the various analytical approaches to the detection and
quantitation of drugs and metabolites in biological fluids and tissues
Understand the role of both laboratory testing and point of care testing in the management
and detection of drug misuse
Understand the principles and practice of therapeutic drug monitoring

Drug misuse, poisoning and its epidemiology


The use and abuse of substances used as both medicines and poisons
International epidemiology of poisoning and its medical definitions and current trends
Available information sources, for example Toxbase, poisons information services
Current issues including chemical poisons and counterfeit pharmaceuticals
Clinical management of poisoning
Signs and symptoms of poisoning
Antidotes, enhanced elimination techniques
Clinical care of poisoned patients
Alcohol (ethanol)
The clinical and forensic toxicology of alcohol, its abuse and consequences
Measurement of alcohol in body fluids and it relationship to effects & toxicity
Pharmacokinetics and metabolism of alcohol
Acute and chronic alcohol toxicity and suspected alcohol related deaths
Methanol and Glycols
The clinical and forensic toxicology of methyl and other alcohols including ethylene,
propylene and diethylene glycol
Laboratory techniques for the measurement of alcohols and glycols in body fluids
Pharmacokinetics and metabolism of methanol and ethylene glycol
Acute and chronic toxicity and fatalities
Amphetamines
The clinical and forensic toxicology of amphetamine and closely related drugs including
methamphetamine, and MDMA (ecstasy)
Laboratory techniques used in the investigation of suspected poisoning or abuse
The pharmacokinetics and metabolism of amphetamines
Acute and chronic toxicity and fatalities
Cannabis and Cannabinoids
The clinical and forensic toxicology of cannabis

Cocaine

Laboratory techniques used in the investigation of suspected cannabis abuse and


intoxication
The pharmacokinetics and metabolism of cannabinoids
Acute and chronic toxicity

The clinical and forensic toxicology of cocaine


Laboratory techniques used in the investigation of suspected cocaine abuse and intoxication
The pharmacokinetics and metabolism of cocaine
Acute and chronic toxicity and fatalities

Opiates
The clinical and forensic toxicology of opiates including morphine , heroin , codeine and
dihydrocodeine
Laboratory techniques used in the investigation of suspected opiate abuse and overdosage
The pharmacokinetics and metabolism of opiates
Acute and chronic toxicity and fatalities
Opioids
The clinical and forensic toxicology of important opioid drugs including methadone,
dextropropoxyphene and buprenorphine
Laboratory techniques used in the investigation of suspected opioid abuse and overdosage
The pharmacokinetics and metabolism of opioids
Acute and chronic toxicity and fatalities
Newer drugs: GHB, Ketamine and Piperazines
The clinical and forensic toxicology of GHB, ketamine and piperazines
Laboratory techniques used in the investigation of suspected abuse and overdosage
The pharmacokinetics and metabolism of GHB, ketamine and piperazines
Acute and chronic toxicity and fatalities
Benzodiazepines
The clinical and forensic Toxicology of benzodiazepines and related hypnotic drugs such as
zopiclone, zolpidem and related drugs.
Laboratory techniques for the detection and measurement of these drugs in biological fluids
in the investigation of suspected abuse and overdose.
The pharmacokinetics and metabolism of this group of drugs and relationship to acute and
chronic toxicity and suspected fatalities will be reviewed
Psychotropic drugs
The clinical and forensic toxicology of major groups of psychotropic drugs such as the
antidepressants and antipsychotics
Laboratory techniques for the detection and measurement of these drugs in biological fluids
in the investigation of suspected poisoning and overdosage
The pharmacokinetics and metabolism of important drugs and relationship to acute and
chronic toxicity and suspected fatalities will be reviewed
Pesticides: OPs , Paraquat , Phosphides , Strychnine
The clinical and forensic toxicology of important pesticides
Laboratory techniques for detection and measurement of these agents in biological fluids in
the investigation of suspected poisoning
The pharmacokinetics and metabolism of these agents and relationship to acute and chronic
toxicity and suspected fatalities will be reviewed

Non opioid analgesics


The clinical and forensic toxicology of important non-opioid analgesics such as paracetamol
and aspirin
Laboratory techniques for the detection and measurement of these drugs and their
metabolites in biological fluids in the investigation of suspected poisoning
The pharmacokinetics and metabolism of these drugs and relationship to acute and chronic
toxicity and suspected fatalities will be reviewed
Cardiovascular agents
The clinical and forensic toxicology of important cardiovascular agents, including beta
blockers, antiarrhythmic agents, calcium antagonists and digoxin
Laboratory techniques for the detection and measurement of various agents in biological
fluids
The pharmacokinetics and metabolism of cardiovascular agents and relationship to acute
and chronic toxicity including fatalities will be reviewed
Metallic poisons
The clinical and forensic toxicology of important metals, including arsenic, lead, mercury and
thallium
Laboratory techniques for the detection and measurement of heavy metals in biological
fluids and tissues
The pharmacokinetics and metabolism of important heavy metals and relationship to acute
and chronic toxicity, including fatalities will be reviewed
Therapeutic Drug Monitoring
Anticonvulsants
Lithium
Cardiovascular agents
Cancer therapy
Fire deaths: Carbon monoxide and Cyanide

The clinical and forensic toxicology of carbon monoxide and cyanide


Laboratory techniques for the detection and measurement of carbon monoxide and cyanide
in biological fluids in the investigation of suspected poisoning
The pharmacokinetics and metabolism of these agents and relationship to acute and chronic
toxicity and suspected fatalities will be reviewed
Volatile substance abuse
The clinical and forensic toxicology of volatile substance abuse. Laboratory techniques for
the identification and measurement of common volatile substances in biological fluids and
tissues
The pharmacokinetics and metabolism of volatile substances and relationship to acute and
chronic toxicity, including fatalities will be reviewed
Drugs in Sport
The toxicology of anabolic steroids, protein hormones (human growth hormone, insulin and
erythropoietin) and other drugs used by bodybuilders and athletes.
Laboratory techniques employed for the detection of doping, as developed by laboratories
accredited by the World Anti-doping Agency
Poisonous plants and herbal medicines
The clinical and forensic toxicology of major poisonous plants and common herbal medicines
Laboratory and other techniques for the identification and analysis of plant material
including the analysis of biological fluids
Role of laboratory investigations

The role of the laboratory investigations in the diagnosis and management of suspected
poisoning
The role of the laboratory in the investigation of sudden or unexpected deaths
Best practice for the provision of laboratory investigations including the techniques used and
their application to patient care

Legislation and medico-legal aspects of drug abuse and poisoning

International conventions on drugs and poisons

Role of the coroner and medical examiner

Role of the toxicologist in court

Preparation of legal reports and oral presentation

Laboratory visit
To be arranged.

Workshops
Investigation of drug misuse - analytical, clinical, occupational and forensic aspects .
Introduction to trends in drug misuse
Specimen collection , adulteration and chain of custody
Analytical techniques for screening and confirmation
Post - mortem Toxicology
The legal system and introduction to the role of the coroner /medical examiner, coroners
officers and police.
The post-mortem examination , post mortem changes and physiology
and collection of specimens . The role of the pathologist
Laboratory investigation of unexpected or sudden death What to look for ,
Selection of analytical techniques, Choice of specimens and Interpretation
of results .The role of the toxicologist .
Problems and pitfalls in the interpretation of post mortem findings
for drugs and alcohol e.g. putrefaction , post mortem redistribution
and exhumation - some illustrative cases .
Investigation of heroin related deaths, mechanisms of death and current clinical and
forensic issues
Use of hair analysis in understanding historical drug use in the deceased .

Forensic Evidence
Writing court reports and presentation of evidence in court.
Exercise based on a real case .

MODULE 4

ADVANCED ANALYTICAL TOXICOLOGY

Module Organisers: Dr Andrew Kicman & Dr Mark Parkin


The aim of Module 4 is to provide the student with an understanding of sample handling,
immunoassay, on-site testing for drugs and alcohol and mass spectrometry.

At the end of the module the student should be able to:

Describe the principles and practice of sample handling


Describe the principles of immunoassay, the importance of specificity and know how to
interpret the data
Compare the advantages to the disadvantages of on-site drug testing
Describe the mass spectrometric instrumentation used and explain how it operates to enable
analysis
Compare and contrast the various ionisation techniques giving advantages and
disadvantages of each technique
Compare and contrast the various mass analysers giving advantages and disadvantages of
each technique
Explain the scientific principles underlying the technique
Decide which technique(s) would be most appropriate for the analysis of a given
chemical/biological sample and explain the reasons for your decision
Give examples of the uses for each technique in analytical chemistry, with particular
emphasis on analytical science.

Advanced sample handling


Consideration of matrices (hair, oral fluid, blood, urine, vitreous humor)
Solid Phase Extraction (reverse phase, normal phase, ion exchange, mixed-mode)
Solid Phase Microextraction
Critical Evaluation of Published Methods
Dilute and inject dilute and shoot (for mass spectrometry)
Immunoassay
Heterogenous and homogenous assays
Non-isotopic versus isotopic assays
Immunoglobulin Affinity, including Scatchard Plot
Antibody Titre
Specificity
Data Analysis
On-site testing for drugs
Near patient testing devices; roadside testing
Technology
Specificity
Matrices (urine versus saliva); breath alcohol
Advantages and disadvantages
Future role

ADVANCED MASS SPECTROMETRY

Ionisation techniques in practice


Electron ionisation
Chemical ionisation
Atmospheric pressure ionisation
Atmospheric pressure chemical ionisation (APCI)
Electrospray ionisation (ESI)
Desorption electrospray ionisation (DESI) and Direct Analysis in Real Time (DART)
Inductively coupled plasma resonance MS
Collision cell
Ionisation and hyphenated techniques

Mass analysers
Linear quadrupoles
3D ion traps
2D ion traps
Magnetic sector instruments (isotope ratio mass spectrometry)
Time Of Flight (TOF) including reflectron and delayed extraction

MS in Analytical Toxicology
Chromatography and mass spectrometry refresher
Gas Chromatography-Mass Spectrometry (GC-MS)
Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)
Liquid Chromatography-Time Of Flight Mass Spectrometry (LC-TOFMS)
Inductively Coupled Plasma Mass Spectrometry (ICP-MS)
Gas Chromatography-Combustion-Isotope Ratio Mass Spectrometry (GC-C-IRMS)
Direct Analysis in Real Time (DART)
n

MS techniques in practice
Selected reaction monitoring
Product ion scanning
Precursor ion scanning
Neutral loss scanning
Effect of time domain & energies
Collision Activated Dissociation (CAD)
Software
Interpretation of Mass Spectra
Isotope Abundance
Accurate Mass And Elemental Composition`
Fragmentation Pathways
Method Development and Validation
Infusion
Chromatography
Instrument Tuning
Matrix Effects
Identification Criteria for Confirmatory Purposes
Quantification
Large Molecule Mass Spectrometry
Protein identification
Analysis of intact and digested proteins
Peptide sequencing
Peptide mass mapping
Post-translational modifications
Examples, e.g. Insulin

Practicals and Workshops


In these practical classes and workshops, you will have the opportunity to gain hands on in order to
learn the key elements of sample extraction, immunoassay, on-site testing and mass spectrometry.
These will include:

Sample Extraction using Reverse Phase, Normal Phase, Ion-Exchange and Mixed Mode
Cartridges
Competitive Immunoassay for Recreational Drugs
Handling of On-site Testing Devices, including alcometers
Mass Spectrometry including infusion and hyphenated Separation Techniques
Tuning the Mass Spectrometer
Resolution, sensitivity and mass accuracy
Mass Range, Scan and Selected Ion Monitoring
Sensitivity and speed
MS/MS selected reaction monitoring
Identification of a Substance in a Matrix
Quantification of target analytes

MODULE 5

Project

All students have an analytical toxicology placement, either of a clinical or forensic nature between
May and August. The total length of the placement is 16 weeks, which includes preparation of the
project report.
This academic year, placements will begin in May.
Students generally undertake the placement in a laboratory or organisation exterior to King's,
although staff at King's also offer placements that may be of interest to students and some
organisations offer placements where some of the practical work takes place in the organisation and
some takes place at Kings.
You must recognise that it is not always possible to assign you your first or second (or sometimes
even your third) placement choice, although we will do our best. If you refuse to select an alternative
placement from those on offer, one will be assigned to you and you will be expected to take it,
irrespective of where in the UK it is located.
Security clearance is mandatory for placements in some laboratories. The host laboratory will arrange
for the security clearance.
It should be noted that we reserve the right to refuse an external placement. In these circumstances,
the individual will carry out their placement within King's.
Assessment
Assessment will be via a written review of the literature pertaining to the research topic; submission
of a journal article; presentation of a poster with oral questioning by assessors.

Literature Review
Research Article, in the style of a journal publication
Poster presentation, including oral examination
Supervisors evaluation

Vous aimerez peut-être aussi